US20180291047A1 - Alkyne compounds for treatment of complement mediated disorders - Google Patents

Alkyne compounds for treatment of complement mediated disorders Download PDF

Info

Publication number
US20180291047A1
US20180291047A1 US16/006,533 US201816006533A US2018291047A1 US 20180291047 A1 US20180291047 A1 US 20180291047A1 US 201816006533 A US201816006533 A US 201816006533A US 2018291047 A1 US2018291047 A1 US 2018291047A1
Authority
US
United States
Prior art keywords
alkyl
compound
carboxamide
independently chosen
hydrogen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/006,533
Inventor
Jason Allan Wiles
Godwin Pais
Akihiro Hashimoto
Venkat Rao Gadhachanda
Qiuping Wang
Dawei Chen
Xiangzhu Wang
Atul Agarwal
Milind Deshpande
Avinash S. Phadke
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Achillion Pharmaceuticals Inc
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
Application filed by Achillion Pharmaceuticals Inc filed Critical Achillion Pharmaceuticals Inc
Priority to US16/006,533 priority Critical patent/US20180291047A1/en
Publication of US20180291047A1 publication Critical patent/US20180291047A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic 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
    • C07F9/553Heterocyclic 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
    • 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 O), which associates with factor B to form the C3(H 2 O)B complex.
  • Complement factor D acts to cleave factor B within the C3(H 2 O)B complex to form Ba and Bb. The Bb fragment remains associated with C3(H 2 O) to form the alternative pathway C3 convertase C3(H 2 O)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 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.
  • 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.
  • disorders mediated by the complement pathway include age-related macular degeneration (AMD), paroxysmal nocturnal hemoglobinuria (PNH), multiple sclerosis, and rheumatoid arthritis.
  • AMD age-related macular degeneration
  • PNH paroxysmal nocturnal hemoglobinuria
  • multiple sclerosis multiple sclerosis
  • rheumatoid arthritis Some examples include age-related macular degeneration (AMD), paroxysmal nocturnal hemoglobinuria (PNH), multiple sclerosis, and
  • Age-related macular degeneration is a leading cause of vision loss in industrialized countries.
  • ATD Age-related 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.
  • 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.
  • treatment with eculizumab requires life-long intravenous injections. Thus, there is an unmet need to develop novel inhibitors of the complement pathway.
  • 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.
  • 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.
  • Biocryst Pharmaceuticals U.S. Pat. No. 6,653,340 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.
  • 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.
  • Alexion Pharmaceuticals PCT patent publication WO1995/029697 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).
  • a compound of Formula I, or a pharmaceutically acceptable salt or composition thereof, wherein R 12 or R 13 on the A group is an alkyne (R 32 ), is a superior inhibitor of complement factor D.
  • the alkyne is directly linked to the A group.
  • the alkyne is indirectly linked to the A group through a linking moiety, defined further below.
  • a method for the treatment of a disorder associated with a dysfunction, including increased activity, of the complement pathway 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.
  • 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.
  • 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 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.
  • 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.
  • 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.
  • Q 1 is N(R 1 ) or C(R 1 R 1′ );
  • Q 2 is C(R 2 R 2′ ), C(R 2 R 2′ )—C(R 2 R 2′ ), S, O, N(R 2 ) or C(R 2 R 2′ )O;
  • Q 3 is N(R 3 ), S, or C(R 3 R 3′ );
  • X 1 and X 2 are independently N, CH, or CZ, or X 1 and X 2 together are C ⁇ C;
  • R 1 , R 1′ , R 2 , R 2′ , R 3 , and R 3′ are illustrated below (any of which can be otherwise substituted with R 1 , R 1′ , R 2 , R 2′ , R 3 , and R 3′ ) as described in more detail below.
  • R and R′ are independently chosen from H, alkyl, cycloalkyl, cycloalkylalkyl, heterocycle, heterocycloalkyl, aryl, arylalkyl, 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 mixtures 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, Cl, NH 2 , CH 3 , CH 2 D, CHD 2 , or CD 3 .
  • R 1 , R 1′ , R 2 , R 2′ , R 3 , and R 3′ are independently chosen at each occurrence, as appropriate, and only where a stable compound results, from hydrogen, halogen, hydroxyl, nitro, cyano, amino, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkoxy, C 2 -C 6 alkynyl, C 2 -C 6 alkanoyl, C 1 -C 6 thioalkyl, hydroxyC 1 -C 6 alkyl, aminoC 1 -C 6 alkyl, —C 0 -C 4 alkylNR 9 R 10 , —C(O)OR 9 , —OC(O)R 9 , —NR 9 C(O)R 10 , —C(O)NR 9 R 10 , —OC(O)NR 9 R 10 , —NR 9 C(O)OR 10
  • 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; or
  • R 2 and R 2′ may be taken together to form a 3- to 6-membered heterocyclic spiro ring; each of which spiro ring each of which ring may be unsubstituted or substituted with 1 or more substituents independently chosen from halogen (and in particular F), hydroxyl, cyano, —COOH, C 1 -C 4 alkyl (including in particular methyl), C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 4 alkoxy, C 2 -C 4 alkano
  • 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 or aryl ring or a 4- to 6-membered heterocyclic or heteroaryl ring containing 1 or 2 heteroatoms independently chosen from N, O, and S; or
  • R 2 and R 3 if bound to adjacent carbon atoms, may be taken together to form a 3- to 6-membered carbocyclic or aryl ring or a 3- to 6-membered heterocyclic or heteroaryl ring; each of which ring may be unsubstituted or substituted with 1 or more substituents independently chosen from halogen (and in particular F), hydroxyl, cyano, —COOH, C 1 -C 4 alkyl (including in particular methyl), C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 4 alkoxy, C 2
  • R 1 and R 1′ , R 2 and R 2′ , or R 3 and R 3′ can be taken together to form a carbonyl group. In alternative embodiments, 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:
  • R 4 is chosen from —CHO, —CONH 2 , C 2 -C 6 alkanoyl, hydrogen, —SO 2 NH 2 , —C(CH 2 ) 2 F, —CH(CF 3 )NH 2 , C 1 -C 6 alkyl, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl),
  • 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, C 1 -C 2 alkyl, C 1 -C 2 alkoxy, —C 0 -C 2 alkyl(mono- and di-C 1 -C 4 alkylamino), C 1 -C 2 haloalkyl, and C 1 -C 2 haloalkoxy.
  • R 5 and R 6 are independently chosen from —CHO, —C(O)NH 2 , —C(O)NH(CH 3 ), C 2 -C 6 alkanoyl, hydrogen, hydroxyl, halogen, cyano, nitro, —COOH, —SO 2 NH 2 , vinyl, C 1 -C 6 alkyl (including methyl), C 2 -C 6 alkenyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), P—C(O)C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), —P(O)(OR 9 ) 2 , —OC(O)R 9 , —C(O)OR 9 , —C(O)N(CH 2 CH 2 R 9 )(R 10 ), —NR 9 C(O)R 10 , phenyl, or 5- to 6-membered heteroaryl.
  • R 5 of 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, C 1 -C 2 alkyl, C 1 -C 4 alkoxy, —C 0 -C 2 alkyl(mono- and di-C 1 -C 4 alkylamino), C 1 -C 2 haloalkyl, and C 1 -C 2 haloalkoxy.
  • R 6′ is hydrogen, halogen, hydroxyl, C 1 -C 4 alkyl, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), or C 1 -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, C 1 -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, C 1 -C 6 alkyl, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), C 1 -C 6 alkoxy, and (C 1 -C 4 alkylamino)C 0 -C 2 alkyl; or R 8 and R 8′ are taken together to form an oxo group; or R 8 and R 8′ can be taken together with the carbon that they are bonded to form a 3-membered carbocyclic ring.
  • R 16 is absent or may include one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkyl(mono- and di-C 1 -C 6 alkylamino), —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), C 1 -C 2 haloalkyl, and C 1 -C 2 haloalkoxy.
  • R 19 is hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, —SO 2 C 1 -C 6 alkyl, (mono- and di-C 1 -C 6 alkylamino)C 1 -C 4 alkyl, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), —C 0 -C 4 alkyl(C 3 -C 7 heterocycloalkyl), —C 0 -C 4 alkyl(aryl), C 0 -C 4 alkyl(heteroaryl), and wherein R 19 other than hydrogen is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, amino, —COOH, and —C(O)OC 1 -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 .
  • No more than 2 of X 11 , X 12 , X 13 , and X 14 are N.
  • R 12 and R 13 is chosen from R 31 and the other of R 12 and R 13 is chosen from R 32 :
  • R 31 is chosen from hydrogen, halogen, hydroxyl, nitro, cyano, amino, —COOH, C 1 -C 2 haloalkyl, C 1 -C 2 haloalkoxy, C 1 -C 6 alkyl, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, C 2 -C 6 alkenyloxy, —C(O)OR 9 , C 1 -C 6 thioalkyl, —C 0 -C 4 alkylNR 9 R 10 , —C(O)NR 9 R 10 , —SO 2 R 9 , —SO 2 NR 9 R 10 , —OC(O)R 9 , and —C(NR 9 )NR 9 R 10 , each of which R 31 other than hydrogen, halogen, hydroxyl, nitro,
  • R 32 is —C 2 -C 6 alkynylR 30 , and each R 32 can be optionally substituted.
  • R 32 include, for example, but are not limited to,
  • R 12 and R 13 are each independently selected from an R 32 moiety.
  • R 11 , R 14 , and R 15 are independently chosen at each occurrence from hydrogen, halogen, hydroxyl, nitro, cyano, —O(PO)(OR 9 ) 2 , —(PO)(OR 9 ) 2 , C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, 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 -C 6 alkynyl(heteroaryl), C 2 -C 6 alkynyl(heter
  • R 21 and R 22 are independently chosen at each occurrence from hydrogen, hydroxyl, cyano, amino, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, (phenyl)C 0 -C 4 alkyl, —C 1 -C 4 alkylOC(O)OC 1 -C 6 alkyl, —C 1 -C 4 alkylOC(O)C 1 -C 6 alkyl, —C 1 -C 4 alkylC(O)OC 1 -C 6 alkyl, —C 1 -C 4 alkylC(O)OC 1 -C 6 alkyl, (4- to 7-membered heterocycloalkyl)C 0 -C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S, and (5- or 6-membered unsaturated or
  • R 23 is independently chosen at each occurrence from C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, (aryl)C 0 -C 4 alkyl, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, (phenyl)C 0 -C 4 alkyl, (4- to 7-membered heterocycloalkyl)C 0 -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)C 0 -C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S, and each R 23 can be optionally substituted.
  • R 24 and R 25 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, and each R 24 and R 25 can be optionally substituted.
  • R 30 is independently chosen at each occurrence from hydrogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, (aryl)C 0 -C 4 alkyl, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, (phenyl)C 0 -C 4 alkyl, (4- to 7-membered heterocycloalkyl)C 0 -C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; (5- or 6-membered unsaturated or aromatic heterocycle)C 0 -C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; COOH, Si(CH 3 ) 3 , COOR 30a , C 2 -C 6 alkanoyl, —B(OH) 2 , —C(O)(CH 2 ) 1-4 S(O)R 21 , —P(O)(OR 21 )(OR 22 ),
  • R 30a is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl-, (aryl)C 0 -C 4 alkyl-, (3- to 7-membered heterocycloalkyl)C 0 -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)C 0 -C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S, each of which R 30a can be optionally substituted.
  • L is a bond or is chosen from the formulas
  • R 17 is hydrogen, C 1 -C 6 alkyl, or —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl) 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 or bicyclic carbocyclic; a monocyclic or bicyclic carbocyclic-oxy group; 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; C 2 -C 6 alkenyl; C 2 -C 6 alkynyl; —(C 0 -C 4 alkyl)(aryl); —(C 0 -C 4 alkyl)(heteroaryl); or —(C 0 -C 4 alkyl)(biphenyl).
  • B is unsubstituted or substituted with one or more substituents independently chosen from R 33 and R 34 , and 0 or 1 substituents chosen from R 35 and R 36 .
  • R 33 is independently chosen from halogen, hydroxyl, —COOH, cyano, C 1 -C 6 alkyl, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkylNR 9 R 10 , —SO 2 R 9 , C 1 -C 2 haloalkyl, and C 1 -C 2 haloalkoxy.
  • R 34 is independently chosen from nitro, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 thioalkyl, -JC 3 -C 7 cycloalkyl, —B(OH) 2 , -JC(O)NR 9 R 23 , -JOSO 2 OR 21 , —C(O)(CH 2 ) 1-4 S(O)R 21 , —O(CH 2 ) 1-4 S(O)NR 21 R 22 , -JOP(O)(OR 21 )(OR 22 ), -JP(O)(OR 21 )(OR 22 ), -JOP(O)(OR 21) R 22 , -JP(O)(OR 21 )R 22 , -JOP(O)R 21 R 22 , -JP(O)R 21 R 22 , -JSP(O)(OR 21 )(OR 22 ), -JSP(O)(OR 21 )(R 22
  • R 35 is independently chosen from naphthyl, naphthyloxy, indanyl, (4- to 7-membered heterocycloalkyl)C 0 -C 4 alkyl 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 R 35 is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, (mono- and di-C 1 -C 6 alkylamino)C 0 -C 4 alkyl, C 1 -C 6 alkylester, —C 0 -C 4 alkyl(C 3 -C 7 cycloal
  • R 36 is independently chosen from tetrazolyl, (phenyl)C 0 -C 2 alkyl, (phenyl)C 1 -C 2 alkoxy, phenoxy, and 5- or 6-membered heteroaryl containing 1, 2, or 3 heteroatoms independently chosen from N, O, B, and S, each of which R 36 is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, (mono- and di-C 1 -C 6 alkylamino)C 0 -C 4 alkyl, C 1 -C 6 alkylester, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), —SO 2 R 9 , —OSi(CH 3 ) 2 C(CH 3 )
  • J is independently chosen at each occurrence from a covalent bond, C 1 -C 4 alkylene, —OC 1 -C 4 alkylene, C 2 -C 4 alkenylene, and C 2 -C 4 alkynylene.
  • compositions comprising a compound or salt of Formula I together with a pharmaceutically acceptable carrier are also disclosed.
  • complement cascade factor D diseases 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.
  • AMD age-related macular degeneration
  • 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 mediated 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;
  • 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;
  • AMD age-related macular degeneration
  • PNH paroxysymal nocturnal hemoglobinuria
  • MS multiple sclerosis
  • RA rheumatoid arthritis
  • 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 examples include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 18 F 31 P, 32 P, 35 S, 36 Cl, 125 I respectively.
  • the invention includes isotopically modified compounds of Formula I.
  • isotopically labelled compounds can be used in metabolic studies (with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), 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.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • an 18 F 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.
  • isotopes of hydrogen for example, deuterium ( 2 H) and tritium ( 3 H) may be used anywhere in described structures that achieves the desired result.
  • isotopes of carbon e.g., 13 C and 14 C, may be used.
  • the 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.
  • the deuterium can be bound to carbon in a location of bond breakage during metabolism (an ⁇ -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. 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.
  • 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 at R 4′ R 5 , R 6 , R 6′ , R 7 , R 8 , R 8′ , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 19 , R 21 , R 22 , R 23 , R 30 , and/or R 30a .
  • certain substituents on the proline ring are selectively deuterated.
  • the substitution of a hydrogen atom for a deuterium atom occurs at R, R′, R 1 , R 1′ , R 2 , R 2′ , R 3 , and/or R 3′ .
  • the alkyl residue is optionally deuterated, e.g., CD 3 or OCD 3 .
  • the unsubstituted methylene carbon is deuterated.
  • 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 3 , CH 2 CD 3 or CD 2 CD 3 .
  • the compound of the present invention may form a solvate with solvents (including water). Therefore, in one embodiment, the invention includes a solvated form of the active compound.
  • solvate refers to a molecular complex of a compound of the present invention (including salts thereof) with one or more solvent molecules. Examples of solvents are water, ethanol, dimethyl sulfoxide, acetone and other common organic solvents.
  • 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. D 2 O, d 6 -acetone, d 6 -DMSO.
  • a solvate can be in a liquid or solid form.
  • a dash (“-”) that is not between two letters or symbols is used to indicate a point of attachment for a substituent.
  • —(C ⁇ O)NH 2 is attached through carbon of the keto (C ⁇ O) group.
  • substituted 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.
  • substituent oxo (i.e., ⁇ O) then two hydrogens on the atom are replaced.
  • an oxo group replaces two hydrogens in an aromatic moiety, the corresponding partially unsaturated ring replaces the aromatic ring.
  • a pyridyl group substituted by oxo is a pyridone.
  • 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.
  • 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, Cl, Br or I); cyano; hydroxyl; nitro; azido; alkanoyl (such as a C 2 -C 6 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
  • “optionally substituted” includes one or more substituents independently chosen from halogen, hydroxyl, amino, cyano, —CHO, —COOH, —CONH 2 , C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, —C 1 -C 6 alkoxy, C 2 -C 6 alkanoyl, C 1 -C 6 alkylester, (mono- and di-C 1 -C 6 alkylamino)C 0 -C 2 alkyl, C 1 -C 2 haloalkyl, hydoxyC 1 -C 6 alkyl, ester, carbamate, urea, sulfonamide, —C 1 -C 6 alkyl(heterocyclo), C 1 -C 6 alkyl(heteroaryl), —C 1 -C 6 alkyl
  • 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 C 1 -C 2 , C 1 -C 3 , or C 1 -C 6 .
  • the specified ranges as used herein indicate an alkyl group having each member of the range described as an independent species.
  • C 1 -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.
  • C 1 -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 is used herein in conjunction with another group, for example, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, or —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), the indicated group, in this case cycloalkyl, is either directly bound by a single covalent bond (C 0 alkyl), 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 —O—C 0 -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 -C 8 alkenyl, 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. Examples of 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 -C 8 alkynyl or C 2 -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 C 1 -C 4 alkylene, C 1 -C 3 alkylene, or C 1 -C 2 alkylene.
  • 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 C 2 -C 4 alkenylene.
  • 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.
  • Alkoxy is an alkyl group as defined above covalently bound through an oxygen bridge (—O—).
  • 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.
  • 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.
  • Alkenyloxy is an alkenyl group as defined covalently bound to the group it substitutes by an oxygen bridge (—O—).
  • Alkanoyl is an alkyl group as defined above covalently bound through a carbonyl (C ⁇ O) bridge.
  • the carbonyl carbon is included in the number of carbons, that is C 2 alkanoyl is a CH 3 (C ⁇ O)— group.
  • the alkanoyl group is optionally substituted as described above.
  • 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 —O(C ⁇ O)alkyl or a group of the formula —(C ⁇ O)Oalkyl.
  • “Amide” or “carboxamide” is —C(O)NR a R b wherein R a and R b are each independently selected from hydrogen, alkyl, for example, C 1 -C 6 alkyl, alkenyl, for example, C 2 -C 6 alkenyl, alkynyl, for example, C 2 -C 6 alkynyl, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), —C 0 -C 4 alkyl(C 3 -C 7 heterocycloalkyl), —C 0 -C 4 alkyl(aryl), and —C 0 -C 4 alkyl(heteroaryl); or together with the nitrogen to which they are bonded, R a and R b can form a C 3 -C 7 heterocyclic ring.
  • the R a and R b groups are each independently optionally substituted as described above.
  • Carbocyclic group 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.
  • 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.
  • carbocyclic rings examples include cyclohexenyl, cyclohexyl, cyclopentenyl, cyclopentyl, cyclobutenyl, cyclobutyl and cyclopropyl rings.
  • the carbocyclic ring is optionally substituted as described above.
  • the cycloalkyl is a partially unsaturated (i.e., not aromatic) group containing all carbon ring atoms.
  • the cycloalkyl is a saturated group containing all carbon ring atoms.
  • 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, —O—, linker.
  • 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.
  • Haloalkoxy indicates a haloalkyl group as defined herein attached through an oxygen bridge (oxygen of an alcohol radical).
  • Hydroalkyl is an alkyl group as previously described, substituted with at least one hydroxyl substituent.
  • aminoalkyl is an alkyl group as previously described, substituted with at least one amino substituent.
  • 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. 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.
  • 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.
  • 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.
  • 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, di
  • 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 ( ⁇ O) moieties are pyrimidinonyl and 1,1-dioxo-thiomorpholinyl.
  • the heterocycle groups herein are optionally substituted independently with one or more substituents described herein.
  • Heterocyclicoxy group is a monocyclic heterocyclic ring or a bicyclic heterocyclic group as described previously linked to the group it substitutes via an oxygen, —O—, linker.
  • 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.
  • the only heteroatom is nitrogen.
  • the only heteroatom is oxygen.
  • the only heteroatom is sulfur.
  • Monocyclic heteroaryl groups typically have from 5 to 7 ring atoms.
  • 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.
  • the total number of S and O atoms in the heteroaryl group exceeds 1, these heteroatoms are not adjacent to one another.
  • the total number of S and O atoms in the heteroaryl group is not more than 2.
  • the total number of S and O atoms in the aromatic heterocycle is not more than 1.
  • 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, puriny
  • 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 heteroatom. 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.
  • 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.
  • mono- and di-alkylamino groups include ethylamino, dimethylamino, and methyl-propyl-amino.
  • a “dosage form” means a unit of administration of an active agent.
  • 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.
  • “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, non-toxic, 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.
  • salts of the present compounds further include solvates of the compounds and of the compound salts.
  • 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.
  • conventional non-toxic 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—(CH 2 ) 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
  • carrier applied to pharmaceutical compositions/combinations of the invention refers to a diluent, excipient, or vehicle with which an active compound is provided.
  • 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.
  • 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.
  • a “prodrug” as used herein means a compound which when administered to a host in vivo is converted into a parent drug.
  • 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.
  • “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.
  • 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.
  • 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.
  • 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.
  • Formula I can be considered to have a central core, an L-B substituent, and a (C ⁇ O)A substituent. It has been surprisingly discovered that a compound of Formula I, or a pharmaceutically acceptable salt or composition thereof, wherein R 12 or R 13 on the A group is an alkyne (R 32 ), 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.
  • Non-limiting examples of compounds falling within Formula I with variations in the variables e.g., A, B, R 1 -R 3′ , and L, are illustrated below. The disclosure includes all combinations of these definitions so long as a stable compound results.
  • 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 XXXXX 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.
  • 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.
  • a compound of Formula I, a pharmaceutically acceptable salt or composition thereof, wherein R 12 or R 13 on the A group is an alkyne is a superior inhibitor of complement factor D.
  • the alkyne is directly linked to the A group.
  • the alkyne is indirectly linked to the A group through a linking moiety, wherein the linking moiety is C 1 -C 4 alkylene.
  • R 12 and R 13 is chosen from R 31 and the other of R 12 and R 13 is chosen from R 32 . In another embodiment each of R 12 and R 13 can be independently selected from R 32 .
  • R 31 is chosen from hydrogen, halogen, hydroxyl, nitro, cyano, amino, —COOH, C 1 -C 2 haloalkyl, C 1 -C 2 haloalkoxy, C 1 -C 6 alkyl, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, C 2 -C 6 alkenyloxy, —C(O)OR 9 , C 1 -C 6 thioalkyl, —C 0 -C 4 alkylNR 9 R 10 , —C(O)NR 9 R 10 , —SO 2 R 9 , —SO 2 NR 9 R 10 , —OC(O)R 9 , and —C(NR 9 )NR 9 R 10 , each of which R 31 other than hydrogen, halogen, hydroxyl, nitro,
  • R 32 is —C 2 -C 6 alkynylR 30 , and each R 32 can be optionally substituted.
  • R 32 include, for example, but are not limited to,
  • R 32 is selected from:
  • R 12 is R 32 .
  • R 13 is R 32 .
  • R 12 is R 32 , which is C 2 -C 4 alkynylR 30 .
  • R 13 is R 32 , which is C 2 -C 4 alkynylR 30 .
  • the disclosure provides compounds of Formula I, wherein;
  • R 12 and R 13 is H and the other of R 12 and R 13 is R 32 , where
  • R 32 is C 2 -C 6 alkynylR 30 (which can be, for example, C 2 -C 3 alkynylR 30 , C 2 -C 4 alkynylR 30 , C 2 -C 5 alkynylR 30 , or C 2 -C 6 alkynylR 30 ), wherein R 30 is independently chosen at each occurrence from hydrogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, (aryl)C 0 -C 4 alkyl, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, (phenyl)C 0 -C 4 alkyl, (4- to 7-membered heterocycloalkyl)C 0 -C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; (5- or 6-membered unsaturated or aromatic heterocycle)C 0 -C 4 alkyl having 1, 2, or 3 heteroatoms
  • R 9 , R 10 , R 21 , R 22 , R 24 , R 25 , and R 30a are as defined in the summary section above.
  • the disclosure provides compounds of Formula I, wherein;
  • R 1 , R 1′ , R 2 , and R 3′ are all hydrogen
  • R 2 is fluoro and R 3 is hydrogen, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), or —O—C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl);
  • R 5 is hydrogen, halogen, or C 1 -C 2 alkyl
  • R 11 , R 13 , R 14 , and R 15 if present, are independently chosen at each occurrence from hydrogen, halogen, hydroxyl, amino, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, —C 0 -C 2 alkyl(mono- and di-C 1 -C 2 alkylamino), trifluoromethyl, and trifluoromethoxy;
  • X 12 is CR 12 ;
  • R 12 is C 2 -C 6 alkynylR 30 .
  • the disclosure provides compounds of Formula I, wherein;
  • n 0 or 1
  • R 2 is halogen
  • R 2′ is hydrogen or halogen
  • R 3 is hydrogen, halogen, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), or —O—C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl);
  • R 6 is —C(O)C 1 -C 4 alkyl, —C(O)NH 2 , —C(O)CF 3 , —C(O)(C 3 -C 7 cycloalkyl), or -ethyl(cyanoimino);
  • R 12 and R 13 is selected from hydrogen, halogen, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, trifluoromethyl, and trifluoromethoxy; the other of R 12 and R 13 is R 32 , where
  • R 32 is C 2 -C 6 alkynylR 30 , wherein R 30 is independently chosen at each occurrence from hydrogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, (aryl)C 0 -C 4 alkyl, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, (phenyl)C 0 -C 4 alkyl, (4- to 7-membered heterocycloalkyl)C 0 -C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; (5- or 6-membered unsaturated or aromatic heterocycle)C 0 -C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; COOH, Si(CH 3 ) 3 , COOR 30a , C 2 -C 6 alkanoyl, —B(OH) 2 , —C(O)(CH 2 ) 1-4 S(O)R
  • R 9 , R 10 , R 21 , R 22 , R 24 , R 25 , and R 30a are as defined in the summary section above.
  • the disclosure provides compounds of Formula I, wherein;
  • R 12 and R 13 is hydrogen, hydroxyl, halogen, methyl, or methoxy; and the other of R 12 and R 13 is R 32 , where
  • R 32 is C 2 -C 6 alkynylR 30 , wherein R 30 is independently chosen at each occurrence from hydrogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, (aryl)C 0 -C 4 alkyl, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, (phenyl)C 0 -C 4 alkyl, (4- to 7-membered heterocycloalkyl)C 0 -C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; (5- or 6-membered unsaturated or aromatic heterocycle)C 0 -C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; COOH, Si(CH 3 ) 3 , COOR 30a , C 2 -C 6 alkanoyl, —B(OH) 2 , —C(O)(CH 2 ) 1-4 S(O)R
  • R 9 , R 10 , R 21 , R 22 , R 24 , R 25 , and R 30a are as defined in the summary section above.
  • R 32 may be unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, amino, oxo, —B(OH) 2 , —Si(CH 3 ) 3 , —COOH, —CONH 2 , —P(O)(OH) 2 , C 1 -C 6 alkyl, C 1 -C 6 alkoxy, —C 0 -C 2 alkyl(mono- and di-C 1 -C 4 alkylamino), C 1 -C 6 alkylester, C 1 -C 4 alkylamino, C 1 -C 4 hydroxylalkyl, C 1 -C 2 haloalkyl, and C 1 -C 2 haloalkoxy.
  • substituents independently chosen from halogen, hydroxyl, nitro, cyano, amino, oxo, —B(OH) 2 , —Si(CH 3 ) 3
  • Q 1 is N(R 1 ) or C(R 1 R 1′ );
  • Q 2 is C(R 2 R 2′ ), C(R 2 R 2′ )—C(R 2 R 2′ ), S, O, N(R 2 ) or C(R 2 R 2′ )O;
  • Q 3 is N(R 3 ), S, or C(R 3 R 3′ );
  • X 1 and X 2 are independently N or CH, or X 1 and X 2 together are C ⁇ C;
  • R 1 , R 1′ , R 2 , R 2′ , R 3 , and R 3′ are illustrated below (any of which can be otherwise substituted with R 1 , R 1′ , R 2 , R 2′ , R 3 , and R 3′ ) as described in more detail below.
  • R and R′ are independently chosen from H, alkyl, cycloalkyl, 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 mixtures 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, Cl, NH 2 , CH 3 , CH 2 D, CHD 2 , or CD 3 .
  • R 1 , R 1′ , R 2 , R 2′ , R 3 , and R 3′ are independently chosen at each occurrence, as appropriate, and only where a stable compound results, from hydrogen, halogen, hydroxyl, nitro, cyano, amino, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkoxy, C 2 -C 6 alkynyl, C 2 -C 6 alkanoyl, C 1 -C 6 thioalkyl, hydroxyC 1 -C 6 alkyl, aminoC 1 -C 6 alkyl, —C 0 -C 4 alkylNR 9 R 10 , —C(O)OR 9 , —OC(O)R 9 , —NR 9 C(O)R 10 , —C(O)NR 9 R 10 , —OC(O)NR 9 R 10 , —NR 9 C(O)OR 10
  • 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; or
  • R 2 and R 2′ may be taken together to form a 3- to 6-membered heterocyclic spiro ring each of which ring may be unsubstituted or substituted with 1 or more substituents independently chosen from halogen (and in particular F), hydroxyl, cyano, —COOH, C 1 -C 4 alkyl (including in particular methyl), C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 4 alkoxy, C 2 -C 4 alkanoyl, hydroxyC 1 -C
  • 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 or aryl ring or a 4- to 6-membered heterocyclic or heteroaryl ring containing 1 or 2 heteroatoms independently chosen from N, O, and S; or R 2 and R 3 , if bound to adjacent carbon atoms, may be taken together to form a 3- to 6-membered carbocyclic or aryl ring or a 3- to 6-membered heterocyclic or heteroaryl ring each of which ring may be unsubstituted or substituted with one or more substituents independently chosen from halogen (and in particular F), hydroxyl, cyano, —COOH, C 1 -C 4 alkyl (including in particular methyl), C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 4 alkoxy, C 2 -
  • the central core moiety is proline.
  • 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.
  • R 1 , R 1′ , R 2′ , and R 3′ if present, are all hydrogen; and R 2 is fluoro and R 3 is —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl) or —O—C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl).
  • R 1 and R 2 are taken together to form a 3- to 6-membered cycloalkyl group, and R 1′ , R 2′ , R 3 , and R 3′ , where present, are all hydrogen.
  • R 1 , R 1′ , R 3 , and R 3′ if present, are all hydrogen, and R 2 and R 2′ are taken together to form a 5- or 6-membered heterocycloalkyl group having 1 or 2 oxygen atoms.
  • R 1 is hydrogen and R 2 is fluoro.
  • R 1 and R 2 are joined to form a 3 membered ring.
  • the disclosure includes compounds of Formula I in which the central pyrrolidine is vinyl substituted, for example:
  • 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:
  • 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:
  • L is a bond or is chosen from the formulas:
  • R 17 is hydrogen, C 1 -C 6 alkyl, or —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl) 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 or bicyclic carbocyclic; a monocyclic or bicyclic carbocyclic-oxy group; 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; C 2 -C 6 alkenyl; C 2 -C 6 alkynyl; —(C 0 -C 4 alkyl)(aryl); —(C 0 -C 4 alkyl)(heteroaryl); or —(C 0 -C 4 alkyl)(biphenyl).
  • R 33 is independently chosen from halogen, hydroxyl, —COOH, cyano, C 1 -C 6 alkyl, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkylNR 9 R 10 , —SO 2 R 9 , C 1 -C 2 haloalkyl, and C 1 -C 2 haloalkoxy;
  • R 34 is independently chosen from nitro, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 thioalkyl, -JC 3 -C 7 cycloalkyl, —B(OH) 2 , -JC(O)NR 9 R 23 , -JOSO 2 OR 21 , —C(O)(CH 2 ) 1-4 S(O)R 21 , —O(CH 2 ) 1-4 S(O)NR 21 R 22 , -JOP(O)(OR 21 )(OR 22 ), -JP(O)(OR 21 )(OR 22 ), -JOP(O)(OR 21 )R 22 , -JP(O)(OR 21 )R 22 , -JOP(O)R 21 R 22 , -JP(O)R 21 R 22 , -JSP(O)(OR 21)(O R 22 ), -JSP(O)(OR 21 )(R
  • R 35 is independently chosen from naphthyl, naphthyloxy, indanyl, (4- to 7-membered heterocycloalkyl)C 0 -C 4 alkyl 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 R 35 is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, (mono- and di-C 1 -C 6 alkylamino)C 0 -C 4 alkyl, C 1 -C 6 alkylester, —C 0 -C 4 alkyl(C 3 -C 7 cycloal
  • R 36 is independently chosen from tetrazolyl, (phenyl)C 0 -C 2 alkyl, (phenyl)C 1 -C 2 alkoxy, phenoxy, and 5- or 6-membered heteroaryl containing 1, 2, or 3 heteroatoms independently chosen from N, O, B, and S, each of which R 36 is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, (mono- and di-C 1 -C 6 alkylamino)C 0 -C 4 alkyl, C 1 -C 6 alkylester, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), —SO 2 R 9 , —OSi(CH 3 ) 2 C(CH 3 )
  • J is independently chosen at each occurrence from a covalent bond, C 1 -C 4 alkylene, —OC 1 -C 4 alkylene, C 2 -C 4 alkenylene, and C 2 -C 4 alkynylene.
  • -L-B— is
  • R 26 and R 27 are independently chosen from hydrogen, halogen, hydroxyl, nitro, cyano, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, C 1 -C 6 thioalkyl, —C 0 -C 4 alkyl(mono- and di-C 1 -C 6 alkylamino), —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), —C 0 -C 4 alkoxy(C 3 -C 7 cycloalkyl), C 1 -C 2 haloalkyl, C 1 -C 2 haloalkoxy, and C 1 -C 2 haloalkylthio.
  • R 18 and R 18′ are independently chosen from hydrogen, halogen, hydroxymethyl, and methyl; and m is 0 or 1; and
  • R 26 , R 27 , and R 28 are independently chosen from hydrogen, halogen, hydroxyl, nitro, cyano, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, C 1 -C 6 thioalkyl, (mono- and di-C 1 -C 6 alkylamino)C 0 -C 4 alkyl, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, (aryl)C 0 -C 4 alkyl-, (heteroaryl)C 0 -C 4 alkyl-, and —C 0 -C 4 alkoxy(C 3 -C 7 cycloalkyl); each of which R 26 , R 27 , and R 28 other than hydrogen, halogen, hydroxyl, nitro, cyano, is unsubstituted or substituted with one or more substituent
  • R 29 is hydrogen, C 1 -C 2 alkyl, C 1 C 2 haloalkyl or —Si(CH 3 ) 2 C(CH 3 ) 3 .
  • m is 0.
  • 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, 1-(2,2,2-trifluoroethyl)-1H-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, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, C 1 -C 6 thioalkyl, (mono- and di-C 1 -C 6 alkylamino)C 0 -C 4 alkyl, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, —C 0 -C 4 alkoxy(C 3 -C 7 cycloalkyl), (phenyl)C 0 -C 2 alkyl, (pyridyl)C 0 -C 2 alkyl; each of which substituents other than hydrogen, halogen, hydroxyl, nitro,
  • B is phenyl or pyridyl substituted with 1, 2, or 3 substituents chosen from chloro, bromo, hydroxyl, —SCF 3 , C 1 -C 2 alkyl, C 1 -C 2 alkoxy, trifluoromethyl, phenyl and trifluoromethoxy each of which substituents other than chloro, bromo, hydroxyl, —SCF 3 , can be optionally substituted.
  • B is a 2-fluoro-3-chlorophenyl or a 2-fluoro-3-trifluoromethoxyphenyl group.
  • B is pyridyl, optionally substituted with halogen, C 1 -C 2 alkoxy, and trifluoromethyl.
  • B is phenyl, substituted with 1, 2, or 3 substituents independently selected from halogen, C 1 -C 2 alkyl, C 1 -C 2 alkoxy, trifluoromethyl, and optionally substituted phenyl.
  • R 23 is independently chosen at each occurrence from (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, (phenyl)C 0 -C 4 alkyl, (4- to 7-membered heterocycloalkyl)C 0 -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)C 0 -C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S.
  • B is selected from
  • R 27 is hydrogen, methyl, or trifluoromethyl
  • R 28 is hydrogen or halogen
  • R 29 is hydrogen, methyl, trifluoromethyl, or —Si(CH 3 ) 2 C(CH 3 ) 3 .
  • A is a group chosen from:
  • R 4 is chosen from —CHO, —CONH 2 , C 2 -C 6 alkanoyl, hydrogen, —SO 2 NH 2 , —C(CH 2 ) 2 F, —CH(CF 3 )NH 2 , C 1 -C 6 alkyl, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), —C(O)C 0 -C 2 alkyl(C 3 -C 7 cycloalkyl),
  • 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, C 1 -C 2 alkyl, C 1 -C 2 alkoxy, —C 0 -C 2 alkyl(mono- and di-C 1 -C 4 alkylamino), C 1 -C 2 haloalkyl, and C 1 -C 2 haloalkoxy.
  • R 5 and R 6 are independently chosen from —CHO, —C(O)NH 2 , —C(O)NH(CH 3 ), C 2 -C 6 alkanoyl, hydrogen, hydroxyl, halogen, cyano, nitro, —COOH, —SO 2 NH 2 , vinyl, C 1 -C 6 alkyl (including methyl), C 2 -C 6 alkenyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), —C(O)C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), —P(O)(OR 9 ) 2 , —OC(O)R 9 , —C(O)OR 9 , —C(O)N(CH 2 CH 2 R 9 )(R 10 ), —NR 9 C(O)R 10 , phenyl, or 5- to 6-membered heteroaryl.
  • 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, —COOH, may be substituted with one or more substituents independently chosen from halogen, hydroxyl, amino, imino, cyano, cyanoimino, C 1 -C 2 alkyl, C 1 -C 4 alkoxy, —C 0 -C 2 alkyl(mono- and di-C 1 -C 4 alkylamino), C 1 -C 2 haloalkyl, and C 1 -C 2 haloalkoxy.
  • R 6′ is hydrogen, halogen, hydroxyl, C 1 -C 4 alkyl, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), or C 1 -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, C 1 -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, C 1 -C 6 alkyl, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), C 1 -C 6 alkoxy, and (C 1 -C 4 alkylamino)C 0 -C 2 alkyl; or R 8 and R 8′ are taken together to form an oxo group; or R 8 and R 8′ can be taken together with the carbon that they are bonded to form a 3-membered carbocyclic ring.
  • R 16 is absent or may include one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkyl(mono- and di-C 1 -C 6 alkylamino), —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), C 1 -C 2 haloalkyl, and C 1 -C 2 haloalkoxy.
  • R 19 is hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, —SO 2 C 1 -C 6 alkyl, (mono- and di-C 1 -C 6 alkylamino)C 1 -C 4 alkyl, —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), —C 0 -C 4 alkyl(C 3 -C 7 heterocycloalkyl), —C 0 -C 4 alkyl(aryl), C 0 -C 4 alkyl(heteroaryl), and wherein R 19 other than hydrogen is unsubstituted or substituted with 1 or more substituents independently chosen from halogen, hydroxyl, amino, —COOH, and —C(O)OC 1 -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 .
  • No more than 2 of X 11 , X 12 , X 13 , and X 14 are N.
  • R 11 , R 14 , and R 15 are independently chosen at each occurrence from hydrogen, halogen, hydroxyl, nitro, cyano, —O(PO)(OR 9 ) 2 , —(PO)(OR 9 ) 2 , C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, 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 -C 6 alkynyl(heteroaryl), C 2 -C 6 alkynyl(heter
  • R 5 and R 6 are independently chosen from —CHO, —C(O)NH 2 , —C(O)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, C 1 -C 2 alkyl, C 1 -C 4 alkoxy, —C 0 -C 2 alkyl(mono- and di-C 1 -C 4 alkylamino), C 1 -C 2 haloalkyl, and C 1 -C 2 haloalkoxy.
  • R 8 and R 8′ are independently hydrogen or methyl.
  • R 8 and R 8′ are hydrogen.
  • R 7 is hydrogen or methyl.
  • R 7 is hydrogen
  • 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 ID.
  • the variables may include any of the definitions set forth herein that results in a stable compound. In certain embodiments, the following conditions apply for Formula IB and IC.
  • the disclosure includes compounds and salts of Formula VII:
  • R 1 , R 2 , R 2′ , and R 3 are independently chosen from hydrogen, halogen, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, —C 0 -C 2 alkylNR 9 R 10 , —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), —O—C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), C 1 -C 2 haloalkyl, and C 1 -C 2 haloalkoxy;
  • R 8 and R 8′ are independently chosen from hydrogen, halogen, and methyl
  • R 5 is hydrogen, hydroxyl, cyano, —COOH, C 1 -C 6 alkyl, C 1 -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, C 1 -C 2 haloalkyl, or C 1 -C 2 haloalkoxy;
  • R 6 is —C(O)CH 3 , —C(O)NH 2 , —C(O)CF 3 , —C(O)(cyclopropyl), or -ethyl(cyanoimino);
  • R 11 and R 14 are independently chosen from hydrogen, halogen, hydroxyl, amino, nitro, cyano, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, C 1 -C 6 thioalkyl, —C 0 -C 4 alkyl(mono- and di-C 1 -C 6 alkylamino), —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), —OC 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), C 1 -C 2 haloalkyl, and C 1 -C 2 haloalkoxy.
  • Prodrugs of Formula I are also within the scope of the disclosure.
  • 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 anti-inflammatory 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 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.
  • 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:
  • Protease inhibitors plasma-derived C1-INH concentrates, for example Cetor® (Sanquin), Berinert-P® (CSL Behring, Lev Pharma), and Cinryze®; and recombinant human C1-inhibitors, for example Rhucin®;
  • Soluble complement regulators Soluble complement receptor 1 (TP10) (Avant Immunotherapeutics); sCR1-sLex/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 Compstatin/POT-4 (Potentia Pharmaceuticals); ARC1905 (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.
  • therapeutic agents examples 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, itraconazole,
  • 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
  • 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.
  • 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 Celec
  • 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-1a), Betaseron (interferon beta-1b), Copaxone (glatiramer acetate), Extavia (interferon beta-1b), Gilenya (fingolimod), Lemtrada (alemtuzumab), Novantrone (mitoxantrone), Plegridy (peginterferon beta-1a), Rebif (interferon beta-1a), Tecfidera (dimethyl fumarate), Tysabri (natalizumab), Solu-Medrol (methylpredni solone), High-dose oral Deltasone (prednisone), H.P. Acthar Gel (ACTH), and combinations thereof.
  • Aubagio teriflunomide
  • Avonex interferon beta-1a
  • Betaseron interferon beta-1b
  • Copaxone glatiramer a
  • 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 S1P 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-CD 25 , 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,
  • anti-inflammatory agents examples 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, diclofe
  • 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 compound of the present invention can be prepared, for example, from a central core.
  • the central core is coupled to an amine to generate an amide of Structure 2 (wherein L-B includes a C(O)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.
  • central core Structure 5 is reacted with a heterocyclic or heteroaryl compound to generate a compound of Structure 6.
  • Structure 6 is deprotected to generate a carboxylic acid, Structure 7.
  • Structure 7 is coupled to an amine to generate a compound of Formula I. This chemistry is illustrated in Route 2.
  • 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.
  • 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 X 11 , X 12 , X 13 and X 14 positions to generate compounds of Formula I. This chemistry is illustrated in Route 4.
  • 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.
  • 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 chemistry is illustrated in Route 6.
  • Z a is halogen
  • deuterated L-proline synthons are disclosed.
  • Deuterated synthons include, but are not limited to, for example, the following compounds:
  • 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.
  • 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.
  • 5-azaspiro[2.4]heptane-4,5-dicarboxylic acid, 5-(1,1-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.
  • the base is diisopropylethylamine.
  • the coupling reagent is HATU.
  • the 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.
  • (4S) 4-oxazolidinecarboxylic acid, hydrochloride is treated with an amine protecting reagent.
  • the amine protecting reagent is di-tert-butyl dicarbonate.
  • 3,4-oxazolidinedicarboxylic acid, 3-(1,1-dimethylethyl) ester, (4S)- is commercially available from JPM2 Pharmaceuticals.
  • the reaction is carried out in an organic solvent in the presence of a base.
  • the organic solvent is acetonitrile.
  • the base is 4-dimentylaminopyridine (DMAP).
  • 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.
  • the amine is (3-chloro-2-fluorophenyl) methanamine.
  • the organic solvent is DMF.
  • the base is diisopropylethylamine.
  • the coupling reagent is HATU.
  • the 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.
  • 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.
  • the amine is (3-chloro-2-fluorophenyl) methanamine.
  • the organic solvent is DMF.
  • the base is diisopropylethylamine.
  • the coupling reagent is HATU.
  • Step 3 the 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.
  • 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.
  • Additional starting materials that can readily be converted to Central-L-B-Synthons include, but are not limited to: (S)-1-(tert-butoxycarbonyl)-2,3-dihydro-1H-pyrrole-2-carboxylic acid, CAS 90104-21-5, available from Ark Pharm; cyclopent-1-ene-1,2-dicarboxylic acid, CAS 3128-15-2, purchased from Ark Pharm; imidazole, 1H-imidazole-1,2-dicarboxylic acid, 1-(1,1-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,
  • the compounds (2S,3S,4S)-4-fluoro-3-methoxy-pyrrolidine-1,2-dicarboxylic acid 1-tert-butyl ester and (2R,3R,4R)-3-fluoro-4-methoxy-pyrrolidine-1,2-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.
  • Alkynyl synthons include but are not limited to:
  • R 30 is independently chosen at each occurrence from hydrogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, (aryl)C 0 -C 4 alkyl, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, (phenyl)C 0 -C 4 alkyl, (4- to 7-membered heterocycloalkyl)C 0 -C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; (5- or 6-membered unsaturated or aromatic heterocycle)C 0 -C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; COOH, Si(CH 3 ) 3 , COOR 30a , C 2 -C 6 alkanoyl, —B(OH) 2 , —C(O)(CH 2 ) 1-4 S(O)R 21 , —P(O)(OR 21 )(OR 22 ),
  • R 30a is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl-, (aryl)C 0 -C 4 alkyl-, (3- to 7-membered heterocycloalkyl)C 0 -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)C 0 -C 4 alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S, each of which R 30a can be optionally substituted.
  • Alkyne synthons can be prepared and coupled by a person skilled in the art.
  • Amashiro discloses alkynyl amines in WO 2008/047831.
  • Aryl alkynes are disclosed in Tang, T. et al., J. Org. Chem., 2013, 78, 3170-3175.
  • Alkynes can be synthesized from primary alcohols; see, Quesada, R. and Taylor, R. J. K., Tett. Lett., 2005, 46, 6473-6476.
  • Alkynes can be synthesized from aldehydes using an in situ preparation of dimethyldiazomethylphosphonate; see Roth, G. J. et al., Synthesis, 2004, 59-62.
  • Bromoalkenes can be converted to alkynes; see, Okutani, M. and Mori, Y., J. Org. Chem., 2009, 74, 442-444.
  • (Z)-1-Bromoalkenes can be converted to alkynes; see, Kuang, C., et al., Tetrahedron, 2005, 61, 4043-4052.
  • the stating material is treated with an organic solvent, an acylating reagent and a catalyst to add the R 6 substitutent.
  • the organic solvent is toluene.
  • the acylating reagent is acetyl chloride.
  • the catalyst is tin tetrachloride.
  • the starting material in Step 1 is treated with an inorganic cyanide and organometallic catalyst to generate a cyano group at the R 6 position. The cyano group is treated with a oxime to generate an amide at the R 6 position.
  • Step 2 the nitrogen of the heteroaryl ring is treated with a tert-butyl haloacetate, an organic solvent and a base.
  • the halo group, LG 2 is bromine.
  • the organic solvent is acetonitrile.
  • the base is potassium carbonate.
  • the tert-butyl ester is treated with an organic acid and an organic solvent to generate an acid.
  • the organic acid is trifluoroacetic acid.
  • the organic solvent is dichloromethane. This chemistry is illustrated in Route 5a.
  • the stating material is treated with an organic solvent, an acylating reagent and a catalyst to add the R 6 substitutent.
  • the organic solvent is toluene.
  • the acylating reagent is acetyl chloride.
  • the catalyst is tin tetrachloride.
  • the starting material in Step 1 is treated with an inorganic cyanide and organometallic catalysts to add a cyano group at the R 6 position. The cyano group is treated with an oxime to generate an amide at the R 6 position.
  • Step 2 the nitrogen of the heteroaryl ring is treated with a tert-butyl haloacetate, an organic solvent and a base.
  • the halo group, LG 2 is bromine.
  • the organic solvent is acetonitrile.
  • the base is potassium carbonate.
  • the tert-butyl ester is treated with an organic acid and an organic solvent to generate a carboxylic acid.
  • the organic acid is trifluoroacetic acid.
  • the organic solvent is dichloromethane. This chemistry is illustrated in Route 5b.
  • central-LB synthons and a carboxylic acid (C(O)-A) are combined and treated with an organic solvent, a base, and coupling agent.
  • the solvent is DMF.
  • the base is N,N-diisopropylethylamine.
  • the coupling reagent is (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate) (HATU).
  • the central-LB synthons and carboxylic acid (C(O)-A) are combined and treated with an organic solvent, a base, and coupling agent.
  • the solvent is DMF.
  • the base is N,N-diisopropylethylamine.
  • the coupling reagent is (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate) (HATU).
  • the proline derivative from Example 6 Scheme 6b is treated with two organometallic catalysts, trimethylsilylacetylene, a base and organic solvent at an elevated temperature to generate a trimethylsilyl protected alkyne.
  • the first organometallic catalyst is copper iodide.
  • the second organometallic catalyst is tetrakis(triphenylphosphine)palladium(0).
  • the base is triethylamine.
  • the organic solvent is DMF.
  • the reaction is deoxygenated by bubbling argon into the pressure vessel.
  • the reaction is treated at an elevated temperature. In one embodiment, the reaction is treated at about 80 to about 105° C.
  • the trimethylsilyl protected akyne is treated with a fluoride reagent and an organic solvent.
  • the fluoride reagent is tetrabutylammonium fluoride.
  • the organic solvent is tetrahydrofuran.
  • the alkyne is treated with LG-R 30 , two organometallic catalysts, a base, and an organic solvent at an elevated temperature.
  • the first organometallic catalyst is copper iodide.
  • the second organometallic catalyst is bis(triphenylphosphine)palladium(II) dichloride.
  • the base is triethylamine.
  • the organic solvent is DMF.
  • the elevated temperature is at about 80 to about 95° C.
  • the LG R 30 group is an heteroaryl halide.
  • the LG-R 30 group is an aryl halide. This chemistry is illustrated in Scheme 7a.
  • the proline derivative from Example 6 Scheme 6b is treated with two organometallic catalysts, an alkyne, a base and organic solvent at an elevated temperature to generate an alkyne of Formula I.
  • the first organometallic catalyst is copper iodide.
  • the second organometallic catalyst is tetrakis(triphenylphosphine)palladium(0).
  • the base is triethylamine.
  • the organic solvent is DMF.
  • the reaction is deoxygenated by bubbling argon into the pressure vessel.
  • the reaction is treated at an elevated temperature. In one embodiment, the reaction is treated at about 80 to about 105° C.
  • the proline derivative from Example 6 Scheme 6b is treated with two organometallic catalysts, trimethylsilylacetylene, a base and organic solvent at an elevated temperature to generate a trimethylsilyl protected alkyne.
  • the first organometallic catalyst is copper iodide.
  • the second organometallic catalyst is tetrakis(triphenylphosphine)palladium(0).
  • the base is triethylamine.
  • the organic solvent is DMF.
  • the reaction is deoxygenated by bubbling argon into the pressure vessel.
  • the reaction is treated at an elevated temperature. In one embodiment, the reaction is treated at about 80 to about 105° C.
  • the trimethylsilyl protected akyne is treated with a fluoride reagent and an organic solvent.
  • the fluoride reagent is tetrabutylammonium fluoride.
  • the organic solvent is tetrahydrofuran.
  • the alkyne is treated with LG-R 30 , two organometallic catalysts, a base, and an organic solvent at an elevated temperature.
  • the first organometallic catalyst is copper iodide.
  • the second organometallic catalyst is bis(triphenylphosphine)palladium(II) dichloride.
  • the base is triethylamine.
  • the organic solvent is DMF.
  • the elevated temperature is at about 80 to about 95° C.
  • the LG R 30 group is an heteroaryl halide.
  • the LG-R 30 group is an aryl halide. This chemistry is illustrated in Scheme 7b.
  • the proline derivative from Example 6 Scheme 6b is treated with two organometallic catalysts, an alkyne, a base and organic solvent at an elevated temperature to generate an alkyne of Formula I.
  • the first organometallic catalyst is copper iodide.
  • the second organometallic catalyst is tetrakis(triphenylphosphine)palladium(0).
  • the base is triethylamine.
  • the organic solvent is DMF.
  • the reaction is deoxygenated by bubbling argon into the pressure vessel.
  • the reaction is treated at an elevated temperature. In one embodiment, the reaction is treated at about 80 to about 105° C.
  • a TMS alkyne from Example 7 Scheme 7a or 7b can be treated with an inorganic fluoride, an organic solvent and carbon dioxide to generate a carboxylic acid.
  • the inorganic fluoride is cesium fluoride.
  • the organic solvent is dimethyl sulfoxide.
  • the resulting carboxylic acid can be treated with a sulfonamide anion to generate a compound of Formula I.
  • reaction mixture was diluted with 1M citric acid solution (200 ml+NaCl solid 20 gm) and extracted with DCM (150 mL ⁇ 2), the organic layer was then washed with an aqueous solution of NaHCO 3 (100 ml) and washed with water (100 ml), brine (100 ml) and dried over Na 2 SO 4 and concentrated under reduced pressure. The remaining residue was purified by column chromatography (eluted with DCM/EtOAc) to give (2S,4R)-tert-butyl 2-((3-chloro-2-fluorobenzyl)carbamoyl)-4-fluoropyrrolidine-1-carboxylate.
  • 1-(5-Bromo-1H-indol-3-yl)ethanone (2) was prepared from 5-bromoindole according to the procedure published by MacKay et al. (MacKay, J. A.; Bishop, R.; Rawal, V. H. Org. Lett. 2005, 7, 3421-3424.)
  • Triethylamine (2.53 mL, 18.1 mmol) and 2.56 mL (18.1 mmol) of ethynyltrimethylsilane were added under argon.
  • the pressure vessel was capped and heated at 100° C. overnight. Then the reaction mixture was cooled to RT and the solvent was removed under reduced pressure.
  • Triethylamine (2.0 mL) and 0.250 g (0.5 mmol) of (2S,4R)-1-(2-(3-Acetyl-5-ethynyl-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide were added under argon.
  • the pressure vessel was capped and heated at 100° C. overnight. Then the reaction mixture was cooled to RT and the solvent was removed under reduced pressure.
  • Compound 2 3-(3-Acetyl-1-(2-((2S,4R)-2-((3-chloro-2-fluorobenzyl)carbamoyl)-4-fluoropyrrolidin-1-yl)-2-oxoethyl)-1H-indol-5-yl)propiolic acid
  • (2S,4R)-1-(tert-butoxycarbonyl)-4-fluoropyrrolidine-2-carboxylic acid (2a, 2.33 g) was dissolved in DMF (50 ml) and DIEA (8.6 ml) was added, followed by the addition of (3-chloro-2-fluorophenyl) methanamine (2b, 3.18 gm) at 5° C. Then HATU (8 gm) was added slowly at same temperature. The reaction mixture was then stirred for 18 h at RT.
  • reaction mixture was diluted with 1M citric acid solution (200 ml+NaCl solid 20 gm) and extracted with DCM (150 mL ⁇ 2), the organic layer was then washed with an aqueous solution of NaHCO 3 (100 ml) and washed with water (100 ml), brine (100 ml) and dried over Na 2 SO 4 and concentrated under reduced pressure. The remaining residue was purified by column chromatography (eluent: DCM/EtOAc) to give (2S,4R)-tert-butyl 2-((3-chloro-2-fluorobenzyl)carbamoyl)-4-fluoropyrrolidine-1-carboxylate (2c).
  • 1-(6-Bromo-1H-indol-3-yl)ethanone (2f) was prepared from 6-bromoindole (2e) according to the procedure published by MacKay et al. (MacKay, J. A.; Bishop, R.; Rawal, V. H. Org. Lett. 2005, 7, 3421-3424.)
  • tert-Butyl 2-(3-acetyl-5-bromo-1H-indol-1-yl)acetate (6c, 4.5 g) was stirred in 1:1 mixture of CH 2 Cl 2 -TFA (90 mL) at RT for 4 h. The volatiles were removed under reduced pressure. The residue was dissolved in 45 mL of DMF. 4.16 g (13.4 mmol) of hydrochloride salt of 2d was added to this solution, followed by 11 mL of N,N-diisopropylethylamine (63.7 mmol). The reaction mixture was cooled in an ice bath and 5.82 g of (15.3 mmol) HATU was added.
  • 1,1′-Carbonyldiimidazole (20 mg) was added with stirring to a heterogeneous solution of 6 (50 mg) in THF (4 mL) and stirred overnight at room temperature.
  • Compound 10 was synthesized using the procedure similar to the synthesis of compound 8. Thus, 100 mg of 2 in DMF was coupled with 93 mg of ethanesulfonamide using 45 mg of 1,1′-carbonyldiimidazole and 37 mg of NaH. The reaction mixture was poured into 50 mL of water and acidified with 2N aq. HCl. The separated solid was isolated by filtration, washed successively with water and tert-butylmethyl ether. The solid was dried under high vacuum to yield 11 mg of 10 as brown solid.
  • Compound 22b was prepared using the procedure similar to the synthesis of 6c. Thus, 23.5 g of 22a was coupled with 17.3 mL of tert-butyl bromoacetate using 32.5 g of potassium carbonate in 470 mL of acetonitrile at 90° C. for 3 h. Then the solvent was removed and water was added to the residue. The product was isolated by filtration, washed with water and dried. Finally, the product was washed with 2:1 mixture of tert-butylmethyl ether and heptane to get 29 g of 22b.
  • Compound 22c was prepared following similar procedure to the synthesis of 6f. 7.0 g of 22b was coupled with 28 mL of trimethylsilylacetylene using 4.69 g of Pd(PPh 3 ) 4 , 1.5 g of cuprous iodide and 28 mL of triethylamine in 70 mL of DMF at 100° C. overnight. After removal of the solvent, the crude product was purified by column chromatography over silica gel (eluent: 0-1% MeOH in dichloromethane) to get 15 g of 22c.
  • Compound 27 1-(2-((1R,3S,5R)-3-((2′-Chloro-2-fluoro-[1,1′-biphenyl]-3-yl)carbamoyl)-2-azabicyclo[3.1.0]hexan-2-yl)-2-oxoethyl)-5-(pyrimidin-2-ylethynyl)-1H-indazole-3-carboxamide
  • (2S,4R)-1-(tert-butoxycarbonyl)-4-fluoropyrrolidine-2-carboxylic acid (2a) was coupled with amine 28e using Ghosez's reagent following procedure described for synthesis of 22j (scheme 8).
  • 0.249 g of compound 2a was coupled with 0.292 g of amine 28e using 0.145 mL of Ghosez's reagent, after chromatographic purification, to get 0.286 g of 28f.
  • 2-Amino-6-bromopyridine (0.191 g) was coupled with 0.23 g of trans-fluoroproline 2a using 0.146 mL of Ghosez's reagent in presence of 0.522 ml of DIEA following the procedure used for the synthesis of 22j.
  • Crude product was purified by chromatography over silica gel (eluent: 0-0.1% MeOH in dichloromethane) to get 0.2 g of 39a as white solid.
  • Compound 64 1-(2-((1R,3S,5R)-3-((2′-chloro-2,4′-difluoro-[1,1′-biphenyl]-3-yl)carbamoyl)-2-azabicyclo[3.1.0]hexan-2-yl)-2-oxoethyl)-5-((5-fluoropyrimidin-2-yl)ethynyl)-1H-indazole-3-carboxamide
  • the acid 27a was coupled with 3-bromo-2fluoroaniline using Ghosez's reagent as described for the synthesis of 22j.
  • Compound 65 1-(2-((1R,3S,5R)-3-((6-bromopyridin-2-yl)carbamoyl)-2-azabicyclo[3.1.0]hexan-2-yl)-2-oxoethyl)-5-((4-fluoropyridin-2-yl)ethynyl)-1H-indazole-3-carboxamide
  • Table 1 shows illustrative compounds of Formula I with characterizing data.
  • the assay of Example 12 was used to determine the IC 50 '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 ⁇ M each.
  • the increase in color is recorded at OD 405 nm in a microplate in kinetic mode over 30 minutes with 30 second time points in a spectrofluorimeter.
  • IC 50 values are calculated by non-linear regression from the percentage of inhibition of complement factor D activity as a function of test compound concentration.
  • red blood cells RBC
  • rabbit erythrocyctes purchased from Complement Technologies
  • GVB Buffer 0.1% gelatin, 5 mM Veronal, 145 mM NaCl, 0.025% NaN 3 , pH 7.3
  • 10 mM final Mg-EGTA 10 mM final Mg-EGTA.
  • Cells are used at a concentration of 1 ⁇ 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 Normal Human Serum
  • 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, wherein R12 or R13 on the A group is an alkyne (R32) 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

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 15/818,559, filed Nov. 20, 2017, which is a continuation of U.S. application Ser. No. 14/631,090, filed Feb. 25, 2015, now U.S. Pat. No. 9,828,396, which claims the benefit of provisional U.S. Application No. 61/944,189, filed Feb. 25, 2014, provisional U.S. Application No. 62/022,916, filed Jul. 10, 2014, and provisional U.S. Application 62/046,783, filed Sep. 5, 2014. The entirety of each of these applications is hereby incorporated by reference for all purposes.
  • BACKGROUND
  • 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).
  • 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(H2O), which associates with factor B to form the C3(H2O)B complex. Complement factor D acts to cleave factor B within the C3(H2O)B complex to form Ba and Bb. The Bb fragment remains associated with C3(H2O) to form the alternative pathway C3 convertase C3(H2O)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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • Biocryst Pharmaceuticals U.S. Pat. No. 6,653,340 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.
  • 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.
  • 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.
  • 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.
  • Japan Tobacco Inc. PCT patent publication WO1999/048492 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.
  • Ferring B. V. and Yamanouchi Pharmaceutical Co. lTD. PCT patent publication WO1993/020099 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.
  • Alexion Pharmaceuticals PCT patent publication WO1995/029697 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).
  • 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
  • It has been discovered that a compound of Formula I, or a pharmaceutically acceptable salt or composition thereof, wherein R12 or R13 on the A group is an alkyne (R32), is a superior inhibitor of complement factor D. In one embodiment, the alkyne is directly linked to the A group. In another embodiment, the alkyne is indirectly linked to the A group through a linking moiety, defined further below.
  • 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.
  • 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.
  • 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.
  • 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.
  • The disclosure provides compounds of Formula I
  • Figure US20180291047A1-20181011-C00001
  • and the pharmaceutically acceptable salts and compositions thereof, wherein:
  • Q1 is N(R1) or C(R1R1′);
  • Q2 is C(R2R2′), C(R2R2′)—C(R2R2′), S, O, N(R2) or C(R2R2′)O;
  • Q3 is N(R3), S, or C(R3R3′);
  • X1 and X2 are independently N, CH, or CZ, or X1 and X2 together are C═C; and
  • wherein Q1, Q2, Q3, X1, and X2 are selected such that a stable compound results.
  • Non-limiting examples of the
  • Figure US20180291047A1-20181011-C00002
  • ring are illustrated below (any of which can be otherwise substituted with R1, R1′, R2, R2′, R3, and R3′) as described in more detail below.
  • Figure US20180291047A1-20181011-C00003
    Figure US20180291047A1-20181011-C00004
    Figure US20180291047A1-20181011-C00005
  • wherein q is 0, 1, 2 or 3 and r is 1, 2 or 3.
  • R and R′ are independently chosen from H, alkyl, cycloalkyl, cycloalkylalkyl, heterocycle, heterocycloalkyl, aryl, arylalkyl, 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 mixtures 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, Cl, NH2, CH3, CH2D, CHD2, or CD3.
  • R1, R1′, R2, R2′, R3, and R3′ are independently chosen at each occurrence, as appropriate, and only where a stable compound results, from hydrogen, halogen, hydroxyl, nitro, cyano, amino, C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C6alkoxy, C2-C6alkynyl, C2-C6alkanoyl, C1-C6thioalkyl, hydroxyC1-C6alkyl, aminoC1-C6alkyl, —C0-C4alkylNR9R10, —C(O)OR9, —OC(O)R9, —NR9C(O)R10, —C(O)NR9R10, —OC(O)NR9R10, —NR9C(O)OR10, C1-C2haloalkyl, and C1-C2haloalkoxy, where R9 and R10 are independently chosen at each occurrence from hydrogen, C1-C6alkyl, (C3-C7cycloalkyl)C0-C4alkyl, —C0-C4alkyl(C3-C7cycloalkyl), and —O—C0-C4alkyl(C3-C7cycloalkyl).
  • In alternative embodiments, 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; R2 and R2′ may be taken together to form a 3- to 6-membered carbocyclic spiro ring; or R2 and R2′ may be taken together to form a 3- to 6-membered heterocyclic spiro ring; each of which spiro ring each of which ring may be unsubstituted or substituted with 1 or more substituents independently chosen from halogen (and in particular F), hydroxyl, cyano, —COOH, C1-C4alkyl (including in particular methyl), C2-C4alkenyl, C2-C4alkynyl, C1-C4alkoxy, C2-C4alkanoyl, hydroxyC1-C4alkyl, (mono- and di-C1-C4alkylamino)C0-C4alkyl, —C0-C4alkyl(C3-C7cycloalkyl), —O—C0-C4alkyl(C3-C7cycloalkyl), C1-C2haloalkyl, and C1-C2haloalkoxy.
  • In alternative embodiments, R1 and R2 may be taken together to form a 3-membered carbocyclic ring; R1 and R2 may be taken together to form a 4- to 6-membered carbocyclic or aryl ring or a 4- to 6-membered heterocyclic or heteroaryl ring containing 1 or 2 heteroatoms independently chosen from N, O, and S; or R2 and R3, if bound to adjacent carbon atoms, may be taken together to form a 3- to 6-membered carbocyclic or aryl ring or a 3- to 6-membered heterocyclic or heteroaryl ring; each of which ring may be unsubstituted or substituted with 1 or more substituents independently chosen from halogen (and in particular F), hydroxyl, cyano, —COOH, C1-C4alkyl (including in particular methyl), C2-C4alkenyl, C2-C4alkynyl, C1-C4alkoxy, C2-C4alkanoyl, hydroxyC1-C4alkyl, (mono- and di-C1-C4alkylamino)C0-C4alkyl, —C0-C4alkyl(C3-C7cycloalkyl), —O—C0-C4alkyl(C3-C7cycloalkyl), C1-C2haloalkyl, and C1-C2haloalkoxy.
  • In alternative embodiments, R1 and R1′, R2 and R2′, or R3 and R3′ 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.
  • A is a group chosen from:
  • Figure US20180291047A1-20181011-C00006
    Figure US20180291047A1-20181011-C00007
  • R4 is chosen from —CHO, —CONH2, C2-C6alkanoyl, hydrogen, —SO2NH2, —C(CH2)2F, —CH(CF3)NH2, C1-C6alkyl, —C0-C4alkyl(C3-C7cycloalkyl),
  • Figure US20180291047A1-20181011-C00008
  • 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, C1-C2alkyl, C1-C2alkoxy, —C0-C2alkyl(mono- and di-C1-C4alkylamino), C1-C2haloalkyl, and C1-C2haloalkoxy.
  • R5 and R6 are independently chosen from —CHO, —C(O)NH2, —C(O)NH(CH3), C2-C6alkanoyl, hydrogen, hydroxyl, halogen, cyano, nitro, —COOH, —SO2NH2, vinyl, C1-C6alkyl (including methyl), C2-C6alkenyl, C1-C6alkoxy, —C0-C4alkyl(C3-C7cycloalkyl), P—C(O)C0-C4alkyl(C3-C7cycloalkyl), —P(O)(OR9)2, —OC(O)R9, —C(O)OR9, —C(O)N(CH2CH2R9)(R10), —NR9C(O)R10, phenyl, or 5- to 6-membered heteroaryl.
  • Each R5 of 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, C1-C2alkyl, C1-C4alkoxy, —C0-C2alkyl(mono- and di-C1-C4alkylamino), C1-C2haloalkyl, and C1-C2haloalkoxy.
  • R6′ is hydrogen, halogen, hydroxyl, C1-C4alkyl, —C0-C4alkyl(C3-C7cycloalkyl), or C1-C4alkoxy; or R6 and R6′ may be taken together to form an oxo, vinyl, or imino group.
  • R7 is hydrogen, C1-C6alkyl, or —C0-C4alkyl(C3-C7cycloalkyl).
  • R8 and R8′ are independently chosen from hydrogen, halogen, hydroxyl, C1-C6alkyl, —C0-C4alkyl(C3-C7cycloalkyl), C1-C6alkoxy, and (C1-C4alkylamino)C0-C2alkyl; or R8 and R8′ are taken together to form an oxo group; or R8 and R8′ can be taken together with the carbon that they are bonded to form a 3-membered carbocyclic ring.
  • R16 is absent or may include one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, C1-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, —C0-C4alkyl(mono- and di-C1-C6alkylamino), —C0-C4alkyl(C3-C7cycloalkyl), C1-C2haloalkyl, and C1-C2haloalkoxy.
  • R19 is hydrogen, C1-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, —SO2C1-C6alkyl, (mono- and di-C1-C6alkylamino)C1-C4alkyl, —C0-C4alkyl(C3-C7cycloalkyl), —C0-C4alkyl(C3-C7heterocycloalkyl), —C0-C4alkyl(aryl), C0-C4alkyl(heteroaryl), and wherein R19 other than hydrogen is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, amino, —COOH, and —C(O)OC1-C4alkyl.
  • X11 is N or CR11.
  • X12 is N or CR12.
  • X13 is N or CR13.
  • X14 is N or CR14.
  • No more than 2 of X11, X12, X13, and X14 are N.
  • One of R12 and R13 is chosen from R31 and the other of R12 and R13 is chosen from R32:
  • R31 is chosen from hydrogen, halogen, hydroxyl, nitro, cyano, amino, —COOH, C1-C2haloalkyl, C1-C2haloalkoxy, C1-C6alkyl, —C0-C4alkyl(C3-C7cycloalkyl), C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, C2-C6alkenyloxy, —C(O)OR9, C1-C6thioalkyl, —C0-C4alkylNR9R10, —C(O)NR9R10, —SO2R9, —SO2NR9R10, —OC(O)R9, and —C(NR9)NR9R10, each of which R31 other than hydrogen, halogen, hydroxyl, nitro, cyano, C1-C2haloalkyl, and C1-C2haloalkoxy is unsubstituted or substituted with one or more substituents independently selected from halogen, hydroxyl, nitro, cyano, amino, —COOH, —CONH2 C1-C2haloalkyl, and C1-C2haloalkoxy, and each of which R31 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, C1-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, (mono- and di-C1-C6alkylamino)C0-C4alkyl, C1-C6alkylester, —C0-C4alkyl)(C3-C7cycloalkyl), C1-C2haloalkyl, and C1-C2haloalkoxy;
  • R32 is —C2-C6alkynylR30, and each R32 can be optionally substituted. Examples of R32 include, for example, but are not limited to,
  • Figure US20180291047A1-20181011-C00009
  • In an alternative embodiment, R12 and R13 are each independently selected from an R32 moiety.
  • R11, R14, and R15 are independently chosen at each occurrence from hydrogen, halogen, hydroxyl, nitro, cyano, —O(PO)(OR9)2, —(PO)(OR9)2, C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, 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, C1-C6alkoxy, C1-C6thioalkyl, —C0-C4alkyl(mono- and di-C1-C6alkylamino), —C0-C4alkyl(C3-C7cycloalkyl), —C0-C4alkoxy(C3-C7cycloalkyl), C1-C2haloalkyl, and C1-C2haloalkoxy.
  • R21 and R22 are independently chosen at each occurrence from hydrogen, hydroxyl, cyano, amino, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, (C3-C7cycloalkyl)C0-C4alkyl, (phenyl)C0-C4alkyl, —C1-C4alkylOC(O)OC1-C6alkyl, —C1-C4alkylOC(O)C1-C6alkyl, —C1-C4alkylC(O)OC1-C6alkyl, (4- to 7-membered heterocycloalkyl)C0-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, and each R21 and R22 can be optionally substituted.
  • R23 is independently chosen at each occurrence from C1-C6alkyl, C1-C6haloalkyl, (aryl)C0-C4alkyl, (C3-C7cycloalkyl)C0-C4alkyl, (phenyl)C0-C4alkyl, (4- to 7-membered heterocycloalkyl)C0-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, and each R23 can be optionally substituted.
  • 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, and each R24 and R25 can be optionally substituted.
  • R30 is independently chosen at each occurrence from hydrogen, C1-C6alkyl, C1-C6haloalkyl, (aryl)C0-C4alkyl, (C3-C7cycloalkyl)C0-C4alkyl, (phenyl)C0-C4alkyl, (4- to 7-membered heterocycloalkyl)C0-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; (5- or 6-membered unsaturated or aromatic heterocycle)C0-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; COOH, Si(CH3)3, COOR30a, C2-C6alkanoyl, —B(OH)2, —C(O)(CH2)1-4S(O)R21, —P(O)(OR21)(OR22), —P(O)(OR21)R22, —P(O)R21R22, —NR9P(O)(NHR21)(NHR22), —NR9P(o)(OR21)(NHR22), —NR9P(O)(OR21)(OR22), —C(S)R21, —NR21SO2R22, —NR9S(O)NR10R22, —NR9SO2NR10R22, —SO2NR9COR22, —SO2NR9CONR21R22, —NR21SO2R22, —C(O)NR21SO2R22, —C(NH2)NR9R22, —C(NH2)NR9S(O)2R22, —NR9C(O)OR10, —NR21OC(O)R22, —(CH2)1-4C(O)NR21R22, —C(O)R24R25, —NR9C(O)R21, —C(O)R21, —NR9C(O)NR9R10, —NR9C(O)NR24R25, —(CH2)1-4OC(O)R21, each of which R30 can be optionally substituted.
  • R30a is C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, (C3-C7cycloalkyl)C0-C4alkyl-, (aryl)C0-C4alkyl-, (3- to 7-membered heterocycloalkyl)C0-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, each of which R30a can be optionally substituted.
  • L is a bond or is chosen from the formulas
  • Figure US20180291047A1-20181011-C00010
  • where R17 is hydrogen, C1-C6alkyl, or —C0-C4alkyl(C3-C7cycloalkyl) and R18 and R18′ are independently chosen from hydrogen, halogen, hydroxymethyl, and methyl; and m is 0, 1, 2, or 3.
  • B is a monocyclic or bicyclic carbocyclic; a monocyclic or bicyclic carbocyclic-oxy group; 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; C2-C6alkenyl; C2-C6alkynyl; —(C0-C4alkyl)(aryl); —(C0-C4alkyl)(heteroaryl); or —(C0-C4alkyl)(biphenyl).
  • Each of which B is unsubstituted or substituted with one or more substituents independently chosen from R33 and R34, and 0 or 1 substituents chosen from R35 and R36.
  • R33 is independently chosen from halogen, hydroxyl, —COOH, cyano, C1-C6alkyl, C2-C6alkanoyl, C1-C6alkoxy, —C0-C4alkylNR9R10, —SO2R9, C1-C2haloalkyl, and C1-C2haloalkoxy.
  • R34 is independently chosen from nitro, C2-C6alkenyl, C2-C6alkynyl, C1-C6thioalkyl, -JC3-C7cycloalkyl, —B(OH)2, -JC(O)NR9R23, -JOSO2OR21, —C(O)(CH2)1-4S(O)R21, —O(CH2)1-4S(O)NR21R22, -JOP(O)(OR21)(OR22), -JP(O)(OR21)(OR22), -JOP(O)(OR21)R22, -JP(O)(OR21)R22, -JOP(O)R21R22, -JP(O)R21R22, -JSP(O)(OR21)(OR22), -JSP(O)(OR21)(R22), -JSP(O)(R21)(R22), -JNR9P(O)(NHR21)(NHR22), -JNR9P(o)(OR21)(NHR22), -JNR9P(O)(OR21)(OR22), -JC(S)R21, -JNR21SO2R22, -JNR9S(O)NR10R22, -JNR9SO2NR10R22, -JSO2NR9COR22, -JSO2NR9CONR21R22, -JNR21SO2R22, -JC(O)NR21SO2R22, -JC(NH2)NR22, -JC(NH2)NR9S(O)2R22, -JOC(O)NR21R22, -JNR21C(O)OR22, -JNR21OC(O)R22, —(CH2)1-4C(O)NR21R22, -JC(O)R24R25, -JNR9C(O)R21, -JC(O)R21, -JNR9C(O)NR10R22, —CCR21, —(CH2)1-4OC(O)R21, and -JC(O)OR23; each of which R34 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(O)(OH)2, C1-C6alkyl, —C0-C4alkyl(C3-C7cycloalkyl), C1-C6alkoxy, —C0-C2alkyl(mono- and di-C1-C4alkylamino), C1-C6alkylester, C1-C4alkylamino, C1-C4hydroxylalkyl, C1-C2haloalkyl, and C1-C2haloalkoxy.
  • R35 is independently chosen from naphthyl, naphthyloxy, indanyl, (4- to 7-membered heterocycloalkyl)C0-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 R35 is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, C1-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, (mono- and di-C1-C6alkylamino)C0-C4alkyl, C1-C6alkylester, —C0-C4alkyl(C3-C7cycloalkyl), —SO2R9, C1-C2haloalkyl, and C1-C2haloalkoxy.
  • R36 is independently chosen from tetrazolyl, (phenyl)C0-C2alkyl, (phenyl)C1-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 R36 is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, C1-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, (mono- and di-C1-C6alkylamino)C0-C4alkyl, C1-C6alkylester, —C0-C4alkyl(C3-C7cycloalkyl), —SO2R9, —OSi(CH3)2C(CH3)3, —Si(CH3)2C(CH3)3, C1-C2haloalkyl, and C1-C2haloalkoxy.
  • J is independently chosen at each occurrence from a covalent bond, C1-C4alkylene, —OC1-C4alkylene, C2-C4alkenylene, and C2-C4alkynylene.
  • Pharmaceutical compositions comprising a compound or salt of Formula I together with a pharmaceutically acceptable carrier are also disclosed.
  • 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.
  • 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 mediated 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.
  • 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
  • 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.
  • 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.
  • 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.
  • 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.
  • 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, 11C, 13C, 14C, 15N, 18F 31P, 32P, 35S, 36Cl, 125I respectively. The invention includes isotopically modified compounds of Formula I. In one embodiment, isotopically labelled compounds can be used 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.
  • 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 that achieves the desired result. Alternatively or in addition, isotopes of carbon, e.g., 13C and 14C, may be used. In one embodiment, the 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 α-deuterium kinetic isotope effect) or next to or near the site of bond breakage (a β-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. 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.
  • 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 R18, R18′, R33, R34, R35, and/or R36. 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, R30, and/or R30a. 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.
  • 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., CD3, CH2CD3 or CD2CD3.
  • The compound of the present invention may form a solvate with solvents (including water). Therefore, in one embodiment, the invention includes a solvated form of the active compound. The term “solvate” refers to a molecular complex of a compound of the present invention (including salts thereof) with one or more solvent molecules. Examples of solvents are 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. D2O, d6-acetone, d6-DMSO. A solvate can be in a liquid or solid form.
  • A dash (“-”) that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, —(C═O)NH2 is attached through carbon of the keto (C═O) group.
  • 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., ═O) 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.
  • 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.
  • 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, Cl, 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, C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, —C1-C6alkoxy, C2-C6alkanoyl, C1-C6alkylester, (mono- and di-C1-C6alkylamino)C0-C2alkyl, C1-C2haloalkyl, hydoxyC1-C6alkyl, ester, carbamate, urea, sulfonamide, —C1-C6alkyl(heterocyclo), C1-C6alkyl(heteroaryl), —C1-C6alkyl(C3-C7cycloalkyl), O—C1-C6alkyl(C3-C7cycloalkyl), B(OH)2, phosphate, phosphonate and C1-C2haloalkoxy.
  • “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 C1-C2, C1-C3, or C1-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 C1-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 C1-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)C0-C4 alkyl, or —C0-C4alkyl(C3-C7cycloalkyl), the indicated group, in this case cycloalkyl, is either directly bound by a single covalent bond (C0alkyl), 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 —O—C0-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.
  • “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-C8alkenyl, 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.
  • “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-C8alkynyl 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.
  • “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 C1-C4alkylene, C1-C3alkylene, or C1-C2alkylene.
  • “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.
  • “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.
  • “Alkoxy” is an alkyl group as defined above covalently bound through an oxygen bridge (—O—). 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.
  • “Alkenyloxy” is an alkenyl group as defined covalently bound to the group it substitutes by an oxygen bridge (—O—).
  • “Alkanoyl” is an alkyl group as defined above covalently bound through a carbonyl (C═O) bridge. The carbonyl carbon is included in the number of carbons, that is C2alkanoyl is a CH3(C═O)— group. In one embodiment, the alkanoyl group is optionally substituted as described above.
  • “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 —O(C═O)alkyl or a group of the formula —(C═O)Oalkyl.
  • “Amide” or “carboxamide” is —C(O)NRaRb wherein Ra and Rb are each independently selected from hydrogen, alkyl, for example, C1-C6alkyl, alkenyl, for example, C2-C6alkenyl, alkynyl, for example, C2-C6alkynyl, —C0-C4alkyl(C3-C7cycloalkyl), —C0-C4alkyl(C3-C7heterocycloalkyl), —C0-C4alkyl(aryl), and —C0-C4alkyl(heteroaryl); or together with the nitrogen to which they are bonded, Ra and Rb can form a C3-C7heterocyclic ring. In one embodiment, the Ra and Rb groups are each independently optionally substituted as described above.
  • “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. 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.
  • “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, —O—, linker.
  • “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.
  • “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 substituent.
  • “Aminoalkyl” is an alkyl group as previously described, substituted with at least one amino substituent.
  • “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. 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.
  • 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 (═O) moieties are pyrimidinonyl and 1,1-dioxo-thiomorpholinyl. The heterocycle groups herein are optionally substituted independently with one or more substituents described herein.
  • “Heterocyclicoxy group” is a monocyclic heterocyclic ring or a bicyclic heterocyclic group as described previously linked to the group it substitutes via an oxygen, —O—, linker.
  • “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, —O—, linker.
  • “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 heteroatom. 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.
  • 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.
  • 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.
  • “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, non-toxic, 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.
  • 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 non-toxic 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).
  • 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.
  • 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.
  • 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.
  • 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.
  • “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.
  • 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
  • According to the present invention, a compound of Formula I is provided:
  • Figure US20180291047A1-20181011-C00011
  • 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═O)A substituent. It has been surprisingly discovered that a compound of Formula I, or a pharmaceutically acceptable salt or composition thereof, wherein R12 or R13 on the A group is an alkyne (R32), 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.
  • Non-limiting examples of compounds falling within Formula I with variations in the variables e.g., A, B, R1-R3′, and L, are illustrated below. The disclosure includes all combinations of these definitions so long as a stable compound results.
  • Formulas II-XXX
  • 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 US20180291047A1-20181011-C00012
    Figure US20180291047A1-20181011-C00013
    Figure US20180291047A1-20181011-C00014
    Figure US20180291047A1-20181011-C00015
    Figure US20180291047A1-20181011-C00016
    Figure US20180291047A1-20181011-C00017
    Figure US20180291047A1-20181011-C00018
  • In these embodiments, it should be understood that where R1 or R3 is attached to a carbon, there can be two independent attachments as in R2/R2′ and these formulas should be considered to include all such variations.
  • 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 R12 and R13 Alkynyl Substituents
  • It has been surprisingly discovered that a compound of Formula I, a pharmaceutically acceptable salt or composition thereof, wherein R12 or R13 on the A group is an alkyne, is a superior inhibitor of complement factor D. In one embodiment, the alkyne is directly linked to the A group. In another embodiment, the alkyne is indirectly linked to the A group through a linking moiety, wherein the linking moiety is C1-C4alkylene.
  • One of R12 and R13 is chosen from R31 and the other of R12 and R13 is chosen from R32. In another embodiment each of R12 and R13 can be independently selected from R32.
  • R31 is chosen from hydrogen, halogen, hydroxyl, nitro, cyano, amino, —COOH, C1-C2haloalkyl, C1-C2haloalkoxy, C1-C6alkyl, —C0-C4alkyl(C3-C7cycloalkyl), C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, C2-C6alkenyloxy, —C(O)OR9, C1-C6thioalkyl, —C0-C4alkylNR9R10, —C(O)NR9R10, —SO2R9, —SO2NR9R10, —OC(O)R9, and —C(NR9)NR9R10, each of which R31 other than hydrogen, halogen, hydroxyl, nitro, cyano, C1-C2haloalkyl, and C1-C2haloalkoxy is unsubstituted or substituted with one or more substituents independently selected from halogen, hydroxyl, nitro, cyano, amino, —COOH, —CONH2 C1-C2haloalkyl, and C1-C2haloalkoxy, and each of which R31 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, C1-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, (mono- and di-C1-C6alkylamino)C0-C4alkyl, C1-C6alkylester, —C0-C4alkyl)(C3-C7cycloalkyl), C1-C2haloalkyl, and C1-C2haloalkoxy;
  • R32 is —C2-C6alkynylR30, and each R32 can be optionally substituted. Examples of R32 include, for example, but are not limited to,
  • Figure US20180291047A1-20181011-C00019
  • In certain embodiments, R32 is selected from:
  • Figure US20180291047A1-20181011-C00020
    Figure US20180291047A1-20181011-C00021
    Figure US20180291047A1-20181011-C00022
    Figure US20180291047A1-20181011-C00023
    Figure US20180291047A1-20181011-C00024
    Figure US20180291047A1-20181011-C00025
  • Non-Limiting R12/R13 Embodiments
  • In one embodiment, R12 is R32.
  • In one embodiment, R13 is R32.
  • In one embodiment, R12 is R32, which is C2-C4alkynylR30.
  • In one embodiment, R13 is R32, which is C2-C4alkynylR30.
  • In one embodiment, the disclosure provides compounds of Formula I, wherein;
  • one of R12 and R13 is H and the other of R12 and R13 is R32, where
  • R32 is C2-C6alkynylR30 (which can be, for example, C2-C3alkynylR30, C2-C4alkynylR30, C2-C5alkynylR30, or C2-C6alkynylR30), wherein R30 is independently chosen at each occurrence from hydrogen, C1-C6alkyl, C1-C6haloalkyl, (aryl)C0-C4alkyl, (C3-C7cycloalkyl)C0-C4alkyl, (phenyl)C0-C4alkyl, (4- to 7-membered heterocycloalkyl)C0-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; (5- or 6-membered unsaturated or aromatic heterocycle)C0-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; COOH, Si(CH3)3, COOR30a, C2-C6alkanoyl, —C(O)(CH2)1-4S(O)R21, —P(O)(OR21)(OR22), —P(O)R21R22, —NR21SO2R22, —NR9SO2NR10R22, —SO2NR9COR22, —SO2NR9CONR21R22, —C(O)NR21SO2R22, —NR9C(O)OR10, —NR10C(O)R22, —(CH2)1-4C(O)NR21R22, —C(O)R24R25, —C(O)R21, —NR9C(O)NR9R10, —NR9C(O)NR24R25, —(CH2)1-4OC(O)R21, each of which R30 can be optionally substituted;
  • wherein R9, R10, R21, R22, R24, R25, and R30a are as defined in the summary section above.
  • In another embodiment, the disclosure provides compounds of Formula I, wherein;
  • R1, R1′, R2, and R3′ are all hydrogen;
  • R2 is fluoro and R3 is hydrogen, —C0-C4alkyl(C3-C7cycloalkyl), or —O—C0-C4alkyl(C3-C7cycloalkyl);
  • R5 is hydrogen, halogen, or C1-C2alkyl;
  • R11, R13, R14, and R15 if present, are independently chosen at each occurrence from hydrogen, halogen, hydroxyl, amino, C1-C4alkyl, C1-C4alkoxy, —C0-C2alkyl(mono- and di-C1-C2alkylamino), trifluoromethyl, and trifluoromethoxy;
  • X12 is CR12; and
  • R12 is C2-C6alkynylR30.
  • In one embodiment, the disclosure provides compounds of Formula I, wherein;
  • m is 0 or 1;
  • R2 is halogen, R2′ is hydrogen or halogen, and R3 is hydrogen, halogen, —C0-C4alkyl(C3-C7cycloalkyl), or —O—C0-C4alkyl(C3-C7cycloalkyl);
  • R6 is —C(O)C1-C4alkyl, —C(O)NH2, —C(O)CF3, —C(O)(C3-C7cycloalkyl), or -ethyl(cyanoimino);
  • one of R12 and R13 is selected from hydrogen, halogen, C1-C4alkyl, C1-C4alkoxy, trifluoromethyl, and trifluoromethoxy; the other of R12 and R13 is R32, where
  • R32 is C2-C6alkynylR30, wherein R30 is independently chosen at each occurrence from hydrogen, C1-C6alkyl, C1-C6haloalkyl, (aryl)C0-C4alkyl, (C3-C7cycloalkyl)C0-C4alkyl, (phenyl)C0-C4alkyl, (4- to 7-membered heterocycloalkyl)C0-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; (5- or 6-membered unsaturated or aromatic heterocycle)C0-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; COOH, Si(CH3)3, COOR30a, C2-C6alkanoyl, —B(OH)2, —C(O)(CH2)1-4S(O)R21, —P(O)(OR21)(OR22), —P(O)(OR21)R22, —P(O)R21R22, —NR9P(O)(NHR21)(NHR22), —NR9P(O)(OR21)(NHR22), —NR9P(O)(OR21)(OR22), —C(S)R21, —NR21SO2R22, —NR9S(O)NR10R22, —NR9SO2NR10R22, —SO2NR9COR22, —SO2NR9CONR21R22, —NR21SO2R22, —C(O)NR21SO2R22, —C(NH2)NR9R22, —C(NH2)NR9S(O)2R22, —NR9C(O)OR1, —NR21OC(O)R22, —(CH2)1-4C(O)NR21R22, —C(O)R24R25, —NR9C(O)R21, —C(O)R21, —NR9C(O)NR9R10, —NR9C(O)NR24R25, —(CH2)1-4OC(O)R21, each of which R30 can be optionally substituted;
  • wherein R9, R10, R21, R22, R24, R25, and R30a are as defined in the summary section above.
  • In one embodiment, the disclosure provides compounds of Formula I, wherein;
  • one of R12 and R13 is hydrogen, hydroxyl, halogen, methyl, or methoxy; and the other of R12 and R13 is R32, where
  • R32 is C2-C6alkynylR30, wherein R30 is independently chosen at each occurrence from hydrogen, C1-C6alkyl, C1-C6haloalkyl, (aryl)C0-C4alkyl, (C3-C7cycloalkyl)C0-C4alkyl, (phenyl)C0-C4alkyl, (4- to 7-membered heterocycloalkyl)C0-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; (5- or 6-membered unsaturated or aromatic heterocycle)C0-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; COOH, Si(CH3)3, COOR30a, C2-C6alkanoyl, —B(OH)2, —C(O)(CH2)1-4S(O)R21, —P(O)(OR21)(OR22), —P(O)(OR21)R22, —P(O)R21R22, —NR9P(O)(NHR21)(NR22), —NR9P(O)(OR21)(NHR22), —NR9P(O)(OR21)(OR22), —C(S)R21, —NR21SO2R22, —NR9S(O)NR10R22, —NR9SO2NR10R22, —SO2NR9COR22, —SO2NR9CONR21R22, —NR21SO2R22, —C(O)NR21SO2R22, —C(NH2)NR9R22, —C(NH2)NR9S(O)2R22, —NR9C(O)OR1, —NR21OC(O)R22, —(CH2)1-4C(O)NR21R22, —C(O)R24R25, —NR9C(O)R21, —C(O)R21, —NR9C(O)NR9R10, —NR9C(O)NR24R25, —(CH2)1-4OC(O)R21, each of which R30 can be optionally substituted;
  • wherein R9, R10, R21, R22, R24, R25, and R30a are as defined in the summary section above.
  • In one embodiment, R32 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(O)(OH)2, C1-C6alkyl, C1-C6alkoxy, —C0-C2alkyl(mono- and di-C1-C4alkylamino), C1-C6alkylester, C1-C4alkylamino, C1-C4hydroxylalkyl, C1-C2haloalkyl, and C1-C2haloalkoxy.
  • Central Core Moiety
  • The central core moiety in Formula I is illustrated below:
  • Figure US20180291047A1-20181011-C00026
  • wherein:
  • Q1 is N(R1) or C(R1R1′);
  • Q2 is C(R2R2′), C(R2R2′)—C(R2R2′), S, O, N(R2) or C(R2R2′)O;
  • Q3 is N(R3), S, or C(R3R3′);
  • X1 and X2 are independently N or CH, or X1 and X2 together are C═C; and
  • wherein Q1, Q2, Q3, X1, and X2 are selected such that a stable compound results.
  • Non-limiting examples of the
  • Figure US20180291047A1-20181011-C00027
  • ring are illustrated below (any of which can be otherwise substituted with R1, R1′, R2, R2′, R3, and R3′) as described in more detail below.
  • Figure US20180291047A1-20181011-C00028
    Figure US20180291047A1-20181011-C00029
    Figure US20180291047A1-20181011-C00030
  • wherein q is, 2 or 3 and r is, 2 or 3.
  • R and R′ are independently chosen from H, alkyl, cycloalkyl, 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 mixtures 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, Cl, NH2, CH3, CH2D, CHD2, or CD3.
  • R1, R1′, R2, R2′, R3, and R3′ are independently chosen at each occurrence, as appropriate, and only where a stable compound results, from hydrogen, halogen, hydroxyl, nitro, cyano, amino, C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C6alkoxy, C2-C6alkynyl, C2-C6alkanoyl, C1-C6thioalkyl, hydroxyC1-C6alkyl, aminoC1-C6alkyl, —C0-C4alkylNR9R10, —C(O)OR9, —OC(O)R9, —NR9C(O)R10, —C(O)NR9R10, —OC(O)NR9R10, —NR9C(O)OR10, C1-C2haloalkyl, and C1-C2haloalkoxy, where R9 and R10 are independently chosen at each occurrence from hydrogen, C1-C6alkyl, (C3-C7cycloalkyl)C0-C4alkyl, —C0-C4alkyl(C3-C7cycloalkyl), and —O—C0-C4alkyl(C3-C7cycloalkyl).
  • Non-Limiting Central Core Embodiments
  • In alternative embodiments, 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; R2 and R2′ may be taken together to form a 3- to 6-membered carbocyclic spiro ring; or R2 and R2′ may be taken together to form a 3- to 6-membered heterocyclic spiro ring each of which ring may be unsubstituted or substituted with 1 or more substituents independently chosen from halogen (and in particular F), hydroxyl, cyano, —COOH, C1-C4alkyl (including in particular methyl), C2-C4alkenyl, C2-C4alkynyl, C1-C4alkoxy, C2-C4alkanoyl, hydroxyC1-C4alkyl, (mono- and di-C1-C4alkylamino)C0-C4alkyl, —C0-C4alkyl(C3-C7cycloalkyl), —O—C0-C4alkyl(C3-C7cycloalkyl), C1-C2haloalkyl, and C1-C2haloalkoxy.
  • In alternative embodiments, R1 and R2 may be taken together to form a 3-membered carbocyclic ring; R1 and R2 may be taken together to form a 4- to 6-membered carbocyclic or aryl ring or a 4- to 6-membered heterocyclic or heteroaryl ring containing 1 or 2 heteroatoms independently chosen from N, O, and S; or R2 and R3, if bound to adjacent carbon atoms, may be taken together to form a 3- to 6-membered carbocyclic or aryl ring or a 3- to 6-membered heterocyclic or heteroaryl ring each of which ring may be unsubstituted or substituted with one or more substituents independently chosen from halogen (and in particular F), hydroxyl, cyano, —COOH, C1-C4alkyl (including in particular methyl), C2-C4alkenyl, C2-C4alkynyl, C1-C4alkoxy, C2-C4alkanoyl, hydroxyC1-C4alkyl, (mono- and di-C1-C4alkylamino)C0-C4alkyl, —C0-C4alkyl(C3-C7cycloalkyl), —O—C0-C4alkyl(C3-C7cycloalkyl), C1-C2haloalkyl, and C1-C2haloalkoxy.
  • In one embodiment, the central core moiety is proline.
  • In one embodiment, the central core moiety is 4-fluoroproline.
  • In one embodiment, R1, R1′, R2′, R3, and R3′, if present, are all hydrogen; and R2 is fluoro.
  • In one embodiment, R1, R1′, R2′, and R3′, if present, are all hydrogen; and R2 is fluoro and R3 is —C0-C4alkyl(C3-C7cycloalkyl) or —O—C0-C4alkyl(C3-C7cycloalkyl).
  • 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.
  • 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.
  • In one embodiment, R1 is hydrogen and R2 is fluoro.
  • In one embodiment, R1 and R2 are joined to form a 3 membered ring.
  • The disclosure includes compounds of Formula I in which the central pyrrolidine is vinyl substituted, for example:
  • Figure US20180291047A1-20181011-C00031
  • In one embodiment, the compound of Formula I has the structure:
  • Figure US20180291047A1-20181011-C00032
  • 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 US20180291047A1-20181011-C00033
  • 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 US20180291047A1-20181011-C00034
  • Example of compounds having the modifications disclosed above include:
  • Figure US20180291047A1-20181011-C00035
  • Central Core L-B Substituents
  • The central core L-B substituents in Formula I are illustrated below:
  • Figure US20180291047A1-20181011-C00036
  • L is a bond or is chosen from the formulas:
  • Figure US20180291047A1-20181011-C00037
  • where R17 is hydrogen, C1-C6alkyl, or —C0-C4alkyl(C3-C7cycloalkyl) and R18 and R18′ are independently chosen from hydrogen, halogen, hydroxymethyl, and methyl; and m is 0, 1, 2, or 3.
  • B is a monocyclic or bicyclic carbocyclic; a monocyclic or bicyclic carbocyclic-oxy group; 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; C2-C6alkenyl; C2-C6alkynyl; —(C0-C4alkyl)(aryl); —(C0-C4alkyl)(heteroaryl); or —(C0-C4alkyl)(biphenyl).
  • Each of which B is unsubstituted or substituted with one or more substituents independently chosen from R33 and R34, and 0 or 1 substituents chosen from R35 and R36:
  • R33 is independently chosen from halogen, hydroxyl, —COOH, cyano, C1-C6alkyl, C2-C6alkanoyl, C1-C6alkoxy, —C0-C4alkylNR9R10, —SO2R9, C1-C2haloalkyl, and C1-C2haloalkoxy;
  • R34 is independently chosen from nitro, C2-C6alkenyl, C2-C6alkynyl, C1-C6thioalkyl, -JC3-C7cycloalkyl, —B(OH)2, -JC(O)NR9R23, -JOSO2OR21, —C(O)(CH2)1-4S(O)R21, —O(CH2)1-4S(O)NR21R22, -JOP(O)(OR21)(OR22), -JP(O)(OR21)(OR22), -JOP(O)(OR21)R22, -JP(O)(OR21)R22, -JOP(O)R21R22, -JP(O)R21R22, -JSP(O)(OR21)(OR22), -JSP(O)(OR21)(R22), -JSP(O)(R21)(R22), -JNR9P(O)(NHR21)(NHR22), -JNR9P(o)(OR21)(NHR22), -JNR9P(O)(OR21)(OR22), -JC(S)R21, -JNR21SO2R22, -JNR9S(O)NR1R22, -JNR9SO2NR1R22, -JSO2NR9COR22, -JSO2NR9CONR21R22, -JNR21SO2R22, -JC(O)NR21SO2R22, -JC(NH2)NR22, -JC(NH2)NR9S(O)2R22, -JOC(O)NR21R21R22, -JNR21C(O)OR22, -JNR21OC(O)R22, —(CH2)1-4C(O)NR21R22, -JC(O)R24R25, -JNR9C(O)R21, -JC(O)R21, -JNR9C(O)NR10R22, —CCR21, —(CH2)1-4OC(O)R21, and -JC(O)OR23; each of which R34 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(O)(OH)2, C1-C6alkyl, —C0-C4alkyl(C3-C7cycloalkyl), C1-C6alkoxy, —C0-C2alkyl(mono- and di-C1-C4alkylamino), C1-C6alkylester, C1-C4alkylamino, C1-C4hydroxylalkyl, C1-C2haloalkyl, and C1-C2haloalkoxy;
  • R35 is independently chosen from naphthyl, naphthyloxy, indanyl, (4- to 7-membered heterocycloalkyl)C0-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 R35 is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, C1-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, (mono- and di-C1-C6alkylamino)C0-C4alkyl, C1-C6alkylester, —C0-C4alkyl(C3-C7cycloalkyl), —SO2R9, C1-C2haloalkyl, and C1-C2haloalkoxy; and
  • R36 is independently chosen from tetrazolyl, (phenyl)C0-C2alkyl, (phenyl)C1-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 R36 is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, C1-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, (mono- and di-C1-C6alkylamino)C0-C4alkyl, C1-C6alkylester, —C0-C4alkyl(C3-C7cycloalkyl), —SO2R9, —OSi(CH3)2C(CH3)3, —Si(CH3)2C(CH3)3, C1-C2haloalkyl, and C1-C2haloalkoxy.
  • J is independently chosen at each occurrence from a covalent bond, C1-C4alkylene, —OC1-C4alkylene, C2-C4alkenylene, and C2-C4alkynylene.
  • In one embodiment, -L-B— is
  • Figure US20180291047A1-20181011-C00038
  • where
    R26 and R27 are independently chosen from hydrogen, halogen, hydroxyl, nitro, cyano, C1-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, C1-C6thioalkyl, —C0-C4alkyl(mono- and di-C1-C6alkylamino), —C0-C4alkyl(C3-C7cycloalkyl), —C0-C4alkoxy(C3-C7cycloalkyl), C1-C2haloalkyl, C1-C2haloalkoxy, and C1-C2haloalkylthio.
  • Non-Limiting L-B Embodiments
  • In another embodiment, -L-B— is
  • Figure US20180291047A1-20181011-C00039
    Figure US20180291047A1-20181011-C00040
  • wherein
  • R18 and R18′ are independently chosen from hydrogen, halogen, hydroxymethyl, and methyl; and m is 0 or 1; and
  • R26, R27, and R28 are independently chosen from hydrogen, halogen, hydroxyl, nitro, cyano, C1-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, C1-C6thioalkyl, (mono- and di-C1-C6alkylamino)C0-C4alkyl, (C3-C7cycloalkyl)C0-C4alkyl, (aryl)C0-C4alkyl-, (heteroaryl)C0-C4alkyl-, and —C0-C4alkoxy(C3-C7cycloalkyl); each of which R26, R27, and R28 other than hydrogen, halogen, hydroxyl, nitro, cyano, is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, amino, C1-C2alkoxy, C1-C2haloalkyl, (C3-C7cycloalkyl)C0-C4alkyl-, and C1-C2haloalkoxy; and
  • R29 is hydrogen, C1-C2alkyl, C1C2haloalkyl or —Si(CH3)2C(CH3)3.
  • In one embodiment, m is 0.
  • 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, 1-(2,2,2-trifluoroethyl)-1H-pyrazol-3-yl, 2,2-dichlorocyclopropylmethyl, or 2-fluoro-3-trimethylsilylphenyl is used.
  • 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, C1-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, C1-C6thioalkyl, (mono- and di-C1-C6alkylamino)C0-C4alkyl, (C3-C7cycloalkyl)C0-C4alkyl, —C0-C4alkoxy(C3-C7cycloalkyl), (phenyl)C0-C2alkyl, (pyridyl)C0-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, C1-C2alkyl, C1-C2alkoxy, —OSi(CH3)2C(CH3)3, —Si(CH3)2C(CH3)3, C1-C2haloalkyl, and C1-C2haloalkoxy.
  • In another embodiment, B is phenyl or pyridyl substituted with 1, 2, or 3 substituents chosen from chloro, bromo, hydroxyl, —SCF3, C1-C2alkyl, C1-C2alkoxy, trifluoromethyl, phenyl and trifluoromethoxy each of which substituents other than chloro, bromo, hydroxyl, —SCF3, can be optionally substituted.
  • In certain embodiments, B is a 2-fluoro-3-chlorophenyl or a 2-fluoro-3-trifluoromethoxyphenyl group.
  • In one embodiment, B is pyridyl, optionally substituted with halogen, C1-C2alkoxy, and trifluoromethyl.
  • In one embodiment, B is phenyl, substituted with 1, 2, or 3 substituents independently selected from halogen, C1-C2alkyl, C1-C2alkoxy, trifluoromethyl, and optionally substituted phenyl.
  • In one embodiment, R23 is independently chosen at each occurrence from (C3-C7cycloalkyl)C0-C4alkyl, (phenyl)C0-C4alkyl, (4- to 7-membered heterocycloalkyl)C0-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.
  • In one embodiment, B is selected from
  • Figure US20180291047A1-20181011-C00041
    Figure US20180291047A1-20181011-C00042
    Figure US20180291047A1-20181011-C00043
    Figure US20180291047A1-20181011-C00044
    Figure US20180291047A1-20181011-C00045
    Figure US20180291047A1-20181011-C00046
    Figure US20180291047A1-20181011-C00047
    Figure US20180291047A1-20181011-C00048
    Figure US20180291047A1-20181011-C00049
    Figure US20180291047A1-20181011-C00050
  • where R27 is hydrogen, methyl, or trifluoromethyl; R28 is hydrogen or halogen; and R29 is hydrogen, methyl, trifluoromethyl, or —Si(CH3)2C(CH3)3.
  • Central Core (C═O)A Substituent
  • The central core (C═O)A substituent in Formula I is illustrated below:
  • Figure US20180291047A1-20181011-C00051
  • A is a group chosen from:
  • Figure US20180291047A1-20181011-C00052
    Figure US20180291047A1-20181011-C00053
  • R4 is chosen from —CHO, —CONH2, C2-C6alkanoyl, hydrogen, —SO2NH2, —C(CH2)2F, —CH(CF3)NH2, C1-C6alkyl, —C0-C4alkyl(C3-C7cycloalkyl), —C(O)C0-C2alkyl(C3-C7cycloalkyl),
  • Figure US20180291047A1-20181011-C00054
  • 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, C1-C2alkyl, C1-C2alkoxy, —C0-C2alkyl(mono- and di-C1-C4alkylamino), C1-C2haloalkyl, and C1-C2haloalkoxy.
  • R5 and R6 are independently chosen from —CHO, —C(O)NH2, —C(O)NH(CH3), C2-C6alkanoyl, hydrogen, hydroxyl, halogen, cyano, nitro, —COOH, —SO2NH2, vinyl, C1-C6alkyl (including methyl), C2-C6alkenyl, C1-C6alkoxy, —C0-C4alkyl(C3-C7cycloalkyl), —C(O)C0-C4alkyl(C3-C7cycloalkyl), —P(O)(OR9)2, —OC(O)R9, —C(O)OR9, —C(O)N(CH2CH2R9)(R10), —NR9C(O)R10, phenyl, or 5- to 6-membered heteroaryl.
  • 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, —COOH, may be substituted with one or more substituents independently chosen from halogen, hydroxyl, amino, imino, cyano, cyanoimino, C1-C2alkyl, C1-C4alkoxy, —C0-C2alkyl(mono- and di-C1-C4alkylamino), C1-C2haloalkyl, and C1-C2haloalkoxy.
  • R6′ is hydrogen, halogen, hydroxyl, C1-C4alkyl, —C0-C4alkyl(C3-C7cycloalkyl), or C1-C4alkoxy; or R6 and R6′ may be taken together to form an oxo, vinyl, or imino group.
  • R7 is hydrogen, C1-C6alkyl, or —C0-C4alkyl(C3-C7cycloalkyl).
  • R8 and R8′ are independently chosen from hydrogen, halogen, hydroxyl, C1-C6alkyl, —C0-C4alkyl(C3-C7cycloalkyl), C1-C6alkoxy, and (C1-C4alkylamino)C0-C2alkyl; or R8 and R8′ are taken together to form an oxo group; or R8 and R8′ can be taken together with the carbon that they are bonded to form a 3-membered carbocyclic ring.
  • R16 is absent or may include one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, C1-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, —C0-C4alkyl(mono- and di-C1-C6alkylamino), —C0-C4alkyl(C3-C7cycloalkyl), C1-C2haloalkyl, and C1-C2haloalkoxy.
  • R19 is hydrogen, C1-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, —SO2C1-C6alkyl, (mono- and di-C1-C6alkylamino)C1-C4alkyl, —C0-C4alkyl(C3-C7cycloalkyl), —C0-C4alkyl(C3-C7heterocycloalkyl), —C0-C4alkyl(aryl), C0-C4alkyl(heteroaryl), and wherein R19 other than hydrogen is unsubstituted or substituted with 1 or more substituents independently chosen from halogen, hydroxyl, amino, —COOH, and —C(O)OC1-C4alkyl.
  • X11 is N or CR11.
  • X12 is N or CR12.
  • X13 is N or CR13.
  • X14 is N or CR14.
  • 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, —O(PO)(OR9)2, —(PO)(OR9)2, C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, 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, C1-C6alkoxy, C1-C6thioalkyl, —C0-C4alkyl(mono- and di-C1-C6alkylamino), —C0-C4alkyl(C3-C7cycloalkyl), —C0-C4alkoxy(C3-C7cycloalkyl), C1-C2haloalkyl, and C1-C2haloalkoxy.
  • In one embodiment, R5 and R6 are independently chosen from —CHO, —C(O)NH2, —C(O)NH(CH3), C2-C6alkanoyl, and hydrogen.
  • 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, C1-C2alkyl, C1-C4alkoxy, —C0-C2alkyl(mono- and di-C1-C4alkylamino), C1-C2haloalkyl, and C1-C2haloalkoxy.
  • In one embodiment, R8 and R8′ are independently hydrogen or methyl.
  • In one embodiment, R8 and R8′ are hydrogen.
  • In one embodiment, R7 is hydrogen or methyl.
  • In one embodiment, R7 is hydrogen.
  • Embodiments of Formulas IA, IB, IC, and ID
  • 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.
  • In one aspect, this disclosure includes compounds and salts of Formula IA:
  • Figure US20180291047A1-20181011-C00055
  • where
    R6, R13, and B may carry any of the definitions set forth herein for this variable.
  • In another aspect, this disclosure includes compounds and salts of Formula IB, IC, and ID.
  • Figure US20180291047A1-20181011-C00056
  • In Formulas IA, IB, IC, and ID, the variables may include any of the definitions set forth herein that results in a stable compound. In certain embodiments, the following conditions apply for Formula IB and IC.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 is H, R2 is F, R6 is alkanoyl, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 and R2 are joined to form a 3 membered ring, R6 is alkanoyl, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 is H, R2 is F, R6 is amide, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 and R2 are joined to form a 3 membered ring, R6 is amide, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 is H, R2 is F, R6 is alkanoyl, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 and R2 are joined to form a 3 membered ring, R6 is alkanoyl, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 is H, R2 is F, R6 is amide, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 and R2 are joined to form a 3 membered ring, R6 is amide, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 is H, R2 is F, R6 is alkanoyl, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is phenyl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 and R2 are joined to form a 3 membered ring, R6 is alkanoyl, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is phenyl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 is H, R2 is F, R6 is amide, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is phenyl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 and R2 are joined to form a 3 membered ring, R6 is amide, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is phenyl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 is H, R2 is F, R6 is alkanoyl, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is phenyl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 and R2 are joined to form a 3 membered ring, R6 is alkanoyl, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is phenyl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 is H, R2 is F, R6 is amide, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is phenyl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=0, R1 and R2 are joined to form a 3 membered ring, R6 is amide, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is phenyl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 is H, R2 is F, R6 is alkanoyl, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 and R2 are joined to form a 3 membered ring, R6 is alkanoyl, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 is H, R2 is F, R6 is amide, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 and R2 are joined to form a 3 membered ring, R6 is amide, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 is H, R2 is F, R6 is alkanoyl, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 and R2 are joined to form a 3 membered ring, R6 is alkanoyl, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 is H, R2 is F, R6 is amide, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 and R2 are joined to form a 3 membered ring, R6 is amide, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is heteroaryl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 is H, R2 is F, R6 is alkanoyl, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is phenyl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 and R2 are joined to form a 3 membered ring, R6 is alkanoyl, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is phenyl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 is H, R2 is F, R6 is amide, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is phenyl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 and R2 are joined to form a 3 membered ring, R6 is amide, R12 is alkynylR30, R30 is heteroaryl, R13 is H, and B is phenyl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 is H, R2 is F, R6 is alkanoyl, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is phenyl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 and R2 are joined to form a 3 membered ring, R6 is alkanoyl, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is phenyl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 is H, R2 is F, R6 is amide, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is phenyl.
  • In some embodiments, structures are provided including Formulas IB and IC, wherein m=1, R1 and R2 are joined to form a 3 membered ring, R6 is amide, R12 is H, R13 is alkynylR30, R30 is heteroaryl, and B is phenyl.
  • Embodiments of Formula VII
  • To further illustrate the invention, various embodiments of Formula VII. In one aspect, the disclosure includes compounds and salts of Formula VII:
  • Figure US20180291047A1-20181011-C00057
  • wherein:
  • R1, R2, R2′, and R3 are independently chosen from hydrogen, halogen, C1-C4alkyl, C1-C4alkoxy, —C0-C2alkylNR9R10, —C0-C4alkyl(C3-C7cycloalkyl), —O—C0-C4alkyl(C3-C7cycloalkyl), C1-C2haloalkyl, and C1-C2haloalkoxy;
  • R8 and R8′ are independently chosen from hydrogen, halogen, and methyl;
  • R5 is hydrogen, hydroxyl, cyano, —COOH, C1-C6alkyl, C1-C6alkoxy, C2-C6alkanoyl —C0-C4alkyl(C3-C7cycloalkyl), —C(O)C0-C4alkyl(C3-C7cycloalkyl, C1-C2haloalkyl, or C1-C2haloalkoxy;
  • R6 is —C(O)CH3, —C(O)NH2, —C(O)CF3, —C(O)(cyclopropyl), or -ethyl(cyanoimino); and
  • R11 and R14 are independently chosen from hydrogen, halogen, hydroxyl, amino, nitro, cyano, C1-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, C1-C6thioalkyl, —C0-C4alkyl(mono- and di-C1-C6alkylamino), —C0-C4alkyl(C3-C7cycloalkyl), —OC0-C4alkyl(C3-C7cycloalkyl), C1-C2haloalkyl, and C1-C2haloalkoxy.
  • Prodrugs of Formula I are also within the scope of the disclosure.
  • III. Pharmaceutical Preparations
  • 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 anti-inflammatory 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.
  • 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.
  • 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
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • “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.
  • 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.
  • V. Combination Therapy
  • 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.
  • 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.
  • 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.
  • Nonlimiting examples of active agents in these categories are:
  • Protease inhibitors: plasma-derived C1-INH concentrates, for example Cetor® (Sanquin), Berinert-P® (CSL Behring, Lev Pharma), and Cinryze®; and recombinant human C1-inhibitors, for example Rhucin®;
  • Soluble complement regulators: Soluble complement receptor 1 (TP10) (Avant Immunotherapeutics); sCR1-sLex/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: Compstatin/POT-4 (Potentia Pharmaceuticals); ARC1905 (Archemix);
  • Receptor agonists: PMX-53 (Peptech Ltd.); JPE-137 (Jerini); JSM-7717 (Jerini);
  • Others: Recombinant human MBL (rhMBL; Enzon Pharmaceuticals).
  • 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.
  • In another embodiment, 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. 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.
  • 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).
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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 Auranofin (Ridaura); Aurothioglucose (Solganal); Aurolate; Myochrysine; or any combination thereof.
  • 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.
  • 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-1a), Betaseron (interferon beta-1b), Copaxone (glatiramer acetate), Extavia (interferon beta-1b), Gilenya (fingolimod), Lemtrada (alemtuzumab), Novantrone (mitoxantrone), Plegridy (peginterferon beta-1a), Rebif (interferon beta-1a), Tecfidera (dimethyl fumarate), Tysabri (natalizumab), Solu-Medrol (methylpredni solone), High-dose oral Deltasone (prednisone), H.P. Acthar Gel (ACTH), and combinations thereof.
  • 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.
  • 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 S1P 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.
  • 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)2O di-tert-butyl dicarbonate
  • ACN Acetonitrile
  • ACOET, ETOAC ethyl acetate
  • CH3OH, MeOH Methanol
  • CsF Cesium fluoride
    CuI Cuprous iodide
    DCM, CH2Cl2 Dichloromethane
  • DIEA, DIPEA N,N-diisopropylethylamine DMA N,N-dimethylacetamide DMF N,N-dimethylformamide DMSO Dimethylsulfoxide
  • DPPA diphenyl phosphoryl azide
  • Et3N, TEA Triethylamine
  • EtOAc Ethyl acetate
    HATU 1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate
    iPr2NEt N,N-diisopropylethylamine
    K2CO3 Potassium carbonate
    Na2SO4 Sodium sulfate
    NaCl Sodium chloride
    NaH Sodium hydride
    NaHCO3 Sodium bicarbonate
  • NEt3 Trimethylamine
  • 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)
    RT Room temperature
    tBuOK potassium tert-butoxide
  • TEA Trimethylamine
  • Tf2O trifluoromethanesulfonic anhydride
    TFA Trifluoroacetic acid
  • THF Tetrahydrofuran TMSBr Bromotrimethylsilane
  • tR Retention time
  • General Methods
  • 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 C18 2.1×50 mm, 1.7 μm
      • Column Temperature: 40° C.
      • Mobile Phase: Solvent A: H2O+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, 50×4.6 mm, 5 m
      • Column Temperature: 40° C.
      • Mobile Phase: Solvent A: H2O/CH3OH/FA=90/10/0.1; Solvent B: H2O/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
  • 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(O)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.
  • Figure US20180291047A1-20181011-C00058
  • 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.
  • Figure US20180291047A1-20181011-C00059
  • 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.
  • Figure US20180291047A1-20181011-C00060
  • 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 US20180291047A1-20181011-C00061
  • 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.
  • Figure US20180291047A1-20181011-C00062
  • 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 chemistry is illustrated in Route 6.
  • Figure US20180291047A1-20181011-C00063
  • Example 2. Examples of Central Synthons
  • Figure US20180291047A1-20181011-C00064
    Figure US20180291047A1-20181011-C00065
  • Za is halogen.
  • In one embodiment, deuterated L-proline synthons are disclosed. Deuterated synthons include, but are not limited to, for example, the following compounds:
  • Figure US20180291047A1-20181011-C00066
  • 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 US20180291047A1-20181011-C00067
  • In Route 1a, 5-azaspiro[2.4]heptane-4,5-dicarboxylic acid, 5-(1,1-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.
  • In Route 1b, (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-(1,1-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.
  • In Route 1c, (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 US20180291047A1-20181011-C00068
  • 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.
  • 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.
  • In Route 2c, commercially available (2S,4R)-1-(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.
  • In Route 2d, commercially available (S)-1-(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.
  • Additional starting materials that can readily be converted to Central-L-B-Synthons include, but are not limited to: (S)-1-(tert-butoxycarbonyl)-2,3-dihydro-1H-pyrrole-2-carboxylic acid, CAS 90104-21-5, available from Ark Pharm; cyclopent-1-ene-1,2-dicarboxylic acid, CAS 3128-15-2, purchased from Ark Pharm; imidazole, 1H-imidazole-1,2-dicarboxylic acid, 1-(1,1-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,
  • Figure US20180291047A1-20181011-C00069
  • 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.; (1S,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)-1-(tert-butoxycarbonyl)-4-chloropyrrolidine-2-carboxylic acid is available from Arch Bioscience; (1S,3aR,6aS)-2-(tert-butoxycarbonyl)octahydrocyclopenta[c]pyrrole-1-carboxylic acid is available from Ark Pharm; 1,2-pyrrolidinedicarboxylic acid, 3-[[(phenylmethoxy)carbonyl]amino]-, 1-(1,1-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.
  • The compound
  • Figure US20180291047A1-20181011-C00070
  • can be prepared as disclosed by Braun, J. V.; Heymons, Albrecht Berichte der Deutschen Chemischen Gesellschaft [Abteilung] B: Abhandlungen (1930) 63B, 502-7.
  • The compounds (2S,3S,4S)-4-fluoro-3-methoxy-pyrrolidine-1,2-dicarboxylic acid 1-tert-butyl ester and (2R,3R,4R)-3-fluoro-4-methoxy-pyrrolidine-1,2-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 Alkynyl Synthons
  • Alkynyl synthons include but are not limited to:
  • Figure US20180291047A1-20181011-C00071
  • wherein
  • R30 is independently chosen at each occurrence from hydrogen, C1-C6alkyl, C1-C6haloalkyl, (aryl)C0-C4alkyl, (C3-C7cycloalkyl)C0-C4alkyl, (phenyl)C0-C4alkyl, (4- to 7-membered heterocycloalkyl)C0-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; (5- or 6-membered unsaturated or aromatic heterocycle)C0-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S; COOH, Si(CH3)3, COOR30a, C2-C6alkanoyl, —B(OH)2, —C(O)(CH2)1-4S(O)R21, —P(O)(OR21)(OR22), —P(O)(OR21)R22, —P(O)R21R22, —NR9P(O)(NHR21)(NHR22), —NR9P(O)(OR21)(NHR22), —NR9P(O)(OR21)(OR22), —C(S)R21, —NR21SO2R22, —NR9S(O)NR0R22, —NR9SO2NR10R22, —SO2NR9COR22, —SO2NR9CONR21R22, —NR21SO2R22, —C(O)NR21SO2R22, —C(NIH2)NR9R22, —C(NH2)NR9S(O)2R22, —NR9C(O)OR10, —NR21OC(O)R22, —(CH2)1-4C(O)NR21R22, —C(O)R24R25, —NR9C(O)R21, —C(O)R21, —NR9C(O)NR9R10, —NR9C(O)NR24R25, —(CH2)1-4OC(O)R21, each of which R30 can be optionally substituted;
  • R30a is C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, (C3-C7cycloalkyl)C0-C4alkyl-, (aryl)C0-C4alkyl-, (3- to 7-membered heterocycloalkyl)C0-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, each of which R30a can be optionally substituted.
  • Alkyne synthons can be prepared and coupled by a person skilled in the art. For example, Amashiro discloses alkynyl amines in WO 2008/047831. Aryl alkynes are disclosed in Tang, T. et al., J. Org. Chem., 2013, 78, 3170-3175. Alkynes can be synthesized from primary alcohols; see, Quesada, R. and Taylor, R. J. K., Tett. Lett., 2005, 46, 6473-6476. Alkynes can be synthesized from aldehydes using an in situ preparation of dimethyldiazomethylphosphonate; see Roth, G. J. et al., Synthesis, 2004, 59-62. Bromoalkenes can be converted to alkynes; see, Okutani, M. and Mori, Y., J. Org. Chem., 2009, 74, 442-444. (Z)-1-Bromoalkenes can be converted to alkynes; see, Kuang, C., et al., Tetrahedron, 2005, 61, 4043-4052.
  • Example 5. Preparation of A-C(O)— Moiety
  • Figure US20180291047A1-20181011-C00072
  • In Step 1 of Route 5a, the stating material is treated with an organic solvent, an acylating reagent and a catalyst to add the R6 substitutent. In one embodiment, the organic solvent is toluene. In one embodiment, the acylating reagent is acetyl chloride. In one embodiment, the catalyst is tin tetrachloride. In another embodiment, the starting material in Step 1 is treated with an inorganic cyanide and organometallic catalyst to generate a cyano group at the R6 position. The cyano group is treated with a oxime to generate an amide at the R6 position. In Step 2, the nitrogen of the heteroaryl ring is treated with a tert-butyl haloacetate, an organic solvent and a base. In one embodiment, the halo group, LG2, is bromine. In one embodiment, the organic solvent is acetonitrile. In one embodiment, the base is potassium carbonate. In Step 3, the tert-butyl ester is treated with an organic acid and an organic solvent to generate an acid. In one embodiment, the organic acid is trifluoroacetic acid. In one embodiment, the organic solvent is dichloromethane. This chemistry is illustrated in Route 5a.
  • Figure US20180291047A1-20181011-C00073
  • In Step 1 of Route 5b, the stating material is treated with an organic solvent, an acylating reagent and a catalyst to add the R6 substitutent. In one embodiment, the organic solvent is toluene. In one embodiment, the acylating reagent is acetyl chloride. In one embodiment, the catalyst is tin tetrachloride. In another embodiment, the starting material in Step 1 is treated with an inorganic cyanide and organometallic catalysts to add a cyano group at the R6 position. The cyano group is treated with an oxime to generate an amide at the R6 position. In Step 2, the nitrogen of the heteroaryl ring is treated with a tert-butyl haloacetate, an organic solvent and a base. In one embodiment, the halo group, LG2, is bromine. In one embodiment, the organic solvent is acetonitrile. In one embodiment, the base is potassium carbonate. In Step 3, the tert-butyl ester is treated with an organic acid and an organic solvent to generate a carboxylic acid. In one embodiment, the organic acid is trifluoroacetic acid. In one embodiment, the organic solvent is dichloromethane. This chemistry is illustrated in Route 5b.
  • Example 6: Coupling of Central-L-B Synthons to —C(O)— Moieties
  • Figure US20180291047A1-20181011-C00074
  • As illustrated in Scheme 6a, central-LB synthons and a carboxylic acid (C(O)-A) are combined and treated with an organic solvent, a base, and coupling agent. In one embodiment, the solvent is DMF. In one embodiment, the base is N,N-diisopropylethylamine. In one embodiment the coupling reagent is (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate) (HATU).
  • Figure US20180291047A1-20181011-C00075
  • As illustrated in Scheme 6b, the central-LB synthons and carboxylic acid (C(O)-A) are combined and treated with an organic solvent, a base, and coupling agent. In one embodiment, the solvent is DMF. In one embodiment, the base is N,N-diisopropylethylamine. In one embodiment the coupling reagent is (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate) (HATU).
  • Example 7. Synthesis of Alkynes of Formula I
  • Figure US20180291047A1-20181011-C00076
  • As an example, the proline derivative from Example 6 Scheme 6b is treated with two organometallic catalysts, trimethylsilylacetylene, a base and organic solvent at an elevated temperature to generate a trimethylsilyl protected alkyne. In one embodiment, the first organometallic catalyst is copper iodide. In one embodiment, the second organometallic catalyst is tetrakis(triphenylphosphine)palladium(0). In one embodiment, the base is triethylamine. In one embodiment, the organic solvent is DMF. In one embodiment, the reaction is deoxygenated by bubbling argon into the pressure vessel. In one embodiment, the reaction is treated at an elevated temperature. In one embodiment, the reaction is treated at about 80 to about 105° C. In Step 2, the trimethylsilyl protected akyne is treated with a fluoride reagent and an organic solvent. In one embodiment, the fluoride reagent is tetrabutylammonium fluoride. In one embodiment, the organic solvent is tetrahydrofuran. In Step 3, the alkyne is treated with LG-R30, two organometallic catalysts, a base, and an organic solvent at an elevated temperature. In one embodiment, the first organometallic catalyst is copper iodide. In one embodiment, the second organometallic catalyst is bis(triphenylphosphine)palladium(II) dichloride. In one embodiment, the base is triethylamine. In one embodiment, the organic solvent is DMF. In one embodiment, the elevated temperature is at about 80 to about 95° C. In one embodiment the LG R30 group is an heteroaryl halide. In one embodiment, the LG-R30 group is an aryl halide. This chemistry is illustrated in Scheme 7a.
  • In an alternate embodiment, the proline derivative from Example 6 Scheme 6b is treated with two organometallic catalysts, an alkyne, a base and organic solvent at an elevated temperature to generate an alkyne of Formula I. In one embodiment, the first organometallic catalyst is copper iodide. In one embodiment, the second organometallic catalyst is tetrakis(triphenylphosphine)palladium(0). In one embodiment, the base is triethylamine. In one embodiment, the organic solvent is DMF. In one embodiment, the reaction is deoxygenated by bubbling argon into the pressure vessel. In one embodiment, the reaction is treated at an elevated temperature. In one embodiment, the reaction is treated at about 80 to about 105° C.
  • Figure US20180291047A1-20181011-C00077
  • The proline derivative from Example 6 Scheme 6b is treated with two organometallic catalysts, trimethylsilylacetylene, a base and organic solvent at an elevated temperature to generate a trimethylsilyl protected alkyne. In one embodiment, the first organometallic catalyst is copper iodide. In one embodiment, the second organometallic catalyst is tetrakis(triphenylphosphine)palladium(0). In one embodiment, the base is triethylamine. In one embodiment, the organic solvent is DMF. In one embodiment, the reaction is deoxygenated by bubbling argon into the pressure vessel. In one embodiment, the reaction is treated at an elevated temperature. In one embodiment, the reaction is treated at about 80 to about 105° C. In Step 2, the trimethylsilyl protected akyne is treated with a fluoride reagent and an organic solvent. In one embodiment, the fluoride reagent is tetrabutylammonium fluoride. In one embodiment, the organic solvent is tetrahydrofuran. In Step 3, the alkyne is treated with LG-R30, two organometallic catalysts, a base, and an organic solvent at an elevated temperature. In one embodiment, the first organometallic catalyst is copper iodide. In one embodiment, the second organometallic catalyst is bis(triphenylphosphine)palladium(II) dichloride. In one embodiment, the base is triethylamine. In one embodiment, the organic solvent is DMF. In one embodiment, the elevated temperature is at about 80 to about 95° C. In one embodiment the LG R30 group is an heteroaryl halide. In one embodiment, the LG-R30 group is an aryl halide. This chemistry is illustrated in Scheme 7b.
  • In an alternate embodiment, the proline derivative from Example 6 Scheme 6b is treated with two organometallic catalysts, an alkyne, a base and organic solvent at an elevated temperature to generate an alkyne of Formula I. In one embodiment, the first organometallic catalyst is copper iodide. In one embodiment, the second organometallic catalyst is tetrakis(triphenylphosphine)palladium(0). In one embodiment, the base is triethylamine. In one embodiment, the organic solvent is DMF. In one embodiment, the reaction is deoxygenated by bubbling argon into the pressure vessel. In one embodiment, the reaction is treated at an elevated temperature. In one embodiment, the reaction is treated at about 80 to about 105° C.
  • In an alternative embodiment, a TMS alkyne from Example 7 Scheme 7a or 7b can be treated with an inorganic fluoride, an organic solvent and carbon dioxide to generate a carboxylic acid. In one embodiment, the inorganic fluoride is cesium fluoride. In one embodiment, the organic solvent is dimethyl sulfoxide. The resulting carboxylic acid can be treated with a sulfonamide anion to generate a compound of Formula I.
  • Example 8. Synthesis of 1A. (2S,4R)-tert-butyl 2-((3-chloro-2-fluoro-benzyl)carbamoyl)-4-fluoropyrrolidine-1-carboxylate
  • Figure US20180291047A1-20181011-C00078
  • (2S,4R)-1-(tert-butoxycarbonyl)-4-fluoropyrrolidine-2-carboxylic acid (2.33 gm, 10 mmol) was dissolved in DMF (50 ml) and iPr2NEt (8.6 ml, 5 eq.) was added, followed by the addition of (3-chloro-2-fluorophenyl) methanamine (3.18 gm 20 mmol) at 5° C. Then HATU (8 gm, 2.1 eq) was added slowly at same temperature. The reaction mixture was then stirred for 18 h at RT. After completion of the reaction monitored by HPLC, The reaction mixture was diluted with 1M citric acid solution (200 ml+NaCl solid 20 gm) and extracted with DCM (150 mL×2), the organic layer was then washed with an aqueous solution of NaHCO3 (100 ml) and washed with water (100 ml), brine (100 ml) and dried over Na2SO4 and concentrated under reduced pressure. The remaining residue was purified by column chromatography (eluted with DCM/EtOAc) to give (2S,4R)-tert-butyl 2-((3-chloro-2-fluorobenzyl)carbamoyl)-4-fluoropyrrolidine-1-carboxylate.
  • 1B. (2S,4R)—N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide hydrochloride (A)
  • Figure US20180291047A1-20181011-C00079
  • (2S,4R)-tert-butyl 2-((3-chloro-2-fluorobenzyl)carbamoyl)-4-fluoropyrrolidine-1-carboxylate (500 mg) was taken in 4N HCl dioxane (30 ml) and resulting reaction mixture was stirred at rt for 3 h. After completion of the reaction monitored by HPLC solvent was removed under reduced pressure. The residue, A, was used for next reaction.
  • Example 9. Synthesis of (2S,4R)-1-(2-(3-acetyl-5-(pyrimidin-2-ylethynyl)-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide
  • Figure US20180291047A1-20181011-C00080
    Figure US20180291047A1-20181011-C00081
  • 1-(5-Bromo-1H-indol-3-yl)ethanone (2) was prepared from 5-bromoindole according to the procedure published by MacKay et al. (MacKay, J. A.; Bishop, R.; Rawal, V. H. Org. Lett. 2005, 7, 3421-3424.)
  • A mixture of 3.9 g (16.4 mmol) of 1-(5-bromo-1H-indol-3-yl)ethanone, 2.63 mL (18.02 mmol) of tert-butyl bromoacetate and 2.50 g (18.02 mmol) potassium carbonate in anhydrous acetonitrile (80 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 1:1 mixture of CH2Cl2 and water (100 mL: 100 mL). The two layers were separated and the organic layer was washed with water (2×100 mL). Finally, the organic layer was dried (Na2SO4) and concentrated. The resulting residue was stirred with 50 mL of heptane for 30 min., cooled in an ice bath and filtered, washing the solid with cold heptane (10 mL). This cream colored solid was dried under high vacuum to give 5.6 g of product tert-Butyl 2-(3-acetyl-5-bromo-1H-indol-1-yl)acetate.
  • 4.5 g of tert-Butyl 2-(3-acetyl-5-bromo-1H-indol-1-yl)acetate was stirred in 1:1 mixture of CH2Cl2-TFA (90 mL) at RT for 4 h. The volatiles were removed under reduced pressure. The residue was dissolved in 45 mL of DMF. 4.16 g (13.4 mmol) of hydrochloride salt of 5 was added to this solution, followed by 11 mL of N,N-diisopropylethylamine (63.7 mmol). The reaction mixture was cooled in an ice bath and 5.82 g of (15.3 mmol) HATU was added. Following the addition of HATU, the cooling bath was removed and the reaction mixture was stirred overnight at RT. This reaction mixture was then poured in 450 mL of 1.0 M aq. citric acid solution. The separated product was isolated by filtration and the solid was washed thoroughly with water. This gray solid was dried under high vacuum to give 7.4 g of product (2S,4R)-1-(2-(3-Acetyl-5-bromo-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide.
  • A mixture of 1 g (1.8 mmol) of (2S,4R)-1-(2-(3-Acetyl-5-bromo-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide, 0.419 g (0.36 mmol) of tetrakis(triphenylphosphine)palladium(0), 0.138 g 0.72 mmol) of cuprous iodide in DMF (10 mL) was deoxygenated by bubbling argon in a pressure vessel. Triethylamine (2.53 mL, 18.1 mmol) and 2.56 mL (18.1 mmol) of ethynyltrimethylsilane were added under argon. The pressure vessel was capped and heated at 100° C. overnight. Then the reaction mixture was cooled to RT and the solvent was removed under reduced pressure. The residue was purified by column chromatography (silica gel, 0-2% MeOH in CH2Cl2) to give 0.56 g of (2S,4R)-1-(2-(3-Acetyl-5-((trimethylsilyl)ethynyl)-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide as a yellow solid.
  • To a solution of (2S,4R)-1-(2-(3-Acetyl-5-((trimethylsilyl)ethynyl)-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide (0.5 g, 1 mmol) in THF (5 mL) was added 1.5 mL of 1.0 M tetrabutylammonium fluoride in THF at RT. The reaction mixture was stirred for 1 h. The solvent was removed under reduced pressure and the residue was purified by column chromatography (silica gel, 0-2% MeOH in CH2Cl2) to give 0.26 g of (2S,4R)-1-(2-(3-acetyl-5-ethynyl-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide as a light yellow solid.
  • A mixture of 0.16 g (1.0 mmol) of 2-bromopyrimidine, 0.141 g (0.2 mmol) of PdCl2(PPh3)2, 0.043 g (0.23 mmol) of cuprous iodide in DMF (4 mL) was deoxygenated by bubbling argon in a pressure vessel. Triethylamine (2.0 mL) and 0.250 g (0.5 mmol) of (2S,4R)-1-(2-(3-Acetyl-5-ethynyl-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide were added under argon. The pressure vessel was capped and heated at 100° C. overnight. Then the reaction mixture was cooled to RT and the solvent was removed under reduced pressure. The residue was purified by column chromatography (silica gel, 0-4.5% MeOH in CH2Cl2) to give 35 mg of the desired product (2S,4R)-1-(2-(3-acetyl-5-(pyrimidin-2-ylethynyl)-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide as a light red solid. 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ: 1.99-2.18 (m, 1H), 2.46 (s, 3H), 3.92 (ddd, J=24.4, 12.4, 2.8 Hz, 1H), 4.26 (dd, J=12.8, 20.8 Hz, 1H), 4.32 (dd, J=28.4, 6.0 Hz, 1H), 4.39-4.49 (m, 2H), 5.24 (d, J=17.2 Hz, 1H), 5.45 (d, J=17.2 Hz, 1H), 5.51 (d, J=52.8 Hz, 1H), 6.96 (t, J=7.2 Hz, 1H), 7.23 (t, J=6.4 Hz, 1H), 7.40-7.51 (m, 4H), 7.59 (d, J=8.4 Hz, 1H), 8.36 (s, 1H), 8.47 (d, J=0.8 Hz, 1H), 8.61 (t, J=5.6 Hz, 1H), 8.85 (d, J=4.8 Hz, 2H). 31F NMR (376 MHz, DMSO-d6, 300 K): (major rotamer) δ: −121.7, −176.1. LC (method 1): tR=1.73 min. LC/MS (EI) m/z: [M+H]+ calcd for C30H24ClF2N5O3, 575.9; found, 576.4.
  • Example 10: Additional Syntheses of Non-Limiting Examples of Alkyne Compounds of Formula I Compound 2: 3-(3-Acetyl-1-(2-((2S,4R)-2-((3-chloro-2-fluorobenzyl)carbamoyl)-4-fluoropyrrolidin-1-yl)-2-oxoethyl)-1H-indol-5-yl)propiolic acid
  • Figure US20180291047A1-20181011-C00082
  • As shown in Scheme 1, (2S,4R)-1-(tert-butoxycarbonyl)-4-fluoropyrrolidine-2-carboxylic acid (2a, 2.33 g) was dissolved in DMF (50 ml) and DIEA (8.6 ml) was added, followed by the addition of (3-chloro-2-fluorophenyl) methanamine (2b, 3.18 gm) at 5° C. Then HATU (8 gm) was added slowly at same temperature. The reaction mixture was then stirred for 18 h at RT. After completion of the reaction monitored by HPLC, The reaction mixture was diluted with 1M citric acid solution (200 ml+NaCl solid 20 gm) and extracted with DCM (150 mL×2), the organic layer was then washed with an aqueous solution of NaHCO3 (100 ml) and washed with water (100 ml), brine (100 ml) and dried over Na2SO4 and concentrated under reduced pressure. The remaining residue was purified by column chromatography (eluent: DCM/EtOAc) to give (2S,4R)-tert-butyl 2-((3-chloro-2-fluorobenzyl)carbamoyl)-4-fluoropyrrolidine-1-carboxylate (2c).
  • (2S,4R)-tert-butyl 2-((3-chloro-2-fluorobenzyl)carbamoyl)-4-fluoropyrrolidine-1-carboxylate (500 mg) was taken in 4N HCl dioxane (30 ml) and resulting reaction mixture was stirred at RT for 3 h. After completion of the reaction, solvent was removed under reduced pressure. The residue, 2d, was used for next reaction.
  • Figure US20180291047A1-20181011-C00083
  • As shown in scheme 2, 1-(6-Bromo-1H-indol-3-yl)ethanone (2f) was prepared from 6-bromoindole (2e) according to the procedure published by MacKay et al. (MacKay, J. A.; Bishop, R.; Rawal, V. H. Org. Lett. 2005, 7, 3421-3424.)
  • A mixture of 0.5 g of 1-(6-bromo-1H-indol-3-yl)ethanone, 0.35 mL of tert-butyl bromoacetate and 0.318 g potassium carbonate in anhydrous acetonitrile (10 mL) was refluxed for 5 h. The reaction mixture was then cooled to RT and diluted with dichloromethane. K2CO3 was filtered off and the filtrate was concentrated under reduced pressure. The residue was purified by chromatography over silica gel (eluent: 0-25% EtOAc in hexanes) to get 0.7 g of white solid of tButyl 2-(3-acetyl-5-bromo-1H-indol-1-yl)acetate (2g).
  • Compound 2e (31.0 g) was taken in 4.0 N HCl in dioxane and stirred overnight at RT. Then 150 mL of additional 4.0 N HCl in dioxane was added and stirred for 6 h at RT. The volatiles were then removed under reduced pressure and the residue was dried under high vacuum to get 25.3 g of 2-(3-acetyl-6-bromo-1H-indol-1-yl)acetic acid (2h).
  • Compound 2h (25.3 g) was dissolved in 250 mL of DMF. 27.86 g of hydrochloride salt of 2d was added to this solution, followed by 83.82 mL of N,N-diisopropylethylamine. The reaction mixture was cooled in an ice bath and 38.98 g of HATU was added. Following the addition of HATU, the cooling bath was removed and the reaction mixture was stirred for 1 h at RT. This reaction mixture was then poured in 250 mL of 1.0 M aq. citric acid solution. The separated product was isolated by filtration and the solid was washed thoroughly with water. This cream solid was dried under high vacuum to give 48 g of product (2S,4R)-1-(2-(3-Acetyl-6-bromo-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide (2i). This solid was then stirred with 250 mL of EtOAc for 30 min at RT and filtered, washing the solid with 60 mL of EtOAc to get 45 g of the product 2i.
  • A mixture of 0.8 g of (2S,4R)-1-(2-(3-Acetyl-6-bromo-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide (2i), 0.669 g of Pd(PPh3)4, 0.124 g of cuprous iodide in DMF (8 mL) was deoxygenated by bubbling argon in a pressure vessel. Triethylamine (4 mL), and 2.6 mL of ethynyltrimethylsilane were added under argon. The pressure vessel was capped and heated at 100° C. overnight. Then the reaction mixture was cooled to RT and the solvent was removed under reduced pressure. The residue was purified by chromatography over silica gel (eluent: 0-2.5% MeOH in CH2Cl2) to give 0.68 g of (2S,4R)-1-(2-(3-Acetyl-6-((trimethylsilyl)ethynyl)-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide (2j) as orange yellow solid.
  • Synthesized following the procedure of Kobayashi et al (Kobayashi et al, Org. Biomol. Chem. 2013, 11, 3773-3775). A flask was charged with 96 mg of CsF in anhydrous DMSO (3 mL) under CO2 atmosphere. Then 0.3 g of 2j was added and stirred under CO2 atmosphere in a balloon overnight at RT. The reaction mixture was diluted with water (30 mL) and extracted with dichloromethane. The aq. layer was acidified with aq. 6N HCl at 0° C. and extracted with EtOAc. Then the EtOAc layer was dried (Na2SO4) and concentrated. The residue was purified by chromatography over silica gel (eluent: 0-3.2% MeOH in CH2Cl2) to get 75 mg of 3-(3-Acetyl-1-(2-((2S,4R)-2-((3-chloro-2-fluorobenzyl)carbamoyl)-4-fluoropyrrolidin-1-yl)-2-oxoethyl)-1H-indol-5-yl)propiolic acid (2) as cream colored solid. 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 2.05-2.18 (m, 1H), 2.45 (s, 3H), 2.50-2.58 (m, 1H), 3.87-4.08 (m, 1H), 4.11-4.16 (m, 1H), 4.23-4.29 (m, 1H), 4.35-4.49 (m, 2H), 5.25 (d, J=17.2 Hz, 1H), 5.45 (d, J=17.2 Hz, 1H), 5.52 (d, J=53.2 Hz, 1H), 6.90 (t, J=8 Hz, 1H), 7.19 (t, J=6.8 Hz, 1H), 7.33-7.55 (m, 2H), 7.91 (s, 1H), 8.23 (d, J=8.4 Hz, 1H), 8.39 (s, 1H), 8.61 (t, J=5.6 Hz, 1H), 13.70 (brs, 1H). 19F NMR (376 MHz, DMSO-d6, 300 K): (major rotamer) δ −121.8, −176.1. LC (method 1): tR=1.53 min. LC/MS (EI) m/z: [M+H]+ calcd for C27H22ClF2N3O5, 542; found, 542.
  • Compound 6: 3-(3-Acetyl-1-(2-((2S,4R)-2-((3-chloro-2-fluorobenzyl)carbamoyl)-4-fluoropyrrolidin-1-yl)-2-oxoethyl)-1H-indol-5-yl)propiolic acid
  • Figure US20180291047A1-20181011-C00084
  • Same procedure as for the synthesis of compound 2 was followed, except the starting material was 5-bromoindole 6a.
  • 1-(5-Bromo-1H-indol-3-yl)ethanone (6b) was prepared from 5-bromoindole according to the procedure published by MacKay et al. (MacKay, J. A.; Bishop, R.; Rawal, V. H. Org. Lett. 2005, 7, 3421-3424.)
  • A mixture of 3.9 g of 1-(5-bromo-1H-indol-3-yl)ethanone, 2.63 mL of tert-butyl bromoacetate and 2.50 g potassium carbonate in anhydrous acetonitrile (80 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 1:1 mixture of CH2Cl2 and water (100 mL: 100 mL). The two layers were separated and the organic layer was washed with water (2×100 mL). Finally, the organic layer was dried (Na2SO4) and concentrated. The resulting residue was stirred with 50 mL of heptane for 30 min., cooled in an ice bath and filtered, washing the solid with cold heptane (10 mL). This cream colored solid was dried under high vacuum to give 5.6 g of product tert-Butyl 2-(3-acetyl-5-bromo-1H-indol-1-yl)acetate (6c).
  • tert-Butyl 2-(3-acetyl-5-bromo-1H-indol-1-yl)acetate (6c, 4.5 g) was stirred in 1:1 mixture of CH2Cl2-TFA (90 mL) at RT for 4 h. The volatiles were removed under reduced pressure. The residue was dissolved in 45 mL of DMF. 4.16 g (13.4 mmol) of hydrochloride salt of 2d was added to this solution, followed by 11 mL of N,N-diisopropylethylamine (63.7 mmol). The reaction mixture was cooled in an ice bath and 5.82 g of (15.3 mmol) HATU was added. Following the addition of HATU, the cooling bath was removed and the reaction mixture was stirred overnight at RT. This reaction mixture was then poured in 450 mL of 1.0 M aq. citric acid solution. The separated product was isolated by filtration and the solid was washed thoroughly with water. This gray solid was dried under high vacuum to give 7.4 g of product (2S,4R)-1-(2-(3-Acetyl-5-bromo-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide (6e).
  • A mixture of 1 g of (2S,4R)-1-(2-(3-Acetyl-5-bromo-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide (6e), 0.419 g of tetrakis(triphenylphosphine)palladium(0), 0.138 g of cuprous iodide in DMF (10 mL) was deoxygenated by bubbling argon in a pressure vessel. Triethylamine (2.53 mL) and 2.56 mL of ethynyltrimethylsilane were added under argon. The pressure vessel was capped and heated at 100° C. overnight. Then the reaction mixture was cooled to RT and the solvent was removed under reduced pressure. The residue was purified by column chromatography over silica gel (eluent: 0-2% MeOH in CH2Cl2) to give 0.56 g of (2S,4R)-1-(2-(3-acetyl-5-((trimethylsilyl)ethynyl)-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide (6f) as a yellow solid.
  • Synthesized following the procedure of Kobayashi et al (Kobayashi et al, Org. Biomol. Chem. 2013, 11, 3773-3775) as described for 2. A flask was charged with 176 mg of CsF in anhydrous DMSO (5 mL) under CO2 atmosphere. Then 0.55 g of 6f was added and stirred under CO2 atmosphere in a balloon overnight at RT. The reaction mixture was diluted with water (30 mL) and extracted with dichloromethane. The aq. layer was acidified with aq. 6N HCl at 0° C. and extracted with EtOAc. Then the EtOAc layer was dried (Na2SO4) and concentrated. The residue was washed with dichloromethane to get 130 mg of 3-(3-Acetyl-1-(2-((2S,4R)-2-((3-chloro-2-fluorobenzyl)carbamoyl)-4-fluoropyrrolidin-1-yl)-2-oxoethyl)-1H-indol-5-yl)propiolic acid (6) as light orange solid. 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 2.05-2.18 (m, 1H), 2.45 (s, 3H), 2.49-2.58 (m, 1H), 3.84-3.97 (m, 1H), 4.09-4.18 (m, 1H), 4.23-4.28 (m, 1H), 4.34-4.49 (m, 2H), 5.23 (d, J=17.2 Hz, 1H), 5.43 (d, J=17.2 Hz, 1H), 5.50 (d, J=52.4 Hz, 1H), 6.96 (t, J=7.6 Hz, 1H), 7.22 (t, J=6.4 Hz, 1H), 7.42-7.44 (m, 2H), 7.57 (d, J=8.4 Hz, 1H), 8.36 (s, 1H), 8.44 (d, J=1.2 Hz, 1H), 8.59 (t, J=6 Hz, 1H), 13.58 (brs, 1H). 19F NMR (376 MHz, DMSO-d6, 300 K): (major rotamer) δ −121.7, −176.1. LC (method 1): tR=1.26 min. LC/MS (EI) m/z: [M+H]+ calcd for C27H22ClF2N3O5, 542; found, 542.
  • Compound 8: (2S,4R)-1-(2-(3-acetyl-5-(3-(ethylsulfonamido)-3-oxoprop-1-yn-1-yl)-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide
  • Figure US20180291047A1-20181011-C00085
  • 1,1′-Carbonyldiimidazole (20 mg) was added with stirring to a heterogeneous solution of 6 (50 mg) in THF (4 mL) and stirred overnight at room temperature.
  • In another round bottom flask, NaH (15 mg) was added portion-wise to an ice cooled solution of ethanesulfonamide (50 mg) in THF (2 mL). After the addition of NaH was complete, the cooling bath was removed and the reaction mixture was stirred at RT for 1 h. This sulfonamide anion was then added to the flask containing the activated ester of 6 drop-wise. The resulting reaction mixture was stirred overnight at RT and the volatiles were removed under reduced pressure. The residue was purified by chromatography over silica gel (eluent: 0-8.5% MeOH in dichloromethane). 10 mg of 8 was obtained as a light yellow solid.
  • 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 1.18-1.28 (m, 5H), 1.95-2.17 (m, 1H), 2.45 (s, 3H), 2.42-2.48 (m, 1H), 3.87-3.99 (m, 1H), 4.09-4.18 (m, 1H), 4.26 (dd, J=10, 5.6 Hz, 1H), 4.34-4.51 (m, 2H), 5.22 (d, J=17.2 Hz, 1H), 5.43 (d, J=17.2 Hz, 1H), 5.51 (d, J=52.4 Hz, 1H), 6.97 (t, J=4.8 Hz, 1H), 7.22 (t, J=7.2 Hz, 1H), 7.39-7.48 (m, 2H), 7.55 (d, J=8 Hz, 1H), 8.35 (s, 1H), 8.45 (brs, 1H), 8.59 (t, J=6 Hz, 1H). 19F NMR (376 MHz, DMSO-d6, 300 K): (major rotamer) δ −121.7, −176.1. LC (method 1): tR=1.73 min. LC/MS (EI) m/z: [M+H]+ calcd for C29H72ClF2N406S, 633; found 633.
  • Compound 9: (2S,4R)-1-(2-(3-Acetyl-5-(pyrimidin-2-ylethynyl)-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide
  • Figure US20180291047A1-20181011-C00086
  • To a solution of (2S,4R)-1-(2-(3-Acetyl-5-((trimethylsilyl)ethynyl)-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide (6f, synthesized as described for compound 6, in scheme 3) (0.5 g) in THF (5 mL) was added 1.5 mL of 1.0 M tetrabutylammonium fluoride in THF at RT. The reaction mixture was stirred for 1 h. The solvent was removed under reduced pressure and the residue was purified by column chromatography over silica gel (eluent: 0-2% MeOH in CH2Cl2) to give 0.26 g of (2S, 4R)-1-(2-(3-acetyl-5-ethynyl-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide (9a) as a light yellow solid.
  • A mixture of 0.16 g of 2-bromopyrimidine, 0.141 g of PdCl2(PPh3)2, 0.043 g of cuprous iodide in DMF (4 mL) was deoxygenated by bubbling argon in a pressure vessel. Triethylamine (2.0 mL) and 0.250 g of (2S,4R)-1-(2-(3-Acetyl-5-ethynyl-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide (9a) were added under argon. The pressure vessel was capped and heated at 90° C. overnight. Then the reaction mixture was cooled to RT and the solvent was removed under reduced pressure. The residue was purified by column chromatography over silica gel (eluent: 0-4.5% MeOH in CH2Cl2) to give 35 mg of the desired product (2S,4R)-1-(2-(3-Acetyl-5-(pyrimidin-2-ylethynyl)-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide (9) as a light orange solid. 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) 31.99-2.18 (m, 1H), 2.46 (s, 3H), 3.92 (ddd, J=24.4, 12.4, 2.8 Hz, 1H), 4.26 (dd, J=12.8, 20.8 Hz, 1H), 4.32 (dd, J=28.4, 6.0 Hz, 1H), 4.39-4.49 (m, 2H), 5.24 (d, J=17.2 Hz, 1H), 5.45 (d, J=17.2 Hz, 1H), 5.51 (d, J=52.8 Hz, 1H), 6.96 (t, J=7.2 Hz, 1H), 7.23 (t, J=6.4 Hz, 1H), 7.40-7.51 (m, 4H), 7.59 (d, J=8.4 Hz, 1H), 8.36 (s, 1H), 8.47 (d, J=0.8 Hz, 1H), 8.61 (t, J=5.6 Hz, 1H), 8.85 (d, J=4.8 Hz, 2H). 31F NMR (376 MHz, DMSO-d6, 300 K): (major rotamer) 3-121.7, −176.1. LC (method 1): tR=1.74 min. LC/MS (EI) m/z: [M+H]+ calcd for C30H24ClF2N5O3, 576; found, 576.
  • Compound 10: (2S,4R)-1-(2-(3-acetyl-6-(3-(ethylsulfonamido)-3-oxoprop-1-yn-1-yl)-1H-indol-1-yl)acetyl)-N-(3-chloro-2-fluorobenzyl)-4-fluoropyrrolidine-2-carboxamide
  • Figure US20180291047A1-20181011-C00087
  • Compound 10 was synthesized using the procedure similar to the synthesis of compound 8. Thus, 100 mg of 2 in DMF was coupled with 93 mg of ethanesulfonamide using 45 mg of 1,1′-carbonyldiimidazole and 37 mg of NaH. The reaction mixture was poured into 50 mL of water and acidified with 2N aq. HCl. The separated solid was isolated by filtration, washed successively with water and tert-butylmethyl ether. The solid was dried under high vacuum to yield 11 mg of 10 as brown solid.
  • 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 1.11-1.22 (m, 5H), 2.01-2.19 (m, 1H), 2.45 (s, 3H), 2.48-2.51 (m, 1H), 3.86-3.98 (m, 1H), 4.09-4.17 (m, 1H), 4.26 (dd, J=10, 6 Hz, 1H), 4.34-4.49 (m, 2H), 5.25 (d, J=17.2 Hz, 1H), 5.44 (d, J=17.2 Hz, 1H), 5.51 (d, J=53.2 Hz, 1H), 6.95 (t, J=8 Hz, 1H), 7.21 (t, J=6 Hz, 1H), 7.38-7.43 (m, 2H), 7.91 (s, 1H), 8.25 (d, J=8.4 Hz, 1H), 8.39 (s, 1H), 8.61 (t, J=5.6 Hz, 1H). 19F NMR (376 MHz, DMSO-d6, 300 K): (major rotamer) δ −121.8, −176.0. LC (method 1): tR=1.73 min. LC/MS (EI) m/z: [M+H]+ calcd for C29H27ClF2N4O6S, 633; found 633.
  • Compound 22: 1-(2-((2S,4R)-2-((2′-chloro-2-fluoro-[1,1′-biphenyl]-3-yl)carbamoyl)-4-fluoropyrrolidin-1-yl)-2-oxoethyl)-5-(pyrimidin-2-ylethynyl)-1H-indazole-3-carboxamide
  • Figure US20180291047A1-20181011-C00088
  • Compound 22b was prepared using the procedure similar to the synthesis of 6c. Thus, 23.5 g of 22a was coupled with 17.3 mL of tert-butyl bromoacetate using 32.5 g of potassium carbonate in 470 mL of acetonitrile at 90° C. for 3 h. Then the solvent was removed and water was added to the residue. The product was isolated by filtration, washed with water and dried. Finally, the product was washed with 2:1 mixture of tert-butylmethyl ether and heptane to get 29 g of 22b.
  • Compound 22c was prepared following similar procedure to the synthesis of 6f. 7.0 g of 22b was coupled with 28 mL of trimethylsilylacetylene using 4.69 g of Pd(PPh3)4, 1.5 g of cuprous iodide and 28 mL of triethylamine in 70 mL of DMF at 100° C. overnight. After removal of the solvent, the crude product was purified by column chromatography over silica gel (eluent: 0-1% MeOH in dichloromethane) to get 15 g of 22c.
  • Compound 22d was synthesized following the same procedure as for the synthesis of 9a. So, 1.6 g of 22c was deprotected using 3.0 mL of 1.0 M TBAF in THF at 0° C. for 15 min. Crude product was purified by chromatography over silica gel (eluent: 0-2.5% MeOH in dichloromethane) to get 1.09 g of 22d as cream-colored solid.
  • A mixture of 0.31 g of 2-bromopyrimidine, 0.282 g of PdCl2(PPh3)2, 0.086 g of cuprous iodide in DMF (4 mL) was deoxygenated by bubbling argon in a pressure vessel. Triethylamine (1.5 mL) and 0.3 g of 22d were added under argon. The pressure vessel was capped and heated at 90° C. overnight. Then the reaction mixture was cooled to RT and the solvent was removed under reduced pressure. The residue was purified by column chromatography over silica gel (eluent: 0-2% MeOH in CH2Cl2) to give 0.15 g of the desired product tert-butyl 2-(3-carbamoyl-5-(pyrimidin-2-ylethynyl)-1H-indazol-1-yl)acetate (22e) light orange solid.
  • A solution of 1.81 g of 22e in CH2Cl2 (3 mL) and TFA (3 mL) was stirred for 2 h at RT. Then the volatiles were removed under reduced pressure. The residue 2-(3-Carbamoyl-5-(pyrimidin-2-ylethynyl)-1H-indazol-1-yl)acetic acid, 22f, was used as such for the next step.
  • Figure US20180291047A1-20181011-C00089
  • 3-Bromo-2-fluoroaniline (22g, 30 g) and 2-chlorophenylboronic acid, 22h (61.75 g), Pd(dppf)Cl2 (19.25 g) and K2CO3 (91 g) were taken in a round bottom flask kept under argon. 400 mL of dioxane and 100 mL of water were added to this mixture. The mixture was bubbled with argon for 5 min and heated with stirring at 100° C. overnight. Then the reaction mixture was filtered through a Celite® pad and the filtrate was concentrated under reduced pressure. The residue was purified by chromatography over silica gel (eluent: 0-0.5% MeOH in CH2Cl2) to get a white oil. 300 mL of saturated HCl in methanol was added to this oil and the solvent was evaporated to get 30 g of the hydrochloride salt of 22i as white solid.
  • To an ice cold solution of (2S,4R)-1-(tert-butoxycarbonyl)-4-fluoropyrrolidine-2-carboxylic acid (2a, 24.64 g) in 600 mL of CH2Cl2, Ghosez's reagent (15.4 mL) was added drop-wise with stirring. The stirring was continued for 3 h. at same temperature. Then solid hydrochloride salt of 22i (30 g) was added, followed by 55.2 mL of DIEA (3 equiv.). The cooling bath was removed and the reaction mixture was stirred overnight at RT. The solvent was co-evaporated with 30 mL of MeOH. The crude product was purified by chromatography over silica gel (eluent: 10-50% CH2Cl2 in hexanes, then dichloromethane) to get 37 g of 22j as white solid.
  • A solution of 35.6 g of compound 22j in 400 mL of 4.0 N HCl in dioxane was stirred at RT for 4 h. The thick heterogeneous mixture was then concentrated under reduced pressure. The residue was sonicated with 350 mL of dichloromethane and filtered and the solid washed with 250 mL of dichloromethane. The isolated solid was dried under high vacuum to get 29.2 g of 22k as white solid.
  • Compound 22f from above and 1.69 g of 22k were taken in 20 mL of DMF. 4 mL of DIEA was added to this reaction mixture and cooled in an ice bath. HATU (2.09 g) was added to this cooled solution. After the addition of HATU, the cooling bath was removed and the reaction mixture was stirred at RT for 1 h. DMF was removed under reduced pressure and the residue was purified by chromatography over silica gel (eluent: 0-2.5% MeOH in dichloromethane) to get 2.46 g of 1-(2-((2S,4R)-2-((2′-chloro-2-fluoro-[1,1′-biphenyl]-3-yl)carbamoyl)-4-fluoropyrrolidin-1-yl)-2-oxoethyl)-5-(pyrimidin-2-ylethynyl)-1H-indazole-3-carboxamide (22).
  • 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 2.08-2.29 (m, 1H), 2.55-2.63 (m, 1H), 3.90-4.03 (m, 1H), 4.24 (dd, J=21.6, 12 Hz, 1H), 4.78 (t, J=8.4 Hz, 1H), 5.56 (d, J=52.8 Hz, 1H), 5.53 (d, J=17.2 Hz, 1H), 5.74 (d, J=17.2 Hz, 1H), 7.07 (t, J=8 Hz, 1H), 7.23 (t, J=8 Hz, 1H), 7.37-7.54 (m, 5H), 7.58-7.61 (m, 1H), 7.65 (dd, J=8.8, 1.6 Hz, 1H), 7.77 (dd, J=8.8, 0.4 Hz, 1H), 7.79 (brs, 1H), 7.97 (t, J=6.8 Hz, 1H), 8.48 (d, J=2.4 Hz, 1H), 8.87 (d, J=4.8 Hz, 2H), 9.99 (s, 1H). 19F NMR (376 MHz, DMSO-d6, 300 K): (major rotamer) δ −126.6,-175.8. LC (method 1): tR=1.99 min. LC/MS (EI) m/z: [M+H]+ calcd for C33H24ClF2N7O3, 640; found 640.
  • Compound 27: 1-(2-((1R,3S,5R)-3-((2′-Chloro-2-fluoro-[1,1′-biphenyl]-3-yl)carbamoyl)-2-azabicyclo[3.1.0]hexan-2-yl)-2-oxoethyl)-5-(pyrimidin-2-ylethynyl)-1H-indazole-3-carboxamide
  • Figure US20180291047A1-20181011-C00090
  • (1R,3S,5R)-2-(tert-butoxycarbonyl)-2-azabicyclo[3.1.0]hexane-3-carboxylic acid (27a) was coupled with 22i using Ghosez's reagent as described in synthesis of 22j (scheme 8). Thus, 0.2 g of 27a was coupled with 0.249 g of hydrochloride salt of 22i using 0.128 mL of Ghosez's reagent to get 0.3 g of 27b as colorless solid.
  • A solution of 0.114 g of 27b in CH2Cl2 (1.0 mL) and TFA (1.0 mL) was stirred for 30 min. Then the volatiles were removed under reduced pressure. The residue, 27c, was used as such for the next step.
  • Figure US20180291047A1-20181011-C00091
  • Compound 22e (0.05 g) was deprotected using 1 mL of TFA and 1 mL of CH2Cl2 to get 22f as described in scheme 7. This crude 22f was then coupled with 27c following the procedure similar to the synthesis of 22 using 0.121 g of HATU and 0.231 mL of DIEA in 1.5 mL of DMF to get 44 mg of 1-(2-((1R,3S,5R)-3-((2′-Chloro-2-fluoro-[1,1′-biphenyl]-3-yl)carbamoyl)-2-azabicyclo[3.1.0]hexan-2-yl)-2-oxoethyl)-5-(pyrimidin-2-ylethynyl)-1H-indazole-3-carboxamide (27) as white solid.
  • 1H NMR (400 MHz, CD3OD, 300 K): (major rotamer) δ 0.649 (br, 1H), 1.09-1.12 (m, 1H), 2.02-2.07 (m, 1H), 2.41-2.58 (m, 2H), 3.69-3.77 (m, 1H), 4.65 (dd, J=8.8, 4.8 Hz, 1H), 5.67 (d, J=17.2 Hz, 1H), 5.78 (d, J=17.2 Hz, 1H), 7.09 (t, J=7.2 Hz, 1H), 7.22 (t, J=8 Hz, 1H), 7.30-7.36 (m, 3H), 7.41 (t, J=5.2 Hz, 1H), 7.47-7.49 (m, 1H), 7.69 (dd, J=18, 8.8 Hz, 2H), 7.97 (t, J=7.6 Hz, 1H), 8.57 (s, 1H), 8.84 (d, J=5.2 Hz, 2H). 19F NMR (376 MHz, CD3OD, 300 K): (major rotamer) δ −128.8. LC (method 1): tR=2.10 min. LC/MS (EI) m/z: [M+H]+ calcd for C34H25ClFN7O3, 634; found 634.
  • Compound 28: 1-(2-((2S,4R)-2-((3-(3-chloropyridin-2-yl)-2-fluorophenyl)carbamoyl)-4-fluoropyrrolidin-1-yl)-2-oxoethyl)-5-(pyrimidin-2-ylethynyl)-1H-indazole-3-carboxamide: Scheme 11
  • Figure US20180291047A1-20181011-C00092
  • A mixture of 2.5 g of nitrobromide (28a), 7.21 g of bis(pinacolato)diboron, 1.24 g of PdCl2(dppf), 3.35 g of potassium acetate in dioxane (25 mL) was deoxygenated by bubbling argon in a pressure vessel. The pressure vessel was capped and heated at 90° C. overnight. Then the reaction mixture was cooled to RT and the solvent was removed under reduced pressure. The residue was diluted with dichloromethane and water. The organic layer was dried (Na2SO4) and concentrated. The crude product was purified by chromatography over silica gel (eluent: 20% EtOAc in hexanes) to get 2.94 g of 28c.
  • Same procedure as for the synthesis of 22i was followed, except that the reaction mixture was heated at 110° C. for 2 days. Thus, 2.8 g of 28c was coupled with 2.02 g of 2-bromo-2-chloropyridine using 85 mg of Pd(dppf)Cl2 and 3.63 g of potassium carbonate in dioxane-water. Purification of crude product by chromatography over silica gel gave 2.25g of 28d.
  • Compound 28d (2.25 g) was taken in 1:1 mixture of MeOH-4 N aq. HCl (60 mL) and 5.8 g of Zn powder was added to this reaction mixture. This heterogeneous reaction mixture was stirred overnight at RT. Finally, the reaction mixture was carefully neutralized with sat. aq. NaHCO3, the volatiles were removed under reduced pressure and the residue was extracted with EtOAc. Then the organic layer was dried (Na2SO4) and concentrated. The crude product was purified by chromatography over silica gel and the product was dissolved in methanolic HCl and taken to dryness to get 2.2 g of 28e.
  • (2S,4R)-1-(tert-butoxycarbonyl)-4-fluoropyrrolidine-2-carboxylic acid (2a) was coupled with amine 28e using Ghosez's reagent following procedure described for synthesis of 22j (scheme 8). Thus, 0.249 g of compound 2a was coupled with 0.292 g of amine 28e using 0.145 mL of Ghosez's reagent, after chromatographic purification, to get 0.286 g of 28f.
  • Compound 28f (0.286 g) was deprotected using 3 mL of TFA and 3 mL of dichloromethane to get 28g. Similarly, 0.240 g of 22e was hydrolyzed using 3 mL of TFA and 3 mL of dichloromethane to get crude 22f. Finally 22f was coupled with 28g as described for the synthesis of 22 using 0.298 g of HATU and 1 mL of DIEA in 5 mL of DMF. After the removal of DMF, the residue was purified by chromatography over silica gel (eluent: 0-3% MeOH in dichloromethane) to get 200 mg of 1-(2-((2S,4R)-2-((3-(3-chloropyridin-2-yl)-2-fluorophenyl)carbamoyl)-4-fluoropyrrolidin-1-yl)-2-oxoethyl)-5-(pyrimidin-2-ylethynyl)-1H-indazole-3-carboxamide (28).
  • 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 2.12-2.29 (m, 1H), 2.58-2.63 (m, 1H), 3.90-4.02 (m, 1H), 4.23 (dd, J=21.6, 12.4 Hz, 1H), 4.76 (t, J=8 Hz, 1H), 5.53 (d, J=17.6 Hz, 1H), 5.55 (d, J=52.8 Hz, 1H), 5.73 (d, J=17.6 Hz, 1H), 7.18 (t, J=6.8 Hz, 1H), 7.25 (t, J=8 Hz, 1H), 7.49-7.53 (m, 3H), 7.63 (d, J=8.8 Hz, 1H), 7.77 (d, J=8.8 Hz, 1H), 7.78 (brs, 1H), 8.08 (d, J=8 Hz, 1H), 8.25 (brs, 1H), 8.46 (s, 1H), 8.64 (d, J=4.8 Hz, 1H), 8.86 (d, J=4.8 Hz, 2H), 10.01 (s, 1H). 19F NMR (376 MHz, DMSO-d6, 300 K): (major rotamer) δ −126.9, −175.9. LC (method 1): tR=1.54 min. LC/MS (EI) m/z: [M+H]+ calcd for C32H23ClF2N8O3, 641; found 641.
  • Compound 39: 1-(2-((2S,4R)-2-((6-bromopyridin-2-yl)carbamoyl)-4-fluoropyrrolidin-1-yl)-2-oxoethyl)-5-(pyrimidin-2-ylethynyl)-1H-indazole-3-carboxamide
  • Figure US20180291047A1-20181011-C00093
  • 2-Amino-6-bromopyridine (0.191 g) was coupled with 0.23 g of trans-fluoroproline 2a using 0.146 mL of Ghosez's reagent in presence of 0.522 ml of DIEA following the procedure used for the synthesis of 22j. Crude product was purified by chromatography over silica gel (eluent: 0-0.1% MeOH in dichloromethane) to get 0.2 g of 39a as white solid.
  • 0.05 g of 22e and 65 mg of 39a were deprotected following procedure described for compound 22 (schemes 7 and 8). Coupling was done using the same procedure that was used for the synthesis of 22. Crude product was purified by chromatography over silica gel (eluent: 0-4% MeOH in dichloromethane) gave 50 mg of 1-(2-((2S,4R)-2-((6-bromopyridin-2-yl)carbamoyl)-4-fluoropyrrolidin-1-yl)-2-oxoethyl)-5-(pyrimidin-2-ylethynyl)-1H-indazole-3-carboxamide (39) as cream colored solid.
  • 1H NMR (400 MHz, CD3OD, 300 K): (major rotamer) δ 2.16-2.37 (m, 1H), 2.68-2.73 (m, 1H), 3.95-4.09 (m, 1H), 4.17-4.30 (m, 1H), 4.76 (t, J=8.4 Hz, 1H), 5.49 (d, J=52 Hz, 1H), 5.49 (d, J=17.2 Hz, 1H), 5.64 (d, J=17.2 Hz, 1H), 7.27 (d, J=7.6 Hz, 1H), 7.47 (t, J=4.8 Hz, 1H), 7.61 (t, J=8 Hz, 1H), 7.71 (s, 2H), 8.08 (d, J=8 Hz, 1H), 8.59 (s, 1H), 8.82 (d, J=4.8 Hz, 1H). 19F NMR (376 MHz, CD3OD, 300 K): (major rotamer) δ −178.6. LC (method 1): tR=1.48 min. LC/MS (EI) m/z: [M+]+ calcd for C26H20BrFN8O3, 591; found 591.
  • Compound 41: 1-(2-((1R,3S,5R)-3-((6-bromopyridin-2-yl)carbamoyl)-2-azabicyclo[3.1.0]hexan-2-yl)-2-oxoethyl)-5-(pyrimidin-2-ylethynyl)-1H-indazole-3-carboxamide
  • Figure US20180291047A1-20181011-C00094
  • 1.5 g of 27a was coupled with 1.26 g of 2-amino-6-bromopyridine using 961 μL of Ghosez's reagent as described for the synthesis of 22j. Crude product was purified by chromatography over silica gel (eluent: 0-0.1% MeOH in dichloromethane) to get 1.5 g of 41a as a white solid.
  • Compound 22e (0.05 g) and 56 mg of 41a were deprotected as described for compound 22 (scheme 7& 8). Coupling was done using the same procedure that was used for the synthesis of 22. Crude product was purified by chromatography over silica gel (eluent: 0-3% MeOH in dichloromethane) to get 50 mg of 1-(2-((1R,3S,5R)-3-((6-bromopyridin-2-yl)carbamoyl)-2-azabicyclo[3.1.0]hexan-2-yl)-2-oxoethyl)-5-(pyrimidin-2-ylethynyl)-1H-indazole-3-carboxamide (41) as white solid. 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 0.76 (brs, 1H), 0.99-1.05 (m, 1H), 1.86-1.92 (m, 1H), 2.07-2.29 (m, 2H), 3.80 (br, 1H), 4.45 (dd, J=8.8, 5.2 Hz, 1H), 5.54 (d, J=17.2 Hz, 1H), 5.87 (d, J=17.2 Hz, 1H), 7.32 (d, J=8 Hz, 1H), 7.50-7.53 (m, 2H), 7.68 (d, J=9.2 Hz, 1H), 7.72 (d, J=7.6 Hz, 1H), 7.79 (d, J=8.8 Hz, 2H), 8.02 (d, J=8 Hz, 1H), 8.46 (s, 1H), 8.85 (d, J=4.8 Hz, 2H), 10.76 (s, 1H). LC (method 1): tR=1.60 min. LC/MS (EI) m/z: [M+]+ calcd for C27H21BrFN8O3, 585; found 585.
  • Compound 46: 1-(2-((2S,4R)-2-((6-bromopyridin-2-yl)carbamoyl)-4-fluoropyrrolidin-1-yl)-2-oxoethyl)-5-((5-fluoropyrimidin-2-yl)ethynyl)-1H-indazole-3-carboxamide
  • Figure US20180291047A1-20181011-C00095
  • Procedure similar to the synthesis of 22e was followed. Thus, 0.1 g of 22d was coupled with 0.592 g of bromopyrimidine 46a in presence of 77 mg of Pd(PPh3)4, 25 mg of CuI and 0.245 mL of TEA in 2 mL of DMF at 90° C. Crude product was purified by chromatography over silica gel (eluent: 0-2% MeOH in dichloromethane) to get 0.13 g of 46b as cream colored solid.
  • Compound 46b (31 g) and 37 mg of 39a were deprotected as described for compound 22 (scheme 7&8). Coupling was done using the same procedure that was used for the synthesis of 22. Crude product was purified by chromatography over silica gel (eluent: 0-2.5% MeOH in dichloromethane) to get 36 mg of 1-(2-((2S,4R)-2-((6-bromopyridin-2-yl)carbamoyl)-4-fluoropyrrolidin-1-yl)-2-oxoethyl)-5-((5-fluoropyrimidin-2-yl)ethynyl)-1H-indazole-3-carboxamide (46) as white solid.
  • 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 2.03-2.23 (m, 1H), 2.52-2.56 (m 1H), 3.88-4.03 (m, 1H), 4.21 (dd, J=22.4, 12.8 Hz, 1H), 4.66 (t, J=8 Hz, 1H), 5.53 (d, J=52.8 Hz, 1H), 5.52 (d, J=17.2 Hz, 1H), 5.73 (d, J=17.2 Hz, 1H), 7.33 (d, J=7.6 Hz, 1H), 7.53 (s, 1H), 7.66-7.69 (m, 1H), 7.72 (d, J=8 Hz, 1H), 7.78 (d, J=8 Hz, 1H), 7.84 (s, 1H), 8.02 (d, J=8 Hz, 1H), 8.45 (s, 1H), 8.96 (d, J=0.8 Hz, 2H), 11.02 (s, 1H). 19F NMR (376 MHz, DMSO-d6, 300 K): (major rotamer) δ −135.7, −175.7. LC (method 1): tR=1.69 min. LC/MS (EI) m/z: [M+H]+ calcd for C26H19BrF2N6O3, 609; found 609.
  • Compound 49: 1-(2-((2S,4R)-2-((2′-chloro-2,4′-difluoro-[1,1′-biphenyl]-3-yl)carbamoyl)-4-fluoropyrrolidin-1-yl)-2-oxoethyl)-5-(pyrimidin-2-ylethynyl)-1H-indazole-3-carboxamide
  • Figure US20180291047A1-20181011-C00096
  • Same procedure as for the synthesis of 2c in scheme 1 was followed. Thus, 1.0 g of 3-bromo-2-fluoroaniline was coupled with 1.35 g of trans-fluoroproline 2a in presence of 2.4 g of HATU in DMF to get 0.695 g of 49a.
  • Prepared following the procedure for the synthesis of 22i (scheme 8). Thus, 0.3 g of 49a was coupled with 0.387 g of boronic acid 49b using 0.108 g of Pd(dppf)Cl2 and 0.512 g of K2CO3 in dioxane-water to get 280 mg of 49c.
  • 0.150 g of 49c and 0.137 g of 22e were deprotected as described in synthesis of compound 22 (schemes 7, 8). Coupling was done using the same procedure that was used for the synthesis of 22 to get 1-(2-((2S,4R)-2-((2′-Chloro-2,4′-difluoro-[1,1′-biphenyl]-3-yl)carbamoyl)-4-fluoropyrrolidin-1-yl)-2-oxoethyl)-5-(pyrimidin-2-ylethynyl)-1H-indazole-3-carboxamide (49).
  • 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 2.11-2.28 (m, 1H), 2.53-2.60 (m, 1H), 3.89-4.02 (m, 1H), 4.23 (dd, J=21.6, 12.8 Hz, 1H), 4.77 (t, J=8.4 Hz, 1H), 5.55 (d, J=52.4 Hz, 1H), 5.52 (d, J=17.2 Hz, 1H), 5.73 (d, J=17.2 Hz, 1H), 7.06 (t, J=7.2 Hz, 1H), 7.22 (t, J=8 Hz, 1H), 7.42-7.46 (m, 1H), 7.52 (t, J=4.8 Hz, 1H), 7.57-7.66 (m, 4H), 7.76 (d, J=9.2 Hz, 1H), 7.78 (s, 1H), 7.97 (t, J=7.6 Hz, 1H), 8.46 (s, 1H), 8.86 (d, J=4.8 Hz, 2H), 9.99 (s, 1H). 19F NMR (376 MHz, DMSO-d6, 300 K): (major rotamer) δ −111.6, −126.7, −175.9. LC (method 1): tR=2.04 min. LC/MS (EI) m/z: [M+H]+ calcd for C33H23ClF3N7O3, 658; found 658.
  • Compound 64: 1-(2-((1R,3S,5R)-3-((2′-chloro-2,4′-difluoro-[1,1′-biphenyl]-3-yl)carbamoyl)-2-azabicyclo[3.1.0]hexan-2-yl)-2-oxoethyl)-5-((5-fluoropyrimidin-2-yl)ethynyl)-1H-indazole-3-carboxamide
  • Figure US20180291047A1-20181011-C00097
  • The acid 27a was coupled with 3-bromo-2fluoroaniline using Ghosez's reagent as described for the synthesis of 22j.
  • Same procedure as for the synthesis of 49c was followed. Thus 0.210 g of 64a was coupled with 0.275 g of 49b in presence of 77 mg of Pd(dppf)Cl2 to get 0.184 g of 64b.
  • Compound 64b (0.136 g) and 0.100 g of 46b were deprotected as described in synthesis of compound 22 (scheme 7&8). Coupling was done using the same procedure that was used for the synthesis of 22 to give 1-(2-((1R,3S,5R)-3-((2′-Chloro-2,4′-difluoro-[1,1′-biphenyl]-3-yl)carbamoyl)-2-azabicyclo[3.1.0]hexan-2-yl)-2-oxoethyl)-5-((5-fluoropyrimidin-2-yl)ethynyl)-1H-indazole-3-carboxamide (64). 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 0.8 (m, 1H), 1.01-1.05 (m, 1H), 1.85-1.95 (m, 1H), 2.28-2.31 (m, 2H), 3.79 (t, J=4.8 Hz, 1H), 4.54 (t, J=6.4 Hz, 1H), 5.55 (d, J=17.2 Hz, 1H), 5.87 (d, J=17.2 Hz, 1H), 7.08 (t, J=6.8 Hz, 1H), 7.23 (t, J=8 Hz, 1H), 7.31-7.35 (m, 1H), 7.45 (t, J=8.8 Hz, 1H), 7.51 (s, 1H), 7.59 (dd, J=8.8, 2.4 Hz, 1H), 7.64 (d, J=8.8 Hz, 1H), 7.78 (d, J=8.8 Hz, 1H), 7.79 (s, 1H), 7.92 (t, J=7.6 Hz, 1H), 8.46 (s, 1H), 8.96 (s, 2H), 9.75 (s, 1H). 19F NMR (376 MHz, DMSO-d6, 300 K): (major rotamer) δ −111.6, −126.5, −135.7. LC (method 1): tR=2.56 min. LC/MS (EI) m/z: [M+H]+ calcd for C34H23ClF3N7O3, 670; found 670.
  • Compound 65: 1-(2-((1R,3S,5R)-3-((6-bromopyridin-2-yl)carbamoyl)-2-azabicyclo[3.1.0]hexan-2-yl)-2-oxoethyl)-5-((4-fluoropyridin-2-yl)ethynyl)-1H-indazole-3-carboxamide
  • Figure US20180291047A1-20181011-C00098
  • Procedure similar to the synthesis of 22e was followed. Thus, 0.1 g of 22d was coupled with 0.589 g of 2-bromo-4-fluoropyridine in presence of 77 mg of Pd(PPh3)4, 25 mg of CuI and 0.245 mL of TEA in 2 mL of DMF at 90° C. Crude product was purified by chromatography over silica gel (eluent: 0-1.5% MeOH in dichloromethane) to get 0.12 g of 65a as orange solid.
  • 0.05 g of 65a and 0.53 g of 41a were deprotected as described in synthesis of compound 22 (scheme 7&8). Coupling was done using the same procedure that was used for the synthesis of 22. 53 mg of 1-(2-((1R,3S,5R)-3-((6-Bromopyridin-2-yl)carbamoyl)-2-azabicyclo[3.1.0]hexan-2-yl)-2-oxoethyl)-5-((4-fluoropyridin-2-yl)ethynyl)-1H-indazole-3-carboxamide (65) was isolated as white solid.
  • 1H NMR (400 MHz, DMSO-d6, 300 K): (major rotamer) δ 0.75 (brm, 1H), 0.99-1.02 (m, 1H), 1.87-1.91 (m, 1H), 2.19-2.24 (m, 1H), 2.29-2.35 (m, 1H), 3.80 (t, J=4.8 Hz, 1H), 4.45 (dd, J=8.8, 5.2 Hz, 1H), 5.53 (d, J=17.2 Hz, 1H), 5.86 (d, J=17.2 Hz, 1H), 7.32 (d, J=8 Hz, 1H), 7.36-7.40 (m, 1H), 7.49 (s, 1H), 7.65 (d, J=8.8 Hz, 1H), 7.69-7.73 (m, 2H), 7.78 (d, J=9.2 Hz, 1H), 7.79 (s, 1H), 8.02 (d, J=8 Hz, 1H), 8.46 (s, 1H), 8.64 (dd, J=8.8, 6 Hz, 1H). 19F NMR (376 MHz, DMSO-d6, 300 K): (major rotamer) δ −103. LC (method 1): tR=2.41 min. LC/MS (EI) m/z: [M+H]+ calcd for C28H21BrFN7O3, 603; found 603.
  • Example 11. Non-Limiting Examples of Compounds of Formula I
  • Table 1 shows illustrative compounds of Formula I with characterizing data. The assay of Example 12 was used to determine the IC50'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
    RT min
    Cmp (Method A MS
    No. Structure Name IC50 or B) (M + 1)
     1
    Figure US20180291047A1-20181011-C00099
    (2S,4R)-1-(2-(3-acetyl-6- ethynyl-1H-indol-1- yl)acetyl)-N-(3-chloro-2- fluorobenzyl)-4- fluoropyrrolidine-2- carboxamide *** 1.97 (A) 498
     2
    Figure US20180291047A1-20181011-C00100
    3-(3-acetyl-1-(2-((2S,4R)-2- (3-chloro-2- fluorobenzylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-1H-indol-6- yl)propiolic acid *** 1.52 (A) 542
     3
    Figure US20180291047A1-20181011-C00101
    (2S,4R)-1-(2-(3-acetyl-6- ((trimethylsilyl)ethynyl)-1H- indol-1-yl)acetyl)-N-(3- chloro-2-fluorobenzyl)-4- fluoropyrrolidine-2- carboxamide *** 2.71 (A) 570
     4
    Figure US20180291047A1-20181011-C00102
    methyl 3-(3-acetyl-1-(2- ((2S,4R)-2-(3-chloro-2- fluorobenzylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-1H-indol-6- yl)propiolate *** 1.91 (A) 556
     5
    Figure US20180291047A1-20181011-C00103
    (2S,4R)-1-(2-(3-acetyl-6- ((4-amino-2-oxo-1,2- dihydropyrimidin-5- yl)ethynyl)-1H-indol-1- yl)acetyl)-N-(3-chloro-2- fluorobenzyl)-4- fluoropyrrolidine-2- carboxamide *** 1.02 (A) 607
     6
    Figure US20180291047A1-20181011-C00104
    3-(3-acetyl-1-(2-((2S,4R)-2- (3-chloro-2- fluorobenzylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-1H-indol-5- yl)propiolic acid *** 1.26 (A) 542
     7
    Figure US20180291047A1-20181011-C00105
    (2S,4R)-1-(2-(3-acetyl-5- ((4-amino-2-oxo-1,2- dihydropyrimidin-5- yl)ethynyl)-1H-indol-1- yl)acetyl)-N-(3-chloro-2- fluorobenzyl)-4- fluoropyrrolidine-2- carboxamide *** 1.17 (A) 607
     8
    Figure US20180291047A1-20181011-C00106
    (2S,4R)-1-(2-(3-acetyl-5-(3- (ethylsulfonamido)-3- oxoprop-1-ynyl)-1H-indol- 1-yl)acetyl)-N-(3-chloro-2- fluorobenzyl)-4- fluoropyrrolidine-2- carboxamide *** 1.73 (A) 633
     9
    Figure US20180291047A1-20181011-C00107
    (2S,4R)-1-(2-(3-acetyl-5- (pyrimidin-2-ylethynyl)-1H- indol-1-yl)acetyl)-N-(3- chloro-2-fluorobenzyl)-4- fluoropyrrolidine-2- carboxamide *** 1.74 (A) 576
    10
    Figure US20180291047A1-20181011-C00108
    (2S,4R)-1-(2-(3-acetyl-6-(3- (ethylsulfonamido)-3- oxoprop-1-ynyl)-1H-indol- 1-yl)acetyl)-N-(3-chloro-2- fluorobenzyl)-4- fluoropyrrolidine-2- carboxamide *** 1.73 (A) 633
    11
    Figure US20180291047A1-20181011-C00109
    3-(1-(2-((2S,4R)-2-(3- chloro-2- fluorobenzylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-3-(2,2,2- trifluoroacetyl)-1H-indol-6- yl)propiolic acid *** 2.11 (A) 596
    12
    Figure US20180291047A1-20181011-C00110
    (2S,4R)-1-(2-(3-acetyl-5- ((2-fluoro-3- (trifluoromethoxy)phenyl) ethynyl)-1H-indol-1- yl)acetyl)-N-(3-chloro-2- fluorobenzyl)-4- fluoropyrrolidine-2- carboxamide ** 2.83 (A) 676
    13
    Figure US20180291047A1-20181011-C00111
    (2S,4R)-1-(2-(3-acetyl-5- ((5-hydroxypyrimidin-2- yl)ethynyl)-1H-indol-1- yl)acetyl)-N-(3-chloro-2- fluorobenzyl)-4- fluoropyrrolidine-2- carboxamide *** 1.44 (A) 592
    14
    Figure US20180291047A1-20181011-C00112
    (2S,4R)-1-(2-(3-acetyl-5- (phenylethynyl)-1H-indol-1- yl)acetyl)-N-(3-chloro-2- fluorobenzyl)-4- fluoropyrrolidine-2- carboxamide *** 2.54 (A) 574
    15
    Figure US20180291047A1-20181011-C00113
    1-(2-((2S,4R)-2-(3-ethynyl- 2-fluorophenylcarbamoyl)- 4-fluoropyrrolidin-1-yl)-2- oxoethyl)-1H-pyrazolo[3,4- c]pyridine-3-carboxamide *** 0.91 (A) 453
    16
    Figure US20180291047A1-20181011-C00114
    (2S,4R)-1-(2-(3-acetyl-5- (pyrimidin-5-ylethynyl)-1H- indol-1-yl)acetyl)-N-(3- chloro-2-fluorobenzyl)-4- fluoropyrrolidine-2- carboxamide *** 1.84 (A) 576
    17
    Figure US20180291047A1-20181011-C00115
    (2S,4R)-1-(2-(3-acetyl-5- (pyridin-4-ylethynyl)-1H- indol-1-yl)acetyl)-N-(3- chloro-2-fluorobenzyl)-4- fluoropvrrolidine-2- carboxamide *** 1.52 (A) 575
    18
    Figure US20180291047A1-20181011-C00116
    (2S,4R)-1-(2-(3-acetyl-5- (isothiazol-4-ylethynyl)-1H- indol-1-yl)acetyl)-N-(3- chloro-2-fluorobenzyl)-4- fluoropyrrolidine-2- carboxamide *** 2.18 (A) 581
    19
    Figure US20180291047A1-20181011-C00117
    (2S,4R)-1-(2-(3-acetyl-5- (pyridin-2-ylethynyl)-1H- indol-1-yl)acetyl)-N-(3- chloro-2-fluorobenzyl)-4- fluoropyrrolidine-2- carboxamide *** 1.86 (A) 575
    20
    Figure US20180291047A1-20181011-C00118
    1-(2-((2S,4R)-2-(3-chloro-2- fluorobenzylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyrimidin-2- ylethynyl)-1H-indazole-3- carboxamide *** 1.52 (A) 578
    21
    Figure US20180291047A1-20181011-C00119
    (1R,3S,5R)-2-(2-(3-acetyl-5- (pyrimidin-2-ylethynyl)-1H- indol-1-yl)acetyl)-N-(6- bromopyridin-2-yl)-2- azabicyclo[3.1.0]hexane-3- carboxamide *** 184 (A) 583
    22
    Figure US20180291047A1-20181011-C00120
    1-(2-((2S,4R)-2-(2′-chloro- 2-fluorobiphenyl-3- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyrimidin-2- ylethynyl)-1H-indazole-3- carboxamide *** 1.99 (A) 640
    23
    Figure US20180291047A1-20181011-C00121
    1-(2-((2S,4R)-2-(3-chloro-2- fluorobenzylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyridin-2- ylethynyl)-1H-indazole-3- carboxamide *** 1.66 (A) 577
    24
    Figure US20180291047A1-20181011-C00122
    (2S,4R)-1-(2-(3-acetyl-5- ((3-cyano-1H-pyrazol-4- yl)ethynyl)-1H-indol-1- yl)acetyl)-N-(3-chloro-2- fluorobenzyl)-4- fluoropyrrolidine-2- carboxamide *** 1.84 (A) 589
    25
    Figure US20180291047A1-20181011-C00123
    1-(2-((2S,4R)-2-(3-chloro-2- fluorophenylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyrimidin-2- ylethynyl)-1H-indazole-3- carboxamide *** 1.56 (A) 564
    26
    Figure US20180291047A1-20181011-C00124
    (2S,4R)-1-(2-(3-acetyl-5- (pyrazin-2-ylethynyl)-1H- indol-1-yl)acetyl)-N-(3- chloro-2-fluorobenzyl)-4- fluoropyrrolidine-2- carboxamide *** 1.86 (A) 576
    27
    Figure US20180291047A1-20181011-C00125
    1-(2-((1R,3S,5R)-3-(2′- chloro-2-fluorobiphenyl-3- ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5- (pyrimidin-2-ylethynyl)-1H- indazole-3-carboxamide *** 2.10 (A) 634
    28
    Figure US20180291047A1-20181011-C00126
    1-(2-((2S,4R)-2-(3-(3- chloropyridin-2-yl)-2- fluorophenylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyrimidin-2- ylethynyl)-1H-indazole-3- carboxamide *** 1.54 (A) 641
    29
    Figure US20180291047A1-20181011-C00127
    5-((1H-pyrazol-4- yl)ethynyl)-1-(2-((2S,4R)-2- (3-chloro-2- fluorobenzylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-1H-indazole-3- carboxamide *** 1.49 (A) 566
    30
    Figure US20180291047A1-20181011-C00128
    1-(2-((2S,4R)-2-(3′-chloro- 2-fluorobiphenyl-3- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyrimidin-2- ylethynyl)-1H-indazole-3- carboxamide *** 2.10 (A) 640
    31
    Figure US20180291047A1-20181011-C00129
    1-(2-((2S,4R)-4-fluoro-2- (2,3,4,9-tetrahydro-1H- pyrido[3,4-b]indole-2- carbonyl)pyrrolidin-1-yl)-2- oxoethyl)-5-(pyrimidin-2- ylethynyl)-1H-indazole-3- carboxamide * 1.65 (A) 591
    32
    Figure US20180291047A1-20181011-C00130
    1-(2-((2S,4R)-2-(3-chloro-2- fluorobenzylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyrimidin-2- ylethynyl)-1H-pyrazolo[3,4- c]pyridine-3-carboxamide *** 1.32 (A) 579
    33
    Figure US20180291047A1-20181011-C00131
    1-(2-((2S,4R)-4-fluoro-2-(6- (trifluoromethyl)pyridin-2- ylcarbamoyl)pyrrolidin-1- yl)-2-oxoethyl)-5- (pyrimidin-2-ylethynyl)-1H- indazole-3-carboxamide *** 1.61 (A) 581
    34
    Figure US20180291047A1-20181011-C00132
    1-(2-((1R,3S,5R)-3-(2- fluoro-3- (trifluoromethoxy)phenyl- carbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5- (pyrimidin-2-ylethynyl)-1H- indazole-3-carboxamide *** 1.87 (A) 608
    35
    Figure US20180291047A1-20181011-C00133
    1-(2-((2S,4R)-2-(3-chloro-2- fluorobenzylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- fluoropyridin-2-yl)ethynyl)- 1H-indazole-3-carboxamide *** 1.91 (A) 595
    36
    Figure US20180291047A1-20181011-C00134
    5-((6-aminopyridin-2- yl)ethynyl)-1-(2-((2S,4R)-2- (3-chloro-2- fluorobenzylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-1H-indazole-3- carboxamide *** 1.27 (A) 592
    37
    Figure US20180291047A1-20181011-C00135
    1-(2-((2S,4R)-2-(3-chloro-2- fluorobenzylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((6- fluoropyridin-2-yl)ethynyl)- 1H-indazole-3-carboxamide *** 1.96 (A) 595
    38
    Figure US20180291047A1-20181011-C00136
    1-(2-((2S,4R)-4-fluoro-2-(2- fluoro-3- (trifluoromethoxy)phenyl- carbamoyl)pyrrolidin- 1-yl)-2- oxoethyl)-5-(pyrimidin-2- ylethynyl)-1H-indazole-3- carboxamide *** 1.77 (A) 614
    39
    Figure US20180291047A1-20181011-C00137
    1-(2-((2S,4R)-2-(6- bromopyridin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyrimidin-2- ylethynyl)-1H-indazole-3- carboxamide *** 1.48 (A) 591
    40
    Figure US20180291047A1-20181011-C00138
    1-(2-((2S,4R)-2-(3-chloro-2- fluorobenzylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 1.73 (A) 596
    41
    Figure US20180291047A1-20181011-C00139
    1-(2-((1R,3S,5R)-3-(6- bromopyridin-2- ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5- (pyrimidin-2-ylethynyl)-1H- indazole-3-carboxamide *** 1.60 (A) 585
    42
    Figure US20180291047A1-20181011-C00140
    (2S,4R)-1-(2-(3-acetyl-5- (pyrimidin-2-ylethynyl)-1H- indol-1-yl)acetyl)-N-(6- bromopyridin-2-yl)-4- fluoropyrrolidine-2- carboxamide *** 1.70 (A) 589
    43
    Figure US20180291047A1-20181011-C00141
    1-(2-((1R,3S,5R)-3-(6- bromopyridin-2- ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 1.81 (A) 603
    44
    Figure US20180291047A1-20181011-C00142
    1-(2-((2S,4R)-2-(3,3- dimethylcyclohexyl- carbamoyl)-4- fluoropyrrolidin-1-yl)- 2-oxoethyl)-5-(pyrimidin-2- ylethynyl)-1H-indazole-3- carboxamide *** 1.69 (A) 546
    45
    Figure US20180291047A1-20181011-C00143
    1-(2-((2S,4R)-4-fluoro-2-(2- fluoro-3-methylbut-2- enylcarbamoyl)pyrrolidin-1- yl)-2-oxoethyl)-5- (pyrimidin-2-ylethynyl)-1H- indazole-3-carboxamide *** 1.31 (A) 522
    46
    Figure US20180291047A1-20181011-C00144
    1-(2-((2S,4R)-2-(6- bromopyridin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 1.69 (A) 609
    47
    Figure US20180291047A1-20181011-C00145
    1-(2-((2S,4R)-2-(6-(2- chlorophenyl)pyridin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyrimidin-2- ylethynyl)-1H-indazole-3- carboxamide *** 1.91 (A) 623
    48
    Figure US20180291047A1-20181011-C00146
    1-(2-((2S,4R)-2-(6- bromopyridin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyridin-2- ylethynyl)-1H-indazole-3- carboxamide *** 1.60 (A) 590
    49
    Figure US20180291047A1-20181011-C00147
    1-(2-((2S,4R)-2-(2′-chloro- 2,4′-difluorobiphenyl-3- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyrimidin-2- ylethynyl)-1H-indazole-3- carboxamide *** 2.04 (A) 658
    50
    Figure US20180291047A1-20181011-C00148
    1-(2-((2S,4R)-2-(6-(1H- pyrazol-4-yl)pyridin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyrimidin-2- ylethynyl)-1H-indazole-3- carboxamide *** 1.11 (A) 579
    51
    Figure US20180291047A1-20181011-C00149
    1-(2-((1R,3S,5R)-3-(3- chloro-2- fluorobenzylcarbamoyl)-6,6- difluoro-2- azabicyclo[3.2.0]heptan-2- yl)-2-oxoethyl)-5- (pyrimidin-2-ylethynyl)-1H- indazole-3-carboxamide ** 1.79 (A) 622
    52
    Figure US20180291047A1-20181011-C00150
    1-(2-((2S,3R)-2-(3-chloro-2- fluorobenzylcarbamoyl)-3- hydroxypyrrolidin-1-yl)-2- oxoethyl)-5-(pyrimidin-2- ylethynyl)-1H-indazole-3- carboxamide *** 1.28 (A) 576
    53
    Figure US20180291047A1-20181011-C00151
    1-(2-((1R,3S,5R)-3-(2′- chloro-2-fluorobiphenyl-3- ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 2.31 (A) 652
    54
    Figure US20180291047A1-20181011-C00152
    1-(2-((1R,3S,5R)-3-(6- cyclopropylpyridin-2- ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5- (pyrimidin-2-ylethynyl)-1H- indazole-3-carboxamide *** 1.62 (A) 547
    55
    Figure US20180291047A1-20181011-C00153
    1-(2-((2S,4R)-2-(2′-chloro- 2-fluorobiphenyl-3- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 2.20 (A) 658
    56
    Figure US20180291047A1-20181011-C00154
    1-(2-((1R,3S,5R)-3-(6- chloropyridin-2- ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5- (pyrimidin-2-ylethynyl)-1H- indazole-3-carboxamide *** 1.56 (A) 541
    57
    Figure US20180291047A1-20181011-C00155
    1-(2-((1R,3S,5R)-3-(6- cyanopyridin-2- ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5- (pyrimidin-2-ylethynyl)-1H- indazole-3-carboxamide *** 1.39 (A) 532
    58
    Figure US20180291047A1-20181011-C00156
    1-(2-((1R,3S,5R)-3-(6- bromopyridin-2- ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5-((5- fluoropyridin-2-yl)ethynyl)- 1H-indazole-3-carboxamide *** 2.01 (A) 602
    59
    Figure US20180291047A1-20181011-C00157
    1-(2-((1R,3S,5R)-3-(6- bromopyridin-2- ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5-((5- (trifluoromethyl)pyridin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 2.33 (A) 654
    60
    Figure US20180291047A1-20181011-C00158
    1-(2-((1R,3S,5R)-3-(6- bromopyridin-2- ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5-((5- cyanopyridin-2-yl)ethynyl)- 1H-indazole-3-carboxamide *** 1.93 (A) 609
    61
    Figure US20180291047A1-20181011-C00159
    1-(2-((1R,3S,5R)-3-(6- bromopyridin-2- ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5-((5- (trifluoromethyl)pyrimidin- 2-yl)ethynyl)-1H-indazole- 3-carboxamide *** 2.17 (A) 653
    62
    Figure US20180291047A1-20181011-C00160
    1-(2-((1R,3S,5R)-3-(6- fluoropyridin-2- ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5- (pyrimidin-2-ylethynyl)-1H- indazole-3-carboxamide *** 1.44 (A) 525
    63
    Figure US20180291047A1-20181011-C00161
    1-(2-((2S,4R)-2-(2′-chloro- 2,4′,5′-trifluorobiphenyl-3- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 2.32 (A) 694
    64
    Figure US20180291047A1-20181011-C00162
    1-(2-((1R,3S,5R)-3-(2′- chloro-2,4′- difluorobiphenyl-3- ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 2.56 (A) 670
    65
    Figure US20180291047A1-20181011-C00163
    1-(2-((1R,3S,5R)-3-(6- bromopyridin-2- ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5-((4- fluoropyridin-2-yl)ethynyl)- 1H-indazole-3-carboxamide *** 2.41 (A) 604
    66
    Figure US20180291047A1-20181011-C00164
    (S)-3-(2-(3-carbamoyl-5- (pyrimidin-2-ylethynyl)-1H- indazol-1-yl)acetyl)-N-(2′- chloro-2-fluorobiphenyl-3- yl)thiazolidine-2- carboxamide *** 2.47 (A) 640
    67
    Figure US20180291047A1-20181011-C00165
    1-(2-((2S,4R)-2-(3-chloro-2- fluorobenzylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5- (phenylethynyl)-1H- pyrazolo[3,4-c]pyridine-3- carboxamide *** 2.46 (A) 577
    68
    Figure US20180291047A1-20181011-C00166
    1-(2-((2S,4R)-2-(5- bromopyridin-3- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 2.03 (A) 609
    69
    Figure US20180291047A1-20181011-C00167
    1-(2-((2S,4R)-4-fluoro-2-(2- fluoro-3-(pyrimidin-5- yl)phenylcarbamoyl) pyrrolidin-1-yl)-2- oxoethyl)-5- (pyrimidin-2-ylethynyl)-1H- indazole-3-carboxamide *** 1.16 (A) 608
    70
    Figure US20180291047A1-20181011-C00168
    1-(2-((2S,4R)-4-fluoro-2-(6- (trifluoromethyl)pyrazin-2- ylcarbamoyl)pyrrolidin-1- yl)-2-oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 1.65 (A) 600
    71
    Figure US20180291047A1-20181011-C00169
    1-(2-((2S,4R)-2-(8- chloroquinolin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 1.94 (A) 615
    72
    Figure US20180291047A1-20181011-C00170
    5-((5-fluoropyrimidin-2- yl)ethynyl)-1-(2-oxo-2- ((1R,3S,5R)-3-(1-(2,2,2- trifluoroethyl)-1H-pyrazol- 3-ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)ethyl)-1H-indazole-3- carboxamide *** 1.54 (A) 596
    73
    Figure US20180291047A1-20181011-C00171
    1-(2-((2S,4R)-2-(2′-chloro- 2-fluorobiphenyl-3- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyridazin-3- ylethynyl)-1H-indazole-3- carboxamide *** 1.93 (A) 640
    74
    Figure US20180291047A1-20181011-C00172
    1-(2-((2S,4R)-2-(6-chloro-4- (trifluoromethyl)pyridin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 2.06 (A) 633
    75
    Figure US20180291047A1-20181011-C00173
    1-(2-((2S,4R)-2-(6- chloropyridin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyridazin-3- ylethynyl)-1H-indazole-3- carboxamide *** 1.37 (A) 547
    76
    Figure US20180291047A1-20181011-C00174
    1-(2-((2S,4R)-2-(6-chloro-5- (trifluoromethyl)pyridin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 2.05 (A) 633
    77
    Figure US20180291047A1-20181011-C00175
    1-(2-((1R,3S,5R)-3-(6- chloropyridin-2- ylcarbamoyl)-2- azabicyclo[3.1.0]hexan-2- yl)-2-oxoethyl)-5- (pyridazin-3-ylethynyl)-1H- indazole-3-carboxamide *** 1.48 (A) 541
    78
    Figure US20180291047A1-20181011-C00176
    1-(2-((2S,4R)-2-(2′-chloro- 2-fluorobiphenyl-3- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5-fluoro-4- methoxypyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 2.37 (A) 688
    79
    Figure US20180291047A1-20181011-C00177
    1-(2-((2S,4R)-2-(2′-chloro- 2-fluorobiphenyl-3- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyridazin-4- ylethynyl)-1H-indazole-3- carboxamide *** 1.93 (A) 640
    80
    Figure US20180291047A1-20181011-C00178
    1-(2-((2S,4R)-2-(6- chloropyridin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyridazin-4- ylethynyl)-1H-indazole-3- carboxamide *** 1.37 (A) 547
    81
    Figure US20180291047A1-20181011-C00179
    1-(2-((2S,4R)-2-(6-chloro-3- fluoropyridin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 1.44 (A) 583
    82
    Figure US20180291047A1-20181011-C00180
    1-(2-((2S,4R)-2-(6- chloropyridin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyrazin-2- ylethynyl)-1H-indazole-3- carboxamide *** 1.53 (A) 547
    83
    Figure US20180291047A1-20181011-C00181
    1-(2-((2S,4R)-2-(2′-chloro- 2-fluorobiphenyl-3- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyrazin-2- ylethynyl)-1H-indazole-3- carboxamide *** 2.08 (A) 640
    84
    Figure US20180291047A1-20181011-C00182
    1-(2-((2S,4R)-2-(6- bromopyrazin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 1.52 (A) 611
    85
    Figure US20180291047A1-20181011-C00183
    1-(2-((2S,4R)-2-(6-(2- chlorophenyl)pyrazin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 1.88 (A) 642
    86
    Figure US20180291047A1-20181011-C00184
    1-(2-((2S,4R)-2-(6-(2- chloro-6- fluorophenyl)pyrazin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 1.87 (A) 660
    87
    Figure US20180291047A1-20181011-C00185
    1-(2-((2S,4R)-2-(6-(2- chloro-4,5- difluorophenyl)pyrazin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 2.01 (A) 678
    88
    Figure US20180291047A1-20181011-C00186
    1-(2-((2S,4R)-2-(6- chloropyridin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- fluoropyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 1.64 (A) 565
    89
    Figure US20180291047A1-20181011-C00187
    1-(2-((2S,4R)-2-(6- bromopyrazin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-(pyrazin-2- ylethynyl)-1H-indazole-3- carboxamide *** 1.41 (A) 594
    90
    Figure US20180291047A1-20181011-C00188
    (2S,4R)-1-(2-(3-acetyl-5- ((5-fluoropyrimidin-2- yl)ethynyl)-1H-indazol-1- yl)acelyl)-N-(6- bromopyridin-2-yl)-4- fluoropyrrolidine-2- carboxamide *** 2.14 (A) 608
    91
    Figure US20180291047A1-20181011-C00189
    1-(2-((2S,4R)-2-(6- bromopyridin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((5- methylpyrimidin-2- yl)ethynyl)-1H-indazole-3- carboxamide *** 1.68 (A) 605
    92
    Figure US20180291047A1-20181011-C00190
    1-(2-((2S,4R)-2-(6- bromopyridin-2- ylcarbamoyl)-4- fluoropyrrolidin-1-yl)-2- oxoethyl)-5-((2- methoxypyrimidin-5- yl)ethynyl)-1H-indazole-3- carboxamide *** 1.83 (A) 623
  • Example 12. Human Factor D Assay
  • 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 μM 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 spectrofluorimeter. 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 13. Hemolysis Assay
  • 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 erythrocyctes (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 1×108 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).
  • 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 (11)

1. A compound of Formula I
Figure US20180291047A1-20181011-C00191
or a pharmaceutically acceptable salt thereof, wherein:
Q1 is C(R1R1′);
Q2 is C(R2R2′);
Q3 is C(R3R3′);
X1 is N and X2 is CH;
R1, R1′, R2, R2′, R3, and R3′ are independently chosen from hydrogen, halogen, and C1-C6alkyl;
A is:
Figure US20180291047A1-20181011-C00192
R4 is chosen from —CHO, —CONH2, C2-C6alkanoyl, hydrogen, —SO2NH2, —C(CH2)2F, —CH(CF3)NH2, C1-C6alkyl, —C0-C4alkyl(C3-C7cycloalkyl), —C(O)C0-C2alkyl(C3-C7cycloalkyl),
Figure US20180291047A1-20181011-C00193
R5 is selected from hydrogen —CHO, —C(O)NH2, —C(O)NH(CH3), and C2-C6alkanoyl;
R9 and R10 are independently chosen at each occurrence from hydrogen, and C1-C6alkyl;
X12 is CR12;
X13 is CR13;
one of R12 and R13 is chosen from R31 and the other of R12 and R13 is chosen from R32:
R31 is chosen from hydrogen, halogen, hydroxyl, nitro, cyano, amino, —COOH, C1-C2haloalkyl, C1-C2haloalkoxy, C1-C6alkyl, —C0-C4alkyl(C3-C7cycloalkyl), C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, C2-C6alkenyloxy, —C(O)OR9, C1-C6thioalkyl, —C0-C4alkylNR9R10, —C(O)NR9R10, —SO2R9, —SO2NR9R10, —OC(O)R9, and —C(NR9)NR9R10;
R9 and R10 are independently chosen at each occurrence from hydrogen and C1-C6alkyl;
R32 is
Figure US20180291047A1-20181011-C00194
R30 is independently chosen at each occurrence from hydrogen, (phenyl)C0-C4alkyl, and (5- or 6-membered unsaturated or aromatic heterocycle)C0-C4alkyl having 1, 2, or 3 heteroatoms independently chosen from N, O, and S;
L is
Figure US20180291047A1-20181011-C00195
R17 is hydrogen;
R18 and R18′ are hydrogen;
m is 0, 1, 2, or 3;
B is —(C0-C4alkyl)(aryl); —(C0-C4alkyl)(heteroaryl); or —(C0-C4alkyl)(biphenyl) each of which B is unsubstituted or substituted with one or more substituents independently chosen from R33 and R34, and 0 or 1 substituents chosen from R36;
R33 is independently chosen from halogen, hydroxyl, —COOH, cyano, C1-C6alkyl, C2-C6alkanoyl, C1-C6alkoxy, —C0-C4alkylNR9R10, —SO2R9, C1-C2haloalkyl, and C1-C2haloalkoxy;
R34 is independently chosen from nitro, C2-C6alkenyl, C2-C6alkynyl, and C1-C6thioalkyl; and
R36 is independently chosen from (phenyl)C0-C2alkyl, (phenyl)C1-C2alkoxy, phenoxy, and 6-membered heteroaryl containing 1, 2, or 3 heteroatoms independently chosen from N, O, B, and S, each of which R36 is unsubstituted or substituted with one or more substituents independently chosen from halogen, hydroxyl, nitro, cyano, C1-C6alkyl, C2-C6alkenyl, C2-C6alkanoyl, C1-C6alkoxy, (mono- and di-C1-C6alkylamino)C0-C4alkyl, C1-C6alkylester, —C0-C4alkyl(C3-C7cycloalkyl), —SO2R9, C1-C2haloalkyl, and C1-C2haloalkoxy.
2. The compound of claim 1, wherein the
Figure US20180291047A1-20181011-C00196
ring is selected from:
Figure US20180291047A1-20181011-C00197
3. The compound of claim 1, wherein R5 is hydrogen.
4. The compound of claim 1, wherein R4 is chosen from —CHO, —CONH2, and C2-C6alkanoyl.
5. The compound of claim 4, wherein R31 is hydrogen.
6. The compound of claim 5, wherein m is 0 or 1.
7. The compound of claim 1, wherein B is selected from:
Figure US20180291047A1-20181011-C00198
8. The compound of claim 1, wherein B is —(CH2)(aryl) substituted with two substituents independently chosen from R33 and R34.
9. The compound of claim 8, wherein R33 is halogen.
10. The compound of claim 9, wherein there are two R33 substituents and one is chlorine and the other is fluorine.
11. The compound of claim 1, wherein B is aryl or heteroaryl each of which B is unsubstituted or substituted with one or more substituents independently chosen from R33 and R34.
US16/006,533 2014-02-25 2018-06-12 Alkyne compounds for treatment of complement mediated disorders Abandoned US20180291047A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/006,533 US20180291047A1 (en) 2014-02-25 2018-06-12 Alkyne 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
US201462022916P 2014-07-10 2014-07-10
US201462046783P 2014-09-05 2014-09-05
US14/631,090 US9828396B2 (en) 2014-02-25 2015-02-25 Alkyne compounds for treatment of complement mediated disorders
US15/818,559 US20180072762A1 (en) 2014-02-25 2017-11-20 Alkyne compounds for treatment of complement mediated disorders
US16/006,533 US20180291047A1 (en) 2014-02-25 2018-06-12 Alkyne compounds for treatment of complement mediated disorders

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/818,559 Continuation US20180072762A1 (en) 2014-02-25 2017-11-20 Alkyne compounds for treatment of complement mediated disorders

Publications (1)

Publication Number Publication Date
US20180291047A1 true US20180291047A1 (en) 2018-10-11

Family

ID=53881572

Family Applications (33)

Application Number Title Priority Date Filing Date
US14/631,625 Active 2035-12-02 US9796741B2 (en) 2014-02-25 2015-02-25 Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US14/631,683 Active US9732104B2 (en) 2014-02-25 2015-02-25 Ether compounds for treatment of complement mediated disorders
US14/631,440 Active US9732103B2 (en) 2014-02-25 2015-02-25 Carbamate, ester, and ketone compounds for treatment of complement mediated disorders
US14/631,312 Active US9598446B2 (en) 2014-02-25 2015-02-25 Amino compounds for treatment of complement mediated disorders
US14/631,090 Active 2035-07-18 US9828396B2 (en) 2014-02-25 2015-02-25 Alkyne compounds for treatment of complement mediated disorders
US14/630,959 Active US9643986B2 (en) 2014-02-25 2015-02-25 Factor D inhibitors useful for treating inflammatory disorders
US14/631,828 Active 2035-08-26 US9758537B2 (en) 2014-02-25 2015-02-25 Compounds for treatment of complement mediated disorders
US14/631,785 Active US9663543B2 (en) 2014-02-25 2015-02-25 Phosphonate compounds for treatment of complement mediated disorders
US14/631,233 Active US9695205B2 (en) 2014-02-25 2015-02-25 Amide compounds for treatment of complement mediated disorders
US15/246,049 Abandoned US20160361329A1 (en) 2014-02-25 2016-08-24 Compounds for treatment of complement mediated disorders
US15/245,712 Active US10005802B2 (en) 2014-02-25 2016-08-24 Amide compounds for treatment of complement mediated disorders
US15/245,788 Abandoned US20160362399A1 (en) 2014-02-25 2016-08-24 Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US15/245,872 Abandoned US20160362433A1 (en) 2014-02-25 2016-08-24 Ether compounds for treatment of complement mediated disorders
US15/245,945 Abandoned US20160362432A1 (en) 2014-02-25 2016-08-24 Phosphonate compounds for treatment of complement mediated disorders
US15/463,701 Active US10189869B2 (en) 2014-02-25 2017-03-20 Amino compounds for treatment of complement mediated disorders
US15/494,926 Abandoned US20170226142A1 (en) 2014-02-25 2017-04-24 Factor D Inhibitors Useful for Treating Inflammatory Disorders
US15/607,120 Active US10100072B2 (en) 2014-02-25 2017-05-26 Phosphonate compounds for treatment of complement mediated disorders
US15/638,081 Active US10370394B2 (en) 2014-02-25 2017-06-29 Carbamate, ester, and ketone compounds for treatment of complement mediated disorders
US15/638,076 Active US10428094B2 (en) 2014-02-25 2017-06-29 Amide compounds for treatment of complement mediated disorders
US15/676,411 Active US10106563B2 (en) 2014-02-25 2017-08-14 Ether compounds for treatment of complement mediated disorders
US15/700,550 Active US10087203B2 (en) 2014-02-25 2017-09-11 Compounds for treatment of complement mediated disorders
US15/711,794 Active US10081645B2 (en) 2014-02-25 2017-09-21 Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US15/818,559 Abandoned US20180072762A1 (en) 2014-02-25 2017-11-20 Alkyne compounds for treatment of complement mediated disorders
US16/006,533 Abandoned US20180291047A1 (en) 2014-02-25 2018-06-12 Alkyne compounds for treatment of complement mediated disorders
US16/006,520 Abandoned US20180291046A1 (en) 2014-02-25 2018-06-12 Alkyne compounds for treatment of complement mediated disorders
US16/006,476 Abandoned US20180298043A1 (en) 2014-02-25 2018-06-12 Alkyne Compounds for Treatment of Complement Mediated Disorders
US16/140,148 Active US10253053B2 (en) 2014-02-25 2018-09-24 Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US16/148,622 Active US10428095B2 (en) 2014-02-25 2018-10-01 Compounds for treatment of complement mediated disorders
US16/162,162 Active US10301336B2 (en) 2014-02-25 2018-10-16 Phosphonate compounds for treatment of complement mediated disorders
US16/164,632 Active US10550140B2 (en) 2014-02-25 2018-10-18 Ether compounds for treatment of complement mediated disorders
US16/246,832 Active US10689409B2 (en) 2014-02-25 2019-01-14 Amino compounds for treatment of complement mediated disorders
US16/276,139 Active US10464956B2 (en) 2014-02-25 2019-02-14 Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US16/672,114 Pending US20200062790A1 (en) 2014-02-25 2019-11-01 Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders

Family Applications Before (23)

Application Number Title Priority Date Filing Date
US14/631,625 Active 2035-12-02 US9796741B2 (en) 2014-02-25 2015-02-25 Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US14/631,683 Active US9732104B2 (en) 2014-02-25 2015-02-25 Ether compounds for treatment of complement mediated disorders
US14/631,440 Active US9732103B2 (en) 2014-02-25 2015-02-25 Carbamate, ester, and ketone compounds for treatment of complement mediated disorders
US14/631,312 Active US9598446B2 (en) 2014-02-25 2015-02-25 Amino compounds for treatment of complement mediated disorders
US14/631,090 Active 2035-07-18 US9828396B2 (en) 2014-02-25 2015-02-25 Alkyne compounds for treatment of complement mediated disorders
US14/630,959 Active US9643986B2 (en) 2014-02-25 2015-02-25 Factor D inhibitors useful for treating inflammatory disorders
US14/631,828 Active 2035-08-26 US9758537B2 (en) 2014-02-25 2015-02-25 Compounds for treatment of complement mediated disorders
US14/631,785 Active US9663543B2 (en) 2014-02-25 2015-02-25 Phosphonate compounds for treatment of complement mediated disorders
US14/631,233 Active US9695205B2 (en) 2014-02-25 2015-02-25 Amide compounds for treatment of complement mediated disorders
US15/246,049 Abandoned US20160361329A1 (en) 2014-02-25 2016-08-24 Compounds for treatment of complement mediated disorders
US15/245,712 Active US10005802B2 (en) 2014-02-25 2016-08-24 Amide compounds for treatment of complement mediated disorders
US15/245,788 Abandoned US20160362399A1 (en) 2014-02-25 2016-08-24 Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US15/245,872 Abandoned US20160362433A1 (en) 2014-02-25 2016-08-24 Ether compounds for treatment of complement mediated disorders
US15/245,945 Abandoned US20160362432A1 (en) 2014-02-25 2016-08-24 Phosphonate compounds for treatment of complement mediated disorders
US15/463,701 Active US10189869B2 (en) 2014-02-25 2017-03-20 Amino compounds for treatment of complement mediated disorders
US15/494,926 Abandoned US20170226142A1 (en) 2014-02-25 2017-04-24 Factor D Inhibitors Useful for Treating Inflammatory Disorders
US15/607,120 Active US10100072B2 (en) 2014-02-25 2017-05-26 Phosphonate compounds for treatment of complement mediated disorders
US15/638,081 Active US10370394B2 (en) 2014-02-25 2017-06-29 Carbamate, ester, and ketone compounds for treatment of complement mediated disorders
US15/638,076 Active US10428094B2 (en) 2014-02-25 2017-06-29 Amide compounds for treatment of complement mediated disorders
US15/676,411 Active US10106563B2 (en) 2014-02-25 2017-08-14 Ether compounds for treatment of complement mediated disorders
US15/700,550 Active US10087203B2 (en) 2014-02-25 2017-09-11 Compounds for treatment of complement mediated disorders
US15/711,794 Active US10081645B2 (en) 2014-02-25 2017-09-21 Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US15/818,559 Abandoned US20180072762A1 (en) 2014-02-25 2017-11-20 Alkyne compounds for treatment of complement mediated disorders

Family Applications After (9)

Application Number Title Priority Date Filing Date
US16/006,520 Abandoned US20180291046A1 (en) 2014-02-25 2018-06-12 Alkyne compounds for treatment of complement mediated disorders
US16/006,476 Abandoned US20180298043A1 (en) 2014-02-25 2018-06-12 Alkyne Compounds for Treatment of Complement Mediated Disorders
US16/140,148 Active US10253053B2 (en) 2014-02-25 2018-09-24 Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US16/148,622 Active US10428095B2 (en) 2014-02-25 2018-10-01 Compounds for treatment of complement mediated disorders
US16/162,162 Active US10301336B2 (en) 2014-02-25 2018-10-16 Phosphonate compounds for treatment of complement mediated disorders
US16/164,632 Active US10550140B2 (en) 2014-02-25 2018-10-18 Ether compounds for treatment of complement mediated disorders
US16/246,832 Active US10689409B2 (en) 2014-02-25 2019-01-14 Amino compounds for treatment of complement mediated disorders
US16/276,139 Active US10464956B2 (en) 2014-02-25 2019-02-14 Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders
US16/672,114 Pending US20200062790A1 (en) 2014-02-25 2019-11-01 Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders

Country Status (26)

Country Link
US (33) US9796741B2 (en)
EP (10) EP3110419A4 (en)
JP (9) JP2017506672A (en)
KR (7) KR102366025B1 (en)
CN (8) CN112250673A (en)
AP (5) AP2016009435A0 (en)
AU (7) AU2015223072A1 (en)
BR (1) BR112016019526B1 (en)
CA (6) CA2940769A1 (en)
CY (1) CY1122550T1 (en)
DK (1) DK3110418T3 (en)
EA (5) EA201691727A1 (en)
ES (2) ES2771950T3 (en)
HR (1) HRP20200182T1 (en)
HU (1) HUE047295T2 (en)
IL (8) IL301427A (en)
LT (1) LT3110418T (en)
MX (6) MX2016011038A (en)
PL (1) PL3110418T3 (en)
PT (1) PT3110418T (en)
RS (1) RS59854B1 (en)
RU (4) RU2016137524A (en)
SA (1) SA516371744B1 (en)
SI (1) SI3110418T1 (en)
WO (8) WO2015130838A1 (en)
ZA (2) ZA201605832B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11926617B2 (en) 2015-08-26 2024-03-12 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of immune and inflammatory disorders

Families Citing this family (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2016137524A (en) * 2014-02-25 2018-04-02 Ачиллион Фармасьютикалс, Инк. AMIDIC COMPOUNDS FOR TREATMENT OF COMPLETE-MEDIATED DISORDERS
CA2977981C (en) 2015-02-27 2023-03-21 The Johns Hopkins University Assay to diagnose and treat disorders of the alternative pathway of complement activation
AR106018A1 (en) 2015-08-26 2017-12-06 Achillion Pharmaceuticals Inc ARYL, HETEROARYL AND HETEROCYCLIC 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
EP3340982B1 (en) 2015-08-26 2021-12-15 Achillion Pharmaceuticals, Inc. Compounds for treatment of immune and inflammatory disorders
WO2017035401A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of immune and inflammatory disorders
AR105809A1 (en) 2015-08-26 2017-11-08 Achillion Pharmaceuticals Inc 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
WO2017035355A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Ether 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
AR105808A1 (en) 2015-08-26 2017-11-08 Achillion Pharmaceuticals Inc AMIDA COMPOUNDS FOR THE TREATMENT OF MEDICAL DISORDERS
WO2017035405A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of immune and inflammatory disorders
EP3365340B1 (en) 2015-10-19 2022-08-10 Incyte Corporation Heterocyclic compounds as immunomodulators
SG10202004618TA (en) 2015-11-19 2020-06-29 Incyte Corp Heterocyclic compounds as immunomodulators
CA3007922A1 (en) * 2015-12-11 2017-06-15 Lifesci Pharmaceuticals, Inc. Therapeutic inhibitory compounds
PL3394033T3 (en) 2015-12-22 2021-05-31 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017127761A1 (en) 2016-01-20 2017-07-27 Vitrisa Therapeutics, Inc. Compositions and methods for inhibiting factor d
TW202222786A (en) * 2016-02-01 2022-06-16 美商百歐克斯製藥公司 Benzopyrazole compounds and analogues thereof
EP3452476B1 (en) 2016-05-06 2021-12-15 Incyte Corporation Heterocyclic compounds as immunomodulators
ES2905980T3 (en) 2016-05-26 2022-04-12 Incyte Corp Heterocyclic compounds as immunomodulators
UA125391C2 (en) 2016-06-20 2022-03-02 Інсайт Корпорейшн Heterocyclic compounds as immunomodulators
EP3484866B1 (en) 2016-07-14 2022-09-07 Incyte Corporation Heterocyclic compounds as immunomodulators
US20190292155A1 (en) * 2016-07-15 2019-09-26 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
WO2018044783A1 (en) 2016-08-29 2018-03-08 Incyte Corporation Heterocyclic compounds as immunomodulators
CN115819417A (en) 2016-09-09 2023-03-21 因赛特公司 Pyrazolopyridine derivatives as HPK1 modulators and their use for the treatment of cancer
WO2018049214A1 (en) 2016-09-09 2018-03-15 Incyte Corporation Pyrazolopyridine derivatives as hpk1 modulators and uses thereof for the treatment of cancer
US20180072741A1 (en) 2016-09-09 2018-03-15 Incyte Corporation Pyrazolopyrimidine compounds and uses thereof
US20180179201A1 (en) 2016-12-22 2018-06-28 Incyte Corporation Heterocyclic compounds as immunomodulators
PT3558990T (en) 2016-12-22 2022-11-21 Incyte Corp Tetrahydro imidazo[4,5-c]pyridine derivatives as pd-l1 internalization inducers
EP3558973B1 (en) 2016-12-22 2021-09-15 Incyte Corporation Pyridine derivatives as immunomodulators
MY197635A (en) 2016-12-22 2023-06-29 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
US20180228786A1 (en) 2017-02-15 2018-08-16 Incyte Corporation Pyrazolopyridine compounds and uses thereof
MX2019010219A (en) * 2017-02-27 2020-02-07 Regeneron Pharma Humanized model of kidney and liver disorders.
ES2933513T3 (en) * 2017-03-01 2023-02-09 Achillion Pharmaceuticals Inc Macrocyclic compounds for the treatment of medical disorders
WO2018160891A1 (en) * 2017-03-01 2018-09-07 Achillion Pharmaceutical, Inc. Pharmaceutical compounds for treatment of medical disorders
EP3985002A1 (en) 2017-03-01 2022-04-20 Achillion Pharmaceuticals, Inc. Aryl, heteroary, and heterocyclic pharmaceutical compounds for treatment of medical disorders
WO2018229543A2 (en) * 2017-06-14 2018-12-20 Lifesci Pharmaceuticals, Inc. Therapeutic inhibitory compounds
WO2019028284A1 (en) * 2017-08-02 2019-02-07 Achillion Pharmaceuticals, Inc. Therapeutic regimens for treatment of paroxysmal nocturnal hemoglobinuria
WO2019051199A1 (en) 2017-09-08 2019-03-14 Incyte Corporation 6-cyano-indazole compounds as hematopoietic progenitor kinase 1 (hpk1) modulators
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
PE20210397A1 (en) 2018-02-20 2021-03-02 Incyte Corp DERIVATIVES OF N- (PHENYL) -2- (PHENYL) PYRIMIDINE-4-CARBOXAMIDE AND RELATED COMPOUNDS AS HPKI INHIBITORS TO TREAT CANCER
WO2019164847A1 (en) 2018-02-20 2019-08-29 Incyte Corporation Indazole compounds and uses thereof
HUE061258T2 (en) 2018-03-30 2023-05-28 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
RS64188B1 (en) 2018-05-11 2023-06-30 Incyte Corp 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
WO2020041301A1 (en) * 2018-08-20 2020-02-27 Achillion Pharmaceuticals, Inc. Pharmaceutical compounds for the treatment of complement factor d medical disorders
JP7443375B2 (en) 2018-09-06 2024-03-05 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 Macrocyclic compounds for the treatment of medical disorders
JP2021536511A (en) 2018-09-06 2021-12-27 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 Form of Danico bread
EP3856348B1 (en) 2018-09-25 2024-01-03 Incyte Corporation Pyrazolo[4,3-d]pyrimidine compounds as alk2 and/or fgfr modulators
CA3114039A1 (en) 2018-09-25 2020-04-02 Achillion Pharmaceuticals, Inc. Morphic forms of complement factor d inhibitors
EP3873464A4 (en) * 2018-11-01 2022-06-08 Merck Sharp & Dohme Corp. Novel substituted pyrazole compounds as indoleamine 2,3-dioxygenase 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
WO2020109343A1 (en) 2018-11-29 2020-06-04 F. Hoffmann-La Roche Ag Combination therapy for treatment of macular degeneration
AU2020245434A1 (en) * 2019-03-22 2021-09-30 Achillion Pharmaceuticals, Inc. Pharmaceutical compounds for the treatment of complement mediated disorders
US11066394B2 (en) 2019-08-06 2021-07-20 Incyte Corporation Solid forms of an HPK1 inhibitor
JP2022544189A (en) 2019-08-09 2022-10-17 インサイト・コーポレイション Salts of PD-1/PD-L1 inhibitors
AR120109A1 (en) 2019-09-30 2022-02-02 Incyte Corp PYRIDO[3,2-D]PYRIMIDINE COMPOUNDS AS IMMUNOMODULATORS
JP2023500395A (en) 2019-11-11 2023-01-05 インサイト・コーポレイション Salts and Crystal Forms of PD-1/PD-L1 Inhibitors
CN111704617B (en) * 2020-06-15 2022-08-23 嘉兴特科罗生物科技有限公司 Small molecule compound
CN116437913A (en) * 2020-09-23 2023-07-14 艾其林医药公司 Pharmaceutical compounds for treating complement-mediated disorders
CR20230230A (en) 2020-11-06 2023-07-27 Incyte Corp Process for making a pd-1/pd-l1 inhibitor and salts and crystalline forms thereof
WO2022099018A1 (en) 2020-11-06 2022-05-12 Incyte Corporation Process of preparing a pd-1/pd-l1 inhibitor
WO2022099075A1 (en) 2020-11-06 2022-05-12 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
WO2023051793A1 (en) * 2021-09-30 2023-04-06 武汉朗来科技发展有限公司 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

Family Cites Families (62)

* 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
US6653340B1 (en) 1997-06-03 2003-11-25 Biocryst Pharmaceuticals, Inc. Compounds useful in the complement, coagulat and kallikrein pathways and method for their preparation
WO1999048492A1 (en) 1998-03-26 1999-09-30 Japan Tobacco Inc. Amide derivatives and nociceptin antagonists
CZ297264B6 (en) 1998-09-09 2006-10-11 Metabasis Therapeutics, Inc. Novel heteroaromatic inhibitors of fructose 1,6-bisphosphatase
EP1219611A4 (en) 1999-09-03 2003-03-19 Ajinomoto Kk Novel processes for preparing oxazepine derivatives
US7528165B2 (en) 2001-12-13 2009-05-05 National Health Research Institutes Indole compounds
EP1546150B1 (en) 2002-07-16 2009-03-04 Amura Therapeutics Limited Pyrrole derivatives as inhibitors of cyteine proteases
AU2003302084A1 (en) 2002-11-15 2004-06-15 Bristol-Myers Squibb Company 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
DK1713503T3 (en) 2004-02-10 2013-11-04 Univ Colorado Regents INHIBITION OF FACTOR B, THE ALTERNATIVE COMPLEMENT PATHWAY AND RELATED PROCEDURE
RU2393154C2 (en) 2004-03-24 2010-06-27 Йерини Аг Novel antiogenesis inhibiting compounds and use thereof
US7417063B2 (en) 2004-04-13 2008-08-26 Bristol-Myers Squibb Company Bicyclic heterocycles useful as serine protease inhibitors
EP1814576A2 (en) 2004-07-20 2007-08-08 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
JPWO2008047831A1 (en) 2006-10-17 2010-02-25 協和発酵キリン株式会社 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)
JP5758298B2 (en) 2009-10-16 2015-08-05 持田製薬株式会社 Nonalcoholic steatohepatitis related marker
EP2558446B1 (en) 2010-04-16 2019-06-12 AC Immune S.A. Novel compounds for the treatment of diseases associated with amyloid or amyloid-like proteins
US9085555B2 (en) * 2011-01-04 2015-07-21 Novartis Ag Complement pathway modulators and uses thereof
CA2877474A1 (en) * 2011-06-20 2012-12-27 Myrexis, Inc. Compounds and therapeutic uses thereof
AU2012272706B2 (en) 2011-06-22 2017-07-06 Apellis Pharmaceuticals, Inc. Methods of treating chronic disorders with complement inhibitors
NZ742005A (en) 2012-05-03 2019-04-26 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
CN104603126B (en) 2012-06-28 2017-05-31 诺华股份有限公司 Pyrrolidin derivatives and its purposes as complement pathway conditioning agent
WO2014002054A1 (en) * 2012-06-28 2014-01-03 Novartis Ag Pyrrolidine derivatives and their use as complement pathway modulators
ES2712190T3 (en) 2012-06-28 2019-05-09 Novartis Ag Modulators of the complement pathway and its uses
CN105121429B (en) 2012-06-28 2017-12-12 诺华股份有限公司 Complement pathway conditioning agent and its purposes
EP2867229B1 (en) 2012-06-28 2017-07-26 Novartis AG Pyrrolidine derivatives and their use as complement pathway modulators
CA2876993A1 (en) * 2012-06-28 2014-01-03 Novartis Ag Pyrrolidine derivatives and their use as complement pathway modulators
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
ITMI20121156A1 (en) * 2012-06-29 2013-12-30 Consiglio Nazionale Ricerche METHOD OF PROCESSING IMAGES OF TOMOGRAPHY WITH OPTICAL CONSISTENCY
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
ES2687983T3 (en) 2012-07-12 2018-10-30 Novartis Ag Modulators of the complement path and uses 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
CA2899034A1 (en) 2013-01-23 2014-07-31 Musc Foundation For Research Development 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
RU2016137524A (en) 2014-02-25 2018-04-02 Ачиллион Фармасьютикалс, Инк. AMIDIC COMPOUNDS FOR TREATMENT OF COMPLETE-MEDIATED DISORDERS
AR106018A1 (en) 2015-08-26 2017-12-06 Achillion Pharmaceuticals Inc ARYL, HETEROARYL AND HETEROCYCLIC 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
WO2017035352A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Carbamate, ester, and ketone compounds for treatment of medical disorders
US10000516B2 (en) 2015-08-26 2018-06-19 Achillion Pharmaceuticals, Inc. Phosphonate 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
AR105809A1 (en) 2015-08-26 2017-11-08 Achillion Pharmaceuticals Inc COMPOUNDS FOR THE 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
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
EP3340983B1 (en) 2015-08-26 2023-10-04 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic 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
WO2017035417A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Phosphonate compounds for treatment of immune and inflammatory disorders
AR105808A1 (en) 2015-08-26 2017-11-08 Achillion Pharmaceuticals Inc AMIDA COMPOUNDS FOR THE TREATMENT OF MEDICAL DISORDERS
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
WO2017035355A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Ether compounds for treatment of medical disorders
WO2017035415A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Alkyne compounds for treatment of immune and inflammatory disorders
EP3340982B1 (en) 2015-08-26 2021-12-15 Achillion Pharmaceuticals, Inc. 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
WO2017035405A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of immune and inflammatory disorders
CA3007922A1 (en) 2015-12-11 2017-06-15 Lifesci Pharmaceuticals, Inc. Therapeutic inhibitory compounds
WO2017127761A1 (en) 2016-01-20 2017-07-27 Vitrisa Therapeutics, Inc. Compositions and methods for inhibiting factor d

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11926617B2 (en) 2015-08-26 2024-03-12 Achillion Pharmaceuticals, Inc. Aryl, heteroaryl, and heterocyclic compounds for treatment of immune and inflammatory disorders

Also Published As

Publication number Publication date
KR20160116018A (en) 2016-10-06
US9732104B2 (en) 2017-08-15
CA2940775A1 (en) 2015-09-03
JP2017506672A (en) 2017-03-09
PL3110418T3 (en) 2020-05-18
US10550140B2 (en) 2020-02-04
MX2016011034A (en) 2017-03-03
MX2016011036A (en) 2017-02-28
IL285114A (en) 2021-08-31
US20160362433A1 (en) 2016-12-15
EP3110423A4 (en) 2017-09-20
IL285114B2 (en) 2023-08-01
US10100072B2 (en) 2018-10-16
JP6688352B2 (en) 2020-04-28
JP2020121979A (en) 2020-08-13
JP2017506269A (en) 2017-03-02
HRP20200182T1 (en) 2020-05-01
JP2017507184A (en) 2017-03-16
RU2016137524A (en) 2018-04-02
US9758537B2 (en) 2017-09-12
US9663543B2 (en) 2017-05-30
IL247449A0 (en) 2016-11-30
US20150239919A1 (en) 2015-08-27
WO2015130842A2 (en) 2015-09-03
US10106563B2 (en) 2018-10-23
EP3110804A4 (en) 2017-11-22
PT3110418T (en) 2020-01-22
US10301336B2 (en) 2019-05-28
US20150239868A1 (en) 2015-08-27
RU2016137678A (en) 2018-04-02
US20180291046A1 (en) 2018-10-11
WO2015130806A1 (en) 2015-09-03
EP3110416A1 (en) 2017-01-04
KR20160118368A (en) 2016-10-11
US20150239920A1 (en) 2015-08-27
RU2707749C2 (en) 2019-11-29
US10370394B2 (en) 2019-08-06
IL285114B1 (en) 2023-04-01
KR102366025B1 (en) 2022-02-21
EP3110804A1 (en) 2017-01-04
RU2016137677A (en) 2018-04-02
US20160362432A1 (en) 2016-12-15
CN106413707A (en) 2017-02-15
CA2940645A1 (en) 2015-09-03
AP2016009434A0 (en) 2016-09-30
CN106456605A (en) 2017-02-22
CA2940769A1 (en) 2015-09-03
KR20160116016A (en) 2016-10-06
EA201691728A1 (en) 2017-02-28
JP6948426B2 (en) 2021-10-13
AU2015223068B2 (en) 2020-07-30
CN107073000A (en) 2017-08-18
IL269806A (en) 2019-11-28
US10428094B2 (en) 2019-10-01
US10689409B2 (en) 2020-06-23
US20190085005A1 (en) 2019-03-21
JP2017508788A (en) 2017-03-30
EP3110418A4 (en) 2017-08-02
US20170226142A1 (en) 2017-08-10
CN106458981A (en) 2017-02-22
US10005802B2 (en) 2018-06-26
WO2015130830A9 (en) 2016-06-23
EP3110805A1 (en) 2017-01-04
AP2016009435A0 (en) 2016-09-30
MX2016011037A (en) 2017-02-28
JP6400738B2 (en) 2018-10-03
CA2940777A1 (en) 2015-09-03
EP3110419A4 (en) 2017-07-26
US9732103B2 (en) 2017-08-15
EA201691730A1 (en) 2017-01-30
WO2015130795A1 (en) 2015-09-03
US10189869B2 (en) 2019-01-29
US20180298043A1 (en) 2018-10-18
CY1122550T1 (en) 2021-01-27
EP3110417A1 (en) 2017-01-04
IL301427A (en) 2023-05-01
US10087203B2 (en) 2018-10-02
AU2020260434B2 (en) 2022-02-17
AU2015223068A1 (en) 2016-09-01
US20150239894A1 (en) 2015-08-27
CA2940774A1 (en) 2015-09-03
ES2771950T3 (en) 2020-07-07
WO2015130830A1 (en) 2015-09-03
EP3110805B1 (en) 2019-12-11
EP3623367A1 (en) 2020-03-18
WO2015130854A1 (en) 2015-09-03
WO2015130845A9 (en) 2016-06-30
CN106456604A (en) 2017-02-22
AU2015223072A1 (en) 2016-09-01
JP7374967B2 (en) 2023-11-07
JP2021193128A (en) 2021-12-23
RU2703995C2 (en) 2019-10-23
EP3110419A1 (en) 2017-01-04
US20190185498A1 (en) 2019-06-20
IL269806B (en) 2021-08-31
CA2940772A1 (en) 2015-09-03
EA035501B1 (en) 2020-06-25
EP3623367B1 (en) 2022-09-28
JP2018199714A (en) 2018-12-20
US20190023729A1 (en) 2019-01-24
IL247450A0 (en) 2016-11-30
WO2015130845A1 (en) 2015-09-03
JP6537532B2 (en) 2019-07-03
LT3110418T (en) 2020-02-25
US9695205B2 (en) 2017-07-04
US20150239838A1 (en) 2015-08-27
US20180022767A1 (en) 2018-01-25
EP3110418A1 (en) 2017-01-04
MX2020004950A (en) 2021-10-26
ES2933956T3 (en) 2023-02-15
US9828396B2 (en) 2017-11-28
US9796741B2 (en) 2017-10-24
KR20160116014A (en) 2016-10-06
US20180072762A1 (en) 2018-03-15
US20180022766A1 (en) 2018-01-25
SA516371744B1 (en) 2020-11-26
KR20160116017A (en) 2016-10-06
AP2016009438A0 (en) 2016-09-30
EA201691727A1 (en) 2017-01-30
AU2015223132A1 (en) 2016-09-01
US20160362398A1 (en) 2016-12-15
AU2015223084A1 (en) 2016-09-01
US10081645B2 (en) 2018-09-25
KR20160119241A (en) 2016-10-12
AP2016009437A0 (en) 2016-09-30
US20170189410A1 (en) 2017-07-06
US20190031692A1 (en) 2019-01-31
US20160361329A1 (en) 2016-12-15
CN112250673A (en) 2021-01-22
CN111437278A (en) 2020-07-24
HUE047295T2 (en) 2020-04-28
RU2016137677A3 (en) 2018-10-19
US20170260219A1 (en) 2017-09-14
RS59854B1 (en) 2020-02-28
EP3110417A4 (en) 2017-07-26
KR102383714B1 (en) 2022-04-07
US20150239895A1 (en) 2015-08-27
EP4129407A1 (en) 2023-02-08
IL247450B (en) 2019-10-31
MX2016011038A (en) 2017-05-12
CN106414427A (en) 2017-02-15
US20150239893A1 (en) 2015-08-27
EP3110423A2 (en) 2017-01-04
RU2016137832A (en) 2018-04-02
AU2015223084B2 (en) 2019-11-07
AU2020260434A1 (en) 2020-11-26
US20150239921A1 (en) 2015-08-27
IL247453A0 (en) 2016-11-30
US20190144473A1 (en) 2019-05-16
IL247454A0 (en) 2016-11-30
RU2016137832A3 (en) 2018-10-29
CN106456604B (en) 2020-11-06
KR20220047666A (en) 2022-04-18
CN106458981B (en) 2019-09-10
ZA201605829B (en) 2019-01-30
EP3110805A4 (en) 2017-07-26
DK3110418T3 (en) 2020-02-10
EA201691725A1 (en) 2017-01-30
US10464956B2 (en) 2019-11-05
EP3110806A1 (en) 2017-01-04
AP2016009436A0 (en) 2016-09-30
SI3110418T1 (en) 2020-03-31
JP2017511815A (en) 2017-04-27
WO2015130842A3 (en) 2016-04-07
US9598446B2 (en) 2017-03-21
EA201691731A1 (en) 2017-02-28
US20190048033A1 (en) 2019-02-14
US20200062790A1 (en) 2020-02-27
WO2015130784A1 (en) 2015-09-03
ZA201605832B (en) 2020-05-27
US9643986B2 (en) 2017-05-09
BR112016019526A2 (en) 2017-08-15
IL247452B (en) 2019-09-26
EP3110806A4 (en) 2017-11-22
WO2015130838A1 (en) 2015-09-03
MX2016011035A (en) 2017-03-09
IL247452A0 (en) 2016-11-30
US20170298084A1 (en) 2017-10-19
US20170298085A1 (en) 2017-10-19
US10428095B2 (en) 2019-10-01
WO2015130842A9 (en) 2016-05-19
AU2015223075A1 (en) 2016-09-01
US20160362399A1 (en) 2016-12-15
EP3110416A4 (en) 2017-07-26
WO2015130806A9 (en) 2016-08-11
US20150239837A1 (en) 2015-08-27
AU2015223121A1 (en) 2016-09-01
BR112016019526B1 (en) 2023-03-28
EA032690B1 (en) 2019-07-31
JP2017508789A (en) 2017-03-30
US20180030075A1 (en) 2018-02-01
EP3110418B1 (en) 2019-11-06
US10253053B2 (en) 2019-04-09

Similar Documents

Publication Publication Date Title
US10464956B2 (en) Aryl, heteroaryl, and heterocyclic compounds for treatment of complement mediated disorders

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION