WO2005121130A2 - Chemical compounds and pharmaceutical compositions containing them for the treatment of inflammatory disorders - Google Patents

Chemical compounds and pharmaceutical compositions containing them for the treatment of inflammatory disorders Download PDF

Info

Publication number
WO2005121130A2
WO2005121130A2 PCT/US2005/019131 US2005019131W WO2005121130A2 WO 2005121130 A2 WO2005121130 A2 WO 2005121130A2 US 2005019131 W US2005019131 W US 2005019131W WO 2005121130 A2 WO2005121130 A2 WO 2005121130A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
group
mmol
alkyl
heteroaryl
Prior art date
Application number
PCT/US2005/019131
Other languages
French (fr)
Other versions
WO2005121130A3 (en
Inventor
Zhuyan Guo
Peter Orth
Zhaoning Zhu
Robert D. Mazzola
Tin Yau Chan
Henry A. Vaccaro
Brian Mc Kittrick
Joseph A. Kozlowski
Brian J. Lavey
Guowei Zhou
Sunil Paliwal
Shing-Chun Wong
Neng-Yang Shih
Pauline C. Ting
Kristin E. Rosner
Gerald W. Shipps, Jr.
M. Arshad Siddiqui
David B. Belanger
Chaoyang Dai
Dansu Li
Vinay M. Girijavallabhan
Janeta Popovici-Muller
Wensheng Yu
Lianyun Zhao
Original Assignee
Schering Corporation
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=35355800&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2005121130(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Schering Corporation filed Critical Schering Corporation
Priority to CA002569111A priority Critical patent/CA2569111A1/en
Priority to JP2007515501A priority patent/JP2008501691A/en
Priority to AU2005252201A priority patent/AU2005252201A1/en
Priority to MXPA06014054A priority patent/MXPA06014054A/en
Priority to EP05759261A priority patent/EP1773821A2/en
Publication of WO2005121130A2 publication Critical patent/WO2005121130A2/en
Publication of WO2005121130A3 publication Critical patent/WO2005121130A3/en
Priority to IL179674A priority patent/IL179674A0/en

Links

Classifications

    • 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
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • 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
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/08Plasma substitutes; Perfusion solutions; Dialytics or haemodialytics; Drugs for electrolytic or acid-base disorders, e.g. hypovolemic shock
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/04Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C235/06Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton being acyclic and saturated having the nitrogen atoms of the carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/04Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C235/08Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton being acyclic and saturated having the nitrogen atom of at least one of the carboxamide groups bound to an acyclic carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/04Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C235/10Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton being acyclic and saturated having the nitrogen atom of at least one of the carboxamide groups bound to an acyclic carbon atom of a hydrocarbon radical substituted by nitrogen atoms not being part of nitro or nitroso groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/18Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D207/20Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/18Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D207/22Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/24Oxygen or sulfur atoms
    • C07D207/262-Pyrrolidones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/44Iso-indoles; Hydrogenated iso-indoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/12Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D215/14Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/02Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with only hydrogen atoms or radicals containing only carbon and hydrogen atoms, directly attached to carbon atoms of the nitrogen-containing ring; Alkylene-bis-isoquinolines
    • C07D217/06Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with only hydrogen atoms or radicals containing only carbon and hydrogen atoms, directly attached to carbon atoms of the nitrogen-containing ring; Alkylene-bis-isoquinolines with the ring nitrogen atom acylated by carboxylic or carbonic acids, or with sulfur or nitrogen analogues thereof, e.g. carbamates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/30Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D263/32Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon 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
    • C07D295/16Heterocyclic 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 acylated on ring nitrogen atoms
    • C07D295/18Heterocyclic 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 acylated on ring nitrogen atoms by radicals derived from carboxylic acids, or sulfur or nitrogen analogues thereof
    • 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
    • C07D295/16Heterocyclic 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 acylated on ring nitrogen atoms
    • C07D295/18Heterocyclic 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 acylated on ring nitrogen atoms by radicals derived from carboxylic acids, or sulfur or nitrogen analogues thereof
    • C07D295/182Radicals derived from carboxylic acids
    • C07D295/185Radicals derived from carboxylic acids from aliphatic carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/38Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D307/52Radicals substituted by nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/06Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to the ring carbon atoms
    • C07D333/14Radicals substituted by singly bound hetero atoms other than halogen
    • C07D333/20Radicals substituted by singly bound hetero atoms other than halogen by nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic 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
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • 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
    • C07D417/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D471/10Spiro-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
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/06Systems containing only non-condensed rings with a five-membered ring
    • C07C2601/08Systems containing only non-condensed rings with a five-membered ring the ring being saturated

Definitions

  • This invention relates generally to tartaric acid functional compounds that can inhibit matrix metalloproteinases (MMPs), a disintegrin and metalloproteases (ADAMs) and/or tumor necrosis factor alpha - converting enzyme (TACE) and in so doing prevent the release of tumor necrosis factor alpha (TNF- ⁇ ), pharmaceutical compositions comprising such compounds, and methods of treatment using such compounds.
  • MMPs matrix metalloproteinases
  • ADAMs disintegrin and metalloproteases
  • TACE tumor necrosis factor alpha - converting enzyme
  • Osteo- and rheumatoid arthritis are destructive diseases of articular cartilage characterized by localized erosion of the cartilage surface. Findings have shown that articular cartilage from the femoral heads of patients with OA, for example, had a reduced incorporation of radiolabeled sulfate over controls, suggesting that there must be an enhanced rate of cartilage degradation in OA (Mankin et al. J. Bone Joint Surg. 52A (1970) 424-434).
  • MMPs Metalloproteases
  • these catabolic enzymes are tightly regulated at the level of their synthesis as well as at their level of extracellular activity through the action of specific inhibitors, such as alpha-2-macroglobulins and TIMPs (tissue inhibitor of MPs), which form inactive complexes with the MMP's.
  • specific inhibitors such as alpha-2-macroglobulins and TIMPs (tissue inhibitor of MPs), which form inactive complexes with the MMP's.
  • Tumor necrosis factor alpha is a cell-associated cytokine that is processed from a 26 kDa precursor form to a 17 kd active form. See Black R.A. "Tumor necrosis factor-alpha converting enzyme” lnt J Biochem Cell Biol. 2002 Jan;34(1):1-5 and Moss ML, White JM, Lambert MH, Andrews RC.'TACE and other ADAM proteases as targets for drug discovery" Drug Discov Today. 2001 Apr 1 ;6(8):417-426, each of which is incorporated by reference herein.
  • TNF- ⁇ has been shown to play a pivotal role in immune and inflammatory responses. Inappropriate or over-expression of TNF- ⁇ is a hallmark of a number of diseases, including RA, Crohn's disease, multiple sclerosis, psoriasis and sepsis. Inhibition of TNF- ⁇ production has been shown to be beneficial in many preclinical models of inflammatory disease, making inhibition of TNF- ⁇ production or signaling an appealing target for the development of novel anti-inflammatory drugs.
  • TNF- ⁇ is a primary mediator in humans and animals of inflammation, fever and acute phase responses, similar to those observed during acute infection and shock. Excess TNF- ⁇ has been shown to be lethal. Blocking the effects of TNF- ⁇ with specific antibodies can be beneficial in a variety of conditions, including autoimmune diseases such as RA (Feldman et al, Lancet, (1994) 344, 1105), non-insulin dependent diabetes mellitus (Lohmander L. S. et al., Arthritis Rheum. 36 (1993) 1214-22) and Crohn's disease (Macdonald T. et al., Clin. Exp. Immunol. 81 (1990) 301 ).
  • RA Paindman et al, Lancet, (1994) 344, 1105
  • non-insulin dependent diabetes mellitus Lihmander L. S. et al., Arthritis Rheum. 36 (1993) 1214-22
  • Crohn's disease Macdonald T.
  • TNF- ⁇ Compounds that inhibit the production of TNF- ⁇ are therefore of therapeutic importance for the treatment of inflammatory disorders. Recently it has been shown that metalloproteases, such as TACE, are capable of converting TNF- ⁇ from its inactive to active form (Gearing et al Nature, 1994, 370, 555). Since excessive TNF- ⁇ production has been noted in several disease conditions also characterized by MMP-mediated tissue degradation, compounds which inhibit both MMPs and TNF- ⁇ production may also have a particular advantage in diseases where both mechanisms are involved.
  • TACE is a member of the ADAM family of type I membrane proteins and mediates the ectodomain shedding of various membrane-anchored signaling and adhesion proteins. TACE has become increasingly important in the study of several diseases, including inflammatory disease, because of its role in cleaving TNF- ⁇ from its "stalk" sequence and thus releasing the soluble form of the TNF- ⁇ protein (Black R.A. Int J Biochem Cell Biol. 2002 34,1-5).
  • TNF- ⁇ MMPs, ADAMs, TACE, and TNF- ⁇
  • the inhibition of TNF- ⁇ , TACE and or other MMPs can prevent the degradation of cartilage by these enzymes, thereby alleviating the pathological conditions of OA and RA as well as many other auto-immune diseases.
  • the present invention provides a novel class of compounds as inhibitors of TACE, the production of TNF- ⁇ , MMPs, ADAMs or any combination thereof, methods of preparing such compounds, pharmaceutical compositions comprising one or more such compounds, methods of preparing pharmaceutical formulations comprising one or more such compounds, and methods of treatment, prevention, inhibition or amelioration of one or more diseases associated with TACE, TNF- ⁇ , MMPs, ADAMs or any combination thereof using such compounds or pharmaceutical compositions.
  • the present application discloses a compound, or pharmaceutically acceptable salts or solvates of said compound, said compound having the general structure shown in formula (I):
  • A is selected from the group consisting of:
  • J is selected from the group consisting of: O, S, and NR 5 ;
  • E is selected from the group consisting of: O, S, and NR 5 ;
  • T O or S
  • R 1 and R 2 are the same or different, each being independently selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, and heteroaryl; or alternatively R 1 and R 2 , taken together with the N to which R 1 and R 2 are shown attached, represent a 4-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein each of said alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, heteroaryl and 4-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties below;
  • R 10 is selected from the group consisting of H, alkyl, and fluoroalkyl
  • R 20 is selected from the group consisting of H, alkyl, and fluoroalkyl
  • R 30 is H or alkyl, or alternatively R 30 and R 40 taken together with the N to which R 40 is shown attached to in Formula I, are joined to form a 4-7 membered heterocylic ring, wherein said heterocylic ring is unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties below;
  • R 40 is H or alkyl
  • R 50 is H or alkyl
  • W is -(CR 13 2 )n-, wherein n is 0 to 5 or a covalent bond, or alternatively two R 3 groups can fuse to form a 3-8 membered cycloalkyl, wherein said 3-8 membered cycloalkyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 6 moieties below;
  • X is absent or present, and if present X is selected from the group consisting of a covalent bond, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties below;
  • Y is absent or present, and if present Y is selected from the group consisting of a covalent bond, -[C(R 6 ) 2 ] n - wherein n is 1 to 2, -0-, -S-, -NR 1 -, -SO v - wherein v is 1 to 2, -SO n (CR 6 2) p - wherein n is 1 or 2 and p is 1 to 4, -0(CR 6 2 ) q - or -(CR 6 2 ) q O- wherein q is 1 to 4, -N(R 7 )S(0) n - or -S(0) n N(R 7 )- wherein n is 1 or 2, and -N(R 7 )C(0)- or -C(0)N(R 7 )-;
  • Z is selected from the group consisting of H, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, said cycloalkyl, heterocyclyl, aryl, and heteroaryl being optionally fused with aryl, heterocyclyl, heteroaryl or cycloalky; wherein each of said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties below;
  • R 5 is selected from the group consisting of hydrogen, alkyl, and alkylaryl; each R 6 is the same or different and is independently selected from the group consisting of hydrogen, halogen, -SR ⁇ , -S(0) q R ⁇ 5 wherein q is 1 to 2, alkyl, cycloalkyl, heterocyclyl, alkoxyl, hydroxy, nitro, cyano, amino, alkenyl, alkynyl, arylalkyl, aminocarbonyl, alkylcarbonyl, and alkoxycarbonyl; each R 7 is the same or different and is independently selected from the group consisting of hydrogen, alkyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, alkenyl, alkynyl, arylalkyl, alkylcarbonyl, and alkoxycarbonyl, wherein each of the aryl, heteroaryl and heterocyclyl can be unsubstituted or optionally independently substituted with one or more moieties
  • R 13 is the same or different and is independently selected from the group consisting of hydrogen, halogen, -OH, -OR 14 , alkyl, cycloalkyl, heterocyclyl, alkenyl, alkynyl, alkylaryl, alkylamino, and alkylcarbonyl;
  • alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkenyl and alkynyl are independently unsubstituted or substituted by 1 to 5 groups independently selected from the group consisting of alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, halo, -CF3,
  • each R , R and R are independently selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, or alternatively R 15 and R 16 taken together with the N to which they are shown attached, are joined to form a 4-8 membered heterocylic ring, wherein said 4-8 membered cycloalkyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 75 moieties below; each R 75 is independently selected from the group consisting of alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkenyl and alkynyl, and wherein each of the alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkenyl and alkyn
  • each R 19 is independently selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl.
  • the compounds of Formula I can be useful as inhibitors and may be useful in the treatment and prevention of diseases associated with TACE, TNF- ⁇ , MMPs, ADAMs or any combination thereof.
  • the present invention provides a novel class of inhibitors of TACE, the production of TNF- ⁇ , MMPs, ADAMs or any combination thereof, pharmaceutical compositions containing one or more of the compounds, methods of preparing pharmaceutical formulations comprising one or more such compounds, and methods of treatment, prevention or amelioration of one or more of the symptoms of inflammation.
  • the present invention provides compounds which are represented by structural Formula (I) above or a pharmaceutically acceptable salt, solvate or isomer thereof, wherein the various moieties are as described above.
  • R 1 and R 2 are the same or different, each being independently selected from the group consisting of alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, and heteroaryl; or alternatively R 1 and R 2 , taken together with the N to which R 1 and R 2 are shown attached, represent a 4-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein each of said alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, heteroaryl and 4-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 .
  • R 1 and R 2 are the same or different, each being independently selected from the group consisting of H, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, and heteroaryl; or alternatively R 1 and R 2 , taken together with "the N to which R 1 and R 2 are shown attached, represent a 4-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein each of said alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, heteroaryl and 4-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 .
  • A is selected from the group consisting of:
  • R 1 and R 2 are shown attached, represent a 4-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R 70 , or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 4-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties.
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • R 1 and R 2 are the same or different, each being independently selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, and heteroaryl; wherein said alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl and heteroaryl can be unsubstituted or optionally independently substituted with one or more moieties which can e-the.same or.different.-each-moiety-being independently selected from the group of R 70 moieties.
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • R ⁇ R 2 2 or R ⁇ R 2 2 wherein R 1 and R 2 are the same or different, each being independently selected from the group consisting of alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, and heteroaryl; wherein said alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl and heteroaryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties.
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • R 1 and R 2 are the same or different, each being independently selected from the group consisting of H, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, and heteroaryl; wherein said alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl and heteroaryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties.
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • A is wherein each x idependently is 0 to 4.
  • A is selected from the group consisting of:
  • R 2 taken together with the N to which R 1 and R 2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R 70 , or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties; wherein a ring is formed from -NR 1 R 2 and said ring is
  • each R 9 is a substituent for H where indicated and can be the same or different, each being independently selected from the group consisting of -OH,
  • R 11 and R 12 taken together with the carbon to which each R 11 and R 12 are shown attached are fused heteroaryl or fused cycloalkyl, wherein said fused heteroaryl and fused cycloalkyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties; and x is 0 to 4, and when x is greater than 1 , each R 9 moiety can be the same or different, each moiety being independently selected from the group of R 9 moieties.
  • A is selected from the group consisting of:
  • R 1 and R 2 taken together with the N to which R and R 2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R 70 , or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties; wherein a ring is formed from -NR 1 R 2 and said ring is
  • each R 9 is a substituent for H where indicated and can be the same or different, each being independently selected from the group consisting of -OH, -OR 14 , -C(0)0R15 -C(0)N(R15)(R16) ) a j ky
  • G is selected from the group consisting of CH 2 , NR 7 , O, S, or SO 2 .
  • A is selected from the group consisting of:
  • R and R taken together with the N to which R 1 and R 2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R 70 , or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties; a ring is formed from -NR 1 R 2 and said ring is selected from the group consisting of
  • each R 70 moiety can be the same or different, each moiety being independently selected from the group of R 70 moieties; and each R 18 is the same or different and is independently H or alkyl.
  • A is selected from the group consisting of:
  • R and R taken together with the N to which R 1 and R 2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R 70 , or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties; a ring is formed from -NR 1 R 2 and said ring is selected from the group consisting of
  • each R 70 moiety can be the same or different, each moiety being independently selected from the group of R 70 moieties.
  • A is selected from the group consisting of:
  • R 1 and R 2 taken together with the N to which R 1 and R 2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R 70 , or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties; a ring is formed from -NR 1 R 2 and said ring is
  • each R 9 is a substituent for H where indicated and can be the same or different, each being independently selected from the group consisting of -OH,
  • J is O.
  • E is O.
  • R 10 is H or alkyl.
  • R 20 , R 30 , R 40 , and R 50 are all the same and are
  • R 6 is H or alkyl.
  • R 6 is H.
  • R 13 is H or alkyl.
  • R 3 is H or -CH 3 .
  • A is selected from the group consisting of
  • R 1 and R 2 taken together with the N to which R 1 and R 2 are shown attached, represent a 4-6 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R 70 , wherein R 70 is aryl.
  • Y is a covalent bond or -[C(R 6 ) 2 ] n - wherein n is 1 to 2, -CH 2 -.
  • Y is selected from the group consisting of a covalent bond, -CH 2 -, -C(H)(OH)-, -C(O)- and -0-.
  • Y is a covalent bond or -CH 2 -.
  • Y is a covalent bond.
  • W is -(CR 13 ) n -, in which n is 0-5.
  • W is -(CR 13 2 ) n -, in which n is 1-5 and each R 13 is H or alkyl. In another embodiment, W is selected from the group consisting of - CH 2 -, -C(H)(CH 3 )-, -C(CH 3 ) 2 - and -CH 2 CH 2 -.
  • W is -CH 2 -.
  • W is -C(H)(CH 3 )-.
  • X is selected from the group consisting of alkyl, aryl, heterocyclyl and heteroaryl.
  • X is aryl
  • X is heteroaryl
  • X is selected from the group consisting of phenyl, azetidinyl, pyrrolidinyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl.
  • X is selected from the group consisting of phenyl, pyridinyl and piperidinyl.
  • X is selected from the group consisting of
  • each R 70 moiety can be the same or different, each moiety being independently selected from the group of R 70 moieties.
  • X is the moiety enclosed by ⁇ and W and Y are shown only to indicate which end of the X is attached to W and which end to Y. Similar depictions occur throughout this application with similar connotations.
  • X is selected from the group consisting of
  • each R 70 moiety can be the same or different, each moiety being independently selected from the group of R 70 moieties.
  • Z is selected from the group consisting of H, aryl and heteroaryl.
  • Z is selected from the group consisting of H, phenyl, indolyl, benzimidazolyl, pyrazolyl, thienyl, pyridinyl, thiazolyl, thiadiazolyl, imidazolyl, pyrrolidinyl, pyrazinyl, triazolyl, tetrazolyl and tetrazinyl, wherein said phenyl, indolyl, benzimidazolyl, pyrazolyl, thienyl, pyridinyl, thiazolyl, thiadiazolyl, imidazolyl, pyrrolidinyl, pyrazinyl, triazolyl, tetrazolyl and tetrazinyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group consisting of R 70 moieties.
  • Z is selected from the group consisting of
  • each R 70 moiety can be the same or different, each moiety being independently selected from the group of R 70 moieties.
  • Z is phenyl
  • Z is a phenyl substituted with at least one substituent selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy, amino and tetrazole.
  • Z is thienyl.
  • Z is a thienyl substituted with at least one substituent selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy, amino and tetrazole.
  • Z is pyrazolyl
  • Z is a pyrazolyl substituted with at least one substituent selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy, amino and tetrazole.
  • Z is pyridinyl
  • Z is a pyridinyl substituted with at least one substituent selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy, amino and tetrazole.
  • Z is imidazolyl
  • Z is an imidazolyl substituted with at least one substituent selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy, amino and tetrazole.
  • R 1 and R 2 taken together with the N to which R 1 and R 2 are shown attached, represent a 4-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R 70 , or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 4-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties; E, J and T are the same and are O; R 10 is H or alkyl; R 20 , R 30 , R 40 , and R 50 are the same and are H; W is -(CR 13 2 ) n -,wherein n is 0 to 5; X is selected from the group consisting of aryl, heteroaryl and heterocyclyl; Y is selected from the group consisting of a
  • A is:
  • each R 9 is a substituent for H where indicated and can be the same or different, each being independently selected from the group consisting of -OH,
  • -OR 14 -C(0)OR 1 5, -C(0)N(R 5 )(R 16 ), alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl, wherein said alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties; and x is 0 to 4, and when x is greater than 1 , each R 9 moiety can be the same or different, each moiety being independently selected from the group of R 9 moieties;
  • R 10 is H or alkyl
  • R 20 , R 30 , R 40 , and R 50 are the same and are H
  • W is -(CR 13 2 ) n -,wherein n is 0 to 5
  • X is selected from the group consisting of aryl, heteroaryl and heterocyclyl
  • Y is selected from the group consisting of a covalent bond, -[C(R 6 ) ] n - wherein n is 1 to 2, - 0-, -S-, and -NR 1 -
  • Z is selected from the group consisting of
  • A is: O
  • R ⁇ wherein R and R 2 , taken together with the N to which R 1 and R 2 are shown attached, represent a 4-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R 70 , or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 4-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties; E, J and T are the same and are O; R 10 is H or alkyl; R 20 , R 30 , R 40 , and R 50 are the same and are H; W is -(CR 13 2 ) n -,wherein n is 0 to 5; X is selected from the group consisting of aryl, heteroaryl and heterocyclyl; Y is selected from the group consisting of
  • A is: O
  • R1 R R 2 wherein R 1 and R 2 , taken together with the N to which R 1 and R 2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R 70 , or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties; wherein E, J and T are the same and are O; R 10 is H or alkyl; R 20 , R 30 , R 40 , and R 50 are the same and are H; W is -(CR 6 ) n -, wherein n is 0 to 5; X is selected from the group consisting of phenyl, piperidinyl, pyridinyl,
  • R and R taken together with the N to which R and R 2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R 70 , or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties;
  • R 10 is H or alkyl
  • R 20 , R 30 , R 40 , and R 50 are the same and are H
  • W is -(CR 6 2 ) n -, wherein n is 0 to 5
  • X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl and pyrazolyl
  • Y is selected from the group consisting of 0-, -S-, and -NR 1 -
  • Z is selected from the group consisting of
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • R 1 and R 2 taken together with the N to which R 1 and R 2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R 70 , or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties; wherein E, J and T are the same and are O; R 10 is H or alkyl; R 20 , R 30 , R 40 , and R 50 are the same and are H; W is -(CR 6 2) n -, wherein n is 0 to 5; X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl,
  • R 1 and R 2 taken together with the N to which R 1 and R 2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R 70 , or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties; wherein E, J and T are the same and are O; R 10 is H or alkyl; R 20 , R 30 , R 40 , and R 50 are the same and are H; W is -(CR 13 2 ) n - wherein n is 1 or 2; X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl,
  • A is o
  • R R 2 • wherein R 1 and R 2 , taken together with the N to which R 1 and R 2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R 70 , or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R 70 moieties; wherein E, J and T are the same and
  • R 1U is H or alkyl
  • R ", R ou , R 4U , and R ou are the same and are H
  • W is (CR )13 2 )rr wherein n is 1 or 2
  • X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl and pyrazolyl
  • Y is a covalent bond and Z is selected from the group consisting of
  • X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl, and pyrazolyl.
  • X is selected from the group consisting of phenyl, piperidinyl, thienyl, and pyridinyl and Y is selected from the group consisting of a covalent bond, -[C(R 6 ) 2 ] n - wherein n is 1 to 2 and -0-.
  • X is phenyl and Y is selected from the group consisting of a covalent bond, -CH 2 - and -0-.
  • X is piperidinyl
  • Y is a covalent bond
  • Z is aryl or heteroaryl having two substituents which can be the same or different, each moiety being independently selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy and amino.
  • W is selected from the group consisting of - CH 2 -, -C(H)CH 3 -, -C(CH 3 ) 2 - and Y is a covalent bond.
  • W is -CH 2 - and Y is -CH 2 -.
  • Z is selected from the group consisting of
  • Another embodiment of the invention discloses the compounds shown in Table 1 below.
  • Patient includes both human and animals.
  • “Mammal” means humans and other mammalian animals.
  • Alkyl means an aliphatic hydrocarbon group which may be straight or branched and comprising about 1 to about 20 carbon atoms in the chain. Preferred alkyl groups contain about 1 to about 12 carbon atoms in the chain. More preferred alkyl groups contain about 1 to about 6 carbon atoms in the chain. Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl, are attached to a linear alkyl chain. "Lower alkyl” means a group having about 1 to about 6 carbon atoms in the chain which may be straight or branched.
  • substituted alkyl means that the alkyl group may be substituted by one or more substituents which may be the same or different, each substituent being independently selected from the group consisting of halo, alkyl, aryl, cycloalkyl, cyano, hydroxy, alkoxy, alkylthio, amino, -NH(alkyl), -NH(cycloalkyl), -N(alkyl) 2) carboxy and -C(0)0-alkyl.
  • suitable alkyl groups include methyl, ethyl, n-propyl, isopropyl and t-butyl.
  • Fluoroalkyl means an alkyl group in which alkyl is as previously described wherein one or more hydrogens are replaced with fluorine atoms.
  • Alkenyl means an aliphatic hydrocarbon group containing at least one carbon-carbon double bond and which may be straight or branched and comprising about 2 to about 15 carbon atoms in the chain.
  • Preferred alkenyl groups have about 2 to about 12 carbon atoms in the chain; and more preferably about 2 to about 6 carbon atoms in the chain.
  • Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl, are attached to a linear alkenyl chain.
  • “Lower alkenyl” means about 2 to about 6 carbon atoms in the chain which may be straight or branched.
  • suitable alkenyl groups include ethenyl, propenyl, n-butenyl, 3-methylbut-2- enyl, n-pentenyl, octenyl and decenyl.
  • Alkynyl means an aliphatic hydrocarbon group containing at least one carbon-carbon triple bond and which may be straight or branched and comprising about 2 to about 15 carbon atoms in the chain.
  • Preferred alkynyl groups have about 2 to about 12 carbon atoms in the chain; and more preferably about 2 to about 4 carbon atoms in the chain.
  • Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl, are attached to a linear alkynyl chain.
  • “Lower alkynyl” means about 2 to about 6 carbon atoms in the chain which may be straight or branched.
  • Non-limiting examples of suitable alkynyl groups include ethynyl, propynyl, 2-butynyl and 3- methylbutynyl.
  • substituted alkynyl means that the alkynyl group may be substituted by one or more substituents which may be the same or different, each substituent being independently selected from the group consisting of alkyl, aryl and cycloalkyl.
  • Aryl means an aromatic monocyclic or multicyclic ring system comprising about 6 to about 14 carbon atoms, preferably about 6 to about 10 carbon atoms.
  • the aryl group can be optionally substituted with one or more "ring system substituents" which may be the same or different, and are as defined herein.
  • suitable aryl groups include phenyl and naphthyl.
  • Heteroaryl means an aromatic monocyclic or multicyclic ring system comprising about 5 to about 14 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the ring atoms is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination. Preferred heteroaryls contain about 5 to about 6 ring atoms.
  • the "heteroaryl” can be optionally substituted by one or more "ring system substituents" which may be the same or different, and are as defined herein.
  • the prefix aza, oxa or thia before the heteroaryl root name means that at least a nitrogen, oxygen or sulfur atom respectively, is present as a ring atom.
  • a nitrogen atom of a heteroaryl can be optionally oxidized to the corresponding N-oxide.
  • suitable heteroaryls include pyridyl, pyrazinyl, furanyl, thienyl, pyrimidinyl, pyridone (including N-substituted pyridones), isoxazolyl, isothiazolyl, oxazolyl, thiazolyl, pyrazolyl, furazanyl, pyrrolyl, pyrazolyl, triazolyl, 1 ,2,4-thiadiazolyl, pyrazinyl, pyridazinyl, quinoxalinyl, phthalazinyl, oxindolyl, imidazo[1 ,2-a]pyridinyI, imidazo[2,1-b]thiazolyl, benzofurazanyl, indolyl, azaindolyl, benzimidazolyl,
  • Aralkyl or “arylalkyl” means an aryl-alkyl- group in which the aryl and alkyl are as previously described. Preferred aralkyls comprise a lower alkyl group. Non-limiting examples of suitable aralkyl groups include benzyl, 2- phenethyl and naphthalenylmethyl. The bond to the parent moiety is through the alkyl.
  • Alkylaryl means an alkyl-aryl- group in which the alkyl and aryl are as previously described. Preferred alkylaryls comprise a lower alkyl group. Non- limiting example of a suitable alkylaryl group is tolyl. The bond to the parent moiety is through the aryl.
  • Cycloalkyl means a non-aromatic mono- or multicyclic ring system comprising about 3 to about 10 carbon atoms, preferably about 5 to about 10 carbon atoms. Preferred cycloalkyl rings contain about 5 to about 7 ring atoms.
  • the cycloalkyl can be optionally substituted with one or more "ring system substituents" which may be the same or different, and are as defined above.
  • suitable monocyclic cycloalkyls include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl and the like.
  • Non-limiting examples of suitable multicyclic cycloalkyls include 1 -decalinyl, norbornyl, adamantyl and the like, as well as partially saturated species such as, for example, indanyl, tetrahydronaphthyl and the like.
  • Halogen means fluorine, chlorine, bromine, or iodine. Preferred are fluorine, chlorine and bromine.
  • Ring system substituent means a substituent attached to an aromatic or non-aromatic ring system which, for example, replaces an available hydrogen on the ring system.
  • Ring system substituents may be the same or different, each being independently selected from the group consisting of alkyl, alkenyl, alkynyl, aryl, heteroaryl, aralkyl, alkylaryl, heteroaralkyl, heteroarylalkenyl, heteroarylalkynyl, alkylheteroaryl, hydroxy, hydroxyalkyl, alkoxy, aryloxy, aralkoxy, acyl, aroyl, halo, nitro, cyano, carboxy, alkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, alkylthio, arylthio, heteroarylthio, aralkylthio
  • Ring system substituent may also mean a single moiety which simultaneously replaces two available hydrogens on two adjacent carbon atoms (one H on each carbon) on a ring system.
  • Examples of such moiety are methylene dioxy, ethylenedioxy, -C(CH 3 ) 2 - and the like which form moieties such as, for example:
  • Heterocyclyl means a non-aromatic saturated monocyclic or multicyclic ring system comprising about 3 to about 10 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the atoms in the ring system is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination. There are no adjacent oxygen and/or sulfur atoms present in the ring system.
  • Preferred heterocyclyls contain about 5 to about 6 ring atoms.
  • the prefix aza, oxa or thia before the heterocyclyl root name means that at least a nitrogen, oxygen or sulfur atom respectively is present as a ring atom.
  • Any -NH in a heterocyclyl ring may exist protected such as, for example, as an -N(Boc), -N(CBz), -N(Tos) group and the like; such protections are also considered part of this invention.
  • the heterocyclyl can be optionally substituted by one or more "ring system substituents" which may be the same or different, and are as defined herein.
  • the nitrogen or sulfur atom of the heterocyclyl can be optionally oxidized to the corresponding N-oxide, S-oxide or S,S-dioxide.
  • Non-limiting examples of suitable monocyclic heterocyclyl rings include piperidyl, pyrrolidinyl, piperazinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, 1 ,4-dioxanyl, tetrahydrofuranyl, tetrahydrothiophenyl, lactam, lactone, and the like.
  • hetero-atom containing ring systems of this invention there are no hydroxyl groups on carbon atoms adjacent to a N, O or S, as well as there are no N or S groups on carbon adjacent to another heteroatom.
  • N, O or S there are no hydroxyl groups on carbon atoms adjacent to a N, O or S, as well as there are no N or S groups on carbon adjacent to another heteroatom.
  • Alkynylalkyl means an alkynyl-alkyl- group in which the alkynyl and alkyl are as previously described. Preferred alkynylalkyls contain a lower alkynyl and a lower alkyl group. The bond to the parent moiety is through the alkyl. Non-limiting examples of suitable alkynylalkyl groups include propargylmethyl. "Heteroaralkyl” means a heteroaryl-alkyl- group in which the heteroaryl and alkyl are as previously described. Preferred heteroaralkyls contain a lower alkyl group. Non-limiting examples of suitable aralkyl groups include pyridylmethyl, and quinolin-3-ylmethyl. The bond to the parent moiety is through the alkyl.
  • Hydroxyalkyl means a HO-alkyl- group in which alkyl is as previously defined. Preferred hydroxyalkyls contain lower alkyl. Non-limiting examples of suitable hydroxyalkyl groups include hydroxymethyl and 2-hydroxyethyl.
  • acyl means an H-C(O)-, alkyl-C(O)- or cycloalkyl-C(O)-, group in which the various groups are as previously described.
  • the bond to the parent moiety is through the carbonyl.
  • Preferred acyls contain a lower alkyl.
  • suitable acyl groups include formyl, acetyl and propanoyl.
  • Aroyl means an aryl-C(O)- group in which the aryl group is as previously described. The bond to the parent moiety is through the carbonyl.
  • suitable groups include benzoyl and 1- naphthoyl.
  • Alkoxy means an alkyl-O- group in which the alkyl group is as previously described.
  • suitable alkoxy groups include methoxy, ethoxy, n-propoxy, isopropoxy and n-butoxy.
  • the bond to the parent moiety is through the ether oxygen.
  • Aryloxy means an aryl-O- group in which the aryl group is as previously described.
  • suitable aryloxy groups include phenoxy and naphthoxy.
  • the bond to the parent moiety is through the ether oxygen.
  • Alkyloxy means an aralkyl-O- group in which the aralkyl group is as previously described.
  • Alkylthio means an alkyl-S- group in which the alkyl group is as previously described.
  • suitable alkylthio groups include methylthio and ethylthio.
  • the bond to the parent moiety is through the sulfur.
  • Arylthio means an aryl-S- group in which the aryl group is as previously described.
  • suitable arylthio groups include phenylthio and naphthylthio.
  • the bond to the parent moiety is through the sulfur.
  • Alkylthio means an aralkyl-S- group in which the aralkyl group is as previously described.
  • Non-limiting example of a suitable aralkylthio group is benzylthio.
  • the bond to the parent moiety is through the sulfur.
  • Alkoxycarbonyl means an alkyl-O-CO- group.
  • suitable alkoxycarbonyl groups include methoxycarbonyl and ethoxycarbonyl.
  • the bond to the parent moiety is through the carbonyl.
  • Aryloxycarbonyl means an aryl-O-C(O)- group.
  • suitable aryloxycarbonyl groups include phenoxycarbonyl and naphthoxycarbonyl.
  • the bond to the parent moiety is through the carbonyl.
  • Alkoxycarbonyl means an aralkyl-O-C(O)- group.
  • a suitable aralkoxycarbonyl group is benzyloxycarbonyl.
  • the bond to the parent moiety is through the carbonyl.
  • Alkylsulfonyl means an alkyl-S(0 2 )- group. Preferred groups are those in which the alkyl group is lower alkyl. The bond to the parent moiety is through the sulfonyl.
  • Arylsulfonyl means an aryl-S(0 2 )- group. The bond to the parent moiety is through the sulfonyl.
  • substituted means that one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • stable compound' or “stable structure” is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • isolated or “in isolated form” for a compound refers to the physical state of said compound after being isolated from a synthetic process or natural source or combination thereof.
  • purified or “in purified form” for a compound refers to the physical state of said compound after being obtained from a purification process or processes described herein or well known to the skilled artisan, in sufficient purity to be characterizable by standard analytical techniques described herein or well known to the skilled artisan.
  • protecting groups When a functional group in a compound is termed "protected", this means that the group is in modified form to preclude undesired side reactions at the protected site when the compound is subjected to a reaction. Suitable protecting groups will be recognized by those with ordinary skill in the art as well as by reference to standard textbooks such as, for example, T. W. Greene et al, Protective Groups in organic Synthesis (1991 ), Wiley, New York.
  • variable e.g., aryl, heterocycle, R 2 , etc.
  • its definition on each occurrence is independent of its definition at every other occurrence.
  • composition is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • Prodrugs and solvates of the compounds of the invention are also contemplated herein.
  • the term "prodrug”, as employed herein, denotes a compound that is a drug precursor which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of Formula I or a salt and/or solvate thereof.
  • a discussion of prodrugs is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems (1987) 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, (1987) Edward B. Roche, ed., American Pharmaceutical Association and Pergamon Press, both of which are incorporated herein by reference thereto.
  • Solvate means a physical association of a compound of this invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. “Solvate” encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates, and the like. "Hydrate” is a solvate wherein the solvent molecule is H 2 0.
  • Effective amount or “therapeutically effective amount” is meant to describe an amount of compound or a composition of the present invention effective in inhibiting the CDK(s) and thus producing the desired therapeutic, ameliorative, inhibitory or preventative effect.
  • the compounds of Formula I can form salts which are also within the scope of this invention.
  • Reference to a compound of Formula I herein is understood to include reference to salts thereof, unless otherwise indicated.
  • the term "salt(s)", as employed herein, denotes acidic salts formed with inorganic and/or organic acids, as well as basic salts formed with inorganic and/or organic bases.
  • zwitterions inner salts may be formed and are included within the term "salt(s)" as used herein.
  • Salts of the compounds of the Formula I may be formed, for example, by reacting a compound of Formula I with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an -aqueous-medium-followed-by lyophilizationr
  • Exemplary acid addition salts include acetates, ascorbates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, fumarates, hydrochlorides, hydrobromides, hydroiodides, lactates, maleates, methanesulfonates, naphthalenesulfonates, nitrates, oxalates, phosphates, propionates, salicylates, succinates, sulfates, tartarates, thiocyanates, toluenesulfonates (also known as tosylates,) and the like.
  • Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as dicyclohexylamines, t-butyl amines, and salts with amino acids such as arginine, lysine and the like.
  • Basic nitrogen-containing groups may be quarternized with agents such as lower alkyl halides (e.g. methyl, ethyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g.
  • dimethyl, diethyl, and dibutyl sulfates dimethyl, diethyl, and dibutyl sulfates
  • long chain halides e.g. decyl, lauryl, and stearyl chlorides, bromides and iodides
  • aralkyl halides e.g. benzyl and phenethyl bromides
  • All stereoisomers for example, geometric isomers, optical isomers and the like
  • of the present compounds including those ofthe salts, solvates and prodrugs of the compounds as well as the salts and solvates of the prodrugs
  • those which may exist due to asymmetric carbons on various substituents including enantiomeric forms (which may exist even in the absence of asymmetric carbons), rotameric forms, atropisomers, and diastereomeric forms, are contemplated within the scope of this invention, as are positional isomers (such as, for example, 4-pyridyl and 3-pyridyl).
  • Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers.
  • the chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations.
  • the use of the terms "salt”, “solvate” "prodrug” and the like, is intended to equally apply to the salt, solvate and prodrug of enantiomers, stereoisomers, rotamers, tautomers, positional isomers, racemates or prodrugs of the inventive compounds.
  • the compounds according to the invention have pharmacological properties; in particular, the compounds of Formula I can be inhibitors of TACE, TNF- ⁇ , ADAM and/or MMP activity.
  • the invention provides a pharmaceutical composition comprising as an active ingredient at least one compound of formula 1.
  • the invention provides a pharmaceutical composition of formula 1 additionally comprising at least one pharmaceutically acceptable carrier.
  • the invention provides a method of treating disorders associated with TACE, TNF- ⁇ , MMPs, ADAMs or any combination thereof or any combination thereof, said method comprising administering to a patient in need of such treatment a pharmaceutical composition which comprises therapeutically effective amounts of at least one compound of formula 1.
  • the invention provides a use of a compound of ⁇ formula ⁇ for the ⁇ manufact ⁇ re " of a ⁇ medicament to treat disorders associated with TACE, TNF- ⁇ , MMPs, ADAMs or any combination thereof.
  • the compounds of Formula I can have anti-inflammatory activity and/or immunomodulatory activity and can be useful in the treatment of diseases including but not limited to septic shock, haemodynamic shock, sepsis syndrome, post ischaemic reperfusion injury, malaria, mycobacterial infection, meningitis, psoriasis, congestive heart failure, fibrotic diseases, cachexia, graft rejection, cancers such as cutaneous T-cell lymphoma, diseases involving angiogenesis, autoimmune diseases, skin inflammatory diseases, inflammatory bowel diseases such as Crohn's disease and colitis, OA and RA, ankylosing spondylitis, psoriatic arthritis, adult Still's disease, ureitis, Wegener's granulomatosis, Behcehe disease, Sjogren's syndrome, sarcoidosis, polymyositis, dermatomyositis, multiple sclerosis, sciatica, complex regional pain syndrome, radiation damage, hyperoxic alveolar injury, periodontal
  • the invention provides a method of preparing a pharmaceutical composition for treating the disorders associated with TACE, TNF- ⁇ , MMPs, ADAMs or any combination thereof, said method comprising bringing into intimate contact at least one compound of formula 1 and at least one pharmaceutically acceptable carrier.
  • the invention provides a compound of formula (I) exhibiting TACE, TNF- ⁇ , MMPs, ADAMs or any combination thereof inhibitory activity, including enantiomers, stereoisomers and tautomers of said compound, and pharmaceutically acceptable salts or solvates of said compound, said compound being selected from the compounds of structures listed below:
  • the compound is selected from the group consisting of:
  • the above preferred compounds have TACE Ki values of less than 20nM.
  • the compound is selected from the group consting of :
  • the above more preferred compounds have TACE Kj values of less than 5 nM.
  • the invention provides a pharmaceutical composition for treating disorders associated with TACE, TNF- ⁇ , MMP, ADAM or any combination thereof in a subject comprising, administering to the subject in need of such treatment a therapeutically effective amount of a compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
  • the invention provides a compound of formula 1 in purified form.
  • the invention provides a method of treating a condition or disease mediated by TACE, MMPs, TNF- ⁇ , aggrecanase (such as aggrecanase 1 or aggrecanase 2), or any combination thereof in a subject comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
  • the invention provides a method of treating a condition or disease selected from the group consisting of rheumatoid arthritis, osteoarthritis, periodontitis, gingivitis, corneal ulceration, solid tumor growth and tumor invasion by secondary metastases, neovascular glaucoma, inflammatory bowel disease, multiple sclerosis and psoriasis in a subject, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
  • the invention provides a method of treating a condition or disease selected from the group consisting of fever, cardiovascular conditions, hemorrhage, coagulation, cachexia, anorexia, alcoholism, acute phase response, acute infection, shock, graft versus host reaction, autoimmune disease and HIV infection in a subject comprising administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
  • the invention provides a method of treating a condition or disease selected from the group consisting of septic shock, haemodynamic shock, sepsis syndrome, post ischaemic reperfusion injury, malaria, mycobacterial infection, meningitis, psoriasis, congestive heart failure, fibrotic diseases, cachexia, graft rejection, cancers such as cutaneous T-cell lymphoma, diseases involving angiogenesis, autoimmune diseases, skin inflammatory diseases, inflammatory bowel diseases such as Crohn's disease and colitis, osteo and rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, adult Still's disease, ureitis, Wegener's granulomatosis, Behcehe disease, Sjogren's syndrome, sarcoidosis, polymyositis, dermatomyositis, multiple sclerosis, sciatica, complex regional pain syndrome, radiation damage, hyperoxic alveolar injury, periodontal disease, HIV
  • the invention provides a method of treating a condition or disease associated with COPD, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
  • the invention provides a method of treating a condition or disease associated with rheumatoid arthritis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
  • the invention provides a method of treating a condition or disease associated with Crohn's disease, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
  • the invention provides a method of treating a condition or disease associated with psoriasis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
  • the-invention provides- a- method of treating a condition or disease associated with ankylosing spondylitis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
  • the invention provides a method of treating a condition or disease associated with sciatica, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
  • the invention provides a method of treating a condition or disease associated with complex regional pain syndrome, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
  • the invention provides a method of treating a condition or disease associated with psoriatic arthritis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
  • the invention provides a method of treating a condition or disease associated with multiple sclerosis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof, in combination with a compound selected from the group consisting of Avonex®, Betaseron, Copaxone or other compounds indicated for the treatment of multiple sclerosis.
  • a compound of the present invention may be co- administered or used in combination with disease-modifying antirheumatic drugs (DMARDS) such as methotrexate, azathioprine, leflunomide, pencillinamine, gold salts, mycophenolate mofetil, cyclophosphamide and other similar drugs.
  • DARDS disease-modifying antirheumatic drugs
  • NSAIDS such as piroxicam, naproxen, indomethacin, ibuprofen and the like
  • COX-2 selective inhibitors Ts ⁇ ch as Vioxx® and Celebrex®
  • Jmmunosuppressives such as steroids, cyclosporin, Tacrolimus, rapamycin and the like
  • biological response modifiers BRMs
  • other anti-inflammatory agents such as p38 kinase inhibitors, PDE4 inhibitors, other chemically different TACE inhibitors, chemokine receptor antagonists, Thalidomide and other small molecule inhibitors of pro-inflammatory cytokine production.
  • a compound of the present invention may be co-administered or used in combination with an H1 antagonist for the treatment of seasonal allergic rhinitis and/or asthma.
  • H1 antagonists may be, for example, Claritin®, Clarinex®, Allegra®, or Zyrtec®.
  • the invention provides a method of treating a condition or disease mediated by TACE, MMPs, TNF- ⁇ , aggrecanase, or any combination thereof in a subject comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof in combination with a therapeutically effective amount of at least one medicament selected from the group consisting of disease modifying anti-rheumatic drugs (DMARDS), NSAIDs, COX-2 inhibitors, COX-1 inhibitors, immunosuppressives, biological response modifiers (BRMs), anti- infammatory agents and H1 antagonists.
  • DARDS disease modifying anti-rheumatic drugs
  • NSAIDs NSAIDs
  • COX-2 inhibitors COX-1 inhibitors
  • immunosuppressives biological response modifiers
  • BRMs biological response modifiers
  • the invention provides a method of treating a condition or disease selected from the group consisting of rheumatoid arthritis, osteoarthritis, periodontitis, gingivitis, corneal ulceration, solid tumor growth and tumor invasion by secondary metastases, neovascular glaucoma, inflammatory bowel disease, multiple sclerosis and psoriasis in a subject, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof in combination with a therapeutically effective amount of at least one medicament selected from the group consisting of DMARDS, NSAIDs, COX-2 inhibitors, COX-1 inhibitors, immunosuppressives, BRMs, anti-infammatory agents and H1 antagonists.
  • a condition or disease selected from the group consisting of rheumatoid arthritis, osteoarthritis, periodontitis, gingivitis, corneal ulceration, solid tumor growth and tumor
  • the invention provides a method of treating a condition or disease selected from the group consisting of septic shock, haemodynamic shock, sepsis syndrome, post ischaemic reperfusion injury, malaria, mycobacterial infection, meningitis, psoriasis, congestive heart failure, fibrotic diseases, cachexia, graft rejection, cancers such as cutaneous T-cell lymphoma, diseases involving angiogenesis, autoimmune diseases, skin inflammatory diseases, inflammatory bowel diseases such as Crohn's disease and colitis, osteo and rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, adult Still's disease, ureitis, Wegener's granulomatosis, Behcehe disease, Sjogren's syndrome, sarcoidosis, polymyositis, dermatomyositis, multiple sclerosis, sciatica, complex regional pain syndrome, radiation damage, hyperoxic alveolar injury, periodontal disease, HIV
  • the invention provides a method for treating RA comprising administering a compound of the formula I in combination with compound selected from the class consisting of a COX-2 inhibitor e.g. Celebrex® or Vioxx®; a COX-1 inhibitor e.g. Feldene®; an immunosuppressive e.g. methotrexate or cyclosporin; a steroid e.g. ⁇ - inethasonerand anti-TNF ⁇ " ⁇ cdmpound; eTg. ⁇ nbrel® or Remicade®; a PDE IV inhibitor, br other classes of compounds indicated for the treatment of RA.
  • a COX-2 inhibitor e.g. Celebrex® or Vioxx®
  • COX-1 inhibitor e.g. Feldene®
  • an immunosuppressive e.g. methotrexate or cyclosporin
  • a steroid e.g. ⁇ - inethasonerand anti-TNF ⁇ " ⁇ cdmpound;
  • the invention provides a method for treating multiple sclerosis comprising administering a compound of the formula I in combination with a compound selected from the group consisting of Avonex®, Betaseron, Copaxone or other compounds indicated for the treatment of multiple sclerosis.
  • TACE activity is determined by a kinetic assay measuring the rate of increase in fluorescent intensity generated by TACE catalyzed cleavage of an internally quenched peptide substrate (SPDL-3).
  • SPDL-3 internally quenched peptide substrate
  • the purified catalytic domain of recombinant human TACE rhTACEc, Residue 215 to 477 with two mutation (S266A and N452Q) and a 6xHis tail
  • rhTACEc recombinant human TACE
  • the substrate SPDL-3 is an internally quenched peptide (MCA-Pro-Leu-Ala-Gln-Ala-Val-Arg-Ser-Ser-Ser-Dpa-Arg-NH2), with its sequence derived from the pro-TNF cleavage site.
  • MCA is (7- Methoxycoumarin-4-yl)acetyl.
  • Dpa is N-3-(2,4-Dinitrophenyl)-L-2,3- diaminopropionyl.
  • a 50 ⁇ l assay mixture contains 20 mM HEPES, pH 7.3, 5 mM CaCI 2 , 100 ⁇ M ZnCI 2 , 2 % DMSO, 0.04% Methylcellulose, 30 ⁇ M SPDL-3, 70 pM rhTACEc and a test compound.
  • RhTACEc is pre-incubated with the testing compound for 90 min. at 25 °C. Reaction is started by addition of the substrate. The fluorescent intensity (excitation at 320 nm, emission at 405 nm) was measured every 45 seconds for 30 min. using a fluorospectrometer (GEMINI XS, Molecular Devices). Rate of enzymatic reaction is shown as Units per second. Effect of a test compound is shown as % of TACE activity in the absence of the compound.
  • TACE inhibitory activities for representative compounds are shown in the table below.
  • Kj values greater than 1000 nM (1 ⁇ M) are designated as D class.
  • Kj values between 100 nM (0.1 ⁇ M) and 1000 nM (1 ⁇ M) are designated as C class.
  • Kj values between 20 nM (0.02 ⁇ M) and 100 nM (0.1 ⁇ M) are designated as B class.
  • Kj values less than 20 nM (0.02 ⁇ M) are designated as A class.
  • compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be coated by the technique described in the U.S. Pat. Nos. 4,256,108; 4,166,452; and 4,265,874 to form osmotic therapeutic tablets for controlled release.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredients is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or a soft gelatin capsules where in the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • an oil medium for example peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example, heptadecaethylene-oxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example, polyethylene
  • the aqueous suspensions may also contain one or more preservatives, for example, ethyl or n-propyl, p- hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame.
  • preservatives for example, ethyl or n-propyl, p- hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example, arachis oil, olive oil, sesame oil or coconut oil, or in mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example, beeswax, hard paraffin or cetyl alcohol.
  • Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation.
  • These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, e.g., sweetening, flavoring and coloring agents, may also be present.
  • the pharmaceutical compositions of the invention may also be in the form of an oil-in-water emulsion.
  • the oily phase may be a vegetable oil, e.g., olive oil or arachis oil, or a mineral oil, e.g., liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring phosphatides, e.g., soy beans, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example, sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, e.g., polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • sweetening agents for example, glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, e.g., as a solution in 1 ,3-butane diol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials are cocoa butter and polyethylene glycols.
  • topical application For topical use, creams, ointments, jellies, solutions or suspensions, etc., containing the compound of The invention are employed. (For purposes of this application, topical application shall include mouthwashes and gargles.)
  • the compounds for the present invention can be administered in the intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • Compounds of the present invention may also be delivered as a suppository employing bases such as cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol.
  • the dosage regimen utilizing the compounds of the present invention is selected in accordance with a variety of factors including type, species, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound thereof employed.
  • a physician or veterinarian of ordinary skill can readily determine and prescribe the effective amount of the drug required to prevent, counter, arrest or reverse the progress of the condition.
  • Optimal precision in achieving concentration of drug within the range that yields efficacy without toxicity requires a regimen based on the kinetics of the drug's availability to target sites. This involves a consideration of the distribution, equilibrium, and elimination of a drug.
  • doses of the compound of Formula I useful in the method of the present invention range from 0.01 to 1000 mg per day. More preferably, dosages range from 0.1 to 1000 mg/day. Most preferably, dosages range from 0.1 to 500 mg/day.
  • the compositions are preferably provided in the form of tablets containing 0.01 to 1000 milligrams of the active ingredient, particularly 0.01 , 0.05, 0.1 , 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100 and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • An effective amount of the drug is ordinarily supplied at a dosage level of from about 0.0002 mg/kg to about 50 mg/kg of body weight per day. The range is more particularly from about 0.001 mg/kg to 1 mg/kg of body weight per day.
  • the active agent of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in dividend doses of two, three or four time daily.
  • the amount of active ingredient that may be combined with the carrier materials to produce single dosage form will vary depending upon the host treated and the particular mode of administration.
  • the compounds of the invention may be produced by processes known to those skilled in the art and as shown in the following reaction schemes and in the preparations and examples described below.
  • TPP Bis-(triphenylphosphine)palladium(ll) chloride
  • PdCI 2 (ddppf) Dichloro[1 ,1 '-bis(diphenylphosphino)ferrocene]palladium(ii) dichloride
  • Pd 2 (dba) 3 Tris(dibenzylideneacetone)dipalladium(0) PyBrOP Bromo-tris-pyrrolidino-phosphonium hexafluorophosphate
  • NMR spectra were acquired on a Mercuryplus 400 MHz NMR Spectrometer (Varian), using CDCI3 or DMSO-d6 as solvents.
  • LC-MS data was obtained using an Agilent 1100 Series LC/MSD (quadrupole, API-ES (Atmospheric Pressure Interface Electrospray)) with a capillary voltage set to 3500 V and running in positive mode.
  • Reported analytical HPLC (LC/MS) retention times were obtained using a C18 (150 x 4.6 mm) reverse-phase column eluting with a 5 or 10 minute gradient of 0.1 % trifluoroacetic acid in water to 95:5 acetonitrile:water at a flow rate of 3 mL/min.
  • the compounds of formula (I) may be produced by processes known to those skilled in the art and as shown in the following reaction schemes and in the preparations and examples described below. These preparations and examples should not be construed to limit the scope of the disclosure. Alternate mechanistic pathways and analogous structures may be apparent to those skilled in the art. Some of the compounds made by these processes are listed in Table 1. All kinds of isomeric forms of the compounds are considered to be within the scope of this invention.
  • Example 1 Two general routes exist for the synthesis of tartrate diamides from amines.
  • the first (Example 1 ) utilizes an acetonide-protected monoacid/monoester intermediate prepared from the commercially available acetonide dimethyl ester using a literature procedure (J. Am. Chem. Soc. 1978, 100, 4865-4872).
  • Example 1 or Example 2 could be used interchangeably, although Example 2 was found to be more preferred for compounds that contained functional groups that were unstable towards acidic deprotection conditions (such as 113).
  • amide bond coupling reagents were acceptable, including HATU, GDI, EDC, DCC/HOBt, PyBrOP, polymer supported CDI with HOBt, polymer supported carbodiimide, and polymer supported EDC (PS-EDC) with HOBt.
  • These coupling reagents could be used with a variety of bases, including triethylamine (TEA), diisopropylethylamine (DIEA), N-methyl morpholine, pyridine, dimethylaminopyridine (DMAP) and imidazole.
  • TAA triethylamine
  • DIEA diisopropylethylamine
  • DMAP dimethylaminopyridine
  • imidazole imidazole.
  • the excess amines in the peptide coupling steps were removed using liquid/liquid extraction or polymeric scavenging resins such as polymer supported isocyanate (PS-NCO) and/or polymer supported tosic acid (MP- TsOH). Unreacted acids could be removed using MP-carbonate resin or polymer resins containing basic functional groups such as trisamine (i.e. PS- trisamine), amberiite, or morpholine.
  • Peptide couplings were conducted in a variety of solvents, including DMF, THF, dioxane, acetonitrile, NMP, and DCM. These solvents can also be combined in various proportions to optimize the reaction conditions.
  • PFP pentafluorophenyl
  • Cleavage (saponification) of the intermediate methyl ester could be accomplished under a variety of well-known conditions; including: a slight excess (1.1-3 equivalents) of base, KOH in methanol, LiOH in THF/water, LiOH in methanol/water, and NaOH/THF/MeOH/water. Removal of the acetonide protecting group could be afforded using a variety of acidic conditions, including TFA:water combinations (such as 80:20).
  • (+)-Diacetyl -L-tartaric anhydride (48) is an article of commerce and reacts readily with a variety of amines to generate monoacid/monoamide intermediate.
  • the preferred solvent for the ring opening is DCM; although DMF, THF or dioxane may also be compatible.
  • the subsequent peptide coupling step proceeds under a variety of standard conditions which are given for Example 1.
  • the acetate groups can be removed using a variety of conditions, including hydrazine/methanol, NaOMe in methanol, ammonia/methanol, lithium hydroxide/THF/water (saponification), potassium carbonate in methanol, or MP-carbonate.
  • the preferred set of conditions for parallel synthesis is to use the MP-carbonate. It has also been observed that one or both of the acetate protecting groups can be cleaved during synthetic transformations involving nucleophiles, basic reagents, heating in protic solvents or exposure to organometallic reagents.
  • Compound 53 was prepared according to the procedure described by D. Miller et al (J. Med. Chem. 1999, 42, 2287).
  • Compound 55 was prepared using procedures similar to those described in
  • Polystyrene EDC resin (30mg, 0.045 mmol) was added to 96-wells of a deep well polypropylene microtiter plate followed by a MeCN/THF/DMF (6:6:1 ) stock solution (1 mL) of B4 (0.015 mmol) and HOBT (0.0225 mmol). Then 1 M stock solutions of each of the individual acids (R -96COOH) (0.023 mL, 0.021 mmol) were added to the wells, which was then sealed and shaken at 25 °C for 20h.
  • Polystyrene DIEA resin (30mg, 0.045 mmol) was added to 72-wells of a deep well polypropylene microtiter plate followed by a MeCN/THF/DMF (6:6:1 ) stock solution (1 mL) of 155 (0.015 mmol). Then 1M stock solutions of each of the individual sulfonyl chlorides (R 1 -72S0 2 CI) (0.023 mL, 0.021 mmol) were added to the wells, which was then sealed and shaken at 25 °C for 20h.
  • a compound precursor may be prepared such that the synthesis of direct analogs is facilitated. Examples of schemes for biaryl synthesis are listed below.
  • Aryl halides and 'pseudo-halides' i.e. triflates
  • boronic acids under a variety of established conditions (Top. Curr. Chem. 2002, 219, 12-49).
  • a variety of bases for this reaction are known in the literature, including sodium carbonate, potassium carbonate, cesium carbonate, potassium t-butoxide, sodium t-butoxide, TEA, DIEA, potassium fluoride, and potassium phosphate. For most applications potassium phosphate was preferred and gave acceptable yields and chemoselectivity.
  • solvents have also been used in the literature for Suzuki reactions, including THF, dioxane, NMP, DMF, DME, DMA, toluene, and water. In general we have found that THF or dioxane are preferred solvents. Solvents may also be mixed in various proportions to enhance reactivity and/or chemoselectivity. Palladium sources for this reaction are numerous as well, including Pd(TPP) 4> Pd(OAc) 2 , PdCI 2 (TPP) 2 , PdCI 2 (dppf), Pd 2 (dba) 3 . In general PdCl2(dppf) was found to be a preferred palladium source.
  • Example 1 part A To 188 (prepared as described in Example 4 Part C from (+)-diacetyl -L-tartaric anhydride (48) and 2- chlorophenylpiperazine) (916 mg, 2.22 mmol) in DMF (5 mL) was added 187 (502 mg, 2.61 mmol), DIEA (850 ⁇ L, 4.88 mmol) and HATU (928 mg, 2.44 mmol). Purification by column chromatography (Si0 2> 20% to 50% EtOAc/DCM) afforded 189 as an off-white foam (652 mg, 50%).
  • Stepl To 2-chlorophenyl boronic acid (24 mg, 0.152 mmol), potassium phosphate (65 mg, 0.306 mmol) and PdCI 2 (dppf) (3.1 mg, 5 mol%) under argon in a 4-mL vial was added 211 (40 mg, 0.076 mmol) in dioxane (0.5 mL). The reaction mixture was heated at 100 °C for 16 hours. The reaction mixture was cooled to room temperature, diluted with ethyl acetate and filtered through celite. The filtrate was concentrated.
  • Pd-mediated Negishi-type couplings were an efficient way to generate a set of relevant compounds for this series where Ar was a phenyl ring. While a range of palladium sources are known we found that Pd(P f Bu 3 ) 2 was preferred.
  • the reaction can be run in a variety of solvent systems, including dioxane, THF, or DMA. A preferred method utilized the solvent from the zinc reagent, which was typically available as a THF stock solution. Removal of the t-BOC group can be completed using a range of acidic conditions including TFA/water, HCI in methanol, and HCI in dioxane; ail of which proceeded equally well with most reaction substrates.
  • the reactions are typically concentrated and freeze-dried to give HCI or TFA salts.
  • the Negishi reactions proceed comparably, and in many cases better, when the amine was protected by a phthalimide.
  • the phthalimide protecting group could be removed by heating in ethanol containing hydrazine hydrate.
  • Compound 237 was prepared according to the procedure described by A. J. Hutchison et al (European Patent 0323807 A2, Dec. 7, 1989)
  • N-(Hydroxymethyl)phthalimide (1.0 g, 5.6 mmol) was dissolved in 1% triflic acid in trifluoroacetic acid (10 mL) at 0 °C. To this mixture was added 330 (0.52 mL, 5.6 mmol). The reaction mixture was warmed to room temperature slowly and stirred overnight. The reaction mixture was poured into ice water (100 mL) and extracted with DCM. The combined organic layers were washed with sodium bicarbonate solution and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si0 2 , 30% EtOAc/Hex) afforded 331 (619 mg, 41 %).
  • a solution of LDA was formed by the addition of 1.6 M ⁇ -butyl lithium (34 mL, 54.5 mmol) to diisopropylamine (8.47 mL, 60 mmoL) in THF (20 mL) at -78 °C. The reaction mixture was stirred at -78 °C for 20 minutes and warmed to 0 °C gradually. The LDA solution was added dropwise to a mixture of N-Boc- D-proline methyl ester (469) (2.5 g, 10.9 mmol) and chloroiodomethane (3.17 mL, 43.6 mmol) in THF (20 mL) at - 78 °C via a cannula over 30 minutes.
  • Oxalyl chloride (1.87 g, 1.3 mL, 14.7 mmol) was dissolved in dry CH 2 CI 2 (35 mL) and cooled to -78° C under a nitrogen atmosphere.
  • DMSO (2.30 g, 2.1 mL, 29.5 mmol) dissolved in dry CH 2 CI 2 (5 mL) was added dropwise via addition funnel. The solution was stirred at -78° C for 15 mins then compound 514 (3.27 g, 11.8 mmol) dissolved in CH 2 CI 2 (15 mL) was added. The reaction mixture was stirred at -78° C for 60 mins then triethylamine (3.58 g, 4.9 mL, 35.4 mmol) was added.
  • Example 12 Suzuki Aryl-Aryl couplings
  • Example 12A Suzuki Aryl-Aryl couplings
  • Part A A solution of ethyl 2-bromobenzoate (548) (0.25 g, 1.1 mmol), (4- aminomethylphenyl)boronic acid (547) (0.20 g, 1.1 mmol), and Pd(dppf)CI 2 (0.040 g, 0.055 mmol) in CH 3 CN (1 mL) and 1 N K 2 C0 3 (1 mL) was heated in a SmithCreator microwave (2-5 mL vessel, 100 °C for 5 minutes). The reaction was diluted with H 2 0 and EtOAc. The organic layer was removed, and the aqueous phase was extracted with EtOAc (3x). The combined organics were dried (Na 2 S0 4 ), filtered, and concentrated.
  • Part D To a solution of 551 (55 mg, 0.104 mmol) in CH 2 CI2 (1 mL) was added polystyrene-bound HOBt (109 mg, 0.095 mmol), DIC (0.067 mL, 0.427 mmol), and DMAP (7 mg, 0.057 mmol) and was shaken overnight. The mixture was filtered, then washed with DMF (3 x 3 mL), CH 2 CI 2 (3 x 3 mL), DMF (3 x 3 mL), and THF (3 x 3 mL). The resin was dried under vacuum overnight.
  • Compound 563 was prepared using procedures similar to those described in Example 11 A.
  • Compound 564 was prepared from 562 and 563 using procedures similar to those described in Example 1 Part A.
  • HPLC-MS t R 2.49 min (UV 254 nm); mass calculated for formula C 31 H 30 CIF 3 N 2 O 5 602.2, observed LCMS m/z 603.2 (M+H).
  • Compound 631 was prepared by the procedures described in Example 11 A. Part A.
  • a Schlenck flask was flame dried under N 2 flow, capped with a septum, and allowed to cool to rt.
  • LiAIH 4 (1.26 g, 33.2 mmol) and AICI 3 (1.47 g, 11.0 mmol) were added to the flask, followed by anhydrous THF.
  • the flask was placed in an ice-water bath immediately and allowed to cool with stirring.
  • Compound 641 was added to the flask in portions.
  • the reaction mixture was stirred for 3 h during which time it warmed to 15 °C.
  • the reaction was recooled to 0 °C and water (3 mL) was added.
  • Aqueous 3.0 N sodium hydroxide was added (6 mL) followed by water (9 mL).
  • the reaction mixture was filtered through a pad of Celite which was rinsed with EtOAc. The resulting filtrate was concentrated to dryness giving a green solid.
  • the crude product was partially purified via flash sgc using a 40%-50% EtOAc/hexanes gradient, followed by 98%EtOAc/2% Et 3 N. The fractions containing the impure desired product were combined to give a blue oil. This material was dissolved in CH 2 CI 2 (6 mL). TFA (4 mL) and triethyl silane (2 mL) were added and the reaction mixture was left stirring ON. The reaction mixture was concentrated to give a green oil.
  • Compound 675 was prepared as described in Example 4A Part C.
  • Step A1

Abstract

This invention relates to compounds of the Formula (I) or a pharmaceutically acceptable salt, solvate or isomer thereof, which can be useful for the treatment of diseases or conditions mediated by MMPs, ADAMs, TACE, TNF-α or combinations thereof.

Description

COMPOUNDS FOR THE TREATMENT OF INFLAM ATORY DISORDERS
BACKGROUND OF THE INVENTION Field of the Invention
This invention relates generally to tartaric acid functional compounds that can inhibit matrix metalloproteinases (MMPs), a disintegrin and metalloproteases (ADAMs) and/or tumor necrosis factor alpha - converting enzyme (TACE) and in so doing prevent the release of tumor necrosis factor alpha (TNF-α), pharmaceutical compositions comprising such compounds, and methods of treatment using such compounds.
Description
Osteo- and rheumatoid arthritis (OA and RA, respectively) are destructive diseases of articular cartilage characterized by localized erosion of the cartilage surface. Findings have shown that articular cartilage from the femoral heads of patients with OA, for example, had a reduced incorporation of radiolabeled sulfate over controls, suggesting that there must be an enhanced rate of cartilage degradation in OA (Mankin et al. J. Bone Joint Surg. 52A (1970) 424-434). There are four classes of protein degradative enzymes in mammalian cells: serine, cysteine, aspartic and metalloproteases. The available evidence supports the belief that it is the metalloproteases that are responsible for the degradation of the extracellular matrix of articullar cartilage in OA and RA. Increased activities of collagenases and stromelysin have been found in OA cartilage and the activity correlates with severity of the lesion (Mankin et al. Arthritis Rheum. 21 , 1978, 761-766, Woessner et al. Arthritis Rheum. 26, 1983, 63-68 and Ibid. 27, 1984, 305-312). In addition, aggrecanase (a newly identified metalloprotease) has been identified that provides the specific cleavage product of proteoglycan, found in RA and OA patients (Lohmander L S. et al. Arthritis Rheum, 36, 1993, 1214-22).
Metalloproteases (MPs) have been implicated as the key enzymes in the destruction of mammalian cartilage and bone. It can be expected that the pathogenesis of such diseases can be modified in a beneficial manner by the administration of MP inhibitors (see Wahl et al. Ann. Rep. Med. Chem. 25, 175-184, AP, San Diego, 1990). MMPs are a family of over 20 different enzymes that are involved in a variety of biological processes important in the uncontrolled breakdown of connective tissue, including proteoglycan and collagen, leading to resorption of the extracellular matrix. This is a feature of many pathological conditions, such as RA and OA, comeal, epidermal or gastric ulceration; tumor metastasis or invasion; periodontal disease and bone disease. Normally these catabolic enzymes are tightly regulated at the level of their synthesis as well as at their level of extracellular activity through the action of specific inhibitors, such as alpha-2-macroglobulins and TIMPs (tissue inhibitor of MPs), which form inactive complexes with the MMP's.
Tumor necrosis factor alpha (TNF-α) is a cell-associated cytokine that is processed from a 26 kDa precursor form to a 17 kd active form. See Black R.A. "Tumor necrosis factor-alpha converting enzyme" lnt J Biochem Cell Biol. 2002 Jan;34(1):1-5 and Moss ML, White JM, Lambert MH, Andrews RC.'TACE and other ADAM proteases as targets for drug discovery" Drug Discov Today. 2001 Apr 1 ;6(8):417-426, each of which is incorporated by reference herein.
TNF-α has been shown to play a pivotal role in immune and inflammatory responses. Inappropriate or over-expression of TNF-α is a hallmark of a number of diseases, including RA, Crohn's disease, multiple sclerosis, psoriasis and sepsis. Inhibition of TNF-α production has been shown to be beneficial in many preclinical models of inflammatory disease, making inhibition of TNF-α production or signaling an appealing target for the development of novel anti-inflammatory drugs.
TNF-α is a primary mediator in humans and animals of inflammation, fever and acute phase responses, similar to those observed during acute infection and shock. Excess TNF-α has been shown to be lethal. Blocking the effects of TNF-α with specific antibodies can be beneficial in a variety of conditions, including autoimmune diseases such as RA (Feldman et al, Lancet, (1994) 344, 1105), non-insulin dependent diabetes mellitus (Lohmander L. S. et al., Arthritis Rheum. 36 (1993) 1214-22) and Crohn's disease (Macdonald T. et al., Clin. Exp. Immunol. 81 (1990) 301 ). Compounds that inhibit the production of TNF-α are therefore of therapeutic importance for the treatment of inflammatory disorders. Recently it has been shown that metalloproteases, such as TACE, are capable of converting TNF-α from its inactive to active form (Gearing et al Nature, 1994, 370, 555). Since excessive TNF-α production has been noted in several disease conditions also characterized by MMP-mediated tissue degradation, compounds which inhibit both MMPs and TNF-α production may also have a particular advantage in diseases where both mechanisms are involved.
One approach to inhibiting the harmful effects of TNF-α is to inhibit the enzyme, TACE before it can process TNF-α to its soluble form. TACE is a member of the ADAM family of type I membrane proteins and mediates the ectodomain shedding of various membrane-anchored signaling and adhesion proteins. TACE has become increasingly important in the study of several diseases, including inflammatory disease, because of its role in cleaving TNF- α from its "stalk" sequence and thus releasing the soluble form of the TNF-α protein (Black R.A. Int J Biochem Cell Biol. 2002 34,1-5).
There are several patents which disclose hydroxamate, carboxylate and/or lactam based MMP inhibitors.
US 6,677,355 and US 6,534,491 (B2), describe compounds that are hydroxamic acid derivatives and MMP inhibitors.
US 6,495,565 discloses lactam derivatives that are potential inhibitors of matrix metalloproteases and/or TNF-α.
There is a need in the art for inhibitors of MMPs, ADAMs, TACE, and TNF-α, which can be useful as anti-inflammatory compounds and cartilage protecting therapeutics. The inhibition of TNF-α, TACE and or other MMPs can prevent the degradation of cartilage by these enzymes, thereby alleviating the pathological conditions of OA and RA as well as many other auto-immune diseases.
SUMMARY OF THE INVENTION
In its many embodiments, the present invention provides a novel class of compounds as inhibitors of TACE, the production of TNF-α, MMPs, ADAMs or any combination thereof, methods of preparing such compounds, pharmaceutical compositions comprising one or more such compounds, methods of preparing pharmaceutical formulations comprising one or more such compounds, and methods of treatment, prevention, inhibition or amelioration of one or more diseases associated with TACE, TNF-σ, MMPs, ADAMs or any combination thereof using such compounds or pharmaceutical compositions.
In one embodiment, the present application discloses a compound, or pharmaceutically acceptable salts or solvates of said compound, said compound having the general structure shown in formula (I):
Figure imgf000006_0001
formula (I)
or a pharmaceutically acceptable salt or solvate thereof, wherein: A is selected from the group consisting of:
Figure imgf000006_0002
-C02R1; d is 0 to 4;
J is selected from the group consisting of: O, S, and NR5;
E is selected from the group consisting of: O, S, and NR5;
T is O or S;
R1 and R2 are the same or different, each being independently selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, and heteroaryl; or alternatively R1 and R2, taken together with the N to which R1 and R2 are shown attached, represent a 4-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein each of said alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, heteroaryl and 4-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties below;
R10 is selected from the group consisting of H, alkyl, and fluoroalkyl;
R20 is selected from the group consisting of H, alkyl, and fluoroalkyl;
R30 is H or alkyl, or alternatively R30 and R40 taken together with the N to which R40 is shown attached to in Formula I, are joined to form a 4-7 membered heterocylic ring, wherein said heterocylic ring is unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties below;
R40 is H or alkyl;
R50 is H or alkyl;
W is -(CR13 2)n-, wherein n is 0 to 5 or a covalent bond, or alternatively two R 3 groups can fuse to form a 3-8 membered cycloalkyl, wherein said 3-8 membered cycloalkyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R6 moieties below;
X is absent or present, and if present X is selected from the group consisting of a covalent bond, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties below;
Y is absent or present, and if present Y is selected from the group consisting of a covalent bond, -[C(R6)2]n- wherein n is 1 to 2, -0-, -S-, -NR1-, -SOv- wherein v is 1 to 2, -SOn(CR62)p- wherein n is 1 or 2 and p is 1 to 4, -0(CR6 2)q- or -(CR6 2)qO- wherein q is 1 to 4, -N(R7)S(0)n- or -S(0)nN(R7)- wherein n is 1 or 2, and -N(R7)C(0)- or -C(0)N(R7)-;
Z is selected from the group consisting of H, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, said cycloalkyl, heterocyclyl, aryl, and heteroaryl being optionally fused with aryl, heterocyclyl, heteroaryl or cycloalky; wherein each of said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties below;
R5 is selected from the group consisting of hydrogen, alkyl, and alkylaryl; each R6 is the same or different and is independently selected from the group consisting of hydrogen, halogen, -SR^, -S(0)qR^ 5 wherein q is 1 to 2, alkyl, cycloalkyl, heterocyclyl, alkoxyl, hydroxy, nitro, cyano, amino, alkenyl, alkynyl, arylalkyl, aminocarbonyl, alkylcarbonyl, and alkoxycarbonyl; each R7 is the same or different and is independently selected from the group consisting of hydrogen, alkyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, alkenyl, alkynyl, arylalkyl, alkylcarbonyl, and alkoxycarbonyl, wherein each of the aryl, heteroaryl and heterocyclyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties below;
R13 is the same or different and is independently selected from the group consisting of hydrogen, halogen, -OH, -OR14, alkyl, cycloalkyl, heterocyclyl, alkenyl, alkynyl, alkylaryl, alkylamino, and alkylcarbonyl;
14
R is alkyl; each R70 is a substituent for H where indicated and is the same or different and is independently selected from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, -CN, -CF3, -OCF3, -OR15, -C(0)R15, -C(0)OR 5 -C(0)N(R15)(R16); _SR15, -S(0)qN(R15)(R 6) wherein q is 1 to 2, -C(=NOR15)R16J _N(R15)(R16), -alkyl-N(Rl 5)(Rl6)t -N(R15)C(0)R16, - CH2-N(R15)C(0)R16, -N(R 15)S(0)R16, -N(R 5)S(0)2R16, -CH2- N(R15)S(0)2R 6, -N(R17)S(0)2N(R16)(R15)t -N(R 7)S(0)N(R16)(R 5),
-N(R17)C(0)N(R16)(R 5), -CH2-N(R17)C(0)N(R16)(R15). -N(R15)C(0)0R16, -CH2-N(R1 5)C(0)OR 16, and -S(0)qR15 wherein q is 1 to
2; and wherein each of the alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkenyl and alkynyl are independently unsubstituted or substituted by 1 to 5 groups independently selected from the group consisting of alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, halo, -CF3,
-CN, -OR15, -N(R15)(R16)J _c(0)OR15, -C(0)N(R15)(R16)J and
-N(R15)S(0)R16;
15 16 17 each R , R and R are independently selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, or alternatively R15 and R16 taken together with the N to which they are shown attached, are joined to form a 4-8 membered heterocylic ring, wherein said 4-8 membered cycloalkyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R75 moieties below; each R75 is independently selected from the group consisting of alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkenyl and alkynyl, and wherein each of the alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkenyl and alkynyl are independently unsubstituted or substituted by 1 to 5 groups independently selected from the group consisting _of alkyi, cycloalkyLheterocyclyl, aryl, heteroaryl, halo, -CF3,
-CN, -0R
Figure imgf000009_0001
; and each R19 is independently selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl.
The compounds of Formula I can be useful as inhibitors and may be useful in the treatment and prevention of diseases associated with TACE, TNF-α, MMPs, ADAMs or any combination thereof. DETAILED DESCRIPTION OF THE INVENTION
In its several embodiments, the present invention provides a novel class of inhibitors of TACE, the production of TNF-α, MMPs, ADAMs or any combination thereof, pharmaceutical compositions containing one or more of the compounds, methods of preparing pharmaceutical formulations comprising one or more such compounds, and methods of treatment, prevention or amelioration of one or more of the symptoms of inflammation.
In one embodiment, the present invention provides compounds which are represented by structural Formula (I) above or a pharmaceutically acceptable salt, solvate or isomer thereof, wherein the various moieties are as described above.
In one embodiment, R1 and R2 are the same or different, each being independently selected from the group consisting of alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, and heteroaryl; or alternatively R1 and R2, taken together with the N to which R1 and R2 are shown attached, represent a 4-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein each of said alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, heteroaryl and 4-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70.
In one embodiment, R1 and R2 are the same or different, each being independently selected from the group consisting of H, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, and heteroaryl; or alternatively R1 and R2, taken together with"the N to which R1 and R2 are shown attached, represent a 4-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein each of said alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, heteroaryl and 4-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70. in one embodiment, A is selected from the group consisting of:
Figure imgf000011_0001
R1 and R2 are shown attached, represent a 4-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 4-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties.
In another embodiment, A is
Figure imgf000011_0002
wherein R1 and R2 are the same or different, each being independently selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, and heteroaryl; wherein said alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl and heteroaryl can be unsubstituted or optionally independently substituted with one or more moieties which can e-the.same or.different.-each-moiety-being independently selected from the group of R70 moieties.
In another embodiment, A is
o s
II II
R \ R2 2 or R \ R2 2 wherein R1 and R2 are the same or different, each being independently selected from the group consisting of alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, and heteroaryl; wherein said alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl and heteroaryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties.
In another embodiment, A is
Figure imgf000012_0001
wherein R1 and R2 are the same or different, each being independently selected from the group consisting of H, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, and heteroaryl; wherein said alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl and heteroaryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties.
In another embodiment, A is
Figure imgf000012_0002
In another embodiment, A is
Figure imgf000012_0003
wherein a is 0 to 4 and x is 0 to 4. In another embodiment, A is
Figure imgf000013_0001
wherein each x idependently is 0 to 4.
In another embodiment, A is selected from the group consisting of:
Figure imgf000013_0002
R2, taken together with the N to which R1 and R2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; wherein a ring is formed from -NR1R2 and said ring is
Figure imgf000013_0003
wherein each R9 is a substituent for H where indicated and can be the same or different, each being independently selected from the group consisting of -OH,
-OR14, -C(0)OR15, -C(0)N(R15)(R16), alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl"and heteT^yOlyl,"wh^rein"s d^ll^l aryr, "heteroarylTalRenyl, alkynyl, cycloalkyl and heterocyclyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties;
R11and R12 taken together with the carbon to which each R11and R12 are shown attached are fused heteroaryl or fused cycloalkyl, wherein said fused heteroaryl and fused cycloalkyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; and x is 0 to 4, and when x is greater than 1 , each R9 moiety can be the same or different, each moiety being independently selected from the group of R9 moieties.
In another embodiment, A is selected from the group consisting of:
Figure imgf000014_0001
wherein R1 and R2, taken together with the N to which R and R2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; wherein a ring is formed from -NR1R2 and said ring is
Figure imgf000014_0002
wherein each R9 is a substituent for H where indicated and can be the same or different, each being independently selected from the group consisting of -OH, -OR14, -C(0)0R15 -C(0)N(R15)(R16)) ajky|f ary t heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl, wherein said alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; x is 0 to 4, and when x is greater than 1 , each R9 moiety can be the same or different, each moiety being independently selected from the group of R9 moieties; and
G is selected from the group consisting of CH2, NR7, O, S, or SO2.
In another embodiment, A is selected from the group consisting of:
Figure imgf000015_0001
wherein R and R , taken together with the N to which R1 and R2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; a ring is formed from -NR1R2 and said ring is selected from the group consisting of
Figure imgf000016_0001
Figure imgf000016_0002
wherein x is 0 to 4, and when x is greater than 1 , each R70 moiety can be the same or different, each moiety being independently selected from the group of R70 moieties; and each R18 is the same or different and is independently H or alkyl. In another embodiment, A is selected from the group consisting of:
Figure imgf000017_0001
wherein R and R , taken together with the N to which R1 and R2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; a ring is formed from -NR1R2 and said ring is selected from the group consisting of
Figure imgf000017_0002
wherein x is 0 to 4, and when x is greater than 1 , each R70 moiety can be the same or different, each moiety being independently selected from the group of R70 moieties.
In another embodiment, A is selected from the group consisting of:
Figure imgf000017_0003
wherein R1 and R2, taken together with the N to which R1 and R2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; a ring is formed from -NR1R2 and said ring is
Figure imgf000018_0001
wherein each R9 is a substituent for H where indicated and can be the same or different, each being independently selected from the group consisting of -OH,
-OR14, -C(0)OR15, -C(0)N(R15)(R16), alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl, wherein said alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; and x is 0 to 4, and when x is greater than 1 , each R9moiety can be the same or different, each moiety being independently selected from the group of
R9moieties.
In another embodiment, J is O.
In another embodiment, E is O.
In another embodiment, R10 is H or alkyl.
In another embodiment, R20, R30, R40, and R50 are all the same and are
H.
In another embodiment, R6 is H or alkyl.
In another embodiment, R6 is H.
In another embodiment, R13 is H or alkyl.
In another embodiment, R 3 is H or -CH3.
In another embodiment, A is selected from the group consisting of
Figure imgf000019_0001
wherein R1 and R2, taken together with the N to which R1 and R2 are shown attached, represent a 4-6 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R70, wherein R70 is aryl.
In another embodiment, Y is a covalent bond or -[C(R6)2]n- wherein n is 1 to 2, -CH2-.
In another embodiment, Y is selected from the group consisting of a covalent bond, -CH2-, -C(H)(OH)-, -C(O)- and -0-.
In another embodiment, Y is a covalent bond or -CH2-.
In another embodiment, Y is a covalent bond.
In another embodiment, W is -(CR13 )n-, in which n is 0-5.
In another embodiment, W is -(CR13 2)n-, in which n is 1-5 and each R13 is H or alkyl. In another embodiment, W is selected from the group consisting of - CH2-, -C(H)(CH3)-, -C(CH3)2- and -CH2CH2-.
In another embodiment, W is -CH2-.
In another embodiment, W is -C(H)(CH3)-.
In another embodiment, X is selected from the group consisting of alkyl, aryl, heterocyclyl and heteroaryl.
In another embodiment, X is aryl.
In another embodiment, X is heteroaryl.
In another embodiment, X is selected from the group consisting of phenyl, azetidinyl, pyrrolidinyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl.
In another embodiment, X is selected from the group consisting of phenyl, pyridinyl and piperidinyl.
In another embodiment, X is selected from the group consisting of
Figure imgf000020_0001
wherein x is 0 to 4, and wherein x is greater than 1 , each R70 moiety can be the same or different, each moiety being independently selected from the group of R70 moieties.
In the above shown moieties for X, X is the moiety enclosed by < and W and Y are shown only to indicate which end of the X is attached to W and which end to Y. Similar depictions occur throughout this application with similar connotations. In another embodiment X is selected from the group consisting of
Figure imgf000021_0001
wherein x is 0 to 4, and when x is greater than 1 , each R70 moiety can be the same or different, each moiety being independently selected from the group of R70 moieties.
In another embodiment, Z is selected from the group consisting of H, aryl and heteroaryl.
In another embodiment, Z is selected from the group consisting of H, phenyl, indolyl, benzimidazolyl, pyrazolyl, thienyl, pyridinyl, thiazolyl, thiadiazolyl, imidazolyl, pyrrolidinyl, pyrazinyl, triazolyl, tetrazolyl and tetrazinyl, wherein said phenyl, indolyl, benzimidazolyl, pyrazolyl, thienyl, pyridinyl, thiazolyl, thiadiazolyl, imidazolyl, pyrrolidinyl, pyrazinyl, triazolyl, tetrazolyl and tetrazinyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group consisting of R 70 moieties.
In another embodiment, Z is selected from the group consisting of
Figure imgf000022_0001
wherein x is 0 to 4, and when x is greater than 1 , each R70 moiety can be the same or different, each moiety being independently selected from the group of R70 moieties.
In another embodiment, Z is phenyl.
In another embodiment, Z is a phenyl substituted with at least one substituent selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy, amino and tetrazole.
In another embodiment, Z is thienyl. In another embodiment, Z is a thienyl substituted with at least one substituent selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy, amino and tetrazole.
In another embodiment, Z is pyrazolyl.
In another embodiment, Z is a pyrazolyl substituted with at least one substituent selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy, amino and tetrazole.
In another embodiment, Z is pyridinyl.
In another embodiment, Z is a pyridinyl substituted with at least one substituent selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy, amino and tetrazole.
In another embodiment, Z is imidazolyl.
In another embodiment, Z is an imidazolyl substituted with at least one substituent selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy, amino and tetrazole.
Figure imgf000023_0001
wherein R1 and R2, taken together with the N to which R1 and R2 are shown attached, represent a 4-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 4-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; E, J and T are the same and are O; R10 is H or alkyl; R20, R30, R40, and R50 are the same and are H; W is -(CR13 2)n-,wherein n is 0 to 5; X is selected from the group consisting of aryl, heteroaryl and heterocyclyl; Y is selected from the group consisting of a covalent bond, - [C(R6)2]n- wherein n is 1 to 2, -0-, -S-, and -NR1-; and Z is selected from the group consisting of
Figure imgf000024_0001
In another embodiment, A is:
Figure imgf000024_0002
wherein;" the-NR1R2of
Figure imgf000024_0003
IS
Figure imgf000025_0001
wherein each R9 is a substituent for H where indicated and can be the same or different, each being independently selected from the group consisting of -OH,
-OR14, -C(0)OR15, -C(0)N(R 5)(R16), alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl, wherein said alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; and x is 0 to 4, and when x is greater than 1 , each R9moiety can be the same or different, each moiety being independently selected from the group of R9 moieties;
E, J and T are the same and are O; R10 is H or alkyl; R20, R30, R40, and R50 are the same and are H; W is -(CR13 2)n-,wherein n is 0 to 5; X is selected from the group consisting of aryl, heteroaryl and heterocyclyl; Y is selected from the group consisting of a covalent bond, -[C(R6) ]n- wherein n is 1 to 2, - 0-, -S-, and -NR1-; and Z is selected from the group consisting of
Figure imgf000026_0001
In another embodiment, A is: O
II
R 1 \
R^ wherein R and R2, taken together with the N to which R1 and R2 are shown attached, represent a 4-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 4-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; E, J and T are the same and are O; R10 is H or alkyl; R20, R30, R40, and R50 are the same and are H; W is -(CR13 2)n-,wherein n is 0 to 5; X is selected from the group consisting of aryl, heteroaryl and heterocyclyl; Y is selected from the group consisting of a covalent bond, - [C(R6)2]n- wherein n is 1 to 2, -0-, -S-, and -NR1-; and Z is selected from the group consisting of
Figure imgf000027_0001
Figure imgf000027_0002
attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; wherein E, J and T are the same and are O; R10 is H or alkyl; R20, R30, R40, and R50 are the same and are H; W is -(CR6 2)n-, wherein n is 0 to 5; X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl and pyrazolyl; Y is -[C(R6)2]n- wherein n is 1 to 2; and Z is selected from the group consisting of
Figure imgf000028_0001
In another embodiment, A is: O
N ' \ y
\
R1 R R2 wherein R1 and R2, taken together with the N to which R1 and R2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; wherein E, J and T are the same and are O; R10 is H or alkyl; R20, R30, R40, and R50 are the same and are H; W is -(CR6 )n-, wherein n is 0 to 5; X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl and pyrazolyl; Y is -[C(R6)2]n- wherein n is 1 to 2; and Z is selected from the group consisting of
Figure imgf000030_0001
Figure imgf000030_0002
Figure imgf000030_0003
and
Figure imgf000030_0004
Figure imgf000030_0005
Figure imgf000030_0006
wherein R and R , taken together with the N to which R and R2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties;
E, J and T are the same and are O; R10 is H or alkyl; R20, R30, R40, and R50 are the same and are H; W is -(CR6 2)n-, wherein n is 0 to 5; X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl and pyrazolyl; Y is selected from the group consisting of 0-, -S-, and -NR1-; and Z is selected from the group consisting of
Figure imgf000031_0001
In another embodiment, A is
Figure imgf000031_0002
wherein R1 and R2, taken together with the N to which R1 and R2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; wherein E, J and T are the same and are O; R10 is H or alkyl; R20, R30, R40, and R50 are the same and are H; W is -(CR62)n-, wherein n is 0 to 5; X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl and pyrazolyl; Y is selected from the group consisting of 0-, -S-, and -NR1-; and Z is selected from the group consisting of
Figure imgf000032_0002
Figure imgf000032_0003
Figure imgf000032_0001
Figure imgf000032_0005
Figure imgf000032_0004
Figure imgf000032_0006
In an ing of:
Figure imgf000033_0001
wherein R1 and R2, taken together with the N to which R1 and R2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; wherein E, J and T are the same and are O; R10 is H or alkyl; R20, R30, R40, and R50 are the same and are H; W is -(CR13 2)n- wherein n is 1 or 2; X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl and pyrazolyl; Y is a covalent bond and Z is selected from the group consisting of
Figure imgf000034_0001
In another embodiment, A is o
II
R^ \
R R2 • wherein R1 and R2, taken together with the N to which R1 and R2 are shown attached, represent a 3-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 3-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; wherein E, J and T are the same and
)10 520 D30 D40 50 are O; R1U is H or alkyl; R ", Rou, R4U, and Rou are the same and are H; W is (CR )132)rr wherein n is 1 or 2; X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl and pyrazolyl; Y is a covalent bond and Z is selected from the group consisting of
Figure imgf000035_0001
In another embodiment, X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl, and pyrazolyl.
In another embodiment, X is selected from the group consisting of phenyl, piperidinyl, thienyl, and pyridinyl and Y is selected from the group consisting of a covalent bond, -[C(R6)2]n- wherein n is 1 to 2 and -0-.
In another embodiment, X is phenyl and Y is selected from the group consisting of a covalent bond, -CH2- and -0-.
In another embodiment, X is piperidinyl, Y is a covalent bond, and Z is aryl or heteroaryl having two substituents which can be the same or different, each moiety being independently selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy and amino.
In another embodiment, W is selected from the group consisting of - CH2-, -C(H)CH3-, -C(CH3)2- and Y is a covalent bond.
In another embodiment, W is -CH2- and Y is -CH2-.
In another embodiment, Z is selected from the group consisting of
Figure imgf000036_0001
Another embodiment of the invention discloses the compounds shown in Table 1 below.
As used above, and throughout this disclosure, the following terms, unless otherwise indicated, shall be understood to have the following meanings:
"Patient" includes both human and animals.
"Mammal" means humans and other mammalian animals.
"Alkyl" means an aliphatic hydrocarbon group which may be straight or branched and comprising about 1 to about 20 carbon atoms in the chain. Preferred alkyl groups contain about 1 to about 12 carbon atoms in the chain. More preferred alkyl groups contain about 1 to about 6 carbon atoms in the chain. Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl, are attached to a linear alkyl chain. "Lower alkyl" means a group having about 1 to about 6 carbon atoms in the chain which may be straight or branched. The term "substituted alkyl" means that the alkyl group may be substituted by one or more substituents which may be the same or different, each substituent being independently selected from the group consisting of halo, alkyl, aryl, cycloalkyl, cyano, hydroxy, alkoxy, alkylthio, amino, -NH(alkyl), -NH(cycloalkyl), -N(alkyl)2) carboxy and -C(0)0-alkyl. Non- limiting examples of suitable alkyl groups include methyl, ethyl, n-propyl, isopropyl and t-butyl. The term "Fluoroalkyl" means an alkyl group in which alkyl is as previously described wherein one or more hydrogens are replaced with fluorine atoms.
"Alkenyl" means an aliphatic hydrocarbon group containing at least one carbon-carbon double bond and which may be straight or branched and comprising about 2 to about 15 carbon atoms in the chain. Preferred alkenyl groups have about 2 to about 12 carbon atoms in the chain; and more preferably about 2 to about 6 carbon atoms in the chain. Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl, are attached to a linear alkenyl chain. "Lower alkenyl" means about 2 to about 6 carbon atoms in the chain which may be straight or branched. Non-limiting examples of suitable alkenyl groups include ethenyl, propenyl, n-butenyl, 3-methylbut-2- enyl, n-pentenyl, octenyl and decenyl.
"Alkynyl" means an aliphatic hydrocarbon group containing at least one carbon-carbon triple bond and which may be straight or branched and comprising about 2 to about 15 carbon atoms in the chain. Preferred alkynyl groups have about 2 to about 12 carbon atoms in the chain; and more preferably about 2 to about 4 carbon atoms in the chain. Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl, are attached to a linear alkynyl chain. "Lower alkynyl" means about 2 to about 6 carbon atoms in the chain which may be straight or branched. Non-limiting examples of suitable alkynyl groups include ethynyl, propynyl, 2-butynyl and 3- methylbutynyl. The term "substituted alkynyl" means that the alkynyl group may be substituted by one or more substituents which may be the same or different, each substituent being independently selected from the group consisting of alkyl, aryl and cycloalkyl.
"Aryl" means an aromatic monocyclic or multicyclic ring system comprising about 6 to about 14 carbon atoms, preferably about 6 to about 10 carbon atoms. The aryl group can be optionally substituted with one or more "ring system substituents" which may be the same or different, and are as defined herein. Non-limiting examples of suitable aryl groups include phenyl and naphthyl.
"Heteroaryl" means an aromatic monocyclic or multicyclic ring system comprising about 5 to about 14 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the ring atoms is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination. Preferred heteroaryls contain about 5 to about 6 ring atoms. The "heteroaryl" can be optionally substituted by one or more "ring system substituents" which may be the same or different, and are as defined herein. The prefix aza, oxa or thia before the heteroaryl root name means that at least a nitrogen, oxygen or sulfur atom respectively, is present as a ring atom. A nitrogen atom of a heteroaryl can be optionally oxidized to the corresponding N-oxide. Non- limiting examples of suitable heteroaryls include pyridyl, pyrazinyl, furanyl, thienyl, pyrimidinyl, pyridone (including N-substituted pyridones), isoxazolyl, isothiazolyl, oxazolyl, thiazolyl, pyrazolyl, furazanyl, pyrrolyl, pyrazolyl, triazolyl, 1 ,2,4-thiadiazolyl, pyrazinyl, pyridazinyl, quinoxalinyl, phthalazinyl, oxindolyl, imidazo[1 ,2-a]pyridinyI, imidazo[2,1-b]thiazolyl, benzofurazanyl, indolyl, azaindolyl, benzimidazolyl, benzothienyl, quinolinyl, imidazolyl, thienopyridyl, quinazolinyl, thienopyrimidyl, pyrrolopyridyl, imidazopyridyl, isoquinolinyl, benzoazaindolyl, 1 ,2,4-triazinyl, benzothiazolyl and the like. The term "heteroaryl" also refers to partially saturated heteroaryl moieties such as, for example, tetrahydroisoquinolyl, tetrahydroquinolyl and the like.
"Aralkyl" or "arylalkyl" means an aryl-alkyl- group in which the aryl and alkyl are as previously described. Preferred aralkyls comprise a lower alkyl group. Non-limiting examples of suitable aralkyl groups include benzyl, 2- phenethyl and naphthalenylmethyl. The bond to the parent moiety is through the alkyl.
"Alkylaryl" means an alkyl-aryl- group in which the alkyl and aryl are as previously described. Preferred alkylaryls comprise a lower alkyl group. Non- limiting example of a suitable alkylaryl group is tolyl. The bond to the parent moiety is through the aryl.
"Cycloalkyl" means a non-aromatic mono- or multicyclic ring system comprising about 3 to about 10 carbon atoms, preferably about 5 to about 10 carbon atoms. Preferred cycloalkyl rings contain about 5 to about 7 ring atoms. The cycloalkyl can be optionally substituted with one or more "ring system substituents" which may be the same or different, and are as defined above. Non-limiting examples of suitable monocyclic cycloalkyls include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl and the like. Non-limiting examples of suitable multicyclic cycloalkyls include 1 -decalinyl, norbornyl, adamantyl and the like, as well as partially saturated species such as, for example, indanyl, tetrahydronaphthyl and the like.
"Halogen" means fluorine, chlorine, bromine, or iodine. Preferred are fluorine, chlorine and bromine.
"Ring system substituent" means a substituent attached to an aromatic or non-aromatic ring system which, for example, replaces an available hydrogen on the ring system. Ring system substituents may be the same or different, each being independently selected from the group consisting of alkyl, alkenyl, alkynyl, aryl, heteroaryl, aralkyl, alkylaryl, heteroaralkyl, heteroarylalkenyl, heteroarylalkynyl, alkylheteroaryl, hydroxy, hydroxyalkyl, alkoxy, aryloxy, aralkoxy, acyl, aroyl, halo, nitro, cyano, carboxy, alkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, alkylthio, arylthio, heteroarylthio, aralkylthio, heteroaralkylthio, cycloalkyl, heterocyclyl, -C(=N-CN)-NH2, -C(=NH)-NH2, -C(=NH)-NH(alkyl), Y-^N-, YiYzN-alkyl-, YιY2NC(0)-, Y1Y2NS02- and -SO2NY1Y2, wherein Y-i and Y2 can be the same or different and are independently selected from the group consisting of hydrogen, alkyl, aryl, cycloalkyl, and aralkyl. "Ring system substituent" may also mean a single moiety which simultaneously replaces two available hydrogens on two adjacent carbon atoms (one H on each carbon) on a ring system. Examples of such moiety are methylene dioxy, ethylenedioxy, -C(CH3)2- and the like which form moieties such as, for example:
Figure imgf000039_0001
"Heterocyclyl" means a non-aromatic saturated monocyclic or multicyclic ring system comprising about 3 to about 10 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the atoms in the ring system is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination. There are no adjacent oxygen and/or sulfur atoms present in the ring system. Preferred heterocyclyls contain about 5 to about 6 ring atoms. The prefix aza, oxa or thia before the heterocyclyl root name means that at least a nitrogen, oxygen or sulfur atom respectively is present as a ring atom. Any -NH in a heterocyclyl ring may exist protected such as, for example, as an -N(Boc), -N(CBz), -N(Tos) group and the like; such protections are also considered part of this invention. The heterocyclyl can be optionally substituted by one or more "ring system substituents" which may be the same or different, and are as defined herein. The nitrogen or sulfur atom of the heterocyclyl can be optionally oxidized to the corresponding N-oxide, S-oxide or S,S-dioxide. Non-limiting examples of suitable monocyclic heterocyclyl rings include piperidyl, pyrrolidinyl, piperazinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, 1 ,4-dioxanyl, tetrahydrofuranyl, tetrahydrothiophenyl, lactam, lactone, and the like.
It should be noted that in hetero-atom containing ring systems of this invention, there are no hydroxyl groups on carbon atoms adjacent to a N, O or S, as well as there are no N or S groups on carbon adjacent to another heteroatom. Thus, for example, in the ring:
Figure imgf000040_0001
there is no -OH attached directly to carbons marked 2 and 5.
It should also be noted that tautomeric forms such as, for example, the moieties:
Figure imgf000040_0002
are considered equivalent in certain embodiments of this invention.
"Alkynylalkyl" means an alkynyl-alkyl- group in which the alkynyl and alkyl are as previously described. Preferred alkynylalkyls contain a lower alkynyl and a lower alkyl group. The bond to the parent moiety is through the alkyl. Non-limiting examples of suitable alkynylalkyl groups include propargylmethyl. "Heteroaralkyl" means a heteroaryl-alkyl- group in which the heteroaryl and alkyl are as previously described. Preferred heteroaralkyls contain a lower alkyl group. Non-limiting examples of suitable aralkyl groups include pyridylmethyl, and quinolin-3-ylmethyl. The bond to the parent moiety is through the alkyl.
"Hydroxyalkyl" means a HO-alkyl- group in which alkyl is as previously defined. Preferred hydroxyalkyls contain lower alkyl. Non-limiting examples of suitable hydroxyalkyl groups include hydroxymethyl and 2-hydroxyethyl.
"Acyl" means an H-C(O)-, alkyl-C(O)- or cycloalkyl-C(O)-, group in which the various groups are as previously described. The bond to the parent moiety is through the carbonyl. Preferred acyls contain a lower alkyl. Non- limiting examples of suitable acyl groups include formyl, acetyl and propanoyl.
"Aroyl" means an aryl-C(O)- group in which the aryl group is as previously described. The bond to the parent moiety is through the carbonyl. Non-limiting examples of suitable groups include benzoyl and 1- naphthoyl.
"Alkoxy" means an alkyl-O- group in which the alkyl group is as previously described. Non-limiting examples of suitable alkoxy groups include methoxy, ethoxy, n-propoxy, isopropoxy and n-butoxy. The bond to the parent moiety is through the ether oxygen.
"Aryloxy" means an aryl-O- group in which the aryl group is as previously described. Non-limiting examples of suitable aryloxy groups include phenoxy and naphthoxy. The bond to the parent moiety is through the ether oxygen.
"Aralkyloxy" means an aralkyl-O- group in which the aralkyl group is as previously described. Non-limiting examples of suitable aralkyloxy groups include-benzyloxy-and-1— or-2=naphthalenemethoxy; The-bond-to the-parent moiety is through the ether oxygen.
"Alkylthio" means an alkyl-S- group in which the alkyl group is as previously described. Non-limiting examples of suitable alkylthio groups include methylthio and ethylthio. The bond to the parent moiety is through the sulfur.
"Arylthio" means an aryl-S- group in which the aryl group is as previously described. Non-limiting examples of suitable arylthio groups include phenylthio and naphthylthio. The bond to the parent moiety is through the sulfur.
"Aralkylthio" means an aralkyl-S- group in which the aralkyl group is as previously described. Non-limiting example of a suitable aralkylthio group is benzylthio. The bond to the parent moiety is through the sulfur.
"Alkoxycarbonyl" means an alkyl-O-CO- group. Non-limiting examples of suitable alkoxycarbonyl groups include methoxycarbonyl and ethoxycarbonyl. The bond to the parent moiety is through the carbonyl.
"Aryloxycarbonyl" means an aryl-O-C(O)- group. Non-limiting examples of suitable aryloxycarbonyl groups include phenoxycarbonyl and naphthoxycarbonyl. The bond to the parent moiety is through the carbonyl.
"Aralkoxycarbonyl" means an aralkyl-O-C(O)- group. Non-limiting example of a suitable aralkoxycarbonyl group is benzyloxycarbonyl. The bond to the parent moiety is through the carbonyl.
"Alkylsulfonyl" means an alkyl-S(02)- group. Preferred groups are those in which the alkyl group is lower alkyl. The bond to the parent moiety is through the sulfonyl.
"Arylsulfonyl" means an aryl-S(02)- group. The bond to the parent moiety is through the sulfonyl.
The term "substituted" means that one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. By "stable compound' or "stable structure" is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
The term "optionally substituted" means optional substitution with the specified groups, radicals or moieties.
The term "isolated" or "in isolated form" for a compound refers to the physical state of said compound after being isolated from a synthetic process or natural source or combination thereof. The term "purified" or "in purified form" for a compound refers to the physical state of said compound after being obtained from a purification process or processes described herein or well known to the skilled artisan, in sufficient purity to be characterizable by standard analytical techniques described herein or well known to the skilled artisan.
It should also be noted that any carbon as well as heteroatom with unsatisfied valences in the text, schemes, examples and Tables herein is assumed to have the sufficient number of hydrogen atom(s) to satisfy the valences.
When a functional group in a compound is termed "protected", this means that the group is in modified form to preclude undesired side reactions at the protected site when the compound is subjected to a reaction. Suitable protecting groups will be recognized by those with ordinary skill in the art as well as by reference to standard textbooks such as, for example, T. W. Greene et al, Protective Groups in organic Synthesis (1991 ), Wiley, New York.
When any variable (e.g., aryl, heterocycle, R2, etc.) occurs more than one time in any constituent or in Formula I, its definition on each occurrence is independent of its definition at every other occurrence.
As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
Prodrugs and solvates of the compounds of the invention are also contemplated herein. The term "prodrug", as employed herein, denotes a compound that is a drug precursor which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of Formula I or a salt and/or solvate thereof. A discussion of prodrugs is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems (1987) 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, (1987) Edward B. Roche, ed., American Pharmaceutical Association and Pergamon Press, both of which are incorporated herein by reference thereto.
"Solvate" means a physical association of a compound of this invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate" encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates, and the like. "Hydrate" is a solvate wherein the solvent molecule is H20.
"Effective amount" or "therapeutically effective amount" is meant to describe an amount of compound or a composition of the present invention effective in inhibiting the CDK(s) and thus producing the desired therapeutic, ameliorative, inhibitory or preventative effect.
The compounds of Formula I can form salts which are also within the scope of this invention. Reference to a compound of Formula I herein is understood to include reference to salts thereof, unless otherwise indicated. The term "salt(s)", as employed herein, denotes acidic salts formed with inorganic and/or organic acids, as well as basic salts formed with inorganic and/or organic bases. In addition, when a compound of Formula I contains both a basic moiety, such as, but not limited to a pyridine or imidazole, and an acidic moiety, such as, but not limited to a carboxylic acid, zwitterions ("inner salts") may be formed and are included within the term "salt(s)" as used herein. Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred, although other salts are also useful. Salts of the compounds of the Formula I may be formed, for example, by reacting a compound of Formula I with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an -aqueous-medium-followed-by lyophilizationr
Exemplary acid addition salts include acetates, ascorbates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, fumarates, hydrochlorides, hydrobromides, hydroiodides, lactates, maleates, methanesulfonates, naphthalenesulfonates, nitrates, oxalates, phosphates, propionates, salicylates, succinates, sulfates, tartarates, thiocyanates, toluenesulfonates (also known as tosylates,) and the like. Additionally, acids which are generally considered suitable for the formation of pharmaceutically useful salts from basic pharmaceutical compounds are discussed, for example, by P. Stahl et al, Camille G. (eds.) Handbook of Pharmaceutical Salts. Properties, Selection and Use. (2002) Zurich: Wiley-VCH; S. Berge et al, Journal of Pharmaceutical Sciences (1977) 66(1 ) 1-19; P. Gould, International J. of Pharmaceutics (1986) 33 201-217; Anderson et al, The Practice of Medicinal Chemistry (1996), Academic Press, New York; and in The Orange Book (Food & Drug Administration, Washington, D.C. on their website). These disclosures are incorporated herein by reference thereto.
Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as dicyclohexylamines, t-butyl amines, and salts with amino acids such as arginine, lysine and the like. Basic nitrogen-containing groups may be quarternized with agents such as lower alkyl halides (e.g. methyl, ethyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g. dimethyl, diethyl, and dibutyl sulfates), long chain halides (e.g. decyl, lauryl, and stearyl chlorides, bromides and iodides), aralkyl halides (e.g. benzyl and phenethyl bromides), and others.
All such acid salts and base salts are intended to be pharmaceutically acceptable salts within the scope of the invention and all acid and base salts are considered equivalent to the free forms of the corresponding compounds for purposes of the invention.
Compounds of Formula I, and salts, solvates and prodrugs thereof, may exist in their tautomeric form (for example, as an amide or imino ether). All such tautomeric forms are contemplated herein as part of the present invention.
All stereoisomers (for example, geometric isomers, optical isomers and the like) of the present compounds (including those ofthe salts, solvates and prodrugs of the compounds as well as the salts and solvates of the prodrugs), such as those which may exist due to asymmetric carbons on various substituents, including enantiomeric forms (which may exist even in the absence of asymmetric carbons), rotameric forms, atropisomers, and diastereomeric forms, are contemplated within the scope of this invention, as are positional isomers (such as, for example, 4-pyridyl and 3-pyridyl). Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers. The chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations. The use of the terms "salt", "solvate" "prodrug" and the like, is intended to equally apply to the salt, solvate and prodrug of enantiomers, stereoisomers, rotamers, tautomers, positional isomers, racemates or prodrugs of the inventive compounds.
Polymorphic forms of the compounds of Formula I, and of the salts, solvates and prodrugs of the compounds of Formula I, are intended to be included in the present invention.
The compounds according to the invention have pharmacological properties; in particular, the compounds of Formula I can be inhibitors of TACE, TNF-α, ADAM and/or MMP activity.
In one aspect, the invention provides a pharmaceutical composition comprising as an active ingredient at least one compound of formula 1.
In another aspect, the invention provides a pharmaceutical composition of formula 1 additionally comprising at least one pharmaceutically acceptable carrier.
In another aspect, the invention provides a method of treating disorders associated with TACE, TNF-α, MMPs, ADAMs or any combination thereof or any combination thereof, said method comprising administering to a patient in need of such treatment a pharmaceutical composition which comprises therapeutically effective amounts of at least one compound of formula 1.
In another aspect, the invention provides a use of a compound of ~formula~ for the~manufactϋre"of a~medicament to treat disorders associated with TACE, TNF-α, MMPs, ADAMs or any combination thereof.
The compounds of Formula I can have anti-inflammatory activity and/or immunomodulatory activity and can be useful in the treatment of diseases including but not limited to septic shock, haemodynamic shock, sepsis syndrome, post ischaemic reperfusion injury, malaria, mycobacterial infection, meningitis, psoriasis, congestive heart failure, fibrotic diseases, cachexia, graft rejection, cancers such as cutaneous T-cell lymphoma, diseases involving angiogenesis, autoimmune diseases, skin inflammatory diseases, inflammatory bowel diseases such as Crohn's disease and colitis, OA and RA, ankylosing spondylitis, psoriatic arthritis, adult Still's disease, ureitis, Wegener's granulomatosis, Behcehe disease, Sjogren's syndrome, sarcoidosis, polymyositis, dermatomyositis, multiple sclerosis, sciatica, complex regional pain syndrome, radiation damage, hyperoxic alveolar injury, periodontal disease, HIV, non-insulin dependent diabetes mellitus, systemic lupus erythematosus, glaucoma, sarcoidosis, idiopathic pulmonary fibrosis, bronchopulmonary dysplasia, retinal disease, scleroderma, osteoporosis, renal ischemia, myocardial infarction, cerebral stroke, cerebral ischemia, nephritis, hepatitis, glomerulonephritis, cryptogenic fibrosing aveolitis, psoriasis, transplant rejection, atopic dermatitis, vasculitis, allergy, seasonal allergic rhinitis, reversible airway obstruction, adult respiratory distress syndrome, asthma, chronic obstructive pulmonary disease (COPD) and/or bronchitis. It is contemplated that a compound of this invention may be useful in treating one or more of the diseases listed.
In another aspect, the invention provides a method of preparing a pharmaceutical composition for treating the disorders associated with TACE, TNF-α, MMPs, ADAMs or any combination thereof, said method comprising bringing into intimate contact at least one compound of formula 1 and at least one pharmaceutically acceptable carrier.
In another aspect, the invention provides a compound of formula (I) exhibiting TACE, TNF-α, MMPs, ADAMs or any combination thereof inhibitory activity, including enantiomers, stereoisomers and tautomers of said compound, and pharmaceutically acceptable salts or solvates of said compound, said compound being selected from the compounds of structures listed below:
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
50
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
17
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
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
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000100_0002
Figure imgf000100_0003
Figure imgf000100_0004
Figure imgf000100_0005
Figure imgf000100_0006
Figure imgf000101_0001
Figure imgf000101_0002
Figure imgf000101_0003
Figure imgf000101_0004
Figure imgf000101_0005
Figure imgf000101_0006
Figure imgf000101_0007
Figure imgf000101_0008
Figure imgf000101_0009
Figure imgf000102_0001
Figure imgf000102_0002
Figure imgf000102_0003
Figure imgf000102_0004
Figure imgf000102_0005
Figure imgf000102_0006
XX X C σ NA OH OJL o
Figure imgf000102_0007
Figure imgf000103_0001
Figure imgf000103_0002
r χ^ζl0 rQ
Figure imgf000103_0003
Figure imgf000103_0004
Figure imgf000103_0005
Figure imgf000103_0006
Figure imgf000103_0007
Figure imgf000103_0008
Figure imgf000103_0009
Figure imgf000104_0001
Figure imgf000104_0002
Figure imgf000104_0003
Figure imgf000104_0004
Figure imgf000104_0005
Figure imgf000104_0006
Figure imgf000104_0007
Figure imgf000105_0001
Figure imgf000105_0002
Figure imgf000105_0003
Figure imgf000105_0004
Figure imgf000105_0005
Figure imgf000105_0006
Figure imgf000105_0007
σ a W^ nθ
Figure imgf000106_0001
Figure imgf000106_0002
Figure imgf000106_0003
Figure imgf000106_0004
Figure imgf000106_0005
σ1
Figure imgf000106_0006
Figure imgf000106_0007
Figure imgf000106_0008
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000108_0002
Figure imgf000109_0001
Figure imgf000109_0002
Figure imgf000109_0003
Figure imgf000109_0004
Figure imgf000109_0005
Figure imgf000110_0001
Figure imgf000110_0002
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
In a preferred embodiment, the compound is selected from the group consisting of:
Figure imgf000163_0002
Figure imgf000164_0001
Figure imgf000165_0001
Figure imgf000166_0001
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
The above preferred compounds have TACE Ki values of less than 20nM.
In a more preferred embodiment, the compound is selected from the group consting of :
Figure imgf000185_0002
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
The above more preferred compounds have TACE Kj values of less than 5 nM.
In another aspect, the invention provides a pharmaceutical composition for treating disorders associated with TACE, TNF-α, MMP, ADAM or any combination thereof in a subject comprising, administering to the subject in need of such treatment a therapeutically effective amount of a compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
In another aspect, the invention provides a compound of formula 1 in purified form.
In another aspect, the invention provides a method of treating a condition or disease mediated by TACE, MMPs, TNF-σ, aggrecanase (such as aggrecanase 1 or aggrecanase 2), or any combination thereof in a subject comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof. In another aspect, the invention provides a method of treating a condition or disease selected from the group consisting of rheumatoid arthritis, osteoarthritis, periodontitis, gingivitis, corneal ulceration, solid tumor growth and tumor invasion by secondary metastases, neovascular glaucoma, inflammatory bowel disease, multiple sclerosis and psoriasis in a subject, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease selected from the group consisting of fever, cardiovascular conditions, hemorrhage, coagulation, cachexia, anorexia, alcoholism, acute phase response, acute infection, shock, graft versus host reaction, autoimmune disease and HIV infection in a subject comprising administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease selected from the group consisting of septic shock, haemodynamic shock, sepsis syndrome, post ischaemic reperfusion injury, malaria, mycobacterial infection, meningitis, psoriasis, congestive heart failure, fibrotic diseases, cachexia, graft rejection, cancers such as cutaneous T-cell lymphoma, diseases involving angiogenesis, autoimmune diseases, skin inflammatory diseases, inflammatory bowel diseases such as Crohn's disease and colitis, osteo and rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, adult Still's disease, ureitis, Wegener's granulomatosis, Behcehe disease, Sjogren's syndrome, sarcoidosis, polymyositis, dermatomyositis, multiple sclerosis, sciatica, complex regional pain syndrome, radiation damage, hyperoxic alveolar injury, periodontal disease, HIV, non- insulin dependent diabetes mellitus, systemic lupus erythematosus, glaucoma, sarcoidosis, idiopathic pulmonary fibrosis, bronchopulmonary dysplasia, retinal disease, scleroderma, osteoporosis, renal ischemia, myocardial infarction, cerebral stroke, cerebral ischemia, nephritis, hepatitis, glomerulonephritis, cryptogenic fibrosing aveolitis, psoriasis, transplant rejection, atopic dermatitis, vasculitis, allergy, seasonal allergic rhinitis, reversible airway obstruction, adult respiratory distress syndrome, asthma, chronic obstructive pulmonary disease (COPD) and bronchitis in a subject comprising administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with COPD, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with rheumatoid arthritis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with Crohn's disease, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with psoriasis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
In another aspectrthe-invention provides- a- method of treating a condition or disease associated with ankylosing spondylitis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with sciatica, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with complex regional pain syndrome, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with psoriatic arthritis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
In another aspect, the invention provides a method of treating a condition or disease associated with multiple sclerosis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof, in combination with a compound selected from the group consisting of Avonex®, Betaseron, Copaxone or other compounds indicated for the treatment of multiple sclerosis.
Additionally, a compound of the present invention may be co- administered or used in combination with disease-modifying antirheumatic drugs (DMARDS) such as methotrexate, azathioprine, leflunomide, pencillinamine, gold salts, mycophenolate mofetil, cyclophosphamide and other similar drugs. They may also be co-administered with or used in combination with NSAIDS such as piroxicam, naproxen, indomethacin, ibuprofen and the like; COX-2" selective inhibitors Tsϋch as Vioxx® and Celebrex®; Jmmunosuppressives such as steroids, cyclosporin, Tacrolimus, rapamycin and the like; biological response modifiers (BRMs) such as Enbrel®, Remicade®, IL-1 antagonists, anti-CD40, anti-CD28, IL-10, anti- adhesion molecules and the like; and other anti-inflammatory agents such as p38 kinase inhibitors, PDE4 inhibitors, other chemically different TACE inhibitors, chemokine receptor antagonists, Thalidomide and other small molecule inhibitors of pro-inflammatory cytokine production. Also, a compound of the present invention may be co-administered or used in combination with an H1 antagonist for the treatment of seasonal allergic rhinitis and/or asthma. Suitable H1 antagonists may be, for example, Claritin®, Clarinex®, Allegra®, or Zyrtec®.
In another aspect, the invention provides a method of treating a condition or disease mediated by TACE, MMPs, TNF-α, aggrecanase, or any combination thereof in a subject comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of formula 1 or a pharmaceutically acceptable salt, solvate or isomer thereof in combination with a therapeutically effective amount of at least one medicament selected from the group consisting of disease modifying anti-rheumatic drugs (DMARDS), NSAIDs, COX-2 inhibitors, COX-1 inhibitors, immunosuppressives, biological response modifiers (BRMs), anti- infammatory agents and H1 antagonists.
In another aspect, the invention provides a method of treating a condition or disease selected from the group consisting of rheumatoid arthritis, osteoarthritis, periodontitis, gingivitis, corneal ulceration, solid tumor growth and tumor invasion by secondary metastases, neovascular glaucoma, inflammatory bowel disease, multiple sclerosis and psoriasis in a subject, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof in combination with a therapeutically effective amount of at least one medicament selected from the group consisting of DMARDS, NSAIDs, COX-2 inhibitors, COX-1 inhibitors, immunosuppressives, BRMs, anti-infammatory agents and H1 antagonists.- -
In another aspect, the invention provides a method of treating a condition or disease selected from the group consisting of septic shock, haemodynamic shock, sepsis syndrome, post ischaemic reperfusion injury, malaria, mycobacterial infection, meningitis, psoriasis, congestive heart failure, fibrotic diseases, cachexia, graft rejection, cancers such as cutaneous T-cell lymphoma, diseases involving angiogenesis, autoimmune diseases, skin inflammatory diseases, inflammatory bowel diseases such as Crohn's disease and colitis, osteo and rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, adult Still's disease, ureitis, Wegener's granulomatosis, Behcehe disease, Sjogren's syndrome, sarcoidosis, polymyositis, dermatomyositis, multiple sclerosis, sciatica, complex regional pain syndrome, radiation damage, hyperoxic alveolar injury, periodontal disease, HIV, non- insulin dependent diabetes mellitus, systemic lupus erythematosus, glaucoma, sarcoidosis, idiopathic pulmonary fibrosis, bronchopulmonary dysplasia, retinal disease, scleroderma, osteoporosis, renal ischemia, myocardial infarction, cerebral stroke, cerebral ischemia, nephritis, hepatitis, glomerulonephritis, cryptogenic fibrosing aveolitis, psoriasis, transplant rejection, atopic dermatitis, vasculitis, allergy, seasonal allergic rhinitis, reversible airway obstruction, adult respiratory distress syndrome, asthma, chronic obstructive pulmonary disease (COPD) and bronchitis in a subject comprising administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof in combination with a therapeutically effective amount of at least one medicament selected from the group consisting of DMARDS, NSAIDs, COX-2 inhibitors, COX-1 inhibitors, immunosuppressives, BRMs, anti-infammatory agents and H1 antagonists.
In another aspect, the invention provides a method for treating RA comprising administering a compound of the formula I in combination with compound selected from the class consisting of a COX-2 inhibitor e.g. Celebrex® or Vioxx®; a COX-1 inhibitor e.g. Feldene®; an immunosuppressive e.g. methotrexate or cyclosporin; a steroid e.g. β- inethasonerand anti-TNF^"~cdmpound; eTg.Εnbrel® or Remicade®; a PDE IV inhibitor, br other classes of compounds indicated for the treatment of RA.
In another aspect, the invention provides a method for treating multiple sclerosis comprising administering a compound of the formula I in combination with a compound selected from the group consisting of Avonex®, Betaseron, Copaxone or other compounds indicated for the treatment of multiple sclerosis. TACE activity is determined by a kinetic assay measuring the rate of increase in fluorescent intensity generated by TACE catalyzed cleavage of an internally quenched peptide substrate (SPDL-3). The purified catalytic domain of recombinant human TACE (rhTACEc, Residue 215 to 477 with two mutation (S266A and N452Q) and a 6xHis tail) is used in the assay. It is purified from the baculovirus/Hi5 cells expression system using affinity chromatography. The substrate SPDL-3 is an internally quenched peptide (MCA-Pro-Leu-Ala-Gln-Ala-Val-Arg-Ser-Ser-Ser-Dpa-Arg-NH2), with its sequence derived from the pro-TNF cleavage site. MCA is (7- Methoxycoumarin-4-yl)acetyl. Dpa is N-3-(2,4-Dinitrophenyl)-L-2,3- diaminopropionyl.
A 50 μl assay mixture contains 20 mM HEPES, pH 7.3, 5 mM CaCI2, 100 μM ZnCI2, 2 % DMSO, 0.04% Methylcellulose, 30 μM SPDL-3, 70 pM rhTACEc and a test compound. RhTACEc is pre-incubated with the testing compound for 90 min. at 25 °C. Reaction is started by addition of the substrate. The fluorescent intensity (excitation at 320 nm, emission at 405 nm) was measured every 45 seconds for 30 min. using a fluorospectrometer (GEMINI XS, Molecular Devices). Rate of enzymatic reaction is shown as Units per second. Effect of a test compound is shown as % of TACE activity in the absence of the compound.
TACE inhibitory activities for representative compounds are shown in the table below. Kj values greater than 1000 nM (1 μM) are designated as D class. Kj values between 100 nM (0.1 μM) and 1000 nM (1 μM)are designated as C class. Kj values between 20 nM (0.02 μM) and 100 nM (0.1 μM) are designated as B class. Kj values less than 20 nM (0.02 μM) are designated as A class.
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
Figure imgf000207_0001
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
Figure imgf000211_0001
Figure imgf000212_0001
Figure imgf000213_0001
Figure imgf000214_0001
Figure imgf000215_0001
Figure imgf000216_0001
Figure imgf000217_0001
Figure imgf000218_0001
Figure imgf000219_0001
Figure imgf000220_0001
Figure imgf000221_0001
Figure imgf000222_0001
Figure imgf000223_0001
Figure imgf000224_0001
Figure imgf000225_0001
Figure imgf000226_0001
Figure imgf000227_0001
Figure imgf000228_0001
Figure imgf000229_0001
Figure imgf000230_0001
Figure imgf000231_0001
Figure imgf000232_0001
Figure imgf000233_0001
Figure imgf000234_0001
Figure imgf000235_0001
Figure imgf000236_0001
Figure imgf000237_0001
Figure imgf000238_0001
Figure imgf000239_0001
Figure imgf000240_0001
Figure imgf000241_0001
Figure imgf000242_0001
Figure imgf000243_0001
Figure imgf000244_0001
Figure imgf000245_0001
Figure imgf000246_0001
Figure imgf000247_0001
Figure imgf000248_0001
Figure imgf000249_0001
Figure imgf000250_0001
Figure imgf000251_0001
Figure imgf000252_0001
Figure imgf000253_0001
Figure imgf000254_0001
Figure imgf000255_0001
Figure imgf000256_0001
Figure imgf000257_0001
Figure imgf000258_0001
Figure imgf000259_0001
Figure imgf000260_0001
Figure imgf000261_0001
Figure imgf000262_0001
Figure imgf000263_0001
Figure imgf000264_0001
Figure imgf000265_0001
Figure imgf000266_0001
Figure imgf000267_0001
Figure imgf000268_0001
Figure imgf000269_0001
Figure imgf000270_0001
Figure imgf000271_0001
Figure imgf000272_0001
Figure imgf000273_0001
Figure imgf000274_0001
Figure imgf000275_0001
Figure imgf000276_0001
Figure imgf000277_0001
Figure imgf000278_0001
Figure imgf000279_0001
Figure imgf000280_0001
Figure imgf000281_0001
Figure imgf000282_0001
Figure imgf000283_0001
Figure imgf000284_0001
Figure imgf000285_0001
Figure imgf000286_0001
Figure imgf000287_0001
Figure imgf000288_0001
Figure imgf000289_0001
Figure imgf000290_0001
Figure imgf000291_0001
Figure imgf000292_0001
Figure imgf000293_0001
Figure imgf000294_0001
Figure imgf000295_0001
Figure imgf000296_0001
Figure imgf000297_0001
Figure imgf000298_0001
Figure imgf000299_0001
Figure imgf000300_0001
Figure imgf000301_0001
Figure imgf000302_0001
Figure imgf000303_0001
Figure imgf000304_0001
Figure imgf000305_0001
Figure imgf000306_0001
Figure imgf000307_0001
Figure imgf000308_0001
Figure imgf000309_0001
Figure imgf000310_0001
Figure imgf000311_0001
Figure imgf000312_0001
Representative examples of compounds of the invention with their respective Kj values are listed in the table below:
Figure imgf000312_0002
Figure imgf000313_0001
Figure imgf000314_0001
Figure imgf000315_0001
Figure imgf000316_0001
, The pharmaceutical compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets. These excipients , may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be coated by the technique described in the U.S. Pat. Nos. 4,256,108; 4,166,452; and 4,265,874 to form osmotic therapeutic tablets for controlled release.
Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredients is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or a soft gelatin capsules where in the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
Aqueous suspensions contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example, heptadecaethylene-oxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example, polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives, for example, ethyl or n-propyl, p- hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame.
Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example, arachis oil, olive oil, sesame oil or coconut oil, or in mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example, beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid. Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, e.g., sweetening, flavoring and coloring agents, may also be present.
The pharmaceutical compositions of the invention may also be in the form of an oil-in-water emulsion. The oily phase may be a vegetable oil, e.g., olive oil or arachis oil, or a mineral oil, e.g., liquid paraffin or mixtures of these. Suitable emulsifying agents may be naturally-occurring phosphatides, e.g., soy beans, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example, sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, e.g., polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening and flavoring agents.
Syrups and elixirs may be formulated with sweetening agents, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, e.g., as a solution in 1 ,3-butane diol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
Compounds of the invention may also be administered in the form of suppositories for rectal administration of the drug. The compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials are cocoa butter and polyethylene glycols.
For topical use, creams, ointments, jellies, solutions or suspensions, etc., containing the compound of The invention are employed. (For purposes of this application, topical application shall include mouthwashes and gargles.)
The compounds for the present invention can be administered in the intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art. To be administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen. Compounds of the present invention may also be delivered as a suppository employing bases such as cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol.
The dosage regimen utilizing the compounds of the present invention is selected in accordance with a variety of factors including type, species, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound thereof employed. A physician or veterinarian of ordinary skill can readily determine and prescribe the effective amount of the drug required to prevent, counter, arrest or reverse the progress of the condition. Optimal precision in achieving concentration of drug within the range that yields efficacy without toxicity requires a regimen based on the kinetics of the drug's availability to target sites. This involves a consideration of the distribution, equilibrium, and elimination of a drug. Preferably, doses of the compound of Formula I useful in the method of the present invention range from 0.01 to 1000 mg per day. More preferably, dosages range from 0.1 to 1000 mg/day. Most preferably, dosages range from 0.1 to 500 mg/day. For oral administration, the compositions are preferably provided in the form of tablets containing 0.01 to 1000 milligrams of the active ingredient, particularly 0.01 , 0.05, 0.1 , 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100 and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. An effective amount of the drug is ordinarily supplied at a dosage level of from about 0.0002 mg/kg to about 50 mg/kg of body weight per day. The range is more particularly from about 0.001 mg/kg to 1 mg/kg of body weight per day.
Advantageously, the active agent of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in dividend doses of two, three or four time daily.
The amount of active ingredient that may be combined with the carrier materials to produce single dosage form will vary depending upon the host treated and the particular mode of administration.
It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the age, body weight, general health, sex, diet, time of administration, route or administration, rate of excretion, drug combination and the severity of the particular disease undergoing therapy.
The compounds of the invention may be produced by processes known to those skilled in the art and as shown in the following reaction schemes and in the preparations and examples described below.
EXAMPLES
The following abbreviations are used in the procedures and schemes:
ACN Acetonitrile
AcOH Acetic acid
ADDP 1 ,11-(Azodicarbonyl)dipiperidine
Anh. Anhydrous
Aq Aqueous
BOC tert-Butoxycarbonyl
°C degrees Celsius
CBZCI Benzyl chloroformate
GDI Carbodiimide
DBU 1 ,8-Diazabicyclo[5.4.0]undec-7-ene
DCC Dicyclohexylcarbodiimide
DCM Dichloromethane DEAD Diethyl azodicarboxylate
(DHQ)2PHAL Hydroquinine 1 ,4-phthalazinediyl diether
DIAD Diisopropylazodicarboxylate
DIEA Diisopropylethylamine
DMA N,N-Dimethylacetamide
DMAP 4-Dimethylaminopyridine
DME Dimethoxyethane
DMF Dimethylformamide
DMFDMA N,N-Dimethylformamide dimethylacetal
DMPU 1 ,3-Dimethyl-3,4,5,6-tetrahydro-2(1 h)-pyrimidinone
DMSO Dimethyl sulfoxide
EDC 1 -(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
El Electron ionization
Eq Equivalents
EtOAc Ethyl acetate
EtOH Ethanol g grams h. hours
1H proton
HATU N,N,N',N'-Tetramethyl-0-(7-Azabenzotriazol-1-yl)Uronium hexafluorophosphate
Hex hexanes
HOBt 1 -Hydroxybenzotriazole
HPLC High pressure liquid chromatography
LAH Lithium aluminum hydride
LDA Lithium diisopropylamide
M Molar mCPBA meta-Chloroperoxybenzoic acid
Me Methyl
MeCN Acetonitrile
MeOH Methanol min Minutes mg Milligrams
MHz Megahertz ml Milliliter
MS Mass Spectroscopy
NMM N-Methylmorpholine
NMP 1-methyl-2-pyrrolidone
ON Overnight
Pd(*Bu3P)2 Bis-(tri-terf-butylophosphine)palladium
Pd(TPP) Tetrakis-(triphenylphosphine)palladium
Pd(Oac)2 Palladium(ll) acetate
PdC (TPP)2 Bis-(triphenylphosphine)palladium(ll) chloride
PdCI2(ddppf) Dichloro[1 ,1 '-bis(diphenylphosphino)ferrocene]palladium(ii) dichloride Pd2(dba)3 Tris(dibenzylideneacetone)dipalladium(0) PyBrOP Bromo-tris-pyrrolidino-phosphonium hexafluorophosphate
Pyr Pyridine
RT Room temperature
Si02 Silica gel 60 chromatography sgc Silica gel 60 chromatography tBOC tert-Butoxycarbonyl
TACE TNF-alpha converting enzyme
TEA Triethylamine
TFA Trifluoroacetic acid
THF Tetrahydrofuran
TLC Thin layer chromatography
TPP Triphenylphosphine tR Retention time
NMR spectra were acquired on a Mercuryplus 400 MHz NMR Spectrometer (Varian), using CDCI3 or DMSO-d6 as solvents. LC-MS data was obtained using an Agilent 1100 Series LC/MSD (quadrupole, API-ES (Atmospheric Pressure Interface Electrospray)) with a capillary voltage set to 3500 V and running in positive mode. Reported analytical HPLC (LC/MS) retention times were obtained using a C18 (150 x 4.6 mm) reverse-phase column eluting with a 5 or 10 minute gradient of 0.1 % trifluoroacetic acid in water to 95:5 acetonitrile:water at a flow rate of 3 mL/min. Purification via reverse phase chromatography was accomplished using a C18 reverse phase column with a gradient of 0.1 % trifluoroacetic acid in water to 95:5 acetonitrile:water at a flow rate of 20 mL/min. Samples were collected using a UV (Gilson, 254 nm) or mass spectra (Agilent 1100 Series LC/MSD model SL) signal.
Normal phase silica gel chromatography on a Biotage instrument was accomplished using a Quad UV System (P/N 07052) utilizing KP-SIL 32-63 um columns, 6θA with flash cartridges 12+M or 25+M.
The compounds of formula (I) may be produced by processes known to those skilled in the art and as shown in the following reaction schemes and in the preparations and examples described below. These preparations and examples should not be construed to limit the scope of the disclosure. Alternate mechanistic pathways and analogous structures may be apparent to those skilled in the art. Some of the compounds made by these processes are listed in Table 1. All kinds of isomeric forms of the compounds are considered to be within the scope of this invention.
Example 1 : General synthesis of tartrate inhibitors
Two general routes exist for the synthesis of tartrate diamides from amines. The first (Example 1 ) utilizes an acetonide-protected monoacid/monoester intermediate prepared from the commercially available acetonide dimethyl ester using a literature procedure (J. Am. Chem. Soc. 1978, 100, 4865-4872). In general either Example 1 or Example 2 could be used interchangeably, although Example 2 was found to be more preferred for compounds that contained functional groups that were unstable towards acidic deprotection conditions (such as 113).
A variety of amide bond coupling reagents were acceptable, including HATU, GDI, EDC, DCC/HOBt, PyBrOP, polymer supported CDI with HOBt, polymer supported carbodiimide, and polymer supported EDC (PS-EDC) with HOBt. These coupling reagents could be used with a variety of bases, including triethylamine (TEA), diisopropylethylamine (DIEA), N-methyl morpholine, pyridine, dimethylaminopyridine (DMAP) and imidazole. In some cases the excess amines in the peptide coupling steps were removed using liquid/liquid extraction or polymeric scavenging resins such as polymer supported isocyanate (PS-NCO) and/or polymer supported tosic acid (MP- TsOH). Unreacted acids could be removed using MP-carbonate resin or polymer resins containing basic functional groups such as trisamine (i.e. PS- trisamine), amberiite, or morpholine. Peptide couplings were conducted in a variety of solvents, including DMF, THF, dioxane, acetonitrile, NMP, and DCM. These solvents can also be combined in various proportions to optimize the reaction conditions. A less preferred but viable route using pentafluorophenyl (PFP) ester intermediates may be also utilized to prepare mono or diamides; preferred solvents for this approach should include THF. General peptide coupling strategies, including PFP-based approaches, can be found in Bodanszky & Bodanszky, The Practice of Peptide Synthesis, second edition, Springer-Verlag, 1994.
Cleavage (saponification) of the intermediate methyl ester could be accomplished under a variety of well-known conditions; including: a slight excess (1.1-3 equivalents) of base, KOH in methanol, LiOH in THF/water, LiOH in methanol/water, and NaOH/THF/MeOH/water. Removal of the acetonide protecting group could be afforded using a variety of acidic conditions, including TFA:water combinations (such as 80:20).
Figure imgf000324_0001
Part A: To 2,2-dimethyl-[1 ,3]dioxolane-4R,5R-dicarboxylic acid monomethyl ester (1) (Musich, J.A.; Rapoport, H.; J. Am. Chem. Soc. 1978, 00, 4865-4872) (500 mg, 2.45 mmol) in DMF (5 mL) was added 2-thiopheneethyIamine (316 μL, 2.70 mmol), DIEA (0.94 mL, 5.4 mmol) and HATU (989 mg, 2.60 mmol). The reaction mixture was stirred overnight and the DMF was removed in vacuo. The residue was dissolved in EtOAc, washed with water, saturated bicarbonate solution, 0.1 N HCI, and brine. The organic layer was dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 10% EtOAc/DCM) afforded 2 as an oil (491 mg, 64%). 1H NMR (400 MHz, CDCI3) δ 7.17 (dd, 1 H , J = 1.2, 5.2 Hz), 6.95 (dd, 1 H, J = 2.1 , 5.2 Hz), 6.84 (d, 1 H, J = 2.4 Hz), 6.66 (bs, 1 H, NH), 4.72 (ABq, 2H, J = 13.2 Hz), 3.84 (s, 3H), 3.65 (m, 1 H), 3.57 (m, 1 H), 3.09 (t, 2H, J = 6.4 Hz), 1.47 (s, 3H), 1.40 (s, 3H); HPLC-MS tR = 1.63 min (UV254 nm); Mass calculated for formula C14H19N05S 313.1 , observed LCMS m/z 314.2 (M+H).
Part B:
To 2 (869 mg, 2.77 mmol) in THF (10 mL) was added 1.0 M LiOH (3 mL, 3 mmol) and the reaction was stirred overnight at room temperature. The reaction mixture was diluted with water (10 mL) and the THF was removed in vacuo. The basic aqueous layer was extracted with diethyl ether and the ether wash was discarded. The aqueous layer was made acidic with 1.0 N HCI and extracted with diethyl ether. The combined organic layers were dried over sodium sulfate and concentrated to afford 3 as a yellow solid (443 mg, 53%). 1H NMR (400 MHz, CDCI3) δ 7.21 (dd, 1 H, J = 1.2, 5.2 Hz), 6.97 (dd, 1 H, J = 3.6, 5.2 Hz), 6.90 (bs, 1 H, NH), 6.86 (d, 1 H, J = 3.2 Hz), 4.48 (ABq, 2H, J = 9.2 Hz), 3.76 (m, 1 H), 3.60 (m, 1 H), 3.15 (m, 2H), 1.53 (s, 3H), 1.41 (s, 3H); HPLC-MS tR = 1.34 min (UV254 nm); Mass calculated for formula Cι3H17N05S 299.1 , observed LCMS m/z 300.1 (M+H).
Part C:
To 3 (150 mg, 0.5 mmol) in DMF (2 mL) was added racemic 2-(4-fluoro- phenyl)-pyrrolidine (99 mg, 0.6 mmol), DIEA (261 μL, 1.5 mmol), and HATU (228 mg, 0.6 mmol) and the reaction mixture was stirred overnight at room temperature. The DMF was removed in vacuo and the residue was dissolved in ethyl acetate. The organic layer was washed with 0.1 N NaOH, 0.1 N HCI, water, and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 10% to 50% EtOAc/DCM) afforded an oil (220 mg, 99%). The diastereomers were resolved (as described in Example 18) by reverse phase HPLC to afford 4 the desired isomer (78 mg, 35%) after lypholization. HPLC-MS tR = 1.93 min (UV254 nm); Mass calculated for formula C23H27FN2O4S 446.2, observed LCMS m/z 447.3 (M+H).
Part D:
Compound 4 (78 mg, 0.17 mmol) was dissolved in 90:10 TFA:water (5 mL) and stirred for 4 hours at room temperature. The reaction mixture was quenched with 1 :1 ACN:water (10 mL) and concentrated. The residue was re-dissolved in 1 :1 ACN:water (10 mL) and concentrated. Lypholization afforded 5 as white solid (62 mg, 87%). 1H NMR (400 MHz, CDCI3) mixture of rotomers δ 7.16- 6.8 (m, 8H), 5.23-5.11 (m, 1 H), 4.85 (m, 1 H), 4.40-4.15 (m, 1 H), 3.90-3.48 (m, 6H), 3.00 (m, 2H), 2.33 (m, 1 H), 2.05-1.83 (m, 3H); HPLC- MS tR = 1.50 min (UV254 nm); Mass calculated for formula C2oH23FN204S 406.1 , observed LCMS m/z 407.2 (M+H).
The following table contains compounds synthesized using the above procedures.
Figure imgf000326_0001
Figure imgf000327_0001
Figure imgf000328_0001
Figure imgf000329_0001
Figure imgf000330_0001
Figure imgf000331_0001
Figure imgf000332_0002
Example 2: Anhydride route to tartrate diamide inhibitors
A second general route to diamide compounds starting from an anhydride intermediate is delineated below in Example 2. (+)-Diacetyl -L-tartaric anhydride (48) is an article of commerce and reacts readily with a variety of amines to generate monoacid/monoamide intermediate. The preferred solvent for the ring opening is DCM; although DMF, THF or dioxane may also be compatible. The subsequent peptide coupling step proceeds under a variety of standard conditions which are given for Example 1. The acetate groups can be removed using a variety of conditions, including hydrazine/methanol, NaOMe in methanol, ammonia/methanol, lithium hydroxide/THF/water (saponification), potassium carbonate in methanol, or MP-carbonate. Often times the preferred set of conditions for parallel synthesis is to use the MP-carbonate. It has also been observed that one or both of the acetate protecting groups can be cleaved during synthetic transformations involving nucleophiles, basic reagents, heating in protic solvents or exposure to organometallic reagents.
Example 2A:
Figure imgf000332_0001
Part A:
To 48 (26 mg, 0.1 mmol) in DMF (1 mL) was added 2,4- difluorophenylpiperazine (20 mg, 0.1 mmol). The reaction mixture was stirred for 1.5 hours. To the crude mixture was added 2-thiopheneethylamine (14 μL, 0.12 mmol), DIEA (38 μL, 0.22 mmol) and HATU (42 mg, 0.11 mmol). The reaction was stirred overnight at room temperature. The reaction mixture was poured into water and extracted with EtOAc. The combined organic layers were washed with 0.1 N NaOH, 0.1 N HCI, and brine; dried over sodium sulfate and concentrated in vacuo to yield 49 as an oil (49 mg, 94%). HPLC- MS tR = 2.10 min (UV254 nm); mass calculated for formula C-24H27F2N3O6S 523.2, observed LCMS m/z 524.4 (M+H); purity > 95% (ELSD).
Part B:
To 49 (49 mg, 0.09 mmol) in MeOH (2 mL) was added anhydrous hydrazine (5 μL, 0.16 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was concentrated in vacuo and freeze- dried to afford 50 as a white powder (40 mg, 100%). 1H NMR (400 MHz, CDCI3) δ 7.17 (d, 1 H, J = 5.2 Hz), 6.96 (m, 2H), 6.85 (m, 4H), 4.87 (s, 1 H), 4.24 (s, 1 H), 3.90 (m, 2H), 3.72 (m, 2H), 3.60 (m, 4H), 3.08 (m, 4H); HPLC- MS tR = 1.85 min (UV254 nm); mass calculated for formula C20H23F2N3O4S 439.1 , observed LCMS m/z 440.2 (M+H).
Example 2B: KA>
Figure imgf000333_0001
Part A:
To 48 (26 mg, 0.1 mmol) in DMF (1 mL) was added N-methyl-(1f?-phenyl- ethyl)-amine (15 μL, 0.1 mmol). The reaction mixture was stirred for 1 hour. To the crude mixture was added 2-thiopheneethylamine (47 μL, 0.4 mmol), HOBt (27 mg, 0.2 mmol), PS-carbodiimide resin (312 mg, 0.4 mmol). To the reaction mixture was added PS-TsOH resin (0.6 mmol) and MP-carbonate resin (0.4 mmol). The reaction was tumbled overnight, filtered and concentrated. Compound 51 was used without further purification. HPLC-MS tR = 1.82 min (UV254 nm); mass calculated for formula C23H28N2O6S 460.2, observed LCMS m/z 461.1 (M+H).
Part B:
To 51 in MeOH (2 mL) was added anhydrous hydrazine (5 μL, 0.16 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was concentrated in vacuo, purified by reverse phase prep-HPLC and freeze-dried to afford 52 as a white powder (6.7 mg, 18% overall). 1H NMR (400 MHz, CDCI3) major rotomer δ 7.40- 7.25 (m, 6H), 7.16 (d, 1 H, J = 4.8 Hz), 6.95 (app. t, 1 H, J = 4.8 Hz), 6.87 (bs, 1 H, NH), 5.96 (q, 1 H, J = 7.2 Hz), 4.91 (d, 1 H, J = 1.2 Hz), 4.22 (d, 1 H, J = 1.2 Hz), 3.60 (m, 2H), 3.08 (m, 2H), 2.79 (s, 3H), 1.56 (d, 3H, J = 5.7Hz); HPLC-MS tR = 3.80 min (UV254 nm , 10 min); mass calculated for formula CιgH24N2θ4S 376.2, observed LCMS m/z 377.2 (M+H).
Example 2C:
Figure imgf000334_0001
Compound 53 was prepared according to the procedure described by D. Miller et al (J. Med. Chem. 1999, 42, 2287).
Parts A & B:
Compound 55 was prepared using procedures similar to those described in
Example 14, Parts D & E.
Data for 54: 1HNMR (400 MHz, CDCI3) δ 7.51-7.25 (m, 6H), 6.22 (s, 1 H), 5.75-
5.67 (m, 2H), 5.36 (s, 2H), 5.28-4.61 (m, 4H), 3.69-3.40 (m, 2H), 2.74 (s, 1 H),
2.24 (s, 3H), 2.18 (s, 3H). Data for 55: 1HNMR (400 MHz, CDCI3) δ 7.54-7.26
(m, 6H), 6.24 (s, 1 H), 5.22-5.02 (m, 2H), 4.94-4.82 (m, 3H) 4.42 (s, 1H), 3.72-
3.50 (m, 2H), 2.92-2.76 (m, 2H) MS (El) m/z M+H Obsd 377.1
Figure imgf000335_0001
Figure imgf000336_0001
Figure imgf000337_0001
Figure imgf000338_0001
Figure imgf000339_0001
Figure imgf000340_0001
Example 2D:
Method A
Figure imgf000340_0002
48 106 107 108
Method A, Step 1 :
Compound 106 (R1 = R2 = CH2Ph) was prepared from 48 following the procedure described in Example 4A, Part C. Method A, Step 2:
To a THF/MeCN (1 :1 , 0.5 mL) solution of polystyrene EDC resin (57 mg, 3 eq) in a 6 mL fritted cartridge was added a THF/MeCN (1 :1 ) solution of 106 (0.028 mmol), a THF solution of HOBT (0.3 mL, 1.5 eq) and a 1 M THF/MeCN (1 :1 ) solution of 2-furanethylamine (0.056 mL, 2 eq). The cartridge was capped and let shake at 25°C for 20 h. Polystryrene isocyanate resin (57 mg, 3 eq) and polystyrene trisamine (39 mg, 6 eq) was added followed by 0.5 mL of THF and the mixture was shaken for 6 h. The suspension was filtered and the filtrate was concentrated to afford 107 (R1 = R2 = CH2Ph, R3 = CH2CH2(C4H40).
Method A, Step 3:
Compound 107 was treated with a 2N solution of NH3 in MeOH (3 mL) for 1.5 h. The solvent was then removed in vacuo to provide 108 (R1 = R2 = CH2Ph, R3 = CH2CH2(C4H40).
Example 2D Table A
Figure imgf000341_0001
Figure imgf000342_0001
Figure imgf000343_0001
Method B
Figure imgf000344_0001
48 B2 B3 B4
Q OAcμ μ O OH u
**- A V2 OAcYOYV O - A RV2 OH OY O *
B5 B6
Method B, Step 1 :
Compound B2 (R1 = CH2Ph, R2 = CH3) was prepared from 48 following the procedure described in Example 4A Part C.
Method B, Step 2:
To a DMF (40 mL) solution of B2 (2.0 g, 5.93 mmol), HOBT (880 mg, 6.52 mmol), and N-Boc-1 ,3-propylenediamine (1.14 g, 6.52 mmol) was added
EDCI (1.48 g, 7.61 mmol) at 25 °C under N2. After stirring for 20 h, 1 N HCI was added, the products were extracted with ethyl acetate (3X), combined, then washed with sat. NaHC03, water (3x), and dried over MgS04. The products were then filtered and concentrated in vacuo to give B3 (R1 =
CH2Ph, R2 = CH3, n = 3).
Method B, Step 3:
To a MeCN (30 mL) solution of B3 (1.18 g, 2.4 mmol) at 25°C was added 20 mL of a 4N solution of HCI in dioxane. The solution was capped and stirred at
25 °C for 2.5 h. The solvent was then removed in vacuo. The product was then dissolved in 45 mL of THF/MeCN/DMF (4:4:1 ) and polystyrene NEt2 resin (4.5 g, 14.4 mmol) added. After stirring for 1.5 h, the solution was filtered off and the resin washed with THF:MeCN (1 :1 ). The filtrates were then diluted to 120 mL with additional THF/MeCN (1 :1 ) and B4 (R1 = CH2Ph, R2 = CH3, n
= 3) was used in the preparation of the following library:
Method B, Step 4:
Polystyrene EDC resin (30mg, 0.045 mmol) was added to 96-wells of a deep well polypropylene microtiter plate followed by a MeCN/THF/DMF (6:6:1 ) stock solution (1 mL) of B4 (0.015 mmol) and HOBT (0.0225 mmol). Then 1 M stock solutions of each of the individual acids (R -96COOH) (0.023 mL, 0.021 mmol) were added to the wells, which was then sealed and shaken at 25 °C for 20h. The solutions were filtered through a polypropylene frit into a 2nd microtiter plate containing polystyrene isocyanate resin (3 equivalents, 0.045 mmol) and polystyrene trisamine resin (6 equivalents, 0.09 mmol). After the top plate was washed with MeCN (0.5 mL), the plate was removed, the bottom microtiter plate sealed and shaken at 25 °C for 16h. Then the solutions were filtered through a polypropylene frit into a 96-well collection plate. The wells of the top plate were then washed with MeCN (0.5 mL), and the plate removed. Then the resultant solutions in the collection plate were transferred into vials and the solvents removed in vacuo via a SpeedVac to provide amides B5. Method B, Step 5:
Compounds B6 were prepared from B5 following the procedure described in Method A, Step 3 (see above).
Example 2D Table B
Figure imgf000345_0001
Figure imgf000346_0001
Method C
Figure imgf000347_0001
steP4,
Figure imgf000347_0002
156 157
Method C, Step 1 :
Compound 153 (R R2N = 4-phenylpiperazine) was prepared from 48 following the procedure described in Method B, Step 1 (see above). Method C, Step 2:
Compound 154 (R1R2N = 4-phenylpiperazine, n = 3) was prepared from 153 following the procedure described in Method B, Step 2 (see above). Method C, Step 3:
Compound 155 (R1R2N = 4-phenylpiperazine, n = 3) was prepared from 154 following the procedure described in Method B, Step 3 (see above) and was used in the preparation of the following library: Method C, Step 4:
Polystyrene DIEA resin (30mg, 0.045 mmol) was added to 72-wells of a deep well polypropylene microtiter plate followed by a MeCN/THF/DMF (6:6:1 ) stock solution (1 mL) of 155 (0.015 mmol). Then 1M stock solutions of each of the individual sulfonyl chlorides (R1-72S02CI) (0.023 mL, 0.021 mmol) were added to the wells, which was then sealed and shaken at 25 °C for 20h. The solutions were filtered through a polypropylene frit into a 2nd microtiter plate containing polystyrene isocyanate resin (3 equivalents, 0.045 mmol) and polystyrene trisamine resin (6 equivalents, 0.09 mmol). After the top plate was washed with MeCN (0.5 mL), the plate was removed, the bottom microtiter plate sealed and shaken at 25 °C for 16h. Then the solutions were filtered through a polypropylene frit into a 96-well collection plate. The wells of the top plate were then washed with MeCN (0.5 mL), and the plate removed. Then the resultant solutions in the collection plate were transferred into vials and the solvents removed in vacuo via a SpeedVac to provide sulfonamides 156. Method C, Step 5:
Compounds 157 were prepared from 156 following the procedure described in Method A, Step 3 (see above).
Example 2D Table C
Figure imgf000348_0001
These compounds were prepared using Method A: Example 2D Table D
Figure imgf000348_0002
Figure imgf000349_0001
Figure imgf000350_0001
These compounds were prepared using Method A: Example 2D Table E
Figure imgf000350_0002
Figure imgf000351_0001
Example 3: Heteroaryl biaryl compounds
A compound precursor may be prepared such that the synthesis of direct analogs is facilitated. Examples of schemes for biaryl synthesis are listed below. Aryl halides and 'pseudo-halides' (i.e. triflates) are known to react with boronic acids under a variety of established conditions (Top. Curr. Chem. 2002, 219, 12-49). A variety of bases for this reaction are known in the literature, including sodium carbonate, potassium carbonate, cesium carbonate, potassium t-butoxide, sodium t-butoxide, TEA, DIEA, potassium fluoride, and potassium phosphate. For most applications potassium phosphate was preferred and gave acceptable yields and chemoselectivity. A number of solvents have also been used in the literature for Suzuki reactions, including THF, dioxane, NMP, DMF, DME, DMA, toluene, and water. In general we have found that THF or dioxane are preferred solvents. Solvents may also be mixed in various proportions to enhance reactivity and/or chemoselectivity. Palladium sources for this reaction are numerous as well, including Pd(TPP)4> Pd(OAc)2, PdCI2(TPP)2, PdCI2(dppf), Pd2(dba)3. In general PdCl2(dppf) was found to be a preferred palladium source.
Example 3A:
Figure imgf000352_0001
Part A:
To an ice cold methanol (100 mL) solution of 4-bromo-thiophene-2-carbaldehyde (183) (90%, 11.1 g, 58.1 mmol, 1.00 equiv) was added sodium borohydride in portions (2.20 g, 58.1 mmol, 1.00 equiv) over ca. 10 minutes. The cooling bath was removed and the reaction solution was aged 30 min. The reaction was quenched at rt by the addition of acetone (until evolution of gas ceased), concentrated, and partitioned between ethyl acetate and 0.1 N HCI. The organic phase was washed with water, brine, dried (sodium sulfate), filtered, and then concentrated to give 184 as an orange oil (10.5 g, 95%) that was used without further purification. 1H NMR (400 MHz, CDCI3) δ 7.18 (s, 1 H), 6.93 (s, 1 H), 4.81 (s, 2H).
Part B:
To 184 (4.38 g, 22.7 mmol) in toluene (25 mL) was added phosphorous tribromide (2.36 mL, 25 mmol). The reaction mixture was heated in an oil bath at 90 °C for 15 minutes. After cooling to room temperature the reaction mixture was poured over ice and extracted with ethyl acetate. The combined organic layer was washed with water (2x), saturated sodium bicarbonate solution (1x) and saturated sodium chloride solution, dried over magnesium sulfate and concentrated to afford 185 as a light brown oil (5.51 g, 95%). The material was used without further purification. 1H NMR (400 MHz, CDCI3) δ 7.21 (s, 1 H), 7.03 (s, 1 H), 4.66 (s, 2H). Part C:
To 185 (5.51 g, 21.5 mmol) in DMF (20 mL) was added phthalimide (3.80 g, 25.8 mmol) and cesium carbonate (7.72 g, 23.7 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was filtered and the filtrate was concentrated. The residue was dissolved in ethyl acetate and water. The organic layer was separated, washed with saturated sodium chloride solution, dried over sodium sulfate and concentrated to afford a brown solid. Recrystallization from -30 % ethyl acetate/hexanes afforded 186 as a peach solid (6.38 g, 2 crops, 92%). 1H NMR (400 MHz, CDCI3) δ 7.86 (m, 2H), 7.72 (m, 2H), 7.11 (s, 1 H), 7.07 (s, 1 H), 4.97 (s, 2H). HPLC-MS tR = 2.02 min (UV254 nm); mass calculated for formula C13H8BrN02S 321.0, observed LCMS m/z 322.0 (M+H).
Part D:
A suspension of 186 (5.76 g, 17.8 mmol) and hydrazine hydrate (3.5 mL, 72 mmol) in ethanol (50 mL) was heated to reflux. The suspension cleared upon heating before a thick white precipitate formed. The reaction mixture was heated for 1 hour and cooled. The white precipitate was broken up by the addition of ethanol (50 mL) and sonication of the mixture. The precipitate was removed by filtration and the solids were thoroughly washed with ethanol and ethyl acetate. The filtrate was concentrated in vacuo. The residue was dissolved in ethyl acetate and water. The layers were separated. The organic layer was washed with saturated sodium chloride solution, dried over sodium sulfate and concentrated to afford 187 as an orange-brown oil (2.66 g, 78%). 1H NMR (400 MHz, CDCI3) δ 7.10 (s, 1 H), 6.84 (s, 1 H), 4.04 (s, 2H). HPLC-MS tR = 0.58 min (UV254 nm); Mass calculated for formula C5H6BrNS 190.9, observed LCMS m/z 192.0(M+H).
Part E:
Following the procedure described in Example 1 part A: To 188 (prepared as described in Example 4 Part C from (+)-diacetyl -L-tartaric anhydride (48) and 2- chlorophenylpiperazine) (916 mg, 2.22 mmol) in DMF (5 mL) was added 187 (502 mg, 2.61 mmol), DIEA (850 μL, 4.88 mmol) and HATU (928 mg, 2.44 mmol). Purification by column chromatography (Si02> 20% to 50% EtOAc/DCM) afforded 189 as an off-white foam (652 mg, 50%). 1H NMR (400 MHz, CDCI3) δ 7.37 (dd, 1 H, J = 1.2, 7.6 Hz), 7.25 (dt, 1 H, J = 1.2, 7.6 Hz), 7.14 (d, 1 H, J = 1.2 Hz), 7.03 (m, 2H), 6.92 (d, 1 H, J = 1.2 Hz), 6.74 (m, 1 H, NH), 5.92 (d, 1 H, J = 3.6 Hz), 5.70 (d, 1 H, J = 4.0 Hz), 4.73 (dd, 1 H, J = 6.4, 15.2 Hz), 4.50 (dd, 1H, J = 5.6, 15.6 Hz), 3.83 (m, 3H), 3.65 (m, 1 H), 3.20 - 3.00 (m, 4H), 2.21 (s, 3H), 2.11 (s, 3H); Mass calculated for formula
Figure imgf000354_0001
585.0, observed LCMS m/z 586.1 (M+H).
Part F:
To 189 (59 mg, 0.1 mmol), phenyl boronic acid (13 mg, 0.11 mmol), potassium phosphate (42 mg, 0.2 mmol), and PdCI2(dppf) (4 mg, 0.005 mmol) was added dioxane (2 mL). The reaction mixture was flushed with argon and heated to 80 °C overnight. The reaction mixture was cooled, poured into water and extracted with EtOAc. The combined organics were washed with brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 20% EtOAc/DCM) afforded 190 as an off-white foam (36 mg, 58%, 94% purity). HPLC-MS tR = 5.61 min (UV254 nm, 10 min.); mass calculated for formula C29H30CIN3O6S 583.2, observed LCMS m/z 584.2 (M+H).
Part G:
To 190 (36 mg, 0.062 mmol) in methanol was added 0.5 M sodium methoxide in methanol (6 μL, 0.003 mmol). The reaction mixture was stirred for 1.5 hours at room temperature, quenched with 0.1 N HCI and concentrated. Purification by reverse- phase prep-LC afforded 191 as a white solid (3.2 mg, 10%). 1H NMR (400 MHz, CDCI3) δ 7.55 (dd, 2H, J = 1.2, 8.4 Hz), 7.40 to 7.20 (m, 9H), 7.09 (m, 1 H, NH), 4.91 (d, 1 H, J = 2.4 Hz), 4.70 (m, 2H), 4.33 (d, 1 H, J = 2.0 Hz), 4.00 (m, 2H), 3.85 (m, 2H), 3.20 (m, 4H); HPLC-MS tR = 1.94 min (UV254 nm); Mass calculated for formula C25H26CIN304S 499.1 , observed LCMS m/z 500.2 (M+H).
Figure imgf000354_0002
Example 3B: 3-Pyridyl-Biaryl
Figure imgf000355_0001
Part A:
To 193 (prepared as described in Example 1 ) (1.5 g, 4.24 mmol) in DMF (10 mL) was added 194 (0.725 g, 5.09 mmol) and HATU (1.94 g, 5.09 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was diluted with ethyl acetate, washed with saturated sodium bicarbonate and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 80% ethyl acetate/hexanes) afforded 195 as a colorless oil (1.73 g, 86%). HPLC-MS tR = 1.92 and 1.97 min (UV254 nm); Mass calculated for formula C23H25Cl2N304477.1 , observed LCMS m/z 478.1 (M+H).
Part B:
Stepl : To phenyl boronic acid (10 mg, 0.084 mmol), potassium phosphate (36 mg, 0.168 mmol) and PdCI2(dppf) (3.1 mg, 10 mol%) under argon in a 4- mL vial was added 195 (20 mg, 0.042 mmol) in dioxane (0.5 mL). The reaction mixture was heated at 100 °C for 16 hours. The reaction mixture was cooled to room temperature, diluted with ethyl acetate and filtered through celite. The filtrate was concentrated. HPLC-MS tR = 2.01 min (UV254 „m); Mass calculated for formula C29H30CIN3O4 519.2, observed LCMS m/z 520.2 (M+H).
Step 2: The crude material from Step 1 was dissolved in 80:20 TFA:water (1 mL) at 0 °C and stirred at room temperature for 2 hours. The reaction mixture was quenched with 1 :1 water:acetonitrile (2 mL) and concentrated. Purification by reverse phase prep-LC afforded 196 as a solid after lypholization. HPLC-MS tR = 3.54 min (UV254 nm, 10 min); Mass calculated for formula C26H26CiN3θ4479.2, observed LCMS m/z 480.2 (M+H).
Figure imgf000356_0001
Figure imgf000357_0001
Example 3C: 2-Pyridyl-Biaryl
Figure imgf000357_0002
Part A:
To 5-bromo-pyridine-2-carbonitrile (209) (1.0 g, 5.46 mmol) in tetrahydrofuran (10 mL) at -78 °C was added lithium aluminum hydride (1.0 M, 13.66 mL, 13.66 mmol). The reaction was stirred for 2 hours at -78 °C. The mixture was quenched at -78 °C with 10:1 THF:water. The mixture was warmed to room temperature, diluted with ethyl acetate and stirred with 1.0 N sodium hydroxide for 10 minutes. The reaction mixture was filtered through celite and the layers were separated. The organic layer was washed with 1.0 N sodium hydroxide and brine, dried over sodium sulfate and concentrated to afford 210 as an oil (600 mg). The material was used without further purification. Part B:
To 193 (1.13 g, 3.2 mmol) in DMF (10 mL) was added 210 (0.600 g, 3.2 mmol) and HATU (1.34 g, 3.52 mmol). The reaction mixture was stirred for 5 hours at room temperature. The reaction mixture was diluted with ethyl acetate, washed with saturated sodium bicarbonate and brine, dried over sodium sulfate and concentrated to afford 211 as an oil (380 mg). HPLC-MS tR = 2.00 and 2.05 min (UV254 nm); Mass calculated for formula C23H25BrCIN304 521.1 , observed LCMS m/z 522.1 (M+H).
Part C:
Stepl : To 2-chlorophenyl boronic acid (24 mg, 0.152 mmol), potassium phosphate (65 mg, 0.306 mmol) and PdCI2(dppf) (3.1 mg, 5 mol%) under argon in a 4-mL vial was added 211 (40 mg, 0.076 mmol) in dioxane (0.5 mL). The reaction mixture was heated at 100 °C for 16 hours. The reaction mixture was cooled to room temperature, diluted with ethyl acetate and filtered through celite. The filtrate was concentrated.
Step 2: The crude material from Step 1 was dissolved in 80:20 TFA:water (1 mL) at 0 °C and stirred at room temperature for 2 hours. The reaction mixture was quenched with 1 :1 wateπacetonitrile (2 mL) and concentrated. Purification by reverse phase prep-LC afforded 212 as a solid after lypholization. HPLC-MS tR = 4.10 and 4.13 min (UV254 nm, 10 min); Mass calculated for formula C26H25Cl2N304 513.2, observed LCMS m/z 514.2 (M+H).
The following compounds were prepared using the above procedures.
Figure imgf000358_0001
Figure imgf000359_0001
Figure imgf000360_0001
Example 4: Thiophene-benzyl compounds
Pd-mediated Negishi-type couplings were an efficient way to generate a set of relevant compounds for this series where Ar was a phenyl ring. While a range of palladium sources are known we found that Pd(PfBu3)2 was preferred. The reaction can be run in a variety of solvent systems, including dioxane, THF, or DMA. A preferred method utilized the solvent from the zinc reagent, which was typically available as a THF stock solution. Removal of the t-BOC group can be completed using a range of acidic conditions including TFA/water, HCI in methanol, and HCI in dioxane; ail of which proceeded equally well with most reaction substrates. The reactions are typically concentrated and freeze-dried to give HCI or TFA salts. The Negishi reactions proceed comparably, and in many cases better, when the amine was protected by a phthalimide. The phthalimide protecting group could be removed by heating in ethanol containing hydrazine hydrate.
Example 4A:
Figure imgf000361_0001
Part A:
To 186 (3.47 g, 10.8 mmol) and bis-(tri-terf-butyl-phosphine) palladium (0.28 g, 0.54 mmol) in a flask under argon was added 2-chlorobenzyl zinc chloride (0.5 M in THF, 54 mL, 27 mmol). The reaction was stirred at room temperature for 3 hours. The reaction mixture was diluted with EtOAc (200 mL) and washed with saturated ammonium chloride solution (100 mL), bicarbonate solution (100 mL) and brine (100 mL). The organic layer was dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 20% EtOAc/Hex) afforded 228 as a white solid (3.59 g, 91%). 1H NMR (400 MHz, CDCI3) δ 7.85 (dd, 2H, J = 3.2, 5.6 Hz), 7.71 (dd, 2H, J = 3.2, 5.6 Hz), 7.36 (m, 1 H), 7.17 (m, 3H), 6.99 (d, 1 H, J = 1.0 Hz), 6.78 (d, 1 H, J = 1.0 Hz), 4.95 (s, 2H), 4.00 (s, 2H).
Part B:
To 228 (3.55 g, 9.72 mmol) suspended in ethanol (35 mL) was added hydrazine monohydrate (1.9 mL, 38.9 mmol). The reaction mixture was heated at reflux for 2 hours. After cooling the solids were removed by filtration and washed with ethanol (100 mL) and ethyl acetate (50 mL). The filtrate was concentrated and the residue was dissolved in ethyl acetate (150 mL) and water (100 mL). The organic layer was separated, washed with brine (100 ml), dried over sodium sulfate and concentrated to yield 229 as a yellow oil (2.54 g, 99%).
Part C: To 48 (2.16 mg, 10.0 mmol) in DCM (10 mL) was added isoindoline (1.13 mL, 10.0 mmol). The reaction was stirred at room temperature for 2 hours. The solvent was removed in vacuo and the residue was partitioned between ethyl acetate and 1.0 N HCI. The organic layer was separated, washed with water and brine, dried over sodium sulfate and concentrated to afford 230 as a dark solid (3.35 g, 100%). 1H NMR (400 MHz, DMSO-d6) δ 13.63 (bs, 1 H), 7.38 - 7.28 (m, 4H), 5.01 (d, 1 H, J = 13.2 Hz), 4.93 (d, 1 H, J = 14.0 Hz), 4.71 (d, 1 H, J = 16.4 Hz), 4.56 (d, 1 H, J = 15.6 Hz), 2.12 (s, 3H), 2.07 (s, 3H); HPLC-MS tR = 1.10 min (UV254 nm); mass calculated for formula C16H17N07 335.1 , observed LCMS m/z 336.1 (M+H).
Part D:
To 230 (18 mg, 0.053 mmol) in NMP (2 mL) was added 229 (19 mg, 0.08 mmol), DIEA (26 μL, 0.148 mmol) and HATU (30 mg, 0.08 mmol). The reaction mixture was stirred overnight. The reaction mixture was poured into water and extracted with ethyl acetate. The combined organic layer was washed with 0.1 N NaOH, 0.1 N HCI and brine, dried over sodium sulfate and concentrated to afford 231.
Part E:
To 231 (118 mg, 0.216 mmol) in methanol (3 mL) and DCM (3 mL) was added MP- carbonate resin (2.54 mmol/g, 85 mg). The reaction was stirred for 3 hours, filtered and concentrated to afford 232 as a white solid (94 mg, 92%). 1H NMR (400 MHz, DMSO-de) δ 8.34 (t, 1 H, J = 2.0 Hz) 7.79 (dd 1 H, J = 0.8, 8.0 Hz), 7.64 (dt, 1 H, J =1.6, 8.0 Hz), 7.47 (d, 1 H, J = 7.6 Hz), 7.40 (dt, 1 H, J = 0.8, 7.6 Hz), 7.35 - 7.27 (m, 4H), 6.97 (d, 1 H, J = 1.6 Hz), 6.81 (d, 1 H, J = 1.2 Hz), 5.67 (d, 1 H, J = 6.4 Hz), 5.05 (d, 1H, J = 12.8 Hz), 4.97 (d, 1 H, J = 8.0 Hz), 4.90 (d, 1H, J = 13.2 Hz), 4.75 (d, 1 H, J = 13.2 Hz), 4.60 (m, 2H), 4.36 (m, 2H, J = 6.4, 15.2 Hz) 4.24 (dd, 1H, J = 3.2, 6.8 Hz); HPLC-MS tR = 1.85 (UV254 nm); mass calculated for formula C24H23CIN204S 470.1 , observed LCMS m/z 471.1
Example 4B:
Figure imgf000363_0001
Part A:
To 233 (4.00 g, 13.7 mmol) and bis-(tri-terf-butyl-phosphine) palladium (0.28 g, 0.54 mmol) in a flask under argon was added 2-chlorobenzyl zinc chloride (0.5 M in THF, 69 mL, 34.5 mmol). The reaction was stirred at room temperature overnight. The reaction mixture was diluted with EtOAc (200 mL) and washed with saturated ammonium chloride solution (100 mL), bicarbonate solution (100 mL) and brine (100 mL). The organic layer was dried over sodium sulfate and concentrated. Purification by column chromatography (Siθ2, 15% EtOAc/Hex) afforded 234 as a white solid (4.5 g, 95%). 1H NMR (400 MHz, CDCI3) δ 7.36 (d, 1 H J =3.6 Hz), 7.25 - 7.18 (m, 4H), 6.74 (s, 1 H), 6.64 (d, 1 H, J = 3.6 Hz), 4.40 (d, 2H, J = 5.2 Hz), 4.22 (s, 2H).
Part B:
To 234 (450 mg, 1.34 mmol) was added 4M HCI in dioxane (1.5 mL) and the reaction was stirred for 30 minutes. The dioxane was removed in vacuo. The residue was dissolved in THF (10 mL) and DIEA (500 μL, 2.68 mmol) and 48 (275 mg, 1.27 mmol) were added. The reaction was stirred overnight at room temperature. The reaction mixture was diluted with ethyl acetate (60 mL) and washed with 1.0 N HCI. The organic layer was dried over sodium sulfate and concentrated to afford 235 as a yellow foam (505 mg, 90%).
Part C:
To 235 (40 mg, 0.09 mmol) in DMF (2 mL) were added DIEA (50 μL, 0.29 mmol), isoindoline (14 mg, 0.12 mmol), and HATU (45 mg, 0.12 mmol). The reaction mixture was stirred for 6 hours. The reaction mixture was diluted with ethyl acetate (30 mL) and washed with 0.1 N NaOH, 0.1 N HCI and brine. The organic layer was dried over sodium sulfate. The crude product was dissolved in methanol (3 mL) and potassium carbonate (100 mg) in water (1 mL) was added. After stirring for 30 minutes the reaction mixture was diluted with ethyl acetate (20 mL) and washed with brine. The organic layer was dried over sodium sulfate and concentrated. Purification by reverse phase prep-LC afforded 236 as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.29 (t, 1 H, J = 6.4 Hz) 7.42 (dd, 1 H, J = 1.6, 7.8 Hz), 7.37 - 7.23 (m, 7H), 6.74 (d, 1 H, J = 3.6 Hz), 6.65 (d, 1 H, J = 3.2 Hz), 5.66 (d, 1H, J = 5.6 Hz), 5.04 (d, 1 H, J = 14.8 Hz), 4.97 (bs, 1 H), 4.89 (d, 1 H, J = 15.2 Hz), 4.75 (d, 1 H, J = 16.4 Hz), 4.60 (m, 2H), 4.38 (dd, 1 H, J = 6.4, 15.2 Hz), 4.23 (bs, 1 H), 4.15 (s, 2H); HPLC-MS tR = 4.92 min (UV254 nm, 10 min); mass calculated for formula C24H23CIN204S 470.1 , observed LCMS m/z 471.1
Example 4C: Thiophene-Benzyl
B
Figure imgf000364_0001
Compound 237 was prepared according to the procedure described by A. J. Hutchison et al (European Patent 0323807 A2, Dec. 7, 1989)
Parts A:
Compound 238 was prepared by procedures similar to those described in Example 5
Part A.
Parts B-E:
Compound 242 was prepared by procedures similar to those described in Example 4B Parts A to C. HPLC-MS tR = 2.05 min (UV254 nm); mass calculated for formula C27H28CIFN204S 530.1 , observed LCMS m/z 531.1 (M+H). The following compounds were prepared by the procdures desribed in Example 4A 4C.
Figure imgf000365_0001
Figure imgf000366_0001
Figure imgf000367_0001
Figure imgf000368_0001
Figure imgf000369_0001
Figure imgf000370_0001
Figure imgf000371_0001
Figure imgf000372_0001
Example 5:
Figure imgf000372_0002
Part A:
To a solution of 187 (2.68 g, 14.0 mmol) and pyridine (2.26 mL, 28 mmol) in dichloromethane (25 mL) was added di-ferf-butyl dicarbonate (3.21 g, 14.7 mmol). The mixture was stirred overnight at room temperature. The reaction mixture was poured into water (25 mL) and the layers were separated. The organic layer was washed with 1.0 N HCI (20 mL), water (20 mL), and saturated sodium chloride solution (20 mL), dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, DCM) afforded 305 as a light orange solid (3.64 g, 89%). 1H NMR (400 MHz, CDCI3) δ 7.11 (s, 1 H), 6.87 (s, 1 H), 4.01 (bs, 1 H), 4.44 (d, 2H, J = 6.0 Hz), 1.48 (s, 9H).
Part B:
To π-butyl lithium (2.5 M, 0.31 mL, 0.78 mmol) in THF (2 mL) at -78 °C under argon was added 305 (100 mg, 0.34 mmol) in THF (1.5 mL) dropwise. The reaction mixture was stirred for 30 minutes at -78 °C. Then 2-thiophene carbaldehyde (60 μL, 0.68 mmol) was added. The reaction mixture was stirred for 30 minutes at -78 °C. The reaction mixture was quenched with saturated ammonium chloride solution and warmed to room temperature. The mixture was diluted with ethyl acetate (10 mL) and the layers were separated. The organic layer was washed with water and brine. Dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 20% EtOAc/hexane) afforded 306 (47 mg, 42%). 1H NMR (400 MHz, CDCI3) δ 7.28 (m, 1 H), 7.16 (s, 1 H) 6.95 (m, 2H), 6.93 (s, 1 H), 6.05 (s, 1 H). 4.88 (bs, 1 H), 4.43 (d, 2H, J = 5.6 Hz), 2.43 (bs, 1 H), 1.48 (s, 9H).
Part C:
To 306 (47 mg, 0.14 mmol) in DCM (2 mL) was added triethylsilane (0.2 mL). The mixture was cooled in an ice bath and TFA (0.2 mL) was added. The mixture was warmed to room temperature and stirred overnight. The solvents were removed in vacuo and 307 was used without further purification. 1H NMR (400 MHz, CD3OD) δ 7.20 (m, 2H), 7.09 (s, 1 H) 6.90 (m, 1 H), 6.86 (m, 1 H), 4.26(s, 2H), 4.15 (s, 2H).
Part D:
To 230 (19 mg, 0.06 mmol) in DMF (2 mL) was added 307 (18 mg, 0.06 mmol), DIEA (29 μL, 0.17 mmol) and HATU (28 mg, 0.07 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was diluted with ethyl acetate (20 mL) and water (20 mL). The layers were separated and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with sodium bicarbonate solution and brine, dried over sodium sulfate and concentrated. Compound 308 was used without further purification. HPLC-MS tR 1.91 min (UV254 nm); mass calculated for formula C26H26N2O6S2 526.1 , observed LCMS m/z 269.9 (M+H).
Part E:
Compound 308 and potassium carbonate (20 mg) were mixed in methanol (2.5 mL) and water (0.5 mL) and were stirred for 30 minutes. The reaction mixture was diluted with ethyl acetate, washed with water and brine, dried over sodium sulfate and concentrated. Purification by reverse phase prep-LC afforded 309 as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.33 (t, 1 H, J = 5.6 Hz), 7.30 (m, 5H), 7.03 (s, 1 H), 6.92 (s, 1 H), 6.67 (s, 1 H), 6.82 (s, 1 H), 5.04 (d, 1H, J = 14.0 Hz), 4.90 (d, 1H, J = 14.0 Hz), 4.75 (d, 1 H, J = 16.0 Hz), 4.60 (m, 2H), 4.41 (dd, 1 H, J = 6.8, 14.8 Hz), 4.33 (dd, 1 H, J = 5.6, 14.4 Hz), 4.25 (s, 1 H), 4.04 (s, 2H); HPLC-MS tR = 4.45 min (UV254 nm, 10 min); mass calculated for formula C22H22N2O4S2442.1 , observed LCMS m/z 443.0 (M+H).
The following compounds were prepared using the procedures described above. Part C did not result in dehydration for compounds like 314 and 317.
Figure imgf000374_0001
Figure imgf000375_0001
Example 6
Example 6A:
Figure imgf000376_0001
Part A:
A mixture of 2-(aminomethyl)thiophene (319) (1.57 g, 13.8 mmol), monomethyl phthalate (2.75 g, 15.3 mmol), EDC (3.26 g, 16.6 mmol), HOBt (2.79 g, 20.7 mmol) and triethylamine (5 mL, 27.6 mmol) in DCM (40 mL) was stirred for 12 hours. The reaction mixture was partitioned between ethyl acetate and water. The aqueous layer was extracted with ethyl acetate (2 x 50 mL). The combined organic layers were washed with 1.0 N HCI, bicarbonate solution and brine, dried over sodium sulfate and concentrated. Recrystallization of the mixture from ethyl acetate/hexanes afforded 320 as a solid (2.20 g, 67%). 1H NMR (400 MHz, CDCI3) δ 7.85 (m, 2H), 7.71 (m, 2H), 7.21 (m, 1 H), 7.15 (m, 1 H), 6.94 (m, 1 H), 5.03 (s, 2H).
Part B: (Feigel, M., Lugert, G., Heichert, C; Liebigs Ann. Chem. 1987, 367) To paraformaldehyde (250 mg) in HCI (cone, 5 mL) was added 0.5 M zinc chloride in THF (1.7 mL, 0.85 mmol). To this mixture was added 320 (235 mg, 0.85 mmol) in portions and then dioxane (3 mL). The reaction mixture was stirred at 60 °C for 45 minutes. The reaction mixture was cooled and diluted with water (50 mL) and ethyl acetate (50 mL). The layers were separated and the aqueous layer was extracted with ethyl acetate (2 x 30 mL). The combined organic layers were dried over sodium sulfate and concentrated. Recrystallization from ethyl acetate / hexane afforded 321 as a solid (190 mg, 78%). 1H NMR (400 MHz, CDCI3) δ 7.86 (dd, 2H, J = 3.2, 5.6 Hz), 7.72 (dd, 2H, J = 3.2, 5.6 Hz), 6.98 (d, 1 H, J = 3.6 Hz), 6.90 (d, 1 H, J = 3.6 Hz), 4.98 (s, 2H), 4.72 (s, 2H).
Part C:
A mixture 321 (190 mg, 0.65 mmol), 2-methyibenzimidazole (112 mg, 0.85 mmol), sodium iodide (catalytic) and cesium carbonate (275 mg, 0.85 mmol) in DMF (6 mL) were stirred overnight at room temperature. The reaction mixture was partitioned between ethyl acetate and water. The layers were separated and the aqueous layer was extracted with ethyl acetate (2 x 50 mL). The combined organic layers were washed with sodium bicarbonate solution and brine, dried over sodium sulfate and concentrated. Recrystallization from ethyl acetate / hexane afforded 322 (65 mg) and 322 contaminated with 2-methylbenzimidazole (120 mg). 1H NMR (400 MHz, CDCI3) δ 7.83 (dd, 2H, J = 3.2, 5.6 Hz), 7.70 (dd, 2H, J = 3.2, 5.6 Hz), 7.68 (m, 1 H), 7.29 (m, 1 H), 7.23 (m, 2H), 6.95 (d, 1 H, J = 3.6 Hz), 6.70 (d, 1 H, J = 3.6 Hz), 5.36 (s, 2H), 4.91 (s, 2H), 2.64 (s, 3H); HPLC-MS tR = 1.14 min (UV254 nm); mass calculated for formula C22H17N3O2S 387.1 , observed LCMS m/z 388.1 (M+H).
Part D:
Compound 322 (90 mg, 0.23 mmol) and hydrazine hydrate (50 μL, 0.93 mmol) in ethanol (10 mL) and DCM (5 mL) were refluxed for 1.5 hours. The reaction mixture was cooled and filtered. The filtrate was concentrated and the crude product was used without further purification.
Part E:
To 230 (83 mg, 0.24 mmol) in DMF (5 mL) was added 323 (82 mg, 0.32 mmol), DIEA (300 μL, 1.6 mmol) and HATU (135 mg, 0.36 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was diluted with ethyl acetate (50 mL) and water (50 mL). The layers were separated and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with sodium bicarbonate solution and brine, dried over sodium sulfate and concentrated. Compound 324 was used without further purification. HPLC-MS tR = 1.13 min (UV254 nm); mass calculated for formula C30H30N4O6S 574.2, observed LCMS m/z 575.1 (M+H).
Part F:
Compound 324 (-120 mg, 0.21 mmol) and potassium carbonate (100 mg) were mixed in methanol (5 mL) and water (1 mL). After 5 minutes a solid precipitated and the reaction was stirred for 30 minutes. The solid was collected by filtration. Purification by reverse phase prep-LC afforded 325 as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.35 (t, 1 H, J = 6.0 Hz), 7.93 (d, 1 H, J = 8.0 Hz), 7.73 (d, 1H, J = 6.8 Hz), 7.48 (m, 2H), 7.33 (m, 1H), 7.27 (m, 3H), 7.08 (d, 1 H, J = 3.6 Hz), 6.82 (d, 1 H, J = 3.6 Hz), 5.80 (s, 2H), 5.02 (d, 1 H, J = 14.4 Hz), 4.86 (d, 1 H, J = 15.2 Hz), 4.74 (d, 1 H, J = 16.8 Hz), 4.58 (d, 1 H, J = 16.4 Hz), 4.57 (s, 1 H), 4.33 (m, 2H), 4.22 (s, 1 H), 2.84 (s, 3H); HPLC-MS tR = 2.74 min (UV254 nm> 10 min); mass calculated for formula C26H26N404S 490.2, observed LCMS m/z 491.2 (M+H).
Example 6B:
Figure imgf000379_0001
Part A:
To n-butyl lithium (2.5 M, 0.91 mL, 2.28 mmol) in THF (5 mL) at -78 °C under argon was added 305 (300 mg, 1.03 mmol) in THF (5 mL) dropwise. The reaction mixture was stirred for 30 minutes at -78 °C. Then dimethyl formamide (151 mg, 2.06 mmol) was added. The reaction mixture was stirred for 30 minutes at -78 °C. The reaction mixture was quenched with saturated ammonium chloride solution and warmed to room temperature. The mixture was diluted with ethyl acetate (10 mL) and the layers were separated. The organic layer was washed with water and brine. Dried over sodium sulfate and concentrated. The residue was dissolved in methanol and sodium borohydride (156 mg, 4.12 mmol) was added. The reaction mixture was stirred overnight at room temperature. The reaction mixture was diluted with ethyl acetate and washed with 0.1 N HCI, water and brine. The organic layer was dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 25% EtOAc/hexane) afforded 326 (147 mg, 58%). 1H NMR (400 MHz, CDCI3) δ 7.10 (s, 1 H), 6.94 (s, 1 H), 4.63 (s, 2H), 4.45 (d, 2H, J = 4.7 Hz), 1.48 (s, 9H).
Part B:
To 326 (30 mg, 0.12 mmol) in THF (2 mL) was added benzimidazole (19 mg, 0.16 mmol), triphenylphosphine (42 mg, 0.16 mmol) and DIAD (32 mg, 0.16 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was concentrated. The residue was dissolved in dichloromethane (2 mL) and TFA (0.5 mL) was added. The reaction mixture was stirred for 30 minutes and concentrated. The residue was dissolved in diethyl ether and washed twice with water. The combined aqueous layer was made basic DIEA and extracted with ethyl acetate. The combined organic layer was washed with brine, dried and concentrated to afford 327 as a film (41 mg). The material was used without further purification.
Part C:
To 230 (20 mg, 0.06 mmol) in DMF (2 mL) was added 327 (19 mg, 0.08 mmol), DIEA (31 μL, 0.18 mmol), DMAP (1 mg) and HATU (30 mg, 0.08 mmol). The reaction mixture was stirred overnight. The mixture was poured into water and extracted with ethyl acetate. The combined organic layers were washed with 0.1 N NaOH, water and brine, dried over sodium sulfate and concentrated. Compound 328 was used without further purification. Mass calculated for formula C2gH28N406S 560.2, observed LCMS m/z 561.2 (M+H).
Part D:
Compound 328 and potassium carbonate (20 mg) were mixed in methanol (2.5 mL) and water (0.5 mL) and were stirred for 30 minutes. The reaction mixture was diluted with ethyl acetate, washed with water and brine, dried over sodium sulfate and concentrated. Purification by reverse phase prep-LC afforded 329 as a white solid (8 mg). HPLC-MS tR = 2.77 min (UV254 nm, 10 min); mass calculated for formula C25H24N40 S 476.2, observed LCMS m/z 477.1 (M+H).
Example 6C
Figure imgf000381_0001
Part A:
N-(Hydroxymethyl)phthalimide (1.0 g, 5.6 mmol) was dissolved in 1% triflic acid in trifluoroacetic acid (10 mL) at 0 °C. To this mixture was added 330 (0.52 mL, 5.6 mmol). The reaction mixture was warmed to room temperature slowly and stirred overnight. The reaction mixture was poured into ice water (100 mL) and extracted with DCM. The combined organic layers were washed with sodium bicarbonate solution and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 30% EtOAc/Hex) afforded 331 (619 mg, 41 %). 1H NMR (400 MHz, CDCI3) δ 9.85 (d, 1 H, J = 1.6 Hz), 7.86 (m, 2H), 7.81 (d, 1 H, J = 1.6 Hz), 7.76 (s, 1H), 7.73 (m, 2H), 4.87 (s, 2H).
Part B:
Compound 331 (315 mg, 1.16 mmol) was dissolved in 1 :1 methylene chloride:methanol (10 mL) and cooled in an ice bath. To this solution was added sodium borohydride (11 mg, 0.29 mmol) and the reaction was stirred for 30 minutes. Additional sodium borohydride (3 mg, 0.08 mmol) was added and the reaction was stirred for 30 minutes. The reaction mixture was diluted with methylene chloride and washed with saturated ammonium chloride solution, water and brine. The reaction mixture was dried over sodium sulfate and concentrated to afford 332 as a white solid (310 mg, 98%). 1H NMR (400 MHz, CD3OD) δ 7.83 (m, 2H), 7.79 (m, 2H), 7.24 (s, 1 H), 6.97 (s, 1 H), 4.76 (s, 2H), 4.65 (s, 2H). Part C:
A mixture of 332 (97 mg, 0.36 mmol) and phosphorous tribromide (34 μL, 0.39 mmol) in DCM (5 mL) was stirred for 30 minutes at room temperature. The reaction mixture was poured into ice water and extracted with ethyl acetate. The combined organic layers were washed with sodium bicarbonate solution and brine, dried over sodium sulfate and concentrated to afford 333 as a white solid (104 mg, 87%). 1H NMR (400 MHz, CDCI3) δ 7.85 (dd, 2H, J = 2.8, 5.2 Hz), 7.72 (dd, 2H, J = 2.8, 5.2 Hz), 7.32 (s, 1 H), 7.15 (s, 1 H), 4.77 (s, 2H), 4.66 (s, 2H).
Part D:
A mixture of 333 (450 mg, 1.34 mmol), 2-methyl-benzimidazole (355 mg, 2.69 mmol) and cesium carbonate (875 mg, 2.69 mmol) in DMF (5 mL) was stirred overnight at room temperature. The reaction mixture was diluted with ethyl acetate and washed with water and brine, dried over sodium sulfate and concentrated. Recrystallization from ethyl acetate / hexanes afforded 334 as a white solid (252 mg, 49%). 1H NMR (400 MHz, CDCI3) δ 7.84 (dd, 2H, J = 2.8, 5.2 Hz), 7.72 (dd, 2H, J = 2.8, 5.2 Hz), 7.68 (m, 1 H), 7.23 (m, 3H), 7.19 (s, 1 H), 7.05 (s, 1 H), 5.38 (s, 2H), 4.75 (s, 2H), 2.66 (s, 3H).
Part E:
A mixture of 334 (225 mg, 0.58 mmol) and hydrazine hydrate (113 μL, 2.32 mmol) in ethanol (4 mL) was refluxed for 4 hours. The reaction mixture was cooled and filtered. The filtrate was concentrated and the residue was dissolved in ethyl acetate. The organic layer was washed with water and brine, dried over sodium sulfate and concentrated to afford a 335 as yellow solid (148 mg, 99%).
Part F:
To 230 (12 mg, 0.036 mmol) in DMF (2 mL) was added 335 (12 mg, 0.045 mmol), DIEA (18 μL, 0.11 mmol) and HATU (17 mg, 0.045 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was diluted with ethyl acetate (20 mL) and water (20 mL). The layers were separated and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with sodium bicarbonate solution and brine, dried over sodium sulfate and concentrated. Compound 336 was used without further purification. HPLC-MS tR = 1.22 min (UV254 nm); mass calculated for formula C3oH3oN406S 574.2, observed LCMS m/z 575.0 (M+H).
Part G:
Compound 336 and potassium carbonate (20 mg) were mixed in methanol (1.5 mL) and stirred for 30 minutes. The reaction mixture was diluted with ethyl acetate, washed with water and brine, dried over sodium sulfate and concentrated. Purification by reverse phase prep-LC afforded 337 as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.23 (t, 1 H, J = 6.4 Hz), 7.89 (m, 1 H), 7.70 (m, 1 H), 7.44 (m, 2H), 7.33 (m, 1 H), 7.27 (m, 3H), 7.20 (s, 1 H), 7.14 (s, 1 H), 5.80 (s, 2H), 5.04 (d, 1 H, J = 13.2 Hz), 4.88 (d, 1 H, J = 15.2 Hz), 4.73 (m, 2H), 4.60 (m, 2H), 4.25 (m, 2H), 2.83 (s, 3H); HPLC-MS tR = 2.04 min (UV254 n . 10 min); mass calculated for formula CH26N 0 S 490.2, observed LCMS m/z 491.1 (M+H).
The following compounds were prepared using the procedures described in Example 6A to 6C.
Figure imgf000383_0001
Figure imgf000384_0001
Figure imgf000385_0002
Example 7:
Figure imgf000385_0001
Part A:
To 232 (26 mg, 0.056 mmol) in toluene (5 mL) was added dibutyltin oxide (20 mg, 0.08 mmol). The mixture was heated for 3 hours at reflux with a Dean Stark trap. The mixture was concentrated. The residue was dissolved in NMP (3 mL) and treated with cesium fluoride (8.5 mg, 0.056 mmol) and iodomethane (14 μL, 0.224 mmol). The reaction mixture was heated at 50 °C overnight. The reaction mixture was poured into water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated. Purification by prep-HPLC afforded 350 (3.4 mg, 13%). HPLC-MS tR = 5.26 min (UV254 nm, 10 min.); mass calculated for formula C25H25CIN2O4S 484.1 , observed LCMS m/z 485.0 (M+H).
Figure imgf000385_0003
Example 8:
Figure imgf000386_0001
PartG
Figure imgf000386_0002
362
Part A:
To BOC-glycine (353) (25.0 g, 0.143 mol) dissolved in dry THF (100 mL) and cooled to -20 °C under a nitrogen atmosphere was added Hunig's base (23.1 g, 31.1 mL, 0.357 mol) then isobutylchloroformate (21.4 g, 20.4 mL, 0.157 mol). Stirred at -20 °C for 60 mins. Serine methyl ester (354) (24.4 g, 0.157 mol) then Hunig's base (23.1 g, 31.1 mL, 0.357 mol) was added. The reaction mixture was warmed to room temperature, stirred for 18 h and concentrated. The residue was partitioned between 0.5 N NaOH (300 mL) CH2CI2. The layers were separated and the acqueous layer was extracted with CH2CI2. The combined organic layer was dried over magnesium sulfate and concentrated. Purified by silica gel chromatography (eluant: 1% MeOH- CH2CI2 to 3% MeOH-CH2CI2) to give 20.5 g (52%) of the product 355 as a yellow oil. MS m/e: 277 (M+H). For n=2: MS m/e: 291 (M+H) Part B:
To compound 355 (4.55g, 16.5 mmol) dissolved in CH2CI2 (200 mL) and cooled to -50 °C under a nitrogen atmosphere was added diethylaminosulfur trifluoride (3.19 g, 2.4 mL, 19.8 mmol) dropwise via syringe. Stirred at -50 °C for 2 h, added potassium carbonate (3.88 g, 28.1 mmol), and warmed slowly to room temperature over 2 h. Added 0.2 N NaOH (100 mL) and separated layers. Extracted aqueous layer with CH2CI2, dried combined organic extracts (MgS04), filtered, and concentrated. Purified by silica gel chromatography (eluant: 2% MeOH-CH2CI2 to 4% MeOH-CH2CI2) to give 3.06 g (72%) of the product 356 as a yellow oil. MS m/e: 259 (M+H). For n=2: MS m/e: 273 (M+H)
Part C:
To compound 356 (6.10 g, 23.6 mmol) dissolved in CH2CI2 (250 mL) and cooled to 0 °C was added DBU (12.59 g, 12.4 mL, 82.7 mmol) and bromotrichloromethane (16.40 g, 8.1 mL, 82.7 mmol). Warmed slowly to room temperature over 2 h and stirred for 16 h. Added 0.1 N NaOH (200 mL) and separated layers. Extracted aqueous layer with CH2CI2, dried combined organic extracts (MgS04), filtered, and concentrated. Purified by silica gel chromatography (eluant: 2% MeOH-CH2CI2 to 3% MeOH-CH2CI2) to give 4.27 g (71%) of the product 357 as an orange oil. MS m/e: 257 (M+H). For n=2: MS m/e: 271 (M+H)
Part D:
To compound 357 (4.25 g, 16.6 mmol) dissolved in Et20 (100 mL) was added lithium borohydride (1.44 g, 66.3 mmol) and MeOH (2.13 g, 2.7 mL, 66.3 mmol). Refluxed for 3 h, cooled to room temperature, and concentrated. Added water (100 mL), extracted with CH2CI2, dried combined organic extracts (MgS0 ), filtered, and concentrated. Purified by silica gel chromatography (eluant: 5% MeOH-CH2CI2 to 10% MeOH-CH2CI2) to give 2.43 g (64%) of the product 358 as a yellow oil. MS m/e: 229 (M+H). For n=2: MS m/e: 243 (M+H)
Part E: To compound 358 (1.41 g, 6.18 mmol) dissolved in CH2CI2 (35 mL) and cooled to -25 °C was added triethylamine (1.25 g, 1.7 mL, 12.4 mmol) then mesyl chloride (0.85 g, 0.57 mL, 7.41 mmol) dropwise. Warmed to 0 °C slowly over 60 mins. Added water (50 mL), extracted with CH2CI2, dried combined organic extracts (MgS0 ), filtered, and concentrated to give 1.89 g (100%) of the product 359 as a yellow oil. MS m/e: 251 (M+2-tBu). For n=2: MS m/e: 265 (M+2-tBu)
Part F:
Suspended copper cyanide (1.66 g, 18.5 mmol) in dry THF (70 mL) under a nitrogen atmosphere and cooled to -25 °C. Added phenyl magnesium bromide (3.0 M in Et20, 12.3 mL, 37.0 mmol) dropwise via syringe such that internal temperature < -20 °C. Stirred at -20 °C for 30 mins then at 0 °C for 30 mins. Warmed to 15 °C internal temperature then recooied to -25 °C. Added compound 359 (1.89 g, 6.18 mmol) dissolved in dry THF (20 mL) dropwise via syringe. Stirred at -25 °C internal temperature for 2 h then at 0 °C for 16 h. Concentrated, added 2 N NH4OH (100 mL), extracted with CH2CI2, dried combined organic extracts (MgS0 ), filtered, and concentrated. Purified by silica gel chromatography (eluant: 5% EtOAc-hexane to 20% EtOAc-hexane) to give 0.82 g (46%) of the product 360A as a yellow oil. MS m/e: 289 (M+H).
The following intermediates were prepared according to the above procedure:
Figure imgf000388_0001
Part G:
To compound 360A (1.00 g, 3.47 mmol) dissolved in 1 :1 CH2CI2:MeOH (30 mL) was added 4 N HCI in dioxane (7.8 mL, 31.2 mmol). Stirred at room temperature for 4 h then concentrated to give 0.78 g (100%) of the product 361 A (hydrochloride salt) as a yellow solid. MS m/e: 189 (M+H).
The following intermediates were prepared according to the above procedure:
Figure imgf000389_0001
Part H: (JFL 80324-023)
Combined compound 361 A (0.20 g, 0.677 mmol), compound 362 (0.30 g, 0.812 mmol), HATU (0.51 g, 1.35 mmol), and triethylamine (0.21 g, 0.28 mL, 2.03 mmol) in dry DMF (8 mL). Stirred at room temperature for 16 h. Concentrated, added 0.5 N NaOH (15 mL), extracted with CH2CI2, dried combined organic extracts (MgS0 ), filtered, and concentrated. Purified by silica gel chromatography (eluant: 3% MeOH-CH2CI2) to give 0.25 g (70%) of the product 363 as a beige oil. MS m/e: 534 (M+H).
The following intermediates were prepared according to the above procedure:
Figure imgf000389_0002
Figure imgf000390_0001
Part I:
To compound 363 (0.24 g, 0.450 mmol) dissolved in MeOH (10 mL) was added 0.5 M NaOMe in MeOH (0.09 mL, 0.045 mmol). Stirred at room temperature for 60 mins. Added 4 N HCI in dioxane (0.11 mL, 0.450 mmol) and concentrated to give 0.20 g (100%) of the product 364 as a brown foam. MS m/e: 450 (M+H).
The following compounds were prepared according to Example 8:
Figure imgf000391_0001
Figure imgf000392_0001
Example 9: Thiazole-benzyl inhibitors
Example 9A: Scheme 1
Figure imgf000393_0001
Part F
Part H
Figure imgf000393_0002
Part A:
To glycinamide HCI 385 (60.0 g, 0.543 mol) suspended in MeOH (1000 mL) and cooled to 0 °C was added triethylamine (109.9 g, 151.4 mL, 1.09 mol) and tBOC anhydride (148.0 g, 0.678 mol) portionwise. Warmed to room temperature and stirred for 24 h. Concentrated, added 1 N NaOH (600 mL), extracted with CH2CI2, dried combined organic extracts (MgS04), filtered, and concentrated to give 53.0 g (56%) of the product 396 as a white solid. MS m/e: 175 (M+H).
For n=2: MS m/e: 189 (M+H)
Part B:
To compound 386 (21.63 g, 0.124 mol) dissolved in THF (400 mL) was added Lawesson reagent (30.13 g, 0.074 mol). Stirred at room temperature for 16 h then concentrated. Purified by silica gel chromatography (eluant: 3% MeOH- CH2CI ) then re-purified by silica gel chromatography (eluant: 2% MeOH- CH2CI2) to give 23.59 g (100%) of the product 387 as a light green solid. MS m/e: 135 (M+2-tBu).
For n=2: MS m/e: 149 (M+2-tBu)
Part C:
To compound 387 (6.00 g, 31.5 mmol) dissolved in CH2CI2 (150 mL) was added ethyl bromopyruvate (6.76 g, 4.4 mL, 34.7 mmol). Stirred at room temperature for 5 h then concentrated. Added 3 A sieves (6 g) and EtOH (150 mL) and refluxed for 16 h. Filtered and concentrated to give a dark foam. Dissolved foam in 1 :1 CH2CI :EtOH (100 mL) and added triethylamine (6.40 g, 8.8 mL, 63.1 mmol) and tBOC anhydride (7.60 g, 34.7 mmol). Stirred at room temperature for 5 h then concentrated. Added 0.25 N NaOH (100 mL), extracted with CH2CI2, dried combined organic extracts (MgS0 ), filtered, and concentrated. Purified by silica gel chromatography (eluant: 10% EtOAc- CH2CI2 to 30% EtOAc-CH2CI2) to give 6.00 g (67%) of the product 388 as a brown oil. MS m/e: 287 (M+H). For n=2: MS m/e: 301 (M+H)
Part D:
To compound 388 (4.70 g, 16.4 mmol) dissolved in Et20 (140 mL) was added lithium borohydride (1.43 g, 65.7 mmol) and MeOH (2.10 g, 2.7 mL, 65.7 mmol). Refluxed for 16 h, cooled to room temperature, and concentrated. Added water (100 mL), extracted with CH2CI2, dried combined organic extracts (MgS0 ), filtered, and concentrated. Purified by silica gel chromatography (eluant: 2% MeOH-CH2CI2 to 5% MeOH-CH2CI2) to give 3.70 g (92%) of the product 389 as a yellow solid. MS m/e: 245 (M+H). For n=2: MS m/e: 259 (M+H)
Part E:
To compound 389 (5.30 g, 21.7 mmol) dissolved in CH2CI2 (130 mL) and cooled to -25 °C was added triethylamine (4.40 g, 6.0 mL, 43.4 mmol) then mesyl chloride (3.00 g, 2.0 mL, 26.0 mmol) dropwise. Warmed to 0 °C slowly over 60 mins. Added water (100 mL), extracted with CH2CI2, dried combined organic extracts (MgS0 ), filtered, and concentrated to give 7.00 g (100%) of the product 390 as a yellow oil. MS m/e: 223 (M+2-tBOC). For n=2: MS m/e: 237 (M+2-tBOC)
Part F:
To compound 390 (0.60 g, 1.86 mmol) dissolved in dry DMF (25 mL) was added 4-hydroxypyridine (0.22 g, 2.30 mmol) and cesium carbonate (1.20 g, 3.72 mmol). Stirred at room temperature for 16 h then concentrated. Added water (25 mL), extracted with CH2CI2, dried combined organic extracts (MgS0 ), filtered, and concentrated. Purified by silica gel chromatography (eluant: 10% MeOH with NH3-CH2CI2) to give 0.40 g (68%) of the product 391 A as a colorless oil. MS m/e: 322 (M+H).
The following intermediates were prepared according to the above procedure:
Figure imgf000395_0001
Part G:
Suspended copper cyanide (1.65 g, 18.4 mmol) in dry THF (70 mL) under a nitrogen atmosphere and cooled to -25 °C. Added phenyl magnesium bromide (3.0 M in Et20, 12.3 mL, 37.0 mmol) dropwise via syringe such that internal temperature < -20 °C. Stirred at -20 °C for 30 mins then at 0 °C for 30 mins. Warmed to 15 °C internal temperature then recooled to -25 °C. Added compound 390 (1.98 g, 6.14 mmol) dissolved in dry THF (20 mL) dropwise via syringe. Stirred at -25 °C internal temperature for 2 h then at 0 °C for 16 h. Concentrated, added 2 N NH4OH (100 mL), extracted with CH2CI2, dried combined organic extracts (MgS04), filtered, and concentrated. Purified by silica gel chromatography (eluant: 5% EtOAc-hexane to 20% EtOAc-hexane) to give 1.14 g (66%) of the product 392A as a yellow oil. MS m/e: 305 (M+H).
The following intermediates were prepared according to the above procedure:
Figure imgf000396_0001
Figure imgf000397_0001
Part H:
To compound 392A (0.30 g, 0.986 mmol) dissolved in 1 :1 CH2CI2:MeOH (10 mL) was added 4 N HCI in dioxane (2.4 mL, 9.86 mmol). Stirred at room temperature for 16 h then concentrated to give 0.23 g (100%) of the product 393A (hydrochloride salt) as a beige foam. MS m/e: 205 (M+H).
The following intermediates were prepared according to the above procedure:
Figure imgf000397_0002
Figure imgf000398_0001
Figure imgf000399_0001
Part I:
Combined compound 393A (100 mg, 0.416 mmol), compound 362 (181 mg, 0.499 mmol), HATU (316 mg, 0.832 mmol), and triethylamine (126 mg, 0.17 mL, 1.25 mmol) in dry DMF (6 mL). Stirred at room temperature for 16 h. Concentrated, added 0.5 N NaOH (15 mL), extracted with CH2CI2, dried combined organic extracts (MgS04), filtered, and concentrated. Purified by silica gel chromatography (eluant: 3% MeOH-CH2CI2) to give 120 mg (52%) of the product 394A as a colorless oil. MS m/e: 550 (M+H).
The following intermediates were prepared according to the above procedure:
Figure imgf000399_0002
Figure imgf000400_0001
Figure imgf000401_0001
66ε iεi6I0/£00ZSfl/13d oεi /sooz OΛV
Figure imgf000402_0001
Figure imgf000403_0001
Part J:
To compound 394A (0.12 g, 0.218 mmol) dissolved in MeOH (6 mL) was added 0.5 M NaOMe in MeOH (0.044 mL, 0.0218 mmol). Stirred at room temperature for 60 mins. Added 4 N HCI in dioxane (0.055 mL, 0.218 mmol) and concentrated to give 0.10 g (100%) of the product 395 as a light yellow foam. MS m/e: 466 (M+H).
The following compounds were prepared according to the above procedure:
Figure imgf000404_0001
Figure imgf000405_0001
Figure imgf000406_0001
Figure imgf000407_0001
Figure imgf000408_0001
Example 9B
Scheme 1
HC N Br Part A MsθV*N -D, Part B s ~Br >- s^~Br * NYV ^
449 450 451 HV 452
Figure imgf000408_0002
Reference for compound 449:
K. C. Nicolaou, N. P. King, M. R. V. Finlay, Y. He, F. Roschangar, D. Vourloumis, H. Vallberg, F. Sarabia, S. Ninkovic, D. Hepworth; Bioorg. Med. Chem. 1999, 7, 665-697.
Part A: To compound 449 (4.86 g, 25.0 mmol) dissolved in CH2CI2 (200 mL) and cooled to -30 °C was added triethylamine (5.07 g, 7.0 mL, 50.1 mmol) and then mesyl chloride (3.44 g, 2.3 mL, 30.1 mmol) dropwise via syringe. Warmed slowly to 0 °C over 60 mins. Added water (200 mL), extracted with CH2CI2, dried combined organic extracts (MgS04), filtered, and concentrated to give 6.80 g (100%) of the product 450 as an orange oil. MS m/e: 272 (M+H).
Part B:
To compound 450 (6.80 g, 25.0 mmol) dissolved in DMF (100 mL) was added sodium azide (3.25 g, 50.0 mmol) and heated at 80 °C for 2 h. Cooled to room temperature and concentrated. Added water (200 mL), extracted with CH2CI2, dried combined organic extracts (MgS0 ), filtered, and concentrated. Purified by silica gel chromatography (eluant: 5% EtOAc-hexane to 10% EtOAc-hexane) to give 4.18 g (76%) of the product 451 as an orange oil. MS m/e: 219 (M+H).
Part C:
To compound 451 (4.18 g, 19.1 mmol) dissolved in 10:1 THF:water by volume (150 mL) was added triphenylphosphine (20.0 g, 76.3 mmol) and refluxed for 2 h. Cooled to room temperature and concentrated. Purified by silica gel chromatography (eluant: 3% MeOH with NH3-CH2CI2) to give 6.18 g of the product 452 (with triphenylphosphine oxide) as a yellow solid. 100% yield of product 452 would be 3.68 g. MS m/e: 194 (M+H).
Part D:
To compound 452 (3.68 g, 19.1 mmol) dissolved in CH2CI2 (100 mL) was added tBOC anhydride (5.21 g, 23.9 mmol). Stirred at room temperature for 2 h then concentrated. Purified by silica gel chromatography (eluant: 3% MeOH-CH2CI2) to give 3.97 g (71 %) of the product 453 as a yellow oil. MS m/e: 293 (M+H).
Part E: To compound 453 (1.07 g, 3.65 mmol) dissolved in dry THF (10 mL) under a nitrogen atmosphere was added 2-chlorobenzylzinc chloride (0.5 M in THF, 14.6 mL, 7.30 mmol) and bis(tri-t-butylphosphine)palladium (0.14 g, 0.274 mmol). Heated at 60 °C for 2 h. Cooled to room temperature and concentrated. Added 0.2 N HCI (30 mL), extracted with CH2CI2, dried combined organic extracts (MgS04), filtered, and concentrated. Purified by silica gel chromatography (eluant: 5% EtOAc-hexane to 15% EtOAc-hexane) to give 0.61 g (49%) of the product 454 as a yellow oil. MS m/e: 339 (M+H).
Scheme 2
Figure imgf000410_0001
CBZNHAA CHO J=L CBZNHAVV ϋ CBZV"
459 ζj 458 ζ 457 ' -\ OMs
Part K
Figure imgf000410_0002
Part F:
To compound 389 (2.00 g, 8.18 mmol) dissolved in MeOH (40 mL) was added 4 N HCI in dioxane (20.5 mL, 81.8 mmol). Stirred at room temperature for 3 h. Concentrated to give 1.48 g (100%) of the product 455 as a white solid. MS m/e: 145 (M+H).
Part G:
To compound 455 (1.48 g, 8.18 mmol) suspended in CH2CI (50 mL) was added triethylamine (2.48 g, 3.4 mL, 24.5 mmol) and cooled to 0 °C. Added CBZCI (1.54 g, 1.3 mL, 9.00 mmol) dissolved in CH2CI2 (10 mL) dropwise via addition funnel. Stirred at 0 °C for 30 mins then at room temperature for 2 h. Added 0.2 N NaOH (100 mL), extracted with CH2CI2, dried combined organic extracts (MgS0 ), filtered, and concentrated. Purified by silica gel chromatography (eluant: 5% MeOH-CH2CI2 to 10% MeOH-CH2CI2) to give 1.41 g (62%) of the product 456 as a white solid. MS m/e: 279 (M+H).
Part H:
To compound 456 (1.40 g, 5.03 mmol) dissolved in CH2CI2 (40 mL) and cooled to -30 °C was added triethylamine (1.02 g, 1.4 mL, 10.1 mmol) and then mesyl chloride (0.69 g, 0.47 mL, 6.04 mmol) dropwise via syringe. Warmed slowly to 0 °C over 60 mins. Added water (50 mL), extracted with CH2CI2, dried combined organic extracts (MgS0 ), filtered, and concentrated to give 1.79 g (100%) of the product 457 as a yellow oil. MS m/e: 357 (M+H).
Part i:
To 2-(1 ,3-dioxan-2-yl)phenylmagnesium bromide (0.25 M in THF, 100 mL, 25.0 mmol) under a nitrogen atmosphere and cooled to -25 °C internal temperature was added copper cyanide (1.12 g, 12.5 mmol). Stirred at -25 °C for 1 h then at 0 °C for 1 h. Warmed to 15 °C internal temperature then recooled to -25 °C. Added compound 457 (1.78 g, 4.99 mmol) dissolved in dry THF (15 mL) dropwise via syringe. Stirred at -25 °C internal temperature for 1 h then at 0 °C for 16 h. Concentrated, added 2 N NH4OH (100 mL) and CH2CI2 (100 mL), and filtered through celite. Separated layers of filtrate, extracted with CH2CI2, dried combined organic extracts (MgS0 ), filtered, and concentrated. Purified by silica gel chromatography (eluant: 10% EtOAc- hexane to 40% EtOAc-hexane) to give 1.35 g (64%) of the product 458 as a white solid. MS m/e: 425 (M+H).
Part J:
To compound 458 (1.34 g, 3.16 mmol) dissolved in CH CI2 (10 mL) was added water (2 mL) and TFA (8 mL). Stirred at room temperature for 5 h then concentrated. Added 1 N NaOH (50 mL), extracted with CH2CI2, dried combined organic extracts (MgS04), filtered, and concentrated. Purified by silica gel chromatography (eluant: 20% EtOAc-CH2CI2 to 30% EtOAc-CH2CI2) to give 1.05 g (91%) of the product 459 as a yellow oil. MS m/e: 367 (M+H). Part K:
To compound 459 (0.74 g, 2.02 mmol) dissolved in CH2CI2 (20 mL) was added dimethylamine (2 M in THF, 2.0 mL, 4.04 mmol), 3A sieves (0.60 g), glacial acetic acid (0.12 g, 0.12 mL, 2.02 mmol), then sodium triacetoxyborohydride (0.64 g, 3.03 mmol). Stirred at room temperature for 16 h. Added 0.5 N NaOH (25 mL), extracted with CH2CI2, dried combined organic extracts (MgS04), filtered, and concentrated. Purified by silica gel chromatography (eluant: 5% MeOH with NH3-CH2CI2 to 15% MeOH with NH3- CH2CI2) to give 0.65 g (81 %) of the product 460 as a yellow oil. MS m/e: 396 (M+H).
Part L:
To compound 460 (0.64 g, 1.62 mmol) dissolved in MeOH (5 mL) was added THF (2 mL) and 6.25 N NaOH (5 mL). Refluxed for 3 h. Cooled to room temperature and concentrated. Purified by silica gel chromatography (eluant: 10% MeOH with NH3-CH2Cl2 to 15% MeOH with NH3-CH2CI2) to give 0.31 g (74%) of the product 461 as an orange oil. MS m/e: 262 (M+H).
Select compounds prepared using the procedures from Example 9B are exemplified in the preceding table.
Example 10: 2-Heteroaryl pyrrolidines & derivatives
Example 10A:
Figure imgf000413_0001
Part A:
To D-1-N-Boc-prolinamide (462) (2.5 g, 11.7 mmol) in THF (15 mL) was added Lawesson's reagent (2.36 g, 5.8 mmol) portionwise at room temperature. The reaction mixture was stirred for 3.5 hours under argon atmosphere. The solvents were removed in vacuo. Purification by column chromatography (Si02, 5% MeOH/DCM) afforded 463 as a light yellow solid (2.5 g, 93%).
Part B:
A mixture of 463 (500 mg, 2.15 mmol) and potassium hydrogencarbonate (1.74 g, 17.35 mmol) in DME (12 mL) was stirred for 10 minutes. Ethyl bromopyruvate (0.81 mL, 6.45 mmol) was added dropwise via a syringe to the reaction mixture. The reaction mixture was stirred for 30 minutes. The reaction mixture was cooled to 0 °C and a mixture of trifluoroacetic anhydride (1.21 mL, 8.6 mmol) and 2,6-lutidine (2.12 mL, 18.3 mmol) was added dropwise via syringe over 10 minutes. The reaction mixture was stirred for 30 minutes at 0 °C. The solvents were removed in vacuo. The residue was dissolved in chloroform, washed with 1.0 N HCI, bicarbonate solution and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02) 30% EtOAc/hexane) afforded 464 as a light yellow solid (520 mg, 74%). HPLC-MS tR = 1.88 min (UV254 nm); Mass calculated for formula C15H22N2θ4S 326.1 , observed LCMS m/z 327.1 (M+H).
Part C:
To 464 (486 mg, 1.49 mmol) in dioxane (1 mL) was added 4 N HCI in dioxane (1 mL). The reaction mixture was stirred for 1 hour at room temperature and concentrated. HPLC-MS tR = 0.60 min (UV25 nm); Mass calculated for formula C10H14N2O2S 226.1 , observed LCMS m/z 227.1 (M+H).
Part D:
To 48 (216 mg, 1.0 mmol) in THF (2 mL) was added material from 229 (238 mg, 1 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was concentrated and freeze-dried to afford a white powder (410 mg, 90%). HPLC-MS tR = 1.74 min (UV254 nm);
Mass calculated for formula C2oH2oClN0 S 453.0, observed LCMS m/z 454.0
(M+H).
Part E:
To 465 (26 mg, 0.1 mmol) in DMF (0.5 mL) was added DIEA (35 μL, 0.2 mmol), 466 (54 mg, 0.12 mmol) in DMF (1 mL) and then HATU (57 mg, 0.15 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was diluted with ethyl acetate (20 mL) and water (20 mL) and the layers were separated. The organic layer was washed with 0.1 N NaOH, 0.1 N HCI and brine, dried over sodium sulfate and concentrated. Compound 467 was used without further purification. HPLC-MS tR = 2.06 min (UV254 nm); Mass calculated for formula C3oH32CIN308S2 661.1 , observed LCMS m/z 662.0 (M+H).
Part F:
To 467 in methanol (1 mL) was added 7.0 M ammonia in methanol (1 mL). The reaction mixture was stirred for 1 hour at room temperature and concentrated. Purification by reverse phase prep-LC afforded 468 as a white powder (2 mg). HPLC-MS tR = 5.04 min (UV254 nm, 10 min); Mass calculated for formula C26H28CIN306S2 577.1 , observed LCMS m/z 578.0 (M+H). Example 10B:
Figure imgf000415_0001
Part A:
A solution of LDA was formed by the addition of 1.6 M π-butyl lithium (34 mL, 54.5 mmol) to diisopropylamine (8.47 mL, 60 mmoL) in THF (20 mL) at -78 °C. The reaction mixture was stirred at -78 °C for 20 minutes and warmed to 0 °C gradually. The LDA solution was added dropwise to a mixture of N-Boc- D-proline methyl ester (469) (2.5 g, 10.9 mmol) and chloroiodomethane (3.17 mL, 43.6 mmol) in THF (20 mL) at - 78 °C via a cannula over 30 minutes. The reaction mixture was stirred at -78 °C for 30 minutes. A solution of acetic acid (15 mL) in THF (15 mL) was added slowly over 20 minutes at -78 °C. The reaction mixture was stirred for 20 minutes and then warmed to room temperature. The reaction mixture was diluted with ethyl acetate and washed with water, sodium bicarbonate solution and brine. The organic layer was dried over sodium sulfate and concentrated. Purification by column chromatography (Siθ2, 20% EtOAc/hexane) afforded 470 as a light brown solid (2.0 g, 74%). HPLC-MS tR = 1.80 min (UV254 nm); Mass calculated for formula CnH18CIN03 247.1 , observed LCMS m/z 248.1 (M+H).
Part B:
A mixture of 470 (250 mg, 1.0 mmol) and thiourea (152 mg, 2 mmol) were stirred for 72 hours. The reaction mixture was diluted with ethyl acetate and washed with sodium bicarbonate solution and brine. The organic layer was dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 80% EtOAc/hexane) afforded 471 as a white solid (201 mg, 75%). HPLC-MS tR = 0.67 min (UV2 4 nm); Mass calculated for formula C12H19N302S 269.1 , observed LCMS m/z 270.1 (M+H).
Part C:
To 471 (201 mg, 0.75 mmol) in dioxane (1 mL) was added 4 N HCI in dioxane
(1 mL). The reaction mixture was stirred for 1 hour at room temperature and concentrated.
Part D:
A mixture of 466 (45 mg, 0.1 mmol), 472 (29 mg, 0.12 mmol), DIEA (50 μL, 0.28 mmol) and HATU (57 mg, 0.15 mmol) in DMF (2 mL) was stirred overnight at room temperature. The reaction mixture was diluted with ethyl acetate (20 mL) and water (20 mL) and the layers were separated. The organic layer was washed with 0.1 N NaOH, 0.1 N HCI and brine, dried over sodium sulfate and concentrated. The residue was dissolved in methanol (1 mL) and 7.0 M ammonia in methanol (1 mL) was added. The reaction mixture was stirred for 1 hour at room temperature and concentrated. Purification by reverse phase prep-LC afforded 473 as a white powder. HPLC-MS tR = 1.42 min (UV254 nm); Mass calculated for formula C23H25CIN40 S2 520.1 , observed LCMS m/z 521.1 (M+H).
Example 10C:
Figure imgf000416_0001
Part A:
To 1-Benzyl-pyrroiidine-2-carboxylic acid (474) (Belokon, Y. N. at al; Tetrahedron Asymmetry 1998, 9, 4249-4252) in CH2CI2 (20 mL) was added 2,2-dimethoxy-ethylamine (614 mg, 5.84 mmol), EDCI (1.12 g, 5.84 mmol), HOBT (658 mg, 4.87 mmol), and NMM (1.08 mL, 9.8 mmol). The reaction mixture was stirred overnight at 25 °C. Saturated aqueous NaHC03 solution (50 mL) was added. The aqueous layer was extracted with CH2CI2 (50 mL x3). The combined organic layers was washed with brine (50 mL), dried over Na2S04, and concentrated by rotary evaporator to give 1-benzyl-pyrrolidine-2- carboxylic acid (2,2-dimethoxy-ethyl)-amide as yellow oil. To 1-benzyl- pyrrolidine-2-carboxylic acid (2,2-dimethoxy-ethyl)-amide was added acetic acid (10 mL) and ammonium acetate (11 g). The reaction mixture was heated to 140 °C overnight. After it was cooled down, it was poured into 100 mL ice- water with stirring. Solid NaHC03 was added in small portion with stirring to adjust the pH of the solution to 8-9. The aqueous solution was extracted with EtOAc (100 mL x2). The combined organic layers was washed with brine, dried over Na2S04, and concentrated with rotary evaporator. Compound 475 (110 mg) was isolated by Si02 chromatography (CH2CI2/MeOH/NH3: 40:1 :0.1 to 20: 1 :0.1 ). MS: m/z 228.3 [M+H]+.
Part B: 5-Pyrrolidine-2-yl-1H-imidazole:
To 475 (110 mg) in 10 mL EtOH was added Pd/C (10%, 50 mg). The reaction mixture was hydrogenated under hydrogen (50 psi) for 24 hours. The solid was filtrated and the solution was evaporated by rotary evaporator to give 5- Pyrrolidine-2-yl-1H-imidazole (476) (67 mg). MS m/z 138.2 [M+H]+.1H-NMR (300 MHz, CDCI3): δ ppm: 6.95 (s, 2H), 4.35 (t, 1 H, J = 7.4 Hz), 3.00 (m, 2H), 2.22-2.03 (m, 2H), 1.85 (m, 3H).
The following compounds were prepared by procedures described in Example 11. The 2-(pyridyl)-pyrrolidines are commericial amines (entries 495 - 497). The amino-thiazole of 471 was further functionalized via acylation chemistry to afford 500 - 501.
Figure imgf000418_0001
Figure imgf000419_0001
Figure imgf000420_0001
Example 11 : 2-Phenyl pyrrolidines & derivatives
Example 11 A:
Figure imgf000421_0001
Part A:
To 1 (925 mg, 4.53 mmol) in DMF (5 mL) was added 2-(3-bromophenyl)- pyrrolidine (503) (prepared by the method of Sorgi, K.L.; Maryanoff, C. A.; McComsey, D. F.; Graden, D. W.; Maryanoff, B. E.; J. Am. Chem. Soc. 1990, 112, 3567) (1.12 g, 4.95 mmol), DIEA (1.75 mL, 10.0 mmol) and HATU (1.88 g, 4.95 mmol). The reaction mixture was stirred overnight at room temperature. The DMF was removed in vacuo and the residue was partitioned between ethyl acetate (20 mL) and water (20 mL). The layers were separated and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with sodium bicarbonate solution, 1.0 N HCI and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 10 - 20% EtOAc/DCM) afforded 504 (1.22 g, 65%). HPLC-MS tR = 2.00 min (UV254 nm); mass calculated for formula C18H22BrN05411.1 , observed LCMS m/z 412.2 (M+H).
Part B:
To piperazine-1 -carboxylic acid tert-butyl ester (279 mg, 1.5 mmol), potassium phosphate (530 mg, 2.5 mmol), Pd2(dba)3 (23 mg, 0.025 mmol) and 2- (dicyclohexylphosphino)biphenyl (35 mg, 0.1 mmol) under argon atmosphere was added the material from Part A (414 mg, 1.0 mmol). The reaction mixture was evacuated and flushed with argon. The mixture was heated overnight at 90 °C. The reaction mixture was diluted with ethyl acetate, filtered through celite and concentrated to give a mixture of methyl ester 505A and acid 505B as an orange film (679 mg). The material was used without further purification. 505A: HPLC-MS tR = 2.08 & 2.14 min (ester, UV254 nm); mass calculated for formula C27H39N307 517.3, observed LCMS m/z 518.0 (M+H). 505B: HPLC-MS tR = 1.83 & 1.91 min (acid, UV254 nm); mass calculated for formula C26H37N307 503.3, observed LCMS m/z 504.1 (M+H)
Part C:
To the mixture of 505A and 505B (~1 mmol) in THF (4 mL) and water (1 mL) was added 1.0 M lithium hydroxide (1.2 mL, 1.2 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was diluted with water (10 mL) and the THF was removed in vacuo. The aqueous layer was washed with diethyl ether (3 x 10 mL), made acidic with 1.0 N HCI and extracted with ethyl acetate (3 x 10 mL). The combined ethyl acetate layers were dried over sodium sulfate and concentrated. Purification by reverse phase prep-LC afforded the desired isomer 506 (107 mg, 96% purity). HPLC-MS tR = 1.83 min (UV254 nm); mass calculated for formula C26H37N307 503.3, observed LCMS m/z 504.1 (M+H).
Part D:
To 506 (107 mg, 0.21 mmol) in DMF (5 mL) was added 229 (55 mg, 0.23 mmol), DIEA (80 μL, 0.46 mmol) and HATU (87 mg, 0.23 mmol). The reaction mixture was stirred overnight at room temperature. The DMF was removed in vacuo and the residue was partitioned between ethyl acetate and water. The layers were separated. The organic layer was washed with 0.1 N NaOH, 0.1 N HCI, and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 50% EtOAc/hexane) afforded 507 (98 mg, 64%). HPLC-MS tR = 2.49 min (UV254 nm); mass calculated for formula C38H47CIN406S 722.2, observed LCMS m/z 723.1 (M+H).
Part E:
To 507 (98 mg, 0.14 mmol) was added 80:20 TFA:water (4 mL) and the mixture was stirred for 4 hours at room temperature. The reaction was quenched with 1 :1 acetonitrile:water (10 mL) and concentrated. The residue was dissolved in ethyl acetate and washed with sodium bicarbonate solution. To the aqueous layer was added sodium chloride and it was extracted with ethyl acetate. The combined organic layer was dried over sodium sulfate and concentrated. The residue was dissolve in acetonitrile (2 mL) and 1.0 N HCI (0.3 mL) and concentrated. The material was lyophilized to afford 508 as the HCI salt as a white powder (75 mg, 86%). HPLC-MS tR = 1.27 min (UV254 nm); mass calculated for formula C30H35CIN O4S 582.2, observed LCMS m/z 583.2 (M+H).
Example 11 B
Figure imgf000423_0001
Part A
To a solution of compound 503 (5.0 g, 22.1 mmol) in dry CH2CI2 (80 mL) was added di-tert-butyl dicarbonate (5.55 g, 25.4 mmol). The solution was stirred at room temperature for 2 hours. The solvent was removed by rotary evaporator. The product was isolated by silica gel chromatography (Hexane/EtOAc 5:1 to 3:1 ) to give compound 509 (5.7 g, 79%).
Part B
To a two neck flask was charged with compound 509 (1.0 g, 3.06 mmol), MeOH (8 mL), triethylamine (6 mL), DMF (6 mL), and Pd(PPh3)2CI2. A condenser with a three-way-valve on the top was attached to the flask. A balloon and a carbon monoxide tank were attached to the three-way-valve. The balloon was filled with CO and flashed the system twice. Then the balloon was filled with CO and was connected to the flask system. The flask was heated in a 80 °C oil bath for 36 hours. After cooling down to room temperature, water (50 mL) and EtOAc (100 mL) were added. The organic phase was separated, washed with water (50 mL) twice and brine, dried over Na24, concentrated with rotary evaporator, the product was isolated with silica gel chromatography (Hexane/EtOAc 10:1 to 5:1) to give compound 510 (610 mg, 65%).
Part C
Compound 510 (384 mg, 1.26 mmol) was dissolved in dioxane/water (3:1 , 4 mL) and LiOH (100 mg. 2.38 mmol) was added. The solution was stirred at room temperature for four hours. Saturated NH CI solution (20 mL) was added. The aqueous phase was extracted with EtOAc (25 mL) twice. The organic phases were combined, washed with brine, dried over Na2S0 , concentrated by rotary evaporator, and dried under vacuum to give compound 511 (368 mg, 100%).
Part E
Compound 511 (76 mg, 0.26 mmol) was dissolved in dry CH2Cl2 (1.5 mL). N- methyl piperazine (0.035 mL, 0.31 mmol), EDCI (75 mg, 0.39 mmol), HOBT (43 mg, 0.31 mmol), and NMM (0.086 mL, 0.78 mmol) were added. The solution was stirred at room temperature for 16 hours. Saturated NaHC03 solution (5 mL) and CH CI2 (5 mL) was added and the layers were separated. The aqueous phase was extracted with CH2CI2 (5 mL) twice. The organic phases were combined, dried over Na2S0 , concentrated by rotary evaporator. The product was isolated by silica gel chromatography to give compound 512 (90 mg, 92%).
Part F
Compound 512 (90 mg, 0.24 mmol) was dissolved in MeOH (1 mL) and HCI (4M in dioxane, 0.25 mL, 1 mmol) was added. The solution was stirred at room temperature for overnight. The solvent was removed by rotary evaporator to give compound 513 (83 mg, 100%).
Example 11C:
Figure imgf000425_0001
Part G:
Compound 510 (3.68 g, 12.0 mmol) was dissolved in dry THF (60 mL), and lithium borohydride (0.78 g, 36.2 mmol) was added. The solution was heated at reflux for 16 h then cooled to room temperature. MeOH (4 mL) was added, and the solvent was removed by rotary evaporator. Water (75 mL) was added, and the aqueous solution was extracted with CH2Cl2 (75 mL) three times. The combined organic extracts were dried (MgS0 ), filtered, and concentrated. The product was purified by silica gel chromatography (eluant: 5% MeOH-CH2CI2 to 10% MeOH-CH2CI2) to give 3.28 g (98%) of compound
514. MS (m/e for M+1 ): 278.
Part H:
Oxalyl chloride (1.87 g, 1.3 mL, 14.7 mmol) was dissolved in dry CH2CI2 (35 mL) and cooled to -78° C under a nitrogen atmosphere. DMSO (2.30 g, 2.1 mL, 29.5 mmol) dissolved in dry CH2CI2 (5 mL) was added dropwise via addition funnel. The solution was stirred at -78° C for 15 mins then compound 514 (3.27 g, 11.8 mmol) dissolved in CH2CI2 (15 mL) was added. The reaction mixture was stirred at -78° C for 60 mins then triethylamine (3.58 g, 4.9 mL, 35.4 mmol) was added. The reaction mixture was stirred at -78° C for 15 mins then warmed to 0° C. Water (75 mL) was added, and the layers separated. The aqueous phase was extracted with CH2CI2 (75 mL) two times. The combined organic extracts were dried (MgSO4), filtered, and concentrated. The product was purified by silica gel chromatography (eluant: 5% EtOAc-CH2CI2 to 10% EtOAc-CH2CI2) to give 2.94 g (90%) of the product
515. MS (m/e for M+1): 276.
Part i: Compound 515 (0.50 g, 1.82 mmol) was dissolved in CH2CI2 (10 mL), and 3A sieves (0.50 g), dimethylamine in THF (2 M, 1.8 mL, 3.64 mmol), glacial acetic acid (0.109 g, 1.82 mmol), and sodium triacetoxyborohydride (0.579 g, 2.73 mmol) were added. The reaction mixture was stirred at room temperature for 24 h. 1 N NaOH (25 mL) was added, and the aqueous solution was extracted with CH2CI2 (25 mL) four times. The combined organic extracts were dried (MgS04), filtered, and concentrated. The product was purified by silica gel chromatography (eluant: 5% MeOH-CH2CI2 to 15% MeOH-CH2CI2) to give 0.396 g (72%) of the product 516. MS (m/e for M+1 ): 305.
The following intermediates were also prepared.
Figure imgf000426_0001
Part J:
The following compounds were prepared using procedure described in
Example 11 B Part F.
Figure imgf000427_0001
The following compounds were prepared using the procedures described in Example 11.
Figure imgf000427_0002
Figure imgf000428_0001
Figure imgf000429_0002
Example 12: Suzuki Aryl-Aryl couplings Example 12A:
Scheme 1 Biaryl Preparation (preferred method)
Figure imgf000429_0001
Compound 543 was prepared by procedures described in Example 1.
Part A:
To a solution of 543 (1.01 g, 3.17 mmol) and 4-bromobenzyl amine (0.71 g, 3.83 mmol) in CH2CI2 (10 mL) cooled to 0 °C was added DIEA (1.10 mL, 6.31 mmol) followed by PyBrOP (1.10 g, 3.43 mmol). The reaction was warmed to room temperature and stirred for 16 hours. The liquid was concentrated, and the thick oil was taken up in EtOAc. The organic layer was washed with 0.5 N KHS04 (1x), sat. NaHC03 (1x), dried (Na2S04), filtered, and concentrated. The residue was purified by silica gel chromatography (eluting 0% to 100% EtOAc/hexanes) to furnish 544 (1.27 g, 2.61 mmol, 82% yield) as a tan oil. MS m/e: 487.1 (M+H).
Part B:
A solution of 544 (0.105 g, 0.215 mmol), 2-cyanophenyl boronic acid (0.032 g, 0.215 mmol), and Pd(dppf)CI2 (0.016 g, 0.021 mmol) in CH3CN (1 mL) and 1 N l<2C03 (1 mL) was heated in a SmithCreator microwave (2-5 mL vessel, 150 °C for 10 minutes). The mixture was concentrated and the residue was purified by silica gel chromatography (eluting 0% to 100 % EtOAc/hexanes) to furnish 545 (0.062 g, 0.12 mmol, 56% yield) as a tan oil. MS m/e: 510.1 (M+H).
Part C:
A solution of 545 (0.049 g, 0.096 mmol) in a 70% TFA/20% CH2CI2/10% H20 mixture was stirred at room temperature for 2 hours. The mixture was concentrated, and the residue was purified by reverse phase HPLC (eluting 5:95 to 95:5 CH3CN/H20 (0.1 % HC02H)) to provide 546 (0.023 g, 0.049 mmol, 51 % yield) as a white solid. MS m/e: 470.1 (M+H).
Example 12B:
Figure imgf000430_0001
Part A: A solution of ethyl 2-bromobenzoate (548) (0.25 g, 1.1 mmol), (4- aminomethylphenyl)boronic acid (547) (0.20 g, 1.1 mmol), and Pd(dppf)CI2 (0.040 g, 0.055 mmol) in CH3CN (1 mL) and 1 N K2C03 (1 mL) was heated in a SmithCreator microwave (2-5 mL vessel, 100 °C for 5 minutes). The reaction was diluted with H20 and EtOAc. The organic layer was removed, and the aqueous phase was extracted with EtOAc (3x). The combined organics were dried (Na2S04), filtered, and concentrated. The residue was purified by silica gel chromatography (eluting 0% to 20% CH2CI2/MeOH) to furnish 549 (0.14 g, 0.55 mmol, 50 % yield) as a brown oil. MS m/e: 256.1 (M+H).
Part B:
To a solution of 549 (0.21 g, 0.82 mmol) and 543 (0.19 g, 0.59 mmol) in CH2CI2 (5 mL) cooled to 0 °C was added DIEA followed by PyBrOP. The reaction was stirred for 20 hours gradually warming to room temperature. The mixture was concentrated, and the brown oil was taken up in EtOAc. The organic phase was washed with 0.5 N KHS04 (1x), sat. NaHC03 (1x), dried (Na2S0 ), filtered, and concentrated. The residue was purified by silica gel chromatography (eluting 0% to 100% EtOAc/hexanes) to furnish 550 (0.29 g, 0.51 mmol, 87% yield) as a brown oil. MS m/e: 557.1 (M+H).
Part C:
To a solution of 550 (0.26 g, 0.47 mmol) in a THF (2 mL)/MeOH (2 mL)/H20 (1 mL) mixture was added LiOH*H20 (0.052 g, 1.24 mmol) in one solid portion. The reaction was stirred overnight and then acidified to pH ~ 3 with 1N HCI. After dilution with EtOAc, the organic layer was removed, and the aqueous phase was extracted with EtOAc (3x). The combined organics were dried (Na2S04), filtered, and concentrated. The residue was purified by reverse phase HPLC (eluting 10:90 to 100:0 CH3CN/H20 (0.1% HC02H) to furnish 551 (0.22 g, 0.38 mmol, 88 % yield). MS m/e: 529.1 (M+H).
Part D: To a solution of 551 (55 mg, 0.104 mmol) in CH2CI2 (1 mL) was added polystyrene-bound HOBt (109 mg, 0.095 mmol), DIC (0.067 mL, 0.427 mmol), and DMAP (7 mg, 0.057 mmol) and was shaken overnight. The mixture was filtered, then washed with DMF (3 x 3 mL), CH2CI2 (3 x 3 mL), DMF (3 x 3 mL), and THF (3 x 3 mL). The resin was dried under vacuum overnight. To the dried resin-bound acid (82 mg, 0.034 mmol) was added 3- chlorobenzylamine (8.33 μL, 0.068 mmol) in CH2CI2 (1 mL) and the mixture was shaken overnight. To the mixture was added polystyrene-bound isocyanate resin (70 mg, 0.102 mmol) and shaken for 5 hours. Filter the desired product and wash the filtrate with CH2CI2 (3 x 3 mL) and THF (3 x 3 mL). Concentrate the organic portions in vacuo to afford 552 (14.6 mg, 0.022 mmol, 66 % yield). MS m/e: 652.2 (M+H).
Part E:
To 552 (14.6mg, 0.022 mmol) was added a 2 mL of a mixture of
TFA/CH2CI2/H2θ (7/2/1 ) and stirred at room temperature for 2 h. The mixture was concentrated in vacuo. The mixture was purified via reverse-phase
HPLC to furnish 553 (3.7 mg, 0.006 mmol, 27 % yield). MS m/e: 612.1
(M+H).
Figure imgf000432_0001
Compound 554 was prepared by the procedure desribed in Example 4 Part C. HPLC-MS tR = 1.39 min (UV254 nm); mass calculated for formula Cι Hι6CIN05 397.1 , observed LCMS m/z 398.1 (M+H). Part A:
To 554 (355 mg, 0.89 mmol) in DMF (5 mL) was added 2-fluoro-4- bromobenzylamine hydrochloride (257 mg, 1.07 mmol), DIEA (530 uL, 3.03 mmol) and HATU (407 mg, 1.07 mmol). The reaction was stirred overnight at room temperature. The reaction mixture was poured into water and extracted with EtOAc. The combined organic layers were washed with 0.1 N NaOH, 0.1 N HCI, and brine; dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 80% ethyl acetate/hexanes) afforded 555 as a foam (320 mg, 62%). HPLC-MS tR = 2.03 min (UV254 nm); mass calculated for formula C25H25BrCIFN2θ6 582.1 , observed LCMS m/z 583.0 (M+H).
Part B:
To 555 (320 mg, 0.55 mmol) in MeOH (5 mL) was added anhydrous hydrazine (28 μL, 0.88 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was concentrated in vacuo and lypholized to afford 556 as a white powder (275 mg, 100%). HPLC-MS tR = 1.81 min (UV254 nm); mass calculated for formula C2iH2iBrCIFN204 498.0, observed LCMS m/z 499.0 (M+H).
Part C:
Compound 556 (46 mg, 0.092 mmol) in dioxane (1 mL) was added to a solution of 2-methoxyphenyl boronic acid (22 mg, 0.14 mmol), potassium phosphate (42 mg, 0.2 mmol), and PdCI2(dppf) (4 mg, 0.005 mmol) under argon atmosphere. The reaction mixture was heated to 80 °C overnight. After cooling the mixture was filtered through celite and the pad was rinsed with ethyl acetate. The filtrate was concentrated. Purification by reverse phase prep-LC afforded 557 as a white solid (26 mg) after lypholization. HPLC-MS tR = 1.97 min (UV254 nm); mass calculated for formula C28H28CIFN2O5 526.2, observed LCMS m/z 527.0 (M+H).
Example 12D:
Figure imgf000434_0001
Part A:
A mixture of 4-bromo-benzylamine hydrochloride (558) (1.0 g, 4.5 mmol), di- terf-butyl dicarbonate (1.48 g, 6.8 mmol) and DIEA (2.4 mL, 13.8 mmol) in chloroform (40 mL) was stirred overnight at room temperature. The reaction mixture was washed with 1.0 N HCI, water and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 20% ethyl acetate/hexane) afforded a mixture of 559 (1.17 g, 91 %). 1H NMR (400 MHz, CDCI3) 57.44 (d, 2H, J = 8.8 Hz), 7.15 (d, 2H, J = 8.8 Hz), 4.85 (bs, 1 H), 4.27 (d, 2H, J = 5.7 Hz), 1.48 (s, 9H).
Part B:
A mixture of 559 (100 mg, 0.35 mmol), 560 (107 mg, 0.52 mmol) potassium phosphate (223 mg, 1.05 mmol) and PdCI2(dppf) (14 mg, 0.018 mmol) in dioxane (5 mL) was under argon atmosphere was heated to 80 °C overnight. The reaction mixture was cooled and filtered through celite. The celite pad was washed with ethyl acetate. The filtrate was washed with saturated sodium bicarbonate, water and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 20% ethyl acetate/hexane) afforded slightly impure 561 (146 mg). HPLC-MS tR = 2.35 min (UV254 nm); mass calculated for formula CιgH2oF3N03 367.1 , observed LCMS m/z 390.1 (M+Na).
Part C:
Compound 561 (129 mg, 0.35 mmol) in 3:1 DCM:TFA (4 mL) was stirred for 1 hour. The residue was dissolved in DCM (5 mL) and concentrated. The residue was dissolved in diethyl ether (20 mL) and treated with 1.0 M HCI in diethyl ether (2 mL). The resulting white solid was collected by filtration and washed with diethyl ether to afford 562 (90 mg, 85% 2 steps). 1H NMR (400 MHz, DMSO-d6) δ 8.3 (bs, 3H), 7.50 (m, 8H), 4.09 (s, 2H).
Part D:
Compound 563 was prepared using procedures similar to those described in Example 11 A. Compound 564 was prepared from 562 and 563 using procedures similar to those described in Example 1 Part A. HPLC-MS tR = 2.49 min (UV254 nm); mass calculated for formula C31H30CIF3N2O5 602.2, observed LCMS m/z 603.2 (M+H).
Part E:
Compound 565 was prepared using the procedures described in Example 1 Part B. HPLC-MS tR = 2.16 min (UV254 nm); mass calculated for formula C28H26CIF3N2θ5 562.2, observed LCMS m/z 563.0 (M+H).
Example 12E:
Figure imgf000435_0001
Part A:
To 2-bromo-5-fluoro-phenol (566) (2.28 g, 11.94 mmol) in DMF (15 mL) was added iodoethane (1.16 mL, 14.32 mmol) and cesium carbonate (4.28 g, 13.13 mmol). The reaction mixture was stirred for 72 hours. The mixture was filtered and the DMF was removed in vacuo. The residue was partitioned in ethyl acetate and water. The layers were separated and the organic layer was washed with water and brine, dried over sodium sulfate and concentrated to afford 567 (1.76 g, 67%) as a colorless oil. 1H NMR (400 MHz, CDCI3) δ 7.46 (dd, 1 H, J = 6.3, 8.8 Hz), 6.62 (dd, 1 H, J = 2.7, 10.5 Hz), 6.57 (m, 1 H, J = 2.7), 4.08 (q, 2H, J = 6.9 Hz), 1.50 (t, 3H, J = 7.0 Hz).
Part B:
Compound 569 (607 mg, 58%) was prepared from 567 and 568 (prepared by the procedures of Maku, S. et. al. (J. Comb. Chem. 2003, 5, 379)) using the procedures described in Example 12D Part B. HPLC-MS tR = 2.13 min (UV25 nrn); mass calculated for formula C175F4N02 341.1 , observed LCMS m/z
342.1 (M+H).
Part C:
To 569 (607 mg, 1.78 mmol) in methanol ( 6 mL) was added 10% potassium carbonate in 2:1 methanokwater (20 ml). To obtain a clear solution additional water (5 mL) was added. The reaction mixture was stirred overnight at room temperature. The methanol was removed in vacuo. The residue was portioned between water and ethyl acetate. The layers were separated and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated. The residue was dissolved in diethyl ether (20 mL) and treated with 1.0 M HCI in diethyl ether (5 mL). The resulting white solid was collected by filtration and washed with diethyl ether to afford 570 (377 mg, 75%). 1H NMR (400 MHz, DMSO-d6) δ 8.27 (bs, 3H), 7.49 (m, 4H), 7.29 (dd, 1 H, J = 6.9, 8.3 Hz), 7.00 (dd, 1 h J = 2.5, 11.4 Hz), 6.83 (dt, 1 H, J = 2.8, 8.6, 10.8Hz) 4.05 (m, 4H), 1.27 (t, 3H, J = 6.5 Hz).
Part D:
Compound 571 was prepared using procedures described in Example 12E Part D. Purification by column chromatography (Si02, 20% ethyl acetate/hexane) afforded 571 (104 mg, 95%). HPLC-MS tR = 2.40 min (UV254 nm); mass calculated for formula C32H34CIFN205 580.2, observed LCMS m/z
581.2 (M+H).
Part E: Compound 572 was prepared using the procedures described in Example 12D Part E. HPLC-MS tR = 2.06 min (UV254 nm); mass calculated for formula C29H3oCIFN205 540.2, observed LCMS m/z 541.2 (M+H).
Example 12F:
Figure imgf000437_0001
Part A:
To a mixture of 2-propyl phenol (573) (0.5 mL, 3.63 mmol) and DIEA (0.950 mL, 5.45 mmol) in DCM (20 mL) at 0 °C was added triflic anhydride (0.734 mL, 4.36 mmol) in DCM (10 mL) via an addition funnel. The reaction mixture was stirred for 45 minutes. The mixture was poured into water. The layers were separated and the organic layer was washed with saturated sodium bicarbonate solution and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 5% ethyl acetate/hexane) afforded 574 (916 mg, 100%). 1H NMR (400 MHz, CDCI3) δ 7.30 (m, 2H), 7.26 (m, 2H), 2.70 (m, 2H), 1.68 (m, 2H), 1.00 (t, 3H, J = 7.3 Hz).
Part B:
Compound 575 (288 mg, 89%) was prepared using the procedures described in Example 12D Part B and a reaction temperature of 100 °C. HPLC-MS tR = 2.30 min (UV254 nm); mass calculated for formula Ci8HιsF3NO 321.1 , observed LCMS m/z 322.2 (M+H).
Part C: Compound 576 (203 mg, 86%) was prepared using the procedure described in Example 12E Part C. 1H NMR (400 MHz, DMSO-d6) δ 8.27 (bs, 3H), 7.52 (d, 2H, J = 8.0 Hz), 7.32 (d, 2H, J = 8.1 Hz), 7.31 (m, 2H), 7.22 (m, 1 H), 7.10 (d, 1 H, J = 7.1 Hz), 4.08 (s, 2H,) 2.54 (m, 2H), 1.44 (m, 2H), 0.75 (t, 3H, J = 7.7 Hz).
Part D:
Compound 577 (62 mg, 92%) was prepared according to the procedure described in Example 12D Part D. HPLC-MS tR = 2.55 min (UV254 nm); mass calculated for formula C33H37CIN204 560.2, observed LCMS m/z 561.2 (M+H).
Part E:
Compound 578 (53 mg, 93%) was prepared according to the procedure described in Example 12D Part E. . HPLC-MS tR = 2.21 min (UV254 nm); mass calculated for formula C30H33CIN2O4 520.2, observed LCMS m/z 521.2 (M+H).
Example 12G:
Figure imgf000438_0001
Part A:
A mixture of 4-bromo-3-fluoro-toluene (579) (2.0 mL, 15.8 mmol), N- bromosuccinimide (3.38 g, 19.0 mmol) and benzoylperoxide (48 mg, 0.2 mmol) in carbon tetrachloride (50 mL) was heated to reflux under a nitrogen atmosphere for 16 hours. The reaction mixture was cooled and filtered. The filtrate was washed with water (2x), saturated sodium bicarbonate solution and brine, dried over sodium sulfate and concentrated to afford a mixture of 580 and dibromonated product (4.02 g). The material was used without further purification. 1H NMR (400 MHz, CDCI3) δ 7.52 (dd, 1 H, J = 6.6, 7.6 Hz), 7.17 (dd, 1 H, J = 2.0, 8.9 Hz), 7.11 (dd, 1 H, J = 2.0, 8.2 Hz), 4.42 (s, 2H).
Part B:
A mixture of 580 (4.02 g, 15.0 mmol), phthalimide (2.65 g, 18 mmol) and cesium carbonate (5.38 g, 16.5 mmol) in DMF (30 mL) was stirred for 72 hours. The reaction mixture was poured into water (100 mL) and extracted with ethyl acetate. The combined organic layers were washed with water (3X) and brine, dried over sodium sulfate and concentrated. Recrystallization from 30% ethyl acetate/hexanes afforded slightly impure 581 (3.28 g) as a yellow solid. 1H NMR (400 MHz, CDCI3) δ 7.86 (m, 2H), 7.74 (m, 2H), 7.49 (dd, 1 H, J = 7.3, 8.4 Hz), 7.20 (dd, 1 H, J = 1.9, 9.3 Hz), 7.06 (dd, 1 H, J = 2.0, 7.9 Hz), 4.80 (s, 2H).
Part C:
A mixture of 581 (1.00 g, 3.0 mmol) and hydrazine monohydrate (580 uL, 12.0 mmol) in ethanol (25 mL) was heated to reflux for 1 hour. The reaction mixture was diluted with ethyl acetate and filtered. The precipitate was washed with ethyl acetate. The filtrate was concentrated and the residue was dissolved in water and ethyl acetate. The layers were separated. The organic layer was washed with saturated sodium bicarbonate solution, water and brine, dried over sodium sulfate and concentrated to afford 582 (375 mg) as a yellow oil. 1H NMR (400 MHz, CDCI3) δ 7.48 (t, 1 H, J = 8.5 Hz), 7.12 (dd, 1 H, J = 2.2, 9.5 Hz), 6.99 (dd, 1 H, J = 1.6, 8.2 Hz), 3.86 (s, 2H).
Part D:
Compound 583 was prepared using procedures similar to those described in Example 12C Part A. HPLC-MS tR = 1.82 min (UV254 nm); mass calculated for formula C23H22BrFN2θ6 520.0, observed LCMS m/z 521.0 (M+H).
Part E: Compound 584 was prepared using the procedures described in Example 12C Part B. HPLC-MS tR = 1.81 min (UV254 nm); mass calculated for formula C19H18BrFN204436.0, observed LCMS m/z 437.0 (M+H).
Part F:
Compound 585 was prepared using procedures similar to those described in Example 12C Part C. HPLC-MS tR = 4.12 min (UV254 nm, 10 min); mass calculated for formula C26H22FN304459.2, observed LCMS m/z 460.1 (M+H).
The following table contains compounds prepared using the procedures described in Example 12 A - G.
Figure imgf000440_0001
Figure imgf000441_0001
Figure imgf000442_0001
Figure imgf000443_0001
Figure imgf000444_0001
Figure imgf000445_0001
Example 13
Figure imgf000445_0002
Compound 631 was prepared by the procedures described in Example 11 A. Part A.
To a solution of 631 (0.46 g) and 632 (1.2 eq) in 10 ml DMF at OC were added HATU (1.5 eq) and HOBt (1.5 eq). The reaction was stirred at r.t. overnight before the solvent was removed in vacuo and residue chromatographed using Ethyl Acetate in Hexane (0 - 100 %) to give 0.46 gram of desired product 633.
Part B.
To a solution of 0.46 g of 633 in 10 ml of DCM was added Dess-Martin Reagent (1.1 eq) and the reaction was stirred at r.t. for 30 min before it was treated with sat. aqueous solution of sodium bicarbonate and sodium thiosulfate 7:1 (w/w). The aqueous layer was extracted with DCM (2X) and the combined organic layers was dried over anhydrous sodium sulfate. After removal of solvent, the crude reaction product was dissolved in 2 ml pyridine. To the solution was added 1.5 eq of hydroxylamine hydrochloride and the solution was heated to reflux for 30 min before the solvent was removed in vacuo and residue chromatographed using Ethyl Acetate in Hexane (0 - 100 %) to give 0.4 gram of product 634.
Part C
To a flask containing 634 (0.42 g) in anhydrous DMF at 0 °C was added NBS (1 eq) and the solution was allowed to warm up to r.t. overnight before another equivalent of NBS was added and solution stirred for another overnight. The final reaction mixture was poured over ice and extracted with DCM. The organic phase was washed with water and brine, dried over sodium sulfate. After removal of the solvent, the residue was chromatographed using Ethyl Acetate in Hexane (0 - 100 %) to give 0.38 gram of desired product.
To a solution of 0.035 gram of above compound in 2 ml DCM was added vinylacetate (2 eq) and DIEA (3.5 eq) and the solution was stirred at r.t. overnight. After removal of solvent, the residue was chromatographed using Ethyl Acetate in Hexane (0 - 100 %) to give 20 mg product which was treated with 50% TFA in DCM for 2h. before it was purified using a RP HPLC system to give 10 mg of desired product 635. The following compounds were, generated using similar procedures.
Figure imgf000447_0001
Example 14:
Figure imgf000448_0001
Part A:
A Schlenck flask was flame dried under N2 flow, capped with a septum, and allowed to cool to rt. Phthalimide (640) (4.13 g, 28.1 mmol) was added followed by anhydrous THF (100 mL). After the phthalimide had dissolved, the flask was placed in an ice water bath and allowed to cool for 20 min. A 1 M solution of 3-fluorophenyl magnesium bromide in THF (25 mL) was added causing a precipitate to form. DMPU (5 mL) was added, causing the reaction mixture to become clear again. Additional 3-fluorophenyl magnesium bromide was added (35 mL) over 10 min. The reaction mixture was stirred at 0 °C for 3.5 h, then quenched with 1.0 M pH 6.5 sodium phosphate buffer. The resulting mixture was diluted with EtOAc and the layers were separated. The organic layer was washed with water and brine, then dried with MgS04. Evaporation of the solvent gave and off white solid (7.4 g). This material was triturated in CH2CI2/hexanes to give 4.0 g of pure 641. MS (El) m/z Obsd. M+H 244.0
Part B:
A Schlenck flask was flame dried under N2 flow, capped with a septum, and allowed to cool to rt. LiAIH4 (1.26 g, 33.2 mmol) and AICI3 (1.47 g, 11.0 mmol) were added to the flask, followed by anhydrous THF. The flask was placed in an ice-water bath immediately and allowed to cool with stirring. Compound 641 was added to the flask in portions. The reaction mixture was stirred for 3 h during which time it warmed to 15 °C. The reaction was recooled to 0 °C and water (3 mL) was added. Aqueous 3.0 N sodium hydroxide was added (6 mL) followed by water (9 mL). The reaction mixture was filtered through a pad of Celite which was rinsed with EtOAc. The resulting filtrate was concentrated to dryness giving a green solid. The crude product was partially purified via flash sgc using a 40%-50% EtOAc/hexanes gradient, followed by 98%EtOAc/2% Et3N. The fractions containing the impure desired product were combined to give a blue oil. This material was dissolved in CH2CI2 (6 mL). TFA (4 mL) and triethyl silane (2 mL) were added and the reaction mixture was left stirring ON. The reaction mixture was concentrated to give a green oil. This material was purified via flash sgc using 2:1 hexanes: EtOAc, followed by 98%EtOAc/2% Et3N as the mobile phase. Compound 642 was isolated as a yellow oil (0.56 g). MS (El) m/z Obsd M+H 214.2.
Part C:
Compound 642 (0.56 g, 2.6 mmol) was dissolved in CH2CI2 (8 mL). Triethyl amine was added (1 mL) followed by (+)-diacetyl-(L)-tartaric anhydride (0.65 g, 3.00 mmol). The reaction mixture was left stirring at rt ON. The reaction mixture was concentrated to dryness and purified via sgc using 2%-5% MeoH/ CH2CI2 mobile phase with 1 % acetic acid added to it. After evaporating the solvent and azeotopic removal of the acetic acid with heptane, compound 643 was obtained as a white solid (0.33 g). MS (El) m/z Obsd M+H 430.1
Part D:
Compound 643 (92 mg, 0.214 mmol), HOBT (36 mg, 0.26 mmol), DMF(1.5 mL), 2-thiophene ethylamine (44 mg, 0.34 mmol), and N-methylmorpholine (50 μL) were added to a flask. EDC was added and the reaction mixture was left stirring ON at rt. The reaction mixture was diluted with EtOAc and washed with aqueous NaHC03, citric acid, water, and brine. The organic layer was dried with MgS0 and concentrated to give a brown oil. The crude product was purified via flash sgc using a 10%-80% EtOAc/Hexanes gradient. Two diastereomeric compounds were isolated-644 Diastereomer A (0.02 g) MS (El) m/z Obsd M+H 539.06 and 645 Diastereomer B (0.03 g) MS (El) m/z Obsd M+H 539.03.
Part E:
Compound 645 was dissolved in 2 M methanolic ammonia and stirred at rt for 30 m. The reaction was purified to dryness. The crude product was purified via prep TLC on silica plates using 1 :1 EtOAc: Hexanes as the mobile phase. 646B: MS (El) m/z Obsd M+H 455.1.
Figure imgf000450_0001
Example 15:
Figure imgf000451_0001
Part A:
Compound 650 (8.36 g, 73.9 mmol) and tert-butyl carbamate (25.96 g, 222 mmol) were dissolved in acetonitrile (300 mL) and trifluoroacetic acid was added. The reaction was stirred overnight at rt under N2, then concentrated. The resulting mixture was dissolved in EtOAc and washed with water. The organic layer was dried with Na2S04 and concentrated to dryness. The crude product was purified via flash sgc using 1 :3 EtOAc: Hexanes as the mobile phase to give 30.27 g of product 651. 1HNMR (400 MHz, CDCI3) δ 7.78 (s, 1 H), 7.40 (s, 1 H), 6.38-6.30 (m, 1 H), 1.51 (s, 18H) MS (El) m/z Obsd M+H 330.1
Part B:
Compound 651 (30.27 g, 91.89 mmol) was dissolved in 2-propanol (1500 mL) and sodium borohydride (17.0 g, 46 mmol) was added. The reaction mixture was refluxed for 3 h, then concentrated on the rotovap. The resulting material was diluted with EtOAc, washed with water, and dried with Na2S04. The solvents were evaporated and the crude product was purified via flash sgc using 1 :2 EtOAc: Hexanes as the mobile phase to give 1 1.58 g of 652. Yield = 73% over steps one and two. 1HNMR (400 MHz, CDCI3) δ 7.78 (s, 1 H), 7.33 (s, 1 H), 5.35 (s, 1 H), 4.69 (s, 2H), 1.53 (s, 9H); MS (El) MS (El) m/z Obsd M+H 215.0
Part C: Compound 652 (0.30 g, 1.40 mmol) was dissolved in 5 mL of anhydrous THF (5 mL). The flask was capped with a septum, placed under N2 blanket, and cooled in a dry ice/2-propanol bath. A solution of LDA (1.71 mL, 1.8 M) was added via syringe and the reaction mixture was stirred for 0.5 h. A solution of 4-methoxy benzaldehyde (0.21 g, 1.54 mmol) in 5 mL of anhydrous THF was added via syringe. The reaction mixture was stirred for 1 h. The ice bath was removed and the reaction was left stirring overnight at rt. Water was added and the reaction mixture was diluted with EtOAc. The layers were separated. The organic layer was washed with water and dried with Na2S04. The solvent was evaporated and the crude product was purified via flash sgc using 1 :1 EtOAc: Hexanes to give 0.28 g of 653. 1HNMR (400 MHz, CDCI3) δ 7.42 (s, 1 H), 7.37-7.32 (m, 2H), 6.89-6.94 (m, 2H), 6.02 (s, 1 H), 5.38 (broad s, 1 H), 4.55 (s, 2H), 3.86 (s, 3H) 3.28 (s, 1 H), 1.49 (s, 9H).
Part D:
Compound 653 (0.28 g, 0.80 mmol) was dissolved in triethylsilane (2 mL) and TFA (0.6 mL). The reaction mixture was refluxed for 3 h then allowed to cool to rt and stirred overnight. The reaction mixture was concentrated. Methanol was added and the reaction mixture was concentrated. Lithium hydroxide (10 mL, 1.0 M aq) and dioxane (10 mL) were added. The reaction mixture was stirred for 3 h then partially concentrated. EtOAc was added and the layers were separated. The organic layer was washed with water and dried with Na2S04. The reaction mixture was concentrated to give 0.23 g of compound 654. 1HNMR (400 MHz, CDCI3) δ 7.44 (s, 1 H), 7.19 (d, J=9 Hz, 2 H), 6.89 (d, J=9 Hz, 2H), 4.16 (s, 2H), 4.11 (s, 2H), 3.84 (s, 3H), 1.77 (broad s, 2H).
Parts E and F:
Compound 654 was converted to compound 655 using procedures described in Example 2. Data for 655: 1HNMR (400 MHz, CDCI3) δ 7.41-7.26 (m, 5H), 7.22-7.14 (M, 4H), 6.89-6.81 (m, 2H), 5.50-5.17 (m, 1 H), 4.98-4.54 (m, 4H), 4.53-4.31 (m, 2H), 4.06-3.98 (m, 2H), 3.84 (s, 3H), 2.47-2.27 (m, 1 H), 2.12- 1.75 (m, 1 H). MS (El) m/z Obsd M+H 496.1.
Figure imgf000453_0001
Figure imgf000454_0002
Example 16: Thioamides
Example 16A:
Figure imgf000454_0001
Part A:
To a solution of compound 193 (200 mg, 0.54 mmol) in THF (5 mL) was added portionwise of Lawesson's reagent (203 mg, 0.5 mmol). The reaction mixture was stirred overnight under argon at room temperature, and diluted with EtOAc. The organic layer was washed with saturated NaHCO3, brine, dried over Na2SO4, and concentrated. Column chromatography over silica gel (EtOAc/hexane, 20:80) afforded compound 665. Mass calculated for formula C18H22CINO4S 383.1 , observed LCMS m/z 384.1 (M+H).
Part B:
To compound 665 in MeOH (5 mL) was added powdered K2C03 (50 mg). Mixture was stirred at room temperature for 1 h and the solvent was removed in vacuo. The residue was dissolved in water and acidified with 1 N HCI. It was extracted with EtOAc, the combined organic layers was washed with 1 N HCI, brine, dried over Na2SO4, and concentrated to afford compound 666 as an oil (45 mg). Mass calculated for formula C17H20CINO4S 369.1 , observed LCMS m/z 370.1 (M+H).
Part D:
To compound 666 (10 mg, 0.027 mmol) in DMF (0.5 mL) was 229 (11 mg, 0.041 mmol) and HATU (20.5 mg, 0.054 mmol). The reaction mixture was stirred at room temperature for 4 h, and diluted with ethyl acetate and water. The organic layer was washed with 1 N HCI, saturated NaHCO3, and brine. It was dried over Na2SO4, and concentrated, resulting in compound 667 as an oil. Mass calculated for formula C29H30CI2N2O3S2 588.1, observed LCMS m/z 589.1 (M+H).
Part E:
Compound 667 was dissolved in 0.5 mL of TFA/H2O (80:20) and stirred at room temperature for 2 h. The reaction mixture was quenched with ACN/H2O (50:50) and concentrated in vacuo. Purification by reverse phase prep-LC afforded compound 668 as a white solid. HPLC-MS tR = 6.97 min (UV254 nm) 10 min), Mass calculated for formula C26H26CI2N2O3S2 548.1 , observed LCMS m/z 549.1 (M+H).
Example 16B:
Figure imgf000455_0001
Part A:
To 1 (204 mg, 1mmol) in DMF (2 mL) was added 229 (320 mmol, 1.2 mmol) and HATU (570 mg, 1.5 mmol). The reaction mixture was stirred overnight at room temperature, and diluted with ethyl acetate and water. The organic layer was washed with 1 N HCI, saturated NaHC03, and brine. It was dried over Na S0 , and concentrated, resulting in 669 as a slightly yellow oil (280 mg, 66%).
Part B:
To a solution of 669 (280 mg, 0.54 mmol) in THF (5 mL) was added portionwise of Lawesson's reagent (202 mg, 0.5 mmol). The reaction mixture was stirred overnight under argon at room temperature, and diluted with EtOAc. The organic layer was washed with saturated NaHC03, brine, dried over Na2S04, and concentrated. Column chromatography over silica gel (EtOAc/hexane, 20:80) afforded compound 670. Mass calculated for formula C20H22CINO4S 439.1 , observed LCMS m/z 440.1 (M+H).
Part C:
To 670 in MeOH (5 mL) was added powdered K2C03 (50 mg). The mixture was stirred at room temperature for 1 h and the solvent was removed in vacuo. The residue was dissolved in water and acidified with 1 N HCI. It was extracted with EtOAc, the combined organic layers was washed with 1 N HCI, Brine, dried over Na2S0 , and concentrated to afford compound 671 as an oil (80 mg). Mass calculated for formula C 7H2oCIN04S 425.1 , observed LCMS m/z 426.1 (M+H).
Part D:
To compound 671 (10 mg, 0.027 mmol) in DMF (0.5 mL) was added racemic 2-(3-chlorophenyl)pyrrolidine (6.5 mg, 0.035 mmol) and HATU (17.5 mg, 0.046 mmol). The reaction mixture was stirred at room temperature for 4 h, and diluted with ethyl acetate and water. The organic layer was washed with 1 N HCI, saturated NaHC03, and brine. It was dried over Na2S0 , and concentrated, resulting in compound 672 as an oil. Mass calculated for formula C29H30CI2N2O3S2 588.1 , observed LCMS m/z 589.1 (M+H).
Part E:
Compound 672 was dissolved in 0.5 mL of TFA/H20 (80:20) and stirred at room temperature for 2 h. The reaction mixture was quenched with ACN/H20 (50:50) and concentrated. in vacuo. Purification by reverse phase prep-LC afforded compound 673 as a white solid. HPLC-MS tR = 7.04 min (UV254 nm, 10 min), Mass calculated for formula C26H26CI2N203S2 548.1 , observed LCMS m/z 549.1 (M+H).
The following compounds were prepared by the methods described above. Belleau's reagent was substituted for Lawesson's reagent.
Figure imgf000457_0001
Example 17
Figure imgf000457_0002
Compound 675 was prepared as described in Example 4A Part C.
Part A:
To 675 (1.34 g, 3.38 mmol) in DMF (20 mL) was added 4- hydroxybenzylamine (0.5 g, 4.06 mmol), DIEA (0.71 mL, 4.06 mmol) and
HATU (1.54 g, 4.06 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was diluted with water and the aqueous layer was extracted with ethyl acetate. The combined organic layers were dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 80% EtOAc/hexane) afforded 676 (1.41 g, 83%). HPLC-MS tR = 1.56 min (UV254 nm); Mass calculated for formula C25H28FN307 501.2, observed LCMS m/z 502.1 (M+H).
Part B:
To a mixture of 676 (50 mg, 0.1 mmol), triphenylphosphine (79 mg, 0.3 mmol), 3-hexyn-1-ol (33 μL, 0.3 mmol) in THF (0.5 mL) at 0 °C was added DEAD (47 μL, 0.3 mmol). The reaction mixture was warmed to room temperature and stirred for 5 hours. The reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 20% EtOAc/hexane to 100% EtOAc) afforded 677 (9 mg, 15%). HPLC-MS tR = 2.09 min (UV254 nm); Mass calculated for formula C31H36FN307 581.3, observed LCMS m/z 582.2 (M+H).
Part C:
To 677 (9 mg, 0.02 mmol) in methanol (0.5 mL) was added anhydrous hydrazine (2 μL, 0.04 mmol). The reaction mixture was stirred overnight at room temperature. The solvents were removed in vacuo and the material was freeze-dried to yield 678 as a white powder (2 mg). HPLC-MS tR = 1.89 min (UV25 nm); Mass calculated for formula C27H32FN305497.2, observed LCMS m/z 498.2 (M+H).
Example 17B:
Figure imgf000459_0001
Part A:
Compound 680 was prepared from compounds 230 & 679 using procedures described in Example 17A Part A. HPLC-MS tR = 1.37 min (UV254 nm); Mass calculated for formula C24H26N207454.2, observed LCMS m/z 455.2 (M+H).
Part B:
A mixture of 680 (49 mg, 0.11 mmol), 681 (14 uL, 0.14 mmol) and cesium carbonate (46 mg, 0.14 mmol) in DMF (2 mL) was stirred 24 hours. The reaction mixture was poured into water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated to afford 682 (33 mg). The material was used without further purification. HPLC-MS tR = 1.91 min (UV25 nm); Mass calculated for formula C28H32N207 508.2, observed LCMS m/z 509.2 (M+H).
Part C:
To compound 682 (33 mg, 0.06 mmol) in methanol (2 mL) was added 7.0 M ammonia in methanol (2 ml). The reaction mixture was stirred for 1 hour and concentrated. Purification by reverse phase prep-HPLC afforded 683 (8 mg). HPLC-MS tR = 4.28 min (UV254 nm, 10 min); Mass calculated for formula C24H28N2θ5424.2, observed LCMS m/z 425.2 (M+H).
The following compounds were prepared using procedures described in Example 17A & 17B.
Figure imgf000460_0001
Example 17C:
Figure imgf000461_0001
Step A1 :
To pre-swelled MB-CHO resin (5.0 g, 1.0 mmol/g) in 40 ml DCE was added 4- hydroxybenzylamine (1.5 eq) and triacetoxyborohydride (2 eq) and the mixture was agitated overnight before the solution was drained and resin washed with MeOH, DCM and THF 5 cycles to give resin 692 after drying in vacuo overnight.
Step A2:
To a pre-swelled resin 692 (150 mg, 1 mmol/g) in anhydrous THF was added benzylalcohol (5 eq) and PPh3 (7 eq) in 1.5 ml of THF and ADDP (5 eq) in 0.5 ml of DCM. The reaction was agitated at room temperature over the weekend. The reaction solution was drained and resin washed with 5 cycles of MeOH, DCM and THF and dried in vaccuo before it was swelled in NMP followed by addition of 1.5 ml 1 M NMP solution of compound 48. The reaction mixture was agitated overnight before the solution was drained and resin washed with 5 cycles of MeOH, DCM and THF and dried in vacuo to give resin 694.
Step B:
Resin 694 was pre-swelled in DCM, before 1-phenylpiperazine hydrochloric salt (695)(6 eq) was added followed by addition of PyBrop (3 eq) and DIEA (9 eq) in 3 ml of DCM. The reaction solution was agitated overnight before it was drained and resin washed with 5 cycles of MeOH, DCM and THF before it was treated with 10% hydrazine in methanol for 2h. The resin was then further washed with 5 cycles of MeOH, DCM and THF followed by cleavage using 50% of TFA in DCM. The cleavage solution was evaporated and residue purified with a RP-HPLC system to give 5 mg of desired product 696.
The following compounds were generated using similar methods.
Figure imgf000462_0001
Figure imgf000463_0001
Figure imgf000464_0001
Figure imgf000465_0002
Example 17D:
Figure imgf000465_0001
Step A.
To resin 692 ( 2.6 g, 1 mmol/g), preswelled in NMP was added 48 (5 eq) and the mixture was agitated for 48 h. before the solution was drained and resin washed with 5 cycles of MeOH, DCM and THF and dried in vacuo. The resin was then swelled in anhydrous DCM (25 ml) before 769 (6 eq, HCI salt) and DIEA (9 eq) were added followed by addition of PyBrop. The reaction was agitated at r.t. overnight before the solution was drained and resin washed with 5 cycles of MeOH, DCM and THF and dried in vacuo to give resin 770 (0.9 mmol/g loading).
Step B.
To a pre-swelled resin 770 (0.095 g, 0.9 mmol/g) in anhydrous DCM was added anhydrous Cu(OAc)2 (3 eq), 771 (5 eq), 4 A molecular sieves (δmicron particle size, 100 mg) and DIEA (7eq) in 2 ml anh. DCM. The reaction was agitated for 48 h. before the mixture was drained and resin washed with 5 cycles of H2O, MeOH, DCM and THF before it was treated with 10% hydrazine in Methanol for 2h. The resin was then further washed with 5 cycles of MeOH, DCM and THF followed by cleavage using 50% of TFA in DCM. The cleavage solution was evaporated and residue purified with a RP- HPLC system to give 5 mg of desired product 772.
The following compounds were synthesized in similar fashion.
Figure imgf000466_0001
Figure imgf000467_0001
Figure imgf000468_0001
Figure imgf000469_0002
Example 18
Example 18A:
Figure imgf000469_0001
Part A:
Following the procedure described in Example 1 Part A, 1 (163 mg, 0.8 mmol), 2S-phenyl-pyrrolidine (798) (Burgess, L.E.; Meyers, A.I.; J. Org. Chem. 1991, 56, 2294) (147 mg, 0.8 mmol), DIEA (560 μL, 3.2 mmol) and HATU (304 mg, 0.8 mmol) were mixed together in DMF (2 mL). Purification by column chromatography (SiO2, 5%-20% EtOAc/DCM) afforded 799 as an oil (115 mg, 43%). HPLC-MS tR = 1.82 min (UV254 nm); Mass calculated for formula C18H23NO5 333.2, observed LCMS m/z 334.1 (M+H).
Part B:
Following the procedure in Example 1 Part B the material from Part A was saponified. 800: HPLC-MS tR = 1.54 min (UV254 nm); Mass calculated for formula C17H21NO5 319.1 , observed LCMS m/z 320.2 (M+H).
Part C:
Compound 800, 2-thiopheneethylamine (6 μL, 0.05 mmol), DIEA (18 μL, 0.103 mmol) and HATU (19 mg, 0.05 mmol) were mixed together following the procedure described in Example 1 Part A. 801: HPLC-MS tR = 2.01 min (UV254 nm); Mass calculated for formula C23H28N204S 428.2, observed LCMS m/z 429.2 (M+H).
Part D:
Compound 801 was dissolved in 90:10 TFA:water (2 mL) and stirred for 4 hours. The reaction mixture was quenched with 1 :1 acetonitrile:water (4 mL) and concentrated. Purification by reverse phase prep-LC afforded 802 as a white powder (9 mg, 50%, 2 steps). HPLC-MS tR = 1.54 min (UV254 nm); Mass calculated for formula C2oH24N204S 388.2, observed LCMS m/z 389.2 (M+H).
Example 18B:
Figure imgf000470_0001
Part A: Compound 2 (42 mg, 0.14 mmol), 2-phenyl pyrrolidine (22 mg, 0.15 mmol), DIEA (63 μL, 0.36 mmol) and HATU (61 mg, 0.16 mmol) were mixed together following the procedure described in Example 1 Part A. The desired isomer was separated by reverse phase prep-LC to afford 803 as a white solid. HPLC-MS tR = 1.96 min (UV254 nm); Mass calculated for formula C23H28N204S 428.2, observed LCMS m/z 429.1 (M+H).
Part B:
Compound 803 was deprotected using the procedure described in Example 1 Part D to afford 804 as a white powder (5 mg, 19% 2 steps). HPLC-MS tR = 1.50 min (UV254 nm); Mass calculated for formula C2oH24N2θ4S 388.2, observed LCMS m/z 389.2 (M+H).
Figure imgf000471_0001
Example 19:
Figure imgf000472_0001
Part A: 2-Phenyl piperidine
To 5-chlorovaleronitrile (809) (1.23 mL, 11 mmol) and trimethylsilylchloride (4.32 mL, 34 mmol) in toluene (50 mL) under argon at 0 °C was added phenylmagnesium bromide (810) (3M in diethyl ether, 3.67 mL, 11 mmol) dropwise. The reaction mixture was stirred at 0 °C for 1 hour. To the reaction mixture was added methanol (50 mL). Sodium borohydride (1.04 g, 27.5 mmol) was added portionwise. After the vigorous bubbling had subsided the reaction mixture was stirred for 1 hour at room temperature. A solution of 50% sodium hydroxide (5 mL) was added to the mixture and it was stirred overnight. The reaction mixture was filtered to remove the precipitate and the solids were washed with ethyl acetate. The filtrate was concentrated in vacuo. The crude reaction mixture was dissolved in ethyl acetate and water. The aqueous layer had a pH of 9. The layers were separated and the organic layer was washed with brine solution, dried over sodium sulfate and concentrated in vacuo to afford an orange oil. The product was purified by column chromatography (Si02, 20% ethyl acetate/hexane to 20% ethyl acetate hexane + 2% triethylamine) to afford 811 as a polar yellow oil (927 mg, 52%). 1H NMR (400 MHz, CDCI3) δ 7.4 - 7.3 (m, 4H), 7.24 (m, 1 H), 3.60 (dd, 1 H, J = 2.4, 10.4 Hz), 3.21 (m, 1 H), 2.81 (dt, 1 H, J = 3.2, 10.8 Hz), 1.90 (m, 1 H), 1.81 (m, 1 H), 1.68 (m, 1 H), 1.56 (m, 3H); HPLC-MS tR = 0.73 min (MS); mass calculated for formula CnHι5N 161.1 , observed LCMS m/z 162.1 (M+H).
Part B: To 48 (108 mg, 0.5 mmol) in DCM (3 mL) was added 811 (80 mg, 0.5 mmol). The reaction mixture was stirred overnight before being poured into 1.0 N HCI and extracted with DCM. The combined organic layers were dried over sodium sulfate, concentrated and freeze-dried to afford 812 as a white solid (119 mg, 63%). Mass calculated for formula C19H23NO7 377.2, observed LCMS m/z 378.1 (M+H).
Part C:
Prepared as described in Example 1 Part A using 812 (19 mg, 0.05 mmol) to afford a foam 813 (24 mg, 100%). Mass calculated for formula C19H23NO7 486.2, observed LCMS m/z 487.1 (M+H).
Part D:
Compound 813 (24 mg, 0.05 mmol) was deprotected using the procedure described in Example 2A Part B. Purification by reverse-phase prep-LC afforded 814 as a white powder (9 mg, 45%). HPLC-MS tR = 4.38 and 4.42 min (UV254 nm, 10 min); Mass calculated for formula C2ιH26N2θ S 402.2, observed LCMS m/z 403.1 (M+H).
Figure imgf000473_0001
Example 20:
Example 20A:
Figure imgf000474_0001
Part A:
To 1H-Pyrazoie-3-carboxylic acid (816) (274 mg, 2.44 mmol) in DMF (10 mL) was added 2-chlorobenzyl bromide (698 μL, 5.38 mmol) and cesium carbonate (1.67 g, 5.12 mmoi). The reaction mixture was stirred overnight at room temperature. The solids were removed by filtration and the filtrate was concentrated. Purification by column chromatography (Si02) 20% to 30% EtOAc/Hex) separated the regioisomers to afford the 81 A as an oil (136 mg, 15%) and 817B as a white solid (685 mg, 78%).
1-(2-Chloro-benzyl)-1 H-pyrazole-5-carboxylic acid 2-chloro-benzyl ester (817A): 1H NMR (400 MHz, CDCI3) δ 7.61 (d, 1 H, J = 2.0 Hz), 7.37 (m, 3H), 7.31 - 7.11 (m, 4H), 7.02 (d, 1 H, J = 2.0 Hz), 6.53 (dd. 1 H, J = 2.0, 7.6 Hz), 5.91 (s, 2H), 5.39 (s, 2H). HPLC-MS tR = 2.40 min (UV254 nm); mass calculated for formula Ci8Hι4CI2N2θ2 360.0, observed LCMS m/z 361.0 (M+H). 1 -(2-Chloro-benzyl)-1.H-pyrazole-3-carboxylic acid 2-chloro-benzyl ester (817B): 1H NMR (400 MHz, CDCI3) δ 7.51 (m, 1 H), 7.44 (d, 1 H, J = 2.4 Hz), 7.40 (m, 2H), 7.27 (m, 4H), 7.08 (dd, 1 H, J = 1.6, 7.6 Hz), 6.87 (d, 1 H, J = 2.4 Hz), 5.54 (s, 2H), 5.51 (s, 2H); HPLC-MS tR = 2.22 min (UV254 nM); mass calculated for formula Cι8H14CI2N202 360.0, observed LCMS m/z 361.0 (M+H).
Part B:
To 817B (685 mg, 1.90 mmol) in THF (10 mL) was added 1.0 M LiAIH4 (1.14 mL, 1.14 mmol) with ice cooling. The reaction mixture was stirred for 1 hour. The reaction was quenched with water (1 mL), 3 M NaOH (1 mL) and water (3 mL). The organic layer was decanted and the precipitate was washed with EtOAc. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 1 :1 DCM:EtOAc) yielded 818 as an oil (350 mg, 83%). 1H NMR (400 MHz, CDCI3) δ 7.42 (d, 1H, J = 2.4 Hz), 7.39 (dd, 1H, J = 2.4, 8.0 Hz), 7.25 (m, 2H), 7.02 (dd, 1 H, J = 1.6, 7.2 Hz), 6.30 (d, 1 H, J = 2.4 Hz), 5.43 (s, 2H), 4.73 (s, 2H), 2.5 (bs, 1 H, OH); HPLC-MS tR = 1.29 min (UV25 nm); mass calculated for formula CnHιιCIN20 222.1 , observed LCMS m/z 223.1 (M+H).
Part C:
To 818 (350 mg, 1.57 mmol) in toluene (5 mL) was added phosphorous tribromide (163 μL, 1.73 mmol). The reaction mixture was heated to reflux in a pre-heated oil bath for 15 minutes. The mixture was cooled, poured over ice and extracted with EtOAc. The combined organic layer was washed with bicarbonate solution and brine, dried over sodium sulfate and concentrated to yield 819 as a white solid (414 mg, 93%). 1H NMR (400 MHz, CDCI3) δ 7.39 (m, 2H), 7.25 (m, 2H), 7.00 (dd, 1 H, J = 2.0, 7.2 Hz), 6.36 (d, 1 H, J = 2.4 Hz), 5.41 (s, 2H), 4.53 (s, 2H); HPLC-MS tR = 1.96 min (UV25 nm); mass calculated for formula CnH10BrCIN2284.0, observed LCMS m/z 285.1 (M+H).
Part D: To 819 (412 mg, 1.44 mmol) in DMF (5 mL) was added phthalimide (255 mg, 1.73 mmol) and cesium carbonate (515 mg, 1.58 mmol). The reaction mixture was stirred overnight at room temperature. The solids were removed by filtration and the filtrate was concentrated. The residue was dissolved in EtOAc and water. The layers were separated. The organic layer was washed with brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 5% EtOAc/DCM) yielded 820 as a solid (418 mg, 82%). HPLC-MS tR = 1.96 min (UV254 nm); mass calculated for formula C19H14CIN302 351.1 , observed LCMS m/z 352.2 (M+H).
Part E:
To 820 (418 mg, 1.19 mmol) in ethanol (20 mL) was added hydrazine monohydrate (231 μL, 4.75 mmol) and the reaction was heated to reflux for 3 hours. The mixture was cooled and diluted with 50% EtOAc/hexanes (40 mL). The solids were removed by filtration and were thoroughly washed with 50% EtOAc/hexanes (30 mL). The filtrate was concentrated. The residue was dissolved in EtOAc and washed with water and brine, dried over sodium sulfate and concentrated to afford 821 as a semi-solid (205 mg, 78%). 1H NMR (400 MHz, CDCI3) δ 7.39 (m, 1 H), 7.38 (d, 1 H, J = 2.0 Hz), 7.23 (m, 2H), 6.92 (dd, 1 H, J = 2.0, 7.2 Hz), 6.22 (d, 1 H, J = 2.0 Hz), 5.39 (s, 2H), 3.91 (s, 2H); HPLC-MS tR = 0.91 min (UV254 nm); mass calculated for formula CnH-izCINs 221.07, observed LCMS m/z 222.1 (M+H).
Part F:
To 821 (49 mg, 0.22 mmol) in DMF (2 ml) was added 230 (67 mg, 0.2 mmol), DIEA (77 μL, 0.44 mmol) and HATU (84 mg, 0.22 mmol). The reaction mixture was stirred overnight. The DMF was removed in vacuo and the residue was dissolved in EtOAc. The organic layer was washed with bicarbonate solution, 0.1 N HCI, and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Siθ2, 80% EtOAc/Hex) yielded 822 as a solid (70 mg, 65%). HPLC-MS tR = 1.80 min (UV254 nm); mass calculated for formula C27H27CIN406 538.2, observed LCMS m/z 539.2 (M+H). Part G:
A mixture of 822 (70 mg, 0.13 mmol) and potassium carbonate (90 mg, 0.65 mmol) in MeOH (2 mL) was stirred 1 hour. The reaction was diluted with EtOAc and poured into brine solution. Additional salt was added and the layers were separated. The aqueous layer was extracted with EtOAc. The combined organic layer was dried over sodium sulfate and concentrated to yield 823 as a white solid (45 mg, 76%). 1H NMR (400 MHz, DMSO-d6) δ 8.05 (t, 1 H, J = 6.0 Hz), 7.70 (d, 1 H, J = 2.0 Hz), 7.46 (dd, 1 H, J = 2.0, 8.0 Hz), 7.36 - 7.27 (m, 6H), 6.94 (dd, 1 H, J = 2.0, 7.6 Hz), 6.20 (d, 1 H, J = 2.0 Hz), 5.68 (d, 1 H, J = 6.8 Hz), 5.35 (s, 2H), 5.06 (d, 1 H, J = 14.4 Hz), 5.03 (d, 1 H, J = 7.6 Hz), 4.91 (d, 1 H, J = 14.4 Hz), 4.76 (d, 1 H, J = 15.6 Hz), 4.62 (dd, 1 H, J = 2.8, 7.6 Hz), 4.61 (d, 1 H, J = 14.8 Hz), 4.26 (m, 3H); HPLC-MS tR = 1.55 min (UV254 nm); mass calculated for formula C23H23CIN404454.1 , observed LCMS m/z 455.2 (M+H).
Example 20B: Pyrazole Spacer
Figure imgf000477_0001
Part A:
To the Weinreb amide 824 (prepared using the method of De Luca, L.; Giacomelli, G.; Taddei, M. J. Org. Chem. 2001 , 66, 2534) (200 mg, 0.85 mmol) in THF (15 mL) was added lithium (trimethylsilyl)acetylide (4.3 mL, 2.14 mmol) dropwise at 0 °C. The reaction mixture was stirred for 1 hour then diluted with EtOAc (50 mL). The mixture was washed with 0.1 N HCI (50 mL), dried over sodium sulfate a d. concentrated. Purification by column chromatography (Si02, 20% EtOAc/Hex) afforded 825 (91 mg, 54%).
Part B:
A mixture of 825 (71 mg, 0.33 mmol), (2-chloro-phenyl)-hydrazine dihydrochloride (89 mg, 0.39 mmol) and potassium carbonate (200 mg, 1.64 mmol) in methanol (5 mL) were stirred at reflux for 12 hours. The reaction mixture was cooled and diluted with EtOAc (50 mL) and water (50 mL). The organic layer was separated, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 20% EtOAc/Hex) afforded 826 (50 mg, 15%). HPLC-MS tR = 2.10 min (ELSD); mass calculated for formula C17H22CIN302 335.1 , observed LCMS m/z 336.2 (M+H).
Part C:
Compound 826 (45 mg, 0.13 mmol) was dissolved in 25% TFA/DCM (4 mL) and stirred for 30 minutes. The solvents were removed in vacuo and the material was used without further purification. The residue was dissolved in DMF (5 mL) and 230 (25 mg, 0.07 mmol), DIEA (300 μL, 1.68 mmol) and HATU (32 mg, 0.09 mmol) were added. The reaction mixture was stirred overnight. The DMF was removed in vacuo and the residue was dissolved in EtOAc and water. The organic layer was separated and washed with 0.1 N NaOH, 0.1 N HCI and brine, dried over sodium sulfate and concentrated. Compound 827 was used without further purification.
Part D:
Compound 827 (-25 mg, 0.05 mmol) was dissolved in methanol (5 mL) and a solution on potassium carbonate (50 mg) in water (1 mL) was added. The reaction was stirred for 30 minutes. The reaction mixture was diluted with ethyl acetate and brine. The organic layer was separated, dried over sodium sulfate and concentrated. Purification by reverse phase prep-LC afforded 828 as a white powder (15 mg, 65%). 1H NMR (400 MHz, DMSO-d6) δ 7.81 (d, 1 H, J = 8.0 Hz), 7.70 (d, 1 H, J = 1.6 Hz), 7.47 (dd, 1 H, J = 1.6, 7.6 Hz), 7.35 - 7.26 (m, 6H), 6.89 (dd, 1H, J = 1.6, 7.2 Hz), 6.29 (d, 1H, J = 2.0 Hz), 5.38 (s, 2H), 5.04 (d, 1 H, J = 15.2 Hz),.4.97fm, 2H), 4.90 (d, 1 H, J = 14.0 Hz), 4.76 (d, 1 H, J = 16.4 Hz), 4.61 (m, 2H), 4.27 (d, 1 H, J = 2.4 Hz), 1.39 (d, 3H, J = 6.8 Hz); HPLC-MS tR = 4.25 min (UV254 nm, 10 min); mass calculated for formula C24H25CIN40 468.2, observed LCMS m/z 469.1 (M+H).
Figure imgf000479_0001
Example 21 : α-Methyl benzyl amines
Example 21 A:
Figure imgf000480_0001
Part A:
A mixture of 1 R-(4-bromo-phenyl)-ethylamine (837) (0.18 mL, 1.25 mmol), 230 (349 mg, 1.0 mmol), DIEA (357 μL, 2.0 mmol) and HATU (456 mg, 1.2 mmol) in NMP (10 mL) was stirred overnight at room temperature. The reaction mixture was poured into water and extracted with EtOAc. The organic layer was washed with bicarbonate solution, 0.1 N HCI, and brine, dried over sodium sulfate and concentrated. 838: 1H NMR (400 MHz, DMSO-de) δ 8.72 (d, 1 H, J = 7.6 Hz), 7.49 (dd, 2H, J = 2.0, 6.8 Hz), 7.37 - 7.25 (m, 6H), 5.56 (ABq, 2H, J = 6.8 Hz), 4.99 (d, 1 H. J = 13.2 Hz), 4.89 (m, 2H), 4.70 (d, 1 H, J = 16.0 Hz), 4.51 (d, 1 H, J = 16.4 Hz), 2.09 (s, 3H), 2.03 (s, 3H), 1.32 (d, 3H, J = 7.2 Hz).
Part B:
To 838 (135 mg, 0.26 mmol), PdCI2(dppf) (22 mg, 0.03 mmol), potassium phosphate (166 mg, 0.78 mmol) in dioxane (5 mL) under argon atmosphere was added benzyl-9-BBN (0.5 M in THF, 1.3 mL, 0.65 mmol). The reaction mixture was heated to 60 °C overnight. The reaction mixture was filtered through a celite pad and the pad was rinsed with ethyl acetate. The filtrate was concentrated. The crude product was dissolved in methanol (5 mL) and potassium carbonate (4 mg) was added. The reaction mixture was stirred for 1 hour, filtered and concentrated to afford 839 as a solid (50 mg, 42%). 1H NMR (400 MHz, DMSO-d6) δ 7.97 (d, 1 H, J = 7.6 Hz), 7.34 - 7.10 (m, 13H), 5.03 (d, 1H. J = 14.4 Hz), 4.90 (m, 2H), 4.72 (d, 1 H, J = 16.8 Hz), 4.58 (m, 2H), 4.20 (d, 1 H, J = 3.2 Hz), 3.87 (s, 2H), 1.37 (d, 3H, J = 7.2 Hz); HPLC-MS t = 1.89 min (UV254nm); mass calculated for formula C27H28N204444.2, observed LCMS m/z 445.1 (M+H). Example 21 B:
Figure imgf000481_0001
Part A:
CH2CI (300 mL) and trifluoroacetic anhydride (100 g, 0.476 mol) were added to a 2 liter 3-necked flask. The flask was equipped with a drying tube and cooled in an ice-water bath. A solution of (R)- -methylbenzylamine (840) (53.72 g, 0.443 mol) dissolved in 100 mL of CH2CI2was added over 40 min. The ice bath was removed and the reaction mixture was stirred at rt for 3 h. The reaction mixture was cooled with an ice-water bath and methanesulfonic acid (110 mL, 1.69 mol) was added. Dibromodimethylhydantoin (65 g, 0.227 mol) was added in portions over 1 hr 15 min. The reaction mixture was left stirring overnight then poured into a solution prepared from 60 mL of 1 M NaHS03 and 500 mL of ice and water. The layers were separated and the aqueous layer was extracted with 100 mL of CH2CI2. The combined organic layer was washed with water and brine, then dried with MgS04. The solvent was evaporated to give a white solid. This material was recrystallized from Et20/Hexanes to give compound 841. MS (El) m/z M+H Obsd 297.16
Part B:
Compound 841 (8.09 g, 27.3 mmol) was dissolved in 40 mL of dioxane, 10 mL of methanol, and 40 mL of 1 M aqueous LiOH. The reaction mixture was stirred at rt for 2.5 h under N2. Boc anhydride (7.45 g, 34.1 mmol) and EtOAc (25 mL) were added and the reaction mixture was stirred at rt under N2 for 1.5 h. The reaction mixture was diluted with EtOAc and brine. The layers were separated and the aqueous layer was extracted with EtOAc. The combined organic layer was washed with citric acid, water, and brine, then dried with MgS04. The solvents were evaporated and the crude product was purified via flash sgc using a 5%-10% EtOAc/Hexanes gradient as the mobile phase. White solid 842 (7.95 g) was obtained as product. MS (El) m/z M+Na Obsd 324.01
Part C:
Compound 842 (1.11 g, 3.69) and 4-pyridine boronic acid (0.55 g, 4.48 mmol) were suspended in 1-propanol (8 mL) and stirred for 25 min at 40 °C. Palladium (II) acetate (55 mg, 0.24 mmol) and water (4 mL), were added, followed by sodium carbonate (0.47 g, 4.43 mmol) and triphenyl phosphine (197 mg 0.75 mmol). The reaction mixture was stirred under N2 at 80 °C for 21 h. The reaction mixture was allowed to cool to rt and diluted with EtOAc and 0.5 M NaHCθ3. The layers were separated and the aqueous layer was extracted with EtOAc. The combined organic layer was washed with brine, dried with MgS04, filtered, and concentrated to give an orange solid. The crude product was purified via sgc using 30% EtOAc/Hexanes, followed by 30% EtOAc/Hexanes with 2% added diisopropylethylamine added, followed by 40% EtOAc/Hexanes as the mobile phase. White solid 843 was obtained as product (0.71 g). MS (El) m/z M+H Obsd 299.10.
Part D:
Compound 843 (0.70, 2.3 mmol) was suspended in 22 mL of 4 M HCI in dioxane and 8 mL of CH2Cl2. The reaction mixture was stirred under N2 at rt for 6 h, then concentrated to give white solid 844 (0.70 g). MS (El) m/z M+H Obsd 199.09.
Parts E & F:
Compound 846 was prepared using procedures similar to those described in
Example 14, Steps E&F. MS (El) m/z M+H Obsd 460.1
Example 21 C:
Figure imgf000483_0001
Part A:
A 500 mL Schlenck Flask equipped with a stir bar was flame dried under N2 flow, capped with a septum, and allowed to cool to rt. A solution of n-butyl lithium in hexanes (55 mL, 2.5 Molar, 137.4 mmol) was added via syringe. The flask was cooled in a dry ice/2-propanol bath. Tetramethylethylene diamine (TMEDA-19.0 g, 167 mmol) was added. Compound 842 (20 g, 66.8 mmol) was dissolved in anhydrous THF (200 mL) and added to the reaction mixture over 30 min via addition funnel. The reaction mixture was stirred at - 78 C for 15 min. Triethyl borate was added (21.4 mL, 176 mmol) as a solution in 40 mL of anhydrous THF. The reaction mixture was stirred for 30 min. The reaction mixture was quenched at -78 °C with 1.0 M aq HCI to a pH of 2, then allowed to warm to rt. The reaction mixture was extracted with EtOAc. The organic layer was dried with MgS04 and concentrated to dryness. The crude product was purified via sgc using 5:95 MeOH:CH2CI2 as the mobile phase. White solid 848 was obtained as product (3.8 grams). Data for 848: 1HNMR (400 MHz, CD3OD) δ 7.56 (d, J = 8Hz, 2H), 7.29 (d, J = 8Hz, 2H), 4.58-4.64 (m, 1 H), 1.41-1.36 (m, 12H).
Part B:
Compound 848 (300 mg, 1.13 mmol), 3-bromo-2-cyanothiophene (192 mg, 1.02 mmol), acetonitrile (4 mL), Dichloro[1 , l'-bis(diphenylphosphino) ferrocene] palladium(ll) dichloride (Strem, 171 mg, 0.23 mmol), and 1.25 mL of 1 M aq K2C03 were added to a microwave tube equipped with a stir bar. N2 was bubbled through the soln. and the tube was capped. The reaction mixture was irradiated at 150 °C for 5 min in a Personal Chemistry "Companion" microwave oven. The resulting material was filtered through Celite which was washed with EtOAc. The filtrate was partitioned between EtOAc and water. The organic layer was washed with water and treated with Na2S04 and Darco activated carbon. The mixture was filtered and concentrated to dryness. The crude product was purified via sgc using 60:40 Hexanes: EtOAc as the mobile phase. Compound 849 was obtained as a yellow oil (224 mg) which crystallized on standing, m/z Obsd. M+Na 350.93
Part C:
Compound 849 (224 mg, 0.682 mmol) was dissolved in 2.8 mL of CH2CI2 and 1.2 mL of trifluoroacetic acid. The reaction mixture was stirred at rt for 2 hours, then concentrated to dryness. The crude product was purified via sgc using 9:1 CH2CI2 :MeOH (NH ) as the mobile phase to give 110 mg of 850 as an oil. m/z Obsd. M+H 229.10
Parts D and E: Compounds 851 and 852 were prepared via procedures similar to those described in Example 14, Parts D and E. Data for 851 : m/z Obsd. M+H 574.05. Data for 852: m/z Obsd. M+Na 511.91.
Example: 21 D:
Figure imgf000485_0001
855
Compound 853 was synthesized using the procedures described in Example
1.
Part A:
Compound 853 (25 mg, 0.050 mmol) was dissolved in THF (1mL) and PdP(t-
Bu3)2 (5 mg, 0.0097 mmol) was added under an argon atmosphere. 6-
Methoxy-2-pyridylzinc bromide (0.5 M in THF, 0.2 mL) was added and the reaction was stirred at 50 °C overnight. The reaction mixture was filtered over a bed of celite and then evaporated under reduced pressure. Purification by reverse phase prep-LC afforded an off-white solid 854 (10 mg, 38%) after lyophilization. HPLC-MS tR = 6.138 min (UV25 nm, 10 min); mass calculated for formula C31H34CIN3O5 563.2, observed LCMS m/z 564.1 (M+H).
Part B:
Compound 854 (10 mg, 0.0188 mmol) was dissolved in MeOH (0.3 mL) and TFA (4 mL) and stirred for 2 hours at room temperature. The solvent was removed to provide the pure product 855 (9.36 mg, 95%). HPLC-MS tR = 5.098 min (UV254 nm, 10 min); mass calculated for formula C28H30CIN3O5 523.1 , observed LCMS m/z 524.1 (M+H). Example: 21 E:
PartA
Figure imgf000486_0001
Figure imgf000486_0002
856 857
Compound 856 was synthesized using the Suzuki procedures desribed in Example 12. Part A:
Compound 856 (160 mg, 0.30 mmol) was dissolved in acetonitrile (5 mL) and sodium iodide (140 mg, 1.0 mmol) was added followed by trimethylsilylchloride (105 mg, 1.0 mmol). Water (0.1 mL) was added and the reaction mixture was stirred at reflux for 4 hours. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with bicarbonate solution and brine; dried over sodium sulfate and concentrated. Purification by reverse phase prep-LC afforded an off- white solid 857 (86 mg, 56%) after lyophilization. HPLC-MS tR = 3.72 min (UV254 nm, 10 min); mass calculated for formula C27H28CIN3O5 509.1 , observed LCMS m/z 510.1 (M+H).
The following table contains compounds made using the procedures described in Example 21 A - E.
Figure imgf000486_0003
Figure imgf000487_0001
Figure imgf000488_0001
Figure imgf000489_0002
Example 22
Figure imgf000489_0001
Part A:
To 305 (100 mg, 0.34 mmol) in THF (2 mL) at -90 °C under argon was added n-butyl lithium (1.6 M, 0.140 mL, 0.22 mmol) dropwise. The reaction mixture was stirred for 30 minutes at -90 °C. Then diphenyl disulfide (90 mg, 0.4 mmol) in THF (1 mL) was added slowly. The reaction mixture was stirred for 30 minutes at -90 °C. The reaction mixture was quenched with saturated ammonium chloride solution and warmed to room temperature. The mixture was diluted with ethyl acetate (10 L) and the layers were separated. The organic layer was washed with water and brine. Dried over sodium sulfate and concentrated. Purification by column chromatography (SiO2, 5% EtOAc/hexane) afforded 881 (19 mg, 59%). HPLC-MS tR = 2.28 min (UV254 nm,); mass calculated for formula C16H19NO2S2321.09, observed LCMS m/z 322.2 (M+H). Part B:
To 881 (66 mg, 0.2 mmol) was added 4N HCI in dioxane (1 mL). The reaction mixture was stirred for 1 hour. The solvents were removed in vacuo and the crude product was used without further purification. HPLC-MS tR = 1.12 min (UV254 nm); mass calculated for formula CnHnNS2 221.0, observed LCMS m/z 222.1 (M+H).
Part C:
To 563 (56 mg, 0.16 mmol) in DMF (2 mL) was added 882 (66 mg, 0.26 mmol), DIEA (82 μL, 0.46 mmol) and HATU (78 mg, 0.24 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was diluted with ethyl acetate (20 mL) and water (20 mL). The layers were separated and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with sodium bicarbonate solution and brine, dried over sodium sulfate and concentrated. Compound 883 (43 mg, 48%) was used without further purification. HPLC-MS tR = 2.40 min (UV254 nm); mass calculated for formula C28H29CIN2θ4S2 556.1 , observed LCMS m/z 557.0 (M+H).
Part D:
Compound 883 (25 mg, 0.04 mmol) was dissolved in 80:20 TFA:water (1 mL) and stirred for 1.5 hours. The reaction was quenched with 1 :1 wateπacetonitrile (1 mL) and the solvents were removed in vacuo. Purification by reverse phase prep-LC afforded 884 as a white solid (5 mg, 22%). HPLC-MS tR = 2.04 min (UV254 nm); mass calculated for formula C25H25CIN2θ4S2 516.1 , observed LCMS m/z 517.1 (M+H).
Example 23:
Figure imgf000491_0001
Part A:
A mixture of DD'-dibromo-p-xylene (885) (528 mg, 2.0 mmol), phthalimide (294 mg, 2.0 mmol) and cesium carbonate (717 mg, 2.2 mmol) in DMF (5 mL) was stirred for 4 hours. The reaction mixture was filtered and the filtrate was concentrated. The residue was dissolved in DCM and washed with bicarbonate solution and brine. The organic layer was dried over sodium sulfate and concentrated. Recrystallization from 75% EtOAc/hexanes removed the dialkylated product. The mother liquor was concentrated and the monoalkylated product 886 was isolated by column chromatography (Si02) 50% DCM/Hex) as a white solid (110 mg, 16%). 1H NMR (400 MHz, CDCI3) δ 7.84 (d, 2H, J = 2.4 Hz), 7.72 (d, 2H, J = 2.4 Hz), 7.41 (d, 2H, J = 7.2 Hz), 7.34 (d, 2H, J = 7.2 Hz), 4.84 (s, 2H), 4.46 (s, 2H).
Part B:
A mixture of 886 (110 mg, 0.33 mmol), 2-methyl benzimidazole (44 mg, 0.33 mmol) and cesium carbonate (114 mg, 0.35 mmol) in DMF (5 mL) was stirred overnight at room temperature. The reaction mixture was filtered and concentrated. The residue was partitioned between bicarbonate solution and ethyl acetate. The layers were separated and the aqueous layer was extracted with ethyl acetate. The combined organic layer was washed with brine, dried over sodium sulfate and concentrated to afford 887 as an off- white solid (120 mg, 95%). 1H NMR (400 MHz, CDCI3) δ 7.82 (m, 2H), 7.70 (m, 2H), 7.37 (d, 2H, J = 8.0 Hz), 7.25 - 7.18 (m, 4H), 7.00 (d, 2H, J = 8.7 Hz), 5.30 (s, 2H), 4.82 (s, 2H), 2.57 (s, 3H). Part C:
A mixture of 887 (120 mg, 0.31 mmol) and hydrazine hydrate (61 μL, 1.26 mmol) in ethanol (10 mL) was heated to reflux for 3 hours. The reaction mixture was concentrated. The residue was dissolved in ethyl acetate and washed with bicarbonate solution. The aqueous layer was extracted with ethyl acetate. The combined organic layer was washed with brine, dried over sodium sulfate and concentrated to afford 888 as a yellow oil (31 mg, 40%). 1H NMR (400 MHz, CDCI3) δ 7.72 (dd, 1 H, J = 1.2, 8.4 Hz), 7.51 (dd, 1 H, J = 3.2, 6.4 Hz), 7.26 - 7.17 (m, 4H), 7.01 (d, 2H, J = 8 Hz), 5.30 (s, 2H), 3.85 (s, 2H), 2.60 (s, 2H), 2.57 (s, 3H).
Part D:
A mixture of 888 (31 mg, 0.12 mmol), 554 (40 mg, 0.1 mmol), DIEA (38 μL, 0.22 mmol) and HATU (46 mg, 0.12 mmol) in DMF (2 mL) was stirred overnight. The DMF was removed in vacuo. The residue was dissolved in ethyl acetate, washed with bicarbonate solution and brine, dried over sodium sulfate and concentrated to afford 889 as a film (61 mg, 96%). HPLC-MS tR = 1.38 min (UV254 nm); mass calculated for formula C34H35CIN406 630.2, observed LCMS m/z 631.2 (M+H).
Part E:
To 889 (61 mg, 0.097 mmol) in methanol (2 mL) was added anhydrous hydrazine (5 μL, 0.16 mmol). The reaction mixture was stirred overnight, concentrated, purified by reverse phase prep-LC and treated with HCI to afford 890 as an HCI salt (19 mg, 33%). HPLC-MS tR = 3.42 min (UV254 nm, 10 min); mass calculated for formula C30H31CIN4O4 546.2, observed LCMS m/z 547.2 (M+H).
Figure imgf000493_0002
Example 24: Alkyne linkages
Example 24A:
Figure imgf000493_0001
Part A:
To 193 (200 mg, 0.57 mmol) in DCM (5 mL) was added propargyl amine (62 mg, 1.13 mmol), DIEA (396 μL, 1.71 mmol), DMAP (7 mg, 0.06 mmol) and EDC (140 mg, 0.74 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was diluted with ethyl acetate, washed with water and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 60% ethyl acetate/hexanes) afforded 892 as an oil (136 mg, 62%). HPLC-MS tR = 1.76 and 1.83 min (UV254 nm); Mass calculated for formula C2oH23CIN2θ4390.1, observed LCMS m/z 391.1 (M+H).
Part B: To 892 (20 mg, 0.05 mmol) in DMF(1 mL) was added 4-chloro-iodobenzene (24 mg, 0.10 mmol), copper(l) iodide (1 mg, 0.005 mmol), PdCI2(Ph3P)2 (1.8 mg, 0.003 mmol) and triethylamine (1 mL). The reaction mixture was stirred overnight at 50 °C under argon atmosphere. The reaction mixture was diluted with ethyl acetate, washed with 0.1 N HCI, dried over sodium sulfate and concentrated. The material was used without further purification. 893: HPLC-MS tR = 2.28 and 2.32 min (UV254 nm); Mass calculated for formula C26H26Cl2N204 500.1 , observed LCMS m/z 501.1 (M+H).
Part C:
Compound 893 was dissolved in 4:1 TFA:water (2 mL) and stirred at room temperature for 1.5 hours. The reaction was quenched with 1 :1 wateπacetonitrile (2 mL) and concentrated. Purification by reverse phase prep-LC afforded 894 as a solid (2 mg). HPLC-MS tR = 5.02 min (UV254 nm, 10 min); Mass calculated for formula C23H22CI2N204460.1 , observed LCMS m/z 461.2 (M+H).
Example 24 B:
Figure imgf000494_0001
Compound 895 was obtained via procedures described in Example 2A Part A using isoindoline to open the (+)-diacetyl-L-tartaric anhydride and propargyl amine in the coupling procedure.
Part A:
Compound 895 (138 mg, 0.371 mmol), p-bromofluorobenzene (244 mg, 1.39 mmol), and dichlorobis(triphenylphosphine) palladium (II) (26 mg, 0.037 mmol) were dissolved in 1.5 mL CH3CN and 1.5 mL Et3N. The reaction mixture was heated in a sealed tube under N2 at 80 °C for 2 h. The reaction mixture was allowed to cool to rt. EOAc and 1.0 M pH 7.0 sodium phosphate buffer were added. The layers were separated. The organic layer was washed with water and dried with MgS0 . The solvent was evaporated and the crude product was purified via sgc using 3:1 EtOAc:hexanes as the mobile phase to give 20 mg of compound 896. MS (El) m/z Obsd M+H 467.19
Part B:
Compound 896 (20 mg, 0.043 mmol) was dissolved in 3 mL of 2 M methanolic ammonia. The reaction mixture was stirred at rt for 1 h. The reaction mixture was concentrated to dryness. The crude product was purified via prep TLC on silica plates using 97:3 CH2CI2 :MeOH as the mobile phase to give 12 mg of 897. MS (El) m/z Obsd M+H 383.2.
Figure imgf000495_0001
Example 25: N-Linked biaryls
Example 25A:
Figure imgf000496_0001
Part A:
To 193 (42 mg, 0.12 mmol) in DMF (2 mL) was added 4-pyrazol-1-yl- benzylamine (25 mg, 0.14 mmol), DIEA (50 μL, 0.29 mmol) and HATU (53 mg, 0.13 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was poured into water and extracted with ethyl acetate. The combined organic layers were washed with 0.1 N NaOH and brine, dried over sodium sulfate and concentrated. The crude material was used without further purification. 904: HPLC-MS tR = 1.97 and 2.03 min (UV254 nm); Mass calculated for formula C27H29CIN404 508.2, observed LCMS m/z 509.2 (M+H).
Part B:
Compound 904 was dissolved in 4:1 TFA:water (2 mL) and stirred at room temperature for 2 hours. The reaction was quenched by the addition of 1 :1 acetonitrile:water (4 mL) and the solvents were removed in vacuo. Purification by reverse phase prep-LC afforded 905 as a white solid (33 mg). HPLC-MS tR = 4.16 min (UV254 nm, 10 min); Mass calculated for formula C24H25CIN404 468.2, observed LCMS m/z 469.2 (M+H).
Example 25B:
Figure imgf000497_0001
Part A:
A mixture of 4-bromobenzyl amine hydrochloride (558) (2.0 g, 9.0 mmol), monomethyl phthalate (1.94 g, 10.8 mmol), EDC (2.07 g, 10.8 mmol), HOBt (1.82 g, 13.5 mmol) and triethylamine (3.8 mL, 27.0 mmol) in DCM (50 mL) was stirred overnight. The reaction mixture was diluted DCM and washed with 1.0 N HCI, water, bicarbonate solution and brine, dried over sodium sulfate and concentrated. Recrystallization of the mixture from ethyl acetate afforded 906 as a solid (1.72 g, 61%). 1H NMR (400 MHz, CDCI3) δ 7.83 (m, 2H), 7.70 (m, 2H), 7.42 (d, 2H, J = 8.2 Hz), 7.29 (d, 2H, J = 8.2 Hz), 4.79 (s, 2H). HPLC-MS tR = 2.05 min (UV254 nm); Mass calculated for formula Cι5H10BrNO2 315.0, observed LCMS m/z 316.0 (M+H).
Part B:
A mixture of 906 (100 mg, 0.32 mmol), pyrrolidine (34 mg, 0.48 mmol), potassium phosphate (171 mg, 0.80 mmol), Pd2(dba)3 (8 mg, 0.008 mmol) and 2-(dicyclohexylphosphino)biphenyl (11 mg, 0.032 mmol) in dioxane (3 mL) were stirred under argon atmosphere at 90 °C overnight. The reaction mixture was filtered through celite and concentrated. The yellow residue was purified by column chromatography (Si02, 5% EtOAc/DCM) to afford 907 (82 mg, 84%). HPLC-MS tR = 2.13 min (UV254nm); Mass calculated for formula Cι9H18N202 306.1 , observed LCMS m/z 307.2 (M+H).
Part C:
To 907 (82 mg, 0.268 mmol) in 1 :1 ethanol:DCM (4 mL) was added hydrazine monohydrate (52 μL, 1.07 mmol). The reaction mixture was heated to reflux overnight. The precipitate was removed by filtration and the filtrate was concentrated. The residue was dissolved in ethyl acetate, washed with water and brine, dried over sodium sulfate and concentrated to afford 908 as a white solid (23 mg, 49%). HPLC-MS tR = 0.86 min (UV254nm); Mass calculated for formula CnHι6N2 176.1 , observed LCMS m/z 177.1 (M+H).
Part D:
A mixture of 554 (30 mg, 0.076 mmol), 908 (17.3 mg, 0.098 mmol), DIEA (40 μL, 0.226 mmol), DMAP (1 mg) and HATU (37 mg, 0.098 mmol) in DMF (1 mL) was stirred overnight. The reaction mixture was poured in water and extracted with ethyl acetate. The combined organic layers were washed with 0.1 N NaOH, water and brine, dried over sodium sulfate and concentrated. Compound 909 was used without further purification.
Part E:
To 909 in 3:1 methanol:water (2 mL) was added potassium carbonate (20 mg). The reaction mixture was stirred for 30 minutes. The reaction mixture was partitioned between ethyl acetate and water. The organic layer was separated, washed with brine, dried over sodium sulfate and concentrated. Purification by reverse phase prep-LC afforded 910 as a solid (2 mg). HPLC- MS tR = 3.57 min (UV25 nm 10 min); Mass calculated for formula C25H30CIN3O4 471.2, observed LCMS m/z 472.2 (M+H).
Example 25C:
Figure imgf000498_0001
Compound 568 was prepared according to the procedure of Maku, S. et. al. (J. Comb. Chem. 2003, 5, 379)
Part A:
A mixture of compound 568 (740 mg, 3 mmol), 3-methyl pyrazole (123 mg, 1.5 mmol), copper (II) acetate (409 mg, 2.25 mmol), pyridine (0.25 ml), 3 mmol) in tetrahydrofuran (10 mL) was stirred at room temperature open to the air for 60 hours. The reaction mixture was diluted with ethyl acetate, filtered through celite and concentrated. Purification by column chromatography (Siθ2, 30% ethyl acetate/hexanes to 50% ethyl acetate/ hexane) afforded the product as an impure white solid (450 mg). Further purification by column chromatography (Si02, dichloromethane to 5% ethyl acetate / dichloromethane) afforded 911 as a solid (295 mg, 70% purity). 1H NMR (400 MHz, acetone-de) δ 8.97 (bs, 1H, NH), 8.16 (d, 1H, J = 2.4 Hz), 7.76 (d, 2H, J = 8.4 Hz), 7.42 (d, 2H, J = 8.8 Hz), 6.27 (d, 1H, J = 2.4 Hz), 4.55 (d, 2H, J = 5.6 Hz), 2.28 (s, 3H); HPLC-MS tR = 1.62 min (UV25 nm); Mass calculated for formula C13H12F3N3O 283.1 , observed LCMS m/z 284.2 (M+H).
Part B:
To 911 (165 mg, 0.43 mmol) in 1 :1 methanol: water (2 mL) was added 10% potassium carbonate in 2:1 methanol:water (7mL). The reaction mixture was stirred overnight at room temperature. The reaction mixture was concentrated and the residue was dissolved in ethyl acetate. The ethyl acetate layer was washed with water and brine, dried over sodium sulfate and concentrated to afford 912 as a white solid (69 mg, 86%). HPLC-MS tR = 0.75 min (UV254 nm); Mass calculated for formula CnHι3N 187.1 , observed LCMS m/z 188.1 (M+H).
Part C:
To 193 (65 mg, 0.18 mmol) in DMF (2 mL) was added 912 (38 mg, 0.2 mmol), DIEA (70 μL, 0.4 mmol) and HATU (76 mg, 0.21 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was poured into water and extracted with ethyl acetate. The combined organic layers were washed with 0.1 N NaOH and brine, dried over sodium sulfate and concentrated. Compound 913 was used without further purification. HPLC- MS tR = 2.05 and 2.09 min (UV254 nm); Mass calculated for formula CH3iCIN4θ4 522.2, observed LCMS m/z 523.2 (M+H).
Part D:
Compound 913 was dissolved in 4:1 TFA:water (2 mL) and stirred at room temperature for 2 hours. The reaction was quenched by the addition of 1 :1 acetonitrile :water (4 mL) and the solvents were removed in vacuo. Purification by reverse phase prep-LC afforded 914 as a white solid (35 mg). HPLC-MS tR = 4.31 min (UV254 nm, 0 min); Mass calculated for formula C25H27CIN4O4482.1 , observed LCMS m/z 483.1 (M+H).
Example 25D:
Figure imgf000500_0001
Part A: Compound 841 (0.5 g, 1.69 mmol), pyrazole (0.14 g, 2.05 mmol), Cul (0.064 g, 0.336 mmol), 1 ,10-phenanthroline (0.12 g, 0.66 mmol), and Cs2C03 (1.1 g, 3.37 mmol) were dissolved in dimethylacetamide and stirred for 20 h at 140 °C under N2. The reaction mixture was allowed to cool to rt and diluted with EtOAc. The resulting mixture was washed with water and concentrated to dryness. The resulting material was purified via flash sgc using CH2CI2:MeOH:conc NH4OH- (200:10:1 ) as the mobile phase to give 0.30 g of 915. Data for 915: 1HNMR (400 MHz, CDCI3) δ 7.98 (s, 1 H), 7.80-7.74 (m, 3H), 7.48 (d, 2H), 6.55 (s, 1 H), 5.23 (m, 1 H), 1.68 (d, J=4 Hz, 2H). MS (El) of m/z Obsd. M+H 283.97
Part B:
Compound 915 (0.20 g, 0.67 mmol) was dissolved in 10 mL of MeOH. Aqueous LiOH (10 mL/ 1.0 M) was added. The reaction mixture was stirred at rt for 4 h. The solution was partially concentrated in vacuo to remove the MeOH. The resulting material was extracted with EtOAc. The organic layer was washed with water, dried with Na2S04, filtered, and concentrated to give 0.14 g of 916. Data for 916: 1HNMR (400 MHz, CDCI3) δ 7.94 (s, 1 H), 7.75 (s, 1 H), 7.68 (d, J= 9 Hz, 2H), 7.47 (d, J= 9 Hz, 2H), 4.26-4.13 (m, 1 H), 1.44 (d, J= 7 Hz, 3H).
Parts C and D:
Compounds 917 and 918 were prepared via procedures similar to those described in Example 14 -Parts D and E. Data for 917: 1HNMR (400 MHz, CDCI3) δ 7.98-7.62 (m, 4H), 7.46-7.13 (m, 8H), 6.68-6.47 (m, 2H), 5.86-5.41 (m, 2H), 5.24-5.10 (m, 2H), 4.05-3.48 (m, 2H), 2.22-1.79 (m, 10H), 1.58-1.55 (m, 2H). Data for 918: 1HNMR (400 MHz, CDCI3) δ 7.96 (s, 1 H), 7.80 (s, 1H), 7.75-7.59 (m, 2H), 7.48-6.95 (m, 8H), 6.52 (s, 1 H), 5.32-5.13 (m, 2H), 4.97- 4.05 (m, 2H), 3.99-3.68 (m, 3H), 2.46-1.80 (m, 4H), 1.62-1.51 (m, 3H); MS (El) m/z Obsd. M+H 449.1.
Figure imgf000501_0001
Figure imgf000502_0001
Figure imgf000503_0001
Figure imgf000504_0001
Figure imgf000505_0001
Example 26: Aryl-heteroaryl biaryl compounds
Example 26A:
Figure imgf000505_0002
Part A:
To 193 (42 mg, 0.12 mmol) in DMF (2 mL) was added 4-[1 ,2,3]thiadiazol-4-yl- benzylamine (25 mg, 0.13 mmol), DIEA (45 μL, 0.26 mmol) and HATU (50 mg, 0.13 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was poured into water and extracted with ethyl acetate. The combined organic layers were washed with 0.1 N NaOH and brine, dried over sodium sulfate and concentrated. The crude material was used without further purification. 953: Mass calculated for formula C26H27CIN4O4S 526.1 , observed LCMS m/z 527.1 (M+H).
Part B:
Compound 953 was dissolved in 4:1 TFA:water (2 mL) and stirred at room temperature for 2 hours. The reaction was quenched by the addition of 1 :1 acetonitrile:water (4 mL) and the solvents were removed in vacuo. Purification by reverse phase prep-LC afforded 954 as a white solid (21 mg). HPLC-MS t = 4.21 min (UV254nm, 0 min); Mass calculated for formula C23H23CIN4O4S 486.1 , observed LCMS m/z 487.1 (M+H).
Example 26B: Biaryl C-C linkage
Figure imgf000506_0001
Part A:
To 4-aminomethyl-benzoic acid methyl ester (955) (2 g, 9.92 mmol) in dichloromethane (25 mL) was added Boc anhydride (2.27 g, 10.4 mmol) and triethylamine (2.76 mL, 19.84 mmol). The reaction mixture was stirred overnight. The reaction mixture was diluted with dichloromethane, washed with water and brine, dried over sodium sulfate and concentrated to afford 956 as a white solid (2.40 g, 91%). HPLC-MS tR = 1.76 min (UV254nm); Mass calculated for formula C14H19NO4265.1, observed LCMS m/z 288.2 (M+Na). Part B:
Compound 957 (257 mg, 80%) was synthesized following the procedure described in Example 10B Part A. HPLC-MS tR = 1.77 min (UV254nm); Mass calculated for formula C1 H18CIN03 283.1 , observed LCMS m/z 306.1
(M+Na).
Part C:
To 957 (49 mg, 0.17 mmol) in DMF (2 mL) was added thioformamide (21 mg, 0.35 mmol) and pyridine (50 μL). The reaction mixture was stirred for 72 hours. The mixture diluted with ethyl acetate, washed with 0.1 N sodium hydroxide, water and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 25% ethyl acetate/hexanes) afforded 958 (25 mg, 50%). HPLC-MS tR = 1.80 min (UV254nm); Mass calculated for formula C158N202S 290.1 , observed LCMS m/z 291.1 (M+H).
Part D:
Compound 958 (25 mg, 0.084 mmol) was stirred in dichloromethane (2 mL) and TFA (1 mL) at room temperature for 1 hour. The solvents were removed in vacuo to afford 959 as an oil (26 mg). 1H NMR (400 MHz, CD3OD) δ 9.07 (d, 1 H, J = 2.0 Hz), 8.03 (d, 2H, J = 8.4 Hz), 7.96 (d, 1 H, J = 1.6 Hz), 7.52 (d, 2H, J = 8.0 Hz), 4.16 (s, 2H).
Part E:
To 193 (23 mg, 0.065 mmol) in DMF (2 mL) was added 959 (26 mg, 0.084 mmol), DIEA (34 μL, 0.20 mmol) and HATU (32 mg, 0.084 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was poured into water and extracted with ethyl acetate. The combined organic layers were washed with 0.1 N NaOH and brine, dried over sodium sulfate and concentrated. The crude material was used without further purification. 960: HPLC-MS tR = 2.00 and 2.05 min (UV254 nm); Mass calculated for formula C27H28CiN3θ4S 525.2, observed LCMS m/z 526.1 (M+H). Part F:
Compound 960 was dissolved in 4:1 TFA:water (2 ml) and stirred at room temperature for 2 hours. The reaction was quenched by the addition of 1 :1 acetonitrile:water (4 mL) and the solvents were removed in vacuo. Purification by reverse phase prep-LC afforded 961 as a white solid (14 mg). HPLC-MS tR = 4.17 min (UV254 nm, 10 min); Mass calculated for formula C24H24CIN304S 485.1 , observed LCMS m/z 486.1 (M+H).
Example 26C: Biaryl C-C linkage
Figure imgf000508_0001
Part A:
To 4-(tert-butoxycarbonylamino-methyl)-benzoic acid (962) (500 mg, 1.99 mmol) in tetrahydrofuran (5 mL) in an ice bath was added DIEA (347 μL, 1.99 mmol). The reaction mixture was stirred for 15 minutes and ethyl chloroformate (190 μL, 1.99 mmol) was added. The reaction mixture was stirred for an additional 15 minutes and ammonia in dioxane (0.5 M, 4.18 mL, 2.09 mmol) was added. The reaction mixture was warmed to room temperature and stirred for 2 hours. The solvent was removed in vacuo and the residue was partitioned between ethyl acetate and water. The layers were separated and the aqueous layer was saturated with sodium chloride and extracted with ethyl acetate. The combined organic layers were washed brine, dried over sodium sulfate and concentrated to afford 963 as a white solid (540 mg), which contained some mixed anhydride as an impurity. 1H NMR (400 MHz, CD3OD) δ7.81 (d, 2H, J = 8.0 Hz), 7.35 (d, 2H, J = 8.4 Hz), 7.21 (bs, 1 H, NH), 4.28 (s, 2H), 1.47 (s, 9H).
Part B:
To 963 (500 mg, 2.0 mmol) in tetrahydrofuran (20 mL) was added Lawesson's reagent (485 mg, 1.2 mmol). The reaction mixture was stirred overnight at room temperature. The solvent was removed in vacuo and the residue was dissolved in ethyl acetate. The mixture was washed with 0.1 N sodium hydroxide, water and brine, dried over sodium sulfate and concentrated to a yellow residue. Purification by column chromatography (Siθ2, 40% ethyl acetate/hexanes) afforded 964 as a pale yellow solid (432 mg, 81%). 1H NMR (400 MHz, CDCI3) δ 7.84 (d, 2H, J = 8.4 Hz), 7.32 (d, 2H, J = 8.4 Hz), 4.36 (d, 2H, J = 5.6 Hz), 1.48 (s, 9H).
Part C:
To 964 (70 mg, 0.263 mmol) in DMF (2 mL) was added chloroacetaldehyde (50% in water, 41 mg, 0.526 mmol). The reaction was stirred overnight at room temperature. An additional 2 equivalents of chloroacetaldehyde was added and the reaction was stirred for 24 hours. The reaction was still not complete so the mixture was heated to 50 °C for 2 days. The mixture diluted with ethyl acetate, washed with 0.1 N sodium hydroxide, water and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 25% ethyl acetate/hexanes) afforded 965 (31 mg). HPLC-MS tR = 1.84 min (UV254 nm); Mass calculated for formula Cι58N202S 290.1 , observed LCMS m/z 291.1 (M+H).
Part D:
Compound 965 (31 mg, 0.106 mmol) was stirred in dichloromethane (2 mL) and TFA (1 mL) at room temperature for 1 hour. The solvents were removed in vacuo to afford 966 as an oil (60 mg). 1H NMR (400 MHz, CD3OD) δ 8.03 (d, 2H, J = 8.8 Hz), 7.90 (d, 1 H, J = 3.2 Hz), 7.65 (d, 1 H, J = 3.6 Hz), 7.56 (d, 2H, J = 8.8 Hz), 4.18 (s, 2H).
Part E:
To 193 (29 mg, 0.082 mmol) in DMF (2 mL) was added 966 (32 mg, 0.106 mmol), DIEA (43 μL, 0.25 mmol) and HATU (40 mg, 0.106 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was poured into water and extracted with ethyl acetate. The combined organic layers were washed with 0.1 N NaOH and brine, dried over sodium sulfate and concentrated. The crude material was used without further purification. 967: HPLC-MS tR = 2.03 and 2.08 min (UV25 nm); Mass calculated for formula C27H28CIN304S 525.2, observed LCMS m/z 526.1 (M+H).
Part F:
Compound 967 was dissolved in 4:1 TFA:water (2 mL) and stirred at room temperature for 2 hours. The reaction was quenched by the addition of 1 :1 acetonitriIe:water (4 mL) and the solvents were removed in vacuo. Purification by reverse phase prep-LC afforded 968 as a white solid (15 mg). HPLC-MS tR = 4.21 min (UV254 nm, 10 min); Mass calculated for formula C24H2 CIN3θ4S 485.1 , observed LCMS m/z 486.1 (M+H).
Figure imgf000510_0001
Figure imgf000511_0001
Example 27: Piperidine-aryl compounds
Example 27A: Piperidine-aryl
, N
977
Figure imgf000511_0002
PartA: Step 1 : A mixture of piperidin-4-ylmethyl-carbamic acid tert-butyl ester (977) (2.5 g, 11.66 mmol), 2-fluoro-3-cyanobenzene (1.55 g, 12.8 mmol) and DIEA (3 mL, 17.5 mmol) in NMP (5 mL) under argon atmosphere was stirred overnight at 120 °C. The reaction mixture was diluted with ethyl acetate, washed with water and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Siθ2, 25% ethyl acetate/hexanes) afforded a white solid (3.1 g).
Step 2: To the material from Step 1 (2 g) in dichloromethane (5 mL) at 0 °C was added TFA (5 mL). The mixture was stirred at room temperature for 1 hour. The mixture was quenched with acetonitrile and concentrated. The residue was dissolved in ethyl acetate, washed with sodium carbonate solution and brine, dried over sodium sulfate and concentrated to yield 978 as an oil which solidified to a waxy solid (920 mg). HPLC-MS tR = 0.63 min (UV254 nm); Mass calculated for formula Ci3H17N3215.1 , observed LCMS m/z 216.2 (M+H).
Part B:
To 978 (120 mg, 0.55 mmol) in DMF (2 mL) was added 230 (167 mg, 0.5 mmol), DIEA (0.21 mL, 1.2 mmol) and HATU (211 mg, 0.55 mmol). The mixture was stirred for 3 hours at room temperature. The reaction mixture was diluted with ethyl acetate, washed with saturated sodium bicarbonate and brine, dried over sodium sulfate and concentrated. The material was used without further purification.
Part C:
To 979 in methanol (4 mL) was added 7.0 N ammonia in methanol (2 mL) was added. The mixture was stirred for 2 hours at room temperature and concentrated. Purification by reverse phase prep-LC afforded 980 as a white powder (80 mg) upon lypholization. HPLC-MS tR = 4.03 min (UV254nm); Mass calculated for formula C25H28N4θ4448.2, observed LCMS m/z 449.2 (M+H). Example 27B: Piperidine-aryl
Figure imgf000513_0001
Part A:
A mixture of 977 (200 mg, 0.93 mmol), 3-iodothiophene (981 ) (294 mg, 1.4 mmol), copper(l)iodide (36 mg, 0.19 mmol), proline (43 mg, 0.37 mmol) and potassium carbonate (258 mg, 1.87 mmol) in DMSO (1.5 mL) was stirred overnight at 80 °C in a 4 mL vial. The reaction mixture was diluted with ethyl acetate and washed with water (3X) and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 25% ethyl acetate / hexanes) afforded 982 (94 mg, 34%).
Part B:
Compound 982 (42 mg, 0.14 mmol) was dissolved in DCM (2 mL) and TFA (1 mL). The mixture was stirred for 1 hour and concentrated to afford 983 in quantitative yield. HPLC-MS tR = 0.35 min (UV25 nm); Mass calculated for formula Cι0H16N2S 196.1 , observed LCMS m/z 197.2 (M+H).
Part C:
Compound 984 was prepared according to the procedure described in Example 27A Part B. HPLC-MS tR = 1.36 min (UV254nm); Mass calculated for formula C26H31N3O6S 513.2, observed LCMS m/z 514.2 (M+H).
Part D: Compound 985 was prepared according to the method described in Example 25 Part E. HPLC-MS tR = 2.85 min (UV254nm, 10 min.); Mass calculated for formula C22H27N304S 429.2, observed LCMS m/z 430.1 (M+H).
Example 27C:
Figure imgf000514_0001
Part D
H0X> Br
W A }X Part E
NC NC
989 990
Figure imgf000514_0002
Part A:
Compound 986 (1.13 g, 7.23 mmol) was dissolved in N-methylpyrrolidinone (4 mL) and 2-fluorobenzonitrile (0.90 g, 7.5 mmol) was added followed by DIEA (2.5 mL, 14.4 mmol). The reaction was stirred at 120 °C overnight. The cooled reaction mixture was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with bicarbonate solution and brine; dried over sodium sulfate and concentrated. Purification by column chromatography (Siθ2, 25% ethyl acetate/hexanes) afforded a white solid 987 (1.5 g, 81 %). HPLC-MS tR = 1.95 (UV254nm); mass calculated for formula C 5H18N2O2 258.14, observed LCMS m/z 259.1 (M+H). Part B:
Diisopropylamine (0.330 mL, 2.32 mmol) was dissolved in THF (20 mL) and cooled in an ice bath. A solution of n-BuLi (2.5M in hexanes, 1 mL) was added dropwise and stirred for 15 minutes. The reaction mixture was then cooled to -78 °C and a solution of compound 987 (400 mg, 1.6 mmol) in THF (10 mL) was added dropwise. Stirring was continued at this temperature for 30 minutes before a solution of iodomethane (450 mg, 3.2 mmol) in THF (10 mL) was added dropwise. The reaction was stirred for 30 minutes at this temperature and then 1 hour at room temperature. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with bicarbonate solution and brine; dried over sodium sulfate and concentrated to provide the product 988 that was used without purification. HPLC-MS tR = 2.10 (UV254nm); mass calculated for formula C16H2oN202 272.15, observed LCMS m/z 273.2 (M+H).
Part C:
Compound 988 was dissolved in THF (20 mL) and lithium borohydride (60 mg, 2.6 mmol) was added. The reaction mixture was refluxed for 5 hours. The cooled reaction mixture was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with bicarbonate solution and brine; dried over sodium sulfate and concentrated. Purification by column chromatography (Siθ2, 50% ethyl acetate/hexanes) afforded a white solid 989 (300 mg). HPLC-MS tR = 1.57 (UV254 nm); mass calculated for formula Cι4H18N20 230.1 , observed LCMS m/z 231.3 (M+H).
Part D:
Compound 989 (300 mg, 1.3 mmol) was dissolved in toluene (5 mL) and phosphorus tribromide (0.06 mL, 0.52 mmol) was added. The reaction was stirred at reflux for 3 hours. The cooled reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with bicarbonate solution and brine; dried over sodium sulfate and concentrated to provide the product 990 that was used without purification. Part E:
Compound 990 was dissolved in DMF (10 mL) and cesium carbonate (850 mg, 2.6 mmol) and phthalimide (190 mg, 1.3 mmol) were added and stirred overnight. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with bicarbonate solution and brine; dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 25% ethyl acetate/hexanes) afforded a white solid 991 (90 mg). HPLC-MS tR = 2.14 min (UV254 nm); mass calculated for formula C22H2ιN302 359.1 , observed LCMS m/z 360.1.
Part F:
Compound 991 (90 mg, 0.25 mmol) was dissolved in ethanol (3 mL) and hydrazine hydrate (0.5 mL) was added. The reaction was stirred for 3 hours and the solids were filtered. The solvent was evaporated to provide the desired product 992 that was used without purification.
Part G:
Compound 992 was dissolved in DMF (5 mL) and compound 230 (84 mg, 0.25 mmol) and HATU (114 mg, 0.30 mmol) were added and stirred overnight. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with bicarbonate solution and brine; dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, ethyl acetate) afforded a white foam 993 (100 mg). HPLC-MS tR = 1.99 min (UV25 n ); mass calculated for formula C3oH3 N406 546.2, observed LCMS m/z 547.2.
Part H:
Compound 993 (100 mg, 0.182 mmol) was dissolved in methanol (5 mL) and potassium carbonate (100 mg) was added and stirred for 30 minutes. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with bicarbonate solution and brine; dried over sodium sulfate and concentrated. Purification by reverse phase prep-LC afforded a white solid 994 (7 mg, 8%) after lyophilization. HPLC-MS tR = 4.32 min (UV254 nm, 10 min); mass calculated for formula C26H3oN4θ4 462.23, observed LCMS m/z 463.1 (M+H).
The following table contains compounds that were prepared using the procedures described in Example 27 A - C. Compounds such as 1002 were made from the chloropyridyl precursor using the procedures described in Example 27A Part A.
Figure imgf000517_0001
Figure imgf000518_0001
Figure imgf000519_0001
Figure imgf000520_0001
Figure imgf000521_0001
Figure imgf000522_0001
Example 28: Alkyiated phenyl pyrrolidines
Figure imgf000523_0001
Part A:
2-Phenyl pyrrolidine (1041) (3.14 g, 21.4 mmol) was dissolved in THF (40 mL). Aqueous NaOH (12 mL, 25%) was added, followed by Boc anhydride. The reaction mixture was stirred at rt for 70 h. The layers were separated. The aqueous layer was extracted with EtOAc. The combined organic layer was washed with 1.0 M pH=7.0 sodium phosphate buffer and brine, then dried with MgSθ4. Evaporation of the solvents gave a yellow oil which was purified by column chromatography (Siθ2, 9:1 Hexanes: EtOAc) to give a 5.1 g of a clear oil as product 1042. MS (El) m/z M+Na Obsd 270.08
Part B:
A 100 mL Schlenck flask was equipped with a stir bar, flame dried under N2 flow, capped with a septum, and allowed to cool to rt. Compound 1042 (0.50 g, 2.02 mmol) was added and the flask was recapped. Anhydrous THF (8.5 mL) was added via syringe and the flask was cooled in a dry ice/2-propanol bath. Sec butyl lithium (1.6 mL, 2.34 mmol) was added via syringe. The reaction mixture was left stirring at -78 °C for 30 m. lodoethane (180 μL, 2.25 mmol) was added and the reaction mixture was stirred for 2 hr. Additional iodoethane was added (80 μL, 1.0 mmol) was added and the reaction mixture was stirred for 45 minutes. The reaction mixture was quenched with 1.0 M pH 7.0 sodium phosphate buffer and diluted with EtOAc. The layers were separated and the aqueous layer was extracted with EtOAc. The combined organic layer was washed with water and brine, then dried with MgS04 and concentrated to a clear oil. The crude product was purified by column chromatography (Si02, 0%-20% EtOAc/Hexanes) to give 0.12 g of compound 1043. MS (El) m/z M+Na Obsd 298.15.
Part C:
Compound 1043 (0.12 g, 0.43 mmol) was dissolved in 10 mL of 4 M HCI in dioxane. The reaction mixture was stirred at rt for 2.25 h. The reaction mixture was concentrated to dryness giving white solid 1044 (112 mg). MS (El) m/z Obsd M+H 176.12.
Part D:
Compound 1044 (104 mg, 0.49 mmol) and 1 (0.120 g, 0.588 mmol) were dissolved in CH2CI2 (2 mL). Diisopropyl ethylamine (200 μL, 1.12 mmol) was added, followed by PyBrop (249 mg, 0.78 mmol). The reaction mixture was left stirring overnight at rt under N2. The reaction mixture was diluted with CH2CI2 then washed with 1.0 m pH 7.0 sodium phosphate buffer, aq NaHCθ3, and brine. The organic layer was dried with MgS04 and concentrated to a clear oil. The crude product was purified purified by column chromatography (Si02, 0%-40% EtOAc/Hexanes) to give 0.16 g of compound 1045. MS (El) m/z Obsd M+H 362.08
Part E:
Compound 1045 (0.15 g, 0.42 mmol) was dissolved in dioxane (1.6 mL) and water (0.4 mL). Lithium hydroxide was added (19 mg, 0.45 mmol). The reaction mixture was stirred at rt for 2 h 15 m. The solution was concentrated to give 1046 as a clear oil. MS (El) m/z Obsd M+H 348.09
Part F:
Compound 1046 (69 mg, 0.19 mmol) and 845 (51 mg, 0.22 mmol) were dissolved in DMF (1 mL) and diisopropylamine (100 μL, 0.58 mmol). PyBrop was added (108 mg, 0.338 mmol) and the reaction was stirred overnight at rt. The reaction mixture was diluted with EtOAc and washed with 1.0 M pH 8.0 sodium phosphate buffer and brine. The organic layer was dried with MgS04 and concentrated to a yellow oil. The crude product was purified via prep TLC on silica plates using 95:5 CH2CI2:MeOH as the mobile phase. Compound 1047 was obtained as a clear oil (74 mg). MS (El) m/z Obsd M+H 528.20.
Part G:
Compound 1047 (74 mg, 0.14 mmol) was dissolved in 5 mL of 9:1 trifluoroacetic acid:water solution. The reaction mixture was stirred at rt for 3 h 10 m, then concentrated to dryness. The crude product was purified purified by column chromatography (Si02, 0%-7% MeOH/ CH2CI2) to give 26 mg of compound 863. MS (El) m/z Obsd M+H 488.1.
Compound 1048A,B was prepared from Boc-proline OMe using the above alkylation procedures and the thiazole ring was constructed as described in Example 10A.
Figure imgf000525_0001
Example 29:
Example 29A:
Figure imgf000526_0001
Part A:
A mixture of 2-amino-benzenethiol (1049) (247 uL, 2.31 mmol) and 48 (500 mg, 2.31 mmol) in benzene was heated at reflux overnight with a dean-stark trap. The reaction mixture was cooled and concentrated. The mixture was recrystallized from 30% ethyl acetate/hexanes to afford 1050 (267 mg) as a brown powder. HPLC-MS tR = 1.35 min (UV254 nm); mass calculated for formula C14H13N06S 323.1 , observed LCMS m/z 324.1 (M+H).
Part B:
Compound 1051 was prepared from compound 1050 (32 mg, 0.1 mmol) using the standard HATU coupling strategy described in Example 2 Part A. Purification by column chromatography (Si02, 10% ethyl acetate/dichloromethane) afforded the product as an orange solid (27 mg). HPLC-MS tR = 1.78 min (UV254 nm); mass calculated for formula C20H20N2O5S2 432.1 , observed LCMS m/z 433.0 (M+H).
Part C:
Compound 1052 was prepared following the procedure described in Example 2 Part B. HPLC-MS tR = 1.39 min (UV254 nm); mass calculated for formula C16Hi6N2θ3S2 348.1 , observed LCMS m/z 349.0 (M+H).
Figure imgf000526_0002
Figure imgf000527_0002
Example 29B:
Figure imgf000527_0001
Part A:
To sodium hydride (95%, 115 mg, 4.55 mmol) in DME (10 mL) under argon was added triethylphophonacetate dropwise. Within a few minutes the reaction mixture was clear. After 1 hour the aldehyde 1055 (400 uL, 4.55 mmol) was added. After 15 minutes water and diethyl ether were added to the reaction mixture. The layers were separated and the aqueous layer was extracted with diethyl ether. The combined organic layer was washed with brine, dried over sodium sulfate and concentrated to afford 1056 as an orange oil (666 mg, 80%). 1H NMR (400 MHz, CDCI3) δ 7.92 (d, 1 H, J = 3.2 Hz), 7.79 (d, 1 H, J = 16.0 Hz), 7.43 (d, 1 H, J = 3.2 Hz), 6.71 (d, 1 H, J = 16.0 Hz), 4.29 (q, 2H, J = 7.5 Hz), 1.36 (t, 3H, J = 7.5 Hz).
Part B:
To ester 1056 (84 mg, 0.46 mmol) in THF (2 mL) was added 1.0 M lithium hydroxide solution ( 0.5 mL, 0.5 mmol). The reaction mixture was stirred overnight. The reaction mixture was made acidic with 1.0 N HCI solution and extracted with ethyl acetate. Sodium chloride was added to the aqueous layer during the extractions. The combined organic layer was dried over sodium sulfate and concentrated to afford 1057 as a film (65 mg, 92%). 1H NMR (400 MHz, CDCI3) δ 7.97 (d, 1 H, J = 3.0 Hz), 7.89 (d, 1 H, J = 15.7 Hz), 7.49 (d, 1 H, J = 3.0 Hz), 6.75 (d, 1H, J = 15.7 Hz).
Part C:
Compound 1058 was prepared from Compound 1057 (32 mg, 0.21 mmol) using the standard HATU coupling strategy described in Example 2 Part A. Purification by column chromatography (Si02, 50% ethyl acetate/hexane) afforded the product as a film (35 mg, 63%). HPLC-MS tR = 1.35 min (UV25 nm); mass calculated for formula Cι22N2OS2 264.0, observed LCMS m/z 265.1 (M+H).
Part D:
Compound 1059 was prepared using a modified Sharpless dihydroxylation procedure (Chem. Rev. 1994, 94, 2483). A flask was charged with (DHQ)2PHAL (10 mg, 10 mol %), K3Fe(CN)6 (128 mg, 0.39 mmol), postassium carbonate (54 mg, 0.39 mmol), methane sulfonamide (24 mg, 0.26 mmol) and potassium osmium tetraoxide dihydrate (1 mg, 2 mol%). To the solids was added alkene 4 (35 mg, 0.13 mmol) in tert-butanol:water (1 :1 , 2.5 mL). The reaction mixture was stirred overnight at room temperature. The reaction mixture was cooled in an ice bath and sodium metabisulfite (38 mg, 0.2 mmol) was added. The mixture was stirred for 30 minutes. The reaction mixture was diluted with ethyl acetate and water. The layers were separated and the aqueous layer was extracted with ethyl acetate. The combined organic layer was washed with 2.0 N potassium hydroxide solution and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 80% ethyl acetate/hexane) afforded the product as a white solid after lypholization (5 mg, 13%). HPLC-MS tR = 1.00 min (UV254 nm); mass calculated for formula Ci2Hi4N2θ3S2 298.0, observed LCMS m/z 299.0 (M+H).
Figure imgf000529_0002
Example 29C:
Figure imgf000529_0001
Part A:
Compound 1062 was prepared following the procedures described in
Example 1.
HPLC-MS tR = 1.69 min (UV254 nm); mass calculated for formula Cι8H28N2θ6S
400.2, observed LCMS m/z 401.2 (M+H).
Part B:
Compound 1062 (198 mg, 0.69 mmol) and ammonium acetate (1.9 g, 24.7 mmol) in acetic acid (6 ml) were heated overnight at 110 °C. The reaction mixture was concentrated. The residue was suspended in DCM and the solids removed by filtration. The filtrate was concentrated to afford a mixture of acetonide protected product and 1063. The residue was treated with 80% TFA:water (2 mL) and stirred overnight. The solvents were removed. Purification by prep-HPLC afforded 1063 (11 mg, 4%) as a TFA salt. HPLC- MS tR = 0.79 min (UV254 nm); mass calculated for formula C13H17N303S 295.1 , observed LCMS m/z 296.1 (M+H).
Example 29D:
Figure imgf000530_0001
Part A:
Compound 1064 was prepared following the procedures described in
Example 1.
HPLC-MS tR = 1.77 min (UV254 nm); mass calculated for formula C21H24N2O5S
416.14, observed LCMS m/z 417.1 (M+H).
Part B:
Compound 1064 (216 mg, 0.52 mmol) and ammonium acetate (2 g, 25.9 mmol) in acetic acid (6 ml) were heated overnight at 110 °C. The reaction mixture was concentrated. The residue was suspended in DCM and the solids removed by filtration. The filtrate was concentrated and purified by reverse phase chromatography (Gilson) to afford 1065 (80 mg, 30%) as a TFA salt (80% purity). HPLC-MS tR = 1.38 min (UV254 nm); mass calculated for formula C21H23N3O3S 397.1 , observed LCMS m/z 398.2 (M+H).
Part C:
A mixture of compound 1065 (63 mg, 0.127 mmol), iodomethane (10 μL, 0.16 mmol) and cesium carbonate (206 mg, 0.634 mmol) in DMF (6 ml) was stirred overnight at room temperature. The reaction mixture was concentrated. The residue was suspended in ethyl acetate and the solids removed by filtration. The filtrate was concentrated to afford the crude acetonide protected product. The residue was treated with 90% TFA:water (3 mL) and stirred for 3 h at 50 °C. The solvents were removed. Purification by prep-HPLC afforded 1066 (12 mg, 19%) as a TFA salt. HPLC-MS tR = 2.87 min (10 min; UV25 nm); mass calculated for formula C19H21N3O3S 371.1 , observed LCMS m/z 372.1 (M+H).
Example 29E:
Figure imgf000531_0001
Part A:
According to a modification of a procedure by Goker, H. et. al. (II Farmaco 1998, 53, 415-420) a mixture of 1-fluoro-2-nitro-benzene (1067) (1.1 mL, 10.4 mmol) and benzylamine (2.35 mL, 21.5 mmol) in DMF (5 mL) was heated overnight at 80 °C. The reaction mixture was concentrated to give an orange solid which was filtered, washed with water and dried to afford benzyl-(2-nitro- phenyl)-amine (1068) (2.5 g, 100%). 1H NMR δ (400 MHz, CDCI3) 8.44 (b, 1 H, NH), 8.22-8.19 (dd, 1H), 7.41-7.27 (m, 6H), 6.84-6.81 (dd, 1H), 6.70-6.66 (m, 1 H), 4.58-4.57 (d, 2H).
Part B:
To a solution of benzyl-(2-nitro-phenyl)-amine (1068) (2.5 g, 10.43 mmol) in ethanol (150 mL) and water (10 mL) was added iron (8.7 g, 155.7 mmol) followed by 30 drops of concentrated HCI, and the resulting mixture was heated overnight at reflux. The reaction mixture was quenched with water, diluted with DCM and the layers were separated. The organic layer was washed with saturated sodium bicarbonate solution, brine, dried over sodium sulfate and concentrated to give a brown oily residue. Further purification by column chromatography (Si02, 20 % ethyl acetate/hexanes) afforded N- benzyl-benzene-1 ,2-diamine (1069) as a dark yellow oil (1.18 g, 57%). 1H NMR (400 MHz, CDCI3) δ 7.41-7.24 (m, 5H), 6.82-6.67 (m, 4H), 4.32 (s, 2H), 3.56 (b, 3H, NH).
Part C:
Compound 1070 was prepared following the procedures described in Example 29C Part A. HPLC-MS tR = 2.13 min (UV254 nm); mass calculated for formula C26H29N3O4S 479.2, observed LCMS m/z 480.1 (M+H).
Part D:
A mixture of compound 1070 (75 mg, 0.156 mmol) and p-toluenesulfonic acid monohydrate (30 mg, 0.156 mmol) in toluene (3 ml) was heated overnight at reflux. The reaction mixture was concentrated to a residue identified as the acetonide deprotected product by LC-MS. Further purification by reverse phase chromatography afforded 1071 (40 mg, 48%) as a TFA salt (95% purity). HPLC-MS tR = 3.52 min (10 min; UV254 nm); mass calculated for formula C23H23N3O3S 421.1 , observed LCMS m/z 422.2 (M+H).
The following compounds were synthesized by procedures described in Example 29C-E.
Figure imgf000532_0001
Figure imgf000533_0002
Example 29F:
1 PartA^ [T-' -CCHHOO Part B, VN Part C, ft o -o C02Et C02H
1074 1075 1076 1077
Figure imgf000533_0001
Part A:
According to a procedure Hodges, J. C. et al (J. Org. Chem. 1991 , 56, 449- 452) to a cold (-78 °C) solution of oxazole (1074) (1g, 14.4 mmol) in 45 mL of THF was added dropwise nBuLi (9 mL, 14.4 mmol, 1.6 M soln in hexane), and the resulting solution was stirred at -78 °C for 25 min. Then DMF (1.12 mL, 14.4 mmol) was added dropwise, followed by THF (5 mL) and the resulting mixture was allowed to warm up slowly to room temperature, and was stirred overnight. The reaction mixture was quenched by the addition of silica gel (3 g), concentrated, and the resulting slurry was diluted with DCM and loaded on a silica gel column. Flash chromatography (3% ethyl acetate/DCM) afforded oxazole-2-carbaldehyde (1075) (165 mg, 12%) as a yellow oil; 1H NMR (400 MHz, CDCI3) δ 9.79 (s, 1 H), 7.9 (s, 1 H), 7.46 (s, 1 H). Part B:
Compound 1076 was prepared from compound 1075 (165 mg, 1.7 mmol) using the procedure described in Example 29B Part A. Purification by column chromatography (Si02, 20% ethyl acetate/hexane) afforded the product as a yellow solid (107 mg, 34%); 1H NMR (400 MHz, CDCI3) δ 7.69 (s, 1 H), 7.45 (d, 1 H, J = 16.0 Hz), 7.27 (s, 1 H), 6.74 (d, 1 H, J = 16.0 Hz), 4.29 (q, 2H, J = 7.3 Hz), 1.36 (t, 3H, J = 7.3 Hz). Part C:
Compound 1077 was prepared from compound 1076 (107 mg, 0.58 mmol) using the procedure described in Example 29B Part B. Work-up afforded the product as a pale yellow solid (77 mg, 85%). 1H NMR (400 MHz, DMSO-d6) δ 12.9 (s, 1 H), 8.25 (s, 1 H), 7.44 (s, 1 H), 7.27 (d, 1 H, J = 16.0 Hz), 6.62 (d, 1 H, J = 16.0 Hz).
Part D:
Compound 1078 was prepared from compound 1077 (165 mg, 1.7 mmol) using the procedure described in Example 29B Part C. After standard HATU coupling work-up the solid residue was triturated with 1 :1 DCM/hexane, filtered and washed with 1 :1 DCM/hexane to give 1078 (123 mg, 70%) as a pale yellow solid; 1H NMR (400 MHz, DMSO-d6) δ 8.49 (t, 1 H), 8.20 (s, 1 H), 7.69-7.66 (dd, 1 H), 7.49-7.44 (m, 1 H), 7.38 (s, 1 H), 7.20-7.16 (dd, 1 H), 7.14 (d, 1 H, J = 16.0 Hz), 6.94 (d, 1 H, J = 16.0 Hz), 3.41-3.38 (d, 2H), 3.16 (t, 2H), 2.72 (t, 2H), 1.81-1.78 (d, 2H), 1.64-1.58 (m, 1 H), 1.40-1.30 (m, 2H). HPLC- MS tR = 1.63 min (UV254 nm); mass calculated for formula Ci9H-ιgFN402 354.15, observed LCMS m/z 355.1 (M+H).
Part E:
Compound 1079 was prepared from compound 1078 (118 mg, 0.33 mmol) using the procedure described in Example 29B Part D. Purification by column chromatography (Siθ2, 5% MeOH/DCM) afforded the product as a white solid (28 mg, 22%); 1H NMR (400 MHz, DMSO-d6) δ 8.02 (s, 1 H), 7.88 (t, 1H), 7.69-7.66 (dd, 1 H), 7.48-7.43 (m, 1 H), 7.20-7.16 (dd, H), 7.14 (s, 1 H), 5.83- 5.81 (d, 1 H), 5.72-5.70 (d, 1 H), 4.93-4.90 (dd, 1 H), 4.23-4.21 (d of d, 1 H), 3.38-3.36 (d, 2H), 3.04 (t, 2H), 2.69 (t, 2H), 1.77-1.72 (d, 2H), 1.62-1.56 (m, 1 H), 1.32-1.23 (m, 2H). 19F NMR (400 MHz, DMSO-d6) δ -120.6 ppm. HPLC- MS tR = 3.32 min (10 min; UV254 nm); mass calculated for formula Cι9H2ιFN40 388.15, observed LCMS m/z 389.2 (M+H).
Example 30: Example 30A:
Figure imgf000535_0001
Part A:
A mixture of 2-(5-bromo-pentyl)-isoindole-1 ,3-dione (1080) (2.0 g, 6.77 mmol), 2-trifluoromethyl-1 H-benzoimidazole (1.2 g, 6.45 mmol) and potassium carbonate (1.78 g, 12.9 mmol) in NMP (5 ml) was stirred overnight at room temperature. The reaction mixture was partitioned between ethyl acetate and water, and the resulting organic layer was washed with water, brine, dried over sodium sulfate and concentrated to give an oil. Further purification by column chromatography (Si02, 25 % ethyl acetate/hexanes) afforded compound 1082 as a colorless oil (1.88 g, 73%). 1H NMR (400 MHz, CDCI3) δ 7.88-7.86 (d, 1 H), 7.85-7.83 (m, 2H), 7.73-7.71 (m, 2H), 7.49-7.34 (m, 3H), 4.31 (t, 2H), 3.72 (t, 2H), 1.99-1.92 (m, 2H), 1.80-1.73 (m, 2H), 1.53-1.45 (m, 2H).
Part B:
To a solution of compound 1082 (1.88 g, 4.68 mmol) in EtOH (50 mL) was added hydrazine monohydrate (0.45 mL, 9.36 mmol) and the resulting mixture was heated for 3 h at 60 °C, during which time a white precipitate formed. The reaction mixture was filtered, washed with EtOH and the filtrate was concentrated. The residue was suspended in ether, stirred for 10 min at room temperature, filtered and washed with ether, and the resulting filtrate was concentrated to afford compound 1083 as an oil (1.10 g, 87%). 1H NMR (400 MHz, CDCI3) δ 7.90-7.88 (d, 1 H), 7.46-7.37 (m, 3H), 4.33 (t, 2H), 2.76 (t, 2H), 2.02-1.40 (m, 6H).
Part C:
Compound 1084 was prepared from 1083 and 230 following the procedures described in Example 2.
Part D:
Compound 1085 was prepared following the procedures described in Example 2. HPLC-MS tR = 4.14 min (10 min; UV25 nm); mass calculated for formula C25H27F3N4θ4 504.2, observed LCMS m/z 505.1 (M+H).
Example 30B:
Part A:
According to a modification of a procedure by Lown, J. W. et. al. (J. Org. Chem. 1982, 47, 2027-2033) a mixture of 2-(2-amino-ethylsulfanyl)-ethanol (1086) (1.82, 15 mmol) and phthalimide (2.22 g, 15 mmol) in toluene (35 ml) was heated overnight at reflux. The reaction mixture was concentrated to a residue, which was purified by column chromatography (Siθ2, 25 % ethyl acetate/DCM to 50% ethyl acetate/DCM) to afford compound 1087 as an off- white solid (2.98 g, 79%). 1H NMR (400 MHz, CDCI3) δ7.87-7.85 (m, 2H), 7.75-7.72 (m, 2H), 3.94 (t, 2H), 3.80 (t, 2H), 2.89-2.82 (m, 4H).
Part B:
According to a modification of a procedure by Nair, S. A. et. al. (Synthesis 1995, 810-814) to an ice-cold solution of compound 1087 (628 mg, 2.5 mmol) and carbon tetrabromide (1.04 g, 3.12 mmol) in DCM (25 mL) was added triphenylphosphine (920 mg, 3.5 mmol) in two portions over 10 min. The reaction mixture was allowed to warm up to room temperature and stirred overnight at room temperature. The mixture was concentrated and the resulting residue was purified by column chromatography (Si02, 30 % ethyl acetate/hexanes) to afford compound 1088 as a white solid (733 mg, 93%). H NMR (400 MHz, CDCI3) δ7.88-7.86 (m, 2H), 7.75-7.73 (m, 2H), 3.92 (t, 2H), 3.53 (t, 2H), 3.04 (t, 2H), 2.90 (t, 2H).
Part C:
Compound 1089 was prepared following the procedures described in Example 30A, Part A. 1H NMR (400 MHz, CDCI3) δ7.89-7.87 (d, 1 H), 7.86- 7.84 (m, 2H), 7.74-7.72 (m, 2H), 7.58-7.56 (d, 1 H), 7.46 (t, 1 H), 7.38 (t, 1 H), 4.52 (t, 2H), 3.92 (t, 2H), 3.02 (t, 2H), 2.89 (t, 2H); 19F NMR (400 MHz, CDCI3) δ-64.7 ppm.
Part D:
To a solution of compound 1089 (250 mg, 0.6 mmol) in DCM (5 mL) was added m-chloroperoxybenzoic acid (336 mg, 1.5 mmol) and the resulting mixture was stirred overnight at room temperature. The reaction mixture was diluted with DCM, washed with saturated sodium bicarbonate solution, brine, dried over sodium sulfate and concentrated to give compound 1090 (284 mg) as a white solid which was used without further purification. 1H NMR (400 MHz, CDCI3) δ7.92-7.90 (d, 1 H), 7.90-7.88 (m, 2H), 7.78-7.76 (m, 2H), 7.66- 7.62 (d of t, 1 H), 7.54-7.51 (t of d, 1 H), 7.46-7.44 (t of d, 1 H), 4.89 (t, 2H), 4.20 (t, 2H), 3.65 (t, 2H), 3.46 (t, 2H); 19F NMR (400 MHz, CDCI3) δ -62.5 ppm.
Part E:
Compound 1091 was prepared following the procedures described in Example 30A, Part B. 1H NMR (400 MHz, CDCI3) δ7.91-7.89 (d, 1 H), 7.63- 7.61 (d, 1 H), 7.51 -7.48 (t of d, 1 H), 7.44-7.40 (t of d, 1 H), 4.90 (t, 2H), 3.71 (t, 2H), 3.31 (t, 2H), 3.12 (t, 2H); 19F NMR (400 MHz, CDCl3) δ-62.5 ppm.
Part F:
Compound 1092 was prepared following the procedures described in Example 27A, Part B, and used without further purification. HPLC-MS tR = 1.65 min (UV254 nm); mass calculated for formula C28H29F3N408S 638.17, observed LCMS m/z 639.1 (M+H).
Part G:
Compound 1093 was prepared following the procedures described in
Example 27A, Part C. HPLC-MS tR = 3.57 min (10 min; UV254 nm); mass calculated for formula C24H25F3N 06S 554.14, observed LCMS m/z 555.1
(M+H).
Example 30 C:
Figure imgf000539_0001
1096
Figure imgf000539_0002
Part A:
According to a modification of a procedure by Brown, R. F. et. al. (J. Am. Chem. Soc. 1955, 77, 1083-1089) a mixture of 3,3-dimethyl glutaric anhydride (1094) (7.1 g, 50 mmol) and methanol (40 mL) was heated overnight at reflux. The reaction mixture was concentrated to give compound 1095 as a colorless oil (8.46 g, 97%). 1H NMR (400 MHz, CDCI3) δ 3.70 (s, 3H), 2.50 (s, 2H), 2.48 (s, 2H), 1.16 (s, 6H).
Part B:
To an ice-cold solution of compound 1095 (8.46 g, 48.5 mmol) in DCM (30 mL) was added dropwise oxalyl chloride (7 mL, 80 mmol), followed by DMF (2 drops) and the resulting mixture was heated for 3.5 h at reflux. The reaction mixture was concentrated to give the corresponding acid chloride (4- chlorocarbonyl-3,3-dimethyl-butyric acid methyl ester) as a brown oil (7.89 g,
84%) which was used without further purification in the next step.
To an ice-cold solution of ammonia in 1 ,4-dioxane (30 mL of a solution containing 2.2 g ammonia dissolved in 100 mL of 1 ,4-dioxane, 38.8 mmol approx) was added dropwise a solution of 4-chlorocarbonyl-3,3-dimethyl- butyric acid methyl ester (3 g, 15.5 mmol) in 1 ,4-dioxane (3 mL) and after the addition was complete the resulting white slurry was stirred 45 min at room temperature. The reaction mixture was diluted with ethyl acetate, filtered and concentrated to give compound 1096 as a viscous yellow oil (2.62 g, 97%). 1H NMR (400 MHz, CDCI3) δ 3.71 (s, 3H), 2.43 (s, 2H), 2.31 (s, 2H), 1.14 (s, 6H).
Part C:
To an ice-cold solution of LAH (42 mL, 1 M sol in THF, 42 mmol) was added dropwise a solution of compound 1096 (2.02 g, 11.6 mmol) in THF and the resulting mixture was heated overnight at reflux. The reaction mixture was cooled to 0 °C and quenched by dropwise addition of brine, when a paste-like solid formed. This solid was triturated and sonicated several times with ethyl acetate, and the combined organic extracts were concentrated to give compound 1097 as an oil (1.17 g, 76%). 1H NMR (400 MHz, CDCI3) δ 3.71 (t, 2H), 2.71 (t, 2H), 1.53 (t, 2H), 1.41 (t, 2H), 0.92 (s, 6H).
Part D:
Compound 1098 was prepared following the procedures described in Example 30B, Part A. HPLC-MS tR = 1.88 min (UV254 nm); mass calculated for formula C159N03 261.14, observed LCMS m/z 262.0 (M+H).
Part E:
Compound 1099 was prepared following the procedures described in Example 30B, Part B. 1H NMR (400 MHz, CDCI3) δ 7.84-7.82 (m, 2H), 7.71- 7.69 (m, 2H), 3.70-3.66 (m, 2H), 3.47-3.42 (m, 2H), 1.97-1.92 (m, 2H), 1.61- 1.57 (m, 2H), 1.03 (s, 6H).
Part F:
Compound 1100 was prepared following the procedures described in Example 30B, Part C. 1H NMR (400 MHz, CDCI3) δ 7.89-7.87 (d, 1 H), 7.86- 7.84 (m, 2H), 7.73-7.71 (m, 2H), 7.5-7.57 (d, 1 H), 7.47-7.43 (t of d, 1 H), 7.39- 7.35 (t of d, 1 H), 4.46-4.41 (m, 2H), 3.79-3.75 (m, 2H), 1.89-1.85 (m, 2H), 1.74-1.69 (m, 2H), 1.18 (s, 6H); 19F NMR (400 MHz, CDCI3) δ-62.7 ppm. Part G:
Compound 1101 was prepared following the procedures described in Example 30B, Part E. HPLC-MS tR = 1.13 min (UV25 nm); mass calculated for formula C15H20F3N3 299.16, observed LCMS m/z 300.1 (M+H).
Part H:
Compound 1102 was prepared following the procedures described in Example 27A, Part B, and used without further purification. HPLC-MS tR = 2.0 min (UV25 nm); mass calculated for formula C31 H35F3N406 616.25, observed LCMS m/z 617.2 (M+H).
Part i:
Compound 1103 was prepared following the procedures described in
Example 27A, Part C. HPLC-MS tR = 4.52 min (10 min, UV254 nm); mass calculated for formula C27H3iF3N404 532.23, observed LCMS m/z 533.2
(M+H).
The following compounds were synthesized using procedures described in Example 30 A-C.
Figure imgf000541_0001
Figure imgf000542_0001
Example 31 :
Figure imgf000543_0001
1120 1121
Part A:
Compound 1115 (790 mg, 7.5 mmol) was dissolved in THF (10 mL) and cooled in an ice bath. 2-Methylphenylisocyanate (500 mg, 3.75 mmol) was added in portions and the resulting solution was stirred at room temperature for 2 hours. The reaction mixture was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with 1 N HCI solution, bicarbonate solution and brine; dried over sodium sulfate and concentrated. Purification by column chromatography (Si0 , 50% ethyl acetate/hexanes) afforded a white solid 1116 (1.02 g, 56%). HPLC-MS tR = 0.95 (UV254nm); mass calculated for formula C12H18N2O3238.1 , observed LCMS m/z 239.2 (M+H).
Part B:
Compound 1116 (1.02 g, 4.2 mmol) was dissolved in THF (25 mL) and cooled in an ice bath. Potassium f-butoxide (1.5 g, 13.2 mmol) was added in portions and the reaction mixture was stirred for 10 minutes. A solution of p- toluenesulfonyl chloride (1.91 g, 10.8 mmol) in THF (5 mL) was added dropwise and the reaction was stirred 30 minutes at 0°C and 30 minutes at room temperature. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with 1 N HCI solution, bicarbonate solution and brine; dried over sodium sulfate and concentrated. Purification by column chromatography (Siθ2, 50% ethyl acetate/hexanes) afforded a white solid 1117 (0.750 g, 48%). HPLC-MS tR = 1.87 min (UV254 nm); mass calculated for formula Cι9H22N204S 374.1 , observed LCMS m/z 375.1 (M+H).
Part C:
Compound 1117 (750 mg, 2.0 mmol) was dissolved in DMF (10 mL) and phthalimide (441 mg, 3.0 mmol) and cesium carbonate (1.95 g, 6.0 mmol) were added and stirred overnight. The reaction mixture was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with 1 N HCI solution, bicarbonate solution and brine; dried over sodium sulfate and concentrated. The solid residue was recrystallized from ethyl acetate/hexanes to provide 1118 (500 mg, 72%) as a white solid. HPLC-MS tR = 1.63 (UV25 nm); mass calculated for formula C2oH19N3θ3 349.1 , observed LCMS m/z 350.2 (M+H).
Part D:
Compound 1118 (110 mg, 0.29 mmol) was dissolved in ethanol (3 mL) and hydrazine hydrate (0.10 mL) was added. The reaction was stirred for 3 hours at room temperature. The solids were removed by filtration and the solution was evaporated under reduced pressure. The residue was dissolved in 1 N NaOH solution and extracted with ethyl acetate. The combined organic layers were washed with brine; dried over sodium sulfate and concentrated to provide compound 1119 that was used without purification.
Part E:
Compound 1119, 230 (97 mg, 0.29 mmol), DIEA (0.125 mL, 0.7 mmol) and HATU (133 mg, 0.348 mmol) were combined in DMF (3 ml) and stirred overnight. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with 1 N HCI solution, bicarbonate solution and brine; dried over sodium sulfate and concentrated to provide compound 1120 that was used in the next step without purification. HPLC-MS tR = 1.68 min (UV254 nm); mass calculated for formula C28H32N θ7 536.2, observed LCMS m/z 537.3(M+H). Part F:
Compound 1120 was dissolved in methanol (5 mL) and potassium carbonate (150 mg) was added and stirred for 30 minutes. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with 1 N HCI solution, bicarbonate solution and brine; dried over sodium sulfate and concentrated. Purification by reverse phase prep-LC afforded 1121 as a white solid (16 mg) after lyophilization. HPLC-MS tR = 3.48 min (UV25 nm> 10 min); mass calculated for formula C24H28N4O5452.2, observed LCMS m/z 453.1 (M+H).
Figure imgf000545_0002
Example 32:
Figure imgf000545_0001
Part A:
Compound 1124 (360 mg, 2.00 mmol) was dissolved in diethylether (10 mL) and cooled to -70 °C. Titanium isopropoxide (624 mg, 2.2 mmol) was added and stirred for 10 minutes. Ethylmagnesium chloride (1 M in THF, 4 mL, 4 mmol) was added dropwise and the reaction was stirred another 10 minutes at this temperature before warming to room temperature and stirring for an additional 30 minutes. Boron trifluoroetherate (864 mg, 4 mmol) was added and the solution was stirred for 2 hours. The reaction was quenched with 1 N HCI and washed with diethylether. The aqueous layer was made basic by addition of 1 N NaOH solution and then extracted with ethyl acetate. The combined organic layers were washed with brine; dried over sodium sulfate and concentrated to provide compound 1125 that was used without purification.
Part B:
Compound 1125 was dissolved in DMF (5 mL) and compound 230 (50 mg, 0.140 mmol) and HATU (69 mg, 0.182 mmol) were added and stirred overnight. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with 1 N HCI, bicarbonate solution and brine; dried over sodium sulfate and concentrated. Purification by column chromatography (Siθ2, 50% ethyl acetate/hexanes) afforded a white solid 1126 (60 mg, 81 %). HPLC-MS tR = 1.928 (UV254 nm); mass calculated for formula
Figure imgf000546_0001
528.0, observed LCMS m/z 529.1 (M+H).
Part C:
Compound 1126 (60 mg, 0.1427 mmol) was dissolved in MeOH (5 mL) and potassium carbonate (100 mg) was added followed by stirring for 30 minutes. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with bicarbonate solution and brine; dried over sodium sulfate and concentrated. Purification by reverse phase prep-LC afforded a white solid 1127 (10 mg, 15.8%) after lyophilization. HPLC-MS tR = 4.192 min (UV254 nm, 10 min); mass calculated for formula C2ιH2iBrN2θ4444.0, observed LCMS m/z 445.1 (M+H). Part D:
Compound 1127 (7.6 mg, 0.017 mmol) was dissolved in dioxane (2 mL) and Pd(dba)3 (3 mg), triphenylphosphine (4.4 mg, 0.017 mmol), potassium phosphate (8 mg, 0.0377 mmol), and 2-methylphenylboronic acid (4.3 mg, 0.034 mmol) were added under a nitrogen atmosphere. The reaction mixture was heated to 90 °C overnight and then filtered over a bed of celite and concentrated. Purification by reverse phase prep-LC afforded a white solid 1128 (5 mg, 65%) after lyophilization. HPLC-MS tR = 4.095 min (UV254 nm, 10 min); mass calculated for formula C28H28N2θ4 456.2, observed LCMS m/z 457.3 (M+H).
Figure imgf000547_0002
Example 33: Constrained Analogs
Example 33A:
Figure imgf000547_0001
Part A:
To 2-amino-3-thiophen-2-yl-propionic acid methyl ester hydrochloride (1130)
(120 mg, 0.54 mmol) in DMF (2 mL) was added 230 (150 mg, 0.45 mmol), DIEA (160 DM, 0.9 mmol) and HATU (205 mg, 0.54 mmol). The reaction mixture was stirred overnight at room temperature, and diluted with ethyl acetate and water. The organic layer was washed with 1 N HCI, saturated NaHC03, and brine. It was dried over Na2S04, and concentrated, resulting in compound 1131.
Part B:
Compound 1131 was dissolved in 5 mL of MeOH, 10 mL of 10% K2C03 aqueous solution was added. After stirring at room temperature for 1 h, the reaction mixture was concentrated in vacuo, and then acified with 1 N HCL to PH 2. The solution was then extracted with EtOAc, the organic layer was washed with brine, dried over Na24, and concentrated, affording compound 1132 as a white solid. Mass calculated for formula Cι9H20N2O6S 404.1 , observed LCMS m/z 405.0.0 (M+H).
Part C:
Compound 1132 (40 mg, 0.1 mmol) in 1 mL of DMF was added with PS carbodiimide resin (117 mg, 1.28 mmol/g loading, 0.15 mmol) and dimethylaminopyridine (3 mg, 0.024 mmol). After stirring overnight at 50 °C, the solid was filtered off, and the solution was concentrated in vacuo. Purification by reverse phase prep-LC afforded compound 1133 as a white solid. HPLC-MS tR = 3.59 min (UV254 nm, 10 min), Mass calculated for formula C25H26CIN3θ4S 386.1 , observed LCMS m/z 387.0 (M+H).
Figure imgf000548_0001
Example 33B:
Figure imgf000549_0001
1137 1138
Part A:
2-Benzylamino-ethanol (1135) (0.676 g, 4.5 mmol) in DMF (10 mL) was added 230 (1.0 g, 3 mmol), and HATU (1.71 g, 4.5 mmol). The reaction mixture was stirred overnight at room temperature, and diluted with ethyl acetate and water. The organic layer was washed with 1 N HCI, saturated NaHC03, and brine. It was dried over Na2S04, and concentrated, resulting in compound 1136 (1.1 g, 80%). Mass calculated for formula C25H28N2θ7 468.2, observed LCMS m/z 469.1 (M+H).
Part B:
Compound 1136 (468 mg, 1 mmol) and DIEA (261 mL, 1.5 mmol) in CH2CI2 (5 mL) was added dropwise with mesyl chloride (116 mL, 1.5 mmol) at 0 °C. After stirring at 0 °C for 10 min, the reaction mixture was allowed to stir at room temperature for 1.5 h, then concentrated to dryness. The residue was extracted with ethyl acetate, washed with 1 N HCI, saturated NaHC03, and brine. It was dried over Na2Sθ4, and concentrated, resulting in 1137 (511 mg, 93%).
Part C:
Compound 1137 (389 mg, 0.71 mmol) in MeOH (20 mL) was added with solid K2CO3 (323 mg, 2.34 mmol). After stirring at room temperature for 2 h, it was concentrated to remove the solvent, then dissolved in EtOAc and water. The organic layer was washed with 1 N HCI, saturated NaHC03, brine, and concentrated. Purification by reverse phase prep-LC afforded 1138 as a white solid. HPLC-MS tR = 3.89 min (UV254 nm, 10 min), Mass calculated for formula C21H22N2O4 366.16, observed LCMS m/z 367.1 (M+H).
Example 33C:
Figure imgf000550_0001
Part A:
Step 1 : (R)- Boc-prolinol (1139) (2.0 g, 9.94 mmol) in THF (5 mL) was added into NaH (0.437 g, 60% in mineral oil, 10.93 mmol) in 10 mL of THF at 0 °C. After stirring at 0 °C for 10 min, allyl bromide (1.32 g, 10.93 mmol) was added. Te reaction mixture was allowed to stir overnight at room temperature. After filtration, the solution was concentrated to dryness, the residue was taken up with EtOAc, washed with 1 N citric acid, saturated NaHC03, and brine, dried over Na2S0 , and concentrated, resulting in a colorless oil (2.30 g, 96%). Step 2: The above material in CH2CI2 (10 mL) was added dropwise with 3 mL of TFA at 0 °C. After stirring at room temperature for 1 h, the solution was concentrated. The residue was dissolved in EtOAc, washed with concentrated Na23, brine, dried over over Na24, and concentrated, resulting in compound 1140 as a colorless oil (1.06 g, 75% for two steps).
Part B: Step 1 : To compound 1140 (500 mg, 3.54 mmol) in DMF (10 mL) was added 1 (867 mg, 4.25 mmol) DIEA (1.48 mL, 8.5 mmol) and HATU (1.616 g, 4.25 mmol) at 0 °C. After stirring at room temperature for 5 h, the reaction mixture was diluted with ethyl acetate and water. The organic phase was washed with 1 N citric acid, saturated NaHC0 , and brine. It was dried over Na2S0 , and concentrated. Column purification (Si02, EtOAc/Hexane 40:60) afforded a colorless oil (650 mg, 57%).
Step 2: The above material was treated with 5 mL of TFA/H20 (80:20) at 0 °C. After stirring at room temperature for 2 h, it was concentrated to dryness. Column purification (Si02, EtOAc/Hexane 70:30) afforded compound 1141 (500 mg, 88%) as a colorless oil.
Part C:
Step 1 : To compound 1141 (250 mg, 0.87 mmol) in toluene (20 mL) was added dibutyltin oxide (217 mg, 0.87 mmol). The reaction mixture was refluxed for 1 h, using Dean-stark apparatus for azotropic removal of water. After cooling to room temperature, the solvent was stripped to dryness under reduced pressure. The resulting oil was dried in vacuo for 2 h. Step 2: To the above oil was added CsF (198 mg, 1.30 mmol). The mixture was then placed in vacuo for 2 h. Allyl bromide (220 mg, 1.83 mmol) in DMF (10 mL) was added at 0 °C. After stirring at room temperature under argone for 30 h, the reaction mixture was concentrated, and then taken up with EtOAc. The solution was washed with saturated KF solution, and brine, dried over Na2S04, and concentrated. The organic layer was washed with 1 N HCI, saturated NaHC03, brine, and concentrated. Column chromatography (S1O2, EtOAc/Hexane 60:40) afforded compound 1142 as a colorless oil (130 mg, 45%). Mass calculated for formula C16H25N06 327.17, observed LCMS m/z 328.1 (M+H).
Part D:
Compound 1142 (110 mg, 0.33 mmol) in dichloromethane (200 mL) was added with Grubbs' second-generation catalyst (Scholl, M.; Ding, S.; Lee, C.W.; Grubbs, R.H., Org. Lett, 1999, 1, 953-956.). After stirring at room temperature under argone for 3.5 h, the solution was concentrated under reduced pressure. Column chromatography (Si02, EtOAc) afforded 1143 as a white solid (60 mg, 60%), Mass calculated for formula C14H2ιN06 299.14, observed LCMS m/z 300.1 (M+H).
Part E:
To 1143 (60 mg, 0.2 mmol) in EtOAc (10 mL) was added 10% Pd/C (10 mg). The reaction mixture was allowed to stir at room temperature under H2 for 2 h, then concentrated. Column chromatography (Siθ2, EtOAc) afforded 1144 as a white solid (50 mg, 83%), Mass calculated for formula C14H23N06 301.15, observed LCMS m/z 302.1 (M+H).
Part F:
Compound 1144 (20 mg, 0.067 mmol) in MeOH (2 mL) was mixed with solid K2CO3 (20 mg, 0.14 mmol). After stirring at room temperature for 5 h, the reaction mixture was concentrated, and then taken up with EtOAc and water. It was acidified with 1 N HCI. The organic layer was separated, washed with brine, dried over Na2S04, and concentrated to afford 1145 as a white solid (12 mg, 63%), Mass calculated for formula Cι3H21N06 287.1 , observed LCMS m/z 288.1 (M+H).
Part G:
A mixture of 1145 (12 mg, 0.042 mmol), 978 (13.5 mg, 0.063 mmol), DIEA (0.018 mL, 0.1 mmol) and HATU (24 mg, 0.063 mmol) in DMF (1 mL) was stirred overnight at room temperature, and then diluted with ethyl acetate and water. The organic layer was washed with 1 N HCI, saturated NaHC03, and brine. It was dried over Na2S04, and concentrated. Purification by reverse phase prep-LC afforded 1146 as a white solid. HPLC-MS tR = 4.22 min (UV254 nm, 10 min), mass calculated for formula C2ιH22N204484.3, observed LCMS m/z 485.2 (M+H).
Example 34
Example 34A:
Figure imgf000553_0001
Part A
Step 1 : A mixture of 3-(aminomethyl)-1-N-(fe/f-butoxycarbonyl)-pyrrolidine (1147) ( 200 mg, 1.0 mmol), monomethyl phthalate (216 mg, 1.2 mmol), EDC (229 mg, 1.2 mmol), HOBt ( 162 mg, 1.2 mmol) and triethylamine (0.79 mL, 2.0 mmol) in DCM (4 mL) was stirred overnight. The reaction mixture was diluted with DCM and washed with 0.1 N HCI, water, saturated sodium bicarbonate solution, water, and brine, dried over sodium sulfate and concentrated. The residue was purified by column chromatography (Siθ2, 33% EtOAc/hexane) to afford a white solid (110 mg, 33%). HPLC-MS tR = 1.89 min (UV25 nrTl); mass calculated for formula Cι8H22N2θ4 330.2, observed LCMS m/z 353.2 (M+Na).
Step 2: Compound 1148 was prepared from the material from step 1 following the procedure as described in Example 27 Part A step 2.
Part B
Compound 1149 was prepared following the procedure as described in Example 27 Part A step 1. HPLC-MS tR = 1.96 min (UV254 nm); mass calculated for formula C20H17N3O2 331.1 , observed LCMS m/z 332.2 (M+H).
Part C
Compound 1149 (0.33 mmol) and hydrazine hydrate (65 μL, 1.34 mmol) in ethanol (3 mL) were refluxed for 2 hours. The reaction mixture was cooled and filtered. The filtrate was concentrated and 1150 was used without further purification.
Part D and E
Compounds 1151 and 1152 were prepared following the procedures described in Example 5 Part D and E. Data for 1151 : HPLC-MS tR = 1.78 min (UV254 nm); mass calculated for formula C28H30N4O6 518.2, observed LCMS m/z 519.2 (M+H). Data for 1152: HPLC-MS tR = 1.52 min (UV254 nm); mass calculated for formula C24H26N404 434.2, observed LCMS m/z 435.2 (M+H).
Example 34B:
Figure imgf000554_0001
Part A
To a solution of 1-benzyl-5-oxo-3-pyrrolidinecarboxamide (1153) (50 mg, 0.23 mmol) in THF (1 mL) was added 1 M solution of lithium aluminum hydride in THF (0.69 mL, 0.69 mmol). The reaction mixture was stirred at 50 °C for 3 hours, cooled, quenched with aqueous ammonium chloride solution. After addition of 1 N sodium hydroxide solution, the mixture was diluted with ethyl acetate, stirred for 20 minutes, and filtered through a pad of Celite. The filtrate was washed with 1 N sodium hydroxide solution and brine, dried over sodium sulfate and concentrated to give 1154 as an oil (14 mg, 32%). HPLC-MS tR = 0.22 min (UV254 nm); mass calculated for formula Cι2HιsN2 190.2, observed LCMS m/z 191.2 (M+H).
Part B and C
Compounds 1155 and 1156 were prepared following the procedures described in Example 5 Part D and E. Data for 1156: HPLC-MS tR = 0.91 min (UV254 nm); mass calculated for formula C24H29N304423.2, observed LCMS m/z 424.1 (M+H).
Example 35
Example 35A:
Figure imgf000555_0001
Part A and B and C:
Compounds 1158, 1159 and 1160 were prepared following the procedures described in Example 27 Part A, B and C. Data for 1158: HPLC-MS tR = 0.76 min (UV25 nm); mass calculated for formula C11H13N3 187.1 , observed LCMS m/z 188.1 (M+H). Data for 1159: HPLC-MS tR = 1.72 min (UV254 nm); mass calculated for formula C27H28N4θ6 504.2, observed LCMS m/z 505.1 (M+H). Data for 1160: HPLC-MS tR = 1.43 min (UV254 nm); mass calculated for formula C23H24N4O4 420.2, observed LCMS m/z 421.1 (M+H). Example 35B:
Figure imgf000556_0001
Part A
Step 1 : A mixture of azetidin-3-ylmethyl-carbamic acid tert-butyl ester (1157)
(20 mg, 0.107 mmol), 4-chloro-1-iodobenzene (38.4 mg, 0.161 mmol), copper iodide (4.0 mg, 0.021 mmol), L-proline (4.8 mg, 0.042 mmol) and potassium carbonate (29.6 mg, 0.214 mmol) in DMSO (2 mL) under Argon atmosphere was stirred at 80 °C overnight. The reaction mixture was diluted with EtOAc, washed with water and brine, dried over sodium sulfate and concentrated.
HPLC-MS tR = 2.13 min (UV254 nm); mass calculated for formula C15H2iCIN203
296.1 , observed LCMS m/z 297.2 (M+H).
Step 2: Compound 1161 was prepared from the material from step 1 following the procedure as described in Example 27 Part A step 2.
Part B and C
Compounds 1162 and 1163 were prepared following the procedures described in Example 5 Part D and E. Data for 1162: HPLC-MS tR = 1.93 min
(UV254 nm); mass calculated for formula C26H28CIN306 513.2, observed
LCMS m/z 514.2 (M+H). Data for 1163: HPLC-MS tR = 1.63 min (UV254 nm); mass calculated for formula C22H24CIN304429.2, observed LCMS m/z 430.1
(M+H). Example 36:
Figure imgf000557_0001
Compound 1164 was prepared using procedures described in Example 2. HPLC-MS tR = 1.88 min (UV254 nm); mass calculated for formula C-21H23CIIN3O4 543.0, observed LCMS m/z 544.0 (M+H).
Part A:
Compound 1165 was prepared from 1164 and styrenyl boronic acid using the coupling conditions described in Example 3 Part F. HPLC-MS tR = 2.11 min (UV254 nm); mass calculated for formula C29H30CIN3 519.2, observed LCMS m/z 520.0 (M+H).
Part B:
A mixture of 1165 (10 mg, 0.02 mmol) and palladium on carbon (10 wt%, 2 mg) in THF (2 mL) under hydrogen (atmospheric pressure) was stirred overnight. The reaction mixture was filtered through celite and concentrated. Purification by prep-LC afforded 1166 as an off-white solid (5 mg). HPLC-MS tR = 5.42 min (UV254 nm, 10 min); mass calculated for formula C29H32CIN3O4 521.2, observed LCMS m/z 522.2 (M+H).
Figure imgf000557_0002
Figure imgf000558_0001
Example 37: Alkyl Suzuki
Example 37A:
Figure imgf000558_0002
Part A:
Compound 1172 was prepared from 189 using the procedures described in
Example 2A Part B. HPLC-MS tR = 4.83 min (UV254 nm, 10 min); mass calculated for formula C19H2iBrCIN304S 501.0, observed LCMS m/z 502.0
(M+H).
Part B: Compound 1173 was prepared using the procedures described in Example 36 Part A. HPLC-MS tR = 5.37 min (UV254 nm, 10 min); mass calculated for formula C27H28CIN304S 525.2, observed LCMS m/z 526.2 (M+H).
Example 37B:
Figure imgf000559_0001
Part A:
Compound 1174 was prepared from 237 using the procedures described in Example 2. HPLC-MS tR = 4.76 min (UV254 nm, 10 min); Mass calculated for formula C2oH23BrCIN304S 515.0, observed LCMS m/z 516.0 (M+H).
Part B:
Compound 1175 was prepared using the procedures described in Example 36 Part A. HPLC-MS tR = 2.29 min (UV254 nm,); mass calculated for formula C28H30CIN3O4S 539.2, observed LCMS m/z 540.2 (M+H).
Example 37C:
Figure imgf000559_0002
Part A:
Compound 1176 was prepared using procedures described in Example 36
Part B.
HPLC-MS tR = 5.50 min (UV254 nm, 10 min); mass calculated for formula
C28H32CIN3O4S 541.2, observed LCMS m/z 542.2 (M+H).
Figure imgf000560_0001
Figure imgf000561_0002
Example 38: Isoindoline analogs
Figure imgf000561_0001
Step l
To 3-chloro-2-methylbenzoic acid 1189 (4.95 g, 0.0290 mol) dissolved in DMF (70 mL) was added cesium carbonate (14.18 g, 0.0435 mol) and methyl iodide (5.35 g, 2.3 mL, 0.0377 mol). The reaction mixture was stirred at room temperature for 20 h. The reaction mixture was concentrated, and EtOAc (200 mL) was added. The organic solution was washed with water (2 x 100 mL), dried (MgS04), filtered, and concentrated to give 4.43 g (83%) of the product 1190 as a yellow oil. MS for M-CL149
Figure imgf000561_0003
Figure imgf000562_0003
Figure imgf000562_0001
Step 2
To compound 1190 (4.42 g, 0.0239 mol) dissolved in CCI4 (100 mL) was added n-bromosuccinimide (5.11 g, 0.287 mol) and benzoyl peroxide (0.58 g, 0.00239 mol). The reaction mixture was heated at reflux for 16 h then cooled to room temperature. The solid was removed by filtration and washed with CH2CI2. The filtrate was concentrated to give 7.80 g of the product 1193 with succinimide as a yellow oil and solid mixture.
Figure imgf000562_0004
Figure imgf000562_0002
Step 3
Compound 1193 (6.31 g, 0.0239 mol) was dissolved in 7 N NH3 in MeOH (50 mL) and stirred at room temperature for 3 h then heated at reflux for 4 h. The reaction mixture was cooled to room temperature and concentrated. The residue was suspended in CH2CI2 (150 mL), and the solid was removed by filtration and washed with CH2CI2. The filtrate was concentrated, and the crude product was purified by silica gel flash chromatography (eluant: 5% MeOH-CH2CI2 to 10% MeOH-CH2Cl2) to give 3.68 g (92%) of the product 1196 as a white solid. MS for M+1 : 168.
Figure imgf000563_0002
Figure imgf000563_0001
Step 4
To compound 1196 (1.00 g, 5.97 mmol) suspended in dry THF (15 mL) was added borane (1 M in THF, 14.9 mL, 14.0 mmol). The reaction mixture was heated at reflux for 5 h then cooled to 0 °C. EtOH (15 mL) and potassium carbonate (2 g) were carefully added portionwise. The reaction mixture was heated at reflux for 16 h then cooled to 0 °C, and (tBOC)20 (1.95 g, 8.95 mmol) was added. The reaction mixture was stirred at room temperature for 3 h then concentrated. Water (30 mL) was added, and the aqueous solution was extracted with 3 x 30 mL of CH2CI2. The combined organic extracts were dried (MgS04), filtered, and concentrated. The crude product was purified by silica gel flash chromatography (eluant: 5% EtOAc- CH2CI2) to give 0.56 g (37%) of the product 1199 as a yellow oil. MS for M+1-tBu: 198.
Figure imgf000564_0001
The following intermediates were prepared according to the procedure of Example 11 B Part F.
Figure imgf000564_0002
The following intermediates were prepared according to the procedure of Example 2
Figure imgf000565_0001
The following compounds were prepared according to the procedure of Example 2
Figure imgf000565_0002
Figure imgf000566_0002
Example 39: Hydrazinoamides
Figure imgf000566_0001
Part A:
1-Phenyl-pyrazolidin-3-one (1215)(1.0 g, 6.16 mmol) in 15 mL of THF was added dropwise to the mixture of AICI3 (0.82 g, 6.16 mmol) and 12 mL of 1 M L1AIH4 (12 mmol) in THF at 0°C under argone. The reaction mixture was then allowed to stir at room temperature for 4 h. It was quenched with 10 mL of 1 N NH4CI solution, diluted with EtOAc and 15% NaOH aqueous solution. After filtration, the organic layer was separated, dried over Na2S04, and concentrated. Column chromatography (Si02, EtOAc/Hexane, 20:80) afforded 1216 as colorless oil.
Part B:
Compound 1217 was prepared according to the procedure described in
Example 2A Part A from 1216 (15 mg, 0.1 mmol).
Part C:
Compound 1217 was dissolved in 1 mL of MeOH, 0.5 mL of 7N NH3 in MeOH was added. After stirring at room temperature for 1 h, the reaction mixture was concentrated in vacuo. Purification by reverse phase prep-LC afforded compound 1218 as a white solid. HPLC-MS tR = 5.09 min (UV254 nm, 10 min),
Mass calculated for formula C25H26CIN304S 499.1 , observed LCMS m/z 500.0
(M+H).
Example 40: Piperidine sulfonamides
BocH
Figure imgf000567_0001
Figure imgf000567_0002
Part A:
Phenylsulfonyl chloride (212 mg, 1.2 mmol) was slowly added to a solution of 4-(Boc-aminomethyl)piperidine (977) (214 mg, 1 mmol) in 1.0 mL of pyridine at 0 °C. The reaction mixture was allowed to stir at room temperature overnight, and diluted with ethyl acetate and water. The organic layer was washed with 1 N citric acid, saturated NaHCθ3, and brine. It was dried over Na2S04, and concentrated, resulting in 1219 (320 mg, 90%). Mass calculated for formula C17H26N204S 354.2, observed LCMS m/z 355.2 (M+H), 299.2 (M- 55)
Part B:
To the solution of 1219 (200 mg, 0.54 mmol) in THF (5 mL) was added 2 mL of 4 N HCI in dioxane. The reaction mixture was stirred at room temperature for 1 h, and concentrated to afford 1220 (105 mg, 66 %).
Part C:
To 1220 (105 mg, 0.36 mmol) in DMF (2 mL) was added 230 (100 mg, 0.3 mmol), DIEA (750 μL, 0.6 mmol) and HATU (137 mg, 0.36 mmol). The reaction mixture was stirred overnight at room temperature, and diluted with ethyl acetate and water. The organic layer was washed with 1 N HCI, saturated NaHC03, and brine. It was dried over Na24, and concentrated, resulting in 1221.
Part D: -
Compound 1221 was dissolved in 4 mL of MeOH, 2 mL of 7N NH in MeOH was added. After stirring at room temperature for 1 h, the reaction mixture was concentrated in vacuo. Purification by reverse phase prep-LC afforded 1222 as a white solid. HPLC-MS tR =-3.59 min (UV254 nm, 10 min), Mass calculated for formula C24H29N306S 488.2, observed LCMS m/z 488.1 (M+H).
Figure imgf000568_0001
Example 41 : Heterocyclic isoindoline analogs Example 41A:
Figure imgf000569_0001
Part A:
Compound 1225 (70% overall yield) was prepared from 1 according to the procedures of Fukui, H. et. al. (Heterocycles 2002, 56, 257)
The following analogs were prepared according to this reference:
Figure imgf000569_0003
Figure imgf000569_0002
Part B:
Compound 1228 was prepared according to procedures described in Example 27A. HPLC-MS tR = 0.95 min (UV254nm); mass calculated for formula C13H16FN3 233.1 , observed LCMS m/z 234.2 (M+H). Compound 1229 was prepared according to the procedures described in Example 2A Part A. HPLC-MS tR = 1.57 min (UV254 Π ); mass calculated for formula C2 H3iFN6θ6 554.23, observed LCMS m/z 555.2 (M+H).
Figure imgf000570_0001
Part C:
Comppound 1036 was synthesized using the procedure found in Example 2A, Part B. Purification by reverse phase prep-LC afforded a white solid after lyophilization. HPLC-MS tR = 3.39 min (UV254 ΠM, 10 min); mass calculated for formula C23H27FN6O4 470.21 , observed LCMS m/z 471.2 (M+H).
Example 41 B:
Figure imgf000570_0002
1035
Part A:
Compound 1230 (2.18 g, 17.7 mmol) was dissolved in methylene chloride (35 mL) and cooled in an ice bath. Triethylamine (5 mL, 35.4 mmol) was added followed by p-toluenesulfonyl chloride (3.35 g, 17.7 mmol) in portions. The reaction mixture was stirred for 3 hours at room temperature and then quenched with water and extracted with methylene chloride. The combined organic layers were washed with 1 N HCI solution, bicarbonate solution and brine; dried over sodium sulfate and concentrated to provide the desired product 1231 (3.85 g), that was used without purification. HPLC-MS tR = 1.26 min (UV254nm); mass calculated for formula C11H15NO3S 241.08, observed LCMS m/z 242.2 (M+H).
Part B:
Compound 1231 (3.82 g, 15.78 mmol) was dissolved in methylene chloride (100 mL) and pyridinium chlorochromate (6.7 g, 31 mmol) was added followed by crushed molecular sieves (3.5 g). The reaction was stirred overnight and then diethylether (150 mL) was added and stirring was continued for an additional hour. The solids were filtered and the solvent was evaporated under reduced pressure. Purification by column chromatography (Siθ2, 50% ethyl acetate/hexanes) afforded a white solid 1232 (3.20 g, 84%). HPLC-MS tR = 1.43 min (UV254 ΠM); mass calculated for formula CnH13N03S 239.06, observed LCMS m/z 240.1 (M+H).
Part C:
Reactions were performed using procedures from Heterocycles, Vol. 41 , No.
7, 1995.
Part D:
Reaction was performed using the procedure found in Example 2A, Part A. 1234: HPLC-MS tR = 1.702 min (UV254 ΠM); mass calculated for formula C27H3oFN506S 571.19, observed LCMS m/z 572.2 (M+H).
Part E:
Reaction was performed using the procedure found in Example 2A, Part B.
Purification by reverse phase prep-LC afforded a white solid after lyophilization. 1035: HPLC-MS tR = 3.66 min (UV254 nM, 10 min); mass calculated for formula C23H26FN504S 487.2, observed LCMS m/z 488.2
(M+H).
Figure imgf000572_0005
Example 42:
Example 42A:
Figure imgf000572_0001
PartD PartE
Figure imgf000572_0003
PartF
Figure imgf000572_0004
Figure imgf000572_0002
1242 1243A.B Part A:
To a solution of dimethyl 2,3-O-isopropylidene-L-tartrate 1237 (10.9 g, 50 mmol) in dry THF (150 mL) and DMPU (25 mL) was added methyl iodide (4.7 L, 75.5 mmol). The reaction mixture was cooled to -78 °C and a 1 M solution in THF of LiN(TMS)2 (55mL, 55 mmol) was added via dropping funnel in 45 minutes period to the reaction mixture. THF reaction mixture was stirred at - 78 °C for 2 hours then warmed to -20 °C and quenched carefully with water (20 mL). The reaction mixture was poured into EtOAc (150 mL) and the organic layer was separated and washed with water (1 x 50 mL) followed by brine (1 x 50 mL). The organic layer was dried over MgS04, filtered and concentrated. Purification by column chromatography (Si02, 10% Et20/hexane followed by 15% Et2θ/hexane) afforded the desired compound 1238 as clear oil (9.7 g, 83%). LCMS m/z: 233.1 (M+H).
Part B:
A pH 8 buffer solution was prepared by titrating a solution of 0.1 M sodium phosphate (300 mL) with 1M HCI to final pH of 8. To a mixture of compound 1238 in pH 8 buffer solution was added esterase (41 units/mg,15 mg, 615 units). Added aq. 1 N NaOH (6.5 mL) slowly via syringe during a period of 3.5 hours to the reaction mixture to maintain the pH of the reaction mixture in the range of 7.9-8.2. The reaction mixture was extracted with Et20 (2 x 50 mL). The aqueous layer was acidified to pH 3 using 12N HCI then extracted with EtOAc (4 x 25 mL). The combined organic layers were dried over MgSθ4, filtered and concentrated in vacuo to afford the desired compound 1239 (1.36 g, 72%). LCMS m/z: 219.1 (M+H).
Part C:
To a solution of compound 916 (0.1 g, 0.535 mmol) in DMF (1.5 mL) was added compound 1239 (0.092 g, 0.5 mmol), triethyl amine (0.2 mL, 1.4 mmol) and HATU (0.365 g, 0.96 mmol). The reaction mixture was stirred for 18 hours and then diluted with CH2CI2 (5 mL) and washed with aq. 0.5N NaOH solution (3 mL). The organic layer was dried over MgS04 and concentrated. Purification by column chromatography (Si02, 30% EtOAc/hexane followed by 40% EtOAc/hexane) afforded the compound 1240 as a white foam (0.162 g, 67%). LCMS m/z 388.1 (M+H).
Part D:
To a solution of compound 1240 (0.175 g, 0.452 mmoi) in THF (2 mL) and MeOH (2 mL) at 0 °C was added aq. 1.0 M LiOH (0.91 mL, 0.91 mmol). The reaction mixture was warmed to room temperature and stirred for 1 hour. The reaction mixture was acidified with aq. 1 N HCI (0.95 mL) and diluted with brine (1.5 mL) and extracted with EtOAc (2 x 10 mL). The combined organic layers were dried over MgS0 and concentrated to afford compound 1241 as a white foam (0.182 g, 100%). LCMS m/z: 374.1 (M+H).
Part E:
To a solution of compound 1241 (0.179 g, 0.48 mmol) in DMF (3 mL) was added 2-phenylpyrrolidine hydrogen chloride salt (0.092 g, 0.5 mmol), triethyl amine (0.2 mL, 1.4 mmol) and HATU (0.365 g, 0.96 mmol). The reaction mixture was stirred for 4.5 hours and then diluted with EtOAc (10 mL) and washed with water (1.5 mL) followed by brine (1.5 mL). The organic layer was dried over MgS0 and concentrated. The residue was purified by prep, plate chromatography (Si02, 66%EtOAc/hexane). Two diastereomeric compounds 1242 were isolated. Diastereomer compound 1242A (0.067 g, 28%), LCMS m/z 503.1 (M+H) and diastereomer compound 1242B (0.095 g, 28%), LCMS m/z 503.1 (M+H).
Part F:
To a solution of compound 1242A (0.06 g, 0.119 mmol) in CH2CI2 (1 mL) was added TFA (4 mL) followed by water (0.5 mL). The reaction mixture was stirred at 80 °C temperature for 3 hours and then concentrated. The residue was purified by column chromatography (Si02, 5% MeOH/CH2CI2) to afford 1243A (0.012 g, 22%), LCMS m/z 463.1 (M+H).
To a solution of compound 1242B (0.088 g, 0.18 mmol) in CH2CI2 (1 mL) was added TFA (4 mL) followed by water (0.5 mL). The reaction mixture was stirred at 23 °C temperature for 18 hours and then concentrated. The residue was purified by column chromatography (S1O2, 5% MeOH/CH Cl2) to afford 1243B (0.05 g, 66%), LCMS m/z 463.1 (M+H).
Figure imgf000575_0003
Example 42B:
Figure imgf000575_0001
1244 1245
Figure imgf000575_0002
1247A and 1247B
Part A:
To a solution of compound 1239 (200 mg, 0.917 mmol) and 2- phenylpyrrolidine (177 mg, 0.96 mmol) in CH2CI2 (5 mL) at 23 °C was added DIEA (0.35 mL, 2.01 mmol) followed by PyBOP (0.367 g, 1.146 mmol) and the reaction mixture was stirred for 18 hours. The reaction mixture was concentrated and the residue was purified by column chromatography (Si02, 20% EtOAc/hexane followed by 30% EtOAc/hexane) to afford the desired compound 1244 as an oil (0.16 g, 50%). LCMS m/z: 348.1 (M+H).
Part B:
Compound 1244 was converted to compound 1245 using the procedure as described in Example 42A Part D. LCMS m/z: 374.1 (M+H).
Part C:
Compound 1245 was converted to compound 1246 using the procedure as described in Example 42A Part E. LCMS m/z: 503.1 (M+H).
Part D:
Compound 1246 was converted to 1247A and 1247B using the procedure as described in Example 42A Part F. Two diastereomeric compounds 1247 were isolated. Diastereomer 1247A (16% yield), LCMS m/z: 463.1 (M+H) and diastereomer 1247B (16% yield), LCMS m/z: 463.1 (M+H).
Example 43:
Example 43A:
Figure imgf000577_0001
Step l :
Compound 1248 was prepared according to the literature procedure: Shiao,
M.-J.; Liu, K.-H.; Lin, P.-Y Hetereocycles 1993, 36, 3, 507.
A solution of 1248 (2.20 g, 9.10 mmol), NBS (1.81 g, 10.20 mmol), 1 ,1 '- azobis(cyclohexanecarbonitrile) (0.113 g, 0.46 mmol) in CCU (40 mL) was stirred at 75 °C for 24 hours. The reaction was cooled to room temperature, and the mixture was diluted with CH2CI2 and sat. Na C03. The organic layer was removed, and the aqueous phase was extracted with CH2CI2 (4x). The combined organics were dried (Na2S04), filtered, and concentrated. The crude product mixture was used without further purification. A solution of the crude product mixture prepared above and sodium azide (0.75 g, 11.5 mmol) in a MeOH (15 mL)/DMF(15 mL)/DMSO (15 mL) solvent mixture was stirred at room temperature for 24 hours. The reaction was diluted with H20 and EtOAc. The organic layer was removed, and the aqueous phase was extracted with EtOAc (1x). The combined organics were washed with H20 (3x), brine (1x), dried (Na2S04), filtered, and concentrated. The resulting brown oil was purified by silica gel chromatography to furnish 1249 (1.2 g, 4.25 mmol, 47% yield over 2 steps). MS m/e: 283.1 (M+H).
Step 2:
To a solution of 1249 (1.2 g, 4.2 mmol) in THF (20 mL) at room temperature was added PPh (1.1g, 4.2 mmol). The reaction was stirred at room temperature for 1 hour at which time H20 (2 mL) was added. The resulting mixture was stirred for 24 hours. After concentration, the mixture was taken up in Et20 and 0.25 N HCI. The organic phase was removed, and the aqueous phase was basicified with sat. Na2C03 and extracted with EtOAc (4x). The combined organics were dried (Na24), filtered, and concentrated. The compound was purified by silica gel chromatography to furnish 1250 (0.51 g, 47% yield). MS m/e: 257.1 (M+H).
Step 3:
Compound 1251 was prepared with 1250 in a similar manner as previously stated in Example 12B.
Step 4:
Compound 1252 was prepared in a similar fashion from 1251 as previously stated in Example 12B. MS m/e: 518.1 (M+H).
Step 5:
To a solution of 1251 (0.20 g, 0.36 mmol) in THF (2 mL)/MeOH (2 mL)/ H20
(2 mL) was added LiOH (0.050 g, 1.2 mmol). The reaction was stirred at room temperature for 20 hours. The reaction was acidified to pH ~ 4 with 1 N
HCI and diluted with EtOAc. The organic layer was removed and the aqueous phase was extracted with EtOAc (3x). The combined organics were dried
(Na2Sθ4), filtered, and concentrated. The compound was used without further purification.
To the crude product mixture prepared above in CH2CI2 (2 mL) was added NMM (0.200 mL, 1.82 mmol), EDCI (0.100 g, 0.52 mmol), and HOBt (0.059 g, 0.38 mmol). This mixture was stirred for 10 minutes at which time NH4CI (0.100 g, 1.86 mmol) was added. After 72 hours of stirring, the reaction was quenched with H20 and diluted with EtOAc. The organic layer was removed, and the aqueous phase was extracted with EtOAc (3x). The combined organics were dried (Na2S04), filtered, and concentrated. The residue was purified by silica gel chromatography to yield 1253 (0.062 g, 61 % yield over 2 steps). MS m/e: 529.1 (M+H).
Step 6:
To a solution of 1253 (0.050 g, 0.095 mmol) in CH2CI2 (2 mL) cooled to 0 °C was added DIEA (0.050 mL, 0.28 mmol) followed by TFAA (0.030 mL, 0.22 mmol). The reaction was stirred at 0 °C for 3 hours and then quenched with 1 N NaOH. The organic layer was removed, and the aqueous phase was acidified with 1 N HCI to pH ~ 4. The aqueous phase was then extracted with EtOAc (3x). The combined organics were dried (Na2S04), filtered, and concentrated. The residue was purified by silica gel chromatography to furnish 1254 (0.042 g, 0.082 mmol, 88% yield). MS m/e: 511.1 (M+H).
Step 7:
Compound 1255 was prepared in a similar fashion from 1253 as previously stated in Example 12B. MS m/e: 489.1 (M+H).
Step 8:
Compound 1256 was prepared in a similar fashion from 1254 as previously stated in Example12A. MS m/e: 471.1 (M+H).
Example 43B:
Figure imgf000580_0001
Step 9:
A solution of aminomethylphenyl boronic acid hydrochloride (1257) (2.0 g, 10.7 mmol), 2,3-dibromothiophene (2.7 g, 11.0 mmol), and Pd(dppf)CI2 (0.51 g, 0.69 mmol) in CH3CN (40 mL)/1 M K2C03 (40 mL) was stirred at 80 °C for 3 hours. The reaction was cooled to room temperature and diluted with EtOAc. The organic layer was removed, and the aqueous phase was extracted with EtOAc (3x). The combined organics were dried (Na2S04), filtered, and concentrated. The brown oil was purified by silica gel chromatography to furnish 1258 (1.4 g, 5.3 mmol, 50% yield) as a brown oil. MS m/e: 268.1 (M+H).
Step 10:
Compound 1259 was prepared in a similar fashion from 1258 as previously stated in Example 12B. MS m/e: 569.1 (M+H).
Step 11 :
A solution of 1259 (0.059 g, 0.10 mmol), pyridine-4-yl boronic acid (0.017 g, 0.14 mmol), and Pd(dppf)CI2 (0.010 g, 0.014 mmol) in CH3CN (1 mL)/1 M K2CO3 (1 mL) was heated in SmithCreator microwave (2-5 mL vessel, 150 °C for 5 minutes). The liquid was concentrated, and the residue was purified by silica gel chromatography to furnish 1260 (0.057 g, 0.10 mmol, 99% yield). MS m/e: 568.1 (M+H).
Step 12:
Compound 1261 was prepared in a similar fashion from 1260 as previously stated in Example 12A MS m/e: 528.1 (M+H).
Example 44:
Part A Part B
-Br Br \ -Br
HO N H2N N=
Figure imgf000581_0001
Part A:
Isopropylmagnesium chloride (2.0 M solution in THF, 5 mL, 10 mmol) was added dropwise to the solution of 2-iodo-5-bromopyridine (1262) in THF (20 mL) at -40°C in an acetonitrile/dry ice bath. After stirring for 1 h at this temperature for 1 h, acetaldehyde (0.615 mL, 1 mmol) in 5 mL of THF was slowly added. The reaction mixture was allowed to stir at -40 °C for 0.5 h, then quenched with 1 N NH4CI (5 mL) solution, and concentrated under reduced pressure. The residue was dissolved in ethyl acetate and water. Organic layer was separated, washed with 1 N NH CI, brine, dried over Na2S04, and concentrated, affording 1263 as a colorless oil (1.95 g, 97%). 1H NMR (CDCI3, 300 MHz), δ 8.58 (d, J = 2.4 Hz, 1 H), 7.80 (dd, J = 8.6, 2.4 Hz, 1 H), 7.21 (d, J = 8.6Hz, 1 H), 4.87 (br q, J = 6.2 Hz, 1 H), 3.79 (br s, 1 H), 1.50 (d, J = 6.2 Hz, 3H). Mass calculated for formula C7H8BrNO 200.98, observed LCMS m/z 202.0 (M+H). Part B:
Diisopropyl azodicaboxylate (1.2 g, 6 mmol) in THF (5 mL) was added into a solution of 1263 (1.0 g, 5 mmol), phththalimide (0.88 g, 6 mmol) and triphenylphosphine (1.57 g, 6 mmol) in 20 mL of THF. After stirring overnight at room temperature, the reaction mixture was concentrated to dryness. It was then dissolved in 30 mL of ethanol, added with hydrazine monohydrate (5 mL), heated to reflux for 3 h. The reaction mixture was filtered and the filtrate was concentrated. The residue was dissolved with ethyl acetate and 1 N HCI. The aqueous phase was backed extracted with ethyl acetate. The organic extract was discarded. The aqueous phase was added with 1N NaOH to pH 12. It was extracted with EtOAc (30 mL x 3). The ester extract was combined, washed with brine, dried over Na2S0 ) and concentrated, resulting in 1264 (0.61 g, 61%). 1H NMR (CDCI3, 300 MHz), δ 8.58 (s, 1 H), 7.75 (dd, J = 8.1 , 2.0 Hz, 1 H), 7.23 (d, J = 8.5 Hz, 1 H), 4.15 (q, J = 6.5 Hz, 1 H), 1.43 (d, J = 6.5 Hz, 3H). Mass calculated for formula C7H9BrN2 199.99, observed LCMS m/z 201.0 (M+H).
Part C:
To 1264 (400 mg, 2 mmol) in DMF (5 mL) was added 563 (707 mg, 2 mmoi), and HATU (912 mg, 2.4 mmol). The reaction mixture was stirred overnight at room temperature, and diluted with ethyl acetate and water. The organic layer was washed with saturated NaHCθ3, and brine. It was dried over Na2S04, and concentrated, resulting in 1265 (600 mg, 56%). Mass calculated for formula C24H27BrCIN304 535.09, observed LCMS m/z 534.1 (M+H).
Part D:
Step 1 : To 1265 (120 mg, 0.22 mmol) in 1 mL of dioxane, was added with 2- cyanophenylboronic acid (49. mg, 0.33 mmol), potassium phosphate (142 mg,
0.66 mmol) and PdCI2(dppf) (7.2 mg, 0.0088 mmol, 4 mol%). The reaction mixture was stirred overnight at 80 °C under argon, then diluted with EtOAc and water, washed with saturated NaHCθ3, brine, dried over Na24, and concentrated. Step 2: The above residue was dissolved in 0.5 mL of TFA/H20 (80:20) and stirred at room temperature for 2 h. The reaction mixture was quenched with ACN/H20 (50:50) and concentrated in vacuo. Purification by reverse phase prep-LC afforded 1266 as a white solid. HPLC-MS tR = 4.04 min (UV254 nm, 10 min), Mass calculated for formula C28H2 CIN 0 518.17, observed LCMS m/z 519.1 (M+H).
Figure imgf000583_0001
Example 45:
Figure imgf000584_0001
Part A:
Compound 1257 (1.00 g, 5.34 mmol) was dissolved in THF (15 mL) and saturated sodium bicarbonate solution (20 mL). Di- -butyldicarbonate (1.28 g, 5.88 mmol) was added and the solution was stirred overnight at room temperature. The reaction mixture was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with bicarbonate solution and brine; dried over sodium sulfate and concentrated to provide a white solid 1270 (560 mg, 42%). HPLC-MS tR = 1.34 (UV254nm); mass calculated for formula Cι28BN04 251.13, observed LCMS m/z 274.1 (M+Na).
Part B:
Compound 1271 (2.0 g, 8.5 mmol) was dissolved in f-butanol (25 mL) and potassium f-butoxide (10g) was added and stirred at reflux for 24 hours. The solvents were removed under reduced pressure and the reaction was quenched with water and extracted with chloroform. The aqueous layer was acidified and extracted with chloroform. The combined organic layers were dried over sodium sulfate and concentrated to provide a yellow solid 1272 (1.10 g, 74%). HPLC-MS tR = 0.76 min (UV254 nm); mass calculated for formula C5H4BrNO 172.95, observed LCMS m/z 174.0 (M+H).
Part C:
Compound 1273 was prepared according to the procedure of Lui, H. et. al.
(Tet. Lett. 1995, 36, 8917). Data for 1273: HPLC-MS tR = 1.12 (UV254 nm); mass calculated for formula C7H8BrNO 200.98, observed LCMS m/z 202.0
(M+H).
Part D:
Compound 1273 (50 mg, 0.247 mmol) was dissolved in dioxane (5 mL) and compound 1270 (93 mg, 0.370 mmol), potassium phosphate (108 mg, 0.51 mmol), triphenylphosphine (10 mg), and Pd(dba)3 (5 mg) were added and stirred at 90 °C overnight. The reaction was filtered through celite and purified by column chromatography (EtOAc) to provide the desired product 1274 as a white solid (60 mg, 75%). HPLC-MS tR = 1.64 (UV254 nm); mass calculated for formula C19H24N2θ3 328.18, observed LCMS m/z 329.2 (M+H).
Part E:
Compound 1274 (60 mg, 0.183 mmol) was dissolved in methylene chloride (4 mL) and TFA (1 mL) was added. The reaction mixture was stirred for 1 hour and then the solvent was removed under reduced pressure. The residue was dissolved in DMF (5 mL) and compound 563 (63.5 mg, 0.183 mmol), HATU (90.5 mg, .0.2379 mmol), and DIEA (0.5 mL) were added and stirred overnight. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with 1 N HCI solution, bicarbonate solution and brine; dried over sodium sulfate and concentrated. Purification of the residue by column chromatography (EtOAc) provided compound 1275 as a white solid (87 mg, 85%). HPLC-MS tR = 1.81 min (UV254 nm); mass calculated for formula C31H34CIN3O5 563.22, observed LCMS m/z 564.1 (M+H). Part F:
Compound 1276 was synthesized using procedures similar to Example 1 , Part D. HPLC-MS tR = 1.48 min (UV254 nm); mass calculated for formula C28H3oCIN3θ5 523.19, observed LCMS m/z 524.2(M+H).
Example 46:
Figure imgf000586_0001
Part A:
Compound 1277 was prepared from 305 according to the procedures described in Example 4A Part A.
Part B:
To 1277 (150 mg, 0.44 mmol) in THF (5 mL) was added lithium hexamethyldisilazide (1.0 M, 0.66 mL, 0.66 mmol) at 0 °C. The reaction mixture was stirred for 10 minutes. To the reaction mixture was added methyl iodide (0.034 mL, 0.53 mmol). The reaction mixture was stirred overnight at room temperature. The reaction mixture was diluted with ethyl acetate and washed with 1.0 N citric acid, satureated sodium bicarbonate solution and brine, dried over sodium sulfate and concentrated. The residue was dissolved in DCM (5 ml) and treated with 4N HCI in dioxane (2 mL). The reaction was stirred at room temperature and concentrated. Compound 1278 was used without further purification.
Part C and D:
Compound 1280 was. obtained from 1278 and 230 using the procedures described in Example 4A Part D and E. Data for 1280: HPLC-MS tR = 5.05 min (UV254 nm, 0 min); mass calculated for formula C 5H25CIN204S 484.1 , observed LCMS m/z 485.0 (M+H).
Example 47: Heterocyclic Isoindolines
Example 47A:
Figure imgf000587_0001
Compound 2000 was prepared according to the procedures in US Patent 5,371 ,090.
Compound 2001 was prepared using procedures described in Example 2 and 27. HPLC-MS tR = min (UV254 nm); mass calculated for formula C21 H24FN307 449.2, observed LCMS m/z 450.1 (M+H).
Part A:
Compound 2002 was prepared using the coupling conditions described in Example 2. HPLC-MS tR = 1.60 min (UV254 nm); mass calculated for formula C28H30FN5O6 551.2, observed LCMS m/z 552.1 (M+H). Part B:
Compound 2002 (50 mg) was dissolved in methanol (2 mL). To this solution was added 7.0 M ammonia in methanol (2 mL). The reaction mixture was stirred for 1 hour at room temperature and concentrated. Purification by prep- LC and conversion to a hydrochloric salt afforded 2003 as an off-white solid (32 mg). HPLC-MS tR = 1.35 min (UV254 nm); mass calculated for formula C24H26FN504 467.2, observed LCMS m/z 468.1 (M+H).
Example 47B:
Figure imgf000588_0001
Compound 2004 was prepared according to the procedures in US Patent 5,371 ,090.
Compound 2005 was prepared using procedures described in Example 1 and 27.
Part A:
Compound 2006 was prepared using the coupling conditions described in Example 1. HPLC-MS tR = 1.40 min (UV254 nm); mass calculated for formula C27H30FN5O4 507.2, observed LCMS m/z 508.1 (M+H).
Part B:
Compound 2006 (50 mg) was deprotected using procedures described in Example 1. Purification by prep-LC and conversion to a hydrochloric salt afforded 2007 as an off-white solid (32 mg). HPLC-MS tR = 1.09 min (UV254 nm); mass calculated for formula C24H26FN504467.2, observed LCMS m/z 468.1 (M+H). Example 47C:
Figure imgf000589_0001
Part A:
Sulfonyl chloride 2008 (2.44 g, 9.8 mmol) was dissolved in dioxane (40 mL) and cooled in an ice bath. Ammonia gas was bubbled into the reaction mixture for 10 minutes. The reaction mixture was warmed to room temperature and filtered. The filtrate was concentrated. The crude product was recrystallized from ethyl acetate/hexanes to afford 2009 as an off-white solid (1.74 g). 1H NMR (400 MHz, DMSO-d6) δ 7.09 (s, 2H), 6.74 (s, 1 H), 3.80 (s, 3H), 2.57 (s, 3H), 2.48 (s, 3H), 2.08 (s, 3H); HPLC-MS tR = 1.43 min (UV25 nm); mass calculated for formula C10H15NO3S 229.1 , observed LCMS m/z 230.1 (M+H).
Part B:
To sodium hydride (95%, 131 mg, 5.19 mmol) in DMF (10 mL) was added sulfonamide 2009 (596 mg, 2.6 mmol). The reaction mixture was heated to 70 °C and stirred for 45 minutes. To this mixture was added 2,3- bis(chloromethyl)pyrazine (2010) (Yoshiizumi, K. et. al. Bioorg. Med. Chem. 2003, 11, 433) (448 mg, 2.53 mmol) in DMF (3 mL). The reaction mixture was heated at 70 °C overnight. The reaction mixture was cooled and poured into water. The aqueous layer was salted and extracted with, chloroform. The combined organics were washed with water, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 30% ethyl acetate/hexanes) afforded recovered sulfonamide 2009 (218 mg) and 2011 (245 mg). HPLC-MS tR = 1.82 min (UV25 nm); mass calculated for formula C16H19N303S 333.1 , observed LCMS m/z 334.1 (M+H). Part C:
A mixture of sulfonamide 2011 (245 mg, 0.73 mmol), anhydrous methanesulfonic acid (3 mL) and 1 :9 thioanisole:trifluoroacetic acid (3 mL) was stirred for 3 hours at room temperature. The reaction mixture was poured over ice and treated with 50% sodium hydroxide (10 mL). The aqueous layer was salted and extracted with chloroform. The combined organics were dried over sodium sulfate and concentrated. The residue was dissolved in 1.0 M HCI (5 mL) and extracted with ether. The aqueous layer was lyophilized to afford 2012 (105 mg) as a brown solid. 1H NMR (400 MHz, DMSO-de) δ 10.27 (s, NH), 8.58 (s, 2H), 4.59 (m, 4H).
Example 47D:
Figure imgf000590_0001
Part A:
Compound 2014 was prepared according to the procedure in Helv. Chim.
Acta. 1986, 905-907.
Part B:
Acetyl chloride (20 mL) was added dropwise to methanol (130 mL) cooled in an ice bath. This solution was then added to compound 2014 (5.00 g, 31.8 mmol) and stirred overnight at reflux. The solvent was removed under reduced pressure and the residue was partitioned between ethyl acetate and 1 N NaOH. The organic layer was washed with brine, dried over sodium sulfate, and concentrated to provide 2015 (5.0 g). H NMR (400 MHz, CDCI3) δ 3.72 (s, 3H), 3.69 (s, 3H), 2.77 (t, 2H), 2.5 (t, 2H), 1.32 (s, 6H).
Part C: Compound 2015 (5.00 g, 24 mmol) was dissolved in THF (60 mL) and saturated sodium bicarbonate (60 mL) and cooled in an ice bath. Methyl chloroformate (2.93 g, 31.2 mmol) was added dropwise and the reaction was stirred at room temperature overnight. The reaction mixture was partitioned between ethyl acetate and water. The organic layer was washed with 1 N HCI, saturated sodium bicarbonate, brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 20% ethyl acetate/hexanes) afforded 2016 (5.5 g). 1H NMR (400 MHz, CDCI3) δ 3.72- 3.68 (m, 9H), 3.65-3.6 (t, 2H), 2.66 (t, 2H), 1.5 (s, 6H).
Part D:
Compound 2016 (1.66 g, 6.36 mmol) was dissolved in THF (20 mL) and cooled on an ice bath. Potassium hydride (35% in oil, 1.12 g, 9.54 mmol) was added in portions and stirring was continued for 2 hours at room temperature. The reaction was slowly quenched with water, acidified to pH 3, and extracted with ethyl acetate. The combined organic layers were dried over sodium sulfate and concentrated (1.5 g). A portion of the residue (350 mg) was dissolved in MeOH (5 mL) and 6N HCI (5 mL) and stirred at reflux for 3 hours and room temperature overnight. The solvent was evaporated under reduced pressure and partitioned between ethyl acetate and water. The organic layer was washed with water, dried over sodium sulfate, and concentrated to yield 2017 (150 mg). 1H NMR (400 MHz, CDCI3) δ 3.72-3.55 (m, 5H), 2.5 (t, 2H), 1.4-1.25 (m, 6H).
Part E:
According to a modification of a procedure by Fukui, H. et al. (Heterocycles 2002, 56, 257-264) a mixture of ketone 2017 (200 mg, 1.17 mmol) and N,N- dimethylformamide dimethyl acetal (3 mL) was heated at 100 °C for 2h, and then concentrated to give enamino-ketone 2018 (244 mg, 92%) as a yellow solid. 1H NMR (400 MHz, CDCI3) δ 7.32-7.31 (m, 1 H), 4.54-4.51 (d, 2H), 3.78- 3.22 (d, 3H), 3.12 (s, 6H), 1.48 (s, 3H), 1.40 (s, 3H).
Part F: According to a modification of a procedure by Fukui, H. et al. (Heterocycles 2002, 56, 257-264) a mixture of enamino-ketone 2018 (690 mg, 3.05 mmol) and formamidine acetate (3.17 g, 30.5 mmol) in ethanol (15 mL) was heated at reflux for 3 days, and then concentrated. The residue was partitioned between dichloromethane and water, and extracted with dichloromethane. The combined organic extracts were concentrated to give an oil which was chromatographed (Si02, 50%-80% ethyl acetate/hexane) to give pyrimidine 2019 (334 mg, 53%). 1H NMR (400 MHz, CDCI3) δ 9.15 (s, 1 H), 8.67 (s, 1 H, broad), 4.76-4.71 (d, 2H), 3.84-3.77 (d, 3H), 1.75 (s, 3H), 1.67 (s, 3H).
Part G:
A mixture of compound 2019 (235 mg, 1.13 mmol), powder potassium hydroxide (900 mg, 16 mmol) and hydrazine monohydrate (1.36 mL, 28 mmol) in ethylene glycol (5 mL) was heated at 100 °C overnight. The reaction mixture was cooled to rt, poured into brine and extracted several times with dichloromethane. The combined organic extracts were washed with brine and concentrated to give compound 2020 (121 mg, 72 %). 1H NMR (400 MHz, CDCI3) δ 9.06 (s, 1 H), 8.58 (s, 1 H), 4.24 (s, 2H), 1.47 (s, 6H). HPLC-MS tR = 0.2 min (UV254 nm); mass calculated for formula C8H11 N3 149.1 , observed LCMS m/z 150.1 (M+H).
Example 47E:
Figure imgf000592_0001
Part D Λ--^ Part E ^N NH
2026 2027
Part A:
To a mixture of methyl crotonate (2022) (2.0 g, 20 mmol) and methyl glycine hydrochloride (2021) (5.5 g, 44 mmol) in methanol (27 mL) was added triethylamine (6.4 mL, 46 mmol). The mixture was stirred at room temperature for 2 days and filtered to remove the white precipitate. The filtrate was concentrated and the residue was dissolved in ethyl acetate, washed with sodium bicarbonate solution, water and brine, dried over sodium sulfate and concentrated to afford 2023 as a light yellow oil (2.4 g, 64%). 1H NMR (400 MHz, CDCI3) δ 3.74 (s, 3H), 3.69 (s, 3H), 3.46 (d, J = 3.2 Hz, 2H), 3.14 (m, 1 H), 2.45 (m, 2H), 1.15 (d, J = 7.2 Hz, 3H).
Part B:
Compound 2023 (2.42 g, 13 mmol) was dissolved in THF (20 mL) and saturated sodium bicarbonate (20 mL) and cooled in an ice bath. Methyl chloroformate (1.2 mL, 15 mmol) was added dropwise and the reaction was stirred at room temperature overnight. The reaction mixture was partitioned between ethyl acetate and water. The organic layer was washed with 1 N HCI, saturated sodium bicarbonate and brine, dried over sodium sulfate and concentrated to afford 2024 (2.9 g).
Part C:
Compound 2024 (1.30 g, 5.26 mmol) was dissolved in THF (20 mL) and cooled on an ice bath. Potassium hydride (35% in oil, 1.12 g, 9.54 mmol) was added in portions and the mixture was stirred at room temperature overnight. The reaction was slowly quenched with water, acidified to pH 3, and extracted with ethyl acetate. The combined organic layers were dried over sodium sulfate and concentrated. Purification by column chromatography (Siθ2, 25% ethyl acetate/hexane) afforded an oil (1.27 g). This oil (1.27 g) was dissolved in MeOH (5 mL) and 6N HCI (5 mL) and stirred at reflux overnight. The mixture was extracted with ethyl acetate and the combined organic layers were washed with sodium bicarbonate solution, water and brine, dried over sodium sulfate and concentrated to afford 2025 (663 mg). 1H NMR (400 MHz, CDCI3) δ 4.5 (bs, 1 H), 3.95 (m, 1 H), 3.78 (s, 3H), 3.70 (m, 1 H), 2.73 (m, 1 H), 2.25 (m, 1 H), 1.28 (m, 3H).
Part D:
Compound 2026 was prepared from the material from part C according to procedures described in Example 47D Part E and Part F. 1H NMR (400 MHz, CDCI3) δ 9.13 (s, 1 H), 8.61 (m, 1 H), 5.24 (m, 1 H), 4.88 - 4.64 (m, 2H), 3.82 (m, 3H), 1.59 (m, 3H).
Part E:
A mixture of compound 2026 (300 mg, 1.45 mmol), powder potassium hydroxide (600 mg, 10.7 mmol) and hydrazine monohydrate (1.0 mL, 21 mmol) in ethylene glycol (5 mL) was heated at 100 °C overnight. The reaction mixture was cooled to rt, poured into brine and extracted several times with dichloromethane. The combined organic extracts were washed with brine and concentrated to give compound 2027 (105 mg, 54 %).
Example 47F:
Figure imgf000594_0001
Part A:
To a solution of compound 2028 (0.29 g, 0.63 mmol) in CH2CI2 at 0 °C was added 70% m-CPBA (0.31 g, 1.26 mmol) and the reaction mixture was warmed to room temperature and stirred for 1 hour. Diluted with CH2CI2 (5 mL) and washed with a solution of water (6 mL) and concentrated NH4OH (0.5 mL). Aqueous layer was extracted with CH2CI2 (3 x 10 mL) and 5% MeOH/CH2CI2 (2 x 5 mL). The organic layer was dried over MgS0 , filtered and concentrated. Purification by column chromatography (Si02, 5% MeOH/CH2CI2) afforded the desired product with some impurity. Dissolved the impure product in EtOAc (15 mL) and washed with 10% NH4OH (5 mL). The aqueous layer was extracted with CH2CI2 (3 x 5 mL). The combined organic layers (EtOAc and CH2CI2) were dried over MgS04, filtered and concentrated to give the desired compound 2029 (0.25 g, 92%). Part B:
Compound 2030 was obtained from compound 2029 using the TFA deprotection procedure as described in Example 1.
The following compounds were prepared using previously described procedures.
Figure imgf000595_0001
Figure imgf000596_0001
Figure imgf000597_0001
Figure imgf000598_0002
Example 48: Heterocyclic tetrahydroisoquinoline analogs Example 48A:
Figure imgf000598_0001
Part A:
Compound 2059 was synthesized according to the literature procedures.
(Dow, R.L.; Schneider, S.R. J. Heterocyclic Chem. 2001, 38, 535).
Example 48B: CO m- CO ^NH 2060 2061
Part A:
Compound 2061 was synthesized according to the procedures of US patent
5,037,834.
Example 48C:
Figure imgf000599_0001
Part A:
According to a modification of a procedure by Fukui, H. et al. (Heterocycles 2002, 56, 257-264) a mixture of ketone 2062 (2.5 g, 12.5 mmol) and N,N- dimethylformamide dimethyl acetal (21 mL) was heated at 100 °C for 2h. The mixture was concentrated to a residue which was passed through a short silica gel column (9:1 :90 methanol:triethylamine:dichloromethane) to give enamino-ketone 2063 (3.1 , 97%) as a viscous brown oil. HPLC-MS tR = 1.34 min (UV254 nm); mass calculated for formula C13H22N203 254.1 , observed LCMS m/z 255.1 (M+H).
Part B:
According to a modification of a procedure by Fukui, H. et al. (Heterocycles 2002, 56, 257-264) a mixture of enamino-ketone 2063 (732 mg, 2.88 mmol) and hydrazine monohydrate (0.28 mL, 5.76 mmol) in ethanol (8 mL) was heated at reflux overnight, and then concentrated. The residue was partitioned between dichloromethane and water, and extracted with dichloromethane. The combined organic extracts were concentrated to give an oil which was chromatographed (Si02, 80% ethyl acetate/hexane) to give compound 2064 as an oil (major isomer, 300 mg, 46%). HPLC-MS tR = 1.39 min (UV254 nm); mass calculated for formula C11 H17N302 223.1 , observed LCMS m/z 224.2 (M+H).
Part C: Compound 2064 (300 mg, 1.34 mmol) was deprotected according to a procedure described by Fukui, H. et al. (Heterocycles 2002, 56, 257-264) to give compound 2065 (57 mg, 22 %). 1H NMR (400 MHz, DMSO-d6) δ 10.9 (s, 1 H, broad), 9.4 (s, 1 H, broad), 7.57 (s, 1 H), 4.19 (t, 2H), 3.30-3.26 (m, 2H), 2.78 (t, 2H). HPLC-MS tR = 0.2 min (UV254 nm); mass calculated for formula C6H9N3 123.1 , observed LCMS m/z 124.2 (M+H).
Example 48D:
Figure imgf000600_0001
Part A:
A mixture of ketone 2066 (4.0 g, 20 mmol), morpholine (7.5 mL, 85 mmol) and p-toluenesulfonic acid monohydrate (160 mg, 0.84 mmol) in benzene (30 mL) was heated at reflux for 3 days using a Dean-Stark trap. The mixture was then washed with saturated sodium bicarbonate solution and concentrated to give 2067 as a yellow oil (4.36 g, 65%). 1H NMR (400 MHz, CDCI3) δ 4.58 (s, 1 H, broad), 3.96-3.95 (m, 2H), 3.77-3.73 (m, 4H), 3.55 (t, 2H), 2.83-2.80 (m, 4H), 2.18 (t, 2H), 1.49 (s, 9H).
Part B:
According to a modification of a procedure by Gϋndisch, D. et. al. (Bioorg. Med. Chem. 2002, 10, 1-9) a mixture of enamine 2067 (2.27 g, 8.48 mmol) and 1 ,3,5-triazine (688 mg, 8.48 mmol) in 1 ,4-dioxane (10 mL) was heated at 100 °C in a sealed tube overnight. The reaction mixture was concentrated and chromatographed (Si0 , 50-65% ethyl acetate/hexane) to give pyrimidine 2068 as a yellow oil (665 mg, 35%). 1H NMR (400 MHz, CDCI3) δ 9.01 (s, 1H), 8.49 (s, 1H), 4.64 (s, 2H), 3.80 (t, 2H), 3.00 (t, 2H), 1.54 (s, 9H). HPLC-MS tR = 1.32 min (UV254 nm); mass calculated for formula C12H17N302 235.1 , observed LCMS m/z 236.1 (M+H).
Part C: Compound 2068 (100 mg) was deprotected using procedures described in Example 48C to give amine 2069 as a yellow solid (68 mg, 78%). 1H NMR (400 MHz, DMSO-de) δ 9.67 (s, 2H, broad), 9.01 (s, 1 H), 8.67 (s, 1 H), 4.33 (t, 2H), 3.50-3.45 (m, 2H), 3.09 (t, 2H). HPLC-MS tR = 0.18 min (UV25 nm); mass calculated for formula C7H9N3 135.1 , observed LCMS m/z 136.2 (M+H).
Example 48E: N- OH
2070
Compound 2070 was prepared from methyl vinyl ketone in 6 steps following procedures described in US Patent 5,037,834/1991.
Example 48F:
Figure imgf000601_0001
2071 2072
Compound 2072 was prepared from 2-aminopyrazine (2071) in 2 steps following procedures described by Sablayrolles, C. et. al. (J. Med. Chem. 1984, 27, 206-212) and Bonnet, P. A. et. al. (J. Chem. Res. Miniprint FR 1984, 2, 0468-0480).
The following compounds were prepared using previously described procedures.
Figure imgf000601_0002
Figure imgf000602_0001
Example 49:
Example 49A: BOG N Part Part B Part C o —A rBoc .NfBoc .NH
2085 2086 2087 2088
Part A:
Compound 2086 was prepared according to the procedure in Org. Lett. 2002,
4, 24, 4353-4356.
Part B:
Compound 2087 was prepared according to the procedure in Org. Lett. 2002, 4, 24, 4353-4356. 1H NMR (400 MHz, CDCI3) δ 7.05 (t, 2H), 5.1 (bs, 2H), 2.15 (m, 2H), 1.4 (s, 9H), 1.3 (m, 2H).
Part C:
Compound 2087 (80 mg, 0.30 mmol) was dissolved in 4 M HCI in dioxane (2 mL) and stirred at room temperature for 1 hr. The reaction was diluted with diethyl ether (5 mL) and the resulting solid was filtered to provide 2088 as an HCI salt (45 mg).
Example 49B:
Figure imgf000603_0001
Part A:
Compound 2090 was prepared according to the procedure in Bioorg. Med.
Chem. 2002, 10, 5, 1197-1206.
Part B:
Compound 2091 was prepared according to the procedure in Bioorg. Med. Chem. 2002, 10, 5, 1197-1206. 1H NMR (400 MHz, CDC) 3) δ 9.00 (s, 1 H), 8.40 (s, 1 H), 5.10 (bs, 1 H), 4.70 (bs, 1 H), 4.10 (m, 2H), 3.40 (bs, 1 H), 2.50 (s, 1 H), 2.40-2.30 (m, 2H), 1.80 (m, 2H), 1.75 (m, 2H), 1.25 (m, 3H).
Part C:
Compound 2091 (350 mg, 1.5 mmol) was dissolved in ethylene glycol (4 mL) and KOH (0.5 g) and hydrazine monohydrate (1 mL) were added and stirred at 100 °C for 12 hours. The reaction mixture was poured into brine and extracted with methylene chloride. The combined organic layers were washed with water, dried over sodium sulfate and concentrated to afford 2092 (150 mg). 1H NMR (400 MHz, CDCI3) δ 9.00 (s, 1 H), 8.40 (s, 1 H), 4.30 (d, 1 H), 4.00 (t, 1 H), 3.20 (m, 1 H), 2.80 (m, 1 H), 2.20-2.10 (m, 2H), 2.00-1.90 (m, 2H), 1.80-1.60 (m, 2H).
The following compounds were prepared using previously described procedures.
Figure imgf000604_0001
Figure imgf000605_0002
Example 50: Piperidine-aryl compounds Example 50A:
Figure imgf000605_0001
Part A:
A mixture of 2-bromo-5-fluorophenoI (556) (0.111 mL, 1 mmol), 2,2,2- trifluoroethyl trifluoromethanesulfonate (2100) (279 mg, 1.2 mmol) and cesium carbonate (358 mg, 1.1 mmol) in NMP (5 mL) was stirred at room temperature overnight. The reaction mixture was poured into water and extracted with ethyl acetate. The combined organic layers were washed with water (2x) and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, hexanes to 2% ethyl acetate/hexanes) afforded 2101 (232 mg) as an oil. 1H NMR (400 MHz, CDCI3) δ 7.53 (m, 1 H), 6.70 (m, 2H), 4.40 (q, 2H, J = 8.0 Hz).
Part B: Compound 2102 was prepared according to the procedures in Example 27B Part A. HPLC-MS tR = 2.10 min (UV254 nm); mass calculated for formula C19H26F4N203 406.2, observed LCMS m/z 407.2 (M+H).
Part C:
Compound 2102 (8 mg, 0.02 mmol) was dissolved in 3:1 DCM:TFA (4 mL) and stirred for 1 hour at room temperature. The solvent was removed in vacuo. The residue was dissolved in ether and treated with 1.0 M HCI in ether (1 mL). The solvents were concentrated to afford 2103 as a white solid (8 mg).
Example 50B:
Figure imgf000606_0001
Part A:
A mixture of 2-fluorophenol (2104) (1.0 mL, 10.7 mmol), ethyl iodide (1.05 mL, 13 mmol) and potassium carbonate (1.66 g, 12 mmol) in acetone (20 mL) was stirred at room temperature overnight. Additional ethyl iodide (0.24 mL, 3 mmol) was added to the reaction and the mixture was stirred for 24 hours. The mixture was filtered and concentrated. The residue was partitioned between water and ethyl acetate. The layers were separated. The organic layer was washed with water and brine, dried over sodium sulfate and concentrated to afford 2105 (1.23 g) as an oil. 1H NMR (400 MHz, CDCI3) δ 7.07 (m, 2H), 6.96 (m, 1 H), 6.87 (m, 1 H), 4.12 (q, 2H, J = 7.1 Hz), 1.48 (t, 3H, J = 7.1 Hz).
Part B:
To arene 2105 (1.23 g, 8.8 mmol) and TMEDA (1.32 mL, 8.8 mmol) in THF (20 mL) at -78 °C under argon was added s-Buϋ (1.4 M, 6.3 mL, 8.8 mmol). The reaction mixture was stirred for 2 hours at -78 °C. To the reaction mixture was added iodine (2.23 g, 8.8 mmol) in THF (10 mL) at -78 °C. The reaction was stirred for 10 minutes then warmed to 0 °C. The reaction mixture was poured into water and extracted with diethyl ether. The combined organics were washed with water (2x), 5% sodium hydrogensulfite and brine, dried over sodium sulfate and concentrated. The crude product 2106 (1.70 g) was a mixture of 75:25 productstarting material by 1H NMR. It was used without further prurification. 1H NMR (400 MHz, CDCI3) δ 7.28 (m, 1 H), 6.94 (m, 1 H), 6.81 (m, 1 H), 4.11 (q, 2H, J = 7.1 Hz), 1.47 (t, 3H, J = 7.1 Hz).
Part C:
Compound 2107 was prepared according to the procedures in Example 27B Part A. HPLC-MS tR = 2.15 min (UV254 nm); mass calculated for formula C19H29FN203 352.2, observed LCMS m/z 353.2 (M+H).
Part D:
Compound 2107 (276 mg, 0.78 mmol) was dissolved in 3:1 DCM:TFA (4 mL) and stirred for 1 hour at room temperature. The solvent was removed in vacuo. The residue was dissolved in ether and treated with 1.0 M HCI in ether (1 mL). The solvents were concentrated to afford 2108 as a white solid (241 mg). 1H NMR (400 MHz, DMSO-d6) δ 7.89 (bs, NH), 6.97 (m, 1 H), 6.75 (m, 1 H), 6.63 (m, 1 H), 4.04 (q, 2H, J = 6.9 Hz), 3.33 (m, 2H), 2.76 (m, 2H), 2.62 (m, 2H), 1.80 (m, 2H), 1.70 (m, 1 H), 1.38 (m, 2H), 1.32 (t, 3H, J = 7.1 Hz).
Example 50C:
Figure imgf000607_0001
Compound 2109 was prepared using procedures described in Example 27. 1H NMR (400 MHz, CDCI3) δ 7.00 - 6.80 (m, 4H), 4.64 (m, 1 H), 3.55 (m, 2H), 3.09 (m, 2H), 2.65 (m, 2H), 1.82 (m, 2H), 1.60-1.50 (m, 2H), 1.49 (s, 9H), 1.42 (m, 1 H). Part A:
To compound 2109 (100 mg, 0.32 mmol) and pyridine (0.026 mL, 0.32 mmol) in DMF (2 mL) was added NBS (115 mg, 0.64 mmol). The mixture was stirred at room temperature for 2 hours and diluted with ethyl acetate. The mixture was washed with sodium carbonate solution, water and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 5% ethyl acetate/hexane) afforded 2110 as a white solid (88 mg). 1H NMR (400 MHz, CDCl3) δ 7.3 (m, 1 H), 7.0 (m, 2H), 4.65 (bs, 1 H), 3.28 (m, 2H), 3.10 (m, 2H), 2.61 (m, 2H), 1.8 (m, 2H), 1.65 - 1.4 (m, 12 H).
Part B:
To 2110 (21 mg, 0.055 mmol) in dichloromethane (2 mL) at 0 °C was added TFA (1 mL). The mixture was stirred at room temperature for 1 hour. The mixture was quenched with acetonitrile and concentrated. Compound 2111 was used without further purification.
Example 50D:
Figure imgf000608_0001
Part A:
Compound 2113 was prepared from 1-N-(f-butoxycarbonyl)-4-cyano- piperidine (2112) according to a procedure described in J. Org. Chem. 1992,
57, 4521-4527.
Part B:
Step 1 : To 2113 (100 mg, 0.41 mmol) in THF (2 mL) were added benzyl chloroformate (0.089 mL, 0.619 mmol) and potassium carbonate (114 mg, 0.82 mmol). The mixture was stirred at room temperature overnight, diluted with ethyl acetate, washed with water, brine, dried over sodium sulfate and concentrated. The product was purified by column chromatography (Si02> 10% ethyl acetate/hexane to 25% ethyl acetate/hexane) to give a pale yellow oil (80 mg). HPLC-MS tR = 2.14 min (UV254 nm); Mass calculated for C21 H32N204 376.5, observed LSMS m/z 399.2 (M+Na).
Step 2: The material from step 1 (80 mg) was stirred in 4 N HCI in 1 ,4-dioxane (2mL) for 1 hour. The solvent was removed in vacuum and 2114 was used without further purification.
Part C:
A mixture of 2114, 2,5-difluorobenzonitrile (32.5 mg, 0.234 mmol) and DIEA (0.113 mL, 0.637 mmol) in NMP (2 mL) under argon atmosphere was stirred overnight at 100 °C. The reaction mixture was diluted with ethyl acetate, washed with water and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 25% ethyl acetate/hexane) afforded 2115 as a yellow oil (26 mg). 1H NMR (400 MHz, CDCl3) δ 7.3 - 7.2 (m, 6H), 7.2 (m, 1 H), 7.0 (m, 1 H), 5.05 (s, 2H), 4.7 (m, 1 H), 3.52 (m, 2H), 2.77 (m, 2H), 2.15 (m, 1 H), 1.8 (m, 2H), 1.63 (m, 2H), 1.32 (s, 6H).
Part D:
A mixture of 2115 (26 mg) and 10% palladium on carbon (5 mg) in ethanol (4 mL) under hydrogen atmosphere was stirred for 3 hours. The mixture was filtered through a pad of celite and the filtrate was concentrated to give 2116 as a yellow residue (20 mg), which was used without further purification.
Example 50E:
Bor CO2H
Figure imgf000609_0001
ii. NaNa ζ YHCOOallyl HCI/Et2q fVV'NHCOOallyl
///. Allyl alcohol Boc' ^ part E H' ^
Part D 2120 -HCI 2121
Part A:
Methyl (triphenylphosphoranylidene)acetate (104.5 g, 0.31 mmol) was added to a solution of Λ/-Boc-piperidinone (2066) (49.79 g, 0.25 mol) in toluene (625 mL). The resulting reaction mixture was heated to reflux and stirred for 17 h. The reaction mixture was then cooled to room temperature and concentrated under vacuum. The resulting residue was then pre-adsorbed on silica gel and purified by eluting it through a plug of silica gel with 50% ethyl acetate/hexanes, to yield unsaturated ester 2117 (62.16 g, 0.243 mol) as a white solid.
Part B:
Potassium ferf-butoxide (450 g, 0.41 mol) was added to a solution of trimethylsulfoxonium iodide (90.0 g, 0.41 mol) in DMSO (700 mL), in one portion. The mixture was stirred at room temperature for 3 h. The unsaturated ester 2117 (59.64 g, 0.23 mol) was dissolved in DMSO (0.26 L) and added to the reaction mixture. The reaction mixture was stirred for 20 h at room temperature and then added to brine (1 L). Saturated aqueous NH4CI was then added to the reaction mixture in order to adjust the pH to approximately 7. The reaction mixture was then extracted several times with ether, the ether extracts combined, washed with water and brine, dried with anhydrous MgS0 , filtered, and concentrated under vacuum to yield ester 2118 (53.5 g, 0.20 mol) as an oil.
Part C:
An aqueous LiOH solution (2N, 200 mL) was added to a solution of the ester 2118 (53.5 g, 0.20 mol) in THF (200 mL). The mixture was then stirred at room temperature for 17 h, diluted with water (750 mL) and washed with ether. The ether phase was discarded, and the aqueous phase acidified to a pH of 3-4 with 6N HCI. The acidified aqueous phase was then extracted with ether several times. The ether washes were combined, washed with water and brine, dried with anhydrous MgSθ4, filtered, and concentrated under vacuum to provide carboxylic acid 2119 (49.25 g, 0.19 mol) as a white solid.
Part D:
Triethylamine (8.7 g, 0.086 mol) followed by ethyl chloroformate (9.4 g, 0.086 mol) was added to a solution of carboxylic acid 2119 (20.0 g, 0.078 mol) in acetone (78 mL) at 0 °C. The resulting mixture was stirred at 0 °C for 40 minutes. Sodium azide (10.2 g, 0.15 mol) in water (50 mL) was then added to the mixture. The mixture was then allowed to warm to room temperature and stirred for 4 h. Water was then added, and then the mixture was extracted several times with CH2CI2. The organic extracts were combined and washed with water and brine, dried over magnesium sulfate and concentrated under vacuum to provide an oil. The oil was taken up into toluene (200 mL), allyl alcohol (5.5 g, 0.094 mol) was added, and the mixture was heated to reflux and stirred at reflux for 17 h. The reaction mixture was then cooled to room temperature and EtOAc (250 mL) was added. Then, the mixture was washed with water and brine, dried over magnesium sulfate and concentrated under vacuum. The resulting residue was purified by silica gel chromatography (35% ethyl acetate/hexanes) to provide the carbamate 2120 (24.4 g, 0.061 mol).
Part E:
A solution of HCI/Et20 (2 N, 50 mL) was added to a solution of the carbamate 2120 (24.4 g, 0.061 mol) in CH2CI2 (100 mL). The reaction mixture was stirred overnight and then concentrated under vacuum to yield 2121 as a hygroscopic foam (17.4 g, 0.052 mol).
Example 50F:
Figure imgf000611_0001
NHBoc
Figure imgf000611_0002
Starting alcohol (2122) (1 , 5a, 6σ)-3-benzyl-6-hydroxymethyl-3- azabicyclo[3.1.0]hexane was prepared by known methods (i.e., Brighty, K. E; Castaldi, M. J Synlett, 1996, 1097). Part A: Alcohol 2122 (11 g, 54 mmol) and triethylamine (38 mL, 27 mmol) were dissolved in CH2CI2 (200 mL) and cooled to 0 °C. The cooled solution was stirred, and CH3S02CI (as a CH2CI2 solution; 6 mL, 78 mmol, 25 mL CH2CI2) was added dropwise, and the stirring was continued for 3-4 h. The reaction mixture was then washed two times with 100 mL of water and two times with 100 ml of brine. The organic and aqueous phases were separated, and the organic phase was dried, and concentrated to provide a crude product. The crude product was purified by silica gel chromatography (eluted with 1 :6 ethyl acetate:hexane). The appropriate fractions were collected from the chromatography column and concentrated to provide pure chloro compound 2123 as an oil (7 g, 59%).
Part B:
The chloro compound 2123 was dissolved in DMF (100 mL) and treated with NaN3 (10.3 g, 157 mmol), and the mixture was stirred vigorously for 36-48 h. The reaction mixture was then diluted with 100 mL water and extracted with ethyl acetate (2 x 100 mL). The organic extracts were combined, dried, and concentrated to yield pure azide 2124 (6.2 g, 87%).
Part C:
The azide 2124 (6.2 g, 27 mmol) and triphenylphosphine (15 g, 57 mmol) were dissolved in 100 mL of THF, and then water (6 mL, 333 mmol) was added. The resulting mixture was stirred vigorously for 16-24 h. The solvent was removed and the crude amine 2125 was obtained without further purification.
Part D:
The crude amine 2125 and N, N-dimethylaminopyridine (0.66 g, 5.4 mmol) were dissolved in CH2CI2 (100 mL). To this solution was added di-tert- butyldicarbonate (7 g, 33 mmol), in portions, and the reaction mixture was stirred for 16 h. The reaction mixture was then washed two times (50 mL) with water and once with brine (50 mL). The organic phase was isolated and dried, and the solvent was removed under reduced pressure. The crude product was subjected to silica gel chromatography using 1 :3 ethyl acetate:hexane as the eluting solvent. The eluted fractions were combined and concentrated to yield 6.9 g of pure carbamate 2126 (84%).
Part E:
The carbamate 2126 (1.9 g, 6.3 mmol) was dissolved in methanol (100 mL) and mixed with palladium hydroxide (20%, 0.4 g). The mixture was transferred to a Parr bottle, which was then charged with hydrogen (20 psi). The Parr bottle was shaken for 10 h. The remaining hydrogen was removed from the Parr bottle under vacuum, and the reaction was filtered through Celite. The filtrate was then concentrated to provide pure amine 2127 (1.4 g).
Example 50G
Figure imgf000613_0001
N-Benzyltropinanecarbonitrile 2128 was prepared using known procedures (see, for example: Montzka, T. A.; Matiskella, J. D.; Partyka, R. A. Tetrahedron Letters 1974, 14, 1325; Lowe, J. A.; Drozda, S. E.; McLean, S.; Bryce, D. K.; Crawford, R.T.; Snider, R. M.;Tsuchiya, M. J. Med. Chem. 1994, 37, 2831 ).
Part A:
L1AIH4 was added to dry THF (40 mL) and the mixture was then cooled to 0 °C. Then carbonitrile 2128 (0.9 g, 3.8 mmol, in a 10 mL THF solution) was added to the mixture dropwise. The reaction mixture was allowed to warm to ambient temperature and stirred for 48 h, then cooled to back to 0 °C and quenched by the sequential addition of 1 mL of water, 2 mL of 0.5 N aq. NaOH, and 1 mL of water. The resulting mixture was stirred vigorously for 2 h and then filtered through Celite. The filtrate was concentrated to yield pure 2129 as an oil (0.9 g, 100%).
Part B: The crude product 2129 and triethylamine (TEA) (0.6 mL, 4.3 mmol) were dissolved in CH2CI2 (50 mL). Di-tert-butyldicarbonate (0.85 g, 3.9 mmol) was added to this solution in portions, and the reaction mixture was stirred for 16 h. The reaction mixture was then washed two times with 50 mL of water and once with 50 mL of brine. The organic phase was dried and the solvent was removed under reduced pressure. The crude product was subjected to silica gel chromatography using 2.5% ammonia saturated methanol in CH2CI2 as the eluting solvent. The eluted fractions were combined and concentrated to yield 0.78 g of pure carbamate product 2130 (61 %).
Part C:
The carbamate 2130 (0.8 g, 2.3 mmol) was dissolved in methanol (60 mL) and treated with palladium hydroxide (20%, 0.08 g). The mixture was transferred to a Parr bottle, which was then charged with hydrogen (20 psi). The Parr bottle was shaken for 10 h. The hydrogen was removed from the Parr bottle under vacuum, and the reaction mixture was filtered through Celite. The filtrate was then concentrated to provide pure amine 2131 (0.6 g).
Example 50H:
Part B-D
Figure imgf000614_0001
2138
Figure imgf000614_0002
2133 2134
oc
Figure imgf000614_0003
Part A:
Tosylmethyl isocyanide (TosMIC) (7.2 g, 37 mmol) was added to 3-methyl-N-benzyl piperidone (2132) (4.25 g, 20.93 mmol) in DME (150 mL) and cooled to 0 °C. Ethyl alcohol (2.9 mL) and potassium tert-butoxide (7 g, 62.4 mmol) were added to the reaction mixture and stirred at room temperature for 4 hours. The reaction mixture was poured into ice and extracted with ethyl acetate (2 x 50 mL). The organic layer was washed with brine, dried over anhydrous MgSθ4 and concentrated under vacuum. The resulting residue was purified by silica gel chromatography (20% ethyl acetate/hexane) to give first 2133 (trans isomer, 1.85 g, 41 %), and second 2134 (cis isomer, 1.05 g, 22%).
Part B:
The trans intermediate 2133 (2.0 g, 9.34 mmol) was dissolved in ethyl alcohol (50 mL) and Raney Nickel (wet, 5.0 g) was added under N2, followed by addition of cone, ammonium hydroxide (1.0 mL). The mixture was subjected to H2 atmosphere at 50 p.s.i. for 16 hours with vigorous shaking. The reaction was filtered through celite under N2 and the filtrate was concentrated under vacuum to provide 1.8 g (88%) of intermediate 2135.
Part C:
Intermediate 2135 (1.0 g, 4.6 mmol) was dissolved in methyl alcohol (35 mL). A solution of BOC-anhydride (1.2 g, 5.5 mmol) in methyl alcohol (15 mL) was added dropwise at 0 °C. The resulting mixture was stirred at 0 °C for one hour, then at ambient temperature for an additional hour. The solvent was removed under vacuum to provide a crude product that was purified by silica-gel column chromatography using ethyl acetate as the eluting solvent. The relevant fractions were collected and concentrated under reduced pressure to yield 1.1 g (76%) of intermediate 2136.
Part D:
Intermediate 2136 (0.5 g, 1.57 mmol) was dissolved in methyl alcohol (25 mL). Palladium hydroxide (20 wt % Pd, 0.1 g) was added under N2. The resulting mixture was exposed to H2 atmosphere at 20 p.s.i. for 16 hours with vigorous shaking. The reaction mixture was filtered through celite and washed with methyl alcohol (1 X 25 mL). The filtrate was concentrated under vacuum to provide intermediate 2137 (0.38 g) as an oil which was used in the next step without further purification.
Intermediate 2138 was prepared using methods similar to those used in Part B-D. The following compounds were prepared using previously described procedures.
Figure imgf000616_0001
Figure imgf000617_0001
Figure imgf000618_0002
Example 51:
Example 51A:
Figure imgf000618_0001
2160 2161
PartA: Compound 2161 was synthesized according to the procedures of WO02060894A2. 1H NMR (400 MHz, DMSO-d6) δ 9.58 (bs, NH), 8.89 (m, 2H), 7.57 (m, 1 H), 4.88 (m, 1 H), 3.80 (m, 2H), 2.06 (m, 4H).
Example 51 B:
Figure imgf000619_0001
Part A:
Compound 2163 was synthesized from Compound 2162 (made according to the procedures of WO2004/052850) according to the procedures of Hanessian et. al. (Bioorg. Med. Chem Lett. 1998, 8, 2123 and the references therein.) HPLC-MS tR = 1.90 min (UV254 nm); mass calculated for formula C13H21 N04 255.2, observed LCMS m/z 278.2 (M+Na).
Part B:
Compound 2164 was synthesized according to the procedures of Example 10B Part A. HPLC-MS tR = 1.79 min (UV254 nm); mass calculated for formula C12H18CIN03 259.1 , observed LCMS m/z 282.1 (M+Na).
Part C:
Compound 2164 (190 mg) was cyclized with N-methylthiourea in DMF (2 mL) to afford 2165 (81 mg) according to the procedures of Example 10B Part B.
HPLC-MS tR = 1.27 min (UV254 nm); mass calculated for formula
C14H21 N302S 295.1 , observed LCMS m/z 296.2 (M+H).
Part D:
To compound 2165 (81 mg) in methanol (2 mL) was added 4.0 M HCI/dioxane
(1 mL) with ice bath cooling. The reaction mixture was stirred for 1.5 hours and concentrated. The residue was suspended in DCM (5 mL) and concentrated to afford impure 2166. The material was used without purification. Example 51 C:
Figure imgf000620_0001
Part A:
Compound 2167 was synthesized from Compound 2162 (made according to the procedures of WO2004/052850) according to the procedures of Hanessian et. al. Bioorg. Med. Chem Lett. 1998, 8, 2123 and the references therein. HPLC-MS tR = 1.83 min (MS); mass calculated for formula C13H21 N04 255.2, observed LCMS m/z 278.2 (M+Na).
Part B:
Compound 2168 was synthesized according to the procedures of Example 10B Part A. HPLC-MS tR = 1.78 min (MS); mass calculated for formula C12H18CIN03 259.1 , observed LCMS m/z 282.1 (M+Na).
Part C:
Compound 2168 (117 mg) was cyclized with N-methylthiourea in DMF (2 mL) to afford 2169 (35 mg) according to the procedures of Example 10B Part B. HPLC-MS tR = 1.16 min (UV25 n ); mass calculated for formula C14H21 N302S 295.1 , observed LCMS m/z 296.2 (M+H).
Part D:
To compound 2169 (35 mg) in methanol (2 mL) was added 4.0 M HCI/dioxane
(1 mL) with ice bath cooling. The reaction mixture was stirred for 1.5 hours and concentrated. The residue was suspended in DCM (5 mL) and concentrated to afford impure 2170. The material was used without purification.
Example 51 D:
Figure imgf000621_0001
Part A:
To hydroxy proline 2171 (1.52 g, 11.6 mmol) in methanol (25 mL) in an ice bath was added thionyl chloride (0.95 mL, 13 mmol). The ice bath was removed and the reaction mixture was heated to reflux overnight. The reaction mixture was cooled and concentrated. The residue was dissolved in methanol and concentrated. The residue was dissolved in diethyl ether and concentrated to afford 2172 quantitatively. 1H NMR (400 MHz, DMSO-d6) δ 10.25 (bs, 1 H), 8.95 (bs, 1 H), 4.50 (d, 1 H, J = 7.0 Hz), 4.36 (m, 1 H), 3.75 (s, 3H), 3.16 (m, 2H), 2.31 (m, 1 H), 2.13 (m, 1 H).
Part B:
To compound 2172 (2.11 g, 11.6 mmol) in DCM (25 mL) was added DIEA (4.2 mL, 24 mmol) and a solution of BOC-anhydride (3.03 g, 13. 9 mmol) in DCM (25 mL) with ice bath cooling. The reaction mixture was slowly warmed to room temperature and stirred overnight. The reaction mixture was washed with water, 0.1 N HCI, bicarbonate solution and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, loaded with ethyl acetate and eluted with 30% ethyl acetate/hexane to ethyl acetate) afforded 2173 (2.66 g) as an oil. 1H NMR (400 MHz, CDCI3) δ rotomers 4.40-4.29 (m, 2H), 3.81 and 3.80 (s, 3H), 3.75 -3.64 (m, 1 H), 3.57- 3.50 (m, 1 H), 2.35 (m, 1 H), 2.11 (m, 1 H), 1.48 and 1.44 (s, 9H).
Part C:
To compound 2173 (2.66 g, 10.8 mmol) in DCM (50 mL) in an ice bath was added triethyl amine (1.66 mL, 11.9 mmol) and methanesulfonyl chloride (1.09 mL, 14.1 mmol) dropwise. The reaction mixture was slowly warmed to room temperature and stirred overnight. The reaction mixture was quenched with ice water and the layers were separated. The aqueous layer was extracted with DCM. The combined organic layers were washed with 0.1 N HCI and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 50% ethyl acetate/hexanes) afforded 2174 (3.36 g) as an oil. HPLC-MS tR = 1.49 min (ELSD); mass calculated for formula C12H21 N07S 323.1 , observed LCMS m/z 346.1 (M+Na).
Part D:
To diphenylselenide (1.97 g, 6.3 mmol) in ethanol (20 mL) in an ice bath was added sodium borohydride (472 mg, 12.5 mmol) portion wise. The reaction mixture was stirred for 10 minutes after the bubbling had ceased. The above solution was added to compound 2174 (3.36 g, 10. 4 mmol) in ethanol (10 mL). The reaction mixture was heated to reflux for 2 hours. The reaction was cooled and the solvents removed in vacuo. The residue was partitioned between water (50 mL) and ethyl acetate (50 mL). The layers were separated. The organic layer was washed with water and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 5% to 20% ethyl acetate/hexanes) afforded 2175 (2.91 g) as an oil. HPLC-MS t = 2.32 min (UV254 nm); mass calculated for formula C18H25N04Se 399.1 , observed LCMS m/z 422.0 (M+Na).
Part E:
To 2175 (481 mg, 1.21 mmol) in DCM (5 mL) in an ice bath was added dropwise pyridine (0.162 mL, 2.0 mmol) and 50% hydrogen peroxide (0.161 mL, 2.63 mmol). The ice bath was removed and the reaction mixture was stirred at room temperature for 1.25 hours. The reaction mixture was diluted with ethyl acetate (50 mL), washed with 0.1 N HCI, bicarbonate solution and brine, dried over sodium sulfate and concentrated to afford 2176 (272 mg) as a yellow oil. HPLC-MS tR = 1.81 min (MS); mass calculated for formula C12H19N04 241.1 , observed LCMS m/z 264.1 (M+Na).
Part F:
Compound 2177 (123 mg) was prepared from 2176 (202 mg, 0.84 mmol) according to the procedure described in Example 10B Part B. HPLC-MS tR = 1.70 min (MS); mass calculated for formula C11 H16CIN03 245.1 , observed LCMS m/z 264.1 (M+Na).
Part G:
Compound 2178 (48 mg) was prepared as described in Example 51 B Part C from compound 2177 (123 mg, 0.5 mmol). HPLC-MS tR = 1.12 min (UV25 nm); mass calculated for formula C13H19N302S 281.1 , observed LCMS m/z 282.1 (M+H).
Part H:
A mixture of compound 2178 (48 mg, 0.17 mmol), TFA (2 mL) and DCM (2 mL) were stirred at room temperature for 1 hour. The solvents were removed and the residue was dissolved in DCM (5 ml) and concentrated to afford 2179 (70 mg). 1H NMR (400 MHz, CDCI3) δ 9.58 (bs, NH), 6.75 (s, 1H), 6.26 (d, 1 H, J = 6.3 Hz), 5.92 (d, 1 H, J = 7.5 Hz), 5.77 (m, 1 H), 4.43 (d, 1 H, J =17.2 Hz), 4.28 (d, 1 H, J =17.2 Hz), 3.11 (s, 3H).
Example 51 E:
Figure imgf000623_0001
2180 2181 2182
Part A:
Compound 2180 was prepared using a modification of the procedure in J. Org. Chem. 1998, 63, 8, 2451-2455. Compound 2180 (0.400 g, 2.72 mmol) was dissolved in THF (60 mL) and cooled in a dry ice/acetone bath. A solution of methyllithium (1.6 M in diethylether, 18 mL) was added dropwise and the solution was warmed to room temperature. The reaction was quenched with brine after 10 minutes. The reaction mixture was poured into water and extracted with ethyl acetate. The combined organics were washed with 1 N HCI, saturated sodium bicarbonate and water, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 50% ethyl acetate/hexanes) afforded 2181 (0.200 g). HPLC-MS tR = 1.126 min (UV254 nm); mass calculated for formula CioHnNO 161.1 , observed LCMS m/z 162 (M+H).
Part B:
Compound 2181 (0.200 g, 1.24 mmol) was dissolved in THF (10 mL) and a solution of borane (2.5 M in THF, 3 mL) was added dropwise and the solution was stirred at reflux overnight. The reaction was quenched with a solution of 1 M sodium hydroxide (2 mL) and methanol (2 mL) and reflux was continued for 5 hours. The reaction mixture was poured into water and extracted with ethyl acetate. The combined organics were washed with saturated sodium bicarbonate, water, dried over sodium sulfate and concentrated to afford 2182 (65 mg). HPLC-MS tR = 0.595 min (UV25 nm); mass calculated for formula C-KJHHN 147.1 , observed LCMS m/z 148 (M+H).
Example 51 F:
Figure imgf000624_0001
Part A:
Compound 2184 was prepared using a modification of the procedure in J. Med.Chem.. 1994, 37, 23, 3878-3881. Compound 2183 (2.0 g, 12.9 mmol) was dissolved in methylene chloride and cooled in an ice bath. Triethylamine (3.61 mL, 26 mmol) was added followed by the dropwise addition of TMSOTf (2.81 mL, 15.5 mmol). The reaction was warmed to room temperature and stirred for 30 minutes. Saturated sodium bicarbonate was added slowly to quench the reaction and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate. In a separate flask TiCI4 (1 M in toluene, 20 mL) was added to methylene chloride (80 mL) at -78 °C. Acetone (1.2 g, 22 mmol) was added and stirred for 2 minutes. The silyl enol ether from the earlier was added and the reaction was stirred for 4 hours. After warming to room temperature, the reaction was quenched with saturated sodium bicarbonate and extracted with methylene chloride. The combined organic layers were dried over sodium sulfate and evaporated. Purification by column chromatography (Si02, 33% ethyl acetate/hexanes) afforded 2184 (600 mg). 1H NMR (400 MHz, CDCI3) δ 7.90 (d, 1 H), 7.80 (d, 1 H), 7.55 (d, 1 H), 7.40 (t, 1 H), 3.15 (s, 2H), 1.40 (s, 6H).
Part B:
Compound 2184 (600 mg, 2.84 mmol) was dissolved in methylene chloride (2 mL) and pyridine (2 mL) and cooled in an ice bath. TFAA (0.894 g, 4.26 mmol) was added dropwise and stirred 1 hour at room temperature. The reaction mixture was partitioned between ethyl acetate and water. The organic layers were washed with 1 N HCI, saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated. The residue was dissolved in methylene chloride (5 mL) and DBU (0.5 mL) was added and stirred for 1 hour at room temperature. The reaction mixture was partitioned between methylene chloride and water. The organic layers were washed with 1 N HCI, saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated. Purification by column chromatography (Si02, 10% ethyl acetate/hexanes) afforded 2185 (460 mg). 1H NMR (400 MHz, CDCI3) δ 7.90 (d, 1 H), 7.80 (d, 1 H), 7.50 (d, 1 H), 7.40 (t, 1 H), 6.70 (m, 1 H), 2.20 (d, 3H), 2.05 (d, 3H).
Part C:
Compound 2185 (460 mg, 2.38 mmol) was dissolved in nitromethane (295 mg, 4.76 mmol) and benzyltrimethylammonium hydroxide (40% weight in MeOH, 200 mg) and stirred for 2 hours at room temperature. The reaction was quenched with acetic acid (1 mL) and partitioned between diethyl ether and water. The organic layer was dried over sodium sulfate and concentrated to afford 2186 (550 mg). 1H NMR (400 MHz, CDCI3) δ 7.90 (d, 1 H), 7.80 (d, 1 H), 7.58 (d, 1 H), 7.40 (t, 1 H), 4.70 (s, 2H), 3.10 (s, 2H), 1.30 (s, 6H).
Part D:
Compound 2187 was prepared according to the procedure in Chem. Ber.
1958, 91, 1978-1980. 1H NMR (400 MHz, CDCI3) δ 7.38 (m, 1 H), 7.22 (m, 2H), 7.18 (m, 1 H), 4.30 (m, 1 H), 2.90-2.80 (dd, 2H), 2.00 (m, 1 H), 1.50 (m, 1 H), 1.15 (d, 6H).
Example 51 G:
Figure imgf000626_0001
Part A:
Compound 2186 (500 mg, 1.96 mmol) and ammonium chloride (104 mg, 1.96 mmol) were dissolved in THF (5 mL) and water (5 mL) and cooled in an ice bath. Zn dust (637 mg, 9.8 mmol) was added in portions and stirred overnight. The reaction mixture was partitioned between ethyl acetate and water. The organic layers were washed with saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated. The residue was dissolved in methylene chloride (10 mL) and m-CPBA (510 mg, 2.94 mmol) was added and stirred for 2 hours. The reaction mixture was partitioned between methylene chloride and water. The organic layers were washed with saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated to yield 2188 (300 mg). 1H NMR (400 MHz, CDCI3) δ 7.80 (m, 1 H), 7.65 (m, 2H), 7.40-7.30 (m, 2H), 3.80 (t, 2H), 2.80 (t, 2H), 1.20 (s, 6H).
Part B:
Compound 2188 (300 mg, 1.35 mmol) was dissolved in THF (10 mL) and MeMgBr (3M in THF, 5 mL) was added dropwise. After stirring for 30 minutes at room temperature the reaction mixture was partitioned between ethyl acetate and water. The organic layers were washed with saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated to afford 2189 (280 mg). H NMR (400 MHz, CDCI3) δ 7.53 (t, 1 H), 7.42 (m, 1 H), 7.30-7.20 (m, 2H), 3.20 (m, 2H), 2.00 (m, 2H), 1.55 (s, 3H), 1.20 (s, 3H), 1.15 (s, 3H).
Part C: Compound 2189 (280 mg, 1.26 mmol) was dissolved in 1 N HCI (10 mL) and Zn dust (0.5 g) was added. The reaction mixture was stirred at 60 °C for 12 hours. The reaction mixture was partitioned between ethyl acetate and water. The organic layers were washed with saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated to afford 2190 (110 mg). 1H NMR (400 MHz, CDCI3) δ 7.53 (bs, 1 H), 7.38 (m, 1 H), 7.30-7.20 (m, 2H), 2.85 (m, 1 H), 2.75 (m, 1 H), 2.05 (m, 1 H), 1.85 (m, 1 H), 1.50 (s, 3H), 1.20 (s, 3H), 0.95 (s, 3H).
Example 51 H:
Figure imgf000627_0001
Part A:
Compound 2191 (2.8 g, 11.2 mmol) was dissolved in toluene (10 mL) and MeOH (10 mL) and cooled in an ice bath. A solution of TMS diazomethane in hexanes (2M, 8.4 mL) was added dropwise until yellow color persisted. The reaction was quenched with acetic acid until color became clear and the solvent was removed under reduced pressure. The residue was partitioned between ethyl acetate and water. The organic layer was washed with 1 N HCI, saturated sodium bicarbonate, brine, dried over sodium sulfate and concentrated to afford 2192 (3.1 g). The product was used without purification.
Part B:
Diisopropylamine (3.9 mL, 28 mmol) was dissolved in THF (20 mL) and cooled to -40 °C. A solution of n-BuLi (2.5M in hexanes, 8.9 mL) was added dropwise and the reaction was stirred for 30 minutes. Compound 2192 (3.1 g, 11.2 mmol) dissolved in THF (10 mL) was added dropwise at -78 °C and stirred for another 30 minutes. Iodomethane (3.1 g, 22.4 mmol) was added dropwise and the reaction was stirred for one additional hour. The reaction was warmed to room temperature and quenched with brine and extracted with ethyl acetate. The combined organic layers were washed with water, brine, dried over sodium sulfate and concentrated to provide 2193 (2.5 g). 1H NMR (400 MHz, CDCI3) δ 7.40-7.20 (m, 5H), 5.20-5.00 (m, 2H), 3.70-3.40 (d, 3H), 3.70-3.60 (m, 2H), 2.20 (m, 1 H), 1.90 (m, 3H), 1.60 (d, 3H).
Part C:
Diisopropylamine (1.24 mL, 8.85 mmol) was dissolved in THF (10 mL) and cooled to -40 °C. A solution of n-BuLi (2.5M in hexanes, 3.5 mL) was added dropwise and the reaction was stirred for 30 minutes. This solution was added dropwise to a solution of compound 2193 (450 mg, 1.61 mmol) and chloroiodomethane (1.12 g, 6.44 mmol) in THF (10 mL) at -78 °C. The reaction was stirred for 30 minutes and then quenched by the dropwise addition of acetic acid (1 mL) in THF (5 mL). After 10 minutes of stirring the reaction mixture was partitioned between ethyl acetate and water. The organic layers were washed with saturated sodium bicarbonate, brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 25% ethyl acetate/hexanes) afforded 2194 (330 mg). 1H NMR (400 MHz, CDCI3) δ 7.40-7.20 (m, 5H), 5.20-5.00 (m, 2H), 4.40-4.00 (m, 2H), 3.80-3.60 (m, 2H), 2.20 (m, 1 H), 2.00-1.8 (m, 3H), 1.60-1.40 (m, 3H).
Part D:
Compound 2194 (175 mg, 0.59 mmol) was dissolved in DMF (5 mL) and thiourea (91 mg, 1.2 mmol) was added and stirred at room temperature overnight. The reaction mixture was partitioned between ethyl acetate and water. The organic layers were washed with saturated sodium bicarbonate, brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 50% ethyl acetate/hexanes) afforded 2195 (115 mg). HPLC-MS tR = 1.17 min (UV254 nm); mass calculated for formula Cι69N302S 317.1 , observed LCMS m/z 318.1 (M+H).
Part E:
Compound 2195 (115 mg, 0.36 mmol) was dissolved in 30% HBr/AcOH (2 mL) and stirred for 2 hours. The solvent was removed under reduced pressure and the residue was dissolved in water and washed with diethyl ether. The aqueous layer was lyophilized to provide 2196 as a di-HBr salt (100 mg).
Example 511:
Figure imgf000629_0001
2197 2198 2199 2200 2201
Part A:
Compound 2197 (2.3 g, 10.6 mmol), 4-chloro-2,6-dimethoxytriazine (2.4 g, 13.9 mmol) and NMM (7.3 mL, 53 mmol) were dissolved in THF (50 mL) and stirred for 1 hour. N,0- dimethylhydroxylamine hydrochloride (2.05 g, 21.2 mmol) was added and the reaction mixture was stirred overnight at room temperature. The reaction mixture was partitioned between ethyl acetate and water. The organic layers were washed with 1 N HCI, saturated sodium bicarbonate, brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 50% ethyl acetate/hexanes) afforded 2198 (2.0 g). 1H NMR (400 MHz, CDCI3) δ 4.70-4.60 (m, 1 H), 3.75 (m, 3H), 3.60- 3.40 (m, 2H), 3.20 (s, 3H), 2.20 (m, 1 H), 2.00-1.80 (m, 3H), 1.50-1.40 (m, 9H).
Part B:
Compound 2198 (550 mg, 2.13 mmol) was dissolved in THF (10 mL) and a solution of lithium (trimethylsilyl)acetylide (0.5 M in THF, 12 mL) was added dropwise and stirred overnight. The reaction mixture was partitioned between ethyl acetate and 1 N HCI. The organic layer was washed with 1 N HCI, saturated sodium bicarbonate, brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 20% ethyl acetate/hexanes) afforded 2199 (370 mg). HPLC-MS tR = 1.69 min (UV25 nm); mass calculated for formula Cι27N03 223.1 , observed LCMS m/z 168.1 (M-(t-butyl)).
Part C:
Compound 2199 (350 mg, 1.56 mmol) and diethylamine (228 mg, 3.13 mmol) were dissolved in EtOH (1.5 mL) and water (1.5 mL) and stirred for 3 hours. The reaction mixture was partitioned between ethyl acetate and water. The organic layers were washed with saturated sodium bicarbonate, brine, dried over sodium sulfate and concentrated. The residue was combined with methyl guanidine hydrochloride (756 mg, 7.8 mmol) and sodium carbonate (826 mg, 7.8 mmol) in EtOH (6 mL) and refluxed for 48 hours. The reaction mixture was partitioned between ethyl acetate and water. The organic layer was washed with saturated sodium bicarbonate, brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 80% ethyl acetate/hexanes) afforded 2200 (210 mg). HPLC-MS tR = 1.26 min (UV254 nm); mass calculated for formula Cι4H22N4θ2 278.1 , observed LCMS m/z 279.1 (M+H).
Part D:
Compound 2200 (105 mg, 0.377 mmol) was dissolved in 4 M HCI in dioxane (2 mL) and methanol (0.5 mL) and stirred at room temperature for 1 hr. The reaction mixture was concentrated and triturated with diethyl ether to afford 2201 (74 mg) as an HCI salt. 1H NMR (400 MHz, DMSO-d6) δ 10.00 (bs, 1 H), 8.70 (bs, 1 H), 8.30 (s, 1 H), 6.70 (d, 1 H), 4.60 (bs, 1 H), 3.40 (m, 2H), 2.75 (s, 3H), 2.40 (m, 1 H), 2.00-1.80 (m, 3H).
Example 51J:
Figure imgf000630_0001
Part A:
Compound 2203 was prepared according to a procedure described in J. Org.
Chem. 1991, 57, 7034-7038.
Part B:
A mixture of 2203 (200 mg, 1.42 mmol) and zinc dust (278 mg, 4.25 mmol) in concentrated hydrochloric acid (1.4 mL) and water (5.8 mL) was stirred at reflux overnight. The mixture was made basic with potassium hydroxide pellets, decanted and the aqueous solution extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated to give 2204 as a yellow sticky solid (95 mg). 1H NMR (400 MHz, CDCI3) δ 3.35 (m, 2H), 1.88 (s, 2H), 1.66 (s, 6H), 1.30 (s, 6H).
Example 51 K:
\ \ \ \
NH NBoc NBoc NH
Part A Part B Part C N - A N 2 HCI
Y^
VNBoc VNBoc VNBoc CNH
2205 2206 2207 2208
Compound 2205 was prepared using procedures described in Example 10B. Part A:
To the solution of 2-methylaminothiazole 2205 (100 mg, 0.35 mmol) and DMAP (135 mg, 1.0 mmol) in THF (5 mL), Boc20 (218 mg, 1.0 mmol) was added at room temperature. The resulting mixture was stirred overnight and then diluted with ethyl acetate (30 mL). The organics was washed with water, Brine and dried over sodium sulfate and concentrated. The residue which was purified by column chromatography (Si02, 20% ethyl acetate/hexanes) afforded the protected product 2206 (130 mg) as semi-oil. HPLC-MS tR = 2.42 min (UV254 nm); mass calculated for formula C18H29N304S 383.2, observed LCMS m/z 384.1 (M+H).
Part B:
The solution of thiazole 2206 (150 mg, 0.39 mmol) in THF (5 mL) was cooled to -78 °C, and n-BuLi (2.5 M in hexane, 0.19 mL, 0.47 mmol) was added slowly. The resulting mixture was stirred at -78 °C for 30 min, and then iodomethane (0.1 mL, 1.55 mmol) was added. The mixture was stirred for another 30 min followed by the addition of saturated NH4CI solution (15 mL). The aqueous layer was extracted with ethyl acetate. The combined organics were washed with water, brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 10% ethyl acetate/hexanes) afforded recovered thiazole 2206 (36 mg) and 2207 (100 mg). HPLC-MS tR = 2.51 min (UV254 nm); mass calculated for formula C19H31 N304S 397.2, observed LCMS m/z 398.2 (M+H).
Part C:
To a solution of 5-methylthiazole 2207 (100 mg, 0.25 mmol) in dioxane (2 mL) was added HCI (4N in dioxane, 4 mL) followed by water (0.5 mL). The mixture was stirred at room temperature for 1 hour and concentrated. The resulting residue 2208 (72 mg) was dried under vacuum and used in the next step without further purification.
Example 51 L:
Figure imgf000632_0001
2206 2209A 2209B 2210
Part A:
To the solution of thiazole 2206 (190 mg, 0.5 mmol) in chloroform (5 mL), NBS (107 mg, 0.6 mmol) was added. The mixture was heated to 50 °C and stirred for 1 hour. After cooling to room temperature, the mixture was concentrated and purified by column chromatography (Si02, 10% ethyl acetate/hexanes) to afford 5-bromothiazole 2209A (221 mg). HPLC-MS tR = 2.65 min (UV254 nm); mass calculated for formula C18H28BrN304S 461.1 , observed LCMS m/z 462.1 (M+H).
Part B:
A mixture of 5-bromothiazole 2209A (221 mg, 0.48 mmol), Zn(CN)2 (117 mg, 1.0 mmol), Pd2(dba)3 (45 mg, 0.05 mmol) and DPPF (55 mg, 0.1 mmol) in a 25-ml round bottom flask was flushed with argon for 3 min. Under the argon, DMA (3 mL) was added and the flask was sealed under the argon atmosphere. The mixture was heated to 85 °C and stirred overnight. After cooling to room temperature, ethyl acetate (30 mL) was added to dilute the reaction mixture and the solution was filtered through celite. The filtrate was concentrated and purified by column chromatography (Si02, 10% ethyl acetate/hexanes) to afford 5-cyanothiazole 2209B (167 mg) as oil. HPLC-MS tR = 2.39 min (UV254 nm); mass calculated for formula C19H28N404S 408.2, observed LCMS m/z 409.2 (M+H).
Part C:
Compound 2210 (110 mg) was prepared from 2209B (167 mg, 0.41 mmol) according to the procedure described in Example 51 K Part C. HPLC-MS tR = 0.62 min (UV254 nm); mass calculated for formula C9H12N4S 208.1 , observed LCMS m/z 209.1 (M+H).
Example 51 M:
Figure imgf000633_0001
2206 2211 2212 2213
Part A:
A solution of n-BuLi (2.5 M in hexane, 0.24 mL, 0.6 mmol) was added slowly to a solution of thiazole 2206 (190 mg, 0.5 mmol) in THF (5 mL) at -78 °C. The resulting mixture was stirred at -78 °C for 1 hour, and then methyl disulfide (94 mg, 1.0 mmol) was added. The mixture was stirred for another one hour, and then warmed to room temperature slowly. Then saturated NH4CI solution was added to quench the reaction. The mixture was extracted with ethyl acetate (30 mL x 3). The combined organics were dried over sodium sulfate, concentrated and purified by column chromatography (Si02, 10% ethyl acetate/hexanes) to afford 5-methylsulfidethiazole 2211 (137 mg) and recovered thiazole 2206 (38 mg). HPLC-MS tR = 2.62 min (UV254 nm); mass calculated for formula C19H31 N304S2 429.2, observed LCMS m/z 430.1 (M+H). Part B:
A mixture of sulfide 2211 (137 mg, 0.32 mmol) and m-CPBA (-77%, 230 mg, 1.0 mmol) in dichloromethane (10 mL) was stirred at room temperature overnight. Ethyl acetate (80 mL) was added to dilute the mixture and the organics were washed with saturated sodium bicarbonate solution twice followed by brine, dried over sodium sulfate, concentrated. The residue was purified by column chromatography (Si02, 20% ethyl acetate/hexanes) afforded 5-methylsulfonethiazole 2212 (141 mg). HPLC-MS tR = 2.62 min (UV254 nm); mass calculated for formula C19H31 N306S2 461.2, observed LCMS m/z 406.1 (M+H- f-Bu).
Part C:
Compound 2213 (110 mg) was prepared from 2212 (141 mg, 0.30 mmol) according to the procedure described in Example 51 K Part C. HPLC-MS t = 0.63 min (UV254 nm); mass calculated for formula C9H15N302S2 261.1 , observed LCMS m/z 262.0 (M+H).
Example 51 N:
Figure imgf000634_0001
Starting pyrroline derivative 2214 was prepared by known methods (Billet, M; Schoenfelder, A; Klotz, P.; Mann, A.. Tetrahedron Letters, 2002, 1453).
Part A:
Trimethylsilyl iodide (0.44 g, 2.24 mmol) was added dropwise to a solution of 2214 (0.47 g, 1.5 mmol; 20 mL CH2CI2) at room temperature and stirred for 3 hours. The mixture was cooled to 0 °C before quenching with methanol (5 mL). The solvent was removed under reduced pressure to provide crude amine 2215, which was used without further purification. Example 510:
Figure imgf000635_0001
Part A:
Borane dimethyl sulfide (2.0 M solution in toluene, 1.45 mL, 2.9 mmol) was added dropwise to a solution of intermediate 2214 (0.65 g, 2 mmol) in THF (10 mL) at 0 °C. After addition, the mixture was stirred at room temperature for 3 hours. The reaction mixture was quenched with dropwise addition of water (1.0 mL) and 3N NaOH (3.0 mL). The resulting mixture was stirred for 5 minutes and then cooled to 0 °C before adding 30% H202 (6.0 mL) dropwise. The mixture was stirred at ambient temperature for 2 hours and then treated with 5% Na2S2θ3 (25 mL) and stirred additional half hour. The reaction mixture was diluted with ethyl acetate (100 mL), and the organics were washed with brine, dried (Na2S0 ), filtered, and concentrated under vacuum to provide a crude residue which was purified by silica- gel column chromatography (1 :3 to 1 :2 ethyl acetate / hexane) to give first intermediate 2217 (0.3 g, 44%) followed by intermediate 2216 (0.2 g, 29%).
Part B:
Intermediate 2216 (0.075 g, 0.22 mmol) was dissolved in ethyl alcohol (1.5 mL). Then 1 ,4-cyclohexadiene (0.21 mL, 2.24 mmol) was added, followed by addition of 10% Pd/C under N2. The reaction mixture was stirred at r.t. for 2 hours, filtered through celite and concentrated under vacuum to give intermediate 2218 (0.035 g, 80%).
Compound 2219 was prepared by using methods similar to those described in Parts A-B for intermediate 2218. Example 51 P:
Figure imgf000636_0001
Part A:
Intermediate 2214 (0.1 g, 0.3 mmol) was dissolved in -butanol (3 mL) and water (3 mL). Potassium hexacyanoferrate (0.31 g, 0.94 mmol) and potassium carbonate (0.13 g, 0.94 mmol) were added and stirred for 15 minutes. To this mixture osmium tetraoxide (0.005 g, 0.02 mmol) was added and stirred for 16 h. The reaction was diluted with ethyl acetate (50 mL) and the organics were washed once with brine (10 mL), dried (Na2S04), filtered and concentrated under reduced pressure to provide intermediate 2220 (0.1 g, 91%).
Part B:
Diol 2220 (0.1 g, 0.28 mmol) was dissolved in acetone (5 mL) and 2,2- dimethoxypropane (0.045 g, 0.43 mmol) was added, followed by addition of catalytic amount of p-toluenesulfonic acid (0.06 g, 0.03 mmol). The reaction mixture was stirred at room temperature for 14 h. The solvent was removed under vacuum to provide a crude product which was purified by silica-gel preparative chromatography using ethyl acetate/hexane (1/3) as the eluting solvent to yield 2221 (0.075 g, 68%).
Part C:
Intermediate 2221 (0.06 g, 0.15 mmol) was dissolved in CH2CI2 (0.5 mL), followed by addition of trimethylsilyl iodide (0.034 mL, 0.24 mmol) at 0 °C. The mixture was stirred at r.t. for 2 hours. The reaction was quenched with methanol (5 mL) and stirred for another 2 hours. The reaction mixture was concentrated to provide the crude amine 2222, which was used without additional purification.
Example 51 Q:
Figure imgf000637_0001
2227 2228 2229 2230
Figure imgf000637_0002
2231 2232
Part A:
Compound 2223 (2.2 g, 13.9 mmol) was dissolved in methylene chloride (25 mL) and triethylamine (3.9 mL, 27.8 mmol), DMAP (100 mg), and
Figure imgf000637_0003
butyldicarbonate (3.33 g, 15.3 mmol) were added. The reaction mixture was stirred for 5 hours at room temperature and then diluted with water and methylene chloride. The organic layers were washed with 1 N HCI, saturated sodium bicarbonate, water, brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 33% ethyl acetate/hexanes) afforded the desired product (3.0 g). 1H NMR (400 MHz, CDCI3) δ 4.60 (m, 1 H), 4.20 (q, 2H), 2.70-2.60 (m, 1 H), 2.50 (m, 1 H), 2.40-2.30 (m, 1 H), 2.05 (m, 1 H), 1 .50 (s, 9H), 1 .20 (t, 3H).
Part B:
Compound 2162 (630 mg, 2.44 mmol) was added to f-butoxy- bis(dimethylamino)methane (2224) (0.705 mL, 3.42 mmol) and stirred at 80 °C overnight. The excess reagent was removed under reduced pressure to provide 2225 that was used without purification (750 mg). 1H NMR (400 MHz, CDCI3) δ 7.10 (m, 1 H), 4.50 (m, 1 H), 4.30-4.20 (m, 2H), 3.30 (m, 1 H), 3.00 (s, 6H), 2.80 (m, 1 H), 1.50 (s, 9H), 1.30 (m, 3H). Part C:
Compound 2225 (750 mg, 2.4 mmol) was dissolved in ethyl acetate (12 mL) and 10% Pd-C (200 mg) was added under an argon atmosphere. The reaction mixture was placed under a hydrogen atmosphere and stirred for 96 hours at room temperature. The reaction was filtered and the solvent was evaporated. Purification by column chromatography (Si02, 33% ethyl acetate/hexanes) afforded the desired product (350 mg). 1H NMR (400 MHz, CDCI3) δ 4.50 (m, 1 H), 4.25 (q, 2H), 2.60 (m, 2H), 1.65 (m, 1 H), 1.50 (s, 9H), 1.32 (t, 3H), 1.28 (d, 3H).
Part D:
Compound 2226 (200 mg, 0.73 mmol) was dissolved in THF (2 mL) and borane dimethyl sulfide (2M in THF, 1.5 mL) was added. The reaction mixture was stirred for 40 hours and then cooled in an ice bath and quenched slowly with methanol. The solvent was evaporated and the residue was purified by column chromatography (Si02, 33% ethyl acetate/hexanes) to afford the desired product (80 mg). 1H NMR (400 MHz, CDCI3) δ 3.95 (m, 1 H), 3.75- 3.50 (m, 3H), 2.75 (t, 1 H), 2.25 (m, 2H), 1.50 (s, 9H), 1.20 (m, 1 H), 1.00 (d, 3H).
Part E:
Compound 2227 (70 mg, 0.32 mmol) was dissolved in acetone (3 mL) and Jones Reagent (1 mL) was added dropwise. The reaction mixture was stirred overnight and then quenched with methanol. The mixture was then filtered and concentrated. The residue was dissolved in saturated sodium bicarbonate and washed with diethyl ether. The aqueous layer was acidified to pH 2 and then extracted with ethyl acetate. The ethyl acetate layers were dried over sodium sulfate and concentrated to provide the desired product (55 mg). 1H NMR (400 MHz, CDCI3) δ 4.30-4.20 (m, 1 H), 3.80-3.70 (m, 1 H), 2.95 (m, 1 H), 2.50-2.30 (m, 1 H), 2.25 (m, 1 H), 1.80-1.60 (m, 1 H), 1.50 (d, 9H), 1.05 (m, 3H). Part F:
Compound 2228 (370 mg, 1.62 mmol) was dissolved in toluene (2 mL) and methanol (2 mL) and cooled in an ice bath. A solution of trimethylsilyl diazomethane (2M in hexanes, 1.5 mL) was added dropwise. The solution was stirred for 1 hour and then concentrated. The residue was dissolved in ethyl acetate and water. The organic layer was washed with 1 N HCI, saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated to provide the methyl ester 2229 (310 mg).
Part G:
In a separate flask diisopropylamine (1.0 mL) was dissolved in THF (10 mL) and cooled to -78 °C. A solution of n-butyl lithium (2.5 M in hexanes, 2.55 mL) was added dropwise and stirred for 30 minutes. This solution was added dropwise to a solution of the methyl ester 2229 (310 mg, 1.27 mmol) and chloroiodomethane (0.9 g, 5.08 mmol) in THF (5 mL) at -78 °C. After thirty minutes the reaction was quenched dropwise with acetic acid (1 mL) in THF (5 mL). The reaction mixture was warmed to room temperature and diluted with water and ethyl acetate. The organic layer was dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 25% ethyl acetate/hexanes) afforded the desired alpha-chloro ketone 2230 (150 mg).
Part H:
The alpha-chloro ketone 2230 (150 mg, 0.575 mmol) was dissolved in DMF (5 mL) and ethyl thiourea (134 mg, 1.15 mmol) was added and stirred at 80 °C overnight. The reaction mixture was partitioned between ethyl acetate and water. The organic layer was washed with brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, ethyl acetate) afforded the desired product 2231 (85 mg). HPLC-MS tR = 1.35 min (UV254 nm); mass calculated for formula C15H25N302S 311.1 , observed LCMS m/z 312.1 (M+H).
Part i:
Compound 2231 (85 mg, 0.30 mmol) was dissolved in 4M HCI in dioxane
(2mL) and methanol (0.5 mL) and stirred for 30 minutes. The solvents were removed to provide 2232 as a di-HCl salt (65 mg). 1H NMR (400 MHz, DMSO-de) δ 9.50 (bs, 1 H), 8.70 (bs, 1 H), 7.80 (bs, 1 H), 6.70 (s, 1 H), 4.50 (m, 1 H), 3.30-3.20 (m, 2H), 2.75-2.65 (m, 2H), 2.40 (m, 2H), 1.70 (m, 1H), 1.15 (t, 3H), 1.10 (d, 3H).
The following compounds were prepared using previously described procedures.
Figure imgf000640_0001
Figure imgf000641_0001
Figure imgf000642_0001
Figure imgf000643_0001
Figure imgf000644_0002
Example 52:
Example 52A:
Figure imgf000644_0001
Part A:
Sodium pellets (1.0 g, 43 mmol) were dissolved in ethanol (35 mL). Compound 2272 (1.4 g, 7.2 mmol) was added to the solution and the reaction was refluxed overnight. The solvent was removed under reduced pressure and the residue was partitioned between ethyl acetate and water. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated to afford 2273 (1.3 g). The product was used without further purification. HPLC-MS tR = 1.92 min (UV254 πm); mass calculated for formula C7H8NOBr 201.0, observed LCMS m/z 202.1 (M+H).
Part B:
Compound 2273 (171 mg, 0.8592 mmol) was dissolved in toluene (10 mL).
The boronic acid 1270 (409 mg, 1.69 mmol), Pd2(dba)3 (38 mg, 0.0423 mmol), S-PHOS (34 mg, 0.084 mmol), potassium phosphate (358 mg, 1.69 mmol) were added to the solution and stirred at reflux overnight. The reaction mixture was cooled to room temperature and filtered, and the toluene was removed under reduced pressure. The residue was partitioned between ethyl acetate and water. The combined organic layers were dried over sodium sulfate and concentrated to afford 2274 (190 mg). The material was used without further purification. HPLC-MS tR = 2.21 min (UV254 nm); mass calculated for formula Cι9H24N203328.2, observed LCMS m/z 329.2 (M+H).
Part C:
Compound 2274 (190 mg, 0.584 mmol) was dissolved in methylene chloride (4 mL) and TFA (1 mL) and stirred for 30 minutes. The solvents were removed to provide 2275 as the TFA salt (190 mg).
Example 52B: Part B
Figure imgf000645_0001
Figure imgf000645_0002
Part A:
Compound 559 (100 mg, 0.35 mmol) was dissolved in dioxane (5 mL). Then boronic acid 2276 (113 mg, 0.52 mmol), Pd2(dba)3 (27 mg, 0.03 mmol), triphenylphosphine (13.7 mg, 0.052 mmol), potassium phosphate (148 mg, 0.70 mmol) were added to the solution and stirred at reflux overnight. The reaction mixture was cooled to room temperature and filtered, and the toluene was removed under reduced pressure. The residue was partitioned between ethyl acetate and water. The combined organic layers were dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 25% ethyl acetate/hexanes) afforded 2277 (80 mg). 1H NMR (400 MHz, CDCI3) δ 7.50 (d, 1H), 7.45 (m, 2H), 7.30-7.15 (m, 5H), 7.20-7.10 (m, 2H), 7.00-6.90 (m, 3H), 4.30 (d, 2H), 1.45 (s, 9H).
Part B: Compound 2277 (80 mg, 0.21 mmol) was dissolved in 4 M HCI in dioxane (2 mL) and stirred at room temperature for 1 hr. The reaction was diluted with diethyl ether (5 mL) and 2278 (60 mg) was collected by filtration.
Example 52C:
Figure imgf000646_0001
Part A:
Compound 2279 (500 mg, 3.08 mmol), cesium carbonate (3.0 g, 9.25 mmol), and 2-bromophenol (527 mg, 3.08 mmol) were dissolved in DMF (15 mL) and stirred at 60 °C for 2 hours. The reaction mixture was partitioned between ethyl acetate and water. The organic layers were washed with saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated. Purification by column chromatography (Si02, 10% ethyl acetate/hexanes) afforded 2280 (400 mg). 1H NMR (400 MHz, CDCI3) δ 7.50 (m, 1 H), 7.20 (m, 1 H), 6.85 (d, 1 H), 6.80 (t, 1 H), 3.90 (d, 2H), 2.40 (m, 1 H), 1.85 (m, 2H), 1.60- 1.45 (m, 4H), 1.40 (m, 2H).
Part B:
Compound 2280 (157 mg, 0.50 mmol), compound 568 (250 mg, 1.02 mmol), Pd2(dba)3 (9.1 mg, 0.01 mmol), S-PHOS (8.3 mg, 0.02 mmol), potassium fluoride (350 mg, 3.0 mmol) were dissolved in dioxane (8 mL) and stirred at reflux overnight. The reaction mixture was cooled to room temperature and filtered, and the toluene was removed under reduced pressure. The residue was partitioned between ethyl acetate and water. The combined organic layers were dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 10% ethyl acetate/hexanes) afforded 2281 (200 mg). 1H NMR (400 MHz, CDCI3) δ 7.60 (m, 2H), 7.40 (m, 2H), 7.30 (m, 2H), 7.00 (m, 2H), 4.60 (d, 2H), 3.85 (d, 2H), 2.35 (m, 1 H), 1.80 (m, 2H), 1.60 (m, 4H), 1.30 (m, 2H). Part C:
Compound 2281 (100 mg, 0.317 mmol) was dissolved in MeOH (5 mL) and a saturated solution of potassium carbonate (1 mL) was added and the reaction was stirred for 3 hours. The reaction mixture was partitioned between ethyl acetate and water. The organic layers were washed with saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated to afford 2282 (40 mg). 1H NMR (400 MHz, CDCI3) δ 7.55 (d, 2H), 7.40 (m, 4H), 7.00 (m, 2H), 4.60 (d, 2H), 3.85 (d, 2H), 2.35 (m, 1 H), 1.80 (m, 2H), 1.60 (m, 4H), 1.35 (m, 2H).
Example 52D:
Figure imgf000647_0001
Part A:
Compound 567 (500 mg, 2.29 mmol), 4-cyanophenyIboronic acid (483 mg, 3.43 mmol), Pd2(dba)3 (70 mg, 0.08 mmol), triphenylphosphine (70 mg, 0.26 mmol), potassium phosphate (970 mg, 4.58 mmol) were dissolved in dioxane (15 mL) and stirred at reflux overnight. The reaction mixture was cooled to room temperature and filtered and the dioxane was removed under reduced pressure. The residue was partitioned between ethyl acetate and water. The combined organic layers were dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 5% ethyl acetate/hexanes) afforded 2283 (240 mg). 1H NMR (400 MHz, CDCI3) δ 7.70-7.58 (m, 4H), 7.27-7.23 (m, 1 H), 6.75-6.70 (m, 2H), 4.05 (q, 2H), 1.38 (t, 3H).
Part B:
Compound 2284 was prepared according to the procedure in J. Org. Chem.
1992, 57, 4521-4527.
Example 52E:
Figure imgf000648_0001
Part A:
To a mixture of compound 841 (2.96 g, 10 mmol), pyrrole-2-carbonitrile (1.1 g, 12 mmol) and 1 ,10-phenanthroline (1.17 g, 6.5 mmol) in DMA (10 mL) was added cesium carbonate (6.52 g, 20 mmol) and Cul (0.42 g, 2.2 mmol) and the mixture was heated at 150 °C for 18 hours. The mixture was filtered and rinsed with EtOAc (100 mL). The filtrate was washed with water (2 x 10 mL) and brine (10 mL), dried over MgS04 and concentrated. Purification by column chromatography (Si02, 10% EtOAc/hexane followed by 20% EtOAc/hexane) afforded compound 2285 as an oil (0.17 g, 5.7%).
Part B:
A mixture of compound 2285 (0.17 g, 0.57 mmol) and aq. 1M LiOH (1 mL, 1 mmol) in dioxane (3 mL) was stirred at room temperature for 18 hours. The reaction mixture was concentrated and added water (2 mL) and extracted with CH2Cl2 (2 x 10 mL). The organic layer was dried over MgS04, filtered and concentrated. Purification by column chromatography (Si02, 1% MeOH/ CH2CI2 followed by 5% MeOH/ CH2CI2) afforded compound 2286 as an oil (0.085 g, 71%).
Example 52F:
H N' γ OMe
Figure imgf000649_0001
2287 2288 2289
Part C Part Part E
Figure imgf000649_0004
Figure imgf000649_0002
Figure imgf000649_0003
2290 2291 2292
Figure imgf000649_0005
Part A:
To a solution of compound 2287 (20.6 g, 136.4 mmol) in CH2CI2 (200 mL) was added triethylamine (27.5 g, 272.8 mmol) and the reaction mixture was cooled to 0 °C, and (Boc)20 (32.7 g, 150 mmol) was added. The mixture was stirred at 0 °C for 10 minutes and then warmed to room temperature and stirred for 4 hours. The reaction mixture was washed with aq. sat. NH CI (100 mL) and dried over MgS04, filtered and concentrated. Purification by column chromatography (Si02, 10% EtOAc/hexane followed by 15% EtOAc/hexane) afforded the compound 2288 as white solid (32.7 g, 95%).
Part B:
To a solution of compound 2288 (3.95 g, 15.7 mmol) in CH2CI2 (200 mL) at - 78 °C was added a solution of 1M BBr3/CH2CI2 (37 mL) dropwise via a dropping funnel over 15 minutes. The reaction mixture was stirred at -78 °C for 2 hours, then poured carefully into MeOH (100 mL) and concentrated to a brown tar (3.6 g). Trituration with ether (3 x 20 mL) and concentrated afforded compound 2289 as brown tar which was used in next reaction without further purification.
Part C:
The crude compound 2289 was suspended in CH2CI2 (100 mL) and triethylamine (8.8 mL, 63 mmol) was added. The reaction mixture was cooled to 0 °C and (Boc)20 (3.77 g, 17.3 mmol) was added. The mixture was stirred at room temperature for 1 hour. The reaction mixture was then washed with water (100 mL), sat. NaHC03 (25 mL) and brine (10 mL). The organic layer was dried over MgS04, filtered and concentrated. Purification by column chromatography (Si02, 10% EtOAc/hexane followed by 15% EtOAc/hexane and 20% EtOAc/hexane) afforded compound 2290 as an oil (1.85 g, 62% over two steps).
Part D:
To a solution of compound 2290 (0.45 g, 1.9 mmol) in CH2CI2 (10 mL) was added a solution of NBS (0.34 g, 1.9 mmol) in CH2CI2 (10 mL) via a dropping funnel. Stirred at room temperature for 3.5 hours and then washed with aq. 10% H2S04 (10 mL). The organic layer was dried over MgS04, filtered and concentrated. Purification by column chromatography (Si02, 100% CH2CI2 followed by 1 % EtOAc/CH2CI2) afforded compound 2291 (0.13 g, 21%).
Part E:
To a solution of compound 2291 (0.22 g, 0.7 mmol) in acetone (6 mL) was added K2C03 (0.288 g, 2.1 mmol) followed by a solution of methyl iodide (0.065 mL, 1.05 mmol) and the reaction mixture was stirred at room temperature for 18 hours. The mixture was filtered and concentrated to dryness. The residue was dissolved in EtOAc (20 mL) and washed with water (2 mL) followed by brine (2 mL). The organic layer was dried over MgS04, filtered and concentrated to afford the compound 2292 (0.2 g, 86%).
Part F:
Compound 2293 was prepared following the procedure described in Example 52E, Part A.
Part G:
Compound 2294 was prepared from compound 2293 via TFA deprotection as described in Example 1.
Example 52G:
Figure imgf000651_0001
2292 2295
Part A:
Compound 2295 was prepared from compound 2292 2293 via TFA as described in Example 1.
Examples 52H:
H2N
p",c
Figure imgf000651_0002
Figure imgf000651_0003
Part A:
Compound 2289 was converted to compound 2296 via the procedure described in Example 52F Part C and using trifluoroacetic anhydride in place of Boc20.
Part B,C,D:
Compounds 2297, 2298 and 2299 were prepared following the procedures described in Example 52F, Part D, E and F using benzyl bromide and cyclopropyl bromide in place of methyl iodide.
Part E:
Compound 2300 were prepared from compound 2299 following the procedures described in Example 52E, Part B. Example 521:
partB
Figure imgf000652_0001
Figure imgf000652_0002
Part A:
A mixture of compound 2299 (0.27 g, 0.69 mmol), 10% Pd-C (0.18 g) and EtOH (25 mL) was stirred under hydrogen atmosphere at 55 psi for 18 hours. Filtered through celite and rinsed with CH2CI2 and concentrated to afford the desired compound 2301 (0.21 g, 100%).
Part B,C:
Compounds 2302 and 2303 were prepared following the procedures described in Example 52H, Part C and E and using 1-bromo-2-butyne as the alkylating reagent.
Example 52 J:
Figure imgf000652_0003
Part A:
A mixture of compound 2304 (60 mg), 10% Pd-C (30 mg) and MeOH (10 mL) was stirred at room temperature under hydrogen atmosphere for 18 hours. Filtered and rinsed with CH2CI2 and concentrated to afford the desired product 2305 (50 mg). Example 52K:
Figure imgf000653_0001
Part A:
Compound 2307 was prepared from compound 2306 following the procedure described in Example 52F, Part B.
Example 52L:
Figure imgf000653_0002
2307 2308
Part A:
To a mixture of 2307 (0.065 g, 0.18 mmol) in acetone (10 mL) was added K2C03 (0.082 g, 0.59 mmol) followed by benzyl bromide (0.022 mL, 0.19 mmol) and the reaction mixture was refluxed for 18 hours. DMF (0.5 mL) was added to the reaction mixture and heated at 70 °C for 18 hours. The reaction mixture was cooled to room temperature and EtOAc (5 mL) was added, then was washed with water (2 x 1 mL) followed by brine (1 mL). The organic layer was dried over MgS0 , filtered and concentrated. Purification by column chromatography (Si02, 5% MeOH (NH3)/CH2CI2) afforded 2308 (0.019 g, 23%).
Example 52M:
Figure imgf000654_0001
Part A:
To a solution of 5-bromopicoline (20 g, 116 mmol) in chloroform (100 mL) at 0 °C was added portionwise 77% mCPBA (28.7 g, 128 mmol) over 20 min. The reaction mixture was allowed to stir at room temperature overnight, after which time the solid was filtered off, the filtrate was washed with saturated Na2C03, dried over Na2S04, and concentrated to afford 2309 (17.83 g, 82%) as a white solid. 1H NMR (400 MHz, CDCI3) δ 8.39 (s, 1 H), 7.29 (d, J = 8.0 Hz, 1 H), 7.12 (d, J = 8.3 Hz, 1H), 2.47 (s, 3H).
Part B:
Compound 2309 (15 g, 80 mmol) was cooled to 0 °C, and trifluoroacetic anhydride (35 mL, 250 mmol) was slowly added under argon. The mixture was allowed to stir at room temperature for 30 min, then at 55 °C for 30 min after which it was cooled to 0 °C in an ice bath. NaHC03 aqueous solution (10%, 150 mL) was slowly added with caution. After the completion of addition, the mixture was allowed to stir at room temperature for 6 h. The solution was extracted with dichloromethane, dried over Na2S0 , and concentrated. The residue was purified by flash column chromatography on silica gel (EtOAc/hexane 50:50), affording product 2310 as a white solid (9.2 g, 61 %). 1H NMR (400 MHz, CDCI3) δ 8.61 (d, J = 1.9 Hz, 1 H), 7.80 (dd, J = 7.9, 2.4 Hz, 1 H), 7.19 (dd, J = 8.3, 0.9 Hz, 1 H), 4.73 (s, 2H), 3.37 (br s, 1 H).
Part C:
To a solution of 2310 (9.0 g, 47.86 mmol) in toluene (40 mL) was added PBr (5 mL, 52.6 mmol) dropwise. After the completion of addition, the mixture was stirred at 100 °C for 2 h. It was cooled to room temperature; the solid was collected by filtration to afford 2311 as a yellow solid (HBr salt, 14.83 g, 93%). 1H NMR (400 MHz, DMSO-d6) δ 9.74 (br, 1 H), 8.66 (d, J = 1.8 Hz, 1 H), 8.06 (dd, J = 8.4, 1.4 Hz, 1H), 7.19 (dd, J = 8.3, 0.9 Hz, 1 H), 4.67 (s, 2H).
Part D:
To a solution of 2311 (14.83 g, 44.7 mmol) in DMF at 0 °C was slowly added Cs2C03 (29.1 g, 89.4 mmol) and potassium phthalimde (9.1 g, 49.2 mmol). The reaction mixture was allowed to stir at room temperature for 6h after which the solvent was evaporated to dryness. The resulting solid was washed with H20. The solid cake was recrystallized in hot EtOH, affording compound 2312 (7.64 g, 54%) as a white solid. 1H NMR (400 MHz, DMSO- de) δ 8.54 (d, J = 1.9 Hz, 1 H), 8.00 (dd, J = 8.4, 2.3 Hz, 1 H), 7.90-7.83 (m, 4H), 7.41 (d, J = 7.8 Hz, 1 H), 4.88 (s, 2H).
Part E:
A mixture of Pd(OAc)2 (45 mg, 0.2 mmol), 2-bis(tert-butyl)phosphino-biphenyl (120 mg, 0.4 mmol), compound 2312 (3.17 g, 10 mmol), 2- trifluoromethoxyphenyl boronic acid (2.47 g, 12 mmol) and KF (2.1 g, 36 mmol) was charged into a 250-mL round bottom flask. Then THF (30 mL) was added under argon, and the reaction mixture was allowed at stir at 50 °C overnight, after which it was cooled to room temperature, filtered through celite and concentrated. Flash column chromatography on silica gel (EtOAc/hexane (5:95) provided compound 2313 (3.27 g, 82%) as a white solid. 1H NMR (400 MHz, CDCI3) δ 8.59 (d, J = 1.5 Hz, 1 H), 7.91-7.89 (m, 2H), 7.77-7.73 (m, 3H), 7.44-7.34 (m, 5H), 5.08 (s, 2H).
Part F:
Compound 2314 was prepared according to the same procedure as in Example 52M Part A. HPLC-MS tR = 1.74 min (UV 4 nrn); mass calculated for formula C21H13 F3N204414.1 , observed LCMS m/z 415.0 (M+H).
Part G: To a solution of compound 2314 (210 mg, 0.51 mmol) in EtOH (10 mL) was added with hydrazine (0.5 mL). The solution was refluxed for 3 h, then cooled to room temperature. After removing the precipitate by filtration, the solution was concentrated. The resulting residue was extracted with Et20 and concentrated, affording compound 2315 as a pale yellow oil (160 mg, 91 %). HPLC-MS tR = 1.74 min (UV25 „m); mass calculated for formula Cι Hn F3N202 284.1 , observed LCMS m/z 285.1 (M+H).
Example 52N:
Figure imgf000656_0001
Part A:
To compound 2314 (200 mg, 0.48 mmol) in acetonitrile (1 mL) was added Et3N (0.094 mL, 0.672 mmol) and Me3SiCN (0.225 mL, 1.69 mmol) subsequently under argon. The reaction mixture was stirred at 80 °C for 48 h, after which it was cooled to room temperature and concentrated in vacuo. The residue was dissolved in EtOAc, washed with saturated NaHC03, brine, dried (Na24), and concentrated. Flash column chromatography (EtOAc/hexane 40:60) afforded compound 2316 (130 mg, 64%). HPLC-MS tR = 2.06 min (UV25 nm); mass calculated for formula C222F3N303423.1 , observed LCMS m/z 424.0 (M+H).
Part B:
Compound 2317 was prepared using the procedure described in Example 52M Part G. 1H NMR (400 MHz, CDCI3) δ 7.77 (d, J = 8.1 Hz, 1 H), 7.60 (d, J = 8.0 Hz, 1 H), 7.47-7.42 (m, 4H), 4.12 (s, 2H). 1.98 (br s, 2H).
Example 520:
Figure imgf000656_0002
Part A:
Phosphorous trichloride (0.330 mL, 3.62 mmol) was added to a solution of 2314 (300 mg, 0.72 mmol) in CHCI3 (1 mL). The reaction mixture was stirred at 60 °C overnight, after which it was cooled to room temperature and concentrated in vacuo. The residue was dissolved in EtOAc, washed with saturated NaHC03, brine, dried (Na2S04), and concentrated. Flash column chromatography (EtOAc/hexane 30:70) afforded 2318 (180 mg, 58%). HPLC- MS tR = 2.33 min (UV254 nm); mass calculated for formula C2ιHι2CIF3N203 432.1 , observed LCMS m/z 433.0 (M+H).
Part B:
Compound 2319 was prepared using the procedure described in Example 52M Part G. HPLC-MS tR = 1.26 min (UV254 nm); mass calculated for formula C13Hi0CIF3N2O 302.0, observed LCMS m/z 303.0 (M+H).
Example 52P:
Figure imgf000657_0001
Part A:
3-Bromo-2-chloro-6-picoline (1.0 g, 4.84 mmol) and NBS (1.20 g, 6.78 mmol) were dissolved in CCI (20 mL). The solution was briefly flushed with argon, then AIBN (16 mg, 0.097 mmol) was added. The mixture was stirred at room temperature overnight, after which the solvent was evaporated in vacuo. The residue was purified by flash column chromatography (EtOAc/hexane 5:95) providing compound 2320 as a white solid (1.28 g, 92%). 1H NMR (400 MHz, CDCI3) δ 8.01 (d, J = 8.2 Hz, 1 H), 7.65 (d, J = 8.2 Hz, 1 H), 4.19 (s, 2H).
Part B: Compound 2321 was prepared according to the procedure described in Example 52M, Part D. 1H NMR (400 MHz, DMSO-d6) δ 8.19 (d, J = 8.0 Hz, 1 H), 7.92-7.84 (m, 4H), 7.41 (d, J = 8.0 Hz, 1 H), 4.86 (s, 2H).
Part C:
Compound 2322 was prepared according to the procedure described in Example 52M, Part G. 1H NMR (400 MHz, CDCI3) δ 7.87 (d, J = 8.0 Hz, 1 H), 7.41 (d, J = 8.1 Hz, 1 H), 3.93 (s, 2H), 1.78 (br s, 2H).
Part D:
Compound 2322 (370 mg, 1.67 mmol) in THF (10 mL) was added dropwise into the solution of Boc20 (437 mg, 2.0 mmol) in THF (10 mL), followed by DIEA (300 mL). The mixture was stirred at room temperature overnight and concentrated. Column chromatography on silica gel (EtOAc/hexane 20:80) provided compound 2323 (450 mg, 84%) as a white solid. 1H NMR (400 MHz, CDCI3) δ 7.89 (d, J = 7.5 Hz, 1 H), 7.12 (d, J = 8.2 Hz, 1 H), 5.35 (br s, 1H), 4.37 (d, J = 5.9 Hz, 2H), 1.47 (s, 9H).
Part E:
Sodium (4 pellets) was dissolved in MeOH (15 mL). The mixture was stirred at room temperature for 10 min until all the sodium had reacted. To this solution was added compound 2323 (450 mg, 1.4 mmol) in MeOH (5 mL) via syringe. The reaction mixture was stirred at 60 °C for 48 h and concentrated. The residue was dissolved with EtOAc and 1N NH4CI solution, washed with brine, dried over Na2S04, and concentrated. Column chromatography (Si02, EtOAc/hexane 20:80) afforded compound 2324 as a white solid (350 mg, 79%). 1H NMR (400 MHz, CDCI3) δ 7.43 (d, J = 7.3 Hz, 1 H), 6.73 (d, J = 7.5 Hz, 1 H), 5.26 (br s, 1 H), 4.32 (d, J = 5.1 Hz, 2H), 4.02 (s, 3H), 1.48 (s, 9H).
Part F:
Compound 2325 was prepared according to the same procedure as in
Example 52M Part E for the Suzuki coupling. HPLC-MS tR = 2.35 min (UV254 nm); mass calculated for formula CιgH21F3N204 398.2, observed LCMS m/z 399.1 (M+H).
Part G:
Compound 2325 was deprotected with TFA in CH2CI2. HPLC-MS tR = 1.12 min (UV254 nm); mass calculated for formula Cι43F3N202 298.1 , observed LCMS m/z 299.0 (M+H).
Example 52Q:
EtOO
Figure imgf000659_0001
Part A:
Ethyl-4-hydroxycyclohexylcarboxylate (2327) (cis/trans mixture) (4.3 g, 25 mmol), 2-trifluoromethylbenzimidazole (4.65 g, 25 mmol), and triphenylphosphine (9.8 g, 37.5 mmol) were dissolved in dioxane (60 mL). The resulting solution was treated with diethyldiazodicarboxylate (6.5 g, 37.5 mmol), dropwise, over a period of 1 h and stirred for 16h. The reaction mixture was diluted with ethyl acetate (200 mL) and washed in sequence with water (2 X 100 mL) and brine (1 X 100 mL). The organics were dried and concentrated to yield a crude which was subjected to silica-gel column chromatography (10% ethyl acetate/hexane) to yield pure 2328 (0.2 g, 0.6 mmol).
Part B:
LiAIH4 was added to dry THF (40 mL) and the mixture was then cooled to 0 °C. Then ester 2328 (0.2 g, 0.6 mmol) in THF (5 mL) was added to the mixture dropwise. The reaction mixture was allowed to warm to ambient temperature and stirred for 48 h, then cooled back to 0 °C and quenched by the sequential addition of 1 mL of water, 2 mL of 0.5 N NaOH, and 1 mL of water. The resulting mixture was stirred vigorously for 2 h and then filtered through Celite. The filtrate was concentrated to yield pure 2329 as an oil (0.2 g, 0.6 mmol).
Part C:
Alcohol 2329 (0.2 g, 0.6 mmol) and triethylamine (0.18 g, 1.8 mmol) were dissolved in CH2CI2 (200 mL) and cooled to 0 °C. The cooled solution was stirred, and CH3S02CI (as a CH2CI2 solution; 0.2 g, 1.8 mmol, 5 mL CH2CI2) was added dropwise, and the stirring was continued for 24 h. The reaction mixture was then washed two times with 50 mL of water and two times with 50 mL of brine. The organic and aqueous phases were separated, and the organic phase was dried, and concentrated to provide pure product 2330 as an oil (0.21 g).
Part D:
The mesylate 2330 was dissolved in DMF (10 mL) and treated with NaN3 (0.23 g, 1.8 mmol), and the mixture was stirred vigorously for 36-72 h. The reaction mixture was then diluted with 100 mL water and extracted with ethyl acetate (2 x 100 mL). The organic phases were combined, dried, and concentrated to yield pure azide 2331 (0.19 g, 87%).
Part E:
The azide 2331 (0.19 g, 0.6 mmol)) and triphenylphosphine (0.15 g, 0.6 mmol) were dissolved in 5 mL of THF, and then water (0.6 mL, 33.3 mmol) was added. The resulting mixture was stirred vigorously for 16-24 h. The solvent was removed and the crude amine 2332 was taken to the next step without further purification.
The following compounds were prepared using previously described procedures.
Figure imgf000660_0001
Figure imgf000661_0001
Figure imgf000662_0001
Figure imgf000663_0001
Figure imgf000664_0001
Figure imgf000665_0002
Example 53:
Example 53A:
Figure imgf000665_0001
Part A:
Compound 2361 was prepared using procedures described in Adamczyk, M. et al. Synth. Commun. 2002, 32, 3199-3205. According to a modification of a procedure by Neidigh, K. A. et al. (J. Chem. Soc. Perkin Trans. 1 1998, 2527-2531) a mixture of ketone 2361 (1.56 g, 7 mmol), titanium (IV) isopropoxide (4.1 mL, 14 mmol), methylamine hydrochloride (0.94 g, 14 mmol) and triethylamine (1.95 mL, 14 mmol) in ethanol (15 mL) was stirred at rt overnight. Then sodium borohydride (0.4 g, 10.5 mmol) was added and the resulting mixture was stirred at rt for 8 h. The reaction mixture was then poured into aqueous ammonium hydroxide (40 mL), the resulting white inorganic precipitate was filtered off, and washed several times with dichloromethane. The organic layer was separated, and the aqueous phase was extracted with dichloromethane. Combined dichloromethane extracts were washed with brine, concentrated and chromatographed (Si02, 5% methanol/dichloromethane) to give 2362 (1.03 g, 62%). 1H NMR (400 Mhz, CDCI3) δ 7.30-7.17 (m, 5H), 4.31-4.30 (d, 1H), 3.78-3.62 (m, 2H), 3.57-3.47 (m, 2H), 2.97-2.70 (m, 3H), 2.43 (s, 3H), 1.27- 1.20 (m, 6H).
Part B:
Compound 2005 was prepared using procedures described in Example 27. Compound 2363 was prepared using the coupling conditions described in Example 1. HPLC-MS tR = 2.42 min (UV254 nm); mass calculated for formula C34H45FN406 624.3, observed LCMS m/z 625.2 (M+H).
Part C:
Compound 2364 was prepared using the cyclization conditions described in Example 29. HPLC-MS tR = 1.66 min (UV254 nm); mass calculated for formula C30H34FN5O3 531.3, observed LCMS m/z 532.2 (M+H).
Part D:
Compound 2364 was deprotected using procedures described in Example 29.
Purification by prep-LC and conversion to a hydrochloric salt afforded 2365 as an off-white solid. HPLC-MS tR = 3.54 min (10 min; UV2 4 nm); mass calculated for formula C27H30FN5O3 491.2, observed LCMS m/z 492.1
(M+H). Example 53B:
Figure imgf000667_0001
Part B CN
Figure imgf000667_0002
Part A:
Compound 2366 was prepared using procedures described in Waldvogel, E. et al. Helv. Chim. Acta. 1997, 80, 2084-2099.
Compound 2367 was prepared using the coupling conditions described in
Example 1. HPLC-MS tR = 1.88 min (UV254 nm); mass calculated for formula
C26H35FN406 518.2, observed LCMS m/z 519.2 (M+H).
Part B:
Compound 2368 was prepared using the cyclization conditions described in Example 29. HPLC-MS tR = 1.40 min (UV254 nm); mass calculated for formula C24H30FN5O3 455.2, observed LCMS m/z 456.1 (M+H).
Part C:
Compound 2368 was deprotected using procedures described in Example 29.
Purification by prep-LC and conversion to a hydrochloric salt afforded 2369 as an off-white solid. HPLC-MS tR = 2.99 min (10 min; UV254 nm); mass calculated for formula C21 H26FN503 415.2, observed LCMS m/z 416.2
(M+H).
Example 53C:
Figure imgf000667_0003
Part A: Compound 2370 was prepared using previously described procedures in Example 29. HPLC-MS tR = 1.30 min (UV254 nm); mass calculated for formula C23H28FN503 441.2, observed LCMS m/z 442.1 (M+H).
According to a modification of a procedure by Miller, R. D. et al. (Chem. Mater. 1994, 6, 1023-1032) to an ice cold solution of compound 2370 (104 mg, 0.23 mmol) and pyridine (0.039 mL, 0.48 mmol) in dichloromethane (3 mL) was added dropwise a solution of bromine (0.016 mL, 0.31 mmol) in dichloromethane (1 mL) and the resulting solution was stirred at 0 °C for 20 min. The reaction mixture was quenched with 10% sodium thiosulfate solution, and extracted with dichloromethane. Combined organic extracts were dried, concentrated and chromatographed (Si02, gradient elution 50% to 80% ethyl acetate/hexane, ethyl acetate followed by 5% methanol/ethyl acetate) to give 2371 as a white solid (35 mg, 37% based on reacted starting material). HPLC-MS tR = 1.94 min (UV2 nm); mass calculated for formula C23H27BrFN503 519.1 , observed LCMS m/z 520.0 (M+H).
Part B:
Compound 2371 was deprotected using procedures described in Example 29. Purification by prep-LC and conversion to a hydrochloric salt afforded 2372 as a white solid. HPLC-MS tR = 3.1 min (10 min; UV25 nm); mass calculated for formula C20H23BrFN5O3 479.1 , observed LCMS m/z 480.0 (M+H).
Example 53D:
Figure imgf000668_0001
Part A:
Compound 2373 was prepared using previously described procedures in
Examples 1 and 27. To a solution of 2373 (300 mg, 0.71 mmol) in methanol (7 mL) was added hydrazine monohydrate (0.35 mL, 7.1 mmol) and the resulting mixture was heated at 70 °C in a sealed tube overnight. The mixture was then cooled to rt, concentrated, diluted with ethyl acetate, washed with water, brine and concentrated to give 2374 (220 mg, 73%).
1H NMR (400 MHz, CDCI3) δ 7.27-7.24 (m, 1 H), 7.22-7.17 (m, 1 H), 7.00 (t, 1H, NH), 6.99-6.96 (m, 2H), 4.62-4.57 (dd, 2H), 3.51-3.48 (d, 2H), 3.38-3.24 (m, 2H), 2.80-2.74 (m, 2H), 1.88-1.84 (d, 2H), 1.79-1.69 (m, 1H), 1.61-1.50 (m, 2H), 1.53 (s, 3H), 1.52 (s, 3H). HPLC-MS tR = 1.49 min (UV254 „m); mass calculated for formula C20H26FN5O4 419.2 observed LCMS m/z 420.1 (M+H).
Part B:
According to a modification of a procedure by Stocks, M. J. et al. (Org. Lett. 2004, 17, 2969-2971 ) to a solution of hydrazide 2374 (110 mg, 0.26 mmol) in acetonitrile (1 mL) was added N,N-dimethylformamide dimethyl acetal (0.035 mL, 0.26 mmol) and the mixture was heated at 50 °C for 50 min. Then methylamine (0.18 mL of 2 M solution in THF, 0.36 mmol) was added, followed by acetic acid (1 mL) and the resulting mixture was heated at 80 °C overnight. The reaction mixture was then concentrated and chromatographed (Si02, 9:1 :90 methanol:triethylamine:dichloromethane) to give 2375. 1H NMR (400 MHz, CDCI3) δ 8.42 (s, 1 H), 7.27-7.24 (m, 1 H), 7.21-7.18 (m, 1 H), 7.02- 6.97 (m, 1 H), 6.92 (t, 1H, NH), 5.48-5.37 (dd, 2H), 3.51-3.47 (m, 2H), 3.36- 3.23 (m, 2H), 2.80-2.74 (m, 2H), 1.88-1.81 (m, 2H), 1.76-1.70 (m, 1H), 1.58- 1.54 (m, 2H), 1.58 (s, 3H), 1.43 (s, 3H). HPLC-MS tR = 1.58 min (UV254 nm); mass calculated for formula C22H27FN603 442.2 observed LCMS m/z 443.2 (M+H).
Part C:
Compound 2375 was deprotected with 4 M HCI in dioxane (3 mL) and water (0.3 mL) at 55 °C for 1 hour. Purification by prep-LC and conversion to a hydrochloric salt afforded 2376 as an off-white solid (20 mg). HPLC-MS tR = 1.23 min (UV254 nm); mass calculated for formula C19H23FN603 402.2, observed LCMS m/z 403.1 (M+H).
Example 53E:
Figure imgf000670_0001
Part A:
(tøtτf-Butoxycarbonylmethylene)triphenylphosphorane (15.1 g, 40 mmol) was added portionwise to a solution of 2-pyridinecarboxaldehyde (4.28 g, 40 mmol) in THF (60 mL) at 0 °C over a period of 5 min. The reaction mixture was allowed to stir at room temperature overnight. The solution was concentrated, extracted with hexane (120 mL). After filtration to remove the solid, the filtrate was concentrated to give a yellow oil. Purification by flash chromatography on silica gel (EtOAc/hexane 15:85) yielded the desired product 2377 (4.5 g, 55%) as slightly yellow liquid. 1H NMR (400 MHz, CDCI3) δ 8.62 (d, J = 4.9 Hz, 1 H), 7.69 (td, J = 6.6, 1.7 Hz, 1 H), 7.58 (d, J = 15.5 Hz, 1 H), 7,41 (dd, J = 7.8, 0.6 Hz, 1 H), 7.23 (m, 1 H), 6.82 (d, J = 16.1 Hz, 1 H), 1.54 (s, 9H).
Part B:
Compound 2377 (4.5 g, 21.92 mmol) was dissolved in dry DCM (40 mL) and cooled to 0 °C, then 77% m-CPBA (5.41 g, 24.12 mmol) was added in three portions over 5 min. The reaction mixture was allowed to stir overnight at room temperature. It was poured into saturated aqueous NaHC03 (100 mL) and the product was extracted into DCM. The organic layer was washed with water and brine and dried (Na2S04). Flash chromatography on silica gel (MeOH/DCM 5:95) afforded compound 2378 as a pale yellow solid (3.2 g, 66%). 1H NMR (400 MHz, CDCI3) δ 8.24 (m, 1 H), 7.96 (d, J = 16.0 Hz, 1 H), 7.51 (m, 1 H), 7.22 (m, 1 H), 6.85 (d, J = 16.0 Hz, 1 H), 1.55 (s, 1 H).
Part C:
A 250-mL round bottom flask filled with H20 (30 mL) was cooled to 0 °C in an ice bath. Reagents K3[Fe(CN)6] (9.88 g, 30 mmol), K2C03 (6.91 g, 50 mmol), and MeS02NH2 (0.95 g, 10 mmol) were subsequently added, followed by K2[Os02(OH)4] (14.7 mg, 0.04 mmol), (DHQ)PHAL (233.7 mg, 0.3 mmol), compound 2378 (2.21 g, 10 mmol) and f-BuOH (20 mL). The reaction mixture was stirred at 0 °C for 24 h. The solid was filtered off and washed with excess EtOAc. The organic layer was separated. The aqueous solution was concentrated to dryness, and the resulting solid was extracted with CH2CI2. The above EtOAc and CH2CI2 solutions were combined and concentrated. Flash chromatography on silica gel (MeOH/CH2CI2 10:90) provided compound 2379 as a white solid (2.2 g, 86%). Recrystallization from EtOAc afforded analytically pure material (1.51 g, 52% yield). 1H NMR (400 MHz, CDCI3), δ 8.24 (d, J = 6.5 Hz, 1 H), 7.54 (dd, J = 7.7, 2.0 Hz, 1 H), 7.39 (t, J = 7.5 Hz, 1 H), 7.30 (m, 1 H), 5.41 (d, J = 2.6 Hz, 1 H), 4.69 (d, J = 2.7 Hz, 1 H), 1.54 (s, 9H).
Part D:
Compound 2379 (1.0 g, 3.9 mmol) in CH2CI2 (10 mL) was treated with TFA (4 mL). The solution was allowed to stir at room temperature for 2 h, and then concentrated to afford compound 2380 as a white solid (750 mg, 96%). 1H
NMR (400 MHz, DMSO-d6) δ 8.20 (dd, J = 6.3, 1.2 Hz, 1 H), 7.54 (dd, J = 6.4,
2.2 Hz, 1 H), 7.37 - 7.32 (m, 2H), 5.38 (d, J = 2.1 Hz, 1 H), 4.62 (d, J = 2.3 Hz,
1 H).
Part E:
Compound 2381 was prepared using the coupling conditions described in Example 1. Purification by prep-LC afforded 2381 as a white solid (63 mg). HPLC-MS tR = 1.30 min (UV25 nm); mass calculated for formula C2ιH23FN404, 414.2, observed LCMS m/z 415.1 (M+H). Part F:
To compound 2381 (25 mg, 0.06 mmol) in EtOH (5 mL) was added with saturated NH CI aqueous solution (5 mL) and indium (16.6 mg, 0.072 mmol). The reaction mixture was allowed to reflux for 24 h. After cooling to room temperature, it was concentrated and extracted with DCM. The DCM extract was filtered and concentrated. Purification by column chromatography on silica gel (Et3N / MeOH / EtOAc 5:5:95) afforded compound 2382 as a white solid (8.5 mg, 35%). HPLC-MS tR = 1.16 min (UV254 nm); mass calculated for formula C21H23FN403, 398.2, observed LCMS m/z 399.1 (M+H).
Example 53F:
Figure imgf000672_0001
Part A:
Under argon, to a mixture of Pd2(dba)3 (384 mg, 0.42 mmol), tri(t- butyl)phosphinium tetrafluoroborate (243 mg, 0.84 mmol), and 2,6- dichloropyrizine (5.0 g, 33.56 mmol) were added sequentially NMP (10 mL), t- butyl acrylate (2.46 mL, 16.78 mmol) and Et3N (3.51 mL, 25.17 mmol). The reaction mixture was allowed to stir at 50 °C for 12 h. After cooling to room temperature, the mixture was diluted with EtOAc and water, filtered through a pad of celite, washed with water, brine, dried (Na2S04), and concentrated. Flash column chromatography on silica gel (EtOAc / hexane 15:85) afforded compound 2383 as a pale yellow solid (2.32 g, 57%). 1H NMR (400 MHz, acetone-ds) δ 8.84 (s, 1H), 8.68 (s, 1H), 7.61 (d, J = 15.7 Hz, 1H), 6.92 (d, J = 15.5 Hz, 1 H), 1.55 (s, 9H).
Part B:
A 50-mL round bottom flask filled with H20 (9 mL) was cooled to 0 °C in an ice bath. Reagents K3[Fe(CN)6] (1.29 g, 3.93 mmol), K2C03 (905 mg, 6.55 mmol), and MeS02NH2 (124.6 mg, 1.31 mmol) were subsequently added, followed by K2[Os02(OH)4] (2.0 mg, 0.005 mmol), (DHQ)PHAL (31.0 mg, 0.04 mmol), compound 2383 (315 mg, 1 mmol) and f-BuOH (6 mL). The reaction mixture was stirred at 0 °C for 36 h. Then EtOAc and H20 were added to dissolve the solid. The organic layer was separated and the aqueous layer was back extracted with EtOAc. The EtOAc extracts were combined, washed with water and brine, dried (Na2S04), and concentrated. Flash chromatography on silica gel (EtOAc/hexane 40:60) provided compound 2384 as a white solid (285 mg, 79%). 1H NMR (400 MHz, CDCI3) δ 8.68 (s, 1 H), 8.54 (s, 1 H), 5.13 (d, J = 2.3 Hz, 1 H), 4.55 (d, J = 2.9 Hz, 1 H), 1.55 (s, 9H). HPLC-MS t = 1.73 min (UV25 nm); mass calculated for formula CnHι5CIN204, 274.1 , observed LCMS m/z 275.0 (M+H).
Part C:
Compound 2384 (285 mg, 1.04 mmoi) in dioxane (5 mL) was treated with 4N HCI in dioxane (5 mL). After stirring at room temperature for 2 h, the solution was concentrated to afford compound 2385 (200 mg, 88%). 1H NMR (400 MHz, CDCI3) δ 8.70 (s, 1 H), 8.66 (s, 1 H), 5.00 (d, J = 2.6 Hz, 1 H), 4.31 (d, J = 2.2 Hz, 1 H), 1.55 (s, 9H).
Part D:
Compound 2386 was prepared using the coupling conditions described in Example 1. Purification by prep-LC afforded 2386 as a white solid (42 mg). HPLC-MS t = 1.55 min (UV25 nm); mass calculated for formula C2oH2ιCIFN503, 433.1 , observed LCMS m/z 434.0
Part E:
Compound 2386 (20 mg, 0.046 mmol) along with 10% Pd/C (20 mg) in EtOAc (10 mL) was stirred at room temperature for 12 h under hydrogen atmosphere (1 atm). The solution was filtered through celite and concentrated. Purification by prep-LC afforded 2387 as a white solid (15 mg, 81 %). HPLC- MS tR = 1.35 min (UV254 nm); mass calculated for formula C2oH22FN5θ3, 399.2, observed LCMS m/z 400.1 Example 53G:
Figure imgf000674_0001
Part A:
To compound 2385 (150 mg, 0.69 mmol) in acetic acid (2 mL) was added acetic anhydride (0.5 mL). The reaction mixture was stirred at room temperature for 2 h, and then concentrated. The residue was dissolved in EtOAc, washed with H20, brine, dried over Na24, and concentrated, giving rise to compound 2388 as oily solid (180 mg, 86%). HPLC-MS tR = 1.10 min (UV254 nm); mass calculated for formula C-ιιHιιCIN206 302.0, observed LCMS m/z 303.0 (M+H).
Part B:
To compound 2388 (180 mg, 0.59 mmol) and HATU (269 mg, 0.71 mmol) in DMF (4 mL) was added isoindoline (84 mg, 0.71 mmol). The reaction mixture was stirred at room temperature for 12 h, and concentrated. The residue was dissolved with EtOAc and water, washed with saturated bicarbonate solution, and brine, dried over Na2S04, and concentrated. Purification by column chromatography on silica gel (EtOAc/hexane 60:40) afforded the desired product 2389 as a pale yellow solid (150 mg, 63%). 1H NMR (400 MHz, CDCI3) δ 8.67 (s, 1 H), 8.51 (s, 1 H), 7.29 (m, 4H), 6.39 (d, J = 7.1 Hz, 1 H), 5.93 (d, J = 6.7 Hz, 1 H), 5.21 (d, J = 5.4 Hz, 1 H), 5.05 (d, J = 4.1 Hz, 1 H), 4.78 (d, J = 5.6 Hz, 1 H), 4.52 (d, J = 4.7 Hz, 1 H), 2.18 (s, 3H), 2.16 (s, 3H). HPLC-MS tR = 1.70 min (UV25 nm); mass calculated for formula C198CIN3θ5 403.1 , observed LCMS m/z 404.0 (M+H).
Part C: A 4-mL vial was charged with Pd2(dba)3 (2.3 mg, 0.0025 mmol), tri(t- butyl)phosphonium tetrafluoroborate (1.5 mg, 0.0025 mmol), compound 2389 (20 mg, 0.05 mmol), phenylboronic acid (9.1 mg, 0.075 mmol) and potassium fluoride (13 mg, 0.225 mmol). Under argon, dioxane (1 mL) was added via syringe. The vial was sealed with a Teflon coated screw cap and placed into an oil bath preheated to 80 °C. The reaction mixture was allowed to stir at 80 °C for 12 h. LC-MS indicated the reaction was completed. After cooling to room temperature, the reaction mixture was filtered through celite with the aid of EtOAc. The filtrate was washed with saturated NaHC03, brine, dried over Na2S04, and concentrated. The resulting solid (21 mg) was used for next step without purification. HPLC-MS tR = 1.89 min (UV2 4 nm); mass calculated for formula C25H23N3O5445.1 , observed LCMS m/z 446.0 (M+H).
Part D:
Compound 2390 (21 mg) was deprotected using procedures described in Example 2. Purification by prep-LC afforded 2391 as an off-white solid (8.3 mg). HPLC-MS tR = 4.02 min (10 min method) (UV25 nm); mass calculated for formula C21H19N305361.1 , observed LCMS m/z 362.0 (M+H).
Example 53H:
Figure imgf000675_0001
Part A:
To compound 2389 (50 mg, 0.12 mmol) in NMP (1 mL) was added 4-N- phenylpipyzine (29 mg, 0.18 mmol) and DIEA (0.032 mL, 0.18 mmol). The reaction mixture was allowed to stir at 80 °C for 16 h. After cooling to room temperature, the solution was diluted with EtOAc, washed with water and brine, dried over Na2S04, and concentrated. The resulting oily solid (52 mg) was utilized for next step without further purification. HPLC-MS tR = 1.96 min (UV254 nm); mass calculated for formula C29H3iN505 529.2, observed LCMS m/z 530.2 (M+H). Part B:
Compound 2392 (52 mg) was deprotected using procedures described in Example 2. Purification by prep-LC afforded 2393 as an off-white solid (15 mg). HPLC-MS tR = 1.60 min (UV254 nm); mass calculated for formula C25H27N503 445.2, observed LCMS m/z 446.1 (M+H).
The following compounds were prepared using previously described procedures.
Figure imgf000676_0001
Figure imgf000677_0001
Example 54:
Figure imgf000678_0001
Part A:
Methyl pipecolinate hydrochloride (2.0 g, 11 mmol) was dissolved in THF (20 mL) and saturated sodium bicarbonate (20 mL) and cooled in an ice bath. Benzyl chloroformate (1.5 mL, 11 mmol) was added dropwise and the reaction was stirred at room temperature overnight. The reaction mixture was partitioned between ethyl acetate and water. The organic layer was washed with 1 N HCI, saturated sodium bicarbonate, water and brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 10% ethyl acetate/hexane to 15% ethyl acetate/hexane) afforded 2404 as a clear oil (2.9 g, 93%).
Part B and C:
Compounds 2405 and 2406 were prepared following the procedures described in Example 10B part A and part B. Compound 2406: 1H NMR (400 MHz, CDCI3) δ 7.40 - 7.30 (m, 5H), 6.11 (s, 1 H), 5.36 (m, 1 H), 5.18 (s, 2H), 3.72 (m, 1 H), 3.00 (m, 1 H), 2.33 (m, 1 H), 1.90 - 1.40 (m, 7H). HPLC-MS tR = 1.34min (UV254 nm); Mass calculated for C16H19N302S 317.4, observed LSMS m/z 318.1 (M+H).
Part D:
Compound 2406 (75 mg, 0.24 mmol) was stirred in 30% HBr in acetic acid (2 mL) for 1 hour at room temperature. The solvent was evaporated and the residue was dissolved in water and lyophilized to afford 2407 (66 mg) as a red solid.
The following compounds were prepared using previously described procedures.
Figure imgf000679_0001
Figure imgf000680_0001
Example 55:
Example 55A:
Figure imgf000681_0001
Part A:
To an ice cold solution of acid 2426 (1.0 g, 3.8 mmol) in 1 :1 methanol.'toluene (10 mL) was added dropwise TMS-diazomethane (3.2 mL of 2 M solution in hexane, 6.4 mmol) and the resulting solution was stirred at rt for 1 h. The reaction mixture was concentrated, partitioned between ethyl acetate and water and extracted with ethyl acetate. The combined organic extracts were washed with saturated sodium bicarbonate solution, brine, dried and evaporated to give ester 2427 as an off-white solid (0.907 g, 86%). 1H NMR (400 MHz, CDCI3) δ 7.43-7.28 (m, 4H), 5.53-5.46 (dd, 1 H), 4.86-4.69 (m, 2H), 3.77-3.75 (d, 3H), 1.54-1.49 (d, 9H).
Part B:
Chloroketone 2428 was prepared using procedures described in Example 10. 1H NMR (400 MHz, CDCI3) δ 7.46-7.30 (m, 4H), 5.65-5.48 (d of d, 1 H), 4.90- 4.71 (m, 2H), 4.42-4.06 (m, 2H), 1.57-1.52 (d, 9H).
Part C:
Compound 2429 was prepared using procedures described in Example 10. HPLC-MS tR = 1.51 min (UV254 nm); mass calculated for formula C17H21 N302S 331.1 , observed LCMS m/z 332.1 (M+H).
Part D:
Compound 2430 was prepared using procedures described in Example 10. HPLC-MS tR = 0.80 min (UV254 nm); mass calculated for formula C12H13N3S 231.1 , observed LCMS m/z 232.1 (M+H).
Example 55B: Part C
Figure imgf000682_0002
Figure imgf000682_0001
Part A:
According to a modification of a procedure by Mach, U. R. et al. (ChemBioChem 2004, 5, 508-518) to a mixture of 3-chlorophenethylamine 2431 (3.89 g, 25 mmol) and 2 M potassium hydroxide solution (35 mL) was added dropwise benzoyl chloride (3.48 mL, 30 mmol). Within minutes a white precipitate formed, it was filtered and washed thoroughly with water, dried (air) and recrystallized (ethanol) to give amide 2432 as a white solid (4.22 g, 65%). 1H NMR (400 MHz, DMSO-d6) δ 8.53 (t, 1 H, NH), 7.78-7.76 (d, 2H), 7.50-7.17 (m, 7H), 3.50-3.46 (m, 2H), 2.85 (t, 2H). HPLC-MS tR = 1.81 min (UV254 nm); mass calculated for formula C15H14CINO 259.1 , observed LCMS m/z 260.0 (M+H).
Part B:
According to a modification of a procedure by Mach, U. R. et al. (ChemBioChem 2004, 5, 508-518) to a mixture of amide 2432 (1.79 g, 6.9 mmol) and powder zinc chloride (3.29 g, 24.1 mmol) in toluene (30 mL) was added dropwise phosphorus oxychloride (4.4 mL, 48.3 mmol) and the mixture was heated at reflux for 2 h, during which time a polymer-like residue formed. The reaction mixture was cooled in an ice bath, quenched by the addition of 2 M sodium hydroxide solution, extracted and sonicated with dichloromethane (the residue dissolved). The combined dichloromethane extracts were concentrated to a solid residue, which was then triturated with THF. The resulting white solid was filtered and dried to give compound 2433 (1.23 g, 74%). 1H NMR (400 MHz, DMSO-d6) δ 7.82-7.67 (m, 6H), 7.58-7.55 (dd, 1 H), 7.41-7.39 (d, 1 H), 3.98 (t, 2H), 3.23 (t, 2H). HPLC-MS tR = 0.96 min (UV254 nm); mass calculated for formula C15H12CIN 241.1 , observed LCMS m/z 242.1 (M+H).
Part C:
According to a modification of a procedure by Mach, U. R. et al.
(ChemBioChem 2004, 5, 508-518) to a solution of imine 2433 (760 mg, 3.14 mmol) in ethanol (50 mL) was added portionwise sodium borohydride (300 mg, 7.86 mmol) and the resulting mixture was heated at reflux overnight. The reaction mixture was cooled, quenched with water, concentrated, partitioned between ethyl acetate and water, and extracted with ethyl acetate. The combined organic extracts were dried and concentrated to give amine 2434 (418 mg, 55%). 1H NMR (400 MHz, DMSO-d6) δ 7.33-7.04 (m, 7H), 6.62-6.60 (d, 1 H), 5.00 (s, 1 H), 3.36-3.26 (m, 1 H), 3.11-3.06 (m, 1 H), 2.98-2.73 (m, 2H). HPLC-MS tR = 1.14 min (UV254 nm); mass calculated for formula C15H14CIN 243.1 , observed LCMS m/z 244.1 (M+H).
Example 55C:
Figure imgf000683_0001
Part A:
A mixture of compound 2435 (6 g, 36.5 mmol) and urea (1.95 g, 32.5 mmol) was heated over a sand bath at 350 °C for 30 minutes. The resulting black tar was cooled down to 100 "C and water (25 mL) was carefully added. This mixture was stirred for 1 hour. The solid was collected by filtration, dissolved in DMF (40 mL) and stirred with Darco (1 g) for 1 hour. The mixture was filtered and water (100 mL) was added to the filtrate to precipitate out a brown solid. The solid was collected by filtration and air dried. The the solid was dissolved in boiling MeOH (120 mL) with Darco (1 g). The mixture was filtered and the filtrate was concentrated to dryness. The residue was suspended in EtOAc and filtered to afford the desired compound 2436 as brown solid (2.5 g, 43%).
Part B:
Compound 2436 (1 g, 6.2 mmol) was added to a solution of 1 M borane/THF (21 mL, 21 mmol) at 0 °C and the mixture was warmed to room temperature and then refluxed for 18 hours. The reaction mixture was cooled to room temperature and 6N HCI (50 mL) was added dropwise. The reaction mixture was concentrated to remove THF and then cooled to 0 °C and sodium hydroxide pellets (13 g) were added carefully to the reaction mixture. The reaction mixture was extracted with CH2CI2 (1 x 50 mL, 1 x 20 mL). The organic layer was dried over MgS04, filtered and concentrated. Purification by column chromatography (Si02, 3% MeOH(NH3)/CH2CI2, followed by 5% MeOH(NH3)/CH2CI2) afforded the desired compound 2437 as tan solid (64 mg, 7.7%).
The following compounds were prepared using previously described procedures.
Figure imgf000684_0001
Figure imgf000685_0002
Example 56:
Example 56A:
Figure imgf000685_0001
Parts A-D:
Compounds 2448-2451 were prepared according to the procedures in Syn.
Lett. 1999, 12, 1907-1910.
Part E:
Compound 2451 (200 mg, 0.52 mmol), HATU (260 mg, 0.68 mmol), pyrrolidine (0.052 mL, 0.624 mmol), and DIEA (0.1 mL) were dissolved in DMF (5 mL). The reaction mixture was stirred overnight at room temperature and then partitioned between ethyl acetate and water. The organic layers were washed with 1 M HCI, saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated. Purification by column chromatography (Si02, 50% ethyl acetate/hexanes) afforded the desired product (200 mg). HPLC-MS tR = 1.91 min (UV254 nm); mass calculated for formula C22H30N2O7 434.21 , observed LCMS m/z 435.1 (M+H).
Part F:
Compound 2452 (200 mg, 0.46 mmol) was dissolved in methylene chloride (4 mL) and trifluoroacetic acid (1 mL). The reaction mixture was stirred at room temperature for 4 hours. The solvent was evaporated and the material was used without further purification (160 mg). HPLC-MS tR = 1.31 min (UV254 n ); mass calculated for formula Cι8H22N207434.21 , observed LCMS m/z 379.1
(M+H).
Part G:
Compound 2453 (160 mg, 0.423 mmol), compound 3000 (prepared as described in Example 27A) (122 mg, 0.423 mmol), HATU (210 mg, 0.55 mmol), and DIEA (0.5 mL) were dissolved in DMF (10 mL). The reaction mixture was stirred overnight at room temperature and then partitioned between ethyl acetate and water. The organic layers were washed with 1 M HCI, saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated. Purification by column chromatography (Si02, 25% ethyl acetate/hexanes to 100% ethyl acetate) afforded the desired product (175 mg). HPLC-MS tR = 1.94 min (UV254 nm); mass calculated for formula C31H36FN5O6593.2, observed LCMS m/z 594.2 (M+H).
Part H:
Compound 2454 (50 mg, 0.084 mmol) was dissolved in 7 M ammonia in MeOH (0.5 mL) and MeOH (2 mL) and stirred for 30 minutes. The solvent was removed under reduced pressure. The residue was dissolved in MeOH (5 mL) and acetic acid (0.3 mL) and Pd-C (100 mg) was added under an argon atmosphere. The reaction was stirred under a hydrogen atmosphere for 3 hours and then filtered through a pad of celite. The solvent was evaporated and the residue was partitioned between ethyl acetate and water. The combined organic layers were washed with saturated sodium bicarbonate, brine, dried over sodium sulfate and concentrated. The residue was dissolved in ethyl acetate (2 mL) and treated with 4 M HCI in dioxane (0.1 mL). The solids were filtered and washed several times with ethyl acetate to obtain the desired product as an HCI salt (23 mg). HPLC-MS tR = 1.15 min (UV254 nm); mass calculated for formula C21H28FN5O3417.2, observed LCMS m/z 418.1 (M+H).
Example 56B:
Figure imgf000687_0001
Part A:
Compound 2451 (200 mg, 0.52 mmol), compound 3000 (194 mg, 0.68 mmol), HATU (260 mg, 0.68 mmol), and DIEA (0.5 mL) were dissolved in DMF (5 mL). The reaction mixture was stirred overnight at room temperature and then partitioned between ethyl acetate and water. The organic layers were washed with 1 M HCI, saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated. Purification by column chromatography (Si02, 33% ethyl acetate/hexanes to 100% ethyl acetate) afforded the desired product (230 mg). HPLC-MS tR = 2.20 min (UV254 nm); mass calculated for formula C3iH37FN407596.2, observed LCMS m/z 597.2 (M+H).
Part B:
Compound 2457 (230 mg, 0.386 mmol) was dissolved in methylene chloride
(4 mL) and trifluoroacetic acid (1 mL). The reaction mixture was stirred at room temperature for 4 hours. The solvent was evaporated and 2458 (190 mg) was used without further purification. HPLC-MS tR = 1.77 min (UV254 nm); mass calculated for formula C27H2gFN407540.2, observed LCMS m/z 541.2
(M+H).
Part C: Compound 2458 (190 mg, 0.32 mmol), HATU (160 mg, 0.416 mmol), pyrrolidine (0.035 mL, 0.416 mmol), and DIEA (0.5 mL) were dissolved in DMF (5 mL). The reaction mixture was stirred overnight at room temperature and then partitioned between ethyl acetate and water. The organic layers were washed with 1 M HCI, saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated. Purification by column chromatography (Si02, 25% ethyl acetate/hexanes to 100% ethyl acetate) afforded the desired product (200 mg). HPLC-MS tR = 1.92 min (UV254 nm); mass calculated for formula C3ιH36FN506593.2, observed LCMS m/z 594.2 (M+H).
Part D:
Compound 2459 (55 mg, 0.092 mmol) was dissolved in 7 M ammonia in MeOH (0.5 mL) and MeOH (2 mL) and stirred for 30 minutes. The solvent was removed under reduced pressure. The residue was dissolved in MeOH (5 mL) and acetic acid (0.3 mL) and Pd-C (100 mg) was added under an argon atmosphere. The reaction was stirred under a hydrogen atmosphere for 3 hours and then filtered over a bed of celite. The solvent was evaporated and the residue was partitioned between ethyl acetate and water. The combined organic layers were washed with saturated sodium bicarbonate, brine, dried over sodium sulfate and concentrated. The residue was dissolved in ethyl acetate (2 mL) and treated with 4 M HCI in dioxane (0.1 mL). The solids were filtered and washed several times with ethyl acetate to obtain 2460 as an HCI salt (29 mg). HPLC-MS tR = 1.15 min (UV25 nm); mass calculated for formula C21H28FN503417.2, observed LCMS m/z 418.1 (M+H).
Example 56C:
Figure imgf000688_0001
Part A: Compound 2462 was prepared using a modification of the procedure in Chem. Pharm. Bull. 1991 , 39, 8, 1972-1982. Compound 2461 (1.06 g, 5.14 mmol) and Bu2SnO (1.28 g, 5.14 mmol) were dissolved in toluene (20 mL) and stirred at reflux for 1.5 hours using a Dean-Stark trap to azeotrope the water formed in the reaction. The solvent was evaporated under reduced pressure and placed under vacuum for 1 hr. Cesium fluoride (1.08 g, 6.68 mmol) was added to the residue and placed under vacuum for an additional 2 hours. The mixture was then dissolved in DMF (20 mL) and iodomethane (2.99 g, 22.1 mmol) and stirred overnight at room temperature. The reaction mixture was partitioned between ethyl acetate and water. The organic layers were washed with 1 N HCI, saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated. Purification by column chromatography (Si02, 33% ethyl acetate/hexanes) afforded the desired product (800 mg). 1H NMR (400 MHz, CDCI3) δ 4.60 (d, 1 H), 4.30 (m, 4H), 4.15 (d, 1 H), 3.50 (s, 3H), 1.35 (t, 6H). Part B:
Compound 2463 was prepared using a modification to a procedure found in Tetrahedron 1993, 49, 37, 8381-8396. The diester 2462 (0.837 g, 3.6 mmol) was suspended in pH 8 phosphate buffer (75 mL) and pH was monitored at 8.33. Pig liver esterase (20 mg) was added to the suspension and the reaction mixture was stirred at room temperature. The pH of the reaction was maintained between 7.9-8.2 by the dropwise addition of 1M NaOH (3.27 mL, 3.24 mmol) over 1.5 hours. The reaction mixture was partitioned between diethyl ether and water. The aqueous layer was acidified to pH 2 and extracted with ethyl acetate. The combined ethyl acetate layers were dried over sodium sulfate and evaporated to provide the desired product as a 4.5:1 mixture of inseparable acids (275 mg). 1H NMR (400 MHz, CDCI3) δ 4.60 (d, 1H), 4.40 (m, 2H), 4.25 (d, 1H), 3.50 (s, 3H), 1.35 (t, 3H).
Part C:
Compound 2463 (130 mg, 0.65 mmol), HATU (322 mg, 0.845 mmol), 4- phenylbenzylamine (153 mg, 0.845 mmol), and DIEA (0.2 mL) were dissolved in DMF (6 mL). The reaction mixture was stirred overnight at room temperature and then partitioned between ethyl acetate and water. The organic layers were washed with 1 M HCI, saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated. Recrystallization from ethyl acetate provided pure product (50 mg) and slightly impure product (150 mg). HPLC-MS tR = 1.70 min (UV254 nm); mass calculated for formula C20H23NO5357.1 , observed LCMS m/z 358.1 (M+H).
Part D:
Compound 2464 (50 mg, 0.140 mmol) was dissolved in 1 M LiOH (0.151 mL, 0.151 mmol) and THF (5 mL) and stirred for 1 hour. The reaction mixture was partitioned between ethyl acetate and water and the aqueous layer was acidified to pH 2 using 1 N HCI. The organic layers were dried over sodium sulfate and concentrated to provide the desired product (46 mg). HPLC-MS tR = 1.43 min (UV254 nm); mass calculated for formula C189N05329.1 , observed LCMS m/z 330.1 (M+H).
Part E:
Compound 2465 (46 mg, 0.140 mmol), HATU (70 mg, 0.182 mmol), compound 2000 (35 mg, 0.182 mmol), and DIEA (0.12 mL) were dissolved in DMF (6 mL). The reaction mixture was stirred overnight at room temperature and then partitioned between ethyl acetate and water. The organic layers were washed with saturated sodium bicarbonate, brine, dried over sodium sulfate, and concentrated. Recrystallization from ethyl acetate provided pure product (30 mg). HPLC-MS (10 min) tR = 3.51 min (UV254 nm); mass calculated for formula C25H25N304431.2, observed LCMS m/z 432.1 (M+H).
Example 56D: HJ
Figure imgf000691_0001
2474
Part A:
Compound 2467 was prepared according to a modified literature procedure
(Nagashima, N. and Ohno, M., Chem. Pharm. Bull., 1991 , 39, 1972-1982.)
L-Diethyl tartrate (2461) (6.18 g, 30 mmol) and dibutyltin oxide (7.47 g, 30 mmol) in toluene (100 mL) was heated under reflux for 1 h, removing water formed as the azotropic mixture. The solution was evaporated to complete dryness in vacuo to afford a white solid. To this solid was added dry CsF (8.66 g, 59 mmol) and DMF (60 mL). The resulting mixture was cooled to 0 °C, and benzyl bromide (6 mL, 51 mmol) was added dropwise via a syringe. After the completion of addition, the reaction mixture was allowed to stir rigorously at room temperature for 12 h. The solvent was removed under vacuum and the obtained residue was dissolved in EtOAc and H20. The organic layer was washed with NaHCθ3 aqueous solution, brine, dried over Na2S0 , and concentrated. Purification by flask column chromatography on silica gel (EtOAc / hexane 30:70) afforded compound 2467 as colorless liquid (8.10 g, 91 %). 1H NMR (400 MHz, CDCI3) δ 7.37-7.25 (m, 5H), 4.87 (d, J = 12.0 Hz, 1H), 4.58 (br s, 1 H), 4.02 (d, J = 12.0 Hz, 1 H), 4.34 - 4.25 (m, 3H), 4.23-4.21 (m, 1H), 4.08 -4.03 (m, 1 H), 3.12 (br s, 1H), 1.35 (t, J = 7.0 Hz, 3H), 1.19 (t, J = 6.9 Hz, 3H). Part B:
To compound 2467 (2.96 g, 10 mmol) in toluene (10 mL) was added with Ag2θ (4.63 g, 20 mmol) and allyl bromide (1.31 mL, 15 mmol). The reaction mixture was allowed to stir at 110 °C for 16 h and cooled to room temperature. After filtering through a pad of celite, the solution was concentrated. Flash column chromatography on silica gel (EtOAc / hexane 20: 80) afforded the desired product 2468 (2.65 g, 79%). 1H NMR (400 MHz, CDCI3) δ 7.33-7.25 (m, 5H), 4.87 (d, J = 12.0 Hz, 1 H), 5.89-5.79 (m, 1 H), 5.26-5.15 (m, 2H), 4.87 (d, J = 11.4 Hz, 1 H), 4.46 (d, J = 11.7 Hz, 1 H), 4.41-4.38 (m, 2H), 4.34 - 3.92 (m, 6H), 1.32 (t, J = 7.2 Hz, 3H), 1.19 (t, J = 7.2 Hz, 3H).
Part C:
To an ice-cold mixture of compound 2468 (1.0 g, 3 mmoi) and N- methylmorpholine monohydrate (703 mg, 6 mmol) in THF / H 0 3:1 (20 mL) was added Os04 (2.5 wt% in -BuOH, 610 mg, 0.06 mmol). After stirring for 30 min, the ice bath was removed and the reaction mixture was stirred at room temperature overnight. Solid NaHS03 (750 mg, 7.2 mmol) was added, and the mixture was stirred for an additional 30 min. The mixture was filtered through a pad of silica and the solution was concentrated in vacuo giving rise to an oil. This material was dissolved in THF/H20 3:1 (20 mL) and Nal04 (1.28 g, 6 mmol) was added. The mixture was allowed to stir at room temperature for 1 h. It was filtered through silica, and the solvent was evaporated. Flash column chromatography on silica gel (EtOAc/ hexane 50:50) provided the aldehyde 2469 (760 mg, 75%) as a colorless oil). 1H NMR (400 MHz, CDCI3) δ 9.72 (s, 1 H), 7.35-7.26 (m, 5H), 4.89 (d, J = 11.8 Hz, 1 H), 4.52-4.48 (m, 2H), 4.45 - 4.04 (m, 7H), 1.34 (t, J = 7.7 Hz, 3H), 1.19 (t, J = 7.4 Hz, 3H).
Part D:
Compound 2469 (340 mg, 1 mmol) and amine 916 (187 mg, 1 mmol) in 1 ,2- dichloroethane (5 mL) were stirred at room temperature for 5 min, after which sodium tri(acetoxyl)borohydride (254 mg, 1.2 mmol) was added slowly. The reaction mixture was allowed to stir at room temperature overnight. The solution was concentrated, the crude residue was purified by flash column chromatography on silica gel (MeOH/EtOAc 10:90), affording desired product 2470 (350 mg, 69%) as a colorless oil. HPLC-MS tR = 1.49 min (UV254 nm); mass calculated for formula C28H35N3O6 509.2, observed LCMS m/z 510.1 (M+H).
Part E:
To a solution of compound 2470 (170 mg, 0.33 mmol) in dioxane / water 1 :1 (10 mL) was added LiOH (1 M, 1.32 mL, 1.32 mmol) dropwise. The mixture was stirred at room temperature for 2h after which it was neutralized with 1 M HCI and concentrated. The residue was utilized for the following step without further purification. HPLC-MS tR = 1.12 min (UV25.Φ nm); mass calculated for formula C24H2 N3O6453.2, observed LCMS m/z 454.1 (M+H).
Part F:
The crude compound 2471 was dissolved in DMF (10 mL) and cooled to 0 °C in an ice bath, HATU (304 mg, 0.8 mmol) was added slowly and the reaction mixture was stirred at 0 °C for 2h. LC-MS indicated the disappearance of diacid and the formation of the cyclized product 2472. HPLC-MS tR = 1.65 min (UV254 nm); mass calculated for formula C24H25N3θ5435.2, observed LCMS m/z 436.1 (M+H).
Part G:
To the above solution at 0 °C was added isoindoline (48 mg, 0.4 mmol) and the reaction mixture was stirred at room temperature overnight. The DMF was evaporated, the residue was dissolved inn EtOAc and water, washed with 1 N HCI, saturated NaHC03 and brine, dried over Na2S04 and concentrated to afford 2437 as an oily solid (85 mg, 47% yield over three steps). HPLC-MS tR = 2.00 min (UV254 nm); mass calculated for formula C32H32N 04 536.1 , observed LCMS m/z 537.1 (M+H). Part H:
Crude compound 2473 was dissolved in EtOH (20 mL), and palladium on carbon (10%, 40 mg) was added. The mixture was hydrogenated (atmospheric pressure) at room temperature overnight. The suspension was filtered through celite and concentrated. The resulting residue was purified by preparative LC affording 2474 as a white solid (9.4 mg). HPLC-MS tR = 3.57 min (10 min method, UV254 nm); mass calculated for formula C25H26N404 446.2, observed LCMS m/z 447.1 (M+H).
The following compounds were prepared using previously described procedures.
Figure imgf000694_0001
Example 57:
Figure imgf000694_0002
2183 2479 Part A:
Compound 2183(200 mg, 1.3 mmol) and cyclopropyl methyl amine (0.16 mL) were dissolved in MeOH (5 mL) and stirred at 50 °C overnight. Then sodium borohydride (42 mg, 1.3 mmol) was added and the mixture was stirred for 2 hours. The solvent was removed under reduced pressure and the residue was partitioned between diethyl ether and 1 N HCI. The aqueous layer was made basic and extracted with ethyl acetate. The combined ethyl acetate layers were dried over sodium sulfate and concentrated to provide the desired product 2479 (130 mg). 1H NMR (400 MHz, CDCl3) δ 7.32 (m, 1 H), 7.28- 7.18 (m, 3H), 3.79 (m, 1H), 2.50-2.25 (m, 2H), 1.38 (d, 3H), 1.00-0.90 (m, 1H), 0.50-0.45 (m, 2H), 0.10-0.00 (m, 2H).
The following compounds were prepared using previously described procedures.
Figure imgf000695_0001
Figure imgf000696_0002
Example 58:
Example 58A:
Figure imgf000696_0001
Part A:
To a solution of B0C2O (6.1 g, 28 mmol) and EtsN (7.8 mL, 56 mmol) in DMF (20 mL) at 0 °C was added portionwise 3-chlorophenylhyrazine hydrocloride (5.0 g, 28 mmol) in DMF (20 mL). The reaction mixture was allowed to stir at room temperature for 2 h and concentrated. The residue was dissolved in EtOAc and H20, washed with 1 N citric acid and saturated NaHC03, dried over Na2S0 and concentrated, affording 2490 (17.83 g, 97%) as a off-white solid. H NMR (400 MHz, CDCI3) δ 7.13 (t, J = 8.3 Hz, 1 H), 6.86-6.81 (m, 2H), 6.70- 6.66 (m, 1 H), 6.36 (br s, 1 H), 1.48 (s, 10H).
Part B: 3-Chloropropionyl chloride (0.400 mL, 4.12 mmol) was added dropwise into a solution of compound 2490 (1.0 g, 4.12 mmol) and K2C03 (1.14 g, 8.24 mmol) in DMF (10 mL) at 0 °C. The reaction mixture was allowed to stir at 80°C overnight and concentrated. The residue was dissolved in EtOAc and H20, washed with 1 N citric acid and saturated NaHC03, dried over Na2S04, and concentrated. Column chromatography (EtOAc/hexane 25:75) provided compound 2491 (600 mg, 49%) as an off-white solid. 1H NMR (400 MHz, CDCI3) δ 7.57 (t, J = 2.3 Hz, 1 H), 7.51-7.48 (m, 1 H), 7.27 (t, J = 8.3 Hz, 1 H), 7.12-7.09 (m, 1 H), 4.16 (t, J = 7.2 Hz, 2H), 2.77 (t, J = 7.3 Hz, 2H), 1.34 (s, 9H).
Part C:
Compound 2491 was deprotected with TFA in CH2CI2. The material was used without further purification.
Example 58B:
Figure imgf000697_0001
Part A:
To a suspension of NaH (60% in mineral oil, 1.56 g, 39 mmol) in benzene (40 mL) at 0 °C was added with 3-chlorophenylhyrazine (2.13 g, 15 mmol). The mixture was stirred at 80 °C for 1 h, after which 1 ,3-dibromopropane (2.0 g, 10 mmol) was added slowly via syringe. It was stirred at 80 °C overnight and quenched by adding water (20 mL). The organic layer was separated, washed with water and brine, dried over Na2S04, and concentrated. Column chromatography (DCM) provided compound 2493 (850 mg, 47%) as pale yellow oil. 1H NMR (400 MHz, CDCI3) δ 7.12 (t, J = 8.2 Hz, 1 H), 7.04 (t, J = 2.3 Hz, 1H), 6.84-6.81 (m, 1H), 6.74-6.71 (m, 1H), 3.75 (bs, 1 H), 3.35 (t, J = 7.5 Hz, 2H), 3.00 (t, J = 6.7 Hz, 2H), 2.16-2.11 (m, 2H).
The following compounds were prepared using previously described procedures.
Figure imgf000698_0002
Example 59: Example 59A:
Figure imgf000698_0001
2500 2501
Part A:
To compound 471 (100 mg, 0.37 mmol) in EtOH (2 mL) was added chloroacetaldehyde (50 wt.% in water, 233 mg, 1.48 mmol). The reaction mixture was allowed to stir at 90 °C overnight and concentrated. The residue was dissolved in EtOAc , washed with water and brine, dried (Na2SO4) and concentrated. The crude product 2500 (90 mg, 82%) was used without further purification. HPLC-MS tR = 1.10 min; Mass calculated for formula C14H19N3θ2S 293.1 , observed m/z 294.1 (M+H).
Part B:
Compound 2500 (90 mg) was dissolved in 2 mL of 4 N HCL in dioxane/H20
(1 :1 ). After stirring at room temperature for 1 h, the solution was concentrated, affording a brown solid (80 mg, 90%). HPLC-MS tR = 0.22 min; Mass calculated for formula C9H11N3S 193.1 , observed m/z 194.1 (M+H).
Example 59B:
Figure imgf000699_0001
470 2502 2503
Part A:
A mixture of chloroketone 470 (200 mg, 0.81 mmol) and 2-aminopyridine (77 mg, 0.82 mmol) in ethanol (5 ml) was heated to reflux and stirred overnight. The reaction mixture was concentrated and purified by prep-HPLC to afford
2502 (103 mg). HPLC-MS tR = 1.00 min (UV254 nm); mass calculated for formula C16H21 N302 287.2, observed LCMS m/z 288.3 (M+H).
Part B:
To a solution of imidazopyridine 2502 (103 mg, 0.56 mmol) in dioxane (2 mL) was added HCI (4N in dioxane, 4 mL) and water (0.5 mL). The mixture was stirred at room temperature for 1 hour and concentrated. The resulting residue
2503 (81 mg) was dried under vacuum and used in the next step without further purification. HPLC-MS tR = 0.19 min (UV254 nm); mass calculated for formula C11 H13N3 187.1 , observed LCMS m/z 188.1 (M+H).
Example 59C:
Figure imgf000700_0001
470 2504 2505
Part A:
A mixture of chloroketone 471 (200 mg, 0.81 mmol) and 2-aminopyridine (140 mg, 0.81 mmol) in ethanol (5 ml) was heated up to reflux and stirred overnight. The reaction mixture was concentrated and purified by prep-HPLC to afford 2504 (119 mg). HPLC-MS tR = 1.22 min (UV254 nm); mass calculated for formula C19H25N304 359.2, observed LCMS m/z 360.1 (M+H).
Part B:
To a solution of imidazopyridine 2504 (119 mg, 0.33 mmol) in dioxane (2 mL) was added HCI (4N in dioxane, 4 mL) and water (0.5 mL). The mixture was stirred at room temperature for 1 hour and concentrated. The resulting residue 2505 (83 mg) was dried under vacuum and used in the next step without further purification. HPLC-MS tR = 0.70 min (UV254 n ); mass calculated for formula C14H17N302 259.1 , observed LCMS m/z 260.1 (M+H).
The following compounds were prepared using previously described procedures.
Figure imgf000700_0002
Figure imgf000701_0002
Example 60
Figure imgf000701_0001
Part A:
Trifluoroacetic anhydride (16 mL, 115.1 mmol) was dissolved in methylene chloride (60 mL) and cooled in an ice bath. Compound 840 (10.0 g, 82.6 mmol) was added dropwise in methylene chloride (40 mL) and stirred at room temperature for one hour. The solution was cooled in an ice bath and iodine (11.0 g, 43.2 mmol) and compound 2509 (9.8 g, 22 mmol) were added and stirred for 12 hours in the dark (cover flask with aluminum foil). Additional compound 2509 (9.5 g, 20 mmol) was added and stirring was continued for 12 hours. The reaction mixture was added to a cooled solution of methylene chloride and 5% sodium bisulfite and stirred for one hour. The organic layer was washed with saturated sodium bicarbonate, filtered through a pad of silica, and concentrated. The residue was suspended in diethyl ether (30 mL) and hexanes (90 mL) and stirred for 30 minutes. The mixture was filtered to provide compound 2510 as a white solid (15.5 g). The product was stored in the dark at room temperature. 1H NMR (400 MHz, CDCI3) δ 7.70 (d, 2H), 7.06 (d, 2H), 5.10 (m, 1 H), 1.60 (d, 3H).
Part B:
Compound 2510 (4.0 g, 11.66 mmol), pyrazole (0.953 g, 14 mmol), copper (I) iodide (444 mg, 2.33 mmol), cesium carbonate (7.6 g, 23.3 mmol), 1 ,10 phenanthroline (844 mg, 4.66 mmol) and dimethylacetamide (40 mL) were combined in a 80 mL screw-capped vial and stirred in the dark at 120 °C for 12 hours. The reaction mixture was poured into ethyl acetate (200 mL) and filtered. The organic layer was washed with water several times followed by 1 N citric acid solution. The organic layer was dried over sodium sulfate and concentrated. The residue was recrystallized from ethyl acetate and hexanes to provide 2.45 g of compound 915. 1H NMR (400 MHz, CDCI3) δ7.92 (m, 1 H), 7.70 (m, 2H), 7.40 (d, 1 H), 6.45 (t, 1 H), 5.20 (m, 1 H), 1.65 (d, 3H).
Part C:
Compound 915 (2.43 g, 9.13 mmol) was dissolved in methanol (20 mL) and 1 N lithium hydroxide (20 mL) and stirred for 6 hours. The reaction mixture was poured into ethyl acetate and water. The aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, and concentrated to provide compound 916 (1.45 g). 1H NMR (400 MHz, CDCI3) δ7.90 (d, 1 H), 7.70 (d, 1 H), 7.64 (d, 2H), 7.43 (d, 2H), 6.46 (t, 1 H), 1.44 (d, 3H).
Part D:
Compound 916 (1.45 g, 7.7 mmol), compound 1 (1.5 g, 7.35 mmol), HATU (3.63 g, 9.55 mmol), and diisopropylethylamine (1.28 mL, 7.35 mmol) were dissolved in DMF (20 mL) at 0 °C. The reaction mixture was stirred for 12 hours and then diluted with water and extracted with ethyl acetate. The combined organic layers were washed with 1 N HCI, saturated sodium bicarbonate, brine and water, dried over sodium sulfate and concentrated. The residue was purified by column chromatography (2:1 hexanes/ethyl acetate) to provide compound 2511 (2.15 g). 1H NMR (400 MHz, CDCI3) δ7.90 (d, 1 H), 7.72 (m, 1 H), 7.66 (d, 2H), 7.40 (d, 2H), 6.75 (d, 1 H), 6.49 (d, 1 H), 5.20 (m, 1 H), 4.82-4.75 (dd, 2H), 3.83 (s, 3H), 1.57 (d, 3H), 1.54 (s, 3H), 1.50 (s, 3H).
Part E:
Compound 2511 (2.15 g, 5.76 mmol) was dissolved in THF (20 mL) and 1 N lithium hydroxide and stirred for 2 hours. Diethyl ether was added and the aqueous layer was acidified to pH 2 with 1 N HCI and extracted with ethyl acetate. The combined ethyl acetate layers were dried over sodium sulfate and concentrated to provide compound 2512 as a white solid (1.95 g). 1 H NMP (400MHz CDCI3) δ7.9 (d, 1 H), 7.74 (m, 1 H), 7.70 (d, 2H), 7.42 (d, 2H), 6.96 (d, 1 H), 6.49 (t, 1 H), 5.2 (m, 1 H), 4.60-4.50 (dd, 2H), 1.64 (d, 3H), 1.54 (d, 3H). Example 61 : Example 61A:
H
Figure imgf000703_0001
Figure imgf000703_0002
Part A:
(R,S)-BOC-1 ,3-Dihydro-2H-lsoindole carboxylic acid 2520 (3.0 g, 11.39 mmol) and cesium carbonate (4.05 g, 12.43 mmol) were stirred together in DMF (25mL) at r.t. for one hour. Iodomethane (1.05 mL, 16.86 mmol) was added and the reaction mixture was stirred at ambient temperature overnight. The reaction mixture was diluted with ethyl acetate then washed several times with water, brine (1 x 50 mL), dried over Na2S04 and concentrated to give light yellow oil. Purification by column chromatography (Si02, 25% ethyl acetate / hexane) afforded 2521 (2.66 g, 84 %) as white solid.
Part B:
A mixture of 2521 (0.2 g, 0.72 mmol) and hydrazine (0.22 mL, 7.0mmol) in ethanol (20 mL) was heated at 70 °C for 5 hours. The solvent was removed under reduced pressure and the resulting residue was dissolved in ethyl acetate, washed with water (2 x 30mL), brine (1 x 20 mL), dried over Na2S04 and concentrated to provide 2522 (0.18 g, 86%) as an yellow oil, which was used in the next step without further purification.
Part C:
A mixture of 2522 (0.2 g, 0.72 mmol) and ethyl isocyanate (0.077 g, 1.083 mmol) in DCM (5 mL) was stirred at r.t. for 16 hours. Additional ethyl isocyanate (0.077 g, 1.083 mmol) was added and reaction was stirred for another two hours. The solvent was removed under reduced pressure to provide the crude product as a white gum. It was dissolved in DCM (20 mL) and treated triethyl amine (0.5 mL, 3.6 mmol) , DMAP (0.017 g, 0.139 mmol) and p-toluene sulphonyl chloride (0.16 g, 0.83 mmol). The mixture was stirred at room temperature for 64 hours and the solvent was removed. The residue was purified by preparative chromatography using 5% MeOH/CH2CI to provide the product which was again purified using ethyl acetate/Hexane (3/1 ) to yield intermediate 2523 (0.1 g, 44%) as a light brown solid.
Part D:
Intermediate 2523 (0.05 g, 0.15 mmol) was dissolved in DCM (10 mL) then trifluoroacetic acid (0.06 mL, 0.77 mmol) was added. Reaction was stirred at ambient temperature for 3 hours and then concentrated. The residue was dissolved in DCM and washed with saturated sodium bicarbonate solution (1 x 0.8 mL), saline(1 x 0.8 mL), dried over sodium sulfate and concentrated under vacuum to provide amine 2524 (0.03 g, 86%) as a brown oil. It was used in the next step without additional purification.
Figure imgf000705_0001
2521 2525 2526
Part A:
Intermediate 2525 was prepared by known methods (Showell , G.A.; Gibbons, T.L.; Kneen, CO.; Macleod, A.M..; Merchant, K.; Saunders, J.; Freedman, S.B.; Patel, S; Baker, R.. J Med Chem. 1991 , 34 (3), 1086-1094).
Part B:
Intermediate 2526 was prepared using the method described in Example 61 A
Part D.
The following compounds were prepared using previously described procedures.
Figure imgf000705_0002
Example 62: Example 62A:
Figure imgf000706_0001
Part A:
A mixture of 2173 (1.54 g, 6.28 mmol) and Dess-Martin periodinane (2.72 g, 6.41 mmol) in DCM (30 ml) was stirred at room temperature overnight. To the reaction mixture was add saturated sodium bicarbonate solution and the mixture was stirred for 2 hours. The solids were removed by filtration. The filtrate was separated and the organic layer was washed with brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 30% ethyl acetate / hexane) afforded 2530 (939 mg, 61 %) as an oil. HPLC-MS tR = 1.10 min (MS); Mass calculated for formula C11 H17N05 243.1 , observed m/z 266.1 (M+Na).
Part B:
To ketone 2530 (939 mg, 3.86 mmol) in DCM (30 mL) at -78 oC under argon was added (diethylamino)sulfur trifluoride (2.53 mL, 19.3 mmol). The bath was slowly warmed to room temperature while the mixture was stirred overnight. The reaction mixture was poured into saturated sodium bicarbonate and the layers were separated. The organic layer was washed with brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 10% to 30% ethyl acetate / hexane) afforded 2531 (751 mg, 74%) as an oil. HPLC-MS tR = 4.72 min (10 min, MS); Mass calculated for formula C11 H17F2N04 265.1 , observed m/z 288.1 (M+Na).
Part C:
Compound 2531 (751 mg, 2.83 mmol) was converted to 2532 (418 mg, 52 %) using the procedures described in Example 10 Part A. HPLC-MS t = 1.85 min (MS); Mass calculated for formula C11 H16CIF2N03 283.1 , observed m/z 228.1 (M-55).
Part D:
Compound 2532 (418 mg, 1.47 mmol) was cyclized with N-methylthiourea according to the procedures described in Example 51 B Part C to afford 2533 (200 mg, 42%). HPLC-MS tR = 1.38 min (UV 254 nm); Mass calculated for formula C8H11 F2N3S 319.1, observed m/z 320.1 (M+H).
Part E:
Compound 2534 was synthesized in quantitative yield using the procedure described in Example 51 D Part H. 1H NMR (400 MHz, CDCI3) .δ 6.89 (s, 1 H), 4.98 (dd, 1 H, J = 7.2, 11.7 Hz), 3.89 (m, 1 H), 3.72 (m, 1 H), 3.13 (s, 3H), 3.09- 2.85 (m, 2H).
Example 62B:
Figure imgf000707_0001
2209A 2535 2536
Part A:
The mixture of 5-bromothiazole 2209A (210 mg, 0.45 mmol), cyclopropylboronic acid (80 mg, 1.0 mmol), Pd(dppf)CI2 (41 mg, 0.05 mmol) and K3P04 (424 mg, 2.0 mmol) in 20-ml vial was flushed with argon for 3 min. Under the argon, dioxane (5 mL) was added and the vial was sealed under the argon atmosphere. The mixture was heated to 80 °C and stirred overnight. After cooling to room temperature, ethyl acetate (30 mL) was added to the reaction mixture and the solution was filtered through celite. The filtrate was concentrated and purified by column chromatography (Si02, 10% ethyl acetate/hexanes) to afford 5-cyclopropylthiazole 2535 (166 mg) as oil. HPLC- MS tR = 2.65 min (UV254 nm); mass calculated for formula C21 H33N304S 423.2, observed LCMS m/z 424.1 (M+H).
Part B:
To the solution of 5-cyclopropylthiazole 2535 (166 mg, 0.39 mmol) in dioxane (2 mL), HCI (4N in dioxane, 4 mL) was added followed by the addition of water (0.5 mL). The mixture was stirred at room temperature for 1 hour and concentrated. The resulting residue 2536 (111 mg) was dried under vacuum and used in the next step without further purification. HPLC-MS tR = 0.86 min (UV254 nm); mass calculated for formula C11 H17N3S 223.1 , observed LCMS m/z 224.1 (M+H).
Example 62C:
Figure imgf000708_0001
Part A:
To Fmoc-D-Tiq-OH (2537) (1.0 g, 2.5 mmol) in dichloromethane (20 mL) at 0 °C was added (COCI)2 (0.262 mL, 3.0 mmol) and DMF (2 drops, catalytic) under argon. The reaction mixture was stirred at 0 °C for 2 hr and concentrated to dryness in vacuo. The resulting syrup was dissolved in dry THF (20 mL) and cooled to 0 °C Trimethylsilyldiazomethane (2.0 M in Et20, 5 mL, 10 mmol) was added dropwise under argon during a period of 30 min. After the completion of addition, the solution was stirred at 0 °C for 3 hr and concentrated under reduced pressure. Flash column chromatography on silica (EtOAc/hexane 25:75) afforded 2538 (630 mg, 60%) as a yellow oily solid. HPLC-MS tR = 2.23 min (UV254 nm); mass calculated for formula C26H2ιN303423.2, observed LCMS m/z 446.2 (M+Na).
Part B: Compound 2538 (630 mg, 1.49 mmol) in Et20 (20 mL) was cooled to 0 °C To this solution was added 2.15 M hydrobromide acid in Et20 (830 DL, 1.79 mmol) dropwise. The reaction mixture was stirred for 20 min at 0 °C and concentrated. The residue was dissolved in EtOAc, the solution was than washed with saturated bicarbonate solution, brine, dried (Na2S04) and concentrated in vacuo to afford 2539 (630 mg, 89%) as an off white solid. HPLC-MS tR = 2.40 min (UV254 nm); mass calculated for formula C26H22BrN03 475.1 , observed LCMS m/z 476.0 (M+H).
Part C:
Compound 2539 (150 mg, 0.31 mmol) and thiourea (36 mg, 0.47 mmol) in DMF (2 mL) was allowed to stir at room temperature for 3 hr. The solution was concentrated and the residue was dissolved with EtOAc and saturated NaHCθ3 solution. The organic phase was washed with H20, brine, dried over Na2S0 , and concentrated. Flash column chromatography on silica (EtOAc / hexane 60:40) afforded 2540 (120 mg, 84%) as a white solid. HPLC-MS tR = 1.96 min (UV25 nm); mass calculated for formula C2 H23N302S 453.2, observed LCMS m/z 454.1 (M+H).
Part D:
Compound 2540 (150 mg, 0.26 mmol) was treated with 20% piperidine in DMF (10 mL). The solution was stirred at room temperature for 2 hr and concentrated. LC-MS indicated the clean deprotection of the Fmoc group. HPLC-MS tR = 0.72 min (UV254 nrn); mass calculated for formula C12H13N3S 231.1 , observed LCMS m/z 232.1 (M+H).
Example 62D:
Figure imgf000709_0001
Part F
Figure imgf000709_0002
Figure imgf000709_0003
Part A:
According to a modification of a procedure by Ortwine, D. F. et al. (J. Med. Chem. 1992, 35, 1345-1370) to an ice-cold solution of 3- chlorophenethylamine 2431 (10 g, 64.6 mmol) and triethylamine (6.46 mL, 67.8 mmol) in DCM (200 mL) was added dropwise ethyl chloroformate (9.45 mL, 67.8 mmol), and the resulting solution was stirred at 0 °C for 1 h, and then at rt for 1.5 h. The reaction mixture was washed with saturated sodium bicarbonate solution, brine, dried and concentrated to give ethyl carbamate 2542 as a colorless oil (14.36 g, 98%). 1H NMR (400 MHz, CDCI3) δ 7.24-7.19 (m, 3H), 7.09-7.07 (d, 1 H), 4.66 (s, broad, NH), 4.12 (q, 2H), 3.44 (q, 2H), 2.81 (t, 2H), 1.25 (t, 3H).
Part B:
According to a modification of a procedure by Ortwine, D. F. et al. (J. Med. Chem. 1992, 35, 1345-1370) to an ice-cold mixture of carbamate 2542 (6.81 g, 29.9 mmol) and 3:1 glacial acetic acid:sulfuric acid (64 mL) was added (in two portions) glyoxylic acid monohydrate (3.03 g, 32.9 mmol) and the mixture was stirred at rt overnight. The reaction mixture was poured in ice, and extracted with dichloromethane (5 times). The combined dichloromethane extracts were dried and concentrated to an oily residue, which was then taken in toluene and concentrated (3 times) and dried under vacuum overnight to give compound 2543 (7.91 g, 93%). 1H NMR (400 MHz, DMSO-d6) δ 12.9 (s, broad, COOH), 7.50-7.12 (m, 3H), 5.42 (s, 1 H), 4.12-4.03 (m, 2H), 3.77-3.50 (m, 2H), 2.89-2.79 (m, 2H), 1.24-1.16 (m, 3H). HPLC-MS tR = 1.68 min (UV254 nm); mass calculated for formula C13H14CIN04 283.1 , observed LCMS m/z 284.0 (M+H).
Part C:
Diazoketone 2544 was prepared using procedures described in Example 62C Part A. 1H NMR (400 MHz, CDCI3) δ 7.23-7.18 (m, 3H), 5.60-5.41 (m, 2H), 4.26-4.22 (m, 2H), 3.90-3.65 (m, 2H), 2.97-2.82 (m, 2H), 1.36-1.32 (m, 3H). HPLC-MS tR = 1.8 min (UV254 n ); mass calculated for formula C14H14CIN303 307.1 , observed LCMS m/z 280.1 (M-N2+H), and 330.1 (M+Na).
Part D:
Bromoketone 2545 was prepared using procedures described in Example 62C Part B. 1H NMR (400 MHz, CDCI3) δ 7.31-7.20 (m, 3H), 5.87-5.81 (m, 1 H), 4.25-4.04 (m, 4H), 3.90-3.82 (m, 1 H), 3.60-3.51 (m, 1 H), 2.98-2.79 (m, 2H), 1.34-1.31 (m, 3H). HPLC-MS tR = 2.07 min (UV254 nm); mass calculated for formula C14H15BrCIN03 359.0, observed LCMS m/z 360.0 (M+H).
Part E:
Thiazole 2546 was prepared using procedures described in Example 10. HPLC-MS tR = 2.05 min (UV25 nm); mass calculated for formula C18H22C1N302S 379.1 , observed LCMS m/z 380.1 (M+H).
Part F:
To a solution of thiazole 2546 (117 mg, 0.31 mmol) in chloroform (1.5 mL) was added iodotrimethylsilane (0.32 mL, 2.24 mmol) and the resulting mixture was heated at 50 °C for 2 h. The reaction mixture was diluted with dichloromethane, washed with saturated sodium bicarbonate, brine and concentrated to give amine 2547 as a dark yellow solid (93 mg, 97%). 1H NMR (400 MHz, CDCI3) δ 7.12-6.97 (m, 3H), 6.01 (s, 1 H), 5.26 (s, broad, 1 H), 5.15 (s, 1H), 3.60-3.53 (m, 1H), 3.18-2.80 (m, 4H), 1.28-1.26 (d of d, 6H). HPLC-MS tR = 1.24 min (UV254 nm); mass calculated for formula C15H18CIN3S 307.1 , observed LCMS m/z 308.2 (M+H).
Example 62E: Part B
Part C
Figure imgf000712_0001
Figure imgf000712_0002
Part A:
According to a modification of a procedure by Ortwine, D. F. et al. (J. Med. Chem. 1992, 35, 1345-1370) to an ice-cold solution of 3- chlorophenethylamine 2431 (11.6 g, 75 mmol) and triethylamine (10.9 mL, 78.7 mmol) in DCM (300 mL) was added dropwise benzyl chloroformate (11.1 mL, 78.1 mmol), and the resulting solution was stirred at 0 °C for 30 min, and then at rt overnight. The reaction mixture was washed with saturated sodium bicarbonate solution, brine, dried and concentrated to give benzyl carbamate 2548 as a colorless oil (21.4 g, 99%). 1H NMR (400 MHz, CDCI3) δ 7.38-7.30 (m, 5H), 7.23-7.05 (m, 4H), 5.1 (s, 2H), 4.75 (s, broad, NH), 3.46 (q, 2H), 2.82 ( 2H).
Part B:
According to a modification of a procedure by Ortwine, D. F. et al. (J. Med. Chem. 1992, 35, 1345-1370) to an ice-cold mixture of benzyl carbamate 2548 (8.6 g, 31.4 mmol) and 3:1 glacial acetic acid:sulfuric acid (64 mL) was added (in two portions) glyoxylic acid monohydrate (3.0 g, 32.6 mmol) and the mixture was stirred at rt overnight. The reaction mixture was poured in ice, and washed with dichloromethane (4 times). The aqueous phase was concentrated, diluted with tetrahydrofuran (200 mL), and treated with 6M NaOH solution (130 mL) to pH 12-13, followed by di-tert-butyi dicarbonate (7.94 g, 36.4 mmol), and then stirred at rt overnight. The reaction mixture was concentrated, diluted with water, acidified with 2 M HCI solution to pH 2, and extracted with ethyl acetate. The combined organic extracts were concentrated and chromatographed (Si02, 5% methanol/dichloromethane) to give compound 2549 (0.95 g, 15% based on reacted starting material). 1H NMR (400 MHz, CDCI3) δ 7.54-7.27 (m, 3H), 5.67-5.50 (d, 1 H), 3.87-3.79 (m, 2H), 3.01-2.88 (m, 2H), 1.61-1.57 (9H).
Part C:
Weinreb amide 2550 was prepared using procedures described in Example 511, Part A. 1H NMR (400 MHz, CDCI3) δ 7.37-7.14 (m, 3H), 6.1-5.88 (d, 1 H), 4.0 (s, 3H), 3.84-3.67 (m, 2H), 3.21 (s, 3H), 3.17-3.09 (m, 1 H), 2.79-2.72 (m, 1 H), 1.5 (s, 9H). HPLC-MS tR = 2.15 min (UV254 nm); mass calculated for formula C17H23CIN204 354.1 , observed LCMS m/z 255.1 (M-BOC+H).
Part D:
Acetylene 2551 was prepared using procedures described in Example 511, Part B. HPLC-MS tR = 2.22 min (UV254 nm); mass calculated for formula C17H18CIN03 319.1 , observed LCMS m/z 264.0 (M-(t-butyl)).
Part E:
Pyrazole 2552 was prepared using procedures described in Example 62G Part A. HPLC-MS tR = 1.97 min (UV254 nm); mass calculated for formula C17H20CIN3O2 333.1 , observed LCMS m/z 234 (M-BOC+H).
Part F:
Amine 2553 was prepared using a procedure similar to that described in Example 511, Part D. HPLC-MS tR = 0.89 min (UV254 nm); mass calculated for formula C12H12CIN3 233.1 , observed LCMS m/z 234.1 (M+H).
Example 62F:
Figure imgf000714_0001
Compound 2554 (3.7 g, 13.4 mmol) was dissolved in methylene chloride (35 mL) and Dess-Martin periodinane (8.55 g, 20.18 mmol) was added and stirred at room temperature overnight. The reaction was quenched with saturated sodium bicarbonate and stirred for 1 hour. The reaction mixture was filtered through celite. The aqueous layer was extracted with methylene chloride. The combined organic layers were dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 50% ethyl acetate/hexanes) afforded 2555 (1.75 g). 1H NMR (400 MHz, CDCI3) δ 7.40- 7.30 (m, 5 H), 5.25-5.20 (m, 2H), 4.90-4.80 (m, 1 H), 4.00-3.80 (m, 2H), 3.80- 3.60 (m, 3H), 3.00 (m, 1 H), 2.60 (m, 1 H).
Part B:
Compound 2556 was prepared according to the procedure in Bioorg. Med.
Chem. 2002, 10, 5, 1197-1206. 1H NMR (400 MHz, CDCI3) δ 8.25 (s, 1 H),
7.4-7.3 (m, 5 H), 5.3-5.15 (m, 3H), 4.7-4.55 (m, 2H), 4.3-4.2 (m, 2H), 1.3 (t,
3H).
Part C:
Compound 2557 was prepared from compound 2556 according to the procedure described in Example 10B Part B. 1H NMR (400 MHz, CDCI3) δ 8.00 (m, 1 H), 7.60-7.40 (m, 5H), 5.30-5.20 (m, 2H), 5.10 (m, 2H), 4.80-4.60 (m, 1 H). Part D:
Compound 2558 was prepared as described in Example 51 B Part C from compound 2557. 1H NMR (400 MHz, CDCI3) δ 8.00 (s, 1 H), 7.40-7.20 (m, 5H), 6.20 (s, 1 H), 5.30-5.00 (m, 3H), 4.70 (m, 2H), 3.25 (m, 2H), 1.30 (m, 3H).
Part E:
Compound 2558 (80 mg, 0.202 mmol) was dissolved in 30% HBr in acetic acid (3 mL) and stirred for 2 hours. The solvent was removed and the residue was dissolved in water and washed with ethyl acetate. The aqueous layer was lyophilized to provide 2559 as a red solid (100 mg). HPLC-MS tR = 0.414 min (UV254 nm); mass calculated for formula C11 H14N6S 262.1 , observed LCMS m/z 263.1 (M+H).
Example 62G:
Figure imgf000715_0001
2560 2561 2562
Part A:
Compound 2199 (1.11 g, 3.7 mmol), isopropyl hydrazine hydrochloride (616 mg, 5.55 mmol), and sodium carbonate (980 mg, 9.25 mmol) were dissolved in ethanol (12 mL) and stirred at 80 °C for 5 hours. The reaction mixture was cooled to room temperature and diluted with ethyl acetate and water. The aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with water, brine, dried over sodium sulfate and concentrated. Purification by column chromatography (Si02, 33% ethyl acetate/hexanes) afforded 2560 (750 mg). 1H NMR (400 MHz, CDCI3) .δ 7.35 (d, 1 H), 6.00 (d, 1 H), 5.10-4.90 (m, 1 H), 4.50 (m, 1 H), 3.60-3.40 (m, 2H), 2.30-2.10 (m, 1 H), 2.10-1.80 (m, 3H), 1.50 (m, 9H), 1.30 (s, 6H).
Part B: Compound 2560 (541 mg, 1.93 mmol) was dissolved in chloroform (10 mL) and N-bromosuccinimide (412 mg, 2.31 mmol) was added and stirred at room temperature overnight. The reaction was diluted with methylene chloride and water. The organic layer was washed with 1 N NaOH, brine, dried over sodium sulfate, and concentrated. Purification by column chromatography (Si02, 15% ethyl acetate/hexanes) afforded 2561 (350 mg). 1H NMR (400 MHz, CDCI3) δ 7.35 (s, 1 H), 5.00-4.80 (m, 1 H), 4.40 (m, 1 H), 3.60-3.40 (m, 2H), 2.30-2.20 (m, 1 H), 2.10-2.00 (m, 1 H), 2.00-1.80 (m, 2H), 1.40 (m, 9H), 1.20 (s, 6H).
Part C:
Compound 2561 (86 mg, 0.238 mmol) was dissolved in 4M HCI in dioxane (2.0 mL) and stirred for 1 hour. The solvent was removed to provide 2562 as a white solid (82 mg). 1H NMR (400 MHz, DMSO-d6) δ 9.60.(bs, 1 H), 8.80 (bs, 1 H), 8.18 (s, 1 H), 4.60 (m, 1 H), 4.50 (m, 1 H), 3.25 (m, 2H), 2.35 (m, 1 H), 2.10-1.90 (m, 3H), 1.40 (d, 6H).
Example 62H:
Figure imgf000716_0001
1 2563
Part A:
To 471 (81 mg, 0.30 mmol) and DIEA (0.156 mL, 0.90 mmol) in dichloromethane (5 mL) was added slowly acetyl chloride (0.052 mL, 0.72 mmol) at 0 °C. The reaction mixture was allowed to stir at room temperature for 2 hr and concentrated. The residue was then dissolved in EtOAc and H20, washed with 1 N citric acid, NaHCθ3, brine, dried over Na2S04, and concentrated. The resulting sticky solid was dissolved in methanol (5 mL) and stirred with solid K2C03 (100 mg) for 2 hr. After removing the solvent, the residue was dissolved in EtOAc and H20, washed with H20, brine, dried (Na2S04) and concentrated to afford 2563 (82 mg, 87 %) as an oily solid. HPLC-MS tR = 1.60 min (UV254 nm); mass calculated for formula C14H2ιN304S 311.1 , observed LCMS m/z 312.1 (M+H).
Example 62I:
Figure imgf000717_0001
2205 2564
Part A:
To compound 2205 (100 mg, 0.35 mmol) and DIEA (0.092 mL, 0.53 mmol) in dichloromethane (5 mL) was slowly added methylsulfonic chloride (0.033 mL, 0.42 mmol) at 0 °C. The reaction mixture was allowed to stir at room temperature for 2 hr and concentrated. The residue was dissolved in EtOAc, washed with water, 1 N citric acid, NaHC03, and brine, dried over Na24 and concentrated to afford 2564 (120 mg, 94%) as an oily solid. HPLC-MS tR = 1.96 min (UV254 nm); mass calculated for formula C-i4H23N304S2 361.1 , observed LCMS m/z 362.0 (M+H).
Example 62 J:
Figure imgf000717_0002
2205 2565
Part A:
To compound 2205 (80 mg, 0.28 mmol) and DIEA (0.098 mL, 0.56 mmol) in dichloromethane (5 mL) was added slowly ethyl chloroformate (0.054 mL, 0.56 mmol) at 0 °C. The reaction mixture was allowed to stir at room temperature for 2 hr and concentrated. The residue was dissolved in EtOAc, washed with water, 1 N citric acid, NaHC03 and brine, dried over Na2S0 and concentrated to afford 2565(88 mg, 88%) as an oily solid. HPLC-MS tR = 2.23 min (UV254 nm); mass calculated for formula CnH25N3θ4S 355.2, observed LCMS m/z 356.2 (M+H). The following compounds were prepared using previously described procedures.
Figure imgf000718_0001
Figure imgf000719_0001
Figure imgf000720_0001
Figure imgf000721_0001
Figure imgf000722_0001
It will be appreciated by those skilled in the art that changes could be made to the embodiments described above without departing from the broad inventive concept thereof. It is understood, therefore, that this invention is not limited to the particular embodiments disclosed, but it is intended to cover modifications that are within the spirit and scope of the invention, as defined by the appended claims.

Claims

ClaimsWhat is claimed is:
1. A compound of formula (I):
Figure imgf000723_0001
formula (I)
or a pharmaceutically acceptable salt or solvate thereof, wherein: A is selected from the group consisting of:
Figure imgf000723_0002
-C02R1;
d is 0 to 4;
J is selected from the group consisting of O, S, and NR5;
E is selected from the group consisting of: O, S, and NR5;
T is O or S;
R1 and R2 are the same or different, each being independently selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, and heteroaryl; or alternatively R1 and R2, taken together with the N to which R1 and R2 are shown attached, represent a 4-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein each of said alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, heteroaryl and 4-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties below;
R10 is selected from the group consisting of H, alkyl, and fluoroalkyl;
R20 is selected from the group consisting of H, alkyl, and fluoroalkyl;
R30 is H or alkyl, or alternatively R30 and R40 taken together with the N to which R40 is shown attached to in Formula I, are joined to form a 4-7 membered heterocylic ring, wherein said heterocylic ring is unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties below;
R40 is H or alkyl;
R50 is H or alkyl;
W is -(CR13 2)n-, wherein n is 0 to 5 or a covalent bond, or alternatively two R13 groups can fuse to form a 3-8 membered cycloalkyl, wherein said 3-8 membered cycloalkyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R6 moieties below;
X is absent or present, and if present X is selected from the group consisting of a covalent bond, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties below;
Y is absent or present, and if present Y is selected from the group consisting of a covalent bond, -[C(R6)2]n- wherein n is 1 or 2, -0-, -S-, -NR1-, - SOv- wherein v is 1 to 2, -SOn(CR6 2)p- wherein n is 1 or 2 and p is 1 to 4, - 0(CR6 2)q- or -(CR6 2)qO- wherein q is 1 to 4, -N(R7)S(0)n- or -S(0)nN(R7)- wherein n is 1 or 2, and -N(R7)C(0)- or -C(0)N(R7)-;
Z is selected from the group consisting of H, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, said cycloalkyl, heterocyclyl, aryl, and heteroaryl being optionally fused with aryl, heterocyclyl, heteroaryl or cycloalky; wherein each of said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties below;
R5 is selected from the group consisting of hydrogen, alkyl, and alkylaryl; each R6 is the same or different and is independently selected from the group consisting of hydrogen, halogen, -SR^, -S(0)qRl 5 wherein q is 1 to 2, alkyl, cycloalkyl, heterocyclyl, alkoxyl, hydroxy, nitro, cyano, amino, alkenyl, alkynyl, arylalkyl, aminocarbonyl, alkylcarbonyl, and alkoxycarbonyl; each R7 is the same or different and is independently selected from the group consisting of hydrogen, alkyl, aryl, cycloalkyl, heteroaryl, heterocyclyl, alkenyl, alkynyl, arylalkyl, alkylcarbonyl, and alkoxycarbonyl, wherein each of the aryl, heteroaryl and heterocyclyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties below;
R13 is the same or different and is independently selected from the group consisting of hydrogen, halogen, -OH, -OR14, alkyl, cycloalkyl, heterocyclyl, alkenyl, alkynyl, alkylaryl, alkylamino, and alkylcarbonyl;
14
R is alkyl; each R70 is a substituent for H where indicated and is the same or different and is independently selected from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, -CN, -CF3, -OCF3, -OR15, -C(0)R15, - C(0)OR15, -C(0)N(R15)(R16); _SR15J -S(0)qN(Rl5)(Rl6) wherein q is 1 to
2, -C(=N0R15)R16; -N(R 5)(Rl6)f -alkyl-N(R15)(R16), -N(R15)C(0)R16, . CH2-N(R15)C(0)R16, -N(R"l 5)S(0)R16, _N(R15)S(0)2R16, -
CH2-N(R15)S(0)2R16, -N(R1 7)S(0)2N(R16)(R15)J N(R17)S(0)N(R16)(R15); -N(R17)C(0)N(R16)(R15), -CH2-
N(R17)C(0)N(R16)(R15), -N(R15)C(0)OR16, -CH2-N(R15)C(0)OR ^, and -S(0)qR15 wherein q is 1 to 2; and wherein each of the alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkenyl and alkynyl are independently unsubstituted or substituted by 1 to 5 groups independently selected from the group consisting of alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, halo, -CF3, -CN, -OR15, -N(R15)(R16)f -C(0)OR 5,-
C(0)N(R15)(R16)) an -N(R15)S(0)R16; and
15 16 17 each R , R and R are independently selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, or alternatively R15 and R16 taken together with the N to which they are shown attached, are joined to form a 4-8 membered heterocylic ring, wherein said 4- 8 membered cycloalkyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R75 moieties below; each R75 is independently selected from the group consisting of alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkenyl and alkynyl, and wherein each of the alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkenyl and alkynyl are independently unsubstituted or substituted by 1 to 5 groups independently selected from the group consisting of alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, halo, -CF3,
1 19 19 19 19
-CN, -OR 9, -N(R )2, -C(0)OR , -C(0)N(R )2, and -N(R )S(0)R ; and each R19 is independently selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl.
2. The compound of claim 1 wherein A is selected from the group
Figure imgf000726_0001
wherein R and R , taken together with the N to which R1 and R2 are shown attached, represent a 4-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 4-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties.
3. The compound of claim 1 wherein A is
Figure imgf000727_0001
wherein R1 and R2 are the same or different, each being independently selected from the group consisting of H, alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl, and heteroaryl; wherein said alkyl, cycloalkyl, heterocyclyl, aryl, arylalkyl, heteroaralkyl and heteroaryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties.
4. The compound of claim 3 wherein A is
Figure imgf000727_0002
5. The compound of claim 2 wherein A is
Figure imgf000727_0003
wherein a is 0 to 4; and x is 0 to 4.
The compound of claim 2 wherein A is
Figure imgf000728_0001
wherein each x idependently is 0 to 4.
7. The compound of claim 2 wherein a ring is formed from -NR1R2 and said ring is
Figure imgf000728_0002
wherein each R9 is a substituent for H where indicated and can be the same or different, each being independently selected from the group consisting of -OH,
-OR14, -C(0)OR15, -C(0)N(R 5)(R16), alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl, wherein said alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties;
R11and R12 taken together with the carbon to which each R11and R12 are shown attached are fused heteroaryl or fused cycloalkyl, wherein said fused heteroaryl and fused cycloalkyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; and x is 0 to 4, and when x is greater than 1 , each R9 moiety can be the same or different, each moiety being independently selected from the group of R9 moieties.
8. The compound of claim 2 wherein a ring is formed from -NR1R2 and said ring is (R9)x wherein each R9 is a substituent for H where indicated and can be the same or different, each being independently selected from the group consisting of -OH,
-OR14, -C(0)OR15, -C(0)N(R15)(R16), alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl, wherein said alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; x is 0 to 4, and when x is greater than 1 , each R9 moiety can be the same or different, each moiety being independently selected from the group of R9 moieties; and
G is selected from the group consisting of CH2, NR7, O, S, or S02.
9. The compound of claim 2 wherein a ring is formed from -NR1R2 and said ring is selected from the group consisting of
Figure imgf000730_0001
wherein x is 0 to 4, and when x is greater than 1 , each R70 moiety can be the same or different, each moiety being independently selected from the group of R70 moieties; and each R18 is the same or different and is independently H or alkyl.
10. The compound of claim 2 wherein a ring is formed from -NR1R2 and said ring is selected from the group consisting of:
Figure imgf000731_0001
x is 0 to 4, and when x is greater than 1 , each R70 moiety can be the same or different, each moiety being independently selected from the group of R70 moieties.
11. The compound of claim 2 wherein a ring is formed from -NR1R2 and is
Figure imgf000732_0001
wherein each R9 is a substituent for H where indicated and can be the same or different, each being independently selected from the group consisting of -OH, -OR14, -C(0)OR15, -C(0)N(R15)(R16), alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl, wherein said alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; and x is 0 to 4, and when x is greater than 1 , each R9moiety can be the same or different, each moiety being independently selected from the group of R9moieties.
12. The compound of claim 2 wherein J is O.
13. The compound of claim 2 wherein E is O.
14. The compound of claim 2 wherein R10 is H or alkyl.
15. The compound of claim 2 wherein R20 is H.
16. The compound of claim 2 wherein R30 is H.
17. The compound of claim 2 wherein R40 is H.
18. The compound of claim 2 wherein R50 is H.
19. The compound of claim 2 wherein R13 is H or alkyl.
20. The compound of claim 19 wherein R13 is H or -CH3.
21. The compound of claim 2 wherein R1 and R2, taken together with the N to which R1 and R2 are shown attached, represent a 4-6 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with R70, wherein R70 is aryl.
22. The compound of claim 2 wherein Y is a covalent bond or - . [C(R6)2]n- wherein n is 1 to 2.
23. The compound of claim 2 wherein Y is selected from the group consisting of a covalent bond, -CH2-, -C(H)(OH)-, -C(O)- and -0-.
24. The compound of claim 23 wherein Y is a covalent bond or - CH2-.
25. The compound of claim 24 wherein Y is a covalent bond.
26. The compound of claim 2 wherein W is -(CR13 2)n-, in which n is 1-5 and each R13 is H, or alkyl.
27. The compound of claim 2 wherein W is selected from the group consisting of -CH2-, -C(H)(CH3)-, -C(CH3)2- and -CH2CH2-.
28. The compound of claim 27 wherein W is -CH2- or -C(H)(CH3)-.
29. The compound of claim 2 wherein X is selected from the group consisting of alkyl, aryl, heterocyclyl and heteroaryl.
30. The compound of claim 29 wherein X is selected from the group consisting of phenyl, azetidinyl, pyrrolidinyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl, imidazolyl and pyrazolyl.
31. The compound of claim 30 wherein X is selected from the group consisting of phenyl, piperidinyl and pyridinyl.
32. The compound of claim 29 wherein X is selected from the group consisting of
Figure imgf000734_0001
wherein x is 0 to 4, and wherein x is greater than 1 , each R70 moiety can be the same or different, each moiety being independently selected from the group of R70 moieties.
33. The compound of claim 29, wherein X is selected from the group consisting of
Figure imgf000735_0001
wherein x is 0 to 4, and when x is greater than 1 , each R70 moiety can be the same or different, each moiety being independently selected from the group of R70 moieties.
34. The compound of claim 2 wherein Z is selected from the group consisting of H, aryl and heteroaryl.
35. The compound of claim 34 wherein Z is selected from the group consisting of H, phenyl, indolyl, benzimidazolyl, pyrazolyl, thienyl, pyridinyl, thiazolyl, thiadiazolyl, imidazolyl, pyrrolidinyl, pyrazinyl, triazolyl, tetrazolyl and tetrazinyl, wherein said phenyl, indolyl, benzimidazolyl, pyrazolyl, thienyl, pyridinyl, thiazolyl, thiadiazolyl, imidazolyl, pyrrolidinyl, pyrazinyl, triazolyl, tetrazolyl and tetrazinyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group consisting of R70 moieties.
36. The compound of claim 35 wherein Z is selected from the group consisting of
Figure imgf000736_0001
wherein x is 0 to 4, and when x is greater than 1 , each R70 moiety can be the same or different, each moiety being independently selected from the group of R70 moieties.
37. The compound of claim 35 wherein Z is phenyl.
38. The compound of claim 37 wherein said phenyl is substituted with at least one substituent selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy, amino and tetrazole.
39. The compound of claim 35 wherein Z is thienyl.
40. The compound of claim 39 wherein said thienyl is substituted with at least one substituent selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy, amino and tetrazole.
41. The compound of claim 35 wherein Z is pyrazolyl.
42. The compound of claim 41 wherein said pyrazolyl is substituted with at least one substituent selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy, amino and tetrazole.
43. The compound of claim 35 wherein Z is pyridinyl.
44. The compound of claim 43 wherein said pyridinyl is substituted with at least one substituent selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy, amino and tetrazole.
45. The compound of claim 35 wherein Z is imidazolyl.
46. The compound of claim 45 wherein said imidazolyl is substituted with at least one substituent selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy, amino and tetrazole.
47. The compound of claim 2 wherein A is
Figure imgf000737_0001
wherein R and R , taken together with the N to which R1 and R2 are shown attached, represent a 4-8 membered heterocyclic ring having 1-3 heteroatoms including said N, said heterocyclic ring being optionally substituted with one or more R70, or optionally fused with aryl, heteroaryl, cycloalkyl, or heterocyclyl, wherein said 4-8 membered heterocyclic ring can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; E, J and T are the same and are O; R10 is H or alkyl;
R20, R30, R40, and R50 are the same and are H; W is -(CR13 2)n-, wherein n is 0 to 5; X is selected from the group consisting of aryl, heteroaryl and heterocyclyl; Y is selected from the group consisting of a covalent bond, -
[^(R6)^- wherein n is 1 to 2, -0-, -S-, and -NR1-; and Z is selected from the group consisting of
Figure imgf000738_0001
48. The compound of claim 47 wherein the -NR >1'DR2 of
Figure imgf000738_0002
IS
Figure imgf000739_0001
wherein each R9 is a substituent for H where indicated and can be the same or different, each being independently selected from the group consisting of -OH,
-OR14, -C(0)OR15, -C(0)N(R1 5)(R 6), alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl, wherein said alkyl, aryl, heteroaryl, alkenyl, alkynyl, cycloalkyl and heterocyclyl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different, each moiety being independently selected from the group of R70 moieties; and x is 0 to 4, and when x is greater than 1 , each R9moiety can be the same or different, each moiety being independently selected from the group of R9moieties.
49. The compound of claim 47 wherein X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl, and pyrazolyl.
50. The compound of claim 48 wherein X is selected from the group consisting of phenyl, piperidinyl, thienyl, and pyridinyl and Y is selected from the group consisting of a covalent bond, -[C(R6)2]n- wherein n is 1 to 2 and -O-
51. The compound of claim 48 wherein X is piperidinyl, Y is a covalent bond, and Z is aryl or heteroaryl having two substituents which can be the same or different, each moiety being independently selected from the group consisting of cyano, alkoxy, halogen, alkyl, haloalkyl, hydroxy, aryl, heteroaryl, aryloxy and amino.
52. The compound of claim 47 wherein W is selected from the group consisting of -CH2-, -C(H)CH3-, -C(CH3)2- and Y is a covalent bond.
53. The compound of claim 47 wherein W is -CH2- or -C(H)(CH3)- and Y is -CH2-.
54. The compound of claim 47 wherein X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl and pyrazolyl; Y is -[C(R6)2]n- wherein n is 1 to 2; and Z is selected from the group consisting of
Figure imgf000741_0001
55. The compound of claim 47 wherein X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl and pyrazolyl; Y is selected from the group consisting of 0-, -S-, and -NR1-; and Z is selected from the group consisting of
Figure imgf000742_0001
56. The compound of claim 47 wherein W is -(CR13 2)n- wherein n is 1 or 2; X is selected from the group consisting of phenyl, piperidinyl, pyridinyl, thienyl, thiazolyl, oxazolyl and pyrazolyl; Y is a covalent bond and Z is selected from the group consisting of
Figure imgf000743_0001
Figure imgf000743_0002
Figure imgf000743_0003
57. The compound of claim 53 wherein Z is selected from the group consisting of
Figure imgf000743_0004
58. The compound of claim 54 wherein Z is selected from the group consisting of
Figure imgf000744_0001
59. The compound of claim 55 wherein Z is selected from the group consisting of
Figure imgf000744_0002
60. A compound exhibiting TACE, TNF-α, MMP or any combination thereof inhibitory activity, including enantiomers, stereoisomers and tautomers of said compound, and pharmaceutically acceptable salts or solvates of said compound, said compound being selected from the compounds of structures listed below:
Figure imgf000745_0001
Figure imgf000746_0001
Figure imgf000747_0001
Figure imgf000748_0001
Figure imgf000749_0001
Figure imgf000750_0001
Figure imgf000751_0001
Figure imgf000751_0002
Figure imgf000751_0003
Figure imgf000751_0004
Figure imgf000751_0005
Figure imgf000751_0006
Figure imgf000752_0001
Figure imgf000753_0001
Figure imgf000754_0001
Figure imgf000755_0001
Figure imgf000756_0001
Figure imgf000757_0001
Figure imgf000758_0001
Figure imgf000759_0001
Figure imgf000760_0001
Figure imgf000761_0001
Figure imgf000762_0001
Figure imgf000763_0001
Figure imgf000764_0001
763
Figure imgf000765_0001
Figure imgf000766_0001
Figure imgf000767_0001
Figure imgf000768_0001
Figure imgf000769_0001
Figure imgf000770_0001
Figure imgf000771_0001
Figure imgf000772_0001
Figure imgf000773_0001
Figure imgf000774_0001
Figure imgf000775_0001
Figure imgf000776_0001
Figure imgf000777_0001
Figure imgf000778_0001
Figure imgf000779_0001
Figure imgf000780_0001
Figure imgf000781_0001
Figure imgf000782_0001
Figure imgf000783_0001
Figure imgf000784_0001
Figure imgf000785_0001
Figure imgf000786_0001
Figure imgf000787_0001
Figure imgf000788_0001
Figure imgf000789_0001
Figure imgf000790_0001
Figure imgf000791_0001
Figure imgf000792_0001
Figure imgf000793_0001
Figure imgf000794_0001
Figure imgf000795_0001
Figure imgf000796_0001
Figure imgf000797_0001
Figure imgf000797_0002
Figure imgf000797_0003
Figure imgf000797_0004
Figure imgf000797_0005
Figure imgf000797_0006
Figure imgf000798_0001
Figure imgf000799_0001
Figure imgf000799_0002
jorΛft' σ°
Figure imgf000799_0003
Figure imgf000799_0004
Figure imgf000799_0005
Figure imgf000799_0006
Figure imgf000799_0007
Figure imgf000799_0008
Figure imgf000800_0001
Figure imgf000800_0002
Figure imgf000800_0003
Figure imgf000800_0004
Figure imgf000800_0005
Figure imgf000800_0006
Figure imgf000800_0007
Figure imgf000800_0008
ΛV<vo σ1
Figure imgf000801_0001
Figure imgf000801_0002
Figure imgf000801_0003
Figure imgf000801_0004
Figure imgf000801_0005
Figure imgf000801_0006
iYttø ø' °
A
Figure imgf000801_0007
Figure imgf000802_0001
Figure imgf000802_0002
Figure imgf000802_0003
Figure imgf000802_0004
Vua
Figure imgf000802_0005
Figure imgf000802_0006
Figure imgf000802_0007
Figure imgf000802_0008
Figure imgf000803_0001
Figure imgf000803_0002
Figure imgf000803_0003
Figure imgf000803_0004
Figure imgf000803_0005
V
O' ΛVVy-O
Figure imgf000803_0006
Figure imgf000803_0007
Figure imgf000803_0008
Figure imgf000804_0001
Figure imgf000805_0001
Figure imgf000805_0002
Figure imgf000806_0001
Figure imgf000806_0002
Figure imgf000806_0003
Figure imgf000806_0004
Figure imgf000807_0001
Figure imgf000807_0002
O OH .
VJ OHY N O
Figure imgf000807_0003
Figure imgf000808_0001
Figure imgf000809_0001
Figure imgf000810_0001
Figure imgf000811_0001
Figure imgf000812_0001
Figure imgf000813_0001
Figure imgf000814_0001
Figure imgf000815_0001
Figure imgf000816_0001
Figure imgf000817_0001
Figure imgf000818_0001
Figure imgf000819_0001
Figure imgf000820_0001
Figure imgf000821_0001
Figure imgf000822_0001
Figure imgf000823_0001
Figure imgf000824_0001
Figure imgf000825_0001
Figure imgf000826_0001
Figure imgf000827_0001
Figure imgf000828_0001
Figure imgf000829_0001
Figure imgf000831_0001
Figure imgf000832_0001
Figure imgf000833_0001
Figure imgf000834_0001
Figure imgf000835_0001
Figure imgf000836_0001
Figure imgf000837_0001
Figure imgf000839_0001
Figure imgf000840_0001
Figure imgf000841_0001
Figure imgf000842_0001
Figure imgf000843_0001
Figure imgf000844_0001
Figure imgf000845_0001
Figure imgf000846_0001
Figure imgf000847_0001
Figure imgf000848_0001
Figure imgf000849_0001
Figure imgf000850_0001
Figure imgf000851_0001
Figure imgf000852_0001
Figure imgf000853_0001
Figure imgf000854_0001
Figure imgf000855_0001
Figure imgf000856_0001
Figure imgf000857_0001
Figure imgf000858_0001
Figure imgf000859_0001
Figure imgf000860_0001
61. The compound of claim 60, said compound being selected from the compounds of structures listed below:
Figure imgf000861_0001
Figure imgf000862_0001
Figure imgf000863_0001
Figure imgf000864_0001
Figure imgf000865_0001
Figure imgf000866_0001
Figure imgf000867_0001
Figure imgf000868_0001
Figure imgf000869_0001
Figure imgf000870_0001
Figure imgf000871_0001
Figure imgf000872_0001
Figure imgf000873_0001
Figure imgf000874_0001
Figure imgf000874_0002
Figure imgf000875_0001
Figure imgf000876_0001
Figure imgf000877_0001
Figure imgf000878_0001
Figure imgf000879_0001
Figure imgf000880_0001
Figure imgf000881_0001
Figure imgf000882_0001
62. The compound of claim 61 , said compound being selected from the compounds of structures listed below:
Figure imgf000882_0002
Figure imgf000883_0001
Figure imgf000884_0001
Figure imgf000885_0001
63. A pharmaceutical composition comprising as an active ingredient at least one compound of claim 1.
64. The pharmaceutical composition of claim 1 additionally comprising at least one pharmaceutically acceptable carrier.
65. A method of treating disorders associated with tumor necrosis factor-alpha-converting enzyme (TACE), tumor necrosis factor-alpha (TNF-α), matrix metalloproteinases (MMPs), a disintegrin and metalloproteases (ADAMs) or any combination thereof, said method comprising administering to a patient in need of such treatment a pharmaceutical composition which comprises therapeutically effective amounts of at least one compound of claim 1.
66. The use of a compound of claim 1 for the manufacture of a medicament to treat disorders associated with TACE, TNF-α, MMPs, ADAMs or any combination thereof.
67. A method of preparing a pharmaceutical composition for treating the disorders associated with TACE, TNF-α, MMPs, ADAMs or any combination thereof, said method comprising bringing into intimate contact at least one compound of claim 1 and at least one pharmaceutically acceptable carrier.
68. A pharmaceutical composition for treating disorders associated with TACE, TNF-α, MMP, ADAM or any combination thereof in a subject comprising, administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
69. A compound of claim 1 in purified form.
70. A method of treating a condition or disease mediated by TACE, MMPs, TNF-σ, aggrecanase, or any combination thereof in a subject comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
71. The method of claim 70, wherein said aggrecanase is aggrecanase 1 or aggrecanase 2.
72. A method of treating a condition or disease selected from the group consisting of rheumatoid arthritis, osteoarthritis, periodontitis, gingivitis, corneal ulceration, solid tumor growth and tumor invasion by secondary metastases, neovascular glaucoma, inflammatory bowel disease, multiple sclerosis and psoriasis in a subject, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
73. A method of treating a condition or disease selected from the group consisting of fever, cardiovascular conditions, hemorrhage, coagulation, cachexia, anorexia, alcoholism, acute phase response, acute infection, shock, graft versus host reaction, autoimmune disease and HIV infection in a subject comprising administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
74. A method of treating a condition or disease selected from the group consisting of septic shock, haemodynamic shock, sepsis syndrome, post ischaemic reperfusion injury, malaria, mycobacterial infection, meningitis, psoriasis, congestive heart failure, fibrotic diseases, cachexia, graft rejection, cancers such as cutaneous T-cell lymphoma, diseases involving angiogenesis, autoimmune diseases, skin inflammatory diseases, inflammatory bowel diseases such as Crohn's disease and colitis, osteo and rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, adult Still's disease, ureitis, Wegener's granulomatosis, Behcehe disease, Sjogren's syndrome, sarcoidosis, polymyositis, dermatomyositis, multiple sclerosis, sciatica, complex regional pain syndrome, radiation damage, hyperoxic alveolar injury, periodontal disease, HIV, non-insulin dependent diabetes mellitus, systemic lupus erythematosus, glaucoma, sarcoidosis, idiopathic pulmonary fibrosis, bronchopulmonary dysplasia, retinal disease, scleroderma, osteoporosis, renal ischemia, myocardial infarction, cerebral stroke, cerebral ischemia, nephritis, hepatitis, glomerulonephritis, cryptogenic fibrosing aveolitis, psoriasis, transplant rejection, atopic dermatitis, vasculitis, allergy, seasonal allergic rhinitis, reversible airway obstruction, adult respiratory distress syndrome, asthma, chronic obstructive pulmonary disease (COPD) and bronchitis in a subject comprising administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
75. A method of treating a condition or disease associated with COPD, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
76. A method of treating a condition or disease associated with rheumatoid arthritis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
77. A method of treating a condition or disease associated with Crohn's disease, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
78. A method of treating a condition or disease associated with psoriasis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
79. A method of treating a condition or disease associated with ankylosing spondylitis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
80. A method of treating a condition or disease associated with sciatica, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
81. A method of treating a condition or disease associated with complex regional pain syndrome, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
82. A method of treating a condition or disease associated with psoriatic arthritis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof.
83. A method of treating a condition or disease associated with multiple sclerosis, comprising: administering to the subject in need of such treatment a therapeutically effective amount of at least one compound of claim 1 or a pharmaceutically acceptable salt, solvate or isomer thereof, in combination with a compound selected from the group consisting of AvonexD, Betaseron, Copaxone or other compounds indicated for the treatment of multiple sclerosis.
84. The method of claim 71 further comprising administering to said subject a therapeutically effective amount of at least one medicament selected from the group consisting of disease modifying anti-rheumatic drugs (DMARDS), NSAIDs, COX-2 inhibitors, COX-1 inhibitors, immunosuppressives, biological response modifiers (BRMs), anti-infammatory agents and H1 antagonists.
85. A method of claim 73, further comprising administering to said subject a therapeutically effective amount of at least one medicament selected from the group consisting of DMARDS, NSAIDs, COX-2 inhibitors, COX-1 inhibitors, immunosuppressives, BRMs, anti-infammatory agents and H1 antagonists.
86. A method of claim 75, further comprising administering to said subject a therapeutically effective amount of at least one medicament selected from the group consisting of DMARDS, NSAIDs, COX-2 inhibitors, COX-1 inhibitors, immunosuppressives, BRMs, anti-infammatory agents and H1 antagonists.
PCT/US2005/019131 2004-06-02 2005-06-01 Chemical compounds and pharmaceutical compositions containing them for the treatment of inflammatory disorders WO2005121130A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
CA002569111A CA2569111A1 (en) 2004-06-02 2005-06-01 Chemical compounds and pharmaceutical compositions containing them for the treatment of inflammatory disorders
JP2007515501A JP2008501691A (en) 2004-06-02 2005-06-01 Pharmaceutical compounds and pharmaceutical compositions comprising the chemical compounds for treating inflammatory diseases
AU2005252201A AU2005252201A1 (en) 2004-06-02 2005-06-01 Chemical compounds and pharmaceutical compositions containing them for the treatment of inflammatory disorders
MXPA06014054A MXPA06014054A (en) 2004-06-02 2005-06-01 Compounds for the treatment of inflammatory disorders.
EP05759261A EP1773821A2 (en) 2004-06-02 2005-06-01 Chemical compounds and pharmaceutical compositions containing them for the treatment of inflammatory disorders
IL179674A IL179674A0 (en) 2004-06-02 2006-11-28 Chemical compounds and pharmaceutical compositions containing them for the treatment of inflammatory disorders

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US57615304P 2004-06-02 2004-06-02
US60/576,153 2004-06-02

Publications (2)

Publication Number Publication Date
WO2005121130A2 true WO2005121130A2 (en) 2005-12-22
WO2005121130A3 WO2005121130A3 (en) 2006-07-20

Family

ID=35355800

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/019131 WO2005121130A2 (en) 2004-06-02 2005-06-01 Chemical compounds and pharmaceutical compositions containing them for the treatment of inflammatory disorders

Country Status (14)

Country Link
US (2) US7652020B2 (en)
EP (1) EP1773821A2 (en)
JP (1) JP2008501691A (en)
KR (1) KR20070103671A (en)
CN (1) CN101027295A (en)
AR (1) AR051172A1 (en)
AU (1) AU2005252201A1 (en)
CA (1) CA2569111A1 (en)
IL (1) IL179674A0 (en)
MX (1) MXPA06014054A (en)
PE (1) PE20060426A1 (en)
TW (1) TW200612911A (en)
WO (1) WO2005121130A2 (en)
ZA (1) ZA200610055B (en)

Cited By (73)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007064749A1 (en) 2005-12-01 2007-06-07 Schering Corporation Compounds for the treatment of inflammatory disorders and microbial diseases
WO2007064732A1 (en) * 2005-12-01 2007-06-07 Schering Corporation Compounds for the treatment of inflammatory disorders and microbial diseases
US7598257B2 (en) 2005-12-13 2009-10-06 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as janus kinase inhibitors
US7834022B2 (en) 2007-06-13 2010-11-16 Incyte Corporation Metabolites of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
EP2272846A1 (en) 2009-06-23 2011-01-12 Bayer CropScience AG Thiazolylpiperidine derivatives as fungicide
EP2351743A1 (en) * 2008-10-27 2011-08-03 Takeda Pharmaceutical Company Limited Bicyclic compound
US8158616B2 (en) 2008-03-11 2012-04-17 Incyte Corporation Azetidine and cyclobutane derivatives as JAK inhibitors
WO2012118850A1 (en) 2011-02-28 2012-09-07 Array Biopharma Inc. Serine/threonine kinase inhibitors
WO2012135581A1 (en) * 2011-03-31 2012-10-04 Idenix Pharmaceuticals, Inc. Methods for treating drug-resistant hepatitis c virus infection with a 5,5-fused arylene or heteroarylene hepatitis c virus inhibitor
US8513270B2 (en) 2006-12-22 2013-08-20 Incyte Corporation Substituted heterocycles as Janus kinase inhibitors
US8563541B2 (en) 2005-09-22 2013-10-22 Incyte Corporation Azepine inhibitors of Janus kinases
WO2014047020A1 (en) 2012-09-19 2014-03-27 Novartis Ag Dihydropyrrolidino-pyrimidines as kinase inhibitors
US8722693B2 (en) 2007-06-13 2014-05-13 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US8987242B2 (en) 2008-12-05 2015-03-24 Merck Sharp & Dohme Corp. Morpholinone compounds as factor IXA inhibitors
WO2015042297A1 (en) 2013-09-20 2015-03-26 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Compounds for treating prostate can cancer
US9187496B2 (en) 2009-12-18 2015-11-17 Idenix Pharmaceuticals Llc 5,5-fused arylene or heteroarylene hepatitis C virus inhibitors
WO2015172196A1 (en) * 2014-05-13 2015-11-19 Monash University Heterocyclic compounds and use of same
US9358229B2 (en) 2011-08-10 2016-06-07 Novartis Pharma Ag JAK PI3K/mTOR combination therapy
US9512161B2 (en) 2009-10-09 2016-12-06 Incyte Corporation Hydroxyl, keto, and glucuronide derivatives of 3-(4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US9611267B2 (en) 2012-06-13 2017-04-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US9611269B2 (en) 2011-06-20 2017-04-04 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US9623029B2 (en) 2009-05-22 2017-04-18 Incyte Holdings Corporation 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]octane- or heptane-nitrile as JAK inhibitors
US9655854B2 (en) 2013-08-07 2017-05-23 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9714233B2 (en) 2013-03-06 2017-07-25 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US9718834B2 (en) 2011-09-07 2017-08-01 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US9745311B2 (en) 2012-08-10 2017-08-29 Incyte Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9801889B2 (en) 2015-02-20 2017-10-31 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
EP2116526B1 (en) * 2006-12-29 2018-04-18 Institute of Pharmacology and Toxicology Academy of Military Medical Sciences P.L.A. China Derivates of substituted tartaric aicd and usage for preparating beta secretase inhibitors
US9993480B2 (en) 2011-02-18 2018-06-12 Novartis Pharma Ag mTOR/JAK inhibitor combination therapy
US9999619B2 (en) 2010-03-10 2018-06-19 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US10004730B2 (en) 2011-10-12 2018-06-26 University of Pittsburgh—of the Commonwealth System of Higher Education Small molecules targeting androgen receptor nuclear localization and/or level in prostate cancer
US10040790B2 (en) 2013-04-19 2018-08-07 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10166191B2 (en) 2012-11-15 2019-01-01 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US10213427B2 (en) 2010-12-22 2019-02-26 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US10471108B2 (en) 2015-11-20 2019-11-12 4D Pharma Research Limited Compositions comprising bacterial strains
US10485830B2 (en) 2016-12-12 2019-11-26 4D Pharma Plc Compositions comprising bacterial strains
US10493112B2 (en) 2015-06-15 2019-12-03 4D Pharma Research Limited Compositions comprising bacterial strains
US10500237B2 (en) 2015-06-15 2019-12-10 4D Pharma Research Limited Compositions comprising bacterial strains
US10544110B2 (en) 2013-09-20 2020-01-28 University of Pittsburgh—of the Commonwealth System of Higher Education Small molecule inhibitors of the nuclear translocation of androgen receptor for the treatment of castration-resistant prostate cancer
US10583158B2 (en) 2016-03-04 2020-03-10 4D Pharma Plc Compositions comprising bacterial strains
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US10610549B2 (en) 2016-07-13 2020-04-07 4D Pharma Plc Composition comprising bacterial strains
US10610550B2 (en) 2015-11-20 2020-04-07 4D Pharma Research Limited Compositions comprising bacterial strains
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US10640506B2 (en) 2010-11-19 2020-05-05 Incyte Holdings Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidines derivatives as JAK inhibitors
US10736926B2 (en) 2015-06-15 2020-08-11 4D Pharma Research Limited Compositions comprising bacterial strains
US10758543B2 (en) 2010-05-21 2020-09-01 Incyte Corporation Topical formulation for a JAK inhibitor
US10780134B2 (en) 2015-06-15 2020-09-22 4D Pharma Research Limited Compositions comprising bacterial strains
US10851137B2 (en) 2013-04-10 2020-12-01 4D Pharma Research Limited Polypeptide and immune modulation
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10899736B2 (en) 2018-01-30 2021-01-26 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US10973872B2 (en) 2014-12-23 2021-04-13 4D Pharma Research Limited Pirin polypeptide and immune modulation
US10980806B2 (en) 2016-03-24 2021-04-20 University of Pittsburgh—of the Commonwealth System of Higher Education Small molecule inhibitors of the nuclear translocation of androgen receptor for the treatment of castration-resistant prostate cancer
US10987387B2 (en) 2017-05-24 2021-04-27 4D Pharma Research Limited Compositions comprising bacterial strain
US11007233B2 (en) 2017-06-14 2021-05-18 4D Pharma Research Limited Compositions comprising a bacterial strain of the genus Megasphera and uses thereof
US11084818B2 (en) 2016-11-11 2021-08-10 Hepo Pharmaceutical Co., Ltd. Nitrogen-containing heterocyclic compound, preparation method, intermediate, pharmaceutical composition and use
US11123379B2 (en) 2017-06-14 2021-09-21 4D Pharma Research Limited Compositions comprising bacterial strains
US11123378B2 (en) 2017-05-22 2021-09-21 4D Pharma Research Limited Compositions comprising bacterial strains
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11224620B2 (en) 2016-07-13 2022-01-18 4D Pharma Plc Compositions comprising bacterial strains
US11266698B2 (en) 2011-10-07 2022-03-08 4D Pharma Research Limited Bacterium for use as a probiotic for nutritional and medical applications
US11304949B2 (en) 2018-03-30 2022-04-19 Incyte Corporation Treatment of hidradenitis suppurativa using JAK inhibitors
US11389493B2 (en) 2015-06-15 2022-07-19 4D Pharma Research Limited Compositions comprising bacterial strains
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
US11723933B2 (en) 2014-12-23 2023-08-15 Cj Bioscience, Inc. Composition of bacteroides thetaiotaomicron for immune modulation
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
US11939331B2 (en) 2021-06-09 2024-03-26 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors

Families Citing this family (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PE20060426A1 (en) * 2004-06-02 2006-06-28 Schering Corp TARTARIC ACID DERIVATIVES AS INHIBITORS OF MMPs, ADAMs, TACE AND TNF-alpha
US20070167426A1 (en) * 2004-06-02 2007-07-19 Schering Corporation Compounds for the treatment of inflammatory disorders and microbial diseases
CN101282962A (en) * 2005-08-12 2008-10-08 先灵公司 Compounds for the treatment of inflammatory disorders
PL2155736T3 (en) 2007-05-14 2012-09-28 Sk Biopharmaceuticals Co Ltd Novel carbamoyloxy arylalkanoyl arylpiperazine compound, pharmaceutical compositions comprising the compound and method for treating pain, anxiety and depression by administering the compound
CA2728767A1 (en) * 2008-06-26 2009-12-30 Amgen Inc. Alkynyl alcohols as kinase inhibitors
US8642582B2 (en) * 2008-12-05 2014-02-04 Merck Sharp & Dohme Corp. Morpholinone compounds as factor IXa inhibitors
TW201038559A (en) * 2009-04-09 2010-11-01 Bristol Myers Squibb Co Hepatitis C virus inhibitors
AU2010249380B2 (en) 2009-05-22 2015-08-20 Incyte Holdings Corporation N-(hetero)aryl-pyrrolidine derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines and pyrrol-3-yl-pyrrolo[2,3-d]pyrimidines as Janus kinase inhibitors
US9249145B2 (en) 2009-09-01 2016-02-02 Incyte Holdings Corporation Heterocyclic derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
CN102127061B (en) * 2010-01-15 2016-02-17 浙江华海药业股份有限公司 One prepares improving one's methods of fluoro-3, the 4-dihydro-2 H-1-benzopyran-2-epoxy ethanes of 6-
CN102190647A (en) * 2010-03-12 2011-09-21 浙江海翔药业股份有限公司 Preparation method of nebivolol intermediate
US9034884B2 (en) 2010-11-19 2015-05-19 Incyte Corporation Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as JAK inhibitors
CN103492384B (en) * 2011-02-25 2016-05-11 诺华股份有限公司 As compound and the composition of TRK inhibitor
TW201313721A (en) 2011-08-18 2013-04-01 Incyte Corp Cyclohexyl azetidine derivatives as JAK inhibitors
US10130635B2 (en) 2012-05-04 2018-11-20 Rhizen Pharmaceuticals Sa Process for preparation of optically pure and optionally substituted 2-(1-hydroxy-alkyl)-chromen-4-one derivatives and their use in preparing pharmaceuticals
SI2844647T1 (en) * 2012-05-04 2020-12-31 Rhizen Pharmaceuticals S.A. Process for preparation of optically pure and optionally substituted 2-(1-hydroxy-alkyl)-chromen-4-one derivatives and their use in preparing pharmaceuticals
AR091079A1 (en) 2012-05-18 2014-12-30 Incyte Corp DERIVATIVES OF PIRROLOPIRIMIDINA AND PIRROLOPIRIDINA REPLACED WITH PIPERIDINILCICLOBUTILO AS JAK INHIBITORS
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
CN103772233B (en) * 2014-01-06 2015-06-10 华东师范大学 Multi-halothane five-member cyclic nitrone derivative and preparation method thereof
WO2015184305A1 (en) 2014-05-30 2015-12-03 Incyte Corporation TREATMENT OF CHRONIC NEUTROPHILIC LEUKEMIA (CNL) AND ATYPICAL CHRONIC MYELOID LEUKEMIA (aCML) BY INHIBITORS OF JAK1
CN104894013B (en) * 2015-05-19 2018-04-20 辽宁师范大学 Lamprey oral gland parasitism bacterial strain LJ1, secretory protein, separation method and purposes
CN108017571A (en) * 2016-10-29 2018-05-11 浙江九洲药物科技有限公司 A kind of preparation method for substituting isoindoline compounds
EP3684375A4 (en) 2017-09-22 2021-09-22 John Mansell Compositions and methods for treatment of sepsis-related disorders
CN107474006A (en) * 2017-09-25 2017-12-15 上海馨远医药科技有限公司 A kind of preparation method of 5 bromine isoindoline hydrochloride
WO2021007269A1 (en) 2019-07-09 2021-01-14 Incyte Corporation Bicyclic heterocycles as fgfr inhibitors
WO2021076728A1 (en) 2019-10-16 2021-04-22 Incyte Corporation Bicyclic heterocycles as fgfr inhibitors
US11945796B2 (en) 2020-09-16 2024-04-02 Nura Bio, Inc. Substituted pyridine derivatives as SARM1 inhibitors
CN112479936A (en) * 2020-12-16 2021-03-12 怀化学院 Preparation method of benzil derivative nitrone
CN113307765A (en) * 2021-05-24 2021-08-27 上海泰坦科技股份有限公司 Pyridine methylamine compound and preparation method thereof
TW202313009A (en) 2021-07-28 2023-04-01 美商努拉生物科技公司 Substituted pyridine derivatives as sarm1 inhibitors

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR1484601A (en) * 1955-01-22 1967-06-16 Leinfelder Hermann Process for preparing polyelectrolytes
US5618842A (en) * 1991-12-31 1997-04-08 Lifegroup S.P.A. N-acyl derivatives of aminoalcohols with polycarboxylic acids able to modulate mast cells in inflammatory processes having neuroimmunogenic origin
WO2001021661A1 (en) * 1999-09-20 2001-03-29 Novo Nordisk A/S BIVALENT INHIBITOR OF FVIIa/TF/FXa COMPLEX
WO2001077075A2 (en) * 2000-04-06 2001-10-18 Inotek Corporation Inhibitors of inflammation and reperfusion injury
US6344457B1 (en) * 1997-04-22 2002-02-05 Chiesi Farmaceutici S.P.A. Amino acid derivatives inhibiting extracellular matrix metalloproteinase and TNF alpha release
US6495565B2 (en) * 2000-03-17 2002-12-17 Bristol-Myers Squibb Pharma Company β-amino acid derivatives as inhibitors of matrix metalloproteases and TNF-α
US6534491B2 (en) * 1996-10-16 2003-03-18 American Cyanamid Company Preparation and use of ortho-sulfonamido bicyclic heteroaryl hydroxamic acids as matrix metalloproteinase and tace inhibitors
WO2003076461A2 (en) * 2002-03-12 2003-09-18 Novo Nordisk A/S Dimeric tf antagonist comprising two factor vii polypeptides
US6677355B1 (en) * 1999-08-18 2004-01-13 Warner-Lambert Company Hydroxamic acid compounds useful as matrix metalloproteinase inhibitors

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003534239A (en) 1999-12-17 2003-11-18 ヴァージコア・インコーポレーテッド Novel succinate compounds, compositions, and methods of use and preparation
PL365694A1 (en) * 2000-06-28 2005-01-10 Teva Pharmaceutical Industries Ltd. Carvedilol
DK1406893T3 (en) 2001-06-15 2007-08-06 Vicuron Pharm Inc Bicyclic pyrrolidine compounds
EP1660471B1 (en) 2003-08-23 2011-05-04 Vernalis (R&D) Limited Derivatives of hydroxamic acid as metalloproteinase inhibitors
PE20060426A1 (en) * 2004-06-02 2006-06-28 Schering Corp TARTARIC ACID DERIVATIVES AS INHIBITORS OF MMPs, ADAMs, TACE AND TNF-alpha
US20070167426A1 (en) 2004-06-02 2007-07-19 Schering Corporation Compounds for the treatment of inflammatory disorders and microbial diseases
US7638513B2 (en) * 2004-06-02 2009-12-29 Schering Corporation Compounds for the treatment of inflammatory disorders
CA2632921A1 (en) 2005-12-01 2007-06-07 Schering Corporation Compounds for the treatment of inflammatory disorders and microbial diseases

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR1484601A (en) * 1955-01-22 1967-06-16 Leinfelder Hermann Process for preparing polyelectrolytes
US5618842A (en) * 1991-12-31 1997-04-08 Lifegroup S.P.A. N-acyl derivatives of aminoalcohols with polycarboxylic acids able to modulate mast cells in inflammatory processes having neuroimmunogenic origin
US6534491B2 (en) * 1996-10-16 2003-03-18 American Cyanamid Company Preparation and use of ortho-sulfonamido bicyclic heteroaryl hydroxamic acids as matrix metalloproteinase and tace inhibitors
US6344457B1 (en) * 1997-04-22 2002-02-05 Chiesi Farmaceutici S.P.A. Amino acid derivatives inhibiting extracellular matrix metalloproteinase and TNF alpha release
US6677355B1 (en) * 1999-08-18 2004-01-13 Warner-Lambert Company Hydroxamic acid compounds useful as matrix metalloproteinase inhibitors
WO2001021661A1 (en) * 1999-09-20 2001-03-29 Novo Nordisk A/S BIVALENT INHIBITOR OF FVIIa/TF/FXa COMPLEX
US6495565B2 (en) * 2000-03-17 2002-12-17 Bristol-Myers Squibb Pharma Company β-amino acid derivatives as inhibitors of matrix metalloproteases and TNF-α
WO2001077075A2 (en) * 2000-04-06 2001-10-18 Inotek Corporation Inhibitors of inflammation and reperfusion injury
WO2003076461A2 (en) * 2002-03-12 2003-09-18 Novo Nordisk A/S Dimeric tf antagonist comprising two factor vii polypeptides

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
ALLENMARK S ET AL: "Chiral selectors based on C2-symmetric dicarboxylic acids" TETRAHEDRON: ASYMMETRY, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 11, no. 17, 8 September 2000 (2000-09-08), pages 3527-3534, XP004224132 ISSN: 0957-4166 *
C. GIORDANO ET AL: "First Asymmetric Synthesis of Enantiomerically Pure (1R,2S)-(-)-(1,2-Epoxypropyl)phosphonic Acid (Fosfomycin)" JOURNAL OF ORGANIC CHEMISTRY, vol. 54, no. 6, 1989, pages 1470-1473, XP002363363 *
F. MASSICOT ET AL: "Solvent-Free Synthesis of Tartramides Under Microwave Activation" SYNTHESIS, no. 16, 2001, pages 2441-2444, XP002363367 *
GAWRONSKI J ET AL: "Factors Affecting Conformation of (R,R)-Tartaric Acid Ester, Amide and Nitrile Derivatives. X-Ray Diffraction, Circular Dichroism, Nuclear Magnetic Resonance and Ab Initio Studies" TETRAHEDRON, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 53, no. 17, April 1997 (1997-04), pages 6113-6144, XP004105837 ISSN: 0040-4020 *
J. D. D'IANNI ET AL: "Hydrogenation of Hydroxyamides" JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 61, 1939, pages 1675-1681, XP002363369 *
J. R. COGGINS ET AL: "Use of Dimethyl Suberimidate and Novel Perodate-Cleavable Bis(imido esters) to Study the Quaternary Structure of the Pyruvate Dehydrogenase Multienzyme Complex of Escherichia coli" BIOCHEMISTRY, vol. 15, no. 12, 1976, pages 2527-2533, XP002363368 *
K. REHSE ET AL: "Antiaggregatorische und anticoagulante Eigenschaften von Oligoaminen, 6. Mitt.: Oligohydroxyalkandiamine" ARCHIV DER PHARMAZIE, vol. 320, 1987, pages 1155-1161, XP008058598 *
KOENIG S ET AL: "SYNTHESE VON N-TERT-ALKYLGLYOXYSAEUREAMIDEN" SYNTHESIS, GEORG THIEME VERLAG, STUTTGART, DE, December 1993 (1993-12), pages 1233-1234, XP001156323 ISSN: 0039-7881 *
L. D.-L. LU ET AL: "Two New Asymmetric Epoxidation Catalysts. Unusual Stoichiometry and Inverse Enantiofacial Selection" JOURNAL OF ORGANIC CHEMISTRY, vol. 49, no. 4, 1984, pages 728-731, XP002363364 *
PING XU ET AL: "Synthesis of a Peptidomimetic HCMV Protease Inhibitor Library" SYNTHESIS, no. 8, 2002, pages 1017-1026, XP002363366 *
RUO WANG ET AL: "A Search for Pyrophosphate Mimics for the Development of Substrates and Inhibitors of Glycosyltransferases" BIOORGANIC & MEDICINAL CHEMISTRY, vol. 5, no. 4, 1997, pages 661-672, XP002363365 *

Cited By (146)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7638513B2 (en) 2004-06-02 2009-12-29 Schering Corporation Compounds for the treatment of inflammatory disorders
US8835423B2 (en) 2005-09-22 2014-09-16 Incyte Corporation Azepine inhibitors of janus kinases
US8563541B2 (en) 2005-09-22 2013-10-22 Incyte Corporation Azepine inhibitors of Janus kinases
WO2007064732A1 (en) * 2005-12-01 2007-06-07 Schering Corporation Compounds for the treatment of inflammatory disorders and microbial diseases
US7553864B2 (en) 2005-12-01 2009-06-30 Schering Corporation Compounds for the treatment of inflammatory disorders and microbial diseases
WO2007064749A1 (en) 2005-12-01 2007-06-07 Schering Corporation Compounds for the treatment of inflammatory disorders and microbial diseases
US9974790B2 (en) 2005-12-13 2018-05-22 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as janus kinase inhibitors
US11331320B2 (en) 2005-12-13 2022-05-17 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US10398699B2 (en) 2005-12-13 2019-09-03 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as janus kinase inhibitors
US9814722B2 (en) 2005-12-13 2017-11-14 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as janus kinase inhibitors
US10639310B2 (en) 2005-12-13 2020-05-05 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US9662335B2 (en) 2005-12-13 2017-05-30 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as janus kinase inhibitors
US11744832B2 (en) 2005-12-13 2023-09-05 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US7598257B2 (en) 2005-12-13 2009-10-06 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as janus kinase inhibitors
US9079912B2 (en) 2005-12-13 2015-07-14 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as Janus kinase inhibitors
US8513270B2 (en) 2006-12-22 2013-08-20 Incyte Corporation Substituted heterocycles as Janus kinase inhibitors
US8841318B2 (en) 2006-12-22 2014-09-23 Incyte Corporation Substituted heterocycles as janus kinase inhibitors
EP2116526B1 (en) * 2006-12-29 2018-04-18 Institute of Pharmacology and Toxicology Academy of Military Medical Sciences P.L.A. China Derivates of substituted tartaric aicd and usage for preparating beta secretase inhibitors
US8822481B1 (en) 2007-06-13 2014-09-02 Incyte Corporation Salts of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d] pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US9376439B2 (en) 2007-06-13 2016-06-28 Incyte Corporation Salts of the janus kinase inhibitor (R)-3(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US7834022B2 (en) 2007-06-13 2010-11-16 Incyte Corporation Metabolites of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US8722693B2 (en) 2007-06-13 2014-05-13 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US8889697B2 (en) 2007-06-13 2014-11-18 Incyte Corporation Metabolites of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US10016429B2 (en) 2007-06-13 2018-07-10 Incyte Corporation Salts of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US10610530B2 (en) 2007-06-13 2020-04-07 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US11213528B2 (en) 2007-06-13 2022-01-04 Incyte Holdings Corporation Salts of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US10463667B2 (en) 2007-06-13 2019-11-05 Incyte Incorporation Metabolites of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US8829013B1 (en) 2007-06-13 2014-09-09 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US8158616B2 (en) 2008-03-11 2012-04-17 Incyte Corporation Azetidine and cyclobutane derivatives as JAK inhibitors
US8420629B2 (en) 2008-03-11 2013-04-16 Incyte Corporation Azetidine and cyclobutane derivatives as JAK inhibitors
EP2351743A1 (en) * 2008-10-27 2011-08-03 Takeda Pharmaceutical Company Limited Bicyclic compound
EP2351743A4 (en) * 2008-10-27 2012-05-09 Takeda Pharmaceutical Bicyclic compound
US8501804B2 (en) 2008-10-27 2013-08-06 Takeda Pharmaceutical Company Limited Bicyclic compound
US8987242B2 (en) 2008-12-05 2015-03-24 Merck Sharp & Dohme Corp. Morpholinone compounds as factor IXA inhibitors
US9623029B2 (en) 2009-05-22 2017-04-18 Incyte Holdings Corporation 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]octane- or heptane-nitrile as JAK inhibitors
EP2272846A1 (en) 2009-06-23 2011-01-12 Bayer CropScience AG Thiazolylpiperidine derivatives as fungicide
US9512161B2 (en) 2009-10-09 2016-12-06 Incyte Corporation Hydroxyl, keto, and glucuronide derivatives of 3-(4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US9187496B2 (en) 2009-12-18 2015-11-17 Idenix Pharmaceuticals Llc 5,5-fused arylene or heteroarylene hepatitis C virus inhibitors
US9999619B2 (en) 2010-03-10 2018-06-19 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US10695337B2 (en) 2010-03-10 2020-06-30 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US11285140B2 (en) 2010-03-10 2022-03-29 Incyte Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US10758543B2 (en) 2010-05-21 2020-09-01 Incyte Corporation Topical formulation for a JAK inhibitor
US11571425B2 (en) 2010-05-21 2023-02-07 Incyte Corporation Topical formulation for a JAK inhibitor
US10869870B2 (en) 2010-05-21 2020-12-22 Incyte Corporation Topical formulation for a JAK inhibitor
US11590136B2 (en) 2010-05-21 2023-02-28 Incyte Corporation Topical formulation for a JAK inhibitor
US11219624B2 (en) 2010-05-21 2022-01-11 Incyte Holdings Corporation Topical formulation for a JAK inhibitor
US10640506B2 (en) 2010-11-19 2020-05-05 Incyte Holdings Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidines derivatives as JAK inhibitors
US10813930B2 (en) 2010-12-22 2020-10-27 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US10213427B2 (en) 2010-12-22 2019-02-26 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US9993480B2 (en) 2011-02-18 2018-06-12 Novartis Pharma Ag mTOR/JAK inhibitor combination therapy
WO2012118850A1 (en) 2011-02-28 2012-09-07 Array Biopharma Inc. Serine/threonine kinase inhibitors
WO2012135581A1 (en) * 2011-03-31 2012-10-04 Idenix Pharmaceuticals, Inc. Methods for treating drug-resistant hepatitis c virus infection with a 5,5-fused arylene or heteroarylene hepatitis c virus inhibitor
US11214573B2 (en) 2011-06-20 2022-01-04 Incyte Holdings Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US9611269B2 (en) 2011-06-20 2017-04-04 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US10513522B2 (en) 2011-06-20 2019-12-24 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US9358229B2 (en) 2011-08-10 2016-06-07 Novartis Pharma Ag JAK PI3K/mTOR combination therapy
US9718834B2 (en) 2011-09-07 2017-08-01 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US11266698B2 (en) 2011-10-07 2022-03-08 4D Pharma Research Limited Bacterium for use as a probiotic for nutritional and medical applications
US10004730B2 (en) 2011-10-12 2018-06-26 University of Pittsburgh—of the Commonwealth System of Higher Education Small molecules targeting androgen receptor nuclear localization and/or level in prostate cancer
US11840534B2 (en) 2012-06-13 2023-12-12 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US9611267B2 (en) 2012-06-13 2017-04-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US11053246B2 (en) 2012-06-13 2021-07-06 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US10131667B2 (en) 2012-06-13 2018-11-20 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US9745311B2 (en) 2012-08-10 2017-08-29 Incyte Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
WO2014047020A1 (en) 2012-09-19 2014-03-27 Novartis Ag Dihydropyrrolidino-pyrimidines as kinase inhibitors
US10874616B2 (en) 2012-11-15 2020-12-29 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US11337927B2 (en) 2012-11-15 2022-05-24 Incyte Holdings Corporation Sustained-release dosage forms of ruxolitinib
US10166191B2 (en) 2012-11-15 2019-01-01 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US11576864B2 (en) 2012-11-15 2023-02-14 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US11896717B2 (en) 2012-11-15 2024-02-13 Incyte Holdings Corporation Sustained-release dosage forms of ruxolitinib
US11576865B2 (en) 2012-11-15 2023-02-14 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US9714233B2 (en) 2013-03-06 2017-07-25 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US10851137B2 (en) 2013-04-10 2020-12-01 4D Pharma Research Limited Polypeptide and immune modulation
US11414463B2 (en) 2013-04-10 2022-08-16 4D Pharma Research Limited Polypeptide and immune modulation
US10040790B2 (en) 2013-04-19 2018-08-07 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10947230B2 (en) 2013-04-19 2021-03-16 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US10450313B2 (en) 2013-04-19 2019-10-22 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US11530214B2 (en) 2013-04-19 2022-12-20 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US9655854B2 (en) 2013-08-07 2017-05-23 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US11045421B2 (en) 2013-08-07 2021-06-29 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US10561616B2 (en) 2013-08-07 2020-02-18 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US10882834B2 (en) 2013-09-20 2021-01-05 University of Pittsburgh—of the Commonwealth System of Higher Education Compounds for treating prostate cancer
EP3046561A4 (en) * 2013-09-20 2017-07-26 University of Pittsburgh - Of the Commonwealth System of Higher Education Compounds for treating prostate cancer
US10544110B2 (en) 2013-09-20 2020-01-28 University of Pittsburgh—of the Commonwealth System of Higher Education Small molecule inhibitors of the nuclear translocation of androgen receptor for the treatment of castration-resistant prostate cancer
WO2015042297A1 (en) 2013-09-20 2015-03-26 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Compounds for treating prostate can cancer
WO2015172196A1 (en) * 2014-05-13 2015-11-19 Monash University Heterocyclic compounds and use of same
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11723933B2 (en) 2014-12-23 2023-08-15 Cj Bioscience, Inc. Composition of bacteroides thetaiotaomicron for immune modulation
US10973872B2 (en) 2014-12-23 2021-04-13 4D Pharma Research Limited Pirin polypeptide and immune modulation
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10251892B2 (en) 2015-02-20 2019-04-09 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10632126B2 (en) 2015-02-20 2020-04-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10214528B2 (en) 2015-02-20 2019-02-26 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9801889B2 (en) 2015-02-20 2017-10-31 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11173162B2 (en) 2015-02-20 2021-11-16 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11667635B2 (en) 2015-02-20 2023-06-06 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10016438B2 (en) 2015-02-20 2018-07-10 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11014923B2 (en) 2015-02-20 2021-05-25 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10738048B2 (en) 2015-02-20 2020-08-11 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10744167B2 (en) 2015-06-15 2020-08-18 4D Pharma Research Limited Compositions comprising bacterial strains
US10493112B2 (en) 2015-06-15 2019-12-03 4D Pharma Research Limited Compositions comprising bacterial strains
US11040075B2 (en) 2015-06-15 2021-06-22 4D Pharma Research Limited Compositions comprising bacterial strains
US11389493B2 (en) 2015-06-15 2022-07-19 4D Pharma Research Limited Compositions comprising bacterial strains
US11433106B2 (en) 2015-06-15 2022-09-06 4D Pharma Research Limited Compositions comprising bacterial strains
US11331352B2 (en) 2015-06-15 2022-05-17 4D Pharma Research Limited Compositions comprising bacterial strains
US10736926B2 (en) 2015-06-15 2020-08-11 4D Pharma Research Limited Compositions comprising bacterial strains
US10780134B2 (en) 2015-06-15 2020-09-22 4D Pharma Research Limited Compositions comprising bacterial strains
US10864236B2 (en) 2015-06-15 2020-12-15 4D Pharma Research Limited Compositions comprising bacterial strains
US11273185B2 (en) 2015-06-15 2022-03-15 4D Pharma Research Limited Compositions comprising bacterial strains
US10500237B2 (en) 2015-06-15 2019-12-10 4D Pharma Research Limited Compositions comprising bacterial strains
US10471108B2 (en) 2015-11-20 2019-11-12 4D Pharma Research Limited Compositions comprising bacterial strains
US11058732B2 (en) 2015-11-20 2021-07-13 4D Pharma Research Limited Compositions comprising bacterial strains
US10610550B2 (en) 2015-11-20 2020-04-07 4D Pharma Research Limited Compositions comprising bacterial strains
US10583158B2 (en) 2016-03-04 2020-03-10 4D Pharma Plc Compositions comprising bacterial strains
US11766433B2 (en) 2016-03-24 2023-09-26 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Small molecule inhibitors of the nuclear translocation of androgen receptor for the treatment of castration-resistant prostate cancer
US10980806B2 (en) 2016-03-24 2021-04-20 University of Pittsburgh—of the Commonwealth System of Higher Education Small molecule inhibitors of the nuclear translocation of androgen receptor for the treatment of castration-resistant prostate cancer
US10610548B2 (en) 2016-07-13 2020-04-07 4D Pharma Plc Compositions comprising bacterial strains
US10610549B2 (en) 2016-07-13 2020-04-07 4D Pharma Plc Composition comprising bacterial strains
US10960031B2 (en) 2016-07-13 2021-03-30 4D Pharma Plc Compositions comprising bacterial strains
US11224620B2 (en) 2016-07-13 2022-01-18 4D Pharma Plc Compositions comprising bacterial strains
US10967010B2 (en) 2016-07-13 2021-04-06 4D Pharma Plc Compositions comprising bacterial strains
US11084818B2 (en) 2016-11-11 2021-08-10 Hepo Pharmaceutical Co., Ltd. Nitrogen-containing heterocyclic compound, preparation method, intermediate, pharmaceutical composition and use
US10485830B2 (en) 2016-12-12 2019-11-26 4D Pharma Plc Compositions comprising bacterial strains
US11376284B2 (en) 2017-05-22 2022-07-05 4D Pharma Research Limited Compositions comprising bacterial strains
US11382936B2 (en) 2017-05-22 2022-07-12 4D Pharma Research Limited Compositions comprising bacterial strains
US11123378B2 (en) 2017-05-22 2021-09-21 4D Pharma Research Limited Compositions comprising bacterial strains
US10987387B2 (en) 2017-05-24 2021-04-27 4D Pharma Research Limited Compositions comprising bacterial strain
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US11472801B2 (en) 2017-05-26 2022-10-18 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US11660319B2 (en) 2017-06-14 2023-05-30 4D Pharma Research Limited Compositions comprising bacterial strains
US11007233B2 (en) 2017-06-14 2021-05-18 4D Pharma Research Limited Compositions comprising a bacterial strain of the genus Megasphera and uses thereof
US11779613B2 (en) 2017-06-14 2023-10-10 Cj Bioscience, Inc. Compositions comprising a bacterial strain of the genus Megasphera and uses thereof
US11123379B2 (en) 2017-06-14 2021-09-21 4D Pharma Research Limited Compositions comprising bacterial strains
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US11278541B2 (en) 2017-12-08 2022-03-22 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US10899736B2 (en) 2018-01-30 2021-01-26 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US11304949B2 (en) 2018-03-30 2022-04-19 Incyte Corporation Treatment of hidradenitis suppurativa using JAK inhibitors
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms
US11939331B2 (en) 2021-06-09 2024-03-26 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors

Also Published As

Publication number Publication date
AU2005252201A1 (en) 2005-12-22
TW200612911A (en) 2006-05-01
KR20070103671A (en) 2007-10-24
CA2569111A1 (en) 2005-12-22
JP2008501691A (en) 2008-01-24
US8039467B2 (en) 2011-10-18
PE20060426A1 (en) 2006-06-28
ZA200610055B (en) 2009-05-27
US20060252778A1 (en) 2006-11-09
IL179674A0 (en) 2007-05-15
AR051172A1 (en) 2006-12-27
US7652020B2 (en) 2010-01-26
CN101027295A (en) 2007-08-29
US20100145048A1 (en) 2010-06-10
WO2005121130A3 (en) 2006-07-20
MXPA06014054A (en) 2007-01-31
EP1773821A2 (en) 2007-04-18

Similar Documents

Publication Publication Date Title
US7652020B2 (en) Compounds for the treatment of inflammatory disorders
US7638513B2 (en) Compounds for the treatment of inflammatory disorders
US20070167426A1 (en) Compounds for the treatment of inflammatory disorders and microbial diseases
EP1778676B1 (en) Hydantoin derivatives for the treatment of inflammatory disorders
US8673911B2 (en) Inhibitors of histone deacetylase
EP1973900B1 (en) Hydantoin compounds for the treatment of inflammatory disorders
AU2006320604A1 (en) Compounds for the treatment of inflammatory disorders and microbial diseases
WO2003042174A1 (en) Cannabinoid receptor ligands
JP2008505963A (en) Amide derivatives as inhibitors of histone deacetylase (HDAC)
US7034057B2 (en) Compounds for the treatment of inflammatory disorders
KR101386679B1 (en) Novel arylpiperazine-containing imidazole 4-carboxamide derivatives and pharmaceutical composition comprising same
JP2009509931A (en) Amino acid derivatives as histone deacetylase (HDAC) inhibitors
JP2011522801A (en) Alpha-substituted N-sulfonylglycine (GYLCINE) amide antagonists of CCR10, compositions containing them and methods for using them
JP2014517812A (en) Sulfamide derivative having adamantyl group and pharmaceutically acceptable salt thereof
MX2008007092A (en) Compounds for the treatment of inflammatory disorders and microbial diseases
AU2002346366A1 (en) Cannabinoid receptor ligands

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 06117761

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 2005252201

Country of ref document: AU

Ref document number: 551478

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 179674

Country of ref document: IL

Ref document number: 2569111

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 200610055

Country of ref document: ZA

Ref document number: 12006502413

Country of ref document: PH

Ref document number: PA/a/2006/014054

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2007515501

Country of ref document: JP

Ref document number: 4431/CHENP/2006

Country of ref document: IN

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

WWE Wipo information: entry into national phase

Ref document number: 1020067026812

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2005759261

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2005252201

Country of ref document: AU

Date of ref document: 20050601

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005252201

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 200580026189.0

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2005759261

Country of ref document: EP