WO2011031934A1 - Hepatitis c virus inhibitors - Google Patents

Hepatitis c virus inhibitors Download PDF

Info

Publication number
WO2011031934A1
WO2011031934A1 PCT/US2010/048377 US2010048377W WO2011031934A1 WO 2011031934 A1 WO2011031934 A1 WO 2011031934A1 US 2010048377 W US2010048377 W US 2010048377W WO 2011031934 A1 WO2011031934 A1 WO 2011031934A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
alkyl
compound
heteroaryl
ring
Prior art date
Application number
PCT/US2010/048377
Other languages
French (fr)
Inventor
Yao-Ling Qiu
Ce Wang
Lu Ying
Xiaowen Peng
Yat Sun Or
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.
Priority to EP10816134.0A priority Critical patent/EP2475254A4/en
Publication of WO2011031934A1 publication Critical patent/WO2011031934A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/18Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D207/22Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/24Oxygen or sulfur atoms
    • C07D207/262-Pyrrolidones
    • C07D207/2732-Pyrrolidones with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to other ring carbon atoms
    • C07D207/277Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D207/282-Pyrrolidone-5- carboxylic acids; Functional derivatives thereof, e.g. esters, nitriles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/08Bridged systems

Definitions

  • the present invention relates to novel antiviral agents. More specifically, the present invention relates to compounds which can inhibit the function of the NS5A protein encoded by Hepatitis C virus (HCV), compositions comprising such compounds, methods for inhibiting HCV viral replication, methods for treating or preventing HCV infection, and processes for making the compounds.
  • HCV Hepatitis C virus
  • HCV infection is responsible for 40-60% of all chronic liver disease and 30% of all liver transplants.
  • Chronic HCV infection accounts for 30%) of all cirrhosis, end-stage liver disease, and liver cancer in the U.S. The CDC estimates that the number of deaths due to HCV will minimally increase to 38,000/year by the year 2010.
  • Alpha-interferon (alone or in combination with ribavirin) has been widely used since its approval for treatment of chronic HCV infection.
  • adverse side effects are commonly associated with this treatment: flulike symptoms, leukopenia, thrombocytopenia, depression from interferon, as well as anemia induced by ribavirin (Lindsay, K. L. (1997) Hepatology, 26 (suppl 1): 71S-77S).
  • HCV is now widely accepted as the most common causative agent of posttransfusion non-A, non-B hepatitis (NANBH) (Kuo, G et al (1989) Science, 244:362-364). Due to its genome structure and sequence homology, this virus was assigned as a new genus in the Flaviviridae family. Like the other members of the Flaviviridae, such as flaviviruses (e.g. yellow fever virus and Dengue virus types 1-4) and pestiviruses (e.g.
  • HCV bovine viral diarrhea virus, border disease virus, and classic swine fever virus
  • the HCV genome is approximately 9.6 kilobases (kb) with a long, highly conserved, noncapped 5' nontranslated region (NTR) of approximately 340 bases which functions as an internal ribosome entry site (IRES) (Wang CY et al 'An RNA pseudoknot is an essential structural element of the internal ribosome entry site located within the hepatitis C virus 5' noncoding region' RNA - A Publication of the RNA
  • This element is followed by a region which encodes a single long open reading frame (ORF) encoding a polypeptide of -3000 amino acids comprising both the structural and nonstructural viral proteins.
  • ORF long open reading frame
  • this RNA Upon entry into the cytoplasm of the cell, this RNA is directly translated into a polypeptide of -3000 amino acids comprising both the structural and nonstructural viral proteins.
  • This large polypeptide is subsequently processed into the individual structural and nonstructural proteins by a combination of host and virally-encoded proteinases (Rice, CM. (1996) in B.N. Fields, D.M.Knipe and P.M. Howley (eds) Virology, 2 nd Edition, p931-960; Raven Press, N.Y.).
  • host and virally-encoded proteinases 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 several 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.
  • NS5A is a membrane-anchored phosphoprotein that is observed in basally phosphorylated (56 kDa) and hyperphosphorylated (58 kDa) forms.
  • NS5A While its function has not fully been elucidated, NS5A is believed to be important in viral replication.
  • the NS5B protein (591 amino acids, 65 kDa) of HCV (Behrens, S.E. et al (1996) EMBO J. , 151 2-22) encodes an R A-dependent RNA polymerase (RdRp) activity and contains canonical motifs present in other RNA viral polymerases.
  • RdRp R A-dependent RNA polymerase
  • the NS5B protein is fairly well conserved both intra- typically (-95-98% amino acid (aa) identity across lb isolates) and inter-typically (-85% aa identity between genotype la and lb isolates).
  • 3' NTR which roughly consists of three regions: an -40 base region which is poorly conserved among various genotypes, a variable length poly(U)/polypyrimidine tract, and a highly conserved 98 base element also called the "3' X-tail" (Kolykhalov, A. et al (1996) J. Virology 70:3363-3371; Tanaka, T. et al (1995) Biochem Biophys. Res. Commun., 215744-749; Tanaka, T. et al (1996) J. Virology, 70:3307-3312; Yamada, N. et al (1996) Virology, 223:255-261).
  • the 3' NTR is predicted to form a stable secondary structure which is essential for HCV growth in chimps and is believed to function in the initiation and regulation of viral RNA replication.
  • HCV NS5A protein is described, for example, in Tan, S.-L., Katzel, M.G. Virology, 2001, 284, 1; and in Rice, C. M. Nature, 2005, 435, 374.
  • the present invention relates to novel antiviral compounds represented herein below, pharmaceutical compositions comprising such compounds, and methods for the treatment or prophylaxis of viral (particularly HCV) infection in a subject in need of such therapy with said compounds.
  • 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 provides a compound of Formula
  • Ring A is absent or a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C 3 -C 8 cycloalkyl, and C 3 -C 8 cycloalkenyl, each optionally substituted; preferably optionally substituted aryl or optionally substituted heteroaryl;
  • Ring B is a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C 3 -C 8 cycloalkyl, and C 3 -C 8 cycloalkenyl, each optionally substituted; preferably optionally substituted aryl or optionally substituted heteroaryl;
  • L is absent or selected from the group consisting of optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, and optionally substituted C 2 -C 4 alkynyl;
  • G is optionally substituted 5 -membered heteroaryl or optionally substituted 5/6- member fused heteroaryl; wherein the 5 -membered heteroaryl contains one or more nitrogen atoms, and wherein the 6-membered ring of said 5/6-fused membered heteroaryl is attached to one of Ring A, L, and Ring B, and is aryl or heteroaryl; preferably optionally substituted imidazolyl or optionally substituted benzimidazolyl;
  • J is selected from -N(R lc )-C(0)-, -N(R lc )-C(0)0- and -N(R lc )-C(0)-N(R lc )-; preferably -N(R lc )-C(0)-;
  • W at each occurrence is independently O or -N(R lb )-; preferably -N(R lb )-;
  • R 1 , R la , R lb , R lc , R 9 , and R 9a at each occurence are each independently hydrogen or optionally substituted C 1 -C 4 alkyl; alternatively R la and R 9a , R la and R 9 , R 1 and R 9a , or R 1 and R 9 can be taken together with the carbon atom(s) to which they are attached to form an optionally substituted C 3 -C 8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or yet alternatively R lb and R 9a , or R lb and R 9 can be taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic ring;
  • R 6 at each occurence is independently selected from the group consisting of optionally substituted 0(C 1 -C 8 alkyl), optionally substituted amino, C 1 -C 8 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted; preferably optionally substituted C 1 -C 8 alkyl; more preferably C 1 -C8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C 1 -C 4 alkyl);
  • Q is selected from: and ;
  • X is absent, O, S, CH 2 , or CH 2 CH 2 ;
  • Y is absent, O, S, C(R , C(R 1 ) 2 C(R 7 ) 2 , C(R 1 ) 2 C(R 7 ) 2 C(R 7 ) 2 , C(R 1 ) 2 OC(R 1 ) 2 , or C(R ⁇ SC(R 1 ) 2 ;
  • At least one of X and Y is not O or S;
  • U is absent or independently selected from O, S, S(O), S0 2 , NC(0)-(C 1 -C 4 alkyl),
  • R 7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C 1 -C 4 alkyl), S(C 1 -C 4 alkyl), amino optionally substituted with one or two C 1 -C 4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C 1 -C 4 alkyl; preferably hydrogen, halogen or hydroxy;
  • two geminal R 7 groups can be taken together with the cabon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkenyl and 3- to 8- membered heterocyclic, each optionally substituted; preferably two geminal R 7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted cyclopropyl or a spiro, optionally substituted 5- to 6-membered heterocyclic;
  • R 2 at each occurence is independently hydrogen, optionally substituted C 1 -C 8 alkyl, or -NR a R b ;
  • R a at each occurence is independently hydrogen or optionally substituted C 1 -C 8 alkyl;
  • R b at each occurence is -C(0)-R 6 ;
  • R a and R b can be taken together with the nitrogen atom to which they are attached to form an optionally substituted heterocyclic or optionally substituted heteroaryl group;
  • R 3 and R 4 are each independently selected from the group consisting of hydrogen, optionally substituted C 1 -C 8 alkyl, optionally substituted C 2 -C 8 alkenyl, and optionally substituted C 3 -C 8 cycloalkyl; preferably hydrogen or optionally substituted C1-C 4 alkyl; alternatively, R 3 and R 4 can be taken together with the carbon atom to which they are attached to form optionally substituted C 3 -C 8 cycloalkyl or optionally substituted heterocyclic;
  • R 5 is independently hydrogen, optionally substituted C 1 -C 8 alkyl, or optionally substituted C 3 -C 8 cycloalkyl; preferably hydrogen or optionally substituted C1-C 4 alkyl; and
  • the present invention provides a compound of Formula (2-1)
  • Ring A and Ring B are each independently a monocyclic or polycyclic group selected from aryl, heteroaryl, heterocyclic, C 3 -C 8 cycloalkyl, and C 3 -C 8 cycloalkenyl, each optionally substituted; preferably optionally substituted aryl or optionally substituted heteroaryl; L is absent or selected from the group consisting of optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, and optionally substituted C 2 -C 4 alkynyl; wherein taken together is not optionally substituted phenyl-thiazolyl or 3,4'-biphenyl;
  • J is selected from the group consisting of -N(R lc )-C( O)-, -N(R lc )-C(0)0- and
  • W at each occurrence is independently O or -N(R lb )-; preferably -N(R lb )-;
  • R 1 , R la , R lb , R lc , R 9 , and R 9a at each occurrence are each independently hydrogen or optionally substituted C 1 -C 4 alkyl; alternatively, R la and R 9a , R la and R 9 , R 1 and R 9a , or R J and R 9 can be taken together with the carbon atom(s) to which they are attached to form an optionally substituted C 3 -C 8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or yet alternatively R lb and R 9a , or R lb and R 9 can be taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic;
  • R 6 at each occurrence is independently selected from the group consisting of optionally substituted 0(C 1 -C 8 alkyl); optionally substituted amino; C 1 -C 8 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted; preferably optionally substituted C 1 -C 8 alkyl; more preferably C 1 -C 8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C 1 -C 4 alkyl);
  • G is selected from the group consisting of -N(R lc )-C(0)-, -N(R lc )-C(0)0- and -N(R lc )-C( -N(R lc )-; preferably -N(R lc )-C(0)-;
  • X is absent, O, S, CH 2 , or CH 2 CH 2 ;
  • Y at is absent, O, S, C(R , CCR ⁇ C(R 7 ⁇ , CCR ⁇ C ⁇ C ⁇ , C(R ⁇ OCCR 1 ⁇ , or C(R ⁇ SCCR 1 ⁇ ; Wherein at least one of X and Y is present;
  • At least one of X and Y is not O or S;
  • R 7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C 1 -C 4 alkyl), S(C 1 -C 4 alkyl), amino optionally substituted with one or two C 1 -C 4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C 1 -C 4 alkyl; preferably hydrogen, halogen or hydroxy;
  • R 2 at each occurrence is independently hydrogen, optionally substituted C 1 -C 8 alkyl, or -NR a R b ;
  • R a at each occurrence is independently hydrogen or optionally substituted C 1 -C 8 alkyl
  • R b at each occurrence is -C(0)-R 6 ;
  • R a and R b can be taken together with the nitrogen atom to which they are attached to form an optionally substituted heterocyclic or optionally substituted heteroaryl group.
  • the present invention provides a compound of Formula
  • Ring A and Ring B are each independently absent or a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C3-C8 cycloalkyl, and C 3 -C 8 cycloalkenyl, each optionally substituted; preferably optionally substituted aryl or optionally substituted heteroaryl;
  • L is absent or selected from the group consisting of optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, and optionally substituted C 2 -C 4 alkynyl;
  • Ring A, Ring B and L is present;
  • G and J are each independently optionally substituted 5-membered heteroaryl or optionally substituted 5/6-member fused heteroaryl; wherein the 5-membered heteroaryl contains one or more nitrogen atoms, and wherein the 6-membered ring of said 5/6-fused membered heteroaryl is attached to one of Ring A, Ring B and L, and is aryl or heteroaryl; preferably optionally substituted imidazolyl or optionally substituted benzimidazolyl; W at each occurrence is independently O or -N(R lb )-; preferably -N(R lb )-;
  • R 1 , R la , R lb , R 9 , and R 9a at each occurrence are each independently hydrogen or optionally substituted C 1 -C 4 alkyl; alternatively R la and R 9a , R la and R 9 , R 1 and R 9a , or R 1 and R 9 can be taken together with the carbon atom(s) to which they are attached to form an optionally substituted C 3 -C 8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or yet alternatively R lb and R 9a , or R lb and R 9 can be taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic;
  • R 6 at each occurrence is independently selected from the group consisting of optionally substituted 0(C 1 -C 8 alkyl); optionally substituted amino; C 1 -C 8 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, C 3 -C8 cycloalkyl, C 3 -C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted; preferably optionally substituted C 1 -C 8 alkyl; more preferably C 1 -C8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C 1 -C 4 alkyl);
  • Q is selected from: , and;
  • X is absent, O, S, CH 2 , or CH 2 CH 2 ;
  • Y is absent, O, S, C(R , C(R 1 ) 2 C(R 7 ) 2 , C(R 1 ) 2 C(R 7 ) 2 C(R 7 ) 2 , C(R 1 ) 2 OC(R 1 ) 2 , or C(R ⁇ SC(R 1 ) 2 ;
  • At least one of X and Y is not O or S;
  • U is absent or independently selected from O, S, S(O), S0 2 , NC(0)-(C 1 -C 4 alkyl), C(O), protected carbonyl, OCH 2 , OCH 2 CH 2 , SCH 2 , SCH 2 CH 2 , C(R 7 ) 2 , and C(R 7 ) 2 C(R 7 ) 2 ; preferably CH 2 ;
  • R 7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C 1 -C 4 alkyl), S(C 1 -C 4 alkyl), amino optionally substituted with one or two C 1 -C 4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C 1 -C 4 alkyl; preferably hydrogen, halogen or hydroxy;
  • two geminal R 7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C 3 -C 8 cycloalkyl, C3-C 8 cycloalkenyl and 3- to 8- membered heterocyclic, each optionally substituted; preferably an optionally substituted cyclopropyl or an optionally substituted 5- to 6-membered heterocyclic;
  • R 2 at each occurrence is independently hydrogen, optionally substituted C 1 -C 8 alkyl, or -NR a R b ;
  • R a at each occurrence is independently hydrogen or optionally substituted C 1 -C 8 alkyl
  • R b at each occurrence is -C(0)-R 6 ;
  • R a and R b can be taken together with the nitrogen atom to which they are attached to form an optionally substituted heterocyclic or optionally substituted heteroaryl group;
  • R 3 and R 4 are each independently selected from the group consisting of hydrogen, optionally substituted C 1 -C 8 alkyl, optionally substituted C 2 -C 8 alkenyl, and optionally substituted C 3 -C 8 cycloalkyl; preferably hydrogen or optionally substituted C1-C 4 alkyl; alternatively, R 3 and R 4 can be taken together with the carbon atom to which they are attached to form optionally substituted C 3 -C 8 cycloalkyl or optionally substituted heterocyclic;
  • R 5 is independently hydrogen, optionally substituted C 1 -C 8 alkyl, or optionally substituted C 3 -C 8 cycloalkyl; preferably hydrogen or optionally substituted C1-C 4 alkyl; and
  • the present invention provides a compound of Formula (4-1):
  • Ring A is absent or a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C 3 -C 8 cycloalkyl, and C 3 -C 8 cycloalkenyl, each optionally substituted; preferably optionally substituted aryl or optionally substituted heteroaryl;
  • Ring B is a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C 3 -C 8 cycloalkyl, and C 3 -C 8 cycloalkenyl, each optionally substituted; preferably optionally substituted aryl or optionally substituted heteroaryl;
  • L is absent or selected from the group consisting of optionally substituted C1-C 4 alkyl, optionally substituted C2-C 4 alkenyl, and optionally substituted C2-C 4 alkynyl;
  • R 1 at each occurrence is independently hydrogen or optionally substituted C1-C 4 alkyl
  • R 6 at each occurrence is independently selected from the group consisting of optionally substituted 0(C 1 -C 8 alkyl); optionally substituted amino; C 1 -C 8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted; preferably optionally substituted C 1 -C 8 alkyl; more preferably C 1 -C 8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C 1 -C 4 alkyl);
  • U is absent or independently selected from O, S, S(O), S0 2 , NC(0)-(C 1 -C 4 alkyl), C(O), protected carbonyl, OCH 2 , OCH 2 CH 2 , SCH 2 , SCH 2 CH 2 , C(R 7 ) 2 , and C(R 7 ) 2 C(R 7 ) 2 ; preferably CH 2 ;
  • R 7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C 1 -C 4 alkyl), S(C 1 -C 4 alkyl), amino optionally substituted with one or two C1-C 4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C1-C 4 alkyl; preferably hydrogen, halogen or hydroxy;
  • two geminal R 7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkenyl and 3- to 8- membered heterocyclic, each optionally substituted; preferably an optionally substituted C 3 -C 8 cyclopropyl or an optionally substituted 5- to 6-membered heterocyclic;
  • G is optionally substituted 5 -membered heteroaryl or optionally substituted 5/6- member fused heteroaryl; wherein the 5 -membered heteroaryl contains one or more nitrogen atoms, and wherein the 6-membered ring of said 5/6-fused membered heteroaryl is attached to one of Ring A, L and Ring B, and is aryl or heteroaryl; preferably optionally substituted imidazolyl or optionally substituted benzimidazolyl;
  • Q is selected from: ,
  • V is selected from the group consisting of -N(R lc )-, -N(R lc )-C(0)-, -N(R lc )- C(0)0- and -N(R lc )-C(0)-N(R lc )-; preferably -N(R lc )- or -N(R lc )-C(0)-;
  • W is O or -N(R lb )-; preferably -N(R lb )-;
  • R la , R lb , R lc , R ld , R 9 , and R 9a at each occurrence are each independently hydrogen or optionally substituted C 1 -C 4 alkyl; alternatively R la and R 9a , R la and R 9 , R ld and R 9a , or R ld and R 9 can be taken together with the carbon atom(s) to which they are attached to form an optionally substituted C 3 -C 8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or yet alternatively R lb and R 9a , or R lb and R 9 can be taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic;
  • X is absent, O, S, CH 2 , or CH 2 CH 2 ;
  • Y is absent, O, S, C(R , C(R 1 ) 2 C(R 7 ) 2 , C(R 1 ) 2 C(R 7 ) 2 C(R 7 ) 2 , C(R 1 ) 2 OC(R 1 ) 2 , or C(R ⁇ SC(R 1 ) 2 ;
  • At least one of X and Y is not O or S;
  • R 2 is hydrogen, optionally substituted C 1 -C 8 alkyl, or
  • R a is hydrogen or optionally substituted C 1 -C 8 alkyl
  • R b at each occurrence is -C(0)-R 6 ;
  • R a and R b can be taken together with the nitrogen atom to which they are attached to form an optionally substituted heterocyclic or optionally substituted heteroaryl group;
  • R 3 and R 4 are each independently selected from the group consisting of hydrogen, optionally substituted C 1 -C 8 alkyl, optionally substituted C 2 -C 8 alkenyl, and optionally substituted C 3 -C 8 cycloalkyl; preferably hydrogen or optionally substituted C 1 -C 4 alkyl; alternatively, R 3 and R 4 can be taken together with the carbon atom to which they are attached to form optionally substituted C 3 -C 8 cycloalkyl or optionally substituted heterocyclic; and
  • R 5 is independently hydrogen, optionally substituted C 1 -C 8 alkyl, or optionally substituted C3-C8 cycloalkyl; preferably hydrogen or optionally substituted C 1 -C 4 alkyl.
  • the present invention provides a compound of Formula
  • Ring A is absent or a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C 3 -C 8 cycloalkyl, and C 3 -C 8 cycloalkenyl, each optionally substituted; preferably optionally substituted aryl or optionally substituted heteroaryl;
  • Ring B is an optionally substituted aryl or optionally substituted heteroaryl
  • L is absent or selected from the group consisting of optionally substituted C 1 -C 4 alkyl, optionally substituted C 2 -C 4 alkenyl, and optionally substituted C 2 -C 4 alkynyl;
  • W at each occurrence is independently O or -N(R lb )-; preferably -N(R lb )-;
  • R 1 , R la , R lb , R lc , R 9 , and R 9a at each occurrence are each independently hydrogen or optionally substituted C 1 -C 4 alkyl; alternatively R la and R 9a , R la and R 9 , R 1 and R 9a , or R 1 and R 9 can be taken together with the carbon atom(s) to which they are attached to form an optionally substituted C 3 -C 8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or yet alternatively R lb and R 9a , or R lb and R 9 can be taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic;
  • R 6 at each occurrence is independently selected from the group consisting of optionally substituted 0(C 1 -C 8 alkyl); optionally substituted amino; C 1 -C 8 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted; preferably optionally substituted C 1 -C 8 alkyl; more preferably C 1 -C 8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C 1 -C 4 alkyl);
  • G is optionally substituted 5 -membered heteroaryl or optionally substituted 5/6- member fused heteroaryl; wherein the 5 -membered heteroaryl contains one or more nitrogen atoms, and wherein the 6-membered ring of said 5/6-fused membered heteroaryl is attached to one of Ring A, L and Ring B and is aryl or heteroaryl; preferably optionally substituted imidazolyl or optionally substituted benzimidazolyl;
  • Q is selected from: ;
  • V is selected from the group consisting of -N(R lc )-, -N(R lc )-C(0)-, -N(R lc )- C(0)0- and -N(R lc )-C(0)-N(R lc )-;
  • X is absent, O, S, CH 2 , or CH 2 CH 2 ;
  • Y is absent, O, S, C(R , C(R 1 ) 2 C(R 7 ) 2 , C(R 1 ) 2 C(R 7 ) 2 C(R 7 ) 2 , C(R 1 ) 2 OC(R 1 ) 2 , or C(R ⁇ SC(R 1 ) 2 ;
  • At least one of X and Y is not O or S;
  • R 7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C 1 -C 4 alkyl), S(C 1 -C 4 alkyl), amino optionally substituted with one or two C 1 -C 4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C 1 -C 4 alkyl; preferably hydrogen, halogen or hydroxy;
  • U is absent or selected from O, S, S(O), S0 2 , NC(0)-(C C 4 alkyl), C(O), protected carbonyl, OCH 2 , OCH 2 CH 2 , SCH 2 , SCH 2 CH 2 , C(R 7 ) 2 , and C(R 7 ) 2 C(R 7 ) 2 ; preferably CH 2 ;
  • Optionally two geminal R 7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkenyl and 3- to 8- membered heterocyclic, each optionally substituted; preferably an optionally substituted cyclopropyl or an optionally substituted 5- to 6-membered heterocyclic;
  • R 2 is hydrogen, optionally substituted C 1 -C 8 alkyl, or -NR a R b ;
  • R a is hydrogen or optionally substituted C 1 -C 8 alkyl
  • R b is -C(0)-R 6 ;
  • R a and R b can be taken together with the nitrogen atom to which they are attached to form an optionally substituted heterocyclic or optionally substituted heteroaryl group;
  • R 3 and R 4 are each independently selected from the group consisting of hydrogen, optionally substituted C 1 -C 8 alkyl, optionally substituted C 2 -C 8 alkenyl, and optionally substituted C 3 -C 8 cycloalkyl; preferably hydrogen or optionally substituted C 1 -C 4 alkyl; alternatively, R 3 and R 4 can be taken together with the carbon atom to which they are attached to form optionally substituted C 3 -C 8 cycloalkyl or optionally substituted heterocyclic;
  • R 5 is hydrogen, optionally substituted C 1 -C 8 alkyl, or optionally substituted C 3 -C 8 cycloalkyl; preferably hydrogen or optionally substituted C 1 -C 4 alkyl; and
  • the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound or combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable carrier or excipient.
  • the present invention provides a method of inhibiting the replication of a RNA-containing virus comprising contacting said virus with said pharmaceutical composition. Particularly, this invention is directed to methods of inhibiting the replication of HCV.
  • the present invention provides a method of treating or preventing infection caused by an RNA-containing virus comprising administering to a patient in need of such treatment a therapeutically effective amount of a compound or combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof.
  • this invention is directed to methods of treating or preventing infection caused by HCV.
  • Yet another aspect of the present invention provides the use of a compound or combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof, as defined hereinafter, in the preparation of a medicament for the treatment or prevention of infection caused by RNA-containing virus, specifically HCV.
  • the compounds of the invention have utility in inhibiting the replication of RNA- containing virus, including, for example, HCV.
  • Other compounds useful for inhibiting the replication of RNA-containing viruses and/or for the treatment or prophylaxis of HCV infection have been described in copending U.S. Application Serial No. 12/702,673 filed February 9, 2010 entitled “Linked Dibenzimidiazole Derivatives"; U.S. Application Serial No. 12/702,692 filed February 9, 2010 entitled “Linked Dibenzimidiazole Derivatives"; U.S. Application Serial No. 12/702,802 filed February 9, 2010 entitled "Linked
  • Provisional Application Serial No. 61/241,578 filed September 11, 2009 entitled “Hepatitis C Virus Inhibitors”; U.S. Provisional Application Serial No. 61/241,595 filed September 11, 2009 entitled “Hepatitis C Virus Inhibitors”; U.S. Provisional Application Serial No. 61/241,617 filed September 11, 2009 entitled “Hepatitis C Virus Inhibitors”; U.S. Provisional Application Serial No. 61/241,577 filed September 11, 2009 entitled “Hepatitis C Virus Inhibitors”; U.S. Provisional Application Serial No. 61/241,598 filed September 11, 2009 entitled “Hepatitis C Virus Inhibitors”; U.S. Provisional Application Serial No.
  • a general strategy for the development of antiviral agents is to inactivate virally encoded proteins, including NS5A, that are essential for the replication of the virus.
  • the relevant patent disclosures describing the synthesis of HCV NS5A inhibitors are: US 2009/0202478; US 2009/0202483; WO 2004/014852; WO
  • the present invention relates to compounds of Formula (1-1) as illustrated above, and pharmaceutically acceptable salts thereof.
  • the present invention relates to compounds of Formula (1- I), and pharmaceutically acceptable salts thereof; wherein J is -NH-C(O)-.
  • the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein J is -NH-C(0)0-.
  • the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein J is -NH-C(O)-
  • the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein R la and R 9a , R la and R 9 , R 1 and R 9a , or R 1 and R 9 are taken together to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic.
  • the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein R lb and R 9a , or R lb and R 9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic.
  • the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein the moiety of is illustrated by one of the following core groups:
  • R 6 is as previously defined in Formula (1-1) and each of the above core groups is optionally substituted.
  • the invention relates to compounds of Formula (1-1) and pharmaceutically acceptable salts thereof; wherein R ⁇ is independently an optionally substituted C 1 -C 8 alkyl. In a further embodiment, the invention relates to compounds of Formula (1-1) and pharmaceutically acceptable salts thereof; wherein -C(0)R 6 is an a-amino acid residue.
  • the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein G is an optionally substituted five-membered heteroaryl containing one or more nitrogen atoms, and is C-attached.
  • the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein G is an optionally substituted 5/6-membered fused heteroaryl; wherein the 5-membered ring of said 5/6-membered fused heteroaryl is a heteroaryl containing one or more nitrogen atoms and wherein the 5- membered ring is C-attached to group Q, and wherein the 6-membered ring of said 5/6- membered fused heteroaryl is aryl or heteroaryl and is C-attached to group one of Ring A, L and Ring B.
  • the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein G is illustrated by one of the following heteroaryl groups:
  • the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of
  • Ring A, Ring B, G, J, L, U, W, X, Y, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 9 , R la , R lb , R 7a , R 7b , and R 9a are as previously defined in Formula (1-1).
  • the present invention relates to compounds of
  • Ring A, Ring B, G, J, L, R 1 , R 6 , R 9 , R la , R lb , and R 9a are as previously defined in Formula (1-1 ).
  • the present invention relates to compounds of Formula (l-Ia-7), and pharmaceutically acceptable salts thereof:
  • Ring A, Ring B, G, J, L, R 1 , R 6 , R 9 , R la , R lb , and R 9a are as previously defined in Formula (1-1).
  • the present invention relates to compounds of Formula (l-Ia-7), and pharmaceutically acceptable salts thereof; wherein R 6 at each occurrence is independently selected from the group consisting of C 1 -C 8 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted.
  • the present invention relates to compounds of Formula (l-Ia-7), and pharmaceutically acceptable salts thereof; wherein R 6 at each independently occurrence is independently C 1 -C 8 alkyl optionally substituted with amino, hydoxy, protected amino, or 0(C 1 -C 4 alkyl).
  • the present invention relates to compounds of Formulae (1-Ib-l ⁇ l-Ib-4), and pharmaceutically acceptable salts thereof: (1-lb-4)
  • Ring B, G, J, Q, R 1 , R 6 , R 9 , R la , R lb , and R 9a are as previously defined in Formula (1-1); in Formula (1-Ib-l), Ring A and L are each present and as previously defined in Formula (1-1); in Formula (l-Ib-2), Ring A is present and as previously defined in Formula (1-1); in Formula (l-Ib-3), L is present and as previously defined in Formula (1- I)
  • the present invention relates to compounds of Formulae (1-Ib-l ⁇ l-Ib-4), and pharmaceutically acceptable salts thereof; wherein R 6 is independently C 1 -C 8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C 1 -C 4 alkyl).
  • the present invention relates to compounds of Formula (1- Ib-1), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or optionally substituted monocyclic heteroaryl; L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (l-Ib-2), and pharmaceutically acceptable salts thereof; wherein one of Ring A and Ring B is optionally substituted phenyl or optionally substituted monocyclic heteroaryl, the other of Ring A and Ring B is optionally substituted bicyclic aryl or bicyclic heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (l-Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted bicyclic aryl or bicyclic heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (1-
  • Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (l-Ib-3), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted bicyclic aryl or bicyclic heteroaryl; L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (l-Ib-3), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (1- Ib-4), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted polycyclic aryl or polycyclic heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of
  • the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein two geminal R 7 groups can be taken together with the carbon atoms to which they are attached to form a spiro, optionally substituted cyclopropyl or a spiro, optionally substituted 5- to 6- membered heterocyclic.
  • the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein R 7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C 1 -C 4 alkyl), S(C 1 -C 4 alkyl), amino optionally substituted with one or two C 1 -C 4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C 1 -C 4 alkyl; preferably hydrogen, halogen or hydroxyl.
  • the present invention relates to com ounds of
  • Representative compounds of the present invention are those selected from compounds 1-1 to 1-374 and 1-377 to 1-382 compiled in the following tables and/or shown below:
  • any substituent or variable e.g., R , R , etc.
  • a particular location in a molecule be independent of its definitions elsewhere in that molecule.
  • U is C(R 7 ) 2
  • each of the two R 7 groups may be the same or different.
  • the present invention relates to compounds of Formula (2-1) as illustrated above, and pharmaceutically acceptable salts thereof.
  • the present invention relates to compounds of Formula (2- I), and pharmaceutically acceptable salts thereof; wherein G is -NH-C(O)-.
  • the present invention relates to compounds of Formula (2-1), and pharmaceutically acceptable salts thereof; wherein G is -NH-C(0)0-.
  • the present invention relates to compounds of Formula (2-1), and pharmaceutically acceptable salts thereof; wherein G is -NH-C(O)- NH-.
  • the present invention relates to compounds of Formula (2-1), and pharmaceutically acceptable salts thereof; wherein J is -NH-C(O)-.
  • the present invention relates to compounds of Formula (2-1), and pharmaceutically acceptable salts thereof; wherein J is -NH-C(0)0-.
  • the present invention relates to compounds of
  • the present invention relates to compounds of Formula (2-1), and pharmaceutically acceptable salts thereof;; wherein each of G and J is -NH-C(O)-.
  • the present invention relates to compounds of Formula (2-1), and pharmaceutically acceptable salts thereof; wherein R la and R 9a , R la and R 9 , R 1 and R 9a , or R 1 and R 9 are taken together to form an optionally substituted C3-C8 cycloalkyl or an optionally substituted 4- to 8-membered heterocyclic.
  • the present invention relates to compounds of Formula
  • R lb and R 9a , or R lb and R 9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic.
  • the present invention relates to compounds of Formula (2-1), and pharmaceutically acceptable salts thereof; wherein the moiety of is illustrated by the one of the following core groups:
  • R 6 is as previously defined in Formula (2-1) and each of the above core groups is optionally substituted.
  • the invention relates to compounds of Formula (2-1) and pharmaceutically acceptable salts thereof; wherein R6 is independently an optionally substituted C 1 -C 8 alkyl.
  • the invention relates to compounds of Formula (2-1) and pharmaceutically acceptable salts thereof; wherein -C(0)R 6 is an a-amino acid residue.
  • the present invention relates to compounds of Formulae (2-Ia-l ⁇ 2-Ia-2), and pharmaceutically acceptable salts thereof:
  • Ring A, Ring B, G, J, L, ,W, X, Y, R 1 , R 2 , , R 6 , R 9 , R la , R lb , R lc , and R 9a are as previously defined in Formula (2-1).
  • the present invention relates to compounds of Formulae (2-Ia-3 ⁇ 2-Ia-4),and pharmaceutically acceptable salts thereof:
  • Ring A, Ring B, G, J, L, Y, R 1 , R 2 , R 6 , R 9 , R la , R lb , R lc , and R 9a are as previously defined in Formula (2-1).
  • the present invention relates to compounds of Formula (2- Ia-5), and pharmaceutically acceptable salts thereof:
  • Ring A, Ring B, G, J, L, Y, R 1 , R 6 , R 9 , R a , R la , R lb , R lc , and R 9a are as previously defined in Formula (2-1).
  • the present invention relates to compounds of Formula (2-Ia-5), and pharmaceutically acceptable salts thereof; wherein R 6 at each occurrence is independently selected from the group consisting of C 1 -C 8 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted.
  • the present invention relates to compounds of Formula (2-Ia-5), and pharmaceutically acceptable salts thereof; wherein R 6 at each occurrence is independently C 1 -C 8 alkyl optionally substituted with amino, hydoxy, protected amino, or 0(C 1 -C 4 alkyl).
  • the present invention relates to compounds of Formulae (2-Ib-l ⁇ 2-Ib-2), and pharmaceutically acceptable salts thereof: ) (2-lb-2)
  • Ring A, Ring B, G, J, Q, R 1 , R 6 , R 9 , R la , R lb , and R 9a are as previously defined; and in Formula (2-lb-l), L is present and as previously defined in Formula (2-1).
  • the present invention relates to compounds of Formulae (2-Ib-l ⁇ 2-Ib-2), and pharmaceutically acceptable salts thereof; wherein R 6 is C 1 -C 8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C 1 -C 4 alkyl).
  • the present invention relates to compounds of Formula (2-Ib-l), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; and L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl.
  • the present invention relates to compounds of
  • the present invention relates to compounds of Formula (2-Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted bicyclic aryl or heteroaryl.
  • the present invention relates to compounds of Formula (2- Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl.
  • Representative compounds of the present invention are those selected from compounds 2-1 to 2-295 compiled in the following tables and/or shown below:
  • any linker or spacer e.g., -NH- C(O)-, -OC(0)-NH-, etc.
  • a linker e.g., -NH- C(O)-, it is meant to include -NH-C(O)- and -C(0)-NH- for attachments.
  • the present invention relates to compounds of Formula (3-1) as illustrated above, and pharmaceutically acceptable salts thereof.
  • the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein R la and R 9a , R la and R 9 , R 1 and R 9a , or R 1 and R 9 are taken together to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic.
  • the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein R lb and R 9a , or R lb and R 9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic.
  • the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein the moiety of is illustrated by one of the following core groups:
  • R 6 is as previously defined and each of the above core groups is optionally substituted.
  • the invention relates to compounds of Formula (3-1) and pharmaceutically acceptable salts thereof; wherein R6 is independently an optionally substituted C 1 -C 8 alkyl.
  • the invention relates to compounds of Formula (3-1) and pharmaceutically acceptable salts thereof; wherein -C(0)R 6 is an a-amino acid residue.
  • the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein G and J are each
  • the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein G and J are each
  • the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein one of G and J is an optionally substituted five-membered heteroaryl containing one or more nitrogen, and is each C-attached; the other of G and J is an optionally substituted 5/6-membered fused heteroaryl; wherein the 5-membered ring of said 5/6-membered fused heteroaryl is a heteroaryl containing one or more nitrogen atoms, and wherein the 6-membered ring of said 5/6-membered fused aryl is aryl or heteroaryl and is C-attached to one of Ring A, L and Ring B.
  • the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein G and J are each
  • the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein G and J are each
  • the present invention relates to compounds of F rmulae (3-Ia-l ⁇ 3-Ia-4), or a pharmaceutically acceptable salt thereof:
  • Ring A, Ring B, G, J, L, U, W, X, Y, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 9 , R la , R lb , R 7a and R 9a are as previously defined in Formula (3-1).
  • the present invention relates to compounds of F rmulae (3-Ia-5 ⁇ 3-Ia-6), and pharmaceutically acceptable salts thereof:
  • Ring A, Ring B, G, J, L, R 1 , R 6 , R 9 , R la , R lb , and R 9a are as previously defined in Formula (3-1).
  • the present invention relates to compounds of Formula (3-Ia-7), and pharmaceutically acceptable salts thereof:
  • Ring A, Ring B, G, J, L, R 1 , R 6 , R 9 , R la , R lb , and R 9a are as previously defined in Formula (3-1).
  • the present invention relates to compounds of Formula (3-Ia-7), and pharmaceutically acceptable salts thereof; wherein R 6 at each occurrence is independently selected from the group consisting of C 1 -C 8 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted.
  • the present invention relates to compounds of Formula (3-Ia-7), and pharmaceutically acceptable salts thereof; wherein R 6 at each occurrence is independently C 1 -C 8 alkyl optionally substituted with amino, hydoxy, protected amino, or 0(C 1 -C 4 alkyl).
  • the present invention relates to compounds of Formulae 3-Ib-l ⁇ 3-Ib-6), and pharmaceutically acceptable salts thereof:
  • G, J, Q, R 1 , R 6 , R 9 , R la , R lb , and R 9a are as previously defined in Formula (3-1); in Formula (3-Ib-l), Ring A, Ring B, and L are each present and as previously defined; in Formula (3-Ib-2), Ring A and Ring B are each present and as previously defined in Formula (3-1); in Formula (3-Ib-3), Ring B and L are each present and as previously defined in Formula (3-1); in Formula (3-Ib-4), Ring A and L are each present and as previously defined in Formula (3-1); in Formula (3-Ib-5), Ring B is present and as previously defined in Formula (3-1); and in Formula (3-Ib-6), L is present and as previously defined in Formula (3-1).
  • the present invention relates to compounds of Formulae (3-Ib-l ⁇ 3-Ib-6), and pharmaceutically acceptable salts thereof; wherein R 6 is independently C 1 -C 8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C 1 -C 4 alkyl).
  • the present invention relates to compounds of Formula (3-Ib-l), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl; and G and J are each independently optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (3-Ib-2), and pharmaceutically acceptable salts thereof; wherein one of Ring A and Ring B is optionally substituted phenyl or optionally substituted monocyclic heteroaryl, the other of Ring A and Ring B is optionally substituted bicyclic aryl or heteroaryl; and G and J are each independently optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (3-Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted bicyclic aryl or bicyclic heteroaryl; and G and J are each independently optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (3-Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; and G and J are each independently optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of
  • Ring B or Ring A is optionally substituted bicyclic aryl or bicyclic heteroaryl
  • L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl
  • G and J are each independently optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of
  • Ring B or Ring A is optionally substituted phenyl or monocyclic heteroaryl
  • L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl
  • G and J are each independently optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of
  • the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein two geminal R 7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkenyl and 3- to 8-membered heterocyclic, each optionally substituted.
  • the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein two geminal R 7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted cyclopropyl or a spiro, optionally substituted 5- to 6- membered heterocyclic.
  • the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein R 7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C 1 -C 4 alkyl), S(C 1 -C 4 alkyl), amino optionally substituted with one or two C 1 -C 4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C 1 - C 4 alkyl; preferably hydrogen, halogen or hydroxy.
  • the present invention relates to com ounds of
  • Representative compounds of the present invention are those selected from compounds 3-1 to 3-347compiled in the following tables and/or shown below:
  • any substituent or variable e.g., R , R , etc.
  • each of the two R 7 groups may be the same or different.
  • the drawn formula of any linker or spacer e.g., -NH- C(O)-, -OC(0)-NH-, etc.
  • linker e.g., -NH- C(O)-, -OC(0)-NH-, etc.
  • a linker is -NH- C(O)-, it is meant to include -NH-C(O)- and -C(0)-NH- for attachments.
  • the present invention relates to compounds of Formula (4-1) as illustrated above, and pharmaceutically acceptable salts thereof.
  • the present invention relates to compounds of Formula (4-1), and pharmaceutically acceptable salts thereof; wherein two geminal R 7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkenyl and 3- to 8-membered heterocyclic, each optionally substituted.
  • the present invention relates to compounds of Formula (4-1), and pharmaceutically acceptable salts thereof; wherein two geminal R 7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted cyclopropyl or a spiro, optionally substituted 5- to 6- membered heterocyclic.
  • the present invention relates to com ounds of Formula (4-
  • R la and R 9a , R la and R 9 , R ld and R 9a , or R ld and R 9 are taken together with the carbon atom to which they are attached to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic.
  • R lb and R 9a are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic.
  • the present invention relates to compounds of Formula
  • R 6 is as previously defined in Formula (4-1) and each of the above core groups is optionally substituted.
  • the invention relates to compounds of Formula (4-1) and pharmaceutically acceptable salts thereof; wherein R ⁇ is independently an optionally substituted C 1 -C 8 alkyl.
  • the invention relates to compounds of Formula (4-1) and pharmaceutically acceptable salts thereof; wherein -C(0)R 6 is an a-amino acid residue.
  • the present invention relates to compounds of Formula (4-1), and pharmaceutically acceptable salts thereof; wherein G is an optionally substituted five-membered heteroaryl containing one or more nitrogen atoms, and is C-attached.
  • the present invention relates to compounds of Formula (4-1), and pharmaceutically acceptable salts thereof; wherein G is an optionally substituted 5/6-membered fused heteroaryl; wherein the 5-membered ring of said 5/6-membered fused heteroaryl is a heteroaryl containing one or more nitrogen atoms and wherein the 5- membered ring is C-attached to group Q, and wherein the 6-membered ring of said 5/6- membered fused heteroaryl is aryl or heteroaryl and is C-attached to one of Ring A, L and Ring B.
  • the present invention relates to compounds of Formula (4-1), and pharmaceutically acceptable salts thereof; wherein G is illustrated by one of the following heteroaryl groups:
  • the present invention relates to compounds of Formula (4-1), and pharmaceutically acceptable salts thereof; wherein G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of
  • Ring A, Ring B, G, J, L, U, W, X, Y, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 9 , R la , R lb , R ld , R 7a , R 7b and R 9a are as previously defined in Formula (4-1).
  • the present invention relates to compounds of
  • Ring A, Ring B, G, J, L, U, W, X, Y, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 9 , R la , R lb , R lc , R ld , R 7a , R 7b and R 9a are as previously defined in Formula (4-1).
  • the present invention relates to compounds of
  • Ring A, Ring B, G, L, R 6 , R 7a and R 7b are as previously defined in Formula (4-1).
  • the present invention relates to compounds of
  • Ring A, Ring B, G, L, R 6 , R 7a and R 7b are as previously defined in Formula (4-1).
  • the present invention relates to compounds of
  • R 6 at each occurrence is independently selected from the group consisting of C 1 -C 8 alkyl, C 2 - C 8 alkenyl, C 2 -C 8 alkynyl, C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted.
  • the present invention relates to compounds of Formula (4-Ia-13 ⁇ 4-Ia-16), and pharmaceutically acceptable salts thereof; wherein R 6 at each occurrence is independently C 1 -C 8 alkyl optionally substituted with amino, hydoxy, protected amino, or 0(C 1 -C 4 alkyl).
  • the present invention relates to compounds of Formulae (4-Ib- ⁇ 4-Ib-4), and pharmaceutically acceptable salts thereof;
  • Ring B, G, Q, U, R 1 , R 6 , R 7a and R 7b are as previously defined in Formula (4-1); in Formula (4-Ib-l), A and L are each present and as previously defined; in Formula (4-1). in Formula (4-Ib-2), Ring A is present and as previously defined in Formula (4-1); and in Formula (4-Ib-3), L is present and as previously defined in Formula (4-1).
  • the present invention relates to compounds of Formula (4-Ib-l ⁇ 4-Ib-4), and pharmaceutically acceptable salts thereof; wherein R 6 is independently C 1 -C 8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C 1 -C 4 alkyl).
  • the present invention relates to compounds of Formula (4-Ib-l), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (4-Ib-2), and pharmaceutically acceptable salts thereof; wherein one of Ring A and Ring B is optionally substituted phenyl or optionally substituted monocyclic heteroaryl, the other of Ring A and Ring B is optionally substituted bicyclic aryl or heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (4-Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted bicyclic aryl or heteroaryl; and G is optionally substituted imidazolyl.
  • the present invention relates to compounds of
  • Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; and G is optionally substituted benzimidazolyl.
  • the present invention relates to compounds of Formula (4-Ib-3), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted bicyclic aryl or heteroaryl; L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (4-Ib-3), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (4-Ib-4), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted polycyclic aryl or heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (4-Ib- 4), and pharmaceutically acceptable salts thereof; wherein R 7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C 1 -C 4 alkyl), S(C 1 -C 4 alkyl), amino optionally substituted with one or two C 1 -C 4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C 1 - C 4 alkyl; preferably hydrogen, halogen or hydroxyl.
  • Representative compounds of the present invention are those selected from compounds 4-1 to 4-348 compiled in the following tables and/or are shown below:
  • any substituent or variable e.g., R 3 , R 7 , etc.
  • each of the two R 7 groups may be the same or different.
  • the present invention relates to compounds of Formula (5-1) as illustrated above, and pharmaceutically acceptable salts thereof.
  • the present invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; wherein R la and R 9a , R la and R 9 , R 1 and R 9a , or R 1 and R 9 are taken together to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic.
  • the present invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; wherein R lb and R 9a , or R lb and R 9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic.
  • the present invention relates to compounds of Formula (5-
  • R 6 is as previously defined and each of the above core groups is optionally substituted.
  • the invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; wherein R ⁇ is independently an optionally substituted C 1 -C 8 alkyl.
  • the invention relates to compounds of Formula (5-1) and pharmaceutically acceptable salts thereof; wherein -C(0)R 6 is an a-amino acid residue.
  • the present invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; G is an optionally substituted five- membered heteroaryl containing one or more nitrogen atoms, and is C-attached. In yet another embodiment, the present invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; wherein G is an optionally substituted 5/6-membered fused heteroaryl; wherein the 5-membered ring of said 5/6-membered fused heteroaryl is a heteroaryl containing one or more nitrogen and wherein the 5-membered ring is C-attached to group Q, and wherein the 6-membered ring of said 5/6-membered fused heteroaryl is aryl or heteroaryl and is C-attached to one of Ring A, L and Ring B.
  • the present invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; wherein G is illustrated by one of the following heteroaryl groups:
  • the present invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; wherein G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of
  • Ring A, Ring B, G, L, U, W, X, Y, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 9 , R la , R lb , R lc , and R 9a are as previously defined in Formula (5-1).
  • the present invention relates to compounds of
  • Ring A, Ring B, G, L, U, W, X, Y, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 9 , R la , R lb , R lc , and R 9a are as previously defined in Formula (5-1).
  • the present invention relates to compounds of Formulae (5-Ia-8 ⁇ 5-Ia-9), and pharmaceutically acceptable salts thereof;
  • Ring A, Ring B, G, L, R 1 , R 6 , R 9 , R la , R lb , R lc , and R 9a are as previously defined in Formula (5-1).
  • the present invention relates to compounds of Formula (5-Ia-ll), and pharmaceutically acceptable salts thereof;
  • Ring A, Ring B, G, L, R 1 , R 6 , R 9 , R la , R lb , R lc , and R 9a are as previously defined in Formula (5-1).
  • the present invention relates to compounds of Formula (5-Ia-ll), and pharmaceutically acceptable salts thereof; wherein R 6 at each occurrence is independently selected from the group consisting of C 1 -C 8 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl, C 3 -C 8 cycloalkyl, C 3 -C 8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted.
  • the present invention relates to compounds of Formula (5-Ia-ll), and pharmaceutically acceptable salts thereof; wherein R 6 at each occurrence is independently C 1 -C 8 alkyl optionally substituted with amino, hydoxy, protected amino, or 0(C 1 -C 4 alkyl).
  • the present invention relates to compounds of Formulae (5-Ib-l ⁇ 5-Ib-4), and pharmaceutically acceptable salts thereof;
  • Ring B, G, Q, R 1 , R 6 , R 9 , R la , R lb , R lc , and R 9a are as previously defined in Formula (5-1); in Formula (5-Ib-l), Ring A and L are each present and as previously defined in Formula (5-1); in Formula (5-Ib-2), Ring A is present and as previously defined in Formula (5-1); and in Formula (5-Ib-3), L is present and as previously defined in Formula (5-1).
  • the present invention relates to compounds of Formulae (5-Ib-l ⁇ 5-Ib-4), and pharmaceutically acceptable salts thereof; wherein R 6 at each occurrence is independently C 1 -C 8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C 1 -C 4 alkyl).
  • the present invention relates to compounds of Formula (5-Ib-l), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (5-Ib-2), and pharmaceutically acceptable salts thereof; wherein one of Ring A and Rng B is optionally substituted phenyl or optionally substituted monocyclic heteroaryl, the other of Ring A and Ring B is optionally substituted bicyclic aryl or heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (5-Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted bicyclic aryl or heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (5-Ib-2) and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (5-Ib-3), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted bicyclic aryl or heteroaryl; L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (5-Ib-3), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formula (5-Ib-4), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted polycyclic aryl or heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
  • the present invention relates to compounds of Formulae (5-Ib-5 ⁇ 5-Ib-8), and pharmaceutically acceptable salts thereof:
  • Ring B, Q, V, R 1 , R 6 , R 9 , R la , R lb , R lc , and R 9a are as previously defined in Formula (5-1); in Formula (5-Ib-5), Ring A and L are each present and as previously defined in Formula (5-1); in Formula (5-Ib-6), Ring A is present and as previously defined in Formula (5-1); and in Formula (5-Ib-7), L is present and as previously defined in Formula (5-1).
  • the present invention relates to compounds of Formula (5-Ib-5), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl.
  • the present invention relates to compounds of Formula (5-Ib-6), and pharmaceutically acceptable salts thereof; wherein one of Ring A and Ring B is optionally substituted phenyl or optionally substituted monocyclic heteroaryl, the other of Ring A and Ring B is optionally substituted bicyclic aryl or heteroaryl.
  • the present invention relates to compounds of Formula (5-Ib-6), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl.
  • the present invention relates to compounds of Formula (5-Ib-7), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted bicyclic aryl or heteroaryl; L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl.
  • the present invention relates to compounds of
  • Ring B is optionally substituted phenyl or monocyclic heteroaryl
  • L is optionally substituted C 2 -C 4 alkenyl or optionally substituted C 2 -C 4 alkynyl.
  • the present invention relates to compounds of Formula (5-Ib-8), or a pharmaceutically acceptable salt thereof; wherein Ring B is optionally substituted polycyclic aryl or heteroaryl.
  • the present invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; wherein two geminal R 7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C 3 -C 8 cycloalkyl, C3-C 8 cycloalkenyl and 3- to 8-membered heterocyclic, each optionally substituted.
  • the present invention relates to compounds of
  • the present invention relates to compounds of Formula
  • R 7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy,
  • the present invention relates to com ounds of
  • Representative compounds of the present invention are those selected from compounds 5-1 to 5-394 compiled in the following tables and/or shown below:
  • any substituent or variable e.g., R 3 , R 7 , etc.
  • each of the two R 7 groups may be the same or different.
  • any linker or spacer e.g., -NH- C(O)-, -OC(0)-NH-, etc.
  • a linker e.g., -NH- C(O)-, it is meant to include -NH-C(O)- and -C(0)-NH- for attachments.
  • the description of the present invention herein should be construed in congruity with the laws and principals of chemical bonding. In some instances it may be necessary to remove a hydrogen atom in order to accommodate a substitutent at any given location. It will be yet appreciated that the compounds of the present invention may contain one or more asymmetric carbon atoms and may exist in racemic, diastereoisomeric, and optically active forms. It will still be appreciated that certain compounds of the present invention may exist in different tautomeric forms. All tautomers are contemplated to be within the scope of the present invention.
  • the compounds encompassed by the present invention are those that are suitably stable for use as pharmaceutical agent.
  • references herein to therapy and/or treatment includes, but is not limited to, prevention, retardation, prophylaxis, therapy and/or cure of the disease. It will further be appreciated that references herein to treatment or prophylaxis of HCV infection includes treatment or prophylaxis of HCV-associated disease such as liver fibrosis, cirrhosis and hepatocellular carcinoma.
  • a further embodiment of the present invention includes pharmaceutical compositions comprising any single compound a combination of two or more compounds delineated herein, or a pharmaceutically acceptable salt thereof, with a pharmaceutically acceptable carrier or excipient.
  • Yet a further embodiment of the present invention is a pharmaceutical composition
  • a pharmaceutical composition comprising any single compound or a combination of two or more compounds delineated herein, or a pharmaceutically acceptable salt thereof, in combination with one or more agents known in the art, with a pharmaceutically acceptable carrier or excipient.
  • 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.
  • agents to be administered in combination with a compound or combination of compounds of the present invention include therapies for disease caused by HCV infection that suppresses HCV viral replication by direct or indirect mechanisms.
  • agents include, but are not limited to, host immune modulators (for example, interferon-alpha, pegylated interferon-alpha, consensus interferon, interferon-beta, interferon-gamma, CpG oligonucleotides and the like); antiviral compounds that inhibit host cellular functions such as inosine monophosphate dehydrogenase (for example, ribavirin and the like); cytokines that modulate immune function (for example, interleukin 2, interleukin 6, and interleukin 12); a compound that enhances the development of type 1 helper T cell response; interfering R A; anti-sense R A; vaccines comprising HCV antigens or antigen adjuvant combinations directed against HCV; 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
  • compositions of the present invention may further comprise other inhibitor(s) of targets in the HCV life cycle, including, but not limited to, helicase, polymerase, metalloprotease, NS4A protein, NS5A protein, and internal ribosome entry site (IRES).
  • targets in the HCV life cycle including, but not limited to, helicase, polymerase, metalloprotease, NS4A protein, NS5A protein, and internal ribosome entry site (IRES).
  • one embodiment of the present 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 or more antiviral agents, or a combination thereof, with a therapeutically effective amount of a compound or combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof.
  • Examples of the host immune modulator include, but are 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.
  • An example of the RNA-containing virus includes, but is not limited to, hepatitis C virus (HCV).
  • a further embodiment of the present 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 present invention, or a pharmaceutically acceptable salt thereof.
  • An example of the RNA-containing virus includes, but is not limited to, hepatitis C virus (HCV).
  • Yet another embodiment of the present 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 present invention, or a pharmaceutically acceptable salt 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.
  • An example of the R A-containing virus includes, but is not limited to, hepatitis C virus (HCV).
  • a further embodiment of the present 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 present invention, or a pharmaceutically acceptable salt 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-114, 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.
  • An example of the RNA- containing virus includes, but is not limited to, hepatitis C virus (HCV).
  • 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 present invention provides the use of a compound or a combination of compounds of the invention, or a pharmaceutically acceptable salt thereof, and one or more agents selected from the group consisting of a host immune modulator and a second or more antiviral agents, 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.
  • the host immune modulator examples include, but are 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 present 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 thereof.
  • 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
  • compositions can be used for inhibiting the replication of an R A-containing virus, particularly Hepatitis C virus (HCV), by contacting said virus with said pharmaceutical composition.
  • R A-containing virus particularly Hepatitis C virus (HCV)
  • HCV Hepatitis C virus
  • compositions are useful for the treatment or prevention of an infection caused by an RNA-containing virus, particularly Hepatitis C virus (HCV).
  • a still further embodiment 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 thereof, and one or more agents as defined hereinabove, with a pharmaceutically acceptable carrier.
  • an RNA-containing virus particularly a hepatitis C virus (HCV)
  • 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.
  • cytochrome P450 monooxygenase inhibitor also referred to herein as a CYP inhibitor
  • the cytochrome P450 monooxygenase inhibitor would be in an amount effective to inhibit metabolism of the compounds of this invention.
  • the CYP inhibitor is administered in an amount such that the bioavailiablity of the protease inhibitor is increased in comparison to the bioavailability in the absence of the CYP inhibitor.
  • the invention provides methods for improving the
  • one embodiment of this invention provides a method for administering an inhibitor of CYP3 A4 and a compound of the invention.
  • Another embodiment of this invention provides a method for administering a compound of the invention and an inhibitor of isozyme 3A4 ("CYP3A4"), isozyme 2C19 (“CYP2C19”), isozyme 2D6 (“CYP2D6"), isozyme 1 A2 (“CYP1 A2”), isozyme 2C9 (“CYP2C9”), or isozyme 2E1 ("CYP2E1").
  • the CYP inhibitor preferably inhibits CYP3A4. Any CYP inhibitor that improves the pharmacokinetics of the relevant NS3/4A protease may be used in a method of this invention.
  • CYP inhibitors include, but are not limited to, ritonavir (see, for example, WO 94/14436), ketoconazole, troleandomycin, 4-methyl pyrazole, cyclosporin, clomethiazole, cimetidine, itraconazole, fluconazole, miconazole, fluvoxamine, fluoxetine, nefazodone, sertraline, indinavir, nelfmavir, amprenavir, fosamprenavir, saquinavir, lopinavir, delavirdine, erythromycin, VX-944, and VX-497.
  • Preferred CYP inhibitors include ritonavir, ketoconazole, troleandomycin, 4-methyl pyrazole, cyclosporin, and clomethiazole.
  • the pharmaceutical pack further comprises one or more of additional agent as described herein.
  • the additional agent or agents may be provided in the same pack or in separate packs.
  • kits for a patient to use in the treatment of HCV infection or in the prevention of HCV infection comprising: a single or a plurality of pharmaceutical formulation of each pharmaceutical component; a container housing the pharmaceutical formulation(s) during storage and prior to administration; and instructions for carrying out drug administration in a manner effective to treat or prevent HCV infection.
  • kits for the simultaneous or sequential administration of a compound of the invention and a CYP inhibitor (and optionally an additional agent) or derivatives thereof are prepared in a conventional manner.
  • a kit will comprise, e.g. a composition of each inhibitor and optionally the additional agent(s) in a pharmaceutically acceptable carrier (and in one or in a plurality of pharmaceutical formulations) and written instructions for the simultaneous or sequential administration.
  • a packaged kit contains one or more dosage forms for self administration; a container means, preferably sealed, for housing the dosage forms during storage and prior to use; and instructions for a patient to carry out drug administration.
  • the instructions will typically be written instructions on a package insert, a label, and/or on other components of the kit, and the dosage form or forms are as described herein.
  • Each dosage form may be individually housed, as in a sheet of a metal foil- plastic laminate with each dosage form isolated from the others in individual cells or bubbles, or the dosage forms may be housed in a single container, as in a plastic bottle.
  • the present kits will also typically include means for packaging the individual kit components, i. e., the dosage forms, the container means, and the written instructions for use.
  • Such packaging means may take the form of a cardboard or paper box, a plastic or foil pouch, etc.
  • 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
  • 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, cortico-steroids, colony-stimulating factors, chemotactic factors, etc.
  • aryl refers to a mono- or polycyclic carbocyclic ring system including, but not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, and idenyl.
  • a polycyclic aryl is a polycyclic ring system that comprises at least one aromatic ring.
  • Polycyclic aryls can comprise fused rings, covalently attached rings or a
  • heteroaryl refers to a mono- or polycyclic ring system comprising at least one aromatic ring having one or more ring atom selected from S, O and N; and the remaining ring atoms are carbon, 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, and quinoxalinyl.
  • a polycyclic heteroaryl can comprise fused rings, covalently attached rings or a combination thereof.
  • any of the aryls, substituted aryls, heteroaryls and substituted heteroaryls described herein, can be any aromatic group.
  • Aromatic groups can be substituted or unsubstituted.
  • bicyclic aryl or "bicyclic heteroaryl” refers to a ring system consisting of two rings wherein at least one ring is aromatic; and they can be fused or covalently attached.
  • tricyclic aryl or "tricyclic heteroaryl” refers to a ring system consisting of three rings wherein at least one ring is aromatic.
  • C 1 -C 4 alkyl refers to saturated, straight- or branched-chain hydrocarbon radicals containing between one and four, one and six, one and eight carbon atoms, or the like, respectively.
  • Examples of C 1 -Cs alkyl radicals include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, tert-butyl, neopentyl, n-hexyl, heptyl and octyl radicals.
  • C 2 -C 8 alkenyl refers to straight- or branched-chain hydrocarbon radicals containing from two to eight, or two to four carbon atoms, or the like, having at least one carbon-carbon double bond by the removal of a single hydrogen atom.
  • Alkenyl groups include, but are not limited to, for example, ethenyl, propenyl, butenyl, l-methyl-2-buten-1-yl, heptenyl, octenyl, and the like.
  • C 2 -C 8 alkynyl refers to straight- or branched-chain hydrocarbon radicals containing from two to eight, or two to four carbon atoms, or the like, having at least one carbon-carbon triple bond by the removal of a single hydrogen atom.
  • Representative alkynyl groups include, but are not limited to, for example, ethynyl, 1-propynyl, 1-butynyl, heptynyl, octynyl, and the like.
  • C 3 -C8-cycloalkyl refers to a monocyclic or polycyclic saturated carbocyclic ring compound, and the carbon atoms may be optionally oxo-substituted.
  • C 3 -C8-cycloalkyl examples include, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopentyl and cyclooctyl; and examples of Cs-Cycycloalkyl include, but not limited to, cyclopentyl, cyclohexyl, bicyclo [2.2.1] heptyl, and the like.
  • C 3 -C 8 cycloalkenyl or “C5-C7 cycloalkenyl,” as used herein, refers to monocyclic or polycyclic carbocyclic ring compound having at least one carbon-carbon double bond, and the carbon atoms may be optionally oxo-substituted.
  • C 3 -C 8 cycloalkenyl examples include, but not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, and the like; and examples of C5-C7 cycloalkenyl include, but not limited to, cyclopentenyl, cyclohexenyl, cycloheptenyl, and the like.
  • arylalkyl refers to an aryl-substituted alkyl group. More preferred arylalkyl groups are aryl-C 1 -C 6 -alkyl groups.
  • heteroarylalkyl refers to a heteroaryl-substituted alkyl group. More preferred heteroarylalkyl groups are heteroaryl-C 1 -C6-alkyl groups.
  • any alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclic and cycloalkenyl moiety described herein can also be an aliphatic group or an alicyclic group.
  • an "aliphatic” group is a non-aromatic moiety comprised of any combination of carbon atoms, hydrogen atoms, halogen atoms, oxygen, nitrogen or other atoms, and optionally contains one or more units of unsaturation, e.g., double and/or triple bonds.
  • Examples of aliphatic groups are functional groups, such as, O, OH, NH, NH 2 , C(O), S(0) 2 , C(0)0, C(0)NH, OC(0)0, OC(0)NH, OC(0)NH 2 , S(0) 2 NH, S(0) 2 NH 2 , NHC(0)NH 2 , NHC(0)C(0)NH, NHS(0) 2 NH, NHS(0) 2 NH 2 , C(0)NHS(0) 2 ,
  • C(0)NHS(0) 2 NH or C(0)NHS(0) 2 NH 2 groups comprising one or more functional groups, non-aromatic hydrocarbons (optionally substituted), and groups wherein one or more carbons of a non-aromatic hydrocarbon (optionally substituted) is replaced by a functional group.
  • Carbon atoms of an aliphatic group can be optionally oxo- substituted.
  • An aliphatic group may be straight chained, branched, cyclic, or a
  • aliphatic groups expressly include, for example, alkoxyalkyls, polyalkoxyalkyls, such as polyalkylene glycols, polyamines, and
  • Aliphatic groups may be optionally substituted.
  • a linear aliphatic group is a non-cyclic aliphatic group. It is to be understood that when an aliphatic group or a linear aliphatic group is said to "contain” or “include” or “comprise” one or more specified functional groups, the aliphatic group can be selected from one or more of the specified functional groups or a combination thereof, or a group wherein one or more carbons of a non-aromatic hydrocarbon (optionally substituted) is replaced by a specified functional group.
  • the linear aliphatic group can be represented by the formula M-V'-M', where M and M' are each independently absent or an alkyl, alkenyl or alkynyl, each optionally substituted, and V is a functional group.
  • V is selected from the group consisting of C(O), S(0) 2 , C(0)0,
  • an exemplary linear aliphatic group is an alkyl, alkenyl or alkynyl, each optionally substituted, which is interrupted or terminated by a functional group such as described herein.
  • alicyclic denotes a monovalent group derived from a monocyclic or bicyclic saturated carbocyclic ring compound by the removal of a single hydrogen atom, and the carbon atoms may be optionally oxo-substituted. Examples include, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo
  • heterocyclic or “heterocycloalkyl” can be used interchangeably and referred to a non-aromatic ring or a bi- or tri-cyclic group fused system, where (i) each ring system contains at least one heteroatom independently selected from oxygen, sulfur and nitrogen, (ii) each ring system can be saturated or unsaturated, (iii) the nitrogen and sulfur heteroatoms may optionally be oxidized, (iv) the nitrogen heteroatom may optionally be quaternized, (v) any of the above rings may be fused to an aromatic ring, and (vi) the remaining ring atoms are carbon atoms which may be optionally oxo-substituted.
  • heterocycloalkyl groups include, but are not limited to, 1,3-dioxolane, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, quinoxalinyl, pyridazinonyl, and tetrahydrofuryl. Such heterocyclic groups may be further substituted. Heteroaryl or heterocyclic groups can be C-attached or N-attached (where possible).
  • any alkyl, alkenyl, alkynyl, alicyclic, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclic, aliphatic moiety or the like, described herein can also be a divalent or multivalent group when used as a linkage to connect two or more groups or substituents, which can be at the same or different atom(s).
  • substituted when used with alkyl, alkenyl, alkynyl, alicyclic, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclic, or aliphatic as described herein refers to substitution by independent replacement of one, two, or three or more of the hydrogen atoms thereon with substituents including, but not limited to, -F, -CI, -Br, -I, - OH, protected hydroxy, -N0 2 , -N 3 , -CN, -NH 2 , protected amino, oxo, thioxo, -NH-C1-C12- alkyl, -NH-C 2 -C 8 -alkenyl, -NH-C 2 -C 8 -alkynyl, -NH-C 3 -C 12 -cycloalkyl, -NH-aryl, -NH- heteroaryl, -NH-hetero
  • halogen refers to an atom selected from fluorine, chlorine, bromine and iodine.
  • hydrox includes hydrogen and deuterium.
  • recitation of an atom includes other isotopes of that atom so long as the resulting compound is pharmaceutically acceptable.
  • hydroxy activating group refers to a labile chemical moiety which is known in the art to activate a hydroxyl group so that it will depart during synthetic procedures such as in a substitution or an elimination reaction.
  • hydroxyl activating group include, but not limited to, mesylate, tosylate, triflate, p- nitrobenzoate, phosphonate and the like.
  • activated hydroxyl refers to a hydroxy group activated with a hydroxyl activating group, as defined above, including mesylate, tosylate, triflate, p-nitrobenzoate, phosphonate groups, for example.
  • hydroxy protecting group refers to a labile chemical moiety which is known in the art to protect a hydroxyl 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 art 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).
  • hydroxyl protecting groups include benzyloxycarbonyl, 4- methoxybenzyloxycarbonyl, tert-butoxycarbonyl, isopropoxycarbonyl,
  • protected hydroxy refers to a hydroxy group protected with a hydroxy protecting group, as defined above, including benzoyl, acetyl,
  • carbonyl protecting group refers to a labile chemical moiety which is known in the art to protect a carbonyl group against undesired reactions during synthetic procedures. After said synthetic procedure(s) the carbonyl protecting group as described herein may be selectively removed.
  • Carbonyl protecting groups as known in the art 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).
  • carbonyl protecting groups include acetals, ketals, cyclic acetals, cyclic ketals, mono- or dithio acetals, mono- or dithioketals, optionally substituted hydrazones or oximes.
  • protected carbonyl refers to a carbonyl group protected with a carbonyl protecting group, as defined above, including dimethyl acetal, 1,3- dioxolane, 1,3-dioxane, S,S'-dimethylketal, 1,3-dithiane, 1,3-dithiolane, 1,3-oxathiolane, N,N-dimethylhydrazone, oxime, for example.
  • 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 art 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, methoxycarbonyl, t- butoxycarbonyl, 9-fluorenylmethoxycarbonyl, benzyloxycarbonyl, and the like.
  • protected amino refers to an amino group protected with an amino protecting group as defined above.
  • substituted amino refers to substitution by replacement of one or two hydrogen atoms of -NH 2 with substituents independently selected from the group consisting of optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cycloalkyl, optionally substituted heteroaryl, and optionally substituted heterocyclic; alternatively, when disubstituted, the two substitutents can be optionally taken together with the nitrogen atom to which they are attached to form an optionallysubstituted heterocyclic group.
  • leaving group means a functional group or atom which can be displaced by another functional group or atom in a substitution reaction, such as a nucleophilic substitution reaction.
  • representative leaving groups include chloro, bromo and iodo groups; sulfonic ester groups, such as mesylate, tosylate, brosylate, nosylate and the like; hydroxy; imidazolyl; and acyloxy groups, such as acetoxy, trifluoroacetoxy 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.
  • protic solvent refers to a solvent that tends to provide protons, such as an alcohol, for example, methanol, ethanol, propanol, isopropanol, butanol, t-butanol, and the like.
  • alcohol for example, methanol, ethanol, propanol, isopropanol, butanol, t-butanol, and the like.
  • solvents are well known to those skilled in the art, and it will be obvious to those skilled in the art that 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
  • 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
  • subject refers to an animal. Preferably the animal is a mammal. More preferably the mammal is a human. A subject also refers to, for example, dogs, cats, horses, cows, pigs, guinea pigs, fish, birds and the like.
  • the compounds of this invention may be modified by appending appropriate functionalities to enhance selective biological properties. Such modifications are known in the art and may 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.
  • 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.
  • 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 or carbon- heteroatom double bond depicted arbitrarily herein as trans may be cis, trans, or a mixture of the two in any proportion.
  • Certain compounds of the present invention may also exist in different stable conformational forms which may be separable. Torsional asymmetry due to restricted rotation about an asymmetric single bond, for example because of steric hindrance or ring strain, may permit separation of different conformers.
  • the present invention includes each conformational isomer of these compounds and mixtures thereof.
  • the term "pharmaceutically acceptable salt” refers to those salts 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. For example, S. M. Berge, et al. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66:
  • 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.
  • suitable organic acid examples include, but are not limited to, nontoxic acid addition salts are 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,
  • 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.
  • ester refers to esters 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
  • prodrugs refers to those prodrugs of the compounds 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
  • Prodrug means a compound which is convertible in vivo by metabolic means (e.g. by hydrolysis) to a compound of the 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).
  • the present invention also relates to solvates of the compounds of Formula (1-1),
  • This invention also encompasses pharmaceutical compositions containing, and methods of treating viral infections through administering, pharmaceutically acceptable prodrugs of compounds of the invention.
  • compounds of the invention having free amino, amido, hydroxy or carboxylic groups can be converted into prodrugs.
  • Prodrugs include compounds wherein an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues is covalently joined through an amide or ester bond to a free amino, hydroxy or carboxylic acid group of compounds of the invention.
  • the amino acid residues include, but are not limited to, the 20 naturally occurring amino acids commonly designated by three letter symbols and also includes 4- hydroxyproline, hydroxylysine, demosine, isodemosine, 3-methylhistidine, norvalin, beta- alanine, gamma-aminobutyric acid, citrulline, homocysteine, homoserine, ornithine and methionine sulfone. Additional types of prodrugs are also encompassed. For instance, free carboxyl groups can be derivatized as amides or alkyl esters.
  • Free hydroxy groups may be derivatized using groups including but not limited to hemisuccinates, phosphate esters, dimethylaminoacetates, and phosphoryloxymethyloxycarbonyls, as outlined in Advanced Drug Delivery Reviews, 1996, 19, 115.
  • Carbamate prodrugs of hydroxy and amino groups are also included, as are carbonate prodrugs, sulfonate esters and sulfate esters of hydroxy groups.
  • acyl group may be an alkyl ester, optionally substituted with groups including but not limited to ether, amine and carboxylic acid functionalities, or where the acyl group is an amino acid ester as described above, are also encompassed.
  • Prodrugs of this type are described in J. Med. Chem. 1996, 39, 10. Free amines can also be derivatized as amides, sulfonamides or phosphonamides. All of these prodrug moieties may incorporate groups including but not limited to ether, amine and carboxylic acid functionalities.
  • compositions of the present invention comprise a
  • the term "pharmaceutically acceptable carrier or excipient” 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 as propylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminun hydroxide; algin
  • 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 includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, 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
  • 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.
  • the 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 that 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 and other coatings well known in the pharmaceutical formulating art. 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 that 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.
  • 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.
  • a therapeutic composition of the invention is formulated and administered to the patient in solid or liquid particulate form by direct administration e.g., inhalation into the respiratory system.
  • Solid or liquid particulate forms of the active compound prepared for practicing the present invention include particles of respirable size: that is, particles of a size sufficiently small to pass through the mouth and larynx upon inhalation and into the bronchi and alveoli of the lungs. Delivery of aerosolized therapeutics, particularly aerosolized antibiotics, is known in the art (see, for example U.S. Pat. No. 5,767,068 to VanDevanter et al, U.S. Pat. No.
  • An inhibitory amount or dose of the compounds of the present invention may range from about 0.01 mg/Kg to about 500 mg/Kg, alternatively from about 0.1 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 patient such as a human or another animal by administering to the subject a therapeutically effective amount of a compound of the 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.
  • terapéuticaally effective amount of a compound of the invention means an amount of the compound which confers a therapeutic effect on the treated subject, at a reasonable benefit/risk ratio applicable to any medical treatment.
  • the therapeutic effect may be objective (i.e., measurable by some test or marker) or subjective (i.e., subject gives an indication of or feels an effect).
  • inhibitory amount of a compound of the present invention means a sufficient amount to decrease the viral load in a biological sample or a subject (e.g., resulting in at least 10%, preferably at least 50%, more preferably at least 80%, and most preferably at least 90%> or 95%, reduction in viral load). 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), as used herein, means a substance of biological origin intended for administration to a subject.
  • 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.
  • Effective doses will also vary depending on route of administration, as well as the possibility of co-usage with other agents. 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 therapeutically effective dose level 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 contemporaneously with the specific compound employed; and like factors well known in the medical arts.
  • the total daily dose of the compounds of this invention administered to a human or other animal 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.
  • 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.
  • Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
  • compositions of this invention comprise a combination of a compound of the Formula (1-1), Formula (2-1), Formula (3-1), Formula (4-1) and Formula (5-1), described herein and one or more additional therapeutic or prophylactic agents
  • both the compound and the additional agent should be present at dosage levels of between about 1 to 100%, and more preferably between about 5 to 95% of the dosage normally
  • agents administered in a monotherapy regimen.
  • the additional agents may be administered separately, as part of a multiple dose regimen, from the compounds of this invention.
  • those agents may be part of a single dosage form, mixed together with the compounds of this invention in a single composition.
  • additional therapeutic or prophylactic agents include, but are not limited to, immune therapies (eg. interferon), therapeutic vaccines, antifibrotic agents, anti-inflammatory agents such as corticosteroids or NSAIDs, bronchodilators such as beta- 2 adrenergic agonists and xanthines (e.g. theophylline), mucolytic agents, anti- muscarinics, anti-leukotrienes, inhibitors of cell adhesion (e.g.
  • compositions according to the invention may also be used in combination with gene replacement therapy.
  • Drug resistance most typically occurs by mutation of a gene that encodes for a protein such as an enzyme used in viral replication, and most typically in the case of HCV, RNA polymerase, protease, or helicase.
  • a compound of the present invention can also be administered in combination or alternation with antiviral agent.
  • Examplary antiviral agents include ribavarin, interferon, interleukin or a stabilized prodrug of any of them. More broadly described, the compound can be administered in combination or alternation with any of the anti-HCV drugs listed in Table 53 below.
  • cyanoborohydride NaN(TMS)2 for sodium bis(trimethylsilyl)amide; NaCl for sodium chloride; NaH for sodium hydride; NaHC0 3 for sodium bicarbonate or sodium hydrogen carbonate; Na 2 C0 3 sodium carbonate; NaOH for sodium hydroxide; Na 2 S0 4 for sodium sulfate; NaHS0 3 for sodium bisulfite or sodium hydrogen sulfite; Na 2 S 2 0 3 for sodium thiosulfate; NH 2 NH 2 for hydrazine; NH 4 HC0 3 for ammonium bicarbonate; NH 4 C1 for ammonium chloride; NMMO for N-methylmorpholine N-oxide; NaI0 4 for sodium periodate; Ni for nickel; OH for hydroxyl; Os0 4 for osmium tetroxide; Pd for palladium; Ph for phenyl; PMB for p-methoxybenzyl; POPd for dihydrogen dichlorobis(di
  • PPh 3 for triphenyl-phosphine
  • Troc for 2,2,2-trichloroethyl carbonyl
  • Ts for tosyl or -S0 2 - C 6 H 4 CH 3
  • Ts 2 0 for tolylsulfonic anhydride or tosyl-anhydride
  • TsOH for p-tolylsulfonic acid
  • TMS for trimethylsilyl
  • TMSC1 trimethylsilyl chloride.
  • the compounds of the present invention may be prepared via several different synthetic routes from a variety of 5/6-membered fused heteroaryl, 5-membered heteroaryl, and related intermediates.
  • a retro-synthesis of the compounds include direct formation of a suitably linked core structure (5/6-membered fused heteroaryl or 5-membered heteroaryl) followed by attachment of a suitable capping group (such as -C(0)R 6 ), plus some functional group manipulations in between and/or after.
  • a suitable capping group such as -C(0)R 6
  • Various 5/6-membered fused heteroaryl or 5-membered heteroaryl intermediates are known to those skilled in the art, for example see the encyclopedic volumns edited by A. R. Katrizky, et al,
  • imidazopyridine or benzimidazole 1-2 which may be obtained by condensation of an amino acid or its derivative 1-1.1 or 1-1.2 and a 2,3-diaminopyridine or 1,2- diaminobenzene 1-1 under the conditions to those skilled in the art.
  • the imidazole ring closure may be realized either in one pot by heat, optionally in the presence of an acid and/or with a dehydration reagent such as polyphosphoric acid; or in two steps: 1) amide formation between diamine 1-1 and amino acid 1-1.1 or 1-1.2 in the presence of a condensation reagent such as EDC C1 , DCC or the like; or through mixed anhydride approach by reacting acid 1-1.1 or 1-1.2 with a chloroformate such as methyl
  • chloroformate isobutyl chloroformate, or the like, in the presence of a base such as TEA, DIPEA, DMAP, N-methylmorpholine, or the like, followed by treating the mixed anhydride with diamine 1-1; and 2) the heterocyclic ring closure in the presence of an acid such as acetic acid, sulfuric acid or the like or a dehydration reagent such as HATU or the like, optionally with heat.
  • a base such as TEA, DIPEA, DMAP, N-methylmorpholine, or the like
  • the NH group in the newly formed imidazopyridine or benzimidazole ring of 1-2 may be protected with an amino protecting group, such as SEM (i.e. SEM-C1, NaH), Boc, Cbz, Teoc, Troc, or the like.
  • SEM i.e. SEM-C1, NaH
  • Boc Boc
  • Cbz Boc
  • Teoc Teoc
  • Troc Troc
  • the protected imidazopyridine or benzimidazole 1-2 may be subjected to lithium-halogen exchange with various (n-, s-, or t-) butyl lithium and the resulting lithiate can be trapped with a nucleophile, i.e. a halide such as various allyl halide to give the allylated 1-6 as a key intermediate.
  • a nucleophile i.e. a halide such as various allyl halide
  • 1-6 may be obtained from the Stille reaction conditions to those skilled in the art (see reviews: A. Anastasia, et al, Handbook of Organopalladium Chemistry for Organic Synthesis, 2002, 1, 311; F. Bellina, et al, Synthesis 2004, 2419; M. G. Organ, et al, Synthesis 2008, 2776; A. T. Lindhardt, et al, Chem. - A European J., 2008, 14, 8756; E. A. B. Kantchev, et al, Angew. Chem. Int. Ed., 2007, 46, 2768; V.
  • allylstanne such as allyltributylstanne
  • allyltributylstanne an allylstanne
  • a key vinyl intermediates 1-3 may be prepared by Stille reaction from bromide 1-2 with tributylvinylstanne.
  • Sonogashira coupling between bromide 1-2 and propargyl alcohol or trimethylsilyl-acetylene can generate propargyl alcohol 1-4 or alkyne 1-5 after removal of TMS. Further bromination of intermediate 1-4 may form the propargyl bromide 1-9.
  • the bromide 1-2 may be converted to methyl ketone 1-7 by coupling with tributyl(l-ethoxyvinyl)tin under Stille coupling conditions followed by acidic hydrolysis.
  • bromination procedure which can be further functionalized to amine 1-20 through azide substitution followed by reduction.
  • Aldehyde 1-8 can then either be reduced to alcohol 1- 11, or be converted to a, ⁇ -unsatuated acid 1-10 through Horner- Wadsworth-Emmons aldehyde homologation reaction followed by saponification.
  • Alcohol 1-11 may be similarly converted to the correponding amine intermediate 1-14 and bromide intermediate 1-13 as described previously.
  • Bromide 1-13 can be homologated to alkyne intermediate 1- 19 with a metal acetylide.
  • bromide 1-13 may be also tranformed to thiol 1-16 through nucleophilic substitution, which can be further oxidized to sulfonic acid 1-17.
  • Sulfonamide 1-18 may then be derived from 1-17 through the sulfonyl chloride activation process.
  • the NH group of all the imidazopyridine or benzimidazole related intermediates listed above may be protected with an amino protecting group, such as SEM (i.e. SEM-C1, NaH), Boc, Cbz, Teoc, Troc, or the like.
  • SEM i.e. SEM-C1, NaH
  • Boc Boc
  • Cbz Boc
  • Teoc Teoc
  • Troc Troc
  • bromo- imidazole 2-4 can be synthesized in a three-step sequence: 1) condensation between amino acid derived aldehyde 2-1.1 or 2-1.2 and glyoxal 2-1.3 in the presence of methanolic ammonia to generate imidazole 2-2; 2) bromination of 2-2 with excess amount of bromination reagent such as 2,4,4,6-tetrabromo-2,5-cyclohexadienone, NBS, etc. to afford dibromide 2-3; and 3) selective reduction of the dibromide 2-3 by heating in aq. Na 2 S03 or aq. NaHS0 3 . 2-4 then may be served as a universal intermediate further elaborable to many other imidazole derivatives using the chemistry discussed in Scheme 1, some of which are listed in the table 54 below.
  • the NH group of imidazole related intermediates listed above may be protected with an amino protecting group (shown in Scheme 2 as PG, theoretically the reaction will generate two regio-isomers, but only one is drawn for clarity), such as SEM (i.e. SEM-C1, NaH), Boc, Cbz, Teoc, Troc, or the like.
  • an amino protecting group shown in Scheme 2 as PG, theoretically the reaction will generate two regio-isomers, but only one is drawn for clarity
  • SEM i.e. SEM-C1, NaH
  • Boc Boc
  • Cbz Boc
  • Teoc Teoc
  • Troc Troc
  • the protected imidazole 2-5 may be deprotonated with a strong base such as LDA, BuLi, etc to generate a carbon anion, which may either undergo a nucleophilic substitution with an activated halide such as 2- 5.2 to afford aryl or heteroaryl substituted imidazole 2-6 or couple with an aryl or heteroaryl halide 2-5.1 in the presence appropriate transition metal salt to generate bicyclic heteroaryl 2-7.
  • the protected bromo imidazole 2-8 may be subjected to lithium- halogen exchange with various (n-, s-, or t-) butyl lithium, the resulting lithiate may undergo similar reactions to afford 2-6 and 2-7.
  • aryl or heteroaryl bromide 2-5.1 may be converted to methyl ketone 2-9 under Stille coupling conditions with tributyl(l-ethoxyvinyl)tin 2-9.1.
  • 2-9 may be brominated under conditions to those skilled in the art to afford bromide 2-10, which may be either converted to the corresponding amine 2-11, or coupled with protected amino acid 1-1.1 or 1-1.2 in the presence of a base such as Et 3 N and DIPEA to afford keto-ester 2-12.
  • amine 2-11 may be converted to the corresponding keto-amide 2-13 via condensation with appropriate amino acid under standard amide formation conditions.
  • 2- 12 and 2-13 may be tranformed to key intermediate 2-14 via heating with (NH 4 )OAc under thermal or microwave conditions.
  • requisite buiding blocks for the syntheses of the molecule of the present invention are various amide, carbamate or urea-related intermediates, which can be prepared from a suitable, commercially available amine through standard amide, carbamate or urea formation by direct condensation of an carboxylic acid with a dehydration and/or condensation reagent, such as HATU, DCC, BtOH, EDC or the like; or a chloroformate; or an isocyanate; in the presence of a suitable base such as pyridine, TEA, DIPEA, DMAP, NaHC0 3 , K 2 C0 3 or the like.
  • a suitable base such as pyridine, TEA, DIPEA, DMAP, NaHC0 3 , K 2 C0 3 or the like.
  • the Boc-protected ⁇ -amino acid 3- la can be condensed with 4-bromoaniline 3-2a in the presence of HATU and DIPEA in CH 2 C1 2 to afford amide 3-3a.
  • heteroaryl or aryl amine related intermediates which can be prepared from a suitable, commercially available amine through standard nucleophilic substitution by direct condensation of a heteroaryl halide such as 5-bromo-2-chloropyrimidine, 2,6-dibromoquinazoline, 6-bromo- 2-chlorobenzothiazole, or the like; with an appropriate amine such as 2-aminomethyl-N- Boc-pyrrolidine, l-Boc-3-aminopyrrolidine, trans-N-Boc1,2-cyclopentanediamine or the like; in the presence of a suitable base such as pyridine, TEA, DIPEA, DMAP, NaHC0 3 , K 2 C0 3 or the like; under thermal or microwave conditions.
  • a heteroaryl halide such as 5-bromo-2-chloropyrimidine, 2,6-dibromoquinazoline, 6-bromo- 2-chlorobenzothiazole, or the like
  • heteroaryl or aryl amino related intermediates can be prepared through transition metal catalyzed amination process between an suitable amine as mentioned above and an aryl or heteroaryl iodide or bromide such as 5-bromo-2-iodopyridine, 1 ,4-diiodobenzene, or the like; in the presence of a suitable base such as Cs 2 C0 3 , K 3 P0 4 , K 2 C0 3 or the like; and an appropriate transition metal catalyst such as Cul or various Pd related catalysts; and an related ligand according to the used transition metal catalyst; under inert atmosphere.
  • a suitable base such as Cs 2 C0 3 , K 3 P0 4 , K 2 C0 3 or the like
  • an appropriate transition metal catalyst such as Cul or various Pd related catalysts
  • an related ligand according to the used transition metal catalyst
  • the Boc-protected ⁇ -diamine 3-lb can be condensed with 5-bromo-2-chloropyrimidine 3-2b in the presence of DIPEA in 1,4-dioxane under thermal or microwave conditions to afford pyrimidynyl amine 3-3b.
  • 3-lb can be reacted with 5-bromo-2-iodopyridine 3-4b in the presence of Cs 2 C0 3 , catalytic amount of Cul, and 2-isobutyrylcyclohexanone as ligand, in DMF under N 2 atmosphere to yield the desired pyridinyl amine 3-5b as described by S. L. Buchwald (J. Am. Chem. Soc. 2006, 128, 8742).
  • the compounds of the present invention may be prepared through various coupling strategy or a combination of strategies to connect two fragments, optionally with a suitable cyclic or acyclic linker or formation of a cyclic or acyclic linker.
  • the said strategy includes, but not limited to, Stille coupling, Suzuki coupling, Sonogashira coupling, Heck coupling, Buchwald amidation, Buchwald amination, alkylation, pericyclic reaction with different variations, or the like.
  • bromides 4-1 and 4-2 can be prepared using similar procedures described in Schemes 1-3 and 3a.
  • Bromide 4-2 can be converted to the corresponding metallated aryl 4-3 (boronate or stanne) under Suzuki or Stille conditions with bis(pinacolato)diboron, hexamethylditin or hexabutylditin in the presence of Pd- catalyst.
  • the latter may be further coupled with imidazopyridine bromide 4-1 under similar conditions to generate a core structure 4-4.
  • Compound 4-4 then may be served as a common intermediate for further derivatizations to 4-5 in two steps: 1) mono-deprotection of the linear or cyclic amine moiety may be accomplished, for example, treatment to hydrogenolytic conditions under Pd catalyst to remove the Cbz protection group; and 2) the released amine functionality may be acylated with an carboxylic acid under standard acylation conditions, for example a coupling reagent such as HATU in combination with an organic base such as DIPEA can be used in this regard; alternatively, the released amine may be reacted with an isocyanate, carbamoyl chloride or chloroformate to provide an urea or carbamate.
  • Various carboxylic acids including amino acids in racemic or optical form are commercially available, and/or can be synthesized in racemic or optical form, see references cited in reviews by D.
  • the compounds of the present invention may also be derived from bromoimidazopyridine or bromobenzimidazole 4- la and amide 4-2a using the procedures described previously.
  • the intermediates 4- la and 4-2a have the desired acyl groups already installed using similar sequences shown in Scheme 4.
  • the compounds of the present invention containing five-membered heteroaryl other than imidazole may be prepared using similar procedures described above in Schemes 1-4 and 4a.
  • some intermediates containing a desired, suitably substituted five-membered heteroaryl have been published in US 2008/0311075A1 by C. Bachand, et al from Bristol-Myers Squibb, Co., which is incorporated by reference.
  • the synthesis of the compounds of the present invention involves 5/6-membered fused heteroaryl intermediates other than imidazopyridines or benzimidazoles, various 5/6-membered fused heteroaryl are known in the literature.
  • the synthesis of other 5/6-membered fused heteroaryl intermediates depends on the chemical features of each structure. For example, a typical synthesis of indole intermediate is illustrated in Scheme 5.
  • the commercially available bromoiodoaniline 5-1 may be coupled to the commercially available acetylene 5-1.1 under the Sonogashira conditions to give phenylacetylene 5-2.
  • the latter may be cyclized to indole 5-3 under heat or microwave condition in the presence of a copper catalyst.
  • the invention encompasses a process of making a compound of the invention comprising:
  • Ring A 1 is absent, optionally substituted aryl or optionally substituted heteroaryl
  • Ring B 1 is optionally substituted aryl or optionally substituted heteroaryl
  • L 1 is absent, optionally substituted C 2 -C 4 alkenyl or C 2 -C 4 alkynyl;
  • Z a and Z b are each independently an amino protecting group or -C(0)-R 6 ; wherein R 6 is as defined above;
  • Z c is hydrogen, an amino protecting group or -C(0)-R 6 ;
  • Z d is an amino protecting group -C(0)-R 6 ;
  • the invention is a process of making a compound according to Formula (l-I) comprising:
  • Ring A 1 is absent, optionally substituted aryl or optionally substituted heteroaryl; Ring B 1 is optionally substituted aryl or optionally substituted heteroaryl;
  • L 1 is absent, optionally substituted C2-C 4 alkenyl or C2-C 4 alkynyl;
  • Z a and Z b are each independently an amino protecting group or -C(0)-R 6 ; wherein R 6 is as defined in in Formula (1-1);
  • Z c is hydrogen, an amino protecting group or -C(0)-R 6 ;
  • Z d is an amino protecting group -C(0)-R 6 ;
  • the invention is a process of making a compound accordingormula (2-1) comprising:
  • Ring A 1 and Ring B 1 are each independently optionally substituted aryl or optionally substituted heteroaryl;
  • L 1 is absent, optionally substituted C2-C 4 alkenyl or C2-C 4 alkynyl; and Z a and Z b are each independently an amino protecting group or -C(0)-R 6 ; wherein R 6 is as defined in Formula (2-1);
  • Z c is hydrogen, an amino protecting group or -C(0)-R 6 ;
  • Z d is an amino protecting group -C(0)-R 6 ;
  • the invention is a process of making a compound of Formula (3-1) comprising:
  • R 1 , R la , R lb , R 7a , R 7b , R 9 , and R 9a are as defined in Formula (3-1);
  • Ring A 1 and Ring B 1 are each independently absent or an optionally substituted aryl or optionally substituted heteroaryl;
  • L 1 is absent, optionally substituted C 2 -C 4 alkenyl or C 2 -C 4 alkynyl;
  • Z a and Z b are each independently an amino protecting group or -C(0)-R 6 ; wherein R 6 is as defined in Formula (3-1); ii) when Z a or Z b is an amino protecting group, fully or selectively deprotecting a compound of Formula (3 -II) to give the corresponding amine of Formula (3 -III):
  • Z c is hydrogen, an amino protecting group or -C(0)-R 6 ;
  • Z d is an amino protecting group -C(0)-R 6 ;
  • the invention is a process of making a compound of Formula (4-omprising:
  • R 1 , R la , R lb , R lc , R 7a , R 7b , R 9 , and R 9a are as defined in Formula (4-1);
  • Ring A 1 is absent, or optionally substituted aryl or optionally substituted heteroaryl
  • Ring B 1 is optionally substituted aryl or optionally substituted heteroaryl
  • L 1 is absent, or optionally substituted C2-C 4 alkenyl or C2-C 4 alkynyl
  • Z a or Z b are each independently an amino protecting group or -C(0)-R 6 ; wherein R 6 is as defined in Formula (4-1); ii) when Z a or Z b is an amino protecting group, fully or selectively deprotecting a compound of Formula (4-II) to give the corresponding amine of Formula (4-III):
  • Z c is hydrogen, an amino protecting group or -C(0)-R 6 ;
  • Z d is an amino protecting group or -C(0)-R 6 ;
  • the invention is a process of making a compound of Formula (5-1) comprising:
  • Ring B, G, U, R 1 , R la , R lb , R lc , R 7a , R 7b , R 9 , and R 9a are as defined in Formula (5-1);
  • Ring A 1 is absent, optionally substituted aryl or optionally substituted heteroaryl
  • L 1 is absent, optionally substituted C 2 -C 4 alkenyl or C 2 -C 4 alkynyl;
  • Z a and Z b are each independently an amino protecting group or -C(0)-R 6 ; wherein R 6 is as defined in Formula (5-1);
  • Z c is hydrogen, an amino protecting group or -C(0)-R 6 ;
  • Z d is an amino protecting group or -C(0)-R 6 ;
  • Step l-257a A solution of the compound from example 1-353 (32.5 ⁇ at most) in
  • Step l-257b A mixture of the crude compound from step l-257a (32.5 ⁇ at most) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
  • Step l-348a A mixture of 4-bromoaniline (0.500 g, 2.91 mmol) and trans (+/-) 2-(tert- butoxycarbonylamino)-cyclopentanecarboxylic acid (0.733 g, 3.20 mmol) in DMF (12 mL) were added EDC HCl (0.724 g, 3.78 mmol), HOBt (0.646 g, 3.78 mmol) and DIPEA (0.54 mL, 4.36 mmol). The mixture was stirred at room temperature until the
  • Step l-348b A mixture of 2,4'-dibromoacetophenone (5.00 g, 18.0 mmol) and N-Boc-L- proline (3.87 g, 18.0 mmol) in CH 3 CN (60 mL) was added TEA (5.40 mL, 37.8 mmol) slowly. The mixture was stirred at room temperature until the disappearence of the starting material. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-348c A solution of the compound from step l-348b (6.73 g, 16.3 mmol) in toluene (100 mL) was added ammonium acetate (25.1 g, 0.327 mol) and the resultant mixture was heated up to 100 °C for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - aq. NaHC0 3 ). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a yellow foam (6.10 g, 95%).
  • Step l-348d A mixture of the compound from step 348c (1.00 g, 2.55 mmol), bis-
  • Step l-348e A mixture of the compound from step l-348a (0.200 g, 0.522 mmol), the compound from step 348d (0.229 g, 0.522 mmol) and NaHC0 3 (0.175 g, 2.09 mmol) in DME (12 mL) and H 2 0 (4 mL) was added Pd(PPh 3 ) 4 (30.2 mg, 26.1 ⁇ ). The resultant mixture were degassed and heated to 85°C under N 2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated. The residue was purified by
  • Step l-349b A mixture of the crude compound from step l-348a (97.5 ⁇ at most) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
  • Step l-350a A mixture of 2-amino-6-bromonaphthalene (0.500 g, 2.25 mmol) and trans (+/-) 2-(tert-butoxycarbonylamino)-cyclopentanecarboxylic acid (0.567 g, 2.48 mmol) in DMF (10 mL) were added EDC C1 (0.561 g, 2.93 mmol), HOBt (0.500 g, 2.93 mmol) and DIPEA (0.42 mL, 3.38 mmol). The mixture was stirred at room temperature until the disappearence of the starting material.
  • Step l-350b A mixture of the compound from step l-350a (0.200 g, 0.462 mmol), the compound from step 348d (0.184 g, 0.420 mmol) and NaHC0 3 (0.141 g, 1.68 mmol) in DME (12 mL) and H 2 0 (4 mL) was added Pd(PPh 3 ) 4 (24.2 mg, 20.9 ⁇ ). The resultant mixture were degassed and heated to 85°C under N 2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-351a A solution of the compound from example 1-350 (50.0 mg, 75.1 ⁇ ) in CH 2 CI 2 (5 mL) was treated with C1 in 1,4-dioxane (4 M, 1 mL) for 30 min. The volatiles 5 were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step.
  • ESIMS m/z 466.24 [M+H] + .
  • Step l-351b A mixture of the crude compound from step 1-35 la (75.1 ⁇ at most) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
  • Step l-352a A mixture of (i?)-(+)-tert-butyl pyrrolidin-3-ylcarbamate (0.100 g, 0.537 mmol) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO 2008/021927, 0.103 g, 0.590 mmol) in DMF (3 mL) was treated with HATU (0.204 g, 0.537 mmol) in the presence of DIPEA (0.13 mL, 1.07 mmol) for 2 hours at rt and the 20 volatiles were evaporated off to provide a brown syrup.
  • Step l-352a A solution of the compound from step l-352a (0.537 mmol at most) in CH 2 CI 2 (6 mL) was treated with C1 in 1,4-dioxane (4 M, 6 mL) for 30 min. The volatiles 25 were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step.
  • ESIMS m/z 244.24 [M+H] + .
  • Step l-352d A solution of the compound from step l-348c (1.500 g, 3.824 mmol) in 1,4- dioxane (12 mL) was treated with C1 in 1,4-dioxane (4 M, 24 mL) at room temperature for 1.5 hours. The volatiles were evaporated off to give the crude desired compound as a yellow solid, which was used directly in the next step.
  • ESIMS m/z 292.06, 294.06
  • Step l-352e A mixture of the crude compound from step l-352d (3.824 mmol at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 0.670 g, 3.824 mmol) in DMF (12 mL) was treated with HATU (1.381 g, 3.633 mmol) in the presence of DIPEA (6.66 mL, 38.24 mmol) for 1 hour at room temperature and the volatiles were evaporated off. The residue was purified by
  • Step l-352f A mixture of the compound from step l-352e (1.50 g, 3.34 mmol), bis-
  • Step l-352g A mixture of the compound from step l-350c (39.8 mg, 84.1 ⁇ ), the compound from step l-352f (54.2 mg, 0.109 mmol) and NaHC0 3 (28.3 g, 0.336 mmol) in DME (6 mL) and H 2 0 (2 mL) was added Pd(PPh 3 ) 4 (4.9 mg, 4.2 ⁇ ). The resultant mixture were degassed and heated to 85 °C under N 2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-353b A mixture of the compound from step l-353a (49.0 mg, 0.114 mmol), the compound from step l-348d (60.0 mg, 0.137 mmol) and NaHC0 3 (38.3 mg, 0.456 mmol) in DME (6 mL) and H 2 0 (2 mL) was added Pd(PPh 3 ) 4 (6.5 mg, 5.6 ⁇ ). The resultant mixture were degassed and heated to 85 °C under N 2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-354b A mixture of the compound from step l-354a (50.3 mg, 0.136 mmol), the compound from step l-348d (65.8 mg, 0.150 mmol) and NaHC0 3 (45.8 mg, 0.545 mmol) in DME (6 mL) and H 2 0 (2 mL) was added Pd(PPh 3 ) 4 (7.9 mg, 6.8 ⁇ ). The resultant mixture were degassed and heated to 85°C under N 2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-355a A solution of the compound from example 1-354 (20.0 mg, 33.2 ⁇ ) in CH 2 C1 2 (4 mL) was treated with C1 in 1,4-dioxane (4 M, 3 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step.
  • ESIMS m/z 402.38 [M+H] + .
  • Step l-355b A mixture of the crude compound from step l-355a (max. 33.2 ⁇ ) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
  • Step l-356a A mixture of trans-N-Boc-1,2-cyclopentanediamine (60.0 mg, 0.300 mmol) and 3-iodobenzoic acid (74.3 mg, 0.300 mmol) in CH 2 C1 2 (6 mL) was treated with HATU (0.114 g, 0.300 mmol) in the presence of DIPEA (75 ⁇ ⁇ , 0.600 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a white solid (0.120 g, 93%).
  • Step l-356c The compound from step l-356b (2.89 g, 11.6 mmol) in acetic acid (60 mL) was treated with bromine (0.59 mL, 11.6 mmol) dropwise for 1 hour. The volatiles were evaporated and the residue was partitioned (EtOAc - saturated aqueous NaHC0 3 ). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated to give the crude desired compound as a light yellow solid (3.898 g).
  • Step l-356e A mixture of the compound from step l-356d (0.200 g, 0.452 mmol), bis(pinaco-lato)diboron (0.144 g, 0.565 mmol), PdCl 2 (dppf) 2 (36.9 mg, 0.0452 mmol) and potassium acetate (88.7 mg, 0.904 mmol) in DMSO (5 mL) was degassed and heated at 80°C under N 2 for 17 hours. The reaction mixture was allowed to cool down and partitioned (EtOAc - water). The organic layer was washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-356f A mixture of the compound from step l-356a (29.4 mg, 68.3 ⁇ ), the compound from step l-356e (40.0 mg, 81.9 ⁇ ) and NaHC0 3 (22.9 mg, 0.273 mmol) in DME (6 mL) and H 2 0 (2 mL) was added Pd(PPh 3 ) 4 (3.9 mg, 3.4 ⁇ ). The resultant mixture were degassed and heated to 85 °C under N 2 for 16 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-357a A solution of the compound from example 1-356 (20.0 mg, 30.0 ⁇ ) in CH 2 CI 2 (4 mL) was treated with C1 in 1,4-dioxane (4 M, 3 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step.
  • ESIMS m/z 466.43 [M+H] + .
  • Step l-357b A mixture of the crude compound from step l-357a (max. 30.0 ⁇ ) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
  • Step l-358a A mixture of N-Boc-L-proline (5.754 g, 26.7 mmol) and TEA (3.73 mL, 26.7 mmol) in THF (60 mL) at -20 °C was treated with ethyl chloroformate (2.55 mL, 26.7 mmol) for 30 minutes before a slow addition of 4-bromo-1,2-diaminobenzene (5.00 g, 26.7 mmol) in THF (20 mL). It was then kept at -20°C for 1 hour and then slowly warmed up to rt and stirred at rt overnight. The volatiles were evaporated and the residue was partitioned (EtOAc - water).
  • Step l-358b A solution of the crude compound from step l-358a (10.7 g, 26.7 mmol at most) in glacial acetic acid (100 mL) was heated at 50°C for 2 hours. The volatiles were evaporated off and the residue was partitioned (EtOAc - aqueous NaHC0 3 ). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-358c A mixture of the compound from step l-358b (2.010 g, 5.488 mmol), trimethylsilyl-acetylene (2.33 ml, 16.46 mmol), Cul (0.110 g, 0.576 mmol) and
  • Step l-358e A mixture of the compound from step l-354a (52.6 mg, 0.143 mmol), the compound from step l-358d (48.8 mg, 0.157 mmol) in Et 3 N (6 mL) was added Cul (0.8 mg, 4.2 ⁇ ) and Pd(PPh 3 ) 4 (16.4 mg, 14.2 ⁇ ). The resultant mixture were degassed and heated to 85°C under N 2 for 20 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-359a A solution of the compound from example 1-358 (27.4 mg, 45.7 ⁇ ) in CH 2 C1 2 (4 mL) was treated with C1 in 1,4-dioxane (4 M, 3 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step.
  • ESIMS m/z 400.35 [M+H] + .
  • Step l-359b A mixture of the crude compound from step l-359a (max. 45.7 ⁇ ) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
  • Step l-360b A solution of the compound from step l-356d (0.120 g, 0.271 mmol) in 1,4- dioxane (3 mL) was treated with C1 in 1,4-dioxane (4 M, 6 mL) at rt for 1.5 hours. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was used directly in the next step.
  • Step l-360c A mixture of the crude compound from step l-360b (0.271 mmol at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (47.5 mg, 0.271 mmol) in DMF (2 mL) was treated with HATU (98.0 mg, 0.258 mmol) in the presence of DIPEA (0.47 mL, 2.713 mmol) for 1.5 hours at rt and the volatiles were evaporated off . The residue was patitioned (EtOAc/CH 2 Cl 2 - H 2 0). The organic layer was washed with saturated NaHC0 3 , brine, dried (Na 2 S0 4 ), filtered and concentrated.
  • Step l-360d A mixture of the compound from step l-360c (0.130 g, 0.260 mmol), bis(pinacolato)diboron (79.3 mg, 0.312 mmol), PdCl 2 (dppf) 2 (21.3 mg, 26.0 ⁇ ) and potassium acetate (51.0 mg, 0.521 mmol) in DMSO (4 mL) was degassed and heated at 80 °C under N 2 overnight. The mixture was allowed to cool down and partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-360e A mixture of the compound from step l-360a (18.4 mg, 43.9 ⁇ ), the compound from step 360d (20.0 mg, 36.6 ⁇ ) and NaHC0 3 (12.3 mg, 0.146 mmol) in DME (6 mL) and H 2 0 (2 mL) was added Pd(PPh 3 ) 4 (2.1 mg, 1.8 ⁇ ). The resultant mixture were degassed and heated to 85°C under N 2 for 16 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-362a A mixture of N-Boc-L-proline (5.754 g, 26.7 mmol) and TEA (3.73 mL, 26.7 mmol) in THF (60 mL) at -20°C was treated with ethyl chloroformate (2.55 mL, 26.7 mmol) for 30 minutes before a slow addition of 4-bromo-1,2-diaminobenzene (5.00 g, 26.7 mmol) in THF (20 mL). It was then kept at -20 °C for 1 hour and then slowly warmed up to rt and stirred at rt overnight. The volatiles were evaporated and the residue was partitioned (EtOAc - water).
  • Step l-362c A mixture of the compound from step l-362b (1 g, 2.73 mmol), bis- (pinacolato)-diboron (763 mg, 3.0 mmol), potassium acetate (402 mg, 4.0 mmol) in 1,4- dioxane (9.1 mL) was added tetrakis(triphenylphosphine)palladium(0) (158 mg, 0.14 mmol). The resulting solution was degased and then heated at 80°C under N 2 overnight before being evaporated.
  • Step l-362d A mixture of the compound from step l-354a (20.0 mg, 54.2 ⁇ ), the compound from step l-362c (24.6 mg, 59.6 ⁇ ) and NaHC0 3 (18.2 mg, 0.217 mmol) in DME (6 mL) and H 2 0 (2 mL) was added Pd(PPh 3 ) 4 (3.1 mg, 2.7 ⁇ ). The resultant mixture were degassed and heated to 90°C under N 2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-364a A mixture of ( ⁇ ) -l-N-Boc-3 ⁇ 4eto-proline (0.100 g, 0.465 mmol) and 2-amino- 6-bromonaphthalene (0.103 g, 0.465 mmol) in CH 2 CI 2 (6 mL) was treated with HATU (0.177 g, 0.465 mmol) in the presence of DIPEA (0.12 mL, 0.929 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a white solid (0.191 g, 98%).
  • Step l-364b A mixture of the compound from step l-364a (30.0 mg, 71.5 ⁇ ), the compound from step l-348d (34.5 mg, 78.6 ⁇ ) and NaHC0 3 (24.0 mg, 0.286 mmol) in DME (6 mL) and H 2 0 (2 mL) was added Pd(PPh 3 ) 4 (4.0 mg, 3.5 ⁇ ). The resultant mixture were degassed and heated to 95°C under N 2 for 16 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-367a To a suspension of trans- 1 ,2-cyclopentanediamine dihydrochloride
  • Step l-367b A mixture of the crude compound from step l-367a (0.578 mmol at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 101.2 mg, 0.578 mmol) in DMF (3 mL) was treated with HATU (0.220 g, 0.578 mmol) in the presence of DIPEA (1.00 mL, 5.778 mmol) for 1 hours at room temperature and the volatiles were evaporated off. The residue was taken up in
  • Step l-367c A mixture of compound from step l-367b (17.0 mg, 34.9 ⁇ ), the compound from step 352f (17.2 mg, 34.9 ⁇ ), Pd(PPh 3 ) 4 (4.0 mg, 3.49 ⁇ ) and NaHC0 3 (10.3 mg, 0.122 mmol) in DME (2.1 mL) and H 2 0 (0.7 mL) was degassed and heated at 97 °C under N 2 for 15 hours. The mixture was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-368b A mixture of compound from step l-368a (45.0 mg, 0.105 mmol), the compound from step l-348d (46.0 mg, 0.105 mmol), Pd(PPh 3 ) 4 (12.1 mg, 10.5 ⁇ ) and NaHC0 3 (30.8 mg, 0.366 mmol) in DME (3 mL) and H 2 0 (1 mL) was degassed and heated at 97°C under N 2 for 15 hours. The mixture was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-369a A solution of the compound of example 1-368 (63.2 mg, 0.103 mmol) in 1,4-dioxane (1.25 mL) was treated with C1 in 1,4-dioxane (4 M, 5 mL) at 30°C for 30 minutes. The volatiles were evaporated off to give the crude desired compound as a yellow solid, which was used directly in the next step.
  • Step l-368b A mixture of the crude compound from step l-369a (0.103 mmol at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 36.0 mg, 0.205 mmol) in DMF (3 mL) was treated with HATU (74.1 mg, 0.195 mmol) in the presence of DIPEA (0.36 mL, 2.053 mmol) for 30 minutes at rt and the volatiles were evaporated off.
  • Step l-370b A mixture of the compound from step l-362c (0.250 g, 0.605 mmol), 1- bromo-4-iodobenzene (0.257 g, 0.908 mmol), NaHC0 3 (0.203 g, 2.42 mmol) and
  • Step l-370c To a mixture of the compound from step l-370b (50.0 mg, 0.113 mmol), bis-(pinacolato)-diboron (35.9 mg, 0.141 mmol), potassium acetate (22.2 mg, 0.226 mmol) in DMSO (2 mL) was added PdCl 2 (dppf) 2 (9.2 mg, 11.3 ⁇ ). The resulting solution was degased and then heated at 80 °C under N 2 overnight before being partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-370d A mixture of compound from step l-370a (32.0 mg, 74.4 ⁇ ), the compound from step l-370c (36.4 mg, 74.4 ⁇ ), Pd(PPh 3 ) 4 (8.6 mg, 7.4 ⁇ ) and NaHC0 3 (21.9 mg, 0.260 mmol) in DME (2.1 mL) and H 2 0 (0.7 mL) was degassed and heated at 97 °C under N 2 for 15 hours. The mixture was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-371a A solution of the compound of example 1-370 (31.7 mg, 47.6 ⁇ ) in 1,4- dioxane (1 mL) was treated with C1 in 1,4-dioxane (4 M, 4 mL) at 30 °C for 30 minutes. The volatiles were evaporated off to give the crude desired compound as a yellow solid, which was used directly in the next step.
  • Step l-371b A mixture of the crude compound from step 1-37 la (47.6 ⁇ at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 16.7 mg, 95.2 ⁇ ) in DMF (3 mL) was treated with HATU (34.4 mg, 90.5 ⁇ ) in the presence of DIPEA (0.17 mL, 0.952 mmol) for 30 minutes at rt and the volatiles were evaporated off.
  • Step l-372a A mixture of (5)-3-(Boc-amino)pyrrolidine (0.150 g, 0.805 mmol) and (S)- 2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO
  • Step l-372b A solution of the compound from step l-372a (0.680 g, 0.805 mmol at most) in 1,4-dioxane (1.5 mL) was treated with C1 in 1,4-dioxane (4 M, 6 mL) at rt for 1 hours. The volatiles were evaporated off to give the crude desired compound as a yellow oil, which was used directly in the next step.
  • ESIMS m/z 244.17 [M+H] + .
  • Step l-372d A mixture of compound from step l-372c (30.0 mg, 63.4 ⁇ ), the compound from step l-352f (40.9 mg, 82.4 ⁇ ), Pd(PPh 3 ) 4 (7.3 mg, 6.34 ⁇ ) and NaHC0 3 (21.3 mg, 0.254 mmol) in DME (3 mL) and H 2 0 (1 mL) was degassed and heated at 98 °C under N 2 for 2.5 hours. The mixture was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et 3 N and 20% MeOH in EtOAc) to give the title compound as a light brown solid (42.7 mg, 94%).
  • Step l-373a A mixture of (i?)-N-Boc-pyrrolidine-3-carboxylic acid (1.000 g, 4.646 mmol) and 4-bromoaniline (0.799 g, 4.646 mmol) in CH 2 CI 2 (20 mL) was treated with HATU (1.766 g, 4.646 mmol) in the presence of DIPEA (1.62 mL, 9.292 mmol) for 1 hours at rt and the volatiles were evaporated off. The residue was purified by
  • Step l-373b A mixture of compound from step l-373a (0.100 g, 0.271 mmol), the compound from step l-348d (0.131 g, 0.298 mmol), Pd(PPh 3 ) 4 (31.2 mg, 27.1 ⁇ ) and NaHC0 3 (91.0 mg, 1.083 mmol) in DME (3 mL) and H 2 0 (1 mL) was degassed and heated at 97°C under N 2 for 15 hours. The mixture was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-374a A solution of the compound of example 1-373 (0.111 g, 0.184 mmol) in 1,4- dioxane (2 mL) was treated with C1 in 1,4-dioxane (4 M, 6 mL) at room temperature for 1 hour. The volatiles were evaporated off to give the crude desired compound as a yellow solid, which was used directly in the next step.
  • Step l-374b A mixture of the crude compound from step l-374a (0.184 mmol at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 64.6 mg, 0.369 mmol) in DMF (4 mL) was treated with HATU (0.133 g, 0.351 mmol) in the presence of DIPEA (0.64 mL, 3.689 mmol) for 1 hour at room temperature and the volatiles were evaporated off. The residue was purified by
  • Step l-375a A mixture of trans-aminocyclopentanol hydrochloride (racemic, 0.100 g, 0.727 mmol) and 4-iodobenzoic acid (0.180 g, 0.727 mmol) in CH 3 CN (10 mL) was treated with HATU (0.276 g, 0.727 mmol) in the presence of DIPEA (1.27 mL, 7.266 mmol) for 30 minutes at rt and the volatiles were evaporated off. The residue was purified by chromatography (silica, hexanes-ethyl acetate with 1% EtOH) to give the desired compound as a white solid (0.226 g, 94%).
  • ESIMS m/z 332.04 [M+H] + .
  • Step l-375b A mixture of compound from step l-375a (45.0 mg, 0.136 mmol), the compound from step l-348d (46.0 mg, 0.105 mmol), Pd(PPh 3 ) 4 (15.7 mg, 13.6 ⁇ ) and NaHC0 3 (45.7 mg, 0.544 mmol) in DME (3 mL) and H 2 0 (1 mL) was degassed and heated at 97°C under N 2 for 15 hours. The mixture was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step l-376a A solution of the compound of example 1-375 (54.2 mg, 0.105 mmol) in 1,4-dioxane (1.25 mL) was treated with C1 in 1,4-dioxane (4 M, 5 mL) at room temperature for 1 hour. The volatiles were evaporated off to give the crude desired compound as a yellow solid, which was used directly in the next step.
  • Step l-376b A mixture of the crude compound from step l-376a (0.103 mmol at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 36.8 mg, 0.210 mmol) in DMF (4 mL) was treated with HATU (37.9 mg, 0.100 mmol) in the presence of DIPEA (0.18 mL, 1.049 mmol) for 15 minutes at rt and the volatiles were evaporated off.
  • HATU 37.9 mg, 0.100 mmol
  • Step l-378a A mixture of (5)-N-Boc-pyrrolidine-3-carboxylic acid (80.0 mg, 0.372 mmol) and 2-amino-5-bromopyridine (64.3 mg, 0.372 mmol) in CH 2 CI 2 (3 mL) was treated with HATU (0.141 g, 0.372 mmol) in the presence of DIPEA (0.13 mL, 0.743 mmol) for 1 hours at rt. More (5)-N-Boc-pyrrolidine-3-carboxylic acid (40.0 mg, 0.186 mmol), DIPEA (0.065 mL, 0.372 mmol) and HATU (70.5 mg, 0.186 mmol) were added.
  • Step l-378b A mixture of compound from step l-378a (48.0 mg, 0.130 mmol), the compound from step l-370c (62.7 mg, 0.143 mmol), Pd(PPh 3 ) 4 (15.0 mg, 13.0 ⁇ ) and NaHC0 3 (38.1 mg, 0.454 mmol) in DME (3 mL) and H 2 0 (1 mL) was degassed and heated at 97°C under N 2 for 15 hours. The volatiles were evaporated off. The residue was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et 3 N in EtOAc) to give the title compound as a yellow solid (61.8 mg, 79%).
  • ESIMS m/z 603.15 [M+H] + .
  • Step l-379a A solution of the compound of example 1-378 (61.8 mg, 0.102 mmol) in 1,4-dioxane (1 mL) was treated with C1 in 1,4-dioxane (4 M, 4mL) at room temperature for 1.5 hours. The volatiles were evaporated off to give the crude desired compound as a yellow solid, which was used directly in the next step.
  • Step l-379b A mixture of the crude compound from step l-379a (0.102 mmol at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 35.9 mg, 0.205 mmol) in DMF (2 mL) was treated with HATU (74.1 mg, 0.195 mmol) in the presence of DIPEA (0.36 mL, 2.051 mmol) for 15 minutes at room temperature and the volatiles were evaporated off.
  • Step l-381a Into a mixture of (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 90 mg, 0.5 mmol) and (R)-tert-butyl 2- (aminomethyl)pyrrolidine-1-carboxylate (100 mg, 0.50 mmol) in acetonitrile (2 mL) was added diisopropylethylamine (0.11 mL, 0.6 mmol) and HATU (200 mg, 0.55 mmol). The resulting mixture was stirred at room temperature for 1 hour before being partitioned between water and EtOAc. The organic phase was separated, dried (Na 2 S0 4 ) and concentrated to afford a brown slurry, which was purified by flash column
  • Step l-381b Into a mixture of compound firm step l-381a (144 mg, 0.4 mmol) was added hydrochloric acid in 1,4-dioxane (4M, 2 mL). The resulting mixture was stirred at room temperature for 1 hour before all volatiles were removed to afford the crude desired compound, which was used directly for the next step without further purification.
  • ESIMS m/z 258.28 [M+H] + .
  • Step l-381c The title compound was prepared from 4-iodobenzoic acid and the compound from step l-348d using procedures similar to that described in example 1-348.
  • ESIMS m/z 730.45 [M+H] + .
  • Examples 2-1 to 2-293 may be prepared using procedures similar to those described in examples 2-294 and 2-295 (described below), and/or as described in the Synthetic Methods.
  • Step 2-294a A mixture of l-Boc-pyrrolidine-3-carboxylic acid (0.100 g, 0.465 mmol) and 4-bromoaniline (79.9 mg, 0.465 mmol) in CH 2 CI 2 (6 mL) was treated with HATU (0.177 g, 0.465 mmol) in the presence of DIPEA (0.12 mL, 0.929 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a colorless oil (0.156 g, 91%).
  • Step 2-294b A mixture of the compound from step 2-294a (0.184 g, 0.499 mmol), bis- (pinacolato) diboron (0.253 g, 0.997 mmol) and potassium acetate (0.122 g, 1.25 mmol) in 1,4-dioxane (8 mL) was added Pd(PPh 3 ) 4 (28.8 mg, 24.9 ⁇ ). The resultant mixture were degassed for three times and heated up to 85°C under N 2 for 13 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - water).
  • Step 2-294c A mixture of the compound from step 2-294a (30.0 mg, 81.3 ⁇ ), the compound from step 2-294b (40.6 mg, 97.5 ⁇ ) and NaHC0 3 (27.3 mg, 0.325 mmol) in DME (6 mL) and H 2 0 (2 mL) was added Pd(PPh 3 ) 4 (4.6 mg, 4.0 ⁇ ). The resultant mixture were degassed and heated to 85°C under N 2 for 16 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step 2-295a A solution of the compound from example 2-294 (23.5 mg, 40.6 ⁇ ) in CH 2 C1 2 (4 mL) was treated with C1 in 1 ,4-dioxane (4 M, 3 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step.
  • ESIMS m/z 379.35 [M+H] + .
  • Step 2-295b A mixture of the crude compound from step 2-295 a (40.6 ⁇ at most) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
  • Step 3-336a A mixture of 2,4'-dibromoacetophenone (1.00 g, 3.60 mmol) and trans (+/-) 2-(fert-butoxycarbonylamino)-cyclopentanecarboxylic acid (0.825 g, 3.60 mmol) in CH 3 CN (12 mL) was added DIPEA (0.94 mL, 7.55 mmol) slowly. The mixture was stirred at room temperature until the disappearence of the starting material. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step 3-336c A mixture of 2,4'-dibromoacetophenone (5.00 g, 18.0 mmol) and N-Boc-L- proline (3.87 g, 18.0 mmol) in CH 3 CN (60 mL) was added TEA (5.40 mL, 37.8 mmol) slowly. The mixture was stirred at room temperature until the disappearence of the starting material. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step 3-336d A solution of the compound from step 3-336c (6.73 g, 16.3 mmol) in toluene (100 mL) was added ammonium acetate (25.1 g, 0.327 mol) and the resultant mixture was heated up to 100°C for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - aq. NaHC0 3 ). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a yellow foam (6.10 g, 95%).
  • Step 3-336e A mixture of the compound from step 3-336d (1.00 g, 2.55 mmol), bis-
  • Step 3-336f A mixture of the compound from step 3_ 336b (0.315 g, 0.777 mmol), the compound from step 3 ⁇ 3366 (0.310 g, 0.706 mmol) and NaHC0 3 (0.237 g, 2.82 mmol) in DME (12 mL) and H 2 0 (4 mL) was added Pd(PPh 3 ) 4 (40.8 mg, 35.3 ⁇ ). The resultant mixture were degassed and heated to 85°C under N 2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step 3-337a A solution of the compound of example 3-336 (80.0 mg, 0.125 mmol) in CH 2 C1 2 (3 mL) was treated with C1 in 1,4-dioxane (4 M, 6 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step.
  • ESIMS m/z 439.23 [M+H] + .
  • Step 3-337b A mixture of the crude compound from step 3 ⁇ 337a (theo 0.125 mmol) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
  • Step 3-338a A mixture of N-Boc-L-proline (5.754 g, 26.7 mmol) and TEA (3.73 mL, 26.7 mmol) in THF (60 mL) at -20°C was treated with ethyl chloroformate (2.55 mL, 26.7 mmol) for 30 minutes before a slow addition of 4-bromo-1,2-diaminobenzene (5.00 g, 26.7 mmol) in THF (20 mL). It was then kept at -20 °C for 1 hour and then slowly warmed up to rt and stirred at rt overnight. The volatiles were evaporated and the residue was partitioned (EtOAc - water).
  • Step 3-338c A mixture of the compound from 3_ 2 338b (0.559 g, 1.425 mmol), trimethylsilyl acetylene (0.60 ml, 4.275 mmol), Cul (28.5 mg, 0.150 mmol) and
  • Step 3-338e A mixture of the compound from step 3 ⁇ 336b (150 mg, 0.369 mmol), the compound from step 3 ⁇ 338(1 (127 mg, 0.406 mmol) in Et 3 N (4 mL) and CH 3 CN (4 mL) were added Cul (2.1 mg, 11.0 ⁇ ) and Pd(PPh 3 ) 4 (21.3 mg, 18.4 ⁇ ). The resultant mixture were degassed and heated to 85°C under N 2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step 3-339a A solution of the compound of example 3 ⁇ 338 (0.137 g, 0.215 mmol) in CH 2 C1 2 (3 mL) was treated with C1 in 1,4-dioxane (4 M, 6 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step.
  • ESIMS m/z 437.20 [M+H] + .
  • Step 3-339b A mixture of the crude compound from step 3_-339a (theo 0.215 mmol) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
  • Step 3-340a A mixture of the compound from step 3_ 338b (1 g, 2.73 mmol), bis- (pinacolato)-diboron (763 mg, 3.0 mmol), potassium acetate (402 mg, 4.0 mmol) in 1,4- dioxane (9.1 mL) was added tetrakis(triphenylphosphine)palladium(0) (158 mg, 0.14 mmol). The resulting solution was degased and then heated at 80°C under N 2 overnight before being evaporated.
  • Step 3-340b A mixture of the compound from step 3 2 338b (0.100 g, 0.246 mmol), the compound from step 3-340a (0.102 g, 0.246 mmol) and NaHC0 3 (82.8 mg, 0.985 mmol) in DME (6 mL) and H 2 0 (2 mL) was added Pd(PPh 3 ) 4 (14.2 mg, 12.3 ⁇ ). The resultant mixture were degassed and heated to 85°C under N 2 for 20 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step 3-341b A mixture of the crude compound from step 3-34 la (theo 0.183 mmol) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
  • Step 3-342a A mixture of l-Boc-pyrrolidine-3-carboxylic acid (0.159 g, 0.739 mmol), 4-bromo-1,2-diaminobenzene (0.142 g, 0.739 mmol) in DMF (6 mL) were added
  • Step 3-342c A mixture of the compound from step 3 ⁇ 342b (30.2 mg, 82.5 ⁇ ), the compound from step 3 ⁇ 3366 (36.2 mg, 82.5 ⁇ ) and NaHCOs (27.7 mg, 0.330 mmol) in DME (6 mL) and H 2 0 (2 mL) was added Pd(PPh 3 ) 4 (4.7 mg, 4.1 ⁇ ). The resultant mixture were degassed and heated to 85°C under N 2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step 3-344a A mixture of 2,4'-dibromoacetophenone (0.258 g, 0.929 mmol) and 1-Boc- pyrrolidine-3-carboxylic acid (0.200 g, 0.929 mmol) in CH3CN (9 mL) was added DIPEA (0.23 mL, 1.86 mmol) slowly. The mixture was stirred at room temperature until the disappearence of the starting material. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step 3-344c A mixture of the compound from step 3-344b (0.115 g, 0.293 mmol), bis- (pinacolato) diboron (0.149 g, 0.587 mmol) and potassium acetate (71.8 mg, 0.733 mmol) in 1,4-dioxane (8 mL) was added Pd(PPh 3 ) 4 (16.9 mg, 14.6 ⁇ ). The resultant mixture were degassed for three times and heated up to 85°C under N 2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step 3-344f A mixture of the compound from step 3-344c (30.0 mg, 68.3 ⁇ ), the compound from step 3 ⁇ 3446 (30.0 mg, 68.3 ⁇ ) and NaHC0 3 (22.9 mg, 0.273 mmol) in DME (6 mL) and H 2 0 (2 mL) was added Pd(PPh 3 ) 4 (3.9 mg, 3.4 ⁇ ). The resultant mixture were degassed and heated to 85°C under N 2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.
  • Step 3-346a To a solution of Boc-trans-2-aminocyclopentane carboxylic acid (racemic, 0.350 g, 1.526 mmol) in THF (8 mL) at 0°C was added iodomethane (0.76 mL, 12.21 mmol), followed by NaH (60% in mineral oil, 0.244 g, 6.106 mmol). The milky solution was stirred at 0°C for 15 minutes and then at room temperature for 3 hours before being quenched carefully with H 2 0. The volatiles were evaporated. The residue was diluted with H 2 0, washed with Et 2 0 (* 1). The aqueous layer was acidified with citric acid to pH ⁇ 3, extracted with dichloromethane.
  • Step 3-346d A mixture of compound from step 3-346c (89.0 mg, 0.212 mmol), the compound from step 3 ⁇ 3366 (93.0 mg, 0.212 mmol), Pd(PPh 3 ) 4 (24.5 mg, 21.2 ⁇ ) and NaHC0 3 (62.3 mg, 0.741 mmol) in DME (3 mL) and H 2 0 (1 mL) was degassed and heated at 98°C under N 2 for 15 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H 2 0). The organics were washed with brine, dried (Na 2 S0 4 ), filtered and evaporated.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention discloses compounds or pharmaceutically acceptable salts, esters, or prodrugs thereof, which inhibit RNA-containing virus, particularly the hepatitis C virus (HCV). 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. The present invention relates to novel antiviral compounds represented herein above, pharmaceutical compositions comprising such compounds, and methods for the treatment or prophylaxis of viral (particularly HCV) infection in a subject in need of such therapy with said compounds.

Description

HEPATITIS C VIRUS INHIBITORS
RELATED APPLICATION
This application claims the benefit of U.S. Provisional Application No. 61/241,489 filed September 11, 2009; U.S. Provisional Application No. 61/241,578 filed September 11, 2009; U.S. Provisional Application No. 61/241,595 filed September 11, 2009; U.S. Provisional Application No. 61/241,617 filed September 11, 2009; U.S. Provisional Application No. 61/241,577 filed September 11, 2009. The entire teachings of the above applications are incorporated herein by reference.
TECHNICAL FIELD
The present invention relates to novel antiviral agents. More specifically, the present invention relates to compounds which can inhibit the function of the NS5A protein encoded by Hepatitis C virus (HCV), compositions comprising such compounds, methods for inhibiting HCV viral replication, methods for treating or preventing HCV infection, and processes for making the compounds. BACKGROUND OF THE INVENTION
Infection with HCV is a major cause of human liver disease throughout the world. In the US, an estimated 4.5 million Americans are chronically infected with HCV.
Although only 30% of acute infections are symptomatic, greater than 85% of infected individuals develop chronic, persistent infection. Treatment costs for HCV infection have been estimated at $5.46 billion for the US in 1997. Worldwide over 200 million people are estimated to be infected chronically. HCV infection is responsible for 40-60% of all chronic liver disease and 30% of all liver transplants. Chronic HCV infection accounts for 30%) of all cirrhosis, end-stage liver disease, and liver cancer in the U.S. The CDC estimates that the number of deaths due to HCV will minimally increase to 38,000/year by the year 2010.
Due to the high degree of variability in the viral surface antigens, existence of multiple viral genotypes, and demonstrated specificity of immunity, the development of a successful vaccine in the near future is unlikely. Alpha-interferon (alone or in combination with ribavirin) has been widely used since its approval for treatment of chronic HCV infection. However, adverse side effects are commonly associated with this treatment: flulike symptoms, leukopenia, thrombocytopenia, depression from interferon, as well as anemia induced by ribavirin (Lindsay, K. L. (1997) Hepatology, 26 (suppl 1): 71S-77S). This therapy remains less effective against infections caused by HCV genotype 1 (which constitutes -75% of all HCV infections in the developed markets) compared to infections caused by the other 5 major HCV genotypes. Unfortunately, only -50-80% of the patients respond to this treatment (measured by a reduction in serum HCV R A levels and normalization of liver enzymes) and, of responders, 50-70% relapse within 6 months of cessation of treatment. Recently, with the introduction of pegylated interferon (Peg-IFN), both initial and sustained response rates have improved substantially, and combination treatment of Peg-IFN with ribavirin constitutes the gold standard for therapy. However, the side effects associated with combination therapy and the impaired response in patients with genotype 1 present opportunities for improvement in the management of this disease.
First identified by molecular cloning in 1989 (Choo, Q-L et al (1989) Science, 244:359-362), HCV is now widely accepted as the most common causative agent of posttransfusion non-A, non-B hepatitis (NANBH) (Kuo, G et al (1989) Science, 244:362-364). Due to its genome structure and sequence homology, this virus was assigned as a new genus in the Flaviviridae family. Like the other members of the Flaviviridae, such as flaviviruses (e.g. yellow fever virus and Dengue virus types 1-4) and pestiviruses (e.g. bovine viral diarrhea virus, border disease virus, and classic swine fever virus) (Choo, Q-L et al (1989) Science, 244:359-362; Miller, R.H. and R.H. Purcell (1990) Proc. Natl. Acad. Sci., USA 87:2057-2061), HCV is an enveloped virus containing a single strand RNA molecule of positive polarity. The HCV genome is approximately 9.6 kilobases (kb) with a long, highly conserved, noncapped 5' nontranslated region (NTR) of approximately 340 bases which functions as an internal ribosome entry site (IRES) (Wang CY et al 'An RNA pseudoknot is an essential structural element of the internal ribosome entry site located within the hepatitis C virus 5' noncoding region' RNA - A Publication of the RNA
Society, 1(5): 526-537, 1995 Jul). This element is followed by a region which encodes a single long open reading frame (ORF) encoding a polypeptide of -3000 amino acids comprising both the structural and nonstructural viral proteins.
Upon entry into the cytoplasm of the cell, this RNA is directly translated into a polypeptide of -3000 amino acids comprising both the structural and nonstructural viral proteins. This large polypeptide is subsequently processed into the individual structural and nonstructural proteins by a combination of host and virally-encoded proteinases (Rice, CM. (1996) in B.N. Fields, D.M.Knipe and P.M. Howley (eds) Virology, 2nd Edition, p931-960; Raven Press, N.Y.). 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 several 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. NS5A is a membrane-anchored phosphoprotein that is observed in basally phosphorylated (56 kDa) and hyperphosphorylated (58 kDa) forms. While its function has not fully been elucidated, NS5A is believed to be important in viral replication. The NS5B protein (591 amino acids, 65 kDa) of HCV (Behrens, S.E. et al (1996) EMBO J. , 151 2-22) encodes an R A-dependent RNA polymerase (RdRp) activity and contains canonical motifs present in other RNA viral polymerases. The NS5B protein is fairly well conserved both intra- typically (-95-98% amino acid (aa) identity across lb isolates) and inter-typically (-85% aa identity between genotype la and lb isolates). The essentiality of the HCV NS5B RdRp activity for the generation of infectious progeny virions has been formally proven in chimpanzees (A. A. Kolykhalov et al, (2000) Journal of Virology, 74(4): 2046-2051). Thus, inhibition of NS5B RdRp activity (inhibition of RNA replication) is predicted to be useful to treat HCV infection.
Following the termination codon at the end of the long ORF, there is a 3' NTR which roughly consists of three regions: an -40 base region which is poorly conserved among various genotypes, a variable length poly(U)/polypyrimidine tract, and a highly conserved 98 base element also called the "3' X-tail" (Kolykhalov, A. et al (1996) J. Virology 70:3363-3371; Tanaka, T. et al (1995) Biochem Biophys. Res. Commun., 215744-749; Tanaka, T. et al (1996) J. Virology, 70:3307-3312; Yamada, N. et al (1996) Virology, 223:255-261). The 3' NTR is predicted to form a stable secondary structure which is essential for HCV growth in chimps and is believed to function in the initiation and regulation of viral RNA replication.
Compounds useful for treating HCV-infected patients are desired which selectively inhibit HCV viral replication. In particular, compounds which are effective to inhibit the function of the NS5A protein are desired. The HCV NS5A protein is described, for example, in Tan, S.-L., Katzel, M.G. Virology, 2001, 284, 1; and in Rice, C. M. Nature, 2005, 435, 374.
Based on the foregoing, there exists a significant need to identify compounds with the ability to inhibit HCV. SUMMARY OF THE INVENTION
The present invention relates to novel antiviral compounds represented herein below, pharmaceutical compositions comprising such compounds, and methods for the treatment or prophylaxis of viral (particularly HCV) infection in a subject in need of such therapy with said compounds. Compounds of the present invention interfere with the life cycle of the hepatitis C virus and are also useful as antiviral agents.
In its first principal aspect, the present invention provides a compound of Formula
(1-1):
Figure imgf000005_0001
or a pharmaceutically acceptable salt thereof, wherein:
Ring A is absent or a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C3-C8 cycloalkyl, and C3-C8 cycloalkenyl, each optionally substituted; preferably optionally substituted aryl or optionally substituted heteroaryl;
Ring B is a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C3-C8 cycloalkyl, and C3-C8 cycloalkenyl, each optionally substituted; preferably optionally substituted aryl or optionally substituted heteroaryl;
L is absent or selected from the group consisting of optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, and optionally substituted C2-C4 alkynyl;
G is optionally substituted 5 -membered heteroaryl or optionally substituted 5/6- member fused heteroaryl; wherein the 5 -membered heteroaryl contains one or more nitrogen atoms, and wherein the 6-membered ring of said 5/6-fused membered heteroaryl is attached to one of Ring A, L, and Ring B, and is aryl or heteroaryl; preferably optionally substituted imidazolyl or optionally substituted benzimidazolyl;
J is selected from -N(Rlc)-C(0)-, -N(Rlc)-C(0)0- and -N(Rlc)-C(0)-N(Rlc)-; preferably -N(Rlc)-C(0)-;
W at each occurrence is independently O or -N(Rlb)-; preferably -N(Rlb)-;
R1, Rla, Rlb, Rlc, R9, and R9a at each occurence are each independently hydrogen or optionally substituted C1-C4 alkyl; alternatively Rla and R9a, Rla and R9, R1 and R9a, or R1 and R9 can be taken together with the carbon atom(s) to which they are attached to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or yet alternatively Rlb and R9a, or Rlb and R9 can be taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic ring;
R6 at each occurence is independently selected from the group consisting of optionally substituted 0(C1-C8 alkyl), optionally substituted amino, C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted; preferably optionally substituted C1-C8 alkyl; more preferably C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C1-C4 alkyl);
7 R R9a
Q is selected from: and ;
Figure imgf000006_0001
Figure imgf000006_0002
X is absent, O, S, CH2, or CH2CH2;
Y is absent, O, S, C(R , C(R1)2C(R7)2, C(R1)2C(R7)2C(R7)2, C(R1)2OC(R1)2, or C(R^SC(R1)2;
Wherein at least one of X and Y is present;
Wherein at least one of X and Y is not O or S;
U is absent or independently selected from O, S, S(O), S02, NC(0)-(C1-C4 alkyl),
C(O), protected carbonyl, OCH2, OCH2CH2, SCH2, SCH2CH2, C(R7)2, and C(R7)2C(R7)2; preferably CH2;
R7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C1-C4 alkyl), S(C1-C4 alkyl), amino optionally substituted with one or two C1-C4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C1-C4 alkyl; preferably hydrogen, halogen or hydroxy;
Optionally two geminal R7 groups can be taken together with the cabon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C3-C8 cycloalkyl, C3-C8 cycloalkenyl and 3- to 8- membered heterocyclic, each optionally substituted; preferably two geminal R7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted cyclopropyl or a spiro, optionally substituted 5- to 6-membered heterocyclic;
R2 at each occurence is independently hydrogen, optionally substituted C1-C8 alkyl, or -NRaRb; Ra at each occurence is independently hydrogen or optionally substituted C1-C8 alkyl;
Rb at each occurence is -C(0)-R6;
Alternatively Ra and Rb can be taken together with the nitrogen atom to which they are attached to form an optionally substituted heterocyclic or optionally substituted heteroaryl group;
R3 and R4 are each independently selected from the group consisting of hydrogen, optionally substituted C1-C8 alkyl, optionally substituted C2-C8 alkenyl, and optionally substituted C3-C8 cycloalkyl; preferably hydrogen or optionally substituted C1-C4 alkyl; alternatively, R3 and R4 can be taken together with the carbon atom to which they are attached to form optionally substituted C3-C8 cycloalkyl or optionally substituted heterocyclic;
R5 is independently hydrogen, optionally substituted C1-C8 alkyl, or optionally substituted C3-C8 cycloalkyl; preferably hydrogen or optionally substituted C1-C4 alkyl; and
R7a and R7b at each occurrence are each independently selected from the group consisting of hydrogen, optionally substituted aryl, and optionally substituted C1-C4 alkyl; alternatively, CHR7a-U or CHR7b-U can be taken together to form a group selected from CH=CH, optionally substituted C3-C8 cycloalkyl, optionally substituted aryl, or optionally substituted heterocyclic; or yet alternatively, U, R7a, and R7b can be taken together with the carbon atoms to which they are attached to form a bridged, optionally substituted 4- to 7- membered ring including C4-C7 cycloalkyl, C4-C7 cycloalkenyl and 4- to 7-membered heterocyclic.
In its second principal aspect, the present invention provides a compound of Formula (2-1)
Figure imgf000007_0001
or a pharmaceutically acceptable salt thereof, wherein:
Ring A and Ring B are each independently a monocyclic or polycyclic group selected from aryl, heteroaryl, heterocyclic, C3-C8 cycloalkyl, and C3-C8 cycloalkenyl, each optionally substituted; preferably optionally substituted aryl or optionally substituted heteroaryl; L is absent or selected from the group consisting of optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, and optionally substituted C2-C4 alkynyl; wherein
Figure imgf000008_0002
taken together is not optionally substituted phenyl-thiazolyl or 3,4'-biphenyl;
J is selected from the group consisting of -N(Rlc)-C( O)-, -N(Rlc)-C(0)0- and
-N(Rlc)-C(0)-N(Rlc)-; preferably -N(Rlc)-C(0)-;
W at each occurrence is independently O or -N(Rlb)-; preferably -N(Rlb)-;
R1, Rla, Rlb, Rlc, R9, and R9a at each occurrence are each independently hydrogen or optionally substituted C1-C4 alkyl; alternatively, Rla and R9a, Rla and R9, R1 and R9a, or RJ and R9 can be taken together with the carbon atom(s) to which they are attached to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or yet alternatively Rlb and R9a, or Rlb and R9 can be taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic;
R6 at each occurrence is independently selected from the group consisting of optionally substituted 0(C1-C8 alkyl); optionally substituted amino; C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted; preferably optionally substituted C1-C8 alkyl; more preferably C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C1-C4 alkyl);
G is selected from the group consisting of -N(Rlc)-C(0)-, -N(Rlc)-C(0)0- and -N(Rlc)-C( -N(Rlc)-; preferably -N(Rlc)-C(0)-;
; or alternatively G and Q are taken together to form
Figure imgf000008_0001
X is absent, O, S, CH2, or CH2CH2;
Y at is absent, O, S, C(R , CCR^C(R7^, CCR^C^C^, C(R^OCCR1^, or C(R^SCCR1^; Wherein at least one of X and Y is present;
Wherein at least one of X and Y is not O or S;
R7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C1-C4 alkyl), S(C1-C4 alkyl), amino optionally substituted with one or two C1-C4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C1-C4 alkyl; preferably hydrogen, halogen or hydroxy;
R2 at each occurrence is independently hydrogen, optionally substituted C1-C8 alkyl, or -NRaRb;
Ra at each occurrence is independently hydrogen or optionally substituted C1-C8 alkyl;
Rb at each occurrence is -C(0)-R6; and
Alternatively Ra and Rb can be taken together with the nitrogen atom to which they are attached to form an optionally substituted heterocyclic or optionally substituted heteroaryl group.
In its third principal aspect, the present invention provides a compound of Formula
(3-1):
Figure imgf000009_0001
or a pharmaceutically acceptable salt thereof, wherein:
Ring A and Ring B are each independently absent or a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C3-C8 cycloalkyl, and C3-C8 cycloalkenyl, each optionally substituted; preferably optionally substituted aryl or optionally substituted heteroaryl;
L is absent or selected from the group consisting of optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, and optionally substituted C2-C4 alkynyl;
Wherein at least one of Ring A, Ring B and L is present;
G and J are each independently optionally substituted 5-membered heteroaryl or optionally substituted 5/6-member fused heteroaryl; wherein the 5-membered heteroaryl contains one or more nitrogen atoms, and wherein the 6-membered ring of said 5/6-fused membered heteroaryl is attached to one of Ring A, Ring B and L, and is aryl or heteroaryl; preferably optionally substituted imidazolyl or optionally substituted benzimidazolyl; W at each occurrence is independently O or -N(Rlb)-; preferably -N(Rlb)-;
R1, Rla, Rlb, R9, and R9a at each occurrence are each independently hydrogen or optionally substituted C1-C4 alkyl; alternatively Rla and R9a, Rla and R9, R1 and R9a, or R1 and R9 can be taken together with the carbon atom(s) to which they are attached to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or yet alternatively Rlb and R9a, or Rlb and R9 can be taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic;
R6 at each occurrence is independently selected from the group consisting of optionally substituted 0(C1-C8 alkyl); optionally substituted amino; C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted; preferably optionally substituted C1-C8 alkyl; more preferably C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C1-C4 alkyl);
Q is selected from:
Figure imgf000010_0001
, and;
Figure imgf000010_0002
X is absent, O, S, CH2, or CH2CH2;
Y is absent, O, S, C(R , C(R1)2C(R7)2, C(R1)2C(R7)2C(R7)2, C(R1)2OC(R1)2, or C(R^SC(R1)2;
Wherein at least one of X and Y is present;
Wherein at least one of X and Y is not O or S;
U is absent or independently selected from O, S, S(O), S02, NC(0)-(C1-C4 alkyl), C(O), protected carbonyl, OCH2, OCH2CH2, SCH2, SCH2CH2, C(R7)2, and C(R7)2C(R7)2; preferably CH2;
R7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C1-C4 alkyl), S(C1-C4 alkyl), amino optionally substituted with one or two C1-C4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C1-C4 alkyl; preferably hydrogen, halogen or hydroxy;
Optionally two geminal R7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C3-C8 cycloalkyl, C3-C8 cycloalkenyl and 3- to 8- membered heterocyclic, each optionally substituted; preferably an optionally substituted cyclopropyl or an optionally substituted 5- to 6-membered heterocyclic;
R2 at each occurrence is independently hydrogen, optionally substituted C1-C8 alkyl, or -NRaRb;
Ra at each occurrence is independently hydrogen or optionally substituted C1-C8 alkyl;
Rb at each occurrence is -C(0)-R6;
Alternatively Ra and Rb can be taken together with the nitrogen atom to which they are attached to form an optionally substituted heterocyclic or optionally substituted heteroaryl group;
R3 and R4 are each independently selected from the group consisting of hydrogen, optionally substituted C1-C8 alkyl, optionally substituted C2-C8 alkenyl, and optionally substituted C3-C8 cycloalkyl; preferably hydrogen or optionally substituted C1-C4 alkyl; alternatively, R3 and R4 can be taken together with the carbon atom to which they are attached to form optionally substituted C3-C8 cycloalkyl or optionally substituted heterocyclic;
R5 is independently hydrogen, optionally substituted C1-C8 alkyl, or optionally substituted C3-C8 cycloalkyl; preferably hydrogen or optionally substituted C1-C4 alkyl; and
R7a and R7b at each occurrence are each independently selected from the group consisting of hydrogen, optionally substituted aryl, and optionally substituted C1-C4 alkyl; alternatively, CHR7a-U or CHR7b-U can be taken together to form a group selected from CH=CH, optionally substituted C3-C8 cycloalkyl, optionally substituted aryl, or optionally substituted heterocyclic; or yet alternatively, U, R7a, and R7b can be taken together with the carbon atoms to which they are attached to form a bridged, optionally substituted 4- to 7- membered ring including C4-C7 cycloalkyl, C4-C7 cycloalkenyl and 4- to 7-membered heterocyclic, each optionally substituted.
In its fourth principal aspect, the present invention provides a compound of Formula (4-1):
Figure imgf000011_0001
or a pharmaceutically acceptable salt thereof, wherein:
Ring A is absent or a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C3-C8 cycloalkyl, and C3-C8 cycloalkenyl, each optionally substituted; preferably optionally substituted aryl or optionally substituted heteroaryl;
Ring B is a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C3-C8 cycloalkyl, and C3-C8 cycloalkenyl, each optionally substituted; preferably optionally substituted aryl or optionally substituted heteroaryl;
L is absent or selected from the group consisting of optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, and optionally substituted C2-C4 alkynyl;
R1 at each occurrence is independently hydrogen or optionally substituted C1-C4 alkyl;
R6 at each occurrence is independently selected from the group consisting of optionally substituted 0(C1-C8 alkyl); optionally substituted amino; C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted; preferably optionally substituted C1-C8 alkyl; more preferably C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C1-C4 alkyl);
U is absent or independently selected from O, S, S(O), S02, NC(0)-(C1-C4 alkyl), C(O), protected carbonyl, OCH2, OCH2CH2, SCH2, SCH2CH2, C(R7)2, and C(R7)2C(R7)2; preferably CH2;
R7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C1-C4 alkyl), S(C1-C4 alkyl), amino optionally substituted with one or two C1-C4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C1-C4 alkyl; preferably hydrogen, halogen or hydroxy;
Optionally two geminal R7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C3-C8 cycloalkyl, C3-C8 cycloalkenyl and 3- to 8- membered heterocyclic, each optionally substituted; preferably an optionally substituted C3-C8 cyclopropyl or an optionally substituted 5- to 6-membered heterocyclic;
R7a and R7b at each occurrence are each independently selected from the group consisting of hydrogen, optionally substituted aryl, and optionally substituted C1-C4 alkyl; alternatively, CHR7a-U or CHR7b-U can be taken together to form a group selected from CH=CH, optionally substituted C3-C8 cycloalkyl, optionally substituted aryl, and optionally substituted heterocyclic;
G is optionally substituted 5 -membered heteroaryl or optionally substituted 5/6- member fused heteroaryl; wherein the 5 -membered heteroaryl contains one or more nitrogen atoms, and wherein the 6-membered ring of said 5/6-fused membered heteroaryl is attached to one of Ring A, L and Ring B, and is aryl or heteroaryl; preferably optionally substituted imidazolyl or optionally substituted benzimidazolyl;
Q is selected from:
Figure imgf000013_0001
,
Figure imgf000013_0002
Alternatively G and Q are taken together to form
Figure imgf000013_0003
Figure imgf000013_0004
V is selected from the group consisting of -N(Rlc)-, -N(Rlc)-C(0)-, -N(Rlc)- C(0)0- and -N(Rlc)-C(0)-N(Rlc)-; preferably -N(Rlc)- or -N(Rlc)-C(0)-;
W is O or -N(Rlb)-; preferably -N(Rlb)-;
Rla, Rlb, Rlc, Rld, R9, and R9a at each occurrence are each independently hydrogen or optionally substituted C1-C4 alkyl; alternatively Rla and R9a, Rla and R9, Rld and R9a, or Rld and R9 can be taken together with the carbon atom(s) to which they are attached to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or yet alternatively Rlb and R9a, or Rlb and R9 can be taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic;
X is absent, O, S, CH2, or CH2CH2;
Y is absent, O, S, C(R , C(R1)2C(R7)2, C(R1)2C(R7)2C(R7)2, C(R1)2OC(R1)2, or C(R^SC(R1)2;
Wherein at least one of X and Y is present;
Wherein at least one of X and Y is not O or S;
R2 is hydrogen, optionally substituted C1-C8 alkyl, or
Ra is hydrogen or optionally substituted C1-C8 alkyl;
Rb at each occurrence is -C(0)-R6; Alternatively Ra and Rb can be taken together with the nitrogen atom to which they are attached to form an optionally substituted heterocyclic or optionally substituted heteroaryl group;
R3 and R4 are each independently selected from the group consisting of hydrogen, optionally substituted C1-C8 alkyl, optionally substituted C2-C8 alkenyl, and optionally substituted C3-C8 cycloalkyl; preferably hydrogen or optionally substituted C1-C4 alkyl; alternatively, R3 and R4 can be taken together with the carbon atom to which they are attached to form optionally substituted C3-C8 cycloalkyl or optionally substituted heterocyclic; and
R5 is independently hydrogen, optionally substituted C1-C8 alkyl, or optionally substituted C3-C8 cycloalkyl; preferably hydrogen or optionally substituted C1-C4 alkyl.
In its fifth principle aspect, the present invention provides a compound of Formula
(5-1):
Figure imgf000014_0001
or a pharmaceutically acceptable salt thereof, wherein:
Ring A is absent or a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C3-C8 cycloalkyl, and C3-C8 cycloalkenyl, each optionally substituted; preferably optionally substituted aryl or optionally substituted heteroaryl;
Ring B is an optionally substituted aryl or optionally substituted heteroaryl;
L is absent or selected from the group consisting of optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, and optionally substituted C2-C4 alkynyl;
W at each occurrence is independently O or -N(Rlb)-; preferably -N(Rlb)-;
R1, Rla, Rlb, Rlc, R9, and R9a at each occurrence are each independently hydrogen or optionally substituted C1-C4 alkyl; alternatively Rla and R9a, Rla and R9, R1 and R9a, or R1 and R9 can be taken together with the carbon atom(s) to which they are attached to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or yet alternatively Rlb and R9a, or Rlb and R9 can be taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic;
R6 at each occurrence is independently selected from the group consisting of optionally substituted 0(C1-C8 alkyl); optionally substituted amino; C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted; preferably optionally substituted C1-C8 alkyl; more preferably C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C1-C4 alkyl);
G is optionally substituted 5 -membered heteroaryl or optionally substituted 5/6- member fused heteroaryl; wherein the 5 -membered heteroaryl contains one or more nitrogen atoms, and wherein the 6-membered ring of said 5/6-fused membered heteroaryl is attached to one of Ring A, L and Ring B and is aryl or heteroaryl; preferably optionally substituted imidazolyl or optionally substituted benzimidazolyl;
Q is selected from:
Figure imgf000015_0004
Figure imgf000015_0003
;
tively G and Q are taken together to form
Figure imgf000015_0001
or
Figure imgf000015_0002
V is selected from the group consisting of -N(Rlc)-, -N(Rlc)-C(0)-, -N(Rlc)- C(0)0- and -N(Rlc)-C(0)-N(Rlc)-;
X is absent, O, S, CH2, or CH2CH2;
Y is absent, O, S, C(R , C(R1)2C(R7)2, C(R1)2C(R7)2C(R7)2, C(R1)2OC(R1)2, or C(R^SC(R1)2;
Wherein at least one of X and Y is present;
Wherein at least one of X and Y is not O or S;
R7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C1-C4 alkyl), S(C1-C4 alkyl), amino optionally substituted with one or two C1-C4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C1-C4 alkyl; preferably hydrogen, halogen or hydroxy;
U is absent or selected from O, S, S(O), S02, NC(0)-(C C4 alkyl), C(O), protected carbonyl, OCH2, OCH2CH2, SCH2, SCH2CH2, C(R7)2, and C(R7)2C(R7)2; preferably CH2; Optionally two geminal R7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C3-C8 cycloalkyl, C3-C8 cycloalkenyl and 3- to 8- membered heterocyclic, each optionally substituted; preferably an optionally substituted cyclopropyl or an optionally substituted 5- to 6-membered heterocyclic;
R2 is hydrogen, optionally substituted C1-C8 alkyl, or -NRaRb;
Ra is hydrogen or optionally substituted C1-C8 alkyl;
Rb is -C(0)-R6;
Alternatively Ra and Rb can be taken together with the nitrogen atom to which they are attached to form an optionally substituted heterocyclic or optionally substituted heteroaryl group;
R3 and R4 are each independently selected from the group consisting of hydrogen, optionally substituted C1-C8 alkyl, optionally substituted C2-C8 alkenyl, and optionally substituted C3-C8 cycloalkyl; preferably hydrogen or optionally substituted C1-C4 alkyl; alternatively, R3 and R4 can be taken together with the carbon atom to which they are attached to form optionally substituted C3-C8 cycloalkyl or optionally substituted heterocyclic;
R5 is hydrogen, optionally substituted C1-C8 alkyl, or optionally substituted C3-C8 cycloalkyl; preferably hydrogen or optionally substituted C1-C4 alkyl; and
R7a and R7b at each occurrence are each independently selected from the group consisting of hydrogen, optionally substituted aryl, and optionally substituted C1-C4 alkyl; alternatively, CHR7a-U or CHR7b-U can be taken together to form a group selected from CH=CH, optionally substituted C3-C8 cycloalkyl, optionally substituted aryl, or optionally substituted heterocyclic; or yet alternatively, U, R7a , and R7b can be taken together with the carbon atoms to which they are attached to form a bridged, optionally substituted 4- to 7-membered ring including C4-C7 cycloalkyl, C4-C7 cycloalkenyl and 4- to 7-membered heterocyclic, each optionally substituted.
Each preferred group stated above can be taken in combination with one, any or all other preferred groups.
In another aspect, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound or combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable carrier or excipient. In yet another aspect, the present invention provides a method of inhibiting the replication of a RNA-containing virus comprising contacting said virus with said pharmaceutical composition. Particularly, this invention is directed to methods of inhibiting the replication of HCV.
In still another aspect, the present invention provides a method of treating or preventing infection caused by an RNA-containing virus comprising administering to a patient in need of such treatment a therapeutically effective amount of a compound or combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof. Particularly, this invention is directed to methods of treating or preventing infection caused by HCV.
Yet another aspect of the present invention provides the use of a compound or combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof, as defined hereinafter, in the preparation of a medicament for the treatment or prevention of infection caused by RNA-containing virus, specifically HCV. DETAILED DESCRIPTION OF THE INVENTION
The compounds of the invention have utility in inhibiting the replication of RNA- containing virus, including, for example, HCV. Other compounds useful for inhibiting the replication of RNA-containing viruses and/or for the treatment or prophylaxis of HCV infection have been described in copending U.S. Application Serial No. 12/702,673 filed February 9, 2010 entitled "Linked Dibenzimidiazole Derivatives"; U.S. Application Serial No. 12/702,692 filed February 9, 2010 entitled "Linked Dibenzimidiazole Derivatives"; U.S. Application Serial No. 12/702,802 filed February 9, 2010 entitled "Linked
Dibenzimidiazole Derivatives"; U.S. Application Serial No. 12/707,190 filed February 17, 2010 entitled "Linked Diimidazole Antivirals"; U.S. Application Serial No. 12/707,200 filed February 17, 2010 entitled "Linked Diimidazole Derivatives"; U.S. Application
Serial No. 12/707,210 filed February 17, 2010 entitled "Hepatitis C Virus Inhibitors"; U.S. Application Serial No. 12/714,583 filed March 1, 2010 entitled "Novel Benzimidazole Derivatives"; and U.S. Application Serial No. 12/714,576 filed March 1, 2010 entitled "Hepatitis C Virus Inhibitors"; U.S. Application Serial No. 12/816,148 filed June 15, 2010 entitled "Hepatitis C Virus Inhibitors"; U.S. Application Serial No. 12/816,171 filed June 15, 2010 entitled "Hepatitis C Virus Inhibitors"; U.S. Provisional Application Serial No. 61/241,489 filed September 11, 2009 entitled "Hepatitis C Virus Inhibitors"; U.S.
Provisional Application Serial No. 61/241,578 filed September 11, 2009 entitled "Hepatitis C Virus Inhibitors"; U.S. Provisional Application Serial No. 61/241,595 filed September 11, 2009 entitled "Hepatitis C Virus Inhibitors"; U.S. Provisional Application Serial No. 61/241,617 filed September 11, 2009 entitled "Hepatitis C Virus Inhibitors"; U.S. Provisional Application Serial No. 61/241,577 filed September 11, 2009 entitled "Hepatitis C Virus Inhibitors"; U.S. Provisional Application Serial No. 61/241,598 filed September 11, 2009 entitled "Hepatitis C Virus Inhibitors"; U.S. Provisional Application Serial No. 61/286,178 filed December 14, 2009 entitled "Hepatitis C Virus Inhibitors"; U.S. Provisional Application Serial No. 61/297,918 filed January 25, 2010 entitled "Hepatitis C Virus Inhibitors"; U.S. Provisional Application Serial No. 61/314,304 filed March 16, 2010 entitled "Hepatitis C Virus Inhibitors"; U.S. Provisional Application Serial No. 61/322,438 filed April 9, 2010 entitled "Hepatitis C Virus Inhibitors"; U.S. Provisional Application Serial No. 61/351,327 filed June 4, 2010 entitled "Hepatitis C Virus Inhibitors"; U.S. Provisional Application Serial No. 61/372,999 filed August 12, 2010 entitled "Hepatitis C Virus Inhibitors"; and the contents of each of which are expressly incorporated by reference herein.
As discussed above, a general strategy for the development of antiviral agents is to inactivate virally encoded proteins, including NS5A, that are essential for the replication of the virus. The relevant patent disclosures describing the synthesis of HCV NS5A inhibitors are: US 2009/0202478; US 2009/0202483; WO 2004/014852; WO
2006/079833; WO 2006/1333262; WO 2007/031791; WO 2007/070556; WO
2007/070600; WO 2007/082554; WO 2008/021927; WO 2008/021928; WO
2008/021936; WO 2008/048589; WO 2008/064218; WO 2008/070447; WO
2008/144380; WO 2008/154601; WO 2009/020825; WO 2009/020828; WO
2009/034390; WO 2009/102318; WO 2009/102325; WO 2009/102694; WO
2010/017401; WO 2010/039793; WO 2010/065668; WO 2010/065674; WO
2010/065681; WO 2010/091413; WO 2010/096777; WO 2010/096462 and WO
2010/096302„the contents of each of which are expressly incorporated by reference herein.
I. Compounds having Formula (1-1)
In certain aspects, the present invention relates to compounds of Formula (1-1) as illustrated above, and pharmaceutically acceptable salts thereof. In another embodiment, the present invention relates to compounds of Formula (1- I), and pharmaceutically acceptable salts thereof; wherein J is -NH-C(O)-.
In still another embodiment, the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein J is -NH-C(0)0-.
In still another embodiment, the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein J is -NH-C(O)-
NH
In yet another embodiment, the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein Rla and R9a, Rla and R9, R1 and R9a, or R1 and R9 are taken together to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic.
In a further embodiment, the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein Rlb and R9a, or Rlb and R9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic.
In yet another embodiment, the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein the moiety of
Figure imgf000019_0001
is illustrated by one of the following core groups:
Figure imgf000019_0002
wherein R6 is as previously defined in Formula (1-1) and each of the above core groups is optionally substituted.
In an additional embodiment, the invention relates to compounds of Formula (1-1) and pharmaceutically acceptable salts thereof; wherein R^ is independently an optionally substituted C1-C8 alkyl. In a further embodiment, the invention relates to compounds of Formula (1-1) and pharmaceutically acceptable salts thereof; wherein -C(0)R6 is an a-amino acid residue.
In yet another embodiment, the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein G is an optionally substituted five-membered heteroaryl containing one or more nitrogen atoms, and is C-attached.
In yet another embodiment, the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein G is an optionally substituted 5/6-membered fused heteroaryl; wherein the 5-membered ring of said 5/6-membered fused heteroaryl is a heteroaryl containing one or more nitrogen atoms and wherein the 5- membered ring is C-attached to group Q, and wherein the 6-membered ring of said 5/6- membered fused heteroaryl is aryl or heteroaryl and is C-attached to group one of Ring A, L and Ring B.
In yet another embodiment, the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein G is illustrated by one of the following heteroaryl groups:
Figure imgf000020_0001
wherein each of the above said heteroaryl groups is optionally substituted.
In yet another embodiment, the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein G is optionally substituted imidazolyl or benzimidazolyl.
In yet another embodiment, the present invention relates to compounds of
Formulae (1-Ia-l ~ l-Ia-4), and pharmaceutically acceptable salts thereof:
Figure imgf000021_0001
wherein Ring A, Ring B, G, J, L, U, W, X, Y, R1, R2, R3, R4, R5, R6, R9, Rla, Rlb, R7a, R7b, and R9a are as previously defined in Formula (1-1).
In yet another embodiment, the present invention relates to compounds of
Formulae (l-Ia-5 ~ l-Ia-6), and pharmaceutically acceptable salts thereof:
Figure imgf000021_0002
wherein Ring A, Ring B, G, J, L, R1, R6, R9, Rla, Rlb, and R9a are as previously defined in Formula (1-1 ).
In yet another embodiment, the present invention relates to compounds of Formula (l-Ia-7), and pharmaceutically acceptable salts thereof:
Figure imgf000021_0003
wherein Ring A, Ring B, G, J, L, R1, R6, R9, Rla, Rlb, and R9a are as previously defined in Formula (1-1).
In yet another embodiment, the present invention relates to compounds of Formula (l-Ia-7), and pharmaceutically acceptable salts thereof; wherein R6 at each occurrence is independently selected from the group consisting of C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted.
In still another embodiment, the present invention relates to compounds of Formula (l-Ia-7), and pharmaceutically acceptable salts thereof; wherein R6 at each independently occurrence is independently C1-C8 alkyl optionally substituted with amino, hydoxy, protected amino, or 0(C1-C4 alkyl).
In still another embodiment, the present invention relates to compounds of Formulae (1-Ib-l ~ l-Ib-4), and pharmaceutically acceptable salts thereof:
Figure imgf000022_0001
(1-lb-4)
wherein Ring B, G, J, Q, R1, R6, R9, Rla, Rlb, and R9a are as previously defined in Formula (1-1); in Formula (1-Ib-l), Ring A and L are each present and as previously defined in Formula (1-1); in Formula (l-Ib-2), Ring A is present and as previously defined in Formula (1-1); in Formula (l-Ib-3), L is present and as previously defined in Formula (1- I)
In still another embodiment, the present invention relates to compounds of Formulae (1-Ib-l ~ l-Ib-4), and pharmaceutically acceptable salts thereof; wherein R6 is independently C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C1-C4 alkyl).
In another embodiment, the present invention relates to compounds of Formula (1- Ib-1), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or optionally substituted monocyclic heteroaryl; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
In yet another embodiment, the present invention relates to compounds of Formula (l-Ib-2), and pharmaceutically acceptable salts thereof; wherein one of Ring A and Ring B is optionally substituted phenyl or optionally substituted monocyclic heteroaryl, the other of Ring A and Ring B is optionally substituted bicyclic aryl or bicyclic heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
In a further embodiment, the present invention relates to compounds of Formula (l-Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted bicyclic aryl or bicyclic heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
In another embodiment, the present invention relates to compounds of Formula (1-
Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl. In yet another embodiment, the present invention relates to compounds of Formula (l-Ib-3), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted bicyclic aryl or bicyclic heteroaryl; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
In a further embodiment, the present invention relates to compounds of Formula (l-Ib-3), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
In another embodiment, the present invention relates to compounds of Formula (1- Ib-4), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted polycyclic aryl or polycyclic heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of
Formula (1-1), and pharmaceutically acceptable salts thereof; wherein two geminal R7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C3-C8 cycloalkyl, C3-C8 cycloalkenyl and 3- to 8-membered heterocyclic, each optionally substituted.
In still another embodiment, the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein two geminal R7 groups can be taken together with the carbon atoms to which they are attached to form a spiro, optionally substituted cyclopropyl or a spiro, optionally substituted 5- to 6- membered heterocyclic.
In still another embodiment, the present invention relates to compounds of Formula (1-1), and pharmaceutically acceptable salts thereof; wherein R7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C1-C4 alkyl), S(C1-C4 alkyl), amino optionally substituted with one or two C1-C4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C1-C4 alkyl; preferably hydrogen, halogen or hydroxyl. In still another embodiment, the present invention relates to com ounds of
Formula (1-1), and pharmaceutically acceptable salts thereof; wherein
Figure imgf000024_0001
at each occurence is independently illustrated by one of the following groups:
Figure imgf000024_0002
Representative compounds of the present invention are those selected from compounds 1-1 to 1-374 and 1-377 to 1-382 compiled in the following tables and/or shown below:
Figure imgf000024_0003
Figure imgf000025_0001
Figure imgf000026_0001
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
It is intended that the definition of any substituent or variable (e.g., R , R , etc.) a particular location in a molecule be independent of its definitions elsewhere in that molecule. For example, when U is C(R7)2, each of the two R7 groups may be the same or different.
It is also intended that the drawn formula of any linker or spacer (e.g., -NH- C(O)-, -OC(0)-NH-, etc.) at a particular location in a molecule be independent of its directions of attachment, unless otherwise indicated. For example, when a linker is -NH- C(O)-, it is meant to include -NH-C(O)- and -C(0)-NH- for attachments. II. Compounds having Formula (2-1)
In certain aspects, the present invention relates to compounds of Formula (2-1) as illustrated above, and pharmaceutically acceptable salts thereof.
In another embodiment, the present invention relates to compounds of Formula (2- I), and pharmaceutically acceptable salts thereof; wherein G is -NH-C(O)-.
In still another embodiment, the present invention relates to compounds of Formula (2-1), and pharmaceutically acceptable salts thereof; wherein G is -NH-C(0)0-.
In still another embodiment, the present invention relates to compounds of Formula (2-1), and pharmaceutically acceptable salts thereof; wherein G is -NH-C(O)- NH-.
In still another embodiment, the present invention relates to compounds of Formula (2-1), and pharmaceutically acceptable salts thereof; wherein J is -NH-C(O)-.
In still another embodiment, the present invention relates to compounds of Formula (2-1), and pharmaceutically acceptable salts thereof; wherein J is -NH-C(0)0-.
In still another embodiment, the present invention relates to compounds of
Formula (2-1), and pharmaceutically acceptable salts thereof; wherein J is -NH-C(O)- NH-.
In still another embodiment, the present invention relates to compounds of Formula (2-1), and pharmaceutically acceptable salts thereof;; wherein each of G and J is -NH-C(O)-.
In yet another embodiment, the present invention relates to compounds of Formula (2-1), and pharmaceutically acceptable salts thereof; wherein Rla and R9a, Rla and R9, R1 and R9a, or R1 and R9 are taken together to form an optionally substituted C3-C8 cycloalkyl or an optionally substituted 4- to 8-membered heterocyclic.
In a further embodiment, the present invention relates to compounds of Formula
(2-1), and pharmaceutically acceptable salts thereof; wherein Rlb and R9a, or Rlb and R9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic.
In yet another embodiment, the present invention relates to compounds of Formula (2-1), and pharmaceutically acceptable salts thereof; wherein the moiety of
Figure imgf000038_0001
is illustrated by the one of the following core groups:
Figure imgf000039_0001
wherein R6 is as previously defined in Formula (2-1) and each of the above core groups is optionally substituted.
In an additional embodiment, the invention relates to compounds of Formula (2-1) and pharmaceutically acceptable salts thereof; wherein R6 is independently an optionally substituted C1-C8 alkyl.
In a further embodiment, the invention relates to compounds of Formula (2-1) and pharmaceutically acceptable salts thereof; wherein -C(0)R6 is an a-amino acid residue.
In yet another embodiment, the present invention relates to compounds of Formulae (2-Ia-l ~ 2-Ia-2), and pharmaceutically acceptable salts thereof:
Figure imgf000039_0002
wherein Ring A, Ring B, G, J, L, ,W, X, Y, R1, R2, , R6, R9, Rla, Rlb, Rlc, and R9a are as previously defined in Formula (2-1).
In yet another embodiment, the present invention relates to compounds of Formulae (2-Ia-3 ~ 2-Ia-4),and pharmaceutically acceptable salts thereof:
Figure imgf000039_0003
wherein Ring A, Ring B, G, J, L, Y, R1, R2, R6, R9, Rla, Rlb, Rlc, and R9a are as previously defined in Formula (2-1).
In another embodiment, the present invention relates to compounds of Formula (2- Ia-5), and pharmaceutically acceptable salts thereof:
Figure imgf000040_0001
wherein Ring A, Ring B, G, J, L, Y, R1, R6, R9, Ra, Rla, Rlb, Rlc, and R9a are as previously defined in Formula (2-1).
In yet another embodiment, the present invention relates to compounds of Formula (2-Ia-5), and pharmaceutically acceptable salts thereof; wherein R6 at each occurrence is independently selected from the group consisting of C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted.
In still another embodiment, the present invention relates to compounds of Formula (2-Ia-5), and pharmaceutically acceptable salts thereof; wherein R6 at each occurrence is independently C1-C8 alkyl optionally substituted with amino, hydoxy, protected amino, or 0(C1-C4 alkyl).
In a further embodiment, the present invention relates to compounds of Formulae (2-Ib-l ~ 2-Ib-2), and pharmaceutically acceptable salts thereof: ) (2-lb-2)
Figure imgf000040_0002
wherein Ring A, Ring B, G, J, Q, R1, R6, R9, Rla, Rlb, and R9a are as previously defined; and in Formula (2-lb-l), L is present and as previously defined in Formula (2-1).
In another embodiment, the present invention relates to compounds of Formulae (2-Ib-l ~ 2-Ib-2), and pharmaceutically acceptable salts thereof; wherein R6 is C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C1-C4 alkyl).
In yet another embodiment, the present invention relates to compounds of Formula (2-Ib-l), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; and L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl.
In still another embodiment, the present invention relates to compounds of
Formula (2-Ib-2), and pharmaceutically acceptable salts thereof; wherein one of Ring A and Ring B is optionally substituted phenyl or optionally substituted monocyclic heteroaryl, and the other of Ring A and Ring B is optionally substituted bicyclic aryl or heteroaryl. In a further embodiment, the present invention relates to compounds of Formula (2-Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted bicyclic aryl or heteroaryl.
In another embodiment, the present invention relates to compounds of Formula (2- Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl.
Representative compounds of the present invention are those selected from compounds 2-1 to 2-295 compiled in the following tables and/or shown below:
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000047_0002
Figure imgf000048_0001
Figure imgf000049_0002
It is intended that the definition of any substituent or variable (e.g., R , R , etc.) at a particular location in a molecule be independent of its definitions elsewhere in that molecule. For example, when Y is C(R1)2, each of the two R1 groups may be the same or different.
It is also intended that the drawn formula of any linker or spacer (e.g., -NH- C(O)-, -OC(0)-NH-, etc.) at a particular location in a molecule be independent of its directions of attachment, unless otherwise indicated. For example, when a linker is -NH- C(O)-, it is meant to include -NH-C(O)- and -C(0)-NH- for attachments.
III. Compounds having Formula (3-1)
In certain aspects, the present invention relates to compounds of Formula (3-1) as illustrated above, and pharmaceutically acceptable salts thereof.
In yet another embodiment, the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein Rla and R9a, Rla and R9, R1 and R9a, or R1 and R9 are taken together to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic.
In a further embodiment, the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein Rlb and R9a, or Rlb and R9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic.
In one embodiment, the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein the moiety of
Figure imgf000049_0001
is illustrated by one of the following core groups:
Figure imgf000050_0001
wherein R6 is as previously defined and each of the above core groups is optionally substituted.
In an additional embodiment, the invention relates to compounds of Formula (3-1) and pharmaceutically acceptable salts thereof; wherein R6 is independently an optionally substituted C1-C8 alkyl.
In a further embodiment, the invention relates to compounds of Formula (3-1) and pharmaceutically acceptable salts thereof; wherein -C(0)R6 is an a-amino acid residue.
In yet another embodiment, the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein G and J are each
independently an optionally substituted five-membered heteroaryl containing one or more nitrogen atoms, and are each C-attached.
In yet another embodiment, the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein G and J are each
independently an optionally substituted 5/6-membered fused heteroaryl; wherein the 5- membered ring of said 5/6-membered fused heteroaryl is a heteroaryl containing one or more nitrogen atoms, and wherein the 6-membered ring of said 5/6-membered fused aryl is aryl or heteroaryl and is C-attached to one of Ring A, Ring B and L.
In yet another embodiment, the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein one of G and J is an optionally substituted five-membered heteroaryl containing one or more nitrogen, and is each C-attached; the other of G and J is an optionally substituted 5/6-membered fused heteroaryl; wherein the 5-membered ring of said 5/6-membered fused heteroaryl is a heteroaryl containing one or more nitrogen atoms, and wherein the 6-membered ring of said 5/6-membered fused aryl is aryl or heteroaryl and is C-attached to one of Ring A, L and Ring B. In yet another embodiment, the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein G and J are each
independently illustrated by one of the following heteroaryl groups:
Figure imgf000051_0001
wherein each of the above said heteroaryl groups is optionally substituted.
In yet another embodiment, the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein G and J are each
independently optionally substituted imidazolyl or benzimidazolyl.
In yet another embodiment, the present invention relates to compounds of F rmulae (3-Ia-l ~ 3-Ia-4), or a pharmaceutically acceptable salt thereof:
Figure imgf000051_0002
wherein Ring A, Ring B, G, J, L, U, W, X, Y, R1, R2, R3, R4, R5, R6, R9, Rla, Rlb, R7a and R9a are as previously defined in Formula (3-1).
In yet another embodiment, the present invention relates to compounds of F rmulae (3-Ia-5 ~ 3-Ia-6), and pharmaceutically acceptable salts thereof:
Figure imgf000051_0003
wherein Ring A, Ring B, G, J, L, R1, R6, R9, Rla, Rlb, and R9a are as previously defined in Formula (3-1).
In yet another embodiment, the present invention relates to compounds of Formula (3-Ia-7), and pharmaceutically acceptable salts thereof:
Figure imgf000052_0001
wherein Ring A, Ring B, G, J, L, R1, R6, R9, Rla, Rlb, and R9a are as previously defined in Formula (3-1).
In yet another embodiment, the present invention relates to compounds of Formula (3-Ia-7), and pharmaceutically acceptable salts thereof; wherein R6 at each occurrence is independently selected from the group consisting of C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted.
In still another embodiment, the present invention relates to compounds of Formula (3-Ia-7), and pharmaceutically acceptable salts thereof; wherein R6 at each occurrence is independently C1-C8 alkyl optionally substituted with amino, hydoxy, protected amino, or 0(C1-C4 alkyl).
In still another embodiment, the present invention relates to compounds of Formulae 3-Ib-l ~ 3-Ib-6), and pharmaceutically acceptable salts thereof:
Figure imgf000052_0002
wherein G, J, Q, R1, R6, R9, Rla, Rlb, and R9a are as previously defined in Formula (3-1); in Formula (3-Ib-l), Ring A, Ring B, and L are each present and as previously defined; in Formula (3-Ib-2), Ring A and Ring B are each present and as previously defined in Formula (3-1); in Formula (3-Ib-3), Ring B and L are each present and as previously defined in Formula (3-1); in Formula (3-Ib-4), Ring A and L are each present and as previously defined in Formula (3-1); in Formula (3-Ib-5), Ring B is present and as previously defined in Formula (3-1); and in Formula (3-Ib-6), L is present and as previously defined in Formula (3-1).
In still another embodiment, the present invention relates to compounds of Formulae (3-Ib-l ~ 3-Ib-6), and pharmaceutically acceptable salts thereof; wherein R6 is independently C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C1-C4 alkyl). In still another embodiment, the present invention relates to compounds of Formula (3-Ib-l), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and G and J are each independently optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of Formula (3-Ib-2), and pharmaceutically acceptable salts thereof; wherein one of Ring A and Ring B is optionally substituted phenyl or optionally substituted monocyclic heteroaryl, the other of Ring A and Ring B is optionally substituted bicyclic aryl or heteroaryl; and G and J are each independently optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of Formula (3-Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted bicyclic aryl or bicyclic heteroaryl; and G and J are each independently optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of Formula (3-Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; and G and J are each independently optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of
Formulae (3-Ib-3 ~ 3-Ib-4), and pharmaceutically acceptable salts thereof; wherein Ring B or Ring A is optionally substituted bicyclic aryl or bicyclic heteroaryl; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and G and J are each independently optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of
Formulae (3-Ib-3 ~ 3-Ib-4), and pharmaceutically acceptable salts thereof; wherein Ring B or Ring A is optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and G and J are each independently optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of
Formula (3-Ib-5), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted polycyclic aryl or polycyclic heteroaryl; and G and J are each independently optionally substituted imidazolyl or benzimidazolyl. In still another embodiment, the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein two geminal R7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C3-C8 cycloalkyl, C3-C8 cycloalkenyl and 3- to 8-membered heterocyclic, each optionally substituted.
In still another embodiment, the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein two geminal R7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted cyclopropyl or a spiro, optionally substituted 5- to 6- membered heterocyclic.
In yet another embodiment, the present invention relates to compounds of Formula (3-1), and pharmaceutically acceptable salts thereof; wherein R7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C1-C4 alkyl), S(C1-C4 alkyl), amino optionally substituted with one or two C1-C4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C1- C4 alkyl; preferably hydrogen, halogen or hydroxy.
In still another embodiment, the present invention relates to com ounds of
Formula (3-1), and pharmaceutically acceptable salts thereof; wherein
Figure imgf000054_0001
at each occurence is independently illustrated by one of the following groups:
Figure imgf000055_0001
Representative compounds of the present invention are those selected from compounds 3-1 to 3-347compiled in the following tables and/or shown below:
Figure imgf000055_0002
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
It is intended that the definition of any substituent or variable (e.g., R , R , etc.) at a particular location in a molecule be independent of its definitions elsewhere in that molecule. For example, when U is C(R7)2, each of the two R7 groups may be the same or different. It is also intended that the drawn formula of any linker or spacer (e.g., -NH- C(O)-, -OC(0)-NH-, etc.) at a particular location in a molecule be independent of its directions of attachment, unless otherwise indicated. For example, when a linker is -NH- C(O)-, it is meant to include -NH-C(O)- and -C(0)-NH- for attachments.
IV. Compounds having Formula (4-1)
In certain aspects, the present invention relates to compounds of Formula (4-1) as illustrated above, and pharmaceutically acceptable salts thereof.
In still another embodiment, the present invention relates to compounds of Formula (4-1), and pharmaceutically acceptable salts thereof; wherein two geminal R7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C3-C8 cycloalkyl, C3-C8 cycloalkenyl and 3- to 8-membered heterocyclic, each optionally substituted.
In still another embodiment, the present invention relates to compounds of Formula (4-1), and pharmaceutically acceptable salts thereof; wherein two geminal R7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted cyclopropyl or a spiro, optionally substituted 5- to 6- membered heterocyclic.
In another embodiment, the present invention relates to com ounds of Formula (4-
I), and pharmaceutically acceptable salts thereof; wherein
Figure imgf000066_0001
at each occurence is independently illustrated by one of the following groups:
Figure imgf000067_0001
each of which can be optionally substituted.
In yet another embodiment, Rla and R9a, Rla and R9, Rld and R9a, or Rld and R9 are taken together with the carbon atom to which they are attached to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic.
In a further embodiment Rlb and R9a, or Rlb and R9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic.
In yet another embodiment, the present invention relates to compounds of Formula
(4-1), and pharmaceutically acceptable salts thereof; wherein the moiety of
Figure imgf000067_0002
is illustrated by one of the following core groups:
Figure imgf000067_0003
wherein R6 is as previously defined in Formula (4-1) and each of the above core groups is optionally substituted. In an additional embodiment, the invention relates to compounds of Formula (4-1) and pharmaceutically acceptable salts thereof; wherein R^ is independently an optionally substituted C1-C8 alkyl.
In a further embodiment, the invention relates to compounds of Formula (4-1) and pharmaceutically acceptable salts thereof; wherein -C(0)R6 is an a-amino acid residue.
In yet another embodiment, the present invention relates to compounds of Formula (4-1), and pharmaceutically acceptable salts thereof; wherein G is an optionally substituted five-membered heteroaryl containing one or more nitrogen atoms, and is C-attached.
In yet another embodiment, the present invention relates to compounds of Formula (4-1), and pharmaceutically acceptable salts thereof; wherein G is an optionally substituted 5/6-membered fused heteroaryl; wherein the 5-membered ring of said 5/6-membered fused heteroaryl is a heteroaryl containing one or more nitrogen atoms and wherein the 5- membered ring is C-attached to group Q, and wherein the 6-membered ring of said 5/6- membered fused heteroaryl is aryl or heteroaryl and is C-attached to one of Ring A, L and Ring B.
In yet another embodiment, the present invention relates to compounds of Formula (4-1), and pharmaceutically acceptable salts thereof; wherein G is illustrated by one of the following heteroaryl groups:
Figure imgf000068_0001
wherein each of the above said heteroaryl groups is optionally substituted.
In yet another embodiment, the present invention relates to compounds of Formula (4-1), and pharmaceutically acceptable salts thereof; wherein G is optionally substituted imidazolyl or benzimidazolyl.
In yet another embodiment, the present invention relates to compounds of
Formulae (4-Ia-l ~ 4-Ia-4), and pharmaceutically acceptable salts thereof:
Figure imgf000069_0001
wherein Ring A, Ring B, G, J, L, U, W, X, Y, R1, R2, R3, R4, R5, R6, R9, Rla, Rlb, Rld, R7a, R7b and R9a are as previously defined in Formula (4-1).
In yet another embodiment, the present invention relates to compounds of
Formulae (4-Ia-5 ~ 4-Ia-8), and pharmaceutically acceptable salts thereof:
Figure imgf000069_0002
wherein Ring A, Ring B, G, J, L, U, W, X, Y, R1, R2, R3, R4, R5, R6, R9, Rla, Rlb, Rlc, Rld, R7a, R7b and R9a are as previously defined in Formula (4-1).
In yet another embodiment, the present invention relates to compounds of
Formulae (4-Ia-9 ~ 4-Ia-12), and pharmaceutically acceptable salts thereof;
Figure imgf000069_0003
wherein Ring A, Ring B, G, L, R6, R7a and R7b are as previously defined in Formula (4-1).
In yet another embodiment, the present invention relates to compounds of
Formulae (4-Ia-13 ~ 4-Ia-16), and pharmaceutically acceptable salts thereof:
Figure imgf000070_0001
wherein Ring A, Ring B, G, L, R6, R7a and R7b are as previously defined in Formula (4-1).
In yet another embodiment, the present invention relates to compounds of
Formulae (4-Ia-13 ~ 4-Ia-16), and pharmaceutically acceptable salts thereof; wherein R6 at each occurrence is independently selected from the group consisting of C1-C8 alkyl, C2- C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted.
In still another embodiment, the present invention relates to compounds of Formula (4-Ia-13 ~ 4-Ia-16), and pharmaceutically acceptable salts thereof; wherein R6 at each occurrence is independently C1-C8 alkyl optionally substituted with amino, hydoxy, protected amino, or 0(C1-C4 alkyl).
In still another embodiment, the present invention relates to compounds of Formulae (4-Ib- ~ 4-Ib-4), and pharmaceutically acceptable salts thereof;
Figure imgf000070_0002
wherein Ring B, G, Q, U, R1, R6, R7a and R7b are as previously defined in Formula (4-1); in Formula (4-Ib-l), A and L are each present and as previously defined; in Formula (4-1). in Formula (4-Ib-2), Ring A is present and as previously defined in Formula (4-1); and in Formula (4-Ib-3), L is present and as previously defined in Formula (4-1).
In still another embodiment, the present invention relates to compounds of Formula (4-Ib-l ~ 4-Ib-4), and pharmaceutically acceptable salts thereof; wherein R6 is independently C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C1-C4 alkyl). In still another embodiment, the present invention relates to compounds of Formula (4-Ib-l), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of Formula (4-Ib-2), and pharmaceutically acceptable salts thereof; wherein one of Ring A and Ring B is optionally substituted phenyl or optionally substituted monocyclic heteroaryl, the other of Ring A and Ring B is optionally substituted bicyclic aryl or heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of Formula (4-Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted bicyclic aryl or heteroaryl; and G is optionally substituted imidazolyl.
In still another embodiment, the present invention relates to compounds of
Formula (4-Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; and G is optionally substituted benzimidazolyl.
In still another embodiment, the present invention relates to compounds of Formula (4-Ib-3), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted bicyclic aryl or heteroaryl; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of Formula (4-Ib-3), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of Formula (4-Ib-4), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted polycyclic aryl or heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
In yet another aspect, the present invention relates to compounds of Formula (4-Ib- 4), and pharmaceutically acceptable salts thereof; wherein R7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C1-C4 alkyl), S(C1-C4 alkyl), amino optionally substituted with one or two C1-C4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C1- C4 alkyl; preferably hydrogen, halogen or hydroxyl.
Representative compounds of the present invention are those selected from compounds 4-1 to 4-348 compiled in the following tables and/or are shown below:
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
It will be appreciated that the description of the present invention herein should be construed in congruity with the laws and principals of chemical bonding. In some instances it may be necessary to remove a hydrogen atom in order to accommodate a substitutent at any given location.
It is intended that the definition of any substituent or variable (e.g., R3, R7, etc.) at a particular location in a molecule be independent of its definitions elsewhere in that molecule. For example, when U is C(R7)2, each of the two R7 groups may be the same or different.
It is also intended that the drawn formula of any linker or spacer (e.g., -NH- C(O)-, -OC(0)-NH-, etc.) at a particular location in a molecule be independent of its directions of attachment, unless otherwise indicated. For example, when a linker is -NH- C(O)-, it is meant to include -NH-C(O)- and -C(0)-NH- for attachments. V. Compounds having Formula (5-1)
In certain aspects, the present invention relates to compounds of Formula (5-1) as illustrated above, and pharmaceutically acceptable salts thereof.
In yet another embodiment, the present invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; wherein Rla and R9a, Rla and R9, R1 and R9a, or R1 and R9 are taken together to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic.
In a further embodiment, the present invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; wherein Rlb and R9a, or Rlb and R9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic.
In another embodiment, the present invention relates to compounds of Formula (5-
I), and pharmaceutically acceptable salts thereof; wherein the moiety of
Figure imgf000082_0002
is illustrated by one of the following core groups:
Figure imgf000082_0001
wherein R6 is as previously defined and each of the above core groups is optionally substituted.
In an additional embodiment, the invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; wherein R^ is independently an optionally substituted C1-C8 alkyl.
In a further embodiment, the invention relates to compounds of Formula (5-1) and pharmaceutically acceptable salts thereof; wherein -C(0)R6 is an a-amino acid residue.
In yet another embodiment, the present invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; G is an optionally substituted five- membered heteroaryl containing one or more nitrogen atoms, and is C-attached. In yet another embodiment, the present invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; wherein G is an optionally substituted 5/6-membered fused heteroaryl; wherein the 5-membered ring of said 5/6-membered fused heteroaryl is a heteroaryl containing one or more nitrogen and wherein the 5-membered ring is C-attached to group Q, and wherein the 6-membered ring of said 5/6-membered fused heteroaryl is aryl or heteroaryl and is C-attached to one of Ring A, L and Ring B.
In yet another embodiment, the present invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; wherein G is illustrated by one of the following heteroaryl groups:
Figure imgf000083_0001
wherein each of the above said heteroaryl groups is optionally substituted.
In yet another embodiment, the present invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; wherein G is optionally substituted imidazolyl or benzimidazolyl.
In yet another embodiment, the present invention relates to compounds of
Form lae (5-Ia-l ~ 5-Ia-4), and pharmaceutically acceptable salts thereof;
Figure imgf000083_0002
wherein Ring A, Ring B, G, L, U, W, X, Y, R1, R2, R3, R4, R5, R6, R9, Rla, Rlb, Rlc, and R9a are as previously defined in Formula (5-1).
In yet another embodiment, the present invention relates to compounds of
Formulae (5-Ia-5 ~ 5-Ia-7), and pharmaceutically acceptable salts thereof;
Figure imgf000084_0001
wherein Ring A, Ring B, G, L, U, W, X, Y, R1, R2, R3, R4, R5, R6, R9, Rla, Rlb, Rlc, and R9a are as previously defined in Formula (5-1).
In yet another embodiment, the present invention relates to compounds of Formulae (5-Ia-8 ~ 5-Ia-9), and pharmaceutically acceptable salts thereof;
Figure imgf000084_0002
wherein Ring A, Ring B, G, L, R1, R6, R9, Rla, Rlb, Rlc, and R9a are as previously defined in Formula (5-1).
In yet another embodiment, the present invention relates to compounds of Formula (5-Ia-ll), and pharmaceutically acceptable salts thereof;
Figure imgf000084_0003
wherein Ring A, Ring B, G, L, R1, R6, R9, Rla, Rlb, Rlc, and R9a are as previously defined in Formula (5-1).
In yet another embodiment, the present invention relates to compounds of Formula (5-Ia-ll), and pharmaceutically acceptable salts thereof; wherein R6 at each occurrence is independently selected from the group consisting of C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted.
In still another embodiment, the present invention relates to compounds of Formula (5-Ia-ll), and pharmaceutically acceptable salts thereof; wherein R6 at each occurrence is independently C1-C8 alkyl optionally substituted with amino, hydoxy, protected amino, or 0(C1-C4 alkyl).
In still another embodiment, the present invention relates to compounds of Formulae (5-Ib-l ~ 5-Ib-4), and pharmaceutically acceptable salts thereof;
Figure imgf000085_0001
wherein Ring B, G, Q, R1, R6, R9, Rla, Rlb, Rlc, and R9a are as previously defined in Formula (5-1); in Formula (5-Ib-l), Ring A and L are each present and as previously defined in Formula (5-1); in Formula (5-Ib-2), Ring A is present and as previously defined in Formula (5-1); and in Formula (5-Ib-3), L is present and as previously defined in Formula (5-1).
In still another embodiment, the present invention relates to compounds of Formulae (5-Ib-l ~ 5-Ib-4), and pharmaceutically acceptable salts thereof; wherein R6 at each occurrence is independently C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino or 0(C1-C4 alkyl).
In still another embodiment, the present invention relates to compounds of Formula (5-Ib-l), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of Formula (5-Ib-2), and pharmaceutically acceptable salts thereof; wherein one of Ring A and Rng B is optionally substituted phenyl or optionally substituted monocyclic heteroaryl, the other of Ring A and Ring B is optionally substituted bicyclic aryl or heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of Formula (5-Ib-2), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted bicyclic aryl or heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of Formula (5-Ib-2) and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl. In still another embodiment, the present invention relates to compounds of Formula (5-Ib-3), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted bicyclic aryl or heteroaryl; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of Formula (5-Ib-3), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and G is optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of Formula (5-Ib-4), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted polycyclic aryl or heteroaryl; and G is optionally substituted imidazolyl or benzimidazolyl.
In still another embodiment, the present invention relates to compounds of Formulae (5-Ib-5 ~ 5-Ib-8), and pharmaceutically acceptable salts thereof:
Figure imgf000086_0001
wherein Ring B, Q, V, R1, R6, R9, Rla, Rlb, Rlc, and R9a are as previously defined in Formula (5-1); in Formula (5-Ib-5), Ring A and L are each present and as previously defined in Formula (5-1); in Formula (5-Ib-6), Ring A is present and as previously defined in Formula (5-1); and in Formula (5-Ib-7), L is present and as previously defined in Formula (5-1).
In still another embodiment, the present invention relates to compounds of Formula (5-Ib-5), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl.
In still another embodiment, the present invention relates to compounds of Formula (5-Ib-6), and pharmaceutically acceptable salts thereof; wherein one of Ring A and Ring B is optionally substituted phenyl or optionally substituted monocyclic heteroaryl, the other of Ring A and Ring B is optionally substituted bicyclic aryl or heteroaryl.
In still another embodiment, the present invention relates to compounds of Formula (5-Ib-6), and pharmaceutically acceptable salts thereof; wherein Ring A and Ring B are each independently optionally substituted phenyl or monocyclic heteroaryl.
In still another embodiment, the present invention relates to compounds of Formula (5-Ib-7), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted bicyclic aryl or heteroaryl; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl.
In still another embodiment, the present invention relates to compounds of
Formula (5-Ib-7), and pharmaceutically acceptable salts thereof; wherein Ring B is optionally substituted phenyl or monocyclic heteroaryl; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl.
In still another embodiment, the present invention relates to compounds of Formula (5-Ib-8), or a pharmaceutically acceptable salt thereof; wherein Ring B is optionally substituted polycyclic aryl or heteroaryl.
In still another embodiment, the present invention relates to compounds of Formula (5-1), and pharmaceutically acceptable salts thereof; wherein two geminal R7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C3-C8 cycloalkyl, C3-C8 cycloalkenyl and 3- to 8-membered heterocyclic, each optionally substituted.
In still another embodiment, the present invention relates to compounds of
Formula (5-1), and pharmaceutically acceptable salts thereof; wherein two geminal R7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted cyclopropyl or a spiro, optionally substituted 5- to 6- membered heterocyclic.
In yet another embodiment, the present invention relates to compounds of Formula
(5-1), and pharmaceutically acceptable salts thereof; wherein R7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy,
0(C1-C4 alkyl), S(C1-C4 alkyl), amino optionally substituted with one or two C1-C4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C1-
C4 alkyl; preferably hydrogen, halogen or hydroxy. In still another embodiment, the present invention relates to com ounds of
Formula (5-1), and pharmaceutically acceptable salts thereof; wherein
Figure imgf000088_0001
each occurence is independently illustrated by one of the following groups:
Figure imgf000088_0002
Representative compounds of the present invention are those selected from compounds 5-1 to 5-394 compiled in the following tables and/or shown below:
Figure imgf000088_0003
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
It will be appreciated that the description of the present invention herein should construed in congruity with the laws and principals of chemical bonding. In some instances it may be necessary to remove a hydrogen atom in order to accommodate a substitutent at any given location.
It is intended that the definition of any substituent or variable (e.g., R3, R7, etc.) at a particular location in a molecule be independent of its definitions elsewhere in that molecule. For example, when U is C(R7)2, each of the two R7 groups may be the same or different.
It is also intended that the drawn formula of any linker or spacer (e.g., -NH- C(O)-, -OC(0)-NH-, etc.) at a particular location in a molecule be independent of its directions of attachment, unless otherwise indicated. For example, when a linker is -NH- C(O)-, it is meant to include -NH-C(O)- and -C(0)-NH- for attachments.
It will be appreciated that the description of the present invention herein should be construed in congruity with the laws and principals of chemical bonding. In some instances it may be necessary to remove a hydrogen atom in order to accommodate a substitutent at any given location. It will be yet appreciated that the compounds of the present invention may contain one or more asymmetric carbon atoms and may exist in racemic, diastereoisomeric, and optically active forms. It will still be appreciated that certain compounds of the present invention may exist in different tautomeric forms. All tautomers are contemplated to be within the scope of the present invention.
It should be understood that in some embodiments, the compounds encompassed by the present invention are those that are suitably stable for use as pharmaceutical agent.
It will be further appreciated that reference herein to therapy and/or treatment includes, but is not limited to, prevention, retardation, prophylaxis, therapy and/or cure of the disease. It will further be appreciated that references herein to treatment or prophylaxis of HCV infection includes treatment or prophylaxis of HCV-associated disease such as liver fibrosis, cirrhosis and hepatocellular carcinoma.
A further embodiment of the present invention includes pharmaceutical compositions comprising any single compound a combination of two or more compounds delineated herein, or a pharmaceutically acceptable salt thereof, with a pharmaceutically acceptable carrier or excipient.
Yet a further embodiment of the present invention is a pharmaceutical composition comprising any single compound or a combination of two or more compounds delineated herein, or a pharmaceutically acceptable salt thereof, in combination with one or more agents known in the art, with a pharmaceutically acceptable carrier or excipient.
It will be further appreciated that 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 present invention include therapies for disease caused by HCV infection that suppresses HCV viral replication by direct or indirect mechanisms. These agents include, but are not limited to, host immune modulators (for example, interferon-alpha, pegylated interferon-alpha, consensus interferon, interferon-beta, interferon-gamma, CpG oligonucleotides and the like); antiviral compounds that inhibit host cellular functions such as inosine monophosphate dehydrogenase (for example, ribavirin and the like); cytokines that modulate immune function (for example, interleukin 2, interleukin 6, and interleukin 12); a compound that enhances the development of type 1 helper T cell response; interfering R A; anti-sense R A; vaccines comprising HCV antigens or antigen adjuvant combinations directed against HCV; 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; and any agent or combination of agents that inhibit the replication of HCV by targeting other proteins of the viral genome involved in the viral replication and/or interfere with the function of other viral targets, such as inhibitors of NS3/NS4A protease, NS3 helicase, NS5B polymerase, NS4A protein and NS5 A protein.
According to yet another embodiment, the pharmaceutical compositions of the present invention may further comprise other inhibitor(s) of targets in the HCV life cycle, including, but not limited to, helicase, polymerase, metalloprotease, NS4A protein, NS5A protein, and internal ribosome entry site (IRES).
Accordingly, one embodiment of the present 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 or more antiviral agents, or a combination thereof, with a therapeutically effective amount of a compound or combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof. Examples of the host immune modulator include, but are 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. An example of the RNA-containing virus includes, but is not limited to, hepatitis C virus (HCV).
A further embodiment of the present 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 present invention, or a pharmaceutically acceptable salt thereof. An example of the RNA-containing virus includes, but is not limited to, hepatitis C virus (HCV).
Yet another embodiment of the present 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 present invention, or a pharmaceutically acceptable salt 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. An example of the R A-containing virus includes, but is not limited to, hepatitis C virus (HCV).
A further embodiment of the present 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 present invention, or a pharmaceutically acceptable salt 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-114, 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. An example of the RNA- containing virus includes, but is not limited to, hepatitis C virus (HCV).
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.
In addition, the present invention provides the use of a compound or a combination of compounds of the invention, or a pharmaceutically acceptable salt thereof, and one or more agents selected from the group consisting of a host immune modulator and a second or more antiviral agents, 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 include, but are 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 present 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 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 thereof, 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 R A-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, a still further embodiment 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 thereof, and one or more agents as defined hereinabove, with 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 agents to be administered in combination with a compound of the present invention include a cytochrome P450 monooxygenase inhibitor (also referred to herein as a CYP inhibitor), which is expected to inhibit metabolism of the compounds of the invention. Therefore, the cytochrome P450 monooxygenase inhibitor would be in an amount effective to inhibit metabolism of the compounds of this invention. Accordingly, the CYP inhibitor is administered in an amount such that the bioavailiablity of the protease inhibitor is increased in comparison to the bioavailability in the absence of the CYP inhibitor.
In one embodiment, the invention provides methods for improving the
pharmacokinetics of compounds of the invention. The advantages of improving the pharmacokinetics of drugs are recognized in the art (see, for example, US Patent Pub. Nos. 2004/0091527; US 2004/0152625; and US 2004/0091527). Accordingly, one embodiment of this invention provides a method for administering an inhibitor of CYP3 A4 and a compound of the invention. Another embodiment of this invention provides a method for administering a compound of the invention and an inhibitor of isozyme 3A4 ("CYP3A4"), isozyme 2C19 ("CYP2C19"), isozyme 2D6 ("CYP2D6"), isozyme 1 A2 ("CYP1 A2"), isozyme 2C9 ("CYP2C9"), or isozyme 2E1 ("CYP2E1"). In a preferred embodiment, the CYP inhibitor preferably inhibits CYP3A4. Any CYP inhibitor that improves the pharmacokinetics of the relevant NS3/4A protease may be used in a method of this invention. These CYP inhibitors include, but are not limited to, ritonavir (see, for example, WO 94/14436), ketoconazole, troleandomycin, 4-methyl pyrazole, cyclosporin, clomethiazole, cimetidine, itraconazole, fluconazole, miconazole, fluvoxamine, fluoxetine, nefazodone, sertraline, indinavir, nelfmavir, amprenavir, fosamprenavir, saquinavir, lopinavir, delavirdine, erythromycin, VX-944, and VX-497. Preferred CYP inhibitors include ritonavir, ketoconazole, troleandomycin, 4-methyl pyrazole, cyclosporin, and clomethiazole.
It will be understood that the administration of the combination of the invention by means of a single patient pack, or patient packs of each formulation, containing within a package insert instructing the patient to the correct use of the invention is a desirable additional feature of this invention.
According to a further aspect of the invention is a pack comprising at least a compound of the invention and a CYP inhibitor of the invention and an information insert containing directions on the use of the combination of the invention. In an alternative embodiment of this invention, the pharmaceutical pack further comprises one or more of additional agent as described herein. The additional agent or agents may be provided in the same pack or in separate packs.
Another aspect of this involves a packaged kit for a patient to use in the treatment of HCV infection or in the prevention of HCV infection, comprising: a single or a plurality of pharmaceutical formulation of each pharmaceutical component; a container housing the pharmaceutical formulation(s) during storage and prior to administration; and instructions for carrying out drug administration in a manner effective to treat or prevent HCV infection.
Accordingly, this invention provides kits for the simultaneous or sequential administration of a compound of the invention and a CYP inhibitor (and optionally an additional agent) or derivatives thereof are prepared in a conventional manner. Typically, such a kit will comprise, e.g. a composition of each inhibitor and optionally the additional agent(s) in a pharmaceutically acceptable carrier (and in one or in a plurality of pharmaceutical formulations) and written instructions for the simultaneous or sequential administration.
In another embodiment, a packaged kit is provided that contains one or more dosage forms for self administration; a container means, preferably sealed, for housing the dosage forms during storage and prior to use; and instructions for a patient to carry out drug administration. The instructions will typically be written instructions on a package insert, a label, and/or on other components of the kit, and the dosage form or forms are as described herein. Each dosage form may be individually housed, as in a sheet of a metal foil- plastic laminate with each dosage form isolated from the others in individual cells or bubbles, or the dosage forms may be housed in a single container, as in a plastic bottle. The present kits will also typically include means for packaging the individual kit components, i. e., the dosage forms, the container means, and the written instructions for use. Such packaging means may take the form of a cardboard or paper box, a plastic or foil pouch, etc.
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 "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, cortico-steroids, colony-stimulating factors, chemotactic factors, etc.
The term "aryl," as used herein, refers to a mono- or polycyclic carbocyclic ring system including, but not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, and idenyl. A polycyclic aryl is a polycyclic ring system that comprises at least one aromatic ring. Polycyclic aryls can comprise fused rings, covalently attached rings or a
combination thereof.
The term "heteroaryl," as used herein, refers to a mono- or polycyclic ring system comprising at least one aromatic ring having one or more ring atom selected from S, O and N; and the remaining ring atoms are carbon, 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, and quinoxalinyl. A polycyclic heteroaryl can comprise fused rings, covalently attached rings or a combination thereof.
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.
The term "bicyclic aryl" or "bicyclic heteroaryl" refers to a ring system consisting of two rings wherein at least one ring is aromatic; and they can be fused or covalently attached.
The term "tricyclic aryl" or "tricyclic heteroaryl" refers to a ring system consisting of three rings wherein at least one ring is aromatic.
The terms "C1-C4 alkyl," "C1-C6 alkyl," "C1-C8 alkyl," "C2-C4 alkyl," or "C3-C6 alkyl," as used herein, refer to saturated, straight- or branched-chain hydrocarbon radicals containing between one and four, one and six, one and eight carbon atoms, or the like, respectively. Examples of C1-Cs alkyl radicals include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, tert-butyl, neopentyl, n-hexyl, heptyl and octyl radicals.
The terms "C2-C8 alkenyl," "C2-C4 alkenyl," "C3-C4 alkenyl," or "C3-C6 alkenyl," as used herein, refer to straight- or branched-chain hydrocarbon radicals containing from two to eight, or two to four carbon atoms, or the like, having at least one carbon-carbon double bond by the removal of a single hydrogen atom. Alkenyl groups include, but are not limited to, for example, ethenyl, propenyl, butenyl, l-methyl-2-buten-1-yl, heptenyl, octenyl, and the like.
The terms "C2-C8 alkynyl," "C2-C4 alkynyl," "C3-C4 alkynyl," or "C3-C6 alkynyl," as used herein, refer to straight- or branched-chain hydrocarbon radicals containing from two to eight, or two to four carbon atoms, or the like, having at least one carbon-carbon triple bond by the removal of a single hydrogen atom. Representative alkynyl groups include, but are not limited to, for example, ethynyl, 1-propynyl, 1-butynyl, heptynyl, octynyl, and the like.
The term "C3-C8-cycloalkyl", or "Cs-Cy-cycloalkyl," as used herein, refers to a monocyclic or polycyclic saturated carbocyclic ring compound, and the carbon atoms may be optionally oxo-substituted. Examples of C3-C8-cycloalkyl include, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopentyl and cyclooctyl; and examples of Cs-Cycycloalkyl include, but not limited to, cyclopentyl, cyclohexyl, bicyclo [2.2.1] heptyl, and the like.
The term "C3-C8 cycloalkenyl" or "C5-C7 cycloalkenyl," as used herein, refers to monocyclic or polycyclic carbocyclic ring compound having at least one carbon-carbon double bond, and the carbon atoms may be optionally oxo-substituted. Examples of C3-C8 cycloalkenyl include, but not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, and the like; and examples of C5-C7 cycloalkenyl include, but not limited to, cyclopentenyl, cyclohexenyl, cycloheptenyl, and the like.
The term "arylalkyl," as used herein, refers to an aryl-substituted alkyl group. More preferred arylalkyl groups are aryl-C1-C6-alkyl groups.
The term "heteroarylalkyl," as used herein, refers to a heteroaryl-substituted alkyl group. More preferred heteroarylalkyl groups are heteroaryl-C1-C6-alkyl groups.
It is understood that any alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclic and cycloalkenyl moiety described herein can also be an aliphatic group or an alicyclic group.
An "aliphatic" group is a non-aromatic moiety comprised of any combination of carbon atoms, hydrogen atoms, halogen atoms, oxygen, nitrogen or other atoms, and optionally contains one or more units of unsaturation, e.g., double and/or triple bonds. Examples of aliphatic groups are functional groups, such as, O, OH, NH, NH2, C(O), S(0)2, C(0)0, C(0)NH, OC(0)0, OC(0)NH, OC(0)NH2, S(0)2NH, S(0)2NH2, NHC(0)NH2, NHC(0)C(0)NH, NHS(0)2NH, NHS(0)2NH2, C(0)NHS(0)2,
C(0)NHS(0)2NH or C(0)NHS(0)2NH2, and the like, groups comprising one or more functional groups, non-aromatic hydrocarbons (optionally substituted), and groups wherein one or more carbons of a non-aromatic hydrocarbon (optionally substituted) is replaced by a functional group. Carbon atoms of an aliphatic group can be optionally oxo- substituted. An aliphatic group may be straight chained, branched, cyclic, or a
combination thereof 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, as used herein, aliphatic groups expressly include, for example, alkoxyalkyls, polyalkoxyalkyls, such as polyalkylene glycols, polyamines, and
polyimines, for example. Aliphatic groups may be optionally substituted. A linear aliphatic group is a non-cyclic aliphatic group. It is to be understood that when an aliphatic group or a linear aliphatic group is said to "contain" or "include" or "comprise" one or more specified functional groups, the aliphatic group can be selected from one or more of the specified functional groups or a combination thereof, or a group wherein one or more carbons of a non-aromatic hydrocarbon (optionally substituted) is replaced by a specified functional group. In some examples, the linear aliphatic group can be represented by the formula M-V'-M', where M and M' are each independently absent or an alkyl, alkenyl or alkynyl, each optionally substituted, and V is a functional group. In some examples, V is selected from the group consisting of C(O), S(0)2, C(0)0,
C(0)N(Rn), OC(0)0, OC(0)N(Rn), S(0)2N(Rn), N(Rn)C(0)N(Rn),
N(Rn)C(0)C(0)N(Rn), N(Rn)S(0)2N(Rn), C(0)N(Rn)S(0)2 or
C(0)N(Rn)S(0)2N(Rn); wherein R11 is as previously defined. In another aspect of the invention, an exemplary linear aliphatic group is an alkyl, alkenyl or alkynyl, each optionally substituted, which is interrupted or terminated by a functional group such as described herein.
The term "alicyclic," as used herein, denotes a monovalent group derived from a monocyclic or bicyclic saturated carbocyclic ring compound by the removal of a single hydrogen atom, and the carbon atoms may be optionally oxo-substituted. 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 terms "heterocyclic" or "heterocycloalkyl" can be used interchangeably and referred to a non-aromatic ring or a bi- or tri-cyclic group fused system, where (i) each ring system contains at least one heteroatom independently selected from oxygen, sulfur and nitrogen, (ii) each ring system can be saturated or unsaturated, (iii) the nitrogen and sulfur heteroatoms may optionally be oxidized, (iv) the nitrogen heteroatom may optionally be quaternized, (v) any of the above rings may be fused to an aromatic ring, and (vi) the remaining ring atoms are carbon atoms which may be optionally oxo-substituted. Representative heterocycloalkyl groups include, but are not limited to, 1,3-dioxolane, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, quinoxalinyl, pyridazinonyl, and tetrahydrofuryl. Such heterocyclic groups may be further substituted. Heteroaryl or heterocyclic groups can be C-attached or N-attached (where possible).
It is understood that any alkyl, alkenyl, alkynyl, alicyclic, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclic, aliphatic moiety or the like, described herein can also be a divalent or multivalent group when used as a linkage to connect two or more groups or substituents, which can be at the same or different atom(s).
The term "substituted" when used with alkyl, alkenyl, alkynyl, alicyclic, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclic, or aliphatic as described herein refers to substitution by independent replacement of one, two, or three or more of the hydrogen atoms thereon with substituents including, but not limited to, -F, -CI, -Br, -I, - OH, protected hydroxy, -N02, -N3, -CN, -NH2, protected amino, oxo, thioxo, -NH-C1-C12- alkyl, -NH-C2-C8-alkenyl, -NH-C2-C8-alkynyl, -NH-C3-C12-cycloalkyl, -NH-aryl, -NH- heteroaryl, -NH-heterocycloalkyl, -dialkylamino, -diarylamino, -diheteroarylamino, -O- C1-C12-alkyl, -0-C2-C8-alkenyl, -0-C2-C8-alkynyl, -0-C3-C12-cycloalkyl, -O-aryl, -O- heteroaryl, -O-heterocycloalkyl, -C(0)-C1-C12-alkyl, -C(0)-C2-C8-alkenyl, -C(0)-C2-C8- alkynyl, -C(0)-C3-C12-cycloalkyl, -C(0)-aryl, -C(0)-heteroaryl, -C(0)-heterocycloalkyl, - CONH2, -CONH-C1-C12-alkyl, -CONH-C2-C8-alkenyl, -CONH-C2-C8-alkynyl, -CONH- C3-C12-cycloalkyl, -CONH-aryl, -CONH-heteroaryl, -CONH-heterocycloalkyl, -OC02-d- C12-alkyl, -OC02-C2-C8-alkenyl, -OC02-C2-C8-alkynyl, -OC02-C3-C12-cycloalkyl, - OC02-aryl, -OC02-heteroaryl, -OC02-heterocycloalkyl, -C02-C1-C12 alkyl, -C02-C2-C8 alkenyl, -C02-C2-C8 alkynyl, C02-C3-C12-cycloalkyl, -C02- aryl, C02-heteroaryl, C02- heterocyloalkyl, -OCONH2, -OCONH-C1-C12-alkyl, -OCONH-C2-C8-alkenyl, -OCONH- C2-C8-alkynyl, -OCONH-C3-C12-cycloalkyl, -OCONH-aryl, -OCONH-heteroaryl, - OCONH- heterocycloalkyl, -NHC(0)H, -NHC(0)-C1-C12-alkyl, -NHC(0)-C2-C8-alkenyl, -NHC(0)-C2-C8-alkynyl, -NHC(0)-C3-C12-cycloalkyl, -NHC(0)-aryl, -NHC(O)- heteroaryl, -NHC(0)-heterocycloalkyl, -NHC02-C1-C12-alkyl, -NHC02-C2-C8-alkenyl, - NHCO2- C2-C8-alkynyl, -NHC02-C3-C12-cycloalkyl, -NHC02-aryl, -NHC02-heteroaryl, - NHC02- heterocycloalkyl, -NHC(0)NH2, -NHC(0)NH-C1-C12-alkyl, -NHC(0)NH-C2-C8- alkenyl, -NHC(0)NH-C2-C8-alkynyl, -NHC(0)NH-C3-C12-cycloalkyl, -NHC(0)NH-aryl, -NHC(0)NH-heteroaryl, -NHC(0)NH-heterocycloalkyl, NHC(S)NH2, -NHC(S)NH-C C12-alkyl, -NHC(S)NH-C2-C8-alkenyl, -NHC(S)NH-C2-C8-alkynyl, -NHC(S)NH-C3-C12- cycloalkyl, -NHC(S)NH-aryl, -NHC(S)NH-heteroaryl, -NHC(S)NH-heterocycloalkyl, - NHC(NH)NH2, -NHC(NH)NH-C1-C12-alkyl, -NHC(NH)NH-C2-C8-alkenyl, - NHC(NH)NH-C2-C8-alkynyl, -NHC(NH)NH-C3-C12-cycloalkyl, -NHC(NH)NH-aryl, - NHC(NH)NH-heteroaryl, -NHC(NH)NH-heterocycloalkyl, -NHC(NH)-C1-C12-alkyl, - NHC(NH)-C2-C8-alkenyl, -NHC(NH)-C2-C8-alkynyl, -NHC(NH)-C3-C12-cycloalkyl, -
NHC(NH)-aryl, -NHC(NH)-heteroaryl, -NHC(NH)-heterocycloalkyl, -C(NH)NH-C1-C12- alkyl, -C(NH)NH-C2-C8-alkenyl, -C(NH)NH-C2-C8-alkynyl, -C(NH)NH-C3-C12- cycloalkyl, -C(NH)NH-aryl, -C(NH)NH-heteroaryl, -C(NH)NH-heterocycloalkyl, -S(O)- C1-C12-alkyl, -S(0)-C2-C8-alkenyl, - S(0)-C2-C8-alkynyl, -S(0)-C3-C12-cycloalkyl, -S(O)- aryl, -S(0)-heteroaryl, -S(0)-heterocycloalkyl, -S02NH2, -S02NH-C1-C12-alkyl, -S02NH- C2-C8-alkenyl, -S02NH- C2-C8-alkynyl, -S02NH-C3-C12-cycloalkyl, -S02NH-aryl, - S02NH-heteroaryl, -S02NH- heterocycloalkyl, -NHS02-C1-C12-alkyl, -NHS02-C2-C8- alkenyl, - NHS02-C2-C8-alkynyl, -NHS02-C3-C12-cycloalkyl, -NHS02-aryl, -NHS02- heteroaryl, -NHS02-heterocycloalkyl, -CH2NH2, -CH2S02CH3, -aryl, -arylalkyl, - heteroaryl, -heteroarylalkyl, -heterocycloalkyl, -C3-C12-cycloalkyl, polyalkoxyalkyl, polyalkoxy, -methoxymethoxy, -methoxyethoxy, -SH, -S-C1-C12-alkyl, -S-C2-C8-alkenyl, - S-C2-C8-alkynyl, -S-C3-C12-cycloalkyl, -S-aryl, -S-heteroaryl, -S-heterocycloalkyl, or methylthiomethyl. It is understood that the aryls, heteroaryls, alkyls, and the like can be further substituted.
The term "halogen," as used herein, refers to an atom selected from fluorine, chlorine, bromine and iodine.
The term "hydrogen" includes hydrogen and deuterium. In addition, the recitation of an atom includes other isotopes of that atom so long as the resulting compound is pharmaceutically acceptable.
The term "hydroxy activating group," as used herein, refers to a labile chemical moiety which is known in the art to activate a hydroxyl group so that it will depart during synthetic procedures such as in a substitution or an elimination reaction. Examples of hydroxyl activating group include, but not limited to, mesylate, tosylate, triflate, p- nitrobenzoate, phosphonate and the like. The term "activated hydroxyl," as used herein, refers to a hydroxy group activated with a hydroxyl activating group, as defined above, including mesylate, tosylate, triflate, p-nitrobenzoate, phosphonate groups, for example.
The term "hydroxy protecting group," as used herein, refers to a labile chemical moiety which is known in the art to protect a hydroxyl 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 art 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 hydroxyl protecting groups include benzyloxycarbonyl, 4- methoxybenzyloxycarbonyl, tert-butoxycarbonyl, isopropoxycarbonyl,
diphenylmethoxycarbonyl, 2,2,2-trichloroethoxycarbonyl, allyloxycarbonyl, acetyl, formyl, chloroacetyl, trifluoroacetyl, methoxyacetyl, phenoxyacetyl, benzoyl, methyl, t- butyl, 2,2,2-trichloroethyl, 2-trimethylsilyl ethyl, allyl, benzyl, triphenylmethyl (trityl), methoxymethyl, methylthiomethyl, benzyloxymethyl, 2-(trimethylsilyl)ethoxymethyl, methanesulfonyl, trimethylsilyl, triisopropylsilyl, and the like.
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, methoxymethyl groups, for example.
The term "carbonyl protecting group," as used herein, refers to a labile chemical moiety which is known in the art to protect a carbonyl group against undesired reactions during synthetic procedures. After said synthetic procedure(s) the carbonyl protecting group as described herein may be selectively removed. Carbonyl protecting groups as known in the art 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 carbonyl protecting groups include acetals, ketals, cyclic acetals, cyclic ketals, mono- or dithio acetals, mono- or dithioketals, optionally substituted hydrazones or oximes.
The term "protected carbonyl," as used herein, refers to a carbonyl group protected with a carbonyl protecting group, as defined above, including dimethyl acetal, 1,3- dioxolane, 1,3-dioxane, S,S'-dimethylketal, 1,3-dithiane, 1,3-dithiolane, 1,3-oxathiolane, N,N-dimethylhydrazone, oxime, for example.
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 art 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, methoxycarbonyl, t- butoxycarbonyl, 9-fluorenylmethoxycarbonyl, benzyloxycarbonyl, and the like.
The term "protected amino," as used herein, refers to an amino group protected with an amino protecting group as defined above.
The term "substituted amino," as used herein, refers to substitution by replacement of one or two hydrogen atoms of -NH2 with substituents independently selected from the group consisting of optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cycloalkyl, optionally substituted heteroaryl, and optionally substituted heterocyclic; alternatively, when disubstituted, the two substitutents can be optionally taken together with the nitrogen atom to which they are attached to form an optionallysubstituted heterocyclic group.
The term "leaving group" means a functional group or atom which can be displaced by another functional group or atom in a substitution reaction, such as a nucleophilic substitution reaction. By way of example, representative leaving groups include chloro, bromo and iodo groups; sulfonic ester groups, such as mesylate, tosylate, brosylate, nosylate and the like; hydroxy; imidazolyl; and acyloxy groups, such as acetoxy, trifluoroacetoxy 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 compounds are well known to those skilled in the art, and it will be obvious to those skilled in the art that 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 term "protic solvent," as used herein, refers 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 it will be obvious to those skilled in the art that 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 Formula 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. 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, 2nd Ed. Wiley-VCH (1999); T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley and Sons (1999); 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), and subsequent editions thereof.
The term "subject," as used herein, refers to an animal. Preferably the animal is a mammal. More preferably the mammal is a human. A subject also refers to, for example, dogs, cats, horses, cows, pigs, guinea pigs, fish, birds and the like. The compounds of this invention may be modified by appending appropriate functionalities to enhance selective biological properties. Such modifications are known in the art and may 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.
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 olefmic double bonds, other unsaturation, or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers or cis- and trans- isomers. Likewise, all tautomeric forms are also intended to be included. Tautomers may be in cyclic or acyclic. 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 or carbon- heteroatom double bond depicted arbitrarily herein as trans may be cis, trans, or a mixture of the two in any proportion.
Certain compounds of the present invention may also exist in different stable conformational forms which may be separable. Torsional asymmetry due to restricted rotation about an asymmetric single bond, for example because of steric hindrance or ring strain, may permit separation of different conformers. The present invention includes each conformational isomer of these compounds and mixtures thereof.
As used herein, the term "pharmaceutically acceptable salt" refers to those salts 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. For example, S. M. 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 are 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.
As used herein, the term "pharmaceutically acceptable ester" refers to esters 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 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 a compound of the 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 present invention also relates to solvates of the compounds of Formula (1-1),
Formula (2-1), Formula (3-1), Formula (4-1) and Formula (5-1), for example hydrates.
This invention also encompasses pharmaceutical compositions containing, and methods of treating viral infections through administering, pharmaceutically acceptable prodrugs of compounds of the invention. For example, compounds of the invention having free amino, amido, hydroxy or carboxylic groups can be converted into prodrugs. Prodrugs include compounds wherein an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues is covalently joined through an amide or ester bond to a free amino, hydroxy or carboxylic acid group of compounds of the invention. The amino acid residues include, but are not limited to, the 20 naturally occurring amino acids commonly designated by three letter symbols and also includes 4- hydroxyproline, hydroxylysine, demosine, isodemosine, 3-methylhistidine, norvalin, beta- alanine, gamma-aminobutyric acid, citrulline, homocysteine, homoserine, ornithine and methionine sulfone. Additional types of prodrugs are also encompassed. For instance, free carboxyl groups can be derivatized as amides or alkyl esters. Free hydroxy groups may be derivatized using groups including but not limited to hemisuccinates, phosphate esters, dimethylaminoacetates, and phosphoryloxymethyloxycarbonyls, as outlined in Advanced Drug Delivery Reviews, 1996, 19, 115. Carbamate prodrugs of hydroxy and amino groups are also included, as are carbonate prodrugs, sulfonate esters and sulfate esters of hydroxy groups. Derivatization of hydroxy groups as (acyloxy)methyl and (acyloxy)ethyl ethers wherein the acyl group may be an alkyl ester, optionally substituted with groups including but not limited to ether, amine and carboxylic acid functionalities, or where the acyl group is an amino acid ester as described above, are also encompassed. Prodrugs of this type are described in J. Med. Chem. 1996, 39, 10. Free amines can also be derivatized as amides, sulfonamides or phosphonamides. All of these prodrug moieties may incorporate groups including but not limited to ether, amine and carboxylic acid functionalities.
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 or excipients.
As used herein, the term "pharmaceutically acceptable carrier or excipient" 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 as propylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminun 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 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, intraarticular, 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 that 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 and other coatings well known in the pharmaceutical formulating art. 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 that 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.
For pulmonary delivery, a therapeutic composition of the invention is formulated and administered to the patient in solid or liquid particulate form by direct administration e.g., inhalation into the respiratory system. Solid or liquid particulate forms of the active compound prepared for practicing the present invention include particles of respirable size: that is, particles of a size sufficiently small to pass through the mouth and larynx upon inhalation and into the bronchi and alveoli of the lungs. Delivery of aerosolized therapeutics, particularly aerosolized antibiotics, is known in the art (see, for example U.S. Pat. No. 5,767,068 to VanDevanter et al, U.S. Pat. No. 5,508,269 to Smith et al, and WO 98/43650 by Montgomery, all of which are incorporated herein by reference). A discussion of pulmonary delivery of antibiotics is also found in U.S. Pat. No. 6,014,969, incorporated herein by reference. ANTIVIRAL ACTIVITY
An inhibitory amount or dose of the compounds of the present invention may range from about 0.01 mg/Kg to about 500 mg/Kg, alternatively from about 0.1 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 patient such as a human or another animal by administering to the subject a therapeutically effective amount of a compound of the 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 "therapeutically effective amount" of a compound of the invention, as used herein, means an amount of the compound which confers a therapeutic effect on the treated subject, at a reasonable benefit/risk ratio applicable to any medical treatment. The therapeutic effect may be objective (i.e., measurable by some test or marker) or subjective (i.e., subject gives an indication of or feels an effect).
The term "inhibitory amount" of a compound of the present invention means a sufficient amount to decrease the viral load in a biological sample or a subject (e.g., resulting in at least 10%, preferably at least 50%, more preferably at least 80%, and most preferably at least 90%> or 95%, reduction in viral load). 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.
Effective doses will also vary depending on route of administration, as well as the possibility of co-usage with other agents. 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 therapeutically effective dose level 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 contemporaneously with the specific compound employed; and like factors well known in the medical arts.
The total daily dose of the compounds of this invention administered to a human or other animal 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.
Upon improvement of a patient'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. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
When the compositions of this invention comprise a combination of a compound of the Formula (1-1), Formula (2-1), Formula (3-1), Formula (4-1) and Formula (5-1), described herein and one or more additional therapeutic or prophylactic agents, both the compound and the additional agent should be present at dosage levels of between about 1 to 100%, and more preferably between about 5 to 95% of the dosage normally
administered in a monotherapy regimen. The additional agents may be administered separately, as part of a multiple dose regimen, from the compounds of this invention. Alternatively, those agents may be part of a single dosage form, mixed together with the compounds of this invention in a single composition.
The said "additional therapeutic or prophylactic agents" include, but are not limited to, immune therapies (eg. interferon), therapeutic vaccines, antifibrotic agents, anti-inflammatory agents such as corticosteroids or NSAIDs, bronchodilators such as beta- 2 adrenergic agonists and xanthines (e.g. theophylline), mucolytic agents, anti- muscarinics, anti-leukotrienes, inhibitors of cell adhesion (e.g. ICAM antagonists), antioxidants (eg N-acetylcysteine), cytokine agonists, cytokine antagonists, lung surfactants and/or antimicrobial and anti-viral agents (eg ribavirin and amantadine). The compositions according to the invention may also be used in combination with gene replacement therapy.
COMBINATION AND ALTERNATION THERAPY FOR HCV
It has been recognized that drug-resistant variants of HCV can emerge after prolonged treatment with an antiviral agent. Drug resistance most typically occurs by mutation of a gene that encodes for a protein such as an enzyme used in viral replication, and most typically in the case of HCV, RNA polymerase, protease, or helicase.
Recently, it has been demonstrated that the efficacy of a drug against a viral infection, such as HIV, can be prolonged, augmented, or restored by administering the drug in combination or alternation with a second, and perhaps third, antiviral compound that induces a different mutation from that caused by the principal drug. Alternatively, the pharmacokinetics, biodistribution, or other parameter of the drug can be altered by such combination or alternation therapy. In general, combination therapy is typically preferred over alternation therapy because it induces multiple simultaneous stresses on the virus.
A compound of the present invention can also be administered in combination or alternation with antiviral agent. Examplary antiviral agents include ribavarin, interferon, interleukin or a stabilized prodrug of any of them. More broadly described, the compound can be administered in combination or alternation with any of the anti-HCV drugs listed in Table 53 below.
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0002
Unless otherwise defined, all technical and scientific terms used herein are accorded the meaning commonly known to one of 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 may be used in the descriptions of the scheme and the examples that follow are: Ac for acetyl; AcOH for acetic acid; AIBN for
azobisisobutyronitrile; BINAP for 2,2'-bis(diphenylphosphino)-l, -binaphthyl; Boc20 for di-fert-butyl-dicarbonate; Boc for t-butoxycarbonyl; Bpoc for 1 -methyl- 1 -(4- biphenylyl)ethyl carbonyl; BtOH for 1-hydroxy-benzotriazole; Bz for benzoyl; Bn for benzyl; BocNHOH for tert-butyl N-hydroxycarbamate; t-BuOK for potassium tert- butoxide; Bu3SnH for tributyltin hydride; BOP for (benzotriazol-1- yloxy)tris(dimethylamino)phos-phonium Hexafluorophosphate; Brine for sodium chloride solution in water; Cbz for carbobenzyloxy; CDI for carbonyldiimidazole; CH2C12 for dichloromethane; CH3 for methyl; CH3CN for acetonitrile; Cs2C03 for cesium carbonate; CuCl for copper (I) chloride; Cul for copper (I) iodide; dba for dibenzylidene acetone; dppb for diphenylphosphino butane; DBU for 1,8-diazabicyclo[5.4.0]undec-7-ene; DCC for Ν,Ν'-dicyclohexylcarbodiimide; DEAD for diethylazodicarboxylate; DIAD for diisopropyl azodicarboxylate; DIPEA or (i-Pr)2EtN for Ν,Ν-diisopropylethyl amine; Dess- Martin periodinane for l,l,l-tris(acetyloxy)-l,l-dihydro-1,2-benziodoxol-3-(1H)-one; DMAP for 4-dimethylaminopyridine; DME for 1 ,2-dimethoxy-ethane; DMF for N,N- dimethylformamide; DM SO for dimethyl sulfoxide; DMT for
Figure imgf000126_0001
methoxyphenyl)phenylmethyl or dimethoxytrityl; DPPA for diphenylphosphoryl azide; EDC for N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide; EDC HCl for N-(3- dimethylamino-propyl)-N'-ethylcarbodiimide hydrochloride; EtOAc for ethyl acetate; EtOH for ethanol; Et20 for diethyl ether; Fmoc for 9-fluorenylmethoxycarbonyl; HATU for 0-(7-azabenzotriazol-1-yl)-N,N,N',N',-tetramethyluronium Hexafluorophosphate; HCl for hydrogen chloride; HOBT for 1-hydroxybenzotriazole; K2C03 for potassium carbonate; n-BuLi for n-butyl lithium; z'-BuLi for z'-butyl lithium; t-BuLi for t-butyl lithium; PhLi for phenyl lithium; LDA for lithium diisopropylamide; LiTMP for lithium 2,2,6, 6-tetramethylpiperidinate; MeOH for methanol; Mg for magnesium; MOM for methoxymethyl; Ms for mesyl or -SO2-CH3; Ms20 for methanesulfonic anhydride or mesyl-anhydride; NaBH4 for sodium borohydride; NaBH3CN for sodium
cyanoborohydride; NaN(TMS)2 for sodium bis(trimethylsilyl)amide; NaCl for sodium chloride; NaH for sodium hydride; NaHC03 for sodium bicarbonate or sodium hydrogen carbonate; Na2C03 sodium carbonate; NaOH for sodium hydroxide; Na2S04 for sodium sulfate; NaHS03 for sodium bisulfite or sodium hydrogen sulfite; Na2S203 for sodium thiosulfate; NH2NH2 for hydrazine; NH4HC03 for ammonium bicarbonate; NH4C1 for ammonium chloride; NMMO for N-methylmorpholine N-oxide; NaI04 for sodium periodate; Ni for nickel; OH for hydroxyl; Os04 for osmium tetroxide; Pd for palladium; Ph for phenyl; PMB for p-methoxybenzyl; POPd for dihydrogen dichlorobis(di-tert- butylphosphinito-KP)palladate(II); Pd2(dba)3 for tris(dibenzylidene-acetone) dipalladium (0); Pd(PPh3)4 for tetrakis(triphenylphosphine)palladium (0); PdCl2(PPh3)2 for trans- dichlorobis(triphenyl-phosphine)palladium (II); Pt for platinum; Rh for rhodium; rt for romm temperature; Ru for ruthenium; SEM for (trimethylsilyl)ethoxymethyl; TBAF for tetrabutylammonium fluoride; TBS for tert-butyl dimethylsilyl; TEA or Et3N for triethylamine; Teoc for 2-trimethylsilyl-ethoxy-carbonyl; TFA for trifluoroacetic acid; THF for tetrahydrofuran; TMEDA for Ν,Ν,Ν' ,Ν'-tetramethylethylenediamine; TPP or
PPh3 for triphenyl-phosphine; Troc for 2,2,2-trichloroethyl carbonyl; Ts for tosyl or -S02- C6H4CH3; Ts20 for tolylsulfonic anhydride or tosyl-anhydride; TsOH for p-tolylsulfonic acid; TMS for trimethylsilyl; or TMSC1 for trimethylsilyl chloride.
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. Starting materials can be obtained from commercial sources or prepared by well-established literature methods known to those of ordinary skill in the art. It will be readily apparent to one of ordinary skill in the art that the compounds defined above can be synthesized by substitution of the appropriate reactants and agents in the syntheses shown below. It will also be readily apparent to one skilled in the art that the selective protection and deprotection steps, as well as the order of the steps themselves, can be carried out in varying order, depending on the nature of the variables to successfully complete the syntheses below. The variables are as defined above unless otherwise noted below.
The compounds of the present invention may be prepared via several different synthetic routes from a variety of 5/6-membered fused heteroaryl, 5-membered heteroaryl, and related intermediates. A retro-synthesis of the compounds include direct formation of a suitably linked core structure (5/6-membered fused heteroaryl or 5-membered heteroaryl) followed by attachment of a suitable capping group (such as -C(0)R6), plus some functional group manipulations in between and/or after. Various 5/6-membered fused heteroaryl or 5-membered heteroaryl intermediates are known to those skilled in the art, for example see the encyclopedic volumns edited by A. R. Katrizky, et al,
"Comprehensive Heterocyclic Chemistry" 1984; "Comprehensive Heterocyclic Chemistry II" 1996; "Comprehensive Heterocyclic Chemistry III" 2008.
A general synthesis and further elaboration of some 6-membered ring fused imidazole related intermediates are summarized in Scheme 1, in which Z is N or CH.
The synthesis starts from the construction of an optionally substituted
imidazopyridine or benzimidazole 1-2, which may be obtained by condensation of an amino acid or its derivative 1-1.1 or 1-1.2 and a 2,3-diaminopyridine or 1,2- diaminobenzene 1-1 under the conditions to those skilled in the art. The imidazole ring closure may be realized either in one pot by heat, optionally in the presence of an acid and/or with a dehydration reagent such as polyphosphoric acid; or in two steps: 1) amide formation between diamine 1-1 and amino acid 1-1.1 or 1-1.2 in the presence of a condensation reagent such as EDC C1 , DCC or the like; or through mixed anhydride approach by reacting acid 1-1.1 or 1-1.2 with a chloroformate such as methyl
chloroformate, isobutyl chloroformate, or the like, in the presence of a base such as TEA, DIPEA, DMAP, N-methylmorpholine, or the like, followed by treating the mixed anhydride with diamine 1-1; and 2) the heterocyclic ring closure in the presence of an acid such as acetic acid, sulfuric acid or the like or a dehydration reagent such as HATU or the like, optionally with heat.
Figure imgf000129_0001
Optionally, the NH group in the newly formed imidazopyridine or benzimidazole ring of 1-2 may be protected with an amino protecting group, such as SEM (i.e. SEM-C1, NaH), Boc, Cbz, Teoc, Troc, or the like. The protected imidazopyridine or benzimidazole 1-2 may be subjected to lithium-halogen exchange with various (n-, s-, or t-) butyl lithium and the resulting lithiate can be trapped with a nucleophile, i.e. a halide such as various allyl halide to give the allylated 1-6 as a key intermediate. Alternatively, 1-6 may be obtained from the Stille reaction conditions to those skilled in the art (see reviews: A. Anastasia, et al, Handbook of Organopalladium Chemistry for Organic Synthesis, 2002, 1, 311; F. Bellina, et al, Synthesis 2004, 2419; M. G. Organ, et al, Synthesis 2008, 2776; A. T. Lindhardt, et al, Chem. - A European J., 2008, 14, 8756; E. A. B. Kantchev, et al, Angew. Chem. Int. Ed., 2007, 46, 2768; V. Farina, et al, Advances in Metal-Organic Chem., 1996, 5, 1), using an allylstanne such as allyltributylstanne as the allyl donor. Analogously a key vinyl intermediates 1-3 may be prepared by Stille reaction from bromide 1-2 with tributylvinylstanne. Also, Sonogashira coupling between bromide 1-2 and propargyl alcohol or trimethylsilyl-acetylene can generate propargyl alcohol 1-4 or alkyne 1-5 after removal of TMS. Further bromination of intermediate 1-4 may form the propargyl bromide 1-9. In addition, the bromide 1-2 may be converted to methyl ketone 1-7 by coupling with tributyl(l-ethoxyvinyl)tin under Stille coupling conditions followed by acidic hydrolysis.
Further elaboration of the imidazopyridine or benzimidazole intermediates starts from the vinyl intermediate 1-3, which may be transformed to aldehyde 1-8 through ozonolysis or periodate/OsC^ cleavage or to alcohol 1-12 by hydroboration-oxidation sequence. Alcohol 1-12 may be converted to bromide 1-15 by the well-known
bromination procedure, which can be further functionalized to amine 1-20 through azide substitution followed by reduction. Aldehyde 1-8 can then either be reduced to alcohol 1- 11, or be converted to a, β-unsatuated acid 1-10 through Horner- Wadsworth-Emmons aldehyde homologation reaction followed by saponification. Alcohol 1-11 may be similarly converted to the correponding amine intermediate 1-14 and bromide intermediate 1-13 as described previously. Bromide 1-13 can be homologated to alkyne intermediate 1- 19 with a metal acetylide. In addition, bromide 1-13 may be also tranformed to thiol 1-16 through nucleophilic substitution, which can be further oxidized to sulfonic acid 1-17. Sulfonamide 1-18 may then be derived from 1-17 through the sulfonyl chloride activation process.
It should be noted that optionally the NH group of all the imidazopyridine or benzimidazole related intermediates listed above may be protected with an amino protecting group, such as SEM (i.e. SEM-C1, NaH), Boc, Cbz, Teoc, Troc, or the like.
Figure imgf000131_0001
A typical syntheses of imidazole related intermediates are analogous to that of the imidazopyridine or benzimidazole intermediates. As shown in Scheme 2, bromo- imidazole 2-4 can be synthesized in a three-step sequence: 1) condensation between amino acid derived aldehyde 2-1.1 or 2-1.2 and glyoxal 2-1.3 in the presence of methanolic ammonia to generate imidazole 2-2; 2) bromination of 2-2 with excess amount of bromination reagent such as 2,4,4,6-tetrabromo-2,5-cyclohexadienone, NBS, etc. to afford dibromide 2-3; and 3) selective reduction of the dibromide 2-3 by heating in aq. Na2S03 or aq. NaHS03. 2-4 then may be served as a universal intermediate further elaborable to many other imidazole derivatives using the chemistry discussed in Scheme 1, some of which are listed in the table 54 below.
Figure imgf000131_0002
Optionally, the NH group of imidazole related intermediates listed above may be protected with an amino protecting group (shown in Scheme 2 as PG, theoretically the reaction will generate two regio-isomers, but only one is drawn for clarity), such as SEM (i.e. SEM-C1, NaH), Boc, Cbz, Teoc, Troc, or the like. The protected imidazole 2-5 may be deprotonated with a strong base such as LDA, BuLi, etc to generate a carbon anion, which may either undergo a nucleophilic substitution with an activated halide such as 2- 5.2 to afford aryl or heteroaryl substituted imidazole 2-6 or couple with an aryl or heteroaryl halide 2-5.1 in the presence appropriate transition metal salt to generate bicyclic heteroaryl 2-7. Similarly, the protected bromo imidazole 2-8 may be subjected to lithium- halogen exchange with various (n-, s-, or t-) butyl lithium, the resulting lithiate may undergo similar reactions to afford 2-6 and 2-7. Also, when 2-8 is treated with metallated aryl or heteroaryl 2-8.1, in which M at each occurrence is independently a boron, tin, silicon, zinc, zirconium, or copper species, under Suzuki, Stille or related coupling conditions known to those skilled in the art (see reviews: A. Suzuki, Pure Applied Chem. 1991, 63, 419; A. Suzuki, Handbook of Organopalladium Chemistry for Organic
Synthesis, 2002, 1, 249; A. Anastasia, et al, Handbook of Organopalladium Chemistry for Organic Synthesis, 2002, 1, 311; F. Bellina, et al, Synthesis 2004, 2419; M. G. Organ, et al, Synthesis, 2008, 2776; A. T. Lindhardt, et al, Chem. - A European J., 2008, 14, 8756; E. A. B. Kantchev, et al, Angew. Chem. Int. Ed., 2007, 46, 2768; V. Farina, et al, Advances in Metal-Organic Chem., 1996, 5, 1), to provide coupling product 2-7. In addition to these direct coupling strategy, aryl or heteroaryl bromide 2-5.1 may be converted to methyl ketone 2-9 under Stille coupling conditions with tributyl(l-ethoxyvinyl)tin 2-9.1. 2-9 may be brominated under conditions to those skilled in the art to afford bromide 2-10, which may be either converted to the corresponding amine 2-11, or coupled with protected amino acid 1-1.1 or 1-1.2 in the presence of a base such as Et3N and DIPEA to afford keto-ester 2-12. Similarly, amine 2-11 may be converted to the corresponding keto-amide 2-13 via condensation with appropriate amino acid under standard amide formation conditions. 2- 12 and 2-13 may be tranformed to key intermediate 2-14 via heating with (NH4)OAc under thermal or microwave conditions.
Similarly when Q is a fused bicyclic group as previously defined, compounds of the present invention may be synthesized from intermediates 3-2 and 3-3 (as shown in Scheme 3), which may be synthesized using similar chemistry described in Schemes 1 and 2 from the bicyclic carboxylic acid 3-1. Bicyclic carboxylic acids or derivatives with various structural features are either commercially available or known in the literature, and have been extensively reviewed by S. Hanessian, et al {Tetrahedron, 1997, 53, 12789) and A. Trabocchi, et al {Amino Acids, 2008, 34, 1) which are incorporated herein by reference.
Scheme 3
Figure imgf000133_0001
Other requisite buiding blocks for the syntheses of the molecule of the present invention are various amide, carbamate or urea-related intermediates, which can be prepared from a suitable, commercially available amine through standard amide, carbamate or urea formation by direct condensation of an carboxylic acid with a dehydration and/or condensation reagent, such as HATU, DCC, BtOH, EDC or the like; or a chloroformate; or an isocyanate; in the presence of a suitable base such as pyridine, TEA, DIPEA, DMAP, NaHC03, K2C03 or the like. For example, as shown in Scheme 3a, the Boc-protected β-amino acid 3- la can be condensed with 4-bromoaniline 3-2a in the presence of HATU and DIPEA in CH2C12 to afford amide 3-3a.
Figure imgf000133_0002
3-1 a 3-2a 3-3a
Other examples of some of the amide -related intermediates that may be used to construct the title compounds of the present invention are compiled in the table 55 below.
Table 55
Figure imgf000133_0003
Figure imgf000134_0001
In addition to the intermediates mentioned above, other requisite buiding blocks for the syntheses of the molecule of the present invention are various heteroaryl or aryl amine related intermediates, which can be prepared from a suitable, commercially available amine through standard nucleophilic substitution by direct condensation of a heteroaryl halide such as 5-bromo-2-chloropyrimidine, 2,6-dibromoquinazoline, 6-bromo- 2-chlorobenzothiazole, or the like; with an appropriate amine such as 2-aminomethyl-N- Boc-pyrrolidine, l-Boc-3-aminopyrrolidine, trans-N-Boc1,2-cyclopentanediamine or the like; in the presence of a suitable base such as pyridine, TEA, DIPEA, DMAP, NaHC03, K2C03 or the like; under thermal or microwave conditions. Alternatively, some of these heteroaryl or aryl amino related intermediates can be prepared through transition metal catalyzed amination process between an suitable amine as mentioned above and an aryl or heteroaryl iodide or bromide such as 5-bromo-2-iodopyridine, 1 ,4-diiodobenzene, or the like; in the presence of a suitable base such as Cs2C03, K3P04, K2C03 or the like; and an appropriate transition metal catalyst such as Cul or various Pd related catalysts; and an related ligand according to the used transition metal catalyst; under inert atmosphere. For example, as shown in Scheme 3b, the Boc-protected β-diamine 3-lb can be condensed with 5-bromo-2-chloropyrimidine 3-2b in the presence of DIPEA in 1,4-dioxane under thermal or microwave conditions to afford pyrimidynyl amine 3-3b. Alternatively, 3-lb can be reacted with 5-bromo-2-iodopyridine 3-4b in the presence of Cs2C03, catalytic amount of Cul, and 2-isobutyrylcyclohexanone as ligand, in DMF under N2 atmosphere to yield the desired pyridinyl amine 3-5b as described by S. L. Buchwald (J. Am. Chem. Soc. 2006, 128, 8742).
Figure imgf000135_0001
3-1 b 3-4 b 3-5b
Other examples of some aryl or heteroaryl amine related intermediates that may be used to construct the title compounds of the present invention are compiled in the table 56 below.
Figure imgf000135_0002
imidazoles such as those listed in Schemes 1-3 and the amide-related derivatives such as that in Scheme 3a, and the tables above in hand, the compounds of the present invention may be prepared through various coupling strategy or a combination of strategies to connect two fragments, optionally with a suitable cyclic or acyclic linker or formation of a cyclic or acyclic linker. The said strategy includes, but not limited to, Stille coupling, Suzuki coupling, Sonogashira coupling, Heck coupling, Buchwald amidation, Buchwald amination, alkylation, pericyclic reaction with different variations, or the like.
An example of the strategies that may be used to prepare the compounds of the present invention is shown in Scheme 4. Both bromides 4-1 and 4-2 can be prepared using similar procedures described in Schemes 1-3 and 3a. Bromide 4-2 can be converted to the corresponding metallated aryl 4-3 (boronate or stanne) under Suzuki or Stille conditions with bis(pinacolato)diboron, hexamethylditin or hexabutylditin in the presence of Pd- catalyst. The latter may be further coupled with imidazopyridine bromide 4-1 under similar conditions to generate a core structure 4-4.
Compound 4-4 then may be served as a common intermediate for further derivatizations to 4-5 in two steps: 1) mono-deprotection of the linear or cyclic amine moiety may be accomplished, for example, treatment to hydrogenolytic conditions under Pd catalyst to remove the Cbz protection group; and 2) the released amine functionality may be acylated with an carboxylic acid under standard acylation conditions, for example a coupling reagent such as HATU in combination with an organic base such as DIPEA can be used in this regard; alternatively, the released amine may be reacted with an isocyanate, carbamoyl chloride or chloroformate to provide an urea or carbamate. Various carboxylic acids including amino acids in racemic or optical form are commercially available, and/or can be synthesized in racemic or optical form, see references cited in reviews by D.
Seebach, et al, Synthesis, 2009, 1; C. Cativiela and M. D. Diaz-de-Villegas, Tetrahedron: Asymmetry, 2007, 18, 569; 2000, 11, 645; and 1998, 9, 3517; and experimental examples compiled in patent application WO 08/021927 A2 by C. Bachand, et al, from Brisol- Myers Squibb, Co., which is incorporated herein by reference. 4-5 may be further deprotected under hydrolytic conditions in the presence of an acid such as TFA or hydrogen chloride to remove the Boc protection group and the released amine
functionality can be further derivatized to the title compounds 1-1, with a carboxylic acid using the conditions described above.
Figure imgf000137_0001
Alternatively, as shown in Scheme 4a, the compounds of the present invention (for example I-l) may also be derived from bromoimidazopyridine or bromobenzimidazole 4- la and amide 4-2a using the procedures described previously. The intermediates 4- la and 4-2a have the desired acyl groups already installed using similar sequences shown in Scheme 4.
Figure imgf000137_0002
The compounds of the present invention containing five-membered heteroaryl other than imidazole may be prepared using similar procedures described above in Schemes 1-4 and 4a. For example, some intermediates containing a desired, suitably substituted five-membered heteroaryl have been published in US 2008/0311075A1 by C. Bachand, et al from Bristol-Myers Squibb, Co., which is incorporated by reference.
Theses intermediates are compiled in the following table 57. Table 57
Figure imgf000138_0003
The synthesis of the compounds of the present invention involves 5/6-membered fused heteroaryl intermediates other than imidazopyridines or benzimidazoles, various 5/6-membered fused heteroaryl are known in the literature. The synthesis of other 5/6- membered fused heteroaryl intermediates depends on the chemical features of each structure. For example, a typical synthesis of indole intermediate is illustrated in Scheme 5. The commercially available bromoiodoaniline 5-1 may be coupled to the commercially available acetylene 5-1.1 under the Sonogashira conditions to give phenylacetylene 5-2. The latter may be cyclized to indole 5-3 under heat or microwave condition in the presence of a copper catalyst.
Figure imgf000138_0001
It will be appreciated that, with appropriate manipulation and protection of any chemical functionality, synthesis of compounds of Formula (I) is accomplished by methods analogous to those above and to those described in the Experimental section. Suitable protecting groups can be found, but are not restricted to, those found in T W Greene and P G M Wuts "Protective Groups in Organic Synthesis", 3rd Ed (1999), J Wiley and Sons.
In certain aspects, the invention encompasses a process of making a compound of the invention comprising:
i) preparing a compound of Formula (II):
Figure imgf000138_0002
via a transition-metal catalyzed cross-coupling reaction; wherein:
G, J, U, R1, Rla, Rlb, R7a, R7b, R9, and R9a are as defined above in Formula (l-I), (2-1),
(3-1), (4-1) or (5-1);
Ring A1 is absent, optionally substituted aryl or optionally substituted heteroaryl;
Ring B1 is optionally substituted aryl or optionally substituted heteroaryl;
L1 is absent, optionally substituted C2-C4 alkenyl or C2-C4 alkynyl; and
Za and Zb are each independently an amino protecting group or -C(0)-R6; wherein R6 is as defined above;
iiii)) wwhheenn ZZaa oorr ZZbb iiss an amino protecting group, fully or selectively deprotecting a compound of Formula (II) to give the corresponding amine of Formula (III):
Figure imgf000139_0001
wherein Zc is hydrogen, an amino protecting group or -C(0)-R6;
iii) capping the released amino group of a compound of Formula (III) with LG-C(O)-
R6,
wherein LG is a leaving group; to give the compound of Formula (IV):
Figure imgf000139_0002
wherein Zd is an amino protecting group -C(0)-R6; and
iv) repeated reaction sequence of deprotecting and capping (step ii-iii) to give the compound of Formula (V):
Figure imgf000139_0003
In another aspect, the invention is a process of making a compound according to Formula (l-I) comprising:
i) preparing a compound of Formula (l-II):
Figure imgf000139_0004
via a transition-metal catalyzed cross-coupling reaction; wherein:
G, J, U, R1, Rla, Rlb, R7a, R7b, R9, and R9a are as defined in Formula (l-I);
Ring A1 is absent, optionally substituted aryl or optionally substituted heteroaryl; Ring B1 is optionally substituted aryl or optionally substituted heteroaryl;
L1 is absent, optionally substituted C2-C4 alkenyl or C2-C4 alkynyl; and
Za and Zb are each independently an amino protecting group or -C(0)-R6; wherein R6 is as defined in in Formula (1-1);
ii) when Za or Zb is an amino protecting group, fully or selectively deprotecting a compound of Formula (l-II) to give the corresponding amine of Formula (l-III):
Figure imgf000140_0001
wherein Zc is hydrogen, an amino protecting group or -C(0)-R6;
iii) capping the released amino group of a compound of Formula (l-III) with LG-
C(0)-R6,
wherein LG is a leaving group; to give the compound of Formula (1-IV):
Figure imgf000140_0002
wherein Zd is an amino protecting group -C(0)-R6; and
iv) repeated reaction sequence of deprotecting and capping (step ii-iii) to give the compound of Formula (1-V):
Figure imgf000140_0003
In yet another aspect, the invention is a process of making a compound accordingormula (2-1) comprising:
i) preparing a compound of Formula (2-II):
Figure imgf000140_0004
via a transition-metal catalyzed cross-coupling reaction; wherein:
G, J, U, R1, Rla, Rlb, R7a, R7b, R9, and R9a are as defined in Formula (2-1);
Ring A1 and Ring B1 are each independently optionally substituted aryl or optionally substituted heteroaryl;
L1 is absent, optionally substituted C2-C4 alkenyl or C2-C4 alkynyl; and Za and Zb are each independently an amino protecting group or -C(0)-R6; wherein R6 is as defined in Formula (2-1);
ii) when Za or Zb is an amino protecting group, fully or selectively deprotecting a compound of Formula 2-II) to give the corresponding amine of Formula (2-III):
Figure imgf000141_0001
wherein Zc is hydrogen, an amino protecting group or -C(0)-R6;
iii) capping the released amino group of a compound of Formula (2-III) with LG-
C(0)-R6,
wherein LG is a leavin group; to give the compound of Formula (2-IV):
Figure imgf000141_0002
wherein Zd is an amino protecting group -C(0)-R6; and
iv) repeated reaction sequence of deprotecting and capping (step ii-iii) to give the compound of Formula 2-V):
Figure imgf000141_0003
In a further aspect, the invention is a process of making a compound of Formula (3-1) comprising:
i) preparing a compound of Formula (3 -II):
Figure imgf000141_0004
via a transition-metal catalyzed cross-coupling reaction; wherein:
G, J, U, R1, Rla, Rlb, R7a, R7b, R9, and R9a are as defined in Formula (3-1);
Ring A1 and Ring B1 are each independently absent or an optionally substituted aryl or optionally substituted heteroaryl;
L1 is absent, optionally substituted C2-C4 alkenyl or C2-C4 alkynyl; and
Za and Zb are each independently an amino protecting group or -C(0)-R6; wherein R6 is as defined in Formula (3-1); ii) when Za or Zb is an amino protecting group, fully or selectively deprotecting a compound of Formula (3 -II) to give the corresponding amine of Formula (3 -III):
Figure imgf000142_0001
wherein Zc is hydrogen, an amino protecting group or -C(0)-R6;
iii) capping the released amino group of a compound of Formula (3 -III) with LG- C(0)-R6,
wherein LG is a leaving group; to give the compound of Formula (3-IV):
Figure imgf000142_0002
wherein Zd is an amino protecting group -C(0)-R6; and
iv) repeated reaction sequence of deprotecting and capping (step ii-iii) to give the compound of Formula (3-V):
Figure imgf000142_0003
In another aspect, the invention is a process of making a compound of Formula (4-omprising:
i) preparing a compound of Formula (4-II):
Figure imgf000142_0004
via a transition-metal catalyzed cross-coupling reaction;
wherein:
G, U, R1, Rla, Rlb, Rlc, R7a, R7b, R9, and R9a are as defined in Formula (4-1);
Ring A1 is absent, or optionally substituted aryl or optionally substituted heteroaryl;
Ring B1 is optionally substituted aryl or optionally substituted heteroaryl;
L1 is absent, or optionally substituted C2-C4 alkenyl or C2-C4 alkynyl; and
Za or Zb are each independently an amino protecting group or -C(0)-R6; wherein R6 is as defined in Formula (4-1); ii) when Za or Zb is an amino protecting group, fully or selectively deprotecting a compound of Formula (4-II) to give the corresponding amine of Formula (4-III):
Figure imgf000143_0001
wherein Zc is hydrogen, an amino protecting group or -C(0)-R6;
iii) capping the released amino group of a compound of Formula (4-III) with LG- C(0)-R6,
wherein LG is a leaving group; to give the compound of Formula (4-IV):
Figure imgf000143_0002
wherein Zd is an amino protecting group or -C(0)-R6; and
iv) repeated reaction sequence of deprotecting and capping (step ii-iii) to give the compound of Formula (4-V):
Figure imgf000143_0003
In an additional aspect, the invention is a process of making a compound of Formula (5-1) comprising:
i) preparing a compound of Formula (5-II):
Figure imgf000143_0004
via a transition-metal catalyzed cross-coupling reaction; wherein:
Ring B, G, U, R1, Rla, Rlb, Rlc, R7a, R7b, R9, and R9a are as defined in Formula (5-1);
Ring A1 is absent, optionally substituted aryl or optionally substituted heteroaryl;
L1 is absent, optionally substituted C2-C4 alkenyl or C2-C4 alkynyl; and
Za and Zb are each independently an amino protecting group or -C(0)-R6; wherein R6 is as defined in Formula (5-1);
ii) when Za or Zb is an amino protecting group, fully or selectively deprotecting a compound of Formula (5-II) to give the corresponding amine of Formula (5-III):
Figure imgf000144_0001
wherein Zc is hydrogen, an amino protecting group or -C(0)-R6;
iii) capping the released amino group of a compound of Formula (5-III) with LG- C(0)-R6,
wherein LG is a leaving group; to give the compound of Formula (5-IV):
Figure imgf000144_0002
wherein Zd is an amino protecting group or -C(0)-R6; and
iv) repeated reaction sequence of deprotecting and capping (step ii-iii) to give the compound of Formula (5-V):
Figure imgf000144_0003
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 limiting of 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.
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. The compounds of examples 1-1 to 1-82, 1-84 to 1-256, 1-258 to 1-326 and 1-328 1-347 may be prepared using procedures similar to those described in examples 1-83, -257 and 1-327 (described below), and/or as described in the Synthetic Methods.
-1 to 1-219.
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0004
Example 1-83.
Figure imgf000154_0001
The title compound was prepared from (S)-1-(tert-butoxycarbonyl)pyrrolidine-3- carboxylic acid and 4-bromoaniline 4-iodobenzoic acid using procedures similar described in examples 1-354 and 1-355. ESIMS m/z = Ί 16.51 [M+H]+.
-257.
Figure imgf000154_0002
Step l-257a. A solution of the compound from example 1-353 (32.5 μιηοΐ at most) in
CH2C12 (4 mL) was treated with C1 in 1,4-dioxane (4 M, 3 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step. ESIMS m/z = 416.37 [M+H]+.
Step l-257b. A mixture of the crude compound from step l-257a (32.5 μιηοΐ at most) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
2008/021927, 11.4 mg, 65.0 μιηοΐ) in DMF (3 mL) was treated with HATU (23.4 mg, 61.7 μιηοΐ) in the presence of DIPEA (81 μΐ^, 0.650 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, CH2Cl2-MeOH) to give the title compound as a light yellow solid
(16.9 mg, 3 steps 71%). ESIMS m/z = 730.63 [M+H]+.
Example 1-327.
Figure imgf000154_0003
The title compound was prepared from the compound of Example 1-366 using procedures similar to that described in example 1-359. ESIMS m/z = 740.43 [M+H]+.
-348a and l-348b.
Figure imgf000155_0001
Step l-348a. A mixture of 4-bromoaniline (0.500 g, 2.91 mmol) and trans (+/-) 2-(tert- butoxycarbonylamino)-cyclopentanecarboxylic acid (0.733 g, 3.20 mmol) in DMF (12 mL) were added EDC HCl (0.724 g, 3.78 mmol), HOBt (0.646 g, 3.78 mmol) and DIPEA (0.54 mL, 4.36 mmol). The mixture was stirred at room temperature until the
disappearence of the starting material. The mixture was partitioned (EtOAc - water) and the organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by fiash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a yellow white solid (0.787 g, 71%). ESIMS m/z = 404.99, 406.98 [M+H+Na]+.
Step l-348b. A mixture of 2,4'-dibromoacetophenone (5.00 g, 18.0 mmol) and N-Boc-L- proline (3.87 g, 18.0 mmol) in CH3CN (60 mL) was added TEA (5.40 mL, 37.8 mmol) slowly. The mixture was stirred at room temperature until the disappearence of the starting material. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by fiash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a light yellow foam (6.73 g, 91%). 1H NMR (CDC13) 7.76 (t, J= 8.0 Hz, 2H), 7.63 (dd, J= 5.0, 8.5 Hz, 2H), 5.51, 5.16 (2d, J= 16.0 Hz, 1H), 5.32, 5.28 (2d, J= 16.5 Hz, 1H), 4.48, 4.40 (dd, J= 5.0, 8.5 Hz, 1H), 3.56 (m, 1H), 3.43 (m, 1H), 2.30 (m, 2H), 2.06 (m, 1H), 1.92 (m, 1H), 1.46, 1.43 (2s, 9H).
Step l-348c. A solution of the compound from step l-348b (6.73 g, 16.3 mmol) in toluene (100 mL) was added ammonium acetate (25.1 g, 0.327 mol) and the resultant mixture was heated up to 100 °C for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - aq. NaHC03). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a yellow foam (6.10 g, 95%). ESIMS m/z = 392.24, 394.24 [M+H]+. 1H NMR (CDC13) 7.57 (bs, 1H), 7.48 (m, 3H), 7.23 (s, 1H), 4.97 (m, 1H), 3.42 (m, 2H), 2.99 (m, 1H), 2.16 (m, 2H), 1.97 (m, 1H), 1.46 (s, 9H).
Step l-348d. A mixture of the compound from step 348c (1.00 g, 2.55 mmol), bis-
(pinacolato) diboron (1.35 g, 5.33 mmol) and potassium acetate (0.640 g, 6.53 mmol) in 1,4-dioxane (20 mL) was added Pd(PPh3)4 (0.147 g, 0.128 mmol). The resultant mixture were degassed for three times and heated up to 80 °C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give intermediate 1 as a light yellow solid (0.978 g, 87%). ESIMS m/z = 440.39 [M+H]+. 1H NMR (CDC13) 11.03, 10.55 (2s, 1H), 7.79 (m, 3H), 7.45 (m, 1H), 7.26 (m, 1H), 4.97 (m, 1H), 3.41 (m, 2H), 3.06, 2.91 (2m, 1H), 2.17 (m, 2H), 1.97 (m, 1H), 1.49 (s, 9H), 1.35 (s, 12H).
Step l-348e. A mixture of the compound from step l-348a (0.200 g, 0.522 mmol), the compound from step 348d (0.229 g, 0.522 mmol) and NaHC03 (0.175 g, 2.09 mmol) in DME (12 mL) and H20 (4 mL) was added Pd(PPh3)4 (30.2 mg, 26.1 μπιοΐ). The resultant mixture were degassed and heated to 85°C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by
chromatography (silica, hexanes-ethyl acetate) to give a mixture of the title compounds as a light yellow solid (0.180 g, 56%). ESIMS m/z = 616.30 [M+H]+.
-349a and l-349b.
Figure imgf000156_0001
Step l-349a. A solution of the compound of example 1-348 (60.0 mg, 97.5 μιηοΐ) in CH2C12 (5 mL) was treated with C1 in 1,4-dioxane (4 M, 1 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step. ESIMS m/z = 416.20 [M+H]+. Step l-349b. A mixture of the crude compound from step l-348a (97.5 μιηοΐ at most) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
2008/021927, 35.8 mg, 0.205 mmol) in DMF (3 mL) was treated with HATU (74.1 mg, 0.195 mmol) in the presence of DIPEA (0.24 mL, 1.95 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, CH2Cl2-MeOH) to give a mixture of the title compounds as a white solid (63.2 mg, 2 steps 89%). ESIMS m/z = 730.33 [M+H]+.
-350a and l-350b.
Figure imgf000157_0001
Step l-350a. A mixture of 2-amino-6-bromonaphthalene (0.500 g, 2.25 mmol) and trans (+/-) 2-(tert-butoxycarbonylamino)-cyclopentanecarboxylic acid (0.567 g, 2.48 mmol) in DMF (10 mL) were added EDC C1 (0.561 g, 2.93 mmol), HOBt (0.500 g, 2.93 mmol) and DIPEA (0.42 mL, 3.38 mmol). The mixture was stirred at room temperature until the disappearence of the starting material. The mixture was partitioned (EtOAc - water) and the organics were washed with brine, dried (Na2S04), filtered and evaporated to give the desired compound as a red white solid (0.911 g, 93%). ESIMS m/z = 433.04, 435.04
[M+Hf.
Step l-350b. A mixture of the compound from step l-350a (0.200 g, 0.462 mmol), the compound from step 348d (0.184 g, 0.420 mmol) and NaHC03 (0.141 g, 1.68 mmol) in DME (12 mL) and H20 (4 mL) was added Pd(PPh3)4 (24.2 mg, 20.9 μπιοΐ). The resultant mixture were degassed and heated to 85°C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give a mixture of the title compounds as a light yellow white solid (0.198 g, 71%). ESIMS m/z = 666.33 [M+H]+. Examples 1-35 la and 1-35 lb.
Figure imgf000158_0001
Step l-351a. A solution of the compound from example 1-350 (50.0 mg, 75.1 μιηοΐ) in CH2CI2 (5 mL) was treated with C1 in 1,4-dioxane (4 M, 1 mL) for 30 min. The volatiles 5 were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step. ESIMS m/z = 466.24 [M+H]+.
Step l-351b. A mixture of the crude compound from step 1-35 la (75.1 μιηοΐ at most) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
2008/021927, 27.6 mg, 0.158 mmol) in DMF (3 mL) was treated with HATU (57.1 mg, 10 0.150 mmol) in the presence of DIPEA (0.19 mL, 1.50 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, CH2Cl2-MeOH) to give a mixture of the title compounds as a yellow solid (52.1 mg, 2 steps 89%). ESIMS m/z = 780.40 [M+H]+.
Example 1-352
Figure imgf000158_0002
Step l-352a. A mixture of (i?)-(+)-tert-butyl pyrrolidin-3-ylcarbamate (0.100 g, 0.537 mmol) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO 2008/021927, 0.103 g, 0.590 mmol) in DMF (3 mL) was treated with HATU (0.204 g, 0.537 mmol) in the presence of DIPEA (0.13 mL, 1.07 mmol) for 2 hours at rt and the 20 volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, CLLCb-MeOH) to give the desired compound as a colorless oil (0.194 g, ca.100%). ESIMS m/z = 344.37 [M+H]+.
Step l-352a. A solution of the compound from step l-352a (0.537 mmol at most) in CH2CI2 (6 mL) was treated with C1 in 1,4-dioxane (4 M, 6 mL) for 30 min. The volatiles 25 were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step. ESIMS m/z = 244.24 [M+H]+. Step l-352c. A mixture of the crude compound from step l-352b (0.537 mmol at most) and 4-iodobenzoic acid (0.146 g, 0.590 mmol) in DMF (3 mL) was treated with HATU (0.204 g, 0.537 mmol) in the presence of DIPEA (0.13 mL, 1.07 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, CH2Cl2-MeOH) to give the desired compound as a light yellow oil (0.198 g, 3 steps 78%). ESIMS m/z = 474.25 [M+H]+.
Step l-352d. A solution of the compound from step l-348c (1.500 g, 3.824 mmol) in 1,4- dioxane (12 mL) was treated with C1 in 1,4-dioxane (4 M, 24 mL) at room temperature for 1.5 hours. The volatiles were evaporated off to give the crude desired compound as a yellow solid, which was used directly in the next step. ESIMS m/z = 292.06, 294.06
[M+H]+.
Step l-352e. A mixture of the crude compound from step l-352d (3.824 mmol at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 0.670 g, 3.824 mmol) in DMF (12 mL) was treated with HATU (1.381 g, 3.633 mmol) in the presence of DIPEA (6.66 mL, 38.24 mmol) for 1 hour at room temperature and the volatiles were evaporated off. The residue was purified by
chromatography (silica, hexanes-ethyl acetate, with 1% Et3N in EtOAc) to give the desired compound as a yellow oil (1.500 g, 87% over 2 steps). ESIMS m/z = 449.12, 451.12
[M+H]+.
Step l-352f. A mixture of the compound from step l-352e (1.50 g, 3.34 mmol), bis-
(pinacolato) diboron (1.69 g, 6.68 mmol) and potassium acetate (0.818 g, 8.35 mmol) in 1,4-dioxane (20 mL) was added Pd(PPh3)4 (0.193 g, 0.167 mmol). The resultant mixture were degassed for three times and heated up to 85°C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give intermediate 2 as a colorless oil (1.15 g, 69%). ESIMS m/z = 497.35 [M+H]+.
Step l-352g. A mixture of the compound from step l-350c (39.8 mg, 84.1 μιηοΐ), the compound from step l-352f (54.2 mg, 0.109 mmol) and NaHC03 (28.3 g, 0.336 mmol) in DME (6 mL) and H20 (2 mL) was added Pd(PPh3)4 (4.9 mg, 4.2 μιηοΐ). The resultant mixture were degassed and heated to 85 °C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, CH2Cl2-MeOH) to give the title compound as a light yellow solid (50.8 mg, 84%). ESIMS m/z = 716.51 [M+H]+.
-353.
Figure imgf000160_0001
Step l-353a. A mixture of (S)-pyrrolidin-2-ylmethanamine (0.132 g, 0.661 mmol) and 4- iodobenzoic acid (0.167 g, 0.661 mmol) in CH2C12 (6 mL) was treated with HATU (0.251 g, 0.661 mmol) in the presence of DIPEA (0.16 mL, 1.32 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a colorless oil (0.256 g, 90%). ESIMS m/z = 431.22 [M+H]+.
Step l-353b. A mixture of the compound from step l-353a (49.0 mg, 0.114 mmol), the compound from step l-348d (60.0 mg, 0.137 mmol) and NaHC03 (38.3 mg, 0.456 mmol) in DME (6 mL) and H20 (2 mL) was added Pd(PPh3)4 (6.5 mg, 5.6 μιηοΐ). The resultant mixture were degassed and heated to 85 °C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the title compound as a light yellow oil (70.0 mg, ca.100%). ESIMS m/z = 616.48 [M+H]+.
-354.
Figure imgf000160_0002
Step l-354a. A mixture of l-Boc-pyrrolidine-3-carboxylic acid (0.100 g, 0.465 mmol) and 4-bromoaniline (79.9 mg, 0.465 mmol) in CH2C12 (6 mL) was treated with HATU (0.177 g, 0.465 mmol) in the presence of DIPEA (0.12 mL, 0.929 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a colorless oil (0.156 g, 91%). ESIMS m/z = 369.21 , 371.25 [M+H]+.
Step l-354b. A mixture of the compound from step l-354a (50.3 mg, 0.136 mmol), the compound from step l-348d (65.8 mg, 0.150 mmol) and NaHC03 (45.8 mg, 0.545 mmol) in DME (6 mL) and H20 (2 mL) was added Pd(PPh3)4 (7.9 mg, 6.8 μπιοΐ). The resultant mixture were degassed and heated to 85°C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the title compound as a colorless oil (72.6 mg, 89%). ESIMS m/z = 602.40 [M+H]+.
-355.
Figure imgf000161_0001
Step l-355a. A solution of the compound from example 1-354 (20.0 mg, 33.2 μιηοΐ) in CH2C12 (4 mL) was treated with C1 in 1,4-dioxane (4 M, 3 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step. ESIMS m/z = 402.38 [M+H]+.
Step l-355b. A mixture of the crude compound from step l-355a (max. 33.2 μιηοΐ) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
2008/021927, 12.2 mg, 69.7 μιηοΐ) in DMF (3 mL) was treated with HATU (25.2 mg, 66.4 μιηοΐ) in the presence of DIPEA (83 μΐ^, 0.664 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, CH2Cl2-MeOH) to give the title compound as a light yellow solid (17.0 mg, 2 steps 71%). ESIMS m/z = 716.64 [M+H]+.
-356a and l-356b.
Figure imgf000161_0002
Step l-356a. A mixture of trans-N-Boc-1,2-cyclopentanediamine (60.0 mg, 0.300 mmol) and 3-iodobenzoic acid (74.3 mg, 0.300 mmol) in CH2C12 (6 mL) was treated with HATU (0.114 g, 0.300 mmol) in the presence of DIPEA (75 μΐ^, 0.600 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a white solid (0.120 g, 93%). ESIMS m/z = 431.27 [M+H]+. Step l-356b. 6-bromo-N-methoxy-N-methyl-2-naphthamide (prepared according to J. Med. Chem., 2006, 49, 4721-4736, 3.57 g, 12.1 mmol) in THF (60 mL) was treated with methyl magnesium bromide (3M in Et20, 8.09 mL, 24.3 mmol) slowly at 0 °C for 1 hour. The solution was warmed up to rt for 2 hours before being quenched with aqueous NH4C1. The volatiles were removed and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated to give the crude desired compound as a white solid (2.89 g, 96%).
Step l-356c. The compound from step l-356b (2.89 g, 11.6 mmol) in acetic acid (60 mL) was treated with bromine (0.59 mL, 11.6 mmol) dropwise for 1 hour. The volatiles were evaporated and the residue was partitioned (EtOAc - saturated aqueous NaHC03). The organics were washed with brine, dried (Na2S04), filtered and evaporated to give the crude desired compound as a light yellow solid (3.898 g).
Step l-356d. A mixture of the compound from step l-356c (at most 11.6 mmol) and N- Boc-L-proline (3.75 g, 17.4 mmol) in CH3CN (60 mL) was added DIPEA (2.89 mL, 23.2 mmol) slowly. The mixture was stirred at rt until the disappearence of the starting material. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated to give the crude desired compound as a yellow-white foam (4.762 g). ESIMS m/z = 462.03, 464.02 [M+H]+.
Step l-356e. A mixture of the compound from step l-356d (0.200 g, 0.452 mmol), bis(pinaco-lato)diboron (0.144 g, 0.565 mmol), PdCl2(dppf)2 (36.9 mg, 0.0452 mmol) and potassium acetate (88.7 mg, 0.904 mmol) in DMSO (5 mL) was degassed and heated at 80°C under N2 for 17 hours. The reaction mixture was allowed to cool down and partitioned (EtOAc - water). The organic layer was washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by chromatography (silica, hexanes- ethyl acetate) to give the desired compound as a yellow solid (0.188 g, 85%). ESIMS m/z = 490.12 [M+H]+.
Step l-356f. A mixture of the compound from step l-356a (29.4 mg, 68.3 μιηοΐ), the compound from step l-356e (40.0 mg, 81.9 μιηοΐ) and NaHC03 (22.9 mg, 0.273 mmol) in DME (6 mL) and H20 (2 mL) was added Pd(PPh3)4 (3.9 mg, 3.4 μπιοΐ). The resultant mixture were degassed and heated to 85 °C under N2 for 16 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give a mixture of title compounds as a light yellow solid (42.2 mg, 93%). ESIMS m/z = 666.45 [M+H]+.
-357a and l-357b.
Figure imgf000163_0001
Step l-357a. A solution of the compound from example 1-356 (20.0 mg, 30.0 μιηοΐ) in CH2CI2 (4 mL) was treated with C1 in 1,4-dioxane (4 M, 3 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step. ESIMS m/z = 466.43 [M+H]+.
Step l-357b. A mixture of the crude compound from step l-357a (max. 30.0 μιηοΐ) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
2008/021927, 11.0 mg, 63.0 μιηοΐ) in DMF (3 mL) was treated with HATU (22.8 mg, 60.0 μιηοΐ) in the presence of DIPEA (75 μΐ^, 0.600 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, CH2Cl2-MeOH) to give a mixture of the title compounds as a white solid (16.0 mg, 2 steps 69%). ESIMS m/z = 780.72 [M+H]+.
Example 1-358.
Figure imgf000163_0002
Step l-358a. A mixture of N-Boc-L-proline (5.754 g, 26.7 mmol) and TEA (3.73 mL, 26.7 mmol) in THF (60 mL) at -20 °C was treated with ethyl chloroformate (2.55 mL, 26.7 mmol) for 30 minutes before a slow addition of 4-bromo-1,2-diaminobenzene (5.00 g, 26.7 mmol) in THF (20 mL). It was then kept at -20°C for 1 hour and then slowly warmed up to rt and stirred at rt overnight. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated to give the crude desired compound as a dark brown foam (10.7 g). ESIMS m/z = 384.18, 386.18 [M+H]+. Step l-358b. A solution of the crude compound from step l-358a (10.7 g, 26.7 mmol at most) in glacial acetic acid (100 mL) was heated at 50°C for 2 hours. The volatiles were evaporated off and the residue was partitioned (EtOAc - aqueous NaHC03). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a brown foam (5.78 g, 59%). ESIMS m/z = 366.17, 368.17 [M+H]+. 1H NMR (CDC13) 10.96, 10.93 (2 s, 1H), 7.81, 7.30 (2 s, 1H), 7.53, 7.17 (2d, J= 8.5 Hz, 1H), 7.23, 7.03 (2d, J= 8.5 Hz, 1H), 5.09, 5.07 (2s, 1H), 3.42-3.49 (m, 2H), 2.75-2.85 (m, 1H), 2.13-2.23 (m, 2H), 1.97-2.00 (m, 1H), 1.48 (s, 9H).
Step l-358c. A mixture of the compound from step l-358b (2.010 g, 5.488 mmol), trimethylsilyl-acetylene (2.33 ml, 16.46 mmol), Cul (0.110 g, 0.576 mmol) and
Pd(PPh3)2Cl2 (0.308 g, 0.439 mmol) in Et3N (50 mL) was degased and then heated at 80°C under N2 overnight before being evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate with 1% Et3N in ethyl acetate) to give the desired compound as a yellow foam (1.140 g, 54%). ESIMS m/z = 384.22 [M+H]+.
Step l-358d. A suspension of the compound from step l-358c (1.140 g, 2.972 mmol) and K2C03 (1.027 g, 7.430 mmol) in methanol (30 ml) was stirred at rt for 2 hour. The volatiles were evaporated off. The residue was patitioned (EtOAc - H20). The organic layer was washed with brine, dried (Na2S04), filtered and concentrated. The residue was purified by chromatography (silica, hexanes-ethyl acetate with 1% Et3N in ethyl acetate) to give the desired compound as a yellow foam (0.792 g, 86%>). ESIMS m/z = 312.18
[M+H]+.
Step l-358e. A mixture of the compound from step l-354a (52.6 mg, 0.143 mmol), the compound from step l-358d (48.8 mg, 0.157 mmol) in Et3N (6 mL) was added Cul (0.8 mg, 4.2 μιηοΐ) and Pd(PPh3)4 (16.4 mg, 14.2 μιηοΐ). The resultant mixture were degassed and heated to 85°C under N2 for 20 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the title compound as a light yellow solid (27.4 mg, 32%). ESIMS m/z = 600.45 [M+H]+. -359.
Figure imgf000165_0001
Step l-359a. A solution of the compound from example 1-358 (27.4 mg, 45.7 μιηοΐ) in CH2C12 (4 mL) was treated with C1 in 1,4-dioxane (4 M, 3 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step. ESIMS m/z = 400.35 [M+H]+.
Step l-359b. A mixture of the crude compound from step l-359a (max. 45.7 μιηοΐ) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
2008/021927, 16.8 mg, 95.9 μιηοΐ) in DMF (3 mL) was treated with HATU (34.7 mg, 91.4 μιηοΐ) in the presence of DIPEA (0.11 mL, 0.914 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, CH2Cl2-MeOH) to give the title compound as a light yellow solid (16.8 mg, 2 steps 52%). ESIMS m/z = 714.65 [M+H]+.
-360.
Figure imgf000165_0002
Step l-360a. A mixture of (S)-tert-butyl 2-(aminomethyl)pyrrolidine-1-carboxylate (0.109 g, 0.544 mmol) and 3-iodobenzoic acid (0.135 g, 0.544 mmol) in CH2C12 (6 mL) was treated with HATU (0.206 g, 0.544 mmol) in the presence of DIPEA (0.14 mL, 1.09 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a colorless oil (0.204 g, 87%). ESIMS m/z = 431.21 [M+H]+.
Step l-360b. A solution of the compound from step l-356d (0.120 g, 0.271 mmol) in 1,4- dioxane (3 mL) was treated with C1 in 1,4-dioxane (4 M, 6 mL) at rt for 1.5 hours. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was used directly in the next step.
Step l-360c. A mixture of the crude compound from step l-360b (0.271 mmol at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (47.5 mg, 0.271 mmol) in DMF (2 mL) was treated with HATU (98.0 mg, 0.258 mmol) in the presence of DIPEA (0.47 mL, 2.713 mmol) for 1.5 hours at rt and the volatiles were evaporated off . The residue was patitioned (EtOAc/CH2Cl2 - H20). The organic layer was washed with saturated NaHC03, brine, dried (Na2S04), filtered and concentrated. The crude product was purified by flash column chromatography (silica, hexanes-ethyl acetate with 1% Et3N in ethyl acetate) to give the desired compound as a light yellow solid (0.132 g, 97% over 2 steps). ESIMS m/z = 498.86, 500.85 [M+H]+.
Step l-360d. A mixture of the compound from step l-360c (0.130 g, 0.260 mmol), bis(pinacolato)diboron (79.3 mg, 0.312 mmol), PdCl2(dppf)2 (21.3 mg, 26.0 μιηοΐ) and potassium acetate (51.0 mg, 0.521 mmol) in DMSO (4 mL) was degassed and heated at 80 °C under N2 overnight. The mixture was allowed to cool down and partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate with 1% Et3N in ethyl acetate) to give the desired compound as a yellow foam (0.105 g, 74%). ESIMS m/z = 547.08 [M+H]+.
Step l-360e. A mixture of the compound from step l-360a (18.4 mg, 43.9 μιηοΐ), the compound from step 360d (20.0 mg, 36.6 μιηοΐ) and NaHC03 (12.3 mg, 0.146 mmol) in DME (6 mL) and H20 (2 mL) was added Pd(PPh3)4 (2.1 mg, 1.8 μπιοΐ). The resultant mixture were degassed and heated to 85°C under N2 for 16 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the title compound as a colorless oil (32.6 mg, ca. 100%). ESIMS m/z = 723.45 [M+Hf.
-361.
Figure imgf000166_0001
The title compound was prepared from the compound of example v360 using procedures similar to that described in example 1-359. ESIMS m/z = 780.75 [M+H]+.
-362.
Figure imgf000166_0002
Step l-362a. A mixture of N-Boc-L-proline (5.754 g, 26.7 mmol) and TEA (3.73 mL, 26.7 mmol) in THF (60 mL) at -20°C was treated with ethyl chloroformate (2.55 mL, 26.7 mmol) for 30 minutes before a slow addition of 4-bromo-1,2-diaminobenzene (5.00 g, 26.7 mmol) in THF (20 mL). It was then kept at -20 °C for 1 hour and then slowly warmed up to rt and stirred at rt overnight. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated to give the crude desired compound as a dark brown foam (10.7 g). ESIMS m/z = 384.18, 386.18 [M+H]+.
Step l-362b. A solution of the crude compound from step l-362a (10.7 g, 26.7 mmol at most) in glacial acetic acid (100 mL) was heated at 50°C for 2 hours. The volatiles were evaporated off and the residue was partitioned (EtOAc - aqueous NaHC03). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a brown foam (5.78 g, 59%). ESIMS m/z = 366.17, 368.17 [M+H]+. 1H NMR (CDC13) 10.96, 10.93 (2 s, 1H), 7.81, 7.30 (2 s, 1H), 7.53, 7.17 (2d, J= 8.5 Hz, 1H), 7.23, 7.03 (2d, J= 8.5 Hz, 1H), 5.09, 5.07 (2s, 1H), 3.42-3.49 (m, 2H), 2.75-2.85 (m, 1H), 2.13-2.23 (m, 2H), 1.97-2.00 (m, 1H), 1.48 (s, 9H).
Step l-362c. A mixture of the compound from step l-362b (1 g, 2.73 mmol), bis- (pinacolato)-diboron (763 mg, 3.0 mmol), potassium acetate (402 mg, 4.0 mmol) in 1,4- dioxane (9.1 mL) was added tetrakis(triphenylphosphine)palladium(0) (158 mg, 0.14 mmol). The resulting solution was degased and then heated at 80°C under N2 overnight before being evaporated. The residue was purified by chromatography (silica, hexanes- ethyl acetate with 1% Et3N in ethyl acetate) to give the desired compound as a yellow solid (680 mg, 60%). ESIMS m/z = 414.24 [M+H]+.
Step l-362d. A mixture of the compound from step l-354a (20.0 mg, 54.2 μιηοΐ), the compound from step l-362c (24.6 mg, 59.6 μιηοΐ) and NaHC03 (18.2 mg, 0.217 mmol) in DME (6 mL) and H20 (2 mL) was added Pd(PPh3)4 (3.1 mg, 2.7 μπιοΐ). The resultant mixture were degassed and heated to 90°C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the title compound as a colorless oil (29.3 mg, 93%). ESIMS m/z = 576.41 [M+H]+. -363.
Figure imgf000168_0001
The title compound was prepared from the compound of example 1-362 using procedures similar to that described in example 1-359. ESIMS m/z = 690.54 [M+H]+.
-364.
Figure imgf000168_0002
Step l-364a. A mixture of (な)-l-N-Boc-¾eto-proline (0.100 g, 0.465 mmol) and 2-amino- 6-bromonaphthalene (0.103 g, 0.465 mmol) in CH2CI2 (6 mL) was treated with HATU (0.177 g, 0.465 mmol) in the presence of DIPEA (0.12 mL, 0.929 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a white solid (0.191 g, 98%).
Step l-364b. A mixture of the compound from step l-364a (30.0 mg, 71.5 μιηιηοΐ), the compound from step l-348d (34.5 mg, 78.6 μιηιηοΐ) and NaHC03 (24.0 mg, 0.286 mmol) in DME (6 mL) and H20 (2 mL) was added Pd(PPh3)4 (4.0 mg, 3.5 μπιοΐ). The resultant mixture were degassed and heated to 95°C under N2 for 16 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the title compound as a white solid (41.6 mg, 89%). ESIMS m/z = 652.28 [M+H]
-365.
Figure imgf000168_0003
The title compound was prepared from the compound of example 1-364 using procedures similar to that described in example 1-359. ESIMS m/z = 766.37 [M+H]+. -366.
Figure imgf000169_0001
A mixture of the compound from step l-364a (30.0 mg, 71.5 μιηιηοΐ), the compound from step l-362c (32.4 mg, 78.6 μιηιηοΐ) and NaHC03 (24.0 mg, 0.286 mmol) in DME (6 mL) and H20 (2 mL) was added Pd(PPh3)4 (4.0 mg, 3.5 μιηοΐ). The resultant mixture were degassed and heated to 95°C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column
chromatography (silica, hexanes-ethyl acetate) to give the title compound as a white solid (40.2 mg, 90%). ESIMS m/z = 626.36 [M+H]+.
-367a and l-367b.
Figure imgf000169_0002
Step l-367a. To a suspension of trans- 1 ,2-cyclopentanediamine dihydrochloride
(racemic, 0.100 g, 0.578 mmol) in THF (6 mL) at 0°C was added NaH (60% in mineral oil, 46.2 mg, 1.155 mmol). After 5 minutes at 0°C, the milky solution was stirred at room temperature for 15 minutes before 9-BBN (0.5 M in THF, 1.15 mL, 0.578 mmol) was added. After 1 hour at room temperature, a solution of 4-iodobenzoyl chloride (0.146 g, 0.548 mmol) in THF (1.2 mL) was added. The suspension was stirred at room temperature for 2 hr being quenched carefully with H20. The mixture was partitioned (EtOAc - aqueous NaHC03). The organiclayer was washed with brine, dried (Na2S04), filtered and evaporated to give the crude product as a mixture of monoamidation and diamidation products as a white semi-solid, which was used directly for next step.
Step l-367b. A mixture of the crude compound from step l-367a (0.578 mmol at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 101.2 mg, 0.578 mmol) in DMF (3 mL) was treated with HATU (0.220 g, 0.578 mmol) in the presence of DIPEA (1.00 mL, 5.778 mmol) for 1 hours at room temperature and the volatiles were evaporated off. The residue was taken up in
CH2Cl2/MeOH (4/1), filtered and concentrated. The residue was purified by
chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a white solid (17.0 mg). ESIMS m/z = 488.07 [M+H]+.
Step l-367c. A mixture of compound from step l-367b (17.0 mg, 34.9 μιηοΐ), the compound from step 352f (17.2 mg, 34.9 μιηοΐ), Pd(PPh3)4 (4.0 mg, 3.49 μιηοΐ) and NaHC03 (10.3 mg, 0.122 mmol) in DME (2.1 mL) and H20 (0.7 mL) was degassed and heated at 97 °C under N2 for 15 hours. The mixture was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified first by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N and 20% MeOH in EtOAc) and then by preparative TLC (CH2Cl2/MeOH 4/1) to give a mixture the title compounds as an off-white solid (8.4 mg, 33%). ESIMS m/z = 730.28 [M+H]+.
Example 1-368.
Figure imgf000170_0001
Step l-368a. A mixture of (IR, 2i?)-trans-N-Boc-1,2-cyclopentanediamine (80.0 mg, 0.399 mmol) and 4-iodobenzoic acid (100.1 mg, 0.403 mmol) in CH3CN (7 mL) was treated with HATU (0.152 g, 0.399 mmol) in the presence of DIPEA (0.70 mL, 3.994 mmol) for 1 hours at rt and the volatiles were evaporated off. The residue was purified by chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a white solid (0.160 g, 93%). ESIMS m/z = 331.10 [M+H]+.
Step l-368b. A mixture of compound from step l-368a (45.0 mg, 0.105 mmol), the compound from step l-348d (46.0 mg, 0.105 mmol), Pd(PPh3)4 (12.1 mg, 10.5 μιηοΐ) and NaHC03 (30.8 mg, 0.366 mmol) in DME (3 mL) and H20 (1 mL) was degassed and heated at 97°C under N2 for 15 hours. The mixture was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N and 10% MeOH in EtOAc) to give the title compound as an off-white solid (63.2 mg, 98%). ESIMS m/z = 616.36 [M+H]+. -369.
Figure imgf000171_0001
Step l-369a. A solution of the compound of example 1-368 (63.2 mg, 0.103 mmol) in 1,4-dioxane (1.25 mL) was treated with C1 in 1,4-dioxane (4 M, 5 mL) at 30°C for 30 minutes. The volatiles were evaporated off to give the crude desired compound as a yellow solid, which was used directly in the next step.
Step l-368b. A mixture of the crude compound from step l-369a (0.103 mmol at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 36.0 mg, 0.205 mmol) in DMF (3 mL) was treated with HATU (74.1 mg, 0.195 mmol) in the presence of DIPEA (0.36 mL, 2.053 mmol) for 30 minutes at rt and the volatiles were evaporated off. It was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N and 20% MeOH in EtOAc) to give the title compound as a yellow solid (56.0 mg, 75% over 2 steps). ESIMS m/z = 730.45 [M+H]+.
Examples l-370a and l-370b.
Figure imgf000171_0002
Step l-370a. A mixture of tran -N-Boc-1,2-cyclopentanediamine (60.0 mg, 0.300 mmol) and 4-iodobenzoic acid (75.0 mg, 0.303 mmol) in CH3CN (5 mL) was treated with HATU (0.114 g, 0.300 mmol) in the presence of DIPEA (0.52 mL, 3.000 mmol) for 1 hours at rt and the volatiles were evaporated off. The residue was purified by chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a white solid (0.119 g, 92%>). ESIMS m/z = 331.10 [M+H]+.
Step l-370b. A mixture of the compound from step l-362c (0.250 g, 0.605 mmol), 1- bromo-4-iodobenzene (0.257 g, 0.908 mmol), NaHC03 (0.203 g, 2.42 mmol) and
Pd(PPh3)4 (34.9 mg, 30.2 μιηοΐ) in DME (12 mL) and water (4 mL) was degassed and heated to 85°C under N2 overnight. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a very light yellow solid (0.246 g, 92%). ESIMS m/z = 442.00, 444.00 [M+H]+.
Step l-370c. To a mixture of the compound from step l-370b (50.0 mg, 0.113 mmol), bis-(pinacolato)-diboron (35.9 mg, 0.141 mmol), potassium acetate (22.2 mg, 0.226 mmol) in DMSO (2 mL) was added PdCl2(dppf)2 (9.2 mg, 11.3 μιηοΐ). The resulting solution was degased and then heated at 80 °C under N2 overnight before being partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate with 1% Et3N in ethyl acetate) to give the desired compound as a colorless oil (36.4 mg, 66%). ESIMS m/z = 490.37 [M+H]+.
Step l-370d. A mixture of compound from step l-370a (32.0 mg, 74.4 μιηοΐ), the compound from step l-370c (36.4 mg, 74.4 μιηοΐ), Pd(PPh3)4 (8.6 mg, 7.4 μιηοΐ) and NaHC03 (21.9 mg, 0.260 mmol) in DME (2.1 mL) and H20 (0.7 mL) was degassed and heated at 97 °C under N2 for 15 hours. The mixture was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N and 10% MeOH in EtOAc) to give a mixture of the title compounds as an off-white solid (31.7 mg, 64%). ESIMS m/z = 666.37 [M+H]+.
-37 la and 1-37 lb.
Figure imgf000172_0001
Step l-371a. A solution of the compound of example 1-370 (31.7 mg, 47.6 μιηοΐ) in 1,4- dioxane (1 mL) was treated with C1 in 1,4-dioxane (4 M, 4 mL) at 30 °C for 30 minutes. The volatiles were evaporated off to give the crude desired compound as a yellow solid, which was used directly in the next step.
Step l-371b. A mixture of the crude compound from step 1-37 la (47.6 μιηοΐ at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 16.7 mg, 95.2 μιηοΐ) in DMF (3 mL) was treated with HATU (34.4 mg, 90.5 μιηοΐ) in the presence of DIPEA (0.17 mL, 0.952 mmol) for 30 minutes at rt and the volatiles were evaporated off. The residue was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N and 20% MeOH in EtOAc) to give a mixture of title compounds as a white solid (31.6 mg, 85% over 2 steps). ESIMS m/z = 780.52 [M+H]+.
-372.
Figure imgf000173_0001
Step l-372a. A mixture of (5)-3-(Boc-amino)pyrrolidine (0.150 g, 0.805 mmol) and (S)- 2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO
2008/021927, 0.148 g, 0.846 mmol) in DMF (3 mL) was treated with HATU (0.306 g, 0.805 mmol) in the presence of DIPEA (1.40 mL, 8.05 mmol) for 1 hours at rt and the volatiles were evaporated off. The residue was purified by chromatography (silica, CH2Cl2-MeOH) to give the desired compound as a yellow oil (0.680 g). ESIMS m/z = 344.21 [M+H]+.
Step l-372b. A solution of the compound from step l-372a (0.680 g, 0.805 mmol at most) in 1,4-dioxane (1.5 mL) was treated with C1 in 1,4-dioxane (4 M, 6 mL) at rt for 1 hours. The volatiles were evaporated off to give the crude desired compound as a yellow oil, which was used directly in the next step. ESIMS m/z = 244.17 [M+H]+.
Step l-372c. A mixture of the crude compound from step l-372b (0.805 mmol at most) and 4-iodobenzoic acid (0.200 g, 0.805 mmol) in CH3CN (4 mL) was treated with HATU (0.306 g, 0.805 mmol) in the presence of DIPEA (1.40 mL, 8.05 mmol) for 1 hours at rt and the volatiles were evaporated off. The residue was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N in EtOAc) to give the desired compound as a white foam (0.232 g, 61% over 3 steps). ESIMS m/z = 474.07 [M+H]+.
Step l-372d. A mixture of compound from step l-372c (30.0 mg, 63.4 μιηοΐ), the compound from step l-352f (40.9 mg, 82.4 μιηοΐ), Pd(PPh3)4 (7.3 mg, 6.34 μιηοΐ) and NaHC03 (21.3 mg, 0.254 mmol) in DME (3 mL) and H20 (1 mL) was degassed and heated at 98 °C under N2 for 2.5 hours. The mixture was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N and 20% MeOH in EtOAc) to give the title compound as a light brown solid (42.7 mg, 94%).
ESIMS m/z = 716.27 [M+H]+. -373.
Figure imgf000174_0001
Step l-373a. A mixture of (i?)-N-Boc-pyrrolidine-3-carboxylic acid (1.000 g, 4.646 mmol) and 4-bromoaniline (0.799 g, 4.646 mmol) in CH2CI2 (20 mL) was treated with HATU (1.766 g, 4.646 mmol) in the presence of DIPEA (1.62 mL, 9.292 mmol) for 1 hours at rt and the volatiles were evaporated off. The residue was purified by
chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a white foam (1.699 g, 99%).
Step l-373b. A mixture of compound from step l-373a (0.100 g, 0.271 mmol), the compound from step l-348d (0.131 g, 0.298 mmol), Pd(PPh3)4 (31.2 mg, 27.1 μπιοΐ) and NaHC03 (91.0 mg, 1.083 mmol) in DME (3 mL) and H20 (1 mL) was degassed and heated at 97°C under N2 for 15 hours. The mixture was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N in EtOAc) to give the title compound as a white solid (0.111 g, 68%). ESIMS m/z = 602.30 [M+H]+.
-374.
Figure imgf000174_0002
Step l-374a. A solution of the compound of example 1-373 (0.111 g, 0.184 mmol) in 1,4- dioxane (2 mL) was treated with C1 in 1,4-dioxane (4 M, 6 mL) at room temperature for 1 hour. The volatiles were evaporated off to give the crude desired compound as a yellow solid, which was used directly in the next step.
Step l-374b. A mixture of the crude compound from step l-374a (0.184 mmol at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 64.6 mg, 0.369 mmol) in DMF (4 mL) was treated with HATU (0.133 g, 0.351 mmol) in the presence of DIPEA (0.64 mL, 3.689 mmol) for 1 hour at room temperature and the volatiles were evaporated off. The residue was purified by
chromatography (silica, hexanes-ethyl acetate, with 1% Et3N and 20% MeOH in EtOAc) to give the title compound as a light yellow solid (0.122 g, 92% over 2 steps). ESIMS m/z = 716.37 [M+H]+.
Figure imgf000175_0001
Step l-375a. A mixture of trans-aminocyclopentanol hydrochloride (racemic, 0.100 g, 0.727 mmol) and 4-iodobenzoic acid (0.180 g, 0.727 mmol) in CH3CN (10 mL) was treated with HATU (0.276 g, 0.727 mmol) in the presence of DIPEA (1.27 mL, 7.266 mmol) for 30 minutes at rt and the volatiles were evaporated off. The residue was purified by chromatography (silica, hexanes-ethyl acetate with 1% EtOH) to give the desired compound as a white solid (0.226 g, 94%). ESIMS m/z = 332.04 [M+H]+.
Step l-375b. A mixture of compound from step l-375a (45.0 mg, 0.136 mmol), the compound from step l-348d (46.0 mg, 0.105 mmol), Pd(PPh3)4 (15.7 mg, 13.6 μπιοΐ) and NaHC03 (45.7 mg, 0.544 mmol) in DME (3 mL) and H20 (1 mL) was degassed and heated at 97°C under N2 for 15 hours. The mixture was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N and 1% EtOH in EtOAc) to give a mixture of title compounds as a white solid (54.2 mg, 77%). ESIMS m/z = 517.31 [M+H]+.
Examples l-376a and l-376b.
Figure imgf000175_0002
Step l-376a. A solution of the compound of example 1-375 (54.2 mg, 0.105 mmol) in 1,4-dioxane (1.25 mL) was treated with C1 in 1,4-dioxane (4 M, 5 mL) at room temperature for 1 hour. The volatiles were evaporated off to give the crude desired compound as a yellow solid, which was used directly in the next step.
Step l-376b. A mixture of the crude compound from step l-376a (0.103 mmol at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 36.8 mg, 0.210 mmol) in DMF (4 mL) was treated with HATU (37.9 mg, 0.100 mmol) in the presence of DIPEA (0.18 mL, 1.049 mmol) for 15 minutes at rt and the volatiles were evaporated off. It was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N and 15% MeOH in EtOAc) to give a mixture of title compounds as a white solid (50.1 mg, 83% over 2 steps). ESIMS m/z = 574.35 [M+H]+.
-377a and l-377b.
Figure imgf000176_0001
To a solution of (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to the method described in WO 2008/021927, 17.8 mg, 0.138 mmol) and Et3N (19.2 μΐ, 0.138 mmol) in THF (2 mL) at -20 °C was added z-butyl chloroformate (17.9 μΐ, 0.138 mmol) in THF (0.1 mL). After 20 minutes at -20 °C, a solution of the compound of example 1-376 (36.0 mg, 62.8 μιηοΐ) in THF (1 mL) was added. After 15 minutes at - 20°C, the mixture was stirred at room temperature overnight. The volatiles were evaporated off. The residue was purified by HPLC to give a mixture of the title compounds as a white solid (8.2 mg, 18%). ESIMS m/z = 731.40 [M+H]+.
Example 1-378.
Figure imgf000176_0002
Step l-378a. A mixture of (5)-N-Boc-pyrrolidine-3-carboxylic acid (80.0 mg, 0.372 mmol) and 2-amino-5-bromopyridine (64.3 mg, 0.372 mmol) in CH2CI2 (3 mL) was treated with HATU (0.141 g, 0.372 mmol) in the presence of DIPEA (0.13 mL, 0.743 mmol) for 1 hours at rt. More (5)-N-Boc-pyrrolidine-3-carboxylic acid (40.0 mg, 0.186 mmol), DIPEA (0.065 mL, 0.372 mmol) and HATU (70.5 mg, 0.186 mmol) were added. The mixture was heated at 100 °C with a microwave for 30 minutes. The volatiles were evaporated off. The residue was purified by chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a white foam (87.3 mg, 63%>).
Step l-378b. A mixture of compound from step l-378a (48.0 mg, 0.130 mmol), the compound from step l-370c (62.7 mg, 0.143 mmol), Pd(PPh3)4 (15.0 mg, 13.0 μιηοΐ) and NaHC03 (38.1 mg, 0.454 mmol) in DME (3 mL) and H20 (1 mL) was degassed and heated at 97°C under N2 for 15 hours. The volatiles were evaporated off. The residue was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N in EtOAc) to give the title compound as a yellow solid (61.8 mg, 79%). ESIMS m/z = 603.15 [M+H]+.
-379.
Figure imgf000177_0001
Step l-379a. A solution of the compound of example 1-378 (61.8 mg, 0.102 mmol) in 1,4-dioxane (1 mL) was treated with C1 in 1,4-dioxane (4 M, 4mL) at room temperature for 1.5 hours. The volatiles were evaporated off to give the crude desired compound as a yellow solid, which was used directly in the next step.
Step l-379b. A mixture of the crude compound from step l-379a (0.102 mmol at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 35.9 mg, 0.205 mmol) in DMF (2 mL) was treated with HATU (74.1 mg, 0.195 mmol) in the presence of DIPEA (0.36 mL, 2.051 mmol) for 15 minutes at room temperature and the volatiles were evaporated off. The residue was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N and 20% MeOH in EtOAc) to give the title compound as a light yellow solid (62.4 mg, 85% over 2 steps). ESIMS m/z = 717.15 [M+H]+.
-380.
Figure imgf000177_0002
The title compound was prepared from (R)-tert-butyl 2-(aminomethyl)pyrrolidine-1- carboxylate using procedures similar to that described in example 1-257. ESIMS m/z = 730.43 [M+H]+.
-381.
Figure imgf000177_0003
Step l-381a. Into a mixture of (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 90 mg, 0.5 mmol) and (R)-tert-butyl 2- (aminomethyl)pyrrolidine-1-carboxylate (100 mg, 0.50 mmol) in acetonitrile (2 mL) was added diisopropylethylamine (0.11 mL, 0.6 mmol) and HATU (200 mg, 0.55 mmol). The resulting mixture was stirred at room temperature for 1 hour before being partitioned between water and EtOAc. The organic phase was separated, dried (Na2S04) and concentrated to afford a brown slurry, which was purified by flash column
chromatography (silica, EtOAc-hexanes) to give the desired product as a light yellow solid (144 mg, 80%). ESIMS m/z = 358.41 [M+H]+.
Step l-381b. Into a mixture of compound firm step l-381a (144 mg, 0.4 mmol) was added hydrochloric acid in 1,4-dioxane (4M, 2 mL). The resulting mixture was stirred at room temperature for 1 hour before all volatiles were removed to afford the crude desired compound, which was used directly for the next step without further purification. ESIMS m/z = 258.28 [M+H]+.
Step l-381c. The title compound was prepared from 4-iodobenzoic acid and the compound from step l-348d using procedures similar to that described in example 1-348. ESIMS m/z = 730.45 [M+H]+.
-382a and l-382b.
Figure imgf000178_0001
A mixture of the title compounds was prepared from l-(tert- butoxycarbonyl)piperidine-3-carboxylic acid using procedures similar to that described examples 1-354 and 1-355. ESIMS m/z = 730.32 [M+H]+.
The compounds of examples 2-1 to 2-293 may be prepared using procedures similar to those described in examples 2-294 and 2-295 (described below), and/or as described in the Synthetic Methods.
Examples 2-1 to 2-219.
Figure imgf000178_0002
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Step 2-294a. A mixture of l-Boc-pyrrolidine-3-carboxylic acid (0.100 g, 0.465 mmol) and 4-bromoaniline (79.9 mg, 0.465 mmol) in CH2CI2 (6 mL) was treated with HATU (0.177 g, 0.465 mmol) in the presence of DIPEA (0.12 mL, 0.929 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a colorless oil (0.156 g, 91%). ESIMS m/z = 369.21 , 371.25 [M+H]+. Step 2-294b. A mixture of the compound from step 2-294a (0.184 g, 0.499 mmol), bis- (pinacolato) diboron (0.253 g, 0.997 mmol) and potassium acetate (0.122 g, 1.25 mmol) in 1,4-dioxane (8 mL) was added Pd(PPh3)4 (28.8 mg, 24.9 μιηοΐ). The resultant mixture were degassed for three times and heated up to 85°C under N2 for 13 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a white solid (0.142 g, 68%). ESIMS m/z = 439.32 [M+Na]+.
Step 2-294c. A mixture of the compound from step 2-294a (30.0 mg, 81.3 μιηοΐ), the compound from step 2-294b (40.6 mg, 97.5 μιηοΐ) and NaHC03 (27.3 mg, 0.325 mmol) in DME (6 mL) and H20 (2 mL) was added Pd(PPh3)4 (4.6 mg, 4.0 μπιοΐ). The resultant mixture were degassed and heated to 85°C under N2 for 16 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the title compound as a white solid (23.5 mg, 50%). ESIMS m/z = 579.09 [M+H]+.
-295.
Figure imgf000187_0001
Step 2-295a. A solution of the compound from example 2-294 (23.5 mg, 40.6 μιηοΐ) in CH2C12 (4 mL) was treated with C1 in 1 ,4-dioxane (4 M, 3 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step. ESIMS m/z = 379.35 [M+H]+.
Step 2-295b. A mixture of the crude compound from step 2-295 a (40.6 μιηοΐ at most) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
2008/021927, 14.9 mg, 85.2 μιηοΐ) in DMF (3 mL) was treated with HATU (30.9 mg, 81.2 μιηοΐ) in the presence of DIPEA (0.10 mL, 0.812 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, CH2Cl2-MeOH) to give the title compound as a white solid (22.2 mg, 2 steps 79%) and diastereomeric mixture. ESIMS m/z = 693.39 [M+H]+. The compounds of examples 3-1 to 3-335 may be prepared using procedures similar to those described in examples 3-336 to 347 (described below), and/or as described in the Synthetic Methods.
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
Step 3-336a. A mixture of 2,4'-dibromoacetophenone (1.00 g, 3.60 mmol) and trans (+/-) 2-(fert-butoxycarbonylamino)-cyclopentanecarboxylic acid (0.825 g, 3.60 mmol) in CH3CN (12 mL) was added DIPEA (0.94 mL, 7.55 mmol) slowly. The mixture was stirred at room temperature until the disappearence of the starting material. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a white solid (1.548 g, 100%). ESIMS m/z = 426.03, 428.03 [M+H]+.
Step 3-336b. A solution of the compound from step 3_-336a (1.54 g, 3.60 mmol) in toluene (20 mL) was added ammonium acetate (5.54 g, 71.9 mol) and the resultant mixture was heated up to 100 °C for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - aq. NaHC03). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give intermediate 1 as a yellow brown solid (1.34 g, 91%). ESIMS m/z = 406.09, 408.09 [M+H]+.
Step 3-336c. A mixture of 2,4'-dibromoacetophenone (5.00 g, 18.0 mmol) and N-Boc-L- proline (3.87 g, 18.0 mmol) in CH3CN (60 mL) was added TEA (5.40 mL, 37.8 mmol) slowly. The mixture was stirred at room temperature until the disappearence of the starting material. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a light yellow foam (6.73 g, 91 >). 1H NMR (CDC13) 7.76 (t, J= 8.0 Hz, 2H), 7.63 (dd, J= 5.0, 8.5 Hz, 2H), 5.51, 5.16 (2d, J= 16.0 Hz, 1H), 5.32, 5.28 (2d, J= 16.5 Hz, 1H), 4.48, 4.40 (dd, J= 5.0, 8.5 Hz, 1H), 3.56 (m, 1H), 3.43 (m, 1H), 2.30 (m, 2H), 2.06 (m, 1H), 1.92 (m, 1H), 1.46, 1.43 (2s, 9H).
Step 3-336d. A solution of the compound from step 3-336c (6.73 g, 16.3 mmol) in toluene (100 mL) was added ammonium acetate (25.1 g, 0.327 mol) and the resultant mixture was heated up to 100°C for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - aq. NaHC03). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a yellow foam (6.10 g, 95%). ESIMS m/z = 392.24, 394.24 [M+H]+. 1H NMR (CDC13) 7.57 (bs, 1H), 7.48 (m, 3H), 7.23 (s, 1H), 4.97 (m, 1H), 3.42 (m, 2H), 2.99 (m, 1H), 2.16 (m, 2H), 1.97 (m, 1H), 1.46 (s, 9H).
Step 3-336e. A mixture of the compound from step 3-336d (1.00 g, 2.55 mmol), bis-
(pinacolato) diboron (1.35 g, 5.33 mmol) and potassium acetate (0.640 g, 6.53 mmol) in 1,4-dioxane (20 mL) was added Pd(PPh3)4 (0.147 g, 0.128 mmol). The resultant mixture were degassed for three times and heated up to 80°C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give intermediate 2 as a light yellow solid (0.978 g, 87%). ESIMS m/z = 440.39 [M+H]+. 1H NMR (CDC13) 11.03, 10.55 (2s, 1H), 7.79 (m, 3H), 7.45 (m, 1H), 7.26 (m, 1H), 4.97 (m, 1H), 3.41 (m, 2H), 3.06, 2.91 (2m, 1H), 2.17 (m, 2H), 1.97 (m, 1H), 1.49 (s, 9H), 1.35 (s, 12H).
Step 3-336f. A mixture of the compound from step 3_ 336b (0.315 g, 0.777 mmol), the compound from step 3^3366 (0.310 g, 0.706 mmol) and NaHC03 (0.237 g, 2.82 mmol) in DME (12 mL) and H20 (4 mL) was added Pd(PPh3)4 (40.8 mg, 35.3 μπιοΐ). The resultant mixture were degassed and heated to 85°C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give a mixture of the title compounds as a light yellow solid (0.326 g, 72%). ESIMS m/z = 639.34 [M+H]+.
Examples 3 -337a and 3 -337b.
Figure imgf000198_0001
Step 3-337a. A solution of the compound of example 3-336 (80.0 mg, 0.125 mmol) in CH2C12 (3 mL) was treated with C1 in 1,4-dioxane (4 M, 6 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step. ESIMS m/z = 439.23 [M+H]+.
Step 3-337b. A mixture of the crude compound from step 3^337a (theo 0.125 mmol) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
2008/021927, 46.0 mg, 0.263 mmol) in DMF (3 mL) was treated with HATU (95.2 mg, 0.251 mmol) in the presence of DIPEA (0.31 mL, 2.51 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, CH2Cl2-MeOH) to give a mixture of the title compounds as a light yellow solid (52.6 mg, 2 steps 56%). ESIMS m/z = 753.41 [M+H]+. -338a and 3-338b.
Figure imgf000199_0001
Step 3-338a. A mixture of N-Boc-L-proline (5.754 g, 26.7 mmol) and TEA (3.73 mL, 26.7 mmol) in THF (60 mL) at -20°C was treated with ethyl chloroformate (2.55 mL, 26.7 mmol) for 30 minutes before a slow addition of 4-bromo-1,2-diaminobenzene (5.00 g, 26.7 mmol) in THF (20 mL). It was then kept at -20 °C for 1 hour and then slowly warmed up to rt and stirred at rt overnight. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated to give the crude desired compound as a dark brown foam (10.7 g). ESIMS m/z = 384.18, 386.18 [M+H]+.
Step 3-338b. A solution of the crude compound from step 3_ 338a (10.7 g, theo. 26.7 mmol) in glacial acetic acid (100 mL) was heated at 50°C for 2 hours. The volatiles were evaporated off and the residue was partitioned (EtOAc - saturated aqueous NaHC03). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give intermediate 3 as a brown foam (5.78 g, 59%). ESIMS m/z = 366.17, 368.17 [M+H]+. 1H NMR (CDCls) 10.96, 10.93 (2 s, 1H), 7.81, 7.30 (2 s, 1H), 7.53, 7.17 (2d, J= 8.5 Hz, 1H), 7.23, 7.03 (2d, J= 8.5 Hz, 1H), 5.09, 5.07 (2s, 1H), 3.42-3.49 (m, 2H), 2.75-2.85 (m, 1H), 2.13-2.23 (m, 2H), 1.97-2.00 (m, 1H), 1.48 (s, 9H).
Step 3-338c. A mixture of the compound from 3_2338b (0.559 g, 1.425 mmol), trimethylsilyl acetylene (0.60 ml, 4.275 mmol), Cul (28.5 mg, 0.150 mmol) and
Pd(PPh3)2Cl2 (80.0 mg, 0.114 mmol) in Et3N (15 mL) was evacuated and refilled with N2 three times while being cooled with an aceton-dry ice bath. The mixture was then heated at 80°C under N2 for 6 hr before being evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate with 1% Et3N in ethyl acetate) to give the desired compound as a yellow foam (0.484 g, 83%). ESIMS m/z = 410.24
[M+H]+. Step 3-338d. A suspension of the compound from step 3^338ΰ (0.484 g, 1.182 mmol) and K2CO3 (0.408 g, 2.954 mmol) in methanol (12 ml) was stirred at rt for 3 hr. The volatiles were evaporated off. The residue was purified by flash column chromatography (silica, dichloromethane-ethyl acetate) to give intermediate 4 as a yellow foam (0.370 g, 93%). ESIMS m/z = 338.24 [M+H]+.
Step 3-338e. A mixture of the compound from step 3^336b (150 mg, 0.369 mmol), the compound from step 3^338(1 (127 mg, 0.406 mmol) in Et3N (4 mL) and CH3CN (4 mL) were added Cul (2.1 mg, 11.0 μιηοΐ) and Pd(PPh3)4 (21.3 mg, 18.4 μιηοΐ). The resultant mixture were degassed and heated to 85°C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give a mixture of the title compounds as a light yellow solid (0.137 g, 58%). ESIMS m/z = 637.32 [M+H]+.
-339a and 3 -339b.
Figure imgf000200_0001
Step 3-339a. A solution of the compound of example 3^338 (0.137 g, 0.215 mmol) in CH2C12 (3 mL) was treated with C1 in 1,4-dioxane (4 M, 6 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step. ESIMS m/z = 437.20 [M+H]+.
Step 3-339b. A mixture of the crude compound from step 3_-339a (theo 0.215 mmol) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
2008/021927, 79.2 mg, 0.452 mmol) in DMF (3 mL) was treated with HATU (0.164 g, 0.431 mmol) in the presence of DIPEA (0.54 mL, 4.31 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, CH2Cl2-MeOH) to give a mixture of the title compounds as a light yellow solid (0.144 g, 2 steps 89%). ESIMS m/z = 751.34 [M+H]+. Examples 3-340a and 3-340b.
Figure imgf000201_0002
Step 3-340a. A mixture of the compound from step 3_ 338b (1 g, 2.73 mmol), bis- (pinacolato)-diboron (763 mg, 3.0 mmol), potassium acetate (402 mg, 4.0 mmol) in 1,4- dioxane (9.1 mL) was added tetrakis(triphenylphosphine)palladium(0) (158 mg, 0.14 mmol). The resulting solution was degased and then heated at 80°C under N2 overnight before being evaporated. The residue was purified by chromatography (silica, hexanes- ethyl acetate with 1% Et3N in ethyl acetate) to give the desired compound as a yellow solid (680 mg, 60%). ESIMS m/z = 414.24 [M+H]+.
Step 3-340b. A mixture of the compound from step 32338b (0.100 g, 0.246 mmol), the compound from step 3-340a (0.102 g, 0.246 mmol) and NaHC03 (82.8 mg, 0.985 mmol) in DME (6 mL) and H20 (2 mL) was added Pd(PPh3)4 (14.2 mg, 12.3 μπιοΐ). The resultant mixture were degassed and heated to 85°C under N2 for 20 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give a mixture of the title compounds as a light yellow solid (0.112 g, 75%). ESIMS m/z = 613.17 [M+H]+.
-34 la and 3-34 lb.
Figure imgf000201_0001
Step 3-341a. A solution of the compound from example 3-340 (0.112 g, 0.183 mmol) in CH2C12 (3 mL) was treated with C1 in 1,4-dioxane (4 M, 2 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step. ESIMS m/z = 413.23 [M+H]+. Step 3-341b. A mixture of the crude compound from step 3-34 la (theo 0.183 mmol) and (S)-(methoxycarbonyl)amino-3 -methyl-butyric acid (prepared according to WO
2008/021927, 67.3 mg, 0.384 mmol) in DMF (3 mL) was treated with HATU (0.139 g, 0.366 mmol) in the presence of DIPEA (0.46 mL, 3.66 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, CH2Cl2-MeOH) to give a mixture of the title compounds as a light yellow solid (87.8 mg, 2 steps 66%). ESIMS m/z = 727.40 [M+H]+.
-342.
Figure imgf000202_0001
Step 3-342a. A mixture of l-Boc-pyrrolidine-3-carboxylic acid (0.159 g, 0.739 mmol), 4-bromo-1,2-diaminobenzene (0.142 g, 0.739 mmol) in DMF (6 mL) were added
EDC HCl (0.184 g, 0.960 mmol) and DMAP (9.0 mg, 73.8 μπιοΐ). The resultant mixture were stirred at rt for two days. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated to give the desired crude compound as a brown sticky oil (0.261 g, 92%), which was directly used in the next step.
Step 3-342b. A solution of the crude compound from step 3_2342a (0.261 g, 0.680 mmol) in glacial acetic acid (8 mL) was heated at 40°C for 14 h. The volatiles were evaporated off and the residue was partitioned (EtOAc - saturated aqueous NaHC03). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a brown oil (0.195 g, 78%). ESIMS m/z = 366.24, 368.24.
Step 3-342c. A mixture of the compound from step 3^342b (30.2 mg, 82.5 μιηοΐ), the compound from step 3^3366 (36.2 mg, 82.5 μιηοΐ) and NaHCOs (27.7 mg, 0.330 mmol) in DME (6 mL) and H20 (2 mL) was added Pd(PPh3)4 (4.7 mg, 4.1 μπιοΐ). The resultant mixture were degassed and heated to 85°C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the title compound as a yellow solid (28.1 mg, 60%). ESIMS m/z = 599.37 [M+H]+. Example 3-343.
Figure imgf000203_0001
The title compound was prepared from the compound from example 3-342 using
procedures similar to that described in example 3-341. ESIMS m/z = 713.68 [M+H]+.
-344.
Figure imgf000203_0002
Step 3-344a. A mixture of 2,4'-dibromoacetophenone (0.258 g, 0.929 mmol) and 1-Boc- pyrrolidine-3-carboxylic acid (0.200 g, 0.929 mmol) in CH3CN (9 mL) was added DIPEA (0.23 mL, 1.86 mmol) slowly. The mixture was stirred at room temperature until the disappearence of the starting material. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a light yellow solid (0.376 g, 98%). ESIMS m/z = 412.22, 414.22 [M+H]+.
Step 3-344b. A solution of the compound from step 3-344a (0.376 g, 0.912 mmol) in toluene (15 mL) was added ammonium acetate (1.41 g, 18.2 mmol) and the resultant mixture was heated up to 100°C for 16 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - aq. NaHC03). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a yellow brown oil (0.318 g, 89%). ESIMS m/z = 392.07, 394.07 [M+H]+.
Step 3-344c. A mixture of the compound from step 3-344b (0.115 g, 0.293 mmol), bis- (pinacolato) diboron (0.149 g, 0.587 mmol) and potassium acetate (71.8 mg, 0.733 mmol) in 1,4-dioxane (8 mL) was added Pd(PPh3)4 (16.9 mg, 14.6 μιηοΐ). The resultant mixture were degassed for three times and heated up to 85°C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a light yellow solid (91.0 mg, 71%). ESIMS m/z = 440.30 [M+H]+. Step 3-344d. Into a mixture of 2-bromo-1-(4-iodophenyl)ethanone (5g, 15.4 mmol) and (S)-1-(tert-butoxycarbonyl)pyrrolidine-2-carboxylic acid (3.48g, 16.1 mmol) in acetonitrile (40 mL) was added diisopropylethylamine (2.4 mL, 17 mmol). The resulting mixture was stirred at rt for 3 hours before being partitioned between EtOAc and aqueous NaHC03. The organic phase was separated, dried (Na2S04) and concentrated to afford a brown oil. It was purified by flash column chromatography (silica, hexane-EtOAc) to give the desired product as light yellow oil (6.0 g, 86%). ESIMS m/z = 481.94 [M+Na]+. Step 3-344e. The mixture of compound from step 3-344d (6.0g, 12.5 mmol) and ammonium acetate (15.1g, 196 mmol) in toluene (80 mL) was stirred at 80°C for 3 hours before being partitioned between water and aqueous NaHC03. The organic phase was separated, dried (Na2S04) and concentrated to afford a deep red oil. It was purified by flash column chromatography (silica, hexane-EtOAc) to give the desired product as light yellow solid (5.34 g, 93%). ESIMS m/z = 439.83 [M+H]+.
Step 3-344f. A mixture of the compound from step 3-344c (30.0 mg, 68.3 μιηοΐ), the compound from step 3^3446 (30.0 mg, 68.3 μιηοΐ) and NaHC03 (22.9 mg, 0.273 mmol) in DME (6 mL) and H20 (2 mL) was added Pd(PPh3)4 (3.9 mg, 3.4 μπιοΐ). The resultant mixture were degassed and heated to 85°C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the title compound as a light yellow solid (31.2 mg, 73%). ESIMS m/z = 625.30 [M+H]+.
-345.
Figure imgf000204_0001
The title compound was prepared from the compound from example 3-344 using procedures similar to that described in example 3-341. ESIMS m/z = 739.57 [M+H]+.
Examples 3-346a and 3-346b.
Figure imgf000204_0002
Step 3-346a. To a solution of Boc-trans-2-aminocyclopentane carboxylic acid (racemic, 0.350 g, 1.526 mmol) in THF (8 mL) at 0°C was added iodomethane (0.76 mL, 12.21 mmol), followed by NaH (60% in mineral oil, 0.244 g, 6.106 mmol). The milky solution was stirred at 0°C for 15 minutes and then at room temperature for 3 hours before being quenched carefully with H20. The volatiles were evaporated. The residue was diluted with H20, washed with Et20 (* 1). The aqueous layer was acidified with citric acid to pH ~3, extracted with dichloromethane. The organics were dried (Na2S04), filtered and evaporated to give the crude desired compound as a yellow oil (0.389 g), which was used directly for next step. ESIMS m/z = 307.04 [M+2Na+CH3CN-H]+.
Step 3-346b. A solution of the crude compound from step 3-346a (0.389 g, 1.526 mmol at most) and 2,4'-dibromoacetophenone (0.445 g, 1.603 mmol) in CH3CN (7 mL) was treated with TEA (0.43 mL, 3.053 mmol) at room temperature for 1.5 hours. The volatiles were evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a colorless oil (0.530 g, 77% over 2 steps). ESIMS m/z = 439.93, 441.93 [M+H]+.
Step 3-346c. A mixture of the compound from step 3-346b (0.530 g, 1.204 mmol) and ammonium acetate (1.021 g, 13.24 mmol) in xylenes (8 mL) was heated at 140°C with a microwave for 80 minutes. The volatiles were evaporated and the residue was partitioned (dichloromethane - aqueous NaHC03). The organic layer was dried (Na2S04), filtered and evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate, with 1%) Et3N in EtOAc) to give the desired compound as a yellow foam (0.420 g, 83%). ESIMS m/z = 419.91, 421.90 [M+H]+.
Step 3-346d. A mixture of compound from step 3-346c (89.0 mg, 0.212 mmol), the compound from step 3^3366 (93.0 mg, 0.212 mmol), Pd(PPh3)4 (24.5 mg, 21.2 μιηοΐ) and NaHC03 (62.3 mg, 0.741 mmol) in DME (3 mL) and H20 (1 mL) was degassed and heated at 98°C under N2 for 15 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N in EtOAc) to give a mixture of the title compounds as a yellow solid (0.104 g, 75%). ESIMS m/z = 653.03 [M+H]+. Examples 3-347a and 3-347b.
Figure imgf000206_0001
Step 3-347a. A solution of the compound of example 3-46 (0.104 g, 0.159 mmol) in 1,4- dioxane (1.5 mL) was treated with C1 in 1,4-dioxane (4 M, 6 mL) at rt for 2 hours. The volatiles were evaporated off to give the crude desired compound as a yellow solid, which was used directly in the next step.
Step 3-347b. A mixture of the crude compound from step 3-347a (0.159 mmol at most) and (5)-2-(methoxycarbonylamino)-3-methylbutanoic acid (prepared according to WO 2008/021927, 55.8 mg, 0.319 mmol) in DMF (3 mL) was treated with HATU (0.115 g, .0302 mmol) in the presence of DIPEA (0.55 mL, 3.186 mmol) for 1 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N and 20% MeOH in EtOAc) to give a mixture of the title compounds as a yellow solid (89.2 mg, 73% over 2 steps). ESIMS m/z = 767.02 [M+H]+.
The compounds of Examples 4-1 to 4-347 may be prepared using procedures similar to those described in Example 4-348 (described below), and/or as described in the Synthetic Methods.
Examples 4-1 to 4-219.
Figure imgf000206_0002
Figure imgf000207_0001
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
Figure imgf000211_0001
Figure imgf000212_0001
Figure imgf000213_0001
Figure imgf000214_0001
Figure imgf000215_0001
Step 4-348a. Into a mixture of (S)-1-(tert-butoxycarbonyl)pyrrolidine-2-carboxylic acid (486 mg, 2.26 mmol) and 5-iodopyrimidin-2-amine (500 mg, 2.26 mmol) in acetonitrile (10 mL) was added diisopropylethylamine (0.45 mL, 2.48 mmol) and HATU (859 mg, 2.26 mmol). The resulting mixture was stirred at 150°C for 1.5 hours (heating with microwave) before being partitioned between water and EtOAc. The organic phase was separated, dried (Na2S04) and concentrated to afford a brown slurry, which was purified by flash column chromatography (silica, EtOAc-hexanes) to give the desired product as a light yellow solid (146 mg, 15%). ESIMS m/z = 440.93 [M+Na]+.
Step 4-348b. 6-bromo-N-methoxy-N-methyl-2-naphthamide (prepared according to J. Med. Chem., 2006, 49, 4721-4736, 3.57 g, 12.1 mmol) in THF (60 mL) was treated with methyl magnesium bromide (3M in Et20, 8.09 mL, 24.3 mmol) slowly at 0 °C for 1 hour. The solution was warmed up to rt for 2 hours before being quenched with aqueous NH4C1. The volatiles were removed and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated to give the crude desired compound as a white solid (2.89 g, 96%).
Step 4-348c. The compound from step 4-348b (2.89 g, 11.6 mmol) in acetic acid (60 mL) was treated with bromine (0.59 mL, 11.6 mmol) dropwise for 1 hour. The volatiles were evaporated and the residue was partitioned (EtOAc - saturated aqueous NaHC03). The organics were washed with brine, dried (Na2S04), filtered and evaporated to give the crude desired compound as a light yellow solid (3.898 g).
Step 4-348d. A mixture of the compound from step 4-348c (at most 11.6 mmol) and N- Boc-L-proline (3.75 g, 17.4 mmol) in CH3CN (60 mL) was added DIPEA (2.89 mL, 23.2 mmol) slowly. The mixture was stirred at rt until the disappearence of the starting material. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated to give the crude desired compound as a yellow-white foam (4.762 g). ESIMS m/z = 462.03, 464.02 [M+H]+.
Step 4-348e. A mixture of the compound from step 4-348d (0.200 g, 0.452 mmol), bis(pinaco-lato)diboron (0.144 g, 0.565 mmol), PdCl2(dppf)2 (36.9 mg, 0.0452 mmol) and potassium acetate (88.7 mg, 0.904 mmol) in DMSO (5 mL) was degassed and heated at 80 °C under N2 for 17 hours. The reaction mixture was allowed to cool down and partitioned (EtOAc - water). The organic layer was washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a yellow solid (0.188 g, 85%). ESIMS m/z = 490.12
[M+H]+.
Step 4-348f. Into a mixture of compound from step 4-348a (146 mg, 0.35 mmol) and the compound from step 4^3486 (171 mg, 0.35 mmol) in 1 ,2-dimethoxyethane (5 mL) and water (2.5 mL) was added tetrakis(triphenylphosphine)palladium(0) (40 mg, 0.035 mmol) and NaHCC"3 (371 mg, 4.42 mmol). The resulting mixture was stirred at 65 °C under nitrogen atmosphere for 1.5 hours before being partitioned between water and EtOAc. The organic phase was separated, dried (Na2S04) and concentrated to afford a brown slurry, which was purified by flash column chromatography (silica, ethyl acetate -hexanes) to give the desired product as a light yellow solid (38 mg, 17%). ESIMS m/z = 654.13
[M+H]+.
Step 4-348g. A solution of the compound from step 4-348f (38 mg, 0.058 mmol) in dichloromethane (1 mL) and methanol (0.1 mL) was treated with C1 in 1,4-dioxane (4 M, 2 mL) at room temperature for 2 hours. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was used directly in the next step.
Step 4-348h. The mixture of compounds from step 4-348g (0.048 mml at most) and (S)- (methoxycarbonyl)amino-3-methyl-butyric acid (prepared according to WO 2008/021927, 20 mg, 0.11 mmol) in MeCN (2 mL) was added diisopropylethylamine (0.11 mL) and HATU (7.7 mg). The resulting solution was stirred at room temperature for 35 minutes before being partitioned between EtOAc and aqueous NaOH (0.5M). The organic phase was separated, dried (Na2S04) and concentrated to afford a brown slurry, which was purified by HPLC (CI 8, methanol-water) to give the title compound (2.0 mg). ESIMS m/z = 768.35 [M+H]+.
The compounds of examples 5-2 to 5-82, 5-84 to 5-293, 5-295, 5-297 to 5-304, 5- 306 to 5-312, 5-314 to 5-321, 5-323 to 5-369 and 5-371 may be prepared using procedures similar to those described in examples 5-1, 5-83, 5-294, 5-296, 5-305, 5-313, 5-322, 5- 370, 5-372 to 5-378, and 5-381 to 5-394 (described below), and/or as described in the Synthetic Methods.
Examples 5-1 to 5-219.
Figure imgf000217_0001
Figure imgf000218_0001
Figure imgf000219_0001
Figure imgf000220_0001
Figure imgf000221_0001
Figure imgf000222_0001
-220 to 5-229.
Figure imgf000222_0002
Figure imgf000223_0001
Figure imgf000224_0001
Figure imgf000225_0001
Figure imgf000226_0001
Figure imgf000227_0002
Example 5-1.
Figure imgf000227_0001
Step 5-la. A mixture of 5-bromo-2-chloropyrimidine (0.101 g, 0.524 mmol) and (S)-2- (aminomethyl)-l-N-Boc- pyrrolidine (0.105 g, 0.524 mmol) in 1,4-dioxane (2 mL) were heated up to 100 °C for 5h. The mixture was cooled down and the volatiles were evaporated. The residue was purified by flash column chromatography (silica, hexanes- ethyl acetate) to give the desired compound as a colorless oil (75.5 mg, 40%). ESIMS m/z = 357.25, 359.25 [M+H]+.
Step 5-lb. 6-bromo-N-methoxy-N-methyl-2-naphthamide (prepared according to J. Med. Chem., 2006, 49, 4721-4736, 3.57 g, 12.1 mmol) in THF (60 mL) was treated with methyl magnesium bromide (3M in Et20, 8.09 mL, 24.3 mmol) slowly at 0 °C for 1 hour. The solution was warmed up to rt for 2 hours before being quenched with aqueous NH4C1. The volatiles were removed and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated to give the crude desired compound as a white solid (2.89 g, 96%).
Step 5-lc. The compound from step 5-lb (2.89 g, 11.6 mmol) in acetic acid (60 mL) was treated with bromine (0.59 mL, 11.6 mmol) dropwise for 1 hour. The volatiles were evaporated and the residue was partitioned (EtOAc - saturated aqueous NaHC03). The organics were washed with brine, dried (Na2S04), filtered and evaporated to give the crude desired compound as a light yellow solid (3.898 g).
Step 5-ld. A mixture of the compound from step 5-lc (at most 11.6 mmol) and N-Boc-L- proline (3.75 g, 17.4 mmol) in CH3CN (60 mL) was added DIPEA (2.89 mL, 23.2 mmol) slowly. The mixture was stirred at rt until the disappearence of the starting material. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated to give the crude desired compound as a yellow-white foam (4.762 g). ESIMS m/z = 462.03, 464.02 [M+H]+. Step 5-le. A mixture of the compound from step 5-ld (0.200 g, 0.452 mmol), bis(pinaco- lato)diboron (0.144 g, 0.565 mmol), PdCl2(dppf)2 (36.9 mg, 0.0452 mmol) and potassium acetate (88.7 mg, 0.904 mmol) in DMSO (5 mL) was degassed and heated at 80 °C under N2 for 17 hours. The reaction mixture was allowed to cool down and partitioned (EtOAc - water). The organic layer was washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a yellow solid (0.188 g, 85%). ESIMS m/z = 490.12 [M+H]+.
Step 5-lf. A mixture of the compound from step 5- la (30.2 mg, 84.5 μιηοΐ), the compound from step le (53.7 mg, 0.110 mmol) and NaHC03 (28.4 mg, 0.338 mmol) in DME (6 mL) and H20 (2 mL) was added Pd(PPh3)4 (4.9 mg, 4.2 μιηοΐ). The resultant mixture were degassed and heated to 85°C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the title compound as a light yellow oil (47.1 mg, 87%). ESIMS m/z = 640.61 [M+H]+.
-83.
Figure imgf000228_0001
Step 5-83a. A solution of the compound of example 5-1 (20.0 mg, 31.2 μιηοΐ) in CH2CI2 (4 mL) was treated with C1 in 1,4-dioxane (4 M, 3 mL) for 30 min. The volatiles were evaporated off to give the crude desired compound as a yellow solid which was directly used in the next step. ESIMS m/z = 440.47 [M+H]+.
Step 5-83b. A mixture of the crude compound from step 5-83a (max. 31.2 μιηοΐ) and (S)- (methoxycarbonyl)amino-3-methyl-butyric acid (prepared according to WO 2008/021927, 11.5 mg, 65.5 μιηοΐ) in DMF (3 mL) was treated with HATU (23.7 mg, 62.4 μιηοΐ) in the presence of DIPEA (78 μΐ^, 0.624 mmol) for 2 hours at rt and the volatiles were evaporated off to provide a brown syrup. It was purified by flash column chromatography (silica, CH2Cl2-MeOH) to give the title compound as a white solid (18.9 mg, 2 steps 80%). ESIMS m/z = 754.81 [M+H]+.
Figure imgf000229_0001
The title compound was prepared from the compound of example 5-391 using procedures similar to that described in example 5-83. ESIMS m/z = 754.14 [M+H]+.
-296.
Figure imgf000229_0002
The title compound was prepared from the compound of example 5-378 using procedures similar to that described in example 5-83. ESIMS m/z = 753.44 [M+H]+.
-305.
Figure imgf000229_0003
The title compound was prepared from the compound of example 5-375 using procedures similar to that described in example 5-83. ESIMS m/z = 759.52 [M+H]+. Example 5-313.
Figure imgf000230_0001
Step 5-313a. The desired compound was prepared from 5-bromo-2-chloropyrimidine and (S)-tert-butyl 2-(aminomethyl)pyrrolidine-1-carboxylate using procedures similar to that described in step 5-la. ESIMS m/z = 357.12 [M+H]+.
Step 5-313b. The desired compound was prepared from compound of step 5-313a and trimethylsiltyl acetylene using procedures similar to that described in steps 5 -392c and 5- 392d. ESIMS m/z = 303.14 [M+H]+.
Step 5-313c. The desired compound was prepared from the compound from step 5-313b and the compounds from step 5 -392b using procedures similar to that described in step 5- 392e. ESIMS m/z = 588.26 [M+H]+.
Step 5-313d. The title compound was prepared from the compound from step 5-313a and the compound from step 5-392d using procedures similar to that described in example 5- 83. ESIMS m/z = 702.34 [M+H]+.
-322.
Figure imgf000230_0002
The title compound was prepared from the compound of example 5-392 using procedures similar to that described in example 5-83. ESIMS m/z = 752.12 [M+H]+.
Example 5-370
Figure imgf000230_0003
The title compound was prepared from the compound of example 5-374 using procedures similar to that described in example 5-83. ESIMS m/z = 745.57 [M+H]+. Examples 5 -372a and 5 -372b.
B
B
Figure imgf000231_0003
Step 5-372a. The desired compound was prepared from trans-N-Boc-1,2- cyclopentanediamine and 5-bromo-2-chloropyrimidine using procedures similar to that described in step 5-la. ESIMS m/z = 357.12, 359.12 [M+H]+.
Step 5-372b. A mixture of the title compounds was prepared from the compound from step 5-372a using precedures similar to that described in step 5- If. ESIMS m/z = 640.49 [M+H]+.
-373 a and 5 -373b.
Figure imgf000231_0001
A mixture of the title compounds were prepared from the compounds of example 5 -372a and 5-372b using procedures similar to that described in example 5-83. ESIMS m/z = 754.72 [M+H]+.
-374.
Figure imgf000231_0002
A mixture of the compound from step 5-la (40.0 mg, 0.112 mmol), benzene- 1 ,4-diboronic acid (8.4 mg, 50.9 μιηοΐ) in toluene (3 mL), EtOH (3 mL) and 2M aqueous Na2C03 (0.2 mL) was added Pd(PPh3)4 (5.8 mg, 5.0 μιηοΐ). The resultant mixture were degassed and heated to reflux under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the title compound as a light yellow solid (18.4 mg, 57%). ESIMS m/z = 631.31 [M+H]+.
-375.
Figure imgf000232_0001
Step 5-375a. A mixture of 6-bromo-2-chlorobenzothiazole (0.126 g, 0.508 mmol) and (5)-2-(aminomethyl)-l-N-Boc- pyrrolidine (0.102 g, 0.508 mmol) in 2-propanol (6 mL) were added K2CO3 (70.1 mg, 0.508 mmol). The resultant mixture were degassed and heated to 85 °C under N2 for 16 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a light yellow oil (76.1 mg, 36%). ESIMS m/z = 412.15, 414.12 [M+H]+.
Step 5-375b. A mixture of 2,4'-dibromoacetophenone (5.00 g, 18.0 mmol) and N-Boc-L- proline (3.87 g, 18.0 mmol) in CH3CN (60 mL) was added TEA (5.40 mL, 37.8 mmol) slowly. The mixture was stirred at room temperature until the disappearence of the starting material. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a light yellow foam (6.73 g, 91 >). 1H NMR (CDC13) 7.76 (t, J= 8.0 Hz, 2H), 7.63 (dd, J= 5.0, 8.5 Hz, 2H), 5.51, 5.16 (2d, J= 16.0 Hz, 1H), 5.32, 5.28 (2d, J= 16.5 Hz, 1H), 4.48, 4.40 (dd, J= 5.0, 8.5 Hz, 1H), 3.56 (m, 1H), 3.43 (m, 1H), 2.30 (m, 2H), 2.06 (m, 1H), 1.92 (m, 1H), 1.46, 1.43 (2s, 9H).
Step 5-375c. A solution of the compound from step 5-375b (6.73 g, 16.3 mmol) in toluene (100 mL) was added ammonium acetate (25.1 g, 0.327 mol) and the resultant mixture was heated up to 100°C for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - aq. NaHC03). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a yellow foam (6.10 g, 95%). ESIMS m/z = 392.24, 394.24 [M+H]+. 1H NMR (CDC13) 7.57 (bs, 1H), 7.48 (m, 3H), 7.23 (s, 1H), 4.97 (m, 1H), 3.42 (m, 2H), 2.99 (m, 1H), 2.16 (m, 2H), 1.97 (m, 1H), 1.46 (s, 9H). Step 5-375d. A mixture of the compound from step 5-375c (1.00 g, 2.55 mmol), bis- (pinacolato) diboron (1.35 g, 5.33 mmol) and potassium acetate (0.640 g, 6.53 mmol) in 1,4-dioxane (20 mL) was added Pd(PPh3)4 (0.147 g, 0.128 mmol). The resultant mixture were degassed for three times and heated up to 80 °C under N2 for 14 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give intermediate 1 as a light yellow solid (0.978 g, 87%). ESIMS m/z = 440.39 [M+H]+. 1H NMR (CDC13) 11.03, 10.55 (2s, 1H), 7.79 (m, 3H), 7.45 (m, 1H), 7.26 (m, 1H), 4.97 (m, 1H), 3.41 (m, 2H), 3.06, 2.91 (2m, 1H), 2.17 (m, 2H), 1.97 (m, 1H), 1.49 (s, 9H), 1.35 (s, 12H).
Step 5-375e. The title compound was prepared from the compound from step 5-375a and the compound from step 5-375d using precedures similar to that described in step 5- If ESIMS m/z = 645.30 [M+H]+.
-376.
Figure imgf000233_0001
Step 5-376a. A mixture of the compound from step 5-375a (29.8 mg, 72.3 μιηοΐ), bis- (pinacolato) diboron (22.9 mg, 90.3 μιηοΐ) and potassium acetate (14.2 mg, 0.145 mmol) in DMSO (5 mL) was added Pd(dppf)Cl2-CH2Cl2 (5.9 mg, 7.2 μπιοΐ). The resultant mixture were degassed for three times and heated up to 80 °C under N2 for 34 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a colorless oil (17.5 mg, 53%). ESIMS m/z = 460.29 [M+H]+.
Step 5-376b. The title compound was prepared from the compound from step 5-376a and the compound from step 5 -375 a using precedures similar to that described in step 5- If ESIMS m/z = 665.30 [M+H]+.
-377.
Figure imgf000233_0002
The title compound was prepared from the compound of example 5-376 using procedures similar to that described in example 5-83. ESIMS m/z = 779.65 [M+H]+. -378.
Figure imgf000234_0001
Step 5-378a. A mixture of 5-bromo-2-iodopyridine (0.100 g, 0.352 mmol), Cul (3.4 mg, 17.6 μιηοΐ) and Cs2C03 (0.230 g, 0.704 mmol) in DMF (3 mL) were added (S)-2- (Aminomethyl)-l-N-Boc- pyrrolidine (0.104 g, 0.528 mmol) and 2- isobutyrylcyclohexanone (11.8 mg, 7.04 μιηοΐ). The resultant mixture were degassed stirred under N2 for 20 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - H20). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a colorless oil (0.112 g, 90%). ESIMS m/z = 356.27, 358.27 [M+H]+.
Step 5-378b. The title compound was prepared from the compound from step 5-378a and the compound from step le using precedures similar to that described in step If ESIMS m/z = 639.44 [M+H]+.
-381.
Figure imgf000234_0002
Step 5-381a. The desired compound was prepared from l-N-Boc-3-aminopyrrolidine and 5-bromo-2-chloropyrimidine using procedures similar to that described in step 5- la. ESIMS m/z = 343.02, 345.02 [M+H]+.
Step 5-381b. The title compound was prepared from the compound from step 5-3815a and the compound from step 5-375d using precedures similar to that described in step 5- lf. ESIMS m/z = 626.35 [M+H]+.
-382.
Figure imgf000234_0003
The title compound was prepared from the compound of example 5-381 using procedures similar to that described in example 5-83. ESIMS m/z = 740.61 [M+H]+. Example 5-383.
Figure imgf000235_0001
The title compound was prepared from the compound from step 5-38 la and the compound from step 5-375d using precedures similar to that described in step 5-lf. ESIMS m/z = 576.34 [M+H]+.
-384.
Figure imgf000235_0002
The title compound was prepared from the compound from example 5-383 using procedures similar to that described in example 5-83. ESIMS m/z = 690.57 [M+H]+.
-385.
Figure imgf000235_0003
The title compound was prepared from the compound from step 5 -375 a and the compound from step 5-le using precedures similar to that described in step 5-lf. ESIMS m/z = 695.33 [M+H]+.
-386.
Figure imgf000235_0004
The title compound was prepared from the compound of example 5-385 using procedures similar to that described in example 5-83 ESIMS m/z = 809.72 [M+H]+.
-387.
Figure imgf000235_0005
Step 5-387a. The desired compound was prepared from 2-N-Boc-butane-1,2-diamine- C1 and 5-bromo-2-chloropyrimidine using procedures similar to that described in step 5- la. ESIMS m/z = 345.22, 347.23 [M+H]+. Step 5-387b. The title compound was prepared from the compound from step 5-387a and the compound from step 5-1 e using precedures similar to that described in step 5- If.
ESIMS m/z = 628.43 [M+H]+.
-388.
Figure imgf000236_0001
The title compound was prepared from the compound from example 5-387 using procedures similar to that described in example 5-83. ESIMS m/z = 742.60 [M+H]+.
-389.
Figure imgf000236_0002
Step 5-389a. A mixture of 2-N-Boc-butane-1,2-diamine-HCl (0.300 g, 1.34 mmol), Et3N (0.56 mL, 4.00 mmol) in THF (5 mL) was added N-ethoxycarbonylphthalimide (0.439 g, 2.00 mmol). The resultant mixture were degassed for three times and heated to reflux under N2 for 5 hours. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated to give the desired crude compound as a white solid (0.524 g). ESIMS m/z = 319.18 [M+H]+.
Step 5-389b. A mixture of the compound from step 5-389a (1.34 mmol at most), Mel (1.25 mL, 2.00 mmol) in THF (8 mL) at 0°C was added NaH (60% in mineral oil, 0.160 g, 4.00 mmol). The resultant mixture were gradually warmed up to rt and stirred for 20 hours. The reaction was quenched by the addition of aqueous NH4C1 and the volatiles were evaporated. The residue was partitioned (EtOAc - water) and the organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by flash column chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a colorless oil (0.305 g, 2 steps 69%). ESIMS m/z = 333.21 [M+H]+.
Step 5-389c. A solution of the compound from step 5-389b (0.150 g, 0.452 mmol) in EtOH (10 mL) was added hydrazine (71 μί, 2.26 mmol). The resultant mixture were heated to reflux for 2 hours. The volatiles were evaporated to give the desired crude compound as a white solid (97.5 mg). ESIMS m/z = 203.22 [M+H]+. Step 5-389d. The desired compound was prepared from the compound from step 5-389c and 5-bromo-2-chloropyrimidine using procedures similar to that described in step 5- la. ESIMS m/z = 359.17, 361.17 [M+H]+.
Step 5-389e. The title compound was prepared from the compound from step 5-389d and the compound from step 5-1 e using precedures similar to that described in step 5- If. ESIMS m/z = 642.52 [M+H]+.
-390.
Figure imgf000237_0001
The title compound was prepared from the compound of example 5-389 using procedures similar to that described in example 5-83. ESIMS m/z = 756.72 [M+H]+.
-391.
Figure imgf000237_0002
Step 5-391a. A suspension of 2,6-dibromoquinazoline (0.120 g, 0.417 mmol) and (S)-N- Boc-2-(aminomethyl)pyrrolidine (83.5 mg, 0.417 mmol) in z'-PrOH (8 mL) and DIPEA (0.15 mL, 0.833 mmol) was heated at 85 oC for 1 hour. The reaction mixture was combined with another batch of 70.0 mg of 2,6-dibromoquinazoline and concentrated. The residue was purified by chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a yellow oil (0.120 g, 45%). ESIMS m/z = 407.15, 409.15 [M+H]+. Step 5-391b. A mixture of compound from step 5-39 la (55.0 mg, 0.135 mmol), the compound from step 5-375d (59.3 mg, 0.135 mmol), Pd(PPh3)4 (15.6 mg, 13.5 μιηοΐ) and NaHC03 (39.7 mg, 0.473 mmol) in DME (3 mL) and H20 (1 mL) was degassed and heated at 97 °C under N2 for 15 hours. The volatiles were evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N in EtOAc) to give the title compound as a yellow foam (64.0 mg, 74%). ESIMS m/z = 640.37 [M+H]+.
-392.
Figure imgf000237_0003
Step 5-392a. A mixture of N-Boc-L-proline (5.754 g, 26.7 mmol) and TEA (3.73 mL, 26.7 mmol) in THF (60 mL) at -20°C was treated with ethyl chloroformate (2.55 mL, 26.7 mmol) for 30 minutes before a slow addition of 4-bromo-1,2-diaminobenzene (5.00 g, 26.7 mmol) in THF (20 mL). It was then kept at -20°C for 1 hour and then slowly warmed up to rt and stirred at rt overnight. The volatiles were evaporated and the residue was partitioned (EtOAc - water). The organics were washed with brine, dried (Na2S04), filtered and evaporated to give the crude desired compound as a dark brown foam (10.7 g). ESIMS m/z = 384.18, 386.18 [M+H]+.
Step 5-392b. A solution of the crude compound from step 5-392a (10.7 g, 26.7 mmol at most) in glacial acetic acid (100 mL) was heated at 50°C for 2 hours. The volatiles were evaporated off and the residue was partitioned (EtOAc - aqueous NaHC03). The organics were washed with brine, dried (Na2S04), filtered and evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate) to give the desired compound as a brown foam (5.78 g, 59%). ESIMS m/z = 366.17, 368.17 [M+H]+. 1H NMR (CDC13) 10.96, 10.93 (2 s, 1H), 7.81, 7.30 (2 s, 1H), 7.53, 7.17 (2d, J= 8.5 Hz, 1H), 7.23, 7.03 (2d, J= 8.5 Hz, 1H), 5.09, 5.07 (2s, 1H), 3.42-3.49 (m, 2H), 2.75-2.85 (m, 1H), 2.13-2.23 (m, 2H), 1.97-2.00 (m, 1H), 1.48 (s, 9H).
Step 5-392c. A mixture of the compound from step 5-392b (2.010 g, 5.488 mmol), trimethylsilyl-acetylene (2.33 ml, 16.46 mmol), Cul (0.110 g, 0.576 mmol) and
Pd(PPh3)2Cl2 (0.308 g, 0.439 mmol) in Et3N (50 mL) was degased and then heated at 80°C under N2 overnight before being evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate with 1% Et3N in ethyl acetate) to give the desired compound as a yellow foam (1.140 g, 54%). ESIMS m/z = 384.22 [M+H]+.
Step 5-392d. A suspension of the compound from step 5-392c (1.140 g, 2.972 mmol) and K2C03 (1.027 g, 7.430 mmol) in methanol (30 ml) was stirred at rt for 2 hour. The volatiles were evaporated off. The residue was patitioned (EtOAc - H20). The organic layer was washed with brine, dried (Na2S04), filtered and concentrated. The residue was purified by chromatography (silica, hexanes-ethyl acetate with 1% Et3N in ethyl acetate) to give the desired compound as a yellow foam (0.792 g, 86%>). ESIMS m/z = 312.18
[M+H]+.
Step 5-392e. A mixture of compound from step 5-391a (50.4 mg, 0.124 mmol), the compound from step 392d (48.2 mg, 0.15 mmol), Pd(PPh3)4 (7.1 mg, 6.2 μιηοΐ) and Cul (0.7 mg, 3.7 μιηοΐ) in THF (2 mL) and Et3N (1 mL) was degassed and heated at 85°C under N2 for 15 hours. The volatiles were evaporated. The residue was purified by chromatography (silica, hexanes-ethyl acetate, with 1% Et3N in EtOAc) to give the title compound as a yellow foam (44.7 mg, 57%). ESIMS m/z = 638.31 [M+H]+. Example 5-393.
Figure imgf000239_0001
The title compound was prepared from the (R)-tert-butyl 2-(aminomethyl)pyrrolidine-1- carboxylate using procedures similar to that described in examples 5-1 and 5-83. ESIMS m/z = 754.42 [M+H]+.
-394.
Figure imgf000239_0002
The title compound was prepared from the compound from step 5-313a and the compound from step 5 -375c using procedures similar to that described in example 5-313. ESIMS m/z = 728.17 [M+H]+.
BIOLOGICAL ACTIVITY
1. HCV Replicon Cell Lines
HCV replicon cell lines (kindly provided by R. Bartenschlager) isolated from colonies as described by Lohman et. al. (Lohman et al. (1999) Science 285: 110-113, expressly incorporated by reference in its entirety) and used for all experiments. The HCV replicon has the nucleic acid sequence set forth in EMBL Accession No.: AJ242651, the coding sequence of which is from nucleotides 1801 to 8406.
The coding sequence of the published HCV replicon was synthesized and subsequently assembled in a modified plasmid pBR322 (Promega, Madison, WI) using standard molecular biology techniques. One replicon cell line ("SGR 11-7") stably expresses HCV replicon RNA which consists of (i) the HCV 5'UTR fused to the first 12 amino acids of the capsid protein, (ii) the neomycin phosphotransferase gene (neo), (iii) the IRES from encephalomyocarditis virus (EMCV), and (iv) HCV NS2 to NS5B genes and the HCV 3'UTR. Another replicon cell line ("Huh-luc/neo-ET") described by Vrolijk et. al. (Vrolijk et. al. (2003) Journal of Virological Methods 110:201-209, expressly incorporated by reference in its entirety) stably expresses HCV replicon RNA which consists of (i) the HCV 5'UTR fused to the first 12 amino acids of the capsid protein, (ii) the firefly luciferase reporter gene, (iii) the ubiquitin gene, (iv) the neomycin
phosphotransferase gene (neo), (v) the IRES from encephalomyocarditis virus (EMCV), and (vi) HCV NS3 to NS5B genes that harbor cell culture adaptive mutations (E1202G, T1280I, K1846T) and the HCV 3*UTR.
These cell lines were maintained at 37°C, 5% C02, 100% relative humidity in DMEM (Cat# 11965-084, Invitrogen), with 10% fetal calf serum ("FCS", Invitrogen), 1% non-essential amino acids (Invitrogen), 1% of Glutamax (Invitrogen), 1% of 100X penicillin/streptomycin (Cat# 15140-122, Invitrogen) and Geneticin (Cat# 10131-027, Invitrogen) at 0.75 mg/ml or 0.5 mg/ml for 11-7 and Huh-luc/neo-ET cells, respectively. 2. HCV Replicon Assay - qRT-PCR
EC50 values of single agent compounds and combinations were determined by HCV RNA detection using quantitative RT-PCR, according to the manufacturer's instructions, with a TAQMAN® One-Step RT-PCR Master Mix Reagents Kit (Cat# AB 4309169, Applied Biosystems) on an ABI Model 7500 thermocycler. The TaqMan primers used for detecting and quantifying HCV RNA were obtained from Integrated DNA Technologies. HCV RNA was normalized to GAPDH RNA levels in drug-treated cells, which was detected and quantified using the Human GAPDH Endogenous Control Mix (Applied Biosystems, AB 4310884E). Total cellular RNA was purified from 96-well plates using the RNAqueous 96 kit (Ambion, Cat# AMI 812). Chemical agent
cytotoxicity was evaluated using an MTS assay according to the manufacturer's directions (Promega).
3. HCV Replicon Assay - Luciferase
Since clinical drug resistance often develops in viral infections following single agent therapies, there is a need to assess the additive, antagonistic, or synergistic properties of combination therapies. We use the HCV replicon system to assess the potential use of the compound of the present invention or in combination therapies with Interferon alpha, cyclosporine analogs and inhibitors targeting other HCV proteins. The acute effects of a single or combinations of drugs are studied in the "Huh-luc/neo-ET" replicon with each chemical agent titrated in an X or Y direction in a 6 point two-fold dilution curve centered around the EC50 of each drug. Briefly, replicon cells are seeded at 7,000 cells per well in 90 ul DMEM (without phenol red, Invitrogen Cat.# 31053-036) per well with 10%> FCS, 1% non-essential amino acids, 1% of Glutamax and 1% of 100X penicillin/streptomycin and incubated overnight at 37°C, 5% C02, 100% relative humidity. 16-20h after seeding cells, test compounds previously solubilized and titrated in dimethyl sulfoxide ("DMSO") from each X plate and Y plate are diluted 1 : 100 in DMEM (without phenol red, Invitrogen Cat.# 31053-036) with 10% FCS, 1% non-essential amino acids, 1% of Glutamax and 1% of 100X penicillin/streptomycin and added directly to the 96-well plate containing cells and growth medium at a 1 : 10 dilution for a final dilution of compound and DMSO of 1 : 1000 (0.2% DMSO final concentration). Drug treated cells are incubated at 37°C, 5% C02, 100% relative humidity for 72 hours before performing a luciferase assay using 100 ul per well BriteLite Plus (Perkin Elmer) according to the manufacturer's instructions. Data analysis utilizes the method published by Prichard and Shipman (Antiviral Research, 1990. 14: 181-205). Using this method, the combination data are analyzed for antagonistic, additive, or synergistic combination effects across the entire combination surface created by the diluted compounds in combination.
The compounds of the present invention may inhibit HCV by mechanisms in addition to or other than NS5A inhibition. In one embodiment, the compounds of the present invention inhibit HCV replicon and in another embodiment the compounds of the present invention inhibit NS5A.
The compounds of the present invention can be effective against the HCV lb genotype. It should also be understood that the compounds of the present invention can inhibit multiple genotypes of HCV. In one embodiment compound of the present invention are active against the la, lb, 2a, 2b, 3a, 4a, and 5a genotypes. Table 58 shows the EC50 values of representative compounds of the present invention against the HCV lb genotype from the above described qRT-PCR or luciferase assay. EC50 ranges against HCV lb are as follows: A >10 nM; B 1-10 nM; C < InM. Where two compound are indicated under the Example column (e.g., l-349a and l-349b, it is to be understood that a mixture of the two compounds was tested in the above described qRT-PCR or luciferase assay.
Figure imgf000241_0001
Figure imgf000242_0001
While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims

WHAT IS CLAIMED IS: 1. A compound represented by Formula (1-1):
Figure imgf000243_0001
or a pharmaceutically acceptable salt thereof, wherein:
Ring A is absent or a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C3-C8 cycloalkyl, and C3-C8 cycloalkenyl, each optionally substituted;
Ring B is a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C3-C8 cycloalkyl, and C3-C8 cycloalkenyl, each optionally substituted;
L is absent or selected from the group consisting of optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, and optionally substituted C2-C4 alkynyl;
G is optionally substituted 5 -membered heteroaryl or optionally substituted 5/6- member fused heteroaryl; wherein the 5 -membered heteroaryl contains one or more nitrogen atoms, and wherein the 6-membered ring of said 5/6-fused membered heteroaryl is attached to one of Ring A, L, and Ring B, and is aryl or heteroaryl;
J is selected from -N(Rlc)-C(0)-, -N(Rlc)-C(0)0- and -N(Rlc)-C(0)-N(Rlc)-;
W at each occurrence is independently O or -N(Rlb)-;
R1, Rla, Rlb, Rlc, R9, and R9a at each occurrence are each independently hydrogen or optionally substituted C1-C4 alkyl; alternatively Rla and R9a, Rla and R9, R1 and R9a, or R1 and R9 can be taken together with the carbon atom(s) to which they are attached to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or yet alternatively Rlb and R9a, or Rlb and R9 can be taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic;
R6 at each occurrence is independently selected from the group consisting of optionally substituted 0(C1-C8 alkyl); optionally substituted amino; C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted;
Q is selected from:
Figure imgf000244_0001
X is absent, O, S, CH2, or CH2CH2;
Y is absent, O, S, C(R1)2, C(R1)2C(R7)2, C(R1)2C(R7)2C(R7)2, C(R^OC(R1)2, or C(R^SC(R1)2;
Wherein at least one of X and Y is present;
Wherein at least one of X and Y is not O or S;
U is absent or independently selected from O, S, S(O), S02, NC(0)-(C1-C4 alkyl), C(O), protected carbonyl, OCH2, OCH2CH2, SCH2, SCH2CH2, C(R7)2, and C(R7)2C(R7)2;
R7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C1-C4 alkyl), S(C1-C4 alkyl), amino optionally substituted with one or two C1-C4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C1-C4 alkyl;
Alternatively, two geminal R7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of optionally substituted C3-C8 cycloalkyl, optionally substituted C3-C8 cycloalkenyl and optionally substituted 3- to 8-membered heterocyclic;
R2 at each occurrence is independently hydrogen, optionally substituted C1-C8 alkyl, or -NRaRb;
Ra at each occurrence is independently hydrogen or optionally substituted C1-C8 alkyl; Rb at each occurrence is -C(0)-R6;
Alternatively Ra and Rb can be taken together with the nitrogen atom to which they are attached to form an optionally substituted heterocyclic or optionally substituted heteroaryl group;
R3 and R4 are each independently selected from the group consisting of hydrogen, optionally substituted C1-C8 alkyl, optionally substituted C2-C8 alkenyl, and optionally substituted C3-C8 cycloalkyl; alternatively, R3 and R4 can be taken together with the carbon atom to which they are attached to form optionally substituted C3-C8 cycloalkyl or optionally substituted heterocyclic; R5 is independently hydrogen, optionally substituted C1-C8 alkyl, or optionally substituted C3-C8 cycloalkyl; and
R7a and R7b at each occurrence are each independently selected from the group consisting of hydrogen, optionally substituted aryl, and optionally substituted C1-C4 alkyl; alternatively, CHR7a-U or CHR7b-U can be taken together to form a group selected from CH=CH, optionally substituted C3-C8 cycloalkyl, optionally substituted aryl, or optionally substituted heterocyclic; or yet alternatively, U, R7a, and R7b can be taken together with the carbon atoms to which they are attached to form a bridged, optionally substituted 4- to 7- membered ring selected from the group consisting of C4-C7 cycloalkyl, C4-C7 cycloalkenyl and 4- to 7-membered heterocyclic, each optionally substituted.
2. The compound of claim 1 , wherein G is
Figure imgf000245_0001
wherein said imidazolyl or benzimidazolyl groups are each optionally substituted; or a pharmaceutically acceptable salt thereof.
3. The compound of claim 2, wherein Q is
Figure imgf000245_0002
or
Figure imgf000245_0003
; and Ring B optionally substituted aryl or optionally substituted heteroaryl; or a pharmaceutically acceptable salt thereof.
4. The compound of claim 3, wherein J is -NH-C(O)-; R6 is independently C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino, or 0(C1-C4 alkyl); or a pharmaceutically acceptable salt thereof.
5. The compound of claim 4, wherein Rlb and R9a, or Rlb and R9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic; or a pharmaceutically acceptable salt thereof.
6. The compound of claim 4, wherein Rla and R9, or R1 and R9a are taken together with the carbon atom(s) to which they are attached to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or a pharmaceutically acceptable salt thereof.
7. The compound of claim 4, wherein Ring A and L are absent; Ring B is optionally substituted bicyclic aryl or optionally substituted bicyclic heteroaryl; and R6 is independently C1-C8 alkyl optionally substituted with protected amino or 0(C1-C4 alkyl); or a pharmaceutically acceptable salt thereof.
8. The compound of claim 4, wherein Ring A is absent; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; Ring B is phenyl, monocyclic heteroaryl, bicyclic aryl, or bicyclic heteroaryl, each optionally substituted; and R6 is independently C1-C8 alkyl optionally substituted with protected amino or 0(C1-C4 alkyl); or a pharmaceutically acceptable salt thereof.
9. The compound of claim 1 selected from the group of compounds 1-1 to 1-374 and 1-377 to 1-382 compiled in the following tables:
Figure imgf000246_0001
Figure imgf000247_0001
Figure imgf000248_0001
Figure imgf000249_0001
Figure imgf000250_0001
Figure imgf000251_0001
Figure imgf000252_0001
Figure imgf000253_0001
Figure imgf000254_0001
Figure imgf000255_0001
Figure imgf000256_0001
Figure imgf000257_0001
Figure imgf000258_0001
Figure imgf000259_0002
10. A com ound represented by Formula (2-1):
Figure imgf000259_0001
or a pharmaceutically acceptable salt thereof, wherein:
Ring A and Ring B are each independently a monocyclic or polycyclic group selected from aryl, heteroaryl, heterocyclic, C3-C8 cycloalkyl, and C3-C8 cycloalkenyl, each optionally substituted;
L is absent or selected from the group consisting of optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, and optionally substituted C2-C4 alkynyl; Wherein
Figure imgf000260_0001
taken together is not optionally substituted phenyl- thiazolyl or 3,4'-biphenyl;
J is selected from the group consisting of -N(Rlc)-C(0)-, -N(Rlc)-C(0)0- and -N(Rlc)-C(0)-N(Rlc)-;
W at each occurrence is independently O or -N(Rlb)-;
R1, Rla, Rlb, Rlc, R9, and R9a at each occurrence are each independently hydrogen or optionally substituted C1-C4 alkyl; alternatively Rla and R9a, Rla and R9, R1 and R9a, or R1 and R9 can be taken together with the carbon atom(s) to which they are attached to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or yet alternatively Rlb and R9a, or Rlb and R9 can be taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic;
R6 at each occurrence is independently selected from the group consisting of optionally substituted 0(C1-C8 alkyl); optionally substituted amino; C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted;
G is selected from the group consisting of -N(Rlc)-C(0)-, -N(Rlc)-C(0)0- and -N lc)-C(0)-N(Rlc)-;
Figure imgf000260_0002
Or alternatively G and Q taken together to form
Figure imgf000260_0003
X is absent, O, S, CH2, or CH2CH2;
Y is absent, O, S, C(R1)2, C(R1)2C(R7)2, C(R1)2C(R7)2C(R7)2, C(R1)2OC(R1)2, or C(R1)2S C(R1)2;
Wherein at least one of X and Y is present;
Wherein at least one of X and Y is not O or S;
R7 at each occurrence is independently selected from the group consisting of hydrog halogen, cyano, hydroxy, 0(C1-C4 alkyl), S(C1-C4 alkyl), amino optionally substituted with one or two C1-C4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C1-C4 alkyl;
R2 at each occurrence is independently hydrogen, optionally substituted C1-C8 alkyl, or -NRaRb;
Ra at each occurrence is independently hydrogen or optionally substituted C1-C8 alkyl;
Rb at each occurrence is -C(0)-R6; and
Alternatively Ra and Rb can be taken together with the nitrogen atom to which they are attached to form an optionally substituted heterocyclic or optionally substituted heteroaryl group.
11. The compound of claim 10, wherein one or both of G and J is -NH-C(O)-; or a pharmaceutically acceptable salt thereof.
12. The compound of claim 11, wherein Q is
Figure imgf000261_0001
; and Ring B is optionally substituted aryl or optionally substituted heteroaryl; or a pharmaceutically acceptable salt thereof.
13. The compound of claim 12, wherein R6 is independently C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino, or 0(C1-C4 alkyl); or a
pharmaceutically acceptable salt thereof.
14. The compound of claim 13, wherein Rlb and R9a, or Rlb and R9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic; or a pharmaceutically acceptable salt thereof. 15. The compound of claim 13, wherein Rla and R9, or R1 and R9a are taken together with the carbon atom(s) to which they are attached to form an optionally substituted C3-C8 cycloalkyl or an optionally substituted 4- to 8-membered heterocyclic; or a
pharmaceutically acceptable salt thereof.
16. The compound of claim 13, wherein Ring A and L are absent; Ring B is optionally substituted bicyclic aryl or optionally substituted bicyclic heteroaryl; and R6 is C1-C8 alkyl optionally substituted with protected amino or 0(C1-C4 alkyl); or a pharmaceutically acceptable salt thereof.
17. The compound of claim 13, wherein Ring A is absent; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; Ring B is phenyl, monocyclic heteroaryl, bicyclic aryl, or bicyclic heteroaryl, each optionally substituted; and R6 is independently C1-C8 alkyl optionally substituted with protected amino or 0(C1-C4 alkyl); or a pharmaceutically acceptable salt thereof.
18. The compound of claim 10, selected from the group of compounds 2-1 to 2-295 compiled in the following tables:
Figure imgf000262_0001
Figure imgf000263_0001
Figure imgf000264_0001
Figure imgf000265_0001
Figure imgf000266_0001
Figure imgf000267_0001
Figure imgf000268_0001
Figure imgf000269_0001
Figure imgf000270_0002
Compounds 2-294 to 2-295
Figure imgf000270_0003
19. A compound represented by Formula (3-1):
Q
Figure imgf000270_0001
or a pharmaceutically acceptable salt thereof, wherein:
Ring A and Ring B are each independently absent or a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C3-C8 cycloalkyl, and C3-C8 cycloalkenyl, each optionally substituted;
L is absent or selected from the group consisting of optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, and optionally substituted C2-C4 alkynyl;
Wherein at least one of Ring A, Ring B and L is present;
G and J are each independently optionally substituted 5-membered heteroaryl or optionally substituted 5/6-member fused heteroaryl; wherein the 5-membered heteroaryl contains one or more nitrogen, and wherein the 6-membered ring of said 5/6-fused membered heteroaryl is attached to group A, B or L and is aryl or heteroaryl;
W at each occurrence is independently O or -N(Rlb)-;
R1, Rla, Rlb, R9, and R9a at each occurrence are each independently hydrogen or optionally substituted C1-C4 alkyl; alternatively Rla and R9a, Rla and R9, R1 and R9a, or R1 and R9 can be taken together with the carbon atom(s) to which they are attached to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or yet alternatively Rlb and R9a, or Rlb and R9 can be taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic;
R6 at each occurrence is independently selected from the group consisting of optionally substituted 0(C1-C8 alkyl); optionally substituted amino; C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted;
Q is selected from:
Figure imgf000271_0001
X is absent, O, S, CH2, or CH2CH2;
Y is absent, O, S, C(R , C(R1)2C(R7)2, C(R1)2C(R7)2C(R7)2, C(R1)2OC(R1)2, or C(R^SC(R1)2;
Wherein at least one of X and Y is present;
Wherein at least one of X and Y is not O or S;
U is absent or independently selected from O, S, S(O), S02, NC(0)-(C1-C4 alkyl), C(O), protected carbonyl, OCH2, OCH2CH2, SCH2, SCH2CH2, and C(R7)2, and
C(R7)2C(R7)2;
R7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C1-C4 alkyl), S(C1-C4 alkyl), amino optionally substituted with one or two C1-C4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C1-C4 alkyl;
Optionally two geminal R7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C3-C8 cycloalkyl, C3-C8 cycloalkenyl and 3- to 8- membered heterocyclic, each optionally substituted;
R2 at each occurrence is independently hydrogen, optionally substituted C1-C8 alkyl, or -NRaRb;
Ra at each occurrence is independently hydrogen or optionally substituted C1-C8 alkyl; Rb at each occurance is -C(0)-R6;
Alternatively Ra and Rb can be taken together with the nitrogen atom to which they are attached to form an optionally substituted heterocyclic or optionally substituted heteroaryl group; R3 and R4 are each independently selected from the group consisting of hydrogen, optionally substituted C1-C8 alkyl, optionally substituted C2-C8 alkenyl, and optionally substituted C3-C8 cycloalkyl; alternatively, R3 and R4 can be taken together with the carbon atom to which they are attached to form optionally substituted C3-C8 cycloalkyl or optionally substituted heterocyclic;
R5 is independently hydrogen, optionally substituted C1-C8 alkyl, or optionally substituted C3-C8 cycloalkyl; and
R7a and R7b at each occurrence are each independently selected from the group consisting of hydrogen, optionally substituted aryl, and optionally substituted C1-C4 alkyl; alternatively, CHR7a-U or CHR7b-U can be taken together to form a group selected from CH=CH, optionally substituted C3-C8 cycloalkyl, optionally substituted aryl, or optionally substituted heterocyclic; or yet alternatively, U, R7a, and R7b can be taken together with the carbon atoms to which they are attached to form a bridged, optionally substituted 4- to 7- membered ring selected from the group consisting of C4-C7 cycloalkyl, C4-C7 cycloalkenyl and 4- to 7-membered heterocyclic, each optionally substitut mpound of claim 19, wherein G and J are each independently
Figure imgf000272_0003
Figure imgf000272_0001
wherein said imidazolyl or benzimidazolyl groups are each optionally substituted; or a pharmaceutically acceptable salt thereof.
21. The compound of claim 20, wherein Q is
Figure imgf000272_0002
and B is optionally substituted aryl or optionally substituted heteroaryl; or a pharmaceutically acceptable salt thereof.
22. The compound of claim 21, wherein R6 is independently C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino, or 0(C1-C4 alkyl); or a
pharmaceutically acceptable salt thereof.
23. The compound of claim 22, wherein Rlb and R9a, or Rlb and R9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic; or a pharmaceutically acceptable salt thereof. 24. The compound of claim 22, wherein Rla and R9, or R1 and R9a are taken together with the carbon atom(s) to which they are attached to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or a pharmaceutically acceptable salt thereof.
25. The compound of claim 22, wherein Ring A and L are absent; Ring B is optionally substituted bicyclic aryl or optionally substituted bicyclic heteroaryl; and R6 is C1-C8 alkyl optionally substituted with protected amino or 0(C1-C4 alkyl); or a pharmaceutically acceptable salt thereof.
26. The compound of claim 22, wherein Ring A is absent; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; Ring B is optionally substituted monocyclic or bicyclic aryl or optionally substituted monocyclic or bicyclic heteroaryl; and R6 is C1-C8 alkyl optionally substituted with protected amino or 0(C1-C4 alkyl); or a pharmaceutically acceptable salt thereof.
27. The compound of claim 19 selected from the group of compounds 3-1 to 3-347 compiled in the following tables:
Figure imgf000273_0001
Figure imgf000274_0001
Figure imgf000275_0001
Figure imgf000276_0001
Figure imgf000277_0001
Figure imgf000278_0001
Figure imgf000279_0001
Figure imgf000280_0001
Figure imgf000281_0001
Figure imgf000282_0001
Figure imgf000283_0001
Figure imgf000284_0002
28. A compound represented by Formula (4-1):
Figure imgf000284_0001
or a pharmaceutically acceptable salt thereof, wherein:
Ring A is absent or a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C3-C8 cycloalkyl, and C3-C8 cycloalkenyl, each optionally substituted;
Ring B is a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C3-C8 cycloalkyl, and C3-C8 cycloalkenyl, each optionally substituted;
L is absent or selected from the group consisting of optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, and optionally substituted C2-C4 alkynyl;
R1 at each occurrence is hydrogen or optionally substituted C1-C4 alkyl;
R6 at each occurrence is independently selected from the group consisting of optionally substituted 0(C1-C8 alkyl); optionally substituted amino; C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted;
U is absent or independently selected from the group consisting of O, S, S(O), S02, NC(0)-(C1-C4 alkyl), C(O), protected carbonyl, OCH2, OCH2CH2, SCH2, SCH2CH2, C(R7)2, and C(R7)2C(R7)2;
R7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C1-C4 alkyl), S(C1-C4 alkyl), amino optionally substituted with one or two C1-C4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C1-C4 alkyl; Optionally two geminal R7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring consisting of C3-C8 cycloalkyl, C3-C8 cycloalkenyl and 3- to 8-membered heterocyclic; R7a and R7b at each occurrence are each independently selected from the group consisting of hydrogen, optionally substituted aryl, and optionally substituted C1-C4 alkyl;
alternatively, CHR7a-U or CHR7b-U can be taken together to form a group selected from CH=CH, optionally substituted C3-C8 cycloalkyl, optionally substituted aryl, or optionally substituted heterocyclic;
G is optionally substituted 5-membered heteroaryl or optionally substituted 5/6- member fused heteroaryl; wherein the 5-membered heteroaryl contains one or more nitrogen, and wherein the 6-membered ring of said 5/6-fused membered heteroaryl is attached to group Ring A, L or Ring B and is aryl or heteroaryl;
Q is selected from:
Figure imgf000285_0001
;
Alternatively G and Q are taken together to form
Figure imgf000285_0002
, or
Figure imgf000285_0003
; V is selected from the group consisting of -N(Rlc)-, -N(Rlc)-C(0)-, -N(Rlc)-
C(0)0- and -N(Rlc)-C(0)-N(Rlc)-;
W at each occurrence is independently O or -N(Rlb)-;
Rla, Rlb, Rlc, Rld, R9, and R9a at each occurrence are each independently hydrogen or optionally substituted C1-C4 alkyl; alternatively Rla and R9a, Rla and R9, Rld and R9a, or Rld and R9 can be taken together with the carbon atom(s) to which they are attached to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or yet alternatively Rlb and R9a, or Rlb and R9 can be taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic;
X is absent, O, S, CH2, or CH2CH2;
Y is absent, O, S, C(R , C(R1)2C(R7)2, C(R1)2C(R7)2C(R7)2, C(R1)2OC(R1)2, or C(R^SC(R1)2;
Wherein at least one of X and Y is present;
Wherein at least one of X and Y is not O or S; R2 is hydrogen, optionally substituted C1-C8 alkyl, or -NRaRb;
Ra is hydrogen or optionally substituted C1-C8 alkyl;
Rb is -C(0)-R6;
Alternatively Ra and Rb can be taken together with the nitrogen atom to which they are attached to form an optionally substituted heterocyclic or optionally substituted heteroaryl group;
R3 and R4 are each independently selected from the group consisting of hydrogen, optionally substituted C1-C8 alkyl, optionally substituted C2-C8 alkenyl, and optionally substituted C3-C8 cycloalkyl; alternatively, R3 and R4 can be taken together with the carbon atom to which they are attached to form optionally substituted C3-C8 cycloalkyl or optionally substituted heterocyclic; and
R5 is independently hydrogen, optionally substituted C1-C8 alkyl, or optionally substituted C3-C8 cycloalkyl; preferably hydrogen or optionally substituted C1-C4 alkyl.
29. The compound of claim 28, wherein G is
Figure imgf000286_0003
or
Figure imgf000286_0004
; wherein said imidazolyl or benzimidazolyl groups are each optionally substituted; or a pharmaceutically acceptable salt thereof.
30. The compound of claim 29, wherein Q is
Figure imgf000286_0001
or ; and Ring B is optionally substituted aryl or optionally substituted heteroaryl; or a pharmaceutically acceptable salt thereof.
31. The compound of claim 29, wherein G and Q are taken together to form
Figure imgf000286_0002
; and Ring B is optionally substituted aryl or optionally substituted heteroaryl; or a pharmaceutically acceptable salt thereof.
32. The compound of claim 30, wherein U is CH2; V is -NH- or -NH-C(O)-; R1 is hydrogen; Rlb and R9a, or Rlb and R9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic; R6 is independently C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino, or 0(C1-C4 alkyl); and R7a and R7b at each occurrence are hydrogen; or a pharmaceutically acceptable salt thereof.
33. The compound of claim 31 , wherein U is CH2; V is -NH- or -NH-C(O)-; R1 is hydrogen; Rlb and R9a, or Rlb and R9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic; R6 is independently C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino, or 0(C1-C4 alkyl); and R7a and R7b at each occurrence are hydrogen; or a pharmaceutically acceptable salt thereof.
34. The compound of claim 30, wherein U is CH2; V is -NH- or -NH-C(O)-; R1 is hydrogen; Rla and R9a, Rla and R9, Rld and R9a, or Rld and R9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic; R6 is independently C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino, or 0(C1-C4 alkyl); and R7a and R7b at each occurrence are hydrogen; or a pharmaceutically acceptable salt thereof.
35. The compound of claim 31 , wherein U is CH2; V is -NH- or -NH-C(O)-; R1 is hydrogen; Rla and R9a, Rla and R9, Rld and R9a, or Rld and R9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic; R6 is independently C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino, or 0(C1-C4 alkyl); and R7a and R7b at each occurrence are hydrogen; or a pharmaceutically acceptable salt thereof. 36. The compound of claim 32, wherein Ring A and L are absent; and B is optionally substituted bicyclic aryl or optionally substituted bicyclic heteroaryl; or a
pharmaceutically acceptable salt thereof.
37. The compound of claim 33, wherein Ring A and L are absent; and B is optionally substituted bicyclic aryl or optionally substituted bicyclic heteroaryl; or a
pharmaceutically acceptable salt thereof.
38. The compound of claim 34, wherein Ring A and L are absent; and B is optionally substituted bicyclic aryl or optionally substituted bicyclic heteroaryl; or a
pharmaceutically acceptable salt thereof. 39. The compound of claim 35, wherein Ring A and L are absent; and B is optionally substituted bicyclic aryl or optionally substituted bicyclic heteroaryl; or a
pharmaceutically acceptable salt thereof.
40. The compound of claim 32, wherein Ring A is absent; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and Ring B is phenyl, monocyclic heteroaryl, bicyclic aryl, or bicyclic heteroaryl, each optionally substituted; or a pharmaceutically acceptable salt thereof.
41. The compound of claim 33, wherein Ring A is absent; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and Ring B is phenyl, monocyclic heteroaryl, bicyclic aryl, or bicyclic heteroaryl, each optionally substituted; or a pharmaceutically acceptable salt thereof.
42. The compound of claim 34, wherein Ring A is absent; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and Ring B is phenyl, monocyclic heteroaryl, bicyclic aryl, or bicyclic heteroaryl, each optionally substituted; or a pharmaceutically acceptable salt thereof.
43. The compound of claim 35, wherein Ring A is absent; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; and Ring B is phenyl, monocyclic heteroaryl, bicyclic aryl, or bicyclic heteroaryl, each optionally substituted; or a pharmaceutically acceptable salt thereof.
44. The compound of claim 28 selected from the group of compounds 4-1 to 4-348 compiled in the following tables:
Compounds 4-1 to 4-219
Figure imgf000288_0001
Figure imgf000289_0001
Figure imgf000290_0001
Figure imgf000291_0001
Figure imgf000292_0001
Figure imgf000293_0001
Figure imgf000294_0001
Figure imgf000295_0001
Figure imgf000296_0001
Figure imgf000297_0001
Figure imgf000298_0003
Figure imgf000298_0001
45. A compound represented by Formula (5-1):
Figure imgf000298_0002
or a pharmaceutically acceptable salt thereof, wherein:
Ring A is absent or a monocyclic or polycyclic group independently selected from aryl, heteroaryl, heterocyclic, C3-C8 cycloalkyl, and C3-C8 cycloalkenyl, each optionally substituted;
Ring B is an optionally substituted aryl or optionally substituted heteroaryl;
L is absent or selected from the group consisting of optionally substituted C1-C4 alkyl, optionally substituted C2-C4 alkenyl, and optionally substituted C2-C4 alkynyl;
W at each occurrence is independently O or -N(Rlb)-;
R1, Rla, Rlb, Rlc, R9, and R9a at each occurrence are each independently hydrogen or optionally substituted C C4 alkyl; alternatively Rla and R9a, Rla and R9, R1 and R9a, or R1 and R9 can be taken together with the carbon atom(s) to which they are attached to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or yet alternatively Rlb and R9a, or Rlb and R9 can be taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered heterocyclic;
R6 at each occurrence is independently selected from the group consisting of optionally substituted 0(C1-C8 alkyl); optionally substituted amino; C1-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, heterocyclic, aryl, and heteroaryl, each optionally substituted;
G is optionally substituted 5-membered heteroaryl or optionally substituted 5/6- member fused heteroaryl; wherein the 5-membered heteroaryl contains one or more nitrogen, and wherein the 6-membered ring of said 5/6-fused membered heteroaryl is attached to one of Ring A, L and Ring B and is aryl or heteroaryl;
Q is selected from:
Figure imgf000299_0001
Alternatively G and Q are taken together to form
Figure imgf000299_0002
V is selected from the group consisting of -N(Rlc)-, -N(Rlc)-C(0)-, -N(Rlc)- C(0)0- and -N(Rlc)-C(0)-N(Rlc)-;
X is absent, O, S, CH2, or CH2CH2;
Y is absent, O, S, C(R , C(R1)2C(R7)2, C(R1)2C(R7)2C(R7)2, C(R1)2OC(R1)2, or C(R^SC(R1)2;
Wherein at least one of X and Y is present;
Wherein at least one of X and Y is not O or S;
R7 at each occurrence is independently selected from the group consisting of hydrogen, halogen, cyano, hydroxy, 0(C1-C4 alkyl), S(C1-C4 alkyl), amino optionally substituted with one or two C1-C4 alkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C1-C4 alkyl;
U is absent or selected from O, S, S(O), S02, NC(0)-(d-C4 alkyl), C(O), protected carbonyl, OCH2, OCH2CH2, SCH2, SCH2CH2, C(R7)2, and C(R7)2C(R7)2;
Optionally two geminal R7 groups can be taken together with the carbon atom to which they are attached to form a spiro, optionally substituted 3- to 8-membered ring selected from the group consisting of C3-C8 cycloalkyl, C3-C8 cycloalkenyl and 3- to 8- membered heterocyclic, each optionally substituted;
R2 is hydrogen, optionally substituted C1-C8 alkyl, or -NRaRb;
Ra is hydrogen or optionally substituted C1-C8 alkyl;
Rb is -C(0)-R6; Alternatively Ra and Rb can be taken together with the nitrogen atom to which they are attached to form an optionally substituted heterocyclic or optionally substituted heteroaryl group;
R3 and R4 are each independently selected from the group consisting of hydrogen, optionally substituted C1-C8 alkyl, optionally substituted C2-C8 alkenyl, and optionally substituted C3-C8 cycloalkyl; alternatively, R3 and R4 can be taken together with the carbon atom to which they are attached to form optionally substituted C3-C8 cycloalkyl or optionally substituted heterocyclic;
R5 is independently hydrogen, optionally substituted C1-C8 alkyl, or optionally substituted C3-C8 cycloalkyl; and
R7a and R7b at each occurrence are each independently selected from the group consisting of hydrogen, optionally substituted aryl, and optionally substituted C1-C4 alkyl; alternatively, CHR7a-U or CHR7b-U can be taken together to form a group selected from CH=CH, optionally substituted C3-C8 cycloalkyl, optionally substituted aryl, or optionally substituted heterocyclic; or yet alternatively, U, R7a , and R7b can be taken together with the carbon atoms to which they are attached to form a bridged, optionally substituted 4- to 7-membered ring selected from the group consisting of C4-C7 cycloalkyl, C4-C7 cycloalkenyl and 4- to 7- membered heterocyclic, each optionally substituted.
46. The compound of claim 45, wherein G is
Figure imgf000300_0003
or
Figure imgf000300_0004
; wherein said imidazolyl or benzimidazolyl groups are each optionally substituted; or a pharmaceutically acceptable salt thereof.
47. The compound of claim 46, wherein Q is
Figure imgf000300_0001
and R6 is independently C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino, or 0(C1-C4 alkyl); or a pharmaceutically acceptable salt thereof.
The compound of claim 45, wherein G and Q are taken together to form
Figure imgf000300_0002
; V is -NH- or -NH-C(O)-; and R6 is independently C1-C8 alkyl optionally substituted with amino, hydroxy, protected amino, or 0(C1-C4 alkyl); or a pharmaceutically acceptable salt thereof.
49. The compound of claim 47, wherein Rlb and R9a, or Rlb and R9 are taken together with the nitrogen or carbon atom(s) to which they are attached to form an optionally substituted 4- to 8-membered monocyclic, or fused or bridged bicyclic heterocyclic; or a pharmaceutically acceptable salt thereof.
50. The compound of claim 47, wherein Rla and R9, or R1 and R9a are taken together with the carbon atom(s) to which they are attached to form an optionally substituted C3-C8 cycloalkyl or optionally substituted 4- to 8-membered heterocyclic; or a pharmaceutically acceptable salt thereof.
51. The compound of claim 47, wherein L is absent; one of Ring A and Ring B is optionally substituted monocyclic aryl or heteroaryl; and the other one of Ring A and Ring B is optionally substituted monocyclic or bicyclic aryl or heteroaryl; and R6 is
independently C1-C8 alkyl optionally substituted with protected amino or 0(C1-C4 alkyl); or a pharmaceutically acceptable salt thereof.
52. The compound of claim 47, wherein Ring A is absent; L is optionally substituted C2-C4 alkenyl or optionally substituted C2-C4 alkynyl; Ring B is phenyl, monocyclic heteroaryl, bicyclic aryl, or bicyclic heteroaryl, each optionally substituted; and R6 is independently C1-C8 alkyl optionally substituted with protected amino or 0(C1-C4 alkyl); or a pharmaceutically acceptable salt thereof.
53. The compound of claim 48 selected from the group of compounds 5-1 to 5-378 and 5-381 to 5-394 compiled in the following tables:
Figure imgf000301_0001
Figure imgf000302_0001
Figure imgf000303_0001
Figure imgf000304_0001
Figure imgf000305_0001
Figure imgf000306_0001
Figure imgf000307_0001
Figure imgf000308_0001
Figure imgf000310_0001
Figure imgf000311_0001
Figure imgf000312_0002
Compounds 5-372 to 5-378 and 5-381 to 5-394
Figure imgf000312_0001
Figure imgf000313_0001
Figure imgf000314_0001
54. A pharmaceutical composition comprising a compound or a combination of compounds according to any one of claims 1 to 53 or a pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable carrier or excipient.
55. A method of inhibiting the replication of an R A-containing virus comprising contacting said virus with a therapeutically effective amount of a compound or combination of compounds of any one of claims 1 to 53, or a pharmaceutically acceptable salt thereof.
56. A method of treating or preventing infection caused by an RNA-containing virus comprising administering to a subject in need of such treatment a therapeutically effective amount of a compound or combination of compounds of any one of claims 1 to 53, or a pharmaceutically acceptable salt thereof.
57. The method of claim 56, wherein the RNA-containing virus is hepatitis C virus.
58. The method of claim 56, further comprising the step of co-administering one or more agents selected from the group consisting of a host immune modulator, a second or more antiviral agents, and or a combination thereof.
59. The method of claim 58, wherein the host immune modulator is selected from the group consisting of interferon-alpha, pegylated-interferon-alpha, interferon-beta, interferon-gamma, consensus interferon, a cytokine, and a vaccine.
60. The method of claim 58, wherein the second or more antiviral agents inhibit replication of HCV by inhibiting host cellular functions associated with viral replication.
61. The method of claim 58, wherein the second or more antiviral agents inhibit the replication of HCV by targeting proteins of the viral genome.
62. The method of claim 61, wherein said targeting protein is selected from the group consisting of helicase, protease, polymerase, metalloprotease, NS4A, NS4B, NS5A, assembly, entry, and IRES.
63. The method of claim 58, further comprising the step of co-administering an agent or combination of agents that treat or alleviate symptoms of HCV infection including cirrhosis and inflammation of the liver.
64. The pharmaceutical composition of claim 54, further comprising an agent selected from interferon, pegylated interferon, ribavirin, amantadine, an HCV protease inhibitor, an HCV polymerase inhibitor, an HCV helicase inhibitor, or an internal ribosome entry site inhibitor.
65. The composition of claim 54, further comprising a cytochrome P450
monooxygenase inhibitor or a pharmaceutically acceptable salt thereof.
66. The composition of claim 65, wherein the cytochrome P450 mooxygenase inhibitor is ritonavir.
67. A method of co-adminstering to a subject in need of anti-hepatitis C viral treatment comprising administering to said subject a cytochrome P450 monooxygenase inhibitor or a pharmaceutically acceptable salt thereof and a compound of any one of claims 1 to 53 or a pharmaceutically acceptable salt thereof.
68. A process of making a compound according to claim 1 comprising:
i) preparing a compound of Formula (l-II):
Figure imgf000315_0001
via a transition-metal catalyzed cross-coupling reaction; wherein:
G, J, U, ft1, Rla, Rlb, R7a, R7b, Pv9, and R9a are as defined in claim 1; Ring A1 is absent, optionally substituted aryl or optionally substituted heteroaryl;
Ring B1 is optionally substituted aryl or optionally substituted heteroaryl;
L1 is absent, optionally substituted C2-C4 alkenyl or C2-C4 alkynyl; and
Za and Zb are each independently an amino protecting group or -C(0)-R6; wherein R6 is as defined in claim 1 ;
ii) when Za or Zb is an amino protecting group, fully or selectively deprotecting a compound of Formula (l-II) to give the corresponding amine of Formula (l-III):
Figure imgf000316_0001
wherein Zc is hydrogen, an amino protecting group or -C(0)-R6;
iii) capping the released amino group of a compound of Formula (l-III) with LG-
C(0)-R6,
wherein LG is a leaving group; to give the compound of Formula (1-IV):
Figure imgf000316_0002
wherein Zd is an amino protecting group -C(0)-R6; and
iv) repeated reaction sequence of deprotecting and capping (step ii-iii) to give the compound of Formula (1-V):
Figure imgf000316_0003
69. A process of making a compound according to claim 10 comprising:
i) preparing a compound of Formula (2-II):
Figure imgf000316_0004
via a transition-metal catalyzed cross-coupling reaction; wherein:
G, J, U, R1, Rla, Rlb, R7a, R7b, R9, and R9a are as defined in claim 10;
Ring A1 and Ring B1 are each independently optionally substituted aryl or optionally substituted heteroaryl;
L1 is absent, optionally substituted C2-C4 alkenyl or C2-C4 alkynyl; and Za and Zb are each independently an amino protecting group or -C(0)-R6; wherein R6 is as defined in claim 10;
ii) when Za or Zb is an amino protecting group, fully or selectively deprotecting a compound of Formula 2-II) to give the corresponding amine of Formula (2-III):
Figure imgf000317_0001
wherein Zc is hydrogen, an amino protecting group or -C(0)-R6;
iii) capping the released amino group of a compound of Formula (2-III) with LG-
C(0)-R6,
wherein LG is a leavin group; to give the compound of Formula (2-IV):
Figure imgf000317_0002
wherein Zd is an amino protecting group -C(0)-R6; and
iv) Repeated reaction sequence of deprotecting and capping (step ii-iii) to give the compound of Formula 2-V):
Figure imgf000317_0003
70. A process of making a compound according to one or more of claims 19 to 27 comprising:
i) preparing a compound of Formula (3 -II):
Figure imgf000317_0004
via a transition-metal catalyzed cross-coupling reaction; wherein:
G, J, U, R1, Rla, Rlb, R7a, R7b, R9, and R9a are as defined in claim 19;
Ring A1 and Ring B1 are each independently absent or an optionally substituted aryl or optionally substituted heteroaryl;
L1 is absent, optionally substituted C2-C4 alkenyl or C2-C4 alkynyl; and
Za and Zb are each independently an amino protecting group or -C(0)-R6; wherein R6 is as defined in claim 19; ii) when Za or Zb is an amino protecting group, fully or selectively deprotecting a compound of Formula (3 -II) to give the corresponding amine of Formula (3 -III):
Figure imgf000318_0001
wherein Zc is hydrogen, an amino protecting group or -C(0)-R6;
iii) capping the released amino group of a compound of Formula (3 -III) with LG- C(0)-R6,
wherein LG is a leaving group; to give the compound of Formula (3-IV):
Figure imgf000318_0002
wherein Zd is an amino protecting group -C(0)-R6; and
iv) repeated reaction sequence of deprotecting and capping (step ii-iii) to give the compound of Formula (3-V):
Figure imgf000318_0003
71. A process of making a compound according to claim 28 comprising:
i) preparing a compound of Formula (4-II):
Figure imgf000318_0004
via a transition-metal catalyzed cross-coupling reaction;
wherein:
G, U, R1, Rla, Rlb, Rlc, R7a, R7b, R9, and R9a are as defined in claim 28;
Ring A1 is absent, or optionally substituted aryl or optionally substituted heteroaryl;
Ring B1 is optionally substituted aryl or optionally substituted heteroaryl;
L1 is absent, or optionally substituted C2-C4 alkenyl or C2-C4 alkynyl; and
Za or Zb are each independently an amino protecting group or -C(0)-R6; wherein R6 is as defined in claim 28;
ii) when Za or Zb is an amino protecting group, fully or selectively deprotecting a compound of Formula (4-II) to give the corresponding amine of Formula (4-III):
Figure imgf000319_0001
wherein Zc is hydrogen, an amino protecting group or -C(0)-R6;
iii) capping the released amino group of a compound of Formula (4-III) with LG- C(0)-R6,
wherein LG is a leaving group; to give the compound of Formula (4-IV):
Figure imgf000319_0002
wherein Zd is an amino protecting group or -C(0)-R6; and
iv) repeated reaction sequence of deprotecting and capping (step ii-iii) to g
compo nd of Formula (4-V):
Figure imgf000319_0003
A process of making a compound according to claim 45 comprising:
i) preparing a compound of Formula (5-II):
Figure imgf000319_0004
via a transition-metal catalyzed cross-coupling reaction; wherein:
Ring B, G, U, R1, Rla, Rlb, Rlc, R7a, R7b, R9, and R9a are as defined in claim 45;
Ring A1 is absent, optionally substituted aryl or optionally substituted heteroaryl;
L1 is absent, optionally substituted C2-C4 alkenyl or C2-C4 alkynyl; and
Za and Zb are each independently an amino protecting group or -C(0)-R6; wherein R6 is as defined in claim 45;
ii) when Za or Zb is an amino protecting group, fully or selectively deprotecting a compound of Formula (5-II) to give the corresponding amine of Formula (5-III):
.
Figure imgf000319_0005
wherein Zc is hydrogen, an amino protecting group or -C(0)-R6;
iii) capping the released amino group of a compound of Formula (5-III) with LG- C(0)-R6,
wherein LG is a leaving group; to give the compound of Formula 5-IV):
Figure imgf000320_0001
wherein Zd is an amino protecting group or -C(0)-R6; and
iv) repeated reaction sequence of deprotecting and capping (step ii-iii) to give the compound of Formula (5-V):
Figure imgf000320_0002
PCT/US2010/048377 2009-09-11 2010-09-10 Hepatitis c virus inhibitors WO2011031934A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP10816134.0A EP2475254A4 (en) 2009-09-11 2010-09-10 Hepatitis c virus inhibitors

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US24159509P 2009-09-11 2009-09-11
US24148909P 2009-09-11 2009-09-11
US24157709P 2009-09-11 2009-09-11
US24161709P 2009-09-11 2009-09-11
US24157809P 2009-09-11 2009-09-11
US61/241,617 2009-09-11
US61/241,595 2009-09-11
US61/241,578 2009-09-11
US61/241,489 2009-09-11
US61/241,577 2009-09-11

Publications (1)

Publication Number Publication Date
WO2011031934A1 true WO2011031934A1 (en) 2011-03-17

Family

ID=43732807

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/048377 WO2011031934A1 (en) 2009-09-11 2010-09-10 Hepatitis c virus inhibitors

Country Status (2)

Country Link
EP (1) EP2475254A4 (en)
WO (1) WO2011031934A1 (en)

Cited By (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100215616A1 (en) * 2009-02-17 2010-08-26 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
WO2011146401A1 (en) * 2010-05-17 2011-11-24 Intermune, Inc. Novel inhibitors of hepatitis c virus replication
US8088368B2 (en) 2009-05-13 2012-01-03 Gilead Sciences, Inc. Antiviral compounds
US8101643B2 (en) 2009-02-27 2012-01-24 Enanta Pharmaceuticals, Inc. Benzimidazole derivatives
US8138215B2 (en) 2009-05-29 2012-03-20 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8143301B2 (en) 2009-04-09 2012-03-27 Bristol Myers Squibb Company Hepatitis C virus inhibitors
US8143414B2 (en) 2009-04-13 2012-03-27 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8188132B2 (en) 2009-02-17 2012-05-29 Enanta Pharmaceuticals, Inc. Linked dibenzimidazole derivatives
WO2012087976A2 (en) * 2010-12-21 2012-06-28 Intermune, Inc. Novel inhibitors of hepatitis c virus replication
US8211928B2 (en) 2009-05-29 2012-07-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8221737B2 (en) 2009-06-16 2012-07-17 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US8242156B2 (en) 2009-02-17 2012-08-14 Enanta Pharmaceuticals, Inc. Linked dibenzimidazole derivatives
US8314135B2 (en) 2009-02-09 2012-11-20 Enanta Pharmaceuticals, Inc. Linked dibenzimidazole antivirals
US8362020B2 (en) 2009-12-30 2013-01-29 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8377980B2 (en) 2009-12-16 2013-02-19 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8394968B2 (en) 2009-02-17 2013-03-12 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2012072713A3 (en) * 2010-11-30 2013-04-04 Oryzon Genomics, S.A. Lysine demethylase inhibitors such as cyclylcyclopropanamine derivatives for use in the treatment of diseases and disorders associated with flaviviridae
EP2575866A1 (en) * 2010-05-24 2013-04-10 Presidio Pharmaceuticals, Inc. Inhibitors of hcv ns5a
WO2013079942A1 (en) * 2011-11-30 2013-06-06 The University Of Sheffield Catalyst compounds
WO2013095275A1 (en) * 2011-12-20 2013-06-27 Medivir Ab Novel hepatitis c virus inhibitors
US8524717B2 (en) 2008-10-17 2013-09-03 Oryzon Genomics, S.A. Oxidase inhibitors and their use
US8541424B2 (en) 2008-12-23 2013-09-24 Abbott Laboratories Anti-viral compounds
US8546405B2 (en) 2008-12-23 2013-10-01 Abbott Laboratories Anti-viral compounds
US8552047B2 (en) 2011-02-07 2013-10-08 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8618153B2 (en) 2009-11-12 2013-12-31 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8673954B2 (en) 2009-02-27 2014-03-18 Enanta Pharmaceuticals, Inc. Benzimidazole derivatives
US8686026B2 (en) 2010-06-10 2014-04-01 Abbvie Inc. Solid compositions
US8691938B2 (en) 2009-06-11 2014-04-08 Abbvie Inc. Anti-viral compounds
US8697704B2 (en) 2010-08-12 2014-04-15 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US8716454B2 (en) 2009-06-11 2014-05-06 Abbvie Inc. Solid compositions
US8722743B2 (en) 2010-04-19 2014-05-13 Oryzon Genomics S.A. Lysine specific demethylase-1 inhibitors and their use
US8778938B2 (en) 2010-06-04 2014-07-15 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US8785487B2 (en) 2010-01-25 2014-07-22 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US8796466B2 (en) 2009-03-30 2014-08-05 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8859555B2 (en) 2009-09-25 2014-10-14 Oryzon Genomics S.A. Lysine Specific Demethylase-1 inhibitors and their use
CN104230946A (en) * 2013-06-06 2014-12-24 爱博新药研发(上海)有限公司 Compounds for inhibiting hepatitis C virus, pharmaceutical compositions of compounds and applications of pharmaceutical compositions
US8921372B2 (en) 2010-11-04 2014-12-30 Theravance Biopharma R&D Ip, Llc Inhibitors of hepatitis C virus
US8927739B2 (en) 2011-05-18 2015-01-06 Enanta Pharmaceuticals, Inc. Processes for the preparation of 5-azaspiro[2.4]heptane-6-carboxylic acid and its derivatives
US8933110B2 (en) 2010-01-25 2015-01-13 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US8937150B2 (en) 2009-06-11 2015-01-20 Abbvie Inc. Anti-viral compounds
US8937303B2 (en) 2010-10-22 2015-01-20 Commonwealth Scientific And Industrial Research Organisation Organic electroluminescent device
US8946296B2 (en) 2009-10-09 2015-02-03 Oryzon Genomics S.A. Substituted heteroaryl- and aryl-cyclopropylamine acetamides and their use
JP2015508088A (en) * 2012-02-24 2015-03-16 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Antiviral compounds
US8993808B2 (en) 2009-01-21 2015-03-31 Oryzon Genomics, S.A. Phenylcyclopropylamine derivatives and their medical use
US9006449B2 (en) 2010-07-29 2015-04-14 Oryzon Genomics, S.A. Cyclopropylamine derivatives useful as LSD1 inhibitors
US9006455B2 (en) 2009-11-11 2015-04-14 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9034832B2 (en) 2011-12-29 2015-05-19 Abbvie Inc. Solid compositions
JP2015514807A (en) * 2012-04-25 2015-05-21 セラヴァンス バイオファーマ アール&ディー アイピー, エルエルシー Piperazine-piperidine compounds as hepatitis C virus inhibitors
US9061966B2 (en) 2010-10-08 2015-06-23 Oryzon Genomics S.A. Cyclopropylamine inhibitors of oxidases
US9073943B2 (en) 2012-02-10 2015-07-07 Lupin Limited Antiviral compounds with a dibenzooxaheterocycle moiety
US9127021B2 (en) 2010-04-09 2015-09-08 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US9181198B2 (en) 2010-07-29 2015-11-10 Oryzon Genomics S.A. Arylcyclopropylamine based demethylase inhibitors of LSD1 and their medical use
US9186337B2 (en) 2010-02-24 2015-11-17 Oryzon Genomics S.A. Lysine demethylase inhibitors for diseases and disorders associated with Hepadnaviridae
US9278922B2 (en) 2009-04-15 2016-03-08 Abbvie Inc. Anti-viral compounds
US9326973B2 (en) 2012-01-13 2016-05-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9333204B2 (en) 2014-01-03 2016-05-10 Abbvie Inc. Solid antiviral dosage forms
US9393256B2 (en) 2011-09-16 2016-07-19 Gilead Pharmasset Llc Methods for treating HCV
US9394279B2 (en) 2009-06-11 2016-07-19 Abbvie Inc. Anti-viral compounds
US9469597B2 (en) 2011-10-20 2016-10-18 Oryzon Genomics S.A. (Hetero)aryl cyclopropylamine compounds as LSD1 inhibitors
US9487512B2 (en) 2011-10-20 2016-11-08 Oryzon Genomics S.A. (Hetero)aryl cyclopropylamine compounds as LSD1 inhibitors
US9546160B2 (en) 2011-05-12 2017-01-17 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9616058B2 (en) 2010-02-24 2017-04-11 Oryzon Genomics, S.A. Potent selective LSD1 inhibitors and dual LSD1/MAO-B inhibitors for antiviral use
US9717712B2 (en) 2013-07-02 2017-08-01 Bristol-Myers Squibb Company Combinations comprising tricyclohexadecahexaene derivatives for use in the treatment of hepatitis C virus
US9765087B2 (en) 2009-02-27 2017-09-19 Enanta Pharmaceuticals, Inc. Benzimidazole derivatives
US9770439B2 (en) 2013-07-02 2017-09-26 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9776981B2 (en) 2009-11-11 2017-10-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9775831B2 (en) 2013-07-17 2017-10-03 Bristol-Myers Squibb Company Combinations comprising biphenyl derivatives for use in the treatment of HCV
US9908859B2 (en) 2011-02-08 2018-03-06 Oryzon Genomics, S.A. Lysine demethylase inhibitors for myeloproliferative disorders
US10039779B2 (en) 2013-01-31 2018-08-07 Gilead Pharmasset Llc Combination formulation of two antiviral compounds
US10086011B2 (en) 2013-08-27 2018-10-02 Gilead Pharmasset Llc Combination formulation of two antiviral compounds
US10201584B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
US10617675B2 (en) 2015-08-06 2020-04-14 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
RU2723482C1 (en) * 2019-10-22 2020-06-11 Андрей Александрович Иващенко Pangenotypic ns5a protein inhibitor of hepatitis c virus, a pharmaceutical composition and methods for preparing and using
US10710986B2 (en) 2018-02-13 2020-07-14 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US10774071B2 (en) 2018-07-13 2020-09-15 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US10899735B2 (en) 2018-04-19 2021-01-26 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
WO2021249364A1 (en) * 2020-06-12 2021-12-16 Wang, Jinping Novel analogs of pterostilbene amino acid bearing carbonates for treating a non-alcoholic fatty liver disease and nonalcoholic steatohepatitis
US11236085B2 (en) 2018-10-24 2022-02-01 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US11247987B2 (en) 2017-10-06 2022-02-15 Forma Therapeutics, Inc. Inhibiting ubiquitin specific peptidase 30
US11484534B2 (en) 2013-03-14 2022-11-01 Abbvie Inc. Methods for treating HCV
US11535618B2 (en) 2018-10-05 2022-12-27 Forma Therapeutics, Inc. Fused pyrrolines which act as ubiquitin-specific protease 30 (USP30) inhibitors
US11667651B2 (en) 2017-12-22 2023-06-06 Hibercell, Inc. Aminopyridine derivatives as phosphatidylinositol phosphate kinase inhibitors
WO2023243601A1 (en) * 2022-06-13 2023-12-21 モジュラス株式会社 Azacycloalkyl carbonyl cyclic amine compound

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US68140A (en) * 1867-08-27 John zoocerman
US20040039043A1 (en) * 2000-07-24 2004-02-26 Hans-Ludwig Elbe Biphenyl carboxamides
US20080044380A1 (en) * 2006-08-11 2008-02-21 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US20080044379A1 (en) * 2006-08-11 2008-02-21 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US20080050336A1 (en) * 2006-08-11 2008-02-28 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US20090202478A1 (en) * 2008-02-13 2009-08-13 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US20090202483A1 (en) * 2008-02-13 2009-08-13 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8143288B2 (en) * 2005-06-06 2012-03-27 Bristol-Myers Squibb Company Inhibitors of HCV replication
KR20080050490A (en) * 2005-09-16 2008-06-05 애로우 쎄라퓨틱스 리미티드 Biphenyl derivatives and their use in treating hepatitis c
JP5419685B2 (en) * 2006-05-16 2014-02-19 ファーマサイエンス・インコーポレイテッド IAPBIR domain binding protein
AU2009322400A1 (en) * 2008-12-03 2011-06-30 Presidio Pharmaceuticals, Inc. Inhibitors of HCV NS5A
EA201490853A1 (en) * 2009-05-13 2014-09-30 Джилид Сайэнс, Инк. ANTI-VIRUS COMPOUNDS

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US68140A (en) * 1867-08-27 John zoocerman
US20040039043A1 (en) * 2000-07-24 2004-02-26 Hans-Ludwig Elbe Biphenyl carboxamides
US20080044380A1 (en) * 2006-08-11 2008-02-21 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US20080044379A1 (en) * 2006-08-11 2008-02-21 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US20080050336A1 (en) * 2006-08-11 2008-02-28 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US20090202478A1 (en) * 2008-02-13 2009-08-13 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US20090202483A1 (en) * 2008-02-13 2009-08-13 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2475254A4 *

Cited By (122)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8524717B2 (en) 2008-10-17 2013-09-03 Oryzon Genomics, S.A. Oxidase inhibitors and their use
US8546405B2 (en) 2008-12-23 2013-10-01 Abbott Laboratories Anti-viral compounds
US8541424B2 (en) 2008-12-23 2013-09-24 Abbott Laboratories Anti-viral compounds
US9163017B2 (en) 2008-12-23 2015-10-20 Abbvie Inc. Anti-viral compounds
US9249138B2 (en) 2008-12-23 2016-02-02 Abbvie Inc. Anti-viral compounds
US8993808B2 (en) 2009-01-21 2015-03-31 Oryzon Genomics, S.A. Phenylcyclopropylamine derivatives and their medical use
US8314135B2 (en) 2009-02-09 2012-11-20 Enanta Pharmaceuticals, Inc. Linked dibenzimidazole antivirals
US8242156B2 (en) 2009-02-17 2012-08-14 Enanta Pharmaceuticals, Inc. Linked dibenzimidazole derivatives
US8188132B2 (en) 2009-02-17 2012-05-29 Enanta Pharmaceuticals, Inc. Linked dibenzimidazole derivatives
US20100215616A1 (en) * 2009-02-17 2010-08-26 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US8809548B2 (en) * 2009-02-17 2014-08-19 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8394968B2 (en) 2009-02-17 2013-03-12 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9765087B2 (en) 2009-02-27 2017-09-19 Enanta Pharmaceuticals, Inc. Benzimidazole derivatives
US8673954B2 (en) 2009-02-27 2014-03-18 Enanta Pharmaceuticals, Inc. Benzimidazole derivatives
US8101643B2 (en) 2009-02-27 2012-01-24 Enanta Pharmaceuticals, Inc. Benzimidazole derivatives
US8796466B2 (en) 2009-03-30 2014-08-05 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8143301B2 (en) 2009-04-09 2012-03-27 Bristol Myers Squibb Company Hepatitis C virus inhibitors
US8143414B2 (en) 2009-04-13 2012-03-27 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9278922B2 (en) 2009-04-15 2016-03-08 Abbvie Inc. Anti-viral compounds
US8822430B2 (en) 2009-05-13 2014-09-02 Gilead Pharmasset Llc Antiviral compounds
US8273341B2 (en) 2009-05-13 2012-09-25 Gilead Sciences, Inc. Antiviral compounds
US8841278B2 (en) 2009-05-13 2014-09-23 Gilead Pharmasset Llc Antiviral compounds
US8669234B2 (en) 2009-05-13 2014-03-11 Gilead Sciences, Inc. Antiviral compounds
US9511056B2 (en) 2009-05-13 2016-12-06 Gilead Pharmasset Llc Antiviral compounds
US9981955B2 (en) 2009-05-13 2018-05-29 Gilead Pharmasset Llc Antiviral compounds
US8088368B2 (en) 2009-05-13 2012-01-03 Gilead Sciences, Inc. Antiviral compounds
US8138215B2 (en) 2009-05-29 2012-03-20 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8211928B2 (en) 2009-05-29 2012-07-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9394279B2 (en) 2009-06-11 2016-07-19 Abbvie Inc. Anti-viral compounds
US10028937B2 (en) 2009-06-11 2018-07-24 Abbvie Inc. Anti-viral compounds
US9586978B2 (en) 2009-06-11 2017-03-07 Abbvie Inc. Anti-viral compounds
US8691938B2 (en) 2009-06-11 2014-04-08 Abbvie Inc. Anti-viral compounds
US10039754B2 (en) 2009-06-11 2018-08-07 Abbvie Inc. Anti-viral compounds
US8716454B2 (en) 2009-06-11 2014-05-06 Abbvie Inc. Solid compositions
US8921514B2 (en) 2009-06-11 2014-12-30 Abbvie Inc. Anti-viral compounds
US8937150B2 (en) 2009-06-11 2015-01-20 Abbvie Inc. Anti-viral compounds
US8221737B2 (en) 2009-06-16 2012-07-17 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US8859555B2 (en) 2009-09-25 2014-10-14 Oryzon Genomics S.A. Lysine Specific Demethylase-1 inhibitors and their use
US8946296B2 (en) 2009-10-09 2015-02-03 Oryzon Genomics S.A. Substituted heteroaryl- and aryl-cyclopropylamine acetamides and their use
US9006455B2 (en) 2009-11-11 2015-04-14 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9776981B2 (en) 2009-11-11 2017-10-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8618153B2 (en) 2009-11-12 2013-12-31 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8377980B2 (en) 2009-12-16 2013-02-19 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8735398B2 (en) 2009-12-30 2014-05-27 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8362020B2 (en) 2009-12-30 2013-01-29 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8785487B2 (en) 2010-01-25 2014-07-22 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US8933110B2 (en) 2010-01-25 2015-01-13 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US9186337B2 (en) 2010-02-24 2015-11-17 Oryzon Genomics S.A. Lysine demethylase inhibitors for diseases and disorders associated with Hepadnaviridae
US9616058B2 (en) 2010-02-24 2017-04-11 Oryzon Genomics, S.A. Potent selective LSD1 inhibitors and dual LSD1/MAO-B inhibitors for antiviral use
US9127021B2 (en) 2010-04-09 2015-09-08 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US9149447B2 (en) 2010-04-19 2015-10-06 Oryzon Genomics S.A. Lysine specific demethylase-1 inhibitors and their use
US8722743B2 (en) 2010-04-19 2014-05-13 Oryzon Genomics S.A. Lysine specific demethylase-1 inhibitors and their use
US10202330B2 (en) 2010-04-19 2019-02-12 Oryzon Genomics, Sa Lysine specific demethylase-1 inhibitors and their use
WO2011146401A1 (en) * 2010-05-17 2011-11-24 Intermune, Inc. Novel inhibitors of hepatitis c virus replication
EP2575866A4 (en) * 2010-05-24 2013-10-16 Presidio Pharmaceuticals Inc Inhibitors of hcv ns5a
EP2575866A1 (en) * 2010-05-24 2013-04-10 Presidio Pharmaceuticals, Inc. Inhibitors of hcv ns5a
US8877707B2 (en) 2010-05-24 2014-11-04 Presidio Pharmaceuticals, Inc. Inhibitors of HCV NS5A
US8778938B2 (en) 2010-06-04 2014-07-15 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US8686026B2 (en) 2010-06-10 2014-04-01 Abbvie Inc. Solid compositions
US10233178B2 (en) 2010-07-29 2019-03-19 Oryzon Genomics, S.A. Arylcyclopropylamine based demethylase inhibitors of LSD1 and their medical use
US9708309B2 (en) 2010-07-29 2017-07-18 Oryzon Genomics, S.A. Arylcyclopropylamine based demethylase inhibitors of LSD1 and their medical use
US9181198B2 (en) 2010-07-29 2015-11-10 Oryzon Genomics S.A. Arylcyclopropylamine based demethylase inhibitors of LSD1 and their medical use
US9006449B2 (en) 2010-07-29 2015-04-14 Oryzon Genomics, S.A. Cyclopropylamine derivatives useful as LSD1 inhibitors
US9676701B2 (en) 2010-07-29 2017-06-13 Oryzon Genomics, S.A. Cyclopropylamine derivatives useful as LSD1 inhibitors
US8697704B2 (en) 2010-08-12 2014-04-15 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US9061966B2 (en) 2010-10-08 2015-06-23 Oryzon Genomics S.A. Cyclopropylamine inhibitors of oxidases
US8937303B2 (en) 2010-10-22 2015-01-20 Commonwealth Scientific And Industrial Research Organisation Organic electroluminescent device
US8921372B2 (en) 2010-11-04 2014-12-30 Theravance Biopharma R&D Ip, Llc Inhibitors of hepatitis C virus
US9260414B2 (en) 2010-11-04 2016-02-16 Theravance Biopharma R&D Ip, Llc Inhibitors of hepatitic C virus
WO2012072713A3 (en) * 2010-11-30 2013-04-04 Oryzon Genomics, S.A. Lysine demethylase inhibitors such as cyclylcyclopropanamine derivatives for use in the treatment of diseases and disorders associated with flaviviridae
US9790196B2 (en) 2010-11-30 2017-10-17 Oryzon Genomics S.A. Lysine demethylase inhibitors for diseases and disorders associated with Flaviviridae
WO2012087976A3 (en) * 2010-12-21 2012-11-29 Intermune, Inc. Novel inhibitors of hepatitis c virus replication
WO2012087976A2 (en) * 2010-12-21 2012-06-28 Intermune, Inc. Novel inhibitors of hepatitis c virus replication
US9340520B2 (en) 2011-02-07 2016-05-17 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8552047B2 (en) 2011-02-07 2013-10-08 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9908859B2 (en) 2011-02-08 2018-03-06 Oryzon Genomics, S.A. Lysine demethylase inhibitors for myeloproliferative disorders
US9546160B2 (en) 2011-05-12 2017-01-17 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US10201541B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
US10201584B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
US8927739B2 (en) 2011-05-18 2015-01-06 Enanta Pharmaceuticals, Inc. Processes for the preparation of 5-azaspiro[2.4]heptane-6-carboxylic acid and its derivatives
US10456414B2 (en) 2011-09-16 2019-10-29 Gilead Pharmasset Llc Methods for treating HCV
US9393256B2 (en) 2011-09-16 2016-07-19 Gilead Pharmasset Llc Methods for treating HCV
US9469597B2 (en) 2011-10-20 2016-10-18 Oryzon Genomics S.A. (Hetero)aryl cyclopropylamine compounds as LSD1 inhibitors
US9670136B2 (en) 2011-10-20 2017-06-06 Oryzon Genomics S.A. (Hetero)aryl cyclopropylamine compounds as LSD1 inhibitors
US10329256B2 (en) 2011-10-20 2019-06-25 Oryzon Genomics, S.A. (Hetero)aryl cyclopropylamine compounds as LSD1 inhibitors
US9487512B2 (en) 2011-10-20 2016-11-08 Oryzon Genomics S.A. (Hetero)aryl cyclopropylamine compounds as LSD1 inhibitors
US10214477B2 (en) 2011-10-20 2019-02-26 Oryzon Genomics S.A. (Hetero)aryl cyclopropylamine compounds as LSD1 inhibitors
US9944601B2 (en) 2011-10-20 2018-04-17 Oryzon Genomics, S.A. (Hetero)aryl cyclopropylamine compounds as LSD1 inhibitors
WO2013079942A1 (en) * 2011-11-30 2013-06-06 The University Of Sheffield Catalyst compounds
WO2013095275A1 (en) * 2011-12-20 2013-06-27 Medivir Ab Novel hepatitis c virus inhibitors
US9034832B2 (en) 2011-12-29 2015-05-19 Abbvie Inc. Solid compositions
US9326973B2 (en) 2012-01-13 2016-05-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9073943B2 (en) 2012-02-10 2015-07-07 Lupin Limited Antiviral compounds with a dibenzooxaheterocycle moiety
US9073942B2 (en) 2012-02-10 2015-07-07 Lupin Limited Antiviral compounds with a heterotricycle moiety
JP2015508088A (en) * 2012-02-24 2015-03-16 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Antiviral compounds
JP2015514807A (en) * 2012-04-25 2015-05-21 セラヴァンス バイオファーマ アール&ディー アイピー, エルエルシー Piperazine-piperidine compounds as hepatitis C virus inhibitors
US10039779B2 (en) 2013-01-31 2018-08-07 Gilead Pharmasset Llc Combination formulation of two antiviral compounds
US11484534B2 (en) 2013-03-14 2022-11-01 Abbvie Inc. Methods for treating HCV
CN104230946A (en) * 2013-06-06 2014-12-24 爱博新药研发(上海)有限公司 Compounds for inhibiting hepatitis C virus, pharmaceutical compositions of compounds and applications of pharmaceutical compositions
CN104230946B (en) * 2013-06-06 2017-03-08 爱博新药研发(上海)有限公司 The compound of suppression hepatitis C virus, pharmaceutical composition and its application
US9770439B2 (en) 2013-07-02 2017-09-26 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9717712B2 (en) 2013-07-02 2017-08-01 Bristol-Myers Squibb Company Combinations comprising tricyclohexadecahexaene derivatives for use in the treatment of hepatitis C virus
US9775831B2 (en) 2013-07-17 2017-10-03 Bristol-Myers Squibb Company Combinations comprising biphenyl derivatives for use in the treatment of HCV
US10086011B2 (en) 2013-08-27 2018-10-02 Gilead Pharmasset Llc Combination formulation of two antiviral compounds
US11116783B2 (en) 2013-08-27 2021-09-14 Gilead Pharmasset Llc Combination formulation of two antiviral compounds
US11707479B2 (en) 2013-08-27 2023-07-25 Gilead Sciences, Inc. Combination formulation of two antiviral compounds
US10105365B2 (en) 2014-01-03 2018-10-23 Abbvie Inc. Solid antiviral dosage forms
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
US10617675B2 (en) 2015-08-06 2020-04-14 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US11247987B2 (en) 2017-10-06 2022-02-15 Forma Therapeutics, Inc. Inhibiting ubiquitin specific peptidase 30
US11667651B2 (en) 2017-12-22 2023-06-06 Hibercell, Inc. Aminopyridine derivatives as phosphatidylinositol phosphate kinase inhibitors
US11555029B2 (en) 2018-02-13 2023-01-17 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US10710986B2 (en) 2018-02-13 2020-07-14 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US10899735B2 (en) 2018-04-19 2021-01-26 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US10774071B2 (en) 2018-07-13 2020-09-15 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
US11535618B2 (en) 2018-10-05 2022-12-27 Forma Therapeutics, Inc. Fused pyrrolines which act as ubiquitin-specific protease 30 (USP30) inhibitors
US11814386B2 (en) 2018-10-05 2023-11-14 Forma Therapeutics, Inc. Fused pyrrolines which act as ubiquitin-specific protease 30 (USP30) inhibitors
US11236085B2 (en) 2018-10-24 2022-02-01 Gilead Sciences, Inc. PD-1/PD-L1 inhibitors
RU2723482C1 (en) * 2019-10-22 2020-06-11 Андрей Александрович Иващенко Pangenotypic ns5a protein inhibitor of hepatitis c virus, a pharmaceutical composition and methods for preparing and using
WO2021249364A1 (en) * 2020-06-12 2021-12-16 Wang, Jinping Novel analogs of pterostilbene amino acid bearing carbonates for treating a non-alcoholic fatty liver disease and nonalcoholic steatohepatitis
WO2023243601A1 (en) * 2022-06-13 2023-12-21 モジュラス株式会社 Azacycloalkyl carbonyl cyclic amine compound

Also Published As

Publication number Publication date
EP2475254A4 (en) 2013-05-22
EP2475254A1 (en) 2012-07-18

Similar Documents

Publication Publication Date Title
US8815928B2 (en) Hepatitis C virus inhibitors
US8927709B2 (en) Hepatitis C virus inhibitors
WO2011031934A1 (en) Hepatitis c virus inhibitors
US8759332B2 (en) Hepatitis C virus inhibitors
US8609648B2 (en) Hepatitis C virus inhibitors
US8221737B2 (en) Hepatitis C virus inhibitors
US9156818B2 (en) Hepatitis C virus inhibitors
US8242156B2 (en) Linked dibenzimidazole derivatives
US8703938B2 (en) Hepatitis C virus inhibitors
US8314135B2 (en) Linked dibenzimidazole antivirals
US8822700B2 (en) Hepatitis C virus inhibitors
US8507522B2 (en) Hepatitis C virus inhibitors
US8188132B2 (en) Linked dibenzimidazole derivatives
WO2010148006A1 (en) Hepatitis c virus inhibitors
EP2575819A1 (en) Hepatitis c virus inhibitors
WO2011081918A1 (en) Hepatitis c virus inhibitors
WO2010091413A1 (en) Linked dibenzimidazole derivatives
WO2010096462A1 (en) Linked diimidazole derivatives
KR20120107529A (en) Hepatitis c virus inhibitors
EP2555622A1 (en) Hepatitis c virus inhibitors
WO2010099527A1 (en) Hepatitis c virus inhibitors
WO2012021704A1 (en) Hepatitis c virus inhibitors
WO2013059278A2 (en) Hepatitis c virus inhibitors
WO2013052362A1 (en) Hepatitis c virus 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: 10816134

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2010816134

Country of ref document: EP