WO2018118829A1 - Heterocyclic compounds as hiv protease inhibitors - Google Patents

Heterocyclic compounds as hiv protease inhibitors Download PDF

Info

Publication number
WO2018118829A1
WO2018118829A1 PCT/US2017/067163 US2017067163W WO2018118829A1 WO 2018118829 A1 WO2018118829 A1 WO 2018118829A1 US 2017067163 W US2017067163 W US 2017067163W WO 2018118829 A1 WO2018118829 A1 WO 2018118829A1
Authority
WO
WIPO (PCT)
Prior art keywords
imino
dihydro
carbonyl
benzyl
pyrrolo
Prior art date
Application number
PCT/US2017/067163
Other languages
French (fr)
Inventor
Brian Mckittrick
John P. Caldwell
John A. Mccauley
Henry Vaccaro
Tin-Yau Chan
Hyunjin Kim
Elizabeth Smith
Liwu Hong
Tanweer Khan
Shihong Ying
Hongwu Wang
Peter D. Williams
Jae-Hun Kim
M. Brad Nolt
Mary Ann Caplen
Original Assignee
Merck Sharp & Dohme Corp.
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 Merck Sharp & Dohme Corp. filed Critical Merck Sharp & Dohme Corp.
Priority to EP17883610.2A priority Critical patent/EP3558305A4/en
Priority to US16/461,899 priority patent/US20200069691A1/en
Publication of WO2018118829A1 publication Critical patent/WO2018118829A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • A61K31/41Heterocyclic 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
    • A61K31/41881,3-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
    • A61K31/42Oxazoles
    • A61K31/424Oxazoles condensed with heterocyclic ring systems, e.g. clavulanic acid
    • 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
    • A61K31/425Thiazoles
    • A61K31/429Thiazoles 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
    • A61K31/435Heterocyclic 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
    • A61K31/4355Heterocyclic 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 oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic 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
    • A61K31/535Heterocyclic 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
    • A61K31/53831,4-Oxazines, e.g. morpholine ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic 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
    • 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
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/88Nitrogen atoms, e.g. allantoin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D403/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • 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
    • C07D413/10Heterocyclic 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 aromatic 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
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/14Ortho-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
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • Rl heterocyclyl is unsubstituted or substituted pyrrolidinyl.
  • Rl heterocyclyl is unsubstituted or substituted
  • R2 is selected from the group consisting of CH3, CH2CH3, (CH2) n CH(CH3)2, (CH2) n OCH(CH3)2, (CH2)nC3-6cycloalkyl wherein the cycloalkyl is cyclopropyl, cyclobutyl or cyclopentyl, or (CHR) n C 6 -10phenyl.
  • R2 is hydrogen
  • R3 and R4 are independently selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, pyridyl, pyranyl, (CH2)ntetrahydropyranyl, and
  • R4 is optionally substituted phenyl and R3 is selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, said isobutyl, isopentyl, cyclopropyl and phenyl optionally substituted with 1 to 3 groups of Rd.
  • R3 is optionally substituted phenyl.
  • C(O) refers to carbonyl.
  • S(O) refers to sulfinyl.
  • the invention includes all possible enantiomers and diastereomers and mixtures of two or more stereoisomers, for example mixtures of enantiomers and/or diastereomers, in all ratios.
  • enantiomers are a subject of the invention in enantiomerically pure form, both as levorotatory and as dextrorotatory antipodes, in the form of racemates and in the form of mixtures of the two enantiomers in all ratios.
  • the invention includes both the cis form and the trans form as well as mixtures of these forms in all ratios.
  • Additional embodiments of the invention include the pharmaceutical compositions, combinations and methods set forth in (a)-(r) above and the uses (i)(a)-(e) through (iii)(a)-(e) set forth in the preceding paragraph, wherein the compound of the present invention employed therein is a compound of one of the embodiments, aspects, classes or subclasses described above. In all of these embodiments, the compound can optionally be used in the form of a pharmaceutically acceptable salt.
  • TPP triphenylphosphine
  • the compounds of formula I can be prepared using the general synthetic reaction schemes shown in Methods A through C.
  • Method A provides a route to compounds V and then to formula I compounds by first elaborating the monosubstituted amino acid ester II to the requisite disubstitiuted amino ester III.
  • II is reacted with benzaldehyde under dehydrating conditions to provide an intermediate imine which can be treated with a strong base such as LiHMDS or LDA and reacted with an alkylating agent such as a alkyl halide or triflate (R 4 X - X is halide or trifiate) followed by subsequent hydrolysis of the imine to give di-substituted amino acid esters III.

Abstract

The present invention is directed to compounds of Formula I pharmaceutical compositions comprising the same, and their use in the inhibition of HIV protease, the inhibition of HIV replication, the prophylaxis of infection by HIV, the treatment of infection by HIV, and the prophylaxis, treatment, and delay in the onset or progression of AIDS.

Description

TITLE OF THE INVENTION
HETEROCYCLIC COMPOUNDS AS HIV PROTEASE INHIBITORS
BACKGROUND OF THE INVENTION
A retrovirus designated human immunodeficiency virus (HIV), particularly the strains known as HIV type-1 (HIV-1) virus and type-2 (HIV-2) virus, is the etiological agent of acquired immunodeficiency syndrome (AIDS). AIDS is a disease characterized by the destruction of the immune system, particularly of CD4 T-cells, with attendant susceptibility to opportunistic infections, and its precursor AIDS-related complex ("ARC"), a syndrome characterized by symptoms such as persistent generalized lymphadenopathy, fever and weight loss. This virus was previously known as LAV, HTLV-III, or ARV. A common feature of retrovirus replication is the extensive post-translational processing of precursor polyproteins by a virally encoded protease to generate mature viral proteins required for virus assembly and function. Inhibition of this processing prevents the production of normally infectious virus. For example, Kohl et al, Proc. Nat'lAcad. Sci. 1988, 85: 4686, demonstrated that genetic inactivation of the HIV encoded protease resulted in the production of immature, non-infectious virus particles. These results indicated that inhibition of the HIV protease represents a viable method for the treatment of AIDS and the prevention or treatment of infection by HIV.
Nucleotide sequencing of HIV shows the presence of a pol gene in one open reading frame [Ratner et al, Nature 1985, 313: 277]. Amino acid sequence homology provides evidence that the pol sequence encodes reverse transcriptase, an endonuclease, HIV protease and gag, which encodes the core proteins of the virion (Toh et al, EMBO J. 1985, 4: 1267; Power et al, Science 1986, 231 : 1567; Pearl et al, Nature 1987, 329: 351].
Several HIV protease inhibitors are presently approved for clinical use in the treatment of AIDS and HIV infection, including indinavir (see US 5413999), amprenavir (US
5585397), saquinavir (US 5196438), ritonavir (US 5484801) and nelfinavir (US 5484926). Each of these protease inhibitors is a peptide-derived peptidomimetic, competitive inhibitor of the viral protease which prevents cleavage of the HIV gag-pol polyprotein precursor. Tipranavir (US 5852195) is a non-peptide peptidomimetic protease inhibitor also approved for use in treating HIV infection. The protease inhibitors are administered in combination with at least one and typically at least two other HIV antiviral agents, particularly nucleoside reverse transcriptase inhibitors such as zidovudine (AZT) and lamivudine (3TC) and/or non-nucleoside reverse transcriptase inhibitors such as efavirenz and nevirapine. Indinavir, for example, has been found to be highly effective in reducing HIV viral loads and increasing CD4 cell counts in
HIV-infected patients, when used in combination with nucleoside reverse transcriptase inhibitors. See, for example, Hammer et al., New England J. Med. 1997, 337: 725-733 and Gulick et al, New England J. Med. 1997, 337: 734-739. There is a continuing need for new compounds which are capable of treating infectious diseases, in particular, for inhibiting HIV protease and suitable for use in the treatment or prophylaxis of infection by HIV and/or for the treatment or prophylaxis or delay in the onset or progression of AIDS.
SUMMARY OF THE INVENTION
The present invention is directed to heterocyclic derivatives, pharmaceutical compositions comprising the same, and their use in the inhibition of HIV protease, the inhibition of HIV replication, the prophylaxis of infection by HIV, the treatment of infection by HIV, and the prophylaxis, treatment, and delay in the onset or progression of AIDS.
DETAILED DESCRIPTION OF THE INVENTION
The invention encompasses compounds of structural formula I
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof, wherein:
A is selected from the group consisting of (CHR2)pC6-10aryl and (CHR2)pC4-nheteroaryl; R is selected from the group consisting of hydrogen and Ci-6alkyl;
Rx is selected from the group consisting of Ci-6alkyl, Ci-3haloalkyl, halogen, S02Ci-6alkyl, and OCi-6alkyl;
R1 is selected from the group consisting of a bicyclic, monocyclic, or tricyclic C4- 13heterocyclyl, said heterocyclyl optionally substituted with 1 to 3 groups of Ra;
R2 is selected from the group consisting of hydrogen, Ci-6alkyl, Ci-3haloalkyl, halogen, S02Ci-6alkyl, (CH2)nOCi-6alkyl, (CH2)nC3-6cycloalkyl, (CHR)nC6-10aryl, (CHR)nC5- lOheteroaryl, said aryl and heteroaryl optionally substituted with 1 to 3 groups of Ra;
R3 and R4 are independently selected from the group consisting of Ci-6alkyl, (CH2)nCl- 3haloalkyl, (CR2)nC3-6cycloalkyl, (CH2)nC6-10aryl, (CH2)nC5-ioheterocyclyl; said alkyl, aryl, and heterocyclyl optionally substituted with 1 to 3 groups of Rd; R5 is selected from the group consisting of hydrogen, Ci-6alkyl, C(0)OR, C3-6cycloalkyl, S02R, O(CH2)nC6-10aryl, and (CH2)nC6-10aryl;
Ra is selected from the group consisting of (CH2)nC5-l lheterocyclyl, Ci-6alkyl, C2-6alkenyl, C(0)Ci-6alkyl, OCi -6alkyl, =0, Ci-3haloalkyl, OCi-3haloalkyl, C3-6cycloalkyl, C(0)C3- 6cycloalkyl, halogen, CN, SCi-6alkyl, S02Cl -6alkyl, (CH2)nC6-12aryl, OC6-12aryl, C(0)C5- 12heterocyclyl, C(0)OR, C(0)OC2-6alkenyl, C(0)NR2, NR2, NHC(0)OR, -NHC3- 6cycloalkyl, -NR(CH2)C3-6cycloalkyl, NHC5-ioheterocyclyl, NHC6-10aryl,
(CH2)nNHC(0)R, said alkyl, alkenyl, aryl, and heterocyclyl optionally substituted with 1 to 3 groups of Rb;
Rb is selected from the group consisting of Ci-6alkyl, Cl -6alkylOR, OR, =0, C(0)R, SOCl - 6alkyl, S02Ci-6alkyl, S02N(R)2, NRR5, Ci-3haloalkyl, OCi-3haloalkyl, C3-6cycloalkyl, halogen, (CH2)nCN, (CH2)nC6-12aryl, and (CH2)nC5-ioheteroaryl, said alkyl, aryl and heteroaryl optionally substituted with 1 to 3 groups of Rc;
Rc is selected from the group consisting of Ci-6alkyl, Ci-6alkylOR, OR, and halogen;
Rd is selected from the group consisting of Ci-6alkyl, Ci-3haloalkyl, CN, C(0)NR2, C5- loheteroaryl, C6-10aryl, and halogen, said heteroaryl, alkyl and aryl optionally substituted with 1 to 3 groups of halogen and CN; n is 0, 1, 2, 3, or 4; p is O or l ; and q is 0 or 1.
An embodiment of the invention of formula I is realized when A is (CHR2)pC6- loaryl. An embodiment of the invention of formula I is realized when A is aryl and p=l. A subembodiment of this aspect of the invention is realized when A is an aryl selected from the group consisting of phenyl, tetrahydronaphthalenyl, dihydroindenyl, and
tetrahydrobenzoannulenyl. Another subembodiment of this aspect of the invention is realized when A is phenyl and p=l . Another subembodiment of this aspect of the invention is realized when A is tetrahydronaphthalenyl and p=0. Still another subembodiment of this aspect of the invention is realized when A is dihydroindenyl and p=0 . Yet another subembodiment of this aspect of the invention is realized when A is tetrahydrobenzoannulenyl and p=0. Another embodiment of the invention of formula I is realized when A is
(CHR2)pC 5-i iheteroaryl. Another embodiment of the invention of formula I is realized when A is (CHR2)pC5-iiheteroaryl and p=l . Another embodiment of the invention of formula I is realized when A is (CHR2)pC9-nheteroaryl and p=0. A subembodiment of this aspect of the invention is realized when A is a heteroaryl selected from the group consisting of pyridyl, thiazolyl, thiophenyl, dihydrochromenyl, and dihydrothiochromenyl. Another subembodiment of this aspect of the invention is realized when A is pyridyl. Another subembodiment of this aspect of the invention is realized when A is thiazolyl. Still another subembodiment of this aspect of the invention is realized when A is thiophenyl. Yet another subembodiment of this aspect of the invention is realized when A is dihydrochromenyl. Yet another subembodiment of this aspect of the invention is realized when A is dihydrothiochromenyl. Another embodiment of the invention of formula I is realized when A is is C5-i iheteroaryl which is pyridyl and p=l
Another embodiment of the invention of formula I is realized when Rl is (CH2)nC4-l lheterocyclyl, said heterocyclyl unsubstituted or substituted with 1 to 3 groups of
Ra. A subembodiment of this aspect of the invention is realized when Rl is unsubstituted (CH2)nC4-l lheterocyclyl. A subembodiment of this aspect of the invention is realized when Rl is substituted (CH2)nC4-H heterocyclyl. A subembodiment of this aspect of the invention is realized when the unsubstituted or substituted Rl is (CH2)nC4-l lheterocyclyl is linked to the C(O) group in formula I via nitrogen atom. Another subembodiment of this aspect of the invention is realized when the heterocyclyl is a bicyclic ring having at least one nitrogen atom. Still another subembodiment of this aspect of the invention is realized when the heterocyclyl is a bicyclic ring having two to four nitrogen atoms. Another subembodiment of this aspect of the invention is realized when the heterocyclyl is selected from the group consisting of substituted or unsubstituted dihydropyrrolopyrazinyl, dihydropyrrolopyrimidinyl, dihydrotriazolopyrazinyl, piperazinyl, piperazinonyl, piperidinyl, hexahydrooxazolopyrazinonyl,
tetrahydropyrazinoindolyl, tetrahydrobenzapinyl, dihydropyrrolooxazolyl,
tetrahydropyrrolopyrrolidione, pyrrolidinyl, hexahydroisoxazolyl, tetrahydropyrazolopyridyl, azetindinyl, tetrahydropyrrolotriazolooxazinyl, tetrahydropyrroloisoxazolyl,
tetrahydrofuropyridinyl, dihydroisoindolyl, dihydropyrrolopyrazolyl, tetrahydropyrrolothiazolyl, tetrahydroimidazopyrazinyl, dihydropyrrolopyridinyl, dihydropyrroloimidazolyl,
dihydroisoindolopyridinyl, dihydroisoquinolinyl, dihydronaphthyridinyl, and
dihydroimidazopyrazinyl.
Another subembodiment of this aspect of the invention is realized when the heterocyclyl is selected from unsubstituted or substituted dihydropyrrolopyrazinyl, piperazinyl, piperazinonyl, pyrrolidinyl, dihydropyrrolopyrimidinyl, dihydrotriazolopyrazinyl, and dihydroisoindolyl. Another subembodiment of this aspect of the invention is realized when the Rl heterocyclyl is unsubstituted or substituted dihydropyrrolopyrazinyl. Another subembodiment of this aspect of the invention is realized when the Rl heterocyclyl is unsubstituted or substituted piperazinyl. Another subembodiment of this aspect of the invention is realized when the Rl heterocyclyl is unsubstituted or substituted piperazinonyl.
Another subembodiment of this aspect of the invention is realized when the Rl heterocyclyl is unsubstituted or substituted pyrrolidinyl. Another subembodiment of this aspect of the invention is realized when the Rl heterocyclyl is unsubstituted or substituted
dihydropyrrolopyrimidinyl.
Another subembodiment of this aspect of the invention is realized when the Rl heterocyclyl is unsubstituted or substituted dihydrotriazolopyrazinyl. Another subembodiment of this aspect of the invention is realized when the Rl heterocyclyl is unsubstituted or substituted dihydroisoindolyl.
Another embodiment of the invention of formula I is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra selected from the group consisting of Ci-6alkyl, OCi-6alkyl, Ci-3haloalkyl, OCi-3haloalkyl, halogen, CN, SCi-
6alkyl, S02Ci-6alkyl, C(0)OR, C(0)NR2, NR2, and NHC(0)OR, said alkyl, aryl, and heterocyclyl optionally substituted with 1 to 3 groups of Rb. A subembodiment of this aspect of the invention is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra selected from the group consisting of CH3, CH2CH3, OCH3, CF3, OCF2,
CH2CF3, fluoro, chloro, bromo, C(0)CH3, SCH3, SO2CH3, CN, COOCH3, COOCH2CH3, NHC(0)OCH3, NHC(0)CH3, CON(CH3)2, CONHCH3, CONHCH(CH3)2, N¾, NHCH3, N(CH3)2, and NHCH(CH3)2. A subembodiment of this aspect of the invention is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra selected from the group consisting of CH3, OCH3, CF3, fluoro, chloro, SO2CH3, CN, COOCH3, NHC(0)OCH3, CON(CH3)2, CONHCH3, CONHCH(CH3)2, N(CH3)2, and NHCH(CH3)2. Still another subembodimemnt of this aspect of the invention is realized when the heterocyclyl of is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra selected from the group consisting of CH3, OCH3, CF3, fluoro, chloro, SO2CH3, CN, COOCH3, and NHC(0)OCH3.
Another subembodiment of this aspect of the invention is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra selected from the group consisting (CH2)nC6-12aryl, (CH2)nC5-i2heterocyclyl, C(0)C5-i2heterocyclyl, -NHC3- 6cycloalkyl, -NR(CH2)C3-6cycloalkyl, NHC5-ioheterocyclyl, and NHC6-10aryl, said heterocyclyl, cycloalkyl and aryl unsubstituted or substituted with 1 to 3 groups of Rb.
A further subembodiment of this aspect of the invention is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra wherein Ra is unsubstituted or substituted (CH2)nC6-12aryl or NHC6-10aryl. A subembodiment of this aspect of the invention is realized when the aryl is unsubstituted or substituted phenyl. A further subembodiment of this aspect of the invention is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra wherein Ra is unsubstituted or substituted (CH2)nC5-i2heterocyclyl, NHC5-ioheterocyclyl, or C(0)C5- 12heterocyclyl. A subembodiment of this aspect of the invention is realized when the heterocyclyl is selected from the group consisting of unsubstituted or substituted pyrazolyl, pyridinyl, indazolyl, pyrrolyl, triazolyl, indolyl, pyrimidinyl, thiophenyl,
tetrahydropyrazolopyridinyl, triazolopyridinyl, dihydropyrrolopyrazolyl, dihydropyridooxazinyl, isoquinolyl, isoxazolyl, dihydropyrrolyl, benzisoxazolyl, thiomorpholinyl, oxadiazolyl, pyrrolodinyl, oxazolyl, oxophenylimidazolidinyl, dihydroimidazopyridinone, furanyl, dihydrobenzimidazolone, and benzoxazolone.
Another subembodiment of this aspect of the invention is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra selected from the group consisting of unsubstituted or substituted pyrazolyl, pyridinyl, pyrimidinyl,
dihydropyrrolopyrazolyl, pyrrolodinyl, dihydroimidazopyridinone, and dihydrobenzimidazolone.
Another subembodiment of this aspect of the invention is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra wherein Ra is unsubstituted or substituted pyrazolyl.
Another subembodiment of this aspect of the invention is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra wherein Ra is unsubstituted or substituted pyridinyl.
Another subembodiment of this aspect of the invention is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra wherein Ra is unsubstituted or substituted pyrimidinyl.
Another subembodiment of this aspect of the invention is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra wherein Ra is unsubstituted or substituted dihydropyrrolopyrazolyl.
Another subembodiment of this aspect of the invention is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra wherein Ra is unsubstituted or substituted pyrrolodinyl.
Another subembodiment of this aspect of the invention is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra wherein Ra is unsubstituted or substituted dihydroimidazopyridinone.
Another subembodiment of this aspect of the invention is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra wherein Ra is unsubstituted or substituted dihydrobenzimidazolone.
A further subembodiment of this aspect of the invention is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra wherein Ra is unsubstituted or substituted -NHC3-6cycloalkyl or -NR(CH2)C3-6cycloalkyl. A subembodiment of this aspect of the invention is realized when the heterocyclyl of Rl is substituted with 0 to 1, 1 to 2, or 2 to 3 groups of Ra wherein Ra is cycloalkyl selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
An embodiment of the invention of formula I is realized when R2 is hydrogen. Another embodiment of the invention of formula I is realized when R2 is Ci-
6alkyl or (CH2)nOCi-6alkyl. A subembodiment of this aspect of the invention is realized when R2 is Ci-6alkyl. Another subembodiment of this aspect of the invention is realized when R2 is (CH2)nOCi-6alkyl. Another subembodiment of this aspect of the invention is realized when the alkyl is selected from the group consisting of CH3, CH2CH3, (CH2)nCH(CH3)2, and
(CH2)nOCH(CH3)2.
Another embodiment of the invention of formula I is realized when R2 is (CH2)nC3-6cycloalkyl. A subembodiment of this aspect of the invention is realized when the cycloalkyl is selected from the group consisting of cyclopropyl, cyclobutyl and cyclopentyl.
Another embodiment of the invention of formula I is realized when R2 is (CHR)nC6-10aryl. A subembodiment of this aspect of the invention is realized when the aryl is phenyl.
Another embodiment of the invention of formula I is realized when R2 is (CHR)nC5-ioheteroaryl. A subembodiment of this aspect of the invention is realized when the heteroaryl is pyrrollidinone.
An embodiment of the invention of formula I is realized when one of R3 and R4 is optionally substituted Ci-6alkyl, (CH2)nCl-3haloalkyl, or (CR2)nC3-6cycloalkyl and the other is (CH2)nC6-10aryl, (CH2)nC5-10heterocyclyl; said alkyl, aryl, and heterocyclyl optionally substituted with 1 to 3 groups of Rd.
Another embodiment of the invention of formula I is realized when R3 and R4 are independently selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3,
(CH2)ncyclopropyl, phenyl, pyridyl, pyranyl, (CH2)ntetrahydropyranyl, and
(CH2)ntetrahydrofuranyl, said isobutyl, isopentyl, cyclopropyl, phenyl, pyridyl, pyranyl, tetrahydropyranyl, and tetrahydrofuranyl optionally substituted with 1 to 3 groups of Rd. A subembodiment of this aspect of the invention is realized when R3 and R4 are independently selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, and phenyl, said isobutyl, isopentyl and phenyl optionally substituted with 1 to 3 groups of Rd. Another subembodiment of this aspect of the invention is realized when one of R3 and R4 is optionally substituted phenyl. Another subembodiment of this aspect of the invention is realized when both of R3 and R4 are optionally substituted phenyl. Another subembodiment of this aspect of the invention is realized when R4 is optionally substituted phenyl. Still another subembodiment of this aspect of the invention is realized when R4 is optionally substituted phenyl and R3 is selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, said isobutyl, isopentyl, cyclopropyl and phenyl optionally substituted with 1 to 3 groups of Rd. Still another subembodiment of this aspect of the invention is realized when R3 is optionally substituted phenyl and R4 is selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, said isobutyl, isopentyl, cyclopropyl and phenyl optionally substituted with 1 to 3 groups of Rd.
Another embodiment of the invention of formula I is realized when Rb is selected from the group consisting of (CH2)2CH3, CH2OCH3, (CH2)20H, CH3, (CH2)CH(CH3)2, OCH3, C(0)CH3, (CH2)nCN, N(CH3>2, NHSO2CH3, SOCH3, S02N(CH3)2, and halogen. A subembodiment of this aspect of the invention is realized when Rb is selected from the group consisting of (C¾) 2CH3, CH3, (CH2)CH(CH3)2, fluoro, and chloro.
Yet another embodiment of the invention of formula I is realized when Rb is selected from the group consisting of (CH2)nC6-12aryl, and (CH2)nC5-10heteroaryl, said aryl and heteroaryl optionally substituted with 1 to 3 groups of Rc. A subembodiment of this aspect the invention is realized when aryl and heteroaryl of Rb is selected from the group consisting of phenyl, piperazinyl, pyrazolyl, and triazolyl, said phenyl, piperizinyl, pyrazolyl, and triazolyl optionally substituted with 1 to 3 groups of Rc. A subembodiment of this aspect the invention is realized when aryl of Rb is optionally substituted phenyl. A subembodiment of this aspect the invention is realized when aryl of Rb is optionally substituted piperazinyl. A subembodiment of this aspect the invention is realized when aryl of Rb is optionally substituted pyrazolyl. A subembodiment of this aspect the invention is realized when aryl of Rb is optionally substituted triazolyl.
Another embodiment of the invention of formula I is realized when Rc is selected from CH3, fluoro, chloro, and OH.
Another embodiment of the invention of formula I is realized when Rd is selected the group consisting of CF3, fluoro, chloro, bromo, CN, C(0)NH2, C(0)N(CH3)2, phenyl, pyridyl and furanyl, said phenyl, pyridyl and furanyl optionally substituted with 1 to 3 groups of halogen and CN.
Another embodiment of the invention of formula I is realized when Rx is selected from the group consisting of CH3, OCH3, CF3, SO2CH3, fluoro, and chloro.
Another embodiment of the invention of formula I is realized when p is 0 which means a bond is the linking group. Another embodiment of the invention of formula I is realized when p is 1.
Still another embodiment of the invention of formula I is realized when q is 0 which means the Rx group is not present.
Another embodiment of the invention of formula I is realized when q is 1.
Yet another embodiment of the invention of formula I is realized when n is 0.
Another embodiment of the invention of formula I is realized when n is 1. Another embodiment of the invention of formula I is realized when n is 2. Another embodiment of the invention of formula I is realized when n is 3. Another embodiment of the invention of formula I is realized when n is 4.
In another embodiment of this invention are compounds of formula I having structural formula II, or the pharmaceutically acceptable salts thereof,
Figure imgf000010_0001
wherein R2, R35 R45 RA and Rx are as originally described and B, C, D, E, respectively, are selected from:
a) N=C-C=N;
b) C=N-C=N;
c) C=C-C=C;
d) N=C-C=C;
e) C=C-N=C; and
f) C=C-C=N.
An embodiment of the invention of formula II is realized when B, C, D, and E, respectively, is a) N=C-C=N.
Another embodiment of the invention of formula II is realized when B, C, D, and E, respectively is b) C=N-C=N.
Another embodiment of the invention of formula II is realized when B, C, D, and E, respectively is c) C=C-C=C.
Another embodiment of the invention of formula II is realized when B, C, D, and E, respectively is d) N=C-C=C or g) C=C-C=N.
Another embodiment of the invention of formula II is realized when Ra is 0 and therefore absent.
Another embodiment of the invention of formula II is realized when there is one Ra group, selected from the group consisting of CH3, CH2CH3, OCH3, CF3, OCF2, CH2CF3, fluoro, chloro, bromo, C(0)CH3, SCH3, SO2CH3, CN, COOCH3, COOCH2CH3,
NHC(0)OCH3, NHC(0)CH3, CON(CH3)2, CONHCH3, CONHCH(CH3)2, Ν¾, NHCH3, N(CH3)2, NHCH(CH3)2, unsubstituted or substituted (CH2)nC6-12phenyl, or NHC6-10phenyl, and unsubstituted or substituted (CH2)nC5-i2heterocyclyl, NHC5-ioheterocyclyl, or C(0)C5- 12heterocyclyl, wherein the heterocyclyl is selected from the group consisting of pyrazolyl, pyridinyl, pyrimidinyl, dihydropyrrolopyrazolyl, pyrrolodinyl, dihydroimidazopyridinone, and dihydrobenzimidazolone, wherein when substituted is substituted with 1 to 3 groups of Rb. Another embodiment of the invention of formula II is realized when R2 is selected from the group consisting of CH3, CH2CH3, (CH2)nCH(CH3)2, (CH2)nOCH(CH3)2, (CH2)nC3-6cycloalkyl wherein the cycloalkyl is cyclopropyl, cyclobutyl or cyclopentyl, or (CHR)nC6-10phenyl.
Another embodiment of the invention of formula II is realized when R2 is hydrogen or Ci-6alkyl, R3 and R4 are independently selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, pyridyl, pyranyl,
(CH2)ntetrahydropyranyl, and (CH2)ntetrahydrofuranyl, said isobutyl, isopentyl, cyclopropyl, phenyl, pyridyl, pyranyl, tetrahydropyranyl, and tetrahydrofuranyl optionally substituted with 1 to 3 groups of Rd, and q is 0. A subembodiment of this aspect of the invention of formula II is realized when B, C, D, and E, respectively, is C=C-C=C. A subembodiment of this aspect of the invention of formula II is realized when B, C, D, and E, respectively, is N=C-C=N. Still another subembodiment of this aspect of the invention of formula II is realized when one of R3 and R4 is optionally substituted isobutyl or isopentyl.
Another subembodiment of the invention of formula II is realized when R2 is hydrogen, R3 and R4 are independently selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, pyridyl, pyranyl, (CH2)ntetrahydropyranyl, and (CH2)ntetrahydrofuranyl, said isobutyl, isopentyl, cyclopropyl, phenyl, pyridyl, pyranyl, tetrahydropyranyl, and tetrahydrofuranyl optionally substituted with 1 to 3 groups of Rd, and q is 1. A subembodiment of this aspect of the invention is realized when q is 1 and Rx is selected from the group consisiting of CH3, OCH3, CF3, SO2CH3, fluoro, and chloro. A further subembodiment of this aspect of the invention is realized when Rx is in the para position on the phenyl ring. Still another subembodiment of this aspect of the invention is realized when Rx is CF3 in the para position of the phenyl ring. Yet another subembodiment of this aspect of the invention of formula II is realized when B, C, D, and E, respectively, is C=C-C=C. Another subembodiment of this aspect of the invention of formula II is realized when B, C, D, and E, respectively, is N=C-C=N. Still another subembodiment of this aspect of the invention of formula II is realized when one of R3 and R4 is optionally substituted isobutyl or isopentyl.
Another embodiment of the invention of formula II is realized when R3 and R4 are independently selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, pyridyl, pyranyl, (CH2)ntetrahydropyranyl, and
(CH2)ntetrahydrofuranyl, said isobutyl, isopentyl, cyclopropyl, phenyl, pyridyl, pyranyl, tetrahydropyranyl, and tetrahydrofuranyl optionally substituted with 1 to 3 groups of Rd.
Another subembodiment of this aspect of the invention is realized when one of R3 and R4 is optionally substituted phenyl. Another subembodiment of this aspect of the invention is realized when both of R3 and R4 are optionally substituted phenyl. Another subembodiment of this aspect of the invention is realized when R4 is optionally substituted phenyl. Still another subembodiment of this aspect of the invention is realized when R4 is optionally substituted phenyl and R3 is selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, said isobutyl, isopentyl, cyclopropyl and phenyl optionally substituted with 1 to 3 groups of Rd. Another subembodiment of this aspect of the invention is realized when R3 is optionally substituted phenyl. Still another subembodiment of this aspect of the invention is realized when R3 is optionally substituted phenyl and R4 is selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, said isobutyl, isopentyl, cyclopropyl and phenyl optionally substituted with 1 to 3 groups of Rd.
Another embodiment of the invention of formula I is represented by structural formula III, or the pharmaceutically acceptable salts thereof,:
Figure imgf000012_0001
wherein R2, R35 and R4 are as originally described, and wherein Y is selected from the group consisting of:
Figure imgf000012_0002
wherein
Ra is as originally described,
G is N or CH when— represents a double bond and is CH2 when— represents a single bond; J is N or CH;
represents the point of attachment, and
— represents a single or double bond.
An embodiment of the invention of formula III is realized when Y is A.
An embodiment of the invention of formula III is realized when Y is B.
An embodiment of the invention of formula III is realized when Y is C.
An embodiment of the invention of formula III is realized when Y is D.
An embodiment of the invention of formula III is realized when Y is E
An embodiment of the invention of formula III is realized when Y is F.
Another embodiment of the invention of formula III is realized when Ra for A, B, E and F is 0 or not present. Another embodiment of the invention of formula III is realized when one of Ra is present for A, B, E and F and is selected from the group consisting of CH3, CH2CH3, OCH3, CF3, OCF2, CH2CF3, fluoro, chloro, bromo, C(0)CH3, SCH3, SO2CH3, CN, COOCH3, COOCH2CH3, NHC(0)OCH3, NHC(0)CH3, CON(CH3)2, CONHCH3, CONHCH(CH3)2, NH2, NHCH3, N(CH3)2, NHCH(CH3)2, unsubstituted or substituted (CH2)nC6-12phenyl, or NHC6-10phenyl, and unsubstituted or substituted (CH2)nC5-i2heterocyclyl, NHC5- lOheterocyclyl, or C(0)C5-i2heterocyclyl, wherein the heterocyclyl is pyrazolyl, pyridinyl, pyrimidinyl, dihydropyrrolopyrazolyl, pyrrolodinyl, dihydroimidazopyridinone, or
dihydrobenzimidazolone, wherein when substituted is substituted with 1 to 3 groups of Rb.
Another embodiment of the invention of formula III is realized when R2 is selected from the group consisting of CH3, CH2CH3, (CH2)nCH(CH3)2, (CH2)nOCH(CH3)2, (CH2)nC3-6cycloalkyl wherein the cycloalkyl is cyclopropyl, cyclobutyl or cyclopentyl, or (CHR)nC6-10phenyl.
Another embodiment of the invention of formula III is realized when R2 is hydrogen, R3 and R4 are independently selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, pyridyl, pyranyl, (CH2)ntetrahydropyranyl, and
(CH2)ntetrahydrofuranyl, said isobutyl, isopentyl, cyclopropyl, phenyl, pyridyl, pyranyl, tetrahydropyranyl, and tetrahydrofuranyl optionally substituted with 1 to 3 groups of Rd, and q is
0.
Another subembodiment of the invention of formula III is realized when R2 is hydrogen, R3 and R4 are independently selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, pyridyl, pyranyl, (CH2)ntetrahydropyranyl, and (CH2)ntetrahydrofuranyl, said isobutyl, isopentyl, cyclopropyl, phenyl, pyridyl, pyranyl, tetrahydropyranyl, and tetrahydrofuranyl optionally substituted with 1 to 3 groups of Rd, and q is 1. A subembodiment of this aspect of the invention is realized when q is 1 and Rx is selected from the group consisiting of CH3, OCH3, CF3, SO2CH3, fluoro, and chloro. A further subembodiment of this aspect of the invention is realized when Rx is in the para position on the phenyl ring. Still another subembodiment of this aspect of the invention is realized when Rx is CF3 in the para position of the phenyl ring.
Another embodiment of the invention of formula III is realized when R3 and R4 are independently selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, pyridyl, pyranyl, (CH2)ntetrahydropyranyl, and
(CH2)ntetrahydrofuranyl, said isobutyl, isopentyl, cyclopropyl, phenyl, pyridyl, pyranyl, tetrahydropyranyl, and tetrahydrofuranyl optionally substituted with 1 to 3 groups of Rd.
Another subembodiment of this aspect of the invention is realized when one of R3 and R4 is optionally substituted phenyl. Another subembodiment of this aspect of the invention is realized when both of R3 and R4 are optionally substituted phenyl. Another subembodiment of this aspect of the invention is realized when R4 is optionally substituted phenyl. Still another subembodiment of this aspect of the invention is realized when R4 is optionally substituted phenyl and R3 is selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, said isobutyl, isopentyl, cyclopropyl and phenyl optionally substituted with 1 to 3 groups of Rd. Another subembodiment of this aspect of the invention is realized when R3 is optionally substituted phenyl. Still another subembodiment of this aspect of the invention is realized when R3 is optionally substituted phenyl and R4 is selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, said isobutyl, isopentyl, cyclopropyl and phenyl optionally substituted with 1 to 3 groups of Rd.
All structural Formulas, embodiments and classes thereof described herein include the pharmaceutically acceptable salts of the compounds defined therein. Reference to the compounds of Formula I herein encompasses the compounds of formulas I, II, and III and all embodiments and classes thereof. Reference to the compounds of this invention as those of a specific formula or embodiment, e.g., formula I, II, and III or embodiments thereof, or any other generic structural formula or specific compound described or claimed herein, is intended to encompass the specific compound or compounds falling within the scope of the formula or embodiment, including salts thereof, particularly pharmaceutically acceptable salts, solvates (including hydrates) of such compounds and solvated salt forms thereof, where such forms are possible, unless specified otherwise
The present invention includes each of the Examples desribed herein, and pharmaceutically acceptable salts thereof. The invention also encompasses pharmaceutical compositions comprising an effective amount of a compound of the invention or a
pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
As used herein, the term "alkyl" refers to a straight or branched chain, saturated aliphatic hydrocarbon radical having a number of carbon atoms in the specified range. Thus, for example, "-Ci-6 alkyl" (or "-C1-C6 alkyl") means linear or branched chain alkyl groups, including all isomers, having the specified number of carbon atoms and includes each of the hexyl and pentyl isomers as well as each of n-, iso-, sec- and tert-butyl (butyl, s-butyl, z-butyl, t- butyl; Bu = butyl), n- and z-propyl (Pr = propyl), ethyl (Et) and methyl (Me). As another example, "-C1-4 alkyl" refers to each of n-, iso-, sec- and /-butyl; n- and wo-propyl; ethyl and methyl. As another example, "-C1-3 alkyl" refers to each of w-propyl, z'sopropyl, ethyl and methyl. An alkyl group, when viewed in context within a chemical structure, may be univalent (e.g., when R2a 1S unsubstituted -Ci-6alkyl), bivalent (e.g., when R2a 1S mono-substituted -Ci- 6alkyl), or multi-valent (e.g., when R2a is -Ci-6alkyl having two or more substituents).
The term "halogen" (or "halo") refers to fluorine, chlorine, bromine and iodine (alternatively referred to as fluoro, chloro, bromo, and iodo). Fluoro or chloro are preferred.
"Cycloalkyl" is a cyclized alkyl ring having the indicated number of carbon atoms. Thus, for example, "-C3-6 cycloalkyl" (or "-C3-C6 cycloalkyl") refers to each of cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. The term "haloalkyl" refers to an alkyl group as defined above in which one or more of the hydrogen atoms have been replaced (i.e., substituted) with a halogen (i.e., F, CI, Br and/or I). Thus, for example, "-Ci-6 haloalkyl" (or "-C1-C6 haloalkyl") refers to a -Ci to C6 linear or branched alkyl group as defined above with one or more halogen substituents;
particularly 1-6 halogen substituents; and more particularly 1-3 halogen substituents. The term "fluoroalkyl" has an analogous meaning except that the halogen substituents are restricted to fluoro. Suitable fluoroalkyls include the series -(CH2)0-4CF3 (i.e., trifluoromethyl, 2,2,2- trifluoroethyl, 3,3,3-trifluoro-n-propyl, etc.). A fluoroalkyl of particular interest is CF3.
The term "C(O)" refers to carbonyl. The terms "S(0)2" and "SO2" each refer to sulfonyl. The term "S(O)" refers to sulfinyl.
As used herein =0 includes oxo (e.g., an annular -CH- substituted with oxo is -C(O) or carbonyl).
The term "aryl," by itself or as part of another substituent, means an aromatic cyclic hydrocarbon radical. Preferred aryl groups have from six to ten carbons atoms. The term "aryl" includes multiple ring systems as well as single ring systems. Preferred aryl groups for use in the invention include phenyl and naphthyl.
The term "aryl" also includes fused cyclic hydrocarbon rings which are partially aromatic (i.e., one of the fused rings is aromatic and the other is non-aromatic). An exemplary aryl group which is partially aromatic is indanyl.
The term heterocyclyl, heterocycle or heterocyclic, as used herein, represents a stable 4- to 7-membered monocyclic, stable 8- to 11-membered bicyclic heterocyclic, or 8- to 13 tricyclic heterocyclic ring which is either saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O, and S, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure. The term heterocyclyl, heterocycle or heterocyclic includes heteroaryl moieties and heterocycloalkyl moieties. Examples of such heterocyclic elements include, but are not limited to, azepinyl, benzodioxolyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzotriazolyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydroisobenzofuranyl, dihydrobenzothienyl, dihydrobenzothiopyranyl,
dihydrobenzothiopyranyl sulfone, 1,3-dioxolanyl, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, 2-oxopiperazinyl, 2-oxopiperdinyl, 2-oxopyrrolidinyl, piperidyl, piperazinyl, pyridyl, pyrazinyl, pyrazolidinyl, pyrazolyl, pyrazolopyridinyl, pyridazinyl, pyrimidinyl, pyrrolidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiazolyl, thiazolinyl, thienofuryl, thienothienyl, thienyl, and triazolyl.
The term "heteroaryl", as used herein except where noted, represents a stable 5- to 7-membered monocyclic- or stable 9- to 11-membered fused bi cyclic heterocyclic ring system which contains an aromatic ring. Any additional ring or rings fused to the aromatic ring may be saturated, such as piperidinyl, partially saturated, or unsaturated, such as pyridinyl, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O and S, and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
When a heterocyclyl group as defined herein is substituted, the substituent may be bonded to a ring carbon atom of the heterocyclic group, or on a ring heteroatom (i.e., a nitrogen, oxygen or sulfur), which has a valence which permits substitution. Preferably, the substituent is bonded to a ring carbon atom. Similarly, when a heterocyclic group is defined as a substituent herein, the point of attachment may be at a ring carbon atom of the heterocyclic group, or on a ring heteroatom (i.e., a nitrogen, oxygen or sulfur), which has a valence which permits attachment. Preferably, the attachment is at a ring carbon atom.
It is understood that the specific rings listed above are not a limitation on the rings which can be used in the present invention. These rings are merely representative.
Unless expressly stated to the contrary in a particular context, any of the various cyclic rings and ring systems described herein may be attached to the rest of the compound at any ring atom (i.e., any carbon atom or any heteroatom) provided that a stable compound results.
Unless expressly stated to the contrary, all ranges cited herein are inclusive. For example, a heteroaromatic ring described as containing from " 1 to 3 heteroatoms" means the ring can contain 1, 2 or 3 heteroatoms. It is also understood that any range cited herein includes within its scope all of the sub-ranges within that range. Thus, for example, a heterocyclic ring described as containing from " 1 to 4 heteroatoms" is intended to include as aspects thereof, heterocyclic rings containing 2 to 4 heteroatoms, 3 or 4 heteroatoms, 1 to 3 heteroatoms, 2 or 3 heteroatoms, 1 or 2 heteroatoms, 1 heteroatom, 2 heteroatoms, 3 heteroatoms, or 4 heteroatoms. As another example, a moeity described as optionally substituted with "from 1 to 3 substituents" is intended to include as aspects thereof, such moeity substituted with 1 to 3 substituents, 2 or 3 substituents, 3 substituents, 1 or 2 substituents, 2 substituents, or 1 substituent.
When any variable (e.g., R3 or R3a) occurs more than one time in any constituent or in Formula I or in any other formula depicting and describing compounds of the present invention, its definition on each occurrence is independent of its definition at every other occurrence. Also, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. Unless expressly depicted or described otherwise, variables depicted in a structural formula with a "floating" bond attached to a ring, such as Ra, are permitted to be a substituent on any available carbon or nitrogen atom in the ring to which the variable is attached. When a moiety is noted as being "optionally substituted" in formula I or any embodiment thereof, it means that formula I or the embodiment thereof encompasses compounds that are substituted with the noted substituent (or substituents) on the moiety and compounds that do not contain the noted substituent (or substituents) on the moiety (i.e., wherein the moiety is unsubstituted).
Unless expressly stated to the contrary, substitution by a named substituent is permitted on any atom in a chain or ring provided such substitution is chemically allowed and results in a stable compound. A "stable" compound is a compound which can be prepared and isolated and whose structure and properties remain or can be caused to remain essentially unchanged for a period of time sufficient to allow use of the compound for the purposes described herein (e.g., therapeutic or prophylactic administration to a subject). The compounds of the present invention are limited to stable compounds embraced by Formula I.
To the extent substituents and substituent patterns provide for the existence of tautomers (e.g., keto-enol tautomers) in the compounds of the invention, all tautomeric forms of these compounds, whether present individually or in mixtures, are within the scope of the present invention. Compounds of the present invention having a hydroxy substituent on a carbon atom of a heteroaromatic ring are understood to include compounds in which only the hydroxy is present, compounds in which only the tautomeric keto form (i.e., an oxo substitutent) is present, and compounds in which the keto and enol forms are both present.
The compounds of Formula I may have one or more chiral (asymmetric) centers. The present invention encompasses all stereoisomeric forms of the compounds of Formula I. Centers of asymmetry that are present in the compounds of Formula I can all independently of one another have (R) or (S) configuration. When bonds to a chiral carbon are depicted as straight lines in the structural Formulas of the invention, or when a compound name is recited without an (R) or (S) chiral designation for a chiral carbon, it is understood that both the (R) and (S) configurations of each such chiral carbon, and hence each enantiomer or diastereomer and mixtures thereof, are embraced within the Formula or by the name. The production of specific stereoisomers or mixtures thereof may be identified in the Examples where such stereoisomers or mixtures were obtained, but this in no way limits the inclusion of all stereoisomers and mixtures thereof from being within the scope of this invention.
The invention includes all possible enantiomers and diastereomers and mixtures of two or more stereoisomers, for example mixtures of enantiomers and/or diastereomers, in all ratios. Thus, enantiomers are a subject of the invention in enantiomerically pure form, both as levorotatory and as dextrorotatory antipodes, in the form of racemates and in the form of mixtures of the two enantiomers in all ratios. In the case of a cis/trans isomerism the invention includes both the cis form and the trans form as well as mixtures of these forms in all ratios. The preparation of individual stereoisomers can be carried out, if desired, by separation of a mixture by customary methods, for example by chromatography or crystallization, by the use of stereochemically uniform starting materials for the synthesis or by stereoselective synthesis. Optionally a derivatization can be carried out before a separation of stereoisomers. The separation of a mixture of stereoisomers can be carried out at an intermediate step during the synthesis of a compound of Formula I or it can be done on a final racemic product. Absolute stereochemistry may be determined by X-ray crystallography of crystalline products or crystalline intermediates which are derivatized, if necessary, with a reagent containing a stereogenic center of known configuration. Alternatively, absolute stereochemistry may be determined by Vibrational Circular Dichroism (VCD) spectroscopy analysis. The present invention includes all such isomers, as well as salts, solvates (which includes hydrates) and solvated salts of such racemates, enantiomers, diastereomers and tautomers and mixtures thereof.
In the compounds of Formula I, the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature. The present invention is meant to include all suitable isotopic variations of the compounds of Formula I. For example, different isotopic forms of hydrogen (H) include protium (lH) and deuterium (¾T). Protium is the predominant hydrogen isotope found in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements, or may provide a compound useful as a standard for characterization of biological samples. Isotopically- enriched compounds within Formula I can be prepared without undue experimentation by conventional techniques well known to those skilled in the art or by processes analogous to those described in the Schemes and Examples herein using appropriate isotopically-enriched reagents and/or intermediates.
The compounds can be administered in the form of pharmaceutically acceptable salts. The term "pharmaceutically acceptable salt" refers to a salt which possesses the effectiveness of the parent compound and which is not biologically or otherwise undesirable (e.g., is neither toxic nor otherwise deleterious to the recipient thereof).
When the compounds of Formula I contain one or more acidic or basic groups the invention also includes the corresponding pharmaceutically acceptable salts. Thus, the compounds of Formula I which contain acidic groups can be used according to the invention as, for example but not limited to, alkali metal salts, alkaline earth metal salts or as ammonium salts. Examples of such salts include but are not limited to sodium salts, potassium salts, calcium salts, magnesium salts or salts with ammonia or organic amines such as, for example, ethylamine, ethanolamine, triethanolamine or amino acids. Compounds of Formula I which contain one or more basic groups, i.e. groups which can be protonated, can be used according to the invention in the form of their acid addition salts with inorganic or organic acids as, for example but not limited to, salts with hydrogen chloride, hydrogen bromide, phosphoric acid, sulfuric acid, nitric acid, benzenesulfonic acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acids, oxalic acid, acetic acid, trifluoroacetic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, formic acid, propionic acid, pivalic acid, diethylacetic acid, malonic acid, succinic acid, pimelic acid, fumaric acid, maleic acid, malic acid, sulfaminic acid, phenylpropionic acid, gluconic acid, ascorbic acid, isonicotinic acid, citric acid, adipic acid, etc. If the compounds of Formula I simultaneously contain acidic and basic groups in the molecule the invention also includes, in addition to the salt forms mentioned, inner salts or betaines (zwitterions). Salts can be obtained from the compounds of Formula I by customary methods which are known to the person skilled in the art, for example by combination with an organic or inorganic acid or base in a solvent or dispersant, or by anion exchange or cation exchange from other salts. The present invention also includes all salts of the compounds of Formula I which, owing to low
physiological compatibility, are not directly suitable for use in pharmaceuticals but which can be used, for example, as intermediates for chemical reactions or for the preparation of
pharmaceutically acceptable salts.
Another embodiment of the present invention is a compound of Formula I, or a pharmaceutically acceptable salt thereof, as originally defined or as defined in any of the foregoing embodiments, aspects, classes, or subclasses, wherein the compound or its salt is in a substantially pure form. As used herein "substantially pure" means suitably at least about 60 wt.%, typically at least about 70 wt.%, preferably at least about 80 wt.%, more preferably at least about 90 wt.% (e.g., from about 90 wt.% to about 99 wt.%), even more preferably at least about 95 wt.% (e.g., from about 95 wt.% to about 99 wt.%, or from about 98 wt.% to 100 wt.%), and most preferably at least about 99 wt.% (e.g., 100 wt.%) of a product containing a compound of Formula I or its salt (e.g., the product isolated from a reaction mixture affording the compound or salt) consists of the compound or salt. The compounds of the invention have two or more asymmetric centers and can occur as mixtures of stereoisomers. It is understood that a substantially pure compound can be either a substantially pure mixture of stereoisomers or a substantially pure individual diastereomer or enantiomer. The level of purity of the compounds and salts can be determined using a standard method of analysis such as thin layer
chromatography, gel electrophoresis, high performance liquid chromatography, and/or mass spectrometry. If more than one method of analysis is employed and the methods provide experimentally significant differences in the level of purity determined, then the method providing the highest level of purity governs. A compound or salt of 100% purity is one which is free of detectable impurities as determined by a standard method of analysis.
Furthermore, compounds of the present invention may exist in amorphous form and/or one or more crystalline forms, and as such all amorphous and crystalline forms and mixtures thereof of the compounds of Formula I are intended to be included within the scope of the present invention. In addition, some of the compounds of the instant invention may form solvates with water (i.e., a hydrate) or common organic solvents. Such solvates and hydrates, particularly the pharmaceutically acceptable solvates and hydrates, of the instant compounds are likewise encompassed within the scope of this invention, along with un-solvated and anhydrous forms.
Any pharmaceutically acceptable pro-drug modification of a compound of this invention which results in conversion in vivo to a compound within the scope of this invention is also within the scope of this invention. For example, esters can optionally be made by esterification of an available carboxylic acid group or by formation of an ester on an available hydroxy group in a compound. Similarly, labile amides can be made. Pharmaceutically acceptable esters or amides of the compounds of this invention may be prepared to act as prodrugs which can be hydrolyzed back to an acid (or -COO" depending on the pH of the fluid or tissue where conversion takes place) or hydroxy form particularly in vivo and as such are encompassed within the scope of this invention. Examples of pharmaceutically acceptable pro- drug modifications include, but are not limited to, -Ci-6alkyl esters and -Ci-6alkyl substituted with phenyl esters.
Accordingly, the compounds within the generic structural formulas, embodiments and specific compounds described and claimed herein encompass salts, all possible
stereoisomers and tautomers, physical forms (e.g., amorphous and crystalline forms), solvate and hydrate forms thereof and any combination of these forms, as well as the salts thereof, pro-drug forms thereof, and salts of pro-drug forms thereof, where such forms are possible unless specified otherwise.
The invention also encompasses methods for the treatment or prophylaxis of infection by HIV or for the treatment, prophylaxis, or delay in the onset of AIDS in a subject in need thereof, which comprises administering to the subject an effective amount of a compound of the invention or a pharmaceutically acceptable salt thereof.
The invention also encompasses a compound of the invention, or a pharmaceutically acceptable salt thereof, for use in the preparation of a medicament for the inhibition of HIV protease, for the treatment or prophylaxis of infection by HIV, or for the treatment, prophylaxis, or delay in the onset of AIDS in a subject in need thereof.
The invention also encompasses a pharmaceutical composition comprising an effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier and further comprising an effective amount of an anti-HIV agent selected from the group consisting of HIV antiviral agents, immunomodulators, and anti-infective agents. Within this embodiment, the anti-HIV agent is an antiviral selected from the group consisting of HIV protease inhibitors, HIV reverse transcriptase inhibitors, HIV integrase inhibitors, HIV fusion inhibitors, HIV entry inhibitors, and HIV maturation inhibitors. Compounds of formula II and III each form a subset of the compounds included in formula I. Any description which follows that refers to a compound of Formula I also applies to a compound of formula II and III and all embodiments thereof.
Other embodiments of the present invention include the following: (a) A pharmaceutical composition comprising an effective amount of a compound of formula I as defined above, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
(b) A pharmaceutical composition which comprises the product prepared by combining (e.g., mixing) an effective amount of a compound of formula I as defined above, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
(c) The pharmaceutical composition of (a) or (b), further comprising an effective amount of an anti-HIV agent selected from the group consisting of HIV antiviral agents, immunomodulators, and anti-infective agents.
(d) The pharmaceutical composition of (c), wherein the anti-HIV agent is an antiviral selected from the group consisting of HIV protease inhibitors, HIV reverse transcriptase inhibitors, HIV integrase inhibitors, HIV fusion inhibitors, HIV entry inhibitors, and HIV maturation inhibitors.
(e) The pharmaceutical composition of (d), wherein the antiviral is selected from the group consisting of HIV reverse transcriptase inhibitors and HIV integrase inhibitors.
(f) A combination which is (i) a compound of Formula I as defined above, or a pharmaceutically acceptable salt thereof, and (ii) an anti-HIV agent selected from the group consisting of HIV antiviral agents, immunomodulators, and anti-infective agents; wherein Compound I and the anti-HIV agent are each employed in an amount that renders the combination effective for inhibition of HIV protease, for treatment or prophylaxis of infection by HIV, or for treatment, prophylaxis of, or delay in the onset or progression of AIDS.
(g) The combination of (f), wherein the anti-HIV agent is an antiviral selected from the group consisting of HIV protease inhibitors, HIV reverse transcriptase inhibitors, HIV integrase inhibitors, HIV fusion inhibitors, HIV entry inhibitors, and HIV maturation inhibitors.
(h) The combination of (g), wherein the antiviral is selected from the group consisting of HIV reverse transcriptase inhibitors and HIV integrase inhibitors.
(i) A method for the inhibition of HIV protease in a subject in need thereof which comprises administering to the subject an effective amount of a compound of Formula I or a pharmaceutically acceptable salt thereof.
(j) A method for the prophylaxis or treatment of infection by HIV (e.g., HIV-1) in a subject in need thereof which comprises administering to the subject an effective amount of a compound of Formula I or a pharmaceutically acceptable salt thereof.
(k) The method of (j), wherein the compound of Formula I is administered in combination with an effective amount of at least one other HIV antiviral selected from the group consisting of HIV protease inhibitors, HIV reverse transcriptase inhibitors, HIV integrase inhibitors, HIV fusion inhibitors, HIV entry inhibitors, and HIV maturation inhibitors.
(1) The method of (k), wherein the at least one other HIV antiviral is selected from the group consisting of HIV reverse transcriptase inhibitors and HIV integrase inhibitors.
(m) A method for the prophylaxis, treatment or delay in the onset or progression of AIDS in a subject in need thereof which comprises administering to the subject an effective amount of a compound of Formula I or a pharmaceutically acceptable salt thereof.
(n) The method of (m), wherein the compound is administered in combination with an effective amount of at least one other HIV antiviral, selected from the group consisting of HIV protease inhibitors, HIV reverse transcriptase inhibitors, HIV integrase inhibitors, HIV fusion inhibitors, HIV entry inhibitors, and HIV maturation inhibitors.
(o) The method of (n), wherein the at least one other HIV antiviral is selected from the group consisting of HIV reverse transcriptase inhibitors and HIV integrase inhibitors.
(p) A method for the inhibition of HIV protease in a subject in need thereof which comprises administering to the subject the pharmaceutical composition of (a), (b), (c) or (d) or the combination of (e) or (f).
(q) A method for the prophylaxis or treatment of infection by HIV (e.g., HIV-1) in a subject in need thereof which comprises administering to the subject the pharmaceutical composition of (a), (b), (c), (d) or (e).
(r) A method for the prophylaxis, treatment, or delay in the onset or progression of AIDS in a subject in need thereof which comprises administering to the subject the pharmaceutical composition of (a), (b), (c), (d) or (e).
The present invention also includes a compound of formula I, or a pharmaceutically acceptable salt thereof, (i) for use in, (ii) for use as a medicament for, or (iii) for use in the manufacture/preparation of a medicament for: (a) therapy (e.g., of the human body), (b) medicine, (c) inhibition of HIV protease, (d) treatment or prophylaxis of infection by HIV, or (e) treatment, prophylaxis of, or delay in the onset or progression of AIDS. In these uses, the compounds of the present invention can optionally be employed in combination with one or more other anti-HIV agents selected from HIV antiviral agents, anti-infective agents, and immunomodulators.
Additional embodiments of the invention include the pharmaceutical compositions, combinations and methods set forth in (a)-(r) above and the uses (i)(a)-(e) through (iii)(a)-(e) set forth in the preceding paragraph, wherein the compound of the present invention employed therein is a compound of one of the embodiments, aspects, classes or subclasses described above. In all of these embodiments, the compound can optionally be used in the form of a pharmaceutically acceptable salt.
Additional embodiments of the present invention include each of the pharmaceutical compositions, combinations, methods and uses set forth in the preceding paragraphs, wherein the compound of the present invention or its salt employed therein is substantially pure. With respect to a pharmaceutical composition comprising a compound of formula I or a pharmaceutically acceptable carrier and optionally one or more excipients, it is understood that the term "substantially pure" is in reference to a compound of formula I or its salt per se.
The methods of the present invention involve the use of compounds of the present invention in the inhibition of HIV protease (e.g., wild type HIV-1 and/or mutant strains thereof), the prophylaxis or treatment of infection by human immunodeficiency virus (HIV) and the prophylaxis, treatment or delay in the onset or progression of consequent pathological conditions such as AIDS. Prophylaxis of AIDS, treating AIDS, delaying the onset or progression of AIDS, or treating or prophylaxis of infection by HIV is defined as including, but not limited to, treatment of a wide range of states of HIV infection: AIDS, ARC (AIDS related complex), both symptomatic and asymptomatic, and actual or potential exposure to HIV. For example, the present invention can be employed to treat infection by HIV after suspected past exposure to HIV by such means as blood transfusion, exchange of body fluids, bites, accidental needle stick, or exposure to patient blood during surgery.
In general, compounds that are HIV protease inhibitors can be identified as those compounds which, when tested in the "Cell-based HIV Infection Assay using a Reporter" assay described below, have an inflection point (IP) of ΙΟμΜ, parti culalry 5 μΜ or less, preferably 1 μΜ or less, and more preferably 0.25 μΜ or less.
The term "administration" and variants thereof (e.g., "administering" a compound) in reference to a compound of formula I mean providing the compound to the individual in need of treatment or prophylaxis and includes both self-administration and administration to the patient by another person. When a compound is provided in combination with one or more other active agents (e.g., antiviral agents useful for treating or prophylaxis of HIV infection or AIDS), "administration" and its variants are each understood to include provision of the compound and other agents at the same time or at different times. When the agents of a combination are administered at the same time, they can be administered together in a single composition or they can be administered separately.
As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients, as well as any product which results from combining the specified ingredients.
By "pharmaceutically acceptable" is meant that the ingredients of the pharmaceutical composition must be compatible with each other and not deleterious to the recipient thereof.
The term "subject" as used herein refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment. The term "effective amount" as used herein means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician. In one embodiment, the effective amount is a "therapeutically effective amount" which is an amount effective for inhibiting HIV protease (wild type and/or mutant strains thereof), inhibiting HIV replication (either of the foregoing which may also be referred to herein as an "inhibition effective amount"), treating HIV infection, treating AIDS, delaying the onset of AIDS and/or slowing progression of AIDS. In another embodiment, the effective amount is a "prophylactically effective amount" which is an amount effective for prophylaxis of HIV infection or prophylaxis of AIDS. It is understood that an effective amount can simultaneously be both a therapeutically effective amount, e.g., for treatment HIV infection, and a
prophylactically effective amount, e.g., for prevention or reduction of risk of developing AIDS. When the active compound (i.e., active ingredient) is administered as the salt, references to the amount of active ingredient are to the free form (i.e., the non-salt form) of the compound.
In the methods of the present invention (e.g., inhibiting HIV protease, treating or prophylaxis of HIV infection, inhibiting HIV replication, treating or prophylaxis of AIDS, delaying the onset of AIDS, or delaying or slowing progression of AIDS), the compounds of formula I, optionally in the form of a salt, can be administered by any means that produces contact of the active agent with the agent's site of action. They can be administered by any conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic agents or in a combination of therapeutic agents. They can be administered alone, but typically are administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice. The compounds of the invention can, for example, be administered by one or more of the follwing routes: orally, parenterally
(including subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques), by inhalation spray, or rectally, in the form of a unit dosage of a pharmaceutical composition containing an effective amount of the compound and conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles. Liquid preparations suitable for oral administration (e.g., suspensions, syrups, elixirs and the like) can be prepared according to techniques known in the art and can employ any of the usual media such as water, glycols, oils, alcohols and the like. Solid preparations suitable for oral administration (e.g., powders, pills, capsules and tablets) can be prepared according to techniques known in the art and can employ such solid excipients as starches, sugars, kaolin, lubricants, binders, disintegrating agents and the like. Parenteral compositions can be prepared according to techniques known in the art and typically employ sterile water as a carrier and optionally other ingredients, such as a solubility aid. Injectable solutions can be prepared according to methods known in the art wherein the carrier comprises a saline solution, a glucose solution or a solution containing a mixture of saline and glucose. Further description of methods suitable for use in preparing pharmaceutical compositions for use in the present invention and of ingredients suitable for use in said compositions is provided in Remington's Pharmaceutical Sciences. 18th edition, edited by A. R. Gennaro, Mack Publishing Co., 1990 and in Remington - The Science and Practice of Pharmacy. 21st edition, Lippincott Williams & Wilkins, 2005.
The compounds of formula I can be administered orally in a dosage range of 0.001 to 1000 mg/kg of mammal (e.g., human) body weight per day in a single dose or in divided doses. One dosage range is 0.01 to 500 mg/kg body weight per day orally in a single dose or in divided doses. Another dosage range is 0.1 to 100 mg/kg body weight per day orally in single or divided doses. For oral administration, the compositions can be provided in the form of tablets or capsules containing 1.0 to 500 milligrams of the active ingredient, particularly 1, 5, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. The specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy. In some cases, depending on the potency of the compound or the individual response, it may be necessary to deviate upwards or downwards from the given daily dose. Furthermore, the compound may be formulated for immediate or modified release such as extended or controlled release.
As noted above, the present invention is also directed to use of a compound of formula I with one or more additional anti-HIV agents. An "anti-HIV agent" is any agent which is directly or indirectly effective in the inhibition of HIV reverse transcriptase, protease, or another enzyme required for HIV replication or infection, the inhibition of HIV replication, the treatment or prophylaxis of HIV infection, and/or the treatment, prophylaxis or delay in the onset or progression of AIDS. It is understood that an anti-HIV agent is effective in treating, preventing, or delaying the onset or progression of HIV infection or AIDS and/or diseases or conditions arising therefrom or associated therewith. For example, the compounds of this invention may be effectively administered, whether at periods of pre-exposure and/or postexposure, in combination with effective amounts of one or more anti-HIV agents selected from HIV antiviral agents, imunomodulators, antiinfectives, or vaccines useful for treating HIV infection or AIDS, such as those disclosed in Table 1 of WO 01/38332 or in the Table in WO 02/30930. Suitable HIV antivirals for use in combination with the compounds of the present invention include, for example, those listed in Table A as follows:
Table A: Antiviral A ents for Treatin HIV infection or AIDS
Figure imgf000025_0001
Figure imgf000026_0001
Figure imgf000027_0001
EI = entry inhibitor; FI = fusion inhibitor; Inl = integrase inhibitor; PI = protease inhibitor; nRTI = nucleoside reverse transcriptase inhibitor; nnRTI = non-nucleoside reverse transcriptase inhibitor. Some of the drugs listed in the table are used in a salt form; e.g., abacavir sulfate, delavirdine mesylate, indinavir sulfate, atazanavir sulfate, nelfinavir mesylate, saquinavir mesylate.
It is understood that the scope of combinations of the compounds of this invention with anti-HIV agents is not limited to the HIV antivirals listed in Table A and/or listed in the above-referenced Tables in WO 01/38332 and WO 02/30930, but includes in principle any combination with any pharmaceutical composition useful for the treatment or prophylaxis of AIDS. The HIV antiviral agents and other agents will typically be employed in these combinations in their conventional dosage ranges and regimens as reported in the art, including, for example, the dosages described in the Physicians' Desk Reference. Thomson PDR, Thomson PDR, 57th edition (2003), the 58th edition (2004), or the 59th edition (2005) and the current Physicians' Desk Reference (68th ed.). (2014), Montvale, NJ: PDR Network. The dosage ranges for a compound of the invention in these combinations are the same as those set forth above.
The compounds of this invention are also useful in the preparation and execution of screening assays for antiviral compounds. For example, the compounds of this invention are useful for isolating enzyme mutants, which are excellent screening tools for more powerful antiviral compounds. Furthermore, the compounds of this invention are useful in establishing or determining the binding site of other antivirals to HIV protease, e.g., by competitive inhibition. Thus the compounds of this invention are commercial products to be used for these purposes.
Abbreviations and acronymns employed herein include the following: Bn = benzyl; BOC (or Boc) = t-butyloxycarbonyl; B0C2O = di-t-butyl carbonate; BOP = benzotriazol- l-yloxytris-(dimethylamino)phosphonium; BSA = bovine serum albumin; CBS = Corey, Bakshi, Shibata chiral oxazaborolidine mediated ketone reduction; Cbz = benzyloxycarbonyl; DBU = l,8-diazabicyclo[5.4.0]undec-7-one; DCAD = di-(4-chlorobenzyl) azodicarboxylate; DCE = 1,2- dichloroethane; DCM = dichloromethane; DEAD = diethyl azodicarboxylate; DIAD = diisopropylazodicarboxylate; Dibal-H = diisobutylaluminum hydride; DMAP = 4- dimethylaminopyridine; DMF = dimethylformamide; DMSO = dimethyl sulfoxide; e.g. = for example (but not limited to); EDC = l-ethyl-3-(3-dimethylaminopropyl) carbodiimide; Et = ethyl; EtOAc = ethyl acetate; EtOH = ethanol; G-2G = Grubbs catalyst, 2nd generation; HOAt = l-hydroxy-7-azabenzotriazole; HPLC = high performance liquid chromatography; HSU = hydroxysuccinimide; i-PrOH = isopropanol; LAH = lithium aluminum hydride; LCMS = liquid chromatography-mass spectroscopy; Me = methyl; MeOH = methanol; MOC = methoxycarbonyl; Ms = mesyl or methanesulfonyl; NMR = nuclear magnetic resonance; Ph = phenyl; RCM = ring closing metathesis; Piv = pivaloyl; PPTS = pyridinium p-toluene sulfonate; PyBrOP = bromo-tris-pyrrolidinophosphonium hexafluorophosphate; ); rt, r.t. or RT = room temperature; SCX = strong cation exchange resin; STP = standard temperature and pressure (i.e., 25°C & 1 atmosphere); TBS = tert-butyldimethylsilyl; TBDPS = tert-butyl(diphenyl) silyl; TBDPSC1 = tert-butyl(dimethyl)silyl chloride; TEA = triethylamine; TFA = trifluoroacetic acid; THF = tetrahydrofuran; TLC = thin layer chromatography; TMAF = tetramethyl ammonium fluoride; TMSCHN2 = trimethylsilyl diazomethane; TPAP = tetrapropyl ammonium
perruthenate; TPP = triphenylphosphine.
The compounds of the present invention can be readily prepared according to the following reaction schemes and examples, or modifications thereof, using readily available starting materials, reagents and conventional synthesis procedures. In these reactions, it is also possible to make use of variants which are themselves known to those of ordinary skill in this art, but are not mentioned in greater detail. Furthermore, other methods for preparing compounds of the invention will be readily apparent to the person of ordinary skill in the art in light of the following reaction schemes and examples. Unless otherwise indicated, all variables are as defined above. In the examples that follow, when a nitrogen atom is depicted without the necessary hydrogen atoms to complete the valence, it is assumed those hydrogen atoms are present unless specifically stated to the contrary.
This invention relates to the preparation and use of compounds represented by
Formula I:
Figure imgf000028_0001
The compounds of formula I can be prepared using the general synthetic reaction schemes shown in Methods A through C.
Method A
Figure imgf000029_0001
Method A provides a route to compounds V and then to formula I compounds by first elaborating the monosubstituted amino acid ester II to the requisite disubstitiuted amino ester III. For example II is reacted with benzaldehyde under dehydrating conditions to provide an intermediate imine which can be treated with a strong base such as LiHMDS or LDA and reacted with an alkylating agent such as a alkyl halide or triflate (R4X - X is halide or trifiate) followed by subsequent hydrolysis of the imine to give di-substituted amino acid esters III.
Condensation of III with with thioureas IV provide compounds V in a similar manner as described in the literature (see McKittrick et al, Bioorganic & Medicinal Chemistry
Letters (2015), 25(7), 1592-1596 and references therein) using a coupling agent such as a carbodiimide under standard peptide coupling conditions. The resultant compounds V are saponified to their corresponding carboxylic acids VI. Acids VI are condensed under standard peptide coupling conditions with amines represented by structure R1 to provide amides VII. The resultant amides are then treated with an acid such as TFA or HCl to remove the BOC protecting group to provide compounds of formula I.
Method B
Figure imgf000030_0001
Alternatively, esters V can be prepared by condensing alpha diketones VIII with substituted guanidines IX according to literature procedures. The compounds V are then converted to compounds of formula I according to method A.
Method C
Figure imgf000030_0002
Method C provides another route to compounds V and then to formula I compounds by first condensing the disubstituted amino acid ester III with orthogonally protected thioureas X. For example, one embodiment of an orthogonally protected thiourea has a BOC protecting group on one nitrogen and a dimethoxybenzyl protecting group on the other nitrogen. The resultant compounds XI are then treated with palladium under an atmosphere of hydrogen to provide intermediates XII. Reaction of compounds XII with suitable alcohols XIII under Mitsunobu conditions provides compounds V and then compounds of formula I according to the appropriate steps from method A.
Detailed procedures for specific illustrative examples are shown below. Method A, Scheme 1;
Figure imgf000031_0001
SCHEME 1, STEP 1:
Methyl 2-amino-2-phenylacetate
Figure imgf000031_0002
To a cooled solution of phenylglycine (40.3 g, 266.3 mmol) in MeOH (250 ml), was added thionyl chloride (29.0 mL, 399.5 mmol) dropwise and stirred for 12 h until a colorless solution was obtained. The solvents were evaporated to give a pale yellow solid, more methanol was added to dissolve the solid and the solution was evaporated to dryness. The solid was placed on a high vacuum pump for 24 h. (53.0 g). XH NMR (300 MHz, CD3OD) δ 7.51 (d, J= 1.2 Hz, 5H), 5.22 (s, 1H), 3.8 (s, 3H).
SCHEME 1, STEP 2:
Methyl 2-(benzylideneamino)-2-phenylacetate
Figure imgf000031_0003
To a suspension of methyl 2-amino-2-phenylacetate (53.0 g, 263 mmol) in dichloromethane (250 mL) was added Et3N (44.7 mL, 315 mmol) dropwise and stirred for 1 h. Benzaldehyde (27 mL, 263 mmol) was added and the reaction was stirred for 12 h. Water (50 mL) was added to the reaction mixture and transferred to a separatory funnel. The organic layer was washed with brine and dried over Na2SC>4, filtered, and concentrated to give an oil which eventually solidified on the high vacuum pump to afford methyl 2-(benzylideneamino)-2-phenylacetate (65.9 g). XH NMR (300 MHz, CDC13) δ 10.02 (s, 1H), 7.84 - 7.79 (m, 2H) 7.53 (d, J=1.5 Hz) 7.5 - 7.28 (m, 8H), 5.20 (s, 1H), 3.74 (s, 3H).
SCHEME 1, STEP 3:
Methyl 2-amino-4-methyl-2-phenylpent-4-enoate
Figure imgf000032_0001
To an oven dried round bottom flask was added methyl 2-(benzylideneamino)-2-phenylacetate (5.0 g, 19.7 mmol), THF (75 mL), and 3-bromo-2-methylpropene (3.2 g, 23.9 mmol) and the mixture was cooled to -78 °C. LiHMDS (45 mL, 1M THF solution, 45 mmol) was added dropwise and stirred for 12 h as the reaction mixture warmed to room temperature. The mixture was cooled in an ice bath as 2N HC1 (2 x 30 mL) was added then transferred to a separatory funnel. The aqueous layer was separated and basified to pH 8-10 with 2N NaOH. The mixture was transferred back to the separatory funnel and extracted with EtOAc (3 x 30 mL). Then the organic portion was dried over Na2S04, filtered, and concentrated to give an oil. The oil was purified by silica gel chromatography (0-30% EtOAc/Hexanes) to give methyl 2-amino-4- methyl-2-phenylpent-4-enoate (1.45 g). XH NMR (300 MHz, CDC13) δ 7.60 - 7.51 (m, 2H), 7.37 - 7.27 (m, 3H), 4.93 (t, J= 1.6 Hz,lH), 4.80 (d, J= 0.7 Hz, 1H), 3.71 (s, 3H), 3.11 (d, J= 13.7 Hz, 1H), 2.69 (d, J= 13.3 Hz, 1H), 2.04 (bs, 2H), 1.28 (s, 3H). SCHEME 1, STEP 4:
Methyl 3-((2-((tert-butoxycarbonyl)imino)-4-(2-methylallyl)-5-oxo-4-phenylimidazolidin-l- yl)methyl)benzoate
Figure imgf000032_0002
To a solution of methyl 2-amino-4-methyl-2-phenylpent-4-enoate (1.4 g, 6.61 mmol) in DMF (10 mL) was added the thiourea (2.4 g, 7.27 mmol), EDCI (1.46 g, 7.60 mmol), and DIPEA (1.75 mL, 9.9 mmol). The mixture was heated to 60 °C for 24 h. The mixture was cooled to room temperature and then diluted with EtOAc (20 mL) then water (10 mL) and transferred to a separatory funnel. The EtOAc was washed with brine (3 x 10 mL) then dried over Na2S04, filtered, and concentrated to give an oil. The oil was purified by silica gel chromatography, eluting with 0-30% (3:2 mixture of CH2C12: EtOAc)/ hexanes to give methyl 3-((2-((tert- butoxycarbonyl)iniino)-4-(2-methylallyl)-5-oxo-4-phenyliniidazolidin-l-yl)methyl)benzoate (0.95 g). Mass Spec. (ESI+) m/z = 378 [C27H31N3O5 - C5H802 +H]+
SCHEME 1, STEP 5:
Methyl 3-((2-((tert-butoxycarbonyl)imino)-4-isobutyl-5-oxo-4-phenylimidazolidin-l- yl)methyl)benzoate
Figure imgf000033_0001
To a solution of methyl 3-((2-((tert-butoxycarbonyl)imino)-4-(2-methylallyl)-5-oxo-4- phenylimidazolidin-l-yl)methyl)benzoate (0.95 g, 1.99 mmol) in EtOH (20 mL) was added 10 wt. % Pd/C (0.25 g). The mixture was evacuated and purged with ¾ then stirred under an atmosphere of hydrogen. After 4 h the mixture was filtered and concentrated to dryness then subjected to the reaction conditions above by adding EtOH (20 mL) and 10 wt. % Pd/C (0.3 g) and stirred for an additional 48 h. The mixture was filtered and concentrated to give methyl 3- ((2-((tert-butoxycarbonyl)inrino)-4-isobu†yl-5-oxo-4-phe
(0.58 g). Mass Spec. (ESI+) m/z = 379 [C27H33N3O5 - Boc group +H]+
SCHEME 1, STEP 6:
3-((2-((teri-butoxycarbonyl)imino)-4-isobutyl-5-oxo-4-phenylimidazolidin-l
yl)methyl)benzoic acid
Figure imgf000033_0002
A suspension of methyl 3-((2-((/er/-butoxycarbonyl)imino)-4-isobu†yl-5-oxo-4- phenylimidazolidin-l-yl)methyl)benzoate (3.5 g, 7.4 mmol) and potassium trimethylsilanolate (2.1 g 16.2 mmol) in THF (75 mL) was heated to 75 °C for 3 h. The mixture stirred overnight at room temperature concentrated to dryness. EtOAc (50 mL) was added to the residue and 2N NaOH (30 mL). This mixture was transferred to a separatory funnel and the aqueous layer was separated, cooled in an ice bath, and the pH adjusted to to 5-6 with 2N HCl. The aqueous layer was added back to the EtOAc layer and, separated, and the aqueous layer was extracted with EtOAc (2 x 20 mL). The combined organic layers were dried over Na2SC>4, filtered and concentrated to give a solid. The solid was dissolved in enough CH2CI2 and purified by silica gel chromatography (0-20% EtOAc/Hexanes) to give compound 3-((2-((tert- butoxycarbonyl)imino)-4-isobutyl-5-oxo-4-phenylimidazolidin-l-yl)methyl)benzoic acid (2.9 g). Mass Spec. (ESI+) m/z = 366 [C26H31N3O5 - C5H802 +H]+
SCHEME 1, STEP 7:
Methyl 4-(3-((2-imino-4-isobutyl-5-oxo-4-phenylimidazolidin-l- yl)methyl)benzoyl)piperazine-l-carboxylate hydrochloride
Figure imgf000034_0001
HC1 (1.5 mL, 4N in dioxane, 2.03 mmol) was added to a solution of 3-((2-((tert- butoxycarbonyl)imino)-4-isobutyl-5-oxo-4-phenylimidazolidin-l-yl)methyl)benzoic acid (60 mg, 0.10 mmol) in CH2CI2 and stirred overnight at room temperature. The reaction mixture was concentrated and the residue was purified by silica gel chromatography (0 - 5 % MeOH/CH2Cl2) to give methyl 4-(3-((2-imino-4-isobutyl-5-oxo-4-phenylimidazolidin-l- yl)methyl)benzoyl)piperazine-l-carboxylate as the HCI salt. The residue was dissolved in acetonitrile (6 mL) and added 0.4N HCI (2 mL) solution, freeze dried, and lyophilized to furnish compound methyl 4-(3-((2-imino-4-isobutyl-5-oxo-4-phenylimidazolidin-l- yl)methyl)benzoyl)piperazine-l-carboxylate hydrochloride (32 mg). Mass Spec. (ESI+) m/z = 492 [C27H33N5O4 +H]+, 500 MHz ¾ NMR Spectrum (DMSO-c¾) δ 11.0 (s, 1H), 9.9 (b, 2H), 7.5 - 7.34 (m, 8H), 7.23 (s, 1H), 4.96 (d, J = 3.6, 2H), 3.63 (s, 3H), 3.30 - 3.16 (m, 8H), 2.15 (d, J= 6.3 Hz, 1H), 2.08 (d, J= 5.6 Hz, 1H), 1.54 (t, J = 6.4 Hz, 1H), 0.78 (d, J = 6.5 Hz, 3H), 0.72 (d, J = 6.6 Hz, 3H).
A variation using this modified sequence of Method A was also utilized as illustrated in Scheme 2 which in some cases gave improved results.
Method A, Scheme 2
Figure imgf000035_0001
SCHEME 2, STEP 1:
Methyl 2-amino-4-methyl-2-phenylpentanoate
Figure imgf000035_0002
To a solution methyl 2-amino-4-methyl-2-phenylpent-4-enoate (2.8 g, 12.77 mmol) in EtOH (100 mL) was added 10 wt. % Pd/C (0.7 g). The mixture was evacuated, then purged under nitrogen, then switched to hydrogen. After 17 h, the reaction mixture was purged with nitrogen, filtered, and concentrated to give compound methyl 2-amino-4-methyl-2-phenylpentanoate (2.8 g). ¾ NMR (300 MHz, CDC13) δ 7.55 - 7.50 (m, 2H), 7.36 - 7.22 (m, 3H), 3.69 (s, 3H), 2.15 - 2.00 (m, 1H), 1.98-1.94 (m, 1H), 1.84 - 1.71 (m, 1H), 0.90 (d, J= 6.7 Hz, 3H) 0.84 (d, J= 6.6 Hz, 3H). SCHEME 2, STEP 2:
Methyl 3-((2-((tert-butoxycarbonyl)imino)-4-isobutyl-5-oxo-4-phenylimidazolidin-l- yl)methyl)benzoate
Figure imgf000035_0003
To a solution of methyl 2-amino-4-methyl-2-phenylpentanoate (4.3 g, 19.4 mmol) in DMF (75 mL) was added methyl 3-((3-(tert-butoxycarbonyl)thioureido)methyl)benzoate (6.9 g, 21.4 mmol), EDCI (4.3 g, 22.4 mmol) and DIPEA (5.0 mL, 29.2 mmol). The mixture was heated to 65 °C for 24 h. Diluted the mixture with EtOAc (50 mL), added H20 (30 mL) and transferred to a separatory funnel. The EtOAc layer was washed with brine (2 x 30 mL), then dried over Na2SC>4, filtered and concentrated to give an oil. The oil was purified by silica gel
chromatography, eluting with 0-20% (3:2 mixture of CLLC^EtOAc) in Hexanes to give methyl 3-((2-((tert-butoxycarbonyl)inrino)-4-isobutyl-5-oxo-4^
(6.8 g). Mass Spec. (ESI+) m/z = 380 [C27H33N3O5 - C5H802 + H]+ Method B, Scheme 3
Figure imgf000036_0002
SCHEME 3, STEP 1:
Methyl 3-((4-(3-bromophenyl)-4-(4-fluorophenyl)-2-imino-5-oxoimidazolidin- 1- yl)methyl)benzoate
Figure imgf000036_0001
A mixture of l-(3-bromophenyl)-2-(4-fluorophenyl)ethane-l,2-dione (300 mg, 0.97 mmol), methyl 3-(guanidinomethyl)benzoate (304 mg, 1.46 mmol), and ΕΪ Ν (0.54 mL, 3.90 mmol) in anhydrous MeOH (8 mL) was heated to refluxing temperature for 20 h under nitrogen. After this period the reaction mixture was cooled to room temperature and the precipitate was filtered off. The filtrate was concentrated under reduced pressure and the crude mixture was purified by silica gel chromatography eluting with 0 to 5% MeOH in DCM to furnish methyl 3-((4-(3- bromophenyl)-4-(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l-yl)methyl)benzoate (240 m Mass Spec. (APCI+) m/z =496 (M+H).
SCHEME 3, STEP 2:
Methyl 3-((4-(3-bromophenyl)-2-(teri-butoxycarbonylimino)-4-(4-fluorophenyl)-5- oxoimidazolidin-l-yl)methyl)benzoate
Figure imgf000037_0001
To a solution of methyl 3-((4-(3-bromophenyl)-4-(4-fluorophenyl)-2-imino-5-oxoimidazolidin- l-yl)methyl)benzoate (233 mg, 0.46 mmol) in DCM (4 mL) at 0 °C, was added (Boc)20 (0.12 mL, 0.51 mmol) and ΕΪ Ν (0.13 mL, 0.93 mmol) under nitrogen. The reaction mixture was allowed to warm up to room temperature and stirred for 16 h. After this period solvent was evaporated and residue was re-dissolved in DCM (20 mL). The organic layer was washed with water (5 mL) and brine (5 mL), dried over Na2SC>4, filtered and concentrated to dryness. The crude mixture was purified by silica gel chromatography eluting with 5 to 10% EtOAc in Hexanes to afford methyl 3-((4-(3-bromophenyl)-2-(tert-butoxycarbonylimino)-4-(4- fluorophenyl)-5-oxoimidazolidin-l-yl)methyl)benzoate (260 mg).1!! NMR (400 MHz, CDC13) δ 9.20 (s, 1H), 8.30-8.05 (m, 1H), 7.99-7.94 (m, 1H), 7.57-7.53 (m, 1H), 7.52-7.47 (m, 1H), 7.43-7.36 (m, 2H), 7.30-7.19 (m, 4H), 4.91 (s, 2H), 1.54 (s, 9H).
SCHEME 3, STEP 3:
3-((4-(3-bromophenyl)-2-(teri-butoxycarbonylimino)-4-(4-fluorophenyl)-5-oxoimidazolidin- l-yl)methyl)benzoic acid
Figure imgf000037_0002
To a solution of methyl 3-((4-(3-bromophenyl)-2-(ter^butoxycarbonylimino)-4-(4- fluorophenyl)-5-oxoimidazolidin-l-yl)methyl)benzoate (255 mg, 0.42 mmol) in THF (3 mL), was added KOTMS (110 mg, 0.85 mmol) and the reaction mixture was heated at 45 °C for 1.5 h under nitrogen. After this period solvent was evaporated, water added and the mixture was basified to pH 10 with aq NaOH (6N, 2 mL). This basic aqueous solution was then acidified to pH 4 by drop-wise addition of aq HCl (6N) at 0 °C. The acidic mixture was further acidified to pH 2 by drop-wise addition of aq HCl (6N) resulting in the formation of a thick precipitate which was filtered off quickly. The filtered cake was washed with water (2 x 3 mL) and then dried on the lyophilizer to furnish 3-((4-(3-bromophenyl)-2-(ter^butoxycarbonylimino)-4-(4- fluorophenyl)-5-oxoimidazolidin-l-yl)methyl)benzoic acid (220 mg). XH NMR (300 MHz, DMSO-c e) δ 10.35 (s, 1H), 7.88-7.81 (m, 1H), 7.78-7.73 (m, 1H), 7.64-7.57 (m, 1H), 7.54-7.43 (m, 3H), 7.43-7.32 (m, 4H), 7.32-7.20 (m, 2H), 4.83 (s, 2H), 1.44 (s, 9H).
SCHEME 3, STEP 4:
tert-Butyl 4-(3-bromophenyl)-4-(4-fluorophenyl)-l-(3-(isoindoline-2-carbonyl)benzyl)-5- oxoimidazolidin-2-ylidenecarbamate
Figure imgf000038_0001
To a mixture of 3-((4-(3-bromophenyl)-2-(tert-butoxycarbonylimino)-4-(4-fluorophenyl)-5- oxoimidazolidin-l-yl)methyl)benzoic acid (215 mg, 0.36 mmo), isoindoline (88 mg, 0.73 mmol), and HATU (210 mg, 0.55 mmol) in anhydrous DMF (3 mL) was added DIPEA (0.2 mL, 1.10 mmol, 3 eq) and the reaction mixture was heated at 45 °C for 16 h under nitrogen. After this period DMF evaporated, added water, then extracted with DCM (3 x 5 mL). Combined organic layer was washed with brine (5 mL), dried over Na2SC>4, filtered, and concentrated to dryness. The crude mixture was purified by silica gel chromatography eluting with 0 to 1% MeOH in DCM to furnish fert-butyl 4-(3-bromophenyl)-4-(4-fluorophenyl)-l-(3-(isoindoline-2- carbonyl)benzyl)-5-oxoimidazolidin-2-ylidenecarbamate (220 mg). MS (ESI+) m/z 685
[(M+H)+2].
SCHEME 3, STEP 5: 3'-(4-(4-fluorophenyl)-2-imino-l-(3-(isoindoline-2-carbonyl)benzyl)-5-oxoimidazolidin-4- yl)biphenyl-3-carbonitrile
Figure imgf000039_0001
A mixture of fert-butyl 4-(3-bromophenyl)-4-(4-fluorophenyl)-l-(3-(isoindoline-2- carbonyl)benzyl)-5-oxoimidazolidin-2-ylidenecarbamate (215 mg, 0.31 mmol), (3- cyanophenyl)boronic acid (92.4 mg, 0.62 mmol) and K2C03 (174 mg in 0.63 mL H20, 2M, 1.25 mmol) in 1,4-dioxane (5 mL) was bubbled with nitrogen for 10 min. To this mixture was added Pd(PPh3)4 (36.3 mg, 0.03 mmol) and the reaction mixture was refluxed for 12 h. After this period the reaction mixture was cooled to room temperature, the solid was filtered off. The filter cake was washed with DCM (5 mL) and the combined filtrate was evaporated to dryness. The crude mixture was purified by silica gel chromatography eluting with 0 to 5% MeOH in DCM to furnish 3'-(4-(4-fluorophenyl)-2-imino-l-(3-(isoindoline-2-carbonyl)benzyl)-5-oxoimidazolidin- 4-yl)biphenyl-3-carbonitrile (83 mg). Mass Spec. (ESI+) m/z =606 (M+H).
SCHEME 3, STEP 6:
TFA salt of 3'-(4-(4-fluorophenyl)-2-imino-l-(3-(isoindoline-2-carbonyl)benzyl)-5- oxoimidazolidin-4-yl)biphenyl-3-carbonitrile
Figure imgf000039_0002
To a solution of 3'-(4-(4-fluorophenyl)-2-imino-l-(3-(isoindoline-2-carbonyl)benzyl)-5- oxoimidazolidin-4-yl)biphenyl-3-carbonitrile (83 mg, 0.13 mmol) in anhydrous DCM (1 mL), was added TFA (0.05 mL, 0.68 mmol) at 0 °C and then the reaction mixture was warmed up to room temperature and stirred for 15 min. After this period, DCM was evaporated to dryness. The crude mixture was dissolved in a 1 :2 mixture of water:CH3CN (0.5 mL: l mL) and was lyophilized to furnish the TFA salt of 3'-(4-(4-fluorophenyl)-2-imino-l-(3-(isoindoline-2- carbonyl)benzyl)-5-oxoimidazolidin-4-yl)biphenyl-3-carbonitrile (90 mg). 1HNMR (300 MHz, DMSO-d6) δ 12.20-11.60 (bs, 1H), 10.20-9.60 (bs, 2H), 8.08-8.00 (m, 1H), 7.93-7.82 (m, 2H), 7.80-7.64 (m, 3H), 7.62-7.10 (m, 15H), 5.06 (s, 2H), 4.80 (s, 2H), 4.52 (s, 2H). Mass Spec. (APCI+) m/z =606 (M+H).
Method C, Scheme 4
Figure imgf000040_0001
SCHEME 4, STEP 1:
Isopropyl 5-(l-hydroxypentyl)-2-(trifluoromethyl)benzoate
Figure imgf000040_0002
A solution of isopropyl 5-formyl-2-(trifluoromethyl)benzoate (197 mg, 0.759 mmol) in THF (10 niL) was cooled to 0 °C and treated with w-BuMgCl (0.5 mL, 2.0 M in Et20). The reaction mixture was stirred at 0 °C for 1 h. Then the reaction mixture was quenched by addition of sat NH4Cl(aq) (2 mL). The reaction mixture was diluted with EtOAc (80 mL), H20 (20 mL), and sat NH4Cl(aq) (20 mL). The layers were separated, and the organic layer was washed with brine (1 x 20 mL), dried over Na2SC>4, filtered, and concentrated. The crude product was purified by column chromatography using EtOAc and hexanes as eluents to yield isopropyl 5-(l- hydroxypentyl)-2-(trifluoromethyl)benzoate (95 mg) as a solid. Mass Spec. (ESI+) m/z =319.2 (M + H+).
SCHEME 4, STEP 2:
Isopropyl 5-(l-((R)-2-((tert-butoxycarbonyl)imino)-4-(4-fluorophenyl)-4-isopentyl-5- oxoimidazolidin-l-yl)pentyl)-2-(trifluoromethyl)benzoate.
Figure imgf000041_0001
To a solution of isopropyl 5-(l-hydroxypentyl)-2-(trifluoromethyl)benzoate (95 mg, 0.3 mmol) and fert-butyl f7^-(4-(4-fluorophenyl)-4-isopentyl-5-oxoimidazolidin-2-ylidene)carbamate (91 mg, 0.25 mmol) in THF (1 mL) were added «-Β¾Ρ (0.1 mL, 0.40 mmol) and diethyl azodicarboxylate (0.078 mL, 0.50 mmol) at room temperature. The reaction mixture was stirred at room temperature under nitrogen for 12 h. The reaction mixture was concentrated down and purified by column chromatography using EtOAc and hexanes as eluents to yield isopropyl 5-(l- ((Kj-2-((tert-butoxycarbonyl)iimno)-4-(4-fl
yl)pentyl)-2-(trifluoromethyl)benzoate (67 mg) as a oil. Mass Spec. (ESI+) m/z =664.4 (M + H+).
SCHEME 4, STEP 3:
5-(l-((R)-2-((tert-butoxycarbonyl)imino)-4-(4-fl
l-yl)pentyl)-2-(trifluoromethyl)benzoic acid
Figure imgf000041_0002
To a solution of isopropyl 5-(l-((R)-2-((tert-butoxycarbonyl)imino)-4-(4-fluorophenyl)-4- isopentyl-5-oxoimidazolidin-l-yl)pentyl)-2-(trifluoromethyl)benzoate (67 mg, 0.10 mmol) in THF (0.5 mL) and MeOH (0.5 mL) was added a solution of LiOH (0.3 mL, 4N in H20). The reaction mixture was stirred at 50 °C. After 22 h, the reaction mixture was cooled to room temperature, diluted with H20 (10 mL) and acidified to ~pH 2 by addition of IN HCl(aq). The reaction mixture was extracted with CH2CI2 (2 x 30 mL) and EtOAc (2 x 30 mL). All the organic extracts were combined, dried over Na2SC>4, filtered, and concentrated to yield 5-(\-((R)- 2-((ter^butoxycarbonyl)imino)-4-(4-fluorophenyl)-4-isopentyl-5-oxoimidazolidin-l-yl)penfy 2-(trifluoromethyl)benzoic acid (63 mg) as a solid. Mass Spec. (ESI+) m/z = 622.2 (M + H+).
SCHEME 4, STEP 4:
tert-butyl ((4R)-l-(l-(3-(6,7-dihydro-5H-pyrrolo[3,4-b]pyrazine-6-carbonyl)-4- (trifluoromethyl)phenyl)pentyl)-4-(4-fluorophenyl)-4-isopentyl-5-oxoimidazolidin-2- ylidene)carbamate.
Figure imgf000042_0001
To a solution of 5-(l-((R)-2-((tert-butoxycarbonyl)irnino)-4-(4-fluorophenyl)-4-isopentyl-5- oxoimidazolidin-l-yl)pentyl)-2-(trifluoromethyl)benzoic acid (63 mg, 0.10 mmol) and hydrochloric salt of 6,7-dihydro-5H-pyrrolo[3,4-b]pyrazine (28 mg, 0.12 mmol) in DMF (2 mL) were added catalytic amount of DMAP, diisopropylethylamine (0.089 mL, 0.51 mmol), and HATU (46 mg, 0.12 mmol) The reaction mixture was stirred at room temperature for 20 h. Then, the reaction mixture was diluted with EtOAc (50 mL), washed with saturated NaHC03(aq) (2 x 50 mL), water (4 x 50 mL), brine (1 x 50 mL), dried over Na2SC>4, filtered, and
concentrated. The crude product was purified by column chromatography using EtOAc and hexanes as eluents to yield amide tert-butyl ((4R)-l-(l-(3-(6,7-dihydro-5H-pyrrolo[3,4- b]pyrazine-6-carbonyl)-4-(trifluoromethyl)phenyl)pentyl)-4-(4-fluorophenyl)-4-isopentyl-5- oxoimidazolidin-2-ylidene)carbamate as a solid. Mass Spec. (ESI+) m/z =725.4 (M + H+)
SCHEME 4, STEP 5 :
(5R)-3-(l-(3-(6,7-dihydro-5H-pyrrolo[3,4-b]pyrazine-6-carbonyl)-4-
(trifluoromethyl)phenyl)pentyl)-5-(4-fluorophenyl)-2-imino-5-isopentylimidazolidin-4-one
Figure imgf000043_0001
To a solution of fert-butyl ((4R)-l-(l-(3-(6,7-dihydro-5H-pyrrolo[3,4-b]pyrazine-6-carbonyl)-4- (trifluoromethyl)phenyl)pentyl)-4-(4-fluorophenyl)-4-isopentyl-5-oxoimidazolidin-2- ylidene)carbamate (40 mg, 0.056 mmol) in CH2CI2 (2 mL) was added trifluoroacetic acid (0.6 mL, TFA). The reaction mixture was stirred at room temperature under nitrogen for 2 h. Then, the reaction mixture was transferred into a separatory funnel containing sat NaHCC (30 mL). The pH of the resulting mixture was adjusted to 9 by addition of 1 N NaOH(aq) and extracted with CH2CI2 (3 x 50 mL). The organic extracts were combined, dried over Na2SC>4, filtered, and concentrated. The crude products were purified by chiral semi-preparative HPLC using iPrOH and hexanes as eluents to give (5R)-3-(l-(3-(6,7-dihydro-5H-pyrrolo[3,4-b]pyrazine-6- carbonyl)-4-(trifluoromethyl)phenyl)pentyl)-5-(4-fluorophenyl)-2-imino-5- isopentylimidazolidin-4-one (8 mg) as a solid. Mass Spec. (ESI+) m/z = 625.4 (M + H+).
Preparation of Intermediates
Preparation of Compounds IV.
Figure imgf000043_0002
Compounds IV were generally prepared in a similar method described in the literature
(reference: Synthesis 2010, 6, 991). A solution of NN-bisboc-thiourea was treated with sodium hydride followed by treatment with TFAA. After which a solution of requisite amine was added to the reaction mixture to afford compound IV.
An example of the preparation of compound IV is shown below in Scheme 5
SCHEME 5:
Methyl 3-((3-(tert-butoxycarbonyl)thioureido)methyl)-5-fluorobenzoate
Figure imgf000044_0001
To an ice cold solution of NN-bisboc-thiourea (335.8 g, 1.21 mol) in THF (4.5 L) was added NaH (35 g) in a 12 L 3-neck RBF. After 1 hr at this temperature TFAA was added over 30 min, keeping the temperature close to 3°C. After stirring for an additional 1 hr a solution of methyl 3- (aminomethyl)-5-fluorobenzoate (133.7 g, 0.72 mol) in THF (0.3 L) was added over 30 min, keeping the temperature at 3°C. After stirring for 90 minutes the reaction mixture was poured into 18 L cold water. Then brine (2L) was added to this solution and the resulting solution was extracted with 8L EtOAc and, subsequently, 4L EtOAc. The combined organic layer was dried over MgS04, filtered, and concentrated to dryness. The residue was further azeotroped with hexane 4L, and solidified upon cooling. The solid was filtered, and washed with cold pentane. The solid residue was triturated with 40/60 isopropanol: pentane, chilled and filtered. The solid was further washed with 40/60 isopropanol: pentane (cold) 3 x 200 ml, filtered and dried under vacuum to give the desired product as a solid (230 g). Preparation of Compounds IX.
Figure imgf000044_0002
Compounds IX were generally prepared in the following manner. A solution of S- methylisothiourea sulfate and requisite amine were heated in water to afford compounds IX.
An example of the preparation of compound IX is shown below in Scheme 5
SCHEME 6:
l-(2,4-dimethoxybenzyl)guanidine
Figure imgf000044_0003
The mixture of S-methylisothiourea sulfate (6.95g, 50 mmol) and 2,4-dimethoxybenzylamine (8.36g, 50 mmol) in 10 mL water was heated at 105 °C for 2 h. After this period the reaction mixture was cooled to room temperature, filtered, and rinsed with water. The filter cake was suspended in 40 mL water and was heated to boiling. The suspension was cooled to room temperature, filtered, rinsed with water and a 9: 1 mixture of Et^O and zPrOH. The crystalline product was dried in vacuo to provide 10.2 g of l-(2,4-dimethoxybenzyl)guanidine.
Preparation of Compound X.
An example of the preparation of compound X is shown below in Scheme 7.
SCHEME 7:
Compound X
Figure imgf000045_0001
SCHEME 7, STEP 1
l-(Isothiocyanatomethyl)-2,4-dimethoxybenzene
To a solution of 2,4-dimethoxybenzylamine (6.6 mL, 43.5 mmol) in DCM (85 mL) was added saturated aqueous sodium bicarbonate solution (85 mL) and the mixture was stirred vigorously at RT for 15 min. Stirring was stopped then thiophosgene (6.6 mL, 87 mmol) was added via syringe to the bottom layer. The mixture was stirred at RT for 90 min then the aqueous layer was separated and the organic layer was washed with brine, dried over Na2SC>4 and concentrated in vacuo twice from DCM to give 9.1 g of l-(isothiocyanatomethyl)-2,4-dimethoxybenzene as an oil.
SCHEME 7, STEP 2
Compound X
To a suspension of 60% sodium hydride in hexanes (3.4 g, 85 mmol) in anhydrous THF (100 mL) at 0 °C was added fert-butylcarbamate (7.4 g, 63 mmol) and the mixture was stirred for 15 min. A solution of l-(isothiocyanatomethyl)-2,4-dimethoxybenzene (9.1 g, 43.5 mmol) in anhydrous THF (50 mL) was then added over 15 min and the reaction was allowed to warm up to RT and stirred overnight. The final mixture was quenched with water and 10% aqueous phosphoric acid until neutral pH, extracted with EtOAc, dried over Na2SC>4 and concentrated in vacuo. The residue was purified by chromatography over silica gel (eluting with hexanes/EtOAc 100:0 to 80:20) to afford 10.61 g of compound X as a solid. Preparation of Compounds XIII.
Figure imgf000046_0001
Precursors include, but are not limited to, requisite aldehydes, carboxylic esters, or carboxylic acids which may be treated with reducing reagents to afford the corresponding alcohols. Alternatively, requisite aldehydes may be treated with organometalic reagents to afford the corresponding secondary alcohols. Examples of the preparation of compounds XIII are shown below in Schemes 8 and 9.
SCHEME 8:
Methyl 5-(hydroxymethyl)thiophene-3-carboxylate
Figure imgf000046_0002
To a solution of methyl 5-formylthiophene-3-carboxylate (1.01 g, 5.93 mmol) in MeOH (60 mL) held at 0 °C was added sodium borohydride (0.45 g, 11.9 mmol). After the bubbling ceased, the reaction was allowed to warm to room temperature and stirred an additional 30 min. The mixture was extracted with EtOAc (200 mL) and brine (200 mL). Theorganic portion was collected, dried over sodium sulfate, filtered, and concentrated in vacuo to afford methyl 5- (hydroxymethyl)thiophene-3-carboxylate. (0.89 g) XH NMR (400 MHz, CDC13) δ 8.04 (d, 1H), 7.40 (m, 1H), 4.82 (s, 2H), 3.85 (s, 3H).
SCHEME 9:
Methyl 4-(hydroxymethyl)-6-methylpicolinate
SCHEME 9, STEP 1:
Methyl 2-cyano-6-methylisonicotinate
Figure imgf000046_0004
To a degassed solution of methyl 2-chloro-6-methylisonicotinate (2.03g, 10.9 mmol) in DMF (20 mL) was added tetrakis(triphenylphosphine)palladium(0) (1.18 g, 1.02 mmol) and zinc cyanide (1.14g, 9.71 mmol). This mixture was heated to 80 °C while stirring for 18 h under an atmosphere of nitrogen. The mixture was cooled to room temperature, then extracted with EtOAc (100 mL) and sat'd NH4OH (aq) solution (2 x 100 mL) then brine (2 x 100 mL). The combined organic extracts were dried over sodium sulfate, filtered, and concentrated in vacuo. The resultant residue was purified by silica gel chromatography eluting with 0 to 30% EtOAc in Hexanes to afford methyl 2-cyano-6-methylisonicotinate (1.29 g). Mass Spec. (ESI+) m/z =177.2 (M + H+).
SCHEME 9, STEP 2:
4-(hydroxymethyl)-6-methylpicolinonitrile
Figure imgf000047_0001
A solution of methyl 2-cyano-6-methylisonicotinate (602 mg, 3.42 mmol) in THF (5 mL) was cooled to 0 °C while stirring. To this cooled solution was added sodium borohydride (388 mg, 10.25 mmol). Then, to this solution was added MeOH (12 mL) and continue to stir at 0 °C for 90 min. The solution was allowed to warm to room temperature and stirred an additional 15 min. The reaction mixture was quenched with 1 N HC1 (aq) solution to adjust the pH to ~7. The mixture was extracted with EtOAc (50 mL) and brine (50 mL) and the combined organic portions collected, dried over sodium sulfate, filtered, and concentrated in vacuo to afford 4- (hydroxymethyl)-6-methylpicolinonitrile (0.50 g). lH NMR (400 MHz, CDC13) δ 7.52 (s, 1H), 7.38 (s, 1H), 4.75 (s, 2H), 2.56 (s, 3H).
SCHEME 9, STEP 3:
Methyl 4-(hydroxymethyl)-6-methylpicolinate
Figure imgf000047_0002
A solution of 4-(hydroxymethyl)-6-methylpicolinonitrile (0.50 g, 3.4 mmol) in water (3 mL) and cone, sulfuric acid (2 mL) was heated to 135 °C while stirring for 18 h. After this time the reaction was cooled to 95 °C and MeOH (8 mL) was added to the mixture and was allowed to stir at 95 °C for an additional 1 h. The reaction was allowed to cool to room temperature. Then the contents of the reaction mixture added directly over ice. Water (100 mL) and EtOAc (100 mL) was added to this mixture. Solid sodium bicarbonate was added to the solution to adjust to pH ~8. The aqueous layer was extracted with EtOAc (2 x 100 mL). The combined organic portions were washed with brine (100 mL), dried over sodium sulfate, and concentrated to afford methyl 4-(hydroxymethyl)-6-methylpicolinate (170.9 mg). XH NMR (400 MHz, CDC13) δ 7.90 (s, 1H), 7.36 (s, 1H), 4.76 (s, 2H), 3.97 (s, 3H), 2.63 (s, 3H). Mass Spec. (ESI+) m/z = 182.2 (M + H+).
The compounds shown in Table 1 were made by following procedures analogous to the schemes and Examples herein.
TABLE 1
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
- Ill -
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Assay for Inhibition of Microbial Expressed HIV Protease
Inhibition of Escherichia coli expressed wild-type HIV-1 protease protein was carried out with a peptide substrate [Val-Ser-Gln-Asn-( naphtyl)Ala-Pro-Ile-Val]. The inhibitor compound was preincubated with HIV-1 protease enzyme in assay buffer (50 mM sodium acetate, pH 5.5, 100 mM NaCl, and 0.1% BSA) for 30 minutes at room temperature. Peptide substrate was added to 400 μΜ in a total volume of 20 containing 20 pM HIV-1 protease (final) after which the reaction was incubated for 1 hour at 30°C. The reaction was quenched by the addition of formic acid and HIV protease inhibitor indinavir to 0.012% and 150 nM final concentrations, respectively. Product formation was determined after separation of product and substrate on a ZORBAX Eclipse XDB-C18 column (Aligent Technologies, Santa Clara, CA, USA) connected to an API 4000™ mass spectrometer (AB Sciex, Pte. Ltd., Concord Ontario, Canada) with multiple reaction monitoring (transitions were 644.5/428.9 and 615.4/422.2 (M1/M3) for product and indinavir respectively). The extent of inhibition of the reaction was determined from the peak area of the products. Analysis of the products, independently synthesized, provided quantitation standards and confirmation of the product composition. Representative compounds of the present invention exhibit inhibition of HIV-1 protease in this assay. Ic50's refer to the 50% inhibition of the cleavage of a peptide substrate by hiv protease.
Table 2 shows data obtained from the above described assays for the Compounds herein. Data shown in the table reflects the mean of at least two independent experiments.
TABLE 2
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0002
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
While the foregoing specification teaches the principles of the present invention, with examples provided for the purpose of illustration, the practice of the invention encompasses all of the usual variations, adaptations and/or modifications that come within the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. A compound of structural Formula I:
Figure imgf000137_0001
or a pharmaceutically acceptable salt thereof, wherein:
A is selected from the group consisting of (CHR2)pC6-10aryl and (CHR2)pC4-nheteroaryl; R is selected from the group consisting of hydrogen and Ci-6alkyl;
Rx is selected from the group consisting of Ci-6alkyl, Ci-3haloalkyl, halogen, S02Ci-6alkyl, and OCi-6alkyl;
R1 is selected from the group consisting of a bicyclic, monocyclic, or tricyclic C4- 13heterocyclyl, said heterocyclyl optionally substituted with 1 to 3 groups of Ra;
R2 is selected from the group consisting of hydrogen, Ci-6alkyl, Ci-3haloalkyl, halogen, S02Ci-6alkyl, (CH2)nOCi-6alkyl, (CH2)nC3-6cycloalkyl, (CHR)nC6-10aryl, (CHR)nC5- lOheteroaryl, said aryl and heteroaryl optionally substituted with 1 to 3 groups of Ra;
R3 and R4 are independently selected from the group consisting of Ci-6alkyl, (CH2)nCl- 3haloalkyl, (CR2)nC3-6cycloalkyl, (CH2)nC6-10aryl, (CH2)nC5-ioheterocyclyl; said alkyl, aryl, and heterocyclyl optionally substituted with 1 to 3 groups of Rd;
R5 is selected from the group consisting of hydrogen, Ci-6alkyl, C(0)OR, C3-6cycloalkyl, SO2R, O(CH2)nC6-10aryl, and (CH2)nC6-10aryl;
Ra is selected from the group consisting of (CH2)nC5-l lheterocyclyl, Ci-6alkyl, C2-6alkenyl, C(0)Ci-6alkyl, OCi-6alkyl, =0, Cl -3haloalkyl, OCi-3haloalkyl, C3-6cycloalkyl, C(0)C3- 6cycloalkyl, halogen, CN, SCi -6alkyl, S02Ci-6alkyl, (CH2)nC6-12aryl, OC6-12aryl, C(0)C5- 12heterocyclyl, C(0)OR, C(0)OC2-6alkenyl, C(0)NR2, NR2, NHC(0)OR, -NHC3- 6cycloalkyl, -NR(CH2)C3-6cycloalkyl, NHC5-ioheterocyclyl, NHC6-10aryl,
(CH2)nNHC(0)R, said alkyl, alkenyl, aryl, and heterocyclyl optionally substituted with 1 to 3 groups of Rb; Rb is selected from the group consisting of Ci -6alkyl, Ci -6alkylOR, OR, =0, C(0)R, SOCi . 6alkyl, S02Ci-6alkyl, S02N(R)2, NRR5, Ci -3haloalkyl, OCi-3haloalkyl, C3-6cycloalkyl, halogen, (CH2)nCN, (CH2)nC6-12aryl, and (CH2)nC5-ioheteroaryl, said alkyl, aryl and heteroaryl optionally substituted with 1 to 3 groups of Rc;
Rc is selected from the group consisting of Ci-6alkyl, Ci-6alkylOR, OR, and halogen;
Rd is selected from the group consisting of Ci-6alkyl, Ci-3haloalkyl, CN, C(0)NR2, C5- loheteroaryl, C6-10aryl, and halogen, said heteroaryl, alkyl and aryl optionally substituted with 1 to 3 groups of halogen and CN; n is 0, 1, 2, 3, or 4; p is O or l ; and q is 0 or 1.
2. The compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein A is (CHR2)pC6-10aryl.
3. The compound according to any one of claims 1 and 2, or a pharmaceutically acceptable salt thereof , wherein the aryl of A is selected from the group consisting of phenyl, tetrahydronaphthalenyl, dihydroindenyl, and tetrahydrobenzoannulenyl.
4. The compound according to claim 1, or a pharmaceutically acceptable salt thereof wherein A is (CHR2)pC 5-nheteroaryl said heteroaryl selected from the group consisting pyridyl, thiazolyl, thiophenyl, dihydrochromenyl, and dihydrothiochromenyl.
5. The compound according to anyone of claims 1 to 4, or a pharmaceutically acceptable salt thereof, wherein Rl is (CH2)nC4-nheterocyclyl, said heterocyclyl unsubstituted or substituted with 1 to 3 groups of Ra.
6. The compound according to claim 5, or a pharmaceutically acceptable salt thereof, wherein Rl is unsubstituted or substituted (CH2)nC4-l lheterocyclyl wherein the heterocyclyl is selected from the group consisting of dihydropyrrolopyrazinyl,
dihydropyrrolopyrimidinyl, dihydrotriazolopyrazinyl, piperazinyl, piperazinonyl, piperidinyl, hexahydrooxazolopyrazinonyl, tetrahydropyrazinoindolyl, tetrahydrobenzapinyl, dihydropyrrolooxazolyl, tetrahydropyrrolopyrrolidione, pyrrolidinyl, hexahydroisoxazolyl, tetrahydropyrazolopyridyl, azetindinyl, tetrahydropyrrolotriazolooxazinyl,
tetrahydropyrroloisoxazolyl, tetrahydrofuropyridinyl, dihydroisoindolyl,
dihydropyrrolopyrazolyl, tetrahydropyrrolothiazolyl, tetrahydroimidazopyrazinyl,
dihydropyrrolopyridinyl, dihydropyrroloimidazolyl, dihydroisoindolopyridinyl,
dihydroisoquinolinyl, dihydronaphthyridinyl, and dihydroimidazopyrazinyl.
7. The compound according to claim 6, or a pharmaceutically acceptable salt thereof, wherein Rl is unsubstituted or substituted (CH2)nC4-l lheterocyclyl wherein the heterocyclyl is selected from the group consisting of dihydropyrrolopyrazinyl, piperazinyl, piperazinonyl, pyrrolidinyl, dihydropyrrolopyrimidinyl, dihydrotriazolopyrazinyl, and dihydroisoindolyl.
8. The compound according to any one of claims 1, 6, and 7, or a pharmaceutically acceptable salt thereof, wherein Rl is substituted with 1 to 3 groups of Ra selected from the group consisting of Ci-6alkyl, OCi-6alkyl, Ci-3haloalkyl, OCi-3haloalkyl, halogen, CN, SCi -6alkyl, S02Ci -6alkyl, (CH2)nC6-12aryl, (CH2)nC5-12heterocyclyl, C(0)OR, C(0)NR2, NR2, NHC(0)OR, C(0)C5-12heterocyclyl, -NHC3-6cycloalkyl, - NR(CH2)C3-6cycloalkyl, NHC5-ioheterocyclyl, and NHC6-10aryl, said alkyl, heterocyclyl, cycloalkyl and aryl unsubstituted or substituted with 1 to 3 groups of Rb.
9. The compound according to any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, wherein Rl is substituted with 0 to 1, 1 to 2, or 1 to 3 groups of Ra selected from the group consisting of CH3, CH2CH3, OCH3, CF3, OCF2, CH2CF3, fluoro, chloro, bromo, C(0)CH3, SCH3, SO2CH3, CN, COOCH3, COOCH2CH3, NHC(0)OCH3, NHC(0)CH3, CON(CH3)2, CONHCH3, CONHCH(CH3)2, NH2, NHCH3, N(CH3)2, and NHCH(CH3)2, (CH2)nC6-12phenyl, (CH2)nC5-12heterocyclyl, C(0)C5- 12heterocyclyl, -NHC3-6cycloalkyl, -NR(CH2)C3-6cycloalkyl, NHC5-ioheterocyclyl, and NHC6-10phenyl, said heterocyclyl, cycloalkyl and aryl unsubstituted or substituted with 1 to 3 groups of Rb.
10. The compound according to claim 9, or a pharmaceutically acceptable salt thereof, wherein the heterocyclyl of Ra is selected from the group consisting of unsubstituted or substituted pyrazolyl, pyridinyl, indazolyl, pyrrolyl, triazolyl, indolyl, pyrimidinyl, thiophenyl, tetrahydropyrazolopyridinyl, triazolopyridinyl, dihydropyrrolopyrazolyl, dihydropyridooxazinyl, isoquinolyl, isoxazolyl, dihydropyrrolyl, benzisoxazolyl, thiomo holinyl, oxadiazolyl, pyrrolodinyl, oxazolyl, oxophenylimidazolidinyl, dihydroimidazopyridinone, furanyl, dihydrobenzimidazolone, and benzoxazolone.
11. The compound according to any one of claims 1 to 10, or a
pharmaceutically acceptable salt thereof, wherein R2 is hydrogen.
12. The compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt thereof, wherein R2 is is selected from the group consisting of CH3, CH2CH3, (CH2)nCH(CH3)2, and (CH2)nOCH(CH3)2 , cyclopropyl, cyclobutyl, cyclopentyl, (CHR)nC6-10phenyl, and (CHR)nC5-ioheteroaryl.
13. The compound according to any one of claims 1 to 12, or a pharmaceutically acceptable salt thereof, wherein R3 and R4 are independently selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, pyridyl, pyranyl, (CH2)ntetrahydropyranyl, and (CH2)ntetrahydrofuranyl, said isobutyl, isopentyl, cyclopropyl, phenyl, pyridyl, pyranyl, tetrahydropyranyl, and tetrahydrofuranyl optionally substituted with 1 to 3 groups of Rd.
14. The compound according to claim 1 wherein the compounds of formula I are represented by structural formula II, or a pharmaceutically acceptable salts thereof,
Figure imgf000140_0001
wherein R2, R35 R4 Ra and RX are as defined in claim 1 and B, C, D, E, respectively, are selected from:
a) N=C-C=N;
b) C=N-C=N;
c) C=C-C=C;
d) N=C-C=C;
e) C=C-N=C; and
f) C=C-C=N.
15. The compound according to claim 14, or a pharmaceutically acceptable salt thereof, wherein one Ra is present and is selected from the group consisting of CH3, CH2CH3, OCH3, CF3, OCF2, CH2CF3, fluoro, chloro, bromo, C(0)CH3, SCH3, SO2CH3, CN, COOCH3, COOCH2CH3, NHC(0)OCH3, NHC(0)CH3, CON(CH3)2, CONHCH3, CONHCH(CH3)2, NH2, NHCH3, N(CH3>2, NHCH(CH3)2, unsubstituted or substituted (CH2)nC6-12phenyl, NHC6-10phenyl, and unsubstituted or substituted (CH2)nC5- 12heterocyclyl, NHC5-ioheterocyclyl, or C(0)C5-i2heterocyclyl, wherein the heterocyclyl is selected from the group consisting of optionally substituted pyrazolyl, pyridinyl, pyrimidinyl, dihydropyrrolopyrazolyl, pyrrolodinyl, dihydroimidazopyridinone, and dihydrobenzimidazolone, and R3 and R4 are independently selected from the group consisting of isobutyl, isopentyl, (CH2)nCF3, (CH2)ncyclopropyl, phenyl, pyridyl, pyranyl, (CH2)ntetrahydropyranyl, and (CH2)ntetrahydrofuranyl, said isobutyl, isopentyl, cyclopropyl, phenyl, pyridyl, pyranyl, tetrahydropyranyl, and tetrahydrofuranyl optionally substituted with 1 to 3 groups of Rd.
16. The compound according to claim 1 wherein the compound of formula I is represented by structural formula III, or a pharmaceutically acceptable salts thereof,:
Figure imgf000141_0001
wherein R2, R35 and R4 are as defined in claim 1, and wherein Y is selected from the group consisting of:
Figure imgf000141_0002
wherein
Ra is as originally described,
G is N or CH when— represents a double bond and is CH2 when— represents a single bond;
J is N or CH;
represents the point of attachment, and
— represents a single or double bond.
17. The compound of claim 1 which is:
(5R)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)phenyl]-3- methylbufyl}-5-(4-fluorophenyl)-2-imino-5-(tetrahydro-2H-pyran-4-yl)imidazolidin-4-one, (5R)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)phenyl]-4- phenylbufyl}-5-(4-fluorophenyl)-2-imino-5-(3-methylbu†yl)irnidazolidin-4-one, (5R)-5-(2-cyclopropylethyl)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-
(trifluoromethyl)phenyl]-3-phenylpropyl}-5-(4-fluorophenyl)-2-iminoimidazolidin-4-one,
(5R)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)phenyl]-2- phenylethyl}-5-(4-fluorophenyl)-2-imino-5-(3-methylbutyl)imidazolidin-4-one,
(5R)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)phenyl]-3- methylbutyl}-2-imino-5-(3-methylbutyl)-5-phenylimidazolidin-4-one,
(5R)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)phenyl]-2- phenylethyl}-5-(4-fluorophenyl)-2-imino-5-(3-methylbutyl)imidazolidin-4-one,
(5R)-5-(2-cyclopropylethyl)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-
(trifluoromethyl)phenyl]-2-(2-oxopyrrolidm^
one,
(5R)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)phenyl]-2-(l- methylethoxy)ethyl}-5-(4-fluorophenyl)-2-imino-5-(3-methylbutyl)imidazolidin-4-one,
(5R)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)phenyl]-4- methylpentyl}-2-imino-5-(3-methylbutyl)-5-phenylimidazolidin-4-one,
(5R)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)phenyl]-3- methylbutyl}-5-(4-fluorophenyl)-2-imino-5-[(3R) etrahydrofuran-3-ylmethyl]imidazolidin^
(5R)-3-{2-cyclopentyl-l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-
(trifluoromethyl)phenyl]ethyl}-5-(4-fluorophenyl)-2-imino-5-(3-methylbutyl)imidazolidin-4
(5R)-5-(4-fluorophenyl)-3-[3-({2-[l-(2-hydroxyethyl)-lH-pyrazol-4-yl]-5,7-dihydro-6H- pyrrolo[3,4-d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-2-imino-5-(4,4,4- trifluorobutyl)imidazolidin-4-one,
(5R)-3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-5-(4- fluorophenyl)-2-imino-5-(4,4,4-trifluorobutyl)imidazolidin-4-one,
(5R)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)phenyl]-3- methylbutyl}-5-(4-fluorophenyl)-2-imino-5-(tetrahydro-2H-pyran-4-ylmethyl)imidazolidin-^
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[2-(l-methyl-lH-pyrazol-4-yl)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,44rifluorobutyl)imidazolidin-4-one,
(5R)-3-[3-({2-[6-(dimethylamino)pyridin-3-yl]-5 ,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6- yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4-fluorophenyl)-2-imino-5-(4,4,4- trifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-{3-[(2-{l-[2-(methylsulfonyl)ethyl]-lH-pyrazol-4-yl}-5,7- dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl)carbonyl]-4-(trifluoromethyl)benzyl}-5-(4,4,4- trifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[2-(l-propyl-lH-pyrazol-4-yl)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,44rifluorobutyl)imidazolidin-4-one, (5R)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)phenyl]-3- methylbutyl}-5-(4-fluorophenyl)-2-imino-5 (3 (5R)-5-(4-fluorophenyl)-2-imino-3-[3-({2-[l-(2-methoxyethyl)-lH-pyrazol-4-yl]-5,7-dihydro-6H- pyrrolo[3,4-d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4- trifluorobutyl)iirddazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-({2-[3-(methylsulfinyl)phenyl]-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4 rifluorobutyl)imidazolidin-4-one,
(5R)-5 -(4-fluoropheny l)-2-imino-3- [3- { [2-( 1 H-indazol-4-y l)-5 ,7-dihy dro-6H-pyrrolo [3,4- d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,4-trifluorobutyl)imidazolidin-4-one,
(5R)-2-imino-5 3-methylbutyl)-3-{4-methyl-1 4-(trifluoromethyl)-3-{[3-(trifluoromethyl)-5,6- dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl}phenyl]pentyl}-5-phenylimidazolidin-4- one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-({2-[3-(methylsulfonyl)phenyl]-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4 rifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-({2-[5-(4-methylpiperazin-l-yl)pyridin-3-yl]-5,7-dihydro-6H- pyrrolo[3,4-d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4- trifluorobutyl)imidazolidin-4-one,
5-(2-cyclopropylethyl)-3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4- (trifluoromethyl)benzyl]-5-(3-fluorophenyl)-2-iminoimidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[2-(3-methyl-lH-pyrazol-4-yl)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,4-trifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[2-(4,5,6,7-tetrahydropyrazolo[l,5-a]pyridin-3-yl)-5,7- dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,4- trifluorobutyl)imidazolidin-4-one,
(5S)-5-(4-fluorophenyl)-2-imino-3-{3-methyl-l-[4-(trifluoromethyl)-3-{[3-(trifluoromethyl)-5,6- dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl}phenyl]butyl}-5-[(3R)-tetrahydrofuran-3- ylmethyl]imidazolidin-4-one,
(5R)-3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-2-imino- 5-(3-methylbutyl)-5-phenylimidazolidin-4-one,
(5R)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)phenyl]-3- methylbutyl}-2-imino-5-(3-methylbutyl)-5-phenylimidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-{3-[(2-[l,2,4]triazolo[l,5-a]pyridin-6-yl-5,7-dihydro-6H- pyrrolo[3,4-d]pyrimidin-6-yl)carbonyl]-4-(trifluoromethyl)benzyl}-5-(4,4,4- trifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3 3-({2-[l 2-morpholin-4-ylethyl)-lH-pyrazol-4-yl]-5,7-dihydro-
6H-pyrrolo[3,4-d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4- trifluorobutyl)imidazolidin-4-one,
(5R)-3-[3-{[2-(5,6-dihydro-4H-pyrrolo[l,2-b]pyrazol-3-yl)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4-fluorophenyl)-2-imino-5-(4,4,4- trifluorobutyl)iirddazolidin-4-one, (5R)-3-{3-[(2-chloro-5 ,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl)carbonyl]-4- (trifluoromethyl)benzy 1 } -5 -(4-fluorophen^
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-({2-[(3R) etrahydrofuran-3-ylamino]-5,7-dihydro-6H- pyrrolo[3,4-d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4- trifluorobutyl)imidazolidin-4-one,
(5R)-5-(2-cyclopropylethyl)-5-(4-fluorophenyl)-2-imino-3-{3-phenyl-l-[4-(trifluoromethyl)-3-{[3-
(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- yl]carbonyl}phenyl]propyl}imidazolidin-4-one,
(5R)-5-(2-cyclopropylethyl)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-
(trifluoromethyl)phenyl]-2-(l-methylethoxy)ethyl}-5-(4-fluorophenyl)-2-iminoimidazolidin-4-o^
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[2-(lH-pyrrolo[2,3-b]pyridin-5-yl)-5,7-dihydro-6H- pyrrolo[3,4-d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,4- trifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-({2-[3-(lH-pyrazol-5-yl)phenyl]-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4 rifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[2-(l-methyl-lH-indol-4-yl)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,44rifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-{3-methyl-l-[4-(trifluoromethyl)-3-{[3-(trifluoromethyl)-5,6- dihydro[l,2,4]triazolo[4,3-a] yrazine-7(8H)-yl]carbonyl}phenyl]butyl}-5-[(3R)-tetrahydrofuran-3- ylmethyl]imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[2-(l-methyl-lH-indazol-5-yl)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,44rifluorobutyl)imidazolidin-4-one,
N-[3-(6-{[5-{[(4R)-4-(4-fluorophenyl)-2-imino-5-oxo-4-(4,4,4-trifluorobu1yl)imidazolidin-l- yl]methyl}-2-(trifluoromethyl)phenyl]carbonyl}-6 -dihydro-5H-pyrrolo[3,4-d]pyrimidin-2- yl)phenyl]methanesulfonamide,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[2-(4-methyl-3,4-dihydro-2H-pyrido[3,2-b] [l,4]oxazin-7- yl)-5 -dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,4- trifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-({2-[l-(2-methylpropyl)-lH-pyrazol-4-yl]-5,7-dihydro-6H- pyrrolo[3,4-d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4- trifluorobutyl)iirddazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-{3-[(2-pyridin-3-yl-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6- yl)carbonyl]-4-(trifluoromethyl)benzyl}-5-(4,4,4 rifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-({2-[3-(5-methyl-l,3,4-oxadiazol-2-yl)phenyl]-5,7-dihydro-
6H-pyrrolo[3,4-d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4- trifluorobutyl)iirddazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-{3-[(2-pyrimidin-5-yl-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin- 6-yl)carbonyl]-4-(trifluoromethyl)benzyl}-5-(4,4,44rifluorobutyl)imidazolidin-4-one, (5R)-5-(4-fluorophenyl)-2-imino-3 3-{[3 methylsulfanyl)-5,6-dihydro[l,2,4]triazolo[4,3- a]pyrazin-7(8H)-yl]carbonyl}-4-(trifluoromethyl)ben^
(5R)-5-(4-fluorophenyl)-2-imino-3 3-({2 (l-methylethyl)amino]-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4 rifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[2-(2-methoxypyridin-4-yl)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,44rifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-{3-[(2-pyridin-4-yl-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6- yl)carbonyl]-4-(trifluoromethyl)benzyl}-5-(4,4,4 rifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-5-(3-methylbutyl)-3-{2-phenyl-l-[4-(trifluoromethyl)-3-{[3-
(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- y 1] carbonyl } phenyl] ethyl } imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-{3-[(2-isoquinolin-6-yl-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-
6-yl)carbonyl]-4-(trifluoromethyl)benzyl}-5-(4,4,44rifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-5-(3-methylbutyl)-3-{4-phenyl-l-[4-(trifluoromethyl)-3-{[3-
(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- yl]carbonyl}phenyl]butyl}imidazolidin-4-one,
(5R)-3-{3-[(2-amino-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl)carbonyl]-4- (trifluoromethyl)benzyl}-5-(4-fluorophenyl)-2^
6-{[5-{[(4R)-4-(4-fluorophenyl)-2-imino-5-oxo-4-(4,4,4-trifluorobu1yl)imidazolidin-l-yl]me (trifluoromethyl)phenyl]carbonyl}-6 -dihydro-5H-pyrrolo[3,4-d]pyrimidine-2-carbonitrile, (5R)-3-[3-{[2-(3-acetylphenyl)-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl]carbonyl}-4- (trifluoromethyl)benzyl]-5-(4-fluorophenyl)-2-i^
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[2-(3-methylisoxazol-4-yl)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,44rifluorobutyl)imidazolidin-4-one, [5-(6- { [5 - { [(4R)-4-(4-fluorophenyl)-2-imino-5 -oxo-4-(4,4,4-trifluorobuty l)imidazolidin- 1 - yl]methyl}-2-(trifluoromethyl)phenyl]carbonyl}-6 -dihydro-5H-pyrrolo[3,4-d]pyrimidin-2- y l)py ridin-2-y 1] acetonitrile,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[2-(3-pyrrolidin-l-ylphenyl)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,44rifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-{3-[(2-thiophen-3-yl-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6- yl)carbonyl]-4-(trifluoromethyl)benzyl}-5-(4,4,4 rifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[2-(3-methyl-l,2-benzisoxazol-5-yl)-5,7-dihydro-6H- pyrrolo[3,4-d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,4- trifluorobutyl)imidazolidin-4-one,
(5R)-3-{3-[(3-ethyl-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl)carbonyl]-4- (trifluoromethyl)benzyl}-5-(4-fluorophenyl)-2-i^
3 6-{[5-{[(4R)-4 4-fluorophenyl)-2-imino-5-oxo-4-(4,4,4-trifluorobutyl)imidazolidin-l- yl]methyl}-2-(trifluoromethyl)phenyl]carbonyl}-6 -dihydro-5H-pyrrolo[3,4-d]pyrimidin-2-yl)- Ν,Ν-dimethylbenzenesulfonamide,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-({2-[3-(thiomo holin-4-ylcarbonyl)phenyl]-5,7-dihydro-6H pyrrolo[3,4-d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4- trifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[2-(methylamino)-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-
6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,44rifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3 3-({2 2-(lH-pyrazol-l-yl)pyrimidin-5-yl]-5,7-dihydro-6H- pyrrolo[3,4-d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4- trifluorobutyl)imidazolidin-4-one,
(5R)-3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-2-imino- 5^henyl-5-[(3R)4etrahydrofuran-3-ylmethyl]imidazolidin-4-one,
(5R)-3-[3-{[2-(cyclopentylamino)-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl]carbonyl}-4-
(trifluoromethyl)benzyl]-5-(4-fluorophenyl)-2-imino-5-(4,4,4 rifluorobutyl)imidazolidin-4-on
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[2-(l-phenyl-lH-pyrazol-4-yl)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,44rifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-5-(4,4,4-trifluorobutyl)-3-[4-(trifluoromethyl)-3-{[2-
(trifluoromethyl)-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl]carbonyl}benzyl]imidazolidm^
(5R)-3-[3-({2-[(cyclopropylmethyl)amino]-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl}carbonyl)-
4- (trifluoromethyl)benzyl]-5-(4-fluoroph^
5- (2-cyclopropylethyl)-3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4- (trifluoromethyl)benzyl]-2-imino-5-[3-(trifluoromethyl)phenyl]imidazolidin-4-one,
5-(2-cyclopropylethyl)-3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4- (trifluoromethyl)benzyl]-2-imino-5-(4^yridin-3-ylphenyl)imidazolidin-4-one,
(6S)-6-(cyclopropylmethyl)-3-{ l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4- (trifluoromethyl)phenyl]-3-methylbutyl}-2-imino-6-phenyltetrahydropyrimidin-4(lH)-one, 5-(2-cyclopropylethyl)-5-(3-fluorophenyl)-2-imino-3-[4-(trifluoromethyl)-3-{[3-(trifluoromethyl)- 5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl}benzyl]imidazolidin-4-one,
3- (6-{[5-{[(4R)-4-(4-fluorophenyl)-2-imino-5-oxo-4-(4,4,4-trifluorobu1yl)imidazolidin-l- yl]methyl}-2-(trifluoromethyl)phenyl]carbonyl}-6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidin-2- yl)benzonitrile,
(5R)-3-[3-{[2-(dimethylamino)-5 -dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl]carbonyl}-4-
(trifluoromethyl)benzyl]-5-(4-fluorophenyl)-2-imino-5-(4,4,4 rifluorobutyl)imidazolidin-4-on methyl [6-({(8R)-8 (4R)-2-imino-4-(3-methylbutyl)-5-oxo-4^henylimidazolidin-l-yl]-5,6,7,8- tetrahydronaphthalen-2-yl}carbonyl)-6 -dihydro-5H^yrrolo[3,4-d]pyrimidin-2-yl]carbamate,
5-(2-cyclopropylethyl)-5-(3,5-difluorophenyl)-2-imino-3-[4-(trifluoromethyl)-3-{[3-
(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl}benzyl]imidazolidin-
4- one,
(5R)-5-(4-fluorophenyl)-2-imino-3-{3-[(2-mo holin-4-yl-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin- 6- yl)carbonyl]-4-(trifluoromethyl)benzyl}-5-(4,4,44rifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[4-(4-methyl-l,2,5-oxadiazol-3-yl)piperazin-l-yl]carbonyl}-
4- (trifluoromethyl)benzyl]-5-(4,4,4-trifluorobutyl)imidazolidin-4-one,
(5R)-3-[3-{[2-(cyclobutylamino)-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl]carbonyl}-4- (trifluoromethyl)benzyl]-5-(4-fluorophenyl)-2-imino-5-(4,4,4 rifluorobutyl)imidazolidin-4
(5R)-5-(4-fluorophenyl)-3-[3-{[2-(3-fluorophenyl)-5 -dihydro-6H-pyrrolo[3,4-d]pyrimidin-6- y 1] carbonyl } -4-(trifluoromethyl)benzy 1] -2-imino-5-(4,4,4-trifluorobuty l)imidazolidin-4-one,
7- {[5-{[(4R)-4-(4-fluorophenyl)-2-imino-5-oxo-4-(4,4,4-trifluorobu1yl)imidazolidin-l-yl]me (trifluoromethyl)phenyl]carbonyl}hexahydro[l,3]oxazolo[3,4-a]pyrazin-3-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-{3-[(4-pyrimidin-5-ylpiperazin-l-yl)carbonyl]-4- (trifluoromethyl)benzyl}-5-(4,4,4-trifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-({2-[6-(lH-pyrazol-l-yl)pyridin-3-yl]-5,7-dihydro-6H- pyrrolo[3,4-d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4- trifluorobutyl)iirddazolidin-4-one,
(5R)-5-(4-fluorophenyl)-3-[3-{[4-(2-hydroxy-2-methylpropanoyl)piperazin-l-yl]carbonyl}-4- (trifluoromethyl)benzyl]-2-imino-5-(4,4,4 rifluorobutyl)imidazolidin-4-one,
5- (2-cyclopropylethyl)-5-(3,5-difluorophenyl)-2-imino-3-[4-(trifluoromethyl)-3-{[3- (trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl}benzyl]imidazolidin- 4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[4-(methylsulfonyl)piperazin-l-yl]carbonyl}-4- (trifluoromethyl)benzyl]-5-(4,4,4-trifluorobutyl)imidazolidin-4-one,
(5R)-5-(2-cyclopropylethyl)-3-[(5R)-3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-
6,7,8,9 etrahydro-5H-benzo[7]annulen-5-yl]-5-(4-fluorophenyl)-2-iminoimidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-{3-[(2-phenyl-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6- yl)carbonyl]-4-(trifluoromethyl)benzyl}-5-(4,4,4 rifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-({2-[3-(2H-l,2,3-triazol-2-yl)phenyl]-5,7-dihydro-6H- pyrrolo[3,4-d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4- trifluorobutyl)imidazolidin-4-one,
methyl [6-({(3R)-3 (4R)-2-imino-4-(3-methylbutyl)-5-oxo-4-phenylimidazolidin-l-yl]-2,3-dihydro- lH-inden-5-yl}carbonyl)-6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidin-2-yl]carbamate,
(5R)-5-(4-fluorophenyl)-3-[3-{[2-(4-fluorophenyl)-5 -dihydro-6H-pyrrolo[3,4-d]pyrimidin-6- y 1] carbonyl } -4-(trifluoromethyl)benzy 1] -2-imino-5-(4,4,4-trifluorobuty l)imidazolidin-4-one,
(5R)-3-[3-(5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-ylcarbonyl)-4-(trifluoromethyl)benzyl]-5-
(4-fluorophenyl)-2-imino-5-(4,4,4-trifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-{3-[(4-pyridin-2-ylpiperazin-l-yl)carbonyl]-4-
(trifluoromethyl)benzyl}-5-(4,4,4-trifluorobutyl)imidazolidin-4-one,
(5R)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)phenyl]-3- methylbutyl}-5-(4-fluorophenyl)-2-imino-5-(tetrahydro-2H-pyran-4-yl)imidazolidin-4-one, (5R)-3-{cyclopentyl[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-
(trifluoromethyl)phenyl]methyl}-5-(4-fluorophenyl)-2-imino-5-(3-methylbutyl)imidazolidin-4-one, prop-2-en-l-yl 4-{[5-{[(4R)-4-(4-fluorophenyl)-2-imino-5-oxo-4-(4,4,4 rifluorobutyl)imidazolidin-
1 -yl] methyl } -2-(trifluoromethyl)phenyl] carbonyl } piperazine- 1 -carboxy late,
(5R)-5-(2-cyclopropylethyl)-5-(3-fluorophenyl)-2-imino-3-[4-(trifluoromethyl)-3-{[3-
(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl}benzyl]imidazolidin-
4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-{[2-(3-methylphenyl)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]carbonyl}-4-(trifluoromethyl)benzyl]-5-(4,4,44rifluorobutyl)imidazolidin-4-one,
2-imino-5-phenyl-5-[(3R)-tetrahydrofuran-3-ylmethyl]-3-[4-(trifluoromethyl)-3-{[3-
(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl}benzyl]imidazolidin-
4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-{3-[(2-pyrrolidin-l-yl-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-
6-yl)carbonyl]-4-(trifluoromethyl)benzyl}-5-(4,4,44rifluorobutyl)imidazolidin-4-one,
(5R)-5-(2-cyclopropylethyl)-3-[(4R)-6-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-3,4- dihydro-2H-chromen-4-yl]-5-(4-fluorophenyl)-2-iminoimidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-{3-[(4-pyridin-3-ylpiperazin-l-yl)carbonyl]-4-
(trifluoromethyl)benzyl}-5-(4,4,4-trifluorobutyl)imidazolidin-4-one,
(5R)-3-{3-[(4-acetylpiperazin-l-yl)carbonyl]-4-(trifluoromethyl)benzyl}-5-(4-fluorophenyl)-2- imino-5-(4,4,4-trifluorobutyl)imidazolidin-4-one,
(5R)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)phenyl]-3- methylbutyl}-5-(4-fluorophenyl)-2-imino-5-[(^
(5R)-3-[(4R)-6-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-l,l-dioxido-3,4-dihydro-2H- thiochromen-4-yl]-2-imino-5-(3-methylbutyl)-5-phenylimidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3- { [4-(methoxy acetyl)piperazin- 1 -yl] carbonyl } -4- (trifluoromethyl)benzyl]-5-(4,4,4-trifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-3-[3-({2-[2-(4-fluorophenyl)-l,3-oxazol-5-yl]-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-2-imino-5-(4,4,4 rifluorobutyl)imidazoli
4- one,
5- (2-cyclopropylethyl)-3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4- (trifluoromethyl)benzyl]-2-imino-5-[3-(trifluoromethyl)phenyl]imidazolidin-4-one,
4-{[5-{[(4R)-4-(4-fluorophenyl)-2-imino-5-oxo-4-(4,^^
(trifluoromethyl)phenyl] carbonyl } -N-methy lpiperazine- 1 -carboxamide,
(5R)-3-[(lR)-7-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-l,2,3,4-tetrahydronaphthalen- l-yl]-5-(4-fluorophenyl)-2-imino-5-(4,4,4 rifluorobutyl)imidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-({2-[4-(2H-l,2,3-triazol-2-yl)phenyl]-5,7-dihydro-6H- pyrrolo[3,4-d]pyrimidin-6-yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4- trifluorobutyl)imidazolidin-4-one, (5R)-3-{2-cyclopentyl-l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4- (trifluoromethyl)phenyl]ethyl}-5-(4-fluorophenyl)-2-imino-5-(3-methylbutyl)imidazolidin-4-o (5R)-5-(4-fluorophenyl)-2-imino-3-{3-[(4-phenylpiperazin-l-yl)carbonyl]-4- (trifluoromethyl)benzyl}-5-(4,4,4-trifluorobutyl)imidazolidin-4-one,
(5R)-3-{l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)phenyl]-3- methylbutyl}-5-(4-fluorophenyl)-2-imino-5-[(3R) etrahydrofuran-3-ylmethyl]imidazolidin^ (5R)-3-[3-{[2-(3,5-difluorophenyl)-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl]carbonyl}-4- (trifluoromethyl)benzyl]-5-(4-fluorophenyl)-^
(5R)-3-{3-[(2-cyclopropyl-5,6-dihydro[l,2,4]triazolo[l,5-a]pyrazin-7(8H)-yl)carbonyl]-4-
(trifluoromethyl)benzyl}-5-(4-fluorophenyl)-2-imino-5-(4,4,44rifluorobutyl)imidazolidin-4-on
(5R)-5-(4-fluorophenyl)-2-imino-3-{3-methyl-l-[4-(trifluoromethyl)-3-{[3-(trifluoromethyl)-5,6- dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl}phenyl]butyl}-5-[(3R)-tetrahydrofuran-3- ylmethyl]imidazolidin-4-one,
(5R)-3-[(lR)-7-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-l,2,3,4-tetrahydronaphthalen- l-yl]-2-imino-5-(3-methylbutyl)-5-phenylimidazolidin-4-one,
5-(2-cyclopropylethyl)-2-imino-5-[4-(trifluoromethyl)phenyl]-3-[4-(trifluoromethyl)-3-{[3- (trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl}benzyl]imidazolidm^
4- one,
(5R)-3-[3-(5 ,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(methylsulfonyl)benzyl]-2-imino-
5- (3-methylbutyl)-5-phenylimidazolidin-4-one,
(5R)-5-(4-fluorophenyl)-2-imino-5 4,4,4-trifluorobutyl)-3-[3-{[4-(2,2,2-trifluoroethyl)piperazin-l- y 1] carbonyl } -4-(trifluoromethyl)benzy l]imidazolidin-4-one,
(5R)-3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-2-imino- 5-phenyl-5-[(3R)4etrahydrofuran-3-ylmethyl]imidazolidin-4-one,
(5R)-5-(2-cyclopropylethyl)-3-[(lR)-7-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)- l,2,3,4 etrahydronaphthalen-l-yl]-5-(4-fluorophenyl)-2-iminoimidazolidin-4-one,
(5R)-3-[(lR)-7-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-l,2,3,4-tetrahydronaphthalen- l-yl]-5-(4-fluorophenyl)-2-imino-5-(tetrahydro-2H-pyran-4-yl)imidazolidin-4-one,
5- (2-cyclopropylethyl)-2-imino-5-[4-(trifluoromethyl)phenyl]-3-[4-(trifluoromethyl)-3-{[3- (trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl}benzyl]imidazolidin- 4-one,
(5R)-5-(4-fluorophenyl)-2-imino-3-[3-(3,4, 10,10a-tetrahydropyrazino[l,2-a]indol-2(lH)- ylcarbonyl)-4-(trifluoromethyl)benzyl]-5-(4,4,4-trifluorobutyl)imidazolidin-4-one,
(5R)-3-[3-({2-[l-(3-chlorophenyl)-lH-pyrazol-4-yl]-5,7-dihydro-6H-pyn-olo[3,4-d]pyrimidin-6- yl}carbonyl)-4-(trifluoromethyl)benzyl]-5-(4-fluorophenyl)-2-imino-5-(4,4,4- trifluorobutyl)imidazolidin-4-one,
6- {[5-{[4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l-yl]methyl}-2- (trifluoromethyl)phenyl]carbonyl}-6 -dihydro-5H-pyrrolo[3,4-d]pyrimidine-2-carbonitrile, methyl (6-{[5-{(lR)-1 (4R)-2-imino-4 3-methylbutyl)-5-oxo-4-phenylimidazolidin-l-yl]ethyl}-2- (trifluoromethyl)phenyl]carbonyl}-6 -dihydro-5H-pyrrolo[3,4-d]pyrimidin-2-yl)carbamate, 6-({5-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]-2-(trifluoromethyl)phenyl}carbo 6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidine-2-carbonitrile,
methyl (6-{[5-{[2-imino-4-(3-methylbutyl)-5-oxo-4-phenylimidazolidin-l-yl]methyl}-2- (trifluoromethyl)phenyl]carbonyl}-6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidin-2-yl)carbamate, 3-[3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-5,5-bis(4- fluorophenyl)-2-iminoimidazolidin-4-one,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-5,5-bis(4- fluorophenyl)-2-iminoimidazolidin-4-one,
6- { [5- { [2-imino-4-(2-methy lpropy l)-5 -oxo-4-phenylimidazolidin- 1 -y ljmethy 1 } -2-
(trifluoromethyl)phenyl]carbonyl}-6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidine-2-carbonitrile,
(5R)-3-{(lR)-l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-
(trifluoromethyl)phenyl]ethyl}-2-imino-5-(3-methylbutyl)-5-phenylimidazolidin-4-one,
3-[3-(5 J-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-2-imino-5-(2- methylpropyl)-5-phenylimidazolidin-4-one,
3-[3-(5 J-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-2-imino-5,5- diphenylimidazolidin-4-one,
3-[3-(5 J-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-5-(4- fluorophenyl)-2-imino-5-(3-methylbutyl)imidazolidin-4-one,
3-[3-(5 J-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-2-imino- 5,5-diphenylimidazolidin-4-one,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-5-(4- fluorophenyl)-2-imino-5-(3-methylbutyl)imidazolidin-4-one,
(5R)-3-{(lR)-l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-
(trifluoromethyl)phenyl]ethyl}-2-imino-5-(2-methylpropyl)-5-phenylimidazolidin-4-one,
6-{[5-{[2-imino-4-(3-methylbutyl)-5-oxo-4-phenylimidazolidin-l-yl]methyl}-2-
(trifluoromethyl)phenyl]carbonyl}-6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidine-2-carbonitrile,
(5R)-3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-2-imino-
5-(2-methylpropyl)-5-phenylimidazolidin-4-one,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-2-imino-5- (2-methylpropyl)-5-phenylimidazolidin-4-one,
3-[3-(5 J-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-2-imino-5-(3- methylbutyl)-5-phenylimidazolidin-4-one,
3- [3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-2-imino-5- (3-methylbutyl)-5-phenylimidazolidin-4-one,
4- ({5-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]-2-(trifluoromethyl)phenyl}carbonyl)- 1 -methylpiperazin-2-one, 2-imino-3-{3-[(7-methoxy-l,2,4,5-tetrahydro-3H-3-benzazepin-3-yl)carbonyl]-4-
(trifluoromethyl)benzyl}-5-(3-methylbutyl)-5-phenylimidazolidin-4-one,
1 -(1 - { [5- { [2-imino-4-(3-methylbutyl)-5-oxo-4-phenylimidazolidin- 1 -yl]methyl} -2-
(trifluoromethyl)phenyl]carbonyl}piperidin-4-yl)-l,3-dihydro-2H-benzimidazol-2-one,
4- { [5- { [2-imino-4-(2-methy lpropy l)-5 -oxo-4-phenylimidazolidin- 1 -y l]methy 1 } -2-
(trifluoromethyl)pheny 1] carbonyl } - 1 -methy lpiperazin-2-one,
4- {[5-{[4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l-yl]methyl}-2- (trifluoromethyl)pheny 1] carbonyl } - 1 -methy lpiperazin-2-one,
5- (4-fluorophenyl)-2-imino-5-(3-methylbutyl)-3-{3-[(2-phenyl-4,6-dihydro-5H-pyrrolo[3,4- d] [ 1 ,3] oxazol-5-yl)carbony 1] -4-(trifluoromethy l)benzyl } imidazolidin-4-one,
(3aR,6aR)-5-{[5-{[4-(4-fluorophenyl)-2-imino-4-(3-methylbutyl)-5-oxoimidazolidin-l-yl]methyl}-
2- (trifluoromethyl)phenyl]carbonyl}-3a-phenyltetrahydropyrrolo[3,4-c]pyrrole-l,3(2H,3aH)-dione,
4- ( { 5- [(2-imino-5-oxo-4,4-diphenylimidazolidin- 1 -y l)methy 1] -2- (trifluoromethy l)pheny 1 } carbonyl)piperazin-2-one,
methyl 4-({5-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]-2- (trifluoromethyl)pheny 1 } carbonyl)piperazine- 1 -carboxylate,
3- {3-[(3,3-difluoropyrrolidin-l-yl)carbonyl]-4-(trifluoromethyl)benzyl}-2-imino-5-(3-methylbutyl)-
5- phenylimidazolidin-4-one,
5-(4-fluorophenyl)-2-imino-5-(3-methylbutyl)-3-[3-{[(3aS,7aR)-7a-phenylhexahydroisoxazolo[4,3- c]pyridin-5(3H)-yl]carbonyl}-4-(trifluoromethyl)benzyl]imidazolidin-4-one,
5-(4-fluorophenyl)-2-imino-5-(3-methylbutyl)-3-{3-[(l-phenyl-l,4,6,7-tetrahydro-5H-pyrazolo[4,3- c]pyridin-5-yl)carbonyl]-4-(trifluoromethyl)benzyl}imidazolidin-4-one,
5,5-bis(4-fluorophenyl)-2-imino-3-{3-[(7-methoxy-l,2,4,5-tetrahydro-3H-3-benzazepin-3- yl)carbonyl]-4-(trifluoromethyl)benzyl}imidazolidin-4-one,
3-[3-(azetidin-l-ylcarbonyl)-4-(trifluoromethyl)benzyl]-2-imino-5-(3-methylbutyl)-5- phenylimidazolidin-4-one,
3-[3-(5 J-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-5-(4- fluorophenyl)-2-imino-5-(3-methylbutyl)imidazolidin-4-one,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-5-(4- fluorophenyl)-2-imino-5-(3-methylbutyl)imidazolidin-4-one,
5-(4-fluorophenyl)-2-imino-5-(3-methylbutyl)-3-{3-[(5aR,8aS)-5a,6,8,8a-tetrahydro-4H,7H- pyrrolo [3 ,4-b] [ 1 ,2,3] triazolo[ 1 ,5 -d] [ 1 ,4] oxazin-7-y lcarbonyl] -4- (trifluoromethyl)benzyl}irrddazolidin-4-one,
2-imino-5-(3-methylbutyl)-5-phenyl-3-[3-(pyrrolidin-l-ylcarbonyl)-4- (trifluoromethyl)benzyl]imidazolidin-4-one,
2- imino-5,5-diphenyl-3-{3-[(4-pyrimidin-2-ylpiperazin-l-yl)carbonyl]-4- (trifluoromethyl)benzyl}irrddazolidin-4-one,
3- {3-[(3,3-difluoropyrrolidin-l-yl)carbonyl]-4-(trifluoromethyl)benzyl}-5,5-bis(4-fluorophenyl)-2- iminoimidazolidin-4-one,
3-[3-(azetidin-l-ylcarbonyl)-4-(trifluoromethyl)benzyl]-5,5-bis(4-fluorophenyl)-2- iminoimidazolidin-4-one,
(5S)-3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-(trifluoromethyl)benzyl]-2-imino- 5-(2-methylpropyl)-5-phenylimidazolidin-4-one,
5-(4-fluorophenyl)-2-imino-5-(3-methylbutyl)-3-[3-{[(3aS,6aR)-3-(phenylamino)-3a,4,6,6a- tetrahydro-5H-pyrrolo[3,4-d]isoxazol-5-yl]carbonyl}-4-(trifluoromethyl)benzyl]imidazolidin-4-one,
3-{3-[(3,3-dimethylpyrrolidin-l-yl)carbonyl]-4-(trifl^
5- phenylimidazolidin-4-one,
3 - { 3 -[(3,3 -difluoropyrrolidin- 1 -yl)carbony 1] -4-(trifluoromethy l)benzy 1 } -2-imino-5 -(2- methylpropyl)-5-phenylimidazolidin-4-one,
N-[(5- {[5- { [4-(4-fluorophenyl)-2-imino-4-(3-methylbutyl)-5-oxoimidazolidin-l -yl]methyl} -2- (trifluoromethyl)phenyl]carbonyl}-4,5,6,74etrahydrofuro[3,2-c]pyridin-2-yl)methyl]acetami 3-[3-(azetidin-l-ylcarbonyl)-4-(trifluoromethyl)benzyl]-2-imino-5-(2-methylpropyl)-5- phenylimidazolidin-4-one,
5,5-bis(4-fluorophenyl)-2-imino-3-[3-(pyrrolidin-l-ylcarbonyl)-4- (trifluoromethyl)benzyl]imidazolidin-4-one,
6- [(3-{[4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l-yl]methyl}phenyl)carbonyl]-6,7- dihydro-5H-pyrrolo[3,4-d]pyrimidine-2-carbonitrile,
methyl {6-[(3-{[(4R)-2-imino-4-(3-methylbutyl)-5-oxo-4-phenylimidazolidin-l- yl]methyl}phenyl)carbonyl]-6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidin-2-yl}carbamate,
2- imino-5,5-diphenyl-3-(3-{[3-(trifluoromethyl)-4,6-dihydropyrrolo[3,4-c]pyrazol-5(lH)- y 1] carbonyl } benzy l)imidazolidin-4-one,
3- {(lR)-l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)phenyl]ethyl}-2-imino-5,5- diphenylimidazolidin-4-one,
3-{(lR)-l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)phenyl]ethyl}-5,5-bis(4- fluorophenyl)-2-iminoimidazolidin-4-one,
3-{(lR)-l-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)phenyl]ethyl}-2-imino-5,5- diphenylimidazolidin-4-one,
methyl {6-[(3- { [2-imino-4-(2-methylpropyl)-5-oxo-4-phenylimidazolidin- 1 - yl]methyl}phenyl)carbonyl]-6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidin-2-yl}carbamate,
methyl {6-[(3-{[4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l-yl]methyl}phenyl)carbonyl]-
6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidin-2-yl}carbamate,
methyl [6-({3-[(2-irnino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)-6,7- dihydro-5H-pyrrolo[3,4-d]pyrimidin-2-yl]carbamate,
6-[(3-{[2-irnino-4-(3-methylbutyl)-5-oxo-4-phenylimidazolidin-l-yl]methyl}phenyl)carbonyl]-6,7- dihydro-5H-pyrrolo[3,4-d]pyrimidine-2-carbonitrile,
5-biphenyl-4-yl-3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)benzyl]-2-imino-5- phenylimidazolidin-4-one,
3-[3-(5 J-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)benzyl]-2-imino-5-(3-methylbutyl)-5- phenylimidazolidin-4-one,
3- {3-[(4-fluoro-l,3-dihydro-2H-isoindol-2-yl)carbonyl]benzyl}-2-imino-5,5-diphenylimidazolidin-
4- one,
3-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-5,5-bis(4-fluorophenyl)-2-iminoimidazolidin-4- one,
3-{3-[(4-fluoro-l,3-dihydro-2H-isoindol-2-yl)carbonyl]benzyl}-5,5-bis(4-fluorophenyl)-2- iminoimidazolidin-4-one,
3-{3-[(5,6-difluoro-l,3-dihydro-2H-isoindol-2-yl)carbonyl]benzyl}-5,5-bis(4-fluorophenyl)-2- iminoimidazolidin-4-one,
3-{(lR)-l-[3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)phenyl]ethyl}-5,5-bis(4- fluorophenyl)-2-iminoimidazolidin-4-one,
6-({3-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)-6,7-dihydro-5H- pyrrolo[3,4-d]pyrimidine-2-carbonitrile,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)benzyl]-5,5-bis(4-fluorophenyl)-2- iminoimidazolidin-4-one,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)benzyl]-5-(4'-fluorobiphenyl-4-yl)-2- imino-5-phenylimidazolidin-4-one,
5,5-bis(4-fluorophenyl)-2-imino-3-[(lR)-l-{3-[(l-methyl-4,6-dihydropyrrolo[3,4-c]pyrazol-5(lH)- yl)carbonyl] phenyl} ethyl] imidazolidin-4-one,
3- {3-[(5-fluoro-l,3-dihydro-2H-isoindol-2-yl)carbonyl]benzyl}-2-imino-5,5-diphenylimidazolidin-
4- one,
3-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-2-imino-5-(3-methylbutyl)-5- phenylimidazolidin-4-one,
5,5-bis(4-fluorophenyl)-2-imino-3-{3-[(2-methoxy-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6- yl)carbonyl]benzyl}imidazolidin-4-one,
5- ({3-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)-2-methyl-3,4,5,6- tetrahy dropyrrolo[3,4-d] [ 1 ,3]thiazol- 1 -ium,
(5R)-3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-5-fluorobenzyl]-2-imino-5-(3- methylbutyl)-5-phenylimidazolidin-4-one,
3-[3-(5 ,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)benzyl]-2-imino-5-(3-methylbutyl)-5- phenylimidazolidin-4-one,
ethyl 3-chloro-7-({3-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)- 5,6,7,8-tetrahydroimidazo[l,2-a]pyrazine-2-carboxylate,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)benzyl]-5-(3-fluorophenyl)-2-imino-5-(2- methylpropyl)imidazolidin-4-one,
5,5-bis(4-fluorophenyl)-2-imino-3-(3-{[2-(trifluoromethyl)-5,7-dihydro-6H-pyrrolo[3,4- d]pyrimidin-6-yl]carbonyl}benzyl)imidazolidin-4-one,
5-(3-bromophenyl)-5-(4-fluorophenyl)-2-imino-3-(3-{[3-(trifluoromethyl)-4,6-dihydropyrrolo[3,4- c] py razol-5( 1 H)-y 1] carbonyl } benzyl)imidazolidin-4-one,
5- (4-bromophenyl)-3-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-2-imino-5- phenylimidazolidin-4-one,
2- imino-3-{3-[(6-methyl-l,3-dihydro-2H-pyrrolo[3,4-c]pyridin-2-yl)carbonyl]benzyl}-5,5- diphenylimidazolidin-4-one,
3- [3-(5 J-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)benzyl]-5,5-bis(4-fluorophenyl)-2- iminoimidazolidin-4-one,
3-{(lR)-l-[3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)phenyl]ethyl}-2-imino-5-(3- methylbutyl)-5-phenylimidazolidin-4-one,
3-{(lR)-l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)phenyl]ethyl}-2-imino-5-(3- methylbutyl)-5-phenylimidazolidin-4-one,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyridin-6-ylcarbonyl)benzyl]-2-imino-5-(3-methylbutyl)-5- phenylimidazolidin-4-one,
3 -[3 -(5 ,7-dihy dro-6H-pyrrolo [3,4-b] pyrazin-6-y lcarbony l)benzy 1] -2-imino-5 ,5 - diphenylimidazolidin-4-one,
3-(3-{[4-(dimethylamino)-5 ,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl]carbonyl}benzyl)-5,5-bis(4- fluorophenyl)-2-iminoimidazolidin-4-one,
methyl 4-[(3-{(lR)-l-[2-imino-4-(3-methylbutyl)-5-oxo-4-phenylimidazolidin-l- y 1] ethyl } pheny l)carbonyl] piperazine- 1 -carboxy late,
6- [(3-{[2-imino-4-(2-methylpropyl)-5-oxo-4-phenylimidazolidin-l-yl]methyl}phenyl)carbonyl]-6,7- dihydro-5H-pyrrolo[3,4-d]pyrimidine-2-carbonitrile,
3-{[5-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)cyclohexa-l,5-dien-l-yl]methyl}-2-imino-5-(2- methylpropyl)-5-phenylimidazolidin-4-one,
3-{ l-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)phenyl]ethyl}-2-imino-5,5-diphenylimidazolidin-4- one,
3- {l-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)phenyl]propyl}-2-imino-5,5-diphenylimidazolidin-
4- one,
5,5-bis(4-fluorophenyl)-2-imino-3-{3-[(6-methyl-l,3-dihydro-2H-pyrrolo[3,4-c]pyridin-2- yl)carbonyl]benzyl}imidazolidin-4-one,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)benzyl]-2-imino-5-(3-methylbutyl)-5- phenylimidazolidin-4-one,
3-{(lR)-l-[3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)phenyl]ethyl}-2-imino-5,5- diphenylimidazolidin-4-one,
3-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-5-(4-fluorophenyl)-2-imino-5-(3- methylbutyl)imidazolidin-4-one,
2-imino-5,5-diphenyl-3-{3-[(4-pyrimidin-2-ylpiperazin-l-yl)carbonyl]benzyl}imidazolidin-4- 3-{(lR)-l-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)phenyl]ethyl}-5,5-bis(4-fluorophenyl)-2- iminoimidazolidin-4-one,
3-{3-[(5-fluoro-l,3-dihydro-2H-isoindol-2-yl)carbonyl]benzyl}-5,5-bis(4-fluorophenyl)-2- iminoimidazolidin-4-one,
2- imino-5,5-diphenyl-3-(3-{[5-(trifluoromethyl)-l,3-dihydro-2H-isoindol-2- y 1] carbonyl } benzy l)imidazolidin-4-one,
methyl 4-[(3-{[4-(3'-cyanobiphenyl-3-yl)-4-(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l- yl]methyl}phenyl)carbonyl]piperazine-l-carboxylate,
3- [3-(5 ,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)benzyl]-2-imino-5-(2-methylpropyl)-5- phenylimidazolidin-4-one,
3-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-2-imino-5-phenyl-5-propylimidazolidin-4-one,
3-{(lR)-l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)phenyl]ethyl}-2-imino-5-(2- methylpropyl)-5-phenylimidazolidin-4-one,
3-(3-{[4-(2-chlorophenyl)piperazin-l-yl]carbonyl}benzyl)-2-imino-5,5-diphenylimidazolidin-4-one, methyl 4- [(3 - { [4-(3 -bromophenyl)-4-(4-fluorophenyl)-2-imino-5-oxoimidazolidin- 1 - yl]methyl}phenyl)carbonyl]piperazine-l-carboxylate,
2-imino-3-{3-[(l-methyl-4,6-dihydropyrrolo[3,4-c]pyrazol-5(lH)-yl)carbonyl]benzyl}-5,5- diphenylimidazolidin-4-one,
2-imino-5,5-diphenyl-3-(3-{[2-(trifluoromethyl)-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6- y 1] carbonyl } benzy l)imidazolidin-4-one,
6-({3-[(2-imino-5-oxo-4-phenyl-4-propyliinidazolidin-l-yl)methyl]phenyl}carbonyl)-6,7-dihydro- 5H-pyrrolo[3,4-d]pyrimidine-2-carbonitrile,
2-imino-5-(3-methylbutyl)-5-phenyl-3-{3-[(4-pyrimidin-2-ylpiperazin-l- yl)carbonyl]benzyl}imidazolidin-4-one,
ethyl 7-({3-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl} carbonyl)-5, 6,7,8- tetrahy droimidazo [ 1 ,2-a] pyrazine-2-carboxy late,
2- imino-3-(3-{[3-(methylsulfanyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- yl] carbonyl} benzy l)-5, 5 -diphenylimidazolidin-4-one,
ethyl 3-chloro-7-({3-[(2-irnino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)- 5,6,7,8-tetrahydroimidazo[l,2-a]pyrazine-2-carboxylate,
5-(4-fluorophenyl)-2-imino-5-pyridin-2-yl-3-(3-{[3-(trifluoromethyl)-4,6-dihydropyrrolo[3,4- c] py razol-5( 1 H)-y 1] carbonyl } benzyl)imidazolidin-4-one,
3- [3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)benzyl]-2-imino-5-(3-methylbutyl)-5- phenylimidazolidin-4-one,
3 -(3- {[2-(cyclopropy lcarbonyl)-5,6-dihy droimidazo [l,2-a]py razin-7(8H)-yl] carbonyl }benzyl)-2- imino-5,5-diphenylimidazolidin-4-one,
3-chloro-7-({3-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)-N-methyl- 5,6,7,8 etrahydroimidazo[l,2-a]pyrazine-2-carboxarnide, 3-[3-(5 J-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)benzyl]-5-(4-fluorophenyl)-2-imino-5-(3- methylbutyl)imidazolidin-4-one,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)benzyl]-2-imino-5-(2-methylpropyl)-5- phenylimidazolidin-4-one,
5,5-bis(4-fluorophenyl)-2-imino-3-{3-[(2-methyl-5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6- yl)carbonyl]benzyl}imidazolidin-4-one,
methyl 4-[(3-{[2-imino-4-(3-methylbutyl)-5-oxo-4-phenylimidazolidin-l- yl]methyl}phenyl)carbonyl]piperazine-l-carboxylate,
3-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-5-(4-fluorophenyl)-2-imino-5-pyridin-2- ylimidazolidin-4-one,
2- imino-5 3-methylbutyl)-3-{3 (2-methyl-4,6-dihydropyrrolo[3,4-d]imidazol-5(lH)- yl)carbonyl]benzyl}-5-phenylimidazolidin-4-one,
3- [3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)-5-fluorobenzyl]-2-imino-5,5- diphenylimidazolidin-4-one,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyridin-6-ylcarbonyl)benzyl]-2-imino-5-(2-methylpropyl)-5- phenylimidazolidin-4-one,
benzyl 4-( {3 - [(2-imino-5-oxo-4,4-dipheny limidazolidin- 1 -y l)methyl] phenyl } carbony l)piperazine- 1 - carboxylate,
3-{(lR)-l-[3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)phenyl]ethyl}-2-imino-5-(2- methylpropyl)-5-phenylimidazolidin-4-one,
3-{l-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-4-(4-fluorophenyl)-2-imino-5- oxoimidazolidin-4-yl}benzonitrile,
3-{ l-[3-(5 -dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)phenyl]propyl}-2-imino-5,5- diphenylimidazolidin-4-one,
3-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-5-(3-fluorophenyl)-2-imino-5-(2- methylpropyl)imidazolidin-4-one,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)benzyl]-5-(3-fluorophenyl)-2-imino-5-(3- methylbutyl)imidazolidin-4-one,
3-{ l-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)phenyl]ethyl}-2-imino-5,5- diphenylimidazolidin-4-one,
5,5-bis(4-fluorophenyl)-3-(3-{[4-(furan-2-ylcarbonyl)piperazin-l-yl]carbonyl}benzyl)-2- iminoimidazolidin-4-one,
3-(3-{[4-(furan-2-ylcarbonyl)piperazin-l-yl]carbonyl}benzyl)-2-imino-5-(2-methylpropyl)-5- phenylimidazolidin-4-one,
3-{3-[(7-fluoro-3,4-dihydroisoquinolin-2(lH)-yl)carbonyl]benzyl}-2-imino-5,5- diphenylimidazolidin-4-one,
3-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-5-(3-fluorophenyl)-2-imino-5-(3- methylbutyl)imidazolidin-4-one, 3- {3-[(5,6-dichloro-l,3-dihydro-2H-isoindol-2-yl)carbonyl]benzyl}-2-imino-5,5- diphenylimidazolidin-4-one,
4- [(3-{(lR)-l-[2-imino-4-(3-methylbutyl)-5-oxo-4-phenylimidazolidin-l-yl]ethyl}phenyl)carbonyl]- 1 -methylpiperazin-2-one,
5- biphenyl-4-yl-3-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-2-imino-5-phenylimidazolidin- 4-one,
2- imino-3-{3-[(2-methyl-4,6-dihydropyrrolo[3,4-d]imidazol-5(lH)-yl)carbonyl]benzyl}-5-(2- methylpropyl)-5-phenylimidazolidin-4-one,
5,5-bis(4-fluorophenyl)-2-imino-3-(3-{[2-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[l,5-a]pyrazin- 7(8H)-yl]carbonyl}benzyl)imidazolidin-4-one,
3- { 1 -[(3- { [4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin- 1 - yl]methyl}phenyl)carbonyl]piperidin-4-yl}-5-fluoro-l,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one,
3-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-2-imino-5-(3-methylbutyl)-5-(3-pyridin-3- ylphenyl)imidazolidin-4-one,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)benzyl]-2-imino-5-phenyl-5- propylimidazolidin-4-one,
methyl 4-[(3-{(lR)-l-[2-imino-4-(2-methylpropyl)-5-oxo-4-phenylimidazolidin-l- y 1] ethyl } pheny l)carbonyl] piperazine- 1 -carboxy late,
5,5-bis(4-fluorophenyl)-2-imino-3-{3-[(4-pyrimidin-2-ylpiperazin-l- yl)carbonyl]benzyl}imidazolidin-4-one,
3- {3-[(5-chloro-3,4-dihydroisoquinolin-2(lH)-yl)carbonyl]benzyl}-2-imino-5,5- diphenylimidazolidin-4-one,
2- imino-3-(3-{[5-methyl-3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- yl]carbonyl}benzyl)-5,5-diphenylimidazolidin-4-one,
4- [(3-{[2-irnino-4-(3-methylbutyl)-5-oxo-4-phenylimidazolidin-l-yl]methyl}phenyl)carbonyl]-l- methylpiperazin-2-one,
3- [3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-5-(4-fluorophenyl)-2-imino-5-(3-pyridin-3- ylphenyl)imidazolidin-4-one,
5,5-bis(4-fluorophenyl)-2-imino-3-{3-[(4-pyridin-2-ylpiperazin-l-yl)carbonyl]benzyl}imidazolidm^
4- one,
3-{3-[(5,6-dichloro-l,3-dihydro-2H-isoindol-2-yl)carbonyl]benzyl}-5,5-bis(4-fluorophenyl)-2- iminoimidazolidin-4-one,
methyl 2-({3-[(2-irnino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)-2,3-dihydro- lH-isoindole-l-carboxylate,
methyl 4-[(3-{[4-(4-fluorophenyl)-2-imino-4-(2-methylpropyl)-5-oxoimidazolidin-l- yl]methyl}phenyl)carbonyl]piperazine-l-carboxylate,
5 -(3 -bromophenyl)-3 -[3-( 1 ,3 -dihy dro-2H-isoindol-2-ylcarbonyl)benzyl] -5 -(4-fluorophenyl)-2- iminoimidazolidin-4-one, 2-imino-3-{3-[(2-methyl-5 ,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-yl)carbonyl]benzyl}-5,5- diphenylimidazolidin-4-one,
2- imino-5,5-diphenyl-3-(3-{[3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- y 1] carbonyl } benzy l)imidazolidin-4-one,
3'-{ l-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-4-(4-fluorophenyl)-2-imino-5- oxoimidazolidin-4-yl}biphenyl-3-carbonitrile,
3- {l-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-4-(4-fluorophenyl)-2-imino-5- oxoimidazolidin-4-yl}benzamide,
3- [3-(7,8-dihydropyrido[3,4-b]pyrazin-6(5H)-ylcarbonyl)benzyl]-2-imino-5,5-diphenylimidazolidin-
4- one,
3-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)-5-fluorobenzyl]-2-imino-5,5-diphenylimidazolidin-4- one,
3-(3-{[3-ethenyl-2-(trifluoromethyl)-5,6-dihydroimidazo[l,2-a]pyrazin-7(8H)-yl]carbonyl}benzyl)- 2-imino-5 ,5 -diphenylimidazolidin-4-one,
methyl 4-({3-fluoro-5-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l- yl)methyl] phenyl } carbony l)piperazine- 1 -carboxylate,
2- ({3-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)-N,N-dimethyl-2,3- dihydro-lH-isoindole-l-carboxamide,
3- (3-{[5,5-dimethyl-3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- yl]carbonyl}benzyl)-2-imino-5,5-diphenylimidazolidin-4-one,
2- imino-5,5-diphenyl-3-(3-{[2-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[l,5-a]pyrazin-7(8H)- y 1] carbonyl } benzy l)imidazolidin-4-one,
3- [3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)benzyl]-2-imino-5-phenyl-5- propylimidazolidin-4-one,
2- imino-5-(2-methylpropyl)-5-phenyl-3-{3-[(4-pyrimidin-2-ylpiperazin-l- yl)carbonyl]benzyl}imidazolidin-4-one,
3- [3-(5 J-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)benzyl]-2-imino-5-(3-methylbutyl)-5-(3- pyridin-3-ylphenyl)imidazolidin-4-one,
methyl 4-[(3-{[4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l- yl]methyl}phenyl)carbonyl]piperazine-l -carboxylate,
3-{l-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-4-(4-fluorophenyl)-2-imino-5- oxoimidazolidin-4-yl}-N,N-dimethylbenzamide,
2- imino-5 ,5 -diphenyl-3 - {3 -[(4-pyridin-2-y lpiperazin- 1 -yl)carbonyl] benzy 1 } imidazolidin-4-one,
3- {3-[(6-fluoro-3,4-dihydroisoquinolin-2(lH)-yl)carbonyl]benzyl}-2-imino-5,5- diphenylimidazolidin-4-one,
4- ({3-fluoro-5-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)-N,N- dimethylpiperazine- 1 -carboxamide,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrirrddin-6-ylcarbonyl)benzyl]-2-imino-5,5- diphenylimidazolidin-4-one,
5,5-bis(4-fluorophenyl)-2-imino-3-{3-[(l-methyl-l,3-dihydro-2H-isoindol-2- yl)carbonyl]benzyl}imidazolidin-4-one,
3-[3-(2,3-dihydro-lH-indol-l-ylcarbonyl)benzyl]-2-imino-5,5-diphenylimidazolidin-4-one,
3-[3-(3,4-dihydro-2J-naphthyridin-2(lH)-ylcarbonyl)b^
one,
1- { l-[(3-{[2-imino-4-(3-methylbutyl)-5-oxo-4-phenylimidazolidin-l- yl]methyl}phenyl)carbonyl]piperidin-4-yl}-l,3-dihydro-2H-benzimidazol-2-one,
3-(3-{[4-(2-chlorophenyl)piperazin-l-yl]carbonyl}benzyl)-2-imino-5-(2-methylpropyl)-5- phenylimidazolidin-4-one,
1 - { 1 -[(3- { [4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin- 1 - yl]methyl}phenyl)carbonyl]piperidin-4-yl}-5-fluoro-l,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one,
5,5-bis(4-fluorophenyl)-2-imino-3-(3-{[5-(trifluoromethyl)-l,3-dihydro-2H-isoindol-2- y 1] carbonyl } benzyl)imidazolidin-4-one,
3-[3-(3,4-dihydroisoquinolin-2(lH)-ylcarbonyl)benzyl]-2-imino-5-(2-methylpropyl)-5- phenylimidazolidin-4-one,
3- { 1 -[(3- { [4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin- 1 - yl]methyl}phenyl)carbonyl]piperidin-4-yl}-l,3-benzoxazol-2(3H)-one,
2- imino-5 -phenyl-5 -propyl-3 - { 3- [(4-pyrimidin-2-y Ipiperazin- 1 -yl)carbony 1] benzyl } imidazolidin-4- one,
2- imino-5-(2-methylpropyl)-5-phenyl-3-{3-[(4-pyridin-2-ylpiperazin-l- yl)carbonyl]benzyl}imidazolidin-4-one,
3- {3-[(5,6-dichloro-l-methyl-l,3-dihydro-2H-isoindol-2-yl)carbonyl]benzyl}-2-imino-5,5- diphenylimidazolidin-4-one,
3 - { 3 -[(4-acetylpiperazin- 1 -y l)carbonyl] -5-fluorobenzy 1 } -2-imino-5,5 -diphenylimidazolidin-4-one,
2- imino-5,5-diphenyl-3-{3-[(4-phenylpiperazin-l-yl)carbonyl]benzyl}imidazolidin-4-one,
3- {(lS)-l-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)phenyl]ethyl}-5,5-bis(4-fluorophenyl)-2- iminoimidazolidin-4-one,
4- ({3-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)-N-(l- methy lethyl)piperazine- 1 -carboxamide,
3- chloro-7-({3-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)-N,N- dimethyl-5,6,7,8-tetrahydroiinidazo[l,2-a]pyrazine-2-carboxarnide,
4- [(3-{(lR)-l-[2-irnino-4-(2-methylpropyl)-5-oxo-4-phenylimidazolidin-l- yl]ethyl}phenyl)carbonyl]-l-methylpiperazin-2-one,
2-imino-5,5-diphenyl-3-(3-{[3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- y 1] carbonyl } benzy l)imidazolidin-4-one,
methyl 4-({3-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)piperazine-l- carboxylate, 3-{(lS)-l-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)phenyl]ethyl}-2-imino-5,5- diphenylimidazolidin-4-one,
1 - { 1 -[(3- { [4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin- 1 - yl]methyl}phenyl)carbonyl]piperidin-4-yl}-l,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one,
2-imino-3-{3-[(4-phenoxypiperidin-l-yl)carbonyl]benzyl}-5,5-diphenylimidazolidin-4-one, 2-imino-5,5-diphenyl-3-(3-{[7-(trifluoromethyl)-3,4-dihydroisoquinolin-2(lH)- y 1] carbonyl } benzy l)imidazolidin-4-one,
3 - { 3 -[(4-acetylpiperazin- 1 -y l)carbonyl] benzyl } -5,5 -bis(4-fluoropheny l)-2-iminoimidazolidin-4-one,
2- imino-5,5-diphenyl-3-(3-{[6-(trifluoromethyl)-3,4-dihydroisoquinolin-2(lH)- y 1] carbonyl } benzy l)imidazolidin-4-one,
3- {3-[(3-benzyl-5,6-dihydroimidazo[l,2-a]pyrazin-7(8H)-yl)carbonyl]benzyl}-2-imino-5,5- diphenylimidazolidin-4-one,
1 - { 1 -[(3- { [4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin- 1 - yl]methyl}phenyl)carbonyl]piperidin-4-yl}-l,3-dihydro-2H-benzimidazol-2-one,
3-[3-(5,6-dihydroiinidazo[l,2-a]pyrazin-7(8H)-ylcarbonyl)benzyl]-2-imino-5,5- diphenylimidazolidin-4-one,
2- imino-3-{3-[(3-methoxy-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl)carbonyl]benzyl}-5,5- diphenylimidazolidin-4-one,
3- (3-fluoro-5-{[3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- yl]carbonyl}benzyl)-2-imino-5,5-diphenylimidazolidin-4-one,
2-irnino-5-(2-methylpropyl)-5-phenyl-3-{3-[(4-phenylpiperazin-l-yl)carbonyl]benzyl}imidazolidin-
4- one,
2-imino-5,5-diphenyl-3-(3-{[2-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[l,5-a]pyrazin-7(8H)- y 1] carbonyl } benzy l)imidazolidin-4-one,
ethyl [ 1 -( {3 -fluoro-5 -[(2-imino-5 -oxo-4,4-dipheny limidazolidin- 1 - yl)methyl] phenyl }carbonyl)piperidin-4-yl] carbamate,
2-imino-3-{3-[(2-methyl-5,7-dihydro-6H-pyrrolo[3,4-d]pyrirnidin-6-yl)carbonyl]benzyl}-5-phenyl-
5- propylimidazolidin-4-one,
5 -fluoro- 1 - { 1 -[(3- { [2-imino-4-(2-methy lpropy l)-5 -oxo-4-phenylimidazolidin- 1 - yl]methyl}phenyl)carbonyl]piperidin-4-yl}-l,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one, methyl 4-[(3-{[4-(3-carbamoylphenyl)-4-(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l- yl]methyl}phenyl)carbonyl]piperazine-l-carboxylate,
methyl 4-[(3-{[4-(3-fluorophenyl)-2-imino-4-(3-methylbutyl)-5-oxoimidazolidin-l- yl]methyl}phenyl)carbonyl]piperazine-l-carboxylate,
2-irnino-5,5-diphenyl-3-(3-{[2-(trifluoromethyl)-5,6-dihydroimidazo[l,2-a]pyrazin-7(8H)- y 1] carbonyl } benzy l)imidazolidin-4-one,
methyl 4-[(3-{[4-(3-cyanophenyl)-4-(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l- yl]methyl}phenyl)carbonyl]piperazine-l-carboxylate, 4-[(3- { [2-imino-4-(3-methylbutyl)-5-oxo-4-phenylimidazolidin-l - yl]methyl}phenyl)carbonyl]piperazin-2-one,
3 - { 3 -[(4-acetylpiperazin- 1 -yl)carbony 1] benzyl } -2-imino-5 ,5-diphenylimidazolidin-4-one,
4-({3-fluoro-5-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)-l- methylpiperazin-2-one,
methyl 4-[(3-{[2-imino-4-(3-methylbutyl)-5-oxo-4-(3-pyridin-3-ylphenyl)imidazolidin-l- yl]methyl}phenyl)carbonyl]piperazine-l-carboxylate,
ethyl [l-({3-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)piperidin-4- yl] carbamate,
3- {3-[(3-ethyl-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl)carbonyl]benzyl}-2-imino-5,5- diphenylimidazolidin-4-one,
4- ( { 3- [(2-imino-5-oxo-4,4-diphenylimidazolidin- 1 -yl)methyl]phenyl } carbonyl)-N,N- dimethylpiperazine- 1 -carboxamide,
4-({3-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)-l-methylpiperazin-
2- one,
3- (3-{[4-(4-fluorophenyl)piperazin-l-yl]carbonyl}benzyl)-2-imino-5,5-diphenylimidazolidin-4-one, 3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyridin-6-ylcarbonyl)benzyl]-2-imino-5-(3-methylbutyl)-5-(3- pyridin-3-ylphenyl)imidazolidin-4-one,
N ert-butyl-7-({3-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)-5,6,7,8- tetrahydro[l,2,4]triazolo[4,3-a]pyrazine-3-carboxamide,
3-[4-chloro-3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)benzyl]-2-imino-5,5- diphenylimidazolidin-4-one,
6-[(5-{[4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l-yl]methyl}-2-chlorophenyl)carbonyl]-
6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidine-2-carbonitrile,
6-[(2-chloro-5-{[2-imino-4-(2-methylpropyl)-5-oxo-4-phenylimidazolidin-l- yl]methyl}phenyl)carbonyl]-6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidine-2-carbonitrile,
3-[4-chloro-3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)benzyl]-2-imino-5-(2- methylpropyl)-5-phenylimidazolidin-4-one,
3-[4-chloro-3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyridin-6-ylcarbonyl)benzyl]-2-imino-5-(2- methylpropyl)-5-phenylimidazolidin-4-one,
6-({2-chloro-5-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)-6,7- dihydro-5H-pyrrolo[3,4-d]pyrimidine-2-carbonitrile,
3-[4-chloro-3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyridin-6-ylcarbonyl)benzyl]-2-imino-5,5- diphenylimidazolidin-4-one,
5,5-bis(4-chlorophenyl)-3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4- methoxybenzyl]-2-iminoimidazolidin-4-one,
methyl {6-[(5-{[4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l-yl]methyl}-2- fluorophenyl)carbonyl]-6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidin-2-yl}carbamate, 3- [3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)-4-methoxybenzyl]-2-imino-5,5-diphenylimid
4- one,
3-{3-[(4-fluoro-l,3-dihydro-2H-isoindol-2-yl)carbonyl]-4-methoxybenzyl}-2-imino-5,5- diphenylimidazolidin-4-one,
3-[4-chloro-3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)benzyl]-2-imino-5-(2- methylpropyl)-5-phenylimidazolidin-4-one,
3-[4-chloro-3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)benzyl]-2-imino-5,5- diphenylimidazolidin-4-one,
3-[4-chloro-3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)benzyl]-5,5-bis(4-fluorophenyl)-2- iminoimidazolidin-4-one,
3-[4-chloro-3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)benzyl]-5,5-bis(4- fluorophenyl)-2-iminoimidazolidin-4-one,
3- {4-chloro-3-[(5-fluoro-l,3-dihydro-2H-isoindol-2-yl)carbonyl]benzyl}-5,5-bis(4-fluorophenyl)-2- iminoimidazolidin-4-one,
4- ({2-chloro-5-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)-l- methylpiperazin-2-one,
3- [3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-fluorobenzyl]-2-imino-5,5- diphenylimidazolidin-4-one,
4- ({2-chloro-5-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)piperazin-2- one,
methyl 4-[(5-{[4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l-yl]methyl}-2- chlorophenyl)carbonyl]piperazine-l-carboxylate,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-4-fluorobenzyl]-5,5-bis(4-fluorophenyl)-
2- iminoimidazolidin-4-one,
1 -[ 1 -( {2-chloro-5-[(2-imino-5-oxo-4,4-diphenylimidazolidin- 1 - yl)methyl]phenyl}carbonyl)piperidin-4-yl]-l,3-dihydro-2H-benzimidazol-2-one,
6-({2-fluoro-5-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]phenyl}carbonyl)-6,7- dihydro-5H-pyrrolo[3,4-d]pyrimidine-2-carbonitrile,
3- [3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)-4-fluorobenzyl]-2-imino-5,5- diphenylimidazolidin-4-one,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-b]pyridin-6-ylcarbonyl)-4-fluorobenzyl]-2-imino-5,5- diphenylimidazolidin-4-one,
3-[3-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)-4-methoxybenzyl]-2-imino-5,5- diphenylimidazolidin-4-one,
5,5-bis(4-chlorophenyl)-3-[3-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)-4-methoxybenzyl]-2- iminoimidazolidin-4-one,
(5R)-3-{[5-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-6-(trifluoromethyl)pyridin-3- yl]methyl}-2-imino-5-(2-methylpropyl)-5-phenylimidazolidin-4-one, 2- (benzyloxy)ethyl 4-[(5-{[4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l-yl]methyl}pyridin-
3- yl)carbonyl]piperazine-l-carboxylate,
l-{ l-[(5-{[4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l-yl]methyl}pyridin-3- yl)carbonyl]piperidin-4-yl}-l,3-dihydro-2H-benzimidazol-2-one,
6-[(5-{[4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l-yl]methyl}pyridin-3-yl)carbonyl]-6,7- dihydro-5H-pyrrolo[3,4-d]pyrimidine-2-carbonitrile,
3-{[5-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)pyridin-3-yl]methyl}-5,5-bis(4-fluorophenyl)-2- iminoimidazolidin-4-one,
benzyl 4-[(5-{[4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l-yl]methyl}pyridin-3- yl)carbonyl]piperazine-l-carboxylate,
3-{[4-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)pyridin-2-yl]methyl}-2-imino-5,5- diphenylimidazolidin-4-one,
6-({2-[(2-imino-5-oxo-4,4-diphenylimidazolidin-l-yl)methyl]pyridin-4-yl}carbonyl)-6,7-dihydro- 5H-pyrrolo[3,4-d]pyrimidine-2-carbonitrile,
3-{[4-(l,3-dihydro-2H-isoindol-2-ylcarbonyl)pyridin-2-yl]methyl}-5,5-bis(4-fluorophenyl)-2- iminoimidazolidin-4-one,
3-{[4-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)pyridin-2-yl]methyl}-2-imino-5,5- diphenylimidazolidin-4-one,
3-{[4-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)pyridin-2-yl]methyl}-2-imino-5-(3- methylbutyl)-5-phenylimidazolidin-4-one,
3-{[4-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)pyridin-2-yl]methyl}-2-imino-5,5- diphenylimidazolidin-4-one,
3-{[4-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)pyridin-2-yl]methyl}-5,5-bis(4- fluorophenyl)-2-iminoimidazolidin-4-one,
3-{[4-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)pyridin-2-yl]methyl}-2-imino-5-(2- methylpropyl)-5-phenylimidazolidin-4-one,
methyl 4-[(2-{[4,4-bis(4-fluorophenyl)-2-imino-5-oxoimidazolidin-l-yl]methyl}pyridin-4- yl)carbonyl]piperazine-l-carboxylate,
methyl 4-[(2-{[2-imino-4-(3-methylbutyl)-5-oxo-4-phenylimidazolidin-l-yl]methyl}pyridin-4- yl)carbonyl]piperazine-l-carboxylate,
3-{[2-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)-6-methylpyridin-4-yl]methyl}-5,5-bis(4- fluorophenyl)-2-iminoimidazolidin-4-one,
3-{[4-(5,7-dihydro-6H-pyrrolo[3,4-b]pyrazin-6-ylcarbonyl)thiophen-2-yl]methyl}-2-imino-5,5- diphenylimidazolidin-4-one,
3-{[4-(5,7-dihydro-6H-pyrrolo[3,4-d]pyrimidin-6-ylcarbonyl)thiophen-2-yl]methyl}-2-imino-5,5- diphenylimidazolidin-4-one,
or a pharmaceutically acceptable salt thereof.
18. A pharmaceutical composition comprising an effective amount of a compound according to any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
19. A method for the treatment or prophylaxis of infection by HIV or for the treatment, prophylaxis, or delay in the onset of AIDS in a subject in need thereof, which comprises administering to the subject an effective amount of the compound according to any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof.
20. A compound of any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, for use in the preparation of a medicament for the inhibition of HIV protease, for the treatment or prophylaxis of infection by HIV, or for the treatment, prophylaxis, or delay in the onset of AIDS in a subject in need thereof.
21. A compound of any one of claims 1 to 17, for use in therapy.
22. A pharmaceutical composition comprising an effective amount of a compound of any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier and further comprising an effective amount of an anti-HIV agent selected from the group consisting of HIV antiviral agents, immunomodulators, and anti-infective agents.
23. The pharmaceutical composition of Claim 22, wherein the anti-HIV agent is an antiviral selected from the group consisting of HIV protease inhibitors, HIV reverse transcriptase inhibitors, HIV integrase inhibitors, HIV fusion inhibitors, HIV entry inhibitors, and HIV maturation inhibitors.
PCT/US2017/067163 2016-12-22 2017-12-19 Heterocyclic compounds as hiv protease inhibitors WO2018118829A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP17883610.2A EP3558305A4 (en) 2016-12-22 2017-12-19 Heterocyclic compounds as hiv protease inhibitors
US16/461,899 US20200069691A1 (en) 2016-12-22 2017-12-19 Heterocyclic compounds as hiv protease inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662437919P 2016-12-22 2016-12-22
US62/437,919 2016-12-22

Publications (1)

Publication Number Publication Date
WO2018118829A1 true WO2018118829A1 (en) 2018-06-28

Family

ID=62627320

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/067163 WO2018118829A1 (en) 2016-12-22 2017-12-19 Heterocyclic compounds as hiv protease inhibitors

Country Status (3)

Country Link
US (1) US20200069691A1 (en)
EP (1) EP3558305A4 (en)
WO (1) WO2018118829A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019075291A1 (en) * 2017-10-13 2019-04-18 Gilead Sciences, Inc. 1-benzyl-2-imino-4-phenyl-5-oxoimidazolidine derivatives as hiv protease inhibitors
CN110862395A (en) * 2019-11-13 2020-03-06 株洲千金药业股份有限公司 Preparation method of raw material compound for preparing tadalafil important impurities
WO2020214647A1 (en) * 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of an hiv protease inhibitor
WO2020214716A1 (en) * 2019-04-17 2020-10-22 Gilead Sciences, Inc. 2-imino-5-oxo-imidazolidine inhibitors of hiv protease

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023064331A1 (en) 2021-10-15 2023-04-20 Ssw Advanced Technologies, Llc Illuminated shelf assemblies

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008103351A2 (en) * 2007-02-23 2008-08-28 Schering Corporation Heterocyclic aspartyl protease inhibitors
US20090111832A1 (en) * 2005-11-16 2009-04-30 Barrow James C Imidazolidinone Compounds Useful as Beta-Secretase Inhibitors for the Treatment of Alzheimer's Disease
US8722708B2 (en) * 2005-06-14 2014-05-13 Merck Sharp & Dohme Inc. Substituted isoindolines as aspartyl protease inhibitors
US20140296532A1 (en) * 2013-03-26 2014-10-02 Guangzhou Institutes Of Biomedicine And Health, Chinese Academy Of Sciences Compositions and methods for the treatment of malaria

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7700603B2 (en) * 2003-12-15 2010-04-20 Schering Corporation Heterocyclic aspartyl protease inhibitors
US7592348B2 (en) * 2003-12-15 2009-09-22 Schering Corporation Heterocyclic aspartyl protease inhibitors
US20190046507A1 (en) * 2016-02-18 2019-02-14 Merck Sharp & Dohme Corp. Compounds for the treatment of malaria

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8722708B2 (en) * 2005-06-14 2014-05-13 Merck Sharp & Dohme Inc. Substituted isoindolines as aspartyl protease inhibitors
US20090111832A1 (en) * 2005-11-16 2009-04-30 Barrow James C Imidazolidinone Compounds Useful as Beta-Secretase Inhibitors for the Treatment of Alzheimer's Disease
WO2008103351A2 (en) * 2007-02-23 2008-08-28 Schering Corporation Heterocyclic aspartyl protease inhibitors
US20140296532A1 (en) * 2013-03-26 2014-10-02 Guangzhou Institutes Of Biomedicine And Health, Chinese Academy Of Sciences Compositions and methods for the treatment of malaria

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3558305A4 *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019075291A1 (en) * 2017-10-13 2019-04-18 Gilead Sciences, Inc. 1-benzyl-2-imino-4-phenyl-5-oxoimidazolidine derivatives as hiv protease inhibitors
KR20200071098A (en) * 2017-10-13 2020-06-18 길리애드 사이언시즈, 인코포레이티드 1-benzyl-2-imino-4-phenyl-5-oxoimidazolidine derivatives as HIV protease inhibitors
US10774053B2 (en) 2017-10-13 2020-09-15 Gilead Sciences, Inc. HIV protease inhibitors
US11352329B2 (en) 2017-10-13 2022-06-07 Gilead Sciences, Inc. HIV protease inhibitors
KR102515284B1 (en) 2017-10-13 2023-03-31 길리애드 사이언시즈, 인코포레이티드 1-Benzyl-2-imino-4-phenyl-5-oxoimidazolidine derivatives as HIV protease inhibitors
WO2020214647A1 (en) * 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of an hiv protease inhibitor
WO2020214716A1 (en) * 2019-04-17 2020-10-22 Gilead Sciences, Inc. 2-imino-5-oxo-imidazolidine inhibitors of hiv protease
US11318119B2 (en) 2019-04-17 2022-05-03 Gilead Sciences, Inc. HIV protease inhibitors
CN110862395A (en) * 2019-11-13 2020-03-06 株洲千金药业股份有限公司 Preparation method of raw material compound for preparing tadalafil important impurities
CN110862395B (en) * 2019-11-13 2020-09-29 株洲千金药业股份有限公司 Preparation method of raw material compound for preparing tadalafil important impurities

Also Published As

Publication number Publication date
EP3558305A1 (en) 2019-10-30
US20200069691A1 (en) 2020-03-05
EP3558305A4 (en) 2020-08-19

Similar Documents

Publication Publication Date Title
EP2922840B1 (en) Bicyclic heterocycle substituted pyridyl compounds useful as kinase modulators
EP3558305A1 (en) Heterocyclic compounds as hiv protease inhibitors
CA2871650C (en) Imidazothiadiazole and imidazopyridazine derivatives as protease activated receptor 4 (par4) inhibitors for treating platelet aggregation
CA2515343C (en) Antiparasitic terpene alkaloids
US7968553B2 (en) Tetrahydro-4H-pyrido[1,2-a] pyrimidines useful as HIV integrase inhibitors
JP7097373B2 (en) Aminotriazolopyridine as a kinase inhibitor
CA2859764C (en) Dihydro-benzo-oxazine and dihydro-pyrido-oxazine derivatives
US20040097492A1 (en) Anti-infective agents
TWI822713B (en) Modulators of the beta-3 adrenergic receptor useful for the treatment or prevention of disorders related thereto
WO2013085802A1 (en) Pyrrolopyrimidines as janus kinase inhibitors
KR102345381B1 (en) Carbazole carboxamide compounds useful as kinase inhibitors
CA2657287A1 (en) 1-hydroxy naphthyridine compounds as anti-hiv agents
AU2009257712A1 (en) Pyrrolo [2, 3-c] pyridine derivatives as p38 kinase inhibiting agents
NZ577637A (en) Non-nucleoside reverse transcriptase inhibitors
JP2007528394A (en) HIV integrase inhibitor
JP2011525183A (en) Imidazopyridine and imidazopyrazine compounds useful as kinase inhibitors
AU2015335703A1 (en) Carbazole derivatives
WO2016100166A1 (en) SUBSTITUTED DIHYDRO-1H-PYRROLO[3,2-c]PYRIDIN-4(5H)-ONES AS RIPK3 INHIBITORS
WO2017046737A1 (en) 1-phenylpyrrolidin-2-one derivatives as perk inhibitors
WO2015091584A1 (en) Thiazolopyridine compounds, compositions and their use as tyk2 kinase inhibitors
JP2011525184A (en) Triazolopyridine compounds useful as kinase inhibitors
JP2009513640A (en) HIV integrase inhibitor
WO2016177690A1 (en) Tricyclic piperidine compounds
CN108341819B (en) Phosphodiesterase inhibitors and uses thereof
WO2009154870A1 (en) Hiv integrase inhibitors

Legal Events

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

Ref document number: 17883610

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2017883610

Country of ref document: EP

Effective date: 20190722