WO2009053828A2 - P3 hydroxyamino macrocyclic hepatitis c serine protease inhibitors - Google Patents

P3 hydroxyamino macrocyclic hepatitis c serine protease inhibitors Download PDF

Info

Publication number
WO2009053828A2
WO2009053828A2 PCT/IB2008/002839 IB2008002839W WO2009053828A2 WO 2009053828 A2 WO2009053828 A2 WO 2009053828A2 IB 2008002839 W IB2008002839 W IB 2008002839W WO 2009053828 A2 WO2009053828 A2 WO 2009053828A2
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
compound
aryl
heteroaryl
cycloalkyl
Prior art date
Application number
PCT/IB2008/002839
Other languages
French (fr)
Other versions
WO2009053828A3 (en
Inventor
Deqiang Niu
Dong Liu
Yonghua Gai
Yat Sun Or
Zhe Wang
Original Assignee
Enanta Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Enanta Pharmaceuticals, Inc. filed Critical Enanta Pharmaceuticals, Inc.
Publication of WO2009053828A2 publication Critical patent/WO2009053828A2/en
Publication of WO2009053828A3 publication Critical patent/WO2009053828A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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

Definitions

  • the present invention relates to novel macrocycles having activity against the hepatitis C virus (HCV) and useful in the treatment of HCV infections. More particularly, the invention relates to N-hydroxyl macrocyclic compounds, compositions containing such compounds and methods for using the same, as well as processes for making such compounds.
  • HCV hepatitis C virus
  • HCV is the principal cause of non-A, non-B hepatitis and is an increasingly severe public health problem both in the developed and developing world. It is estimated that the virus infects over 200 million people worldwide, surpassing the number of individuals infected with the human immunodeficiency virus (HIV) by nearly five fold. HCV infected patients, due to the high percentage of individuals inflicted with chronic infections, are at an elevated risk of developing cirrhosis of the liver, subsequent hepatocellular carcinoma and terminal liver disease. HCV is the most prevalent cause of hepatocellular cancer and cause of patients requiring liver transplantations in the western world.
  • HIV human immunodeficiency virus
  • anti-HCV therapeutics There are considerable barriers to the development of anti-HCV therapeutics, which include, but are not limited to, the persistence of the virus, the genetic diversity of the virus during replication in the host, the high incident rate of the virus developing drug -resistant mutants, and the lack of reproducible infectious culture systems and small-animal models for HCV replication and pathogenesis. In a majority of cases, given the mild course of the infection and the complex biology of the liver, careful consideration must be given to antiviral drugs, which are likely to have significant side effects.
  • NS3 hepatitis C non- structural protein-3
  • HCV is a flaviridae type RNA virus.
  • the HCV genome is enveloped and contains a single strand RNA molecule composed of circa 9600 base pairs. It encodes a polypeptide comprised of approximately 3010 amino acids.
  • the HCV polyprotein is processed by viral and host peptidase into 10 discreet peptides which serve a variety of functions. There are three structural proteins, C, El and E2.
  • the P7 protein is of unknown function and is comprised of a highly variable sequence.
  • NS2 is a zinc-dependent metalloproteinase that functions in conjunction with a portion of the NS3 protein.
  • NS3 incorporates two catalytic functions (separate from its association with NS2): a serine protease at the N-terminal end, which requires NS4A as a cofactor, and an ATP-ase-dependent helicase function at the carboxyl terminus.
  • NS4A is a tightly associated but non-covalent cofactor of the serine protease.
  • the NS3-NS4A protease is responsible for cleaving four sites on the viral polyprotein.
  • the NS3-NS4A cleavage is autocatalytic, occurring in cis.
  • the remaining three hydrolyses, NS4A-NS4B, NS4B-NS5A and NS5A-NS5B all occur in trans.
  • NS3 is a serine protease which is structurally classified as a chymotrypsin-like protease. While the NS serine protease possesses proteolytic activity by itself, the HCV protease enzyme is not an efficient enzyme in terms of catalyzing polyprotein cleavage. It has been shown that a central hydrophobic region of the NS4A protein is required for this enhancement. The complex formation of the NS3 protein with NS4A seems necessary to the processing events, enhancing the proteolytic efficacy at all of the sites.
  • a general strategy for the development of antiviral agents is to inactivate virally encoded enzymes, including NS3, that are essential for the replication of the virus.
  • Current efforts directed toward the discovery of NS3 protease inhibitors were reviewed by S. Tan, A. Pause, Y. Shi, N. Sonenberg, Hepatitis C Therapeutics: Current Status and Emerging Strategies, Nature Rev. Drug Discov., 1, 867-881 (2002).
  • Other patent disclosures describing the synthesis of HCV protease inhibitors are: WO 2006/007700; US 2005/0261200; WO 2004/113365; WO 03/099274 (2003); US 2003/0008828;
  • the present invention relates to novel N-hydroxyl macrocyclic compounds and pharmaceutically acceptable salts, esters or prodrugs thereof, methods of using the same to treat hepatitis C infection in a subject in need of such therapy .
  • Macrocyclic compounds of the present invention interfere with the life cycle of the hepatitis C virus and are also useful as antiviral agents.
  • the present invention further relates to pharmaceutical compositions comprising the aforementioned compounds, salts, esters or prodrugs for administration to a subject suffering from HCV infection.
  • the present invention further features pharmaceutical compositions comprising a compound of the present invention (or a pharmaceutically acceptable salt, ester or prodrug thereof) and another anti-HCV agent, such as interferon (e.g., alpha-interferon, beta-interferon, consensus interferon, pegylated interferon, or albumin or other conjugated interferon), ribavirin, amantadine, another HCV protease inhibitor, or an HCV polymerase, helicase or internal ribosome entry site inhibitor.
  • interferon e.g., alpha-interferon, beta-interferon, consensus interferon, pegylated interferon, or albumin or other conjugated interferon
  • ribavirin e.g., amantadine
  • another HCV protease inhibitor e.g., alpha-interferon, beta-interferon, consensus interferon, pegylated interferon, or albumin or other conjugated interferon
  • the present invention further relates to pharmaceutical compositions comprising the compounds of the present invention, or pharmaceutically acceptable salts, esters, or prodrugs thereof, in combination with a pharmaceutically acceptable carrier or excipient.
  • a pharmaceutically acceptable carrier or excipient in one embodiment of the present invention there are disclosed compounds represented by Formulas I, or pharmaceutically acceptable salts, esters, or prodrugs thereof:
  • each Ri is independently selected from the group consisting of: (i) aryl; substituted aryl; heteroaryl; substituted heteroaryl;
  • heterocycloalkyl or substituted heterocycloalkyl (ii) heterocycloalkyl or substituted heterocycloalkyl; (iii) -Ci-C 8 alkyl, -C 2 -C 8 alkenyl, or -C 2 -C 8 alkynyl containing O, 1, 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-C 8 alkyl, substituted -C 2 -C 8 alkenyl, or substituted -C 2 -C 8 alkynyl containing O, 1 , 2, or 3 heteroatoms selected from O, S or N; -C 3 -C) 2 cycloalkyl, or substituted -C 3 -
  • Ci 2 cycloalkyl -C 3 -Ci 2 cycloalkenyl, or substituted -C 3 -Ci 2 cycloalkenyl;
  • Each R 2 is independently selected from the group consisting of: (i) hydrogen; (ii) aryl; substituted aryl; heteroaryl; substituted heteroaryl;
  • heterocycloalkyl or substituted heterocycloalkyl (iii) heterocycloalkyl or substituted heterocycloalkyl; (iv) -Ci-C 8 alkyl, -C 2 -C 8 alkenyl, or -C 2 -C 8 alkynyl containing O, 1 , 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-C 8 alkyl, substituted -C 2 -C 8 alkenyl, or substituted -C 2 -C 8 alkynyl containing O, 1 , 2, or 3 heteroatoms selected from O, S or N; -C 3 -Ci 2 cycloalkyl, or substituted -C 3 -
  • Ci 2 cycloalkyl -C 3 -Ci 2 cycloalkenyl, or substituted -C 3 -Ci 2 cycloalkenyl;
  • G is selected from the group consisting of -OH, -NH-S(O) 2 -R 3 , -NH-S(O) 2 NR 4 R 5 ;
  • Each R 3 is independently selected from the group consisting of :
  • heterocycloalkyl or substituted heterocycloalkyl (ii) heterocycloalkyl or substituted heterocycloalkyl; (iii) -C)-C 8 alkyl, -C 2 -C 8 alkenyl, or -C 2 -C 8 alkynyl containing O, 1 , 2, or 3 heteroatoms selected from O, S or N, substituted -C 1 -C 8 alkyl, substituted -C 2 -C 8 alkenyl, or substituted -C 2 -C 8 alkynyl containing O, 1 , 2, or 3 heteroatoms selected from O, S or N; -C 3 -Ci 2 cycloalkyl, or substituted -C 3 - C 12 cycloalkyl; -C 3 -C 12 cycloalkenyl, or substituted -C 3 -C )2 cycloalkenyl;
  • each R 4 and R 5 are independently selected from the group consisting of :
  • heterocycloalkyl or substituted heterocycloalkyl (iii) heterocycloalkyl or substituted heterocycloalkyl; (iv) -Ci-C 8 alkyl, -C 2 -C 8 alkenyl, or -C 2 -C 8 alkynyl containing O, 1 , 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-C 8 alkyl, substituted -C 2 -C 8 alkenyl, or substituted -C 2 -C 8 alkynyl containing O, 1, 2, or 3 heteroatoms selected from O, S or N; -C 3 -Ci 2 cycloalkyl, or substituted -C 3 - Ci 2 cycloalkyl; -C 3 -Cj 2 cycloalkenyl, or substituted -C 3 -Ci 2 cycloalkenyl;
  • L is selected from the group consisting of -CH 2 -, -0-, -S-, or -S(O) 2 -;
  • X is absent or is selected from the group consisting of: (1) oxygen; (2) sulfur;
  • Y is absent or is selected from the group consisting of:
  • Z is selected from the group consisting of aryl, substituted aryl, heteroaryl, substituted heteroaryl, Heterocycloalkyl, substituted heterocycloalkyl;
  • each Zi, Z 2 are independently selected from the group consisting of: i) hydrogen; ii) aryl; iii) substituted aryl; iv) heteroaryl; v) substituted heteroaryl; vi) heterocyclic or substituted heterocyclic; vii)-Ci-C 8 alkyl, -C 2 -C 8 alkenyl, or -C 2 -C 8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S or N; viii) substituted -Ci-C 8 alkyl, substituted -C 2 -C 8 alkenyl, or substituted -C 2 -C 8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S or N; ix) -C 3 -Ci 2 cycloalkyl; x) substituted -C 3 -Ci 2 cycloalkyl; xi)
  • a cyclic moiety selected from the group consisting of : substituted or unsubstituted cycloalkyl, cycloalkenyl, or heterocylic; substituted or unsubstituted cycloalkyl, cycloalkenyl, and heterocyclic fused with one or more aryl, substituted aryl, heteroaryl; substituted heteroaryl, heterocyclic or substituted heterocyclic;
  • a first embodiment of the invention is a compound represented by Formula I as described above, or a pharmaceutically acceptable salt, ester or prodrug thereof, alone or in combination with a pharmaceutically acceptable carrier or excipient.
  • Another embodiment of the invention is a compound represented by Formula II:
  • R 6 is selected from the group consisting of aryl, substituted aryl, heteroaryl, and substituted heteroaryl; J is absent or is selected from the group consisting of O, S, NR 5 , CO, (CO)NR 5 , (CO)O, NR 5 (CO), NH(CO)NH, NR 5 SO 2 ; wherein R 5 are as defined in the first embodiment;
  • Each R 7 i, R 72 , R 73 and R 74 is independently selected from the group consisting of : (i) hydrogen;
  • G can be -NH-SO 2 -NR 4 R 5 or -NHSO 2 - R 3 , where R 3 is selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C 3 -Ci 2 cycloalkyl, -C 3 -Ci 2 cycloalkenyl, substituted -C 3 -Ci 2 cycloalkyl, or substituted -C 3 -Ci 2 cycloalkenyl, and R 4 and R 5 are each independently selected from hydrogen, -Ci-C 8 alkyl, -C 2 -C 8 alkenyl, -C 2 -C 8 alkynyl, substituted -Cj-C 8 alkyl, substituted -C 2 -C 8 alkenyl, substituted -C 2 -C 8 alkynyl, aryl, substituted aryl, heteroaryl, substituted
  • Each R 7J , R 72 , R 73 and R 74 is independently selected from the group consisting of hydrogen, halogen, -NO 2 , -CN, -M-R 4 ; wherein M is absent, or O, S, NH, NR 5 , aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycloalkyl, and substituted heterocycloalkyl, wherein R 4 , R 5 are as defined previously.
  • a and B are independently selected from the group consisting of H, R 1 , where Ri is selected from -Ci-C 8 alkyl, -C 2 -Cg alkenyl, or -C 2 -C 8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-C 8 alkyl, substituted -C 2 -C 8 alkenyl, or substituted -C 2 -C 8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S or N; -C 3 -Ci 2 cycloalkyl, or substituted -C 3 -Ci 2 cycloalkyl; -C 3 -Ci 2 cycloalkenyl, or substituted -C 3 -Cj 2 cycloalkenyl; wherein R 6 is selected from the group consisting of aryl, substituted aryl, heteroaryl, and substituted heteroaryl.
  • G is -NHSO 2 -R 3 , where R 3 is selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C 3 -Ci 2 cycloalkyl, -C 3 -Ci 2 cycloalkenyl, substituted -C 3 -Ci 2 cycloalkyl, or substituted -C 3 -Cj 2 cycloalkenyl.
  • R 7 i, R 72 , R 73 and R 74 is independently selected from the group consisting of hydrogen, halogen, -NO 2 , -CN.
  • A is H.
  • B is selected from the group consisting of -Ci-C 8 alkyl, substituted -Ci-C 8 alkyl, -C 3 -Ci 2 cycloalkyl, or substituted -C 3 -Ci 2 cycloalkyl; wherein R 6 is selected from the group consisting of aryl, substituted aryl, heteroaryl, and substituted heteroaryl. J is absent, j is 1 or 2.
  • G is -NHSO 2 -R 3 , where R 3 is selected from -C 3 -Ci 2 cycloalkyl or substituted -C 3 - Ci 2 cycloalkyl.
  • each R 6 , R 7 i, R 72 , R 73 , R 74 and J are as defined previously in Formulae III; and A, B, G, j are as defined in the first embodiment.
  • R 6 is selected from the group consisting of aryl, substituted aryl, heteroaryl, and substituted heteroaryl. J is absent, j is 1 or 2.
  • G can be -NH-SO 2 -NR 4 R 5 or -NHSO 2 -R 3 , where R 3 is selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C 3 -Ci 2 cycloalkyl, -C 3 -Cj 2 cycloalkenyl, substituted -C 3 -Cj 2 cycloalkyl, or substituted -C 3 -Cj 2 cycloalkenyl, and R 4 and R 5 are each independently selected from hydrogen, -Cj-C 8 alkyl, -C 2 -C 8 alkenyl, -C 2 -C 8 alkynyl, substituted -Cj-C 8 alkyl, substituted -C 2 -C 8 alkeny
  • Each R 7J , R 72 , R 73 and R 74 is independently selected from the group consisting of hydrogen, halogen, -NO 2 , -CN, -M-R 4 , wherein M is absent, or O, S, NH, NR 5 , aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycloalkyl, and substituted heterocycloalkyl, wherein R 4 , R 5 are as defined previously.
  • a and B are independently selected from the group consisting of H, Rj, where Ri is selected from -Cj-C 8 alkyl, -C 2 -C 8 alkenyl, or -C 2 -C 8 alkynyl containing O, 1, 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-C 8 alkyl, substituted -C 2 -C 8 alkenyl, or substituted -C 2 -C 8 alkynyl containing O, 1, 2, or 3 heteroatoms selected from O, S or N; -C 3 -Cj 2 cycloalkyl, or substituted -C 3 -Cj 2 cycloalkyl; -C 3 -Ci 2 cycloalkenyl, or substituted -C 3 -Cj 2 cycloalkenyl.
  • R 6 is selected from the group consisting of aryl, substituted aryl, heteroaryl, and substituted heteroaryl. J is absent, j is 1 or 2.
  • G is -NHSO 2 -R 3 , where R 3 is selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C 3 -Cj 2 cycloalkyl, -C 3 -Ci 2 cycloalkenyl, substituted -C 3 -Cj 2 cycloalkyl, or substituted -C 3 -Cj 2 cycloalkenyl.
  • Each R 71 , R 7 2, R 73 and R 74 is independently selected from the group consisting of hydrogen, halogen, - NO 2 , -CN.
  • A is H.
  • B is selected from the group consisting of -Ci-C 8 alkyl, substituted -Ci-C 8 alkyl, -C 3 -Ci 2 cycloalkyl, or substituted -C 3 -Ci 2 cycloalkyl.
  • R 6 is selected from the group consisting of aryl, substituted aryl, heteroaryl, and substituted heteroaryl.
  • J is absent, j is 1 or 2.
  • G is -NHSO 2 -R 3 , where R 3 is selected from - C 3 -Cj 2 cycloalkyl or substituted -C 3 -Ci 2 cycloalkyl.
  • each R 71 , R 72 , R 73 , R 74 are as defined previously in Formulae III; and A, B, GJ are as defined in the first embodiment.
  • G can be -NH-SO 2 -NR 4 R 5 or -NHSO 2 -R 3 , where R 3 is selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C 3 -Ci 2 cycloalkyl, -C 3 -Ci 2 cycloalkenyl, substituted -C 3 -Ci 2 cycloalkyl, or substituted -C 3 -Ci 2 cycloalkenyl, and R 4 and R 5 are each independently selected from hydrogen, -Ci-C 8 alkyl, -C 2 -C 8 alkenyl, -C 2 - Cg alkynyl, substituted -Ci-C 8 alkyl, substituted -C 2 -C 8 alkenyl, substituted -C 2 -C 8 alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl,
  • Each R 7 i, R 72 , R 73 and R 74 is independently selected from the group consisting of hydrogen, halogen, -NO 2 , -CN, -M-R 4 , wherein M is absent, or O, S, NH, NR 5 , aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycloalkyl, and substituted heterocycloalkyl, wherein R 4 , R 5 are as defined previously.
  • a and B are independently selected from the group consisting of H, Ri, where Ri is selected from -Cj-C 8 alkyl, -C 2 -C 8 alkenyl, or -C 2 -C 8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S, or N; substituted -Cj-C 8 alkyl, substituted -C 2 -C 8 alkenyl, or substituted -C 2 -C 8 alkynyl containing 0, 1 , 2, or 3 heteroatoms selected from O, S or N; -C 3 -Cj 2 cycloalkyl, or substituted -C 3 -Cn cycloalkyl; -C 3 -Cj 2 cycloalkenyl, or substituted -C 3 -Ci 2 cycloalkenyl.
  • G is -NHSO 2 -R 3 , where R 3 is selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C 3 -Ci 2 cycloalkyl, -C 3 -Cj 2 cycloalkenyl, substituted -C 3 -Cj 2 cycloalkyl, or substituted -C 3 -Ci 2 cycloalkenyl.
  • R 7J , R 72 , R 73 and R 74 is independently selected from the group consisting of hydrogen, halogen, -NO 2 , -CN.
  • A is H.
  • B is selected from the group consisting of -Cj-C 8 alkyl, substituted -Cj-C 8 alkyl, -C 3 -Cj 2 cycloalkyl, or substituted -C 3 -Cj 2 cycloalkyl.
  • j is 1 or 2.
  • G is -NHSO 2 -R 3 , where R 3 is selected from -C 3 -Ci 2 cycloalkyl or substituted -C 3 -Cj 2 cycloalkyl.
  • Each R 7J , R 72 , R 73 and R 74 is independently selected from the group consisting of hydrogen, halogen.
  • Zl, Z2 and A, B, G, j are as defined in the first embodiment.
  • Zl and Z2 are independently selected from the group consisting of hydrogen, aryl, substituted aryl, heteroaryl, and substituted heteroaryl, or Z 1 and Z 2 taken together with the carbon atom to which they are attached form a cyclic moiety selected from the group consisting of substituted or unsubstiruted cycloalkyl, cycloalkenyl, or heterocylic, substituted or unsubstituted cycloalkyl, cycloalkenyl, and heterocyclic fused with one or more aryl, substituted aryl, heteroaryl, and substituted heteroaryl.
  • a and B are independently selected from the group consisting of H, Ri, where Ri is selected from -Ci- C 8 alkyl, -C 2 -C 8 alkenyl, or -C 2 -C 8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-Cg alkyl, substituted -C 2 -C 8 alkenyl, or substituted -C 2 -C 8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S or N; -C 3 -Cj 2 cycloalkyl, or substituted -C 3 -C 12 cycloalkyl; -C 3 -Ci 2 cycloalkenyl, or substituted -C 3 -Ci 2 cycloalkenyl.
  • G is -NHSO 2 -R 3 , where R 3 is selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C 3 -Ci 2 cycloalkyl, -C 3 -Ci 2 cycloalkenyl, substituted -C 3 -Ci 2 cycloalkyl, or substituted -C 3 -Ci 2 cycloalkenyl.
  • a and B are selected from the group consisting of hydrogen, -Ci-C 8 alkyl, substituted -Ci-C 8 alkyl, -C 3 -Ci 2 cycloalkyl, or substituted -C 3 -Ci 2 cycloalkyl.
  • j is 1 or 2.
  • G is -NHSO 2 -R 3 , where R 3 is selected from -C 3 - Ci 2 cycloalkyl or substituted -C 3 -Ci 2 cycloalkyl.
  • Representative compounds of the invention include, but are not limited to, the following compounds (Table 1) according to Formula VII, wherein A, B, Q and G are delineated in Table 1 below for each example:
  • the present invention also features pharmaceutical compositions comprising a compound of the present invention, or a pharmaceutically acceptable salt, ester or prodrug thereof.
  • Compounds of the present invention can be administered as the sole active pharmaceutical agent, or used in combination with one or more agents to treat or prevent hepatitis C infections or the symptoms associated with HCV infection.
  • Other agents to be administered in combination with a compound or combination of compounds of the invention include therapies for disease caused by HCV infection that suppresses HCV viral replication by direct or indirect mechanisms. These include agents such as host immune modulators (for example, interferon-alpha, pegylated interferon-alpha, interferon-beta, interferon-gamma, CpG oligonucleotides and the like), or antiviral compounds that inhibit host cellular functions such as inosine monophosphate dehydrogenase (for example, ribavirin and the like).
  • host immune modulators for example, interferon-alpha, pegylated interferon-alpha, interferon-beta, interferon-gamma, CpG oligonucleotides and the like
  • cytokines that modulate immune function.
  • vaccines comprising HCV antigens or antigen adjuvant combinations directed against HCV.
  • agents to be administered in combination with a compound of the present invention include any agent or combination of agents that inhibit the replication of HCV by targeting proteins of the viral genome involved in the viral replication.
  • These agents include but are not limited to other inhibitors of HCV RNA dependent RNA polymerase such as, for example, nucleoside type polymerase inhibitors described in WOOl 90121(A2), or U.S. Pat. No. 6,348,587Bl or WO0160315 or WOO 132153 or non-nucleoside inhibitors such as, for example, benzimidazole polymerase inhibitors described in EP 1162196Al or WO0204425 or inhibitors of HCV protease such as, for example, peptidomimetic type inhibitors such as BILN2061 and the like or inhibitors of HCV helicase.
  • inhibitors of HCV RNA dependent RNA polymerase such as, for example, nucleoside type polymerase inhibitors described in WOOl 90121(A2), or U.S. Pat. No. 6,348,587Bl or WO0160315 or WOO 132153
  • non-nucleoside inhibitors such as, for example,
  • agents to be administered in combination with a compound of the present invention include any agent or combination of agents that inhibit the replication of other viruses for co-infected individuals.
  • agents include but are not limited to therapies for disease caused by hepatitis B (HBV) infection such as, for example, adefovir, lamivudine, and tenofovir or therapies for disease caused by human immunodeficiency virus (HIV) infection such as, for example, protease inhibitors: ritonavir, lopinavir, indinavir, nelfinavir, saquinavir, amprenavir, atazanavir, tipranavir, TMC-114, fosamprenavir; reverse transcriptase inhibitors: zidovudine, lamivudine, didanosine, stavudine, tenofovir, zalcitabine, abacavir, efavirenz, nevirapine, delavirdine, TMC-125;
  • one aspect of the invention is directed to a method for treating or preventing an infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment one or more agents selected from the group consisting of a host immune modulator and a second antiviral agent, or a combination thereof, with a therapeutically effective amount of a compound or combination of compounds of the invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, prodrug, salt of a prodrug, or combination thereof.
  • Examples of the host immune modulator are, but not limited to, interferon-alpha, pegylated-interferon-alpha, interferon-beta, interferon-gamrna, a cytokine, a vaccine, and a vaccine comprising an antigen and an adjuvant, and said second antiviral agent inhibits replication of HCV either by inhibiting host cellular functions associated with viral replication or by targeting proteins of the viral genome.
  • Further aspect of the invention is directed to a method of treating or preventing infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment an agent or combination of agents that treat or alleviate symptoms of HCV infection including cirrhosis and inflammation of the liver, with a therapeutically effective amount of a compound or combination of compounds of the invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, prodrug, salt of a prodrug, or combination thereof.
  • Yet another aspect of the invention provides a method of treating or preventing infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment one or more agents that treat patients for disease caused by hepatitis B (HBV) infection, with a therapeutically effective amount of a compound or a combination of compounds of the invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, prodrug, salt of a prodrug, or combination thereof.
  • An agent that treats patients for disease caused by hepatitis B (HBV) infection may be for example, but not limited thereto, L- deoxythymidine, adefovir, lamivudine or tenfovir, or any combination thereof.
  • Example of the RNA-containing virus includes, but not limited to, hepatitis C virus (HCV).
  • Another aspect of the invention provides a method of treating or preventing infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment one or more agents that treat patients for disease caused by human immunodeficiency virus (HIV) infection, with a therapeutically effective amount of a compound or a combination of compounds of the invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, prodrug, salt of a prodrug, or combination thereof.
  • HIV human immunodeficiency virus
  • the agent that treats patients for disease caused by human immunodeficiency virus (HIV) infection may include, but is not limited thereto, ritonavir, lopinavir, indinavir, nelfmavir, saquinavir, amprenavir, atazanavir, tipranavir, TMC-1 14, fosamprenavir, zidovudine, lamivudine, didanosine, stavudine, tenofovir, zalcitabine, abacavir, efavirenz, nevirapine, delavirdine, TMC-125, L-870812, S-1360, enfuvirtide (T-20) or T-1249, or any combination thereof.
  • HIV human immunodeficiency virus
  • Example of the RNA-containing virus includes, but not limited to, hepatitis C virus (HCV).
  • HCV hepatitis C virus
  • the present invention provides the use of a compound or a combination of compounds of the invention, or a therapeutically acceptable salt form, stereoisomer, or tautomer, prodrug, salt of a prodrug, or combination thereof, and one or more agents selected from the group consisting of a host immune modulator and a second antiviral agent, or a combination thereof, to prepare a medicament for the treatment of an infection caused by an RNA-containing virus in a patient, particularly hepatitis C virus.
  • HCV hepatitis C virus
  • Examples of the host immune modulator are, but not limited to, interferon-alpha, pegylated- interferon-alpha, interferon-beta, interferon-gamma, a cytokine, a vaccine, and a vaccine comprising an antigen and an adjuvant, and said second antiviral agent inhibits replication of HCV either by inhibiting host cellular functions associated with viral replication or by targeting proteins of the viral genome.
  • combination of compound or compounds of the invention, together with one or more agents as defined herein above can be employed in pure form or, where such forms exist, in pharmaceutically acceptable salt form, prodrug, salt of a prodrug, or combination thereof.
  • combination of therapeutic agents can be administered as a pharmaceutical composition containing a therapeutically effective amount of the compound or combination of compounds of interest, or their pharmaceutically acceptable salt form, prodrugs, or salts of the prodrug, in combination with one or more agents as defined hereinabove, and a pharmaceutically acceptable carrier.
  • Such pharmaceutical compositions can be used for inhibiting the replication of an RNA-containing virus, particularly Hepatitis C virus (HCV), by contacting said virus with said pharmaceutical composition.
  • such compositions are useful for the treatment or prevention of an infection caused by an RNA-containing virus, particularly Hepatitis C virus (HCV).
  • further aspect of the invention is directed to a method of treating or preventing infection caused by an RNA-containing virus, particularly a hepatitis C virus (HCV), comprising administering to a patient in need of such treatment a pharmaceutical composition comprising a compound or combination of compounds of the invention or a pharmaceutically acceptable salt, stereoisomer, or tautomer, prodrug, salt of a prodrug, or combination thereof, one or more agents as defined hereinabove, and a pharmaceutically acceptable carrier.
  • HCV hepatitis C virus
  • the therapeutic agents When administered as a combination, the therapeutic agents can be formulated as separate compositions which are given at the same time or within a predetermined period of time, or the therapeutic agents can be given as a single unit dosage form.
  • Antiviral agents contemplated for use in such combination therapy include agents (compounds or biologicals) that are effective to inhibit the formation and/or replication of a virus in a mammal, including but not limited to agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of a virus in a mammal.
  • agents can be selected from another anti-HCV agent; an HIV inhibitor; an HAV inhibitor; and an HBV inhibitor.
  • Other anti-HCV agents include those agents that are effective for diminishing or preventing the progression of hepatitis C related symptoms or disease.
  • Such agents include but are not limited to immunomodulatory agents, inhibitors of HCV NS3 protease, other inhibitors of HCV polymerase, inhibitors of another target in the HCV life cycle and other anti-HCV agents, including but not limited to ribavirin, amantadine, levovirin and viramidine.
  • Immunomodulatory agents include those agents (compounds or biologicals) that are effective to enhance or potentiate the immune system response in a mammal.
  • Immunomodulatory agents include, but are not limited to, inosine monophosphate dehydrogenase inhibitors such as VX-497 (merimepodib, Vertex Pharmaceuticals), class I interferons, class II interferons, consensus interferons, asialo-interferons pegylated interferons and conjugated interferons, including but not limited to interferons conjugated with other proteins including but not limited to human albumin.
  • Class I interferons are a group of interferons that all bind to receptor type I, including both naturally and synthetically produced class I interferons, while class II interferons all bind to receptor type II.
  • Examples of class I interferons include, but are not limited to, [alpha]-, [beta]-, [delta]-, [omega]-, and [tau] -interferons, while examples of class II interferons include, but are not limited to, [gamma] -interferons.
  • Inhibitors of HCV NS3 protease include agents (compounds or biologicals) that are effective to inhibit the function of HCV NS3 protease in a mammal.
  • Inhibitors of HCV NS3 protease include, but are not limited to, those compounds described in WO 99/07733, WO 99/07734, WO 00/09558, WO 00/09543, WO 00/59929, WO 03/064416, WO 03/064455, WO 03/064456, WO 2004/030670, WO 2004/037855, WO 2004/039833, WO 2004/101602, WO 2004/101605, WO 2004/103996, WO 2005/028501 , WO 2005/070955, WO 2006/000085, WO 2006/007700 and WO 2006/007708 (all by Boehringer Ingelheim), WO 02/060926, WO 03/053349, WO03/099274, WO
  • Inhibitors of HCV polymerase include agents (compounds or biologicals) that are effective to inhibit the function of an HCV polymerase.
  • Such inhibitors include, but are not limited to, non-nucleoside and nucleoside inhibitors of HCV NS5B polymerase.
  • inhibitors of HCV polymerase include but are not limited to those compounds described in: WO 02/04425, WO 03/007945, WO 03/010140, WO 03/010141 , WO 2004/064925, WO 2004/065367, WO 2005/080388 and WO 2006/007693 (all by Boehringer Ingelheim), WO 2005/049622 (Japan Tobacco), WO 2005/014543 (Japan Tobacco),WO 2005/012288 (Genelabs), WO 2004/087714 (IRBM), WO 03/101993 (Neogenesis), WO 03/026587 (BMS), WO 03/000254 (Japan Tobacco), and WO 01/47883 (Japan Tobacco), and the clinical candidates XTL-2125, HCV 796, R-1626 and NM 283.
  • Inhibitors of another target in the HCV life cycle include agents (compounds or biologicals) that are effective to inhibit the formation and/or replication of HCV other than by inhibiting the function of the HCV NS3 protease. Such agents may interfere with either host or HCV viral mechanisms necessary for the formation and/or replication of HCV.
  • Inhibitors of another target in the HCV life cycle include, but are not limited to, entry inhibitors, agents that inhibit a target selected from a helicase, a NS2/3 protease and an internal ribosome entry site (IRES) and agents that interfere with the function of other viral targets including but not limited to an NS5A protein and an NS4B protein.
  • a patient may be co-infected with hepatitis C virus and one or more other viruses, including but not limited to human immunodeficiency virus (HIV), hepatitis A virus (HAV) and hepatitis B virus (HBV).
  • HAV human immunodeficiency virus
  • HAV hepatitis A virus
  • HBV hepatitis B virus
  • combination therapy to treat such co-infections by co-administering a compound according to the present invention with at least one of an HIV inhibitor, an HAV inhibitor and an HBV inhibitor.
  • the pharmaceutical compositions of the present invention may further comprise another anti-viral, anti-bacterial, anti-fungal or anti-cancer agent, or an immune modulator, or another thearapeutic agent.
  • the present invention includes methods of treating viral infection such as, but not limited to, hepatitis C infections in a subject in need of such treatment by administering to said subject an effective amount of a compound of the present invention or a pharmaceutically acceptable salt, ester, or prodrug thereof.
  • the present invention includes methods of treating hepatitis C infections in a subject in need of such treatment by administering to said subject an anti-HCV virally effective amount or an inhibitory amount of a pharmaceutical composition of the present invention.
  • An additional embodiment of the present invention includes methods of treating biological samples by contacting the biological samples with the compounds of the present invention.
  • Yet a further aspect of the present invention is a process of making any of the compounds delineated herein employing any of the synthetic means delineated herein.
  • viral infection refers to the introduction of a virus into cells or tissues, e.g., hepatitis C virus (HCV). In general, the introduction of a virus is also associated with replication. Viral infection may be determined by measuring virus antibody titer in samples of a biological fluid, such as blood, using, e.g., enzyme immunoassay. Other suitable diagnostic methods include molecular based techniques, such as RT-PCR, direct hybrid capture assay, nucleic acid sequence based amplification, and the like. A virus may infect an organ, e.g., liver, and cause disease, e.g., hepatitis, cirrhosis, chronic liver disease and hepatocellular carcinoma.
  • HCV hepatitis C virus
  • anti-cancer agent refers to a compound or drug capable of preventing or inhibiting the advancement of cancer.
  • agents include cis-platin, actinomycin D, doxorubicin, vincristine, vinblastine, etoposide, amsacrine, mitoxantrone, tenipaside, taxol, colchicine, cyclosporin A, phenothiazines or thioxantheres.
  • anti-fungal agent shall used to describe a compound which may be used to treat a fungus infection other than 3-AP, 3-AMP or prodrugs of 3-AP and 3-AMP according to the present invention.
  • Anti-fungal agents according to the present invention include, for example, terbinafine, fluconazole, itraconazole, posaconazole, clotrimazole, griseofulvin, nystatin, tolnaftate, caspofungin, amphotericin B, liposomal amphotericin B, and amphotericin B lipid complex.
  • antibacterial agent refers to both naturally occurring antibiotics produced by microorganisms to suppress the growth of other microorganisms, and agents synthesized or modified in the laboratory which have either bactericidal or bacteriostatic activity, e.g., ⁇ -lactam antibacterial agents, glycopeptides, macrolides, quinolones, tetracyclines, and aminoglycosides.
  • bacteriostatic it means that the agent essentially stops bacterial cell growth (but does not kill the bacteria); if the agent is bacteriocidal, it means that the agent kills the bacterial cells (and may stop growth before killing the bacteria).
  • immune modulator refers to any substance meant to alter the working of the humoral or cellular immune system of a subject.
  • immune modulators include inhibitors of mast cell-mediated inflammation, interferons, interleukins, prostaglandins, steroids, corticosteroids, colony-stimulating factors, chemotactic factors, etc.
  • compositions of the present invention may further comprise inhibitor(s) of other targets in the HCV life cycle, including, but not limited to, helicase, polymerase, metalloprotease, and internal ribosome entry site (IRES).
  • inhibitor(s) of other targets in the HCV life cycle including, but not limited to, helicase, polymerase, metalloprotease, and internal ribosome entry site (IRES).
  • IRS internal ribosome entry site
  • Ci-C 6 alkyl or “Ci-C 8 alkyl,” as used herein, refer to saturated, straight- or branched-chain hydrocarbon radicals containing between one and six, or one and eight carbon atoms, respectively.
  • Ci-C 6 alkyl radicals include, but are not limited to, methyl, ethyl, propyl, isopropyl, «-butyl, tert-butyl, neopentyl, n-hexyl radicals; and examples of Ci-C 8 alkyl radicals include, but are not limited to, methyl, ethyl, propyl, isopropyl, «-butyl, tert-butyl, neopentyl, n-hexyl, heptyl, octyl radicals.
  • C 2 -C 6 alkenyl or "C 2 -C 8 alkenyl,” as used herein, denote a monovalent group derived from a hydrocarbon moiety by the removal of a single hydrogen atom wherein the hydrocarbon moiety has at least one carbon-carbon double bond and contains from two to six, or two to eight carbon atoms, respectively.
  • Alkenyl groups include, but are not limited to, for example, ethenyl, propenyl, butenyl, 1 -methyl -2 -buten-1-yl, heptenyl, octenyl and the like.
  • C 2 -C 6 alkynyl or "C 2 -C 8 alkynyl,” as used herein, denote a monovalent group derived from a hydrocarbon moiety by the removal of a single hydrogen atom wherein the hydrocarbon moiety has at least one carbon-carbon triple bond and contains from two to six, or two to eight carbon atoms, respectively.
  • Representative alkynyl groups include, but are not limited to, for example, ethynyl, 1-propynyl, 1-butynyl, heptynyl, octynyl and the like.
  • C 3 -Cg-cycloalkyl or "C 3 -Ci 2 -cycloalkyl,” as used herein, denotes a monovalent group derived from a monocyclic or polycyclic saturated carbocyclic ring compound by the removal of a single hydrogen atom where the saturated carbocyclic ring compound has from 3 ot 8, or from 3 to 12, ring atoms, respectively.
  • C 3 -C 8 - cycloalkyl examples include, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopentyl and cyclooctyl; and examples of C 3 -Ci 2 -cycloalkyl include, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo [2.2.1] heptyl, and bicyclo [2.2.2] octyl.
  • C 3 -C 8 -cycloalkenyl or "C 3 -Ci 2 -cycloalkenyl” as used herein, denote a monovalent group derived from a monocyclic or polycyclic carbocyclic ring compound having at least one carbon-carbon double bond by the removal of a single hydrogen atom where the carbocyclic ring compound has from 3 to 8, or from 3 to 12, ring atoms, respectively.
  • C 3 -C 8 -cycloalkenyl examples include, but not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, and the like; and examples of C 3 -Ci 2 -cycloalkenyl include, but not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, and the like.
  • aryl refers to a mono- or bicyclic carbocyclic ring system having one or two aromatic rings including, but not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, idenyl and the like.
  • arylalkyl refers to a Ci-C 3 alkyl or Ci-C 6 alkyl residue attached to an aryl ring. Examples include, but are not limited to, benzyl, phenethyl and the like.
  • heteroaryl refers to a mono-, bi-, or tri-cyclic aromatic radical or ring having from five to ten ring atoms of which at least one ring atom is selected from S, O and N; wherein any N or S contained within the ring may be optionally oxidized.
  • Heteroaryl includes, but is not limited to, pyridinyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzooxazolyl, quinoxalinyl, and the like.
  • heteroarylalkyl refers to a Ci-C 3 alkyl or Ci-C 6 alkyl residue residue attached to a heteroaryl ring.
  • heterocycloalkyl refers to a non-aromatic 3-, 4-, 5-, 6- or 7-membered ring or a bi- or tri-cyclic group fused system, where (i) each ring contains between one and three heteroatoms independently selected from oxygen, sulfur and nitrogen, (ii) each 5-membered ring has 0 to 1 double bonds and each 6-membered ring has 0 to 2 double bonds, (iii) the nitrogen and sulfur heteroatoms may optionally be oxidized, (iv) the nitrogen heteroatom may optionally be quaternized, and (iv) any of the above rings may be fused to a benzene ring.
  • heterocycloalkyl groups include, but are not limited to, [l,3]dioxolane, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, and tetrahydrofuryl.
  • substituted refers to independent replacement of one, two, or three or more of the hydrogen atoms thereon with substituents including, but not limited to, -F, -Cl, -Br, -I, -OH, protected hydroxy, -NO 2 , -CN, -NH 2 , protected amino, -NH -d-C ⁇ -alkyl, -NH -C 2 -C 12 -alkenyl, -NH -C 2 -C 12 -alkenyl, -NH -C 3 -C 12 -cycloalkyl, -NH - aryl, -NH -heteroaryl, -NH -heterocycloalkyl, -dialkylamino, -diarylamino, - diheteroarylamino, -O-Ci-Ci 2 -alkyl, -O-C 2 -Ci 2 -alkeny
  • each substituent in a substituted moiety is additionally optionally substituted with one or more groups, each group being independently selected from -F, -Cl, -Br, -I, -OH, -NO 2 , -CN, or -NH 2 .
  • any of the aryls, substituted aryls, heteroaryls and substituted heteroaryls described herein, can be any aromatic group.
  • Aromatic groups can be substituted or unsubstituted.
  • any alkyl, alkenyl, alkynyl, cycloalkyl and cycloalkenyl moiety described herein can also be an aliphatic group, an alicyclic group or a heterocyclic group.
  • An "aliphatic group” is non-aromatic moiety that may contain any combination of carbon atoms, hydrogen atoms, halogen atoms, oxygen, nitrogen or other atoms, and optionally contain one or more units of unsaturation, e.g., double and/or triple bonds.
  • An aliphatic group may be straight chained, branched or cyclic and preferably contains between about 1 and about 24 carbon atoms, more typically between about 1 and about 12 carbon atoms.
  • aliphatic groups include, for example, polyalkoxyalkyls, such as polyalkylene glycols, polyamines, and polyimines, for example. Such aliphatic groups may be further substituted. It is understood that aliphatic groups may be used in place of the alkyl, alkenyl, alkynyl, alkylene, alkenylene, and alkynylene groups described herein.
  • alicyclic denotes a monovalent group derived from a monocyclic or polycyclic saturated carbocyclic ring compound by the removal of a single hydrogen atom.
  • Examples include, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo [2.2.1] heptyl, and bicyclo [2.2.2] octyl. Such alicyclic groups may be further substituted.
  • heterocyclic refers to a non-aromatic 5-, 6- or 7- membered ring or a bi- or tri-cyclic group fused system, where (i) each ring contains between one and three heteroatoms independently selected from oxygen, sulfur and nitrogen, (ii) each 5-membered ring has 0 to 1 double bonds and each 6-membered ring has 0 to 2 double bonds, (iii) the nitrogen and sulfur heteroatoms may optionally be oxidized, (iv) the nitrogen heteroatom may optionally be quaternized, (iv) any of the above rings may be fused to a benzene ring, and (v) the remaining ring atoms are carbon atoms which may be optionally oxo-substituted.
  • heterocycloalkyl groups include, but are not limited to, [l,3]dioxolane, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, quinoxalinyl, pyridazinonyl, and tetrahydrofuryl.
  • Such heterocyclic groups may be further substituted to give substituted heterocyclic.
  • alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, arylalkyl, heteroarylalkyl, and heterocycloalkyl are intended to be monovalent or divalent.
  • alkylene, alkenylene, and alkynylene, cycloaklylene, cycloalkenylene, cycloalkynylene, arylalkylene, hetoerarylalkylene and heterocycloalkylene groups are to be included in the above definitions, and are applicable to provide the formulas herein with proper valency.
  • hydroxy activating group refers to a labile chemical moiety which is known in the art to activate a hydroxy group so that it will depart during synthetic procedures such as in a substitution or elimination reactions.
  • hydroxy activating group include, but not limited to, mesylate, tosylate, triflate, p-nitrobenzoate, phosphonate and the like.
  • activated hydroxy refers to a hydroxy group activated with a hydroxy activating group, as defined above, including mesylate, tosylate, triflate, p- nitrobenzoate, phosphonate groups, for example.
  • protected hydroxy refers to a hydroxy group protected with a hydroxy protecting group, as defined above, including benzoyl, acetyl, trimethylsilyl, triethylsilyl, and methoxymethyl groups.
  • halo and halogen, as used herein, refer to an atom selected from fluorine, chlorine, bromine and iodine.
  • the compounds described herein contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- , or as (D)- or (L)- for amino acids.
  • the present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms.
  • Optical isomers may be prepared from their respective optically active precursors by the procedures described above, or by resolving the racemic mixtures. The resolution can be carried out in the presence of a resolving agent, by chromatography or by repeated crystallization or by some combination of these techniques, which are known to those skilled in the art.
  • subject refers to a mammal.
  • a subject therefore refers to, for example, dogs, cats, horses, cows, pigs, guinea pigs, and the like.
  • the subject is a human.
  • the subject may be referred to herein as a patient.
  • the term "pharmaceutically acceptable salt” refers to those salts of the compounds formed by the process of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art.
  • hydroxy protecting group refers to a labile chemical moiety which is known in the art to protect a hydroxy group against undesired reactions during synthetic procedures. After said synthetic procedure(s) the hydroxy protecting group as described herein may be selectively removed. Hydroxy protecting groups as known in the are described generally in T.H. Greene and P. G., S. M. Wuts, Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, New York (1999).
  • hydroxy protecting groups include benzyloxycarbonyl, 4-nitrobenzyloxycarbonyl, 4- bromobenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, methoxycarbonyl, tert- butoxycarbonyl, isopropoxycarbonyl, diphenylmethoxycarbonyl, 2,2,2- trichloroethoxycarbonyl, 2-(trimethylsilyl)ethoxycarbonyl, 2-furfuryloxycarbonyl, allyloxycarbonyl, acetyl, formyl, chloroacetyl, trifluoroacetyl, methoxyacetyl, phenoxyacetyl, benzoyl, methyl, t-butyl, 2,2,2-trichloroethyl, 2-trimethylsilyl ethyl, 1,1- dimethyl-2-propenyl, 3-methyl- 3 -butenyl, allyl, benzyl, para- methoxybenzyl
  • Preferred hydroxy protecting groups for the present invention are acetyl (Ac or -C(O)CH 3 ), benzoyl (Bz or -C(O)C 6 H 5 ), and trimethylsilyl (TMS or-Si(CH 3 ) 3 ).Berge, et al. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66: 1-19 (1977). The salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or separately by reacting the free base function with a suitable organic acid.
  • salts include, but are not limited to, nontoxic acid addition salts e.g., salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • nontoxic acid addition salts e.g., salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • salts include, but are not limited to, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamo
  • alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, alkyl having from 1 to 6 carbon atoms, sulfonate and aryl sulfonate.
  • amino protecting group refers to a labile chemical moiety which is known in the art to protect an amino group against undesired reactions during synthetic procedures. After said synthetic procedure(s) the amino protecting group as described herein may be selectively removed.
  • Amino protecting groups as known in the are described generally in T.H. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, New York (1999). Examples of amino protecting groups include, but are not limited to, t-butoxycarbonyl, 9- fluorenylmethoxycarbonyl, benzyloxycarbonyl, and the like.
  • ester refers to esters of the compounds formed by the process of the present invention which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof.
  • Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms.
  • esters include, but are not limited to, formates, acetates, propionates, butyrates, acrylates and ethylsuccinates.
  • prodrugs refers to those prodrugs of the compounds formed by the process of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the present invention.
  • Prodrug as used herein means a compound, which is convertible in vivo by metabolic means (e.g. by hydrolysis) to afford any compound delineated by the formulae of the instant invention.
  • prodrugs are known in the art, for example, as discussed in Bundgaard, (ed.), Design of Prodrugs, Elsevier (1985); Widder, et al. (ed.), Methods in Enzymology, vol. 4, Academic Press (1985); Krogsgaard-Larsen, et al., (ed). "Design and Application of Prodrugs, Textbook of Drug Design and Development, Chapter 5, 113-191 (1991); Bundgaard, et al., Journal of Drug Deliver Reviews, 8:1-38(1992); Bundgaard, J. of Pharmaceutical Sciences, 77:285 et seq.
  • acyl includes residues derived from acids, including but not limited to carboxylic acids, carbamic acids, carbonic acids, sulfonic acids, and phosphorous acids. Examples include aliphatic carbonyls, aromatic carbonyls, aliphatic sulfonyls, aromatic sulfinyls, aliphatic sulfinyls, aromatic phosphates and aliphatic phosphates. Examples of aliphatic carbonyls include, but are not limited to, acetyl, propionyl, 2-fluoroacetyl, butyryl, 2-hydroxy acetyl, and the like.
  • aprotic solvent refers to a solvent that is relatively inert to proton activity, i.e., not acting as a proton-donor.
  • examples include, but are not limited to, hydrocarbons, such as hexane and toluene, for example, halogenated hydrocarbons, such as, for example, methylene chloride, ethylene chloride, chloroform, and the like, heterocyclic compounds, such as, for example, tetrahydrofuran and N-methylpyrrolidinone, and ethers such as diethyl ether, bis-methoxymethyl ether.
  • solvents are well known to those skilled in the art, and individual solvents or mixtures thereof may be preferred for specific compounds and reaction conditions, depending upon such factors as the solubility of reagents, reactivity of reagents and preferred temperature ranges, for example. Further discussions of aprotic solvents may be found in organic chemistry textbooks or in specialized monographs, for example: Organic Solvents Physical Properties and Methods of Purification. 4th ed., edited by John A. Riddick et al, Vol. II, in the Techniques of Chemistry Series, John Wiley & Sons, NY, 1986.
  • protogenic organic solvent or “protic solvent” as used herein, refer to a solvent that tends to provide protons, such as an alcohol, for example, methanol, ethanol, propanol, isopropanol, butanol, t-butanol, and the like.
  • solvents are well known to those skilled in the art, and individual solvents or mixtures thereof may be preferred for specific compounds and reaction conditions, depending upon such factors as the solubility of reagents, reactivity of reagents and preferred temperature ranges, for example. Further discussions of protogenic solvents may be found in organic chemistry textbooks or in specialized monographs, for example: Organic Solvents Physical Properties and Methods of Purification, 4th ed., edited by John A. Riddick et al, Vol. II, in the Techniques of Chemistry Series. John Wiley & Sons, NY, 1986.
  • stable refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., therapeutic or prophylactic administration to a subject).
  • the synthesized compounds can be separated from a reaction mixture and further purified by a method such as column chromatography, high pressure liquid chromatography, or recrystallization.
  • a method such as column chromatography, high pressure liquid chromatography, or recrystallization.
  • further methods of synthesizing the compounds of the formulae herein will be evident to those of ordinary skill in the art.
  • the various synthetic steps may be performed in an alternate sequence or order to give the desired compounds.
  • the solvents, temperatures, reaction durations, etc. delineated herein are for purposes of illustration only and one of ordinary skill in the art will recognize that variation of the reaction conditions can produce the desired bridged macrocyclic products of the present invention.
  • Synthetic chemistry transformations and protecting group methodologies useful in synthesizing the compounds described herein are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations. VCH Publishers (1989); T. W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 2d. Ed., John Wiley and Sons (1991); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995).
  • the compounds of this invention may be modified by appending various functionalities via any synthetic means delineated herein to enhance selective biological properties.
  • modifications are known in the art and include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.
  • compositions of the present invention comprise a therapeutically effective amount of a compound of the present invention formulated together with one or more pharmaceutically acceptable carriers.
  • pharmaceutically acceptable carrier means a non-toxic, inert solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
  • materials which can serve as pharmaceutically acceptable carriers are 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 a propylene 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, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulf
  • compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), buccally, or as an oral or nasal spray.
  • the pharmaceutical compositions of this invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir, preferably by oral administration or administration by injection.
  • the pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles.
  • the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra- articular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • the oral compositions can also include adjuvants such as wetting agents, e
  • sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • the injectable formulations can be sterilized, for example, by filtration through a bacterial -retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • the rate of drug release can be controlled.
  • biodegradable polymers include poly(orthoesters) and poly(anhydrides).
  • Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.
  • compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or: a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and g
  • 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 sugar as well as high molecular weight polyethylene glycols and the like.
  • the active compounds can also be in micro-encapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.
  • the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • buffering agents include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required.
  • Ophthalmic formulation, ear drops, eye ointments, powders and solutions are also contemplated as being within the scope of this invention.
  • 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 the compounds 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.
  • Transdermal patches have the added advantage of providing controlled delivery of a compound to the body.
  • dosage forms can be made by dissolving or dispensing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin.
  • the rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • an inhibitory amount or dose of the compounds of the present invention may range from about 0.1 mg/Kg to about 500 mg/Kg, alternatively from about 1 to about 50 mg/Kg. Inhibitory amounts or doses will also vary depending on route of administration, as well as the possibility of co-usage with other agents.
  • viral infections are treated or prevented in a subject such as a human or lower mammal by administering to the subject an anti-hepatitis C virally effective amount or an inhibitory amount of a compound of the present invention, in such amounts and for such time as is necessary to achieve the desired result.
  • An additional method of the present invention is the treatment of biological samples with an inhibitory amount of a compound of composition of the present invention in such amounts and for such time as is necessary to achieve the desired result.
  • anti-hepatitis C virally effective amount of a compound of the invention, as used herein, mean a sufficient amount of the compound so as to decrease the viral load in a biological sample or in a subject.
  • an anti-hepatitis C virally effective amount of a compound of this invention will be at a reasonable benefit/risk ratio applicable to any medical treatment.
  • inhibitory amount of a compound of the present invention means a sufficient amount to decrease the hepatitis C viral load in a biological sample or a subject. It is understood that when said inhibitory amount of a compound of the present invention is administered to a subject it will be at a reasonable benefit/risk ratio applicable to any medical treatment as determined by a physician.
  • biological sample(s), means a substance of biological origin intended for administration to a subject. Examples of biological samples include, but are not limited to, blood and components thereof such as plasma, platelets, subpopulations of blood cells and the like; organs such as kidney, liver, heart, lung, and the like; sperm and ova; bone marrow and components thereof; or stem cells.
  • another embodiment of the present invention is a method of treating a biological sample by contacting said biological sample with an inhibitory amount of a compound or pharmaceutical composition of the present invention.
  • a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary.
  • the dosage or frequency of administration, or both may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level, treatment should cease.
  • the subject may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
  • the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific inhibitory dose for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts.
  • the total daily inhibitory dose of the compounds of this invention administered to a subject in single or in divided doses can be in amounts, for example, from 0.01 to 50 mg/kg body weight or more usually from 0.1 to 25 mg/kg body weight.
  • Single dose compositions may contain such amounts or submultiples thereof to make up the daily dose.
  • treatment regimens according to the present invention comprise administration to a patient in need of such treatment from about 10 mg to about 1000 mg of the compound(s) of this invention per day in single or multiple doses. Unless otherwise defined, all technical and scientific terms used herein are accorded the meaning commonly known to one with ordinary skill in the art. All publications, patents, published patent applications, and other references mentioned herein are hereby incorporated by reference in their entirety.
  • BME for 2-mercaptoethanol
  • BOP for benzotriazol- 1 -yloxy-tris(dimethylamino)phosphonium hexafluorophosphate ;
  • DIBAL-H for diisobutylaluminum hydride
  • DIEA for diisopropyl ethylamine
  • DMAP for N,N-dimethylaminopyridine
  • DME for ethylene glycol dimethyl ether
  • DMEM Dulbecco's Modified Eagles Media
  • DMF for N,N-dimethyl formamide
  • DMSO for dimethylsulfoxide
  • KHMDS potassium bis(trimethylsilyl) amide
  • Ph for phenyl
  • RCM for ring-closing metathesis
  • TFA for trifluoroacetic acid
  • THF for tetrahydrofuran
  • TPP or PPh 3 for triphenylphosphine
  • tBOC or Boc for tert-butyloxy carbonyl
  • the quinoxaline and quinoline analogs of the present invention were prepared via several different synthetic routes.
  • the simplest method, shown in Scheme 2, was to condense lH-quinoxalin-2-one analogs (2-2), or Hydroxyquinolines (2-3), where R6, R71, R72, R73, R74 and J are as defined previously, with key intermediate ⁇ -l by using Mitsunobu conditions to give compound 2-1.
  • Mitsunobu conditions see O. Mitsunobu, Synthesis 1981, 1-28; D. L. Hughes, Org. React. 29, 1-162 (1983); D. L. Hughes, Organic Preparations and Procedures Int. 28, 127-164 (1996); and J. A. Dodge, S. A. Jones, Recent Res. Dev. Org. Chem. 1, 273-283 (1997).
  • the macrocyclic starting material 3-1 was prepared following Scheme- 1 by starting with the commercially available trans-Boc-hydroxyproline.
  • Compounds of Formula 3-3 (the carbamates) were prepared by reacting 3-1 with CDI and isoindoline derivatives 3-2.
  • Scheme 3 R71, R72, R73 and R74 are as previously defined in Formula I.
  • Scheme 4 illustrates the general synthetic method of the Oximyl macrocyclics 4-3.
  • a suitable leaving group such as, but not limited to OMs, OTs, OTf, bromide, or iodide.
  • Compound (4-1) was subsequently treated with an aryl Oxime (i.e. compound 4-2) at the presence of a base such as, but not limited to K 2 CO 3 , Pyridine, TEA, DBU in a suitable solvent like DMF, DMSO, THF etc. to provide compound 4-3.
  • a base such as, but not limited to K 2 CO 3 , Pyridine, TEA, DBU in a suitable solvent like DMF, DMSO, THF etc.
  • Scheme 5 illustrates the oxidation of the p3 nitrogen.
  • Compound 5-1 was subjected to the Boc deprotection procedure, such as, but not limited to hydrochloric acid, to provide the free amino compound, which reacted with aryl aldehyde under reductive amination condition to give compound 5-2.
  • the p3 nitrogen of formula (5-2) was oxidized with H2O2 using Na2WO4 as catalyst.
  • the resulting compound was hydrolyzed to give free amino alcohol 5-3.
  • Alkylated or acylated with appropriate alkyl halide or activated acyl groups (A-X) followed by hydrolysis to give compounds of formula (5-4).
  • the carboxylic acid was treated with sulfonamide to provide compounds of formula (5-5).
  • Scheme 6 illustrates a method to prepare compounds with substitution groups on the hydroxyl group (i.e. compounds 6-3).
  • Compound 6-1 was prepared by treating the ⁇ - bromocarboxylic acid with the hydroxylamine following the procedure described in the literature (J. Org. Chem. 2000, 65(9), 2684-2695).
  • Compound 6-2 was made by: 1) introducing the Q group into hydroxyl proline following the procedure described in schemes 2-4; 2) hydrolysis of the resulting ester followed by coupling with the Pl aminoacid piece; 3) hydrolysis of the resulting ester followed by coupling with the sulfonamide following the procedures described in schemes 1 and 5.
  • Compounds 6-3 was then made by coupling 6-1 and 6-2 followed by metathesis as described in scheme- 1.
  • MS Found 516.28, M+Na + .
  • Step 2B The title compound of Step 2A (200 mg, 0.28 mmol) was treated with HCl (4 M in dioxane, 3 mL, 12 mmol). The reaction mixture was stirred at room temperature for 1 h until LCMS showed the complete consumption of starting material. The solvent was removed in vacuo. The residue was dissolved in DCM (3 ml). The solvent was removed in vacuo and the residue was used directly in next step.
  • Step 2D To a solution of compound E-2-4 (13 mg) in 3 ml dry DCM was treated with cyclopentyl chloroformate (3 eqiv.) at the presence of /Pr 2 NEt (5 eqiv.). The reaction mixture is stirred for 2 hours. The reaction mixture was diluted with 20 mL EtOAc, and washed with NaHCO 3 aq. (10 ml) and brine (10 ml). The organic phase was dried over anhydrous Na 2 SO 4 , filtered, and then concentrated in vacuo. The residue was dissolved in 3 mL of dioxane and 1 mL of 1 N LiOH aqueous solution, and the resulting mixture was stirred at room temperature for 20 hours.
  • Step 3A Cyclopropylsulfonyl chloride (1.4g, 10 mmol) was dissolved in 0.5 M ammonia in dioxane (50 ml, 25 mmol) at RT. The reaction was kept at RT for 3 days. The large amount of precipitation was filtered and discarded. The clear filtrate was evaporated in vacuo and the white residue was dried on vacuum for 24 hours to give the cyclopropylsulfonamide (0.88 g, 74%).
  • 1 H-NMR 500 MHz, CD 3 Cl): ⁇ 4.62 (2H, s), 2.59 (IH, m), 1.20 (2H, m), 1.02 (2H, m).
  • Step 3B The title compound from Example 2 (2.0 mg) and carbonyldiimidazole (1.0 mg) were dissolved in 0.7 ml anhydrous DMF and the resulting solution was heated to 4O 0 C for 1 hour. Cyclopropylsulfonamide (1.0 mg) was added to the reaction followed by DBU (1.0 mg). The reaction mixture was stirred at 4O 0 C for 10 hour. LCMS showed the formation of the desired product. The reaction was cooled down and 10 ml ethyl acetate was added to the solution. The mixture was washed with saturated aqueous NaHCO 3 solution, water and brine. The organic layer was dried over anhydrous sodium sulfate.
  • the title compound is prepared following the procedures described in Example 2 by using appropriate chloroformate reagents.
  • the title compound is made by reacting the title compound of Example 1 with
  • the title compound is prepared following the procedures described in Example 3 by starting with the title compound of Example 2 and 2-thiophenesulfonamide.
  • the mesylate compound from step 14A (1.0 eqiv.), (1.2 eqiv.), and K2CO3 (2 eqiv.) are dissolved in DMF or DMSO.
  • the resulting reaction mixture is stirred at 40-80°C for 10 hours, cooled and extracted with ethyl acetate.
  • the organic extract is washed with water (2x30ml), and the organic solution is concentrated in vacuo, subsequently purified by column chromatography eluting with 50% ethyl acetate in hexanes to give the product.
  • the title compound is prepared following the procedures described in Example 14 by using appropriate chloroformate reagents.
  • the title compound is prepared following the procedures described in Example 14 by using appropriate chloroformate reagents.
  • the title compound 28-3 is made following the procedures described in Example 14 (steps 14A and 14B).
  • Examples 29 to Examples 32 below are made following the procedures described in Examples 28 and 3 by starting with the approporiate materials.
  • Compound 33-4 was prepared from 1.-3 by the standard hydrolysis reaction as described in the PCT WO 2004113365.
  • Compound 33-6 was prepared from the standard coupling reaction of 33-4 and 33-5 as described in the PCT WO 2004113365.
  • oxim core intermediate 33-8 (0.053mmol) and DIPEA (0.09ml, 0.516mmolmmol) in DMF (1.5ml) at 0 0 C was added HATU (26mg, 0.068mmol). The mixture was stirred at room temperature for 18h, diluted with EtOAc and washed with half-sat.-aq. NaCl four times. The organic phase was dried over anhydrous MgSO 4 , filtered, and then concentrated in vacuo.
  • the compounds of the present invention exhibit potent inhibitory properties against the HCV NS3 protease.
  • the following examples elucidate assays in which the compounds of the present invention are tested for anti-HCV effects.
  • HCV protease activity and inhibition is assayed using an internally quenched fluorogenic substrate.
  • a DABCYL and an EDANS group are attached to opposite ends of a short peptide. Quenching of the EDANS fluorescence by the DABCYL group is relieved upon proteolytic cleavage. Fluorescence was measured with a Molecular Devices Fluoromax (or equivalent) using an excitation wavelength of 355 nm and an emission wavelength of 485 nm.
  • the assay is run in Corning white half-area 96-well plates (VWR 29444-312 [Corning 3693]) with full-length NS3 HCV protease Ib tethered with NS4A cofactor (final enzyme concentration 1 to 15 nM).
  • the assay buffer is complemented with 10 ⁇ M NS4A cofactor Pep 4A (Anaspec 25336 or in-house, MW 1424.8).
  • RET S 1 (Ac- Asp-Glu- ASP(EDANS)-GIu-GIu-AbU-[COO]AIa-SCr-LyS-(D ABCYL)- NH 2 , . AnaSpec 22991, MW 1548.6) is used as the fluorogenic peptide substrate.
  • the assay buffer contained 50 mM Hepes at pH 7.5, 30 mM NaCl and 10 mM BME.
  • the enzyme reaction is followed over a 30 minutes time course at room temperature in the absence and presence of inhibitors.
  • the peptide inhibitors HCV Inh 1 (Anaspec 25345, MW 796.8) Ac-Asp-Glu-Met- Glu-Glu-Cys-OH, [-20 0 C] and HCV Inh 2 (Anaspec 25346, MW 913.1) Ac- Asp-Glu-Dif-Cha-Cys-OH, were used as reference compounds.
  • IC50 values were calculated using XLFit in ActivityBase (IDBS) using equation
  • HCV Cell Based Assay Cell lines, including Huh- 11 -7 or Huh 9-13, harboring HCV replicons (Lohmann, et al Science 285:110-113, 1999) are seeded at 5xlO 3 cells/well in 96 well plates and fed media containing DMEM (high glucose), 10% fetal calf serum, penicillin- streptomycin and non-essential amino acids. Cells are incubated in a 5% CO 2 incubator at 37 0 C. At the end of the incubation period, total RNA is extracted and purified from cells using Qiagen Rneasy 96 Kit (Catalog No. 74182). To amplify the
  • HCV RNA so that sufficient material can be detected by an HCV specific probe (below), primers specific for HCV (below) mediate both the reverse transcription of the HCV RNA and the amplification of the cDNA by polymerase chain reaction (PCR) using the TaqMan One-Step RT-PCR Master Mix Kit (Applied Biosystems catalog no. 4309169).
  • the nucleotide sequences of the RT-PCR primers which are located in the NS5B region of the HCV genome, are the following: HCV Forward primer "RBNS5bfor" 5 'GCTGCGGCCTGTCGAGCT: HCV Reverse primer "RBNS5Brev”: 5 'CAAGGTCGTCTCCGCATAC.
  • Detection of the RT-PCR product is accomplished using the Applied Biosystems (ABI) Prism 7700 Sequence Detection System (SDS) that detects the fluorescence that is emitted when the probe, which is labeled with a fluorescence reporter dye and a quencher dye, is processed during the PCR reaction.
  • SDS Sequence Detection System
  • the increase in the amount of fluorescence is measured during each cycle of PCR and reflects the increasing amount of RT-PCR product.
  • quantification is based on the threshold cycle, where the amplification plot crosses a defined fluorescence threshold. Comparison of the threshold cycles of the sample with a known standard provides a highly sensitive measure of relative template concentration in different samples (ABI User Bulletin #2 December 11, 1997).
  • the data is analyzed using the ABI SDS program version 1.7.
  • the relative template concentration can be converted to RNA copy numbers by employing a standard curve of HCV RNA standards with known copy number (ABI User Bulletin #2 December 11, 1997).
  • the RT reaction is performed at 48 0 C for 30 minutes followed by PCR.
  • Thermal cycler parameters used for the PCR reaction on the ABI Prism 7700 Sequence Detection System are: one cycle at 95 0 C, 10 minutes followed by 35 cycles each of which include one incubation at 95 0 C for 15 seconds and a second incubation for 60
  • RT-PCR is performed on the cellular messenger RNA glyceraldehydes-3 -phosphate dehydrogenase (GAPDH).
  • GAPDH messenger RNA glyceraldehydes-3 -phosphate dehydrogenase
  • the GAPDH copy number is very stable in the cell lines used.
  • GAPDH RT-PCR is performed on the same exact RNA sample from which the HCV copy number is determined.
  • the GAPDH primers and probes, as well as the standards with which to determine copy number, are contained in the ABI Pre- Developed TaqMan Assay Kit (catalog no. 4310884E).
  • the ratio of HCV/GAPDH RNA is used to calculate the activity of compounds evaluated for inhibition of HCV
  • HCV replicon RNA levels in Huh- 1 1-7 or 9-13 cells is determined by comparing the amount of HCV RNA normalized to GAPDH (e.g. the ratio of HCV/GAPDH) in the cells exposed to compound versus cells exposed to the 0% inhibition and the 100% inhibition controls.
  • cells are seeded at 5x 10 3 cells/well in a 96 well plate and are incubated either with: 1) media containing 1% DMSO (0% inhibition control), 2) 100 international units, IU/ml Interferon-alpha 2b in media/1 %DMSO or 3) media/1 %DMSO containing a fixed concentration of compound.
  • 96 well plates as described above are then incubated at 37 0 C for 3 days (primary screening assay) or 4 days (IC50 determination). Percent inhibition is defined as:
  • C2 the ratio of HCV RNA copy number/GAPDH RNA copy number in the 100% inhibition control ( 100 IU/ml Interferon-alpha 2b).
  • HCV NS3 proteases of different HCV genotypes including genotypes 1, 2, 3 and 4.
  • Representative compounds were tested in the above assays (Example 122 and Example 123).

Abstract

The present invention relates to compounds of Formula (I), or a pharmaceutically acceptable salt, ester, or prodrug, thereof: which inhibit serine protease activity, particularly the activity of hepatitis C virus (HCV) NS3-NS4A protease. Consequently, the compounds of the present invention interfere with the life cycle of the hepatitis C virus and are also useful as antiviral agents. The present invention further relates to pharmaceutical compositions comprising the aforementioned compounds for administration to a subject suffering from HCV infection. The invention also relates to methods of treating an HCV infection in a subject by administering a pharmaceutical composition comprising the compounds of the present invention.

Description

P3 HYDROXYAMINO MACROCYCLIC HEPATITIS C SERINE PROTEASE
INHIBITORS
Inventors: Deqiang Niu, Dong Liu, Yonghua Gai, Yat Sun Or, Zhe Wang
RELATED APPLICATIONS
This application claims the benefit of US provisional application number 60/981,613 filed on October 22, 2007. The contents of the above application is incorporated herein by reference. TECHNICAL FIELD
The present invention relates to novel macrocycles having activity against the hepatitis C virus (HCV) and useful in the treatment of HCV infections. More particularly, the invention relates to N-hydroxyl macrocyclic compounds, compositions containing such compounds and methods for using the same, as well as processes for making such compounds.
BACKGROUND OF THE INVENTION
HCV is the principal cause of non-A, non-B hepatitis and is an increasingly severe public health problem both in the developed and developing world. It is estimated that the virus infects over 200 million people worldwide, surpassing the number of individuals infected with the human immunodeficiency virus (HIV) by nearly five fold. HCV infected patients, due to the high percentage of individuals inflicted with chronic infections, are at an elevated risk of developing cirrhosis of the liver, subsequent hepatocellular carcinoma and terminal liver disease. HCV is the most prevalent cause of hepatocellular cancer and cause of patients requiring liver transplantations in the western world. There are considerable barriers to the development of anti-HCV therapeutics, which include, but are not limited to, the persistence of the virus, the genetic diversity of the virus during replication in the host, the high incident rate of the virus developing drug -resistant mutants, and the lack of reproducible infectious culture systems and small-animal models for HCV replication and pathogenesis. In a majority of cases, given the mild course of the infection and the complex biology of the liver, careful consideration must be given to antiviral drugs, which are likely to have significant side effects.
Only two approved therapies for HCV infection are currently available. The original treatment regimen generally involves a 3-12 month course of intravenous interferon-α (IFN-α), while a new approved second-generation treatment involves co- treatment with IFN-α and the general antiviral nucleoside mimics like ribavirin. Both of these treatments suffer from interferon related side effects as well as low efficacy against HCV infections. There exists a need for the development of effective antiviral agents for treatment of HCV infection due to the poor tolerability and disappointing efficacy of existing therapies.
In a patient population where the majority of individuals are chronically infected and asymptomatic and the prognoses are unknown, an effective drug would desirably possess significantly fewer side effects than the currently available treatments. The hepatitis C non- structural protein-3 (NS3) is a proteolytic enzyme required for processing of the viral polyprotein and consequently viral replication. Despite the huge number of viral variants associated with HCV infection, the active site of the NS 3 protease remains highly conserved thus making its inhibition an attractive mode of intervention. Recent success in the treatment of HIV with protease inhibitors supports the concept that the inhibition of NS3 is a key target in the battle against HCV.
HCV is a flaviridae type RNA virus. The HCV genome is enveloped and contains a single strand RNA molecule composed of circa 9600 base pairs. It encodes a polypeptide comprised of approximately 3010 amino acids.
The HCV polyprotein is processed by viral and host peptidase into 10 discreet peptides which serve a variety of functions. There are three structural proteins, C, El and E2. The P7 protein is of unknown function and is comprised of a highly variable sequence. There are six non-structural proteins. NS2 is a zinc-dependent metalloproteinase that functions in conjunction with a portion of the NS3 protein. NS3 incorporates two catalytic functions (separate from its association with NS2): a serine protease at the N-terminal end, which requires NS4A as a cofactor, and an ATP-ase-dependent helicase function at the carboxyl terminus. NS4A is a tightly associated but non-covalent cofactor of the serine protease.
The NS3-NS4A protease is responsible for cleaving four sites on the viral polyprotein. The NS3-NS4A cleavage is autocatalytic, occurring in cis. The remaining three hydrolyses, NS4A-NS4B, NS4B-NS5A and NS5A-NS5B all occur in trans. NS3 is a serine protease which is structurally classified as a chymotrypsin-like protease. While the NS serine protease possesses proteolytic activity by itself, the HCV protease enzyme is not an efficient enzyme in terms of catalyzing polyprotein cleavage. It has been shown that a central hydrophobic region of the NS4A protein is required for this enhancement. The complex formation of the NS3 protein with NS4A seems necessary to the processing events, enhancing the proteolytic efficacy at all of the sites.
A general strategy for the development of antiviral agents is to inactivate virally encoded enzymes, including NS3, that are essential for the replication of the virus. Current efforts directed toward the discovery of NS3 protease inhibitors were reviewed by S. Tan, A. Pause, Y. Shi, N. Sonenberg, Hepatitis C Therapeutics: Current Status and Emerging Strategies, Nature Rev. Drug Discov., 1, 867-881 (2002). Other patent disclosures describing the synthesis of HCV protease inhibitors are: WO 2006/007700; US 2005/0261200; WO 2004/113365; WO 03/099274 (2003); US 2003/0008828;
US2002/0037998 (2002); WO 00/59929 (2000); WO 00/09543 (2000); WO 99/50230 (1999); US5861297 (1999); WO 99/07733 (1999).
SUMMARY OF THE INVENTION The present invention relates to novel N-hydroxyl macrocyclic compounds and pharmaceutically acceptable salts, esters or prodrugs thereof, methods of using the same to treat hepatitis C infection in a subject in need of such therapy . Macrocyclic compounds of the present invention interfere with the life cycle of the hepatitis C virus and are also useful as antiviral agents. The present invention further relates to pharmaceutical compositions comprising the aforementioned compounds, salts, esters or prodrugs for administration to a subject suffering from HCV infection. The present invention further features pharmaceutical compositions comprising a compound of the present invention (or a pharmaceutically acceptable salt, ester or prodrug thereof) and another anti-HCV agent, such as interferon (e.g., alpha-interferon, beta-interferon, consensus interferon, pegylated interferon, or albumin or other conjugated interferon), ribavirin, amantadine, another HCV protease inhibitor, or an HCV polymerase, helicase or internal ribosome entry site inhibitor. The invention also relates to methods of treating an HCV infection in a subject by administering to the subject a pharmaceutical composition of the present invention. The present invention further relates to pharmaceutical compositions comprising the compounds of the present invention, or pharmaceutically acceptable salts, esters, or prodrugs thereof, in combination with a pharmaceutically acceptable carrier or excipient. In one embodiment of the present invention there are disclosed compounds represented by Formulas I, or pharmaceutically acceptable salts, esters, or prodrugs thereof:
Figure imgf000005_0001
Wherein
A is selected from the group consisting of H, R1, -(C=O)-O-Ri, -(C=O)-R2, -CC=O)-NH-R2, or -S(O)2-R1, -S(O)2NHR2;
each Ri is independently selected from the group consisting of: (i) aryl; substituted aryl; heteroaryl; substituted heteroaryl;
(ii) heterocycloalkyl or substituted heterocycloalkyl; (iii) -Ci-C8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl containing O, 1, 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl containing O, 1 , 2, or 3 heteroatoms selected from O, S or N; -C3-C)2 cycloalkyl, or substituted -C3-
Ci2 cycloalkyl; -C3-Ci2 cycloalkenyl, or substituted -C3-Ci2 cycloalkenyl;
Each R2 is independently selected from the group consisting of: (i) hydrogen; (ii) aryl; substituted aryl; heteroaryl; substituted heteroaryl;
(iii) heterocycloalkyl or substituted heterocycloalkyl; (iv) -Ci-C8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl containing O, 1 , 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl containing O, 1 , 2, or 3 heteroatoms selected from O, S or N; -C3-Ci2 cycloalkyl, or substituted -C3-
Ci2 cycloalkyl; -C3-Ci2 cycloalkenyl, or substituted -C3-Ci2 cycloalkenyl; B is selected from the group consisting of H, R1, -(C=O)-O-R1, -(C=O)-R2, -C(=0)-NH-R2; where Rj, R2 are as previously defined;
G is selected from the group consisting of -OH, -NH-S(O)2-R3, -NH-S(O)2NR4R5;
Each R3 is independently selected from the group consisting of :
(i) aryl; substituted aryl; heteroaryl; substituted heteroaryl;
(ii) heterocycloalkyl or substituted heterocycloalkyl; (iii) -C)-C8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl containing O, 1 , 2, or 3 heteroatoms selected from O, S or N, substituted -C1-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl containing O, 1 , 2, or 3 heteroatoms selected from O, S or N; -C3-Ci2 cycloalkyl, or substituted -C3- C12 cycloalkyl; -C3-C12 cycloalkenyl, or substituted -C3-C)2 cycloalkenyl;
each R4 and R5 are independently selected from the group consisting of :
(i) hydrogen;
(ii) aryl; substituted aryl; heteroaryl; substituted heteroaryl;
(iii) heterocycloalkyl or substituted heterocycloalkyl; (iv) -Ci-C8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl containing O, 1 , 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl containing O, 1, 2, or 3 heteroatoms selected from O, S or N; -C3-Ci2 cycloalkyl, or substituted -C3- Ci2 cycloalkyl; -C3-Cj2 cycloalkenyl, or substituted -C3-Ci2 cycloalkenyl;
L is selected from the group consisting of -CH2-, -0-, -S-, or -S(O)2-;
X is absent or is selected from the group consisting of: (1) oxygen; (2) sulfur;
(3) NR4; where R4 is as previously defined above;
(4) -0-NH-; Y is absent or is selected from the group consisting of:
(i) -C(=O)-, -C(=0)-NH-, -S(O)2-, -S(O)2NH-;
(ii) -Ci-C6 alkyl containing 0, 1, 2, or 3 heteroatoms selected from O, S, or N, optionally substituted with one or more substituent selected from halogen, aryl, substituted aryl, heteroaryl, or substituted heteroaryl;
(iii) -C2-C6 alkenyl containing 0, 1, 2, or 3 heteroatoms selected from O, S, or N, optionally substituted with one or more substituent selected from halogen, aryl, substituted aryl, heteroaryl, or substituted heteroaryl;
(iv) -C2-C6 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S, or N, optionally substituted with one or more substituent selected from halogen, aryl, substituted aryl, heteroaryl, or substituted heteroaryl; (v) -C3-Ci2 cycloalkyl, substituted -C3-Ci2 cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl;
Z is selected from the group consisting of aryl, substituted aryl, heteroaryl, substituted heteroaryl, Heterocycloalkyl, substituted heterocycloalkyl;
,z2
so
Or -X-Y-Z taken together to form -J**, wherein each Zi, Z2 are independently selected from the group consisting of: i) hydrogen; ii) aryl; iii) substituted aryl; iv) heteroaryl; v) substituted heteroaryl; vi) heterocyclic or substituted heterocyclic; vii)-Ci-C8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S or N; viii) substituted -Ci-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S or N; ix) -C3-Ci2 cycloalkyl; x) substituted -C3-Ci2 cycloalkyl; xi) -C3-Ci2 cycloalkenyl; xii) substituted -C3-Ci2 cycloalkenyl; xiii) -V-R8, where V is (CO), (CO)O, (CO)NR4, (SO), (SO2), (SO2)NR4; and R4 is as previously defined, R8 is selected from the group consisting of: (1) Hydrogen;
(2) aryl;
(3) substituted aryl;
(4) heteroaryl;
(5) substituted heteroaryl; (6) heterocyclic or substituted heterocyclic;
(7) -Ci-C8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl containing O, 1, 2, or 3 heteroatoms selected from O, S or N;
(8) substituted -Ci-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl containing O, 1, 2, or 3 heteroatoms selected from O, S or N; (9) -C3-Ci2 cycloalkyl;
(10) substituted -C3-Cj2 cycloalkyl;
(11) -C3-Ci2 cycloalkenyl;
(12) substituted -C3-Ci2 cycloalkenyl;
or Zi and Z2 taken together with the carbon atom to which they are attached form a cyclic moiety selected from the group consisting of : substituted or unsubstituted cycloalkyl, cycloalkenyl, or heterocylic; substituted or unsubstituted cycloalkyl, cycloalkenyl, and heterocyclic fused with one or more aryl, substituted aryl, heteroaryl; substituted heteroaryl, heterocyclic or substituted heterocyclic;
j = 0, 1, 2, 3, or 4; k=l, 2, or 3; m = 0, 1, or 2; n = 0, 1, or 2; and
÷=÷ denotes a carbon-carbon single or double bond. DETAILED DESCRIPTION OF THE INVENTION
A first embodiment of the invention is a compound represented by Formula I as described above, or a pharmaceutically acceptable salt, ester or prodrug thereof, alone or in combination with a pharmaceutically acceptable carrier or excipient. Another embodiment of the invention is a compound represented by Formula II:
Figure imgf000009_0001
( H ) or a pharmaceutically acceptable salt, ester or prodrug thereof, alone or in combination with a pharmaceutically acceptable carrier or excipient, where A, B, G, X, Y, and Z are as defined in the previous embodiment. Another embodiment of the invention is a compound represented by Formula III:
Figure imgf000009_0002
( III ) or a pharmaceutically acceptable salt, ester or prodrug thereof, alone or in combination with a pharmaceutically acceptable carrier or excipient; where A, B, G and j are as previously defined in the first embodiment. Wherein R6 is selected from the group consisting of aryl, substituted aryl, heteroaryl, and substituted heteroaryl; J is absent or is selected from the group consisting of O, S, NR5, CO, (CO)NR5, (CO)O, NR5(CO), NH(CO)NH, NR5SO2; wherein R5 are as defined in the first embodiment;
Each R7i, R72, R73 and R74 is independently selected from the group consisting of : (i) hydrogen;
(ii) halogen; (iii) -NO2;
(iv) -CN;
(V) -M-R4, wherein M is absent, or O, S, NH, NR5;
(vi) aryl;
(vϋ) substituted aryl;
(viϋ) heteroaryl;
(ix) substituted heteroaryl;
(x) heterocycloalkyl; and
(Xi) substituted heterocycloalkyl; wherein R4, R5 are as defined previously in the first embodiment.
In one example, A and B are independently selected from the group consisting of H, R1, -(C=O)-O-R1, where R1 is selected from aryl; substituted aryl; heteroaryl; substituted heteroaryl; heterocycloalkyl or substituted heterocycloalkyl; -C1-Cg alkyl, -C2-Cg alkenyl, or -C2-C8 alkynyl containing O, 1, 2, or 3 heteroatoms selected from O, S, or N; substituted -Cj-C8 alkyl, substituted -C2-Cg alkenyl, or substituted -C2-Cg alkynyl containing O, 1, 2, or 3 heteroatoms selected from O, S or N; -C3-Cj2 cycloalkyl, or substituted -C3-C]2 cycloalkyl; -C3-Ci2 cycloalkenyl, or substituted -C3-Ci2 cycloalkenyl; wherein R6 is selected from the group consisting of aryl, substituted aryl, heteroaryl, and substituted heteroaryl. J is absent, j is 1 or 2. G can be -NH-SO2-NR4R5 or -NHSO2- R3, where R3 is selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C3-Ci2 cycloalkyl, -C3-Ci2 cycloalkenyl, substituted -C3-Ci2 cycloalkyl, or substituted -C3-Ci2 cycloalkenyl, and R4 and R5 are each independently selected from hydrogen, -Ci-C8 alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl, substituted -Cj-C8 alkyl, substituted -C2-C8 alkenyl, substituted -C2-C8 alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, - C3-Ci2 cycloalkyl, -C3-Ci2 cycloalkenyl, substituted -C3-Ci2 cycloalkyl, or substituted -C3- Cj2 cycloalkenyl. Each R7J, R72, R73 and R74 is independently selected from the group consisting of hydrogen, halogen, -NO2, -CN, -M-R4; wherein M is absent, or O, S, NH, NR5, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycloalkyl, and substituted heterocycloalkyl, wherein R4, R5 are as defined previously. In still another example, A and B are independently selected from the group consisting of H, R1, where Ri is selected from -Ci-C8 alkyl, -C2-Cg alkenyl, or -C2-C8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S or N; -C3-Ci2 cycloalkyl, or substituted -C3-Ci2 cycloalkyl; -C3-Ci2 cycloalkenyl, or substituted -C3-Cj2 cycloalkenyl; wherein R6 is selected from the group consisting of aryl, substituted aryl, heteroaryl, and substituted heteroaryl. J is absent, j is 1 or 2. G is -NHSO2-R3, where R3 is selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C3-Ci2 cycloalkyl, -C3-Ci2 cycloalkenyl, substituted -C3-Ci2 cycloalkyl, or substituted -C3-Cj2 cycloalkenyl. Each R7i, R72, R73 and R74 is independently selected from the group consisting of hydrogen, halogen, -NO2, -CN.
In still another example, A is H. B is selected from the group consisting of -Ci-C8 alkyl, substituted -Ci-C8 alkyl, -C3-Ci2 cycloalkyl, or substituted -C3-Ci2 cycloalkyl; wherein R6 is selected from the group consisting of aryl, substituted aryl, heteroaryl, and substituted heteroaryl. J is absent, j is 1 or 2.
G is -NHSO2-R3, where R3 is selected from -C3-Ci2 cycloalkyl or substituted -C3- Ci2 cycloalkyl.
Another embodiment of the invention is a compound represented by Formula IV:
Figure imgf000011_0001
( IV )
Wherein each R6, R7i, R72, R73, R74 and J are as defined previously in Formulae III; and A, B, G, j are as defined in the first embodiment. In one example, A and B are independently selected from the group consisting of H, Ri, -(C=O)-O-Ri, where Ri is selected from aryl; substituted aryl; heteroaryl; substituted heteroaryl; heterocycloalkyl or substituted heterocycloalkyl; -Ci-C8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S or N; -C3-C12 cycloalkyl, or substituted -C3-Cj2 cycloalkyl; -C3-Ci2 cycloalkenyl, or substituted -C3-Ci2 cycloalkenyl. Wherein R6 is selected from the group consisting of aryl, substituted aryl, heteroaryl, and substituted heteroaryl. J is absent, j is 1 or 2. G can be -NH-SO2-NR4R5 or -NHSO2-R3, where R3 is selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C3-Ci2 cycloalkyl, -C3-Cj2 cycloalkenyl, substituted -C3-Cj2 cycloalkyl, or substituted -C3-Cj2 cycloalkenyl, and R4 and R5 are each independently selected from hydrogen, -Cj-C8 alkyl, -C2-C8 alkenyl, -C2-C8 alkynyl, substituted -Cj-C8 alkyl, substituted -C2-C8 alkenyl, substituted -C2-C8 alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C3-Cj2 cycloalkyl, -C3-Ci2 cycloalkenyl, substituted -C3-Cj2 cycloalkyl, or substituted -C3-Ci2 cycloalkenyl. Each R7J, R72, R73 and R74 is independently selected from the group consisting of hydrogen, halogen, -NO2, -CN, -M-R4, wherein M is absent, or O, S, NH, NR5, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycloalkyl, and substituted heterocycloalkyl, wherein R4, R5 are as defined previously.
In still another example, A and B are independently selected from the group consisting of H, Rj, where Ri is selected from -Cj-C8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl containing O, 1, 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl containing O, 1, 2, or 3 heteroatoms selected from O, S or N; -C3-Cj2 cycloalkyl, or substituted -C3-Cj2 cycloalkyl; -C3-Ci2 cycloalkenyl, or substituted -C3-Cj2 cycloalkenyl. Wherein R6 is selected from the group consisting of aryl, substituted aryl, heteroaryl, and substituted heteroaryl. J is absent, j is 1 or 2. G is -NHSO2-R3, where R3 is selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C3-Cj2 cycloalkyl, -C3-Ci2 cycloalkenyl, substituted -C3-Cj2 cycloalkyl, or substituted -C3-Cj2 cycloalkenyl. Each R71, R72, R73 and R74 is independently selected from the group consisting of hydrogen, halogen, - NO2, -CN. In still another example, A is H. B is selected from the group consisting of -Ci-C8 alkyl, substituted -Ci-C8 alkyl, -C3-Ci2 cycloalkyl, or substituted -C3-Ci2 cycloalkyl. Wherein R6 is selected from the group consisting of aryl, substituted aryl, heteroaryl, and substituted heteroaryl. J is absent, j is 1 or 2. G is -NHSO2-R3, where R3 is selected from - C3-Cj2 cycloalkyl or substituted -C3-Ci2 cycloalkyl.
Another embodiment of the invention is a compound represented by Formula V:
Figure imgf000013_0001
( V )
Wherein each R71, R72, R73, R74 are as defined previously in Formulae III; and A, B, GJ are as defined in the first embodiment.
In one example, A and B are independently selected from the group consisting of H, Ri, -(C=O)-O-Ri, where R1 is selected from aryl; substituted aryl; heteroaryl; substituted heteroaryl; heterocycloalkyl or substituted heterocycloalkyl; -C1-C8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl containing O, 1, 2, or 3 heteroatoms selected from O, S, or N; substituted -C1-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl containing O, 1, 2, or 3 heteroatoms selected from O, S or N; -C3-C12 cycloalkyl, or substituted -C3-Ci2 cycloalkyl; -C3-Cj2 cycloalkenyl, or substituted -C3-C)2 cycloalkenyl. j is 1 or 2. G can be -NH-SO2-NR4R5 or -NHSO2-R3, where R3 is selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C3-Ci2 cycloalkyl, -C3-Ci2 cycloalkenyl, substituted -C3-Ci2 cycloalkyl, or substituted -C3-Ci2 cycloalkenyl, and R4 and R5 are each independently selected from hydrogen, -Ci-C8 alkyl, -C2-C8 alkenyl, -C2- Cg alkynyl, substituted -Ci-C8 alkyl, substituted -C2-C8 alkenyl, substituted -C2-C8 alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C3-Ci2 cycloalkyl, -C3-Cj2 cycloalkenyl, substituted -C3-Ci2 cycloalkyl, or substituted -C3-Cj2 cycloalkenyl. Each R7i, R72, R73 and R74 is independently selected from the group consisting of hydrogen, halogen, -NO2, -CN, -M-R4, wherein M is absent, or O, S, NH, NR5, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycloalkyl, and substituted heterocycloalkyl, wherein R4, R5 are as defined previously.
In still another example, A and B are independently selected from the group consisting of H, Ri, where Ri is selected from -Cj-C8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S, or N; substituted -Cj-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl containing 0, 1 , 2, or 3 heteroatoms selected from O, S or N; -C3-Cj2 cycloalkyl, or substituted -C3-Cn cycloalkyl; -C3-Cj2 cycloalkenyl, or substituted -C3-Ci2 cycloalkenyl. j is 1 or 2. G is -NHSO2-R3, where R3 is selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C3-Ci2 cycloalkyl, -C3-Cj2 cycloalkenyl, substituted -C3-Cj2 cycloalkyl, or substituted -C3-Ci2 cycloalkenyl. Each R7J, R72, R73 and R74 is independently selected from the group consisting of hydrogen, halogen, -NO2, -CN.
In still another example, A is H. B is selected from the group consisting of -Cj-C8 alkyl, substituted -Cj-C8 alkyl, -C3-Cj2 cycloalkyl, or substituted -C3-Cj2 cycloalkyl. j is 1 or 2. G is -NHSO2-R3, where R3 is selected from -C3-Ci2 cycloalkyl or substituted -C3-Cj2 cycloalkyl. Each R7J, R72, R73 and R74 is independently selected from the group consisting of hydrogen, halogen.
Another embodiment of the invention is a compound represented by Formula VI:
Figure imgf000014_0001
( VI)
Wherein Zl, Z2 and A, B, G, j are as defined in the first embodiment. In one example, Zl and Z2 are independently selected from the group consisting of hydrogen, aryl, substituted aryl, heteroaryl, and substituted heteroaryl, or Z1 and Z2 taken together with the carbon atom to which they are attached form a cyclic moiety selected from the group consisting of substituted or unsubstiruted cycloalkyl, cycloalkenyl, or heterocylic, substituted or unsubstituted cycloalkyl, cycloalkenyl, and heterocyclic fused with one or more aryl, substituted aryl, heteroaryl, and substituted heteroaryl. A and B are independently selected from the group consisting of H, Ri, where Ri is selected from -Ci- C8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-Cg alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl containing 0, 1, 2, or 3 heteroatoms selected from O, S or N; -C3-Cj2 cycloalkyl, or substituted -C3-C12 cycloalkyl; -C3-Ci2 cycloalkenyl, or substituted -C3-Ci2 cycloalkenyl. j is 1 or 2. G is -NHSO2-R3, where R3 is selected from aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, -C3-Ci2 cycloalkyl, -C3-Ci2 cycloalkenyl, substituted -C3-Ci2 cycloalkyl, or substituted -C3-Ci2 cycloalkenyl.
In another example,
Figure imgf000015_0001
A and B are selected from the group consisting of hydrogen, -Ci-C8 alkyl, substituted -Ci-C8 alkyl, -C3-Ci2 cycloalkyl, or substituted -C3-Ci2 cycloalkyl. j is 1 or 2. G is -NHSO2-R3, where R3 is selected from -C3- Ci2 cycloalkyl or substituted -C3-Ci2 cycloalkyl.
Representative compounds of the invention include, but are not limited to, the following compounds (Table 1) according to Formula VII, wherein A, B, Q and G are delineated in Table 1 below for each example:
Figure imgf000015_0002
( VII) TABLE 1
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
The present invention also features pharmaceutical compositions comprising a compound of the present invention, or a pharmaceutically acceptable salt, ester or prodrug thereof.
Compounds of the present invention can be administered as the sole active pharmaceutical agent, or used in combination with one or more agents to treat or prevent hepatitis C infections or the symptoms associated with HCV infection. Other agents to be administered in combination with a compound or combination of compounds of the invention include therapies for disease caused by HCV infection that suppresses HCV viral replication by direct or indirect mechanisms. These include agents such as host immune modulators (for example, interferon-alpha, pegylated interferon-alpha, interferon-beta, interferon-gamma, CpG oligonucleotides and the like), or antiviral compounds that inhibit host cellular functions such as inosine monophosphate dehydrogenase (for example, ribavirin and the like). Also included are cytokines that modulate immune function. Also included are vaccines comprising HCV antigens or antigen adjuvant combinations directed against HCV. Also included are agents that interact with host cellular components to block viral protein synthesis by inhibiting the internal ribosome entry site (IRES) initiated translation step of HCV viral replication or to block viral particle maturation and release with agents targeted toward the viroporin family of membrane proteins such as, for example, HCV P7 and the like. Other agents to be administered in combination with a compound of the present invention include any agent or combination of agents that inhibit the replication of HCV by targeting proteins of the viral genome involved in the viral replication. These agents include but are not limited to other inhibitors of HCV RNA dependent RNA polymerase such as, for example, nucleoside type polymerase inhibitors described in WOOl 90121(A2), or U.S. Pat. No. 6,348,587Bl or WO0160315 or WOO 132153 or non-nucleoside inhibitors such as, for example, benzimidazole polymerase inhibitors described in EP 1162196Al or WO0204425 or inhibitors of HCV protease such as, for example, peptidomimetic type inhibitors such as BILN2061 and the like or inhibitors of HCV helicase.
Other agents to be administered in combination with a compound of the present invention include any agent or combination of agents that inhibit the replication of other viruses for co-infected individuals. These agent include but are not limited to therapies for disease caused by hepatitis B (HBV) infection such as, for example, adefovir, lamivudine, and tenofovir or therapies for disease caused by human immunodeficiency virus (HIV) infection such as, for example, protease inhibitors: ritonavir, lopinavir, indinavir, nelfinavir, saquinavir, amprenavir, atazanavir, tipranavir, TMC-114, fosamprenavir; reverse transcriptase inhibitors: zidovudine, lamivudine, didanosine, stavudine, tenofovir, zalcitabine, abacavir, efavirenz, nevirapine, delavirdine, TMC-125; integrase inhibitors: L- 870812, S- 1360, or entry inhibitors: enfuvirtide (T-20), T- 1249.
Accordingly, one aspect of the invention is directed to a method for treating or preventing an infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment one or more agents selected from the group consisting of a host immune modulator and a second antiviral agent, or a combination thereof, with a therapeutically effective amount of a compound or combination of compounds of the invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, prodrug, salt of a prodrug, or combination thereof. Examples of the host immune modulator are, but not limited to, interferon-alpha, pegylated-interferon-alpha, interferon-beta, interferon-gamrna, a cytokine, a vaccine, and a vaccine comprising an antigen and an adjuvant, and said second antiviral agent inhibits replication of HCV either by inhibiting host cellular functions associated with viral replication or by targeting proteins of the viral genome.
Further aspect of the invention is directed to a method of treating or preventing infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment an agent or combination of agents that treat or alleviate symptoms of HCV infection including cirrhosis and inflammation of the liver, with a therapeutically effective amount of a compound or combination of compounds of the invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, prodrug, salt of a prodrug, or combination thereof. Yet another aspect of the invention provides a method of treating or preventing infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment one or more agents that treat patients for disease caused by hepatitis B (HBV) infection, with a therapeutically effective amount of a compound or a combination of compounds of the invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, prodrug, salt of a prodrug, or combination thereof. An agent that treats patients for disease caused by hepatitis B (HBV) infection may be for example, but not limited thereto, L- deoxythymidine, adefovir, lamivudine or tenfovir, or any combination thereof. Example of the RNA-containing virus includes, but not limited to, hepatitis C virus (HCV).
Another aspect of the invention provides a method of treating or preventing infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment one or more agents that treat patients for disease caused by human immunodeficiency virus (HIV) infection, with a therapeutically effective amount of a compound or a combination of compounds of the invention, or a pharmaceutically acceptable salt, stereoisomer, tautomer, prodrug, salt of a prodrug, or combination thereof. The agent that treats patients for disease caused by human immunodeficiency virus (HIV) infection may include, but is not limited thereto, ritonavir, lopinavir, indinavir, nelfmavir, saquinavir, amprenavir, atazanavir, tipranavir, TMC-1 14, fosamprenavir, zidovudine, lamivudine, didanosine, stavudine, tenofovir, zalcitabine, abacavir, efavirenz, nevirapine, delavirdine, TMC-125, L-870812, S-1360, enfuvirtide (T-20) or T-1249, or any combination thereof. Example of the RNA-containing virus includes, but not limited to, hepatitis C virus (HCV). In addition, the present invention provides the use of a compound or a combination of compounds of the invention, or a therapeutically acceptable salt form, stereoisomer, or tautomer, prodrug, salt of a prodrug, or combination thereof, and one or more agents selected from the group consisting of a host immune modulator and a second antiviral agent, or a combination thereof, to prepare a medicament for the treatment of an infection caused by an RNA-containing virus in a patient, particularly hepatitis C virus. Examples of the host immune modulator are, but not limited to, interferon-alpha, pegylated- interferon-alpha, interferon-beta, interferon-gamma, a cytokine, a vaccine, and a vaccine comprising an antigen and an adjuvant, and said second antiviral agent inhibits replication of HCV either by inhibiting host cellular functions associated with viral replication or by targeting proteins of the viral genome.
When used in the above or other treatments, combination of compound or compounds of the invention, together with one or more agents as defined herein above, can be employed in pure form or, where such forms exist, in pharmaceutically acceptable salt form, prodrug, salt of a prodrug, or combination thereof. Alternatively, such combination of therapeutic agents can be administered as a pharmaceutical composition containing a therapeutically effective amount of the compound or combination of compounds of interest, or their pharmaceutically acceptable salt form, prodrugs, or salts of the prodrug, in combination with one or more agents as defined hereinabove, and a pharmaceutically acceptable carrier. Such pharmaceutical compositions can be used for inhibiting the replication of an RNA-containing virus, particularly Hepatitis C virus (HCV), by contacting said virus with said pharmaceutical composition. In addition, such compositions are useful for the treatment or prevention of an infection caused by an RNA-containing virus, particularly Hepatitis C virus (HCV).
Hence, further aspect of the invention is directed to a method of treating or preventing infection caused by an RNA-containing virus, particularly a hepatitis C virus (HCV), comprising administering to a patient in need of such treatment a pharmaceutical composition comprising a compound or combination of compounds of the invention or a pharmaceutically acceptable salt, stereoisomer, or tautomer, prodrug, salt of a prodrug, or combination thereof, one or more agents as defined hereinabove, and a pharmaceutically acceptable carrier.
When administered as a combination, the therapeutic agents can be formulated as separate compositions which are given at the same time or within a predetermined period of time, or the therapeutic agents can be given as a single unit dosage form.
Antiviral agents contemplated for use in such combination therapy include agents (compounds or biologicals) that are effective to inhibit the formation and/or replication of a virus in a mammal, including but not limited to agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of a virus in a mammal. Such agents can be selected from another anti-HCV agent; an HIV inhibitor; an HAV inhibitor; and an HBV inhibitor. Other anti-HCV agents include those agents that are effective for diminishing or preventing the progression of hepatitis C related symptoms or disease. Such agents include but are not limited to immunomodulatory agents, inhibitors of HCV NS3 protease, other inhibitors of HCV polymerase, inhibitors of another target in the HCV life cycle and other anti-HCV agents, including but not limited to ribavirin, amantadine, levovirin and viramidine.
Immunomodulatory agents include those agents (compounds or biologicals) that are effective to enhance or potentiate the immune system response in a mammal. Immunomodulatory agents include, but are not limited to, inosine monophosphate dehydrogenase inhibitors such as VX-497 (merimepodib, Vertex Pharmaceuticals), class I interferons, class II interferons, consensus interferons, asialo-interferons pegylated interferons and conjugated interferons, including but not limited to interferons conjugated with other proteins including but not limited to human albumin. Class I interferons are a group of interferons that all bind to receptor type I, including both naturally and synthetically produced class I interferons, while class II interferons all bind to receptor type II. Examples of class I interferons include, but are not limited to, [alpha]-, [beta]-, [delta]-, [omega]-, and [tau] -interferons, while examples of class II interferons include, but are not limited to, [gamma] -interferons.
Inhibitors of HCV NS3 protease include agents (compounds or biologicals) that are effective to inhibit the function of HCV NS3 protease in a mammal. Inhibitors of HCV NS3 protease include, but are not limited to, those compounds described in WO 99/07733, WO 99/07734, WO 00/09558, WO 00/09543, WO 00/59929, WO 03/064416, WO 03/064455, WO 03/064456, WO 2004/030670, WO 2004/037855, WO 2004/039833, WO 2004/101602, WO 2004/101605, WO 2004/103996, WO 2005/028501 , WO 2005/070955, WO 2006/000085, WO 2006/007700 and WO 2006/007708 (all by Boehringer Ingelheim), WO 02/060926, WO 03/053349, WO03/099274, WO 03/099316, WO 2004/032827, WO 2004/043339, WO 2004/094452, WO 2005/046712, WO 2005/051410, WO 2005/054430 (all by BMS), WO 2004/072243, WO 2004/093798, WO 2004/113365, WO 2005/010029 (all by Enanta), WO 2005/037214 (Intermune) and WO 2005/051980 (Schering), and the candidates identified as VX-950, ITMN-191 and SCH 503034.
Inhibitors of HCV polymerase include agents (compounds or biologicals) that are effective to inhibit the function of an HCV polymerase. Such inhibitors include, but are not limited to, non-nucleoside and nucleoside inhibitors of HCV NS5B polymerase. Examples of inhibitors of HCV polymerase include but are not limited to those compounds described in: WO 02/04425, WO 03/007945, WO 03/010140, WO 03/010141 , WO 2004/064925, WO 2004/065367, WO 2005/080388 and WO 2006/007693 (all by Boehringer Ingelheim), WO 2005/049622 (Japan Tobacco), WO 2005/014543 (Japan Tobacco),WO 2005/012288 (Genelabs), WO 2004/087714 (IRBM), WO 03/101993 (Neogenesis), WO 03/026587 (BMS), WO 03/000254 (Japan Tobacco), and WO 01/47883 (Japan Tobacco), and the clinical candidates XTL-2125, HCV 796, R-1626 and NM 283.
Inhibitors of another target in the HCV life cycle include agents (compounds or biologicals) that are effective to inhibit the formation and/or replication of HCV other than by inhibiting the function of the HCV NS3 protease. Such agents may interfere with either host or HCV viral mechanisms necessary for the formation and/or replication of HCV. Inhibitors of another target in the HCV life cycle include, but are not limited to, entry inhibitors, agents that inhibit a target selected from a helicase, a NS2/3 protease and an internal ribosome entry site (IRES) and agents that interfere with the function of other viral targets including but not limited to an NS5A protein and an NS4B protein. It can occur that a patient may be co-infected with hepatitis C virus and one or more other viruses, including but not limited to human immunodeficiency virus (HIV), hepatitis A virus (HAV) and hepatitis B virus (HBV). Thus also contemplated is combination therapy to treat such co-infections by co-administering a compound according to the present invention with at least one of an HIV inhibitor, an HAV inhibitor and an HBV inhibitor. According to another embodiment, the pharmaceutical compositions of the present invention may further comprise another anti-viral, anti-bacterial, anti-fungal or anti-cancer agent, or an immune modulator, or another thearapeutic agent.
According to still another embodiment, the present invention includes methods of treating viral infection such as, but not limited to, hepatitis C infections in a subject in need of such treatment by administering to said subject an effective amount of a compound of the present invention or a pharmaceutically acceptable salt, ester, or prodrug thereof.
According to a further embodiment, the present invention includes methods of treating hepatitis C infections in a subject in need of such treatment by administering to said subject an anti-HCV virally effective amount or an inhibitory amount of a pharmaceutical composition of the present invention.
An additional embodiment of the present invention includes methods of treating biological samples by contacting the biological samples with the compounds of the present invention.
Yet a further aspect of the present invention is a process of making any of the compounds delineated herein employing any of the synthetic means delineated herein.
Definitions Listed below are definitions of various terms used to describe this invention. These definitions apply to the terms as they are used throughout this specification and claims, unless otherwise limited in specific instances, either individually or as part of a larger group.
The term "viral infection" refers to the introduction of a virus into cells or tissues, e.g., hepatitis C virus (HCV). In general, the introduction of a virus is also associated with replication. Viral infection may be determined by measuring virus antibody titer in samples of a biological fluid, such as blood, using, e.g., enzyme immunoassay. Other suitable diagnostic methods include molecular based techniques, such as RT-PCR, direct hybrid capture assay, nucleic acid sequence based amplification, and the like. A virus may infect an organ, e.g., liver, and cause disease, e.g., hepatitis, cirrhosis, chronic liver disease and hepatocellular carcinoma.
The term "anti-cancer agent" refers to a compound or drug capable of preventing or inhibiting the advancement of cancer. Examples of such agents include cis-platin, actinomycin D, doxorubicin, vincristine, vinblastine, etoposide, amsacrine, mitoxantrone, tenipaside, taxol, colchicine, cyclosporin A, phenothiazines or thioxantheres.
The term "anti-fungal agent" shall used to describe a compound which may be used to treat a fungus infection other than 3-AP, 3-AMP or prodrugs of 3-AP and 3-AMP according to the present invention. Anti-fungal agents according to the present invention include, for example, terbinafine, fluconazole, itraconazole, posaconazole, clotrimazole, griseofulvin, nystatin, tolnaftate, caspofungin, amphotericin B, liposomal amphotericin B, and amphotericin B lipid complex.
The term "antibacterial agent" refers to both naturally occurring antibiotics produced by microorganisms to suppress the growth of other microorganisms, and agents synthesized or modified in the laboratory which have either bactericidal or bacteriostatic activity, e.g., β-lactam antibacterial agents, glycopeptides, macrolides, quinolones, tetracyclines, and aminoglycosides. In general, if an antibacterial agent is bacteriostatic, it means that the agent essentially stops bacterial cell growth (but does not kill the bacteria); if the agent is bacteriocidal, it means that the agent kills the bacterial cells (and may stop growth before killing the bacteria).
The term "immune modulator" refers to any substance meant to alter the working of the humoral or cellular immune system of a subject. Such immune modulators include inhibitors of mast cell-mediated inflammation, interferons, interleukins, prostaglandins, steroids, corticosteroids, colony-stimulating factors, chemotactic factors, etc.
According to yet another embodiment, the pharmaceutical compositions of the present invention may further comprise inhibitor(s) of other targets in the HCV life cycle, including, but not limited to, helicase, polymerase, metalloprotease, and internal ribosome entry site (IRES). The term "Ci-C6 alkyl," or "Ci-C8 alkyl," as used herein, refer to saturated, straight- or branched-chain hydrocarbon radicals containing between one and six, or one and eight carbon atoms, respectively. Examples of Ci-C6 alkyl radicals include, but are not limited to, methyl, ethyl, propyl, isopropyl, «-butyl, tert-butyl, neopentyl, n-hexyl radicals; and examples of Ci-C8 alkyl radicals include, but are not limited to, methyl, ethyl, propyl, isopropyl, «-butyl, tert-butyl, neopentyl, n-hexyl, heptyl, octyl radicals.
The term "C2-C6 alkenyl," or "C2-C8 alkenyl," as used herein, denote a monovalent group derived from a hydrocarbon moiety by the removal of a single hydrogen atom wherein the hydrocarbon moiety has at least one carbon-carbon double bond and contains from two to six, or two to eight carbon atoms, respectively. Alkenyl groups include, but are not limited to, for example, ethenyl, propenyl, butenyl, 1 -methyl -2 -buten-1-yl, heptenyl, octenyl and the like.
The term "C2-C6 alkynyl," or "C2-C8 alkynyl," as used herein, denote a monovalent group derived from a hydrocarbon moiety by the removal of a single hydrogen atom wherein the hydrocarbon moiety has at least one carbon-carbon triple bond and contains from two to six, or two to eight carbon atoms, respectively. Representative alkynyl groups include, but are not limited to, for example, ethynyl, 1-propynyl, 1-butynyl, heptynyl, octynyl and the like. The term "C3-Cg-cycloalkyl", or "C3-Ci2-cycloalkyl," as used herein, denotes a monovalent group derived from a monocyclic or polycyclic saturated carbocyclic ring compound by the removal of a single hydrogen atom where the saturated carbocyclic ring compound has from 3 ot 8, or from 3 to 12, ring atoms, respectively. Examples of C3-C8- cycloalkyl include, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopentyl and cyclooctyl; and examples of C3-Ci2-cycloalkyl include, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo [2.2.1] heptyl, and bicyclo [2.2.2] octyl.
The term "C3-C8-cycloalkenyl", or "C3-Ci2-cycloalkenyl" as used herein, denote a monovalent group derived from a monocyclic or polycyclic carbocyclic ring compound having at least one carbon-carbon double bond by the removal of a single hydrogen atom where the carbocyclic ring compound has from 3 to 8, or from 3 to 12, ring atoms, respectively. Examples of C3-C8-cycloalkenyl include, but not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, and the like; and examples of C3-Ci 2-cycloalkenyl include, but not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, and the like.
The term "aryl," as used herein, refers to a mono- or bicyclic carbocyclic ring system having one or two aromatic rings including, but not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, idenyl and the like. The term "arylalkyl," as used herein, refers to a Ci-C3 alkyl or Ci-C6 alkyl residue attached to an aryl ring. Examples include, but are not limited to, benzyl, phenethyl and the like.
The term "heteroaryl," as used herein, refers to a mono-, bi-, or tri-cyclic aromatic radical or ring having from five to ten ring atoms of which at least one ring atom is selected from S, O and N; wherein any N or S contained within the ring may be optionally oxidized. Heteroaryl includes, but is not limited to, pyridinyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzooxazolyl, quinoxalinyl, and the like. The term "heteroarylalkyl," as used herein, refers to a Ci-C3 alkyl or Ci-C6 alkyl residue residue attached to a heteroaryl ring. Examples include, but are not limited to, pyridinylmethyl, pyrimidinylethyl and the like. The term "heterocycloalkyl," as used herein, refers to a non-aromatic 3-, 4-, 5-, 6- or 7-membered ring or a bi- or tri-cyclic group fused system, where (i) each ring contains between one and three heteroatoms independently selected from oxygen, sulfur and nitrogen, (ii) each 5-membered ring has 0 to 1 double bonds and each 6-membered ring has 0 to 2 double bonds, (iii) the nitrogen and sulfur heteroatoms may optionally be oxidized, (iv) the nitrogen heteroatom may optionally be quaternized, and (iv) any of the above rings may be fused to a benzene ring. Representative heterocycloalkyl groups include, but are not limited to, [l,3]dioxolane, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, and tetrahydrofuryl.
The term "substituted" as used herein, refers to independent replacement of one, two, or three or more of the hydrogen atoms thereon with substituents including, but not limited to, -F, -Cl, -Br, -I, -OH, protected hydroxy, -NO2, -CN, -NH2, protected amino, -NH -d-Cπ-alkyl, -NH -C2-C12-alkenyl, -NH -C2-C12-alkenyl, -NH -C3-C12-cycloalkyl, -NH - aryl, -NH -heteroaryl, -NH -heterocycloalkyl, -dialkylamino, -diarylamino, - diheteroarylamino, -O-Ci-Ci2-alkyl, -O-C2-Ci2-alkenyl, -O-C2-Ci2-alkenyl, -0-C3-Ci2- cycloalkyl, -O-aryl, -O-heteroaryl, -O-heterocycloalkyl, -C(O)- CrC12-alkyl, -C(O)- C2- C12-alkenyl, -C(O)- C2-C12-alkenyl, -C(O)-C3-Ci2-cycloalkyl, -C(O)-aryl, -C(O)-heteroaryl, -C(O)-heterocycloalkyl, -CONH2, -CONH- C,-Ci2-alkyl, -CONH- C2-C12-alkenyl, -CONH- C2-Ci2-alkenyl, -CONH-C3-C,2-cycloalkyl, -CONH-aryl, -CONH-heteroaryl, -CONH- heterocycloalkyl, -OCO2- Ci-C12-alkyl, -OCO2- C2-C,2-alkenyl, -OCO2- C2-C,2-alkenyl, - OCO2-C3-Ci2-cycloalkyl, -OCO2-aryl, -OCO2-heteroaryl, -OC02-heterocycloalkyl, - OCONH2, -OCONH- C,-Ci2-alkyl, -OCONH- C2-C12-alkenyl, -OCONH- C2-C,2-alkenyl, - OCONH- C3-C12-cycloalkyl, -OCONH- aryl, -OCONH- heteroaryl, -OCONH- heterocycloalkyl, -NHC(O)- C,-C12-alkyl, -NHC(O)-C2-C 12-alkenyl, -NHC(O)-C2-C12- alkenyl, -NHC(O)-C3-C 12-cycloalkyl, -NHC(0)-aryl, -NHC(O)-heteroaryl, -NHC(O)- heterocycloalkyl, -NHCO2- Ci-C12-alkyl, -NHCO2- C2-C]2-alkenyl, -NHCO2- C2-Ci2- alkenyl, -NHCO2- C3-C)2-cycloalkyl, -NHCO2- aryl, -NHCO2- heteroaryl, -NHCO2- heterocycloalkyl, -NHC(O)NH2, -NHC(O)NH- Ci-C)2-alkyl, -NHC(O)NH-C2-Ci2-alkenyl, -NHC(O)NH-C2-C 12-alkenyl, -NHC(O)NH-C3-Ci2-cycloalkyl, -NHC(O)NH-aryl, -
NHC(O)NH-heteroaryl, -NHC(O)NH-heterocycloalkyl, NHC(S)NH2, -NHC(S)NH- Ci-Ci2- alkyl, -NHC(S)NH-C2-C i2-alkenyl, -NHC(S)NH-C2-C,2-alkenyl, -NHC(S)NH-C3-C12- cycloalkyl, -NHC(S)NH-aryl, -NHC(S)NH-heteroaryl, -NHC(S)NH-heterocycloalkyl, - NHC(NH)NH2, -NHC(NH)NH- d-C12-alkyl, -NHC(NH)NH-C2-C 12-alkenyl, - NHC(NH)NH-C2-C i2-alkenyl, -NHC(NH)NH-C3-C π-cycloalkyl, -NHC(NH)NH-aryl, - NHC(NH)NH-heteroaryl, -NHC(NH)NH-heterocycloalkyl, -NHC(NH)-C1 -C I2-alkyl, - NHC(NH)-C2-C 12-alkenyl, -NHC(NH)-C2-C12-alkenyl, -NHC(NH)-C3-C 12-cycloalkyl, - NHC(NH)-aryl, -NHC(NH)-heteroaryl, -NHC(NH)-heterocycloalkyl, -C(NH)NH-Ci-Ci2- alkyl, -C(NH)NH-C2-C iralkenyl, -C(NH)NH-C2-C 12-alkenyl, -C(NH)NH-C3-C2- cycloalkyl, -C(NH)NH-aryl, -C(NH)NH-heteroaryl, -C(NH)NH-heterocycloalkyl, -S(O)- Ci-C]2-alkyl, - S(O)-C2-Ci2-alkenyl, - S(O)-C2-C i2-alkenyl, - S(O)-C3-C 12-cycloalkyl, - S(O)-aryl, - S(O)-heteroaryl, - S(O)-heterocycloalkyl -SO2NH2, -SO2NH- Ci-Ci2-alkyl, - SO2NH- C2-C12-alkenyl, -SO2NH- C2-C,2-alkenyl, -SO2NH- C3-C,2-cycloalkyl, -SO2NH- aryl, -SO2NH- heteroaryl, -SO2NH- heterocycloalkyl, -NHSO2-Ci-C,2-alkyl, -NHSO2-C2- Ci2-alkenyl, - NHSO2-C2-Ci2-alkenyl, -NHSO2-C3-C12-cycloalkyl, -NHSO2-aryl, -NHSO2- heteroaryl, -NHSO2-heterocycloalkyl, -CH2NH2, -CH2SO2CH3, -aryl, -arylalkyl, - heteroaryl, -heteroarylalkyl, -heterocycloalkyl, -C3-Ci2-cycloalkyl, polyalkoxyalkyl, polyalkoxy, -methoxymethoxy, -methoxyethoxy, -SH, -S-Ci-Ci2-alkyl, -S-C2-Ci2-alkenyl, - S-C2-Ci2-alkenyl, -S-C3-Ci2-cycloalkyl, -S-aryl, -S-heteroaryl, -S-heterocycloalkyl, methylthiomethyl, or -L'-R', wherein L' is Ci-C6alkylene, C2-C6alkenylene or C2- C6alkynylene, and R' is aryl, heteroaryl, heterocyclic, C3-Ci2cycloalkyl or C3- C2cycloalkenyl. It is understood that the aryls, heteroaryls, alkyls, and the like can be further substituted. In some cases, each substituent in a substituted moiety is additionally optionally substituted with one or more groups, each group being independently selected from -F, -Cl, -Br, -I, -OH, -NO2, -CN, or -NH2.
In accordance with the invention, any of the aryls, substituted aryls, heteroaryls and substituted heteroaryls described herein, can be any aromatic group. Aromatic groups can be substituted or unsubstituted.
It is understood that any alkyl, alkenyl, alkynyl, cycloalkyl and cycloalkenyl moiety described herein can also be an aliphatic group, an alicyclic group or a heterocyclic group. An "aliphatic group" is non-aromatic moiety that may contain any combination of carbon atoms, hydrogen atoms, halogen atoms, oxygen, nitrogen or other atoms, and optionally contain one or more units of unsaturation, e.g., double and/or triple bonds. An aliphatic group may be straight chained, branched or cyclic and preferably contains between about 1 and about 24 carbon atoms, more typically between about 1 and about 12 carbon atoms. In addition to aliphatic hydrocarbon groups, aliphatic groups include, for example, polyalkoxyalkyls, such as polyalkylene glycols, polyamines, and polyimines, for example. Such aliphatic groups may be further substituted. It is understood that aliphatic groups may be used in place of the alkyl, alkenyl, alkynyl, alkylene, alkenylene, and alkynylene groups described herein. The term "alicyclic," as used herein, denotes a monovalent group derived from a monocyclic or polycyclic saturated carbocyclic ring compound by the removal of a single hydrogen atom. Examples include, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo [2.2.1] heptyl, and bicyclo [2.2.2] octyl. Such alicyclic groups may be further substituted. The term "heterocyclic" as used herein, refers to a non-aromatic 5-, 6- or 7- membered ring or a bi- or tri-cyclic group fused system, where (i) each ring contains between one and three heteroatoms independently selected from oxygen, sulfur and nitrogen, (ii) each 5-membered ring has 0 to 1 double bonds and each 6-membered ring has 0 to 2 double bonds, (iii) the nitrogen and sulfur heteroatoms may optionally be oxidized, (iv) the nitrogen heteroatom may optionally be quaternized, (iv) any of the above rings may be fused to a benzene ring, and (v) the remaining ring atoms are carbon atoms which may be optionally oxo-substituted. Representative heterocycloalkyl groups include, but are not limited to, [l,3]dioxolane, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, quinoxalinyl, pyridazinonyl, and tetrahydrofuryl. Such heterocyclic groups may be further substituted to give substituted heterocyclic.
It will be apparent that in various embodiments of the invention, the substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, arylalkyl, heteroarylalkyl, and heterocycloalkyl are intended to be monovalent or divalent. Thus, alkylene, alkenylene, and alkynylene, cycloaklylene, cycloalkenylene, cycloalkynylene, arylalkylene, hetoerarylalkylene and heterocycloalkylene groups are to be included in the above definitions, and are applicable to provide the formulas herein with proper valency.
The term "hydroxy activating group", as used herein, refers to a labile chemical moiety which is known in the art to activate a hydroxy group so that it will depart during synthetic procedures such as in a substitution or elimination reactions. Examples of hydroxy activating group include, but not limited to, mesylate, tosylate, triflate, p-nitrobenzoate, phosphonate and the like. The term "activated hydroxy", as used herein, refers to a hydroxy group activated with a hydroxy activating group, as defined above, including mesylate, tosylate, triflate, p- nitrobenzoate, phosphonate groups, for example.
The term "protected hydroxy," as used herein, refers to a hydroxy group protected with a hydroxy protecting group, as defined above, including benzoyl, acetyl, trimethylsilyl, triethylsilyl, and methoxymethyl groups.
The terms "halo" and "halogen," as used herein, refer to an atom selected from fluorine, chlorine, bromine and iodine.
The compounds described herein contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- , or as (D)- or (L)- for amino acids. The present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optical isomers may be prepared from their respective optically active precursors by the procedures described above, or by resolving the racemic mixtures. The resolution can be carried out in the presence of a resolving agent, by chromatography or by repeated crystallization or by some combination of these techniques, which are known to those skilled in the art. Further details regarding resolutions can be found in Jacques, et al., Enantiomers, Racemates, and Resolutions (John Wiley & Sons, 1981). When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included. The configuration of any carbon-carbon double bond appearing herein is selected for convenience only and is not intended to designate a particular configuration unless the text so states; thus a carbon-carbon double bond depicted arbitrarily herein as trans may be cis, trans, or a mixture of the two in any proportion.
The term "subject" as used herein refers to a mammal. A subject therefore refers to, for example, dogs, cats, horses, cows, pigs, guinea pigs, and the like. Preferably the subject is a human. When the subject is a human, the subject may be referred to herein as a patient.
As used herein, the term "pharmaceutically acceptable salt" refers to those salts of the compounds formed by the process of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art.
The term "hydroxy protecting group," as used herein, refers to a labile chemical moiety which is known in the art to protect a hydroxy group against undesired reactions during synthetic procedures. After said synthetic procedure(s) the hydroxy protecting group as described herein may be selectively removed. Hydroxy protecting groups as known in the are described generally in T.H. Greene and P. G., S. M. Wuts, Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, New York (1999). Examples of hydroxy protecting groups include benzyloxycarbonyl, 4-nitrobenzyloxycarbonyl, 4- bromobenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, methoxycarbonyl, tert- butoxycarbonyl, isopropoxycarbonyl, diphenylmethoxycarbonyl, 2,2,2- trichloroethoxycarbonyl, 2-(trimethylsilyl)ethoxycarbonyl, 2-furfuryloxycarbonyl, allyloxycarbonyl, acetyl, formyl, chloroacetyl, trifluoroacetyl, methoxyacetyl, phenoxyacetyl, benzoyl, methyl, t-butyl, 2,2,2-trichloroethyl, 2-trimethylsilyl ethyl, 1,1- dimethyl-2-propenyl, 3-methyl- 3 -butenyl, allyl, benzyl, para- methoxybenzyldiphenylmethyl, triphenylmethyl (trityl), tetrahydrofuryl, methoxymethyl, methylthiomethyl, benzyloxymethyl, 2,2,2-triehloroethoxymethyl, 2- (trimethylsilyl)ethoxymethyl, methanesulfonyl, para-toluenesulfonyl, trimethylsilyl, triethylsilyl, triisopropylsilyl, and the like. Preferred hydroxy protecting groups for the present invention are acetyl (Ac or -C(O)CH3), benzoyl (Bz or -C(O)C6H5), and trimethylsilyl (TMS or-Si(CH3)3).Berge, et al. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66: 1-19 (1977). The salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or separately by reacting the free base function with a suitable organic acid. Examples of pharmaceutically acceptable salts include, but are not limited to, nontoxic acid addition salts e.g., salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include, but are not limited to, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, /?-toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, alkyl having from 1 to 6 carbon atoms, sulfonate and aryl sulfonate.
The term "amino protecting group," as used herein, refers to a labile chemical moiety which is known in the art to protect an amino group against undesired reactions during synthetic procedures. After said synthetic procedure(s) the amino protecting group as described herein may be selectively removed. Amino protecting groups as known in the are described generally in T.H. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, New York (1999). Examples of amino protecting groups include, but are not limited to, t-butoxycarbonyl, 9- fluorenylmethoxycarbonyl, benzyloxycarbonyl, and the like.
As used herein, the term "pharmaceutically acceptable ester" refers to esters of the compounds formed by the process of the present invention which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms. Examples of particular esters include, but are not limited to, formates, acetates, propionates, butyrates, acrylates and ethylsuccinates.
The term "pharmaceutically acceptable prodrugs" as used herein refers to those prodrugs of the compounds formed by the process of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the present invention. "Prodrug", as used herein means a compound, which is convertible in vivo by metabolic means (e.g. by hydrolysis) to afford any compound delineated by the formulae of the instant invention. Various forms of prodrugs are known in the art, for example, as discussed in Bundgaard, (ed.), Design of Prodrugs, Elsevier (1985); Widder, et al. (ed.), Methods in Enzymology, vol. 4, Academic Press (1985); Krogsgaard-Larsen, et al., (ed). "Design and Application of Prodrugs, Textbook of Drug Design and Development, Chapter 5, 113-191 (1991); Bundgaard, et al., Journal of Drug Deliver Reviews, 8:1-38(1992); Bundgaard, J. of Pharmaceutical Sciences, 77:285 et seq. (1988); Higuchi and Stella (eds.) Prodrugs as Novel Drug Delivery Systems, American Chemical Society (1975); and Bernard Testa & Joachim Mayer, "Hydrolysis In Drug And Prodrug Metabolism: Chemistry, Biochemistry And Enzymology," John Wiley and Sons, Ltd. (2002).
The term "acyl" includes residues derived from acids, including but not limited to carboxylic acids, carbamic acids, carbonic acids, sulfonic acids, and phosphorous acids. Examples include aliphatic carbonyls, aromatic carbonyls, aliphatic sulfonyls, aromatic sulfinyls, aliphatic sulfinyls, aromatic phosphates and aliphatic phosphates. Examples of aliphatic carbonyls include, but are not limited to, acetyl, propionyl, 2-fluoroacetyl, butyryl, 2-hydroxy acetyl, and the like.
The term "aprotic solvent," as used herein, refers to a solvent that is relatively inert to proton activity, i.e., not acting as a proton-donor. Examples include, but are not limited to, hydrocarbons, such as hexane and toluene, for example, halogenated hydrocarbons, such as, for example, methylene chloride, ethylene chloride, chloroform, and the like, heterocyclic compounds, such as, for example, tetrahydrofuran and N-methylpyrrolidinone, and ethers such as diethyl ether, bis-methoxymethyl ether. Such solvents are well known to those skilled in the art, and individual solvents or mixtures thereof may be preferred for specific compounds and reaction conditions, depending upon such factors as the solubility of reagents, reactivity of reagents and preferred temperature ranges, for example. Further discussions of aprotic solvents may be found in organic chemistry textbooks or in specialized monographs, for example: Organic Solvents Physical Properties and Methods of Purification. 4th ed., edited by John A. Riddick et al, Vol. II, in the Techniques of Chemistry Series, John Wiley & Sons, NY, 1986. The terms "protogenic organic solvent" or "protic solvent" as used herein, refer to a solvent that tends to provide protons, such as an alcohol, for example, methanol, ethanol, propanol, isopropanol, butanol, t-butanol, and the like. Such solvents are well known to those skilled in the art, and individual solvents or mixtures thereof may be preferred for specific compounds and reaction conditions, depending upon such factors as the solubility of reagents, reactivity of reagents and preferred temperature ranges, for example. Further discussions of protogenic solvents may be found in organic chemistry textbooks or in specialized monographs, for example: Organic Solvents Physical Properties and Methods of Purification, 4th ed., edited by John A. Riddick et al, Vol. II, in the Techniques of Chemistry Series. John Wiley & Sons, NY, 1986.
Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds. The term "stable", as used herein, refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., therapeutic or prophylactic administration to a subject).
The synthesized compounds can be separated from a reaction mixture and further purified by a method such as column chromatography, high pressure liquid chromatography, or recrystallization. As can be appreciated by the skilled artisan, further methods of synthesizing the compounds of the formulae herein will be evident to those of ordinary skill in the art. Additionally, the various synthetic steps may be performed in an alternate sequence or order to give the desired compounds. In addition, the solvents, temperatures, reaction durations, etc. delineated herein are for purposes of illustration only and one of ordinary skill in the art will recognize that variation of the reaction conditions can produce the desired bridged macrocyclic products of the present invention. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations. VCH Publishers (1989); T. W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 2d. Ed., John Wiley and Sons (1991); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995).
The compounds of this invention may be modified by appending various functionalities via any synthetic means delineated herein to enhance selective biological properties. Such modifications are known in the art and include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.
PHARMACEUTICAL COMPOSITIONS The pharmaceutical compositions of the present invention comprise a therapeutically effective amount of a compound of the present invention formulated together with one or more pharmaceutically acceptable carriers. As used herein, the term "pharmaceutically acceptable carrier" means a non-toxic, inert solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. Some examples of materials which can serve as pharmaceutically acceptable carriers are 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 a propylene 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, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator. The pharmaceutical compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), buccally, or as an oral or nasal spray. The pharmaceutical compositions of this invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir, preferably by oral administration or administration by injection. The pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles. In some cases, the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form. The term parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra- articular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl 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, and perfuming agents.
Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.
The injectable formulations can be sterilized, for example, by filtration through a bacterial -retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
In order to prolong the effect of a drug, it is often 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 with 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 drug in biodegradable polymers such as polylactide-polyglycolide. Depending upon 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 which are compatible with body tissues.
Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or: a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form 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 sugar as well as high molecular weight polyethylene glycols and the like.
The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes.
Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, ear drops, eye ointments, powders and solutions are also contemplated as being within the scope of this invention. 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 the compounds 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.
Transdermal patches have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
Antiviral Activity An inhibitory amount or dose of the compounds of the present invention may range from about 0.1 mg/Kg to about 500 mg/Kg, alternatively from about 1 to about 50 mg/Kg. Inhibitory amounts or doses will also vary depending on route of administration, as well as the possibility of co-usage with other agents. According to the methods of treatment of the present invention, viral infections are treated or prevented in a subject such as a human or lower mammal by administering to the subject an anti-hepatitis C virally effective amount or an inhibitory amount of a compound of the present invention, in such amounts and for such time as is necessary to achieve the desired result. An additional method of the present invention is the treatment of biological samples with an inhibitory amount of a compound of composition of the present invention in such amounts and for such time as is necessary to achieve the desired result.
The term "anti-hepatitis C virally effective amount" of a compound of the invention, as used herein, mean a sufficient amount of the compound so as to decrease the viral load in a biological sample or in a subject. As well understood in the medical arts, an anti-hepatitis C virally effective amount of a compound of this invention will be at a reasonable benefit/risk ratio applicable to any medical treatment.
The term "inhibitory amount" of a compound of the present invention means a sufficient amount to decrease the hepatitis C viral load in a biological sample or a subject. It is understood that when said inhibitory amount of a compound of the present invention is administered to a subject it will be at a reasonable benefit/risk ratio applicable to any medical treatment as determined by a physician. The term "biological sample(s)," as used herein, means a substance of biological origin intended for administration to a subject. Examples of biological samples include, but are not limited to, blood and components thereof such as plasma, platelets, subpopulations of blood cells and the like; organs such as kidney, liver, heart, lung, and the like; sperm and ova; bone marrow and components thereof; or stem cells. Thus, another embodiment of the present invention is a method of treating a biological sample by contacting said biological sample with an inhibitory amount of a compound or pharmaceutical composition of the present invention. Upon improvement of a subject's condition, a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level, treatment should cease. The subject may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific inhibitory dose for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts.
The total daily inhibitory dose of the compounds of this invention administered to a subject in single or in divided doses can be in amounts, for example, from 0.01 to 50 mg/kg body weight or more usually from 0.1 to 25 mg/kg body weight. Single dose compositions may contain such amounts or submultiples thereof to make up the daily dose. In general, treatment regimens according to the present invention comprise administration to a patient in need of such treatment from about 10 mg to about 1000 mg of the compound(s) of this invention per day in single or multiple doses. Unless otherwise defined, all technical and scientific terms used herein are accorded the meaning commonly known to one with ordinary skill in the art. All publications, patents, published patent applications, and other references mentioned herein are hereby incorporated by reference in their entirety.
Abbreviations
Abbreviations which have been used in the descriptions of the schemes and the examples that follow are:
ACN for acetonitrile;
BME for 2-mercaptoethanol; BOP for benzotriazol- 1 -yloxy-tris(dimethylamino)phosphonium hexafluorophosphate ;
COD for cyclooctadiene;
DAST for diethylaminosulfur trifluoride;
DABCYL for 6-(N-4'-carboxy-4-(dimethylamino)azobenzene)- aminohexyl- 1 -O-(2-cyanoethyl)-(N,N-diisopropyl)-phosphoramidite;
DCM for dichloromethane;
DIAD for diisopropyl azodicarboxylate;
DIBAL-H for diisobutylaluminum hydride; DIEA for diisopropyl ethylamine; DMAP for N,N-dimethylaminopyridine; DME for ethylene glycol dimethyl ether; DMEM for Dulbecco's Modified Eagles Media; DMF for N,N-dimethyl formamide; DMSO for dimethylsulfoxide;
DUPHOS for
Figure imgf000042_0001
EDANS for 5-(2-Amino-ethylamino)-naphthalene-l -sulfonic acid;
EDCI or EDC for l-(3-diethylaminopropyl)-3-ethylcarbodiirnide hydrochloride; EtOAc for ethyl acetate;
HATU for O (7-Azabenzotriazole-l-yl)-N,N,N',N' - tetramethyluronium hexafluorophosphate ;
Hoveyda's Cat. for DichloroCo-isopropoxyphenylmethylene)
(tricyclohexylphosphine)ruthenium(II); KHMDS is potassium bis(trimethylsilyl) amide;
Ms for mesyl;
NMM for N-4-methylmorpholine;
PyBrOP for Bromo-tri-pyrolidino-phosphonium hexafluorophosphate;
Ph for phenyl; RCM for ring-closing metathesis;
RT for reverse transcription;
RT-PCR for reverse transcription-polymerase chain reaction;
TEA for triethyl amine;
TFA for trifluoroacetic acid; THF for tetrahydrofuran;
TLC for thin layer chromatography;
TPP or PPh3 for triphenylphosphine; tBOC or Boc for tert-butyloxy carbonyl; and
Xantphos for 4,5-Bis-diphenylphosphanyl-9,9-dimethyl-9H-xanthene. Synthetic Methods
The compounds and processes of the present invention will be better understood in connection with the following synthetic schemes that illustrate the methods by which the compounds of the invention may be prepared.
Scheme 1
Figure imgf000043_0001
LiOH H2O
Figure imgf000043_0002
All of the quinoxaline analogs were prepared from the common intermediate 1.-7. The synthesis of compound 1.-7 is outlined in Scheme 1. Deprotection of commercially available Boc-hydroxyproline 1.-1 with HCl in dioxane followed by coupling with acid 1.-2 using HATU, afforded intermediate .1-3. Other amino acid derivatives containing a terminal alkene may be used in place of 1.-2 in order to generate varied macrocyclic structures (for further details see WO/0059929). Hydrolysis of 1-3 with LiOH followed by subsequent peptide coupling with cyclopropyl-containing amine 1.-5 yielded tri-peptide 1.-6. Finally, ring-closing metathesis with a ruthenium-based catalyst gave the desired key intermediate 1.-7 (for further details on ring closing metathesis see recent reviews: Grubbs et al., Ace. Chem. Res., 1995, 28, 446; Shrock et al., Tetrahedron 1999, 55, 8141; Furstner, A. Angew. Chem. Int. Ed. 2000, 39, 3012; Trnka et al., Ace. Chem. Res. 2001, 34, 18; and Hoveyda et al., Chem. Eur. J. 2001, 7, 945). Scheme 2
Figure imgf000044_0001
The quinoxaline and quinoline analogs of the present invention were prepared via several different synthetic routes. The simplest method, shown in Scheme 2, was to condense lH-quinoxalin-2-one analogs (2-2), or Hydroxyquinolines (2-3), where R6, R71, R72, R73, R74 and J are as defined previously, with key intermediate \-l by using Mitsunobu conditions to give compound 2-1. For further details on the Mitsunobu reaction, see O. Mitsunobu, Synthesis 1981, 1-28; D. L. Hughes, Org. React. 29, 1-162 (1983); D. L. Hughes, Organic Preparations and Procedures Int. 28, 127-164 (1996); and J. A. Dodge, S. A. Jones, Recent Res. Dev. Org. Chem. 1, 273-283 (1997).
Scheme 3
Figure imgf000044_0002
The macrocyclic starting material 3-1 was prepared following Scheme- 1 by starting with the commercially available trans-Boc-hydroxyproline. Compounds of Formula 3-3 (the carbamates) were prepared by reacting 3-1 with CDI and isoindoline derivatives 3-2. (Scheme 3) R71, R72, R73 and R74 are as previously defined in Formula I. Scheme 4
Figure imgf000045_0001
4-3
Scheme 4 illustrates the general synthetic method of the Oximyl macrocyclics 4-3. First, the hydroxy group of compound h-7 was converted to a suitable leaving group such as, but not limited to OMs, OTs, OTf, bromide, or iodide. Compound (4-1) was subsequently treated with an aryl Oxime (i.e. compound 4-2) at the presence of a base such as, but not limited to K2CO3, Pyridine, TEA, DBU in a suitable solvent like DMF, DMSO, THF etc. to provide compound 4-3.
Figure imgf000045_0002
Scheme 5 illustrates the oxidation of the p3 nitrogen. Compound 5-1 was subjected to the Boc deprotection procedure, such as, but not limited to hydrochloric acid, to provide the free amino compound, which reacted with aryl aldehyde under reductive amination condition to give compound 5-2. The p3 nitrogen of formula (5-2) was oxidized with H2O2 using Na2WO4 as catalyst. The resulting compound was hydrolyzed to give free amino alcohol 5-3. Alkylated or acylated with appropriate alkyl halide or activated acyl groups (A-X) followed by hydrolysis to give compounds of formula (5-4). The carboxylic acid was treated with sulfonamide to provide compounds of formula (5-5).
Scheme 6 illustrates a method to prepare compounds with substitution groups on the hydroxyl group (i.e. compounds 6-3). Compound 6-1 was prepared by treating the α- bromocarboxylic acid with the hydroxylamine following the procedure described in the literature (J. Org. Chem. 2000, 65(9), 2684-2695). Compound 6-2 was made by: 1) introducing the Q group into hydroxyl proline following the procedure described in schemes 2-4; 2) hydrolysis of the resulting ester followed by coupling with the Pl aminoacid piece; 3) hydrolysis of the resulting ester followed by coupling with the sulfonamide following the procedures described in schemes 1 and 5. Compounds 6-3 was then made by coupling 6-1 and 6-2 followed by metathesis as described in scheme- 1. All references cited herein, whether in print, electronic, computer readable storage media or other form, are expressly incorporated by reference in their entirety, including but not limited to, abstracts, articles, journals, publications, texts, treatises, internet web sites, databases, patents, and patent publications.
Examples
The compounds and processes of the present invention will be better understood in connection with the following examples, which are intended as an illustration only and not to limit the scope of the invention. Various changes and modifications to the disclosed embodiments will be apparent to those skilled in the art and such changes and modifications including, without limitation, those relating to the chemical structures, substituents, derivatives, formulations and/or methods of the invention may be made without departing from the spirit of the invention and the scope of the appended claims.
Example 1. Synthesis of the cyclic peptide precursor:
BocHN UO
Figure imgf000047_0001
Figure imgf000047_0002
IA. To a solution of Boc-L-2-amino-8-nonenoic acid 1-1 (1.36g, 5 mol) and the commercially available c/s-L-hydroxyproline methyl ester 1-2 (1.09g, 6 mmol) in 15 ml DMF, DIEA (4 ml, 4eq.) and HATU (4g, 2eq) were added. The coupling was carried out at O °C over a period of 1 hour. The reaction mixture was diluted with 100 mL EtOAc, and directly washed with 5% citric acid (2x 20 ml), water (2x 20 ml), IM NaHCO3 (4x 20 ml) and brine (2x 10 ml). The organic phase was dried over anhydrous Na2SO4, filtered, and then concentrated in vacuo, affording the dipeptide 1-3 (1.91g, 95.8%) that was identified by HPLC (Retention time = 8.9 min, 30-70%, 90%B), and MS (found 421.37, M+Na+).
IB. Dipeptide 1-3 (1.9Ig) was dissolved in 15 mL of dioxane and 15 mL of 1 N LiOH aqueous solution, and the resulting mixture was stirred at room temperature for 4 hours. The reaction mixture was acidified by 5% citric acid and extracted with 100 mL EtOAc. The organic portion was then washed with water (2x 20 ml), IM NaHCO3 (2x 20 ml) and brine (2x 20 ml). The organic phase was dried over anhydrous Na2SO4, filtered, and then concentrated in vacuo, yielding the free carboxylic acid compound 1-4 (1.79g, 97%), which was used directly without the need for further purification.
1C. To a solution of the free acid obtained above (1.77, 4.64 mmol) in 5 ml DMF, D-β-vinyl cyclopropane amino acid ethyl ester 1-5 (0.95g, 5 mmol), DIEA (4 ml, 4eq.) and HATU (4g, 2eq) were added. The coupling was carried out at 0 0C over a period of 5 hours. The reaction mixture was diluted with 80 mL EtOAc, and washed with 5% citric acid (2x 20 ml), water (2x 20 ml), IM NaHCO3 (4x 20 ml), and brine
(2x 10 ml). The organic phase was dried over anhydrous Na2SO4, filtered, and then concentrated in vacuo. The residue was purified by silica gel flash chromatography using gradient elution with hexanes:EtOAc (5:1-»3:1— »1 :1— »1:2— »1 :5). The linear tripeptide 1-6 was isolated as an oil (1.59g, 65.4%) and identified by HPLC (Retention time = 11.43 min) and MS (found 544.84, M+Na+).
ID. Ring Closing Metathesis (RCM). A solution of the linear tripeptide 1-6 (1.5 Ig, 2.89 mmol) in 200 ml dry DCM was deoxygenated by N2 bubbling. Hoveyda's 1st generation catalyst (5 mol% eq.) was then added as a solid. The reaction was refluxed under N2 atmosphere for 12 hours. The solvent was evaporated and the residue was purified by silica gel flash chromatography using gradient elution with hexanes:EtOAc (9:1-»5:1-»3:1-»1 :1-»1 :2-»1 :5). The cyclic peptide precursor 1 was isolated as a white powder (1.24g, 87%), and identified by HPLC (Retention time = 7.84 min, 30-70%, 90%B), and MS (found 516.28, M+Na+). For further details of the synthetic methods employed to produce the cyclic peptide precursor 1, see WO 00/059929 (2000).
Figure imgf000048_0001
( VII) Example 2. Compound of Formula VII, wherein A = CX °Λ /, B= H, Q=
Figure imgf000049_0001
Figure imgf000049_0002
Step 2A. To a cooled mixture of macrocyclic precursor 1, 3-(thiophen-2-yl)-lH- quinoxalin-2-one E2-1 (1.1 equiv.), and triphenylphosphine (2 equiv.) in THF was added DIAD (2 equiv.) dropwise at O0C. The resulting mixture was held at O0C for 15 min. before being warmed to room temperature. After 18 hours, the mixture was concentrated under vacuum and the residue was purified by chromatography eluting with 60% EtOAc in hexanes to give E2-2 as a clear oil (1.0 g, 95%). MS (ESI) m/z = 704.4 (M+H)+.
H'-NMR [CDCl3, δ (ppm)]: 8.6 (d, IH), 8.0 (d, IH), 7.8 (d, IH), 7.6 (m, 2H), 7.5 (d, 2H), 7.2 (t, IH), 7.0 (brs, IH), 6.0 (brt, IH), 5.5 (m, IH), 5.3 (brd, IH), 5.2 (t, IH), 5.0 (m. IH), 4.6 (brt, IH), 4.1-4.3 (m, 3H), 3.1 (m, IH), 5.3 (m, IH), 2.1-2.3 (m, 2H), 1.3 (brs, 9H), 1.2 (t, 3H).
Step 2B. The title compound of Step 2A (200 mg, 0.28 mmol) was treated with HCl (4 M in dioxane, 3 mL, 12 mmol). The reaction mixture was stirred at room temperature for 1 h until LCMS showed the complete consumption of starting material. The solvent was removed in vacuo. The residue was dissolved in DCM (3 ml). The solvent was removed in vacuo and the residue was used directly in next step.
The residue was dissolved in AcOH (3 mL). The solution was charged with benzylaldehyde (43 μL) and NaBH3CN (34 mg). The resulting mixture was stirred at rt for 18h. The reaction mixture was diluted with 20 mL EtOAc, and washed with water (20 ml), NaHCO3 aq. (10 ml) and brine (10 ml). The organic phase was dried over anhydrous Na2SO4, filtered, and then concentrated in vacuo, purified by column chromatography to afford compound E-2-3 (40 mg, 20%, 2 steps). MS (ESI) m/z = 694.44 (M+H)+.
Step 2C. To a solution of compound E-2-3 (40 mg) in 8 mL MeOH was added Na2WO4 (60 mg) and H2O2 (30wt%, 1 mL). The reaction was carried out at RT over a period of 40 hours. The reaction mixture was diluted with 20 mL EtOAc, and washed with brine (2X10 ml). The organic phase was dried over anhydrous Na2SO4, filtered, and then concentrated in vacuo. The residue was dissolved in MeOH, and then treated with NH20H HCl (40 mg). The reaction mixture was stirred at rt for 2h, and then concentrated in vacuo, purified by column chromatography to afford compound E-2-4. MS (ESI) m/z = 620.39 (M+H)+.
Step 2D. To a solution of compound E-2-4 (13 mg) in 3 ml dry DCM was treated with cyclopentyl chloroformate (3 eqiv.) at the presence of /Pr2NEt (5 eqiv.). The reaction mixture is stirred for 2 hours. The reaction mixture was diluted with 20 mL EtOAc, and washed with NaHCO3 aq. (10 ml) and brine (10 ml). The organic phase was dried over anhydrous Na2SO4, filtered, and then concentrated in vacuo. The residue was dissolved in 3 mL of dioxane and 1 mL of 1 N LiOH aqueous solution, and the resulting mixture was stirred at room temperature for 20 hours. The reaction mixture was acidified by HCl (IM) to pH~3 and extracted with 20 mL EtOAc. The organic phase was dried over anhydrous Na2SO4, filtered, and then concentrated in vacuo. The residue was purified by HPLC (40-90% acetonitrile in water) to yield the title compound E-2-S (5.5 mg, 37% two steps). MS (ESI) m/z = 704.11 (M+H)+.
Example 3. Compound of Formula VII, wherein A = ° / , B= H, Q=
Figure imgf000051_0001
Step 3A: Cyclopropylsulfonyl chloride (1.4g, 10 mmol) was dissolved in 0.5 M ammonia in dioxane (50 ml, 25 mmol) at RT. The reaction was kept at RT for 3 days. The large amount of precipitation was filtered and discarded. The clear filtrate was evaporated in vacuo and the white residue was dried on vacuum for 24 hours to give the cyclopropylsulfonamide (0.88 g, 74%). 1H-NMR (500 MHz, CD3Cl): δ 4.62 (2H, s), 2.59 (IH, m), 1.20 (2H, m), 1.02 (2H, m).
Step 3B: The title compound from Example 2 (2.0 mg) and carbonyldiimidazole (1.0 mg) were dissolved in 0.7 ml anhydrous DMF and the resulting solution was heated to 4O0C for 1 hour. Cyclopropylsulfonamide (1.0 mg) was added to the reaction followed by DBU (1.0 mg). The reaction mixture was stirred at 4O0C for 10 hour. LCMS showed the formation of the desired product. The reaction was cooled down and 10 ml ethyl acetate was added to the solution. The mixture was washed with saturated aqueous NaHCO3 solution, water and brine. The organic layer was dried over anhydrous sodium sulfate. The organic phase was then filtered, concentrated in vacuo and subsequently purified by flash chromatography (ethyl acetate/hexanes 1 :1) to give 1.0 mg (50%) of the title compound. MS (ESI) m/z = 807.31 (M+H)+.
Example 4. Compound of Formula VII, wherein A
Figure imgf000051_0002
G =-OH.
The title compound is prepared following the procedures described in Example 2 by using appropriate chloroformate reagents.
Example 5. Compound of Formula VII, wherein A = ° /,
Figure imgf000051_0003
/ V
G = H V . The title compound is prepared following the procedures described in Example 3 by using the title compound of Example 4.
Example 6. Compound of Formula VII, wherein A = ? , B= H,
Figure imgf000052_0001
G =-
OH.
The title compound is prepared following the procedures described in Example 2 without the chloroformate formation step.
Example 7. Compound of Formula VII, wherein A = ? , B= H, Q=
Figure imgf000052_0002
G
-Vv.
The title compound is prepared following the procedures described in Example 3 by using the title compound of Example 6.
Example 8. Compound of Formula VII, wherein A =
Figure imgf000052_0003
° /, B= H, Q=
G =-0H. The title compound is prepared following the procedures described in Example 2 by using appropriate chloroformate reagents.
Example 9. Compound of Formula VII, wherein A = CX ° Λ /, B= H, Q=
Figure imgf000052_0004
G = H V. The title compound is prepared following the procedures described in Example 3 by using the title compound of Example 8. Example 10. Compound of Formula VII, wherein A = CX °x /, B= H, Q=
Figure imgf000053_0001
The title compound is made by reacting the title compound of Example 1 with
MeO.
Mecr I^-^l rrl-OH under the Mitsunobu conditions described in step 2A, then followed by the procedures described in Example 2.
Example 11. Compound of Formula VII, wherein A = ° /, B= H, Q=
Figure imgf000053_0002
The title compound is prepared following the procedures described in Example 3 by using the title compound of Example 10.
Example 12. Compound of Formula VII, wherein A = CX °x /, B= H, Q=
Figure imgf000053_0003
The title compound is prepared following the procedures described in Example 3 by starting with the title compound of Example 2 and 2-thiophenesulfonamide.
Example 13. Compound of Formula VII, wherein A
Figure imgf000053_0004
H, Q=
Figure imgf000053_0005
The title compound is prepared following the procedures described in Example 3 by starting with the title compound of Example 10 and 2-thiophenesulfonamide. Example 14. Compound of Formula VII, wherein A
Figure imgf000054_0001
B= H, Q=
Figure imgf000054_0002
Step 14A Mesylate formation:
To a solution of the title compound from Example 1 (macrocyclic peptide precursor 1, j=2, 1.0 eqiv.) and DIEA (2.0 eqiv.) in DCM is added Methanesulfonyl chloride
(1.1 eqiv.) slowly at 0 °C. The reaction is kept for 3 hours. EtOAc is then added and followed by washing with water, NaHCO3 aq. solution and brine, respectively. The organic phase is dried over anhydrous Na2SO4 and evaporated, yielding the mesylate compound that was used for next step without further purification.
Step 14B Substitution of the Mesylate:
The mesylate compound from step 14A (1.0 eqiv.),
Figure imgf000054_0003
(1.2 eqiv.), and K2CO3 (2 eqiv.) are dissolved in DMF or DMSO. The resulting reaction mixture is stirred at 40-80°C for 10 hours, cooled and extracted with ethyl acetate. The organic extract is washed with water (2x30ml), and the organic solution is concentrated in vacuo, subsequently purified by column chromatography eluting with 50% ethyl acetate in hexanes to give the product.
The title compound is prepared following the procedures described in Example 2(2B, 2C, 2D) from compound 14B.
Example 15. Compound of Formula VII, wherein A =ux B= H, Q=
Figure imgf000054_0004
The title compound is prepared following the procedures described in Example 3 by using the title compound of Example 14.
Example 16. Compound of Formula VII, wherein A =^ Ό /; β= H, Q=
Figure imgf000055_0001
The title compound is prepared following the procedures described in Example 14 by using appropriate chloroformate reagents.
I o Example 17. Compound of Formula VII, wherein A = ° /, B= H, Q=
Figure imgf000055_0002
The title compound is prepared following the procedures described in Example 3 by using the title compound of Example 16.
r-Λ °
Example 18. Compound of Formula VII, wherein A = ° /, B= H, Q=
Figure imgf000055_0003
, G =-0H.
The title compound is prepared following the procedures described in Example 14 by using appropriate chloroformate reagents.
r-~\ O
Example 19. Compound of Formula VII, wherein A '& V, B= H, Q=
Figure imgf000055_0004
The title compound is prepared following the procedures described in Example 3 by using the title compound of Example 18.
A-I O Example 20. Compound of Formula VII, wherein A = ° /, B= H, Q=
Figure imgf000056_0001
20-1
Step 2OA
Cyclic compound 20-l(j=2) is prepared following the procedures described in Example 1 by starting with commercially available 7><ms-Boc-hydroxyproline, which is condensed with CDI (1.2 eqiv.) in dichloromethane at RT. Once this coupling is complete as confirmed by MS analysis, 4-Fluoro-2, 3-dihydro-lH- isoindole (3 eqiv.) is added and the resulting mixture is stirred overnight. The reaction mixture is diluted with dichloromethane (20 mL) and washed with IN aq. HCl and brine. The organic portion is then dried (Na2SO4), filtered, and concentrated in vacuo. The crude is purified via flash chromatography (silica gel) to afford the corresponding carbamate.
Step 2OB, C, D
The title compound is prepared following the procedures described in Example 2 (2B, 2C, 2D) from compound 20-3.
Examples 21 to Examples 27 below are made following the procedures described in Examples 20 and 3 by starting with the approporiate materials. Example 21. Compound of Formula VII, wherein A = ° /, B= H, Q=
Figure imgf000057_0001
Example 22. Compound of Formula VII, wherein A =
Figure imgf000057_0002
G =-OH.
Example 23. Compound of Formula VII, wherein A = ° /, B= H, Q=
Figure imgf000057_0003
Figure imgf000057_0004
Example 24. Compound of Formula VII, wherein A -0 B= H, Q=
Figure imgf000057_0005
o
Example 25. Compound of Formula VII, wherein A = ° /, B= H, Q=
Figure imgf000057_0006
.
Example 26. Compound of Formula VII, wherein A = Cl °X / , B= H, Q=
Figure imgf000057_0007
Example 27. Compound of Formula VII, wherein A = ° /, B= H, Q=
Figure imgf000058_0001
Example 28. Compound of Formula VII, wherein A = CXX ° /, B= H, Q=
Figure imgf000058_0002
G =-OH.
Figure imgf000058_0003
Step 28A
The title compound 28-3 is made following the procedures described in Example 14 (steps 14A and 14B).
Step 28B, C, D
The title compound is prepared following the procedures described in Example 2 (2B, 2C, 2D) from compound 28-3.
Examples 29 to Examples 32 below are made following the procedures described in Examples 28 and 3 by starting with the approporiate materials.
Example 29. Compound of Formula VII, wherein A =
Figure imgf000058_0004
G = H V. Example 30. Compound of Formula VII,
Figure imgf000059_0001
=-OH.
Example 31. Compound of Formula VII, wherein A =
Figure imgf000059_0002
= / H V V .
Example 32. Compound of Formula VII, wherein A = ° /, B= H, Q=
Figure imgf000059_0003
G = H V .
Example 33. Compound of Formula VII, wherein A = -H,
Figure imgf000059_0004
G = H V.
Step 33A
Figure imgf000059_0005
8A
To a solution of Boc cw-L-hydroxyproline methyl ester (2g, 8.15mmol) 33-1 and Et3N (1.7ml, 12.23mmol) in dichloromethan at 00C was added slowly MsCl (0.7ml, 8.96mmol). The resulting mixture was stirred at room temperature for l~2h, diluted with EtOAc, washed with brine, dried (MgSO4) and concentrated in vacuo to dryness to give crude 33-2 which was directly used in next step. Step 33B
Figure imgf000060_0001
8B
A mixture of the above crude 33-2, 9H-fluoren-9-one oxime (1.8g, 8.97mmol), cesium carbonate (4g, 12.2mmol) and DMF (12ml) was stirred at 5O0C for 2Oh, diluted with EtOAc, washed with brine, dried (MgSO4) and concentrated in vacuo. The residue was purified by silica gel chromatography (Hexane/EtOAc = 9 : 1 to 4 : 1) to afford 33-3 (2.736g).
Step 33C
Figure imgf000060_0002
33-3 33-4
Compound 33-4 was prepared from 1.-3 by the standard hydrolysis reaction as described in the PCT WO 2004113365.
Figure imgf000061_0001
Compound 33-6 was prepared from the standard coupling reaction of 33-4 and 33-5 as described in the PCT WO 2004113365.
Step 33E
Figure imgf000061_0002
To a solution of compound 33-6 (301mg, 0.55mmol) in THF/MeOH (3.4ml-1.7ml) was added aqueous lithium hydroxide (IM, 1.7ml, 1.7mmol). The mixture was stirred at room temperature for 18 hours. Most organic solvents were evaporated in vacuo, and the resulting residue was diluted with water and acidified to pH 5 to 6. The mixture was extracted with EtOAc three times. The combined organic extracts were dried (MgSO4), filtered and concentrated in vacuo to afford 33-7 (-100%). Step 33F
Figure imgf000062_0001
Compound 33-7 (0.55 mmol) and carbonyldiimidazole (134mg, 0.825 mmol) were dissolved in 5 ml of anhydrous DMF and the resulting solution was stirred at 4O0C for 1 hour. Cyclopropylsulfonamide (133mg, l.lmmol) was added to the reaction followed by DBU (1 lOul, 0.715 mmol). The reaction mixture was stirred at 4O0C for 20 hours. The reaction mixture was diluted with ethyl acetate and washed with half- saturated-aqueous NaCl solution three times. The organic layer was dried over anhydrous (MgSO4) and concentrated in vacuo. The residue was purified by silica gel chromatography (Hexans/ EtOAc = 1 : 1 to 0 : 1 then ACOEt/MeOH = 95 : 5 to 90 : 10) to give 33-8 (300mg).
Figure imgf000062_0002
Compound 33-8 (lOOmg, 0.161 mmol) was treated with 4N HCl in 1, 4-dioxane (4ml, 16mmoL). The mixture was stirred at room temperature for an hour, concentrated to dryness to afford HCl salt of 33-9 (-100%). Step 33H
Figure imgf000063_0001
A mixture of compound 33-9 (lOmg, 0.04mmol, prepared according to the procedures described in J. Org. Chem. 2000, 65(9), 2684-2695), compound 33-10 (25mg, 0.24mmol), triethyl amine (17ul, 0.12mmol) and alcohol was stirred at 8O0C for 1 day, colled to rt, concentrated to dryness to afford the crude product 33-11 which was used directly in next step.
Step 331
Figure imgf000063_0002
Figure imgf000063_0003
To a solution of the crude 33-11, oxim core intermediate 33-8 (0.053mmol) and DIPEA (0.09ml, 0.516mmolmmol) in DMF (1.5ml) at 0 0C was added HATU (26mg, 0.068mmol). The mixture was stirred at room temperature for 18h, diluted with EtOAc and washed with half-sat.-aq. NaCl four times. The organic phase was dried over anhydrous MgSO4, filtered, and then concentrated in vacuo. The residue was purified by preparative HPLC to afford tcompound 33-12A (8mg), MS (ESI): m/z 760.57 (M+H) and compound 33-12B(8mg), MS (ESI): m/z 760.58 (M+H).
Step 33J
Figure imgf000064_0001
Example 33
A solution of compound 33-12 (4mg, 0.005mmol) in 3ml dry DCM was deoxygenated by N2 bubbling. Hoveyda's 1st generation catalyst (1.5mg.) was then added as a solid. The reaction mixture was stirred at 4OC for 12h. The solvent was evaporated and the residue was purified by preparative HPLC to give the title compound (lmg), MS(ESI), m/z 732.53 (M+H).
Example 34. Compound of Formula VII, wherein A =-H,
Figure imgf000064_0002
G
/ V
= H ^V
Step 34A
Figure imgf000064_0003
To a mixture of Boc cw-L-hydroxyproline methyl ester 34-1 (1.073g, 4.37mmol), 3- (thiophen-2-yl)-lH-quinoxalin-2-one 34-2 (0.999g, 4.38mmol)) and triphenylphosphine (2.29g, 8.74mmol) in THF at O0C was added dropwise DIAD (1.72ml, 8.7mmol). The resulting mixture was held at 0°C for 15 min. before being warmed to room temperature. After 18 hours, the mixture was concentrated under vacuum and the residue was purified by chromatography (Hexane/EtOAC = 1 :0 to 8:2) to give 34-3 (2.28g).
Step 34B
Figure imgf000065_0001
To a solution of compound 34-3 (2.05g, 4.5mmol) in THF/MeOH (2OmI-IOmI) was added aqueous lithium hydroxide (IM, 10ml, 10mmol).The mixture was stirred at room temperature for 20 hours. Most organic solvents were evaporated in vacuo, and the resulting residue was diluted with water and acidified to pH 5 to 6. The mixture was extracted with EtOAc three times. The combined organic extracts were dried (MgSO4), filtered and concentrated in vacuo to afford 34-4 (1.76g).
Step 34C
Figure imgf000065_0002
To a solution of 34-4 (Ug, 3.85mmol), (IR, 2S)-l-Amino-2-vinyl- cyclopropanecarboxylic acid ethyl ester HCl salt 34-5 (0.74g, 3.86mmol) and DIPEA (2ml, l l .όmmol) in DMF (25ml) at 0 °C was added in portions HATU (1.75g, 4.6mmol). The mixture was stirred at rt for 18h, diluted with EtOAc and washed with half-sat.-aq. NaCl four times. The organic phase was dried over anhydrous MgSO4, filtered, and then concentrated in vacuo. The residue was purified by silica gel chromatography (Hexane/EtOAC = 9 : 1 to 7 : 3) to afford compound 34-6 (l.lg). Step 34D
Figure imgf000066_0001
To a solution of compound 34-6 (0.2 Ig, 0.363mmol) in THF/MeOH (6ml-3ml) was added aqueous lithium hydroxide (IM, 3ml, 3mmol).The mixture was stirred at room temperature for 20 hours. Most organic solvents were evaporated in vacuo, and the resulting residue was diluted with water and acidified to pH 5 to 6. The mixture was extracted with EtOAc three times. The combined organic extracts were dried (MgSO4), filtered and concentrated in vacuo to afford 34-7 (0.205g). MS (ESI): m/e 551.23 (M+H).
Step 34E
Figure imgf000066_0002
Compound 34-7 (175mg, 0.317 mmol) and carbonyldiimidazole (80mg, 0.476 mmol) were dissolved in 3 ml of anhydrous DMF and the resulting solution was stirred at 4O0C for 1 hour. Cyclopropylsulfonamide (77 mg, 0.634 mmol) was added to the reaction followed by DBU (71ul, 0.476 mmol). The reaction mixture was stirred at 4O0C for 20 hour. The reaction mixture was diluted with ethyl acetate and washed with half-saturated-aqueous NaCl solution three times. The organic layer was dried over anhydrous (MgSO4) and concentrated in vacuo. The residue was purified by silica gel chromatography (Hexans/ EtOAc = 1 : 1 to 1 : 2) to give 34-8 (96mg). MS (ESI): m/e 654.26 (M+H).
Step 34F
Figure imgf000067_0001
Compound 34-8 (77mg, 0.118 mmol) was treated with 4N HCl in 1, 4-dioxane (2ml, 8mmoL). The mixture was stirred at room temperature for an hour, concentrated to dryness to affored HCl salt of 34-9 (-100%). MS (ESI): m/e 554.20 (M+H).
Step 34G
Figure imgf000067_0002
Figure imgf000067_0003
Example 34
34-10A
Compound 34- 1OA was prepared from 33-11 and 34-9 according to the same procedure described in the step 331. MS(ESI): 793.41 (M+H). Step 34H
Figure imgf000068_0001
Figure imgf000068_0002
Example 34
34-10A
Compound 34- 1OA was subjected to the same raction conditions as described in step
33J to afford the title compound. MS(ESI): 765.19 (M+H).
The compounds of the present invention exhibit potent inhibitory properties against the HCV NS3 protease. The following examples elucidate assays in which the compounds of the present invention are tested for anti-HCV effects.
Example 35 NS3/NS4a Protease Enzyme Assay
HCV protease activity and inhibition is assayed using an internally quenched fluorogenic substrate. A DABCYL and an EDANS group are attached to opposite ends of a short peptide. Quenching of the EDANS fluorescence by the DABCYL group is relieved upon proteolytic cleavage. Fluorescence was measured with a Molecular Devices Fluoromax (or equivalent) using an excitation wavelength of 355 nm and an emission wavelength of 485 nm.
The assay is run in Corning white half-area 96-well plates (VWR 29444-312 [Corning 3693]) with full-length NS3 HCV protease Ib tethered with NS4A cofactor (final enzyme concentration 1 to 15 nM). The assay buffer is complemented with 10 μM NS4A cofactor Pep 4A (Anaspec 25336 or in-house, MW 1424.8). RET S 1 (Ac- Asp-Glu- ASP(EDANS)-GIu-GIu-AbU-[COO]AIa-SCr-LyS-(D ABCYL)- NH2,.AnaSpec 22991, MW 1548.6) is used as the fluorogenic peptide substrate. The assay buffer contained 50 mM Hepes at pH 7.5, 30 mM NaCl and 10 mM BME. The enzyme reaction is followed over a 30 minutes time course at room temperature in the absence and presence of inhibitors. The peptide inhibitors HCV Inh 1 (Anaspec 25345, MW 796.8) Ac-Asp-Glu-Met- Glu-Glu-Cys-OH, [-200C] and HCV Inh 2 (Anaspec 25346, MW 913.1) Ac- Asp-Glu-Dif-Cha-Cys-OH, were used as reference compounds.
IC50 values were calculated using XLFit in ActivityBase (IDBS) using equation
205: y=A+((B-A)/(l+((C/x)ΛD))).
Example 36 - Cell-Based Replicon Assay
Quantification of HCV replicon RNA in cell lines (HCV Cell Based Assay) Cell lines, including Huh- 11 -7 or Huh 9-13, harboring HCV replicons (Lohmann, et al Science 285:110-113, 1999) are seeded at 5xlO3 cells/well in 96 well plates and fed media containing DMEM (high glucose), 10% fetal calf serum, penicillin- streptomycin and non-essential amino acids. Cells are incubated in a 5% CO2 incubator at 37 0C. At the end of the incubation period, total RNA is extracted and purified from cells using Qiagen Rneasy 96 Kit (Catalog No. 74182). To amplify the
HCV RNA so that sufficient material can be detected by an HCV specific probe (below), primers specific for HCV (below) mediate both the reverse transcription of the HCV RNA and the amplification of the cDNA by polymerase chain reaction (PCR) using the TaqMan One-Step RT-PCR Master Mix Kit (Applied Biosystems catalog no. 4309169). The nucleotide sequences of the RT-PCR primers, which are located in the NS5B region of the HCV genome, are the following: HCV Forward primer "RBNS5bfor" 5 'GCTGCGGCCTGTCGAGCT: HCV Reverse primer "RBNS5Brev": 5 'CAAGGTCGTCTCCGCATAC.
Detection of the RT-PCR product is accomplished using the Applied Biosystems (ABI) Prism 7700 Sequence Detection System (SDS) that detects the fluorescence that is emitted when the probe, which is labeled with a fluorescence reporter dye and a quencher dye, is processed during the PCR reaction. The increase in the amount of fluorescence is measured during each cycle of PCR and reflects the increasing amount of RT-PCR product. Specifically, quantification is based on the threshold cycle, where the amplification plot crosses a defined fluorescence threshold. Comparison of the threshold cycles of the sample with a known standard provides a highly sensitive measure of relative template concentration in different samples (ABI User Bulletin #2 December 11, 1997). The data is analyzed using the ABI SDS program version 1.7. The relative template concentration can be converted to RNA copy numbers by employing a standard curve of HCV RNA standards with known copy number (ABI User Bulletin #2 December 11, 1997).
The RT-PCR product was detected using the following labeled probe: 5 ' FAM-CGAAGCTCCAGGACTGCACGATGCT-TAMRA FAM= Fluorescence reporter dye. TAMRA:=Quencher dye.
The RT reaction is performed at 480C for 30 minutes followed by PCR. Thermal cycler parameters used for the PCR reaction on the ABI Prism 7700 Sequence Detection System are: one cycle at 950C, 10 minutes followed by 35 cycles each of which include one incubation at 950C for 15 seconds and a second incubation for 60
0C for 1 minute.
To normalize the data to an internal control molecule within the cellular RNA, RT- PCR is performed on the cellular messenger RNA glyceraldehydes-3 -phosphate dehydrogenase (GAPDH). The GAPDH copy number is very stable in the cell lines used. GAPDH RT-PCR is performed on the same exact RNA sample from which the HCV copy number is determined. The GAPDH primers and probes, as well as the standards with which to determine copy number, are contained in the ABI Pre- Developed TaqMan Assay Kit (catalog no. 4310884E). The ratio of HCV/GAPDH RNA is used to calculate the activity of compounds evaluated for inhibition of HCV
RNA replication.
Activity of compounds as inhibitors of HCV replication (Cell based Assay) in replicon containing Huh-7 cell lines
The effect of a specific anti-viral compound on HCV replicon RNA levels in Huh- 1 1-7 or 9-13 cells is determined by comparing the amount of HCV RNA normalized to GAPDH (e.g. the ratio of HCV/GAPDH) in the cells exposed to compound versus cells exposed to the 0% inhibition and the 100% inhibition controls. Specifically, cells are seeded at 5x 103 cells/well in a 96 well plate and are incubated either with: 1) media containing 1% DMSO (0% inhibition control), 2) 100 international units, IU/ml Interferon-alpha 2b in media/1 %DMSO or 3) media/1 %DMSO containing a fixed concentration of compound. 96 well plates as described above are then incubated at 370C for 3 days (primary screening assay) or 4 days (IC50 determination). Percent inhibition is defined as:
% Inhibition= [100-((S-C2)/Cl-C2))]xl00 where S= the ratio of HCV RNA copy number/GAPDH RNA copy number in the sample;
Cl= the ratio of HCV RNA copy number/GAPDH RNA copy number in the
0% inhibition control (media/1 %DMSO); and
C2= the ratio of HCV RNA copy number/GAPDH RNA copy number in the 100% inhibition control ( 100 IU/ml Interferon-alpha 2b).
The dose-response curve of the inhibitor is generated by adding compound in serial, three-fold dilutions over three logs to wells starting with the highest concentration of a specific compound at lOuM and ending with the lowest concentration of 0.0 IuM. Further dilution series (IuM to 0.00 IuM for example) is performed if the IC50 value is not in the linear range of the curve. IC50 is determined based on the IDBS Activity Base program using Microsoft Excel "XL Fit" in which A=I 00% inhibition value (100IU/ml Interferon-alpha 2b), B= 0% inhibition control value (media/1 %DMSO) and C= midpoint of the curve as defined as C=(B-A/2)+A. A, B and C values are expressed as the ratio of HCV RNA/GAPDH RNA as determined for each sample in each well of a 96 well plate as described above. For each plate the average of 4 wells are used to define the 100% and 0% inhibition values.
In the above assays, representative compounds of the present invention were found to have HCV replication inhibitory activity and HCV NS3 protease inhibitory activity.
These compounds were also effective in inhibiting HCV NS3 proteases of different HCV genotypes including genotypes 1, 2, 3 and 4. Representative compounds were tested in the above assays (Example 122 and Example 123). Exemplary compounds disclosed herein were found to have activities in the ranges of <= 0.2 nM-100 nM in the NS3/NS4a Protease Enzyme Assay and <= 0.2 nM- 1000 nM in the Cell-Based Replicon Assay.
Although the invention has been described with respect to various preferred embodiments, it is not intended to be limited thereto, but rather those skilled in the art will recognize that variations and modifications may be made therein which are within the spirit of the invention and the scope of the appended claims.

Claims

WHAT IS CLAIMED:
1. A compound of Formula I:
Figure imgf000073_0001
Wherein
A is selected from the group consisting of H, Ri, -(C=O)-O-R1, -(C=O)-R2, -C(=0)-NH-R2, or -S(O)2-Ri, -S(O)2NHR2; each Ri is independently selected from the group consisting of: (i) aryl; substituted aryl; heteroaryl; substituted heteroaryl;
(ii) heterocycloalkyl or substituted heterocycloalkyl; and (iii) -C1-C8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl each containing O, 1 , 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl each containing O, 1, 2, or 3 heteroatoms selected from O, S or N; -C3-Ci2 cycloalkyl, or substituted -C3-
Ci2 cycloalkyl; -C3-C]2 cycloalkenyl, or substituted -C3-Ci2 cycloalkenyl;
Each R2 is independently selected from the group consisting of: (i) hydrogen; (ii) aryl; substituted aryl; heteroaryl; substituted heteroaryl;
(iii) heterocycloalkyl or substituted heterocycloalkyl; and (iv) -Ci-C8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl each containing O, 1 , 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl each containing O, 1, 2, or 3 heteroatoms selected from O, S or N; -C3-Ci2 cycloalkyl, or substituted -C3-
Ci2 cycloalkyl; -C3-Ci2 cycloalkenyl, or substituted -C3-Ci2 cycloalkenyl; B is selected from the group consisting of H, Ri, -(C=O)-O-R], -(C=O)-R2, and -C(=0)-NH-R2;
G is selected from the group consisting of -OH, -NH-S(O)2-Ri, and -NH- S(O)2NR4R5;
each R4 and R5 are independently selected from the group consisting of: (i) hydrogen;
(ii) aryl; substituted aryl; heteroaryl; substituted heteroafyl; (iii) heterocycloalkyl or substituted heterocycloalkyl;
(iv) -CpC8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl each containing O, 1, 2, or 3 heteroatoms selected from O, S, or N; substituted -Ci-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl each containing O, 1 , 2, or 3 heteroatoms selected from O, S or N; -C3-C)2 cycloalkyl, or substituted -C3- Cj2 cycloalkyl; -C3-Ci2 cycloalkenyl, or substituted -C3-Ci2 cycloalkenyl;
L is selected from the group consisting Of -CH2-, -0-, -S-, NR4R5 or -S(O)2-;
X is absent or is selected from the group consisting of: (1) oxygen;
(2) sulfur;
(3) NR4; and
(4) -0-NH-;
Y is absent or is selected from the group consisting of:
(i) -CC=O)-, -C(=0)-NH-, -S(O)2-, -S(O)2NH-;
(ii) -Ci-C6 alkyl- containing O, 1, 2, or 3 heteroatoms selected from O, S, or N; (iii) -C2-C6 alkenyl- containing O, 1, 2, or 3 heteroatoms selected from O, S, or
N; (iv) -C2-C6 alkynyl- containing O, 1, 2, or 3 heteroatoms selected from O, S, or
N; (v) -C3-Ci2 cycloalkyl-, substituted -C3-C]2 cycloalkyl-, -heterocycloalkyl-, substituted -heterocycloalkyl-;
Z is selected from the group consisting of aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycloalkyl, substituted heterocycloalkyl;
. Z2
M
\>
Or -X-Y-Z taken together to form -J«v, wherein each Zi, Z2 are independently selected from the group consisting of: i) hydrogen; ii) aryl; iii) substituted aryl; iv) heteroaryl; v) substituted heteroaryl; vi) heterocyclic or substituted heterocyclic;
VU)-Ci-C8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl each containing 0, 1, 2, or 3 heteroatoms selected from O, S or N; viii) substituted -Ci-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl each containing 0, 1 , 2, or 3 heteroatoms selected from O, S or N; ix) -C3-Ci2 cycloalkyl; x) substituted -C3-Ci2 cycloalkyl; xi) -C3-Ci2 cycloalkenyl; xii) substituted -C3-Ci2 cycloalkenyl; xiii) -V-R8, where V is (CO), (CO)O, (CO)NR4, (SO), (SO2), (SO2)NR4; and R4 is as previously defined, R8 is selected from the group consisting of: (1) hydrogen; (2) aryl;
(3) substituted aryl;
(4) heteroaryl;
(5) substituted heteroaryl;
(6) heterocyclic or substituted heterocyclic; (7) -Ci-C8 alkyl, -C2-C8 alkenyl, or -C2-C8 alkynyl each containing 0, 1 , 2, or 3 heteroatoms selected from O, S or N; (8) substituted -Ci-C8 alkyl, substituted -C2-C8 alkenyl, or substituted -C2-C8 alkynyl each containing 0, 1, 2, or 3 heteroatoms selected from O, S or N;
(9) -C3-Ci2 cycloalkyl;
(10) substituted -C3-C12 cycloalkyl; (11) -C3-C12 cycloalkenyl;
(12) substituted -C3-C12 cycloalkenyl;
or Zi and Z2 taken together with the carbon atom to which they are attached form a cyclic moiety selected from the group consisting of: substituted or unsubstituted cycloalkyl, cycloalkenyl, or heterocyclic; substituted or unsubstituted cycloalkyl, cycloalkenyl, and heterocyclic fused with one or more aryl, substituted aryl, heteroaryl; substituted heteroaryl, heterocyclic or substituted heterocyclic; J = O, 1, 2, 3, or 4; k=l, 2, or 3; m = 0, 1, or 2; n = 0, 1, or 2; and
denotes a carbon-carbon single or double bond.
2. The compound of claim 1, wherein the compound is of Formula II:
Figure imgf000076_0001
( H ) or pharmaceutically acceptable salts, esters, or prodrugs thereof, wherein A, B, G, X, Y, Z are as defined previously in claim 1.
3. The compound of claim 1, wherein the compound is of Formula III:
Figure imgf000077_0001
( III ) or pharmaceutically acceptable salts, esters, or prodrugs thereof, wherein R6 is selected from the group consisting of aryl, substituted aryl, heteroaryl, and substituted heteroaryl; J is absent or is selected from the group consisting of O, S, alkylene, alkenylene, alkynylene, NR5, CO, (CO)NR5, (CO)O, NR5(CO), NH(CO)NH, NR5SO2; wherein R5 are as defined in claim 1 ;
Each R7i, R72, R73 and R74 is independently selected from the group consisting of: (i) hydrogen; (ii) halogen; (iii) -NO2;
(iv) -CN;
(v) -M-R4, wherein M is absent, or O, S, NH, NR5; (vi) aryl;
(vii) substituted aryl; (viii) heteroaryl;
(ix) substituted heteroaryl; (x) heterocycloalkyl; (xi) substituted heterocycloalkyl; wherein R4, R5 are as defined previously in claim 1 ; wherein A, B, G, j are as defined previously in claim 1.
4. The compound of claim 1, wherein the compound is of Formula IV:
Figure imgf000078_0001
( IV ) or pharmaceutically acceptable salts, esters, or prodrugs thereof, wherein each R6, R7I, R72, R73, R74 and J are as defined previously in claim 3; and A, B, GJ are as defined in claim 1.
5. The compound of claim 1, wherein the compound is of Formula V:
Figure imgf000078_0002
( V ) or pharmaceutically acceptable salts, esters, or prodrugs thereof, wherein each R71, R72, R73, R74 are as defined previously in claim 3; and A, B, G, j are as defined in claim 1.
6. The compound of claim 1, wherein the compound is of Formula VI:
Figure imgf000078_0003
( VI) or pharmaceutically acceptable salts, esters, or prodrugs thereof, wherein Zi, Z2 and A, B, G, j are as defined in claim 1.
7. A compound according to claim 1 which is selected from compounds of Formula VII wherein A, Q and G are delineated in Table 1 :
Figure imgf000079_0001
( VII)
TABLE 1
Figure imgf000079_0002
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
8. A pharmaceutical composition comprising an inhibitory amount of a compound according to claim 1 in combination with a pharmaceutically acceptable carrier or excipient.
9. A method of treating a viral infection in a subject, comprising administering to the subject an inhibitory amount of a pharmaceutical composition according to claim 8.
10. The method of claim 9, wherein the viral infection is hepatitis C.
11. A method of inhibiting the replication of hepatitis C virus, the method comprising supplying a hepatitis C viral NS3 protease inhibitory amount of the pharmaceutical composition of claim 8.
12. The method of claim 9 further comprising administering concurrently an additional anti- hepatitis C virus agent.
13. The method of claim 12, wherein said additional anti-hepatitis C virus agent is selected from the group consisting of: α-interferon, β-interferon, ribavarin, and adamantine.
14. The method of claim 12, wherein said additional anti-hepatitis C virus agent is an inhibitor of hepatitis C virus helicase, polymerase, metalloprotease, or IRES.
15. The pharmaceutical composition of claim 8, further comprising another anti-HCV agent.
16. The pharmaceutical composition of claim 8, further comprising an agent selected from interferon, ribavirin, amantadine, another HCV protease inhibitor, an HCV polymerase inhibitor, an HCV helicase inhibitor, or an internal ribosome entry site inhibitor.
17. The pharmaceutical composition of claim 8, further comprising pegylated interferon.
18. The pharmaceutical composition of claim 8, further comprising another anti-viral, antibacterial, anti-fungal or anti-cancer agent, or an immune modulator.
19. The pharmaceutical composition of claim 8, further comprising ritonavir.
PCT/IB2008/002839 2007-10-22 2008-10-23 P3 hydroxyamino macrocyclic hepatitis c serine protease inhibitors WO2009053828A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US98161307P 2007-10-22 2007-10-22
US60/981,613 2007-10-22

Publications (2)

Publication Number Publication Date
WO2009053828A2 true WO2009053828A2 (en) 2009-04-30
WO2009053828A3 WO2009053828A3 (en) 2009-12-30

Family

ID=40580157

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2008/002839 WO2009053828A2 (en) 2007-10-22 2008-10-23 P3 hydroxyamino macrocyclic hepatitis c serine protease inhibitors

Country Status (1)

Country Link
WO (1) WO2009053828A2 (en)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011017389A1 (en) 2009-08-05 2011-02-10 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors useful against viral infections, particularly hcv
WO2011075615A1 (en) 2009-12-18 2011-06-23 Idenix Pharmaceuticals, Inc. 5,5-fused arylene or heteroarylene hepatitis c virus inhibitors
US8222203B2 (en) 2007-12-14 2012-07-17 Enanta Pharmaceuticals, Inc. Macrocyclic oximyl hepatitis C serine protease inhibitors
US8232246B2 (en) 2009-06-30 2012-07-31 Abbott Laboratories Anti-viral compounds
WO2012109398A1 (en) 2011-02-10 2012-08-16 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors, pharmaceutical compositions thereof, and their use for treating hcv infections
WO2012135581A1 (en) 2011-03-31 2012-10-04 Idenix Pharmaceuticals, Inc. Methods for treating drug-resistant hepatitis c virus infection with a 5,5-fused arylene or heteroarylene hepatitis c virus inhibitor
US8283309B2 (en) 2007-12-20 2012-10-09 Enanta Pharmaceuticals, Inc. Bridged carbocyclic oxime hepatitis C virus serine protease inhibitors
US8361958B2 (en) 2007-12-05 2013-01-29 Enanta Pharmaceuticals, Inc. Oximyl HCV serine protease inhibitors
US8372802B2 (en) 2008-03-20 2013-02-12 Enanta Pharmaceuticals, Inc. Fluorinated macrocyclic compounds as hepatitis C virus inhibitors
US8420596B2 (en) 2008-09-11 2013-04-16 Abbott Laboratories Macrocyclic hepatitis C serine protease inhibitors
US8426360B2 (en) 2007-11-13 2013-04-23 Enanta Pharmaceuticals, Inc. Carbocyclic oxime hepatitis C virus serine protease inhibitors
WO2013090929A1 (en) * 2011-12-15 2013-06-20 Gilead Sciences, Inc. Amino quinoline derivatives inhibitors of hcv
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
WO2015042375A1 (en) 2013-09-20 2015-03-26 Idenix Pharmaceuticals, Inc. Hepatitis c virus inhibitors
US8993595B2 (en) 2009-04-08 2015-03-31 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors
EP2899207A1 (en) 2014-01-28 2015-07-29 Amikana.Biologics New method for testing HCV protease inhibition
WO2015134560A1 (en) 2014-03-05 2015-09-11 Idenix Pharmaceuticals, Inc. Solid forms of a flaviviridae virus inhibitor compound and salts thereof
WO2015134561A1 (en) 2014-03-05 2015-09-11 Idenix Pharmaceuticals, Inc. Pharmaceutical compositions comprising a 5,5-fused heteroarylene flaviviridae inhibitor and their use for treating or preventing flaviviridae infection
US9334279B2 (en) 2012-11-02 2016-05-10 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9333204B2 (en) 2014-01-03 2016-05-10 Abbvie Inc. Solid antiviral dosage forms
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
US10201584B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070021330A1 (en) * 2003-07-03 2007-01-25 Enanta Pharmaceuticals, Inc. Aza-peptide macrocyclic hepatitis c serine protease inhibitors
US20070054842A1 (en) * 2005-07-25 2007-03-08 Blatt Lawrence M Novel macrocyclic inhibitors of hepatitis C virus replication
US20070060510A1 (en) * 2003-04-18 2007-03-15 Enanta Pharmaceuticals, Inc. Quinoxalinyl macrocyclic hepatitis C serine protease inhibitors
US20070161575A1 (en) * 2003-06-05 2007-07-12 Enanta Pharmaceuticals, Inc. Tri-peptide hepatitis C serine protease inhibitors
US20070179167A1 (en) * 2005-08-26 2007-08-02 Cottrell Kevin M Inhibitors of serine proteases

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070060510A1 (en) * 2003-04-18 2007-03-15 Enanta Pharmaceuticals, Inc. Quinoxalinyl macrocyclic hepatitis C serine protease inhibitors
US20070161575A1 (en) * 2003-06-05 2007-07-12 Enanta Pharmaceuticals, Inc. Tri-peptide hepatitis C serine protease inhibitors
US20070021330A1 (en) * 2003-07-03 2007-01-25 Enanta Pharmaceuticals, Inc. Aza-peptide macrocyclic hepatitis c serine protease inhibitors
US20070054842A1 (en) * 2005-07-25 2007-03-08 Blatt Lawrence M Novel macrocyclic inhibitors of hepatitis C virus replication
US20070179167A1 (en) * 2005-08-26 2007-08-02 Cottrell Kevin M Inhibitors of serine proteases

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8426360B2 (en) 2007-11-13 2013-04-23 Enanta Pharmaceuticals, Inc. Carbocyclic oxime hepatitis C virus serine protease inhibitors
US8361958B2 (en) 2007-12-05 2013-01-29 Enanta Pharmaceuticals, Inc. Oximyl HCV serine protease inhibitors
US8222203B2 (en) 2007-12-14 2012-07-17 Enanta Pharmaceuticals, Inc. Macrocyclic oximyl hepatitis C serine protease inhibitors
US8283309B2 (en) 2007-12-20 2012-10-09 Enanta Pharmaceuticals, Inc. Bridged carbocyclic oxime hepatitis C virus serine protease inhibitors
US8372802B2 (en) 2008-03-20 2013-02-12 Enanta Pharmaceuticals, Inc. Fluorinated macrocyclic compounds as hepatitis C virus inhibitors
US8642538B2 (en) 2008-09-11 2014-02-04 Abbvie, Inc. Macrocyclic hepatitis C serine protease inhibitors
US8420596B2 (en) 2008-09-11 2013-04-16 Abbott Laboratories Macrocyclic hepatitis C serine protease inhibitors
US9309279B2 (en) 2008-09-11 2016-04-12 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
WO2011017389A1 (en) 2009-08-05 2011-02-10 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors useful against viral infections, particularly hcv
WO2011075615A1 (en) 2009-12-18 2011-06-23 Idenix Pharmaceuticals, Inc. 5,5-fused arylene or heteroarylene 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
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
WO2012109398A1 (en) 2011-02-10 2012-08-16 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors, pharmaceutical compositions thereof, and their use for treating hcv infections
WO2012135581A1 (en) 2011-03-31 2012-10-04 Idenix Pharmaceuticals, Inc. Methods for treating drug-resistant hepatitis c virus infection with a 5,5-fused arylene or heteroarylene hepatitis c virus inhibitor
US9527885B2 (en) 2011-05-05 2016-12-27 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8957203B2 (en) 2011-05-05 2015-02-17 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US10201584B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
US10201541B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
WO2013090929A1 (en) * 2011-12-15 2013-06-20 Gilead Sciences, Inc. Amino quinoline derivatives inhibitors of hcv
US9499550B2 (en) 2012-10-19 2016-11-22 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9334279B2 (en) 2012-11-02 2016-05-10 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
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
WO2015042375A1 (en) 2013-09-20 2015-03-26 Idenix Pharmaceuticals, Inc. 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
EP2899207A1 (en) 2014-01-28 2015-07-29 Amikana.Biologics New method for testing HCV protease inhibition
WO2015134561A1 (en) 2014-03-05 2015-09-11 Idenix Pharmaceuticals, Inc. Pharmaceutical compositions comprising a 5,5-fused heteroarylene flaviviridae inhibitor and their use for treating or preventing flaviviridae infection
WO2015134560A1 (en) 2014-03-05 2015-09-11 Idenix Pharmaceuticals, Inc. Solid forms of a flaviviridae virus inhibitor compound and salts thereof

Also Published As

Publication number Publication date
WO2009053828A3 (en) 2009-12-30

Similar Documents

Publication Publication Date Title
US8383583B2 (en) Macrocyclic, pyridazinone-containing hepatitis C serine protease inhibitors
US8273709B2 (en) Triazole-containing macrocyclic HCV serine protease inhibitors
US8304385B2 (en) Macrocyclic tetrazolyl hepatitis C serine protease inhibitors
WO2009053828A2 (en) P3 hydroxyamino macrocyclic hepatitis c serine protease inhibitors
US8263549B2 (en) C5-substituted, proline-derived, macrocyclic hepatitis C serine protease inhibitors
EP2219453B1 (en) Quinoxaline-containing compounds as hepatitis c virus inhibitors
US8222203B2 (en) Macrocyclic oximyl hepatitis C serine protease inhibitors
EP2429568B1 (en) Macrocyclic compounds as hepatitis c virus inhibitors
US8426360B2 (en) Carbocyclic oxime hepatitis C virus serine protease inhibitors
US8501682B2 (en) Difluorinated tripeptides as HCV serine protease inhibitors
WO2009076166A2 (en) Oximyl hcv serine protease inhibitors
WO2009085978A1 (en) Bridged carbocyclic oxime hepatitis c virus serine protease inhibitors
WO2009070689A1 (en) Bicyclic, c5-substituted proline derivatives as inhibitors of the hepatitis c virus ns3 protease
EP2224942A2 (en) Fluorinated tripeptide hcv serine protease inhibitors
WO2008021956A2 (en) Acylaminoheteroaryl hepatitis c virus protease inhibitors
WO2009117594A1 (en) Fluorinated macrocyclic compounds as hepatitis c virus inhibitors
WO2008134395A1 (en) Aza-peptide macrocyclic hepatitis c serine protease inhibitors
WO2008021733A2 (en) Tetrazolyl acyclic hepatitis c serine protease inhibitors
WO2008022006A2 (en) Arylalkoxyl hepatitis c virus protease inhibitors
WO2008019289A2 (en) Tetrazolyl macrocyclic hepatitis c serine protease inhibitors
WO2008021871A2 (en) Triazolyl acyclic hepatitis c serine protease inhibitors
WO2008021960A2 (en) Triazolyl macrocyclic hepatitis c serine protease inhibitors
WO2009134987A1 (en) Difluoromethyl-containing macrocyclic compounds as hepatitis c virus inhibitors
WO2009094438A1 (en) Heteroaryl-containing tripeptide hcv serine protease inhibitors

Legal Events

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

Ref document number: 08841639

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08841639

Country of ref document: EP

Kind code of ref document: A2