WO2014125444A1 - Heterocyclic amides as kinase inhibitors - Google Patents

Heterocyclic amides as kinase inhibitors Download PDF

Info

Publication number
WO2014125444A1
WO2014125444A1 PCT/IB2014/059004 IB2014059004W WO2014125444A1 WO 2014125444 A1 WO2014125444 A1 WO 2014125444A1 IB 2014059004 W IB2014059004 W IB 2014059004W WO 2014125444 A1 WO2014125444 A1 WO 2014125444A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
mmol
alkyl
methyl
salt according
Prior art date
Application number
PCT/IB2014/059004
Other languages
French (fr)
Inventor
Deepak BANDYOPADHYAY
Patrick M. Eidam
Peter J. GOUGH
Philip Anthony Harris
Jae U. Jeong
Jianxing Kang
Bryan Wayne KING
Ami Lakdawala Shah
Jr. Robert W. Marquis
Lara Kathryn LEISTER
Attiq Rahman
Joshi M. Ramanjulu
Clark A Sehon
JR. Robert SINGHAUS
Daohua Zhang
Original Assignee
Glaxosmithkline Intellectual Property Development Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Glaxosmithkline Intellectual Property Development Limited filed Critical Glaxosmithkline Intellectual Property Development Limited
Priority to PL14705891T priority Critical patent/PL2956452T3/en
Priority to CN201480021120.8A priority patent/CN105121432B/en
Priority to RS20180698A priority patent/RS57332B1/en
Priority to BR122017002949A priority patent/BR122017002949A2/en
Priority to CA2900695A priority patent/CA2900695C/en
Priority to UAA201508769A priority patent/UA118259C2/en
Priority to BR112015019627-6A priority patent/BR112015019627B1/en
Priority to EP19154374.3A priority patent/EP3508484B1/en
Priority to SI201430738T priority patent/SI2956452T1/en
Priority to US14/763,183 priority patent/US9556152B2/en
Priority to MA44820A priority patent/MA44820B1/en
Priority to KR1020157024819A priority patent/KR102228764B1/en
Priority to BR122017002946A priority patent/BR122017002946A2/en
Priority to EA201591506A priority patent/EA028991B1/en
Priority to NZ628447A priority patent/NZ628447A/en
Priority to CR20190511A priority patent/CR20190511A/en
Priority to CR20190512A priority patent/CR20190512A/en
Priority to EP14705891.1A priority patent/EP2956452B1/en
Priority to MA38326A priority patent/MA38326B1/en
Priority to PL18155836T priority patent/PL3342771T3/en
Priority to BR122017002951A priority patent/BR122017002951A2/en
Priority to DK14705891.1T priority patent/DK2956452T3/en
Priority to ES14705891.1T priority patent/ES2672530T3/en
Priority to JP2015557554A priority patent/JP6321045B2/en
Priority to AU2014217453A priority patent/AU2014217453B2/en
Priority to KR1020197004425A priority patent/KR102267977B1/en
Priority to LTEP14705891.1T priority patent/LT2956452T/en
Priority to SG11201505796TA priority patent/SG11201505796TA/en
Priority to EP18152185.7A priority patent/EP3375780B1/en
Priority to MX2015010572A priority patent/MX360715B/en
Priority to EP18155836.2A priority patent/EP3342771B1/en
Publication of WO2014125444A1 publication Critical patent/WO2014125444A1/en
Priority to ZA2015/05267A priority patent/ZA201505267B/en
Priority to PH12015501675A priority patent/PH12015501675A1/en
Priority to IL240280A priority patent/IL240280B/en
Priority to CR20150420A priority patent/CR20150420A/en
Priority to HK16101405.9A priority patent/HK1213545A1/en
Priority to US15/271,581 priority patent/US9624202B2/en
Priority to US15/459,141 priority patent/US10292987B2/en
Priority to US15/835,747 priority patent/US20180098998A1/en
Priority to IL256500A priority patent/IL256500B/en
Priority to IL256502A priority patent/IL256502B/en
Priority to PH12018500342A priority patent/PH12018500342A1/en
Priority to PH12018500341A priority patent/PH12018500341A1/en
Priority to PH12018500343A priority patent/PH12018500343A1/en
Priority to HRP20180900TT priority patent/HRP20180900T1/en
Priority to CY20181100604T priority patent/CY1120320T1/en
Priority to US16/299,254 priority patent/US10933070B2/en
Priority to US16/408,718 priority patent/US10940154B2/en
Priority to CY20211100121T priority patent/CY1123915T1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • 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
    • 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
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • 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
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • 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
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • A61P29/02Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID] without antiinflammatory effect
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • 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
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • 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
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D223/00Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom
    • C07D223/02Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings
    • C07D223/06Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings 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
    • C07D223/08Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D267/00Heterocyclic compounds containing rings of more than six members having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D267/02Seven-membered rings
    • C07D267/08Seven-membered rings having the hetero atoms in positions 1 and 4
    • C07D267/12Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
    • C07D267/14Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D281/00Heterocyclic compounds containing rings of more than six members having one nitrogen atom and one sulfur atom as the only ring hetero atoms
    • C07D281/02Seven-membered rings
    • C07D281/04Seven-membered rings having the hetero atoms in positions 1 and 4
    • C07D281/08Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
    • C07D281/10Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • 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
    • 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
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic 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
    • 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
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems

Definitions

  • the present invention relates to heterocyclic amides that inhibit RIP1 kinase and methods of making and using the same.
  • Receptor-interacting protein- 1 (RIP1) kinase is a TKL family serine/threonine protein kinase involved in innate immune signaling.
  • RIPl kinase is a RHIM domain containing protein, with an N-terminal kinase domain and a C-terminal death domain ((2005) Trends Biochem. Sci. 30, 151-159).
  • the death domain of RIPl mediates interaction with other death domain containing proteins including Fas and TNFR- 1 ((1995) Cell 81 513-523), TRAIL-Rl and TRAIL-R2 ((1997) Immunity 7, 821-830) and TRADD ((1996) Immunity 4, 387-396), while the RHIM domain is crucial for binding other RHFM domain containing proteins such as TRIF ((2004) Nat Immunol. 5, 503-507), DAI ((2009) EMBO Rep. 10, 916-922) and RIP3 ((1999) J. Biol. Chem. 274, 16871- 16875); (1999) Curr. Biol. 9, 539-542) and exerts many of its effects through these interactions.
  • RIPl is a central regulator of cell signaling, and is involved in mediating both pro-survival and programmed cell death pathways which will be discussed below.
  • TLR3 (2004) Nat Immunol. 5, 503-507), TLR4 ((2005) J. Biol. Chem. 280,
  • complex I provides a platform for pro-survival signaling through the activation of the NFKB and MAP kinases pathways ((2010) Sci. Signal. 115, re4).
  • RIPl by proteins such as A20 and CYLD or inhibition of the cIAPs
  • DISC death-inducing signaling complex
  • formation of the DISC which contains RIPl, TRADD, FADD and caspase 8, results in the activation of caspase 8 and the onset of programmed apoptotic cell death also in a RIPl kinase independent fashion ((2012) FEBS J 278, 877-887).
  • Apoptosis is largely a quiescent form of cell death, and is involved in routine processes such as development and cellular homeostasis.
  • RIP3 can now enter this complex, become phosphorylated by RIPl and initiate a caspase-independent programmed necrotic cell death through the activation of MLKL and PGAM5 ((2012) Cell 148, 213-227); ((2012) Cell 148, 228-243); ((2012) Proc. Natl. Acad. Sci. USA. 109, 5322-5327).
  • DAMPs danger associated molecular patterns
  • DAMPs are capable of providing a "danger signal” to surrounding cells and tissues, eliciting proinflammatory responses including inflammasome activation, cytokine production and cellular recruitment ((2008 Nat. Rev. Immunol 8, 279-289).
  • Dysregulation of RIPl kinase-mediated programmed cell death has been linked to various inflammatory diseases, as demonstrated by use of the RIP3 knockout mouse (where RIPl -mediated programmed necrosis is completely blocked) and by Necrostatin-1 (a tool inhibitor of RIPl kinase activity with poor oral bioavailability).
  • the RIP3 knockout mouse has been shown to be protective in inflammatory bowel disease (including Ulcerative colitis and Crohn's disease) ((2011) Nature 477, 330-334), Psoriasis ((2011) Immunity 35, 572- 582), retinal-detachment-induced photoreceptor necrosis ((2010) PNAS 107, 21695- 21700), retinitis pigmentosa ((2012) Proc. Natl. Acad. Sci., 109:36, 14598-14603), cerulein-induced acute pancreatits ((2009) Cell 137, 1100-1111) and Sepsis/systemic inflammatory response syndrome (SIRS) ((2011) Immunity 35, 908-918).
  • inflammatory bowel disease including Ulcerative colitis and Crohn's disease
  • Psoriasis (2011) Immunity 35, 572- 582)
  • retinal-detachment-induced photoreceptor necrosis (2010) PNAS 107, 21695
  • Necrostatin-1 has been shown to be effective in alleviating ischemic brain injury ((2005) Nat. Chem. Biol. 1, 112-119), retinal ischemia/reperfusion injury ((2010) J. Neurosci. Res. 88, 1569- 1576), Huntington's disease ((2011) Cell Death Dis. 2 el 15), renal ischemia reperfusion injury ((2012) Kidney Int. 81, 751-761), cisplatin induced kidney injury ((2012) Ren. Fail. 34, 373-377) and traumatic brain injury ((2012) Neurochem. Res. 37, 1849-1858).
  • Other diseases or disorders regulated at least in part by RIPl -dependent apoptosis, necrosis or cytokine production include hematological and solid organ malignancies ((2013) Genes Dev. 27: 1640-1649), bacterial infections and viral infections ((2014) Cell Host & Microbe 15, 23-35) (including, but not limited to, tuberculosis and influenza ((2013) Cell 153, 1- 14)) and Lysosomal storage diseases (particularly, Gaucher Disease, Nature Medicine Advance Online Publication, 19 January 2014, doi: 10.1038/nm.3449).
  • a potent, selective, small molecule inhibitor of RIP1 kinase activity would block RIP 1 -dependent cellular necrosis and thereby provide a therapeutic benefit in diseases or events associated with DAMPs, cell death, and/or inflammation.
  • the invention is directed to compounds according to Formula (I):
  • X is O, S, SO, S0 2 , NH, CO, CH 2 , CF 2 , CH(CH 3 ), CH(OH), or N(CH 3 );
  • Y is CH 2 or CH 2 CH 2;
  • Z 1 is N, CH or CR 1 ;
  • Z 2 is CH or CR 2 ;
  • Z 3 is N, CH or CR 3 ;
  • Z 4 is CH or CR 4 ;
  • R 1 is fluoro or methyl
  • R 2 and R 3 is halogen, cyano, (Ci-C 6 )alkyl, halo(Ci-C4)alkyl,
  • R 2 and R 3 is halogen or (Ci-C 6 )alkyl
  • R 4 is fluoro, chloro, or methyl
  • R 5 is H or methyl
  • A is phenyl, 5-6 membered heteroaryl, or 5-6 membered
  • heterocycloalkyl wherein the carbonyl moiety and L are substituted 1,3 on ring
  • n 0 or m is 1 and R A is (Ci-C4)alkyl
  • L is O, S, NH, N(CH 3 ), CH 2 , CH 2 CH 2 , CH(CH 3 ), CHF, CF 2 , CH 2 0,
  • B is an optionally substituted (C 3 -C 6 )cycloalkyl, phenyl, 5-6 membered
  • heteroaryl or 5-6 membered heterocycloalkyl
  • the compounds according to Formula (I), or salts, particularly pharmaceutically acceptable salts, thereof, are inhibitors of RIPl kinase.
  • the present invention is also directed to a method of inhibiting RIPl kinase which method comprises contacting a cell with a compound according to
  • the invention is further directed to a method of treating a RIPl kinase-mediated disease or disorder which comprises administering a therapeutically effective amount of a compound according to Formula (I), or a salt, particularly a pharmaceutically acceptable salt thereof, to a patient (a human or other mammal, particularly, a human) in need thereof.
  • Such RIP1 kinase-mediated diseases or disorders include inflammatory bowel disease (including Crohn's disease and ulcerative colitis), psoriasis, retinal detachment, retinitis pigmentosa, arthritis (including rheumatoid arthritis, spondyloarthritis, gout, and SoJIA), transplant rejection, ischemia reperfusion injury of solid organs, multiple sclerosis, and tumor necrosis factor receptor-associated periodic syndrome.
  • the present invention is further directed to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound according to Formula (I), or a salt, particularly a pharmaceutically acceptable salt, thereof and a pharmaceutically acceptable excipient.
  • this invention is directed to a pharmaceutical composition for the treatment of a RIP1 kinase-mediated disease or disorder, where the composition comprises a compound according to Formula (I), or a salt, particularly a pharmaceutically acceptable salt, thereof and a pharmaceutically acceptable excipient.
  • Figure 1 A shows the temperature loss over time in mice after oral pre-dosing with the compound of Example 12 or vehicle followed by simultaneous i.v. administration of mouse TNF and zVAD.
  • Figure IB shows the temperature loss in mice 2.5 hours after oral pre-dosing with the compound of Example 12 or vehicle followed by simultaneous i.v. administration of mouse TNF and zVAD.
  • Figure 2A shows the temperature loss over time in mice after oral pre-dosing with the compound of Example 12 or vehicle followed by i.v. administration of mouse TNF.
  • Figure 2B shows the temperature loss in mice 6 hours after oral pre-dosing with the compound of Example 12 or vehicle followed by i.v. administration of mouse TNF.
  • Figure 3 A shows the cellular levels of ATP in mouse L929 fibrosarcoma cells pre- treated with the compound of Example 77 followed by treatment with TNFa + QvD.
  • Figure 3B shows the cellular levels of ATP in human monocytic leukemia U937 fibrosarcoma cells pre-treated with the compound of Example 77 followed by treatment with TNFa + QvD.
  • Figure 4 A shows the temperature loss over time in mice after oral pre-dosing with the compound of Example 161 or vehicle followed by simultaneous i.v. administration of mouse TNF and zVAD.
  • Figure 4B shows the temperature loss in mice 6 hours after oral pre-dosing with the compound of Example 161 or vehicle followed by simultaneous i.v. administration of mouse TNF and zVAD.
  • Figure 5 A shows the temperature loss over time in mice after oral pre-dosing with the compound of Example 161 or vehicle followed by i.v. administration of mouse TNF.
  • Figure 5B shows the temperature loss in mice 6 hours after oral pre-dosing with the compound of Example 161 or vehicle followed by i.v. administration of mouse TNF.
  • Figure 6A shows the cellular levels of ATP in mouse L929 fibrosarcoma cells pre- treated with the compound of Example 161 followed by treatment with TNFa + QvD.
  • Figure 6B shows an IC 50 curve of ATP in human monocytic leukemia U937 fibrosarcoma cells pre-treated with the compound of Example 161 followed by treatment with TNFa + QvD. Data are normalized to lOmM Nec-1 which was set at 100% survival.
  • Figure 7 is a powder x-ray diffraction (PXRD) pattern of a crystalline form of anhydrous (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)- 4H-l,2,4-triazole-3-carboxamide (free base).
  • PXRD powder x-ray diffraction
  • This invention is also directed to a compound of Formula (I) wherein:
  • X is O, S, SO, S0 2 , NH, CO, CH 2 , CF 2 , CH(CH 3 ), CH(OH), or N(CH 3 );
  • Y is CH 2 or CH 2 CH 2;
  • Z 1 is N, CH or CR 1 ;
  • Z 2 is CH or CR 2 ;
  • Z 3 is N, CH or CR 3 ;
  • Z 4 is CH or CR 4 ;
  • R 1 is fluoro or methyl
  • R 2 and R 3 is halogen, cyano, (Ci-C 6 )alkyl, halo(Ci-C 4 )alkyl,
  • 5-6 membered heterocycloalkyl and 5-6 membered heteroaryl are optionally substituted by 1 or 2 substituents each independently selected from the group consisting of (Ci-C 4 )alkyl and -(Ci-C 4 )alkyl-CN;
  • R 2 and R 3 is halogen or (Ci-C 6 )alkyl
  • R 4 is fluoro, chloro, or methyl
  • R 5 is H or methyl
  • A is phenyl, 5-6 membered heteroaryl, or 5-6 membered
  • heterocycloalkyl wherein the carbonyl moiety and L are substituted 1,3 on ring
  • n 0 or m is 1 and R A is (Ci-C 4 )alkyl
  • L is O, S, NH, N(CH 3 ), CH 2 , CH 2 CH 2 , CH(CH 3 ), CHF, CF 2 , CH 2 0,
  • B is an optionally substituted (C 3 -C 6 )cycloalkyl, phenyl, 5-6 membered
  • heteroaryl or 5-6 membered heterocycloalkyl
  • alkyl represents a saturated, straight or branched hydrocarbon group having the specified number of carbon atoms.
  • (Ci-C 4 )alkyl refers to an alkyl moiety containing from 1 to 4 carbon atoms.
  • Exemplary alkyls include, but are not limited to methyl, ethyl, ⁇ -propyl, isopropyl, «-butyl, isobutyl, s-butyl, and t- butyl.
  • substituent term such as “alkyl”
  • another substituent term for example as in “hydroxy(Ci-C 4 )alkyl” or "aryl(Ci-C 4 )alkyl
  • the linking substituent term e.g., alkyl
  • alkyl is intended to encompass a divalent moiety, wherein the point of attachment is through that linking substituent.
  • aryl(Ci-C 4 )alkyl groups include, but are not limited to, benzyl (phenylmethyl), 1-methylbenzyl
  • hydroxy(Ci-C 4 )alkyl examples include, but are not limited to, hydroxymethyl, hydroxyethyl, and hydroxyisopropyl.
  • halo(Ci-C 4 )alkyl represents a group having one or more halogen atoms, which may be the same or different, at one or more carbon atoms of an alkyl moiety containing from 1 to 4 carbon atoms.
  • halo(Ci-C 4 )alkyl groups include, but are not limited to, -CF 3 (trifluorom ethyl), -CC1 3 (trichloromethyl), 1, 1-difluoroethyl, 2,2,2- trifluoroethyl, and hexafluoroisopropyl.
  • Alkenyl refers to straight or branched hydrocarbon group having at least 1 and up to 3 carbon-carbon double bonds. Examples include ethenyl and propenyl.
  • Alkoxy refers to an "alkyl-oxy-" group, containing an alkyl moiety attached through an oxygen linking atom.
  • (Ci-C 4 )alkoxy represents a saturated, straight or branched hydrocarbon moiety having at least 1 and up to 4 carbon atoms attached through an oxygen linking atom.
  • Exemplary "(Ci-C 4 )alkoxy” groups include, but are not limited to, methoxy, ethoxy, «-propoxy, isopropoxy, «-butoxy, s- butoxy, and t-butoxy.
  • halo(Ci-C 4 )alkoxy refers to a "haloalkyl-oxy-" group, containing a “halo(Ci-C 4 )alkyl” moiety attached through an oxygen linking atom, which
  • halo(Ci-C 4 )alkyl refers to a moiety having one or more halogen atoms, which may be the same or different, at one or more carbon atoms of an alkyl moiety containing from 1 to 4 carbon atoms.
  • exemplary "halo(Ci-C 4 )alkoxy" groups include, but are not limited to, - OCFIF 2 (difluoromethoxy), -OCF 3 (trifluoromethoxy), -OCH 2 CF 3 (trifluoroethoxy), and -OCH(CF 3 ) 2 (hexafluoroisopropoxy).
  • a carbocyclic group is a cyclic group in which all of the ring members are carbon atoms, which may be saturated, partially unsaturated (non-aromatic) or fully unsaturated (aromatic).
  • the term “carbocyclic” includes cycloalkyl and aryl groups. "Cycloalkyl” refers to a non-aromatic, saturated, cyclic hydrocarbon group containing the specified number of carbon atoms. For example, the term
  • (C 3 -C 6 )cycloalkyl refers to a non-aromatic cyclic hydrocarbon ring having from three to six ring carbon atoms.
  • Exemplary "(C 3 -C 6 )cycloalkyl” groups include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • cycloalkyloxy or “cycloalkoxy” refer to a group containing a cycloalkyl moiety, defined hereinabove, attached through an oxygen linking atom.
  • Exemplary "(C 3 -C 6 )cycloalkyloxy” groups include cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, and cyclohexyloxy.
  • Aryl refers to a group or moiety comprising an aromatic, monocyclic or bicyclic hydrocarbon radical containing from 6 to 10 carbon ring atoms and having at least one aromatic ring.
  • aryl groups are phenyl, naphthyl, indenyl, and
  • dihydroindenyl indanyl
  • aryl is phenyl
  • a heterocyclic group is a cyclic group having, as ring members, atoms of at least two different elements, which cyclic group may be saturated, partially unsaturated
  • heterocyclic or “heterocyclyl” includes heterocycloalkyl and heteroaryl groups. It is to be understood that the terms heterocyclic, heterocyclyl, heteroaryl, and heterocycloalkyl, are intended to encompass stable groups where a ring nitrogen heteroatom is optionally oxidized (e.g., heteroaryl groups containing an N-oxide, such as oxo-pyridyl (pyridyl-N-oxide) and oxo-oxadiazolyl (oxo-4,5-dihydro-l,3,4-oxadiazolyl) or where a ring sulfur heteroatom is optionally oxidized (e.g., heterocycloalkyl groups containing sulfones or sulfoxide moieties, such as tetrahydrothienyl-1 -oxide (tetrahydrothienyl sulfoxide,
  • Heterocycloalkyl refers to a non-aromatic, monocyclic or bicyclic group containing 3-10 ring atoms, being saturated or having one or more degrees of unsaturation and containing one or more (generally one or two) heteroatom substitutions independently selected from oxygen, sulfur, and nitrogen.
  • heterocycloalkyl groups include, but are not limited to, aziridinyl, thiiranyl, oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrrolinyl, pyrazolidinyl, pyrazolinyl, imidazolidinyl, imidazolinyl, oxazolinyl, thiazolinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, 1,3- dioxolanyl, piperidinyl, piperazinyl, tetrahydropyranyl, dihydropyranyl,
  • Examples of "4-membered heterocycloalkyl” groups include oxetanyl, thietanyl and azetidinyl.
  • 5-6-membered heterocycloalkyl represents a non aromatic, monocyclic group, which is saturated or partially unsaturated, containing 5 or 6 ring atoms, which includes one or two heteroatoms selected independently from oxygen, sulfur, and nitrogen.
  • Illustrative examples of 5 to 6-membered heterocycloalkyl groups include, but are not limited to pyrrolidinyl, piperidinyl, piperazinyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, tetrahydrothiopyranyl, morpholinyl, and thiomorpholinyl.
  • Heteroaryl represents a group or moiety comprising an aromatic monocyclic or bicyclic radical, containing 5 to 10 ring atoms, including 1 to 4 heteroatoms independently selected from nitrogen, oxygen and sulfur. This term also encompasses bicyclic heterocyclic-aryl groups containing either an aryl ring moiety fused to a heterocycloalkyl ring moiety or a heteroaryl ring moiety fused to a cycloalkyl ring moiety.
  • heteroaryls include, but are not limited to, furanyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiadiazolyl, isothiazolyl, pyridinyl (pyridyl), oxo-pyridyl (pyridyl-N-oxide), pyridazinyl, pyrazinyl, pyrimidinyl, triazinyl, benzofuranyl, isobenzofuryl, 2,3- dihydrobenzofuryl, 1,3-benzodioxolyl, dihydrobenzodioxinyl, benzothienyl, indolizinyl, indolyl, isoindolyl, dihydroindolyl, benzimid
  • 5-6-membered heteroaryl represents an aromatic monocyclic group containing 5 or 6 ring atoms, including at least one carbon atom and 1 to 4 heteroatoms independently selected from nitrogen, oxygen and sulfur.
  • Selected 5- membered heteroaryl groups contain one nitrogen, oxygen, or sulfur ring heteroatom, and optionally contain 1, 2, or 3 additional nitrogen ring atoms.
  • Selected 6-membered heteroaryl groups contain 1, 2, or 3 nitrogen ring heteroatoms.
  • Examples of 5- membered heteroaryl groups include furyl (furanyl), thienyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, thiazolyl, isothiazolyl, thiadiazolyl, oxazolyl, isoxazolyl, oxadiazolyl and oxo- oxadiazolyl.
  • Selected 6-membered heteroaryl groups include pyridinyl, oxo-pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl and triazinyl.
  • Bicyclic heteroaryl groups include 6,5-fused heteroaryl (9-membered heteroaryl) and 6,6-fused heteroaryl (10-membered heteroaryl) groups.
  • 6,5-fused heteroaryl (9-membered heteroaryl) groups include benzothienyl, benzofuranyl, indolyl, indolinyl, isoindolyl, isoindolinyl, indazolyl, indolizinyl, isobenzofuryl,
  • 6,6-fused heteroaryl (10-membered heteroaryl) groups include quinolyl, isoquinolyl, phthalazinyl, naphthridinyl (1,5-naphthyridinyl, 1,6-naphthyridinyl, 1,7-naphthyridinyl, 1,8-naphthyridinyl), quinazolinyl, quinoxalinyl, 4H-quinolizinyl, tetrahydroquinolinyl, cinnolinyl, and pteridinyl.
  • bicyclic ring systems may be attached at any suitable position on either ring.
  • Hydroxo or “hydroxyl” is intended to mean the radical -OH.
  • cyano refers to the group - CN.
  • the term "optionally substituted” indicates that a group (such as an alkyl, cycloalkyl, alkoxy, heterocycloalkyl, aryl, or heteroaryl group) or ring or moiety (such as a carbocyclic or heterocyclic ring or moiety) may be unsubstituted, or the group, ring or moiety may be substituted with one or more substituent(s) as defined.
  • groups may be selected from a number of alternative groups, the selected groups may be the same or different.
  • pharmaceutically acceptable refers to those compounds, materials, compositions, and dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • the terms "compound(s) of the invention” or “compound(s) of this invention” mean a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), as defined herein, in any form, i.e., any salt or non-salt form (e.g., as a free acid or base form, or as a salt, particularly a pharmaceutically acceptable salt thereof) and any physical form thereof (e.g., including non-solid forms (e.g., liquid or semi-solid forms), and solid forms (e.g., amorphous or crystalline forms, specific polymorphic forms, solvate forms, including hydrate forms (e.g., mono-, di-and hemi- hydrates)), and mixtures of various forms.
  • any salt or non-salt form e.g., as a free acid or base form, or as a salt, particularly a pharmaceutically acceptable salt thereof
  • any physical form thereof e.g., including non-solid forms (e.g., liquid or
  • Formula (I), particularly, compounds of any one of Formulas (I-IV), as defined herein, in any salt or non-salt form, and in any physical form thereof, may have varying levels of activity, different bioavailabilities and different handling properties for formulation purposes.
  • X is O, S, SO,
  • X is O, S, SO, S0 2 , NH, CO, CH 2 , or N(CH 3 ).
  • X is S, SO, S0 2 , or CO. In yet another embodiment, X is CF 2 ,
  • X is O, CH 2 , NH or N(CH 3 ).
  • X is O or CH 2 .
  • Y is CH 2 or
  • Y is CH 2 CH 2 . In selected embodiments, Y is
  • Z 1 is CR 1 and Z 2 , Z 3 and Z 4 are each CH.
  • Z 1 , Z 2 , and Z 4 are each CH and Z 3 is CR 3 . In a further embodiment, Z 1 , Z 3 , and Z 4 are each CH and Z 2 is CR 2 . In a still further embodiment,
  • Z 1 , Z 2 , and Z 3 are each CH and Z 4 is CR 4 .
  • Z 1 and Z 2 are CH, Z 3 is CR 3 , and Z 4 is CR 4 .
  • Z 1 and Z 3 are CH, Z 2 is CR 2 , and Z 4 is CR 4 .
  • Z 1 is CH, Z 2 is CR 2 , Z 3 is CR 3 ,
  • Z 4 is CR 4 .
  • Z 1 and Z 3 are both N, Z 2 is CH and Z 4 is CH or CR 4 .
  • Z 4 is CH or CR 4 .
  • Z 1 and Z 3 are both N, Z 2 is CH or CR 2 and Z 4 is
  • Z 1 is N
  • Z 2 is CR 2 and Z 3 and Z 4 are CH.
  • Z 3 is N
  • Z 2 , Z 3 and Z 4 are CH.
  • R 1 is fluoro
  • R 1 is methyl
  • one of R 2 and R 3 is halogen, cyano, (Ci-C 6 )alkyl,
  • R 2 and R 3 is halogen, cyano or (Ci-C 6 )alkyl.
  • one of R 2 and R 3 is halogen, cyano, (Ci-C 6 )alkyl, halo(Ci-C 4 )alkyl, (Ci-C 6 )alkoxy, hydroxyl, B(OH) 2 , -COOH, halo(Ci-C 4 )alkylC(OH) 2 -, (Ci-C 4 )alkoxy(Ci-C 4 )alkoxy, (Ci-C 4 )alkylS0 2 -,
  • 5-6 membered heterocycloalkyl and 5-6 membered heteroaryl are optionally substituted by 1 or 2 substituents each independently selected from the group consisting of (Ci-C )alkyl and -(Ci-C )alkyl-CN;
  • R 2 and R 3 is halogen or (Ci-C 6 )alkyl.
  • R 2 is halogen, cyano, (Ci-C 6 )alkyl, (Ci-C 6 )alkoxy, halo(Ci-C 4 )alkoxy, hydroxyl, B(OH) 2 , -COOH, halo(Ci-C 4 )alkylC(OH) 2 -,
  • R 2 is halogen, cyano, (Ci-C 6 )alkyl, hydroxyl, B(OH) 2 , -COOH, halo(Ci-C 4 )alkylC(OH) 2 -, (Ci-C 4 )alkoxy(Ci-C 4 )alkoxy, or 5-6 membered heteroaryl, wherein said 5-6 membered heteroaryl is optionally substituted by a (Ci-C 3 )alkyl substituent; and Z 3 is CH.
  • R 3 is halogen, (Ci-C 6 )alkyl, halo(Ci-C 4 )alkyl,
  • R 3 is halogen, (Ci-C 6 )alkyl, halo(Ci-C )alkyl,
  • heterocycloalkyl-C(O)- 5-6 membered heterocycloalkyl-(Ci-C 4 )alkyl- HC(0)-, 5-6 membered heterocycloalkyl-(Ci-C 4 )alkoxy-, 5-6 membered heteroaryl, or 5-6 membered heteroaryl-C(0) H
  • said 5-6 membered heterocycloalkyl and 5-6 membered heteroaryl are optionally substituted by (Ci-C 3 )alkyl or -(Ci-C 3 )alkyl-CN; and Z 2 is CH.
  • R 2 is fluoro, chloro, bromo, -CN, -CH 3 ,
  • R 2 is fluoro, chloro, bromo, -CN,
  • R 2 is chloro, bromo, -CN, -CH 3 ,
  • R 3 is fluoro, chloro, bromo, -CN, -OCH 3 ,
  • pyrazol-4-yl l-methyl-pyrazol-3-yl, l-methyl-pyrrol-4-yl-C(0)NH-, 5- methyl-l,3,4-oxadiazol-2-yl, or 5-oxo-4,5-dihydro-l,3,4-oxadiazol-2-yl.
  • R 3 is fluoro, chloro, bromo, -OCH 3 ,
  • R is fluoro, chloro, bromo, -OCH 3 ,
  • R 4 is fluoro, chloro, methyl, or trifluorom ethyl. In another embodiment, R 4 is fluoro. In yet another embodiment, R 4 is methyl.
  • R 5 is H. In another embodiment, R 5 is methyl.
  • A is phenyl, 5-6 membered heteroaryl, or 5-6 membered heterocycloalkyl, wherein the carbonyl moiety and L are substituted 1,3 on ring A.
  • A is a 5 membered heteroaryl containing one oxygen or sulfur atom and optionally containing one or two nitrogen atoms;
  • A is furyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, or oxadiazolyl (more specifically, 1, 2, 4-oxadiazolyl or 1, 3, 4-oxadiazolyl).
  • A is a 5 membered heteroaryl containing one nitrogen atom and optionally containing one, two or three additional nitrogen atoms, specifically;
  • A is pyrrolyl, pyrazolyl, imidazolyl, triazolyl (more specifically, 1, 2, 3-triazolyl or 1, 2, 4-triazolyl) or tetrazolyl. In selected embodiments, A is triazolyl. In yet embodiment of this invention, A is a 5 or 6 membered heterocycloalkyl
  • A is piperidinyl or pyrrolidinyl.
  • A is a 6 membered aromatic group selected from phenyl and pyridyl.
  • Another embodiment of this invention is directed to a compound according to Formula (II):
  • a 1 is C
  • a 4 is C or N
  • a 2 , A 3 , and A 5 are each independently selected from CH, CR A , O, S, N, NH and NR A to form a furyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl or tetrazolyl ring moiety, wherein said ring moiety contains 0 or 1 of CR A and NR A ; and wherein X, Z 1 , Z 2 , Z 3 , Z 4 , R 5 , L, and B are as defined herein,
  • a 1 is C
  • a 4 is C or N
  • a 2 , A 3 , and A 5 are each independently selected from CH, O, N, and NH to form an oxazolyl, isoxazolyl, oxadiazolyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl or tetrazolyl ring moiety.
  • a 1 and A 4 are each C, and A 2 , A 3 and A 5 are each independently selected from N and NH to form a triazolyl ring moiety.
  • A is piperidinyl or pyrrolidinyl
  • A is piperidinyl or pyrrolidinyl
  • s is 0 or 1
  • a 10 is N and X, Z 1 , Z 2 , Z 3 , Z 4 , R 5 R A , m, L, and B are as defined herein.
  • m is 0 and A is an unsubstituted piperidinyl or pyrrolidinyl moiety.
  • m is 0.
  • m is 1 and R A is (Ci-C 4 )alkyl, specifically R A is (Ci-C 2 )alkyl.
  • R A is methyl.
  • a further embodiment of this invention wherein A is phenyl, pyridinyl, or pyridinyl- -oxide, may be represented by Formula (IV):
  • a 6 , A 7 , A 8 , and A 9 are each CH;
  • one of A 6 , A 7 , A 8 , and A 9 is CR A and the others of A 6 , A 7 , A 8 , and A 9 are CH;
  • a 6 , A 7 , A 8 , and A 9 is N and the others of A 6 , A 7 , A 8 , and A 9 are
  • one of A 6 , A 7 , A 8 , and A 9 is N-0 and the other of A 6 , A 7 , A 8 , and A 9 are
  • L is O, S, H, N(CH 3 ), CH 2 , CH 2 CH 2 , CH(CH 3 ), CHF, CF 2 , CH 2 0, CH 2 N(CH 3 ), CH 2 H, or CH(OH).
  • L is O, S, N(CH 3 ), CH 2 , CH 2 CH 2 , CH(CH 3 ), CF 2 , CH 2 0, CH 2 N(CH 3 ), or CH(OH).
  • L is CH 2 0,
  • L is N(CH 3 ),
  • L is CH(CH 3 ), or CH(OH).
  • L is -(R)CH(CH 3 ).
  • L is O, CH 2 , or NH.
  • L is O.
  • L is CH 2 .
  • B is an
  • B is an optionally substituted 5-6 membered heteroaryl or 5-6 membered heterocycloalkyl.
  • B is an optionally substituted pyrazolyl, thienyl, pyridinyl (pyridyl), oxo-pyridyl, pyrimidinyl, isoxazolyl, morpholinyl, tetrahydropyranyl or tetrahydrofuranyl, wherein the pyrazolyl, thienyl, pyridinyl (pyridyl), oxo-pyridyl, pyrimidinyl, isoxazolyl, morpholinyl, tetrahydropyranyl or tetrahydrofuranyl is optionally substituted by one or two independently selected (Ci-C 4 )alkyl substituents.
  • Bis an optionally substituted pyrazolyl, thienyl, pyridinyl (pyridyl), oxo-pyridyl, pyrimidinyl, isoxazolyl, morpholinyl, or tetrahydrofuranyl wherein the pyrazolyl, thienyl, pyridinyl (pyridyl), oxo-pyridyl, pyrimidinyl, isoxazolyl, morpholinyl, or tetrahydrofuranyl is optionally substituted by one or two independently selected (Ci-C 4 )alkyl substituents.
  • B is thien-2-yl (thiophen-2-yl), 5-methyl-thien-2-yl (5-methyl-thiophen-2-yl), pyrazol-l-yl, 3,5-dimethylpyrazol-l-yl, 4-methylpyrazol-l-yl,
  • B is thien-2-yl (thiophen-2-yl), 5-methyl- thien-2-yl (5-methyl-thiophen-2-yl), pyrazol-l-yl, 3,5-dimethylpyrazol-l-yl, 4-methylpyrazol-l-yl, 3,5-dimethylisoxazol-4-yl, tetrahydrofuran-2-yl, morpholin-4-yl, pyridin-2-yl, 2-oxo-pyridin-l-yl, 6-methylpyridin-3-yl, and 2-methylpyrimidin-5-yl.
  • B is thien-2-yl, pyrazol-l-yl,
  • B is unsubstituted (C 3 -C 6 )cycloalkyl or phenyl.
  • B is unsubstituted cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
  • B is unsubstituted cyclopentyl or cyclohexyl.
  • B is
  • B is substituted phenyl.
  • B is phenyl, substituted by 1 or 2 substituents independently
  • B is phenyl, substituted by 1 or 2 substituents independently
  • B is phenyl, substituted by 1 or 2
  • B is phenyl, substituted by 1 or 2 fluoro substituents.
  • B is cyclopentyl, cyclohexyl, 2-methylphenyl, 4-methylphenyl,
  • B is
  • the moiety -L-B is (C 3 -C 6 )alkyl, (C 3 -C 6 )alkoxy, halo(C 3 -C 6 )alkoxy, (C 3 -C 6 )alkenyl, or
  • -OCH 2 CH CH 2 , -CH 2 CH 2 CH 2 CH 2 CH 3 , -OCH 2 CH 2 CH 2 CH 3 , -CH 2 CH 2 CH 3 ,
  • -L-B is
  • -OCH 2 CH CH 2 , -CH 2 CH 2 CH 2 CH 2 CH 3 , -OCH 2 CH 2 CH 2 CH 3 , -CH 2 CH 2 CH 3 , or
  • Representative compounds of this invention include the compounds of the
  • the present invention encompasses compounds of Formula (I) as the free base and as salts thereof, for example as a pharmaceutically acceptable salt thereof.
  • the invention relates to compounds of Formula (I) in the form of a free base.
  • the invention relates to compounds of Formula (I) in the form of a salt, particularly, a pharmaceutically acceptable salt.
  • the invention relates to compounds of the Examples in the form of a free base.
  • the invention relates to compounds of the Examples in the form of a salt, particularly, a pharmaceutically acceptable salt.
  • this invention is directed to (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide; (S)-5-benzyl-N- (6-fluoro-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b] [l,4]oxazepin-3-yl)-4H-l,2,4- triazole-3-carboxamide; and 5-benzyl-N-(7-bromo-2-oxo-2,3,4,5-tetrahydro-lH- benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3-carboxamide, or a salt thereof, particularly a pharmaceutically acceptable salt, thereof.
  • this invention is directed to (S)-5- benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4- triazole-3-carboxamide; (S)-5-benzyl-N-(6-fluoro-5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b] [l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide; or 5 -benzyl -N- (7-bromo-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3- carboxamide, or a pharmaceutically acceptable salt, thereof.
  • this invention is directed to (S)-5-benzyl-N-(5-methyl-4-oxo- 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide and (S)-5- benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3- carboxamide, or a pharmaceutically acceptable salt thereof.
  • This invention is directed to (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide or a salt, particularly a pharmaceutically acceptable salt, thereof.
  • one particular compound of the invention is (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide (free base).
  • the compound of the invention is a salt of (S)-5-benzyl-N-(5-methyl- 4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide.
  • the compound of the invention is a pharmaceutically acceptable salt of (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H- l,2,4-triazole-3-carboxamide.
  • the compound of the invention is a base-addition salt of (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide.
  • the compound of the invention is a crystalline form of anhydrous (S)-5- benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4- triazole-3 -carboxamide (free base) characterized by the PXRD pattern of Figure 7.
  • a particular compound of the invention is a crystalline form of anhydrous (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)- 4H-l,2,4-triazole-3 -carboxamide (free base) characterized by the diffraction data in Table 1.
  • This invention is directed (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH- benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3-carboxamide, or a salt, particularly a
  • one particular compound of the invention is (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)- 4H-l,2,4-triazole-3 -carboxamide (free base).
  • the compound of the invention is a salt of (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH- benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3-carboxamide.
  • the compound of the invention is a pharmaceutically acceptable salt of (S)-5-benzyl-N-(7- chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3- carboxamide.
  • the compound of the invention is a base-addition salt of (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4- tri azol e-3 -carb oxami de .
  • this invention is directed to a compound of
  • X is O, S, SO, S0 2 , NH, CO, CH 2 , or N(CH 3 );
  • Y is CH 2 or CH 2 CH 2 ;
  • Z 1 , Z 2 , Z 3 , and Z 4 are each CH; or Z 1 is CR 1 and Z 2 , Z 3 and Z 4 are each
  • Z 2 is CR 2 ; or Z l , Z 2 , and Z 3 are each CH and Z 4 is CR 4 ; or Z 1 and Z 2 are CH,
  • Z 3 is CR 3 , and Z 4 is CR 4 ; or Z 1 and Z 4 are CH, Z 2 is CR 2 , and Z 3 is CR 3 ; or Z 1
  • Z 3 are CH, Z 2 is CR 2 , and Z 4 is CR 4 ; or Z 1 is CH, Z 2 is CR 2 , Z 3 is CR 3 , and
  • Z 4 is CR 4 ; or Z 1 and Z 3 are both N, Z 2 is CH and Z 4 is CH or CR 4 ; or Z 1 and Z 3
  • Z 2 is CH or CR 2 and Z 4 is CH; or Z 1 is N, Z 2 is CR 4 and Z 3 and Z 4
  • R 1 is methyl
  • R 2 is fluoro, chloro, bromo, -CN, -CH 3 , -OCH 3 , -OCHF 2 , -OH, B(OH) 2 , CF 3 C(OH) 2 -, CH 3 OCH 2 CH 2 O-, cyclopropyl, 5H-tetrazol-5-yl, pyrazol-3-yl, or 5-methyl-l,3,4-oxadiazol-2-yl;
  • R 3 is fluoro, chloro, bromo, -CN, -OCH 3 , -OCHF 2 , B(OH) 2 , -COOH, CH 3 S0 2 -, CH 3 S0 2 NHC(0)-, CH 3 C(0)NH-, (CH 3 ) 2 NC(0)-, CH 3 OC(0)-, (CH 3 )C(0)N(CH 3 )-, HOCH 2 CH 2 C(0)NH-, CH 3 OCH 2 CH 2 NHC(0)NH-, CH 3 S0 2 CH 2 CH 2 NHC(0)-, CH 3 CH 2 NHC(0)NH-, CH 3 OC(0)NH-,
  • morpholin-4-yl-CO- pyrrolidin-l-yl-CH 2 CH 2 NHC(0)-, pyridin-2-yl, tetrahydrofuran-2-yl-CH 2 0-, pyrrolidin-l-yl-CH 2 CH 2 0-, tetrazol-5-yl, l-(2- cyanoethyl)-tetrazol-5-yl, pyrazol-l-yl, pyrazol-3-yl, pyrazol-4-yl, 1-methyl- pyrazol-3-yl, 1 -methyl-pyrrol -4-yl-C(0)NH-, 5-methyl-l,3,4-oxadiazol-2-yl, or 5 -oxo-4, 5 -dihy dro- 1 , 3 ,4-oxadiazol-2-yl ;
  • R 4 is fluoro, chloro, methyl, or trifluoromethyl
  • R 5 is H or methyl
  • A is furyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, 1, 2, 4-oxadiazolyl, 1, 3, 4-oxadiazolyl, pyrrolyl, pyrazolyl, imidazolyl, 1, 2, 3-triazolyl, 1, 2, 4- triazolyl, tetrazolyl, piperidinyl, pyrrolidinyl, phenyl or pyridyl;
  • n 0 or m is 1 and R A is methyl
  • L is O, S, N(CH 3 ), CH 2 , CH 2 CH 2 , CH(CH 3 ), -(R)CH(CH 3 ), CF 2 , CH 2 0, CH 2 N(CH 3 ), or CH(OH);
  • B is thien-2-yl, 5-methyl-thien-2-yl, pyrazol-l-yl,
  • this invention is directed to a compound of Formula (I) wherein:
  • X is O, S, SO, S0 2 , NH, CO, CH 2 , or N(CH 3 );
  • Y is CH 2 or CH 2 CH 2 ;
  • Z 1 , Z 2 , Z 3 , and Z 4 are each CH; or Z 1 is CR 1 and Z 2 , Z 3 and Z 4 are each
  • Z 1 , Z 2 , and Z 4 are each CH and Z 3 is CR 3 ; or Z 1 , Z 3 , and Z 4 are each CH and Z 2 is CR 2 ; or Z l , Z 2 , and Z 3 are each CH and Z 4 is CR 4 ; or Z 1 and Z 3 are CH, Z 2 is CR 2 , and Z 4 is CR 4 ; or Z 1 and Z 3 are both N, Z 2 is CH and Z 4 is CH or CR 4 ; or Z 1 is N, Z 2 is CR 4 andZ 3 and Z 4 are CH; or Z 3 is N, and Z 2 , Z 3 and Z 4 are CH;
  • R 1 is methyl
  • R 2 is fluoro, chloro, bromo, -CN, -CH 3 , -OH, B(OH) 2 , CF 3 C(OH) 2 -, CH 3 OCH 2 CH 2 0-, 5H-tetrazol-5-yl, pyrazol-3-yl, or 5-methyl-l,3,4-oxadiazol- 2-yl;
  • R 3 is fluoro, chloro, bromo, -OCH 3 , B(OH) 2 , -COOH, CH 3 S0 2 -,
  • R 4 is fluoro, chloro, methyl, or trifluoromethyl
  • R 5 is H or methyl
  • A is furyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, 1, 2, 4-oxadiazolyl, 1, 3, 4-oxadiazolyl, pyrrolyl, pyrazolyl, imidazolyl, 1, 2, 3-triazolyl, 1, 2, 4- triazolyl, tetrazolyl, piperidinyl, pyrrolidinyl, phenyl or pyridyl;
  • n 0 or m is 1 and R A is methyl
  • L is O, S, N(CH 3 ), CH 2 , CH 2 CH 2 , CH(CH 3 ), CF 2 , CH 2 0, CH 2 N(CH 3 ), or
  • B is thien-2-yl, 5-methyl-thien-2-yl, pyrazol-l-yl,
  • this invention is directed to a compound of Formula (I) wherein:
  • X is O, CH 2 , NH or N(CH 3 );
  • Y is CH 2 or CH 2 CH 2 ;
  • Z 1 , Z 2 , Z 3 , and Z 4 are each CH; or Z 1 is CR 1 and Z 2 , Z 3 and Z 4 are each CH; or Z l , Z 2 , and Z 4 are each CH and Z 3 is CR 3 ; or Z l , Z 3 , and Z 4 are each CH and Z 2 is CR 2 ; or Z l , Z 2 , and Z 3 are each CH and Z 4 is CR 4 ; or Z 1 and Z 3 are CH, Z 2 is CR 2 , and Z 4 is CR 4 ; or Z 1 and Z 3 are both N, Z 2 is CH and Z 4 is CH or CR 4 ; or Z 1 is N, Z 2 is CR 4 andZ 3 and Z 4 are CH; or Z 3 is N, and Z 2 , Z 3 and Z 4 are CH;
  • R 1 is methyl
  • R 2 is chloro, bromo, -CN, -CH 3 , -OH, B(OH) 2 , CF 3 C(OH) 2 -, CH 3 OCH 2 CH 2 0-, 5H-tetrazol-5-yl, , pyrazol-3-yl, or 5-methyl-l,3,4- oxadiazol-2-yl;
  • R 3 is fluoro, chloro, bromo, -OCH 3 , B(OH) 2 , -COOH, CH 3 S0 2 -, CH 3 S0 2 HC(0)-, CH 3 C(0) H-, (CH 3 ) 2 NC(0)-, CH 3 OC(0)-,
  • morpholin-4-yl-CO- pyrrolidin- 1 -yl-CH 2 CH 2 HC(0)-, tetrahydrofuran-2-yl- CH 2 0-, pyrrolidin- l-yl-CH 2 CH 2 0-, tetrazol-5-yl, l-(2-cyanoethyl)-tetrazol-5- yl, pyrazol-l-yl, pyrazol-3-yl, l-methyl-pyrazol-3-yl, 1 -methyl-pyrrol -4-yl- C(0) H-, or 5-oxo-4,5-dihydro-l,3,4-oxadiazol-2-yl;
  • R 4 is fluoro or methyl;
  • A is furyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, 1, 2, 4-oxadiazolyl, 1,
  • n 0 or m is 1 and R A is methyl
  • L is O, S, N(CH 3 ), CH 2 , CH 2 CH 2 , CH(CH 3 ), CF 2 , CH 2 0, CH 2 N(CH 3 ), or
  • B is thien-2-yl, pyrazol-l-yl, 3,5-dimethylpyrazol-l-yl,
  • this invention is directed to a compound of Formula (I) wherein X is O or CH 2; Y is CH 2 ; Z 1 , Z 2 , and Z 4 are each CH
  • Z 3 is CR 3 ; or Z 1 , Z 3 , and Z 4 are each CH and Z 2 is CR 2 ; or Z 1 , Z 2 , and Z 3 are each CH and Z 4 is CR 4 ; or Z 1 and Z 3 are CH, Z 2 is CR 2 , and Z 4 is CR 4 ; R 2 is
  • R 3 is 5-methyl-l,3,4-oxadiazol-2-yl; R 4 is
  • R 5 is H or methyl
  • A is triazolyl
  • m is 0
  • L is CH 2
  • B is
  • the compounds of this invention contain one or more asymmetric centers (also referred to as a chiral center), such as a chiral carbon, or a chiral -SO- moiety.
  • asymmetric centers also referred to as a chiral center
  • the stereochemistry of the chiral carbon center present in compounds of this invention is generally represented in the compound names and/or in the chemical structures illustrated herein.
  • Compounds of this invention containing one or more chiral centers may be present as racemic mixtures, diastereomeric mixtures, enantiomerically enriched mixtures, diastereomerically enriched mixtures, or as enantiomerically or diastereomerically pure individual stereoisomers.
  • Individual stereoisomers of a compound of this invention may be resolved (or mixtures of stereoisomers may be enriched) using methods known to those skilled in the art. For example, such resolution may be carried out (1) by formation of diastereoisomeric salts, complexes or other derivatives; (2) by selective reaction with a stereoisomer-specific reagent, for example by enzymatic oxidation or reduction; or (3) by gas-liquid or liquid chromatography in a chiral environment, for example, on a chiral support such as silica with a bound chiral ligand or in the presence of a chiral solvent.
  • stereoisomers may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer to the other by asymmetric transformation.
  • the invention also includes various deuterated forms of the compounds of this invention, a specific example of which is N-[(3S)-7-deuterio -l-methyl-2-oxo-2,3,4,5- tetrahydro-lH-l-benzazepin-3-yl]-5-(phenylmethyl)-lH-pyrazole-3-carboxamide.
  • Each available hydrogen atom attached to a carbon atom may be independently replaced with a deuterium atom.
  • a person of ordinary skill in the art will know how to synthesize deuterated forms of the compounds of this invention.
  • a-deuterated a-amino acids are commercially available or may be prepared by conventional techniques (see for example: Elemes, Y. and Ragnarsson, U. J.
  • deuterated reagents e.g. by reduction using lithium aluminum deuteride or sodium borodeuteride or by metal-halogen exchange followed by quenching with D 2 0 or methanol- ⁇
  • solvates (particularly, hydrates) of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), including solvates of salts of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), may be formed when solvent molecules are incorporated into the crystalline lattice during crystallization.
  • the present invention includes within its scope all possible stoichiometric and non-stoichiometric salt and/or hydrate forms.
  • the compound or salt including solvates (particularly, hydrates) thereof, may exist in crystalline forms, non-crystalline forms or a mixture thereof.
  • the compound or salt, or solvates (particularly, hydrates) thereof may also exhibit polymorphism (i.e. the capacity to occur in different crystalline forms). These different crystalline forms are typically known as "polymorphs.”
  • polymorphs typically known as “polymorphs.”
  • the disclosed compound, or solvates (particularly, hydrates) thereof also include all polymorphs thereof. Polymorphs have the same chemical composition but differ in packing, geometrical arrangement, and other descriptive properties of the crystalline solid state.
  • Polymorphs may have different physical properties such as shape, density, hardness, deformability, stability, and dissolution properties. Polymorphs typically exhibit different melting points, IR spectra, and X-ray powder diffraction patterns, which may be used for identification. One of ordinary skill in the art will appreciate that different polymorphs may be produced, for example, by changing or adjusting the conditions used in crystallizing/recrystallizing the compound. It is well known and understood to those skilled in the art that the apparatus employed, humidity, temperature, orientation of the powder crystals, and other parameters involved in obtaining a powder X-ray diffraction (PXRD) pattern may cause some variability in the appearance, intensities, and positions of the lines in the diffraction pattern.
  • PXRD powder X-ray diffraction
  • a powder X-ray diffraction pattern that is "substantially in accordance" with that of the Figure provided herein is a PXRD pattern that would be considered by one skilled in the art to represent a compound possessing the same crystal form as the compound that provided the PXRD pattern of the Figure.
  • the PXRD pattern may be identical to that of Figure 7, or more likely it may be somewhat different.
  • Such a PXRD pattern may not necessarily show each of the lines of the diffraction patterns presented herein, and/or may show a slight change in appearance, intensity, or a shift in position of said lines resulting from differences in the conditions involved in obtaining the data.
  • a person skilled in the art is capable of determining if a sample of a crystalline compound has the same form as, or a different form from, a form disclosed herein by comparison of their PXRD patterns. For example, one skilled in the art can overlay a PXRD pattern of a sample of a crystalline form of anhydrous (S)-5-benzyl-N- (5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3- carboxamide (free base) with the PXRD pattern of Fig.
  • the sample form can be readily and accurately identified as having the same form as the crystalline form of anhydrous (S)-5-benzyl-N-(5- methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3- carboxamide (free base) described herein.
  • S anhydrous
  • a person skilled in the art is capable of determining if a given diffraction angle (expressed in °2 ⁇ ) obtained from a PXRD pattern is at about the same position as a recited value.
  • Suitable pharmaceutically acceptable salts can include acid or base addition salts.
  • salts and solvates e.g. hydrates and hydrates of salts
  • the counterion or associated solvent is pharmaceutically acceptable.
  • Salts and solvates having non-pharmaceutically acceptable counterions or associated solvents are within the scope of the present invention, for example, for use as intermediates in the preparation of other compounds of the invention and their salts and solvates.
  • Salts may be prepared in situ during the final isolation and purification of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV). If a basic compound of Formula (I-IV) is isolated as a salt, the corresponding free base form of that compound may be prepared by any suitable method known to the art, including treatment of the salt with an inorganic or organic base, suitably an inorganic or organic base having a higher pK a than the free base form of the compound.
  • a disclosed compound containing a carboxylic acid or other acidic functional group is isolated as a salt
  • the corresponding free acid form of that compound may be prepared by any suitable method known to the art, including treatment of the salt with an inorganic or organic acid, suitably an inorganic or organic acid having a lower pK a than the free acid form of the compound.
  • This invention also provides for the conversion of one salt of a compound of this invention, e.g., a hydrochloride salt, into another salt of a compound of this invention, e.g., a sulfate salt.
  • Salts of the compounds of Formula (I), particularly compounds of Formulas (I-IV), containing a basic amine or other basic functional group may be prepared by any suitable method known in the art, such as treatment of the free base with an acid.
  • suitable method known in the art, such as treatment of the free base with an acid.
  • pharmaceutically acceptable salts so formed include acetate, adipate, ascorbate, aspartate, benzenesulfonate, benzoate, camphorate, camphor-sulfonate (camsylate), caprate
  • ethanesulfonate (esylate), formate, fumarate, galactarate (mucate), gentisate (2,5- dihydroxybenzoate), glucoheptonate (gluceptate), gluconate, glucuronate, glutamate, glutarate, glycerophosphorate, glycolate, hippurate, hydrobromide, hydrochloride, hydroiodide, isobutyrate, lactate, lactobionate, laurate, maleate, malate, malonate, mandelate, methanesulfonate (mesylate), naphthalene-l,5-disulfonate (napadisylate), naphthalene-sulfonate (napsylate), nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, phosphate, diphosphate, proprionate, pyroglutamate, salicylate, sebacate, stea
  • Salts of the disclosed compounds containing a carboxylic acid or other acidic functional group can be prepared by reacting with a suitable base.
  • a suitable base which affords a pharmaceutically acceptable cation, which includes alkali metal salts (especially sodium and potassium), alkaline earth metal salts (especially calcium and magnesium), aluminum salts and ammonium salts, as well as salts made from physiologically acceptable organic bases such as trimethylamine, triethylamine, morpholine, pyridine, piperidine, picoline, dicyclohexylamine, N P- dibenzylethylenediamine, 2-hydroxyethylamine, /s-(2-hydroxyethyl)amine, tri-(2- hydroxyethyl)amine, procaine, dibenzylpiperidine, dehydroabietylamine, N P- bisdehydroabietylamine, glucamine, N-methylglucamine, collidine, choline, quinine, quinoline, and
  • the compounds of this invention are intended for use in pharmaceutical compositions it will readily be understood that they are each preferably provided in substantially pure form, for example at least 60% pure, more suitably at least 75% pure and preferably at least 85%, especially at least 98% pure (% are on a weight for weight basis). Impure preparations of the compounds may be used for preparing the more pure forms used in the pharmaceutical compositions.
  • the compounds of this invention may be prepared using synthetic procedures illustrated in the Schemes below or by drawing on the knowledge of a skilled organic chemist.
  • the synthesis provided in these Schemes are applicable for producing compounds of the invention having a variety of different R groups employing appropriate precursors, which are suitably protected if needed, to achieve compatibility with the reactions outlined herein. Subsequent deprotection, where needed, affords compounds of the nature generally disclosed. While the Schemes are shown with compounds only of Formulas (I-IV), they are illustrative of processes that may be used to make the compounds of the invention.
  • (S)-N-(4-Oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)carboxamides may be prepared via the general method outlined in Scheme 1.
  • Boc-L-serine can be condensed with an appropriate substituted l-fluoro-2-nitrobenzene with a base, followed by reduction of the nitro group to the amine and cyclisation to the Boc protected (S)-3-amino-2,3- dihydrobenzo[b][l,4]oxazepin-4(5H)-one using an amide coupling agent.
  • the Boc protecting group can be removed using acidic conditions and the resulting free amine can be coupled to the appropriate acid using an amide coupling agent.
  • Boc protected (S)-3- amino-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one can also be methylated to give the Boc protected (S)-3-amino-5-methyl-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one, which can then be deprotected using acidic conditions, and the resulting free amine can be coupled to an appropriate acid using an amide coupling agent.
  • (R)-N-(4-Oxo-2,3,4,5-tetrahydrobenzo[b][l,4]thiazepin-3-yl)carboxamides may be prepared via the general method outlined in Scheme 2.
  • Boc-L-cysteine can be condensed with an appropriate substituted l-fluoro-2-nitrobenzene with a base, followed by reduction of the nitro group to the amine and cyclisation to the Boc protected (R)-3-amino-2,3- dihydrobenzo[b][l,4]thiazepin-4(5H)-one using an amide coupling agent.
  • the Boc protecting group can be removed using acidic conditions and the resulting free amine can be coupled to an appropriate acid using an amide coupling agent.
  • Boc protected (R)-3- amino-2,3-dihydrobenzo[b][l,4]thiazepin-4(5H)-one can also be methylated to give the Boc protected (R)-3-amino-5-methyl-2,3-dihydrobenzo[b][l,4]thiazepin-4(5H)-one, which can then be deprotected using acidic conditions, and the resulting free amine can be coupled to an appropriate acid using an amide coupling agent.
  • (S)-N-(2-Oxo-2,3,4,5-tetrahydro-lH-benzo[b][l,4]diazepin-3-yl)carboxamides may be prepared via the general method outlined in Scheme 3.
  • 3-Aminoalanine can be condensed with the appropriate substituted l-fluoro-2-nitrobenzene with a base, followed by reduction of the nitro group to the amine and cyclisation to the Boc protected (S)-3- amino-4,5-dihydro-lH-benzo[b][l,4]diazepin-2(3H)-one using an amide coupling agent.
  • the Boc protecting group can be removed using acidic conditions and the resulting free amine can be coupled to the appropriate acid using an amide coupling agent.
  • the Boc protected (S)-3-amino-4,5-dihydro-lH-benzo[b][l,4]diazepin-2(3H)-one can also be methylated with methyl iodide using sodium hydride as base to give the Boc protected (S)- 3-amino-l-methyl-4,5-dihydro-lH-benzo[b][l,4]diazepin-2(3H)-one, which can then be deprotected using acidic conditions, and the resulting free amine can be coupled to the appropriate acid using an amide coupling agent.
  • Boc protected (S)-3- amino-4,5-dihydro-lH-benzo[b][l,4]diazepin-2(3H)-one can also be methylated with methyl iodide using potassium carbonate as base in ethanol to give the Boc protected (S)-3- amino-5-methyl-4,5-dihydro-lH-benzo[b][l,4]diazepin-2(3H)-one, which can then be deprotected using acidic conditions, and the resulting free amine can be coupled to the appropriate acid using an amide coupling agent.
  • (S)-N-(6-Oxo-6,7,8,9-tetrahydro-5H-pyrimido[4,5-b][l,4]diazepin-7- yl)carboxamides may be prepared via the general method outlined in Scheme 4.
  • Boc-L- serine can be condensed with 4,6-dichloropyrimidin-5-amine in the presence of a base such as triethylamine, followed by cyclisation to the Boc protected (S)-7-amino-4-chloro-8,9- dihydro-5H-pyrimido[4,5-b][l,4]diazepin-6(7H)-one using an amide coupling agent.
  • the chloride can be removed by reduction using hydrogenation with a palladium/carbon catalyst.
  • the Boc protecting group can then be removed using acidic conditions and the resulting free amine can be coupled to the appropriate acid using an amide coupling agent.
  • (S)-N-(2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl) carboxamides may be prepared via the general method outlined in Scheme 5.
  • the appropriately substituted tetralone can be converted to the l,3,4,5-tetrahydro-l-benzazepin-2-one via either an acid- mediated Schmidt reaction with sodium azide, or Beckmann rearrangement of the corresponding ketoximes formed from reaction with hydroxylamine.
  • the 1,3,4,5- tetrahydro-l-benzazepin-2-one can then be converted to the a-iodobenzlactam by iodotrimethylsilane-mediate iodination, subsequently converted to the a-azidobenzlactam with sodium azide, and following a Staudinger reduction with triphenylphosphine yields the a-aminobenzlactam.
  • a racemization/resolution of the a-aminobenzlactam can be accomplished using L-pyroglutamic acid and 5-nirosalicylaldehyde to yield the (S)-3- amino-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one as described by Armstrong et. al. in Tetrahedron Letters 1994, pages 3239-42.
  • This amine can then be coupled to the appropriate acid using an amide coupling agent.
  • the amine can protected with a Boc protecting group, then methylated at the lactam nitrogen with methyl iodide, followed by deprotected using acidic conditions. The resulting free amine can be coupled to the appropriate acid using an amide coupling agent.
  • 5-Substituted-4H-l,2,4-triazole-3-carboxylic acids may be prepared via the general method outlined in Scheme 6.
  • the appropriately substituted acetohydrazide is condensed with ethyl 2-ethoxy-2-iminoacetate in ethanol.
  • the resulting ethyl 2-amino-2-(2- substituted hydrazono)acetate is then heated neat or in a high boiling solvent such as xylenes resulting in cyclization to the 5-substituted-4H-l,2,4-triazole-3-carboxylic ethyl ester.
  • This can then by hydrolyzed using aqueous base, for example lithium hydroxide in THF and water.
  • Scheme 6 xylene or neat
  • 5-Benzyl-lH-pyrazole-3-carboxylic acids can be prepared following the route shown in Scheme 7.
  • the appropriately substituted benzyl methyl ketone is condensed with diethyl oxalate in the presence of a base such as potassium tert-butoxide in ethanol.
  • the resulting ethyl 5-(substituted-phenyl)-2,4-dioxopentanoate is then condensed with hydrazine in ethanol resulting in cyclization to the ethyl 5-substituted benzyl- lH-pyrazole- 3-carboxylate.
  • This can then by hydrolyzed using aqueous base, for example lithium hydroxide in THF and water.
  • EtOH 5-Methyl-l-substituted-lH-pyrazole-3-carboxylic acids can be prepared following the route shown in Scheme 8.
  • Ethyl 3-methyl-lH-pyrazole-5-carboxylate is alkylated in the presence of a base such as potassium hydroxide with the appropriate alkylating agent, such as an alkyl or arylbromide, to give a mixture of desired ethyl 5-methyl-l-propyl-lH- pyrazole-3-carboxylate and undesired regioisomer ethyl 3 -methyl- 1 -substituted- 1H- pyrazole-5-carboxylate ethyl oxalate.
  • a base such as potassium hydroxide
  • the appropriate alkylating agent such as an alkyl or arylbromide
  • the ethyl 5-methyl-l-substituted-lH-pyrazole-3- carboxylates can be isolated using chromatography and then hydrolyzed using aqueous base, for example lithium hydroxide in THF and water to yield 5-methyl-l-propyl-lH- pyrazole-3-carboxylic acids.
  • the compounds of this invention may be particularly useful for the treatment of RIPl kinase-mediated diseases or disorders.
  • RIPl kinase-mediated diseases or disorders are diseases or disorders that are mediated by activation of RIPl kinase, and as such, are diseases or disorders where inhibition of RIPl kinase would provide benefit.
  • the compounds of this invention may be particularly useful for the treatment of
  • diseases/disorders which are likely to be regulated at least in part by programmed necrosis, particularly inflammatory bowel disease (including Crohn's disease and ulcerative colitis), psoriasis, retinal detachment, retinitis pigmentosa, macular degeneration, pancreatitis, atopic dermatitis, arthritis (including rheumatoid arthritis, spondylarthritis, gout, SoJIA), systemic lupus erythematosus (SLE), Sjogren's syndrome, systemic scleroderma, anti- phospholipid syndrome (APS), vasculitis, osteoarthritis, liver damage/diseases (non-alcohol steatohepatitis, alcohol steatohepatitis, autoimmune hepatitis autoimmune hepatobiliary diseases, primary sclerosing cholangitis (PSC)), nephritis, Celiac disease, autoimmune ITP
  • the compounds of this invention may be particularly useful for the treatment of diseases/disorders which are likely to be regulated at least in part by programmed necrosis, apoptosis or the production of inflammatory cytokines, particularly inflammatory bowel disease (including Crohn's disease and ulcerative colitis), psoriasis, retinal detachment, retinitis pigmentosa, macular degeneration, pancreatitis, atopic dermatitis, arthritis
  • rheumatoid arthritis including rheumatoid arthritis, spondyloarthritis, gout, systemic onset juvenile idiopathic arthritis (SoJIA), psoriatic arthritis), systemic lupus erythematosus (SLE), Sjogren's syndrome, systemic scleroderma, anti-phospholipid syndrome (APS), vasculitis, osteoarthritis, liver damage/diseases (non-alcohol steatohepatitis, alcohol steatohepatitis, autoimmune hepatitis, autoimmune hepatobiliary diseases, primary sclerosing cholangitis (PSC), acetaminophen toxicity, hepatotoxicity), kidney damage/injury (nephritis, renal transplant, surgery, administration of nephrotoxic drugs e.g. cisplatin, acute kidney injury(AKI)) Celiac disease, autoimmune idiopathic thrombocyto
  • autoimmune ITP autoimmune ITP
  • transplant rejection transplant rejection
  • ischemia reperfusion injury of solid organs sepsis
  • systemic inflammatory response syndrome SIRS
  • cerebrovascular accident CVA, stroke
  • myocardial infarction MI
  • atherosclerosis Huntington's disease, Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis (ALS), allergic diseases (including asthma and atopic dermatitis), multiple sclerosis, type I diabetes, Wegener's
  • ICE interleukin-1 converting enzyme
  • COPD chronic obstructive pulmonary disease
  • TRAPS tumor necrosis factor receptor-associated periodic syndrome
  • NEMO-deficiency syndrome F-kappa-B essential modulator gene (also known as IKK gamma or IKKG) deficiency syndrome)
  • HOIL-1 deficiency (also known as RBCKl) heme-oxidized IRP2 ubiquitin ligase-1 deficiency), linear ubiquitin chain assembly complex (LUBAC) deficiency syndrome
  • hematological and solid organ malignancies bacterial infections and viral infections (such as tuberculosis and influenza), and Lysosomal storage diseases (particularly, Gaucher Disease, and including GM2
  • Gangliosidosis Alpha-mannosidosis, Aspartylglucosaminuria, Cholesteryl Ester storage disease, Chronic Hexosaminidase A Deficiency, Cystinosis, Danon disease, Fabry disease, Farber disease, Fucosidosis, Galactosialidosis, GM1 gangliosidosis, Mucolipidosis, Infantile Free Sialic Acid Storage Disease, Juvenile Hexosaminidase A Deficiency, Krabbe disease, Lysosomal acid lipase deficiency, Metachromatic Leukodystrophy,
  • Mucopolysaccharidoses disorders Multiple sulfatase deficiency, Niemann-Pick Disease, Neuronal Ceroid Lipofuscinoses, Pompe disease, Pycnodysostosis, Sandhoff disease, Schindler disease, Sialic Acid Storage Disease, Tay-Sachs and Wolman disease).
  • the treatment of the above-noted diseases/disorders may concern, more specifically, the amelioration of organ injury or damage sustained as a result of the noted diseases.
  • the compounds of this invention may be particularly useful for amelioration of brain tissue injury or damage following ischemic brain injury or traumatic brain injury, or for amelioration of heart tissue injury or damage following myocardial infarction, or for amelioration of brain tissue injury or damage associated with
  • the treatment of diseases/disorders selected from those described herein may concern, more specifically, the amelioration of liver tissue injury or damage associated with overdose of acetaminophen, or for amelioration of kidney tissue injury or damage following renal transplant or the administration of nephrotoxic drugs or substances e.g. cisplatin.
  • the compounds of this invention may be particularly useful for the treatment of inflammatory bowel disease (including Crohn's disease and ulcerative colitis), psoriasis, retinal detachment, retinitis pigmentosa, arthritis (including rheumatoid arthritis, spondylarthritis, gout, and SoJIA), transplant rejection, ischemia reperfusion injury of solid organs, multiple sclerosis, and/or tumor necrosis factor receptor-associated periodic syndrome.
  • inflammatory bowel disease including Crohn's disease and ulcerative colitis
  • psoriasis retinal detachment
  • retinitis pigmentosa retinitis pigmentosa
  • arthritis including rheumatoid arthritis, spondylarthritis, gout, and SoJIA
  • transplant rejection transplant rejection
  • ischemia reperfusion injury of solid organs multiple sclerosis
  • multiple sclerosis multiple sclerosis
  • tumor necrosis factor receptor-associated periodic syndrome including
  • the compounds of this invention may be particularly useful for the treatment of inflammatory bowel disease (including Crohn's disease and ulcerative colitis), psoriasis, retinal detachment, retinitis pigmentosa, arthritis (including rheumatoid arthritis, spondyloarthritis, gout, and systemic onset juvenile idiopathic arthritis
  • inflammatory bowel disease including Crohn's disease and ulcerative colitis
  • psoriasis retinal detachment
  • retinitis pigmentosa including arthritis, spondyloarthritis, gout, and systemic onset juvenile idiopathic arthritis
  • SoJIA SoJIA
  • Treatment of RIP 1 -mediated disease conditions may be achieved using a compound of this invention as a monotherapy, or in dual or multiple combination therapy, particularly for the treatment of refractory cases, such as in combination with other anti-inflammatory and/or anti-TNF agents, which may be administered in therapeutically effective amounts as is known in the art.
  • Combination therapies according to the present invention thus comprise the administration of at least one compound of any one of Formulas (I-IV) or a pharmaceutically acceptable salt thereof, and at least one other theraputically active agent.
  • combination therapies according to the present invention comprise the administration of at least one compound of any one of Formulas (I-IV) or a pharmaceutically acceptable salt thereof, and at least one other therapeutically active agent.
  • the compound(s) of any one of Formulas (I-IV) and pharmaceutically acceptable salts thereof, and the other therapeutically active agent(s) may be administered together in a single pharmaceutical composition or separately and, when administered separately this may occur simultaneously or sequentially in any order.
  • a combination comprising a compound of any one of Formulas (I-IV) or a pharmaceutically acceptable salt thereof, together with one or more other therapeutically active agents.
  • a combination comprising (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide, or a
  • a compound of any one of Formulas (I-IV) or a pharmaceutically acceptable salt thereof, and a pharmaceutical composition comprising a compound of any one of Formulas (I-IV) or a pharmaceutically acceptable salt thereof may be used in combination with or include one or more other therapeutic agents, for example an anti-inflammatory agent and/or an anti-TNF agent.
  • the compounds of this invention may be administered in combination with other anti-inflammatory agents for any of the indications above, including oral or topical corticosteroids (such as prednisone (Deltasone®) and bundesonide), anti-TNF agents (including anti-TNF biologic agents), 5-aminosalicyclic acid and mesalamine preparations, hydroxycloroquine, thiopurines (azathioprin, mercaptopurin ), methotrexate, cyclophosphamide, cyclosporine, JAK inhibitors (tofacitinib), anti-IL6 biologies, anti-ILl or IL 12 or IL23 biologics(ustekinumab(Stelara®)), anti-integrin agents(natalizumab
  • corticosteroids such as prednisone (Deltasone®) and bundesonide
  • anti-TNF agents including anti-TNF biologic agents
  • 5-aminosalicyclic acid and mesalamine preparations
  • the compounds of this invention may be administered in combination with other anti-inflammatory agents for any of the indications above, including oral or topical corticosteroids (such as prednisone (Deltasone®) and bundesonide), anti-TNF agents, and other anti-inflammatory agents for any of the indications above, including oral or topical corticosteroids (such as prednisone (Deltasone®) and bundesonide), anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents, anti-TNF agents
  • cyclophosphamide cyclosporine, calcineurin inhibitors (cyclosporine, pimecrolimus, tacrolimus), mycophenolic acid (CellCept®), mTOR inhibitors (temsirolimus, everolimus), JAK inhibitors (tofacitinib), (Xeljan®)), Syk inhibitors (fostamatinib), anti-IL6 biologies, anti-ILl (anakinra (Kineret®), canakinumab (Ilaris®), rilonacept (Arcalyst®)), anti-IL12 and IL23 biologies (ustekinumab(Stelara®)), anti-ILl 7 biologies (secukinumab), anti- CD22 (epratuzumab), anti-integrin agents(natalizumab (Tysabri®)), vedolizumab
  • anti-inflammatory biologic agents include Actemra® (anti- IL6R mAb), anti-CD20 mAbs (rituximab (Rituxan®) and ofatumumab (Arzerra®)), abatacept (Orencia®), anakinra (Kineret®), ustekinumab (Stelara®), and belimumab (Benlysta®).
  • anti-inflammatory biologic agents examples include Actemra® (tocilizumab, anti-IL6R mAb), anti-CD20 mAbs (rituximab (Rituxan®) and ofatumumab (Arzerra®)), abatacept (Orencia®), anakinra (Kineret®), Canakinumab (Ilaris®), rilonacept (Arcalyst®), secukinumab, epratuzumab, sifalimumab, ustekinumab (Stelara®), and belimumab (Benlysta®).
  • Suitable anti-TNF agents biologic agents include etanecerpt (Enbrel®), adalimumab (Humira®), infliximab (Remicade®), certolizumab (Cimzia®), and golimumab (Simponi®).
  • one embodiment of this invention is directed to a method of inhibiting RIP1 kinase comprising contacting a cell with a compound of the invention.
  • the invention is directed to a method of treating a RIP1 kinase-mediated disease or disorder (specifically, a disease or disorder recited herein) comprising
  • the invention is directed to a method of treating a RIPl kinase-mediated disease or disorder (specifically, a disease or disorder recited herein) comprising administering a therapeutically effective amount of (S)-5-benzyl-N-(5-methyl- 4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to a human in need thereof.
  • the invention is directed to a method of treating a RIPl kinase-mediated disease or disorder (specifically, a disease or disorder recited herein) comprising
  • this invention provides a compound of the invention for use in therapy.
  • This invention also provides a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a pharmaceutically acceptable salt thereof, for use in therapy.
  • This invention particularly provides a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a pharmaceutically acceptable salt thereof, for use in the treatment of a RIPl kinase-mediated disease or disorder (for example, a disease or disorder recited herein).
  • this invention provides a compound described herein, or a
  • this invention provides (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide, or a
  • This invention further provides (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4- triazole-3-carboxamide, or a pharmaceutically acceptable salt thereof, for use in therapy.
  • this invention provides a compound of the invention for use in the treatment of a RIPl kinase-mediated disease or disorder.
  • this invention provides a compound described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment of a RIPl kinase-mediated disease or disorder.
  • this invention provides a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a pharmaceutically acceptable salt thereof, for use in the treatment of diseases/disorders which are likely to be regulated at least in part by programmed necrosis, apoptosis or the production of inflammatory cytokines, particularly inflammatory bowel disease (including Crohn's disease and ulcerative colitis), psoriasis, retinal detachment, retinitis pigmentosa, macular degeneration, pancreatitis, atopic dermatitis, arthritis
  • autoimmune damage/diseases non-alcohol steatohepatitis, alcohol steatohepatitis, autoimmune hepatitis, autoimmune hepatobiliary diseases, primary sclerosing cholangitis (PSC)), nephritis, Celiac disease, autoimmune idiopathic thrombocytopenic purpura (autoimmune ITP), transplant rejection, ischemia reperfusion injury of solid organs, sepsis, systemic inflammatory response syndrome (SIRS), cerebrovascular accident (CVA, stroke), myocardial infarction (MI), atherosclerosis, Huntington's disease, Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis (ALS), allergic diseases (including asthma and atopic dermatitis), multiple sclerosis, type I diabetes, Wegener's granulomatosis, pulmonary sarcoidosis, Behcet's disease, interleukin-1 converting enzyme (ICE, also known as caspase-1)
  • EMO-deficiency syndrome F-kappa-B essential modulator gene (also known as IKK gamma or IKKG) deficiency syndrome), HOIL-1 deficiency ((also known as RBCK1) heme-oxidized IRP2 ubiquitin ligase-1 deficiency), linear ubiquitin chain assembly complex (LUBAC) deficiency syndrome, hematological and solid organ malignancies, bacterial infections and viral infections (such as tuberculosis and influenza), and Lysosomal storage diseases (particularly, Gaucher Disease, and including GM2 Gangliosidosis, Alpha- mannosidosis, Aspartylglucosaminuria, Cholesteryl Ester storage disease, Chronic
  • this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of inflammatory bowel disease. In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of Crohn's disease. In another embodiment, this invention provides a compound of Formula (I), or a
  • this invention provides a compound of Formula (I), or a
  • this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of psoriasis.
  • this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of retinal detachment.
  • this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of retinitis pigmentosa.
  • this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of arthritis.
  • this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of rheumatoid arthritis.
  • this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of spondylarthritis. In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of gout. In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of systemic onset juvenile idiopathic arthritis. In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of transplant rejection. In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of ischemia reperfusion injury of solid organs.
  • This invention specifically provides for the use of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a pharmaceutically acceptable salt thereof, as an active therapeutic substance. More specifically, this provides for the use of the compounds described herein for the treatment of a RIPl kinase-mediated disease or disorder. Accordingly, the invention provides for the use of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a pharmaceutically acceptable salt thereof, as an active therapeutic substance in the treatment of a human in need thereof with a RIPl kinase-mediated disease or disorder.
  • the invention further provides for the use of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a salt thereof, particularly a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of a RIPl kinase-mediated disease or disorder, for example the diseases and disorders recited herein.
  • the invention also provides for the use of a compound described herein, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of a RIPl kinase-mediated disease or disorder, for example the diseases and disorders recited herein.
  • the invention also provides for the use of (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b] [ 1 ,4]oxazepin-3 -yl)-4H- 1 ,2,4-triazole-3 -carboxamide, or a
  • the invention further provides for the use of (S)-5-benzyl-N-(7- chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3- carboxamide, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of a RIPl kinase-mediated disease or disorder, for example the diseases and disorders recited herein.
  • the invention provides for the use of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of a human in need thereof with a RIPl kinase-mediated disease or disorder.
  • this invention provides for the use of a compound of Formula
  • autoimmune damage/diseases non-alcohol steatohepatitis, alcohol steatohepatitis, autoimmune hepatitis, autoimmune hepatobiliary diseases, primary sclerosing cholangitis (PSC)), nephritis, Celiac disease, autoimmune idiopathic thrombocytopenic purpura (autoimmune ITP), transplant rejection, ischemia reperfusion injury of solid organs, sepsis, systemic inflammatory response syndrome (SIRS), cerebrovascular accident (CVA, stroke), myocardial infarction (MI), atherosclerosis, Huntington's disease, Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis (ALS), allergic diseases (including asthma and atopic dermatitis), multiple sclerosis, type I diabetes, Wegener's granulomatosis, pulmonary sarcoidosis, Behcet's disease, interleukin-1 converting enzyme (ICE, also known as caspase-1)
  • EMO-deficiency syndrome F-kappa-B essential modulator gene (also known as IKK gamma or IKKG) deficiency syndrome), HOIL-1 deficiency ((also known as RBCK1) heme-oxidized IRP2 ubiquitin ligase-1 deficiency), linear ubiquitin chain assembly complex (LUBAC) deficiency syndrome, hematological and solid organ malignancies, bacterial infections and viral infections (such as tuberculosis and influenza), and Lysosomal storage diseases (particularly, Gaucher Disease, and including GM2 Gangliosidosis, Alpha- mannosidosis, Aspartylglucosaminuria, Cholesteryl Ester storage disease, Chronic
  • diseases/disorders may concern, more specifically, the amelioration of organ injury or damage sustained as a result of the noted diseases.
  • this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of inflammatory bowel disease.
  • this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of Crohn's disease.
  • this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the
  • this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of psoriasis. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of retinal detachment. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of retinitis pigmentosa.
  • this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of arthritis. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of rheumatoid arthritis. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of spondyloarthritis. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of gout.
  • this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of systemic onset juvenile idiopathic arthritis. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of transplant rejection. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of ischemia reperfusion injury of solid organs.
  • a therapeutically "effective amount” is intended to mean that amount of a compound that, when administered to a patient in need of such treatment, is sufficient to effect treatment, as defined herein.
  • a therapeutically effective amount of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a pharmaceutically acceptable salt thereof is a quantity of an inventive agent that, when administered to a human in need thereof, is sufficient to modulate and/or inhibit the activity of RIP 1 kinase such that a disease condition which is mediated by that activity is reduced, alleviated or prevented.
  • the amount of a given compound that will correspond to such an amount will vary depending upon factors such as the particular compound (e.g., the potency (pIC 5 o), efficacy (EC 50 ), and the biological half-life of the particular compound), disease condition and its severity, the identity (e.g., age, size and weight) of the patient in need of treatment, but can nevertheless be routinely determined by one skilled in the art.
  • the particular compound e.g., the potency (pIC 5 o), efficacy (EC 50 ), and the biological half-life of the particular compound
  • disease condition and its severity e.g., the identity of the patient in need of treatment, but can nevertheless be routinely determined by one skilled in the art.
  • duration of treatment and the time period of administration (time period between dosages and the timing of the dosages, e.g., before/with/after meals) of the compound will vary according to the identity of the mammal in need of treatment (e.g., weight), the particular compound and its properties (e.g., pharmacokinetic properties), disease or disorder and its severity and the specific composition and method being used, but can nevertheless be determined by one of skill in the art.
  • Treating is intended to mean at least the mitigation of a disease or disorder in a patient.
  • the methods of treatment for mitigation of a disease or disorder include the use of the compounds in this invention in any conventionally acceptable manner, for example for prevention, retardation, prophylaxis, therapy or cure of a RIPl kinase-mediated disease or disorder, as described hereinabove.
  • the compounds of the invention may be administered by any suitable route of administration, including both systemic administration and topical administration.
  • Systemic administration includes oral administration, parenteral administration, transdermal administration, rectal administration, and administration by inhalation.
  • Parenteral administration refers to routes of administration other than enteral, transdermal, or by inhalation, and is typically by injection or infusion.
  • Parenteral administration includes intravenous, intramuscular, and subcutaneous injection or infusion.
  • Inhalation refers to administration into the patient's lungs whether inhaled through the mouth or through the nasal passages.
  • Topical administration includes application to the skin.
  • the compounds of the invention may be administered once or according to a dosing regimen wherein a number of doses are administered at varying intervals of time for a given period of time. For example, doses may be administered one, two, three, or four times per day. Doses may be administered until the desired therapeutic effect is achieved or indefinitely to maintain the desired therapeutic effect. Suitable dosing regimens for a compound of the invention depend on the pharmacokinetic properties of that compound, such as absorption, distribution, and half-life, which can be determined by the skilled artisan.
  • suitable dosing regimens including the duration such regimens are administered, for a compound of the invention depend on the disease or disorder being treated, the severity of the disease or disorder being treated, the age and physical condition of the patient being treated, the medical history of the patient to be treated, the nature of concurrent therapy, the desired therapeutic effect, and like factors within the knowledge and expertise of the skilled artisan. It will be further understood by such skilled artisans that suitable dosing regimens may require adjustment given an individual patient's response to the dosing regimen or over time as individual patient needs change. Total daily dosages range from 1 mg to 2000 mg, preferably, total daily dosages range from 1 mg to 250 mg.
  • the compounds of the invention will be normally, but not necessarily, formulated into a pharmaceutical composition prior to administration to a patient. Accordingly, the invention also is directed to pharmaceutical compositions comprising a compound of the invention and one or more pharmaceutically acceptable excipients.
  • a pharmaceutical composition comprising (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4- triazole-3-carboxamide (free base), and one or more pharmaceutically acceptable excipients.
  • composition comprising (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3- yl)-4H-l,2,4-triazole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipients.
  • a pharmaceutical composition comprising crystalline (S)-5-benzyl-N-(5-methyl-4-oxo- 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide (free base) having the PXRD pattern of Figure 7 and one or more pharmaceutically acceptable excipients.
  • a pharmaceutical composition comprising crystalline (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b] [ 1 ,4]oxazepin-3 -yl)-4H- 1 ,2,4-triazole-3 -carboxamide (free base) characterized by the diffraction data in Table 1, and one or more pharmaceutically acceptable excipients.
  • a pharmaceutical composition comprising (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)- 4H-l,2,4-triazole-3 -carboxamide (free base) and one or more pharmaceutically acceptable excipients.
  • composition comprising (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)- 4H-l,2,4-triazole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipients.
  • compositions of the invention may be prepared and packaged in bulk form wherein an effective amount of a compound of the invention can be extracted and then given to the patient such as with powders, syrups, and solutions for injection.
  • the pharmaceutical compositions of the invention may be prepared and packaged in unit dosage form.
  • one or more tablets or capsules may be administered.
  • a dose of the pharmaceutical composition contains at least a therapeutically effective amount of a compound of this invention (i.e., a compound of
  • the pharmaceutical compositions may contain from 1 mg to 1000 mg of a compound of this invention.
  • unit dosage forms containing from 1 mg to 1000 mg of a compound of the invention may be administered one, two, three, or four times per day, preferably one, two, or three times per day, and more preferably, one or two times per day, to effect treatment of a RIPl kinase-mediated disease or disorder.
  • compositions of the invention typically contain one compound of the invention. However, in certain embodiments, the pharmaceutical compositions of the invention contain more than one compound of the invention. In addition, the pharmaceutical compositions of the invention may optionally further comprise one or more additional pharmaceutically active compounds.
  • pharmaceutically acceptable excipient means a material, composition or vehicle involved in giving form or consistency to the composition.
  • Each excipient must be compatible with the other ingredients of the pharmaceutical composition when commingled such that interactions which would substantially reduce the efficacy of the compound of the invention when administered to a patient and interactions which would result in pharmaceutical compositions that are not pharmaceutically acceptable are avoided.
  • each excipient must of course be of sufficiently high purity to render it pharmaceutically acceptable.
  • the compounds of the invention and the pharmaceutically acceptable excipient or excipients will typically be formulated into a dosage form adapted for administration to the patient by the desired route of administration.
  • Conventional dosage forms include those adapted for (1) oral administration such as tablets, capsules, caplets, pills, troches, powders, syrups, elixirs, suspensions, solutions, emulsions, sachets, and cachets; (2) parenteral administration such as sterile solutions, suspensions, and powders for reconstitution; (3) transdermal administration such as transdermal patches; (4) rectal administration such as suppositories; (5) inhalation such as aerosols and solutions; and (6) topical administration such as creams, ointments, lotions, solutions, pastes, sprays, foams, and gels.
  • oral administration such as tablets, capsules, caplets, pills, troches, powders, syrups, elixirs, suspensions, solutions, emulsions, sachets, and cachets
  • parenteral administration such as sterile solutions, suspensions, and powders for reconstitution
  • transdermal administration such as transdermal patches
  • rectal administration such as s
  • Suitable pharmaceutically acceptable excipients will vary depending upon the particular dosage form chosen.
  • suitable pharmaceutically acceptable excipients may be chosen for a particular function that they may serve in the composition.
  • certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the production of uniform dosage forms.
  • Certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the production of stable dosage forms.
  • Certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the carrying or transporting the compound or compounds of the invention once administered to the patient from one organ, or portion of the body, to another organ, or portion of the body.
  • Certain pharmaceutically acceptable excipients may be chosen for their ability to enhance patient compliance.
  • Suitable pharmaceutically acceptable excipients include the following types of excipients: diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweeteners, flavoring agents, flavor masking agents, coloring agents, anti-caking agents, humectants, chelating agents, plasticizers, viscosity increasing agents, antioxidants, preservatives, stabilizers, surfactants, and buffering agents.
  • excipients may serve more than one function and may serve alternative functions depending on how much of the excipient is present in the formulation and what other ingredients are present in the formulation.
  • Skilled artisans possess the knowledge and skill in the art to enable them to select suitable pharmaceutically acceptable excipients in appropriate amounts for use in the invention.
  • resources that are available to the skilled artisan which describe pharmaceutically acceptable excipients and may be useful in selecting suitable pharmaceutically acceptable excipients. Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), The Handbook of Pharmaceutical Additives (Gower Publishing Limited), and The Handbook of Pharmaceutical Excipients (the American Pharmaceutical Association and the Pharmaceutical Press).
  • compositions of the invention are prepared using techniques and methods known to those skilled in the art. Some of the methods commonly used in the art are described in Remington's Pharmaceutical Sciences (Mack Publishing Company). Accordingly, another embodiment of this invention is a method of preparing a
  • composition comprising the step of admixing crystalline (S)-5-benzyl-N- (5-methyl-4-oxo-2,3 ,4,5-tetrahydrobenzo[b] [ 1 ,4]oxazepin-3 -yl)-4H- 1 ,2,4-triazole-3 - carboxamide (free base) having the PXRD pattern of Figure 7 with one or more pharmaceutically acceptable excipients.
  • a method of preparing a pharmaceutical composition comprising the step of admixing crystalline (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)- 4H-l,2,4-triazole-3 -carboxamide (free base) characterized by the diffraction data in Table 1, with one or more pharmaceutically acceptable excipients.
  • the invention is directed to a solid oral dosage form such as a tablet or capsule comprising an effective amount of a compound of the invention and a diluent or filler.
  • Suitable diluents and fillers include lactose, sucrose, dextrose, mannitol, sorbitol, starch (e.g. corn starch, potato starch, and pre-gelatinized starch), cellulose and its derivatives (e.g. microcrystalline cellulose), calcium sulfate, and dibasic calcium
  • the oral solid dosage form may further comprise a binder.
  • Suitable binders include starch (e.g. corn starch, potato starch, and pre-gelatinized starch), gelatin, acacia, sodium alginate, alginic acid, tragacanth, guar gum, povidone, and cellulose and its derivatives (e.g. microcrystalline cellulose).
  • the oral solid dosage form may further comprise a disintegrant. Suitable disintegrants include crospovidone, sodium starch glycolate, croscarmelose, alginic acid, and sodium carboxymethyl cellulose.
  • the oral solid dosage form may further comprise a lubricant. Suitable lubricants include stearic acid, magnesium stearate, calcium stearate, and talc.
  • the reaction mixture was partitioned between ethyl acetate (1000 mL) and 0.5 M HC1 solution (1000 mL). The layers were separated, the organic layer was washed with water (3 x 400 ml), brine (400 mL), and concentrated under reduced pressure to provide the crude product.
  • the crude compound was purified by silica gel column using 0-10% MeOH in DCM to afford (S)-2-((tert-butoxycarbonyl)amino)-3-(2- nitrophenoxy)propanoic acid (32 g, 76 mmol, 62.3 % yield) as redish yellow semi solid.
  • Step 1 To a solution of 3-iodo-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one (16.8 g, 58.5 mmol) in ⁇ , ⁇ -Dimethylformamide (DMF) (100 mL) was added sodium azide (4.57 g, 70.2 mmol) (mild exotherm) and mixture was stirred at rt for 1 hour. A precipitate formed after 30 minutes. Ice was added to the reaction and it was then diluted with 300 mL water. More solid precipitated out and the mixture was stirred for 10 minutes. Filtered tan solid, rinsed with water and used as-is in next step (was not dried because next step contained water). Small amount dried for HNMR analysis.
  • DMF ⁇ , ⁇ -Dimethylformamide
  • Step 2 To a solution of 3-azido-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one in THF (120 mL) was added 1.0 mL water and PPh 3 resin (21.5 g, 3 mmol/g loading, 1.1 eq, 64.4 mmol, Aldrich). Stirred at rt for 20 hours. Reaction was filtered to remove resin, rinsed with THF and filtrate was concentrated. Triturated solid in 10% DCM/diethyl ether, filtered and dried to give a tan solid (9.13 g, 85% yield over 2 steps). 1H NMR (DMSO-d 6 ) ⁇ : 9.68 (br.
  • Step 1 To a mixture of (S)-3-amino-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one (0.615 g, 3.49 mmol) in DCM (20 mL) was added TEA (0.730 mL, 5.24 mmol) and BOC 2 0 (0.851 mL, 3.66 mmol). The reaction was stirred at rt for 1 hour, diluted with water and layers were separated. The organics were concentrated and dried to give 950 mg of (S)-tert-butyl (2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate as an off- white solid.
  • Step 2 To a mixture of cesium carbonate (1.592 g, 4.89 mmol) and (S)-tert-butyl (2-oxo-2,3, 4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate (950 mg, 3.40 mmol) in N,N- Dimethylformamide (DMF) (10 mL) was added iodomethane (0.262 mL, 4.19 mmol). The reaction was stirred at rt for 20 hours, then water (30 mL) was added and mixture was stirred vigorously for 15 minutes.
  • DMF N,N- Dimethylformamide
  • Step 3 To a solution of (S)-tert-butyl (l-methyl-2-oxo-2,3,4,5-tetrahydro-lH- benzo[b]azepin-3-yl)carbamate (800 mg, 2.73 mmol) in DCM (20 mL) was added HC1 (4.0 M in dioxane) (4.0 mL, 16.00 mmol). The mixture was stirred at rt for 1.5 hours, then concentrated and dried to give 670 mg of (S)-3-amino-l-methyl-4,5-dihydro-lH- benzo[b]azepin-2(3H)-one, HC1 salt as a tan solid.
  • Step 1 To a solution of sodium acetate (7.47 g, 91 mmol) in Water (13.33 mL) was added hydroxylamine hydrochloride (6.33 g, 91 mmol), then Ethanol (40 mL) and 7- bromo-3,4-dihydronaphthalen-l(2H)-one (10.25 g, 45.5 mmol). The white slurry was heated at 80°C for 45 minutes. Reaction was removed from heat, stirred for 10 minutes, then poured over ice and stirred until all ice melted. Filtered resulting solid, rinsed with water and dried to give a white solid (10.58 g, 95%).
  • DPP A diphenylphosphorylazide
  • TEA 0.091 mL, 0.649 mmol
  • tBuOH 0.439 mL, 4.59 mmol
  • Jert-butyl (7-bromo-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate 200 mg, 0.563 mmol
  • zinc cyanide 99 mg, 0.845 mmol
  • Pd 2 dba 3 258 mg, 0.282 mmol
  • S-Phos 277 mg, 0.676 mmol
  • Ethyl 5-benzoylisoxazole-3-carboxylate 400 mg, 1.631 mmol was dissolved in 5mL MeOH, and then NaBH 4 (93 mg, 2.447 mmol) was added at 0 °C. The reaction mixture was maintained at rt for 16 h. The mixture was concentrated, and then partitioned between sat. NaHC0 3 (aq) and DCM. The organic layer was concentrated and dissolved in lmL THF. An aqueous solution of LiOH (1.2 mL, 50 mg/ml solution) was added to this THF solution. The mixture was maintained at rt for 16 h.

Abstract

Disclosed are compounds having the formula (I) wherein X, Y, Z1, Z2, Z3, Z4, R5, RA, m, A. L, and B are as defined herein, and methods of making and using the same.

Description

HETEROCYCLIC AMIDES AS KINASE INHIBITORS
Field of the Invention
The present invention relates to heterocyclic amides that inhibit RIP1 kinase and methods of making and using the same.
Background of the Invention
Receptor-interacting protein- 1 (RIP1) kinase, originally referred to as RIP, is a TKL family serine/threonine protein kinase involved in innate immune signaling. RIPl kinase is a RHIM domain containing protein, with an N-terminal kinase domain and a C-terminal death domain ((2005) Trends Biochem. Sci. 30, 151-159). The death domain of RIPl mediates interaction with other death domain containing proteins including Fas and TNFR- 1 ((1995) Cell 81 513-523), TRAIL-Rl and TRAIL-R2 ((1997) Immunity 7, 821-830) and TRADD ((1996) Immunity 4, 387-396), while the RHIM domain is crucial for binding other RHFM domain containing proteins such as TRIF ((2004) Nat Immunol. 5, 503-507), DAI ((2009) EMBO Rep. 10, 916-922) and RIP3 ((1999) J. Biol. Chem. 274, 16871- 16875); (1999) Curr. Biol. 9, 539-542) and exerts many of its effects through these interactions. RIPl is a central regulator of cell signaling, and is involved in mediating both pro-survival and programmed cell death pathways which will be discussed below.
The role for RIPl in cell signaling has been assessed under various conditions
[including TLR3 ((2004) Nat Immunol. 5, 503-507), TLR4 ((2005) J. Biol. Chem. 280,
36560-36566), TRAIL ((2012) J .Virol. Epub, ahead of print), FAS ((2004) J. Biol. Chem. 279, 7925-7933)], but is best understood in the context of mediating signals downstream of the death receptor TNFRl ((2003) Cell 114, 181-190). Engagement of the TNFR by TNF leads to its oligomerization, and the recruitment of multiple proteins, including linear K63- linked polyubiquitinated RIPl ((2006) Mol. Cell 22, 245-257), TRAF2/5 ((2010) J. Mol. Biol. 396, 528-539), TRADD ((2008) Nat. Immunol. 9, 1037-1046) and cIAPs ((2008) Proc. Natl. Acad. Sci. USA. 105, 1 1778-11783), to the cytoplasmic tail of the receptor. This complex which is dependent on RIPl as a scaffolding protein (i.e. kinase
independent), termed complex I, provides a platform for pro-survival signaling through the activation of the NFKB and MAP kinases pathways ((2010) Sci. Signal. 115, re4).
Alternatively, binding of TNF to its receptor under conditions promoting the
deubiquitination of RIPl (by proteins such as A20 and CYLD or inhibition of the cIAPs) results in receptor internalization and the formation of complex II or DISC (death-inducing signaling complex) ((2011) Cell Death Dis. 2, e230). Formation of the DISC, which contains RIPl, TRADD, FADD and caspase 8, results in the activation of caspase 8 and the onset of programmed apoptotic cell death also in a RIPl kinase independent fashion ((2012) FEBS J 278, 877-887). Apoptosis is largely a quiescent form of cell death, and is involved in routine processes such as development and cellular homeostasis.
Under conditions where the DISC forms and RJP3 is expressed, but apoptosis is inhibited (such as FADD/caspase 8 deletion, caspase inhibition or viral infection), a third RIPl kinase-dependent possibility exists. RIP3 can now enter this complex, become phosphorylated by RIPl and initiate a caspase-independent programmed necrotic cell death through the activation of MLKL and PGAM5 ((2012) Cell 148, 213-227); ((2012) Cell 148, 228-243); ((2012) Proc. Natl. Acad. Sci. USA. 109, 5322-5327). As opposed to apoptosis, programmed necrosis (not to be confused with passive necrosis which is not programmed) results in the release of danger associated molecular patterns (DAMPs) from the cell.
These DAMPs are capable of providing a "danger signal" to surrounding cells and tissues, eliciting proinflammatory responses including inflammasome activation, cytokine production and cellular recruitment ((2008 Nat. Rev. Immunol 8, 279-289).
Dysregulation of RIPl kinase-mediated programmed cell death has been linked to various inflammatory diseases, as demonstrated by use of the RIP3 knockout mouse (where RIPl -mediated programmed necrosis is completely blocked) and by Necrostatin-1 (a tool inhibitor of RIPl kinase activity with poor oral bioavailability). The RIP3 knockout mouse has been shown to be protective in inflammatory bowel disease (including Ulcerative colitis and Crohn's disease) ((2011) Nature 477, 330-334), Psoriasis ((2011) Immunity 35, 572- 582), retinal-detachment-induced photoreceptor necrosis ((2010) PNAS 107, 21695- 21700), retinitis pigmentosa ((2012) Proc. Natl. Acad. Sci., 109:36, 14598-14603), cerulein-induced acute pancreatits ((2009) Cell 137, 1100-1111) and Sepsis/systemic inflammatory response syndrome (SIRS) ((2011) Immunity 35, 908-918). Necrostatin-1 has been shown to be effective in alleviating ischemic brain injury ((2005) Nat. Chem. Biol. 1, 112-119), retinal ischemia/reperfusion injury ((2010) J. Neurosci. Res. 88, 1569- 1576), Huntington's disease ((2011) Cell Death Dis. 2 el 15), renal ischemia reperfusion injury ((2012) Kidney Int. 81, 751-761), cisplatin induced kidney injury ((2012) Ren. Fail. 34, 373-377) and traumatic brain injury ((2012) Neurochem. Res. 37, 1849-1858). Other diseases or disorders regulated at least in part by RIPl -dependent apoptosis, necrosis or cytokine production include hematological and solid organ malignancies ((2013) Genes Dev. 27: 1640-1649), bacterial infections and viral infections ((2014) Cell Host & Microbe 15, 23-35) (including, but not limited to, tuberculosis and influenza ((2013) Cell 153, 1- 14)) and Lysosomal storage diseases (particularly, Gaucher Disease, Nature Medicine Advance Online Publication, 19 January 2014, doi: 10.1038/nm.3449).
A potent, selective, small molecule inhibitor of RIP1 kinase activity would block RIP 1 -dependent cellular necrosis and thereby provide a therapeutic benefit in diseases or events associated with DAMPs, cell death, and/or inflammation.
SUMMARY OF THE INVENTION
The invention is directed to compounds according to Formula (I):
Figure imgf000005_0001
wherein:
X is O, S, SO, S02, NH, CO, CH2, CF2, CH(CH3), CH(OH), or N(CH3);
Y is CH2 or CH2CH2;
Z1 is N, CH or CR1;
Z2 is CH or CR2;
Z3 is N, CH or CR3;
Z4 is CH or CR4;
R1 is fluoro or methyl;
one of R2 and R3 is halogen, cyano, (Ci-C6)alkyl, halo(Ci-C4)alkyl,
(Ci-Ce)alkoxy, halo(Ci-C4)alkoxy, hydroxyl, B(OH)2, -COOH, halo(Ci-C4)alkylC(OH) (Ci-C4)alkoxy(Ci-C4)alkoxy, (Ci-C4)alkylS02-, (Ci-C4)alkylS02NHC(0)-,
(Ci-C4)alkylC(0)NH-, ((Ci-C4)alkyl)((Ci-C4)alkyl)NC(0)-, (Ci-C4)alkylOC(0)-, (Ci-C4)alkylC(0)N(Ci-C4)alkyl)-, (Ci-C4)alkylNHC(0)-,
(Ci-C4)alkoxy(C2-C4)alkylNHC(0)-, (Ci-C4)alkoxy(C2-C4)alkylC(0)NH-,
(Ci-C4)alkoxy(C2-C4)alkylNHC(0)NH-, (Ci-C4)alkylS02(C2-C4)alkylNHC(0)-,
(Ci-C4)alkylNHC(0)NH-, (Ci-C4)alkylOC(0)NH-, hydroxy(Ci-C4)alkylOC(0)NH-, 5- membered heterocycloalkyl-C(O)-, 5-6 membered heterocycloalkyl-(Ci-C4)alkyl-NHC(0)-, 5-6 membered heterocycloalkyl-(Ci-C4)alkoxy-, 3-6 membered cycloalkyl, 5-6 membered heteroaryl, or 5-6 membered heteroaiyl-C(0)NH, wherein said 3-6 membered cycloalkyl, 5-6 membered heterocycloalkyl and 5-6 membered heteroaryl are optionally substituted by 1 or 2 substituents each independently selected from the group consisting of (Ci-C4)alkyl and -(Ci-C4)alkyl-CN;
and the other of R2 and R3 is halogen or (Ci-C6)alkyl;
R4 is fluoro, chloro, or methyl;
R5 is H or methyl;
A is phenyl, 5-6 membered heteroaryl, or 5-6 membered
heterocycloalkyl, wherein the carbonyl moiety and L are substituted 1,3 on ring
A;
m is 0 or m is 1 and RA is (Ci-C4)alkyl; and
L is O, S, NH, N(CH3), CH2, CH2CH2, CH(CH3), CHF, CF2, CH20,
CH2N(CH3), CH2NH, or CH(OH);
B is an optionally substituted (C3-C6)cycloalkyl, phenyl, 5-6 membered
heteroaryl, or 5-6 membered heterocycloalkyl;
wherein said (C3-C6)cycloalkyl, phenyl, 5-6 membered heteroaryl, or 5-6 membered heterocycloalkyl is unsubstituted or is substituted by one or two
substituents each independently selected from halogen, (Ci-C4)alkyl,
halo(Ci-C4)alkyl, (Ci-C4)alkoxy, halo(Ci-C4)alkoxy, nitro, and
(Ci-C4)alkylC(0)-;
or the moiety -L-B is (C3-C6)alkyl, (C3-C6)alkoxy, halo(C3-C6)alkoxy,
(C3-C6)alkenyl, or (C3-C6)alkenyloxy;
or a salt, particularly a pharmaceutically acceptable salt, thereof.
The compounds according to Formula (I), or salts, particularly pharmaceutically acceptable salts, thereof, are inhibitors of RIPl kinase.
Accordingly, the present invention is also directed to a method of inhibiting RIPl kinase which method comprises contacting a cell with a compound according to
Formula (I), or a salt, particularly a pharmaceutically acceptable salt, thereof.
The invention is further directed to a method of treating a RIPl kinase-mediated disease or disorder which comprises administering a therapeutically effective amount of a compound according to Formula (I), or a salt, particularly a pharmaceutically acceptable salt thereof, to a patient (a human or other mammal, particularly, a human) in need thereof. Such RIP1 kinase-mediated diseases or disorders include inflammatory bowel disease (including Crohn's disease and ulcerative colitis), psoriasis, retinal detachment, retinitis pigmentosa, arthritis (including rheumatoid arthritis, spondyloarthritis, gout, and SoJIA), transplant rejection, ischemia reperfusion injury of solid organs, multiple sclerosis, and tumor necrosis factor receptor-associated periodic syndrome.
The present invention is further directed to a pharmaceutical composition comprising a compound according to Formula (I), or a salt, particularly a pharmaceutically acceptable salt, thereof and a pharmaceutically acceptable excipient. Particularly, this invention is directed to a pharmaceutical composition for the treatment of a RIP1 kinase-mediated disease or disorder, where the composition comprises a compound according to Formula (I), or a salt, particularly a pharmaceutically acceptable salt, thereof and a pharmaceutically acceptable excipient.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 A shows the temperature loss over time in mice after oral pre-dosing with the compound of Example 12 or vehicle followed by simultaneous i.v. administration of mouse TNF and zVAD.
Figure IB shows the temperature loss in mice 2.5 hours after oral pre-dosing with the compound of Example 12 or vehicle followed by simultaneous i.v. administration of mouse TNF and zVAD.
Figure 2A shows the temperature loss over time in mice after oral pre-dosing with the compound of Example 12 or vehicle followed by i.v. administration of mouse TNF.
Figure 2B shows the temperature loss in mice 6 hours after oral pre-dosing with the compound of Example 12 or vehicle followed by i.v. administration of mouse TNF.
Figure 3 A shows the cellular levels of ATP in mouse L929 fibrosarcoma cells pre- treated with the compound of Example 77 followed by treatment with TNFa + QvD.
Figure 3B shows the cellular levels of ATP in human monocytic leukemia U937 fibrosarcoma cells pre-treated with the compound of Example 77 followed by treatment with TNFa + QvD.
Figure 4 A shows the temperature loss over time in mice after oral pre-dosing with the compound of Example 161 or vehicle followed by simultaneous i.v. administration of mouse TNF and zVAD. Figure 4B shows the temperature loss in mice 6 hours after oral pre-dosing with the compound of Example 161 or vehicle followed by simultaneous i.v. administration of mouse TNF and zVAD.
Figure 5 A shows the temperature loss over time in mice after oral pre-dosing with the compound of Example 161 or vehicle followed by i.v. administration of mouse TNF.
Figure 5B shows the temperature loss in mice 6 hours after oral pre-dosing with the compound of Example 161 or vehicle followed by i.v. administration of mouse TNF.
Figure 6A shows the cellular levels of ATP in mouse L929 fibrosarcoma cells pre- treated with the compound of Example 161 followed by treatment with TNFa + QvD.
Figure 6B shows an IC50 curve of ATP in human monocytic leukemia U937 fibrosarcoma cells pre-treated with the compound of Example 161 followed by treatment with TNFa + QvD. Data are normalized to lOmM Nec-1 which was set at 100% survival.
Figure 7 is a powder x-ray diffraction (PXRD) pattern of a crystalline form of anhydrous (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)- 4H-l,2,4-triazole-3-carboxamide (free base).
DETAILED DESCRIPTION OF THE INVENTION
This invention is also directed to a compound of Formula (I) wherein:
X is O, S, SO, S02, NH, CO, CH2, CF2, CH(CH3), CH(OH), or N(CH3);
Y is CH2 or CH2CH2;
Z1 is N, CH or CR1;
Z2 is CH or CR2;
Z3 is N, CH or CR3;
Z4 is CH or CR4;
R1 is fluoro or methyl;
one of R2 and R3 is halogen, cyano, (Ci-C6)alkyl, halo(Ci-C4)alkyl,
(Ci-C6)alkoxy, hydroxyl, B(OH)2, -COOH, halo(Ci-C4)alkylC(OH)2-,
(Ci-C4)alkoxy(Ci-C4)alkoxy, (Ci-C4)alkylS02-, (Ci-C4)alkylS02NHC(0)-,
(Ci-C4)alkylC(0)NH-, ((Ci-C4)alkyl)((Ci-C4)alkyl)NC(0)-, (Ci-C4)alkylOC(0)-, (Ci-C4)alkylC(0)N(Ci-C4)alkyl)-, (Ci-C4)alkylNHC(0)-,
(Ci-C4)alkoxy(C2-C4)alkylNHC(0)-, (Ci-C4)alkoxy(C2-C4)alkylC(0)NH-,
(Ci-C4)alkoxy(C2-C4)alkylNHC(0)NH-, (Ci-C4)alkylS02(C2-C4)alkylNHC(0)-,
(Ci-C4)alkylNHC(0)NH-, (Ci-C4)alkylOC(0)NH-, hydroxy(Ci-C4)alkylOC(0)NH-, 5-6 membered heterocycloalkyl-C(O)-, 5-6 membered
heterocycloalkyl-(Ci-C4)alkyl-NHC(0)-, 5-6 membered heterocycloalkyl-(Ci-C4)alkoxy-, 5-6 membered heteroaryl, or 5-6 membered heteroaiyl-C(0) H,
wherein said 5-6 membered heterocycloalkyl and 5-6 membered heteroaryl are optionally substituted by 1 or 2 substituents each independently selected from the group consisting of (Ci-C4)alkyl and -(Ci-C4)alkyl-CN;
and the other of R2 and R3 is halogen or (Ci-C6)alkyl;
R4 is fluoro, chloro, or methyl;
R5 is H or methyl;
A is phenyl, 5-6 membered heteroaryl, or 5-6 membered
heterocycloalkyl, wherein the carbonyl moiety and L are substituted 1,3 on ring
A;
m is 0 or m is 1 and RA is (Ci-C4)alkyl; and
L is O, S, NH, N(CH3), CH2, CH2CH2, CH(CH3), CHF, CF2, CH20,
CH2N(CH3), CH2NH, or CH(OH);
B is an optionally substituted (C3-C6)cycloalkyl, phenyl, 5-6 membered
heteroaryl, or 5-6 membered heterocycloalkyl;
wherein said (C3-C6)cycloalkyl, phenyl, 5-6 membered heteroaryl, or 5-6 membered heterocycloalkyl is unsubstituted or is substituted by one or two
substituents each independently selected from halogen, (Ci-C4)alkyl,
halo(Ci-C4)alkyl, (Ci-C4)alkoxy, halo(Ci-C4)alkoxy, nitro, and
(Ci-C4)alkylC(0)-;
or the moiety -L-B is (C3-C6)alkyl, (C3-C6)alkoxy, halo(C3-C6)alkoxy,
(C3-C6)alkenyl, or (C3-C6)alkenyloxy;
or a salt, particularly a pharmaceutically acceptable salt, thereof.
The alternative definitions for the various groups and substituent groups of Formula (I) provided throughout the specification are intended to particularly describe each compound species disclosed herein, individually, as well as groups of one or more compound species. The scope of this invention includes any combination of these group and substituent group definitions. The compounds of the invention are only those which are contemplated to be "chemically stable" as will be appreciated by those skilled in the art.
As used herein, the term "alkyl" represents a saturated, straight or branched hydrocarbon group having the specified number of carbon atoms. The term "(Ci-C4)alkyl" refers to an alkyl moiety containing from 1 to 4 carbon atoms. Exemplary alkyls include, but are not limited to methyl, ethyl, ^-propyl, isopropyl, «-butyl, isobutyl, s-butyl, and t- butyl.
When a substituent term such as "alkyl" is used in combination with another substituent term, for example as in "hydroxy(Ci-C4)alkyl" or "aryl(Ci-C4)alkyl", the linking substituent term (e.g., alkyl) is intended to encompass a divalent moiety, wherein the point of attachment is through that linking substituent. Examples of "aryl(Ci-C4)alkyl" groups include, but are not limited to, benzyl (phenylmethyl), 1-methylbenzyl
(1-phenylethyl), and phenethyl (2-phenylethyl). Examples of "hydroxy(Ci-C4)alkyl" groups include, but are not limited to, hydroxymethyl, hydroxyethyl, and hydroxyisopropyl.
The term "halo(Ci-C4)alkyl" represents a group having one or more halogen atoms, which may be the same or different, at one or more carbon atoms of an alkyl moiety containing from 1 to 4 carbon atoms. Examples of "halo(Ci-C4)alkyl" groups include, but are not limited to, -CF3 (trifluorom ethyl), -CC13 (trichloromethyl), 1, 1-difluoroethyl, 2,2,2- trifluoroethyl, and hexafluoroisopropyl.
"Alkenyl" refers to straight or branched hydrocarbon group having at least 1 and up to 3 carbon-carbon double bonds. Examples include ethenyl and propenyl.
"Alkoxy" refers to an "alkyl-oxy-" group, containing an alkyl moiety attached through an oxygen linking atom. For example, the term "(Ci-C4)alkoxy" represents a saturated, straight or branched hydrocarbon moiety having at least 1 and up to 4 carbon atoms attached through an oxygen linking atom. Exemplary "(Ci-C4)alkoxy" groups include, but are not limited to, methoxy, ethoxy, «-propoxy, isopropoxy, «-butoxy, s- butoxy, and t-butoxy.
The term "halo(Ci-C4)alkoxy" refers to a "haloalkyl-oxy-" group, containing a "halo(Ci-C4)alkyl" moiety attached through an oxygen linking atom, which
halo(Ci-C4)alkyl" refers to a moiety having one or more halogen atoms, which may be the same or different, at one or more carbon atoms of an alkyl moiety containing from 1 to 4 carbon atoms. Exemplary "halo(Ci-C4)alkoxy" groups include, but are not limited to, - OCFIF2 (difluoromethoxy), -OCF3 (trifluoromethoxy), -OCH2CF3 (trifluoroethoxy), and -OCH(CF3)2 (hexafluoroisopropoxy).
A carbocyclic group is a cyclic group in which all of the ring members are carbon atoms, which may be saturated, partially unsaturated (non-aromatic) or fully unsaturated (aromatic). The term "carbocyclic" includes cycloalkyl and aryl groups. "Cycloalkyl" refers to a non-aromatic, saturated, cyclic hydrocarbon group containing the specified number of carbon atoms. For example, the term
"(C3-C6)cycloalkyl" refers to a non-aromatic cyclic hydrocarbon ring having from three to six ring carbon atoms. Exemplary "(C3-C6)cycloalkyl" groups include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
The terms "cycloalkyloxy" or "cycloalkoxy" refer to a group containing a cycloalkyl moiety, defined hereinabove, attached through an oxygen linking atom.
Exemplary "(C3-C6)cycloalkyloxy" groups include cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, and cyclohexyloxy.
"Aryl" refers to a group or moiety comprising an aromatic, monocyclic or bicyclic hydrocarbon radical containing from 6 to 10 carbon ring atoms and having at least one aromatic ring. Examples of "aryl" groups are phenyl, naphthyl, indenyl, and
dihydroindenyl (indanyl). Generally, in the compounds of this invention, aryl is phenyl.
A heterocyclic group is a cyclic group having, as ring members, atoms of at least two different elements, which cyclic group may be saturated, partially unsaturated
(non-aromatic) or fully unsaturated (aromatic). The terms "heterocyclic" or "heterocyclyl" includes heterocycloalkyl and heteroaryl groups. It is to be understood that the terms heterocyclic, heterocyclyl, heteroaryl, and heterocycloalkyl, are intended to encompass stable groups where a ring nitrogen heteroatom is optionally oxidized (e.g., heteroaryl groups containing an N-oxide, such as oxo-pyridyl (pyridyl-N-oxide) and oxo-oxadiazolyl (oxo-4,5-dihydro-l,3,4-oxadiazolyl) or where a ring sulfur heteroatom is optionally oxidized (e.g., heterocycloalkyl groups containing sulfones or sulfoxide moieties, such as tetrahydrothienyl-1 -oxide (tetrahydrothienyl sulfoxide, tetrahydrothiophenyl sulfoxide) and tetrahydrothienyl- 1,1 -dioxide (tetrahydrothienyl sulfone)).
"Heterocycloalkyl" refers to a non-aromatic, monocyclic or bicyclic group containing 3-10 ring atoms, being saturated or having one or more degrees of unsaturation and containing one or more (generally one or two) heteroatom substitutions independently selected from oxygen, sulfur, and nitrogen. Examples of "heterocycloalkyl" groups include, but are not limited to, aziridinyl, thiiranyl, oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrrolinyl, pyrazolidinyl, pyrazolinyl, imidazolidinyl, imidazolinyl, oxazolinyl, thiazolinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, 1,3- dioxolanyl, piperidinyl, piperazinyl, tetrahydropyranyl, dihydropyranyl,
tetrahydrothiopyranyl, 1,3-dioxanyl, 1,4-dioxanyl, 1,3-oxathiolanyl, 1,3-oxathianyl, 1,3- dithianyl, 1,4-oxathiolanyl, 1,4-oxathianyl, 1,4-dithianyl, morpholinyl, thiomorpholinyl, hexahydro- IH- 1 ,4-diazepinyl, azabicylo[3.2.1 joctyl, azabicylo[3.3.1 Jnonyl,
azabicylo[4.3.0]nonyl, oxabicylo[2.2.1]heptyl, l, l-dioxidotetrahydro-2H-thiopyranyl, and 1 , 5 , 9-tri azacy cl ododecy 1.
Examples of "4-membered heterocycloalkyl" groups include oxetanyl, thietanyl and azetidinyl.
The term "5-6-membered heterocycloalkyl" represents a non aromatic, monocyclic group, which is saturated or partially unsaturated, containing 5 or 6 ring atoms, which includes one or two heteroatoms selected independently from oxygen, sulfur, and nitrogen. Illustrative examples of 5 to 6-membered heterocycloalkyl groups include, but are not limited to pyrrolidinyl, piperidinyl, piperazinyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, tetrahydrothiopyranyl, morpholinyl, and thiomorpholinyl.
"Heteroaryl" represents a group or moiety comprising an aromatic monocyclic or bicyclic radical, containing 5 to 10 ring atoms, including 1 to 4 heteroatoms independently selected from nitrogen, oxygen and sulfur. This term also encompasses bicyclic heterocyclic-aryl groups containing either an aryl ring moiety fused to a heterocycloalkyl ring moiety or a heteroaryl ring moiety fused to a cycloalkyl ring moiety.
Illustrative examples of heteroaryls include, but are not limited to, furanyl, thienyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiadiazolyl, isothiazolyl, pyridinyl (pyridyl), oxo-pyridyl (pyridyl-N-oxide), pyridazinyl, pyrazinyl, pyrimidinyl, triazinyl, benzofuranyl, isobenzofuryl, 2,3- dihydrobenzofuryl, 1,3-benzodioxolyl, dihydrobenzodioxinyl, benzothienyl, indolizinyl, indolyl, isoindolyl, dihydroindolyl, benzimidazolyl, dihydrobenzimidazolyl, benzoxazolyl, dihydrobenzoxazolyl, benzothiazolyl, benzoisothiazolyl, dihydrobenzoisothiazolyl, indazolyl, imidazopyridinyl, pyrazolopyridinyl, benzotriazolyl, triazolopyridinyl, purinyl, quinolinyl, tetrahydroquinolinyl, isoquinolinyl, tetrahydroisoquinolinyl, quinoxalinyl, cinnolinyl, phthalazinyl, quinazolinyl, 1,5-naphthyridinyl, 1,6-naphthyridinyl, 1,7- naphthyridinyl, 1,8-naphthyridinyl, and pteridinyl.
As used herein, "5-6-membered heteroaryl" represents an aromatic monocyclic group containing 5 or 6 ring atoms, including at least one carbon atom and 1 to 4 heteroatoms independently selected from nitrogen, oxygen and sulfur. Selected 5- membered heteroaryl groups contain one nitrogen, oxygen, or sulfur ring heteroatom, and optionally contain 1, 2, or 3 additional nitrogen ring atoms. Selected 6-membered heteroaryl groups contain 1, 2, or 3 nitrogen ring heteroatoms. Examples of 5- membered heteroaryl groups include furyl (furanyl), thienyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, thiazolyl, isothiazolyl, thiadiazolyl, oxazolyl, isoxazolyl, oxadiazolyl and oxo- oxadiazolyl. Selected 6-membered heteroaryl groups include pyridinyl, oxo-pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl and triazinyl.
Bicyclic heteroaryl groups include 6,5-fused heteroaryl (9-membered heteroaryl) and 6,6-fused heteroaryl (10-membered heteroaryl) groups. Examples of 6,5-fused heteroaryl (9-membered heteroaryl) groups include benzothienyl, benzofuranyl, indolyl, indolinyl, isoindolyl, isoindolinyl, indazolyl, indolizinyl, isobenzofuryl,
2,3-dihydrobenzofuryl, benzoxazolyl, benzthiazolyl, benzimidazolyl, benzoxadiazolyl, benzthiadiazolyl, benzotriazolyl, l,3-benzoxathiol-2-on-yl (2-oxo-l,3-benzoxathiolyl), purinyl and imidazopyridinyl.
Examples of 6,6-fused heteroaryl (10-membered heteroaryl) groups include quinolyl, isoquinolyl, phthalazinyl, naphthridinyl (1,5-naphthyridinyl, 1,6-naphthyridinyl, 1,7-naphthyridinyl, 1,8-naphthyridinyl), quinazolinyl, quinoxalinyl, 4H-quinolizinyl, tetrahydroquinolinyl, cinnolinyl, and pteridinyl.
Unless otherwise specified, all bicyclic ring systems may be attached at any suitable position on either ring.
The terms "halogen" and "halo" represent chloro, fluoro, bromo, or iodo
substituents. "Oxo" represents a double-bonded oxygen moiety; for example, if attached directly to a carbon atom forms a carbonyl moiety (C = O). "Hydroxy" or "hydroxyl" is intended to mean the radical -OH. As used herein, the term "cyano" refers to the group - CN.
As used herein, the term "optionally substituted" indicates that a group (such as an alkyl, cycloalkyl, alkoxy, heterocycloalkyl, aryl, or heteroaryl group) or ring or moiety (such as a carbocyclic or heterocyclic ring or moiety) may be unsubstituted, or the group, ring or moiety may be substituted with one or more substituent(s) as defined. In the case where groups may be selected from a number of alternative groups, the selected groups may be the same or different.
The term "independently" means that where more than one substituent is selected from a number of possible substituents, those substituents may be the same or different.
The term "pharmaceutically acceptable" refers to those compounds, materials, compositions, and dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
As used herein, the terms "compound(s) of the invention" or "compound(s) of this invention" mean a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), as defined herein, in any form, i.e., any salt or non-salt form (e.g., as a free acid or base form, or as a salt, particularly a pharmaceutically acceptable salt thereof) and any physical form thereof (e.g., including non-solid forms (e.g., liquid or semi-solid forms), and solid forms (e.g., amorphous or crystalline forms, specific polymorphic forms, solvate forms, including hydrate forms (e.g., mono-, di-and hemi- hydrates)), and mixtures of various forms.
Accordingly, included within the present invention are the compounds of Formula (I), particularly, compounds of any one of Formulas (I-IV), as defined herein, in any salt or non-salt form and any physical form thereof, and mixtures of various forms. While such are included within the present invention, it will be understood that the compounds of
Formula (I), particularly, compounds of any one of Formulas (I-IV), as defined herein, in any salt or non-salt form, and in any physical form thereof, may have varying levels of activity, different bioavailabilities and different handling properties for formulation purposes.
In one embodiment of the compounds of this invention, X is O, S, SO,
S02, H, CO, CH2, CF2, CH(CH3), N(CH3), or CH(OH). In a specific
embodiment, X is O, S, SO, S02, NH, CO, CH2, or N(CH3). In another
embodiment, X is S, SO, S02, or CO. In yet another embodiment, X is CF2,
CH(CH3), or CH(OH). In a further embodiment, X is O, CH2, NH or N(CH3).
In selected embodiments, X is O or CH2.
In one embodiment of the compounds of this invention, Y is CH2 or
CH2CH2. In another embodiment, Y is CH2CH2. In selected embodiments, Y is
CH2.
In one embodiment of the compounds this invention, Z1, Z2, Z3, and Z4
are each CH. In another embodiment, Z1 is CR1 and Z2, Z3 and Z4 are each CH.
In a further embodiment, Z1, Z2, and Z4 are each CH and Z3 is CR3. In a further embodiment, Z1, Z3, and Z4 are each CH and Z2 is CR2. In a still further
embodiment, Z1, Z2, and Z3 are each CH and Z4 is CR4. In another embodiment, Z1 and Z2 are CH, Z3 is CR3, and Z4 is CR4. In another embodiment, Z1 and Z4
are CH, Z 2 is CR 2 , and Z 3 is CR 3. In another embodiment, Z 1 and Z 3 are CH, Z 2 is CR2, and Z4 is CR4. In another embodiment, Z1 is CH, Z2 is CR2, Z3 is CR3,
and Z4 is CR4.
In yet another embodiment of the compounds of this invention Z1 and Z3 are both N, Z2 is CH and Z4 is CH or CR4. In yet another embodiment of the
compounds of this invention Z1 and Z3 are both N, Z2 is CH or CR2 and Z4 is
CH. In still other embodiments, Z1 is N, Z2 is CR2 and Z3 and Z4 are CH. In yet other embodiments, Z3 is N, and Z2, Z3 and Z4 are CH.
In one embodiment of the compounds of this invention, R1 is fluoro. In
another embodiment, R1 is methyl.
In one embodiment, one of R2 and R3 is halogen, cyano, (Ci-C6)alkyl,
halo(Ci-C4)alkyl, (Ci-C6)alkoxy, halo(d-C4)alkoxy, hydroxyl, B(OH)2, -COOH, halo(Ci-C4)alkylC(OH)2-, (Ci-C4)alkoxy(Ci-C4)alkoxy, (Ci-C4)alkylS02-,
(Ci-C4)alkylS02 HC(0)-, (Ci-C4)alkylC(0) H-, ((Ci-C4)alkyl)((Ci-C4)alkyl)NC(0)-, (Ci-C4)alkylOC(0)-, (Ci-C4)alkylC(0)N(Ci-C4)alkyl)-, (Ci-C4)alkyl HC(0)-,
(Ci-C4)alkoxy(C2-C4)alkyl HC(0)-, (Ci-C4)alkoxy(C2-C4)alkylC(0) H-,
(Ci-C4)alkoxy(C2-C4)alkyl HC(0)NH-, (Ci-C4)alkylS02(C2-C4)alkyl HC(0)-,
(Ci-C4)alkyl HC(0) H-, (Ci-C4)alkylOC(0) H-, hydroxy(Ci-C4)alkylOC(0) H-, 5-6 membered heterocycloalkyl-C(O)-, 5-6 membered
heterocycloalkyl-(Ci-C )alkyl-NHC(0)-, 5-6 membered heterocycloalkyl-(Ci-C )alkoxy-, 3-6 membered cycloalkyl, 5-6 membered heteroaryl, or 5-6 membered heteroaiyl-C(0)NH, wherein said 3-6 membered cycloalkyl, 5-6 membered heterocycloalkyl and 5-6 membered heteroaryl are optionally substituted by 1 or 2 substituents each independently selected from the group consisting of (Ci-C4)alkyl and -(Ci-C4)alkyl-CN;
and the other of R2 and R3 is halogen, cyano or (Ci-C6)alkyl.
In one embodiment of the compounds of this invention, one of R2 and R3 is halogen, cyano, (Ci-C6)alkyl, halo(Ci-C4)alkyl, (Ci-C6)alkoxy, hydroxyl, B(OH)2, -COOH, halo(Ci-C4)alkylC(OH)2-, (Ci-C4)alkoxy(Ci-C4)alkoxy, (Ci-C4)alkylS02-,
(Ci-C4)alkylS02 HC(0)-, (Ci-C4)alkylC(0) H-, ((Ci-C4)alkyl)((Ci-C4)alkyl)NC(0)-, (Ci-C4)alkylOC(0)-, (Ci-C4)alkylC(0)N(Ci-C4)alkyl)-, (Ci-C4)alkyl HC(0)-,
(Ci-C4)alkoxy(C2-C4)alkyl HC(0)-, (Ci-C4)alkoxy(C2-C4)alkylC(0) H-,
(Ci-C4)alkoxy(C2-C4)alkyl HC(0)NH-, (Ci-C4)alkylS02(C2-C4)alkyl HC(0)-, (Ci-C4)alkyl HC(0) H-, (Ci-C4)alkylOC(0) H-, hydroxy(Ci-C4)alkylOC(0) H-, 5-6 membered heterocycloalkyl-C(O)-, 5-6 membered
heterocycloalkyl-(Ci-C4)alkyl-NHC(0)-, 5-6 membered heterocycloalkyl-(Ci-C4)alkoxy-, 5-6 membered heteroaryl, or 5-6 membered heteroaiyl-C(0) H,
wherein said 5-6 membered heterocycloalkyl and 5-6 membered heteroaryl are optionally substituted by 1 or 2 substituents each independently selected from the group consisting of (Ci-C )alkyl and -(Ci-C )alkyl-CN;
and the other of R2 and R3 is halogen or (Ci-C6)alkyl.
In another embodiment, R2 is halogen, cyano, (Ci-C6)alkyl, (Ci-C6)alkoxy, halo(Ci-C4)alkoxy, hydroxyl, B(OH)2, -COOH, halo(Ci-C4)alkylC(OH)2-,
(Ci-C )alkoxy(Ci-C )alkoxy, 3-5 membered cycloalkyl, or 5-6 membered heteroaryl, wherein said 3-5 membered cycloalkyl or 5-6 membered heteroaryl is optionally substituted by a (Ci-C3)alkyl substituent; and Z3 is CH or CR3 and R3 is cyano, (Ci-C6)alkyl, or a 5-6 membered heteroaryl, optionally substituted by a (Ci-C3)alkyl substituent. In another embodiment, R2 is halogen, cyano, (Ci-C6)alkyl, hydroxyl, B(OH)2, -COOH, halo(Ci-C4)alkylC(OH)2-, (Ci-C4)alkoxy(Ci-C4)alkoxy, or 5-6 membered heteroaryl, wherein said 5-6 membered heteroaryl is optionally substituted by a (Ci-C3)alkyl substituent; and Z3 is CH.
In another embodiment, R3 is halogen, (Ci-C6)alkyl, halo(Ci-C4)alkyl,
(Ci-C6)alkoxy, halo(Ci-C6)alkoxy, B(OH)2, -COOH, (Ci-C4)alkylS02-,
(Ci-C4)alkylS02 HC(0)-, (Ci-C4)alkylC(0) H-, ((Ci-C4)alkyl)((Ci-C4)alkyl)NC(0)-, (Ci-C4)alkylOC(0)-, (Ci-C4)alkylC(0)N(Ci-C4)alkyl)-,
(Ci-C4)alkoxy(C2-C4)alkyl HC(0)NH-, (Ci-C4)alkylS02(C2-C4)alkyl HC(0)-,
(Ci-C4)alkyl HC(0) H-, (Ci-C4)alkylOC(0) H-, hydroxy(Ci-C4)alkylOC(0) H-, 5-6 membered heterocycloalkyl-C(O)-, 5-6 membered
heterocycloalkyl-(Ci-C )alkyl-NHC(0)-, 5-6 membered heterocycloalkyl-(Ci-C )alkoxy-, 5-6 membered heteroaryl, or 5-6 membered heteroaiyl-C(0) H, herein said 5-6 membered heterocycloalkyl and 5-6 membered heteroaryl are optionally substituted by (Ci-C3)alkyl or -(Ci-C3)alkyl-CN; and Z2 is CH.
In another embodiment, R3 is halogen, (Ci-C6)alkyl, halo(Ci-C )alkyl,
(Ci-C6)alkoxy, B(OH)2, -COOH, (Ci-C4)alkylS02-, (Ci-C4)alkylS02 HC(0)-,
(Ci-C4)alkylC(0) H-, ((Ci-C4)alkyl)((Ci-C4)alkyl)NC(0)-, (Ci-C4)alkylOC(0)-, (Ci-C4)alkylC(0)N(Ci-C4)alkyl)-, (Ci-C4)alkoxy(C2-C4)alkyl HC(0) H-, (Ci-C4)alkylS02(C2-C4)alkyl HC(0)-, (Ci-C4)alkyl HC(0) H-,
(Ci-C4)alkylOC(0) H-, hydroxy(Ci-C4)alkylOC(0) H-, 5-6 membered
heterocycloalkyl-C(O)-, 5-6 membered heterocycloalkyl-(Ci-C4)alkyl- HC(0)-, 5-6 membered heterocycloalkyl-(Ci-C4)alkoxy-, 5-6 membered heteroaryl, or 5-6 membered heteroaryl-C(0) H, herein said 5-6 membered heterocycloalkyl and 5-6 membered heteroaryl are optionally substituted by (Ci-C3)alkyl or -(Ci-C3)alkyl-CN; and Z2 is CH.
In specific embodiments, R2 is fluoro, chloro, bromo, -CN, -CH3,
-OCH3, -OCHF2, -OH, B(OH)2, CF3C(OH)2-, CH3OCH2CH20-,
cyclopropyl, 5H-tetrazol-5-yl, pyrazol-3-yl, or 5-methyl-l,3,4-oxadiazol-2-yl.
In other specific embodiments, R2 is fluoro, chloro, bromo, -CN,
-CH3, -OH, B(OH)2, CF3C(OH)2-, CH3OCH2CH20-, 5H-tetrazol-5-yl,
pyrazol-3-yl, or 5-methyl-l,3,4-oxadiazol-2-yl.
In another specific embodiment, R2 is chloro, bromo, -CN, -CH3,
-OH, B(OH)2, CF3C(OH)2-, CH3OCH2CH20-, 5H-tetrazol-5-yl, , pyrazol-3- yl, or 5-methyl-l,3,4-oxadiazol-2-yl.
In specific embodiments, R3 is fluoro, chloro, bromo, -CN, -OCH3,
-OCHF2, B(OH)2, -COOH, CH3S02-, CH3S02NHC(0)-, CH3C(0)NH-,
(CH3)2NC(0)-, CH3OC(0)-, (CH3)C(0)N(CH3)-, HOCH2CH2C(0)NH-,
CH3OCH2CH2NHC(0)NH-, CH3S02CH2CH2NHC(0)-, CH3CH2NHC(0)NH-, CH3OC(0)NH-, morpholin-4-yl-CO-, pyrrolidin-l-yl-CH2CH2NHC(0)-,
pyridin-2-yl, tetrahydrofuran-2-yl-CH20-, pyrrolidin-l-yl-CH2CH20-,
tetrazol-5-yl, l-(2-cyanoethyl)-tetrazol-5-yl, pyrazol-l-yl, pyrazol-3-yl,
pyrazol-4-yl, l-methyl-pyrazol-3-yl, l-methyl-pyrrol-4-yl-C(0)NH-, 5- methyl-l,3,4-oxadiazol-2-yl, or 5-oxo-4,5-dihydro-l,3,4-oxadiazol-2-yl.
In other specific embodiments, R3 is fluoro, chloro, bromo, -OCH3,
B(OH)2, -COOH, CH3S02-, CH3S02NHC(0)-, CH3C(0)NH-,
(CH3)2NC(0)-, CH3OC(0)-, (CH3)C(0)N(CH3)-, HOCH2CH2C(0)NH-,
CH3OCH2CH2NHC(0)NH-, CH3S02CH2CH2NHC(0)-, CH3CH2NHC(0)NH-, CH3OC(0)NH-, morpholin-4-yl-CO-, pyrrolidin-l-yl-CH2CH2NHC(0)-,
tetrahydrofuran-2-yl-CH20-, pyrrolidin-l-yl-CH2CH20-, tetrazol-5-yl, l-(2- cyanoethyl)-tetrazol-5-yl, pyrazol-l-yl, pyrazol-3-yl, l-methyl-pyrazol-3-yl, l-methyl-pyrrol-4-yl-C(0)NH-, 5-methyl-l,3,4-oxadiazol-2-yl, or 5-oxo-4,5- dihydro-l,3,4-oxadiazol-2-yl. In still other embodiments, R is fluoro, chloro, bromo, -OCH3,
B(OH)2, -COOH, CH3SO2-, CH3S02 HC(0)-, CH3C(0) H-,
(CH3)2NC(0)-, CH3OC(0)-, (CH3)C(0)N(CH3)-, HOCH2CH2C(0) H-,
CH3OCH2CH2 HC(0) H-, CH3S02CH2CH2 HC(0)-, CH3CH2 HC(0) H-, CH3OC(0) H-, morpholin-4-yl-CO-, pyrrolidin-l-yl-CH2CH2NHC(0)-, tetrahydrofuran-2-yl-CH20-, pyrrolidin-l-yl-CH2CH20-, tetrazol-5-yl, l-(2- cyanoethyl)-tetrazol-5-yl, pyrazol-l-yl, pyrazol-3-yl, l-methyl-pyrazol-3-yl, l-methyl-pyrrol-4-yl-C(0)NH-, or 5-oxo-4,5-dihydro-l,3,4-oxadiazol-2-yl.
In one embodiment of the compounds of this invention, R4 is fluoro, chloro, methyl, or trifluorom ethyl. In another embodiment, R4 is fluoro. In yet another embodiment, R4 is methyl.
In one embodiment of the compounds of this invention, R5 is H. In another embodiment, R5 is methyl.
In one embodiment of the compounds of this invention, A is phenyl, 5-6 membered heteroaryl, or 5-6 membered heterocycloalkyl, wherein the carbonyl moiety and L are substituted 1,3 on ring A.
In another embodiment, A is a 5 membered heteroaryl containing one oxygen or sulfur atom and optionally containing one or two nitrogen atoms;
specifically A is furyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, or oxadiazolyl (more specifically, 1, 2, 4-oxadiazolyl or 1, 3, 4-oxadiazolyl). In another embodiment, A is a 5 membered heteroaryl containing one nitrogen atom and optionally containing one, two or three additional nitrogen atoms, specifically;
A is pyrrolyl, pyrazolyl, imidazolyl, triazolyl (more specifically, 1, 2, 3-triazolyl or 1, 2, 4-triazolyl) or tetrazolyl. In selected embodiments, A is triazolyl. In yet embodiment of this invention, A is a 5 or 6 membered heterocycloalkyl
specifically, A is piperidinyl or pyrrolidinyl. In a further embodiment of this invention, A is a 6 membered aromatic group selected from phenyl and pyridyl.
Another embodiment of this invention is directed to a compound according to Formula (II):
Figure imgf000019_0001
wherein:
A1 is C,
A4 is C or N,
and A2, A3, and A5 are each independently selected from CH, CRA, O, S, N, NH and NRA to form a furyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl or tetrazolyl ring moiety, wherein said ring moiety contains 0 or 1 of CRA and NRA; and wherein X, Z1, Z2, Z3, Z4, R5, L, and B are as defined herein,
or a salt, particularly a pharmaceutically acceptable salt, thereof.
In selected embodiments, A1 is C, A4 is C or N, and A2, A3, and A5 are each independently selected from CH, O, N, and NH to form an oxazolyl, isoxazolyl, oxadiazolyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl or tetrazolyl ring moiety.
In other selected embodiments, A1 and A4 are each C, and A2, A3 and A5 are each independently selected from N and NH to form a triazolyl ring moiety.
Another embodiment of this invention, wherein A is piperidinyl or pyrrolidinyl, may be represented by Formula (III):
Figure imgf000019_0002
wherein s is 0 or 1, A10 is N and X, Z1, Z2, Z3, Z4, R5 RA, m, L, and B are as defined herein. In specific embodiments, m is 0 and A is an unsubstituted piperidinyl or pyrrolidinyl moiety. In one embodiment of the compounds of this invention, m is 0. In another embodiment, m is 1 and RA is (Ci-C4)alkyl, specifically RA is (Ci-C2)alkyl. In selected embodiments, RA is methyl.
A further embodiment of this invention, wherein A is phenyl, pyridinyl, or pyridinyl- -oxide, may be represented by Formula (IV):
Figure imgf000020_0001
wherein:
A6, A7, A8, and A9 are each CH;
one of A6, A7, A8, and A9 is CRA and the others of A6, A7, A8, and A9 are CH;
one of A6, A7, A8, and A9 is N and the others of A6, A7, A8, and A9 are
CH;
one of A6, A7, A8, and A9 is N-0 and the other of A6, A7, A8, and A9 are
CH;
and X, Z1, Z2, Z3, Z4, R5, L, and B are as defined herein.
In one embodiment of the compounds of this invention, L is O, S, H, N(CH3), CH2, CH2CH2, CH(CH3), CHF, CF2, CH20, CH2N(CH3), CH2 H, or CH(OH). In another embodiment, L is O, S, N(CH3), CH2, CH2CH2, CH(CH3), CF2, CH20, CH2N(CH3), or CH(OH). In another embodiment, L is CH20,
CH2CH2, CH2 H, or CH2N(CH3). In a further embodiment, L is N(CH3),
CH(CH3), or CH(OH). In another further embodiment, L is -(R)CH(CH3). In a still further embodiment, L is O, CH2, or NH. In one selected embodiment, L is O. In another selected embodiment, L is CH2.
In one embodiment of the compounds of this invention, B is an
optionally substituted (C3-C6)cycloalkyl, phenyl, 5-6 membered heteroaryl, or 5-6 membered heterocycloalkyl; wherein said (C3-C6)cycloalkyl, phenyl, 5-6 membered heteroaryl, or 5-6 membered heterocycloalkyl is unsubstituted or is substituted by one or two substituents each independently selected from halogen, (Ci-C4)alkyl, halo(Ci-C4)alkyl, (Ci-C4)alkoxy, halo(Ci-C4)alkoxy, nitro, and (Ci-C4)alkylC(0)-.In one embodiment of the compounds of this invention, B is an optionally substituted 5-6 membered heteroaryl or 5-6 membered heterocycloalkyl. In one embodiment, B is an optionally substituted pyrazolyl, thienyl, pyridinyl (pyridyl), oxo-pyridyl, pyrimidinyl, isoxazolyl, morpholinyl, tetrahydropyranyl or tetrahydrofuranyl, wherein the pyrazolyl, thienyl, pyridinyl (pyridyl), oxo-pyridyl, pyrimidinyl, isoxazolyl, morpholinyl, tetrahydropyranyl or tetrahydrofuranyl is optionally substituted by one or two independently selected (Ci-C4)alkyl substituents. In another embodiment, Bis an optionally substituted pyrazolyl, thienyl, pyridinyl (pyridyl), oxo-pyridyl, pyrimidinyl, isoxazolyl, morpholinyl, or tetrahydrofuranyl, wherein the pyrazolyl, thienyl, pyridinyl (pyridyl), oxo-pyridyl, pyrimidinyl, isoxazolyl, morpholinyl, or tetrahydrofuranyl is optionally substituted by one or two independently selected (Ci-C4)alkyl substituents. In specific embodiments, B is thien-2-yl (thiophen-2-yl), 5-methyl-thien-2-yl (5-methyl-thiophen-2-yl), pyrazol-l-yl, 3,5-dimethylpyrazol-l-yl, 4-methylpyrazol-l-yl,
3,5-dimethylisoxazol-4-yl, tetrahydropyran-3-yl, tetrahydrofuran-2-yl, morpholin-4-yl, pyridin-2-yl, 2-oxo-pyridin-l-yl, 6-methylpyridin-3-yl, or 2-methylpyrimidin-5-yl.
In other specific embodiments, B is thien-2-yl (thiophen-2-yl), 5-methyl- thien-2-yl (5-methyl-thiophen-2-yl), pyrazol-l-yl, 3,5-dimethylpyrazol-l-yl, 4-methylpyrazol-l-yl, 3,5-dimethylisoxazol-4-yl, tetrahydrofuran-2-yl, morpholin-4-yl, pyridin-2-yl, 2-oxo-pyridin-l-yl, 6-methylpyridin-3-yl, and 2-methylpyrimidin-5-yl.
In specific embodiments, B is thien-2-yl, pyrazol-l-yl,
3,5-dimethylpyrazol-l-yl, 4-methylpyrazol-l-yl, 3,5-dimethylisoxazol-4-yl, tetrahydrofuran-2-yl, morpholin-4-yl, pyridin-2-yl, 2-oxo-pyridin-l-yl,
6-methylpyridin-3-yl, and 2-methylpyrimidin-5-yl.
In another embodiment of the compounds of this invention, B is unsubstituted (C3-C6)cycloalkyl or phenyl. In a selected embodiment of this invention, B is unsubstituted cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl. In a specific embodiment, B is unsubstituted cyclopentyl or cyclohexyl. In another selected embodiment of the compounds of this invention, B is
unsubstituted phenyl.
In another selected embodiment, B is substituted phenyl. In one
embodiment, B is phenyl, substituted by 1 or 2 substituents independently
selected from halogen, (Ci-C4)alkyl, halo(Ci-C4)alkyl, (Ci-C4)alkoxy,
halo(Ci-C4)alkoxy, nitro, and (Ci-C4)alkylC(0)-. In other embodiments, B is
phenyl, substituted by 1 or 2 substituents independently selected from halogen,
(Ci-C3)alkyl and (Ci-C3)alkoxy. In specific embodiments, B is phenyl,
substituted by a substituent selected from fluoro, chloro, bromo, iodo, nitro,
methyl, ethyl, isopropyl, trifluoromethyl, methoxy, and -COCH3. In specific
embodiments, B is phenyl, substituted by 1 or 2 substituents independently
selected from iodo, fluoro, chloro, bromo, methyl and methoxy.
In one selected embodiment, B is phenyl, substituted by 1 or 2
substituents independently selected from fluoro, chloro, bromo, and methyl,
specifically B is phenyl, substituted by 1 or 2 fluoro substituents. In specific
embodiments, B is cyclopentyl, cyclohexyl, 2-methylphenyl, 4-methylphenyl,
2-trifluoromethylphenyl, 2-fluorophenyl, 3 -fluorophenyl, 4-fluorophenyl,
2-iodophenyl, 3-bromophenyl, 4-bromophenyl, 4-chlorophenyl,
2,5-difluorophenyl, 2,4-difluorophenyl, 3,4-difluorophenyl,
3,5-difluorophenyl, or 4-methoxyphenyl. In other embodiments, B is
2,3-difluorophenyl or 2,6-difluorophenyl.
In one embodiment of the compounds of this invention, the moiety -L-B is (C3-C6)alkyl, (C3-C6)alkoxy, halo(C3-C6)alkoxy, (C3-C6)alkenyl, or
(C3-C6)alkenyloxy. In another embodiment, the moiety -L-B is (C3-C6)alkyl,
(C3-C6)alkoxy, or (C3-C5)alkenyloxy. In specific embodiments, -L-B is
-OCH2CH=CH2, -CH2CH2CH2CH2CH3, -OCH2CH2CH2CH3, -CH2CH2CH3,
-CH2CH(CH3)2 or -CH2CH2CH(CH3)2. In other specific embodiments, -L-B is
-OCH2CH=CH2, -CH2CH2CH2CH2CH3, -OCH2CH2CH2CH3, -CH2CH2CH3, or
-CH2CH(CH3)2.
Representative compounds of this invention include the compounds of the
Examples. It will be appreciated that the present invention encompasses compounds of Formula (I) as the free base and as salts thereof, for example as a pharmaceutically acceptable salt thereof. In one embodiment the invention relates to compounds of Formula (I) in the form of a free base. In another embodiment the invention relates to compounds of Formula (I) in the form of a salt, particularly, a pharmaceutically acceptable salt. It will be further appreciated that, in one embodiment, the invention relates to compounds of the Examples in the form of a free base. In another embodiment the invention relates to compounds of the Examples in the form of a salt, particularly, a pharmaceutically acceptable salt.
Specifically, this invention is directed to (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide; (S)-5-benzyl-N- (6-fluoro-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b] [l,4]oxazepin-3-yl)-4H-l,2,4- triazole-3-carboxamide; and 5-benzyl-N-(7-bromo-2-oxo-2,3,4,5-tetrahydro-lH- benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3-carboxamide, or a salt thereof, particularly a pharmaceutically acceptable salt, thereof. Specifically, this invention is directed to (S)-5- benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4- triazole-3-carboxamide; (S)-5-benzyl-N-(6-fluoro-5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b] [l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide; or 5 -benzyl -N- (7-bromo-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3- carboxamide, or a pharmaceutically acceptable salt, thereof.
More specifically, this invention is directed to (S)-5-benzyl-N-(5-methyl-4-oxo- 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide and (S)-5- benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3- carboxamide, or a pharmaceutically acceptable salt thereof.
This invention is directed to (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide or a salt, particularly a pharmaceutically acceptable salt, thereof. Accordingly, one particular compound of the invention is (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide (free base). In another embodiment, the compound of the invention is a salt of (S)-5-benzyl-N-(5-methyl- 4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide. In another embodiment, the compound of the invention is a pharmaceutically acceptable salt of (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H- l,2,4-triazole-3-carboxamide. In another embodiment, the compound of the invention is a base-addition salt of (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide. In still another embodiment, the compound of the invention is a crystalline form of anhydrous (S)-5- benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4- triazole-3 -carboxamide (free base) characterized by the PXRD pattern of Figure 7. In yet another embodiment, a particular compound of the invention is a crystalline form of anhydrous (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)- 4H-l,2,4-triazole-3 -carboxamide (free base) characterized by the diffraction data in Table 1.
This invention is directed (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH- benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3-carboxamide, or a salt, particularly a
pharmaceutically acceptable salt, thereof. Accordingly, one particular compound of the invention is (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)- 4H-l,2,4-triazole-3 -carboxamide (free base). In another embodiment, the compound of the invention is a salt of (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH- benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3-carboxamide. In another embodiment, the compound of the invention is a pharmaceutically acceptable salt of (S)-5-benzyl-N-(7- chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3- carboxamide. In another embodiment, the compound of the invention is a base-addition salt of (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4- tri azol e-3 -carb oxami de .
In a specific embodiment, this invention is directed to a compound of
Formula (I) wherein:
X is O, S, SO, S02, NH, CO, CH2, or N(CH3);
Y is CH2 or CH2CH2;
Z1, Z2, Z3, and Z4 are each CH; or Z1 is CR1 and Z2, Z3 and Z4 are each
CH; or Zl, Z2, and Z4 are each CH and Z3 is CR3; or Zl, Z3, and Z4 are each CH
and Z2 is CR2; or Zl, Z2, and Z3 are each CH and Z4 is CR4; or Z1 and Z2 are CH,
Z3 is CR3, and Z4 is CR4; or Z1 and Z4 are CH, Z2 is CR2, and Z3 is CR3; or Z1
and Z3 are CH, Z2 is CR2, and Z4 is CR4; or Z1 is CH, Z2 is CR2, Z3 is CR3, and
Z4 is CR4; or Z1 and Z3 are both N, Z2 is CH and Z4 is CH or CR4; or Z1 and Z3
are both N, Z2 is CH or CR2 and Z4 is CH; or Z1 is N, Z2 is CR4 and Z3 and Z4
are CH; or Z3 is N, and Z2, Z3 and Z4 are CH;
R1 is methyl, R2 is fluoro, chloro, bromo, -CN, -CH3, -OCH3, -OCHF2, -OH, B(OH)2, CF3C(OH)2-, CH3OCH2CH2O-, cyclopropyl, 5H-tetrazol-5-yl, pyrazol-3-yl, or 5-methyl-l,3,4-oxadiazol-2-yl;
R3 is fluoro, chloro, bromo, -CN, -OCH3, -OCHF2, B(OH)2, -COOH, CH3S02-, CH3S02NHC(0)-, CH3C(0)NH-, (CH3)2NC(0)-, CH3OC(0)-, (CH3)C(0)N(CH3)-, HOCH2CH2C(0)NH-, CH3OCH2CH2NHC(0)NH-, CH3S02CH2CH2NHC(0)-, CH3CH2NHC(0)NH-, CH3OC(0)NH-,
morpholin-4-yl-CO-, pyrrolidin-l-yl-CH2CH2NHC(0)-, pyridin-2-yl, tetrahydrofuran-2-yl-CH20-, pyrrolidin-l-yl-CH2CH20-, tetrazol-5-yl, l-(2- cyanoethyl)-tetrazol-5-yl, pyrazol-l-yl, pyrazol-3-yl, pyrazol-4-yl, 1-methyl- pyrazol-3-yl, 1 -methyl-pyrrol -4-yl-C(0)NH-, 5-methyl-l,3,4-oxadiazol-2-yl, or 5 -oxo-4, 5 -dihy dro- 1 , 3 ,4-oxadiazol-2-yl ;
R4 is fluoro, chloro, methyl, or trifluoromethyl;
R5 is H or methyl;
A is furyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, 1, 2, 4-oxadiazolyl, 1, 3, 4-oxadiazolyl, pyrrolyl, pyrazolyl, imidazolyl, 1, 2, 3-triazolyl, 1, 2, 4- triazolyl, tetrazolyl, piperidinyl, pyrrolidinyl, phenyl or pyridyl;
m is 0 or m is 1 and RA is methyl;
L is O, S, N(CH3), CH2, CH2CH2, CH(CH3), -(R)CH(CH3), CF2, CH20, CH2N(CH3), or CH(OH); and
B is thien-2-yl, 5-methyl-thien-2-yl, pyrazol-l-yl,
3,5-dimethylpyrazol-l-yl, 4-methylpyrazol-l-yl, 3,5-dimethylisoxazol-4-yl, tetrahydropyran-3-yl, tetrahydrofuran-2-yl, morpholin-4-yl, pyridin-2-yl, 2-oxo-pyridin-l-yl, 6-methylpyridin-3-yl, 2-methylpyrimidin-5-yl, cyclopentyl, cyclohexyl, phenyl, 2-methylphenyl, 4-methylphenyl,
2-trifluoromethylphenyl, 2-fluorophenyl, 3 -fluorophenyl, 4-fluorophenyl, 2-iodophenyl, 3-bromophenyl, 4-bromophenyl, 4-chlorophenyl,
2,5-difluorophenyl, 2,4-difluorophenyl, 3,4-difluorophenyl,
3,5-difluorophenyl, 4-methoxyphenyl, 2,3-difluorophenyl or 2,6-difluorophenyl; or -L-B-RB is -OCH2CH=CH2, -CH2CH2CH2CH2CH3,
-OCH2CH2CH2CH3, -CH2CH2CH3, -CH2CH(CH3)2 or -CH2CH2CH(CH3)2; or a salt, particularly, a pharmaceutically acceptable salt thereof. In another specific embodiment, this invention is directed to a compound of Formula (I) wherein:
X is O, S, SO, S02, NH, CO, CH2, or N(CH3);
Y is CH2 or CH2CH2;
Z1, Z2, Z3, and Z4 are each CH; or Z1 is CR1 and Z2, Z3 and Z4 are each
CH; or Z1, Z2, and Z4 are each CH and Z3 is CR3; or Z1, Z3, and Z4 are each CH and Z2 is CR2; or Zl, Z2, and Z3 are each CH and Z4 is CR4; or Z1 and Z3 are CH, Z2 is CR2, and Z4 is CR4; or Z1 and Z3 are both N, Z2 is CH and Z4 is CH or CR4; or Z1 is N, Z2 is CR4 andZ3 and Z4 are CH; or Z3 is N, and Z2, Z3 and Z4 are CH;
R1 is methyl,
R2 is fluoro, chloro, bromo, -CN, -CH3, -OH, B(OH)2, CF3C(OH)2-, CH3OCH2CH20-, 5H-tetrazol-5-yl, pyrazol-3-yl, or 5-methyl-l,3,4-oxadiazol- 2-yl;
R3 is fluoro, chloro, bromo, -OCH3, B(OH)2, -COOH, CH3S02-,
CH3S02 HC(0)-, CH3C(0) H-, (CH3)2NC(0)-, CH3OC(0)-,
(CH3)C(0)N(CH3)-, HOCH2CH2C(0) H-, CH3OCH2CH2NHC(0) H-, CH3S02CH2CH2NHC(0)-, CH3CH2 HC(0) H-, CH3OC(0) H-,
mo holin-4-yl-CO-, pyrrolidin- 1 -yl-CH2CH2 HC(0)-, tetrahydrofuran-2-yl- CH20-, pyrrolidin- l-yl-CH2CH20-, tetrazol-5-yl, l-(2-cyanoethyl)-tetrazol-5- yl, pyrazol-l-yl, pyrazol-3-yl, l-methyl-pyrazol-3-yl, 1 -methyl-pyrrol -4-yl- C(0) H-, 5-methyl-l,3,4-oxadiazol-2-yl, or 5-oxo-4,5-dihydro-l,3,4- oxadiazol-2-yl;
R4 is fluoro, chloro, methyl, or trifluoromethyl;
R5 is H or methyl;
A is furyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, 1, 2, 4-oxadiazolyl, 1, 3, 4-oxadiazolyl, pyrrolyl, pyrazolyl, imidazolyl, 1, 2, 3-triazolyl, 1, 2, 4- triazolyl, tetrazolyl, piperidinyl, pyrrolidinyl, phenyl or pyridyl;
m is 0 or m is 1 and RA is methyl;
L is O, S, N(CH3), CH2, CH2CH2, CH(CH3), CF2, CH20, CH2N(CH3), or
CH(OH); and
B is thien-2-yl, 5-methyl-thien-2-yl, pyrazol-l-yl,
3,5-dimethylpyrazol-l-yl, 4-methylpyrazol-l-yl, 3,5-dimethylisoxazol-4-yl, tetrahydrofuran-2-yl, morpholin-4-yl, pyridin-2-yl, 2-oxo-pyridin-l-yl,
6-methylpyridin-3-yl, 2-methylpyrimidin-5-yl, cyclopentyl, cyclohexyl, phenyl,
2- methylphenyl, 4-methylphenyl, 2-trifluoromethylphenyl, 2-fluorophenyl,
3 - fluorophenyl, 4-fluorophenyl, 2-iodophenyl, 3-bromophenyl,
4-bromophenyl, 4-chlorophenyl, 2,5-difluorophenyl, 2,4-difluorophenyl, 3,4-difluorophenyl, 3,5-difluorophenyl, or 4-methoxyphenyl;
or -L-B-RB is -OCH2CH=CH2, -CH2CH2CH2CH2CH3,
-OCH2CH2CH2CH3, -CH2CH2CH3, -CH2CH(CH3)2 or -CH2CH2CH(CH3)2; or a salt, particularly, a pharmaceutically acceptable salt thereof.
In another specific embodiment, this invention is directed to a compound of Formula (I) wherein:
X is O, CH2, NH or N(CH3);
Y is CH2 or CH2CH2;
Z1, Z2, Z3, and Z4 are each CH; or Z1 is CR1 and Z2, Z3 and Z4 are each CH; or Zl, Z2, and Z4 are each CH and Z3 is CR3; or Zl, Z3, and Z4 are each CH and Z2 is CR2; or Zl, Z2, and Z3 are each CH and Z4 is CR4; or Z1 and Z3 are CH, Z2 is CR2, and Z4 is CR4; or Z1 and Z3 are both N, Z2 is CH and Z4 is CH or CR4; or Z1 is N, Z2 is CR4 andZ3 and Z4 are CH; or Z3 is N, and Z2, Z3 and Z4 are CH;
R1 is methyl,
R2 is chloro, bromo, -CN, -CH3, -OH, B(OH)2, CF3C(OH)2-, CH3OCH2CH20-, 5H-tetrazol-5-yl, , pyrazol-3-yl, or 5-methyl-l,3,4- oxadiazol-2-yl;
R3 is fluoro, chloro, bromo, -OCH3, B(OH)2, -COOH, CH3S02-, CH3S02 HC(0)-, CH3C(0) H-, (CH3)2NC(0)-, CH3OC(0)-,
(CH3)C(0)N(CH3)-, HOCH2CH2C(0) H-, CH3OCH2CH2NHC(0) H-, CH3S02CH2CH2NHC(0)-, CH3CH2 HC(0) H-, CH3OC(0) H-,
morpholin-4-yl-CO-, pyrrolidin- 1 -yl-CH2CH2 HC(0)-, tetrahydrofuran-2-yl- CH20-, pyrrolidin- l-yl-CH2CH20-, tetrazol-5-yl, l-(2-cyanoethyl)-tetrazol-5- yl, pyrazol-l-yl, pyrazol-3-yl, l-methyl-pyrazol-3-yl, 1 -methyl-pyrrol -4-yl- C(0) H-, or 5-oxo-4,5-dihydro-l,3,4-oxadiazol-2-yl;
R4 is fluoro or methyl; A is furyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, 1, 2, 4-oxadiazolyl, 1,
3, 4-oxadiazolyl, pyrrolyl, pyrazolyl, imidazolyl, 1, 2, 3-triazolyl, 1, 2, 4- triazolyl, tetrazolyl, piperidinyl, pyrrolidinyl, phenyl or pyridyl;
m is 0 or m is 1 and RA is methyl;
L is O, S, N(CH3), CH2, CH2CH2, CH(CH3), CF2, CH20, CH2N(CH3), or
CH(OH); and
B is thien-2-yl, pyrazol-l-yl, 3,5-dimethylpyrazol-l-yl,
4-methylpyrazol-l-yl, 3,5-dimethylisoxazol-4-yl, tetrahydrofuran-2-yl,
morpholin-4-yl, pyridin-2-yl, 2-oxo-pyridin-l-yl, 6-methylpyridin-3-yl,
2-methylpyrimidin-5-yl, cyclopentyl, cyclohexyl, phenyl, 2-methylphenyl,
4-methylphenyl, 2-trifluoromethylphenyl, 2-fluorophenyl, 3 -fluorophenyl,
4-fluorophenyl, 2-iodophenyl, 3-bromophenyl, 4-bromophenyl,
4-chlorophenyl, 2,5-difluorophenyl, 2,4-difluorophenyl, 3,4-difluorophenyl,
3,5-difluorophenyl, or 4-methoxyphenyl;
or -L-B is -OCH2CH=CH2, -CH2CH2CH2CH2CH3, -OCH2CH2CH2CH3,
-CH2CH2CH3, or -CH2CH(CH3)2;
or a pharmaceutically acceptable salt thereof.
In another specific embodiment, this invention is directed to a compound of Formula (I) wherein X is O or CH2; Y is CH2; Z1, Z2, and Z4 are each CH
and Z3 is CR3; or Z1, Z3, and Z4 are each CH and Z2 is CR2; or Z1, Z2, and Z3 are each CH and Z4 is CR4; or Z1 and Z3 are CH, Z2 is CR2, and Z4 is CR4; R2 is
fluoro, chloro, bromo, or -CH3; R3 is 5-methyl-l,3,4-oxadiazol-2-yl; R4 is
fluoro; R5 is H or methyl; A is triazolyl; m is 0; L is CH2; and B is
cyclopentyl or phenyl; or a salt, particularly a pharmaceutically acceptable salt thereof.
The compounds of this invention contain one or more asymmetric centers (also referred to as a chiral center), such as a chiral carbon, or a chiral -SO- moiety. The stereochemistry of the chiral carbon center present in compounds of this invention is generally represented in the compound names and/or in the chemical structures illustrated herein. Compounds of this invention containing one or more chiral centers may be present as racemic mixtures, diastereomeric mixtures, enantiomerically enriched mixtures, diastereomerically enriched mixtures, or as enantiomerically or diastereomerically pure individual stereoisomers. Individual stereoisomers of a compound of this invention may be resolved (or mixtures of stereoisomers may be enriched) using methods known to those skilled in the art. For example, such resolution may be carried out (1) by formation of diastereoisomeric salts, complexes or other derivatives; (2) by selective reaction with a stereoisomer-specific reagent, for example by enzymatic oxidation or reduction; or (3) by gas-liquid or liquid chromatography in a chiral environment, for example, on a chiral support such as silica with a bound chiral ligand or in the presence of a chiral solvent. The skilled artisan will appreciate that where the desired stereoisomer is converted into another chemical entity by one of the separation procedures described above, a further step is required to liberate the desired form. Alternatively, specific stereoisomers may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer to the other by asymmetric transformation.
The invention also includes various deuterated forms of the compounds of this invention, a specific example of which is N-[(3S)-7-deuterio -l-methyl-2-oxo-2,3,4,5- tetrahydro-lH-l-benzazepin-3-yl]-5-(phenylmethyl)-lH-pyrazole-3-carboxamide. Each available hydrogen atom attached to a carbon atom may be independently replaced with a deuterium atom. A person of ordinary skill in the art will know how to synthesize deuterated forms of the compounds of this invention. For example, a-deuterated a-amino acids are commercially available or may be prepared by conventional techniques (see for example: Elemes, Y. and Ragnarsson, U. J. Chem. Soc, Perkin Trans. 1, 1996, 6, 537-40). Employing such compounds may allow for the preparation of compounds in which the hydrogen atom at a chiral center is replaced with a deuterium atom. Other commercially available deuterated starting materials may be employed in the preparation of deuterated analogs of the compounds of this invention (see for example: methyl-i¾-amine available from Aldrich Chemical Co., Milwaukee, WI), or they may be synthesized using
conventional techniques employing deuterated reagents (e.g. by reduction using lithium aluminum deuteride or sodium borodeuteride or by metal-halogen exchange followed by quenching with D20 or methanol-^)-
The skilled artisan will appreciate that solvates (particularly, hydrates) of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), including solvates of salts of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), may be formed when solvent molecules are incorporated into the crystalline lattice during crystallization. The present invention includes within its scope all possible stoichiometric and non-stoichiometric salt and/or hydrate forms.
When a disclosed compound or its salt is named or depicted by structure, it is to be understood that the compound or salt, including solvates (particularly, hydrates) thereof, may exist in crystalline forms, non-crystalline forms or a mixture thereof. The compound or salt, or solvates (particularly, hydrates) thereof, may also exhibit polymorphism (i.e. the capacity to occur in different crystalline forms). These different crystalline forms are typically known as "polymorphs." It is to be understood that when named or depicted by structure, the disclosed compound, or solvates (particularly, hydrates) thereof, also include all polymorphs thereof. Polymorphs have the same chemical composition but differ in packing, geometrical arrangement, and other descriptive properties of the crystalline solid state. Polymorphs, therefore, may have different physical properties such as shape, density, hardness, deformability, stability, and dissolution properties. Polymorphs typically exhibit different melting points, IR spectra, and X-ray powder diffraction patterns, which may be used for identification. One of ordinary skill in the art will appreciate that different polymorphs may be produced, for example, by changing or adjusting the conditions used in crystallizing/recrystallizing the compound. It is well known and understood to those skilled in the art that the apparatus employed, humidity, temperature, orientation of the powder crystals, and other parameters involved in obtaining a powder X-ray diffraction (PXRD) pattern may cause some variability in the appearance, intensities, and positions of the lines in the diffraction pattern. A powder X-ray diffraction pattern that is "substantially in accordance" with that of the Figure provided herein is a PXRD pattern that would be considered by one skilled in the art to represent a compound possessing the same crystal form as the compound that provided the PXRD pattern of the Figure. For example, the PXRD pattern may be identical to that of Figure 7, or more likely it may be somewhat different. Such a PXRD pattern may not necessarily show each of the lines of the diffraction patterns presented herein, and/or may show a slight change in appearance, intensity, or a shift in position of said lines resulting from differences in the conditions involved in obtaining the data. A person skilled in the art is capable of determining if a sample of a crystalline compound has the same form as, or a different form from, a form disclosed herein by comparison of their PXRD patterns. For example, one skilled in the art can overlay a PXRD pattern of a sample of a crystalline form of anhydrous (S)-5-benzyl-N- (5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3- carboxamide (free base) with the PXRD pattern of Fig. 7, and using expertise and knowledge in the art, readily determine whether the PXRD pattern of the sample is substantially in accordance with the PXRD pattern of Figure 7. If the PXRD pattern is substantially in accordance with Fig. 7, the sample form can be readily and accurately identified as having the same form as the crystalline form of anhydrous (S)-5-benzyl-N-(5- methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3- carboxamide (free base) described herein. Similarly, a person skilled in the art is capable of determining if a given diffraction angle (expressed in °2Θ) obtained from a PXRD pattern is at about the same position as a recited value.
Because of their potential use in medicine, the salts of the compounds of Formula
(I), particularly a compound of any one of Formulas (I-IV), are preferably pharmaceutically acceptable. Suitable pharmaceutically acceptable salts can include acid or base addition salts.
As used herein, the term "pharmaceutically acceptable" means a compound which is suitable for pharmaceutical use. Salts and solvates (e.g. hydrates and hydrates of salts) of the compounds of the invention which are suitable for use in medicine are those wherein the counterion or associated solvent is pharmaceutically acceptable. Salts and solvates having non-pharmaceutically acceptable counterions or associated solvents are within the scope of the present invention, for example, for use as intermediates in the preparation of other compounds of the invention and their salts and solvates.
Salts may be prepared in situ during the final isolation and purification of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV). If a basic compound of Formula (I-IV) is isolated as a salt, the corresponding free base form of that compound may be prepared by any suitable method known to the art, including treatment of the salt with an inorganic or organic base, suitably an inorganic or organic base having a higher pKa than the free base form of the compound. Similarly, if a disclosed compound containing a carboxylic acid or other acidic functional group is isolated as a salt, the corresponding free acid form of that compound may be prepared by any suitable method known to the art, including treatment of the salt with an inorganic or organic acid, suitably an inorganic or organic acid having a lower pKa than the free acid form of the compound. This invention also provides for the conversion of one salt of a compound of this invention, e.g., a hydrochloride salt, into another salt of a compound of this invention, e.g., a sulfate salt. Salts of the compounds of Formula (I), particularly compounds of Formulas (I-IV), containing a basic amine or other basic functional group may be prepared by any suitable method known in the art, such as treatment of the free base with an acid. Examples of pharmaceutically acceptable salts so formed include acetate, adipate, ascorbate, aspartate, benzenesulfonate, benzoate, camphorate, camphor-sulfonate (camsylate), caprate
(decanoate), caproate (hexanoate), caprylate (octanoate), carbonate, bicarbonate, cinnamate, citrate, cyclamate, dodecyl sulfate (estolate), ethane-l,2-disulfonate (edisylate),
ethanesulfonate (esylate), formate, fumarate, galactarate (mucate), gentisate (2,5- dihydroxybenzoate), glucoheptonate (gluceptate), gluconate, glucuronate, glutamate, glutarate, glycerophosphorate, glycolate, hippurate, hydrobromide, hydrochloride, hydroiodide, isobutyrate, lactate, lactobionate, laurate, maleate, malate, malonate, mandelate, methanesulfonate (mesylate), naphthalene-l,5-disulfonate (napadisylate), naphthalene-sulfonate (napsylate), nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, phosphate, diphosphate, proprionate, pyroglutamate, salicylate, sebacate, stearate, succinate, sulfate, tartrate, thiocyanate, ^-toluenesulfonate (tosylate), undecylenate, 1- hydroxy-2-naphthoate, 2,2-dichloroacetate, 2-hydroxyethanesulfonate (isethionate), 2- oxoglutarate, 4-acetamidobenzoate, and 4-aminosalicylate.
Salts of the disclosed compounds containing a carboxylic acid or other acidic functional group can be prepared by reacting with a suitable base. Such a pharmaceutically acceptable salt may be made with a base which affords a pharmaceutically acceptable cation, which includes alkali metal salts (especially sodium and potassium), alkaline earth metal salts (especially calcium and magnesium), aluminum salts and ammonium salts, as well as salts made from physiologically acceptable organic bases such as trimethylamine, triethylamine, morpholine, pyridine, piperidine, picoline, dicyclohexylamine, N P- dibenzylethylenediamine, 2-hydroxyethylamine, /s-(2-hydroxyethyl)amine, tri-(2- hydroxyethyl)amine, procaine, dibenzylpiperidine, dehydroabietylamine, N P- bisdehydroabietylamine, glucamine, N-methylglucamine, collidine, choline, quinine, quinoline, and basic amino acids such as lysine and arginine. In one embodiment, the pharmaceutically acceptable base-addition salt of a compound of Formula (I) is a sodium salt or a potassium salt thereof.
Because the compounds of this invention are intended for use in pharmaceutical compositions it will readily be understood that they are each preferably provided in substantially pure form, for example at least 60% pure, more suitably at least 75% pure and preferably at least 85%, especially at least 98% pure (% are on a weight for weight basis). Impure preparations of the compounds may be used for preparing the more pure forms used in the pharmaceutical compositions.
GENERAL SYNTHETIC METHODS
The compounds of this invention may be prepared using synthetic procedures illustrated in the Schemes below or by drawing on the knowledge of a skilled organic chemist. The synthesis provided in these Schemes are applicable for producing compounds of the invention having a variety of different R groups employing appropriate precursors, which are suitably protected if needed, to achieve compatibility with the reactions outlined herein. Subsequent deprotection, where needed, affords compounds of the nature generally disclosed. While the Schemes are shown with compounds only of Formulas (I-IV), they are illustrative of processes that may be used to make the compounds of the invention.
Intermediates (compounds used in the preparation of the compounds of the invention) may also be present as salts. Thus, in reference to intermediates, the phrase "compound(s) of formula (number)" means a compound having that structural formula or a pharmaceutically acceptable salt thereof.
(S)-N-(4-Oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)carboxamides may be prepared via the general method outlined in Scheme 1. Boc-L-serine can be condensed with an appropriate substituted l-fluoro-2-nitrobenzene with a base, followed by reduction of the nitro group to the amine and cyclisation to the Boc protected (S)-3-amino-2,3- dihydrobenzo[b][l,4]oxazepin-4(5H)-one using an amide coupling agent. The Boc protecting group can be removed using acidic conditions and the resulting free amine can be coupled to the appropriate acid using an amide coupling agent. The Boc protected (S)-3- amino-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one can also be methylated to give the Boc protected (S)-3-amino-5-methyl-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one, which can then be deprotected using acidic conditions, and the resulting free amine can be coupled to an appropriate acid using an amide coupling agent. Scheme 1
Figure imgf000034_0001
(R)-N-(4-Oxo-2,3,4,5-tetrahydrobenzo[b][l,4]thiazepin-3-yl)carboxamides may be prepared via the general method outlined in Scheme 2. Boc-L-cysteine can be condensed with an appropriate substituted l-fluoro-2-nitrobenzene with a base, followed by reduction of the nitro group to the amine and cyclisation to the Boc protected (R)-3-amino-2,3- dihydrobenzo[b][l,4]thiazepin-4(5H)-one using an amide coupling agent. The Boc protecting group can be removed using acidic conditions and the resulting free amine can be coupled to an appropriate acid using an amide coupling agent. The Boc protected (R)-3- amino-2,3-dihydrobenzo[b][l,4]thiazepin-4(5H)-one can also be methylated to give the Boc protected (R)-3-amino-5-methyl-2,3-dihydrobenzo[b][l,4]thiazepin-4(5H)-one, which can then be deprotected using acidic conditions, and the resulting free amine can be coupled to an appropriate acid using an amide coupling agent. Scheme 2
Figure imgf000035_0001
EDC NMM
Figure imgf000035_0002
HATU
R1C02H
DIEA HCI
Figure imgf000035_0003
(S)-N-(2-Oxo-2,3,4,5-tetrahydro-lH-benzo[b][l,4]diazepin-3-yl)carboxamides may be prepared via the general method outlined in Scheme 3. 3-Aminoalanine can be condensed with the appropriate substituted l-fluoro-2-nitrobenzene with a base, followed by reduction of the nitro group to the amine and cyclisation to the Boc protected (S)-3- amino-4,5-dihydro-lH-benzo[b][l,4]diazepin-2(3H)-one using an amide coupling agent. The Boc protecting group can be removed using acidic conditions and the resulting free amine can be coupled to the appropriate acid using an amide coupling agent. The Boc protected (S)-3-amino-4,5-dihydro-lH-benzo[b][l,4]diazepin-2(3H)-one can also be methylated with methyl iodide using sodium hydride as base to give the Boc protected (S)- 3-amino-l-methyl-4,5-dihydro-lH-benzo[b][l,4]diazepin-2(3H)-one, which can then be deprotected using acidic conditions, and the resulting free amine can be coupled to the appropriate acid using an amide coupling agent. Alternatively, the Boc protected (S)-3- amino-4,5-dihydro-lH-benzo[b][l,4]diazepin-2(3H)-one can also be methylated with methyl iodide using potassium carbonate as base in ethanol to give the Boc protected (S)-3- amino-5-methyl-4,5-dihydro-lH-benzo[b][l,4]diazepin-2(3H)-one, which can then be deprotected using acidic conditions, and the resulting free amine can be coupled to the appropriate acid using an amide coupling agent. Schem
Figure imgf000036_0001
(S)-N-(6-Oxo-6,7,8,9-tetrahydro-5H-pyrimido[4,5-b][l,4]diazepin-7- yl)carboxamides may be prepared via the general method outlined in Scheme 4. Boc-L- serine can be condensed with 4,6-dichloropyrimidin-5-amine in the presence of a base such as triethylamine, followed by cyclisation to the Boc protected (S)-7-amino-4-chloro-8,9- dihydro-5H-pyrimido[4,5-b][l,4]diazepin-6(7H)-one using an amide coupling agent. The chloride can be removed by reduction using hydrogenation with a palladium/carbon catalyst. The Boc protecting group can then be removed using acidic conditions and the resulting free amine can be coupled to the appropriate acid using an amide coupling agent. Alternatively, the Boc protected (S)-7-amino-8,9-dihydro-5H-pyrimido[4,5- b][l,4]diazepin-6(7H)-one can be methylated in the presence of a base, deprotected using acidic conditions, and the resulting free amine can be coupled to the appropriate acid using an amide coupling agent to yield (S)-N-(5-methyl-6-oxo-6,7,8,9-tetrahydro-5H- pyrimido[4,5-b][l,4]diazepin-7-yl)carboxamides (Scheme 4, R=H). If the chloride is not removed, then this sequence can be repeated to yield the corresponding (S)-N-(4-chloro-5- methyl-6-oxo-6,7,8,9-tetrahydro-5H-pyrimido[4,5-b][l,4]diazepin-7-yl)carboxamides (Scheme 4, R=C1).
Scheme 4
Figure imgf000037_0001
Pd/C
Figure imgf000037_0002
R.1CO2H HATU
DIEA TFA
Figure imgf000037_0003
R = H or CI
(S)-N-(2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl) carboxamides may be prepared via the general method outlined in Scheme 5. The appropriately substituted tetralone can be converted to the l,3,4,5-tetrahydro-l-benzazepin-2-one via either an acid- mediated Schmidt reaction with sodium azide, or Beckmann rearrangement of the corresponding ketoximes formed from reaction with hydroxylamine. The 1,3,4,5- tetrahydro-l-benzazepin-2-one can then be converted to the a-iodobenzlactam by iodotrimethylsilane-mediate iodination, subsequently converted to the a-azidobenzlactam with sodium azide, and following a Staudinger reduction with triphenylphosphine yields the a-aminobenzlactam. A racemization/resolution of the a-aminobenzlactam can be accomplished using L-pyroglutamic acid and 5-nirosalicylaldehyde to yield the (S)-3- amino-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one as described by Armstrong et. al. in Tetrahedron Letters 1994, pages 3239-42. This amine can then be coupled to the appropriate acid using an amide coupling agent. Alternatively the amine can protected with a Boc protecting group, then methylated at the lactam nitrogen with methyl iodide, followed by deprotected using acidic conditions. The resulting free amine can be coupled to the appropriate acid using an amide coupling agent.
Scheme 5
Figure imgf000038_0001
5-Substituted-4H-l,2,4-triazole-3-carboxylic acids may be prepared via the general method outlined in Scheme 6. The appropriately substituted acetohydrazide is condensed with ethyl 2-ethoxy-2-iminoacetate in ethanol. The resulting ethyl 2-amino-2-(2- substituted hydrazono)acetate is then heated neat or in a high boiling solvent such as xylenes resulting in cyclization to the 5-substituted-4H-l,2,4-triazole-3-carboxylic ethyl ester. This can then by hydrolyzed using aqueous base, for example lithium hydroxide in THF and water. Scheme 6
Figure imgf000039_0001
xylene or neat
N-N LiOH
N-N
ΰ THF/Water Eto2c N
M H
5-Benzyl-lH-pyrazole-3-carboxylic acids can be prepared following the route shown in Scheme 7. The appropriately substituted benzyl methyl ketone is condensed with diethyl oxalate in the presence of a base such as potassium tert-butoxide in ethanol. The resulting ethyl 5-(substituted-phenyl)-2,4-dioxopentanoate is then condensed with hydrazine in ethanol resulting in cyclization to the ethyl 5-substituted benzyl- lH-pyrazole- 3-carboxylate. This can then by hydrolyzed using aqueous base, for example lithium hydroxide in THF and water.
Scheme 7
Figure imgf000039_0002
H2NNH2
EtOH
Figure imgf000039_0003
5-Methyl-l-substituted-lH-pyrazole-3-carboxylic acids can be prepared following the route shown in Scheme 8. Ethyl 3-methyl-lH-pyrazole-5-carboxylate is alkylated in the presence of a base such as potassium hydroxide with the appropriate alkylating agent, such as an alkyl or arylbromide, to give a mixture of desired ethyl 5-methyl-l-propyl-lH- pyrazole-3-carboxylate and undesired regioisomer ethyl 3 -methyl- 1 -substituted- 1H- pyrazole-5-carboxylate ethyl oxalate. The ethyl 5-methyl-l-substituted-lH-pyrazole-3- carboxylates can be isolated using chromatography and then hydrolyzed using aqueous base, for example lithium hydroxide in THF and water to yield 5-methyl-l-propyl-lH- pyrazole-3-carboxylic acids.
Scheme 8
Figure imgf000040_0001
The compounds of this invention may be particularly useful for the treatment of RIPl kinase-mediated diseases or disorders. Such RIPl kinase-mediated diseases or disorders are diseases or disorders that are mediated by activation of RIPl kinase, and as such, are diseases or disorders where inhibition of RIPl kinase would provide benefit. The compounds of this invention may be particularly useful for the treatment of
diseases/disorders which are likely to be regulated at least in part by programmed necrosis, particularly inflammatory bowel disease (including Crohn's disease and ulcerative colitis), psoriasis, retinal detachment, retinitis pigmentosa, macular degeneration, pancreatitis, atopic dermatitis, arthritis (including rheumatoid arthritis, spondylarthritis, gout, SoJIA), systemic lupus erythematosus (SLE), Sjogren's syndrome, systemic scleroderma, anti- phospholipid syndrome (APS), vasculitis, osteoarthritis, liver damage/diseases (non-alcohol steatohepatitis, alcohol steatohepatitis, autoimmune hepatitis autoimmune hepatobiliary diseases, primary sclerosing cholangitis (PSC)), nephritis, Celiac disease, autoimmune ITP, transplant rejection, ischemia reperfusion injury of solid organs, sepsis, systemic inflammatory response syndrome (SIRS), cerebrovascular accident (CVA), myocardial infarction (MI), Huntington's disease, Alzheimer's disease, Parkinson's disease, allergic diseases (including asthma and atopic dermatitis), multiple sclerosis, type I diabetes, Wegener's granulomatosis, pulmonary sarcoidosis, Behcet's disease, interleukin-1 converting enzyme (ICE, also known as caspase-1) associated fever syndrome, chronic obstructive pulmonary disease (COPD), tumor necrosis factor receptor-associated periodic syndrome (TRAPS) and peridontitis.
The compounds of this invention may be particularly useful for the treatment of diseases/disorders which are likely to be regulated at least in part by programmed necrosis, apoptosis or the production of inflammatory cytokines, particularly inflammatory bowel disease (including Crohn's disease and ulcerative colitis), psoriasis, retinal detachment, retinitis pigmentosa, macular degeneration, pancreatitis, atopic dermatitis, arthritis
(including rheumatoid arthritis, spondyloarthritis, gout, systemic onset juvenile idiopathic arthritis (SoJIA), psoriatic arthritis), systemic lupus erythematosus (SLE), Sjogren's syndrome, systemic scleroderma, anti-phospholipid syndrome (APS), vasculitis, osteoarthritis, liver damage/diseases (non-alcohol steatohepatitis, alcohol steatohepatitis, autoimmune hepatitis, autoimmune hepatobiliary diseases, primary sclerosing cholangitis (PSC), acetaminophen toxicity, hepatotoxicity), kidney damage/injury (nephritis, renal transplant, surgery, administration of nephrotoxic drugs e.g. cisplatin, acute kidney injury(AKI)) Celiac disease, autoimmune idiopathic thrombocytopenic purpura
(autoimmune ITP), transplant rejection, ischemia reperfusion injury of solid organs, sepsis, systemic inflammatory response syndrome (SIRS), cerebrovascular accident (CVA, stroke), myocardial infarction (MI), atherosclerosis, Huntington's disease, Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis (ALS), allergic diseases (including asthma and atopic dermatitis), multiple sclerosis, type I diabetes, Wegener's
granulomatosis, pulmonary sarcoidosis, Behcet's disease, interleukin-1 converting enzyme (ICE, also known as caspase-1) associated fever syndrome, chronic obstructive pulmonary disease (COPD), tumor necrosis factor receptor-associated periodic syndrome (TRAPS), peridontitis, NEMO-deficiency syndrome ( F-kappa-B essential modulator gene (also known as IKK gamma or IKKG) deficiency syndrome), HOIL-1 deficiency ((also known as RBCKl) heme-oxidized IRP2 ubiquitin ligase-1 deficiency), linear ubiquitin chain assembly complex (LUBAC) deficiency syndrome, hematological and solid organ malignancies, bacterial infections and viral infections (such as tuberculosis and influenza), and Lysosomal storage diseases (particularly, Gaucher Disease, and including GM2
Gangliosidosis, Alpha-mannosidosis, Aspartylglucosaminuria, Cholesteryl Ester storage disease, Chronic Hexosaminidase A Deficiency, Cystinosis, Danon disease, Fabry disease, Farber disease, Fucosidosis, Galactosialidosis, GM1 gangliosidosis, Mucolipidosis, Infantile Free Sialic Acid Storage Disease, Juvenile Hexosaminidase A Deficiency, Krabbe disease, Lysosomal acid lipase deficiency, Metachromatic Leukodystrophy,
Mucopolysaccharidoses disorders, Multiple sulfatase deficiency, Niemann-Pick Disease, Neuronal Ceroid Lipofuscinoses, Pompe disease, Pycnodysostosis, Sandhoff disease, Schindler disease, Sialic Acid Storage Disease, Tay-Sachs and Wolman disease).
The treatment of the above-noted diseases/disorders may concern, more specifically, the amelioration of organ injury or damage sustained as a result of the noted diseases. For example, the compounds of this invention may be particularly useful for amelioration of brain tissue injury or damage following ischemic brain injury or traumatic brain injury, or for amelioration of heart tissue injury or damage following myocardial infarction, or for amelioration of brain tissue injury or damage associated with
Huntington's disease, Alzheimer's disease or Parkinson's disease, or for amelioration of liver tissue injury or damage associated with non-alcohol steatohepatitis, alcohol steatohepatitis, autoimmune hepatitis autoimmune hepatobiliary diseases, or primary sclerosing cholangitis. In addition, the treatment of diseases/disorders selected from those described herein may concern, more specifically, the amelioration of liver tissue injury or damage associated with overdose of acetaminophen, or for amelioration of kidney tissue injury or damage following renal transplant or the administration of nephrotoxic drugs or substances e.g. cisplatin.
The compounds of this invention may be particularly useful for the treatment of inflammatory bowel disease (including Crohn's disease and ulcerative colitis), psoriasis, retinal detachment, retinitis pigmentosa, arthritis (including rheumatoid arthritis, spondylarthritis, gout, and SoJIA), transplant rejection, ischemia reperfusion injury of solid organs, multiple sclerosis, and/or tumor necrosis factor receptor-associated periodic syndrome. More specifically, the compounds of this invention may be particularly useful for the treatment of inflammatory bowel disease (including Crohn's disease and ulcerative colitis), psoriasis, retinal detachment, retinitis pigmentosa, arthritis (including rheumatoid arthritis, spondyloarthritis, gout, and systemic onset juvenile idiopathic arthritis
(SoJIA)),transplant rejection, and/or ischemia reperfusion injury of solid organs.
Treatment of RIP 1 -mediated disease conditions, or more broadly, treatment of immune mediated disease, such as, but not limited to, allergic diseases, autoimmune diseases, prevention of transplant rejection and the like, may be achieved using a compound of this invention as a monotherapy, or in dual or multiple combination therapy, particularly for the treatment of refractory cases, such as in combination with other anti-inflammatory and/or anti-TNF agents, which may be administered in therapeutically effective amounts as is known in the art.
The compounds of any one of Formulas (I-IV) and pharmaceutically acceptable salts thereof may be employed alone or in combination with other therapeutic agents.
Combination therapies according to the present invention thus comprise the administration of at least one compound of any one of Formulas (I-IV) or a pharmaceutically acceptable salt thereof, and at least one other theraputically active agent. Preferably, combination therapies according to the present invention comprise the administration of at least one compound of any one of Formulas (I-IV) or a pharmaceutically acceptable salt thereof, and at least one other therapeutically active agent. The compound(s) of any one of Formulas (I-IV) and pharmaceutically acceptable salts thereof, and the other therapeutically active agent(s) may be administered together in a single pharmaceutical composition or separately and, when administered separately this may occur simultaneously or sequentially in any order. The amounts of the compound(s) of any one of Formulas (I-IV) and
pharmaceutically acceptable salts thereof, and the other therapeutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect. Thus in a further aspect, there is provided a combination comprising a compound of any one of Formulas (I-IV) or a pharmaceutically acceptable salt thereof, together with one or more other therapeutically active agents. In one aspect, there is provided a combination comprising (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide, or a
pharmaceutically acceptable salt thereof, together with one or more other therapeutically active agents. In another aspect, there is provided a combination comprising (S)-5-benzyl- N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3- carboxamide, or a pharmaceutically acceptable salt thereof, together with one or more other therapeutically active agents. Thus in one aspect of this invention, a compound of any one of Formulas (I-IV) or a pharmaceutically acceptable salt thereof, and a pharmaceutical composition comprising a compound of any one of Formulas (I-IV) or a pharmaceutically acceptable salt thereof, may be used in combination with or include one or more other therapeutic agents, for example an anti-inflammatory agent and/or an anti-TNF agent.
For example, the compounds of this invention may be administered in combination with other anti-inflammatory agents for any of the indications above, including oral or topical corticosteroids (such as prednisone (Deltasone®) and bundesonide), anti-TNF agents (including anti-TNF biologic agents), 5-aminosalicyclic acid and mesalamine preparations, hydroxycloroquine, thiopurines (azathioprin, mercaptopurin ), methotrexate, cyclophosphamide, cyclosporine, JAK inhibitors (tofacitinib), anti-IL6 biologies, anti-ILl or IL 12 or IL23 biologics(ustekinumab(Stelara®)), anti-integrin agents(natalizumab
(Tysabri®)), anti-CD20 or CD4 biologies and other cytokine inhibitors or biologies to T- cell or B-cell receptors or interleukins.
The compounds of this invention may be administered in combination with other anti-inflammatory agents for any of the indications above, including oral or topical corticosteroids (such as prednisone (Deltasone®) and bundesonide), anti-TNF agents
(including anti-TNF biologic agents), 5-aminosalicyclic acid and mesalamine preparations, hydroxycloroquine, thiopurines (azathioprin, mercaptopurin ), methotrexate,
cyclophosphamide, cyclosporine, calcineurin inhibitors (cyclosporine, pimecrolimus, tacrolimus), mycophenolic acid (CellCept®), mTOR inhibitors (temsirolimus, everolimus), JAK inhibitors (tofacitinib), (Xeljan®)), Syk inhibitors (fostamatinib), anti-IL6 biologies, anti-ILl (anakinra (Kineret®), canakinumab (Ilaris®), rilonacept (Arcalyst®)), anti-IL12 and IL23 biologies (ustekinumab(Stelara®)), anti-ILl 7 biologies (secukinumab), anti- CD22 (epratuzumab), anti-integrin agents(natalizumab (Tysabri®)), vedolizumab
(Entyvio®)), anti-IFNa (sifalimumab), anti-CD20 or CD4 biologies and other cytokine inhibitors or biologies to T-cell or B-cell receptors or interleukins.
Examples of suitable anti-inflammatory biologic agents include Actemra® (anti- IL6R mAb), anti-CD20 mAbs (rituximab (Rituxan®) and ofatumumab (Arzerra®)), abatacept (Orencia®), anakinra (Kineret®), ustekinumab (Stelara®), and belimumab (Benlysta®). Examples of other suitable anti-inflammatory biologic agents include Actemra® (tocilizumab, anti-IL6R mAb), anti-CD20 mAbs (rituximab (Rituxan®) and ofatumumab (Arzerra®)), abatacept (Orencia®), anakinra (Kineret®), Canakinumab (Ilaris®), rilonacept (Arcalyst®), secukinumab, epratuzumab, sifalimumab, ustekinumab (Stelara®), and belimumab (Benlysta®). Examples of suitable anti-TNF agents biologic agents include etanecerpt (Enbrel®), adalimumab (Humira®), infliximab (Remicade®), certolizumab (Cimzia®), and golimumab (Simponi®).
Accordingly, one embodiment of this invention is directed to a method of inhibiting RIP1 kinase comprising contacting a cell with a compound of the invention. In another embodiment, the invention is directed to a method of treating a RIP1 kinase-mediated disease or disorder (specifically, a disease or disorder recited herein) comprising
administering a therapeutically effective amount of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a salt, particularly a pharmaceutically acceptable salt thereof, to a human in need thereof.
In one specific embodiment, the invention is directed to a method of treating a RIPl kinase-mediated disease or disorder (specifically, a disease or disorder recited herein) comprising administering a therapeutically effective amount of (S)-5-benzyl-N-(5-methyl- 4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to a human in need thereof. In another specific embodiment, the invention is directed to a method of treating a RIPl kinase-mediated disease or disorder (specifically, a disease or disorder recited herein) comprising
administering a therapeutically effective amount of (S)-5-benzyl-N-(7-chloro-2-oxo- 2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to a human in need thereof.
Specifically, this invention provides a compound of the invention for use in therapy.
This invention also provides a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a pharmaceutically acceptable salt thereof, for use in therapy. This invention particularly provides a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a pharmaceutically acceptable salt thereof, for use in the treatment of a RIPl kinase-mediated disease or disorder (for example, a disease or disorder recited herein).
Specifically, this invention provides a compound described herein, or a
pharmaceutically acceptable salt thereof, for use in therapy. More specifically, this invention provides (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide, or a
pharmaceutically acceptable salt thereof, for use in therapy. This invention further provides (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4- triazole-3-carboxamide, or a pharmaceutically acceptable salt thereof, for use in therapy.
In another embodiment, this invention provides a compound of the invention for use in the treatment of a RIPl kinase-mediated disease or disorder. Specifically, this invention provides a compound described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment of a RIPl kinase-mediated disease or disorder. In another embodiment, this invention provides a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a pharmaceutically acceptable salt thereof, for use in the treatment of diseases/disorders which are likely to be regulated at least in part by programmed necrosis, apoptosis or the production of inflammatory cytokines, particularly inflammatory bowel disease (including Crohn's disease and ulcerative colitis), psoriasis, retinal detachment, retinitis pigmentosa, macular degeneration, pancreatitis, atopic dermatitis, arthritis
(including rheumatoid arthritis, spondyloarthritis, gout, systemic onset juvenile idiopathic arthritis (SoJIA)), systemic lupus erythematosus (SLE), Sjogren's syndrome, systemic scleroderma, anti-phospholipid syndrome (APS), vasculitis, osteoarthritis, liver
damage/diseases (non-alcohol steatohepatitis, alcohol steatohepatitis, autoimmune hepatitis, autoimmune hepatobiliary diseases, primary sclerosing cholangitis (PSC)), nephritis, Celiac disease, autoimmune idiopathic thrombocytopenic purpura (autoimmune ITP), transplant rejection, ischemia reperfusion injury of solid organs, sepsis, systemic inflammatory response syndrome (SIRS), cerebrovascular accident (CVA, stroke), myocardial infarction (MI), atherosclerosis, Huntington's disease, Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis (ALS), allergic diseases (including asthma and atopic dermatitis), multiple sclerosis, type I diabetes, Wegener's granulomatosis, pulmonary sarcoidosis, Behcet's disease, interleukin-1 converting enzyme (ICE, also known as caspase-1) associated fever syndrome, chronic obstructive pulmonary disease (COPD), tumor necrosis factor receptor-associated periodic syndrome (TRAPS), peridontitis,
EMO-deficiency syndrome ( F-kappa-B essential modulator gene (also known as IKK gamma or IKKG) deficiency syndrome), HOIL-1 deficiency ((also known as RBCK1) heme-oxidized IRP2 ubiquitin ligase-1 deficiency), linear ubiquitin chain assembly complex (LUBAC) deficiency syndrome, hematological and solid organ malignancies, bacterial infections and viral infections (such as tuberculosis and influenza), and Lysosomal storage diseases (particularly, Gaucher Disease, and including GM2 Gangliosidosis, Alpha- mannosidosis, Aspartylglucosaminuria, Cholesteryl Ester storage disease, Chronic
Hexosaminidase A Deficiency, Cystinosis, Danon disease, Fabry disease, Farber disease, Fucosidosis, Galactosialidosis, GM1 gangliosidosis, Mucolipidosis, Infantile Free Sialic Acid Storage Disease, Juvenile Hexosaminidase A Deficiency, Krabbe disease, Lysosomal acid lipase deficiency, Metachromatic Leukodystrophy, Mucopolysaccharidoses disorders, Multiple sulfatase deficiency, Niemann-Pick Disease, Neuronal Ceroid Lipofuscinoses, Pompe disease, Pycnodysostosis, Sandhoff disease, Schindler disease, Sialic Acid Storage Disease, Tay-Sachs and Wolman disease), wherein treatment of the above-noted diseases/disorders may concern, more specifically, the amelioration of organ injury or damage sustained as a result of the noted diseases.
In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of inflammatory bowel disease. In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of Crohn's disease. In another embodiment, this invention provides a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, for use in the treatment of ulcerative colitis. In another embodiment, this invention provides a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, for use in the treatment of psoriasis. In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of retinal detachment. In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of retinitis pigmentosa. In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of arthritis. In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of rheumatoid arthritis. In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of spondylarthritis. In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of gout. In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of systemic onset juvenile idiopathic arthritis. In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of transplant rejection. In another embodiment, this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of ischemia reperfusion injury of solid organs.
This invention specifically provides for the use of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a pharmaceutically acceptable salt thereof, as an active therapeutic substance. More specifically, this provides for the use of the compounds described herein for the treatment of a RIPl kinase-mediated disease or disorder. Accordingly, the invention provides for the use of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a pharmaceutically acceptable salt thereof, as an active therapeutic substance in the treatment of a human in need thereof with a RIPl kinase-mediated disease or disorder.
The invention further provides for the use of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a salt thereof, particularly a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of a RIPl kinase-mediated disease or disorder, for example the diseases and disorders recited herein. Specifically, the invention also provides for the use of a compound described herein, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of a RIPl kinase-mediated disease or disorder, for example the diseases and disorders recited herein. More specifically, the invention also provides for the use of (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b] [ 1 ,4]oxazepin-3 -yl)-4H- 1 ,2,4-triazole-3 -carboxamide, or a
pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of a RIPl kinase-mediated disease or disorder, for example the diseases and disorders recited herein. The invention further provides for the use of (S)-5-benzyl-N-(7- chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3- carboxamide, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of a RIPl kinase-mediated disease or disorder, for example the diseases and disorders recited herein. Accordingly, the invention provides for the use of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of a human in need thereof with a RIPl kinase-mediated disease or disorder.
In one embodiment, this invention provides for the use of a compound of Formula
(I), particularly a compound of any one of Formulas (I-IV), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of diseases/disorders which are likely to be regulated at least in part by programmed necrosis, apoptosis or the production of inflammatory cytokines, particularly inflammatory bowel disease (including Crohn's disease and ulcerative colitis), psoriasis, retinal detachment, retinitis pigmentosa, macular degeneration, pancreatitis, atopic dermatitis, arthritis (including rheumatoid arthritis, spondyloarthritis, gout, systemic onset juvenile idiopathic arthritis (SoJIA)), systemic lupus erythematosus (SLE), Sjogren's syndrome, systemic scleroderma, anti-phospholipid syndrome (APS), vasculitis, osteoarthritis, liver
damage/diseases (non-alcohol steatohepatitis, alcohol steatohepatitis, autoimmune hepatitis, autoimmune hepatobiliary diseases, primary sclerosing cholangitis (PSC)), nephritis, Celiac disease, autoimmune idiopathic thrombocytopenic purpura (autoimmune ITP), transplant rejection, ischemia reperfusion injury of solid organs, sepsis, systemic inflammatory response syndrome (SIRS), cerebrovascular accident (CVA, stroke), myocardial infarction (MI), atherosclerosis, Huntington's disease, Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis (ALS), allergic diseases (including asthma and atopic dermatitis), multiple sclerosis, type I diabetes, Wegener's granulomatosis, pulmonary sarcoidosis, Behcet's disease, interleukin-1 converting enzyme (ICE, also known as caspase-1) associated fever syndrome, chronic obstructive pulmonary disease (COPD), tumor necrosis factor receptor-associated periodic syndrome (TRAPS), peridontitis,
EMO-deficiency syndrome ( F-kappa-B essential modulator gene (also known as IKK gamma or IKKG) deficiency syndrome), HOIL-1 deficiency ((also known as RBCK1) heme-oxidized IRP2 ubiquitin ligase-1 deficiency), linear ubiquitin chain assembly complex (LUBAC) deficiency syndrome, hematological and solid organ malignancies, bacterial infections and viral infections (such as tuberculosis and influenza), and Lysosomal storage diseases (particularly, Gaucher Disease, and including GM2 Gangliosidosis, Alpha- mannosidosis, Aspartylglucosaminuria, Cholesteryl Ester storage disease, Chronic
Hexosaminidase A Deficiency, Cystinosis, Danon disease, Fabry disease, Farber disease, Fucosidosis, Galactosialidosis, GM1 gangliosidosis, Mucolipidosis, Infantile Free Sialic Acid Storage Disease, Juvenile Hexosaminidase A Deficiency, Krabbe disease, Lysosomal acid lipase deficiency, Metachromatic Leukodystrophy, Mucopolysaccharidoses disorders, Multiple sulfatase deficiency, Niemann-Pick Disease, Neuronal Ceroid Lipofuscinoses, Pompe disease, Pycnodysostosis, Sandhoff disease, Schindler disease, Sialic Acid Storage Disease, Tay-Sachs and Wolman disease), wherein treatment of the above-noted
diseases/disorders may concern, more specifically, the amelioration of organ injury or damage sustained as a result of the noted diseases.
In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of inflammatory bowel disease. In another
embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of Crohn's disease. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the
manufacture of a medicament for use in the treatment of ulcerative colitis. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of psoriasis. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of retinal detachment. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of retinitis pigmentosa. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of arthritis. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of rheumatoid arthritis. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of spondyloarthritis. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of gout. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of systemic onset juvenile idiopathic arthritis. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of transplant rejection. In another embodiment, this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in the treatment of ischemia reperfusion injury of solid organs.
A therapeutically "effective amount" is intended to mean that amount of a compound that, when administered to a patient in need of such treatment, is sufficient to effect treatment, as defined herein. Thus, e.g., a therapeutically effective amount of a compound of Formula (I), particularly a compound of any one of Formulas (I-IV), or a pharmaceutically acceptable salt thereof, is a quantity of an inventive agent that, when administered to a human in need thereof, is sufficient to modulate and/or inhibit the activity of RIP 1 kinase such that a disease condition which is mediated by that activity is reduced, alleviated or prevented. The amount of a given compound that will correspond to such an amount will vary depending upon factors such as the particular compound (e.g., the potency (pIC5o), efficacy (EC50), and the biological half-life of the particular compound), disease condition and its severity, the identity (e.g., age, size and weight) of the patient in need of treatment, but can nevertheless be routinely determined by one skilled in the art. Likewise, the duration of treatment and the time period of administration (time period between dosages and the timing of the dosages, e.g., before/with/after meals) of the compound will vary according to the identity of the mammal in need of treatment (e.g., weight), the particular compound and its properties (e.g., pharmacokinetic properties), disease or disorder and its severity and the specific composition and method being used, but can nevertheless be determined by one of skill in the art.
"Treating" or "treatment" is intended to mean at least the mitigation of a disease or disorder in a patient. The methods of treatment for mitigation of a disease or disorder include the use of the compounds in this invention in any conventionally acceptable manner, for example for prevention, retardation, prophylaxis, therapy or cure of a RIPl kinase-mediated disease or disorder, as described hereinabove.
The compounds of the invention may be administered by any suitable route of administration, including both systemic administration and topical administration.
Systemic administration includes oral administration, parenteral administration, transdermal administration, rectal administration, and administration by inhalation.
Parenteral administration refers to routes of administration other than enteral, transdermal, or by inhalation, and is typically by injection or infusion. Parenteral administration includes intravenous, intramuscular, and subcutaneous injection or infusion. Inhalation refers to administration into the patient's lungs whether inhaled through the mouth or through the nasal passages. Topical administration includes application to the skin.
The compounds of the invention may be administered once or according to a dosing regimen wherein a number of doses are administered at varying intervals of time for a given period of time. For example, doses may be administered one, two, three, or four times per day. Doses may be administered until the desired therapeutic effect is achieved or indefinitely to maintain the desired therapeutic effect. Suitable dosing regimens for a compound of the invention depend on the pharmacokinetic properties of that compound, such as absorption, distribution, and half-life, which can be determined by the skilled artisan. In addition, suitable dosing regimens, including the duration such regimens are administered, for a compound of the invention depend on the disease or disorder being treated, the severity of the disease or disorder being treated, the age and physical condition of the patient being treated, the medical history of the patient to be treated, the nature of concurrent therapy, the desired therapeutic effect, and like factors within the knowledge and expertise of the skilled artisan. It will be further understood by such skilled artisans that suitable dosing regimens may require adjustment given an individual patient's response to the dosing regimen or over time as individual patient needs change. Total daily dosages range from 1 mg to 2000 mg, preferably, total daily dosages range from 1 mg to 250 mg.
For use in therapy, the compounds of the invention will be normally, but not necessarily, formulated into a pharmaceutical composition prior to administration to a patient. Accordingly, the invention also is directed to pharmaceutical compositions comprising a compound of the invention and one or more pharmaceutically acceptable excipients.
In one embodiment, there is provided a pharmaceutical composition comprising (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4- triazole-3-carboxamide (free base), and one or more pharmaceutically acceptable excipients. In another embodiment, there is provided a pharmaceutical composition comprising (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3- yl)-4H-l,2,4-triazole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipients. In another embodiment, there is provided a pharmaceutical composition comprising crystalline (S)-5-benzyl-N-(5-methyl-4-oxo- 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4-triazole-3-carboxamide (free base) having the PXRD pattern of Figure 7 and one or more pharmaceutically acceptable excipients. In another embodiment, there is provided a pharmaceutical composition comprising crystalline (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b] [ 1 ,4]oxazepin-3 -yl)-4H- 1 ,2,4-triazole-3 -carboxamide (free base) characterized by the diffraction data in Table 1, and one or more pharmaceutically acceptable excipients. In one embodiment, there is provided a pharmaceutical composition comprising (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)- 4H-l,2,4-triazole-3 -carboxamide (free base) and one or more pharmaceutically acceptable excipients. In another embodiment, there is provided a pharmaceutical composition comprising (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)- 4H-l,2,4-triazole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipients.
The pharmaceutical compositions of the invention may be prepared and packaged in bulk form wherein an effective amount of a compound of the invention can be extracted and then given to the patient such as with powders, syrups, and solutions for injection. Alternatively, the pharmaceutical compositions of the invention may be prepared and packaged in unit dosage form. For oral application, for example, one or more tablets or capsules may be administered. A dose of the pharmaceutical composition contains at least a therapeutically effective amount of a compound of this invention (i.e., a compound of
Formula (I), particularly a compound of any one of Formulas (I-IV), or a salt, particularly a pharmaceutically acceptable salt, thereof). When prepared in unit dosage form, the pharmaceutical compositions may contain from 1 mg to 1000 mg of a compound of this invention.
As provided herein, unit dosage forms (pharmaceutical compositions) containing from 1 mg to 1000 mg of a compound of the invention may be administered one, two, three, or four times per day, preferably one, two, or three times per day, and more preferably, one or two times per day, to effect treatment of a RIPl kinase-mediated disease or disorder.
The pharmaceutical compositions of the invention typically contain one compound of the invention. However, in certain embodiments, the pharmaceutical compositions of the invention contain more than one compound of the invention. In addition, the pharmaceutical compositions of the invention may optionally further comprise one or more additional pharmaceutically active compounds.
As used herein, "pharmaceutically acceptable excipient" means a material, composition or vehicle involved in giving form or consistency to the composition. Each excipient must be compatible with the other ingredients of the pharmaceutical composition when commingled such that interactions which would substantially reduce the efficacy of the compound of the invention when administered to a patient and interactions which would result in pharmaceutical compositions that are not pharmaceutically acceptable are avoided. In addition, each excipient must of course be of sufficiently high purity to render it pharmaceutically acceptable. The compounds of the invention and the pharmaceutically acceptable excipient or excipients will typically be formulated into a dosage form adapted for administration to the patient by the desired route of administration. Conventional dosage forms include those adapted for (1) oral administration such as tablets, capsules, caplets, pills, troches, powders, syrups, elixirs, suspensions, solutions, emulsions, sachets, and cachets; (2) parenteral administration such as sterile solutions, suspensions, and powders for reconstitution; (3) transdermal administration such as transdermal patches; (4) rectal administration such as suppositories; (5) inhalation such as aerosols and solutions; and (6) topical administration such as creams, ointments, lotions, solutions, pastes, sprays, foams, and gels.
Suitable pharmaceutically acceptable excipients will vary depending upon the particular dosage form chosen. In addition, suitable pharmaceutically acceptable excipients may be chosen for a particular function that they may serve in the composition. For example, certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the production of uniform dosage forms. Certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the production of stable dosage forms. Certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the carrying or transporting the compound or compounds of the invention once administered to the patient from one organ, or portion of the body, to another organ, or portion of the body. Certain pharmaceutically acceptable excipients may be chosen for their ability to enhance patient compliance.
Suitable pharmaceutically acceptable excipients include the following types of excipients: diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweeteners, flavoring agents, flavor masking agents, coloring agents, anti-caking agents, humectants, chelating agents, plasticizers, viscosity increasing agents, antioxidants, preservatives, stabilizers, surfactants, and buffering agents. The skilled artisan will appreciate that certain pharmaceutically acceptable excipients may serve more than one function and may serve alternative functions depending on how much of the excipient is present in the formulation and what other ingredients are present in the formulation.
Skilled artisans possess the knowledge and skill in the art to enable them to select suitable pharmaceutically acceptable excipients in appropriate amounts for use in the invention. In addition, there are a number of resources that are available to the skilled artisan which describe pharmaceutically acceptable excipients and may be useful in selecting suitable pharmaceutically acceptable excipients. Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), The Handbook of Pharmaceutical Additives (Gower Publishing Limited), and The Handbook of Pharmaceutical Excipients (the American Pharmaceutical Association and the Pharmaceutical Press).
The pharmaceutical compositions of the invention are prepared using techniques and methods known to those skilled in the art. Some of the methods commonly used in the art are described in Remington's Pharmaceutical Sciences (Mack Publishing Company). Accordingly, another embodiment of this invention is a method of preparing a
pharmaceutical composition comprising the step of admixing crystalline (S)-5-benzyl-N- (5-methyl-4-oxo-2,3 ,4,5-tetrahydrobenzo[b] [ 1 ,4]oxazepin-3 -yl)-4H- 1 ,2,4-triazole-3 - carboxamide (free base) having the PXRD pattern of Figure 7 with one or more pharmaceutically acceptable excipients. In another embodiment, there is provided a method of preparing a pharmaceutical composition comprising the step of admixing crystalline (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)- 4H-l,2,4-triazole-3 -carboxamide (free base) characterized by the diffraction data in Table 1, with one or more pharmaceutically acceptable excipients.
In one aspect, the invention is directed to a solid oral dosage form such as a tablet or capsule comprising an effective amount of a compound of the invention and a diluent or filler. Suitable diluents and fillers include lactose, sucrose, dextrose, mannitol, sorbitol, starch (e.g. corn starch, potato starch, and pre-gelatinized starch), cellulose and its derivatives (e.g. microcrystalline cellulose), calcium sulfate, and dibasic calcium
phosphate. The oral solid dosage form may further comprise a binder. Suitable binders include starch (e.g. corn starch, potato starch, and pre-gelatinized starch), gelatin, acacia, sodium alginate, alginic acid, tragacanth, guar gum, povidone, and cellulose and its derivatives (e.g. microcrystalline cellulose). The oral solid dosage form may further comprise a disintegrant. Suitable disintegrants include crospovidone, sodium starch glycolate, croscarmelose, alginic acid, and sodium carboxymethyl cellulose. The oral solid dosage form may further comprise a lubricant. Suitable lubricants include stearic acid, magnesium stearate, calcium stearate, and talc.
EXAMPLES
The following examples illustrate the invention. These examples are not intended to limit the scope of the present invention, but rather to provide guidance to the skilled artisan to prepare and use the compounds, compositions, and methods of the present invention. While particular embodiments of the present invention are described, the skilled artisan will appreciate that various changes and modifications can be made without departing from the spirit and scope of the invention.
The reactions described herein are applicable for producing compounds of the invention having a variety of different substituent groups (e.g., R1, R2, etc.), as defined herein. The skilled artisan will appreciate that if a particular substituent is not compatible with the synthetic methods described herein, the substituent may be protected with a suitable protecting group that is stable to the reaction conditions. The protecting group may be removed at a suitable point in the reaction sequence to provide a desired intermediate or target compound. Suitable protecting groups and the methods for protecting and de- protecting different substituents using such suitable protecting groups are well known to those skilled in the art; examples of which may be found in T. Greene and P. Wuts,
Protecting Groups in Chemical Synthesis (3rd ed.), John Wiley & Sons, NY (1999).
Names for the intermediate and final compounds described herein were generated using the software naming program ACD/Name Pro V6.02 available from Advanced Chemistry Development, Inc., 110 Yonge Street, 14th Floor, Toronto, Ontario, Canada, M5C 1T4 (http://www.acdlabs.com/) or the naming program in ChemDraw, Struct=Name Pro 12.0, as part of ChemBioDraw Ultra, available from CambridgeSoft. 100
CambridgePark Drive, Cambridge, MA 02140 USA (www.cambridgesoft.com).
It will be appreciated by those skilled in the art that in certain instances these programs may name a structurally depicted compound as a tautomer of that compound. It is to be understood that any reference to a named compound or a structurally depicted compound is intended to encompass all tautomers of such compounds and any mixtures of tautomers thereof.
EXAMPLES
In the following experimental descriptions, the following abbreviations may be used:
Figure imgf000056_0001
BuOH butanol
CDCI3 deuterated chloroform
CDI 1 , 1 '-carbony ldiimidazole
CH2C12 or DCM methylene chloride or dichloromethane
CH3CN or MeCN acetonitrile
CH3NH2 methylamine
d day
DAST diethylaminosulfur trifluoride
DCE 1 ,2-dichloroethane
DCM 1 ,2-dichloromethane
DIEA or DIPEA diisopropyl ethylamine
DMA dimethy lacetamide
DMAP 4-Dimethylaminopyridine
DMF N, Λ^-dimethy lformamide
DMSO dimethy lsulfoxide
EDC 1 -ethy 1-3 -(3 -dimethy laminopropy 1) carbodiimide equiv equivalents
Et ethyl
Et3N or TEA triethylamine
Et20 diethyl ether
EtOAc ethyl acetate
FCC flash column chromatography
h, hr hour(s)
HATU ^-(T-Azabenzotriazol-ly^-A'.A'.iV'.iV'-tetramethylyronium hexafluorophosphate
HC1 hydrochloric acid
HPLC high-performance liquid chromatography
IC1 iodine monochloride
/'-Pr2NEt N ', N '-diisopropylethy lamine
KOi-Bu potassium ter/-butoxide
KOH potassium hydroxide
LCMS liquid chromatography -mass spectroscopy
LiHDMS lithium hexamethyldisilazide
LiOH lithium hydroxide
Me methyl
MeOH or CH3OH methanol
MgS04 magnesium sulfate
min minute(s)
MS mass spectrum μ\ν microwave
NaBH4 sodium borohydride
Na2C03 sodium carbonate
NaHC03 sodium bicarbonate
NaOH sodium hydroxide
Na2S04 sodium sulfate
NBS N-Bromosuccinimide
N2H2 hydrazine
NH4CI ammonium chloride
NH4OH ammonium hydroxide
NiCl2 «6H20 nickel (II) chloride hexahydrate
NMP iV-methyl-2-pyrrolidone
NMR nuclear magnetic resonance
Pd/C palladium on carbon
Ph phenyl
POCI3 phosphoryl chloride
PSI pound-force per square inch
rm or rxn mixture reaction mixture
rt room temperature
satd. saturated
sm starting material
TEA thiethylamine
TFA trifluoroacetic acid
THF tetrahydrofuran
TMEDA tetramethylethylenediamine
TMSI trimethylsilyl iodide
TMSN3 trimethylsilyl azide
T3P 2,4,6-tripropyl-l,3,5,2,4,6-trioxatriphosphorinane-2,4,6-trioxide
¾or Rf retention time
Preparation 1
(S)-2-((tert-butoxycarbonyl)amino)-3-(2-nitrophenoxy)propanoic acid
Figure imgf000058_0001
To a suspension of sodium hydride (9.75 g, 244 mmol) in DMF (250 mL) was added a solution of (S)-2-((tert-butoxycarbonyl)amino)-3-hydroxypropanoic acid (25 g, 122 mmol) in 50 mL of DMF dropwise over 10 min at 0°C. Vigorous gas evolution was observed. Once gas evolution had ceased, l-fluoro-2-nitrobenzene (12.85 mL, 122 mmol) was added dropwise neat at 0°C.The reaction mixture was allowed to stir at room temperature for 16 hr. The reaction mixture was partitioned between ethyl acetate (1000 mL) and 0.5 M HC1 solution (1000 mL). The layers were separated, the organic layer was washed with water (3 x 400 ml), brine (400 mL), and concentrated under reduced pressure to provide the crude product. The crude compound was purified by silica gel column using 0-10% MeOH in DCM to afford (S)-2-((tert-butoxycarbonyl)amino)-3-(2- nitrophenoxy)propanoic acid (32 g, 76 mmol, 62.3 % yield) as redish yellow semi solid. 1H MR (400 MHz, CDC13) d ppm 7.88 (dd, J=8.46, 1.64 Hz, 1 H), 7.52 - 7.61 (m, 1 H), 7.06 - 7.15 (m, 2 H), 5.68 (br. d., 1 H), 4.75 (br. s., 1 H), 4.60 - 4.72 (m, 1 H), 2.07 (s, 2 H), 1.48 (s, 9 H). MS (m/z) 325.13 (M-H+).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000060_0002
The following intermediates used for the preparation of titled example compounds were synthesized using (S)-3-amino-2-((tert-butoxycarbonyl)amino)-propanoic acid as described by Scott B. Hoyt et. al. in patent application WO 2008/106077.
Figure imgf000060_0003
Preparation 2
,-aminophenoxy)-2-((tert-butoxycarbonyl)amino)propanoic acid
Figure imgf000061_0001
A solution of (S)-2-((tert-butoxycarbonyl)amino)-3-(2-nitrophenoxy)propanoic acid (32 g, 98 mmol) and Pd/C (2.82 g, 2.65 mmol) in methanol (500 mL) using Parr apparatus was hydrogenated with 60 PSI at rt for 2 hr. The TLC (10% MeOH in DCM; Rf: 0.4) shows complete conversion of all of the starting material and the reaction mixture was filtered through a celite bed. The celite bed was washed with methanol (130 mL, 3 times) and the combined filtrate was concentrated to afford (S)-3-(2-aminophenoxy)-2-((tert- butoxycarbonyl)amino)propanoic acid (32 g, 95 mmol, 97 % yield) as pale brown semi solid. The residue was used in the next step without further purification. 1H MR
(DMSO-de) δ: 7.42 (br. s., 1H), 6.74 (d, J = 7.1 Hz, 1H), 6.64 - 6.70 (m, 1H), 6.57 - 6.62 (m, 1H), 6.47 (td, J = 7.6, 1.6 Hz, 1H), 4.40 (d, J = 4.3 Hz, 1H), 4.24 (dd, J = 9.5, 4.9 Hz, 1H), 4.00 (dd, J = 9.6, 3.5 Hz, 1H), 1.41 (s, 9H). MS (m/z) 295.19 (M-H+), 222.15 (- tBuO group).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000062_0001
Preparation 3
(S)-tert-butyl (4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate
Figure imgf000063_0001
To a solution of (S)-3-(2-aminophenoxy)-2-((tert-butoxycarbonyl)amino)propanoic acid (23 g, 78 mmol) and DIPEA (14.91 mL, 85 mmol) in DMSO (230 mL) stirred under nitrogen at 10°C was added HATU (29.5 g, 78 mmol) portionwise during 15 min. The reaction mixture was stirred at room temperature for 16 hr. Reaction was quenched with water (900 mL) (resulting in formation of a solid) and was stirred at 18°C for 20 min. The resultant solid was filtered, washed with excess water (3 times) and dried in vacuo (high vacuum) to afford (S)-tert-butyl (4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3- yl)carbamate (18g, 61.6 mmol, 79 % yield) as a pale brown solid. TLC: 50% EtOAc in Hexane; Rf: 0.55. 1H MR (400 MHz, DMSO-^6) d ppm 9.92 (s, 1 H), 6.99 - 7.21 (m, 5 H), 4.17 - 4.45 (m, 3 H), 1.36 (s, 9 H). MS (m/z) : 179.16 ([M-BOC] + H+).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000063_0002
Figure imgf000064_0001
Preparation 4
(S)-tert-butyl (7-fluoro-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3- yl)carbamate
Figure imgf000064_0002
To a suspension of (S)-tert-butyl (7-fluoro-4-oxo-2, 3,4,5 - tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate (0.89g, 2.70 mmol) and K2C03 (0.392 g, 2.84 mmol) in DMF ( 10.0 mL) at rt was added a solution of Mel (0.161 mL, 2.57 mmol) in DMF. The reaction mixture was stirred at rt overnight, then additional 0.74 eq of Mel and K2C03 were added and the reaction was monitored by LCMS. The reaction mixture was diluted with EtOAc then washed with water (2x), sat. aq. H4CI and brine. The organic phase was concentrated in vacuo then purified by FCC [EtO Ac/Hex: 15-50%] to yield the desired product (640 mg, 76%). 1H MR (DMSO-d6) δ: 7.44 (dd, J = 9.9, 3.0 Hz, 1H), 7.17 - 7.25 (m, 2H), 7.10 (td, J = 8.5, 3.0 Hz, 1H), 4.23 - 4.42 (m, 3H), 3.28 (s, 3H), 1.35 (s, 9H). MS (m/z) 211.1 ([M-BOC] + H+).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the one described above.
Figure imgf000065_0001
Preparation 5
(S)-tert-butyl (5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate
Figure imgf000065_0002
To a solution of (S)-tert-butyl (4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3- yl)carbamate (30 g, 108 mmol) and Cs2C03 (49.2 g, 151 mmol) in DMF (300 mL) stirred under nitrogen at room temp was added methyl iodide (8.09 mL, 129 mmol) dropwise during 15 min. The reaction mixture was stirred at rt for 16 hr. TLC (30% EtOAc in
Hexane; Rf: 0.4) shows reaction was complete. The reaction was poured into cold water (1500 mL) which formed a solid, the resultant solid was filtered, the filter cake was washed with water (two times) and dried in vacuo to afford the crude compound. This was triturated with 5% Et20 in hexane (300 mL) to afford (S)-tert-butyl (5-methyl-4-oxo- 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate (19g, 62.7 mmol, 58.1 % yield) as a brown solid. 1H MR (DMSO-d6) δ: 7.47 (dd, J = 7.7, 1.6 Hz, 1H), 7.23 - 7.33 (m, 2H), 7.14 - 7.21 (m, 2H), 4.25 - 4.41 (m, 3H), 3.28 (s, 3H), 1.34 (s, 9H). MS (m/z) 193.33 ([M- BOC] + H+).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the one described above.
Figure imgf000066_0001
Preparation 6
(S)-3-amino-7-fluoro-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one trifluoroacetate
Figure imgf000066_0002
To a suspension of (S)-tert-butyl (7-fluoro-4-oxo-2, 3,4,5 - tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate (105.0 mg, 0.354 mmol) in DCM (1.5 mL) was added TFA (0.191 mL, 2.481 mmol). The reaction became homogeneous almost immediately and was allowed to stir at rt and monitored by LCMS. (about 2h). The reaction was diluted with ethyl ether then concentrated under reduced pressure (repeated 3 times) to give the desired product as a TFA salt. The sample was once azeotroped with toluene. Quantitative recovery was assumed.
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the one described above.
Figure imgf000066_0003
Figure imgf000067_0001
Preparation 7
4 5-dihydro-lH-benzo[b]azepin-2(3H)-one
Figure imgf000067_0002
To a solution of 3,4-dihydronaphthalen-l(2H)-one (4.55 mL, 34.2 mmol) in methanesulfonic acid (40 mL) cooled in an ice/brine bath was added sodium azide (2.5 g, 38.5 mmol) in 5 portions over 15 minutes. Moderate gas evolution. Mixture was stirred cooled for 15 minutes, then at rt for 30 minutes. Reaction was poured over ice and stirred for 10 minutes. The resulting solid was filtered, rinsed with water and hexanes, and dried to give 6.10 g tan solid. 1H MR (DMSO-d6) δ: 9.51 (br. s., 1H), 7.16 - 7.29 (m, 2H), 7.04 - 7.12 (m, 1H), 6.96 (d, J = 7.6 Hz, 1H), 2.68 (t, J = 6.8 Hz, 2H), 2.04 - 2.19 (m, 4H); MS (m/z) 162.0 (M+H+).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000068_0001
Preparation 8
3-iodo-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one
Figure imgf000068_0002
To a mixture of 4,5-dihydro-lH-benzo[b]azepin-2(3H)-one (10.6 g, 65.8 mmol) in DCM (150 mL) cooled in an ice/water bath was added TMEDA (29.8 mL, 197 mmol), then dropwise over 20 minutes was added TMSI (26.9 mL, 197 mmol). The mixture was stirred cooled for 60 minutes, iodine (25.03 g, 99 mmol) was added and the mixture was stirred cooled for another 60 minutes. The reaction was quenched with 5% Na2S2C"3 and stirred
15 minutes. The resulting solid was filtered and dried to give 11.3 g tan solid. Layers of filtrate were separated. Organics were concentrated to a solid, triturated in diethyl ether and solid was filtered and dried to give 5.52 g light brown solid (87% yield, both batches). 1H MR (DMSO-de) δ: 9.93 (s, 1H), 7.22 - 7.30 (m, 2H), 7.09 - 7.17 (m, 1H), 6.99 (d, J = 7.3 Hz, 1H), 4.63 (dd, J = 9.1, 6.8 Hz, 1H), 2.52 - 2.81 (m, 4H); MS (m/z) 288.0 (M+H+).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000069_0001
Preparation 9
3-amino-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one
Figure imgf000069_0002
Step 1 : To a solution of 3-iodo-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one (16.8 g, 58.5 mmol) in Ν,Ν-Dimethylformamide (DMF) (100 mL) was added sodium azide (4.57 g, 70.2 mmol) (mild exotherm) and mixture was stirred at rt for 1 hour. A precipitate formed after 30 minutes. Ice was added to the reaction and it was then diluted with 300 mL water. More solid precipitated out and the mixture was stirred for 10 minutes. Filtered tan solid, rinsed with water and used as-is in next step (was not dried because next step contained water). Small amount dried for HNMR analysis. 1H MR (DMSO-d6) δ: 10.05 (s, 1H), 7.20 - 7.33 (m, 2H), 7.06 - 7.17 (m, 1H), 7.00 (d, J = 7.8 Hz, 1H), 3.89 (dd, J = 11.6, 8.1 Hz, 1H), 2.65 - 2.81 (m, 2H), 2.41 (tt, J = 12.7, 7.8 Hz, 1H), 2.04 - 2.17 (m, 1H); MS (m/z) 203.0 (M+H+).
Step 2: To a solution of 3-azido-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one in THF (120 mL) was added 1.0 mL water and PPh3 resin (21.5 g, 3 mmol/g loading, 1.1 eq, 64.4 mmol, Aldrich). Stirred at rt for 20 hours. Reaction was filtered to remove resin, rinsed with THF and filtrate was concentrated. Triturated solid in 10% DCM/diethyl ether, filtered and dried to give a tan solid (9.13 g, 85% yield over 2 steps). 1H NMR (DMSO-d6) δ: 9.68 (br. s., 1H), 7.18 - 7.29 (m, 2H), 7.04 - 7.13 (m, 1H), 6.96 (d, J = 7.8 Hz, 1H), 3.13 (dd, J = 11.4, 7.8 Hz, 1H), 2.55 - 2.70 (m, 2H), 2.27 (tt, J = 12.9, 7.7 Hz, 1H), 1.70 - 1.83 (m, 1H), 1.62 (br. s., 2H); MS (m/z) 177.0 (M+H+). The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000070_0001
Preparation 10
-amino-4,5-dihydro-lH-benzo[b]azepin-2(3H)-
Figure imgf000070_0002
To a solution of 3-amino-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one (24.1 g, 127 mmol) in isopropanol (300 mL) at 70°C was added L-pyroglutamic acid (16.42 g, 127 mmol). Stirred for 30 minutes. Added 400 mL more isopropanol to facilitate stirring. Then 2-hydroxy-5-nitrobenzaldehyde (0.638 g, 3.82 mmol) was added and mixture was stirred at 70°C for 3.5 days. Mixture was cooled to rt, solid was filtered, rinsing with isopropanol and hexanes. Solid was dried to give a tan solid as the pyroglutamic acid salt (33 g, 84%). The %ee = 97.4% @ 220 nm and 97.8% @ 254 nm. MS (m/z) 177.0 (M+H+).
(S)-3-amino-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one, pyroglutamic acid salt (33 g) was basified with minimum amount of concentrated H4OH and extracted with DCM four times. The combined organics were concentrated to a solid which was triturated in diethyl ether, filtered and dried to give a light orange/tan solid as free base (19.01 g, 81%).
1H MR (DMSO-d6) δ: 9.70 (br. s., 1H), 7.17 - 7.30 (m, 2H), 7.05 - 7.13 (m, 1H), 6.96 (d, J = 7.8 Hz, 1H), 3.15 (dd, J = 11.5, 8.0 Hz, 1H), 2.56 - 2.73 (m, 2H), 2.28 (tt, J = 12.9, 7.6 Hz, 1H), 2.04 (br. s., 2H), 1.69 - 1.83 (m, 1H); MS (m/z) 177.0 (M+H+). The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000071_0001
Preparation 11
(S)-3-amino-l-methyl-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one, HC1 salt
Figure imgf000071_0002
Step 1 : To a mixture of (S)-3-amino-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one (0.615 g, 3.49 mmol) in DCM (20 mL) was added TEA (0.730 mL, 5.24 mmol) and BOC20 (0.851 mL, 3.66 mmol). The reaction was stirred at rt for 1 hour, diluted with water and layers were separated. The organics were concentrated and dried to give 950 mg of (S)-tert-butyl (2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate as an off- white solid. 1H MR (DMSO-d6) δ: 9.71 (s, 1H), 7.22 - 7.30 (m, 2H), 7.08 - 7.15 (m, 1H), 6.95-7.03 (m, 2H), 3.87 (dt, J = 12.1, 8.2 Hz, 1H), 2.61 - 2.70 (m, 2H), 2.19 (ddd, J = 12.0, 8.0, 4.0 Hz, 1H), 2.01 - 2.12 (m, 1H), 1.34 (s, 9H); MS (m/z) 277 (M+H+).
Step 2: To a mixture of cesium carbonate (1.592 g, 4.89 mmol) and (S)-tert-butyl (2-oxo-2,3, 4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate (950 mg, 3.40 mmol) in N,N- Dimethylformamide (DMF) (10 mL) was added iodomethane (0.262 mL, 4.19 mmol). The reaction was stirred at rt for 20 hours, then water (30 mL) was added and mixture was stirred vigorously for 15 minutes. The resulting solid was filtered, rinsed with water and hexanes and dried to give 800 mg of (S)-tert-butyl (l-methyl-2-oxo-2,3,4,5-tetrahydro-lH- benzo[b]azepin-3-yl)carbamate as an off-white solid. 1H MR (DMSO-d6) δ: 7.34 - 7.39 (m, 2H), 7.29 (d, J = 7.3 Hz, 1H), 7.18 - 7.24 (m, 1H), 7.03 (d, J = 8.6 Hz, 1H), 3.86 (dt, J = 11.6, 8.3 Hz, 1H), 3.27 (s, 3H), 2.60 - 2.66 (m, 2H), 2.01 - 2.13 (m, 2H), 1.33 (s, 9H); MS (m/z) 291 (M+H+). Step 3 : To a solution of (S)-tert-butyl (l-methyl-2-oxo-2,3,4,5-tetrahydro-lH- benzo[b]azepin-3-yl)carbamate (800 mg, 2.73 mmol) in DCM (20 mL) was added HC1 (4.0 M in dioxane) (4.0 mL, 16.00 mmol). The mixture was stirred at rt for 1.5 hours, then concentrated and dried to give 670 mg of (S)-3-amino-l-methyl-4,5-dihydro-lH- benzo[b]azepin-2(3H)-one, HC1 salt as a tan solid. 1H NMR (DMSO-d6) δ: 8.33 (br. s., 3H), 7.39 - 7.43 (m, 2H), 7.36 (d, J = 7.1 Hz, 1H), 7.24 - 7.30 (m, 1H), 3.62 (dd, J = 11.4, 8.1 Hz, 1H), 3.57 (s, 3H), 2.70 - 2.77 (m, 2H), 2.44 (ddd, J = 12.1, 8.0, 4.2 Hz, 1H), 2.07 - 2.17 (m, 1H); MS (m/z) 191 (M+H+).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000072_0001
Preparation 12
8-bromo-4 5-dihydro-lH-benzo[b]azepin-2(3H)-one
Figure imgf000072_0002
Step 1 : To a solution of sodium acetate (7.47 g, 91 mmol) in Water (13.33 mL) was added hydroxylamine hydrochloride (6.33 g, 91 mmol), then Ethanol (40 mL) and 7- bromo-3,4-dihydronaphthalen-l(2H)-one (10.25 g, 45.5 mmol). The white slurry was heated at 80°C for 45 minutes. Reaction was removed from heat, stirred for 10 minutes, then poured over ice and stirred until all ice melted. Filtered resulting solid, rinsed with water and dried to give a white solid (10.58 g, 95%). 1H NMR (DMSO-d6) δ: 11.29 (s, 1H), 7.94 (d, J = 2.0 Hz, 1H), 7.42 (dd, J = 8.2, 2.1 Hz, 1H), 7.17 (d, J = 8.3 Hz, 1H), 2.66 (dt, J = 16.9, 6.3 Hz, 4H), 1.74 (quin, J = 6.4 Hz, 2H); MS (m/z) 240/242 (M+H+), bromine splitting pattern. Step 2: To methanesulfonic acid (100 mL) was added phosphorus pentoxide (9.70 g, 68.3 mmol) and mixture was heated at 90°C for 1.5 hours. Removed from heat and added 7-bromo-3,4-dihydronaphthalen-l(2H)-one oxime (10.58 g, 43.2 mmol) in portions over 10 minutes. Mixture was heated at 80°C for 20 hours. Reaction was removed from heat and poured over ice, then 50% w/w NaOH was added slowly along with ice to control temperature. Resulting precipitate was stirred for 10 minutes, filtered, rinsed with water and dried to give a pink powder that was 80% pure (9.81 g, 74%). 1H NMR (DMSO-d6) δ: 9.61 (s, 1H), 7.19 - 7.32 (m, 2H), 7.13 (d, J = 2.0 Hz, 1H), 2.66 (t, J = 6.9 Hz, 2H), 2.04 - 2.21 (m, 4H); MS (m/z) 240/242 (M+H+), bromine splitting pattern.
Preparation 13
(R)-2-((tert-butoxycarbonyl)amino)-3-((2-nitrophenyl)thio)propanoic acid
,Boc
HN
N02
Figure imgf000073_0001
N(¾
To a solution of (R)-2-((tert-butoxycarbonyl)amino)-3-mercaptopropanoic acid (5.02 g, 22.69 mmol)in Water (32 mL) was added NaHC03 (5.72 g, 68.1 mmol) stirred at 25°C was slowly added a solution of l-fluoro-2-nitrobenzene (3.20 g, 22.69 mmol) in Ethanol (40 mL). The reaction mixture was stirred at reflux for 4h and cooled to rt. LCMS indicated the reaction was completed. The ethanol was removed under vacuum and the resulting aqueous phase was diluted with water (50 ml), washed with ether (2 x 100 ml), (discarded the ether phase LCMS showed minor product). The aqueous was acidified to pH 4 with IN aqueous HC1 and extracted with DCM (2x300 mL). The organic layers were combined, washed with brine, dried over Na2S04 and concentrated in vacuo to afford the title compound as yellow solid (R)-2-((tert-butoxycarbonyl)amino)-3-((2- nitrophenyl)thio)propanoic acid (7 g, 20.4 mmol, 90 % yield). MS (m/z) 343 (M+H+). Preparation 14
-aminophenyl)thio)-2-((tert-butoxycarbonyl)amino)propanoic acid
Figure imgf000074_0001
To a solution of (R)-2-((tert-butoxycarbonyl)amino)-3-((2- nitrophenyl)thio)propanoic acid (0.8 g, 2.337 mmol) in MeOH (100 mL) was added ammonium chloride (0.250 g, 4.67 mmol) and zinc (1.528 g, 23.37 mmol) at 25°C. After stirring at rt for lh, the mixture was heated to 75 C for 2 h. The resulting mixture was then directly filtered through celite and celite was washed with boiling MeOH (2x100 ml). The combined organic were partially concentrated under vacuum (25 ml) and the residue was allowed to stand overnight at rt. Solid salts were eliminated by filtration, then DCM (100 ml) and water (100 ml) was added to the filtrate, the resulting organic phase was washed with water (3x100 ml), dried over Na2S04 and concentrated in vacuo to afford the title compound as solid (R)-3-((2-aminophenyl)thio)-2-((tert-butoxycarbonyl)amino)propanoic acid (700 mg, 2.241 mmol, 96 % yield). MS (m/z) 313 (M+H+).
Preparation 15
(R)-tert-butyl (4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]thiazepin-3-yl)carbamate
Figure imgf000074_0002
To a solution of (R)-3-((2-aminophenyl)thio)-2-((tert- butoxycarbonyl)amino)propanoic acid (3.3g, 10.56 mmol) in DCM (100 mL) was added Nl-((ethylimino)methylene)-N3,N3-dimethylpropane-l,3-diamine hydrochloride (2.228 g, 11.62 mmol). Stirred at rt for 5min, then added 4-methylmorpholine (1.742 mL, 15.85 mmol). The reaction mixture was stirred at 25 °C for 5h. LCMS showed product and the reaction was completed. Removed all the DCM and added 200 ml of EtOAc and the mixture was washed with water, 0. IN HCl(aq), NaHC03(aq) and brine. The organic phase was dried over Na2S04 and concentrated under reduced pressure to afford crude product. ISCO purification (eluting with 0-70% of EtOAc in hexane) to afford the pure title compound as (R)-tert-butyl (4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]thiazepin-3- yl)carbamate (1.5 g, 5.10 mmol, 48.2 % yield). 1H MR (400MHz, CDC13) δ = 7.73 - 7.57 (m, 1 H), 7.39 (td, J= 1.4, 7.6 Hz, 2 H), 7.27 - 7.03 (m, 2 H), 5.58 (br. s., 1 H), 4.49 (dt, J = 7.2, 11.8 Hz, 1 H), 3.85 (dd, 7= 6.7, 11.0 Hz, 1 H), 2.95 (t, 7= 11.4Hz, 1 H), 1.42 (s, 9 H). MS (m/z) 295 (M+H+).
Preparation 16
(R)-3-amino-2,3-dihydrobenzo[b][l,4]thiazepin-4(5H)-one, Hydrochloride
Figure imgf000075_0001
To a solution of (R)-tert-butyl (4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]thiazepin-3- yl)carbamate (100 mg, 0.340 mmol) in dioxane (3 mL) was added HC1 (0.425 mL, 1.699 mmol, 4M in dioxane). The reaction mixture was stirred at 25 °C for 18h. LCMS indicated the product w/o starting material. Removed all the solvents and washed the solid with ether and the solid was used without further purification. MS (m/z) 195 (M+H*).
The following intermediates used for the preparation of titled example compounds was synthesized using methods analogous to the ones described above.
Figure imgf000075_0002
Preparation 17
(R)-tert-butyl (5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]thiazepin-3-yl)carbamate
Figure imgf000076_0001
To a solution of (R)-tert-butyl (4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]thiazepin-3- yl)carbamate (200 mg, 0.679 mmol) in Ν,Ν-Dimethylformamide (DMF) (5 mL) was added Cs2C03 (332 mg, 1.019 mmol) The reaction mixture was stirred at rt for 5 min, then Mel (0.051 mL, 0.815 mmol) was added. The reaction mixture was stirred at rt for 3h and LCMS showed the reaction was completed. Added EtOAc and washed with water, brine and dried over Na2SC>4. Removed all the solvent to afford the title compound as (R)-tert- butyl (5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]thiazepin-3-yl)carbamate (200 mg, 0.649 mmol, 95 % yield). MS (m/z) 309 (M+H+). Preparation 18
(R)-3-amino-5-methyl-2,3-dihydrobenzo[b][l,4]thiazepin-4(5H)-one hydrochloride
Figure imgf000076_0002
To a solution of (R)-tert-butyl (5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]thiazepin-3-yl)carbamate (290 mg, 0.940 mmol) in DCM (3 mL) was added HC1 (7.05 mL, 28.2 mmol, 4M in dioxane). The reaction mixture was stirred at 25 °C for 3h. LCMS indicated the product w/o starting material. Removed all the solvents and the solid (200mg, 87%) was washed with ether and hexane and used without further purification. MS (m/z) 209 (M+H+).
Preparation 19
(S)-3-((tert-butoxycarbonyl)amino)-5-methyl-4-oxo-2
tetrahydrobenzo[b][l,4]oxazepine-7-carboxylic ac
Figure imgf000076_0003
To a solution of ester (500.0 mg, 1.427 mmol) in THF (16 mL)/Water (5 mL) was added LiOH (2.141 mL, 2.141 mmol) as a solution in water (1.0 mL). After 3h, the reaction mixture was poured into cold water (70 mL) then extracted twice with EtOAc. The aqueous phase was acidified to pH~3 then extracted with EtOAc twice to extract the desired product. The organic phase was dried over Na2S04, filtered, then concentrated in vacuo. The solid was azeotroped twice with toluene then concentrated to a final solid that was sufficiently pure to use in the next step. No further purification appeared necessary. Yield: 456 mg (90%) white solid. MS (m/z) 337.3 (M+H+).
Preparation 20
(S)-tert-butyl (7-(hydrazinecarbonyl)-5-methyl-4-oxo-2,3,4,5-tetrahydro benzo[b][l,4] xazepin-3-yl) carbamate
Figure imgf000077_0001
To a suspension of (S)-3-((tert-butoxycarbonyl)amino)-5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine-7-carboxylic acid (228 mg, 0.678 mmol) in dry DCM (5.5 ml) was added CDI (115 mg, 0.712 mmol) as a solid. . The reaction mixture was stirred at rt for lh 30 min then this mixture was added slowly dropwise to a separate stirring solution of anhydrous hydrazine (217 mg, 6.78 mmol) in 3.0 mL of dry DCM at rt. After lh, the reaction mixture was diluted with DCM then washed with water and brine. After drying the sample over Na2S04 and concentration, the solid product had sufficient purity to carry to the next step, (164 mg, 69%). 1H NMR (DMSO-d6) δ: 9.84 (s, 1H), 7.88 (d, J = 2.0 Hz, 1H), 7.71 (dd, J = 8.3, 2.0 Hz, 1H), 7.26 (d, J = 8.3 Hz, 1H), 7.21 (d, J = 7.8 Hz, 1H), 4.55 (br. s., 1H), 4.30 - 4.41 (m, 4H), 3.31 (s, 3H), 1.34 (s, 9H). MS (m/z) 351.3 (M+H+).
Preparation 21
(S)-tert-butyl (5-methyl-4-oxo-7-(5-oxo-4,5-dihydro-l,3,4-oxadiazol-2-yl)-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate
Figure imgf000078_0001
To a solution of (S)-tert-butyl (7-(hydrazinecarbonyl)-5-methyl-4-oxo-2,3,4,5- tetrahydro benzo[b][l,4] xazepin-3-yl)carbamate (191.0 mg, 0.545 mmol) in N,N- Dimethylformamide (DMF) (3.0 mL) was added TEA (0.114 mL, 0.818 mmol) followed by CDI (97 mg, 0.600 mmol). The mixture was stirred at rt. The reaction mixture was diluted with EtOAc then washed with cold dilute HCl, water (2x), and brine. The organic phase was dried over Na2S04, filtered, then concentrated in vacuo. The residue was washed with small amount of DCM then filtered and collected as a white powder then used in the next step without further purification (150.0 mg, 80%). 1H MR (DMSO-d6) δ: 12.67 (br. s., 1H), 7.83 (d, J = 2.0 Hz, 1H), 7.68 (dd, J = 8.3, 2.0 Hz, 1H), 7.35 (d, J = 8.3 Hz, 1H), 7.23 (d, J = 7.8 Hz, 1H), 4.32 - 4.45 (m, 3H), 1.35 (s, 9H). MS (m/z) 377.3 (M+H+).
Preparation 22
(S)-3-amino-5-methyl-7-(5-oxo-4,5-dihydro-l,3,4-oxadiazol-2-yl)-2,3-dihydrobenzo[b]
[1,4] oxazepin-4(5H)-one, Hydrochloride
Figure imgf000078_0002
To a solution of (S)-tert-butyl (5-methyl-4-oxo-7-(5-oxo-4,5-dihydro-l,3,4- oxadiazol-2-yl)-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate (40.0 mg, 0.106 mmol) in DCM (1.0 mL) was added a solution of 4M HCl in 1,4 dioxane (0.531 mL, 2.126 mmol). The mixture was stirred at rt for lh. The reaction mixture was concentrated in vacuo, then it was azeotroped twice with toluene to yield the desired product. MS (m/z) 277.1 (M+H+).
Preparation 23 (S)-3-(5-benzylisoxazole-3-carboxamido)-5-methyl-4-oxo-2,3,4,5-tetrahydro
-7-carboxylic acid
Figure imgf000079_0001
To a solution of (S)-methyl 3-(5-benzylisoxazole-3-carboxamido)-5-methyl-4-oxo-
2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine-7-carboxylate (332 mg, 0.762 mmol) in THF (6 mL)/Water (2.0 mL) was added LiOH (1.144 mL, 1.144 mmol) as a solution in water. Reaction was stirred at rt for about 2h. The reaction mixture was diluted with water then extracted with EtOAc twice. The aqueous phase was acidified to pH~3.0 then it was extracted with EtOAc. The latter organic phase was dried over Na2S04 then filtered and concentrated in vacuo to yield the desired product as a solid. The solid was warmed in toluene then decanted to give the final solid product that was used directly in the next step. 1H MR (DMSO-de) δ: 13.18 (br. s., 1H), 8.87 (d, J = 8.1 Hz, 1H), 7.98 (d, J = 2.0 Hz, 1H), 7.85 (dd, J = 8.3, 2.0 Hz, 1H), 7.25 - 7.38 (m, 6H), 6.55 (s, 1H), 4.87 (dt, J = 11.8, 7.7 Hz, 1H), 4.64 (dd, J = 11.6, 10.1 Hz, 1H), 4.46 (dd, J = 9.9, 7.6 Hz, 1H), 4.22 (s, 2H). MS (m/z) 422.3 (M+H+).
Preparation 24
(S)-5-benzyl-N-(7-(hydrazinecarbonyl)-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo [b][l,4] oxazepin-3-yl)isoxazole-3-carboxamide
Figure imgf000079_0002
To a suspension of (S)-3-(5-benzylisoxazole-3-carboxamido)-5-methyl-4-oxo- 2,3,4,5-tetrahydro benzo[b][l,4]oxazepine-7-carboxylic acid (178.0 mg, 0.422 mmol) in
DCM (6.0 mL) was added CDI (75 mg, 0.465 mmol). The reaction mixture was stirred at rt for lh 30 min. The mixture was then added slowly dropwise to a solution of hydrazine
(0.199 mL, 6.34 mmol) in 0.50 mL DCM. After lh LCMS indicated about 79 % conversion to the desired product. The reaction mixture was diluted with DCM then washed with water and brine. After drying the sample over Na2S04 and concentration, the solid product had sufficient purity to cany to next step. 1H MR (DMSO-d6) δ: 9.86 (s, 1H), 8.90 (d, J = 8.1 Hz, 1H), 7.91 (d, J = 2.3 Hz, 1H), 7.73 (dd, J = 8.3, 2.0 Hz, 1H), 7.27 - 7.38 (m, 6H), 6.55 (s, 1H), 4.85 (dt, J = 11.8, 7.9 Hz, 1H), 4.62 (dd, J = 11.6, 10.1 Hz, 1H), 4.54 (br. s., 2H), 4.44 (dd, J = 9.9, 7.8 Hz, 1H), 4.22 (s, 2H), 3.33 (s, 3H). MS (m/z) 436.2 (M+H+). Preparation 25
(S)-5-benzyl-N-(7-(hydrazinecarbonyl)-5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b] [ 1 ,4]oxazepin-3 -yl)- lH-pyrazole-3 -carboxamide
Figure imgf000080_0001
To a solution of (S)-methyl 3-(5-benzyl-lH-pyrazole-3-carboxamido)-5-methyl-4- oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine-7-carboxylate (191.0 mg, 0.440 mmol) in MeOH (5.0 mL) was added hydrazine monohydrate (0.058 mL, 1.199 mmol) as a solution in MeOH (1.0 mL). The reaction mixture was refluxed overnight then it was diluted with EtOAc and partitioned with water. The organic phase was dried over Na2S04, filtered, then concentrated in vacuo. The residue was purified by FCC (MeOH-DCM: 0-7.0%]) to yield the desired the product (119.0 mg, 62.3%). 1H MR (DMSO-d6) δ: 13.22 (s, 1H), 9.85 (s, 1H), 8.09 (d, J = 8.1 Hz, 1H), 7.90 (d, J = 2.0 Hz, 1H), 7.73 (dd, J = 8.3, 2.0 Hz, 1H), 7.18 - 7.34 (m, 6H), 6.37 (d, J = 1.5 Hz, 1H), 4.84 (dt, J = 11.6, 7.8 Hz, 1H), 4.48 - 4.62 (m, 3H), 4.36 - 4.47 (m, 1H), 3.99 (s, 2H). MS (m/z) 435.2 (M+H+).
Preparation 26
(S)-tert-butyl (7-((2-cyanoethyl)carbamoyl)-5-methyl-4-oxo-2,3,4,5-tetra hydro
benzo[b][l,4]oxazepin-3-yl)carbamate
Figure imgf000080_0002
To a suspension of (S)-3-((tert-butoxycarbonyl)amino)-5-methyl-4-oxo-2,3,4,5- tetra-hydrobenzo[b][l,4]oxazepine-7-carboxylic acid (185.0 mg, 0.550 mmol) in DCM (5.0 mL) was added l-chloro-N,N,2-trimethylprop-l -en- 1 -amine (88 mg, 0.660 mmol) as a solution in DCM (0.10 ml) dropwise over 1 min. The reaction mixture was stirred at rt for lh and became a homogeneous solution. The reaction mixture was cooled in an ice-bath then 3-aminopropanenitrile (154 mg, 2.200 mmol) was added dropwise as a solution in DCM (0.25 mL). After 10 min, the ice-bath was removed then 10 % aq citric acid solution was added and the mixture was stirred vigorously for 15 min . The organic phase was separated, washed with sat. aq sodium bicarbonate, brine then dried over Na2S04 and concentrated in vacuo. The residue was purified by FCC [EtO Ac-Hex: 45-80%] to yield the desired product (190.0 mg, 89%).MS (m/z) 389.3 (M+H+).
Preparation 27
(S)-tert-butyl (7-(l-(2-cyanoethyl)-lH-tetrazol-5-yl)-5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate
Figure imgf000081_0001
To a solution of (S)-tert-butyl (7-((2-cyanoethyl)carbamoyl)-5-methyl-4-oxo- 2,3,4,5-tetra hydro benzo[b][l,4]oxazepin-3-yl)carbamate (188.0 mg, 0.484 mmol) and pyridine (0.243 mL, 3.00 mmol) in DCM (2.0 mL) was added phosphorus pentachloride (161 mg, 0.774 mmol). The reaction mixture was heated to reflux for 3.0 h. The reaction mixture was cooled to rt then TMSN3 (0.257 mL, 1.936 mmol) was added and the reaction mixture was stirred overnight. At 20 h, 5.0 eq of TMS-N3 followed by 3.0 eq pyridine was added. The reaction mixture was warmed in an oil bath at 45 deg C for ~ 4h. The reaction mixture was carefully quenched with a few drops of sat. aq. NaHC03 initially, then after 5 min excess NaHC03 was added and the mixture was stirred for 15 min. The organic phase was separated and washed with 10% aq citric acid and brine. The organic solution was dried over Na2S04, filtered and concentrated in vacuo. The residue was purified by FCC (EtO Ac-Hex: 50 -70 %) to yield the desired product (152.0 mg, 72%). 1H MR (DMSO- de) δ: 7.87 (d, J = 2.0 Hz, 1H), 7.68 (dd, J = 8.2, 2.1 Hz, 1H), 7.44 (d, J = 8.3 Hz, 1H), 7.21 (d, J = 8.6 Hz, 1H), 4.73 - 4.87 (m, 2H), 4.35 - 4.54 (m, 3H), 3.24 (t, J = 6.3 Hz, 2H), 1.36 (s, 9H). MS (m/z) 414.3 (M+H+). Preparation 28
-amino-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-7-yl)-lH- tetrazol-l-yl)propanenitrile, Trifluoroacetic acid salt
Figure imgf000082_0001
To a solution of (S)-tert-butyl (7-(l-(2-cyanoethyl)-lH-tetrazol-5-yl)-5-methyl-4- oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate (152 mg, 0.368 mmol) in DCM (1.0 mL) was added TFA (0.50 mL, 6.49 mmol). The mixture was stirred at rt for lh then the reaction mixture was concentrated in vacuo to a residue that was azeotroped with toluene to yield solid product that was used directly in the next step (149.0 mg, 95%). MS (m/z) 314.2 (M+H+).
Preparation 29
(S)-3-(5-benzylisoxazole-3-carboxamido)-N-(2-cyanoethyl)-5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine-7-carboxamide
Figure imgf000082_0002
To a suspension of (S)-3-(5-benzylisoxazole-3-carboxamido)-5-methyl-4-oxo- 2,3,4,5-tetrahydrobenzo [b][l,4]oxazepine-7-carboxylic acid (87.0 mg, 0.206 mmol) in DCM (2.0 mL) was added l-chloro-N,N,2-trimethylprop-l -en- 1 -amine (33.1 mg, 0.248 mmol) as a solution in DCM (0.10 ml) dropwise over 1 min. The reaction mixture was stirred at rt for lh and became a homogeneous solution. The reaction mixture was cooled in an ice-bath then 3-aminopropanenitrile (57.9 mg, 0.826 mmol) was added dropwise as a solution in DCM (0.25 mL). The ice-bath was removed then 10 % aq citric acid solution was added and the mixture was stirred vigorously for 15 min . The organic phase was separated, washed with sat. aq sodium bicarbonate and brine then dried over sodium sulfate and concentrated in vacuo. The residue was purified by FCC( EtO Ac-Hex: 60-80%) to yield the desired product(67.0 mg, 68.5 %). MS (m/z) 474.4 (M+H+). Preparation 30
(S)-tert-butyl (5-methyl-7-(morpholine-4-carbonyl)-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate
Figure imgf000083_0001
To a solution of (S)-3-((tert-butoxycarbonyl)amino)-5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b] [1,4] oxazepine-7-carboxylic acid (100.0 mg, 0.297 mmol DMSO (2.0 mL) was added DIEA (0.109 mL, 0.624 mmol) then HATU (113 mg, 0.297 mmol). After 5 min dimethylamine (0.156 mL, 0.312 mmol was added and the reaction mixture was stirred at rt. The reaction mixture was diluted with EtO Ac then washed with sat. aq. H4C1, water and brine. The organic phase was dried over Na2S04, filtered and concentrated in vacuo, The residue was purified by FCC [EtO Ac-Hex: 15-50%] to yield the desired product (44.0 mg, 40.7 %). 1H MR (DMSO-d6) δ: 7.53 (d, J = 1.8 Hz, 1H), 7.27 - 7.31 (m, 1H), 7.21 - 7.25 (m, 1H), 7.18 (d, J = 8.1 Hz, 1H), 4.27 - 4.44 (m, 3H), 3.29 (s, 3H), 2.99 (br. s., 6H), 1.35 (s, 9H). MS (m/z) 364.0 (M+H+).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000083_0002
Preparation 31
(S)-3-amino-N,N,5-trimethyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine-7- carboxamide trifluoroacetate
Figure imgf000083_0003
To a suspension of (S)-tert-butyl (5-methyl-7-(morpholine-4-carbonyl)-4-oxo-
2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate (46.0mg, 0.113 mmol) in DCM (1.5 mL) was added TFA (0.175 mL, 2.269 mmol). The reaction mixture was stirred for 4h at rt and found to be complete by LC/MS. The reaction mixture was concentrated in vacuo then azeotroped with toluene twice. The residue was used without further purification and used directly in the next step. MS (m/z) 264.0 (M+H+).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000084_0001
Preparation 32
(S)-di-tert-butyl (5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine-3,7- diyl)dicarbamate
Figure imgf000084_0002
A mixture of (S)-3-((tert-butoxycarbonyl)amino)-5-methyl-4-oxo-2,3,4,5-tetrahydro benzo[b][l,4]oxazepine-7-carboxylic acid (78.0 mg, 0.232 mmol),
diphenylphosphorylazide (DPP A) (0.070 mL, 0.325 mmol), TEA (0.091 mL, 0.649 mmol), and tBuOH (0.439 mL, 4.59 mmol) in toluene were first heated to 70 deg C for 30 min then to 100 deg C for overnight. After 20 min some desired product was observed; the reaction mixture was left overnight and appeared to be complete. The solvent was removed in vacuo then the residue was purified by FCC [E/H 25 %].( No work-up was needed and the sample was well purified by column.); 1H NMR (DMSO-d6) δ: 9.47 (br. s., 1H), 7.54 (d, J = 1.5 Hz, 1H), 7.25 (dd, J = 8.5, 1.9 Hz, 1H), 7.15 (d, J = 8.8 Hz, 1H), 7.07 (d, J = 8.6 Hz, 1H), 4.30 - 4.39 (m, 1H), 4.20 - 4.26 (m, 2H), 3.23 (s, 3H), 1.48 (s, 9H), 1.35 (s, 9H). MS (m/z) 408.3 (M+H+).
Preparation 33 (S)- -diamino-5-methyl-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one
Figure imgf000085_0001
To a suspension of (S)-di-tert-butyl (5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine-3,7-diyl)dicarbamate (75.0 mg, 0.184 mmol) in DCM (1.5 mL) was added HC1 (0.782 mL, 3.13 mmol). The reaction mixture was stirred overnight at rt. The solvent was evaporated, then the residue was azeotroped with toluene to obtain a solid residue that was used in the next step without further purification;
quantitative yield was assumed. MS (m/z) 208.1 (M+FT ). Preparation 34
(S)-N-(7-amino-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-5- b enzy li soxazol e-3 -carb oxami de
/Ύ° NH2 ° N- HATU °1 )
A solution of 5-benzylisoxazole-3-carboxylic acid (37.4 mg, 0.184 mmol) and HATU (77 mg, 0.202 mmol) in acetonitrile (2.5 mL) (1 mL) was stirred for lh. This mixture was added slowly to a second mixture of mixture of (9.0 eq) N-methylmorpholine (0.182 mL, 1.656 mmol) and (S)-3,7-diamino-5-methyl-2,3- dihydrobenzo[b][l,4]oxazepin-4(5H)-one, 2 hydrochloride (51.5 mg, 0.184 mmol) . LCMS showed predominantly one product 70 % plus bis-coupled 23%. The reaction mixture was diluted with EtO Ac then washed with water and brine. After drying over sodium sulfate, and filtering the sample was concentrated in vacuo and purified by FCC [EtO Ac-Hex: 20 - 60%]. 1H NMR (DMSO-de) δ: 8.78 (d, J = 8.3 Hz, 1H), 7.27 - 7.39 (m, 5H), 6.87 (d, J = 8.3 Hz, 1H), 6.58 (d, J = 2.8 Hz, 1H), 6.55 (s, 1H), 6.43 (dd, J = 8.6, 2.5 Hz, 1H), 5.16 (s, 2H), 4.77 - 4.87 (m, 1H), 4.36 - 4.44 (m, 1H), 4.20 - 4.27 (m, 3H), 3.22 (s, 3H). MS (m/z) 393.2 (M+H+).
Preparation 35 (S)-tert-butyl (3-((tert-butoxycarbonyl)amino)-5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-7-yl)(methyl)carbamate
Figure imgf000086_0001
To a suspension of (S)-di-tert-butyl (5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine-3,7-diyl)dicarbamate (60.0 mg, 0.147 mmol) DMF (1.0 mL) was added Cs2C03 (48.0 mg, 0.147 mmol) then Mel (9.21 μΐ., 0.147 mmol). The reaction mixture was combined with a second batch reaction of 40 mg scale and both were processed together. The reaction mixture was diluted with EtOAc then washed successively with water and brine. The organic phase was dried over Na2S04 and filtered. The concentrated residue was purified by FCC (EtO Ac-Hex: 15 -35 %) to yield an 86.0 mg mixture of desired product with some starting material (7:3 by LC/MS) that was used in the next step. MS (m/z) 422.4 (M+H+).
Preparation 36
(S)-3-amino-5-meth l-7- meth l- amino -2 3-dih drobenzo b l 4 oxaze in-4(5H)-one
Figure imgf000086_0002
To a mixture of (S)-tert-butyl (3-((tert-butoxycarbonyl)amino)-5-methyl-4-oxo- 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-7-yl)(methyl)carbamate from previous step (86.0 mg, 0.204 mmol) in DCM (3.0 ml) was added HCI (1.020 mL, 4.08 mmol) as a 4 M HCI solution in 1,4 dioxane. The reaction mixture was stirred at rt overnight then additional 0.5 mL of 4M HCI in 1,4 dioxane was added and stirring was continued lh. The reaction mixture was concentrated in vacuo, then azeotroped with toluene to yield residual solid having the desired product that was used directly in the next step. MS (m/z) 222.1 (M+H+). Preparation 37
(S)-3-((5-Amino-6-chloropyrimidin-4-yl)amino)-2-((tert-butoxycarbonyl)amino)propanoic acid
Figure imgf000087_0001
To a suspension of 4,6-dichloropyrimidin-5-amine (0.402 g, 2.448 mmol) and triethylamine (0.751 mL, 5.39 mmol) in BuOH (10 mL) was added (S)-3-amino-2-((tert- butoxycarbonyl)amino)propanoic acid (0.5 g, 2.448 mmol) at rt. The reaction mixture was heated at 90 °C. After heating for lhr, another 10 mL of BuOH and EtOH (15 mL) were added to the reaction mixture. After heating for 2 days (Note: still some starting material remained), the reaction mixture was concentrated, then diluted with water and EtOAc. After separation, the aqueous solution was extracted with EtOAc (x2), and then the aqueous solution was acidified with IN HC1 (pH around 3). After extraction with EtOAc (x3), the combined organic solution was washed with brine, dried over MgS04. After filtration and evaporation in vacuo, (S)-3-((5-amino-6-chloropyrimidin-4-yl)amino)-2-((tert- butoxycarbonyl)amino)propanoic acid (430 mg, 1.296 mmol, 52.9 % yield) was obtained as pale brownish solids, which was used for the next reaction without further purification. MS (m/z) 332.2 (M+H+). 1H NMR (DMSO-d6) δ: 12.67 (br. s., 1H), 7.75 (s, 1H), 7.09 (d, J = 8.3 Hz, 1H), 6.85 - 6.91 (m, 1H), 5.05 (s, 2H), 4.24 (td, J = 8.0, 5.3 Hz, 1H), 3.71 - 3.82 (m, 1H), 3.55 - 3.68 (m, 1H), 1.35 - 1.41 (m, 9H).
The following intermediates used for the preparation of titled example compounds were synthesized using a method analogous to the ones described above using 4,6-dichloro- 2-methylpyrimidin-5-amine and 2-chloro-5-fluoro-3-nitropyridine in DMSO as solvent at
70 °C.
Figure imgf000087_0002
Preparation 38
(S)-tert-butyl (4-chloro-6-oxo-6,7,8,9-tetrahydro-5H-pyrimido[4,5-b][l,4]diazepin-7- yl)carbamate
Figure imgf000088_0001
To a solution of (S)-3-((5-amino-6-chloropyrimidin-4-yl)amino)-2-((tert- butoxycarbonyl)amino)propanoic acid (300 mg, 0.904 mmol) and HATU (378 mg, 0.995 mmol) in DMSO (4.0 mL) was added DIEA (0.237 mL, 1.356 mmol) at rt. After 5 hr at rt, another 378 mg of HATU and 0.24 mL of DIEA were added. After stirring for overnight at rt, water was added, then extracted with EtOAc (x3). The combined organic solution was washed with brine, and dried over MgS04. After filtration and evaporation in vacuo, the crude material was purified by silica gel column chromatography (Biotage, 25 g cartridge, 10% to 60% EtOAc in hexane) to give (S)-tert-butyl (4-chloro-6-oxo-6,7,8,9-tetrahydro- 5H-pyrimido[4,5-b][l,4]diazepin-7-yl)carbamate (125 mg, 0.394 mmol, 43.6 % yield) as colorless solid. MS (m/z) 314.2 (M+H+). 1H NMR (DMSO-d6) δ: 9.49 (s, 1H), 8.14 - 8.22 (m, 1H), 8.07 (s, 1H), 6.95 (d, J = 7.1 Hz, 1H), 4.23 - 4.34 (m, 1H), 3.41 - 3.51 (m, 2H), 1.39 (s, 9H).
Preparation 39
butyl (6-oxo-6,7,8, 9-tetrahydro-5H-pyrimido[4,5-b][l,4]diazepin-7-yl)carbamate
Figure imgf000088_0002
To a suspension of (S)-tert-butyl (4-chloro-6-oxo-6,7,8,9-tetrahydro-5H- pyrimido[4,5-b][l,4]diazepin-7-yl)carbamate (300 mg, 0.956 mmol) in EtOH (5 mL), EtOAc (5.00 mL), and MeOH (7.5 mL) was added Pd/C (153 mg, 0.143 mmol) at rt. The reaction mixture was stirred under H2 balloon for 3 hr. The reaction mixture was filtered and washed with EtOAc and MeOH. The combined filtrate was evaporated in vacuo and the resultant solid (S)-tert-butyl (6-oxo-6,7,8,9-tetrahydro-5H-pyrimido[4,5- b][l,4]diazepin-7-yl)carbamate (260 mg, 0.912 mmol, 95 % yield) was used for the next reaction without further purification. MS (m/z) 280.2 (M+H+). 1H NMR (DMSO-d6) δ: 10.32 (s, 1H), 9.41 (br. s., 1H), 8.50 (s, 1H), 8.01 (s, 1H), 7.06 (d, J = 7.1 Hz, 1H), 4.36 - 4.50 (m, 1H), 3.53 - 3.63 (m, 1H), 3.40 - 3.52 (m, 1H), 1.40 (s, 9H).
The following intermediate used for the preparation of titled example compounds was synthesized from (S)-tert-butyl (4-chloro-2,5-dimethyl-6-oxo-6,7,8,9-tetrahydro-5H- pyrimido[4,5-b][l,4]diazepin-7-yl)carbamate using methods analogous to the ones described above.
Figure imgf000089_0001
Preparation 40
(S)-tert-butyl (4-chloro-5-methyl-6-oxo-6,7,8,9-tetrahydro-5H-pyrimido[4,5- -7-yl)carbamate
Figure imgf000089_0002
To a solution of (S)-tert-butyl (4-chloro-6-oxo-6,7,8,9-tetrahydro-5H-pyrimido[4,5- b][l,4]diazepin-7-yl)carbamate (0.7 g, 2.231 mmol) in DMF (10 ml) was added NaH (0.094 g, 2.343 mmol) at rt. After 30 min at rt, iodomethane (0.146 ml, 2.343 mmol) was added and stirred for 1 hr 20 min. The addition of water triggered a precipitation. The solid was filtered and washed with water and hexane. The wet solid was collected and dried at 50°C in a vacuum oven to give (S)-tert-butyl (4-chloro-5-methyl-6-oxo-6,7,8,9-tetrahydro- 5H-pyrimido[4,5-b][l,4]diazepin-7-yl)carbamate (620 mg, 1.797 mmol, 81 % yield) as a colorless solid, which was used for the next reaction without further purification. MS (m/z) 328.2 (M+H+). 1H MR (DMSO-d6) δ: 8.18 (s, 1H), 8.09 (br. d, 1H), 7.02 (d, J = 7.6 Hz, 1H), 4.38 - 4.48 (m, 1H), 3.37 - 3.54 (m, 2H), 3.12 (s, 3H), 1.38 (s, 9H).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000089_0003
Preparation 41 (S)-tert-butyl (7-bromo-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate
Figure imgf000090_0001
To a mixture of (S)-3-amino-7-bromo-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one (800 mg, 3.14 mmol) in DCM (20 mL) was added Et3 (0.656 mL, 4.70 mmol) and BOC20 (0.764 mL, 3.29 mmol). The mixture was stirred at rt for 1 h, and then was diluted with H20 (20 mL). The organic layer was separated and concentrated. The resulting residue was purified by Isco Combiflash (20%-80% EtOAc/Hexane; 40g RediSep column).
Collected fractions containing the product were combined and concentrated to give the desired product as a white solid (900 mg, 81 % yield). 1H MR (CDC13) δ ppm 9.21 (s, 1H), 7.32 (d, J = 2.0 Hz, 1H), 7.24 (dd, J = 8.3, 2.0 Hz, 1H), 6.83 (d, J = 8.3 Hz, 4H), 5.68 (d, J = 7.8 Hz, 1H), 4.17 - 4.31 (m, 1H), 2.76 - 2.95 (m, 1H), 2.52 - 2.68 (m, 2H), 1.94 - 2.01 (m, 1H), 1.39 (s, 9H); MS (m/z): 355 (M+H+).
Preparation 42
Jert-butyl (7-cyano-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate
Figure imgf000090_0002
Jert-butyl (7-bromo-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate (200 mg, 0.563 mmol), zinc cyanide (99 mg, 0.845 mmol), Pd2dba3 (258 mg, 0.282 mmol), and S-Phos (277 mg, 0.676 mmol) were mixed in a 5 ml microwave vial. The vial was flushed with N2 3 times, and then 2ml of DMF was added. The reaction mixture was microwaved using an Emrys Optimizer (150W, absorption normal, 120 °C, 20 min). The mixture was then filtered and the filtrate was concentrated. The residue was purified by Isco Combiflash (20%-50% EtOAc/Hexane; 40g RediSep column). Collected fractions containing the product were combined and concentrated to give the desired product as a brown oil. This oil was lyophilized to a pale yellow solid (146 mg, 86 % yield). 1H MR (CDC13) δ ppm 9.18 (s, 1H), 7.47 - 7.59 (m, 2H), 7.13 (d, J = 8.1 Hz, 1H), 5.50 (d, J = 7.8 Hz, 1H), 4.26 (dt, J = 11.4, 7.7 Hz, 1H), 2.84 - 3.00 (m, 1H), 2.62 - 2.79 (m, 2H), 1.98 - 2.12 (m, 1H), 1.41 (s, 9H); MS (m/z): 302 (M+H+).
Preparation 43 3-amino-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepine-7-carbonitrile
Figure imgf000091_0001
To a mixture of tert-butyl (7-cyano-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3- yl)carbamate (70 mg, 0.234 mmol) in DCM (5 mL) was added HC1 (4N in dioxane) (0.31 mL, 1.23 mmol). The reaction mixture was stirred at rt for 1 h. The mixture was then concentrated and dried. This crude material was taken to the next step without purification (47 mg, 100% yield). MS (m/z): 202 (M+H+). Preparation 44
(S)-tert-butyl (2-oxo-7-(lH-tetrazol-5-yl)-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3- e
Figure imgf000091_0002
(S)-tert-butyl (7-cyano-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate (100 mg, 0.332 mmol) was dissolved in DMF (2 mL), and then sodium azide (64.9 mg,
0.999 mmol) and ammonium chloride (53.8 mg, 1.006 mmol) were added. The mixture was maintained at 120 °C for 16 h. The mixture was filtered and the filtrate was then
concentrated and the residue was purified by Isco Combiflash (2%-10% MeOH/CH2Cl2, 10%) Et3 in MeOH; 12g RediSep column). Collected fractions containing the product were combined and concentrated to give the desired product as a colorless oil (114 mg, 100 % yield). MS (m/z): 345 (M+H+).
Preparation 45
(S)-3-amino-7-(lH-tetrazol-5-yl)-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one
Figure imgf000091_0003
(S)-tert-butyl (2-oxo-7-(lH-tetrazol-5-yl)-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3- yl)carbamate (114 mg, 0.332 mmol) was dissolved in DCM (2 mL), and then HC1 (4 N in dioxane, 0.83 mL) was added. The mixture was maintained at rt for 2 h. The rxn mixture was concentrated to an off-white solid. MS (m/z): 245 (M+H+).
Preparation 46
tert-butyl (7-bromo-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate
Figure imgf000092_0001
To a mixture of 3-amino-7-bromo-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one (1.7g, 6.66 mmol) in DCM (50 mL) was added Et3 (1.393 mL, 10.00 mmol) and BOC20 (1.625 mL, 7.00 mmol). The mixture was maintained at rt for 1 h. The reaction mixture was then diluted with water and the organic layer was separated, concentrated and dried under high vacuum for 16 h. This crude material was taken to the next step without further purification (2.36 g, 100 % yield). MS (m/z): 355 (M+H+).
Preparation 47
tert-butyl (7-bromo-l-methyl-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate
Figure imgf000092_0002
To a mixture of cesium carbonate (3.04 g, 9.33 mmol) and tert-butyl (7-bromo-2- oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate (2.36 g, 6.66 mmol) in 1ml DMF and THF (50 mL) mixed solution was added iodomethane (0.500 mL, 8.00 mmol). The reaction mixture was maintained at rt for 20 h. The mixture was then filtered and the filtrate was concentrated. The residue was purified by Isco Combiflash (10%-50%
EtOAc/Hexane; 330 g RediSep column). Collected fractions containing the product were combined and concentrated to give the desired product as a white solid (1.6 g, 65 % yield). 1H MR (CDC13) δ ppm 7.44 (dd, J = 8.5, 2.1 Hz, 1H), 7.37 (d, J = 2.3 Hz, 1H), 7.07 (d, J = 8.6 Hz, 1H), 5.47 (d, J = 7.6 Hz, 1H), 4.23 (dt, J = 11.5, 7.5 Hz, 1H), 3.39 (s, 3H), 2.73 - 2.91 (m, 1H), 2.49 - 2.65 (m, 2H), 1.87 - 2.03 (m, 1H), 1.42 (s, 9H); MS (m/z): 369
(M+H+). Preparation 48
1 , 1 -dimethyleth -methyl-2-oxo-2,3 ,4, 5-tetrahy dro- 1H- 1 -benzazepin-3 -yl)carbamate-d 1
Figure imgf000093_0001
Tert-butyl (7-bromo-l-methyl-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3- yl)carbamate (80 mg, 0.217 mmol) was dissolved in 50ml THF and then cooled to -78 °C. N-butyllithium (2.5M in Hexane) (0.217 mL, 0.542 mmol) was added dropwise at -78 °C. The reaction mixture was maintained at -78 °C for 30 min, and then was quenched by MeOD. The mixture was washed by sat.NaHC03(aq). The organic layer was separated and purified by Isco Combiflash (20%-80% EtOAc/Hexane; 12g RediSep column). Collected fractions containing the product were combined and concentrated to give the desired product as a white solid (64 mg, 100 % yield). 1H NMR (400 MHz, CDC13) δ ppm 1.41 (s, 9 H) 1.95 (dd, J=7.45, 3.92 Hz, 1 H) 2.51 - 2.68 (m, 2 H) 2.73 - 2.98 (m, 1 H) 3.41 (s, 3 H) 4.19 - 4.34 (m, 1 H) 5.37 - 5.64 (m, 1 H) 7.11 - 7.24 (m, 2 H) 7.23 - 7.38 (m, 1 H); MS (m/z): 292 (M+H+).
Preparation 49
-amino- -methyl- 1 ,3,4,5-tetrahydro-2H- 1 -benzazepin-2-
Figure imgf000093_0002
1,1 -dimethylethyl (l-methyl-2-oxo-2,3,4,5-tetrahydro-lH-l-benzazepin-3- yl)carbamate-dl ( 64 mg) was dissolved in 2mL DCM, and then 0.54 mL HC1 (4N in dioxane) was added dropwise. The reaction mixture was maintained at rt for 4 h. The mixture was then concentrated and the crude material was taken to the next step without further purification (35 mg, 84 % yield). MS (m/z): 192 (M+H+).
Preparation 50
tert-butyl (l-methyl-2-oxo-7-(2,2,2-trifluoroacetyl)-2,3,4,5-tetrahydro-lH-benzo[b]azepin-
3-yl)carbamate
Figure imgf000094_0001
Tert-butyl (7-bromo-l-methyl-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3- yl)carbamate (100 mg, 0.271 mmol) was dissolved in 50ml THF and then cooled to -78 °C. N-butyllithium (2.5M in Hexane, 0.271 mL, 0.677 mmol) was added dropwise at -78 °C. This mixture was maintained at -78 °C for 30 min, and then ethyl 2,2,2-trifluoroacetate (0.129 mL, 1.083 mmol) was added dropwise at -78 °C. The mixture turned into colorless solution after addition. This mixture was maintained at -78 °C for 1 h, and then was slowly warmed up to rt. The rxn mixture was quenched by MeOH, and then washed by
sat. H4Cl(aq). The organic layer was separated and concentrated. The residue was purified by Isco CombiFlash (20%-80% EtOAc/Hexane; 40g RediSep column). Collected fractions containing the product were combined and concentrated to give the desired product as a yellow oil (24 mg, 23 % yield). MS (m/z): 404 (M+H+).
1 , 1 -Dimethylethyl [ 1 -methyl-2-oxo-7-(2,2,2-trifluoro- 1 , 1 -dihydroxyethyl)-2,3 ,4,5- tetrahydro-lH-l-benzazepin-3-yl]carbamate (125 mg, 0.324 mmol) was dissolved in DCM (2 mL), and then HCI (4N in dioxane) (0.809 mL, 3.24 mmol) was added. The yellow solution was maintained at room temperature for 16 hours. This solution was then concentrated to give the 3 -amino- l-methyl-7-(2,2,2-trifluoro-l,l-dihydroxy ethyl)- 1, 3,4,5- tetrahydro-2H-l-benzazepin-2-one as a yellow oil (92 mg, 100% yield). MS (m/z): 304 (M+H+).
Preparation 51
(S)-tert-butyl (2-oxo-7-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3,4,5-tetrahydro- lH-benzo[b]azepin-3-yl)carbamate
Figure imgf000095_0001
(S)-tert-butyl (7-bromo-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3- yl)carbamate (200 mg, 0.563 mmol), bis(pinacolato)diboron (157 mg, 0.619 mmol), PdC12(dppf)-CH2Cl2 adduct (46.0 mg, 0.056 mmol) and potassium acetate (182 mg, 1.858 mmol) were mixed in 1,4-Dioxane (2mL). The reaction mixture was put in an Emrys Optimizer (150W, absorption normal, 120 °C, 10 min). The reaction mixture was then partitioned between H20 and DCM. The organic layer was washed by brine, dried over
MgS04 and concentrated to a brown residue. This residue was purified by Isco Combiflash (10%-80% EtOAc/Hexane; 40g RediSep column). Collected fractions containing the product were combined and concentrated to give the desired product as a white solid (82 mg, 36 % yield). MS (m/z): 402 (M+H+).
Preparation 52
-3-amino-7-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-4,5-dihydi benzo[b]azepin-2(3H)-one
Figure imgf000095_0002
(S)-tert-butyl (2-oxo-7-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3,4,5- tetrahydro-lH-benzo[b]azepin-3-yl)carbamate (82 mg, 0.205 mmol) was dissolved in 2mL DCM, and then HCI (4N in dioxane, 1.408 mL, 5.63 mmol) was added dropwise. The reaction solution was maintained at rt for 16 h. The solution was then concentrated to a yellow oil (62 mg, 100 % yield). MS (m/z): 302 (M+H+). Preparation 53
(S)-5-benzyl-N-(2-oxo-7-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3,4,5-tetrahydro- lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3-carboxamide
Figure imgf000096_0001
To a magnetically stirred solution of 5-benzyl-4H-l,2,4-triazole-3-carboxylic acid
(41.7 mg, 0.205 mmol) in 5mL DCM at rt was added 4-methylmorpholine (66.4 mg, 0.657 mmol) and HATU (94 mg, 0.246 mmol). A solution of (S)-3-amino-7-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one (62 mg, 0.205 mmol) and in 2mL DCM was add to this mixture. The reaction mixture was maintained at rt for 16 h. The crude mixture was then purified by Isco Combiflash (20%-50% EtOAc/Hexane; 40g RediSep column). Collected fractions containing the product were combined and concentrated to give the desired product as a yellow solid (65 mg, 65 % yield). MS (m/z): 488 (M+H+). Preparation 54
(S)-3-amino-8-hydroxy-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one
Figure imgf000096_0002
(S)-tert-butyl (8-methoxy-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3- yl)carbamate (300 mg, 0.973 mmol) was dissolved in DCM (15 mL) and cooled in an ice/brine bath. Then boron tribromide (2.92 mL, 2.92 mmol) was added and reaction was stirred cooled for 10 minutes. Removed ice bath and stirred at rt for 60 minutes. Added another 2.0 mL BBr3 and stirred for 45 minutes, then added another 2 mL BBr3 and stirred for another 20 minutes. Reaction was cooled in an ice bath, quenched with 5 mL satd. NaHC03 and stirred vigorously for 5 minutes. The pH of the aqueous was - 7-8. The layers were separated and aqueous was extracted with 10% MeOH/DCM: both organics contained impurities and aqueous contained majority product. Concentrated aqueous to give 2.4 g crude solid (contained inorganic salts). Used as is in next step. MS (m/z) 195.0 (M+H+). Preparation 55
(S)-3-amino-5-me hyl-7-(lH-pyrazol-3-yl)-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one
Figure imgf000097_0001
(S)-3-amino-7-bromo-5-methyl-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one (50.5 mg, 0.186 mmol), (lH-pyrazol-3-yl)boronic acid (31.3 mg, 0.279 mmol), sodium carbonate (59.2 mg, 0.559 mmol) and Pd(PPli3)4 (21.53 mg, 0.019 mmol) were combined in DME (2 mL) and Water (0.7 mL) and reaction was heated at 85°C in oil bath for 3 hours. After 2 hours, more Pd(PPh3)4 (15 mg) and boronic acid (15 mg) were added. After 3 hours in oil bath, reaction was put in microwave at 120°C for 15 min. Reaction was partitioned between 10% MeOH/DCM and water. Concentrated organics and purified by Biotage (4 g silica column; 0.5-5% MeOH/DCM (plus NH4OH), 15 min.) to give 30 mg light yellow oil in 60% yield. 1H MR (DMSO-d6) δ: 7.79 (m, 2H), 7.66 (m, 1H), 7.19 (m, 1H), 6.76 (m, 1H), 4.27 (m, 1H), 4.00 (m, 1H), 3.65 (m, 1H), 3.35 (s, 3H), 1.72 (br. s., 2H); MS (m/z) 259.1 (M+H+).
Preparation 56
(S)-3-amino-5-methyl-7-(lH-pyrazol-l-yl)-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one
Figure imgf000097_0002
(S)-3-amino-7-bromo-5-methyl-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one (100 mg, 0.369 mmol), lH-pyrazole (50.2 mg, 0.738 mmol) and potassium carbonate (153 mg, 1.107 mmol) were added to 1,4-Dioxane (2.0 mL) and mixture was degassed for 10 minutes under nitrogen. Then copper(I) iodide (35.1 mg, 0.184 mmol) and N1,N2- dimethylethane-l,2-diamine (0.020 mL, 0.184 mmol) were added and mixture was heated at 100°C for 3 days. Cooled to rt, diluted with water and 10% MeOH/DCM and separated layers. Concentrated organics and purified by Biotage (12 g silica column; 0.5-3% MeOH/DCM (plus NH4OH), 15 min.; 3-4.5%, 3 min; 4.5% 5 min.) to give 18 mg light brown solid in 19% yield. 1H NMR (DMSO-d6) δ: 8.54 (d, J = 2.5 Hz, 1H), 7.85 (d, J = 2.5 Hz, 1H), 7.76 (d, J = 1.5 Hz, 1H), 7.69 (dd, J = 8.7, 2.7 Hz, 1H), 7.27 (d, J = 8.8 Hz, 1H), 6.54 - 6.59 (m, 1H), 4.29 (dd, J = 9.9, 7.6 Hz, 1H), 4.02 (t, J = 10.7 Hz, 1H), 3.68 (br. s., 1H), 3.36 (s, 3H), 1.75 (br. s., 2H); MS (m/z) 259.1 (M+H+).
Preparation 57
■butyl (2-oxo-8-(2-(pyrrolidin-l-yl)ethoxy)-2,3,4,5-tetrahydro-lH-benzo[b]azepin- -yl)carbamate
Figure imgf000098_0001
Polymer bound PPh3 (3 mmol/g loading, 2.5 eq, 330 mg), (S)-tert-butyl (8-hydroxy- 2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate (130 mg, 0.400 mmol) and 2- (pyrrolidin-l-yl)ethanol (0.094 mL, 0.800 mmol) were combined in THF (4 mL). Next added di-tert-butyl azodicarboxylate (184 mg, 0.800 mmol) and stirred mixture at rt for 3 days. Added 0.5 eq more of the following reagents: polymer bound PPh3, di-tert-butyl azodicarboxylate and 2-(pyrrolidin-l-yl)ethanol and stirred for another 24 hours. Reaction was filtered through a small plug of Celite, rinsing with 10% MeOH/DCM. Filtrate was concentrated, partitioned between DCM and 6N NaOH and layers were separated. Crude was concentrated and purified by Biotage (4 g silica column; 1-5% MeOH/DCM (plus
NH4OH), 15 min.) to give 74 mg white foam in 46% yield. 1H NMR (DMSO-d6) δ: 9.64 (s, 1H), 7.15 (d, J = 8.3 Hz, 1H), 6.97 (d, J = 8.3 Hz, 1H), 6.70 (dd, J = 8.3, 2.5 Hz, 1H), 6.57 (d, J = 2.5 Hz, 1H), 3.96 - 4.10 (m, 2H), 3.88 (dt, J = 11.9, 8.3 Hz, 1H), 2.77 (t, J = 5.9 Hz, 2H), 2.54 - 2.62 (m, 2H), 2.53 (m, 1H), 2.09 - 2.22 (m, 2H), 1.95 - 2.07 (m, 2H), 1.62 - 1.71 (m, 5H), 1.34 (s, 9H); MS (m/z) 390.3 (M+H+). Preparation 58
-butoxypicolinate
Figure imgf000099_0001
To a mixture of 4-chloropicolinic acid (1 g, 6.35 mmol) and butan-l-ol (5.80 ml, 63.5 mmol) was added sulfuric acid (0.101 ml, 1.904 mmol) and heated to 80 °C for 2 days. After cooling down to rt, the reaction mixture was diluted with water and neutralized with IN NaOH solution to pH 5-6, then extracted with EtOAc (x3). After drying over MgS04, filtration, and evaporation in vacuo, the residue was purified by Biotage (50 g cartridge, 0% to 40% EtOAc in hexane) to give butyl 4-butoxypicolinate (765 mg, 3.04 mmol, 48.0 % yield). MS (m/z) 252.1 (M+H+). 1H NMR (CDC13) δ: 8.55 (d, J = 6.1 Hz, 1H), 7.65 (d, J = 2.3 Hz, 1H), 6.95 (dd, J = 5.7, 2.7 Hz, 1H), 4.42 (t, J = 6.8 Hz, 2H), 4.09 (t, J = 6.4 Hz, 2H), 1.77 - 1.87 (m, 4H), 1.43-1.57 (m, 4H), 0.97-1.03 (m, 6H).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000099_0002
Preparation 59
Ethyl 2-amino-2-(2-(2-(3 -fluorophenyl)acetyl)hy drazono)acetate
Figure imgf000099_0003
2-(3-Fluorophenyl)acetohydrazide (2.90 g, 17.22 mmol) and ethyl 2-ethoxy-2- iminoacetate (2.5 g, 17.22 mmol) in ethanol (30 mL) was stirred under nitrogen at rt for overnight, the resultant suspension was filtered. The white solid was washed with EtOH and dried under vacuum to give the title compound ethyl 2-amino-2-(2-(2-(3- fluorophenyl)acetyl)hydrazono)acetate (3 g, 11.23 mmol, 65.2 % yield) which was used without further purification. MS (m/z) 268 (M+H+). The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000100_0001
Preparation 60
Ethyl 5-(3-fluorobenzyl)-4H-l,2,4-triazole-3-carboxylate
Figure imgf000100_0002
Ethyl 2-amino-2-(2-(2-(3-fluorophenyl)acetyl)hydrazono)acetate (3 g, 11.23 mmol) in a flask was placed in a pre-heated oil bath at 200°C for 15 minutes. The melt was allowed to cool, the resultant solid taken up into MeOH (20 mL), and then the solvent was evaporated. The resultant white solid was suspended in ether (30 mL), stirred for 10 minutes, filtered off, washed with ether (40 mL), and dried under vacuum to give the title compound ethyl 5-(3-fluorobenzyl)-4H-l,2,4-triazole-3-carboxylate (1.2 g, 4.81 mmol, 42.9 % yield), which was used without further purification. MS (m/z) 250 (M+H+). The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000101_0001
Preparation 61
5-Benzyl-4H-l,2,4-triazole-3-carboxylic acid
Figure imgf000101_0002
To a solution of ethyl 5-benzyl-4H-l,2,4-triazole-3-carboxylate (8.29 g, 35.85 mmol) in THF (100 mL) was added a solution of lithium hydroxide (2.00 g, 84 mmol) in water (20 mL). The mixture was stirred for 20 hours at room temperature. The reaction was concentrated to remove THF and cone. HC1 was added until pH ~ 2 at which point a solid precipitated out. The suspension was stirred for 15 minutes in an ice/water bath, filtered, rinsed with cold water and dried under vacuum to give 6.93 g (80% yield) of 5- benzyl-4H-l,2,4-triazole-3-carboxylic acid hydrochloride. MS (m/z) 204 (M+H+). The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000102_0001
Preparation 62
ethyl l-(3-fluorobenzyl)-lH-imidazole-4-carboxylate
Figure imgf000102_0002
To a solution of ethyl lH-imidazole-4-carboxylate (1 g, 7.14 mmol), CS2CO3 (2.56 g, 7.85 mmol) and in Ν,Ν-Dimethylformamide (DMF) (5 mL) was l-(bromomethyl)-3- fluorobenzene (1.349 g, 7.14 mmol). The reaction mixture was stirred at for 5h. LCMS showed the reaction was completed with product. Added 150 ml of EtOAc and extracted with water, brine and dried over Na2S04. Evaporated all the solvents to afford the crude product as ethyl l-(3-fluorobenzyl)-lH-imidazole-4-carboxylate (1.7 g, 6.85 mmol, 96 % yield). MS (m/z) 250 (M+H+).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000102_0003
Preparation 63
l-(3-fluorobenzyl)-lH-imidazole-4-carboxylic acid
Figure imgf000103_0001
Freshly prepared lithium hydroxide (34.2 mL, 68.5 mmol) was added to a stirring, room temperature solution of ethyl l-(3-fluorobenzyl)-lH-imidazole-4-carboxylate (1.7 g, 6.85 mmol) in THF (25 mL) under N2. The reaction was then stirred at rt overnight and LCMS showed completed. The reaction was concentrated and then dissolved in H20 (10 mL). 2N HC1 was added dropwise until the pH=3. The white solid that precipitated from the reaction was filtered off and washed with cold H20. The solid was dried under vacuum overnight to obtain title product. l-(3-fluorobenzyl)-lH-imidazole-4-carboxylic acid (1.2 g, 79.5%). MS (m/z) 221 (M+H+).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000103_0002
Preparation 64
-fluorophenyl)-2,4-dioxopentanoate
Figure imgf000103_0003
To a solution of l-(4-fluorophenyl)propan-2-one (25 g, 164 mmol) diethyl oxalate (28.8 g, 197 mmol) in toluene (300 mL) stirred under nitrogen at 0°C was added potassium tert-butoxide (23.97 g, 214 mmol) in toluene (300 mL). The reaction mixture was stirred at 0C for 2 more hours and then at rt for over night. LCMS indicated the reaction was completed. Removed all the toluene and dissolved the residue in water and neutralized to pH =6 and extracted with EtOAc twice. The organic phase was combined and washed with brine, and dried over Na2S04. Removed all the solvents to afford the title compound used without further purification (32g, 77%). MS (m/z) 253 (M+H+). The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000104_0001
Preparation 65
Ethyl 5-(4-fluorobenzyl)-lH-pyrazole-3-carboxylate
Figure imgf000104_0002
Hydrazine (1.095 mL, 34.9 mmol) was added to a stirring room temperature solution of ethyl 5-(4-fluorophenyl)-2,4-dioxopentanoate (8 g, 31.7 mmol) in ethanol (100 mL) under N2. The reaction was then heated to reflux (95 C oil bath) until judged complete by HPLC (3h). The reaction was concentrated and purified by silica gel chromatography (solid loading, Isco, 0-45% of EtOAc in hexane). Only pure fractions were combined and concentrated to obtain product as ethyl 5-(4-fluorobenzyl)-lH-pyrazole-3-carboxylate (4 g, 16.11 mmol, 50.8 % yield). MS (m/z) 249 (M+H+).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000104_0003
Preparation 66
5-(4-fluorobenzyl)-lH-pyrazole-3-carboxylic acid
Figure imgf000105_0001
Freshly prepared 2M LiOH aqueous solution (64.5 mL, 129 mmol) was added to a stirring, room temperature solution of ethyl 5-(4-fluorobenzyl)-lH-pyrazole-3-carboxylate (4 g, 16.11 mmol) in THF (65 mL) under N2. The reaction was then stirred at rt for 12 hours and LCMS showed 70% completed. Heated to 50 C for 2h and reaction was completed. The reaction was concentrated and then dissolved in 20 mL H20. To a stirring aqueous solution, 2N HCl was added dropwise until the pH=4. The white solid that precipitated from the reaction was filtered off and washed with cold H20. The solid was dried under vacuum overnight (at 40 °C) to obtain title product as 5-(4-fluorobenzyl)-lH- pyrazole-3-carboxylic acid (3g, 85%). MS (m/z) 221 (M+H+). 1H MR (DMSO-d6) δ: 12.59 - 13.70 (m, 1H), 7.01 - 7.41 (m, 4H), 6.46 (s, 1H), 3.95 (s, 2H).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000105_0002
Preparation 67
-methyl-l-(4-methylbenzyl)-lH-pyrazole-3-carboxylate
Figure imgf000105_0003
To a solution of ethyl 3-methyl-lH-pyrazole-5-carboxylate (607 mg, 3.94 mmol) in THF (20 mL) was added KOH (221 mg, 3.94 mmol). After stirring for 1 hr at rt, the reaction mixture turned to the suspension, then l-(bromomethyl)-4-methylbenzene (729 mg, 3.94 mmol) was added and heated to reflux. After overnight, the reaction mixture was cooled down to rt and concentrated. The residue was subjected to Biotage (cartridge 50g / pre-wet 5% EtO Ac/Hex / eluent: 5% to 25% EtOAc, then maintained 25% EtO Ac/Hex) to give ethyl 5-methyl-l-(4-methylbenzyl)-lH-pyrazole-3-carboxylate (833 mg, 3.16 mmol, 80 % yield) as a desired product and the regioisomer ethyl 3 -methyl- l-(4-methylbenzyl)- lH-pyrazole-5-carboxylate (58 mg, 0.220 mmol, 5.59 % yield). Ethyl 5-methyl-l-(4- methylbenzyl)-lH-pyrazole-3-carboxylate: 1H NMR (CDC13) 5: 7.13 (d, J = 7.1 Hz, 2H), 7.03 (d, 2H), 6.62 (br. s., 1H), 5.36 (br. s., 2H), 4.42 (dd, J = 7.1, 1.3 Hz, 2H), 2.34 (br. s., 3H), 2.19 (s, 3H), 1.35 - 1.50 (m, 3H); MS (m/z) 259.1 (M+H+). The regioisomer ethyl 3- methyl-l-(4-methylbenzyl)-lH-pyrazole-5-carboxylate: 1H NMR (CDC13) d: 7.05 - 7.24 (m, 4H), 6.66 (br. s., 1H), 5.67 (br. s., 2H), 4.19 - 4.42 (m, 2H), 2.32 (d, J = 3.0 Hz, 6H), 1.27 - 1.40 (m, 3H).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000106_0001
Figure imgf000107_0001
Preparation 68
5-Meth -l-(4-methylbenzyl)-lH-pyrazole-3-carboxylic acid
Figure imgf000107_0002
To a solution of ethyl 5-methyl-l-(4-methylbenzyl)-lH-pyrazole-3-carboxylate (830 mg, 3.21 mmol) in THF (3.0 mL) and water (3.0 mL) was added lithium hydroxide, H20 (539 mg, 12.85 mmol) at rt. After stirring for overnight at rt, the reaction mixture was concentrated in vacuo. The aqueous solution was diluted with water (5 mL) and acidified with IN HC1 (about 5.1 mL) to pH 3-4. The resultant white solid was collected and dried under a vacuum oven to give 5-methyl-l-(4-methylbenzyl)-lH-pyrazole-3-carboxylic acid (670 mg, 2.88 mmol, 90 % yield) as white solids. MS (m/z) 231.1 (M+H+). 1H NMR (DMSO-de) d: 12.58 (br. s., 1H), 7.16 (d, J = 7.8 Hz, 2H), 7.03 (d, J = 7.8 Hz, 2H), 6.51 (s, 1H), 5.32 (s, 2H), 2.27 (s, 3H), 2.22 (s, 3H).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000108_0001
Preparation 69
-b enzy 1 -2H-tetrazol e- 5 -carb oxy 1 ate
Figure imgf000108_0002
To a solution of ethyl 2H-tetrazole-5-carboxylate, sodium salt (800 mg, 4.85 mmol) in DMF (8 mL) was added (bromomethyl)benzene (1.151 mL, 9.69 mmol) at rt. After stirring for 48 hr at rt, Et3N (1.013 mL, 7.27 mmol) was added to the reaction mixture, then stirred for overnight. After adding water, the reaction mixture was extracted with EtOAc. The combined organic solution was washed with water and brine, dried over MgSC After filtration and concentration, the residue was subjected to Biotage (50g of silica gel cartridge; eluent: 5% to 15% EtOAc, then maintained 15% EtO Ac/Hex) to give ethyl 2- benzyl-2H-tetrazole-5-carboxylate (342 mg, 1.473 mmol, 30.4 % yield, unoptimized) as a major product: MS (m/z) 233.1 (M+H+); 1H NMR (DMSO-d6) d: 7.36 - 7.46 (m, 5H), 6.05 (s, 2H), 4.40 (q, J = 7.1 Hz, 2H), 1.33 (t, 3H). The regioisomer ethyl 1 -benzyl- lH-tetrazole- 5-carboxylate (87 mg, 0.375 mmol, 7.73 % yield) was obtained as a minor product: 1H NMR (DMSO-de) d: 7.29 - 7.43 (m, 5H), 5.92 (s, 2H), 4.44 (q, J = 7.1 Hz, 2H), 1.33 (t, 3H) (Note: some mixture of both products was also obtained).
The following intermediate used for the preparation of titled example compounds was synthesized using methods analogous to the ones described above.
Figure imgf000109_0001
Preparation 70
-Benzyl-2H-tetrazole-5-carboxylic acid
Figure imgf000109_0002
To a solution of ethyl 2-benzyl-2H-tetrazole-5-carboxylate (338 mg, 1.455 mmol) in THF (3 mL) and water (3.00 mL) was added LiOH (183 mg, 4.37 mmol). After stirring for lhr at rt, the reaction mixture was concentrated in vacuo and the residual aqueous solution was acidified with IN HC1 (around pH -2-3). A small amount of white solids was precipitated out. After collecting solids, the aqueous solution was placed in a hood and allowed to slow evaporation of water. Another white solid was obtained (followed this step two more times. Note: some product was still detected in water). The combined solid was dried in a vacuum oven at 50 °C to give 2-benzyl-2H-tetrazole-5-carboxylic acid (167.4 mg, 0.820 mmol, 56.3 % yield) as white solids. MS (m/z) 205.0 (M+H+). 1H MR
(DMSO-de) δ: 14.30 (br. s., 1H), 7.33 - 7.49 (m, 5H), 6.03 (s, 2H).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000110_0001
Preparation 71
Ethyl -(difluoro(phenyl)methyl)isoxazole-3-carboxylate
Figure imgf000110_0002
Ethyl 5-benzoylisoxazole-3-carboxylate (630 mg, 2.57 mmol) was dissolved in 2 mL of dichloroethane (DCE), and then a solution of DAST (0.944 mL, 7.71 mmol) in 2 mL of DCE was added dropwise at 0 °C. The reaction mixture was maintained at 50 °C for 16 h, and then the mixture was concentrated. The resulting brown residue was purified by Isco Combiflash (10 %-30 % EtOAc/Hexane; 80 g Isco RediSep column). Collected fractions containing the product were combined and concentrated to give the desired product as a yellow oil (207 mg, 31 % yield). 1H MR (CDC13) δ ppm 7.56 - 7.64 (m, 2H), 7.45 - 7.56 (m, 3H), 6.87 (s, 1H), 4.45 (q, J = 7.2 Hz, 4H), 1.41 (t, J = 7.1 Hz, 3H); MS (m/z): 268 (M+H+). Preparation 72
5-(difluoro(phenyl)methyl)isoxazole-3-carboxylic acid
Figure imgf000110_0003
Ethyl 5-(difluoro(phenyl)methyl)isoxazole-3-carboxylate (207 mg, 0.775 mmol) was dissolved in 2 mL of THF, and then lithium hydroxide monohydrate (48.8 mg, 1.162 mmol) was added. The reaction mixture was maintained at rt for 16 h. The rxn mixture was neutralized by adding a solution of 4N HCl/dioxane dropwise. The mixture was then filtered and the filtrate was concentrated to a yellow oil (185 mg, 100 % yield). MS (m/z): 240 (M+H+). Preparation 73
5-(hydroxy(phenyl)methyl)isoxazole-3-carboxylic acid
Figure imgf000111_0001
Ethyl 5-benzoylisoxazole-3-carboxylate (400 mg, 1.631 mmol) was dissolved in 5mL MeOH, and then NaBH4 (93 mg, 2.447 mmol) was added at 0 °C. The reaction mixture was maintained at rt for 16 h. The mixture was concentrated, and then partitioned between sat. NaHC03(aq) and DCM. The organic layer was concentrated and dissolved in lmL THF. An aqueous solution of LiOH (1.2 mL, 50 mg/ml solution) was added to this THF solution. The mixture was maintained at rt for 16 h. A solution of HCl (0.8 mL, 4N in dioxane) was added to the mixture. The organic layer was separated and concentrated to a yellow oil (190 mg, 53 % yield). 1H MR (DMSO-d6) δ ppm 7.26 - 7.54 (m, 6H), 6.50 (s, 2H), 5.91 (s, 1H); MS (m/z): 220 (M+H+).
Preparation 74
Ethyl 5 -b enzy li soxazol e-3 -carb oxy 1 ate
Figure imgf000111_0002
A solution of the ethyl 2-chloro-2-(hydroxyimino)acetate (39.1 g, 258 mmol) was dispensed into a solution of prop-2-yn-l-ylbenzene (10 g, 86 mmol) and triethylamine (29.4 mL, 430 mmol) in CH3CN (300 mL). After standing for overnight at 80 °C the solvent was removed in vacuum. The crude was dissolved in EtO Ac (200 mL) and was washed with saturated sodium bicarbonate solution (50 mL), water (50 mL) and saturated brine (50 mL). The organic phase was separated and dried over sodium sulphate and evaporated in vacuo to give ethyl 5-benzylisoxazole-3-carboxylate (6 g, 25.9 mmol, 30 % yield) as a yellow solid. Used directly in the next step without further purification. MS (m/z): 232 (M+H+). Preparation 75
5-Benzylisoxazole-3-carboxylic acid
Figure imgf000112_0001
A solution of the ethyl 5-benzylisoxazole-3-carboxylate (6 g, 25.9 mmol) was dispensed into a solution of the sodium hydroxide (2.1 mL, 78 mmol) in methanol (100 mL) and water (10 mL). After standing for 2 h at 20 °C, the solvent was removed in vacuum. The residue was acidified with dilute HCl (20 mL) and then extracted with EtOAc (50 mL). The organic phase was washed with water (20 mL) and saturated brine (20 mL), and dried over sodium sulphate. Evaporation in vacuo gave 5-benzylisoxazole-3-carboxylic acid (3.2 g, 15.31 mmol, 59.0 % yield) as a yellow solid. 1H MR (DMSO-d6) δ ppm 14.0 (bs, 1H), 7.2 - 7.4 (m, 5H), 6.6 (s, 1H), 4.2 (s, 2H); MS (m/z): 204 (M+H+).
Preparation 76
(S)-3-amino-5-methyl-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one hydrochloride
Figure imgf000112_0002
To a solution of (S)-tert-butyl (5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate (28 g, 96 mmol) ) in DCM (300 mL) was added 4M HCl (71.8 mL, 287 mmol) and reaction stirred under nitrogen at room temp for 3 hr. The solvents were evaporated to dryness to yield the crude compound which was triturated with diethyl ether (200 mL), filtered and dried in vacuo to afford (S)-3-amino-5- methyl-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one hydrochloride (22.2 g, 97 mmol, 101 % yield) as brown solid. MS (m/z): 193.20 (M+H+).
The following intermediates used for the preparation of titled example compounds were synthesized using methods analogous to the ones described above.
Figure imgf000112_0003
Figure imgf000113_0001
Figure imgf000113_0002
Preparation 77
-(4-Chlorobenzyl)isoxazole-3-carboxylic acid
Figure imgf000113_0003
To a solution of l-(4-chlorophenyl)propan-2-one (10 g, 59.3 mmol) in THF (150 mL) in an ice bath was added NaH (1.423 g, 59.3 mmol) portion wise over 30min.
Dimethyl oxalate (7.0 g, 59.3 mmol) was added at room temperature for lhour and the mixture was stirred at 25 °C for 2 hours. The solvent was removed in vacuo and the residue was dissolved in EtOAc which was washed with water. The aqueous phase was separated and extracted with EtOAc. The combined organic phases were dried over Na2S04, filtered, and concentrated in vacuo to give methyl 5-(4-chlorophenyl)-2,4-dioxopentanoate (15g, 50.1 mmol, 84 % yield) as an oil which was used in next step without further purification. MS (m/z): 255/257 (M+H+).
To a solution of methyl 5-(4-chlorophenyl)-2,4-dioxopentanoate (5 g, 19.63 mmol) in ethanol (80 mL) was added hydroxylamine hydrochloride (1.364 g, 19.63 mmol) and then the mixture was stirred at 78 °C for 2 hours monitored. The solvent was removed in vacuo and the residue was dissolved in EtOAc which was washed with water. The aqueous phase was separated and extracted with EtOAc. The combined organic phases were dried over Na2S04, filtered, and concentrated in vacuo to give methyl 5-(4- chlorobenzyl)isoxazole-3-carboxylate (4.7 g, 16.81 mmol, 86 % yield) as a solid, which was used in next step without further purification. MS (m/z): 252/254 (M+H+).
To a solution of methyl 5-(4-chlorobenzyl)isoxazole-3-carboxylate (100 mg, 0.397 mmol) in THF (5 mL) in an ice bath was added a solution of NaOH (15.89 mg, 0.397 mmol) in water (5 mL). The mixture was stirred at room temperature for 2 hours. The solvent was removed in vacuo and the residue was dissolved in water. The aqueous solution was acidified by addition of IN HC1 to pH=2-3. The resulting solid which deposited was collected by filtration and dried in vacuo to give pure 5-(4- chlorobenzyl)isoxazole-3-carboxylic acid (90 mg, 0.360 mmol, 91 % yield) as a white solid. MS (m/z): 238/240 (M+H+).
Preparation 78
5-((Methyl(phenyl)amino)methyl)-lH-pyrazole-3-carboxylic acid
Figure imgf000114_0001
To a mixture of ethyl l-acetyl-5-methyl-lH-pyrazole-3-carboxylate (2 g, 10.19 mmol) and NBS (1.996 g, 11.21 mmol) in CC14 (20 mL) was added benzoic
peroxyanhydride (0.123 g, 0.51 mmol) at room temperature followed by reflux for 5 hours. LCMS showed product with some starting material left. Removed all the solvent and the residue was purified by flash chromatography by solid loading (eluting 0-30% of ethyl acetate in hexane) to afford the product. Combined the fractions and removed all the solvents to afford the crude product as ethyl l-acetyl-5-(bromomethyl)-lH-pyrazole-3- carboxylate (1.6 g, 5.82 mmol, 57.1 % yield) MS (m/z) 232 /234 (M + H+, - Acetyl)
To a solution of N-methylaniline (42.9 mg, 0.4 mmol) in DMF (2 mL) at 20°C was added NaH (21.84 mg, 0.546 mmol). The reaction mixture was stirred for 5 min. Then ethyl l-acetyl-5-(bromomethyl)-lH-pyrazole-3-carboxylate (100 mg, 0.364 mmol) was added and the mixture were stirred at room temperature for 3 more hours. LCMS indicated the reaction was completed. The reaction was quenched with drop of water and the solvents were removed to dryness. The crude ethyl l-acetyl-5-
((methyl(phenyl)amino)methyl)-lH-pyrazole-3-carboxylate (100 mg, 91%) was used for hydrolysis without further purification. MS (m/z) 260 (M + H+ - Acetyl).
To a solution of ethyl l-acetyl-5-((methyl(phenyl)amino)methyl)-lH-pyrazole-3- carboxylate (110 mg, 0.365 mmol) in THF (2 mL) was added a solution of LiOH (1.825 mL, 3.65 mmol) in water (1.0 mL) . The reaction mixture was stirred at room temperature for 16 hours and then heated to 50°C for 4 h at which time LCMS showed hydrolysis was completed. Cooled to 0°C and added IN HCl until pH=2. The resulting solid was filtered and dried under vacuum. The 5-((methyl(phenyl)amino)methyl)-lH-pyrazole-3-carboxylic acid (80mg, 95%) was used as such without further purification. MS (m/z) = 231 (M + H+)
Preparation 79
-(3-fluorobenzyl)-4H-l,2,4-triazole-3-carboxylic acid
Figure imgf000115_0001
Freshly prepared LiOH (11.03 mL, 22.07 mmol) was added to a stirring, room temperature solution of ethyl 5-(3-fluorobenzyl)-4H-l,2,4-triazole-3-carboxylate (1.1 g, 4.41 mmol) in THF (10 mL) under N2. The reaction was then stirred at rt for 5h and LCMS showed the reaction was complete. The reaction was concentrated and then dissolved in 10.0 mL H20. 2N HCl was added dropwise until the pH= 4. The white solid that precipitated from the reaction was filtered off and washed with cold H20. The solid was dried under vacuum overnight to obtain title product.5-(3-fluorobenzyl)-4H-l,2,4-triazole- 3-carboxylic acid (750 mg, 3.39 mmol, 77 % yield). MS (m/z) 222 (M+H+).
Preparation 80
(S)-tert-butyl (l-methyl-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b][l,4]diazepin-3- yl)carbamate
Figure imgf000116_0001
To a suspension of NaH (72.1 mg, 1.803 mmol) in THF (25 mL) stirred under nitrogen at room temp was added a solution of (S)-tert-butyl (2-oxo-2, 3,4,5 -tetrahydro-1 H- benzo[b][l,4]diazepin-3-yl)carbamate (500 mg, 1.803 mmol) in THF (10 mL) dropwise during 5 min. The reaction mixture was stirred at room temperature for 1 hour and then iodomethane (0.114 mL, 1.821 mmol) was added dropwise during 2 min. The reaction mixture was stirred at room temperature for 36 hours. Reaction was quenched with water (40 mL) and extracted with EtOAc (3 x 75 mL). The organic layer was dried over anh. Na2S04 and concentrated to provide the crude product (700 mg). This was purified by silica gel column using 25-50% EtOAc in Hexane to afford (S)-tert-butyl (l-methyl-2-oxo- 2,3,4,5-tetrahydro-lH-benzo[b][l,4]diazepin-3-yl)carbamate (340 mg, 1.162 mmol, 64 % yield) as off-white solid. MS (m/z) 192.15 ([M-BOC] + H+).
Preparation 81
(S)-Tert-butyl (l,5-dimethyl-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b][l,4]diazepin-3- yl)carbamate
Figure imgf000116_0002
To a solution of (S)-tert-butyl (2-oxo-2,3,4,5-tetrahydro-lH-benzo[b][l,4]diazepin- 3-yl)carbamate (167 mg, 0.602 mmol) in DMF (2 mL) was added Cs2C03 (785 mg, 2.409 mmol) followed by Mel (0.113 mL, 1.807 mmol). The reaction mixture was stirred at room temperature overnight. The solution was diluted with EtOAc and washed with water and brine, dried over Na2S04 and concentrated. The crude material was purified by biotage column (10 to 60% EA/Hexane) to provide (S)-tert-butyl (l,5-dimethyl-2-oxo-2,3,4,5- tetrahydro-lH-benzo[b][l,4]diazepin-3-yl)carbamate (123 mg, 0.403 mmol, 66.9 % yield).
Preparation 82 (S)-tert-butyl (l-methyl-4-oxo-2,3,4,5-tetrahydro-lH-benzo[b][l,4]diazepin-3-yl)carbamate
Figure imgf000117_0001
To a suspension (S)-tert-butyl (2-oxo-2,3,4,5-tetrahydro-lH-benzo[b][l,4]diazepin-3- yl)carbamate (500 mg, 1.803 mmol) and potassium carbonate (336 mg, 2.434 mmol) in acetone (10 mL) was added iodomethane (1.240 mL, 19.83 mmol). The reaction mixture was sealed and heated 80 °C using CEM Microwave operator for 40min. After cooling, to the reaction mixture was filtered and concentrated under reduced pressure to afford the crude product (600 mg). The crude was purified by silica gel (100-200 mesh) flash chromatography using 20-40 % ethyl acetate in hexane as an eluent to afford (S)-tert-butyl (l-methyl-4-oxo-2,3,4,5-tetrahydro-lH-benzo[b][l,4]diazepin-3-yl)carbamate (390 mg, 1.298 mmol, 72.0 % yield). TLC: 30% EtOAc in Hexane; Rf: 0.35. MS (m/z) 290.21 (M+H+).
Preparation 83
2-(4-Fluoro-3-nitrophenyl)-5-methyl-l,3,4-oxadiazole
Figure imgf000117_0002
To a suspension of 4-fluorobenzohydrazide (18 g, 117 mmol) in 1, 1,1- triethoxy ethane (85 ml, 467 mmol) was heated at 150 °C. After refluxing for 24 hr at 150 °C, the reaction solution was cooled down to room temperature, then a solid precipitated out. After flushing with nitrogen for 2-3 minutes, the resultant solid was treated with 1-2% ethyl ether in hexane. After collecting and washing solid with hexane followed by drying in vacuo at 50°C, 2-(4-fluorophenyl)-5-methyl-l,3,4-oxadiazole (17.80 g, 99 mmol, 85 % yield) was obtained as light brown solid: MS (m/z) 179.0 (M+H+); 1H MR (DMSO-d6) δ: 7.97 - 8.10 (m, 2H), 7.38 - 7.51 (m, 2H), 2.58 (s, 3H).
To a suspension (a partial solution, close to dark red color) of 2-(4-fluorophenyl)-5- methyl-l,3,4-oxadiazole (2 g, 11.23 mmol) in concentrated (fuming) H2S04 (8 ml) at 0 °C was added nitric acid (1.394 ml, 28.1 mmol) (the solution color was changed to orange yellow). After 30 min at 0°C, the reaction solution was poured into cold water (around 300 ml). The resultant solid was collected by filtration and washed with water. The light tan solid was dried over in a vacuum oven at 50°C for overnight to give 2-(4-fluoro-3- nitrophenyl)-5-methyl-l,3,4-oxadiazole (2.28 g, 10.22 mmol, 91 % yield); MS (m/z) 224.0 (M+H+); 1H NMR (DMSO-d6) δ: 8.61 (dd, J = 7.1, 2.3 Hz, 1H), 8.38 (ddd, J = 8.8, 4.3, 2.3 Hz, 1H), 7.84 (dd, J = 11.1, 8.8 Hz, 1H), 2.62 (s, 3H).
The following intermediates used for the preparation of titled example compounds were syn hesized using methods analogous to the ones described above.
Figure imgf000118_0001
Preparation 84
(S)-3-amino-5-methyl-8-(5-methyl-l,3,4-oxadiazol-2-yl)-2,3- dihydrobenzo[b] [ 1 ,4]oxazepin-4(5H)-one, bis hydrochloride · chloride
Figure imgf000118_0002
A mixture of (S)-ethyl 3-((tert-butoxycarbonyl)amino)-5-methyl-4-oxo-2,3,4,5- tetrahydro benzo[b] [1,4] oxazepine-8-carboxylate (0.45 g, 1.235 mmol) and anhydrous hydrazine (0.465 mL, 14.82 mmol) in EtOH (5.0 mL) was heated to reflux overnight. The solvent was removed in vacuo then the resulting residue was suspended in ethyl ether, filtered and washed with ethyl ether having 5% ethanol to yield (S)-tert-butyl (8- (hydrazinecarbonyl)-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b] [1,4] oxazepin-3- yl)carbamate (253.0mg, 58.5 %) ; MS (m/z): 351.3 (M+H+).
A suspension of (S)-tert-butyl (8-(hydrazinecarbonyl)-5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b] [l,4]oxazepin-3-yl)carbamate (253.0 mg, 0.722 mmol) in 1,1,1- triethoxy ethane (2896 μΐ, 15.89 mmol) was heated to 150 °C under nitrogen. After 2h the reaction mixture was cooled to room temperature. On cooling, the solid product precipitated and was collected by filtration then washed with a small amount of ethyl ether to give (S)-tert-butyl (5-methyl-8-(5-methyl-l,3,4-oxadiazol-2-yl)-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate (165.0 mg, yield 61 %). 1H MR
(DMSO-de) d: 7.89 (dd, J = 8.5, 1.9 Hz, 1H), 7.66 - 7.78 (m, 2H), 7.25 (d, J = 6.6 Hz, 1H), 4.42 (br. s., 3H), 3.33 (br. s., 3H), 2.60 (s, 3H), 1.35 (s, 9H) ; MS (m/z): 375.3 (M+H+).
To a solution of (S)-tert-butyl (5-methyl-8-(5-methyl-l,3,4-oxadiazol-2-yl)-4-oxo- 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate (165.0 mg, 0.441 mmol) in DCM (3.0 mL) was added a solution of 4M HC1 in 1,4 dioxane (1.653 mL, 6.61 mmol). The mixture was stirred at room temperature for lh. The solid product precipitated from the reaction mixture and it was collected by filtration, then washed with ethyl ether, to yield (S)-3-amino-5-methyl-8-(5-methyl-l,3,4-oxadiazol-2-yl)-2,3- dihydrobenzo[b][l,4]oxazepin-4(5H)-one, bis hydrochloride- chloride salt (150.0 mg, 89%). MS (m/z): 275.1 (M+H+).
Preparation 85
2-amino-2-(2-(2-phenylacetyl)hydrazono)acetate
Figure imgf000119_0001
To a solution of 2-phenylacetohydrazide (20 g, 133 mmol) in ethanol (75 mL) and diethyl ether (250 mL) was added ethyl 2-ethoxy-2-iminoacetate (20 g, 138 mmol). Stirred at room temperature for 4 hours. A precipitate started to form after 10 minutes. The resulting solid was filtered off, rinsed with diethyl ether and dried to give ethyl 2-amino-2- (2-(2-phenylacetyl)hydrazono)acetate (27.85 g, yield 82%) as a white solid, which was used without further purification. MS (m/z) 250 (M+H+). Preparation 86
Ethyl 5 -b enzy 1 -4H- 1 ,2, 4-tri azol e-3 -carb oxy 1 ate
Figure imgf000120_0001
Ethyl 2-amino-2-(2-(2-phenylacetyl)hydrazono)acetate (27.85 g, 109 mmol) was suspended in xylenes (300 mL) and heated at 170 °C for 24 hours with a Dean-Stark trap. Initially a very thick mixture formed which became light yellow and homogeneous as the reaction progressed. The reaction was cooled to room temperature and a solid precipitated out. Diethyl ether was added and the reaction mixture stirred for 15 minutes in an ice/water bath. The solid was filtered off, rinsing with diethyl ether and hexanes and dried to give ethyl 5-benzyl-4H-l,2,4-triazole-3-carboxylate (24.67 g, 95% yield) as a white solid, which was used without further purification. MS (m/z) 232 (M+H+).
Preparation 87
(S)-tert-butyl (6,8-difluoro-7-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate
Figure imgf000120_0002
To a solution of l,3,5-trifluoro-2-methylbenzene (6.0 g, 41.1 mmol) in sulfuric acid (46.0 ml, 862 mmol) stirred at -10°C was added a mixture of nitric acid (1.835 ml, 41.1 mmol) in sulfuric acid (15.32 ml, 287 mmol) dropwise over 10 min. The reaction mixture was stirred at -10 to 15 °C for 1.5 hr, at which time TLC indicated starting material was consumed. The reaction mixture was poured on to ice-water (500 mL), extracted with DCM (250 mL), and the organic layer was washed with water (100 mL), dried over anh. Na2S04 and concentrated to afford l,3,5-trifluoro-2-methyl-4-nitrobenzene (6.9 g, 31.3 mmol, 76 % yield) as yellow liquid. TLC: 20% EtOAc in hexane; Rf: 0.65. GCMS (m/z) 191 (M+).
To a solution of l,3,5-trifluoro-2-methyl-4-nitrobenzene (6.9 g, 36.1 mmol) and NaH (2.89 g, 72.2 mmol) in DMF (70 mL) stirred under nitrogen at 0°C was added a solution of (S)-2-((tert-butoxycarbonyl)amino)-3-hydroxypropanoic acid (7.41 g, 36.1 mmol) in DMF (70 mL) dropwise during 5 min. The reaction mixture was stirred at 0-25 °C for 3 hr at which time TLC indicated starting material was consumed. 0.5M HCl (250 mL) was then added, and the aqueous phase extracted with EtOAc (500 mL). The organic layer was washed with water (100 mL x 2), followed by brine (100 mL). The organic layer was dried over anh. Na2S04, filtered and concentrated to yield the crude (S)-2-((tert- butoxycarbonyl)amino)-3-(3,5-difluoro-2-methyl-6-nitrophenoxy)propanoic acid and (S)-2- ((tert-butoxycarbonyl)amino)-3-(3,5-difluoro-4-methyl-2-nitrophenoxy)propanoic acid (1 : 1 mixture) (12 g, 32 mmol, 89 % yield) as yellow gum. This crude compound was carried over to next step. TLC: 10%MeOH in DCM; Rf: 0.3. MS (m/z) 375 (M-H+).
To a solution of (S)-2-((tert-butoxycarbonyl)amino)-3-(3,5-difluoro-2-methyl-6- nitrophenoxy)propanoic acid compound and (S)-2-((tert-butoxycarbonyl)amino)-3-(3,5- difluoro-4-methyl-2-nitrophenoxy)propanoic acid (1 : 1) (12 g, 32 mmol) in methanol (250 mL) stirred under nitrogen at room temp was added palladium on carbon (2.55 g, 2.392 mmol) portionwise during 5 min. The contents were stirred under hydrogen at a pressure of 60 PSI at 25 °C for 3 hr, at which time TLC indicated starting material was consumed. The reaction mixture was filtered through a celite pad, washed with excess methanol (300 mL) and concentrated to provide the crude (S)-3-(2-amino-3,5-difluoro-4-methylphenoxy)-2- ((tert-butoxycarbonyl)amino)propanoic acid compound and (S)-3-(2-amino-3,5-difluoro-6- methylphenoxy)-2-((tert-butoxycarbonyl)amino)propanoic acid (1 : 1 mixture) (9 g, 92% recovery). TLC: 10% MeOH in DCM; Rf: 0.4. This compound was used to next step without further purification.
To a solution of (S)-3-(2-amino-3,5-difluoro-4-methylphenoxy)-2-((tert- butoxycarbonyl)amino)propanoic acid compound and (S)-3-(2-amino-3,5-difluoro-6- methylphenoxy)-2-((tert-butoxycarbonyl)amino)propanoic acid (1 : 1) (9 g, 26.0 mmol) and DIEA (6.81 mL, 39.0 mmol) in DMSO (90 mL) stirred under nitrogen at 20°C was added HATU (9.88 g, 26.0 mmol) portionwise during 5 min. The reaction mixture was stirred at 25 °C for 20 h, at which time TLC indicated starting material was consumed. Reaction was quenched with water (500 mL), extracted with EtOAc (300 mL) and the organic layer was dried over anh. Na2SC>4, filtered and concentrated to yield the crude product. This was purified by prep. HPLC to afford a regioisomeric mixture of (S)-tert-butyl (6,8-difluoro-7- methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate and (S)-tert-butyl (6,8-difluoro-9-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate (1.98 g, 23%) as a pale brown solid. The desired regioisomer was isolated by chiral HPLC (column: Ο1ι^^- :(250*30*5.0μ), mobile phase: n-hexane:IPA ( 80:20), flow rate: 30 ml/min) of the regioisomeric mixture (2.57 g, 7.8 mmol) to afford (S)-tert-butyl (6,8- difluoro-7-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)carbamate (710 mg, 27% recovery) as an off-white solid. 1H MR (DMSO-d6) δ: 9.8 (s, 1H), 7.15 (m, 1H), 6.95 (m, 1H), 4.35 (m, 3H), 3.15 (s, 3H), 1.35 (s, 9H). MS (m/z) 329 (M+H+).
Preparation 88
7-Chloro-9-fluoro-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one
Figure imgf000122_0001
water
To a solution of 6-chloro-8-fluoro-3,4-dihydronaphthalen-l(2H)-one (3.2 g, 16.11 mmol) in ethanol (32 mL) and water (16 mL) stirred in air at room temp was added sodium acetate (2.64 g, 32.2 mmol) and hydroxylamine hydrochloride (2.239 g, 32.2 mmol) in one charge. The reaction mixture was stirred at 100 °C for lh. Reaction was monitored by TLC using 20%) of ethyl acetate in hexane as mobile phase (twice elution). On completion of the reaction, the reaction mixture was evaporated to dryness and 25mL of water was added. The suspension was stirred for 5 min and filtered, washed with water (10 mL), hexane (10 mL) and dried under vacuum to give 6-chloro-8-fluoro-3,4-dihydronaphthalen-l(2H)-one oxime (3.35g, 15.68 mmol, 97 %> yield) as a pale brown solid, as a 1 : 1 mixture of syn and anti stereoisomers. MS (m/z) 214/216 (M+H+). To a solution of 6-chloro-8-fluoro-3,4-dihydronaphthalen-l(2H)-one oxime (3.2 g, 14.98 mmol) and TEA (6.26 mL, 44.9 mmol) in DCM (90 mL) stirred under nitrogen at 5°C was added tosyl-chloride (8.57 g, 44.9 mmol) portionwise during 5 min. The reaction mixture was allowed to stir at rt for 18 hr. After completion of the reaction (monitored by TLC, 20% ethyl acetate in hexane), the reaction mixture was diluted with water, extracted with DCM (2 x 90 mL). The organic layer was separated and washed with water (2 x 50mL), and the combined organic phases were washed with brine, dried over anhydrous sodium sulfate and evaporated under reduced pressure to afford the crude product. The crude product was added to a silica gel column and was eluted with Hexane/EtOAc.
Collected product fractions: from 5-6% EtOAc elution were concentrated under reduced pressure to afford 6-chloro-8-fluoro-3,4-dihydronaphthalen-l(2H)-one O-tosyl oxime (3.0g, 6.61 mmol, 44.1 % yield) as a light brown solid, as a 4 : 1 unassigned mixture of syn and anti stereoisomers. 1H NMR (DMSO-d6) δ: 7.95 (m, 2H), 7.35 (m, 2H), 7.0 (m, 2H), 2.85 (m, 2H), 2.65 (m, 2H), 1.8 (m, 2H); MS (m/z) 368/370 (M+H+).
To a solution of 6-chloro-8-fluoro-3,4-dihydronaphthalen-l(2H)-one O-tosyl oxime
(3g, 8.16 mmol) in ethanol (150 mL) and water (80 mL) was added potassium acetate (17.61 g, 179 mmol) and reaction mixture was stirred at 100 °C for 16h. After the completion of the reaction (monitored the reaction by TLC 30% ethyl acetate in hexane), the reaction mixture was evaporated under reduced pressure to remove the ethanol, remaining aqueous layer was further diluted with water (5mL) and cooled for 30min at 5°C. The resulting solid precipitate formed was filtered off, washed with cold water, hexane and dried under vacuum to afford 7-chloro-9-fluoro-4,5-dihydro-lH- benzo[b]azepin-2(3H)-one (lg, 4.16 mmol, 51.0 % yield) as brown solid. 1H MR
(DMSO-de) δ: 9.43 (s, 1H), 7.35 (d, 1H), 7.22 (s, 1H), 2.75 (m, 2H), 2.15 (m, 4H); MS (m/z) 213/215 (M+H+).
Example 1
Method A
(R)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]thiazepin-3-yl)isoxazole-
3-carboxamide
Figure imgf000124_0001
To a solution of 5-benzylisoxazole-3-carboxylic acid (208 mg, 0.919 mmol) in DCM (30 mL) was added Nl-((ethylimino)methylene)-N3,N3-dimethylpropane-l,3- diamine hydrochloride (129 mg, 0.674 mmol) and lH-benzo[d][l,2,3]triazol-l-ol hydrate (103 mg, 0.674 mmol), then 4-methylmorpholine (0.202 mL, 1.839 mmol). Stirred at rt for 5min, then added (R)-3-amino-5-methyl-2,3-dihydrobenzo[b][l,4]thiazepin-4(5H)-one, hydrochloride (150 mg, 0.613 mmol). The reaction mixture was stirred at 25 °C for 5h. LCMS showed product and the reaction was completed. Removed all the DCM and added 200 ml of EtOAc and the mixture was washed with water, 0. IN HC1 aq, NaHC03 aq and brine. The organic phase was dried over Na2S04 and concentrated under reduced pressure to afford crude product. ISCO purification (eluting with 0-50% of EtOAc in hexane ) to afford the title compound as (R)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]thiazepin-3-yl)isoxazole-3-carboxamide. The residue was triturated with ether and hexane. The resulting solid was filtered and rinsed with hexane and collected (200 mg, 83%). 1H NMR (400MHz, DMSO-d6) δ = 8.96 (d, J= 7.8 Hz, 1 H), 7.67 (d, J = 7.3 Hz, 1 H), 7.59 (d, J= 4.0 Hz, 2H), 7.44 - 7.19 (m, 6 H), 6.52 (s, 1 H), 4.62 - 4.43 (m, 1 H), 4.21 (s, 2 H), 3.52 (dd, 7= 6.8, 11.4 Hz, 1 H), 3.71- 3.44 (m, 1 H), 3.30 (s, 3 H). MS (m/z) 394 (M+H+).
Example 2
Method B
(R)-5-benzyl-N-(5-methyl-l,l-dioxido-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]thiazepin-3- yl)isoxazole-3-carboxamide
Figure imgf000124_0002
To a solution of (R)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]thiazepin-3-yl)isoxazole-3-carboxamide (69 mg, 0.175 mmol) in DCM (15 mL) was added 3-chlorobenzoperoxoic acid (101 mg, 0.438 mmol) at 0 °C. The reaction mixture was stirred for 1 hr at 0 °C and warmed up to rt, then stirred for 12 hr at rt. The reaction was quenched with cold IN aq. NaOH solution. After extraction with DCM, the combined organic solution was washed with 2.5% aq. Na2S203 solution and brine. After drying over MgS04, filtration, and concentration, the residue was subjected to the column chromatography (ISCO 40g, eluent: 5% to 50% EtO Ac/Hex) to provide (R)-5-benzyl-N-(5- methyl-l, l-dioxido-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]thiazepin-3-yl)isoxazole-3- carboxamide (60 mg, 0.141 mmol, 80 % yield) as gum. Trituration with DCM and hexane to afford the white solid. 1H NMR (400MHz, DMSO-d6) 5 = 9.14 (d, J= 7.6 Hz, 1 H), 8.06 - 7.90 (m, 2 H), 7.81 (d, J = 8.1 Hz, 1 H),
7.64 (t, J= 7.6 Hz, 1 H), 7.49 - 7.19 (m, 5 H), 6.52 (s, 1 H), 5.76 (s, 1 H), 4.72 (dt, J = 7.5, 11.2 Hz, 1 H), 4.22(s, 2 H), 4.14 - 3.95 (m, 2 H), 3.29 (s, 3 H). MS (m/z) 426 (M+H+).
Examples 3 and 4
Method C
5-benzyl-N-((l S,3R)-5-methyl-l-oxido-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]thiazepin-3- yl)isoxazole-3-carboxamide and 5-benzyl-N-((lR,3R)-5-methyl-l-oxido-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]thiazepin-3-yl)isoxazole-3-carboxamide
Figure imgf000125_0001
To a solution of (R)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]thiazepin-3-yl)isoxazole-3-carboxamide (75 mg, 0.191 mmol) in DCM (15 mL) was added 3-chlorobenzoperoxoic acid (54.8 mg, 0.238 mmol) at 0 °C. The reaction mixture was stirred for 1 hr at 0 °C and warmed up to rt, then stirred for 12 hr at rt. The reaction was quenched with cold IN aq. NaOH solution. After extraction with DCM, the combined organic solution was washed with 2.5% aq. Na2S203 solution and brine. After drying over MgS04, filtration, and concentration, the residue was subjected to the column chromatography (ISCO 40g, eluent: 0% to 40%, then to 60% EtO Ac/Hex) to provide 2 isomers as 5-benzyl-N-((lR,3R)-5-methyl-l-oxido-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]thiazepin-3-yl)isoxazole-3-carboxamide (64 mg, 0.156 mmol, 82 % yield)) and 5-benzyl-N-((l S,3R)-5-methyl-l-oxido-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]thiazepin-3-yl)isoxazole-3-carboxamide (14 mg, 0.034 mmol, 17.94 % yield. 5-benzyl-N-((l S,3R)-5-methyl-l-oxido-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]thiazepin-3-yl)isoxazole-3-carboxamide. MS (m/z) 410 (M+H+). 1H NMR (400MHz, DMSO-d6) δ = 9.06 (d, J = 7.8 Hz, 1 H), 7.88 - 7.75 (m, 1 H), 7.73 - 7.63 (m, 2 H), 7.55 -7.44 (m, 1 H), 7.42 - 7.20 (m, 4 H), 6.54 (s, 1 H), 5.76 (s, 1 H), 4.73 (dt, J = 7.6, 11.1 Hz, 1 H), 4.22 (s, 2 H),3.83 (dd, J = 7.5, 14.5 Hz, 1 H), 3.53 (dd, J = 11.1, 14.4 Hz, 1 H), 3.25 (s, 3 H). 5-benzyl-N-((lR,3R)-5-methyl-l-oxido-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]thiazepin-3-yl)isoxazole-3-carboxamide. MS (m/z) 410 (M+H+). 1H MR (DMSO-d6) 8.89 - 9.19 (m, 1H), 7.56 - 7.86 (m, 4H), 7.22 - 7.48 (m, 5H), 6.51 (s, 1H), 4.45 - 4.65 (m, 2H), 4.13 - 4.35 (m, 3H), 3.30 (s, 3H). Example 5
Method D
3-Benzyl-N-((S)-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)piperidine-
1-carboxamide
Figure imgf000126_0001
To a suspension of (S)-3-amino-5-methyl-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)- one, hydrochloride (100 mg, 0.437 mmol) and Et3N (0.152 ml, 1.093 mmol) in THF (4 ml) was added 4-nitrophenyl carbonochloridate (97 mg, 0.481 mmol) at 0 °C. After 45 min, 3- benzylpiperidine (0.085 ml, 0.481 mmol) and Et3N (0.091 ml, 0.656 mmol) were added and warmed up to rt. After 2 hr at rt, the reaction mixture was concentrated, then diluted with MeOH-DMSO (2 mL, 1 : 1). After filtration through Acrodisc CR 25 mm syringe filter with 0.2 uM PTFE membrane, the solution was purified by HPLC (Waters, column: Waters Sunfire 30x150mm, eluent: Acetonitrile:Water TFA 50-100%, flow rate: 50 ml/min) to give 3-benzyl-N-((S)-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3- yl)piperidine-l-carboxamide (52.2 mg, 0.123 mmol, 28.2 % yield, unoptimized) as a mixture of diastereomers. MS (m/z) 393.9 (M+H+). After separation using a chiral column (Chiralpak IA-H, eluent: Co-solvent: IP A, % Co-solvent: 30% Isocratic, Flow rate = 4 mL/min), two diastereomers were obtained without assigning the absolute stereochemistry at the C-3 piperdine. Isomer A (9.9 mg): 1H MR (CDC13) δ: 7.14-7.36 (m, 9H), 5.50 (d, J = 6.3 Hz, 1H), 4.86 (dt, J = 11.1, 6.8 Hz, 1H), 4.63 (dd, J = 9.7, 7.5 Hz, 1H), 4.16 (dd, J = 11.2, 9.7 Hz, 1H), 3.87 (dt, J = 13.1, 1.6 Hz, 1H), 3.72 - 3.81 (m, 1H), 3.43 (s, 3H), 2.88 (ddd, J = 12.9, 11.4, 3.2 Hz, 1H), 2.52 - 2.69 (m, 2H), 2.45 (dd, J = 13.6, 8.1 Hz, 1H), 1.65 - 1.81 (m, 3H), 1.36 - 1.46 (m, 1H), 1.06 - 1.18 (m, 1H). MS (m/z) 393.9 (M+H+). Isomer B (13.9 mg): 1H NMR (CDC13) δ: 7.09 - 7.73 (m, 9H), 5.51 (br. s., 1H), 4.79 - 4.92 (m, 1H), 4.59 - 4.70 (m, 1H), 4.10 - 4.20 (m, 1H), 3.89 (m, 1H), 3.77 (m, 1H), 3.42 (s, 3H), 2.77 - 2.93 (m, 1H), 2.40 - 2.67 (m, 3H), 1.63 - 1.81 (m, 3H), 1.38 - 1.49 (m, 1H), 1.06 - 1.20 (m, 1H). MS (m/z) 393.9 (M+H+).
Example 6
Method E
3-Benzyl-N-((S)-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)piperidine-
1-carboxamide
o
Figure imgf000127_0001
Example 21
Polymer bound PPh3 (1.6 mmol/g loading, 2.5 eq, 268 mg), (S)-5-benzyl-N-(8- hydroxy-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)isoxazole-3-carboxamide (65 mg, 0.171 mmol) and 2-methoxyethanol (0.027 mL, 0.343 mmol) were combined in THF (3 mL). Next added di-tert-butyl azodicarboxylate (79 mg, 0.343 mmol) and stirred mixture at rt for 20 hours. TFA (0.066 mL, 0.857 mmol) was added and mixture was stirred for 1 hour. Reaction was filtered through Celite, rinsing with 10% MeOH/DCM and concentrated. Crude was partitioned between DCM and satd. NaHC03 and layers were separated. Organics were concentrated and purified by flash chromatography, Biotage (4 g silica column; 0.5-3% MeOH/DCM (plus NH4OH), 15 min.) to give 37 mg white foam in 48% yield. 1H NMR (DMSO-d6) δ: 9.92 (s, 1H), 8.77 (d, J = 8.1 Hz, 1H), 7.25 - 7.40 (m, 5H), 7.02 (d, J = 8.8 Hz, 1H), 6.70 - 6.79 (m, 2H), 6.57 (s, 1H), 4.80 (dt, J = 10.8, 7.4 Hz, 1H), 4.50 (t, J = 10.6 Hz, 1H), 4.41 (dd, J = 10.5, 6.7 Hz, 1H), 4.23 (s, 2H), 4.01 - 4.13 (m, 2H), 3.60 - 3.68 (m, 2H), 3.30 (s, 3H); MS (m/z) 438.3 (M+H+).
Example 7
5-Benzyl-N-(l-hydroxy-5-methyl-4-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3- yl)isoxazole-3-carboxamide
Figure imgf000128_0001
Benzyl (2,5-dioxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate (400 mg, 1.233 mmol) was dissolved in 10 mL of THE, and then CS2CO3 (1.0 g, 3.08 mmol) was added, followed by methyl iodide (0.116 mL, 1.850 mmol). The reaction mixture was maintained at room temperature for 16 hours. The mixture was then filtered. The filtrate was concentrated, and then purified by Isco Combiflash (15 %-80 % EtOAc/Hexane; 40 g Isco RediSep column). The fractions containing the product were combined and concentrated to give benzyl (l-methyl-2,5-dioxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3- yl) carbamate as a yellow oil (290 mg, 70 % yield). 1H NMR (CDC13) d ppm 7.53 - 7.72 (m, 2H), 7.26 - 7.44 (m, 6H), 7.21 (d, J = 8.1 Hz, 1H), 6.14 (d, J = 6.6 Hz, 1H), 5.02 - 5.18 (m, 2H), 4.95 (ddd, J = 12.6, 6.6, 4.0 Hz, 1H), 3.38 (s, 3H), 3.33 (dd, 1H), 2.94 (dd, J = 19.3, 12.8 Hz, 1H); MS (m/z): 339 (M+H+).
Benzyl (l-methyl-2,5-dioxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl) carbamate (290 mg, 0.857 mmol) was dissolved in ethanol (20 mL). Palladium on Carbon (lOwt % loading, 91 mg, 0.857 mmol) was added. The reaction mixture was maintained at room temperature for 3 h under a hydrogen balloon. The reaction mixture was filtered and the filtrate was concentrated to a yellow oil, which was then turned into white solid upon standing under high vacuum for 16 h to yield 3-amino-l-methyl-3,4-dihydro-lH- benzo[b]azepine-2,5-dione (158 mg, 90 % yield). 1H NMR (MeOH-d4) d ppm 7.60 - 7.88 (m, 2H), 7.23 - 7.59 (m, 2H), 4.50 - 4.72 (m, 1H), 3.43 (s, 3H), 3.11 - 3.38 (m, 1H), 2.08 - 2.31 (m, 1H); MS (m/z): 205 (M+H+).
5-Benzylisoxazole-3-carboxylic acid (29.8 mg, 0.147 mmol) was dissolved in
DCM(2 mL), and then N-hydroxybenzotriazole (24.75 mg, 0.162 mmol) and EDC (31.0 mg, 0.162 mmol) were added. The mixture was maintained at room temperature for lOmin. N-methylmorpholine (0.057 mL, 0.514 mmol) and 3-amino-5-methyl-2,3-dihydro-lH- benzo[b]azepine-l,4(5H)-dione (30 mg, 0.147 mmol) were then added. The reaction mixture was maintained at room temperature for 16 h. The mixture was then concentrated and the residue was purified by Isco Combiflash (10%-50% EtOAc/Hexane; 24 g Isco RediSep column). The fractions containing the product were combined and concentrated to give 5-benzyl-N-(5-methyl-l,4-dioxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3- yl)isoxazole-3-carboxamid as a clear oil, which turned into a white solid upon standing under high vacuum for 16 h (42 mg, 73 % yield). 1H NMR (400 MHz, CDC13) d ppm 3.00 (dd, J=19.33, 12.76 Hz, 1 H) 3.39 - 3.48 (m, 4 H) 4.13 (s, 2 H) 5.26 (ddd, J=12.76, 6.44, 3.79 Hz, 1 H) 6.35 (s, 1 H) 7.19 - 7.43 (m, 7 H) 7.55 - 7.72 (m, 2 H) 7.99 (d, J=6.57 Hz, 1 H); MS (m/z): 390 (M+H+).
5-Benzyl-N-(5-methyl-l,4-dioxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3- yl)isoxazole-3-carboxamide (20 mg, 0.051 mmol) was dissolved in 2mL of MeOH, and then NaBH4 (2.91 mg, 0.077 mmol) was added at rt. The mixture was maintained at rt for 16 h. The mixture was then concentrated and partitioned between sat.NaHC03 (aq) and DCM. The organic layer was concentrated and the residue was purified by Isco Combiflash (1 %-10 % MeOH/CH2Cl2, 10% NEt3 in MeOH; 4 g RediSep column). Collected fractions containing the product were combined and concentrated to give 5-benzyl-N-(l-hydroxy-5- methyl-4-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)isoxazole-3-carboxamide as a clear oil (14 mg, 70 % yield). 1H MR (400 MHz, CDC13) δ ppm 2.34 (d, J=4.55 Hz, 1 H) 2.44 (ddd, J=12.13, 10.86, 7.58 Hz, 1 H) 2.67 (td, J=11.68, 8.21 Hz, 1 H) 3.43 (s, 3 H) 4.12 (s, 2 H) 4.51 (dt, J=10.48, 7.89 Hz, 1 H) 5.06 (t, J=3.66 Hz, 1 H) 6.32 (s, 1 H) 7.13 - 7.45 (m, 8 H) 7.67 (dd, J=6.69, 2.15 Hz, 1 H) 7.81 (d, J=7.07 Hz, 1 H); MS (m/z): 392 (M+H+). Example 8
(S)-5-benzyl-N-(5-methyl-4-oxo-7-(lH-tetrazol-5-yl)-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)isoxazole-3-carboxamide
Figure imgf000130_0001
Step 1 : (S)-5-benzyl-N-(7-(l-(2-cyanoethyl)-lH-tetrazol-5-yl)-5-methyl-4-oxo-
2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)isoxazole-3-carboxamide. To a solution of (S)-3-(5- benzylisoxazole-3-carboxamido)-N-(2-cyanoethyl)-5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine-7-carboxamide (69.0 mg, 0.146 mmol) and pyridine
(0.071 mL, 0.874 mmol) in DCM (2.0 mL) was added phosphorus pentachloride (45.5 mg, 0.219 mmol). The reaction mixture was heated to reflux for 3.0 hr then additional 0.25 eq PC15 was added. The reaction mixture was cooled to rt then TMSN3 (0.110 mL, 0.831 mmol) was added and the reaction mixture was stirred overnight at rt. At 20 h, additional 4.0 eq. of TMS-N3 and 3.0 eq. pyridine were added to the reaction mixture. The reaction mixture was carefully quenched with a few drops of sat. aq. NaHC03 followed after 5 min with excess NaHC03. The mixture was stirred for 15 min. The organic phase was separated and washed with 10 % aq citric acid and brine. The organic phase was dried over Na2S04, filtered and concentrated in vacuo. The residue was purified by FCC (EtO Ac-Hex: 50 -70 %). MS (m/z) 499.3 (M+H+).
Step 2: (S)-5-benzyl-N-(5-methyl-4-oxo-7-(lH-tetrazol-5-yl)-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)isoxazole-3-carboxamide. To a solution of (S)-5- benzyl-N-(7-(l-(2-cyanoethyl)-lH-tetrazol-5-yl)-5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)isoxazole-3-carboxamide (42.0 mg, 0.084 mmol) in THF (2.0 mL) was added 2.0 M NaOH (0.051 mL, 0.101 mmol). The reaction mixture was stirred for 2h then quenched with cold IN HC1 and extracted with EtO Ac. The organic phase was washed with brine, dried over Na2S04, filtered then concentrated in vacuo. The solid product obtained was used without further purification (36.0 mg, 96%). 1H NMR (DMSO-de) δ: 8.92 (d, J = 8.1 Hz, 1H), 8.12 (d, J = 2.0 Hz, 1H), 7.94 (dd, J = 8.3, 2.3 Hz, 1H), 7.46 (d, J = 8.3 Hz, 1H), 7.25 - 7.39 (m, 6H), 6.55 (s, 1H), 4.87 - 4.96 (m, 1H), 4.67 (dd, J = 11.6, 10.1 Hz, 1H), 4.49 (dd, J = 9.9, 7.6 Hz, 1H), 4.22 (s, 2H), 3.39 (s, 3H). MS (m/z) 446.3 (M+H+).
Example 9
(S)-3-(5-benzylisoxazole-3-carboxamido)-5-methyl-N-(methylsulfonyl)-4-oxo-2,3,4,5- tetrahydro benzo[b] [ 1 ,4]oxazepine-7-carboxamide
Figure imgf000131_0001
To a suspension of (S)-3-(5-benzylisoxazole-3-carboxamido)-5-methyl-4-oxo- 2,3,4,5-tetrahydro benzo[b][l,4]oxazepine-7-carboxylic acid (60.0 mg, 0.142 mmol) in DCM (2.0 mL) was added l-chloro-N,N,2-trimethylprop-l -en- 1 -amine (22.83 mg, 0.171 mmol) as a solution in DCM (0.10 ml) dropwise over 1 min. The reaction mixture was stirred at rt for lh and became a homogeneous solution. This mixture was added dropwise to a mixture of methanesulfonamide (54.2 mg, 0.570 mmol), TEA (0.079 mL, 0.570 mmol) and DMAP (1.044 mg, 8.54 μιηοΐ) in 1.0 mL DCM and stirring was continued over 2h at rt. The reaction mixture was diluted with EtOAc, then washed with 10% aq citric acid, water and brine. The organic phase was dried over Na2SC>4, filtered then concentrated in vacuo. The residue was purified by FCC[MeOH-DCM: 0-4.0 %] to yield the desired product (26.0 mg, 36.6%). 1H MR (DMSO-de) δ: 8.82 (d, J = 8.1 Hz, 1H), 7.95 (d, J = 1.8 Hz, 1H), 7.83 (dd, J = 8.3, 2.0 Hz, 1H), 7.25 - 7.38 (m, 6H), 7.20 (d, J = 8.1 Hz, 1H), 6.56 (s, 1H), 4.84 (dt, J = 11.6, 7.8 Hz, 1H), 4.59 (dd, J = 11.7, 10.0 Hz, 1H), 4.42 (dd, J = 9.7, 7.7 Hz, 1H), 4.22 (s, 2H), 3.32 (br. s., 3H), 2.95 (s, 3H). MS (m/z) 499.1 (M+H+). Example 10
Method F
(S)-5-benzyl-N-(7-fluoro-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)isoxazole-3- carboxamide
Figure imgf000132_0001
To a solution of 5-benzylisoxazole-3-carboxylic acid (79 mg, 0.390 mmol) and DIEA (0.186 mL, 1.064 mmol) in DMSO (1 mL) was added HATU (135 mg, 0.355 mmol) in one portion. After stirring at rt for 5 min, a solution of (S)-3-amino-7-fluoro-2,3- dihydrobenzo [b][l,4]oxazepin-4(5H)-one, trifluoroacetic acid salt (110.0 mg, 0.355 mmol) in DMSO (1 mL) was added dropwise to the mixture. The reaction was allowed to stir at rt for 2 h. LCMS analysis indicated starting material still remained. An additional amount of DIEA (0.20 mL) and HATU (0.11 g) were added and the reaction allowed to stir for 2 h. The reaction mixture was diluted with EtOAc then washed with water (3x), H4CI and brine. After drying the solution over Na2S04 and concentrating in vacuo, the residue was purified by FCC [EtOAc/Hex: 25-60%] to yield the desired product (50 mg, 37 %). 1H
MR (DMSO-de) δ: 10.21 (s, 1H), 8.88 (d, J = 8.1 Hz, 1H), 7.25 - 7.40 (m, 5H), 7.17 (dd, J = 8.6, 5.6 Hz, 1H), 6.92 - 7.00 (m, 2H), 6.55 - 6.59 (m, 1H), 4.83 (dt, J = 10.5, 7.5 Hz, 1H), 4.38 - 4.53 (m, 2H), 4.23 (s, 2H). MS (m/z) 382.9(M+H+).
Example 11
Method G
(S)-5-benzyl-N-(7-(3-isopropylureido)-5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-yl)isoxazole-3-carboxamide
Figure imgf000132_0002
To a solution of (S)-N-(7-amino-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b]
[l,4]oxazepin-3-yl)-5-benzylisoxazole-3-carboxamide (50.0 mg, 0.112 mmol) in DMF (0.50 mL) at 0 °C was added 2-isocyanatopropane (0.023 mL, 0.235 mmol). After 2 d, additional 2-isocyanatopropane (0.023 mL, 0.235 mmol) was added and the reaction was continued. The reaction mixture was diluted with EtOAc then it was washed in succession with sat. H4CI, water, and brine. The organic phase was dried over Na2S04, filtered then concentrated in vacuo. The residue was purified by FCC [EtOAc/Hex-45 -80 %]. 1H MR (DMSO-de) δ: 8.83 (d, J = 8.3 Hz, 1H), 8.44 (s, 1H), 7.57 (d, J = 2.5 Hz, 1H), 7.25 - 7.38 (m, 5H), 7.12 - 7.16 (m, 1H), 7.05 - 7.09 (m, 1H), 6.55 (s, 1H), 6.06 (d, J = 7.6 Hz, 1H), 4.83 (dt, J = 11.4, 8.1 Hz, 1H), 4.46 - 4.53 (m, 1H), 4.33 (dd, J = 9.9, 7.8 Hz, 1H), 4.22 (s, 2H), 3.71 - 3.81 (m, 1H), 3.26 (s, 3H), 1.11 (s, 3H), 1.09 (s, 3H). MS (m/z) 478.2 (M+H+).
Example 12
Method H
(S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-4H-l,2,4- triazole-3-carboxamide
Figure imgf000133_0001
A mixture of (S)-3-amino-5-methyl-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)-one, hydrochloride (4.00 g, 16.97 mmol), 5-benzyl-4H-l,2,4-triazole-3-carboxylic acid, hydrochloride (4.97 g, 18.66 mmol) and DIEA (10.37 mL, 59.4 mmol) in isopropanol (150 mL) was stirred vigorously for 10 minutes and then 2,4,6-tripropyl-l,3,5,2,4,6- trioxatriphosphinane 2,4,6-trioxide (T3P) (50% by wt. in EtOAc) (15.15 mL, 25.5 mmol) was added. The mixture was stirred at rt for 10 minutes and then quenched with water and concentrated to remove isopropanol. The resulting crude material is dissolved in EtOAc and washed with 1M HC1, satd. NaHC03 and brine. Organics were concentrated and purified by column chromatography (220 g silica column; 20-90% EtOAc/hexanes, 15 min.; 90%, 15 min.) to give the title compound as a light orange foam (5.37 g, 83%). 1H NMR (MeOH-d4) δ: 7.40 - 7.45 (m, 1H), 7.21 - 7.35 (m, 8H), 5.01 (dd, J = 11.6, 7.6 Hz, 1H), 4.60 (dd, J = 9.9, 7.6 Hz, 1H), 4.41 (dd, J = 11.4, 9.9 Hz, 1H), 4.17 (s, 2H), 3.41 (s, 3H); MS (m/z) 378.3 (M+H+). Alternative Preparation:
To a solution of (S)-3-amino-5-methyl-2,3-dihydrobenzo[b][l,4]oxazepin-4(5H)- one hydrochloride (100 g, 437 mmol), 5-benzyl-4H-l,2,4-triazole-3-carboxylic acid hydrochloride (110 g, 459 mmol) in DCM (2.5 L) was added DIPEA (0.267 L, 1531 mmol) at 15 °C. The reaction mixture was stirred for 10 min. and 2,4,6-tripropyl-l, 3, 5,2,4,6- trioxatriphosphinane 2,4,6-trioxide >50 wt. % in ethyl acetate (0.390 L, 656 mmol) was slowly added at 15 °C. After stirring for 60 mins at RT the LCMS showed the reaction was complete, upon which time it was quenched with water, partitioned between DCM and washed with 0.5N HCl aq (2 L), saturated aqueous NaHC03 (2 L), brine (2 L) and water (2 L). The organic phase was separated and activated charcoal (100 g) and sodium sulfate
(200 g) were added. The dark solution was shaken for 1 h before filtering. The filtrate was then concentrated under reduced pressure to afford the product as a tan foam (120 g). The product was dried under a high vacuum at 50 °C for 16 h. 1H MR showed 4-5% wt of ethyl acetate present. The sample was dissolved in EtOH (650 ml) and stirred for 30 mins, after which the solvent was removed using a rotavapor (water-bath T=45 °C). The product was dried under high vacuum for 16 h at RT (118 g, 72% yield). The product was further dried under high vacuum at 50 °C for 5 h. 1H NMR showed <1% of EtOH and no ethyl acetate. 1H NMR (400 MHz, DMSO-i¾) δ ppm 4.12 (s, 2 H), 4.31 - 4.51 (m, 1 H), 4.60 (t, J=10.36 Hz, 1 H), 4.83 (dt, 7=11.31, 7.86 Hz, 1 H), 7.12 - 7.42 (m, 8 H), 7.42 - 7.65 (m, 1 H), 8.45 (br. s., 1 H), 14.41 (br. s., 1 H). MS (m/z) 378 (M + H+).
Crystallization:
(S)-5-Benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)- 4H-l,2,4-triazole-3-carboxamide (100 mg) was dissolved in 0.9 mL of toluene and 0.1 mL of methylcyclohexane at 60 °C, then stirred briskly at room temperature (20 °C) for 4 days. After 4 days, an off-white solid was recovered (76 mg, 76% recovery). The powder X-ray diffraction (PXRD) pattern of this material is shown in Figure 7 and the corresponding diffraction data is provided in Table 1.
The PXRD analysis was conducted using a PANanalytical X'Pert Pro
diffractometer equipped with a copper anode X-ray tube, programmable slits, and
X'Celerator detector fitted with a nickel filter. Generator tension and current were set to 45kV and 40mA respectively to generate the copper Ka radiation powder diffraction pattern over the range of 2 - 40°2Θ. The test specimen was lightly triturated using an agate mortar and pestle and the resulting fine powder was mounted onto a silicon background plate.
Table 1.
Figure imgf000135_0001
Example 13
Method I
(S)-5-benzyl-N-(2-oxo-7-(lH-pyrazol-3-yl)-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)- - 1 ,2,4-triazole-3 -carboxamide
Figure imgf000136_0001
(S)-5-benzyl-N-(7-bromo-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H- l,2,4-triazole-3 -carboxamide (60 mg, 0.136 mmol), tert-butyl 3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole-l-carboxylate (40.1 mg, 0.136 mmol), Pd(PPh3)4 (29.8 mg, 0.026 mmol) and K2C03 (107 mg, 0.775 mmol) were mixed inl,4-dioxane (2 mL) and water (1 mL). The reaction mixture was put in an Emrys Optimizer (150W, absorption normal) and microwaved at 130 °C, for 20 min. The reaction mixture was filtered and the filtrate was concentrated. The residue was purified by reverse phase HPLC (Waters Sunfire 30x150mm, 26-60% CH3CN:H20 (0.1 % TFA), 50 mg/mL). Collected fractions containing the product were combined, neutralized by NaHC03, and then concentrated to give the desired product as a white solid (6 mg, 11 % yield). 1H MR (DMSO-d6) δ ppm 10.05 (s, 1H), 8.31 (br. s., 1H), 7.77 (s, 1H), 7.65 - 7.75 (m, 2H), 7.19 - 7.42 (m, 5H), 7.07 (d, J = 8.0 Hz, 1H), 6.71 (d, J = 1.5 Hz, 1H), 4.38 (dt, J = 11.2, 7.9 Hz, 1H), 4.12 (s, 2H), 2.66 - 2.90 (m, 2H), 2.42 - 2.51 (m, 1H), 2.28 (br. s., 1H); MS (m/z): 428 (M+H+). Example 14
(S)-3-(5-benzylisoxazole-3-carboxamido)-5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine-7-carboxylic acid
Figure imgf000136_0002
Example 84
To a solution of (S)-methyl 3-(5-benzylisoxazole-3-carboxamido)-5-methyl-4-oxo- 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine-7-carboxylate (332 mg, 0.762 mmol) in THF (6 mL)/Water (2.0 mL) was added LiOH (1.144 mL, 1.144 mmol) as a solution in water. Reaction was stirred at rt for about 2h. The reaction mixture was diluted with water then extracted with EtOAc twice. The aqueous phase was acidified to pH~3.0 then it was extracted with EtOAc. The latter organic phase was dried over Na2S04 then filtered and concentrated in vacuo to yield the desired product as a solid. The solid was warmed in toluene then decanted to give the final solid product that was used directly in the next step. 1H MR (DMSO-d6) 5: 13.18 (br. s., 1H), 8.87 (d, J = 8.1 Hz, 1H), 7.98 (d, J = 2.0 Hz, 1H), 7.85 (dd, J = 8.3, 2.0 Hz, 1H), 7.25 - 7.38 (m, 6H), 6.55 (s, 1H), 4.87 (dt, J = 11.8, 7.7 Hz, 1H), 4.64 (dd, J = 11.6, 10.1 Hz, 1H), 4.46 (dd, J = 9.9, 7.6 Hz, 1H), 4.22 (s, 2H). MS (m/z) 422.3 (M+H+). Example 15
Method J
(S)-N-(7-acetamido-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-5- -3 -carb oxami de
Figure imgf000137_0001
(S)-N-(7-acetamido-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)- 5-benzylisoxazole-3-carboxamide. To a solution of (S)-N-(7-amino-5-methyl-4-oxo- 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-5-benzylisoxazole-3-carboxamide (60.0 mg, 0.153 mmol) in THF (2.0 mL) at 0 °C was added DIEA (0.061 mL, 0.352 mmol) then AcCl (10.87 μΐ., 0.153 mmol). The reaction mixture was monitored by LCMS. Reaction showed desired mass after 10 min with all sm consumed. The reaction mixture was concentrated to a solid residue. The solid was suspended in small amount of DCM and lmL of 25%
EtO Ac/Hex. The suspension was lightly warmed then cooled and filtered to collect the solid product. The solid was washed with ethyl ether .yield=56 mg solid powder; NMR shows much impurity, so the sample was subjected to FCC [MeOH-DCM:0-3.0 %].
yield=18 mg. 1H NMR (DMSO-d6) δ: 10.11 (s, 1H), 8.86 (d, J = 8.1 Hz, 1H), 7.71 (d, J = 2.3 Hz, 1H), 7.25 - 7.42 (m, 6H), 7.15 (d, J = 8.6 Hz, 1H), 6.55 (s, 1H), 4.84 (dt, J = 11.6, 8.0 Hz, 1H), 4.49 - 4.56 (m, 1H), 4.35 (dd, J = 9.9, 7.8 Hz, 1H), 4.22 (s, 2H), 3.27 (s, 3H), 2.06 (s, 3H). MS (m/z) 435.3 (M+H+). Example 16
Method K
(S)-(3-(5-benzyl-4H-l,2,4-triazole-3-carboxamido)-2-oxo-2,3,4,5-tetrahydi
-7-yl)boronic acid
Figure imgf000138_0001
(S)-5-benzyl-N-(2-oxo-7-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3,4,5- tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3-carboxamide (65 mg, 0.133 mmol) was dissolved in 2mL THF, and then sodium periodate (28.5 mg, 0.133 mmol) was added, followed by HCl (IN in H20, 0.041 mL, 1.334 mmol). The mixture was maintained at rt for 2 h. The mixture was then concentrated and the residue was purified by Isco Combiflash (2%-10% MeOH/CH2Cl2, 10% NEt3 in MeOH; 40g RediSep column). Collected fractions containing the product were combined and concentrated to give the desired product as a colorless oil, which was then lyophilized to a white solid (36 mg, 67 % yield). 1H NMR (400 MHz, MeOD-d4) δ ppm 2.11 - 2.32 (m, 1 H) 2.57 - 2.74 (m, 1 H) 2.74 - 2.89 (m, 1 H) 2.97 (td, J=13.33, 7.96 Hz, 1 H) 4.12 - 4.22 (m, 2 H) 4.49 - 4.65 (m, 1 H) 6.98 - 7.17 (m, 1 H) 7.19 - 7.43 (m, 5 H) 7.51 - 7.66 (m, 1 H) 7.71 (br. s., 1 H); MS (m/z): 406 (M+H+).
Example 17
(S)-(3-(3-benzyl-lH-pyrazole-5-carboxamido)-2-oxo-2,3,4,5-tetrahydro-lH- benzo[b]azepin-8-yl)boronic acid
Figure imgf000139_0001
KOAc, bis(pinacolato)diboron PdCUdppf
Figure imgf000139_0002
To a mixture of (S)-3-amino-8-bromo-4,5-dihydro-lH-benzo[b]azepin-2(3H)-one (1.0 g, 3.92 mmol) in DCM (30 mL) was added TEA (0.820 mL, 5.88 mmol) and BOC20 (0.956 mL, 4.12 mmol). Mixture was stirred at room temperature for 1.5 hours and a solid precipitated out. Water was added and the mixture was stirred for 5 minutes and solid was filtered and dried to give 869 mg of (S)-tert-butyl (8-bromo-2-oxo-2,3,4,5-tetrahydro-lH- benzo[b]azepin-3-yl)carbamate as a pale yellow solid. Layers of the filtrate were separated and organics were concentrated to a solid. Solid was triturated in diethyl ether, filtered and dried to give 390 mg pale yellow solid. Yield = 87%. 1H NMR (DMSO-d6) δ: 9.81 (s, 1H), 7.29 - 7.33 (m, 1H), 7.24 (d, J = 8.1 Hz, 1H), 7.18 (d, J = 1.8 Hz, 1H), 7.05 (d, J = 8.3
Hz, 1H), 3.86 (dt, J = 12.0, 8.1 Hz, 1H), 2.56 - 2.72 (m, 2H), 2.19 (m, 1H), 2.06 (td, J =
12.3, 7.3 Hz, 1H), 1.34 (s, 9H); MS (m/z) 355/357 (bromine splitting pattern) (M+H+). Nitrogen was bubbled through a mixture of (S)-tert-butyl (8-bromo-2-oxo-2,3,4,5- tetrahydro-lH-benzo[b]azepin-3-yl)carbamate (385 mg, 1.084 mmol), potassium acetate (532 mg, 5.42 mmol) and bis(pinacolato)diboron (330 mg, 1.301 mmol) in 1,4-Dioxane (10 mL) for 5 min. Then PdCl2(dppf)-CH2Cl2 adduct (89 mg, 0.108 mmol) was added and mixture was heated at 95°C for 2 hours. Reaction was cooled to room temperature, diluted with water and ethyl acetate and filtered through a Celite plug. Layers of filtrate were separated. Organics were concentrated and purified by Biotage (10 g silica column, 10- 50% E/H, 10 min.; 70%, 5 min.) to give 344 mg of (S)-tert-butyl (2-oxo-8-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate as a tan solid in 77% yield. 1H NMR (DMSO-d6) δ: 9.70 (s, 1H), 7.41 (d, J = 7.3 Hz, 1H), 7.28 - 7.33 (m, 2H), 7.00 (d, J = 8.3 Hz, 1H), 3.83 (dt, J = 11.9, 8.3 Hz, 1H), 2.68 (m, 2H), 2.18 (m, 1H), 2.08 (m, 1H), 1.34 (s, 9H), 1.30 (s, 12H); MS (m/z) 403.4 (M+H+).
To a mixture of (S)-tert-butyl (2-oxo-8-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)carbamate (70 mg, 0.174 mmol) in DCM (3 mL) was added 4.0 M HC1 in dioxane (218 μΐ, 0.870 mmol). Mixture was stirred at room temperature for 2 days, concentrated to remove solvents to yield (S)-3-benzyl-N-(2-oxo-8- (4,4,5, 5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)- lH-pyrazole-5-carboxamide which was used as-is in the next step. MS (m/z) 303.3 (M+H+).
A solution of 3-benzyl-lH-pyrazole-5-carboxylic acid (38.7 mg, 0.191 mmol) and
HATU (79 mg, 0.209 mmol) in CH3CN (1 mL) and DMSO (0.3 mL) was stirred for 40 minutes. Then it was added to a mixture of N-methylmorpholine (0.067 mL, 0.609 mmol) and (S)-3-amino-8-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-4,5-dihydro-lH- benzo[b]azepin-2(3H)-one (from the prior step) in CH3CN (1 mL). The mixture was stirred at room temperature for 40 minutes. Water (5 mL) was slowly added while stirring vigorously. A solid precipitated out and it was stirred for 5 minutes, filtered and dried to give 60 mg light brown solid in 70% yield. 1H NMR (MeOH-d4) δ: 7.58 (d, J = 7.3 Hz, 1H), 7.46 (s, 1H), 7.27 - 7.37 (m, 4H), 7.19 - 7.26 (m, 4H), 6.49 (br. s., 1H), 4.54 (dd, J = 11.6, 8.1 Hz, 1H), 4.02 (s, 2H), 2.98 (td, J = 13.2, 8.0 Hz, 1H), 2.75 - 2.83 (m, 1H), 2.61 (m, 1H), 2.16 - 2.28 (m, 1H), 1.36 (s, 12H); MS (m/z) 487.5 (M+H+).
To a solution of (S)-3-benzyl-N-(2-oxo-8-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-
2-yl)-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-lH-pyrazole-5-carboxamide (45 mg,
0.093 mmol) in THF (2 mL) was added benzeneboronic acid, polymer bound [5 eq, 0.46 mmol, 170 mg (assuming 2.6 mmol/g loading), 2.6-3.2 mmol/g loading] and cone. HC1 (0.039 mL, 0.463 mmol). Mixture was stirred at room temperature for 3 days. Reaction was not quite complete, so more benzeneboronic acid, polymer bound (50 mg) was added and mixture was stirred for another 4 hours, then filtered to remove resin and concentrated. Water (3 mL) was added and a solid formed. Solid was filtered and purified by column chromatography (4 g silica column; 50-100% ethyl acetate/hexanes, then 10%
methanol/ethyl acetate) to give 9 mg of (S)-(3-(3-benzyl-lH-pyrazole-5-carboxamido)-2- oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-8-yl)boronic acid as an off-white solid in 33% yield. 1H MR (MeOH-d4) δ: 7.46 (d, J = 7.3 Hz, 1H), 7.28 - 7.36 (m, 4H), 7.21 - 7.26 (m, 3H), 6.48 (br. s., 1H), 4.56 (dd, J = 11.2, 8.0 Hz, 1H), 4.03 (s, 2H), 2.92 - 3.03 (m, 1H), 2.73 - 2.81 (m, 1H), 2.63 (br. s., 1H), 2.20 (m, 1H); MS (m/z) 405.4 (M+H+).
The following compounds were prepared via coupling of the appropriate amine and acid using the method indicated.
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
. ppm s,
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
carboxamide ppm (s, 3 H)
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
diastereomers) 0.99- 1.17 (m, IH)
Figure imgf000158_0001
Figure imgf000159_0001
carboxamide 6^ 3.98 (s, 2H), 3.31 (s, 3H)
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
Figure imgf000166_0001
(S)-2-(2,5- ¾ NMR (DMSO-d6) δ:
difluorobenzyl)-N-(4- 10.17 (s, IH), 9.15 (d, J = 8.0
oxo-2,3,4,5- Hz, IH), 7.43 - 7.51 (m, IH),
144 tetrahy drobenzo [b] 7.33 - 7.41 (m, 2H), 7.09 - 401 F
[1,4] oxazepin-3 -y 1)- 7.19 (m, 4H), 6.10 (s, 2H),
2H-tetrazole-5- 4.80 - 4.90 (m, IH), 4.57 (t, J
carboxamide = 10.7 Hz, IH), 4.45 (dd, IH)
¾ NMR (DMSO-d6) δ:
(S)-2-benzyl-N-(4- 10.17 (s, IH), 9.14 (d, J = 8.0
oxo-2,3,4,5- Hz, IH), 7.36 - 7.46 (m, 5H),
tetrahy drobenzo [b]
145 7.10 - 7.18 (m, 4H), 6.05 (s, 365 F
[1,4] oxazepin-3 -y 1)- 2H), 4.86 (dt, J = 10.5, 7.3
2H-tetrazole-5- Hz, IH), 4.57 (t, J = 10.7 Hz,
carboxamide
IH), 4.45 (dd, IH)
(S)-5-benzyl-N-(5- ¾ NMR (DMSO-dg) δ: 9.43
methyl-4-oxo- (br. s., IH), 7.52 (dd, J = 7.7,
2,3,4,5- 1.6 Hz, IH), 7.21 - 7.41 (m,
146 tetrahy drobenzo [b] 8H), 4.80 (br. s., IH), 4.65 379 F
[1,4] oxazepin-3 -y 1)- SNH (dd, J = 11.6, 9.9 Hz, IH),
1 ,3 ,4-oxadiazole-2- 4.35 - 4.45 (m, 3H), 3.31 (s,
carboxamide 3H)
¾ NMR (DMSO-d6) δ:
(S)-5-benzyl-N-(4- 10.17 (s, IH), 9.43 (d, J = 8.0
oxo-2,3,4,5- Hz, IH), 7.28 - 7.42 (m, 5H),
tetrahy drobenzo [b] 7.11 - 7.17 (m, 4H), 4.79 (dt,
147 365 F
[1,4] oxazepin-3 -y 1)- J = 10.5, 7.3 Hz, IH), 4.56 (t,
l,3,4-oxadiazole-2- J = 10.5 Hz, IH), 4.44 (dd, J
carboxamide = 10.5, 6.5 Hz, IH), 4.39 (s,
2H)
¾ NMR (DMSO-d6) δ:
(S)-3-benzyl-N-(4- 13.23 (br. s., IH), 8.22 - 8.31
chloro-5-methyl-6-
HN / (m, IH), 8.20 (s, IH), 8.02
oxo-6,7,8,9- (br. s., IH), 7.29 - 7.34 (m,
tetrahydro-5H-
148 2H), 7.21 - 7.27 (m, 3H), 412 F pyrimido[4,5-b]
6.41 (br. s., IH), 4.80 - 4.92
[l,4]diazepin-7-yl)- (m, IH), 3.99 (br. s., 2H),
lH-pyrazole-5- 3.64 (br. s., IH), 3.39 - 3.50
carboxamide CI (m, IH), 3.17 (s, 3H)
Figure imgf000168_0001
(S)-l-benzyl-N-(5- ¾ NMR (DMSO-d6) δ: 9.10 methyl-6-oxo- (br. s., 1H), 8.87 (s, 1H), 8.52
6,7,8,9-tetrahydro- (s, 1H), 8.42 - 8.49 (m, 2H),
154 5H-pyrimido[4,5-b] 7.30 - 7.43 (m, 5H), 5.51 (s, 379 F [l,4]diazepin-7-yl)- 2H), 4.82 - 4.89 (m, 1H),
lH-l,2,4-triazole-3- 3.58 - 3.71 (m, 2H), 3.34 (s,
carboxamide 3H)
Figure imgf000169_0001
(S)-5-benzyl-N-(4-
¾ NMR (DMSO-d6) δ:
oxo-2,3,4,5- 10.17 (s, 1H), 9.07 (d, J = 8.0
tetrahy drobenzo [b]
155 Hz, 1H), 7.08 - 7.46 (m, 9H), 365 F
[1,4] oxazepin-3 -y 1)- 4.76 - 4.89 (m, 1H), 4.37 - l,2,4-oxadiazole-3- 4.61 (m, 4H)
carboxamide
¾ NMR (400 MHz, CDC13)
(S)-5- 5 ppm 4.34 (t, 1 H) 4.79 (dd, (difluoro(phenyl)
.7=10.36, 6.57 Hz, 1 H) 5.09
methyl)-N-(4-oxo- (dt, J= 10.55, 6.60 Hz, 1 H)
2,3,4,5-
156 6.87 (s, 1 H) 7.05 - 7.10 (m, 400 A tetrahy drobenzo [b]
1 H) 7.13 - 7.23 (m, 3 H)
[ 1,4] oxazepin-3 - 7.45 - 7.68 (m, 5 H) 7.83 (d,
yl)isoxazole-3- .7=6.82 Hz, 1 H) 8.26 (s, 1 H)
carboxamide
¾ NMR (400 MHz, CDC13)
(S)-5- 5 ppm 3.46 (s, 3 H) 4.28 (dd, (difluoro(phenyl)met
.7=11.12, 9.85 Hz, 1 H) 4.76
hyl)-N-(5-methyl-4-
(dd, .7=9.85, 7.33 Hz, 1 H)
oxo-2,3,4,5-
157 5.04 (dt, .7=11.12, 7.07 Hz, 1 414 A tetrahy drobenzo [b]
H) 6.83 (s, 1 H) 7.18 - 7.32
[ 1,4] oxazepin-3 - (m, 5 H) 7.45 - 7.56 (m, 3 H)
yl)isoxazole-3- 7.56 - 7.64 (m, 2 H) 7.83 (d,
carboxamide
.7=6.82 Hz, 1 H)
1H NMR (400 MHz, CDC13)
5 ppm 3.34 (s, 3 H) 3.91 (s, 2
(S)-5-(3- H) 4.21 (dd, J=l 1.37, 9.85
bromobenzyl)-N-(5- Hz, 1 H) 4.59 (dd, J=9.85,
methyl-4-oxo- 7.58 Hz, 1 H) 5.06 (dt,
2,3,4,5-
158 J=l 1.49, 7.64 Hz, 1 H) 6.43 455 A tetrahy drobenzo [b]
(s, 1 H) 7.00 - 7.21 (m, 6 H)
[1,4] oxazepin-3 -y 1)- 7.21 - 7.32 (m, 2 H) 8.07 (d,
lH-pyrazole-3- J=7.58 Hz, 1 H) 12.45 (br. s.,
carboxamide c5 ° 1 H)
Figure imgf000170_0001
Example 161
(S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4- -3-carboxamide
Figure imgf000170_0002
Preparation 1 : The title compound was prepared via coupling of the appropriate amine and acid using Method H.
Preparation 2: To a solution of (S)-3-amino-4,5-dihydro-lH-benzo[b]azepin-2(3H)- one (50 g, 284 mmol), 5-benzyl-4H-l,2,4-triazole-3-carboxylic acid (72.1 g, 355 mmol) in dichloromethane (1500 ml) was added DIPEA (173 ml, 993 mmol) at 15 °C. The reaction mixture was stirred for 20 minutes and 2,4,6-tripropyl-l,3,5,2,4,6-trioxatriphosphinane
2,4,6-trioxide (236 ml, 397 mmol) was slowly added at 15 °C. The reaction was stirred for over night. The resulting solid was filtered and the solid was washed with DCM. The solid was dried under vacuum at 50 °C for over night. For the filtration, it was concentrated under rotary-evaporator and to the sticky residue added plenty of cold water and stirring, the slowly precipitated white solid was collected and washed the solid with water and ethyl ether. The solid was dried under vacuum at 50 °C for 3 days to afford the product (S)-5- benzyl -N-(2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3- carboxamide (Total recovery: 102 g, 282 mmol, 99 % yield). 1H NMR (MeOH-d4) δ: 7.18 - 7.48 (m, 8H), 7.10 (d, J = 7.6 Hz, 1H), 4.58 (m, 1H), 4.17 (s, 2H), 2.97 (m 1H), 2.77 (m, 1H), 2.67 (m, 1H), 2.23 (m, 1H). MS (m/z) 362 (M+H+).
To a solution of (S)-5-benzyl-N-(2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3- yl)-4H-l,2,4-triazole-3-carboxamide (35 g, 97 mmol) in DMA (700 ml) was added NCS (14.87 g, 111 mmol) at 0 °C. After 30 mins, the reaction mixture was warmed up to RT and continued stirring for 5 hrs. A second portion of NCS (3.88 g, 29.1 mmol) was added to the reaction mixture and stirring continued for an additional 24 hrs. A third portion of NCS (1.293 g, 9.68 mmol) was then added and the solution was stirred at RT for 16 h further.
The reaction was then quenched with cold water. The white solid was collected by filtration and washed with water 3 times to provide (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5- tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3-carboxamide (36 g, 91 mmol, 94 % yield). The product was air dried overnight. Additional purification was achieved by suspending (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5-tetrahydro-lH-benzo[b]azepin-3-yl)- 4H-l,2,4-triazole-3-carboxamide (10 g, 25.3 mmol) in hot methanol (500 mL) for lh. The solution was then cooled to RT, filtered and the solid was washed with methanol 2 times (75 mL) to give the product (7 g, 70% yield). 1H NMR (DMSO-d6) δ: 10.06 (s, 1H), 8.31 (br. s., 1H), 7.44 (d, J=2.5 Hz, 1H), 7.18-7.40 (m, 7H), 7.05 (d, J=8.6 Hz, 1H), 4.32 (dt, J=11.5, 7.9 Hz, 1H), 4.11 (s, 2H), 2.63-2.80 (m, 2H), 2.37-2.49 (m, 1H), 2.25 (br. s., 1H). MS (m/z) 396/398 (M+H+).
The following compounds were prepared via coupling of the appropriate amine and acid using the method indicated.
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
carboxamide 3H), 3.32 (s, 3H)
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
carboxamide c5 ° 3H)
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
The following compounds were prepared via the oxidation method indicated.
Figure imgf000188_0002
The following compounds were prepared via acylation or isocyante addition of the appropriate amine using the method indicated.
Figure imgf000188_0003
Figure imgf000189_0001
Figure imgf000190_0001
The following compounds were prepared via Suzuki coupling using the method indicated.
Figure imgf000190_0002
The following compounds were prepared via coupling of the appropriate amine and acid using the method indicated.
Figure imgf000190_0003
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
carboxamide 3H), 2.44 (s, 3H)
Figure imgf000197_0001
carboxamide 1.10-1.29 (m, 2H)
Example 286
Figure imgf000198_0001
Using the procedure described in Example 12, (S)-3-amino-5-methyl-2,3- dihydrobenzo[b][l,4]oxazepin-4(5H)-one hydrochloride (220 mg, 0.96 mmol) was reacted with 5-(l-phenylethyl)-4H-l,2,4-triazole-3-carboxylic acid hydrochloride (256 mg, 1.0 mmol) to yield N-((S)-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-5-(l- phenylethyl)-4H-l,2,4-triazole-3-carboxamide (330 mg, 89% yield) as a mixture of 2 diastereoisomers. Separation of the 2 diastereoisomers was achieved using a Gilson LC eluting with 20:80 EtOAc/Hexane with 0.1% DEA. The 2 diastereoisomers were each isolated with a diastereomeric excess > 99% and a yield of 138 mg of each.
N-((S)-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-5-((S)-l- phenylethyl)-4H-l,2,4-triazole-3-carboxamide: 1H MR (DMSO-d6) δ: 8.46 (br. s., 1H), 7.51 (dd, J = 7.7, 1.6 Hz, 1H), 7.08 - 7.45 (m, 8H), 4.84 (dd, J = 11.2, 8.0 Hz, 1H), 4.52 - 4.72 (m, 1H), 4.25 - 4.49 (m, 2H), 3.32 (s, 3H), 1.63 (d, J = 7.3 Hz, 3H). MS (m/z) 392 (M+H+).
N-((S)-5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-yl)-5-((R)-l- phenylethyl)-4H-l,2,4-triazole-3-carboxamide: 1H NMR (DMSO-d6) δ: 8.45 (br. s., 1H), 7.51 (dd, J = 7.7, 1.9 Hz, 1H), 7.12 - 7.42 (m, 9H), 4.76 - 4.94 (m, 1H), 4.53 - 4.70 (m, 1H), 4.28 - 4.49 (m, 2H), 3.26 - 3.42 (m, 3H), 1.63 (d, J = 7.3 Hz, 3H). MS (m/z) 392 (M+H+).
Pharmaceutical Compositions
Example A
Tablets are prepared using conventional methods and are formulated as follows:
Ingredient Amount per tablet
Compound 5mg
Microcrystalline cellulose lOOmg
Lactose lOOmg Sodium starch glycollate 30mg
Magnesium stearate 2mg
Total 237mg
Example B
Capsules are prepared using conventional methods and are formulated as follows: Ingredient Amount per tablet
Compound 15mg
Dried starch 178mg
Magnesium stearate 2mg
Total 195mg
Biological Assays:
Biological in vitro assay
A fluorescent polarization based binding assay was developed to quantitate interaction of novel test compounds at the ATP binding pocket of RIP 1, by competition with a fluorescently labeled ATP competitive ligand. GST-RipKl (1-375) was purified from a Baculovirus expression system and was used at a final assay concentration of lOnM . A fluorescent labeled ligand (14-(2-{[3-({2-{[4-(cyanomethyl)phenyl]amino}-6-[(5- cyclopropyl-lH-pyrazol-3-yl)amino]-4-pyrimidinyl}amino)propyl]amino}-2-oxoethyl)- 16,16, 18,18-tetramethyl-6,7,7a, 8a,9, 10,16, 18- octahydrobenzo[2",3"]indolizino[8",7":5',6']pyrano[3',2':3,4]pyrido[l,2-a]indol-5-ium-2- sulfonate (prepared as described below) was used at a final assay concentration of 5nM. Both the enzyme and ligand were prepared in solutions in 50mM HEPES pH7.5, lOmM NaCl, 50mM MgC12, 0.5mM DTT, and 0.02% CHAPS. Test compounds were prepared in neat DMSO and lOOnL was dispensed to individual wells of a multiwell plate. Next, 5ul GST-RipKl (1-375) was added to the test compounds at twice the final assay
concentration, and incubated at room temperature for 10 minutes. Following the incubation, 5ul of the fluorescent labeled ligand solution, was added to each reaction, at twice the final assay concentration, and incubated at room temperature for at least 15 minutes. Finally, samples were read on an instrument capable of measuring fluorescent polarization. Test compound inhibition was expressed as percent (%) inhibition of internal assay controls. For concentration response experiments, normalized data were fit and pIC5os determined using conventional techniques. The pICsos are averaged to determine a mean value, for a minimum of 2 experiments.
As determined using the above method, the compounds of Examples 1-286 exhibited a pIC5o between approximately 5.0 and 9.0. For instance, the compounds of Examples 12, 91, 102, 161, 163 and 169 inhibited RIPl kinase in the above method with a mean pIC50 of approximately 7.6, 7.6, 7.8, 7.9, 7.9 and 7.2 respectively. Continued testing resulted in a slight change in the reported average pIC50 for these compounds (Example 161 (7.7) and Example 169 (7.3)).
Preparation of (14-(2-{[3-({2-{[4-(cyanomethyl)phenyl]amino}-6-[(5-cyclopropyl- lH-pyrazol-3-yl)amino]-4-pyrimidinyl}amino)propyl]amino}-2-oxoethyl)-16, 16,18, 18- tetramethyl-6,7,7a,8a,9, 10, 16,18-octahydrobenzo[2",3"]indolizino[8",7":5',6']
pyrano[3',2':3,4] pyrido[l,2-a]indol-5-ium-2-sulfonate
Figure imgf000200_0001
A solution of 2,4,6-trichloropyrimidine (Alfa, 12.25 g, 66.8 mmol), 3-amino-5- cyclopropyl-lH-pyrazole (Fluorochem 8.23 g, 66.8 mmol) and triethylamine (11.2 mL, 80.4 mmol) in ethanol (100 mL) was stirred at room temperature for 16 hours under Ar (balloon). The solvent was removed in vacuo and the crude material was dissolved in ethyl acetate. The solution was washed with water and dried (Na2S04), filtered and evaporated to give a beige solid. Pure product was obtained as a white crystalline solid after recrystallisation from acetonitrile. It was possible to obtain a second crop. From two runs performed under the same conditions, 29.0 g (88%) of 2,6-dichloro-N-(5-cyclopropyl-lH- pyrazol-3-yl)pyrimidin-4-amine. The product contained -10 % acetonitrile but was carried through to the next step regardless.
A suspension of (5-cyclopropyl-lH-pyrazol-3-yl)-(2,6-dichloro-pyrimidin-4-yl)- amine (25.8 g, 0.1 mol) and 4-aminophenylacetonitrile (Alfa, 13.91 g, 0.11 mol) in diisopropylethylamine (Alfa, 342 mL) was stirred at 110 °C for 16 hours under Ar
(balloon). The resultant gummy suspension was dissolved in DCM, washed with water and dried (Na2S04), filtered and concentrated. When the DCM was reduced to a small volume the material was left to stand and the product dropped out of solution. After filtration and washing with DCM, formation of 2-(4-((4-chloro-6-((5-cyclopropyl-lH-pyrazol-3- yl)amino)pyrimidin-2-yl)amino)phenyl)acetonitrile was obtained as a beige powder (10.6 g, 30.3 %).
A mixture of {4-[4-chloro-6-(5-cyclopropyl-lH-pyrazol-3-ylamino)-pyrimidin-2- ylamino]-phenyl}-acetonitrile (1.54 g, 4.2 mmol) and tert-butyl N-(2- aminopropyl)carbamate (Aldrich, 2.20 g, 12.6 mmol, 3.0 eq) were heated at 115°C for 16 hours under Ar (balloon). The resultant glassy solid was purified by column
chromatography (at least 25 cm depth of silica, eluent = DCM→ 5% MeOH in DCM). Recovered starting material is the first yellow band to elute from the column (eluent ~ 2% MeOH in DCM) and the product elutes when the second yellow band has moved through the column (eluent ~ 4% MeOH in DCM). A purple band elutes once almost all the product has eluted. A good eluent for TLC analysis of the fractions is 1 : 1 EtO Ac/Pet. Ether. The initial fractions of the product are contaminated with a trace of higher Rf material whereas the final fractions of product contain traces of a lower Rf material. Therefore only the middle product fractions were combined. tert-Butyl (3-((2-((4-
(cyanomethyl)phenyl)amino)-6-((5-cyclopropyl-lH-pyrazol-3-yl)amino)pyrimidin-4- yl)amino)propyl)carbamate was obtained as a yellow foam (0.7 g, 33.0 %).
tert-Butyl (3-((2-((4-(cyanomethyl)phenyl)amino)-6-((5-cyclopropyl-lH-pyrazol-3- yl)amino)pyrimidin-4-yl)amino)propyl)carbamate (20 mg, 0.040 mmol) was dissolved in an ice-cold solution of water (0.1 mL) in trifluoroacetic acid (TFA) (1.9 mL). The reaction mixture was allowed to warm to room temperature and left for a total of 2 hours. Excess acid was removed under reduced pressure and the oily residue triturated with several portions of dry ether. The residual solid was dried under reduced pressure. MS (m/z) 403 (M+H+). Analytical CI 8 HPLC showed only one major component. Yield of 2-(4-((4-((3- aminopropyl)amino)-6-((5-cyclopropyl-lH-pyrazol-3-yl)amino)pyrimidin-2- yl)amino)phenyl)acetonitrile estimated at approximately 98%.
2-(4-((4-((3-Aminopropyl)amino)-6-((5-cyclopropyl-lH-pyrazol-3- yl)amino)pyrimidin-2-yl)amino)phenyl)acetonitrile trifluoroacetic acid salt (3.2 mg, 6.18 μπιοΐ) and 14-{2-[(2,5-dioxo-l-pyrrolidinyl)oxy]-2-oxoethyl}-16, 16,18, 18-tetramethyl- 6,7,7a,8a,9, 10,16, 18- octahydrobenzo[2",3"]indolizino[8",7":5',6']pyrano[3',2':3,4]pyrido[l,2-a]indol-5-ium-2- sulfonate trifluoroacetic acid salt (2.6 mg, 3.37 μιηοΐ) were placed in a 2ml Eppendorf tube and DMF (200 μΐ) added. The mixture was stirred till all solid had dissolved and then the mixture was basified by the addition of DIPEA (2 μΐ, 0.011 mmol). The reaction was stirred overnight at room temperature. The reaction mixture was evaporated to dryness and redissolved in DMSO/MeOH (<lml), filtered (0.2μπι) and applied to a Phenomenex Jupiter C18 preparative column and eluted with the following gradient (A = 0.1% trifluoroacetic acid in water, B= 0.1% TFA/90%acetonitrile/10% water): Flow rate = lOml/min., AU = 20/10 (214nm). The target component was eluted in two fractions. Both fractions were combined and evaporated to dryness to yield 1.4 mg of 14-(2-{[3-({2-{[4- (cyanomethyl)phenyl]amino}-6-[(5-cyclopropyl-lH-pyrazol-3-yl)amino]-4- pyrimidinyl}amino)propyl]amino}-2-oxoethyl)-16, 16,18, 18-tetramethyl- 6,7,7a,8a,9, 10,16, 18- octahydrobenzo[2",3"]indolizino[8",7":5',6']pyrano[3',2':3,4]pyrido[l,2-a]indol-5-ium-2- sulfonate.
GST-RipKl Preparation: His.GST.TEV.RIPKl 1-375
The RIPK1 gene [receptor (T FRSF)-interacting serine-threonine kinase 1] was cloned from human adrenal gland cDNA. Primers were designed from the reference sequence NM 003804.3 with an added CACC Kozak directional tag for cloning into pENTR/TEV/D-TOPO. Gateway® LR cloning was used to site-specifically recombine RIPK1 downstream to an N-terminal HisGST contained within the destination vector pDEST8-His.GST according to the protocol described by Invitrogen. A stop codon was inserted after amino acid 375 using Quikchange Stratagene mutagenesis kit according to manufacturer's protocol and resulted in pDEST8.His.GST.TEV.human RIPK1 1-375. His. GST. Tev. human RIPKl 1-375 baculovirus was generated using the bac to bac system (Invitrogen) according to manufacturer's specifications. Transfection of Spodoptera frugiperda(Sf9) insect cells was performed using Fugene 6 (Roche), according to the manufacturer's protocol. His.GST.TEV.human RJPK1 1-375 baculovirus infected insect cells (BIICs) were prepared during the baculovirus generation according to David Wasilko and S Edward Lee, TIPS: Titerless Infected Cells Preservation and Scale up, BioProcessing Journal Fall 2006 p29-32. 20L Sf9 cells were grown in serum free Hyclone, SFX media (HyClone Laboratories, 925 West 1800 South Logan, Utah 84321) at 27°C in wave bags seeded at a density of.8X10A6cell/ml with a rock rate of 25rpm, airflow of .18 to .22. in wave reactor (WAVE Bioreactor, System 20/50EH). Cells were grown ON at 27C.
His.GST.TEV.human RIPK1 1-375 baculovirus infected insect cells (BIICs) were used to infect Sf9s at a cell density of 1.7 to 2.4 X10A6. 2ml of BIIC (1 X10A7cells/mL) were added to 20L cells. Rock rate is increased to 25rom at infection. Harvest 72 hrs post infection using the Viafuge. Weigh pellets, seal wave bags and freeze at -80.
A 50g cell pellet was re-suspended in 250ml lysis buffer (50mM Tris pH 7.5, 250mM NaCl, ImM DTT and Complete Protease Inhibitor tablets (l/50ml, from Roche Diagnostics). The cells were lysed by sonication on ice, 3x30" at power level 4 using the large probe on a Branson Sonicator. The suspension was then clarified by centrifugation at 15,000g for 30 minutes, at 4°C. The lysate was decanted from the insoluble pellet and batch bound to 10ml of Glutathione Agarose (Pierce) for 2h at 4C with gentle end over end rotation. The beads were then packed into a column and washed to baseline with lysis buffer (no protease inhibitors) and then eluted with 20mM reduced glutathione in 50mM Tris, pH8.
Fractions identified by SDS-PAGE as containing protein of interest were pooled (10ml total volume), concentrated to about 5ml and loaded onto a 300ml SDX200 SEC column (GE Healthcare) which had been equilibrated in 50mM Tris, pH7.5, 150mM NaCl, ImM DTT and 10% Glycerol. The Ripl protein eluted as a dimer off the SEC column.
The protein concentration was determined by Bradford assay using BSA as a standard. The yield was 12.5mg at 0.63mg/ml. The purity was >95% as determined by scanning a Coomassie stained SDS-PAGE gel.
LCMS analysis showed that the major species had lost the N-terminal methionine, was acetylated and had one phosphorylated site. The protein was aliquoted and frozen at - 80°C for use as needed.
Biological in vivo assay The efficacy of RIP 1 inhibitors can be tested in mice in vivo using a T F-driven systemic inflammatory response syndrome model (Duprez, L., et al. 2011. Immunity 35(6):908-918). The model can be run in a long modality (using TNF alone i.v.) which results in the termination of the study in ~7hrs (under IACUC guidelines for temperature loss) or a short modality (using TNF plus the caspase inhibitor zVAD i.v.) which needs to be terminated at ~3hrs (under IACUC guidelines for temperature loss). TNF (or
TNF/zVAD) induced manifestations include temperature loss, the production of numerous cytokines (including IL-6, IL-lb, ΜΤΡΙβ and MIP2) in the periphery, liver and intestinal inflammation and an increase of markers of cellular (LDH and CK) and liver damage (AST and ALT) in the serum. Inhibition of these TNF (or TNF/zVAD) induced manifestations can be shown by orally or IP pre-dosing with selected compounds of this invention.
Each test compound is run through the TNF/zVAD and TNF (alone) versions of the model. For example, mice (7 mice per group) were orally pre-dosed with vehicle or test compound at 50mg/kg 15 minutes before i.v. administration of mouse TNF (30μg/mouse) and zVAD (0.4mg/mouse) simultaneously. Temperature loss in the mice was measured by rectal probe. The study was terminated when the control group lost 7 degrees, per our IACUC protocol. Representative data expressed over time or at the 2.5 hour time point is provided in Figures 1 A, IB, 4A and 4B, respectively. All data are shown as means ± standard error of the mean. Data for compounds tested in this model are provided in Table 2.
Table 2.
Example No. Dose (mg/kg) % Inhibition
12 30 93
20 30 52
45 10 91
64 30 62
125 30 25
108 30 56
161 50 85
163 10 73
176 30 34 190 30 85
97 30 70
235 30 58
236 30 23
In addition to the T F/zVAD model, each compound is also tested in a TNF alone model. For the TNF (alone) version of the model, mice (7 mice per group) were orally pre- dosed with vehicle or test compound at 50mg/kg 15 minutes before i.v. administration of mouse TNF (30μg/mouse). An example of the TNF (alone) model over time and at the 6 hour time point can be seen in Figures 2A, 2B, 5A and 5B, respectively. All data are shown as means ± standard error of the mean. Data for compounds tested in this model are provided in Table 3. Table 3.
Example No. Dose (mg/kg) % Inhibition
12 50 87
20 50 51
161 50 82
190 50 56
235 50 73
Biological in vitro cell assay
The efficacy of RIP 1 inhibitors can be tested in mice in vitro using a human monocytic leukemia U937 or mouse L929 fibrosarcoma cells in a necroptosis assay (He, S. et al. 2009. Cell 137(6): 1100-1111). Cells were maintained in RPMI supplemented with 10% fetal bovine serum lOOU/ml penicillin, lOOug/ml streptomycin. For the assay, cells were suspended at 5e5 cells/ml in phenol red free RPMI supplemented with 1% fetal bovine serum, lOOU/ml penicillin, lOOug/ml streptomycin. Thirty-five (35) ul of the cell suspension was aliquotted into a white, half area assay plate. Five (5) ul each of QVD (final concentration 50uM) or compound was added to the cells and incubated at 37°C for
30 min to lh. Following the incubation, 5ul TNFa (final concentration lOOng/ml) was added to the cells and the samples were incubated overnight. The next day, cellular levels of ATP was determined using the Cell Titer-Glo Luminescent Cell Viability kit (available from Promega Corporation, Madison, Wisconsin, USA). For example, L929 (Figure 3 A) or U937 (Figure 3B) cells were treated with vehicle or ΙΟμΜ of Example 77. For example, L929 (Figure 6A) or U937 (Figure 6B) cells were treated with vehicle or indicated concentrations of the compound of Example 161. Viability was measured by quantitating cellular levels of ATP using the Cell Titer-Glo kit. All data are shown as means ± standard deviation of the mean.

Claims

What is claimed is:
1. A compound according to Formula (I):
Figure imgf000207_0001
wherein:
X is O, S, SO, S02, NH, CO, CH2, CF2, CH(CH3), CH(OH), or N(CH3);
Y is CH2 or CH2CH2;
Z1 is N, CH or CR1;
Z2 is CH or CR2;
Z3 is N, CH or CR3;
Z4 is CH or CR4;
R1 is fluoro or methyl;
one of R2 and R3 is halogen, cyano, (Ci-C6)alkyl, halo(Ci-C4)alkyl,
(Ci-C6)alkoxy, hydroxyl, B(OH)2, -COOH, halo(Ci-C4)alkylC(OH)2-,
(Ci-C4)alkoxy(Ci-C4)alkoxy, (Ci-C4)alkylS02-, (Ci-C4)alkylS02 HC(0)-,
(Ci-C4)alkylC(0) H-, ((Ci-C4)alkyl)((Ci-C4)alkyl)NC(0)-, (Ci-C4)alkylOC(0)-, (Ci-C4)alkylC(0)N(Ci-C4)alkyl)-, (Ci-C4)alkyl HC(0)-,
(Ci-C4)alkoxy(C2-C4)alkyl HC(0)-, (Ci-C4)alkoxy(C2-C4)alkylC(0) H-,
(Ci-C4)alkoxy(C2-C4)alkyl HC(0)NH-, (Ci-C4)alkylS02(C2-C4)alkyl HC(0)-,
(Ci-C4)alkyl HC(0) H-, (Ci-C4)alkylOC(0) H-, hydroxy(Ci-C4)alkylOC(0) H-, 5-6 membered heterocycloalkyl-C(O)-, 5-6 membered
heterocycloalkyl-(Ci-C )alkyl-NHC(0)-, 5-6 membered heterocycloalkyl-(Ci-C )alkoxy-, 5-6 membered heteroaryl, or 5-6 membered heteroaiyl-C(0) H,
wherein said 5-6 membered heterocycloalkyl and 5-6 membered heteroaryl are optionally substituted by 1 or 2 substituents each independently selected from the group consisting of (Ci-C )alkyl and -(Ci-C )alkyl-CN;
and the other of R2 and R3 is halogen or (Ci-C6)alkyl;
R4 is fluoro, chloro, or methyl;
R5 is H or methyl; A is phenyl, 5-6 membered heteroaryl, or 5-6 membered heterocycloalkyl, wherein the carbonyl moiety and L are substituted 1,3 on ring
A;
m is 0 or m is 1 and RA is (Ci-C4)alkyl; and
L is O, S, NH, N(CH3), CH2, CH2CH2, CH(CH3), CHF, CF2, CH20,
CH2N(CH3), CH2NH, or CH(OH);
B is an optionally substituted (C3-C6)cycloalkyl, phenyl, 5-6 membered heteroaryl, or 5-6 membered heterocycloalkyl;
wherein said (C3-C6)cycloalkyl, phenyl, 5-6 membered heteroaryl, or 5-6 membered heterocycloalkyl is unsubstituted or is substituted by one or two substituents each independently selected from halogen, (Ci-C4)alkyl, halo(Ci-C4)alkyl, (Ci-C4)alkoxy, halo(Ci-C4)alkoxy, nitro, and
(Ci-C4)alkylC(0)-;
or the moiety -L-B is (C3-C6)alkyl, (C3-C6)alkoxy, halo(C3-C6)alkoxy, (C3-C6)alkenyl, or (C3-C6)alkenyloxy;
or a salt thereof.
2. The compound or salt according to claim 1 wherein X is S, SO, S02, or CO.
3. The compound or salt according to claim 1 wherein X is CF2, CH(CH3), or CH(OH).
4. The compound or salt according to claim 1 wherein X is O, CH2, NH or N(CH3).
5. The compound or salt according to claim 1 wherein X is O or CH2.
6. The compound or salt according to any one of claims 1-5, wherein Y is CH2.
7. The compound or salt according to any one of claims 1-6, wherein Z2, Z3, and Z4 are each CH.
8. The compound or salt according to any one of claims 1-6, wherein Z1 is CR1 and Z2, Z3 and Z4 are each CH.
9. The compound or salt according to any one of claims 1-6, wherein Z2, and Z4 are each CH and Z3 is CR3.
10. The compound or salt according to any one of claims 1-6, wherein Z3, and Z4 are each CH and Z2 is CR2.
1 1. The compound or salt according to any one of claims 1-6, wherein Z2, and Z3 are each CH and Z4 is CR4.
12. The compound or salt according to any one of claims 1-6, wherein Z1 and Z3 are CH, Z2 is CR2, and Z4 is CR4.
13. The compound or salt according to any one of claims 1-6, wherein Z1 and Z3 are both N, Z2 is CH and Z4 is CH or CR4.
14. The compound or salt according to any one of claims 1-6, wherein Z1 is N, Z2 is CR2 and Z3 and Z4 are CH.
15. The compound or salt according to any one of claims 1-6, wherein Z3 is N, and Z2, Z3 and Z4 are CH.
16. The compound or salt according to any one of claims 1-6 or 8, wherein R1 is methyl.
17. The compound or salt according to any one of claims 1-6, 10, 12, or 14, wherein R2 is halogen, cyano, (d-C6)alkyl, hydroxyl, B(OH)2, -COOH, halo(Ci-C4)alkylC(OH)2-, (Ci-C4)alkoxy(Ci-C4)alkoxy, or 5-6 membered heteroaryl, wherein said 5-6 membered heteroaryl is optionally substituted by a (Ci-C3)alkyl substituent.
18. The compound or salt according to any one of claims 1-6, wherein Z3 is CH.
19. The compound or salt according to any one of claims 1-6, or 9 , wherein R3 is halogen, (Ci-C6)alkyl, halo(Ci-C4)alkyl, (Ci-C6)alkoxy, B(OH)2, -COOH, (Ci-C4)alkylS02-, (Ci-C4)alkylS02 HC(0)-,
(Ci-C4)alkylC(0) H-, ((Ci-C4)alkyl)((Ci-C4)alkyl)NC(0)-,
(Ci-C4)alkylOC(0)-, (Ci-C4)alkylC(0)N(Ci-C4)alkyl)-,
(Ci-C4)alkoxy(C2-C4)alkyl HC(0) H-,
(Ci-C4)alkylS02(C2-C4)alkyl HC(0)-, (Ci-C4)alkyl HC(0) H-,
(Ci-C4)alkylOC(0) H-, hydroxy(Ci-C4)alkylOC(0) H-, 5-6 membered heterocycloalkyl-C(O)-, 5-6 membered
heterocycloalkyl-(Ci-C4)alkyl-NHC(0)-, 5-6 membered
heterocycloalkyl-(Ci-C )alkoxy-, 5-6 membered heteroaryl, or 5-6 membered heteroaryl-C(0) H, herein said 5-6 membered heterocycloalkyl and 5-6 membered heteroaryl are optionally substituted by (Ci-C3)alkyl or
-(Ci-C3)alkyl-CN.
20. The compound or salt to any one of claims 1-6, wherein Z2 is CH.
21. The compound or salt according to any one of claims 1-6, 10, 12, or 14, wherein R2 is fluoro, chloro, bromo, -CN, -CH3, -OH, B(OH)2, CF3C(OH)2-, CH3OCH2CH20-, 5H-tetrazol-5-yl, pyrazol-3-yl, or 5-methyl- l,3,4-oxadiazol-2-yl.
22. The compound or salt according to any one of claims 1-6, or 9, wherein R3 is fluoro, chloro, bromo, -OCH3, B(OH)2, -COOH, CH3S02-, CH3S02 HC(0)-, CH3C(0) H-, (CH3)2NC(0)-, CH3OC(0)-,
(CH3)C(0)N(CH3)-, HOCH2CH2C(0) H-, CH3OCH2CH2NHC(0) H-, CH3S02CH2CH2NHC(0)-, CH3CH2 HC(0) H-, CH3OC(0) H-, mo holin-4-yl-CO-, pyrrolidin- 1 -yl-CH2CH2 HC(0)-, tetrahydrofuran-2-yl- CH20-, pyrrolidin- l-yl-CH2CH20-, tetrazol-5-yl, l-(2-cyanoethyl)-tetrazol-5- yl, pyrazol-l-yl, pyrazol-3-yl, l-methyl-pyrazol-3-yl, l-methyl-pyrrol-4-yl- C(0) H-, 5-methyl-l,3,4-oxadiazol-2-yl or 5-oxo-4,5-dihydro-l,3,4- oxadiazol-2-yl.
23. The compound or salt according to any one of claims 1-6, or 11-13, , wherein R4 is fluoro or R4 is methyl.
24. The compound or salt according to any one of claims 1-23, wherein R5 is H.
25. The compound or salt according to any one of claims 1-23, wherein R5 is methyl.
26. The compound or salt according to any one of claims 1-25, wherein
A is phenyl, 5-6 membered heteroaryl, or 5-6 membered heterocycloalkyl, wherein the carbonyl moiety and L are substituted 1,3 on ring A.
27. The compound or salt according to any one of claims 1-25, wherein A is furyl, thienyl, oxazolyl, isoxazolyl, thiazolyl or oxadiazolyl.
28. The compound or salt according to any one of claims 1-25, wherein A is pyrrolyl, pyrazolyl, imidazolyl, triazolyl, or tetrazolyl.
29. The compound or salt according to any one of claims 1-25, wherein
A is piperidinyl, pyrrolidinyl, phenyl or pyridyl.
30. The compound or salt according to any one of claims 1-25, according to Formula (II):
Figure imgf000211_0001
wherein:
A1 is C,
A4 is C or N, and A2, A3, and A5 are each independently selected from CH, CRA, O, S, N, NH and NRA to form a furyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl or tetrazolyl ring moiety, wherein said ring moiety contains 0 or 1 of CRA and NRA.
31. The compound or salt according to claim 30, wherein A1 is C, A4 is C or N, and A2, A3, and A5 are each independently selected from CH, O, N, and NH to form an oxazolyl, isoxazolyl, oxadiazolyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl or tetrazolyl ring moiety.
32. The compound or salt according to claim 30, wherein A1 and A4 are each C, and A2, A3 and A5 are each independently selected from N and NH to form a triazolyl ring moiety.
33. The compound or salt according to any one of claims 1-25 according to Formula (III):
Figure imgf000212_0001
wherein s is 0 or 1, A is N .
34. The compound or salt according to claim 33, wherein m is 0 and A is an unsubstituted piperidinyl or pyrrolidinyl moiety.
35. The compound or salt according to any one of claims 1-25, wherein m is O.
36. The compound or salt according to any one of claims 1-25 wherein m is 1 and RA is methyl.
37. The compound or salt according to any one of claims 1-25, according to Formula (IV):
Figure imgf000213_0001
wherein:
A6, A7, A8, and A9 are each CH;
one of A6, A7, A8, and A9 is CRA and the others of A6, A7, A8, and A9 are CH;
one of A6, A7, A8, and A9 is N and the others of A6, A7, A8, and A9 are
CH;
one of A6, A7, A8, and A9 is N-0 and the other of A6, A7, A8, and A9 are
CH.
38. The compound or salt according to any one of claims 1-37, wherein L is O, S, N(CH3), CH2, CH2CH2, CH(CH3), CF2, CH20, CH2N(CH3), or CH(OH).
39. The compound or salt according to any one of claims 1-37, wherein
L is O, CH2, or H.
40. The compound or salt according to any one of claims 1-39, wherein Bis an optionally substituted pyrazolyl, thienyl, pyndinyl (pyridyl), oxo-pyridyl, pyrimidinyl, isoxazolyl, morpholinyl, or tetrahydrofuranyl, wherein the pyrazolyl, thienyl, pyndinyl, oxo-pyridyl, pyrimidinyl, isoxazolyl, morpholinyl, or tetrahydrofuranyl is optionally substituted by one or two independently selected (Ci-C4)alkyl substituents.
41. The compound or salt according to any one of claims 1-39, wherein B is thien-2-yl, 5-methyl-thien-2-yl, pyrazol-l-yl, 3,5-dimethylpyrazol-l-yl, 4-methylpyrazol-l-yl, 3,5-dimethylisoxazol-4-yl, tetrahydrofuran-2-yl, morpholin-4-yl, pyridin-2-yl, 2-oxo-pyridin-l-yl, 6-methylpyridin-3-yl, and 2-methylpyrimidin-5-yl.
42. The compound or salt according to any one of claims 1-39, wherein B is unsubstituted cyclopentyl or cyclohexyl.
43. The compound or salt according to any one of claims 1-39, wherein B is unsubstituted phenyl.
44. The compound or salt according to any one of claims 1-39, wherein B is phenyl, substituted by 1 or 2 substituents independently selected from halogen, (Ci-C4)alkyl, halo(Ci-C4)alkyl, (Ci-C4)alkoxy, halo(Ci-C4)alkoxy, nitro, and (Ci-C4)alkylC(0)-.
45. The compound or salt according to any one of claims 1-39, wherein B is phenyl, substituted by 1 or 2 substituents independently selected from halogen, (Ci-C3)alkyl and (Ci-C3)alkoxy.
46. The compound or salt according to any one of claims 1-39, wherein B is phenyl, substituted by 1 or 2 substituents independently selected from iodo, fluoro, chloro, bromo, methyl and methoxy.
47. The compound or salt according to any one of claims 1-39, wherein B is 2-methylphenyl, 4-methylphenyl, 2-trifluoromethylphenyl,
2-fluorophenyl, 3 -fluorophenyl, 4-fluorophenyl, 2-iodophenyl,
3-bromophenyl, 4-bromophenyl, 4-chlorophenyl, 2,5-difluorophenyl, 2,4-difluorophenyl, 3,4-difluorophenyl, 3,5-difluorophenyl, or
4-methoxyphenyl .
48. The compound or salt according to any one of claims 1-37, wherein -L-B is (C3-C6)alkyl, (C3-C6)alkoxy, or (C3-C5)alkenyloxy.
49. The compound or salt according to any one of claims 1-37, wherein
-L-B is -OCH2CH=CH2, -CH2CH2CH2CH2CH3, -OCH2CH2CH2CH3,
-CH2CH2CH3, -CH2CH(CH3)2 or -CH2CH2CH(CH3)2.
50. A compound which is (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b] [ 1 ,4]oxazepin-3 -yl)-4H- 1 ,2,4-triazole-3-carboxamide.
51. A compound which is (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5- tetrahydrobenzo[b] [ 1 ,4]oxazepin-3 -yl)-4H- 1 ,2,4-triazole-3-carboxamide, or a
pharmaceutically acceptable salt thereof.
52. A compound which is (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5- tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3-carboxamide.
53. A compound which is (S)-5-benzyl-N-(7-chloro-2-oxo-2,3,4,5- tetrahydro-lH-benzo[b]azepin-3-yl)-4H-l,2,4-triazole-3-carboxamide, or a
pharmaceutically acceptable salt thereof.
54. The compound or salt according to any one of claims 1-49, wherein the salt is a pharmaceutically acceptable salt of said compound.
55. λ A pharmaceutical composition comprising the compound or pharmaceutically acceptable salt thereof according to any one of claims 50-54, and one or more
pharmaceutically acceptable excipients.
56. A method of treating a RIPl kinase-mediated disease or disorder comprising administering a therapeutically effective amount of the compound or pharmaceutically acceptable salt thereof according to any one of claims 50-54 to a human in need thereof.
57. The compound, or pharmaceutically acceptable salt thereof, according to any one of claims 50-54 for use in therapy.
58. Use of the compound, or pharmaceutically acceptable salt thereof, according to any one of claims 50-54 in the manufacture of a medicament for the treatment of a RIP1 kinase-mediated disease or disorder.
59. The method according to claim 56, wherein the disease or disorder is selected from inflammatory bowel disease, Crohn's disease, ulcerative colitis, psoriasis, retinal detachment, retinitis pigmentosa, macular degeneration, pancreatitis, atopic dermatitis, rheumatoid arthritis, spondyloarthritis, gout, SoJIA, systemic lupus erythematosus, Sjogren's syndrome, systemic scleroderma, anti-phospholipid syndrome, vasculitis, osteoarthritis, non-alcohol steatohepatitis, alcohol steatohepatitis, autoimmune hepatitis autoimmune hepatobiliary diseases, primary sclerosing cholangitis, nephritis, Celiac disease, autoimmune ITP, transplant rejection, ischemia reperfusion injury of solid organs, sepsis, systemic inflammatory response syndrome, cerebrovascular accident, myocardial infarction, Huntington's disease, Alzheimer's disease, Parkinson's disease, allergic diseases, asthma, atopic dermatitis, multiple sclerosis, type I diabetes, Wegener's granulomatosis, pulmonary sarcoidosis, Behcet's disease, interleukin-1 converting enzyme associated fever syndrome, chronic obstructive pulmonary disease, tumor necrosis factor receptor-associated periodic syndrome, and peridontitis.
60. The use according to claim 58, wherein the disease or disorder is selected from inflammatory bowel disease, Crohn's disease, ulcerative colitis,
psoriasis, retinal detachment, retinitis pigmentosa, macular degeneration,
pancreatitis, atopic dermatitis, rheumatoid arthritis, spondyloarthritis, gout,
SoJIA, systemic lupus erythematosus, Sjogren's syndrome, systemic
scleroderma, anti-phospholipid syndrome, vasculitis, osteoarthritis, non-alcohol steatohepatitis, alcohol steatohepatitis, autoimmune hepatitis autoimmune
hepatobiliary diseases, primary sclerosing cholangitis, nephritis, Celiac disease, autoimmune ITP, transplant rejection, ischemia reperfusion injury of solid
organs, sepsis, systemic inflammatory response syndrome, cerebrovascular
accident, myocardial infarction, Huntington's disease, Alzheimer's disease,
Parkinson's disease, allergic diseases, asthma, atopic dermatitis, multiple
sclerosis, type I diabetes, Wegener's granulomatosis, pulmonary sarcoidosis,
Behcet's disease, interleukin-1 converting enzyme associated fever syndrome, chronic obstructive pulmonary disease, tumor necrosis factor receptor- associated periodic syndrome, and peridontitis.
PCT/IB2014/059004 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors WO2014125444A1 (en)

Priority Applications (49)

Application Number Priority Date Filing Date Title
PL14705891T PL2956452T3 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
CN201480021120.8A CN105121432B (en) 2013-02-15 2014-02-14 Heterocycleamide as kinase inhibitor
RS20180698A RS57332B1 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
BR122017002949A BR122017002949A2 (en) 2013-02-15 2014-02-14 heterocyclic amides as kinase inhibitors
CA2900695A CA2900695C (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
UAA201508769A UA118259C2 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
BR112015019627-6A BR112015019627B1 (en) 2013-02-15 2014-02-14 COMPOUNDS OF HETEROCYCLIC AMIDES, PHARMACEUTICAL COMPOSITIONS COMPRISING SUCH COMPOUNDS AND USES THEREOF TO TREAT ULCERATIVE COLITIS, CROHN'S DISEASE, RHEUMATOID ARTHRITIS, PSORIASIS, SPONDYLOARTHRITIS, SYSTEMIC-ON-APPEARING JUVENILE IDIOPATHIC ARTHRITIS (SOJIA), OSTEOARTHRITIS AND MULTIPLE Sclerosis
EP19154374.3A EP3508484B1 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
SI201430738T SI2956452T1 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
US14/763,183 US9556152B2 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
MA44820A MA44820B1 (en) 2013-02-15 2014-02-14 Heterocyclic amides for use as kinase inhibitors
KR1020157024819A KR102228764B1 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
BR122017002946A BR122017002946A2 (en) 2013-02-15 2014-02-14 heterocyclic amides as kinase inhibitors
EA201591506A EA028991B1 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
NZ628447A NZ628447A (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
CR20190511A CR20190511A (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
CR20190512A CR20190512A (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
EP14705891.1A EP2956452B1 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
MA38326A MA38326B1 (en) 2013-02-15 2014-02-14 Heterocyclic amides for use as kinase inhibitors
PL18155836T PL3342771T3 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
BR122017002951A BR122017002951A2 (en) 2013-02-15 2014-02-14 heterocyclic amides as kinase inhibitors
DK14705891.1T DK2956452T3 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors.
ES14705891.1T ES2672530T3 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
JP2015557554A JP6321045B2 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
AU2014217453A AU2014217453B2 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
KR1020197004425A KR102267977B1 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
LTEP14705891.1T LT2956452T (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
SG11201505796TA SG11201505796TA (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
EP18152185.7A EP3375780B1 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
MX2015010572A MX360715B (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors.
EP18155836.2A EP3342771B1 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
ZA2015/05267A ZA201505267B (en) 2013-02-15 2015-07-21 Heterocyclic amides as kinase inhibitors
PH12015501675A PH12015501675A1 (en) 2013-02-15 2015-07-28 Heterocyclic amides as kinase inhibitors
IL240280A IL240280B (en) 2013-02-15 2015-07-30 Heterocyclic amides as kinase inhibitors
CR20150420A CR20150420A (en) 2013-02-15 2015-08-14 HETEROCYCLIC AMIDES AS INHIBITORS OF KINASES
HK16101405.9A HK1213545A1 (en) 2013-02-15 2016-02-05 Heterocyclic amides as kinase inhibitors
US15/271,581 US9624202B2 (en) 2013-02-15 2016-09-21 Heterocyclic amides as kinase inhibitors
US15/459,141 US10292987B2 (en) 2013-02-15 2017-03-15 Heterocyclic amides as kinase inhibitors
US15/835,747 US20180098998A1 (en) 2013-02-15 2017-12-08 Heterocyclic amides as kinase inhibitors
IL256500A IL256500B (en) 2013-02-15 2017-12-22 Heterocyclic amides as kinase inhibitors
IL256502A IL256502B (en) 2013-02-15 2017-12-22 Heterocyclic amides as kinase inhibitors
PH12018500342A PH12018500342A1 (en) 2013-02-15 2018-02-15 Heterocyclic amides as kinase inhibitors
PH12018500341A PH12018500341A1 (en) 2013-02-15 2018-02-15 Heterocyclic amides as kinase inhibitors
PH12018500343A PH12018500343A1 (en) 2013-02-15 2018-02-15 Heterocyclic amides as kinase inhibitors
HRP20180900TT HRP20180900T1 (en) 2013-02-15 2018-06-08 Heterocyclic amides as kinase inhibitors
CY20181100604T CY1120320T1 (en) 2013-02-15 2018-06-08 Heterocyclic Amides As Motion Suspensions
US16/299,254 US10933070B2 (en) 2013-02-15 2019-03-12 Heterocyclic amides as kinase inhibitors
US16/408,718 US10940154B2 (en) 2013-02-15 2019-05-10 Heterocyclic amides as kinase inhibitors
CY20211100121T CY1123915T1 (en) 2013-02-15 2021-02-12 HETEROCYCLIC AMIDES AS KINASE INHIBITORS

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361765664P 2013-02-15 2013-02-15
US61/765,664 2013-02-15
US201361790044P 2013-03-15 2013-03-15
US61/790,044 2013-03-15

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US14/763,183 A-371-Of-International US9556152B2 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
EP18152185.7A Previously-Filed-Application EP3375780B1 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors
US15/271,581 Continuation US9624202B2 (en) 2013-02-15 2016-09-21 Heterocyclic amides as kinase inhibitors

Publications (1)

Publication Number Publication Date
WO2014125444A1 true WO2014125444A1 (en) 2014-08-21

Family

ID=50151350

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2014/059004 WO2014125444A1 (en) 2013-02-15 2014-02-14 Heterocyclic amides as kinase inhibitors

Country Status (37)

Country Link
US (6) US9556152B2 (en)
EP (4) EP2956452B1 (en)
JP (3) JP6321045B2 (en)
KR (2) KR102228764B1 (en)
CN (3) CN106928203B (en)
AU (1) AU2014217453B2 (en)
BR (4) BR122017002949A2 (en)
CA (1) CA2900695C (en)
CL (1) CL2015002279A1 (en)
CR (3) CR20190512A (en)
CY (2) CY1120320T1 (en)
DK (2) DK2956452T3 (en)
DO (3) DOP2015000196A (en)
EA (2) EA028991B1 (en)
ES (3) ES2814556T3 (en)
HK (3) HK1213545A1 (en)
HR (2) HRP20180900T1 (en)
HU (2) HUE053215T2 (en)
IL (3) IL240280B (en)
LT (2) LT3342771T (en)
MA (1) MA44820B1 (en)
MX (1) MX360715B (en)
MY (2) MY178552A (en)
NZ (1) NZ628447A (en)
PE (1) PE20151752A1 (en)
PH (4) PH12015501675A1 (en)
PL (2) PL3342771T3 (en)
PT (2) PT2956452T (en)
RS (2) RS61439B1 (en)
SG (1) SG11201505796TA (en)
SI (2) SI3342771T1 (en)
TR (1) TR201808112T4 (en)
TW (4) TWI638815B (en)
UA (1) UA118259C2 (en)
UY (1) UY35330A (en)
WO (1) WO2014125444A1 (en)
ZA (1) ZA201505267B (en)

Cited By (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016027253A1 (en) 2014-08-21 2016-02-25 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as rip1 kinase inhibitors as medicaments
WO2016128936A1 (en) 2015-02-13 2016-08-18 Glaxosmithkline Intellectual Property Development Limited Crystalline forms of (s)-5-benzyl-n-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][1,4]oxazepin-3-yl)-4h-1,2,4-triazole-3-carboxamide
WO2016139181A1 (en) 2015-03-02 2016-09-09 Apeiron Biologics Ag Bicyclic tetrahydrothiazepine derivatives useful for the treatment of neoplastic and/or infectious diseases
US9499521B2 (en) 2014-12-11 2016-11-22 President And Fellows Of Harvard College Inhibitors of cellular necrosis and related methods
WO2016185423A1 (en) 2015-05-19 2016-11-24 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
WO2016208592A1 (en) * 2015-06-22 2016-12-29 大日本住友製薬株式会社 Bicyclic heterocyclic amide derivative
WO2017004500A1 (en) * 2015-07-02 2017-01-05 Genentech, Inc. Bicyclic lactams and methods of use thereof
US9556152B2 (en) 2013-02-15 2017-01-31 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
WO2017069279A1 (en) * 2015-10-23 2017-04-27 武田薬品工業株式会社 Heterocyclic compound
WO2017096301A1 (en) * 2015-12-04 2017-06-08 Denali Therapeutics Inc. Isoxazolidine derived inhibitors of receptor interacting protein kinase 1 (ripk 1)
WO2017109724A1 (en) 2015-12-21 2017-06-29 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US9815850B2 (en) 2016-02-05 2017-11-14 Denali Therapeutics Inc. Compounds, compositions and methods
WO2018007973A2 (en) 2016-07-06 2018-01-11 Glaxosmithkline Intellectual Property Development Limited Process and intermediates for preparing benzoxazepines
WO2018073193A1 (en) 2016-10-17 2018-04-26 F. Hoffmann-La Roche Ag Bicyclic pyridone lactams and methods of use thereof
US9957281B2 (en) 2015-07-22 2018-05-01 Enanta Pharmaceuticals, Inc. Benzodiazepine derivatives as RSV inhibitors
WO2018083288A1 (en) 2016-11-07 2018-05-11 Bayer Aktiengesellschaft Substituted sulfonyl amides for controlling animal pests
WO2018092089A1 (en) 2016-11-18 2018-05-24 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
WO2018100070A1 (en) 2016-12-02 2018-06-07 F. Hoffmann-La Roche Ag Bicyclic amide compounds and methods of use thereof
WO2018109097A1 (en) 2016-12-16 2018-06-21 F. Hoffmann-La Roche Ag Inhibitors of rip1 kinase and methods of use thereof
WO2018117196A1 (en) 2016-12-20 2018-06-28 大日本住友製薬株式会社 Drug targeting cancer stem cell
WO2018154520A1 (en) 2017-02-27 2018-08-30 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
WO2019012063A1 (en) 2017-07-14 2019-01-17 F. Hoffmann-La Roche Ag Bicyclic ketone compounds and methods of use thereof
WO2019072942A1 (en) 2017-10-11 2019-04-18 F. Hoffmann-La Roche Ag Bicyclic compounds for use as rip1 kinase inhibitors
WO2019086494A1 (en) 2017-10-31 2019-05-09 F. Hoffmann-La Roche Ag Bicyclic sulfones and sulfoxides and methods of use thereof
WO2019123219A1 (en) 2017-12-20 2019-06-27 Glaxosmithkline Intellectual Property Development Limited Crystalline salt form of (s)-5-benzyl-n-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][1,4]oxazepin-3-yl)-4h-1,2,4-triazole-3-carboxamide
WO2019130230A1 (en) 2017-12-29 2019-07-04 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US10358441B2 (en) 2017-02-16 2019-07-23 Enanta Pharmaceuticals, Inc. Processes for the preparation of benzodiazepine derivatives
US10398706B2 (en) 2017-01-06 2019-09-03 Enanta Pharmaceuticals, Inc. Heteroaryldiazepine derivatives as RSV inhibitors
WO2019204537A1 (en) 2018-04-20 2019-10-24 Genentech, Inc. N-[4-oxo-2,3-dihydro-1,5-benzoxazepin-3-yl]-5,6-dihydro-4h-pyrrolo[1,2-b]pyrazol e-2-carboxamide derivatives and related compounds as rip1 kinase inhibitors for treating e.g. irritable bowel syndrome (ibs)
WO2019213445A1 (en) 2018-05-03 2019-11-07 Rigel Pharmaceuticals, Inc. Rip1 inhibitory compounds and methods for making and using the same
WO2019213447A1 (en) 2018-05-03 2019-11-07 Rigel Pharmaceuticals, Inc. Rip1 inhibitory compounds and methods for making and using the same
WO2019224774A1 (en) 2018-05-23 2019-11-28 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as rip1 kinase inhibitors
WO2019224773A1 (en) 2018-05-23 2019-11-28 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as rip1 kinase inhibitors
US10501422B2 (en) 2017-11-13 2019-12-10 Enanta Pharmaceuticals, Inc. Processes for the resolution of benzodiazepin-2-one and benzoazepin-2-one derivatives
WO2020044206A1 (en) 2018-08-29 2020-03-05 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors for use in the treatment cancer
CN110913858A (en) * 2017-05-17 2020-03-24 戴纳立制药公司 Kinase inhibitors and uses thereof
WO2020088194A1 (en) 2018-11-02 2020-05-07 中国科学院上海药物研究所 Heterocyclic amide for inhibiting rip1 kinase and uses thereof
US10647711B2 (en) 2017-11-13 2020-05-12 Enanta Pharmaceuticals, Inc. Azepin-2-one derivatives as RSV inhibitors
WO2020146615A1 (en) 2019-01-11 2020-07-16 Genentech, Inc. Bicyclic pyrrolotriazolr ketone compounds and methods of use thereof
WO2020151589A1 (en) 2019-01-25 2020-07-30 北京赛特明强医药科技有限公司 Acylamino bridged heterocyclic compound, and composition and application thereof
US10752598B2 (en) 2017-06-07 2020-08-25 Enanta Pharmaceuticals, Inc. Aryldiazepine derivatives as RSV inhibitors
US10759816B2 (en) 2016-01-15 2020-09-01 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as RSV inhibitors
WO2020234716A1 (en) 2019-05-17 2020-11-26 Glaxosmithkline Intellectual Property Development Limited Matrix composition comprising (s)-5-benzyl-n-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][1,4]oxazepin-3-yl)-4h-1,2,4-triazole-3-carboxamide
US10851115B2 (en) 2017-06-30 2020-12-01 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as RSV inhibitors
US10881666B2 (en) 2017-09-29 2021-01-05 Enanta Pharmaceuticals, Inc. Combination pharmaceutical agents as RSV inhibitors
US10898469B2 (en) 2016-02-26 2021-01-26 Sumitomo Dainippon Pharma Co., Ltd. Imidazolylamide derivative
WO2021046382A1 (en) 2019-09-06 2021-03-11 Rigel Pharmaceuticals, Inc. Rip1 inhibitory compounds and methods for making and using the same
WO2021046437A1 (en) 2019-09-06 2021-03-11 Rigel Pharmaceuticals, Inc. Rip1 inhibitory compounds and methods for making and using the same
US10975094B2 (en) 2018-04-11 2021-04-13 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as RSV inhibitors
WO2021092336A1 (en) 2019-11-07 2021-05-14 Rigel Pharmaceuticals, Inc. Heterocyclic rip1 inhibitory compounds
US11072618B2 (en) 2016-12-09 2021-07-27 Denali Therapeutics Inc. Compounds, compositions and methods
US11091501B2 (en) 2017-06-30 2021-08-17 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as RSV inhibitors
US11179400B2 (en) 2019-04-09 2021-11-23 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as RSV inhibitors
EP3872077A4 (en) * 2018-11-02 2021-12-01 Shanghai Institute of Materia Medica, Chinese Academy of Sciences Heterocyclic amide for inhibiting rip1 kinase and uses thereof
WO2021245070A1 (en) 2020-06-02 2021-12-09 Sanofi Isoxazolidines as ripk1 inhibitors and use thereof
US11254664B2 (en) 2019-03-18 2022-02-22 Enanta Pharmaceuticals, Inc. Benzodiazepine derivatives as RSV inhibitors
EP3816163A4 (en) * 2018-06-26 2022-04-13 Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences Cell necrosis inhibitor, preparation method therefor and use thereof
US11420976B2 (en) 2020-01-24 2022-08-23 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as anti-viral agents
WO2022192533A1 (en) * 2021-03-11 2022-09-15 Rigel Pharmaceuticals, Inc. Heterocyclic rip1 kinase inhibitors
WO2022212809A1 (en) 2021-04-02 2022-10-06 Genentech, Inc. Processes for making bicyclic ketone compounds
US11505558B1 (en) 2019-10-04 2022-11-22 Enanta Pharmaceuticals, Inc. Antiviral heterocyclic compounds
US11534439B2 (en) 2020-07-07 2022-12-27 Enanta Pharmaceuticals, Inc. Dihydroquinoxaline and dihydropyridopyrazine derivatives as RSV inhibitors
TWI790227B (en) * 2017-04-14 2023-01-21 美商百健Ma公司 Inhibiting agents for bruton’s tyrosine kinase
US11572367B2 (en) 2019-10-04 2023-02-07 Enanta Pharmaceuticals, Inc. Antiviral heterocyclic compounds
US11584758B2 (en) 2020-04-02 2023-02-21 Rigel Pharmaceuticals, Inc. RIP1K inhibitors
WO2023068881A1 (en) * 2021-10-22 2023-04-27 주식회사 보로노이바이오 Aryl or heteroaryl derivative, and pharmaceutical composition comprising same as active ingredient for prevention or treatment of kinase-related diseases
WO2023083847A1 (en) 2021-11-11 2023-05-19 Sanofi Isoxazolidines as ripk1 inhibitors and use thereof
US11667643B2 (en) 2020-07-01 2023-06-06 Rigel Pharmaceuticals, Inc. RIP1K inhibitors
RU2797922C2 (en) * 2017-07-14 2023-06-13 Ф. Хоффманн-Ля Рош Аг Bicyclic ketones and methods of their use
US11690850B2 (en) 2019-11-26 2023-07-04 Board Of Regents, The University Of Texas System Inhibitors of receptor interacting protein kinase I for the treatment of disease
US11718612B2 (en) 2019-09-06 2023-08-08 Board Of Regents, The University Of Texas System Inhibitors of receptor interacting protein kinase I for the treatment of disease
US11753381B2 (en) 2019-09-27 2023-09-12 Board Of Regents, The University Of Texas System Inhibitors of receptor interacting protein kinase I for the treatment of disease
US11767310B2 (en) 2021-08-10 2023-09-26 Abbvie Inc. Nicotinamide RIPK1 inhibitors
WO2024040155A1 (en) 2022-08-19 2024-02-22 Genzyme Corporation Isoxazolidines as ripk1 inhibitors and use thereof
WO2024044183A1 (en) * 2022-08-22 2024-02-29 Enanta Pharmaceuticals, Inc. Fused heterobicyclic antiviral agents
EP4110765A4 (en) * 2020-02-28 2024-03-06 Univ Texas Inhibitors of receptor interacting protein kinase i for the treatment of disease
US11945824B2 (en) 2020-10-19 2024-04-02 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as anti-viral agents

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10183940B2 (en) * 2014-11-14 2019-01-22 Boehringer Ingelheim International Gmbh Aryl and heteroaryl-fused tetrahydro-1,4-oxazepine amides as somatostatin receptor subtype 4 (SSTR4) agonists
CN110769853B (en) * 2017-04-17 2022-06-03 北京生命科学研究所 Treatment of male aging
CN110407770B (en) * 2018-04-27 2022-12-30 复旦大学 3-substituted-1, 5-benzazepine compound and pharmaceutical use thereof
CN110642874B (en) * 2018-06-26 2023-03-28 中国科学院上海有机化学研究所 Cell necrosis inhibitor and preparation method and application thereof
CN109134448B (en) * 2018-10-16 2020-11-27 中南大学湘雅医院 Heterocyclic compound and salt thereof, preparation method, application and medicine
KR20220016223A (en) * 2019-05-31 2022-02-08 메드샤인 디스커버리 아이엔씨. Bicyclic compounds that are RIP-1 kinase inhibitors and applications thereof
US20210040115A1 (en) * 2019-08-09 2021-02-11 Bisichem Co., Ltd. Fused ring heteroaryl compounds as ripk1 inhibitors
JP2023538097A (en) * 2020-08-18 2023-09-06 ハチメド リミテッド Pyrimidinone compounds and uses thereof
WO2022171111A1 (en) * 2021-02-10 2022-08-18 Zai Lab (Us) Llc Heteroaryl-fused bicyclic compound as rip1-kinase inhibitors and uses thereof
WO2022171110A1 (en) * 2021-02-10 2022-08-18 Zai Lab (Us) Llc Fused tricyclic compounds as rip1-kinase inhibitors and uses thereof
CN113045560B (en) * 2021-03-30 2022-09-06 港科鹏禾生物(苏州)有限公司 Amide derivative and preparation method and application thereof
WO2022231928A1 (en) * 2021-04-27 2022-11-03 Merck Sharp & Dohme Llc Ripk1 inhibitors and methods of use
CN115246796A (en) * 2021-04-27 2022-10-28 中国科学院上海有机化学研究所 Compound for inhibiting programmed cell death and preparation method thereof
CN115806557A (en) * 2021-09-14 2023-03-17 中国科学院上海有机化学研究所 Compound for inhibiting programmed cell death and preparation method thereof
CN113876778B (en) * 2021-11-05 2023-04-14 中日友好医院(中日友好临床医学研究所) Application of KW2449 in preparation of medicine for improving rheumatoid arthritis
WO2023119210A1 (en) * 2021-12-24 2023-06-29 제일약품주식회사 Novel compound as ripk1 inhibitor and pharmaceutical composition comprising same
CN114736197B (en) * 2022-06-13 2022-09-13 南京医工医药技术有限公司 Imidazolidinone derivatives and their use
WO2023240379A1 (en) * 2022-06-13 2023-12-21 南京医工医药技术有限公司 Imidazolinone derivative and use thereof
CN117447460A (en) * 2022-07-13 2024-01-26 南京天印健华医药科技有限公司 Heterocyclic compounds as RIPK1 inhibitors

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004098589A1 (en) * 2003-05-02 2004-11-18 Elan Pharmaceuticals, Inc. 4- bromo - 5 - (2- chloro - benzoylamino) - 1h - pyrazole - 3 - carboxylic acid amide derivatives and related compounds as bradykinin b1 receptor antagonists for the treatment of inflammatory diseases
WO2007075772A2 (en) * 2005-12-20 2007-07-05 President And Fellows Of Harvard College Compounds, screens, and methods of treatment
WO2008106077A1 (en) 2007-02-28 2008-09-04 Merck & Co., Inc. Substituted benzodiazepinones, benzoxazepinones and benzothiazepinones as sodium channel blockers

Family Cites Families (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5239065B2 (en) 1971-11-17 1977-10-03
EP0166357A3 (en) * 1984-06-26 1988-10-26 Merck & Co. Inc. Benzofused lactams and pharmaceutical compositions containing them
US4692522A (en) 1985-04-01 1987-09-08 Merck & Co., Inc. Benzofused lactams useful as cholecystokinin antagonists
US5206234A (en) 1990-10-22 1993-04-27 Merck & Co., Inc. Benzolactam analogs as antagonists of cck
JP3358069B2 (en) 1991-12-24 2002-12-16 武田薬品工業株式会社 Tricyclic heterocycles, their production method and agents
AU720378B2 (en) 1995-06-07 2000-06-01 Merck & Co., Inc. Novel N-(4-oxo-2,3,4,5-tetrahydro-1H-1, 5-benzodiazepin-3yl)-3-amides
WO1998041510A1 (en) 1997-03-14 1998-09-24 Shionogi & Co., Ltd. Novel benzolactam derivatives and medicinal compositions comprising the same
US6228854B1 (en) 1997-08-11 2001-05-08 Cor Therapeutics, Inc. Selective factor Xa inhibitors
FR2781483A1 (en) * 1998-07-21 2000-01-28 Hoechst Marion Roussel Inc Thioazepinone derivatives having Src domain SH2 inhibiting and osteoclast-mediated calcium resorption from bone inhibiting activity, for treatment of osteoporosis
WO2001002358A2 (en) 1999-07-06 2001-01-11 Vertex Pharmaceuticals Incorporated Cyclized amide derivatives for treating or preventing neuronal damage
WO2001035964A1 (en) 1999-11-18 2001-05-25 Antexpharma, Inc. Substituted 1-benzazepines and derivatives thereof
EP1268433A1 (en) 2000-04-03 2003-01-02 Bristol-Myers Squibb Pharma Company Cyclic lactams as inhibitors of a-beta-protein production
US6713476B2 (en) 2000-04-03 2004-03-30 Dupont Pharmaceuticals Company Substituted cycloalkyls as inhibitors of a beta protein production
NZ516746A (en) 2000-06-01 2004-02-27 Bristol Myers Squibb Pharma Lactams substituted by cyclic succinates as inhibitors of A-beta protein production
DE10054279A1 (en) 2000-11-02 2002-05-16 Apotech Res & Dev Ltd Use of ligands from death receptors or RIP to trigger caspase-independent cell death and compounds to inhibit caspase-independent cell death
EP1392317A4 (en) 2001-05-16 2005-10-19 Antexpharma Inc Substituted 1-benzazepines and derivatives thereof
US7057046B2 (en) * 2002-05-20 2006-06-06 Bristol-Myers Squibb Company Lactam glycogen phosphorylase inhibitors and method of use
EP1620567A1 (en) 2003-04-15 2006-02-01 Hans-Jürgen Thiesen Method for diagnosing rheumatoid arthritis or osteoarthritis
WO2005028664A2 (en) 2003-09-17 2005-03-31 University Of Massachusetts Modulation of programmed necrosis
EP1640012A1 (en) 2004-09-24 2006-03-29 Salama, Zoser B. nat.rer.Dr. Pharmaceuticals agents comprising blood components 10 kDa and their use for prophylaxis and treatment of defects of the immune system
US20060067942A1 (en) 2004-09-24 2006-03-30 Salama Zoser B Pharmaceutical agent comprising amino acids, peptides, proteins and/or fractions and fragments thereof and the use of same in the prophylaxis and treatment of immune system deficiency in humans and animals
FR2894968B1 (en) 2005-12-20 2008-02-22 Trophos Sa NOVEL CHOLEST-4-EN-3-ONE OXIME DERIVATIVES, PHARMACEUTICAL COMPOSITIONS COMPRISING THEM, AND PROCESS FOR PREPARING THE SAME
FR2899108B1 (en) 2006-03-31 2012-02-03 Trophos USE OF CHOLEST-4-EN-3-ONE DERIVATIVES FOR THE PRODUCTION OF A CYTOPROTECTIVE MEDICINE
US8007790B2 (en) 2006-04-03 2011-08-30 Stowers Institute For Medical Research Methods for treating polycystic kidney disease (PKD) or other cyst forming diseases
FR2907783A1 (en) 2006-10-30 2008-05-02 Trophos Sa New 4-azacholest-4-ene N-oxide useful to treat or prevent e.g. necrosis, pathological apoptosis and/or necroptosis, diseases of bones, joints, connective tissue and/or cartilage, muscle disease, skin diseases and cardiovascular disease
WO2008086043A2 (en) 2007-01-10 2008-07-17 Wyeth Methods and compositions for assessment and treatment of asthma
JP5239065B2 (en) 2007-04-20 2013-07-17 株式会社大一商会 Game machine
EP2192838A4 (en) 2007-08-15 2011-07-27 Harvard College Heterocyclic inhibitors of necroptosis
FR2934596B1 (en) 2008-07-30 2015-04-10 Trophos NOVEL CHOLEST-4-EN-3-ONE OXIME DERIVATIVES, PHARMACEUTICAL COMPOSITIONS COMPRISING THEM, AND PROCESS FOR PREPARING THE SAME
WO2010075290A1 (en) 2008-12-22 2010-07-01 President And Fellows Of Harvard College Unsaturated heterocyclic inhibitors of necroptosis
US8257921B1 (en) 2008-12-22 2012-09-04 Schering Corporation NRIP1 regulation of apolipoprotein A1
EP3000468A1 (en) 2008-12-23 2016-03-30 President and Fellows of Harvard College Small molecule inhibitors of necroptosis
FR2940650B1 (en) 2008-12-29 2017-01-27 Trophos NOVEL OXIME DERIVATIVES OF 3,5-SECO-4-NOR-CHOLESTANE, PHARMACEUTICAL COMPOSITIONS COMPRISING THE SAME, AND PROCESS FOR PREPARING THE SAME
US7622106B1 (en) 2009-03-06 2009-11-24 Board Of Regents, The University Of Texas System Necrosis assay
KR101155506B1 (en) 2009-07-08 2012-06-15 고려대학교 산학협력단 Screening method of therapeutic and diagnostic agents for tnf-alpha-induced diseases using reactive oxygen species modulator 1
EP2519260A2 (en) 2009-12-31 2012-11-07 Deutsches Krebsforschungszentrum Novel modulators of trail signalling
US20130005726A1 (en) 2010-03-08 2013-01-03 Derek Abbott Compositions and methods for treating inflammatory disorders
AU2011242465B2 (en) 2010-04-23 2017-01-19 Massachusetts Eye And Ear Infirmary Methods and compositions for preserving photoreceptor and retinal pigment epithelial cells
AU2011278998B2 (en) 2010-07-16 2016-06-09 Les Laboratoires Servier Therapeutically active compositions and their method of use
US20120022116A1 (en) 2010-07-20 2012-01-26 Huayun Deng Compositions and methods for the treatment of pathological condition(s) related to gpr35 and/or gpr35-herg complex
US20140024598A1 (en) 2010-11-01 2014-01-23 Demetrios Vavvas Methods and compositions for preserving retinal ganglion cells
US9643977B2 (en) 2011-03-11 2017-05-09 President And Fellows Of Harvard College Necroptosis inhibitors and methods of use therefor
WO2013013826A1 (en) 2011-07-27 2013-01-31 Friedrich-Alexander-Universität Erlangen-Nürnberg Necroptosis inhibitors for the treatment of inflammatory diseases of the gastrointestinal tract
US20140251917A1 (en) 2011-09-22 2014-09-11 Marv Enterprises,LLC Method for the treatment of multiple sclerosis
CN102343102A (en) 2011-10-19 2012-02-08 柳忠辉 Application of activin receptor-interacting protein 2 (ARIP2) gene to preparation of medicament
CA2888805C (en) 2011-10-21 2020-07-14 Massachusetts Eye And Ear Infirmary Methods and compositions for promoting axon regeneration and nerve function
KR101410332B1 (en) 2012-02-29 2014-06-24 코아스템(주) Anti-Oxidant Compositions for Culturing Mesenchymal Stromal Cells and Uses Thereof
ES2446494B1 (en) 2012-03-28 2015-03-16 Consejo Superior De Investigaciones Científicas (Csic) Therapeutic application of necrostatin-1 in steatohepatitis
WO2014022102A1 (en) 2012-08-01 2014-02-06 Amgen Inc. Methods of using anti-apoptotic compounds to modulate one or more properties of a cell culture
JP6362596B2 (en) 2012-08-06 2018-07-25 ブレインストーム セル セラペウティクス リミテッド Method for producing mesenchymal stem cells secreting neurotrophic factor
WO2014102668A2 (en) 2012-12-26 2014-07-03 Koninklijke Philips N.V. Assessment of cellular signaling pathway activity using linear combination(s) of target gene expressions;
WO2014126127A1 (en) 2013-02-13 2014-08-21 国立大学法人北海道大学 Medicinal composition for treating necroptosis-related disease and method for screening active ingredient thereof
TWI638815B (en) 2013-02-15 2018-10-21 英商葛蘭素史克智慧財產發展有限公司 Heterocyclic amides as kinase inhibitors
KR20150126882A (en) 2013-02-27 2015-11-13 더 브로드 인스티튜트, 인코퍼레이티드 T cell balance gene expression, compositions of matters and methods of use thereof
JP2016514693A (en) 2013-03-15 2016-05-23 プレジデント アンド フェローズ オブ ハーバード カレッジ Hybrid necrotosis inhibitor
EP3182974A1 (en) 2014-08-21 2017-06-28 GlaxoSmithKline Intellectual Property Development Limited Heterocyclic amides as rip1 kinase inhibitors as medicaments
KR20180114910A (en) 2016-02-05 2018-10-19 데날리 테라퓨틱스 인크. Inhibitors of receptor-interacting protein kinase 1

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004098589A1 (en) * 2003-05-02 2004-11-18 Elan Pharmaceuticals, Inc. 4- bromo - 5 - (2- chloro - benzoylamino) - 1h - pyrazole - 3 - carboxylic acid amide derivatives and related compounds as bradykinin b1 receptor antagonists for the treatment of inflammatory diseases
WO2007075772A2 (en) * 2005-12-20 2007-07-05 President And Fellows Of Harvard College Compounds, screens, and methods of treatment
WO2008106077A1 (en) 2007-02-28 2008-09-04 Merck & Co., Inc. Substituted benzodiazepinones, benzoxazepinones and benzothiazepinones as sodium channel blockers

Non-Patent Citations (47)

* Cited by examiner, † Cited by third party
Title
"Gaucher Disease", NATURE MEDICINE, 19 January 2014 (2014-01-19)
"Remington's Pharmaceutical Sciences", MACK PUBLISHING COMPANY
"The Handbook of Pharmaceutical Additives", GOWER PUBLISHING LIMITED
"The Handbook of Pharmaceutical Excipients", AMERICAN PHARMACEUTICAL ASSOCIATION AND THE PHARMACEUTICAL PRESS
ARMSTRONG, TETRAHEDRON LETTERS, 1994, pages 3239 - 42
CELL DEATH DIS., vol. 2, 2011, pages E230
CELL DEATH DIS., vol. 2, 2011, pages EL 15
CELL HOST & MICROBE, vol. 15, 2014, pages 23 - 35
CELL, vol. 114, 2003, pages 181 - 190
CELL, vol. 137, 2009, pages 1100 - 1111
CELL, vol. 148, 2012, pages 213 - 227
CELL, vol. 148, 2012, pages 228 - 243
CELL, vol. 153, 2013, pages 1 - 14
CELL, vol. 81, 1995, pages 513 - 523
CURR. BIOL., vol. 9, 1999, pages 539 - 542
DAVID WASILKO; S EDWARD LEE: "TIPS: Titerless Infected Cells Preservation and Scale up", BIOPROCESSING JOURNAL FALL, 2006, pages 29 - 32
DUPREZ, L. ET AL., IMMUNITY, vol. 35, no. 6, 2011, pages 908 - 918
EMBO REP., vol. 10, 2009, pages 916 - 922
FEBS J, vol. 278, 2012, pages 877 - 887
GENES, 2013
HE, S. ET AL., CELL, vol. 137, no. 6, 2009, pages 1100 - 1111
IMMUNITY, vol. 35, 2011, pages 572 - 582
IMMUNITY, vol. 35, 2011, pages 908 - 918
IMMUNITY, vol. 4, 1996, pages 387 - 396
IMMUNITY, vol. 7, 1997, pages 821 - 830
J .VIROL., 2012
J. BIOL. CHEM., vol. 274, 1999, pages 16871 - 16875
J. BIOL. CHEM., vol. 279, 2004, pages 7925 - 7933
J. BIOL. CHEM., vol. 280, 2005, pages 36560 - 36566
J. MOL. BIOL., vol. 396, 2010, pages 528 - 539
J. NEUROSCI. RES., vol. 88, 2010, pages 1569 - 1576
KIDNEY INT., vol. 81, 2012, pages 751 - 761
MOL. CELL, vol. 22, 2006, pages 245 - 257
NAT IMMUNOL., vol. 5, 2004, pages 503 - 507
NAT. CHEM. BIOL., vol. 1, 2005, pages 112 - 119
NAT. IMMUNOL., vol. 9, 2008, pages 1037 - 1046
NAT. REV. IMMUNOL, vol. 8, 2008, pages 279 - 289
NATURE, vol. 477, 2011, pages 330 - 334
NEUROCHEM. RES., vol. 37, 2012, pages 1849 - 1858
PNAS, vol. 107, 2010, pages 21695 - 21700
PROC. NATL. ACAD. SCI. USA., vol. 105, 2008, pages 11778 - 11783
PROC. NATL. ACAD. SCI. USA., vol. 109, 2012, pages 5322 - 5327
PROC. NATL. ACAD. SCI., vol. 109, no. 36, 2012, pages 14598 - 14603
REN. FAIL., vol. 34, 2012, pages 373 - 377
SCI. SIGNAL, vol. 115, 2010, pages RE4
T. GREENE; P. WUTS: "Protecting Groups in Chemical Synthesis", 1999, JOHN WILEY & SONS
TRENDS BIOCHEM. SCI, vol. 30, 2005, pages 151 - 159

Cited By (184)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10292987B2 (en) 2013-02-15 2019-05-21 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US10933070B2 (en) 2013-02-15 2021-03-02 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US9624202B2 (en) 2013-02-15 2017-04-18 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US9556152B2 (en) 2013-02-15 2017-01-31 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US10940154B2 (en) 2013-02-15 2021-03-09 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
CN106573006A (en) * 2014-08-21 2017-04-19 葛兰素史密斯克莱知识产权发展有限公司 Heterocyclic amides as RIP1 kinase inhibitors as medicaments
WO2016027253A1 (en) 2014-08-21 2016-02-25 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as rip1 kinase inhibitors as medicaments
US9499521B2 (en) 2014-12-11 2016-11-22 President And Fellows Of Harvard College Inhibitors of cellular necrosis and related methods
US9944628B2 (en) 2014-12-11 2018-04-17 President And Fellows Of Harvard College Inhibitors of cellular necrosis and related methods
US10508102B2 (en) 2014-12-11 2019-12-17 President And Fellows Of Harvard College Inhibitors of cellular necrosis and related methods
US10287280B2 (en) 2015-02-13 2019-05-14 Glaxosmithkline Intellectual Property Development Limited Crystalline forms of (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][1,4]oxazepin-3-yl)-4H-1,2,4-triazole-3-carboxamide
AU2016217496B2 (en) * 2015-02-13 2019-03-07 Glaxosmithkline Intellectual Property Development Limited Crystalline forms of (S)-5-benzyl-N-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][1,4]oxazepin-3-yl)-4H-1,2,4-triazole-3-carboxamide
JP2018505199A (en) * 2015-02-13 2018-02-22 グラクソスミスクライン、インテレクチュアル、プロパティー、ディベロップメント、リミテッドGlaxosmithkline Intellectual Property Development Limited (S) -5-Benzyl-N- (5-methyl-4-oxo-2,3,4,5 tetrahydrobenzo [b] [1,4] oxazepin-3-yl) -4H-1,2,4 -Crystal form of triazole-3-carboxamide
CN107207485A (en) * 2015-02-13 2017-09-26 葛兰素史密斯克莱知识产权发展有限公司 (S) crystal formation of the formamide of 5 benzyl N (base of 5 methyl, 4 oxo 2,3,4,5 tetrahydro benzo [b] [1,4] oxaza heptantriene 3) 1,2,4 triazoles of 4H 3
WO2016128936A1 (en) 2015-02-13 2016-08-18 Glaxosmithkline Intellectual Property Development Limited Crystalline forms of (s)-5-benzyl-n-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][1,4]oxazepin-3-yl)-4h-1,2,4-triazole-3-carboxamide
CN107889487A (en) * 2015-03-02 2018-04-06 阿派隆生物制剂股份公司 For treating the bicyclic tetrahydro sulphur azepine * derivatives of tumour and/or infectious diseases
CN107889487B (en) * 2015-03-02 2021-09-03 阿派隆生物制剂股份公司 Bicyclic tetrahydrothiazepine derivatives for the treatment of tumor and/or infectious diseases
US10472341B2 (en) 2015-03-02 2019-11-12 Apeiron Biologics Ag Bicyclic tetrahydrothiazepine derivatives useful for the treatment of neoplastic and/or infectious diseases
WO2016139181A1 (en) 2015-03-02 2016-09-09 Apeiron Biologics Ag Bicyclic tetrahydrothiazepine derivatives useful for the treatment of neoplastic and/or infectious diseases
WO2016185423A1 (en) 2015-05-19 2016-11-24 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
CN112370452A (en) * 2015-05-19 2021-02-19 葛兰素史密斯克莱知识产权发展有限公司 Heterocyclic amides as kinase inhibitors
US10899716B2 (en) 2015-05-19 2021-01-26 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US10590085B2 (en) 2015-05-19 2020-03-17 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US11485710B2 (en) 2015-05-19 2022-11-01 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
EA036452B1 (en) * 2015-05-19 2020-11-12 Глэксосмитклайн Интеллекчуал Проперти Дивелопмент Лимитед Heterocyclic amides as kinase inhibitors
JP2020143079A (en) * 2015-05-19 2020-09-10 グラクソスミスクライン、インテレクチュアル、プロパティー、ディベロップメント、リミテッドGlaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
JP2018515555A (en) * 2015-05-19 2018-06-14 グラクソスミスクライン、インテレクチュアル、プロパティー、ディベロップメント、リミテッドGlaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
CN107922348A (en) * 2015-06-22 2018-04-17 大日本住友制药株式会社 Bicyclic heterocycle amide derivatives
US10294237B2 (en) 2015-06-22 2019-05-21 Sumitomo Dainippon Pharma Co., Ltd. Bicyclic heterocyclic amide derivative
WO2016208592A1 (en) * 2015-06-22 2016-12-29 大日本住友製薬株式会社 Bicyclic heterocyclic amide derivative
KR20180023988A (en) * 2015-07-02 2018-03-07 에프. 호프만-라 로슈 아게 Bicyclic lactams and their use
TWI788830B (en) * 2015-07-02 2023-01-01 瑞士商赫孚孟拉羅股份公司 Bicyclic lactams and methods of use thereof
JP2018519319A (en) * 2015-07-02 2018-07-19 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Bicyclic lactam and method of use thereof
WO2017004500A1 (en) * 2015-07-02 2017-01-05 Genentech, Inc. Bicyclic lactams and methods of use thereof
IL274391A (en) * 2015-07-02 2020-06-30 Hoffmann La Roche Bicyclic lactams and methods of use thereof
KR102104911B1 (en) * 2015-07-02 2020-04-28 에프. 호프만-라 로슈 아게 Bicyclic lactam and its use method
RU2716136C2 (en) * 2015-07-02 2020-03-06 Ф. Хоффманн-Ля Рош Аг Bicyclic lactams and methods for use thereof
CN107849023B (en) * 2015-07-02 2021-03-23 豪夫迈·罗氏有限公司 Bicyclic lactams and methods of use thereof
AU2019203444B2 (en) * 2015-07-02 2021-01-21 F. Hoffmann-La Roche Ag Bicyclic lactams and methods of use thereof
JP2021020946A (en) * 2015-07-02 2021-02-18 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Bicyclic lactams and methods for use thereof
JP7190471B2 (en) 2015-07-02 2022-12-15 エフ.ホフマン-ラ ロシュ アーゲー Bicyclic lactam and method of use thereof
KR102603915B1 (en) * 2015-07-02 2023-11-21 에프. 호프만-라 로슈 아게 Bicyclic lactams and methods of use thereof
US10988459B2 (en) 2015-07-02 2021-04-27 Genentech, Inc. Bicyclic lactams and methods of use thereof
TWI763630B (en) * 2015-07-02 2022-05-11 瑞士商赫孚孟拉羅股份公司 Bicyclic lactams and methods of use thereof
EP3760625A1 (en) * 2015-07-02 2021-01-06 F. Hoffmann-La Roche AG Bicyclic lactams as receptor-interacting protein-1 (rip1) kinase inhibitors for treating e.g. inflammatory diseases
CN112979660A (en) * 2015-07-02 2021-06-18 豪夫迈·罗氏有限公司 Bicyclic lactams and methods of use thereof
CN107849023A (en) * 2015-07-02 2018-03-27 豪夫迈·罗氏有限公司 Bicyclic lactam and its application process
KR20190143848A (en) * 2015-07-02 2019-12-31 에프. 호프만-라 로슈 아게 Bicyclic lactams and methods of use thereof
US11952389B2 (en) 2015-07-22 2024-04-09 Enanta Pharmaceuticals, Inc. Benzodiazepine derivatives as RSV inhibitors
US9957281B2 (en) 2015-07-22 2018-05-01 Enanta Pharmaceuticals, Inc. Benzodiazepine derivatives as RSV inhibitors
US10865215B2 (en) 2015-07-22 2020-12-15 Enanta Pharmaceuticals, Inc. Benzodiazepine derivatives as RSV inhibitors
US11390631B1 (en) 2015-07-22 2022-07-19 Enanta Pharmaceuticals, Inc. Benzodiazepine derivatives as RSV inhibitors
CN108431004B (en) * 2015-10-23 2021-02-12 武田药品工业株式会社 Heterocyclic compounds
WO2017069279A1 (en) * 2015-10-23 2017-04-27 武田薬品工業株式会社 Heterocyclic compound
JPWO2017069279A1 (en) * 2015-10-23 2018-08-16 武田薬品工業株式会社 Heterocyclic compounds
AU2016340527B2 (en) * 2015-10-23 2020-09-17 Takeda Pharmaceutical Company Limited Heterocyclic compound
US10787462B2 (en) 2015-10-23 2020-09-29 Takeda Pharmaceutical Company Limited Heterocyclic compound
WO2017096301A1 (en) * 2015-12-04 2017-06-08 Denali Therapeutics Inc. Isoxazolidine derived inhibitors of receptor interacting protein kinase 1 (ripk 1)
CN108602809A (en) * 2015-12-04 2018-09-28 戴纳立制药公司 The inhibitor of receptor interacting protein kinases 1 (RIPK 1) derived from isoxazole alkyl
US10709692B2 (en) 2015-12-04 2020-07-14 Denali Therapeutics Inc. Isoxazolidine derived inhibitors of receptor interacting protein kinase 1 (RIPK1)
CN108602809B (en) * 2015-12-04 2022-09-30 戴纳立制药公司 Isoxazolidine derived inhibitors of receptor interacting protein kinase 1(RIPK 1)
WO2017109724A1 (en) 2015-12-21 2017-06-29 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US10961258B2 (en) 2015-12-21 2021-03-30 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US10759816B2 (en) 2016-01-15 2020-09-01 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as RSV inhibitors
IL287136B2 (en) * 2016-02-05 2023-09-01 Denali Therapeutics Inc Inhibitors of receptor-interacting protein kinase 1
US10604535B2 (en) 2016-02-05 2020-03-31 Denali Therapeutics Inc. Compounds, compositions and methods
US9896458B2 (en) 2016-02-05 2018-02-20 Denali Therapeutics Inc. Compounds, compositions and methods
JP2022017447A (en) * 2016-02-05 2022-01-25 デナリ セラピューティクス インコーポレイテッド Compounds, compositions and methods
US9815850B2 (en) 2016-02-05 2017-11-14 Denali Therapeutics Inc. Compounds, compositions and methods
US10131676B2 (en) 2016-02-05 2018-11-20 Denali Therapeutics Inc. Compounds, compositions and methods
JP2019508407A (en) * 2016-02-05 2019-03-28 デナリ セラピューティクス インコーポレイテッドDenali Therapeutics Inc. Compound, composition and method
JP7381543B2 (en) 2016-02-05 2023-11-15 デナリ セラピューティクス インコーポレイテッド Compounds, compositions and methods
IL287136B1 (en) * 2016-02-05 2023-05-01 Denali Therapeutics Inc Inhibitors of receptor-interacting protein kinase 1
US10898469B2 (en) 2016-02-26 2021-01-26 Sumitomo Dainippon Pharma Co., Ltd. Imidazolylamide derivative
WO2018007973A2 (en) 2016-07-06 2018-01-11 Glaxosmithkline Intellectual Property Development Limited Process and intermediates for preparing benzoxazepines
WO2018007973A3 (en) * 2016-07-06 2018-02-15 Glaxosmithkline Intellectual Property Development Limited Process and intermediates for preparing benzoxazepines
CN109843886A (en) * 2016-10-17 2019-06-04 豪夫迈·罗氏有限公司 Two ring pyridone lactams and its application method
CN109843886B (en) * 2016-10-17 2022-04-19 豪夫迈·罗氏有限公司 Bicyclic pyridone lactams and methods of use thereof
WO2018073193A1 (en) 2016-10-17 2018-04-26 F. Hoffmann-La Roche Ag Bicyclic pyridone lactams and methods of use thereof
US10947226B2 (en) 2016-10-17 2021-03-16 Genentech, Inc. Bicyclic pyridone lactams and methods of use thereof
WO2018083288A1 (en) 2016-11-07 2018-05-11 Bayer Aktiengesellschaft Substituted sulfonyl amides for controlling animal pests
WO2018092089A1 (en) 2016-11-18 2018-05-24 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US11607396B2 (en) 2016-12-02 2023-03-21 Genentech, Inc. Bicyclic amide compounds and methods of use thereof
US11071721B2 (en) 2016-12-02 2021-07-27 Genentech, Inc. Bicyclic amide compounds and methods of use thereof
WO2018100070A1 (en) 2016-12-02 2018-06-07 F. Hoffmann-La Roche Ag Bicyclic amide compounds and methods of use thereof
US11072618B2 (en) 2016-12-09 2021-07-27 Denali Therapeutics Inc. Compounds, compositions and methods
JP7209625B2 (en) 2016-12-16 2023-01-20 エフ.ホフマン-ラ ロシュ アーゲー Inhibitor of RIP1 kinase and method of use thereof
EP4095138A1 (en) 2016-12-16 2022-11-30 F. Hoffmann-La Roche AG Inhibitors of rip1 kinase and methods of use thereof
US11072607B2 (en) 2016-12-16 2021-07-27 Genentech, Inc. Inhibitors of RIP1 kinase and methods of use thereof
JP2020502146A (en) * 2016-12-16 2020-01-23 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft RIP1 kinase inhibitors and methods of use
WO2018109097A1 (en) 2016-12-16 2018-06-21 F. Hoffmann-La Roche Ag Inhibitors of rip1 kinase and methods of use thereof
WO2018117196A1 (en) 2016-12-20 2018-06-28 大日本住友製薬株式会社 Drug targeting cancer stem cell
US10398706B2 (en) 2017-01-06 2019-09-03 Enanta Pharmaceuticals, Inc. Heteroaryldiazepine derivatives as RSV inhibitors
US10906895B2 (en) 2017-02-16 2021-02-02 Enanta Pharmaceuticals, Inc. Processes for the preparation of benzodiazepine derivatives
US10358441B2 (en) 2017-02-16 2019-07-23 Enanta Pharmaceuticals, Inc. Processes for the preparation of benzodiazepine derivatives
CN110573504A (en) * 2017-02-27 2019-12-13 葛兰素史克知识产权开发有限公司 heterocyclic amides as kinase inhibitors
WO2018154520A1 (en) 2017-02-27 2018-08-30 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
TWI790227B (en) * 2017-04-14 2023-01-21 美商百健Ma公司 Inhibiting agents for bruton’s tyrosine kinase
IL270648B1 (en) * 2017-05-17 2023-11-01 Denali Therapeutics Inc Kinase inhibitors and uses thereof
CN110913858A (en) * 2017-05-17 2020-03-24 戴纳立制药公司 Kinase inhibitors and uses thereof
US11203600B2 (en) 2017-05-17 2021-12-21 Denali Therapeutics Inc. Kinase inhibitors and uses thereof
EP3624797A4 (en) * 2017-05-17 2021-07-21 Denali Therapeutics Inc. Kinase inhibitors and uses thereof
IL270648B2 (en) * 2017-05-17 2024-03-01 Denali Therapeutics Inc Kinase inhibitors and uses thereof
US10752598B2 (en) 2017-06-07 2020-08-25 Enanta Pharmaceuticals, Inc. Aryldiazepine derivatives as RSV inhibitors
US10851115B2 (en) 2017-06-30 2020-12-01 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as RSV inhibitors
US11091501B2 (en) 2017-06-30 2021-08-17 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as RSV inhibitors
WO2019012063A1 (en) 2017-07-14 2019-01-17 F. Hoffmann-La Roche Ag Bicyclic ketone compounds and methods of use thereof
US11834461B2 (en) 2017-07-14 2023-12-05 Genentech, Inc. Bicyclic ketone compounds and methods of use thereof
RU2797922C2 (en) * 2017-07-14 2023-06-13 Ф. Хоффманн-Ля Рош Аг Bicyclic ketones and methods of their use
US11098058B2 (en) 2017-07-14 2021-08-24 Genentech, Inc. Bicyclic ketone compounds and methods of use thereof
US10881666B2 (en) 2017-09-29 2021-01-05 Enanta Pharmaceuticals, Inc. Combination pharmaceutical agents as RSV inhibitors
WO2019072942A1 (en) 2017-10-11 2019-04-18 F. Hoffmann-La Roche Ag Bicyclic compounds for use as rip1 kinase inhibitors
US11673892B2 (en) 2017-10-11 2023-06-13 Genentech, Inc. Bicyclic compounds for use as RIP1 kinase inhibitors
WO2019086494A1 (en) 2017-10-31 2019-05-09 F. Hoffmann-La Roche Ag Bicyclic sulfones and sulfoxides and methods of use thereof
US11072617B2 (en) 2017-10-31 2021-07-27 Genentech, Inc. Bicyclic sulfones and sulfoxides and methods of use thereof
US10501422B2 (en) 2017-11-13 2019-12-10 Enanta Pharmaceuticals, Inc. Processes for the resolution of benzodiazepin-2-one and benzoazepin-2-one derivatives
US10647711B2 (en) 2017-11-13 2020-05-12 Enanta Pharmaceuticals, Inc. Azepin-2-one derivatives as RSV inhibitors
WO2019123219A1 (en) 2017-12-20 2019-06-27 Glaxosmithkline Intellectual Property Development Limited Crystalline salt form of (s)-5-benzyl-n-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][1,4]oxazepin-3-yl)-4h-1,2,4-triazole-3-carboxamide
WO2019130230A1 (en) 2017-12-29 2019-07-04 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors
US10975094B2 (en) 2018-04-11 2021-04-13 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as RSV inhibitors
WO2019204537A1 (en) 2018-04-20 2019-10-24 Genentech, Inc. N-[4-oxo-2,3-dihydro-1,5-benzoxazepin-3-yl]-5,6-dihydro-4h-pyrrolo[1,2-b]pyrazol e-2-carboxamide derivatives and related compounds as rip1 kinase inhibitors for treating e.g. irritable bowel syndrome (ibs)
US11634436B2 (en) 2018-04-20 2023-04-25 Genentech, Inc. Pyridine lactam compounds and methods of use thereof
IL278417B1 (en) * 2018-05-03 2024-01-01 Rigel Pharmaceuticals Inc Rip1 inhibitory compounds and methods for making and using the same
IL278416B1 (en) * 2018-05-03 2023-08-01 Rigel Pharmaceuticals Inc Rip1 inhibitory compounds and methods for making and using the same
US10815206B2 (en) 2018-05-03 2020-10-27 Rigel Pharmaceuticals, Inc. RIP1 inhibitory compounds and methods for making and using the same
WO2019213447A1 (en) 2018-05-03 2019-11-07 Rigel Pharmaceuticals, Inc. Rip1 inhibitory compounds and methods for making and using the same
US11332451B2 (en) 2018-05-03 2022-05-17 Rigel Pharmaceuticals, Inc. RIP1 inhibitory compounds and methods for making and using the same
US11370765B2 (en) 2018-05-03 2022-06-28 Rigel Pharmaceuticals, Inc. RIP1 inhibitory compounds and methods for making and using the same
US11370764B2 (en) 2018-05-03 2022-06-28 Rigel Pharmaceuticals Inc. RIP1 inhibitory compounds and methods for making and using the same
US11377428B2 (en) 2018-05-03 2022-07-05 Rigel Pharmaceuticals, Inc. RIP1 inhibitory compounds and methods for making and using the same
US10975064B2 (en) 2018-05-03 2021-04-13 Rigel Pharmaceuticals, Inc. RIP1 inhibitory compounds and methods for making and using the same
JP2021523226A (en) * 2018-05-03 2021-09-02 ライジェル ファーマシューティカルズ, インコーポレイテッド RIP1 inhibitor compound and methods for producing and using it
EP4248975A2 (en) 2018-05-03 2023-09-27 Rigel Pharmaceuticals, Inc. Rip1 inhibitory compounds and methods for making and using the same
EP4248975A3 (en) * 2018-05-03 2024-03-13 Rigel Pharmaceuticals, Inc. Rip1 inhibitory compounds and methods for making and using the same
EP3788044B1 (en) * 2018-05-03 2023-08-09 Rigel Pharmaceuticals, Inc. Rip1 inhibitory compounds and methods for making and using the same
WO2019213445A1 (en) 2018-05-03 2019-11-07 Rigel Pharmaceuticals, Inc. Rip1 inhibitory compounds and methods for making and using the same
US11472782B2 (en) 2018-05-03 2022-10-18 Rigel Pharmaceuticals, Inc. RIP1 inhibitory compounds and methods for making and using the same
US11919890B2 (en) 2018-05-03 2024-03-05 ;Eli Lilly and Company RIP1 inhibitory compounds and methods for making and using the same
AU2019262144B2 (en) * 2018-05-03 2023-11-16 Rigel Pharmaceuticals, Inc. RIP1 inhibitory compounds and methods for making and using the same
WO2019224774A1 (en) 2018-05-23 2019-11-28 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as rip1 kinase inhibitors
WO2019224773A1 (en) 2018-05-23 2019-11-28 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as rip1 kinase inhibitors
EP3816163A4 (en) * 2018-06-26 2022-04-13 Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences Cell necrosis inhibitor, preparation method therefor and use thereof
WO2020044206A1 (en) 2018-08-29 2020-03-05 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides as kinase inhibitors for use in the treatment cancer
WO2020088194A1 (en) 2018-11-02 2020-05-07 中国科学院上海药物研究所 Heterocyclic amide for inhibiting rip1 kinase and uses thereof
EP3872077A4 (en) * 2018-11-02 2021-12-01 Shanghai Institute of Materia Medica, Chinese Academy of Sciences Heterocyclic amide for inhibiting rip1 kinase and uses thereof
WO2020146615A1 (en) 2019-01-11 2020-07-16 Genentech, Inc. Bicyclic pyrrolotriazolr ketone compounds and methods of use thereof
TWI724753B (en) * 2019-01-25 2021-04-11 大陸商北京賽特明強醫藥科技有限公司 Amido bridged heterocyclic compound, and composition and application thereof
WO2020151589A1 (en) 2019-01-25 2020-07-30 北京赛特明强医药科技有限公司 Acylamino bridged heterocyclic compound, and composition and application thereof
AU2020210455B2 (en) * 2019-01-25 2023-02-02 Beijing Scitech-Mq Pharmaceuticals Limited Acylamino bridged heterocyclic compound, and composition and application thereof
CN113166116A (en) * 2019-01-25 2021-07-23 北京赛特明强医药科技有限公司 Amido bridged heterocyclic compound, and composition and application thereof
US11254664B2 (en) 2019-03-18 2022-02-22 Enanta Pharmaceuticals, Inc. Benzodiazepine derivatives as RSV inhibitors
US11912695B2 (en) 2019-03-18 2024-02-27 Enanta Pharmaceuticals, Inc. Benzodiazepine derivatives as RSV inhibitors
US11179400B2 (en) 2019-04-09 2021-11-23 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as RSV inhibitors
WO2020234716A1 (en) 2019-05-17 2020-11-26 Glaxosmithkline Intellectual Property Development Limited Matrix composition comprising (s)-5-benzyl-n-(5-methyl-4-oxo-2,3,4,5-tetrahydrobenzo[b][1,4]oxazepin-3-yl)-4h-1,2,4-triazole-3-carboxamide
US11564930B2 (en) 2019-09-06 2023-01-31 Rigel Pharmaceuticals, Inc. RIP1 inhibitory compounds and methods for making and using the same
WO2021046437A1 (en) 2019-09-06 2021-03-11 Rigel Pharmaceuticals, Inc. Rip1 inhibitory compounds and methods for making and using the same
WO2021046447A1 (en) * 2019-09-06 2021-03-11 Rigel Pharmaceuticals, Inc. Heterocyclic rip1 kinase inhibitors
US11479543B2 (en) 2019-09-06 2022-10-25 Rigel Pharmaceuticals, Inc. Heterocyclic RIP1 kinase inhibitors
AU2020343671B2 (en) * 2019-09-06 2023-11-30 Rigel Pharmaceuticals, Inc. RIP1 inhibitory compounds and methods for making and using the same
US11718612B2 (en) 2019-09-06 2023-08-08 Board Of Regents, The University Of Texas System Inhibitors of receptor interacting protein kinase I for the treatment of disease
WO2021046382A1 (en) 2019-09-06 2021-03-11 Rigel Pharmaceuticals, Inc. Rip1 inhibitory compounds and methods for making and using the same
US11407736B2 (en) 2019-09-06 2022-08-09 Rigel Pharmaceuticals, Inc. RIP1 inhibitory compounds and methods for making and using the same
WO2021046407A1 (en) 2019-09-06 2021-03-11 Rigel Pharmaceuticals, Inc. Rip1 inhibitory compounds and methods for making and using the same
US11753381B2 (en) 2019-09-27 2023-09-12 Board Of Regents, The University Of Texas System Inhibitors of receptor interacting protein kinase I for the treatment of disease
US11572367B2 (en) 2019-10-04 2023-02-07 Enanta Pharmaceuticals, Inc. Antiviral heterocyclic compounds
US11505558B1 (en) 2019-10-04 2022-11-22 Enanta Pharmaceuticals, Inc. Antiviral heterocyclic compounds
US11578078B2 (en) 2019-11-07 2023-02-14 Rigel Pharmaceuticals, Inc. Heterocyclic RIP1 inhibitory compounds
WO2021092336A1 (en) 2019-11-07 2021-05-14 Rigel Pharmaceuticals, Inc. Heterocyclic rip1 inhibitory compounds
US11690850B2 (en) 2019-11-26 2023-07-04 Board Of Regents, The University Of Texas System Inhibitors of receptor interacting protein kinase I for the treatment of disease
US11420976B2 (en) 2020-01-24 2022-08-23 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as anti-viral agents
EP4110765A4 (en) * 2020-02-28 2024-03-06 Univ Texas Inhibitors of receptor interacting protein kinase i for the treatment of disease
US11584758B2 (en) 2020-04-02 2023-02-21 Rigel Pharmaceuticals, Inc. RIP1K inhibitors
WO2021245070A1 (en) 2020-06-02 2021-12-09 Sanofi Isoxazolidines as ripk1 inhibitors and use thereof
US11667643B2 (en) 2020-07-01 2023-06-06 Rigel Pharmaceuticals, Inc. RIP1K inhibitors
US11534439B2 (en) 2020-07-07 2022-12-27 Enanta Pharmaceuticals, Inc. Dihydroquinoxaline and dihydropyridopyrazine derivatives as RSV inhibitors
US11945824B2 (en) 2020-10-19 2024-04-02 Enanta Pharmaceuticals, Inc. Heterocyclic compounds as anti-viral agents
WO2022192533A1 (en) * 2021-03-11 2022-09-15 Rigel Pharmaceuticals, Inc. Heterocyclic rip1 kinase inhibitors
WO2022212809A1 (en) 2021-04-02 2022-10-06 Genentech, Inc. Processes for making bicyclic ketone compounds
US11767310B2 (en) 2021-08-10 2023-09-26 Abbvie Inc. Nicotinamide RIPK1 inhibitors
WO2023068881A1 (en) * 2021-10-22 2023-04-27 주식회사 보로노이바이오 Aryl or heteroaryl derivative, and pharmaceutical composition comprising same as active ingredient for prevention or treatment of kinase-related diseases
US11897876B2 (en) 2021-11-11 2024-02-13 Genzyme Corporation Isoxazolidines as RIPK1 inhibitors and use thereof
WO2023083847A1 (en) 2021-11-11 2023-05-19 Sanofi Isoxazolidines as ripk1 inhibitors and use thereof
WO2024040155A1 (en) 2022-08-19 2024-02-22 Genzyme Corporation Isoxazolidines as ripk1 inhibitors and use thereof
WO2024044183A1 (en) * 2022-08-22 2024-02-29 Enanta Pharmaceuticals, Inc. Fused heterobicyclic antiviral agents

Also Published As

Publication number Publication date
US20190201413A1 (en) 2019-07-04
CN106928203A (en) 2017-07-07
DOP2015000196A (en) 2016-05-15
CR20190511A (en) 2020-01-08
CL2015002279A1 (en) 2016-07-08
ZA201505267B (en) 2017-08-30
HRP20180900T1 (en) 2018-07-13
EP3508484B1 (en) 2021-04-07
EA031971B1 (en) 2019-03-29
DK2956452T3 (en) 2018-06-06
MY195183A (en) 2023-01-11
HK1249100A1 (en) 2018-10-26
EP2956452A1 (en) 2015-12-23
JP2016512488A (en) 2016-04-28
NZ628447A (en) 2016-08-26
US20150353533A1 (en) 2015-12-10
IL256502A (en) 2018-02-28
BR112015019627A2 (en) 2017-07-18
HUE053215T2 (en) 2021-06-28
EP3375780A1 (en) 2018-09-19
KR102267977B1 (en) 2021-06-21
PH12015501675A1 (en) 2015-10-19
EA201591506A1 (en) 2016-01-29
EA028991B1 (en) 2018-01-31
TW201819369A (en) 2018-06-01
BR122017002949A2 (en) 2019-09-10
JP2018111712A (en) 2018-07-19
CN106928203B (en) 2020-02-07
PT2956452T (en) 2018-06-01
HUE037763T2 (en) 2018-09-28
RS61439B1 (en) 2021-03-31
ES2855135T3 (en) 2021-09-23
EP3342771B1 (en) 2020-11-25
EP3375780B1 (en) 2020-06-03
TW201819368A (en) 2018-06-01
DOP2018000088A (en) 2018-10-31
SG11201505796TA (en) 2015-08-28
TW201819370A (en) 2018-06-01
US10933070B2 (en) 2021-03-02
EP3342771A1 (en) 2018-07-04
TW201443042A (en) 2014-11-16
US9624202B2 (en) 2017-04-18
PT3342771T (en) 2021-02-11
ES2672530T3 (en) 2018-06-14
IL256500B (en) 2018-11-29
PL2956452T3 (en) 2018-08-31
PL3342771T3 (en) 2021-06-14
MA44820A1 (en) 2019-09-30
KR20150119174A (en) 2015-10-23
PH12018500341A1 (en) 2018-09-10
PE20151752A1 (en) 2015-11-25
BR122017002946A2 (en) 2019-09-10
CY1120320T1 (en) 2019-07-10
PH12018500342A1 (en) 2018-09-10
KR20190018558A (en) 2019-02-22
TWI637951B (en) 2018-10-11
TWI638815B (en) 2018-10-21
US20180098998A1 (en) 2018-04-12
AU2014217453A1 (en) 2015-08-13
US10940154B2 (en) 2021-03-09
JP2018150372A (en) 2018-09-27
EA201792039A1 (en) 2018-01-31
CA2900695C (en) 2021-10-12
US20170008878A1 (en) 2017-01-12
CN105121432A (en) 2015-12-02
MX2015010572A (en) 2015-11-16
MX360715B (en) 2018-11-13
CN108774214A (en) 2018-11-09
BR122017002951A2 (en) 2020-01-28
TWI648273B (en) 2019-01-21
RS57332B1 (en) 2018-08-31
DOP2018000089A (en) 2018-06-30
EP3508484A1 (en) 2019-07-10
UY35330A (en) 2014-08-29
AU2014217453B2 (en) 2016-04-14
BR112015019627B1 (en) 2022-08-30
TWI648274B (en) 2019-01-21
DK3342771T3 (en) 2021-02-08
UA118259C2 (en) 2018-12-26
CN108774214B (en) 2021-03-09
US20190262356A1 (en) 2019-08-29
HRP20210252T1 (en) 2021-04-02
US9556152B2 (en) 2017-01-31
IL256500A (en) 2018-02-28
CR20150420A (en) 2015-10-19
SI2956452T1 (en) 2018-06-29
MA44820B1 (en) 2020-06-30
CY1123915T1 (en) 2022-05-27
IL240280A0 (en) 2015-09-24
PH12018500343A1 (en) 2018-09-10
CR20190512A (en) 2020-01-08
LT3342771T (en) 2021-03-10
HK1213545A1 (en) 2016-07-08
EP2956452B1 (en) 2018-03-21
TR201808112T4 (en) 2018-07-23
US10292987B2 (en) 2019-05-21
SI3342771T1 (en) 2021-04-30
KR102228764B1 (en) 2021-03-18
ES2814556T3 (en) 2021-03-29
LT2956452T (en) 2018-06-11
JP6352571B1 (en) 2018-07-04
HK1253099A1 (en) 2019-06-06
IL240280B (en) 2018-04-30
CA2900695A1 (en) 2014-08-21
JP6321045B2 (en) 2018-05-09
US20170183332A1 (en) 2017-06-29
CN105121432B (en) 2018-08-03
IL256502B (en) 2018-11-29
MY178552A (en) 2020-10-16

Similar Documents

Publication Publication Date Title
US10940154B2 (en) Heterocyclic amides as kinase inhibitors
AU2013315387B2 (en) Amino-quinolines as kinase inhibitors

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201480021120.8

Country of ref document: CN

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

Ref document number: 14705891

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 14763183

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 240280

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 38326

Country of ref document: MA

ENP Entry into the national phase

Ref document number: 2900695

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 001727-2015

Country of ref document: PE

ENP Entry into the national phase

Ref document number: 2014217453

Country of ref document: AU

Date of ref document: 20140214

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2015557554

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: IDP00201505013

Country of ref document: ID

Ref document number: CR2015-000420

Country of ref document: CR

Ref document number: MX/A/2015/010572

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 15196058

Country of ref document: CO

Ref document number: NC2017/0011320

Country of ref document: CO

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015019627

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 2014705891

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20157024819

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: A201508769

Country of ref document: UA

WWE Wipo information: entry into national phase

Ref document number: 201591506

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 112015019627

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150814

WWE Wipo information: entry into national phase

Ref document number: 12018500342

Country of ref document: PH

Ref document number: 12018500341

Country of ref document: PH

Ref document number: 12018500343

Country of ref document: PH