WO2015130854A1 - Compounds for treatment of complement mediated disorders - Google Patents

Compounds for treatment of complement mediated disorders Download PDF

Info

Publication number
WO2015130854A1
WO2015130854A1 PCT/US2015/017609 US2015017609W WO2015130854A1 WO 2015130854 A1 WO2015130854 A1 WO 2015130854A1 US 2015017609 W US2015017609 W US 2015017609W WO 2015130854 A1 WO2015130854 A1 WO 2015130854A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
independently chosen
cycloalkyl
halogen
hydroxyl
Prior art date
Application number
PCT/US2015/017609
Other languages
French (fr)
Inventor
Avinash S. Phadke
Xiangzhu Wang
Dawei Chen
Akihiro Hashimoto
Venkat Rao GADHACHANDA
Godwin Pais
Qiuping Wang
Atul Agarwal
Milind Deshpande
Jason Allan Wiles
Original Assignee
Achillion Pharmaceuticals, Inc.
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
Priority to EP15755573.1A priority Critical patent/EP3110805B1/en
Priority to KR1020167026303A priority patent/KR102366025B1/en
Priority to CA2940777A priority patent/CA2940777A1/en
Priority to AP2016009437A priority patent/AP2016009437A0/en
Application filed by Achillion Pharmaceuticals, Inc. filed Critical Achillion Pharmaceuticals, Inc.
Priority to AU2015223084A priority patent/AU2015223084B2/en
Priority to EA201691725A priority patent/EA035501B1/en
Priority to RU2016137832A priority patent/RU2703995C2/en
Priority to CN201580010598.5A priority patent/CN106458981B/en
Priority to MX2016011038A priority patent/MX2016011038A/en
Priority to JP2016570944A priority patent/JP6537532B2/en
Publication of WO2015130854A1 publication Critical patent/WO2015130854A1/en
Priority to ZA2016/05832A priority patent/ZA201605832B/en
Priority to IL247452A priority patent/IL247452B/en
Priority to US15/246,049 priority patent/US20160361329A1/en
Priority to SA516371744A priority patent/SA516371744B1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/572Five-membered rings
    • C07F9/5728Five-membered rings condensed with carbocyclic rings or carbocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/401Proline; Derivatives thereof, e.g. captopril
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/4045Indole-alkylamines; Amides thereof, e.g. serotonin, melatonin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41551,2-Diazoles non condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41621,2-Diazoles condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41881,3-Diazoles condensed with other heterocyclic ring systems, e.g. biotin, sorbinil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41921,2,3-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/501Pyridazines; Hydrogenated pyridazines not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/536Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines ortho- or peri-condensed with carbocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/549Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame having two or more nitrogen atoms in the same ring, e.g. hydrochlorothiazide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/683Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • 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
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • 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
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/12Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/14Radicals substituted by nitrogen atoms, not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/30Indoles; Hydrogenated indoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/30Indoles; Hydrogenated indoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to carbon atoms of the hetero ring
    • C07D209/40Nitrogen atoms, not forming part of a nitro radical, e.g. isatin semicarbazone
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/30Indoles; Hydrogenated indoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to carbon atoms of the hetero ring
    • C07D209/42Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/44Iso-indoles; Hydrogenated iso-indoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/80[b, c]- or [b, d]-condensed
    • C07D209/82Carbazoles; Hydrogenated carbazoles
    • C07D209/88Carbazoles; Hydrogenated carbazoles 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 carbon atoms of the ring system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • 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
    • C07D403/08Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing alicyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D487/12Heterocyclic 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 three hetero rings
    • C07D487/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/113Spiro-condensed systems with two or more oxygen atoms as ring hetero atoms in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic System
    • C07F5/02Boron compounds
    • C07F5/025Boronic and borinic acid compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic System
    • C07F5/02Boron compounds
    • C07F5/027Organoboranes and organoborohydrides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic System
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/0803Compounds with Si-C or Si-Si linkages
    • C07F7/081Compounds with Si-C or Si-Si linkages comprising at least one atom selected from the elements N, O, halogen, S, Se or Te
    • C07F7/0812Compounds with Si-C or Si-Si linkages comprising at least one atom selected from the elements N, O, halogen, S, Se or Te comprising a heterocyclic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65583Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system each of the hetero rings containing nitrogen as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • C07F9/65616Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings containing the ring system having three or more than three double bonds between ring members or between ring members and non-ring members, e.g. purine or analogs
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Definitions

  • the complement system is a part of the innate immune system which does not adapt to changes over the course of the host's life, but is recruited and used by the adaptive immune system. For example, it assists, or complements, the ability of antibodies and phagocytic cells to clear pathogens.
  • This sophisticated regulatory pathway allows rapid reaction to pathogenic organisms while protecting host cells from destruction.
  • Over thirty proteins and protein fragments make up the complement system. These proteins act through opsonization (enhancing phaogytosis of antigens), chemotaxis (attracting macrophages and neutrophils), cell lysis (rupturing membranes of foreign cells) and agglutination (clustering and binding of pathogens together).
  • Complement factor D plays an early and central role in activation of the alternative pathway of the complement cascade. Activation of the alternative complement pathway is initiated by spontaneous hydrolysis of a thioester bond within C3 to produce C3(H 2 0), which associates with factor B to form the C3(H 2 0)B complex.
  • Complement factor D acts to cleave factor B within the C3(H 2 0)B complex to form Ba and Bb.
  • the Bb fragment remains associated with C3(H 2 0) to form the alternative pathway C3 convertase C3(H 2 0)Bb.
  • C3b generated by any of the C3 convertases also associates with factor B to form C3bB, which factor D cleaves to generate the later stage alternative pathway C3 convertase C3bBb.
  • This latter form of the alternative pathway C3 convertase may provide important downstream amplification within all three of the defined complement pathways, leading ultimately to the recruitment and assembly of additional factors in the complement cascade pathway, including the cleavage of C5 to C5a and C5b.
  • C5b acts in the assembly of factors C6, C7, C8, and C9 into the membrane attack complex, which can destroy pathogenic cells by lysing the cell.
  • complement pathway contributes to the production of C3a and C5a, both potent anaphylatoxins, which also have roles in a number of inflammatory disorders. Therefore, in some instances, it is desirable to decrease the response of the complement pathway, including the alternative complement pathway.
  • disorders mediated by the complement pathway include age-related macular degeneration (AMD), paroxysmal nocturnal hemoglobinuria (PNH), multiple sclerosis, and rheumatoid arthritis.
  • ASD Age-related macular degeneration
  • Paroxysmal nocturnal hemoglobinuria is a non-malignant, hematological disorder characterized by the expansion of hematopoietic stem cells and progeny mature blood cells which are deficient in some surface proteins. PNH erythrocytes are not capable of modulating their surface complement activation, which leads to the typical hallmark of PNH - the chronic activation of complement mediated intravascular anemia.
  • the anti-C5 monoclonal antibody eculizumab has been approved in the U.S. for treatment of PNH.
  • many of the patients treated with eculizumab remain anemic, and many patients continue to require blood transfusions.
  • factor D inhibitors While initial attempts have been made to develop inhibitors of factor D, there are currently no small molecule factor D inhibitors in clinical trials. Examples of factor D inhibitors or prolyl compounds are described in the following disclosures.
  • Novartis PCT patent publications WO2014/002057 titled “Pyrrolidine derivatives and their use as complement pathway modulators” and WO2014/009833 titled “Complement pathway modulators and uses thereof describe additional factor D inhibitors with heterocyclic substituents. Additional factor D inhibitors are described in Novartis PCT patent publications WO2014/002051, WO2014/002052, WO2014/002053, WO2014/002054, WO2014/002058, WO2014/002059, and WO2014/005150.
  • PCT patent publication WO 1993/020099 titled "CCK and/or gastrin receptor ligands” describes compounds with a proline-like core and heterocyclic substituents connected to the proline core through amide linkages for the treatment of, for example, gastric disorders or pain.
  • Q 3 is N(R 3 ), S, or C(R 3 R 3' ).
  • Q 1 , Q 2 , Q 3 , X 1 , and X 2 are selected such that a stable compound results.
  • R and R' are independently chosen from H, alkyl, cycloalkyl, alkylcycloalkyl, cycloalkylalkyl, heterocycle, heterocycloalkyl, aryl, aralkyl, heteroaryl, heteroarylalkyl wherein each group can be optionally substituted or any other substituent group herein that provides the desired properties.
  • the ring includes one or more chiral carbon atoms.
  • the invention includes embodiments in which the chiral carbon can be provided as an enantiomer, or mixtrues of enantiomers, including a racemic mixture. Where the ring includes more than one stereocenter, all of the enantiomers and diastereomers are included in the invention as individual species.
  • Z is F, CI, NH 2 , CH 3 , CH 2 D, CHD 2 , or CD 3 .
  • R , R , R , R , and R are independently chosen at each occurence from (c) and (d):
  • any one of the following rings (e), (f), (g), (h), (i), or j) may be present: (e) R 1 and R 1 or R 3 and R 3 may be taken together to form a 3- to 6-membered carbocyclic spiro ring or a 3- to 6-membered heterocyclic spiro ring containing 1 or 2 heteroatoms independently chosen from N, O, or S;
  • R 2 and R 2 may be taken together to form a 3- to 6-membered carbocyclic spiro ring
  • R 2 and R 2 may be taken together to form a 3- to 6-membered heterocyclic spiro ring, each of which spiro rings (e), (f), and (g) is unsubstituted or substituted with 1 or more substituents independently chosen from halogen (and in particular F), hydroxyl, cyano, -COOH, Ci-C 4 alkyl (including in particular methyl), C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, Ci-C 4 alkoxy, C 2 - C 4 alkanoyl, hydroxyCi-C 4 alkyl, (mono- and di-Ci-C 4 alkylamino)Co-C 4 alkyl, -Co-C 4 alkyl(C 3 - C 7 cycloalkyl), -0-Co-C 4 alkyl(C 3 -C 7 cycloalkyl), Ci-C 2 haloalkyl, and Ci-C 2 haloalkoxy
  • R 1 and R 2 may be taken together to form a 3-membered carbocyclic ring
  • R 1 and R 2 may be taken together to form a 4- to 6-membered carbocyclic ring or a 4- to 6-membered heterocyclic ring containing 1 or 2 heteroatoms independently chosen from N, O, and S.
  • R 2 and R 3 if bound to adjacent carbon atoms, may be taken together to form a 3- to 6- membered carbocyclic ring or a 3- to 6-membered heterocyclic ring; each of which ring (h), (i), and (j) may be unsubstituted or substituted with 1 or more substituents independently chosen from halogen (and in particular F), hydroxyl, cyano, -COOH, Ci-C 4 alkyl (including in particular methyl), C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, Ci-C 4 alkoxy, C 2 -C 4 alkanoyl, hydroxyCi-C 4 alkyl, (mono- and di-Ci-C 4 alkylamino)Co-C 4 alkyl, -Co-C 4 alkyl(C 3 -C 7 cycloalkyl), -0-Co-C 4 alkyl(C 3 - C 7 cycloalkyl), Ci
  • R and R , R and R , or R and R can be taken together to form a carbonyl group.
  • R 1 and R 2 or R 2 and R 3 can be taken together to form a carbon-carbon double bond.
  • A is a group chosen from (k) and (1) where (k) is
  • X 4 is B(OH) and Y is CHR 9 ; or X 4 is CHR 9 and Y is B(OH).
  • R 101 is hydrogen, alkyl, carboxy.
  • R 4 is (m) or (n):
  • each of which R 4 other than hydrogen, -CHO, and -CONH 2 is unsubstituted or substituted with one or more of amino, imino, halogen, hydroxyl, cyano, cyanoimino, Ci-C 2 alkyl, Ci-C 2 alkoxy, -Co-C 2 alkyl(mono- and di-Ci-C 4 alkylamino), Ci-C 2 haloalkyl, and Ci-C 2 haloalkoxy.
  • R 5 and R 6 other than hydrogen, hydroxyl, cyano, and -COOH is unsubstituted or optionally substituted.
  • R 5 and R 6 other than hydrogen, hydroxyl, cyano, and -COOH may be substituted with one or more substituents independently chosen from halogen, hydroxyl, amino, imino, cyano, cyanoimino, Ci-C 2 alkyl, Ci-C 4 alkoxy, -Co- C 2 alkyl(mono- and di-Ci-C 4 alkylamino), Ci-C 2 haloalkyl, Ci-C 2 haloalkoxy, C(0)alkyl, C(0)cycloalkyl, C(0)aryl, C(0)heterocycle, and C(0)heteroaryl.
  • R 6 is hydrogen, halogen, hydroxyl, Ci-C 4 alkyl, or Ci-C 4 alkoxy; or R 6 and R 6 may be taken together to form an oxo, vinyl, or imino group.
  • R 7 is hydrogen, Ci-C 6 alkyl, or -C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl).
  • two A groups can be bonded together to form a dimer through a suitable linking group that achieves the desired purpose.
  • linking groups include but are not limited to, urea, amide, -C(0)-C(0)-, carbamate and ketone.
  • two heteroaryl rings for example, two indole rings, are linked through a urea to form the dimer.
  • R 16 is 0 or 1 or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, Ci-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, Ci-C 6 alkoxy, -Co-C 4 alkyl(mono- and di- Ci-Cealkylamino), -Co-C 4 alkyl(C 3 -Cycycloalkyl), Ci-C 2 haloalkyl, and Ci-C 2 haloalkoxy.
  • R 19 is hydrogen, C C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, -S0 2 Ci-C 6 alkyl, (mono- and di-Ci-C 6 alkylamino)Ci-C 4 alkyl, -Co-C 4 alkyl(C 3 -Cycycloalkyl), -Co-C 4 alkyl(C 3 - Cyheterocycloalkyl), -C 0 -C 4 alkyl(aryl), C 0 -C 4 alkyl(heteroaryl), and -C(0)(CH 2 )i- 2 C(0)OR 9 each of which R 19 other than hydrogen is substituted with 0 or 1 or more substituents independently chosen from halogen, hydroxyl, amino, -COOH, and -C(0)OCi-C 4 alkyl.
  • X 11 is N or CR 11 .
  • X 12 is N or CR 12 .
  • X 13 is N or CR 13 .
  • X 14 is N or CR 14 .
  • R 17 , R 17 0 , or OH where R 17 is hydrogen or Ci-C 6 alkyl and R 18 and R 18 are independently chosen from hydrogen, halogen, hydroxymethyl, and methyl; and m is 0, 1, 2, or 3;
  • B is a monocyclic, bicyclic carbocyclic or carbocyclic-oxy group or a monocyclic, bicyclic, or tricyclic heterocyclic group having 1 , 2, 3, or 4 heteroatoms independently selected from N, O, and S and from 4 to 7 ring atoms per ring, or B is a C 2 - C 6 alkenyl or C 2 -C 6 alkynyl group, or B is -(Co-C 4 alkyl)(aryl), -(Co-C 4 alkyl)(heteroaryl), or -(C 0 - C 4 alkyl)(biphenyl) .
  • J is independently chosen at each occurrence from a covalent bond, Ci- C 4 alkylene, -OCi-C 4 alkylene, C 2 -C 4 alkenylene, and C 2 -C 4 alkynylene.
  • R 21 and R 22 are independently chosen at each occurrence from hydrogen, hydroxyl, cyano, amino, Ci-C 6 alkyl, Ci-C 6 haloalkyl, Ci-C 6 alkoxy, (C 3 -Cvcycloalkyl)Co-C 4 alkyl, (phenyl)C 0 -C 4 alkyl, -Ci-C 4 alkylOC(0)OCi-C 6 alkyl, -C C 4 alkylOC(0)Ci-C 6 alkyl, -C C 4 alkylC(0)OCi-C 6 alkyl, (4- to 7-membered heterocycloalkyl)Co-C 4 alkyl having 1 , 2, or 3 heteroatoms independently chosen from N, O, and S, and (5- or 6- membered unsaturated or aromatic heterocycle)Co-C 4 alkyl having 1 , 2, or 3 heteroatoms independently chosen from N, O, and S.
  • R 23 is independently chosen at each occurrence from (C -C 7 cycloalkyl)C 0 - C 4 alkyl, (phenyl)Co-C 4 alkyl, (4- to 7-membered heterocycloalkyl)Co-C 4 alkyl having 1 , 2, or 3 heteroatoms independently chosen from N, O, and S, and (5- or 6- membered unsaturated or aromatic heterocycle)Co-C 4 alkyl having 1 , 2, or 3 heteroatoms independently chosen from N, O, and S.
  • Either X 2 is nitrogen or at least one of (d), (e), (g), (i), (1), (n), (p), (s), (v), (x), and (y) is present.
  • Pharmaceutical compositions comprising a compound or salt of Formula I together with a pharmaceutically acceptable carrier are also disclosed.
  • ASD age-related macular degeneration
  • retinal degeneration other ophthalmic diseases (e.g., geographic atrophy), paroxysymal nocturnal hemoglobinuria (PNH), multiple sclerosis (MS), arthritis including rheumatoid arthritis (RA), a respiratory disease or a cardiovascular disease)
  • PNH paroxysymal nocturnal hemoglobinuria
  • MS multiple sclerosis
  • RA rheumatoid arthritis
  • a respiratory disease or a cardiovascular disease comprising administering a therapeutically effective amount of a compound or salt of Formula I to a host, including a human, in need of such treatment are also disclosed.
  • an effective amount of an active factor D inhibiting compound is provided to treat an inflammatory or immune disorder, including an autoimmune disorder, that is meadited or affected by factor D.
  • an inflammatory or immune disorder including an autoimmune disorder, that is meadited or affected by factor D.
  • the compound of Formula I can be used to treat a disorder mediated by the complement pathway, regardless whether it is acting through Factor D.
  • the present invention includes at least the following features:
  • cascade factor D including age-related macular degeneration (AMD), retinal degeneration, paroxysymal nocturnal hemoglobinuria (PNH), multiple sclerosis (MS), and rheumatoid arthritis (RA) and other disorders described further herein;
  • the compounds in any of the Formulas described herein include enantiomers, mixtures of enantiomers, diastereomers, tautomers, racemates and other isomers, such as rotamers, as if each is specifically described.
  • "Formula I” includes all subgeneric groups of Formula I, such as Formula IA and Formula IB and also includes pharmaceutically acceptable salts of a compound of Formula I, unless clearly contraindicated by the context in which this phrase is used.
  • Forma I also includes all subgeneric groups of Formula I, such as Formulas IC - ID, and Formulas II - XXX, and also includes pharmaceutically acceptable salts of all subgeneric groups of Formula I, such as Formulas IA - ID, and Formulas II - XXX, unless contraindicated by the context in which this phrase is used.
  • the present invention includes compounds of Formula I and the use of compounds with at least one desired isotopic substitution of an atom, at an amount above the natural abundance of the isotope, i.e., enriched.
  • Isotopes are atoms having the same atomic number but different mass numbers, i.e., the same number of protons but a different number of neutrons.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as 2 H, 3 H, U C, 13 C, 14 C, 15 N, 18 F, 31 P, 32 P, 35 S, 36 CI, 125 I respectively.
  • the invention includes various isotopically labeled compounds as defined herein, for example those into which radioactive isotopes, such as 3 H, 13 C, and 14 C, are present.
  • the deuterium can be bound to carbon in a location of bond breakage during metabolism (an a-deuterium kinetic isotope effect) or next to or near the site of bond breakage (a ⁇ -deuterium kinetic isotope effect).
  • Isotopic substitutions for example deuterium substitutions, can be partial or complete. Partial deuterium substitution means that at least one hydrogen is substituted with deuterium.
  • the isotope is 90, 95 or 99% or more enriched in an isotope at any location of interest.
  • deuterium is 90, 95 or 99% enriched at a desired location. Unless otherwise stated, the enrichment at any point is above natural abundance and enough to alter a detectable property of the drug in a human.
  • the substitution of a hydrogen atom for a deuterium atom occurs within an R group substituent on the L-B moiety region. In one embodiment, the substitution of a hydrogen atom for a deuterium atom occurs within an R group selected from any of R 18 , R 18' , R 33 , R 34 , R 35 , and/or R 36. In one embodiment, the substitution of a hydrogen atom for a deuterium atom occurs within an R group substituent within the A-carbonyl moiety region.
  • the substitution of a hydrogen atom for a deuterium atom occurs within an R group when at least one of the variables within the R group is hydrogen (e.g., 2 H or D) or alkyl (e.g., CD 3 ).
  • the alkyl residue is typically deuterated, e.g., CD 3i CH 2 CD 3 or CD 2 CD 3 .
  • the hydrogen may be isotopically enriched as deuterium (i.e., 2 H).
  • a dash (“-") that is not between two letters or symbols is used to indicate a point of attachment for a substituent.
  • a stable compound or stable structure refers to a compound leading to a compound that can be isolated and can be formulated into a dosage form with a shelf life of at least one month.
  • "optionally substituted” includes one or more substituents independently chosen from halogen, hydroxyl, amino, cyano, -CHO, -COOH, -CONH 2 , Ci-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, -Ci- C 6 alkoxy, C 2 -C 6 arkanoyl, Ci-Cealkylester, (mono- and di-Ci-C 6 alkylamino)Co-C 2 alkyl, Ci- C 2 haloalkyl, hydoxyCi-C 6 alkyl, ester, carbamate, urea, sulfonamide,-Ci-C 6 alkyl(heterocyclo), Ci-C 6 alkyl(heteroaryl), -Ci-C 6 alkyl(C 3 -C 7 cycloalkyl),
  • Alkyl is a branched or straight chain saturated aliphatic hydrocarbon group. In one embodiment, the alkyl contains from 1 to about 12 carbon atoms, more generally from 1 to about 6 carbon atoms or from 1 to about 4 carbon atoms. In one embodiment, the alkyl contains from 1 to about 8 carbon atoms. In certain embodiments, the alkyl is Ci-C 2 , Ci-C 3 , or Ci-C 6 .
  • the specified ranges as used herein indicate an alkyl group having each member of the range described as an independent species.
  • Ci-C 6 alkyl indicates a straight or branched alkyl group having from 1, 2, 3, 4, 5, or 6 carbon atoms and is intended to mean that each of these is described as an independent species.
  • Ci- C 4 alkyl indicates a straight or branched alkyl group having from 1, 2, 3, or 4 carbon atoms and is intended to mean that each of these is described as an independent species.
  • C 0 -C n alkyl When C 0 -C n alkyl is used herein in conjunction with another group, for example, (C 3 _ C 7 cycloalkyl)Co-C 4 alkyl, or -Co-C 4 alkyl(C 3 -C 7 cycloalkyl), the indicated group, in this case cycloalkyl, is either directly bound by a single covalent bond (Coalkyl), or attached by an alkyl chain in this case 1, 2, 3, or 4 carbon atoms. Alkyls can also be attached via other groups such as heteroatoms as in -0-Co-C 4 alkyl(C 3 -C 7 cycloalkyl).
  • alkyl examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, isopentyl, tert-pentyl, neopentyl, n-hexyl, 2-methylpentane, 3-methylpentane, 2,2- dimethylbutane and 2,3-dimethylbutane.
  • the alkyl group is optionally substituted as described above.
  • alkenyl is a branched or straight chain aliphatic hydrocarbon group having one or more carbon-carbon double bonds that may occur at a stable point along the chain.
  • Nonlimiting examples are C 2 -Csalkenyl, C 2 -C 6 alkenyl and C 2 -C 4 alkenyl.
  • the specified ranges as used herein indicate an alkenyl group having each member of the range described as an independent species, as described above for the alkyl moiety.
  • alkenyl include, but are not limited to, ethenyl and propenyl. In one embodiment, the alkenyl group is optionally substituted as described above.
  • Alkynyl is a branched or straight chain aliphatic hydrocarbon group having one or more carbon-carbon triple bonds that may occur at any stable point along the chain, for example, C 2 -Cgalkynyl or C2-C 6 alkynyl.
  • the specified ranges as used herein indicate an alkynyl group having each member of the range described as an independent species, as described above for the alkyl moiety.
  • alkynyl examples include, but are not limited to, ethynyl, propynyl, 1- butynyl, 2-butynyl, 3-butynyl, 1-pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1-hexynyl, 2- hexynyl, 3-hexynyl, 4-hexynyl and 5-hexynyl.
  • the alkynyl group is optionally substituted as described above.
  • Alkylene is a bivalent saturated hydrocarbon.
  • Alkylenes for example, can be a 1 to 8 carbon moiety, 1 to 6 carbon moiety, or an indicated number of carbon atoms, for example Ci-C 4 alkylene, Ci-C 3 alkylene, or Ci-C 2 alkylene.
  • Alkynylene is a bivalent hydrocarbon having at least one carbon-carbon triple bond.
  • Alkynylenes for example, can be a 2 to 8 carbon moiety, 2 to 6 carbon moiety, or an indicated number of carbon atoms, for example C 2 -C 4 alkynylene.
  • alkenyloxy is an alkenyl group as defined covalently bound to the group it substitutes by an oxygen bridge (-0-).
  • Haloalkyl indicates both branched and straight-chain alkyl groups substituted with 1 or more halogen atoms, up to the maximum allowable number of halogen atoms.
  • haloalkyl include, but are not limited to, trifluoromethyl, monofluoromethyl, difluoromethyl, 2-fluoroethyl, and penta-fluoroethyl.
  • Haloalkoxy indicates a haloalkyl group as defined herein attached through an oxygen bridge (oxygen of an alcohol radical).
  • Hydroxyalkyl is an alkyl group as previously described, substituted with at least one hydroxyl subsitutuent.
  • Halo or "halogen” indicates independently any of fluoro, chloro, bromo, and iodo.
  • Aryl indicates aromatic groups containing only carbon in the aromatic ring or rings.
  • the aryl groups contain 1 to 3 separate or fused rings and is 6 to about 14 or 18 ring atoms, without heteroatoms as ring members.
  • such aryl groups may be further substituted with carbon or non-carbon atoms or groups.
  • substitution may include fusion to a 5 to 7-membered saturated cyclic group that optionally contains 1 or 2 heteroatoms independently chosen from N, O, and S, to form, for example, a 3,4- methylenedioxyphenyl group.
  • Aryl groups include, for example, phenyl and naphthyl, including 1-naphthyl and 2-naphthyl.
  • aryl groups are pendant.
  • An example of a pendant ring is a phenyl group substituted with a phenyl group.
  • the aryl group is optionally substituted as described above.
  • heterocycle refers to a saturated or a partially unsaturated (i.e., having one or more double and/or triple bonds within the ring without aromaticity) carbocyclic radical of 3 to about 12, and more typically 3, 5, 6, 7 to 10 ring atoms in which at least one ring atom is a heteroatom selected from nitrogen, oxygen, phosphorus and sulfur, the remaining ring atoms being C, where one or more ring atoms is optionally substituted independently with one or more substituents described above.
  • a heterocycle may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms and 1 to 4 heteroatoms selected from N, O, P, and S) or a bicycle having 6 to 10 ring members (4 to 9 carbon atoms and 1 to 6 heteroatoms selected from N, O, P, and S), for example: a bicyclo [4,5], [5,5], [5,6], or [6,6] system.
  • the only heteroatom is nitrogen.
  • the only heteroatom is oxygen.
  • the only heteroatom is sulfur.
  • Heterocycles are described in Paquette, Leo A.; "Principles of Modern Heterocyclic Chemistry" (W. A.
  • Heterocycloalkyl is a saturated ring group. It may have, for example, 1, 2, 3, or 4 heteroatoms independently chosen from N, S, and O, with remaining ring atoms being carbon. In a typical embodiment, nitrogen is the heteroatm. Monocyclic heterocycloalkyl groups typically have from 3 to about 8 ring atoms or from 4 to 6 ring atoms. Examples of heterocycloalkyl groups include morpholinyl, piperazinyl, piperidinyl, and pyrrolinyl.
  • “Pharmaceutical compositions” are compositions comprising at least one active agent, such as a compound or salt of Formula I, and at least one other substance, such as a carrier.
  • “Pharmaceutical combinations” are combinations of at least two active agents which may be combined in a single dosage form or provided together in separate dosage forms with instructions that the active agents are to be used together to treat any disorder described herein.
  • “Pharmaceutically acceptable salts” includes derivatives of the disclosed compounds in which the parent compound is modified by making inorganic and organic, nontoxic, acid or base addition salts thereof.
  • the salts of the present compounds can be synthesized from a parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate, or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two.
  • carrier applied to pharmaceutical compositions/combinations of the invention refers to a diluent, excipient, or vehicle with which an active compound is provided.
  • a "patient” or “host” or “subject” is a human or non-human animal in need of modulation of the complement factor D pathway. Typically the host is a human.
  • a "patient” or “host” or “subject” also refers to for example, mammals, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like.
  • Nonlimiting examples of prodrug strategies include covalent attachment of removable groups, or removable portions of groups, for example, but not limited to acylation, phosphorylation, phosphonylation, phosphoramidate derivatives, amidation, reduction, oxidation, esterification, alkylation, other carboxy derivatives, sulfoxy or sulfone derivatives, carbonylation or anhydride, among others.
  • Providing a compound of Formula I with at least one additional active agent means the compound of Formula I and the additional active agent(s) are provided simultaneously in a single dosage form, provided concomitantly in separate dosage forms, or provided in separate dosage forms for administration separated by some amount of time that is within the time in which both the compound of Formula I and the at least one additional active agent are within the blood stream of a patient.
  • the compound of Formula I and the additional active agent need not be prescribed for a patient by the same medical care worker.
  • the additional active agent or agents need not require a prescription.
  • Non- limiting examples of compounds falling within Formula I with variations in the variables e.g., A, B, R ⁇ -R 3 , and L, are illustrated below. The disclosure includes all combinations of these definitions so long as a stable compound results.
  • n 0 or 1.
  • the disclosure includes compounds and salts of Formula I and pharmaceutically acceptable compositions thereof, and any of its subformulae (II-XXX) in which at least one of the following conditions is met in the embodiments described below.
  • R 12 and R 13 are independently chosen at each occurrence from (q), (r) and (s):
  • R 12 or R 13 are independently selected from:
  • the disclosure provides compounds of Formula I, wherein;
  • R 30 is as defined in the summary section above.
  • R 5 is hydrogen, halogen, or Ci-C 2 alkyl
  • R 11 , R 13 , R 14 , and R 15 are independently chosen at each occurrence from hydrogen, halogen, hydroxyl, amino, Ci-C 4 alkyl, Ci-C 4 alkoxy, -Co-C 2 alkyl(mono- and di- Ci-C 2 alkylamino), trifluoromethyl, and trifluoromethoxy;
  • R 12 and R 13 are selected from hydrogen, halogen, Ci-C 4 alkyl, Ci-C 4 alkoxy, trifluoromethyl, and trifluoromethoxy; the other of R 12 and R 13 is chosen from (s):
  • the central core moiety is 4-fluoroproline.
  • R 1 , R 1 ' , R 2 ' , R 3 , and R 3 ' if present, are all hydrogen; and R 2 is fluoro.
  • the compound of Formula I has the structure:
  • the central pyrrolidine is modified by addition of a second heteroatom to a pyrrolidine ring, such as N, O, S, or Si, for example:
  • a second heteroatom such as N, O, S, or Si
  • Another modification within the scope of the disclosure is joining a substituent on the central pyrrolidine ring to R 7 or R 8 to form a 5- to 6- membered heterocyclic ring, for example:
  • R 18 and R 18 are independently chosen from hydrogen, halogen, hydroxymethyl, and methyl; and m is 0 or 1 ; and
  • the disclosure further includes compounds and salts of Formula I in which B is 2-fluoro-3-chlorophenyl.
  • B is 2-fluoro-3-chlorophenyl.
  • another carbocyclic, aryl, heterocyclic, or heteroaryl group such as 2-bromo-pyridin-6-yl, l-(2,2,2-trifluoroethyl)-lH- pyrazol-3-yl, 2,2-dichlorocyclopropylmethyl, or 2-fluoro-3-trimethylsilylphenyl is used.
  • B is phenyl, pyridyl, or indanyl each of which is unsubstituted or substituted with one or more substituents independently chosen from hydrogen, halogen, hydroxyl, nitro, cyano, Ci-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, Ci-C 6 alkoxy, Ci- C 6 thioalkyl, (mono- and di-Ci-C 6 alkylamino)Co-C 4 alkyl, (C 3 -Cvcycloalkyl)Co-C 4 alkyl, -C 0 - C 4 alkoxy(C 3 -C 7 cycloalkyl), (phenyl)Co-C 2 alkyl, (pyridyl)Co-C 2 alkyl; each of which substituents other than hydrogen, halogen, hydroxyl, nitro, cyano, is unsubstituted or substituted with one
  • R 23 is independently chosen at each occurrence from (C 3 - Cycycloalkyl)Co-C 4 alkyl, (phenyl)Co-C 4 alkyl, (4- to 7-membered heterocycloalkyl)Co-C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S, and (5- or 6- membered unsaturated or aromatic heterocycle)Co-C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S.
  • R s y rogen met y, or tr uoromet y
  • R s y rogen or aogen an R s hydrogen, methyl, trifluoromethyl, or -Si(CH 3 )2C(CH 3 ) 3 .
  • A is a group chosen from (k) and (1) where (k) is
  • R 4 is (m) or (n):
  • each of which R 4 other than hydrogen, -CHO, and -CONH 2 is unsubstituted or substituted with one or more of amino, imino, halogen, hydroxyl, cyano, cyanoimino, Ci-C 2 alkyl, Ci-C 2 alkoxy, -Co-C 2 alkyl(mono- and di-Ci-C 4 alkylamino), Ci-C 2 haloalkyl, and Ci-C 2 haloalkoxy.
  • R 5 and R 6 are independently chosen from (o) and (p):
  • R 5 and R 6 other than hydrogen, hydroxyl, cyano, and -COOH is unsubstituted or optionally substituted.
  • R 5 and R 6 other than hydrogen, hydroxyl, cyano, and -COOH may be substituted with one or more substituents independently chosen from halogen, hydroxyl, amino, imino, cyano, cyanoimino, Ci-C 2 alkyl, Ci-C 4 alkoxy, -Co- C 2 alkyl(mono- and di-Ci-C 4 alkylamino), Ci-C 2 haloalkyl, Ci-C 2 haloalkoxy, C(0)alkyl, C(0)cycloalkyl, C(0)aryl, C(0)heterocycle, and C(0)heteroaryl.
  • R 7 is hydrogen, Ci-C 6 alkyl, or -C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl).
  • R 8 and R 8 are independently chosen from hydrogen, halogen, hydroxyl, Ci- C 6 alkyl, Ci-C 6 alkoxy, and (Ci-C 4 alkylamino)Co-C 2 alkyl, or R 8 and R 8 are taken together to form an oxo group, or can be taken together with the carbon that they are bonded to form a 3- membered carbocyclic ring.
  • R 19 is hydrogen, Ci-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, -S0 2 Ci-C 6 alkyl, (mono- and di-Ci-C 6 alkylamino)Ci-C 4 alkyl, -Co-C 4 alkyl(C3-C 7 cycloalkyl), -C 0 -C 4 alkyl(C 3 - C 7 heterocycloalkyl), -Co-C 4 alkyl(aryl), Co-C 4 alkyl(heteroaryl), each of which R 19 other than hydrogen is substituted with 0 or 1 or more substituents independently chosen from halogen, hydroxyl, amino, -COOH, and -C(0)OCi-C 4 alkyl.
  • X 12 is N or CR 12 .
  • X 14 is N or CR 14 .
  • R 11 , R 14 , and R 15 are independently chosen at each occurrence from hydrogen, halogen, hydroxyl, nitro, cyano, -NR 9 C(0)R 10 , C(0)NR 9 R 10 , -0(PO)(OR 9 ) 2 , -(PO)(OR 9 ) 2 , d- C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkenyl(aryl), C 2 -C 6 alkenyl(cycloalkyl), C 2 -C 6 alkenyl(heterocycle), C 2 -C 6 alkenyl(heteroaryl), C 2 -C 6 alkynyl, C 2 -C 6 alkynyl(aryl), C 2 -C 6 alkynyl(cycloalkyl), C 2 - C 6 alkynyl(heterocycle), C 2 -Cealkynyl(heteroaryl), C 2 -C 6 alkyl
  • R 5 and R 6 are independently chosen from -CHO, -C(0)NH 2 , -C(0)NH(CH 3 ), C 2 -C 6 alkanoyl, and hydrogen.
  • each R 5 and R 6 other than hydrogen, hydroxyl, cyano, and -COOH is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, amino, imino, cyano, cyanoimino, Ci-C 2 alkyl, Ci-C 4 alkoxy, -Co- C 2 alkyl(mono- and di-Ci-C 4 alkylamino), Ci-C 2 haloalkyl, Ci-C 2 haloalkoxy i C(0)alkyl, C(0)cycloalkyl, C(0)aryl, C(0)heterocycle, and C(0)heteroaryl.
  • R 8 and R 8 are independently hydrogen or methyl.
  • R 8 and R 8 are hydrogen.
  • R 7 is hydrogen or methyl.
  • R is hydrogen.
  • two A groups can be bonded together to form a dimer through a suitable linking group that achieves the desired purpose.
  • linking groups include but are not limited to, urea, amide, -C(0)-C(0)-, carbamate and ketone.
  • two heteroaryl rings for example, two indole rings, are linked through a urea to form indole-NHC(0)NH-indole.
  • this disclosure includes compounds and salts of Formula IA:
  • R 6 , R 13 , and B may carry any of the definitions set forth herein for this variable.
  • this disclosure includes compounds and salts of Formula IB, IC, and I
  • R 1 , R 2 , R 2 , and R 3 are independently chosen from hydrogen, halogen, Ci- C 4 alkyl, Ci-C 4 alkoxy, -C 0 -C 2 alkylNR 9 R 10 , -Co-C 4 alkyl(C 3 -C 7 cycloalkyl), -O-C 0 -C 4 alkyl(C 3 - C 7 cycloalkyl), Ci-C 2 haloalkyl, and Ci-C 2 haloalkoxy;
  • R 8 and R 8 are independently chosen from hydrogen, halogen, and methyl;
  • R 5 is hydrogen, hydroxyl, cyano, -COOH, Ci-C 6 alkyl, Ci-C 6 alkoxy, C 2 - C 6 alkanoyl -C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), -C(O)C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl, Ci-C 2 haloalkyl, or C i -C 2 haloalkoxy ;
  • R 6 is -C(0)CH 3 , -C(0)NH 2 , -C(0)CF 3 , -C(0)(cyclopropyl), or
  • R 11 and R 14 are independently chosen from hydrogen, halogen, hydroxyl, amino, nitro, cyano, Ci-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, Ci-C 6 alkoxy, Ci-C 6 thioalkyl, -C 0 - C 4 alkyl(mono- and di-Ci-Cealkylamino), -Co-C 4 alkyl(C 3 -C 7 cycloalkyl), -OCo-C 4 alkyl(C 3 - C 7 cycloalkyl), Ci-C 2 haloalkyl, and Ci-C 2 haloalkoxy.
  • compositions comprising an effective amount of compound or pharmaceutically acceptable salt of Formula I, together with at least one pharmaceutically acceptable carrier.
  • the pharmaceutical composition may contain a compound or salt of Formula I as the only active agent, or, in an alternative embodiment, Formula I and at least one additional active agent.
  • the pharmaceutical composition is in a dosage form that contains from about 0.1 mg to about 2000 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg of a compound of Formula I and optionally from about 0.1 mg to about 2000 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg of an additional active agent in a unit dosage form.
  • Examples are dosage forms with at least 25, 50, 100, 200, 250, 300, 400, 500, 600, 700, or 750 mg of active compound, or its salt.
  • the pharmaceutical composition may also include a molar ratio of a compound of Formula I and an additional active agent.
  • the pharmaceutical composition may contain a molar ratio of about 0.5 : 1 , about 1 : 1, about 2: 1, about 3 : 1 or from about 1.5:1 to about 4: 1 of an another antiinflammatory agent.
  • Compounds disclosed herein may be administered orally, topically, parenterally, by inhalation or spray, sublingually, via implant, including ocular implant, transdermally, via buccal administration, rectally, as an ophthalmic solution, injection, including ocular injection, intraveneous, intra-aortal, intracranial, or by other means, in dosage unit formulations containing conventional pharmaceutically acceptable carriers.
  • the pharmaceutical composition may be formulated as any pharmaceutically useful form, e.g., as an aerosol, a cream, a gel, a pill, a capsule, a tablet, a syrup, a transdermal patch, or an ophthalmic solution.
  • Some dosage forms, such as tablets and capsules are subdivided into suitably sized unit doses containing appropriate quantities of the active components, e.g., an effective amount to achieve the desired purpose.
  • Carriers include excipients and diluents and must be of sufficiently high purity and sufficiently low toxicity to render them suitable for administration to the patient being treated.
  • the carrier can be inert or it can possess pharmaceutical benefits of its own.
  • the amount of carrier employed in conjunction with the compound is sufficient to provide a practical quantity of material for administration per unit dose of the compound.
  • Classes of carriers include, but are not limited to binders, buffering agents, coloring agents, diluents, disintegrants, emulsifiers, flavorants, glidents, lubricants, preservatives, stabilizers, surfactants, tableting agents, and wetting agents.
  • Some carriers may be listed in more than one class, for example vegetable oil may be used as a lubricant in some formulations and a diluent in others.
  • Exemplary pharmaceutically acceptable carriers include sugars, starches, celluloses, powdered tragacanth, malt, gelatin; talc, and vegetable oils.
  • Optional active agents may be included in a pharmaceutical composition, which do not substantially interfere with the activity of the compound of the present invention.
  • compositions/combinations can be formulated for oral administration.
  • These compositions can contain any amount of active compound for Formula I that achieves the desired result, for example between 0.1 and 99 weight % (wt.%) of a compound of Formula I and usually at least about 5 wt.% of a compound of Formula I.
  • Some embodiments contain from about 25 wt.% to about 50 wt. % or from about 5 wt.% to about 75 wt.% of the compound of Formula I.
  • the complement factor D inhibitors of the present invention can be administered, for example, either systemically or locally.
  • Systemic administration includes, for example, oral, transdermal, subdermal, intraperitioneal, subcutaneous, transnasal, sublingual, or rectal.
  • Local administration for ocular administration includes: topical, intravitreal, periocular, transscleral, retrobulbar, juxtascleral, sub-tenon, or via an intraocular device.
  • the inhibitors may be delivered via a sustained delivery device implanted intravitreally or transsclerally, or by other known means of local ocular delivery.
  • the compounds and pharmaceutical compositions disclosed herein are useful for treating or preventing a disorder that is mediated by the complement pathway, and in particular, a pathway that is modulated by complement factor D.
  • the disorder is an inflammatory disorder, an immune disorder, an autoimmune disorder, or complement factor D related disorders in a host.
  • the disorder is an ocular disorder.
  • Complement mediated disorders that may be treated or prevented by the compounds and compositions of this disclosure include, but are not limited to, inflammatory effects of sepsis, systemic inflammatory response syndrome (SIRS), ischemia/ reperfusion injury (I/R injury), psoriasis, myasthenia gravis, system lupus erythematosus (SLE), paroxysmal nocturnal hemoglobinuria (PNH), hereditary angioedema, multiple sclerosis, trauma, burn injury, capillary leak syndrome, obesity, diabetes, Alzheimer's dementia, stroke, schizophrenia, epilepsy, age-related macular degeneration, glaucoma, diabetic retinopathy, asthma, allergy, acute respiratory distress syndrome (ARDS), atypical hemolytic uremic syndrome (aHUS), hemolytic uremic syndrome (HUS), cystic fibrosis, myocardial infarction, lupus nephritides, Crohn's disease, rheumatoid arthritis, at
  • lung disease and disorders such as dyspnea, hemoptysis, chronic obstructive pulmonary disease (COPD), emphysema, pulmonary embolisms and infarcts, pneumonia, fibrogenic dust diseases, inert dusts and minerals (e.g., silicon, coal dust, beryllium, and asbestos), pulmonary fibrosis, organic dust diseases, chemical injury (due to irritant gases and chemicals, e.g., chlorine, phosgene, sulfur dioxide, hydrogen sulfide, nitrogen dioxide, ammonia, and hydrochloric acid), smoke injury, thermal injury (e.g., burn, freeze), bronchoconstriction, hypersensitivity pneumonitis, parasitic diseases, Goodpasture's Syndrome, pulmonary vasculitis, Pauci-immune vasculitis, immune complex- associated inflammation, uveitis (including Behcet's disease and other sub-types of uveitis), antiphospholipid syndrome, arthritis,
  • COPD chronic ob
  • complement mediated diseases include ophthalmic diseases (including early or neovascular age-related macular degeneration and geographic atrophy), autoimmune diseases (including arthritis, rheumatoid arthritis), respiratory diseases, cardiovascular diseases.
  • ophthalmic diseases including early or neovascular age-related macular degeneration and geographic atrophy
  • autoimmune diseases including arthritis, rheumatoid arthritis
  • respiratory diseases including cardiovascular diseases.
  • the compounds of the invention are suitable for use in the treatment of diseases and disorders associated with fatty acid metabolism, including obesity and other metabolic disorders.
  • a method for the treatment of paroxysmal nocturnal hemoglobinuria includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
  • a method for the treatment of age- related macular degeneration is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
  • a method for the treatment of rheumatoid arthritis is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
  • a method for the treatment of multiple sclerosis includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
  • a method for the treatment of myasthenia gravis includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
  • a method for the treatment of atypical hemolytic uremic syndrome (aHUS) is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
  • a method for the treatment of C3 glomerulonephritis includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
  • a method for the treatment of abdominal aortic aneurysm includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
  • a method for the treatment of neuromyelitis optica (NMO) is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
  • the present invention provides methods of treating or preventing an inflammatory disorder or a complement related disease, by administering to a host in need thereof an effective amount of a compound of Formula I of the invention.
  • the present invention provides methods of treating or preventing an inflammatory disorder more generally, an immune disorder, autoimmune disorder, or complement factor D related disease, by providing an effective amount of a compound or pharmaceutically acceptable salt of Formula I to patient with a factor D mediated inflammatory disorder.
  • a compound or salt of Formula I may be provided as the only active agent or may be provided together with one or more additional active agents.
  • a method for the treatment of a disorder associated with a dysfunction in the complement cascade includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
  • a method of inhibiting activation of the alternative complement pathway in a subject includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
  • a method of modulating factor D activity in a subject is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
  • prevention means decreasing the likelihood of the appearance of symptoms in a patient administered the compound prophylactically as compared to the likelihood of the appearance of symptoms in patients not administered the compound or decreasing the severity of symptoms in a patient administered the compound prophylactically as compared to the severity of symptoms experienced by patients with the disorder or condition who were not administered the compound.
  • an effective amount of a compound of Formula I is used to prevent or prophylaxis of a complement factor D related disorder.
  • An effective amount of a pharmaceutical composition/ combination of the invention may be an amount sufficient to (a) inhibit the progression of a disorder mediated by the complement pathway, including an inflammatory, immune, including an autoimmune, disorder or complement factor D related disease; (b) cause a regression of an inflammatory, immune, including an autoimmune, disorder or complement factor D related disease; or (c) cause a cure of an inflammatory, immune, including an autoimmune, disorder or complement factor D related disease.
  • An effective amount of a compound or pharmaceutical composition described herein will also provide a sufficient amount of the active agent when administered to a patient to provide a clinical benefit. Such an amount may be ascertained experimentally, for example by assaying blood concentration of the agent, or theoretically, by calculating bioavailability.
  • a compound or salt of Formula I may be provided in combination or alternation with at least one additional inhibitor of the complement system or a second active compound with a different biological mechanism of action.
  • a compound or salt of Formula I may be provided in combination with a complement C5 inhibitor or C5 convertase inhibitor.
  • a compound or salt of Formula I may be provided in combination with eculizumab.
  • a compound or salt of Formula I may be provided in combination with additional inhibitors of factor D.
  • a compound or salt of Formula I may be provided together with a compound that inhibits an enzyme that metabolizes protease inhibitors. In one embodiment, a compound or salt of Formula I may be provided together with ritonavir.
  • a compound or salt of Formula I may be provided together with a protease inhibitor, a soluble complement regulator, a therapeutic antibody (monoclonal or polyclonal), complement component inhibitors, receptor agonists, or siRNAs.
  • Nonlimiting examples of active agents in these categories are: [0246] Protease inhibitors: plasma-derived Cl-INH concentrates, for example Cetor® (Sanquin), Berinert-P® (CSL Behring, Lev Pharma), and Cinryze®; and recombinant human CI -inhibitors, for example Rhucin®;
  • Soluble complement regulators Soluble complement receptor 1 (TP 10) (Avant
  • Immunotherapeutics sCRl-sLe x /TP-20 (Avant Immunotherapeutics); MLN-2222 /CAB-2 (Millenium Pharmaceuticals); Mirococept (Inflazyme Pharmaceuticals);
  • Therapeutic antibodies Eculizumab/Soliris (Alexion Pharmaceuticals); Pexelizumab (Alexion Pharmaceuticals); Ofatumumab (Genmab A/S); TNX-234 (Tanox); TNX- 558 (Tanox); TA106 (Taligen Therapeutics); Neutrazumab (G2 Therapies); Anti-properdin (Novelmed Therapeutics); HuMax-CD38 (Genmab A/S);
  • Complement component inhibitors include Compstatin/POT-4 (Potentia Pharmaceuticals); ARC 1905 (Archemix);
  • Receptor agonists PMX-53 (Peptech Ltd.); JPE-137 (Jerini); JSM-7717 (Jerini);
  • the present invention provides a method of treating or preventing age-related macular degeneration (AMD) by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention.
  • AMD age-related macular degeneration
  • the compositions of the present invention are administered in combination with an anti-VEGF agent.
  • Nonlimiting examples of anti-VEGF agents include, but are not limited to, aflibercept (Eylea®; Regeneron Pharmaceuticals); ranibizumab (Lucentis®: Genentech and Novartis); and pegaptanib (Macugen®; OSI Pharmaceuticals and Pfizer); Bevacizumab (Avastin; Genentech/Roche); anecortane acetate, squalamine lactate, and corticosteroids, including, but not limited to, triamcinolone acetonide.
  • a compound of Formula I can be combined with a second agent in order to treat a disorder of the eye.
  • Examples of types of therapeutic agents that can be used in combination for ocular applications include anti-inflammatory drugs, antimicrobial agents, anti-angiogenesis agents, immunosuppressants, antibodies, steroids, ocular antihypertensive drugs and combinations thereof.
  • therapeutic agents include amikacin, anecortane acetate, anthracenedione, anthracycline, an azole, amphotericin B, bevacizumab, camptothecin, cefuroxime, chloramphenicol, chlorhexidine, chlorhexidine digluconate, clortrimazole, a clotrimazole cephalosporin, corticosteroids, dexamethasone, desamethazone, econazole, eftazidime, epipodophyllotoxin, fluconazole, flucytosine, fluoropyrimidines, fluoroquinolines, gatifloxacin, glycopeptides, imidazoles, itrac
  • eye disorders that may be treated according to the compositions and methods disclosed herein include amoebic keratitis, fungal keratitis, bacterial keratitis, viral keratitis, onchorcercal keratitis, bacterial keratoconjunctivitis, viral keratoconjunctivitis, corneal dystrophic diseases, Fuchs' endothelial dystrophy, Sjogren's syndrome, Stevens-Johnson syndrome, autoimmune dry eye diseases, environmental dry eye diseases, corneal neovascularization diseases, post-corneal transplant rejection prophylaxis and treatment, autoimmune uveitis, infectious uveitis, anterior uveitis, posterior uveitis (including toxoplasmosis), pan-uveitis, an inflammatory disease of the vitreous or retina, endophthalmitis prophylaxis and treatment, macular edema, macular degeneration, age related macular degeneration, proliferative and non-prolife
  • a compound of Formula I, or a combination of Formula I and another active agent can be administered into an eye compartment of via injection into the vitreous chamber, subretinal space, subchoroidal space, the episclera, the conjunctiva, the sclera, the anterior chamber, and the cornea and compartments therein (e.g., subepithelial, intrastromal, endothelial).
  • a compound of Formula I, or a combination of Formula I and another active agent can be administered into an eye compartment via binding to a mucosal penetrating particle to treat a condition located in the vitreous chamber, subretinal space, subchoroidal space, the episclera, the conjunctiva, the sclera or the anterior chamber, and the cornea and compartments therein (e.g., subepithelial, intrastromal, endothelial).
  • Mucosal penetrating particles are known in the art, and are described in, for example, PCT published application WO 2013166436 to Kala Pharmaceuticals, incorporated in its entirety herein.
  • a composition comprising compound of Formula I suitable for topical administration to an eye.
  • the pharmaceutical composition comprises a plurality of coated particles, comprising a core particle comprising a compound of Formula I, wherein Formula I constitutes at least about 80 wt% of the core particle, and a coating comprising one or more surface-altering agents, wherein the one or more surface-altering agents comprise at least one of a poloxamer, a poly(vinyl alcohol), or a polysorbate.
  • the one or more surface-altering agents is present on the outer surface of the core particle at a density of at least 0.01 molecules/nm.
  • the one or more surface-altering agents is present in the pharmaceutical composition in an amount of between about 0.001% to about 5% by weight.
  • the plurality of coated particles have an average smallest cross-sectional dimension of less than about 1 micron.
  • the pharmaceutical composition also includes one or more ophthalmically acceptable carriers, additives, and/or diluents.
  • particles suitable for use with the presently disclosed methods can exist in a variety of shapes, including, but not limited to, spheroids, rods, disks, pyramids, cubes, cylinders, nanohelixes, nanosprings, nanorings, rod- shaped particles, arrow-shaped particles, teardrop-shaped particles, tetrapod- shaped particles, prism-shaped particles, and a plurality of other geometric and non-geometric shapes.
  • the presently disclosed particles have a spherical shape.
  • the present invention provides a method of treating or preventing paroxysmal nocturnal hemoglobinuria (PNH) by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention. In one embodiment, the present invention provides a method of treating or preventing paroxysmal nocturnal hemoglobinuria (PNH) by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention in combination or alternation with additional inhibitors of the complement system or another active compound with a different biological mechanism of action.
  • PNH paroxysmal nocturnal hemoglobinuria
  • the present invention provides a method of treating or preventing paroxysmal nocturnal hemoglobinuria (PNH) by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention in combination or alternation with eculizumab.
  • PNH paroxysmal nocturnal hemoglobinuria
  • a compound of Formula I is administered in combination or alternation with at least one anti-rhuematoid arthritis drug selected from: salicylates including aspirin (Anacin, Ascriptin, Bayer Aspirin, Ecotrin) and salsalate (Mono-Gesic, Salgesic); nonsteroidal anti-inflammatory drugs (NSAIDs); nonselective inhibitors of the cyclo-oxygenase (COX-1 and COX-2) enzymes, including diclofenac (Cataflam, Voltaren), ibuprofen (Advil, Motrin), ketoprofen (Orudis), naproxen (Aleve, Naprosyn), piroxicam (Feldene), etodolac (Lodine), indomethacin, oxaprozin (Daypro), nabumetone (Relafen), and meloxicam (Mobic); selective cyclo-oxygenase-2 (COX-2) inhibitors, including diclofenac
  • the present invention provides a method of treating or preventing multiple sclerosis by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention. In one embodiment, the present invention provides a method of treating or preventing multiple sclerosis by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention in combination or alternation with additional inhibitors of the complement system. In another embodiment, the present invention provides a method of treating or preventing multiple sclerosis by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention in combination or alternation with a corticosteroid. Examples of corticosteroids include, but are not limited to, prednisone, dexamethasone, solumedrol, and methylprednisolone.
  • a compound of Formula I is combined with at least one anti- multiple sclerosis drug selected from: Aubagio (teriflunomide), Avonex (interferon beta- la), Betaseron (interferon beta- lb), Copaxone (glatiramer acetate), Extavia (interferon beta- lb), Gilenya (fmgolimod), Lemtrada (alemtuzumab), Novantrone (mitoxantrone), Plegridy (peginterferon beta- la), Rebif (interferon beta- la), Tecfidera (dimethyl fumarate), Tysabri (natalizumab), Solu-Medrol (methylprednisolone), High-dose oral Deltasone (prednisone), H.P. Acthar Gel (ACTH), and combinations thereof.
  • Aubagio teriflunomide
  • Avonex interferon beta- la
  • Betaseron interferon beta- lb
  • Copaxone
  • a compound or salt of Formula I may be provided in combination or alternation with an immunosuppressive agent or an anti-inflammatory agent.
  • a compound described herein can be administered in combination or alternation with at least one immunosuppressive agent.
  • the immunosuppressive agent as nonlimiting examples, may be a calcineurin inhibitor, e.g. a cyclosporin or an ascomycin, e.g. Cyclosporin A (NEORAL®), FK506 (tacrolimus), pimecrolimus, a mTOR inhibitor, e.g. rapamycin or a derivative thereof, e.g.
  • Sirolimus (RAPAMUNE®), Everolimus (Certican®), temsirolimus, zotarolimus, biolimus-7, biolimus-9, a rapalog, e.g.ridaforolimus, azathioprine, campath 1H, a SIP receptor modulator, e.g. fingolimod or an analogue thereof, an anti IL-8 antibody, mycophenolic acid or a salt thereof, e.g. sodium salt, or a prodrug thereof, e.g.
  • Mycophenolate Mofetil (CELLCEPT®), OKT3 (ORTHOCLONE OKT3®), Prednisone, ATGAM®, THYMOGLOBULIN®, Brequinar Sodium, OKT4, T10B9.A-3A, 33B3.1, 15-deoxyspergualin, tresperimus, Leflunomide ARAVA®, CTLAI-Ig, anti-CD25, anti-IL2R, Basiliximab (SIMULECT®), Daclizumab (ZENAPAX®), mizorbine, methotrexate, dexamethasone, ISAtx-247, SDZ ASM 981 (pimecrolimus, Elidel®), CTLA41g (Abatacept), belatacept, LFA31g, etanercept (sold as Enbrel® by Immunex), adalimumab (Humira®), infliximab (Remicade®), an anti-LFA-1 antibody, natal
  • anti-inflammatory agents include methotrexate, dexamethasone, dexamethasone alcohol, dexamethasone sodium phosphate, fluromethalone acetate, fluromethalone alcohol, lotoprendol etabonate, medrysone, prednisolone acetate, prednisolone sodium phosphate, difluprednate, rimexolone, hydrocortisone, hydrocortisone acetate, lodoxamide tromethamine, aspirin, ibuprofen, suprofen, piroxicam, meloxicam, flubiprofen, naproxan, ketoprofen, tenoxicam, diclofenac sodium, ketotifen fumarate, diclofenac sodium, nepafenac, bromfenac, flurbiprofen sodium, suprofen, celecoxib, naproxen, rofecoxib, glucocorticoids,
  • a compound of Formula I is combined with one or more non-steroidal anti-inflammatory drugs (NSAIDs) selected from naproxen sodium (Anaprox), celecoxib (Celebrex), sulindac (Clinoril), oxaprozin (Daypro), salsalate (Disalcid), diflunisal (Dolobid), piroxicam (Feldene), indomethacin (Indocin), etodolac (Lodine), meloxicam (Mobic), naproxen (Naprosyn), nabumetone (Relafen), ketorolac tromethamine (Toradol), naproxen/esomeprazole (Vimovo), and diclofenac (Voltaren), and combinations thereof.
  • NSAIDs non-steroidal anti-inflammatory drugs
  • a heteroaryl compound, Structure 9-1 is coupled to an activated ester, Structure 9-2, to generate Structure 9-3.
  • the leaving group, LGi is a halide.
  • LG is a halide.
  • Structure 9-3 is treated with an inorganic cyanide and two organometallic catalyst to generate structure 9- 4.
  • the inorganic cyanide is zinc cyanide.
  • the two organometallic catalysts are Pd 2 (dba) 3 and Pd(dppf)Cl 2 .
  • Structure 9-4 is treated with an organic acid to generate Structure 9-5.
  • the organic acid is trifluoroacetic acid.
  • Structure 9-5 is coupled to Structure 3 from Route 1 to generate compounds of Formula I. This chemistry is illustrated in Route 9.
  • a central core that is directed linked to an aryl or heteroaryl group in place of the L-B region can be prepared according to known processes.
  • a central core is alkylated to generate a central core -A compound comprising a carboxylic acid is reduced to an alcohol and used to generate an alkyl halide.
  • the alkyl halide is treated with a heteroaryl compound to generate a compound of Formula I.
  • This chemistry can be carried out by those skilled in the art of organic chemistry. See for example: Advanced Organic Chemistry: Reactions, Mechanisms and Structure by J. March.
  • 5-azaspiro[2.4]heptane-4,5-dicarboxylic acid, 5-(l,l-dimethylethyl) ester, (4S)-, CAS 209269-08-9 can be prepared as described in Tandon, M. et al. Bioorg. Med. Chem. Lett. 1998, 8, 1139-1144.
  • Step 2 the protected azaspiro[2.4]heptane is coupled to an amine in the presence of an organic solvent, a base and a coupling reagent to generate an amide bond; the L-B moiety.
  • the amine is (3-chloro-2-fluorophenyl) methanamine.
  • the organic solvent is DMF.
  • Boc-L-proline is coupled to an amine in the presence of an organic solvent, a base and a coupling reagent to generate an amide bond; the L-B moiety.
  • the amine is (3-chloro-2-fluorophenyl) methanamine.
  • the organic solvent is DMF.
  • the base is diisopropylethylamine.
  • the coupling reagent is HATU.
  • the Boc protecting group is removed.
  • the starting material is reacted with an acid in the presence of an organic solvent.
  • the acid is 4N hydrochloric acid.
  • the organic solvent is dioxane.
  • the compounds (2S,3S,4S)-4-fluoro-3-methoxy-pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester and (2R,3R,4R)-3-fluoro-4-methoxy-pyrrolidine-l,2-dicarboxylic acid 1-tert- butyl ester can be prepared as a mixture according to WO 2012/093101 to Novartis and the regioisomers can be ultimately separated once coupled to generate the central core-L-B synthons.
  • the compound (S)-Boc-5-oxopyrrolidine-2-carboxylic acid is available from the Aldrich Chemical Co.
  • Structure 1-5 from Example 4 Route 4a, is coupled to Structure 3 from Route 1 to generate compounds of Formula I. This chemistry is illustrated in Route 5 a.
  • Structure 3-5 from Example 4 Route 4c, is coupled to Structure 3 from Route 1 to generate compounds of Formula I. This chemistry is illustrated in Route 5c.
  • Step 3 (2S,4R)-N-(2'-Chloro-2-fluoro-[l,l'-biphenyl]-3-yl)-4-fluoropyrrolidine-2- carboxamide hydrochloride (Int-1)
  • SM8 was dissolved in DCM and equal volume of TFA was added. The mixture was stirred for 30 min at rt. The volatiles were removed under reduced pressure and the the residue Int-2 was used without further purification.
  • Step 4 tert-Butyl 2-(3-cyano-lH-thieno[3,2-c]pyrazol-l-yl)acetate(99e)
  • Step 6 l-(2-((2S,4R)-2-((2'-Chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-lH-thieno[3,2-c]pyrazole-3-carboxamide (99)
  • reaction mixture was stirred for 0.5 h at rt , then quenched by aq NaHC0 3 (5 mL).
  • the mixture was extracted with EtOAc (30 mL) and the organic layer was washed with water and brine.
  • the solvent was removed under reduced pressure and the remaining residue was purified by column chromatography (eluent: 5% MeOH in DCM) to give 109 (57 mg).
  • Step 7 l-(2-((2S,4R)-2-((2'-Chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-5-methyl-lH-thieno[3,2-c]pyrazole-3-carboxamide (113)
  • Step 1 Ethyl l-(2-(tert-butoxy)-2-oxoethyl)-4,5,6,7-tetrahydro-lH-indazole-3-carboxylate (104b)
  • Step 2 2-(3-(Ethoxycarbonyl)-4,5,6,7-tetrahydro-lH-indazol-l-yl)acetic acid (104d)
  • Step 3 Ethyl l-(2-((2S,4R)-2-((2'-chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-4,5,6,7-tetrahydro-lH-indazole-3-carboxylate (104e)
  • Step 4 l-(2-((2S,4R)-2-((2'-Chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-4,5,6,7-tetrahydro-lH-indazole-3-carboxylic acid (104f)
  • Step 5 l-(2-((2S,4R)-2-((2'-Chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-4,5,6,7-tetrahydro-lH-indazole-3-carboxamide (104)
  • Acid 104f (252 mg, 0.46 mmol) from Step 4 was mixed with NH 4 C1 (125 mg, 2.32 mmol) in DMF (3 mL). To this solution was added HATU (262 mg, 0.69 mmol) followed by dropwise addition of DIEA (1.38 mmol, 0.24 mL). The mixture was stirred for 3 h at rt and the volatiles were removed under reduced pressure. The remaining residue was diluted with 10%> aq sodium carbonate (15 mL) and water (15 mL), and then extracted with ethyl acetate (3 ⁇ 25 mL). The combined organic solution was washed with water and brine, and then dried over MgS0 4 .
  • Step 1 Methyl 2-(3-acetyl-8-(3-chlorophenyl)-lH-pyrazolo[4,3-g]indolizin-l-yl)acetate (105c)
  • Step 3 (2S,4R)-l-(2-(3-Acetyl-8-(3-chlorophenyl)-lH-imidazo[l,2-a]pyrazolo[3,4-c]pyridin- l-yl)acetyl)-N-(6-bromopyridin-2-yl)-4-fluoropyrrolidine-2-carboxamide (105)
  • Step 5 l-(2-((2S,4R)-2-((2'-Chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-7-cyano-lH-indazole-3-carboxamid (111)
  • Step 1 l-(3'-Amino-6-chloro-2'-fluoro-[l,l'-biphenyl]-3-yl)ethanone (117c)
  • Step 3 (2S,4R)-N-(5 '- Acetyl-2 '-chloro-2-fluoro- [1 , 1 '-biphenyl] -3-yl)-4-fluoropyrrolidine-2- carboxamide TFA salt (117e)
  • Step 4 l-(2-((2S,4R)-2-((5'-Acetyl-2'-chloro-2-fluoro-[l,l , -biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-lH-indazole-3-carboxamide (117)
  • Step 1 tert-Butyl 2-(4-bromo-3-carbamoyl-lH-pyrazol-l-yl)acetate (110b)
  • Step 2 tert-Butyl 2-(3-carbamoyl-4-(2-methoxypyrimidin-5-yl)-lH-pyrazol-l-yl)acetate (110c)
  • Step 4 l-(2-((2S,4R)-2-((2'-Chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-4-(2-methoxypyrimidin-5-yl)-lH-pyrazole-3- carboxamide (110)
  • Step 2 (1R,3 S,5R)-2-(2-(4-Bromo-3-carbamoyl- lH-pyrazol- l-yl)acetyl)-N-(2 '-chloro-2- fluor 0- [1,1 '-biphenyl] -3-yl)-2-azabicyclo [3.1.0] hexane-3-carboxamide (77)
  • the title compound 77 (50 mg) was prepared from 2-(4-bromo-3 -carbamoyl- 1H- pyrazol-l-yl)acetic acid 77a and (lR,3S,5R)-N-(2 , -chloro-2-fluoro-[l,l , -biphenyl]-3-yl)-2- azabicyclo[3.1.0]hexane-3-carboxamide hydrochloride (131 mg) in a manner similar to that described in Scheme 5.
  • the reaction mixture was cooled to rt and the volatiles were removed under reduced pressure. The remaining residue was purified by column chromatography. The desired product fractions were collected and concentrated, and then the HC1 salt 80c was made by treatment with HCl/MeOH. 13.1 g of 80c was obtained.
  • Step 3 (2S,4R)-tert-Butyl l-iil'-chloro-l ⁇ S'-trifluoro-Il ⁇ '-biphen ll-S- carbamo l) ⁇ - fluoropyrrolidine-l-carboxylate (80d)
  • Step 4 (2S,4R)-N-(2'-Chloro-2,4',5 , -trifluoro-[l,l , -biphenyl]-3-yl)-4-fluoropyrrolidine-2- carboxamide hydrochloride (80e)
  • tert-Butyl 2-(3 -carbamoyl- lH-indazol-l-yl)acetate 80g (1.0 g) was taken in 4 N HC1 in dioxane (10 mL) and the resulting reaction mixture was stirred at rt for 2 h. The solvent was then removed under reduced pressure and the remaining residue 80h was used directly without further purification.
  • Step 7 l-il-iilS ⁇ Ri-l-iil'-Chloro-l ⁇ S'-trifluoro-Il ⁇ '-biphenyll-S-y carbamoyl) ⁇ - fluoropyrrolidin-l-yl)-2-oxoethyl)-lH-indazole-3-carboxamide (80)
  • Table 1 shows illustrative compounds of Formula I with characaterizing data.
  • the assay of Example 8 was used to determine the ICso's of the compounds.
  • Other standard factor D inhibition assays are also available.
  • Three ***s are used to denote compounds with an IC 50 less than 1 micromolar; two **s indicate compound with an IC 50 between 1 micromolar and 10 micromolar, and one * denotes compounds with an IC 50 greater than 10 micromolar.
  • Human factor D (purified from human serum, Complement Technology, Inc.) at 80 nM final concentration is incubated with test compound at various concentrations for 5 minutes at room temperature in 50 mM Tris, 1M NaCl, pH 7.5.
  • a synthetic substrate Z-L-Lys- SBzl and DTNB (Ellman's reagent) are added to final concentrations of 100 ⁇ each.
  • the increase in color is recorded at OD405 nm in a microplate in kinetic mode over 30 minutes with 30 second time points in a spectra fluorimeter.
  • IC50 values are calculated by non- linear regression from the percentage of inhibition of complement factor D activity as a function of test compound concentration.
  • the hemolysis assay was previously described by G. Ruiz-Gomez, et al, J. Med. Chem. (2009) 52: 6042-6052.
  • red blood cells RBC
  • rabbit erythrocytes purchased from Complement Technologies
  • GVB Buffer 0.1 % gelatin, 5 mM Veronal, 145 mM NaCl, 0.025 % NaN 3 , pH 7.3
  • Cells are used at a concentration of 1 x 10 8 cells/mL.
  • the optimum concentration of Normal Human Serum (NHS) needed to achieve 100% lysis of rabbit erythrocytes is determined by titration.
  • NHS (Complement Technologies) is incubated with inhibitor for 15 min at 37 °C, rabbit erythrocytes in buffer were added and incubated for an additional 30 min at 37 °C.
  • Positive control (100% lysis) consists of serum and RBC and negative control (0% lysis) of Mg-EGTA buffer and RBC only. Samples are centrifuged at 2000g for 5 min, and supernatants collected. Optical density of the supernatant is monitored at 405 nm using a UV/visible spectrophotometer. Percentage lysis in each sample is calculated relative to positive control (100% lysis).

Abstract

Compounds, methods of use, and processes for making inhibitors of complement factor D comprising Formula I, or a pharmaceutically acceptable salt or composition thereof are provided. The inhibitors described herein target factor D and inhibit or regulate the complement cascade at an early and essential point in the alternative complement pathway, and reduce factor D's ability to modulate the classical and lectin complement pathways. The inhibitors of factor D described herein are capable of reducing the excessive activation of complement, which has been linked to certain autoimmune, inflammatory, and neurodegenerative diseases, as well as ischemia- reperfusion injury and cancer.

Description

COMPOUNDS FOR TREATMENT OF
COMPLEMENT MEDIATED DISORDERS
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of provisional U.S. Application No. 61/944,189 filed February 25, 2014, provisional U.S. Application No. 62/022,916 filed July 10, 2014, and provisional U.S. Application 62/046,783 filed September 5, 2014. The entirety of each of these applications is hereby incorporated by reference for all purposes.
BACKGROUND
[0002] The complement system is a part of the innate immune system which does not adapt to changes over the course of the host's life, but is recruited and used by the adaptive immune system. For example, it assists, or complements, the ability of antibodies and phagocytic cells to clear pathogens. This sophisticated regulatory pathway allows rapid reaction to pathogenic organisms while protecting host cells from destruction. Over thirty proteins and protein fragments make up the complement system. These proteins act through opsonization (enhancing phaogytosis of antigens), chemotaxis (attracting macrophages and neutrophils), cell lysis (rupturing membranes of foreign cells) and agglutination (clustering and binding of pathogens together).
[0003] The complement system has three pathways: classical, alternative and lectin. Complement factor D plays an early and central role in activation of the alternative pathway of the complement cascade. Activation of the alternative complement pathway is initiated by spontaneous hydrolysis of a thioester bond within C3 to produce C3(H20), which associates with factor B to form the C3(H20)B complex. Complement factor D acts to cleave factor B within the C3(H20)B complex to form Ba and Bb. The Bb fragment remains associated with C3(H20) to form the alternative pathway C3 convertase C3(H20)Bb. Additionally, C3b generated by any of the C3 convertases also associates with factor B to form C3bB, which factor D cleaves to generate the later stage alternative pathway C3 convertase C3bBb. This latter form of the alternative pathway C3 convertase may provide important downstream amplification within all three of the defined complement pathways, leading ultimately to the recruitment and assembly of additional factors in the complement cascade pathway, including the cleavage of C5 to C5a and C5b. C5b acts in the assembly of factors C6, C7, C8, and C9 into the membrane attack complex, which can destroy pathogenic cells by lysing the cell.
[0004] The dysfunction of or excessive activation of complement has been linked to certain autoimmune, inflammatory, and neurodegenerative diseases, as well as ischemia- reperfusion injury and cancer. For example, activation of the alternative pathway of the complement cascade contributes to the production of C3a and C5a, both potent anaphylatoxins, which also have roles in a number of inflammatory disorders. Therefore, in some instances, it is desirable to decrease the response of the complement pathway, including the alternative complement pathway. Some examples of disorders mediated by the complement pathway include age-related macular degeneration (AMD), paroxysmal nocturnal hemoglobinuria (PNH), multiple sclerosis, and rheumatoid arthritis.
[0005] Age-related macular degeneration (AMD) is a leading cause of vision loss in industrialized countries. Based on a number of genetic studies, there is evidence of the link between the complement cascade and macular degeneration. Individuals with mutations in the gene encoding complement factor H have a fivefold increased risk of macular degeneration and individuals with mutations in other complement factor genes also have an increased risk of AMD. Individuals with mutant factor H also have increased levels of C-reactive protein, a marker of inflammation. Without adequate functioning factor H, the alternative pathway of the complement cascade is overly activated leading to cellular damage. Inhibition of the alternative pathway is thus desired.
[0006] Paroxysmal nocturnal hemoglobinuria (PNH) is a non-malignant, hematological disorder characterized by the expansion of hematopoietic stem cells and progeny mature blood cells which are deficient in some surface proteins. PNH erythrocytes are not capable of modulating their surface complement activation, which leads to the typical hallmark of PNH - the chronic activation of complement mediated intravascular anemia. Currently, only one product, the anti-C5 monoclonal antibody eculizumab, has been approved in the U.S. for treatment of PNH. However, many of the patients treated with eculizumab remain anemic, and many patients continue to require blood transfusions. In addition, treatment with eculizumab requires life-long intravenous injections. Thus, there is an unmet need to develop novel inhibitors of the complement pathway. [0007] Factor D is an attractive target for inhibition or regulation of the complement cascade due to its early and essential role in the alternative complement pathway, and its potential role in signal amplification within the classical and lectin complement pathways. Inhibition of factor D effectively interrupts the pathway and attenuates the formation of the membrane attack complex.
[0008] While initial attempts have been made to develop inhibitors of factor D, there are currently no small molecule factor D inhibitors in clinical trials. Examples of factor D inhibitors or prolyl compounds are described in the following disclosures.
[0009] Biocryst Pharmaceuticals US Pat. No. 6653340 titled "Compounds useful in the complement, coagulat and kallikrein pathways and method for their preparation" describes fused bicyclic ring compounds that are potent inhibitors of factor D. Development of the factor D inhibitor BCX1470 was discontinued due to lack of specificity and short half-life of the compound.
[0010] Novartis PCT patent publication WO2012/093101 titled "Indole compounds or analogues thereof useful for the treatment of age-related macular degeneration" describes certain factor D inhibitors.
[0011] Novartis PCT patent publications WO2014/002057 titled "Pyrrolidine derivatives and their use as complement pathway modulators" and WO2014/009833 titled "Complement pathway modulators and uses thereof describe additional factor D inhibitors with heterocyclic substituents. Additional factor D inhibitors are described in Novartis PCT patent publications WO2014/002051, WO2014/002052, WO2014/002053, WO2014/002054, WO2014/002058, WO2014/002059, and WO2014/005150.
[0012] Bristol-Myers Squibb PCT patent publication WO2004/045518 titled "Open chain prolyl urea-related modulators of androgen receptor function" describes open chain prolyl urea and thiourea related compounds for the treatment of androgen receptor-associated conditions, such as age-related diseases, for example, sarcopenia.
[0013] Japan Tobacco Inc. PCT patent publication WO 1999/048492 titled "Amide derivatives and nociceptin antagonists" describes compounds with a proline-like core and aromatic substituents connected to the proline core through amide linkages useful for the treatment of pain.
[0014] Ferring B.V. and Yamanouchi Pharmaceutical Co. 1TD. PCT patent publication WO 1993/020099 titled "CCK and/or gastrin receptor ligands" describes compounds with a proline-like core and heterocyclic substituents connected to the proline core through amide linkages for the treatment of, for example, gastric disorders or pain.
[0015] Alexion Pharmaceuticals PCT patent publication WO 1995/029697 titled "Methods and compositions for the treatment of glomerulonephritis and other inflammatory diseases" discloses antibodies directed to C5 of the complement pathway for the treatment of glomerulonephritis and inflammatory conditions involving pathologic activation of the complement system. Alexion Pharmaceutical's anti-C5 antibody eculizumab (Soliris®) is currently the only complement-specific antibody on the market, and is the first and only approved treatment for paroxysmal nocturnal hemoglobinuria (PNH).
[0016] Compounds which mediate the complement pathway, and for example, act as factor D inhibitors are needed for treatment of disorders in a host, including a human, associated with misregulation of the complement cascade.
SUMMARY
[0017] According to the present invention, a compound of Formula I is provided:
Figure imgf000005_0001
[0018] as well as the pharmaceutically acceptable salts and compositions thereof. Formula I can be considered to have a central core, an L-B substituent, and a (C=0)A substituent. It has been discovered that a compound of Formula I, or a pharmaceutically acceptable salt or composition thereof, is a superior inhibitor of complement factor D, and therefore can be used as an effective amount to treat a host in need of complement factor D modulation. As described below, the invention provides compounds of Formula I, wherein either X2 is nitrogen or at least one of (d), (e), (g), (i), (1), (n), (p), (s), (v), (x), and (y) is present, as defined below. Pharmaceutical compositions comprising a compound or salt of Formula I together with a pharmaceutically acceptable carrier are also disclosed. [0019] In one embodiment, a method for the treatment of a disorder associated with a dysfunction, including increased activity, of the complement pathway is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier, as described in more detail below.
[0020] In one embodiment, the disorder is associated with the alternative complement cascade pathway. In yet another embodiment, the disorder is associated with the complement classical pathway. In a further embodiment, the disorder is associated with the complement lectin pathway. The factor D inhibitors provided herein can thus dampen or inhibit detrimental complement activity in a host, by administration of an effective amount in a suitable manner to a host in need thereof.
[0021] Specific embodiments of this invention are directed to certain disease indications. In one embodiment, a method for the treatment of paroxysmal nocturnal hemoglobinuria (PNH) is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. In another embodiment, a method for the treatment of age-related macular degeneration (AMD) is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. In another embodiment, a method for the treatment of rheumatoid arthritis is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. In another embodiment, a method for the treatment of multiple sclerosis is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
[0022] In other embodiments of the invention, an active compound provided herein can be used to treat or prevent a disorder in a host mediated by complement factor D, or by an excessive or detrimental amount of the C3 amplification loop of the complement pathway. As examples, the invention includes methods to treat or prevent complement associated disorders that are induced by antibody-antigen interactions, a component of an immune or autoimmune disorder or by ischemic injury. The invention also provides methods to decrease inflammation or an immune response, including an autoimmune response, where mediated or affected by factor D.
[0023] Formula I carries variables, e.g, A, B, L, X1, X2, Q1, Q2, and Q3, with the following definitions.
[0024] Q1 is N(RJ) or QRV).
[0025] Q2 is C(R2R2'), C(R2R2')-C(R2R2'), S, O, N(R2) or C(R2R2')0.
[0026] Q3 is N(R3), S, or C(R3R3').
[0027] (a) X1 and X2 are independently N, CH, or CZ, or (b) X1 and X2 together are C=C.
[0028] Q1, Q2, Q3, X1, and X2 are selected such that a stable compound results.
[0029] Non-limiting examples of the
Figure imgf000007_0001
ring are illustrated below (any of which c
Figure imgf000007_0002
Figure imgf000008_0001
[0030] R and R' are independently chosen from H, alkyl, cycloalkyl, alkylcycloalkyl, cycloalkylalkyl, heterocycle, heterocycloalkyl, aryl, aralkyl, heteroaryl, heteroarylalkyl wherein each group can be optionally substituted or any other substituent group herein that provides the desired properties. In some embodiments, the ring includes one or more chiral carbon atoms. The invention includes embodiments in which the chiral carbon can be provided as an enantiomer, or mixtrues of enantiomers, including a racemic mixture. Where the ring includes more than one stereocenter, all of the enantiomers and diastereomers are included in the invention as individual species.
[0031] Z is F, CI, NH2, CH3, CH2D, CHD2, or CD3.
1 1 ' 2 2 ' 3 3'
[0032] R , R , R , R , R , and R are independently chosen at each occurence from (c) and (d):
(c) hydrogen, halogen, hydroxyl, nitro, cyano, amino, Ci-C6alkyl, C2-C6alkenyl, Ci- C6alkoxy, C2-C6alkynyl, C2-C6alkanoyl, Ci-C6thioalkyl, hydroxyCi-C6alkyl, aminoCi-C6alkyl, -Co-C4alkylNR9R10, -C(0)OR9, -OC(0)R9, -NR9C(0)R10, -C(0)NR9R10, -OC(0)NR9R10, -O(heteroaryl), -NR9C(0)OR10, Ci-C2haloalkyl, and Ci-C2haloalkoxy, where R9 and R10 are independently chosen at each occurrence from hydrogen, Ci-C6alkyl, and (C3-C7cycloalkyl)Co- C4alkyl;
(d) -C0-C4alkyl(C3-C7cycloalkyl) and -O-C0-C4alkyl(C3-C7cycloalkyl).
[0033] In alternative embodiments, any one of the following rings (e), (f), (g), (h), (i), or j) may be present: (e) R1 and R1 or R3 and R3 may be taken together to form a 3- to 6-membered carbocyclic spiro ring or a 3- to 6-membered heterocyclic spiro ring containing 1 or 2 heteroatoms independently chosen from N, O, or S;
(f) R2 and R2 may be taken together to form a 3- to 6-membered carbocyclic spiro ring,
(g) R2 and R2 may be taken together to form a 3- to 6-membered heterocyclic spiro ring, each of which spiro rings (e), (f), and (g) is unsubstituted or substituted with 1 or more substituents independently chosen from halogen (and in particular F), hydroxyl, cyano, -COOH, Ci-C4alkyl (including in particular methyl), C2-C4alkenyl, C2-C4alkynyl, Ci-C4alkoxy, C2- C4alkanoyl, hydroxyCi-C4alkyl, (mono- and di-Ci-C4alkylamino)Co-C4alkyl, -Co-C4alkyl(C3- C7cycloalkyl), -0-Co-C4alkyl(C3-C7cycloalkyl), Ci-C2haloalkyl, and Ci-C2haloalkoxy.
(h) R1 and R2 may be taken together to form a 3-membered carbocyclic ring;
(i) R1 and R2 may be taken together to form a 4- to 6-membered carbocyclic ring or a 4- to 6-membered heterocyclic ring containing 1 or 2 heteroatoms independently chosen from N, O, and S.
(j) R2 and R3, if bound to adjacent carbon atoms, may be taken together to form a 3- to 6- membered carbocyclic ring or a 3- to 6-membered heterocyclic ring; each of which ring (h), (i), and (j) may be unsubstituted or substituted with 1 or more substituents independently chosen from halogen (and in particular F), hydroxyl, cyano, -COOH, Ci-C4alkyl (including in particular methyl), C2-C4alkenyl, C2-C4alkynyl, Ci-C4alkoxy, C2-C4alkanoyl, hydroxyCi-C4alkyl, (mono- and di-Ci-C4alkylamino)Co-C4alkyl, -Co-C4alkyl(C3-C7cycloalkyl), -0-Co-C4alkyl(C3- C7cycloalkyl), Ci-C2haloalkyl, and Ci-C2haloalkoxy.
1 1 ' 2 2' 3 3'
[0034] In alternative embodiments, R and R , R and R , or R and R can be taken together to form a carbonyl group. In alternative embodiments, R1 and R2 or R2 and R3 can be taken together to form a carbon-carbon double bond.
[0035] A is a group chosen from (k) and (1) where (k) is
Figure imgf000009_0001
and (1) is
Figure imgf000010_0001
[0036] X4 is B(OH) and Y is CHR9; or X 4 is CHR9 and Y is B(OH).
[0037] R101 is hydrogen, alkyl, carboxy.
[0038] R4 is (m) or (n):
(m) -CHO, -CONH2, or C2-C6alkanoyl, including C(0)C3-C7cycloalkyl;
(n) hydrogen, -S02NH2, -C(CH2)2F, -CH(CF3)NH2, Ci-C6alkyl, -C0-C4alkyl(C3- C7cycloalkyl), -C(O)C0-C2alkyl(C3-C7cycloalkyl), Ηΐί Ν Η H I N Η Ν ^ΙΜ °¾ 0ΝΗ or °¾ Η
each of which R4 other than hydrogen, -CHO, and -CONH2, is unsubstituted or substituted with one or more of amino, imino, halogen, hydroxyl, cyano, cyanoimino, Ci-C2alkyl, Ci-C2alkoxy, -Co-C2alkyl(mono- and di-Ci-C4alkylamino), Ci-C2haloalkyl, and Ci-C2haloalkoxy.
[0039] R5 and R6 are independently chosen from (o) and (p):
(o) -CHO, -C(0)NH2, -C(0)NH(CH3), or C2-C6alkanoyl;
(p) hydrogen, hydroxyl, halogen, cyano, nitro, -COOH, -S02NH2, -C(NH2)Ci-C3alkyl, -C(NH2)C C3haloalkyl, -CF(C=CH2), -C(=NCN)Ci-C6alkyl, Ci-C6alkyl, C2-C6alkenyl, C C6alkoxy, -Co-C4alkyl(C3-C7cycloalkyl), -C(O)C0-C4alkyl(C3-C7cycloalkyl), -P(0)(OR9)2, - OC(0)R9, -C(0)OR9, -C(0)N(CH2CH2R9)(R10), -NR9C(0)R10, phenyl, or 5- to 6-membered heteroaryl.
[0040] Each R5 and R6 other than hydrogen, hydroxyl, cyano, and -COOH is unsubstituted or optionally substituted. For example, R5 and R6 other than hydrogen, hydroxyl, cyano, and -COOH may be substituted with one or more substituents independently chosen from halogen, hydroxyl, amino, imino, cyano, cyanoimino, Ci-C2alkyl, Ci-C4alkoxy, -Co- C2alkyl(mono- and di-Ci-C4alkylamino), Ci-C2haloalkyl, Ci-C2haloalkoxy, C(0)alkyl, C(0)cycloalkyl, C(0)aryl, C(0)heterocycle, and C(0)heteroaryl.
[0041] R6 is hydrogen, halogen, hydroxyl, Ci-C4alkyl, or Ci-C4alkoxy; or R6 and R6 may be taken together to form an oxo, vinyl, or imino group.
[0042] R7 is hydrogen, Ci-C6alkyl, or -C0-C4alkyl(C3-C7cycloalkyl).
[0043] In an alternate embodiment, two A groups can be bonded together to form a dimer through a suitable linking group that achieves the desired purpose. Examples of linking groups include but are not limited to, urea, amide, -C(0)-C(0)-, carbamate and ketone. In one embodiment, two heteroaryl rings, for example, two indole rings, are linked through a urea to form the dimer.
[0044] R8 and R8 are independently chosen from hydrogen, halogen, hydroxyl, Ci- C6alkyl, Ci-C6alkoxy, and (Ci-C4alkylamino)Co-C2alkyl, or R8 and R8 are taken together to form an oxo group, or can be taken together with the carbon that they are bonded to form a 3- membered carbocyclic ring.
[0045] R16 is 0 or 1 or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, Ci-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, Ci-C6alkoxy, -Co-C4alkyl(mono- and di- Ci-Cealkylamino), -Co-C4alkyl(C3-Cycycloalkyl), Ci-C2haloalkyl, and Ci-C2haloalkoxy.
[0046] R19 is hydrogen, C C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, -S02Ci-C6alkyl, (mono- and di-Ci-C6alkylamino)Ci-C4alkyl, -Co-C4alkyl(C3-Cycycloalkyl), -Co-C4alkyl(C3- Cyheterocycloalkyl), -C0-C4alkyl(aryl), C0-C4alkyl(heteroaryl), and -C(0)(CH2)i-2C(0)OR9 each of which R19 other than hydrogen is substituted with 0 or 1 or more substituents independently chosen from halogen, hydroxyl, amino, -COOH, and -C(0)OCi-C4alkyl.
[0047] X11 is N or CR11.
[0048] X12 is N or CR12.
[0049] X13 is N or CR13.
[0050] X14 is N or CR14.
[0051] No more than two of X11, X12, X13, and X14 are N.
[0052] R11, R14, and R15 are independently chosen at each occurrence from hydrogen, halogen, hydroxyl, nitro, cyano, -NR9C(0)R10, C(0)NR9R10, -0(PO)(OR9)2, -(PO)(OR9)2, d- C6alkyl, C2-C6alkenyl, C2-C6alkenyl(aryl), C2-C6alkenyl(cycloalkyl), C2-C6alkenyl(heterocycle), C2-C6alkenyl(heteroaryl), C2-C6alkynyl, C2-C6alkynyl(aryl), C2-C6alkynyl(cycloalkyl), C2- C6alkynyl(heterocycle), C2-Cealkynyl(heteroaryl), C2-C6alkanoyl, Ci-C6alkoxy, Ci-C6thioalkyl, - Co-C4alkyl(mono- and di-Ci-Cealkylamino), -Co-C4alkyl(C3-Cycycloalkyl), (phenyl)Co-C4alkyl, (4- to 7-membered heterocycloalkyl)Co-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S, and (5- or 6- membered unsaturated heterocycle or heteroaryl)C0- C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S), -Co-C4alkoxy(C3- Cycycloalkyl), Ci-C2haloalkyl, and Ci-C2haloalkoxy.
[0053] Alternatively, R13 and R14 can together form a bridge that can be an optionally substituted cycloalkyl, heterocycle or heteroaryl ring.
[0054] R12 and R13 are independently chosen at each occurrence from (q), (r) and (s).
(q) hydrogen, halogen, hydroxyl, nitro, cyano, amino, -COOH, Ci-C2haloalkyl,
Ci-C2haloalkoxy; (r) Ci-C6alkyl, -Co-C4alkyl(C3-Cycycloalkyl), C2-C6alkenyl, C2-C6alkanoyl, Ci-C6alkoxy, C2-C6alkenyloxy, -C(0)OR9, Ci-C6thioalkyl, -C0-C4alkylNR9R10, -C(0)NR9R10, -S02R9, - S02NR9R10, -OC(0)R9, and -C(NR9)NR9R10 each of which (r) is unsubstituted or substituted with one or more substituents independently selected from halogen, hydroxyl, nitro, cyano, amino, -COOH, -CONH2? C Cjhaloalkyl, and C Cjhaloalkoxy, and each of which (r) is also optionally substituted with one substituent chosen from phenyl and 4- to 7- membered heterocycle containing 1, 2, or 3 heteroatoms independently chosen from N, O, and S; which phenyl or 4- to 7-membered heterocycle is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, CrC6alkyl, C2- C6alkenyl, C2-C6alkanoyl, CrC6alkoxy, (mono- and di-CrC6alkylamino)C0-C4alkyl, Cr C6alkylester, -(C0-C4alkyl)(C3-C7cycloalkyl), CrC2haloalkyl, and CrC2haloalkoxy;
(s) -C(CH2)2R30.
[0055] R30 is -NR9C(0)R31 or R32.
[0056] R31 and R32 are each independently selected from Ci-C6alkyl, Ci-C6haloalkyl, C2- C6alkenyl, C2-C6alkynyl, (C3-Cvcycloalkyl)Co-C4alkyl, (aryl)Co-C4alkyl, (heterocycle)Co-C4alkyl and (heteroaryl)Co-C4alkyl wherein each group can be optionally substituted; or each of which (s) may be unsubstituted or substituted with one or more substituents independently chosen from, but not limited to, halogen, hydroxyl, nitro, cyano, amino, oxo, -B(OH)2, -Si(CH3)3, - COOH, -CONH2, -P(0)(OH)2, Ci-C6alkyl, Ci-C6alkoxy, -C0-C2alkyl(mono- and di-C C4alkylamino), Ci-Cealkylester, Ci-C4alkylamino, Ci-C4hydroxylalkyl, Ci-C2haloalkyl, and Ci- C2haloalkoxy.
[0057] L is either (t), (u), or (v):
(t) is a group of the formula
O R18 R18' R18 RI B' 0 R18 R18'
R17 , R17 0 , or OH where R17 is hydrogen or Ci-C6alkyl and R18 and R18 are independently chosen from hydrogen, halogen, hydroxymethyl, and methyl; and m is 0, 1, 2, or 3;
(u) is a bond,
(v) or a directly linked optionally substitiuted alkyl, alkyl(heteroaryl), heterocyclic aryl, heteroaryl, moiety including but not limited to:
Figure imgf000014_0001
[0058] B is a monocyclic, bicyclic carbocyclic or carbocyclic-oxy group or a monocyclic, bicyclic, or tricyclic heterocyclic group having 1 , 2, 3, or 4 heteroatoms independently selected from N, O, and S and from 4 to 7 ring atoms per ring, or B is a C2- C6alkenyl or C2-C6alkynyl group, or B is -(Co-C4alkyl)(aryl), -(Co-C4alkyl)(heteroaryl), or -(C0- C4alkyl)(biphenyl) .
[0059] Each of which B is unsubstituted or substituted with one or more substituents independently chosen from (w) and (x) and 0 or 1 substituents chosen from (y) and (z):
(w) halogen, hydroxyl, -COOH, cyano, Ci-C6alkyl, C2-C6alkanoyl, Ci-C6alkoxy, -C0- C4alkylNR9R10, -S02R9, Ci-C2haloalkyl, and Ci-C2haloalkoxy;
(x) nitro, C2-C6alkenyl, C2-C6alkynyl, Ci-C6thioalkyl, -JC3-C7cycloalkyl, -B(OH)2, -JC(0)NR9R23,-JOS02OR21, -C(0)(CH2)i_4S(0)R21, SR9 -0(CH2)i_4S(0)NR21R22
-JOP(0)(OR21)(OR22), -JP(0)(OR21)(OR22), -JOP(0)(OR21)R22, -JP(0)(OR21)R22, -JOP(0)R21R22, -JP(0)R21R22, -JSP(0)(OR21)(OR22), -JSP(0)(OR21)(R22), -JSP(0)(R21)(R22), -JNR9P(0)(NHR21)(NHR22), -JNR9P(0)(OR21)(NHR22), -JNR9P(0)(OR21)(OR22), -JC(S)R21, -JNR21S02R22, -JNR9S(O)NR10R22, -JNR9SO2NR10R22, -JS02NR9COR22, -JS02NR9CONR21R22, -JNR21S02R22, -JC(0)NR21S02R22, -JC(NH2)NR22, -JC(NH2)NR9S(0)2R22, -JOC(0)NR21R22, -JNR21C(0)OR22, -JNR21OC(0)R22, -(CH2)i_4C(0)NR21R22, -JC(0)R24R25, -JNR9C(0)R21, -JC(0)R21, -JNR9C(O)NR10R22, -CCR21, -(CH2)i_4OC(0)R21, and -JC(0)OR23; each of which (x) may be unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, amino, oxo, -B(OH)2, -Si(CH3)3, -COOH, -CONH2, -P(0)(OH)2, Ci-C6alkyl, Ci-C6alkoxy, -Co-C2alkyl(mono- and di-Ci-C4alkylamino), Ci- C6alkylester, Ci-C4alkylamino, Ci-C4hydroxylalkyl, Ci-C2haloalkyl, and Ci-C2haloalkoxy;
(y) naphthyl, naphthyloxy, indanyl, (4- to 7-membered heterocycloalkyl)Co-C4alkyl containing 1 or 2 heteroatoms chosen from N, O, and S, and bicyclic heterocycle containing 1 , 2, or 3 heteroatoms independently chosen from N, O, and S, and containing 4- to 7- ring atoms in each ring; each of which (y) is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, Ci-C6alkyl, C2-C6alkenyl, C2- C6alkanoyl, Ci-C6alkoxy, (mono- and di-Ci-C6alkylamino)Co-C4alkyl, Ci-C6alkylester, -C0- C4alkyl(C3-C7cycloalkyl), -S02R9, Ci-C2haloalkyl, and Ci-C2haloalkoxy; and
(z) tetrazolyl, (phenyl)Co-C2alkyl, (phenyl)Ci-C2alkoxy, phenoxy, and 5- or 6-membered heteroaryl containing 1 , 2, or 3 heteroatoms independently chosen from N, O, B, and S, each of which (z) is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, Ci-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, Ci-C6alkoxy, (mono- and di-Ci-C6alkylamino)Co-C4alkyl, Ci-Cealkylester, -Co-C4alkyl(C3-Cvcycloalkyl), -S02R9, -OSi(CH3)2C(CH3)3, -Si(CH3)2C(CH3)3, Ci-C2haloalkyl, and Ci-C2haloalkoxy.
[0060] J is independently chosen at each occurrence from a covalent bond, Ci- C4alkylene, -OCi-C4alkylene, C2-C4alkenylene, and C2-C4alkynylene.
[0061] R21 and R22 are independently chosen at each occurrence from hydrogen, hydroxyl, cyano, amino, Ci-C6alkyl, Ci-C6haloalkyl, Ci-C6alkoxy, (C3-Cvcycloalkyl)Co-C4alkyl, (phenyl)C0-C4alkyl, -Ci-C4alkylOC(0)OCi-C6alkyl, -C C4alkylOC(0)Ci-C6alkyl, -C C4alkylC(0)OCi-C6alkyl, (4- to 7-membered heterocycloalkyl)Co-C4alkyl having 1 , 2, or 3 heteroatoms independently chosen from N, O, and S, and (5- or 6- membered unsaturated or aromatic heterocycle)Co-C4alkyl having 1 , 2, or 3 heteroatoms independently chosen from N, O, and S.
[0062] R23 is independently chosen at each occurrence from (C -C7cycloalkyl)C0- C4alkyl, (phenyl)Co-C4alkyl, (4- to 7-membered heterocycloalkyl)Co-C4alkyl having 1 , 2, or 3 heteroatoms independently chosen from N, O, and S, and (5- or 6- membered unsaturated or aromatic heterocycle)Co-C4alkyl having 1 , 2, or 3 heteroatoms independently chosen from N, O, and S.
[0063] R24 and R25 are taken together with the nitrogen to which they are attached to form a 4- to 7-membered monocyclic heterocycloalkyl group, or a 6- to 10- membered bicyclic heterocyclic group having fused, spiro, or bridged rings.
[0064] Either X2 is nitrogen or at least one of (d), (e), (g), (i), (1), (n), (p), (s), (v), (x), and (y) is present. Pharmaceutical compositions comprising a compound or salt of Formula I together with a pharmaceutically acceptable carrier are also disclosed.
[0065] Methods of treating or preventing disorders mediated by complement cascade factor D, including but not limited to age-related macular degeneration (AMD), retinal degeneration, other ophthalmic diseases (e.g., geographic atrophy), paroxysymal nocturnal hemoglobinuria (PNH), multiple sclerosis (MS), arthritis including rheumatoid arthritis (RA), a respiratory disease or a cardiovascular disease, are provided, comprising administering a therapeutically effective amount of a compound or salt of Formula I to a host, including a human, in need of such treatment are also disclosed.
[0066] In another embodiment, an effective amount of an active factor D inhibiting compound is provided to treat an inflammatory or immune disorder, including an autoimmune disorder, that is meadited or affected by factor D. In an alternative embodiment, the compound of Formula I can be used to treat a disorder mediated by the complement pathway, regardless whether it is acting through Factor D.
[0067] The present invention includes at least the following features:
(a) a compound of Formula I as described herein, and pharmaceutically acceptable salts and prodrugs thereof (each of which and all subgenuses and species thereof considered individually and specifically described);
(b) Formula I as described herein, and pharmaceutically acceptable salts and prodrugs thereof, for use in treating or preventing disorders mediated by the complement pathway, and for example, cascade factor D, including age-related macular degeneration (AMD), retinal degeneration, paroxysymal nocturnal hemoglobinuria (PNH), multiple sclerosis (MS), and rheumatoid arthritis (RA) and other disorders described further herein;
(c) use of Formula I, and pharmaceutically acceptable salts and prodrugs thereof in the manufacture of a medicament for use in treating or preventing disorders mediated by complement cascade factor D, including age-related macular degeneration (AMD), retinal degeneration, paroxysymal nocturnal hemoglobinuria (PNH), multiple sclerosis (MS), and rheumatoid arthritis (RA) and other disorders described further herein;
(d) a process for manufacturing a medicament intended for the therapeutic use for treating or preventing treating or preventing disorders mediated by complement cascade factor D, including age-related macular degeneration (AMD), retinal degeneration, paroxysymal nocturnal hemoglobinuria (PNH), multiple sclerosis (MS), and rheumatoid arthritis (RA) and other disorders described further herein characterized in that Formula I as described herein is used in the manufacture; (e) a pharmaceutical formulation comprising an effective host-treating amount of the Formula I or a pharmaceutically acceptable salt or prodrug thereof together with a pharmaceutically acceptable carrier or diluent;
(f) Formula I as described herein in substantially pure form, including substantially isolated from other chemical entities (e.g., at least 90 or 95%);
(g) processes for the manufacture of the compounds of Formula I and salts, compositions, dosage forms thereof; and
(h) processes for the preparation of therapeutic products that contain an effective amount of Formula I, as described herein.
DETAILED DESCRIPTION
I. TERMINOLOGY
[0068] Compounds are described using standard nomenclature. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this invention belongs.
[0069] The compounds in any of the Formulas described herein include enantiomers, mixtures of enantiomers, diastereomers, tautomers, racemates and other isomers, such as rotamers, as if each is specifically described. "Formula I" includes all subgeneric groups of Formula I, such as Formula IA and Formula IB and also includes pharmaceutically acceptable salts of a compound of Formula I, unless clearly contraindicated by the context in which this phrase is used. "Formula I" also includes all subgeneric groups of Formula I, such as Formulas IC - ID, and Formulas II - XXX, and also includes pharmaceutically acceptable salts of all subgeneric groups of Formula I, such as Formulas IA - ID, and Formulas II - XXX, unless contraindicated by the context in which this phrase is used.
[0070] The terms "a" and "an" do not denote a limitation of quantity, but rather denote the presence of at least one of the referenced item. The term "or" means "and/or". Recitation of ranges of values are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. The endpoints of all ranges are included within the range and independently combinable. All methods described herein can be performed in a suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of examples, or exemplary language (e.g., "such as"), is intended merely to better illustrate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. Unless defined otherwise, technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this invention belongs.
[0071] The present invention includes compounds of Formula I and the use of compounds with at least one desired isotopic substitution of an atom, at an amount above the natural abundance of the isotope, i.e., enriched. Isotopes are atoms having the same atomic number but different mass numbers, i.e., the same number of protons but a different number of neutrons.
[0072] Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as 2H, 3H, UC, 13C, 14C, 15N, 18F, 31P, 32P, 35S, 36CI, 125I respectively. The invention includes various isotopically labeled compounds as defined herein, for example those into which radioactive isotopes, such as 3H, 13C, and 14C, are present. Such isotopically labelled compounds are useful in metabolic studies (with 14C), reaction kinetic studies (with, for example 2H or 3H), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients. In particular, an 18F labeled compound may be particularly desirable for PET or SPECT studies. Isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
[0073] By way of general example and without limitation, isotopes of hydrogen, for example, deuterium (2H) and tritium (3H) may be used anywhere in described structures. Alternatively or in addition, isotopes of carbon, e.g., 13C and 14C, may be used. A typical isotopic substitution is deuterium for hydrogen at one or more locations on the molecule to improve the performance of the drug, for example, the pharmacodynamics, pharmacokinetics, biodistribution, half-life, stability, AUC, Tmax, Cmax, etc. For example, the deuterium can be bound to carbon in a location of bond breakage during metabolism (an a-deuterium kinetic isotope effect) or next to or near the site of bond breakage (a β-deuterium kinetic isotope effect). [0074] Isotopic substitutions, for example deuterium substitutions, can be partial or complete. Partial deuterium substitution means that at least one hydrogen is substituted with deuterium. In certain embodiments, the isotope is 90, 95 or 99% or more enriched in an isotope at any location of interest. In one embodiments deuterium is 90, 95 or 99% enriched at a desired location. Unless otherwise stated, the enrichment at any point is above natural abundance and enough to alter a detectable property of the drug in a human.
[0075] In one embodiment, the substitution of a hydrogen atom for a deuterium atom occurs within an R group substituent on the L-B moiety region. In one embodiment, the substitution of a hydrogen atom for a deuterium atom occurs within an R group selected from any of R 18 , R 18' , R 33 , R 34 , R 35 , and/or R 36. In one embodiment, the substitution of a hydrogen atom for a deuterium atom occurs within an R group substituent within the A-carbonyl moiety region. In one embodiment, the substitution of a hydrogen atom for a deuterium atom occurs at R4< R5, R6, R6 , R7, R8, R8', R11, R12, R13, R14, R15, R16, R19, R21, R22, R23, and R30. In other embodiments, certain substituents on the proline ring are selectively deuterated. For example, in one embodiment, the substitution of a hydrogen atom for a deuterium atom occurs at R, R , R1, R1', R2, R2', R3, and/or R3'. In one embodiment, for example, when any of the R substituents of the proline ring are methyl or methoxy, the alkyl residue is optionally deuterated, e.g., CD3 or OCD3. In certain other embodiments, when two substituents of the proline ring are combined to form a cyclopropyl ring, the unsubstituted methylene carbon is deuterated.
[0076] In one embodiment, the substitution of a hydrogen atom for a deuterium atom occurs within an R group when at least one of the variables within the R group is hydrogen (e.g., 2H or D) or alkyl (e.g., CD3). For example, when any of R groups are, or contain for example through substitution, methyl or ethyl, the alkyl residue is typically deuterated, e.g., CD3i CH2CD3 or CD2CD3. In certain other embodiments, when any of the above mentioned R groups are hydrogen, the hydrogen may be isotopically enriched as deuterium (i.e., 2H).
[0077] The compounds of the present invention may form solvates with solvents (including water). The term "solvate" refers to a molecular complex of a compound of the present invention (including salts thereof) with one or more solvent molecules. Solvents include water, ethanol, dimethyl sulfoxide, acetone and other common organic solvents. The term "hydrate" refers to a molecular complex comprising a compound of the invention and water. Pharmaceutically acceptable solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D20, d6-acetone, d6-DMSO. A solvate can be in a liquid or solid form.
[0078] A dash ("-") that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, -(C=0)NH2 is attached through carbon of the keto (C=0) group.
[0079] The term "substituted", as used herein, means that any one or more hydrogens on the designated atom or group is replaced with a moiety selected from the indicated group, provided that the designated atom's normal valence is not exceeded. For example, when the substituent is oxo (i.e., =0) then two hydrogens on the atom are replaced. When an oxo group replaces two hydrogens in an aromatic moiety, the corresponding partially unsaturated ring replaces the aromatic ring. For example a pyridyl group substituted by oxo is a pyridone. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds or useful synthetic intermediates.
[0080] A stable compound or stable structure refers to a compound leading to a compound that can be isolated and can be formulated into a dosage form with a shelf life of at least one month.
[0081] Any suitable group may be present on a "substituted" or "optionally substituted" position that forms a stable molecule and advances the desired purpose of the invention and includes, but is not limited to, e.g., halogen (which can independently be F, CI, Br or I); cyano; hydroxyl; nitro; azido; alkanoyl (such as a C2-C6 alkanoyl group); carboxamide; alkyl, cycloalkyl, alkenyl, alkynyl, alkoxy, aryloxy such as phenoxy; alkylthio including those having one or more thioether linkages; alkylsulfinyl; alkylsulfonyl groups including those having one or more sulfonyl linkages; aminoalkyl groups including groups having one or more N atoms; aryl (e.g., phenyl, biphenyl, naphthyl, or the like, each ring either substituted or unsubstituted aromatic); arylalkyl having for example, 1 to 3 separate or fused rings and from 6 to about 14 or 18 ring carbon atoms, with benzyl being an exemplary arylalkyl group; arylalkoxy, for example, having 1 to 3 separate or fused rings with benzyloxy being an exemplary arylalkoxy group; or a saturated, unsaturated, or aromatic heterocyclic group having 1 to 3 separate or fused rings with one or more N, O or S atoms, e.g. coumarinyl, quinolinyl, isoquinolinyl, quinazolinyl, pyridyl, pyrazinyl, pyrimidinyl, furanyl, pyrrolyl, thienyl, thiazolyl, triazinyl, oxazolyl, isoxazolyl, imidazolyl, indolyl, benzofuranyl, benzothiazolyl, tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, morpholinyl, piperazinyl, and pyrrolidinyl. Such heterocyclic groups may be further substituted, e.g. with hydroxy, alkyl, alkoxy, halogen and amino. In certain embodiments "optionally substituted" includes one or more substituents independently chosen from halogen, hydroxyl, amino, cyano, -CHO, -COOH, -CONH2, Ci-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, -Ci- C6alkoxy, C2-C6arkanoyl, Ci-Cealkylester, (mono- and di-Ci-C6alkylamino)Co-C2alkyl, Ci- C2haloalkyl, hydoxyCi-C6alkyl, ester, carbamate, urea, sulfonamide,-Ci-C6alkyl(heterocyclo), Ci-C6alkyl(heteroaryl), -Ci-C6alkyl(C3-C7cycloalkyl), 0-Ci-C6alkyl(C3-C7cycloalkyl), B(OH)2, phosphate, phosphonate and Ci-C2haloalkoxy.
[0082] "Alkyl" is a branched or straight chain saturated aliphatic hydrocarbon group. In one embodiment, the alkyl contains from 1 to about 12 carbon atoms, more generally from 1 to about 6 carbon atoms or from 1 to about 4 carbon atoms. In one embodiment, the alkyl contains from 1 to about 8 carbon atoms. In certain embodiments, the alkyl is Ci-C2, Ci-C3, or Ci-C6. The specified ranges as used herein indicate an alkyl group having each member of the range described as an independent species. For example, the term Ci-C6 alkyl as used herein indicates a straight or branched alkyl group having from 1, 2, 3, 4, 5, or 6 carbon atoms and is intended to mean that each of these is described as an independent species. For example, the term Ci- C4alkyl as used herein indicates a straight or branched alkyl group having from 1, 2, 3, or 4 carbon atoms and is intended to mean that each of these is described as an independent species. When C0-Cn alkyl is used herein in conjunction with another group, for example, (C3_ C7cycloalkyl)Co-C4 alkyl, or -Co-C4alkyl(C3-C7cycloalkyl), the indicated group, in this case cycloalkyl, is either directly bound by a single covalent bond (Coalkyl), or attached by an alkyl chain in this case 1, 2, 3, or 4 carbon atoms. Alkyls can also be attached via other groups such as heteroatoms as in -0-Co-C4alkyl(C3-C7cycloalkyl). Examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, isopentyl, tert-pentyl, neopentyl, n-hexyl, 2-methylpentane, 3-methylpentane, 2,2- dimethylbutane and 2,3-dimethylbutane. In one embodiment, the alkyl group is optionally substituted as described above.
[0083] "Alkenyl" is a branched or straight chain aliphatic hydrocarbon group having one or more carbon-carbon double bonds that may occur at a stable point along the chain. Nonlimiting examples are C2-Csalkenyl, C2-C6alkenyl and C2-C4alkenyl. The specified ranges as used herein indicate an alkenyl group having each member of the range described as an independent species, as described above for the alkyl moiety. Examples of alkenyl include, but are not limited to, ethenyl and propenyl. In one embodiment, the alkenyl group is optionally substituted as described above.
[0084] "Alkynyl" is a branched or straight chain aliphatic hydrocarbon group having one or more carbon-carbon triple bonds that may occur at any stable point along the chain, for example, C2-Cgalkynyl or C2-C6alkynyl. The specified ranges as used herein indicate an alkynyl group having each member of the range described as an independent species, as described above for the alkyl moiety. Examples of alkynyl include, but are not limited to, ethynyl, propynyl, 1- butynyl, 2-butynyl, 3-butynyl, 1-pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1-hexynyl, 2- hexynyl, 3-hexynyl, 4-hexynyl and 5-hexynyl. In one embodiment, the alkynyl group is optionally substituted as described above.
[0085] "Alkylene"is a bivalent saturated hydrocarbon. Alkylenes, for example, can be a 1 to 8 carbon moiety, 1 to 6 carbon moiety, or an indicated number of carbon atoms, for example Ci-C4alkylene, Ci-C3alkylene, or Ci-C2alkylene.
[0086] "Alkenylene" is a bivalent hydrocarbon having at least one carbon-carbon double bond. Alkenylenes, for example, can be a 2 to 8 carbon moiety, 2 to 6 carbon moiety, or an indicated number of carbon atoms, for example C2-C4alkenylene.
[0087] "Alkynylene" is a bivalent hydrocarbon having at least one carbon-carbon triple bond. Alkynylenes, for example, can be a 2 to 8 carbon moiety, 2 to 6 carbon moiety, or an indicated number of carbon atoms, for example C2-C4alkynylene.
[0088] "Alkoxy" is an alkyl group as defined above covalently bound through an oxygen bridge (-0-). Examples of alkoxy include, but are not limited to, methoxy, ethoxy, n-propoxy, i- propoxy, n-butoxy, 2-butoxy, t-butoxy, n-pentoxy, 2-pentoxy, 3-pentoxy, isopentoxy, neopentoxy, n-hexoxy, 2-hexoxy, 3-hexoxy, and 3-methylpentoxy. Similarly an "alkylthio" or a "thioalkyl" group is an alkyl group as defined above with the indicated number of carbon atoms covalently bound through a sulfur bridge (-S-). In one embodiment, the alkoxy group is optionally substituted as described above.
[0089] "Alkenyloxy" is an alkenyl group as defined covalently bound to the group it substitutes by an oxygen bridge (-0-).
[0090] "Alkanoyl" is an alkyl group as defined above covalently bound through a carbonyl (C=0) bridge. The carbonyl carbon is included in the number of carbons, that is C2alkanoyl is a CH3(C=0)- group. In one embodiment, the alkanoyl group is optionally substituted as described above.
[0091] "Alkylester" is an alkyl group as defined herein covalently bound through an ester linkage. The ester linkage may be in either orientation, e.g., a group of the formula -0(C=0)alkyl or a group of the formula -(C=0)Oalkyl.
[0092] "Amide" or "carboxamide" is -C(0)NRaRb wherein Ra and Rb are each independently selected from hydrogen, alkyl, for example, Ci-C6alkyl, alkenyl, for example, C2- C6alkenyl, alkynyl, for example, C2-C6alkynyl, -Co-C4alkyl(C3-Cycycloalkyl), -Co-C4alkyl(C3- Cyheterocycloalkyl), -Co-C4alkyl(aryl), and -Co-C4alkyl(heteroaryl); or together with the nitrogen to which they are bonded, Ra and Rb can form a Cs-Cyheterocyclic ring. In one embodiment, the Ra and Rb groups are each independently optionally substituted as described above.
[0093] "Carbocyclic group", "carbocyclic ring", or "cycloalkyl" is a saturated or partially unsaturated (i.e., not aromatic) group containing all carbon ring atoms. A carbocyclic group typically contains 1 ring of 3 to 7 carbon atoms or 2 fused rings each containing 3 to 7 carbon atoms. In one embodiment, a carbocyclic ring can be fused to an aryl ring. Cycloalkyl substituents may be pendant from a substituted nitrogen or carbon atom, or a substituted carbon atom that may have two substituents can have a cycloalkyl group, which is attached as a spiro group. Examples of carbocyclic rings include cyclohexenyl, cyclohexyl, cyclopentenyl, cyclopentyl, cyclobutenyl, cyclobutyl and cyclopropyl rings. In one embodiment, the carbocyclic ring is optionally substituted as described above. In one embodiment, the cycloalkyl is a partially unsaturated (i.e., not aromatic) group containing all carbon ring atoms. In another embodiment, the cycloalkyl is a saturated group containing all carbon ring atoms.
[0094] "Carbocyclic-oxy group" is a monocyclic carbocyclic ring or a mono- or bi- cyclic carbocyclic group as defined above attached to the group it substitutes via an oxygen, -0-, linker.
[0095] "Haloalkyl" indicates both branched and straight-chain alkyl groups substituted with 1 or more halogen atoms, up to the maximum allowable number of halogen atoms. Examples of haloalkyl include, but are not limited to, trifluoromethyl, monofluoromethyl, difluoromethyl, 2-fluoroethyl, and penta-fluoroethyl. [0096] "Haloalkoxy" indicates a haloalkyl group as defined herein attached through an oxygen bridge (oxygen of an alcohol radical).
[0097] "Hydroxyalkyl" is an alkyl group as previously described, substituted with at least one hydroxyl subsitutuent.
[0098] "Aminoalkyl" is an alkyl group as previously described, substituted with at least one amino subsitutuent.
[0099] "Halo" or "halogen" indicates independently any of fluoro, chloro, bromo, and iodo.
[0100] "Aryl" indicates aromatic groups containing only carbon in the aromatic ring or rings. In one embodiment, the aryl groups contain 1 to 3 separate or fused rings and is 6 to about 14 or 18 ring atoms, without heteroatoms as ring members. When indicated, such aryl groups may be further substituted with carbon or non-carbon atoms or groups. Such substitution may include fusion to a 5 to 7-membered saturated cyclic group that optionally contains 1 or 2 heteroatoms independently chosen from N, O, and S, to form, for example, a 3,4- methylenedioxyphenyl group. Aryl groups include, for example, phenyl and naphthyl, including 1-naphthyl and 2-naphthyl. In one embodiment, aryl groups are pendant. An example of a pendant ring is a phenyl group substituted with a phenyl group. In one embodiment, the aryl group is optionally substituted as described above.
[0101] The term "heterocycle," or "heterocyclic ring" as used herein refers to a saturated or a partially unsaturated (i.e., having one or more double and/or triple bonds within the ring without aromaticity) carbocyclic radical of 3 to about 12, and more typically 3, 5, 6, 7 to 10 ring atoms in which at least one ring atom is a heteroatom selected from nitrogen, oxygen, phosphorus and sulfur, the remaining ring atoms being C, where one or more ring atoms is optionally substituted independently with one or more substituents described above. A heterocycle may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms and 1 to 4 heteroatoms selected from N, O, P, and S) or a bicycle having 6 to 10 ring members (4 to 9 carbon atoms and 1 to 6 heteroatoms selected from N, O, P, and S), for example: a bicyclo [4,5], [5,5], [5,6], or [6,6] system. In one embodiment, the only heteroatom is nitrogen. In one embodiment, the only heteroatom is oxygen. In one embodiment, the only heteroatom is sulfur. Heterocycles are described in Paquette, Leo A.; "Principles of Modern Heterocyclic Chemistry" (W. A. Benjamin, New York, 1968), particularly Chapters 1, 3, 4, 6, 7, and 9; "The Chemistry of Heterocyclic Compounds, A series of Monographs" (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; and J. Am. Chem. Soc. (1960) 82:5566. Examples of heterocyclic rings include, but are not limited to, pyrrolidinyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, piperidonyl, morpholino, thiomorpholino, thioxanyl, piperazinyl, homopiperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 2- pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, dihydroisoquinolinyl, tetrahydroisoquinolinyl, pyrazolidinylimidazolinyl, imidazolidinyl, 2-oxa- 5-azabicyclo[2.2.2]octane, 3-oxa-8-azabicyclo[3.2.1]octane, 8-oxa-3-azabicyclo[3.2.1]octane, 6- oxa-3-azabicyclo[3.1.1]heptane, 2-oxa-5-azabicyclo[2.2.1]heptane, 3-azabicyco[3.1.0]hexanyl, 3-azabicyclo[4.1.0]heptanyl, azabicyclo[2.2.2]hexanyl, 3H-indolyl, quinolizinyl, N-pyridyl ureas, and pyrrolopyrimidine. Spiro moieties are also included within the scope of this definition. Examples of a heterocyclic group wherein 1 or 2 ring carbon atoms are substituted with oxo (=0) moieties are pyrimidinonyl and 1,1-dioxo-thiomorpholinyl. The heterocycle groups herein are optionally substituted independently with one or more substituents described herein.
[0102] "Heterocyclicoxy group" is a monocyclic heterocyclic ring or a bicyclic heterocyclic group as described previously linked to the group it substitutes via an oxygen, -0-, linker.
[0103] "Heteroaryl" indicates a stable monocyclic aromatic ring which contains from 1 to 3, or in some embodiments from 1 to 2, heteroatoms chosen from N, O, and S, with remaining ring atoms being carbon, or a stable bicyclic or tricyclic system containing at least one 5- to 7- membered aromatic ring which contains from 1 to 3, or in some embodiments from 1 to 2, heteroatoms chosen from N, O, and S, with remaining ring atoms being carbon. In one embodiment, the only heteroatom is nitrogen. In one embodiment, the only heteroatom is oxygen. In one embodiment, the only heteroatom is sulfur. Monocyclic heteroaryl groups typically have from 5 to 7 ring atoms. In some embodiments bicyclic heteroaryl groups are 9- to 10-membered heteroaryl groups, that is, groups containing 9 or 10 ring atoms in which one 5- to 7-member aromatic ring is fused to a second aromatic or non-aromatic ring. When the total number of S and O atoms in the heteroaryl group exceeds 1, these heteroatoms are not adjacent to one another. In one embodiment, the total number of S and O atoms in the heteroaryl group is not more than 2. In another embodiment, the total number of S and O atoms in the aromatic heterocycle is not more than 1. Examples of heteroaryl groups include, but are not limited to, pyridinyl (including, for example, 2-hydroxypyridinyl), imidazolyl, imidazopyridinyl, pyrimidinyl (including, for example, 4-hydroxypyrimidinyl), pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxadiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, tetrahydroisoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, triazolyl, thiadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, tetrahydrofuranyl, and furopyridinyl. Heteroaryl groups are optionally substituted independently with one or more substituents described herein. "Heteroaryloxy" is a heteroaryl group as described bound to the group it substituted via an oxygen, -0-, linker.
[0104] "Heterocycloalkyl" is a saturated ring group. It may have, for example, 1, 2, 3, or 4 heteroatoms independently chosen from N, S, and O, with remaining ring atoms being carbon. In a typical embodiment, nitrogen is the heteroatm. Monocyclic heterocycloalkyl groups typically have from 3 to about 8 ring atoms or from 4 to 6 ring atoms. Examples of heterocycloalkyl groups include morpholinyl, piperazinyl, piperidinyl, and pyrrolinyl.
[0105] The term "mono- and/ or di-alkylamino" indicates secondary or tertiary alkylamino groups, wherein the alkyl groups are independently chosen alkyl groups, as defined herein. The point of attachment of the alkylamino group is on the nitrogen. Examples of mono- and di-alkylamino groups include ethylamino, dimethylamino, and methyl-propyl-amino.
[0106] A "dosage form" means a unit of administration of an active agent. Examples of dosage forms include tablets, capsules, injections, suspensions, liquids, emulsions, implants, particles, spheres, creams, ointments, suppositories, inhalable forms, transdermal forms, buccal, sublingual, topical, gel, mucosal, and the like. A "dosage form" can also include an implant, for example an optical implant.
[0107] "Pharmaceutical compositions" are compositions comprising at least one active agent, such as a compound or salt of Formula I, and at least one other substance, such as a carrier. "Pharmaceutical combinations" are combinations of at least two active agents which may be combined in a single dosage form or provided together in separate dosage forms with instructions that the active agents are to be used together to treat any disorder described herein.
[0108] "Pharmaceutically acceptable salts" includes derivatives of the disclosed compounds in which the parent compound is modified by making inorganic and organic, nontoxic, acid or base addition salts thereof. The salts of the present compounds can be synthesized from a parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate, or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two. Generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are typical, where practicable. Salts of the present compounds further include solvates of the compounds and of the compound salts.
[0109] Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts and the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, conventional nontoxic acid salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, mesylic, esylic, besylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, HOOC-(CH2)n-COOH where n is 0-4, and the like. Lists of additional suitable salts may be found, e.g., in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., p. 1418 (1985).
[0110] The term "carrier" applied to pharmaceutical compositions/combinations of the invention refers to a diluent, excipient, or vehicle with which an active compound is provided.
[0111] A "pharmaceutically acceptable excipient" means an excipient that is useful in preparing a pharmaceutical composition/combination that is generally safe, non-toxic and neither biologically nor otherwise inappropriate for administration to a host, and includes, in one embodiment, an excipient that is acceptable for veterinary use as well as human pharmaceutical use. A "pharmaceutically acceptable excipient" as used in the present application includes both one and more than one such excipient.
[0112] A "patient" or "host" or "subject" is a human or non-human animal in need of modulation of the complement factor D pathway. Typically the host is a human. A "patient" or "host" or "subject" also refers to for example, mammals, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like.
[0113] A "prodrug" as used herein, means a compound which when administered to a host in vivo is converted into a parent drug. As used herein, the term "parent drug" means any of the presently described chemical compounds that are useful to treat any of the disorders described herein, or to control or improve the underlying cause or symptoms associated with any physiological or pathological disorder described herein in a host, typically a human. Prodrugs can be used to achieve any desired effect, including to enhance properties of the parent drug or to improve the pharmaceutic or pharmacokinetic properties of the parent. Prodrug strategies exist which provide choices in modulating the conditions for in vivo generation of the parent drug, all of which are deemed included herein. Nonlimiting examples of prodrug strategies include covalent attachment of removable groups, or removable portions of groups, for example, but not limited to acylation, phosphorylation, phosphonylation, phosphoramidate derivatives, amidation, reduction, oxidation, esterification, alkylation, other carboxy derivatives, sulfoxy or sulfone derivatives, carbonylation or anhydride, among others.
[0114] "Providing a compound of Formula I with at least one additional active agent" means the compound of Formula I and the additional active agent(s) are provided simultaneously in a single dosage form, provided concomitantly in separate dosage forms, or provided in separate dosage forms for administration separated by some amount of time that is within the time in which both the compound of Formula I and the at least one additional active agent are within the blood stream of a patient. In certain embodiments the compound of Formula I and the additional active agent need not be prescribed for a patient by the same medical care worker. In certain embodiments the additional active agent or agents need not require a prescription. Administration of the compound of Formula I or the at least one additional active agent can occur via any appropriate route, for example, oral tablets, oral capsules, oral liquids, inhalation, injection, suppositories or topical contact. [0115] A "therapeutically effective amount" of a pharmaceutical composition/combination of this invention means an amount effective, when administered to a patient, to provide a therapeutic benefit such as an amelioration of symptoms, e.g., an amount effective to decrease the symptoms of a macular degeneration. In one embodiment, a therapeutically effective amount is an amount sufficient to prevent a significant increase or will significantly reduce the detectable level of complement factor D in the patient's blood, serum, or tissues.
II. DETAILED DESCRIPTION OF THE ACTIVE COMPOUNDS
[0116] According to the present invention a compound of Formula I is provided:
Figure imgf000029_0001
as well as the pharmaceutically acceptable salts and compositions thereof. Formula I can be considered to have a central core, an L-B substituent, and a (C=0)A substituent. It has been discovered that a compound of Formula I, or a pharmaceutically acceptable salt or composition thereof, is a superior inhibitor of complement factor D, and therefore can be used as an effective amount to treat a host in need of complement factor D modulation.
[0117] Non- limiting examples of compounds falling within Formula I with variations in the variables e.g., A, B, R^-R3 , and L, are illustrated below. The disclosure includes all combinations of these definitions so long as a stable compound results.
Formulas II - XXX
[0118] In one aspect, the disclosure includes compounds and salts of Formula II, III, IV, V, VI, VII, VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXIII, XXIV, XXV, XXVI, XXVII, XXVIII, XXIX and XXX which are within the scope of Formula I. The variables shown in Formula II-XXX carry the definitions set forth in the SUMMARY section for Formula I or any of the definitions set forth in this disclosure.
Figure imgf000030_0001
Formula V Formula VI Formula VII
Figure imgf000030_0002
Formula XI Formula XII Formula XIII
Figure imgf000031_0001
Formula XV Formula XVI
Figure imgf000031_0002
m is 0 or 1. Formula XVII Formula XVIII Formula XIX
Figure imgf000031_0003
Formula XXIII Formula XXIV Formula XXV
Figure imgf000032_0001
m is 0 or 1. m is 0 or 1. m is 0 or 1.
Formula XXVI Formula XXVII Formula XXVIII
Figure imgf000032_0002
m is 0 or 1. m is 0 or 1.
Formula XXIX Formula XXX
[0119] Additionally, the disclosure includes compounds and salts of Formula I and pharmaceutically acceptable compositions thereof, and any of its subformulae (II-XXX) in which at least one of the following conditions is met in the embodiments described below.
The R and R Substituents
[0120] It has been discovered that a compound of Formula I, a pharmaceutically acceptable salt or composition thereof, is a superior inhibitor of complement factor D.
[0121] In one embodiment, R12 and R13 are independently chosen at each occurrence from (q), (r) and (s):
(q) hydrogen, halogen, hydroxyl, nitro, cyano, amino, -COOH, Ci-C2haloalkyl,
Ci-C2haloalkoxy;
(r) Ci-C6alkyl, -Co-C4alkyl(C3-Cycycloalkyl), C2-C6alkenyl, C2-C6alkanoyl, Ci-C6alkoxy, C2-C6alkenyloxy, -C(0)OR9, Ci-C6thioalkyl, -C0-C4alkylNR9R10, -C(0)NR9R10, -S02R9, -S02NR9R10, -OC(0)R9, and -C(NR9)NR9R10 each of which (r) is unsubstituted or substituted with one or more substituents independently selected from halogen, hydroxyl, nitro, cyano, amino, -COOH, -CONH2? C^Cjhaloalkyl, and C Cjhaloalkoxy, and each of which (r) is also optionally substituted with one substituent chosen from phenyl and 4- to 7- membered heterocycle containing 1, 2, or 3 heteroatoms independently chosen from N, O, and S; which phenyl or 4- to 7-membered heterocycle is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, CrC6alkyl, C2- C6alkenyl, C2-C6alkanoyl, CrC6alkoxy, (mono- and di-C1-C6alkylamino)C0-C4alkyl, Cr C6alkylester, -(C0-C4alkyl)(C3-C7cycloalkyl), CrC2haloalkyl, and CrC2haloalkoxy;
(s) -C(CH2)2_4R30.
[0122] R30 is -NR9C(0)R31 or R32.
[0123] R31 and R32 are Ci-C6alkyl, Ci-C6haloalkyl, (C3-C7cycloalkyl)Co-C4alkyl, (phenyl)Co-C4alkyl, (4- to 7-membered heterocycloalkyl)Co-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S, and (5- or 6- membered unsaturated or aromatic heterocycle)Co-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S.
[0124] Each of which (s) may be unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, amino, oxo, -B(OH)2, -Si(CH3)3,
-COOH, -CONH2, -P(0)(OH)2, Ci-C6alkyl, Ci-C6alkoxy, -C0-C2alkyl(mono- and di-C C4alkylamino), Ci-Cealkylester, Ci-C4alkylamino, Ci-C4hydroxylalkyl, Ci-C2haloalkyl, and Ci- C2haloalkoxy.
[0125] In certain embodiments, R12 or R13 are independently selected from:
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001

Figure imgf000036_0001
Non-limiting R'TR Embodiments
[0126] In one embodiment, the disclosure provides compounds of Formula I, wherein;
12 13 12 13
one of R and R is H and the other of R and R is chosen from (s):
[0127] (s) -C(CH2)2R30;
[0128] wherein R30 is as defined in the summary section above.
[0129] In another embodiment, the disclosure provides compounds of Formula I, wherein;
[0130] R1, R1', R2, and R3'are all hydrogen; [0131] R2 is fluoro and R3 is hydrogen, -Co-C4alkyl(C3-C7cycloalkyl), or -O-C0- C4alkyl(C3-C7cycloalkyl);
[0132] R5 is hydrogen, halogen, or Ci-C2alkyl;
[0133] R11, R13, R14, and R15, if present, are independently chosen at each occurrence from hydrogen, halogen, hydroxyl, amino, Ci-C4alkyl, Ci-C4alkoxy, -Co-C2alkyl(mono- and di- Ci-C2alkylamino), trifluoromethyl, and trifluoromethoxy;
[0134] X12 is CR12; and
[0135] R12 is chosen from (s):
[0136] (s) -C(CH2)2R30;
[0137] wherein R30 is as defined in the summary section above.
[0138] In one embodiment, the disclosure provides compounds of Formula I, wherein;
[0139] m is O or 1 ;
[0140] R2 is halogen, R2 is hydrogen or halogen, and R3 is hydrogen, halogen, -Co- C4alkyl(C3-C7cycloalkyl), or -O-C0-C4alkyl(C3-C7cycloalkyl);
[0141] R6 is -C(0)Ci-C4alkyl, -C(0)NH2, -C(0)CF3, -C(0)(C3-C7cycloalkyl), or -ethyl(cyanoimino);
[0142] one of R12 and R13 is selected from hydrogen, halogen, Ci-C4alkyl, Ci-C4alkoxy, trifluoromethyl, and trifluoromethoxy; the other of R12 and R13 is chosen from (s):
[0143] (s) -C(CH2)2R30;
[0144] wherein R30 is as defined in the summary section above.
[0145] In one embodiment, the disclosure provides compounds of Formula I, wherein;
[0146] one of R12 and R13 is hydrogen, hydroxyl, halogen, methyl, or methoxy; and the other of R12 and R13 is chosen from (s):
[0147] (s) -C(CH2)2R30;
[0148] wherein R30 is as defined in the summary section above. Central Core Moiety
[0149] The central core moiety in Formula I is illustrated below:
Figure imgf000038_0001
wherein:
[0150] Q1 is N(RJ) or QRV).
[0151] Q2 is C(R2R2'), C(R2R2')-C(R2R2'), S, O, N(R2) or C(R2R2')0.
[0152] Q3 is N(R3), S, or C(R3R3').
[0153] (a) X1 and X2 are independently N, CH, or CZ, or (b) X1 and X2 together
[0154] Q1, Q2, Q3, X1, and X2 are selected such that a stable compound results.
[0155] Non- limiting examples of the
Figure imgf000038_0002
ring are illustrated below (any of which
Figure imgf000038_0003
Figure imgf000039_0001
[0156] R and R' are independently chosen from H, alkyl, cycloalkyl, alkylcycloalkyl, cycloalkylalkyl, heterocycle, heterocycloalkyl, aryl, aralkyl, heteroaryl, heteroarylalkyl wherein each group can be optionally substituted or any other substituent group herein that provides the desired properties. In some embodiments, the ring includes one or more chiral carbon atoms. The invention includes embodiments in which the chiral carbon can be provided as an enantiomer, or mixtrues of enantiomers, including a racemic mixture. Where the ring includes more than one stereocenter, all of the enantiomers and diastereomers are included in the invention as individual species.
[0157] Z is F, CI, NH2, CH3, CH2D, CHD2, or CD3.
1 1 ' 2 2 ' 3 3'
[0158] R , R , R , R , R , and R are independently chosen at each occurence from (c) and (d):
(c) hydrogen, halogen, hydroxyl, nitro, cyano, amino, Ci-C6alkyl, C2-C6alkenyl, Ci- C6alkoxy, C2-C6alkynyl, C2-C6alkanoyl, Ci-C6thioalkyl, hydroxyCi-C6alkyl, aminoCi-C6alkyl, -Co-C4alkylNR9R10, -C(0)OR9, -OC(0)R9, -NR9C(0)R10, -C(0)NR9R10, -OC(0)NR9R10, -O(heteroaryl), -NR9C(0)OR10, Ci-C2haloalkyl, and Ci-C2haloalkoxy, where R9 and R10 are independently chosen at each occurrence from hydrogen, Ci-C6alkyl, and (C3-C7cycloalkyl)Co- C4alkyl;
(d) -Co-C4alkyl(C3-C7cycloalkyl) and -O-C0-C4alkyl(C3-C7cycloalkyl).
Non-limiting Central Core Embodiments
[0159] In alternative embodiments, any one of the following rings (e), (f), (g), (h), (i), or (j) may be present:
(e) R1 and R1 or R3 and R3 may be taken together to form a 3- to 6-membered carbocyclic spiro ring or a 3- to 6-membered heterocyclic spiro ring containing 1 or 2 heteroatoms independently chosen from N, O, or S;
(f) R2 and R2 may be taken together to form a 3- to 6-membered carbocyclic spiro ring,
(g) R2 and R2 may be taken together to form a 3- to 6-membered heterocyclic spiro ring, each of which spiro rings (e), (f), and (g) is unsubstituted or substituted with 1 or more substituents independently chosen from halogen (and in particular F), hydroxyl, cyano, -COOH, Ci-C4alkyl (including in particular methyl), C2-C4alkenyl, C2-C4alkynyl, Ci-C4alkoxy, C2- C4alkanoyl, hydroxyCi-C4alkyl, (mono- and di-Ci-C4alkylamino)Co-C4alkyl, -Co-C4alkyl(C3- C7cycloalkyl), -0-Co-C4alkyl(C3-C7cycloalkyl), Ci-C2haloalkyl, and Ci-C2haloalkoxy.
(h) R1 and R2 may be taken together to form a 3-membered carbocyclic ring;
(i) R1 and R2 may be taken together to form a 4- to 6-membered carbocyclic ring or a 4- to 6-membered heterocyclic ring containing 1 or 2 heteroatoms independently chosen from N, O, and S.
(j) R2 and R3, if bound to adjacent carbon atoms, may be taken together to form a 3- to 6- membered carbocyclic ring or a 3- to 6-membered heterocyclic ring; each of which ring (h), (i), and (j) may be unsubstituted or substituted with 1 or more substituents independently chosen from halogen (and in particular F), hydroxyl, cyano, -COOH, Ci-C4alkyl (including in particular methyl), C2-C4alkenyl, C2-C4alkynyl, Ci-C4alkoxy, C2-C4alkanoyl, hydroxyCi-C4alkyl, (mono- and di-Ci-C4alkylamino)Co-C4alkyl, -C0-C4alkyl(C3-C7cycloalkyl), -O-C0-C4alkyl(C3- C7cycloalkyl), Ci-C2haloalkyl, and Ci-C2haloalkoxy.
[0160] In one embodiment, the central core moiety is proline.
[0161] In one embodiment, the central core moiety is 4-fluoroproline. [0162] In one embodiment, R 1 , R 1 ' , R 2 ' , R 3 , and R 3 ' , if present, are all hydrogen; and R 2 is fluoro.
[0163] In one embodiment, R1, R1 , R2 , and R3 , if present, are all hydrogen; and R2 is fluoro and R3 is -Co-C4alkyl(C3-Cycycloalkyl) or -0-Co-C4alkyl(C3-Cvcycloalkyl).
[0164] In one embodiment, R1 and R2 are taken together to form a 3- to 6-membered cycloalkyl group, and R1 , R2 , R3, and R3 , where present, are all hydrogen.
[0165] In one embodiment, R1, R1 , R3, and R3 , if present, are all hydrogen, and R2 and R2' are taken together to form a 5- or 6-membered heterocycloalkyl group having 1 or 2 oxygen atoms.
[0166] In one embodiment, R1 is hydrogen and R2 is fluoro.
[0167] In one embodiment, R1 and R2 are joined to form a 3 membered ring.
[0168] The disclosure includes compounds of Formula I in which the central pyrrolidine is vinyl substituted, for example:
Figure imgf000041_0001
[0169] In one embodiment, the compound of Formula I has the structure:
Figure imgf000041_0002
[0170] In one embodiment, the central pyrrolidine is modified by addition of a second heteroatom to a pyrrolidine ring, such as N, O, S, or Si, for example:
Figure imgf000041_0003
[0171] Another modification within the scope of the disclosure is joining a substituent on the central pyrrolidine ring to R7 or R8 to form a 5- to 6- membered heterocyclic ring, for example:
Figure imgf000042_0001
[0172 Example compounds having the modifications disclosed above include:
Figure imgf000042_0002
Central Core L-B Substituents
[0173] The central core L-B substituents in Formula I are illustrated below:
Figure imgf000042_0003
[0174] L is either (t), (u), or (v):
(t) is a group of the formula
O RI B R18' R18 R18' o O R18 R18-
R17 , R17 0 , or OH where R17 is hydrogen or Ci-C6alkyl and R18 and R18 are independently chosen from hydrogen, halogen, hydroxymethyl, and methyl; and m is 0, 1, 2, or 3;
(u) is a bond, (v) or a directly linked optionally substitiuted alkyl, alkyl(heteroaryl), heterocyclic aryl, heteroaryl, moiety including but not limited to:
Figure imgf000043_0001
[0175] B is a monocyclic, bicyclic carbocyclic or carbocyclic-oxy group or a monocyclic, bicyclic, or tricyclic heterocyclic group having 1 , 2, 3, or 4 heteroatoms independently selected from N, O, and S and from 4 to 7 ring atoms per ring, or B is a C2- C6alkenyl or C2-C6alkynyl group, or B is -(C0-C4alkyl)(aryl), -(C0-C4alkyl)(heteroaryl), or -(C0- C4alkyl)(biphenyl) .
[0176] Each of which B is unsubstituted or substituted with one or more substituents independently chosen from (w) and (x) and 0 or 1 substituents chosen from (y) and (z):
(w) halogen, hydroxyl, -COOH, cyano, Ci-C6alkyl, C2-C6alkanoyl, Ci-C6alkoxy, -C0- C4alkylNR9R10, -S02R9, Ci-C2haloalkyl, and Ci-C2haloalkoxy;
(x) nitro, C2-C6alkenyl, C2-C6alkynyl, Ci-Cethioalkyl, -JC3-Cvcycloalkyl, -B(OH)2, -JC(0)NR9R23,-JOS02OR21, -C(0)(CH2)i_4S(0)R21, SR9 -0(CH2)i_4S(0)NR21R22
-JOP(0)(OR21)(OR22), -JP(0)(OR21)(OR22), -JOP(0)(OR21)R22, -JP(0)(OR21)R22, -JOP(0)R21R22, -JP(0)R21R22, -JSP(0)(OR21)(OR22), -JSP(0)(OR21)(R22), -JSP(0)(R21)(R22), -JNR9P(0)(NHR21)(NHR22), -JNR9P(0)(OR21)(NHR22), -JNR9P(0)(OR21)(OR22), -JC(S)R21, -JNR21S02R22, -JNR9S(O)NR10R22, -JNR9SO2NR10R22, -JS02NR9COR22, -JS02NR9CONR21R22, -JNR21S02R22, -JC(0)NR21S02R22, -JC(NH2)NR22, -JC(NH2)NR9S(0)2R22, -JOC(0)NR21R22, -JNR21C(0)OR22, -JNR21OC(0)R22, -(CH2)i_4C(0)NR21R22, -JC(0)R24R25, -JNR9C(0)R21, -JC(0)R21, -JNR9C(O)NR10R22, -CCR21, -(CH2)i_4OC(0)R21, and -JC(0)OR23; each of which (x) may be unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, amino, oxo, -B(OH)2, -Si(CH3)3, -COOH, -CONH2, -P(0)(OH)2, Ci-C6alkyl, Ci-C6alkoxy, -Co-C2alkyl(mono- and di-Ci-C4alkylamino), Ci- C6alkylester, Ci-C4alkylamino, Ci-C4hydroxylalkyl, Ci-C2haloalkyl, and Ci-C2haloalkoxy;
(y) naphthyl, naphthyloxy, indanyl, (4- to 7-membered heterocycloalkyl)Co-C4alkyl containing 1 or 2 heteroatoms chosen from N, O, and S, and bicyclic heterocycle containing 1 , 2, or 3 heteroatoms independently chosen from N, O, and S, and containing 4- to 7- ring atoms in each ring; each of which (y) is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, Ci-C6alkyl, C2-C6alkenyl, C2- C6alkanoyl, Ci-C6alkoxy, (mono- and di-Ci-C6alkylamino)Co-C4alkyl, Ci-Cealkylester, -C0- C4alkyl(C3-Cycycloalkyl), -S02R9, Ci-C2haloalkyl, and Ci-C2haloalkoxy; and
(z) tetrazolyl, (phenyl)C0-C2alkyl, (phenyl)Ci-C2alkoxy, phenoxy, and 5- or 6-membered heteroaryl containing 1, 2, or 3 heteroatoms independently chosen from N, O, B, and S, each of which (z) is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, Ci-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, Ci-C6alkoxy, (mono- and di-Ci-C6alkylamino)Co-C4alkyl, Ci-C6alkylester, -Co-C4alkyl(C3-C7cycloalkyl), -S02R9, -OSi(CH3)2C(CH3)3, -Si(CH3)2C(CH3)3, Ci-C2haloalkyl, and Ci-C2haloalkoxy.
[0177] In one embodiment, -L-B- is
Figure imgf000044_0001
, where
[0178] R and R are independently chosen from hydrogen, halogen, hydroxyl, nitro, cyano, Ci-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, Ci-C6alkoxy, Ci-C6thioalkyl, -C0- C4alkyl(mono- and di-Ci-Cealkylamino), -Co-C4alkyl(C3-C7cycloalkyl), -Co-C4alkoxy(C3- C7cycloalkyl), Ci-C2haloalkyl, Ci-C2haloalkoxy, and Ci-C2haloalkylthio.
Non-Limiting L-B Embodiments
0179] In another embodiment, -L-B- is
Figure imgf000044_0002
Figure imgf000045_0001
[0180] R18 and R18 are independently chosen from hydrogen, halogen, hydroxymethyl, and methyl; and m is 0 or 1 ; and
[0181] R 26 , R 27 , and R 28 are independently chosen from hydrogen, halogen, hydroxyl, nitro, cyano, Ci-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, Ci-C6alkoxy, Ci-C6thioalkyl, (mono- and di-Ci-C6alkylamino)Co-C4alkyl, (C3-Cycycloalkyl)Co-C4alkyl, (aryl)Co-C4alkyl-, (heteroaryl)Co-
C4alkyl-, and -Co-C4alkoxy 26 , R 27 , and R 28
(C3-Cvcycloalkyl); each of which R other than hydrogen, halogen, hydroxyl, nitro, cyano, is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, amino, Ci-C2alkoxy, Ci-C2haloalkyl, (C3-C7cycloalkyl)Co-C4alkyl-, and Ci-C2haloalkoxy; and
[0182] R29 is hydrogen, Ci-C2alkyl, CiC2haloalkyl or -Si(CH3)2C(CH3)3.
[0183] In one embodiment, m is 0.
[0184] In one embodiment, the disclosure further includes compounds and salts of Formula I in which B is 2-fluoro-3-chlorophenyl. In another embodiment, another carbocyclic, aryl, heterocyclic, or heteroaryl group such as 2-bromo-pyridin-6-yl, l-(2,2,2-trifluoroethyl)-lH- pyrazol-3-yl, 2,2-dichlorocyclopropylmethyl, or 2-fluoro-3-trimethylsilylphenyl is used.
[0185] In another embodiment, B is phenyl, pyridyl, or indanyl each of which is unsubstituted or substituted with one or more substituents independently chosen from hydrogen, halogen, hydroxyl, nitro, cyano, Ci-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, Ci-C6alkoxy, Ci- C6thioalkyl, (mono- and di-Ci-C6alkylamino)Co-C4alkyl, (C3-Cvcycloalkyl)Co-C4alkyl, -C0- C4alkoxy(C3-C7cycloalkyl), (phenyl)Co-C2alkyl, (pyridyl)Co-C2alkyl; each of which substituents other than hydrogen, halogen, hydroxyl, nitro, cyano, is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, amino, Ci-C2alkyl, Ci- C2alkoxy, -OSi(CH3)2C(CH3)3, -Si(CH3)2C(CH3)3, Ci-C2haloalkyl, and Ci-C2haloalkoxy. [0186] In another embodiment, B is phenyl or pyridyl substituted with 1, 2, or 3 substituents chosen from chloro, bromo, hydroxyl, -SCF3, Ci-C2alkyl, Ci-C2alkoxy, trifluoromethyl, phenyl and trifluoromethoxy each of which substituents other than chloro, bromo, hydroxyl, -SCF3, can be optionally substitued.
[0187] In certain embodiments, B is a 2-fluoro-3-chlorophenyl or a 2-fluoro-3- trifluoromethoxyphenyl group.
[0188] In one embodiment, B is pyridyl, optionally substituted with halogen, Ci- C2alkoxy, and trifluoromethyl.
[0189] In one embodiment, B is phenyl, substituted with 1, 2, or 3 substituents independently selected from halogen, Ci-C2alkyl, Ci-C2alkoxy, trifluoromethyl, and optionally substituted phenyl.
[0190] In one embodiment, R23 is independently chosen at each occurrence from (C3- Cycycloalkyl)Co-C4alkyl, (phenyl)Co-C4alkyl, (4- to 7-membered heterocycloalkyl)Co-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S, and (5- or 6- membered unsaturated or aromatic heterocycle)Co-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S.
[0191] In one embodiment, B is selected from
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001

Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
w ere R s y rogen, met y, or tr uoromet y; R s y rogen or aogen; an R s hydrogen, methyl, trifluoromethyl, or -Si(CH3)2C(CH3)3.
Central Core (C=0)A Substituent
[0192] The central core (C=0)A substituent in Formula I is illustrated below:
Figure imgf000052_0002
[0193] A is a group chosen from (k) and (1) where (k) is
Figure imgf000052_0003
and (1) is
Figure imgf000053_0001
[0194] X4 is B(OH) and Y is CHR9; or X 4 is CHR9 and Y is B(OH).
[0195] R101 is hydrogen, alkyl, carboxy.
[0196] R4 is (m) or (n):
(m) -CHO, -CONH2, or C2-C6alkanoyl, including C(0)C3-C7cycloalkyl;
(n) hydrogen, -S02NH2, -C(CH2)2F, -CH(CF3)NH2, Ci-C6alkyl, -C0-C4alkyl(C3- C7cycloalkyl), -C(O)C0-C2alkyl(C3-C7cycloalkyl),
Figure imgf000054_0001
each of which R4 other than hydrogen, -CHO, and -CONH2, is unsubstituted or substituted with one or more of amino, imino, halogen, hydroxyl, cyano, cyanoimino, Ci-C2alkyl, Ci-C2alkoxy, -Co-C2alkyl(mono- and di-Ci-C4alkylamino), Ci-C2haloalkyl, and Ci-C2haloalkoxy.
[0197] R5 and R6 are independently chosen from (o) and (p):
(o) -CHO, -C(0)NH2, -C(0)NH(CH3), or C2-C6alkanoyl;
(p) hydrogen, hydroxyl, halogen, cyano, nitro, -COOH, -S02NH2, -C(NH2)Ci-C3alkyl, -C(NH2)C C3haloalkyl, -CF(C=CH2), -C(=NCN)Ci-C6alkyl, Ci-C6alkyl, C2-C6alkenyl, C C6alkoxy, -Co-C4alkyl(C3-C7cycloalkyl), -C(O)C0-C4alkyl(C3-C7cycloalkyl), -P(0)(OR9)2, -OC(0)R9, -C(0)OR9, -C(0)N(CH2CH2R9)(R10), -NR9C(0)R10, phenyl, or 5- to 6-membered heteroaryl.
[0198] Each R5 and R6 other than hydrogen, hydroxyl, cyano, and -COOH is unsubstituted or optionally substituted. For example, R5 and R6 other than hydrogen, hydroxyl, cyano, and -COOH may be substituted with one or more substituents independently chosen from halogen, hydroxyl, amino, imino, cyano, cyanoimino, Ci-C2alkyl, Ci-C4alkoxy, -Co- C2alkyl(mono- and di-Ci-C4alkylamino), Ci-C2haloalkyl, Ci-C2haloalkoxy, C(0)alkyl, C(0)cycloalkyl, C(0)aryl, C(0)heterocycle, and C(0)heteroaryl.
[0199] R6 is hydrogen, halogen, hydroxyl, Ci-C4alkyl, or Ci-C4alkoxy; or R6 and R6 may be taken together to form an oxo, vinyl, or imino group.
[0200] R7 is hydrogen, Ci-C6alkyl, or -C0-C4alkyl(C3-C7cycloalkyl).
[0201] R8 and R8 are independently chosen from hydrogen, halogen, hydroxyl, Ci- C6alkyl, Ci-C6alkoxy, and (Ci-C4alkylamino)Co-C2alkyl, or R8 and R8 are taken together to form an oxo group, or can be taken together with the carbon that they are bonded to form a 3- membered carbocyclic ring.
[0202] R16 is 0 or 1 or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, Ci-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, Ci-C6alkoxy, -Co-C4alkyl(mono- and di- Ci-Cealkylamino), -Co-C4alkyl(C3-C7cycloalkyl), Ci-C2haloalkyl, and Ci-C2haloalkoxy. [0203] R19 is hydrogen, Ci-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, -S02Ci-C6alkyl, (mono- and di-Ci-C6alkylamino)Ci-C4alkyl, -Co-C4alkyl(C3-C7cycloalkyl), -C0-C4alkyl(C3- C7heterocycloalkyl), -Co-C4alkyl(aryl), Co-C4alkyl(heteroaryl), each of which R19 other than hydrogen is substituted with 0 or 1 or more substituents independently chosen from halogen, hydroxyl, amino, -COOH, and -C(0)OCi-C4alkyl.
[0204] X11 is N or CR11.
[0205] X12 is N or CR12.
[0206] X13 is N or CR13.
[0207] X14 is N or CR14.
[0208] No more than 2 of X11, X12, X13, and X14 are N.
[0209] R11, R14, and R15 are independently chosen at each occurrence from hydrogen, halogen, hydroxyl, nitro, cyano, -NR9C(0)R10, C(0)NR9R10, -0(PO)(OR9)2, -(PO)(OR9)2, d- C6alkyl, C2-C6alkenyl, C2-C6alkenyl(aryl), C2-C6alkenyl(cycloalkyl), C2-C6alkenyl(heterocycle), C2-C6alkenyl(heteroaryl), C2-C6alkynyl, C2-C6alkynyl(aryl), C2-C6alkynyl(cycloalkyl), C2- C6alkynyl(heterocycle), C2-Cealkynyl(heteroaryl), C2-C6alkanoyl, Ci-C6alkoxy, Ci-C6thioalkyl, - Co-C4alkyl(mono- and di-Ci-Cealkylamino), -Co-C4alkyl(C3-C7cycloalkyl), (phenyl)Co-C4alkyl, (4- to 7-membered heterocycloalkyl)Co-C4alkyl having 1 , 2, or 3 heteroatoms independently chosen from N, O, and S, and (5- or 6- membered unsaturated or aromatic heterocycle)C0- C4alkyl having 1 , 2, or 3 heteroatoms independently chosen from N, O, and S), -C0-C4alkoxy(C3- C7cycloalkyl), Ci-C2haloalkyl, and Ci-C2haloalkoxy. Alternatively, R13 and R14 can together form a bridge that can be an optionally substituted cycloalkyl, heterocycle or heteroaryl ring.
[0210] In one embodiment, R5 and R6 are independently chosen from -CHO, -C(0)NH2, -C(0)NH(CH3), C2-C6alkanoyl, and hydrogen.
[021 1] In one embodiment, each R5 and R6 other than hydrogen, hydroxyl, cyano, and -COOH is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, amino, imino, cyano, cyanoimino, Ci-C2alkyl, Ci-C4alkoxy, -Co- C2alkyl(mono- and di-Ci-C4alkylamino), Ci-C2haloalkyl, Ci-C2haloalkoxyi C(0)alkyl, C(0)cycloalkyl, C(0)aryl, C(0)heterocycle, and C(0)heteroaryl.
[0212] In one embodiment, R8 and R8 are independently hydrogen or methyl.
[0213] In one embodiment, R8 and R8 are hydrogen.
[0214] In one embodiment, R7 is hydrogen or methyl. [0215] In one embodiment, R is hydrogen.
[0216] In an alternate embodiment, two A groups can be bonded together to form a dimer through a suitable linking group that achieves the desired purpose. Examples of linking groups include but are not limited to, urea, amide, -C(0)-C(0)-, carbamate and ketone. In one embodiment, two heteroaryl rings, for example, two indole rings, are linked through a urea to form indole-NHC(0)NH-indole.
Embodiments of Formulas IA, IB, IC, and ID
[0217] To further illustrate the invention, various embodiments of Formula IA, IB, IC and ID are provided. These are presented by way of example to show some of the variations among presented compounds within the invention and can be applied to any of the Formulas I- XXX.
[0218] In one aspect, this disclosure includes compounds and salts of Formula IA:
Figure imgf000056_0001
where R6, R13, and B may carry any of the definitions set forth herein for this variable.
[0219] In another aspect, this disclosure includes compounds and salts of Formula IB, IC, and I
Figure imgf000056_0002
[0220] In Formulas IA, IB, IC, and ID, the variables may include any of the definitions set forth herein that results in a stable compound. Embodiments of Formula VII
[0221] To further illustrate the invention, various embodiments of Formula VII. In one aspect, the disclosure includes compounds and salts of Formula VII:
Figure imgf000057_0001
(VII), wherein:
[0222] R1, R2, R2 , and R3 are independently chosen from hydrogen, halogen, Ci- C4alkyl, Ci-C4alkoxy, -C0-C2alkylNR9R10, -Co-C4alkyl(C3-C7cycloalkyl), -O-C0-C4alkyl(C3- C7cycloalkyl), Ci-C2haloalkyl, and Ci-C2haloalkoxy;
[0223] R8 and R8 are independently chosen from hydrogen, halogen, and methyl;
[0224] R5 is hydrogen, hydroxyl, cyano, -COOH, Ci-C6alkyl, Ci-C6alkoxy, C2- C6alkanoyl -C0-C4alkyl(C3-C7cycloalkyl), -C(O)C0-C4alkyl(C3-C7cycloalkyl, Ci-C2haloalkyl, or C i -C2haloalkoxy ;
[0225] R6 is -C(0)CH3, -C(0)NH2, -C(0)CF3, -C(0)(cyclopropyl), or
-ethyl(cyanoimino); and
[0226] R11 and R14 are independently chosen from hydrogen, halogen, hydroxyl, amino, nitro, cyano, Ci-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, Ci-C6alkoxy, Ci-C6thioalkyl, -C0- C4alkyl(mono- and di-Ci-Cealkylamino), -Co-C4alkyl(C3-C7cycloalkyl), -OCo-C4alkyl(C3- C7cycloalkyl), Ci-C2haloalkyl, and Ci-C2haloalkoxy.
[0227] Prodrugs of Formula I are also within the scope of the disclosure.
III. PHARMACEUTICAL PREPARATIONS
[0228] Compounds disclosed herein can be administered as the neat chemical, but can also administered as a pharmaceutical composition, that includes an effective amount for a host in need of treatment of the selected compound of Formula I, as described herein. Accordingly, the disclosure provides pharmaceutical compositions comprising an effective amount of compound or pharmaceutically acceptable salt of Formula I, together with at least one pharmaceutically acceptable carrier. The pharmaceutical composition may contain a compound or salt of Formula I as the only active agent, or, in an alternative embodiment, Formula I and at least one additional active agent. In certain embodiments the pharmaceutical composition is in a dosage form that contains from about 0.1 mg to about 2000 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg of a compound of Formula I and optionally from about 0.1 mg to about 2000 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg of an additional active agent in a unit dosage form. Examples are dosage forms with at least 25, 50, 100, 200, 250, 300, 400, 500, 600, 700, or 750 mg of active compound, or its salt. The pharmaceutical composition may also include a molar ratio of a compound of Formula I and an additional active agent. For example the pharmaceutical composition may contain a molar ratio of about 0.5 : 1 , about 1 : 1, about 2: 1, about 3 : 1 or from about 1.5:1 to about 4: 1 of an another antiinflammatory agent.
[0229] Compounds disclosed herein may be administered orally, topically, parenterally, by inhalation or spray, sublingually, via implant, including ocular implant, transdermally, via buccal administration, rectally, as an ophthalmic solution, injection, including ocular injection, intraveneous, intra-aortal, intracranial, or by other means, in dosage unit formulations containing conventional pharmaceutically acceptable carriers. The pharmaceutical composition may be formulated as any pharmaceutically useful form, e.g., as an aerosol, a cream, a gel, a pill, a capsule, a tablet, a syrup, a transdermal patch, or an ophthalmic solution. Some dosage forms, such as tablets and capsules, are subdivided into suitably sized unit doses containing appropriate quantities of the active components, e.g., an effective amount to achieve the desired purpose.
[0230] Carriers include excipients and diluents and must be of sufficiently high purity and sufficiently low toxicity to render them suitable for administration to the patient being treated. The carrier can be inert or it can possess pharmaceutical benefits of its own. The amount of carrier employed in conjunction with the compound is sufficient to provide a practical quantity of material for administration per unit dose of the compound.
[0231] Classes of carriers include, but are not limited to binders, buffering agents, coloring agents, diluents, disintegrants, emulsifiers, flavorants, glidents, lubricants, preservatives, stabilizers, surfactants, tableting agents, and wetting agents. Some carriers may be listed in more than one class, for example vegetable oil may be used as a lubricant in some formulations and a diluent in others. Exemplary pharmaceutically acceptable carriers include sugars, starches, celluloses, powdered tragacanth, malt, gelatin; talc, and vegetable oils. Optional active agents may be included in a pharmaceutical composition, which do not substantially interfere with the activity of the compound of the present invention.
[0232] The pharmaceutical compositions/combinations can be formulated for oral administration. These compositions can contain any amount of active compound for Formula I that achieves the desired result, for example between 0.1 and 99 weight % (wt.%) of a compound of Formula I and usually at least about 5 wt.% of a compound of Formula I. Some embodiments contain from about 25 wt.% to about 50 wt. % or from about 5 wt.% to about 75 wt.% of the compound of Formula I.
[0233] The complement factor D inhibitors of the present invention can be administered, for example, either systemically or locally. Systemic administration includes, for example, oral, transdermal, subdermal, intraperitioneal, subcutaneous, transnasal, sublingual, or rectal. Local administration for ocular administration includes: topical, intravitreal, periocular, transscleral, retrobulbar, juxtascleral, sub-tenon, or via an intraocular device. The inhibitors may be delivered via a sustained delivery device implanted intravitreally or transsclerally, or by other known means of local ocular delivery.
IV. METHODS OF TREATMENT
[0234] The compounds and pharmaceutical compositions disclosed herein are useful for treating or preventing a disorder that is mediated by the complement pathway, and in particular, a pathway that is modulated by complement factor D. In certain embodiments, the disorder is an inflammatory disorder, an immune disorder, an autoimmune disorder, or complement factor D related disorders in a host. In one embodiment, the disorder is an ocular disorder. Complement mediated disorders that may be treated or prevented by the compounds and compositions of this disclosure include, but are not limited to, inflammatory effects of sepsis, systemic inflammatory response syndrome (SIRS), ischemia/ reperfusion injury (I/R injury), psoriasis, myasthenia gravis, system lupus erythematosus (SLE), paroxysmal nocturnal hemoglobinuria (PNH), hereditary angioedema, multiple sclerosis, trauma, burn injury, capillary leak syndrome, obesity, diabetes, Alzheimer's dementia, stroke, schizophrenia, epilepsy, age-related macular degeneration, glaucoma, diabetic retinopathy, asthma, allergy, acute respiratory distress syndrome (ARDS), atypical hemolytic uremic syndrome (aHUS), hemolytic uremic syndrome (HUS), cystic fibrosis, myocardial infarction, lupus nephritides, Crohn's disease, rheumatoid arthritis, atherosclerosis, transplant rejection, prevention of fetal loss, biomaterial reactions (e.g. in hemodialysis, inplants), C3 glomerulonephritis, abdominal aortic aneurysm, neuromyelitis optica (NMO), vasculitis, neurological disorders, Guillain Barre Syndrome, traumatic brain injury, Parkinson's disease, disorders of inappropriate or undesirable complement activation, hemodialysis complications, hyperacute allograft rejection, xenograft rejection, interleukin-2 induced toxicity during I L-2 therapy, inflammatory disorders, inflammation of autoimmune diseases, adult respiratory distress syndrome, thermal injury including burns or frostbite, myocarditis, post-ischemic reperfusion conditions, balloon angioplasty, post-pump syndrome in cardiopulmonary bypass or renal bypass, hemodialysis, renal ischemia, mesenteric artery reperfusion after aortic reconstruction, immune complex disorders and autoimmune diseases, SLE nephritis, proliferative nephritis, liver fibrosis, hemolytic anemia, tissue regeneration and neural regeneration. In addition, other known complement related disease are lung disease and disorders such as dyspnea, hemoptysis, chronic obstructive pulmonary disease (COPD), emphysema, pulmonary embolisms and infarcts, pneumonia, fibrogenic dust diseases, inert dusts and minerals (e.g., silicon, coal dust, beryllium, and asbestos), pulmonary fibrosis, organic dust diseases, chemical injury (due to irritant gases and chemicals, e.g., chlorine, phosgene, sulfur dioxide, hydrogen sulfide, nitrogen dioxide, ammonia, and hydrochloric acid), smoke injury, thermal injury (e.g., burn, freeze), bronchoconstriction, hypersensitivity pneumonitis, parasitic diseases, Goodpasture's Syndrome, pulmonary vasculitis, Pauci-immune vasculitis, immune complex- associated inflammation, uveitis (including Behcet's disease and other sub-types of uveitis), antiphospholipid syndrome, arthritis, autoimmune heart disease, inflammatory bowel disease, ischemia-reperfusion injuries, Barraquer-Simons Syndrome, hemodialysis, systemic lupus, lupus erythematosus, transplantation, diseases of the central nervous system and other neurodegenerative conditions, glomerulonephritis (including membrane proliferative glomerulonephritis), blistering cutaneous diseases (including bullous pemphigoid, pemphigus, and epidermolysis bullosa), ocular cicatrical pemphigoid, MPGN II, uveitis, adult macular degeneration, diabetic retinopathy, retinitis pigmentosa, macular edema, Behcet's uveitis, multifocal choroiditis, Vogt-Koyangi-Harada syndrome, imtermediate uveitis, birdshot retino- chorioditis, sympathetic ophthalmia, ocular dicatricial pemphigoid, ocular pemphigus, nonartertic ischemic optic neuropathy, postoperative inflammation, and retinal vein occlusion.
[0235] In some embodiments, complement mediated diseases include ophthalmic diseases (including early or neovascular age-related macular degeneration and geographic atrophy), autoimmune diseases (including arthritis, rheumatoid arthritis), respiratory diseases, cardiovascular diseases. In other embodiments, the compounds of the invention are suitable for use in the treatment of diseases and disorders associated with fatty acid metabolism, including obesity and other metabolic disorders.
[0236] In one embodiment, a method for the treatment of paroxysmal nocturnal hemoglobinuria (PNH) is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. In another embodiment, a method for the treatment of age- related macular degeneration (AMD) is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. In another embodiment, a method for the treatment of rheumatoid arthritis is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. In another embodiment, a method for the treatment of multiple sclerosis is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. In another embodiment, a method for the treatment of myasthenia gravis is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. In another embodiment, a method for the treatment of atypical hemolytic uremic syndrome (aHUS) is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. In another embodiment, a method for the treatment of C3 glomerulonephritis is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. In another embodiment, a method for the treatment of abdominal aortic aneurysm is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. In another embodiment, a method for the treatment of neuromyelitis optica (NMO) is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
[0237] In some embodiments, the present invention provides methods of treating or preventing an inflammatory disorder or a complement related disease, by administering to a host in need thereof an effective amount of a compound of Formula I of the invention. In some embodiments, the present invention provides methods of treating or preventing an inflammatory disorder more generally, an immune disorder, autoimmune disorder, or complement factor D related disease, by providing an effective amount of a compound or pharmaceutically acceptable salt of Formula I to patient with a factor D mediated inflammatory disorder. A compound or salt of Formula I may be provided as the only active agent or may be provided together with one or more additional active agents.
[0238] In one embodiment, a method for the treatment of a disorder associated with a dysfunction in the complement cascade is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. In one embodiment, a method of inhibiting activation of the alternative complement pathway in a subject is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. In one embodiment, a method of modulating factor D activity in a subject is provided that includes the administration of an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier.
[0239] "Prevention" as used in this disclosure means decreasing the likelihood of the appearance of symptoms in a patient administered the compound prophylactically as compared to the likelihood of the appearance of symptoms in patients not administered the compound or decreasing the severity of symptoms in a patient administered the compound prophylactically as compared to the severity of symptoms experienced by patients with the disorder or condition who were not administered the compound. In an alternative embodiment, an effective amount of a compound of Formula I is used to prevent or prophylaxis of a complement factor D related disorder.
[0240] An effective amount of a pharmaceutical composition/ combination of the invention may be an amount sufficient to (a) inhibit the progression of a disorder mediated by the complement pathway, including an inflammatory, immune, including an autoimmune, disorder or complement factor D related disease; (b) cause a regression of an inflammatory, immune, including an autoimmune, disorder or complement factor D related disease; or (c) cause a cure of an inflammatory, immune, including an autoimmune, disorder or complement factor D related disease.
[0241] An effective amount of a compound or pharmaceutical composition described herein will also provide a sufficient amount of the active agent when administered to a patient to provide a clinical benefit. Such an amount may be ascertained experimentally, for example by assaying blood concentration of the agent, or theoretically, by calculating bioavailability.
V. COMBINATION THERAPY
[0242] In one embodiment, a compound or salt of Formula I may be provided in combination or alternation with at least one additional inhibitor of the complement system or a second active compound with a different biological mechanism of action. In one embodiment, a compound or salt of Formula I may be provided in combination with a complement C5 inhibitor or C5 convertase inhibitor. In another embodiment, a compound or salt of Formula I may be provided in combination with eculizumab. In one embodiment, a compound or salt of Formula I may be provided in combination with additional inhibitors of factor D.
[0243] In one embodiment, a compound or salt of Formula I may be provided together with a compound that inhibits an enzyme that metabolizes protease inhibitors. In one embodiment, a compound or salt of Formula I may be provided together with ritonavir.
[0244] In nonlimiting embodiments, a compound or salt of Formula I may be provided together with a protease inhibitor, a soluble complement regulator, a therapeutic antibody (monoclonal or polyclonal), complement component inhibitors, receptor agonists, or siRNAs.
[0245] Nonlimiting examples of active agents in these categories are: [0246] Protease inhibitors: plasma-derived Cl-INH concentrates, for example Cetor® (Sanquin), Berinert-P® (CSL Behring, Lev Pharma), and Cinryze®; and recombinant human CI -inhibitors, for example Rhucin®;
[0247] Soluble complement regulators: Soluble complement receptor 1 (TP 10) (Avant
Immunotherapeutics); sCRl-sLex/TP-20 (Avant Immunotherapeutics); MLN-2222 /CAB-2 (Millenium Pharmaceuticals); Mirococept (Inflazyme Pharmaceuticals);
[0248] Therapeutic antibodies: Eculizumab/Soliris (Alexion Pharmaceuticals); Pexelizumab (Alexion Pharmaceuticals); Ofatumumab (Genmab A/S); TNX-234 (Tanox); TNX- 558 (Tanox); TA106 (Taligen Therapeutics); Neutrazumab (G2 Therapies); Anti-properdin (Novelmed Therapeutics); HuMax-CD38 (Genmab A/S);
[0249] Complement component inhibitors: Compstatin/POT-4 (Potentia Pharmaceuticals); ARC 1905 (Archemix);
[0250] Receptor agonists: PMX-53 (Peptech Ltd.); JPE-137 (Jerini); JSM-7717 (Jerini);
[0251] Others: Recombinant human MBL (rhMBL; Enzon Pharmaceuticals).
[0252] In an embodiment, the present invention provides a method of treating or preventing age-related macular degeneration (AMD) by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention. In one embodiment, the compositions of the present invention are administered in combination with an anti-VEGF agent. Nonlimiting examples of anti-VEGF agents include, but are not limited to, aflibercept (Eylea®; Regeneron Pharmaceuticals); ranibizumab (Lucentis®: Genentech and Novartis); and pegaptanib (Macugen®; OSI Pharmaceuticals and Pfizer); Bevacizumab (Avastin; Genentech/Roche); anecortane acetate, squalamine lactate, and corticosteroids, including, but not limited to, triamcinolone acetonide.
[0253] In another embodiment, a compound of Formula I can be combined with a second agent in order to treat a disorder of the eye.
[0254] Examples of types of therapeutic agents that can be used in combination for ocular applications include anti-inflammatory drugs, antimicrobial agents, anti-angiogenesis agents, immunosuppressants, antibodies, steroids, ocular antihypertensive drugs and combinations thereof. Examples of therapeutic agents include amikacin, anecortane acetate, anthracenedione, anthracycline, an azole, amphotericin B, bevacizumab, camptothecin, cefuroxime, chloramphenicol, chlorhexidine, chlorhexidine digluconate, clortrimazole, a clotrimazole cephalosporin, corticosteroids, dexamethasone, desamethazone, econazole, eftazidime, epipodophyllotoxin, fluconazole, flucytosine, fluoropyrimidines, fluoroquinolines, gatifloxacin, glycopeptides, imidazoles, itraconazole, ivermectin, ketoconazole, levofloxacin, macrolides, miconazole, miconazole nitrate, moxifloxacin, natamycin, neomycin, nystatin, ofloxacin, polyhexamethylene biguanide, prednisolone, prednisolone acetate, pegaptanib, platinum analogues, polymicin B, propamidine isethionate, pyrimidine nucleoside, ranibizumab, squalamine lactate, sulfonamides, triamcinolone, triamcinolone acetonide, triazoles, vancomycin, anti-vascular endothelial growth factor (VEGF) agents, VEGF antibodies, VEGF antibody fragments, vinca alkaloid, timolol, betaxolol, travoprost, latanoprost, bimatoprost, brimonidine, dorzolamide, acetazolamide, pilocarpine, ciprofloxacin, azithromycin, gentamycin, tobramycin, cefazolin, voriconazole, gancyclovir, cidofovir, foscarnet, diclofenac, nepafenac, ketorolac, ibuprofen, indomethacin, fluoromethalone, rimexolone, anecortave, cyclosporine, methotrexate, tacrolimus and combinations thereof. Examples of eye disorders that may be treated according to the compositions and methods disclosed herein include amoebic keratitis, fungal keratitis, bacterial keratitis, viral keratitis, onchorcercal keratitis, bacterial keratoconjunctivitis, viral keratoconjunctivitis, corneal dystrophic diseases, Fuchs' endothelial dystrophy, Sjogren's syndrome, Stevens-Johnson syndrome, autoimmune dry eye diseases, environmental dry eye diseases, corneal neovascularization diseases, post-corneal transplant rejection prophylaxis and treatment, autoimmune uveitis, infectious uveitis, anterior uveitis, posterior uveitis (including toxoplasmosis), pan-uveitis, an inflammatory disease of the vitreous or retina, endophthalmitis prophylaxis and treatment, macular edema, macular degeneration, age related macular degeneration, proliferative and non-proliferative diabetic retinopathy, hypertensive retinopathy, an autoimmune disease of the retina, primary and metastatic intraocular melanoma, other intraocular metastatic tumors, open angle glaucoma, closed angle glaucoma, pigmentary glaucoma and combinations thereof.
[0255] A compound of Formula I, or a combination of Formula I and another active agent, can be administered into an eye compartment of via injection into the vitreous chamber, subretinal space, subchoroidal space, the episclera, the conjunctiva, the sclera, the anterior chamber, and the cornea and compartments therein (e.g., subepithelial, intrastromal, endothelial).
[0256] In an alternative embodiment, a compound of Formula I, or a combination of Formula I and another active agent, can be administered into an eye compartment via binding to a mucosal penetrating particle to treat a condition located in the vitreous chamber, subretinal space, subchoroidal space, the episclera, the conjunctiva, the sclera or the anterior chamber, and the cornea and compartments therein (e.g., subepithelial, intrastromal, endothelial). Mucosal penetrating particles are known in the art, and are described in, for example, PCT published application WO 2013166436 to Kala Pharmaceuticals, incorporated in its entirety herein.
[0257] In other embodiments, a composition comprising compound of Formula I suitable for topical administration to an eye is provided. The pharmaceutical composition comprises a plurality of coated particles, comprising a core particle comprising a compound of Formula I, wherein Formula I constitutes at least about 80 wt% of the core particle, and a coating comprising one or more surface-altering agents, wherein the one or more surface-altering agents comprise at least one of a poloxamer, a poly(vinyl alcohol), or a polysorbate. The one or more surface-altering agents is present on the outer surface of the core particle at a density of at least 0.01 molecules/nm. The one or more surface-altering agents is present in the pharmaceutical composition in an amount of between about 0.001% to about 5% by weight. The plurality of coated particles have an average smallest cross-sectional dimension of less than about 1 micron. The pharmaceutical composition also includes one or more ophthalmically acceptable carriers, additives, and/or diluents.
[0258] It will be appreciated by one of ordinary skill in the art that particles suitable for use with the presently disclosed methods can exist in a variety of shapes, including, but not limited to, spheroids, rods, disks, pyramids, cubes, cylinders, nanohelixes, nanosprings, nanorings, rod- shaped particles, arrow-shaped particles, teardrop-shaped particles, tetrapod- shaped particles, prism-shaped particles, and a plurality of other geometric and non-geometric shapes. In some embodiments, the presently disclosed particles have a spherical shape.
[0259] In one embodiment, the present invention provides a method of treating or preventing paroxysmal nocturnal hemoglobinuria (PNH) by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention. In one embodiment, the present invention provides a method of treating or preventing paroxysmal nocturnal hemoglobinuria (PNH) by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention in combination or alternation with additional inhibitors of the complement system or another active compound with a different biological mechanism of action. In another embodiment, the present invention provides a method of treating or preventing paroxysmal nocturnal hemoglobinuria (PNH) by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention in combination or alternation with eculizumab.
[0260] In one embodiment, the present invention provides a method of treating or preventing rheumatoid arthritis by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention. In one embodiment, the present invention provides a method of treating or preventing rheumatoid arthritis by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention in combination or alternation with an additional inhibitor of the complement system. In another embodiment, the present invention provides a method of treating or preventing rheumatoid arthritis by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention in combination or alternation with methotrexate.
[0261] In certain embodiments, a compound of Formula I is administered in combination or alternation with at least one anti-rhuematoid arthritis drug selected from: salicylates including aspirin (Anacin, Ascriptin, Bayer Aspirin, Ecotrin) and salsalate (Mono-Gesic, Salgesic); nonsteroidal anti-inflammatory drugs (NSAIDs); nonselective inhibitors of the cyclo-oxygenase (COX-1 and COX-2) enzymes, including diclofenac (Cataflam, Voltaren), ibuprofen (Advil, Motrin), ketoprofen (Orudis), naproxen (Aleve, Naprosyn), piroxicam (Feldene), etodolac (Lodine), indomethacin, oxaprozin (Daypro), nabumetone (Relafen), and meloxicam (Mobic); selective cyclo-oxygenase-2 (COX-2) inhibitors including Celecoxib (Celebrex); disease- modifying antirheumatic drugs (DMARDs), including azathioprine (Imuran), cyclosporine (Sandimmune, Neoral), gold salts (Ridaura, Solganal, Aurolate, Myochrysine), hydroxychloroquine (Plaquenil), leflunomide (Arava), methotrexate (Rheumatrex), penicillamine (Cuprimine), and sulfasalazine (Azulfidine); biologic drugs including abatacept (Orencia), etanercept (Enbrel), infliximab (Remicade), adalimumab (Humira), and anakinra (Kineret); corticosteroids including betamethasone (Celestone Soluspan), cortisone (Cortone), dexamethasone (Decadron), methylprednisolone (SoluMedrol, DepoMedrol), prednisolone (Delta-Cortef), prednisone (Deltasone, Orasone), and triamcinolone (Aristocort); gold salts, including Auranofm (Ridaura); Aurothioglucose (Solganal); Aurolate; Myochrysine; or any combination thereof. [0262] In one embodiment, the present invention provides a method of treating or preventing multiple sclerosis by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention. In one embodiment, the present invention provides a method of treating or preventing multiple sclerosis by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention in combination or alternation with additional inhibitors of the complement system. In another embodiment, the present invention provides a method of treating or preventing multiple sclerosis by administering to a subject in need thereof an effective amount of a composition comprising a compound of the current invention in combination or alternation with a corticosteroid. Examples of corticosteroids include, but are not limited to, prednisone, dexamethasone, solumedrol, and methylprednisolone.
[0263] In one embodiment, a compound of Formula I is combined with at least one anti- multiple sclerosis drug selected from: Aubagio (teriflunomide), Avonex (interferon beta- la), Betaseron (interferon beta- lb), Copaxone (glatiramer acetate), Extavia (interferon beta- lb), Gilenya (fmgolimod), Lemtrada (alemtuzumab), Novantrone (mitoxantrone), Plegridy (peginterferon beta- la), Rebif (interferon beta- la), Tecfidera (dimethyl fumarate), Tysabri (natalizumab), Solu-Medrol (methylprednisolone), High-dose oral Deltasone (prednisone), H.P. Acthar Gel (ACTH), and combinations thereof.
[0264] In one aspect, a compound or salt of Formula I may be provided in combination or alternation with an immunosuppressive agent or an anti-inflammatory agent.
[0265] In one embodiment of the present invention, a compound described herein can be administered in combination or alternation with at least one immunosuppressive agent. The immunosuppressive agent as nonlimiting examples, may be a calcineurin inhibitor, e.g. a cyclosporin or an ascomycin, e.g. Cyclosporin A (NEORAL®), FK506 (tacrolimus), pimecrolimus, a mTOR inhibitor, e.g. rapamycin or a derivative thereof, e.g. Sirolimus (RAPAMUNE®), Everolimus (Certican®), temsirolimus, zotarolimus, biolimus-7, biolimus-9, a rapalog, e.g.ridaforolimus, azathioprine, campath 1H, a SIP receptor modulator, e.g. fingolimod or an analogue thereof, an anti IL-8 antibody, mycophenolic acid or a salt thereof, e.g. sodium salt, or a prodrug thereof, e.g. Mycophenolate Mofetil (CELLCEPT®), OKT3 (ORTHOCLONE OKT3®), Prednisone, ATGAM®, THYMOGLOBULIN®, Brequinar Sodium, OKT4, T10B9.A-3A, 33B3.1, 15-deoxyspergualin, tresperimus, Leflunomide ARAVA®, CTLAI-Ig, anti-CD25, anti-IL2R, Basiliximab (SIMULECT®), Daclizumab (ZENAPAX®), mizorbine, methotrexate, dexamethasone, ISAtx-247, SDZ ASM 981 (pimecrolimus, Elidel®), CTLA41g (Abatacept), belatacept, LFA31g, etanercept (sold as Enbrel® by Immunex), adalimumab (Humira®), infliximab (Remicade®), an anti-LFA-1 antibody, natalizumab (Antegren®), Enlimomab, gavilimomab, antithymocyte immunoglobulin, siplizumab, Alefacept efalizumab, pentasa, mesalazine, asacol, codeine phosphate, benorylate, fenbufen, naprosyn, diclofenac, etodolac and indomethacin, aspirin and ibuprofen.
[0266] Examples of anti-inflammatory agents include methotrexate, dexamethasone, dexamethasone alcohol, dexamethasone sodium phosphate, fluromethalone acetate, fluromethalone alcohol, lotoprendol etabonate, medrysone, prednisolone acetate, prednisolone sodium phosphate, difluprednate, rimexolone, hydrocortisone, hydrocortisone acetate, lodoxamide tromethamine, aspirin, ibuprofen, suprofen, piroxicam, meloxicam, flubiprofen, naproxan, ketoprofen, tenoxicam, diclofenac sodium, ketotifen fumarate, diclofenac sodium, nepafenac, bromfenac, flurbiprofen sodium, suprofen, celecoxib, naproxen, rofecoxib, glucocorticoids, diclofenac, and any combination thereof. In one embodiment, a compound of Formula I is combined with one or more non-steroidal anti-inflammatory drugs (NSAIDs) selected from naproxen sodium (Anaprox), celecoxib (Celebrex), sulindac (Clinoril), oxaprozin (Daypro), salsalate (Disalcid), diflunisal (Dolobid), piroxicam (Feldene), indomethacin (Indocin), etodolac (Lodine), meloxicam (Mobic), naproxen (Naprosyn), nabumetone (Relafen), ketorolac tromethamine (Toradol), naproxen/esomeprazole (Vimovo), and diclofenac (Voltaren), and combinations thereof.
VI. PROCESS OF PREPARATION OF COMPOUNDS OF FORMULA I
ABBREVIATIONS
(Boc)20 di-tert-butyl dicarbonate
ACN Acetonitrile
AcOEt, EtOAc ethyl acetate
CH3OH, MeOH Methanol
CsF Cesium fluoride
Cul Cuprous iodide
DCM, CH2C12 Dichloromethane DIEA, DIPEA N,N-diisopropylethylamine
DMA N,N-dimethylacetamide
DMF N,N-dimethylformamide
DMSO Dimethyl sulfoxide
DPPA Diphenyl phosphoryl azide
Et3N, TEA Triethylamine
EtOAc Ethyl acetate
EtOH Ethanol
HATU 1 - [Bis(dimethylamino)methylene] - 1 H- 1 ,2,3 -triazolo
[4,5-b]pyridinium 3-oxide hexafluorophosphate
HC1 Hydrochloric acid
;ΡΓ2ΝΕΪ N,N-diisopropylethylamine
K2C03 Potassium carbonate
LiOH Lithium hydroxide
MTBE Methyl ylether
Na2S04 Sodium sulfate
NaCl Sodium chloride
NaH Sodium hydride
NaHC03 Sodium bicarbonate
NEt3, TEA Triethylamine
Pd (OAc) 2 Palladium acetate
Pd(dppf)Cl2 [1,1 '-Bis(diphenylphosphino) ferrocene] dichloropalladium(II)
Pd(PPh3)2Cl2 Bis(triphenylphosphine)palladium(II) dichloride
Pd(PPh3)4 Tetrakis(triphenylphosphine)palladium(0)
Pd2 (dba) 3 Tris(dibenzylideneacetone)dipalladium(0)
PP 3 Triphenylphosphine
RT Room temperature
TBTU O-Benzotriazolyl tetramethylisouronium tetrafluoroborate tBuOK potassium tert-butoxide
Tf20 trifluoromethanesulfonic anhydride
TFA Trifluoroacetic acid
THF Tetrahydrofuran TMSBr Bromotrimethylsilane
tR Retention time
Zn (CN)2 Zinc cyanide
GENERAL METHODS
[0267] All nonaqueous reactions were performed under an atmosphere of dry argon or nitrogen gas using anhydrous solvents. The progress of reactions and the purity of target compounds were determined using one of the two liquid chromatography (LC) methods listed below. The structure of starting materials, intermediates, and final products was confirmed by standard analytical techniques, including NMR spectroscopy and mass spectrometry.
LC Method A
Instrument: Waters Acquity Ultra Performance LC
Column: ACQUITY UPLC BEH CI 8 2.1 x 50 mm, 1.7 μιη
Column Temperature: 40 °C
Mobile Phase: Solvent A: H20 + 0.05% FA; Solvent B: CH3CN + 0.05% FA
Flow Rate: 0.8 mL/min
Gradient: 0.24 min @ 15% B, 3.26 min gradient (15-85% B), then 0.5 min @ 85% B. Detection: UV (PDA), ELS, and MS (SQ in EI mode)
LC Method B
Instrument: Shimadzu LC-2010A HT
Column: Athena, C18-WP, 50 x 4.6 mm, 5 μιη
Column Temperature: 40 °C
Mobile Phase: Solvent A: H20/CH3OH/FA = 90/10/0.1; Solvent B: H20/CH3OH/FA = 10/90/0.1
Flow Rate: 3 mL/min
Gradient: 0.4 min @ 30% B, 3.4 min gradient (30-100% B), then 0.8 min @ 100% B Detection: UV (220/254 nm) EXAMPLE 1. GENERAL ROUTE OF SYNTHESIS
[0268] A compound of the present invention can be prepared, for example, from a central core. In one embodiment, for example, the central core Structure 1 is an N-protected aminoacid where X1 is nitrogen and PG = protecting group. In one embodiment, the central core is coupled to an amine to generate an amide of Structure 2 (wherein L-B includes a C(0)N moiety). Structure 2 can then be deprotected to generate Structure 3. Structure 3 is coupled to Structure 4 (A-COOH) to generate a second amide bond, forming a compound within Formula I. The chemistry is illustrated in Route 1.
removal of
PR
Figure imgf000072_0001
Structure 2
Figure imgf000072_0002
Formula I
Route 1
[0269] In an alternative embodiment, central core Structure 5 is reacted with a heterocyclic or heteroaryl compound to generate a compound of Structure 6. In one embodiment, Structure 6 is deprotected to generate a carboxylic acid, Structure 7. In one embodiment, Structure 7 is coupled to an amine to generate a compound of Formula I. This chemistry is illustrated in Route 2. 0ΡΘ removal of
Figure imgf000073_0001
Structure 6
Structure 5
Figure imgf000073_0002
Structure 7
Formula I
Route 2
[0270] In an alternative embodiment, Structure 8 is deprotected to generate an amine which is Structure 9. Structure 9 is then coupled to generate an amide which is Structure 6. Structure 6 is then deprotected to generate a carboxylic acid which is Structure 7. Structure 7 is then coupled to form the amide which falls within Formula I. The chemistry is illustrated in Route 3.
Q1 1 Q1 A-COOH (structur
9 V2 nPG removal of Q · 2 .OPG coupling
PG2
. n Structure 9
Structure 8
Figure imgf000074_0001
Structure 6 Structure 7
Formula I
Route 3
[0271] In an alternate embodiment, a heteroaryl or aryl moiety, 4-1, is coupled to a central core to generate 4-2. The protected acid, 4-2 is deblocked to form the carboxylic acid, 4- 3. The carboxylic acid is then coupled to form an amide (L-B) which is 4-4. The heteroaryl or aryl moiety, A', can then be further derivitized to add substituents at the X11, X12, X13 and X14 positions to generate compounds of Formula I. This chemistry is illustrated in Route 4.
Figure imgf000074_0002
B
Figure imgf000074_0003
4-4
Formula I
Route 4 [0272] In an alternate embodiment, Structure 5-1 is coupled to an acid, Structure 5-2, to generate Structure 5-3. The carboxylic acid, Structure 5-3, is deblocked to generate a carboxylic acid which is Structure 5-4. Carboxylic acid Structure 5-4 is coupled to an amine to form the product amide (L-B) which is a compound within Formula I. This chemistry is illustrated in Route 5.
S
Figure imgf000075_0001
tructure 5-1
Structure 5-4
Structure 5-3
Figure imgf000075_0002
Formula I
Route 5
[0273] In an alternate embodiment, Structure 6-1 is coupled to an amine to generate an amide (L-B) which is Structure 6-2. Structure 6-2 is coupled to an amine to generate compounds within Formula I. This chemistr is illustrated in Route 6.
Figure imgf000075_0003
Formula I
Route 6
[0274] In another embodiment, a heteroaryl compound, for example, Structure 7-1 is iodinated to incorporate an iodide group at the R6 position. Other R6 groups can be introduced by those skilled in the art. For example, Structure 7-1 can be acylated to introduce an acyl group at the R6 position. Structure 7-2 is coupled to an activated ester, Structure 7-3, to generate Structure 7-4. In some embodiments, the leaving group, LG, is a halide. Structure 7-4 is treated with inorganic cyanide and two organometallic catalysts to generate a cyano group at the R6 position. In some embodiments, the inorganic cyanide is zinc cyanide. In some embodiments, the organometallic catalyst are Pd(dppf)2 and Pd2(dba)3. Structure 7-4 is treated with an organic acid to generate Structure 7-5. In some embodiments, the organic acid is trifluoroacetic acid. Structure 7-5 is coupled to Structure 3, from Route 1, to generate compounds of Formula I. This chemistry is illustrated in Route 7.
Figure imgf000076_0001
Formula I
Route 7
[0275] In another embodiment, a heteroaryl compound, for example, Structure 8-1, is coupled to an activated ester, Structure 8-2, to generate Structure 8-3. In some embodiments, R6 is C(0)OEt. In some embodiments, the leaving group, LG, is a halide. Structure 8-3 is treated with an organic acid to generate Structure 8-4. In some embodiments, the organic acid is trifluoroacetic acid. Structure 8-4 is coupled to Structure 3 from Route 1 to generate compounds of Formula I. In some embodiments, wherein R6 is C(0)OEt, the ester can be hydrolyzed and the acid used to generate an amide, -C(0)NH2 at the R position. This chemistry is illustrated in Route 8.
Figure imgf000077_0001
Structure 8-1 Structure 8-3 Structure 8-4
Figure imgf000077_0002
Formula I
Route 8
[0276] In another embodiment, a heteroaryl compound, Structure 9-1, is coupled to an activated ester, Structure 9-2, to generate Structure 9-3. In some embodiments, the leaving group, LGi, is a halide. In some embodiments, LG is a halide. In some embodiments, Structure 9-3 is treated with an inorganic cyanide and two organometallic catalyst to generate structure 9- 4. In some embodiments, the inorganic cyanide is zinc cyanide. In some embodiments, the two organometallic catalysts are Pd2(dba)3 and Pd(dppf)Cl2. Structure 9-4 is treated with an organic acid to generate Structure 9-5. In some embodiments, the organic acid is trifluoroacetic acid. Structure 9-5 is coupled to Structure 3 from Route 1 to generate compounds of Formula I. This chemistry is illustrated in Route 9.
Figure imgf000078_0001
Structure 9-5 Formula I
Route 9
[0277] In another embodiment, a heteroaryl compound, Structure 10-1, is coupled to an activated ester, Structure 10-2, to generate Structure 10-3. In some embodiments, the leaving group, LGi, is a halide. In some embodiments, LG is a halide. In some embodiments, Structure 10-3 is treated with an inorganic cyanide and two organometallic catalyst to generate structure 10-4. In some embodiments, the inorganic cyanide is zinc cyanide. In some embodiments, the two organometallic catalysts are Pd2(dba)3 and Pd(dppf)Cl2. Structure 10-4 is treated with an organic acid to generate Structure 10-5. In some embodiments, the organic acid is trifluoroacetic acid. Structure 10-5 is coupled to Structure 3 from Route 1 to generate compounds of Formula I. This chemistry is illustrated in Route 10.
Figure imgf000079_0001
Structure 10-5 Formula I
Route 10
A central core that is directed linked to an aryl or heteroaryl group in place of the L-B region can be prepared according to known processes. For example, a central core is alkylated to generate a central core -A compound comprising a carboxylic acid is reduced to an alcohol and used to generate an alkyl halide. The alkyl halide is treated with a heteroaryl compound to generate a compound of Formula I. This chemistry can be carried out by those skilled in the art of organic chemistry. See for example: Advanced Organic Chemistry: Reactions, Mechanisms and Structure by J. March.
In one embodiment, a pyrrolidine compound is alkylated to generate a pyrrolidine-C(O)- A compound. The product is chlorinated alpha to the pyrrolidine and treated with base to generate an imine. The imine product is treated with a heteroaryl compound to generate a compound of Formula I. This chemistry can be carried out by those skilled in the art of organic chemistry. See for example: Advanced Organic Chemistry: Reactions, Mechanisms and Structure by J. March. In one embodiment, an amino indole is treated with phosgene or a phosgene equivalent to generate a compound of Formula I. This chemistry can be carried out by those skilled in the art of organic chemistry. See for example: Advanced Organic Chemistry: Reactions, Mechanisms and Structure by J. March.
Figure imgf000080_0001
ZA is halogen. [0278] In one embodiment, deuterated L-proline synthons are disclosed. Deuterated s nthons include, but are not limited to, for example, the following compounds:
Figure imgf000081_0001
J
[0279] Structure A can be treated with deuterium oxide to generate Structure B. See, Barraclough, P. et al. Tetrahedron Lett. 2005, 46, 4653-4655; Barraclough, P. et al. Org. Biomol. Chem. 2006, 4, 1483-1491 and WO 2014/037480 (p.103). Structure B can be reduced to generate Structure C. See, Barraclough, P. et al. Tetrahedron Lett. 2005, 46, 4653-4655; Barraclough, P. et al. Org. Biomol. Chem. 2006, 4, 1483-1491. Structure C can be treated with Mitsunobu reaction conditions to generate Structure D. Structure B can be treated with DAST to generate Structure E. See, WO 2014/037480. Structure A can be treated with sodium borodeuteride to generate Structure F. See, Dormoy, J. -R.; Castro, B. Synthesis 1986, 81-82. Compound F can be used to generate Structure K. See, Dormoy, J. -R.; Castro, B. Synthesis 1986, 81-82. Structure B can be treated with a deuterated reducing agent, for example sodium borodeuteride to generate Structure G. Structure G can be treated with DAST to generate Structure H. Structure F can be used to generate Structure K. See, Dormoy, J. -R.; Castro, B. Synthesis 1986, 81-82. Structure G can be used to generate Structure I. Structure J can be prepared according to Hruby, V. J. et al. J. Am. Chem. Soc. 1979, 101, 202-212. Structures A-J can be used to prepare compounds of Formula I.
EXAMPLE 3. PREPARATION OF CENTRAL-L-B SYNTHONS
Figure imgf000082_0001
PG Step 2
(1 b)
Figure imgf000082_0002
(1c)
Figure imgf000082_0003
Routes la, lb and lc.
[0280] In Route la, 5-azaspiro[2.4]heptane-4,5-dicarboxylic acid, 5-(l,l-dimethylethyl) ester, (4S)-, CAS 209269-08-9, can be prepared as described in Tandon, M. et al. Bioorg. Med. Chem. Lett. 1998, 8, 1139-1144. In Step 2, the protected azaspiro[2.4]heptane is coupled to an amine in the presence of an organic solvent, a base and a coupling reagent to generate an amide bond; the L-B moiety. In one embodiment, the amine is (3-chloro-2-fluorophenyl) methanamine. In one embodiment, the organic solvent is DMF. In one embodiment, the base is diisopropylethylamine. In one embodiment, the coupling reagent is HATU. In Step 3, the protecting group is removed. In one embodiment, the starting material is reacted with an acid in the presence of an organic solvent. In one embodiment, the acid is 4N hydrochloric acid. In one embodiment, the organic solvent is dioxane.
[0281] In Route lb, (4S) 4-oxazolidinecarboxylic acid, hydrochloride is treated with an amine protecting reagent. In one embodiment, the amine protecting reagent is di-tert-butyl dicarbonate. In another embodiment, 3,4-oxazolidinedicarboxylic acid, 3-(l,l-dimethylethyl) ester, (4S)-, is commercially available from JPM2 Pharmaceuticals. In one embodiment the reaction is carried out in an organic solvent in the presence of a base. In one embodiment, the organic solvent is acetonitrile. In one embodiment, the base is 4-dimentylaminopyridine (DMAP). In Step 2, the protected 4-oxazolidinecarboxylic acid is coupled to an amine in the presence of an organic solvent, a base and a coupling reagent to generate an amide bond; the L-B moiety. In one embodiment, the amine is (3-chloro-2-fluorophenyl) methanamine. In one embodiment, the organic solvent is DMF. In one embodiment, the base is diisopropylethylamine. In one embodiment, the coupling reagent is HATU. In Step 3, the protecting group is removed. In one embodiment, the starting material is reacted with an acid in the presence of an organic solvent. In one embodiment, the acid is 4N hydrochloric acid. In one embodiment, the organic solvent is dioxane.
[0282] In Route lc, (S)-5-(tert-Butoxycarbonyl)-5-azaspiro[2.4]heptane-6-caboxylic acid, CAS 1129634-44-1, is commercially available from Ark Pharm. In Step 2, the carboxylic acid is coupled to an amine in the presence of an organic solvent, a base and a coupling reagent to generate an amide bond; the L-B moiety. In one embodiment, the amine is (3-chloro-2- fluorophenyl) methanamine. In one embodiment, the organic solvent is DMF. In one embodiment, the base is diisopropylethylamine. In one embodiment, the coupling reagent is HATU. In Step 3, the protecting group is removed. In one embodiment, the starting material is reacted with an acid in the presence of an organic solvent. In one embodiment, the acid is 4N hydrochloric acid. In one embodiment, the organic solvent is dioxane.
Figure imgf000084_0001
Routes 2a, 2b, 2c, and 2d.
[0283] In Route 2a, commercially available Boc-L-proline is coupled to an amine in the presence of an organic solvent, a base and a coupling reagent to generate an amide bond; the L-B moiety. In one embodiment, the amine is (3-chloro-2-fluorophenyl) methanamine. In one embodiment, the organic solvent is DMF. In one embodiment, the base is diisopropylethylamine. In one embodiment, the coupling reagent is HATU. In Step 2, the Boc protecting group is removed. In one embodiment, the starting material is reacted with an acid in the presence of an organic solvent. In one embodiment, the acid is 4N hydrochloric acid. In one embodiment, the organic solvent is dioxane.
[0284] In Route 2b, commercially available (1R, 3S, 5R)-2-[(tert-butoxy)carbonyl]-2- azabicyclo[3.1.0]hexane-3-carboxylic acid, from Enamine, is coupled to an amine in the presence of an organic solvent, a base and a coupling reagent to generate an amide bond; the L-B moiety. In one embodiment, the amine is (3-chloro-2-fluorophenyl) methanamine. In one embodiment, the organic solvent is DMF. In one embodiment, the base is diisopropylethylamine. In one embodiment, the coupling reagent is HATU. In Step 2, the Boc protecting group is removed. In one embodiment, the starting material is reacted with an acid in the presence of an organic solvent. In one embodiment, the acid is 4N hydrochloric acid. In one embodiment, the organic solvent is dioxane.
[0285] In Route 2c, commercially available (2S,4R)-l-(tert-butoxycarbonyl)-4- fluoropyrrolidine-2-carboxylic acid, from Manchester Organics, is coupled to an amine in the presence of an organic solvent, a base and a coupling reagent to generate an amide bond; the L-B moiety. In one embodiment, the amine is (3-chloro-2-fluorophenyl) methanamine. In one embodiment, the organic solvent is DMF. In one embodiment, the base is diisopropylethylamine. In one embodiment, the coupling reagent is HATU. In Step 2, the Boc protecting group is removed. In one embodiment, the starting material is reacted with an acid in the presence of an organic solvent. In one embodiment, the acid is 4N hydrochloric acid. In one embodiment, the organic solvent is dioxane.
[0286] In Route 2d, commercially available (S)-l-(tert-butoxycarbonyl)indoline-2- carboxylic acid, from Chem-Impex, is coupled to an amine in the presence of an organic solvent, a base and a coupling reagent to generate an amide bond; the L-B moiety. In one embodiment, the amine is (3-chloro-2-fluorophenyl) methanamine. In one embodiment, the organic solvent is DMF. In one embodiment, the base is diisopropylethylamine. In one embodiment, the coupling reagent is HATU. In Step 2, the Boc protecting group is removed. In one embodiment, the starting material is reacted with an acid in the presence of an organic solvent. In one embodiment, the acid is 4N hydrochloric acid. In one embodiment, the organic solvent is dioxane. This chemistry is illustrated in Scheme 2.
[0287] Additional starting materials that can readily be converted to Central-L-B- Synthons include, but are not limited to: (S)-l-(tert-butoxycarbonyl)-2,3-dihydro-lH-pyrrole-2- carboxylic acid, CAS 90104-21-5, available from Ark Pharm; cyclopent-l-ene-l,2-dicarboxylic acid, CAS 3128-15-2, purchased from Ark Pharm; imidazole, lH-imidazole-l,2-dicarboxylic acid, l-(l,l-dimethylethyl) 2-ethyl ester, CAS 553650-00-3, commercially available from FCH Group; Boc-L-octahydroindole-2-carboxylic acid can be purchased from Chem Impex. The compound, [0288]
Figure imgf000086_0001
can be prepared according to the procedures disclosed in WO
2004/111041; (S)-Boc-5-oxopyrrolidine-2-carboxylic acid is available from the Aldrich Chemical Co.; (lS,2S,5R)-3-(tert-butoxycarbonyl)-3-azabicyclo[3.3.0]hexane-2-carboxylic acid is available from Ark Pharm; (S)-3-Boc-thiazolidine-2-carboxylic acid is available from Alfa Aesar; (2S,4R)-l-(tert-butoxycarbonyl)-4-chloropyrrolidine-2-carboxylic acid is available from Arch Bioscience; (lS,3aR,6aS)-2-(tert-butoxycarbonyl)octahydrocyclopenta[c]pyrrole-l- carboxylic acid is available from Ark Pharm; 1 ,2-pyrrolidinedicarboxylic acid, 3- [[(phenylmethoxy)carbonyl]amino]-, l-(l,l-dimethylethyl) ester, (2S,3R) can be prepared as disclosed in WO 2004/007501. The Cbz group can be removed and the amino group can be alkylated to generate central core compounds of the present invention.
[0289] The compound
Figure imgf000086_0002
be prepared as disclosed by Braun, J.V.;
Heymons, Albrecht Berichte der Deutschen Chemischen Gesellschaft [Abteilung] B: Abhandlungen (1930) 63B, 502-7.
[0290] The compounds (2S,3S,4S)-4-fluoro-3-methoxy-pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester and (2R,3R,4R)-3-fluoro-4-methoxy-pyrrolidine-l,2-dicarboxylic acid 1-tert- butyl ester can be prepared as a mixture according to WO 2012/093101 to Novartis and the regioisomers can be ultimately separated once coupled to generate the central core-L-B synthons. The compound (S)-Boc-5-oxopyrrolidine-2-carboxylic acid is available from the Aldrich Chemical Co.
EXAMPLE 4. PREPARATION OF A-C(0)-MOIETY
[0291] Examples of the preparation of the A-C(0)-Moiety can be found in Example 1 and below.
[0292] In an alternate embodiment, a heteroaryl compound of Structure 1-1 is acylated to generate Structure 1-2. In an alternate embodiment, Structure 1-1 is treated with an inorganic cyanide and organometallic catalysts to generate a compound where R6 = cyano. The cyano compound can be treated with an oxime to generatre an amide, -C(0)NH2, at the R position. Structure 1-2 is coupled to an activated ester of Structure 1-3 to generate Structure 1-4. In some embodiements, the leaving group, LG, is a halide. The ester is hydrolyzed to generate acid Structure 1-5. This chemistry is illustrated in Route 4a.
Figure imgf000087_0001
Structure 1 -1
Figure imgf000087_0002
Structure 1-2 Structure 1 -4 Structure 1-5 Route 4a
In an alternate embodiment, a heteroaryl compound of Structure 2-1 is acylated to generate Structure 2-2. In an alternate embodiment, Structure 2-1 is treated with an inorganic cyanide and organometallic catalysts to generate a compound where R6 = cyano. The cyano compound can be treated with an oxime to generatre an amide, -C(0)NH2, at the R6 position. Structure 2-2 is coupled to an activated ester of Structure 2-3 to generate Structure 2-4. In some embodiements, the leaving group, LG, is a halide. The ester group in Structure 2-4 is hydrolyzed to generate acid Structure 2-5. This chemistry is illustrated in Route 4b.
Figure imgf000087_0003
Structure 2-1
Figure imgf000087_0004
Structure 2-2 Structure 2-4 Structure 2-5 Route 4b [0293] In an alternate embodiment, a heteroaryl compound of Structure 3-1 is acylated to generate Structure 3-2. In an alternate embodiment, Structure 3-1 is treated with an inorganic cyanide and organometallic catalysts to generate a compound where R6 = cyano. The cyano compound can be treated with an oxime to generate an amide, -C(0)NH2, at the R6 position. Structure 3-2 is coupled to an activated ester of Structure 3-3 to generate Structure 3-4. In some embodiments, the leaving group, LG, is a halide. The ester is hydrolyzed to generate an acid which is Structure 3-5. This chemistry is illustrated in Route 4c.
Figure imgf000088_0001
Structure 3-1
Figure imgf000088_0002
Structure 3-2 Structure 3-4 Structure 3-5 Route 4c
EXAMPLE 5. COUPLING OF CENTRAL-L-B-SYNTHONS TO A-C(0)-MOIETIES
[0294] Examples of the coupling of central-L-B-synthons to A-C(0)-moieties can be found in Example 1 and below.
[0295] In one embodiment, Structure 1-5, from Example 4 Route 4a, is coupled to Structure 3 from Route 1 to generate compounds of Formula I. This chemistry is illustrated in Route 5 a.
Figure imgf000089_0001
Formula I
Route 5 a
[0296] In one embodiment, Structure 2-5 from Example 4 Route 4b is coupled to a central core-L-B to generate compounds of Formula I. This chemistry is illustrated in Route 5b.
Figure imgf000089_0002
Structure 2-5 Formula I
Route 5 b
[0297] In one embodiment, Structure 3-5, from Example 4 Route 4c, is coupled to Structure 3 from Route 1 to generate compounds of Formula I. This chemistry is illustrated in Route 5c.
Figure imgf000089_0003
Formula I
Route 5 c EXAMPLE 6. SYNTHESES OF NON-LIMITING EXAMPLES OF COMPOUNDS OF FORMULA I
Scheme 1. Synthesis of (2S,4R)-N-(2'-chloro-2-fluoro-[l,l'-biphenyl]-3-yl)-4- fluoropyrrolidine-2-carboxamide hydrochloride (lnt-1)
Figure imgf000090_0001
lnt-1
Step 1: 2'-Chloro-2-fluoro-[l,l'-biphenyl]-3-amine hydrochloride (SM3)
[0298] A mixture of SMI (30 g), SM2 (60 g), K2C03 (91 g), and Pd(dppf)2Cl2 (19.25 g) in solvent (dioxane 400 mL, H20 100 mL) was purged with argon in a pressure vessel for 5 min and stirred for 15 h at 100 °C. The solvent was removed under reduced pressure and the remaining residue was purified by column chromatography. The purified material was then dissolved in MeOH and treated with HCl/MeOH. The solvent was removed under reduced pressure and the remaining solid was washed with IPA-heptane (1/1) to afford SM3. Step 2: (2S,4R)-tert-Butyl 2-((2'-chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidine-l-carboxylate (SMS)
[0299] To an ice-cold solution of SM4 (530 mg) in DCM (20 mL) was added 1-chloro- N,N,2-trimethyl-l-propenylamine (0.333 mL, 1.1 equiv) dropwise with stirring. The stirring was continued for 3 h at this temperature and then solid SM3 (640 mg, 1.1 equiv) was added, followed by DIE A (1.12 mL, 3 equiv). The cooling bath was removed and the reaction mixture was stirred overnight at rt. The reaction mixture was then added to water (20 mL) and extracted with DCM (2 x 25 mL). The organic layer was washed successively with an aqueous solution of NaHCC"3 (20 mL), water (20 mL), and brine (20 mL), then dried over Na2SC"4 and concentrated under reduced pressure. The remaining residue was purified by column chromatography (eluted with hexanes/EtOAc) to give SMS.
Step 3: (2S,4R)-N-(2'-Chloro-2-fluoro-[l,l'-biphenyl]-3-yl)-4-fluoropyrrolidine-2- carboxamide hydrochloride (Int-1)
[0300] (2S,4R)-tert-Butyl 2-((2*-chloro-2-fiuoro-[ 1 , 1 *-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidine-l-carboxylate SMS (700 mg) was taken in 4 N HC1 in dioxane (25 mL) and the resulting reaction mixture was stirred at rt for 3 h. The solvent was then removed under reduced pressure and the remaining residue Int-1 was used directly without further purification.
Scheme 2. Synthesis of (2S,4R)-N-(6-bromopyridin-2-yl)-4-fluoropyrrolidine-2- carboxamide TFA salt (Int-2)
Step 1
Figure imgf000092_0001
DCM, 0 °C to rt
Figure imgf000092_0002
Step 1: (2S,4R)-tert-Butyl 2-((6-bromopyridin-2-yl)carbamoyl)-4-fluoropyrrolidine-l- carboxylate (SM8)
[0301] To an ice-cold solution of SM6 (30 g) in DCM (600 mL) was added 1-chloro- N,N,2-trimethyl-l-propenylamine (18. 7 mL, 1.1 equiv) dropwise with stirring. The stirring was continued for 3 h at this temperature. Solid SM7 (24.48 g, 1.1 equiv) was then added, followed by DIEA (67.2 mL, 3 equiv). The cooling bath was removed and the reaction mixture was stirred overnight at rt. The solvent was co-evaporated with MeOH (30 mL). The residue was then dissolved in chloroform (300 mL) and washed successively with cold 1 N aq HC1 (3 χ 200 mL), water (300 mL), and saturated aq NaHC03 (300 mL). The organic layer was dried (Na2S04) and concentrated under reduced pressure. The remaining residue was stirred in a 1 : 1 solution of DCM and heptane (150 mL). The white solid was isolated by filtration and dried under high vacuum to afford SM8 (37. 3 g). Step 2: (2S,4R)-N-(6-Bromopyridin-2-yl)-4-fluoropyrrolidine-2-carboxamide TFA salt (Int- 2)
[0302] SM8 was dissolved in DCM and equal volume of TFA was added. The mixture was stirred for 30 min at rt. The volatiles were removed under reduced pressure and the the residue Int-2 was used without further purification.
Scheme 3: Synthesis of l-(2-((2S,4R)-2-((2'-chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)- -fluoropyrrolidin-l-yl)-2-oxoethyl)-lH-thieno[3,2-c]pyrazole-3-carboxamide (99)
Figure imgf000093_0001
Step 1: lH-Thieno[3,2-c]pyrazole (99b)
[0303] The title compound 99b (310 mg) was prepared from 3-bromothiophene-2- carbaldehyde 99a according to the literature (Airey, J. et al. Synthesis 2014, 96-100). 1H NMR (400 MHz, CDC13, 300 K): δ 7.11 (s, 1H), 7.60 (s, 1H), 7.76 + 8.03 (1H), 13.33 + 13.0 (1H).
Step 2: 3-Iodo-lH-thieno[3,2-c]pyrazole (99c)
[0304] lH-Thieno[3,2-c]pyrazole 99b (310 mg, 2.5 mmol) in DMF (10 mL) was treated with iodine (954 mg, 3.76 mmol), followed by powdered KOH (421 mg, 7.5 mmol). The mixture was stirred overnight at rt. The mixture was diluted with water (50 mL), followed by addition of 10% Na2S203 solution (5 mL). The mixture was extracted with ethyl acetate (3 χ 50 mL). The combined organic extracts were washed with brine and dried over magnesium sulfate. The solution was filtered and concentrated, and the residue 99c was used in the next step without purification.
Step 3: tert-Butyl 2-(3-iodo-lH-thieno[3,2-c]pyrazol-l-yl)acetate (99d)
[0305] The residue 99c from Step 2 was dissolved in CH3CN (50 mL). To this solution, tert-butyl 2-bromoacetate (634 mg, 0.48 mL, 3.25 mmol) and solid potassium carbonate (1.03 g, 7.5 mmol) were added. The mixture was refluxed overnight under an atmosphere of argon. The reaction mixture was cooled to rt and filtered through a pad of Celite®. The solid cake was washed with CH3CN (20 mL), and the combined solution was concentrated under reduced pressure. The remaining residue was purified by column chromatography to afford pure compound 99d (610 mg) and impure isomer tert-butyl 2-(3-iodo-2H-thieno[3,2-c]pyrazol-2- yl)acetate (130 mg). 1H NMR (400MHz, CDC13): δ 1.45 (s, 9H), 4.93 (s, 2H), 6.93 (d, J = 5.2 Hz, 1H), 7.42 (d, J = 5.2 Hz, 1H). LC/MS (EI) m/z: [M + H]+ 365.
Step 4: tert-Butyl 2-(3-cyano-lH-thieno[3,2-c]pyrazol-l-yl)acetate(99e)
[0306] To a degassed solution of t-butyl 2-(3-iodo-lH-thieno[3,2-c]pyrazol-l-yl)acetate 99d (610 mg, 1.67 mmol) in the co-solvents DMF (14 mL) and water (2 mL) were added Zn(CN)2 (235 mg, 2.0 mmol), Pd(dppf)2 (125 mg, 0.17 mmol), and Pd2(dba)3 (156 mg, 0.17 mmol) under an atmosphere of argon. The mixture was heated at 1 10 °C for 6 h. The reaction mixture was cooled to rt and the volatiles were removed under reduced pressure. The remaining residue was diluted with ethyl acetate (50 mL) and filtered through a pad of Celite®. The solid was washed with ethyl acetate (30 mL). The combined organic solution was concentrated under reduced pressure and the remaining residue was purified by column chromatography to afford the title compound 99e (290 mg). 1H NMR (400MHz, CDC13, 300 K): δ 1.47 (s, 9H), 5.0 (s, 2H), 6.93 (d, J = 5.2 Hz, 1H), 7.52 (d, J = 5.2 Hz, 1H). LC/MS (EI) m/z: [M + H]+ 264.
Step 5: 2-(3-Carbamoyl-lH-thieno[3,2-c]pyrazol-l-yl)acetic acid (99f)
[0307] A solution of tert-butyl 2-(3-cyano-lH-thieno[3,2-c]pyrazol-l-yl)acetate (59 mg, 0.22 mmol) in TFA (2 mL) was subjected to microwave irradiation at 140 °C for 30 min. The mixture was concentrated under reduced pressure, and the residue was co-evaporated with toluene (10 mL) twice. The dried residue 99f was used directly in the next step. LC/MS (EI) m/z: 226.
Step 6: l-(2-((2S,4R)-2-((2'-Chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-lH-thieno[3,2-c]pyrazole-3-carboxamide (99)
[0308] To a solution of the residue 99f (0.22 mmol) from Step 5 was added (2S,4R)-N- (2'-chloro-2-fluoro-[ 1 , 1 '-biphenyl]-3-yl)-4-fluoropyrrolidine-2-carboxamide hydrochloride Int-1 (90 mg, 0.24 mmol) in DMF (2 mL) followed by HATU (109 mg, 0.29 mmol) and dropwise addition of DIEA (0.3 mL) at rt. The reaction mixture was stirred for 1 h at rt and the volatiles were removed under reduced pressure. The residue was diluted with 20 mL of 10% sodium carbonate and extracted with ethyl acetate (3 χ 20 mL). The combined organic solution was washed with water and brine, and then dried over MgSC^. The solution was filtered and the solvent was removed under reduced pressure. The remaining residue was purified by column chromatography to afford 99 (50.7 mg). 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 2.00-2.19 (m, 1H), 2.47-2.53 (m, 1H), 3.75-3.88 (m, 1H), 4.02-4.11 (m, 1H), 4.70 (t, J = 8.8 Hz, 1H), 5.26 (d, J = 17.2 Hz, 1H), 5.38-5.49 (m, 2H), 6.97-7.01 (m, 1H), 7.05 (d, J = 5.2 Hz, 1H), 7.15 (t, J = 7.6 Hz, 1H), 7.30-7.41 (m, 4H), 7.51-7.54 (m, 2H), 7.60-7.62 (m, 1H), 7.89-7.93 (m, 1H), 9.92(s, 1H); 19F NMR (376 MHz, DMSO-d6, 300K): (major rotamer) δ -126.8, -175.8. LC (method A): tR = 1.94 min. LC/MS (EI) m/z: [M + H]+ 544.
Figure imgf000095_0001
[0309] To a solution of 2-(3 -carbamoyl- lH-thieno [3, 2-c]pyrazol-l-yl)acetic acid 99f (0.193 mmol) was added (2S,4R)-N-(6-bromopyridin-2-yl)-4-fluoropyrrolidine-2-carboxamide hydrochloride (0.212 mmol) in DMF(1.5 mL) followed by TBTU (93 mg, 0.29 mmol) and dropwise addition of DIEA (0.21 mL, 1.2 mmol). The reaction mixture was stirred for 0.5 h at rt , then quenched by aq NaHC03 (5 mL). The precipitate was collected by filtration and purified by column chromatography (eluent: 5% MeOH in DCM) to give 106 (68 mg). 1H NMR (400 MHz, CDC13-CD30D, 300 K): (major rotamer) δ 2.37 (m, 1H), 2.39-2.41 (m, 1H), 2.59-2.70 (m, 1H), 3.65 (dd, J = 36.0, 12.4 Hz, 1H), 3.90-3.98 (dd, J = 20.0, 12.0 Hz, 1H), 4.80 (t, J = 8.0 Hz, 1H), 5.18 (d, J = 8.0 Hz, 1H), 5.4 (m, 1H), 6.96 (d, J = 8.0 Hz, 1H), 7.22 (d, J = 8.0 Hz, 1H), 7.50 (d, J = 4.0Hz, 1H), 7.55 (t, J = 8.0 Hz, 1 H), 8.14 (d, J = 8.0 Hz, 1H). 31F NMR (376 MHz, CDC13- CD3OD, 300 K): (major rotamer) δ -177.0. LC (method A): tR = 1.33 min. LC/MS (EI) mlz: [M + H]+ 497.
Scheme 5: Synthesis of l-(2-((lR,3S,5R)-3-((3-chloro-2-fluorobenzyl)carbamoyl)-2- azabicyclo 3.1.0]hexan-2-yl)-2-oxoethyl)-lH-thieno[3,2-c]pyrazole-3-carboxamide (109)
Figure imgf000096_0001
[0310] To a solution of 2-(3 -carbamoyl- lH-thieno [3, 2-c]pyrazol-l-yl)acetic acid 99f (0.287 mmol) was added (lR,3S,5R)-N-(3-chloro-2-fluorobenzyl)-2-azabicyclo[3.1.0]hexane-3- carboxamide hydrochloride (48 mg, 0.158 mmol) in DMF (1.5 mL) followed by TBTU (69 mg, 0.216 mmol) and dropwise addition of DIEA (0.13 mL, 0.75 mmol). The reaction mixture was stirred for 0.5 h at rt , then quenched by aq NaHC03 (5 mL). The mixture was extracted with EtOAc (30 mL) and the organic layer was washed with water and brine. The solvent was removed under reduced pressure and the remaining residue was purified by column chromatography (eluent: 5% MeOH in DCM) to give 109 (57 mg). 1H NMR (400 MHz, CDC13, 300 K): (major rotamer) δ 0.59-0.61 (m, 1H), 1.08-1.12 (m, 1H), 1.95-2.06 (m, 3H), 2.70-2.72 (m, 1H), 3.34-3.62 (m, 1H), 4.41 (s, 2H), 4.57-4.60 (m, 1H), 5.21-5.32 (m, 2H), 6.00 (s, 1H), 6.70 (s, 1H), 6.84-6.85 (m, 1H), 6.92-6.94 (m, 1 H), 7.11-7.13 (m, 1H), 7.21-7.26 (m, 1H), 7.36- 7.38 (m, 1H), 7.43 (m, 1H). 13C NMR (100 MHz, CDC13, 300 K): (major rotamer) δ 19.40, 20.74, 29.97, 37.14, 37.52, 53.43, 65.54, 108.95, 120.76, 120.94, 123.07, 124.58, 124.63, 126.83, 126.98, 127.93, 129.56, 134.78, 137.27, 150.91, 154.84, 157.30, 163.16, 167.60,170.45. 31F NMR (376 MHz, CDC13, 300 K): (major rotamer) δ -120.9. LC (method A): tR = 1.49 min. LC/MS (EI) mlz: [M + H]+ 476.
Scheme 6: Synthesis of l-(2-((2S,4R)-2-((2'-chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-
4-fluoropyrrolidin-l-yl)-2-oxoethyl)-5-methyl-lH-thieno[3,2-c]pyrazole-3-carboxamide
Figure imgf000097_0001
Step 1: 3-Bromo-5-methylthiophene-2-carbaldehyde (113b)
[0311] To a solution of 4-bromo-2-methylthiophene 113a (11 g, 62 mmol) in anhydrous THF (100 mL) cooled to -78 °C was added dropwise a 2 M LDA-THF solution (34 mL, 68 mmol) under an atmosphere of argon. The mixture was stirred for 2 h at -78 °C and then anhydrous DMF (9.6 mL, 124 mmol) was added. The reaction mixture was kept at -78 °C for 1 h and quenched with 10% aq citric acid (10 mL). The volatiles were removed under reduced pressure and the remaining residue was diluted with ethyl acetate (150 mL). The organic layer was successively washed with 5% aq citric acid (80 mL), water (100 mL), and brine (50 mL), and then dried over MgS04. The solution was filtered and concentrated under reduced pressure to afford 113b (12.3 g) as a pale yellow oil, which was used in the next step without further purification.
Step 2: 5-Methyl-lH-thieno[3,2-c]pyrazole (113c)
[0312] The title compound 113c (3.0g) was prepared from 3-bromo-5-methylthiophene- 2-carbaldehyde 113b (12.2 g) in a manner similar to that reported in the literature (Airey, J. et al. Synthesis 2014, 96-100). 1H NMR (400 MHz, CDC13, 300 K): δ 2.49 (s, 3H), 6.86 (s, 1H), 7.65 + 7.89 (1H), 12.83 + 13.10 (1H). LC/MS (EI) mlz: [M + H]+ 139.
Step 3: 3-Iodo-5-methyl-lH-thieno[3,2-c]pyrazole (113d)
[0313] The title compound 113d was prepared from 5-methyl-lH-thieno[3,2-c]pyrazole (1.0 g) in a manner similar to that described in Step 2 of Scheme 3.
Step 4: tert-Butyl 2-(3-iodo-5-methyl-lH-thieno[3,2-c]pyrazol-l-yl)acetate (113e)
[0314] The title compound 113e was prepared from 3 -iodo-5 -methyl- lH-thieno [3,2- c]pyrazole in a similar manner as described in Step 3 of Scheme 3. 1H NMR (400MHz, CDC13): 1.46 (s, 9H), 2.53 (s, 3H), 4.87 (s, 2H), 6.64 (s, 1H) ppm. MH+ 379.09. Minor product is tert-butyl 2-(3-iodo-5-methyl-2H-thieno[3,2-c]pyrazol-2-yl)acetate. 1H NMR (400 MHz, CDC13, 300 K): δ 1.47 (s, 9H), 2.51 (s, 3H), 4.98 (s, 2H), 6.80 (s, 1H). LC/MS (EI) mlz: [M + H]+ 379. Step 5: tert-Butyl 2-(3-cyano-5-methyl-lH-thieno[3,2-c]pyrazol-l-yl)acetate (113f)
[0315] The title compound 113f was prepared from tert-butyl 2-(3 -iodo-5 -methyl- 1 H- thieno[3,2-c]pyrazol-l-yl)acetate (823 mg, 2.15 mmol) in a manner similar to that described in Step 4 of Scheme 3. 1H NMR (400 MHz, CDC13, 300 K): δ 1.47 (s, 9H), 2.56 (s, 3H), 4.93 (s, 2H), 6.65 (s, IH). LC/MS (EI) mlz: [M + H]+ 278.
Step 6: 2-(3-Carbamoyl-5-methyl-lH-thieno[3,2-c]pyrazol-l-yl)acetic acid (113g)
[0316] The title compound 113g was prepared from tert-butyl 2-(3 -cyano-5 -methyl- 1 H- thieno[3,2-c]pyrazol-l-yl)acetate (143 mg) in a manner similar to that described in Step 5 of Scheme 3. LC/MS (EI) mlz: [M + H]+ 240.
Step 7: l-(2-((2S,4R)-2-((2'-Chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-5-methyl-lH-thieno[3,2-c]pyrazole-3-carboxamide (113)
[0317] The title compound 113 (86.9 mg) was prepared from 2-(3-carbamoyl-5-methyl- lH-thieno[3,2-c]pyrazol-l-yl)acetic acid (105 mg, 0.3 mmol) in a manner similar to that described in Step 6 of Scheme 3. 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 2.06-2.19 (m, IH), 2.39 (s, 3H), 2.40-2.55 (m, IH), 3.74-3.87 (m, IH), 4.02-4.10 (m, IH), 4.69 (t, J = 8.8 Hz, IH), 5.18 (d, J = 17.2 Hz, IH), 5.31-5.51 (m, 2H), 6.79 (s, IH), 7.0 (t, J = 7.2 Hz, IH), 7.15 (t, J = 8.0 Hz, IH), 7.24-7.46 (m, 4H), 7.50-7.53 (m, 2H), 7.87-7.92 (m, IH), 9.92 (s, IH); 19F NMR (376 MHz, DMSO-d6, 300K): (major rotamer) δ -126.71, -175.87. LC (method A): tR = 2.10 min. LC/MS (EI) mlz: [M + H]+ 558.
Scheme 7: Synthesis of l-(2-((2S,4R)-2-((2'-chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)- -fluoropyrrolidin-l-yl)-2-oxoethyl)-4,5,6,7-tetrahydro-lH-indazole-3-carboxamide (104)
Figure imgf000100_0001
Step 1: Ethyl l-(2-(tert-butoxy)-2-oxoethyl)-4,5,6,7-tetrahydro-lH-indazole-3-carboxylate (104b)
[0318] To a solution of ethyl 4,5,6,7-tetrahydro-lH-indazole-3-carboxylate 104a (1.03g, 5.35 mmol) in CH3CN (50 mL) was added tert-butyl 2-bromoacetate (1.17g, 0.89 niL, 6.0 mmol) and potassium carbonate (1.5 g, 10.6 mmol). The mixture was refluxed overnight under an atmosphere of argon. LC-MS analysis indicated two isomers were formed in a ratio of 4: 1. The reaction mixture was cooled to rt and filtered through a pad of Celite®. The solid cake was washed with CH3CN (20 mL), and the combined solution was concentrated under reduced pressure. The remaining residue was purified by column chromatography to afford the major isomer ethyl l-(2-(tert-butoxy)-2-oxoethyl)-4,5,6,7-tetrahydro-lH-indazole-3-carboxylate 104b (1.37 g) and the minor isomer ethyl 2-(2-(tert-butoxy)-2-oxoethyl)-4,5,6,7-tetrahydro-2H- indazole-3-carboxylate 104c (0.28 g). 1H NMR (400 MHz, CDC13, 300 K): (major isomer) δ 1.38 (t, J = 7.2 Hz, 3H), 1.45 (s, 9H), 1.75 (m, 2H), 1.82 (m, 2H), 2.52 (t, J = 5.2 Hz, 2H), 2.75 (t, J = 5.2 Hz, 2H), 4.37 (q, J = 7.2 Hz, 2H), 4.77 (s, 2H). LC/MS (EI) m/z: 309. 1H NMR (400 MHz, CDC13, 300 K): (minor isomer) δ 1.35 (t, J = 7.2 Hz, 3H), 1.47 (s, 9H), 1.74-1.80 (m, 4H), 2.67 (t, J = 5.2 Hz, 2H), 2.75 (t, J = 5.2 Hz, 2H), 4.30 (q, J = 7.2 Hz, 2H), 5.11 (s, 2H). LC/MS (EI) m/z: [M + H]+ 309.
Step 2: 2-(3-(Ethoxycarbonyl)-4,5,6,7-tetrahydro-lH-indazol-l-yl)acetic acid (104d)
[0319] Ethyl l-(2-(tert-butoxy)-2-oxoethyl)-4,5,6,7-tetrahydro-lH-indazole-3- carboxylate 104b (1.35 g, 4.38 mmol) was treated with TFA (5 mL) and DCM (5 mL) and stirred overnight at rt. The volatiles were removed under reduced pressure and the remaining residue was co-evaporated with toluene (10 mL) twice. The dried residue 104d was used directly in the next step.
Step 3: Ethyl l-(2-((2S,4R)-2-((2'-chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-4,5,6,7-tetrahydro-lH-indazole-3-carboxylate (104e)
[0320] To a solution of the residue 104d from Step 2 (375 mg, 1.5 mmol) was added (2S,4R)-N-(2'-chloro-2-fluoro-[ 1 , 1 '-biphenyl]-3-yl)-4-fluoropyrrolidine-2-carboxamide hydrochloride (560 mg, 1.5 mmol) in DMF (2 mL) followed by HATU (741 mg, 1.95 mmol) and dropwise addition of DIEA (4.5 mmol, 0.78 mL). The mixture was stirred for 1 h at rt and the volatiles were removed under reduced pressure. The remaining residue was diluted with 10% aq sodium carbonate (20 mL) and water (50 mL), then extracted with ethyl acetate (3 χ 50 mL). The combined organic extract was washed with water and brine, then dried over MgS04. The solution was filtered and the solvent was removed under reduced pressure. The remaining residue was purified by column chromatography to afford the title compound 104e (608 mg). LC/MS (EI) m/z: [M + H]+ 571.
Step 4: l-(2-((2S,4R)-2-((2'-Chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-4,5,6,7-tetrahydro-lH-indazole-3-carboxylic acid (104f)
[0321] Compound 104e from Step 3 (608 mg, 1.06 mmol) was dissolved in a mixture of MeOH-THF-H20 (3 mL-3 mL-3 mL) and treated with LiOH (100 mg, 4.25 mmol). The reaction mixture was stirred overnight at rt. The volatiles were removed under reduced pressure and the remaining residue was acidified with 10% aq citric acid (10 mL). The solid 104f was collected by filtration, washed with water, and dried in vacuo for use in the next step. LC/MS (EI) m/z: [M + H]+ 543.
Step 5: l-(2-((2S,4R)-2-((2'-Chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-4,5,6,7-tetrahydro-lH-indazole-3-carboxamide (104)
[0322] Acid 104f (252 mg, 0.46 mmol) from Step 4 was mixed with NH4C1 (125 mg, 2.32 mmol) in DMF (3 mL). To this solution was added HATU (262 mg, 0.69 mmol) followed by dropwise addition of DIEA (1.38 mmol, 0.24 mL). The mixture was stirred for 3 h at rt and the volatiles were removed under reduced pressure. The remaining residue was diluted with 10%> aq sodium carbonate (15 mL) and water (15 mL), and then extracted with ethyl acetate (3 χ 25 mL). The combined organic solution was washed with water and brine, and then dried over MgS04. The mixture was filtered and the filtrate was concentrated under reduced pressure. The remaining residue was purified by column chromatography to afford the title compound 104 (160 mg). 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 1.53-1.61 (m, 4H), 1.99- 2.16 (m, 1H), 2.38-2.41 (m, 3H), 2.54-2.56 (m, 2H), 3.40-3.53 (m, 1H), 3.93-4.07 (m, 1H), 4.68 (t, J = 8.8 Hz, 1H), 4.90-5.14 (m, 2H), 5.36-5.49 (1H), 6.93-7.19 (m, 4H), 7.32-7.43 (m, 3H), 7.52-7.54 (m, 1H), 7.92 (t, J = 6.8 Hz, 1H), 9.90 (s, 1H); 19F NMR (376 MHz, DMSO-d6, 300K): (major rotamer) δ -126.8, -176.06. LC (method A): tR = 2.02 min. LC/MS (EI) m/z: [M + H]+ 542. Scheme 8: Synthesis of (2S,4R)-l-(2-(3-acetyl-8-(3-chlorophenyl)-lH-imidazo[l,2- a]pyrazolo[3,4-c]pyridin-l-yl)acetyl)-N-(6-bromopyridin-2-yl)-4-fluoropyrrolidine-2- carboxamide (105)
Figure imgf000103_0001
4 A mol. sieves
H
Figure imgf000103_0002
Step 1: Methyl 2-(3-acetyl-8-(3-chlorophenyl)-lH-pyrazolo[4,3-g]indolizin-l-yl)acetate (105c)
[0323] A mixture of l-(l-azidovinyl)-3-chlorobenzene 105a (0.05 g) ( prepared as reported by Donthiri et al. J. Org. Chem 2014, 79, 11277-11284), methyl 2-(3 -acetyl- 1 H- pyrazolo[3,4-c]pyridin-l-yl)acetate 105b (0.195 g), Cul (1 mg) and 4 A molecular sieves was placed in a vial and dry acetonitrile (3 mL) was added. The sealed vial was heated at 65 °C for 24 h. The reaction mixture was allowed to cool to rt and the solvent was removed under reduced pressure. The remaining residue was purified by column chromatography (eluent: DCM) to give 105c (20 mg) as a light orange solid. Step 2: 2-(3-Acetyl-8-(3-chlorophenyl)-lH-pyrazolo[4,3-g]indolizin-l-yl)acetic acid (105d)
[0324] The solid 105c (20 mg, 0.052 mmol) from Step 1 was taken in THF (3 mL), water (0.3 mL), and MeOH (1 mL), and then 1 N aq NaOH (1 mL) was added. The resulting mixture was stirred at rt overnight and concentrated under reduced pressure. The remaining residue was diluted with EtOAc and water. The organic layer was separated and the aq layer was extracted repeatedly with EtOAc. The combined organic layer was dried and concentrated to give 105d as a light yellow solid, which was used as such in the next step.
Step 3: (2S,4R)-l-(2-(3-Acetyl-8-(3-chlorophenyl)-lH-imidazo[l,2-a]pyrazolo[3,4-c]pyridin- l-yl)acetyl)-N-(6-bromopyridin-2-yl)-4-fluoropyrrolidine-2-carboxamide (105)
[0325] To a solution of 2-(3-acetyl-8-(3-chlorophenyl)-lH-pyrazolo[4,3-g]indolizin-l- yl)acetic acid 105d (0.052 mmol) and (2S,4R)-N-(6-bromopyridin-2-yl)-4-fluoropyrrolidine-2- carboxamide TFA salt (0.05 mmol) in DMF (1 mL) cooled to 0-5 °C was added DIEA (47 μί). HATU (24 mg) was then added to this cooled solution and the cooling bath was removed. The reaction mixture was stirred at rt for 30 min and poured into water (10 mL) with stirring. The solid was isolated by filtration and purified by column chromatography (eluent: 0-1.5% MeOH in DCM) to afford 105 (10 mg) as light yellow solid. 1H NMR (400 MHz, CDC13, 300 K): (major rotamer) δ 2.48-2.52 (m, 1H), 2.73 (s, 3H), 2.78-2.94 (m, 1H), 4.04-4.27 (m, 2H), 4.95 (t, J = 7.6 Hz, 1H), 5.51 (d, J = 52.4 Hz, 1H), 5.77 (d, J = 16 Hz, 1H), 5.88 (d, J = 16 Hz, 1H), 6.99 (d, J = 8.4 Hz, 1H), 7.19-7.23 (m, 2H), 7.54-7.56 (m, 1H), 7.69-7.71 (m, 2H), 7.75 (d, J = 8 Hz, 1H), 7.85 (s, 1H), 7.87 (d, J = 7.2 Hz, 1H), 8.97 (br s, 1H); 31F NMR (376 MHz, CDC13, 300 K): (major rotamer) δ -176.04; LC (method A): tR = 2.60 min. LC/MS (EI) mlz: [M + H]+ 640.
Scheme 9: Synthesis of l-(2-((2S,4R)-2-((2'-chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)- 4-fluoropyrrolidin-l-yl)-2-oxoethyl)-7-cyano-lH-indazole-3-carboxamide (111)
Figure imgf000105_0001
Step 1: 7-Bromo-lH-indazole-3-carboxamide (111b)
[0326] To a solution of bromoindazole 111a (3 g) in DMF (30 mL) were added ammonium chloride (1.8 g) and DIEA (10.38 mL). The reaction mixture was cooled in an ice bath and then HATU (5.7 g) was added. The reaction mixture was warmed to rt and stirred overnight. The reaction mixture was poured into water (300 mL) containing LiOH (630 mg). The product was isolated by filtration and washed with water. The tan colored solid was dried under high vacuum to give 111b (2.15 g). Step 2: tert-Buty\ 2-(7-bromo-3-carbamoyl-lH-indazol-l-yl)acetate (111c)
[0327] A mixture of 7-bromo-lH-indazole-3-carboxamidelllb (2.15 g), tert-butyl bromoacetate (1.45 mL), and potassium carbonate (1.36 g) in anhydrous acetonitrile (40 mL) was refluxed for 2 h. The reaction mixture was then cooled to rt and the solvent was removed under reduced pressure. The remaining residue was sonicated with water (40 mL) and filtered. The solid obtained was washed thoroughly with water and further washed with 2:1 mixture of tert-butyl methyl ether and heptane (30 mL) and dried under high vacuum to give 111c (2.5 g).
Step 3: tert- utyl 2-(3-carbamoyl-7-cyano-lH-indazol-l-yl)acetate (Hid)
[0328] A mixture of tert-butyl 2-(7-bromo-3-carbamoyl-lH-indazol-l-yl)acetatelllc (0.5 g), zinc cyanide (0.198 g), Pd(dppf)Cl2 (0.115 g), and Pd2(dba)3 (0.129 g) in DMF (5 mL) and water (1 mL) was bubbled with argon for 5 min. The vial was then sealed and heated at 80 °C for 3 h. After cooling the reaction mixture to rt, it was diluted with EtOAc. The mixture was then washed swith water and a saturated aq NaHC03 solution. The separated organic layer was dried (Na2S04) and concentrated. The remaining residue was purified by column chromatography (0-2% MeOH in DCM) to give a slightly yellow solid (0.3 g). The solid was then washed with ether (3 mL) to give Hid (0.23 g) as a colorless solid.
Step 4: 2-(3-Carbamoyl-7-cyano-lH-indazol-l-yl)acetic acid (Hie)
[0329] The title compound llle was prepared from tert-butyl 2-(3-carbamoyl-7-cyano- lH-indazol-l-yl)acetate (50 mg) in a manner similar to that described in Step 2 of Scheme 7, and used in the next step without further purification.
Step 5: l-(2-((2S,4R)-2-((2'-Chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-7-cyano-lH-indazole-3-carboxamid (111)
[0330] 2-(3-Carbamoyl-7-cyano-lH-indazol-l-yl)acetic acid llle from Step 4 above was coupled with (2S,4R)-N-(2'-chloro-2-fluoro-[ 1 , 1 '-biphenyl]-3-yl)-4-fluoropyrrolidine-2- carboxamide hydrochloride using the procedure described in Step 6 of Scheme 3. The crude product was purified by column chromatography (0-2% MeOH in DCM) to afford the title compound 111 (50 mg). 1H NMR (400 MHz, CD3OD, 300 K): (major rotamer) δ 2.65-2.75 (m, 1H), 4.1-4.14 (m, 1H), 4.16-4.24 (m, 1H), 4.89 (t, J = 8.4 Hz, 1H), 5.52 (d, J = 52.8 Hz, 1H), 5.73 (d, J = 17.6 Hz, 1H), 5.89 (d, J = 17.6 Hz, 1H), 7.07 (t, J = 7.2 Hz, 1H), 7.19 (t, J = 8 Hz, 1H), 7.28-7.44 (m, 5H), 7.49-7.59 (m, 1H), 7.87 (d, J = 7.2 Hz, 1H), 7.91 (s, 1H), 7.97 (t, J = 7.2 Hz, 1H), 8.59 (d, J = 8 Hz, 1H). ). 31F NMR (376 MHz, CD3OD, 300 K): (major rotamer) δ -178.5, -128.8. LC (method A): tR = 2.09 min. LC/MS (EI) m/z: [M + H]+ 563.
Scheme 10: l-il-iilS^RJ-l-iiS'-Acetyl-l'-chloro-l-fluoro-Il^'-bi henyll-S-y carbamoyl)- 4-fluoropyrrolidin-l-yl)-2-oxoethyl)-lH-indazole-3-carboxamide (117)
Figure imgf000107_0001
Step 1: l-(3'-Amino-6-chloro-2'-fluoro-[l,l'-biphenyl]-3-yl)ethanone (117c)
[0331] To a mixture of 3-bromo-2-fluoroanilinell7a (1.0 g), (5-acetyl-2- chlorophenyl)boronic acid 117b (3.132 g), Pd(dppf)Cl2 (0.860 g), and K2C03 (3.64 g) under an atmosphere of argon was added dioxane (40 mL) and water (10 mL). The mixture was bubbled with argon for 5 min and heated with stirring at 100 °C overnight. The reaction mixture was then filtered through a pad of Celite® and the filtrate was concentrated under reduced pressure. The remaining residue was purified by column chromatography (0-0.5% MeOH in DCM) to afford 117c (0.9 g) as orange-yellow oil. Step 2: (2S,4R)-tert-Butyl 2-((5'-acetyl-2,-chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)- 4-fluoropyrrolidine-l-carboxylate (117d)
[0332] To an ice-cold solution of (2S,4R)-l-(tert-butoxycarbonyl)-4-fluoropyrrolidine-2- carboxylic acid (0.25 g, 1.07 mmol) in DCM (15 mL) was added l-chloro-N,N,2-trimethylprop- 1 -en- 1 -amine (1.2 mmol, 0.16 mL, 1.1 equiv) dropwise with stirring. The stirring was continued for 3 h at this temperature. Solid l-(3'-amino-6-chloro-2'-fluoro-[l, -biphenyl]-3-yl)ethanone 117c (256 mg, 0.97 mmol) was then added, followed by DIE A (0.56 mL, 3.6 mmol, 3 equiv). The cooling bath was removed and the reaction mixture was stirred overnight at rt. The solvent was co-evaporated with MeOH (3 mL). The remaining residue was then dissolved in chloroform (30 mL) and washed successively with cold 1 N aq HC1 (3 χ 20 mL), water (30 mL), and a saturated aq NaHC03 solution (30 mL). The organic layer was dried (Na2S04) and concentrated under reduced pressure. Finally, the residue was stirred with a 1 : 1 solution of DCM and heptane (15 mL). The product was isolated by filtration and dried under high vacuum to give 117d (0.2 g) as a white solid.
Step 3 : (2S,4R)-N-(5 '- Acetyl-2 '-chloro-2-fluoro- [1 , 1 '-biphenyl] -3-yl)-4-fluoropyrrolidine-2- carboxamide TFA salt (117e)
[0333] (2S,4R)-tert-Butyl 2-((5*-acetyl-2'-chloro-2-fluoro-[l,r-biphenyl]-3- yl)carbamoyl)-4-fluoropyrrolidine-l-carboxylate 117d (110 mg) was stirred in DCM (2 mL) and TFA (2 mL) for 30 min. The volatiles were removed under reduced pressure and the resulting residue 117e was used as such for the next step.
Step 4: l-(2-((2S,4R)-2-((5'-Acetyl-2'-chloro-2-fluoro-[l,l,-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-lH-indazole-3-carboxamide (117)
[0334] The title compound 117 was prepared in a manner similar to that described in Step 3 of Scheme 8. Thus, (2S,4R)-N-(5*-acetyl-2,-chloro-2-fiuoro-[l,r-biphenyl]-3-yl)-4- fluoropyrrolidine-2-carboxamide TFA salt 117e (obtained from Step 3) was coupled with 2-(3- carbamoyl-lH-indazol-l-yl)acetic acid (0.05 g) using HATU (0.104 g ) and DIEA (0.2 mL) in DMF (1.5 mL). The crude product was purified by column chromatography (0-2% MeOH in DCM) to give 117 (60 mg). 1H NMR (400 MHz, CD3OD, 300 K): (major rotamer) δ 2.65-2.75 (m, 1H), 3.91-4.04 (m, 1H), 4.20-4.29 (m, 1H), 4.84 (t, J = 8 Hz, 1H), 5.48 (d, J = 52 Hz, 1H),
5.42 (d, J = 17.2 Hz, 1H), 5.57 (d, J = 17.2 Hz, 1H), 7.12 (t, J = 6.8 Hz, 1H), 7.21-7.32 (m, 2H),
7.43 (t, J = 7.2 Hz, 1H), 7.59 (d, J = 8.4 Hz, 1H), 7.65 (d, J = 8.4 Hz, 1H), 7.89 (s, 1H), 7.93 (s, 1H), 7.96-8.02 (m, 2H), 8.22 (d, J = 8 Hz, 1H). 31F NMR (376 MHz, CD3OD, 300 K): (major rotamer) δ -128.5, -178.6 ppm. LC (method A): tR = 1.92 min. LC/MS (EI) mlz: [M + H]+ 580.
Scheme 11: Synthesis of l-(2-((2S,4R)-2-((2'-chloro-2-fluoro-[l,l'-biphenyl]-3- yl)carbamoyl)-4-fluoropyrrolidin-l-yl)-2-oxoethyl)-4-(2-methoxypyrimidin-5-yl)-lH- pyrazole-3-carboxamide (110)
Figure imgf000109_0001
Step 1: tert-Butyl 2-(4-bromo-3-carbamoyl-lH-pyrazol-l-yl)acetate (110b)
[0335] A mixture of 4-bromo-lH-pyrazole-3-carboxamidell0a (1.0 g, 5.26 mmol), tert- butyl bromoacetate (1.13 g, 0.84 mL 5.78 mmol), and potassium carbonate (798 mg, 5.78 mmol) in anhydrous acetonitrile (20 mL) was refluxed for 5 h. The reaction mixture was then cooled to rt and the solvent was removed under reduced pressure. The residue was taken in a 1 : 1 mixture of DCM and water (100 mL: 100 mL). The two layers were separated and the organic layer was washed with water (2 χ 100 mL). Finally, the organic layer was dried (Na2S04) and concentrated. The resulting residue was purified by column chromatography to give tert-butyl 2- (4-bromo-3 -carbamoyl- lH-pyrazol-l-yl)acetate 110b.
Step 2: tert-Butyl 2-(3-carbamoyl-4-(2-methoxypyrimidin-5-yl)-lH-pyrazol-l-yl)acetate (110c)
[0336] A mixture of tert-butyl 2-(4-bromo-3 -carbamoyl- lH-pyrazol-l-yl)acetate 110b (150 mg, 0.49 mmol), (2-methoxypyrimidin-5-yl)boronic acid (154 mg, 1 mmol), cesium carbonate (380 mg, 1.17 mmol), and DMF (2 mL) was purged with argon in a pressure vessel for 5 min. Tetrakis(triphenylphosphine)palladium (0) (30 mg, 0.025 mmol) was then added under argon and the pressure vessel was sealed and irradiated with microwaves at 90 °C for 30 min. The reaction mixture was cooled to rt and the solvent was removed under reduced pressure. The remaining residue was purified by column chromatography to give 110c.
Step 3: 2-(3-Carbamoyl-4-(2-methoxypyrimidin-5-yl)-lH-pyrazol-l-yl)acetic acid (llOd)
[0337] tert-Butyl 2-(3-carbamoyl-4-(2-methoxypyrimidin-5-yl)-lH-pyrazol-l-yl)acetate 110c (120 mg, 0.36 mmol) in DCM (5 mL) was treated with TFA (5 mL). After completion of the reaction (as judged by LC-MS analysis), the solvent was removed under reduced pressure. The remaining material llOd was used directly in the next step.
Step 4: l-(2-((2S,4R)-2-((2'-Chloro-2-fluoro-[l,l'-biphenyl]-3-yl)carbamoyl)-4- fluoropyrrolidin-l-yl)-2-oxoethyl)-4-(2-methoxypyrimidin-5-yl)-lH-pyrazole-3- carboxamide (110)
[0338] 2-(3-Carbamoyl-4-(2-methoxypyrimidin-5-yl)-lH-pyrazol-l-yl)acetic acid llOd (0.36 mmol) from Step 3 was dissolved in DMF (4 mL) and DIEA (1.8 mmol) was added, which was followed by the addition of (2S,4R)-N-(2'-chloro-2-fiuoro-[l,r-biphenyl]-3-yl)-4- fluoropyrrolidine-2-carboxamide hydrochloride (120 mg, 0.32 mmol) at 5 °C. HATU (287 mg, 0.76 mmol) was then added slowly at this temperature and the reaction mixture was stirred for 3 h at rt. The reaction mixture was then added to water (50 mL + 10 g solid NaCl) and extracted with DCM (2 x 25 mL). The organic layer was washed successively with an aq solution of NaHCC"3 (20 mL), water (20 mL), and brine (20 mL), then dried over Na2SC"4 and concentrated under reduced pressure. The remaining residue was purified by column chromatography (eluted with DCM/CH3OH) to give the title compound 110. 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 2.20-2.26 (m, 1H), 2.51-2.62 (m, 1H), 3.79-3.91 (m, 1H), 3.94 (s, 3H), 4.09- 4.17 (m, 1H), 4.80 (t, J = 8.0 Hz, 1H), 5.17-5.41 (m, 2H), 5.50 (d, J = 7 Hz, 1H), 7.08 (t, J = 7.2 Hz, 1H), 7.24 (t, J = 8.0 Hz, 1H), 7.30 (s, 1H), 7.39-7.47 (m, 4H), 7.58-7.61 (m, 1H), 7.96-8.13 (m, 2H), 8.74 (s, 2H), 10.05 (s, 1H); 19F NMR (376 MHz, DMSO-d6, 300 K): (major rotamer) δ - 126.84, -175.97 ppm. LC (method A): tR = 1.85 min. LC/MS (EI) m/z: [M + H]+ 596.
Scheme 12: Synthesis of (lR,3S,5R)-2-(2-(4-bromo-3-carbamoyl-lH-pyrazol-l-yl)acetyl)-N- (2 '-chloro-2-fluoro- [1,1 '-biphenyl] -3-yl)-2-azabicyclo [3.1.0] hexane-3-carboxamide (77)
Figure imgf000111_0001
Step 1: 2-(4-Bromo-3-carbamoyl-lH-pyrazol-l-yl)acetic acid (77a)
[0339] tert-Butyl 2-(4-bromo-3 -carbamoyl- lH-pyrazol-l-yl)acetate 110b (120 mg, 0.40 mmol) in DCM (5 mL) was treated with TFA (5 mL) in a similar manner to that described in Step 2 of Scheme 7. The volatiles were removed under reduced pressure and the remaining material was used directly in the next synthetic step.
Step 2 : (1R,3 S,5R)-2-(2-(4-Bromo-3-carbamoyl- lH-pyrazol- l-yl)acetyl)-N-(2 '-chloro-2- fluor 0- [1,1 '-biphenyl] -3-yl)-2-azabicyclo [3.1.0] hexane-3-carboxamide (77)
[0340] The title compound 77 (50 mg) was prepared from 2-(4-bromo-3 -carbamoyl- 1H- pyrazol-l-yl)acetic acid 77a and (lR,3S,5R)-N-(2,-chloro-2-fluoro-[l,l,-biphenyl]-3-yl)-2- azabicyclo[3.1.0]hexane-3-carboxamide hydrochloride (131 mg) in a manner similar to that described in Scheme 5. 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 0.65 (d, J = 1.6 Hz, 1H), 0.97-1.02 (m, 1H), 1.86-1.88 (m, 1H), 2.24-2.29 (m, 2H), 3.66 (t, J = 5.2 Hz, 1H), 4.52-4.56 (m, 1H), 5.19-5.47 (m, 2H), 7.09 (t, J = 7.2 Hz, 1H), 7.24 (t, J = 8.0 Hz, 1H), 7.28 (s, 1H), 7.39-7.47 (m, 4H), 7.59-7.61 (m, 1H), 7.94 (t, J = 7.2 Hz, 1H), 7.99 (s, 1H), 9.79 (s, 1H); 19F NMR (376 MHz, DMSO-d6, 300 K): (major rotamer) δ -126.64. LC (method A): tR = 1.97 min. LC/MS (EI) m/z: [M + H]+ 560.
Scheme 13: Synthesis of l-il-iilS^Ri-l-iil'-chloro-l^ S'-trifluoro-Il^'-bi henylJ-S- yl)carbamoyl)-4-fluoropyrrolidin-l-yl)-2-oxoethyl)-lH-indazole-3-carboxamide (80)
Figure imgf000112_0001
Step 1: 2-Fluoro-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)aniline (80b)
[0341] A solution of 3-bromo-2-fluoroaniline (0.5g, 2.63 mmol), 4,4,4',4',5,5,5',5'- octamethyl-2,2'-bi(l,3,2-dioxaborolane) (1.67g, 6.6 mmol), and KOAc (0.77g) in dioxane (10 mL) was degassed and refilled with argon twice. To this solution was added Pd(d f)2Cl2 (289 mg) under an atmosphere of argon. The solution was heated at 90 °C for 15 h. The reaction mixture was cooled to rt and the volatiles were removed under reduced pressure. The remaining residue was purified by column chromatography to afford 80b (803 mg).
Ill Step 2: 1'-Chloro-l^ S'-trifluoro-Il^'-biphenyll-S-amine hydrochloride (80c)
[0342] A solution of 2-fluoro-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)aniline 80b (26.0 g, 110 mmol), l-bromo-2,4,5-trifluorobenzene (12.5 g, 60 mmol), and K2CO3 (38 g, 275 mmol) in co-solvent of dioxane (250 mL) and water (63 mL) was degassed and refilled with argon twice. To this solution was added Pd(d f)2Cl2 (8.04g) under an atmosphere of argon. The solution was refluxed for 15 h. The reaction mixture was cooled to rt and the volatiles were removed under reduced pressure. The remaining residue was purified by column chromatography. The desired product fractions were collected and concentrated, and then the HC1 salt 80c was made by treatment with HCl/MeOH. 13.1 g of 80c was obtained.
Step 3: (2S,4R)-tert-Butyl l-iil'-chloro-l^ S'-trifluoro-Il^'-biphen ll-S- carbamo l)^- fluoropyrrolidine-l-carboxylate (80d)
[0343] To an ice-cold solution of (2S,4R)-l-(tert-butoxycarbonyl)-4-fluoropyrrolidine-2- carboxylic acid (9.5 g, 40.7 mmol) in DCM (200 mL) was added l-chloro-N,N,2-trimethyl-l- propenylamine (5.92 mL, 1.1 equiv) dropwise with stirring. The stirring was continued for 3 h at this temperature, and then solid 2'-chloro-2,4',5'-trifluoro-[l, -biphenyl]-3-amine hydrochloride 80c (13. lg, 44.5 mmol) was added, followed by DIEA (21.3 mL). The cooling bath was removed and the reaction mixture was stirred overnight at rt. The reaction mixture was then added to water (120 mL) and extracted with DCM (2 χ 120 mL). The organic layer was washed successively with an aq solution of NaHC03 (20 mL), water (20 mL), and brine (20 mL), and then dried over Na2S04 and concentrated under reduced pressure. The remaining residue was purified by column chromatography (e luted with hexanes/EtOAc) to give 14. lg of the desire title compound 80d.
Step 4: (2S,4R)-N-(2'-Chloro-2,4',5,-trifluoro-[l,l,-biphenyl]-3-yl)-4-fluoropyrrolidine-2- carboxamide hydrochloride (80e)
[0344] (2S,4R)-tert-Butyl 2-((2*-chloro-2,4,,5*-trifluoro-[ 1 , 1 *-biphenyl]-3-yl)carbamoyl)- 4-fluoropyrrolidine-l-carboxylate 80d (1.0 g) was taken in 4 N HC1 in dioxane (10 mL) and the resulting reaction mixture was stirred at rt for 2 h. The solvent was then removed under reduced pressure and the remaining residue 80e was used directly without further purification. Step 5: terf-Butyl 2-(3-carbamoyl-lH-indazol-l-yl)acetate (80g)
[0345] lH-Indazole-3-carboxamide 80f (56g, 347 mmol) was dissolved in CH3CN (500 mL). To this solution were added tert-butyl 2-bromoacetate (82 g, 61.5 mL) and potassium carbonate (77.4 g, 560 mmol). The mixture was heated at 90 °C for 3 h under an atmosphere of argon. The reaction mixture was cooled to rt and filtered through a pad of Celite®. The solid cake was washed with CH3CN (120 mL), and the combined filtrate was concentrated under reduced pressure. The remaining residue was purified by column chromatography to afford the title compound 80g (70 g).
Step 6: 2-(3-Carbamoyl-lH-indazol-l-yl)acetic acid (80h)
[0346] tert-Butyl 2-(3 -carbamoyl- lH-indazol-l-yl)acetate 80g (1.0 g) was taken in 4 N HC1 in dioxane (10 mL) and the resulting reaction mixture was stirred at rt for 2 h. The solvent was then removed under reduced pressure and the remaining residue 80h was used directly without further purification.
Step 7: l-il-iilS^Ri-l-iil'-Chloro-l^ S'-trifluoro-Il^'-biphenyll-S-y carbamoyl)^- fluoropyrrolidin-l-yl)-2-oxoethyl)-lH-indazole-3-carboxamide (80)
[0347] To a solution of 2-(3 -carbamoyl- lH-indazol-l-yl)acetic acid 80h (5.3 g, 24.2 mmol), (2S,4R)-N-(2'-chloro-2,4',5'-trifluoro-[ 1 , 1 '-biphenyl]-3-yl)-4-fiuoropyrrolidine-2- carboxamide hydrochloride 80e (9.0 g, 22.0 mmol) in DMF (50 mL) was added HATU (10 g) followed by dropwise addition of DIE A (18.0 mL) at rt. The mixture was stirred for 1 h at rt and the volatiles were removed under reduced pressure. The remaining residue was diluted with 10% aq sodium carbonate (50 mL) and extracted with ethyl acetate. The organic extract was washed with water and brine, then dried over MgSC^. The solution was filtered and the filtrate was evaporated under reduced pressure. The remaining residue was purified by column chromatography to afford the title compound 80 (10.0 g). 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 2.13-2.26 (m, 1H), 2.45-2.57 (m, 1H), 3.88-4.00 (m, 1H), 4.18-4.27 (m, 1H), 4.76 ( t, J = 8.4 Hz, 1H), 5.43-5.68 (m, 3H), 7.07-7.09 (m, 1H), 7.20-7.27 (m, 2H), 7.35- 7.42 (m, 2H), 7.59-7.64 (m, 2H), 7.85-7.89 (m, 1H), 7.91-7.99 (m, 1H), 8.17 (d, J = 8.4 Hz, 1H), 10.00 (s, 1H). 19F NMR (376 MHz, DMSO-d6, 300K): (major rotamer) δ -126.7, -135.8, -139.4, - 175.9. LC (method A): tR = 2.28 min. LC/MS (EI) m/z: [M + H]+ 574. EXAMPLE 7. NON-LIMITING EXAMPLES OF COMPOUNDS OF FORMULA I
[0348] Table 1 shows illustrative compounds of Formula I with characaterizing data. The assay of Example 8 was used to determine the ICso's of the compounds. Other standard factor D inhibition assays are also available. Three ***s are used to denote compounds with an IC50 less than 1 micromolar; two **s indicate compound with an IC50 between 1 micromolar and 10 micromolar, and one * denotes compounds with an IC50 greater than 10 micromolar.
TABLE 1
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
EXAMPLE 8. HUMAN FACTOR D ASSAY
[0349] Human factor D (purified from human serum, Complement Technology, Inc.) at 80 nM final concentration is incubated with test compound at various concentrations for 5 minutes at room temperature in 50 mM Tris, 1M NaCl, pH 7.5. A synthetic substrate Z-L-Lys- SBzl and DTNB (Ellman's reagent) are added to final concentrations of 100 μΜ each. The increase in color is recorded at OD405 nm in a microplate in kinetic mode over 30 minutes with 30 second time points in a spectra fluorimeter. IC50 values are calculated by non- linear regression from the percentage of inhibition of complement factor D activity as a function of test compound concentration.
EXAMPLE 9. HEMOLYSIS ASSAY
[0350] The hemolysis assay was previously described by G. Ruiz-Gomez, et al, J. Med. Chem. (2009) 52: 6042-6052. In the assay red blood cells (RBC), rabbit erythrocytes (purchased from Complement Technologies), are washed using GVB Buffer (0.1 % gelatin, 5 mM Veronal, 145 mM NaCl, 0.025 % NaN3, pH 7.3) plus 10 mM final Mg-EGTA. Cells are used at a concentration of 1 x 108 cells/mL. Prior to the hemolysis assay, the optimum concentration of Normal Human Serum (NHS) needed to achieve 100% lysis of rabbit erythrocytes is determined by titration. NHS (Complement Technologies) is incubated with inhibitor for 15 min at 37 °C, rabbit erythrocytes in buffer were added and incubated for an additional 30 min at 37 °C. Positive control (100% lysis) consists of serum and RBC and negative control (0% lysis) of Mg-EGTA buffer and RBC only. Samples are centrifuged at 2000g for 5 min, and supernatants collected. Optical density of the supernatant is monitored at 405 nm using a UV/visible spectrophotometer. Percentage lysis in each sample is calculated relative to positive control (100% lysis).
[0351] This specification has been described with reference to embodiments of the invention. However, one of ordinary skill in the art appreciates that various modifications and changes can be made without departing from the scope of the invention as set forth in the claims below. Accordingly, the specification is to be regarded in an illustrative rather than a restrictive sense, and all such modifications are intended to be included within the scope of invention.

Claims

CLAIMS What is claimed is: What is claimed is:
1. A compound of the formula
Figure imgf000151_0001
or a pharmaceutically acceptable salt thereof, wherein
Q1 is N(RJ) or QRV);
Q2 is Q2 is C(R2R2'), C(R2R2')-C(R2R2'), S, O, N(R2) or C(R2R2')0;
Q3 is N(R3), S, or C(R3R3');
(a) X1 and X2 are independently N or CH, or
(b) X1 and X2 together are C=C;
1 1 ' 2 2' 3 3'
R , R , R\ R , R\ and RJ are independently chosen at each occurence from (c) and (d)
(c) hydrogen, halogen, hydroxyl, nitro, cyano, amino, Ci-C6alkyl, C2-C6alkenyl, Ci-C6alkoxy,
C2-C6alkynyl, C2-C6alkanoyl, Ci-Cethioalkyl, hydroxyCi-C6alkyl, aminoCi-Cealkyl, -Co-C4alkylNR9R10, -C(0)OR9, -OC(0)R9, -NR9C(0)R10, -C(0)NR9R10, -OC(0)NR9R10, -O(heteroaryl), -NR9C(0)OR10, Ci-C2haloalkyl, and Ci-C2haloalkoxy, where R9 and R10 are independently chosen at each occurrence from hydrogen, Ci-C6alkyl, and (C3- Cycycloalkyl)Co-C4alkyl;
(d) -C0-C4alkyl(C3-C7cycloalkyl) and -O-C0-C4alkyl(C3-C7cycloalkyl);
Additionally any one of the following rings (e), (f), (g), (h), (i), or j) may be present:
(e) R1 and R1' or R3 and R3 may be taken together to form a 3- to 6-membered carbocyclic spiro ring or a 3- to 6-membered heterocyclic spiro ring containing 1 or 2 heteroatoms independently chosen from N, O, or S;
(f) R2 and R2 may be taken together to form a 3- to 6-membered carbocyclic spiro ring,
(g) R2 and R2'may be taken together to form a 3- to 6-membered heterocyclic spiro ring, each of which spiro rings (e), (f), and (g) is unsubstituted or substituted with one or more halogen or methyl substituents; (h) R1 and R2 may be taken together to form a 3-membered carbocyclic ring;
(i) R1 and R2 may be taken together to form a 4- to 6-membered carbocyclic ring or a 4- to 6- membered heterocyclic ring containing 1 or 2 heteroatoms independently chosen from N, O, and S;
(j) R2 and R3, if bound to adjacent carbon atoms, may be taken together to form a 3- to 6- membered carbocyclic ring or a 3- to 6-membered heterocyclic ring; each of which ring (h), (i), and (j) may be unsubstituted or substituted with 1 or more substituents independently chosen from halogen, hydroxyl, cyano, -COOH, Ci-C4alkyl, C2-C4alkenyl, Ci-C4alkoxy, C2-C4alkanoyl, hydroxyCi-C4alkyl, (mono- and di-Ci-C4alkylamino)Co- C4alkyl, -C0-C4alkyl(C3-C7cycloalkyl), -O-C0-C4alkyl(C3-C7cycloalkyl), Ci-C2haloalkyl, and Ci-C2haloalkoxy;
A is a group chosen from (k) and (1) where (k) is
Figure imgf000152_0001
Figure imgf000153_0001
X4 is B(OH) and Y is CHR9; or
X 4 is CHR9 and Y is B(OH);
R101 is hydrogen, alkyl, carboxy.
R4 is (m) or (n)
(m) -CHO, -CONH2, or C2-C6alkanoyl, including C(0)C3-C7cycloalkyl;
(n) hydrogen, -S02NH2, -C(CH2)F, -CH(CF3)NH2, Ci-C6alkyl, -C0-C4alkyl(C3-C7cycloalkyl), - -C2alkyl(C3-C7cycloalkyl),
Figure imgf000153_0002
each of which R4 other than hydrogen, -CHO, and -CONH2, is unsubstituted or substituted with one or more of amino, imino, halogen, hydroxyl, cyano, cyanoimino, Ci-C2alkyl, Ci- C2alkoxy, -Co-C2alkyl(mono- and di-Ci-C4alkylamino), Ci-C2haloalkyl, and Ci- C2haloalkoxy;
R5 and R6 are independently chosen from (o) and (p)
(o) -CHO, -C(0)NH2, -C(0)NH(CH3), or C2-C6alkanoyl; (p) hydrogen, hydroxyl, halogen, cyano, nitro, -COOH, -S02NH2, -C(NH2)Ci-C3alkyl, -C(NH2)Ci-C3haloalkyl, -CF(C=CH2), -C(=NCN)C C6alkyl, Ci-C6alkyl, C2-C6alkenyl, Ci-C6alkoxy, -Co-C4alkyl(C3-C7cycloalkyl), -C(O)C0-C4alkyl(C3-C7cycloalkyl), -P(0)(OR9)2, -OC(0)R9, -C(0)OR9, -C(0)N(CH2CH2R9)(R10), -NR9C(0)R10, phenyl, or 5- to 6-membered heteroaryl.
each R5 and R6 other than hydrogen, hydroxyl, cyano, and -COOH is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, amino, imino, cyano, cyanoimino, Ci-C2alkyl, Ci-C4alkoxy, -Co-C2alkyl(mono- and di- Ci-C4alkylamino), Ci-C2haloalkyl, Ci-C2haloalkoxy, C(0)alkyl, C(0)cycloalkyl, C(0)aryl, C(0)heterocycle, and C(0)heteroaryl.
R6 is hydrogen, halogen, hydroxyl, Ci-C4alkyl, or Ci-C4alkoxy; or
R6 and R6 may be taken together to form an oxo, vinyl, or imino group;
R7 is hydrogen, Ci-C6alkyl, or -Co-C4alkyl(C3-C7cycloalkyl);
R8 and R8' are independently chosen from hydrogen, halogen, hydroxyl, Ci-C6alkyl, Ci- C6alkoxy, and (Ci-C4alkylamino)Co-C2alkyl, or R8 and R8 are taken together to form an oxo group;
R16 is 0 or 1 or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, Ci- C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, Ci-C6alkoxy, -C0-C4alkyl(mono- and di-Ci- Cealkylamino), -Co-C4alkyl(C3-C7cycloalkyl), Ci-C2haloalkyl, and Ci-C2haloalkoxy;
R19 is hydrogen, Ci-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, -S02Ci-C6alkyl, (mono- and di-Ci- C6alkylamino)Ci-C4alkyl, -Co-C4alkyl(C3-C7cycloalkyl), each of which R19 other than hydrogen is substituted with 0 or 1 or more substituents independently chosen from halogen, hydroxyl, amino, -COOH, and -C(0)OCi-C4alkyl;
X11 is N or CR11;
X12 is N or CR12;
X13 is N or CR13;
X14 is N or CR14;
where no more than 2 of X11, X12, X13, and X14 are N;
R11, R14, and R15 are independently chosen at each occurrence from hydrogen, halogen, hydroxyl, nitro, cyano, -NR9C(0)R10, C(0)NR9R10, -0(PO)(OR9)2, -(PO)(OR9)2, Ci- C6alkyl, C2-C6alkenyl, C2-C6alkenyl(aryl), C2-C6alkenyl(cycloalkyl), C2- C6alkenyl(heterocycle), C2-C6alkenyl(heteroaryl), C2-C6alkynyl, C2-C6alkynyl(aryl), C2- C6alkynyl(cycloalkyl), C2-C6alkynyl(heterocycle), C2-C6alkynyl(heteroaryl), C2- C6alkanoyl, Ci-C6alkoxy, Ci-Cethioalkyl, -Co-C4alkyl(mono- and di-Ci-Cealkylamino), - Co-C4alkyl(C3-Cycycloalkyl), (phenyl)Co-C4alkyl, (4- to 7-membered heterocycloalkyl)Co-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N,
0, and S, and (5- or 6- membered unsaturated or aromatic heterocycle)Co-C4alkyl having
1, 2, or 3 heteroatoms independently chosen from N, O, and S), -Co-C4alkoxy(C3- Cycycloalkyl), Ci-C2haloalkyl, and Ci-C2haloalkoxy;
R12 and R13 are independently chosen from (q), (r), and (s),
(q) hydrogen, halogen, hydroxyl, nitro, cyano, amino, -COOH, Ci-C2haloalkyl, and Ci- C2haloalkoxy,
(r) Ci-Cealkyl, C2-C6alkenyl, C2-C6alkanoyl, Ci-C6alkoxy, C2-C6alkenyloxy, -C(0)OR9, Ci- Cethioalkyl, -C0-C4alkylNR9R10, -C(0)NR9R10, -S02R9, -S02NR9R10, -OC(0)R9, and -C(NR9)NR9R10 each of which (r) is unsubstituted or substituted with one or more substituents independently selected from halogen, hydroxyl, nitro, cyano, amino, - COOH,
-CONH2 Ci-C2haloalkyl, and Ci-C2haloalkoxy, and each of which (r) is also optionally substituted with one substituent chosen from phenyl and 4- to 7-membered heterocycle containing 1, 2, or 3 heteroatoms independently chosen from N, O, and S; which phenyl or 4- to 7-membered heterocycle is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, Ci-C6alkyl, C2- C6alkenyl, C2-C6alkanoyl, Ci-C6alkoxy, (mono- and di-Ci-C6alkylamino)Co-C4alkyl, Ci- C6alkylester, -Co-C4alkyl)(C3-C7cycloalkyl), Ci-C2haloalkyl, and Ci-C2haloalkoxy;
(s) -C(CH2)2R30;
R30 is -NR9C(0)R31 or R32;
R31 and R32 are Ci-C6alkyl, Ci-C6haloalkyl, (C3-C7cycloalkyl)C0-C4alkyl, (phenyl)C0-C4alkyl, (4- to 7-membered heterocycloalkyl)Co-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S, and (5- or 6- membered unsaturated or aromatic heterocycle)Co-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S;
L is either (t), (u), or (v) where (t) is a group of the formula
Figure imgf000156_0001
where R17 is hydrogen or
Ci-C6alkyl and R18 and R18 are independently chosen from hydrogen, halogen, and methyl; and m is 0, 1, 2, or 3; and
(u) is a bond,
(v) or a directly linked optionally substitiuted alkyl, alkyl(heteroaryl), heterocyclic aryl, heteroaryl, moiety including but not limited to:
Figure imgf000156_0002
B is a monocyclic or bicyclic carbocyclic or carbocyclic-oxy group or a monocyclic, bicyclic, or tricyclic heterocyclic group having 1, 2, 3, or 4 heteroatoms independently selected from N, O, and S and from 4 to 7 ring atoms per ring, or
B is a C2-C6alkenyl or C2-C6alkynyl group;
each of which B is unsubstituted or substituted with one or more substituents independently chosen from (w) and (x) and 0 or 1 substituents chosen from (y) and (z)
(w) halogen, hydroxyl, -COOH, cyano, Ci-C6alkyl, C2-C6alkanoyl, Ci-C6alkoxy, -C0- C4alkylNR9R10, -S02R9, C C2haloalkyl, and Ci-C2haloalkoxy;
(x) nitro, C2-C6alkenyl, C2-C6alkynyl, Ci-Cethioalkyl, -JC3-Cycycloalkyl, -B(OH)2, - JC(0)NR9R23, -JOS02OR21, SR9 -C(0)(CH2)i_4S(0)R21, -0(CH2)i_4S(0)NR21R22 - JOP(0)(OR21)(OR22), -JP(0)(OR21)(OR22), -JOP(0)(OR21)R22, -JP(0)(OR21)R22, - JOP(0)R21R22, -JP(0)R21R22, -JSP(0)(OR21)(OR22), -JSP(0)(OR21)(R22), JSP(0)(R21)(R22), -JNR9P(0)(NHR21)(NHR22), -JNR9P(0)(OR21)(NHR22), -
JNR9P(0)(OR21)(OR22), -JC(S)R21, -JNR21S02R22, -JNR9S(O)nNR10R22, JS02NR9COR22, -JS02NR9CONR21R22, -JNR21S02R22, -JC(0)NR21S02R22, - JC(NH2)NR22, -JC(NH2)NS(0)2R22, -JOC(0)NR21R22, -JNR21C(0)OR22, JNR21OC(0)R22, -(CH2)i_4C(0)NR21R22, -JC(0)R24R25, -JNR9C(0)R21, -JC(0)R21, -JNR9C(O)NR10R22, -CCR21, -(CH2)i_4OC(0)R21, and -JC(0)OR23;
each of which (x) may be unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, amino, oxo, -B(OH)2, - Si(CH3)3, -COOH, -CONH2, -P(0)(OH)2, Ci-C6alkyl, Ci-C6alkoxy, -C0-C2alkyl(mono- and di-C C4alkylamino), Ci-Cealkylester, Ci-C4alkylamino, Ci-C4hydroxylalkyl, Ci-C2haloalkyl, and Ci-C2haloalkoxy;
(y) naphthyl, naphthyloxy, indanyl, (4- to 7-membered heterocycloalkyl)Co-C4alkyl containing 1 or 2 heteroatoms chosen from N, O, and S, and bicyclic heterocycle containing 1 , 2, or 3 heteroatoms independently chosen from N, O, and S, and containing 4- to 7- ring atoms in each ring; each of which (y) is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, Ci-C6alkyl, C2- C6alkenyl, C2-C6alkanoyl, Ci-C6alkoxy, (mono- and di-Ci-C6alkylamino)Co-C4alkyl, Ci- C6alkylester, -C0-C4alkyl(C3-C7cycloalkyl), -S02R9, Ci-C2haloalkyl, and Ci- C2haloalkoxy; and
(z) tetrazolyl, (phenyl)Co-C2alkyl, (phenyl)Ci-C2alkoxy, phenoxy, and 5- or 6-membered heteroaryl containing 1 , 2, or 3 heteroatoms independently chosen from N, O, B, and S, each of which (z) is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, Ci-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, Ci-C6alkoxy, (mono- and di-Ci-C6alkylamino)Co-C4alkyl, Ci- C6alkylester, -Co-C4alkyl(C3-C7cycloalkyl), -S02R9, -OSi(CH3)2C(CH3)3, C C2haloalkyl, and Ci-C2haloalkoxy;
J is independently chosen at each occurrence from a covalent bond, Ci-C4alkylene, -OCi- C4alkylene, C2-C4alkenylene, and C2-C4alkynylene;
R21 and R22 are independently chosen at each occurrence from hydrogen, hydroxyl, cyano, amino, Ci-C6alkyl, Ci-C6haloalkyl, Ci-C6alkoxy, (C -C7cycloalkyl)C0-C4alkyl, (phenyl)C0-C4alkyl, -Ci-C4alkylOC(0)OCi-C6alkyl, -Ci-C4alkylOC(0)Ci-C6alkyl, -Ci- C4alkylC(0)OCi-C6alkyl, (4- to 7-membered heterocycloalkyl)Co-C4alkyl having 1 , 2, or 3 heteroatoms independently chosen from N, O, and S, and (5- or 6- membered unsaturated or aromatic heterocycle)Co-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S;
R23 is independently chosen at each occurrence from (C3-Cycycloalkyl)Co-C4alkyl, (phenyl)Co- C4alkyl, (4- to 7-membered heterocycloalkyl)Co-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S, and (5- or 6- membered unsaturated or aromatic heterocycle)C0-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S;
R24 and R25 are taken together with the nitrogen to which they are attached to form a 4- to 7- membered monocyclic heterocycloalkyl group, or a 6- to 10- membered bicyclic heterocyclic group having fused, spiro, or bridged rings;
each of which (s) may be unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, amino, oxo, -B(OH)2, - Si(CH3)3, -COOH, -CONH2, -P(0)(OH)2, Ci-C6alkyl, Ci-C6alkoxy, -C0-C2alkyl(mono- and di-Cr C4alkylamino), Ci-Cealkylester, Ci-C4alkylamino, Ci-C4hydroxylalkyl, Ci-C2haloalkyl, and Ci-C2haloalkoxy; and
wherein X2 is nitrogen or at least one of (d), (e), (g), (i), (1), (n), (p), (s), (v), (x), and (y) is present.
2. A pharmaceutically acceptable composition comprising an effective amount of a compound of claim 1 in a pharmaceutically acceptable carrier.
3. A method for the treatment of a complement D mediated disorder in a host comprising administering an effective amount of a compound of claim 1 optionally in a pharmaceutically acceptable carrier.
4. The method of claim 3, wherein the host is a human.
5. Use of a compound of claim 1 in the manufacture of a medicament for the treatment of a disorder mediated by complement Factor D.
6. A compound of claim 1 for use in the treatment of a disorder mediated by complement Factor D.
7. The use of claim 5, wherein the disorder is paroxysymal nocturnal hemoglobinuria (PNH).
8. The use of claim 5, wherein the disorder is multiple sclerosis.
9. The use of claim 5, wherein the disorder is arthritis.
10. The use of claim 5, wherein the disorder is rheumatoid arthritis.
11. The use of claim 5, wherein the disorder is a respiratory disease or a cardiovascular disease.
12. The compound of claim 6, wherein the disorder is multiple sclerosis.
13. The compound of claim 6, wherein the disorder is arthritis.
14. The compound of claim 6, wherein the disorder is rheumatoid arthritis.
15. The compound of claim 6, wherein the disorder is a respiratory disease or a cardiovascular disease.
PCT/US2015/017609 2014-02-25 2015-02-25 Compounds for treatment of complement mediated disorders WO2015130854A1 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
EA201691725A EA035501B1 (en) 2014-02-25 2015-02-25 Compounds for treatment of complement mediated disorders
CA2940777A CA2940777A1 (en) 2014-02-25 2015-02-25 Compounds for treatment of complement mediated disorders
AP2016009437A AP2016009437A0 (en) 2014-02-25 2015-02-25 Compounds for treatment of complement mediated disorders
CN201580010598.5A CN106458981B (en) 2014-02-25 2015-02-25 For treating the compound of the disease of complement-mediated
AU2015223084A AU2015223084B2 (en) 2014-02-25 2015-02-25 Compounds for treatment of complement mediated disorders
KR1020167026303A KR102366025B1 (en) 2014-02-25 2015-02-25 Compounds for treatment of complement mediated disorders
RU2016137832A RU2703995C2 (en) 2014-02-25 2015-02-25 Compounds for treating complement-mediated disorders
EP15755573.1A EP3110805B1 (en) 2014-02-25 2015-02-25 Compounds for treatment of complement mediated disorders
MX2016011038A MX2016011038A (en) 2014-02-25 2015-02-25 Compounds for treatment of complement mediated disorders.
JP2016570944A JP6537532B2 (en) 2014-02-25 2015-02-25 Compounds for the treatment of complement mediated disorders
ZA2016/05832A ZA201605832B (en) 2014-02-25 2016-08-22 Compounds for treatment of complement mediated disorders
IL247452A IL247452B (en) 2014-02-25 2016-08-24 Compounds for treatment of complement mediated disorders
US15/246,049 US20160361329A1 (en) 2014-02-25 2016-08-24 Compounds for treatment of complement mediated disorders
SA516371744A SA516371744B1 (en) 2014-02-25 2016-08-25 Compounds for Treatment of Complement Mediated Disorders

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201461944189P 2014-02-25 2014-02-25
US61/944,189 2014-02-25
US201462022916P 2014-07-10 2014-07-10
US62/022,916 2014-07-10
US201462046783P 2014-09-05 2014-09-05
US62/046,783 2014-09-05

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/246,049 Continuation US20160361329A1 (en) 2014-02-25 2016-08-24 Compounds for treatment of complement mediated disorders

Publications (1)

Publication Number Publication Date
WO2015130854A1 true WO2015130854A1 (en) 2015-09-03

Family

ID=53881572

Family Applications (8)

Application Number Title Priority Date Filing Date
PCT/US2015/017609 WO2015130854A1 (en) 2014-02-25 2015-02-25 Compounds for treatment of complement mediated disorders
PCT/US2015/017538 WO2015130795A1 (en) 2014-02-25 2015-02-25 Amide compounds for treatment of complement mediated disorders
PCT/US2015/017523 WO2015130784A1 (en) 2014-02-25 2015-02-25 Alkyne compounds for treatment of complement mediated disorders
PCT/US2015/017554 WO2015130806A1 (en) 2014-02-25 2015-02-25 Amino compounds for treatment of complement mediated disorders
PCT/US2015/017583 WO2015130830A1 (en) 2014-02-25 2015-02-25 Carbamate, ester, and ketone compounds for treatment of complement mediated disorders
PCT/US2015/017593 WO2015130838A1 (en) 2014-02-25 2015-02-25 Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
PCT/US2015/017600 WO2015130845A1 (en) 2014-02-25 2015-02-25 Phosphonate compounds for treatment of complement mediated disorders
PCT/US2015/017597 WO2015130842A2 (en) 2014-02-25 2015-02-25 Ether compounds for treatment of complement mediated disorders

Family Applications After (7)

Application Number Title Priority Date Filing Date
PCT/US2015/017538 WO2015130795A1 (en) 2014-02-25 2015-02-25 Amide compounds for treatment of complement mediated disorders
PCT/US2015/017523 WO2015130784A1 (en) 2014-02-25 2015-02-25 Alkyne compounds for treatment of complement mediated disorders
PCT/US2015/017554 WO2015130806A1 (en) 2014-02-25 2015-02-25 Amino compounds for treatment of complement mediated disorders
PCT/US2015/017583 WO2015130830A1 (en) 2014-02-25 2015-02-25 Carbamate, ester, and ketone compounds for treatment of complement mediated disorders
PCT/US2015/017593 WO2015130838A1 (en) 2014-02-25 2015-02-25 Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
PCT/US2015/017600 WO2015130845A1 (en) 2014-02-25 2015-02-25 Phosphonate compounds for treatment of complement mediated disorders
PCT/US2015/017597 WO2015130842A2 (en) 2014-02-25 2015-02-25 Ether compounds for treatment of complement mediated disorders

Country Status (26)

Country Link
US (33) US9796741B2 (en)
EP (10) EP3110417A4 (en)
JP (9) JP6400738B2 (en)
KR (7) KR20160116018A (en)
CN (8) CN106414427A (en)
AP (5) AP2016009434A0 (en)
AU (7) AU2015223075A1 (en)
BR (1) BR112016019526B1 (en)
CA (6) CA2940775A1 (en)
CY (1) CY1122550T1 (en)
DK (1) DK3110418T3 (en)
EA (5) EA201691728A1 (en)
ES (2) ES2933956T3 (en)
HR (1) HRP20200182T1 (en)
HU (1) HUE047295T2 (en)
IL (8) IL301427A (en)
LT (1) LT3110418T (en)
MX (6) MX2020004950A (en)
PL (1) PL3110418T3 (en)
PT (1) PT3110418T (en)
RS (1) RS59854B1 (en)
RU (4) RU2016137678A (en)
SA (1) SA516371744B1 (en)
SI (1) SI3110418T1 (en)
WO (8) WO2015130854A1 (en)
ZA (2) ZA201605832B (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9598446B2 (en) 2014-02-25 2017-03-21 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of complement mediated disorders
WO2017136395A1 (en) 2016-02-01 2017-08-10 Biocryst Pharmaceuticals, Inc. Benzopyrazole compounds and analogues thereof
US10000516B2 (en) 2015-08-26 2018-06-19 Achillion Pharmaceuticals, Inc. Phosphonate compounds for treatment of medical disorders
US10011612B2 (en) 2015-08-26 2018-07-03 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of medical disorders
US10092584B2 (en) 2015-08-26 2018-10-09 Achillion Pharmaceuticals, Inc. Compounds for the treatment of medical disorders
US10138225B2 (en) 2015-08-26 2018-11-27 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of medical disorders
US10385097B2 (en) 2015-08-26 2019-08-20 Achillion Pharmaceuticals, Inc. Ether compounds for treatment of medical disorders
WO2020041301A1 (en) 2018-08-20 2020-02-27 Achillion Pharmaceuticals, Inc. Pharmaceutical compounds for the treatment of complement factor d medical disorders
WO2020069024A1 (en) 2018-09-25 2020-04-02 Achillion Pharmaceuticals, Inc. Morphic forms of complement factor d inhibitors
US10660876B2 (en) 2015-08-26 2020-05-26 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of medical disorders
US10662175B2 (en) 2015-08-26 2020-05-26 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of immune and inflammatory disorders
WO2020112581A1 (en) * 2018-11-28 2020-06-04 Merck Sharp & Dohme Corp. Novel substituted piperazine amide compounds as indoleamine 2, 3-dioxygenase (ido) inhibitors
US10807952B2 (en) 2015-08-26 2020-10-20 Achillion Pharmaceuticals, Inc. Compounds for treatment of immune inflammatory disorders
US10906887B2 (en) 2015-08-26 2021-02-02 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of immune and inflammatory disorders
US10919884B2 (en) 2015-08-26 2021-02-16 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of immune and inflammatory disorders
US11001600B2 (en) 2015-08-26 2021-05-11 Achillion Pharmaceuticals, Inc. Disubstituted compounds for treatment of medical disorders
US11053253B2 (en) 2017-03-01 2021-07-06 Achillion Pharmaceuticals, Inc. Macrocyclic compounds for treatment of medical disorders
US11084800B2 (en) 2017-03-01 2021-08-10 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic pharmaceutical compounds for treatment of medical disorders
US11447465B2 (en) 2017-03-01 2022-09-20 Achillion Pharmaceuticals, Inc. Pharmaceutical compounds for treatment of medical disorders
US11814391B2 (en) 2018-09-06 2023-11-14 Achillion Pharmaceuticals, Inc. Macrocyclic compounds for the treatment of medical disorders
US11814363B2 (en) 2018-09-06 2023-11-14 Achillion Pharmaceuticals, Inc. Morphic forms of danicopan

Families Citing this family (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016138520A1 (en) 2015-02-27 2016-09-01 The Johns Hopkins University Assay to diagnose and treat disorders of the alternative pathway of complement activation
EP3365340B1 (en) 2015-10-19 2022-08-10 Incyte Corporation Heterocyclic compounds as immunomodulators
MD3377488T2 (en) 2015-11-19 2023-02-28 Incyte Corp Heterocyclic compounds as immunomodulators
US20190127366A1 (en) * 2015-12-11 2019-05-02 Lifesci Pharmaceuticals, Inc. Therapeutic inhibitory compounds
SG11201805300QA (en) 2015-12-22 2018-07-30 Incyte Corp Heterocyclic compounds as immunomodulators
AU2017210042B2 (en) 2016-01-20 2021-01-21 396419 B.C. Ltd. Compositions and methods for inhibiting Factor D
ES2906460T3 (en) 2016-05-06 2022-04-18 Incyte Corp Heterocyclic compounds as immunomodulators
US20170342060A1 (en) 2016-05-26 2017-11-30 Incyte Corporation Heterocyclic compounds as immunomodulators
CA3028685A1 (en) 2016-06-20 2017-12-28 Incyte Corporation Heterocyclic compounds as immunomodulators
US20180016260A1 (en) 2016-07-14 2018-01-18 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2018015818A2 (en) * 2016-07-15 2018-01-25 Lifesci Pharmaceuticals, Inc. Therapeutic inhibitory compounds
DE102016114392A1 (en) * 2016-08-03 2018-02-08 Eberhard Karls Universität Tübingen Medizinische Fakultät A compound for the treatment of a disease associated with a desegregation of the alternative complement pathway
ES2941716T3 (en) 2016-08-29 2023-05-25 Incyte Corp Heterocyclic compounds as immunomodulators
WO2018049214A1 (en) 2016-09-09 2018-03-15 Incyte Corporation Pyrazolopyridine derivatives as hpk1 modulators and uses thereof for the treatment of cancer
KR102507967B1 (en) 2016-09-09 2023-03-09 인사이트 코포레이션 Pyrazolopyridine derivatives as HPK1 modulators and their use to treat cancer
US20180072741A1 (en) 2016-09-09 2018-03-15 Incyte Corporation Pyrazolopyrimidine compounds and uses thereof
HRP20221216T1 (en) 2016-12-22 2022-12-23 Incyte Corporation Tetrahydro imidazo[4,5-c]pyridine derivatives as pd-l1 internalization inducers
US20180179179A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Heterocyclic compounds as immunomodulators
US20180179201A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Heterocyclic compounds as immunomodulators
ES2934230T3 (en) 2016-12-22 2023-02-20 Incyte Corp Benzooxazole derivatives as immunomodulators
WO2018136827A1 (en) 2017-01-20 2018-07-26 Vitrisa Therapeutics, Inc. Stem-loop compositions and methods for inhibiting factor d
WO2018152220A1 (en) 2017-02-15 2018-08-23 Incyte Corporation Pyrazolopyridine compounds and uses thereof
KR102602199B1 (en) * 2017-02-27 2023-11-15 리제너론 파아마슈티컬스, 인크. Humanized Models of Kidney and Liver Disorders
WO2018229543A2 (en) * 2017-06-14 2018-12-20 Lifesci Pharmaceuticals, Inc. Therapeutic inhibitory compounds
CN111163767A (en) * 2017-08-02 2020-05-15 艾其林医药公司 Treatment regimen for paroxysmal nocturnal hemoglobinuria
US10722495B2 (en) 2017-09-08 2020-07-28 Incyte Corporation Cyanoindazole compounds and uses thereof
KR101969521B1 (en) * 2017-10-26 2019-08-13 에스케이텔레콤 주식회사 Method for managing connection to machine type communication terminal and mobile terminal and base station for operating the same
US10745388B2 (en) 2018-02-20 2020-08-18 Incyte Corporation Indazole compounds and uses thereof
US10752635B2 (en) 2018-02-20 2020-08-25 Incyte Corporation Indazole compounds and uses thereof
MX2020008656A (en) 2018-02-20 2020-12-09 Incyte Corp N-(phenyl)-2-(phenyl)pyrimidine-4-carboxamide derivatives and related compounds as hpk1 inhibitors for treating cancer.
FI3774791T3 (en) 2018-03-30 2023-03-21 Incyte Corp Heterocyclic compounds as immunomodulators
TW202010742A (en) 2018-04-06 2020-03-16 美商百歐克斯製藥公司 Substituted benzofuran, benzopyrrole,benzothiophene, and structurally related complement inhibitors
US11299473B2 (en) 2018-04-13 2022-04-12 Incyte Corporation Benzimidazole and indole compounds and uses thereof
CN112752756A (en) 2018-05-11 2021-05-04 因赛特公司 Tetrahydro-imidazo [4,5-c ] pyridine derivatives as PD-L1 immunomodulators
US10899755B2 (en) 2018-08-08 2021-01-26 Incyte Corporation Benzothiazole compounds and uses thereof
JP7399968B2 (en) 2018-09-25 2023-12-18 インサイト・コーポレイション Pyrazolo[4,3-D]pyrimidine compounds as ALK2 and/or FGFR modulators
EP3873464A4 (en) * 2018-11-01 2022-06-08 Merck Sharp & Dohme Corp. Novel substituted pyrazole compounds as indoleamine 2,3-dioxygenase inhibitors
WO2020109343A1 (en) 2018-11-29 2020-06-04 F. Hoffmann-La Roche Ag Combination therapy for treatment of macular degeneration
CA3134608A1 (en) * 2019-03-22 2020-10-01 Achillion Pharmaceuticals, Inc. Pharmaceutical compounds for the treatment of complement mediated disorders
CN114450276A (en) 2019-08-06 2022-05-06 因赛特公司 Solid forms of HPK1 inhibitor
TW202115059A (en) 2019-08-09 2021-04-16 美商英塞特公司 Salts of a pd-1/pd-l1 inhibitor
AU2020357514A1 (en) 2019-09-30 2022-04-07 Incyte Corporation Pyrido[3,2-d]pyrimidine compounds as immunomodulators
WO2021096849A1 (en) 2019-11-11 2021-05-20 Incyte Corporation Salts and crystalline forms of a pd-1/pd-l1 inhibitor
CN111704617B (en) * 2020-06-15 2022-08-23 嘉兴特科罗生物科技有限公司 Small molecule compound
JP2023542949A (en) * 2020-09-23 2023-10-12 アキリオン ファーマシューティカルズ, インコーポレーテッド Pharmaceutical compounds for the treatment of complement-mediated disorders
US11780836B2 (en) 2020-11-06 2023-10-10 Incyte Corporation Process of preparing a PD-1/PD-L1 inhibitor
IL302590A (en) 2020-11-06 2023-07-01 Incyte Corp Process for making a pd-1/pd-l1 inhibitor and salts and crystalline forms thereof
US11760756B2 (en) 2020-11-06 2023-09-19 Incyte Corporation Crystalline form of a PD-1/PD-L1 inhibitor
WO2023023227A1 (en) 2021-08-20 2023-02-23 Alexion Pharmaceuticals, Inc. Methods for treating sickle cell disease or beta thalassemia using complement alternative pathway inhibitors
CA3233486A1 (en) * 2021-09-30 2023-04-06 Jun LOU Compound as inhibitor of complement factor d, and pharmaceutical composition and use thereof
WO2023158772A1 (en) 2022-02-21 2023-08-24 Teva Pharmaceuticals International Gmbh Solid state forms of danicopan and process thereof
WO2024008121A1 (en) * 2022-07-06 2024-01-11 南京明德新药研发有限公司 Difluoro-substituted azabicyclo compounds and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012093101A1 (en) * 2011-01-04 2012-07-12 Novartis Ag Indole compounds or analogues thereof useful for the treatment of age-related macular degeneration (amd)
WO2014002067A2 (en) * 2012-06-29 2014-01-03 Consiglio Nazionale Delle Ricerche Method of processing optical coherence tomography images

Family Cites Families (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9206757D0 (en) 1992-03-27 1992-05-13 Ferring Bv Novel peptide receptor ligands
US6074642A (en) 1994-05-02 2000-06-13 Alexion Pharmaceuticals, Inc. Use of antibodies specific to human complement component C5 for the treatment of glomerulonephritis
AU7808198A (en) 1997-06-03 1998-12-21 Biocryst Pharmaceuticals, Inc. Novel compounds useful in the complement, coagulat and kallikrein pathways and method for their preparation
US6410561B1 (en) 1998-03-26 2002-06-25 Japan Tobacco Inc. Amide derivatives and nociceptin antagonists
WO2000014095A1 (en) 1998-09-09 2000-03-16 Metabasis Therapeutics, Inc. Novel heteroaromatic inhibitors of fructose 1,6-bisphosphatase
WO2001017980A1 (en) 1999-09-03 2001-03-15 Ajinomoto Co., Inc. Novel processes for preparing oxazepine derivatives
US7528165B2 (en) 2001-12-13 2009-05-05 National Health Research Institutes Indole compounds
ATE424401T1 (en) 2002-07-16 2009-03-15 Amura Therapeutics Ltd PYRROL DERIVATIVES AS INHIBITORS OF CYSTEIN PROTEASES
EP1567487A4 (en) 2002-11-15 2005-11-16 Bristol Myers Squibb Co Open chain prolyl urea-related modulators of androgen receptor function
AU2003902946A0 (en) 2003-06-12 2003-06-26 Fujisawa Pharmaceutical Co., Ltd. Dpp-iv inhibitor
US7999082B2 (en) 2004-02-10 2011-08-16 National Jewish Medical And Research Center Anti-factor B antibodies
EP1727811B1 (en) 2004-03-24 2014-11-12 Shire Orphan Therapies GmbH New compounds for the inhibition of angiogenesis and use of thereof
US7417063B2 (en) 2004-04-13 2008-08-26 Bristol-Myers Squibb Company Bicyclic heterocycles useful as serine protease inhibitors
WO2006012373A2 (en) 2004-07-20 2006-02-02 Critical Therapeutics, Inc. Combination therapies of hmgb and complement inhibitors against inflammation
JP5225098B2 (en) * 2005-12-14 2013-07-03 ブリストル−マイヤーズ スクイブ カンパニー Arylpropionamide, arylacrylamide, arylpropinamide, or arylmethylurea analogs as factor XIa inhibitors
EP2108642A1 (en) 2006-10-17 2009-10-14 Kyowa Hakko Kirin Co., Ltd. Jak inhibitor
CN101583598B (en) * 2007-01-15 2012-09-12 参天制药株式会社 Novel indole derivative having inhibitory activity on i kappa b kinase
WO2009091826A2 (en) 2008-01-14 2009-07-23 The Board Of Regents Of The University Of Texas System Compositions and methods related to a human cd19-specific chimeric antigen receptor (h-car)
WO2011046204A1 (en) 2009-10-16 2011-04-21 持田製薬株式会社 Marker associated with non-alcoholic steatohepatitis
KR101851810B1 (en) 2010-04-16 2018-04-26 에이씨 이뮨 에스.에이. Novel compounds for the treatment of diseases associated with amyloid or amyloid-like proteins
JP2014518223A (en) 2011-06-20 2014-07-28 アルツハイマーズ・インスティテュート・オブ・アメリカ・インコーポレイテッド Compounds and their therapeutic uses
CN103796667A (en) 2011-06-22 2014-05-14 艾普莱斯制药公司 Methods of treating chronic disorders with complement inhibitors
NZ700875A (en) 2012-05-03 2017-03-31 Kala Pharmaceuticals Inc Pharmaceutical nanoparticles showing improved mucosal transport
US9056874B2 (en) * 2012-05-04 2015-06-16 Novartis Ag Complement pathway modulators and uses thereof
US9579360B2 (en) 2012-06-20 2017-02-28 The Trustees Of The University Of Pennsylvania Methods of treating or preventing periodontitis and diseases associated with periodontitis
ES2712190T3 (en) 2012-06-28 2019-05-09 Novartis Ag Modulators of the complement pathway and its uses
WO2014002054A1 (en) 2012-06-28 2014-01-03 Novartis Ag Pyrrolidine derivatives and their use as complement pathway modulators
EA201590118A1 (en) * 2012-06-28 2015-04-30 Новартис Аг PYRROLIDINE DERIVATIVES AND THEIR APPLICATION AS MODULATORS OF THE COMPLEMENT ACTIVATION
WO2014002051A2 (en) 2012-06-28 2014-01-03 Novartis Ag Complement pathway modulators and uses thereof
JP6154897B2 (en) 2012-06-28 2017-06-28 ノバルティス アーゲー Pyrrolidine derivatives and their use as complement pathway regulators
ES2644700T3 (en) 2012-06-28 2017-11-30 Novartis Ag Pyrrolidine derivatives and their use as modulators of the complement pathway
WO2014005150A1 (en) 2012-06-29 2014-01-03 Novartis Ag Crystalline forms of l-(2-((lr,3s,5r)-3-( (2 -fluoro-3 - (trifluoromethoxy) phenyl) carbamoyl) - 2 -azabicycl o [3.1.0] hexan- 2 -yl) - 2 -oxoethyl) - 5 -methyl - 1h - pyrazolo [3, 4 -c] pyridine - 3 - carboxami de and salts thereof
WO2014002059A1 (en) 2012-06-29 2014-01-03 Novartis Ag CRYSTALLINE FORMS OF 1-(2-((1R,3S,5R)-3-(((R)-1-(3-chloro-2-fluorophenyl)ethyl)carbamoyl)-2-azabicyclo[3.1.0]hexan-2-yl)-2-oxoethyl)-1Hpyrazolo[3,4-c]pyridine-3-carboxamide
CN104684910B (en) * 2012-07-12 2016-10-12 诺华股份有限公司 Complement is by way of regulator and application thereof
PE20150776A1 (en) 2012-09-10 2015-05-21 Hoffmann La Roche 6-AMINO ACID-HETEROARYLDIHYDROPYRIMIDINES FOR THE TREATMENT AND PROPHYLAXIS OF HEPATITIS B VIRUS INFECTION
US9773428B2 (en) 2012-12-11 2017-09-26 Fluidity Software, Inc. Computerized system and method for teaching, learning, and assessing step by step solutions to stem problems
AU2014209350B8 (en) 2013-01-23 2019-04-18 Department Of Veterans Affairs (Us) Targeting constructs based on natural antibodies and uses thereof
JP2016169161A (en) 2013-07-19 2016-09-23 大日本住友製薬株式会社 Novel imidazo pyridine compound
EA201691728A1 (en) 2014-02-25 2017-02-28 Ачиллион Фармасьютикалс, Инк. CONNECTIONS WITH ESSENTIAL GROUPS FOR THE TREATMENT OF DISTRESSED MEDIATED DISTRIBUTIONS
AR105809A1 (en) 2015-08-26 2017-11-08 Achillion Pharmaceuticals Inc COMPOUNDS FOR THE TREATMENT OF MEDICAL DISORDERS
AR105808A1 (en) 2015-08-26 2017-11-08 Achillion Pharmaceuticals Inc AMIDA COMPOUNDS FOR THE TREATMENT OF MEDICAL DISORDERS
US10000516B2 (en) 2015-08-26 2018-06-19 Achillion Pharmaceuticals, Inc. Phosphonate compounds for treatment of medical disorders
WO2017035352A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Carbamate, ester, and ketone compounds for treatment of medical disorders
WO2017035401A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of immune and inflammatory disorders
WO2017035418A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Disubstituted compounds for treatment of immune and inflammatory disorders
WO2017035405A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of immune and inflammatory disorders
EP3340983B1 (en) 2015-08-26 2023-10-04 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of immune and inflammatory disorders
WO2017035417A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Phosphonate compounds for treatment of immune and inflammatory disorders
WO2017035411A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Ether compounds for treatment of immune and inflammatory disorders
WO2017035415A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Alkyne compounds for treatment of immune and inflammatory disorders
WO2017035408A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Compounds for treatment of immune and inflammatory disorders
AR106018A1 (en) 2015-08-26 2017-12-06 Achillion Pharmaceuticals Inc ARYL, HETEROARYL AND HETEROCYCLIC COMPOUNDS FOR THE TREATMENT OF MEDICAL DISORDERS
WO2017035362A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Use of complement pathway inhibitor compounds to mitigate adoptive t-cell therapy associated adverse immune responses
WO2017035348A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Alkyne compounds for treatment of medical disorders
WO2017035361A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Disubstituted compounds for the treatment of medical disorders
WO2017035351A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of medical disorders
WO2017035413A2 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Carbamate, ester, and ketone compounds for treatment of immune and inflammatory disorders
WO2017035355A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Ether compounds for treatment of medical disorders
US20190127366A1 (en) 2015-12-11 2019-05-02 Lifesci Pharmaceuticals, Inc. Therapeutic inhibitory compounds
AU2017210042B2 (en) 2016-01-20 2021-01-21 396419 B.C. Ltd. Compositions and methods for inhibiting Factor D

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012093101A1 (en) * 2011-01-04 2012-07-12 Novartis Ag Indole compounds or analogues thereof useful for the treatment of age-related macular degeneration (amd)
WO2014002067A2 (en) * 2012-06-29 2014-01-03 Consiglio Nazionale Delle Ricerche Method of processing optical coherence tomography images

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
COLE, LB ET AL.: "Structure of 3,4-Dichloroisocoumarin-Inhibited Factor D", ACTA CRYST. D54, 1998, pages 711 - 717, XP055220052 *

Cited By (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10550140B2 (en) 2014-02-25 2020-02-04 Achillion Pharmaceuticals, Inc. Ether compounds for treatment of complement mediated disorders
US10428094B2 (en) 2014-02-25 2019-10-01 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of complement mediated disorders
US10689409B2 (en) 2014-02-25 2020-06-23 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of complement mediated disorders
US9598446B2 (en) 2014-02-25 2017-03-21 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of complement mediated disorders
US10464956B2 (en) 2014-02-25 2019-11-05 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US9732103B2 (en) 2014-02-25 2017-08-15 Achillion Pharmaceuticals, Inc. Carbamate, ester, and ketone compounds for treatment of complement mediated disorders
US9732104B2 (en) 2014-02-25 2017-08-15 Achillion Pharmaceuticals, Inc. Ether compounds for treatment of complement mediated disorders
US9796741B2 (en) 2014-02-25 2017-10-24 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US9828396B2 (en) 2014-02-25 2017-11-28 Achillion Pharmaceuticals, Inc. Alkyne compounds for treatment of complement mediated disorders
US10189869B2 (en) 2014-02-25 2019-01-29 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of complement mediated disorders
US10005802B2 (en) 2014-02-25 2018-06-26 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of complement mediated disorders
US10370394B2 (en) 2014-02-25 2019-08-06 Achillion Pharmaceuticals, Inc. Carbamate, ester, and ketone compounds for treatment of complement mediated disorders
US10081645B2 (en) 2014-02-25 2018-09-25 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US10253053B2 (en) 2014-02-25 2019-04-09 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US10100072B2 (en) 2014-02-25 2018-10-16 Achillion Pharmaceuticals, Inc. Phosphonate compounds for treatment of complement mediated disorders
US10106563B2 (en) 2014-02-25 2018-10-23 Achillion Pharmaecuticals, Inc. Ether compounds for treatment of complement mediated disorders
US9663543B2 (en) 2014-02-25 2017-05-30 Achillion Pharmaceuticals, Inc. Phosphonate compounds for treatment of complement mediated disorders
US9695205B2 (en) 2014-02-25 2017-07-04 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of complement mediated disorders
US9643986B2 (en) 2014-02-25 2017-05-09 Achillion Pharmaceuticals, Inc. Factor D inhibitors useful for treating inflammatory disorders
US10092584B2 (en) 2015-08-26 2018-10-09 Achillion Pharmaceuticals, Inc. Compounds for the treatment of medical disorders
US10385097B2 (en) 2015-08-26 2019-08-20 Achillion Pharmaceuticals, Inc. Ether compounds for treatment of medical disorders
US11649229B2 (en) 2015-08-26 2023-05-16 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of immune and inflammatory disorders
US10011612B2 (en) 2015-08-26 2018-07-03 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of medical disorders
US10287301B2 (en) 2015-08-26 2019-05-14 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of medical disorders
US11407738B2 (en) 2015-08-26 2022-08-09 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of immune and inflammatory disorders
US10000516B2 (en) 2015-08-26 2018-06-19 Achillion Pharmaceuticals, Inc. Phosphonate compounds for treatment of medical disorders
US11001600B2 (en) 2015-08-26 2021-05-11 Achillion Pharmaceuticals, Inc. Disubstituted compounds for treatment of medical disorders
US11649223B2 (en) 2015-08-26 2023-05-16 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of immune and inflammatory disorders
US10906887B2 (en) 2015-08-26 2021-02-02 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of immune and inflammatory disorders
US10822352B2 (en) 2015-08-26 2020-11-03 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of medical disorders
US10660876B2 (en) 2015-08-26 2020-05-26 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of medical disorders
US10662175B2 (en) 2015-08-26 2020-05-26 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of immune and inflammatory disorders
US10919884B2 (en) 2015-08-26 2021-02-16 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of immune and inflammatory disorders
US10138225B2 (en) 2015-08-26 2018-11-27 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of medical disorders
US10807952B2 (en) 2015-08-26 2020-10-20 Achillion Pharmaceuticals, Inc. Compounds for treatment of immune inflammatory disorders
US11926617B2 (en) 2015-08-26 2024-03-12 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of immune and inflammatory disorders
EP3925956A1 (en) * 2016-02-01 2021-12-22 BioCryst Pharmaceuticals, Inc. 1h-indazole-3-carboxamide derivatives and related compounds as factor d inhibitors for treating diseases characterized by aberrant complement system activity, such as e.g. immunological disorders
US11559515B2 (en) 2016-02-01 2023-01-24 Biocryst Pharmaceuticals, Inc. Indole compounds and analogues thereof
WO2017136395A1 (en) 2016-02-01 2017-08-10 Biocryst Pharmaceuticals, Inc. Benzopyrazole compounds and analogues thereof
CN109121414A (en) * 2016-02-01 2019-01-01 拜奥克里斯特制药公司 Benzopyrazoles compound and its analog
JP2019507130A (en) * 2016-02-01 2019-03-14 バイオクリスト ファーマスーティカルズ,インコーポレイテッドBiocryst Pharmaceuticals,Inc. Benzopyrazole compounds and their analogues
JP7257462B2 (en) 2016-02-01 2023-04-13 バイオクリスト ファーマスーティカルズ,インコーポレイテッド Benzopyrazole compounds and analogues thereof
JP2021181460A (en) * 2016-02-01 2021-11-25 バイオクリスト ファーマスーティカルズ, インコーポレイテッドBiocryst Pharmaceuticals, Inc. Benzopyrazole compounds and analogues thereof
US10849883B2 (en) 2016-02-01 2020-12-01 Biocryst Pharmaceuticals, Inc. Benzopyrazole compounds and analogues thereof
EP3411411A4 (en) * 2016-02-01 2019-08-28 Biocryst Pharmaceuticals, Inc. Benzopyrazole compounds and analogues thereof
EP3985002A1 (en) 2017-03-01 2022-04-20 Achillion Pharmaceuticals, Inc. Aryl, heteroary, and heterocyclic pharmaceutical compounds for treatment of medical disorders
US11447465B2 (en) 2017-03-01 2022-09-20 Achillion Pharmaceuticals, Inc. Pharmaceutical compounds for treatment of medical disorders
US11084800B2 (en) 2017-03-01 2021-08-10 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic pharmaceutical compounds for treatment of medical disorders
US11053253B2 (en) 2017-03-01 2021-07-06 Achillion Pharmaceuticals, Inc. Macrocyclic compounds for treatment of medical disorders
US11708351B2 (en) 2017-03-01 2023-07-25 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic pharmaceutical compounds for treatment of medical disorders
US11718626B2 (en) 2017-03-01 2023-08-08 Achillion Pharmaceuticals, Inc. Macrocyclic compounds for treatment of medical disorders
WO2020041301A1 (en) 2018-08-20 2020-02-27 Achillion Pharmaceuticals, Inc. Pharmaceutical compounds for the treatment of complement factor d medical disorders
US11814391B2 (en) 2018-09-06 2023-11-14 Achillion Pharmaceuticals, Inc. Macrocyclic compounds for the treatment of medical disorders
US11814363B2 (en) 2018-09-06 2023-11-14 Achillion Pharmaceuticals, Inc. Morphic forms of danicopan
WO2020069024A1 (en) 2018-09-25 2020-04-02 Achillion Pharmaceuticals, Inc. Morphic forms of complement factor d inhibitors
US11807627B2 (en) 2018-09-25 2023-11-07 Achillon Pharmaceuticals, Inc. Morphic forms of complement factor D inhibitors
WO2020112581A1 (en) * 2018-11-28 2020-06-04 Merck Sharp & Dohme Corp. Novel substituted piperazine amide compounds as indoleamine 2, 3-dioxygenase (ido) inhibitors

Also Published As

Publication number Publication date
DK3110418T3 (en) 2020-02-10
US9598446B2 (en) 2017-03-21
CA2940645A1 (en) 2015-09-03
JP2021193128A (en) 2021-12-23
MX2016011034A (en) 2017-03-03
US20150239893A1 (en) 2015-08-27
EP3110419A4 (en) 2017-07-26
BR112016019526B1 (en) 2023-03-28
EP3110418A1 (en) 2017-01-04
EP3623367B1 (en) 2022-09-28
JP2017506269A (en) 2017-03-02
EP3110416A4 (en) 2017-07-26
US10005802B2 (en) 2018-06-26
EP3110806A4 (en) 2017-11-22
US20160361329A1 (en) 2016-12-15
EP3110806A1 (en) 2017-01-04
US9796741B2 (en) 2017-10-24
US20170298085A1 (en) 2017-10-19
US10464956B2 (en) 2019-11-05
KR20160116018A (en) 2016-10-06
US20150239920A1 (en) 2015-08-27
EA035501B1 (en) 2020-06-25
ZA201605829B (en) 2019-01-30
EP3110423A4 (en) 2017-09-20
SI3110418T1 (en) 2020-03-31
RU2016137678A (en) 2018-04-02
US20160362399A1 (en) 2016-12-15
US10428094B2 (en) 2019-10-01
SA516371744B1 (en) 2020-11-26
CN111437278A (en) 2020-07-24
CA2940772A1 (en) 2015-09-03
WO2015130830A9 (en) 2016-06-23
AU2015223084B2 (en) 2019-11-07
RU2703995C2 (en) 2019-10-23
EP4129407A1 (en) 2023-02-08
US20190085005A1 (en) 2019-03-21
EP3110805A1 (en) 2017-01-04
LT3110418T (en) 2020-02-25
IL247450B (en) 2019-10-31
WO2015130784A1 (en) 2015-09-03
ZA201605832B (en) 2020-05-27
KR102383714B1 (en) 2022-04-07
IL285114B2 (en) 2023-08-01
WO2015130806A9 (en) 2016-08-11
JP2020121979A (en) 2020-08-13
IL247454A0 (en) 2016-11-30
US20160362398A1 (en) 2016-12-15
US20170189410A1 (en) 2017-07-06
KR20160116016A (en) 2016-10-06
US10081645B2 (en) 2018-09-25
JP2017506672A (en) 2017-03-09
US20190023729A1 (en) 2019-01-24
RU2016137832A (en) 2018-04-02
AU2015223075A1 (en) 2016-09-01
BR112016019526A2 (en) 2017-08-15
CN106456605A (en) 2017-02-22
US20150239837A1 (en) 2015-08-27
CY1122550T1 (en) 2021-01-27
US9828396B2 (en) 2017-11-28
US10100072B2 (en) 2018-10-16
US10428095B2 (en) 2019-10-01
HUE047295T2 (en) 2020-04-28
WO2015130842A3 (en) 2016-04-07
CN106413707A (en) 2017-02-15
US20180291046A1 (en) 2018-10-11
CN107073000A (en) 2017-08-18
CA2940769A1 (en) 2015-09-03
EA032690B1 (en) 2019-07-31
CN106456604A (en) 2017-02-22
US10301336B2 (en) 2019-05-28
AU2015223072A1 (en) 2016-09-01
US20150239919A1 (en) 2015-08-27
WO2015130845A9 (en) 2016-06-30
US20160362433A1 (en) 2016-12-15
JP7374967B2 (en) 2023-11-07
EA201691730A1 (en) 2017-01-30
US10087203B2 (en) 2018-10-02
US20150239921A1 (en) 2015-08-27
US20190031692A1 (en) 2019-01-31
EA201691728A1 (en) 2017-02-28
RU2707749C2 (en) 2019-11-29
CA2940775A1 (en) 2015-09-03
RU2016137677A (en) 2018-04-02
US20150239868A1 (en) 2015-08-27
MX2020004950A (en) 2021-10-26
EA201691727A1 (en) 2017-01-30
WO2015130830A1 (en) 2015-09-03
RU2016137524A (en) 2018-04-02
US9758537B2 (en) 2017-09-12
ES2771950T3 (en) 2020-07-07
US9663543B2 (en) 2017-05-30
EA201691731A1 (en) 2017-02-28
CN112250673A (en) 2021-01-22
JP2017507184A (en) 2017-03-16
US20150239894A1 (en) 2015-08-27
HRP20200182T1 (en) 2020-05-01
US10253053B2 (en) 2019-04-09
AP2016009438A0 (en) 2016-09-30
EP3110804A1 (en) 2017-01-04
EP3110423A2 (en) 2017-01-04
EP3110417A4 (en) 2017-07-26
IL269806A (en) 2019-11-28
EA201691725A1 (en) 2017-01-30
AU2020260434B2 (en) 2022-02-17
AP2016009437A0 (en) 2016-09-30
AU2015223068A1 (en) 2016-09-01
US10689409B2 (en) 2020-06-23
MX2016011037A (en) 2017-02-28
WO2015130845A1 (en) 2015-09-03
MX2016011035A (en) 2017-03-09
CN106414427A (en) 2017-02-15
US20170226142A1 (en) 2017-08-10
KR20160116017A (en) 2016-10-06
ES2933956T3 (en) 2023-02-15
KR20160116014A (en) 2016-10-06
EP3110416A1 (en) 2017-01-04
US9695205B2 (en) 2017-07-04
US9643986B2 (en) 2017-05-09
IL247450A0 (en) 2016-11-30
AU2015223132A1 (en) 2016-09-01
US20170298084A1 (en) 2017-10-19
US20200062790A1 (en) 2020-02-27
US20190185498A1 (en) 2019-06-20
EP3110805A4 (en) 2017-07-26
US10106563B2 (en) 2018-10-23
JP6688352B2 (en) 2020-04-28
US20180298043A1 (en) 2018-10-18
US20180030075A1 (en) 2018-02-01
WO2015130806A1 (en) 2015-09-03
US20160362432A1 (en) 2016-12-15
US20180022766A1 (en) 2018-01-25
RU2016137677A3 (en) 2018-10-19
JP2018199714A (en) 2018-12-20
EP3110418A4 (en) 2017-08-02
WO2015130842A9 (en) 2016-05-19
US20180291047A1 (en) 2018-10-11
EP3110417A1 (en) 2017-01-04
IL301427A (en) 2023-05-01
MX2016011038A (en) 2017-05-12
AP2016009435A0 (en) 2016-09-30
CN106458981B (en) 2019-09-10
US20150239838A1 (en) 2015-08-27
US20180022767A1 (en) 2018-01-25
JP6948426B2 (en) 2021-10-13
JP2017508789A (en) 2017-03-30
IL247452A0 (en) 2016-11-30
KR102366025B1 (en) 2022-02-21
US10189869B2 (en) 2019-01-29
US9732104B2 (en) 2017-08-15
MX2016011036A (en) 2017-02-28
KR20160118368A (en) 2016-10-11
US20180072762A1 (en) 2018-03-15
US20190048033A1 (en) 2019-02-14
IL247452B (en) 2019-09-26
IL247449A0 (en) 2016-11-30
IL285114A (en) 2021-08-31
EP3110805B1 (en) 2019-12-11
AU2015223121A1 (en) 2016-09-01
JP2017511815A (en) 2017-04-27
AU2015223068B2 (en) 2020-07-30
US10370394B2 (en) 2019-08-06
CA2940777A1 (en) 2015-09-03
EP3110418B1 (en) 2019-11-06
JP6400738B2 (en) 2018-10-03
AU2020260434A1 (en) 2020-11-26
CN106458981A (en) 2017-02-22
US20190144473A1 (en) 2019-05-16
IL269806B (en) 2021-08-31
EP3110419A1 (en) 2017-01-04
CN106456604B (en) 2020-11-06
RU2016137832A3 (en) 2018-10-29
EP3110804A4 (en) 2017-11-22
IL285114B1 (en) 2023-04-01
AP2016009436A0 (en) 2016-09-30
CA2940774A1 (en) 2015-09-03
PL3110418T3 (en) 2020-05-18
US10550140B2 (en) 2020-02-04
JP6537532B2 (en) 2019-07-03
RS59854B1 (en) 2020-02-28
AP2016009434A0 (en) 2016-09-30
US20150239895A1 (en) 2015-08-27
PT3110418T (en) 2020-01-22
KR20220047666A (en) 2022-04-18
IL247453A0 (en) 2016-11-30
KR20160119241A (en) 2016-10-12
AU2015223084A1 (en) 2016-09-01
WO2015130795A1 (en) 2015-09-03
WO2015130838A1 (en) 2015-09-03
EP3623367A1 (en) 2020-03-18
JP2017508788A (en) 2017-03-30
US9732103B2 (en) 2017-08-15
US20170260219A1 (en) 2017-09-14
WO2015130842A2 (en) 2015-09-03

Similar Documents

Publication Publication Date Title
US10428095B2 (en) Compounds for treatment of complement mediated disorders
EA043505B1 (en) ARYL, HETEROARYL AND HETEROCYCLIC COMPOUNDS FOR THE TREATMENT OF COMPLEMENT MEDIATED DISORDERS

Legal Events

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

Ref document number: 15755573

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2016570944

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 247452

Country of ref document: IL

Ref document number: MX/A/2016/011038

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2940777

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2015223084

Country of ref document: AU

Date of ref document: 20150225

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112016019532

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20167026303

Country of ref document: KR

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2015755573

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 201691725

Country of ref document: EA

Ref document number: 2015755573

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2016137832

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112016019532

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20160824