WO2008101665A1 - Macrocyclic compounds as hcv ns3 protease inhibitors - Google Patents

Macrocyclic compounds as hcv ns3 protease inhibitors Download PDF

Info

Publication number
WO2008101665A1
WO2008101665A1 PCT/EP2008/001281 EP2008001281W WO2008101665A1 WO 2008101665 A1 WO2008101665 A1 WO 2008101665A1 EP 2008001281 W EP2008001281 W EP 2008001281W WO 2008101665 A1 WO2008101665 A1 WO 2008101665A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
group
substituted
cycloalkyl
compound
Prior art date
Application number
PCT/EP2008/001281
Other languages
French (fr)
Inventor
Shawn D. Britt
Jiping Fu
David Thomas Parker
Michael A. Patane
Prakash Raman
Branko Radetich
Mohindra Seepersaud
Aregahegn Yifru
Rui Zheng
Trixi Brandl
Sylvain Cottens
Claus Ehrhardt
Stefan Andreas Randl
Pascal Rigollier
Nikolaus Schiering
Oliver Simic
Original Assignee
Novartis Ag
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 Novartis Ag filed Critical Novartis Ag
Priority to KR1020097019520A priority Critical patent/KR20090111353A/en
Priority to AU2008217187A priority patent/AU2008217187A1/en
Priority to EP08707770A priority patent/EP2125757A1/en
Priority to MX2009008872A priority patent/MX2009008872A/en
Priority to CN200880012715A priority patent/CN101663284A/en
Priority to CA002677843A priority patent/CA2677843A1/en
Priority to EA200901101A priority patent/EA200901101A1/en
Priority to BRPI0807887-4A2A priority patent/BRPI0807887A2/en
Priority to JP2009550673A priority patent/JP2010519266A/en
Priority to US12/527,787 priority patent/US20100240638A1/en
Publication of WO2008101665A1 publication Critical patent/WO2008101665A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D515/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen, oxygen, and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D515/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen, oxygen, 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
    • C07D515/04Ortho-condensed 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D257/00Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms
    • C07D257/02Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D285/00Heterocyclic compounds containing rings having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by groups C07D275/00 - C07D283/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic 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
    • C07D487/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/02Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link
    • C07K5/0202Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link containing the structure -NH-X-X-C(=0)-, X being an optionally substituted carbon atom or a heteroatom, e.g. beta-amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/02Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link
    • C07K5/0207Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link containing the structure -NH-(X)4-C(=0), e.g. 'isosters', replacing two amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0827Tripeptides containing heteroatoms different from O, S, or N
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/1008Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atoms, i.e. Gly, Ala
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1024Tetrapeptides with the first amino acid being heterocyclic

Definitions

  • HCV chronic hepatitis C virus
  • the current standard therapy for HCV infection is pegylated interferon alpha (IFN- ⁇ ) in combination with ribavirin.
  • IFN- ⁇ pegylated interferon alpha
  • ribavirin can induce significant adverse effects, ranging from flu-like symptoms (fever and fatigue), hematologic complications (leukopenia, thrombocytopenia), neuropsychiatric issues (depression, insomnia, irritability), weight loss, and autoimmune dysfunctions (hypothyroidism, diabetes) from treatment with interferon to significant hemolytic anemia from treatment with ribavirin. Therefore, more effective and better tolerated drugs are still greatly needed.
  • HCV HCV
  • HCV first identified in 1989 (See e.g. Choo, Q. L. et al. Science (1989) 244:359- 362), is a single-stranded RNA virus with a 9.6-kilobase genome of positive polarity. It encodes a single polyprotein that is cleaved upon translation by cellular and viral proteases into at least ten individual proteins: C, El, E2, p7, NS2, NS3, NS4A, NS4B, NS5A, and NS5B (See e.g. Lindenbach, B. D. et al. (2001). Flaviviridae: the viruses and their replication, p. 991-1041. In D. M. Knipe, P. M.
  • NS3 an approximately 70 kDa protein, has two distinct domains: a N-terminal serine protease domain of 180 amino acids (AA) and a C-terminal helicase/NTPase domain (AA 181 to 631).
  • the NS3 protease is considered a member of the chymotrypsin family because of similarities in protein sequence, overall three-dimensional structure and mechanism of catalysis.
  • the HCV NS3 serine protease is responsible for proteolytic cleavage of the polyprotein at the NS3/NS4A, NS4A/NS4B, NS4B/NS5A and NS5A/NS5B junctions (See e.g. Bartenschlager, R., L. et al. (1993) J. Virol. 67:3835-3844; Grakoui, A. et al. (1993) J. Virol. 67:2832-2843; Tomei, L. et al. (1993) J. Virol. 67:4017-4026).
  • NS4A an approximately 6 kDa protein of 54 AA, is a co-factor for the serine protease activity of NS3 (See e.g. Failla, C. et al. (1994) J. Virol. 68:3753-3760; Tanji, Y. et al. (1995) J. Virol. 69:1575-1581).
  • Autocleavage of the NS3/NS4A junction by the NS3/NS4A serine protease occurs intramolecularly (i.e , cis) while the other cleavage sites are processed intermolecularly (i.e., trans). It has been demonstrated that HCV NS3 protease is essential for viral replication and thus represents an attractive target for antiviral chemotherapy.
  • the invention provides compounds of the Formula I:
  • the macrocycle comprises between 15 to 40 ring atoms; m, x and z are each independently selected from 0 or 1 ; p is selected at each occurrence from the group consisting of 0, 1 and 2; Ri and R 2 are independently selected, at each occurrence, from hydrogen or cyano, or from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, alkoxy, and cycloalkyloxy, each of which is unsubstituted or substituted with 1-6 moieties which can be the same or different and are independently selected from the group consisting of hydroxy, oxo, alkyl, aryl, alkoxy, aryloxy, thio, alkylthio, arylthio, amino, alkylamino, arylamin
  • R 3 is selected from the group consisting of H and Ci- 4 -alkyl
  • E is a divalent residue selected from the group consisting OfC(O)NR 23 , NR 23 S(CO p , NR 23 S(CO p NR 23 ;
  • Li and L 2 are divalent residues independently selected from the group consisting of C 0 - 4 alkylene, (CH 2 ),-FG-(CH 2 ) k , (CH 2 ) r C 3-7 cycloalkylene-(CH 2 ) k , (CH 2 ),-C 3- 7 cycloheteroalkylene-(CH 2 ) k , alkenylene, alkynylene, arylene, heteroarylene, cycloalkylene and heterocycloalkylene, each of which is substituted with 0 to 4 independently selected Xi or X 2 groups; i and k are independently selected integers of from 0 to 7; L 3 is a Co- 4 alkylene or a divalent ethylene or acetylene residue, wherein the C 0- 4alkylene and divalent ethylene residues are substituted by 0-2 substituents selected from alkyl, aryl, heteroaryl, mono- or di-alkylamino-Co-C ⁇ al
  • FG is absent or a divalent residue selected from the group consisting of O, S(O) P , NR 23 , C(O), C(O)NR 23 , NR 23 C(O), OC(O)NR 23 , NR 23 C(O)O, NR 23 C(O)NR 23 , S(O) P NR 23 , NR 23 S(O) p , and NR 23 S(O) P NR 23 ;
  • R 23 is independently selected at each occurrence from hydrogen or the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, aryl, heteroaryl, heteroaralkyl, aralkyl and heteroaralkyl, each of which is substituted with 0-2 substituents independently selected from halogen, alkyl, alkoxy, and mono- and di-alkylamino; or
  • R 9 is absent or selected from hydrogen, Q ⁇ alkyl, C 3-7 cycloalkyl-Co- 4 alkyl, or hydroxy;
  • R 7 , Rio, Rn, Ri 2 , Ri 3 , R1 5 , Ri 6 , R17, and R 22 are each, independently, hydrogen or selected from the group consisting of alkyl, alkenyl, alkynyl, aryl, alkyl-aryl, heteroalkyl, heterocyclyl, heteroaryl, aryl-heteroaryl, alkyl-heteroaryl, cycloalkyl, alkyloxy, alkyl- aryloxy, aryloxy, heteroaryloxy, heterocyclyloxy, cycloalkyloxy, amino, alkylamino, arylamino, alkyl-arylamino, arylamino, heteroarylamino, cycloalkylamino, carboxyalkylamino, aralkyloxy and heterocyclylamino; each of which may be further substituted 0 to 5 times with substituents independently selected from Xi and X 2 ;
  • Xi is alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkyl-alkyl, heterocyclyl, heterocyclylalkyl, aryl, alkylaryl, aralkyl, arylheteroaryl, heteroaryl, heterocyclylamino, alkylheteroaryl, or heteroaralkyl; wherein Xi can be independently substituted with one or more of X 2 moieties which can be the same or different and are independently selected; X 2 is hydroxy, oxo, alkyl, aryl, heteroaryl, alkoxy, aryloxy, heteroaryloxy, thio, alkylthio, arylthio, heteroarylthio, amino, alkylamino, arylamino, heteroarylamino, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, alkylsulfonamido
  • n and g are each, independently, 0, 1 or 2;
  • X is O, S, N, C or CR 5a ;
  • R 4 is hydrogen or is selected from the group consisting of Ci -6 -alkyl, C 3-7 -cycloalkyl, aryl, heterocycle and heteroaryl, all of which may be independently substituted one or more times with a halogen atom or Ci- 4 -alkyl;
  • R 5 is absent, hydrogen or oxo or is selected from the group consisting of hydroxyl, Ci-
  • R 5a is selected from the group consisting of H, hydroxyl, Ci -8 -alkyl, C 2-8 -alkenyl, C 2-8 - alkynyl, C 3-8 -cycloalkyl-C 0-4 -alkyl, aryl-C 0-4 -alkyl and heteroaryl-Co ⁇ -alkyl, or R 4 and R 5 may together form a fused dimethyl cyclopropyl ring, a fused cyclopentane ring, a fused phenyl ring or a fused pyridyl ring, each of which may be substituted with a halogen atom, aryl, heteroaryl, trihalomethyl, Ci- 4 -alkoxy or C 1-4 -alkyl; or R 5 and R 53 may together form a spirocyclic ring having between 3 and 7 ring atoms and having 0, 1, or 2 ring heteroatoms, which is optionally substituted by 0-4 substituten
  • R 6 is independently selected at each occurrence from the group consisting of hydrogen, hydroxy, amino, and C 3- or two R 6 residues may together form a spirocyclic ring having between 3 and 7 ring atoms and having O, 1 , or 2 ring heteroatoms, which is optionally substituted by 0-4 substitutents selected from cyano, halogen, hydroxyl, amino, thiol, Ci -8 -alkyl, C 2-8 -alkenyl, C 2-8 -alkynyl, Ci -8 -alkoxy-C 0-4 alkyl, Ci-g-haloalkyl, C 2-8 -haloalkenyl, C 2-8 -haloalkynyl, Ci -8 - haloalkoxy, Ci -8 -alkylthio, Ci -8 -alkylsulfonyl, Q-g-alkylsulfoxy, Ci -8 -alkylthio, Ci -8 -alky
  • the invention provides a method of treating an HCV-associated disorder comprising administering to a subject in need thereof a pharmaceutically acceptable amount of a compound of the invention, such that the HCV-associated disorder is treated.
  • the invention provides a method of treating an HIV infection comprising administering to a subject in need thereof a pharmaceutically acceptable amount of a compound of the invention.
  • the invention provides a method of treating, inhibiting or preventing the activity of HCV in a subject in need thereof, comprising administering to the subject a pharmaceutically acceptable amount of a compound of the invention.
  • the compounds of the invention inhibit the activity of the NS2 protease, the NS3 protease, the NS3 helicase, the NS5a protein, and/or the NS5b polymerase.
  • the interaction between the NS3 protease and NS4A cofactor is disrupted.
  • the compounds of the invention prevent or alter the severing of one or more of the NS4A-NS4B, NS4B-NS5A and NS5A-NS5B junctions of the HCV.
  • the invention provides a method of inhibiting the activity of a serine protease, comprising the step of contacting said serine protease with a compound of the invention, hi another embodiment, the invention provides a method of treating, inhibiting or preventing the activity of HCV in a subject in need thereof, comprising administering to the subject a pharmaceutically acceptable amount of a compound of the invention, wherein the compound interacts with any target in the HCV life cycle.
  • the target of the HCV life cycle is selected from the group consisting of NS2 protease, NS3 protease, NS3 helicase, NS5a protein andNS5b polymerase.
  • the invention provides a method of decreasing the HCV RNA load in a subject in need thereof comprising administering to the subject a pharmaceutically acceptable amount of a compound of the invention.
  • the compounds of the invention exhibit HCV protease activity.
  • the compounds are an HCV NS3-4A protease inhibitor.
  • the invention provides a method of treating an HCV- associated disorder in a subject, comprising administering to a subject in need thereof a pharmaceutically acceptable amount of a compound of the invention, and a pharmaceutically acceptable carrier, such that the HCV-associated disorder is treated.
  • the invention provides a method of treating an HCV- associated disorder in a subject wherein the subject is suffering from or susceptible to a viral infection which is resistant to one or more anti-viral therapies, the method comprising administering to a subject in need thereof a pharmaceutically acceptable amount of a compound of the invention, and a pharmaceutically acceptable carrier, such that the drug- resistant HCV-associated disorder is treated.
  • the invention provides a method of treating an HCV- associated disorder comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of the invention, in combination with a pharmaceutically effective amount of an additional HCV-modulating compound, such as interferon or derivatized interferon, or a cytochrome P450 monooxygenase inhibitor, such that the HCV- associated disorder is treated.
  • an additional HCV-modulating compound such as interferon or derivatized interferon, or a cytochrome P450 monooxygenase inhibitor
  • the additional HCV-modulating compound is selected from the group consisting of ITMN191, Sch 503034 and VX-950.
  • the invention provides a method of inhibiting hepatitis C
  • the invention provides a packaged HCV-associated disorder treatment, comprising an HCV-modulating compound of the invention, packaged with instructions for using an effective amount of the HCV-modulating compound to treat an HCV-associated disorder.
  • the HCV-associated disorder is selected from the group consisting of HCV infection, liver cirrhosis, chronic liver disease, hepatocellular carcinoma, cryoglobulinaemia, non-Hodgkin's lymphoma, and a suppressed innate intracellular immune response.
  • the invention provides a method of treating HCV infection, liver cirrhosis, chronic liver disease, hepatocellular carcinoma, cryoglobulinaemia, non- Hodgkin's lymphoma, and/or a suppressed innate intracellular immune response in subject in need thereof comprising administering to the subject a pharmaceutically acceptable amount of a compound of the invention.
  • the HCV to be treated is selected of any HCV genotype. In another embodiment, the HCV is selected from HCV genotype 1, 2 and/or 3.
  • This invention is directed to compounds, e.g., peptide compounds, and intermediates thereto, as well as pharmaceutical compositions containing the compounds for use in treatment of HCV infection.
  • This invention is also directed to the compounds of the invention or compositions thereof as protease inhibitors, particularly as serine protease inhibitors, and more particularly as HCV NS3 protease inhibitors.
  • the compounds are particularly useful in interfering with the life cycle of the hepatitis C virus and in treating or preventing an HCV infection or physiological conditions associated therewith.
  • the present invention is also directed to methods of combination therapy for inhibiting HCV replication in cells, or for treating or preventing an HCV infection in patients using the compounds of the invention or pharmaceutical compositions, or kits thereof.
  • the compounds of the invention are compounds of Formula I, in which
  • Ri and R 2 taken in combination form a 3, 4, 5, or 6-membered saturated carbocyclic ring which is substituted with 0-2 substituents independently selected from halogen, alkyl, alkenyl, alkoxy and C 3-6 cycloalkyl.
  • compounds of the invention are compounds of Formula I, in which R
  • compounds of Formula I include those compounds in which Ri and R 2 are taken in combination to form a cyclopropyl ring substituted with 0-2 substituents independently , selected from halogen, alkyl, alkenyl, and alkoxy or substituted with 0 to 2 Ci-C 4 alkyl residues.
  • Still other compounds of Formula I include those in which R t and R 2 are taken in combination to form a cyclopropyl ring which is substituted with 0 or 1 substituents selected Ci ⁇ alkyl, vinyl or cyclopropyl; and E is C(O)NH, NHS(O) 2 , NHSO 2 N(Me), NHSO 2 N(Et) or NHSO 2 N(cyclopropyl).
  • the compounds of the invention are compounds of any one of Formulae I, in which Ri is H or Ci -4 alkyl; and R 2 is H, Ci-C 4 alkyl, Ci-C 4 fluoroalkyl, C 2 - C 4 alkenyl, or C 3 -C 7 cycloalkylCo -2 alkyl.
  • Certain other compounds of Formula I comprise a macrocycle having between 15 and
  • Certain compounds of Formula I comprise a macrocycle having 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 ring atoms. In certain instances, compounds of Formula I comprise a macrocycle having 16, 17, 18, 19, 20, 21, 22, or 23 ring atoms. Certain other compounds of Formula I comprise a macrocycle selected from the group consisting of macrocycles of the formulae:
  • L 1 is Ci-C 6 alkylene, C 3 -C 7 cycloalkylene, arylene or heteroarylene, each of which is substituted by 0-4 residues independently selected from Ci-C 4 alkyl, Ci-C4alkoxy, hydroxyl, amino, mono- and di- Ci-C 4 alkylamino, halogen, cyano, Ci-C 4 fluoroalkyl, Ci-C 4 fluoroalkoxy, COOH, carboxamide (CONH 2 ), mono- and di-Cr C4alkylcarboxamide, aryl, heteroaryl and 5 or 6 membered saturated heterocycles;
  • L 2 is selected from Ci-C ⁇ alkylene and C 2 -C 6 alkenylene, each of which is substituted by 0-4 residues independently selected from Ci-C 4 alkyl, Ci-C 4 alkoxy, hydroxyl, amino, mono- and di- Ci-C 4 alkylamino, halogen, cyano, Ci-C 4 fluoroalkyl, Q ⁇ fluoroalkoxy,
  • L 3 is absent or a divalent ethylene residue which is substituted by O to 2 independently selected methyl or ethyl residues.
  • Li is a divalent residue selected from C 2 -
  • C4alkylene 1,2-phenylene, 1,3-phenylene, 2,4-pyridylene, 2,3-pyridylene, 3,4-pyridylene or 1,7-indolylene, 2,7-indolylene, each of which is substituted with 0-3 residues selected from Cj-C4alkyl, Ci-C 4 alkoxy, hydroxyl, amino, mono- and di- Ci-C 4 alkylamino, halogen, cyano, Ci-C 2 fluoroalkyl, Ci-C 2 fluoroalkoxy, COOH, carboxamide (CONH 2 ), and mono- and di-Ci- C4alkylcarboxamide.
  • Li is C 3 -C 7 cycloalkylene, arylene or heteroarylene which is substituted by 0-4 residues independently selected from Ci-C 4 alkyl, Ci-C4alkoxy, hydroxyl, amino, mono- and di- Ci-C 4 alkylamino, halogen, cyano, Ci- C 4 fluoroalkyl, Ci-Qfluoroalkoxy, COOH, carboxamide (CONH 2 ), mono- and di-Ci- C 4 alkylcarboxamide, aryl, heteroaryl and 5 or 6 membered saturated heterocycles;
  • L 2 is selected from Ci-C ⁇ alkylene and C 2 -C 6 alkenylene, each of which is substituted by 0-4 residues independently selected from Ci-C 4 alkyl, C]-C 4 alkoxy, hydroxyl, amino, mono- and di- Ci-C 4 alkylamino, halogen, cyano, Ci-C 4 fluoroalkyl, C 1 -C 4 HuOrOaIkOXy,
  • L 3 is absent or a divalent ethylene residue which is substituted by O to 2 independently selected methyl or ethyl residues.
  • Li is a divalent residue selected from 1,2- phenylene, 1,3-phenylene, 2,4-pyridylene, 2,3-pyridylene, 3,4-pyridylene or 1,7-indolylene, 2,7-indolylene, each of which is substituted with 0-3 residues selected from Ci-C 4 alkyl, C 1 - C 4 alkoxy, hydroxyl, amino, mono- and di- Ci-C 4 alkylamino, halogen, cyano, Ci- C 2 fluoroalkyl, Ci-C 2 fluoroalkoxy, COOH, carboxamide (CONH 2 ), and mono- and di-Ci- C 4 alkylcarboxamide.
  • Certain compounds of Formula I include compounds of Formula II:
  • R 1 is selected from the group consisting of H and Ci- 4 -alkyl
  • R 2 is selected from the group consisting of Ci ⁇ -alkyl, C(O)C ]-4 -alkyl, C(O)OCi -4 - alkyl, and (CH 2 ) 0-4 -C 3-6 -cycloalkyl; or R 1 and R 2 together form a cyclopropane ring;
  • R 3 is selected from the group consisting of H and Ci ⁇ -alkyl
  • X is O, NR 5 or CR 5 R 5a ;
  • R 4 is hydrogen or is selected from the group consisting of C ⁇ -alkyl, C 3-6 -cycloalkyl, aryl, heterocycle and heteroaryl, each of which may be independently substituted one or more times with a halogen atom or
  • R 5 is hydrogen or oxo or is selected from the group consisting of hydroxyl, Ci- 8 -alkyl, C 2 - 8 -alkenyl, C 2-8 -alkynyl, C 3-8 -cycloalkyl-Co- 4 -alkyl, aryl-Co- 4 -alkyl, aryloxy, heteroaryloxy, heterocycle-Co -4 -alkyl and heteroaryl-Co ⁇ -aUcyl, each of which may be independently substituted one or more times with a halogen atom, aryl, heteroaryl, trihalomethyl, alkoxy or C 1 ⁇ -alkyl;
  • R 52 is selected from the group consisting of H, hydroxyl, Ci-g-alkyl, C 2- g-alkenyl, C 2-8 - alkynyl, C 3-8 -cycloalkyl-Co- 4 -alkyl, aryl-C 0-4 -alkyl and heteroaryl-Co- 4 -alkyl, or R 4 and R 5 may together form a fused dimethyl cyclopropyl ring, a fused cyclopentane ring, a fused phenyl ring or a fused pyridyl ring, each of which may be substituted with a halogen atom, aryl, heteroaryl, trihalomethyl, Ci- 4 -alkoxy or Ci ⁇ -alkyl; or R 5 and Rs 2 may together form a spirocarbocyclic saturated ring having between 3 and 6 carbon ring atoms which is optionally substituted by 0-2 substitutents selected from halogen, Ci
  • Rio and Ru are each, independently, selected from the group consisting of H and Ci -4 - alkyl;
  • R 6 and R] 3 is H;
  • Ri 2 is selected from the group consisting of H, Ci-4-alkyl and C 3-6 -cycloalkyl;
  • Still other compounds of the invention according to Formula II include those compounds in which X is CR 5 Rs 3 , R 4 is H, and R 5 and Rs a taken in combination form a 3 to 6 member spirocyclic carbocycle substituted with 0-2 substitutents selected from halogen, Ci -6 - alkyl, C 2-6 -alkenyl, C 2-6 -alkynyl, Ci -6 -alkoxide, C 3-7 -C ycloalkyl-C 0-4 -alkyl, phenyl-C 0-4 -alkyl, naphthyl-Co-4-alkyl, heteroaryl-Co-4-alkyl, or two substitutents taken together form a fused or spirocyclic 3 to 7 membered carbocyclic ring, each of which is substituted with 0-3 independently selected halogen atoms or Ci ⁇ -alkyl groups.
  • Z 2 is nitrogen or CH; ki and k 2 are 0 or 1 such that a sum of ki and k 2 equals 1 or 2;
  • R a is hydrogen, C ⁇ alkyl, or phenyl;
  • Rb is hydrogen, C 1 . 4 a.kyl, mono- and or phenyl or R a and R b taken together form a fused or spirocyclic 3 to 6 membered ring having 0, 1 or 2 ring heteroatoms selected from N, O and S, which fused or spirocyclic ring has 0 to 2 independently selected substitutents selected from halogen, Ci ⁇ alkyl, Ci- 4 alkoxy, Q- 4 alkanoyl, and phenyl; and
  • Rc represents 0 to 4 substitents which are independently selected at each occurrence of R 0 from the group consisting of halogen, C 1-4 alkyl, and phenyl, or two geminal R 0 substitents, taken in combination form a 3 to 6 member spirocyclic ring.
  • Certain compounds of the invention according to Formula Ha include those compounds in which the divalent residue:
  • Still other compounds of the invention according to Formula II include those compounds in which: X is CRsR 53 ; and
  • R 5 and R 53 taken in combination, form a spirocyclic ring having between 3 and 7 ring atoms and having 0, 1, or 2 ring heteroatoms, which spirocyclic ring is substituted with a spirocyclic 3 to 7 membered ring having 0, 1 or 2 ring heteroatoms selected from N, O and S, and wherein each of the spirocyclic rings has 0 to 2 independently selected substitutents selected from cyano, halogen, hydroxyl, amino, thiol, Ci -8 -alkyl, C 2-8 -alkenyl, C 2-8 -alkynyl, Ci. 8 -alkoxy-Co -4 alkyl, C
  • Certain other compounds according to Formula I or Formula II include those compounds in which X is CR 5 R 53 wherein Rs 3 is hydrogen, methyl or trifluoromethyl; and R 5 is a residue of the formula:
  • Z 3 is NR 23 or O
  • Z 4 , Zs 1 Z 6 , and Z 7 are each independently selected from the group consisting of N, CH, and CR 8 ;
  • R 8 and R 8a each indepently represent O to 2 groups, each of which is independently selected at each occurrence of R 8 and R 8a from the group consisting of hydrogen, halogen, Cj- 4-alkyl, 4alkyl, mono- and heterocycleC 0- 4alkylamino and heterocycleCo ⁇ alkyl; or two R 8a , taken in combination, form a fused- or spiro-cyclic 3-7 membered ring.
  • Still other compounds of Formula I or Formula II include those compounds in which X is CR 5a , R 5a is hydrogen or methyl, and R 5 is a residue selected from the group consisting of:
  • R 8 is selected from hydrogen, methyl, ethyl, mono-, di-, or tri-fluoromethyl, mono-, di-, or tri-fluoromethoxy, fluoro, and chloro.
  • R 8 is selected from hydrogen, methyl, ethyl, mono-, di-, or tri-fluoromethyl, mono-, di-, or tri-fluoromethoxy, fluoro, and chloro.
  • R 6 is hydrogen, methyl, ethyl, and mono-, di-, and tri-fluoromethyl
  • R 8 is selected from R 8 is selected from hydrogen, methyl, ethyl, mono-, di-, or tri- fluoromethyl, mono-, di-, or tri-fluoromethoxy, fluoro, and chloro.
  • Still other compounds of Formula I or Formula II include those compounds in which X is CR 5a , R 5a is hydrogen or methyl, and R 5 is a residue selected from the group consisting of:
  • Still other compounds of the invention according to Formula II include compounds according to Formula lib:
  • Z 2 is nitrogen or CH; ki and k 2 are 0 or 1 such that a sum of k
  • R c represents 0 to 2 substituents which are independently selected at each occurrence OfR 0 from the group consisting of halogen, Ci ⁇ alkyl, and phenyl, or two geminal R 0 substitents, taken in combination form a 3 to 6 member spirocyclic ring;
  • R 4 represents 0, 1, or 2 substituents each of which is independently selected from H and C 1 - 4 - alkyl;
  • R 6 is hydrogen or C h alky!.
  • Certain compounds of Formula II include those compounds, in which the
  • ring is a divalent residue derived from a proline residue selected from the group consisting of:
  • Certain other compounds of Formula II, Formula Ha or Formula lib include compounds in which X is CR. 5 R. 5 a, R 4 is H, and R 5 and R 53 taken in combination form a 3 to 6 member spirocyclic carbocycle substituted with 0-2 substitutents selected from halogen, C 1-6 - alkyl, C 2-6 -alkenyl, C 2-6 -alkynyl, Ci -6 -alkoxide, C 3-7 -cycloalkyl-C 0-4 -alkyl, phenyl-Co- 4 -alkyl, naphthyl-Co-4-alkyl, or two substitutents taken together form a fused or spirocyclic 3 to 7 membered carbocyclic ring, each of which is substituted with 0-3 independently selected halogen atoms or Q ⁇ -alkyl groups.
  • Certain compounds of Formulae I include compounds of Formula III:
  • Certain compounds of the invention according to Formula III include compounds in which:
  • Zi is absent or NR 10 ;
  • Z 2 is nitrogen or CH;
  • R 3 is selected from the group consisting of H, Ci-4-alkyl, and C 3-6 -cycloalkylCo- C 4 alkyl;
  • Rn, Ri 5 and R 22 are selected from the group consisting of H, alkyl-aryl, Ci ⁇ -alkyl, O- Ci-4-alkyl, N(H)-C M -alkyl, and C 3-6 -cycloalkylC 0 -C 4 alkyl;
  • Rio and Rn are each, independently, selected from the group consisting of H, Ci -4 - alkyl and (CH 2 ) 0-4 -C 3-6 -cycloalkyl; or
  • Ri 5 and Ri 6 may together form a 3, 4, 5, 6 or 7-membered ring that may comprise between 0 to 3 additional heteroatoms, wherein the ring may be further substituted with 0-5 substitutents; or
  • Ri 6 and Ri 7 may together form a 3, 4, 5, 6 or 7-membered ring that may comprise between 0 to 3 additional heteroatoms, wherein the ring may be further substituted with 0-5 substitutents;
  • R 3 is selected from the group consisting of H and Ci- 4 -alkyl
  • R 8 , Rio and Rn are each, independently, selected from the group consisting of H, Ci -4 - alkyl, and C 3-7 cycloalkylC 0-4 alkyl;
  • Ri 2 is selected from the group consisting of H, Ci -4 -alkyl and (CH 2 ) 0-4 -C 3-6 -cycloalkyl;
  • Certain compounds of Formula III include compounds represented by Formula Ilia:
  • R 25 and R 26 are each, independently, selected from the group consisting of H, C 1-4 - alkyl, O-Q ⁇ -alkyl, N(R 24 ) 2 , C 3-6 cycloalkylCo-C 4 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted heterocycle, wherein each R 24 is independently selected from the group consisting of H, halogen, hydroxy, COOH, amino, carboxamide, substituted or unsubstitiited-d- 4 -alkyl, substituted or unsubstituted C 3-6 cycloalkylCo-C 4 alkyl, substituted or unsubstituted-Ci- 4 -alkoxy, substituted or unsubstituted C ⁇ cycloalkylCo ⁇ alkyl-oxy-, substituted or unsubstituted arylC 0 -C 4 alkyl, substituted or unsubstituted heterocycleCo- C 4 alkyl, substituted
  • R 25 is H and R 26 is amine, substituted or unsubstiuted phenyl, or substituted or unsubstiuted benzyl.
  • Certain other compounds of Formula III include compounds represented by Formula HIb:
  • R 27 and R 28 are each, independently, selected from the group consisting of H, C 1-4 - alkyl, O-C 1-4 -alkyl, N(R 24 ) 2 , C 3 - 6 cycloalkylCo-C 4 alkyl, substituted or unsubstituted aryl, substituted or unsubstituted O-aryl and substituted or unsubstituted heterocycle, wherein R 24 is independently selected at each occurrence from the group consisting of H, halogen, hydroxy, COOH, amino, carboxamide, substituted or unsubstituted-Ci -4 -alkyl, substituted or unsubstituted C 3-6 cycloalkylCo-C 4 alkyl, substituted or unsubstituted-C 1-4 -alkoxy, substituted or unsubstituted C 3-6 cycloalkylCo-C 4 alkyl-oxy-, substituted or unsubstituted arylCo-C 4 alkyl
  • R 28 is quinoline, or O- quinoline, wherein the quinoline and O-quinoline substituents may be independently substituted one or more times (or preferably between one and five times) with halogen, amino, O-Ci-4-alkyl, substituted or substituted or unsubstituted- (CH 2 )o- 4 -C 3-6 -cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted O-aryl, and substituted or unsubstituted heterocycle.
  • Z 2 is nitrogen or CH
  • R 29 and R 30 are selected from the group consisting of H, Ci-4-alkyl, O-C ⁇ -alkyl, N(R 24 ) 2 , C 3-6 cycloalkylCo-C 4 alkyl, substituted or unsubstituted aryl, substituted or unsubstituted aryl-oxy and substituted or unsubstituted heterocycle, wherein each R 24 is independently selected at each occurrence from the group consisting of H, halogen, hydroxy, COOH, amino, carboxamide, substituted or unsubstituted-Ci- 4 -alkyl, substituted or unsubstituted C 3-6 cycloalkylCo-C 4 alkyl, substituted or unsubstituted-Ci- 4 -alkoxy, substituted or unsubstituted C 3 - 6 cycloalkylCo-C 4 alkyl-oxy-, substituted or unsubstituted arylCo-C 4 alkyl, substituted or un
  • R 2 g is selected from the group consisting of O- phenyl and O-benzyl.
  • Still other compounds of Formula III include compounds represented by Formula IHd:
  • Z 2 is nitrogen or CH
  • R 31 represents one or two residues which are independently selected at each occurrence from the group consisting of H, Ci-4-alkyl, O-Ci-4-alkyl, N(R 24 ) 2 , (CH 2 ) 0-4 -C 3-6 - cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted O-aryl and substituted or unsubstituted heterocycle, wherein each R 24 is independently selected from the group consisting of H, halogen, hydroxy, COOH, amino, carboxamide, substituted or unsubstituted-Ci ⁇ -alkyl, substituted or unsubstituted C 3-6 cycloalkylCo-C4alkyl, substituted or substituted or unsubstituted C 3-6 cycloalkylCo-C 4 alkyl-oxy-, substituted or unsubstituted arylCo-C 4 alkyl, substituted or unsubstituted heterocycleC 0
  • Z 2 is nitrogen or CH;
  • Formula IHd is represented by a compound of the Formula HIf
  • Formula IHd is represented by a compound of the Formula IHg:
  • Certain compounds of Formula III include compounds represented by Formula Illh:
  • R 35 is H, halogen, hydroxy, COOH, amino, carboxamide, substituted or unsubstituted- Ci- 4 -alkyl, substituted or unsubstituted C 3-6 cycloalkylCo-C 4 alkyl, substituted or unsubstituted- Ci-4-alkoxy, substituted or unsubstituted C 3 ⁇ cyclpalkylCo-C 4 alkyl-oxy-, substituted or unsubstituted arylCo-C 4 alkyl, substituted or unsubstituted heterocycleCo-C 4 alkyl, substituted or unsubstituted arylCo-C 4 alkyl-oxy and substituted or unsubstituted heterocycleCo-C 4 alkyl- oxy.
  • R 35 is phenyl, optionally substituted with chloro.
  • Certain compounds of Formula I include compounds of Formula IV:
  • Certain compounds of Formula FV include those compounds in which: y is 0 or 1 ;
  • Z 2 is nitrogen or CH; Ri is selected from the group consisting of H and Ci- 4 -alkyl;
  • Ri 7 is hydrogen or is selected from the group consisting of Ci ⁇ -alkyl, Ci -6 -cycloalkyl, (CH 2 )o- 4 -C 3-6 -cycloalkyl, aryl, alkyl-aryl and heterocycle, each of which may be independently substituted one or more times (or preferably between one and five times); Rio and Rn are each, independently, selected from the group consisting of H and Ci -4 - alkyl;
  • Ri 2 is selected from the group consisting of H, Ci- 4 -alkyl, Ci- ⁇ -cycloalkyl and aryl;
  • Certain other compounds of Formula IV include those compounds in which R] 7 is selected from the group consisting of H, cyclopropylCo-C 2 alkyl, cyclopentylCo-C 2 alkyl, phenylCi-C 2 alkyl, and naphthylCi-C 2 alkyl.
  • Certain other compounds of Formulae I, II (including Ha and lib), III (including Ilia through IIDi), and/or IV include those compounds in which V is selected from the group consisting of C(O)R 24 , C(O)C(O)OR 24 , C(O)N(H)R 24 , C(O)C(O)N(H)R 24 and C(O)OR 24 , wherein each R 24 is independently selected from the group consisting of H, halogen, substituted or unsubstituted-Ci- 4 -alkyl, substituted or unsubstituted C 3 - 6 -cycloalkylCo-C 4 alkyl, substituted or unsubstituted arylCo-C 4 alkyl and substituted or unsubstituted heterocycleCo- C 4 alkyl, and any combination thereof.
  • Still other compounds of Formulae I, II (including Ha and lib), III (including Ilia through IIDi), and/or IV include compounds in which V is C(O)-R 20 , wherein R 20 is selected from the group consisting of tert-butyl, C 3 - 6 -cycloalkyl, phenyl, pyrazine, benzooxazole, 4,4- dimethyl-4,5-dihydro-oxazole, benzoimidazole, pyrimidine, thiazole, benzothiazole, benzothiazole 1,1 -dioxide and quinazoline, each of which may be further independently substituted with 0-5 substitutents selected from a halogen atom, Ci-4alkoxy, C 2 - Qalkenyloxy, C 2 -C 4 alkynyloxy, substituted by one or more halogen atoms, or C 3-6 - cycloalkyl.
  • R 20 is selected from the group consisting of ter
  • Still other compounds of Formulae I, II (including Ha and lib), III (including Ilia through HIh), and/or IV include compounds in which V is R 20 or C(O)-R 2O , wherein R 20 is a residue of the formula:
  • Z 8 is absent or selected from NR 33 or oxygen; g and fare independently selected integers selected from the group consisting of 0, 1,
  • j is an integer selected from the group consisting of 1, 2, 3 and 4, wherein the sum of f + g + j is less than or equal to 5 and greater than or equal to 2 when Z 8 is absent and the sum of f + g + jk is less than or equal to 4 and greater than or equal to 1 when Z 8 is oxygen;
  • R 33 is independently selected at each occurrence from the group consisting of hydrogen, and
  • R 34 represents zero to three residues each independently selected at each occurrence from the group consisting of halogen, hydroxy, amino, C 3-6 cycloalkyl,
  • Still other compounds of Formulae I, II (including IIa and lib), III (including FIIa through HIh), and/or FV include compounds in which V is C(O)-R 2O , wherein R 20 is a residue of the formula:
  • g is an integer selected from the group consisting of 0, 1, 2, 3 and 4; j is an integer selected from the group consisting of 1, 2, 3 and 4, wherein the sum of g + j is less than or equal to 5 and greater than or equal to 2; R 33 is independently selected at each occurrence from the group consisting of hydrogen, Ci ⁇ alkyl, haloCi ⁇ alkyl, C 3-6 cycloalkyl, hydroxyCi ⁇ alkyl, and Ci ⁇ alkoxyCi ⁇ alkyl; and
  • R 34 represents zero to three residues each independently selected at each occurrence from the group consisting of halogen, hydroxy, amino, mono-and and C 1-4 alkoxyC 1-4 alkyl.
  • X is CR 5 Rs a , R 4 and Rs 3 are H and R 5 is aryl-C 0- 3 -alkyl, -O-heterocycle, or heterocycle-Co- 3 -alkyl, wherein aryl and heterocycle may be independently substituted one or more times (or preferably between one and five times) with a halogen atom, aryl, trihalomethyl, C 3 - 6 -cycloalkyl or C 1-4 -alkyl.
  • X is CR 5 Rs 3 , R 4 and R 53 are H and R 5 is selected from the group consisting of piperidine, phenyl, -O-pyridinyl and CH 2 -pyridinyl, wherein the phenyl and pyridinyl groups may be independently substituted one or more times (or preferably between one and five times) with a halogen atom or Ci ⁇ -alkyl.
  • R 5 is 5-chloro-pyridin-2-yl.
  • R 5 is selected from the group consisting of
  • R 2 is independently selected from the group consisting of C 1-4 -alkyl and aryl.
  • CR 5 R 5a taken in combination, form a spirocyclic 3 to 6 member carbocyclic ring.
  • Certain spirocyclic rings include groups of the formula:
  • Rs b and R 5c are independently selected from hydrogen halogen, Ci- 6 -alkyl, C 2-6 -alkenyl, C 2-6 - alkynyl, Ci ⁇ -alkoxide, C 3-7 -cycloalkyl-Co- 4 -alkyl, phenyl-Co ⁇ t -alkyl, naphthyl-Co- 4 -alkyl, heteroaryl 7 Co- 4 -alkyl, or two substitutents taken together form a fused or spirocyclic 3 to 7 membered carbopyclic ring, each of which is substituted with 0-3 independently selected halogen atoms or C 1-4 -alkyl groups.
  • R 2 is selected from the group consisting of propyl and (CH 2 ) 2 -cyclobutyl.
  • Rn is H and R 12 is C 3-6 -cycloalkyl.
  • R 12 is cyclohexyl
  • V is selected from the group consisting of C(O)- N(H)-/-butyl.
  • Yet other compounds of any one of Formulae I, II (including Ha and lib), III (including Ilia through HIh), and/or IV include compounds in which V is C(O)-N(H)-/-butyl or C(O)-R 20 , wherein R 20 is selected from the group consisting of C 3-6 -cycloalkyl, phenyl, pyrazine, benzooxazole, 4,4-dimethyl-4,5-dihydro-oxazole, benzoimidazole, pyrimidine, thiazole, benzothiazole, benzothiazole 1,1 -dioxide and quinazoline, all of which may be further independently substituted with a halogen atom, CF 3 , C ⁇ - 4 -alkyl, Q ⁇ alkoxy, C 2 - C 4 alkenyloxy, C 2 -C 4 alkynyloxy, or C 3-6 -cycloalkyl.
  • V is selected from the group consisting Of C 3-6 - cycloalkyl, phenyl, pyrazine, benzooxazole, 4,4-dimethyl-4,5-dihydro-oxazole, benzoimidazole, pyrimidine, thiazole, benzothiazole, benzothiazole 1,1 -dioxide and quinazoline, all of which may be further independently substituted with a halogen atom, CF 3 , Ci- 4 -alkyl , Ci -4 alkoxy, C 2 -C 4 alkenyloxy, C 2 -C 4 alkynyloxy, or C 3-6 -cycloalkyl.
  • V is R 20 or C(O)-R 20 , wherein R 20 is selected from the group consisting of C 3-6 -cycloalkyl, phenyl, pyrazine, benzooxazole, 4,4-dimethyl-4,5-dihydro- oxazole, benzoimidazole, pyrimidine, benzothiazole 1,1 -dioxide and quinazoline, all of which may be further independently substituted with a halogen atom, CF 3 , Ci-4-alkyl or C 3-6 - cycloalkyl.
  • V is R 20 or C(O)-R 20 , wherein R 2 o is selected from the group consisting of
  • Ri 8 is selected from the group consisting of hydrogen, a halogen atom, aryl, Ci -4 - alkyl, Ci ⁇ alkoxy, C 2 -C 4 alkenyloxy, C 2 -C 4 alkynyloxy, C ⁇ -alkyl substituted by one or more halogen atoms, or C 3-6 -cycloalkyl.
  • V is R 20 or C(O)-R 20 , wherein R 20 is selected from the group consisting of
  • R] 8 is selected from the group consisting of hydrogen, a halogen atom, aryl, Ci -4 - alkyl, Ci ⁇ alkoxy, C 2 -C4alkenyloxy, C 2 -C 4 alkynyloxy, d ⁇ -alkyl substituted by one or more halogen atoms, or C 3-6 -cycloalkyl.
  • V is selected from the group consisting of C 3-6 -cycloalkyl, phenyl, pyrazine, benzooxazole, 4,4-dimethyl-4,5-dihydro-oxazole, benzoimidazole, pyrimidine, thiazole, benzothiazole, benzothiazole 1,1 -dioxide and quinazoline, all of which may be further independently substituted with a halogen atom, Ci- 4 -alkyl, C 1-4 alkoxy, C 2 - C 4 alkenyloxy, C 2 -C 4 alkynyloxy, Ci- 4 -alkyl substituted by one or more halogen atoms, or C 3-6 - cycloalkyl.
  • variable V is selected from the group consisting of R 20 and C(O)- R 20 , wherein R 20 is selected from the group consisting of C 3-6 -cycloalkyl, mono- and di-Ci.
  • variable V is selected from the group consisting of R 20 and C(O)- R 20 , wherein R 20 is selected from the group consisting of
  • R 18 is selected from the group consisting of hydrogen, a halogen atom, aryl, trihalomethyl, and Ci- 4 -alkyl.
  • any of the C 3-6 -cycloalkyl groups of Formula I, or any subformula thereof may be independently substituted one or more times (or preferably between one and five times) with a halogen atom, aryl, heteroaryl, trihalomethyl, C ⁇ -alkoxy or C 1-4 -alkyl.
  • any of the heterocycle groups are independently selected from the group consisting of acridinyl, carbazolyl, cinnolinyl, quinoxalinyl, pyrrazolyl, indolyl, benzotriazolyl, furanyl, thienyl, benzothienyl, benzo furanyl, quinolinyl, isoquinolinyl, oxazolyl, isoxazolyl, indolyl, pyrazinyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolyl, tetrahydroquinoline, benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, imidazoly
  • Certain other compounds of Formula I, and sub formulae thereof include those compounds which contain a fragment selected from the residues of each of Tables C, D, E, F, hand G.
  • compounds of the invention include all P1-P2 compounds formed by combining all possible permutations of the fragments of Tables C, D, E, F and G wherein the bond ending in an asterisk is the point of attachment Pl and P2 fragments are coupled by condensation of the amino residue on the Pl fragment with the carboxylic acid residue on the P2 fragment.
  • the compound C(l)-D(3)-E(10)-F(4)-G(15) is the compound in which the residue of entry 1 of Table C, the residue of entry 3 of Table D, the residue of entry 10 of Table E, the residue of entry 4 of Table F (where n is 1) and the residue of entry 15 of Table G are combined to form a compound of formula I which has the structure:
  • variable, E, of Formula 1 is a residue selected from the group consisting of: Dl ⁇ -C(O)NH-# I D2 @-NHSO 2 -#
  • the fragment of Formula 1 has a residue of the formula
  • the fragment of Formula 1 has a residue of the formula
  • certain compounds of the invention are found to show IC 50 values for HCV inhibition in the range from 10 to more than 100 ⁇ M, or 0.5 to 30 ⁇ M, or show IC 5O values for HCV inhibition of less than 10 ⁇ M.
  • a compound of the present invention is further characterized as a modulator of HCV, including a mammalian HCV, and especially including a human HCV.
  • the compound of the invention is an HCV inhibitor.
  • HCV-associated state or "HCV-associated disorder” include disorders and states (e.g., a disease state) that are associated with the activity of HCV, e.g., infection of HCV in a subject.
  • HCV-associated states include HCV-infection, liver cirrhosis, chronic liver disease, hepatocellular carcinoma, cryoglobulinemia, non-Hodgkin's lymphoma, and a suppressed innate intracellular immune response.
  • HCV-associated states are often associated with the NS3 serine protease of HCV, which is responsible for several steps in the processing of the HCV polyprotein into smaller functional proteins.
  • NS3 protease forms a heterodimeric complex with the NS4A protein, an essential cofactor that enhances enzymatic activity, and is believed to help anchor HCV to the endoplasmic reticulum.
  • NS3 first autocatalyzes hydrolysis of the NS3-NS4A juncture, and then cleaves the HCV polyprotein intermolecularly at the NS4A-NS4B, NS4B-NS5 A and NS5A-NS5B intersections. This process is associated with replication of HCV in a subject.
  • Inhibiting or modulating the activity of one or more of the NS3, NS4A, NS4B, NS5A and NS5B proteins will inhibit or modulate replication of HCV in a subject, thereby preventing or treating the HCV-associated state.
  • the HCV-associated state is associated with the activity of the NS3 protease.
  • the HCV- associated state is associated with the activity of NS3-NS4A heterodimeric complex.
  • the compounds of the invention are NS3/NS4A protease inhibitors.
  • the compounds of the invention are NS2/NS3 protease inhibitors. Without being bound by theory, it is believed that the disruption of the above protein- protein interactions by the compounds of the invention will interfere with viral polyprotein processing by the NS3 protease and thus viral replication.
  • HCV-associated disorders also include HCV-dependent diseases.
  • HVC-dependent diseases include, e.g., any disease or disorder that depend on or related to activity or misregulation of at least one strain of HCV.
  • the present invention includes treatment of HCV-associated disorders as described above, but the invention is not intended to be limited to the manner by which the compound performs its intended function of treatment of a disease.
  • the present invention includes treatment of diseases described herein in any manner that allows treatment to occur, e.g., HCV infection.
  • the compounds of the invention can be useful for treating diseases related to HFV, as well as HIV infection and AIDS (Acquired Immune Deficiency Syndrome).
  • the invention provides a pharmaceutical composition of any of the compounds of the present invention.
  • the invention provides a pharmaceutical composition of any of the compounds of the present invention and a pharmaceutically acceptable carrier or excipient of any of these compounds.
  • the invention includes the compounds as novel chemical entities.
  • the invention includes a packaged HCV-associated disorder treatment. The packaged treatment includes a compound of the invention packaged with instructions for using an effective amount of the compound of the invention for an intended use.
  • the compounds of the present invention are suitable as active agents in pharmaceutical compositions that are efficacious particularly for treating HCV-associated disorders.
  • the pharmaceutical composition in various embodiments has a pharmaceutically effective amount of the present active agent along with other pharmaceutically acceptable excipients, carriers, fillers, diluents and the like.
  • pharmaceutically effective amount indicates an amount necessary to administer to a host, or to a cell, issue, or organ of a host, to achieve a therapeutic result, especially an anti-HCV effect, e.g., inhibition of proliferation of the HCV virus, or of any other HCV-associated disease.
  • the diseases to be treated by compounds of the invention include, for example, HCV infection, liver cirrhosis, chronic liver disease, hepatocellular carcinoma, cryoglobulinemia, non-Hodgkin's lymphoma, and a suppressed innate intracellular immune response.
  • the present invention provides a method for inhibiting the activity of HCV.
  • the method includes contacting a cell with any of the compounds of the present invention.
  • the method further provides that the compound is present in an amount effective to selectively inhibit the activity of one or more of the NS3, NS4A, NS4B, NS5A and NS5B proteins.
  • the method provides that the compound is present in an amount effective to diminish the HCV RNA load in a subject.
  • the present invention provides a use of any of the compounds of the invention for manufacture of a medicament to treat HCV infection in a subject.
  • the invention provides a method of manufacture of a medicament, including formulating any of the compounds of the present invention for treatment of a subject.
  • the term “treat,” “treated,” “treating” or “treatment” includes the diminishment or alleviation of at least one symptom associated or caused by the state, disorder or disease being treated.
  • the treatment comprises the induction of an HCV- inhibited state, followed by the activation of the HCV-modulating compound, which would in turn diminish or alleviate at least one symptom associated or caused by the HCV-associated state, disorder or disease being treated.
  • treatment can be diminishment of one or several symptoms of a disorder or complete eradication of a disorder.
  • subject is intended to include organisms, e.g., prokaryotes and eukaryotes, which are capable of suffering from or afflicted with an HCV-associated disorder.
  • subjects include mammals, e.g., humans, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats, and transgenic non-human animals.
  • the subject is a human, e.g., a human suffering from, at risk of suffering from, or potentially capable of suffering from an HCV-associated disorder, and for diseases or conditions described herein, e.g., HCV infection.
  • the subject is a cell.
  • HCV-modulating compound refers to compounds that modulate, e.g. , inhibit, or otherwise alter, the activity of HCV.
  • an "NS3/NS4A protease inhibitor,” or an “NS2/NS3 protease inhibitor” refers to a compound that modulates, e.g., inhibits, or otherwise alters, the interaction of these proteases with one another.
  • HCV-modulating compounds include compounds of Formula I, as well as Table A and Table B (including pharmaceutically acceptable salts thereof, as well as enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof).
  • the method includes administering to a subject an effective amount of an HCV-modulating compound of the invention, e.g., HCV-modulating compounds of Formula I, as well as Table A and Table B (including pharmaceutically acceptable salts thereof, as well as enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof).
  • an HCV-modulating compound of the invention e.g., HCV-modulating compounds of Formula I, as well as Table A and Table B (including pharmaceutically acceptable salts thereof, as well as enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof).
  • alkyl includes saturated aliphatic groups, including straight-chain alkyl groups (e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, etc.), branched-chain alkyl groups (isopropyl, tert-butyl, isobutyl, etc.), cycloalkyl (alicyclic) groups (cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl), alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups.
  • straight-chain alkyl groups e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl,
  • alkyl also includes alkenyl groups and alkynyl groups.
  • C x -C y -alkyl indicates a particular alkyl group (straight- or branched-chain) of a particular range of carbons.
  • Ci-C 4 -alkyl includes, but is not limited to, methyl, ethyl, propyl, butyl, isopropyl, tert-butyl, isobutyl and sec-butyl.
  • C 3 - 6 -cycloalkyl includes, but is not limited to, cyclopropyl, cyclopentyl, and cyclohexyl. As discussed below, these alkyl groups, as well as cycloalkyl groups, may be further substituted.
  • "Co-C n alkyl” refers to a single covalent bond (Co) or an alkyl group having from 1 to n carbon atoms; for example "Co-C 4 alkyl” refers to a single covalent bond or a C]-C 4 alkyl group; “Co-C 8 alkyl” refers to a single covalent bond or a Ci-C 8 alkyl group.
  • a substituent of an alkyl group is specifically indicated.
  • Ci- C4hydroxyalkyl refers to a Ci-C 4 alkyl group that has at least one hydroxy substituent.
  • Alkylene refers to a divalent alkyl group, as defined above.
  • Co-C 4 alkylene is a single covalent bond or an alkylene group having from 1 to 4 carbon atoms; and C 0 -
  • C ⁇ alkylene is a single covalent bond or an alkylene group having from 1 to 6 carbon atoms.
  • Alkenylene and Alkynylene refer to divalent alkenyl and alkynyl groups respsectively, as defined above.
  • alkyl further includes alkyl groups which can further include oxygen, nitrogen, sulfur or phosphorous atoms replacing one or more carbons of the hydrocarbon backbone.
  • a straight chain or branched chain alkyl has 10 or fewer carbon atoms in its backbone (e.g., Ci-Qo for straight chain, C 3 -C 10 for branched chain), and more preferably 6 or fewer carbons.
  • a “cycloalkyl” is a group that comprises one or more saturated and/or partially saturated rings in which all ring members are carbon, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, adamantyl, decahydro-naphthalenyl, octahydro-indenyl, and partially saturated variants of the foregoing, such as cyclohexenyl.
  • Cycloalkyl groups do not comprise an aromatic ring or a heterocyclic ring.
  • Certain cycloalkyl groups are C 3 -C 8 cycloalkyl, in which the group contains a single ring with from 3 to 8 ring members.
  • a "(C3-C 8 cycloalkyl)Co-C 4 alkyl” is a C 3 -C 8 cycloalkyl group linked via a single covalent bond or a Ci-C 4 alkylene group.
  • C 3-6 -cycloalkyl groups are substituted one or more times (or preferably between one and five times) with substitutents independently selected from a halogen atom, aryl, heteroaryl, trihalomethyl, Ci ⁇ -alkoxy or Ci-4-alkyl.
  • alkyl e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, etc.
  • alkyl include both "unsubstituted alkyl” and “substituted alkyl", the latter of which refers to alkyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone, which allow the molecule to perform its intended function.
  • substituted is intended to describe moieties having substituents replacing a hydrogen on one or more atoms, e.g. C, O or N, of a molecule.
  • substituents can include, for example, alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, al
  • substituents of the invention include moieties selected from straight or branched alkyl (preferably C 1 -C 5 ), cycloalkyl (preferably C 3 -C 8 ), alkoxy (preferably Ci-C 6 ), thioalkyl (preferably Ci-C 6 ), alkenyl (preferably C 2 -C 6 ), alkynyl (preferably C 2 -C 6 ), heterocyclic, carbocyclic, aryl (e.g., phenyl), aryloxy (e.g., phenoxy), aralkyl (e.g., benzyl), aryloxyalkyl (e.g.
  • substituents can include, for example, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, oxime, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, sulfonato, sulfamoyl, sulfonamide, nitro, triflu
  • Cycloalkyls can be further substituted, e.g., with the substituents described above.
  • An "aralkyl” moiety is an alkyl substituted with an aryl (e.g., phenylmethyl (/.e., benzyl)).
  • alkenyl includes unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but which contain at least one double bond.
  • alkenyl includes straight-chain alkenyl groups (e.g. , ethenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl, nonenyl, decenyl, etc.), branched- chain alkenyl groups, cycloalkenyl (alicyclic) groups (cyclopropenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl), alkyl or alkenyl substituted cycloalkenyl groups, and cycloalkyl or cycloalkenyl substituted alkenyl groups.
  • alkenyl includes straight-chain alkenyl groups (e.g. , ethenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl, nonenyl
  • alkenyl further includes alkenyl groups that include oxygen, nitrogen, sulfur or phosphorous atoms replacing one or more carbons of the hydrocarbon backbone.
  • a straight chain or branched chain alkenyl group has 6 or fewer carbon atoms in its backbone (e.g., C 2 -C 6 for straight chain, C 3 -C 6 for branched chain).
  • cycloalkenyl groups may have from 3-8 carbon atoms in their ring structure, and more preferably have 5 or 6 carbons in the ring structure.
  • C 2 -C 6 includes alkenyl groups containing 2 to 6 carbon atoms.
  • alkenyl includes both "unsubstituted alkenyls" and “substituted alkenyls”, the latter of which refers to alkenyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone.
  • substituents can include, for example, alkyl groups, alkynyl groups, halogens, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino
  • alkynyl includes unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but which contain at least one triple bond.
  • alkynyl includes straight-chain alkynyl groups (e.g., ethynyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl, octynyl, nonynyl, decynyl, etc.), branched- chain alkynyl groups, and cycloalkyl or cycloalkenyl substituted alkynyl groups.
  • alkynyl further includes alkynyl groups that include oxygen, nitrogen, sulfur or phosphorous atoms replacing one or more carbons of the hydrocarbon backbone.
  • a straight chain or branched chain alkynyl group has 6 or fewer carbon atoms in its backbone (e.g., C 2 -C 6 for straight chain, C 3 -C 6 for branched chain).
  • C 2 -C 6 includes alkynyl groups containing 2 to 6 carbon atoms.
  • alkynyl includes both "unsubstituted alkynyls" and “substituted alkynyls", the latter of which refers to alkynyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone.
  • substituents can include, for example, alkyl groups, alkynyl groups, halogens, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate,
  • amine or “amino” should be understood as being broadly applied to both a molecule, or a moiety or functional group, as generally understood in the art, and may be primary, secondary, or tertiary.
  • amine or “amino” includes compounds where a nitrogen atom is covalently bonded to at least one carbon, hydrogen or heteroatom.
  • alkylamino comprises groups and compounds wherein the nitrogen is bound to at least one additional alkyl group.
  • dialkyl amino includes groups wherein the nitrogen atom is bound to at least two additional alkyl groups.
  • arylamino and diarylamino include groups wherein the nitrogen is bound to at least one or two aryl groups, respectively.
  • alkylarylamino refers to an amino group which is bound to at least one alkyl group and at least one aryl group.
  • alkaminoalkyl refers to an alkyl, alkenyl, or alkynyl group bound to a nitrogen atom which is also bound to an alkyl group.
  • amide includes compounds or moieties which contain a nitrogen atom which is bound to the carbon of a carbonyl or a thiocarbonyl group.
  • the term includes "alkaminocarbonyl” or “alkylaminocarbonyl” groups which include alkyl, alkenyl, aryl or alkynyl groups bound to an amino group bound to a carbonyl group. It includes.arylaminocarbonyl and arylcarbonylamino groups which include aryl or heteroaryl moibties bound to an amino group which is bound to the carbon of a carbonyl or thiocarbonyl group.
  • alkylaminocarbonyl alkenylaminocarbonyl
  • alkynylaminocarbonyl alkynylaminocarbonyl
  • arylaminocarbonyl alkylcarbonylamino
  • alkenylcarbonylamino alkynylcarbonylamino
  • arylcarbonylamino alkylcarbonylamino
  • alkenylcarbonylamino alkynylcarbonylamino
  • arylcarbonylamino alkylcarbonylamino
  • aryl includes groups, including 5- and 6-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, phenyl, pyrrole, furan, thiophene, thiazole, isothiaozole, imidazole, triazole, tetrazole, pyrazole, oxazole, isoxazole, pyridine, pyrazine, pyridazine, and pyrimidine, and the like.
  • aryl includes multicyclic aryl groups, e.g., tricyclic, bicyclic, e.g., naphthalene, benzoxazole, benzodioxazole, benzothiazole, benzoimidazole, benzothiophene, methylenedioxyphenyl, quinoline, isoquinoline, anthryl, phenanthryl, napthridine, indole, benzofuran, purine, benzofuran, deazapurine, or indolizine.
  • multicyclic aryl groups e.g., tricyclic, bicyclic, e.g., naphthalene, benzoxazole, benzodioxazole, benzothiazole, benzoimidazole, benzothiophene, methylenedioxyphenyl, quinoline, isoquinoline, anthryl, phenanthryl, napthridine, indole, benzofuran, purine,
  • aryl groups having heteroatoms in the ring structure may also be referred to as "aryl heterocycles", “heterocycles,” “heteroaryls” or “heteroaromatics.”
  • the aromatic ring can be substituted at one or more ring positions with such substituents as described above, as for example, alkyl, halogen, hydroxyl, alkoxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkylaminoacarbonyl, aralkylaminocarbonyl, alkenylaminocarbonyl, alkylcarbonyl, arylcarbonyl, aralkylcarbonyl, alkenylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino,
  • aryl groups recited herein are C 6 -CioarylC o -Cgalkyl groups (i.e., groups in which a 6- to 10-membered carbocyclic group comprising at least one aromatic ring is linked via a single covalent bond or a CrCgalkylene group).
  • Such groups include, for example, phenyl and indanyl, as well as groups in which either of the foregoing is linked via Ci- C 8 alkylene, preferably via Ci-C 4 alkylene.
  • Phenyl groups linked via a single covalent bond or Ci-C 6 alkylene group are designated phenylC ⁇ -C ⁇ alkyl (e.g., benzyl, 1-phenyl-ethyl, 1-phe ⁇ yl- propyl and 2-phenyl-ethyl).
  • Arylene refers to a divalent aryl group, as defined above. Arylene is intended to encompass divalent residues of phenyl, naphthyl and biphenyl. "Heteroarylene” refers to divalent heteroaryl groups as defined infra.
  • heteroaryl represents a stable monocyclic or bicyclic ring of up to 7 atoms in each ring, wherein at least one ring is aromatic and contains from 1 to 4 heteroatoms selected from the group consisting of O, N and S.
  • Heteroaryl groups within the scope of this definition include but are not limited to: acridinyl, carbazolyl, cinnolinyl, quinoxalinyl, pyrrazolyl, indolyl, isoindoline, benzotriazolyl, furanyl, thienyl, benzothienyl, benzofuranyl, quinolinyl, isoquinolinyl, oxazolyl, isoxazolyl, indolyl, pyrazinyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolyl, tetrahydroquinoline.
  • heteroaryl is also understood to include the N-oxide derivative of any nitrogen- containing heteroaryl.
  • heteroaryl substituent is bicyclic and one ring is non-aromatic or contains no heteroatoms, it is understood that attachment is via the aromatic ring or via the heteroatom containing ring, respectively.
  • heterocycle or “heterocyclyl” as used herein is intended to mean a 5- to 10-membered aromatic or nonaromatic heterocycle containing from 1 to 4 heteroatoms selected from the group consisting of O, N and S, and includes bicyclic groups.
  • Heterocyclyl therefore includes the above mentioned heteroaryls, as well as dihydro and tetrathydro analogs thereof.
  • heterocyclyl include, but are not limited to the following: benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, imidazolyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridazinyl, pyridazinyl
  • a “heterocycleCo-Cgalkyl” is a heterocyclic group linked via a single covalent bond or Ci-Qalkylene group.
  • a (4- to 7-membered heterocycle)C 0 -C 8 alkyl is a heterocyclic group (e.g., monocyclic or bicyclic) having from 4 to 7 ring members linked via a single covalent bond or an alkylene group having from 1 to 8 carbon atoms.
  • a “(6-membered heteroaryl)Co- C 6 alkyl” refers to a heteroaryl group linked via a direct bond or Ci-C 6 alkyl group.
  • acyl includes compounds and moieties which contain the acyl radical (CH 3 CO-) or a carbonyl group.
  • substituted acyl includes acyl groups where one or more of the hydrogen atoms are replaced by for example, alkyl groups, alkynyl groups, halogens, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, ary
  • acylamino includes moieties wherein an acyl moiety is bonded to an amino group.
  • the term includes alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido groups.
  • alkoxy includes substituted and unsubstituted alkyl, alkenyl, and alkynyl groups covalently linked to an oxygen atom. Examples of alkoxy groups include methoxy, ethoxy, isopropyloxy, propoxy, butoxy, and pentoxy groups and may include cyclic groups such as cyclopentoxy. Examples of substituted alkoxy groups include halogenated alkoxy groups.
  • the alkoxy groups can be substituted with groups such as alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate
  • carbonyl or “carboxy” includes compounds and moieties which contain a carbon connected with a double bond to an oxygen atom, and tautomeric forms thereof.
  • moieties that contain a carbonyl include aldehydes, ketones, carboxylic acids, amides, esters, anhydrides, etc.
  • carboxy moiety refers to groups such as “alkylcarbonyl” groups wherein an alkyl group is covalently bound to a carbonyl group, "alkenylcarbonyl” groups wherein an alkenyl group is covalently bound to a carbonyl group, "alkynylcarbonyl” groups wherein an alkynyl group is covalently bound to a carbonyl group, “arylcarbonyl” groups wherein an aryl group is covalently attached to the carbonyl group.
  • the term also refers to groups wherein one or more heteroatoms are covalently bonded to the carbonyl moiety.
  • the term includes moieties such as, for example, aminocarbonyl moieties, (wherein a nitrogen atom is bound to the carbon of the carbonyl group, e.g., an amide), aminocarbonyloxy moieties, wherein an oxygen and a nitrogen atom are both bond to the carbon of the carbonyl group (e.g., also referred to as a "carbamate").
  • aminocarbonylamino groups e.g., ureas
  • heteroatom can be further substituted with one or more alkyl, alkenyl, alkynyl, aryl, aralkyl, acyl, etc. moieties.
  • thiocarbonyl or “thiocarboxy” includes compounds and moieties which contain a carbon connected with a double bond to a sulfur atom.
  • thiocarbonyl moiety includes moieties that are analogous to carbonyl moieties.
  • thiocarbonyl moieties include aminothiocarbonyl, wherein an amino group is bound to the carbon atom of the thiocarbonyl group, furthermore other thiocarbonyl moieties include, oxythiocarbonyls (oxygen bound to the carbon atom), aminothiocarbonylamino groups, etc.
  • ether includes compounds or moieties that contain an oxygen bonded to two different carbon atoms or heteroatoms.
  • alkoxyalkyl which refers to an alkyl, alkenyl, or alkynyl group covalently bonded to an oxygen atom that is covalently bonded to another alkyl group.
  • esters includes compounds and moieties that contain a carbon or a heteroatom bound to an oxygen atom that is bonded to the carbon of a carbonyl group.
  • ester includes alkoxycarboxy groups such, as methoxycarboayl, ethoxycarbonyl, propoxycarbonyl, butoxycarbonyl, pentoxycarbonyl, etc.
  • alkyl, alkenyl, or alkynyl groups are as defined above.
  • thioether includes compounds and moieties which contain a sulfur atom bonded to two different carbon or hetero atoms.
  • examples of thioethers include, but are not limited to alkthioalkyls, alkthioalkenyls, and alkthioalkynyls.
  • alkthioalkyls include compounds with an alkyl, alkenyl, or alkynyl group bonded to a sulfur atom that is bonded to an alkyl group.
  • alkthioalkenyls and alkthioalkynyls refer to compounds or moieties wherein an alkyl, alkenyl, or alkynyl group is bonded to a sulfur atom which is covalently bonded to an alkynyl group.
  • hydroxy or "hydroxyl” includes groups with an -OH or -O-.
  • halogen includes fluorine, bromine, chlorine, iodine, etc.
  • perhalogenated generally refers to a moiety wherein all hydrogens are replaced by halogen atoms.
  • polycyclyl or “polycyclic radical” include moieties with two or more rings ⁇ e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls) in which two or more carbons are common to two adjoining rings, e.g., the rings are “fused rings". Rings that are joined through non-adjacent atoms are termed "bridged" rings.
  • Each of the rings of the polycycle can be substituted with such substituents as described above, as for example, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkoxycarbonyl, alkylaminoacarbonyl, aralkylaminocarbonyl, alkenylaminocarbonyl, alkylcarbonyl, arylcarbonyl, aralkylcarbonyl, alkenylcarbonyl, aminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and urei
  • Preferred heteroatoms are nitrogen, oxygen, sulfur and phosphorus.
  • any combination thereof implies that any number of the listed functional groups and molecules may be combined to create a larger molecular architecture.
  • bonds and/or hydrogen atoms are added to provide the following number of total bonds to each of the following types of atoms: carbon: four bonds; nitrogen: three bonds; oxygen: two bonds; and sulfur: two bonds.
  • Groups that are "optionally substituted” are unsubstituted or are substituted by other than hydrogen at one or more available positions, typically 1, 2, 3, 4 or 5 positions, by one or more suitable groups (which may be the same or different).
  • Optional substitution is also indicated by the phrase "substituted with from 0 to X substituents," where X is the maximum number of possible substituents.
  • Certain optionally substituted groups are substituted with from 0 to 2, 3 or 4 independently selected substituents (i.e., are unsubstituted or substituted with up to the recited maximum number of substitutents).
  • the structures of some of the compounds of this invention include asymmetric carbon atoms. It is to be understood accordingly that the isomers arising from such asymmetry (e.g., all enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates) are included within the scope of this invention. Such isomers can be obtained in substantially pure form by classical separation techniques and by stereochemical ⁇ controlled synthesis. Furthermore, the structures and other compounds and moieties discussed in this application also include all tautomers thereof. Compounds described herein may be obtained through art recognized synthesis strategies. It will also be noted that the substituents of some of the compounds of this invention include isomeric cyclic structures.
  • tetrazole includes tetrazole, 2H-tetrazole, 3H- tetrazole, 4H-tetrazole and 5H-tetrazole.
  • the compounds of the present invention have valuable pharmacological properties and are useful in the treatment of diseases.
  • compounds of the invention are useful in the treatment of ⁇ CV-associated disorders, e.g., as drugs to treat HC V infection.
  • use includes any one or more of the following embodiments of the invention, respectively: the use in the treatment of HCV-associated disorders; the use for the manufacture of pharmaceutical compositions for use in the treatment of these diseases, e.g., in the manufacture of a medicament; methods of use of compounds of the invention in the treatment of these diseases; pharmaceutical preparations having compounds of the invention for the treatment of these diseases; and compounds of the invention for use in the treatment of these diseases; as appropriate and expedient, if not stated otherwise.
  • diseases to be treated and are thus preferred for use of a compound of the present invention are selected from HCV-associated disorders, including those corresponding to HCV-infection, as well as those diseases that depend on the activity of one or more of the NS3, NS4A, NS4B, NS5A and NS5B proteins, or a NS3-NS4A, NS4A-NS4B, NS4B-NS5A or NS5A-NS5B complex.
  • the term "use” further includes embodiments of compositions herein which bind to an HCV protein sufficiently to serve as tracers or labels, so that when coupled to a fluor or tag, or made radioactive, can be used as a research reagent or as a diagnostic or an imaging agent.
  • a compound of the present invention is used for treating
  • HCV-associated diseases and use of the compound of the present invention as an inhibitor of any one or more HCVs. It is envisioned that a use can be a treatment of inhibiting one or more strains of HCV.
  • Assays The inhibition of HCV activity may be measured as using a number of assays available in the art. An example of such an assay can be found in Anal Biochem. 1996 240(1): 60-7; which is incorporated by reference in its entirety. Assays for measurement of HCV activity are also described in the experimental section below.
  • an effective amount of the compound is that amount necessary or sufficient to treat or prevent an HCV-associated disorder, e.g. prevent the various morphological and somatic symptoms of an HCV-associated disorder, and/or a disease or condition described herein.
  • an effective amount of the HCV -modulating compound is the amount sufficient to treat HCV infection in a subject.
  • an effective amount of the HCV-modulating compound is the amount sufficient to treat HCV infection, liver cirrhosis, chronic liver disease, hepatocellular carcinoma, cryoglobulinaemia, non-Hodgkin's lymphoma, and a suppressed innate intracellular immune response in a subject.
  • the effective amount can vary depending on such factors as the size and weight of the subject, the type of illness, or the particular compound of the invention. For example, the choice of the compound of the invention can affect what constitutes an "effective amount.” One of ordinary skill in the art would be able to study the factors contained herein and make the determination regarding the effective amount of the compounds of the invention without undue experimentation.
  • the regimen of administration can affect what constitutes an effective amount.
  • the compound of the invention can be administered to the subject either prior to or after the onset of an HCV-associated state. Further, several divided dosages, as well as staggered dosages, can be administered daily or sequentially, or the dose can be continuously infused, or can be a bolus injection. Further, the dosages of the compound(s) of the invention can be proportionally increased or decreased as indicated by the exigencies of the therapeutic or prophylactic situation.
  • Compounds of the invention may be used in the treatment of states, disorders or diseases as described herein, or for the manufacture of pharmaceutical compositions for use in the treatment of these diseases. Methods of use of compounds of the present invention in the treatment of these diseases, or pharmaceutical preparations having compounds of the present invention for the treatment of these diseases.
  • composition includes preparations suitable for administration to mammals, e.g., humans.
  • pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
  • phrases "pharmaceutically acceptable carrier” is art recognized and includes a pharmaceutically acceptable material, composition or vehicle, suitable for administering compounds of the present invention to mammals.
  • the carriers include liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer'
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • antioxidants examples include: water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, ⁇ -tocopherol, and the like; and metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin
  • Formulations of the present invention include those suitable for oral, nasal, topical, transdermal, buccal, sublingual, rectal, vaginal and/or parenteral administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
  • the amount of active ingredient that can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound that produces a therapeutic effect. Generally, out of one hundred per cent, this amount will range from about 1 per cent to about ninety-nine percent of active ingredient, preferably from about 5 per cent to about 70 per cent, most preferably from about 10 per cent to about 30 per cent.
  • Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention with the carrier and, optionally, one or more accessory ingredients, hi general, the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • Formulations of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient.
  • a compound of the present invention may also be administered as a bolus, electuary or paste.
  • the active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; humectants, such as glycerol; disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; solution retarding agents, such as paraffin; absorption accelerators, such as quaternary ammonium compounds; wetting agents, such as, for example, cetyl alcohol and glycerol monostea
  • the pharmaceutical compositions may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres.
  • compositions may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions that can be dissolved in sterile water, or some other sterile injectable medium immediately before use.
  • These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • embedding compositions that can be used include polymeric substances and waxes.
  • the active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Liquid dosage forms for oral administration of the compounds of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluent commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsif ⁇ ers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluent commonly used in the art, such as, for example, water or other solvents, solubilizing agents and
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • Suspensions in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • Formulations of the pharmaceutical compositions of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.
  • Formulations of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration of a compound of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that may be required.
  • the ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to a compound of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances.
  • Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • Transdermal patches have the added advantage of providing controlled delivery of a compound of the present invention to the body.
  • dosage forms can be made by dissolving or dispersing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the active compound in a polymer matrix or gel.
  • compositions of this invention suitable for parenteral administration comprise one or more compounds of the invention in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • These compositions may also contain adjuvants such as preservatives, wetting agents,emulsifying agents and dispersing agents.
  • microorganisms Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents that delay absorption such as aluminum monostearate and gelatin.
  • antibacterial and antifungal agents for example, paraben, chlorobutanol, phenol sorbic acid, and the like.
  • isotonic agents such as sugars, sodium chloride, and the like into the compositions.
  • prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents that delay absorption such as aluminum monostearate and gelatin.
  • the absorption of the drug in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally-administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
  • Injectable depot forms are made by forming microencapsule matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions that are compatible with body tissue. The preparations of the present invention may be given orally, parenterally, topically, or rectally. They are of course given by forms suitable for each administration route.
  • they are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, etc., administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories.
  • Oral administration is preferred.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
  • systemic administration means the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient's system and, thus, is subject to metabolism and other like processes, for example ⁇ subcutaneous administration.
  • These compounds may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracisternally and topically, as by powders, ointments or drops, including buccally and sublingually.
  • the compounds of the present invention which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level will depend upon a variety of factors including the activity of the particular compound of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required.
  • the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • a suitable daily dose of a compound of the invention will be that amount of the compound that is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above. Generally, intravenous and subcutaneous doses of the compounds of this invention for a patient, when used for the indicated analgesic effects, will range from about 0.0001 to about 100 mg per kilogram of body weight per day, more preferably from about 0.01 to about 50 mg per kg per day, and still more preferably from about 1.0 to about 100 mg per kg per day. An effective amount is that amount treats an HCV-associated disorder. If desired, the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
  • protecting group a readily removable group that is not a constituent of the particular desired end product of the compounds of the present invention.
  • the protection of functional groups by such protecting groups, the protecting groups themselves, and their cleavage reactions are described for example in standard reference works, such as e.g., Science of Synthesis: Houben-Weyl Methods of Molecular Transformation. Georg Thieme Verlag, Stuttgart, Germany. 2005. 41627 pp. (URL: http://www.science-of-synthesis.com (Electronic Version, 48 Volumes)); J. F. W. McOmie, "Protective Groups in Organic Chemistry", Plenum Press, London and New York 1973, in T. W. Greene and P. G.
  • Salts of compounds of the present invention having at least one salt- forming group may be prepared in a manner known per se.
  • salts of compounds of the present invention having acid groups may be formed, for example, by treating the compounds with metal compounds, such as alkali metal salts of suitable organic carboxylic acids, e.g., the sodium salt of 2-ethylhexanoic acid, with organic alkali metal or alkaline earth metal compounds, such as the corresponding hydroxides, carbonates or hydrogen carbonates, such as sodium or potassium hydroxide, carbonate or hydrogen carbonate, with corresponding calcium compounds or with ammonia or a suitable organic amine, stoichiometric amounts or only a small excess of the salt-forming agent preferably being used.
  • metal compounds such as alkali metal salts of suitable organic carboxylic acids, e.g., the sodium salt of 2-ethylhexanoic acid
  • organic alkali metal or alkaline earth metal compounds such as the corresponding hydroxides, carbonates or hydrogen carbonates, such as sodium or potassium hydroxide, carbonate or hydrogen carbonate, with corresponding calcium compounds or with ammonia or a suitable
  • Acid addition salts of compounds of the present invention are obtained in customary manner, e.g., by treating the compounds with an acid or a suitable anion exchange reagent.
  • Internal salts of compounds of the present invention containing acid and basic salt-forming groups, e.g., a free carboxy group and a free amino group, may be formed, e.g., by the neutralisation of salts, such as acid addition salts, to the isoelectric point, e.g., with weak bases, or by treatment with ion exchangers.
  • Salts can be converted in customary manner into the free compounds; metal and ammonium salts can be converted, for example, by treatment with suitable acids, and acid addition salts, for example, by treatment with a suitable basic agent.
  • diastereoisomers can be separated in a manner known per se into the individual isomers; diastereoisomers can be separated, for example, by partitioning between polyphasic solvent mixtures, recrystallisation and/or chromatographic separation, for example over silica gel or by, e.g., medium pressure liquid chromatography over a reversed phase column, and racemates can be separated, for example, by the formation of salts with optically pure salt-forming reagents and separation of the mixture of diastereoisomers so obtainable, for example by means of fractional crystallisation, or by chromatography over optically active column materials.
  • the process steps to synthesize the compounds of the invention can be carried out under reaction conditions that are known per se, including those mentioned specifically, in the absence or, customarily, in the presence of solvents or diluents, including, for example, solvents or diluents that are inert towards the reagents used and dissolve them, in the absence or presence of catalysts, condensation or neutralizing agents, for example ion exchangers, such as cation exchangers, e.g., in the H + form, depending on the nature of the reaction and/or of the reactants at reduced, normal or elevated temperature, for example in a temperature range of from about -100 0 C to about 190 0 C, including, for example, from approximately - 80 0 C to approximately 150 0 C, for example at from -80 to -60 0 C, at room temperature, at from -20 to 40 0 C or at reflux temperature, under atmospheric pressure or in a closed vessel, where appropriate under pressure, and/or in an inert atmosphere, for example under
  • mixtures of isomers that are formed can be separated into the individual isomers, for example diastereoisomers or enantiomers, or into any desired mixtures of isomers, for example racemates or mixtures of diastereoisomers, for example analogously to the methods described in Science of Synthesis: Houben-Weyl Methods of Molecular Transformation. Georg Thieme Verlag, Stuttgart, Germany. 2005.
  • solvents from which those solvents that are suitable for any particular reaction may be selected include those mentioned specifically or, for example, water, esters, such as lower alkyl-lower alkanoates, for example ethyl acetate, ethers, such as aliphatic ethers, for example diethyl ether, or cyclic ethers, for example tetrahydrofurane or dioxane, liquid aromatic hydrocarbons, such as benzene or toluene, alcohols, such as methanol, ethanol or 1- or 2-propanol, nitriles, such as acetonitrile, halogenated hydrocarbons, such as methylene chloride or chloroform, acid amides, such as dimethylformamide or dimethyl acetamide, bases, such as heterocyclic nitrogen bases, for example pyridine or N-methylpyrrolidin-2- one, carboxylic acid anhydrides, such as lower alkanoic acid anhydrides, for example acetic anhydride,
  • the compounds, including their salts, may also be obtained in the form of hydrates, or their crystals may, for example, include the solvent used for crystallization. Different crystalline forms may be present.
  • the invention relates also to those forms of the process in which a compound obtainable as an intermediate at any stage of the process is used as starting material and the remaining process steps are carried out, or in which a starting material is formed under the reaction conditions or is used in the form of a derivative, for example in a protected form or in the form of a salt, or a compound obtainable by the process according to the invention is produced under the process conditions and processed further in situ.
  • the present invention also relates to pro-drugs of a compound of the present invention that are converted in vivo to the compounds of the present invention as described herein. Any reference to a compound of the present invention is therefore to be understood as referring, also to the corresponding pro-drugs of the compound of the present invention, as appropriate and expedient.
  • Combinations A compound of the present invention may also be used in combination with other agents, e.g., an additional HCV-modulating compound that is or is not of the formula I, for treatment of and HCV-associated disorder in a subject.
  • combination is meant either a fixed combination in one dosage unit form, or a kit of parts for the combined administration where a compound of the present invention and a combination partner may be administered independently at the same time or separately within time intervals that especially allow that the combination partners show a cooperative, e.g., synergistic, effect, or any combination thereof.
  • WO 2005/042020 describes the combination of various HCV inhibitors with a cytochrome P450 ("CYP") inhibitor.
  • CYP cytochrome P450
  • Any CYP inhibitor that improves the pharmacokinetics of the relevant NS3/4A protease may be used in combination with the compounds of this invention.
  • CYP inhibitors include, but are not limited to, ritonavir (WO 94/14436, incorporated herein by reference in its entirety), ketoconazole, ⁇ -oleandomycin, 4-methyl pyrazole, cyclosporin, clomethiazole, cimetidine, itraconazole, fluconazole, miconazole, fluvoxamine, fluoxetine, nefazodone, sertraline, indinavir, nelfinavir, amprenavir, fosamprenavir, saquinavir, lopinavir, delavirdine, erythromycin, VX-944, and VX-497.
  • Preferred CYP inhibitors include ritonavir, ketoconazole, troleandomycin, 4-methyl pyrazole, cyclosporin, and clomethiazole. Methods for measuring the ability of a compound to inhibit CYP activity are known
  • a compound to be evaluated may be incubated with 0.1, 0.5, and 1.0 mg protein/ml, or other appropriate concentration of human hepatic microsomes (e. g., commercially available, pooled characterized hepatic microsomes) for 0, 5, 10, 20, and 30 minutes, or other appropriate times, in the presence of an NADPH- generating system. Control incubations may be performed in the absence of hepatic microsomes for 0 and 30 minutes (triplicate). The samples may be analyzed for the presence of the compound.
  • a compound one concentration, ⁇ K m
  • a CYP inhibitor such as ritonavir
  • control incubations should contain the same concentration of organic solvent as the incubations with the CYP inhibitor.
  • concentrations of the compound in the samples may be quantitated, and the rate of disappearance of parent compound may be determined, with rates being expressed as a percentage of control activity.
  • one embodiment of this invention provides a method for administering an inhibitor of CYP3A4 and a compound of the invention.
  • Another embodiment of this invention provides a method for administering an inhibitor of isozyme 3A4 ("CYP3A4"), isozyme 2Cl 9 ("CYP2C19”), isozyme 2D6 ("CYP2D6"), isozyme 1 A2 (“CYP 1A2”), isozyme 2C9 (“CYP2C9”), or isozyme 2El (“CYP2E1").
  • the protease inhibitor is VX-950 (or a sterereoisomer thereof)
  • the CYP inhibitor preferably inhibits CYP3A4.
  • CYP3A4 activity is broadly observed in humans.
  • this invention provides methods wherein the CYP inhibitor is . administered together with the compound of the invention in the same dosage form or in separate dosage forms.
  • the compounds of the invention may be administered as the sole ingredient or in combination or alteration with other antiviral agents, especially agents active against HCV.
  • combination therapy effective dosages of two or more agents are administered together, whereas in alternation or sequential-step therapy, an effective dosage of each agent is administered serially or sequentially.
  • combination therapy is typically preferred over alternation therapy because it induces multiple simultaneous stresses on the virus.
  • the dosages given will depend on absorption, inactivation and excretion rate of the drug as well as other factors. It is to be noted that dosage values will also vary with the severity of the condition to be alleviated.
  • Daily dosages required in practicing the method of the present invention will vary depending upon, for example, the compound of the invention employed , the host, the mode of administration, the severity of the condition to be treated.
  • a preferred daily dosage range is about from 1 to 50 mg/kg per day as a single dose or in divided doses.
  • Suitable daily dosages for patients are on the order of from e.g. 1 to 20 mg/kg p.o or i.v.
  • Suitable unit dosage forms for oral administration comprise from ca. 0.25 to 10 mg/kg active ingredient, e.g. compound of Formula I or any subformulae thereof, together with one or more pharmaceutically acceptable diluents or carriers therefor.
  • the amount of co-agent in the dosage form can vary greatly, e.g., 0.00001 to 1000mg/kg active ingredient.
  • daily dosages with respect to the co-agent used will vary depending upon, for example, the compound employed, the host, the mode of administration and the severity of the condition to be treated.
  • lamivudine may be administered at a daily dosage of lOOmg.
  • the pegylated interferon may be administered parenterally one to three times per week, preferably once a week, at a total weekly dose ranging from 2 to 10 million IU, more preferable 5 to 10 million IU, most preferable 8 to 10 million IU. Because of the diverse types of co-agent that may be used, the amounts can vary greatly, e.g., .0001 to 5,000 mg/kg per day.
  • the current standard of care for treating hepatitis C is the combination of pegylated interferon alpha with ribavirin, of which the recommended doses are 1.5 ⁇ g/kg/wk peginterferon alfa-2b or 180 ⁇ g/wk peginterferon alfa-2a, plus 1,000 to 1,200 mg daily of ribavirin for 48 weeks for genotype I patients, or 800 mg daily of ribavirin for 24 weeks for genotype 2/3 patients.
  • the compound of the invention e.g., compound of Formula I or subformulae thereof
  • co-agents of the invention may be administered by any conventional route, in particular enterally, e.g. orally, for example in the form of solutions for drinking, tablets or capsules or parenterally, for example in the form of injectable solutions or suspensions.
  • Certain preferred pharmaceutical compositions may be e.g. those based on microemulsions as described in UK 2,222,770 A.
  • the compound of the invention are administered together with other drugs (co-agents) e.g. a drug which has anti-viral activity, especially anti-Flaviviridae activity, most especially anti-HCV activity, e.g. an interferon, e.g. interferon- ⁇ -2a or interferon- ⁇ -2b, e.g. Intron R A, Roferon R , Avonex R , Rebif ⁇ or Betaferon R , or an interferon conjugated to a water soluble polymer or to human albumin, e.g. albuferon, an anti-viral agent, e.g.
  • drugs co-agents
  • drugs co-agents
  • a drug which has anti-viral activity, especially anti-Flaviviridae activity, most especially anti-HCV activity e.g. an interferon, e.g. interferon- ⁇ -2a or interferon- ⁇ -2b, e.g. Intron R A, Roferon R
  • ribavirin lamivudine, the compounds disclosed in US patent no. 6,812,219 and WO 2004/002422 A2 (the disclosures of which are incorporated herein by reference in their entireties), an inhibitor of the HCV or other Flaviviridae virus encoded factors like the NS3/4A protease, helicase or RNA polymerase or a prodrug of such an inhibitor, an anti-fibrotic agent, e.g. a N-phenyl-2-pyrimidine-amine derivative, e.g. imatinib, an immune modulating agent, e.g. mycophenolic acid, a salt or a prodrug thereof, e.g.
  • an anti-fibrotic agent e.g. a N-phenyl-2-pyrimidine-amine derivative, e.g. imatinib
  • an immune modulating agent e.g. mycophenolic acid, a salt or a prodrug thereof, e.g.
  • sodium mycophenolate or mycophenolate mofetil or a SlP receptor agonist, e.g. FTY720 or an analogue thereof optionally phosphorylated, e.g. as disclosed in EP627406A1, EP778263A1, EP1002792A1, WO02/18395, WO02/76995, WO 02/06268, JP2002316985, WO03/29184, WO03/29205, WO03/62252 and WO03/62248, the disclosures of which are incorporated herein by reference in their entireties.
  • SlP receptor agonist e.g. FTY720 or an analogue thereof optionally phosphorylated, e.g. as disclosed in EP627406A1, EP778263A1, EP1002792A1, WO02/18395, WO02/76995, WO 02/06268, JP2002316985, WO03/29184, WO03/29205, WO03/62252 and WO03/62248, the disclosures of
  • Conjugates of interferon to a water-soluble polymer are meant to include especially conjugates to polyalkylene oxide homopolymers such as polyethylene glycol (PEG) or polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereof.
  • polyalkylene oxide-based polymers effectively non- antigenic materials such as dextran, polyvinyl pyrrolidones, polyacrylamides, polyvinyl alcohols, carbohydrate-based polymers and the like can be used.
  • Such interferon-polymer conjugates are described in U.S. Pat. Nos. 4,766,106, 4,917,888, European Patent Application No. 0 236 987, European Patent Application No.
  • Interferon used to prepare polymer conjugates may be prepared from a mammalian extract, such as human, ruminant or bovine interferon, or recombinantly produced. Preferred are conjugates of interferon to polyethylene glycol, also known as pegylated interferons.
  • pegylated alfa-interferons for example pegylated interferon- a -2a, pegylated interferon- a -2b; pegylated consensus interferon or pegylated purified interferon- ⁇ product.
  • pegylated interferon- a -2a is described e.g. in European Patent 593,868 (incorporated herein by reference in its entirety) and commercially available e. g. under the tradename PEGASYS ® (Hoffmann-La Roche).
  • Pegylated interferon- ⁇ -2b is described, e.g.
  • Pegylated consensus interferon is described in WO 96/11953 (incorporated herein by reference in its entirety).
  • the preferred pegylated a- interferons are pegylated interferon- ⁇ -2a and pegylated interferon- ⁇ -2b. Also preferred is pegylated consensus interferon.
  • fusion proteins of an interferon for example fusion proteins of interferon- ⁇ -2a, interferon-Q!-2b; consensus interferon or purified interferon- ⁇ product, each of which is fused with another protein.
  • Certain preferred fusion proteins comprise an interferon (e.g., interferon- ⁇ -2b) and an albumin as described in U.S. Patent 6,973,322 and international publications WO02/60071, WO05/003296 and WO05/077042 (Human Genome Sciences).
  • a preferred interferon conjugated to a human albumin is Albuferon (Human Genome Sciences).
  • Cyclosporins which bind strongly to cyclophilin but are not immunosuppressive include those cyclosporins recited in U.S. Patents 5,767,069 and 5,981,479 and are incorporated herein by reference. Melle 4 -Cyclosporin is a preferred non-immunosuppressive cyclosporin. Certain other cyclosporin derivatives are described in WO2006039668 (Scynexis) and WO2006038088 (Debiopharm SA) and are incorporated herein by reference.
  • a cyclosporin is considered to be non-immunosuppressive when it has an activity in the Mixed Lymphocyte Reaction (MLR) of no more than 5%, preferably no more than 2%, that of cyclosporin A.
  • MLR Mixed Lymphocyte Reaction
  • the Mixed Lymphocyte Reaction is described by T. Meo in
  • Spleen cells (0.5 x 10 6 ) from Balb/c mice (female, 8 - 10 weeks) are co- incubated for 5 days with 0.5 x 10 6 irradiated (2000 rads) or mitomycin C treated spleen cells from CBA mice (female, 8 - 10 weeks).
  • the irradiated allogeneic cells induce a proliferative response in the BaIb c spleen cells which can be measured by labeled precursor incorporation into the DNA.
  • the stimulator cells are irradiated (or mitomycin C treated) they do not respond to the Balb/c cells with proliferation but do retain their antigenicity.
  • the IC 5O found for the test compound in the MLR is compared with that found for cyclosporin A in a parallel experiment.
  • non-immunosuppressive cyclosporins lack the capacity of inhibiting CN and the downstream NF-AT pathway.
  • [Melle] 4 -ciclosporin is a preferred non- immunosuppressive cyclophilin-binding cyclosporin for use according to the invention.
  • Ribavirin (l-/3-D-ribofuranosyl-l-l,2,4-triazole-3-caroxamide) is a synthetic, non- interferon-inducing, broad spectrum antiviral nucleoside analog sold under the trade name, Virazole (The Merk Index, 11 th edition, Editor: Budavar, S, Merck & Co., Inc., Rahway, NJ, pl304,1989). United States Patent No. 3,798,209 and RE29,835 (incorporated herein by reference in their entireties) disclose and claim ribavirin. Ribavirin is structurally similar to guanosine, and has in vitro activity against several DNA and RNA viruses including Flavivi ⁇ dae (Gary L. Davis, Gastroenterology 118:S1O4-S114, 2000).
  • Ribavirin reduces serum amino transferase levels to normal in 40% of patients, but it does not lower serum levels of HCV-RNA (Gary L. Davis, Gastroenterology 118:S1O4-S114, 2000). Thus, ribavirin alone is not effective in reducing viral RNA levels. Additionally, ribavirin has significant toxicity and is known to induce anemia. Ribavirin is not approved for monotherapy against HCV; it is approved in combination with interferon alpha-2a or interferon alpha-2b for the treatment of HCV.
  • a further preferred combination is a combination of a compound of the invention (e.g., a compound of Formula I or any sub formulae thereof) with a non-immunosuppressive cyclophilin-binding cyclosporine, with mycophenolic acid, a salt or a prodrug thereof, and/or with a SlP receptor agonist, e.g. FTY720.
  • a compound of the invention e.g., a compound of Formula I or any sub formulae thereof
  • a non-immunosuppressive cyclophilin-binding cyclosporine e.g., with mycophenolic acid, a salt or a prodrug thereof, and/or with a SlP receptor agonist, e.g. FTY720.
  • Additional examples of compounds that can be used in combination or alternation treatments include:
  • Interferons including interferon alpha 2a or 2b and pegylated (PEG) interferon alpha 2a or 2b, for example:
  • Intron-A® interferon alfa-2b (Schering Corporation, Kenilworth, NJ);
  • PEG-Intron® peginteferon alfa-2b (Schering Corporation, Kenilworth, NJ);
  • Alferon® a mixture of natural alpha interferons (Interferon Sciences, and Purdue Frederick Co., CT);
  • interferon beta Consensus alpha interferon from Amgen, Inc., Newbury Park, CA
  • Other forms of interferon include: interferon beta, gamma, tau and omega, such as Rebif ( Interferon beta Ia) by Serono, Omniferon (natural interferon) by Viragen, REBIF (interferon beta- Ia) by Ares-Serono, Omega Interferon by BioMedicines; oral Interferon Alpha by Amarillo Biosciences; an interferon conjugated to a water soluble polymer or to a human albumin, e.g., Albuferon (Human Genome Sciences), an antiviral agent, a consensus interferon, ovine or bovine interferon-tau
  • Conjugates of interferon to a water-soluble polymer are meant to include especially conjugates to polyalkylene oxide homopolymers such as polyethylene glocol (PEG) or polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereof.
  • polyalkylene oxide homopolymers such as polyethylene glocol (PEG) or polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereof.
  • PEG polyethylene glocol
  • polypropylene glycols polyoxyethylenated polyols
  • copolymers thereof copolymers thereof
  • block copolymers thereof block copolymers thereof.
  • polyalkylene oxid-based polymers effectively non- antigenic materials such as dextran, polyvinyl pyrrolidones, polyacrylamides, polyvinyl alcohols, carbohydrate-based polymers and the like can be used
  • Interferon used to prepare polymer conjugates may be prepared from a mammalian extract, such as human, ruminant or bovine interferon, or recombinantly produced.
  • a mammalian extract such as human, ruminant or bovine interferon, or recombinantly produced.
  • Ribavirin such as ribavirin (l-beta-D-ribofuranosyl-lH-l,2,4-triazole-3- carboxamide) from Valeant Pharmaceuticals, Inc., Costa Mesa, CA); Rebetol® from Schering Corporation, Kenilworth, NJ, and Copegus® from Hoffmann-La Roche, Nutley, NJ; and new ribavirin analogues in development such as Levovirin and Viramidine by Valeant, (3) Thiazolidine derivatives which show relevant inhibition in a reverse-phase HPLC assay with an NS3/4A fusion protein and NS5A/5B substrate (Sudo K.
  • ribavirin l-beta-D-ribofuranosyl-lH-l,2,4-triazole-3- carboxamide
  • Examples include substrate-based NS3 protease inhibitors (Attwood et al., Antiviral peptide derivatives, PCT WO 98/22496, 1998; Attwood et al., Antiviral Chemistry and Chemotherapy 1999, 10, 259-273; Attwood et al, Preparation and use of amino acid derivatives as anti-viral agents, German Patent Pub. DE 19914474; Tung et al.
  • Inhibitors of serine proteases particularly hepatitis C virus NS3 protease; PCT WO 98/17679), including alphaketoamides and hydrazinoureas, and inhibitors that terminate in an electrophile such as a boronic acid or phosphonate (Llinas-Brunet et al. Hepatitis C inhibitor peptide analogues, PCT WO 99/07734) are being investigated.
  • Non-substrate-based NS3 protease inhibitors such as 2,4,6-trihydroxy-3-nitro- benzamide derivatives (Sudo K. et al., Biochemiscal and Biophysical Research Communications, 1997, 238 643-647; Sudo K. et al.
  • Sch 68631 a phenanthrenequinone
  • HCV protease inhibitor Chom M et al., Tetrahedron Letters 37:7229-7232, 1996.
  • Sch 351633 isolated from the fungus Penicillium grieofulvum, was identified as a protease inhibitor (Chu M. et al., Bioorganic and Medicinal Chemistry Letters 9:1949-1952).
  • Nanomolar potency against the HCV NS3 protease enzyme has been achieved by the design of selective inhibitors based on the macromolecule eglin c.
  • Eglin c isolated from leech, is a potent inhibitor of several serine proteases such as S. griseus proteases A and B, V- chymotrypsin, chymase and subtilisin. Qasim M.A. et al., Biochemistry 36:1598-1607, 1997.
  • U.S. patents disclosing protease inhibitors for the treatment of HCV include, for example, U.S. Patent No. 6,004,933 to Spruce et al (incorporated herein by reference in its entirety) which discloses a class of cysteine protease inhibitors for inhibiting HCV endopeptidase 2; U.S. Patent No. 5,990,276 to Zhang et al. (incorporated herein by reference in its entirety) which discloses synthetic inhibitors of hepatitis C virus NS3 protease; U.S. Patent No. 5,538,865 to Reyes et al.(incorporated herein by reference in its entirety).
  • HCV inhibitor tripeptides are disclosed in U.S. Patent Nos. 6,534,523, 6,410,531 and 6,420,380 to Boehringer Ingelheim and WO 02/060926 to Bristol Myers Squibb (incorporated herein by reference in their entireties).
  • Diaryl peptides as NS3 serine protease inhibitors of HCV are disclosed in WO 02/48172 to Schering Corporation (incorporated herein by reference).
  • Imidazoleidinones as NS3 serine protease inhibitors of HCV are disclosed in WO 02/18198 to Schering Corporation and WO 02/48157 to Bristol Myers Squibb (incorporated herein by reference in their entireties).
  • WO 98/17679 to Vertex Pharmaceuticals and WO 02/48116 to Bristol Myers Squibb also disclose HCV protease inhibitors (incorporated herein by reference in their entireties).
  • HCV NS3-4A serine protease inhibitors including BILN 2061 by Boehringer Ingelheim, VX-950 by Vertex, SCH 6/7 by Schering-Plough, and other compounds currently in preclinical development;
  • Substrate-based NS3 protease inhibitors including alphaketoamides and hydrazinoureas, and inhibitors that terminate in an elecrophile such as a boronic acid or phosphonate;
  • Non-substrate-based NS3 protease inhibitors such as 2,4,6-trihydroxy-3-nitro- benzamide derivatives including RD3-4082 and RD3-4078, the former substituted on the amide with a 14 carbon chain and the latter processing a para-phenoxyphenyl group; and Sch68631 , a phenanthrenequinone, an HCV protease inhibitor.
  • Penicillium griseofulvum was identified as a protease inhibitor.
  • EgHn c isolated from leech is a potent inhibitor of several serine proteases such as S. griseus proteases A and B, a-chymotrypsin, chymase and subtilisin.
  • US patent no. 6004933 discloses a class of cysteine protease inhibitors from inhibiting HCV endopeptidase 2; synthetic inhibitors of HCV NS3 protease (pat), HCV inhibitor tripeptides (pat), diaryl peptides such as NS3 serine protease inhibitors of HCV (pat), Imidazolidindiones as NS3 serine protease inhibitors of HCV (pat).
  • Thiazolidines and benzanilides which show relevant inhibition in a reverse-phase HPLC assay with an NS3/4A fusion protein and NS5A/5B substrate especially compound RD- 16250 possessing a fused cinnamoyl moiety substituted with a long alkyl chain, RD4 6205 and RD4 6193 Phenan-threnequinone possessing activity against protease in a SDS-PAGE and autoradiography assay isolated from the fermentation culture broth of Streptomyces sp, Sch68631 and Sch351633, isolated from the fungus Penicillium griseofulvum, which demonstrates activity in a scintillation proximity assay. . . •
  • Nucleoside or non-nucleoside inhibitors of HCV NS5B RNA-dependent RNA polymerase such as 2'-C-methyl-3'-O-L-valine ester ribofuranosyl cytidine (Idenix) as disclosed in WO 2004/002422 A2 (incorporated herein by reference in its entirety), R803 (Rigel), JTK-003 (Japan Tabacco), HCV-086 (ViroPharma/Wyeth) and other compounds currently in preclinical development; gliotoxin (ref) and the natural product cerulenin; 2'-fluoronucleosides; other nucleoside analogues as disclosed in WO 02/057287 A2, WO 02/057425 A2, WO 01/90121, WO 01/92282, and US patent no. 6,812,219, the disclosures of which are incorporated herein by reference in their entirety.
  • Idenix 2'-C-methyl-3'-O-L
  • Idenix Pharmaceuticals discloses the use of branched nucleosides in the treatment of flaviviruses (including HCV) and pestiviruses in International Publication Nos. WO 01/90121 and WO 01/92282 (incorporated herein by reference in their entireties). Specifically, a method for the treatment of hepatitis C infection (and flaviviruses and pestiviruses) in humans and other host animals is disclosed in the Idenix publications that includes administering an effective amount of a biologically active 1 ', 2', 3' or 4'-branced B- D or B-L nucleosides or a pharmaceutically acceptable salt or prodrug thereof, administered either alone or in combination with another antiviral agent, optionally in a pharmaceutically acceptable carrier.
  • a biologically active 1 ', 2', 3' or 4'-branced B- D or B-L nucleosides or a pharmaceutically acceptable salt or prodrug thereof administered either alone or in combination with another antiviral agent, optionally
  • HCV NS3 helicase inhibitors such as VP 50406 by ViroPhama and compounds from Vertex.
  • Other helicase inhibitors (Diana G.D. et al., Compounds, compositions and methods for treatment of hepatitis C, U.S. Patent No. 5,633,358 (incorporated herein by reference in its entirety); Diana G.D. et al., Piperidine derivatives, pharmaceutical compositions thereof and their use in the treatment of hepatitis C, PCT WO 97/36554);
  • S-ODN Antisense phosphorothioate oligodeoxynucleotides (S-ODN) complementary to sequence stretches in the 5' non-coding region (NCR) of the virus (Alt M. et al., Hepatology, 1995, 22, 707-717), or nucleotides 326-348 comprising the 3 1 end of the NCR and nucleotides 371-388 located in the core coding region of the HCV RNA (Alt M. et al., Archives of Virology, 1997, 142, 589-599; Galderisi U.
  • Ribozymes such as nuclease-resistant ribozymes (Maccjak, D.J. et al.,
  • An immune modulating agent such as an IMPDH inhibitor, mycophenolic acid, a salt or a prodrug thereof sodium mycophenolate or mycophenolate mofetil, or Merimebodib
  • VX-497 thymosin alpha- 1 (Zadaxin, by SciClone); or a SlP receptor agonist, e.g. FTY720 or analogue thereof optionally phosphorylated.
  • An anti-fibrotic agent such as a N-phenyl-2-pyrimidine- amine derivative, imatinib (Gleevac), IP-501 by Indevus, and Interferon gamma Ib from InterMune (18) Therapeutic vaccine by Intercell, Epimmune/Genecor, Merix, Tripep (Chron-
  • miscellaneous compounds including 1-amino-alkylcyclohexanes (U.S. Patent No. 6,034,134 to Gold et al.), alkyl lipids (U.S. Pat. No. 5,922,757 to Chojkier et al.), vitamin E and other antioxidants (U.S. Patent. No. 5,922,757 to Chojkier et al.), amantadine, bile acids (U.S. Pat. No. 5,846,99964 to Ozeki et al.), N-(phosphonoacetl)-L-aspartic acid, )U.S. Pat. No.
  • Methods of this invention may also involve administration of another component comprising an additional agent selected from an immunomodulatory agent; an antiviral agent; an inhibitor of HCV protease; an inhibitor of another target in the HCV life cycle; a CYP inhibitor; or combinations thereof.
  • an additional agent selected from an immunomodulatory agent; an antiviral agent; an inhibitor of HCV protease; an inhibitor of another target in the HCV life cycle; a CYP inhibitor; or combinations thereof.
  • this invention provides a method comprising administering a compound of the invention and another anti-viral agent, preferably an anti- HCV agent.
  • anti-viral agents include, but are not limited to, immunomodulatory agents, such as a, ⁇ , and ⁇ interferons, pegylated derivatized interferon-a compounds, and thymosin; other anti-viral agents, such as ribavirin, amantadine, and telbivudine; other inhibitors of hepatitis C proteases (NS2-NS3 inhibitors and NS3-NS4A inhibitors); inhibitors of other targets in the HCV life cycle, including helicase, polymerase, and metalloprotease inhibitors; inhibitors of internal ribosome entry; broad-spectrum viral inhibitors, such as IMPDH inhibitors (e.g., compounds of United States Patent 5,807, 876,6, 498,178, 6,344, 465,6, 054,472, WO 97/
  • a pharmaceutical combination comprising a) a first agent which is a compound of the invention, e.g. a compound of formula I or any subformulae thereof, and b) a co- agent, e.g. a second drug agent as defined above.
  • a method as defined above comprising co-administration, e.g. concomitantly or in sequence, of a therapeutically effective amount of a compound of the invention, e.g. a compound of formula I or any subformulae thereof, and a co-agent, e.g. a second drug agent as defined above.
  • co-administration or “combined administration” or the like as utilized herein are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time. Fixed combinations are also within the scope of the present invention.
  • the administration of a pharmaceutical combination of the invention results in a beneficial effect, e.g. a synergistic therapeutic effect, compared to a monotherapy applying only one of its pharmaceutically active ingredients.
  • Each component of a combination according to this invention may be administered separately, together, or in any combination thereof.
  • dosages of interferon are typically measured in IU (e.g., about 4 million IU to about 12 million IU).
  • each component may be administered in one or more dosage forms.
  • Each dosage form may be administered to the patient in any order.
  • the compound of the invention and any additional agent may be formulated in separate dosage forms.
  • the compound of the invention and any additional agent may be formulated together in any combination.
  • the compound of the invention inhibitor may be formulated in one dosage form and the additional agent may be formulated together in another dosage form. Any separate dosage forms may be administered at the same time or different times.
  • composition of this invention comprises an additional agent as described herein.
  • Each component may be present in individual compositions, combination compositions, or in a single composition.
  • the invention is further illustrated by the following examples, which should not be construed as further limiting.
  • the assays used throughout the Examples are accepted. Demonstration of efficacy in these assays is predictive of efficacy in subjects.
  • Nucleosil Nucleosil®, trademark of Machery & Nagel, Duren, FRG for
  • TLC conditions Rf values for TLC are measured on 5 x 10 cm TLC plates, silica gel F254, Merck, Darmstadt, Germany.
  • API-ES positive/negative
  • linker refers to the L 1 -FG-L 2 -L 3 residue of Formula I
  • Pi refers to the Ri residue of Formula I
  • P 2 subst refers to the R 5 residue of Formula I.
  • linker refers to the Li-FG-L 2 -L 3 residue of Formula I
  • Pi refers to the R] residue of Formula I
  • P 2 subst refers to the R 5 residue of Formula I.
  • Step 3 8- ⁇ 2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)-sulfamoyll-phenylcarbamoyl ⁇ - octanoic acid methyl ester
  • Step 2 (ZS ⁇ R ⁇ -Fluoro-l ⁇ -dihydro-isoindole-l-carbonyloxy ⁇ pyrrolidine-l ⁇ -dicarboxylic acid l-tert-butyl ester
  • Step l 4-Fluoro-l,3-dihydro-isoindo-e-2-carboxy-ic ac j ⁇ j (3R,5S)-l-tert-butoxycarbonyl-5- ⁇ (lR,2S)-l-[2-(8-methoxycarbonyl-octanoylamino)-benzenesulfonylaminocarbonyl]-2- vinyl-cyclopropyIcarbamoylJ-pyrroIidin-3-yl ester
  • step 4 using 200 mg (0.14 mmol) 8- ⁇ 2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl] -phenyl carbamoylj-octanoic acid methyl ester (HCl-salt), 113 mg (0.29 mmol) (2S,4R)-4-(4-Fluoro- l,3-dihydro-isoindole-2-carbonyloxy)-pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester,
  • the title compound is prepared analogously as described for the title compound in Example 2 using 65 mg (0.09 mmol) 8-[2-( ⁇ (lR,2S)-l-[(2-Amino-l,2,3,4-tetrahydro-naphthalene-2- carbonyl)-amino]-2-vinyl-cyclopropanecarbonyl ⁇ -sulfamoyl)-phenylcarbamoyl]-octanoic acid (TFA-salt), 114 mg (0.88 mmol) DIPEA and 167 g (0.44 mmol) HATU.
  • the title compound is prepared analogously as described for the title compound in Example 2 using 58 mg (0.08 mmol) 8-[2-( ⁇ (lR,2S)-l-[2-(Cyclopentylmethyl-amino)-acetylamino]-2- vinyl-cyclopropane-carbonyl ⁇ -sulfamoyl)-phenylcarbamoyl]-octanoic acid (TFA-salt), 106 mg (0.82 mmol) DIPEA and 156 mg (0.41 mmol) HATU in 51 mL DCM/MeOH (50:1).
  • step 4 using 150 mg (0.19 mmol) 8- ⁇ 2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl] -phenyl-carbamoyl ⁇ -octanoic acid methyl ester (HCl-salt), 57 mg (0.22 mmol) (tert-Butoxycarbonyl-cyclopentylmethyl-amino)-acetic acid, 84 mg (0.22 mmol) HBTU and 120 mg (0.93 mmol) DIPEA in 2 mL DMF.
  • Step 3 using 150 mg (0.16 mmol) (2S,4R)-2- ⁇ (lR,2S)-l-[2-(9-Carboxy-nonanoylamino)- benzene-sulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl ⁇ -4-(7-methoxy-2-phenyl- quinolin-4-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester and 1 mL TFA in 5 mL
  • step 1 using 150 mg (0.22 mmol) 9- ⁇ 2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyi)- sulfamoyl]-phenylcarbamoyl ⁇ -nonanoic acid methyl ester, 117 mg (0.22 mmol) (2S,4R)-4-
  • step 3 using 50 mg (0.051 mmol) (2S,4R)-2- ⁇ (lR,2S)-l-[2-(9-Carboxy-nonanoylamino)- benzenesulfonyl-aminocarbonyl]-2-vinyl-cyclopropylcarbamoyl ⁇ -4-[2-(2-isopropylamino- thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-l-carboxylic acid tert-butyl ester and
  • Step 2 using 0.76 g (1.99 mmol) [(lR,2S)-l-(2-Amino-benzenesulfonylaminocarbonyl)-2- vinyl-cyclopropyl]-carbamic acid tert-butyl ester, 0.94 g (4.97 mmol) Monomethyl suberate,
  • step 2 using 73 mg (0.08 mmol) (2S,4R)-2- ⁇ (lR,2S)-l-[2-(8-Methoxycarbonyl- octylamino)-benzenesulfonyl-aminocarbonyl]-2-vinyl-cyclopropylcarbamoyl ⁇ -4-(7-methoxy-
  • step 3 using 71 mg (0.08 mmol) (2S,4R)-2- ⁇ (lR,2S)-l-[2-(8-Carboxy-octylamino)- benzenesulfonyl-amino-carbonyl]-2-vinyl-cyclopropylcarbamoyl ⁇ -4-(7-methoxy-2-phenyl- quinolin-4-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester and 0.3 mL TFA in 5 mL
  • Example 12 (8S,10R)-10-[2-(2-IsopropyIamino-thiazol-4-yl)-7-methoxy-quinoIin-4-yloxy]-5-[(lR,2S)- l-carbonylamino-2-vinyl-cyclopropyl]-2,2-dioxo-2 ⁇ *6*-thia-3,6,12,22-tetraaza- tricyclo[21.4.0.0*8,12*]heptacosa-l(27),23,25-triene-4,7,13-trio ⁇ e
  • step 3 using 135 mg (0.14 mmol) (2S,4R)-2- ⁇ (lR,2S)-l-[2-(8-Carboxy-octylamino)- benzenesulfonylamino-carbonyl]-2-vinyl-cyclopropylcarbamoyl ⁇ -4-[2-(2-isopropylamino- thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-l-carboxylic acid tert-butyl ester and
  • a microwave-vial is charged with 2.2 g (10 mmol) 3-Bromophenylacetic acid, 2.62 g (30 mmol) Methyl acrylate, 0.31 g (1.0 mmol) P(o-tol) 3 , 90 mg (0.4 mmol) Pd(OAc) 2 , amd 1.2 g (12 mmol) NEt 3 .
  • the vial is purged with argon, sealed and heated in the microwave (Personal Chemistry, Emrys Optimizer) for 15 min at 150°C. After cooling to rt the mixture is diluted with water and EtOAc, filtered through a pad of Hyflo and washed thoroughly with EtOAc.
  • Step 4 3-[3-( ⁇ 2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)-sulfainoyl]- phenylcarbamoyl ⁇ -methyl)-phenyl]-propionic acid methyl ester
  • the title compound is prepared analogously as described for the title compound in Example 1 (step 4) using 114 mg (0.59 mmol) 3-[3-( ⁇ 2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylcarbamoyl ⁇ -methyl)-phenyl] -propionic acid methyl ester, 73 mg (0.16 mmol) (2S,4R)-4-(7-Methoxy-2-phenyl-quinolin-4-yloxy)-pyrrolidine-l,2- dicarboxylic acid 1-tert-butyl esterr, 90 mg (0.24 mmol) HATU and 102 mg (0.79 mmol)
  • step 3 using 26 mg (0.03 mmol) (2S,4R)-2-[(lR,2S)-l-(2- ⁇ 2-[3-(2-Carboxy-ethyl)-phenyl]- acetylamino ⁇ -benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-4-(7- methoxy-2-phenyl-quinolin-4-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester and 1 mL
  • the title compound is prepared analogously as described for the title compound in Example 2 using 20 mg (0.02 mmol) 3- ⁇ 2-[2-(2- ⁇ [(lR,2S)-l-( ⁇ (2S,4R)-4-[2-(2-Isopropylamino-thiazol- 4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-2-carbonyl ⁇ -amino)-2-vinyl- cyclopropanecarbonyl]-sulfamoyl ⁇ -phenylamino)-ethoxy]-ethoxy ⁇ -propionic acid (TFA- salt), 22 mg (0.20 mmol) DIPEA and 32 mg (0.09 mmol) HATU in 10 mL DCM and 0.2 mL DMF.
  • step 1 using 235 mg (0.34 mmol) 3-[2-(2- ⁇ 2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylamino ⁇ -ethoxy)-ethoxy]-propionic acid methyl ester, 182 mg (0.34 mmol) (2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy- quinolin-4-yloxy]-pyrrolidine-l,2-dicarboxylic acid 1 -tert-butyl ester, 196 mg (0.52 mmol)
  • step 2 using 170 mg (0.18 mmol) (2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy- quinolin-4-yloxy]-2-[( 1 R,2S)- 1 -(2- ⁇ 2-[2-(2-methoxycarbonyl-ethoxy)-ethoxy]-ethylamino ⁇ - benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-pyrrolidine-l-carboxylic acid tert-butyl ester (TFA-salt) and 76 mg (1.8 mmol) LiOH in 20 mL THF/MeOH/H 2 O
  • Step 2 3-[2-( ⁇ 2-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyI-cyclopropanecarbonyl)- sulfamoyl]-phenylcarbamoyl ⁇ -methoxy)-ethoxy]-propionic acid methyl ester
  • Step 2 using 100 mg (0.26 mmol) [(lR,2S)-l-(2-Amino-benzenesulfonylaminocarbonyl)-2- vinyl-cyclopropyl]-carbamic acid tert-butyl ester, 200 mg (0.97 mmol) 3-(2-
  • Step 3 3-[2-( ⁇ 2-[((lR,2S)-l-Amino-2-vinyI-cyclopropanecarbonyl)-sulfamoyl]- phenylcarbamoyl ⁇ -methoxy)-ethoxy]-propionic acid methyl ester
  • the title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 110 mg (0.10 mmol) (2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy- quinolin-4-yloxy]-2-[( 1R,2S)- 1 -(2- ⁇ 2-[2-(2-methoxycarbonyl-ethoxy)-ethoxy]-acetylamino ⁇ - benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-pyrrolidine-l-carboxylic acid tert-butyl ester (TFA-salt) and 43 mg (1.0 mmol) LiOH in 16 mL THF/MeOH/H 2 O
  • Step 6 3- ⁇ 2-[(2- ⁇ [(lR,2S)-l-( ⁇ (2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy-quinolin- 4-yloxy]-pyrrolidine-2-carbonyl ⁇ -amino)-2-vinyI-cyclopropanecarbonyl]-sulfamoyI ⁇ - phenylcarbamoyl)-methoxy]-ethoxy ⁇ -propionic acid
  • the title compound is prepared analogously as described for the title compound in Example 2 using 56 mg (0.05 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-5- [(lR,2S)-l-(2- ⁇ 2-[2-(2-carboxy-ethoxy)-ethoxy]-ethylamino ⁇ - benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-pyrrolidin-3-yl ester (TFA- salt), 70 mg (0.54 mmol) DIPEA and 103 mg (0.27 mmol) HATU in 50 mL DCM and 1 mL DMF.
  • step 1 using 235 mg (0.34 mmol) 3-[2-(2- ⁇ 2-[((lR,2S)-l-Am ⁇ no-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylamino ⁇ -ethoxy)-ethoxy]-propionic acid methyl ester, 136 mg (0.34 mmol) ((2S,4R)-4-(4-Fluoro-l,3-dihydro-isoindole-2-carbonyloxy)- pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester, 196 mg (0.52 mmol) HATU and 134 mg

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Virology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oncology (AREA)
  • Engineering & Computer Science (AREA)
  • Communicable Diseases (AREA)
  • Immunology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present application describes macrocyclic compounds of formula (I) with NS3 protease inhibitory activity for treating hepatitis C virus infection.

Description

MACROCYCLIC COMPOUNDS AS HCV NS3 PROTEASE INHIBITORS
Background
Chronic hepatitis C virus (HCV) infection is a major global health burden, with an estimated 170 million people infected worldwide and an additional 3 to 4 million infected each year (See e.g. World Health Organization Fact Sheet No.164. October 2000). Although 25% of new infections are symptomatic, 60-80% of patients will develop chronic liver disease, of whom an estimated 20% will progress to cirrhosis with a 1-4% annual risk of developing hepatocellular carcinoma (See e.g. World Health Organization Guide on Hepatitis C. 2002; Pawlotsky, J-M. (2006) Therapy of Hepatitis C: From Empiricism to Eradication. Hepatology 43:S207-S220). Overall, HCV is responsible for 50-76% of all liver cancer cases and two thirds of all liver transplants in the developed world (See e.g. World Health
Organization Guide on Viral Cancers. 2006). And ultimately, 5-7% of infected patients will die from the consequences of HCV infection (See e.g. World Health Organization Guide on Hepatitis C. 2002).
The current standard therapy for HCV infection is pegylated interferon alpha (IFN-α) in combination with ribavirin. However, only up to 50% of patients with genotype 1 virus can be successfully treated with this interferon-based therapy. Moreover, both interferon and ribavirin can induce significant adverse effects, ranging from flu-like symptoms (fever and fatigue), hematologic complications (leukopenia, thrombocytopenia), neuropsychiatric issues (depression, insomnia, irritability), weight loss, and autoimmune dysfunctions (hypothyroidism, diabetes) from treatment with interferon to significant hemolytic anemia from treatment with ribavirin. Therefore, more effective and better tolerated drugs are still greatly needed.
HCV, first identified in 1989 (See e.g. Choo, Q. L. et al. Science (1989) 244:359- 362), is a single-stranded RNA virus with a 9.6-kilobase genome of positive polarity. It encodes a single polyprotein that is cleaved upon translation by cellular and viral proteases into at least ten individual proteins: C, El, E2, p7, NS2, NS3, NS4A, NS4B, NS5A, and NS5B (See e.g. Lindenbach, B. D. et al. (2001). Flaviviridae: the viruses and their replication, p. 991-1041. In D. M. Knipe, P. M. Howley, and D. E. Griffin (ed.), Fields virology, 4th ed, vol. 1. Lippincott Williams & Wilkins, Philadelphia, Pennsylvania). NS3, an approximately 70 kDa protein, has two distinct domains: a N-terminal serine protease domain of 180 amino acids (AA) and a C-terminal helicase/NTPase domain (AA 181 to 631). The NS3 protease is considered a member of the chymotrypsin family because of similarities in protein sequence, overall three-dimensional structure and mechanism of catalysis. The HCV NS3 serine protease is responsible for proteolytic cleavage of the polyprotein at the NS3/NS4A, NS4A/NS4B, NS4B/NS5A and NS5A/NS5B junctions (See e.g. Bartenschlager, R., L. et al. (1993) J. Virol. 67:3835-3844; Grakoui, A. et al. (1993) J. Virol. 67:2832-2843; Tomei, L. et al. (1993) J. Virol. 67:4017-4026). NS4A, an approximately 6 kDa protein of 54 AA, is a co-factor for the serine protease activity of NS3 (See e.g. Failla, C. et al. (1994) J. Virol. 68:3753-3760; Tanji, Y. et al. (1995) J. Virol. 69:1575-1581). Autocleavage of the NS3/NS4A junction by the NS3/NS4A serine protease occurs intramolecularly (i.e , cis) while the other cleavage sites are processed intermolecularly (i.e., trans). It has been demonstrated that HCV NS3 protease is essential for viral replication and thus represents an attractive target for antiviral chemotherapy.
Summary of the Invention
There remains a need for new treatments and therapies for HCV infection, as well as HCV-associated disorders. There is also a need for compounds useful in the treatment or prevention or amelioration of one or more symptoms of HCV, as well as a need for methods of treatment or prevention or amelioration of one or more symptoms of HCV. Furthermore, ' there is a need for methods for modulating the activity of HCV-serine proteases, particularly the HCV NS3/NS4a serine protease, using the compounds provided herein.
In one aspect, the invention provides compounds of the Formula I:
Figure imgf000003_0001
and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof; wherein the macrocycle:
Figure imgf000004_0001
comprises between 15 to 40 ring atoms; m, x and z are each independently selected from 0 or 1 ; p is selected at each occurrence from the group consisting of 0, 1 and 2; Ri and R2 are independently selected, at each occurrence, from hydrogen or cyano, or from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, alkoxy, and cycloalkyloxy, each of which is unsubstituted or substituted with 1-6 moieties which can be the same or different and are independently selected from the group consisting of hydroxy, oxo, alkyl, aryl, alkoxy, aryloxy, thio, alkylthio, arylthio, amino, alkylamino, arylamino, alkylsulfonyl, arylsulfonyl, alkylsulfonamido, arylsulfonamido, heteroarylsulfonamido, ! arylamiήosulfonyl, heteroarylaminosulfonyl, mono and dialkylaminosulfonyl, carboxy, carbalkoxy, amido, carboxamido, alkoxycarbonylamino, aminocarbonyloxy, alkoxycarbonyloxy, alkylureido, arylureido, halogen, cyano, or nitro; wherein each of said alkyl, alkoxy, and aryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different and are independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkyl-alkyl, heterocyclyl, heterocyclylalkyl, aryl, alkylaryl, aralkyl, arylheteroaryl, heteroaryl, heterocyclylamino, alkylheteroaryl and heteroaralkyl;
R3 is selected from the group consisting of H and Ci-4-alkyl; E is a divalent residue selected from the group consisting OfC(O)NR23, NR23S(COp, NR23S(COpNR23;
Li and L2 are divalent residues independently selected from the group consisting of C0-4alkylene, (CH2),-FG-(CH2)k, (CH2)rC3-7cycloalkylene-(CH2)k, (CH2),-C3- 7cycloheteroalkylene-(CH2)k, alkenylene, alkynylene, arylene, heteroarylene, cycloalkylene and heterocycloalkylene, each of which is substituted with 0 to 4 independently selected Xi or X2 groups; i and k are independently selected integers of from 0 to 7; L3 is a Co-4alkylene or a divalent ethylene or acetylene residue, wherein the C0- 4alkylene and divalent ethylene residues are substituted by 0-2 substituents selected from alkyl, aryl, heteroaryl, mono- or di-alkylamino-Co-Cόalkyl, hydroxyl alkyl or alkoxyalkyl;
FG is absent or a divalent residue selected from the group consisting of O, S(O)P, NR23, C(O), C(O)NR23, NR23C(O), OC(O)NR23, NR23C(O)O, NR23C(O)NR23, S(O)PNR23, NR23S(O)p, and NR23S(O)PNR23; R23 is independently selected at each occurrence from hydrogen or the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, aryl, heteroaryl, heteroaralkyl, aralkyl and heteroaralkyl, each of which is substituted with 0-2 substituents independently selected from halogen, alkyl, alkoxy, and mono- and di-alkylamino; or
Two R23 residues, taken in combination, form a monocyclic, bicyclic or tricyclic heterocyclic ring system which is saturated, partially unsaturated, or aromatic, and which is substituted with O to 3 substituents independently selected from Ci-6alkyl,
Figure imgf000005_0001
Ci- δalkoxyCi-όalkoxy, mono- and di-Ci-ealkylaminoCi-όalkoxy, Ci-6haloalkyl, Ci-6haloalkoxy, mono- and di-Ci^alkylamino, halogen, 4 to 7 member heterocycloalkyl, aryl, heteroaryl, and 3 to 6 member spirocycloalkyl or spiroheterocycloalkyl, each of which is substituted with O to 3 substituents independently selected from the group consisting of C^alkyl, Q^alkoxy, hydroxy, amino, and mono- and di-Ci^alkylamino;
R9 is absent or selected from hydrogen, Q^alkyl, C3-7cycloalkyl-Co-4alkyl, or hydroxy;
R7, Rio, Rn, Ri2, Ri3, R15, Ri6, R17, and R22 are each, independently, hydrogen or selected from the group consisting of alkyl, alkenyl, alkynyl, aryl, alkyl-aryl, heteroalkyl, heterocyclyl, heteroaryl, aryl-heteroaryl, alkyl-heteroaryl, cycloalkyl, alkyloxy, alkyl- aryloxy, aryloxy, heteroaryloxy, heterocyclyloxy, cycloalkyloxy, amino, alkylamino, arylamino, alkyl-arylamino, arylamino, heteroarylamino, cycloalkylamino, carboxyalkylamino, aralkyloxy and heterocyclylamino; each of which may be further substituted 0 to 5 times with substituents independently selected from Xi and X2;
Xi is alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkyl-alkyl, heterocyclyl, heterocyclylalkyl, aryl, alkylaryl, aralkyl, arylheteroaryl, heteroaryl, heterocyclylamino, alkylheteroaryl, or heteroaralkyl; wherein Xi can be independently substituted with one or more of X2 moieties which can be the same or different and are independently selected; X2 is hydroxy, oxo, alkyl, aryl, heteroaryl, alkoxy, aryloxy, heteroaryloxy, thio, alkylthio, arylthio, heteroarylthio, amino, alkylamino, arylamino, heteroarylamino, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, alkylsulfonamido, arylsulfonamido, heteroarylsulfonamido, arylaminosulfonyl, heteroarylaminosulfonyl, mono and dialkylaminosulfonyl, carboxy, carbalkoxy, amido, carboxamido, alkoxycarbonylamino, aminocarbonyloxy, alkoxycarbonyloxy, carbamoyl, ureido, alkylureido, arylureido, halogen, cyano, or nitro; wherein each of said alkyl, alkoxy, and aryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different and are independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkyl- alkyl, heterocyclyl, heterocyclylalkyl, aryl, alkylaryl, aralkyl, arylheteroaryl, heteroaryl, heterocyclylamino, alkylheteroaryl and heteroaralkyl; Zi is Co-4alkylene, oxygen or NRi0; Z2 is CR9, O or N;
Figure imgf000006_0001
V is selected from hydrogen or from the group consisting of alkyl, alkyl-aryl, heteroalkyl, heterocyclyl, heteroaryl, aryl-heteroaryl, alkyl-heteroaryl, cycloalkyl, alkyloxy, alkyl-aryloxy, aryloxy, heteroaryloxy, heterocyclyloxy, cycloalkyloxy, amino, alkylamino, arylamino, alkyl-arylamino, arylamino, heteroarylamino, cycloalkylamino, carboxyalkylamino, mono- and di-alkylcarboxamide, aralkyloxy and heterocyclylamino; each of which may be further independently substituted one or more times with X1 and X2; wherein X1 is alkyl, alkenyl, alkynyl, cycloalkyl, .cycloalkyl-alkyl, heterocyclyl, heterocyclylalkyl, aryl, alkylaryl, aralkyl, aryloxy, arylthio, arylheteroaryl, heteroaryl, heterocyclylamino, alkylheteroaryl, or heteroaralkyl; wherein X1 can be independently substituted with one or more X2 moieties which can be the same or different and are independently selected; wherein X2 is hydroxy, oxo, alkyl, cycloalkyl, spirocycloalkyl, heterocycloalkyl, aryl, heteroaryl, alkoxy, aryloxy, thio, alkylthio, amino, mono- and di- alkylamino, arylamino, alkylsulfonyl, arylsulfonyl, alkylsulfonamido, arylsulfonamido, carboxy, carbalkoxy, carboxamido, alkoxycarbonylamino, alkoxycarbonyl, alkoxycarbonyloxy, alkylureido, arylureido, halogen, cyano, or nitro; wherein each X2 residue selected to be alkyl, alkoxy, and aryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different and are independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkyl-alkyl, heterocyclyl, heterocyclylalkyl, aryl, alkylaryl, aralkyl, arylheteroaryl, heteroaryl, heterocyclylamino, alkylheteroaryl and heteroaralkyl; or V is selected from the group consisting of-Q'-Q2, wherein Q1 is absent, C(O),
S(O)2, N(H), N(Ci-4-alkyl), C=N(CN), C=N(SO2CH3), C=N-COH-C M-alkyl, or C=N-COH, and Q2 is hydrogen or is selected from the group consisting of Ci^-alkyl, O-Ci-4-alkyl, NH2,
Figure imgf000006_0002
SO2-aryl, SO2-heteroaryl, SO2-C i-4-alkyl, C3-6-cycloalkyl-C0- 4-alkyl, aryl, heteroaryl and heterocycle, each of which may be independently substituted one or more times with a halogen atom, Ci-4-alkyl, Ci-4-alkyl substituted by one or more halogen atoms, or C3-6-cycloalkyl; or R22 and R16 may together form a 3, 4, 5, 6 or 7-membered ring and may contain one or more heteroatoms, wherein the ring may be further substituted one or more times; or R7 and Ri 5 may together form a 3, 4, 5, 6 or 7-membered ring and may contain one or more heteroatoms, wherein the ring may be further substituted one or more times; or Ri 5 and Ri7 may together form a 3, 4, 5, 6 or 7-membered ring and may contain one or more heteroatoms, wherein the ring may be further substituted one or more times; or R]5 and Ri6 may together form a 4, 5, 6 or 7-membered ring and may contain one or more heteroatoms, wherein the ring may be further substituted one or more times; or Ri5 and Ri6 may together form an arylene or heteroarylene ring and R7 and R22 are absent, wherein the ring may be further substituted one or more times; or Ri and R2 may together form a 3, 4, 5, 6 or 7-membered ring that is saturated or partially unsaturated and may contain one or more heteroatoms, which ring is substituted with 0-3 residues independently selected from C^alkyl, Ci^alkoxy, C2-4alkenyl, C2-4alkynyl, halogen, hydroxy, C3-6cylcoalkyl and C3-6spirocycloalkyl; or Rn and Ri6 may together form a 4, 5,16, 7 or 8-membered ring of the formula:
Figure imgf000007_0001
wherein n and g are each, independently, 0, 1 or 2;
X is O, S, N, C or CR5a;
R4 is hydrogen or is selected from the group consisting of Ci-6-alkyl, C3-7-cycloalkyl, aryl, heterocycle and heteroaryl, all of which may be independently substituted one or more times with a halogen atom or Ci-4-alkyl; R5 is absent, hydrogen or oxo or is selected from the group consisting of hydroxyl, Ci-
8-alkyl, C2-8-alkenyl, C2-8-alkynyl, C3.8-cycloalkyl-C<M-alkyl,
Figure imgf000007_0002
heterocycle-Co- 4-alkyl, heteroaryl-Co-4-alkyl , C3-8-cycloalkyloxy, aryloxy, NR23COR23, CONR23R23, NR23CONHR23, OCONR23R23, NR23COOR23, OCOR23, COOR23, aryl-C(O)O, aryl- C(O)NR23, heteroaryloxy, heteroaryl-C(O)O, heterocycle-C(O)O, heteroaryl-C(O)NR23, heterocycle-C(O)NR23, each of which may be independently substituted one or more times (or more preferably 0, 1, 2, 3, 4, or 5 times) with halogen,
Figure imgf000007_0003
Ci-4-alkoxy, 1IaIoCi-4- alkyl, haloCi-4-alkoxy, amino, mono- and di-CMalkylaminoCo^alkyl, mono- and di-Cj. 4alkylaminoCo-4alkoxy, C3-7cycloalkyl, fused- or spiro-cyclic 3-7 membered ring, heterocycleC0-4alkoxy, heterocycleCo^alkyl, aryl, or heteroaryl;
R5a is selected from the group consisting of H, hydroxyl, Ci-8-alkyl, C2-8-alkenyl, C2-8- alkynyl, C3-8-cycloalkyl-C0-4-alkyl, aryl-C0-4-alkyl and heteroaryl-Co^-alkyl, or R4 and R5 may together form a fused dimethyl cyclopropyl ring, a fused cyclopentane ring, a fused phenyl ring or a fused pyridyl ring, each of which may be substituted with a halogen atom, aryl, heteroaryl, trihalomethyl, Ci-4-alkoxy or C1-4-alkyl; or R5 and R53 may together form a spirocyclic ring having between 3 and 7 ring atoms and having 0, 1, or 2 ring heteroatoms, which is optionally substituted by 0-4 substitutents selected from cyano, halogen, hydroxyl, amino, thiol, Ci-8-alkyl, C2.8-alkenyl, C2-8-alkynyl, Ci-s-alkoxy-Co^alkyl, Ci-g-haloalkyl, C2-8-haloalkenyl, C2-8-haloalkynyl, Ci-8-haloalkoxy, C1- 8-alkylthio, Q.g-alkylsulfonyl, Ci-8-alkylsulfoxy, Ci-8-alkanoyl, C1-8-alkoxycarbonyl, C3-7- cycloalkyl-C0-4-alkyl, aryl-Co-4-alkyl, heteroaryl-Co-4-alkyl, COOH, C(O)NH2, mono- and di- Ci-4-alkyl-carboxamide, mono- and di-C1-4-alkyl-amino-Co-4alkyl, SO3H, SO2NH2, and mono-and di-Ci-4-alkylsulfonamide, or two, substitutents taken together form a fused or spirocyclic 3 to 7 membered ring having ,0, 1 or 2 ring heteroatoms selected from N, O and S, which fused or spirocyclic ring has 0 to 2 independently selected substitutents selected from cyano, halogen, hydroxyl, amino, thiol, Ci-8-alkyl, C2-8-alkenyl, C2-8-alkynyl, Ci-8-alkoxy-Co- 4alkyl, C1-8-haloalkyl, C2-8-haloalkenyl, C2-8-haloalkynyl, Ci-8-haloalkoxy, d-g-alkylthio, Ci- 8-alkylsulfonyl, Ci-8-alkylsulfoxy, Ci-8-alkanoyl, Ci-s-alkoxycarbonyl, C3-7-C ycloalkyl-Co^- alkyl, aryl-C0-4-alkyl, heteroaryl-C0-4-alkyl, COOH, C(O)NH2, mono- and di-C1-4-alkyl- carboxamide, mono- and
Figure imgf000008_0001
SO3H, SO2NH2, and mono-and di-Ci. 4-alkylsulfonamide; and R6 is independently selected at each occurrence from the group consisting of hydrogen, hydroxy, amino,
Figure imgf000008_0002
and C3-
Figure imgf000008_0003
or two R6 residues may together form a spirocyclic ring having between 3 and 7 ring atoms and having O, 1 , or 2 ring heteroatoms, which is optionally substituted by 0-4 substitutents selected from cyano, halogen, hydroxyl, amino, thiol, Ci-8-alkyl, C2-8-alkenyl, C2-8-alkynyl, Ci-8-alkoxy-C0-4alkyl, Ci-g-haloalkyl, C2-8-haloalkenyl, C2-8-haloalkynyl, Ci-8- haloalkoxy, Ci-8-alkylthio, Ci-8-alkylsulfonyl, Q-g-alkylsulfoxy, Ci-8-alkanoyl, Ci-8- alkoxycarbonyl, C3-7-cycloalkyl-Co-4-alkyl, aryl-C0-4-alkyl, heteroaryl-C0-4-alkyl, COOH, C(O)NH2, mono- and di-CM-alkyl-carboxamide, mono- and di-Ci-4-alkyl-amino-Co^alkyl, SO3H, SO2NH2, and mono-and
Figure imgf000009_0001
or two substitutents taken together form a fused or spirocyclic 3 to 7 membered ring having 0, 1 or 2 ring heteroatoms selected from N, O and S, which fused or spirocyclic ring has 0 to 2 independently selected substitutents selected from halogen,
Figure imgf000009_0003
mono- and
Figure imgf000009_0002
alkylamino, mono- and di-C^-alkyl-carboxamide, Ci-4-alkoxycarbonyl, and phenyl.
In one embodiment, the invention provides a method of treating an HCV-associated disorder comprising administering to a subject in need thereof a pharmaceutically acceptable amount of a compound of the invention, such that the HCV-associated disorder is treated. hi another embodiment, the invention provides a method of treating an HIV infection comprising administering to a subject in need thereof a pharmaceutically acceptable amount of a compound of the invention.
In still another embodiment, the invention provides a method of treating, inhibiting or preventing the activity of HCV in a subject in need thereof, comprising administering to the subject a pharmaceutically acceptable amount of a compound of the invention. In one embodiment, the compounds of the invention inhibit the activity of the NS2 protease, the NS3 protease, the NS3 helicase, the NS5a protein, and/or the NS5b polymerase. In another embodiment, the interaction between the NS3 protease and NS4A cofactor is disrupted. In yet another embodiment, the compounds of the invention prevent or alter the severing of one or more of the NS4A-NS4B, NS4B-NS5A and NS5A-NS5B junctions of the HCV. In another embodiment, the invention provides a method of inhibiting the activity of a serine protease, comprising the step of contacting said serine protease with a compound of the invention, hi another embodiment, the invention provides a method of treating, inhibiting or preventing the activity of HCV in a subject in need thereof, comprising administering to the subject a pharmaceutically acceptable amount of a compound of the invention, wherein the compound interacts with any target in the HCV life cycle. In one embodiment, the target of the HCV life cycle is selected from the group consisting of NS2 protease, NS3 protease, NS3 helicase, NS5a protein andNS5b polymerase.
In another embodiment, the invention provides a method of decreasing the HCV RNA load in a subject in need thereof comprising administering to the subject a pharmaceutically acceptable amount of a compound of the invention.
In another embodiment, the compounds of the invention exhibit HCV protease activity. In one embodiment, the compounds are an HCV NS3-4A protease inhibitor. In another embodiment, the invention provides a method of treating an HCV- associated disorder in a subject, comprising administering to a subject in need thereof a pharmaceutically acceptable amount of a compound of the invention, and a pharmaceutically acceptable carrier, such that the HCV-associated disorder is treated.
In another embodiment, the invention provides a method of treating an HCV- associated disorder in a subject wherein the subject is suffering from or susceptible to a viral infection which is resistant to one or more anti-viral therapies, the method comprising administering to a subject in need thereof a pharmaceutically acceptable amount of a compound of the invention, and a pharmaceutically acceptable carrier, such that the drug- resistant HCV-associated disorder is treated.
In still another embodiment, the invention provides a method of treating an HCV- associated disorder comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of the invention, in combination with a pharmaceutically effective amount of an additional HCV-modulating compound, such as interferon or derivatized interferon, or a cytochrome P450 monooxygenase inhibitor, such that the HCV- associated disorder is treated. In one embodiment, the additional HCV-modulating compound is selected from the group consisting of ITMN191, Sch 503034 and VX-950. , In another embodiment, the invention provides a method of inhibiting hepatitis C
.virus replication in a cell, comprising contacting said cell with a compound of the invention. i In yet another embodiment, the invention provides a packaged HCV-associated disorder treatment, comprising an HCV-modulating compound of the invention, packaged with instructions for using an effective amount of the HCV-modulating compound to treat an HCV-associated disorder.
In certain embodiments, the HCV-associated disorder is selected from the group consisting of HCV infection, liver cirrhosis, chronic liver disease, hepatocellular carcinoma, cryoglobulinaemia, non-Hodgkin's lymphoma, and a suppressed innate intracellular immune response.
In another embodiment, the invention provides a method of treating HCV infection, liver cirrhosis, chronic liver disease, hepatocellular carcinoma, cryoglobulinaemia, non- Hodgkin's lymphoma, and/or a suppressed innate intracellular immune response in subject in need thereof comprising administering to the subject a pharmaceutically acceptable amount of a compound of the invention.
In one embodiment, the HCV to be treated is selected of any HCV genotype. In another embodiment, the HCV is selected from HCV genotype 1, 2 and/or 3.
Detailed Description of the Invention This invention is directed to compounds, e.g., peptide compounds, and intermediates thereto, as well as pharmaceutical compositions containing the compounds for use in treatment of HCV infection. This invention is also directed to the compounds of the invention or compositions thereof as protease inhibitors, particularly as serine protease inhibitors, and more particularly as HCV NS3 protease inhibitors. The compounds are particularly useful in interfering with the life cycle of the hepatitis C virus and in treating or preventing an HCV infection or physiological conditions associated therewith. The present invention is also directed to methods of combination therapy for inhibiting HCV replication in cells, or for treating or preventing an HCV infection in patients using the compounds of the invention or pharmaceutical compositions, or kits thereof. In one aspect, the compounds of the invention are compounds of Formula I, in which
Ri and R2 taken in combination form a 3, 4, 5, or 6-membered saturated carbocyclic ring which is substituted with 0-2 substituents independently selected from halogen, alkyl, alkenyl, alkoxy and C3-6cycloalkyl. In other aspects, compounds of the invention are compounds of Formula I, in which R| and R2 taken in combination form a cyclopropyl ring. In certain compounds of Formula I include those compounds in which Ri and R2 are taken in combination to form a cyclopropyl ring substituted with 0-2 substituents independently , selected from halogen, alkyl, alkenyl, and alkoxy or substituted with 0 to 2 Ci-C4alkyl residues. Still other compounds of Formula I include those in which Rt and R2 are taken in combination to form a cyclopropyl ring which is substituted with 0 or 1 substituents selected Ci^alkyl, vinyl or cyclopropyl; and E is C(O)NH, NHS(O)2, NHSO2N(Me), NHSO2N(Et) or NHSO2N(cyclopropyl).
In another aspect, the compounds of the invention are compounds of any one of Formulae I, in which Ri is H or Ci-4 alkyl; and R2 is H, Ci-C4alkyl, Ci-C4fluoroalkyl, C2- C4alkenyl, or C3-C7cycloalkylCo-2alkyl. Certain other compounds of Formula I comprise a macrocycle having between 15 and
40 ring atoms, between 15 and 35, 15 and 30 or 15 and 25 ring atoms, or between 17 and 23 ring atoms. Certain compounds of Formula I comprise a macrocycle having 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 ring atoms. In certain instances, compounds of Formula I comprise a macrocycle having 16, 17, 18, 19, 20, 21, 22, or 23 ring atoms. Certain other compounds of Formula I comprise a macrocycle selected from the group consisting of macrocycles of the formulae:
Figure imgf000012_0001
In certain compounds of Formula I, L1 is Ci-C6alkylene, C3-C7cycloalkylene, arylene or heteroarylene, each of which is substituted by 0-4 residues independently selected from Ci-C4alkyl, Ci-C4alkoxy, hydroxyl, amino, mono- and di- Ci-C4alkylamino, halogen, cyano, Ci-C4fluoroalkyl, Ci-C4fluoroalkoxy, COOH, carboxamide (CONH2), mono- and di-Cr C4alkylcarboxamide, aryl, heteroaryl and 5 or 6 membered saturated heterocycles;
L2 is selected from Ci-Cβalkylene and C2-C6alkenylene, each of which is substituted by 0-4 residues independently selected from Ci-C4alkyl, Ci-C4alkoxy, hydroxyl, amino, mono- and di- Ci-C4alkylamino, halogen, cyano, Ci-C4fluoroalkyl, Q^fluoroalkoxy,
COOH, carboxamide (CONH2), mono- and di-Q^alkylcarboxamide, aryl, heteroaryl and 5 or 6 membered saturated heterocycles; and
L3 is absent or a divalent ethylene residue which is substituted by O to 2 independently selected methyl or ethyl residues. In yet other compounds of Formula I, Li is a divalent residue selected from C2-
C4alkylene, 1,2-phenylene, 1,3-phenylene, 2,4-pyridylene, 2,3-pyridylene, 3,4-pyridylene or 1,7-indolylene, 2,7-indolylene, each of which is substituted with 0-3 residues selected from Cj-C4alkyl, Ci-C4alkoxy, hydroxyl, amino, mono- and di- Ci-C4alkylamino, halogen, cyano, Ci-C2fluoroalkyl, Ci-C2fluoroalkoxy, COOH, carboxamide (CONH2), and mono- and di-Ci- C4alkylcarboxamide. hi certain compounds of Formula I, Li is C3-C7cycloalkylene, arylene or heteroarylene which is substituted by 0-4 residues independently selected from Ci-C4alkyl, Ci-C4alkoxy, hydroxyl, amino, mono- and di- Ci-C4alkylamino, halogen, cyano, Ci- C4fluoroalkyl, Ci-Qfluoroalkoxy, COOH, carboxamide (CONH2), mono- and di-Ci- C4alkylcarboxamide, aryl, heteroaryl and 5 or 6 membered saturated heterocycles;
L2 is selected from Ci-Cβalkylene and C2-C6alkenylene, each of which is substituted by 0-4 residues independently selected from Ci-C4alkyl, C]-C4alkoxy, hydroxyl, amino, mono- and di- Ci-C4alkylamino, halogen, cyano, Ci-C4fluoroalkyl, C1-C4HuOrOaIkOXy,
COOH, carboxamide (CONH2), mono- and di-Ci-C4alkylcarboxamide, aryl, heteroaryl and 5 or 6 membered saturated heterocycles; and
L3 is absent or a divalent ethylene residue which is substituted by O to 2 independently selected methyl or ethyl residues. In yet other compounds of Formula I, Li is a divalent residue selected from 1,2- phenylene, 1,3-phenylene, 2,4-pyridylene, 2,3-pyridylene, 3,4-pyridylene or 1,7-indolylene, 2,7-indolylene, each of which is substituted with 0-3 residues selected from Ci-C4alkyl, C1- C4alkoxy, hydroxyl, amino, mono- and di- Ci-C4alkylamino, halogen, cyano, Ci- C2fluoroalkyl, Ci-C2fluoroalkoxy, COOH, carboxamide (CONH2), and mono- and di-Ci- C4alkylcarboxamide.
Certain compounds of Formula I include compounds of Formula II:
Figure imgf000013_0001
II and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof. Yet other compounds of the invention according to Formula II include those compounds in which: x is O or 1 ; n is O or 1 ; Ri4 is C(O) or S(O)P; Zi is absent or NH; Z2 is nitrogen or CH;
R1 is selected from the group consisting of H and Ci-4-alkyl;
R2 is selected from the group consisting of Ci^-alkyl, C(O)C ]-4-alkyl, C(O)OCi-4- alkyl, and (CH2)0-4-C3-6-cycloalkyl; or R 1 and R2 together form a cyclopropane ring;
R3 is selected from the group consisting of H and Ci^-alkyl;
X is O, NR5 or CR5R5a;
R4 is hydrogen or is selected from the group consisting of C^-alkyl, C3-6-cycloalkyl, aryl, heterocycle and heteroaryl, each of which may be independently substituted one or more times with a halogen atom or
Figure imgf000014_0001
R5 is hydrogen or oxo or is selected from the group consisting of hydroxyl, Ci-8-alkyl, C2-8-alkenyl, C2-8-alkynyl, C3-8-cycloalkyl-Co-4-alkyl, aryl-Co-4-alkyl, aryloxy, heteroaryloxy, heterocycle-Co-4-alkyl and heteroaryl-Co^-aUcyl, each of which may be independently substituted one or more times with a halogen atom, aryl, heteroaryl, trihalomethyl,
Figure imgf000014_0002
alkoxy or C 1 ^-alkyl;
R52 is selected from the group consisting of H, hydroxyl, Ci-g-alkyl, C2-g-alkenyl, C2-8- alkynyl, C3-8-cycloalkyl-Co-4-alkyl, aryl-C0-4-alkyl and heteroaryl-Co-4-alkyl, or R4 and R5 may together form a fused dimethyl cyclopropyl ring, a fused cyclopentane ring, a fused phenyl ring or a fused pyridyl ring, each of which may be substituted with a halogen atom, aryl, heteroaryl, trihalomethyl, Ci-4-alkoxy or Ci^-alkyl; or R5 and Rs2 may together form a spirocarbocyclic saturated ring having between 3 and 6 carbon ring atoms which is optionally substituted by 0-2 substitutents selected from halogen, Ci-6-alkyl, C2-6-alkenyl, C2-6-alkynyl, Ci-6-alkoxide, C3-7-cycloalkyl-Co-4-alkyl, phenyl-Co^-alkyl, naphthyl-Co-4-alkyl, heteroaryl-Co^-alkyl, or two substitutents taken together form a fused or spirocyclic 3 to 7 membered carbocyclic ring, each of which is substituted with 0-3 independently selected halogen atoms or Ci-4-alkyl groups;
Rio and Ru are each, independently, selected from the group consisting of H and Ci-4- alkyl;
R6 and R]3 is H; Ri2 is selected from the group consisting of H, Ci-4-alkyl and C3-6-cycloalkyl; and
V is selected from the group consisting of-Q'-Q2, wherein Q1 is absent, C(O), N(H), N(Ci-4-alkyl), C-N(CN), C=N(SO2CH3), or C=N-COH, and Q2 is H, C)-4-alkyl, C=N-COH- CM-alkyl, d-4-alkoxy, C3-7cycloalkyloxy, heterocycloalkyloxy, NH2, N(H)-Ci -4-alkyl, N(Ci- 4-alkyl)2, SO2-aryl, SO2-C i_4-alkyl, C3-6cycloalkyl-Co-4-alkyl, aryl, heteroaryl and heterocycle, each of which may be independently substituted one or more times with a halogen atom, Ci-4- alkyl, C^alkoxy, C2-C4alkenyloxy, C2-C4alkynyloxy, Ci^-alkyl substituted by one or more halogen atoms, or C3-6-cycloalkyl; or when x is 0, Ri0 and V can form a cyclopropyl ring that may be further substituted by an amide group.
Still other compounds of the invention according to Formula II include those compounds in which X is CR5Rs3, R4 is H, and R5 and Rsa taken in combination form a 3 to 6 member spirocyclic carbocycle substituted with 0-2 substitutents selected from halogen, Ci-6- alkyl, C2-6-alkenyl, C2-6-alkynyl, Ci-6-alkoxide, C3-7-C ycloalkyl-C0-4-alkyl, phenyl-C0-4-alkyl, naphthyl-Co-4-alkyl, heteroaryl-Co-4-alkyl, or two substitutents taken together form a fused or spirocyclic 3 to 7 membered carbocyclic ring, each of which is substituted with 0-3 independently selected halogen atoms or Ci^-alkyl groups.
Yet other compounds of the invention according to Formula II include compounds according to Formula Ha:
Figure imgf000015_0001
Ha wherein
Z2 is nitrogen or CH; ki and k2 are 0 or 1 such that a sum of ki and k2 equals 1 or 2; Ra is hydrogen, C^alkyl, or phenyl; Rb is hydrogen, C1.4a.kyl,
Figure imgf000015_0002
mono- and
Figure imgf000015_0003
or phenyl or Ra and Rb taken together form a fused or spirocyclic 3 to 6 membered ring having 0, 1 or 2 ring heteroatoms selected from N, O and S, which fused or spirocyclic ring has 0 to 2 independently selected substitutents selected from halogen, Ci^alkyl, Ci-4alkoxy, Q- 4alkanoyl, and phenyl; and
Rc represents 0 to 4 substitents which are independently selected at each occurrence of R0 from the group consisting of halogen, C1-4alkyl, and phenyl, or two geminal R0 substitents, taken in combination form a 3 to 6 member spirocyclic ring.
Certain compounds of the invention according to Formula Ha include those compounds in which the divalent residue:
Figure imgf000016_0001
is selected from the group consisting of:
Figure imgf000016_0002
Figure imgf000017_0001
Yet other compounds of the invention according to Formula II include those compounds in which: X is CRsR53; and
R5 and R53, taken in combination, form a spirocyclic ring having between 3 and 7 ring atoms and having 0, 1, or 2 ring heteroatoms, which spirocyclic ring is substituted with a spirocyclic 3 to 7 membered ring having 0, 1 or 2 ring heteroatoms selected from N, O and S, and wherein each of the spirocyclic rings has 0 to 2 independently selected substitutents selected from cyano, halogen, hydroxyl, amino, thiol, Ci-8-alkyl, C2-8-alkenyl, C2-8-alkynyl, Ci.8-alkoxy-Co-4alkyl, C|.8-haloalkyl, C2-8-haloalkenyl, C2-8-haloalkynyl, d-g-haloalkoxy, Ci- 8-alkylthio, Ci-8-alkylsulfonyl, Ci-8-alkylsulfoxy, Ci-8-alkanoyl, Ci-8-alkoxycarbonyl, C3-7- cycloalkyl-Co^-alkyl, aryl-C0^-alkyl, heteroaryl-C0-4-alkyl, COOH, C(O)NH2, mono- and di- Ci-4-alkyl-carboxamide, mono- and di-C1-4-alkyl-amino-Co-4alkyl, SO3H, SO2NH2, and mono-and di-Ci-4-alkylsulfonamide.
Certain other compounds according to Formula I or Formula II include those compounds in which X is CR5R53 wherein Rs3 is hydrogen, methyl or trifluoromethyl; and R5 is a residue of the formula:
Figure imgf000018_0001
wherein n and g are integers independently selected from O, 1, or 2 (preferably n+g = 1, 2, 3 or 4; or more preferably n+g is 2 or 3);
Z3 is NR23 or O;
Z4, Zs1Z6, and Z7 are each independently selected from the group consisting of N, CH, and CR8; and
R8 and R8a each indepently represent O to 2 groups, each of which is independently selected at each occurrence of R8 and R8a from the group consisting of hydrogen, halogen, Cj- 4-alkyl,
Figure imgf000018_0002
4alkyl, mono- and
Figure imgf000018_0003
heterocycleC0- 4alkylamino and heterocycleCo^alkyl; or two R8a, taken in combination, form a fused- or spiro-cyclic 3-7 membered ring.
Yet other compounds of Formula I or Formula II include those compounds in which X is CR5a, R5a is hydrogen or methyl, and R5 is a residue selected from the group consisting of:
Figure imgf000018_0004
Figure imgf000019_0001
wherein R8 is selected from hydrogen, methyl, ethyl, mono-, di-, or tri-fluoromethyl, mono-, di-, or tri-fluoromethoxy, fluoro, and chloro. In still other compounds of Formula I or Formula II include those compounds in
which the residue
Figure imgf000020_0001
is a residue of the formula:
Figure imgf000020_0002
wherein wherein R6 is hydrogen, methyl, ethyl, and mono-, di-, and tri-fluoromethyl; R8 is selected from R8 is selected from hydrogen, methyl, ethyl, mono-, di-, or tri- fluoromethyl, mono-, di-, or tri-fluoromethoxy, fluoro, and chloro.
Still other compounds of Formula I or Formula II include those compounds in which X is CR5a, R5a is hydrogen or methyl, and R5 is a residue selected from the group consisting of:
Figure imgf000020_0003
Figure imgf000021_0001
Still other compounds of the invention according to Formula II include compounds according to Formula lib:
Figure imgf000022_0001
lib
Z2 is nitrogen or CH; ki and k2 are 0 or 1 such that a sum of k| and k2 equals 1 or 2;
Ra and Rb taken together form a spirocyclic 3 to 6 membered ring having 0, 1 or 2 ring heteroatoms selected from N, O and S, which fused or spirocyclic ring has 0 to 2 independently selected substituents selected from halogen, Ci^alkyl, C^alkoxy, Ci-
4alkanoyl, and phenyl;
Rc represents 0 to 2 substituents which are independently selected at each occurrence OfR0 from the group consisting of halogen, Ci^alkyl, and phenyl, or two geminal R0 substitents, taken in combination form a 3 to 6 member spirocyclic ring;
R4 represents 0, 1, or 2 substituents each of which is independently selected from H and C 1-4- alkyl; and
R6 is hydrogen or Chalky!.
In certain compounds of the invention according to Formula lib, the divalent residue:
Figure imgf000022_0002
is selected from the group consisting of:
Figure imgf000023_0001
Certain compounds of Formula II, include those compounds, in which the
Figure imgf000023_0002
ring is a divalent residue derived from a proline residue selected from the group consisting of:
Figure imgf000023_0003
Figure imgf000024_0001
Certain other compounds of Formula II, Formula Ha or Formula lib include compounds in which X is CR.5R.5a, R4 is H, and R5 and R53 taken in combination form a 3 to 6 member spirocyclic carbocycle substituted with 0-2 substitutents selected from halogen, C1-6- alkyl, C2-6-alkenyl, C2-6-alkynyl, Ci-6-alkoxide, C3-7-cycloalkyl-C0-4-alkyl, phenyl-Co-4-alkyl, naphthyl-Co-4-alkyl,
Figure imgf000024_0002
or two substitutents taken together form a fused or spirocyclic 3 to 7 membered carbocyclic ring, each of which is substituted with 0-3 independently selected halogen atoms or Q^-alkyl groups.
Certain compounds of Formulae I include compounds of Formula III:
Figure imgf000024_0003
and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereofs.
Certain compounds of the invention according to Formula III include compounds in which:
Zi is absent or NR10; Z2 is nitrogen or CH;
R3 is selected from the group consisting of H, Ci-4-alkyl, and C3-6-cycloalkylCo- C4alkyl;
Rn, Ri5 and R22 are selected from the group consisting of H, alkyl-aryl, Ci^-alkyl, O- Ci-4-alkyl, N(H)-C M-alkyl, and C3-6-cycloalkylC0-C4alkyl;
Rio and Rn are each, independently, selected from the group consisting of H, Ci-4- alkyl and (CH2)0-4-C3-6-cycloalkyl; or
Ri5 and Ri6 may together form a 3, 4, 5, 6 or 7-membered ring that may comprise between 0 to 3 additional heteroatoms, wherein the ring may be further substituted with 0-5 substitutents; or
Ri6 and Ri7 may together form a 3, 4, 5, 6 or 7-membered ring that may comprise between 0 to 3 additional heteroatoms, wherein the ring may be further substituted with 0-5 substitutents; and
V is selected from the group consisting of -Ql-Q2, wherein Q1 is absent, C(O), N(H), N(C1-4-alkyl), C=N(CN), C=N(SO2CH3), or C=N-COH, and Q2 is H, CM-alkyl, C=N-COH-
Ci-4-alkyl, O-C1-4-alkyl, NH2, N(H)-C 1-4-alkyl, N(CM-alkyl)2, SO2-aryl, SO2-CM-alkyl, C3-6- cycloalkyl-Co-4-alkyl, aryl, heteroaryl and heterocycle, each of which may be independently substituted one or more times with a halogen atom, Ci-4-alkyl, C1-4alkoxy, C2-C4alkenyloxy, C2-C4alkynyloxy, Ci-4-alkyl substituted by one or more halogen atoms, or C3-6-cycloalkyl; Certain other compounds of the invention according to Formula III include compounds in which:
R3 is selected from the group consisting of H and Ci-4-alkyl;
R13 Js H;
R8, Rio and Rn are each, independently, selected from the group consisting of H, Ci-4- alkyl, and C3-7cycloalkylC0-4alkyl;
Ri2 is selected from the group consisting of H, Ci-4-alkyl and (CH2)0-4-C3-6-cycloalkyl; and
V is selected from the group consisting of-Q'-Q2, wherein Q1 is absent, C(O), N(H), N(Ci-4-alkyl), C=N(CN), C=N(SO2CH3), or C=N-COH, and Q2 is H, CM-alkyl, C=N-COH- d-4-alkyl, O-C1-4-alkyl, NH2, N(H)-C ^-alkyl, N(C1-4-alkyl)2, SO2-aryl, SO2-CM-alkyl, C3-6- cycloalkyl-Co-4-alkyl, aryl, heteroaryl and heterocycle, each of which may be independently substituted one or more times with a halogen atom, Q-4-alkyl, Ci^-alkyl substituted by one or more halogen atoms, C^alkoxy, C2-C4alkenyloxy, C2-C4alkynyloxy, or C^e-cycloalkyl.
Certain compounds of Formula III include compounds represented by Formula Ilia:
Figure imgf000026_0001
and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof; wherein Z2 is nitrogen or CH;
R25 and R26 are each, independently, selected from the group consisting of H, C1-4- alkyl, O-Q^-alkyl, N(R24)2, C3-6cycloalkylCo-C4alkyl, substituted or unsubstituted aryl and substituted or unsubstituted heterocycle, wherein each R24 is independently selected from the group consisting of H, halogen, hydroxy, COOH, amino, carboxamide, substituted or unsubstitiited-d-4-alkyl, substituted or unsubstituted C3-6cycloalkylCo-C4alkyl, substituted or unsubstituted-Ci-4-alkoxy, substituted or unsubstituted C^cycloalkylCo^alkyl-oxy-, substituted or unsubstituted arylC0-C4alkyl, substituted or unsubstituted heterocycleCo- C4alkyl, substituted or unsubstituted arylCo-C4alkyl-oxy and substituted or unsubstituted heterocycleCo-C4alkyl-oxy; or R22 or R26 may together form a 3-membered ring that is substituted or unsubstituted.
In another embodiment of Formula Ilia, R25 is H and R26 is amine, substituted or unsubstiuted phenyl, or substituted or unsubstiuted benzyl.
Certain other compounds of Formula III include compounds represented by Formula HIb:
Figure imgf000027_0001
and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof; wherein Z2 is nitrogen or CH;
R27 and R28 are each, independently, selected from the group consisting of H, C1-4- alkyl, O-C1-4-alkyl, N(R24)2, C3-6cycloalkylCo-C4alkyl, substituted or unsubstituted aryl, substituted or unsubstituted O-aryl and substituted or unsubstituted heterocycle, wherein R24 is independently selected at each occurrence from the group consisting of H, halogen, hydroxy, COOH, amino, carboxamide, substituted or unsubstituted-Ci-4-alkyl, substituted or unsubstituted C3-6cycloalkylCo-C4alkyl, substituted or unsubstituted-C1-4-alkoxy, substituted or unsubstituted C3-6cycloalkylCo-C4alkyl-oxy-, substituted or unsubstituted arylCo-C4alkyl, substituted or unsubstituted heterocycleCo-C4alkyl, substituted or unsubstituted arylCo- C4alkyl-oxy and substituted or unsubstituted heterocycleC0-C4alkyl-oxy. In one embodiment of Formula HIb, R28 is quinoline,
Figure imgf000027_0002
or O- quinoline, wherein the quinoline and O-quinoline substituents may be independently substituted one or more times (or preferably between one and five times) with halogen, amino, O-Ci-4-alkyl, substituted or
Figure imgf000027_0003
substituted or unsubstituted- (CH2)o-4-C3-6-cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted O-aryl, and substituted or unsubstituted heterocycle.
Yet other compounds of Formula III include compounds represented by Formula IIIc:
Figure imgf000028_0001
and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof; wherein
Z2 is nitrogen or CH;
R29 and R30 are selected from the group consisting of H, Ci-4-alkyl, O-C^-alkyl, N(R24)2, C3-6cycloalkylCo-C4alkyl, substituted or unsubstituted aryl, substituted or unsubstituted aryl-oxy and substituted or unsubstituted heterocycle, wherein each R24 is independently selected at each occurrence from the group consisting of H, halogen, hydroxy, COOH, amino, carboxamide, substituted or unsubstituted-Ci-4-alkyl, substituted or unsubstituted C3-6cycloalkylCo-C4alkyl, substituted or unsubstituted-Ci-4-alkoxy, substituted or unsubstituted C3-6cycloalkylCo-C4alkyl-oxy-, substituted or unsubstituted arylCo-C4alkyl, substituted or unsubstituted heterocycleCo-C4alkyl, substituted or unsubstituted arylCo- C4alkyl-oxy and substituted or unsubstituted heterocycleCo-C4alkyl-oxy.
In one embodiment of Formula IHc, R2g is selected from the group consisting of O- phenyl and O-benzyl.
Still other compounds of Formula III include compounds represented by Formula IHd:
Figure imgf000028_0002
and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof; wherein
Z2 is nitrogen or CH;
R31 represents one or two residues which are independently selected at each occurrence from the group consisting of H, Ci-4-alkyl, O-Ci-4-alkyl, N(R24)2, (CH2)0-4-C3-6- cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted O-aryl and substituted or unsubstituted heterocycle, wherein each R24 is independently selected from the group consisting of H, halogen, hydroxy, COOH, amino, carboxamide, substituted or unsubstituted-Ci^-alkyl, substituted or unsubstituted C3-6cycloalkylCo-C4alkyl, substituted or
Figure imgf000029_0001
substituted or unsubstituted C3-6cycloalkylCo-C4alkyl-oxy-, substituted or unsubstituted arylCo-C4alkyl, substituted or unsubstituted heterocycleC0- C4alkyl, substituted or unsubstituted arylCo-C4alkyl-oxy and substituted or unsubstituted heterocycleCo-C4alkyl-oxy; or two R31 residues may together form a 3, 4, 5, 6 or 7-membered ring that is aromatic ^ or non-aromatic and may contain one or more heteroatoms selected from N, O or S, wherein the ring may be further substituted one. or more times (or preferably between one and five times). hi another embodiment, Formula Hid is represented by a compound of the Formula IHe:
Figure imgf000029_0002
and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof; wherein
Z2 is nitrogen or CH; R32 is -Q'-Q2, wherein Q1 is absent, C(O), S(O)P, N(H), N(C1-4-alkyl), C=N(CN), C=N(SO2CH3), or C=N-COH, and Q2 is H, CI-4-alkyl, C=N-COH-CM-alkyl, O-C1-4-alkyl, NH2, N(H)-Ci -4-alkyl, N(CM-alkyl)2, SO2-aryl, SO2-C M-alkyl, C3-6-cycloalkyl-C0-4-alkyl, aryl, heteroaryl and heterocycle, each of which may be independently substituted one or more times (or preferably between one and five times) with a halogen atom, Ci-4-alkyl, Ci-4-alkyl substituted by one or more halogen atoms, or C3-^-cycloaUcyl.
In another embodiment, Formula IHd is represented by a compound of the Formula HIf
Figure imgf000030_0001
and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof.
In another embodiment, Formula IHd is represented by a compound of the Formula IHg:
Figure imgf000030_0002
and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof. Certain compounds of Formula III include compounds represented by Formula Illh:
Figure imgf000031_0001
IHh and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof; wherein
R35 is H, halogen, hydroxy, COOH, amino, carboxamide, substituted or unsubstituted- Ci-4-alkyl, substituted or unsubstituted C3-6cycloalkylCo-C4alkyl, substituted or unsubstituted- Ci-4-alkoxy, substituted or unsubstituted C3^cyclpalkylCo-C4alkyl-oxy-, substituted or unsubstituted arylCo-C4alkyl, substituted or unsubstituted heterocycleCo-C4alkyl, substituted or unsubstituted arylCo-C4alkyl-oxy and substituted or unsubstituted heterocycleCo-C4alkyl- oxy.
In one embodiment of Formula Illh, R35 is phenyl, optionally substituted with chloro.
Certain compounds of Formula I include compounds of Formula IV:
Figure imgf000031_0002
and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof.
Certain compounds of Formula FV include those compounds in which: y is 0 or 1 ;
Z2 is nitrogen or CH; Ri is selected from the group consisting of H and Ci-4-alkyl;
Ri7 is hydrogen or is selected from the group consisting of Ci^-alkyl, Ci-6-cycloalkyl, (CH2)o-4-C3-6-cycloalkyl, aryl, alkyl-aryl and heterocycle, each of which may be independently substituted one or more times (or preferably between one and five times); Rio and Rn are each, independently, selected from the group consisting of H and Ci-4- alkyl;
Ri2 is selected from the group consisting of H, Ci-4-alkyl, Ci-β-cycloalkyl and aryl; and
V is selected from the group consisting of-Q'-Q2, wherein Q1 is absent, C(O), N(H), N(Ci-4-alkyl), C=N(CN), C=N(SO2CH3), or C=N-COH, and Q2 is H, CM-alkyl, C=N-COH- CM-alkyl, O-Ci-4-alkyl, NH2, N(H)-C, -4-alkyl, N(C1-4-alkyl)2, SO2-aryl, SO2-CM-alkyl, C3-6- cycloalkyl-Co-4-alkyl, aryl, heteroaryl and heterocycle, each of which may be independently substituted one or more times (or preferably between one and five times) with a halogen atom, Ci-4-alkyl,
Figure imgf000032_0001
C2-C4alkenyloxy, C2-C4alkynyloxy, Ci-4-alkyl substituted by one or more halogen atoms, or C3-6-cycloalkyl; or Ri i and V form the following 5-membered ring which may be further substituted:
Figure imgf000032_0002
R-13
Certain other compounds of Formula IV include those compounds in which R]7 is selected from the group consisting of H, cyclopropylCo-C2alkyl, cyclopentylCo-C2alkyl, phenylCi-C2alkyl, and naphthylCi-C2alkyl.
Certain other compounds of Formulae I, II (including Ha and lib), III (including Ilia through IIDi), and/or IV include those compounds in which V is selected from the group consisting of C(O)R24, C(O)C(O)OR24, C(O)N(H)R24, C(O)C(O)N(H)R24 and C(O)OR24, wherein each R24 is independently selected from the group consisting of H, halogen, substituted or unsubstituted-Ci-4-alkyl, substituted or unsubstituted C3-6-cycloalkylCo-C4alkyl, substituted or unsubstituted arylCo-C4alkyl and substituted or unsubstituted heterocycleCo- C4alkyl, and any combination thereof.
Yet other compounds of Formulae I, II (including Ha and lib), III (including Ilia through IIDi), and/or IV include compounds in which V is C(O)-R20, wherein R20 is selected from the group consisting of tert-butyl, C3-6-cycloalkyl, phenyl, pyrazine, benzooxazole, 4,4- dimethyl-4,5-dihydro-oxazole, benzoimidazole, pyrimidine, thiazole, benzothiazole, benzothiazole 1,1 -dioxide and quinazoline, each of which may be further independently substituted with 0-5 substitutents selected from a halogen atom,
Figure imgf000033_0001
Ci-4alkoxy, C2- Qalkenyloxy, C2-C4alkynyloxy,
Figure imgf000033_0002
substituted by one or more halogen atoms, or C3-6- cycloalkyl.
Still other compounds of Formulae I, II (including Ha and lib), III (including Ilia through HIh), and/or IV include compounds in which V is R20 or C(O)-R2O, wherein R20 is a residue of the formula:
Figure imgf000033_0003
wherein
Z8 is absent or selected from NR33 or oxygen; g and fare independently selected integers selected from the group consisting of 0, 1,
2, 3 and 4; j is an integer selected from the group consisting of 1, 2, 3 and 4, wherein the sum of f + g + j is less than or equal to 5 and greater than or equal to 2 when Z8 is absent and the sum of f + g + jk is less than or equal to 4 and greater than or equal to 1 when Z8 is oxygen; R33 is independently selected at each occurrence from the group consisting of hydrogen,
Figure imgf000033_0004
and
Figure imgf000033_0005
and
R34 represents zero to three residues each independently selected at each occurrence from the group consisting of halogen, hydroxy, amino,
Figure imgf000033_0006
C3-6cycloalkyl,
Figure imgf000033_0007
Figure imgf000033_0008
Yet other compounds of Formulae I, II (including IIa and lib), III (including FIIa through HIh), and/or FV include compounds in which V is C(O)-R2O, wherein R20 is a residue of the formula:
Figure imgf000033_0009
wherein g is an integer selected from the group consisting of 0, 1, 2, 3 and 4; j is an integer selected from the group consisting of 1, 2, 3 and 4, wherein the sum of g + j is less than or equal to 5 and greater than or equal to 2; R33 is independently selected at each occurrence from the group consisting of hydrogen, Ci^alkyl, haloCi^alkyl, C3-6cycloalkyl, hydroxyCi^alkyl, and Ci^alkoxyCi^alkyl; and
R34 represents zero to three residues each independently selected at each occurrence from the group consisting of halogen, hydroxy, amino,
Figure imgf000034_0001
mono-and
Figure imgf000034_0002
and C1-4alkoxyC1-4alkyl.
In another embodiment of Formula I, X is CR5Rsa, R4 and Rs3 are H and R5 is aryl-C0- 3-alkyl, -O-heterocycle, or heterocycle-Co-3-alkyl, wherein aryl and heterocycle may be independently substituted one or more times (or preferably between one and five times) with a halogen atom, aryl, trihalomethyl, C3-6-cycloalkyl or C1-4-alkyl. In yet another embodiment of Formula I, X is CR5Rs3, R4 and R53 are H and R5 is selected from the group consisting of piperidine, phenyl, -O-pyridinyl and CH2-pyridinyl, wherein the phenyl and pyridinyl groups may be independently substituted one or more times (or preferably between one and five times) with a halogen atom or Ci^-alkyl.
In yet another embodiment of formula I, R5 is 5-chloro-pyridin-2-yl. In still another embodiment of formulae I or II (including Ha and lib), R5 is selected from the group consisting of
Figure imgf000034_0003
Figure imgf000035_0001
wherein R2] is independently selected from the group consisting of C1-4-alkyl and aryl.
In still other embodiments, CR5R5a, taken in combination, form a spirocyclic 3 to 6 member carbocyclic ring. Certain spirocyclic rings include groups of the formula:
Figure imgf000035_0002
wherein fis O, 1, 2, 3, 4 or 5;
Rsb and R5c are independently selected from hydrogen halogen, Ci-6-alkyl, C2-6-alkenyl, C2-6- alkynyl, Ci^-alkoxide, C3-7-cycloalkyl-Co-4-alkyl, phenyl-Co^t-alkyl, naphthyl-Co-4-alkyl, heteroaryl7Co-4-alkyl, or two substitutents taken together form a fused or spirocyclic 3 to 7 membered carbopyclic ring, each of which is substituted with 0-3 independently selected halogen atoms or C1-4-alkyl groups.
In yet another embodiment of Formula I, R2 is selected from the group consisting of propyl and (CH2)2-cyclobutyl. In still another embodiment of Formula I, Rn is H and R12 is C3-6-cycloalkyl.
In one embodiment of Formula I, R12 is cyclohexyl.
In another embodiment of formula I, V is selected from the group consisting of C(O)- N(H)-/-butyl.
Yet other compounds of any one of Formulae I, II (including Ha and lib), III (including Ilia through HIh), and/or IV include compounds in which V is C(O)-N(H)-/-butyl or C(O)-R20, wherein R20 is selected from the group consisting of C3-6-cycloalkyl, phenyl, pyrazine, benzooxazole, 4,4-dimethyl-4,5-dihydro-oxazole, benzoimidazole, pyrimidine, thiazole, benzothiazole, benzothiazole 1,1 -dioxide and quinazoline, all of which may be further independently substituted with a halogen atom, CF3, Cι-4-alkyl, Q^alkoxy, C2- C4alkenyloxy, C2-C4alkynyloxy, or C3-6-cycloalkyl.
In certain other compounds of any one of Formulae I, II (including Ha and lib), III (including HIa through IIDi), and/or IV, V is selected from the group consisting Of C3-6- cycloalkyl, phenyl, pyrazine, benzooxazole, 4,4-dimethyl-4,5-dihydro-oxazole, benzoimidazole, pyrimidine, thiazole, benzothiazole, benzothiazole 1,1 -dioxide and quinazoline, all of which may be further independently substituted with a halogen atom, CF3, Ci-4-alkyl , Ci-4alkoxy, C2-C4alkenyloxy, C2-C4alkynyloxy, or C3-6-cycloalkyl.
In yet another embodiment of Formulae I, II (including Ha and lib), III (including Ilia through HIh), and/or FV, V is R20 or C(O)-R20, wherein R20 is selected from the group consisting of C3-6-cycloalkyl, phenyl, pyrazine, benzooxazole, 4,4-dimethyl-4,5-dihydro- oxazole, benzoimidazole, pyrimidine, benzothiazole 1,1 -dioxide and quinazoline, all of which may be further independently substituted with a halogen atom, CF3, Ci-4-alkyl or C3-6- cycloalkyl.
In still another embodiment of Formulae I, II (including Ha and lib), III (including Ilia through IHh), and/or IV, V is R20 or C(O)-R20, wherein R2o is selected from the group consisting of
Figure imgf000036_0001
wherein Ri8 is selected from the group consisting of hydrogen, a halogen atom, aryl, Ci-4- alkyl, Ci^alkoxy, C2-C4alkenyloxy, C2-C4alkynyloxy, C^-alkyl substituted by one or more halogen atoms, or C3-6-cycloalkyl.
In one embodiment of Formulae I, II (including Ha and lib), in (including Ilia through HIh), and/or IV, V is R20 or C(O)-R20, wherein R20 is selected from the group consisting of
Figure imgf000036_0002
wherein R] 8 is selected from the group consisting of hydrogen, a halogen atom, aryl, Ci-4- alkyl, Ci^alkoxy, C2-C4alkenyloxy, C2-C4alkynyloxy, d^-alkyl substituted by one or more halogen atoms, or C3-6-cycloalkyl.
In another embodiment of Formulae I, II (including IIa and lib), III (including Ilia through Illh), and/or IV, V is selected from the group consisting of C3-6-cycloalkyl, phenyl, pyrazine, benzooxazole, 4,4-dimethyl-4,5-dihydro-oxazole, benzoimidazole, pyrimidine, thiazole, benzothiazole, benzothiazole 1,1 -dioxide and quinazoline, all of which may be further independently substituted with a halogen atom, Ci-4-alkyl, C1-4alkoxy, C2- C4alkenyloxy, C2-C4alkynyloxy, Ci-4-alkyl substituted by one or more halogen atoms, or C3-6- cycloalkyl.
In yet another embodiment of Formula I, II (including IIa and lib), III (including Ilia through IIDi), and/or FV, variable V is selected from the group consisting of R20 and C(O)- R20, wherein R20 is selected from the group consisting of C3-6-cycloalkyl, mono- and di-Ci. 4alkylamino, phenyl, pyrazine, benzooxazole, 4,4-dimethyl-4,5-dihydro-oxazole, benzoimidazole, pyrimidine, benzothiazole 1,1 -dioxide and quinazoline, each of which may be further independently substituted with a halogen atom, CF3, Ci-4-alkyl, Ci-4alkoxy, C2- C4alkenyloxy, C2-C4alkynyloxy, or C3-6-cycloalkyl.
In still another embodiment of Formula I, II (including IIa and lib), III (including Ilia through HIh), and/or IV, variable V is selected from the group consisting of R20 and C(O)- R20, wherein R20 is selected from the group consisting of
Figure imgf000037_0001
Figure imgf000038_0001
wherein b is 0, 1, or 2; and R18 is selected from the group consisting of hydrogen, a halogen atom, aryl, trihalomethyl, and Ci-4-alkyl.
In one embodiment, any of the C3-6-cycloalkyl groups of Formula I, or any subformula thereof, may be independently substituted one or more times (or preferably between one and five times) with a halogen atom, aryl, heteroaryl, trihalomethyl, C^-alkoxy or C1-4-alkyl.
In one embodiment of Formula I, or any sub formulae thereof, any of the heterocycle groups are independently selected from the group consisting of acridinyl, carbazolyl, cinnolinyl, quinoxalinyl, pyrrazolyl, indolyl, benzotriazolyl, furanyl, thienyl, benzothienyl, benzo furanyl, quinolinyl, isoquinolinyl, oxazolyl, isoxazolyl, indolyl, pyrazinyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolyl, tetrahydroquinoline, benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, imidazolyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridazinyl, pyridyl, pyrimidyl, pyrrolyl, quinazolinyl, quinolyl, quinoxalinyl, tetrahydropyranyl, tetrazolyl, tetrazolopyridyl, thiadiazolyl, thiazolyl, thienyl, triazolyl, azetidinyl, 1 ,4-dioxanyl, hexahydroazepinyl, piperazinyl, piperidinyl, pyridin-2- onyl, pyrrolidinyl, morpholinyl, thiomorpholinyl, dihydrobenzoimidazolyl, dihydrobenzofuranyl, dihydrobenzothiophenyl, dihydrobenzoxazolyl, dihydrofuranyl, dihydroimidazolyl, dihydroindolyl, dihydroisooxazolyl, dihydroisothiazolyl, dihydrooxadiazolyl, dihydrooxazolyl, dihydropyrazinyl, dihydropyrazolyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dihydroquinolinyl, dihydrotetrazolyl, dihydrothiadiazolyl, dihydrothiazolyl, dihydrothienyl, dihydrotriazolyl, dihydroazetidinyl, methylenedioxybenzoyl, tetrahydrofuranyl, and tetrahydrothienyl, and N-oxides thereof, all of which may be independently further substituted one or more times (or preferably between one and five times) with a halogen atom, Ci-4-alkyl, Ci^-alkyl substituted by one or more halogen atoms, or C3-6-cycloalkyl.
Preferred embodiments of the compounds of the invention (including pharmaceutically acceptable salts thereof, as well as enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof) are shown below in Table A and Table B, and are also considered to be "compounds of the invention."
TABLE A
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
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
Certain additional compounds of Formula I (or sub formulae thereof) which are contemplated in the present invention include compounds depicted in Table B. Table B
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
Certain other compounds of Formula I, and sub formulae thereof, include those compounds which contain a fragment selected from the residues of each of Tables C, D, E, F, hand G. Thus, compounds of the invention include all P1-P2 compounds formed by combining all possible permutations of the fragments of Tables C, D, E, F and G wherein the bond ending in an asterisk is the point of attachment Pl and P2 fragments are coupled by condensation of the amino residue on the Pl fragment with the carboxylic acid residue on the P2 fragment. For example, the compound C(l)-D(3)-E(10)-F(4)-G(15) is the compound in which the residue of entry 1 of Table C, the residue of entry 3 of Table D, the residue of entry 10 of Table E, the residue of entry 4 of Table F (where n is 1) and the residue of entry 15 of Table G are combined to form a compound of formula I which has the structure:
Figure imgf000103_0001
C(I) D(3) E(IO) F(21) where n = 1 G(15)
Figure imgf000103_0002
TABLE C
Figure imgf000103_0003
TABLE D
The variable, E, of Formula 1 is a residue selected from the group consisting of: Dl ^-C(O)NH-# I D2 @-NHSO2-# | D3 @-NHSO2NMe-#
TABLE E
The fragment of Formula 1 has a residue of the formula
Figure imgf000104_0001
selected from the group consisting of:
Figure imgf000104_0002
Figure imgf000105_0001
Figure imgf000106_0001
TABLEF
Figure imgf000106_0002
Figure imgf000107_0001
TABLE G
The fragment of Formula 1 has a residue of the formula
Figure imgf000107_0002
selected from the group consisting of:
Figure imgf000107_0003
#
Figure imgf000108_0001
Figure imgf000109_0001
Using the HCV NS3-4A protease and Luciferase-HCV replicon assays described in the exemplification section below, certain compounds of the invention (including compounds of Table A depicted above) are found to show IC50 values for HCV inhibition in the range from 10 to more than 100 μM, or 0.5 to 30 μM, or show IC5O values for HCV inhibition of less than 10 μM.
In certain embodiments, a compound of the present invention is further characterized as a modulator of HCV, including a mammalian HCV, and especially including a human HCV. In a preferred embodiment, the compound of the invention is an HCV inhibitor. The terms "HCV-associated state" or "HCV-associated disorder" include disorders and states (e.g., a disease state) that are associated with the activity of HCV, e.g., infection of HCV in a subject. HCV-associated states include HCV-infection, liver cirrhosis, chronic liver disease, hepatocellular carcinoma, cryoglobulinemia, non-Hodgkin's lymphoma, and a suppressed innate intracellular immune response. HCV-associated states are often associated with the NS3 serine protease of HCV, which is responsible for several steps in the processing of the HCV polyprotein into smaller functional proteins. NS3 protease forms a heterodimeric complex with the NS4A protein, an essential cofactor that enhances enzymatic activity, and is believed to help anchor HCV to the endoplasmic reticulum. NS3 first autocatalyzes hydrolysis of the NS3-NS4A juncture, and then cleaves the HCV polyprotein intermolecularly at the NS4A-NS4B, NS4B-NS5 A and NS5A-NS5B intersections. This process is associated with replication of HCV in a subject. Inhibiting or modulating the activity of one or more of the NS3, NS4A, NS4B, NS5A and NS5B proteins will inhibit or modulate replication of HCV in a subject, thereby preventing or treating the HCV-associated state. In a particular embodiment, the HCV-associated state is associated with the activity of the NS3 protease. In another particular embodiment, the HCV- associated state is associated with the activity of NS3-NS4A heterodimeric complex. In one embodiment, the compounds of the invention are NS3/NS4A protease inhibitors. In another embodiment, the compounds of the invention are NS2/NS3 protease inhibitors. Without being bound by theory, it is believed that the disruption of the above protein- protein interactions by the compounds of the invention will interfere with viral polyprotein processing by the NS3 protease and thus viral replication.
HCV-associated disorders also include HCV-dependent diseases. HVC-dependent diseases include, e.g., any disease or disorder that depend on or related to activity or misregulation of at least one strain of HCV.
The present invention includes treatment of HCV-associated disorders as described above, but the invention is not intended to be limited to the manner by which the compound performs its intended function of treatment of a disease. The present invention includes treatment of diseases described herein in any manner that allows treatment to occur, e.g., HCV infection.
In a related embodiment, the compounds of the invention can be useful for treating diseases related to HFV, as well as HIV infection and AIDS (Acquired Immune Deficiency Syndrome). In certain embodiments, the invention provides a pharmaceutical composition of any of the compounds of the present invention. In a related embodiment, the invention provides a pharmaceutical composition of any of the compounds of the present invention and a pharmaceutically acceptable carrier or excipient of any of these compounds. In certain embodiments, the invention includes the compounds as novel chemical entities. In one embodiment, the invention includes a packaged HCV-associated disorder treatment. The packaged treatment includes a compound of the invention packaged with instructions for using an effective amount of the compound of the invention for an intended use.
The compounds of the present invention are suitable as active agents in pharmaceutical compositions that are efficacious particularly for treating HCV-associated disorders. The pharmaceutical composition in various embodiments has a pharmaceutically effective amount of the present active agent along with other pharmaceutically acceptable excipients, carriers, fillers, diluents and the like. The phrase, "pharmaceutically effective amount" as used herein indicates an amount necessary to administer to a host, or to a cell, issue, or organ of a host, to achieve a therapeutic result, especially an anti-HCV effect, e.g., inhibition of proliferation of the HCV virus, or of any other HCV-associated disease.
In one embodiment, the diseases to be treated by compounds of the invention include, for example, HCV infection, liver cirrhosis, chronic liver disease, hepatocellular carcinoma, cryoglobulinemia, non-Hodgkin's lymphoma, and a suppressed innate intracellular immune response.
In other embodiments, the present invention provides a method for inhibiting the activity of HCV. The method includes contacting a cell with any of the compounds of the present invention. In a related embodiment, the method further provides that the compound is present in an amount effective to selectively inhibit the activity of one or more of the NS3, NS4A, NS4B, NS5A and NS5B proteins. In another related embodiment, the method provides that the compound is present in an amount effective to diminish the HCV RNA load in a subject.
In other embodiments, the present invention provides a use of any of the compounds of the invention for manufacture of a medicament to treat HCV infection in a subject.
In other embodiments, the invention provides a method of manufacture of a medicament, including formulating any of the compounds of the present invention for treatment of a subject. Definitions The term "treat," "treated," "treating" or "treatment" includes the diminishment or alleviation of at least one symptom associated or caused by the state, disorder or disease being treated. In certain embodiments, the treatment comprises the induction of an HCV- inhibited state, followed by the activation of the HCV-modulating compound, which would in turn diminish or alleviate at least one symptom associated or caused by the HCV-associated state, disorder or disease being treated. For example, treatment can be diminishment of one or several symptoms of a disorder or complete eradication of a disorder.
The term "subject" is intended to include organisms, e.g., prokaryotes and eukaryotes, which are capable of suffering from or afflicted with an HCV-associated disorder. Examples of subjects include mammals, e.g., humans, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats, and transgenic non-human animals. In certain embodiments, the subject is a human, e.g., a human suffering from, at risk of suffering from, or potentially capable of suffering from an HCV-associated disorder, and for diseases or conditions described herein, e.g., HCV infection. In another embodiment, the subject is a cell.
The language "HCV-modulating compound," "modulator of HCV" or "HCV inhibitor" refers to compounds that modulate, e.g. , inhibit, or otherwise alter, the activity of HCV. Similarly, an "NS3/NS4A protease inhibitor," or an "NS2/NS3 protease inhibitor" refers to a compound that modulates, e.g., inhibits, or otherwise alters, the interaction of these proteases with one another. Examples of HCV-modulating compounds include compounds of Formula I, as well as Table A and Table B (including pharmaceutically acceptable salts thereof, as well as enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof).
Additionally, the method includes administering to a subject an effective amount of an HCV-modulating compound of the invention, e.g., HCV-modulating compounds of Formula I, as well as Table A and Table B (including pharmaceutically acceptable salts thereof, as well as enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof).
The term "alkyl" includes saturated aliphatic groups, including straight-chain alkyl groups (e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, etc.), branched-chain alkyl groups (isopropyl, tert-butyl, isobutyl, etc.), cycloalkyl (alicyclic) groups (cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl), alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups. The term "alkyl" also includes alkenyl groups and alkynyl groups. Furthermore, the expression "Cx-Cy-alkyl", wherein x is 1-5 and y is 2-10 indicates a particular alkyl group (straight- or branched-chain) of a particular range of carbons. For example, the expression Ci-C4-alkyl includes, but is not limited to, methyl, ethyl, propyl, butyl, isopropyl, tert-butyl, isobutyl and sec-butyl. Moreover, the term C3-6-cycloalkyl includes, but is not limited to, cyclopropyl, cyclopentyl, and cyclohexyl. As discussed below, these alkyl groups, as well as cycloalkyl groups, may be further substituted. "Co-Cnalkyl" refers to a single covalent bond (Co) or an alkyl group having from 1 to n carbon atoms; for example "Co-C4alkyl" refers to a single covalent bond or a C]-C4alkyl group; "Co-C8alkyl" refers to a single covalent bond or a Ci-C8alkyl group. In some instances, a substituent of an alkyl group is specifically indicated. For example, "Ci- C4hydroxyalkyl" refers to a Ci-C4alkyl group that has at least one hydroxy substituent.
"Alkylene" refers to a divalent alkyl group, as defined above. Co-C4alkylene is a single covalent bond or an alkylene group having from 1 to 4 carbon atoms; and C0-
Cόalkylene is a single covalent bond or an alkylene group having from 1 to 6 carbon atoms.
"Alkenylene" and "Alkynylene" refer to divalent alkenyl and alkynyl groups respsectively, as defined above.
The term alkyl further includes alkyl groups which can further include oxygen, nitrogen, sulfur or phosphorous atoms replacing one or more carbons of the hydrocarbon backbone. In an embodiment, a straight chain or branched chain alkyl has 10 or fewer carbon atoms in its backbone (e.g., Ci-Qo for straight chain, C3-C10 for branched chain), and more preferably 6 or fewer carbons.
A "cycloalkyl" is a group that comprises one or more saturated and/or partially saturated rings in which all ring members are carbon, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, adamantyl, decahydro-naphthalenyl, octahydro-indenyl, and partially saturated variants of the foregoing, such as cyclohexenyl.
Cycloalkyl groups do not comprise an aromatic ring or a heterocyclic ring. Certain cycloalkyl groups are C3-C8cycloalkyl, in which the group contains a single ring with from 3 to 8 ring members. A "(C3-C8cycloalkyl)Co-C4alkyl" is a C3-C8cycloalkyl group linked via a single covalent bond or a Ci-C4alkylene group. In certain aspects, C3-6-cycloalkyl groups are substituted one or more times (or preferably between one and five times) with substitutents independently selected from a halogen atom, aryl, heteroaryl, trihalomethyl, Ci^-alkoxy or Ci-4-alkyl.
Moreover, alkyl (e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, etc.) include both "unsubstituted alkyl" and "substituted alkyl", the latter of which refers to alkyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone, which allow the molecule to perform its intended function. The term "substituted" is intended to describe moieties having substituents replacing a hydrogen on one or more atoms, e.g. C, O or N, of a molecule. Such substituents can include, for example, alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, moφholino, phenol, benzyl, phenyl, piperizine, cyclopentane, cyclohexane, pyridine, 5H-tetrazole, triazole, piperidine, or an aromatic or heteroaromatic moiety.
Further examples of substituents of the invention, which are not intended to be limiting, include moieties selected from straight or branched alkyl (preferably C1-C5), cycloalkyl (preferably C3-C8), alkoxy (preferably Ci-C6), thioalkyl (preferably Ci-C6), alkenyl (preferably C2-C6), alkynyl (preferably C2-C6), heterocyclic, carbocyclic, aryl (e.g., phenyl), aryloxy (e.g., phenoxy), aralkyl (e.g., benzyl), aryloxyalkyl (e.g. , phenyloxyalkyl), arylacetamidoyl, alkylaryl, heteroaralkyl, alkylcarbonyl and arylcarbonyl or other such acyl group, heteroarylcarbonyl, or heteroaryl group, (CR'R")o-3NR'R" (e.g., -NH2), (CR'R")0-3CN (e.g., -CN), -NO2, halogen (e.g., -F, -Cl, -Br, or -I), (CR'R")o-3C(halogen)3 (e.g., -CF3), (CR'R")0-3CH(halogen)2, (CR'R")0-3CH2(halogen), (CR'R")o-3CONR'R", (CR'R")0-3(CNH)NR'R", (CR'R")o_3S(0)1-2NR'R", (CR'R")0-3CHO, (CR'R")0-30(CR'R")o-3H, (CR'R")o-3S(0)o-3R' (e.g., -SO3H, -OSO3H), (CR'R")o-30(CR'R")o-3H (e.g., -CH2OCH3 and -OCH3), (CR'R")O-3S(CR'R")O-3H (e.g., -SH and -SCH3), (CR'R")0-3OH (e.g., -OH), (CR'R")0-3COR\ (CR'R")0-3(substituted or unsubstituted phenyl), (CR'R")o-3(C3-C8 cycloalkyl), (CR'R")0-3CO2R' (e.g., -CO2H), or (CR'R")o-3OR' group, or the side chain of any naturally occurring amino acid; wherein R' and R" are each independently hydrogen, a Ci-C5 alkyl, C2-C5 alkenyl, C2-C5 alkynyl, or aryl group. Such substituents can include, for example, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, oxime, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, sulfonato, sulfamoyl, sulfonamide, nitro, trifluoromethyl, cyano, azido, heterocyclyl, or an aromatic or heteroaromatic moiety. In certain embodiments, a carbonyl moiety (C=O) may be further derivatized with an oxime moiety, e.g., an aldehyde moiety may be derivatized as its oxime (-C=N-OH) analog. It will be understood by those skilled in the art that the moieties substituted on the hydrocarbon chain can themselves be substituted, if appropriate. Cycloalkyls can be further substituted, e.g., with the substituents described above. An "aralkyl" moiety is an alkyl substituted with an aryl (e.g., phenylmethyl (/.e., benzyl)).
The term "alkenyl" includes unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but which contain at least one double bond.
For example, the term "alkenyl" includes straight-chain alkenyl groups (e.g. , ethenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl, nonenyl, decenyl, etc.), branched- chain alkenyl groups, cycloalkenyl (alicyclic) groups (cyclopropenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl), alkyl or alkenyl substituted cycloalkenyl groups, and cycloalkyl or cycloalkenyl substituted alkenyl groups. The term alkenyl further includes alkenyl groups that include oxygen, nitrogen, sulfur or phosphorous atoms replacing one or more carbons of the hydrocarbon backbone. In certain embodiments, a straight chain or branched chain alkenyl group has 6 or fewer carbon atoms in its backbone (e.g., C2-C6 for straight chain, C3-C6 for branched chain). Likewise, cycloalkenyl groups may have from 3-8 carbon atoms in their ring structure, and more preferably have 5 or 6 carbons in the ring structure. The term C2-C6 includes alkenyl groups containing 2 to 6 carbon atoms.
Moreover, the term alkenyl includes both "unsubstituted alkenyls" and "substituted alkenyls", the latter of which refers to alkenyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone. Such substituents can include, for example, alkyl groups, alkynyl groups, halogens, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino
(including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety. The term "alkynyl" includes unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but which contain at least one triple bond. For example, the term "alkynyl" includes straight-chain alkynyl groups (e.g., ethynyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl, octynyl, nonynyl, decynyl, etc.), branched- chain alkynyl groups, and cycloalkyl or cycloalkenyl substituted alkynyl groups. The term alkynyl further includes alkynyl groups that include oxygen, nitrogen, sulfur or phosphorous atoms replacing one or more carbons of the hydrocarbon backbone. In certain embodiments, a straight chain or branched chain alkynyl group has 6 or fewer carbon atoms in its backbone (e.g., C2-C6 for straight chain, C3-C6 for branched chain). The term C2-C6 includes alkynyl groups containing 2 to 6 carbon atoms. Moreover, the term alkynyl includes both "unsubstituted alkynyls" and "substituted alkynyls", the latter of which refers to alkynyl moieties having substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone. Such substituents can include, for example, alkyl groups, alkynyl groups, halogens, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety.
The term "amine" or "amino" should be understood as being broadly applied to both a molecule, or a moiety or functional group, as generally understood in the art, and may be primary, secondary, or tertiary. The term "amine" or "amino" includes compounds where a nitrogen atom is covalently bonded to at least one carbon, hydrogen or heteroatom. The terms include, for example, but are not limited to, "alkylamino," "arylamino," "diarylamino," "alkylarylamino," "alkylaminoaryl," "arylaminoalkyl," "alkaminoalkyl," "amide," "amido," and "aminocarbonyl." The term "alkyl amino" comprises groups and compounds wherein the nitrogen is bound to at least one additional alkyl group. The term "dialkyl amino" includes groups wherein the nitrogen atom is bound to at least two additional alkyl groups. The term "arylamino" and "diarylamino" include groups wherein the nitrogen is bound to at least one or two aryl groups, respectively. The term "alkylarylamino," "alkylaminoaryl" or "arylaminoalkyl" refers to an amino group which is bound to at least one alkyl group and at least one aryl group. The term "alkaminoalkyl" refers to an alkyl, alkenyl, or alkynyl group bound to a nitrogen atom which is also bound to an alkyl group.
The term "amide," "amido" or "aminocarbonyl" includes compounds or moieties which contain a nitrogen atom which is bound to the carbon of a carbonyl or a thiocarbonyl group. The term includes "alkaminocarbonyl" or "alkylaminocarbonyl" groups which include alkyl, alkenyl, aryl or alkynyl groups bound to an amino group bound to a carbonyl group. It includes.arylaminocarbonyl and arylcarbonylamino groups which include aryl or heteroaryl moibties bound to an amino group which is bound to the carbon of a carbonyl or thiocarbonyl group. The terms "alkylaminocarbonyl," "alkenylaminocarbonyl," "alkynylaminocarbonyl," "arylaminocarbonyl," "alkylcarbonylamino," "alkenylcarbonylamino," "alkynylcarbonylamino," and "arylcarbonylamino" are included in term "amide." Amides also include urea groups (aminocarbonylamino) and carbamates (oxycarbonylamino).
The term "aryl" includes groups, including 5- and 6-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, phenyl, pyrrole, furan, thiophene, thiazole, isothiaozole, imidazole, triazole, tetrazole, pyrazole, oxazole, isoxazole, pyridine, pyrazine, pyridazine, and pyrimidine, and the like. Furthermore, the term "aryl" includes multicyclic aryl groups, e.g., tricyclic, bicyclic, e.g., naphthalene, benzoxazole, benzodioxazole, benzothiazole, benzoimidazole, benzothiophene, methylenedioxyphenyl, quinoline, isoquinoline, anthryl, phenanthryl, napthridine, indole, benzofuran, purine, benzofuran, deazapurine, or indolizine. Those aryl groups having heteroatoms in the ring structure may also be referred to as "aryl heterocycles", "heterocycles," "heteroaryls" or "heteroaromatics." The aromatic ring can be substituted at one or more ring positions with such substituents as described above, as for example, alkyl, halogen, hydroxyl, alkoxy, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkylaminoacarbonyl, aralkylaminocarbonyl, alkenylaminocarbonyl, alkylcarbonyl, arylcarbonyl, aralkylcarbonyl, alkenylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety. Aryl groups can also be fused or bridged with alicyclic or heterocyclic rings which are not aromatic so as to form a polycycle (e.g., tetralin).
Certain aryl groups recited herein are C6-CioarylCo-Cgalkyl groups (i.e., groups in which a 6- to 10-membered carbocyclic group comprising at least one aromatic ring is linked via a single covalent bond or a CrCgalkylene group). Such groups include, for example, phenyl and indanyl, as well as groups in which either of the foregoing is linked via Ci- C8alkylene, preferably via Ci-C4alkylene. Phenyl groups linked via a single covalent bond or Ci-C6alkylene group are designated phenylCό-Cόalkyl (e.g., benzyl, 1-phenyl-ethyl, 1-pheήyl- propyl and 2-phenyl-ethyl).
"Arylene" refers to a divalent aryl group, as defined above. Arylene is intended to encompass divalent residues of phenyl, naphthyl and biphenyl. "Heteroarylene" refers to divalent heteroaryl groups as defined infra.
The term "heteroaryl", as used herein, represents a stable monocyclic or bicyclic ring of up to 7 atoms in each ring, wherein at least one ring is aromatic and contains from 1 to 4 heteroatoms selected from the group consisting of O, N and S. Heteroaryl groups within the scope of this definition include but are not limited to: acridinyl, carbazolyl, cinnolinyl, quinoxalinyl, pyrrazolyl, indolyl, isoindoline, benzotriazolyl, furanyl, thienyl, benzothienyl, benzofuranyl, quinolinyl, isoquinolinyl, oxazolyl, isoxazolyl, indolyl, pyrazinyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolyl, tetrahydroquinoline. As with the definition of heterocycle below, "heteroaryl" is also understood to include the N-oxide derivative of any nitrogen- containing heteroaryl. In cases where the heteroaryl substituent is bicyclic and one ring is non-aromatic or contains no heteroatoms, it is understood that attachment is via the aromatic ring or via the heteroatom containing ring, respectively.
The term "heterocycle" or "heterocyclyl" as used herein is intended to mean a 5- to 10-membered aromatic or nonaromatic heterocycle containing from 1 to 4 heteroatoms selected from the group consisting of O, N and S, and includes bicyclic groups. "Heterocyclyl" therefore includes the above mentioned heteroaryls, as well as dihydro and tetrathydro analogs thereof. Further examples of "heterocyclyl" include, but are not limited to the following: benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, imidazolyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridazinyl, pyridyl, pyrimidyl, pyrrolyl, quinazolinyl, quinolyl, quinoxalinyl, tetrahydropyranyl, tetrazolyl, tetrazolopyridyl, thiadiazolyl, thiazolyl, thienyl, triazolyl, azetidinyl, 1 ,4-dioxanyl, hexahydroazepinyl, piperazinyl, piperidinyl, pyridin-2-onyl, pyrrolidinyl, morpholinyl, thiomoφholinyl, dihydrobenzoimidazolyl, dihydrobenzofuranyl, dihydrobenzothiophenyl, dihydrobenzoxazolyl, dihydrofuranyl, dihydroimidazolyl, dihydroindolyl, dihydroisooxazolyl, dihydroisothiazolyl, dihydrooxadiazolyl, dihydrooxazolyl, dihydropyrazinyl, dihydropyrazolyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dihydroquinolinyl, dihydrotetrazolyl, dihydrothiadiazolyl, dihydrothiazolyl, dihydrothienyl, dihydrotriazolyl, dihydroazetidinyl, methylenedioxybenzoyl, tetrahydro furanyl, and tetrahydrothienyl, and N-oxides thereof. Attachment of a heterocyclyl substituent can occur via a carbon atom or via a heteroatom.
A "heterocycleCo-Cgalkyl" is a heterocyclic group linked via a single covalent bond or Ci-Qalkylene group. A (4- to 7-membered heterocycle)C0-C8alkyl is a heterocyclic group (e.g., monocyclic or bicyclic) having from 4 to 7 ring members linked via a single covalent bond or an alkylene group having from 1 to 8 carbon atoms. A "(6-membered heteroaryl)Co- C6alkyl" refers to a heteroaryl group linked via a direct bond or Ci-C6alkyl group.
The term "acyl" includes compounds and moieties which contain the acyl radical (CH3CO-) or a carbonyl group. The term "substituted acyl" includes acyl groups where one or more of the hydrogen atoms are replaced by for example, alkyl groups, alkynyl groups, halogens, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety.
The term "acylamino" includes moieties wherein an acyl moiety is bonded to an amino group. For example, the term includes alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido groups. The term "alkoxy" includes substituted and unsubstituted alkyl, alkenyl, and alkynyl groups covalently linked to an oxygen atom. Examples of alkoxy groups include methoxy, ethoxy, isopropyloxy, propoxy, butoxy, and pentoxy groups and may include cyclic groups such as cyclopentoxy. Examples of substituted alkoxy groups include halogenated alkoxy groups. The alkoxy groups can be substituted with groups such as alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamide, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moieties. Examples of halogen substituted alkoxy groups include, but are not limited to, fluoromethoxy, difiuoromethoxy, trifluoromethoxy, chloromethoxy, dichloromethoxy, trichloromethoxy, etc.
The term "carbonyl" or "carboxy" includes compounds and moieties which contain a carbon connected with a double bond to an oxygen atom, and tautomeric forms thereof. Examples of moieties that contain a carbonyl include aldehydes, ketones, carboxylic acids, amides, esters, anhydrides, etc. The term "carboxy moiety" or "carbonyl moiety" refers to groups such as "alkylcarbonyl" groups wherein an alkyl group is covalently bound to a carbonyl group, "alkenylcarbonyl" groups wherein an alkenyl group is covalently bound to a carbonyl group, "alkynylcarbonyl" groups wherein an alkynyl group is covalently bound to a carbonyl group, "arylcarbonyl" groups wherein an aryl group is covalently attached to the carbonyl group. Furthermore, the term also refers to groups wherein one or more heteroatoms are covalently bonded to the carbonyl moiety. For example, the term includes moieties such as, for example, aminocarbonyl moieties, (wherein a nitrogen atom is bound to the carbon of the carbonyl group, e.g., an amide), aminocarbonyloxy moieties, wherein an oxygen and a nitrogen atom are both bond to the carbon of the carbonyl group (e.g., also referred to as a "carbamate"). Furthermore, aminocarbonylamino groups (e.g., ureas) are also include as well as other combinations of carbonyl groups bound to heteroatoms (e.g., nitrogen, oxygen, sulfur, etc. as well as carbon atoms). Furthermore, the heteroatom can be further substituted with one or more alkyl, alkenyl, alkynyl, aryl, aralkyl, acyl, etc. moieties.
The term "thiocarbonyl" or "thiocarboxy" includes compounds and moieties which contain a carbon connected with a double bond to a sulfur atom. The term "thiocarbonyl moiety" includes moieties that are analogous to carbonyl moieties. For example, "thiocarbonyl" moieties include aminothiocarbonyl, wherein an amino group is bound to the carbon atom of the thiocarbonyl group, furthermore other thiocarbonyl moieties include, oxythiocarbonyls (oxygen bound to the carbon atom), aminothiocarbonylamino groups, etc. The term "ether" includes compounds or moieties that contain an oxygen bonded to two different carbon atoms or heteroatoms. For example, the term includes "alkoxyalkyl" which refers to an alkyl, alkenyl, or alkynyl group covalently bonded to an oxygen atom that is covalently bonded to another alkyl group.
The term "ester" includes compounds and moieties that contain a carbon or a heteroatom bound to an oxygen atom that is bonded to the carbon of a carbonyl group. The term "ester" includes alkoxycarboxy groups such, as methoxycarboayl, ethoxycarbonyl, propoxycarbonyl, butoxycarbonyl, pentoxycarbonyl, etc. The alkyl, alkenyl, or alkynyl groups are as defined above.
The term "thioether" includes compounds and moieties which contain a sulfur atom bonded to two different carbon or hetero atoms. Examples of thioethers include, but are not limited to alkthioalkyls, alkthioalkenyls, and alkthioalkynyls. The term "alkthioalkyls" include compounds with an alkyl, alkenyl, or alkynyl group bonded to a sulfur atom that is bonded to an alkyl group. Similarly, the term "alkthioalkenyls" and alkthioalkynyls" refer to compounds or moieties wherein an alkyl, alkenyl, or alkynyl group is bonded to a sulfur atom which is covalently bonded to an alkynyl group.
The term "hydroxy" or "hydroxyl" includes groups with an -OH or -O-.
The term "halogen" includes fluorine, bromine, chlorine, iodine, etc. The term "perhalogenated" generally refers to a moiety wherein all hydrogens are replaced by halogen atoms. The terms "polycyclyl" or "polycyclic radical" include moieties with two or more rings {e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls) in which two or more carbons are common to two adjoining rings, e.g., the rings are "fused rings". Rings that are joined through non-adjacent atoms are termed "bridged" rings. Each of the rings of the polycycle can be substituted with such substituents as described above, as for example, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkoxycarbonyl, alkylaminoacarbonyl, aralkylaminocarbonyl, alkenylaminocarbonyl, alkylcarbonyl, arylcarbonyl, aralkylcarbonyl, alkenylcarbonyl, aminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkyl, alkylaryl, or an aromatic or heteroaromatic moiety. The term "heteroatom" includes atoms of any element other than carbon or hydrogen.
Preferred heteroatoms are nitrogen, oxygen, sulfur and phosphorus.
Additionally, the phrase "any combination thereof implies that any number of the listed functional groups and molecules may be combined to create a larger molecular architecture. For example, the terms "phenyl," "carbonyl" (or "=O"), "-O-," "-OH," and C1-6 (i.e., -CH3 and -CH2CH2CH2-) can be combined to form a 3-methoxy-4-propoxybenzoic acid substituent. It is to be understood that when combining functional groups and molecules to create a larger molecular architecture, hydrogens can be removed or added, as required to satisfy the valence of each atom.
It is to be understood that all of the compounds of the invention described above will further include bonds between adjacent atoms and/or hydrogens as required to satisfy the valence of each atom. That is, bonds and/or hydrogen atoms are added to provide the following number of total bonds to each of the following types of atoms: carbon: four bonds; nitrogen: three bonds; oxygen: two bonds; and sulfur: two bonds.
Groups that are "optionally substituted" are unsubstituted or are substituted by other than hydrogen at one or more available positions, typically 1, 2, 3, 4 or 5 positions, by one or more suitable groups (which may be the same or different). Optional substitution is also indicated by the phrase "substituted with from 0 to X substituents," where X is the maximum number of possible substituents. Certain optionally substituted groups are substituted with from 0 to 2, 3 or 4 independently selected substituents (i.e., are unsubstituted or substituted with up to the recited maximum number of substitutents).
It will be noted that the structures of some of the compounds of this invention include asymmetric carbon atoms. It is to be understood accordingly that the isomers arising from such asymmetry (e.g., all enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates) are included within the scope of this invention. Such isomers can be obtained in substantially pure form by classical separation techniques and by stereochemical^ controlled synthesis. Furthermore, the structures and other compounds and moieties discussed in this application also include all tautomers thereof. Compounds described herein may be obtained through art recognized synthesis strategies. It will also be noted that the substituents of some of the compounds of this invention include isomeric cyclic structures. It is to be understood accordingly that constitutional isomers of particular substituents are included within the scope of this invention, unless indicated otherwise. For example, the term "tetrazole" includes tetrazole, 2H-tetrazole, 3H- tetrazole, 4H-tetrazole and 5H-tetrazole.
Use in HCV-associated disorders
The compounds of the present invention have valuable pharmacological properties and are useful in the treatment of diseases. In certain embodiments, compounds of the invention are useful in the treatment of ΗCV-associated disorders, e.g., as drugs to treat HC V infection.
The term "use" includes any one or more of the following embodiments of the invention,, respectively: the use in the treatment of HCV-associated disorders; the use for the manufacture of pharmaceutical compositions for use in the treatment of these diseases, e.g., in the manufacture of a medicament; methods of use of compounds of the invention in the treatment of these diseases; pharmaceutical preparations having compounds of the invention for the treatment of these diseases; and compounds of the invention for use in the treatment of these diseases; as appropriate and expedient, if not stated otherwise. In particular, diseases to be treated and are thus preferred for use of a compound of the present invention are selected from HCV-associated disorders, including those corresponding to HCV-infection, as well as those diseases that depend on the activity of one or more of the NS3, NS4A, NS4B, NS5A and NS5B proteins, or a NS3-NS4A, NS4A-NS4B, NS4B-NS5A or NS5A-NS5B complex. The term "use" further includes embodiments of compositions herein which bind to an HCV protein sufficiently to serve as tracers or labels, so that when coupled to a fluor or tag, or made radioactive, can be used as a research reagent or as a diagnostic or an imaging agent. In certain embodiments, a compound of the present invention is used for treating
HCV-associated diseases, and use of the compound of the present invention as an inhibitor of any one or more HCVs. It is envisioned that a use can be a treatment of inhibiting one or more strains of HCV. Assays The inhibition of HCV activity may be measured as using a number of assays available in the art. An example of such an assay can be found in Anal Biochem. 1996 240(1): 60-7; which is incorporated by reference in its entirety. Assays for measurement of HCV activity are also described in the experimental section below. Pharmaceutical Compositions
The language "effective amount" of the compound is that amount necessary or sufficient to treat or prevent an HCV-associated disorder, e.g. prevent the various morphological and somatic symptoms of an HCV-associated disorder, and/or a disease or condition described herein. In an example, an effective amount of the HCV -modulating compound is the amount sufficient to treat HCV infection in a subject. In another example, an effective amount of the HCV-modulating compound is the amount sufficient to treat HCV infection, liver cirrhosis, chronic liver disease, hepatocellular carcinoma, cryoglobulinaemia, non-Hodgkin's lymphoma, and a suppressed innate intracellular immune response in a subject. The effective amount can vary depending on such factors as the size and weight of the subject, the type of illness, or the particular compound of the invention. For example, the choice of the compound of the invention can affect what constitutes an "effective amount." One of ordinary skill in the art would be able to study the factors contained herein and make the determination regarding the effective amount of the compounds of the invention without undue experimentation. The regimen of administration can affect what constitutes an effective amount. The compound of the invention can be administered to the subject either prior to or after the onset of an HCV-associated state. Further, several divided dosages, as well as staggered dosages, can be administered daily or sequentially, or the dose can be continuously infused, or can be a bolus injection. Further, the dosages of the compound(s) of the invention can be proportionally increased or decreased as indicated by the exigencies of the therapeutic or prophylactic situation.
Compounds of the invention may be used in the treatment of states, disorders or diseases as described herein, or for the manufacture of pharmaceutical compositions for use in the treatment of these diseases. Methods of use of compounds of the present invention in the treatment of these diseases, or pharmaceutical preparations having compounds of the present invention for the treatment of these diseases.
The language "pharmaceutical composition" includes preparations suitable for administration to mammals, e.g., humans. When the compounds of the present invention are administered as pharmaceuticals to mammals, e.g., humans, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
The phrase "pharmaceutically acceptable carrier" is art recognized and includes a pharmaceutically acceptable material, composition or vehicle, suitable for administering compounds of the present invention to mammals. The carriers include liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations.
Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions. Examples of pharmaceutically acceptable antioxidants include: water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, α-tocopherol, and the like; and metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
Formulations of the present invention include those suitable for oral, nasal, topical, transdermal, buccal, sublingual, rectal, vaginal and/or parenteral administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient that can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound that produces a therapeutic effect. Generally, out of one hundred per cent, this amount will range from about 1 per cent to about ninety-nine percent of active ingredient, preferably from about 5 per cent to about 70 per cent, most preferably from about 10 per cent to about 30 per cent.
Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention with the carrier and, optionally, one or more accessory ingredients, hi general, the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
Formulations of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient. A compound of the present invention may also be administered as a bolus, electuary or paste.
In solid dosage forms of the invention for oral administration (capsules, tablets, pills, dragees, powders, granules and the like), the active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; humectants, such as glycerol; disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; solution retarding agents, such as paraffin; absorption accelerators, such as quaternary ammonium compounds; wetting agents, such as, for example, cetyl alcohol and glycerol monostearate; absorbents, such as kaolin and bentonite clay; lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and coloring agents. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like. A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
The tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions that can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
Liquid dosage forms for oral administration of the compounds of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluent commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifϊers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
Formulations of the pharmaceutical compositions of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound. Formulations of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
Dosage forms for the topical or transdermal administration of a compound of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that may be required.
The ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
Powders and sprays can contain, in addition to a compound of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
Transdermal patches have the added advantage of providing controlled delivery of a compound of the present invention to the body. Such dosage forms can be made by dissolving or dispersing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the active compound in a polymer matrix or gel.
Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention. Pharmaceutical compositions of this invention suitable for parenteral administration comprise one or more compounds of the invention in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
Examples of suitable aqueous and nonaqueous carriers that may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. These compositions may also contain adjuvants such as preservatives, wetting agents,emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents that delay absorption such as aluminum monostearate and gelatin.
In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally-administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
Injectable depot forms are made by forming microencapsule matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions that are compatible with body tissue. The preparations of the present invention may be given orally, parenterally, topically, or rectally. They are of course given by forms suitable for each administration route. For example, they are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, etc., administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories. Oral administration is preferred.
The phrases "parenteral administration" and "administered parenterally" as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
The phrases "systemic administration," "administered systemically," "peripheral administration" and "administered peripherally" as used herein mean the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient's system and, thus, is subject to metabolism and other like processes, for example^ subcutaneous administration.
These compounds may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracisternally and topically, as by powders, ointments or drops, including buccally and sublingually.
Regardless of the route of administration selected, the compounds of the present invention, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art. Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
The selected dosage level will depend upon a variety of factors including the activity of the particular compound of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
In general, a suitable daily dose of a compound of the invention will be that amount of the compound that is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above. Generally, intravenous and subcutaneous doses of the compounds of this invention for a patient, when used for the indicated analgesic effects, will range from about 0.0001 to about 100 mg per kilogram of body weight per day, more preferably from about 0.01 to about 50 mg per kg per day, and still more preferably from about 1.0 to about 100 mg per kg per day. An effective amount is that amount treats an HCV-associated disorder. If desired, the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
. While it is possible for a compound of the present invention to be administered alone, it is preferable to administer the compound as a pharmaceutical composition. Synthetic Procedure
Compounds of the present invention are prepared from commonly available compounds using procedures known to those skilled in the art, including any one or more of the following conditions without limitation:
Within the scope of this text, only a readily removable group that is not a constituent of the particular desired end product of the compounds of the present invention is designated a "protecting group," unless the context indicates otherwise. The protection of functional groups by such protecting groups, the protecting groups themselves, and their cleavage reactions are described for example in standard reference works, such as e.g., Science of Synthesis: Houben-Weyl Methods of Molecular Transformation. Georg Thieme Verlag, Stuttgart, Germany. 2005. 41627 pp. (URL: http://www.science-of-synthesis.com (Electronic Version, 48 Volumes)); J. F. W. McOmie, "Protective Groups in Organic Chemistry", Plenum Press, London and New York 1973, in T. W. Greene and P. G. M. Wuts, "Protective Groups in Organic Synthesis", Third edition, Wiley, New York 1999, in "The Peptides"; Volume 3 (editors: E. Gross and J. Meienhofer), Academic Press, London and New York 1981, in "Methoden der organischen Chemie" {Methods of Organic Chemistry), Houben Weyl, 4th edition, Volume 15/1, Georg Thieme Verlag, Stuttgart 1974, in H.-D. Jakubke and H. Jeschkeit, "Aminosauren, Peptide, Proteine" {Amino acids. Peptides, Proteins), Verlag Chemie, Weinheim, Deerfield Beach, and Basel 1982, and in Jochen Lehmann, "Chemie der Kohlenhydrate: Monosaccharide und Derivate" {Chemistry of Carbohydrates: Monosaccharides and Derivatives), Georg Thieme Verlag, Stuttgart 1974. A characteristic of protecting groups is that they can be removed readily {i.e., without the occurrence of undesired secondary reactions) for example by solvolysis, reduction, photolysis or alternatively under physiological conditions {e.g., by enzymatic cleavage). ' Salts of compounds of the present invention having at least one salt- forming group may be prepared in a manner known per se. For example, salts of compounds of the present invention having acid groups may be formed, for example, by treating the compounds with metal compounds, such as alkali metal salts of suitable organic carboxylic acids, e.g., the sodium salt of 2-ethylhexanoic acid, with organic alkali metal or alkaline earth metal compounds, such as the corresponding hydroxides, carbonates or hydrogen carbonates, such as sodium or potassium hydroxide, carbonate or hydrogen carbonate, with corresponding calcium compounds or with ammonia or a suitable organic amine, stoichiometric amounts or only a small excess of the salt-forming agent preferably being used. Acid addition salts of compounds of the present invention are obtained in customary manner, e.g., by treating the compounds with an acid or a suitable anion exchange reagent. Internal salts of compounds of the present invention containing acid and basic salt-forming groups, e.g., a free carboxy group and a free amino group, may be formed, e.g., by the neutralisation of salts, such as acid addition salts, to the isoelectric point, e.g., with weak bases, or by treatment with ion exchangers. Salts can be converted in customary manner into the free compounds; metal and ammonium salts can be converted, for example, by treatment with suitable acids, and acid addition salts, for example, by treatment with a suitable basic agent.
Mixtures of isomers obtainable according to the invention can be separated in a manner known per se into the individual isomers; diastereoisomers can be separated, for example, by partitioning between polyphasic solvent mixtures, recrystallisation and/or chromatographic separation, for example over silica gel or by, e.g., medium pressure liquid chromatography over a reversed phase column, and racemates can be separated, for example, by the formation of salts with optically pure salt-forming reagents and separation of the mixture of diastereoisomers so obtainable, for example by means of fractional crystallisation, or by chromatography over optically active column materials.
Intermediates and final products can be worked up and/or purified according to standard methods, e.g., using chromatographic methods, distribution methods, (re-) crystallization, and the like. General process conditions
The following applies in general to all processes mentioned throughout this disclosure.
The process steps to synthesize the compounds of the invention can be carried out under reaction conditions that are known per se, including those mentioned specifically, in the absence or, customarily, in the presence of solvents or diluents, including, for example, solvents or diluents that are inert towards the reagents used and dissolve them, in the absence or presence of catalysts, condensation or neutralizing agents, for example ion exchangers, such as cation exchangers, e.g., in the H+ form, depending on the nature of the reaction and/or of the reactants at reduced, normal or elevated temperature, for example in a temperature range of from about -100 0C to about 1900C, including, for example, from approximately - 800C to approximately 1500C, for example at from -80 to -600C, at room temperature, at from -20 to 400C or at reflux temperature, under atmospheric pressure or in a closed vessel, where appropriate under pressure, and/or in an inert atmosphere, for example under an argon or nitrogen atmosphere. At all stages of the reactions, mixtures of isomers that are formed can be separated into the individual isomers, for example diastereoisomers or enantiomers, or into any desired mixtures of isomers, for example racemates or mixtures of diastereoisomers, for example analogously to the methods described in Science of Synthesis: Houben-Weyl Methods of Molecular Transformation. Georg Thieme Verlag, Stuttgart, Germany. 2005. The solvents from which those solvents that are suitable for any particular reaction may be selected include those mentioned specifically or, for example, water, esters, such as lower alkyl-lower alkanoates, for example ethyl acetate, ethers, such as aliphatic ethers, for example diethyl ether, or cyclic ethers, for example tetrahydrofurane or dioxane, liquid aromatic hydrocarbons, such as benzene or toluene, alcohols, such as methanol, ethanol or 1- or 2-propanol, nitriles, such as acetonitrile, halogenated hydrocarbons, such as methylene chloride or chloroform, acid amides, such as dimethylformamide or dimethyl acetamide, bases, such as heterocyclic nitrogen bases, for example pyridine or N-methylpyrrolidin-2- one, carboxylic acid anhydrides, such as lower alkanoic acid anhydrides, for example acetic anhydride, cyclic, linear or branched hydrocarbons, such as cyclohexane, hexane or isopentane, or mixtures of those solvents, for example aqueous solutions, unless otherwise indicated in the description of the processes. Such solvent mixtures may also be used in working up, for example by chromatography or partitioning.
The compounds, including their salts, may also be obtained in the form of hydrates, or their crystals may, for example, include the solvent used for crystallization. Different crystalline forms may be present.
The invention relates also to those forms of the process in which a compound obtainable as an intermediate at any stage of the process is used as starting material and the remaining process steps are carried out, or in which a starting material is formed under the reaction conditions or is used in the form of a derivative, for example in a protected form or in the form of a salt, or a compound obtainable by the process according to the invention is produced under the process conditions and processed further in situ. Pro-drugs
The present invention also relates to pro-drugs of a compound of the present invention that are converted in vivo to the compounds of the present invention as described herein. Any reference to a compound of the present invention is therefore to be understood as referring, also to the corresponding pro-drugs of the compound of the present invention, as appropriate and expedient. Combinations A compound of the present invention may also be used in combination with other agents, e.g., an additional HCV-modulating compound that is or is not of the formula I, for treatment of and HCV-associated disorder in a subject.
By the term "combination", is meant either a fixed combination in one dosage unit form, or a kit of parts for the combined administration where a compound of the present invention and a combination partner may be administered independently at the same time or separately within time intervals that especially allow that the combination partners show a cooperative, e.g., synergistic, effect, or any combination thereof.
For example, WO 2005/042020, incorporated herein by reference in its entirety, describes the combination of various HCV inhibitors with a cytochrome P450 ("CYP") inhibitor. Any CYP inhibitor that improves the pharmacokinetics of the relevant NS3/4A protease may be used in combination with the compounds of this invention. These CYP inhibitors include, but are not limited to, ritonavir (WO 94/14436, incorporated herein by reference in its entirety), ketoconazole, ^-oleandomycin, 4-methyl pyrazole, cyclosporin, clomethiazole, cimetidine, itraconazole, fluconazole, miconazole, fluvoxamine, fluoxetine, nefazodone, sertraline, indinavir, nelfinavir, amprenavir, fosamprenavir, saquinavir, lopinavir, delavirdine, erythromycin, VX-944, and VX-497. Preferred CYP inhibitors include ritonavir, ketoconazole, troleandomycin, 4-methyl pyrazole, cyclosporin, and clomethiazole. Methods for measuring the ability of a compound to inhibit CYP activity are known
(see, e.g., US 6,037,157 and Yun, et al. Drug Metabolism & Disposition, vol. 21, pp. 403-407 (1993); incorporated herein by reference). For example, a compound to be evaluated may be incubated with 0.1, 0.5, and 1.0 mg protein/ml, or other appropriate concentration of human hepatic microsomes (e. g., commercially available, pooled characterized hepatic microsomes) for 0, 5, 10, 20, and 30 minutes, or other appropriate times, in the presence of an NADPH- generating system. Control incubations may be performed in the absence of hepatic microsomes for 0 and 30 minutes (triplicate). The samples may be analyzed for the presence of the compound. Incubation conditions that produce a linear rate of compound metabolism will be used a guide for further studies. Experiments known in the art can be used to determine the kinetics of the compound metabolism (Kn, and Vm3x). The rate of disappearance of compound may be determined and the data analyzed according to Michaelis-Menten kinetics by using Lineweaver-Burk, Eadie-Hofstee, or nonlinear regression analysis.
Inhibition of metabolism experiments may then be performed. For example, a compound (one concentration, < Km) may be incubated with pooled human hepatic microsomes in the absence or presence of a CYP inhibitor (such as ritonavir) under the conditions determined above. As would be recognized, control incubations should contain the same concentration of organic solvent as the incubations with the CYP inhibitor. The concentrations of the compound in the samples may be quantitated, and the rate of disappearance of parent compound may be determined, with rates being expressed as a percentage of control activity.
Methods for evaluating the influence of co-administration of a compound of the invention and a CYP inhibitor in a subject are also known (see, e.g., US2004/0028755; incorporated herein by reference). Any such methods could be used in connection with this invention to determine the pharmacokinetic impact of a combination. Subjects that would benefit from treatment according to this invention could then be selected.
Accordingly, one embodiment of this invention provides a method for administering an inhibitor of CYP3A4 and a compound of the invention. Another embodiment of this invention provides a method for administering an inhibitor of isozyme 3A4 ("CYP3A4"), isozyme 2Cl 9 ("CYP2C19"), isozyme 2D6 ("CYP2D6"), isozyme 1 A2 ("CYP 1A2"), isozyme 2C9 ("CYP2C9"), or isozyme 2El ("CYP2E1"). In embodiments where the protease inhibitor is VX-950 (or a sterereoisomer thereof), the CYP inhibitor preferably inhibits CYP3A4. As would be appreciated, CYP3A4 activity is broadly observed in humans.
Accordingly, embodiments of this invention involving inhibition of isozyme 3A4 would be expected to be applicable to a broad range of patients.
Accordingly, this invention provides methods wherein the CYP inhibitor is . administered together with the compound of the invention in the same dosage form or in separate dosage forms.
The compounds of the invention (e.g., compound of Formula I or subformulae thereof) may be administered as the sole ingredient or in combination or alteration with other antiviral agents, especially agents active against HCV. In combination therapy, effective dosages of two or more agents are administered together, whereas in alternation or sequential-step therapy, an effective dosage of each agent is administered serially or sequentially. In general, combination therapy is typically preferred over alternation therapy because it induces multiple simultaneous stresses on the virus. The dosages given will depend on absorption, inactivation and excretion rate of the drug as well as other factors. It is to be noted that dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens and schedules should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions. The efficacy of a drug against the viral infection can be prolonged, augmented, or restored by administering the compound in combination or alternation with a second, and perhaps third antiviral compound that induces a different gene mutation than that caused by the principle drug in a drug resistant virus. Alternatively, the pharmacokinetic, biodistribution or other parameters of the drug can be altered by such combination or alternation therapy.
Daily dosages required in practicing the method of the present invention will vary depending upon, for example, the compound of the invention employed , the host, the mode of administration, the severity of the condition to be treated. A preferred daily dosage range is about from 1 to 50 mg/kg per day as a single dose or in divided doses. Suitable daily dosages for patients are on the order of from e.g. 1 to 20 mg/kg p.o or i.v. Suitable unit dosage forms for oral administration comprise from ca. 0.25 to 10 mg/kg active ingredient, e.g. compound of Formula I or any subformulae thereof, together with one or more pharmaceutically acceptable diluents or carriers therefor. The amount of co-agent in the dosage form can vary greatly, e.g., 0.00001 to 1000mg/kg active ingredient.
Daily dosages with respect to the co-agent used will vary depending upon, for example, the compound employed, the host, the mode of administration and the severity of the condition to be treated. For example, lamivudine may be administered at a daily dosage of lOOmg. The pegylated interferon may be administered parenterally one to three times per week, preferably once a week, at a total weekly dose ranging from 2 to 10 million IU, more preferable 5 to 10 million IU, most preferable 8 to 10 million IU. Because of the diverse types of co-agent that may be used, the amounts can vary greatly, e.g., .0001 to 5,000 mg/kg per day.
The current standard of care for treating hepatitis C is the combination of pegylated interferon alpha with ribavirin, of which the recommended doses are 1.5 μg/kg/wk peginterferon alfa-2b or 180 μg/wk peginterferon alfa-2a, plus 1,000 to 1,200 mg daily of ribavirin for 48 weeks for genotype I patients, or 800 mg daily of ribavirin for 24 weeks for genotype 2/3 patients.
The compound of the invention (e.g., compound of Formula I or subformulae thereof) and co-agents of the invention may be administered by any conventional route, in particular enterally, e.g. orally, for example in the form of solutions for drinking, tablets or capsules or parenterally, for example in the form of injectable solutions or suspensions. Certain preferred pharmaceutical compositions may be e.g. those based on microemulsions as described in UK 2,222,770 A.
The compound of the invention (e.g., compound of Formula I or subformulae thereof) are administered together with other drugs (co-agents) e.g. a drug which has anti-viral activity, especially anti-Flaviviridae activity, most especially anti-HCV activity, e.g. an interferon, e.g. interferon-α-2a or interferon-α-2b, e.g. IntronR A, RoferonR, AvonexR, Rebif^ or BetaferonR, or an interferon conjugated to a water soluble polymer or to human albumin, e.g. albuferon, an anti-viral agent, e.g. ribavirin, lamivudine, the compounds disclosed in US patent no. 6,812,219 and WO 2004/002422 A2 (the disclosures of which are incorporated herein by reference in their entireties), an inhibitor of the HCV or other Flaviviridae virus encoded factors like the NS3/4A protease, helicase or RNA polymerase or a prodrug of such an inhibitor, an anti-fibrotic agent, e.g. a N-phenyl-2-pyrimidine-amine derivative, e.g. imatinib, an immune modulating agent, e.g. mycophenolic acid, a salt or a prodrug thereof, e.g. sodium mycophenolate or mycophenolate mofetil, or a SlP receptor agonist, e.g. FTY720 or an analogue thereof optionally phosphorylated, e.g. as disclosed in EP627406A1, EP778263A1, EP1002792A1, WO02/18395, WO02/76995, WO 02/06268, JP2002316985, WO03/29184, WO03/29205, WO03/62252 and WO03/62248, the disclosures of which are incorporated herein by reference in their entireties.
Conjugates of interferon to a water-soluble polymer are meant to include especially conjugates to polyalkylene oxide homopolymers such as polyethylene glycol (PEG) or polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereof. As an alternative to polyalkylene oxide-based polymers, effectively non- antigenic materials such as dextran, polyvinyl pyrrolidones, polyacrylamides, polyvinyl alcohols, carbohydrate-based polymers and the like can be used. Such interferon-polymer conjugates are described in U.S. Pat. Nos. 4,766,106, 4,917,888, European Patent Application No. 0 236 987, European Patent Application No. 0 510 356 and International Application Publication No. WO 95/13090, the disclosures of which are incorporated herein by reference in their entireties. Since the polymeric modification sufficiently reduces antigenic responses, the foreign interferon need not be completely autologous. Interferon used to prepare polymer conjugates may be prepared from a mammalian extract, such as human, ruminant or bovine interferon, or recombinantly produced. Preferred are conjugates of interferon to polyethylene glycol, also known as pegylated interferons.
, Especially preferred conjugates of interferon are pegylated alfa-interferons, for example pegylated interferon- a -2a, pegylated interferon- a -2b; pegylated consensus interferon or pegylated purified interferon-α product. Pegylated interferon- a -2a is described e.g. in European Patent 593,868 (incorporated herein by reference in its entirety) and commercially available e. g. under the tradename PEGASYS® (Hoffmann-La Roche). Pegylated interferon-α-2b is described, e.g. in European Patent 975,369 (incorporated herein by reference in its entirety) and commercially available e.g. under the tradename PEG- INTRON A® (Schering Plough). Pegylated consensus interferon is described in WO 96/11953 (incorporated herein by reference in its entirety). The preferred pegylated a- interferons are pegylated interferon-α-2a and pegylated interferon-α-2b. Also preferred is pegylated consensus interferon.
Other preferred co-agents are fusion proteins of an interferon, for example fusion proteins of interferon-α-2a, interferon-Q!-2b; consensus interferon or purified interferon-α product, each of which is fused with another protein. Certain preferred fusion proteins comprise an interferon (e.g., interferon-α-2b) and an albumin as described in U.S. Patent 6,973,322 and international publications WO02/60071, WO05/003296 and WO05/077042 (Human Genome Sciences). A preferred interferon conjugated to a human albumin is Albuferon (Human Genome Sciences).
Cyclosporins which bind strongly to cyclophilin but are not immunosuppressive include those cyclosporins recited in U.S. Patents 5,767,069 and 5,981,479 and are incorporated herein by reference. Melle4-Cyclosporin is a preferred non-immunosuppressive cyclosporin. Certain other cyclosporin derivatives are described in WO2006039668 (Scynexis) and WO2006038088 (Debiopharm SA) and are incorporated herein by reference. A cyclosporin is considered to be non-immunosuppressive when it has an activity in the Mixed Lymphocyte Reaction (MLR) of no more than 5%, preferably no more than 2%, that of cyclosporin A. The Mixed Lymphocyte Reaction is described by T. Meo in
"Immunological Methods", L. Lefkovits and B. Peris, Eds., Academic Press, N. Y. pp. 227 - 239 (1979). Spleen cells (0.5 x 106) from Balb/c mice (female, 8 - 10 weeks) are co- incubated for 5 days with 0.5 x 106 irradiated (2000 rads) or mitomycin C treated spleen cells from CBA mice (female, 8 - 10 weeks). The irradiated allogeneic cells induce a proliferative response in the BaIb c spleen cells which can be measured by labeled precursor incorporation into the DNA. Since the stimulator cells are irradiated (or mitomycin C treated) they do not respond to the Balb/c cells with proliferation but do retain their antigenicity. The IC5O found for the test compound in the MLR is compared with that found for cyclosporin A in a parallel experiment. In addition, non-immunosuppressive cyclosporins lack the capacity of inhibiting CN and the downstream NF-AT pathway. [Melle]4-ciclosporin is a preferred non- immunosuppressive cyclophilin-binding cyclosporin for use according to the invention.
Ribavirin (l-/3-D-ribofuranosyl-l-l,2,4-triazole-3-caroxamide) is a synthetic, non- interferon-inducing, broad spectrum antiviral nucleoside analog sold under the trade name, Virazole (The Merk Index, 11th edition, Editor: Budavar, S, Merck & Co., Inc., Rahway, NJ, pl304,1989). United States Patent No. 3,798,209 and RE29,835 (incorporated herein by reference in their entireties) disclose and claim ribavirin. Ribavirin is structurally similar to guanosine, and has in vitro activity against several DNA and RNA viruses including Flaviviήdae (Gary L. Davis, Gastroenterology 118:S1O4-S114, 2000).
Ribavirin reduces serum amino transferase levels to normal in 40% of patients, but it does not lower serum levels of HCV-RNA (Gary L. Davis, Gastroenterology 118:S1O4-S114, 2000). Thus, ribavirin alone is not effective in reducing viral RNA levels. Additionally, ribavirin has significant toxicity and is known to induce anemia. Ribavirin is not approved for monotherapy against HCV; it is approved in combination with interferon alpha-2a or interferon alpha-2b for the treatment of HCV. A further preferred combination is a combination of a compound of the invention (e.g., a compound of Formula I or any sub formulae thereof) with a non-immunosuppressive cyclophilin-binding cyclosporine, with mycophenolic acid, a salt or a prodrug thereof, and/or with a SlP receptor agonist, e.g. FTY720. Additional examples of compounds that can be used in combination or alternation treatments include:
(1) Interferons, including interferon alpha 2a or 2b and pegylated (PEG) interferon alpha 2a or 2b, for example:
(a) Intron-A®, interferon alfa-2b (Schering Corporation, Kenilworth, NJ); (b) PEG-Intron®, peginteferon alfa-2b (Schering Corporation, Kenilworth, NJ);
(c) Roferon®, recombinant interferon alfa-2a (Hoffmann-La Roche, Nutley, NJ);
(d) Pegasys®, peginterferon alfa-2a (Hoffmann-La Roche, Nutley, NJ);
(e) Berefor®, interferon alfa 2 available (Boehringer Ingelheim Pharmaceutical, Inc., Ridgefield, CT); (f) Sumiferon®, a purified blend of natural alpha interferons (Sumitomo, Japan)
(g) Wellferon®, lymphoblastoid interferon alpha nl (GlaxoSmithKline); . •■ (h) Infergen®, consensus alpha interferon (InterMune Pharmaceuticals, Inc., Brisbane, CA);
(i) Alferon®, a mixture of natural alpha interferons (Interferon Sciences, and Purdue Frederick Co., CT);
(j) Viraferon®;
(k) Consensus alpha interferon from Amgen, Inc., Newbury Park, CA, Other forms of interferon include: interferon beta, gamma, tau and omega, such as Rebif ( Interferon beta Ia) by Serono, Omniferon (natural interferon) by Viragen, REBIF (interferon beta- Ia) by Ares-Serono, Omega Interferon by BioMedicines; oral Interferon Alpha by Amarillo Biosciences; an interferon conjugated to a water soluble polymer or to a human albumin, e.g., Albuferon (Human Genome Sciences), an antiviral agent, a consensus interferon, ovine or bovine interferon-tau
Conjugates of interferon to a water-soluble polymer are meant to include especially conjugates to polyalkylene oxide homopolymers such as polyethylene glocol (PEG) or polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereof. As an alternative to polyalkylene oxid-based polymers, effectively non- antigenic materials such as dextran, polyvinyl pyrrolidones, polyacrylamides, polyvinyl alcohols, carbohydrate-based polymers and the like can be used. Since the polymeric modification sufficiently reduces antigenic response, the foreign interferon need not be completely autologous. Interferon used to prepare polymer conjugates may be prepared from a mammalian extract, such as human, ruminant or bovine interferon, or recombinantly produced. Preferred are conjugates of interferon to polyethylene glycol, also known as pegylated interferons.
(2) Ribavirin, such as ribavirin (l-beta-D-ribofuranosyl-lH-l,2,4-triazole-3- carboxamide) from Valeant Pharmaceuticals, Inc., Costa Mesa, CA); Rebetol® from Schering Corporation, Kenilworth, NJ, and Copegus® from Hoffmann-La Roche, Nutley, NJ; and new ribavirin analogues in development such as Levovirin and Viramidine by Valeant, (3) Thiazolidine derivatives which show relevant inhibition in a reverse-phase HPLC assay with an NS3/4A fusion protein and NS5A/5B substrate (Sudo K. et al., Antiviral Research, 1996, 32, 9-18), especially compound RD- 1-6250, possessing a fused cinnamoyl moiety substituted with a long alkyl chain, RD4 6205 and RD4 6193;
(4) Thiazolidines and benzanilides identified in Kakiuchi N. et al. J. FEBS Letters 421, 217-220; Takeshita N. et al. Analytical Biochemistry, 1997, 247, 242-246;
(5) A phenan-threnequinone possessing activity against protease in a SDS-PAGE and autoradiography assay isolated from the fermentation culture broth of Streptomyces sp., Sch 68631 (Chu M. et al., Tetrahedron Letters, 1996, 37, 7229-7232), and Sch 351633, isolated from the fungus Penicillium griseofulvum, which demonstrates activity in a scintillation proximity assay (Chu M. et al, Bioorganic and Medicinal Chemistry Letters 9, 1949-1952);
(6) Protease inhibitors.
Examples include substrate-based NS3 protease inhibitors (Attwood et al., Antiviral peptide derivatives, PCT WO 98/22496, 1998; Attwood et al., Antiviral Chemistry and Chemotherapy 1999, 10, 259-273; Attwood et al, Preparation and use of amino acid derivatives as anti-viral agents, German Patent Pub. DE 19914474; Tung et al. Inhibitors of serine proteases, particularly hepatitis C virus NS3 protease; PCT WO 98/17679), including alphaketoamides and hydrazinoureas, and inhibitors that terminate in an electrophile such as a boronic acid or phosphonate (Llinas-Brunet et al. Hepatitis C inhibitor peptide analogues, PCT WO 99/07734) are being investigated. Non-substrate-based NS3 protease inhibitors such as 2,4,6-trihydroxy-3-nitro- benzamide derivatives (Sudo K. et al., Biochemiscal and Biophysical Research Communications, 1997, 238 643-647; Sudo K. et al. Antiviral Chemistry and Chemotherapy, 1998, 9, 186), including RD3-4082 and RD3-4078, the former substituted on the amide with a 14 carbon chain and the latter processing a/?αrø-phenoxyphenyl group are also being investigated.
Sch 68631, a phenanthrenequinone, is an HCV protease inhibitor (Chu M et al., Tetrahedron Letters 37:7229-7232, 1996). In another example by the same authors, Sch 351633, isolated from the fungus Penicillium grieofulvum, was identified as a protease inhibitor (Chu M. et al., Bioorganic and Medicinal Chemistry Letters 9:1949-1952). Nanomolar potency against the HCV NS3 protease enzyme has been achieved by the design of selective inhibitors based on the macromolecule eglin c. Eglin c, isolated from leech, is a potent inhibitor of several serine proteases such as S. griseus proteases A and B, V- chymotrypsin, chymase and subtilisin. Qasim M.A. et al., Biochemistry 36:1598-1607, 1997.
U.S. patents disclosing protease inhibitors for the treatment of HCV include, for example, U.S. Patent No. 6,004,933 to Spruce et al (incorporated herein by reference in its entirety) which discloses a class of cysteine protease inhibitors for inhibiting HCV endopeptidase 2; U.S. Patent No. 5,990,276 to Zhang et al. (incorporated herein by reference in its entirety) which discloses synthetic inhibitors of hepatitis C virus NS3 protease; U.S. Patent No. 5,538,865 to Reyes et al.(incorporated herein by reference in its entirety). Peptides as NS3 serine protease inhibitors of HCV are disclosed in WO 02/008251 to Corvas International, Inc., and WO 02/08187 and WO 02/008256 to Schering Corporation (incorporated herein by reference in their entireties). HCV inhibitor tripeptides are disclosed in U.S. Patent Nos. 6,534,523, 6,410,531 and 6,420,380 to Boehringer Ingelheim and WO 02/060926 to Bristol Myers Squibb (incorporated herein by reference in their entireties). Diaryl peptides as NS3 serine protease inhibitors of HCV are disclosed in WO 02/48172 to Schering Corporation (incorporated herein by reference). Imidazoleidinones as NS3 serine protease inhibitors of HCV are disclosed in WO 02/18198 to Schering Corporation and WO 02/48157 to Bristol Myers Squibb (incorporated herein by reference in their entireties). WO 98/17679 to Vertex Pharmaceuticals and WO 02/48116 to Bristol Myers Squibb also disclose HCV protease inhibitors (incorporated herein by reference in their entireties).
HCV NS3-4A serine protease inhibitors including BILN 2061 by Boehringer Ingelheim, VX-950 by Vertex, SCH 6/7 by Schering-Plough, and other compounds currently in preclinical development;
Substrate-based NS3 protease inhibitors, including alphaketoamides and hydrazinoureas, and inhibitors that terminate in an elecrophile such as a boronic acid or phosphonate; Non-substrate-based NS3 protease inhibitors such as 2,4,6-trihydroxy-3-nitro- benzamide derivatives including RD3-4082 and RD3-4078, the former substituted on the amide with a 14 carbon chain and the latter processing a para-phenoxyphenyl group; and Sch68631 , a phenanthrenequinone, an HCV protease inhibitor.
Sch 351633, isolated from the fungus Penicillium griseofulvum was identified as a protease inhibitor. EgHn c, isolated from leech is a potent inhibitor of several serine proteases such as S. griseus proteases A and B, a-chymotrypsin, chymase and subtilisin.
US patent no. 6004933 (incorporated herein by reference in its entirety) discloses a class of cysteine protease inhibitors from inhibiting HCV endopeptidase 2; synthetic inhibitors of HCV NS3 protease (pat), HCV inhibitor tripeptides (pat), diaryl peptides such as NS3 serine protease inhibitors of HCV (pat), Imidazolidindiones as NS3 serine protease inhibitors of HCV (pat).
Thiazolidines and benzanilides (ref). Thiazolidine derivatives which show relevant inhibition in a reverse-phase HPLC assay with an NS3/4A fusion protein and NS5A/5B substrate especially compound RD- 16250 possessing a fused cinnamoyl moiety substituted with a long alkyl chain, RD4 6205 and RD4 6193 Phenan-threnequinone possessing activity against protease in a SDS-PAGE and autoradiography assay isolated from the fermentation culture broth of Streptomyces sp, Sch68631 and Sch351633, isolated from the fungus Penicillium griseofulvum, which demonstrates activity in a scintillation proximity assay. . . •
(7) Nucleoside or non-nucleoside inhibitors of HCV NS5B RNA-dependent RNA polymerase, such as 2'-C-methyl-3'-O-L-valine ester ribofuranosyl cytidine (Idenix) as disclosed in WO 2004/002422 A2 (incorporated herein by reference in its entirety), R803 (Rigel), JTK-003 (Japan Tabacco), HCV-086 (ViroPharma/Wyeth) and other compounds currently in preclinical development; gliotoxin (ref) and the natural product cerulenin; 2'-fluoronucleosides; other nucleoside analogues as disclosed in WO 02/057287 A2, WO 02/057425 A2, WO 01/90121, WO 01/92282, and US patent no. 6,812,219, the disclosures of which are incorporated herein by reference in their entirety.
Idenix Pharmaceuticals discloses the use of branched nucleosides in the treatment of flaviviruses (including HCV) and pestiviruses in International Publication Nos. WO 01/90121 and WO 01/92282 (incorporated herein by reference in their entireties). Specifically, a method for the treatment of hepatitis C infection (and flaviviruses and pestiviruses) in humans and other host animals is disclosed in the Idenix publications that includes administering an effective amount of a biologically active 1 ', 2', 3' or 4'-branced B- D or B-L nucleosides or a pharmaceutically acceptable salt or prodrug thereof, administered either alone or in combination with another antiviral agent, optionally in a pharmaceutically acceptable carrier. Certain preferred biologically active 1', 2', 3', or 4' branched B-D or B-L nucleosides, including Telbivudine, are describedi n U.S. Patents 6,395,716 and 6,875,751, each of which are incorporated herein by reference.
Other patent applications disclosing the use of certain nucleoside analogs to treat hepatitis C virus include: PCTCAOO/01316 (WO 01/32153; filed November 3, 2000) and PCT/CAOl/00197 (WO 01/60315; filed February 19, 2001) filed by BioChem Pharma, Inc., (now Shire Biochem, Inc.); PCT/US02/01531 (WO 02/057425; filed January 18, 2002) and PCT/US02/03086 (WO 02/057287; filed January 18, 2002) filed by Merck & Co., Inc., PCT/EP01/09633 (WO 02/18404; published August 21, 2001) filed by Roche, and PCT Publication Nos. WO 01/79246 (filed April 13, 2001), WO 02/32920 (filed October 18, 2001) and WO 02/48165 by Pharmasset, Ltd. (the disclosures of which are incorporated herein by reference in their entireties) PCT Publication No. WO 99/43691 to Emory University (incorporated herein by reference in its entirety), entitled "2'-Fluoronucleosides" discloses the use of certain 2'- fluoronucleosides to treat HCV.
Eldrup et al. (Oral Session V, Hepatitis C Virus, Flaviviridae; 16th International Conference on Antiviral Research (April 27, 2003, Savannah, GA)) described the structure activity relationship of 2'-modifϊed nucleosides for inhibition of HCV.
Bhat et al. (Oral Session V, Hepatitis C Virus, Flaviviridae, 2003 (Oral Session V, Hepatitis C Virus, Flaviviridae; 16th International conference on Antiviral Research (April 27, 2003, Savannah, Ga); p A75) describes the synthesis and pharmacokinetic properties of nucleoside analogues as possible inhibitors of HCV RNA replication. The authors report that 2 '-modified nucleosides demonstrate potent inhibitory activity in cell-based replicon assays.
Olsen et al. (Oral Session V, Hepatitis C Virus, Flaviviridae; 16th International Conference on Antiviral Research (April 27, 2003, Savannah, Ga)p A76) also described the effects of the 2 '-modified nucleosides on HCV RNA replication.
(8) Nucleotide polymerase inhibitors and gliotoxin (Ferrari R. et al. Journal of Virology, 1999, 73, 1649-1654), and the natural product cerulenin (Lohmann V. et al.
Virology, 1998, 249, 108-118);
(9) HCV NS3 helicase inhibitors, such as VP 50406 by ViroPhama and compounds from Vertex. Other helicase inhibitors (Diana G.D. et al., Compounds, compositions and methods for treatment of hepatitis C, U.S. Patent No. 5,633,358 (incorporated herein by reference in its entirety); Diana G.D. et al., Piperidine derivatives, pharmaceutical compositions thereof and their use in the treatment of hepatitis C, PCT WO 97/36554);
(10) Antisense phosphorothioate oligodeoxynucleotides (S-ODN) complementary to sequence stretches in the 5' non-coding region (NCR) of the virus (Alt M. et al., Hepatology, 1995, 22, 707-717), or nucleotides 326-348 comprising the 31 end of the NCR and nucleotides 371-388 located in the core coding region of the HCV RNA (Alt M. et al., Archives of Virology, 1997, 142, 589-599; Galderisi U. et al., Journal of Cellular Physiology, 199, 181, 251-257); such as ISIS 14803 by Isis Pharm/Elan, antisense by Hybridon, antisense by AVI bioPharma, (11) Inhibitors of ERES-dependent translation (Dceda N et al., Agent for the prevention and treatment of hepatitis C, Japanese Patent Pub. JP-08268890; Kai Y et al. Prevention and treatment of viral diseases, Japanese Patent Pub. JP-10101591); such as ISIS 14803 by Isis Pharm/Elan, IRES inhibitor by Anadys, IRES inhibitors by Immusol, targeted RNA chemistry by PTC Therapeutics (12) Ribozymes, such as nuclease-resistant ribozymes (Maccjak, D.J. et al.,
Hepatology 1999, 30, abstract 995) and those directed in U.S. Patent No. 6,043,077 to Barber et al., and U.S. Patent Nos. 5,869,253 and 5,610,054 to Draper et al.(incorporated herein by reference in their entireties) for example, HEPTAZYME by RPI (13) siRNA directed against HCV genome (14) HCV replication inhibitor of any other mechanisms such as by
VP50406ViroPharama/Wyeth, inhibitors from Achillion, Arrow
(15) An inhibitor of other targets in the HCV life cycle including viral entry, assembly and maturation
(16) An immune modulating agent such as an IMPDH inhibitor, mycophenolic acid, a salt or a prodrug thereof sodium mycophenolate or mycophenolate mofetil, or Merimebodib
(VX-497); thymosin alpha- 1 (Zadaxin, by SciClone); or a SlP receptor agonist, e.g. FTY720 or analogue thereof optionally phosphorylated.
(17) An anti-fibrotic agent, such as a N-phenyl-2-pyrimidine- amine derivative, imatinib (Gleevac), IP-501 by Indevus, and Interferon gamma Ib from InterMune (18) Therapeutic vaccine by Intercell, Epimmune/Genecor, Merix, Tripep (Chron-
VacC), immunotherapy (Therapore) by Avant, T cell therapy by CellExSys, monoclonal antibody XTL-002 by STL, ANA 246 and ANA 246 BY Anadys,
(19) Other miscellaneous compounds including 1-amino-alkylcyclohexanes (U.S. Patent No. 6,034,134 to Gold et al.), alkyl lipids (U.S. Pat. No. 5,922,757 to Chojkier et al.), vitamin E and other antioxidants (U.S. Patent. No. 5,922,757 to Chojkier et al.), amantadine, bile acids (U.S. Pat. No. 5,846,99964 to Ozeki et al.), N-(phosphonoacetl)-L-aspartic acid, )U.S. Pat. No. 5,830,905 to Diana et al.), benzenedicarboxamides (U.S. Pat. No. 5,633,388 to Diane et al.), polyadenylic acid derivatives (U.S. Pat. No. 5,496,546 to Wang et al.), 2'3'- dideoxyinosine (U.S. Pat. No. 5,026,687 to Yarchoan et al.), benzimidazoles (U.S. Pat. No. 5,891,874 to Colacino et al.), plant extracts (U.S. Pat. No. 5,837,257 to Tsai et al., U.S. Pat. No. 5,725,859 to Omer et al., and U.S. Pat. No. 6,056,961) and piperidines (U.S. Pat. No. 5,830,905 to Diana et al.); the disclosures of which are incorporated herein by reference in their entireties. Also,squalene, telbivudine, N-(phosphonoacetyl)-L-aspartic acid, benzenedicarboxamides, polyadenylic acid derivatives, glycosylation inhibitors, and nonspecific cytoprotective agents that block cell injury caused by the virus infection.
(20) Any other compound currently in preclinical or clinical development for the treatment of HCV, including Interleukin-10 (Schering-Plough), AMANTADINE (Symmetrel) by Endo Labs Solvay, caspase inhibitor EDN-6556 by Idun Pharma, HCV/MF59 by Chiron, CIVACIR (Hepatitis C Immune Globulin) by NABI, CEPLENE (histamine dichloride) by Maxim, IDN-6556 by Idun PHARM, T67, a beta-tubulin inhibitor, by Tularik. a therapeutic vaccine directed to E2 by Innogenetics, FK788 by Fujisawa Helathcare, IdB 1016 (Siliphos, oral silybin-phosphatidyl choline phytosome), fusion inhibitor by : Trimeris, Dication by Immtech, hemopurifϊer by Aethlon Medical, UT 23 IB by United Therapeutics.
(21) Purine nucleoside analog antagonists of T1R7 (toll- like receptors) developed by Anadys, e.g., Isotorabine (ANA245) and its prodrug (ANA975), which are described in European applications EP348446 and EP636372, International Publications WO03/045968, WO05/121162 and WO05/25583, and U.S. Patent 6/973322, each of which is incorporated by reference.
(21) Non-nucleoside inhibitors developed by Genelabs and described in International Publications WO2004/108687, WO2005/12288, and WO2006/076529, each of which is incorporated by reference.
(22) Other co-agents (e.g. , non-immunomodulatory or immunomodulatory compounds) that may be used in combination with a compound of this invention include, but are not limited to, those specified in WO 02/18369, which is incorporated herein by reference.
Methods of this invention may also involve administration of another component comprising an additional agent selected from an immunomodulatory agent; an antiviral agent; an inhibitor of HCV protease; an inhibitor of another target in the HCV life cycle; a CYP inhibitor; or combinations thereof.
Accordingly, in another embodiment, this invention provides a method comprising administering a compound of the invention and another anti-viral agent, preferably an anti- HCV agent. Such anti-viral agents include, but are not limited to, immunomodulatory agents, such as a, β, and δ interferons, pegylated derivatized interferon-a compounds, and thymosin; other anti-viral agents, such as ribavirin, amantadine, and telbivudine; other inhibitors of hepatitis C proteases (NS2-NS3 inhibitors and NS3-NS4A inhibitors); inhibitors of other targets in the HCV life cycle, including helicase, polymerase, and metalloprotease inhibitors; inhibitors of internal ribosome entry; broad-spectrum viral inhibitors, such as IMPDH inhibitors (e.g., compounds of United States Patent 5,807, 876,6, 498,178, 6,344, 465,6, 054,472, WO 97/40028, WO 98/40381, WO 00/56331, and mycophenolic acid and derivatives thereof, and including, but not limited to VX-497, VX- 148, and/or VX-944); or combinations of any of the above. In accordance with the foregoing the present invention provides in a yet further aspect:
• A pharmaceutical combination comprising a) a first agent which is a compound of the invention, e.g. a compound of formula I or any subformulae thereof, and b) a co- agent, e.g. a second drug agent as defined above. • A method as defined above comprising co-administration, e.g. concomitantly or in sequence, of a therapeutically effective amount of a compound of the invention, e.g. a compound of formula I or any subformulae thereof, and a co-agent, e.g. a second drug agent as defined above.
The terms "co-administration" or "combined administration" or the like as utilized herein are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time. Fixed combinations are also within the scope of the present invention. The administration of a pharmaceutical combination of the invention results in a beneficial effect, e.g. a synergistic therapeutic effect, compared to a monotherapy applying only one of its pharmaceutically active ingredients.
Each component of a combination according to this invention may be administered separately, together, or in any combination thereof. As recognized by skilled practitioners, dosages of interferon are typically measured in IU (e.g., about 4 million IU to about 12 million IU).
If an additional agent is selected from another CYP inhibitor, the method would, therefore, employ two or more CYP inhibitors. Each component may be administered in one or more dosage forms. Each dosage form may be administered to the patient in any order.
The compound of the invention and any additional agent may be formulated in separate dosage forms. Alternatively, to decrease the number of dosage forms administered to a patient, the compound of the invention and any additional agent may be formulated together in any combination. For example, the compound of the invention inhibitor may be formulated in one dosage form and the additional agent may be formulated together in another dosage form. Any separate dosage forms may be administered at the same time or different times.
Alternatively, a composition of this invention comprises an additional agent as described herein. Each component may be present in individual compositions, combination compositions, or in a single composition.
Exemplification of the Invention
The invention is further illustrated by the following examples, which should not be construed as further limiting. The assays used throughout the Examples are accepted. Demonstration of efficacy in these assays is predictive of efficacy in subjects.
The following abbreviations are used throughout the examples and the specification.
LIST OF ABBREVIATIONS abs. Absolute
Ac acetyl
ACN Acetonitrile
AcOEt / EtOAc Ethyl acetate
AcOH acetic acid aq aqueous
Ar aryl
Bn benzyl
Bu butyl (nBu = n-butyl, tBu = tert-butyl)
CDI Carbonyldiimidazole
CH3CN Acetonitrile
DBU l,8-Diazabicyclo[5.4.0]-undec-7-ene DCE 1 ,2-Dichloroethane
DCM Dichloromethane
DIPEA N-Ethyldiisopropylamine
DMAP Dimethylaminopyridine DMF N,N'-Dimethylformamide
DMSO Dimethylsulfoxide
EI Electronspray ionisation
Et2O Diethylether
Et3N Triethylamine Ether Diethylether
EtOH Ethanol
FC Flash Chromatography h hour(s)
HATU O-(7-Azabenzotriazole-l-yl)-N,N,N'N'- tetramethyluronium hexafluorophosphate
HBTU O-(Benzotriazol-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate hexafluorophosphate
HCl Hydrochloric acid
HOBt 1 -Hydroxybenzotriazole
HPLC High Performance Liquid Chromatography
H2O Water
L liter(s)
LC-MS Liquid Chromatography Mass Spectrometry
Me methyl
MeI Iodomethane
MeOH Methanol mg milligram min minute(s) mL milliliter
MS Mass Spectrometry
Pd/C palladium on charcoal
PG protecting group
Ph phenyl Prep Preparative
Rf ratio of fronts
RP reverse phase rt Room temperature SiO2 Silica gel
TBAF Tetrabutylammonium fluoride
TEA Triethylamine
TFA Trifluoroacetic acid
THF Tetrahydrofurane TLC Thin Layer Chromatography tR Retention time
Trademarks
Hyflo = Celite® (The Celite Corporation) = filtering aid based on diatomaceous earth
Nucleosil = Nucleosil®, trademark of Machery & Nagel, Duren, FRG for
HPLC materials
Temperatures are measured in degrees Celsius. Unless otherwise indicated, the reactions take place at roome temperature.
TLC conditions: Rf values for TLC are measured on 5 x 10 cm TLC plates, silica gel F254, Merck, Darmstadt, Germany.
HPLC (method A):
Instrument: Agilent system column: Macherey-Nagel Nucleosil 100-3 C18 HD, particle size 3.5 Dm, pore size lOOA, length 70 mm, internal diameter 4 mm, flow 1.0 ml/min solvent: CH3CN (0.1% CF3CO2H); H2O (0.1% CF3CO2H) gradient: 0-6 min : 20-100% CH3CN, 1.5 min : 100% CH3CN, 0.5 min 100-20% CH3CN
HPLC (method B):
Instrument: Kontron, Kroma-System Column: Macherey-Nagel, Lichrosphere 100-5 RP 18
Solvent: CH3CN (0.1% CF3CO2H); H2O (0.1% CF3CO2H)
Gradient: 0-5 min: 10-100% CH3CN; 5-7.5 min: 100% CH3CN (Flow 1.5mL/min)
HPLC (method C):
Instrument: Agilent system column: waters symmetry Cl 8, 3.5 microm , 2.1 x 50mm , flow 0.6 ml/min solvent: CH3CN (0.1 % CF3CO2H); H2O (0.1 % CF3CO2H) gradient: 0-3.5 min : 20-95% CH3CN, 3.5-5 min : 95% CH3CN, 5.5-5.55 min 95 % to 20 % CH3CN
MS (method D):
Instrument: Agilent 1100 Series
Detection: API-ES, positive/negative
LC-MS (method E):
Instrument: Agilent system
Column: Waters symmetry, 3.5 microm , 50 x 2.1 mm, 5 min, 20% to 95% CH3CN solvent: CH3CN (0.1 % HCO2H); H2O (0.1 % HCO2H) gradient: 0-3.5 min : 20-95% CH3CN, 3.5-5 min : 95% CH3CN, 5.5-5.55 min 95 % to 20 % CH3CN
Preparative HPLC (method F): Instrument: Gilson system column: waters Cl 8 ODB, 5 microm, 50 x 19 mm solvent: CH3CN (0.1 % HCO2H); H2O (0.1 % HCO2H)
Preparative HPLC (method G): Instrument: Gilson Column: Sun-Fire prep C18 OBD 5 microm, Column 19 x 50 mm (flow 20mL/min) or Column 30 x 100 mm (flow 40mL/min) Solvent: CH3CN (0.1 % CF3CO2H) and H2O (0.1 % CF3CO2H) Gradient: 0-20 min: 5- 100% CH3CN GENERAL SYNTHETIC METHODS
Scheme 1: Keto-Sulfonamide macrocycles
Figure imgf000152_0002
Figure imgf000152_0001
BOC-De prot.
Figure imgf000152_0003
Figure imgf000152_0004
P2subs
Figure imgf000152_0005
In Scheme 1, the term "linker" refers to the L1-FG-L2-L3 residue of Formula I, the term "Pi" refers to the Ri residue of Formula I, and the term "P2subst" refers to the R5 residue of Formula I.
Scheme 2: Keto-Amide macrocycles (Synthesis of compounds in which L1-FG-L2-L3 is an alkylene-amide-alkylene residue)
Figure imgf000153_0001
in Scheme 2, the term "linker" refers to the Li-FG-L2-L3 residue of Formula I, the term "Pi" refers to the R] residue of Formula I, and the term "P2subst" refers to the R5 residue of Formula I. Example 1
(8S,1 OR)-10-(7-Methoxy-2-phenyl-quinolin-4-yIoxy)-5-[(lR,2S)-l-carbonylamino-2- vinyl-cycIopropyIJ-2,2-dioxo-2λ*6*-thia-3,6,12,22-tetraaza- tricyclo[21.4.0.0*8,12*]heptacosa-l(27),23,25-triene-4,7,13,21-tetraone
Figure imgf000154_0001
To an ice-cold solution of 250 mg (0.25 mmol) (8-{2-[((lR,2S)-l-{[(2S,4R)-4-(7-Methoxy-2- phenyl-quinolin-4-yloxy)-pyrrolidine-2-carbonyl]-amino}-2-vinyl-cyclopropanecarbonyl)- sulfamoyl] -phenyl-carbamoyl }-octanoic acid in 50 mL DCM/DMF (50:1) and 0.43 mL (2.5 mmol) of DIPEA is added 475 mg (1.3 mmol) HATU and the ice bath is removed. After stirring for 2 h the solvent is removed in vacuo and the residue is purified by preparative reverse phase HPLC (method G) to give the title compound as a colorless solid. HPLC (method A) tR = 4.78 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.5 MS (method D): 780 [M+]
Preparation of (8-{2-[((lR,2S)-l-{[(2S,4R)-4-(7-Methoxy-2-phenyl-quinolin-4-yIoxy)- pyrrolidine-2-carbonyl]-amino}-2-vinyl-cyclopropanecarbonyl)-suIfamoyl]- phenylcarbamoyl}-octanoic acid
Step 1
[(lR,2S)-l-(2-Amino-benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropyl]-carbamic acid tert-butyl ester
Figure imgf000154_0002
To a solution of 6.3 g (28 mmol) (lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl-cyclopropane- carboxylic acid (prepared according to WO 2000009558 Al) in 90 mL abs. THF is added 6.95 g (42 mmol) CDI and the mixture is refluxed for 2 h. After cooling to rt 5.1 g (29 mmol) 2-Aminobenzenesulfonamide and 6.5 g (42 mmol) DBU is added and stirring is continued for 45 min. The reaction mixture is diluted with 250 mL EtOAc and washed with 100 mL 0.5 N HCl and brine. The organic phase is dried with Na2SO4, filtered and the solvent is removed in vacuo. The residue is purified by FC on silica (eluent: CH2Cl2/Me0H 98:2) to give the title compound as a colorless solid. HPLC (method A) tR = 3.99 min TLC, Rf (CH2Cl2/Me0H 19:1) = 0.35 MS (method D): 382 [M+H]
Step 2
8-{2-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]- phenylcarbamoyl}-octanoic acid methyl ester
Figure imgf000155_0001
To a solution of 2.65 g (13 mmol) Monomethyl azelate in 20 mL DCM is added at rt a solution of 1.87 g (16 mmol) Benzotriazole and 1.87 g (16 mmol) Thionylchloride in 10 mL DCM. The suspension is stirred for 1 h, filtered, washed with 20 mL DCM and the solvent is removed in vacuo. The residue is dissolved in 10 mL DCM and added at 0°C to a solution of 2.0 g (5.2 mmol) [(lR,2S)-l-(2-Amino-benzenesulfonylaminocarbonyl)-2-vinyl- cyclopropylj-carbamic acid tert-butyl ester, 5.1 g (50 mmol) NEt3 and 100 mg DMAP in 50 mL DCM. After stirring for 15 h at rt the reaction is quenched by addition of aq. bicarbonate, extracted with DCM, dried with Na2SO4, filtered and the solvent is removed in vacuo. The residue is purified by FC on silica (eluent: CH2Cl2/Me0H 98:2 -> 95:5) to give the title compound as a red oil.
HPLC (method A) tR = 5.19 min TLC, Rf (CH2Cl2/MeOH 9:1) = 0.46 MS (method D): 566 [M+]
Step 3 8-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)-sulfamoyll-phenylcarbamoyl}- octanoic acid methyl ester
Figure imgf000156_0001
To a solution of 2.48 g (4.4 mmol) 8-{2-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl- cyclopropane-carbonyl)-sulfamoyl]-phenylcarbamoyl}-octanoic acid methyl ester in 4 mL Dioxane is added 6 mL 4N HCl in Dioxane at rt and the mixture is stirred for 15 h. The solvent is removed in vacuo to give the title compound as a hydrochloride salt which is used without further purification. HPLC (method A) tR = 3.36 min MS (method D): 466 [M+]
Step 4
(2S,4R)-2-{(lR,2S)-l-[2-(8-Methoxycarbonyl-octanoylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-(7-methoxy-2-phenyl- quinolin-4-yloxy)-pyrroHdine-l-carboxylic acid tert-butyl ester
Figure imgf000156_0002
To an ice-cold solution of 0.39 g (0.78 mmol) 8-{2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylcarbamoyl}-octanoic acid methyl ester (HCl-salt) in 25 mL DCM is added 0.44 g (0.94 mmol) (2S,4R)-4-(7-Methoxy-2-phenyl-quinolin-4- yloxy)-pyrrolidine-l,2-dicarboxylic acid 1 -tert-butyl ester (prepared according to WO 2000009543), 0.46 g (1.2 mmol) HBTU and 0.51 g (3.9 mmol) DEPEA and the ice bath is removed. After stirring for 15 h at rt the reaction is quenched by addition of aq. bicarbonate, extracted with DCM, dried with Na2SO4, filtered and the solvent is removed in vacuo. The residue is purified by FC on silica (eluent: CH2Cl2/Me0H 99:1 -> 95:5) to give the title compound as a colorless oil. HPLC (method A) tR = 5.43 min MS (method D): 912 [M+]
Step 5
8-{2-[((lR,2S)-l-{[(2S,4R)-4-(7-Methoxy-2-phenyl-quinolin-4-yloxy)-pyrroIidine-2- carbonyl]-amino}-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]-phenyIcarbamoyl}- octanoic acid
Figure imgf000157_0001
To a solution of 0.45 g (0.39 mmol) (2S,4R)-2-{(lR,2S)-l-[2-(8-Methoxycarbonyl- octanoylamino)-benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-(7- methoxy-2-phenyl-quinolin-4-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester is added 2 mL TFA at rt. After stirring for 1 h the solvent is removed in vacuo, the residue is dissolved in 10 mL THF/MeOH/H2O (2:1:1) and 50 mg (2.1 mmol) LiOH is added at rt. After stirring for 15 h, pH 5 is adjusted by addition of IN HCl, the solvent is removed in vacuo, the residue is taken up in water and extracted with DCM. The combined organic phases are dried with Na2SO4, filtered and the solvent is removed in vacuo to give the title compound as a colorless oil, which is used without further purification. HPLC (method A) tR = 4.49 min MS (method D): 798 [M+]
Example 2
(8S,10R)-10-[2-(2-IsopropyIamino-thiazol-4-yI)-7-methoxy-quinolin-4-yIoxy]-5-[(lR,2S)- l-carbonyIamino-2-vinyl-cycIopropyl]-2,2-dioxo-2λ*6*-thia-3,6,12,22-tetraaza- tricyclo[21.4.0.0*8,12*]heptacosa-l(27),23,25-triene-4,7,13,21-tetraone
Figure imgf000158_0001
To a an ice-cold solution of 90 mg (0.10 mmol) 8-{2-[2-({(2S,4R)-4-[2-(2-Isopropylamino- thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-2-carbonyl}-amino)-2-methyl- propionylsulfamoyl]-phenylcarba-moyl}-octanoic acid in 26 mL DCM/DMF (25:1) is added
135 mg (1.04 mmol) DIPEA followed by 59 mg (0.16 mmol) HATU. After 15 min the ice bath is removed and stirring is continued at rt for 1 h. The solvent is removed in vacuo and the residue is purified by preparative reverse phase HPLC (Method G) to give the title compound as a yellow solid.
HPLC (method A) tR = 5.11 min
TLC, Rf (CH2Cl2/MeOH/H2O/AcOH 75:27:5:0.5) = 0.13
MS (method D): 844 [M+]
Preparation of 8-{2-[2-({(2S,4R)-4-[2-(2-Isopropylamiπo-thiazol-4-yl)-7-methoxy- quinoIin-4-yloxy]-pyrrolidine-2-carbonyI}-amino)-2-methyl-propionylsulfaraoyl]- phenylcarbamoylj-octanoic acid
Step l
(2S,4R)-4-[2-(2-IsopropyIamino-thiazoI-4-yl)-7-methoxy-quinolin-4-yloxyl-2-{(lR,2S)-l- [2-(8-methoxycarbonyl-octanoylamino)-benzenesulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoyl}-pyrrolidine-l-carboxylic acid tert-butyl ester
Figure imgf000159_0001
The title compound is prepared analogously as described for the title compound in Example 1 (step 4) using 91 mg (0.18 mmol) 8-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl]-phenyl-carbamoyl}-octanoic acid methyl ester (HCl-salt), 95 mg (0.18 mmol) (2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-l,2- dicarboxylic acid 1-tert-butyl ester (prepared according to WO 2005073216), 89 mg (0.23 mmol) HATU and 116 mg (0.90 mmol) DIPEA in 5 mL DMF. HPLC (method A) tR = 5.71 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.42 MS (method D): 976 [M+]
Step 2
(2S,4R)-2-{(lR,2S)-l-[2-(8-Carboxy-octanoylamiπo)-benzenesulfonylaniinocarbonyl]-2- vinyl-cyclopropyIcarbamoyl}-4-[2-(2-isopropyIamino-thiazol-4-yI)-7-methoxy-quinoliιi- 4-yloxyJ-pyrrolidine-l-carboxylic acid tert-butyl ester
Figure imgf000159_0002
To a solution of 103 mg (0.10 mmol) (2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7- methoxy-quinolin-4-yloxy]-2- {(1 R,2S)- 1 -[2-(8-methoxycarbonyl-octanoylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-pyrrolidine-l-carboxylic acid tert-butyl ester in 8 mL THF/MeOH/H2O (2:1:1) is added 26 mg (1.1 mmol) LiOH at rt and the mixture is stirred for 2 h at 40°C. The solvent is removed in vacuo, pH 3 is adjusted by addition of IN HCl followed by extraction with DCM. The combined organic phase is washed with brine, dried with Na2SO4, filtered and the solvent is removed in vacuo to give the title compound as a yellow oil, which is used without further purification. HPLC (method A) tR = 5.23 min TLC, Rf (CH2Cl2/MeOH 9:1) = 0.20 MS (method D): 962 [M+]
Step 3
8-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy-quinoIin-4- yloxy]-pyrrolidine-2-carbonyI}-amiπo)-2-vinyl-cyclopropanecarbonyl]-sulfamoyl}- phenylcarbamoyl)-octanoic acid
Figure imgf000160_0001
To a solution of 102 mg (0.11 mmol) (2S,4R)-2-{(lR,2S)-l-[2-(8-Carboxy-octanoylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-[2-(2-isopropylamino- thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-l-carboxylic acid tert-butyl ester in 5 mL DCM is added 0.5 mL TFA at rt. After stirring for 2 h the solvent is removed in vacuo.
To remove excess of TFA the residue is taken up in DCM and the solvent is removed in vacuo. This procedure is repeated three times. The title compound is obtained as a brown oil, which is used without further purification.
HPLC (method A) tR = 4.55 min
TLC, Rf (CH2Cl2/MeOH/H2O/AcOH 90:10: 1 :0.5) = 0.49
MS (method D): 862 [M+]
Example 3
4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (8S,10R)-5-[(lR,2S)-l-carbonylamino- 2-vinyl-cyclopropyl]-2,2,4,7,13,21-hexaoxo-2λ*6*-thia-3,6,12,22-tetraaza- tricyclo[21.4.0.0*8,12*]heptacosa-l(27),23,25-trien-10-yl ester
Figure imgf000161_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 119 mg (0.14 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-5- {(lR,2S)-l-[2-(8-carboxy-octanoylamino)-benzenesulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoyl}-pyrrolidin-3-yl ester (TFA-salt), 182 mg (1.4 mmol) DIPEA and 268 mg (0.71 mmol) HATU. HPLC (method A) tR = 5.00 min TLC, Rf (CH2Cl2/Me0H 19:1) = 0.41
MS (method D): 710 [M+] + 727 [M+H2O]
Preparation of (2S,4R)-4-(4-Fluoro-13-dihydro-isoindole-2-carbonyloxy)-pyrrolidine- 1,2-dicarboxylic acid l-tert-butyl ester
Step l (2S,4R)-4-(4-Fluoro-l,3-dihydro-isoindole-2-carbonyloxy)-pyrrolidine-l,2-dicarboxylic acid l-tert-butyl ester 2-methyl ester
Figure imgf000161_0002
To a solution of 1.79 g (7.1 mmol) 2S,4R)-4-Hydroxy-pyrrolidine-l,2-dicarboxylic acid 1- tert-butyl ester 2-methyl ester in 65 mL DCM is added 1.57 (9.2 mmol) CDI at it and the mixture is stirred for 1 h. A solution of 2.91 g (21.2. mmol) 4-Fluoro-2,3-dihydro-lH- isoindole (prepared according to WO 2005037214) in 5 mL DCM is added and the reaction mixture is stirred at rt overnight. The mixture is diluted with DCM and washed three times with IN HCl, sat. NaHCO3 and brine. The organic phase is dried with Na2SO4, filtered and the solvent is removed in vacuo. The residue is purified by FC (CH2Cl2ZMeOH 98:2) to give the title compound as an oil.
LC-MS (method E) tR= 3.76 min, [M-BOC] - 308 TLC, Rf (CH2Cl2MeOH 9:1) = 0.85
Step 2 (ZS^R^^-Fluoro-l^-dihydro-isoindole-l-carbonyloxy^pyrrolidine-l^-dicarboxylic acid l-tert-butyl ester
Figure imgf000162_0001
To a mixture of 500 mg (1.2 mmol) (2S,4R)-4-(4-Fluoro-l,3-dihydro-isoindole-2- carbonyloxy)-pyrrolidine-l,2-dicarboxylic acid l-tert-butyl ester 2-methyl ester in 10 mL
THF/methanol/water (3:1:1) is added 62 mg (1.5 mmol) lithiumhydroxid-hydrate and the mixture is stirred at rt for 6 h. pH is adjusted to 3 and the mixture is extracted four times with
DCM. The combined organic layers are washed with NaHCO3 and brine, dried over Na2SO4, filtered and concentrated in vacuo to yield the title compound which was used without further purification.
HPLC (method B) tR = 3.15 min
LC-MS (method E) tR = 3.49 min, [M-H] = 394
TLC, Rf (CH2Cl2/Me0H 9:1) - 0.48
Preparation of 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-l-tert- butoxycarbonyl-5-{(lR,2S)-l-[2-(8-carboxy-octanoylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropyIcarbamoyl}-pyrrolidin-3-yI ester
Step l 4-Fluoro-l,3-dihydro-isoindo-e-2-carboxy-ic acj{j (3R,5S)-l-tert-butoxycarbonyl-5- {(lR,2S)-l-[2-(8-methoxycarbonyl-octanoylamino)-benzenesulfonylaminocarbonyl]-2- vinyl-cyclopropyIcarbamoylJ-pyrroIidin-3-yl ester
Figure imgf000163_0001
The title compound is prepared analogously as described for the title compound in Example 1
(step 4) using 200 mg (0.14 mmol) 8-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl] -phenyl carbamoylj-octanoic acid methyl ester (HCl-salt), 113 mg (0.29 mmol) (2S,4R)-4-(4-Fluoro- l,3-dihydro-isoindole-2-carbonyloxy)-pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester,
136 mg (0.36 mmol) HATU and 93 mg (0.71 mmol) DIPEA in 5 mL DCM.
HPLC (method A) = 5.72 min
TLC, Rf(CH2Cl2ZMeOH 9:1) = 0.50
MS (method D): 859 [M+H2O]
Step 2
4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5-
{(lR,2S)-l-[2-(8-carboxy-octanoyIaniino)-benzenesulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoyI}-pyrroIidin-3-yI ester
Figure imgf000163_0002
To a solution of 118 mg (0.14 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5-{(lR,2S)-l-[2-(8-methoxycarbonyl-octanoylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-pyrrolidin-3-yl ester in 16 mL THF/MeOH/H2O (2:1:1) is added 34 mg (1.4 mmol) LiOH at rt and the mixture is stirred for 2 h at 400C. The solvent is removed in vacuo, pH 3 is adjusted by addition of IN HCl followed by extraction with DCM. The combined organic phase is washed with brine, dried with Na2SO4, filtered and the solvent is removed in vacuo to give the title compound as a yellow oil, which is used without further purification. tR HPLC (method A) tR = 5.17 min TLC, Rf (CH2Cl2/Me0H 85:15) = 0.54 MS (method D): 845 [M+H20]
Step 3
4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyI-5-
{(lR,2S)-l-[2-(8-carboxy-octanoyIamino)-benzenesulfonylaminocarbonyl]-2-vinyI- cyclopropyIcarbamoyl}-pyrrolidin-3-yl ester
Figure imgf000164_0001
To a solution of 116 mg (0.14 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)- 1 -tert-butoxycarbonyl-5- {( 1 R,2S)- 1 -[2-(8-carboxy-octanoylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-pyrrolidin-3-yl ester in 25 mL DCM is added 1 mL TFA at rt. After stirring overnight the solvent is removed in vacuo. To remove excess of TFA the residue is taken up in DCM and the solvent is removed in vacuo, which is repeated three times. The title compound is obtained as a brown oil, which is used without further purification. HPLC (method A) tR = 4.22 min TLC, Rf (CH2Cl2/Me0H 85:15) = 0.56 MS (method D): 728 [M+]
Example 4 ll-[2-(l,2,3,4-tetrahydronaphthalene)]-8-[(lR,2S)-l-carbonylamino-2-vinyl- cyclopropyl]-5,5-dioxo-5,8,9,ll,12,15,16,17,18,19,20,22-dodecahydro-6H,14H-5λ*6*- thia-6,9,12,22-tetraaza-benzocycIoicosene-7, 10, 13,21 -tet raone
Figure imgf000165_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 65 mg (0.09 mmol) 8-[2-({(lR,2S)-l-[(2-Amino-l,2,3,4-tetrahydro-naphthalene-2- carbonyl)-amino]-2-vinyl-cyclopropanecarbonyl}-sulfamoyl)-phenylcarbamoyl]-octanoic acid (TFA-salt), 114 mg (0.88 mmol) DIPEA and 167 g (0.44 mmol) HATU.
HPLC (method A) = 5.07 min
TLC, Rf (CH2Cl2/Me0H 85:15) = 0.23
MS (method D): 607 [M+]
Preparation of 8-[2-({(lR,2S)-l-[(2-Amino-l,2,3,4-tetrahydro-naphthalene-2-carbonyl)- amino]-2-vinyl-cydopropanecarbonyl}-sulfamoyl)-phenylcarbamoyl]-octanoic acid
Step l
8-[2-({(lR,2S)-l-[(2-tert-Butoxycarbonylamino-l,2,3,4-tetrahydro-naphthalene-2- carbonyl)-amino]-2-vinyl-cycIopropanecarbonyl}-sulfarnoyl)-phenylcarbamoyl]- octanoic acid methyl ester
Figure imgf000165_0002
The title compound is prepared analogously as described for the title compound in Example 1 (step 4) using 150 mg (0.19 mmol) 8-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl]-phenylcarbamoyl}-octanoic acid methyl ester (HCl-salt), 66 mg (0.22 mmol) 2- tert-Butoxycarbonylamino-l,2,3,4-tetrahydro-naphthalene-2-carboxylic acid, 84 mg (0.22 mmol) HBTU and 120 mg (0.93 mmol) DIPEA in 2 mL DMF. HPLC (method A) tR = 5.77 min TLC, Rf (CH2Cl2/MeOH 19:1) = 0.53 MS (method D): 739 [M+]
Step 2
8-[2-({(lR,2S)-l-[(2-Amino-l,2,3,4-tetrahydro-naphthalene-2-carbonyl)-amino]-2-vinyl- cyclopropanecarbonyl}-sulfamoyl)-phenylcarbamoyl]-octanoic acid
Figure imgf000166_0001
The title compound is prepared analogously as described for the title compound in Example 1 (step 5) using 102 mg (0.14 mmol) 8-[2-({(lR,2S)-l-[(2-tert-Butoxycarbonylamino-l,2,3,4- tetrahydro-naphthalene-2-carbonyl)-amino]-2-vinyl-cyclopropanecarbonyl}-sulfamoyl)- phenylcarbamoylj-octanoic acid methyl ester and 1 mL TFA in 10 mL DCM and 33 mg (1.4 mmol) LiOH in 12 mL THF/MeOH/H2O (2:1:1). HPLC (method A) tR = 3.93 min
TLC, Rf (CH2Cl2/Me0H 19:1) = 0.44 MS (method D): 625 [M+]
Example 5 ll-[2-indanyl]-8-[(lR,2S)-l-carbonyIainino-2-vinyl-cyclopropyI]-5,5-dioxo-
5,8,9,ll,12,15,16,17,18,19,20,22-dodecahydro-6H,14H-5λ*6*-thia-6,9,12,22-tetraaza- benzocycloicosene-7,10,13^1-tetraone
Figure imgf000166_0002
The title compound is prepared analogously as described for the title compound in Example 2 using 83 mg (0.012 mmol) 8-[2-({(lR,2S)-l-[(2-Amino-indane-2-carbonyl)-amino]-2-vinyl- cyclopropane-carbonyl}-sulfamoyl)-phenylcarbamoyl]-octanoic acid (TFA-salt), 149 mg (1.15 mmol) DEPEA and 219 g (0.58 mmol) HATU. HPLC (method A) tR = 4.91 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.25 MS (method D): 593 [M+]
Preparation of 8-[2-({(lR,2S)-l-[(2-Amino-indane-2-carbonyl)-amino]-2-vinyl- cyclopropane-carbonyl}-sulfamoyl)-phenylcarbamoyl]-octanoic acid
Step l
8-[2-({(lR,2S)-l-[(2-tert-Butoxycarbonylaιnino-indane-2-carbonyl)-ainino]-2-vinyl- cyclopropanecarbonyl}-sulfamoyl)-phenylcarbamoyl]-octanoic acid methyl ester
Figure imgf000167_0001
The title compound is prepared analogously as described for the title compound in Example 1 (step 4) using 163 mg (0.20 mmol) 8-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl]-phenylcarbamoyl}-octanoic acid methyl ester (HCl-salt), 67 mg (0.24 mmol) 2- tert-Butoxycarbonylamino-indan-2-carboxylic acid, 91 mg (0.24 mmol) HBTU and 130 mg (1.00 mmol) DIPEA in 2 mL DMF. HPLC (method A) tR = 5.61 min TLC, Rf (CH2Cl2/Me0H 19:1) - 0.41 MS (method D): 725 [M+]
Step 2
8-[2-({(lR,2S)-l-[(2-tert-Butoxycarbonylamino-indane-2-carbonyl)-amino]-2-vinyI- cyclopropanecarbonyl}-sulfamoyl)-phenylcarbamoyl]-octanoic acid
Figure imgf000168_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 84 mg (0.12 mmol) 8-[2-({(lR,2S)-l-[(2-tert-Butoxycarbonylamino-indane-2- carbonyl)-amino]-2-vinyl-cyclopropanecarbonyl}-sulfamoyl)-phenylcarbamoyl]-octanoic acid methyl ester and 28 mg (1.16 mmol) LiOH in 10 mL THF/MeOH/H2O (2:1:1). HPLC (method A) tR = 5.02 min TLC, Rf(CH2Cl2MeOH 9:1) = 0.35 MS (method D): 711 [M+]
Step 3
8-[2-({(lR,2S)-l-[(2-tert-Butoxycarbonylamino-indane-2-carbonyl)-ainino]-2-vinyl- cyclopropanecarbonyl}-sulfamoyl)-phenylcarbamoyl]-octanoic acid
Figure imgf000168_0002
The title compound is prepared analogously as described for the title compound in Example 2 (step 3) using 82 mg (0.12 mmol) 8-[2-({(lR,2S)-l-[(2-tert-Butoxycarbonylamino-indane-2- carbonyl)-amino]-2-vinyl-cyclopropanecarbonyl}-sulfamoyl)-phenylcarbamoyl]-octanoic acid and 1 mL TFA in 25 mL DCM. HPLC (method A) tR = 2.85 min MS (method D): 611 [M+]
Example 6
12-CyclopentylmethyI-8-[(lR,2S)-l-carbonylamino-2-vinyl-cycIopropyl]-5,5-dioxo-
5,8,9,ll,12,15,16,17,18,19,20,22-dodecahydro-6H,14H-5λ*6*-thia-6,9,12,22-tetraaza- benzocycloicosene-?,! 0,13,21 -tetraone
Figure imgf000169_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 58 mg (0.08 mmol) 8-[2-({(lR,2S)-l-[2-(Cyclopentylmethyl-amino)-acetylamino]-2- vinyl-cyclopropane-carbonyl}-sulfamoyl)-phenylcarbamoyl]-octanoic acid (TFA-salt), 106 mg (0.82 mmol) DIPEA and 156 mg (0.41 mmol) HATU in 51 mL DCM/MeOH (50:1).
HPLC (method A) tR = 5.23 min
TLC, Rf (CH2Cl2/Me0H 85:15) = 0.23
MS (method D): 573 [M+H] + 590 [M+H2O]
Preparation of (tert-Butoxycarbonyl-cyclopentylmethyl-amino)-acetic acid
Step 1
(Cyclopentylmethyl-amino)-acetic acid methyl ester
Figure imgf000169_0002
To a solution of 9.0 g (89 mmol) Cyclopentanecarboxaldehyde, 11.3 g (89 mmol) Glycine methylester hydrochloride and 13.1 g (116 mmol) NEt3 in 250 mL MeOH is added 2 g molecular sieves 4A. After stirring for 30 min at rt, 4.5 g (116 mmol) NaBH4 is added at 00C in 5 portions. The ice-bath is removed and stirring is continued for 2 h at rt. The reaction is quenched by addition of aq. bicarbonate, MeOH is evaporated and the residue is diluted with water. After extraction with DCM, the organic phase is washed with brine, dried with Na2SO4, filtered and the solvent is removed in vacuo. The residue is purified by FC on silica (eluent: hexane/EtOAc 3:1) to give the title compound as a yellow oil. TLC, Rf (hexane/EtOAc 1:1) = 0.55 MS (method D): 172 [M+H] Step 2
(tert-Butoxycarbonyl-cycIopentylmethyl-amino)-acetic acid methyl ester
Figure imgf000170_0001
A solution of 1.1 g (6.2 mmol) (Cyclopentylmethyl-amino)-acetic acid methyl ester and 1.25 g (12.4 mmol) NEt3 in 60 mL DCM is cooled to 0°C and 2.03 g (9.3 mmol) (BOC)2O is added. The ice-bath is removed after 15 min and stirring is continued for 2 h at rt. The reaction is quenched by addition of aq. bicarbonate and extracted with DCM. The organic phase is washed with brine, dried with Na2SO4, filtered and the solvent is removed in vacuo.
The residue is purified by FC on silica (eluent: CH2Cl2/MeOH 99:1) to give the title compound as a yellow oil.
TLC, Rf (hexane/EtOAc 1:1) = 0.86 MS (method D): 216 [M+-55]
Step 3 (tert-Butoxycarbonyl-cycIopentylmethyl-ainino)-acetic acid
Figure imgf000170_0002
To a solution of 1.22 g (4.5 mmol) (tert-Butoxycarbonyl-cyclopentylmethyl-amino)-acetic acid methyl ester in 40 mL THF/MeOH/H2O (2:1:1) is added 0.57 g (13.5 mmol) LiOH and the reaction stirred for 15 h at rt. The solvent is removed in vacuo, pH 3 is adjusted by addition of 4N HCl followed by extraction with EtOAc. The combined organic phase is washed with brine, dried with Na2SO4, filtered and the solvent is removed in vacuo. The residue is purified by FC on silica (eluent: CH2Cl2/MeOH 98:2) to give the title compound as a yellow oil. TLC, Rf(CH2Cl2ZMeOH 19:1) = 0.34 MS (method D): 202 [M+-SS]
Preparation of 8-[2-({(lR,2S)-l-[2-(Cyclopentylmethyl-amino)-acetylamino]-2-vinyl- cyclopro-panecarbonyl}-sulfamoyl)-phenylcarbamoyl]-octanoic acid Step 1
8-[2-({(lR,2S)-l-[2-(tert-Butoxycarbonyl-cyclopentylmethyl-amino)-acetyIamino]-2- vinyl-cyclopropanecarbonyl}-sulfamoyl)-phenylcarbamoyl]-octanoic acid methyl ester
Figure imgf000171_0001
The title compound is prepared analogously as described for the title compound in Example 1
(step 4) using 150 mg (0.19 mmol) 8-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl] -phenyl-carbamoyl }-octanoic acid methyl ester (HCl-salt), 57 mg (0.22 mmol) (tert-Butoxycarbonyl-cyclopentylmethyl-amino)-acetic acid, 84 mg (0.22 mmol) HBTU and 120 mg (0.93 mmol) DIPEA in 2 mL DMF. HPLC (method A) tR = 5.98 min TLC, Rf (CH2Cl2/MeOH 19:1) = 0.30 MS (method D): 705 [M+]
Step 2
8-[2-({(lR,2S)-l-[2-(Cyclopentylmethyl-amino)-acetyIamino]-2-vinyl- cyclopropanecarbonyl}-sulfamoyl)-phenylcarbamoyl]-octanoic acid
Figure imgf000171_0002
The title compound is prepared analogously as described for the title compound in Example 1 (step 5) using 102 mg (0.14 mmol) 8-[2-({(lR,2S)-l-[2-(tert-Butoxycarbonyl- cyclopentylmethyl-amino)-acetylamino]-2-vinyl-cyclopropanecarbonyl}-sulfamoyl)- phenylcarbamoyl]-octanoic acid methyl ester and 1 mL TFA in 10 mL DCM and 33 mg (1.4 mmol) LiOH in 12 mL THF/MeOH/H2O (2:1:1). HPLC (method A) tR = 3.99 min TLC, Rf (CH2Cl2MeOH 85:15) = 0.57 MS (method D): 591 [M+]
Example 7
(8S,10R)-10-(7-Methoxy-2-phenyl-quinolin-4-yloxy)-5-[(lR,2S)-l-carbonylamino-2- vinyI-cyclopropyl]-2,2-dioxo-2λ*6*-thia-3,6,12,23-tetraaza- tricyclo[22.4.0.0*8,12*]octacosa-l(28),24,26-triene-4,7,13,22-tetraone
Figure imgf000172_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 150 mg (0.16 mmol) 9-{2-[((lR,2S)-l-{[(2S,4R)-4-(7-Methoxy-2-phenyl-quinolin-4- yloxy)-pyrrolidine-2-carbonyl]-amino}-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]- phenylcarbamoyl}-nonanoic acid (TFA-salt), 207 mg (1.6 mmol) DIPEA and 304 mg (0.80 mmol) HATU in 51 mL DCM/MeOH (50:1). HPLC (method A) tR = 5.00 min TLC, Rf (CH2Cl2/MeOH 9:1) = 0.5 MS (method D): 794 [M+]
Preparation of (8-{2-[((lR,2S)-l-{[(2S,4R)-4-(7-Methoxy-2-phenyl-quinoIin-4-yloxy)- pyrroIidine-2-carbonyl]-amino}-2-vinyl-cyclopropanecarbonyI)-sulfamoyI]- phenylcarbamoyl}-octanoic acid
Step l
9-{2-[((lR,2S)-l-tert-ButoxycarbonyIamino-2-vinyl-cyclopropanecarboπyl)-sulfamoylJ- phenylcarbamoyl}-nonanoic acid methyl ester
Figure imgf000173_0001
The title compound is prepared analogously as described for the title compound in Example 1 (Step 2) using 1.50 g (3.9 mmol) [(lR,2S)-l-(2-Amino-benzenesulfonylaminocarbonyl)-2- vinyl-cyclopropyl]-carbamic acid tert-butyl ester, 2.12 g (9.8 mmol) Monomethyl sebacate, 1.41 g (11.8 mmol) Benzotriazole, 1.41 g (11.8 mmol) Thionylchloride, 1.84 g (20.0 mmol) NEt3 and 100 mg DMAP in 50 mL DCM. HPLC (method A) tR = 5.42 min TLC, Rf (CH2Cl2/MeOH 19:1) = 0.33 MS (method D): 580 [M+]
Step 2
9-{2-[((lR,2S)-l-Amino-2-vinyI-cyclopropanecarbonyI)-sulfamoyl]-phenylcarbamoyl}- nonanoic acid methyl ester
Figure imgf000173_0002
The title compound is prepared analogously as described for the title compound in Example 1
(Step 3) using 1.10 g (1.9 mmol) 9-{2-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylcarbamoyl}-nonanoic acid methyl ester and 3 mL 4
N HCl in Dioxane.
HPLC (method A) tR = 3.65 min
MS (method D): 480 [M+]
Step 3
(2S,4R)-2-{(lR,2S)-l-[2-(9-MethoxycarbonyI-nonanoylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropyIcarbamoyl}-4-(7-methoxy-2-phenyl- quinolin-4-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester
Figure imgf000174_0001
The title compound is prepared analogously as described for the title compound in Example 1 (Step 4) using 280 mg (0.43 mmol) 9-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl]-phenylcarbamoyl}-nonanoic acid methyl ester (HCl-salt), 218 mg (0.47 mmol) (2S,4R)-4-(7-Methoxy-2-phenyl-quinolin-4-yloxy)-pyrrolidine- 1 ,2-dicarboxylic acid 1 -tert- butyl ester, 278 mg (2.15 mmol) DIPEA and 212 mg (0.56 mmol) HBTU in 2 mL DMF. HPLC (method A) tR = 5.59 min TLC, Rf (CH2Cl2/MeOH 19:1) = 0.23 MS (method D): 926 [M+]
Step 4
(2S,4R)-2-{(lR,2S)-l-[2-(9-Carboxy-nonanoylamino)-benzenesuIfonylaminocarbonylJ-2- vinyl-cyclopropylcarbamoylJ^^T-methoxy-l-phenyl-quinolin^-yloxy^pyrroIidine-l- carboxylic acid tert-butyl ester
Figure imgf000174_0002
The title compound is prepared analogously as described for the title compound in Example 2 (Step 2) using 152 mg (0.16 mmol) (2S,4R)-2-{(lR,2S)-l-[2-(9-Methoxycarbonyl- nonanoylamino)-benzene-sulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-(7- methoxy-2-phenyl-quinolin-4-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester and 38 mg (1.6 mmol) LiOH in 8 mL THF/MeOH/H2O (2:1 :1). HPLC (method A) tR = 5.06 min MS (method D): 912 [M+]
Step 5
9-{2-[((lR,2S)-l-{[(2S,4R)-4-(7-Methoxy-2-phenyl-quinolin-4-yloxy)-pyrrolidine-2- carbonyl]-amino}-2-vinyl-cyclopropanecarbonyI)-sulfamoyl]-phenyIcarbamoyl}- nonanoic acid
Figure imgf000175_0001
The title compound is prepared analogously as described for the title compound in Example 2
(Step 3) using 150 mg (0.16 mmol) (2S,4R)-2-{(lR,2S)-l-[2-(9-Carboxy-nonanoylamino)- benzene-sulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-(7-methoxy-2-phenyl- quinolin-4-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester and 1 mL TFA in 5 mL
DCM.
HPLC (method A) tR = 4.61 min
MS (method D): 812 [M+]
Example 8
(8S,10R)-10-[2-(2-Isopropylamino-thiazol-4-yI)-7-methoxy-quinolin-4-yloxy]-5-[(lR,2S)- l-carbonylamino-2-vinyl-cycIopropyl]-2,2-dioxo-2λ*6*-thia-3,6,12,23-tetraaza- tricyclo[22.4.0.0*8,12*]octacosa-l(28),24,26-triene-4,7,13,22-tetraone
Figure imgf000176_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 57 mg (0.05 mmol) 9-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)- 7-methoxy-quinolin-4-yloxy]-pyrrolidine-2-carbonyl}-amino)-2-vinyl- cyclopropanecarbonyl]-sulfamoyl}-phenylcarbamoyl)-nonanoic acid, 67 mg (0.52 mmol) DEPEA and 99 mg (0.26 mmol) HATU in 51 mL DCM/DMF (50:1). HPLC (method A) tR = 5.33 min TLC, Rf (CH2Cl2/MeOH 9:1) = 0.30 MS (method D): 858 [M+]
Preparation of 9-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-Isopropylamino-thiazoI-4-yI)-7- methoxy-quinolin-4-yloxy]-pyrroIidine-2-carbonyl}-aιτiino)-2-vinyl- cyclopropanecarbonyl]-sulfamoyl}-phenylcarbamoyl)-nonanoic acid
Step l
(2S,4R)-4-[2-(2-IsopropyI-thiazoI-4-yl)-7-methoxy-quinolin-4-yloxy]-2-{(lR,2S)-l-[2-(8- methoxycarbonyl-octanoylaminoJ-benzenesulfonylaminocarbonylJ-Z-vinyl- cyclopropylcarbamoyl}-pyrrolidine-l-carboxylic acid tert-butyl ester
Figure imgf000176_0002
The title compound is prepared analogously as described for the title compound in Example 2
(step 1) using 150 mg (0.22 mmol) 9-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyi)- sulfamoyl]-phenylcarbamoyl}-nonanoic acid methyl ester, 117 mg (0.22 mmol) (2S,4R)-4-
[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-l,2- dicarboxylic acid 1-tert-butyl ester, 101 mg (0.27 mmol) HATU and 143 mg (1.1 mmol)
DIPEA in 5 mL DMF.
HPLC (method A) tR = 5.80 min
TLC, Rf (CH2Cl2/MeOH 9:1) = 0.30
MS (method D): 990 [M+]
Step 2
(2S,4R)-2-{(lR,2S)-l-[2-(9-Carboxy-nonanoylamino)-benzenesuIfonylaminocarbonyl]-2- vinyl-cyclopropylcarbamoyl}-4-[2-(2-isopropylamino-thiazol-4-yl)-7-methoxy-quinoliii- 4-yloxy]-pyrrolidine-l-carboxylic acid tert-butyl ester
Figure imgf000177_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 59 mg (0.053 mmol) (2S,4R)-4-[2-(2-Isopropyl-thiazol-4-yl)-7-methoxy- quinolin-4-yloxy]-2- {(1 R,2S)- 1 -[2-(8-methoxycarbonyl-octanoylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropyl-carbamoyl} -pyrrolidine- 1 -carboxylic acid tert-butyl ester and 22 mg (0.53 mmol) LiOH in 8 mL THF/MeOH/H2O (2:1:1). HPLC (method A) tR = 5.28 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.26 MS (method D): 976 [M+]
Step 3
9-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-IsopropyIamino-thiazol-4-yl)-7-methoxy-quinolin-4- yloxy]-pyrrolidine-2-carbonyl}-amino)-2-vinyl-cyclopropanecarbonyI]-sulfamoyl}- phenylcarbamoyl)-nonanoic acid
Figure imgf000178_0001
The title compound is prepared analogously as described for the title compound in Example 2
(step 3) using 50 mg (0.051 mmol) (2S,4R)-2-{(lR,2S)-l-[2-(9-Carboxy-nonanoylamino)- benzenesulfonyl-aminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-[2-(2-isopropylamino- thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-l-carboxylic acid tert-butyl ester and
0.5 mL TFA in 5 mL DCM.
HPLC (method A) tR = 4.74 min
TLC, Rf (CH2Cl2/MeOH/H2O/AcOH 90:10:1:0.5) = 0.16
MS (method D): 876 [M+]
Example 9 (8S,10R)-10-(7-Methoxy-2-phenyl-quinolin-4-yloxy)-5-[(lR,2S)-l-carbonylamino-2- vinyl-cyclopropyl]-2,2-dioxo-2λ*6*-thia-3,6,12,21-tetraaza- tricyclo[20.4.0.0*8,12*]hexacosa-l(26),22,24-triene-4,7,13,20-tetraone
Figure imgf000178_0002
The title compound is prepared analogously as described for the title compound in Example 2 using 121 mg (0.14 mmol) 7-{2-[((lR,2S)-l-{[(2S,4R)-4-(7-Methoxy-2-phenyl-quinolin-4- yloxy)-pyrrolidine-2-carbonyl]-amino}-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]- phenylcarbamoyl}-heptanoic acid (TFA-salt), 174 mg (1.4 mmol) DEPEA and 257 mg (0.66 mmol) HATU in 51 mL DCM/DMF (50:1). HPLC (method A) tR = 4.68 min TLC, Rf (CH2Cl2/Me0H 85:15) = 0.43 MS (method D): 766 [M+]
Preparation of 7-{2-[((lR,2S)-l-{[(2S,4R)-4-(7-Methoxy-2-phenyl-quinolin-4-yIoxy)- pyrrolidine-2-carbonyl]-amino}-2-vinyl-cyclopropanecarbonyl)-sulfamoyI]- phenylcarbamoyl}-heptanoic acid
Step l 7-{2-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyI-cyclopropanecarbonyl)-sulfamoyl]- phenylcarbamoylj-heptanoic acid methyl ester
Figure imgf000179_0001
The title compound is prepared analogously as described for the title compound in Example 1
(Step 2) using 0.76 g (1.99 mmol) [(lR,2S)-l-(2-Amino-benzenesulfonylaminocarbonyl)-2- vinyl-cyclopropyl]-carbamic acid tert-butyl ester, 0.94 g (4.97 mmol) Monomethyl suberate,
0.71 g (5.97 mmol) Benzotriazole, 0.71 g (5.97 mmol) Thionylchloride, 0.92 g (10 mmol)
NEt3 and 70 mg DMAP in 40 mL DCM.
HPLC (method A) tR = 4.95 min
TLC, Rf (CH2Cl2/Me0H 19:1) = 0.23 MS (method D): 552 [M+]
Step 2
7-{2-[((lR,2S)-l-Amino-2-vinyI-cyclopropanecarbonyl)-sulfamoyI]-phenylcarbamoyl}- heptanoic acid methyl ester
Figure imgf000180_0001
The title compound is prepared analogously as described for the title compound in Example 1
(Step 3) using 0.78 g (1.4 mmol) 7-{2-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylcarbamoyl}-heptanoic acid methyl ester and 1 mL
4N HCl in Dioxane.
HPLC (method A) tR = 3.04 min
MS (method D): 452 [M+]
Step 3
(2S,4R)-2-{(lR,2S)-l-[2-(7-MethoxycarbonyI-heptanoylamino)- benzenesulfonyIaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-(7-methoxy-2-phenyl- quinolin-4-yloxy)-pyrroIidine-l-carboxylic acid tert-butyl ester
Figure imgf000180_0002
The title compound is prepared analogously as described for the title compound in Example 1 (Step 4) using 150 mg (0.22 mmol) 7-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl]-phenylcarbamoyl}-heptanoic acid methyl ester, 120 mg (0.26 mmol) (2S,4R)-4- (7-Methoxy-2-phenyl-quinolin-4-yloxy)-pyrrolidine-l,2-dicarboxylic acid 1 -tert-butyl ester, 98 mg (0.26 mmol) HBTU and 139 mg (1.1 mmol) DIPEA in 2 mL DMF. HPLC (method A) tR = 5.19 min TLC, Rf (CH2Cl2/Me0H 19:1) = 0.43 MS (method D): 898 [M+] Step 4
(2S,4R)-2-{(lR,2S)-l-[2-(7-Carboxy-heptanoylamino)-benzenesulfonyIaminocarbonylJ- 2-vinyl-cyclopropylcarbamoyl}-4-(7-methoxy-2-phenyI-quinolin-4-yloxy)-pyrrolidine-l- carboxylic acid tert-butyl ester
Figure imgf000181_0001
The title compound is prepared analogously as described for the title compound in Example 2 (Step 2) using 179 mg (0.17 mmol) (2S,4R)-2-{(lR,2S)-l-[2-(7-Methoxycarbonyl- heptanoylamino)-benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-(7- methoxy-2-phenyl-quinolin-4-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester and 41 mg (1.7 mmol) LiOH in 10 mL THF/MeOH/H2O (2:1:1). HPLC (method A) tR = 4.74 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.32 MS (method D): 884 [M+]
Step 5
7-{2-[((lR,2S)-l-{[(2S,4R)-4-(7-Methoxy-2-phenyI-quinolin-4-yloxy)-pyrrolidine-2- carbonyI]-amino}-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]-phenylcarbamoyl}- heptanoic acid
Figure imgf000182_0001
The title compound is prepared analogously as described for the title compound in Example 2
(Step 3) using 134 mg (0.15 mmol) (2S,4R)-2-{(lR,2S)-l-[2-(7-Carboxy-heptanoylamino)- benzenesulfonyl-aminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-(7-methoxy-2-phenyl- quinolin-4-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester and 1 mL TFA in 25 mL
DCM.
HPLC (method A) tR = 4.04 min
TLC, Rf (CH2Cl2/MeOH 85:15) = 0.54
MS (method D): 784 [M+]
Example 10
(8S,10R)-10-[2-(2-IsopropyIamino-thiazoI-4-yl)-7-methoxy-quinolin-4-yIoxy]-5-[(lR,2S)- l-carbonylamino-2-vinyl-cyclopropyl]-2,2-dioxo-2λ*6*-thia-3,6,12,21-tetraaza- tricyclo[20.4.0.0*8,12*]hexacosa-l(26),22,24-triene-4,7,13,20-tetraone
Figure imgf000182_0002
The title compound is prepared analogously as described for the title compound in Example 2 using 121 mg (0.11 mmol) 7-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)- 7-methoxy-quinolin-4-yloxy]-pyrrolidine-2-carbonyl}-amino)-2-vinyl- cyclopropanecarbonyl]-sulfamoyl}-phenyl-carbamoyl)-heptanoic acid (TFA-salt), 145 mg (1.1 mmol) DIPEA and 213 mg (0.56 mmol) HATU in 51 mL DCM/DMF (50:1). HPLC (method A) tR = 4.98 min TLC, Rf (CH2Cl2/Me0H 85:15) = 0.46 MS (method D): 830 [M+]
Preparation of 7-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7- methoxy-quinolin-4-yloxyJ-pyrrolidine-2-carbonyI}-amino)-2-vinyl- cyclopropanecarbonyl]-sulfamoyl}-phenylcarbamoyl)-heptanoic acid
Step l
(2S,4R)-4-[2-(2-IsopropyIamino-thiazol-4-yl)-7-methoxy-quinolin~4-yloxy]-2-{(lR,2S)-l-
[2-(7-methoxycarbonyl-heptanoylamino)-benzenesulfonylaminocarbonyI]-2-vinyl- cyclopropylcarbamoyl}-pyrrolidine-l-carboxylic acid tert-butyl ester
Figure imgf000183_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 1) using 170 mg (0.24 mmol) 7-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl]-phenylcarbamoyl}-heptanoic acid methyl ester (HCl-salt), 207 mg (0.29 mmol) (2S,4R)-442-(2-Isopropylamino-thiazol-4-yl)-7-methoxy-quinolin-4-yloxy] -pyrrolidine- 1,2- dicarboxylic acid 1-tert-butyl ester, 111 mg (0.29 mmol) HBTU and 158 mg (1.2 mmol) DIPEA in 2 mL DMF. HPLC (method A) tR = 5.35 min TLC, Rf (CH2Cl2/Me0H 19:1) - 0.27 MS (method D): 962 [M+] Step 2
(2S,4R)-2-{(lR,2S)-l-[2-(7-Carboxy-heptanoylamino)-benzenesulfonylaminocarbonyl]- 2-vinyI-cyclopropylcarbamoyl}-4-[2-(2-isopropyIamino-thiazoI-4-yl)-7-methoxy- quinolin-4-yloxy]-pyrrolidine-l-carboxylic acid tert-butyl ester
Figure imgf000184_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 138 mg (0.14 mmol) (2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy- quinolin-4-yloxy]-2-{(lR,2S)-l-[2-(7-methoxycarbonyl-heptanoylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclo-propylcarbamoyl} -pyrrolidine- 1 -carboxylic acid tert-butyl ester and 35 mg (1.4 mmol) LiOH in 10 mL THF/MeOH/H2O (2:1:1). HPLC (method A) tR = 5.07 min TLC, Rf (CH2Cl2/Me0H 85:15) = 0.55 MS (method D): 948 [M+]
Step 3
8-[2-({(lR,2S)-l-[(2-Amino-indane-2-carbonyl)-amino]-2-vinyl-cyclopropanecarbonyl}- sulfamoyl)-phenylcarbamoyl]-octanoic acid
Figure imgf000185_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 3) using 135 mg (0.14 mmol) (2S,4R)-2-{(lR,2S)-l-[2-(7-Carboxy-heptanoylamino)- benzenesulfonyl-aminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-[2-(2-isopropylamino- thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-l-carboxylic acid tert-butyl ester and 1 mL TFA in 25 mL DCM. HPLC (method A) tR = 4.33 min TLC, Rf(CH2Cl2MeOH 85:15) - 0.46 MS (method D): 848 [M+]
Example 11
(8S,10R)-10-(7-Methoxy-2-phenyl-quinoIin-4-yloxy)-5-[(lR,2S)-l-carbonylamino-2- vinyl-cyclopropyl]-2,2-dioxo-2λ*6*-thia-3,6,12,22-tetraaza- tricyclo[21.4.0.0*8,12*Jheptacosa-l(27),23,25-triene-4,7,13-trione
Figure imgf000185_0002
The title compound is prepared analogously as described for the title compound in Example 2 using 80 mg (0.08 mmol) 9-{2-[((lR,2S)-l-{[(2S,4R)-4-(7-Methoxy-2-phenyl-quinolin-4- yloxy)-pyrrolidine-2-carbonyl]-amino}-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]- phenylamino}-nonanoic acid (TFA-salt), 102 mg (0.80 mmol) DEPEA and 150 mg (0.40 mmol) HATU in 25 mL DCM and 0.5 mL DMF. HPLC (method A) tR = 5.43 min TLC, Rf (CH2Cl2/MeOH 19:1) = 0.37 MS (method D): 766 [M+H2O]
Preparation of 9-{2-[((lR,2S)-l-{[(2S,4R)-4-(7-Methoxy-2-phenyl-quinolin-4-yloxy)- pyrrolidine-2-carbonyl]-amino}-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]- phenylaminoj-nonanoic acid
Step l
9-Hydroxy-nonanoic acid methyl ester
Figure imgf000186_0001
To an ice-cold solution of 10.0 g (45 mmol) Mono-methyl azelate in 250 mL THF is added 90 mL (90 mmol) BH3*THF-Komplex (IM in THF), the ice-bath is removed and stirring is continued at rt for 90 min. The reaction is quenched by careful addition of Methanol, the main solvent is evaporated, the residue is diluted with water and extracted with EtOAc. The combined organic phase is dried with Na2SO4, filtered, and the solvent is removed in vacuo to give the title compound as a colorless oil, which is used without further purification. MS (method D): 206 [M+H2O]
Step 2
9-Oxo-nonanoic acid methyl ester
Figure imgf000186_0002
To a solution of 5.2 g (28 mmol) 9-Hydroxy-nonanoic acid methyl ester in 350 mL DCM is added 9.1 g (41 mmol) Pyridinium chlorochromate and the reaction is stirred for 15 h at rt. The reaction is diluted with DCM, silica is added, the mixture is filtered through a pad of Hyflo and thoroughly washed with DCM. The solvent is removed in vacuo to give the title compound as a green oil, which is used without further purification. MS (method D): 204 [M+H2O] Step 3
9-{2-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl-cyclopropanecarbonyl)-sulfamoyI]- phenylamino}-nonanoic acid methyl ester
Figure imgf000187_0001
To a solution of 100 mg (0.26 mmol) [(lR,2S)-l-(2-Amino-benzenesulfonylaminocarbonyl)- 2-vinyl-cyclopropyl]-carbamic acid tert-butyl ester and 98 mg (0.52 mmol) 9-Oxo-nonanoic acid methyl ester in 15 mL 1 ,2-Dichloroethane is added at rt 0.045 mL (0.79 mmol) AcOH followed by 145 mg (0.67 mmol) NaBH(OAc)3. After stirring for 15 h at rt the solvent is removed in vacuo and the residue is purified by preparative reverse phase HPLC (Method G) to give the title compound as a yellow oil. HPLC (method A) tR = 5.68 min MS (method D): 552 [M+]
Step 4
9-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]-phenylamino}- nonanoic acid methyl ester
Figure imgf000187_0002
To a solution of 2.10 g (1.56 mmol) 9-{2-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl- cyclopropane-carbonyl)-sulfamoyl]-phenylamino}-nonanoic acid methyl ester in 50 mL
Dioxane is added 25 mL 4N HCl in Dioxane and the reaction is stirred for 15 h at rt. The solvent is removed in vacuo and the residue is purified by preparative reverse phase HPLC
(Method G) to give the title compound as an orange oil.
HPLC (method A) tR = 4.00 min TLC, Rf (CH2Cl2/Me0H 19:1) - 0.38
MS (method D): 452 [M+] Step 5
(2S,4R)-2-{(lR,2S)-l-[2-(8-Methoxycarbonyl-octylamino)- benzenesulfonyIamiπocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-(7-methoxy-2-phenyl- quinolin-4-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester
Figure imgf000188_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 1) using 105 mg (0.21 mmol) 9-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl]-phenylamino}-nonanoic acid methyl ester, 95 mg (0.21 mmol) (2S,4R)~4-(7- Methoxy-2-phenyl-quinolin-4-yloxy)-pyrrolidine-l,2-dicarboxylic acid 1 -tert-butyl ester, 102 mg (0.27 mmol) HATU and 133 mg (1.0 mmol) DIPEA in 5 mL DMF. HPLC (method A) tR = 5.83 min MS (method D): 898 [M+]
Step 6
(2S,4R)-2-{(lR,2S)-l-[2-(8-Carboxy-octylamino)-benzenesulfonyIaminocarbonyl]-2- vinyI-cycIopropylcarbamoyl}-4-(7-methoxy-2-phenyI-quinoIin-4-yloxy)-pyrrolidine-l- carboxylic acid tert-butyl ester
Figure imgf000188_0002
The title compound is prepared analogously as described for the title compound in Example 2
(step 2) using 73 mg (0.08 mmol) (2S,4R)-2-{(lR,2S)-l-[2-(8-Methoxycarbonyl- octylamino)-benzenesulfonyl-aminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-(7-methoxy-
2-phenyl-quinolin-4-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester and 20 mg LiOH in
8 mL THF/MeOH/H2O (2:1 :1).
HPLC (method A) tR = 5.29 min
TLC, Rf (CH2Cl2/MeOH/H2O/AcOH 90:10:1:0.5) = 0.66
MS (method D): 884 [M+]
Step 7
9-{2-[((lR,2S)-l-{[(2S,4R)-4-(7-Methoxy-2-phenyl-quinolin-4-yloxy)-pyrrolidine-2- carbonyl]-amino}-2-vinyl-cyclopropanecarbonyl)-sulfamoyI]-phenyIamiπo}-nonanoic acid
Figure imgf000189_0001
The title compound is prepared analogously as described for the title compound in Example 2
(step 3) using 71 mg (0.08 mmol) (2S,4R)-2-{(lR,2S)-l-[2-(8-Carboxy-octylamino)- benzenesulfonyl-amino-carbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-(7-methoxy-2-phenyl- quinolin-4-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester and 0.3 mL TFA in 5 mL
DCM.
HPLC (method A) tR = 4.78 min
TLC, Rf (CH2Cl2/MeOH/H2O/AcOH 90:10:1:0.5) = 0.41
MS (method D): 784 [M+]
Example 12 (8S,10R)-10-[2-(2-IsopropyIamino-thiazol-4-yl)-7-methoxy-quinoIin-4-yloxy]-5-[(lR,2S)- l-carbonylamino-2-vinyl-cyclopropyl]-2,2-dioxo-2λ*6*-thia-3,6,12,22-tetraaza- tricyclo[21.4.0.0*8,12*]heptacosa-l(27),23,25-triene-4,7,13-trioπe
Figure imgf000190_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 168 mg (0.14 mmol) 9-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)- 7-methoxy-quinolin-4-yloxy]-pyrrolidine-2-carbonyl}-amino)-2-vinyl- cyclopropanecarbonyl]-sulfamoyl}-phenylamino)-nonanoic acid (TFA-salt), 182 mg (1.4 mmol) DIPEA and 268 mg (0.71 mmol) HATU in 75 mL DCM and 1 mL DMF. HPLC (method A) tR = 5.90 min TLC, Rf (CH2Cl2/Me0H 19:1) = 0.37 MS (method D): 830 [M+]
Preparation of 9-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-Isopropylamino-thiazoI-4-yι)-7- methoxy-quiπolin-4-yloxy]-pyrrolidine-2-carbonyl}-amino)-2-vinyI- cyclopropanecarbonyl]-sulfamoyl}-phenylamino)-nonanoic acid
Step l
(2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy-quinolin-4-yIoxy]-2-{(lR,2S)-l- [2-(8-methoxycarbonyl-octylamino)-beQzenesuIfonyIaminocarbonyl]-2-vinyl- cyclopropylcarbamoyl}-pyrrolidine-l-carboxylic acid tert-butyl ester
Figure imgf000191_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 1) using 200 mg (0.44 mmol) 9-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl] -phenyl-amino }-nonanoic acid methyl ester, 234 mg (0.44 mmol) (2S,4R)-4-[2-(2- Isopropylamino-thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine- 1 ,2-dicarboxylic acid 1-tert-butyl ester, 219 mg (0.58 mmol) HATU and 287 mg (2.2 mmol) DDPEA in 5 mL DMF. HPLC (method A) tR = 6.1 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.81 MS (method D): 962 [M+]
Step 2
(IS^R^l-KlR-lS^l-^^S-Carboxy-octylamino^benzenesulfonylaminocarbonyl]-!- vinyl-cyclopropylcarbamoyI}-4-[2-(2-isopropylamino-thiazoI-4-yl)-7-methoxy-quinolin- 4-yIoxy]-pyrroIidine-l-carboxylic acid tert-butyl ester
Figure imgf000191_0002
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 174 mg (0.18 mmol) (2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy- quinolin-4-yloxy]-2-{(lR,2S)-l-[2-(8-methoxycarbonyl-octylamino)- benzenesulfonylaminocarbonyl] -2 -vinyl-cyclopropyl-carbamoyl } -pyrrolidine- 1 -carboxylic acid tert-butyl ester and 44 mg (1.81 mmol) LiOH in 14 mL THF/MeOH/H2O (2: 1 :1). HPLC (method A) tR = 5.58 min TLC, Rf (CH2Cl2/Me0H) = 0.27 MS (method D): 948 [M+]
Step 3
9-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-Isopropylamino-thiazoI-4-yl)-7-methoxy-quinolin-4- yloxy]-pyrrolidine-2-carboπyl}-amino)-2-vinyl-cyclopropanecarbonyl]-sulfamoyl}- phenylamino)-nonanoic acid
Figure imgf000192_0001
The title compound is prepared analogously as described for the title compound in Example 2
(step 3) using 135 mg (0.14 mmol) (2S,4R)-2-{(lR,2S)-l-[2-(8-Carboxy-octylamino)- benzenesulfonylamino-carbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-[2-(2-isopropylamino- thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-l-carboxylic acid tert-butyl ester and
0.6 mL TFA in 10 mL DCM.
HPLC (method A) tR = 5.20min
TLC, Rf (CHzCyMeOH/HzO/AcOH 90:10:1:0.5) - 0.19
MS (method D): 848 [M+]
Example 13
4-Fluoro-l,3-dihydro-isoindoIe-2-carboxylic acid (8S,10R)-5-[(lR,2S)-l-carbonylamino-
2-vinyI-cyclopropyl]-2,2,4,7,13-pentaoxo-2λ*6*-thia-3,6,12,22-tetraaza- tricyclo[21.4.0.0*8,12*]heptacosa-l(27),23,25-trien-10-yl ester
Figure imgf000193_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 78 mg (0.08 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-5- {( 1 R,2S)- 1 -[2-(8-carboxy-octylamino)-benzenesulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoyl}-pyrrolidin-3-yl ester (TFA-salt), 107 mg (0.83 mmol) DIPEA and 158 mg (0.42 mmol) HATU in 50 mL DCM and 1 mL DMF. HPLC (method A) tR = 5.65 min TLC, Rf (CH2Cl2/MeOH 19:1) = 0.27 MS (method D): 696 [M+]
Preparation of 4-Fluoro-l,3-dihydro-isoindole-2-carboxyIic acid (3R,5S)-5-{(lR,2S)-l- [2-(8-carboxy-octyIamino)-benzenesuIfonyIaminocarbonyl]-2-vinyl- cyclopropylcarbamoyl}-pyrrolidin-3-yl ester
Step l
4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5-
{(lR,2S)-l-[2-(8-methoxycarbonyl-octylamino)-benzenesulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoyl}-pyrroIidin-3-yl ester
Figure imgf000193_0002
The title compound is prepared analogously as described for the title compound in Example 2 (step 1) using 150 mg (0.18 mmol) 9-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl]-phenyl-amino}-nonanoic acid methyl ester, 71 mg (0.18 mmol) (2S,4R)-4-(4- Fluoro- 1 ,3 -dihydro-isoindole-2-carbonyloxy)-pyrrolidine- 1 ,2-dicarboxylic acid 1 -tert-butyl ester, 103 mg (0.27 mmol) HATU and 70 mg (0.54 mmol) DIPEA in 5 mL DCM. HPLC (method A) tR = 6.10 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.69 MS (method D): 828 [M+]
Step 2
4-Fluoro-l,3-dihydro-isoindoIe-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5-
{(lR,2S)-l-[2-(8-carboxy-octylamino)-benzenesuIfonylaniinocarbonyl]-2-vinyl- cycIopropylcarbamoyl}-pyrroIidin-3-yl ester
Figure imgf000194_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 80 mg (0.09 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)- 1 -tert-butoxycarbonyl-5- {( 1 R,2S)- 1 -[2-(8-methoxycarbonyl-octylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-pyrrolidin-3-yl ester and 36 mg (0.85 mmol) LiOH in 12 mL THF/MeOH/H2O (2:1:1). HPLC (method A) tR = 5.53 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.51 MS (method D): 814 [M+]
Step 3
4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-5-{(lR,2S)-l-[2-(8-carboxy- oct>Iamino)-benzeπesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-pyrrolidin-
3-yI ester
Figure imgf000195_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 3) using 68 mg (0.08 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)- l-tert-butoxycarbonyl-5-{(lR,2S)-l-[2-(8-carboxy-octylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-pyrrolidin-3-yl ester and 1 mL TFA in 5 mL DCM. HPLC (method A) tR = 4.74 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.35 MS (method D): 714 [M+]
Example 14
(1R,2S,2'R,25a'S)-2'-[(7-methoxy-2-phenylquinolin-4-yl)oxy]-2-vinyl-
1'H,2'H,3'H,5'H,6'H,7'H,13'H,14'H,15'H,21'H,22'H,24'H,25'H,25a'H- spiro[cyclopropane-1,23'-[20]thia[4,15,21,24]tetraaza[8,12](metheno)pyrrolo[2,1- g][1,2,5,8,19]benzothiatetraazacyclohenicosine]-5',14',22',25'-tetrone 20',20'-dioxide
Figure imgf000195_0002
The title compound is prepared analogously as described for the title compound in Example 2 using 23 mg (0.03 mmol) 3-[3-({2-[((lR,2S)-l-{[(2S,4R)-4-(7-Methoxy-2-phenyl-quinolin- 4-yloxy)-pyrrolidine-2-carbonyl]-amino}-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]- phenylcarbamoyl}-methyl)-phenyl]-propionic acid (TFA-salt), 32 mg (0.25 mmol) DIPEA and 48 mg (0.71 mmol) HATU in 10 mL DCM and 0.2 mL DMF. HPLC (method A) tR = 4.58 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.44 MS (method D): 800 [M+]
Preparation of 3-[3-({2-l((lR,2S)-l-{[(2S,4R)-4-(7-Methoxy-2-phenyl-quinolin-4-yIoxy)- pyrrolidine-2-carbonyl]-amino}-2-vinyI-cycIopropanecarbonyl)-sulfamoyl]- phenylcarbamoyl}-methyl)-phenyl]-propionic acid
Step l (E)-3-(3-CarboxymethyI-phenyl)-acrylic acid methyl ester
Figure imgf000196_0001
A microwave-vial is charged with 2.2 g (10 mmol) 3-Bromophenylacetic acid, 2.62 g (30 mmol) Methyl acrylate, 0.31 g (1.0 mmol) P(o-tol)3, 90 mg (0.4 mmol) Pd(OAc)2, amd 1.2 g (12 mmol) NEt3. The vial is purged with argon, sealed and heated in the microwave (Personal Chemistry, Emrys Optimizer) for 15 min at 150°C. After cooling to rt the mixture is diluted with water and EtOAc, filtered through a pad of Hyflo and washed thoroughly with EtOAc. The filtrate is separated, the aqueous phase is extracted with EtOAc and the combined organic phases are dried with Na2SO4, filtered and the solvent is removed in vacuo. The residue is purified by FC on silica (eluent: CH2Cl2/MeOH 98:2 -> 95:5) to give the title compound as a colorless solid. HPLC (method A) tR = 3.14 min TLC, Rf (CH2Cl2/MeOH 19:1) = 0.22 MS (method D): 221 [M+H]
Step 2 3-(3-Carboxymethyl-phenyl)-propionic acid methyl ester
Figure imgf000197_0001
A shaking flask charged with 3.9 g (16.0 mmol) (E)-3-(3-Carboxymethyl-phenyl)-acrylic acid methyl ester and 0.4 g 10% Pd/C (Engelhard 4505) in 80 mL EtOAc is purged with hydrogen and shaken for 1O h. The catalyst is removed by filtration, washed with EtOAc and the filtrate is concentrated in vacuo to give the title compound as a colorless solid which is used without further purification. HPLC (method A) tR = 2.96 min TLC, Rf(CH2Cl2MeOH 19:1) = 0.19 MS (method D): 240 [M+H20]
Step 3
3-[3-({2-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl]-phenylcarbamoyl}-methyl)-phenyl]-propionic acid methyl ester
Figure imgf000197_0002
The title compound is prepared analogously as described for the title compound in Example 1
(Step 2) using 1.0 g (2.6 mmol) [(lR,2S)-l-(2-Amino-benzenesulfonylaminocarbonyl)-2- vinyl-cyclopropyl]-carbamic acid tert-butyl ester, 2.0 g (9.0 mmol) 3-(3-Carboxymethyl- phenyl)-propionic acid methyl ester, 1.30 g (10.8 mmol) Benzotriazole, 1.30 g (10.8 mmol) Thionylchloride, 2.65 g (26 mmol) NEt3 and 100 mg DMAP in 40 mL DCM. HPLC (method A) tR = 4.90 min TLC, Rf (CH2Cl2/MeOH 19:1) = 0.36 MS (method D): 613 [M+H2O]
Step 4 3-[3-({2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)-sulfainoyl]- phenylcarbamoyl}-methyl)-phenyl]-propionic acid methyl ester
Figure imgf000198_0001
The title compound is prepared analogously as described for the title compound in Example 1 (Step 3) using 0.38 g (0.59 mmol) 3-[3-({2-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl- cyclopropane-carbonyl)-sulfamoyl] -phenylcarbamoyl } -methyl)-phenyl]-propionic acid methyl ester and 5 mL 4N HCl in Dioxane. HPLC (method A) tR = 3.09 min MS (method D): 486 [M+]
Step 5
(2S,4R)-2-[(lR,2S)-l-(2-{2-[3-(2-MethoxycarbonyI-ethyI)-phenyl]-acetylamino}- benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyI]-4-(7-methoxy-2-phenyl- quinolin-4-yloxy)-pyrroIidine-l-carboxyIic acid tert-butyl ester
Figure imgf000198_0002
The title compound is prepared analogously as described for the title compound in Example 1 (step 4) using 114 mg (0.59 mmol) 3-[3-({2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylcarbamoyl} -methyl)-phenyl] -propionic acid methyl ester, 73 mg (0.16 mmol) (2S,4R)-4-(7-Methoxy-2-phenyl-quinolin-4-yloxy)-pyrrolidine-l,2- dicarboxylic acid 1-tert-butyl esterr, 90 mg (0.24 mmol) HATU and 102 mg (0.79 mmol)
DIPEA in 5 mL DMF.
HPLC (method A) tR = 5.20 min
TLC, Rf (CH2Cl2MeOH 9: 1) = 0.38
MS (method D): 932 [M+]
Step 6
(2S,4R)-2-[(lR,2S)-l-(2-{2-[3-(2-Methoxycarbonyl-ethyl)-phenyl]-acetylamino}- benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-4-(7-methoxy-2-phenyl- quinolin-4-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester
Figure imgf000199_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 28 mg (0.03 mmol) (2S,4R)-2-[(lR,2S)-l-(2-{2-[3-(2-Methoxycarbonyl-ethyl)- phenyl]-acetyl-amino}-benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-4-(7- methoxy-2-phenyl-quinolin-4-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester and 13 mg (0.3 mmol) LiOH in 8 mL THF/MeOH/H2O (2:1:1). HPLC (method A) tR = 4.77 min TLC, Rf (CH2Cl2/MeOH 9:1) = 0.17 MS (method D): 918 [M+]
Step 7
3-[3-({2-[((lR,2S)-l-{[(2S,4R)-4-(7-Methoxy-2-phenyl-quinolin-4-yloxy)-pyrrolidine-2- carbonyl]-amino}-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]-phenylcarbamoyI}- methyl)-phenyl]-propionic acid
Figure imgf000200_0001
The title compound is prepared analogously as described for the title compound in Example 2
(step 3) using 26 mg (0.03 mmol) (2S,4R)-2-[(lR,2S)-l-(2-{2-[3-(2-Carboxy-ethyl)-phenyl]- acetylamino}-benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-4-(7- methoxy-2-phenyl-quinolin-4-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester and 1 mL
TFA in 5 mL DCM.
HPLC (method A) tR = 3.82 min
TLC, Rf (CH2Cl2/Me0H 9:1) = 0.35 '
MS (method D): 818 [M+]
Example 15
8S,10R)-10-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy-quinoIin-4-yloxy]-5-[(lR,2S)- l-carbonylamino-2-vinyl-cyclopropylJ-2,2-dioxo-16,19-dioxa-2λ*6*-thia-3,6,12,22- tetraaza-tricyclo[21.4.0.0*8,12*lheptacosa-l(27),23,25-triene-4,7,13,21-tetraone
Figure imgf000200_0002
The title compound is prepared analogously as described for the title compound in Example 2 using 20 mg (0.02 mmol) 3-{2-[2-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-Isopropylamino-thiazol- 4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-2-carbonyl}-amino)-2-vinyl- cyclopropanecarbonyl]-sulfamoyl}-phenylamino)-ethoxy]-ethoxy} -propionic acid (TFA- salt), 22 mg (0.20 mmol) DIPEA and 32 mg (0.09 mmol) HATU in 10 mL DCM and 0.2 mL DMF.
HPLC (method A) tR = 4.65 min TLC, Rf (CH2Cl2MeOH 9: 1) - 0.34 MS (method D): 834 [M+]
Preparation of 3-{2-[2-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7- methoxy-quinolin-4-yloxy]-pyrrolidine-2-carbonyl}-amino)-2-vinyl- cyclopropanecarbonylj-sulfamoyl}-phenylamino)-ethoxy]-ethoxy}-propionic aci(l
Step l 3-(2-AlIyloxy-ethoxy)-propionic acid methyl ester
Figure imgf000201_0001
To a solution of 20 g (0.19 mol) 2-Allyloxyethanol in 250 mL abs. THF is added 44 mg Sodium and the mixture is refluxed until the sodium disappears. After cooling to RT 28.3 g (0.33 mol) methyl acrylate is added and stirring is continued overnight. The solvent is removed in vacuo, 400 mL MeOH and 1 mL cone. H2SO4 is added and the mixture is refluxed overnight. The solvent is removed in vacuo and the residue is purified by FC on silica (eluent: hexane/EtOAc 3:1) to give the title compound as a colorless oil. TLC, Rf (hexane/EtOAc 3:1) = 0.48 MS (method D): 206 [M+ 18]
Step 2
3-[2-(2-Oxo-ethoxy)-ethoxy]-propionic acid methyl ester A suspension of 1.5 g (8.0 mmol) 3-(2-Allyloxy-ethoxy)-propionic acid methyl ester and 134 mg (1.6 mmol) sodium bicarbonate in 160 mL DCM is cooled to -78°C. Ozone is bubbled through until a blue color appears (-15 min). Oxygen is bubbled through the mixture for 2 min to remove excess of ozone, 2.7 g (10 mmol) PPh3 is added and stirring is continued for 1 h at -78°C. After warming to RT, the solvent is removed in vacuo and the residue is used without further purification.
Step 3
3-[2-(2-{2-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl]-phenylamino}-ethoxy)-ethoxy]-propionic acid methyl ester
Figure imgf000202_0001
The title compound is prepared analogously as described for the title compound in Example 11 (step 3) using 200 mg (0.52 mmol) [(lR,2S)-l-(2-Amino-benzenesulfonylaminocarbonyl)-
2-vinyl-cyclopropyl]-carbamic acid tert-butyl ester, 500 mg crude 3-[2-(2-Oxo-ethoxy)- ethoxy] -propionic acid methyl ester (from the previous step), 292 mg (1.31 mmol)
NaBH(OAc)3 and 94 mg (1.6 mmol) AcOH in 20 mL 1,2 DCE
HPLC (method A) tR = 4.57 min MS (method D): 556 [M+]
Step 4
3-[2-(2-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]-phenylamino}- ethoxy)-ethoxy]-propionic acid methyl ester
Figure imgf000202_0002
The title compound is prepared analogously as described for the title compound in Example 1 1 (step 4) using 485 mg (0.58 mmol) 3-[2-(2-{2-[((lR,2S)-l-tert-Butoxycarbonylamino-2- vinyl-cyclopropanecarbonyl)-sulfamoyl]-phenylamino}-ethoxy)-ethoxy]-propionic acid methyl ester and 1.5 mL TFA in 20 mL DCM. HPLC (method A) tR = 2.64 min MS (method D): 456 [M+] Step 5
(ZS^RH-fl-CZ-Isopropylamino-thiazoM-yO-T-methoxy-quinolin^-yloxyl-Z-lClRjZS)-!- (2-{2-[2-(2-methoxycarbonyI-ethoxy)-ethoxyl-ethylaniino}- benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyI]-pyrrolidine-l- carboxylic acid tert-butyl ester
Figure imgf000203_0001
The title compound is prepared analogously as described for the title compound in Example 2
(step 1) using 235 mg (0.34 mmol) 3-[2-(2-{2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylamino}-ethoxy)-ethoxy]-propionic acid methyl ester, 182 mg (0.34 mmol) (2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy- quinolin-4-yloxy]-pyrrolidine-l,2-dicarboxylic acid 1 -tert-butyl ester, 196 mg (0.52 mmol)
HATU and 134 mg (1.0 mmol) DIPEA in 20 mL DCM.
HPLC (method A) tR = 5.08 min TLC, Rf(CH2Cl2/Me0H 9:1) = 0.31
MS (method D): 966 [M+]
Step 6
(2S,4R)-2-[(lR,2S)-l-(2-{2-[2-(2-Carboxy-ethoxy)-ethoxy]-ethylamino}- benzenesulfonyIamino-carbonyl)-2-vinyl-cycIopropylcarbamoyl]-4-[2-(2- isopropylamino-thiazol^-yO-T-methoxy-quinolin^-yloxyJ-pyrrolidine-l-carboxylic acid tert-butyl ester
Figure imgf000204_0001
The title compound is prepared analogously as described for the title compound in Example 2
(step 2) using 170 mg (0.18 mmol) (2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy- quinolin-4-yloxy]-2-[( 1 R,2S)- 1 -(2- {2-[2-(2-methoxycarbonyl-ethoxy)-ethoxy]-ethylamino} - benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-pyrrolidine-l-carboxylic acid tert-butyl ester (TFA-salt) and 76 mg (1.8 mmol) LiOH in 20 mL THF/MeOH/H2O
(2:1:1).
HPLC (method A) tR = 4.79min
TLC, Rf(CH2Cl2ZMeOH 9:1) = 0.33
MS (method D): 952 [M+]
Step 7
(2S,4R)-2-[(lR,2S)-l-(2-{2-[2-(2-Carboxy-ethoxy)-ethoxy]-ethylamino}- benzenesulfonylamino-carbonyl)-2-vinyi-cyclopropylcarbamoyl]-4-[2-(2- isopropyIamino-thiazol-4-yI)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-l-carboxylic acid tert-butyl ester
Figure imgf000204_0002
The title compound is prepared analogously as described for the title compound in Example 2 (step 3) using 12 mg (0.01 mmol) (2S,4R)-2-[(lR,2S)-l-(2-{2-[2-(2-Carboxy-ethoxy)- ethoxy]-ethylamino}-benzenesulfonylamino-carbonyl)-2-vinyl-cyclopropylcarbamoyl]-4-[2-
(2-isopropylamino-thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine- 1 -carboxylic acid tert-butyl ester (TFA-salt) and 0.1 mL TFA in 3 mL DCM.
HPLC (method A) tR = 4.27 min
TLC, Rf (CH2Cl2/MeOH/H2O/AcOH 90: 10:1:0.5) = 0.26
MS (method D): 852 [M+]
Example 16
(8S,10R)-10-[2-(2-Isop^opyIaIllino-thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-5-[(lR,2S)- l-carbonylamino-2-vinyl-cyclopropyl]-2,2-dioxo-16,19-dioxa-2λ*6*-thia-3,6,12,22- tetraaza-tricyclo[21.4.0.0*8,12*]heptacosa-l(27),23,25-triene-4,7,13,21-tetraone
Figure imgf000205_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 108 mg (0.10 mmol) 3-{2-[(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-Isopropylamino-thiazol-
4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-2-carbonyl}-amino)-2-vinyl- cyclopropanecarbonylj-sulfamoyl} -phenylcarbamoyl)-methoxy]-ethoxy} -propionic acid
(TFA-salt), 128 mg (1.0 mmol) DEPEA and 188 mg (0.5 mmol) HATU in 100 mL DCM and
2 mL DMF. HPLC (method A) tR = 4.50 min
TLC, Rf (CH2Cl2/Me0H 9:1) = 0.18
MS (method D): 848 [M+]
Preparation of 33-{2-[(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-Isopropylamino-thiazoI-4-yl)-7- methoxy-quinolin-4-yIoxyJ-pyrrolidine-2-carbonyl}-amino)-2-vinyl- cyclopropanecarbonyl]-sulfamoyl}-phenylcarbamoyl)-methoxy]-ethoxy}-propionic acid Step l
3-(2-Carboxymethoxy-ethoxy)-propionic acid methyl ester
Figure imgf000206_0001
To a solution of 1.0 g (5.3 mmol) 3-(2-Allyloxy-ethoxy)-propionic acid methyl ester (according to example 15 step 1) in 50 mL CCl4ZACNZH2O (2:2:3) is added 5.68 g (27 mmol) Sodium(meta)periodate followed by 135 mg (0.27 mmol) RuCl3 monohydrate at RT. After stirring overnight the reaction is diluted with water and extracted thoroughly with DCM and the organic phase is discarded. The aq. phase is adjusted to pH 1 by addition of 4N HCl, and extracted thoroughly (12 x) with DCM. The organic phase is dried with Na2SO4, filtered and the solvent is removed in vacuo. The residue is used without further purification. TLC, Rf (CH2Cl2ZMeOH 9:1) = 0.16 MS (method D): 224 [M+18]
Step 2 3-[2-({2-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyI-cyclopropanecarbonyl)- sulfamoyl]-phenylcarbamoyl}-methoxy)-ethoxy]-propionic acid methyl ester
Figure imgf000206_0002
The title compound is prepared analogously as described for the title compound in Example 1
(Step 2) using 100 mg (0.26 mmol) [(lR,2S)-l-(2-Amino-benzenesulfonylaminocarbonyl)-2- vinyl-cyclopropyl]-carbamic acid tert-butyl ester, 200 mg (0.97 mmol) 3-(2-
Carboxymethoxy-ethoxy)-propionic acid methyl ester, 140 mg (1.2 mmol) Benzotriazole,
140 mg (1.2 mmol) Thionylchloride, 265 mg (2.6 mmol) NEt3 and 20 mg DMAP in 20 mL
DCM.
HPLC (method A) tR = 4.31 min TLC, Rf (CH2Cl2ZMeOH 19:1) = 0.56
MS (method D): 570 [M+]
Step 3 3-[2-({2-[((lR,2S)-l-Amino-2-vinyI-cyclopropanecarbonyl)-sulfamoyl]- phenylcarbamoyl}-methoxy)-ethoxy]-propionic acid methyl ester
Figure imgf000207_0001
The title compound is prepared analogously as described for the title compound in Example 11 (step 4) using 116 mg (0.20 mmol) 33-[2-({2-[((lR,2S)-l-tert-Butoxycarbonylamino-2- vinyl-cyclopropanecarbonyl)-sulfamoyl]-phenylcarbamoyl}-methoxy)-ethoxy]-propionic acid methyl ester and 0.5 mL TFA in 6 mL DCM. HPLC (method A) tR = 1.95 min TLC, Rf (CH2Cl2/Me0H 19:1) = 0.32 MS (method D): 470 [M+]
Step 4
(2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-2-[(lR,2S)-l- (2-{2-[2-(2-methoxycarbonyl-ethoxy)-ethoxy]-acetylamino}- benzenesulfonyIaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-pyrroIidine-l- carboxylic acid tert-butyl ester
Figure imgf000207_0002
The title compound is prepared analogously as described for the title compound in Example 2 (step 1) using 118 mg (0.20 mmol) 33-[2-({2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylcarbamoyl}-methoxy)-ethoxy]-propionic acid methyl ester, 107 mg (0.20 mmol) (2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy- quinolin-4-yloxy]-pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester, 115 mg (0.30 mmol) HATU and 78 mg (0.61 mmol) DIPEA in 6 mL DCM. HPLC (method A) tR = 5.05 min TLC, Rf (CH2Cl2MeOH 9:1) = 0.35 MS (method D): 980 [M+]
Step 5
(2S,4R)-2-[(lR,2S)-l-(2-{2-[2-(2-Carboxy-ethoxy)-ethoxy]-acetylamino}- benzenesulfonylamino-carbonyl)-2-vinyl-cyclopropylcarbamoyl]-4-[2-(2- isopropylamino-thiazol-4-yl)-7-methoxy-quinolin-4-yIoxy]-pyrrolidine-l-carboxylic acid tert-butyl ester
Figure imgf000208_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 110 mg (0.10 mmol) (2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy- quinolin-4-yloxy]-2-[( 1R,2S)- 1 -(2- {2-[2-(2-methoxycarbonyl-ethoxy)-ethoxy]-acetylamino } - benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-pyrrolidine-l-carboxylic acid tert-butyl ester (TFA-salt) and 43 mg (1.0 mmol) LiOH in 16 mL THF/MeOH/H2O
(2:1: 1). HPLC (method A) tR = 4.73 min
TLC, Rf (CHzCh/MeOH/HiO/AcOH 90:10:1:0.5) = 0.40
MS (method D): 966 [M+]
Step 6 3-{2-[(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy-quinolin- 4-yloxy]-pyrrolidine-2-carbonyl}-amino)-2-vinyI-cyclopropanecarbonyl]-sulfamoyI}- phenylcarbamoyl)-methoxy]-ethoxy}-propionic acid
Figure imgf000209_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 3) using 96 mg (0.10 mmol) ((2S,4R)-2-[(lR,2S)-l-(2-{2-[2-(2-Carboxy-ethoxy)- ethoxy]-acetylamino}-benzenesulfonylamino-carbonyl)-2-vinyl-cyclopropylcarbamoyl]-4-[2- (2-isopropylamino-thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine- 1 -carboxylic acid tert-butyl ester (TFA-salt) and 0.5 mL TFA in 6 mL DCM. HPLC (method A) tR = 3.92 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.42 MS (method D): 866 [M+]
Example 17
4-Fluoro-l,3-dihydro-isoindoIe-2-carboxylic acid (8S,10R)-5-[(lR,2S)-l-carbonylammo-
2-vinyI-cycIopropyI]-2,2,4,7,13-pentaoxo-16,19-dioxa-2λ*6*-thia-3,6,12,22-tetraaza- tricyclo[21.4.0.0*8,12*] heptacosa-l(27),23,25-trien-10-yl ester
Figure imgf000209_0002
The title compound is prepared analogously as described for the title compound in Example 2 using 56 mg (0.05 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-5- [(lR,2S)-l-(2-{2-[2-(2-carboxy-ethoxy)-ethoxy]-ethylamino}- benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-pyrrolidin-3-yl ester (TFA- salt), 70 mg (0.54 mmol) DIPEA and 103 mg (0.27 mmol) HATU in 50 mL DCM and 1 mL DMF.
HPLC (method A) tR = 4.52 min TLC, Rf (CH2Cl2MeOH 9: 1) = 0.45 MS (method D): 700 [M+]
Preparation of 4-FIuoro-l,3-dihydro-isoindoIe-2-carboxylic acjd (3R,5S)-5-[(lR,2S)-l- ^-^-β-^-carboxy-ethoxy^ethoxyJ-ethylaminoJ-benzenesulfonylainiiiocarboiiyl)^- vinyl-cyclopropyl-carbamoyll-pyrrolidiii-θ-yl ester
Step l
4-Fluoro-l,3-dihydro-isoindoIe-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5- ((lR,2S)-l-(2-{2-[2-(2-methoxycarbonyl-ethoxy)-ethoxy]-ethylamino}- benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-pyrrolidin-3-yl ester
Figure imgf000210_0001
The title compound is prepared analogously as described for the title compound in Example 2
(step 1) using 235 mg (0.34 mmol) 3-[2-(2-{2-[((lR,2S)-l-Amϊno-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylamino} -ethoxy)-ethoxy]-propionic acid methyl ester, 136 mg (0.34 mmol) ((2S,4R)-4-(4-Fluoro-l,3-dihydro-isoindole-2-carbonyloxy)- pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester, 196 mg (0.52 mmol) HATU and 134 mg
(1.0 mmol) DIPEA in 20 mL DCM.
HPLC (method A) tR = 5.08 min
TLC, Rf (CH2Cl2/Me0H 19:1) = 0.31 MS (method D): 832 [M+]
Step 2
4-Fiuoro-l,3-dihydro-isoindole-2-carboxyIic acid (3R,5S)-l-tert-butoxycarbonyl-5- [(lR,2S)-l-(2-{2-[2-(2-carboxy-ethoxy)-ethoxy]-ethylamino}- benzenesulfonylaininocarbonyl)-2-vinyl-cyclopropyIcarbamoyl]-pyrroIidin-3-yI ester
Figure imgf000211_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 170 mg (0.18 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5-[(lR,2S)-l-(2-{2-[2-(2-methoxycarbonyl-ethoxy)-ethoxy]- ethylamino}-benzene-sulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-pyrrolidin-3- yl ester (TFA-salt) and 76 mg (1.8 mmol) LiOH in 20 mL THF/MeOH/H2O (2:1:1). HPLC (method A) tR = 4.78 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.33 MS (method D): 818 [M+]
Step 3
4-Fluoro-l,3-dihydro-isoindole-2-carboxyIic acid (3R,5S)-5-[(lR,2S)-l-(2-{2-[2-(2- carboxy-ethoxy)-ethoxy]-ethylamino}-benzenesulfonylaminocarbonyl)-2-vinyl- cycIopropyIcarbamoyl]-pyrroIidin-3-yl ester
Figure imgf000211_0002
The title compound is prepared analogously as described for the title compound in Example 2 (step 3) using 52 mg (0.06 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)- l-tert-butoxycarbonyl-5-[(lR,2S)-l-(2-{2-[2-(2-carboxy-ethoxy)-ethoxy]-ethylamino}- benzenesulfonylamino-carbonyl)-2-vinyl-cyclopropylcarbamoyl]-pyrrolidin-3-yl-ester (TFA- salt) and 0.2 mL TFA in 3 mL DCM. HPLC (method A) tR = 3.85 min TLC, Rf (CH2Cl2/MeOH 9:1) = 0.25 MS (method D): 718 [M+]
Example 18
4-Fluoro-l,3-dihydro-isoindole-2-carboxyIic acid (8S,10R)-5-[(lR,2S)-l-carbonylamino- 2-vinyl-cyclopropyl]-2,2,4,7,13-pentaoxo-15,19-dioxa-2λ*6*-thia-3,6,12,22-tetraaza- tricyclo[21.4.0.0*8,12*]heptacosa-l(27),23,25-trien-10-yl ester
Figure imgf000212_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 8 mg (0.008 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-5-
(( 1 R,2S)- 1 - {2-[2-(3-carboxymethoxy-propoxy)-ethylamino]- benzenesulfonylaminocarbonyl}-2-vinyl-cyclopropylcarbamoyl)-pyrrolidin-3-yl ester (TFA- salt), 10 mg (0.08 mmol) DIPEA and 15 mg (0.04 mmol) HATU in 25 mL DCM and 0.5 mL
DMF.
HPLC (method A) tR = 4.63 min
TLC, Rf (CH2Cl2/MeOH 9:1) = 0.54 MS (method D): 700 [M+]
Preparation of 4-Fluoro-l,3-dihydro-isoindole-2-carboxyIic acid (3R^5S)-5-((lR,2S)-l- {2-[2-(3-carboxymethoxy-propoxy)-ethylamino]-benzenesulfonylaminocarbonyI}-2- vinyl-cyclopropyl-carbamoyl)-pyrrolidin-3-yl ester
Step l (3-AHyloxy-propoxy)-acetic acid
Figure imgf000213_0001
To an ice-cold solution of 7.8 g (67 mmol) 3-Allyloxy-propan-l-ol (prepared according to Synth. Comm. 1992, 22, 189-200) in 250 mL abs. THF is added 12.7 g (61 mmol) Sodium iodocaetate followed by 5.4 g (134 mmol) NaH (60% suspension in mineral oil). The ice-bath is removed and the reaction is refluxed for 5 h. After cooling to RT the reaction is quenched by addition of water and THF is removed in vacuo. The aq. phase is adjusted to pH 1 with 4 N HCl and extracted with DCM. The organic phase is washed with brine, dried with Na2SO4, filtered, the solvent is removed in vacuo and the residue is purified by FC on silica (eluent: CH2Cl2/Me0H 85:15) to give the title compound as a yellow oil. TLC, Rf (CH2Cl2/Me0H 85:15) = 0.62 MS (method D): 175 [M+H]
Step 2
(3-Allyloxy-propoxy)-acetic acid methyl ester
Figure imgf000213_0002
To a solution of 7.5 g (43 mmol) (3-Allyloxy-propoxy)-acetic acid in 300 mL acetone is added 6.9 g (68 mmol) KHCO3 followed by 6.7 mL (108 mmol) Iodomethane and the reaction is refluxed for 3 h. Additional 6.7 mL (108 mmol) Iodomethane is added and reflux is continued for 3 h. A third portion of 6.7 mL (108 mmol) Iodomethane is added and the mixture is refluxed overnight. After cooling to RT the reaction mixture is filtered and the solvent is removed in vacuo. The residue is taken up in EtOAc, washed with sat. NaHCO3- solution and brine, dried with Na2SO4, filtered and the solvent is removed in vacuo. The residue is used without further purification. TLC, Rf (CH2Cl2/Me0H 19:1) = 0.78 MS (method D): 206 [M+18]
Step 3
[3-(2-Oxo-ethoxy)-propoxy]-acetic acid methyl ester
Figure imgf000213_0003
A solution of 2.0 g (1 1.0 mmol) (3-Allyloxy-propoxy)-acetic acid methyl ester in 200 mL DCM is cooled to -78°C. Ozone is bubbled through until a blue color appears (-30 min). Oxygen is bubbled through the mixture for 2 min to remove excess of ozone, 1.0 mL (14 mmol) Dimethylsulfide is added and stirring is continued for 1 h at -78°C. After warming to RT, the solvent is removed in vacuo and the residue is used without further purification.
Step 4
[3-(2-{2-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyI-cyclopropanecarbonyl)- sulfamoyl]-phenylamino}-ethoxy)-propoxy]-acetic acid methyl ester
Figure imgf000214_0001
The title compound is prepared analogously as described for the title compound in Example 11 (step 3) using 1.9 g (5 mmol) [(lR,2S)-l-(2-Amino-benzenesulfonylaminocarbonyl)-2- vinyl-cyclopropyl]-carbamic acid tert-butyl ester, 2.56 g crude [3-(2-Oxo-ethoxy)-propoxy]- acetic acid methyl ester (from the previous step), 3.3 g (15 mmol) NaBH(OAc)3 and 0.90 g (15 mmol) AcOH in 150 mL 1,2 DCE. MS (method D): 556 [M+]
Step 5
[3-(2-{2-[((lR,2S)-l-Amino-2-vinyl-cydopropanecarbonyI)-sulfamoyl]-phenylamino}- ethoxy)-propoxy]-acetic acid methyl ester
Figure imgf000214_0002
The title compound is prepared analogously as described for the title compound in Example 11 (step 4) using 1.78 g (3.2 mmol) [3-(2-{2-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl- cyclopropane-carbonyl)-sulfamoyl]-phenylamino}-ethoxy)-propoxy]-acetic acid methyl ester and 5 mL TFA in 25 mL DCM. MS (method D): 456 [M+]
Step 6
^Fluoro-l-S-dihydro-isoindole^-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5-
((lR,2S)-l-{2-[2-(3-methoxycarbonylmethoxy-propoxy)-ethylamino]- benzenesulfonylaminocarbonyI}-2-vinyI-cyclopropylcarbamoyl)-pyrrolidin-3-yl ester
Figure imgf000215_0001
The title compound is prepared analogously as described for the title compound in Example 2
(step 1) using 520 mg (0.38 mmol) [3-(2-{2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylamino}-ethoxy)-propoxy]-acetic acid methyl ester
(TFA-salt), 150 mg (0.38 mmol) (((2S,4R)-4-(4-Fluoro-l,3-dihydro-isoindole-2- carbonyloxy)-pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester, 217 mg (0.57 mmol)
HATU and 295 mg (2.3 mmol) DIPEA in 10 mL DCM.
HPLC (method A) tR = 5.23 min TLC, Rf (CH2Cl2/Me0H 9: 1) = 0.63
MS (method D): 832 [M+]
Step 7
4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5- ((lR,2S)-l-{2-[2-(3-carboxymethoxy-propoxy)-ethyIamino]- benzenesulfonylaminocarbonyl}-2-vinyl-cycIopropylcarbamoyl)-pyrrolidin-3-yl ester
Figure imgf000216_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 28 mg (0.015 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)- 1 -tert-butoxycarbonyl-5-(( 1R,2S)- 1 - {2-[2-(3-methoxycarbonylmethoxy-propoxy)- ethylamino]-benzene-sulfonylaminocarbonyl}-2-vinyl-cyclopropylcarbamoyl)-pyrrolidin-3- yl ester (TFA-salt) and 7 mg (0.3 mmol) LiOH in 20 mL THF/MeOH/H2O (2:1:1). HPLC (method A) tR - 4.82 min TLC, Rf(CH2Cl2ZMeOH 9:1) - 0.29 MS (method D): 818 [M+]
Step 8
4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-5-((l R,2S)-l-{2-[2-(3- carboxymethoxy-propoxy)-ethylamino]-benzenesulfonylaininocarbonyl}-2-vinyl- cyclopropyIcarbamoyl)-pyrrolidin-3-yl ester
Figure imgf000216_0002
The title compound is prepared analogously as described for the title compound in Example 2 (step 3) using 8 mg (0.01 mmol) (4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)- l-tert-butoxycarbonyl-5-((lR,2S)-l-{2-[2-(3-carboxymethoxy-propoxy)-ethylamino]- benzenesulfonyl-aminocarbonyl} -2-vinyl-cyclopropylcarbamoyl)-pyrrolidin-3-yl ester (TFA- salt) and 0.2 mL TFA in 1 mL DCM. HPLC (method A) tR = 3.88 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.44 MS (method D): 718 [M+]
Example 19
(8S,10R)-10-[2-(2-IsopropyIamino-thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-5-[(lR,2S)- l-carbonylamino-2-vinyl-cyclopropyl]-17-methyl-2,2-dioxo-2λ*6*-thia-3,6,12,17,22- pentaaza-tricyclo [21.4.0.0*8,12*] heptacosa-1 (27),23,25-triene-4,7, 13-trione
Figure imgf000217_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 330 mg (0.16 mmol) 4-{[4-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-Isopropylamino-thiazol- 4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-2-carbonyl}-amino)-2-vinyl- cyclopropanecarbonyl]-sulfamoyl} -phenylamino)-butyl]-methyl-amino} -butyric acid (TFA- salt), 0.29 mL (1.64 mmol) DIPEA and 312 mg (0.82 mmol) HATU in 40 mL DCM and 1 mL DMF. HPLC (method A) tR = 4.24 min MS (method D): 845 [M+]
Preparation of 4-{I4-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7- methoxy-quinolin-4-yloxy]-pyrrolidine-2-carbonyl}-amino)-2-vinyI- cyclopropanecarbonylj-sulfamoyl}-phenylaπiino)-butyl]-methyl-amino}-butyric acid
Step 1
4-Methylamino-butyric acid methyl ester
Figure imgf000217_0002
A solution of 2.3 g (15 mmol) 4-Methylamino-butyric acid hydrochloride and 25 mL (31 mmol) HCl (1.25 M in MeOH) in 150 mL MeOH is stirred overnight at RT. The solvent is removed in vacuo and the residue is used without further purification. MS (method D): 132 [M+H]
Step 2
4-{[4-(tert-Butyl-dimethyl-silanyloxy)-butyI]-methyl-amino}-butyric acid methyl ester
Figure imgf000218_0001
The title compound is prepared analogously as described for the title compound in Example 11 (step 3) using 1.6 g (9.5 mmol) 4-Methylamino-butyric acid methyl ester hydrochloride, 1.93 g (9.5 mmol) 4-(tert-Butyl-dimethyl-silanyloxy)-butyraldehyde (prepared according to J. Org. Chem. 2005, 70(6), 2097), 4.50 g (19 mmol) NaBH(OAc)3 and 1.1 mL (19 mmol) AcOH in 10O mL 1,2 DCE. MS (method D): 318 [M+]
Step 3
4-[(4-Hydroxy-butyI)-methyl-amino]-butyric acid methyl ester
Figure imgf000218_0002
To an ice-cold solution of 2.1 g (6.6 mmol) 4-{[4-(tert-Butyl-dimethyl-silanyloxy)-butyl]- methyl-amino} -butyric acid methyl ester in 10 mL abs. THF is slowly added 7.9 mL (7.9 mmol) TBAF (IM in THF). After 2 h at RT additional 2 mL TBAF is added stirring is continued for 2 h, the solvent is removed in vacuo and the residue is purified by FC on silica (eluent: TBME/MeOH/NHUOH 90:10:1) to give the title compound as a brown oil. MS (method D): 204 [M+H]
Step 4
4-[Methyl-(4-oxo-butyl)-amino]-butyric acid methyl ester
Figure imgf000218_0003
To a solution of 100 mg (0.47 mmol) 4-[(4-Hydroxy-butyl)-methyl-amino]-butyric acid methyl ester in 2 mL DCM is added 220 mg (0.98 mmol) PCC. After stirring overnight at RT, the solvent is removed in vacuo and the residue is purified by FC on silica (eluent: TBME/MeOH/NH4OH 85:15:1) to give the title compound as a brown oil. TLC, Rf (TBME/MeOH/NH4OH 90:10:1) = 0.30
Step 5
4-[(4-{2-[((lR,2S)-l-tert-Butoxycarbonylainino-2-vinyl-cyclopropanecarbonyl)- sulfamoyl]-phenylamino}-butyl)-methyl-amino]-butyric acid methylester
Figure imgf000219_0001
The title compound is prepared analogously as described for the title compound in Example
11 (step 3) using 2.0 g (5.2 mmol) (lR,2S)-l-(2-Amino-benzenesulfonylaminocarbonyl)-2- vinyl-cyclopropyl]-carbamic acid tert-butyl ester, 1.27 g (6.3 mmol) 4-[Methyl-(4-oxo- butyl)-amino]-butyric acid methyl ester, 3.1 g (13 mmol) NaBH(OAc)3 and 0.90 mL (16 mmol) AcOH in 80 mL 1 ,2-Dichloroethane.
HPLC (method B) tR = 5.67 min
MS (method D): 567 [M+]
Step 6
4-[(4-{2-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)-sulfamoyI]-phenylamino}- butyl)-methyl-amino]-butyric acid methyl ester
Figure imgf000219_0002
The title compound is prepared analogously as described for the title compound in Example 11 (step 4) using 210 mg (0.37 mmol) (4-[(4-{2-[((lR,2S)-l-teit-Butoxycarbonylamino-2- vinyl-cyclopropanecarbonyl)-sulfamoyl]-phenylamino}-butyl)-methyl-amino]-butyric acid methylester and 1.4 mL TFA in 15 mL DCM. MS (method D): 467 [M+]
Step 7 (2S,4R)-2-[(lR,2S)-l-(2-{4-[(3-MethoxycarbonyI-propyl)-methyl-amino]-butylamino}- benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-4-[7-methoxy-2-(2- isopropyIamino-thiazoI-4-yl)-quinolin-4-yloxyJ-pyrroIidine-l-carboxylic acid tert-butyl ester
Figure imgf000220_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 1) using 244 mg (0.37 mmol) 4-[(4-{2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylamino} -butyl)-methyl-amino]-butyric acid methyl ester, 210 mg (0.37 mmol) (2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy- quinolin-4-yloxy]-pyrrolidine-l,2-dicarboxylic acid 1 -tert-butyl ester, 212 mg (0.56 mmol) HATU and 0.39 mL (2.23 mmol) DIPEA in 5 mL DCM. HPLC (method B) tR = 5.95 min MS (method D): 977.5 [M+]
Step 8
(2S,4R)-2-[(lR,2S)-l-(2-{4-[(3-Carboxy-propyl)-methyl-araino]-butylamino}- benzenesulfonylaminocarbonyl)-2-vinyI-cyclopropyIcarbamoyl]-4-[2-(2-isopropyIamino- thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrroIidine-l-carboxylic acid tert-butyl ester
Figure imgf000221_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 160 mg (0.16 mmol) (2S,4R)-2-[(lR,2S)-l-(2-{4-[(3-Methoxycarbonyl- propyl)-methyl-amino]-butylamino}-benzenesulfonylaminocarbonyl)-2-vinyl- cyclopropylcarbamoyl]-4-[7-methoxy-2-(2-isopropylamino-thiazol-4-yl)-quinolin-4-yloxy]- pyrrolidine-1-carboxylic acid tert-butyl ester (TFA-salt) and 35 mg (0.82 mmol) LiOH in 10 mL THF/MeOH/H2O (2:1:1). HPLC (method B) tR = 6.06 min MS (method D): 963 [M+]
Step 9
4-{ [4-(2-{ [(I R,2S)-1 -({(2S,4R)-4-[2-(2-Isopropylamino-thiazoI-4-yI)-7-methoxy-quinolin-
4-yIoxy]-pyrrolidine-2-carbonyl}-amino)-2-vinyl-cyclopropanecarbonyl]-suIfamoyl}- phenylamino)-buryl]-methyl-amino}-butyric acid
Figure imgf000221_0002
The title compound is prepared analogously as described for the title compound in Example 2 (step 3) using 176 mg (0.16 mmol) (2S,4R)-2-[(lR,2S)-l-(2-{4-[(3-Carboxy-propyl)-methyl- amino]-butylamino}-benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-4-[2- (2-isopropylamino-thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine- 1 -carboxylic acid tert-butyl ester (TFA-salt) and 0.8 mL TFA in 10 mL DCM. HPLC (method B) tR = 5.66 min MS (method D): 863 [M+]
Example 20
4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (8S,10R)-5-[(lR,2S)-l-carbonylamino-
2-vinyl-cyclopropyl]-17-methyI-2,2,4,7,13-pentaoxo-2λ*6*-thia-3,6,12,17,22-pentaaza- tricyclo[21.4.0.0*8,12* ] heptacosa-1 (27),23,25-trien-l 0-yI ester
Figure imgf000222_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 300 mg (0.19 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-5- [(lR,2S)-l-(2-{4-[(3-carboxy-propyl)-methyl-amino]-butylamino}- benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-pyrrolidin-3-yl ester (TFA- salt), 0.32 mL (1.9 mmol) DIPEA and 361 mg (0.95 mmol) HATU in 50 mL DCM and 1 mL DMF.
HPLC (method A) tR = 4.10 min MS (method D): 711 [M+]
Preparation of 4-Fluoro-l,3-dihydro-isoindoIe-2-carboxyIic acid (3R,5S)-5-[(lR,2S)-l- (2-{4-[(3-carboxy-propyl)-methyl-aitiino]-butylamino}-benzenesulfonylainiαocarbonyl)- 2-vinyl-cyclopropyIcarbamoyl]-pyrroIidin-3-yI ester
Step l
4-FIuoro-l,3-dihydro-isoindoIe-2-carboxyIic acid (3R,5S)-l-tert-butoxycarbonyI-5-
[(lR,2S)-l-(2-{4-[(3-methoxycarbonyl-propyI)-methyl-aminol-butylainino}- benzenesuIfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]-pyrrolidin-3-yl ester
Figure imgf000223_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 1) using 250 mg (0.29 mmol) 4-[(4-{2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylamino} -butyl)-methyl-amino] -butyric acid methyl ester, 110 mg (0.28 mmol) (2S,4R)-4-(4-Fluoro-l,3-dihydro-isoindole-2-carbonyloxy)- pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester, 159 mg (0.42 mmol) HATU and 0.29 mL (1.7 mmol) DIPEA in 5 mL DCM. HPLC (method B) tR = 6.52 min MS (method D): 843 [M+]
Step 2
4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5-
[(lR,2S)-l-(2-{4-[(3-methoxycarbonyI-propyl)-methyl-amino]-butylamino}- benzenesuIfonyIaminocarbonyl)-2-vinyI-cyclopropyIcarbamoyl]-pyrrolidin-3-yl ester
Figure imgf000223_0002
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 160 mg (0.19 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5-[(lR,2S)-l-(2-{4-[(3-methoxycarbonyl-propyl)-methyl- amino]-butylamino}-benzene-sulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]- pyrrolidin-3-yl ester (TFA-salt) and 32 mg (0.76 mmol) LiOH in 10 mL THF/MeOH/H2O (2:1 :1).
HPLC (method B) tR = 6.31 min
MS (method D): 829 [M+]
Step 3
4-Fluoro-l,3-dihydro-isoindole-2-carboxyIic acid (3R,5S)-5-[(lR,2S)-l-(2-{4-[(3- carboxy-propyty-methyl-aminoj-butylaminoj-benzenesulfonylaminocarboiiyl^-vinyl- cyclopropylcarbamoyl]-pyrroIidin-3-yl ester
Figure imgf000224_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 3) using 190 mg (0.19 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5-[(lR,2S)-l-(2-{4-[(3-methoxycarbonyl-propyl)-methyl- amino]-butylamino}-benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropylcarbamoyl]- pyrrolidin-3-yl ester (TFA-salt) and 0.8 mL TFA in 10 mL DCM. HPLC (method B) tR = 5.94 min MS (method D): 729 [M+]
Example 21
{(8S,10R,14S)-10-[2-(2-IsopropyIamino-thiazol-4-yl)-7-methoxy-quinolin-4-yIoxy]-5- [(lR,2S)-l-carbonylamino-2-vinyl-cyclopropyl]-2,2,4,7,13-pentaoxo-2λ*6*-thia-
3,6,12,22-tetraaza-tricyclo[21.4.0.0*8,12*]heptacosa-l(23),24,26-trien-14-yl}-carbamic acid cyclopentyl ester
Figure imgf000225_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 120 mg (0.07 mmol) (S)-2-Cyclopentyloxycarbonylamino-9-(2-{[(lR,2S)-l-({(2S,4R)- 4-[2-(2-isopropyl-amino-thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-2-carbonyl}- amino)-2-vinyl-cyclopropane-carbonyl]-sulfamoyl} -phenylamino)-nonanoic acid (TFA-salt), 92 mg (0.71 mmol) DIPEA and 135 mg (0.36 mmol), HATU in 50 mL DCM and 1 mL DMF. HPLC (method A) tR = 6.03 min TLC, Rf (CH2Cl2MeOH 9:1) = 0.58 MS (method D): 957 [M+]
Preparation of (S)-2-Cyclopentyloxycarbonylamino-9-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2- isopropyl-amino-thiazol-4-yl)-7-methoxy-quinoIin-4-yloxy]-pyrrolidine-2-carbonyI}- amino)-2-vinyl-cycIopropane-carbonyl]-sulfamoyI}-phenyIamino)-nonanoic acid
Step 1
(2S,5R)-3,6-Diethoxy-2-hept-6-enyl-5-isopropyI-2,5-dihydro-pyrazine
Figure imgf000225_0002
A solution of 26.2 g (123 mmol) of (R)-3,6-Diethoxy-2-isopropyl-2,5-dihydro-pyrazine in 450 ml of abs. THF under argon is cooled to -75°C and 77 mL (123 mmol) n-BuLi (1.6 M in Toluene) is added within 45 min while the temperature is maintained below -700C. A solution of 15 g (85 mmol) of 7-bromo-l-heptene in 80 ml of THF abs is added at -70°C. The reaction mixture is stirred for 3h at -70°C, for 17h at -4°C and for 3h at RT. Ice-cold saturated NH4Cl (70 ml) and H2O (500 ml) are added and the resulting mixture is extracted with EtOAc (500 ml). The organic layer is washed with H2O. The combined aqueous phases are extracted with EtOAc (500 ml). The combined organic phases are dried over Na2SO4, concentrated in vacuo and the residue purified by FC on silica gel. (eluent: Hexane/EtOAc 30: 1) to give the title compound as a yellow oil. TLC, Rf (Hexane/ EtOAc 30:1) = 0.46 MS (method D): 309 [M+H]
Step 2 (S)-2-Amino-non-8-enoic acid ethyl ester
Figure imgf000226_0001
To a solution of 19 g (62 mmol) (2S,5R)-3,6-Diethoxy-2-hept-6-enyl-5-isopropyl-2,5- dihydro-pyrazine in 400 mL ACN at RT, is added 250 mL of IN aq HCl. The reaction mixture is stirred for 2 h at RT. Saturated aq. NaHCO3 (250 mL) is added to adjust pH 8. The reaction mixture is stirred overnight at RT and then concentrated in vacuo. The aq. phase is extracted with 500 mL of EtOAc. The organic phase is washed twice with 250 mL H2O, dried over Na2SO4, concentrated in vacuo and the residue is purified on silica gel. (eluent: EtOAc). The product is distilled under high vacuum to give the title compound (S)-2-Amino-non-8- enoic acid ethyl ester as a colorless oil. TLC, Rf (Hexane/ EtOAc 1:2) - 0.21 MS (method D): 200 [M+H]
Step 3 (S)-2-Cyclopentyloxycarboxycarbonylamino-non-8-enoic acid ethyl ester
Figure imgf000227_0001
To a solution of 9.3 mL (100 mmol) of cyclopentanol in 200 mL of THF abs under nitrogen atmosphere at 10°C, is added over a 20-min period 89 mL (169 mmol)) of a phosgen solution (20% in Toluene). The reaction mixture is warmed up to RT and stirred for 2h, while a nitrogen stream is passed through the solution, so that the reaction volume is concentrated to 150 mL. A solution of 8.0 g (41mmol) of (S)-2-amino-non-8-enoic acid ethyl ester in 20 mL abs. THF is added at RT, followed by triethylamine added at 0°C until pH 9.4 is adjusted. The reaction mixture is stirred for Ih at 0°C and concentrated in vacuo. EtOAc (500 mL) is added and the organic layer is washed 3x with H2O (100 mL), with NaHCO3 (100 mL) and with brine (100 mL). The organic layer is dried over Na2SO4, concentrated in vacuo, and the residue is purified by FC on silica gel. (Eluent: Hexane/EtOAc 7:1) to give the title compound as a yellow oil. TLC, Rf (Hexane/ EtOAc 3:1) = 0.33 MS (method D): 312 [M+H]
Step 4 (S)-2-Cyclopentyloxycarbonylamino-non-8-enoic acid
Figure imgf000227_0002
To a solution of 460 g (1.5 mol) of (S)-2-Cyclopenthyloxycarboxycarbonylamino-non-8- enoic acid ethyl ester in 4.0 L of THF 1.8 L of Methanol is added at RT. A solution of 137g (3.25 mol) of LiOH monohydrate in 1.8 L of water is added over a 40-min period. The reaction mixture is stirred at RT for 3 h, concentrated in vacuo, taken up in H2O (2L), washed with 10% aqueous citric acid (2.5 L) and extracted with EtOAc (2.5 L). The organic layer is washed with H2O (2x 2 L) and brine (2 L). The organic layer is dried over Na2SO4, concentrated in vacuo and the residue purified by FC on silica gel (eluent: Hexane/EtOAc 10:1 -> EtOAc) to give the title compound as a red amorphous solid. TLC, Rf (CH2C12/MeOH 9:1) = 0.3 MS (method D): 282 [M-H]
Step 5
(S)-2-Cyclopentyloxycarbonylamino-non-8-enoic acid methyl ester
Figure imgf000228_0001
To a solution of 11.5 g (41 mmol) (S)-2-Cyclopentyloxycarbonylamino-non-8-enoic acid in 200 mL Acetone is added at it 6.5 g (65 mmol) KHCO3 and 14.4 g (101 mmol) Iodomethane and the reaction is reflux ed for 15 h. After cooling to rt the reaction mixture is filtered, washed with Acetone and the solvent removed in vacuo. The residue is dissolved in EtOAc, washed with aq. bicarbonate and brine, dried with Na2SO4, filtered and the solvent is removed in vacuo. The residue is purified by FC on silica (eluent: CH2Cl2/Me0H 99:1 -> 95:5) to give the title compound as a yellow oil. HPLC (method A) tR = 5.29 min TLC, Rf (CH2Cl2/MeOH 99:1) = 0.50 MS (method D): 298 [M+H]
Step 6
(S)-2-Cyclopentyloxycarbonylamino-9-hydroxy-nonanoic acid methyl ester
Figure imgf000228_0002
To an ice-cold solution of 8.1 g (27 mmol) (S)-2-Cyclopentyloxycarbonylamino-non-8-enoic acid methyl ester in 200 mL THF is added 82 mL (41 mmol) 9-BBN (0.5 M in THF) and the ice-bath is removed. After stirring for 2 h the reaction is cooled to 0°C and quenched by addition of 25 mL aq. bicarbonate and 5 mL aq. 35% H2O2. After extraction with EtOAc, the combined organic phase is dried with Na2SO4, filtered and the solvent is removed in vacuo. The residue is purified by FC on silica (eluent: CH2Cl2ZMeOH 98:2 -> 95:5) to give the title compound as a colorless oil. HPLC (method A) tR = 3.95 min TLC, Rf (CH2Cl2/Me0H 19:1) = 0.34 MS (method D): 316 [M+H]
Step 7 (S)-2-CyclopentyIoxycarbonylamino-9-oxo-nonanoic acid methyl ester
Figure imgf000229_0001
To a solution of 2.2 g (7.0 mmol) (S)-2-Cyclopentyloxycarbonylamino-9-hydroxy-nonanoic acid methyl ester in 150 mL DCM is added 2.3 g (10.5 mmol) PCC. After stirring for 4 h at rt silica is added, the reaction is filtered through a pad of Hyflo and thoroughly washed with DCM. The solvent is removed in vacuo to give the title compound as a brown oil, which is used without further purification. TLC, Rf (CH2Cl2/MeOH 19:1) = 0.54 MS (method D): 314 [M+H]
Step 8
(S)-9-{2-[((lR,2S)-l-tert-ButoxycarbonyIamino-2-vinyI-cyclopropanecarbonyI)- sulfamoyl]-phenylamino}-2-cyclopentyloxycarbonylarnino-nonanoic acid methyl ester
Figure imgf000229_0002
The title compound is prepared analogously as described for the title compound in Example 11 (step 3) using 0.95 g (2.5 mmol) lR,2S)-l-(2-Amino-benzenesulfonylaminocarbonyl)-2- vinyl-cyclopropyl]-carbamic acid tert-butyl ester, 1.95 g (4.98 mmol) (S)-2- Cyclopentyloxycarbonylamino-9-oxo-nonanoic acid methyl ester, 1.58 g (7.5 mmol) NaBH(OAc)3 and 0.43 mL (7.5 mmol) AcOH in 100 mL 1,2-Dichloroethane. HPLC (method A) tR = 5.76 min TLC, Rf (CH2Cl2MeOH 19:1) = 0.33 MS (method D): 679 [M+]
Step 9
(SJ-P-II-I^lRjZSVl-Amino-l-vinyl-cyclopropanecarbonylJ-sulfamoyll-phenylamino}-!- cyclopentyloxycarbonylamino-nonanoic acid methyl ester
Figure imgf000230_0001
The title compound is prepared analogously as described for the title compound in Example
11 (step 4) using 310 mg (0.46 mmol) (S)-9-{2-[((lR,2S)-l-tert-Butoxycarbonylamino-2- vinyl-cyclopropane-carbonyty-sulfamoylj-phenylarnino} -2-cyclopentyloxycarbonylamino- nonanoic acid methyl ester and 1 mL TFA in 10 mL DCM.
HPLC (method A) tR = 4.27 min
TLC, Rf (CH2Cl2/MeOH 9:1) = 0.59
MS (method D): 579 [M+]
Step 10
(2S,4R)-2-{(lR,2S)-l-[2-((S)-8-Cyclopentyloxycarbonylamino-8-methoxycarbonyl- octylamino)-benzenesuIfonylaminocarbonyI]-2-vinyl-cyclopropylcarbamoyl}-4-[2-(2- isopropyl-amino-thiazoM-yO-T-methoxy-quinolin^-yloxyJ-pyrroHdine-l-carboxylic acid tert-butyl ester
Figure imgf000231_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 1) using 185 mg (0.32 mmol) (S)-9-{2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylamino}-2-cyclopentyloxycarbonylamino-nonanoic acid methyl ester, 170 mg (0.32 mmol) (2S,4R)-4-[2-(2-Isopropylamino-thiazol-4-yl)-7- methoxy-quinolin-4-yloxy]-pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester, 183 mg (0.48 mmol) HATU and 124 mg (0.96 mmol) DIPEA in 10 mL DCM. HPLC (method A) tR = 6.12 min TLC, Rf (CH2Cl2/Me0H 19:1) = 0.20 MS (method D): 1089 [M+]
Step 11
(2S,4R)-2-{(lR,2S)-l-[2-((S)-8-Carboxy-8-cyclopentyIoxycarbonylamino-octyIamino)- benzenesulfonylaminocarbonyl]-2-vinyI-cyclopropylcarbamoyl}-4-[2-(2-isopropyl- amino-thiazoM-yO-T-methoxy-quinolin-^-yloxyJ-pyrrolidine-l-carboxylic acid tert- butyl ester
Figure imgf000232_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 205 mg (0.14 mmol) (2S,4R)-2-{(lR,2S)-l-[2-((S)-8- Cyclopentyloxycarbonylamino-8-methoxy-carbonyl-octylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-[2-(2-isopropyl-amino- thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-l-carboxylic acid tert-butyl ester (TFA-salt) and 59 mg (1.4 mmol) LiOH in 16 mL THF/MeOH/H2O (2:1:1). HPLC (method A) tR = 5.72 min TLC, Rf(CH2Cl2MeOH 9:1) = 0.34 MS (method D): 1075 [M+]
Step 12
(S)-2-CyclopentyIoxycarbonyIamino-9-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-isopropylainino- thiazoI-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-2-carbonyl}-amino)-2-vinyl- cyclopropanecarbonyl]-sulfamoyl}-phenylamino)-nonanoic acid
Figure imgf000233_0001
The title compound is prepared analogously as described for the title compound in Example 2
(step 3) using 119 mg (0.09 mmol) (2S,4R)-2-{(lR,2S)-l-[2-((S)-8-Carboxy-8- cyclopentyloxycarbonylamino-octylamino)-benzenesulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoyl}-4-[2-(2-isopropyl-amino-thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]- pyrrolidine-1-carboxylic acid tert-butyl ester (TFA-salt) and 0.5 mL TFA in 5 mL DCM.
HPLC (method A) tR = 5.33 min
TLC, Rf (CH2Cl2/MeOH/H2O/AcOH 90:10:1 :0.5) = 0.46
MS (method D): 975 [M+]
Example 22
4-Fluoro-l,3-dihydro-isoindoIe-2-carboxylic acid (8S,10R,14S)-14- cyclopentyloxycarbonyI-amino-5-[(lR,2S)-l-carbonylamino-2-vinyl-cyclopropyl]-
2,2,4,7,13-pentaoxo-2λ*6*-thia-3,6,12,22-tetraaza-tricyclo[21.4.0.0*8,12*]heptacosa- l(23),24,26-trien-10-yl ester
Figure imgf000234_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 217 mg (0.21 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-5-
{(lR,2S)-l-[2-((S)-8-carboxy-8-cyclopentyloxycarbonylamino-octylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclo-propylcarbamoyl} -pyrrolidin-3-yl ester (TFA- salt), 262 mg (2.0 mmol) DIPEA and 386 mg (1.1 mmol) HATU in 50 mL DCM and 1 mL
DMF.
HPLC (method A) tR = 5.97 min
TLC, Rf (CH2Cl2/MeOH 9:1) = 0.75
MS (method D): 823 [M+]
Preparation of ^Fluoro-l^-dihydro-isoindole^-carboxylic acid (3R,5S)-5-{(lR,2S)-l- [2-((S)-8-carboxy-8-cyclopentyloxycarbonylamino-octylamino)- benzenesulfonylaminocarbonyl]-2-vinyI-cyclopropyIcarbamoyI}-pyrrolidin-3-yl ester
Step 1
4-Fluoro-l,3-dihydro-isoindole-2-carboxyIic acid (3R,5S)-l-tert-butoxycarbonyl-5-
{(lR,2S)-l-[2-((S)-8-cyclopentyloxycarbonylamino-8-methoxycarbonyl-octylamino)- benzenesulfonylamino-carbonyl]-2-vinyl-cyclopropylcarbamoyl}-pyrroIidin-3-yl ester
Figure imgf000235_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 1) using 368 mg (0.46 mmol) (S)-9-{2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylamino}-2-cyclopentyloxycarbonylamino-nonanoic acid methyl ester (TFA-salt), 216 mg (0.55 mmol) (2S,4R)-4-(4-Fluoro-l,3-dihydro- isoindole-2-carbonyloxy)-pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester, 260 mg (0.68 mmol) HATU and 354 mg (2.74 mmol) DEPEA in 8 mL DCM. HPLC (method A) tR = 6.15 min TLC, Rf (CH2Cl2/Me0H 19:1) = 0.50 MS (method D): 955 [M+]
Step 2
4-FIuoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5-
{(lR,2S)-l-[2-((S)-8-carboxy-8-cyclopentyIoxycarbonylainino-octylainino)- benzenesulfonyIaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-pyrroIidin-3-yI ester
Figure imgf000236_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 217 mg (0.20 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5 S)- 1 -tert-butoxycarbonyl-5 - {( 1 R,2S)- 1 -[2-((S)-8-cyclopentyloxycarbonylamino-8- methoxycarbonyl-octylamino)-benzenesulfonylamino-carbonyl]-2-vinyl- cyclopropylcarbamoyl}-pyrrolidin-3-yl ester (TFA-salt) and 49 mg (2.0 mmol) LiOH in 16 mL THF/MeOH/H2O (2:1:1). HPLC (method A) tR = 5.59 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.60 MS (method D): 941 [M+]
Step 3
4-Fluoro-l,3-dihydro-isoiπdole-2-carboxylic acid (3R,5S)-5-{(lR,2S)-l-[2-((S)-8- carboxy-8-cyclopentyloxycarbonylamino-octyIainino)-benzenesulfonylaminocarbonyl]-
2-vinyl-cyclopropyIcarbamoyl}-pyrrolidin-3-yI ester
Figure imgf000237_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 3) using 191 mg (0.20 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-1 -tert-butoxycarbonyl-5- {( 1 R,2S)- 1 -[2-((S)-8-carboxy-8- cyclopentyloxycarbonylamino-octylamino)-benzene-sulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoyl}-pyrrolidin-3-yl ester and 1 mL TFA in 25 mL DCM. HPLC (method A) tR = 5.01 min MS (method D): 841 [M+]
Example 23
{(8S,10R,14S)-10-[2-(2-Isopropylamino-thiazol-4-yl)-7-methoxy-quinolin-4-yloxyJ-5- [(lR,2S)-l-carbonylamino-2-vinyl-cyclopropyl)-2,2,4,7,13,21-hexaoxo-2λ*6*-thia- 3,6,12,22-tetraaza-tricyclo[21.4.0.0*8,12*]heptacosa-l(23),24,26-trien-14-yl}-carbamic acid cyclopentyl ester
Figure imgf000238_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 118 mg (0.097 mmol) (S)-2-Cyclopentyloxycarbonylamino-8-(2-{[(lR,2S)-l-
({(2S,4R)-4-[2-(2-isopropyl-amino-thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-2- carbonyl}-amino)-2-vinyl-cyclo-propane-carbonyl]-sulfamoyl}-phenylcarbamoyl)-octanoic acid (TFA-salt), 126 mg (0.97 mmol) DIPEA and 184 mg (0.49 mmol) HATU in 100 mL
DCM and 2 mL DMF.
HPLC (method A) tR = 5.43 min
TLC, Rf (CH2Cl2/Me0H 9:1) = 0.45
MS (method D): 971 [M+]
Preparation of (S)-2-Cyclopentyloxycarbonylamino-8-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2- isopropylamino-thiazol-4-yl)-7-methoxy-quinolin-4-yIoxy]-pyrrolidine-2-carbonyl}- amino)-2-vinyl-cyclo-propanecarbonyl]-sulfamoyl}-phenylcarbamoy!)-octanoic acid
Step l
(S)-2-CyclopentyloxycarbonyIamino-nonanedioic acid 1 -methyl ester
Figure imgf000238_0002
To a solution of 1.88 g (6.0 mmol) ((S)-2-Cyclopentyloxycarbonylamino-9-oxo-nonanoic acid methyl ester in 20 mL tBuOH is added at rt 2.1 g (30 mmol) 2-Methyl-2-buten, 2.81 g (18 mmol) NaH2PO4 (in 15 mL H2O) and 1.62 g (18 mmol) NaClO2 (in 15 mL H2O). After stirring for 1 h, the solvent is removed in vacuo, the residue is diluted with water, acidified with 0.5 N HCl and extracted with EtOAc. The combined organic phase is dried with Na2SO4, filtered and the solvent is removed in vacuo. The residue is purified by FC on silica (eluent: CH2Cl2/Me0H 98:2 -> 95:5) to give the title compound as a colorless oil. HPLC (method A) tR = 3.83 min TLC, Rf (CH2Cl2MeOH 19:1) = 0.26 MS (method D): 330 [M+H]
Step 2
(S)-8-{2-[((lR,2S)-l-tert-ButoxycarbonyIaniino-2-vinyl-cycIopropanecarbonyl)- sulfamoyl]-phenylcarbamoyl}-2-cycIopentyloxycarbonylamino-octanoic acid methyl ester
Figure imgf000239_0001
The title compound is prepared analogously as described for the title compound in Example 1
(Step 2) using 0.85 g (2.23 mmol) [(lR,2S)-l-(2-Amino-benzenesulfonylaminocarbonyl)-2- vinyl-cyclopropylj-carbamic acid tert-butyl ester, 0.96 g (2.9 mmol) (S)-2-
Cyclopentyloxycarbonylamino-nonanedioic acid 1 -methyl ester, 0.40 g (3.3 mmol) Benzotriazole, 0.40 g (3.3 mmol) Thionylchloride, 0.92 g (10 mmol) NEt3 and 100 mg
DMAP in 50 mL DCM.
HPLC (method A) tR = 5.31 min
TLC, Rf (CH2Cl2/Me0H 19:1) = 0.31
MS (method D): 693 [M+]
Step 3
(S)-8-{2-(((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]- phenylcarbamoyl}-2-cyclopentyloxycarbonylamino-octanoic acid methyl ester
Figure imgf000240_0001
The title compound is prepared analogously as described for the title compound in Example 1 (Step 3) using 0.85 g (2.23 mmol) (S)-8-{2-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl- cyclopropane-carbonyl)-sulfamoyl]-phenylcarbamoyl}-2-cyclopentyloxycarbonylamino- octanoic acid methyl ester and 5 mL 4N HCl in Dioxane. HPLC (method A) tR = 3.76 min MS (method D): 593 [M+]
Step 4
(2S,4R)-2-{(lR,2S)-l-[2-((S)-8-CyclopentyloxycarbonyIamino-8-methoxycarbonyl- octanoyl-amino)-benzenesuIfonylaminocarbonyIJ-2-vinyl-cyclopropylcarbamoyI}-4-(-[2- (2-isopropyl-amino-thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-l-carboxyIic acid tert-butyl ester
Figure imgf000240_0002
The title compound is prepared analogously as described for the title compound in Example 2 (step 1) using 190 mg (0.27 mmol) (S)-8-{2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylcarbamoyl}-2-cyclopentyloxycarbonylamino- octanoic acid methyl ester (HCl-salt), 141 mg (0.27 mmol) (2S,4R)-4-[2-(2-Isopropylamino- thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-l ,2-dicarboxylic acid 1 -tert-butyl ester, 152 mg (0.40 mmol) HATU and 103 mg (0.80 mmol) DIPEA in 10 mL DCM. HPLC (method A) tR = 5.63 min TLC, Rf (CH2Cl2/Me0H 9: 1) = 0.20 MS (method D): 1103 [M+]
Step 5
(2S,4R)-2-{(lR,2S)-l-[2-((S)-8-Carboxy-8-cyclopentyloxycarbonylamino- octanoylamino)-benzenesulfonylaminocarbonyI]-2-vinyl-cyclopropylcarbamoyI}-4-(-[2-
(2-isopropyl-amino-thiazol-4-yl)-7-methoxy-quinoIin-4-yloxy]-pyrroIidine-l-carboxylic acid tert-butyl ester
Figure imgf000241_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 120 mg (0.099 mmol) (2S,4R)-2-{(lR,2S)-l-[2-((S)-8- Cyclopentyloxycarbonylamino-8-methoxycarbonyl-octanoyl-amino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-4-(-[2-(2-isopropyl-amino- thiazol-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-l-carboxylic acid tert-butyl ester (TFA-salt) and 42 mg (0.99 mmol) LiOH in 16 mL THF/MeOH/H2O (2:1:1). HPLC (method A) tR = 5.38 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.11 MS (method D): 1089 [M+] Step 6
(S)-2-Cyclopentyloxycarbonylamino-8-(2-{[(lR,2S)-l-({(2S,4R)-4-[2-(2-isopropylamino- thiazoI-4-yl)-7-methoxy-quinolin-4-yloxy]-pyrrolidine-2-carbonyI}-amino)-2-vinyI-cyclo- propanecarbonyl]-sulfamoyl}-phenylcarbamoyl)-octanoic acid
Figure imgf000242_0001
The title compound is prepared analogously as described for the title compound in Example 2
(step 3) using 106 mg (0.097 mmol) (2S,4R)-2-{(lR,2S)-l-[2-((S)-8-Carboxy-8- cyclopentyloxycarbonylamino-octanoylamino)-benzenesulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoyl}-4-(-[2-(2-isopropyl-amino-thiazol-4-yl)-7-methoxy-quinolin-4- yloxy]-pyrrolidine-l-carboxylic acid tert-butyl ester and 1 mL TFA in 5 mL DCM.
HPLC (method A) tR = 4.78 min
TLC, Rf (CH2Cl2/MeOH/H2O/AcOH 90: 10:1:0.5) = 0.21
MS (method D): 989 [M+]
Example 24
[(8S,10R,14S)-10-(5-Chloro-pyridin-2-yIoxy)-5-[(lR,2S)-l-carbonylamino-2-vinyl- cyclopropyl]-2,2,4,7,13-pentaoxo-2λ*6*-thia-3,6,12,22-tetraaza- tricyclo [21.4.0.0*8,12 *] heptacosa-1 (23),24,26-trien-l 4-y I]-carbamic acid cyclopentyl ester
Figure imgf000243_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 250 mg (0.25 mmol) (S)-9-{2-[((lR,2S)-l-{[(2S,4R)-4-(5-Chloro-pyridin-2-yloxy)- pyrτolidine-2-carbonyl]-amino}-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]-phenylamino}-2- cyclopentyloxycarbonyl-amino-nonanoic acid (TFA-salt), 318 mg (2.5 mmol) DIPEA and 468 mg (1.2 mmol) HATU in 50 mL DCM and 1 mL DMF. HPLC (method A) tR = 6.27 min TLC, Rf (CH2Cl2/MeOH 19:1) = 0.37 MS (method D): 771 [M+]
Preparation of (S)-9-{2-[((lR,2S)-l-{[(2S,4R)-4-(5-Chloro-pyridin-2-yloxy)-pyrrolidine- 2-carbonyl]-amino}-2-vinyI-cyclopropanecarbonyI)-sulfamoyl]-phenylamino}-2- cyclopentyloxy-carbonyl-amino-nonanoic acid
Step 1
(2S,4R)-4-(5-ChIoro-pyridin-2-yloxy)-2-{(lR,2S)-l-[2-((S)-8- cycIopentyloxycarbonylamino-8-methoxycarbonyI-octyIainino)- benzenesulfonylaininocarbonyl]-2-vinyl-cyclopropylcarbainoyl}-pyrrolidine-l- carboxylic acid tert-butyl ester
Figure imgf000244_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 1) using 380 mg (0.47 mmol) (S)-9-{2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylamino}-2-cyclopentyloxycarbonylamino-nonanoic acid methyl ester (TFA-salt), 194 mg (0.57 mmol) ((2S,4R)-4-(5-Chloro-pyridin-2-yloxy)- pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester (prepared according to WO 2005035525), 269 mg (0.70 mmol) HATU and 365 mg (2.82 mmol) DIPEA in 10 mL DCM. HPLC (method A) tR = 6.33 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.69 MS (method D): 903 [M+]
Step 2
(2S,4R)-2-{(lR,2S)-l-[2-((S)-8-Carboxy-8-cyclopentyloxycarbonyIainino-octylamino)- benzene-sulfonylaminocarbonyl]-2-vinyI-cyclopropyIcarbamoyl}-4-(5-chloro-pyridin-2- yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester
Figure imgf000245_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 250 mg (0.25 mmol) (2S,4R)-4-(5-Chloro-pyridin-2-yloxy)-2-{(lR,2S)-l-[2- ((S)-8-cyclopentyloxy-carbonylamino-8-methoxycarbonyl-octylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropyl-carbamoyl} -pyrrolidine- 1 -carboxylic acid tert-butyl ester (TFA-salt) and 59 mg (2.5 mmol) LiOH in 16 mL THF/MeOH/H2O (2:1:1).
HPLC (method A) tR = 5.86 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.48 MS (method D): 889 [M+]
Step 3
(S)-9-{2-[((lR,2S)-l-{[(2S,4R)-4-(5-Chloro-pyridiii-2-yloxy)-pyrrolidine-2-carbonyl]- amino}-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]-phenylamino}-2- cyclopenryloxycarbonylamino-nonanoic acid
Figure imgf000246_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 3) using 219 mg (0.25 mmol) (2S,4R)-2-{(lR,2S)-l-[2-((S)-8-Carboxy-8- cyclopentyloxycarbonylamino-octylamino)-benzene-sulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoyl}-4-(5-chloro-pyridin-2-yloxy)-pyrrolidine-l-carboxylic acid tert-butyl ester and 1 mL TFA in 10 mL DCM. :
HPLC (method A) tR = 4.99 min TLC, Rf (CH2Cl2/MeOH 9:1) = 0.51 MS (method D): 789 [M+]
Example 25
((8S,14S)-5-[(lR,2S)-l-carbonylamino-2-vinyl-cyclopropyl]-2,2,4,7,13-pentaoxo-2λ*6*- thia-3,6,12,22-tetraaza-tricyclo[21.4.0.0*8,12*]heptacosa-l(23),24,26-trien-14-yl)- carbamic acid cyclopentyl ester
Figure imgf000246_0002
The title compound is prepared analogously as described for the title compound in Example 2 using 62 mg (0.07 mmol) ((S)-2-Cyclopentyloxycarbonylamino-9-[2-({(lR,2S)-l-[((S)- pyrrolidine-2-carbonyl)-amino]-2-vinyl-cyclopropanecarbonyl}-sulfamoyl)-phenylamino]- nonanoic acid (TFA-salt), 90 mg (0.7 mmol) DIPEA and 133 mg (0.35 mmol) HATU in 25 mL DCM and 0.5 mL DMF. HPLC (method A) tR = 5.68 min
TLC, Rf (CH2Cl2/Me0H 19: 1) = 0.41 MS (method D): 644 [M+]
Preparation of ((S)-2-Cyclopentyloxycarbonylamino-9-[2-({(lR,2S)-l-[((S)-pyrrolidine- 2-carbonyl)-amino]-2-vinyl-cycIopropanecarbonyl}-sulfamoyl)-phenylamino]-nonanoic acid
Step l
(S)-2-{(lR,2S)-l-[2-((S)-8-Cyclopentyloxycarbonylamino-8-methoxycarbonyl- octylamino)-benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropyIcarbamoyl}- pyrrolidine-1-carboxylic acid tert-butyl ester
Figure imgf000247_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 1) using 150 mg (0.19 mmol) (S)-9-{2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylamino}-2-cyclopentyloxycarbonylamino-nonanoic acid methyl ester (TFA-salt), 48 mg (0.22 mmol) (S)-Pyrrolidine-l,2-dicarboxylic acid 1 -tert- butyl ester, 106 mg (0.28 mmol) HATU and 144 mg (1.1 mmol) DIPEA in 10 mL DCM. HPLC (method A) tR = 5.84 min TLC, Rf (CH2Cl2MeOH 19: 1) = 0.63 MS (method D): 776 [M+]
Step 2 (S)-2-{(lR,2S)-l-(2-((S)-8-Carboxy-8-cyc,opentyloxycarbonylamino-octylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyI}-pyrrolidine-l- carboxylic acid tert-butyl ester
Figure imgf000248_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 79 mg (0.09 mmol) (S)-2-{(lR,2S)-l-[2-((S)-8-Cyclopentyloxycarbonylamino- 8-methoxycarbonyl-octylamino)-benzenesulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoyl}-pyrrolidine-l-carboxylic acid tert-butyl ester (TFA-salt) and 21 mg (0.89 mmol) LiOH in 16 mL THF/MeOH/H2O (2:1:1). HPLC (method A) tR = 5.35 min TLC, Rf (CH2Cl2/MeOH 9:1) = 0.37 MS (method D): 762 [M+]
Step 3
(S)-2-CycIopentyloxycarbonylamino-9-[2-({(lR,2S)-l-[((S)-pyrrolidine-2-carbonyl)- amino]-2-vinyl-cyclopropanecarbonyl}-sulfamoyl)-phenylamino]-nonanoic acid
Figure imgf000248_0002
The title compound is prepared analogously as described for the title compound in Example 2 (step 3) using 59 mg (0.08 mmol) (S)-2-{(lR,2S)-l-[2-((S)-8-Carboxy-8- cyclopentyloxycarbonylamino-octylamino)-benzenesulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoyl} -pyrrolidine- 1-carboxylic acid tert-butyl ester and 1 mL TFA in 10 mL DCM.
HPLC (method A) tR = 4.44 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.40 MS (method D): 662 [M+]
Example 26 cyclopentyl HlR^S^yS^-iy^y-dioxido-U^lS^lV-trioxo-l-yiByl-
5t,6l,7l,8l,9',10l,lll,12f,131,14l,16al,17l,17aM7bl,18t,19l,21l,22l-octadecahydro-16tfl- spiro[cyclopropane-l,20'-cyclopropa[3,4]pyrrolo[2,l- g][l,2,5,8,18]benzothiatetraazacycloicosin]-13'-yl]carbamate
Figure imgf000249_0001
The title compound is prepared analogously as described for the title compound in Example 2 using 176 mg (0.20 mmol) (S)-9-[2-({(lR,2S)-l-[(3-Aza-bicyclo[3.1.0]hexane-2-carbonyl)- amino]-2-vinyl-cyclopropane-carbonyl}-sulfamoyl)-phenylamino]-2- cyclopentyloxycarbonylamino-nonanoic acid (TFA-salt), 252 mg (0.98 mmol) DLPEA and 371 mg (1.95 mmol) HATU in 50 mL DCM and 1 mL DMF. HPLC (method A) tR = 5.68 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.62 MS (method D): 656 [M+]
Preparation of (S)-9-[2-({(lR,2S)-l-[(3-Aza-bicyclo[3.1.0]hexane-2-carbonyl)-amino]-2- vinyl-cyclopropane-carbonyl}-sulfamoyl)-phenylamino]-2- cyclopentyloxycarbonylamino-nonanoic acid
Step 1 trans-rac-3-Aza-bicyclo[3.1.0]hexane-2,3-dicarboxylic acid 3-tert-butyI ester
Figure imgf000250_0001
To a solution of 0.70 g (5.5. mmol) trans-rac-3-Aza-bicyclo[3.1.0]hexane-2-carboxylic acid (Aldrich) in 20 mL DCM is added 1.11 g (11.0 mmol) NEt3. 1.68 g (7.7 mmol) (BOC2)O is added in three portions over 10 min and the mixture is stirred overnight at ambient temperature. The reaction is quenched by addition of water, acidified with IN HCl and extracted with DCM. The combined organic phase is washed with brine, dried with Na2SO4, filtered and the solvent is removed in vacuo. The residue is purified by FC on silica (eluent: CH2Cl2/Me0H 98:2) to give the title compound as a colorless solid. TLC, Rf (CH2Cl2/Me0H 9:1) = 0.62 MS (method D): 172 [M-55]
Step 2
2-{(lR,2S)-l-[2-((S)-8-CyclopentyloxycarbonyIamino-8-methoxycarbonyl-octylamino)- benzene-sulfonylaminocarbonyl]-2-vinyl-cycIopropyIcarbamoyl}-3-aza- bicyclo[3.1.0]hexane-3-carboxylic acid tert-butyl ester
Figure imgf000250_0002
The title compound is prepared analogously as described for the title compound in Example 2 (step 1) using 244 mg (0.30 mmol) (S)-9-{2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylamino}-2-cyclopentyloxycarbonylamino-nonanoic acid methyl ester (TFA-salt), 82 mg (0.36 mmol) trans-rac-3-Aza-bicyclo[3.1.0]hexane-2,3- dicarboxylic acid 3-tert-butyl ester, 172 mg (0.45 mmol) HATU and 234 mg (1.8 mmol) DEPEA in 1O mL DCM. HPLC (method A) tR = 5.90 min TLC, Rf(CH2Cl2ZMeOH 9:1) = 0.69 MS (method D): 788 [M+]
Step 3
2-{(lR,2S)-l-[2-((S)-8-Carboxy-8-cyclopentyloxycarbonylamino-octylamino)- benzenesulfonyl-aminocarbonyl]-2-vinyl-cycIopropylcarbamoyl}-3-aza- bicyclo[3.1.0]hexane-3-carboxylic acid tert-butyl ester
Figure imgf000251_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 183 mg (0.20 mmol) 2-{(lR,2S)-l-[2-«S)-8-Gyclopentyloxycarbonylaπύno-8- methoxycarbonyl-octylamino)-benzene-sulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoyl}-3-aza-bicyclo[3.1.0]hexane-3-carboxylic acid tert-butyl ester (TFA- salt) and 49 mg (2.0 mmol) LiOH in 20 mL THF/MeOH/H2O (2:1:1). HPLC (method A) tR = 5.42 min TLC, Rf(CH2Cl2MeOH 9:1) = 0.50 MS (method D): 774 [M+]
Step 4
(S)-9-[2-({(lR,2S)-l-[(3-Aza-bicyclo[3.1.0]hexane-2-carbonyl)-amino]-2-vinyl- cycIopropane-carbonyl}-sulfamoyl)-phenylamino]-2-cyclopentyloxycarbonyIamino- nonanoic acid
Figure imgf000252_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 3) using 157 mg (0.20 mmol) 2-{(lR,2S)-l-[2-((S)-8-Carboxy-8- cyclopentyloxycarbonylamino-octylamino)-benzenesulfonyl-aminocarbonyl]-2-vinyl- cyclopropylcarbamoyl}-3-aza-bicyclo[3.1.0]hexane-3-carboxylic acid tert-butyl ester and 0.5 mL TFA in 5 mL DCM. HPLC (method A) tR = 4.43 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.37 MS (method D): 674 [M+]
Example 27
(lR^S^l'R^Sa'^-ό'^'-Dioxido-l'^'.lP'-trioxo-Z-vinylicosahydrodispiroIcyclopropane- l,3'-pyrrolo[2,l-g][l,2,5,8]thiatriazacyclohenicosine-7',rt-cyclopropan]-22'-yl 4-fluoro- l,3-dihydro-2//-isoiiidole-2-carboxylate
Figure imgf000252_0002
The title compound is prepared in analogy to the procedure described in Example 1 (last step) using 115 mg (0.14 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-5- {( 1 R,2S)- 1 - [ 1 -( 1 1 -carboxy-undecyl)-cyclopropanesulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoyl}-pyrrolidin-3-yl ester (trifluoroacetate) LC MS (method E) tR = 5.135 min, M+H = 687.3 HPLC (method C) tR = 5.681 min
Preparation of 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-5-{(lR,2S)-l- [l-(ll-carboxy-undecyl)-cyclopropanesulfonylaminocarbonyl]-2-vinyI- cyclopropylcarbamoylJ-pyrroIidin-S-yl ester (trifluoroacetate)
Step l (12-Bromo-dodecyloxy)-tert-butyl-dimethyI-silane
Figure imgf000253_0001
To a mixture of 3.8 g (14.3 mmol) 12-Bromo-l-dodecanol and 1.2 g (17.2 mmol) imidazole in 8 mL DMF is added 2.6 g (17.2 mmol) tert-Butyl-chloro-dimethyl-silane. The mixture is stirred at RT for 5 h, then EtOAc is added and the mixture is washed with IN aq. HCl and water. The combined organic phases are dried over Na2SO4 and concentrated in vacuo to give the product which was used in the next step without further purification. TLC, Rf (EtOAc/hexane 1 :9) = 0.70
Step 2 l-IlZ-^ert-Butyl-dimethyl-silanyloxy^dodecylJ-cyclopropanesulfonylamine tert-butyl carbamate
Figure imgf000253_0002
To an ice-cold solution of 4.0 mL (28.2 mmol) diisopropylamine in 45 mL THF is added 17 mL (27 mmol) n-BuLi (1.6 M in hexanes). The mixture is stirred for 1 h at 0°C and cooled to -78°C. A mixture of 2.4 g (10.8 mmol) Cyclopropylsulfonylamine tert-butyl carbamate (prepared as described in US2007/0010455) in 5 mL THF is added and the resulting mixture is stirred for an additional hour. Then 4.5 g (11.9 mmol) (12-Bromo-dodecyloxy)-tert-butyl- dimethyl-silane is added and the mixture is allowed to warm to RT and stirred overnight. Sat. aq. NtLjCl-solution is added and the mixture is extracted with EtOAc. The combined organic layers are dried over Na2SO4 and concentrated in vacuo. The residue is purified by FC on silica (eluent: hexane to EtOAc/hexane 1: 1) to give the title compound. TLC, Rf (EtOAc/hexane 1 :9) = 0.8
Step 3 l^lI-Hydroxy-dodecyO-cyclopropanesulfonylamine tert-butyl carbamate
Figure imgf000254_0001
A mixture of 3.3 g (6.4 mmol) l-[12-(tert-Butyl-dimethyl-silanyloxy)-dodecyl]- cyclopropanesulfonyl-amine tert-butyl carbamate and 13 mL TBAF (1 M in THF) in 400 mL
THF is stirred for 4 h at RT. Sat. aq. NELtCl-solution is added and the mixture is extracted with EtOAc. The combined organic layers are dried over Na2SO4 and concentrated in vacuo.
The residue is purified by FC on silica (eluent: hexane to EtOAc/hexane 1:1) to give the title compound.
TLC, Rf (EtOAc/hexane 1:1) = 0.45
Step 4 l-(12-Oxo-dodecyl)-cyclopropanesuIfonylamine tert-butyl carbamate
Figure imgf000254_0002
The title compound is prepared in analogy to the procedure described in Example 14 (step 3) using 1.8 g (4.4 mmol) l-^-Hydroxy-dodecyO-cyclopropanesulfonylamine tert-butyl carbamate, 1.4 g (6.7 mmol) PCC in 150 mL DCM. TLC, Rf (EtOAc/hexane 1:19) = 0.7 Step 5
12-(l-tert-Butyl carbamoylsulfamoyl-cyclopropyl)-dodecanoic acid
Figure imgf000255_0001
The title compound is prepared in analogy to the procedure described in Example 16 (step 1) using 1.5 g (3.7 mmol) l-(12-Oxo-dodecyl)- cyclopropanesulfonylamine tert-butyl carbamate.
TLC, Rf (EtOAc/hexane 1:19) = 0.42
Step 6
12-(l-Sulfamoyl-cyclopropyl)-dodecanoic acid methyl ester
Figure imgf000255_0002
A mixture of 1.5 g (3.6 mmol) 12-(l-tert-Butyl carbamoylsulfamoyl-cyclopropyl)-dodecanoic acid in 10 mL MeOH is cooled to -15 °C and 1.7 mL (23.6 mmol) thionylchloride is added. The mixture is stirred for 1 h at RT and heated to 60 0C overnight. At RT 1 mL of thionylchloride is added and the mixture is again warmed to 60 0C for 2 h before it is concentrated and filtered over a small plug of silica gel to give the title compound. TLC, Rf (EtOAc/hexane 1:1) = 0.57
Step 7 12-{l-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl-cyclopropanecarbonyl)-su.famoyl]- cyclopropylj-dodecanoic acid methyl ester
Figure imgf000256_0001
A mixture of 610 mg (2.7 mmol) (lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl- cyclopropane-carboxylic acid and 687 mg (4.0 mmol) CDI in 20 mL THF is refluxed for 1 h. The reaction mixture is cooled to RT and 0.6 mL (4.0 mmol) DBU and a mixture of 806 mg (2.4 mmol) 12-(l-Sulfamoyl-cyclopropyl)-dodecanoic acid methyl ester in 5 mL THF is added. The mixture is stirred at RT overnight, concentrated in vacuo, taken up in EtOAc and washed with 0.1 M aq. HCl. The combined organic phases are dried over Na2SO4 and concentrated in vacuo. The residue is purified by FC on silica (Eluent: EtOAc/hexane 1:3) to give the title compound.
LC-MS (method E) tR = 5.132 min, M-H = 543.3 HPLC (method C) tR = 4.472 min
Step 8
12-{l-[((lR,2S)-l-Ainino-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]-cycIopropyl}- dodecanoic acid methyl ester (hydrochloride)
Figure imgf000256_0002
A mixture of 343 mg (0.6 mmol) 12-{l-[((lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl- cyclopropane-carbonyl)-sulfamoyl]-cyclopropyl}-dodecanoic acid methyl ester and 10 mL of a 4 M solution of HCl in dioxane in 10 mL dioxane is stirred at RT overnight. The mixture is concentrated and coevaporated twice with DCM. The obtained product is used without further purification. LC MS (method E) tR = 4.103 min, M-H = 443.2 HPLC (method C) tR = 3.258 min
Step 9
4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5-
{(lR,2S)-l-[l-(ll-methoxycarbonyI-undecyl)-cycIopropanesulfonyIaminocarbonyl]-2- vinyl-cyclopropylcarbamoylJ-pyrrolidin-3-yI ester
To a mixture of 181 m (0.46 mmol) (2S,4R)-4-(4-Fluoro-l,3-dihydro-isoindole-2- carbonyloxy)-pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester in 3 mL DMF is added 0.2 mL (1.25 mmol) DIPEA and 192 mg (0.50 mmol) HBTU at RT. After 30 min 200 mg (0.42 mmol) 12- { 1 -[((1 R,2S)- 1 - Amino-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]-cyclopropyl} - dodecanoic acid methyl ester (hydrochloride) is added and the mixture is stirred at RT overnight. DCM is added and the mixture is washed with aq. K2CC>3-solution. The aq. layer is extracted twice with DCM and the combined organic layers are washed with aq. 10% KHSO4-solution and brine, dried over Na2SO4 and concentrated under reduced pressure. The residue is purified by FC (silica gel, eluent: EtOAc/hexane 1 :3) to give the title compound. LC MS (method E) tR = 4.317 min, M+H = 819.4 HPLC (method C) tR = 4.681 min
Step 9
4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5- {(lR,2S)-l-[l-(ll-carboxy-undecyl)-cyclopropanesulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoylJ-pyrrolidin-3-yl ester
Figure imgf000258_0001
A mixture of 137 mg (0.17 mmol) 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)- 1 -tert-butoxycarbonyl-5- {(1 R,2S)- 1 -[ 1 -( 11 -methoxycarbonyl-undecyl)- cyclopropanesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-pyrrolidin-3-yl ester and 21 mg (0.50 mmol) Lithiumhydroxid-monohydrate in 2 mL THF/MeOH/water (2:1 :1) is stirred at RT overnight. The mixture is concentrated under reduced pressure, the residue is acidified with IN HCl and extracted with DCM (3x). The combined organic layers are dried over Na2SO4 and concentrated in vacuo to give the title compound which is used without further purification. LC MS (method E) tR = 4.623 min, M+H = 805.3
Step 10
4-Fluoro-l^-dihydro-isoindole-2-carboxylic acid (3R,5S)-5-{(lR,2S)-l- [l-(ll-carboxy- undecyl)-cyclopropanesulfonylaminocarbonyI]-2-vinyl-cyclopropylcarbamoyl}- pyrrolidin-3-yl ester (trifluoroacetate)
Figure imgf000259_0001
A mixture of 115 mg (0.14 mmol) 4-fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-
1 -tert-butoxycarbonyl-5- {( 1 R,2S)- 1 -[ 1 -( 11 -carboxy-undecyl)- cyclopropanesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbanioyl}-pyrrolidin-3-yl ester and 0.2 mL (2.93 mmol) TFA in 2 mL DCM is stirred at RT for 1.5 h before the mixture is concentrated in vacuo. The crude product is used without further purification. LC MS (method E) tR = 3.316 min, M+H = 705 3
Example 28
(lR,2S,22'R,23a?S)-7'-methyl-6',6'-dioxido-r,4',19'-trioxo-2-vinyIicosahydro-7'H- spiro[cydopropane-l,3'-pyrroIo[2,l-^][l,2,5,8,21]thiatetraazacycIohenicosin]-22'-yI 4- fluoro-l,3-dihydro-2//-isoindole-2-carboxyIate
Figure imgf000259_0002
The title compound is prepared in analogy to the procedure described in Example 1 (last step) using 700 mg (0.75 mmol) of the title compound obtained in step 8 (trifluoroacetate) LC MS (method E) tR = 4.613 min, M-H - 674.2 HPLC (method C) tR - 4.275 min
Step 1
12-Methylamino-dodecanoic acid methyl ester
Figure imgf000260_0001
To a mixture of 5 g (21.8 mmol) 2-Methylamino-dodecanoic acid in 25 mL MeOH is added 5.5 mL (620 mmol) thionyl chloride at - 15°C. The reaction mixture is refluxed overnight and concentrated under reduced pressure to yield the title compound which is used without further purification. LC MS (method E) tR = 1.819 min, M+H = 244.3
Step 2
Methyl 12-[{[(terr-butoxycarbonyl)amino]sulfonyl}(methyl)amino]dodecanoate
Figure imgf000260_0002
mmol) N-(tert-Butoxycarbonyl)-N-[4-(dimethylazaniumylidene)- l,4-dihydropyridin-l-ylsulfonyl]azanide (prepared according to J.- Y. Winum et. al, Org. Lett. 2001, 3, 2241.), 97 mg (0.35 mmol) 12-Methylamino-dodecanoic acid methyl ester and 0.07 mL (0.40 mmol) DIPEA in 3 mL DCM is stirred at RT overnight. The reaction mixture is diluted with DCM and washed with 10% KHSCvsolution. The aq. layer is extracted with DCM and the combined organic layers are washed with % KHSO4-solution and brine, dried over Na2SO4 and concentrated under reduced pressure to give the title compound which is used without further purification. LC MS (method E) tR = 4.415 min, M+H = 423.1
Step 3
Methyl 12-[(aminosulfonyl)(methyl)amino]dodecanoate
Figure imgf000261_0001
A mixture of 9 g (21 mmol) of the title compound obtained in step 2 and 25 mL (330 mmol)
TFA in 100 mL DCM is stirred at RT for 1.5 h before the mixture is concentrated in vacuo.
The crude product is triturated with water, filtered, dried and used without further purification.
LC MS (method E) tR = 4.00 min, M+H = 321.1
Step 4
Methyl 12-[{[({(lR,2S)-l-[(tert-butoxycarbonyl)amino]-2- vinylcyclopropyl}carbonyl)amino]-sulfonyl}(methyl)amino]dodecanoate
Figure imgf000261_0002
A mixture of 1.41 g (6.2 mmol) (lR,2S)-l-tert-Butoxycarbonylamino-2-vinyl-cyclopropane- carboxylic acid and 1.52 mg (9.31 mmol) CDI in 30 mL THF is refluxed for 1 h. In a second flask to a mixture of 3.0 g (9.31 mmol) of the title compound obtained in step 3 in 30 mL THF 9.3 mL (9.3 mmol) LiHMDS (1 M in THF) is added at 0°C and the mixture is stirred for 30 min. Both mixtures are combined and stirred at RT overnight. Water is added and the mixture is extracted with DCM (3x). The combined organic layers are dried over Na2SO4 and concentrated in vacuo. The residue is purified by FC (silica gel, eluent: EtOAc/hexane 1 :3) to give the title compound. LC-MS (method E) tR = 4.728 min, M-H = 530.2
Step 5
Methyl 12-{[({[(lR,25)-l-amino-2-vinylcyclopropyl]carbonyI}amino)sulfonyIj-
(methyl)amino}-dodecanoate (hydrochloride)
Figure imgf000262_0001
A mixture of 1.91 g (3.6 mmol) of the title compound obtained in step 4 and 18 mL of a 4 M solution of HCl in dioxane in 18 mL dioxane is stirred at RT for 6 h. The mixture is concentrated and coevaporated twice with DCM. The obtained product is used without further purification. LC MS (method E) tR = 3.642 min, M+H - 432.3
Step 6
(3R,5S)-l-(tert-butoxycarbonyl)-5-{[(lR,25)-l-({[(12-methoxy-12- oxododecyl)(methyI)amino]-sulfonyl}carbarnoyl)-2- vinyIcyclopropyl]carbamoyl}pyrrolidin-3-yl 4-fluoro-l,3-dihydro-2/-r-isoindole-2- carboxylate
Figure imgf000262_0002
To a mixture of 447 mg (1.13 mmol) of the title compound obtained in step 5 in 10 mL DMF is added 0.5 mL (3.09 mmol) DIPEA and 474 mg (1.24 mmol) HBTU at RT. After 30 min 569 mg (1.03 mmol) 12-{l-[((lR,2S)-l-Amino-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]- cyclopropyl}-dodecanoic acid methyl ester (hydrochloride) is added and the mixture is stirred at RT overnight. DCM is added and the mixture is washed with aq. K23-solution. The aq. layer is extracted twice with DCM and the combined organic layers are washed with aq. 10% KHSCvsolution and brine, dried over Na2SO4 and concentrated under reduced pressure. The residue is purified by FC (silica gel, eluent: EtOAc/hexane 1 : 1) to give the title compound. LC MS (method E) tR - 5.007 min, M+H = 806.3
Step 7
12-{[({[(lR,2S)-l-{[(4R)-l-(tert-butoxycarbonyl)-4-{[(4-nuoro-l,3-dihydro-2/T-isoindol- 2-yI)carbonyl]oxy}-L-prolyl]amino}-2-vinylcyclopropyl]carbonyl}amino)sulfonyl]- (methyl)amino}-dodecanoic acid
Figure imgf000263_0001
A mixture of 641 mg (0.79 mmol) of the title compound obtained in step 6 and 100 mg (2.38 mmol) Lithiumhydroxid-monohydrate in 8 mL THF/MeOH/water (2: 1:1) is stirred at RT overnight. The mixture is concentrated under reduced pressure, the residue is acidified with IN HCl and extracted with DCM (3x). The combined organic layers are dried over Na2SO4 and concentrated in vacuo to give the title compound which is used without further purification.
LC MS (method E) tR = 4.574 min, M-H = 792.4
Step 8
12-{[({[(l/?,25)-l-{[(4i?)-4-{[(4-nuoro-l,3-dihydro-2Jff-isoindol-2-yl)carbonyl]oxy}-L- prolyl] aminoj^-vinylcyclopropyl] carbonyl} amino)sulfony 1] (methyl)amino} dodecanoic acid
Figure imgf000264_0001
A mixture of 600 mg (0.76 mmol) of the title compound obtained in step 7 and 0.5 mL (6.5 mmol) TFA in 12 mL DCM is stirred at RT for 1.5 h, before the mixture is concentrated in vacuo. The crude product is used without further purification. LC MS (method E) tR = 3.023 min, M-H = 692.2 Example 29
Cyclopentyl [(lS,2"5,6'S,22'R,24a'S)-2,2-dimethyl-19t,19t-dioxido-5'^l',24'-trioxo-2M- vinyHf,5',6',7',8I,9',10Mlt,12',13',14',20f,21',23',24t,24af- hexadecahydrodispiro[cyclopropane-l,2'-pyrrolo[2,l- ^][l,2,5,8,181benzothiatetraazacycloicosine-22',l"-cyclopropan]-6'-yl]carbamate
Figure imgf000264_0002
The title compound is prepared analogously as described for the title compound in Example 2 using 330 mg (0.35 mmol) ((S)-2-Cyclopentyloxycarbonylamino-9-[2-({(lR,2S)-l-[((3S,6S)- l,l-dimethyl-5-aza-spiro[2.4]heptane-6-carbonyl)-amino]-2-vinyl-cyclopropanecarbonyl}- sulfamoyl)-phenylamino]-nonanoic acid (TFA-salt), 452 mg (3.5 mmol) DIPEA and 665 mg (1.75 mmol) HATU in 75 mL DCM and 1.5 mL DMF. HPLC (method A) tR = 6.21 min TLC, Rf (CH2CVMeOH 19:1) = 0.40 MS (method D): 698 [M+]
Preparation of (3S,6S)-l,l-Dimethyl-5-aza-spiro[2.4]heptane-5,6-dicarboxylic acid 5- tert-butyl ester
Step l (3R,7aS)-6-Hydroxymethyl-3-phenyl-tetrahydro-pyrrolo[l,2-c]oxazol-5-one
Figure imgf000265_0001
To a solution of DIPA (12.4 mL, 88.6 mmol, 1.2 equiv) in THF (400 mL) at -30°C is added n-BuLi (50 mL, 1.60 M in hexane, 81.0 mmol, 1.10 equiv). The solution is stirred at this temperature for 30 min, then a solution of (3R,7aS)-3-Phenyl-tetrahydro-pyrrolo[l,2- c]oxazol-5-one (15.0 g, 73.8 mmol, 1.0 equiv, prepared according to J. Org. Chem. 1986, 51, 3140.) is added and the solution is stirred at -30°C for 30 min.
A stream of CHO (22.0g, 738 mmol, 10 equiv) and N2 gas is bubbled through this solution over 10 mins. The reaction mixture is warmed up to 0°C over 30 mins and quenched by addition of 2.0 N HCl aq. solution until pH 3. EtOAc is added and the phases are separated. The aqueous layer is extracted 3 x with EtOAc, the combined organic layer is washed with brine, dried over Na2SO4 and concentrated. The residue was continued to the next step with no further purification.
Step 2
(3R,7aS)-6-Methylene-3-phenyl-tetrahydro-pyrrolo[l,2-c]oxazol-5-one
Figure imgf000266_0001
The residue from step 1 is dissolved in DCM (200 mL). To this solution at 0°C is added TEA (30.9 mL, 222 mmol, 3.0 equiv), DMAP (902 mg, 7.4 mmol, 0.1 equiv), followed by SLOW addition of MsCl (11.5 mL, 148 mmol, 2.0 equiv), while the reaction temperature is maintained below 5°C. The solution is stirred at rt for 2 h, quenched by addition of sat. aq. NH4Cl and followed by 1/1 mixture of EtOAc/TBME. The phases are separated and the aqueous layer is extracted with EtOAc. The organic layers are combined, washed with brine, dried with Na2SO4 and concentrated.
The residue is dissolved in DCM/toluene (20 mL/20 mL). At 0°C, 15 mL of DBU are added and the internal temperature is kept below 20°C. After stirring for 2 h at RT the mixture is loaded directly to a silical gel column and flushed with hexane/EtOAc (2/1 to 1/1) to give the title compound (7.4 g). The product is used immediately in the next step to avoid polymerization. LC-MS (method E) tR = 0.86 min, M+H = 216.1
Step 3 lS^'R^a'S^.Z-dimethyl-S'-phenyldihydro-l'H-spirotcycIopropane-l^'-pyrroloIl,!- c][l,3]oxazol]-5'-one
Figure imgf000266_0002
To a solution of isopropyl triphenyl phosphine iodide (10.4 g, 24.1 mmol, 1.4 equiv) in THF (70 mL) at -300C is added n-BuLi (1.60 M, 13.9 mL, 22.4 mmol). The solution is stirred at 0°C for 30 min, then cooled to -300C. A solution of (3R,7aS)-6-Methylene-3-phenyl- tetrahydro-pyrrolo[l,2-c]oxazol-5-one (3.7 g, 17.2 mmol, 1.0 equiv) is and the reaction is warmed to rt over Ih and stirred at rt for 3h. The reaction is quenched by addition of sat. aq. NaHCO3 solution. After diltution with EtOAc, the mixture is filtered. The two phases are separated and the aqueous layer is extracted with EtOAc. Organic layers are combined, washed with brine, dried over Na2SO4 and concentrated. The residue is purified by silical gel, hexane/EtOAc 3/1 to 2/1 to give the title compound. TLC, Rf (EtO Ac/heptane 1 :2) = 0.53 (diastereomer 1) and 0.46 (diastereomer 2)
Step 5 ((3S,6S)-5-Benzyl-l,l-dimethyl-5-aza-spiro[2.4]hept-6-yl)-methanoI
Figure imgf000267_0001
To an ice-cold solution of 9.9 g (38 mmol) (lS,3'R,7a'S)-2,2-dimethyl-3'-phenyldihydro-lΗ- spirotcyclopropane-l^'-pyrrolofl^-cJtljSjoxazorj-S'-one in 250 ml_ abs. THF is added 4.52 g (115 mmol) LiAlH4 under Argon. The reaction is refluxed for 3 h and quenched at 0°C by addition of 10 mL sat. aq. Na2SO4. After addition of 300 mL EtOAc and stirring for 30 min the mixture is filtered and the filtrate is concentrated to give the titled compound, which is used without further purification. HPLC (method A) tR = 2.64 min TLC, Rf (CH2Cl2ZMeOH 9:1) = 0.48 MS (method D): 246 [M+H]
Step 6 ((3S,6S)-l,l-Dimethyl-5-aza-spiro[2.4]hept-6-yl)-methanol
Figure imgf000267_0002
A suspension of 9.5 g (38 mmol) ((3S,6S)-5-Benzyl-l,l-dimethyl-5-aza-spiro[2.4]hept-6-yl)- methanol and 10% Pd on charcoal (2 g) in 100 mL EtOAc/AcOH (1:1) is stirred for 2.5 h under H2 atmosphere. The reaction is filtered, washed with DCM and concentrated. After addition of 2N aq. NaOH, the aq. phase is extracted with DCM. The combined organic phases are washed with brine, dried with Na2SO4, filtered and the solvent is removed in vacuo. The residue is purified by FC (silica gel, eluent: DCM/MeOH 9:1 -> 4:1) to give the title compound. TLC, Rf (CH2Cl2/Me0H 4:1) - 0.29 MS (method D): 156 [M+H]
Step 7
(SS.βSJ-β-Hydroxymethyl-l.l-dimethyl-S-aza-spiroIZ^Jheptane-S-carboxylic acid tert- butyl ester
Figure imgf000268_0001
To an ice-cold solution of 1.4 g (9.0 mmol) ((3S,6S)-l,l-Dimethyl-5-aza-spiro[2.4]hept-6- yl)-methanol in 30 mL DCM is added 2.5 mL (18 mmol) NEt3 and 2.8 g (12.6 mmol) (BOC)2O and the mixture is stirred overnight at RT. The reaction is quenched by addition of aq. sat. bicarbonate and extracted with DCM. The combined organic phases are washed with brine, dried with Na2SO4, filtered and the solvent is removed in vacuo. The residue is purified by FC (silica gel, eluent: DCM/MeOH 19:1) to give the title compound. TLC, Rf (CH2Cl2/MeOH 19:1) = 0.58 MS (method D): 200 [M-55]
Step 8
(3S,6S)-l,l-DiπiethyI-5-aza-spiro[2.4]heptane-5,6-dicarboxylic acid 5-tert-butyI ester
Figure imgf000268_0002
To a solution of 1.7 g (6.7 mmol) (3S,6S)-6-Hydroxymethyl-l,l-dimethyl-5-aza- spiro[2.4]heptane-5-carboxylic acid tert-butyl ester in 30 mL DCM is added 235 mg (0.67 mmol) TPAP, 1.18 g (10 mmol) NMO followed by 300 mg molecular sieves 4A. The reaction is stirred for 2 h at RT, filtered through a pad of Celite, washed with DCM and the solvent is removed in vacuo. The residue is dissolved in 30 mL tert-butanol and 2.4 g (33.3 mmol) 2-Methyl-2-buten is added, followed by 3.1 g (20 mmol) NaH2PO4 (in 20 mL water) and 1.81 g (20 mmol) NaClO2 (in 20 mL water). After 2 h at RT, 0.5 N aq. HCl is added and extracted with EtOAc. The solvent is removed in vacuo, the residue is dissolved in DCM and extracted 3 x with aq. NaHCO3. The organic phase is discarded, while the bicarbonate phase is acidified with 4 N HCl to pH 1-2 and then extracted with DCM. The combined organic phase is dried with Na2SO4, filtered and the solvent is removed in vacuo to give the title compound, which is used without further purification. TLC, Rf(CH2Cl2MeOH 19:1) = 0.16 MS (method D): 214 [M-55]
Preparation of ((SH-CycIopentyloxycarbonylamino-P-IZ^^lR.lS^l-I^S^S)-!,!- dimethyl-5-aza-spiro[2.4]heptane-6-carbonyl)-amino]-2-vinyI-cyclopropanecarbonyl}- sulfamoyl)-phenylamino]-nonanoic acid
Step l
(3S,6S)-6-{(lR,2S)-l-I2-((S)-8-CyclopentyloxycarboπyIamino-8-methoxycarbonyl- octyIamino)-benzenesulfonylaminocarbonyl]-2-vinyl-cycIopropylcarbamoyI}-l,l- dimethyl-5-aza-spiro[2.4]heptane-5-carboxylic acid tert-butyl ester
Figure imgf000269_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 1) using 403 mg (0.50 mmol) (S)-9-{2-[((lR,2S)-l-Amino-2-vinyl- cyclopropanecarbonyl)-sulfamoyl]-phenylamino}-2-cyclopentyloxycarbonylamino-nonanoic acid methyl ester (TFA-salt), 162 mg (0.60 mmol) (3S,6S)-l,l-Dimethyl-5-aza- spiro[2.4]heptane-5,6-dicarboxylic acid 5-tert-butyl ester, 285 mg (0.75 mmol) HATU and 388 mg (3.0 mmol) DIPEA in 15 mL DCM. HPLC (method A) tR = 6.30 min TLC, Rf (CH2Cl2/Me0H 9:1) = 0.40 MS (method D): 830 [M+]
Step 2
(3S,6S)-6-{(lR,2S)-l-[2-((S)-8-Carboxy-8-cyclopentyloxycarbonylamino-octylamino)- benzenesuIfonylaminocarbonyl]-2-vinyl-cycIopropylcarbamoyl}-l,l-dimethyI-5-aza- spiro[2.4]heptane-5-carboxylic acid tert-butyl ester
Figure imgf000270_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 2) using 330 mg (0.35 mmol) (3S,6S)-6-{(lR,2S)-l-[2-((S)-8-
Cyclopentyloxycarbonylamino-8-methoxycarbonyl-octylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-l,l-dimethyl-5-aza- sphO[2.4]heptane-5-carboxylic acid tert-butyl ester (TFA-salt) and 84 mg (3.5 mmol) LiOH in 20 mL THF/MeOH/H2O (2:1 :1). HPLC (method A) tR = 5.85 min
TLC, Rf (CH2Cl2/Me0H 9:1) = 0.50
MS (method D): 816 [M+]
Step 3 (S)-2-CycIopentyloxycarbonyIamino-9-[2-({(lR,2S)-l-[((3S,6S)-l,l-dimethyl-5-aza- spiro[2.4]heptane-6-carbonyl)-amino]-2-vinyl-cyclopropanecarbonyl}-suIfainoyl)- phenylamino]-nonanoic acid
Figure imgf000271_0001
The title compound is prepared analogously as described for the title compound in Example 2 (step 3) using 258 mg (0.35 mmol) (3S,6S)-6-{(lR,2S)-l-[2-((S)-8-Carboxy-8- cyclopentyloxycarbonylamino-octylamino)-benzenesulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoyl} -1,1 -dimethyl-5-aza-spiro[2.4]heptane-5-carboxylic acid tert-butyl ester and 1.0 mL TFA in 10 mL DCM. HPLC (method A) tR = 4.87 min TLC, Rf(CH2Cl2MeOH 85:15) = 0.73 MS (method D): 716 [M+]
Scheme 3:
Synthetic scheme for Example 30
Figure imgf000272_0001
Figure imgf000272_0002
Example 30
((E)-(3S,13S)-3-Benzyl-7-cyclobutylmethyl-ll-cyclopentylmethyl-2,5,6,9,12-pentaoxo- 1,4,8,11tetraaza-cyclononadec-16-en-13-yl)-carbamic acid tert-butyl ester
Figure imgf000272_0003
A solution of VI (85 mg, 0.12 mmol,) with Hoveyda-Grubbs 2nd generation catalyst (3 mg, ~3 mol %) in Toluene (10 mL) degassed with N2 is heated to 80 °C for 2.5 hours. After 2.5 hours the reaction is cooled to room temp and the catalyst is scavenged by adding the reaction to thiourea bound resin (4 equiv.). The reaction is stirred for 1 hour after which time the solution is filtered and the solvent removed. The crude product is run though a plug of silica gel with EtOAc and is purified by prep HPLC to yield VII. LC-MS (method E): M+H = 694.9
Preparation of [^^-({^-((SVl-But-S-enylcarbamoyl^-pheiiyl-ethylcarbamoyl)-!- cyclobutylmethyl-2-oxo-ethylcarbamoyl]-methyl}-cyclopentyImethyl-carbamoyI)-pent- 4-enyl]-carbamic acid tert-butyl ester VI Step l [2-((S)-l-But-3-enylcarbamoyl-2-phenyl-ethylcarbamoyl)-l-cyclobutylmethyl-2- hydroxy-ethyl]-carbamic acid tert-butyl ester
Figure imgf000273_0001
To a solution of I (500 mg, 1.57 mmol, 1.0 equiv) in CH2Cl2 (2.0 mL) at 0 °C is added TFA (2.0 mL) and the solution is stirred at room temp for 1 hour. After 1 hour the solvent is removed under reduced pressure to yield a crude oil. A solution of II (640 mg, 2.30 mmol, 1.5 equiv), EDC (0.45 g, 2.30 mmol, 1.5 equiv), DIEA (2.0 mL, 11.5 mmol, 7.5 equiv) in CH2Cl2 (5.0 mL) is added at 0 0C. The solution is brought to room temperature and stirred for 18 hours. The reaction mixture is diluted with EtOAc and washed with 0.5 N HCl. The phases are separated and the aqueous layer is extracted with EtOAc. The organic layers are combined and washed with brine, dried over Na2SO4 and concentrated. The residue is purified by silica gel column chromatography (heptane/EtOAc, 1 :3) to give product III.. LC-MS (method E): M+H = 474.3
Step 2: [(S)-2-((S)-l-But-3-enylcarbamoyl-2-phenyl-ethylcarbamoyl)-l-cyclobutylmethyI-2-oxo- ethyl]-carbamic acid tert-butyl ester
Figure imgf000274_0001
III IV
To a solution of III (150 mgs, 0.32 mmol, 1.0 equiv) a in CH3CN (10.0 mL) at °C is added
DMP (0.39 mgs, 2.5 equiv.) and the solution is stirred at room temp for 1 hour. After 1 hour 3mL 1 N sodium thiosulfate is added to the reaction mixture and the solution extracted with EtOAc. The phases are separated and the aqueous layer is extracted with EtOAc. The organic layers are combined and washed with brine, dried over Na2SO4 and concentrated. The residue is purified by silica gel column chromatography (heptane/EtOAc, 1:1) to give product IV. LC-MS (method E): M+H = 472.3
Step 3:
[(S)-l-({[2-((S)-l-But-3-enylcarbamoyl-2-phenyl-ethylcarbamoyl)-l-cyclobutylmethyl-2- oxo-ethylcarbamoyI]-methyl}-cycIopentylmethyl-carbamoyl)-pent-4-enyl]-carbamic acid tert-butyl ester
Figure imgf000274_0002
V
To a solution of IV (102 mg, 0.22mmol, 1.0 equiv) a in CH2Cl2 (2.0 mL) at 0 °C is added
TFA (2.0 mL) and the solution is stirred at room temp for 1 hour. After 1 hour the solvent is removed under reduced pressure to yield a crude oil to which is added a solution of V (85 mg, 0.22 mmol, 1.0 equiv), PyBrOP (108 mgs, 0.22 mmol, 1.0 equiv), DIEA (0.2 mL, 1.15 mmol, 5 equiv) in CH2Cl2 (5.0 mL) at 0 0C. The solution is brought to room temperature and stirred for 18 hours. The reaction mixture is diluted with EtOAc and washed with 0.5 N HCl. The phases are separated and the aqueous layer is extracted with EtOAc. The organic layers are combined and washed with brine, dried over Na2SO4 and concentrated. The residue is purified by silica gel column chromatography (heptane/EtOAc, 1Z3) to give product VI. LC-MS (method E): M+H = 722.9.
Example 31
Cyclopentyl KlR^S^'I^ό'S^a'SH'-hydroxy-^'.lP'-dioxido-SSll'^'-trioxo-Z-vinyl- lI,2',3',5f,6t,7I,8',9t,10t,ll',12t,13',14',20',21f,23f,24l,24al- octadecahydrospiro[cyclopropane-l,22'-pyrrolo[2,l- g][l,2,5,8,18]benzothiatetraazacycloicosin]-6'-ylJcarbamate
Figure imgf000275_0001
The title compound can be prepared as described above for the final step in the synthesis of example 1
LC MS (method E) tR = 4.209 min, M+H = 660.3 HPLC (method C) tR - 3.993 min
Step l tert-butyl (2S,4R)-4-{[tert-butyI(dimethyl)silyl]oxy}-2-{[(lR,2S)-l-{[(2-{[(8S)-8- { [(cyclopentyloxy)carbonylj amino}-9-methoxy-9- oxononyl]amino}phenyl)sulfonyl]carbamoyl}-2- vinylcyclopropyl]carbamoyl}pyrro-idine-l-carboxylate
Figure imgf000275_0002
The title compound can be prepared as described above for the synthesis of (3R,5S)-l-tert- butoxycarbonyl-5- {( 1 R,2S)- 1 -[2-(8-methoxycarbonyl-octanoylamino)- benzenesulfonylaminocarbonyl]-2-vinyl-cyclopropylcarbamoyl}-pyrrolidin-3-yl ester (example 3) using (4R)-l-(tert-butoxycarbonyl)-4-{[tert-butyl(dimethyl)silyl]oxy}-L-proline (for preparation see T. Sato et al. J. Chem. Soc. Perkin Trans. 1 2001, 20, 2623). LC MS (method E) tR = 5.401 min, M+H = 907.2
Step 2
(2S)-9-({2-[({[(lR,2S)-l-{[(4R)-l-(tert-butoxycarbonyl)-4-{[tert- butyl(dimethyl)silyI]oxy}-L-prolyl]amino}-2- vinylcyclopropyl] carbonyi} amino)sulfonyl] phenyl} amino)-2- {[(cyclopentyloxy)carbonyl]amino}nonanoic acid
Figure imgf000276_0001
The title compound can be prepared analogously as described for the title compound in example 21, step 11.
LC MS (method E) tR = 5.097 min
Step 3
(2S)-2-{[(cyclopentyloxy)carbonyl]amino}-9-({2-[({[(lR,2S)-l-{[(4R)-4-hydroxy-L- prolyl]amino}-2-vinylcyclopropyl]carbonyl}amino)suIfonyl]phenyl}amino)nonanoic acid
Figure imgf000276_0002
The title compound can be prepared as described above for the synthesis of (2S)-2- {[(cyclopentyloxy)carbonyl]amino} -9-( {2-[( {[(1 R,2S)- 1- {[(4R)-4- {[(2-nitrophenyl)- sulfonyl] amino} -L-prolyl]amino} -2-vinylcyclopropyl]carbonyl}amino)-sulfonyl]phenyl} - amino)nonanoic acid (hydrochloride salt).
LC MS (method E) tR = 3.009 min, M+H = 678.3
Example 32
Cyclopentyl {(lR,2S,2'R,6'S,24a'S)-19f,19'-dioxido-5',21I,24'-trioxo-2'-[(quinolin-6- ylcarboayl)-amino]-2-vinyl-lI,2',3',5t,6(,71,8I,9',10',llM2t,13',14',20',21t,23',24',24af- octadecahydrospiro- [cyclop ropane-1 ,22'-pyrrolo [2,1 - g][l,2,5,8,18jbenzothiatetraazacycloicosin]-6'-yl}carbamate
Figure imgf000277_0001
To a mixture of 50 mg (0.07 mmol) Cyclopentyl [(lR,2S,2'R,6'S,24atS)-2'-amino-19',19I- dioxido-5',21 ',24'-trioxo-2-vinyl- 1 *,2',31,5\6I,7',8',9', 10', 11 ', 1 T, 131, 14',20\21 ',23',24',24^- octadecahydrospiro[cyclopropane- 1 ,22'-pyrrolo[2, 1 - g][l,2,5,8,18]benzothiatetraazacycloicosin]-6'-yl]carbamate, 0.037 mL (0.21 mmol) DIPEA and 40 mg (0.11 mmol) HATU in 0.7 mL DCM/DMF (50:1) are added at 0°C 16 mg (0.09 mmol) 6-Quinoline carboxlylic acid. The mixture is stirred for 72 h, concentrated in vacuo and purified by prep. HPLC (method C). LC MS (method E) tR = 4.254 min, M+H - 814.3
Preparation of Cyclopentyl [(lR,2S,2fR,6'S,24a'S)-2'-amino-19',19'-dioxido-5t,21',24'- trioxo-2-vinyl-l',2',3t,5',6t,7',8',9',10I,ir,12l,13',14',20',21I,23',24',24a'- octadecahydrospiro[cyclopropane-l,22'-pyrrolo[2,l- g][l,2,5,8,18]benzothiatetraazacycIoicosin]-6'-yl]carbamate
Step 1 l-tert-Butyl 2-methyl (2S,4R)-4-{[(2-nitrophenyI)sulfonyl]amino}pyrrolidine-l,2- dicarboxylate
Figure imgf000278_0001
To a mixture of 3 g (10.6 mmol) N-Boc-trans-4-amino-L-proline methyl ester hydrochloride, 14.8 mL (106 mmol) triethylamine in 260 mL DCM is added 3.6 g (15.9 mmol) 2-nitro- benzolsulfonylchloride at 00C. The mixture is stirred at rt overnight and extracted with brine. The organic layer is dried over Na2SO4, concentrated in vacuo and purified by FC on silica (eluent: DCM to DCMMeOH 95:5). LC MS (method E) tR = 3.284 min, M+H = 430.03 HPLC (method C) tR = 3.306 min
Step 2
(4R)-l-(te/t-butoxycarbonyI)-4-{[(2-nitrophenyl)sulfonyl]amino}-L-proIine
Figure imgf000278_0002
A mixture of 4.2 g (9.8 mmol) 1-tert-butyl 2-methyl (2S,4R)-4-{[(2- nitrophenyl)sulfonyl]amino} -pyrrolidine- 1,2-dicarboxylate and 1.2 g (29 mmol) LiOH in 100 mL THF/water/MeOH (2:1 :1) is stirred at rt for 4 h. The mixture is concentrated in vacuo and the residue is diluted with DCM and IN aq. HCl solution. The formed precipitate is filtered and dried. LC MS (method E) tR = 2.943 min, M-H = 414.1
Step 3
(2S)-2-{[(cyclopentyloxy)carbonyl]amino}-9-({2-[({[(lR,2S)-l-{[(4R)-4-{[(2-nitropheπyl)- sulfonylJaminoJ-L-prolyllaminoJ-Σ-vinylcycIopropyllcarbonylJamino- )sulfonyl]phenyl}amino)-nonanoic acid
Figure imgf000279_0001
The title compound is prepared analogously as described for the title compound in example 21, step 10 using 3.3 g (5.4 mmol) methyl (2S)-9-({2-[({[(lR,2S)-l-amino-2- vinylcyclopropyl]carbonyl } amino)-sulfonyl]phenyl } amino)-2-
{[(cyclopentyloxy)carbonyl] amino }nonanoate hydrochloride, 2.8 g (6.8 mmol) (4R)-l-(tert- butoxycarbonyl)-4-{[(2-nitrophenyl)sulfonyl] amino }-L-proline, 2.6 g (6.9 mmol) HTBU and 3.0 mL (17 mmol) DIPEA in 50 mL DCM/DMF (50:1) LC MS (method E) tR = 4.644 min, M+H = 977.2 HPLC (method C) tR = 4.346 min
Step 4
(2S)-9-({2-[({[(lR,2S)-l-{[(4R)-l-(tert-Butoxycarbonyl)-4-{[(2- nitrophenyl)sulfonyl]amino}-L-prolyl]amino}-2- vinylcyclopropyl] carbonyl} amino)su Ifony 1] phenyl} amino)-2-{ [(cyclopenty loxy )- carbonyl|amino}nonanoic acid
Figure imgf000280_0001
The title compound is prepared analogously as described for the title compound in example 21, step 11 using 3.0 g (3.1 mmol) ((2S)-2-{[(cyclopentyloxy)carbonyl]amino}-9-({2- [( {[( 1R,2S)- 1 - {[(4R)-4- {[(2-nitrophenyl)-sulfonyl]amino} -L-prolyl]amino} -2- vinylcyclopropyl]carbonyl}amino-)sulfonyl]phenyl}-amino)-nonanoic acid and 387 mg (9.22 mmol) LiOH in 30 mL THF/water/MeOH 2:1 :1. LC MS (method E) tR = 4.240 min, M+H = 963.3 HPLC (method C) tR = 4.083 min
Step 5
(2S)-2-{[(cycIopeπtyIoxy)carbonyl]amino}-9-({2-[({[(lR,2S)-l-{[(4R)-4-{[(2-nitrophenyl)- suIfonyllaminoJ-L-prolyllaminoJ^-viπylcyclopropylJcarbonylJamino^sulfonylJphenyl}- amino)nonanoic acid (hydrochloride salt)
Figure imgf000280_0002
A mixture of 1.9 g (2.0 mmol) (2S)-9-({2-[({[(lR,2S)-l-{[(4R)-l-(tert-Butoxycarbonyl)-4-
{[(2-nitrophenyl)sulfonyl]amino}-L-prolyl]amino}-2- vinylcyclopropyl]carbonyl} amino)sulfonyl]phenyl} amino)-2- {[(cyclopentyloxy)- carbonyl]amino}nonanoic acid, 20 mL 4 M HCl in dioxane and 20 mL dioxane is stirred at rt for 3 h.The mixture is concentrated in vacuo and the crude product is used without further purification.
LC MS (method E) tR = 3.346 min, M+H = 863.2
HPLC (method C) tR = 3.484 min
Step 6
CycIopentyl [(lR,2S,2tR,6'S,24a'S)-2'-{[(2-nitrophenyl)sulfonyl]amino}-19',19f-dioxido-
5',21',24l-trioxo-2-vinyI-ll,2t,3',5f,6f,7',8',9',10',ll',12',13',14t,20',21t,23f,24',24al- octadecahydrospiro-[cyclopropane-l,22'-pyrrolo[2,l- g][l,2,5,8,18]benzothiatetraazacycloicosin]-6'-yl]carbamate
Figure imgf000281_0001
The title compound can be prepared analogously as described for the title compound of example 21 using 2.1 g (2.1 mmol) (2S)-2-{[(cyclopentyloxy)carbonyl]amino}-9-({2-
[({[(lR,2S)-l-{[(4R)-4-{[(2-nitrophenyl)-sulfonyl]amino}-L-prolyl]amino}-2- vinylcyclopropyl]carbonyl}amino)-sulfonyl]phenyl}-amino)nonanoic acid (hydrochloride salt), 4.1 g (10.8 mmol) HATU and 3.8 mL (21.5 mmol) DIPEA in 300 mL DCM/DMF
(50:1).
LC MS (method E) tR = 4.470 min, M-H - 842.2
HPLC (method C) tR - 4.371 min
Step 7 Cyclopentyl [(1R,2S,2'R,6'S,24a'S)-2'-amino-19',19'-dioxido-S'^l'^'-trioxo-Z-vinyl- ll,2',3',5',6',7f,8I,9',10l,lll,12',13',14',20',21',23f,24l,24a'- octadecahydrospiro[cyclopropane-l,22'-pyrrolo[2,l- g] [1,2,5,8,18]benzothiatetraazacycloicosin]-6'-yl]carbamate
Figure imgf000282_0001
A mixture of 670 mg (0.7 mmol) Cyclopentyl [(lR,2S,2'R,6'S,24a1S)-2'-{[(2- nitrophenyl)sulfonyl] amino } - 19', 19'-dioxido-5',21 ',24'-trioxo-2-vinyl- lI,2I,3l,5I,6l,7t,8I,9I,10',l l1,12I,13I,14l,20I,21',231,24t,24a'-octadecahydrospiro-[cyclopropane- l,22l-pyrrolo[2,l-g][l,2,5,8,18]benzothiatetraazacycloicosin]-6'-yl]carbamate, 0.2 mL (2.2 mmol) thiophenol and 404 mg (2.9 mmol) K2CO3 in 30 mL acetonitrile is stirred at rt overnight. The mixture is diluted with water and ethyl acetate. The organic layer is washed with brine, dried over Na2SO4 and concentrated in vacuo. The residue is dissolved in hot DCM, ethyl ether is added and the precipitate is filtered and dried. LC MS (method E) tR = 3.150 min, MlH = 659.3
Example 33 (l^S^ό'S^O'^la'S^lβ'-Ktert-butoxycarbonyOaminol^'-methyl-ό'^'-dioxido- l',4',17'-trioxo-2-vinyloctadecahydro-7'H-spiro[cyclopropane-l,3'-pyrroIo[2,l- g][l,2,5,8,19]thiatetraazacyclononadecin]-20'-yl 5-(dimethylamino)-l,3-dihydro-2H- isoindole-2-carboxylate
Figure imgf000282_0002
A mixture of 150 mg (0.15 mmol) (lR^S.lό'S^O'R^la'S^lό'-amino-T'-methyl-ό'.β1- dioxido- r,4', 17'-trioxo-2- vinyloctadecahydro-7'H-spiro[cyclopropane- 1 ,3 '-pyrrolo[2, 1 - g][ 1 ,2,5,8, 19]thiatetraazacyclononadecin]-20'-yl 5-(dimethylamino)- 1 ,3-dihydro-2H- isoindole-2-carboxylate, 37 mg (0.17 mmol) Boc2O and 0.03 mL (0.20 mmol) triethylamine in 4 mL DCM is stirred at rt overnight. The mixture is concentrated and purified by prep.
HPLC.
LC MS (method E) tR = 3.645 min, M+H = 788.2
Step 1
Ethyl (2S)-2- { [(2-n itrophenyl)sulfonyl] amino}dec-9-enoate
Figure imgf000283_0001
To a mixture of 10 g (47 mmol) ethyl (2<S)-2-aminodec-9-enoate (prepared as described above for (S)-2-Amino-non-8-enoic acid ethyl ester) and 67 mL (469 mmol) triethylamine in 800 mL DCM is added 16 g (70 mmol) o-nitro-benzenesulfonylchloride at 0°C. The mixture is stirred at rt overnight and partitioned between EtOAc and water. The aq. layer is extracted with EtOAc and the combined organic layers are dried over Na2SO4 and concentrated in vacuo. The crude product is purified by FC (silica gel). LC MS (method E) tR = 4.361 min, M+H = 399.1 HPLC (method C) tR = 4.335 min
Step 2
Ethyl (2S)-10-hydroxy-2-{[(2-nitrophenyl)suIfonyl]amino}decanoate
Figure imgf000283_0002
This compound can be prepared as described above for the synthesis of (S)-2- Cyclopentyloxycarbonylamino-9-hydroxy-nonanoic acid methyl ester LC MS (method E) tR = 3.550 min, M+H = 417.1 HPLC (method C) tR = 3.635 min
Step 3
Ethyl (2S)-10-(methylamino)-2-{[(2-nitrophenyl)suIfonyI]amino}decanoate
Figure imgf000284_0001
To a mixture of 13 g (31 mmol) Ethyl (2S)-10-hydroxy-2-{[(2- nitrophenyl)sulfonyl]amino}decanoate in 300 mL DCM is added 2.9 mL (37 mmol) methanesulfonylchloride and 8.6 mL (61 mmol) triethylamine at 0°C. After 1 h water is added and the mixture is extracted with DCM. The combined organic layers are dried over Na2SO4 and concentrated. The crude is taken up in 150 mL DMSO and 42 mL methylamine (8 M in EtOH) and the mixture is stirred at rt overnight. The mixture is partitioned between water and ether and the aq. phase is extracted with ether. The combined organic layers are dried over Na2SO4 and concentrated in vacuo to give the title compound which is used without further purification in the next step. LC MS (method E) tR = 0.930 min, M+H = 430.1
Step 4
Ethyl (2S)-10-[ { | (tert-bu toxyca r bon yl )a mino] su lion vϊ } (methyl)amino]-2-{ [(2- nitrophenyl)sulfonyl]amino}decanoate
Figure imgf000285_0001
This compound can be prepared using the method described by J. Y. Winum et al. Org. Lett.
2001, 3, 2241.
LC MS (method E) tR = 4.121 min, M+H = 609.3
HPLC (method C) tR = 4.580 min
Step 5
Ethyl (2S)-10-[(aminosulfonyl)(methyl)amino]-2-{[(2- nitrophenyl)sulfonyl]amino}decanoate
Figure imgf000285_0002
A mixture of 11 g (16 mmol) ethyl (2S)-10-[{[(tert- butoxycarbonyl)amino]sulfonyl} (methyl)amino]-2- {[(2- nitrophenyl)sulfonyl]amino}decanoate and 200 mL HCl in dioxane (4 M) is stirred overnight at rt. The mixture is concentrated and the crude is purified by FC (silica gel, eluent: hexanes to hexanes/EtOAc 1:1).
LC MS (method E) tR = 3.559 min, M+H = 509.0
HPLC (method C) tR = 3.900 min
Step 6 Ethyl (2S)-10-[{[({(lR,2S)-l-[(tert-butoxycarbonyl)amino]-2- vinylcyclopropyl}carbonyl)amino]suIfonyl}(methyl)amino]-2-{[(2- nitrophenyl)sulfonyl]amino}decanoate
Figure imgf000286_0001
The title compound can be prepared as described above for the synthesis of [(lR,2S)-l-(2-
Amino-benzenesulfonylaminocarbonyl)-2-vinyl-cyclopropyl]-carbamic acid tert-butyl ester
(example 1, step 1)
LC MS (method E) tR = 4.270 min, M-H = 718.2
HPLC (method C) tR = 4.289 min
Step 7 ethyl (2S)-10-{ [({[(1 R,2S)-1 -amino-2- vinylcyclopropyl] car bony 1} amino)su Ifonyl] (methyI)amino}-2-{ [(2- nitrophenyl)sulfonyl]amino}decanoate
Figure imgf000286_0002
The title compound can be prepared as described above for the synthesis of 8-{2-[((lR,2S)-l-
Amino-2-vinyl-cyclopropanecarbonyl)-sulfamoyl]-phenylcarbamoyl} -octanoic acid methyl ester (example 1, step 3)
LC MS (method E) tR = 3.368 min, M+H = 618.1
HPLC (method C) tR = 3.279 min
Step 8 (3R,5S)-l-(tert-butoxycarbonyl)-5-({(lR,2S)-l-[({[(9S)-10-ethoxy-9-{[(2- nitrophenylJsulfonyllaininoJ-lO-oxodecylKmethylJaininoJsulfonyOcarbamoyll-Z- vinylcyclopropyl}carbamoyl)pyrroIidin-3-yl 5-(dimethylamino)-l,3-dihydro-2H- isoindoIe-2-carboxylate
Figure imgf000287_0001
The title compound can be prepared as described above for the synthesis of 4-Fluoro-l,3- dihydro-isoindole-2-carboxylic acid (3R,5 S)-I -tert-butoxycarbonyl-5- {( 1 R,2S)- 1 -[2-(8- methoxycarbonyl-octanoylamino)-benzenesulfonylaminocarbonyl]-2-vinyl- cyclopropylcarbamoylJ-pyrrolidin-3-yl ester (example 3, step 4) using (4R)-l-(tert- butoxycarbonyl)-4-({[5-(dimethylamino)-l,3-dihydro-2H-isoindol-2-yl]carbonyl}oxy)-L- proline which can be prepared as described in example 3, steps 1 and 2. LC MS (method E) tR = 4.439 min, M+H = 1020.4
Step 9 (2S)-10-{[({[(lR,2S)-l-{[(4R)-4-({(5-(dimethylamino)-l,3-dihydro-2H-isoindol-2- yl] carbonyl} oxy)-L-prolyl] amino}-2- vinylcycIopropyI]carbonyl}amino)suIfonyi](methyl)amino}-2-{[(2- nitrophenyl)sulfonyl]amino}decanoic acid
Figure imgf000288_0001
The title compound can be prepared as described above for the synthesis of (2S)-2- {[(cyclopentyloxy)carbonyl]amino}-9-({2-[({[(lR,2S)-l-{[(4R)-4-{[(2-nitrophenyl)- sulfonyl] amino } -L-prolyl] amino } -2- vinylcyclopropyl] carbonyl } amino)-sulfonyl]phenyl } - amino)nonanoic acid (hydrochloride salt) (step 4 and 5) LC MS (method E) tR = 2.934 min, M+H - 892.3
Step 10
(H^ZSjlό'S^O'^la'SH'-methyHό'-IP-nitrophenyOsulfonyllaminol-όSό'-dioxido- l',4',17'-trioxo-2-vinyloctadecahydro-7'H-spiro[cyclopropane-l,3'-pyrrolo[2,l- g][l,2,5,8,19]thiatetraazacyclononadecin]-20'-yl 5-(dimethylamino)-l,3-dihydro-2H- isoindole-2-carboxylate
Figure imgf000288_0002
The title compound can be prepared as described above for the final step in the synthesis of example 1 LC MS (method E) tR = 3.927 min, M+H = 874.2
Step 11
(1 R,2S,16'S,20'R,21 a' S)-16'-amino-7'-methyI-6f ,6'-dioxido-l ',4',17'-trioxo-2- vinyloctadecahydro-7Η-spiro[cycIopropane-l,3'-pyrrolo[2,l- g][l,2,5,8,19]thiatetraazacyclononadecin]-20'-yI 5-(dimethylamino)-l,3-dihydro-2H- isoindole-2-carboxylate
Figure imgf000289_0001
A mixture of 760 mg (0.9 mmol) (lR,2S,16'S,20IR,21a'S)-7I-methyl-16'-{[(2- nitrophenyl)sulfonyl] amino } -6',6'-dioxido- 1 ',4', 17'-trioxo-2-vinyloctadecahydro-7Η- spiro[cyclopropane- 1 ,3'-pytτolo[2, 1 -g][ 1 ,2,5,8, 19]thiatetraazacyclononadecin]-20'-yl 5- (dimethylamino)-l,3-dihydro-2H-isoindole-2-carboxylate, 0.3 mL (4.4 mmol) 2-mercapto- ethanol and 0.7 mL (4.4 mmol) DBU in 2 mL acetonitrile is stirred at rt for 5 h. The mixture is partitioned between EtOAc and water. The organic layer is washed with water, dried over Na2SO4 and concentrated to give the crude product which is used in the next step without further purification. LC MS (method E) tR = 1.806 min, M+H = 688.1
Example 34 (l/?,25,2fR,6t5,24atS)-6f-amino-17l-fluoro-19',19t-dioxido-51,21t,24'-trioxo-2-vinyl- l',2t,3I,5',6',71,8f,9f,10I,ll1,12',13',14',20',21',23',24',24al- octadecahydrospiro[cyclopropane-l,22'-pyrrolo[2,l-
^][l,2,5,8,18]benzothiatetraazacycloicosin]-2f-yl 4-fluoro-l,3-dihydro-2iϊ-isoindole-2- carboxylate
Figure imgf000290_0001
To a solution of 2.24 g (2.45 mmol) (l/?,25,21/?,6'5,24aI5)-17I-fluoro-6'-{[(2-nitrophenyl)- sulfonyl] amino } - 19', 19'-dioxido-5',21 ',24'-trioxo-2- vinyl-
Figure imgf000290_0002
1 ,22'-pyrrolo[2, 1 -g][ 1 ,2,5,8, 18]benzothiatetraazacycloicosin]-2'-yl 4-fluoro- 1 ,3-dihydro-2H- isoindole-2-carboxylate (prepared analogously as described starting from ethyl (2S)-2-{[(2- nitrophenyl)sulfonyl]-amino}dec-9-enoate) in 230 mL acetonitrile is added at it 1.85 mL (12.3 mmol) DBU followed by 1.9 mL (27 mmol) 2-mercaptoethanole. After 90 min the reaction mixture is concentrated, aq. bicarbonate is added and extracted with DCM. The organic layer is dried over Na2SO4, concentrated in vacuo and purified by FC on silica (eluent: DCM/MeOΗ 19:1 -> 9:1). MS (method ): M+ = 729.2 ΗPLC (method ) tR = 4.60 min
The following compounds (Table 1) can be prepared according to one of the methods described above.
TABLEl
\23',2 '
Figure imgf000291_0001
Figure imgf000292_0001
Figure imgf000293_0001
Figure imgf000294_0001
Figure imgf000295_0001
Figure imgf000296_0001
Figure imgf000297_0001
',23',2
Figure imgf000298_0001
',23',2
Figure imgf000299_0001
Figure imgf000300_0001
Figure imgf000301_0001
Figure imgf000302_0001
,24a1-
",23", ',
Figure imgf000303_0001
18]
} - ',5',
Figure imgf000304_0001
Figure imgf000305_0001
Figure imgf000307_0001
Figure imgf000308_0001
Figure imgf000309_0001
Figure imgf000310_0001
Figure imgf000311_0001
Figure imgf000312_0001
Figure imgf000313_0001
Figure imgf000314_0001
Figure imgf000315_0001
10',
',23',2
Figure imgf000316_0001
Figure imgf000317_0001
',23',2
4-
Figure imgf000318_0001
11 f,
Figure imgf000319_0001
Figure imgf000320_0001
-
Figure imgf000321_0001
',
Figure imgf000322_0001
Figure imgf000323_0001
Figure imgf000324_0001
Figure imgf000325_0001
Figure imgf000326_0001
-
Figure imgf000327_0001
Figure imgf000328_0001
Figure imgf000329_0001
',23',2
',23',2
Figure imgf000330_0001
Figure imgf000331_0001
Figure imgf000332_0001
Figure imgf000333_0001
Figure imgf000334_0001
Figure imgf000335_0001
Figure imgf000336_0001
Figure imgf000337_0001
Figure imgf000338_0001
Figure imgf000339_0001
Figure imgf000340_0001
Figure imgf000341_0001
Figure imgf000342_0001
Figure imgf000343_0001
Figure imgf000344_0001
Figure imgf000345_0001
Figure imgf000346_0001
Figure imgf000347_0001
Figure imgf000348_0001
Example 226
4-FIuoro-l,3-dihydro-isoindole-2-carboxylic acid (2R,5S,18aS)-16-cyclobutylmethyI-5- cycIopentyloxycarbonylamino-4,14,15,18-tetraoxo-octadecahydro-3a,13,17-triaza- cyclopentacycloheptadecen-2-yl ester
Figure imgf000349_0001
A solution of 0.01 g (0.014 mmol) of 4-fluoro-l,3-dihydro-isoindole-2-carboxylic acid (2R,5S, 18aS)- 1 ό-cyclobutylmethyl-S-cyclopentyloxycarbonylamino- 15-hydroxy-4, 14, 18- trioxo-octadecahydro-3a,13,17-triaza-cyclopentacycloheptadecen-2-yl ester in 0.1 mL of DMSO is treated with 0.012 g (0.042 mmol) of EBX for 3 hours and chromatographed by RP- HPLC (method G) to give the title compound; MS (method D): 712 [M+l]; HPLC (method A) tR (min) 5.24
Preparation of 4-fluoro-l,3-dihydro-isoindole-2-carboxylic acid (2R,5S,18aS)-16-cyclo butylmethyl-5-cyclopentyloxycarbonylamiiio-15-hydroxy-4,14,18-trioxo-octadecahydro- 3 a, 13, 17-triaza-cycIopen tacycloheptadecen-2-y 1 ester
Figure imgf000350_0001
Figure imgf000351_0001
Step l
(S)-2-CycIopentyIoxycarbonyIamino-non-8-enoic acid methyl ester
A solution of 18.1 g (63.87 mmol) of (S)-2-cyclopentyloxycarbonylamino-non-8-enoic acid in 300 mL of acetone is treated with 10.232 g (102.2 mmol) of ICHCO3 and 22.666 g (159.69 mmol) of iodomethane and then heated up to reflux. Upon completion the reaction mixture is cooled down, salts are fϊltered-off and the filtrate is concentrated, taken up in EtOAc, washed with saturated aqueous NaHCO3 and brine. The organics are dried over Na2SO4, concentrated in vacuo to give the title compound; MS (method D): 298 [M+l]
Step 2
(S)-2-CyclopentyIoxycarbonylamino-9-hydroxy-nonanoic acid methyl ester
A solution of 25.65 g (86.25 mmol) of (S)-2-cyclopentyloxycarbonylamino-non-8-enoic acid methyl ester in 400 mL of absolute THF is cooled to 0 0C and treated by drop wise addition of 275 rnL (120.7 mmol) of 9-BBN (0.5M in THF solution) while maintaining temperature below 5 0C. The reaction mixture is stirred at RT under completion, cooled to 0 0C, treated by drop wise addition of 80 itiL of a 5% NaHCO3 aqueous solution, then by careful addition of 16.3 mL of 35% H2O2 in water while maintaining the temperature below 12 0C. The reaction mixture is stirred at RT for 1.5 hour, treated with 100 mL of saturated aqueous NaHCO3 and 100 mL water. The organics are washed with brine and water, combined, dried (Na2SO4), concentrated and chromatographed on silica gel (eluent Hexane / EtOAc 1:1) to give the title compound; MS (method D): 316 [M+l]
Step 3
(S)-9-Bromo-2-cyclopentyloxycarbonylamino-nonanoic acid methyl ester
A solution of 5.5 g (17.44 mmol) of (S)-2-cyclopentyloxycarbonylamino-9-hydroxy- nonanoic acid methyl ester in 60 mL OfCH2Cl2 is treated with 4.851 g (18.31 mmol) of triphenylphosphine and 3.36 g (18.31 mmol) of N-bromosuccinimide and stirred overnight at RT. The crude reaction mixture is chromatographed on silica gel (eluent Hexane / EtOAc 7:2) to give the title compound; MS (method D): 378 [M+l]
Step 4
(S)-9-Azido-2-cyclopentyIoxycarbonyIamino-nonanoic acid methyl ester A solution of 1.8 g (4.76 mmol) of (S)-9-bromo-2-cyclopentyloxycarbonylamino-nonanoic acid methyl ester in 20 mL DMF is treated with 1.25 g (19.03 mmol) of sodium azide and stirred at 50 0C for 2 hours. The reaction mixture is quenched with saturated aqueous NaHC03 and extracted with ethylether. The organics are washed with brine, dried over Na2SO4 and concentrated to give the title compound; MS (method D): 341 [M+l]
Step 5
(S)-9-Amino-2-cyclopentyloxycarbonylamino-nonanoic acid methyl ester
A solution of 1.41 g (4.14 mmol) of (S)-9-azido-2-cyclopentyloxycarbonylamino-nonanoic acid methyl ester in 50 mL ethanol is hydrogenated over Pd/Carbon (0.2 g, 10 %) at RT under H2 atmosphere. The reaction mixture is filtered through Celite and the filtrate concentrated to give the title compound; MS (method D): 315 [M+l]
Step 6 (S)-9-(3-tert-Butoxycarbonylamino-4-cycIobutyl-2-hydroxy-butyrylamino)-2- cyclopentyloxycarbonylamino-nonanoic acid methyl ester
A solution of 0.4 g (1.27 mmol) of (S)-9-amino-2-cyclopentyloxycarbonylamino-nonanoic acid methyl ester and 0.417 g (1.52 mmol) of 3-tert-butoxycarbonylamino-4-cyclobutyl-2- hydroxy-butyric acid in 10 mL CH2Cl2 is treated with 0.212 g (1.53 mmol) of l-hydroxy-7- azabenzotriazole and 0.443 g (2.29 mmol) of N-(3-dimethylaminopropyl)-N'-ethyl- carbodiimide hydrochloride, followed by 0.217 mL (1.53 mmol) of triethylamine. The reaction mixture is stirred overnight at RT and chromatographed on silica gel (eluent Hexane / EtOAc 3:2) to give the title compound; MS (method D): 570 [M+l]
Step 7
(S)-9-(3-Aniino-4-cyclobutyI-2-hydroxy-butyryIainino)-2-cycLopentyloxycarbonylainino- αonanoic acid methyl ester
A solution of 0.358 g (0.63 mmol) of (S)-9-(3-tert-butoxycarbonylamino-4-cyclobutyl-2- hydroxy-butyrylamino)-2-cyclopentyloxycarbonylamino-nonanoic acid methyl ester in 1.57 mL of 4N HCl in dioxane is stirred at RT. Upon completion the reaction mixture is concentrated in vacuo to give the title compound; MS (method D): 470 [M+l]
Step 8 4-Fluoro-l,3-dihydro-isoindoIe-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5-[l- cyclobutylmethyl-2-((S)-8-cyclopentyloxycarbonylamino-8-methoxycarbonyl- octylcarbamoylJ^-hydroxy-ethylcarbamoylJ-pyrrolidin-S-yl ester
A solution of 0.306 g (0.65 mmol) of (S)-9-(3-amino-4-cyclobutyl-2-hydroxy-butyrylamino)- 2-cyclopentyloxycarbonylamino-nonanoic acid methyl ester and 0.283 g (0.72 mmol) of (2S,4R)-4-(4-fluoro- 1 ,3-dihydro-isoindole-2-carbonyloxy)-pyrrolidine- 1 ,2-dicarboxylic acid 1-tert-butyl ester 15 mL CH2Cl2 is treated with 0.109 g (0.78 mmol) of l-hydroxy-7- azabenzotriazole and 0.139 mL (0.98 mmol) of triethylamine, followed by 0.227 g (1.17 mmol) of N-(3-dimethylaminopropyl)-N'-ethyl-carbodiimide hydrochloride. The reaction mixture is stirred overnight at RT and chromatographed by RP-HPLC (method G) to give the title compound; MS (method D): 846 [M+l ]
Step 9 4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-l-tert-butoxycarbonyl-5-[2- ((S)-8-carboxy-8-cyclopentyIoxycarbonyIamino-octylcarbamoyl)-l-cyclobutylmethyl-2- hydroxy-ethylcarbamoyl]-pyrrolidin-3-yl ester
A suspension of 0.353 g (0.42 mmol) of 4-fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-1 -tert-butoxycarbonyl-5-[ 1 -cyclobutylmethyl-2-((S)-8- cyclopentyloxycarbonylamino-8-methoxycarbonyl-octylcarbamoyl)-2-hydroxy- ethylcarbamoyl]-pyrrolidin-3-yl ester in 5 mL methanol and 5 mL water is treated with 0.204 g (8.34 mmol) of LiOH and stirred overnight at RT. Methanol is removed in vacuo, the resulting aqueous phase is acidified to pH 6 with 2N HCl and extracted with CH2Cl2. The organics are dried over Na2SO4 to give the title compound; MS (method D): 832 [M+l]
Step 10
4-Fluoro-l^-dihydro-isoindole-2-carboxylic acid (3R,5S)-5-[2-((S)-8-carboxy-8- cyclopentyloxycarbonylamino-octyIcarbamoyI)-l-cycIobutylmethyl-2-hydroxy- ethylcarbamoyl]-pyrrolidin-3-yl ester
The title compound is obtained from 0.302 g (0.254 mmol) of 4-fluoro-l,3-dihydro-isoindole- 2-carboxylic acid (3R,5S)- 1 -tert-butoxycarbonyl-5-[2-((S)-8-carboxy-8- cyclopentyloxycarbonylamino-octylcarbamoyl)-l-cyclobutylmethyl-2-hydroxy- ethylcarbamoyl]-pyrrolidin-3-yl ester according to the procedure described in step 7; MS (method D): 732 [M+l]
Step 11
4-Fluoro-l,3-dihydro-isoindole-2-carboxylic acid (2R,5S,18aS)-16-cyclobutyImethyl-5- cyclopentyloxycarbonylamino-15-hydroxy-4,14,18-trioxo-octadecahydro-3a,13,17- triaza-cyclopentacycIoheptadecen-2-yI ester
A solution of 0.293 g (0.28 mmol) of 4-fluoro-l,3-dihydro-isoindole-2-carboxylic acid (3R,5S)-5-[2-((S)-8-carboxy-8-cyclopentyloxycarbonylamino-octylcarbamoyl)-l- cyclobutyhnethyl-2-hydroxy-ethylcarbamoyl]-pyrrolidin-3-yl ester in 30 mL OfCH2Cl2 is treated with 0.479 mL (2.8 mmol) of Hunig's base, followed by 0.532 g (1.4 mmol) of HATU. The reaction mixture is stirred at RT under completion and chromatographed by RP- HPLC (method G) to give the title compound; MS (method D): 714 [M+l] Synthesis of intermediates Preparation of 5-chloroisoindoline
Figure imgf000355_0001
Prepared as described by T.-Y-Tsai in Bioorg. Med. Chem. Lett. 2006, 16, 3268 starting from 5-chloro- 1 H-isoindole- 1 ,3(2H)-dione.
Preparation of 5-morpholin-4-yIisoindoline
Step l 5-bromoisoindoline
Figure imgf000355_0002
To a mixture of 4.5 g (20 mmol) 5-bromo-l H-isoindole- l,3(2H)-dione inlO mL THF is added 81 mL Borane-THF complex (1 M) and the mixture is refluxed overnight. After cooling to rt 150 mL MeOH and 80 mL 6 N aq. HCl are carefully added and the mixture is refluxed for 1 h. The mixture is concentrated under reduced pressure, water and DCM are added and the aq. layer is extracted with DCM (2x) and ether (2x). The pH of the aq. layer is adjusted to 11 using cone. Aq. NaOH and extracted with DCM (4x). The combined organic layers of this last extraction are dried over Na2SO4, concentrated in vacuo and the residue is used without further purification. LC MS (method E) tR = 0.346 min, M+H = 200.1
Step 2 terf-butyl 5-bromo-l,3-dihydro-2/y-isoindole-2-carboxylate
Figure imgf000355_0003
To a mixture of 2.2 g (11 mmol) 5-bromoisoindoline in 90 mL DCM is added at 0°C a solution of 2.9 g (13 mmol) Boc2O in 20 mL DCM followed by 3.0 mL (20 mmol) TMEDA. The mixture is stirred at 5 °C overnight and 250 mL 2 N aq. HCl is added and the mixture is stirred for additional 20 min at 5 °C. The aq. layer is extracted with DCM and the combined organic layers are dried over Na2SO4 and concentrated under reduced pressure. The residue is purified by FC on silica. HPLC (method C) tR = 4.141 min
Step 3 tert-butyl 5-morpholin-4-yl-l,3-dihydro-2//-isoindθ-e-2-carboxylate
Figure imgf000356_0001
A mixture of 600 mg (2.0 mmol) /er/-butyl 5-bromo-l,3-dihydro-2H-isoindole-2-carboxylate, 0.2 mL (2.4 mmol) morpholine, 268 mg (2.8 mmol) sodium tert.butoxide, 18 mg (0.02 mmol) Pd2(dba)3 and 37 mg (0.06 mmol) rac-BINAP in 4 mL toluene is stirred at 80 0°C for 3 h. The mixture is cooled to rt, ethyl ether is added and the precipitate is filtered off and dried. LC MS (method E) tR = 3.592 min, M+Η = 305.2 ΗPLC (method C) tR = 2.870 min
Step 4
5-morpholin-4-ylisoindoline (hydrochloride)
Figure imgf000356_0002
A mixture of 130 mg (0.4 mmol) tert-butyl 5-morpholin-4-yl-l,3-dihydro-2H-isoindole-2- carboxylate, 4 mL 4 M HCl in dioxane and 4 mL dioxane is stirred for 3.5 h at rt. The mixture is concentrated and the crude is used without further purification. LC MS (method E) tR = 0.264 min, M+Η = 205.1
The following isoindoline can be prepared as described above:
Figure imgf000357_0001
Preparation of l-(2,3-Dihydro-lH-isoindol-5-yl)cyclopropanamine
Step l
S-Cyano-l^-dihydro-isoindole-Z-carboxylic acid tert-butyl ester
Figure imgf000357_0002
A mixture of 0.5 g (1.5 mmol) tert-butyl 5-bromo-l,3-dihydro-2H-isoindole-2-carboxylate, 626 mg (2.0 mmol) Zinc cyanide and 367 mg (0.2 mmol) Pd(PPh3)4 in 15 mL DMF is heated to 80°C for 2h. The mixture is partitioned between water and EtOAc and the aq. layer is extracted with EtOAc. The combined organic layers are washed with brine, dried and concentrated under reduced pressure to give a crude product which is purified by FC (silica gel)-
LC MS (method E) tR = 4.161 min, M+H = 244.9
Step 2
5-(l-Amino-cycIopropyl)-l,3-dihydro-isoindole-2-carboxylic acid tert-butyl ester
Figure imgf000357_0003
To a mixture of 250 mg (1.1 mmol) S-Cyano-l^-dihydro-isoindole^-carboxylic acid tert- butyl ester and 0.3 mL (1.2 mmol) titanium-(VI)-isopropoxide in 5 mL ether is added 0.8 mL (2.3 mmol) ethylmagnesium bromide (3 M in ether) at -70°C. After 5 min the mixture is allowed to reach rt over 1 h and 0.3 mL (2.1mmol) BF3*Et2O is added. After 1 h the mixture is quenched with IN HCl and ether and a basic pH is adjusted using NaOH solution. The aq. layer is extracted with ether and the combined organic layers are dried and concentrated under reduced pressure. The crude product is purified by FC (silica gel)
Step 3 l-(2,3-D'hydro-lH-isoindol-5-yl)cycIopropanamine (dihydrochloride)
Figure imgf000358_0001
95 mg (0.3 mmol) of the 5-(l-Amino-cyclopropyl)-l,3-dihydro-isoindole-2-carboxylic acid tert-butyl ester is dissolved in 2 mL dioxane and 2 mL 4M HCl in dioxane are added. The mixture is stirred at rt for 3 h and concentrated in vacuo to yield the product which is used in the next step without further purification.
Preparation of N-methyl-l,2,3,4-tetrahydroisoquinolin-5-amine
Step l 5-Amino-3,4-dihydro-lH-isoquinoline-2-carboxylic acid tert-butyl ester
Figure imgf000358_0002
To a mixture of 5 g (34 mmol) l,2,3,4-tetrahydro-5-aminoisoquinoline in 150 mL dioxane are added 11 mL aq. NaOH (3M) and 7.4 g (34 mmol) Boc2O at 0°C. The mixture is stirred at rt overnight, ice water is added and the mixture is extracted with EtOAc. The combined organic layers are washed with sat. NaHCO3-solution and brine, dried and concentrated in vacuo. The crude product is used in the next step without further purification. LC MS (method E) tR = 2.636 min, M-Boc+H = 149.2
Step 2
5-Methylamino-3,4-dihydro-lH-isoquinoline-2-carboxylic acid tert-butyl ester and 5- DimethyIamino-3,4-dihydro-lH-isoquinoline-2-carboxylic acid tert-butyl ester
Figure imgf000359_0001
To a mixture of 8.2 g (33 mmol) 5-Amino-3,4-dihydro-lH-isoquinoline-2-carboxylic acid tert-butyl ester in 200 mL THF is added 3.3 g (83 mmol) NaH (60% in mineral oil) at 0°C. After 15 min 6.2 mL (99 mmol) methyliodid is added and the mixture is stirred at rt for 48 h. The mixture is poured on ice water and extracted with EtOAc. The combined organic layer is dried and concentrated to give a mixture of mono- and dimethylated product. The crude product is triturated with MeOH and the unsoluble solid is filtered off to give the pure monomethylated product. The filtrate is concentrated to give a mixture of mono- and dimethylated product. LC MS (method E) tR = 2.076 min, M +H = 277.1 (dimethyl)
LC MS (method E) tR = 3.261 min, M-Boc+H = 263.2 (monomethyl)
Step 3
Methyl-(l,2,3,4-tetrahydro-isoquinoIin-5-yl)-amine (dihydrochloride salt)
Figure imgf000359_0002
300 mg (1.2 mmol) of the pure 5-Methylamino-3,4-dihydro-lH-isoquinoline-2-carboxylic acid tert-butyl ester obtained in step 2 is dissolved in 5 mL dioxane and 5 mL 4M HCl in dioxane are added. The mixture is stirred at rt for 3 h and concentrated in vacuo to yield the product which is used in the next step without further purification. LC MS (method E) tR = 0.256 min, M +H = 163.1
Preparation of N,N-dimethyl-l,2,3,4-tetrahydroisoquinolin-5-amine (dihydrochloride)
Step l Dimethylamino-3,4-dihydro-lH-isoquinoline-2-carboxylic acid tert-butyl ester
Figure imgf000360_0001
1 g of the mixture of mono- and dimethyl product of step 2 of the previous example is dissolved in 10 mL THF and 190 mg NaH (60% in mineral oil) is added at 0°C. After 15 min 0.35 mL methyliodide is added and the mixture is stirred at rt overnight. The mixture is poured on ice water and extracted with EtOAc. The combined organic layer is dried and concentrated to give the dimethylated product. LC MS (method E) tR = 2.076 min, M +H = 277.1
Step 2
N,N-dimethyl-l,2,3,4-tetrahydroisoquinolin-5-amine (dihydrochloride)
Figure imgf000360_0002
1.3 g (4.7 mmol) of the pure Dimethylamino-3,4-dihydro-lH-isoquinoline-2-carboxylic acid tert-butyl ester obtained in step 1 is dissolved in 15 mL dioxane and 15 mL 4M HCl in dioxane are added. The mixture is stirred at rt overnight and concentrated in vacuo to yield the product which is used in the next step without further purification. LC MS (method E) tR = 0.349 min, M +H = 177.3 Preparation of 2-(4-methyl-piperazin-l-yl)-6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidine
Figure imgf000360_0003
Step 1
3-[l-Dimethylamino-methylidene]-4-oxo-pyrrolidine-l-carboxylic acid tert-butyl ester A mixture of 15.72 g (84.87 mmol) of N-(tert-butoxycarbonyl)-3-pyrrolidinone and 82 mL of N,N-dimethylformamide dimethylacetal is heated up at reflux for 1.5 hour. Excess of N,N- dimethylformamide dimethylacetal is removed in vacuo, the residue is triturated with n- hexane to provide a solid that is dried in vacuo; MS (method D): 241 [M+l]
Step 2
Z-^-Methyl-piperazin-l-yO-S^-dihydro-pyrroloβ^-dlpyrimidine-ό-carboxylic acid tert-butyl ester A mixture of 0.39 g (1.62 mmol) of 3-[l-dimethylamino-methylidene]-4-oxo-pyrrolidine-l- carboxylic acid tert-butyl ester, 0.98 g (2.43 mmol) of 4-methylpiperazine-l- carboximidamide and 1.35 mL of sodium methoxide (5.4M in methanol) in 10 mL of ethanol is heated up at reflux overnight. The reaction mixture is poured into ice-water and extracted with EtOAc, the organics are washed with brine and dried over Na2SO4. Purification by RP- HPLC (method G) gives the title compound; MS (method D): 320 [M+l]
Step 3
2-(4-Methyl-piperazin-l-yI)-6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidine
A solution of 0.16 g (0.5 mmol) of 2-(4-methyl-piperazin-l-yl)-5,7-dihydro-pyrrolo[3,4- d]pyrimidine-6-carboxylic acid tert-butyl ester in 1 mL 1,4-dioxane is treated with 1.9 mL of 4N HCl in dioxane and stirred at RT under completion. The reaction mixture is concentrated in vacuo, taken up in 2N NaOH aqueous solution and extracted with EtOAc. The organics are dried over Na2SO4 and concentrated in vacuo to give the title compound; MS (method D): 220 [M+l]
The following compounds are prepared in an analogous manner
(6,7-Dihydro-5H-pyrrolo[3,4-d]pyrimidin-2-yl)-dimethyl-amine: MS (method D): 165 [M+l]
2-Pyrrolidin-l-yl-6,7-dihydro-5H-pyrrolo[3,4-dlpyrimidine: MS (method D): 191 [M+l] 2-Morpholin-4-yl-6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidine: MS (method D): 207 [M+l] Preparation of (6,7-dihydro-5H-pyrroIo[3,4-d]pyrimidin-4-yI)-dimethyl-ainine
Figure imgf000362_0001
Step 1 l-Chloro^-dimethylamino-S.T-dihydro-pyrroloP^-dlpyrimidine-ό-carboxylic acid tert- butyl ester
A solution of 0.2 g (0.66 mmol) of 2,4-dichloro-5,7-dihydro-pyrrolo[3,4-d]pyrimidine-6- carboxylic acid tert-butyl ester in 8 mL of ethanol is treated with 0.103 mL (0.73 mmol) of triethylamine and 0.118 mL of a dimethylamine solution in ethanol (5.6 M). The vial is sealed and the reaction mixture is stirred at RT for 3 hours. The solvent is removed in vacuo and the residue is chromatographed on silica gel (eluent Hexane/EtOAc 4:1) to give the title compound; MS (method D): 299 [M+l], Rf 0.25 (eluent Hexane/EtOAc 3:1)
Step 2
4-DimethyIamino-5,7-dihydro-pyrrolo[3,4-d]pyriinidine-6-carboxylic acid tert-butyl ester
A solution of 0.08 g (0.27 mmol) of 2-chloro-4-dimethylamino-5,7-dihydro-pyrrolo[3,4- d]pyrimidine-6-carboxylic acid tert-butyl ester in 10 mL of methanol is treated with 4 mL of triethylamine and degassed. Pd on Carbon (10%, 20 mg) is added and the reaction is allowed to stir overnight under an H2 atmosphere. Under completion the catalyst is removed by filtration and the filtrate is chromatographed on silica gel (eluent Hexane / EtOAc 1 : 1) to afford the title compound; MS (method D): 265 [M+l]
Step 3
(6,7-Dihydro-5H-pyrrolo[3,4-d]pyrimidin-4-yl)-dimethyl-ainine
A solution of 0.067 g (0.25 mmol) of 4-dimethylamino-5,7-dihydro-pyrrolo[3,4- d]pyrimidine-6-carboxylic acid tert-butyl ester in 1 mL of 1,4-dioxane is treated with 0.95 mL of 4N HCl in 1,4-dioxane. Under completion the reaction mixture is freeze-dried to give the title compound; MS (method D): 165 [M+ 1]
Preparation of (S)-3-(3-chIoro-phenyl)-l-oxa-2,7-diaza-spiro[4.4]non-2-eiie-7,8- dicarboxylic acid 7-tert-butyl ester
Figure imgf000363_0001
Step l (S)-4-Oxo-pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester 2-methyI ester
A solution of 2.14 g (10.01 mmol) of sodium metaperiodate in 25 mL of water is added to a well stirred suspension of 0.168g (1.26 mmol) of ruthenium(IV)oxide hydrate in 10 mL CCl4 at 00C to give a yellow organic phase. A solution of 1.23 g (5.02 mmol) of Boc-Cis-HYP- OMe in chloroform is added in one portion. The ice bath is removed and the reaction mixture is allowed to stir at RT for 1.5 hour. The organic layer is separated, the water phase is extracted with ethylether. The organics are treated with 2-propanol, dried over Na2SO4, filtered over Celite and concentrated in vacuo to afford the title compound; MS (method D): 242 [M-I]
Step 2
(S)-4-Methylene-pyrroIidine-l,2-dicarboxyIic acid 1-tert-butyl ester 2-methyl ester
A suspension of 0.3 g (2.59 mmol) of potassium tert-butoxide 20 mL of ethylether at O0C is treated with 0.944 g (2.59 mmol) of methyl-triphenylphosphoniumbromide, followed by 0.45 g (1.85 mmol) of (S)-4-oxo-pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester 2-methyl ester after 15 minutes. The resulting brown mixture is heated up to reflux for 4 hours, poured into an ice-cold solution of ammonium chloride, and extracted with ethylether. The organic phase is dried over Na2SO4, concentrated and chromatographed on silica gel (eluent Hexane / EtOAc 6: 1) to give the title compound; Rf 0.44 (eluent Hexane/EtOAc 3:1)
Figure imgf000364_0001
Step l 3-Chloro-benzaldehyde oxime
To a solution of 7.24 g (51.51 mmol) of 3-chlorobenzaldehyde and 3.941 g (56.14 mmol) of hydroxylamine hydrochloride in water (13 mL) and ethanol (13 mL) is added ice (25 g), followed by a 50% NaOH solution (5 mL). The resulting solution is stirred for 1 hour, acidified with concentrated HCl, and extracted with CH2Cl2. The organics are washed with water, dried over Na2SO4 and concentrated to give the title compound; MS (method D): 154 [M-I]
Step 2
3-Chlorobenzohydroximinoyl chloride
A mixture of 0.5 g (3.21 mmol) of 3-chloro-benzaldehyde oxime and 0.447 g (3.21 mmol) of N-chlorosuccinimide in 5 mL DMF is stirred at 60 0C for 45 min. The reaction mixture is poured into ice-water, extracted with ethylether. The organics are washed with brine, dried over Na2SO4 and concentrated in vacuo to give the title compound; HPLC (method A) tR (min) 4.17
Figure imgf000364_0002
Step l
(S)-3-(3-Chloro-phenyl)-l-oxa-2,7-diaza-spiro[4.4]non-2-ene-7,8-dicarboxyIic acid 7- tert-butyl ester 8-methyl ester
A solution of 0.15 g (0.62 mmol) of (S)-4-methylene-pyrrolidine-l,2-dicarboxylic acid 1-tert- butyl ester 2-methyl ester in 10 mL of EtOAc is treated with 0.154 g (0.81 mmol) of 3- chlorobenzohydroximinoyl chloride below 7 0C, followed by 0.114 mL (0.81 mmol) of triethylamine. The reaction mixture is stirred at RT overnight, poured into ice-water / EtOAc. The organics are washed with brine, dried over Na2SO4, concentrated and chromatographed to give the title compound; HPLC (method A) tR (min) 4.8 and 4.9 (4: 1 ratio)
Step 2 (S)-3-(3-Chloro-phenyl)-l-oxa-2,7-diaza-spiro[4.4]non-2-ene-7,8-dicarboxyIic acid 7- tert-butyl ester
A solution of 0.12 g (0.30 mmol) of (S)-3-(3-chloro-phenyl)-l-oxa-2,7-diaza-spiro[4.4]non- 2-ene-7,8-dicarboxylic acid 7-tert-butyl ester 8-methyl ester in methanol (3 mL) and water 1.5 mL) is treated with 0.371 g (15.2 mmol) of LiOH and stirred at RT for 1 hour. The reaction mixture is poured into 6N HCl, extracted with CH2Cl2. The organics are combined, dried over Na2SO4 and concentrated to give the title compound; MS (method D): 379 [M-I]
The following compound is prepared in an analogous manner: (SVS-Pyridin-S-yH-oxa^^-diaza-spiro^^Jnon^-ene^.δ-dicarboxylic acid 7-tert-butyl ester: MS (method D): 348 [M+l]
Preparation of (2S,4R)-4-(6-chloro-benzo[d]isoxazol-3-yloxy)-pyrrolidine-l,2- dicarboxylic acid 1-tert-butyl ester
Figure imgf000365_0001
Step l
(IS^R^-^-Chloro-benzofdlisoxazoW-yloxy^pyrrolidine-l^-dicarboxylic acid 1-tert- butyl ester 2-methyI ester
A solution of 1 g (4.08 mmol) of Boc-cis-HYP-OMe in 70 mL of THF is cooled to 0 0C, treated with 0.784 g (4.48 mmol) of 6-chlorobenzo(d)isoxazol-3-ol, 1.62 g (6.12 mmol) of triphenylphosphine and after 5 minutes, 1.26 mL (6.12 mmol) of diisopropyl azodicarboxylate. The reaction mixture is stirred at RT overnight, concentrated and chromatographed by RP-HPLC (method G) to give the title compound; MS (method D): 297 [M-Boc+ 1]
Step 2 (2S,4R)-4-(6-Chloro-benzo[d]isoxazol-3-yloxy)-pyrrolidine-l,2-dicarboxylic acid 1-tert- butyl ester
A solution of 1.24 g (3.12 mmol) of (2S,4R)-4-(6-chloro-benzo[d]isoxazol-3-yloxy)- pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester 2-methyl ester in methanol (3 mL) and water 1.5 mL) is treated with 0.382 g (15.6 mmol) of LiOH and stirred at RT for 1 hour. The reaction mixture is poured into 6N HCl, extracted with CH2Cl2. The organics are combined, dried over Na2SO4 and concentrated to give the title compound; MS (method D): 381 [M-I]
The following compounds are prepared in an analogous manner: (2S,4R)-4-(Isoxazolo[4,5-bJpyridin-3-yIoxy)-pyrrolidine-l,2-dicarboxylic acid 1-tert- butyl ester: MS (method D): 348 [M-I]
(2S,4R)-4-(Isoxazolo [5,4-c] pyridin-3-y Ioxy)-py rrolidine- 1 ,2-dicarboxy lie acid 1 -tert- butyl ester: MS (method D): 348 [M-I]
Preparation of (lR,2R)-l-tert-butoxycarbonylamino-2-ethyl-cyclopropanecarboxylic acid methyl ester
Figure imgf000366_0001
A solution of 15.94 g (66 mmol) of (lR,2S)-l-tert-butoxycarbonylamino-2-vinyl- cyclopropanecarboxylic acid methyl ester in 300 mL t-butyl-methyl ether is hydrogenated over 1.6 g of Pd(OH)2 on Carbon (20%, wet) under H2 atmosphere at RT, and under atmospheric pressure. The catalyst is filtered-off and the residue concentrated in vacuo to give the title compound; MS (method D): 242 [M-I]
BIOLOGICAL ACTIVITY
Example 227: HCV NS3-4A protease assay The inhibitory activity of certain compounds of Table A against HCV NS3-4A serine protease is determined in a homogenous assay using the full-length NS3-4A protein (genotype Ia, strain HCV-I) and a commercially available internally-quenched fluorogenic peptide substrate as described by Taliani, M., et al. 1996 Anal. Biochem. 240:60-67, which is incorporated by reference in its entirety.
Example 228: Luciferase-based HCV replicon assay
The antiviral activity and cytotoxicity of certain compounds of Table A is determined using a subgenomic genotype Ib HCV replicon cell line (Huh-Luc/neo-ET) containing a luciferase reporter gene, the expression of which is under the control of HCV RNA replication and translation. Briefly, 5,000 replicon cells are seeded in each well of 96- well tissue culture plates and are allowed to attach in complete culture media without G418 overnight. On the next day, the culture media are replaced with media containing a serially diluted compound of Table A in the presence of 10% FBS and 0.5% DMSO. After a 48-h treatment with the compound of Table A, the remaining luciferase activities in the cells are determined using BriteLite reagent (Perkin Elmer, Wellesley, Massachusetts) with a LMaxII plate reader (Molecular Probe, Invitrogen). Each data point represents the average of four replicates in cell culture. IC50 is the concentration of the at which the luciferase activity in the replicon cells is reduced by 50%. The cytotoxicity of the compound of Table A is evaluated using an MTS-based cell viability assay. Compounds Table A supra have been tested in at least one of the protease assay of
Example 227 or the replicon assay of Example 228 and exhibit an IC5O of less than about 10 μM or less in at least one of the assays recited in Example 227 and 228.
Equivalents Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments and methods described herein. Such equivalents are intended to be encompassed by the scope of the following claims.

Claims

CLAIMSWhat is claimed is:
1. A compound of formula I:
Figure imgf000368_0001
and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof; wherein the macrocycle:
Figure imgf000368_0002
comprises between 15 to 40 ring atoms; m, x and z are each independently selected from 0 or 1 ; p is selected at each occurrence from the group consisting of 0, 1 and 2; Ri and R2 are independently selected, at each occurrence, from hydrogen or cyano, or from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, alkoxy, and cycloalkyloxy, each of which is unsubstituted or substituted with 1-6 moieties which can be the same or different and are independently selected from the group consisting of hydroxy, oxo, alkyl, aryl, alkoxy, aryloxy, thio, alkylthio, arylthio, amino, alkylamino, arylamino, alkylsulfonyl, arylsulfonyl, alkylsulfonamido, arylsulfonamido, heteroarylsulfonamido, arylaminosulfonyl, heteroarylaminosulfonyl, mono and dialkylaminosulfonyl, carboxy, carbalkoxy, amido, carboxamido, alkoxycarbonylamino, aminocarbonyloxy, alkoxycarbonyloxy, alkylureido, arylureido, halogen, cyano, or nitro; wherein each of said alkyl, alkoxy, and aryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different and are independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkyl-alkyl, heterocyclyl, heterocyclylalkyl, aryl, alkylaryl, aralkyl, arylheteroaryl, heteroaryl, heterocyclylamino, alkylheteroaryl and heteroaralkyl;
R3 is selected from the group consisting of H and Ci-4-alkyl; E is a divalent residue selected from the group consisting OfC(O)NR23, NR23S(O)P,
NR23S(O)PNR23;
Li and L2 are divalent residues independently selected from the group consisting of Co^alkylene, (CH2),-FG-(CH2)k, (CH2),-C3-7cycloalkylene-(CH2)k, (CH2J1-C3. 7cycloheteroalkylene-(CH2)ic, alkenylene, alkynylene, arylene, heteroarylene, cycloalkylene and heterocycloalkylene, each of which is substituted with 0 to 4 independently selected X1 or X2 groups; i and k are independently selected integers of from 0 to 7;
L3 is a Co-4alkylene or a divalent ethylene or acetylene residue, wherein the Co- 4alkylene and divalent ethylene residues are substituted by 0-2 substituents selected from alkyl, aryl, heteroaryl, mono- or di-alkylamino-Co-Cόalkyl, hydroxyl alkyl or alkoxyalkyl;
FG is absent or a divalent residue selected from the group consisting of O, S(O)P, NR23, C(O), C(O)NR23, NR23C(O), OC(O)NR23, NR23C(O)O, NR23C(O)NR23, S(O)pNR23, NR23S(O)p, and NR23S(O)PNR23;
R23 is independently selected at each occurrence from hydrogen or the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, aryl, heteroaryl, heteroaralkyl, aralkyl and heteroaralkyl, each of which is substituted with 0-2 substituents independently selected from halogen, alkyl, alkoxy, and mono- and di-alkylamino; or
Two R23 residues, taken in combination, form a monocyclic, bicyclic or tricyclic heterocyclic ring system which is saturated, partially unsaturated, or aromatic, and which is substituted with O to 3 substituents independently selected from Ci-6alkyl, C1-6alkoxy, Q- 6alkoxyCi-6alkoxy, mono- and
Figure imgf000369_0001
mono- and di-Ci-6alkylamino, halogen, 4 to 7 member heterocycloalkyl, aryl, heteroaryl, and 3 to 6 member spirocycloalkyl or spiroheterocycloalkyl, each of which is substituted with O to 3 substituents independently selected from
Figure imgf000369_0002
Ci-4alkoxy, hydroxy, amino, and mono- and
Figure imgf000369_0003
R7, Rio, Rn, Ri2, Ri3, Ri5, Ri6, Rn, and R22 are each, independently, selected from hydrogen or the group consisting of alkyl, alkenyl, alkynyl, aryl, alkyl-aryl, heteroalkyl, heterocyclyl, heteroaryl, aryl-heteroaryl, alkyl-heteroaryl, cycloalkyl, alkyloxy, alkyl- aryloxy, aryloxy, heteroaryloxy, heterocyclyloxy, cycloalkyloxy, amino, alkylamino, arylamino, alkyl-arylamino, arylamino, heteroarylamino, cycloalkylamino, carboxyalkylamino, aralkyloxy and heterocyclylamino; all of which may be further substituted 0 to 5 times with substituents independently selected from Xi and X2;
R.9 is absent or selected from hydrogen, C^alkyl, C3-7CyC loalkyl-Co^alkyl, or hydroxy;
Xi is alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkyl-alkyl, heterocyclyl, heterocyclylalkyl, aryl, alkylaryl, aralkyl, arylheteroaryl, heteroaryl, heterocyclylamino, alkylheteroaryl, or heteroaralkyl; wherein X1 can be independently substituted with one or more of X2 moieties which can be the same or different and are independently selected; X2 is hydroxy, oxo, alkyl, aryl, heteroaryl, alkoxy, aryloxy, heteroaryloxy, thio, alkylthio, arylthio, heteroarylthio, amino, alkylamino, arylamino, heteroarylamino, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, alkylsulfonamido, arylsulfonamido, heteroarylsulfonamido, arylaminosulfonyl, heteroarylaminosulfonyl, mono and dialkylaminosulfonyl, carboxy, carbalkoxy, amido, carboxamido, alkoxycarbonylamino, aminocarbonyloxy, alkoxycarbonyloxy, carbamoyl, ureido, alkylureido, arylureido, halogen, cyano, or nitro; wherein each of said alkyl, alkoxy, and aryl can be unsubstituted or optionally independently substituted with one or more moieties which can be the same or different and are independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkyl- alkyl, heterocyclyl, heterocyclylalkyl, aryl, alkylaryl, aralkyl, arylheteroaryl, heteroaryl, heterocyclylamino, alkylheteroaryl and heteroaralkyl; Zi is Co^alkylene, oxygen or NRi0; Z2 is CR9, O or N; R14 is C(O) or S(O)P;
V is selected from hydrogen or from the group consisting of alkyl, alkyl-aryl, heteroalkyl, heterocyclyl, heteroaryl, aryl-heteroaryl, alkyl-heteroaryl, cycloalkyl, alkyloxy, alkyl-aryloxy, aryloxy, heteroaryloxy, heterocyclyloxy, cycloalkyloxy, amino, alkylamino, arylamino, alkyl-arylamino, arylamino, heteroarylamino, cycloalkylamino, carboxyalkylamino, mono- and di-alkylcarboxamide, aralkyloxy and heterocyclylamino; each of which may be further independently substituted one or more times with X1 and X2; wherein X1 is alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkyl-alkyl, heterocyclyl, heterocyclylalkyl, aryl, alkylaryl, aralkyl, aryloxy, arylthio, arylheteroaryl, heteroaryl, heterocyclylamino, alkylheteroaryl, or heteroaralkyl; wherein X1 can be independently substituted with one or more X2 moieties which can be the same or different and are independently selected; wherein X2 is hydroxy, oxo, alkyl, cycloalkyl, spirocycloalkyl, heterocycloalkyl, aryl, heteroaryl, alkoxy, aryloxy, thio, alkylthio, amino, mono- and di- alkylamino, arylamino, alkylsulfonyl, arylsulfonyl, alkylsulfonamido, arylsulfonamido, carboxy, carbalkoxy, carboxamido, alkoxycarbonylamino, alkoxycarbonyl, alkoxycarbonyloxy, alkylureido, arylureido, halogen, cyano, or nitro; wherein each X2 residue selected to be alkyl, alkoxy, and aryl can be unsubstiruted or optionally independently substituted with one or more moieties which can be the same or different and are independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkyl-alkyl, heterocyclyl, heterocyclylalkyl, aryl, alkylaryl, aralkyl, arylheteroaryl, heteroaryl, heterocyclylamino, alkylheteroaryl and heteroaralkyl; or V is selected from the group consisting of-Q'-Q2, wherein Q1 is absent, C(O),
S(O)2, N(H), N(Ci-4-alkyl), C-N(CN), C=N(SO2CH3), C=N-COH-C M-alkyl, or C=N-COH, and Q2 is hydrogen or is selected from the group consisting of Ci-4-alkyl, O-C^-alkyl, NH2, N(H)-C i-4-alkyl, N(Ci-4-alkyl)2, SO2-aryl, SO2-heteroaryl, SO^C^-alkyl, C3-6-cycloalkyl-C0- 4-alkyl, aryl, heteroaryl and heterocycle, each of which may be independently substituted one or more times with a halogen atom, Ci-4-alkyl, C1-4-alkyl substituted by one or more halogen atoms, or C3-6-cycloalkyl; or R22 and Ri6 may together form a 3, 4, 5, 6 or 7-membered ring and may contain one or more heteroatoms, wherein the ring may be further substituted one or more times; or R7 and Ri 5 may together form a 3, 4, 5, 6 or 7-membered ring and may contain one or more heteroatoms, wherein the ring may be further substituted one or more times; or Ri5 and Rn may together form a 3, 4, 5, 6 or 7-membered ring and may contain one or more heteroatoms, wherein the ring may be further substituted one or more times; or Ri5 and Ri6 may together form a 4, 5, 6 or 7-membered ring and may contain one or more heteroatoms, wherein the ring may be further substituted one or more times; or Ri 5 and Ri6 may together form an arylene or heteroarylene ring and R7 and R22 are absent, wherein the ring may be further substituted one or more times; or Ri and R2 may together form a 3, 4, 5, 6 or 7-membered ring that is saturated or partially unsaturated and may contain one or more heteroatoms, which ring is substituted with 0-3 residues independently selected from C^alkyl, Ci ^alkoxy, C2-4alkenyl, C2-4alkynyl, halogen, hydroxy, C3-6cylcoalkyl and C3-6spirocycloalkyl; or Ri7 and Ri6 may together form a 4, 5, 6, 7 or 8-membered ring of the formula:
Figure imgf000372_0001
wherein n and g are each, independently, 0, 1 or 2;
X is O, S, N, C or CR5a; R4 is hydrogen or is selected from the group consisting of Ci^-alkyl, C3-7-cycloalkyl, aryl, heterocycle and heteroaryl, all of which may be independently substituted one or more times with a halogen atom or Ci-4-alkyl;
R5 is absent, hydrogen or oxo or is selected from the group consisting of hydroxyl, C1- 8-alkyl, C2-8-alkenyl, C2-8-alkynyl, C3-8-cycloalkyl-Co-4-alkyl, aryl-C0-4-alkyl, heterocycle-Co- 4-alkyl, heteroaryl-Co-4-alkyl , C3-8-cycloalkyloxy, aryloxy, NR23COR23, CONR23R23, NR23CONHR23, OCONR23R23, NR23COOR23, OCOR23, COOR23, aryl-C(O)O, aryl- C(O)NR23, heteroaryloxy, heteroaryl-C(O)O, heterocycle-C(O)O, heteroaryl-C(O)NR23, heterocycle-C(O)NR23, each of which may be independently substituted one or more times (or more preferably O, 1, 2, 3, 4, or 5 times) with halogen, Ci-4-alkyl, C^-alkoxy, 1IaIoCi-4- alkyl, haloCi-4-alkoxy, amino, mono- and di-Ct^alkylaminoCo^alkyl, mono- and di-Q. 4alkylaminoC0-4alkoxy, C3-7cycloalkyl, fused- or spiro-cyclic 3-7 membered ring, heterocycleC0-4alkoxy, heterocycleCo^alkyl, aryl, or heteroaryl;
R5a is selected from the group consisting of H, hydroxyl, Ci-8-alkyl, C2-8-alkenyl, C2-8- alkynyl, C3-8-cycloalkyl-Co-4-alkyl, aryl-Co-4-alkyl and heteroaryl-Co-4-alkyl, or R4 and R5 may together form a fused dimethyl cyclopropyl ring, a fused cyclopentane ring, a fused phenyl ring or a fused pyridyl ring, each of which may be substituted with a halogen atom, aryl, heteroaryl, trihalomethyl, C^-alkoxy or Q-4-alkyl; or R5 and R5a may together form a spirocyclic ring having between 3 and 7 ring atoms and having O, 1 , or 2 ring heteroatoms, which is optionally substituted by 0-4 substitutents selected from cyano, halogen, hydroxyl, amino, thiol, Ci-8-alkyl, C2-8-alkenyl, C2-g-alkynyl, Ci-8-alkoxy-Co-4alkyl, Ci-8-haloalkyl, C2-8-haloalkenyl, C2-8-haloalkynyl, Ci-8-haloalkoxy, Q- 8-alkylthio, C1-8-alkylsulfonyl, Ci.g-alkylsulfoxy, C1-8-alkanoyl, Cug-alkoxycarbonyl, C3-7- cycloalkyl-Co-4-alkyl, aryl-Co-4-alkyl, heteroaryl-Co-4-alkyl, COOH, C(O)NH2, mono- and di- C1-4-alkyl-carboxamide, mono- and di-Ci-4-alkyl-amino-Co^talkyl, SO3H, SO2NH2, and mono-and di-Ci-4-alkylsulfonamide, or two substitutents taken together form a fused or spirocyclic 3 to 7 membered ring having O, 1 or 2 ring heteroatoms selected from N, O and S, which fused or spirocyclic ring has 0 to 2 independently selected substitutents selected from cyano, halogen, hydroxyl, amino, thiol, Ci-8-alkyl, C2-8-alkenyl, C2-8-alkynyl, C1-8-alkoxy-Co- 4alkyl, Ci-8-haloalkyl, C2-8-haloalkenyl, C2-8-haloalkynyl, Ci-g-haloalkoxy, Ci-8-alkylthio, Ci- 8-alkylsulfonyl, Ci-8-alkylsulfoxy, Ci-8-alkanoyl, Ci-8-alkoxycarbonyl, C3-7-cycloalkyl-Co-4- alkyl, aryl-C0-4-alkyl, heteroaryl-C0-4-alkyl, COOH, C(O)NH2, mono- and di-C]-4-alkyl- carboxamide, mono- and
Figure imgf000373_0001
SO3H, SO2NH2, and mono-and di-Ci. 4-alkylsulfonamide; and
R^ is independently selected at each occurrence from the group consisting of hydrogen, hydroxy, amino, Ci-4alkyl,
Figure imgf000373_0002
and C3- δCycloalkylCo^alkyl; or two R6 residues may together form a spirocyclic ring having between 3 and 7 ring atoms and having O, 1, or 2 ring heteroatoms, which is optionally substituted by 0-4 substitutents selected from cyano, halogen, hydroxyl, amino, thiol, Ci-s-alkyl, C2-8-alkenyl, C2-8-alkynyl, Ci-8-alkoxy-Co-4alkyl, Ci-8-haloalkyl, C2-s-haloalkenyl, C2-8-haloalkynyl, Ci-8- haloalkoxy, Ci-8-alkylthio, Ci-8-alkylsulfonyl, Ci-8-alkylsulfoxy, Ci-8-alkanoyl, Ci-8- alkoxycarbonyl,
Figure imgf000373_0003
COOH, C(O)NH2, mono- and
Figure imgf000373_0005
mono- and
Figure imgf000373_0004
SO3H, SO2NH2, and mono-and di-Ci-4-alkylsulfonamide, or two substitutents taken together form a fused or spirocyclic 3 to 7 membered ring having O, 1 or 2 ring heteroatoms selected from N, O and S, which fused or spirocyclic ring has 0 to 2 independently selected substitutents selected from halogen,
Figure imgf000373_0006
C1-4alkanoyl, mono- and di-Ci-4- alkylamino, mono- and di-Q^-alkyl-carboxamide, Cj-4-alkoxycarbonyl, and phenyl.
2. A compound of claim 1, wherein R] and R2 taken in combination form a 3, 4, 5, or 6-membered saturated carbocyclic ring which is substituted with 0-2 substituents independently selected from halogen, alkyl, alkenyl, alkoxy and C3-6cycloalkyl.
3. A compound of claim 1 wherein R2 and one occurrence of Ri taken in combination form a cyclopropyl ring which is substituted with 0 or 1 substituents selected Ci- 4alkyl, vinyl or cyclopropyl; E is C(O)NH, NHS(O)2, NHSO2N(Me), NHSO2N(Et) or NHSO2N(cyclopropyl).
4. The compound of claim 1, wherein the macrocycle:
Figure imgf000374_0001
comprises between 15 to 25 ring atoms.
5. The compound of claim 1, wherein the macrocycle:
Figure imgf000374_0002
comprises between 17 to 23 ring atoms.
6. The compound of claim 1, wherein L1 is CrQalkylene, C3-C7cycloalkylene, arylene or heteroarylene each of which is substituted by 0-4 residues independently selected from Ci-Cialkyl, Ci-C4alkoxy, hydroxyl, amino, mono- and di- Ci-C4alkylamino, halogen, cyano, Ci-C^fluoroalkyl, Ci- C4fluoroalkoxy, COOH, carboxamide (CONH2), mono- and di-Ci-C4alkylcarboxamide, aryl, heteroaryl and 5 or 6 membered saturated heterocycles; L2 is selected from Ci-C6alkylene and C2-C6alkenylene, each of which is substituted by 0-4 residues independently selected from Ci-C4alkyl, Cι-C4alkoxy, hydroxyl, amino, mono- and di- Ci-C4alkylamino, halogen, cyano, Ci-C4fluoroalkyl, Ci-C4iluoroalkoxy, COOH, carboxamide (CONH2), mono- and
Figure imgf000374_0003
aryl, heteroaryl, and 5 or 6 membered saturated heterocycles; and L3 is absent or a divalent ethylene residue which is substituted by 0 to 2 independently selected methyl or ethyl residues.
7. The compound of claim 6, wherein Lj is a divalent residue selected from C2- C4alkylene, 1 ,2-phenylene, 1,3-phenylene, 2,4-pyridylene, 2,3-pyridylene, 3,4-pyridylene or 1,7-indolylene, 2,7-indolylene, each of which is substituted with 0-3 residues selected from Ci-C4alkyl, Ci-C4alkoxy, hydroxyl, amino, mono- and di- Ci-C4alkylamino, halogen, cyano, Ci-C2fluoroalkyl, Cι-C2fluoroalkoxy, COOH, carboxamide (CONH2), and mono- and di-Q- C4alkylcarboxamide.
8. The compound of claim 1, wherein R5 is a residue of the formula:
Figure imgf000375_0001
wherein n and g are integers independently selected from 0, 1, or 2;
Z3 is NR23 or O;
Z4, Z5, Z6, and Z7 are each independently selected from the group consisting of N, CH, and CR8; and
R8 and Rsa each indepently represent 0 to 2 groups, each of which is independently selected at each occurrence ofR8 and R83 from the group consisting of hydrogen, halogen, Ci- 4-alkyl,
Figure imgf000375_0002
4alkyl, mono- and di-Ci^alkylaminoCo^alkoxy, heterocycleCo^alkoxy, and heterocycleCo- 4alkyl.
9. The compound of claim 1 , wherein E is C(O)NH;
Ri is H or C 1-4 alkyl; and
R2 is H, Ci-C4alkyl, Ci-C4fluoroalkyl, C2-C4alkenyl, or C3-C7cycloalkylC0-2alkyl.
10. A compound of claim 1 wherein the compound is a compound of formula II:
Figure imgf000375_0003
II and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof.
11. The compound of claim 10, wherein x is 0 or 1 ; n is 0 or 1 ; R14 is C(O) or S(O)P;
Zi is absent or NH;
Z2 is nitrogen or CH;
R1 is selected from the group consisting of H and C^-alkyl;
R2 is selected from the group consisting of Ci-4-alkyl, C(O)C i^-alkyl, C(O)OCi-4- alkyl, and (CH2)0-4-C3-6-cycloalkyl; or R1 and R2 together form a cyclopropyl ring which is substituted with 0 or 1 substituents selected Ci^alkyl, vinyl or cyclopropyl;
R3 is selected from the group consisting of H and Ci-4-alkyl;
X is O, NR5 or CR5R53; R4 is hydrogen or is selected from the group consisting of Ci-4-alkyl, C3-ό-cycloalkyl, aryl, heterocycle and heteroaryl, each of which may be independently substituted one or more times with a halogen atom or d^-alkyl;
R5 is hydrogen or oxo or is selected from the group consisting of hydroxyl, Ci-8-alkyl, C2.8-alkenyl, C2-g-alkynyl, C3-8-cycloalkyl-Co-4-alkyl, aryl-C0-4-alkyl, aryloxy, heteroaryloxy, heterocycle-C0-4-alkyl and heteroaryl-Co-4-alkyl, each of which may be independently substituted one or more times with a halogen atom, aryl, heteroaryl, trihalomethyl, Ci-4- alkoxy or d-4-alkyl; or
R5 is a residue of the formula:
Figure imgf000376_0001
wherein n and g are integers independently selected from 0, 1, or 2; Z3 is NR23 or O;
Z4, Z5, Z6, and Z7 are each independently selected from the group consisting of N, CH, and CR8; R8 and R83 each indepently represent 0 to 2 groups, each of which is independently selected at each occurrence OfR8 and R83 from the group consisting of hydrogen, halogen, Ci- 4-alkyl, Ci-4-alkoxy, haloC1-4-alkyl, haloCi-4-alkoxy, amino, mono- and di-Ci^alkylaminoCo- 4alkyl, mono- and
Figure imgf000377_0001
heterocycleC0-4alkoxy, heterocycleCo- 4alkylamino, and heterocycleCo^alkyl;
R53 is selected from the group consisting of H, hydroxyl, Ci-8-alkyl, C2-8-alkenyl, C2-8- alkynyl, C3-8-cycloalkyl-Co-4-alkyl, aryl-Co-4-alkyl and heteroaryl-C0-4-alkyl, or R4 and R5 may together form a fused dimethyl cyclopropyl ring, a fused cyclopentane ring, a fused phenyl ring or a fused pyridyl ring, each of which may be substituted with a halogen atom, aryl, heteroaryl, trihalomethyl, Ci-4-alkoxy or Ci-4-alkyl; or R5 and R5a may together form a spirocarbocyclic saturated ring having between 3 and 6 carbon ring atoms which is optionally substituted by 0-2 substitutents selected from halogen, Ci-6-alkyl, C2-6-alkenyl, C2-6-alkynyl,
Figure imgf000377_0002
C3-7-cycloalkyl-Co-4-alkyl, phenyl-Co-4-alkyl, naphthyl-Co-4-alkyl, heteroaryl-Co-4-alkyl, or two substitutents taken together form a fused or spirocyclic 3 to 7 membered carbocyclic ring, each of which is substituted with 0-3 independently selected halogen atoms or C^-alkyl groups;
Rio and Rn are each, independently, selected from the group consisting of H and Ci-4- alkyl;
R6 and Ri 3 is H; Ri2 is selected from the group consisting of H, Ci-4-alkyl and C3-6-cycloalkyl; and
V is selected from the group consisting of-Q'-Q2, wherein Q1 is absent, C(O), N(H), N(Ci.4-alkyl), C=N(CN), C=N(SO2CH3), or C=N-COH, and Q2 is H, Ci-4-alkyl, C=N-COH- Ci-4-alkyl, Ci-4-alkoxy, C3-7cycloalkyloxy, heterocycloalkyloxy, NH2, N(H)-C i^-alkyl, N(Q- 4-alkyl)2, Sθ2-aryl, SO2-C 1-4-alkyl, C3-6cycloalkyl-Co-4-alkyl, aryl, heteroaryl and heterocycle, each of which may be independently substituted one or more times with a halogen atom, Ci-4- alkyl, Ci^alkoxy, C2-C4alkenyloxy, C2-C4alkynyloxy, Ci-4-alkyl substituted by one or more halogen atoms, or C3-6-cycloalkyl; or when x is 0, Rio and V can form a cyclopropyl ring that may be further substituted by an amide group.
12. The compound of claim 10, wherein X is CR5R53, R4 is H, and R5 and Rsa taken in combination form a 3 to 6 member spirocyclic carbocycle substituted with 0-2 substitutents selected from halogen, Ci-6-alkyl, C2-6-alkenyl, C2-6-alkynyl, Ci-6-alkoxide, C3-7- cycloalkyl-Co-4-alkyl, phenyl-Co-4-alkyl, naphthyl-Co-4-alkyl, heteroaryl-Co-4-alkyl, or two substitutents taken together form a fused or spirocyclic 3 to 7 membered carbocyclic ring, each of which is substituted with 0-3 independently selected halogen atoms or Ci^-alkyl groups.
13. The compound of claim 10, wherein V is R20 or C(O)-R20, wherein R20 is selected from the group consisting of C3-6-cycloalkyl, mono- and
Figure imgf000378_0001
phenyl, pyrazine, benzooxazole, 4,4-dimethyl-4,5-dihydro-oxazole, benzoimidazole, pyrimidine, benzothiazole 1,1 -dioxide and quinazoline, each of which may be further independently substituted with a halogen atom, CF3, C^-alkyl, Ci-4alkoxy, C2-C4alkenyloxy, C2- C4alkynyloxy, or C3-6-cycloalkyl.
14. The compound of claim 10, wherein V is hydrogen or selected from R20 or C(O)R20, wherein R20 is selected from the group consisting of
Figure imgf000378_0002
Figure imgf000379_0001
wherein b is 0, 1, or 2; and Ri8 is selected from the group consisting of hydrogen, a halogen atom, aryl, trihalomethyl, and Ci^-alkyl.
15. The compound of claim 10 according to Formula Ila:
Figure imgf000379_0002
πa wherein
Z2 is nitrogen or CH; k] and k2 are 0 or 1 such that a sum of ki and k2 equals 1 or 2;
Ra is hydrogen, C^alkyl, or phenyl;
Rb is hydrogen,
Figure imgf000379_0003
mono- and
Figure imgf000379_0004
or phenyl or Ra and Rb taken together form a fused or spirocyclic 3 to 6 membered ring having 0, 1 or 2 ring heteroatoms selected from N, O and S, which fused or spirocyclic ring has 0 to 2 independently selected substitutents selected from halogen,
Figure imgf000379_0005
Q- 4alkanoyl, and phenyl; and
Rc represents 0 to 4 substitents which are independently selected at each occurrence of R0 from the group consisting of halogen, Ci^alkyl, and phenyl, or two geminal R0 substitents, taken in combination form a 3 to 6 member spirocyclic ring.
16. The compound of claim 15, wherein the divalent residue:
Figure imgf000380_0001
is selected from the group consisting of:
Figure imgf000380_0002
Figure imgf000381_0001
17. The compound of claim 10, wherein X is CRsRs3; and
R5 and Rs3, taken in combination, form a spirocyclic ring having between 3 and 7 ring atoms and having 0, 1, or 2 ring heteroatoms, which spirocyclic ring is substituted with a spirocyclic 3 to 7 membered ring having 0, 1 or 2 ring heteroatoms selected from N, O and S, and wherein each of the spirocyclic rings has 0 to 2 independently selected substitutents selected from cyano, halogen, hydroxyl, amino, thiol, Ci-8-alkyl, C2-8-alkenyl, C2-8-alkynyl, Ci-8-alkoxy-Co-4alkyl, Ci-8-haloalkyl, C2-8-haloalkenyl, C2-8-haloalkynyl, Ci-8-haloalkoxy, Ci- 8-alkylthio, Ci-8-alkylsulfonyl, Ci-8-alkylsulfoxy, Ci-8-alkanoyl, Ci-8-alkoxycarbonyl, C3-7- cycloalkyl-Co-4-alkyl, aryl-Co-4-alkyl, heteroaryl-Co^-alkyl, COOH, C(O)NH2, mono- and di- CM-alkyl-carboxamide, mono- and di-Ci-4-alkyl-amino-Co-.ialkyl, SO3H, SO2NH2, and mono-and di-CM-alkylsulfonamide.
18. The compound of claim 10 according to Formula lib:
lib
Z2 is nitrogen or CH; ki and k2 are 0 or 1 such that a sum of Ic1 and k2 equals 1 or 2;
R3 and R^ taken together form a spirocyclic 3 to 6 membered ring having 0, 1 or 2 ring heteroatoms selected from N, O and S, which fused or spirocyclic ring has 0 to 2 independently selected substituents selected from halogen, Ci^alkyl, C^alkoxy, Ci- 4alkanoyl, and phenyl;
Rc represents 0 to 2 substituents which are independently selected at each occurrence OfR0 from the group consisting of halogen, Ci^alkyl, and phenyl, or two geminal R0 substitents, taken in combination form a 3 to 6 member spirocyclic ring;
R4 represents 0, 1, or 2 substituents each of which is independently selected from H and Q-4- alkyl; and R<s is hydrogen or Ci^alkyl.
19. The compound of claim 18, wherein the divalent residue:
Figure imgf000383_0001
cted from the group consisting of:
Figure imgf000383_0002
20. A compound of claim 1 , wherein the compound is a compound of formula III:
Figure imgf000384_0001
III and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereofs.
21. The compound of claim 20, wherein:
Z2 is nitrogen or CH; Z1 is absent or NRi0;
R3 is selected from the group consisting of H, Q-4-alkyl, and C3-6-cycloalkylCo- C4alkyl;
Rn, R15 and R22 are selected from the group consisting of H, alkyl-aryl, Ci-4-alkyl, O- Ci-4-alkyl, N(H)-C i-4-alkyl, and C3-6-cycloalkylC0-C4alkyl;
Rio and R]7 are each, independently, selected from the group consisting of H, Ci-4- alkyl and (CH2)0-4-C3-6-cycloalkyl; or R]5 and Ri6 may together form a 3, 4, 5, 6 or 7-membered ring that may comprise between 0 to 3 additional heteroatoms, wherein the ring may be further substituted with 0-5 substitutents; or
Ri6 and Rj7 may together form a 3, 4, 5, 6 or 7-membered ring that may comprise between 0 to 3 additional heteroatoms, wherein the ring may be further substituted with 0-5 substitutents; and
V is selected from the group consisting of-Ql-Q2, wherein Q1 is absent, C(O), N(H), N(CM-alkyl), C=N(CN), C=N(SO2CH3), or C=N-COH, and Q2 is H, CM-alkyl, C=N-COH- Ci^-alkyl, Ci-4-alkoxy, C3-7cycloalkyloxy, heterocycloalkyloxy, NH2, N(H)-C i-4-alkyl, N(Q- 4-alkyl)2, SO2-aryl, SO2-C 1-4-alkyl, C3-6cycloalkyl-Co-4-alkyl, aryl, heteroaryl and heterocycle, each of which may be independently substituted one or more times with a halogen atom, Ci-4- alkyl, Ci^alkoxy, C2-C4alkenyloxy, C2-C4alkynyloxy, Ci-4-alkyl substituted by one or more halogen atoms, or C3-6-CyClOaIlCyI.
22. The compound of claim 20, wherein
R3 is selected from the group consisting of H and Ci-4-alkyl; R13 is H; Rio and Rn are each, independently, selected from the group consisting of H, Ci-4- alkyl, and C3-7cycloalkylC0-4alkyl;
R9 and Ri2 are each, independently, selected from the group consisting of H, Ci-4- alkyl and (CH2)o-4-C3-6-cycloalkyl; and
V is selected from the group consisting of-Q'-Q2, wherein Q1 is absent, C(O), N(H), N(Ci-4-alkyl), C=N(CN), C=N(SO2CH3), or C=N-COH, and Q2 is H, C^-alkyl, C=N-COH- Ci-4-alkyl, C^-alkoxy, C3-7cycloalkyloxy, heterocycloalkyloxy, NH2, N(H)-C i-4-alkyl, N(Ci- 4-alkyl)2, SO2-aryl, SO2-C 1-4-alkyl, C^cycloalkyl-QM-alkyl, aryl, heteroaryl and heterocycle, each of which may be independently substituted one or more times with a halogen atom, Ci-4- alkyl, Ci^alkoxy, C2-C4alkenyloxy, C2-C4alkynyloxy, C1-4-alkyl substituted by one or more halogen atoms, or C3-6-cycloalkyl.
23. The compound of claim 20, wherein V is C(O)-R20, wherein R2o is selected from the group consisting of tert-butyl, C3-6-cycloalkyl, phenyl, pyrazine, benzooxazole, 4,4- dimethyl-4,5-dihydro-oxazole, benzoimidazole, pyrimidine, thiazole, benzothiazole, benzothiazole 1,1 -dioxide and quinazoline, each of which may be further independently substituted with 0-5 substitutents selected from halogen atom, CF3, Ci-4-alkyl or C3-6- cycloalkyl.
24. The compound of claim 20, wherein V is selected from the group consisting of C3-6-cycloalkyl, phenyl, pyrazine, benzooxazole, 4,4-dimethyl-4,5-dihydro-oxazole, benzoimidazole, pyrimidine, thiazole, benzothiazole,benzothiazole 1,1 -dioxide and quinazoline, all of which may be further independently substituted with a halogen atom, CF3, Ci-4-alkyl , Q^alkoxy, C2-C4alkenyloxy, C2-C4alkynyloxy, or C^-cycloalkyl.
25. A compound of claim 1, wherein the compound is a compound of formula FV:
Figure imgf000386_0001
IV and pharmaceutically acceptable salts, enantiomers, stereoisomers, rotamers, tautomers, diastereomers, or racemates thereof.
26. The compound of claim 25, wherein
Z2 is nitrogen or CH;
R3 is selected from the group consisting of H and C^-alkyl; Rn is selected from hydrogen or the group consisting of C1-4-alkyl, Q-6-cycloalkyl, (CH2)o-4-C3-6-cycloalkyl, aryl, alkyl-aryl and heterocycle, each of which may be independently substituted one or more times;
Rio and Rn are each, independently, selected from the group consisting of H and C1-4- alkyl;
Ri2 is selected from the group consisting of H, C^-alkyl, Ci-6-cycloalkyl and aryl; and V is selected from the group consisting of-Q'-Q2, wherein Q1 is absent, C(O), N(H),
Ntd^-alkyl), C=N(CN), C=N(SO2CH3), or C=N-COH, and Q2 is H, C1-4-alkyl, C=N-COH- CM-alkyl, O-C1-4-alkyl, NH2, N(H)-C 1-4-alkyl, N(CI-4-alkyl)2, SO2-aryl, SO2-C M-alkyl, C3-6- cycloalkyl-Co-4-alkyl, aryl, heteroaryl and heterocycle, each of which may be independently substituted one or more times with a halogen atom, Ci-4-alkyl, Ci^-alkyl substituted by one or more halogen atoms, or C3-6-cycloalkyl; or Ri 1 and V form the following 5-membered ring which may be further substituted:
Figure imgf000386_0002
Ri3
27. The compound of claim 25, wherein Ri7 is selected from the group consisting of H, cyclopropylCo-C2alkyl, cyclopentylCo-C2alkyl, phenylCi-C2alkyl, and naphthylCi- C2alkyl.
28. The compound of claim 25, wherein V is C(O)-N(H)-M>utyl or C(O)-R20, wherein R20 is selected from the group consisting of C3.6-cycloalkyl, phenyl, pyrazine, benzooxazole, 4,4-dimethyl-4,5-dihydro-oxazole, benzoimidazole, pyrimidine, thiazole, benzothiazole, benzothiazole 1,1 -dioxide and quinazoline, all of which may be further independently substituted with a halogen atom, CF3, C1-4-alkyl,
Figure imgf000387_0001
C2-C4alkenyloxy, C2-C4alkynyloxy, or C3-6-cycloalkyl.
29. The compound of claim 25, wherein V is selected from the group consisting of C3-6-cycloalkyl, phenyl, pyrazine, benzooxazole, 4,4-dimethyl-4,5-dihydro-oxazole, benzoimidazole, pyrimidine, thiazole, benzothiazole,benzothiazole 1,1 -dioxide and quinazoline, all of which may be further independently substituted with a halogen atom, CF3, Ci-4-alkyl , C^alkoxy, C2-C4alkenyloxy, C2-C4alkynyloxy, or C3-6-cycloalkyl.
30. The compound of claim 1, wherein V is R2o or C(O)-R2O, wherein R2o is a residue of the formula:
Figure imgf000387_0002
wherein
Zg is absent or selected from NR33 or oxygen; g and f are independently selected integers selected from the group consisting of 0, 1, 2, 3 and 4; j is an integer selected from the group consisting of 1, 2, 3 and 4, wherein the sum off
+ g + j is less than or equal to 5 and greater than or equal to 2 when Z8 is absent and the sum of f + g + jk is less than or equal to 4 and greater than or equal to 1 when Z8 is oxygen;
R33 is independently selected at each occurrence from the group consisting of hydrogen,
Figure imgf000387_0003
and
R34 represents zero to three residues each independently selected at each occurrence from the group consisting of halogen, hydroxy, amino, Ci^alkyl, C3-6cycloalkyl, C1-4alkoxy,
Figure imgf000387_0004
31. The compound of claim 1, wherein V is R2o or C(O)-R20, wherein R20 is a residue of the formula:
Figure imgf000388_0001
wherein g is an integer selected from the group consisting of 0, 1, 2, 3 and 4; j is an integer selected from the group consisting of 1, 2, 3 and 4, wherein the sum of g + j is less than or equal to 5 and greater than or equal to 2;
R33 is independently selected at each occurrence from the group consisting of hydrogen,
Figure imgf000388_0002
C3-6cycloalkyl, hydroxyC1-4alkyl, and C1-4alkoxyCi-4alkyl; and
R34 represents zero to three residues each independently selected at each occurrence from the group consisting of halogen, hydroxy, amino, C^alkyl, Ca^cycloalkyl, C^alkoxy, mono-and di-CMalkylamino, hydroxyCi^alkyl, and Ci^alkoxyCi^alkyl.
32. A pharmaceutical composition comprising at least one compound according to any one of claims 1-31 and a pharmaceutically acceptable carrier.
33. The pharmaceutical composition of claim 32, wherein the composition further comprises at least one additional HCV-modulating compound.
34. The pharmaceutical composition of claim 32, wherein the additional HCV- modulating compound is selected from the group consisting of Sch 503034 and VX-950.
35. The pharmaceutical composition of claim 32, wherein the additional HCV- modulating compound is interferon or derivatized interferon.
36. The pharmaceutical composition of claim 32, wherein the interferon is selected from the group consisting of interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A, lymphoblastoid interferon, and interferon tau; and said compound having anti-hepatitis C virus activity is selected from the group consisting of interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, double stranded RNA, double stranded RNA complexed with tobramycin, Imiquimod, ribavirin, an inosine 5 '-monophosphate dehydrogenase inhibitor, amantadine, and rimantadine.
37. The pharmaceutical composition of claim 32, wherein the additional HCV- modulating compound is a cytochrome P450 monooxygenase inhibitor.
38. The pharmaceutical composition of claim 37, wherein the cytochrome P450 inhibitor is selected from the group consisting of ritonavir, ketoconazole, troleandomycin, 4- methyl pyrazole, cyclosporin, and clomethiazole.
39. A method of treating an HCV-associated disorder comprising administering to a subject in need thereof a pharmaceutically acceptable amount of a compound according to any one of claims 1-31.
40. The method of claim 39, wherein the HCV-associated disorder is selected from the group consisting of HCV infection, liver cirrhosis, chronic liver disease, hepatocellular carcinoma, cryoglobulinaemia, non-Hodgkin's lymphoma, and a suppressed innate intracellular immune response.
41. A method of treating an HIV infection comprising administering to a subject in need thereof a pharmaceutically acceptable amount of a compound according to any one of claims 1-31.
42. A method of treating, inhibiting or preventing the activity of HCV in a subject in need thereof, comprising administering to the subject a pharmaceutically acceptable amount of a compound according to any one of claims 1-31.
43. A, method of inhibiting the activity of a serine protease, comprising the step of contacting said serine protease with a compound according to any one of claims 1-31.
44. The method of claim 43, wherein the activity of the NS2 protease is inhibited.
45. The method of claim 43, wherein the activity of the NS3 protease is inhibited.
46. The method of claim 43, wherein the activity of the NS3 helicase is inhibited.
47. The method of claim 43, wherein the activity of the NS5a protein is inhibited.
48. The method of claim 43, wherein the activity of the NS5b polymerase is inhibited.
49. The method of claim 43, wherein the interaction between the NS3 protease and NS4A cofactor is disrupted.
50. The method of claim 43, wherein the severing of one or more of the NS4A- NS4B, NS4B-NS5A and NS5A-NS5B junctions of the HCV is prevented or altered.
51. The method of any one of claims 43-50, wherein an HCV-associated disorder is treated in a subject in need thereof.
52. The method of claim 51, wherein the HCV-associated disorder is selected from the group consisting of HCV infection, liver cirrhosis, chronic liver disease, hepatocellular carcinoma, cryoglobulinaemia, non-Hodgkin's lymphoma, and a suppressed innate intracellular immune response.
53. A method of treating, inhibiting or preventing the activity of HCV in a subject in need thereof, comprising administering to the subject a pharmaceutically acceptable amount of a compound according to any one of claims 1-31, wherein the compound interacts with any target in the HCV life cycle.
54. The method of claim 53, wherein the target is selected from the group consisting of NS2 protease, NS3 protease, NS3 helicase, NS5a protein andNS5b polymerase.
55. A method of decreasing the HCV RNA load in a subject in need thereof comprising administering to the subject a pharmaceutically acceptable amount of a compound according to any one of claims 1-31, such that the HCV RNA load in the subject is decreased.
56. A method of treating an HCV-associated disorder in a subject, comprising administering to a subject in need thereof a pharmaceutically acceptable amount of a compound according to any one of claims 1-31, and a pharmaceutically acceptable carrier, such that the HCV-associated disorder is treated.
57. A method of treating an HCV-associated disorder comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound according to any one of claims 1-31, in combination with a pharmaceutically effective amount of an additional HCV-modulating compound, such that the HCV-associated disorder is treated.
58. The method of claim 57, wherein the additional HCV-modulating compound is selected from the group consisting of ITMNl 91 , Sch 503034 and VX-950.
59. The method of claim 57, wherein the additional HCV-modulating compound is interferon or derivatized interferon.
60. The method of claim 59, wherein the interferon is selected from the group consisting of interferon alpha 2B, pegylated interferon alpha, albufuron, consensus interferon, interferon alpha 2A, lymphoblastoid interferon, and interferon tau; and said compound having anti-hepatitis C virus activity is selected from the group consisting of interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, double stranded RNA, double stranded RNA complexed with tobramycin, Imiquimod, ribavirin, an inosine 5 '-monophosphate dehydrogenase inhibitor, amantadine, and rimantadine.
61. The method of claim 57, wherein the additional HCV-modulating compound is a cytochrome P450 monooxygenase inhibitor.
62. The method of claim 61, wherein the cytochrome P450 inhibitor is selected from the group consisting of ritonavir, ketoconazole, troleandomycin, 4-methyl pyrazole, cyclosporin, and clomethiazole.
63. The method of claim 57, wherein the HCV-associated disorder is selected from the group consisting of HCV infection, liver cirrhosis, chronic liver disease, hepatocellular carcinoma, cryoglobulinemia, non-Hodgkin's lymphoma, and a suppressed innate intracellular immune response.
64. A method of inhibiting hepatitis C virus replication in a cell, comprising contacting said cell with a compound according to any one of claims 1-31.
65. A packaged HCV-associated disorder treatment, comprising an HCV- modulating compound according to any one of claims 1-31, packaged with instructions for using an effective amount of the HCV-modulating compound to treat an HCV-associated disorder.
66. The treatment of claim 65, wherein the HCV-associated disorder is selected from the group consisting of HCV infection, liver cirrhosis, chronic liver disease, hepatocellular carcinoma, cryoglobulinaemia, non-Hodgkin's lymphoma, and a suppressed innate intracellular immune response.
67. A method of treating HCV infection, liver cirrhosis, chronic liver disease, hepatocellular carcinoma, cryoglobulinaemia, non-Hodgkin's lymphoma, and/or a suppressed innate intracellular immune response in subject in need thereof comprising administering to the subject a pharmaceutically acceptable amount of a compound according to any one of claims 1-31.
68. The method of claim 39, wherein the HCV is selected from any HCV genotype.
69. The method of claim 39, wherein the HCV is selected from HCV genotype 1, 2 and/or 3.
70. A method of preventing liver damage in a liver transplant patient, the method comprising administration of a compound of any one of claims 1-31 to a patient who has received a liver transplant or is scheduled for a liver transplant operation.
PCT/EP2008/001281 2007-02-20 2008-02-19 Macrocyclic compounds as hcv ns3 protease inhibitors WO2008101665A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
KR1020097019520A KR20090111353A (en) 2007-02-20 2008-02-19 Macrocyclic compounds as hcv ns3 protease inhibitors
AU2008217187A AU2008217187A1 (en) 2007-02-20 2008-02-19 Macrocyclic compounds as HCV NS3 protease inhibitors
EP08707770A EP2125757A1 (en) 2007-02-20 2008-02-19 Macrocyclic compounds as hcv ns3 protease inhibitors
MX2009008872A MX2009008872A (en) 2007-02-20 2008-02-19 Macrocyclic compounds as hcv ns3 protease inhibitors.
CN200880012715A CN101663284A (en) 2007-02-20 2008-02-19 Macrocyclic compounds as hcv ns3 protease inhibitors
CA002677843A CA2677843A1 (en) 2007-02-20 2008-02-19 Macrocyclic compounds as hcv ns3 protease inhibitors
EA200901101A EA200901101A1 (en) 2007-02-20 2008-02-19 MACROCYCLIC COMPOUNDS AS INHIBITORS OF HCV PROTEASIS NS3
BRPI0807887-4A2A BRPI0807887A2 (en) 2007-02-20 2008-02-19 MACROCYCLIC COMPOUNDS AS HCV NS3 PROTEASE INHIBITORS.
JP2009550673A JP2010519266A (en) 2007-02-20 2008-02-19 Macrocycles as HCV NS3 protease inhibitors
US12/527,787 US20100240638A1 (en) 2007-02-20 2008-02-19 Organic Compounds and their uses

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US89075407P 2007-02-20 2007-02-20
US60/890,754 2007-02-20

Publications (1)

Publication Number Publication Date
WO2008101665A1 true WO2008101665A1 (en) 2008-08-28

Family

ID=39475541

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/001281 WO2008101665A1 (en) 2007-02-20 2008-02-19 Macrocyclic compounds as hcv ns3 protease inhibitors

Country Status (11)

Country Link
US (1) US20100240638A1 (en)
EP (1) EP2125757A1 (en)
JP (1) JP2010519266A (en)
KR (1) KR20090111353A (en)
CN (1) CN101663284A (en)
AU (1) AU2008217187A1 (en)
BR (1) BRPI0807887A2 (en)
CA (1) CA2677843A1 (en)
EA (1) EA200901101A1 (en)
MX (1) MX2009008872A (en)
WO (1) WO2008101665A1 (en)

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2451184A (en) * 2007-07-17 2009-01-21 Angeletti P Ist Richerche Bio Macrocyclic hepatitis C virus NS3 protease inhibitors having a 6-(N-sulphamoyl(carbamoyl))-[7-aza-]indole moiety within the macrocyclic ring path
WO2011041551A1 (en) * 2009-10-01 2011-04-07 Intermune, Inc. Therapeutic antiviral peptides
WO2011091757A1 (en) 2010-01-27 2011-08-04 AB Pharma Ltd. Polyheterocyclic compounds highly potent as hcv inhibitors
US8232246B2 (en) 2009-06-30 2012-07-31 Abbott Laboratories Anti-viral compounds
EP2489669A1 (en) * 2011-02-18 2012-08-22 Helmholtz-Zentrum für Infektionsforschung GmbH Haprolid and derivatives thereof as inhibitors of HCV
US20130079366A1 (en) * 2010-02-11 2013-03-28 Vanderbilt University Benzisoxazoles and Azabenzisoxazoles as MGLUR4 Allosteric Potentiators, Compositions, and Methods of Treating Neurological Dysfunction
US8420596B2 (en) 2008-09-11 2013-04-16 Abbott Laboratories Macrocyclic hepatitis C serine protease inhibitors
WO2013092893A1 (en) 2011-12-21 2013-06-27 Rottapharm Spa Chemical compounds
WO2013173759A2 (en) * 2012-05-17 2013-11-21 Enanta Pharmaceuticals, Inc. Macrocyclic nucleoside phosphoramidate derivatives
US8691757B2 (en) 2011-06-15 2014-04-08 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8877929B2 (en) 2008-09-04 2014-11-04 Bristol-Myers Squibb Company Process for synthesizing substituted isoquinolines
US8889871B2 (en) 2002-05-20 2014-11-18 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8937041B2 (en) 2010-12-30 2015-01-20 Abbvie, Inc. Macrocyclic hepatitis C serine protease inhibitors
US8951964B2 (en) 2010-12-30 2015-02-10 Abbvie Inc. Phenanthridine macrocyclic hepatitis C serine protease inhibitors
US8957203B2 (en) 2011-05-05 2015-02-17 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8993595B2 (en) 2009-04-08 2015-03-31 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors
US9174977B2 (en) 2012-03-19 2015-11-03 Rottapharm Biotech S.R.L. 2-azabicyclo[4.1.0]heptane derivatives as orexin receptor antagonists for the treatment of certain disorders
US9284307B2 (en) 2009-08-05 2016-03-15 Idenix Pharmaceuticals Llc Macrocyclic serine protease inhibitors
US9333204B2 (en) 2014-01-03 2016-05-10 Abbvie Inc. Solid antiviral dosage forms
US9334279B2 (en) 2012-11-02 2016-05-10 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9353100B2 (en) 2011-02-10 2016-05-31 Idenix Pharmaceuticals Llc Macrocyclic serine protease inhibitors, pharmaceutical compositions thereof, and their use for treating HCV infections
US9409943B2 (en) 2012-11-05 2016-08-09 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9499550B2 (en) 2012-10-19 2016-11-22 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9580463B2 (en) 2013-03-07 2017-02-28 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9598433B2 (en) 2012-11-02 2017-03-21 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9643999B2 (en) 2012-11-02 2017-05-09 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2017085098A1 (en) 2015-11-19 2017-05-26 Basf Se Substituted oxadiazoles for combating phytopathogenic fungi
WO2018066718A1 (en) * 2016-10-04 2018-04-12 Takeda Pharmaceutical Company Limited Therapeutic compounds
US10201541B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
US10968249B2 (en) 2014-11-26 2021-04-06 Enanta Pharmaceuticals, Inc. Bile acid derivatives as FXR/TGR5 agonists and methods of use thereof
WO2024038129A1 (en) * 2022-08-17 2024-02-22 Mironid Limited Compounds and their use as pde4 activators

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012048235A1 (en) * 2010-10-08 2012-04-12 Novartis Ag Vitamin e formulations of sulfamide ns3 inhibitors
RU2625796C2 (en) * 2012-01-11 2017-07-19 Ф. Хоффманн-Ля Рош Аг Macrocyclic amides as protease inhibitors
CN108794489B (en) * 2018-07-18 2019-12-17 香港浸会大学深圳研究院 Derivatization reagent and preparation method and application thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997030080A1 (en) * 1996-02-20 1997-08-21 Ortho Pharmaceutical Corporation Macrocyclic peptides useful in the treatment of thrombin related disorders
WO2005030796A1 (en) * 2003-09-26 2005-04-07 Schering Corporation Macrocyclic inhibitors of hepatitis c virus ns3 serine protease
WO2007009109A2 (en) * 2005-07-14 2007-01-18 Gilead Sciences, Inc. Antiviral compounds
WO2008033389A2 (en) * 2006-09-13 2008-03-20 Novartis Ag Macrocyclic hcv inhibitors and their uses

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997030080A1 (en) * 1996-02-20 1997-08-21 Ortho Pharmaceutical Corporation Macrocyclic peptides useful in the treatment of thrombin related disorders
WO2005030796A1 (en) * 2003-09-26 2005-04-07 Schering Corporation Macrocyclic inhibitors of hepatitis c virus ns3 serine protease
WO2007009109A2 (en) * 2005-07-14 2007-01-18 Gilead Sciences, Inc. Antiviral compounds
WO2008033389A2 (en) * 2006-09-13 2008-03-20 Novartis Ag Macrocyclic hcv inhibitors and their uses

Cited By (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9636375B2 (en) 2002-05-20 2017-05-02 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8889871B2 (en) 2002-05-20 2014-11-18 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9227940B2 (en) 2002-05-20 2016-01-05 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
GB2451184A (en) * 2007-07-17 2009-01-21 Angeletti P Ist Richerche Bio Macrocyclic hepatitis C virus NS3 protease inhibitors having a 6-(N-sulphamoyl(carbamoyl))-[7-aza-]indole moiety within the macrocyclic ring path
GB2451184B (en) * 2007-07-17 2011-09-21 Angeletti P Ist Richerche Bio Novel anti-viral macrocyclic indole compounds
US7989438B2 (en) 2007-07-17 2011-08-02 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Therapeutic compounds
US8877929B2 (en) 2008-09-04 2014-11-04 Bristol-Myers Squibb Company Process for synthesizing substituted isoquinolines
US9309279B2 (en) 2008-09-11 2016-04-12 Abbvie Inc. Macrocyclic hepatitis C serine protease inhibitors
US8420596B2 (en) 2008-09-11 2013-04-16 Abbott Laboratories Macrocyclic hepatitis C serine protease inhibitors
US8642538B2 (en) 2008-09-11 2014-02-04 Abbvie, Inc. Macrocyclic hepatitis C serine protease inhibitors
US8993595B2 (en) 2009-04-08 2015-03-31 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors
US8232246B2 (en) 2009-06-30 2012-07-31 Abbott Laboratories Anti-viral compounds
US9284307B2 (en) 2009-08-05 2016-03-15 Idenix Pharmaceuticals Llc Macrocyclic serine protease inhibitors
WO2011041551A1 (en) * 2009-10-01 2011-04-07 Intermune, Inc. Therapeutic antiviral peptides
EP2528922A4 (en) * 2010-01-27 2013-10-16 Pharma Ltd Ab Polyheterocyclic compounds highly potent as hcv inhibitors
JP2013518067A (en) * 2010-01-27 2013-05-20 エービー・ファーマ・リミテッド Polyheterocyclic compounds as hepatitis C virus inhibitors
EP2528922A1 (en) * 2010-01-27 2012-12-05 AB Pharma Ltd Polyheterocyclic compounds highly potent as hcv inhibitors
WO2011091757A1 (en) 2010-01-27 2011-08-04 AB Pharma Ltd. Polyheterocyclic compounds highly potent as hcv inhibitors
KR101781789B1 (en) * 2010-01-27 2017-09-26 에이비 파르마 리미티드. Polyheterocyclic compounds highly potent as hcv inhibitors
US20130079366A1 (en) * 2010-02-11 2013-03-28 Vanderbilt University Benzisoxazoles and Azabenzisoxazoles as MGLUR4 Allosteric Potentiators, Compositions, and Methods of Treating Neurological Dysfunction
US8916584B2 (en) * 2010-02-11 2014-12-23 Vanderbilt University Benzisoxazoles and azabenzisoxazoles as MGLUR4 allosteric potentiators, compositions, and methods of treating neurological dysfunction
US8937041B2 (en) 2010-12-30 2015-01-20 Abbvie, Inc. Macrocyclic hepatitis C serine protease inhibitors
US8951964B2 (en) 2010-12-30 2015-02-10 Abbvie Inc. Phenanthridine macrocyclic hepatitis C serine protease inhibitors
US9353100B2 (en) 2011-02-10 2016-05-31 Idenix Pharmaceuticals Llc Macrocyclic serine protease inhibitors, pharmaceutical compositions thereof, and their use for treating HCV infections
WO2012110249A1 (en) * 2011-02-18 2012-08-23 Helmholtz-Zentrum für Infektionsforschung GmbH Haprolid and derivatives thereof as inhibitors of hcv
EP2489669A1 (en) * 2011-02-18 2012-08-22 Helmholtz-Zentrum für Infektionsforschung GmbH Haprolid and derivatives thereof as inhibitors of HCV
US8957203B2 (en) 2011-05-05 2015-02-17 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9527885B2 (en) 2011-05-05 2016-12-27 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US10201541B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
US10201584B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
US8691757B2 (en) 2011-06-15 2014-04-08 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2013092893A1 (en) 2011-12-21 2013-06-27 Rottapharm Spa Chemical compounds
US9174977B2 (en) 2012-03-19 2015-11-03 Rottapharm Biotech S.R.L. 2-azabicyclo[4.1.0]heptane derivatives as orexin receptor antagonists for the treatment of certain disorders
WO2013173759A2 (en) * 2012-05-17 2013-11-21 Enanta Pharmaceuticals, Inc. Macrocyclic nucleoside phosphoramidate derivatives
WO2013173759A3 (en) * 2012-05-17 2014-03-06 Enanta Pharmaceuticals, Inc. Macrocyclic nucleoside phosphoramidate derivatives
US9499550B2 (en) 2012-10-19 2016-11-22 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9598433B2 (en) 2012-11-02 2017-03-21 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9643999B2 (en) 2012-11-02 2017-05-09 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9334279B2 (en) 2012-11-02 2016-05-10 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9409943B2 (en) 2012-11-05 2016-08-09 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9580463B2 (en) 2013-03-07 2017-02-28 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9333204B2 (en) 2014-01-03 2016-05-10 Abbvie Inc. Solid antiviral dosage forms
US9744170B2 (en) 2014-01-03 2017-08-29 Abbvie Inc. Solid antiviral dosage forms
US10105365B2 (en) 2014-01-03 2018-10-23 Abbvie Inc. Solid antiviral dosage forms
US10968249B2 (en) 2014-11-26 2021-04-06 Enanta Pharmaceuticals, Inc. Bile acid derivatives as FXR/TGR5 agonists and methods of use thereof
WO2017085098A1 (en) 2015-11-19 2017-05-26 Basf Se Substituted oxadiazoles for combating phytopathogenic fungi
WO2018066718A1 (en) * 2016-10-04 2018-04-12 Takeda Pharmaceutical Company Limited Therapeutic compounds
WO2024038129A1 (en) * 2022-08-17 2024-02-22 Mironid Limited Compounds and their use as pde4 activators

Also Published As

Publication number Publication date
CA2677843A1 (en) 2008-08-28
EA200901101A1 (en) 2010-04-30
EP2125757A1 (en) 2009-12-02
AU2008217187A1 (en) 2008-08-28
KR20090111353A (en) 2009-10-26
JP2010519266A (en) 2010-06-03
MX2009008872A (en) 2009-10-30
CN101663284A (en) 2010-03-03
US20100240638A1 (en) 2010-09-23
BRPI0807887A2 (en) 2014-06-17

Similar Documents

Publication Publication Date Title
EP2125757A1 (en) Macrocyclic compounds as hcv ns3 protease inhibitors
ES2543840T3 (en) Spirocyclic HCV / HIV inhibitors and their uses
AU2010233472B2 (en) Organic compounds and their uses
JP5095824B2 (en) Spiropyrrolidines and their use against HCV and HIV infection
EP2007787A2 (en) Hcv inhibitors comprising beta amino acids and their uses
EP2007788A2 (en) Hcv inhibitors
WO2008033389A2 (en) Macrocyclic hcv inhibitors and their uses
US20080045530A1 (en) Organic Compounds and Their Uses
KR20080111127A (en) Hcv inhibitors comprising beta amino acids and their uses
AU2013231160A1 (en) Organic compounds and their uses

Legal Events

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

Ref document number: 200880012715.1

Country of ref document: CN

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

Ref document number: 08707770

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2008217187

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2677843

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2009550673

Country of ref document: JP

Ref document number: MX/A/2009/008872

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2008217187

Country of ref document: AU

Date of ref document: 20080219

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 5864/DELNP/2009

Country of ref document: IN

Ref document number: 2008707770

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1020097019520

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 200901101

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 12527787

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0807887

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20090819