Methods for treating hepatitis C

Information

  • Patent Grant
  • 8013006
  • Patent Number
    8,013,006
  • Date Filed
    Thursday, July 14, 2005
    18 years ago
  • Date Issued
    Tuesday, September 6, 2011
    12 years ago
Abstract
In accordance with the present invention, compounds that inhibit viral replication, preferably Hepatitis C Virus (HCV) replication, have been identified, and methods for their use provided. In one aspect of the invention, compounds useful in the treatment or prevention of a viral infection are provided. In another aspect of the invention, compounds useful in the treatment or prevention of HCV infection are provided.
Description
FIELD OF THE INVENTION

The present invention relates to methods for treating Hepatitis C using Indole compounds that modify translational control of Hepatitis C virus.


BACKGROUND OF THE INVENTION

An estimated 170 million people worldwide are reported to be infected with hepatitis C virus (HCV), the causative agent of hepatitis C. Seventy to eighty percent of HCV infections lead to chronic liver infection, which in turn may result in severe liver disease, including liver fibrosis, cirrhosis, and hepatocellular carcinoma (115).


HCV constitutes the Hepacivirus genus of the family Flaviviridae (106), and contains a positive-stranded 9.6 kb RNA genome. The features of the HCV genome include a 5′-untranslated region (UTR) that encodes an internal ribosome entry site (IRES) that directs the translation of a single long open reading frame (ORF) encoding a polyprotein of 3,010 amino acids. The HCV ORF is followed by a 3′-UTR of variable length, depending on the HCV variant, that encodes the sequences required for the initiation of antigenomic strand synthesis (79).


The HCV IRES and 3′-UTR both encode regions of RNA structures that are required for genome translation and replication. The HCV polyprotein is posttranslationally processed into at least 10 mature viral proteins, including the structural proteins core (putative nucleocapsid), E1 and E2 and the nonstructural (NS) proteins NS2 to NS5B.


Three distinct elements have been shown to be involved in HCV IRES-mediated translation: (1) integrity of the global structure of HCV IRES, (2) the 3′-terminal region of the HCV genome; and (3) trans-acting cellular factors that interact with the HCV IRES element and assist in translation initiation (35).


The initiation of protein synthesis in eukaryotic cells predominantly follows the 5′ cap-dependent, first AUG rule (61). However, an increasing number of viral (6, 12, 28, 31a, 50, 95, 97, 98, 105, 128) and cellular mRNAs (18, 39, 45, 78, 91, 130) have been shown to use an IRES element to direct translation initiation. In 1992, an IRES element was reported in the 5′ UTR of the HCV RNA genome (129), indicating that synthesis of the viral protein is initiated in a cap-independent fashion.


A bicistronic expression system can be used to define and evaluate the function of IRES elements. This test system harbors two different reporter genes in which the 5′-proximal reporter gene is expressed by a cap dependent translation mechanism while the second reporter is expressed only if an upstream sequence inserted in the intergenic space contains an IRES sequence element. Using this system, a putative IRES in the HCV 5′ UTR was unambiguously demonstrated to function as an IRES involved in translational control of viral proteins (133). In vitro translation, RNA transfection, and mutagenesis studies provided further evidence that the HCV 5′ UTR contains an IRES element (23, 41, 42, 108, 129, 132, 133, 134). Both in vitro and cell-based studies demonstrated that the HCV IRES guides cellular translation initiation factors to an internal site of the viral RNA (56, 58, 120), thus functionally demonstrating the HCV IRES activity. Taken together, these results demonstrate that the HCV 5′-UTR contains an IRES element that plays an active and crucial role in the mechanism of internal initiation for HCV protein translation.


The IRES is one of the most conserved regions of the HCV genome, reflecting its essential nature for viral replication and protein synthesis (13, 118, 122). Although both 5′ and 3′ sequences of the IRES appear to play a role in the control of initiation of translation (42, 109, 110, 113, 136), the minimal sequence requirement for HCV IRES function has been mapped to a region between nucleotides 44-354 (40).


Biochemical probing and computer modeling indicate that the HCV IRES and its 5′ sequence is folded into a distinct structure that consists of four major domains and a pseudoknot (11, 42, 122). Domain I contains a small stem-loop structure that does not appear to be a functional part of the IRES element while domains II, III, and IV contain the HCV IRES activity (43, 111). The relationships between secondary and tertiary structures of the HCV IRES and their function have recently been established (5, 55, 56, 99, 124). Both domains II and III consist of multiple stems, loops, and bulges and are important for IRES activity (23, 40, 51, 52, 54, 56, 64, 74, 75, 93, 107, 108, 110, 124, 127, 131, 139). Domain II can induce conformational changes on the ribosome that have been implicated in the decoding process (124). Domain III has the highest degree of structural conservation among the different HCV strains. It comprises the core of the flavivirus IRES and has 6 subdomains (40). Various studies have shown that subdomain IIId forms complex secondary/tertiary structures and is critical for initiation activity (55, 56, 57, 124, 129). Domain IV has one stem-loop that spans the initiation codon and is specific for the HCV IRES (41, 122), but the precise role of domain IV in IRES activity remains controversial (41, 112).


The role of the HCV IRES is to position the translational machinery near an internal initiator codon in the viral mRNA. The translation initiation mechanism of the HCV IRES differs significantly from that of 5′-cap-dependent translation initiation (7, 21, 31, 35, 81, 96, 114, 123). Most cellular capped mRNAs utilize a number of initiation factors (eIFs) that are required for the translation initiation process. The initial steps of the process require proteins that interact with the 5′ cap structure and recruit the 40S ribosomal subunit to the cap-proximal region of mRNA. This complex then scans 3′ of the cap, until reaching an AUG codon at which translation will initiate (21, 114). However, in the case of HCV, the IRES functionally replaces the 5′ cap structure, allowing the 40S ribosomal subunit and eIF3 to bind directly to the RNA. Subdomain IIId of the HCV IRES harbors the binding site for the 40S ribosomal subunit and the only initiation factors required for translation initiation are eIF2, eIF3, and eIF4E (15, 58, 94, 100, 120, 124).


The polypyrimidine track-binding protein (PTB) and La autoantigen are noncanonical translation initiation factors that bind to and enhance HCV IRES activity (1, 2, 3, 4, 5, 30, 48, 49, 53). PTB, a 57-kDa protein involved in RNA splicing, is also necessary for efficient IRES-mediated translation initiation of picornavirus mRNA, and some cellular mRNAs (10, 11, 36, 53, 59, 89, 92). The La autoantigen, a 52 kDa double-stranded RNA unwinding protein, also increases the activity of poliovirus and cellular IRESs (38, 85, 86). Other cellular factors involved in HCV IRES-mediated translation initiation include proteasome α-subunit PSMA7 (62), ribosomal protein S5 (26), ribosomal protein S9 (24, 25, 100), and hnRNPL (33). However, the role of these RNA-binding proteins in HCV IRES-mediated initiation of translation is unclear. Recently, it was reported that the activity of interferon (IFN) α against HCV replication might target HCV IRES-mediated translation initiation by causing a reduction of La protein levels (117). Thus, an inhibitor that blocks interaction between the IRES and the noncanonical factors might efficiently inhibit HCV replication and lack cytotoxicity.


Currently, only interferon (IFN) α and the nucleoside analogue ribavirin, in combination, are marketed for the treatment of HCV infection. However, these two agents are immunomodulators and have limited efficacy, relatively high toxicity, and high cost (80, 83, 84, 138). Although the treatment outcome is variable among the six major HCV genotypes, only about one-half of all treated patients respond to therapy, suggesting that the virus encodes protein products that may directly or indirectly attenuate the antiviral action of IFN. IFNs are naturally produced in response to virus infection, and cellular exposure to IFN leads to the induced expression of a variety of IFN-stimulated genes (ISGs), many of which have an antiviral function. ISG action can limit virus replication at multiple points within the replicative cycle.


There remains a need for a more effective means of treating patients afflicted with HCV. Specifically, a need exists for novel antiviral drugs that have no cross-resistance with existing treatment modalities, and which demonstrate synergy with other anti-HCV agents. The applicants set out to identify drug candidates that inhibit HCV infection and were successful in identifying Indole compounds that are useful as anti-HCV agents. Without being limited to one theory, it is believed that the compounds of the present invention inhibit IRES-mediated initiation, elongation, and termination, i.e. translation.


The compounds of the present invention are also useful for inhibiting translation of other cap-independent viruses that contain an IRES element. Such viruses include those of the picornavirus genus, such as poliovirus, hepatitis A virus and rhinovirus; those of the coronavirus genus, such as SARS; those of the arbovirus genus; those of the flavivirus genus, such as yellow fever, dengue, and West Nile virus, herpesviruses, such as herpes simplex virus and Kaposi's sarcoma-associated herpesvirus, or any other virus with a similar mode of replication. Furthermore, compounds of the invention are also useful for inhibiting HIV, or any other virus with a similar mode of translation.


All documents referred to herein are incorporated by reference into the present application as though fully set forth herein.


SUMMARY OF THE INVENTION

In accordance with the present invention, compounds that can inhibit HCV replication have been identified. Also in accordance with the present invention, compounds that can inhibit HCV infection have been identified, and methods for their use provided.


In one aspect of the invention, compounds of Formula (I) are provided which are useful in the prevention and/or treatment of HCV infection. Without being limited to one theory, it is believed that the compounds of the present invention inhibit IRES-mediated initiation, elongation and termination, i.e., translation. The compounds of Formula (I) may also be useful for inhibiting and/or treating other viral infections where the virus contains an IRES element. Such viruses include those of the picornavirus genus, such as by way of non-limiting example poliovirus, hepatitis A virus and rhinovirus; those of the coronaviridae genus, such as by way of non-limiting example SARS; those of the arbovirus genus; those of the flavivirus genus, such as by way of non-limiting example yellow fever, dengue, and West Nile virus; herpesviruses, such as by way of non-limiting example herpes simplex virus and Kaposi's sarcoma-associated herpesvirus, or any other virus with a similar mode of replication. Furthermore, compounds of the invention are also useful for inhibiting HIV, or any other virus with a similar mode of translation.


In another aspect of the invention, methods are provided for the prevention and/or treatment of HCV infection.


In yet another aspect of the invention, pharmaceutical compositions comprising the compounds of the invention for the prevention and/or treatment of HCV infection are provided.


In one embodiment, the invention is directed to methods for inhibiting HCV IRES-mediated initiation and translation comprising administering an amount of one or more compound of the invention, effective for inhibiting IRES-mediated initiation and translation, to a subject in need thereof.


CERTAIN EMBODIMENTS
Embodiment 1

A pharmaceutical composition for the prevention or treatment of Hepatitis C viral (HCV) infection comprising a therapeutically effective amount of at least one compound having the following formula:




embedded image



wherein:

  • X is:
    • hydrogen;
    • a nitro group;
    • a cyano group;
    • a —CORa group, where Ra is:
      • a C1 to C6 alkyl,
      • a C6 to C8 aryl optionally substituted with an alkoxy or a halogen, or
      • a dialkyl-amino;
    • a —COORx group, where Rx is a C1 to C6 alkyl;
    • a formyl group;
    • a C6 to C8 aryl optionally substituted with an alkoxy; or
    • a 5 or 6-membered heteroaryl optionally substituted with:
      • a C1 to C6 alkyl,
      • a C6 to C8 aryl optionally substituted with an alkoxy or one or more halogens, or
      • a 5 to 6 membered heteroaryl;
  • Y is:
    • a hydrogen;
    • a haloalkyl;
    • a halogen;
    • an amino optionally substituted with one or more C1 to C6 alkyls;
    • a benzofuran;
    • a benzothiophene;
    • a dibenzofuran;
    • a dibenzothiophene;
    • a benzothiazole;
    • a naphthalene;
    • an indole, optionally substituted on the nitrogen with a C1 to C6 alkyl;




embedded image




    •  where Rb is a hydrogen or a C1 to C6 alkyl, and n is 0 or 1;







embedded image




embedded image




    •  where Rc is a hydrogen, a —CONHRx, where Rx is as defined above, or an —SO2Rx, where Rx is as defined above; or







embedded image




    •  where Rd is a C1 to C6 alkyl or a C6 to C8 aryl;

    • a —NHCORe group, where Re is:
      • a C1 to C6 alkyl;
      • a C6 to C8 aryl optionally substituted with:
        • a C1 to C6 alkyl,
        • an alkoxy,
        • a cyano group,
        • a nitro group, or
        • a halogen;

    • a —NHCOORx group, where Rx is as defined above;

    • a —CH2O—Rf group, where Rf is a C6 to C8 aryl;

    • a —NRgRh group, where Rg is a C1 to C6 alkyl or a hydrogen and Rh is a C6 to C8 aryl optionally substituted with an alkoxy;

    • a C1 to C6 alkyl;

    • a 5 or 6 membered heteroaryl, optionally substituted with:
      • a C1 to C6 alkyl, optionally substituted with a C6 to C8 aryl,
      • a C6 to C8 aryl, optionally substituted with —COORx, where Rx is as defined above, or
      • an amino group;

    • a 5 or 6 membered heterocycle optionally substituted with:
      • a —COORx group, where Rx is as defined above, or
      • a —NHCOORx group, where Rx is as defined above;

    • a C6 to C8 aryl, optionally substituted with one or more of the following:
      • an alkoxy, optionally substituted with:
        • an alkoxy,
        • a hydroxy,
        • one or more halogens,
        • a 5 or 6 membered heterocycle, optionally substituted with:
          • a C1 to C6 alkyl, or
          • a hydroxy,
        • an amino group optionally substituted with one or more C1 to C6 alkyls,
        • a —NRiSO2Rx group, where Rx is as defined above and Ri is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a —CORx group, where Rx is as defined above,
          • a haloalkyl, or
          • a haloalkoxy,
        • a —NRjCORk group, where Rk is:
          • a C1 to C6 alkyl,
          • a hydrogen, or
          • an amino optionally substituted with one or more C1 to C6 alkyls, and Rj is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a —CORx group, where Rx is as defined above,
          • a haloalkyl, or
          • a haloalkoxy,
        • a —N═N+═N group, or
        • a —CORl, where Rl is a 5 or 6 membered heterocycle optionally substituted with a hydroxy,
      • an amino optionally substituted with one or more C1 to C6 alkyls,
      • a nitro group,
      • a C1 to C6 alkyl group, optionally substituted with:
        • a —NHSO2Rx group, where Rx is as defined above, or
        • a —NRxSO2Rx group, where Rx is as defined above,
      • a haloalkoxy,
      • a halogen,
      • a hydroxy,
      • a —COORx group, where Rx is as defined above,
      • a —CORm group, where Rm is:
        • an amino optionally substituted with one or more C1 to C6 alkyls, where the C1 to C6 alkyls are optionally substituted with:
          • a hydroxy
          • a 5 or 6 membered heterocycle,
          • an amino optionally substituted with one or more C1 to C6 alkyls,
          • an alkoxy,
        • a 3 to 7 membered heterocycle, optionally substituted with a C1 to C6 alkyl, optionally substituted with a dialkyl-amino,
        • a —NHRn group, where Rn is:
          • a —CH2CONH2, or
          • a C6 to C8 aryl optionally substituted with:
          •  an alkyl,
          •  one or more halogens,
          •  a nitro group, or
          •  one or more alkoxys,
      • a —NRoCORp group, where Rp is:
        • a C1 to C6 alkyl optionally substituted with:
          • a halogen,
          • an alkoxy, or
          • a C6 to C8 aryl,
        • a 5 or 6 membered heterocycle,
        • a C6 to C8 aryl, optionally substituted with a halogen,
        • a 5 or 6 membered heteroaryl optionally substituted with one or more C1 to C6 alkyls,
        • a hydrogen,







embedded image






      • and where Ro is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a —CORx group, where Rx is as defined above,
        • a haloalkyl, or
        • a haloalkoxy,

      • a —NRqCONRqRr group, where Rq is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a haloalkyl,
        • a haloalkoxy, or
        • a —CORx group, where Rx is as defined above,

      • and where Rr is:
        • a C6 to C8 aryl optionally substituted with:









embedded image










          • a C1 to C6 alkyl,

          • a haloalkyl,

          • a —ORs group, where Rs is a C6 to C8 aryl, or

          • a —COORx group, where Rx is as defined above,



        • a C1 to C6 alkyl optionally substituted with one or more of the following:
          • a halogen,
          • an alkylene,
          • a C6 to C8 aryl, or
          • a —COORx group, where Rx is as defined above,

        • a —COORx group, where Rx is as defined above,



      • a —NRtCOORu group, where Ru is:
        • a C1 to C12 alkyl, optionally substituted with:
          • a C6 to C8 aryl optionally substituted with a C1 to C6 alkyl or an alkoxy,
          • an alkylene,
          • an alkoxy,
          • an alkyne,
          • a halogen, or
          • a 5 or 6 membered heterocycle,
        • a C6 to C8 aryl, optionally substituted with:
          • an alkoxy,
          • a halogen, or
          • a C1 to C6 alkyl, or
        • a 5 or 6 membered heterocycle,

      • and Rt is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a —CORx group, where Rx is as defined above,
        • a haloalkyl, or
        • a haloalkoxy,

      • a —NRvSO2Rw group, where Rv is:
        • a hydrogen,
        • a —CORx, where Rx is as defined above, or
        • a C1 to C6 alkyl, optionally substituted with:
          • a halogen,
          • a —CORx group, where Rx is as defined above,
          • a —OCORx group, where Rx is as defined above,
          • a hydroxyl,
          • a hydroxyl, or
          • an alkoxy,

      • and where Rw is:
        • a C1 to C6 alkyl optionally substituted with:
          • a halogen,
          • a haloalkyl,
          • a C6 to C8 aryl, or
          • a 5 or 6 membered heterocycle,
        • a C2 to C6 alkylene,
        • an alkyl- or dialkyl-amino optionally substituted with a halogen,
        • a 5 or 6 membered heterocycle, or
        • a 5 or 6 membered heteroaryl optionally substituted with:
          • a C1 to C6 alkyl,
          • a 5 or 6 membered heterocycle, or









embedded image






      •  optionally substituted with a C1 to C6 alkyl, where Ry is a C1 to C6 alkyl or hydrogen,









embedded image






      • where Rz is hydrogen or a C1 to C6 alkyl, optionally substituted with a C6 to C8 aryl,

      • a —SRx group, where Rx is as defined above,

      • a —SO2Raa group, where Raa is:
        • a C1 to C6 alkyl,
        • an amino group,
        • an alkyl- or dialkyl-amino group optionally substituted with a hydroxy or a —COORx group, where Rx is as defined above,
        • a 5 or 6 membered heteroaryl,

      • a C6 to C8 aryl, or

      • a —NHRbb group, where Rbb is:









embedded image








        • a —C(═S)NH2 group, or

        • a —PO(ORx)2, where Rx is as defined above;











embedded image




    •  Rcc group, where Rcc is:
      • a naphthalene,
      • a 5 or 6 membered heteroaryl,
      • a







embedded image






      • a C6 to C8 aryl, optionally substituted with one or more of the following:
        • an alkoxy,
        • an hydroxy,
        • a halogen,
        • a C1 to C6 alkyl, optionally substituted with a cyano group,
        • an amino optionally substituted with one or more C1 to C6 alkyls,
        • a —NHPORxRx, where Rx is as defined above,
        • a —NReeCONRffRff group, where Ree is a hydrogen or a C1 to C6 alkyl, optionally substituted with a halogen, and Rff is:
          • a hydrogen,
          • a haloalkyl,
          • a haloalkoxy,
          • a C1 to C6 alkyl, or
          • a —CORx, where Rx is as defined above,
        • a —NRggCORhh group, where Rhh is:
          • a hydrogen,
          • a C1 to C6 alkyl optionally substituted with:
          •  an alkoxy,
          •  a halogen, or
          •  an amino optionally substituted with one or more C1 to C6 alkyls,
          • an amino optionally substituted with one or more C1 to C6 alkyls, where the alkyls are optionally substituted with a halogen,
          • a 5 or 6 membered heterocycle,
          • a 5 or 6 membered heteroaryl,
        • and Rgg is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a haloalkyl,
          • a haloalkoxy, or
          • a —CORx group, where Rx is as defined above,
        • a haloalkyl,
        • 5 or 6 membered heterocycle groups,
        • an amino optionally substituted with one or more C1 to C6 alkyls,
        • a —NRiiSO2Rx group, where Rx is as defined above, and Rii is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a haloalkyl,
          • a haloalkoxy,
          • a —CORx group, where Rx is as defined above;





  • Z is:
    • a hydrogen;
    • a C1 to C6 alkyl optionally substituted with:
      • an alkoxy,
      • one or more halogens, or
      • a C6 to C8 aryl;
    • a C2 to C6 alkylene;
    • a C6 to C8 aryl optionally substituted with an alkoxy or one or more C1 to C6 alkyls;
    • a —COORx group, where Rx is as defined above; or





embedded image


  • R is a hydrogen, a halogen or an alkoxy;

  • R1 is:
    • a hydrogen;
    • a hydroxy;
    • a halogen;
    • a haloalkyl;
    • a nitro group;
    • a 5 or 6 membered heteroaryl;
    • a 5 or 6 membered heterocycle;
    • an alkoxy optionally substituted with:
      • one or more halogens,
      • a C6 to C8 aryl, or
      • a 5 or 6 membered heterocycle;
    • a C6 to C8 aryl optionally substituted with an alkoxy;
    • a —CORx group, where Rx is as defined above;
    • a C1 to C6 alkyl optionally substituted with a dialkyl-amino or a 5 or 6 membered heterocycle; or

  • R1 joins together with R2 to form:





embedded image


  • R2 is:
    • a nitro group;
    • a hydrogen;
    • a halogen;
    • a hydroxy group;
    • a C1 to C6 alkyl group, optionally substituted with one or more halogens;
    • an amino group;
    • an alkoxy group optionally substituted with:
      • one or more halogens,
      • an —OCORx group, where Rx is as defined above,
      • a dialkyl-amino optionally substituted with an alkoxy,
      • a 5 or 6 membered heterocycle group optionally substituted with a C1 to C6 alkyl,
      • a 5 or 6 membered heteroaryl group, or
      • a C6 to C8 aryl group;
    • a —COORx group, where Rx is as defined above;
    • a haloalkyl;
    • an amide group optionally substituted with:
      • a hydroxy group, or
      • a C6 to C8 aryl;
    • a 5 or 6 membered heteroaryl;
    • a —OCORx group, where Rx is as defined above;
    • a —NHCORjj group, where Rjj is:
      • an alkoxy, or
      • an amino optionally substituted with one or more C1 to C6 alkyls;
    • a —ORkk group, where Rkk is a 5 to 6 membered heteroaryl;
    • a —NHSO2Rx group, where Rx is as defined above; or

  • R2 joins together with R1 to form:





embedded image


  • R3 is:
    • a hydrogen; or
    • —CH2OCORx, and Rx is as defined above;


      or a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable excipient, and optionally at least one additional anti-HCV agents.



Embodiment 2

The pharmaceutical composition of Embodiment 1, wherein said optional at least one additional anti-HCV agent is selected from the group consisting of pegylated interferon, un-pegylated interferon, ribavirin or prodrugs or derivatives thereof, a glucosidase inhibitor, a protease inhibitor, a polymerase inhibitor, p7 inhibitors, an entry inhibitor, a fusion inhibitor, an anti-fibrotic, a caspase inhibitor, a drug which targets inosine monophosphate dehydrogenase inhibitors (IMPDH), synthetic thymosin alpha 1, therapeutic vaccines, immunomodulators, a glycosidase inhibitor, a helicase inhibitor, a Toll-like receptor agonist, and combinations thereof.


Embodiment 3

The pharmaceutical composition of Embodiment 1, wherein X is selected from the group consisting of —hydrogen; —a cyano group; and —a —CORa group, where Ra is: —a C1 to C6 alkyl, or —a dialkyl-amino.


Embodiment 4

The pharmaceutical composition of Embodiment 1, wherein Y is selected from the group consisting of




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


Embodiment 5

The pharmaceutical composition of Embodiment 1, wherein Y is selected from the group consisting of




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


Embodiment 6

The pharmaceutical composition of Embodiment 1, wherein R is a hydrogen.


Embodiment 7

The pharmaceutical composition of Embodiment 1, wherein R1 is selected from the group consisting of a hydrogen, a nitro group, or an alkoxy.


Embodiment 8

The pharmaceutical composition of Embodiment 1, wherein R2 is selected from the group consisting of a hydroxy, a hydrogen, a haloalkyl group, a nitro group, an amide, —COORx or an alkoxy.


Embodiment 9

The pharmaceutical composition of Embodiment 1, wherein R3 is a hydrogen.


Embodiment 10

The pharmaceutical composition of Embodiment 1, wherein said compound is selected from the compounds of Table A.


Embodiment 11

The pharmaceutical composition of Embodiment 1 wherein said compound is selected from the compounds of Table B or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient.


Embodiment 12

The pharmaceutical composition of Embodiment 17, wherein said composition further comprises at least one additional anti-HCV agent selected from the group consisting of pegylated interferon, un-pegylated interferon, ribavirin or prodrugs or derivatives thereof, a glucosidase inhibitor, a protease inhibitor, a polymerase inhibitor, p7 inhibitors, an entry inhibitor, a fusion inhibitor, an anti-fibrotic, a caspase inhibitor, a drug which targets inosine monophosphate dehydrogenase inhibitors (IMPDH), synthetic thymosin alpha 1, therapeutic vaccines, immunomodulators, a glycosidase inhibitor, a helicase inhibitor, a Toll-like receptor agonist, and combinations thereof.


Embodiment 13

A method for treating a subject for a Hepatitis C viral (HCV) infection comprising administering to said subject a pharmaceutical composition comprising an HCV inhibitory amount of at least one compound having the following formula:




embedded image



wherein:

  • X is:
    • hydrogen;
    • a nitro group;
    • a cyano group;
    • a —CORa group, where Ra is:
      • a C1 to C6 alkyl,
      • a C6 to C8 aryl optionally substituted with an alkoxy or a halogen, or
      • a dialkyl-amino;
    • a —COORx group, where Rx is a C1 to C6 alkyl;
    • a formyl group;
    • a C6 to C8 aryl optionally substituted with an alkoxy; or
    • a 5 or 6-membered heteroaryl optionally substituted with:
      • a C1 to C6 alkyl,
      • a C6 to C8 aryl optionally substituted with an alkoxy or one or more halogens, or
      • a 5 to 6 membered heteroaryl;
  • Y is:
    • a hydrogen;
    • a haloalkyl;
    • a halogen;
    • an amino optionally substituted with one or more C1 to C6 alkyls;
    • a benzofuran;
    • a benzothiophene;
    • a dibenzofuran;
    • a dibenzothiophene;
    • a benzothiazole;
    • a naphthalene;
    • an indole, optionally substituted on the nitrogen with a C1 to C6 alkyl;




embedded image




    •  where Rb is a hydrogen or a C1 to C6 alkyl, and n is 0 or 1;







embedded image




    •  where Rc is a hydrogen, a —CONHRx, where Rx is as defined above, or an —SO2Rx, where Rx is as defined above; or







embedded image




    •  where Rd is a C1 to C6 alkyl or a C6 to C8 aryl;

    • a —NHCORe group, where Re is:
      • a C1 to C6 alkyl;
      • a C6 to C8 aryl optionally substituted with:
        • a C1 to C6 alkyl,
        • an alkoxy,
        • a cyano group,
        • a nitro group, or
        • a halogen;

    • a —NHCOORx group, where Rx is as defined above;

    • a —CH2O—Rf group, where Rf is a C6 to C8 aryl;

    • a —NRgRh group, where Rg is a C1 to C6 alkyl or a hydrogen and Rh is a C6 to C8 aryl optionally substituted with an alkoxy;

    • a C1 to C6 alkyl;

    • a 5 or 6 membered heteroaryl, optionally substituted with:
      • a C1 to C6 alkyl, optionally substituted with a C6 to C8 aryl,
      • a C6 to C8 aryl, optionally substituted with —COORx, where Rx is as defined above, or
      • an amino group;

    • a 5 or 6 membered heterocycle optionally substituted with:
      • a —COORx group, where Rx is as defined above, or
      • a —NHCOORx group, where Rx is as defined above;

    • a C6 to C8 aryl, optionally substituted with one or more of the following:
      • an alkoxy, optionally substituted with:
        • an alkoxy,
        • a hydroxy,
        • one or more halogens,
        • a 5 or 6 membered heterocycle, optionally substituted with:
          • a C1 to C6 alkyl, or
          • a hydroxy,
        • an amino group optionally substituted with one or more C1 to C6 alkyls,
        • a —NRiSO2Rx group, where Rx is as defined above and Ri is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a —CORx group, where Rx is as defined above,
          • a haloalkyl, or
          • a haloalkoxy,
        • a —NRjCORk group, where Rk is:
          • a C1 to C6 alkyl,
          • a hydrogen, or
          • an amino optionally substituted with one or more C1 to C6 alkyls,
        • and Rj is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a —CORx group, where Rx is as defined above,
          • a haloalkyl, or
          • a haloalkoxy,
        • a —N═N+═N group, or
        • a —CORl, where Rl is a 5 or 6 membered heterocycle optionally substituted with a hydroxy,
      • an amino optionally substituted with one or more C1 to C6 alkyls,
      • a nitro group,
      • a C1 to C6 alkyl group, optionally substituted with:
        • a —NHSO2Rx group, where Rx is as defined above, or
        • a —NRxSO2Rx group, where Rx is as defined above,
      • a haloalkoxy,
      • a halogen,
      • a hydroxy,
      • a —COORx group, where Rx is as defined above,
      • a —CORm group, where Rm is:
        • an amino optionally substituted with one or more C1 to C6 alkyls, where the C1 to C6 alkyls are optionally substituted with:
          • a hydroxy
          • a 5 or 6 membered heterocycle,
          • an amino optionally substituted with one or more C1 to C6 alkyls,
          • an alkoxy,
        • a 3 to 7 membered heterocycle, optionally substituted with a C1 to C6 alkyl, optionally substituted with a dialkyl-amino,
        • a —NHRn group, where Rn is:
          • a —CH2CONH2, or
          • a C6 to C8 aryl optionally substituted with:
          •  an alkyl,
          •  one or more halogens,
          •  a nitro group, or
          •  one or more alkoxys,
      • a —NRoCORp group, where Rp is:
        • a C1 to C6 alkyl optionally substituted with:
          • a halogen,
          • an alkoxy, or
          • a C6 to C8 aryl,
        • a 5 or 6 membered heterocycle,
        • a C6 to C8 aryl, optionally substituted with a halogen,
        • a 5 or 6 membered heteroaryl optionally substituted with one or more C1 to C6 alkyls,
        • a hydrogen,







embedded image






      • and where Ro is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a —CORx group, where Rx is as defined above,
        • a haloalkyl, or
        • a haloalkoxy,

      • a —NRqCONRqRr group, where Rq is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a haloalkyl,
        • a haloalkoxy, or
        • a —CORx group, where Rx is as defined above,

      • and where Rr is:
        • a C6 to C8 aryl optionally substituted with:









embedded image










          • a C1 to C6 alkyl,

          • a haloalkyl,

          • a —ORs group, where Rs is a C6 to C8 aryl, or

          • a —COORx group, where Rx is as defined above,



        • a C1 to C6 alkyl optionally substituted with one or more of the following:
          • a halogen,
          • an alkylene,
          • a C6 to C8 aryl, or
          • a —COORx group, where Rx is as defined above,

        • a —COORx group, where Rx is as defined above,



      • a —NRtCOORu group, where Ru is:
        • a C1 to C12 alkyl, optionally substituted with:
          • a C6 to C8 aryl optionally substituted with a C1 to C6 alkyl or an alkoxy,
          • an alkylene,
          • an alkoxy,
          • an alkyne,
          • a halogen, or
          • a 5 or 6 membered heterocycle,
        • a C6 to C8 aryl, optionally substituted with:
          • an alkoxy,
          • a halogen, or
          • a C1 to C6 alkyl, or
        • a 5 or 6 membered heterocycle,

      • and Rt is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a —CORx group, where Rx is as defined above,
        • a haloalkyl, or
        • a haloalkoxy,

      • a —NRvSO2Rw group, where Rv is:
        • a hydrogen,
        • a —CORx, where Rx is as defined above, or
        • a C1 to C6 alkyl, optionally substituted with:
          • a halogen,
          • a —CORx group, where Rx is as defined above,
          • a —OCORx group, where Rx is as defined above,
          • a hydroxyl,
          • a hydroxyl, or
          • an alkoxy,

      • and where Rw is:
        • a C1 to C6 alkyl optionally substituted with:
          • a halogen,
          • a haloalkyl,
          • a C6 to C8 aryl, or
          • a 5 or 6 membered heterocycle,
        • a C2 to C6 alkylene,
        • an alkyl- or dialkyl-amino optionally substituted with a halogen,
        • a 5 or 6 membered heterocycle, or
        • a 5 or 6 membered heteroaryl optionally substituted with:
          • a C1 to C6 alkyl,
          • a 5 or 6 membered heterocycle, or









embedded image






      •  optionally substituted with a C1 to C6 alkyl, where Ry is a C1 to C6 alkyl or hydrogen,









embedded image






      • where Rz is hydrogen or a C1 to C6 alkyl, optionally substituted with a C6 to C8 aryl,

      • a —SRx group, where Rx is as defined above,

      • a —SO2Raa group, where Raa is:
        • a C1 to C6 alkyl,
        • an amino group,
        • an alkyl- or dialkyl-amino group optionally substituted with a hydroxy or a —COORx group, where Rx is as defined above,
        • a 5 or 6 membered heteroaryl,

      • a C6 to C8 aryl, or

      • a —NHRbb group, where Rbb is:









embedded image








        • a —C(═S)NH2 group, or

        • a —PO(ORx)2, where Rx is as defined above;











embedded image




    •  Rcc group, where Rcc is:
      • a naphthalene,
      • a 5 or 6 membered heteroaryl,







embedded image






      • a C6 to C8 aryl, optionally substituted with one or more of the following:
        • an alkoxy,
        • an hydroxy,
        • a halogen,
        • a C1 to C6 alkyl, optionally substituted with a cyano group,
        • an amino optionally substituted with one or more C1 to C6 alkyls,
        • a —NHPORxRx, where Rx is as defined above,
        • a —NReeCONRffRff group, where Ree is a hydrogen or a C1 to C6 alkyl, optionally substituted with a halogen, and Rff is:
          • a hydrogen,
          • a haloalkyl,
          • a haloalkoxy,
          • a C1 to C6 alkyl, or
          • a —CORx, where Rx is as defined above,
        • a —NRggCORhh group, where Rhh is:
          • a hydrogen,
          • a C1 to C6 alkyl optionally substituted with:
          •  an alkoxy,
          •  a halogen, or
          •  an amino optionally substituted with one or more C1 to C6 alkyls,
          • an amino optionally substituted with one or more C1 to C6 alkyls, where the alkyls are optionally substituted with a halogen,
          • a 5 or 6 membered heterocycle,
          • a 5 or 6 membered heteroaryl,
        • and Rgg is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a haloalkyl,
          • a haloalkoxy, or
          • a —CORx group, where Rx is as defined above,
        • a haloalkyl,
        • 5 or 6 membered heterocycle groups,
        • an amino optionally substituted with one or more C1 to C6 alkyls,
        • a —NRiiSO2Rx group, where Rx is as defined above, and Rii is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a haloalkyl,
          • a haloalkoxy,
          • a —CORx group, where Rx is as defined above;





  • Z is:
    • a hydrogen;
    • a C1 to C6 alkyl optionally substituted with:
      • an alkoxy,
      • one or more halogens, or
      • a C6 to C8 aryl;
    • a C2 to C6 alkylene;
    • a C6 to C8 aryl optionally substituted with an alkoxy or one or more C1 to C6 alkyls;
    • a —COORx group, where Rx is as defined above; or





embedded image


  • R is a hydrogen, a halogen or an alkoxy;

  • R1 is:
    • a hydrogen;
    • a hydroxy;
    • a halogen;
    • a haloalkyl;
    • a nitro group;
    • a 5 or 6 membered heteroaryl;
    • a 5 or 6 membered heterocycle;
    • an alkoxy optionally substituted with:
      • one or more halogens,
      • a C6 to C8 aryl, or
      • a 5 or 6 membered heterocycle;
    • a C6 to C8 aryl optionally substituted with an alkoxy;
    • a —CORx group, where Rx is as defined above;
    • a C1 to C6 alkyl optionally substituted with a dialkyl-amino or a 5 or 6 membered heterocycle; or

  • R1 joins together with R2 to form:





embedded image


  • R2 is:
    • a nitro group;
    • a hydrogen;
    • a halogen;
    • a hydroxy group;
    • a C1 to C6 alkyl group, optionally substituted with one or more halogens;
    • an amino group;
    • an alkoxy group optionally substituted with:
      • one or more halogens,
      • an —OCORx group, where Rx is as defined above,
      • a dialkyl-amino optionally substituted with an alkoxy,
      • a 5 or 6 membered heterocycle group optionally substituted with a C1 to C6 alkyl,
      • a 5 or 6 membered heteroaryl group, or
      • a C6 to C8 aryl group;
    • a —COORx group, where Rx is as defined above;
    • a haloalkyl;
    • an amide group optionally substituted with:
      • a hydroxy group, or
      • a C6 to C8 aryl;
    • a 5 or 6 membered heteroaryl;
    • a —OCORx group, where Rx is as defined above;
    • a —NHCORjj group, where Rjj is:
      • an alkoxy, or
      • an amino optionally substituted with one or more C1 to C6 alkyls;
    • a —ORkk group, where Rkk is a 5 to 6 membered heteroaryl;
    • a —NHSO2Rx group, where Rx is as defined above; or

  • R2 joins together with R1 to form:





embedded image


  • R3 is:
    • a hydrogen; or
    • —CH2OCORx, and Rx is as defined above;


      or a pharmaceutically acceptable salt thereof and a pharamceutically acceptable excipient.



Embodiment 14

The method of Embodiment 13, wherein said pharmaceutical composition further comprises at least one additional anti-HCV agent.


Embodiment 15

The method of Embodiment 14, wherein said at least one additional anti-HCV agent is selected from the group consisting of pegylated interferon, un-pegylated interferon, ribavirin or prodrugs or derivatives thereof, a glucosidase inhibitor, a protease inhibitor, a polymerase inhibitor, p7 inhibitors, an entry inhibitor, a fusion inhibitor, an anti-fibrotic, a caspase inhibitor, a drug which targets inosine monophosphate dehydrogenase inhibitors (IMPDH), synthetic thymosin alpha 1, therapeutic vaccines, immunomodulators, a glycosidase inhibitor, a helicase inhibitor, a Toll-like receptor agonist, and combinations thereof.


Embodiment 16

The method of Embodiment 13, wherein X is selected from the group consisting of —hydrogen; —a cyano group; and —a —CORa group, where Ra is: -a C1 to C6 alkyl, or —a dialkyl-amino.


Embodiment 17

The method of Embodiment 13, wherein Y is selected from the group consisting of




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


Embodiment 18

The method of Embodiment 13, wherein Y is selected from the group consisting of




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


Embodiment 19

The method of Embodiment 13, wherein R is a hydrogen.


Embodiment 20

The method of Embodiment 13, wherein R1 is selected from the group consisting of a hydrogen; —a halogen; —a nitro group; —a 5 or 6 membered heterocycle; —an alkoxy optionally substituted with: —a C6 to C8 aryl; -a C6 to C8 aryl optionally substituted with an alkoxy


Embodiment 21

The method of Embodiment 13, wherein R2 is selected from the group consisting of —a nitro group; —a hydrogen; —a halogen; —a hydroxy group; —a C1 to C6 alkyl group, optionally substituted with one or more halogens; —an alkoxy group optionally substituted with: —one or more halogens, —an —OCORx group, where Rx is as defined above, —a dialkyl-amino optionally substituted with an alkoxy, —a 5 or 6 membered heterocycle group optionally substituted with a C1 to C6 alkyl, or —a 5 or 6 membered heteroaryl group; —an amide group; and —a —NHSO2Rx group, where Rx is as defined above


Embodiment 22

The method of Embodiment 13, wherein R3 is a hydrogen.


Embodiment 23

The method of Embodiment 13, wherein said compound is selected from the compounds of Table A.


Embodiment 24

The method of Embodiment 13, wherein said compound is selected from the compounds of Table B.


Embodiment 25

A method for treating or preventing infection by a virus in a subject, wherein said virus comprises a internal ribosome entry site (IRES), comprising administering to said subject a pharmaceutical composition comprising a viral inhibitory amount of one or more compound having the following formula:




embedded image



wherein:

  • X is:
    • hydrogen;
    • a nitro group;
    • a cyano group;
    • a —CORa group, where Ra is:
      • a C1 to C6 alkyl,
      • a C6 to C8 aryl optionally substituted with an alkoxy or a halogen, or
      • a dialkyl-amino;
    • a —COORx group, where Rx is a C1 to C6 alkyl;
    • a formyl group;
    • a C6 to C8 aryl optionally substituted with an alkoxy; or
    • a 5 or 6-membered heteroaryl optionally substituted with:
      • a C1 to C6 alkyl,
      • a C6 to C8 aryl optionally substituted with an alkoxy or one or more halogens, or
      • a 5 to 6 membered heteroaryl;
  • Y is:
    • a hydrogen;
    • a haloalkyl;
    • a halogen;
    • an amino optionally substituted with one or more C1 to C6 alkyls;
    • a benzofuran;
    • a benzothiophene;
    • a dibenzofuran;
    • a dibenzothiophene;
    • a benzothiazole;
    • a naphthalene;
    • an indole, optionally substituted on the nitrogen with a C1 to C6 alkyl;




embedded image




    •  where Rb is a hydrogen or a C1 to C6 alkyl, and n is 0 or 1;







embedded image




    •  where Rc is a hydrogen, a —CONHRx, where Rx is as defined above, or an —SO2Rx, where Rx is as defined above; or







embedded image




    •  where Rd is a C1 to C6 alkyl or a C6 to C8 aryl;

    • a —NHCORe group, where Re is:
      • a C1 to C6 alkyl;
      • a C6 to C8 aryl optionally substituted with:
        • a C1 to C6 alkyl,
        • an alkoxy,
        • a cyano group,
        • a nitro group, or
        • a halogen;

    • a —NHCOORx group, where Rx is as defined above;

    • a —CH2O—Rf group, where Rf is a C6 to C8 aryl;

    • a —NRgRh group, where Rg is a C1 to C6 alkyl or a hydrogen and Rh is a C6 to C8 aryl optionally substituted with an alkoxy;

    • a C1 to C6 alkyl;

    • a 5 or 6 membered heteroaryl, optionally substituted with:
      • a C1 to C6 alkyl, optionally substituted with a C6 to C8 aryl,
      • a C6 to C8 aryl, optionally substituted with —COORx, where Rx is as defined above, or
      • an amino group;

    • a 5 or 6 membered heterocycle optionally substituted with:
      • a —COORx group, where Rx is as defined above, or
      • a —NHCOORx group, where Rx is as defined above;

    • a C6 to C8 aryl, optionally substituted with one or more of the following:
      • an alkoxy, optionally substituted with:
        • an alkoxy,
        • a hydroxy,
        • one or more halogens,
        • a 5 or 6 membered heterocycle, optionally substituted with:
          • a C1 to C6 alkyl, or
          • a hydroxy,
        • an amino group optionally substituted with one or more C1 to C6 alkyls,
        • a —NRiSO2Rx group, where Rx is as defined above and R1 is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a —CORx group, where Rx is as defined above,
          • a haloalkyl, or
          • a haloalkoxy,
        • a —NRjCORk group, where Rk is:
          • a C1 to C6 alkyl,
          • a hydrogen, or
          • an amino optionally substituted with one or more C1 to C6 alkyls,
        • and Rj is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a —CORx group, where Rx is as defined above,
          • a haloalkyl, or
          • a haloalkoxy,
        • a —N═N+═N group, or
        • a —CORl, where Rl is a 5 or 6 membered heterocycle optionally substituted with a hydroxy,
      • an amino optionally substituted with one or more C1 to C6 alkyls,
      • a nitro group,
      • a C1 to C6 alkyl group, optionally substituted with:
        • a —NHSO2Rx group, where Rx is as defined above, or
        • a —NRxSO2Rx group, where Rx is as defined above,
      • a haloalkoxy,
      • a halogen,
      • a hydroxy,
      • a —COORx group, where Rx is as defined above,
      • a —CORm group, where Rm is:
        • an amino optionally substituted with one or more C1 to C6 alkyls, where the C1 to C6 alkyls are optionally substituted with:
          • a hydroxy
          • a 5 or 6 membered heterocycle,
          • an amino optionally substituted with one or more C1 to C6 alkyls,
          • an alkoxy,
        • a 3 to 7 membered heterocycle, optionally substituted with a C1 to C6 alkyl, optionally substituted with a dialkyl-amino,
        • a —NHRn group, where Rn is:
          • a —CH2CONH2, or
          • a C6 to C8 aryl optionally substituted with:
          •  an alkyl,
          •  one or more halogens,
          •  a nitro group, or
          •  one or more alkoxys,
      • a —NRoCORp group, where Rp is:
        • a C1 to C6 alkyl optionally substituted with:
          • a halogen,
          • an alkoxy, or
          • a C6 to C8 aryl,
        • a 5 or 6 membered heterocycle,
        • a C6 to C8 aryl, optionally substituted with a halogen,
        • a 5 or 6 membered heteroaryl optionally substituted with one or more C1 to C6 alkyls,
        • a hydrogen,







embedded image






      • and where Ro is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a —CORx group, where Rx is as defined above,
        • a haloalkyl, or
        • a haloalkoxy,

      • a —NRqCONRqRr group, where Rq is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a haloalkyl,
        • a haloalkoxy, or
        • a —CORx group, where Rx is as defined above,

      • and where Rr is:
        • a C6 to C8 aryl optionally substituted with:









embedded image










          • a C1 to C6 alkyl,

          • a haloalkyl,

          • a —ORs group, where Rs is a C6 to C8 aryl, or

          • a —COORx group, where Rx is as defined above,



        • a C1 to C6 alkyl optionally substituted with one or more of the following:
          • a halogen,
          • an alkylene,
          • a C6 to C8 aryl, or
          • a —COORx group, where Rx is as defined above,

        • a —COORx group, where Rx is as defined above,



      • a —NRtCOORu group, where Ru is:
        • a C1 to C12 alkyl, optionally substituted with:
          • a C6 to C8 aryl optionally substituted with a C1 to C6 alkyl or an alkoxy,
          • an alkylene,
          • an alkoxy,
          • an alkyne,
          • a halogen, or
          • a 5 or 6 membered heterocycle,
        • a C6 to C8 aryl, optionally substituted with:
          • an alkoxy,
          • a halogen, or
          • a C1 to C6 alkyl, or
        • a 5 or 6 membered heterocycle,

      • and Rt is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a —CORx group, where Rx is as defined above,
        • a haloalkyl, or
        • a haloalkoxy,

      • a —NRvSO2Rw group, where Rv is:
        • a hydrogen,
        • a —CORx, where Rx is as defined above, or
        • a C1 to C6 alkyl, optionally substituted with:
          • a halogen,
          • a —CORx group, where Rx is as defined above,
          • a —OCORx group, where Rx is as defined above,
          • a hydroxyl,
          • a hydroxyl, or
          • an alkoxy,

      • and where Rw is:
        • a C1 to C6 alkyl optionally substituted with:
          • a halogen,
          • a haloalkyl,
          • a C6 to C8 aryl, or
          • a 5 or 6 membered heterocycle,
        • a C2 to C6 alkylene,
        • an alkyl- or dialkyl-amino optionally substituted with a halogen,
        • a 5 or 6 membered heterocycle, or
        • a 5 or 6 membered heteroaryl optionally substituted with:
          • a C1 to C6 alkyl,
          • a 5 or 6 membered heterocycle, or









embedded image






      •  optionally substituted with a C1 to C6 alkyl, where Ry is a C1 to C6 alkyl or hydrogen,









embedded image






      • where Rz is hydrogen or a C1 to C6 alkyl, optionally substituted with a C6 to C8 aryl,

      • a —SRx group, where Rx is as defined above,

      • a —SO2Raa group, where Raa is:
        • a C1 to C6 alkyl,
        • an amino group,
        • an alkyl- or dialkyl-amino group optionally substituted with a hydroxy or a —COORx group, where Rx is as defined above,
        • a 5 or 6 membered heteroaryl,

      • a C6 to C8 aryl, or

      • a —NHRbb group, where Rbb is:









embedded image








        • a —C(═S)NH2 group, or

        • a —PO(ORx)2, where Rx is as defined above;











embedded image




    •  Rcc group, where Rcc is:
      • a naphthalene,
      • a 5 or 6 membered heteroaryl,







embedded image






      • a C6 to C8 aryl, optionally substituted with one or more of the following:
        • an alkoxy,
        • an hydroxy,
        • a halogen,
        • a C1 to C6 alkyl, optionally substituted with a cyano group,
        • an amino optionally substituted with one or more C1 to C6 alkyls,
        • a —NHPORxRx, where Rx is as defined above,
        • a —NReeCONRffRff group, where Ree is a hydrogen or a C1 to C6 alkyl, optionally substituted with a halogen, and Rff is:
          • a hydrogen,
          • a haloalkyl,
          • a haloalkoxy,
          • a C1 to C6 alkyl, or
          • a —CORx, where Rx is as defined above,
        • a —NRggCORhh group, where Rhh is:
          • a hydrogen,
          • a C1 to C6 alkyl optionally substituted with:
          •  an alkoxy,
          •  a halogen, or
          •  an amino optionally substituted with one or more C1 to C6 alkyls,
          • an amino optionally substituted with one or more C1 to C6 alkyls, where the alkyls are optionally substituted with a halogen,
          • a 5 or 6 membered heterocycle,
          • a 5 or 6 membered heteroaryl,
        • and Rgg is:
          • a hydrogen,
          • a C1 to C 6 alkyl,
          • a haloalkyl,
          • a haloalkoxy, or
          • a —CORx group, where Rx is as defined above,
        • a haloalkyl,
        • 5 or 6 membered heterocycle groups,
        • an amino optionally substituted with one or more C1 to C6 alkyls,
        • a —NRiiSO2Rx group, where Rx is as defined above, and Rii is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a haloalkyl,
          • a haloalkoxy,
          • a —CORx group, where Rx is as defined above;





  • Z is:
    • a hydrogen;
    • a C1 to C6 alkyl optionally substituted with:
      • an alkoxy,
      • one or more halogens, or
      • a C6 to C8 aryl;
    • a C2 to C6 alkylene;
    • a C6 to C8 aryl optionally substituted with an alkoxy or one or more C1 to C6 alkyls;
    • a —COORx group, where Rx is as defined above; or





embedded image


  • R is a hydrogen, a halogen or an alkoxy;

  • R1 is:
    • a hydrogen;
    • a hydroxy;
    • a halogen;
    • a haloalkyl;
    • a nitro group;
    • a 5 or 6 membered heteroaryl;
    • a 5 or 6 membered heterocycle;
    • an alkoxy optionally substituted with:
      • one or more halogens,
      • a C6 to C8 aryl, or
      • a 5 or 6 membered heterocycle;
    • a C6 to C8 aryl optionally substituted with an alkoxy;
    • a —CORx group, where Rx is as defined above;
    • a C1 to C6 alkyl optionally substituted with a dialkyl-amino or a 5 or 6 membered heterocycle; or

  • R1 joins together with R2 to form:





embedded image


  • R2 is:
    • a nitro group;
    • a hydrogen;
    • a halogen;
    • a hydroxy group;
    • a C1 to C6 alkyl group, optionally substituted with one or more halogens;
    • an amino group;
    • an alkoxy group optionally substituted with:
      • one or more halogens,
      • an —OCORx group, where Rx is as defined above,
      • a dialkyl-amino optionally substituted with an alkoxy,
      • a 5 or 6 membered heterocycle group optionally substituted with a C1 to C6 alkyl,
      • a 5 or 6 membered heteroaryl group, or
      • a C6 to C8 aryl group;
    • a —COORx group, where Rx is as defined above;
    • a haloalkyl;
    • an amide group optionally substituted with:
      • a hydroxy group, or
      • a C6 to C8 aryl;
    • a 5 or 6 membered heteroaryl;
    • a —OCORx group, where Rx is as defined above;
    • a —NHCORjj group, where Rjj is:
      • an alkoxy, or
      • an amino optionally substituted with one or more C1 to C6 alkyls;
    • a —ORkk group, where Rkk is a 5 to 6 membered heteroaryl;
    • a —NHSO2Rx group, where Rx is as defined above; or

  • R2 joins together with R1 to form:





embedded image


  • R3 is:
    • a hydrogen; or
    • —CH2OCORx, and Rx is as defined above;


      and/or a pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable excipient.



Embodiment 33

The method of Embodiment 32 wherein said pharmaceutical composition further comprises at least one additional anti-viral agent.


Embodiment 34

The method of Embodiment 33, wherein said at least one additional anti-viral agent is elected from the group consisting of pegylated interferon, un-pegylated interferon, ribavirin or prodrugs or derivatives thereof, a glucosidase inhibitor, a protease inhibitor, a polymerase inhibitor, p7 inhibitors, an entry inhibitor, a fusion inhibitor, an anti-fibrotic, a caspase inhibitor, a drug which targets inosine monophosphate dehydrogenase inhibitors (IMPDH), synthetic thymosin alpha 1, therapeutic vaccines, immunomodulators, a glycosidase inhibitor, a helicase inhibitor, and a Toll-like receptor agonist.


Embodiment 26

A compound selected from the group consisting of the following:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


Embodiment 27

A compound selected from the group consisting of the following:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


Embodiment 27

A compound selected from the group consisting of the following:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


Embodiment 29

A pharmaceutical composition for affecting viral IRES activity in a subject infected with a virus, comprising one or more compound having the following formula:




embedded image



wherein:

  • X is:
    • hydrogen;
    • a nitro group;
    • a cyano group;
    • a —CORa group, where Ra is:
      • a C1 to C6 alkyl,
      • a C6 to C8 aryl optionally substituted with an alkoxy or a halogen, or
      • a dialkyl-amino;
    • a —COORx group, where Rx is a C1 to C6 alkyl;
    • a formyl group;
    • a C6 to C8 aryl optionally substituted with an alkoxy; or
    • a 5 or 6-membered heteroaryl optionally substituted with:
      • a C1 to C6 alkyl,
      • a C6 to C8 aryl optionally substituted with an alkoxy or one or more halogens, or
      • a 5 to 6 membered heteroaryl;
  • Y is:
    • a hydrogen;
    • a haloalkyl;
    • a halogen;
    • an amino optionally substituted with one or more C1 to C6 alkyls;
    • a benzofuran;
    • a benzothiophene;
    • a dibenzofuran;
    • a dibenzothiophene;
    • a benzothiazole;
    • a naphthalene;
    • an indole, optionally substituted on the nitrogen with a C1 to C6 alkyl;




embedded image




    •  where Rb is a hydrogen or a C1 to C6 alkyl, and n is 0 or 1;







embedded image




    •  where Rc is a hydrogen, a —CONHRx, where Rx is as defined above, or an —SO2Rx, where Rx is as defined above; or







embedded image




    •  where Rd is a C1 to C6 alkyl or a C6 to C8 aryl;

    • a —NHCORe group, where Re is:
      • a C1 to C6 alkyl;
      • a C6 to C8 aryl optionally substituted with:
        • a C1 to C6 alkyl,
        • an alkoxy,
        • a cyano group,
        • a nitro group, or
        • a halogen;

    • a —NHCOORx group, where Rx is as defined above;

    • a —CH2O—Rf group, where Rf is a C6 to C8 aryl;

    • a —NRgRh group, where Rg is a C1 to C6 alkyl or a hydrogen and Rh is a C6 to C8 aryl optionally substituted with an alkoxy;

    • a C1 to C6 alkyl;

    • a 5 or 6 membered heteroaryl, optionally substituted with:
      • a C1 to C6 alkyl, optionally substituted with a C6 to C8 aryl,
      • a C6 to C8 aryl, optionally substituted with —COORx, where Rx is as defined above, or
      • an amino group;

    • a 5 or 6 membered heterocycle optionally substituted with:
      • a —COORx group, where Rx is as defined above, or
      • a —NHCOORx group, where Rx is as defined above;

    • a C6 to C8 aryl, optionally substituted with one or more of the following:
      • an alkoxy, optionally substituted with:
        • an alkoxy,
        • a hydroxy,
        • one or more halogens,
        • a 5 or 6 membered heterocycle, optionally substituted with:
          • a C1 to C6 alkyl, or
          • a hydroxy,
        • an amino group optionally substituted with one or more C1 to C6 alkyls,
        • a —NRiSO2Rx group, where Rx is as defined above and Ri is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a —CORx group, where Rx is as defined above,
          • a haloalkyl, or
          • a haloalkoxy,
        • a —NRjCORk group, where Rk is:
          • a C1 to C6 alkyl,
          • a hydrogen, or
          • an amino optionally substituted with one or more C1 to C6 alkyls, and Rj is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a —CORx group, where Rx is as defined above,
          • a haloalkyl, or
          • a haloalkoxy,
        • a —N═N+═N group, or
        • a —CORl, where Rl is a 5 or 6 membered heterocycle optionally substituted with a hydroxy,
      • an amino optionally substituted with one or more C1 to C6 alkyls,
      • a nitro group,
      • a C1 to C6 alkyl group, optionally substituted with:
        • a —NHSO2Rx group, where Rx is as defined above, or
        • a —NRxSO2Rx group, where Rx is as defined above,
      • a haloalkoxy,
      • a halogen,
      • a hydroxy,
      • a —COORx group, where Rx is as defined above,
      • a —CORm group, where Rm is:
        • an amino optionally substituted with one or more C1 to C6 alkyls, where the C1 to C6 alkyls are optionally substituted with:
          • a hydroxy
          • a 5 or 6 membered heterocycle,
          • an amino optionally substituted with one or more C1 to C6 alkyls,
          • an alkoxy,
        • a 3 to 7 membered heterocycle, optionally substituted with a C1 to C6 alkyl, optionally substituted with a dialkyl-amino,
        • a —NHRn group, where Rn is:
          • a —CH2CONH2, or
          • a C6 to C8 aryl optionally substituted with:
          •  an alkyl,
          •  one or more halogens,
          •  a nitro group, or
          •  one or more alkoxys,
      • a —NRoCORp group, where Rp is:
        • a C1 to C6 alkyl optionally substituted with:
          • a halogen,
          • an alkoxy, or
          • a C6 to C8 aryl,
        • a 5 or 6 membered heterocycle,
        • a C6 to C8 aryl, optionally substituted with a halogen,
        • a 5 or 6 membered heteroaryl optionally substituted with one or more C, to C6 alkyls,
        • a hydrogen,







embedded image






      • and where Ro is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a —CORx group, where Rx is as defined above,
        • a haloalkyl, or
        • a haloalkoxy,

      • a —NRqCONRqRr group, where Rq is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a haloalkyl,
        • a haloalkoxy, or
        • a —CORx group, where Rx is as defined above,

      • and where Rr is:
        • a C6 to C8 aryl optionally substituted with:









embedded image










          • a C1 to C6 alkyl,

          • a haloalkyl,

          • a —ORs group, where Rs is a C6 to C8 aryl, or

          • a —COORx group, where Rx is as defined above,



        • a C1 to C6 alkyl optionally substituted with one or more of the following:
          • a halogen,
          • an alkylene,
          • a C6 to C8 aryl, or
          • a —COORx group, where Rx is as defined above,

        • a —COORx group, where Rx is as defined above,



      • a —NRtCOORu group, where Ru is:
        • a C1 to C12 alkyl, optionally substituted with:
          • a C6 to C8 aryl optionally substituted with a C1 to C6 alkyl or an alkoxy,
          • an alkylene,
          • an alkoxy,
          • an alkyne,
          • a halogen, or
          • a 5 or 6 membered heterocycle,
        • a C6 to C8 aryl, optionally substituted with:
          • an alkoxy,
          • a halogen, or
          • a C1 to C6 alkyl, or
        • a 5 or 6 membered heterocycle,

      • and Rt is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a —CORx group, where Rx is as defined above,
        • a haloalkyl, or
        • a haloalkoxy,

      • a —NRvSO2Rw group, where Rv is:
        • a hydrogen,
        • a —CORx, where Rx is as defined above, or
        • a C1 to C6 alkyl, optionally substituted with:
          • a halogen,
          • a —CORx group, where Rx is as defined above,
          • a —OCORx group, where Rx is as defined above,
          • a hydroxyl,
          • a hydroxyl, or
          • an alkoxy,

      • and where Rw is:
        • a C1 to C6 alkyl optionally substituted with:
          • a halogen,
          • a haloalkyl,
          • a C6 to C8 aryl, or
          • a 5 or 6 membered heterocycle,
        • a C2 to C6 alkylene,
        • an alkyl- or dialkyl-amino optionally substituted with a halogen,
        • a 5 or 6 membered heterocycle, or
        • a 5 or 6 membered heteroaryl optionally substituted with:
          • a C1 to C6 alkyl,
          • a 5 or 6 membered heterocycle, or









embedded image




embedded image






      •  optionally substituted with a C1 to C6 alkyl, where Ry is a C1 to C6 alkyl or hydrogen,









embedded image






      • where Rz is hydrogen or a C1 to C6 alkyl, optionally substituted with a C6 to C8 aryl,

      • a —SRx group, where Rx is as defined above,

      • a —SO2Raa group, where Raa is:
        • a C1 to C6 alkyl,
        • an amino group,
        • an alkyl- or dialkyl-amino group optionally substituted with a hydroxy or a —COORx group, where Rx is as defined above,
        • a 5 or 6 membered heteroaryl,

      • a C6 to C8 aryl, or

      • a —NHRbb group, where Rbb is:









embedded image








        • a —C(═S)NH2 group, or

        • a —PO(ORx)2, where Rx is as defined above;











embedded image




    •  Rcc group, where Rcc is:
      • a naphthalene,
      • a 5 or 6 membered heteroaryl,







embedded image






      • a C6 to C8 aryl, optionally substituted with one or more of the following:
        • an alkoxy,
        • an hydroxy,
        • a halogen,
        • a C1 to C6 alkyl, optionally substituted with a cyano group,
        • an amino optionally substituted with one or more C1 to C6 alkyls,
        • a —NHPORxRx, where Rx is as defined above,
        • a —NReeCONRffRff group, where Ree is a hydrogen or a C1 to C6 alkyl, optionally substituted with a halogen, and Rff is:
          • a hydrogen,
          • a haloalkyl,
          • a haloalkoxy,
          • a C1 to C6 alkyl, or
          • a —CORx, where Rx is as defined above,
        • a —NRggCORhh group, where Rhh is:
          • a hydrogen,
          • a C1 to C6 alkyl optionally substituted with:
          •  an alkoxy,
          •  a halogen, or
          •  an amino optionally substituted with one or more C1 to C6 alkyls,
          • an amino optionally substituted with one or more C1 to C6 alkyls,
          • where the alkyls are optionally substituted with a halogen,
          • a 5 or 6 membered heterocycle,
          • a 5 or 6 membered heteroaryl,
        • and Rgg is:
          • a hydrogen,
          • a C1 to C 6 alkyl,
          • a haloalkyl,
          • a haloalkoxy, or
          • a —CORx group, where Rx is as defined above,
        • a haloalkyl,
        • 5 or 6 membered heterocycle groups,
        • an amino optionally substituted with one or more C1 to C6 alkyls,
        • a —NRiiSO2Rx group, where Rx is as defined above, and Rii is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a haloalkyl,
          • a haloalkoxy,
          • a —CORx group, where Rx is as defined above;





  • Z is:
    • a hydrogen;
    • a C1 to C6 alkyl optionally substituted with:
      • an alkoxy,
      • one or more halogens, or
      • a C6 to C8 aryl;
    • a C2 to C6 alkylene;
    • a C6 to C8 aryl optionally substituted with an alkoxy or one or more C1 to C6 alkyls;
    • a —COORx group, where Rx is as defined above; or





embedded image


  • R is a hydrogen, a halogen or an alkoxy;

  • R1 is:
    • a hydrogen;
    • a hydroxy;
    • a halogen;
    • a haloalkyl;
    • a nitro group;
    • a 5 or 6 membered heteroaryl;
    • a 5 or 6 membered heterocycle;
    • an alkoxy optionally substituted with:
      • one or more halogens,
      • a C6 to C8 aryl, or
      • a 5 or 6 membered heterocycle;
    • a C6 to C8 aryl optionally substituted with an alkoxy;
    • a —CORx group, where Rx is as defined above;
    • a C1 to C6 alkyl optionally substituted with a dialkyl-amino or a 5 or 6 membered heterocycle; or

  • R1 joins together with R2 to form:





embedded image


  • R2 is:
    • a nitro group;
    • a hydrogen;
    • a halogen;
    • a hydroxy group;
    • a C1 to C6 alkyl group, optionally substituted with one or more halogens;
    • an amino group;
    • an alkoxy group optionally substituted with:
      • one or more halogens,
      • an —OCORx group, where Rx is as defined above,
      • a dialkyl-amino optionally substituted with an alkoxy,
      • a 5 or 6 membered heterocycle group optionally substituted with a C1 to C6 alkyl,
      • a 5 or 6 membered heteroaryl group, or
      • a C6 to C8 aryl group;
    • a —COORx group, where Rx is as defined above;
    • a haloalkyl;
    • an amide group optionally substituted with:
      • a hydroxy group, or
      • a C6 to C8 aryl;
    • a 5 or 6 membered heteroaryl;
    • a —OCORx group, where Rx is as defined above;
    • a —NHCORjj group, where Rjj is:
      • an alkoxy, or
      • an amino optionally substituted with one or more C1 to C6 alkyls;
    • a —ORkk group, where Rkk is a 5 to 6 membered heteroaryl;
    • a —NHSO2Rx group, where Rx is as defined above; or

  • R2 joins together with R1 to form:





embedded image


  • R3 is:
    • a hydrogen; or
    • CH2OCORx, and Rx is as defined above;


      or a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable excipient, and optionally one or more compound known in the art to affect IRES activity.



Embodiment 30

The pharmaceutical composition of Embodiment 29, wherein said one or more compound known in the art to affect IRES activity affects IRES mediated translation of the single ORF encoding the polyprotein.


Embodiment 31

A method for affecting viral IRES activity in a subject infected with a virus, comprising administering to said subject one or more compound having the following formula:




embedded image



wherein:

  • X is:
    • hydrogen;
    • a nitro group;
    • a cyano group;
    • a —CORa group, where Ra is:
      • a C1 to C6 alkyl,
      • a C6 to C8 aryl optionally substituted with an alkoxy or a halogen, or
      • a dialkyl-amino;
    • a —COORx group, where Rx is a C1 to C6 alkyl;
    • a formyl group;
    • a C6 to C8 aryl optionally substituted with an alkoxy; or
    • a 5 or 6-membered heteroaryl optionally substituted with:
      • a C1 to C6 alkyl,
      • a C6 to C8 aryl optionally substituted with an alkoxy or one or more halogens, or
      • a 5 to 6 membered heteroaryl;
  • Y is:
    • a hydrogen;
    • a haloalkyl;
    • a halogen;
    • an amino optionally substituted with one or more C1 to C6 alkyls;
    • a benzofuran;
    • a benzothiophene;
    • a dibenzofuran;
    • a dibenzothiophene;
    • a benzothiazole;
    • a naphthalene;
    • an indole, optionally substituted on the nitrogen with a C1 to C6 alkyl;




embedded image




    •  where Rb is a hydrogen or a C1 to C6 alkyl, and n is 0 or 1;







embedded image




    •  where Rc is a hydrogen, a —CONHRx, where Rx is as defined above, or an —SO2Rx, where Rx is as defined above; or







embedded image




    •  where Rd is a C1 to C6 alkyl or a C6 to C8 aryl;

    • a —NHCORe group, where Re is:
      • a C1 to C6 alkyl;
      • a C6 to C8 aryl optionally substituted with:
        • a C1 to C6 alkyl,
        • an alkoxy,
        • a cyano group,
        • a nitro group, or
        • a halogen;

    • a —NHCOORx group, where Rx is as defined above;

    • a —CH2O—Rf group, where Rf is a C6 to C8 aryl;

    • a —NRgRh group, where Rg is a C1 to C6 alkyl or a hydrogen and Rh is a C6 to C8 aryl optionally substituted with an alkoxy;

    • a C1 to C6 alkyl;

    • a 5 or 6 membered heteroaryl, optionally substituted with:
      • a C1 to C6 alkyl, optionally substituted with a C6 to C8 aryl,
      • a C6 to C8 aryl, optionally substituted with —COORx, where Rx is as defined above, or
      • an amino group;

    • a 5 or 6 membered heterocycle optionally substituted with:
      • a —COORx group, where Rx is as defined above, or
      • a —NHCOORx group, where Rx is as defined above;

    • a C6 to C8 aryl, optionally substituted with one or more of the following:
      • an alkoxy, optionally substituted with:
        • an alkoxy,
        • a hydroxy,
        • one or more halogens,
        • a 5 or 6 membered heterocycle, optionally substituted with:
          • a C1 to C6 alkyl, or
          • a hydroxy,
        • an amino group optionally substituted with one or more C1 to C6 alkyls,
        • a —NRiSO2Rx group, where Rx is as defined above and Ri is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a —CORx group, where Rx is as defined above,
          • a haloalkyl, or
          • a haloalkoxy,
        • a —NRjCORk group, where Rk is:
          • a C1 to C6 alkyl,
          • a hydrogen, or
          • an amino optionally substituted with one or more C1 to C6 alkyls,
        • and Rj is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a —CORx group, where Rx is as defined above,
          • a haloalkyl, or
          • a haloalkoxy,
        • a —N═N+═N group, or
        • a —CORl, where Rl is a 5 or 6 membered heterocycle optionally substituted with a hydroxy,
      • an amino optionally substituted with one or more C1 to C6 alkyls,
      • a nitro group,
      • a C1 to C6 alkyl group, optionally substituted with:
        • a —NHSO2Rx group, where Rx is as defined above, or
        • a —NRxSO2Rx group, where Rx is as defined above,
      • a haloalkoxy,
      • a halogen,
      • a hydroxy,
      • a —COORx group, where Rx is as defined above,
      • a —CORm group, where Rm is:
        • an amino optionally substituted with one or more C1 to C6 alkyls, where the C1 to C6 alkyls are optionally substituted with:
          • a hydroxy
          • a 5 or 6 membered heterocycle,
          • an amino optionally substituted with one or more C1 to C6 alkyls,
          • an alkoxy,
        • a 3 to 7 membered heterocycle, optionally substituted with a C1 to C6 alkyl, optionally substituted with a dialkyl-amino,
        • a —NHRn group, where Rn is:
          • a —CH2CONH2, or
          • a C6 to C8 aryl optionally substituted with:
          •  an alkyl,
          •  one or more halogens,
          •  a nitro group, or
          •  one or more alkoxys,
      • a —NRoCORp group, where Rp is:
        • a C1 to C6 alkyl optionally substituted with:
          • a halogen,
          • an alkoxy, or
          • a C6 to C8 aryl,
        • a 5 or 6 membered heterocycle,
        • a C6 to C8 aryl, optionally substituted with a halogen,
        • a 5 or 6 membered heteroaryl optionally substituted with one or more C1 to C6 alkyls,
        • a hydrogen,







embedded image








        •  or



      • and where Ro is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a —CORx group, where Rx is as defined above,
        • a haloalkyl, or
        • a haloalkoxy,

      • a —NRqCONRqRr group, where Rq is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a haloalkyl,
        • a haloalkoxy, or
        • a —CORx group, where Rx is as defined above,

      • and where Rr is:
        • a C6 to C8 aryl optionally substituted with:









embedded image










          • a C1 to C6 alkyl,

          • a haloalkyl,

          • a —ORs group, where Rs is a C6 to C8 aryl, or

          • a —COORx group, where Rx is as defined above,



        • a C1 to C6 alkyl optionally substituted with one or more of the following:
          • a halogen,
          • an alkylene,
          • a C6 to C8 aryl, or
          • a —COORx group, where Rx is as defined above,

        • a —COORx group, where Rx is as defined above,



      • a —NRtCOORu group, where Ru is:
        • a C1 to C12 alkyl, optionally substituted with:
          • a C6 to C8 aryl optionally substituted with a C1 to C6 alkyl or an alkoxy,
          • an alkylene,
          • an alkoxy,
          • an alkyne,
          • a halogen, or
          • a 5 or 6 membered heterocycle,
        • a C6 to C8 aryl, optionally substituted with:
          • an alkoxy,
          • a halogen, or
          • a C1 to C6 alkyl, or
        • a 5 or 6 membered heterocycle,

      • and Rt is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a —CORx group, where Rx is as defined above,
        • a haloalkyl, or
        • a haloalkoxy,

      • a —NRvSO2Rw group, where Rv is:
        • a hydrogen,
        • a —CORx, where Rx is as defined above, or
        • a C1 to C6 alkyl, optionally substituted with:
          • a halogen,
          • a —CORx group, where Rx is as defined above,
          • a —OCORx group, where Rx is as defined above,
          • a hydroxyl,
          • a hydroxyl, or
          • an alkoxy,

      • and where Rw is:
        • a C1 to C6 alkyl optionally substituted with:
          • a halogen,
          • a haloalkyl,
          • a C6 to C8 aryl, or
          • a 5 or 6 membered heterocycle,
        • a C2 to C6 alkylene,
        • an alkyl- or dialkyl-amino optionally substituted with a halogen,
        • a 5 or 6 membered heterocycle, or
        • a 5 or 6 membered heteroaryl optionally substituted with:
          • a C1 to C6 alkyl,
          • a 5 or 6 membered heterocycle, or









embedded image




embedded image






      •  optionally substituted with a C1 to C6 alkyl, where Ry is a C1 to C6 alkyl or hydrogen,









embedded image






      • where Rz is hydrogen or a C1 to C6 alkyl, optionally substituted with a C6 to C8 aryl,

      • a —SRx group, where Rx is as defined above,

      • a —SO2Raa group, where Raa is:
        • a C1 to C6 alkyl,
        • an amino group,
        • an alkyl- or dialkyl-amino group optionally substituted with a hydroxy or a —COORx group, where Rx is as defined above,
        • a 5 or 6 membered heteroaryl,

      • a C6 to C8 aryl, or

      • a —NHRbb group, where Rbb is:









embedded image








        • a —C(═S)NH2 group, or

        • a —PO(ORx)2, where Rx is as defined above;











embedded image




    •  group, where Wcc is:
      • a naphthalene,
      • a 5 or 6 membered heteroaryl,







embedded image






      • a C6 to C8 aryl, optionally substituted with one or more of the following:
        • an alkoxy,
        • an hydroxy,
        • a halogen,
        • a C1 to C6 alkyl, optionally substituted with a cyano group,
        • an amino optionally substituted with one or more C1 to C6 alkyls,
        • a —NHPORxRx, where Rx is as defined above,
        • a —NReeCONRffRff group, where Ree is a hydrogen or a C1 to C6 alkyl, optionally substituted with a halogen, and Rff is:
          • a hydrogen,
          • a haloalkyl,
          • a haloalkoxy,
          • a C1 to C6 alkyl, or
          • a —CORx, where Rx is as defined above,
        • a —NRggCORhh group, where Rhh is:
          • a hydrogen,
          • a C1 to C6 alkyl optionally substituted with:
          •  an alkoxy,
          •  a halogen, or
          •  an amino optionally substituted with one or more C1 to C6 alkyls,
          • an amino optionally substituted with one or more C1 to C6 alkyls, where the alkyls are optionally substituted with a halogen,
          • a 5 or 6 membered heterocycle,
          • a 5 or 6 membered heteroaryl,
        • and Rgg is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a haloalkyl,
          • a haloalkoxy, or
          • a —CORx group, where Rx is as defined above,
        • a haloalkyl,
        • 5 or 6 membered heterocycle groups,
        • an amino optionally substituted with one or more C1 to C6 alkyls,
        • a —NRiiSO2Rx group, where Rx is as defined above, and Rii is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a haloalkyl,
          • a haloalkoxy,
          • a —CORx group, where Rx is as defined above;





  • Z is:
    • a hydrogen;
    • a C1 to C6 alkyl optionally substituted with:
      • an alkoxy,
      • one or more halogens, or
      • a C6 to C8 aryl;
    • a C2 to C6 alkylene;
    • a C6 to C8 aryl optionally substituted with an alkoxy or one or more C1 to C6 alkyls;
    • a —COORx group, where Rx is as defined above; or





embedded image


  • R is a hydrogen, a halogen or an alkoxy;

  • R1 is:
    • a hydrogen;
    • a hydroxy;
    • a halogen;
    • a haloalkyl;
    • a nitro group;
    • a 5 or 6 membered heteroaryl;
    • a 5 or 6 membered heterocycle;
    • an alkoxy optionally substituted with:
      • one or more halogens,
      • a C6 to C8 aryl, or
      • a 5 or 6 membered heterocycle;
    • a C6 to C8 aryl optionally substituted with an alkoxy;
    • a —CORx group, where Rx is as defined above;
    • a C1 to C6 alkyl optionally substituted with a dialkyl-amino or a 5 or 6 membered heterocycle; or
    • R1 joins together with R2 to form:





embedded image


  • R2 is:
    • a nitro group;
    • a hydrogen;
    • a halogen;
    • a hydroxy group;
    • a C1 to C6 alkyl group, optionally substituted with one or more halogens;
    • an amino group;
    • an alkoxy group optionally substituted with:
      • one or more halogens,
      • an —OCORx group, where Rx is as defined above,
      • a dialkyl-amino optionally substituted with an alkoxy,
      • a 5 or 6 membered heterocycle group optionally substituted with a C1 to C6 alkyl,
      • a 5 or 6 membered heteroaryl group, or
      • a C6 to C8 aryl group;
    • a —COORx group, where Rx is as defined above;
    • a haloalkyl;
    • an amide group optionally substituted with:
      • a hydroxy group, or
      • a C6 to C8 aryl;
    • a 5 or 6 membered heteroaryl;
    • a —OCORx group, where Rx is as defined above;
    • a —NHCORjj group, where Rjj is:
      • an alkoxy, or
      • an amino optionally substituted with one or more C1 to C6 alkyls;
    • a —ORkk group, where Rkk is a 5 to 6 membered heteroaryl;
    • a —NHSO2Rx group, where Rx is as defined above; or

  • R2 joins together with R1 to form:





embedded image


  • R3 is:
    • a hydrogen; or
    • CH2OCORx, and Rx is as defined above;


      or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient and optionally one or more compound known in the art to affect IRES activity.



Embodiment 32

The method of Embodiment 31, wherein said compound known in the art to affect IRES activity affects IRES mediated translation of the single ORF encoding the polyprotein.


Embodiment 33

A pharmaceutical composition for affecting viral IRES activity in a subject infected with a virus, comprising one or more compound having the following formula, in an amount effective for affecting viral IRES activity:




embedded image



wherein:

  • X is:
    • hydrogen;
    • a nitro group;
    • a cyano group;
    • a —CORa group, where Ra is:
      • a C1 to C6 alkyl,
      • a C6 to C8 aryl optionally substituted with an alkoxy or a halogen, or
      • a dialkyl-amino;
    • a —COORx group, where Rx is a C1 to C6 alkyl;
    • a formyl group;
    • a C6 to C8 aryl optionally substituted with an alkoxy; or
    • a 5 or 6-membered heteroaryl optionally substituted with:
      • a C1 to C6 alkyl,
      • a C6 to C8 aryl optionally substituted with an alkoxy or one or more halogens, or
      • a 5 to 6 membered heteroaryl;
    • Y is:
    • a hydrogen;
    • a haloalkyl;
    • a halogen;
    • an amino optionally substituted with one or more C1 to C6 alkyls;
    • a benzofuran;
    • a benzothiophene;
    • a dibenzofuran;
    • a dibenzothiophene;
    • a benzothiazole;
    • a naphthalene;
    • an indole, optionally substituted on the nitrogen with a C1 to C6 alkyl;




embedded image




    •  where Rb is a hydrogen or a C1 to C6 alkyl, and n is 0 or 1;







embedded image




    •  where Rc is a hydrogen, a —CONHRx, where Rx is as defined above, or an —SO2Rx, where Rx is as defined above; or







embedded image




    •  where Rd is a C1 to C6 alkyl or a C6 to C8 aryl;

    • a —NHCORe group, where Re is:
      • a C1 to C6 alkyl;
      • a C6 to C8 aryl optionally substituted with:
        • a C1 to C6 alkyl,
        • an alkoxy,
        • a cyano group,
        • a nitro group, or
        • a halogen;

    • a —NHCOORx group, where Rx is as defined above;

    • a —CH2O—Rf group, where Rf is a C6 to C8 aryl;

    • a —NRgRh group, where Rg is a C1 to C6 alkyl or a hydrogen and Rh is a C6 to C8 aryl optionally substituted with an alkoxy;

    • a C1 to C6 alkyl;

    • a 5 or 6 membered heteroaryl, optionally substituted with:
      • a C1 to C6 alkyl, optionally substituted with a C6 to C8 aryl,
      • a C6 to C8 aryl, optionally substituted with —COORx, where Rx is as defined above, or
      • an amino group;

    • a 5 or 6 membered heterocycle optionally substituted with:
      • a —COORx group, where Rx is as defined above, or
      • a —NHCOORx group, where Rx is as defined above;

    • a C6 to C8 aryl, optionally substituted with one or more of the following:
      • an alkoxy, optionally substituted with:
        • an alkoxy,
        • a hydroxy,
        • one or more halogens,
        • a 5 or 6 membered heterocycle, optionally substituted with:
          • a C1 to C6 alkyl, or
          • a hydroxy,
        • an amino group optionally substituted with one or more C1 to C6 alkyls,
        • a —NRiSO2Rx group, where Rx is as defined above and Ri is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a —CORx group, where Rx is as defined above,
          • a haloalkyl, or
          • a haloalkoxy,
        • a —NRjCORk group, where Rk is:
          • a C1 to C6 alkyl,
          • a hydrogen, or
          • an amino optionally substituted with one or more C1 to C6 alkyls,
        • and Rj is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a —CORx group, where Rx is as defined above,
          • a haloalkyl, or
          • a haloalkoxy,
        • a —N═N+═N group, or
        • a —CORl, where Rl is a 5 or 6 membered heterocycle optionally substituted with a hydroxy,
      • an amino optionally substituted with one or more C1 to C6 alkyls,
      • a nitro group,
      • a C1 to C6 alkyl group, optionally substituted with:
        • a —NHSO2Rx group, where Rx is as defined above, or
        • a —NRxSO2Rx group, where Rx is as defined above,
      • a haloalkoxy,
      • a halogen,
      • a hydroxy,
      • a —COORx group, where Rx is as defined above,
      • a —CORm group, where Rm is:
        • an amino optionally substituted with one or more C1 to C6 alkyls, where the C1 to C6 alkyls are optionally substituted with:
          • a hydroxy
          • a 5 or 6 membered heterocycle,
          • an amino optionally substituted with one or more C1 to C6 alkyls,
          • an alkoxy,
        • a 3 to 7 membered heterocycle, optionally substituted with a C1 to C6 alkyl, optionally substituted with a dialkyl-amino,
        • a —NHRn group, where Rn is:
          • a —CH2CONH2, or
          • a C6 to C8 aryl optionally substituted with:
          •  an alkyl,
          •  one or more halogens,
          •  a nitro group, or
          •  one or more alkoxys,
      • a —NRoCORp group, where Rp is:
        • a C1 to C6 alkyl optionally substituted with:
          • a halogen,
          • an alkoxy, or
          • a C6 to C8 aryl,
        • a 5 or 6 membered heterocycle,
        • a C6 to C8 aryl, optionally substituted with a halogen,
        • a 5 or 6 membered heteroaryl optionally substituted with one or more C1 to C6 alkyls,
        • a hydrogen,







embedded image






      • and where Ro is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a —CORx group, where Rx is as defined above,
        • a haloalkyl, or
        • a haloalkoxy,

      • a —NRqCONRqRr group, where Rq is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a haloalkyl,
        • a haloalkoxy, or
        • a —CORx group, where Rx is as defined above,

      • and where Rr is:
        • a C6 to C8 aryl optionally substituted with:









embedded image










          • a C1 to C6 alkyl,

          • a haloalkyl,

          • a —ORs group, where Rs is a C6 to C8 aryl, or

          • a —COORx group, where Rx is as defined above,



        • a C1 to C6 alkyl optionally substituted with one or more of the following:
          • a halogen,
          • an alkylene,
          • a C6 to C8 aryl, or
          • a —COORx group, where Rx is as defined above,

        • a —COORx group, where Rx is as defined above,



      • a —NRtCOORu group, where Ru is:
        • a C1 to C12 alkyl, optionally substituted with:
          • a C6 to C8 aryl optionally substituted with a C1 to C6 alkyl or an alkoxy,
          • an alkylene,
          • an alkoxy,
          • an alkyne,
          • a halogen, or
          • a 5 or 6 membered heterocycle,
        • a C6 to C8 aryl, optionally substituted with:
          • an alkoxy,
          • a halogen, or
          • a C1 to C6 alkyl, or
        • a 5 or 6 membered heterocycle,

      • and Rt is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a —CORx group, where Rx is as defined above,
        • a haloalkyl, or
        • a haloalkoxy,

      • a —NRvSO2Rw group, where Rv is:
        • a hydrogen,
        • a —CORx, where Rx is as defined above, or
        • a C1 to C6 alkyl, optionally substituted with:
          • a halogen,
          • a —CORx group, where Rx is as defined above,
          • a —OCORx group, where Rx is as defined above,
          • a hydroxyl,
          • a hydroxyl, or
          • an alkoxy,

      • and where Rw is:
        • a C1 to C6 alkyl optionally substituted with:
          • a halogen,
          • a haloalkyl,
          • a C6 to C8 aryl, or
          • a 5 or 6 membered heterocycle,
        • a C2 to C6 alkylene,
        • an alkyl- or dialkyl-amino optionally substituted with a halogen,
        • a 5 or 6 membered heterocycle, or
        • a 5 or 6 membered heteroaryl optionally substituted with:
          • a C1 to C6 alkyl,
          • a 5 or 6 membered heterocycle, or









embedded image






      •  optionally substituted with a C1 to C6 alkyl, where Ry is a C1 to C6 alkyl or hydrogen,









embedded image






      • where Rz is hydrogen or a C1 to C6 alkyl, optionally substituted with a C6 to C8 aryl,

      • a —SRx group, where Rx is as defined above,

      • a —SO2Raa group, where Raa is:
        • a C1 to C6 alkyl,
        • an amino group,
        • an alkyl- or dialkyl-amino group optionally substituted with a hydroxy or a —COORx group, where Rx is as defined above,
        • a 5 or 6 membered heteroaryl,

      • a C6 to C8 aryl, or

      • a —NHRbb group, where Rbb is:









embedded image








        • a —C(═S)NH2 group, or

        • a —PO(ORx)2, where Rx is as defined above;











embedded image




    •  Rcc group, where Rcc is:
      • a naphthalene,
      • a 5 or 6 membered heteroaryl,







embedded image






      • a C6 to C8 aryl, optionally substituted with one or more of the following:
        • an alkoxy,
        • an hydroxy,
        • a halogen,
        • a C1 to C6 alkyl, optionally substituted with a cyano group,
        • an amino optionally substituted with one or more C1 to C6 alkyls,
        • a —NHPORxRx, where Rx is as defined above,
        • a —NReeCONRffRff group, where Ree is a hydrogen or a C1 to C6 alkyl, optionally substituted with a halogen, and Rff is:
          • a hydrogen,
          • a haloalkyl,
          • a haloalkoxy,
          • a C1 to C6 alkyl, or
          • a —CORx, where Rx is as defined above,
        • a —NRggCORhh group, where Rhh is:
          • a hydrogen,
          • a C1 to C6 alkyl optionally substituted with:
          •  an alkoxy,
          •  a halogen, or
          •  an amino optionally substituted with one or more C1 to C6 alkyls,
          • an amino optionally substituted with one or more C1 to C6 alkyls, where the alkyls are optionally substituted with a halogen,
          • a 5 or 6 membered heterocycle,
          • a 5 or 6 membered heteroaryl,
        • and Rgg is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a haloalkyl,
          • a haloalkoxy, or
          • a —CORx group, where Rx is as defined above,
        • a haloalkyl,
        • 5 or 6 membered heterocycle groups,
        • an amino optionally substituted with one or more C1 to C6 alkyls,
        • a —NRiiSO2Rx group, where Rx is as defined above, and Rii is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a haloalkyl,
          • a haloalkoxy,
          • a —CORx group, where Rx is as defined above;





  • Z is:
    • a hydrogen;
    • a C1 to C6 alkyl optionally substituted with:
      • an alkoxy,
      • one or more halogens, or
      • a C6 to C8 aryl;
    • a C2 to C6 alkylene;
    • a C6 to C8 aryl optionally substituted with an alkoxy or one or more C1 to C6 alkyls;
    • a —COORx group, where Rx is as defined above; or





embedded image


  • R is a hydrogen, a halogen or an alkoxy;

  • R1 is:
    • a hydrogen;
    • a hydroxy;
    • a halogen;
    • a haloalkyl;
    • a nitro group;
    • a 5 or 6 membered heteroaryl;
    • a 5 or 6 membered heterocycle;
    • an alkoxy optionally substituted with:
      • one or more halogens,
      • a C6 to C8 aryl, or
      • a 5 or 6 membered heterocycle;
    • a C6 to C8 aryl optionally substituted with an alkoxy;
    • a —CORx group, where Rx is as defined above;
    • a C1 to C6 alkyl optionally substituted with a dialkyl-amino or a 5 or 6 membered heterocycle; or

  • R1 joins together with R2 to form:





embedded image


  • R2 is:
    • a nitro group;
    • a hydrogen;
    • a halogen;
    • a hydroxy group;
    • a C1 to C6 alkyl group, optionally substituted with one or more halogens;
    • an amino group;
    • an alkoxy group optionally substituted with:
      • one or more halogens,
      • an —OCORx group, where Rx is as defined above,
      • a dialkyl-amino optionally substituted with an alkoxy,
      • a 5 or 6 membered heterocycle group optionally substituted with a C1 to C6 alkyl,
      • a 5 or 6 membered heteroaryl group, or
      • a C6 to C8 aryl group;
    • a —COORx group, where Rx is as defined above;
    • a haloalkyl;
    • an amide group optionally substituted with:
      • a hydroxy group, or
      • a C6 to C8 aryl;
    • a 5 or 6 membered heteroaryl;
    • a —OCORx group, where Rx is as defined above;
    • a —NHCORjj group, where Rjj is:
      • an alkoxy, or
      • an amino optionally substituted with one or more C1 to C6 alkyls;
    • a —ORkk group, where Rkk is a 5 to 6 membered heteroaryl;
    • a —NHSO2Rx group, where Rx is as defined above; or

  • R2 joins together with R1 to form:





embedded image


  • R3 is:
    • a hydrogen; or

  • —CH2OCORx, and Rx is as defined above;

  • or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient.






BRIEF DESCRIPTION OF THE DRAWING


FIG. 1 illustrates the HCV-PV chimera construct. The cloverleaf-like RNA structure of PV, an essential cis-acting replication signal ending with the genome-linked protein VPg, is located at the 5′ end of the genome. The solid (HCV) and open (PV) boxes depict open reading frames encoding viral polypeptides. The position of the HCV core fragment (the first 123 amino acids) gene is denoted by A Core. Overall, the HCV_specific sequence in the HCV-PV spans from nucleotides 18 to 710 (139).





DETAILED DESCRIPTION OF THE INVENTION

In accordance with the present invention, compounds that modify HCV translation have been identified and methods of using these compounds for preventing or treating HCV infection are provided. Without being limited to one theory, it is believed that the compounds of the present invention inhibit IRES-mediated initiation and translation. The HCV IRES directs the translation of a single long ORF encoding a polyprotein that is posttranslationally processed into at least 10 mature viral proteins, including the structural proteins core (putative nucleocapsid), E1 and E2 and the nonstructural (NS) proteins NS2 to NS5B.


A. Compounds of the Invention


In one aspect of the invention, compounds of the invention are provided which are useful for preventing or treating HCV infection.


Preferred compounds of the present invention useful for preventing or treating HCV infection include those of Formula (I) as shown below.




embedded image



wherein:

  • X is:
    • hydrogen;
    • a nitro group;
    • a cyano group;
    • a —CORa group, where Ra is:
      • a C1 to C6 alkyl,
      • a C6 to C8 aryl optionally substituted with an alkoxy or a halogen, or
      • a dialkyl-amino;
    • a —COORx group, where Rx is a C1 to C6 alkyl;
    • a formyl group;
    • a C6 to C8 aryl optionally substituted with an alkoxy; or
    • a 5 or 6-membered heteroaryl optionally substituted with:
      • a C1 to C6 alkyl,
      • a C6 to C8 aryl optionally substituted with an alkoxy or one or more halogens, or
      • a 5 to 6 membered heteroaryl;
  • Y is:
    • a hydrogen;
    • a haloalkyl;
    • a halogen;
    • an amino optionally substituted with one or more C1 to C6 alkyls;
    • a benzofuran;
    • a benzothiophene;
    • a dibenzofuran;
    • a dibenzothiophene;
    • a benzothiazole;
    • a naphthalene;
    • an indole, optionally substituted on the nitrogen with a C1 to C6 alkyl;




embedded image



where Rb is a hydrogen or a C1 to C6 alkyl, and n is 0 or 1;




embedded image



where Rc is a hydrogen, a —CONHRx, where Rx is as defined above, or an —SO2Rx, where Rx is as defined above; or




embedded image



where Rd is a C1 to C6 alkyl or a C6 to C8 aryl;

    • a —NHCORe group, where Re is:
      • a C1 to C6 alkyl;
      • a C6 to C8 aryl optionally substituted with:
        • a C1 to C6 alkyl,
        • an alkoxy,
        • a cyano group,
        • a nitro group, or
        • a halogen;
    • a —NHCOORx group, where Rx is as defined above;
    • a —CH2O—Rf group, where Rf is a C6 to C8 aryl;
    • a —NRgRh group, where Rg is a C1 to C6 alkyl or a hydrogen and Rh is a C6 to C8 aryl optionally substituted with an alkoxy;
    • a C1 to C6 alkyl;
    • a 5 or 6 membered heteroaryl, optionally substituted with:
      • a C1 to C6 alkyl, optionally substituted with a C6 to C8 aryl,
      • a C6 to C8 aryl, optionally substituted with —COORx, where Rx is as defined above, or
      • an amino group;
    • a 5 or 6 membered heterocycle optionally substituted with:
      • a —COORx group, where Rx is as defined above, or
      • a —NHCOORx group, where Rx is as defined above;
    • a C6 to C8 aryl, optionally substituted with one or more of the following:
      • an alkoxy, optionally substituted with:
        • an alkoxy,
        • a hydroxy,
        • one or more halogens,
        • a 5 or 6 membered heterocycle, optionally substituted with:
          • a C1 to C6 alkyl, or
          • a hydroxy,
        • an amino group optionally substituted with one or more C1 to C6 alkyls,
        • a —NRiSO2Rx group, where Rx is as defined above and Ri is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a —CORx group, where Rx is as defined above,
          • a haloalkyl, or
          • a haloalkoxy,
        • a —NRjCORk group, where Rk is:
          • a C1 to C6 alkyl,
          • a hydrogen, or
          • an amino optionally substituted with one or more C1 to C6 alkyls,
        • and Rj is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a —CORx group, where Rx is as defined above,
          • a haloalkyl, or
          • a haloalkoxy,
        • a —N═N+═N group, or
        • a —CORl, where Rl is a 5 or 6 membered heterocycle optionally substituted with a hydroxy,
      • an amino optionally substituted with one or more C1 to C6 alkyls,
      • a nitro group,
      • a C1 to C6 alkyl group, optionally substituted with:
        • a —NHSO2Rx group, where Rx is as defined above, or
        • a —NRxSO2Rx group, where Rx is as defined above,
      • a haloalkoxy,
      • a halogen,
      • a hydroxy,
      • a —COORx group, where Rx is as defined above,
      • a —CORm group, where Rm is:
        • an amino optionally substituted with one or more C1 to C6 alkyls, where the C1 to C6 alkyls are optionally substituted with:
          • a hydroxy
          • a 5 or 6 membered heterocycle,
          • an amino optionally substituted with one or more C1 to C6 alkyls,
          • an alkoxy,
        • a 3 to 7 membered heterocycle, optionally substituted with a C1 to C6 alkyl, optionally substituted with a dialkyl-amino,
        • a —NHRn group, where Rn is:
          • a —CH2CONH2, or
          • a C6 to C8 aryl optionally substituted with:
          •  an alkyl,
          •  one or more halogens,
          •  a nitro group, or
          •  one or more alkoxys,
      • a —NRoCORp group, where Rp is:
        • a C1 to C6 alkyl optionally substituted with:
          • a halogen,
          • an alkoxy, or
          • a C6 to C8 aryl,
        • a 5 or 6 membered heterocycle,
        • a C6 to C8 aryl, optionally substituted with a halogen,
        • a 5 or 6 membered heteroaryl optionally substituted with one or more C1 to C6 alkyls,
        • a hydrogen,




embedded image






      • and where Ro is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a —CORx group, where Rx is as defined above,
        • a haloalkyl, or
        • a haloalkoxy,

      • a —NRqCONRqRr group, where Rq is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a haloalkyl,
        • a haloalkoxy, or
        • a —CORx group, where Rx is as defined above,

      • and where Rr is:
        • a C6 to C8 aryl optionally substituted with:









embedded image










          • a C1 to C6 alkyl,

          • a haloalkyl,

          • a —ORs group, where Rs is a C6 to C8 aryl, or

          • a —COORx group, where Rx is as defined above,



        • a C1 to C6 alkyl optionally substituted with one or more of the following:
          • a halogen,
          • an alkylene,
          • a C6 to C8 aryl, or
          • a —COORx group, where Rx is as defined above,

        • a —COORx group, where Rx is as defined above,



      • a —NRtCOORu group, where Ru is:
        • a C1 to C12 alkyl, optionally substituted with:
          • a C6 to C8 aryl optionally substituted with a C1 to C6 alkyl or an alkoxy,
          • an alkylene,
          • an alkoxy,
          • an alkyne,
          • a halogen, or
          • a 5 or 6 membered heterocycle,
        • a C6 to C8 aryl, optionally substituted with:
          • an alkoxy,
          • a halogen, or
          • a C1 to C6 alkyl, or
        • a 5 or 6 membered heterocycle,

      • and Rt is:
        • a hydrogen,
        • a C1 to C6 alkyl,
        • a —CORx group, where Rx is as defined above,
        • a haloalkyl, or
        • a haloalkoxy,

      • a —NRvSO2Rw group, where Rv is:
        • a hydrogen,
        • a —CORx, where Rx is as defined above, or
        • a C1 to C6 alkyl, optionally substituted with:
          • a halogen,
          • a —CORx group, where Rx is as defined above,
          • a —OCORx group, where Rx is as defined above,
          • a hydroxyl,
          • a hydroxyl, or
          • an alkoxy,

      • and where Rw is:
        • a C1 to C6 alkyl optionally substituted with:
          • a halogen,
          • a haloalkyl,
          • a C6 to C8 aryl, or
          • a 5 or 6 membered heterocycle,
        • a C2 to C6 alkylene,
        • an alkyl- or dialkyl-amino optionally substituted with a halogen,
        • a 5 or 6 membered heterocycle, or
        • a 5 or 6 membered heteroaryl optionally substituted with:
          • a C1 to C6 alkyl,
          • a 5 or 6 membered heterocycle, or









embedded image






      •  optionally substituted with a C1 to C6 alkyl, where Ry is a C1 to C6 alkyl or hydrogen,









embedded image






      •  where Rz is hydrogen or a C1 to C6 alkyl, optionally substituted with a C6 to C8 aryl,

      • a —SRx group, where Rx is as defined above,

      • a —SO2Raa group, where Raa is:
        • a C1 to C6 alkyl,
        • an amino group,
        • an alkyl- or dialkyl-amino group optionally substituted with a hydroxy or a —COORx group, where Rx is as defined above,
        • a 5 or 6 membered heteroaryl,

      • a C6 to C8 aryl, or

      • a —NHRbb group, where Rbb is:









embedded image








        • a —C(═S)NH2 group, or

        • a —PO(ORx)2, where Rx is as defined above;











embedded image




    •  Rcc group, where Rcc is:
      • a naphthalene,
      • a 5 or 6 membered heteroaryl,







embedded image






      • a C6 to C8 aryl, optionally substituted with one or more of the following:
        • an alkoxy,
        • an hydroxy,
        • a halogen,
        • a C1 to C6 alkyl, optionally substituted with a cyano group,
        • an amino optionally substituted with one or more C1 to C6 alkyls,
        • a —NHPORxRx, where Rx is as defined above,
        • a —NReeCONRffRff group, where Ree is a hydrogen or a C1 to C6 alkyl, optionally substituted with a halogen, and Rff is:
          • a hydrogen,
          • a haloalkyl,
          • a haloalkoxy,
          • a C1 to C6 alkyl, or
          • a —CORx, where Rx is as defined above,
        • a —NRggCORhh group, where Rhh is:
          • a hydrogen,
          • a C1 to C6 alkyl optionally substituted with:
          •  an alkoxy,
          •  a halogen, or
          •  an amino optionally substituted with one or more C1 to C6 alkyls,
          • an amino optionally substituted with one or more C1 to C6 alkyls, where the alkyls are optionally substituted with a halogen,
          • a 5 or 6 membered heterocycle,
          • a 5 or 6 membered heteroaryl,
        • and Rgg is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a haloalkyl,
          • a haloalkoxy, or
          • a —CORx group, where Rx is as defined above,
        • a haloalkyl,
        • 5 or 6 membered heterocycle groups,
        • an amino optionally substituted with one or more C1 to C6 alkyls,
        • a —NRiiSO2Rx group, where Rx is as defined above, and Rii is:
          • a hydrogen,
          • a C1 to C6 alkyl,
          • a haloalkyl,
          • a haloalkoxy,
          • a —CORx group, where Rx is as defined above;





  • Z is:
    • a hydrogen;
    • a C1 to C6 alkyl optionally substituted with:
      • an alkoxy,
      • one or more halogens, or
      • a C6 to C8 aryl;
    • a C2 to C6 alkylene;
    • a C6 to C8 aryl optionally substituted with an alkoxy or one or more C1 to C6 alkyls;
    • a —COORx group, where Rx is as defined above; or





embedded image


  • R is a hydrogen, a halogen or an alkoxy;

  • R1 is:
    • a hydrogen;
    • a hydroxy;
    • a halogen;
    • a haloalkyl;
    • a nitro group;
    • a 5 or 6 membered heteroaryl;
    • a 5 or 6 membered heterocycle;
    • an alkoxy optionally substituted with:
      • one or more halogens,
      • a C6 to C8 aryl, or
      • a 5 or 6 membered heterocycle;
    • a C6 to C8 aryl optionally substituted with an alkoxy;
    • a —CORx group, where Rx is as defined above;
    • a C1 to C6 alkyl optionally substituted with a dialkyl-amino or a 5 or 6 membered heterocycle; or

  • R1 joins together with R2 to form:





embedded image


  • R2 is:
    • a nitro group;
    • a hydrogen;
    • a halogen;
    • a hydroxy group;
    • a C1 to C6 alkyl group, optionally substituted with one or more halogens;
    • an amino group;
    • an alkoxy group optionally substituted with:
      • one or more halogens,
      • an —OCORx group, where Rx is as defined above,
      • a dialkyl-amino optionally substituted with an alkoxy,
      • a 5 or 6 membered heterocycle group optionally substituted with a C1 to C6 alkyl,
      • a 5 or 6 membered heteroaryl group, or
      • a C6 to C8 aryl group;
    • a —COORx group, where Rx is as defined above;
    • a haloalkyl;
    • an amide group optionally substituted with:
      • a hydroxy group, or
      • a C6 to C8 aryl;
    • a 5 or 6 membered heteroaryl;
    • a —OCORx group, where Rx is as defined above;
    • a —NHCORjj group, where Rjj is:
      • an alkoxy, or
      • an amino optionally substituted with one or more C1 to C6 alkyls;
    • a —ORkk group, where Rkk is a 5 to 6 membered heteroaryl;
    • a —NHSO2Rx group, where Rx is as defined above; or

  • R2 joins together with R1 to form:





embedded image


  • R3 is:
    • a hydrogen; or
    • CH2OCORx, and Rx is as defined above;

  • or a pharmaceutically acceptable salt thereof.



In another preferred embodiment, a compound or a composition of the present invention includes a compound of Formula I, wherein the compound of Formula I is not




embedded image


As used herein, the term “alkyl” generally refers to saturated hydrocarbyl radicals of straight or branched configuration, including methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, n-hexyl, n-heptyl, octyl, n-octyl, and the like. In some embodiments, alkyl substituents may be C1 to C12, or C1 to C8 or C1 to C6 alkyl groups.


As used herein, “alkylene” generally refers to linear, branched or cyclic alkene radicals having one or more carbon-carbon double bonds, such as C2 to C6 alkylene groups including 3-propenyl.


As used herein, “aryl” refers to a carbocyclic aromatic ring structure. Included in the scope of aryl groups are aromatic rings having from five to twenty carbon atoms. Aryl ring structures include compounds having one or more ring structures, such as mono-, bi-, or tricyclic compounds. Examples of aryl groups that include phenyl, tolyl, anthracenyl, fluorenyl, indenyl, azulenyl, phenanthrenyl (i.e., phenanthrene), and napthyl (i.e., napthalene) ring structures. In certain embodiments, the aryl group may be optionally substituted.


As used herein, “heteroaryl” refers to cyclic aromatic ring structures in which one or more atoms in the ring, the heteroatom(s), is an element other than carbon. Heteroatoms are typically O, S or N atoms. Included within the scope of heteroaryl, and independently selectable, are O, N, and S heteroaryl ring structures. The ring structure may include compounds having one or more ring structures, such as mono-, bi-, or tricyclic compounds. In some embodiments, the heteroaryl groups may be selected from heteroaryl groups that contain two or more heteroatoms, three or more heteroatoms, or four or more heteroatoms. Heteroaryl ring structures may be selected from those that contain five or more atoms, six or more atoms, or eight or more atoms. Examples of heteroaryl ring structures include: acridine, benzimidazole, benzoxazole, benzodioxole, benzofuran, 1,3-diazine, 1,2-diazine, 1,2-diazole, 1,4-diazanaphthalene, furan, furazan, imidazole, indole, isoxazole, isoquinoline, isothiazole, oxazole, purine, pyridazine, pyrazole, pyridine, pyrazine, pyrimidine, pyrrole, quinoline, quinoxaline, thiazole, thiophene, 1,3,5-triazine, 1,2,4-triazine, 1,2,3-triazine, tetrazole and quinazoline.


As used herein, “heterocycle” refers to cyclic ring structures in which one or more atoms in the ring, the heteroatom(s), is an element other than carbon. Heteroatoms are typically O, S or N atoms. Included within the scope of heterocycle, and independently selectable, are O, N, and S heterocycle ring structures. The ring structure may include compounds having one or more ring structures, such as mono-, bi-, or tricyclic compounds. Example of heterocyclo groups include morpholinyl, pyrrolidinonyl, pyrrolidinyl, piperidinyl, piperazinyl, hydantoinyl, valerolactamyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyridinyl, tetrahydroprimidinyl, tetrahydrothiophenyl or tetrahydrothiopyranyl and the like. In certain embodiments, the heterocycle may optionally be substituted.


As used herein, “alkoxy” generally refers to a group with the structure —O—R, where R is an alkyl group as defined above.


For the purposes of this invention, halo substituents may be independently selected from the halogens such as fluorine, chlorine, bromine, iodine, and astatine. A haloalkyl is an alkyl group, as defined above, substituted with one or more halogens. A haloalkoxy is an alkoxy group, as defined above, substituted with one or more halogens.


For the purposes of this invention, where one or more functionalities encompassing X, Y, Z, R, R1, R2, and R3, are incorporated into a molecule of Formula (I), each functionality appearing at any location within the disclosed compound may be independently selected, and as appropriate, independently substituted. Further, where a more generic substituent is set forth for any position in the molecules of the present invention, it is understood that the generic substituent may be replaced with more specific substituents, and the resulting molecules are within the scope of the molecules of the present invention.


By “substituted” or “optionally substituted” it is meant that the particular substituent may be substituted with a chemical group known to one of skill in the art to be appropriate for the referred-to substituent, unless a chemical group is specifically mentioned.


Exemplary X substituents include the following, where the * indicates the bond of attachment of the scaffold molecule.




embedded image


embedded image


Preferred X substituents include —hydrogen; —a cyano group; and —a —CORa group, where Ra is: —a C1 to C6 alkyl, or —a dialkyl-amino.


Preferred X substituents also include the following, where the * indicates the bond of attachment of the scaffold molecule.




embedded image


More preferred X substituents include the following, where the * indicates the bond of attachment of the scaffold molecule.




embedded image


Exemplary Y substituents include the following, where the * indicates the bond of attachment of the scaffold molecule.




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


Preferred Y substituents include the following, where the * indicates the bond of attachment of the scaffold molecule.




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


More preferred Y substituents include the following, where the * indicates the bond of attachment of the scaffold molecule.




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


Exemplary Z substituents include the following, where the * indicates the bond of attachment of the scaffold molecule.




embedded image


embedded image


Preferred Z substituents include —a hydrogen; —a C1 to C6 alkyl optionally substituted with: —an alkoxy, —one or more halogens, or —a C6 to C8 aryl; —a C2 to C6 alkylene; and —a C6 to C8 aryl optionally substituted with an alkoxy.


Preferred Z substituents also include the following, where the * indicates the bond of attachment of the scaffold molecule.




embedded image


More preferred Z substituents include —a hydrogen; —a C1 to C6 alkyl optionally substituted with: —a C6 to C8 aryl; —a C2 to C6 alkylene; and —a C6 to C8 aryl optionally substituted with an alkoxy.


More preferred Z substituents include the following, where the * indicates the bond of attachment of the scaffold molecule.




embedded image


Exemplary R substituents include the following:




embedded image


Preferred R substituents include the following:




embedded image


Exemplary R1 substituents include the following:




embedded image


embedded image


Preferred R1 substituents include —a hydrogen; —a halogen; —a nitro group; —a 5 or 6 membered heterocycle; —an alkoxy optionally substituted with: —a C6 to C8 aryl; —a C6 to C8 aryl optionally substituted with an alkoxy.


Preferred R1 substituents also include the following:




embedded image


More preferred R1 substituents include the following:




embedded image


Exemplary R2 substituents include the following:




embedded image


embedded image


embedded image


embedded image


Preferred R2 substituents include —a nitro group; —a hydrogen; —a halogen; _13 a hydroxy group; —a C1 to C6 alkyl group, optionally substituted with one or more halogens; an alkoxy group optionally substituted with: —one or more halogens, —an —OCORx group, where Rx is as defined above, —a dialkyl-amino optionally substituted with an alkoxy, —a 5 or 6 membered heterocycle group optionally substituted with a C1 to C6 alkyl, or —a 5 or 6 membered heteroaryl group; —an amide group; and —a —NHSO2Rx group, where Rx is as defined above.


Preferred R2 substituents also include the following:




embedded image


embedded image


embedded image


More preferred R2 substituents include —a hydrogen; _—a C1 to C6 alkyl group, optionally substituted with one or more halogens; —an alkoxy group optionally substituted with: —one or more halogens, —a 5 or 6 membered heterocycle group optionally substituted with a C1 to C6 alkyl, or —a 5 or 6 membered heteroaryl group.


More preferred R2 substituents also include the following:




embedded image


Exemplary R3 substituents include the following:




embedded image


Preferred R3 substituents include the following:




embedded image


Compounds of the invention include the following:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


The above compounds were prepared using the schemes and examples set forth below. Other methods of producing these compounds are known to one of skill in the art.


Preferred compounds include the following compounds in Table A:









TABLE A









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image











More preferred compounds include the following compounds in Table B:









TABLE B









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image









embedded image











B. Preparation of Compounds of the Invention


Indole compounds of the present invention can be obtained via standard, well-known synthetic methodology. Many of the indole starting materials can be prepared the routes described below or by those skilled in the art.


Compounds of formula I, represented by structure II can be prepared by the methodology depicted in Scheme A below:


An α-nitroketone derivative A2 can be derived from treatment of the anion of nitromethane, obtained from the treatment of nitromethane with a base, such as, e.g., sodium or potassium t-butoxide or sodium hydride, with an activated carboxylic acid derivative, e.g., the acyl imidazolide A1. Reaction of the α-nitroketone A2 with amine derivative A3 can afford the nitro enamine A4 by mixing the components A3 and A4 and heating in a suitable solvent such as an alcohol or an aprotic solvent. Treatment of the nitro enamine A4 with quinone A5 in a polar protic solvent such as acetic acid at or near ambient temerature gives the compound of formula II.




embedded image


Compounds of formula I, represented by structure III can be prepared as shown in Scheme B below:


Treatment of B1 with a reactive alkyl or aryl group containing a leaving group L in a suitable solvent, with or without heat in the presence of a base, such an inorganic base, e.g., sodium or potassium carbonate or an organic base, e.g., triethylamine, can afford the compound of structure III. Examples of leaving groups include but are not limited to halogens (e.g., chlorine, bromine or iodine) or alkyl or arylsulfonates.




embedded image


Compounds of formula I, represented by structure IV can be prepared as shown in Scheme C below:


Compounds of structure IV can be obtained by nitrating an indole of structure C1, to give the 3-nitroindole C2. The nitration can be carried out by treatment of C1 with a nitrating agent, such as nitric acid or sodium nitrite in a solvent such as acetic acid, acetic anhydride, sulfuric acid or in a mixed solvent system containing an organic solvent such as dichloromethane. The reaction can be carried out a temperature of −30° C. to +50° C. Treatment of C2 with a reactive functional group R9 containing a suitable leaving group L (C3) can give compounds of structure IV. Reactive functional groups can consist of but are not limited to alkyl and aralkyl. L can represent a halide, particularly chloro, bromo or iodo or an alkylsulfonate. The reaction between C2 and C3 can be carried out in a suitable solvent in the presence of an inorganic base such as potassium carbonate or sodium hydride or an organic base such as a trialkylamine. Alternatively, the group R9 can represent an aryl or heteroaryl group and L can represent a halide, particularly chloro, bromo or iodo. The reaction can be carried out in a polar or nonpolar solvent at a temperature from ambient to 200° C. in the presence of a copper catalyst, e.g., CuI, a base such as Cs2CO3 or K3PO4, and optionally an amine ligand such as 1,2-bis(methylamino)ethane or 1,2-cyclohexanediamine.


An alternative pathway is to convert C1 into C4 in similar fashion as described above and then carry out the nitration reaction to afford compounds of structure IV.




embedded image


Compounds of formula I, represented by structure V can be prepared as shown in Scheme D.


Treatment of β-ketoesters of structure D1 with amines D2 gives the amino crotonate derivatives D3 by heating in a suitable solvent such as an alcohol or an aprotic solvent. Reaction between D3 and quinone D4 in a polar protic solvent, such as acetic acid gives compounds of structure V.




embedded image


Compounds of the present invention, represented by structure VI compounds can be prepared by the chemistry described in scheme E below.


Indole-3-carboxylic esters E1 can be converted to indole-3-carboxylic acids E2 by treatment of compounds of structure E1 with, for example, either acid or base in aqueous or mixed aqueous-organic solvents at ambient or elevated temperature or by treatment with nucleophilic agents, for example, boron tribromide or trimethylsilyl iodide, in a suitable solvent. Compounds of type E2 can then be activated and treated with amines of type E3 to give compounds E4. Activation of the carboxylic acid can be carried out, for example, by any of the standard methods. For example, the acid E2 can be activated with coupling reagents such as EDCI or DCC with or without HOBt in the presence of the amine E3, or alternatively the acid can be activated as the acid chloride by treatment of the acid with, e.g., thionyl chloride or oxalyl chloride or as the acyl imidazolide, obtained by treatment of the acid with carbonyl diimidazole, followed by treatment of the amine E3. Compounds E4 can be converted to compounds of structure VI by treatment of E4 with a reactive functional group R9 containing a suitable leaving group L (E5) as described previously. Alternatively, compounds of type E1 can be converted to compounds of structure E6 by treatment with E5. Indole-3-carboxylic esters E6 can then be converted to indole-3-carboxylic acids E7 by the methods described above. Conversion of E7 to compounds of structure VI can be carried out by the activation and reaction with an amine E3 as described above.




embedded image


Compounds of the present invention, represented by structure VII compounds can be prepared by the chemistry described in scheme F below.


Indoles F1 can be formylated with reagents such as phosphorous oxychloride in the presence of DMF to give the indole-3-carboxaldehydes F2. Conversion to compounds of structure VII can be accomplished by treatment of F2 with compounds F3 as described previously. Alternatively, compounds of type F1 can first be converted to F4 and then be formylated to compounds of structure VII.




embedded image


Compounds of formula G, represented by structure VIII can be prepared as shown in Scheme G.


Indole-3-carboxaldehydes of structure G1 can be converted to the indole-3-carboxylic acid derivatives by oxidation with reagents such as potassium permanganate under aqueous conditions.




embedded image


Compounds of formula H, represented by structure IX can be prepared as shown in Scheme H.


Indole-3-carboxaldehydes of structure H1 can be converted to the indole-3-carbonitrile derivatives H2 by a variety of methods. Treatment of H1 with a nitroalkane, e.g., nitropropane, in the presence of an amine source, e.g., ammonium hydrogen phosphate gives the indole-3-carbonitrile H2 derivative. An alternative pathway to compound H2 is via the intermediate H3. Conversion of H1 to the oxime derivative H3 can be followed by dehydration, e.g., treatment of the oxime with acetic anhydride and a base, or reaction of the oxime with thionyl chloride to give H2. The compound H2 can then be reacted with a reactive functional group R9 containing a suitable leaving group L (H4) as described previously to afford compounds of structure IX.


Alternatively, H1 can be reacted with a reactive functional group R9 containing a suitable leaving group L (H4) to give the intermediate H5 which can be reacted with a nitroalkane as above to give the indole-3-carbonitrile IX compound. Compound IX can also be obtained by conversion to the oxime H6 followed by a dehydration reaction as described above.




embedded image


Compounds of the present invention, represented by structure X can also be prepared as described in scheme I below.


Indoles I1 can be cyanated with an appropriate cyanating agent, e.g., chlorosulfonyl isocyanate (I2) or a dialkyl phosphoryl isocyanate in a suitable solvent or solvent mixture, e.g. DMF, CH3CN or dioxane, to afford compounds of structure I3. The compound I3 can then be reacted with a reactive functional group R9 containing a suitable leaving group L (I4) as described previously afford the compound X.


Alternatively, compound I1 can be reacted with a reactive functional group R9 containing a suitable leaving group L to give compounds of structure 15 which can then be cyanated as above to give compounds of formula X.




embedded image


Compounds of formula J, represented by structure XI can be prepared as shown in Scheme J.


Amino crotonates J1 can be reacted with amines J2 to give J3. Reaction of J3 with quinone in the presence of a polar, protic solvent, e.g., acetic acid, gives the compound of structure XI.




embedded image


Compounds of the present invention, represented by structure XII and XIII can be prepared as described in scheme K below.


Aldehydes of structure K1 can be reacted with an alkyl azidoacetate K2 by heating the components together in a suitable organic solvent, e.g., a protic or non-protic solvent, in the presence of an organic or inorganic base, to give the α-azidoacrylate K3. Heating K3 in the presence of a suitable non-reactive organic solvent, e.g., toluene or xylenes can give the 2-alkoxycarbonylindoles K4. Reduction of the ester functionality with a suitable reducing reagent, for example, lithium aluminum hydride, in a suitable solvent, e.g., ether or THF can give the intermediate K5. Reaction of K5 with a reactive functional group R9 containing a suitable leaving group L (K6) as described in previously affords the compound K7. Cyanation of K7 with a cyanating agent, e.g., chlorosulfonyl isocyanate as described previously can give compound XII. Alternatively, cyanation of K5 with chlorosulfonyl isocyanate gives K8, which can be reacted with a reactive functional group R9 containing a suitable leaving group L (K6) as described previously, affords, the compound XII.


An alternative use of intermediate K4 is exemplified below. Hydrolysis of the 2-alkoxycarbonyl group of the indole K4 either under acidic or basic conditions followed by decarboxylation can give the intermediate K9. Decarboxylation can be carried out thermally, i.e., heating in an appropriate solvent, e.g., toluene, xylenes, or quinoline. Alternatively, a source of copper can be added, for example, copper bronze, to facilitate decarboxylation. Reaction of K9 with a reactive functional group R9 containing a suitable leaving group L (K6) as described above can afford the compounds K10. Cyanation of K10 with a cyanating agent, e.g., chlorosulfonyl isocyanate as described previously can give compound XIII. Alternatively, cyanation of K9 with chlorosulfonyl isocyanate gives K11, which can be reacted with a reactive functional group R9 containing a suitable leaving group L (K6) as described in previously, affords the compound XIII.




embedded image


Compounds of formula L, represented by structure XIV can be prepared as shown in Scheme L.


Compounds of formula L1 can be halogenated on the 2-methyl group to give 2-bromomethyl or chloromethyl indoles L2. The halogenation reaction can be conducted with reagents, e.g., N-bromo- or chlorosuccinimide. The reaction can be conducted in a suitable solvent, such as chloroform, carbon tetrachloride, or THF and carried out in a range between ambient temperature and 80° C. Optionally, a radical initiator may be added, e.g., benzoyl peroxide or AIBN. The compound L2 can then be reacted with a nucleophile R5—W (L3) to give compounds of structure XIV. The reaction can be conducted in a suitable solvent, e.g., THF, CH2Cl2 or DMF, within a temperature range of 0° C. to 120° C. A base, e.g., an inorganic base, such as potassium carbonate or an organic base, such as a trialkylamine can be used to remove the acid formed in the reaction. The group W can refer to an N, O or S atom.




embedded image


Compounds of the present invention, represented by structure XV can be prepared as described in scheme M below.


Anilines of structure M1 can be diazotized and the resulting diazonium salt can be reduced to give the phenyl hydrazine compound M2. Reaction between the hydrazine M2 and a ketone M3 under acidic conditions can give the indole compound M4. The conditions for the cyclization reaction can be carried out under typical conditions utilized by one skilled in the art, for example, acidic conditions, utilizing acids such as a Bronstead acid ,e.g., acetic acid, hydrochloric acid or polyphosphoric acid or a Lewis acid, e.g., zinc chloride. The reaction can be carried out in the presence of a co-solvent, e.g., CH2Cl2 or THF typically within a temperature range of 0° C. to 120° C. Reaction of M4 with a reactive functional group R9 containing a suitable leaving group L (M5) as described previously, can afford compounds M6. Cyanation of the indole M6 with a cyanating agent such as chlorosulfonyl isocyanate can give the compound of structure XV.


Alternatively, the indole M4 can be cyanated to give compounds of structure M7. Reaction of M7 with a reactive functional group R9 containing a suitable leaving group L (M5) as described above can give compounds of structure XV.




embedded image


Compounds of formula I, represented by structure XVI can be prepared as shown in Scheme N.


Compounds of formula N1 can be reacted with a dialkylformamide dialkyl acetal, N2, e.g., dimethylformamide dimethyl acetal, optionally in the presence of a suitable solvent, e.g., DMF or dioxane, at a temperature range from ambient to 150° C. to give the compound of structure N3. Reduction of the nitro group of compounds of type N3 under standard conditions can give the indole compounds of structure N4. The reduction can be carried out via hydrogenation, using a sub-stoichiometric amount of a hydrogenation catalyst, e.g., platinum or palladium, in the presence of a hydrogen source in a protic or aprotic solvent. The reduction can be carried out in a temperature range of ambient to 80° C. Alternatively, the reduction can be carried out via chemical reduction, e.g., in the presence of stoichiometric amounts of Fe or Sn compounds in a suitable solvent at a temperature range of ambient to 100° C. The compound N4 can then be reacted with a reactive functional group R9 containing a suitable leaving group L (N5) as described previously to afford compounds of structure N6. Cyanation of N6 with a cyanating agent such as chlorosulfonyl isocyanate in a suitable solvent can give the compounds of structure XVI.


Alternatively, compounds of structure N4 can be cyanated to give compounds of structure N7. Reaction with N7 with a reactive functional group R9 containing a suitable leaving group L (N5) as described above can give compounds of structure XVI.




embedded image


Compounds of formula I, represented by structure XVII can be prepared as shown in Scheme O.


Compounds of structure O1 can be converted to 2-iodo- or bromoindoles O2. Typically, a strong base, such as n-butyllithium or s-butyllithium or lithium diisopropylamide or lithium or potassium hexamethyldisilazide is employed, with formation of the 2-indolyl anion generated in a suitable unreactive solvent, e.g., ether or THF, or solvent mixtures containing them. The reaction is typically carried out in the range of −78° C. to ambient temperature. The 2-indolyl anion can then be quenched with an electrophilic source of halogen, including but not limited to iodine, bromine or N-bromosuccinimide to give compounds of structure O2. Reaction of 2-iodo- or bromoindoles O2 with a boronic acid (commonly referred to as a Suzuki reaction) or trialkyl stannane (commonly referred to as a Stille reaction) can give the compounds of structure XVII. The coupling reactions are carried out by methods known to those skilled in the art and include conducting the reaction in the presence of a catalyst, such as tetrakis (triphenylphosphine) palladium (0), bis (triphenylphosphine) palladium (II) dichloride or palladium acetate with added phosphine ligand. The reactions are carried out in a suitable solvent, e.g., DMF, toluene, dimethoxy ethane or dioxane at a temperature range of ambient to 150° C. For the Suzuki reaction, a base is usually added. The base can be in aqueous solution, e.g., aqueous sodium carbonate or sodium bicarbonate, or the base can be employed under anhydrous conditions, e.g., cesium or potassium fluoride. For the Stille reaction a copper co-catalyst, e.g., copper iodide, can be added.


Alternatively, indoles O1 can be converted to the indole-2-boronic acid or indole-2-trialkylstannane derivatives O3 by reacting the 2-indolyl anion described above with a trialkylborate or chlorotrialkyl stannane derivative, respectively. Compounds of type O3 can be reacted with aryl and heteroaryl bromides and iodides under similar conditions to those described above to form compounds of structure XVII.




embedded image


Compounds of formula I, represented by structure XVIII can be prepared as shown in Scheme P.


Compounds of structure P1 can be converted to compounds P3 by treatment of P1 with an aryl or heteroaryl halide (P2) in the presence of organometallic catalysis. Such catalyst combinations can include palladium catalysts, e.g., palladium acetate and a source of copper, e.g., copper iodide. The reaction can be carried out in the presence of a base, e.g., cesium carbonate. The reaction can be carried out within a temperature range of ambient temperature to 150° C.




embedded image


Compounds of the present invention, represented by structure XIX can be prepared as described in scheme Q below.


Compounds of structure XIX can be prepared by protecting an indole compound of structure Q1 as e.g., the N-Boc derivative Q2. Alternatively, other protecting groups which can be utilized but not limited to include , e.g., benzyl, alkyl or aryl sulfonyl, or trialkyl silyl. Treatment of Q2 with a strong base, e.g., lithium diisopropyl amide in an aprotic solvent, e.g., THF followed by quenching with a trialkylborate derivative can give the indolyl-2-boronic acid Q3. Reaction with an aryl or heteroaryl halide Q4 in the presence of palladium catalysis, e.g., tetrakis (triphenylphosphine) palladium (0), bis (triphenylphosphine) palladium (II) dichloride or palladium acetate with added phosphine ligand, can give the compound Q5. Removal of the protecting group can give Q6. Reaction with Q6 with a reactive functional group R9 containing a suitable leaving group L as described above can give compounds of structure Q7. Cyanation of compound Q7 can give the compounds of structure XIX.




embedded image


Compounds of formula I, represented by structure XX can be prepared as shown in Scheme R.


Compounds of structure R1 can be prepared by protecting an indole compound of structure R1 as e.g., the N-Boc derivative R2 as above. Compounds of structure R2 can be converted to 2-iodo- or bromoindoles R3. Typically, a strong base, such as n-butyllithium or s-butyllithium or lithium diisopropylamide or lithium or potassium hexamethyldisilazide is employed, with formation of the 2-indolyl anion generated in a suitable unreactive solvent, e.g., ether or THF, or solvent mixtures containing them. The reaction is typically carried out in the range of −78° C. to ambient temperature. The 2-indolyl anion can then be quenched with an electrophilic source of halogen, including but not limited to iodine, bromine or N-bromosuccinimide to give compounds of structure R3. After removal of the protecting group, compounds of R4 can be reacted with aryl or heteroaryl boronic acids or esters (R5) (commonly referred to as a Suzuki reaction) to give compounds of structure R6. The coupling reactions are carried out by methods known to those skilled in the art and include conducting the reaction in the presence of a catalyst, such as tetrakis (triphenylphosphine) palladium (0), bis (triphenylphosphine) palladium (II) dichloride or palladium acetate with added phosphine ligand. Reaction with R6 with a reactive functional group R9 containing a suitable leaving group L as described above can give compounds of structure XX.




embedded image


Compounds of the present invention, represented by structure XXI can be prepared as described in scheme S below.


2-iodo- or bromoindoles of structure S1 can be reacted with alkenes in the presence of a palladium catalyst (commonly referred to as the Heck reaction) to give compounds of type XXI. The coupling reactions can be carried out by methods known to those skilled in the art. The choice of catalyst and solvents are similar to those described previously.




embedded image


Compounds of formula I, represented by structure XXII can be prepared as shown in Scheme T.


2-Iodo- or 2-bromoindoles of structure T1 can be reacted with acetylenes in the presence of a palladium catalyst (commonly referred to as the Sonagashira reaction) to give compounds of type XXII. The coupling reactions can be carried out by methods known to those skilled in the art. A typical set of reaction conditions includes reacting the indoles of structure T1 with an acetylene compound T2 in the presence of a source of palladium, a copper co-catalyst and an amine source. The reaction is carried out in a suitably unreactive solvent and conducted within a temperature range from ambient to 150° C.




embedded image


Compounds of formula I, represented by structure XXIII can be prepared as shown in Scheme U.


Compounds of structure XXIII can be obtained from the reduction of compounds XXI and XXII. Conditions for the reduction can include, but are not limited to catalytic reduction, e.g., hydrogenation over a source of platinum or palladium in a suitable solvent, e.g., CH2Cl2, ether, THF, methanol or solvent combinations.




embedded image


Compounds of the present invention, represented by structure XXIV can be prepared as described in scheme V below.


Indoles of structure VI can be reacted with a suitable base, such as lithium diisopropylamide or potassium hexamethyldisilazide to generate the 2-indolyl anion in a suitable unreactive solvent, e.g., ether or THF, or solvent mixtures containing them. The reaction is typically carried out in the range of −78° C. to ambient temperature. The 2-indolyl anion can then be quenched with a source of zinc halide, e.g., zinc halide metal or soluitions containing them to give organozinc compounds of structure V2. Reaction of V2 with an arylhalide (V3) in the presence of a palladium catalyst (commonly referred to as the Negishi reaction) gives compounds of structure XXIV. Alternatively, 2-iodo or bromoindoles of structure V4, prepared from compounds V1 as described previously, can be reacted with organozinc compounds of structure V5 in the presence of a suitable palladium catalyst to give compounds of structure XXIV. The organozinc compound V5 can be derived from, e.g., an alkyl or alkenyl halide after treatment with activated zinc or an aryl or heteroaryl lithium or magnesium comound after treatment with zinc halide. Furthermore, the reactions of V2 or V4 can be carried out in the presence of a palladium source, e.g., as tetrakis (triphenylphosphine) palladium (0) or bis (triphenylphosphine) palladium (II) dichloride in a suitable solvent and at a temperature range from ambient to 150° C.




embedded image


Compounds of formula I, represented by structure XXV-XXVIII can be prepared as shown in Scheme W.


2-Iodo- or bromoindoles of structure W1 can be reacted with acetylenes of structure W2 in the presence of a palladium catalyst (commonly referred to as the Sonagashira reaction) to give compounds of type XXV. The coupling reactions can be carried out by methods known to those skilled in the art. A typical set of reaction conditions includes reacting the indoles of structure W1 with an acetylene compound W2 in the presence of a source of palladium, an optional copper co-catalyst and an amine source. The reaction is carried out in a suitably unreactive solvent and conducted within a temperature range from ambient to 150° C. Reaction with XXV with a reactive functional group R9 containing a suitable leaving group L as described above can give compounds of structure XXVI.


2-iodo- or bromoindoles of structure W1 can also be reacted with alkenes in the presence of a palladium catalyst (commonly referred to as the Heck reaction) to give compounds of type XXVII. The coupling reactions can be carried out by methods known to those skilled in the art. The choice of catalyst and solvents are similar to those described previously. Reaction with XXVII with a reactive functional group R9 containing a suitable leaving group L as described above can give compounds of structure XXVIII.




embedded image


Compounds of formula I, represented by structure XXIX can be prepared as shown in Scheme X.


Indoles of structure X1 and be acylated with acyl halides of structure X2 to give compounds of structure XXIX. The reaction can be promoted with a Lewis acid. The choice of Lewis acid can be chosen from, but is not limited to aluminum chloride, ferric chloride, stannic chloride or diethyl aluminum. The reaction is typically carried out in a suitable non-reactive solvent including CH2Cl2, carbon disulfide or dichloroethane and is typically conducted within a temperature range of −20° C. to 80° C.




embedded image


Compounds of formula I, represented by structure XXX can be prepared as shown in Scheme Y.


3-Cyanoindoles of structure Y1 can be converted to tetrazoles of structure Y2 by treatment with, e.g., sodium azide. Heating a mixture of Y2 and the reagent Y3 can give the 3-(1,2,4-oxadiazolyl)indole compound XXX. The reagent Y3 can be, e.g., an acyl halide or an acid derivative activated with a reagent such as dicyclohexyl carbodiimide or diisopropyl carbodiimide. The reaction can be carried out in a variety of solvents, including e.g., toluene, dioxane, pyridine and dichloroethane and can be carried out by heating Y2 and Y3 at a temperature range of 30° to 130° C.




embedded image


Compounds of formula I, represented by structure XXXI can be prepared as shown in Scheme Z.


3-Cyanoindoles of structure Z1 can be treated with hydroxylamine to give hydroxyamidine compounds of formula Z2. Reaction of hydroxyamidines of structure Z2 with compounds of structure Z3 can give O-acylhydroxyamidines Z4. Compounds Z3 can represent, for example, acyl halides or carboxylic acids activated with a reagent such as dicyclohexyl carbodiimide or diisopropyl carbodiimide. Heating compounds of structure Z4 in a non-reactive organic solvent, e.g., toluene, dichloroethane or dioxane in a temperature range of 30° C. to 150° C. can give compounds of structure XXXI.




embedded image


Compounds of the present invention, represented by structure XXXII can be prepared as described in scheme AA below.


Ketoindoles of type AA1 can be converted to oximes of structure AA2 by heating the ketoindoles with hydroxylamine (free base or acid salt) in a suitable solvent. Bis-deprotonation of compounds of type AA2 with a strong organic base (e.g., n-butyllityium or sec-butyllithium or tert-butyllithium) followed by reaction with DMF can give compounds of formula XXXII.




embedded image


Compounds of formula I, represented by structure XXXIII can be prepared as shown in Scheme AB.


3-Ketoindoles of structure AB1 can be homologated to vinylogous amides of structure AB3 by reaction with dialkyl amide dialkyl acetals AB2. The dialkyl amides can include e.g., lower alkyl amides such as formamide, acetamide and propionamide. Examples would include dimethlformamide dimethyl acetal and dimethyl acetamide dimethyl acetal. The reaction can be conducted by reacting AB1 and AB2 with or without additional solvent at a temperature from ambient to 150° C. Treatment of AB3 with hydroxylamine (free base or acid salt) in a suitable solvent can give compounds of structure XXXIII. The reaction is typically conducted within a temperature range from ambient to 120° C.




embedded image


Compounds of formula I, represented by structure XXXIV can be prepared as shown in Scheme AC.


Vinylogous amides of structure AC1 (as prepared above) can be treated with hydrazines AC2 in a suitable organic solvent (DMF, alcohol or acetic acid) at temperatures ranging from ambient temperature to 150° C. to give compounds of structure XXXIV.




embedded image


Compounds of the present invention, represented by structure XXXV can be prepared as described in scheme AD below.


Indole-3-carboxaldehydes of structure AD1 (as prepared in Scheme F) can be reacted with p-(toluenesulfonyl)methyl isocyanate (TOSMIC) in the presence of a base to give compounds of structure XXXV. Bases can include potassium carbonate or 1,8-diazabicyclo[5.4.0]undec-7-ene and the reaction can be carried out in a suitable organic solvent from ambient temperature to 150° C.




embedded image


Compounds of formula I, represented by structure XXXVI and XXXVII can be prepared as shown in Scheme AE.


3-Indolecarboxylic acids of structure AE1 (from Scheme E) can be converted to amides of structure AE2. Compounds of structure AE2 can be activated by any of the standard methods. For example, the acid AE1 can be activated with coupling reagents such as EDCI or DCC with or without HOBt in the presence of ammonia. Alternatively, the acid can be activated as the acid chloride or as the acyl imidazolide as described previously, followed by treatment of ammonia.


The indole-3-carboxamides of structure AE2 can be reacted with substituted aldehydes or ketones (AE3) containing a suitable leaving group L, in a suitable solvent at temperatures above ambient and up to 200° C. The reaction can be performed with or without added base to afford oxazoles of structure XXXVI.


The indole-3-carboxamides of structure AE2 can also be converted to thioamides of structure AE4 by treating the primary amides with Lawesson's reagent or phosphorous pentasulfide at or above ambient temperature in a suitable organic solvent. The resulting thioamides AE4 can be reacted with substituted aldehydes or ketones containing a suitable leaving group L (AE3), in a suitable solvent at temperatures above ambient and up to 150° C. The reaction can be performed with or without added base to afford thiazoles of structure XXXVII.




embedded image


Compounds of the present invention, represented by structure XXXVIII and XXXIX can be prepared as described in scheme AF below.


3-Ketoindoles of structure AF1 can be halogenated (e.g., brominated) to give compounds of structure AF3. Suitable brominating agents can include but are not limited to phenyltrimethylammonium tribromide (AF2), N-bromosuccinimide or bromine and can be carried out in a variety of organic solvents.


Treatment of compounds AF3 with amides of type AF4 in a suitable solvent at temperatures above ambient and up to 200° C. with or without added base can give oxazoles of structure XXXVIII.


Treatment of compounds AF3 with thioamides of type AF5 in a suitable solvent at temperatures above ambient and up to 150° C. with or without added base can give thiazoles of structure XXXIX.




embedded image


Compounds of formula I, represented by structure XL can be prepared as shown in Scheme AG.


Indoles of structure AG1 can be brominated or iodinated to give compounds of structure AG2. Brominating agents may include but are notlimited to bromine or N-bromosuccinimide and iodinating reagents may include iodine monochloride or bis-trifluoroacetoxy iodobenzene. Reaction of 3-iodo- or bromoindoles AG2 with a boronic acid AG3 (commonly referred to as a Suzuki reaction) can give the compounds of structure XL. The coupling reactions are carried out by methods known to those skilled in the art and include conducting the reaction in the presence of a catalyst, such as tetrakis (triphenylphosphine) palladium (0), bis (triphenylphosphine) palladium (II) dichloride or palladium acetate with added phosphine ligand. The reactions are carried out in a suitable solvent, e.g., DMF, toluene, dimethoxy ethane or dioxane at a temperature range of ambient to 150° C. and typically in the presence of a base e.g., aqueous sodium carbonate or sodium bicarbonate, or the base can be employed under anhydrous conditions, e.g., cesium or potassium fluoride.


Alternatively, indole AG2 can be converted to the indole-3-boronic acid derivative AG5 by reacting the 3-haloindole AG2 with a strong organic base (alkyllithium or grignard reagent) and reacting the resultant anion with a trialkylborate reagent AG4. Compounds of type AG5 can be reacted with aryl and heteroaryl bromides and iodides under similar conditions to those described above to form compounds of structure XL.




embedded image


Compounds of the present invention, represented by structure XLI can be prepared as described in scheme AH below.


3-iodo- or bromoindoles of structure AH1 can be reacted with alkenes AH2 in the presence of a palladium catalyst (commonly referred to as the Heck reaction) to give compounds of type XLI. The coupling reactions can be carried out by methods known to those skilled in the art. The choice of catalyst and solvents are similar to those described in Scheme AG.




embedded image


Compounds of formula I, represented by structure XLII can be prepared as shown in Scheme AI.


3-Iodo- or bromoindoles of structure AI1 can be reacted with acetylenes AI2 in the presence of a palladium catalyst (commonly referred to as the Sonagashira reaction) to give compounds of type XLII. The coupling reactions can be carried out by methods known to those skilled in the art. A typical set of reaction conditions includes reacting the indole of structure AI1 with an acetylene compound AI2 in the presence of a source of palladium, a copper co-catalyst and an amine source and carrying out the reaction at a temperature range of ambient to 150° C.




embedded image


Compounds of the present invention, represented by structure XLIII and XLIV can be prepared as described in scheme AJ below.


Nitroanilines of structure AJ1 can be converted to indoles of structure XLIII by condensation and cyclization with nitriles of structure AJ2. The reaction can be carried out in a suitable organic solvent, e.g., DMF or dioxane. Treatment of compounds of structure XLIII with a base followed by reaction with a reactive functional group R9 containing a suitable leaving group L can give the compounds of formula XLIV.




embedded image


Compounds of formula I, represented by structure XLV-XLVIII can be prepared as shown in Scheme AK.


2-aminoindoles of structure XLV can be alkylated with a reactive functional group R15 containing a suitable leaving group L in the presence of a base, e.g., sodium hydride or potassium carbonate in a suitable organic solvent to give compounds of structure XLVI. A second alkylation utilizing a reactive functional group R′15 containing a suitable leaving group L similarly can give compounds of structure XLVII.


Acylation of compounds of structure XLV with acyl chlorides of structure AK1 can give compounds of structure XLVIII. The reaction is typically carried out in the presence of an organic base, e.g., a trialkylamine or an inorganic base, e.g., potassium carbonate in a suitable organic solvent.




embedded image


Compounds of the present invention, represented by structure XLIX can be prepared as described in scheme AL below.


Indole-3-carboxylic acids of structure AL1 can be activated to give compounds of structure AL2. Compounds of structure AL2 can represent, for example, acyl halides or carboxylic acids activated with a reagent such as dicyclohexyl carbodiimide or diisopropyl carbodiimide. Reaction of compounds of structure AL2 with hydroxyamidines of structure AL3 can give O-acylhydroxyamidines AL4. Hydroxyamidines may be obtained commercially or by treatment of nitrile compounds with hydroxylamine. Heating compounds of structure AL4 in a non-reactive organic solvent, e.g., toluene, dichloroethane or dioxane in a temperature range of 30° C. to 150° C. can give compounds of structure XLIX.




embedded image


C. Methods of the Invention


The methods of the invention generally comprise administering a therapeutically effective amount of one or more compound of the present invention to a subject in need of treatment for HCV infection. In a preferred embodiment, a therapeutically effective amount of a composition comprising a compound of Formula I as described herein is administered to a subject in need of treatment. In another preferred embodiment, in another preferred embodiment, a compound or a composition used in the methods of the present invention includes a compound of Formula I as described herein wherein the compound of Formula I is not




embedded image



Compound 1.


The compound(s) of the present invention may be administered to the subject via any drug delivery route known in the art. Specific exemplary administration routes include oral, ocular, rectal, buccal, topical, nasal, ophthalmic, subcutaneous, intramuscular, intraveneous (bolus and infusion), intracerebral, transdermal, and pulmonary. Individuals infected with HCV can be treated with the compounds of the present invention to prevent or reduce further replication of HCV.


The term therapeutically effective amount, as used herein, refers to an amount of a compound of the present invention effective to inhibit HCV translation, thereby effectively treating or ameliorating the HCV infection. The effect of the compound can be determined by analyzing (1) the presence of HCV-RNA; (2) the presence of anti-HCV antibodies; (3) the level of serum alanine amino transferase (ALT) and aspartate aminotransferase (AST) (ALT and AST are elevated in patients chronically infected with HCV); and (4) hepatocellular damage. The precise effective amount for a subject will depend upon the subject's body weight, size and health. Therapeutically effective amounts for a given patient can be determined by routine experimentation that is within the skill and judgment of the clinician.


For any compound, the therapeutically effective amount can be estimated initially either in cell culture assays or in relevant animal models, such as marmosets and tarmarins. The animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans. Therapeutic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., ED50 (the dose therapeutically effective in 50% of the population) and LD50 (the dose lethal to 50% of the population). The dose ratio between therapeutic and toxic effects is the therapeutic index, and it can be expressed as the ratio, ED50/LD50. Pharmaceutical compositions that exhibit large therapeutic indices are preferred. The dosage contained in such compositions is preferably within a range of circulating concentrations that include an ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration.


More specifically, the concentration-biological effect relationships observed with regard to the compound(s) of the present invention indicate an initial target plasma concentration ranging from approximately 0.1 μg/ml to approximately 100 μg/mL, preferably from approximately 1 μg/mL to approximately 50 μg/mL, more preferably from approximately 5 μg/mL to approximately 50 μg/mL, even more preferably from approximately 10 μg/mL to approximately 25 μg/mL. To achieve such plasma concentrations, the compounds of the invention may be administered at doses that vary from 0.1 μg to 100,000 mg, depending upon the route of administration. Guidance as to particular dosages and methods of delivery is provided in the literature and is generally available to practitioners in the art. In general the dose will be in the range of about 1 mg/day to about 10 g/day, or about 0.1 g to about 3 g/day, or about 0.3 g to about 3 g/day, or about 0.5 g to about 2 g/day, in single, divided, or continuous doses for a patient weighing between about 40 to about 100 kg (which dose may be adjusted for patients above or below this weight range, particularly children under 40 kg).


The exact dosage will be determined by the practitioner, in light of factors related to the subject that requires treatment. Dosage and administration are adjusted to provide sufficient levels of the active agent(s) or to maintain the desired effect. Factors which may be taken into account include the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. Long-acting pharmaceutical compositions may be administered every 3 to 4 days, every week, or once every two weeks depending on half-life and clearance rate of the particular formulation.


D. Metabolites of the Compounds of the Invention


Also falling within the scope of the present invention are the in vivo metabolic products of the compounds described herein. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, esterification and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising contacting a compound of this invention with a mammalian tissue or a mammal for a period of time sufficient to yield a metabolic product thereof. Such products typically are identified by preparing a radio-labeled (e.g. C14 or H3) compound of the invention, administering it in a detectable dose (e.g., greater than about 0.5 mg/kg) to a mammal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours), and isolating its conversion products from urine, blood or other biological samples. These products are easily isolated since they are labeled (others are isolated by the use of antibodies capable of binding epitopes surviving in the metabolite). The metabolite structures are determined in conventional fashion, e.g., by MS or NMR analysis. In general, analysis of metabolites may be done in the same way as conventional drug metabolism studies well-known to those skilled in the art. The conversion products, so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the invention even if they possess no biological activity of their own.


E. Pharmaceutical Compositions of the Invention


While it is possible for the compounds of the present invention to be administered neat, it may be preferable to formulate the compounds as pharmaceutical compositions. As such, in yet another aspect of the invention, pharmaceutical compositions useful in the methods of the invention are provided. The pharmaceutical compositions of the invention may be formulated with pharmaceutically acceptable excipients such as carriers, solvents, stabilizers, adjuvants, diluents, etc., depending upon the particular mode of administration and dosage form. The pharmaceutical compositions should generally be formulated to achieve a physiologically compatible pH, and may range from a pH of about 3 to a pH of about 11, preferably about pH 3 to about pH 7, depending on the formulation and route of administration. In alternative embodiments, it may be preferred that the pH is adjusted to a range from about pH 5.0 to about pH 8.0.


More particularly, the pharmaceutical compositions of the invention comprise a therapeutically or prophylactically effective amount of one or more compound of the present invention, together with one or more pharmaceutically acceptable excipients. A therapeutically or prophylactically effective amount of a compound of the present invention includes a viral inhibitory amount of said compound or an amount effective for affecting viral IRES activity. By “viral inhibitory amount” it is meant an amount sufficient to inhibit viral replication or infectivity. By “an amount effective for affecting viral IRES activity” it is meant an amount sufficient to inhibit viral IRES mediated initiation and/or translation. Optionally, the pharmaceutical compositions of the invention may comprise a combination of compounds of the present invention, or may include a second active ingredient useful in the treatment of viral infections, such as anti-viral agents that include, but are not limited to: pegylated interferon, including by way of non-limiting example pegylated α-interferon; un-pegylated interferon, including by way of non-limiting example, un-pegylated α-interferon; ribavirin or prodrugs or derivatives thereof; a glucosidase inhibitor; protease inhibitors; polyermase inhibitors; p7 inhibitors; entry inhibitors, including fusion inhibitors such as Fuzeon (Trimeris); helicase inhibitors; a Toll-like receptor agonist, a caspase inhibitor, anti-fibrotics; drugs that target IMPDH (inosine monophosphate dehydrogenase inhibitors), such as Merimepadib™ (Vertex Pharmaceuticals Inc.); synthetic thymosin alpha 1 (ZADAXIN™, SciClone Pharmaceuticals Inc.); a glycosidase inhibitor; therapeutic viral vaccines, such as those produced by Chiron and Immunogenics; and immunomodulators, such as histamine.


Formulations of the present invention, e.g., for parenteral or oral administration, are most typically solids, liquid solutions, emulsions or suspensions, while inhaleable formulations for pulmonary administration are generally liquids or powders, with powder formulations being generally preferred. A preferred pharmaceutical composition of the invention may also be formulated as a lyophilized solid that is reconstituted with a physiologically compatible solvent prior to administration. Alternative pharmaceutical compositions of the invention may be formulated as syrups, creams, ointments, tablets, and the like.


The term “pharmaceutically acceptable excipient” refers to an excipient for administration of a pharmaceutical agent, such as the compounds of the present invention. The term refers to any pharmaceutical excipient that may be administered without undue toxicity. Pharmaceutically acceptable excipients are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there exists a wide variety of suitable formulations of pharmaceutical compositions of the present invention (see, e.g., Remington's Pharmaceutical Sciences).


Suitable excipients may be carrier molecules that include large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive virus particles. Other exemplary excipients include antioxidants such as ascorbic acid; chelating agents such as EDTA; carbohydrates such as dextrin, hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid; liquids such as oils, water, saline, glycerol and ethanol; wetting or emulsifying agents; pH buffering substances; and the like. Liposomes are also included within the definition of pharmaceutically acceptable excipients.


The pharmaceutical compositions of the invention may be formulated in any form suitable for the intended method of administration. When intended for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, non-aqueous solutions, dispersible powders or granules (including micronized particles or nanoparticles), emulsions, hard or soft capsules, syrups or elixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions, and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation.


Pharmaceutically acceptable excipients particularly suitable for use in conjunction with tablets include, for example, inert diluents, such as celluloses, calcium or sodium carbonate, lactose, calcium or sodium phosphate; disintegrating agents, such as croscarmellose sodium, cross-linked povidone, maize starch, or alginic acid; binding agents, such as povidone, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.


Formulations for oral use may be also presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluent, for example celluloses, lactose, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with non-aqueous or oil medium, such as glycerin, propylene glycol, polyethylene glycol, peanut oil, liquid paraffin or olive oil.


In another embodiment, pharmaceutical compositions of the invention may be formulated as suspensions comprising a compound of the present invention in an admixture with at least one pharmaceutically acceptable excipient suitable for the manufacture of a suspension. In yet another embodiment, pharmaceutical compositions of the invention may be formulated as dispersible powders and granules suitable for preparation of a suspension by the addition of suitable excipients.


Excipients suitable for use in connection with suspensions include suspending agents, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth, gum acacia, dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycethanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate); and thickening agents, such as carbomer, beeswax, hard paraffin or cetyl alcohol. The suspensions may also contain one or more preservatives such as acetic acid, methyl and/or n-propyl p-hydroxy-benzoate; one or more coloring agents; one or more flavoring agents; and one or more sweetening agents such as sucrose or saccharin.


The pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these. Suitable emulsifying agents include naturally-occurring gums, such as gum acacia and gum tragacanth; naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids; hexitol anhydrides, such as sorbitan monooleate; and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate. The emulsion may also contain sweetening and flavoring agents. Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.


Additionally, the pharmaceutical compositions of the invention may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous emulsion or oleaginous suspension. This emulsion or suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,2-propane-diol. The sterile injectable preparation may also be prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile fixed oils may be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or di-glycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables.


Generally, the compounds of the present invention useful in the methods of the present invention are substantially insoluble in water and are sparingly soluble in most pharmaceutically acceptable protic solvents and in vegetable oils. However, the compounds are generally soluble in medium-chain fatty acids (e.g., caprylic and capric acids) or triglycerides and have high solubility in propylene glycol esters of medium-chain fatty acids. Also contemplated in the invention are compounds which have been modified by substitutions or additions of chemical or biochemical moieties which make them more suitable for delivery (e.g., increase solubility, bioactivity, palatability, decrease adverse reactions, etc.), for example by esterification, glycosylation, PEGylation, etc.


In a preferred embodiment, the compounds of the present invention may be formulated for oral administration in a lipid-based formulation suitable for low solubility compounds. Lipid-based formulations can generally enhance the oral bioavailability of such compounds. As such, a preferred pharmaceutical composition of the invention comprises a therapeutically or prophylactically effective amount of a compound of the present invention, together with at least one pharmaceutically acceptable excipient selected from the group consisting of: medium chain fatty acids or propylene glycol esters thereof (e.g., propylene glycol esters of edible fatty acids such as caprylic and capric fatty acids) and pharmaceutically acceptable surfactants such as polyoxyl 40 hydrogenated castor oil.


In an alternative preferred embodiment, cyclodextrins may be added as aqueous solubility enhancers. Preferred cyclodextrins include hydroxypropyl, hydroxyethyl, glucosyl, maltosyl and maltotriosyl derivatives of α-, β-, and γ-cyclodextrin. A particularly preferred cyclodextrin solubility enhancer is hydroxypropyl-β-cyclodextrin (HPBC), which may be added to any of the above-described compositions to further improve the aqueous solubility characteristics of the compounds of the present invention. In one embodiment, the composition comprises 0.1% to 20% hydroxypropyl-β-cyclodextrin, more preferably 1% to 15% hydroxypropyl-β-cyclodextrin, and even more preferably from 2.5% to 10% hydroxypropyl-β-cyclodextrin. The amount of solubility enhancer employed will depend on the amount of the compound of the present invention in the composition.


F. Combination Therapy


It is also possible to combine any compound of the present invention with one or more other active ingredients useful in the treatment of HCV infection, including compounds, in a unitary dosage form, or in separate dosage forms intended for simultaneous or sequential administration to a patient in need of treatment. When administered sequentially, the combination may be administered in two or more administrations. In an alternative embodiment, it is possible to administer one or more compounds of the present invention and one or more additional active ingredients by different routes.


The skilled artisan will recognize that a variety of active ingredients may be administered in combination with the compounds of the present invention that may act to augment or synergistically enhance the viral inhibiting activity of the compounds of the invention. Such active ingredients include anti-HCV agents. Anti-HCV agents include agents that target the virus as well as agents that have an immunomodulatory effect. For example, anti-HCV agents include, but are not limited to, interferon, including, for example without limitation, IFN-α, ribavirin or prodrugs or derivatives thereof; a glucosidase inhibitor, protease inhibitors, polymerase inhibitors, helicase inhibitors, a Toll-like receptor agonist, a caspase inhibitor and a glycosidase inhibitor. Furthermore, the compounds of the invention may also be administered in combination with other compounds that affect IRES activity known to one of skill in the art.


According to the methods of the invention, the combination of active ingredients may be: (1) co-formulated and administered or delivered simultaneously in a combined formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by any other combination therapy regimen known in the art. When delivered in alternation therapy, the methods of the invention may comprise administering or delivering the active ingredients sequentially, e.g., in separate solution, emulsion, suspension, tablets, pills or capsules, or by different injections in separate syringes. In general, during alternation therapy, an effective dosage of each active ingredient is administered sequentially, i.e., serially, whereas in simultaneous therapy, effective dosages of two or more active ingredients are administered together. Various sequences of intermittent combination therapy may also be used.


To assist in understanding the present invention, the following Examples are included. The experiments relating to this invention should not, of course, be construed as specifically limiting the invention and such variations of the invention, now known or later developed, which would be within the purview of one skilled in the art are considered to fall within the scope of the invention as described herein and hereinafter claimed.


EXAMPLES

The present invention is described in more detail with reference to the following non-limiting examples, which are offered to more fully illustrate the invention, but are not to be construed as limiting the scope thereof. The examples illustrate the preparation of certain compounds of the invention, and the testing of these compounds in vitro and/or in vivo. Those of skill in the art will understand that the techniques described in these examples represent techniques described by the inventors to function well in the practice of the invention, and as such constitute preferred modes for the practice thereof. However, it should be appreciated that those of skill in the art should in light of the present disclosure, appreciate that many changes can be made in the specific methods that are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.


Example 1
Preparation of Compounds of the Invention
Example 1A
Preparation of 1-ethyl-6-methoxy-1H-indole-3-carbonitrile (compound 5)



embedded image


Step A: A solution of 6-methoxyindole (10.0 g, 68.0 mmol) in DMF (120 mL) was cooled to 0° C. and treated with chlorosulfonyl isocyanate (7.72 mL, 88.4 mmol). After the addition, the reaction mixture was stirred at this temperature for 1 h. The dark solution was poured into ice water (600 mL) and the light brown solid was collected by filtration, washed with additional H2O and dried to afford 9.9 g (85%) of 6-methoxy-1H-indole-3-carbonitrile as a light brown solid.


Step B: To a solution of 6-methoxy-1H-indole-3-carbonitrile (9.9 g, 57.6 mmol) in DMF (150 mL) was added NaH (60% dispersion in mineral oil, 3.45 g, 86.3 mmol). The reaction mixture was stirred for 15 min and then ethyl iodide (5.53 mL, 69.1 mmol) was added and the mixture was stirred at room temperature overnight. The reaction mixture was then diluted with H2O and extracted with EtOAc (2×). The organic phases were washed with H2O (3×) and saturated NaCl and then dried and concentrated to a semi-solid. The crude product was purified via column chromatography on silica gel (200 g) using CH2Cl2/hexanes (50-100%) as eluent to yield 6-methoxy-1-ethyl-1H-indole-3-carbonitrile as a tan solid.


Utilizing steps A and B above and substituting different indoles and alkyl halides gave the following compounds: Compounds 43, 45, 51, 52, 108, 109, 115, 118, 120, 123, 126, 179 and 714.


Example 1B
Preparation of 6-ethoxy-1-ethyl-1H-indole-3-carbonitrile (compound 9)



embedded image


Step A: To a solution of 1-ethyl-6-methoxy-1H-indole-3-carbonitrile (2.85 g, 14.2 mmol), prepared by example 1A, step B, in CH2Cl2 (40 mL) was added a 1M solution of BBr3 in CH2Cl2 (28.5 mL, 28.5 mmol) at 0° C. The mixture was allowed to warm to room temperature and kept for 2.5 h. The dark reaction mixture was then poured onto ice and sufficient 1M NaOH was added until the pH was 8-9. The product was extracted with CH2Cl2 (3×) and the combined organic phases were washed with saturated NaHCO3 , H2O and saturated NaCl. After drying over MgSO4, the solution was concentrated and the product was purified by chromatography (EtOAc/CH2Cl2, 0-10%) to afford 2.15 g (82%) of 6-hydoxy-1-ethyl-1H-indole-3-carbonitrile as a yellow solid.


Step B: To a solution 6-hydoxy-1-ethyl-1H-indole-3-carbonitrile (80 mg, 0.43 mmol) in 5 mL of methyl ethyl ketone was added anhydrous K2CO3 (71 mg, 0.52 mmol) and iodomethane (0.05 mL, 0.60 mmol). After stirring overnight at reflux, the reaction mixture was cooled, diluted with H2O and extracted with EtOAc (3×). The combined organic phases were dried and concentrated. Flash chromatography (CH2Cl2) gave 94 mg (100%) of 6-ethoxy-1-ethyl-1H-indole-3-carbonitrile as a white wax.


In similar fashion, following steps A and B, above, the following compounds were also prepared: compounds 6, 10, 11, 12 and 24


Example 1C
Preparation of 5-(4-methoxyphenyl)-5H-[1,3]dioxolo[4,5-f]indole-7-carbonitrile (compound 44)



embedded image


A mixture of p-iodoanisole (85 mg, 0.36 mmol), anhydrous K3PO4 (102 mg, 0.48 mmol), CuI (4.6 mg, 0.024 mmol) and N,N′-Dimethyl cyclohexane-1,2-diamine (14 mg, 0.096 mmol) was added to 5H-[1,3]dioxolo[4,5-f]indole-7-carbonitrile (45 mg, 0.24 mmol), prepared as described by the method of example 1A, step A, in anhydrous toluene (0.4 mL). After heating at reflux for 24 h, the solvent was evaporated under vacuum. The residue was dissolved with CH2Cl2 (5 mL) and the mixture was filtered. The filtrate was concentrated to afford crude product, which was purified by silica gel chromatography using EtOAc/petroleum ether (1:4) as eluent to yield 5-(4-methoxyphenyl)-5H-[1,3]dioxolo[4,5-f]indole-7-carbonitrile.


Utilizing the procedure above and substituting different aryl iodides gave the following compounds: compounds 4, 8, 102, 103, 111, 112, 117, 119, 124, 125, 127, 154.


Example 1D
Preparation of 1-ethyl-6-(pyrazin-2-yloxy)-1H-indole-3-carbonitrile (compound 13)



embedded image


To a solution of 1-ethyl-6-hydroxy-1H-indole-3-carbonitrile (60 mg, 0.32 mmol) prepared as described in example 1A, step A, in DMF (5 mL) was added K2CO3 (55 mg, 0.40 mmol) and 2-chloropyridazine (45 mg, 0.40 mmol). The mixture was heated at 110° C. for 18 h. After cooling to room temperature, the reaction mixture was diluted with H2O and extracted with EtOAc (3×). The combined organic phases were washed with H2O and saturated NaCl, dried and concentrated. The product was isolated by chromatography (EtOAc/CH2Cl2, 1-3%) over silica gel to afford 76 mg (96%) of the title compound, 1-ethyl-6-(pyrazin-2-yloxy)-1H-indole-3-carbonitrile, as an off-white solid.


Example 1E
Preparation of 3-cyano-1-ethyl-1H-indole-6-carboxylic acid phenylamide (compound 15)



embedded image


Step A: A solution of methyl 3-cyano-1-ethyl-1H-indole-6-carboxylate (1.60 g, 7.02 mmol), prepared by the method described in example 1A from methyl 1H-indole-6-carboxylate, in THF (35 mL) was treated with 1N NaOH (7.7 mL, 7.7 mmol) and heated at reflux for 2.5 h. After cooling to room temperature, most of the THF was removed and the solution was diluted with H2O and extracted with ether (2×). The ether extracts were discarded. The aqueous phase was then acidified with 6N HCl to pH 2 and then extracted with EtOAc (3×). The EtOAc layers were combined, washed with saturated NaCl and then dried and concentrated to afford 1.43 g (95%) of 3-cyano-1-ethyl-1H-indole-6-carboxylic acid as a white solid.


Step B: A suspension of 3-cyano-1-ethyl-1H-indole-6-carboxylic acid (0.42 g, 1.96 mmol) in CH2Cl2 (15 mL) was cooled to 0° C. The suspension was treated with DMF (2 drops) and then oxalyl chloride (0.34 mL, 3.92 mmol) was added via syringe during 2 minutes after which the ice bath was removed and the reaction mixture was allowed to warm to ambient temperature during 1.5 h during which time the reaction became a yellow solution. The solution was then concentrated in vacuo to afford 0.46 g (quantitative yield) of 3-cyano-1-ethyl-1H-indole-6-carbonyl chloride as a yellow solid.


Step C: A suspension of 3-cyano-1-ethyl-1H-indole-6-carbonyl chloride (70 mg, 0.30 mmol) in THF (5 mL) was cooled to 0° C. and treated with aniline (0.08 mL, 0.90 mmol). After the addition the reaction was warmed to ambient temperature and after stirring for an additional 16 hours, the reaction mixture was diluted with H2O and extracted with EtOAc (2×). The combined organic phases were washed with saturated NaCl and then dried and concentrated to afford the product. Chromatography (EtOAc/CH2Cl2, 2/98) over silica gel gave 44 mg (51%) of 3-cyano-1-ethyl-1H-indole-6-carboxylic acid phenylamide.


Utilizing essentially the procedure above gave the following compound: Compound 89.


Example 1F
Preparation of t-butyl (3-cyano-1-ethyl-1H-indol-6-yl)-carbamate (compound 16)



embedded image


A solution of 3-cyano-1-ethyl-1H-indole-6-carboxylic acid (0.60 g, 2.80 mmol) from Example 1E, step A, in t-butanol (20 mL) was treated with Et3N (0.46 mL, 3.36 mmol) and diphenylphosphoryl azide (0.73 mL, 3.36 mmol) and then heated at reflux for 4 h. After cooling to room temperature, most of the t-butanol was removed in vacuo to give an oil, which was then dissolved in EtOAc. After washing with H2O, the organic phase was back-extracted with EtOAc and the organic layers were combined and washed sequentially with additional H2O, saturated NaHCO3 and saturated NaCl. The organic phase was dried, concentrated and the resulting crude product was purified by chromatography over silica gel using EtOAc/CH2Cl2 (0-1%) to afford 0.52 g (65%) of t-butyl (3-cyano-1-ethyl-1H-indol-6-yl)-carbamate as a white solid.


The following compound was made in similar fashion: compound 90.


Example 1Ga
Preparation of 2-(4-aminophenyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile via Suzuki route (compound 55)



embedded image


Step A: A 2M solution of lithium diisopropyl amide in THF/hexanes (Acros) (3.9 mL, 7.8 mmol) was diluted with THF (5 mL) in a flame-dried flask. After cooling the reaction to −30° C., a solution of 1-ethyl-6-methoxy-1H-indole-3-carbonitrile (1.30 g, 6.5 mmol) in THF (10 mL) was added dropwise during 10 min, maintaining the temperature at −30° C. After stirring for an additional 30 min at this temperature, a solution of iodine (2.31 g, 9.1 mmol) in THF (5 mL) was added during 10 min. After the addition, the reaction was warmed to ambient temperature during 1 h. The reaction was then diluted with ice-H2O and extracted with EtOAc (2×). The combined organic phases were washed with 1M sodium thiosulfate and saturated NaCl and then concentrated to a brown solid. Chromatography (CH2Cl2/hexanes, 1/1) over silica gel gave 1.31 g (62%) of 1-ethyl-2-iodo-6-methoxy-1H-indole-3-carbonitrile as an off-white solid.


Step B: A mixture of 1-ethyl-2-iodo-6-methoxy-1H-indole-3-carbonitrile (1.25 g, 3.83 mmol), 4-(4,4,5,5-tetramethyl)-1,3-2-dioxaboralanyl-2-yl-aniline (0.96 g, 4.90 mmol), CsF (1.46 g, 9.58 mmol) and Pd(PPh3)2Cl2 (110 mg, 0.15 mmol) in DME (20 mL) was added to a flask and alternatively evacuated and flushed with N2. The reaction was then heated at reflux for 24 h and then cooled to room temperature. The reaction mixture was diluted with H2O and extracted with EtOAc (2×). The combined organic phases were washed with H2O and saturated NaCl and then dried over MgSO4 and concentrated. The crude reaction mix purified by flash chromatographt on silica gel using EtOAc/CH2Cl2 (5/95) as eluent to afford 765 mg (69%) of 2-(4-aminophenyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile as a yellow solid.


Utilizing essentially the same procedure described above and substituting different boronic acids gave the following compounds: compounds 19, 20, 21, 22, 53, 63, 70, 71, 74, 76, 77, 79, 80, 100, 110, 229, 239, 240, 247, 250, 254, 255, 256, 257, 258, 259, 260, 281, 282, 283, 284, 286, 335, 336, 337, 338, 339, 347, 348, 426, 427, 428, 429, 476, 543, 578, 758.


Example 1Gb
Preparation of 2-(4-aminophenyl)-1-butyl-6-methoxy-1H-indole-3-carbonitrile via alternative Suzuki route



embedded image


To a solution of (i-Pr)2NH (1.35 mL, 9.65 mmol) in THF (30 mL) cooled to −78° C. was added n-BuLi (3.7 mL, 2.5M in hexanes, 9.21 mmol) in one portion. The acetone/dry ice bath was exchanged for ice/water bath and the solution was stirred further for 40 min. The solution was cooled to −78° C. and solution of 1-butyl-6-methoxy-1H-indole-3-carbonitrile, prepared as in example 1A (2.0 g, 8.77 mmol) in THF (10 mL) was added dropwise. This solution was stirred for 15 min at −78° C., following by 20 min at −20° C. Trimethyl borate (1.0 mL, 8.77 mmol) was added, the reaction mixture was stirred for 15 min at −20° C. after which the cooling bath was removed and this solution was stirred further at room temperature for 1 h. A solution of K3PO4 was added (11.7 mL, 3M aqueous solution, 35.1 mmol) followed by a solution of 4-iodoaniline (2.5 g, 11.40 mmol) and PdCl2dppf catalyst (640 mg, 0.88 mmol) in DMF (40 mL, plus a 5 mL rinse). The reaction mixture was stirred overnight (ca. 18 h.) and then water (80 mL) was added and the product was extracted with EtOAc (3×50 mL). The combined organic fractions were dried over MgSO4, filtered and concentrated under reduced pressure. The crude product was purified via flush chromatography on silica gel (5→60% EtOAc/Hexanes as eluant) to afford the desired 2-(4-aminophenyl)-1-butyl-6-methoxy-1H-indole-3-carbonitrile as a tan solid (2.4 g, 86% yield).


The following compounds were prepared in similar fashion utilizing other indole and aryl and hereroaryl bromides and iodides: Compounds 656, 659, 660, 661, 682, 683, 712, 731, 732, 733, 806, 807, 808, 809, 810, 811, 812, 813, 814, 827.


Example 1Gc
Preparation of 2-(4-aminophenyl)-6-methoxy-1-propyl-1H-indole-3-carbonitrile via Negishi route.



embedded image


A nitrogen-purged flask fitted with a septum and a nitrogen needle was charged with dry THF (all additions performed by syringe) (20 mL). Diisopropylamine (Aldrich Sure-Seal, 2.00 mL, 14.3 mmol) was added, and the solution was cooled to 0° C. n-Butyllithium (8.50 mL of 1.6 M solution in hexane, 13.6 mmol) was added slowly. The flask was allowed to warm to room temperature briefly, and then was cooled to −78° C. A concentrated THF solution of 6-methoxy-1-propyl-1H-indole-3-carbonitrile (2.77 g, 12.9 mmol; prepared analogously to compound 5 of Example 1A) was added slowly, and the resulting solution was maintained at −78° C. for 30 min. The flask was then transferred to a water-ice bath and allowed to come to 0° C. for about 15 minutes. The solution was once again cooled to −78° C., and ZnCl2 (0.5 M solution in THF, 27.0 mL, 13.5 mmol) was slowly added. A precipitate was observed at this point, which may have been the bis(indole)zinc compound, but the solution became homogeneous when the entire volume of zinc chloride solution was added. After about 10 minutes, the solution was allowed to come to room temperature, and a THF solution (5 mL) of 4-iodoaniline (3.47 g, 15.8 mmol) and triphenylphosphine (338 mg, 1.29 mmol) was added. The septum was removed, and solid Pd2(dba)3 (295 mg, 0.322 mmol) was added. A reflux condenser was fitted to the flask, and the solution was degassed by three successive cycles of vacuum pumping/N2 purging. The solution was then heated to reflux overnight. After cooling to room temperature, the solution was poured into 4 volumes of water, and 4 volumes of ethyl acetate were added. The resulting mixture was vigorously stirred for 30 minutes, then filtered through celite (with ethyl acetate washing) to remove solid Zn-and Pd-containing material. The phases were separated, and the aqueous phase was extracted with more ethyl acetate. The organic phases were washed in sequence with saturated brine, combined, dried over anhydrous sodium sulfate, filtered and evaporated. A solid precipitate formed at this point, which was sufficiently pure product and was collected by trituration with ether and filtration. The remaining material was purified by column chromatography (eluting 1:2 ethyl acetate-hexane on silica gel 60). Total yield of the product, 2-(4-amino-phenyl)-6-methoxy-1-propyl-1H-indole-3-carbonitrile, was 2.75 g (8.99 mmol, 70%).


The following compounds were made using essentially the same procedure and substituting other aryl or heteroaryl iodides or bromides: Compounds 393, 408, 430, 431, 436, 437, 438, 459, 460, 461, 462, 483, 484, 632, 633, 634, 635, 636, 650, 651.


Example 1Gd
Preparation of 1-ethyl-2-(3-hydroxyphenyl)-6-methoxy-1H-indole-3-carbonitrile (Compound 288)



embedded image


Step A: A solution of THF (60 mL) and diisopropylamine (5.5 mL, 39 mmol) was cooled to −78° C. n-Butyllithium (14.5 mL, 2.5M in hexanes, 36.2 mmol) was added dropwise over 5 minutes. The LDA mixture was stirred at −78° C. for 10 minutes, and then at 0° C. for 20 minutes. The solution was re-cooled to −78° C. 1-ethyl-6-methoxy-1H-indole-3-carbonitrile (5.0 g, 25 mmol), prepared as in example 1A, was taken up in THF (30 mL) and added dropwise to the LDA mixture over 15 minutes. The reaction was stirred at −78° C. for 10 minutes, and at 0° C. for 30 minutes. Once again, the reaction mixture was cooled to −78° C. Tributyltin iodide (10 mL, 35 mmol) was added dropwise. This was stirred at −78° C. for 15 minutes, and then at 0° C. for 30 minutes. The reaction mixture was absorbed onto silica gel and concentrated. Purification by chromatography (CH2Cl2) yielded 1-ethyl-6-methoxy-2-tributylstannanyl-1H-indole-3-carbonitrile (12.05 g, 98%).


Step B: 1-Ethyl-6-methoxy-2-tributylstannanyl-1H-indole-3-carbonitrile (1.0 g, 2.05 mmol), prepared in step A, was combined with 3-iodophenol (474 mg, 2.15 mmol), Pd(PPh3)2Cl2 (67 mg, 0.102 mmol), CuI (75 mg, 0.39 mmol) and THF (4.0 mL). This mixture was heated at 65° C. overnight. The reaction mixture was diluted in EtOAc, and was filtered through celite. The filtrate was concentrated and the residue was purified by silica gel chromatography (4:1, CH2Cl2/EtOAc) to yield crude product. Ether trituration yielded 1-ethyl-2-(3-hydroxy-phenyl)-6-methoxy-1H-indole-3-carbonitrile (430 mg, 72%) as a yellow-white solid.


The following compounds were prepared similarly as above, using other commercially available iodides and bromides, or using iodides derived from a one step amidation of p-iodophenylsulfonyl chloride: Compounds 275, 276, 277, 278, 331, 363, 364, 373, 374, 375, 474, 475, 678.


Example 1Ge
Preparation of ethanesulfonic acid [4-(3-cyano-6-difluoromethoxy-1-ethyl-1H-indol-2-yl)-phenyl]-amide via Heck route (compound 519)



embedded image


Step A: A solution of 6-difluoromethoxy-1-ethyl-1H-indole (402.8 mg, 2.04 mmol), ethanesulfonic acid (4-iodo-phenyl)-amide (712.1 mg, 2.29 mmol), cesium carbonate (733.2 mg, 3.82 mmol), triphenylphosphine (33.1 mg, 0.13 mmol) and palladium acetate (5.7 mg, 0.025 mmol) in DMA (5 ml) was heated to 135° C. for 48 h. The reaction mixture was diluted with water and extracted with EtOAc (2×10 mL). The combined organic phases were washed with brine, dried over MgSO4, and then concentrated. The residue was purified via column chromatogrphy on silica gel (25 g) using EtOAc/Hexanes (10-20%) as eluent to afford 298.2 mg (37.1% yield) of ethanesulfonic acid [4-(6-difluoromethoxy-1-ethyl-1H-iodo-2-yl)-phenyl]-amide, compound 516, as a light brown solid.


Step B: Following the procedure 1A, step A, ethanesulfonic acid [4-(6-difluoromethoxy-1-ethyl-1H-iodo-2-yl)-phenyl]-amide was converted to ethanesulfonic acid [4-(3-cyano-6-difluoromethoxy-1-ethyl-1H-indol-2-yl)-phenyl]-amide, compound 519.


Following steps A and B above, the following compounds were prepared in similar fashion: Compounds 343, 344, 345, 346, 409, 410, 411, 412, 413, 414, 415, 416, 417, 418, 419, 463, 464, 465, 466, 467, 468, 469, 470, 471, 472, 473, 515, 517, 518, 520, 521, 522, 523, 524, 575, 577, 579, 580, 611, 612, 613, 614


Example 1H
Preparation of 1-ethyl-2-(4-fluorophenylethynyl)-6-methoxy-1H-indole-3-carbonitrile (compound 67)



embedded image


A mixture of 1-ethyl-2-iodo-6-methoxy-1H-indole-3-carbonitrile (150 mg, 0.46 mmol), prepared as described in example 1Ga, step A, 4-fluorophenylacetylene (80 mg, 0.0.69 mmol), bis(triphenylphosphine) palladium (II) dichloride (6 mg, 0.009 mmol) and CuI (4 mg, 0.018 mmol) was added to a sealable tube and alternatively evacuated and flushed with N2. To the tube was then added DMF (4 mL) and Et3N (0.25 mL, 1.84 mmol) and the reaction was heated at 80° C. for 20 h and then cooled to room temperature. The reaction mixture was diluted with H2O and extracted with EtOAc (2×). The combined organic phases were washed with H2O (3×) and saturated NaCl and then dried over MgSO4 and concentrated. The crude reaction mix was absorbed on silica gel (0.6 g) and chromatographed over silica gel using EtOAc/hexanes (10-20%) as eluent to afford 120 mg (82%) of 1-ethyl-2-(4-fluorophenylethynyl)-6-methoxy-1H-indole-3-carbonitrile as a yellow solid.


Utilizing essentially the same procedure described above and substituting different acetylene derivatives gave the following compounds: compounds 64, 65, 66, 68, 69, 91, 92, 93, 94, 95, 96, 133, 134, 135, 136, 137, 143, 144, 145, 146, 147, 148, 149, 150, 151, 158, 159, 160, 161, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 184, 185, 186, 187, 188, 196, 197, 198, 199, 200, 201, 202, 223, 230, 231, 232, 233, 234, 235, 236, 237, 238.


Example 1I
Preparation of 1-ethyl-3-(5-ethyl-[1,2,4]oxadiazol-3-yl)-6-methoxy-1H-indole (compound 28)



embedded image


Step A: A solution of 1-ethyl-6-methoxy-1H-indole-3-carbonitrile (1.00 g, 5.00 mmol) in MeOH (10 mL) was treated with a 50% aqueous solution of hydroxylamine (0.38 mL, 6.25 mmol) and heated at reflux for 18 h. After cooling to room temperature, the heterogeneous mixture was filtered to afford 525 mg of desired product as a tan solid. The filtrate was concentrated to an oil, which was then dissolved in CH2Cl2 and chromatographed over silica gel using EtOAc/CH2Cl2 (15-50%) to afford an additional 295 mg of product as a tan solid. Total yield of 1-ethyl-N-hydroxy-6-methoxy-1H-indole-3-carboxamidine was 820 mg (70%).


Step B: The N-hydroxycarboxamidine above (50 mg, 0.21 mmol), polystyrene-diisopropylethylamine 165 mg, 3.90 mmol/g loading) and propionyl chloride (0.03 mL, 0.32 mmol) in CH2Cl2 (10 mL) were placed in a tube and rotated for 22 h at room temperature. After this time, trisamine resin (77 mg, 2.71 mmol/g loading) was then added and the tube rotated for an additional 30 min at room temperature. Solids were filtered and then the filtrate was concentrated and diluted with toluene (5 mL) and heated at 110° C. overnight. The crude reaction mixture was concentrated and purified by chromatography (EtOAc/CH2Cl2, 2/98) to afford 27 mg (46%) of 1-ethyl-3-(5-ethyl-[1,2,4]oxadiazol-3-yl)-6-methoxy-1H-indole as a white solid.


The following compound was prepared utilizing the above procedure with substitution of the appropriate acyl halide: compound 29.


Example 1J
Preparation of 1-ethyl-6-methoxy-3-(5-ethyl-[1,3,4]oxadiazol-2-yl)-1H-indole (compound 54)



embedded image


Step A: A mixture of 1-ethyl-6-methoxy-1H-indole-3-carbonitrile (1.00 g, 5.00 mmol) in toluene (30 mL) was treated with triethylamine hydrochloride (1.03 g, 7.50 mmol) and sodium azide (0.49 g, 7.50 mmol) and was heated at reflux for 16 h. After cooling to room temperature, the reaction mixture was diluted with saturated NaHCO3 and extracted with EtOAc. The organic layer was then washed with additional NaHCO3 (2×). The combined aqueous phases were acidified to pH 2 with 6N HCl. The resultant thick precipitate was extracted with hot EtOAc (3×) and the combined organic phases were washed with saturated NaCl and dried and concentrated to give 0.55 g (45%) of 1-ethyl-6-methoxy-3-(1H-tetrazol-5-yl)-1H-indole as a yellow solid.


Step B: A suspension of the tetrazole above (50 mg, 0.21 mmol) and propionyl chloride (0.03 mL, 0.31 mmol) in dichloroethane (5 mL) was heated at reflux for 21 h. After cooling the reaction mixture to room temperature, polystyrene trisamine resin (70 mg, 3.4 meq/g) was added and the reaction was rotated for 4 h at room temperature. After filtering off the resin, and removal of the solvent, the crude product was absorbed on silica gel and the product was isolated by silica gel chromatography (EtOAc/CH2Cl2, 5-10%) to afford 30 mg (53%) of 1-ethyl-6-methoxy-3-(5-ethyl-[1,3,4]oxadiazol-2-yl)-1H-indole as a tan solid.


Example 1K
Preparation of ethyl 5-difluoromethoxy-1-(4-methoxyphenyl)-2-methyl-1H-indole-3-carboxylate (compound 49)



embedded image


Freon-22 (HCF2Cl) gas was bubbled into a solution of ethyl 5-hydroxy-1-(4-methoxyphenyl)-2-methyl-1H-indole-3-carboxylate (250 mg, 0.77 mmol) in CH2Cl2 (5 mL) at 0° C. containing a small amount of tetrabutylammonium bromide as a phase transfer catalyst. A 50% solution of NaOH was added dropwise at 0° C. After the addition, the mixture was stirred at 0° C. for 2 h. After the addition of H2O, the organic phase was separated and washed with brine and dried over Na2SO4. The solvent was then concentrated and the residue was purified by column chromatography over silica gel using EtOAc/petroleum ether (1/2) as eluent to yield the desired product in 40% yield.


The following compounds were prepared utilizing the above procedure with substitution of the appropriate hydroxyindole: compounds 18, 46, and 50.


Example 1L
Preparation of 1-[5-methoxy-1-(4-methoxyphenyl)-1-H-indol-3-yl]-ethanone (compound 42)



embedded image


5-Methoxy-1-(4-methoxyphenyl)-1-H-indole (50 mg, 0.2 mmol), prepared by the method of example 1C, was dissolved in 1 mL of CH2Cl2 at 0° C. Et2AlCl (300 μL, 1M in hexanes, 0.3 mmol) was then added. After stirring at 0° C. for 30 min, a solution of acetyl chloride (22 μL, 0.3 mmol) in 1 mL of CH2Cl2 was added dropwise. This was stirred at 0° C. for a further 90 min. The reaction mixture was quenched with H2O and was extracted with CH2Cl2 and concentrated in vacuo. Purification by column chromatography on silica gel EtOAc/CH2Cl2 (5/95) yielded the title compound as a white solid (42 mg, 71%).


Utilizing essentially the same procedure described above and substituting different acyl chlorides, the following compounds were prepared: compounds 32, 33, 34, 37, 38, 39, 47, 48.


Example 1M
Preparation of 1-ethyl-3-isoxazol-3-yl-6-methoxy-1-H-indole (compound 57)



embedded image


Step A: A mixture of 1-(1-ethyl-6-methoxy-1-H-indole-3-yl)ethanone (200 mg, 0.92 mmol), prepared from 1-ethyl-6-methoxy-1H-indole by the procedure described in example 1L, hydroxylamine hydrochloride (128 mg, 1.84 mmol), NaOAc (151 mg, 1.84 mmol) and EtOH (7 mL) was heated at 85° C. for 4 h. The reaction mixture was then partitioned between H2O and EtOAc. The organic phase was dried and concentrated in vacuo. Purification by column chromatography using EtOAc/CH2Cl2 (1/9) yielded 1-(1-ethyl-6-methoxy-1-H-indole-3-yl)ethanone oxime as a white solid (189 mg, 92%).


Step B: 1-(1-Ethyl-6-methoxy-1-H-indole-3-yl)ethanone oxime (100 mg, 0.43 mmol) was dissolved in THF (900 μL) at 0° C. n-BuLi (450 μL, 2.5 M in hexanes, 1.12 mol) was added dropwise, resulting in instant precipitation of solids. DMF (70 μL, 0.9 mol) in 260 μL of was then added dropwise. This was stirred at 0° C. for 1 h, then at room temperature for 1 h. The reaction mixture was pipetted into a mixture containing 1 mL of H2O, 1 mL of THF, and 100 μL of concentrated H2 SO4. This mixture was heated at 75° C. for 1 h and then was partitioned between H2O and EtOAc. The organic phase was dried and concentrated. Purification by column chromatography (CH2Cl2) yielded 1-ethyl-3-isoxazol-3-yl-6-methoxy-1-H-indole product as a white solid (13 mg, 12%).


Example 1N
Preparation of 1-ethyl-3-isoxazol-5-yl-6-methoxy-1H-indole (compound 58)



embedded image


1-(1-Ethyl-6-methoxy-1H-indol-3-yl)ethanone (100 mg, 0.46 mmol), prepared from 1-ethyl-6-methoxy-1H-indole by the procedure described in example 1L, was heated with 1.5 mL of dimethylformamide dimethylacetal and 100 μL of pyrrolidine at 110° C. overnight. The dimethylformamide dimethylacetal was then concentrated in vacuo. The residue was redissolved in 1.25 mL of EtOH and 250 μL of H2O, and was treated with hydroxylamine hydrochloride (66 mg, 0.95 mmol) and heated at 80° C. for 2 h. Partitioning between H2O and EtOAc and drying and concentration of the organic phase followed by purification by silica gel chromatography (EtOAc/CH2Cl2, 5/95) gave 1-ethyl-3-isoxazol-5-yl-6-methoxy-1H-indole as a white solid (72 mg, 66%).


Utilizing essentially the same procedure described above, the following compound was prepared: Compound 60.


Example 1O
Preparation of 1-ethyl-6-methoxy-3-(2H-pyrazol-3-yl)-1H-indole (compound 59)



embedded image


1-(1-Ethyl-6-methoxy-1H-indol-3-yl)-ethanone (100 mg, 0.46 mmol), prepared from 1-ethyl-6-methoxy-1H-indole by the procedure described in example 1L, was heated with 1.5 mL of dimethylformamide dimethyl acetal and 100 μL pyrrolidine at 110° C. overnight. The DMF dimethyl acetal was removed in vacuo. The residue was redissolved in 3 mL of acetic acid, hydrazine hydrate (70 μL, 1.38 mmol) was added, and the mixture was heated to 100° C. for 2 h. The acetic acid was removed in vacuo, and the residue was partitioned between EtOAc and saturated NaHCO3. The organic phase was dried and concentrated and the product purified by silica gel chromatography (EtOAc/Hex, 1/1) to give 59 mg of 1-ethyl-6-methoxy-3-(2H-pyrazol-3-yl)-1H-indole (54%) as a colorless semisolid. Trituration in Et2O gave a white crystalline powder.


The following compound was prepared utilizing the above procedure: Compound 61.


Example 1P
Preparation of methyl 1-ethyl-3-oxazol-5-yl-1H-indole-6-carboxylate (compound 72)



embedded image


Step A: 1-Ethyl-1H-indole-6-carboxylic acid methyl ester (900 mg, 4.45 mmol) was dissolved in DMF (3.3 mL). This was added dropwise to an ice-cold solution of POCl3 (430 μL, 4.5 mmol) in DMF (1.5 mL). The reaction mixture was stirred at room temperature for 90 minutes. The reaction mixture was then treated with 6N NaOH (3.5 ml). The mixture was then partitioned between H2O and ethyl acetate. Purification by silica gel chromatography (5-10% EtOAc/CH2Cl2) yielded 1-ethyl-3-formyl-1H-indole-6-carboxylic acid methyl ester (985 mg, 96%) as a white solid.


Step B: 1-Ethyl-3-formyl-1H-indole-6-carboxylic acid methyl ester (100 mg, 0.42 mmol), TOSMIC (100 mg, 0.52 mmol), K2CO3 (178 mg, 1.29 mmol), and MeOH (800 μL) were heated at 80° C. overnight. The reaction mixture was then partitioned between H2O and ether. After drying and concentrating the organic phase, the product was purified by silica gel chromatography (EtOAc/CH2Cl2, 10/90) to give methyl 1-ethyl-3-oxazol-5-yl-1H-indole-6-carboxylate (26 mg, 23%) as an off-white solid.


Example 1Q
Preparation of methyl 1-ethyl-3-oxazol-2-yl-1H-indole-6-carboxylate (compound 75)



embedded image


Step A: 1-Ethyl-3-formyl-1H-indole-6-carboxylic acid methyl ester (800 mg, 3.5 mmol), prepared as shown in example 1P, step A, was dissolved in acetone (98 mL). A solution of KMnO4 (655 mg, 4.15 mmol) in H2O (31 mL) was added. The reaction mixture was stirred at room temperature for 90 minutes. Another addition of KMnO4 (108 mg) in H2O (6 mL), followed by stirring for another 45 minutes was required to drive the reaction to completion. The reaction mixture was then quenched with 10% H2O2 (1.5 mL). The mixture was filtered through celite. The filtrate was stripped down under vacuum to roughly ⅓ of the volume. The residue was acidified with 6N HCl, and was extracted into ethyl acetate. The solids isolated from the ethyl acetate layer were triturated with acetone to yield 1-ethyl-1H-indole-3,6-dicarboxylic acid 6-methyl ester (696 mg, 79%) as a light orange solid.


Step B: 1-Ethyl-1H-indole-3,6-dicarboxylic acid 6-methyl ester (600 mg, 2.43 mmol) was suspended in a solution of CH2Cl2 (27 ml) and DMF (20 μL). Oxalyl chloride (470 μL, 5.38 mmol) was added, and the reaction mixture was stirred for 1 hour at room temperature. This mixture was then slowly poured into a rapidly stirring solution of concentrated NH4OH (10 mL). This was then partitioned in H2O and EtOAc. The residue from the ethyl acetate layer was triturated with acetone to yield 6-methoxycarbonyl-1-ethyl-1H-indole-3-carboxamide (511 mg, 85%) as a white solid.


Step C: A mixture of 150 mg (0.61 mmol) of 6-methoxycarbonyl-1-ethyl-1H-indole-3-carboxamide in diglyme (3.6 mL), and bromoacetaldehyde dimethyl acetal (430 μL, 3.7 mmol) was heated at 125° C. for 2 h. The reaction mixture was cooled and partitioned in H2O and EtOAc. The organic phase was dried and concentrated and the product was purified by silica gel chromatography (EtOAc/CH2Cl2 5-10%). The product containing fractions were combined and concentrated and the solid was triturated with hexanes to yield methyl 1-ethyl-3-oxazol-2-yl-1H-indole-6-carboxylate (75 mg, 46%) as a yellow solid.


Example 1R
Preparation of 1-ethyl-6-methoxy-3-thiazol-2-yl-1H-indole (compound 73)



embedded image


Step A: 1-Ethyl-6-methoxy-1H-indole (900 mg, 5.14 mmol) was dissolved in DMF (1.5 mL). This was added dropwise to an ice-cold solution of POCl3 (500 μL, 5.2 mmol) in DMF (1.75 ml). After stirring at room temperature for 90 minutes, the reaction mixture was re-cooled in an ice bath and was slowly quenched with 6N NaOH (4 mL). The reaction mixture was partitioned between EtOAc and H2O. Purification by silica gel chromatography (EtOAc/CH2Cl2, 5/95) yielded 1-ethyl-6-methoxy-1H-indole-3-carbaldehyde (849 mg, 81%) as a yellow solid.


Step B: 1-Ethyl-6-methoxy-1H-indole-3-carbaldehyde (600 mg, 2.95 mmol) was dissolved in acetone (85 mL). A solution of KMnO4 (450 mg, 2.85 mmol) in H2O (28 mL) was added. This was stirred at room temperature for 5 hours. Another solution of KMnO4 (450 mg, 2.85 mmol) in H2O (25 mL) was then added. After stirring for another hour at room temperature, the reaction was complete. The reaction mixture was quenched with 10% H2O2 (1.5 mL), and was then filtered through celite. The filtrate was stripped down under vacuum to roughly ⅓ of the volume. The residue was acidified with 6N HCl, and was extracted into ethyl acetate. Purification by silica gel column (hexanes/acetone/acetic acid, 70/30/1) yielded crude product. Trituration with ether yielded pure 1-ethyl-6-methoxy-1H-indole-3-carboxylic acid (365 mg, 56%) as a yellow solid.


Step C: 1-Ethyl-6-methoxy-1H-indole-3-carboxylic acid (250 mg, 1.14 mmol) was suspended in a solution of CH2Cl2 (12.5 mL) and DMF (10 μL). Oxalyl chloride (230 μL, 2.64 mmol) was added, and the reaction mixture was stirred for 1 hour at room temperature. This mixture was then slowly poured into a rapidly stirring solution of concentrated NH4OH (5 mL). This was then partitioned in H2O and EtOAc. The residue from the ethyl acetate layer was triturated with acetone to yield 1-ethyl-6-methoxy-1H-indole-3-carboxamide (134 mg, 54%) as a white solid.


Step D: 1-Ethyl-6-methoxy-1H-indole-3-carboxamide (120 mg, 0.55 mmol), Lawesson's reagent (240 mg, 0.6 mmol), and toluene (2 mL) were heated at 90° C. for 90 min. The reaction mixture was concentrated and purified by silica gel chromatography (EtOAc/CH2Cl2, 1/9) to yield 1-ethyl-6-methoxy-1H-indole-3-thiocarboxamide as a yellow solid (92 mg, 71%).


Step E: 1-Ethyl-6-methoxy-1H-indole-3-thiocarboxamide (83 mg, 0.36 mmol), glyme (3.6 mL) and bromoacetaldehyde dimethyl acetal (220 μL, 1.86 mmol) were heated at 80° C. for 16 h. More bromoacetaldehyde dimethyl acetal (250 μL was added. This was heated at 80° C. for 2 h. Addition of 250 μL more bromoacetaldehyde dimethyl acetal was followed by heating for another 2 hours. The reaction mixture was cooled to room temperature, absorbed onto silica and purified by silica gel chromatography (hexanes/EtOAc, 7/3) to afford 1-ethyl-6-methoxy-3-thiazol-2-yl-1H-indole as a brown oil (44 mg, 47%).


The following compounds were prepared following the procedure described above: Compounds 78, 101, 104, 105 and 106.


Example 1S
Preparation of 1-ethyl-6-methoxy-2-phenoxymethyl-1H-indole-3-carbonitrile (compound 99)



embedded image


Step A: To a suspension of LiAlH4 (7.6 g, 0.2 mol) in dioxane (100 mL) was added dropwise a solution of methyl 6-methoxy-1H-indole-2-carboxylate (8.2 g, 0.04 mol) in dioxane (50 mL) at 0° C. After the addition, the mixture was stirred at room temperature for 1 h and then heated at reflux for 5 h. After cooling to 0° C., the reaction was quenched by water (dropwise) and then 15% aqueous NaOH. After stirring at room temperature for 1 h, the mixture was filtered through Celite. The solid was washed with large amount of EtOAc. The solvent was washed with brine, dried over Na2SO4 and evaporated under vacuum. The residue was purified by flash column chromatography on silica gel using EtOAc/petroleum ether (1/5) as eluent to yield 61% of 6-methoxy-2-methyl-1H-indole.


Step B: To a solution of 6-methoxy-2-methyl-1H-indole (3.9 g, 24 mmol) in acetonitrile (200 mL) and DMF (20 mL) was added dropwise a solution of ClSO2NCO (4 mL, 1.3 eq.) in acetonitrile (31 mL) at 0° C. After the addition, the mixture was stirred at room temperature for 3 h. Then it was poured into ice water and saturated NaHCO3 was added to it until it became basic. The aqueous phase was extracted with CH2Cl2 and then evaporated. The residue was purified with flash column chromatography on silica gel using EtOAc/petroleum ether (1/5) as eluent to yield 81% of 6-methoxy-2-methyl-1H-indole-3-carbonitrile.


Step C: To a suspension of NaH (0.6 g, 2 eq.) in DMF (7 mL) was added a solution of 6-methoxy-2-methyl-1H-indole-3-carbonitrile (1.3 g, 7.0 mmol) in DMF (8 mL) followed by ethyl iodide (1.2 mL, 2 eq.) at 0° C. After stirring for 1 h, the mixture was poured into ice water and the extracted with CH2Cl2. The organic layer was washed with brine and dried with Na2SO4. The solvent was evaporated under vacuum and purified with flash column chromatography on silica gel using EtOAc/petroleum ether (1/5) as eluent to yield 92% of 1-ethyl-6-methoxy-2-methyl-1H-indole-3-carbonitrile.


Step D: To a solution of 1-ethyl-6-methoxy-2-methyl-1H-indole-3-carbonitrile (1.38 g, 6.45 mmol) in benzene (130 mL) was added benzoyl peroxide (226 mg) and NBS (1.21 g, 1.05 eq.). Then the mixture was heated to reflux for 3 h. After cooling and filtering, the filtrate was concentrated under vacuum. The crude 2-bromomethyl-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (1.6 g, 86%) was used without further purification.


Step E: To a solution of NaH (44 mg, 4 eq.) in DMF (0.5 mL) was added 2-bromomethyl-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (80 mg, 0.274 mmol) and phenol (2 eq.). After stirring for 20 h, the mixture was poured into ice water and extracted with CH2Cl2. The organic layer was washed with brine and dried with Na2SO4. The solvent was evaporated under vacuum and purified with flash column chromatography on silica gel using EtOAc/petroleum ether (1/5) as eluent to yield 1-ethyl-6-methoxy-2-phenoxymethyl-1H-indole-3-carbonitrile, compound 99.


Example 1T
Preparation of 6-nitro-2-pyrrol-1-yl-1H-indole-3-carbonitrile (compound 7)



embedded image


Step A: A solution of 2-fluoro-5-nitroaniline (11.7 g, 74.9 mmol) in dimethylformamide (120 mL) was treated with malononitrile (5.28 g, 80.0 mmol) and potassium carbonate (11.05 g, 80.0 mmol) (Modification of Chem. Heterocyclic Cpd. (Engl. Trans., 9, 37 (2001). The resulting heterogeneous mixture was heated to gentle reflux for 3 h, then cooled and poured into water (500 mL). The resulting precipitate was collected by filtration and taken up into ethyl acetate (300 mL). This solution was dried over Na2SO4, filtered and partially evaporated to give a precipitate, which was collected by filtration. Further evaporation and filtration gave a second crop. The two crops were combined and dried under vacuum to give 2-amino-1-ethyl-6-nitro-1H-indole-3-carbonitrile (7.90 g, 52%) as an orange powder.


Step B: A solution of 2-amino-6-nitro-1H-indole-3-carbonitrile (362 mg, 1.79 mmol) in acetic acid (5 mL) was treated with 2,5-dimethoxytetrahydrofuran (0.30 mL, 2.27 mmol), and the solution was heated to reflux for 14 h. After cooling to ambient temperature, the solution was poured into water (100 mL), and solid sodium bicarbonate was added until CO2 evolution ceased. The mixture was extracted with EtOAc (2×100 mL), and the extracts were washed with saturated brine, combined, dried over MgSO4, filtered and concentrated. The residual material was separated by silica gel chromatography (EtOAc/hexanes, 1/4) to afford 6-nitro-2-pyrrol-1-yl-1H-indole-3-carbonitrile, compound 5, as a yellow solid (232 mg, 51%).


Example 1U
Preparation of N-(3-cyano-1-ethyl-6-nitro-1H-indol-2-yl)acetamide (compound 25)



embedded image


Step A: Sodium hydride (42 mg, 1.05 mmol, 60% w/w suspension in mineral oil) was washed with hexane and taken up in dimethylsulfoxide (1 mL). A solution of 2-amino-6-nitro-1H-indole-3-carbonitrile, prepared in procedure 1T) in dimethylsulfoxide (1 mL) was added by syringe, and the resulting mixture was stirred for 20 min. Then, iodoethane (77 μL, 0.96 mmol) was added by syringe, and the mixture was stirred for 14 h. The reaction was then poured into EtOAc (50 mL), and this solution was washed with water (3×50 mL) and saturated brine (40 mL). The aqueous phases were back-extracted with EtOAc, and the organic extracts were combined, dried over Na2SO4, filtered and evaporated. The residual material was separated by column chromatography over silica gel (EtOAc/hexanes, 1/1) to afford first a small amount of a dialkylated analog, then the desired compound, 2-amino-1-ethyl-6-nitro-1H-indole-3-carbonitrile (114 mg, 52%), and finally unreacted starting material. The desired product was isolated as an orange powder.


Step B: Sodium hydride (44 mg, 1.10 mmol, 60% w/w in mineral oil) was washed with hexanes and suspended in 1,4-dioxane (3 mL). A solution of 2-amino-1-ethyl-6-nitro-1H-indole-3-carbonitrile (120 mg, 0.521 mmol), prepared in step B, above, in dioxane (2 mL) was added, and the resulting mixture was allowed to stir for 30 min. Then, acetyl chloride (45 μL, 0.63 mmol) was added by syringe, and the solution was stirred for an additional 12 h. The reaction was partitioned between water and EtOAc (20 mL each), and the organic phase was washed with brine. The aqueous phases were back-extracted in sequence with ethyl acetate, and the organic extracts were combined, dried over MgSO4, filtered and evaporated. The resulting solid was triturated with Et2O, collected by filtration and dried under vacuum to afford N-(3-cyano-1-ethyl-6-nitro-1H-indol-2-yl)-acetamide (100 mg, 71%), compound 25, as an off-white powder.


Using this procedure and substituting the appropriate acid chlorides or chloroformates gave the following compounds: Compounds 23, 26, 35, 36, 203, 204, 214, 215, 216.


Example 1V
Preparation of N-ethyl-3-phenyl-5-nitroindole (compound 41)



embedded image


Step A: To a solution of 5-nitroindole (5.00 g, 30.8 mmol) in pyridine (200 mL) at −4° C. was added a solution of pyridinium bromide perbromide (10.99 g, 34.3 mmol) in pyridine (200 mL) dropwise under nitrogen with stirring. After complete addition, the reaction mixture was stirred for 5 min at 0° C. The reaction mixture was diluted in 0° C. water (200 mL) and extracted with 200 mL of Et2O. The organic layer was washed with 6 M HCl (300 mL), 5% NaHCO3 (300 mL), and brine (300 mL). The organic phase was dried over MgSO4 and solvent was removed to give 3-bromo-5-nitroindole as a yellow powder, 80% pure with 20% 5-nitroindole (6.80 g, 74% yield).


Step B: A solution of 3-bromo-5-nitroindole from above (625 mg, 2.1 mmol), phenylboronic acid (381 mg, 3.13 mmol), triphenylphosphine (109.3 mg, 0.417 mmol) in dimethoxyethane (4.16 mL) was degassed. To this mixture 2N sodium carbonate (6.25 mL) was added, and reaction mixture was degassed again. To the reaction was added palladium (II) acetate (23.4 mg, 0.104 mmol), and the reaction was refluxed under dry nitrogen with stirring for 8 hours. The reaction mixture was then diluted with 1 M HCl (100 mL), and extracted with ethyl acetate (100 mL). The organic phase was washed with water (100 mL), and brine (100 mL). The organic phase was dried over MgSO4 and concentrated in vacuo. The crude product was purified by chromatography over silica gel (EtOAc/hexanes, 10/90) to afford 3-phenyl-5-nitroindole as an orange powder (45 mg, 9% yield).


Step C: To a mixture of 60% NaH in mineral oil (8.7 mg, 0.630 mmol) and DMF (1.0 mL) was added dropwise a solution of 3-phenyl-5-nitroindole (40.0 mg, 2.1 mmol) in DMF (0.75 mL). The reaction mixture was stirred for 20 min at 0° C. under N2. Ethyl iodide (14.8 μL, 0.185 mmol) was added dropwise and the reaction mixture was stirred for an additional 3 hours. The reaction mixture was diluted with water (250 mL), and extracted with EtOAc (30 mL). The organic phase was washed with water (250 mL) and was then dried over MgSO4 and the solvent was removed in vacuo. The desired N-ethyl-3-phenyl-5-nitroindole was obtained as a yellow powder (40.0 mg, 89.5% yield).


In similar fashion the following compound was prepared: Compound 40


Example 1W
Preparation of [3-Cyano-1-(4-methoxyphenyl)-1H-indol-6-yl]-carbamic acid propyl ester (compound 97)



embedded image


6-Amino-1-(4-methoxyphenyl)-1H-indole-3-carbonitrile (30 mg, 0.12 mmol), was suspended in EtOH (300 μL). Propyl chloroformate (168 μL, 1.5 mmol) was added, and this mixture was stirred at room temperature overnight. The addition of triethylamine (300 μL), followed by another hour of stirring at room temperature, completed the reaction. This reaction mixture was loaded directly onto a silica column, and was eluted with CH2Cl2. Another silica column (3/2, ether/hexanes) was needed to fully purify the product, [3-cyano-1-(4-methoxy-phenyl)-1H-indol-6-yl]-carbamic acid propyl ester (19 mg, 45%), as a white solid.


Example 1X
Preparation of N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl]-methanesulfonamide (compound 130)



embedded image


2-(4-Aminophenylethynyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (50 mg, 0.16 mmol), prepared as described by the method of Example 1H, was dissolved in pyridine (550 μL) at room temperature. Methanesulfonyl chloride (17 μL, 0.21 mmol) was added dropwise. This was stirred overnight at room temperature. The reaction mixture was then diluted in ethyl acetate and was washed with aqueous HCl, followed by brine. The organic layer was dried and concentrated. Purification by silica gel chromatography (9/1, CH2Cl2/EtOAc) yielded N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl]-methanesulfonamide (58 mg, 92%) as an off-white solid.


The following compounds were made using the procedure shown above, by substituting the appropriate aminophenylethynyl indoles and sulfonyl chlorides: Compounds 131, 132, 208, 209, and 210.


Example 1Y
Preparation of N-[4-(3-Cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-methanesulfonamide (compound 129)



embedded image


A solution of 2-(4-aminophenyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (70 mg, 0.24 mmol), prepared as described in Example 1Ga, step B in THF (3 mL) was cooled to 0° C. and treated with triethylamine (0.04 mL, 0.31 mmol) and methanesulfonylchloride (0.02 mL, 0.29 mmol) at stirred, warming to room temperature overnight. The reaction mixture was then diluted with H2O and extracted with ethyl acetate (3×). The organic phase was washed with H2O and saturated NaCl, dried and concentrated and purified by flash chromatography using EtOAc/hexanes (30-50%) to afford 60 mg (68%) of N-[4-(3-Cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-methanesulfonamide as a tan solid.


Using essentially the same procedure as above and substituting the appropriate aminophenylindole and sulfonyl chloride or carrying out the reaction in pyridine as both base and solvent gave the following compounds: 83, 85, 86, 87, 88, 243, 251, 252, 272, 273, 287, 289, 365, 366, 367, 368, 369, 370, 371, 394, 439, 440, 448, 449, 451, 452, 477, 487, 488, 495, 505, 510, 548, 549, 550, 551, 552, 562, 563, 598, 599, 601, 602, 608, 609, 610, 615, 616, 617, 621, 622, 623, 629, 630, 631, 639, 655, 657, 658, 662, 669, 670, 671, 674, 675, 701, 702, 703, 706, 707, 708, 709, 710, 711, 713, 715, 720, 789, 790, 791, 850, 851.


Example 1Za
Preparation of N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl]-acetamide (compound 138)



embedded image


2-(4-Aminophenylethynyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (95 mg, 0.29 mmol), prepared as described in Example 1H, was dissolved in THF (1.4 mL). Triethylamine (84 μL, 0.6 mmol) was added, followed by dropwise addition of acetyl chloride (44 μL, 0.5 mmol). This was stirred at room temperature for 1 h. The reaction mixture was partitioned between H2O and EtOAc. The organic layer was dried and concentrated. Purification by silica chromatography (9/1, CH2Cl2/EtOAc) yielded N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl]-acetamide (103 mg, 96%) as a yellow solid.


The following compounds were prepared by the procedure shown above, substituting the appropriate aminophenylethynyl indoles and acid chlorides: Compounds 82, 139, 152, 153, 162, 163, 165, 167, 205, 206, 207, 211, 212, 213, 219, 224, 225, 228.


Example 1Zb
Preparation of N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl]-formamide (compound 241)



embedded image


Acetic anhydride (2.5 mL) and 98% formic acid (1.0 mL) were heated at 65° C. for 1 hour. This was cooled to 0° C. 2-(4-Aminophenylethynyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (100 mg, 0.32 mmol), prepared as in example 1H, was taken up in THF (1.2 mL) and added to the formic acetic anhydride mixture. This was stirred at 0° C. for 30 minutes. The reaction mixture was then partitioned between H2O and EtOAc. The EtOAc layer was washed with saturated NaHCO3, followed by saturated brine. The organic layer was dried and concentrated. Purification by silica gel chromatography (4/1, CH2Cl2/EtOAc) yielded of N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl]-formamide (105 mg, 96%) as a yellow solid.


The following compound was prepared similarly as described above: Compound 218.


Example 1AA
Preparation of N-[4-(3-Cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-acetamide (compound 128)



embedded image


A solution of 2-(4-aminophenyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (70 mg, 0.24 mmol), prepared as described in example 1Ga, step B in THF (3 mL) was cooled to 0° C. and treated with triethylamine (0.04 mL, 0.31 mmol) and acetyl chloride (0.02 mL, 0.29 mmol) and stirred, warming to room temperature overnight. The reaction mixture was then diluted with H2O and extracted with ethyl acetate (3×). The organic phase was washed with H2O and saturated NaCl, dried and concentrated and purified by flash chromatography using EtOAc/hexanes (30-50%) to afford 57 mg (71%) of N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]acetamide as a tan solid.


Using essentially the same procedure as above and substituting appropriate aminophenyl indoles and the acid chlorides, the following compounds were prepared: Compounds 81, 242, 244, 324, 325, 326, 327, 328, 329, 330, 383, 420, 421, 422, 423, 424, 425, 544, 558, 559, 560, 561, 565, 566 567, 644, 645, 646, 755, 756, 757, 759, 760, 761, 762, 763, 764, 765, 766, 798, 799, 801, 802, 803, 804, 854, 855, 856, 857, 858, 859.


Example 1AB
Preparation of 1-[3-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)phenyl]-3-ethyl urea (compound 220)



embedded image


2-(3-Aminophenylethynyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (100 mg, 0.32 mmol), prepared as described in Example 1H, was dissolved in pyridine (670 μL). Ethyl isocyanate (62 μL, 0.75 mmol) was added. The reaction mixture was then heated at 100° C. for 2 h. The mixture was then diluted in EtOAc, and was washed with aqueous HCl, followed by brine. The organic layer was dried and concentrated. Purification by silica chromatography (4/1, CH2Cl2/EtOAc), followed by trituration with hexanes/acetone (1/1), yielded 1-[3-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl]-3-ethyl urea (44 mg, 36%) as a white solid.


Example 1AC
Preparation of 1-(2-chloroethyl)-3-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl] urea (compound 156)



embedded image


2-(4-Aminophenylethynyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (100 mg, 0.32 mmol), prepared as described in Example 1H, was suspended in toluene (600 μL). 2-Chloroethyl isocyanate (32 μL, 0.37 mmol) was added, and the mixture was heated at 100° C. for 5 h. The reaction mixture was then cooled, diluted in acetone, and absorbed onto silica. Purification by column chromatography (5-10% EtOAc in CH2Cl2) yielded 1-(2-chloro-ethyl)-3-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl] urea (73 mg, 54%) as a yellow solid.


The following compounds were prepared using the procedure above: Compound 221.


Example 1AD
Preparation of Ethanesulfonic acid [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl]methyl amide (compound 157)



embedded image


N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)phenyl] ethanesulfonamide (70 mg, 0.17 mmol), prepared as in example 1X, was combined with K2CO3 (49 mg, 0.35 mmol), and DMF (1.0 mL). Iodomethane (16 μL, 0.26 mmol) was added, and the mixture was stirred at room temperature for 1 hour. The reaction mixture was then diluted in EtOAc, and was washed with H2O and then brine. The organic layer was dried and concentrated. Purification by silica chromatography (95/5, CH2Cl2/EtOAc) yielded a light tan solid. Trituration gave ethanesulfonic acid [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl]methyl amide (61 mg, 85%) as an orange-white solid.


The following compounds were prepared using the procedure above, substituting the appropriate sulfonamide: Compound 182, 652, 840.


Example 1AE
Preparation of 1-ethyl-5-methoxy-2-[4-(morpholine-4-carbonyl)-phenyl]-1H-indole-3-carbonitrile (compound 245)



embedded image


Step A: Methyl 4-(3-cyano-1-ethyl-5-methoxy-1H-indol-2-yl)-benzoate (350 mg, 1.05 mmol), prepared as described in Example 1Ga step B, was combined with NaOH (40 mg, 1 mmol), H2O (0.8 mL), and THF (3.4 mL) and was heated at 80° C. for 1 hour. The reaction mixture was diluted in H2O and was then ether-washed. The aqueous layer was acidified with aqueous HCl, and was extracted into EtOAc. The organic layer was dried and concentrated to yield 4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-benzoic acid (311 mg, 92%) as a pure white solid.


Step B: 4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-benzoic acid (50 mg, 0.16 mmol) was suspended in CH2Cl2 (2.2 mL) and catalytic DMF (2 μL). Oxalyl chloride (22 μL, 0.25 mmol) was added. The reaction mixture was stirred at room temperature for 1 hour, at which time full dissolution occurred. This reaction mixture was pipetted dropwise into a vigorously stirring solution of morpholine (1.0 mL) in CH2Cl2 (5 ml). After addition was complete, the reaction mixture was washed with aqueous HCl solution. The organic layer was dried and concentrated. Purification by silica column (1:1 CH2Cl2/EtOAc) yielded 1-ethyl-6-methoxy-2-[4-(morpholine-4-carbonyl)-phenyl]-1H-indole-3-carbonitrile (56 mg, 90%) as a white solid.


The following compounds were prepared similarly as described above: Compounds 113, 114, 246, 270, 271 290, 291, 292, 323, 377, 378, 379, 380, 381, 382, 384, 385, 386, 387, 388, 389, 390, 391, 392, 432, 433, 564, 568, 569, 570, 571, 572, 573, 647, 648, 853, 860, 861, 862.


Example 1AF
Preparation of cyclopropanecarboxylic acid [4-(3-cyano-1-ethyl-6-hydroxy-1H-indol-2-ylethynyl)-phenyl] amide (compound 194)



embedded image


Cyclopropanecarboxylic acid [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl]-amide (60 mg, 0.16 mmol), prepared as described in example 1Za, was stirred in BBr3 (800 μL, 1M in CH2Cl2, 0.8 mmol) at room temperature for 1 hour. The reaction mixture was quenched with H2O, and was extracted with CH2Cl2. The organic layer was dried and concentrated. Purification by silica chromatography (EtOAC) gave impure product. These crude product was triturated with 1/1 hexanes/acetone to yield cyclopropanecarboxylic acid [4-(3-cyano-1-ethyl-6-hydroxy-1H-indol-2-ylethynyl)-phenyl]-amide (32 mg, 54%) as an off-white solid.


The following compounds were prepared using the procedure above, substituting the appropriate sulfonamides (from example 1X) or amides (from Example 1Z): Compounds 164, 168, 183, 193, 195.


Example 1AG
Preparation of 1-ethyl-6-methoxy-2-[4-(2-oxo-imidazolidin-1-yl)-phenylethynyl]-1H-indole-3-carbonitrile (compound 166)



embedded image


1-(2-Chloroethyl)-3-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl] urea (55 mg, 0.13 mmol), prepared as in Example 1AC, was combined with K2CO3 (50 mg, 0.36 mmol) and DMF (550 μL). This mixture was stirred at room temperature for 3 hours. The reaction mixture was diluted in EtOAc, and was washed with H2O, and then with brine. The organic layer was dried and concentrated. Purification by silica chromatography (10-50%, EtOAc/CH2Cl2) yielded 1-ethyl-6-methoxy-2-[4-(2-oxo-imidazolidin-1-yl)-phenylethynyl]-1H-indole-3-carbonitrile (47 mg, 94%) as a white solid.


The following compounds were prepared using the above procedure, substituting the appropriate urea: Compound 222.


Example 1AH
Preparation of N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl]-dimethylphosphinic amide (compound 227)



embedded image


2-(3-Aminophenylethynyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (100 mg, 0.32 mmol), prepared as described in Example 1H, was dissolved in pyridine (300 μL) at 0° C. Dimethylphosphinic chloride (60 mg, 0.53 mmol) in THF (300 μL) was added. The reaction was stirred at room temperature for 2 hours. The reaction mixture was diluted in EtOAc, and was washed with aqueous HCl followed by brine. The organic layer was dried and concentrated. Purification by silica chromatography (acetone) yielded N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl]-dimethylphosphinic amide (65 mg, 52%), compound 227, as a pure white solid. The silica column was then flushed with 9/1 CH2Cl2/MeOH to yield 9 mg of N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-ylethynyl)-phenyl]-bis-(dimethylphosphinic) amide as a by-product.


Example 1AI
Preparation of 1-ethyl-6-methoxy-3-[5-(4-methoxyphenyl)-isoxazol-3-yl]-1H-indole (compound 116)



embedded image


Step A: A mixture of 1-ethyl-6-methoxy-1H-indole-3-carbaldehyde oxime (0.20 g, 0.92 mmol), prepared from the aldehyde precursor in example 1R, in dichloroethane (3 mL) was treated with N-chlorosuccinimide (0.12 g, 0.92 mmol) and pyridine (0.04 mL, 0.46 mmol) and stirred at room temperature for 1 h. The reaction mixture was then poured into H2O and acidified with 1N HCl until the pH was 2. The mixture was extracted with EtOAc and the organic phases were washed with H2O and saturated NaCl and dried and concentrated to a mixture of chlorooximes, which were used in the next step without further purification.


Step B: The mixture of chlorooximes prepared above was dissolved in CH2Cl2 (5 mL) and to this was added 4-methoxyphenylacetylene (0.24 g, 1.84 mmol) and triethylamine (0.25 mL, 1.84 mmol) at 0° C. and the reaction was then stirred overnight warming to room temperature. The reaction was then diluted with H2O and extracted with EtOAc (3×). The organic phases were washed with H2O and saturated NaCl and dried and concentrated. Chromatography over silica gel (EtOAc/hexanes, 10-20%) gave 76 mg (24%) of 1-ethyl-6-methoxy-3-[5-(4-methoxy-phenyl)-isoxazol-3-yl]-1H-indole as a tan solid.


Example 1AJ
Preparation of [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-carbamic acid ethyl ester (compound 121)



embedded image


A biphasic mixture of 2-(4-amino-phenyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (70 mg, 0.24 mmol), prepared as described in example 1Ga step B, and ethyl chloroformate (0.03 mL, 0.29 mmol) in EtOAc (3 mL) and saturated NaHCO3 (3 mL) was prepared at 0° C. and then allowed to warm to room temperature and stirred for 24 h. The reaction was then diluted with H2O and extracted with EtOAc (2×). The organic phases were washed with H2O and saturated NaCl and then dried and concentrated. Flash chromatography (EtOAc/hexanes 20-40%) gave 48 mg (55%) of [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-carbamic acid ethyl ester as an off-white solid.


The following compound was prepared in similar fashion: Compound 122, 293, 294, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, 349, 350, 351, 352, 353, 354, 355, 356, 357, 358, 359, 360, 361, 372, 434, 435, 450, 453, 454, 455, 457, 485, 486, 489, 490, 500, 501, 502, 503, 506, 507, 508, 509, 545, 546, 547, 553, 554, 555, 556, 557, 581, 582, 583, 584, 585, 585, 586, 587, 588, 589, 590, 591, 592, 593, 594, 595, 596, 597, 603, 604, 605, 606, 607, 618, 619, 624, 625, 637, 640, 641, 664, 665, 676, 677, 721, 722, 723, 734, 735, 736, 737, 738, 739, 744, 745, 746, 747, 787, 788, 792, 793, 794, 795, 796, 797, 819, 822, 823, 824, 825, 826, 849.


Example 1AK
Preparation of 1-ethyl-5-thiophen-3-yl-1H-indole-3-carbonitrile (compound 141)



embedded image


A tube was charged with a mixture of 5-bromo-1-ethyl-1H-indole-3-carbonitrile (100 mg, 0.40 mmol), thiophene-3-boronic acid (72 mg, 0.56 mmol), PdCl2(PPh3)2 (11 mg, 0.016 mmol) and CsF (152 mg, 1 mmol) and then alternately evacuated and filled with nitrogen (3×) and diluted with dimethoxyethane (3 mL) and then heated to 90° C. for 19 h. After cooling, the crude reaction mixture was diluted with saturated NaHCO3 and extracted with EtOAc (2×). The combined organic phases were washed with saturated NaCl and dried and concentrated. Flash chromatography over silica gel (CH2Cl2/hexanes, 40/60) gave 25 mg (25%) of 1-ethyl-5-thiophen-3-yl-1H-indole-3-carbonitrile as a white solid.


The following compounds were prepared in similar fashion: Compounds 140 and 142.


Example 1AL
Preparation of N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-N-methyl methanesulfonamide (compound 180)



embedded image


A solution of N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]methanesulfonamide (130 mg, 0.35 mmol), prepared as in Example 1Y, in DMF (10 mL) was treated with NaH (21 mg, 0.53 mmol), and stirred at room temperature for 10 min. Iodomethane (0.03 mL, 0.53 mmol) was added, and the mixture was stirred at room temperature for 18 h. The reaction mixture was then diluted with H2O, and extracted with EtOAc (2×). The organic phases were washed with H2O and saturated NaCl and then dried and concentrated. Purification by flash chromatography over silica gel (EtOAc/CH2Cl2, 0-1%) gave 60 mg (45%) of N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-N-methyl methanesulfonamide as a white solid.


In similar fashion the following compounds were prepared: Compounds 181, 642, 643, 672, 673, 816, 852.


Example 1AM
Preparation of N-[4-(3-cyano-1-ethyl-6-hydroxy-1H-indol-2-yl)-phenyl]-methanesulfonamide (compound 189)



embedded image


A solution of N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]methanesulfonamide (85 mg, 0.23 mmol) in CH2Cl2 (2 mL) was cooled to −5° C. A solution of boron tribromide (1.15 mL, 1.15 mmol, 1M solution in CH2Cl2) was added and the reaction mixture was allowed to warm to 110° C. over 4 h. The reaction mixture was poured into H2O and extracted with EtOAc (3×). The combined organic phases were washed with H2O and saturated NaCl and dried and concentrated. Chromatography over silica gel (EtOAc/CH2Cl2, 5-10%) gave 18 mg (22%) of N-[4-(3-cyano-1-ethyl-6-hydroxy-1H-indol-2-yl)-phenyl]methane sulfonamide as a tan solid.


The following compounds were made similarly: Compounds 190, 191, 192.


Example 1AN
Preparation of methyl 3-[5-(3-cyano-6-methoxy-1H-indol-2-yl)-[1,2,4]oxadiazol-3-yl]benzoate (compound 226)



embedded image


Step A: To a mixture of 6-methoxy-1H-indole-3-carbonitrile (5.88 g, 40 mmol), prepared as described in the previous examples, and (Boc)2O (9.59 g, 44.0 mmol) in DCM (50 mL) was added DMAP (0.10 g, 0.8 mmol). The mixture was stirred at room temperature for 48 h, then treated with water (30 mL) and dried over anhydrous Na2SO4. The crude product was chromatographed over silica gel (hexanes/EtOAc, 7/1) to furnish the desired intermediate, 3-cyano-6-methoxyindole-1-carboxylic acid tert-butyl ester (8.48 g, 86%).


Step B: The above intermediate (2.72 g, 10.0 mmol) was dissolved in anhydrous THF (20 mL), and cooled at −78° C., followed by the addition of LDA (1.5 M monoTHF in cyclohexane, 10.0 mL, 15 mmol). After stirring for 45 min, CO2 gas was introduced for 2 h. The mixture was then brought to room temperature and the solvent was removed in vacuo, and the residue was treated with water and acidified to pH=2 with 6 N HCl. The precipitate was collected and washed with water and dried to provide the acid intermediate, 3-cyano-6-methoxy-indole-1,2-dicarboxylic acid 1-tert-butyl ester (2.40 g, 73%).


Step C: To a solution of 3-cyano-6-methoxyindole-1,2-dicarboxylic acid 1-tert-butyl ester (474 mg, 1.5 mmol) prepared above, and HOBt (200 mg, 1.5 mmol) in DCE/DMF (10 mL/1 mL), was added DCC (310 mg, 1.5 mmol), followed by 3-(N-hydroxycarbamimidoyl)benzoic acid methyl ester (291 mg, 1.5 mmol). The mixture was stirred at room temperature for 2 h and filtered. The filtrate was collected and the solvent was replaced with chlorobenzene, followed by the heating at 150° C. for 48 h. After cooling to room temperature, the solvent was removed in vacuo and the residue was chromatographed (silica gel, CH2Cl2/EtOAc, 8/2) to furnish the intermediate, 3-cyano-6-methoxy-2-[3-(3-methoxycarbonylphenyl)-[1,2,4]oxadiazol-5-yl]-indole-1-carboxylic acid tert-butyl ester, which was treated with 50% TFA in DCM (10.0 mL) at room temperature for 1 h. After removal of the volatiles in vacuo, the residue was suspended in water and neutralized with K2CO3 to provide the desired product, methyl 3-[5-(3-cyano-6-methoxy-1H-indol-2-yl-)[1,2,4]oxadiazol-3-yl]benzoate, compound 226 (350 mg, 62%).


Example 1AO
Preparation of 1-ethyl-2-(4-methanesulfonylphenyl)-6-methoxy-1H-indole-3-carbonitrile (compound 265)



embedded image


A solution of 1-ethyl-6-methoxy-2-(4-methylsulfanylphenyl)-1H-indole-3-carbonitrile (0.12 g, 0.37 mmol) in CH2Cl2 (5 mL) was treated with m-chloroperbenzoic acid (Aldrich, <77%, 0.26 g,) in one portion and the reaction was stirred for 10 h at room temperature. The reaction was then diluted with H2O and saturated NaHCO3 and extracted twice with EtOAc. The organic phases were washed with NaHCO3 (2×) and saturated NaCl and dried and concentrated to a dark semi-solid. The crude product was purified by flash chromatography (EtOAc/CH2Cl2, 0-3%) through a 5 gram silica cartridge topped with 1 gram of basic alumina to give 72 mg (55%) of 1-ethyl-6-methoxy-2-(4-methylsulfanylphenyl)-1H-indole-3-carbonitrile as an off-white solid.


Example 1AP
Preparation of N-{4-[3-cyano-1-ethyl-6-(2-morpholin-4-yl-ethoxy)-1H-indol-2-yl]-phenyl} methanesulfonamide (compound 478)



embedded image


A solution of N-{4-[6-(2-chloroethoxy)-3-cyano-1-ethyl-1H-indol-2-yl]-phenyl} methanesulfonamide (90 mg, 0.21 mmol), morpholine (0.06 mL, 0.65 mmol), NaI (32 mg, 0.21 mmol) and diisopropyl ethylamine (0.06 mL, 0.32 mmol) in CH3CN (2 mL) was heated in a sealed tube at 100° C. for 25 h. The reaction mixture was cooled to room temperature, diluted with H2O and extracted with EtOAc (3×). The combined organic phases were washed with saturated NaCl, dried and concentrated. The crude solid was triturated with EtOAc and filtered to give 41 mg (41%) of N-{4-[3-cyano-1-ethyl-6-(2-morpholin-4-yl-ethoxy)-1H-indol-2-yl]-phenyl} methanesulfonamide as a tan solid.


The following compounds were made similarly: Compounds 479, 480, 481, 482, 496, 497 and 498.


Example 1AQ
Preparation of 2-morpholin-4-yl-ethanesulfonic acid [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl] amide (compound 653)



embedded image


Step A: A solution of 2-(4-aminophenyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile, prepared by example 1Ga step B, (0.82 mg, 2.82 mmol), in pyridine (10 mL) was treated dropwise with chloroethyl sulfonylchloride (0.38 mL, 3.66 mmol) at room temperature. After stirring for 4 h, the reaction mixture was quenched with ice-water and enough 6N HCl was added until the pH was lowered to 2. The suspension was extracted with hot EtOAc (3×). The organic phases were then washed sequentially with 1N HCl, H2O and saturated NaCl and dried and concentrated to give ethenesulfonic acid [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl] amide as a pale orange solid which was used directly in the next step without further purification.


Step B: A suspension of ethenesulfonic acid [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl] amide, prepared above, (70 mg, 0.18 mmol), morpholine (0.05 mL, 0.55 mmol) in CH3CN (1.5 mL) was heated at reflux for 1.5 h. After cooling to room temperature, the reaction was concentrated and the residue was purified by flash chromatography (acetone/EtOAc, 2/98) over silica gel to afford 89 mg (100%) of 2-morpholin-4-yl-ethanesulfonic acid [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl] amide as a tan foam.


The following compound was made similarly: Compound 654.


Example 1AR
Preparation of 2-morpholin-4-yl-ethanesulfonic acid [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]methyl amide (compound 668)



embedded image


A solution of 2-morpholin-4-yl-ethanesulfonic acid [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl] amide, prepared in example 1AQ (60 mg, 0.13 mmol) in DMF (3 mL) was treated with K2CO3 (35 mg, 0.26 mmol) and methyl iodide (0.02 mL, 0.26 mmol). After stirring at room temperature for 1.5 h, the reaction mixture was diluted with H2O and extracted with EtOAc (2×). The organic phases were then washed with H2O (3×) and saturated NaCl, and then dried and concentrated to afford a residue. Flash chromatography over silica gel (acetone/EtOAc, 0-2%) gave 31 mg (50%) of 2-morpholin-4-yl-ethanesulfonic acid [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]methyl amide as an off white solid.


The following compounds were made similarly: Compounds 684, 685, 686, 687, 688, 689, 690, 691, 692, 693, 694, 695, 696, 697, 698.


Example 1AS
Preparation of 2-[4-(1,1-dioxo-1λ6-isothiazolidin-2-yl)phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (compound 84)



embedded image


Step A: A solution of 2-(4-aminophenyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile, prepared by example 1Ga step B, (2.78 g, 9.55 mmol) in pyridine (40 mL) was treated dropwise with 3-chloropropanesulfonyl chloride (1.45 mL, 11.9 mmol) and the reaction was stirred for 4 h at room temperature. The reaction was diluted with water and enough 6N HCl to lower the pH to 2. The reaction mixture was extracted with EtOAc (3×) and the combined organic layers were washed sequentially with 1N HCl, water and saturated NaCl and then dried and concentrated to give 3.9 g (95%), of 3-chloropropane-1-sulfonic acid [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl] amide as a brown foam which was used directly in the next step.


Step B: A solution of 3-chloropropane-1-sulfonic acid [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl] amide, prepared above (3.65 g, 2.33 mmol) in DMF (100 mL) was treated with K2CO3 and heated at 70° C. for 2 h. After cooling to room temperature, the reaction mixture was diluted with H2O and extracted 3× with hot EtOAc. The hot organic layers were washed with warm H2O (3×) and saturated NaCl and dried and concentrated to a solid. Trituration (CH2Cl2/hexanes) gave 2.27 g (68%) of 2-[4-(1,1-dioxo-1λ6-isothiazolidin-2-yl)phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile as a light brown solid.


The following compounds were made in similar fashion: Compound 649, 775.


Example 1AT
Preparation of 2-[4-(1,1-dioxo-1λ6-isothiazolidin-2-yl)phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (compound 666)



embedded image


Step A: Following the procedure in example 1B step A, 2-[4-(1,1-dioxo-1λ6-isothiazolidin-2-yl)phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile was treated with 1M BBr3 solution in CH2Cl2 at −15° C. for 1.5 h and then poured into ice-water and filtered and dried to afford 2-[4-(1,1-dioxo-1λ6-isothiazolidin-2-yl)phenyl]-1-ethyl-6-hydroxy-1H-indole-3-carbonitrile in nearly quantitative yield.


Step B: Following the procedure in example 1B step B, 2-[4-(1,1-dioxo-1λ6-isothiazolidin-2-yl)phenyl]-1-ethyl-6-hydroxy-1H-indole-3-carbonitrile, K2CO3, 2-iodopropane and methyl ethyl ketone were heated at reflux to give, after flash chromatography (EtOAc/CH2Cl2, 0-2%), 61% of 2-[4-(1,1-dioxo-1λ6-isothiazolidin-2-yl)phenyl]-1-ethyl-6-isopropoxy-1H-indole-3-carbonitrile as an off-white solid.


The following compounds were made similarly: Compounds 667, 699


Example 1AU
Preparation of 2-[4-(1,1-dioxo-1λ6-isothiazolidin-2-yl)-phenyl]-1-ethyl-6-(2-morpholin-4-yl-ethoxy)-1H-indole-3-carbonitrile (compound 729)



embedded image


A mixture of 2-[4-(1,1-dioxo-1λ6-isothiazolidin-2-yl)phenyl]-1-ethyl-6-hydroxy-1H-indole-3-carbonitrile, prepared in example 1AT above (70 mg, 0.25 mmol), K2CO3 (75 mg, 0.51 mmol), sodium iodide (27 mg, 0.18 mmol), 4-(2-chloroethyl) morpholine hydrochloride (42 mg, 0.25 mmol) in methyl ethyl ketone (3 mL) was heated in a sealed tube at 100° C. After 13 hours, DMF (3 mL) was added and the reaction was heated for an additional 6 h. After this time, an additional 42 mg of 4-(2-chloroethyl) morpholine hydrochloride and 135 mg of K2CO3 was added and the reaction was heated for an additional 6 h to complete the reaction. The reaction mixture was cooled to room temperature, diluted with water, and extracted with EtOAc (3×). The combined organic phases were washed with water (2×) and saturated NaCl and dried and concentrated. Pure 2-[4-(1,1-dioxo-1λ6-isothiazolidin-2-yl)-phenyl]-1-ethyl-6-(2-morpholin-4-yl-ethoxy)-1H-indole-3-carbonitrile was obtained by flash chromatography (MeOH/CH2Cl2, 0-6%) to give 29 mg (34%) of a tan solid.


The following compounds were made similarly: Compounds 728 and 730.


Example 1AV
Preparation of 2-[4-(2,5-dioxo-imidazolidin-1-yl)-phenyl]-6-ethoxy-1-ethyl-1H-indole-3-carbonitrile (compound 779)



embedded image


Step A: A solution of 2-(4-aminophenyl)-6-ethoxy-1-ethyl-1H-indole-3-carbonitrile (585 mg, 1.92 mmol) in 10 mL of 1,4-dioxane was treated with ethyl isocyanatoacetate (0.25 mL, 2.12 mmol), and the resulting solution was heated to reflux overnight. The solution was allowed to cool, and the solvent was removed by rotary evaporation. The residual material was triturated with ether, and the resulting precipitate was collected by filtration and dried under vacuum to afford compound 773 (587 mg, 1.35 mmol, 70%).


A similar procedure was used to prepare methyl 2-{3-[4-(3-cyano-6-ethoxy-1-ethyl-1H-indol-2-yl)-phenyl]-ureido}-3-phenyl-propionate (compound 777)


Step B: A solution of ethyl {3-[4-(3-cyano-6-ethoxy-1-ethyl-1H-indol-2-yl)-phenyl]-ureido}-acetate (compound 773, 101 mg, 0.232 mmol) in THF (10 mL) was treated with a solution of potassium tert-butoxide in tert-butanol (0.30 mL, 1.0 M, 0.30 mmol), and the resulting mixture was allowed to stir overnight. The reaction mixture was partitioned between water and ethyl acetate (50 mL each), and the organic phase was washed with saturated brine. The aqueous phases were extracted with more ethyl acetate, and the extracts were combined, dried over anhydrous magnesium sulfate, filtered and evaporated. The residual material was separated by column chromatography (eluting 2/1 ethyl acetate/hexane on silica gel 60) to afford 2-[4-(2,5-dioxo-imidazolidin-1-yl)-phenyl]-6-ethoxy-1-ethyl-1H-indole-3-carbonitrile, compound 779, which was purified further by trituration with ether, collection by filtration and drying under high vacuum (76 mg, 0.196 mmol, 84%).


Example 1AW
Preparation of 2-[4-(2,4-dioxo-imidazolidin-1-yl)phenyl]-6-ethoxy-1-ethyl-1H-indole-3-carbonitrile (compound 776)



embedded image


A solution of 2-(4-aminophenyl)-6-ethoxy-1-ethyl-1H-indole-3-carbonitrile (319 mg, 1.04 mmol) in 1,4-dioxane (3 mL) was treated with chloroacetyl isocyanate (0.10 mL, 1.17 mmol), and the resulting solution was warmed to 60° C. overnight. The solution was cooled, and DBU (0.20 mL, 1.31 mmol) was added. This mixture was stirred at ambient temperature overnight, and then was partitioned between water and ethyl acetate (50 mL each). The organic layer was washed with saturated brine, and then dried over anhydrous magnesium sulfate, filtered and evaporated. The residual material was triturated with ether, and the resulting solid was collected by filtration and dried under high vacuum to afford the title product (319 mg, 0.821 mmol, 79%).


Example 1AX
Preparation of N,N-Dimethyl-2-[4-(3,4-dimethyl-2,5-dioxo-imidazolidin-1-yl)-phenyl]-6-ethoxy-1-ethyl-1H-indole-3-carboxamide (compound 780) and N,N-Dimethyl-6-ethoxy-1-ethyl-2-[4-(3-methyl-2,5-dioxo-imidazolidin-1-yl)-phenyl]-1H-indole-3-carboxamide (compound 781)



embedded image


Step A. A solution of ethyl {3-[4-(3-cyano-6-ethoxy-1-ethyl-1H-indol-2-yl)-phenyl]-ureido} acetate (compound 773, 325 mg, 0.748 mmol), prepared in procedure 1AV, step A, in acetone (5 mL) was treated with HCl (3 mL, 6 N), and the resulting solution was heated to reflux overnight. The reaction mixture was cooled, and the resulting precipitate was collected by filtration, washed with ether and dried under high vacuum to afford the product, 6-ethoxy-1-ethyl-2-[4-(2,5-dioxo-imidazolidin-1-yl)-phenyl]-1H-indole-3-carboxamide (264 mg, 0.650 mmol, 87%).


Step B. Sodium hydride dispersion in mineral oil (75 mg) was washed with a small portion of hexane, and the hexane layer was decanted off. A solution of 6-ethoxy-1-ethyl-2-[4-(2,5-dioxo-imidazolidin-1-yl)-phenyl]-1H-indole-3-carboxamide (190 mg, 0.468 mmol) in dimethylformamide (2 mL) was added, and the mixture was stirred for 1 hour. Then, methyl iodide (0.10 mL, 1.61 mmol) was added by syringe. The resulting mixture was allowed to stir at ambient temperature overnight and then was poured into 50 mL of ethyl acetate. The organic phase was washed with water (3×50 mL) and saturated brine (20 mL), then dried over anhydrous magnesium sulfate, filtered and evaporated. The residual material was separated by column chromatogaphy (1/1 ethyl acetate/hexane, eluting on silica gel 60) to afford the title products, compounds 780 and 781.


Example 1AY
Preparation of N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-N-(2-hydroxyethyl)-methanesulfonamide (compound 828)



embedded image


Step A: Sodium hydride dispersion in mineral oil (108 mg) was washed with a small portion of hexane, and the hexane layer was decanted off. A solution of N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]methanesulfonamide (compound 129, 500 mg, 1.35 mmol) in DMF (5 mL) was slowly added. After gas evolution was complete, 2-bromoethyl acetate (0.30 mL, 2.64 mmol) and sodium iodide (20 mg) were added. The mixture was stirred at ambient temperature overnight, and then was poured into 50 mL of ethyl acetate. This was washed with water (3×50 mL) and saturated brine (20 mL), then dried over anhydrous magnesium sulfate, filtered and evaporated. The residual material was separated by column chromatogaphy (1/1 ethyl acetate/hexane, eluting on silica gel 60) to afford compound 815 (364 mg, 0.799 mmol, 59%).


Step B: A mixture of N-(2-acetoxyethyl)-N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]methanesulfonamide (compound 815, 164 mg, 0.360 mmol) and lithium hydroxide hydrate (45 mg, 1.07 mmol) in 5 mL THF/1 mL water was warmed to 60° C. overnight. The mixture was cooled and poured into ethyl acetate (50 mL). This was washed with water (50 mL) and brine (20 mL), dried over anhydrous magnesium sulfate, filtered and evaporated to afford a solid. The solid was triturated with ether, collected by filtration and dried under high vacuum to afford N-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-N-(2-hydroxyethyl) methanesulfonamide, compound 828 (137 mg, 0.331 mmol, 92%).


Example 1AZ
Preparation of 1-ethyl-6-methoxy-2-[4-(2-methoxyethoxy)-phenyl]-1H-indole-3-carbonitrile (compound 248)



embedded image


1-Ethyl-2-(4-hydroxy-phenyl)-6-methoxy-1H-indole-3-carbonitrile (40 mg, 0.14 mmol), prepared as in example 1Ga step B, was combined with K2CO3 (77 mg, 0.56 mmol), bromoethyl methyl ether (26 μL, 0.28 mmol), and DMF (450 μL). This was stirred at room temperature for 1 hour, and then at 75° C. for 3 hours. The reaction mixture was then partitioned between H2O and EtOAc. The organic layer was dried and concentrated. Purification by silica gel chromatography (CH2Cl2, 0-5% EtOAc) to yield 1-ethyl-6-methoxy-2-[4-(2-methoxyethoxy)-phenyl]-1H-indole-3-carbonitrile (44 mg, 90%) as a white solid.


The following compound was prepared similarly as above: Compound 249.


Example 1BA
Preparation of 1-ethyl-6-methoxy-2-[4-(2-morpholin-4-yl-ethoxy)-phenyl]-1H-indole-3-carbonitrile (compound 261)



embedded image


Step A: 1-Ethyl-6-methoxy-2-[4-(2-hydroxyethoxy)-phenyl]-1H-indole-3-carbonitrile (450 mg, 1.34 mmol), prepared as in example 1AZ, was combined with PPh3 (878 mg, 3.35 mmol) in CH2Cl2 (32 mL) at 0° C. N-bromosuccinimide (600 mg, 3.37 mmol) was added in one portion. The reaction mixture was stirred at room temperature for 30 minutes. The reaction mixture was washed with aqueous NaHCO3. The organic layer was dried and concentrated, and purified by silica gel chromatography (CH2Cl2) to yield 2-[4-(2-bromoethoxy)-phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (506 mg, 95%), compound 253 as a white solid.


Step B: 2-[4-(2-bromoethoxy)-phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (40 mg, 0.1 mmol), prepared as in step A above, was combined with morpholine (50 μL, 0.58 mmol) and acetonitrile (1.0 mL). This was heated at 85° C. for 2 h. The reaction mixture was then partitioned between CH2Cl2 and H2O. The organic layer was dried and concentrated. Purification by silica gel chromatography (6/4, acetone/hexanes) yielded 1-ethyl-6-methoxy-2-[4-(2-morpholin-4-yl-ethoxy)-phenyl]-1H-indole-3-carbonitrile (39 mg, 96%) as a white solid.


The following compounds were prepared similarly as above, using different amines: Compounds 262, 263, 264.


Example 1BB
Preparation of N-{2-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenoxy]-ethyl} methanesulfonamide (compound 268)



embedded image


Step A: 2-[4-(2-Bromoethoxy)phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (258 mg, 0.65 mmol), prepared in example 1BA, step A, was combined with NaN3 (144 mg, 2.2 mmol), and MeOH (3.2 mL). This was heated overnight at 75° C. The reaction mixture was then partitioned between CH2Cl2 and H2O. The organic layer was dried and concentrated. Purification by silica gel chromatography (CH2Cl2) yielded 2-[4-(2-azidoethoxy)phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (187 mg, 80%), compound 266 as a white solid.


Step B: 2-[4-(2-Azidoethoxy)phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (410 mg, 1.14 mmol), prepared as in step A, above, was suspended in a solution of MeOH (20 mL) and concentrated HCl (500 μL). Pd/C (150 mg, 10%) was added, and this mixture was hydrogenated at 30 p.s.i. for 1 h. This was filtered and the filtrate was concentrated. The filtrate residue was partitioned between EtOAc and 0.5N NaOH. The organic layer was dried and concentrated. Purification by silica gel chromatography (10-30%, MeOH/CH2Cl2) yielded 2-[4-(2-aminoethoxy)phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (298 mg, 78%), compound 267, as a white solid.


Step C: 2-[4-(2-Aminoethoxy)phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (30 mg, 0.09 mmol), prepared in step B, above, was dissolved in pyridine (300 μL). Methanesulfonyl chloride (8 μL, 0.1 mmol) was added. This was stirred at room temperature for 45 minutes. More methansulfonyl chloride (4 μL, 0.05 mmol) was added. Stirring continued for another hour. The reaction mixture was partitioned between EtOAc and aqueous HCl. The organic layer was dried and concentrated. Purification by silica gel chromatography (1/1 CH2Cl2/EtOAc) yielded N-{2-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)phenoxy]ethyl} methanesulfonamide, compound 268 (32 mg, 86%) as a white solid.


The following compound was prepared similarly as above: Compound 269.


Example 1BC
Preparation of N-{2-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenoxy]-ethyl} acetamide (compound 274)



embedded image


2-[4-(2-Aminoethoxy)phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (30 mg, 0.09 mmol), prepared as in example 1BB, step B, was dissolved in THF (400 μL), and Et3N (24 μL, 0.17 mmol). Acetyl chloride (10 μL, 0.14 mmol) was added, and the reaction mixture was stirred at room temperature for 2 h. The reaction mixture was partitioned between EtOAc and H2O. The organic layer was dried and concentrated. Purification by silica gel chromatography (EtOAc) yielded N-{2-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)phenoxy]ethyl} acetamide (33 mg, 97%) as a white solid.


Example 1BD
Preparation of 1-{2-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenoxy]ethyl}-3-ethyl-urea (Compound 279)



embedded image


2-[4-(2-Aminoethoxy)phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (30 mg, 0.09 mmol), prepared as in example 1BB, was combined with ethyl isocyanate (18 μL, 0.21 mmol) and pyridine (300 μL). This mixture was stirred at room temperature for 90 minutes, and was then partitioned between EtOAc and aqueous HCl. The organic layer was dried and concentrated. Purification by silica gel chromatography (EtOAc) yielded 1-{2-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenoxy]-ethyl}-3-ethyl-urea (34 mg, 93%) as a white solid.


Example 1BE
Preparation of N-{2-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenoxy]ethyl}formamide (compound 280)



embedded image


Acetic anhydride (700 μL) and 98% formic acid (280 μL) were heated at 65° C. for 1 h. This was cooled to 0° C. 2-[4-(2-Aminoethoxy)phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (30 mg, 0.09 mmol), prepared as in example 1BB, was taken up in THF (400 μL), and added to the mixed anhydride. This was stirred at 0° C. for 45 minutes. The mixture was then portioned between EtOAc and aqueous NaHCO3. The organic layer was dried and concentrated. Purification by silica gel chromatography (4/1, CH2Cl2/acetone) yielded N-{2-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)phenoxy]-ethyl} formamide (28 mg, 86%) as a white solid.


Example 1BF
Preparation of 1-ethyl-2-{4-[2-(3-hydroxypyrrolidin-1-yl)-2-oxo-ethoxy]phenyl}-6-methoxy-1H-indole-3-carbonitrile (compound 285)



embedded image


Step A: 1-Ethyl-2-(4-hydroxyphenyl)-6-methoxy-1H-indole-3-carbonitrile (559 mg, 1.91 mmol), was used to prepare [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenoxy]-acetic acid tert-butyl ester (780 mg, 100%) utilizing essentially the same procedure as example 1AZ.


Step B: [4-(3-Cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenoxy]-acetic acid tert-butyl ester (745 mg, 1.83 mmol) was stirred in 20% TFA in CH2Cl2 at room temperature for 3 hours. This was concentrated and the residue was partitioned between H2O and EtOAc. The organic layer was dried and concentrated. The residue was triturated with CH2Cl2 to yield [4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenoxy]-acetic acid (634 mg, 99%) as a white solid.


Step C: [4-(3-Cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenoxy]-acetic acid (40 mg, 0.12 mmol) was suspended in CH2Cl2 (1.65 mmol) and DMF (2 μL). Oxalyl chloride (17 μL, 0.19 mmol) was added. This was stirred at room temperature for 30 minutes. The resulting solution was then pipetted into a stirring solution of S-3-hydroxypyrrolidine (150 μL) and CH2Cl2 (3.0 mL). The mixture was washed with aqueous HCl. The organic layer was dried and concentrated. Purification by silica gel chromatography (3/2 CH2Cl2/acetone) yielded 1-ethyl-2-{4-[2-(3-hydroxy-pyrrolidin-1-yl)-2-oxo-ethoxy]-phenyl}-6-methoxy-1H-indole-3-carbonitrile (40 mg, 79%), compound 285 as a white solid.


Example 1BG
Preparation of 1-ethyl-6-methoxy-2-(2-oxo-2,3-dihydro-benzooxazol-5-yl)-1H-indole-3-carbonitrile (Compound 332)



embedded image


Step A: 1-Ethyl-2-(4-hydroxy-3-nitrophenyl)-6-methoxy-1H-indole-3-carbonitrile (369 mg, 1.1 mmol), prepared as in example 1Gd, was combined with EtOAc (20 mL) and Pd/C (150 mg, 10%). This mixture was hydrogenated at 30 p.s.i. for 1 h. This was filtered through celite. The filtrate was concentrated and triturated with ether to yield 2-(3-amino-4-hydroxyphenyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (307 mg, 91%), compound 322, as a white solid.


Step B: 2-(3-Amino-4-hydroxyphenyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (100 mg, 0.33 mmol), prepared as in step A, was combined with CDI (83 mg, 0.51 mmol), and THF (1.1 mL). This was heated at 65° C. for 1 hour. The reaction mixture was partitioned between EtOAc and aqueous HCl. The organic layer was dried and concentrated. Purification by silica gel chromatography (9/1, CH2Cl2/EtOAc) yielded 1-ethyl-6-methoxy-2-(2-oxo-2,3-dihydro-benzooxazol-5-yl)-1H-indole-3-carbonitrile (89 mg, 81%) as a white solid.


Example 1BH
Preparation of 1-ethyl-6-methoxy-2-(3-oxo-3,4-dihydro-2H-benzo [1,4]oxazin-6-yl)-1H-indole-3-carbonitrile (compound 334)



embedded image


Step A: Bromoacetic acid (52 mg, 0.37 mmol) was combined with EDCI hydrochloride (62 mg, 0.4 mmol) and acetonitrile (900 μL) to form a homogeneous solution. 2-(3-Amino-4-hydroxyphenyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (100 mg, 0.33 mmol), prepared as in example 1BG, step B, was added to the solution. A thick paste soon formed. Another 1.1 mL of acetonitrile was added and the mixture was then stirred at room temperature for 2 hours. The reaction mixture was then partitioned between H2O and EtOAc. The organic layer was dried and concentrated. Purification by silica gel chromatography (4/1, CH2Cl2/EtOAc) yielded 2-chloro-N-[5-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-2-hydroxyphenyl] acetamide (82 mg, 60%), compound 333, as a white solid.


Step B: 2-Chloro-N-[5-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-2-hydroxy-phenyl] acetamide (57 mg, 0.13 mmol), prepared in step A, was combined with K2CO3 (55 mg, 0.4 mmol), and DMF (400 μL). This was heated at 80° C. for 1 hour. The reaction mixture was then partitioned between H2O and EtOAc. The organic layer was dried and concentrated. Purification by silica gel chromatography (9/1, CH2Cl2/EtOAc) yielded 1-ethyl-6-methoxy-2-(3-oxo-3,4-dihydro-2H-benzo[1,4]oxazin-6-yl)-1H-indole-3-carbonitrile (45 mg, 90%) as a white solid.


Example 1BI
Preparation of 1-ethyl-6-methoxy-2-(2-oxo-2,3-dihydro-benzooxazol-6-yl)-1H-indole-3-carbonitrile (Compound 340)



embedded image


Step A: 4-Aminosalicylic acid (4.0 g, 26 mmol) was suspended in H2SO4 (26 mL, 2.7M) at −5° C. Sodium nitrite (1.8 g, 26.1 mmol) in H2O (6.5 mL) was cooled to ice bath temperature and was added dropwise to the aminosalicylic acid mixture over 5 minutes. The resulting suspension was stirred at −5° C. for 15 minutes. A solution of KI (6.8 g, 41 mmol) in H2SO4 (13 mL, 1M) was added dropwise to the diazonium salt, with considerable evolution of N2. The reaction mixture was heated at 70° C. for 20 minutes. The reaction mixture was then partitioned between H2O and EtOAc. The organic layer was dried and concentrated. Purification by silica gel chromatography (7/3, hexanes/acetone, 1% acetic acid) yielded 4-iodosalicylic acid (5.33 g, 85-90% pure).


Step B: Crude 4-Iodosalicylic acid (1.0 g, 3.8 mmol) was dissolved in THF (28 mL) and Et3N (1.15 mL, 8.2 mmol). DPPA (1.7 mL, 7.8 mmol) was added. This was heated at 70° C. overnight. The reaction mixture was then partitioned between H2O and EtOAc. The organic layer was dried and concentrated. Purification by silica gel chromatography (9/1, CH2Cl2/EtOAc) yielded 472 mg crude intermediate. Trituration with ether yielded 6-iodo-3H-benzooxazol-2-one (369 mg, 37%) as a white solid.


Step C: 6-Iodo-3H-benzooxazol-2-one (118 mg, 0.45 mmol) was used to prepare 1-ethyl-6-methoxy-2-(2-oxo-2,3-dihydro-benzooxazol-6-yl)-1H-indole-3-carbonitrile, compound 340 (75 mg, 55%), utilizing essentially the same procedure as in example 1Gd.


Example 1BJ
Preparation of 1-ethyl-6-methoxy-2-(4-methyl-3-oxo-3,4,-dihydro-2H-benzo[1,4]oxazin-6-yl)-1H-indole-3-carbonitrile (compound 339)



embedded image


1-Ethyl-6-methoxy-2-(3-oxo-3,4-dihydro-2H-benzo [1,4]oxazin-6-yl)-1H-indole-3-carbonitrile (20 mg, 0.058 mmol), prepared as in example 1BH, was combined with NaH (14 mg, 60% suspension in oil, 0.35 mmol). THF (300 μL) was added. This was stirred at room temperature for 5 minutes. A solution of methyl iodide (4.4 μL) in THF (100 μL) was added. This was stirred at room temperature for 1 hour. The reaction mixture was partitioned between EtOAc and aqueous HCl. The organic layer was dried and concentrated. Purification by silica gel chromatography (9/1, CH2Cl2/EtOAc) yielded 1-ethyl-6-methoxy-2-(4-methyl-3-oxo-3,4,-dihydro-2H-benzo[1,4]oxazin-6-yl)-1H-indole-3-carbonitrile (16 mg, 76%) as a white solid.


The following compound was prepared similarly: Compound 341.


Example 1BK
Preparation of 1-ethyl-2-iodo-6-methoxy-5-nitro-1H-indole-3-carbonitrile (compound 499)



embedded image


1-Ethyl-2-iodo-6-methoxy-1H-indole-3-carbonitrile (50 mg, 0.15 mmol), prepared as in example 1Ga, Step A, was suspended in acetic acid (620 μL) at 0° C. Nitric acid (4.25M in AcOH) was added. This was stirred at room temperature for 2 hours. The reaction mixture was then partitioned between CH2Cl2 and H2O. The organic layer was washed with aqueous NaHCO3, and then was dried and concentrated. Purification by silica gel chromatography (6/4, CH2Cl2/hexanes), followed by ether trituration, yielded 1-ethyl-2-iodo-6-methoxy-5-nitro-1H-indole-3-carbonitrile (16 mg, 29%) as a yellow solid.


Example 1BL
Preparation of 1′-ethanesulfonyl-1-ethyl-6-methoxy-2′,3′-dihydro-1H,1H′-[2,6′]biindolyl-3-carbonitrile (compound 753)



embedded image


Step A: 6-Nitroindoline (3.0 g, 18.3 mmol) was dissolved in THF (45 mL) and Et3N (3.4 mL, 24.4 mmol) at 0° C. Acetyl chloride (1.5 mL, 21 mmol) was added dropwise. The mixture was stirred at room temperature for 30 minutes. The mixture was partitioned between EtOAc and aqueous HCl. The organic layer was dried and concentrated to yield 1-acetyl-6-nitroindoline (3.8 g, 100%) as a yellow solid.


Step B: 1-Acetyl-6-nitroindoline (3.8 g, 18.3 mmol) was suspended in EtOAc (200 mL). Pd/C (650 mg, 10%) was added, and the mixture was hydrogenated at 40-55 p.si.i. for 2 hours. The mixture was then filtered through celite. The filtrate was concentrated, and the residue was triturated with ether to yield 1-acetyl-6-aminoindoline (3.18 g, 99%) as an orange solid.


Step C: 1-Acetyl-6-aminoindoline (1.5 g, 8.5 mmol) was used to prepare 1-acetyl-6-iodoindoline (1.06 g, 43%), utilizing essentially the same procedure in example 1BI, Step A.


Step D: 1-Acetyl-6-iodoindoline (1.06 g, 3.7 mmol), NaOH (1.16 g, 29 mmol), EtOH (8 mL), and H2O (6 mL) were heated at 90° C. overnight. The reaction mixture was then partitioned between H2O and EtOAc. The organic layer was extracted into aqueous HCl. The aqueous layer was in turn basified with NaOH, and was extracted with EtOAc. The organic layer was dried and concentrated. Hexane trituration yielded 6-iodoindoline (577 mg, 64%) as a brown solid.


Step E: 1-Iodoindoline (600 mg, 2.45 mmol) was used to prepare 1-ethyl-6-methoxy-2′,3′-dihydro-1H,1H′-[2,6′]biindolyl-3-carbonitrile (535 mg, 67%), utilizing essentially the same procedure as in example 1Gd, Step B.


Step F: 1-Ethyl-6-methoxy-2′,3′-dihydro-1H,1H′-[2,6′]biindolyl-3-carbonitrile (30 mg, 0.095 mmol) was used to prepare 1′-Ethanesulfonyl-1-Ethyl-6-methoxy-2′,3′-dihydro-1H,1H′-[2,6′]biindolyl-3-carbonitrile (24 mg, 62%), utilizing the procedure in example 1Y.


The following compounds were prepared similarly as above: Compounds 752 and 754.


Example 1BM
Preparation of 5-acetyl-1-ethyl-6-methoxy-2-(4-nitro-phenyl)-1H-indole-3-carbonitrile (compound 844)



embedded image


1-Ethyl-6-methoxy-2-(4-nitrophenyl)-1H-indole-3-carbonitrile (100 mg, 0.3 mmol), prepared by the method of example 1Gc was suspended in 1,2-dichloroethane (500 mL) at 0° C. Acetyl chloride (50 μL, 0.69 mmol) was added, followed by AlCl3 (55 mg, 0.4 mmol) in one portion. This was stirred at 0° C. for 1 hour, at room temperature for 4 hours, and at 45° C. overnight. The reaction mixture was then partitioned between CH2Cl2 and H2O. The organic layer was dried and concentrated. Purification by silica gel chromatography (195:5 CH2Cl2/EtOAc) yielded 5-acetyl-1-ethyl-6-methoxy-2-(4-nitro-phenyl)-1H-indole-3-carbonitrile (33 mg, 29%) as an orange solid.


Example 1BN
Preparation of 1-ethyl-6-methoxy-5-morpholin-4-ylmethyl-2-(4-nitro-phenyl)-1H-indole-3-carbonitrile (compound 845)



embedded image


Step A: 1-Ethyl-6-methoxy-2-(4-nitrophenyl)-1H-indole-3-carbonitrile (100 mg, 0.3 mmol), prepared by the method of example 1Gc, was combined with 1,3,5-trioxane (64 mg, 0.71 mmol) and acetic acid (2.0 mL). 33% HBr in acetic acid (2.0 mL) was added. This was stirred at room temperature for 4 hours. The reaction mixture was then partitioned between CH2Cl2 and H2O. The organic layer was washed with aqueous NaHCO3, and was subsequently dried and concentrated. The crude material was carried through to the next step.


Step B: Crude 6-bromomethyl-1-ethyl-6-methoxy-2-(4-nitro-phenyl)-1H-indole-3-carbonitrile (0.3 mmol) was heated with morpholine (150 μL, 1.75 mmol) and DCE (1.0 mL) at 90° C. overnight. The reaction mixture was then partitioned between H2O and EtOAc. The organic layer was dried and concentrated. Purification by silica gel chromatography (50-100%, EtOAc/CH2Cl2), followed by trituration with 1/1 hexane/acetone yielded 1-ethyl-6-methoxy-5-morpholin-4-ylmethyl-2-(4-nitrophenyl)-1H-indole-3-carbonitrile (57 mg, 44% overall yield) as a yellow solid.


Example 1BO
2-[4-(1,1-dioxidoisothiazolidin-2-yl)phenyl]-1-cyclopropylmethyl-6-methoxy-1H-indole-3-carbonitrile (compound 716)



embedded image


Step A: To a solution of 6-methoxyindole (5.88 g, 40.0 mmol) and di-tert-butyl dicarbonate (9.59 g, 44.0 mmol) in DCM (50 mL) was added, at 40° C. while stirring, DMAP (0.10 g). After stirring overnight, the mixture was washed sequentially with 0.1 N HCl, water and brine and dried over anhydrous Na2SO4. The solvent was evaporated and the residue was chromatographed (silica gel, EtOAc/hexanes, 1/7) to provide tert-butyl 6-methoxy-1H-indole-1-carboxylate (8.48 g, 86%).


Step B: The above Boc-indole (3.08 g, 12.5 mmol) and isopropylborate (4.83 mL, 21.9 mmol) were dissolved in anhydrous THF (20 mL) and the solution was cooled at 0° C. While stirring, LDA (12.5 mL, 1.5 M mono-THF complex in cyclohexane, 18.7 mmol) was added dropwise. The mixture was stirred at 0° C. for 15 min and then room temperature for 0.5 hr, followed by the addition of HCl (6 N, 3.0 mL, 18 mmol) in a ice-water bath. The organic solvent was removed in vacuo and the residue was suspended in H2O (100 mL) and acidified with HCl (6 N) to pH 4˜5. The precipitate was collected via filtration and washed with water and hexanes and dried in air to provide 1-Boc-6-mehoxyindole-2-boronic acid (3.38 g, 93%).


Step C: To a solution of 4-iodoanilline (3.18 g, 14.5 mmol) in pyridine (15 mL) at 0° C., was added 3-chloropropanesulfonyl chloride (2.3 mL, 18.9 mmol). After the addition, the mixture was stirred for 2 hr at room temperature, and poured into ice-water (200 mL). The organic was separated and the aqueous layer was extracted with DCM (2×50 mL). The combined organics were washed with HCl (2 N, 2×15 mL), water (2×50 mL) and brine (20 mL) consecutively and dried over anhydrous Na2SO4. The solvent was then evaporated and the residue was chromatographed to furnish 3-chloro-N-(4-iodophenyl)propane-1-sulfonamide (4.68 g, 90%). The chlorosulfonamide obtained (3.47 g, 9.6 mmol) was then treated with K2CO3 (3.33 g, 24.1 mmol) in DMF (50 mL) at 50° C. for 2 hr. The mixture was poured into ice-water (300 mL) and the precipitate was collected and dried in air to provide essentially pure 2-(4-iodophenyl)isothiazolidine-1,1-dioxide (3.11 g, 100%).


Step D: To a mixture of 1-Boc-6-mehoxyindole-2-boronic acid prepared in step B above (0.36 g, 1.25 mmol), 2-(4-iodophenyl)isothiazolidine-1,1-dioxide (0.32 g, 1.0 mmol) and PdCl2(dppf) (0.037 g, 0.05 mmol) in DMF (4.0 mL), was added aqueous K2CO3 solution (1.5 mL, 2.0 M, 3.0 mmol). The mixture was stirred at room temperature overnight and then poured into ice-water (100 mL). The precipitate was collected and washed with water and purified by flash column chromatography (silica gel, DCM/EtOAc, 9/1) to furnish 1-Boc-2-[4-(1,1-dioxidoisothiazolidin-2-yl)phenyl]-6-methoxy-1H-indole (0.43 g, 98%).


The following compound was made similarly: Compound 768


Step D: 1-Boc-2-[4-(1,1-dioxidoisothiazolidin-2-yl)phenyl]-6-methoxy-1H-indole (1.63 g, 3.7 mmol) was treated with TFA (25 mL) in DCM (25 mL) at room temperature for 4 hr. After the removal of the volatiles, the residue was carefully stirred with saturated NaHCO3 for 0.5 hr. The precipitate was collected via filtration and washed with water thoroughly and dried to provide essentially pure 1-H-2-[4-(1,1-dioxidoisothiazolidin-2-yl)phenyl]-6-methoxyindole (1.17 g, 92%).


Step E: At 0° C., 1-H-2-[4-(1,1-dioxidoisothiazolidin-2-yl)phenyl]-6-methoxyindole (0.95 g, 2.8 mmol) was dissolved in DMF (10 mL) and treated with chlorosulfonyl isocyanate (0.36 mL, 4.2 mmol). The mixture was then stirred at room temperature overnight and poured into ice-water (150 mL) then stirred for 0.5 hr. The precipitate was collected via filtration and washed thoroughly with water and dried in air to furnish 1-H-2-[4-(1,1-dioxidoisothiazolidin-2-yl)phenyl]-6-methoxyindole-3-carbonitrile (0.89 g, 87%).


The following compound was prepared in the same fashion as described above: Compound 829


Step F: To a solution of 1-H-2-[4-(1,1-dioxidoisothiazolidin-2-yl)phenyl]-6-methoxyindole-3-carbonitrile (73 mg, 0.2 mmol) and K2CO3 (69 mg, 0.5 mmol) in DMF (3.0 mL) was added cyclopropylmethyl iodide (0.029 mL, 0.3 mmol). The mixture was stirred at 50° C. overnight and poured into ice-water (10 mL). The precipitate was collected via filtration, washed with water and purified by column chromatography to provide 2-[4-(1,1-dioxidoisothiazolidin-2-yl)phenyl]-6-methoxy-1-cyclopropylmethylindole-3-carbonitrile, compound 716 (73 mg, 87%).


The following compounds were prepared in the same fashion as described above: Compounds 717, 718, 719, 782, 783, 784.


Example 1BP
Preparation of 2-[4-(1,1′-dioxo-1λ6-isothiazolidin-2-yl)-6-methoxy-3-oxazol-5-yl-1-propyl-1H-indole (compound 805)



embedded image


Step A: 2-[4-(1,1′-Dioxo-1λ6-isothiazolidin-2-yl)-6-methoxy-indole (900 mg, 2.62 mmol), prepared in example 1BO, step D was used to prepare 2-[4-(1,1′-dioxo-1λ6-isothiazolidin-2-yl)-6-methoxy-1-propyl-1H-indole (608 mg, 60%), utilizing essentially the same procedure as example 1A, Step B.


Step B: 2-[4-(1,1′-Dioxo-1λ6-isothiazolidin-2-yl)-6-methoxy-1-propyl-1H-indole (50 mg, 0.13 mmol) was used to prepare 2-[4-(1,1′-dioxo-1λ6-isothiazolidin-2-yl)-6-methoxy-3-oxazol-5-yl-1-propyl-1H-indole (9 mg, 15% overall yield) according to the protocol in example 1P.


Example BQ
Preparation of 2-[4-(cyclopropylsulfonyl)piperazin-1-yl]-1-ethyl-6-(trifluoromethyl)-1H-indole-3-carbonitrile (compound 842)



embedded image


Step A: To a solution of 1-ethyl-6-trifluoromethylindole-3-carbonitrile (2.54 g, 10.0 mmol), prepared by the method of procedure 1A, in anhydrous THF (20.0 mL), at −78° C. was added LDA (8.3 mL, 1.5 M mono-THF in cyclohexane, 12.5 mmol) dropwise. The mixture was continued for 0.5 hr after the addition, followed by the addition of hexachloroethane and the mixture was then brought to room temperature slowly and stirred for 0.5 hr. The solvent was then evaporated and the residue was treated with water. The organics were extracted with dichloromethane, washed with water and brine and dried over anhydrous Na2SO4. The crude product obtained after the removal of the solvent was chromatographed (silica gel, dichloromethane/hexanes, 3 /2) to provide 2-chloro-1-ethyl-6-(trifluoromethyl)-1H-indole-3-carbonitrile (1.75 g, 64%).


Step B: The chloroindole obtained above (0.27 g, 1.0 mmol), K2CO3 (0.35 g, 2.5 mmol) and N-Boc-piperazine (0.28 g, 1.5 mmol) were stirred at 70° C. in DMF (5.0 mL) for 3 days and then poured into water (50 mL). The precipitate was collected via filtration and washed with water. Chromatography of this crude product (silica gel, dichloromethane/ethyl acetate, 9/1) provided 4-(3-cyano-1-ethyl-6-trifluoromethyl-1H-indol-2-yl)-piperazine-1-carboxylic acid tert-butyl ester, compound 785 (0.30 g, 71%).


The following compound was prepared in the same fashion as described above, by using other amines: Compounds 514, 785, 786.


Step C: 4-(3-cyano-1-ethyl-6-trifluoromethyl-1H-indol-2-yl)-piperazine-1-carboxylic acid tert-butyl ester (0.26 g, 6.1 mmol) was treated with TFA (5 mL) in dichloromethane (5 mL) for 1 hr at room temperature. After the removal of the volatiles, the residue was treated with saturated NaHCO3 and the precipitate was collected via filtration, washed with water thoroughly and dried in air to furnish essentially pure 1-ethyl-2-piperazin-1-yl-6-(trifluoromethyl)-1H-indole-3-carbonitrile (0.20 g, 100%).


Step D: To a solution of 1-ethyl-2-piperazin-1-yl-6-(trifluoromethyl)-1H-indole-3-carbonitrile (32 mg, 0.1 mmol), pyridine (0.1 mL) in dichloromethaene (1.0 mL) was added cyclopropanesulfonyl chloride (28 mg, 0.2 mmol) and the mixture was stirred at room temperature overnight. This was then diluted with dichloromethane (5 mL), washed with HCl (2 N, 2×2 mL), water (2×5 mL) and brine (5 mL) and chromatographed over silica gel (dichloromethane/ethyl acetate, 9/1) to provide 2-[4-(cyclopropylsulfonyl)piperazin-1-yl]-1-ethyl-6-(trifluoromethyl)-1H-indole-3-carbonitrile, compound 842 (30 mg, 70%).


The following compounds were prepared in the same fashion as described above, using corresponding sulfonyl chlorides: Compounds 841, 843.


Example 1BR
Ethanesulfonic acid [3-cyano-2-(4-ethoxyphenyl)-1-ethyl-1H-indol-6-yl]-amide (compound 835)



embedded image


Step A: 6-Bromo-2-(4-ethoxyphenyl)-1-ethyl-1H-indole-3-carbonitrile (0.74 g, 2.0 mmol), compound 831, prepared from 6-bromoindole as described in example 1Gb, was mixed with K2CO3 (0.55 g, 4.0 mmol), CuI (0.02 g, 0.1 mmol), tert-butyl carbamate (0.35 g, 3.0 mmol), N,N′-dimethylcyclohexane-1,2-diamine ligand (0.028 g, 0.2 mmol) and anhydrous toluene (5.0 mL) in a sealed tube. The reaction system was flushed with nitrogen and then stirred at 110° C. overnight. After cooling, the solvent was replaced with dichloromethane and chromatographed (silica gel, dichloromethane) to provide [3-cyano-2-(4-ethoxy-phenyl)-1-ethyl-1H-indol-6-yl]-carbamic acid tert-butyl ester (0.68 g, 84%), compound 832.


Step B: Compound 832 prepared in step A above (0.63 g, 1.56 mmol) was treated with TFA/DCM (7.5 mL/7.5 mL) at room temperature for 2 hr, and the volatiles were removed in vacuum. The residue was treated with saturated NaHCO3 and the precipitate was collected via filtration and washed thoroughly with water, dried in air to provide 6-amino-2-(4-ethoxyphenyl)-1-ethyl-1H-indole-3-carbonitrile (0.45 g, 96%), compound 833.


Step C: The above amine (31 mg, 0.1 mmol) was treated with ethanesulfonyl chloride (19 mg, 0.15 mmol) in pyridine (1.0 mL) at room temperature overnight to provide, after purification using column chromatography, ethanesulfonic acid [3-cyano-2-(4-ethoxy-phenyl)-1-ethyl-1H-indol-6-yl]-amide (83%), compound 835.


The following compound was prepared in the same fashion as described above: Compounds 830, 834, 836 and 837.


Example 1BS
Preparation of [3-cyano-2-(4-ethoxyphenyl)-1-ethyl-1H-indol-6-yl]-carbamic acid ethyl ester (compound 838)



embedded image


6-Amino-2-(4-ethoxyphenyl)-1-ethyl-1H-indole-3-carbonitrile (31 mg, 0.1 mmol), compound 833, prepared in example 1BR, step B was treated with ethyl chloroformate (16 mg, 0.15 mmol) in pyridine (1.0 mL) at room temperature overnight to furnish, after purification using column chromatography [3-cyano-2-(4-ethoxyphenyl)-1-ethyl-1H-indol-6-yl]-carbamic acid ethyl ester (30 mg, 79%).


Example 1BT
Preparation of 1-[3-cyano-2-(4-ethoxyphenyl)-1-ethyl-1H-indol-6-yl]-3-ethyl-urea (compound 839)



embedded image


6-Amino-2-(4-ethoxyphenyl)-1-ethyl-1H-indole-3-carbonitrile (31 mg, 0.1 mmol) was treated with ethyl isocyanate (14 mg, 0.2 mmol) in dichloromethane (1.0 mL) at 40° C. overnight. The precipitate was collected via filtration, washed with dichloromethane an dried in air to furnish, 1-[3-cyano-2-(4-ethoxy-phenyl)-1-ethyl-1H-indol-6-yl]-3-ethyl-urea (36 mg, 95%).


Example 1BU
Preparation of 1-(2-chloroethyl)-3-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-urea (compound 442)



embedded image


To solution of 2-(4-aminophenyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (50 mg, 0.172 mmole) in THF (2 mL) was added 2-chloroethyl isocyanate (22 uL, 0.258 mmole) at room temperature. After stirring overnight at reflux, the reaction mixture was concentrated in vacuo and the residue was diluted with ethyl acetate. The resulting semi-solid was triturated with hexane and the precipitate collected was collected by filtration and washed well with 50% ethyl acetate in hexane and dried in vacuo to afford (62 mg, 91%) of 1-(2-chloroethyl)-3-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-urea.


Utilizing essentially the same procedure, the following compounds were prepared: Compounds 295, 362, 395, 396, 397, 398, 399, 400, 401, 402, 403, 404, 405, 406, 407, 443, 444, 445, 446, 511, 512, 513, 600, 620, 626, 627, 628, 679, 680, 681, 740, 741, 742, 743, 748, 749, 750, 751, 774, 817, 818, 846, 847, 848.


Example 1BV
Preparation of 1-ethyl-6-methoxy-2-[4-(2-oxo-imidazolidin-1-yl)-phenyl]-1H-indole-3-carbonitrile (compound 771)



embedded image


To a solution of 1-(2-chloroethyl)-3-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-urea (100 mg, 0.252 mmol) in MeOH (10 mL) was added aqueous 1M KOH (504 uL) and then stirred at 49° C. for 24 h. The solvents were removed under reduced pressure. The residue was diluted with ethyl acetate and then washed with water. The organic layer was dried over anhydrous MgSO4, filtered and concentrated under reduced pressure. The residue was diluted with ethyl acetate and then triturated with hexane and the precipitate collected by filtration and washed well with 50% ethyl acetate in hexane and dried in vacuo to afford 1-ethyl-6-methoxy-2-[4-(2-oxo-imidazolidin-1-yl)-phenyl]-1H-indole-3-carbonitrile (56 mg, 62%).


Using essentially the same procedure, the following compounds were prepared: Compounds 770, 778


Example 1BW
Preparation of 1-ethyl-6-isopropoxy-2-[4-(2-oxo-oxazolidin-3-yl)-phenyl]-1H-indole-3-carbonitrile (compound 638)



embedded image


To a solution of [4-(3-cyano-1-ethyl-6-isopropoxy-1H-indol-2-yl)-phenyl]-carbamic acid 2-chloro-ethyl ester (30 mg, 0.07 mmol) in DMF (1 mL) was added aqueous K2CO3 (10 mg) and then stirred at 50° C. for 18 h. The reaction mixture was poured into cold water and the precipitate collected by filtration and washed with hexane and dried in vacuo to afford the title compound (21 mg, 81%).


The following compounds were made in similar fashion: Compounds 820, 821, 863, 864.


Example 1BX
Preparation of {3-[3-cyano-1-ethyl-6-(3-pyrrolidin-1-yl-propoxy)-1H-indol-2-yl]-phenyl}-carbamic acid ethyl ester (compound 530)



embedded image


Step A: To a solution of [3-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-carbamic acid ethyl ester (1.65 g, 4.37 mmole) in DCM (20 mL) was added 1M BBr3 in DCM (13.12 mL) over a period of 20 min. The reaction mixture was stirred further 1 h at room temperature and then the solvents were removed under reduced pressure. The residue was dissolved in MeOH and then poured into cold water. The precipitate was collected by filtration and washed with hexane and dried in vacuo to afford [3-(3-cyano-1-ethyl-6-hydroxy-1H-indol-2-yl)-phenyl]-carbamic acid ethyl ester (1.5 g, 98%).


Step B: To a solution of [3-(3-cyano-1-ethyl-6-hydroxy-1H-indol-2-yl)-phenyl]-carbamic acid ethyl ester (1.2 g, 2.91 mmol) in DMF (10 mL) was added K2CO3 (538 mg, 3.9 mmole) and 3-bromo-1-chloroproane (383 uL, 3.9 mmole) and the reaction was stirred for overnight at 50° C. The reaction mixture was then poured into cold water and the precipitate was collected by filtration and washed with hexane and dried in vacuo to afford 1.1 g, 89% of the desired product.


Step C: To a solution of {3-[3-cyano-1-ethyl-6-(3-pyrrolidin-1-yl-propoxy)-1H-indol-2-yl]-phenyl}-carbamic acid ethyl ester (50 mg, 0.12 mmole) in CH3CN (2 mL) was added DIEA (31 uL, 0.18 mmol), sodium iodide (20 mg, 0.132 mmol) and pyrrolidine (30 uL, 0.36 mmole). The resulting mixture was stirred at reflux temperature for overnight. The solvent was evaporated and the residue was diluted with ethyl acetate and then triturated with hexane and the precipitate collected by filtration and washed well with 50% ethyl acetate in hexane and dried in vacuo to afford 1-ethyl-6-isopropoxy-2-[4-(2-oxo-oxazolidin-3-yl)-phenyl]-1H-indole-3-carbonitrile, compound 638 (46 mg, 85%).


The following compounds were made in similar fashion following steps A-C, above: Compounds 441, 447, 491, 492, 493, 504, 525, 526, 527, 528, 529, 531, 532, 533, 534, 535, 536, 537, 538, 539


Example 1BY
Preparation of [3-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-thiourea (Compound 767)



embedded image


Step A: The starting material 2-(3-amino-phenyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (187 mg, 0.642 mmol) was dissolved in anhydrous acetone (3.0 mL). Benzoyl isothiocyanate (107 mg, 0.656 mmol) was added to the solution at room temperature and the mixture was stirred for 17 h during which time a precipitate had formed. The precipitate was filtered, washed with acetone and dried to give 264 mg of 1-benzoyl-3-[3-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-thiourea (90% yield) as a light yellow solid.


Step B: A suspension of 1-benzoyl-3-[3-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-thiourea (241 mg, 0.530 mmol) in methyl alcohol (2.0 ml) and water (0.5 mL) was stirred at room temperature as sodium hydroxide (31 mg, 0.78 mmol) was added. The reaction mixture was heated to 50° C. for 17 h. The reaction mixture was concentrated to remove methyl alcohol. Water was added to the mixture and the solid was filtered, washed with water and dried to give 179 mg of [3-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)-phenyl]-thiourea, compound 767 (96% yield) as a white solid.


Example 1BZ
Preparation of 1-ethyl-6-methoxy-2-[4-(2-phenylquinazolin-4-ylamino)-phenyl]-1H-indole-3-carbonitrile (Compound 458)



embedded image


A solution of 2-(4-aminophenyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (100 mg, 0.343 mmol), 4-chloro-2-phenyl-quinazoline (83 mg, 0.34 mmol) and diisopropylethylamine (0.10 mL, 0.57 mmol) in absolute ethanol (3 mL) was heated to reflux overnight. The solution was cooled and evaporated, and the residue taken up in ethyl acetate (50 mL). This was washed with water and saturated brine (50 mL each), then dried over anhydrous sodium sulfate, filtered and evaporated. The resulting solid was triturated with ether, collected by filtration and dried under vacuum to afford 1-ethyl-6-methoxy-2-[4-(2-phenylquinazolin-4-ylamino)-phenyl]-1H-indole-3-carbonitrile (139 mg, 0.280 mmol, 82%).


Example 1CA
Preparation of diethyl [4-(3-cyano-6-ethoxy-1-ethyl-1H-indol-2-yl)-phenyl]-phosphoramidate (compound 772)



embedded image


A solution of 2-(4-aminophenyl)-6-ethoxy-1-ethyl-1H-indole-3-carbonitrile (148 mg, 0.484 mmol), diethyl chlorophosphate (0.086 mL, 0.58 mmol) and diisopropylethylamine (0.10 mL, 0.57 mmol) in 1,4-dioxane (5 mL) was stirred at ambient temperature for 12 hours, then heated to 80° C. for an additional 24 hours. The solution was cooled and poured into 50 mL of ethyl acetate. This was washed with water and saturated brine (50 mL each), then dried over anhydrous magnesium sulfate, filtered and evaporated. The residual material was separated by flash chromatography (eluting 2/1 ethyl acetate/hexane on silica gel 60) to afford diethyl [4-(3-cyano-6-ethoxy-1-ethyl-1H-indol-2-yl)-phenyl]-phosphoramidate (108 mg, 0.245 mmol, 51%) as a white powder after evaporation.


Example 1CB
Preparation of 1-ethyl-6-methoxy-2-[4-(5-methyl-1,1-dioxo-1λ6-[1,2,5]thiadiazolidin-2-yl)-phenyl]-1H-indole-3-carbonitrile (compound 726)



embedded image


Step A: To a solution of 2-(4-aminophenyl)-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (202 mg, 0.693 mmol) in pyridine (2.0 mL) was added the N-β-(chloroethylamino)sulfonyl chloride (222 mg, 1.39 mmol). The mixture was stirred at room temperature for 17 h then water (12.0 mL) was added and the mixture was extracted with ethyl acetate (3×2 mL). The extract was washed with 10% aqueous HCl (2×2 mL), water (2×2 mL), dried over MgSO4, filtered and concentrated on a rotary evaporator. The crude product was purified by flash chromatography (0-5%, ethyl acetate/methylene chloride) to give 217 mg of N-(2-chloro-ethyl)-N′-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)phenyl] sulfamide, compound 724, as a tan solid (75% yield).


In similar fashion the following compounds were prepared: Compounds 540, 541, 542, 574, 576, 704


Step B: To a solution of N-(2-chloro-ethyl)-N′-[4-(3-cyano-1-ethyl-6-methoxy-1H-indol-2-yl)phenyl] sulfamide (100 mg, 0.241 mmol) in anhydrous DMF (1.25 mL), was added potassium carbonate (71.0 mg, 0.514 mmol). The mixture was stirred at room temperature for 17 h, then diluted with water (7.5 mL). The reaction mixture was extracted with ethyl acetate (3×2 mL) and the extract was washed with water (2×2 mL), dried over MgSO4 and concentrated to give 2-[4-(1,1-dioxo-1λ6-[1,2,5]thiadiazolidin-2-yl)phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile, compound 725, as a white solid (84 mg, 88% yield).


In similar fashion the following compound was prepared: Compound: 705


Step C: To a solution of 2-[4-(1,1-dioxo-1λ6-[1,2,5]thiadiazolidin-2-yl)phenyl]-1-ethyl-6-methoxy-1H-indole-3-carbonitrile (34 mg, 0.086 mmol) in anhydrous DMF (1.0 mL) was added potassium carbonate (25 mg, 0.18 mmol) and iodomethane (20.4 mg, 0.144 mmol). The mixture was stirred at room temperature for 2 h. and then diluted with water (6.0 mL) to give a precipitate. The precipitate was filtered, washed with water and dried to give 1-ethyl-6-methoxy-2-[4-(5-methyl-1,1-dioxo-1λ6-[1,2,5]thiadiazolidin-2-yl)-phenyl]-1H-indole-3-carbonitrile, compound 726, as a white solid (35 mg, 98% yield).


In similar fashion the following compound was prepared: Compound 727.


Example 2
Screening of Low Molecular Weight Compounds Using a Cell-Based HCV IRES Monocistronic Translation Assay

Chemical libraries are screened using a cell-based monocistronic HCV IRES-regulated translation assay designed to closely mimic natural HCV mRNA translation and then compound analogs are made based on hits in the chemical libraries and screened as well. A DNA construct is prepared, termed pHCVIRESmono, in which HCV IRES sequences (HCV 2b, nucleotides 18-347) are inserted between a promoter and the firefly luciferase (Fluc) reporter gene. A stably transfected HepG 2 (hepatoblastoma) cell line (termed HepGmono-4) or a Huh7 cell line (termed Huhmono 7), or a Hela-cell line (termed Helamono), are established by transfection with the pHCVIRESmono DNA by selecting for resistance to hygromycin.


Example 3
Determination of Selectivity for HCV IRES-Regulated Translation Using the Cell-Based Cap-Dependent Translation Assays

Since translation assays are used to screen HCV IRES inhibitors, the selected hits may specifically act on HCV IRES-driven translation or may modulate general protein synthesis in mammalian cells. The compounds that act on general translation will most likely have significant toxicity. To address this possibility, various cell-based cap-dependent translation assays are established for the further evaluation of all selected compounds. Plasmid DNAs containing 130 nucleotides of vector sequence 5′ to Fluc are constructed. This construct is referred to herein as pLuc. A stable cell line is established in cap-dependent translation assays using 293T cells (a human embryonic kidney cell line). HepGmono-4 and pLuc are treated with compound for 20 hours and activity is determined by quantifying the Fluc signal. A five-fold selectivity between the HCV IRES and cap-dependent translation is considered to be desirable. For example, using these cell-based cap-dependent translation assays, Applicants identified compounds that showed IC50 values that were at least 5-fold greater in the cap-dependent translation assays than in the HCV IRES translation assay. FIG. 1 shows an example of a hit that was selective against HCV IRES-regulated translation over cap-dependent pLuc translation. Importantly, the compound had the same level of activity in an HCV IRES monocistronic 293T cell line as in HepGmono-4 (data not shown). It is thus unlikely that the selectivity of the compounds between HepGmono-4 (HepG 2) and the cap-dependent translations (293T) is due to the different cell types used.


Additionally, western blotting assays are used to further demonstrate that the compounds selectively inhibit HCV IRES-driven translation. Both HepGmono-4 and pLuc cells are treated with the compounds as described above, following treatment with the test compounds for 20 hours, cells are collected and lysed in Laminin buffer containing 0.5% SDS. Proteins are separated on a 10% SDS-PAGE, then transferred onto a nitrocellulose membrane, and blotted using antibodies against Fluc (RDI) and β-actin (Oncogene). For example, some of the compounds of the present invention were tested in this manner and as expected, the compounds that selectively inhibited HCV IRES-driven translation in assays using Fluc signal as an end point showed comparable reductions of the luciferase reporter protein levels in HepGmono-4 cells and were relatively inactive against pLuc in the Western blot (data not shown). Importantly, these compounds did not inhibit the expression of endogenous β-actin, the translation of which is cap-dependent in both cell lines. Consistently, compounds that did not show selectivity in the translation assays inhibited protein accumulation in both the HCV IRES and cap-dependent translation assays (data not shown). As expected, the general protein translation inhibitor puromycin also inhibited both the HCV IRES-driven and cap-dependent protein production (data not shown). Therefore, the Western blot results confirm that the compounds of the present invention selectively inhibit HCV IRES-driven translation.


Testing conditions for these cell lines are optimized and the effects of mRNA level on activity of the compounds are controlled by quantitating Fluc mRNA levels by RT real-time PCR. For example, some of the compounds of the present invention were tested in this manner, and no significant differences in Fluc mRNA levels were observed between the HepGmono-4, or the Helamono cells, or the Huhmono cells, and cap-dependent translation cell lines used (data not shown).


Example 4
Evaluation of the Selectivity for HCV IRES-Driven Translation Using Cellular IRES-Mediated Translation Assays

A number of human mRNAs have been shown to harbor IRES elements (18, 19, 39, 44, 45, 91, 126, 130). Although the primary sequences and secondary structures of the HCV IRES are different from those of cellular IRESs, an important test for selectivity is to determine whether the selected compounds are active against cellular IRESs. The VEGF IRES has poor initiation activity in in vitro assays, but demonstrates substantial activity in cell-based translation assays (18, 45). For example, some of the compounds of the present invention were tested and all of the compounds that had good selectivity with respect to cap-dependent translation exhibited at least 5-fold higher IC50 values against the VEGF IRES than against the HCV IRES (data not shown). These data indicate that the selected compounds have selectivity against viral IRES. In addition to having different structures, the VEGF IRES also have different interactions with non-canonical cellular translation factors. These differences may contribute to the selectivity of the HCV IRES inhibitors that we have identified.


Example 5
Evaluation of Cytotoxicity

Effects on cell proliferation are a critical issue for any drug discovery effort. Therefore, a cell proliferation/cytotoxicity assay is used to eliminate any compounds that affect mammalian cell growth. The effects of the selected hits on cell proliferation are tested in human cell lines 293 T and Huh7 (a human hepatoblastoma cell line). Cells are grown in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum, L-glutamine, penicillin, and streptomycin. Cells in log phase are treated with test compounds for three days, with 250 μM being the highest concentration of test compound used. The effect of the compounds on cell proliferation is assessed by using the CellTiter 96 AQueous One Solution Cell Proliferation Assay (Promega, Madison, Wis.). Compounds that have at least 5-fold higher CC50 values relative to IC50 values in HepGmono-4 are considered to have a sufficient window between activity and cytotoxicity and, hence, are selected for further evaluation. For example, some of the compounds of the present invention were tested in this manner, and importantly, all compounds that had good selectivity with respect to cap-dependent translation also demonstrated a greater than 5-fold ratio of CC50 to IC50 values.


Example 6
Evaluation of the Efficacy of the Compounds in the HCV Replicon System

The lack of reliable and readily accessible cell-culture and small animal models permissive for HCV replication has limited the development of new anti-HCV agents. Self-replicating subgenomic HCV systems, termed HCV replicons, have recently been described and have been widely used to assess the efficacy of anti-HCV inhibitors (8, 70, 104). Interferon (IFN) α and inhibitors of the HCV protease and polymerase have been reported to be active in the HCV replicon system (8, 17, 32, 68, 69, 117).


HCV replicons that include bicistronic and monocistronic systems are identified and assays for testing the HCV IRES inhibitors are established. In the bicistronic replicons, the HCV IRES directs the expression of the selective marker (Neo and/or a Fluc reporter), and the EMCV IRES mediates the expression of viral non-structural proteins. In the monocistronic replicon, the HCV IRES directly mediates viral protein synthesis. The HCV IRES inhibitors are analyzed in the bicistronic replicon by quantitating the Fluc reporter signal. Replicon-containing cells are cultured with the compounds of the invention for 2 days. Interferon (IFN) α is used as a positive control. For example, the compounds of the present invention were tested in this manner, and the experiments showed that compounds that selectively inhibited HCV IRES-mediated translation inhibited Fluc expression in the bicistronic replicon.


In the following table (Table 1),

    • * =Replicon or HCV-PV IC50>2μM
    • ** =Replicon or HCV-PV IC50 between 0.5 μM and 2 μM
    • *** =Replicon or HCV-PV IC50<0.5 μM
  • Replicon IC50 values are determined by firefiy luciferase signal.
  • HCV-PV IC50 values are determined by viral RNA reduction.














TABLE 1







Mass





Compound
Melting
Spec
HCV-PV
Replicon



Number
Point (° C.)
[M+H]
IC50 μM
IC5Os μM
NMR Data




















1



*



2

311.1

*



3

356.0

*



4

279.2

*



5
64-66
201.3

*



6

201.1

*



7
304-305
251.1

**

1H NMR (300 MHz, DMSO-d6):








δ 8.23 (1H, d, J=2.1 Hz), 8.08







(1H, dd, J=8.8, 2.1 Hz), 7.74 (1H,







d, J=8.8 Hz), 7.55 (2H, t, J=2.2







Hz), 6.51 (2H, t, J=2.2 Hz).


8

277.3

**



9
61-63
215.2

*



10
69-71
229.2

*



11
78-80
243.2

*



12
94-97
277.2

**



13
161-164
265.3

*



14
206-207
415.2

*

1H NMR (300 MHz, CDCl3): δ 8.09








(1H, dd, J=8.8, 1.7 Hz), 7.75 (1H, d,







J=1.7 Hz), 7.49 (1H, d, J=8.8 Hz),Hz







7.40 (2H, d, J=8.8 Hz), 7.22 (2H,







d, J=8.8 Hz), 7.17 (2H, d, J=8.8 Hz),







6.93 (2H, d, J=8.8 Hz), 6.15 (1H, s),







3.93 (3H, s), 3.83 (3H, s).


15
225-226
290.3

*



16
175-177
286.3

*



17

248.1

*



18

237.3

*



19

307.4

*



20
159-160
267.2

*



21
125-125
277.3

*



22
146-149
321.1

**



23
234-235
334.2

***



24
123-124
307.1

**



25
291-293
271.2

*

1H NMR (300 MHz, DMSO-d6):








δ 10.81 (1H, s), 8.63 (1H, d,







J=1.7 Hz), 8.10 (1H, dd, J=8.8,







1.7 Hz), 7.76 (1H, d, J=8.8 Hz), 4.35







(2H, q, J=7.0 Hz), 2.22 (3H, s),







1.26 (3H, t, J=7.0 Hz).


26
287-288
335.3

**

1H NMR (300 MHz, CDCl3): δ








10.83 (1H, s), 8.23 (1H, s), 8.00







(1H, d, J=8.8 Hz), 7.92 (2H, d,







J=7.3 Hz), 7.60 (1H, d, J=8.5 Hz),







7.53-7.43 (1H, m), 7.41-7.30 (2H, m),







4.12 (2H, q, J=7.3 Hz), 1.31 (3H, t,







J=7.3 Hz).


27
138-140
236.3

**



28
68-70
272.2

**



29
oil
284.3

*



30
114-116
292.2

*

1H NMR (300 MHz, DMSO-d6):








δ 8.19 (1H, d, J=2.3 Hz), 7.96







(1H, dd, J=8.8, 2.3 Hz), 7.69 (1H,







d, J=8.8 Hz), 7.38 (1H, t, J=7 Hz),







4.32 (2H, q, J=7.0 Hz), 3.81







(3H, s), 3.61 (2H, pentet, J=7 Hz),







1.29 (3H, t, J=6.7 Hz), 1.24 (3H, t,







J=7.0 Hz).


31
189-190
264.1

*

1H NMR (300 MHz, DMSO-d6):








δ 8.11 (1H, d, J=2.0 Hz), 7.94







(1H, dd, J=8.5, 2.0 Hz), 7.65 (1H,







d, J=8.5 Hz), 7.54 (2H, br s), 4.21







(2H, q, J=7.0 Hz), 3.79 (3H, s),







1.20 (3H, t, J=7.0 Hz).


32
oil
280.2

*



33
113-117
280.2

*



34
137-141
232.1

*



35
318-319
 411.0,

*

1H NMR (300 MHz, DMSO-d6):





413.0


δ 11.29 (1H, s), 8.71 (1H, d,




[M-H]


J=2.0 Hz), 8.13 (1H, dd, J=8.8,







2.0 Hz), 7.98 (2H, d, J=8.5 Hz),







7.84 (2H, d, J=8.5 Hz), 7.83 (1H, d,







J=8.8 Hz), 4.41 (2H, q, J=7.0 Hz),







1.28 (3H, t, J=7.0 Hz).


36
273-274
380.1

*

1H NMR (300 MHz, DMSO-d6):








δ 11.58 (1H, s), 8.87 (1H, s),







8.72 (1H, s), 8.53 (1H, d, J=7.0 Hz),







8.47 (1H, d, J=7.0 Hz), 8.14







(1H, d, J=8.5 Hz), 7.91 (1H, t,







J=8.2 Hz), 7.86 (1H, d, J=8.8 Hz),







4.45 (2H, q, J=7.0 Hz), 1.30 (3H, t,







J=7.0 Hz).


37
56-60
246.2

*



38
96-100
310.1

*



39
94-98
310.1

*



40
176-180
269.1

*



41
155-175
267.1

*



42
138-143
296.1

*



43

229.2

*



44

293.2

*



45
130-131
215.5

*



46

376.2

*



47
140-145
376.2

*



48
138-142
296.3

**



49
89-90
376.3

*



50
91-92
259.3

*



51
95-96
229.4





52

243.2

*



53
145-147
363.1

**



54
99-101
272.3

*



55
183-185
292.2

*



56
223-224
301.2

**



57
130-133
243.2





58
76-79
243.2

*



59
81-85
242.2

*



60
133-136
271.2

*



61
153-158
270.2

*



62

262.0

*



63
188-192
333.4

*



64
108-113
301.3

*



65
130-132
331.3

**



66
119-122
331.3

*



67
132-136
319.3

**



68
140-147
315.3

**



69
163-166
316.3

**



70
142-141
321.2

*



71
199.4
348.2

*



72
144-149
271.3

*



73
oil
259.2

*



74
179-182
325.3

*



75
118-123
271.3

*



76
118-120
293.3

*



77
117-118
307.3

*



78
110-114
287.2

*



79
257-260
332.4

*



80
292-294
356.5

**



81
209-211
360.5
***
***



82
223-228
372.5
***
**



83
221-223
384.4
***
***



84
232-237
396.4
***
***



85
163-165
398.3
***
***



86
158-160
410.3

***



87
187-189
396.3
***
***



88
209-213
398.4
***
**



89
148-155
308.3

**



90
80-95
364.4

*



91
160-161
301.2





92
155-156
317.2

*



93
172.4-172.6
305.3

*



94
262-265
314.4

**



95
248-251
344.4

**



96
243-250
329.4

**



97
164-167
350.4

*



98
180-185
363.2

*



99
123.4-123.8
307.0

*



100
128-129
277.2

*



101
204-209
426.6

**



102
136.7-136.9
267.2

*



103
90-93
263.2

*



104
190-194
406.4

**



105
204-206
442.4

*



106
230-243
494.4

*



107
157-158
327.1

*



108
94-96
249.2

*





251.2





109
54-56
263.2

*





265.2





110
128-130
349.3

**



111
208.5
374.3

*

1H NMR (CDCl3, 400 MHz),








δ = 7.75-7.72 (m, 2H), 7.13-7.10







(m, 2 H), 6.97-6.88 (m, 3H), 6.11







(s, 2H)


112
173.1-173.5
277.3

**

1H NMR (CDCl3, 400 MHz), δ = 7.67








(t, J=8.4 Hz, 2 H), 7.24 (d, J=14.8 Hz,







1H), 7.16 (d, J=8.0 Hz, 1 H), 6.96-6.90







(m, 3 H), 6.10 (s, 2 H), 2.46 (s, 3 H)


113
193-194
374.3

*



114
207-298
390.3

*



115
175-177
177.1

*



116
116-118
349.4

*



117
120-123
249.3

*



118
62-65
205.2

**



119
126-128
283.2

**



120
69-71
205.5

*



121
167-169
364.5

**



122
163-169
426.5

*



123
113-117
239.4

*



124
212.2-212.3
291.3

*
CDCl3, 400 MHz, δ = 7.61 (dd, J=8.4 Hz







and 4.4 Hz, 1 H), 7.04-6.96 (m, 2H),







6.77 (m, 3H), 6.12 (s, 2H), 2.42 (s, 3H)


125
151.2-151.7
291.3

**
CDCl3, 400 MHz, δ = 7.57 (d, J=8.0 Hz,







1H), 7.09 (d, J=8.0 Hz, 1H), 6.97 (d,







J=28.0 Hz, 1 H), 6.86 (s, 1 H), 6.79 (dd,







J=11.2 Hz and 2.0 Hz, 2H), 6.12 (s, 2H),







2.42 (s, 3H), 2.40 (s, 3H)


126
146-148
251.3

*



127
173-175
327.3

*



128
218-220
334.5

*



129
188-190
370.4

**



130
227-233
394.5

***



131
199-204
408.5

**



132
209-212
422.5

**



133
144-146
351.5

**



134
155-157
335.4

**



135
168-170
369.5

**



136
159-161
381.4

**



137
129-132
345.5

**



138
235-239
358.5
***
**



139
191-195
388.5
***
**



140
83-96
253.3

*



141
145-146
253.4

*



142
103-105
237.4

*



143
168-171
384.5

*



144
97-100
315.4

**



145
110-113
357.5

**



146
169-172
335.3

**



147
183-186
344.4

**



148
155-157
335.4

**



149
134-136
345.4

**



150
141-143
319.4

**



151
146-148
319.4

**



152
207-211
386.5

*



153
225-228
398.5
*
**



154

293.1

**
DMSO, 400 MHz, δ = 8.13 (d, J=6.4 Hz,







1H), 8.01 (s, 2H), 7.84 (s, 1H), 7.54







(dd, J=12.4 Hz and 3.6 Hz, 1H), 7.15 (d,







J=8.8 Hz, 2H), 3.83 (s, 3H)


155



**
CDCl3, 300 MHz, δ = 9.31 (s, 1H), 8.32







(s, 1H), 8.19-8.17 (m, 1H), 7.75 (s,







1H), 2.70 (s, 3H)


156
205-209
421.5

**



157
149-153
422.5

**



158
153-156
317.5

**



159
147-150
361.3

**



160
117-119
315.7

**



161
174-176
340.3

**



162
185-190
413.5

*



163
235-239
410.5

**



164
273-280
358.5

**



165
211-225
373.5

*



166
236-240
385.5

***



167
196-200
464.5

**



168
199-204
394.5

*



169
147-150
316.5

**



170
148-151
307.4

**



171
134-137
307.4

**



172
221-223
317.4

**



173
150-153
316.5

**



174
139-142
302.4

**



175
132-135
359.5

**



176
162-164
343.5

**



177
125-130
331.4

**



178
119-123
369.4

*



179
79-80
239.4

**



180
170-171
384.5
**
**



181
177-178
398.5

**



182
148-154
408.5

***



183
276-284
344.5

**



184
197-200
337.4

**



185
157-160
355.4

**



186
166-169
317.4

**



187
187-191
321.4

**



188
209-212
287.4

**



189
252-253
356.4

*



190
234-236
370.4

**



191
208-210
370.4
**
*



192
205-207
384.5

**



193
228-232
378.5

**





[M-H]

**



194
278-284
370.4

**



195
271-275
398.4

**



196
187-189
354.3

**



197
147-149
373.6

*



198
163-165
395.7

*



199
156-158
369.2
***
*



200
187-189
354.4

**



201
147-150
381.2

**



202
137-139
369.4

**



203

257.9

**
DMSO, 300 MHz, δ = 10.34 (s, 1H),







7.42 (d, J=8.7 Hz, 1H), 7.14 (d,







J=2.1 Hz, 1H), 6.87 (dd, J=8.7 Hz and







2.1 Hz, 1H), 4.08 (q, J=7.2 Hz, 2H), 3.80







3.80 (s, 3H), 2.12 (s, 3H), 1.21







(t, J=7.2 Hz,3H)


204

320.1

***
DMSO, 300 MHz, δ = 10.81 (s, 1H),







8.02 (t, J=7.2 Hz, 2H), 7.67-7.47 (m,







4H), 7.21 (d, J=2.4 Hz, 1H), 6.90 (dd,







J=8.4 Hz and 2.1 Hz, 1H), 4.12 (q,







J=6.9 Hz, 2H), 3.83 (s, 3H), 1.24(t,







J=27.5 Hz, 3H)


205
181-184
358.4

**



206
187-191.
372.4

**



207
179-183.
386.4

**



208
192-194.
394.4

**



209
180-183.
408.4

**



210
213-216
422.4

**



211
186-191.
384.4

**



212
180-183
400.4

**



213
165-168
398.4

**



214
254.8-255.1
338.1

***

1H NMR (DMSO, 300 MHz), δ = 7.88








(d, J=7.5 Hz, 1H), 7.82 (d, J=9.9 Hz,







1H), 7.64 (q, J=8.1 Hz, 1H), 7.21 (d,







J=2.4 Hz, 1H), 7.56-7.48 (m, 2H), 7.21







(d, J=2.1 Hz, 1H), 6.91 (dd, J=8.7 Hz







and 2.1 Hz, 1H), 4.12 (q, J=7.2 Hz, 2H),







4.13 (s, 3H), 1.23 (t, J=6.9 Hz, 3H)


215
245-246
345.1

**

1H NMR (DMSO, 300 MHz), δ = 11.08








(s, 1H), 8.16 (d, J=8.4 Hz, 2H), 8.07 (d,







J=7.8 Hz, 2H), 7.49 (d, J=8.4 Hz, 1H),







7.21 (d, J=2.1 Hz, 1H), 6.91 (dd,







J=8.4 Hz and 1.8 Hz, 1H), 4.08 (q,







J=2.1 Hz, 2H), 3.80 (s, 3H), 1.25 (t,







J=7.2 Hz, 3H)


216
261
283.9

**

1H NMR (DMSO, 300 MHz), δ = 10.58








(br, 1H), 7.42 (d, J=8.7 Hz, 1H), 7.15







(d, J=2.1 Hz, 1H), 6.86 (dd, J=8.7 Hz







and 2.4 Hz, 1H), 4.08 (q, J=7.2 Hz, 2H),







3.80 (s, 3H), 2.04 (br, 1H), 1.21 (t,







J=6.9 Hz, 3H), 0.89-0.84 (m, 5H)


217
272-273
315.9

***

1H NMR (DMSO, 300 MHz), δ=9.94








(s, 1H), 7.43 (d, J=8.7 Hz, 1H), 7.15







(d, J=2.1 Hz, 1H), 6.87 (dd, J=8.7 Hz







and 2.4 Hz, 1H), 4.12 (q, J=7.2 Hz,







2H), 3.91 (d, J=6.6 Hz, 2H), 3.80







(s, 3H), 1.89-1.92 (m, 1H), 1.21 (t,







J=7.2 Hz, 3H), 0.89 (d, J=6.6 Hz, 6H)


218
174-177
344.3

**



219
145-149
388.4

**



220
219-223
387.7

**



221
195-200
421.3

**



222
216-219
385.4

**



223
210-216
316.4

**



224
245-249
358.3

**



225
231-236
372.3

**



226
294-295
375.9

*

1H NMR (CDCl3, 300 MHz) δ (ppm),








3.72 (s, 3H), 3.80 (s, 3H), 6.75-6.91







(m, 2H), 7.42-7.56 (m, 2H), 8.04 (d,







1H), 8.23 (d, 1H), 8.67 (d, 1H).


227
198-203
392.3

**



228
195-198
388.3

*



229
142-147
321.3

*



230
218-220
323.4

*



231
201-203
289.3

*



232
190-193
307.3

*



233
232-234
329.4

**



234
221-223
288.3

*



235
218-222
316.4

**



236
145-147
287.3

*



237
146-148
303.3

*



238
189-191
273.3

*



239
212-218
292.3

*



240
164-167
335.3

*



241
184-188
344.3

*



242
242-248
334.3

*



243
194-198
370.3

***



244
187-190
360.4

.



245
192-195
390.4

*



246
160-164
374.4

*



247
212-217
293.3

*



248
117-121
351.4

**



249
132-136
337.4

**



250
160-162
323.3

*



251
186-187
398.4
***
**



252
176-177
432.4

**



253
121-125
399.2

**



254
153-154
307.21

*



255
149-151
435.5

**



256
230-232
331.3

*



257
 174-176.
330.3

*



258
146-148
330.3

*



259
foam
358.3

*



260
109-111
359.3

*



261
138-143
406.4

**



262
117-121
365.4

**



263
121-127
419.4

**



264
glass
406.5

**



265
204-206
355.3

*



266
96-99
362.4

**



267
106-112
336.4





268
137-143
414.4

**



269
153-158
428.4

**



270
175-177
404.4

*



271
158-160
418.4

**



272
173-176
396.4

**



273
207-209
404.4

*



274
166-172
378.4

*



275
201-206
384.4

*



276
224-228
426.4

*



277
186-191
370.4

*



278
234-240
356.4

*



279
197-202
407.5

*



280
89-95
364.4

*



281
132-134
283.3

*



282
135-136
317.3

*



283
215-218
353.4

*



284
112-114
267.3

*



285
185-190
420.5

*



286
191-192
333.3

**

1H NMR (300 MHz, CDCl3): δ








7.92-7.88 (2H, m), 7.67 (1H, s),







7.66 (1H, d, J=8.8 Hz), 7.46-7.42







(2H, m), 6.98 (1H, dd, J=8.8, 2.3 Hz),







6.88 (1H, d, J=2.3 Hz), 4.35 (2H,







q, J=7.3 Hz), 3.92 (3H, s), 1.46







(3H, t, J=7.3 Hz).


287
175-177
384.4

*



288
194-199
293.3

*



289
173-175
432.2

*



290

433.1

*

1H NMR (CDCl3, 300MHz), δ7.97-7.92








(m, 2H), 7.70-7.62 (m, 3H), 7.10 (br, 1H),







6.98 (dd, J=8.7 Hz and 2.1 Hz, 1H),







6.88 (d, J=2.1Hz, 1H), 4.16 (q, J=7.5 Hz,







2H), 3.91 (s, 3H), 3.77 (t, J=4.2 Hz, 4H),







3.62 (q, J=5.4Hz, 2H), 2.68 (t, J=5.7 Hz,







2H), 2.59 (br, 4H), 1.36 (t, J=7.2 Hz, 3H)


291

319.9

*

1H NMR (CD3OD, 300MHz), δ 8.07








(d, J=7.2 Hz, 2H), 7.79-7.68 (m, 2H),







7.54 (d, J=8.7 Hz, 1H), 7.12 (d,







J=2.1 Hz, 1H), 6.97 (dd, J=8.7 Hz and







2.1 Hz, 1H), 4.24 (q, J=7.5 Hz, 2H), 3.90







(s, 3H), 1.26 (t, J=7.2 Hz, 3H)


292
176-177
348.0

*

1H NMR (CDCl3, 300 MHz), δ 7.93-








7.88 (m, 2H), 7.69-7.60 (m, 3H), 6.98







(dd, J=8.7 Hz and 2.1 Hz, 1H), 6.88 (d,







J=2.1 Hz, 1H), 6.17 (br, 1H), 4.15 (q,







J=7.2 Hz, 2H), 3.91 (s, 3H), 3.53 (m,







2H), 1.35 (t, J=7.2 Hz, 3H), 1.28 (t,







J=7.2 Hz, 3H)


293

412.5

*



294

364.4

***



295

609.4

**



296

392.4

**



297

378.4

**



298

394.4

**



299

376.4

**



300

392.4

**



301

412.4

**



302

382.4

**



303

374.2

*



304

426.4

**



305

442.4

*



306

446.4

**



307

430.4

**



308

424.3

*



309

490.3

**



310

378.4

**



311

392.1

*



312

378.1

*



313

394.1

*



314

376.1

*



315

412.0

*



316

474.1

*



317

382.1

*



318

446.1

*



319

430.1

*



320

426.1

*



321

490.9

*



322
223-230
308.4

*



323
102.9-103.4
447.2

*

1H NMR (CD3OD, 300 MHz),








δ8.02-8.00 (m, 2H), 7.78-7.68 (m, 2H),







7.55 (d, J=8.4 Hz, 1H), 7.12 (s, 1H),







6.97 (dd, J=6.6 Hz and 1.5Hz, 1H), 4.24







(q, J=7.5Hz, 2H), 3.90 (s, 3H), 3.68







(t, J=4.2Hz, 4H), 3.52 (q, J=5.4Hz, 2H),







2.50 (t, J=6.9Hz, 6H), 1.86 (br, 2H),







1.28 (t, J=7.2Hz, 3H)


324
165.3-165.7
368.1

*
(DMSO, 300 MHz), δ 10.56 (s, 1H),







7.85 (s, 1H), 7.73 (d, J=7.8 Hz, 1H),







7.54 (q, J=7.8 Hz, 2H), 7.31 (d,







J=7.8 Hz, 1H), 7.23 (s, 1H), 6.92 (d,







J=8.7 Hz, 1H), 4.27 (s, 2H), 4.18 ( q,







J=7.5 Hz, 2H) , 3.83 (s, 3H) 1.20 (t,







J=7.2 Hz, 3H)


325

462.1

*
(DMSO, 300 MHz), δ 10.30 (s, 1H),







8.02 (s, 1H), 7.91 (d, J=7.5 Hz, 1H),







7.58-7.54 (m, 4H), 7.51-7.49 (m, 2H),







7.04 (d, J=8.4 Hz, IH), 6.92 (dd,







J=8.7 Hz and 1.8 Hz, 1H), 6.11 (s, 2H),







4.22 (q, J=7.5 Hz, 2H), 3.83 (s, 3H),







1.21 (t, J=6.9 Hz, 3H)


326
137-138
396.1

*



327
154-155
386.1

*
(DMSO, 300 MHz), δ 10.45 (s, 1H),







8.02 (s, 1H), 7.95 (d, J=8.1 Hz, 2H),







7.55 (q, J=7.8 Hz, 2H), 7.33 (q,







J=3.6 Hz, 2H), 7.25 (s, 1H), 6.95 (dd,







J=3.6 and J=2.4 Hz, 1H), 6.70 (q,







J=1.8 Hz, 1H), 4.22 ( q, J=7.5 Hz, 2H) ,







3.84 (s, 3H), 1.22 (t, J=6.9 Hz, 3H)


328
174-175
401.1

*
(CDCl3, 300 MHz), δ 8.68 (s, 1H), 8.00







(s, 1H), 7.65 (m, 2H), 7.55 (t, J=8.4 Hz,







1H), 7.40 (d, J=7.2 Hz, 1H), 6.95 (dd,







J=7.2 and 1.8 Hz, 1H), 6.90 (d, J=2.1,







1H), 6.54 (s, 1H), 4.20 ( q, J=7.2 Hz,







2H) , 3.92 (s, 3H), 2.53 (s, 3H), 1.39







(t, J=7.2Hz, 3H)


329
175-176
334.0

*
(CDCl3, 300 MHz), δ 7.79 (s, 1H), 7.64







(d, J=8.4 Hz, 1H), 7.58 (d, J=8.7 Hz,







1H), 7.49 (t, J=7.8 Hz, 1H), 7.38 (s,







1H), 7.30 (d, J=4.5 Hz, 1H), 6.99 (d,







J=2.1 Hz, 1H), 6.92 (d, J=2.1 Hz, 1H)







4.19 (q, J=6.9 Hz, 2H), 3.91 (s, 3H),







2.22 (s, 3H), 1.39 (t, J=7.2 Hz, 3H)


330
237-238
438.1

*
(DMSO, 300 MHz), δ 10.90 (s, 1H),







8.71 (s, 1H), 8.18 (d, J=9.9 Hz, IH),







8.06 (s, 1H), 7.97 (t, J=9.3 Hz, 2H),







7.63 (t, J=8.1 Hz, 1H), 7.53 (d, J=8.7,







1H), 7.39 (d, J=8.4 Hz, 1H), 7.27 (d,







J=1.8 Hz, 1H), 6.94 (dd, J=8.7 Hz and







2.1 Hz, 1H), 4.23 ( q, J=6.6 Hz, 2H) ,







3.84 (s, 3H), 1.23 (t, J=7.2 Hz, 3H)


331
160-165.
370.5

**



332
239-145
334.5

*



333
237-240
384.5

**



334
242-246
348.5

**



335
87-88
297.5

**



336
129-132
384.5

*



337
135-137
283.4

*



338
125-135
367.5

*





[M-H]





339
215-219
362.5

**



340
243-248
334.2

*



341
227-230
348.3

**



342
258-261
337.3

*

1H NMR (300 MHz, CDCl3): δ 8.70








(1H, s), 7.79 (1H, d, J=1.8 Hz),







7.73 (1H, dd, J=8.5, 1.8 Hz), 7.07 (1H,







d, J=8.5 Hz), 6.95 (2H, d, J=8.8 Hz),







6.65 (2H, d, J=8.8 Hz), 3.97 (2H, q,







J=7.0 Hz), 3.54 (3H, s), 1.12 (3H, t,







J=7.0 Hz).


343
131-133
373.5

*



344
177-178
356.5

**



345
191-192
370.5

**



346
178-180
384.3

***



347
146-148
357.3

*



348
126-128
267.2

*



349

392.3

***



350

374.3

**



351

390.3

**



352

388.3

**



353

350.2

***



354

388.3

**



355

384.2

*



356

404.3

*





[M-H]





357

392.3

*



358

374.3

*



359

390.3

*



360

388.3

*



361

404.3

*



362

609.5

**





[M-H]





363
201-207
394.2

**





[M-H]





364
183-188
398.2

**



365
oil
408.2

***



366
223-225
420.2

***



367
225-227
434.2

**



368
168-170
434.2

**



369
174-177
470.2

**



370
159-164
432.2

*



371
168-170
340.2

*

1H NMR (300 MHz, DMSO-








d6): δ 10.24 (1H, s), 7.75-7.60 (1H,







m), 7.60 (2H, d, J=8.8 Hz), 7.45-







7.25 (3H, m), 7.41 (2H, d, J=8.8







Hz), 4.21 (2H, q, J=7.0 Hz), 3.13







(3H, s), 1.22 (3H, t, J=7.0 Hz).


372
211-212
334.3

**

1H NMR (300 MHz, DMSO-d6):








δ 9.98 (1H, s), 7.73-7.62 (2H, m),







7.69 (2H, d, J = 8.4 Hz), 7.54 (2H,







d, J=8.4 Hz), 7.39-7.24 (2H, m),







4.26-4.12 (4H, m), 1.26 (3H, t,







J=7.0 Hz), 1.20 (3H, t, J=7.3 Hz).


373
222-228
354.2

*





[M-H]





374
180-186
396.2

*





[M-H]





375
161-166
425.6

*





[M-H]





376

278.4

*



377
153-156
403.1

*

1H NMR (DMSO, 300 MHz),








δ7.66-7.51 (m, 5H), 7.25 (d, J=2.1 Hz,







1H), 6.91 (dd, J=8.7 Hz and 2.4 Hz,







1H), 4.20 (q, J=7.5 Hz, 2H), 3.83







(s, 3H), 3.65-3.55 (m, 2H), 2.73-2.68







(m, 4H), 1.71-1.54 (m, 4H), 1.18







(t, J=7.2 Hz, 3H).


378

376.7

*

1H NMR (DMSO, 300 MHz), δ 8.84








(t, J=1.8 Hz, 1H), 8.02 (d, J=8.1 Hz,







2H), 7.67 (d, J=7.8 Hz, 2H), 7.49 (d,







J=8.7 Hz, 1H), 7.38 (s, 1H), 7.22 (s,







1H), 7.02 (s, 1H), 6.89 (d, J=7.2 Hz,







1H), 4.17 (q, J=6.9 Hz, 2H), 3.80-3.77







(m, 5H), 1.12 (t, J=7.2 Hz, 3H).


379
228-232
472.2

**

1H NMR (DMSO, 300 MHz),








δ7.72-7.67 (m, 2H), 7.78-7.68 (m, 2H),







7.55 (d, J=8.4 Hz, 1H), 7.12 (s, 1H),







6.97 (dd, J= 26.6 Hz and 1.5Hz, 1H),







4.20 (q. J=7.5 Hz, 2H), 3.83 (s, 3H),







3.06 (t, J=4.2 Hz, 6H), 2.76-2.69 (m, 6H),







2.50 (s, 6H), 1.96 (br, 2H), 1.18 (t, J=7.2 Hz,







3H).


380



*

1H NMR (DMSO, 300 MHz), δ 10.67








(s, 1H), 8.16 (d, J=2.4 Hz, 1H), 8.11 (d,







J=4.5 Hz, 2H), 7.78 (d, J=8.4 Hz, 2H),







7.73 (d, J=2.4 Hz, 1H), 7.61 (d,







J=9.0 Hz, 1H), 7.54 (d, J=9.0 Hz, 1H),







7.27 (s, 1H), 6.94 (d, J=8.7 Hz, 1H),







4.28-4.26 (m, 2H), 3.84 (s, 3H), 1.17







(t, J=7.2 Hz, 3H).


381
216-220
410.1

*

1H NMR (DMSO, 300 MHz), δ 10.34








(s, 1H), 8.10 (d, J=8.1 Hz, 2H), 7.75 (d,







J=8.1 Hz, 2H), 7.65 (d, J=8.4 Hz, 2H),







7.53 (d, J=8.7 Hz, 1H), 7.26 (s, 1H),







7.14 (d, J=8.1 Hz, 2H), 6.94 (dd,







J=8.7 Hz and 2.4 Hz, 1H), 4.21 (q,







J=6.9 Hz, 2H), 3.83 (s, 3H), 2.25 (s,







3H), 1.17 (t, J=6.9 Hz, 3H).


382
236-238


**

1H NMR (DMSO, 300 MHz), δ 10.55








(s, 1H), 8.10 (d, J=7.8 Hz, 2H),







7.83-7.76 (m, 4H), 7.54 (d, J=8.4 Hz, 1H),







7.41 (d, J=8.1 Hz, 2H), 7.27 (s, 1H),







6.93 (dd, J=9.9 Hz and 1.2 Hz, 1H), 4.22







(q, J=6.9 Hz, 2H), 3.84 (s, 3H), 1.17 (t,







J=7.2 Hz, 3H).


383
161.2-162.2
362.1

*

1H NMR (CDCl3, 300MHz), δ 7.79








(s, 1H), 7.65-7.59 (m, 2H), 7.48 (t,







J=7.8 Hz, 1H), 7.30 (t, J=11.4 Hz, 2H),







6.96 (dd, J=8.7 Hz and 2.1 Hz, 1H), 6.88







(d, J=2.1 Hz, 1H), 4.18 (q, J=7.2 Hz,







2H), 3.91 (s, 3H), 2.57-2.52 (m, 1H),







1.36 (t, J=7.2 Hz, 3H) 1.28-1.23 (m,







6H).


384
199-201
348.0

*

1H NMR (CDCl3, 300 MHz), δ 7.93 (d,








J=8.4 Hz, 2H), 7.67-7.61 (m, 3H), 6.98







(dd, J=8.7 Hz and 2.1 Hz, 1H), 6.88 (d,







J=2.1 Hz, 1H), 6.14 (br, 1H), 4.14 (q,







J=7.5 Hz, 2H), 3.93 (s, 3H), 3.57-3.53







(m, 2H), 1.36-1.26 (m, 6H).


385
213-213.9
362.0

**

1H NMR (DMSO, 300 MHz), δ 8.37








(d, J=6.6 Hz, 1H), 7.98 (d, J=7.8 Hz,







2H), 7.65 (d, J=7.8 Hz, 2H), 7.50 (d,







J=9.0 Hz, 1H), 7.23 (s, 1H), 6.90 (d,







J=7.8 Hz, 1H), 4.19-4.16 (m, 2H), 4.08-







4.06 (m, 1H), 3.80 (s, 3H), 1.14-1.12







(m, 9H).


386
198-199
359.9

*

1H NMR (DMSO, 300 MHz), δ 8.59








(d, J=4.2 Hz, 1H), 7.97 (d, J=8.1 Hz,







2H), 7.67 (d, J=8.1 Hz, 2H), 7.52 (d,







J=8.7 Hz, 1H), 7.25 (d, J=2.1 Hz, 1H),







6.92 (dd, J=8.7 Hz and 2.1 Hz, 1H), 4.19







(q, J=7.2 Hz, 2H), 3.83 (s, 3H), 2.90-







2.80 (m, 1H), 1.16 (t, J=7.2 Hz, 3H),







0.69 (q, J=4.8 Hz, 2H), 0.57 (q,







J=3.3 Hz, 2H).


387
189-189.5
360.1

*

1H NMR (DMSO, 300 MHz), δ 7.81








(d, J=8.4 Hz, 2H), 7.67 (d, J=8.1 Hz,







2H), 7.53 (d, J=8.7 Hz, 1H), 7.25 (d,







J=1.8 Hz, 1H), 6.93 (dd, J=8.7 Hz and







2.1 Hz, 1H), 4.35 (t, J=6.8 Hz, 2H), 4.18







(q, J=7.2 Hz, 2H), 4.06 (t, J=6.8 Hz,







2H), 3.83 (s, 3H), 2.24 (m, 2H), 1.16 (t,







J=7.2 Hz, 3H).


388
160.1-161.8
363.9

*

1H NMR (DMSO, 300 MHz), δ 8.60 (t,








J=2.1 Hz, 1H), 8.02 (d, J=8.4 Hz, 2H),







7.68 (d, J=8.1 Hz, 2H), 7.53 (d,







J=9.0 Hz, 1H), 7.25 (s, 1H), 6.93 (dd,







J=6.9 Hz and 1.8 Hz, 1H), 4.72 (t,







J=5.4 Hz, 1H), 4.19 (q, J=7.2 Hz, 2H),







3.83 (s, 3H), 3.48 (q, J=6.0 Hz, 2H),







3.33 (m, 2H), 1.15 (t, J=7.2 Hz, 3H).


389
130-132
447.1

*

1H NMR (CDCl3, 300 MHz), δ 8.36 (br,








1H), 8.12 (d, J=7.2 Hz, 2H), 7.67-7.59







(m, 3H), 6.97 (dd, J=9.3 Hz and 1.8 Hz,







1H), 6.89 (s, 1H), 4.18 (q, J=7.5 Hz,







2H), 3.98 (br, 2H), 3.84 (s, 3H), 3.67







(q, J=6.9Hz, 2H), 2.87 (br, 4H),







2.17 (s, 1H) 2.14 (br, 1H), 2.09 (s, 1H),







1.40 (t, J=10.5Hz, 3H), 1.31 (t, J=7.5 Hz,







3H).


390
236-236.8
396.1

*

1H NMR (DMSO, 300 MHz), δ 10.42








(s, 1H), 8.12 (d, J=8.4 Hz, 2H),







7.79-7.75 (m, 4H), 7.55 (d, J=8.7 Hz, 1H),







7.35 (t, J=10.6 Hz, 2H), 7.27 (d,







J=2.1 Hz, 1H), 7.09 (t, J=10.6 Hz, 1H),







6.94 (dd, J=8.7Hz and 2.1 Hz, 1H), 4.23







(m, 2H), 3.84 (s, 3H), 1.18 (t, J=6.9 Hz,







3H).


391
240-242


**

1H NMR (DMSO, 300 MHz),δ 10.55








(s, 1H), 8.11 (d, J=8.1 Hz, 2H), 7.82 (m,







4H), 7.55 (d, J=8.7 Hz, 1H), 7.41 (d,







J=9.0 Hz, 2H), 7.27 (d, J=1.8 Hz, 1H),







6.94 (dd, J=8.7 Hz and 2.1 Hz, 1H), 4.23







(q, J=7.1 Hz, 2H), 3.84 (s, 3H), 1.78 (t,







J=7.1 Hz, 3H).


392
243-246
456.1

*

1H NMR (DMSO, 300 MHz), δ 10.27








(s, 1H), 8.11 (d, J=8.4 Hz, 2H), 7.76 (d,







J=8.4 Hz, 2H), 7.54 (d, J=8.7 Hz, 1H),







7.48 (d, J=2.4 Hz, 1H), 7.33 (d,







J=7.2 Hz, 1H), 7.29 (s, 1H), 6.96-6.91







(m, 2H), 4.25 (q, J=8.5 Hz, 2H), 3.84 (s,







3H), 3.73 (s, 3H), 3.72 (s, 3H), 1.18 (t,







J=7.2 Hz, 3H).


393
202-206
296.4

**



394
192-195
372.3

***





[M-H]





395

397.4

**



396

377.2
***
***



397

377.5
***
***



398

391.5

***



399

375.5

***



400

411.3

**



401

363.3
***
***



402

439.5

**



403

453.5

**



404

425.5

*



405

425.5

**



406

503.5

***



407

483.5

**



408
231-235
358.1

**



409
163-164
398.3

**



410
139-141
359.2

**



411
148-149
373.3

**



412
143-144
373.3

*



413
184-185
370.4

***



414
156-157
384.4

**



415
163-165
384.4

***



416
173-175
398.4

***



417
199-201
398.4

***



418
183-184
412.4

***



419
213-215
398.4

**



420
241-243
372.5

**



421
214-216
406.3

*



422
259-261
386.5

**



423
291-294
434.4

*



424
150-153
443.5

*



425
273-275
398.4

**



426
178-180
327.2

**



427
199-202
330.3

**



428
144-146
319.2

**





[M-H]

*



429
200-206
314.3

**



430
180-186
372.4

**





[M-H]

*



431
167-173
296.4

*



432
160-162
318.1

*
(DMSO, 300 MHz), δ 8.64 (t, J=5.4 Hz,







1H), 8.02 (d, J=8.1 Hz, 2H), 7.80-7.65







(m, 4H), 7.40-7.28 (m, 2H), 4.21 (q,







J=6.9 Hz, 2H), 3.29 (q, J=7.2 Hz, 2H),







1.20-1.08 (m, 6H).


433
187-189
329.9

*
(DMSO, 300 MHz), δ 8.61 (d, J=3.6 Hz,







1H), 7.79 (d, J=8.1 Hz, 2H), 7.76-7.64







(m, 4H), 7.40-7.27 (m, 2H), 4.20 (q,







J=7.2 Hz, 2H), 2.88-2.82 (m, 1H), 1.17







(t, J=7.2 Hz, 3H), 0.69 (q, J=7.2 Hz,







2H), 0.56 (q, J=7.8 Hz, 2H).


434
170-175
368.4

*



435
112-117
382.5

*



436
196-202
280.5

*



437
220-227
342.3

*





[M-H]





438
188-194
356.3

*





[M-H]





439
181-186
358.4

**



440
177-181
370.3

**





[M-H]





441

459.5

**



442

397.5

*



443

411.5

*



444

439.5

*



445

453.5

**



446

503.5

*



447

426.5

***



448
301-303
436.5

*

1H NMR (300 MHz, DMSO-d6):








δ 10.65 (1H, s), 7.94 (1H, s),







7.76 (1H, s), 7.48 (1H, d, J=8.5 Hz),







7.45 (2H, d, J=8.5 Hz), 7.33(2H, d,







J=8.5 Hz), 7.21 (1H, d, J=2.0 Hz), 6.90







(1H, dd, J=8.5, 2.0 Hz), (3H, s), 4.13







(2H, q, J=7.0 Hz), 3.82 (3H, s), 3.66







(3H, s), 1.15 (3H, t, J=7.0 Hz).


449
242-243
518.5

*

1H NMR (300 MHz, DMSO-d6):








δ 10.59 (1H, s), 8.49 (1H, d, J=2.3 Hz),







7.81 (1H, dd, J=9.1, 2.3 Hz), 7.52-7.44







(3H, m), 7.29 (2H, d, J=8.5 Hz), 7.21







(1H, d, J=2.3 Hz), 6.90 (2H, d,







J=8.8 Hz), 4.12 (2H, q, J=7.0 Hz),







3.82 (3H, s), 3.63-3.55 (8H, m),







1.12 (3H, t, J=7.0 Hz).


450
217-220
402.5

**



451
179-182
438.4

*

1H NMR (300 MHz, DMSO-d6):








δ 10.87 (1H, s), 7.94 (1H, dt, J=5.0,







1.0 Hz), 7.66 (1H, dt, J=3.5, 1.2 Hz),







7.55-7.48 (3H, m), 7.33 (2H, d,







J=8.0 Hz), 7.22 (1H, d, J=1.8 Hz), 7.14







(1H, ddd, J=5.0, 3.8, 1.0 Hz), 6.90







(1H, dt, J=8.8, 1.0 Hz), 4.13







(2H, q, J=7.3 Hz), 3.83 (3H, s), 1.13







(3H, t, J = 7.3 Hz).


452
299-301
483.4

*

1H NMR (300 MHz, DMSO-d6):








δ 10.62 (1H, s), 9.12 (1H, dd, J=4.1,







1.7 Hz), 8.53-8.45 (2H, m), 8.29







(1H, d, J=8.5 Hz), 7.78-7.68 (2H, m),







7.44 (1H, d, J=8.8 Hz), 7.36 (2H, d,







J=8.8 Hz), 7.27 (2H,d, J=8.8 Hz),







7.16 (1H, d, J=2.0 Hz), 6.87







(1H, dd, J=8.8, 2.0 Hz), 4.04 (2H,







q, J=7.0 Hz), 3.80 (3H, s), 1.06







(3H, t, J=7.0 Hz).


453
198-200
416.5

**



454
180-182
412.4

*

1H NMR (300 MHz, CDCl3): δ7.44-7.11








(4.8H, m), 6.97 (1.2H, d, J=8.4 Hz),







6.78-6.69 (2H, m), 4.00-3.90 (2H, m),







3.713 (1.2H, s), 3.707 (1.8H, s), 2.93







(1H, v br), 2.12 (1.8H, s), 2.09 (1.2H,







s), 1.17 (1.2H, t, J=7.3 Hz), 1.14







(1.8H, t, J=7.3 Hz).


455
221-223
414.4

*



456
157-159
357.5

**



457
156-158
464.5

**



458
272-273
496.5

*

1H NMR (300 MHz, DMSO-d6):








δ 10.10 (1H, s), 8.62 (1H, d, J=8.2 Hz),







8.49-8.45 (2H, m), 8.27 (2H, d,







J=8.8 Hz), 7.90 (2H, d, J=3.8 Hz),







7.72 (2H, d, J=8.8 Hz), 7.70-7.62







(1H, m), 7.55-7.49 (4H, m), 7.26







(1H, d, J = 2.0 Hz), 6.94 (1H, dd,







J=8.8, 2.0 Hz), 4.27 (2H, q, J=7.0 Hz),







3.85 (3H, s), 1.24 (3H, t, J=7.0 Hz).


459
158-163
368.5

**



460
oil
368.3

**





[M-H]





461
200-205
432.3

***



462
135-140
444.2

***





[M-H]





463
178-179
359.5

*



464
164-166
401.5

*



465
99-100
401.5

*



466
164-166
385.5

**



467
169-170
399.5

**



468
201-202
398.5
**
***



469
243-245
384.5

**



470
178-180
426.5

***



471
154-156
412.2

***



472
215-217
410.5

***



473
189-191
424.5

***



474
218-222
293.5

*



475
178-181
293.5

***



476
175-178
340.4

**



477
194-195
418.4

***



478
172-175
469.5

***



479
190-192
467.5

*



480
207-208
485.4

***



481
209-211
482.5

*



482
128-130
471.5

**



483
149-150
292.2

*

1H NMR (300 MHz, CDCl3): δ 7.32-7.25








(2H, m), 7.19 (1H, d, J=2.4 Hz), 6.97







(1H, dd, J=8.9, 2.4 Hz), 6.92-6.80







(3H, m), 4.17 (2H, q, J=7.3 Hz), 3.89







(3H, s), 3.80 (2H, v br), 1.34 (3H, t,







J=7.3 Hz).


484
156-157
334.2

*

1H NMR (300 MHz, CDCl3): δ 8.16








(1H, d, J=8.5 Hz), 8.00 (1H, d,







J=8.7 Hz), 7.68 (1H, d, J=8.8 Hz),







7.60-7.50 (2H, m), 7.25 (1H, obscurred),







7.10 (1H, d, J=8.8 Hz), 5.20 (2H, q,







J=7.3 Hz), 4.02 (3H, s), 1.55 (3H, t,







J=7.3 Hz).


485
128-133
440.2

*



486
116-123
454.2

**



487
175-179
446.2

**



488
158-164
460.2

***



489

418.5

**



490

404.5

**



491

445.2

*



492

463.2

**



493

477.2

***



494

336.2

*



495
126-128
444.3

**



496
236-239


**



497
235-239


*



498
192-194
427.5

**



499
235-250


***



500

468.2

*



501

420.2

*



502

406.2

*



503

406.2

*



504

412.2

*



505
164-166
384.2

*

1H NMR (300 MHz, CDCl3): δ 7.52








(1H, t, J=7.9 Hz), 7.47 (1H, t, J=1.9 Hz),







7.36-7.30 (3H, m), 7.18 (1H, d,







J=2.3 Hz), 7.00 (1H, dd, J=8.9, 2.3







Hz), 6.91 (1H, br s), 4.18 (2H, q,







J=7.3 Hz), 3.90 (3H, s), 3.25 (2H, q,







J=7.3 Hz), 1.43 (3H, t, J=7.3 Hz),







1.38 (3H, t, J=7.3 Hz)


506
156-157
364.2

*

1H NMR (300 MHz, CDCl3): δ 7.67








(1H, s), 7.48-7.44 (2H, m), 7.33







(1H, d, J=9.0 Hz), 7.26-7.23 (1H,







m), 7.19 (1H, d, J=2.3 Hz), 6.98







(1H, dd, J=9.0, 2.3 Hz), 6.79 (1H,







s), 4.24 (2H, q, J=7.3 Hz), 4.19







(2H, q, J=7.3 Hz), 1.36 (3H, t, J=7.3







Hz), 1.32 (3H, t, J=7.3 Hz).


507
152-153
378.2

*

1H NMR (300 MHz, CDCl3): δ 7.66








(1H, s), 7.46 (2H, d, J=5.0 Hz),







7.33 (1H, d, J=9.0 Hz), 7.26-7.21







(1H, m), 7.19 (1H, d, J=2.3 Hz),







6.98 (1H, dd, J=9.0, 2.3 Hz), 6.72







(1H, s), 5.03 (1H, hp, H = 6.1 Hz),







4.19 (2H, q, J=7.3 Hz), 3.89 (3H,







s), 1.36 (3H, t, J=7.3 Hz), 1.30







(6H, d, J=6.1 Hz).


508
glass
378.2

*

1H NMR (300 MHz, CDCl3): δ 7.67








(1H, s), 7.48-7.45 (2H, m), 7.33







(1H, d, J=9.0 Hz), 7.27-7.23 (1H,







m), 7.19 (1H, d, J=2.3 Hz), 6.98







(1H, dd, J=9.0, 2.3 Hz), 6.77 (1H,







s), 4.19 (2H, q, J=7.0 Hz), 4.14







(2H, t, J=6.7 Hz), 3.89 (3H, s),







1.72 (2H, m, J=7.0 Hz), 1.37 (3H,







t, J=7.0 Hz), 0.99 (3H, t, J=6.5







Hz).


509
92-95
412.1

*

1H NMR (300 MHz, CDCl3): δ 7.67








(1H, s), 7.48-7.45 (2H, m), 7.33







(1H, d, J=8.8 Hz), 7.27-7.24 (1H,







m), 7.19 (1H, d, J=2.3 Hz), 6.99







(1H, dd, J=8.8, 2.3 Hz), 6.83 (1H,







s), 4.35 (2H, t, J=6.0 Hz), 4.19







(2H, q, J=7.3 Hz), 3.89 (3H, s),







3.66 (2H, t, J=6.5 Hz), 2.16 (2H, p,







J=6.1 Hz), 1.37 (3H, t, J=7.3 Hz).


510
210-211
368.3

**

1H NMR (300 MHz, 9:1 CDCl3-








DMSO-d6): δ 9.64 (1H, s), 7.28-7.17







(3H, m), 7.14 (1H, d, J=9.0 Hz),







7.04 (1H, d, J=7.3 Hz), 6.91 (1H,







d, J=2.0 Hz), 6.75 (1H, dd, J=9.0,







2.0 Hz), 3.95 (2H, q, J=7.3 Hz),







3.66 (3H, s), 2.81 (3H, s), 1.15 (3H,







t, J=7.3 Hz).


511
146-148
377.3

*

1H NMR (300 MHz, CDCl3): δ








7.34-7.27 (2H, m), 7.19 (1H, d, J=2.3







Hz), 6.97 (1H, dd, J=8.8, 2.3 Hz),







6.88-6.79 (3H, m), 4.17 (2H, q, J=7.0







Hz), 3.89 (3H, s), 3.87 (2H, v br),







1.34 (3H, t, J=7.0 Hz).


512
67-69
407.2

*

1H NMR (300 MHz, CDCl3): δ 10.06








(1H, s), 7.86 (1H, s), 7.56-7.47 (2H,







m), 7.36-7.30 (3H, m), 7.19 (1H, d,







J=2.0 Hz), 7.00-6.96 (1H, m), 4.62







(1H, br), 4.27 (2H, q, J=7.0 Hz),







4.18 (2H, q, J=7.0 Hz), 3.89 (3H,







s), 1.37 (3H, t, J=7.0 Hz), 1.33







(3H, t, J=7.0 Hz).


513
210-211
479.2

*

1H NMR (300 MHz, 9:1 CDCl3-








DMSO-d6): δ 8.44 (2H, d, J=7.0







Hz), 7.61 (1H, s), 7.45 (1H, s), 7.36-







7.13 (3H, m), 7.10 (2H, d, J=8.8







Hz), 6.93 (1H, d, J=7.6 Hz), 6.89







(1H, d, J=7.9 Hz), 6.85 (1H, d, J=2.3







Hz), 6.69 (1H, dd, J=8.8, 2.3 Hz),







3.94 (2H, q, J = 7.0 Hz), 3.60 (3H,







s), 1.08 (3H, t, J=7.0 Hz). 19F NMR







(300 MHz, 9:1 CDCl3-DMSO-







d6): δ −63.02 (3F, s).


514
248-250
268.2

*



515
137-139
381.1

**



516
155-156
395.3

**
. 1H NMR (300 MHz, CDCl3): δ 7.57







(1H, d, J=8.5 Hz), 7.45 (2H, d, J=8.2







Hz), 7.31 (2H, d, J=8.2 Hz), 7.25 (1H, s),







6.95 (1H, dd, J=9.1, 1.7 Hz), 6.67 (1H,







s), 6.54 (1H, t, J=74.8 Hz), 6.48 (1H, s),







4.15 (2H, q), 3.23 (2H, q), 1.45 (3H, t,







J=7.5 Hz), 1.32 (3H, t, J=7.2 Hz).


517
160-161
409.1

*



518
197-199
392.1

**



519
174-175
406.1

***



520
175-177
420.0

***

1H NMR (300 MHz, CDCl3): δ 7.73








(1H, d, J=8.4 Hz), 7.52 (2H, d, J=8.2







Hz), 7.39 (2H, d, J=8.2 Hz), 7.25 (1H,







s), 7.13 (1H, dd, J=7.5, 2.4 Hz), 6.96







(1H, s), 6.56 (1H, t, J=74.1 Hz), 4.16







(2H, q), 3.26 (2H, q), 1.46 (3H, t,







J=7.5 Hz), 1.32 (3H, t, J=6.9 Hz).


521
184-186
[NH-]

***





418.1





522
187-188
[NH-]







432.1

***



523
190-192
[NH-]

***





418.1





524
186-188
434.2

***



525

458.2

**



526

490.3

***



527

493.3

**



528

475.3

**



529

459.2

**



530

461.3

***



531

479.5

**



532

458.2

**



533

490.3

**



534

493.3

***



535

475.3

***



536

459.2

**



537

461.3

***



538

459.2

**



539

477.3

***



540
182-183
385.2

**



541
149-151
399.2

*



542
186-188
413.2

**



543
197-200
324.2

**



544
263-265
392.2
***
***



545
203-205
382.2
**
**



546
208-210
396.2

***



547
180-182
410.2

***



548
171-174
402.1

**



549
150-153
416.1

***



550
183-185
430.0

**



551
187-190
462.3

**



552
185-188
428.1

***



553

412.2

*



554

388.2

*



555

408.2

*



556

440.2

*



557

454.3

**





[M-H]





558
87-90
406.2

**



559
92-94
386.2

*



560
207-210
434.2

*



561
182-185
398.2

*



562
oil
420.1

***



563
210-212
436.1

**



564
210-211
377.1

**

1H NMR (DMSO, 300 MHz), δ 8.61 (t,








J=2.4 Hz, 1H), 8.35 (br, 2H), 8.04 (d,







J=8.4 Hz, 2H), 7.73 (d, J=8.4 Hz, 2H),







7.52 (d, J=8.7 Hz, 2H), 7.26 (d,







J=2.4 Hz, 1H), 6.94 (dd, J=8.7 Hz and







2.1 Hz, 1H), 4.20 (q, J=7.5 Hz, 2H), 3.83







(s, 3H), 3.55 (q, J=6.0 Hz, 2H), 3.09 (br,







2H), 2.60-2.53 (m, 3H), 1.15(t,







J=7.2 Hz, 3H).


565
228-229
304.2

*

1H NMR (DMSO, 300 MHz), δ 10.23








(s, 1H), 7.79 (d, J=8.4 Hz, 2H), 7.69 (d,







J=7.2 Hz, 1H), 7.62 (d, J=7.8 Hz, 1H),







17.54 (d, J=7.8 Hz, 2H), 7.38-7.22 (m,







2H), 4.20 (q, J=7.2 Hz, 2H), 2.07 (s,







3H), 1.19 (t, J=7.2 Hz, 3H).


566

334.1

*

1H NMR (CDCl3, 300 MHz), δ 8.42 (s,








1H), 7.80-7.76 (m, 3H), 7.54 (d,







J=8.4 Hz, 2H), 7.44 (d, J=7.8 Hz, 1H),







7.36-7.26 (m, 2H), 4.20 (q, J=7.2 Hz,







2H), 4.07 (s, 2H), 3.55 (s, 3H), 1.36 (t,







J=7.2 Hz, 3H).


567
238.2-238.6
344.2

**

1H NMR (CDCl3, 300 MHz), δ 7.78-








7.73 (m, 2H), 7.60 (d, J=5.7 Hz, 2H),







7.51 (d, J=5.7 Hz, 1H), 7.38-7.28 (m,







2H), 4.20 (q, J=6.8 Hz, 2H), 3.26 (br,







1H), 2.43 (br, 2H), 2.27 (br, 2H), 2.06-







1.90 (m, 2H), 1.36 (t, J=6.9 Hz, 3H).


568
195-200
320.0

*

1H NMR (CDCl3, 400 MHz), δ 7.99 (d,








J=8.4 Hz, 2H), 7.66 (d, J=7.6 Hz, 2H),







7.35 (d, J=9.2 Hz, 1H), 7.20 (s, 1H),







7.01 (d, J=9.0 Hz, 1H), 6.15 (br, 1H),







5.65 (br, 1H), 4.17 (q, J=6.8 Hz, 2H),







3.90 (s, 3H), 1.36 (t, J=7.6 Hz, 3H).


569

348.1

**

1H NMR (CDCl3, 400 MHz), δ 7.93 (d,








J=8.4 Hz, 2H), 7.62 (d, J=8.0 Hz, 2H),







7.34 (d, J=8.8 Hz, 1H), 7.19 (d,







J=2.0 Hz, 1H), 7.01 (dd, J=8.8 Hz and







J=2.4 Hz, 1H), 6.19 (br, 1H), 4.16 (q,







J=7.2 Hz, 2H), 3.90 (s, 3H), 3.56-3.52







(m, 2H), 1.38-1.23 (m, 6H).


570
220-222
362.1

**

1H NMR (CDCl3, 300 MHz), δ 7.92 (d,








J=8.1 Hz, 2H), 7.62 (d, J=8.1 Hz, 2H),







7.34 (d, J=9.0 Hz, 111), 7.19 (d,







J=2.1 Hz, 1H), 7.00 (dd, J=9.0 Hz and







2.4 Hz, 1H), 6.02 (d, J=7.8 Hz, 1H), 4.35







(m, 1H), 4.16 (q, J=7.2 Hz, 2H), 3.90 (s,







3H), 1.36-1.26 (m, 9H).


571

360.0

**

1H NMR (CDCl3, 400 MHz), δ 7.91 (d,








J=8.0 Hz, 2H), 7.61 (d, J=8.0 Hz, 2H),







7.34 (d, J=8.8 Hz, 1H), 7.19 (d,







J=2.0 Hz, 1H), 7.01 (dd, J=9.2 Hz and







2.4 Hz, 1H), 6.34 (s, 1H), 4.15 (q,







J=7.2 Hz, 2H), 3.90 (s, 3H), 2.97-2.93







(m, 1H), 1.34 (t, J=7.2 Hz, 3H), 0.66







(q, J=7.2 Hz, 2H), 0.07 (t, J=2.1 Hz,







2H).


572
168-171
378.0

**

1H NMR (CDCl3, 300 MHz), δ 7.96 (d,








J=8.4 Hz, 2H), 7.63 (d, J=8.4 Hz, 2H),







7.34 (d, J=8.7 Hz, 1H), 7.19 (d,







J=2.1 Hz, IH), 7.02 (dd, J=8.4 Hz and







2.4 Hz, 1H), 6.60 (br, 1H), 4.16 (q,







J=6.6 Hz, 2H), 3.90 (s, 3H), 3.68 (q,







J=4.5 Hz, 2H), 3.59 (t, J=4.8 Hz, 2H),







3.41 (s, 3H), 1.35 (t, J=6.9 Hz, 3H).


573

417.2

*

1H NMR (CDCl3, 400 MHz), δ 7.59 (s,








4H), 7.34 (d, J=8.8 Hz, 1H), 7.20 (s,







1H), 7.01 (d, J=8.4 Hz, 1H), 4.15 (q,







J=6.4 Hz, 2H), 3.90 (s, 3H), 3.90-3.79







(m, 2H), 3.60 (br, 1H), 3.54 (q,







J=5.6 Hz, 1H), 2.78 (br, 1H), 2.69-2.67







(m, 1H), 2.61 (br, 2H), 2.41 (d,







J=18.4 Hz, 3H), 2.04 (br, 1H), 1.92 (br,







1H), 1.34 (t, J=6.8 Hz, 3H).


574
155-161
427.4

*



575
144-146
345.2

**



576
141-142
425.4

*



577
171-173
345.4

**



578
171-173
317.4

**



579
178-179
368.5

**





[M-H]





580
198-200
370.4

***



581

378.5

**



582

392.5

**



583

410.4

**



584

426.4

*



585

406.5

**



586

440.4

**



587

406.5

**



588

402.5

*



589

422.5

**



590

446.5

**



591

420.5

*



592

416.4

*



593

454.5

*



594

412.4

*



595

420.5

*



596

404.4

**



597

418.5

*



598
169-170
382.5

*

1H NMR (300 MHz, CDCl3): δ 7.64








(1H, d, J=8.8 Hz), 7.53 (1H, t, J=7.9







Hz), 7.46 (1H, t, J=1.9 Hz), 7.34-







7.29 (2H, m), 6.97 (1H, dd, J=8.8,







2.3 Hz), 6.95 (1H, s), 6.86 (1H,







d, J=2.3 Hz), 4.08 (2H, t, J=7.6







Hz), 3.91 (3H, s), 3.13 (3H, s), 1.75







(2H, hx, J=6.6 Hz), 0.80 (3H, t,







J=7.3 Hz).


599
163-164
398.4

*

1H NMR (300 MHz, CDCl3): δ 7.64








(1H, d, J=8.8 Hz), 7.51 (1H, dd,







J=8.5, 7.3 Hz), 7.44 (1H, t, J=1.8







Hz), 7.30 (2H, dd, J=7.9, 2.0 Hz),







6.97 (1H, dd, J=8.8, 2.3 Hz), 6.86







(1H, d, J=2.3 Hz), 6.85 (1H, s),







4.08 (2H, t, J=7.6 Hz), 3.91 (3H,







s), 3.25 (2H, q, J=7.3 Hz), 1.75







(2H, hx, J=7.6 Hz), 1.42 (3H, t,







J=7.5 Hz), 0.80 (3H, t, J=7.4 Hz).


600
144-145
391.5

*

1H NMR (300 MHz, CDCl3): δ 7.60








(1H, d, J=8.8 Hz), 7.57-7.54 (1H,







m), 7.35-7.31 (2H, m), 7.13-7.09







(1H, m), 6.94 (1H, dd, J=8.8, 2.0







Hz), 6.84 (1H, d, J=2.0 Hz), 4.07







(2H, t, J=7.4 Hz), 3.89 (3H, s),







3.21 (2H, t, J=7.0 Hz), 1.68 (2H,







hx, J=7.3 Hz), 1.53 (2H, hx, J=7.3







Hz), 0.91 (3H, t, J=7.3 Hz), 0.73







(3H, t, J=7.4 Hz).


601
195-200
400.4

***





[M-H]





602
179-184
398.4

***





[M-H]





603

422.4

*



604

410.3

*



605
175-180
419.4

*



606
166-170
433.4

***



607
189-194
449.5

***



608
223-228
452.9

**



609
230-234
439.4

**



610
226-231
424.8

**



611
126-128
396.4

**



612
198-200
410.4

**



613
198-200
[NH-]

***





430.4





614
176-177
[NH-]

***





444.4





615
220-225
440.3

*





[M-H]





616
143-149
414.4

***





[M-H]





617
164-167
433.8

***





[M-H]





618
205-211
368.4

***



619
201-206
382.3

**



620
215-223
367.4

**



621
187-188
412.4

**

1H NMR (300 MHz, CDCl3): δ 7.64








(1H, d, J=8.8 Hz), 7.50 (2H, d, J=8.5







Hz), 7.36 (2H, d, J=8.5 Hz), 6.97







(1H, dd, J=8.8, 2.3 Hz), 6.86







(1H, d, J=2.3 Hz), 6.79 (1H, s),







4.05 (2H, t, J=7.6 Hz), 3.91 (3H,







s), 3.23-3.17 (2H, m), 1.93 (2H, hx,







J = 7.3 Hz), 1.73 (2H, hx, J=7.6







Hz), 1.09 (3H, t, J=7.4 Hz), 0.79







(3H, t, J=7.4 Hz).


622
182-183
412.4

**

1H NMR (300 MHz, CDCl3): δ 7.63








(1H, d, J=8.8 Hz), 7.48 (2H, d, J=8.5







Hz), 7.38 (2H, d, J=8.5 Hz), 6.96







(1H, dd, J=8.8, 2.3 Hz), 6.86







(1H, d, J=2.3 Hz), 6.83 (1H, s), 4.05







(2H, t, J=7.6 Hz), 3.90 (3H, s),







3.43 (1H, hp, J=6.6 Hz), 1.73







(2H, hx, J=7.6 Hz), 1.46 (6H, d,







J= 6.6 Hz), 0.79 (3H, t, J = 7.5 Hz).


623
217-218
460.4

**

1H NMR (300 MHz, CDCl3): δ 7.65








(1H, d, J=8.8 Hz), 7.47 (2H, d, J=8.5







Hz), 7.40-7.28 (5H, m), 7.25 (2H,







d, J=8.5 Hz), 6.97 (1H, dd, J=8.8,







2.3 Hz), 6.86 (1H, d, J=2.3 Hz),







6.55 (1H, s), 4.45 (2H, s), 4.05







(2H, t, J=7.6 Hz), 3.91 (3H, s),







1.74 (2H, hx, J=7.3 Hz), 0.80 (3H,







t, J = 7.4 Hz).


624
187-188
378.5

**

1H NMR (300 MHz, CDCl3): δ 7.63








(1H, d, J=8.5 Hz), 7.57 (2H, d, J=8.8







Hz), 7.46 (2H, d, J=8.8 Hz), 6.95







(1H, dd, J=8.8, 2.3 Hz), 6.85 (1H, d,







J=2.3 Hz), 6.79 (1H, s), 4.26 (2H,







q, J=7.0 Hz), 4.05 (2H, t, J=7.5 Hz),







3.90 (3H, s), 1.72 (2H, hx, J=7.6 Hz),







1.34 (3H, t, J=7.0 Hz), 0.78







(3H, t, J=7.4 Hz).


625
152-153
392.4

***

1H NMR (300 MHz, CDCl3): δ 7.63








(1H, d, J=8.5 Hz), 7.57 (2H, d, J=8.5







Hz), 7.46 (2H, d, J=8.5 Hz), 6.95 (1H,







dd, J=8.5, 2.3 Hz), 6.85 (1H, d,







J=2.3 Hz), 6.77 (1H, s), 4.17 (2H, t,







J=6.7 Hz), 4.05 (2H, t, J=7.4 Hz),







3.90 (3H, s), 1.73 (2H, hx, J=7.3 Hz),







1.00 (3H, t, J=7.4 Hz), 0.78







(3H, t, J=7.4 Hz).


626
193-194
377.5

**

1H NMR (300 MHz, CDCl3): δ 7.61








(1H, d, J=8.5 Hz), 7.52 (2H, d, J=8.8







Hz), 7.41 (2H, d, J=8.8 Hz), 7.30







(1H, br), 6.96 (1H, dd, J=8.8, 2.0







Hz), 6.86 (1H, d, J=2.0 Hz), 4.05







(2H, t, J=7.6 Hz), 3.90 (3H, s),







3.32 (2H, q, J=7.3 Hz), 1.71







(2H, hx, J=7.3 Hz), 1.19 (3H, t, J=7.3







Hz), 0.77 (3H, t, J=7.3 Hz).


627
188-189
391.5

***

1H NMR (300 MHz, CDCl3): δ 7.61








(1H, d, J=8.5 Hz), 7.52 (2H, d, J=8.8







Hz), 7.41 (2H, d, J=8.8 Hz), 7.34







(1H, br), 6.96 (1H, dd, J=8.5, 2.0







Hz), 6.86 (1H, d, J=2.0 Hz), 4.05







(2H, t, J=7.4 Hz), 3.90 (3H, s),







3.24 (2H, t, J=7.0 Hz), 1.72 (2H, hx,







J=7.6 Hz), 1.57 (2H, hx, J=7.0 Hz),







0.95 (3H, t, J=7.3 Hz), 0.77 (3H, t,







J=7.3 Hz).


628
221-226
381.4

**



629
204-210
436.3

*



630
205-210
416.3

**



631
177-182
428.5

**





[M-H]





632
176-178
366.4

**





[M-H]





633
159-161
380.5

**





[M-H]





634
163-165
396.3

**



635
200-201
392.5

***





[M-H]





636
97-89
428.4

***





[M-H]





637

398.4

***



638

390.5

*



639
159-160
412.5

*

1H NMR (300 MHz, CDCl3): δ 7.63








(1H, d, J=8.8 Hz), 7.50 (1H, t, J=7.9







Hz), 7.44 (1H, t, J=1.8 Hz), 7.33-7.28







(2H, m), 7.18 (1H, s), 6.96 (1H, dd,







J=8.8, 2.3 Hz), 6.86 (1H, d,







J=2.3 Hz), 4.08 (2H, t, J=7.6 Hz),







3.90 (3H, s), 3.22-3.16 (2H, m),







1.90 (2H, hx, J=7.8 Hz), 1.74 (2H, hx,







J=7.6 Hz), 1.05 (3H, t, J=7.4 Hz),







0.79 (3H, t, J=7.5 Hz).


640
197-198
396.5

**

1H NMR (300 MHz, CDCl3): δ 7.63








(1H, d, J=8.8 Hz), 7.58 (2H, d,







J=8.8 Hz), 7.48 (2H, d, J=8.8 Hz),







6.95 (1H, dd, J=8.8, 2.0 Hz), 6.92







(1H, s), 6.85 (1H, d, J=2.0 Hz),







4.62 (2H, dt, J=74.3, 4.2 Hz), 4.50







(2H, dt, J=55.8, 4.2 Hz), 4.05 (2H,







t, J=7.6 Hz), 3.90 (3H, s), 1.72







(2H, hx, J = 7.6 Hz), 0.78 (3H, t,







J=7.4 Hz). 19F NMR (300 MHz,







CDCl3): δ 5.11 (1F, m).


641
177-178
392.5

**

1H NMR (300 MHz, CDCl3): δ 7.63








(1H, d, J=8.8 Hz), 7.57 (2H, d,







J=8.5 Hz), 7.46 (2H, d, J=8.5 Hz),







6.95 (1H, dd, J=8.5, 2.0 Hz), 6.85







(1H, d, J=2.0 Hz), 6.72 (1H, s),







5.05 (1H, hp, J=7.1 Hz), 4.05 (2H,







t, J = 7.6 Hz), 3.90 (3H, s), 1.72







(2H, hx, J=7.6 Hz), 1.33 (6H, d,







J= 7.1 Hz), 0.78 (3H, t, J=7.4 Hz).


642
152-153
426.5



1H NMR (300 MHz, CDCl3): δ








7.66-7.57 (3H, m), 7.53 (2H, d, J=8.8







Hz), 6.97 (1H, dd, J=8.8, 2.0 Hz),







6.86 (1H, d, J = 2.0 Hz), 4.07 (2H, t,







J=7.4 Hz), 3.91 (3H, s), 3.46 (3H,s),







3.40 (1H, hp, J = 7.0 Hz), 1.74







(2H, hx, J = 7.3 Hz), 1.40 (6H, d,







J=7.0 Hz), 0.79 (3H, t, J =7.4 Hz).


643
150-151
412.4

**

1H NMR (300 MHz, CDCl3): δ 7.64








(1H, d, J=8.8 Hz), 7.58 (2H, d, J=9.0







Hz), 7.53 (2H, d, J = 9.0 Hz), 6.97







(1H, dd, J=8.8, 2.0 Hz), 6.86 (1H,







d, J=2.0 Hz), 4.07 (2H, t, J=7.6







Hz), 3.91 (3H, s), 3.44 (3H, s),







3.12 (2H, q, J=7.6 Hz), 1.75 (2H,







hx, J=7.6 Hz), 1.41 (3H, t, J=7.3







Hz), 0.79 (3H, t, J=7.5 Hz).


644

348.2

*
(DMSO, 300 MHz), δ 10.12 (s, 1H),







7.77 (d, J=8.4 Hz, 2H), 7.58 (d,







J=8.7 Hz, 1H), 7.49 (d, J=8.7 Hz, 2H),







7.03 (s, IH), 6.92 (d, J=8.4 Hz, 1H),







4.13 (q, J=6.9 Hz, 2H), 3.77 (s, 3H),







2.31 (q, J=7.5 Hz, 2H), 1.16 (t,







J=8.7 Hz, 3H), 1.06 (t, J=7.5 Hz, 3H).


645
242-244
410.2

*
(DMSO, 300 MHz), δ 10.33 (s, 1H),







7.99 (d, J=8.4 Hz, 2H), 7.72 (d,







J=7.8 Hz, 1H), 7.64-7.56 (m, 4H), 7.51







(d, J=1.8 Hz, 1H), 7.36-7.07 (m, 2H),







7.06 (d, J=8.1 Hz, IH), 6.12 (s, 2H),







4.22 (q, J=7.2 Hz, 2H), 1.21 (t,







J=7.2 Hz, 3H).


646

356.2

**
(DMSO, 300 MHz), δ10.47 (s, 1H),







8.01-7.96 (m, 3H), 7.72 (d, J=8.4 Hz,







1H), 7.65-7.59 (m, 3H), 7.38-7.29 (m,







3H), 6.71 (dd, J=3.9 Hz and J=1.8 Hz,







1H), 4.22 (q, J=6.9 Hz, 2H), 1.21 (t,







J=7.2 Hz, 3H).


647

430.1

*
(DMSO, 300 MHz), δ 10.56 (s, 1H),







8.12 (d, J=8.4 Hz, 2H), 7.84-7.76 (m,







4H), 7.67 (d, J=9.0 Hz, IH), 7.41 (d,







J=9.0 Hz, 2H), 7.11 (d, J=8.7 Hz, 1H),







6.98 (dd, J=8.7 Hz and 2.1 Hz, 1H),







4.22 (q, J=7.2 Hz, 2H), 3.82 (s, 3H),







1.18 (t, J=7.2 Hz, 3H).


648

464.0

**
(DMSO, 300 MHz), δ 10.69 (s, 1H),







8.18-8.09 (m, 3H), 7.82-7.72 (m, 3H),







7.70-7.56 (m, 2H), 7.11 (d, J=2.1 Hz,







1H), 6.96 (dd, J=8.1 Hz and 2.4 Hz, IH),







4.22 (q, J=7.2 Hz, 2H), 3.82 (s, 3H),







1.19 (t, J=7.2 Hz, 3H).


649
210-212
410.5

**

1H NMR (300 MHz, DMSO-








d6): δ 7.60 (2H, d, J=8.8 Hz), 7.50







(1H, d, J=8.8 Hz), 7.36 (2H, d, J=8.8







Hz), 7.25 (1H, d, J=2.1 Hz),







6.91 (1H, dd, J= 8.8, 2.1 Hz), 4.15







(2H, t, J=7.3 Hz), 3.833 (2H, t, J=6.4







Hz), 3.831 (3H, s), 3.57 (2H, t, J=7.3







Hz), 2.45-2.40 (2H, m), 1.54







(2H, hx, J = 7.3 Hz), 0.62 (3H, t,







J=7.3 Hz).


650
165-166
384.5

**

1H NMR (300 MHz, CDCl3): δ 7.64








(1H, d, J=8.8 Hz), 7.54 (4H, s),







6.96 (1H, dd, J=8.8, 2.0 Hz), 6.87







(1H, d, J=2.0 Hz), 4.84 (1H, br t,







J=6.1 Hz), 4.41 (2H, d, J=6.1 Hz),







4.13 (2H, q, J=7.3 Hz), 3.90 (3H,







s), 2.97 (3H, s), 1.33 (3H, t, J=7.3







Hz).


651
146-147
384.5

**

1H NMR (300 MHz, CDCl3): δ 7.63








(1H, d, J=8.8 Hz), 7.56-7.45 (4H,







m), 6.96 (1H, dd, J=8.8, 2.0 Hz),







6.87 (1H, d, J=2.0 Hz), 4.89 (1H,







br t, J=5.9 Hz), 4.42 (2H, d, J=5.9







Hz), 4.13 (2H, t, J=7.0 Hz), 3.91







(3H, s), 2.94 (3H, s), 1.34 (3H, t,







J=7.0 Hz).


652
191-194
418.5

***



653
foam
469.5

*



654
197-201
485.5

*



655
184-187
424.2

*



656
159-161
346.2

*



657
196-198
388.2

*



658
203-205
400.5

*



659
175-177
458.4

**



660
215-217
394.5

**



661
156-158
458.5

***



662

398.4

***



663

308.3

*



664

424.4

*



665

444.5

*



666
207-209
424.4

***



667
242-244
424.4

***



668
171-174
483.0

**



669
213-215
461.5

***

1H NMR (300 MHz, 9:1 CDCl3-








DMSO-d6): δ 9.73 (1H, s), 8.76 (1H,







s), 8.54 (1H, d, J=5.2 Hz), 8.11







(1H, d, J=7.9 Hz), 7.58 (1H, dd, J=7.9,







5.2 Hz), 7.45 (1H, d, J=9.3 Hz),







7.28 (4H, s), 6.88-6.82 (2H, m),







5.15 (2H, s), 3.97 (2H, q, J=7.2 Hz),







2.99 (2H, q, J=7.3 Hz), 1.21







(3H, t, J=7.3 Hz), 1.15 (3H, t,







J=7.2 Hz).


670
231-233
473.4

***

1H NMR (300 MHz, 9:1 CDCl3-








DMSO-d6): δ 9.59 (1H, s), 8.87 (1H,







s), 8.60 (1H, d, J=5.2 Hz), 8.35







(1H, d, J=7.6 Hz), 7.80 (1H, dd, J=7.6,







5.2 Hz), 7.45 (1H, d, J=8.5 Hz),







7.32-7.23 (4H, m), 6.88 (1H, d,







J=2.0 Hz), 6.83 (1H, dd, J=8.5,







2.0 Hz), 5.21 (2H, s), 3.97 (2H, q,







J=7.3 Hz), 2.44-2.35 (1H, m), 1.14







(3H, t, J=7.3 Hz), 1.07-0.99 (2H,







m), 0.84-0.78 (2H, m).


671
221-222
461.4

***

1H NMR (300 MHz, 9:1 CDCl3-








DMSO-d6): δ 9.72 (1H, s), 8.56 (1H,







d, J=5.0 Hz), 8.13 (1H, br t, J=7







Hz), 7.87 (1H, d, J=7.6 Hz), 7.53-







7.50 (1H, m), 7.41 (1H, d, J=8.5







Hz), 7.25 (4H, s), 7.01 (1H, d,







J=2.0 Hz), 6.86 (1H, dd, J=8.5,







2.0 Hz), 5.50 (2H, s), 3.98 (2H, q,







J=7.3 Hz), 2.97 (2H, q, J=7.3 Hz),







1.19 (3H, t, J=7.3 Hz), 1.11 (3H, t,







J=7.3 Hz).


672
165-166
415.5

***

1H NMR (300 MHz, CDCl3): δ 7.65








(1H, d, J=8.8 Hz), 7.55 (4H, s),







6.97 (1H, dd, J=8.8, 2.3 Hz), 6.88







(1H, d, J=2.3 Hz), 4.41 (2H, s),







4.14 (2H, q, J=7.3 Hz), 3.91 (3H,







s), 2.91 (3H, s), 2.86 (3H, s), 1.35







(3H, t, J=7.3 Hz).


673
131-132
398.4

*

1H NMR (300 MHz, CDCl3): δ 7.65








(1H, d, J=8.8 Hz), 7.57-7.48 (4H,







m), 6.97 (1H, dd, J=8.8, 2.0 Hz),







6.88 (1H, d, J=2.0 Hz), 4.42 (2H,







s), 4.15 (2H, q, J=7.3 Hz), 3.91







(3H, s), 2.89 (3H, s), 2.87 (3H, s),







1.37 (3H, t, J=7.3 Hz).


674
159-160
414.4

*

1H NMR (300 MHz, 9:1 CDCl3-








DMSO-d6): δ 9.67 (1H, s), 7.36 (1H,







d, J=8.8 Hz), 7.31 (2H, d, J=8.8







Hz), 7.23 (2H, d, J=8.8 Hz), 6.76







(1H, d, J =2.0 Hz), 6.72 (1H, dd,







J=8.8, 2.0 Hz), 4.05 (2H, t, J=5.6







Hz), 3.68 (3H, s), 3.44 (2H, t, J=5.6







Hz), 3.00 (3H, s), 2.95 (2H, q, J=7.3







Hz), 1.17 (3H, t, J=7.3 Hz).


675

460.4

**

1H NMR (300 MHz, 9:1 CDCl3-








DMSO-d6): δ 9.83 (1H, s), 7.61-7.27







(5H, m), 6.82-6.78 (2H, m), 4.10







(2H, t, J=5.6 Hz), 3.74 (3H, s),







3.56-3.47 (4H, m), 3.16 (2H, t, J=7.3







Hz), 3.06 (3H, s), 2.21-2.01 (2H, m).


676
174-175
394.4

**

1H NMR (300 MHz, 9:1 CDCl3-








DMSO-d6): 5 8.77 (1H, s), 7.51 (2H,







d, J=8.5 Hz), 7.41 (1H, d, J=8.5







Hz), 7.31 (2H, d, J=8.5 Hz), 6.80







(1H, d, J=2.0 Hz), 6.77 (1H, dd,







J=8.5, 2.0 Hz), 4.12-4.04 (4H, m),







3.72 (3H, s), 3.46 (2H, t, J=5.6







Hz), 3.04 (3H, s), 1.18 (3H, t,







J=7.2 Hz).


677
133-134
408.4

**

1H NMR (300 MHz, CDCl3): δ 7.64-








7.51 (5H, m), 6.98-6.94 (2H, m),







6.78 (1H, s), 4.25 (2H, t, J=5.7







Hz), 4.17 (2H, t, J=6.7 Hz), 3.90







(3H, s), 3.63 (2H, t, J=5.7 Hz),







3.22 (3H, s), 1.73 (2H, hx, J=7.3







Hz), 1.00 (3H, t, J=7.4 Hz).


678
184-188
318.3

*



679
212-220
389.5

**



680
163-168
403.3

***



681
192-197
403.3

**



682
194-195
460.4

**



683
157-159
494.2

**



684
175-176
412.5

*



685
140-141
448.4

**



686
173-174
424.5

**



687
124-125
420.4

***



688
178-179
410.4

**



689
204-205
409.5

**



690
192-193
423.9

**



691
203-205
383.8
**




692
175-176
397.8

**



693
163-164
411.8

***



694
135-136
383.8

**



695
159-160
397.8

**



696
194-196
397.8

**



697
foam
467.0

***



698
foam
499.5

**



699
255-258
427.8

***



701
188-194
395.8

**



702
147-150
409.8

**



703
160-165
424.5

**



704
169-170
433.4

*



705
199-201
397.4

*



706
151-153
396.4

***



707
159-161
412.5

**



708
175-177
426.5

**



709
166-168
424.4

***



710
oil
458.5

*



711
oil
424.5

*



712
259-261
316.5

*



713
199-202
474.5

**



714
52-53
281.4

**



715
174-175
424.1

***



716
204-205
422.4

**

1H NMR (CDCl3, 300 MHz) δ (ppm)








0.04-0.12 (m, 2H), 0.41-0.50 (m, 2H),







0.98-1.13 (m, 1H), 2.54-2.67 (m, 2H),







3.45 (t, 2H), 3.87 (t, 3H), 3.91 (s, 2H),







3.99 (d, 2H), 6.93-6.99 (m, 2H), 7.36-







7.43 (m, 2H), 7.49-7.56 (m, 2H), 7.61-







7.66 (m, 1H).


717
205-207
424.5

**



718
195-196
450.2

**



719
192-193
410.2

*



720
172-178
410.5

**



721
158-160
404.6

**



722
160-162
432.6

**



723
175-180
418.6

**



724
168-170
416.4

**



725
215-216
397.5

**



726
221-222
411.4
**




727
217-218
487.5

**



728
197-199
479.5

***



729
213-217
493.6

*



730
210-214
495.2

***



731
173-174
388.5

***



732
219-221
378.2

**



733
146-148
354.5

*



734
167-169
412.5

**



735
123-125
426.5

***



736
125-126
[NH-]
***






438.5





737
153-155
[NH-]

**





438.5





738
149-151
[NH-]

***





442.4





739
oil
456.5

*



740
203-205
424.9

**



741
194-196
438.9

***



742
171-173
[NH-]

**





451.5





743
129-129
465.5

***



744
92-93
412.5

***



745
oil
426.5

***



746
oil
440.5

*



747
136-137
410.5

***



748
186-188
391.6

***



749
176-178
405.6

***



750
173-174
419.6

***



751
159-162
405.6

**



752
198-202
396.5

**



753
157-161
409.9

***



754
146-150
424.5

**



755

357.2

***

1H NMR (DMSO, 300 MHz), δ11.02 (s,








1H), 8.83 (d, J=1.8 Hz, 1H), 8.01 (d,







J=8.4 Hz, 2H), 7.71 (d, J=7.2 Hz, 1H),







7.66-7.63 (m, 3H), 7.39-7.26 (m, 3H),







4.22 (q, J=7.5 Hz, 2H), 1.20 (t,







J=6.9 Hz, 3H).


756

388.0

***

1H NMR (CDC13, 300 MHz), δ 8.04








(s, 1H), 7.83 (d, J=8.4 Hz, 2H), 7.77 (d,







J=7.5 Hz, 1H), 7.60 (d, J=8.7 Hz, 2H),







7.46 (d, J=7.2 Hz, 1H), 7.42-7.31 (m,







2H), 4.23 (t, J=7.8 Hz, 2H), 3.01 (s,







3H), 1.38 (t, J=7.2, 3H).


757
218-225
358.2

***

1H NMR (CDC13, 300 MHz), δ7.79-








7.76 (m, 3H), 7.58 (d, J=8.4 Hz, 2H),







7.46 (d, J=7.5 Hz, 2H), 7.39-7.32 (m,







2H), 4.23 (q, J=7.5 Hz, 2H), 2.71 (s,







3H), 2.54 (s, 3H), 1.38 (t, J=7.2 Hz,







3 H).


758
149-153
320.2

***



759

376.2

***

1H NMR (DMSO, 300 MHz) δ 10.14 (s,








1H), 7.79 (d, J=8.4 Hz, 2H), 7.58-7.45







(m, 3H), 7.22 (d, J=1.5 Hz, 1H), 6.88







(dd, J=8.7 Hz and 1.8 Hz, 1H), 4.75-4.67







(m, 1H), 4.15 (q, J=6.9 Hz, 2H), 2.34







(q, J=7.6 Hz, 2H), 1.27 (d, J=6.0 Hz,







6H), 1.16-1.07 (m, 6H).


760

402.2

***
(DMSO, 300 MHz) δ9.99 (s, 1H), 7.80







(d, J=8.7 Hz, 2H), 7.51-7.45 (m, 3H),







7.22 (s, 1H), 6.87 (dd, J=9.0 Hz and







1.8 Hz, 1H), 4.72-4.69 (m, 1H), 4.15 (q,







J=6.6 Hz, 2H), 2.23-1.80 (m, 7H), 1.26







(d, J=6.0 Hz, 6H), 1.13 (t, J=7.2 Hz,







3H).


761
169-173
392.1
***

(DMSO, 300 MHz) δ10.06 (s, 1H), 7.90







(d, J=8.7 Hz, 2H), 7.54-7.46 (m, 3H),







7.22 (d, J=1.5 Hz, 1H), 6.88 (dd,







J=8.4 Hz and 2.1 Hz, 1H), 4.75-4.66 (m,







1H), 4.15 (q, J=7.2 Hz, 2H), 4.03 (s,







2H), 3.37 (s, 3H), 1.26 (d, J=6.0 Hz,







6H), 1.16 (t, J=7.2 Hz, 3H).


762
190-193
452.3

***
(DMSO, 300 MHz) δ10.20 (s, 1H), 7.78







(d, J=8.7 Hz, 2H), 7.52-7.45 (m, 3H),







7.30-7.21 (m, 5H), 7.18-7.14 (m, 1H),







6.88 (dd, J=8.4 Hz and 2.1Hz, 1H),







4.73-4.69 (m, 1H), 4.15 (q, J=6.6 Hz,







2H), 2.91 (t, J-7.5 Hz, 2H), 2.65 (t,







J=7.6 Hz, 2H), 1.27 (d, J=6.0 Hz, 6H),







1.14 (t, J=7.2 Hz, 3H).


763
209.2-209.7
424.3

***
(DMSO, 300 MHz) δ10.54 (s, 1H),







8.93-7.96 (m, 4H), 7.62-7.49 (m, 6H),







7.26 (d, J=1.8 Hz, 1H), 6.91 (dd,







J=8.7 Hz and 2.1 Hz, 1H), 4.76-4.72 (m,







1H), 4.21 (q, J=7.2 Hz, 2H), 1.30 (d,







J=6.0 Hz, 6H), 1.18 (t, J=6.9 Hz, 3H).


764
213.5-231.7
442.2

***
(DMSO, 300 MHz) δ10.52 (s, 1H),







8.06-7.96 (m, 4H), 7.58 (d, J=8.4 Hz,







2H), 7.49 (d, J=8.4 Hz, 1H), 7.37 (t,







J=8.7 Hz, 2H), 7.24 (s, 1H), 6.89 (dd,







J=9.0 Hz and 1.5 Hz, 1H), 4.72-4.66 (m,







1H), 4.18 (q, J=7.2 Hz, 2H), 1.27 (d,







J=6.0 Hz, 6H), 1.16 (t, J=6.9 Hz, 3H).


765
288.5-288.9
438.2

**
(DMSO, 300 MHz) δ10.91 (s, 1H), 8.72







(s, 1H), 8.20 (d, J=9.3 Hz, 1H), 8.03-







7.98 (m, 3H), 7.65-7.62 (d, J=9.0 Hz,







3H), 7.08 (d, J=2.4 Hz, 1H), 6.96 (dd,







J=8.7 Hz and 2.4 Hz, 1H), 4.20 (q,







J=6.8 Hz, 2H), 3.81 (s, 31-1), 1.19 (t,







J=7.2 Hz, 3H).


766
192.8-193.1
440.1

**
(MeOD, 300 MHz) δ 7.98 (d, J=8.7 Hz,







2H), 7.60 (d, J=8.1 Hz, 2H), 7.50 (d,







J=9.3 Hz, 1H), 7.11 (s, 1H), 6.99 (d,







J=6.6 Hz, 1H), 6.60 (s, 1H), 4.24 (q,







J=7.4 Hz, 2H), 3.87 (s, 3H), 2.54 (s,







3H), 1.30 (t, J=6.9 Hz, 3H).


767
204-205
350.9

***



768
170-175
429.4

*



769
118-123
473.5

*



770

361.5

*



771

361.5

***



772
211-212
442.4

***

1H NMR (300 MHz, CDCl3): δ 7.62








(1H, d, J=8.5 Hz), 7.42 (2H, d,







J=8.5 Hz), 7.13 (2H, d, J=8.5 Hz),







6.95 (1H, dd, J=8.5, 1.8 Hz), 6.87







(1H, d, J=1.8 Hz), 5.67 (1H, d,







J=8.8 Hz), 4.28-4.08 (8H, m), 1.47







(3H, t, J=6.9 Hz), 1.37 (6H, td,







J=7.0, 0.6 Hz), 1.34 (3H, t, J=7.3 Hz).


773
191-193
434.9

**

1H NMR (300 MHz, CDCl3): δ 7.60








(1H, d, J=8.8 Hz), 7.52 (2H, d,







J=8.8 Hz), 7.43 (2H, d, J=8.8 Hz),







6.94 (1H, dd, J=8.8, 2.2 Hz), 6.86







(1H, d, J=2.2 Hz), 4.24 (2H, q,







J=7.0 Hz), 4.22-4.06 (6H, m), 1.47







(3H, t, J=7.0 Hz), 1.34-1.25 (6H,







m).


774
205-206
410.9

**

1H NMR (300 MHz, CDCl3): δ 7.63








(1H, d, J=8.8 Hz), 7.53 (2H, d,







J=9.0 Hz), 7.46 (2H, d, J=9.0 Hz),







6.98 (1H, br s), 6.96 (1H, dd,







J=8.8, 2.0 Hz), 6.88 (1H, d, J=2.0







Hz), 4.17-4.09 (4H, m), 3.71-3.61







(4H, m), 1.48 (3H, t, J=7.0 Hz),







1.34 (3H, t, J=7.1 Hz).


775

482.1

**

1H NMR (300 MHz, CDCl3): δ 7.64








(1H, d, J=8.5 Hz), 7.52 (2H, d,







J=9.0 Hz), 7.40 (2H, d, J=9.0 Hz),







6.97 (1H, dd, J=8.5, 2.2 Hz), 6.90







(1H, d, J=2.2 Hz), 4.48 (2H, t,







J=4.7 Hz), 4.26 (2H, t, J=4.7 Hz),







4.05 (2H, t, J=7.6 Hz), 3.87 (2H, t,







J=6.6 Hz), 3.45 (2H, t, J=6.9 Hz),







2.61 (2H, p, J=7.3 Hz), 2.13 (3H,







s), 1.72 (2H, hx, J=7.3 Hz), 0.79







(3H, t, J=7.4 Hz).


776
257-258
389.5

***

1H NMR (300 MHz, DMSO-d6): δ








11.32 (1H, s), 7.82 (2H, d, J=8.8







Hz), 7.61 (2H, d, J=8.8 Hz), 7.50







(1H, d, J=8.8 Hz), 7.24 (1H, d,







J=1.8 Hz), 6.91 (1H, dd, J=8.8, 1.8







Hz), 4.52 (2H, s), 4.18 (2H, q,







J=7.0 Hz), 4.11 (2H, q, J=7.0 Hz),







1.36 (3H, t, J=7.0 Hz), 1.16 (3H, t,







J=7.0 Hz).


777
124-127
511.6

***

1H NMR (300 MHz, CDCl3): δ 7.58








(1H, d, J=8.8 Hz), 7.45 (2H, d,







J=8.8 Hz), 7.39 (2H, d, J=8.8 Hz),







7.33 (1H, br s), 7.31-7.15 (5H, m),







6.94 (1H, dd, J=8.8, 2.2 Hz), 6.86







(1H, d, J=2.2 Hz), 4.84 (1H, t,







J=6.0 Hz), 4.11 (2H, q, J=7.0 Hz),







4.08 (2H, q, J=7.3 Hz), 3.74 (3H,







s), 3.18 (1H, dd, J=14.0, 5.6 Hz),







3.07 (1H, dd, J=14.0, 6.4 Hz), 1.47







(3H, t, J=7.0 Hz), 1.30 (3H, t,







J=7.3 Hz).


778
225-227
375.5

***

1H NMR (300 MHz, CDCl3): δ 7.73








(2H, d, J=8.8 Hz), 7.62 (1H, d,







J=8.8 Hz), 7.52 (2H, d, J=8.8 Hz),







6.95 (1H, dd, J=8.8, 2.0 Hz), 6.88







(1H, d, J=2.0 Hz), 4.13 (2H, q,







J=7.0 Hz), 4.12 (2H, q, J=7.0 Hz),







4.04-3.99 (2H, m), 3.67-3.62 (2H,







m), 1.47 (3H, t, J=7.0 Hz), 1.34







(3H, t, J=7.0 Hz).


779
265-267
389.5

***

1H NMR (300 MHz, DMSO-d6): δ








8.42 (1H, s), 7.72 (2H, d, J=8.8







Hz), 7.61 (2H, d, J=8.8 Hz), 7.53







(1H, d, J=8.5 Hz), 7.26 (1H, d,







J=2.0 Hz), 6.93 (1H, dd, J=8.5, 2.0







Hz), 4.25-4.08 (6H, m), 1.36 (3H, t,







J=7.0 Hz), 1.19 (3H, t, J=7.0 Hz).


780
219-220
463.6

**

1H NMR (300 MHz, CDCl3): δ 7.65








(1H, d, J=8.8 Hz), 7.58 (2H, d,







J=8.8 Hz), 7.48 (2H, d, J=8.8 Hz),







6.97 (1H, dd, J=8.8, 2.0 Hz), 6.88







(1H, d, J=2.0 Hz), 4.18-4.08 (5H,







m), 3.09 (3H, s), 3.00 (3H, s), 2.94







(3H, s), 1.53 (3H, d), 1.47 (3H, t,







J=7.0 Hz), 1.34 (3H, t, J=7.0 Hz).


781
207-208
449.8

***

1H NMR (300 MHz, CDCl3): δ 7.64








(2H, d, J=8.8 Hz), 7.60 (4H, s),







6.97 (1H, d, J=8.8, 2.0 Hz), 6.88







(1H, d, J =2.0 Hz), 4.12 (2H, q,







J=7.0 Hz), 4.11 (2H, q, J=7.0 Hz),







3.36 (3H, s), 3.23 (2H, s), 3.14 (3H,







s), 2.88 (3H, s), 1.48 (3H, t, J=7.0







Hz), 1.33 (3H, t, J=7.0 Hz).


782
188-190
396.5

**



783
201-202
410.5

*



784
245-246
410.5

**



785
151-153
423.5

**

1H NMR (CDC13, 300 MHz) δ (ppm)








1.38 (t, 3H), 3.30-3.37 (m, 4H), 3.51-







3.58 (m, 4H), 4.06-4.18 (q, 2H), 7.45







(d, 1H), 7.54 (s, 1H), 7.66 (d, 1H).


786
202-203
437.5

*



787
261-263
412.2

*



788
112-114
453.9

*



789
154-156
398.2

***



790

398.2

***



791
149-152
396.5

***



792
182-185
350.5

***



793
154-155
364.5

***



794
149-151
378.5

***



795
183-185
378.5

***



796
124-125
392.6

**



797
209-212
277.9

**



798
193.4-193.7
429.2

*
(CD3CN, 400 MHz) δ9.19 (s, 1H), 7.95







(d, J=7.6 Hz, 2H), 7.61 (d, J=8.0 Hz,







2H), 7.56 (d, J=8.8 Hz, 1H), 7.10 (s,







1H), 6.92 (dd, J=8.8 Hz and 0.8 Hz, 1H),







6.56 (s, 1H), 4.74-4.64 (m, 1H), 4.18







(q, J=7.2 Hz, 2H), 2.51 (s, 3H), 1.27 (d,







J=6.0 Hz, 6H), 1.27 (t, J=7.2 Hz, 3H).


799

362.2

***

1H NMR (DMSO, 300 MHz), δ 10.14








(s, 1H), 7.79 (d, J=8.4 Hz, 2H), 7.52-







7.50 (m, 3H), 7.22 (s, 1H), 6.91 (d,







J=8.7 Hz, 1H), 4.17 (q, J=7.5 Hz, 2H),







3.82 (s, 3H), 2.24 (t, J=7.2 Hz, 2H),







1.60 (q, J=7.2 Hz, 2H), 1.23 (t,







J=7.2 Hz, 3H), 0.90 (t, J=7.2 Hz, 3H).


801
199-219
374.2

***

1H NMR (CDCl3, 300 Hz), 7.72 (d,








J=8.4 Hz, 2H), 7.63 (d, J=8.7 Hz, 1H),







7.50 (d, J=8.7 Hz, 2H), 7.22 (s, 1H),







6.91 (dd, J=6.6 Hz and 2.1 Hz, 1H), 6.87







(d, J=2.1 Hz, 1H), 4.14 (q, J=7.2 Hz,







2H), 3.90 (s, 3H), 3.24-3.18 (m, 1H),







2.46-2.25 (m, 4H), 2.06-1.97 (m, 2H),







1.33 (t, J=8.1 Hz, 3H).


802
196-198
410.2

**

1H NMR (DMSO, 300 Hz), δ 10.45 (s,








1 H), 7.80 (d, J=8.4 Hz, 2 H), 7.54-7.48







(m, 3 H), 7.33-7.21 (m, 6H), 6.92 (dd,







J=6.6 Hz and 2.1 Hz, 1 H) 4.17 (q,







J=6.6 Hz, 2H), 3.82 (s, 3 H), 3.67 (s,







2H), 1.15(t, J=7.2 Hz, 3 H).


803
216-217
386.1

*

1H NMR (DMSO, 300 Hz), δ 10.90 (s,








1H), 8.71 (s, 1H), 8.19 (d, J=9.3 Hz,







1H), 8.02-7.98 (m, 3H), 7.63 (d,







J=8.7 Hz, 2H), 7.51 (d, J=8.7 Hz, 1H),







7.24 (d, J=2.1 Hz, 1H), 6.92 (dd,







J=6.6 Hz and 2.1 Hz, 1H), 4.20 (q,







J=6.6 Hz, 2H), 3.83 (s, 3H), 1.16 (t,







J=6.6 Hz, 3H).


804
214-216
401.2

*

1H NMR (DMSO, 300 Hz), δ 10.92 (s,








1H), 8.00 (d, J=8.7 Hz, 2H), 7.59 (d,







J=8.7 Hz, 2H), 7.51 (d, J=8.7 Hz, 1H),







7.23 (d, J=2.1 Hz, 1H), 6.92 (dd,







J=6.6 Hz and 2.1 Hz, IH), 6.69 (s, 1H),







4.19 (q, J=6.6 Hz, 2H), 3.83 (s, 3H),







2.49 (s, 3H), 1.17 (t, J=7.2 Hz, 3H).


805
217-222
452.5

*



806
157-159
422.5

**



807
171-172
396.5

***



808
195-197
410.5

**



809
167-168
410.5

**



810
209-211
422.5

**



811
166-168
354.5

*



812
206-208
460.4

**



813
176-177
458.4

***



814
164-165
[NH-]

*





458.5





815
153-154
473.5

*

1H NMR (300 MHz, CDCl3): δ 7.65








(1H, d, J=8.8 Hz), 7.61 (2H, d,







J=8.8 Hz), 7.56 (2H, d, J=8.8 Hz),







6.98 (1H, dd, J=8.8, 2.3 Hz), 6.89







(1H, d, J=2.3 Hz), 4.24 (2H, t,







J=5.4 Hz), 4.17 (2H, q, J=7.3 Hz),







4.02 (2H, q, J=5.4 Hz), 3.91 (3H,







s), 3.00 (3H, s), 2.01 (3H, s), 1.38







(3H, t, J=7.3 Hz).


816
186-187
445.5

**

1H NMR (300 MHz, CDCl3): δ 7.66








(1H, d, J=8.5 Hz), 7.60 (2H, d,







J=8.8 Hz), 7.55 (2H, d, J=8.8 Hz),







6.98 (1H, dd, J=8.5, 2.0 Hz), 6.89







(1H, d, J=2.0 Hz), 4.16 (2H, q,







J=7.3 Hz), 3.93 (2H, t, J=6.0 Hz),







3.91 (3H, s), 3.53 (2H, t, J=6.0







Hz), 3.38 (3H, s), 3.07 (3H, s), 1.38







(3H, t, J=7.3 Hz).


817
188-190
439.2

*



818
158-159
437.0

***



819

438.9

*



820

408.4

**



821

408.4

*



822
144-145
439.9

**



823
93-94
456.2

*



824
169-170
428.2

*



825
109-110
444.2

*



826
160-161
432.1

**



827
189-191
446.1

***



828
198-200
431.2

**

1H NMR (300 MHz, CDCl3): δ 7.65








(1H, d, J=8.8 Hz), 7.62 (2H, d,







J=8.8 Hz), 7.57 (2H, d, J=8.8 Hz),







6.98 (1H, dd, J=8.8, 2.0 Hz), 6.89







(1H, d, J=2.0 Hz), 4.17 (2H, q,







J=7.2 Hz), 3.93 (2H, t, J=5.3 Hz),







3.91 (3H, s), 3.77 (2H, t, J=5.3







Hz), 3.06 (3H, s), 1.38 (3H, t,







J=7.2 Hz).


829
271-275
422.1

**



830
178-179
420.1

**



831
140-141
371.0

*



832
206-207
406.3

**

1H NMR (CDC13, 300 MHz) δ (ppm)








1.34 (t, 3H), 1.47 (t, 3H), 1.55 (s, 9H),







4.04-4.25 (m, 4H), 6.62 (s, 1H), 6.86-







6.92 (m, 1H), 7.02-7.08 (m, 2H), 7.43-







7.49 (m, 2H), 7.62 (d, 1H), 7.95 (d,







1H).


833
183-185
306.3

**

1H NMR (CDC13, 300 MHz) δ (ppm)








1.21 (t, 3H), 1.37 (t, 3H), 3.84-4.09 (m,







6H), 6.54-6.71 (m, 2H), 6.85-7.01 (m,







2H), 7.23-7.45 (m, 3H).


834
179-180
384.2

**



835
178-179
398.2

**

1H NMR (CDCl3, 300 MHz) δ (ppm),








1.32-1.52 (m, 9H), 3.06-3.17 (q, 2H),







4.07-4.24 (m 4H), 6.55 (s, 1H), 6.99-







7.10 (m, 3H), 7.42-7.50 (m, 2H), 7.52-







7.53 (m, 1H), 7.69 (d, 1H).


836
167-169
412.2

*



837
144-145
410.2

*



838
193-194
378.1

**

1H NMR (CDCl3, 300 MHz) δ (ppm),








1.31-1.39 (m, 6H), 1.48 (t, 3H), 4.07-







4.31 (m, 6H), 6.73 (s, 1H), 6.88-6.95







(m, 1H), 7.01-7.08 (m, 2H), 7.42-7.53







(m, 2H), 7.63 (d, 1H), 7.99 (s, 1H).


839
249-250
377.3

*

1H NMR (CDC13, 300 MHz) δ (ppm),








1.13 (t, 3H), 1.32 (t, 3H), 1.43 (t, 3H),







3.20-3.32 (m, 2H), 4.13-4.26 (m, 4H),







5.77 (t, 1H), 7.06-7.18 (m, 3H), 7.48







(d, 1H), 7.52-7.59 (m, 2H), 8.02 (s,







1H), 8.18 (d, 1H).


840
182-183
442.5

*



841
140-141
429.2

*



842
170-171
427.2

*

1H NMR (CDCl3, 300 MHz) δ (ppm)








1.04-1.13 (m, 2H), 1.20-1.28 (m, 2H),







1.44 (t, 3H), 2.30-2.43 (m, 1H) 3.44-







3.61 (m, 8H), 4.10-4.23 (q, 2H), 7.49







(d, 1H), 7.56 (s, 1H), 7.72 (d, 1H).


843
143-146
463.1

*



844
220-227
378.1

*



845
210-215
435.0

*



846
201-202
391.5

**



847
166-167
405.5

**



848
193-194
453.5

**



849
159-161
364.5

**



850
179-180
446.4

**



851
186-187
410.4

**



852
171-172
426.4

**



853
159-161
346.2

***



854

330.2

**



855

332.1

**



856
202-202
368.1

*



857
192-192
364.2

**



858

362.2

***



859
191-192
374.2

**



860
242-244
334.1

**



861

346.2

**



862

360.1

*



863

362.5

**



864

362.5

**



865
187-192
412.9

*









Example 7
Evaluation of the Activity of Compounds Using an HCV-Poliovirus Chimera

In an HCV-poliovirus (HCV-PV) chimera, the PV 5′ UTR is replaced by the HCV 5′ UTR and partial (the first 123 amino acids) core coding sequences (nucleotides 18 to 710 of HCV 1b) as shown in FIG. 1 (140). As a consequence, the expression of poliovirus proteins is proteins is under regulation of the HCV IRES. Poliovirus is a picornavirus in which protein translation initiation is mediated by an IRES element located in the 5′ UTR. At the 5′ end of the HCV-PV chimeric genome, there is the cloverleaf-like RNA structure of PV, an essential cis-acting replication signal ending with the genome-linked protein VPg. Replication kinetics of the HCV-PV chimera matches that of the parental poliovirus (Mahoney) and can result in cytopathic effects (CPE) in cell culture. Heptazyme, a ribozyme that targets the HCV IRES, was shown to be active against the chimeric virus in cell culture (76, 77).


To evaluate compounds for activity against the chimeric virus, HeLa cells are seeded and incubated at 37° C. under 5% CO2 for 24 hours. The cells are then infected with HCV-PV at a multiplicity of infection (MOI) at 0.1 for 30 min and then treated with compound for 1 day (treatment time will be optimized). The activity of compounds is determined by a change in cytopathic effect, plaque assay, and/or viral RNA production (see e.g., Table 1).


Example 8
Evaluation of the Activity of Compounds Against a Wild-Type Poliovirus (WT-PV) and the Poliovirus IRES Translation Assay (WT-PV Mono Luc)

A DNA construct is prepared, termed pPVIRESmono, in which PV IRES sequences are inserted (nucleotide number 1-742) between a promoter and the firefly luciferase (Fluc) reporter gene. A stably transfected 293 T cell line, is established by transfection with the pPVIRESmono DNA by selecting for resistance to hygromycin. As previously described, cells are treated with compounds for 20 hours, and activity is determined by quantifying the Fluc signal. Additionally, to evaluate compounds activity against wild-type poliovirus, Hela cells are seeded and incubated at 37° C. under 5% CO2 for 24 hours. Cells are then infected with wild-type poliovirus at a MOI at 0.1 for 30 minutes, and then treated with compound for one day. The activity of compounds is determined by changes in cytopathic effect, plaque assay, and RT-PCR using poliovirus IRES primers and probes (see e.g., Table 2).


Furthermore, if compounds are active against the poliovirus and other virus IRESs, then the compounds are useful for treating viral infection by any virus containing an IRES.













TABLE 2





Compound
WT-PV CPE
WT-PV CPE
WT-PV CPE
WTPV mono luc


No.
(100 μM) *
(10 μM)*
(1 μM)*
IC50 (μM)



















4
3
2
1
0.8


5
3
2
1
9


9
3
2
2
>100


10
3
2
2
>100


19
3
2
1
15


24
3
2
2
1.5









Example 9
In Vitro Translation Assay

In vitro translation assays can be used to distinguish between the compounds that act on HCV IRES RNA or cellular translation factors. In exemplary assays, the mRNA that will direct translation is a transcribed runoff product from the T7 RNA polymerase promoter of the pHCVIRESmono plasmid DNA generated with Ambion RNA MegaTranscript kit (Ambion, Inc., Austin, Tex.). In vitro translation is performed using HeLa cell lysates using methods known to one of skill in the art. Preliminary results indicate that one or more of the compounds of the present invention has significantly higher activity against HCV IRES regulated translation after preincubating the compound with the HCV IRES RNA transcripts than after preincubating with HeLa cell lysate for 30 min at 37° C. or without preincubation (data not shown). This suggests that this compound may interact with the HCV IRES RNA in the in vitro translation assay. To demonstrate whether the compounds selectively act on the HCV IRES, pLuc is used together with cellular IRES mRNA transcripts as controls for in vitro translation.


All publications and patent applications cited herein are incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.


Although certain embodiments have been described in detail above, those having ordinary skill in the art will clearly understand that many modifications are possible in the embodiments without departing from the teachings thereof. All such modifications are intended to be encompassed within the claims of the invention.


REFERENCES



  • 1. Ali, N., G. J. Pruijn, D. J. Kenan, J. D. Keene, and A. Siddiqui. 2000. Human La antigen is required for the hepatitis C virus internal ribosome entry site-mediated translation. J Biol Chem 275:27531-27540.

  • 2. Ali, N. and A. Siddiqui. 1995. Interaction of polypyrimidine tract-binding protein with the 5′ noncoding region of the hepatitis C virus RNA genome and its functional requirement in internal initiation of translation. J Virol 69:6367-6375.

  • 3. Ali, N. and A. Siddiqui. 1997. The La antigen binds 5′ noncoding region of the hepatitis C virus RNA in the context of the initiator AUG codon and stimulates internal ribosome entry site-mediated translation. Proc Natl Acad Sci USA 94:2249-2254.

  • 4. Anwar, A. N. Ali, R. Tanveer, and A. Siddiqui. 2000. Demonstration of functional requirement of polypyrimidine tract-binding protein by SELEX RNA during hepatitis C virus internal ribosome entry site-mediated translation initiation. J Biol Chem 275:34231-34235.

  • 5. Beales, L. P., D. J. Rowlands, and A. Holzenburg. 2001. The internal ribosome entry site (IRES) of hepatitis C virus visualized by electron microscopy. RNA 7:661-670.

  • 6. Belsham, G. J. and J. K. Brangwyn. 1990. A region of the 5′ noncoding region of foot-and-mouth disease virus RNA directs efficient internal initiation of protein synthesis within cells: involvement with the role of L protease in translational control. J Virol 64:5389-5395.

  • 7. Belsham, G. J. and R. J. Jackson. 2000. Translation initiation on picornavirus RNA., p. 869-900. Cold Spring Harbor Laboratory Press, New York.

  • 8. Blight, K. J., A. A. Kolykhalov, and C. M. Rice. 2000. Efficient initiation of HCV RNA replication in cell culture. Science 290:1972-1974.

  • 9. Blight, K. J., J. A. McKeating, and C. M. Rice. 2002. Highly permissive cell lines for subgenomic and genomic hepatitis C virus RNA replication. J Virol 76:13001-13014.

  • 10. Borvjagin, G., T. Pestova, and I. Shatsky. 1994. Pyrimidine tract binding protein strongly stimulates in vitro encephalomyocarditis virus RNA translation at the level of the preinitiation complex formation. FEBS Lett 351:291-302.

  • 11. Brown, E. A., H. Zhang, L. H. Ping, and S. M. Lemon. 1992. Secondary structure of the 5′ nontranslated regions of hepatitis C virus and pestivirus genomic RNAs. Nucleic Acids Res 20:5041-5045.

  • 12. Buck C B, Shen X, Egan M A, Pierson T C, Walker C M, and Siliciano R F. 2001. The human immunodeficiency virus type 1 gag gene encodes an internal ribosome entry site. J Virol 75:181-191.

  • 13. Bukh, J., R. H. Purcell, and R. H. Miller. 1992. Sequence analysis of the 5′ noncoding region of hepatitis C virus. Proc Natl Acad Sci USA 89:4942-4946.

  • 14. Bukh, J., R. H. Purcell, and R. H. Miller. 1994. Sequence analysis of the core gene of 14 hepatitis C virus genotypes. Proc Natl Acad Sci USA 91:8239-8243.

  • 15. Buratti, E., S. Tisminetzky, M. Zotti, and F. E. Baralle. 1998. Functional analysis of the interaction between HCV 5′UTR and putative subunits of eukaryotic translation initiation factor eIF3. Nucleic Acids Res 26:3179-3187.

  • 16. Chappell, S. A., J. P. LeQuesne, F. E. Paulin, M. L. deSchoolmeester, M. Stoneley, R. L. Soutar, S. H. Ralston, M. H. Helfrich, and A. E. Willis. 2000. A mutation in the c-myc-IRES leads to enhanced internal ribosome entry in multiple myeloma: a novel mechanism of oncogene de-regulation. Oncogene 19:4437-4440.

  • 17. Chung, R. T., W. He, A. Saquib, A. M. Contreras, R. J. Xavier, A. Chawla, T. C. Wang, and E. V. Schmidt. Hepatitis C virus replication is directly inhibited by IFN-alpha in a full-length binary expression system. 2001. Proc Natl Acad Sci USA 98:9847-9852.

  • 18. Coldwell, M. J., S. A. Mitchell, M. Stoneley, M. MacFarlane, and A. E. Willis. 2000. Initiation of Apaf-1 translation by internal ribosome entry. Oncogene 19:899-905.

  • 19. Creancier, L., D. Morello, P. Mercier, and A. C. Prats. 2000. Fibroblast growth factor 2 internal ribosome entry site (IRES) activity ex vivo and in transgenic mice reveals a stringent tissue-specific regulation. J Cell Biol 150:275-281.

  • 20. Das, S., M. Ott, A. Yamane, A. Venkatesan, S. Gupta, and A. Dasgupta. 1998. Inhibition of internal entry site (IRES)-mediated translation by a small yeast RNA: a novel strategy to block hepatitis C virus protein synthesis. Front Biosci 3:D1241-D 1252.

  • 21. Dever, T. E. 2002. Gene-specific regulation by general translation factors. Cell 108 :545-556.

  • 22. Dumas, E., C. Staedel, M. Colombat, S. Reigadas, S. Chabas, T. Astier-Gin, A. Cahour, S. Litvak, and M. Ventura. 2003. A promoter activity is present in the DNA sequence corresponding to the hepatitis C virus 5′ UTR. Nucleic Acids Res 31:1275-1281.

  • 23. Fukushi, S., K. Katayama, C. Kurihara, N. Ishiyama, F. B. Hoshino, T. Ando, and A. Oya. 1994. Complete 5′ noncoding region is necessary for the efficient internal initiation of hepatitis C virus RNA. Biochem Biophys. Res Commun. 199:425-432.

  • 24. Fukushi, S., C. Kurihara, N. Ishiyama, F. B. Hoshino, A. Oya, and K. Katayama. 1997. The sequence element of the internal ribosome entry site and a 25-kilodalton cellular protein contribute to efficient internal initiation of translation of hepatitis C virus RNA. J Virol 71:1662-1666.

  • 25. Fukushi, S., M. Okada, T. Kageyama, F. B. Hoshino, and K. Katayama. 1999. Specific interaction of a 25-kilodalton cellular protein, a 40S ribosomal subunit protein, with the internal ribosome entry site of hepatitis C virus genome. Virus Genes 19:153-161.

  • 26. Fukushi, S., M. Okada, J. Stahl, T. Kageyama, F. B. Hoshino, and K. Katayama. 2001. Ribosomal protein S5 interacts with the internal ribosomal entry site of hepatitis C virus. J Biol Chem 276:20824-20826.

  • 27. Funkhouser, A. W., D. E. Schultz, S. M. Lemon, R. H. Purcell, and S. U. Emerson. 1999. Hepatitis A virus translation is rate-limiting for virus replication in MRC-5 cells. Virology 254:268-278.

  • 28. Glass, M. J., X. Y. Jia, and D. F. Summers. 1993 Identification of the hepatitis A virus internal ribosome entry site: in vivo and in vitro analysis of bicistronic RNAs containing the HAV 5′ noncoding region. Virology. 193:842-852.

  • 29. Gordon S. C., B. R. Bacon, I. M. Jacobson, M. I. Shiffman, N. H. Afdhal, J. G. McHutchison, T. J. Kwoh, and F. A. Dorr. 2002. A Phase II, 12-week study of ISIS 14803, an antisense inhibitor of HCV for the treatment of chronic hepatitis C. AASLD Abst. 795. Hepatology 36:362A.

  • 30. Gosert, R., K. H. Chang, R. Rijnbrand, M. Yi, D. V. Sangar, and S. M. Lemon. 2000. Transient expression of cellular polypyrimidine-tract binding protein stimulates cap-independent translation directed by both picornaviral and flaviviral internal ribosome entry sites In vivo. Mol Cell Biol 20:1583-1595.

  • 31. Gray, N, and M. Wickens. 1998. Control of translation initiation in animals. Annu Rev Cell Dev Biol 14:399-458.

  • 31a. Griffith, A., and D. M. Coen. 2005. An unusual internal ribosome entry site in the herpes simplex virus thymidine kinase gene. Proc Natl Acad Sci USA. 102:9667-72.

  • 32. Guo, J. T., V. V. Bichko, and C. Seeger. 2001. Effect of alpha interferon on the hepatitis C virus replicon. J Virol 75:8516-8523.

  • 33. Hahm, B., Y. K. Kim, J. H. Kim, T. Y. Kim, and S. K. Jang. 1998. Heterogeneous nuclear ribonucleoprotein L interacts with the 3′ border of the internal ribosomal entry site of hepatitis C virus. J Virol 72:8782-8788.

  • 34. Haller, A. A., S. R. Stewart, and B. L. Semler. 1996. Attenuation stem-loop lesions in the 5′ noncoding region of poliovirus RNA: neuronal cell-specific translation defects. J Virol 70:1467-1474.

  • 35. Hellen, C. U. and T. V. Pestova. 1999. Translation of hepatitis C virus RNA. J Viral Hepat 6:79-87.

  • 36. Hellen, C. U., G. W. Witherell, M. Schmid, S. H. Shin, T. V. Pestova, A. Gil, and E. Wimmer. 1993. A cytoplasmic 57-kDa protein that is required for translation of picornavirus RNA by internal ribosomal entry is identical to the nuclear pyrimidine tract-binding protein. Proc Natl Acad Sci USA 90:4672-7646

  • 37. Hendrix, M., E. S. Priestley, G. F. Joyce, and C. H. Wong. 1997. Direct observation of aminoglycoside-RNA interactions by surface plasmon resonance. Journal of the American Chemical Society 119:3641-8.

  • 38. Holcik, M. and R. G. Korneluk. 2000. Functional characterization of the X-linked inhibitor of apoptosis (XIAP) internal ribosome entry site element: role of La autoantigen in XIAP translation. Mol Cell Biol 20:4648-4657.

  • 39. Holcik, M., C. Lefebvre, C. Yeh, T. Chow, and R. G. Korneluk. 1999. A new internal-ribosome-entry-site motif potentiates XIAP-mediated cytoprotection. Nat Cell Biol 1: 190-192.

  • 40. Honda, M., M. R. Beard, L. H. Ping, and S. M. Lemon. 1999. A phylogenetically conserved stem-loop structure at the 5′ border of the internal ribosome entry site of hepatitis C virus is required for cap-independent viral translation. J Virol 1165-1174.

  • 41. Honda, M., E. A. Brown, and S. M. Lemon. 1996. Stability of a stem-loop involving the initiator AUG controls the efficiency of internal initiation of translation on hepatitis C virus RNA. RNA 2:955-968.

  • 42. Honda, M., L. H. Ping, R. C. Rijnbrand, E. Amphlett, B. Clarke, D. Rowlands, and S. M. Lemon. 1996. Structural requirements for initiation of translation by internal ribosome entry within genome-length hepatitis C virus RNA. Virology 222:31-42.

  • 43. Honda, M., R. Rijnbrand, G. Abell, D. Kim, and S. M. Lemon. 1999. Natural variation in translational activities of the 5′ nontranslated RNAs of hepatitis C virus genotypes 1a and 1b: evidence for a long-range RNA-RNA interaction outside of the internal ribosomal entry site. J Virol 73:4941-4951.

  • 44. Huez, I., S. Bornes, D. Bresson, L. Creancier, and H. Prats. 2001. New vascular endothelial growth factor isoform generated by internal ribosome entry site-driven CUG translation initiation. Mol Endocrinol. 15:2197-2210.

  • 45. Huez, I., L. Creancier, S. Audigier, M. C. Gensac, A. C. Prats, and H. Prats. 1998. Two independent internal ribosome entry sites are involved in translation initiation of vascular endothelial growth factor mRNA. Mol Cell Biol 18:6178-6190

  • 46. Ikeda, M., M. Yi, K. Li, and S. M. Lemon. 2002. Selectable subgenomic and genome-length dicistronic RNAs derived from an infectious molecular clone of the HCV-N strain of hepatitis C virus replicate efficiently in cultured Huh7 cells. J Virol 76:2997-3006.

  • 47. Irvine, J. D., L. Takahashi, K. Lockhart, J. Cheong, J. W. Tolan, H. E. Selick, and J. R. Grove. 1999. MDCK (Madin-Darby canine kidney) cells: A tool for membrane permeability screening. J Pharm Sci 88:28-33.

  • 48. Isoyama, T., N. Kamoshita, K. Yasui, A. Iwai, K. Shiroki, H. Toyoda, A. Yamada, Y. Takasaki, and A. Nomoto. 1999. Lower concentration of La protein required for internal ribosome entry on hepatitis C virus RNA than on poliovirus RNA. J Gen Virol 80 (Pt 9):2319-2327.

  • 49. Ito, T. and M. M. Lai. 1999. An internal polypyrimidine-tract-binding protein-binding site in the hepatitis C virus RNA attenuates translation, which is relieved by the 3′-untranslated sequence. Virology 254:288-296.

  • 50. Jang, S. K., H. G. Krausslich, M. J. Nicklin, G. M. Duke, A. C. Palmenberg, and E. Wimmer. 1988. A segment of the 5′ nontranslated region of encephalomyocarditis virus RNA directs internal entry of ribosomes during in vitro translation. J Virol 62:2636-2643.

  • 51. Jubin, R., N. E. Vantuno, J. S. Kieft, M. G. Murray, J. A. Doudna, J. Y. Lau, and B. M. Baroudy. 2000. Hepatitis C virus internal ribosome entry site (IRES) stem loop IIId contains a phylogenetically conserved GGG triplet essential for translation and IRES folding. J Virol 74:10430-10437.

  • 52. Kalliampakou, K. I., L. Psaridi-Linardaki, and P. Mavromara. 2002. Mutational analysis of the apical region of domain II of the HCV IRES. FEBS Lett 511:79-84.

  • 53. Kaminski, A., S. L. Hunt, J. G. Patton, and R. J. Jackson. 1995. Direct evidence that polypyrimidine tract binding protein (PTB) is essential for internal initiation of translation of encephalomyocarditis virus RNA.RNA 1:924-938

  • 54. Kamoshita, N., K. Tsukiyama-Kohara, M. Kohara, and A. Nomoto. 1997. Genetic analysis of internal ribosomal entry site on hepatitis C virus RNA: implication for involvement of the highly ordered structure and cell type-specific transacting factors. Virology 233:9-18.

  • 55. Kieft, J. S., K. Zhou, R. Jubin, M. G. Murray, J. Y. Lau, and J. A. Doudna. 1999. The hepatitis C virus internal ribosome entry site adopts an ion-dependent tertiary fold. J Mol Biol 292:513-529.

  • 56. Kieft, J. S., K. Zhou, R. Jubin, M. G. Murray, J. Y. Lau, and J. A. Doudna. 2001. Mechanism of ribosome recruitment by hepatitis C IRES RNA. RNA 7:194-206.

  • 57. Klinck, R., E. Westhof, S. Walker, M. Afshar, A. Collier, and F. Aboul-Ela. 2000. A potential RNA drug target in the hepatitis C virus internal ribosomal entry site. RNA 6:1423-1431.

  • 58. Kolupaeva V G, Pestova T V, and Hellen C U T. 2000. An enzymatic foot-printing analysis of the interaction of 40S ribosomal subunits with the internal ribosomal entry site of hepatitis C virus. J Virol 74:6242-6250.

  • 59. Kolupaeva, V. G., C. U. Hellen, and I. N. Shatsky. 1996. Structural analysis of the interaction of the pyrimidine tract-binding protein with the internal ribosomal entry site of encephalomyocarditis virus and foot-and-mouth disease virus RNAs. RNA 2:1199-1212.

  • 60. Kolupaeva, V. G., T. V. Pestova, C. U. Hellen, and I. N. Shatsky. 1998. Translation eukaryotic initiation factor 4G recognizes a specific structural element within the internal ribosome entry site of encephalomyocarditis virus RNA. J Biol Chem 273:18599-18604.

  • 61. Kozak, M. 1999. Initiation of translation in prokaryotes and eukaryotes. Gene 234:187-208.

  • 62. Kruger, M., C. Beger, P. J. Welch, J. R. Barber, M. P. Manns, and F. Wong-Staal. 2001. Involvement of proteasome alpha-subunit PSMA7 in hepatitis C virus internal ribosome entry site-mediated translation. Mol Cell Biol 21: 8357-8364

  • 63. La Monica, N. and V. R. Racaniello. 1989. Differences in replication of attenuated and neurovirulent polioviruses in human neuroblastoma cell line SH-SY5Y. J Virol 63:2357-2360.

  • 64. Le, S. Y., N. Sonenberg, and J. V. Maizel, Jr. 1995. Unusual folding regions and ribosome landing pad within hepatitis C virus and pestivirus RNAs. Gene 154:137-143.

  • 65. Lerat, H., Y. K. Shimizu, and S. M. Lemon. 2000. Cell type-specific enhancement of hepatitis C virus internal ribosome entry site-directed translation due to 5′ nontranslated region substitutions selected during passage of virus in lymphoblastoid cells. J Virol 74:7024-7031.

  • 66. L, K., T. M. Davis, C. Bailly, A. Kumar, D. W. Boykin, and W. D. Wilson. 2001. A heterocyclic inhibitor of the REV-RRE complex binds to RRE as a dimer. Biochemistry 40:1150-8.

  • 67. Lipinski, J. 2000. J. Pharm. Tox. Meth. 44:235-249.

  • 68. Llinas-Brunet M. 2002. NS3 serine protease inhibitors as potential antiviral agents for the treatment of hepatitis C virus infections. The 3rd internatl antiviral & vaccine discovery and development summit. March 13-14. Princeton, N.J.

  • 69. Lohmann, V., F. Korner, A. Dobierzewska, and R. Bartenschlager. 2001. Mutations in hepatitis C virus RNAs conferring cell culture adaptation. J Virol 75:1437-1449.

  • 70. Lohmann, V., F. Korner, J. Koch, U. Herian, L. Theilmann, and R. Bartenschlager. 1999. Replication of subgenomic hepatitis C virus RNAs in a hepatoma cell line. Science 285:110-113.

  • 71. Lopez, d. Q., E. Lafuente, and E. Martinez-Salas. 2001. IRES interaction with translation initiation factors: functional characterization of novel RNA contacts with eIF3, eIF4B, and eIF4GII. RNA 7:1213-1226.

  • 72. Lopez, d. Q. and E. Martinez-Salas. 2000. Interaction of the eIF4G initiation factor with the aphthovirus IRES is essential for internal translation initiation in vivo. RNA 6:1380-1392.

  • 73. Lu, H. H. and E. Wimmer. 1996. Poliovirus chimeras replicating under the translational control of genetic elements of hepatitis C virus reveal unusual properties of the internal ribosomal entry site of hepatitis C virus. Proc Natl Acad Sci USA 93:1412-7.

  • 74. Lukavsky, P. J., G. A. Otto, A. M. Lancaster, P. Sarnow, and J. D. Puglisi. 2000. Structures of two RNA domains essential for hepatitis C virus internal ribosome entry site function. Nat Struct Bio 7:1105-1110.

  • 75. Lyons, A. J., J. R. Lytle, J. Gomez, and H. D. Robertson. Hepatitis C virus internal ribosome entry site RNA contains a tertiary structural element in a functional domain of stem-loop II. Nucleic Acids Res 29:2535-2546.

  • 76. Macejak, D. G., K. L. Jensen, S. F. Jamison, K. Domenico, E. C. Roberts, N. Chaudhary, I. von_Carlowitz, L. Bellon, M. J. Tong, A. Conrad, P. A. Pavco, and L. M. Blatt. 2000. Inhibition of hepatitis C virus (HCV)-RNA-dependent translation and replication of a chimeric HCV poliovirus using synthetic stabilized ribozymes. Hepatology (Baltimore, Md.) 31:769-76.

  • 77. Macejak, D. G., K. L. Jensen, P. A. Pavco, K. M. Phipps, B. A. Heinz, J. M. Colacino, and L. M. Blatt. 2001. Enhanced antiviral effect in cell culture of type 1 interferon and ribozymes targeting HCV RNA. J Viral Hepatitis 8:400-405.

  • 78. Macejak, D. G. and P. Sarnow. 1991. Internal initiation of translation mediated by the 5′ leader of a cellular mRNA. Nature 353:90-94.

  • 79. Major M E, Rehermann B, and Feinstone. 2001. Hepatitis C viruses., p. 2535-2541. In D. Knipe and P. Howley (eds.), Fields Virology. Lippincott Williams and Wilkins, Philadelphia, Pa.

  • 80. Manns M P, McHutchison J G, Gordon S C, Rustgi V K, Shiffman M, Reindollar R, Goodman Z D, Koury K, Ling M, and Albrecht J K. 2003. Peginterferon alfa-2b plus ribavirin compared with interferon alfa-2b plus ribavirin for initial treatment of chronic hepatitis C: a randomised trial. Lancet 358:958-965.

  • 81. Martinez-Salas, E., R. Ramos, E. Lafuente, and d. Q. Lopez. 2001. Functional interactions in internal translation initiation directed by viral and cellular IRES elements. J Gen Virol 82:973-984.

  • 82. Mazur, S., F. A. Tanious, D. Ding, A. Kumar, D. W. Boykin, I. J. Simpson, S. Neidle, and W. D. Wilson. 2000. A thermodynamic and structural analysis of DNA minor-groove complex formation. Journal of Molecular Biology 300:321-37.

  • 83. McHutchison J G and Poynard T. 1999. Combination therapy with interferon plus ribavirin for the initial treatment of chronic hepatitis C. Semin. Liver Dis. 19 Suppl 1:57-65.

  • 84. McHutchison, J. G., T. Poynard, R. Esteban-Mur, G. L. Davis, Z. D. Goodman, J. Harvey, M. H. Ling, J. J. Garaud, J. K. Albrecht, K. Patel, J. L. Dienstag, and T. Morgan. 2002. Hepatic HCV RNA before and after treatment with interferon alone or combined with ribavirin. Hepatology 35:688-693.

  • 85. Meerovitch, K., J. Pelletier, and N. Sonenberg. 1989. A cellular protein that binds to the 5′-noncoding region of poliovirus RNA: implications for internal translation initiation. Genes Dev 3:1026-1034.

  • 86. Meerovitch, K., Y. V. Svitkin, H. S. Lee, F. Lejbkowicz, D. J. Kenan, E. K. Chan, V. I. Agol, J. D. Keene, and N. Sonenberg. 1993. La autoantigen enhances and corrects aberrant translation of poliovirus RNA in reticulocyte lysate. J Virol 67: 3798-3807.

  • 87. Mercer, D. F., D. E. Schiller, J. F. Elliott, D. N. Douglas, C. Hao, A. Rinfret, W. R. Addison, K. P. Fischer, T. A. Churchill, J. R. Lakey, D. L. Tyrrell, and N. M. Kneteman. 2001. Hepatitis C virus replication in mice with chimeric human livers. Nature Medicine 7:927-33.

  • 88. Michel, Y. M., A. M. Borman, S. Paulous, and K. M. Kean. 2001. Eukaryotic initiation factor 4G-poly(A) binding protein interaction is required for poly(A) tail-mediated stimulation of picornavirus internal ribosome entry segment-driven translation but not for X-mediated stimulation of hepatitis C virus translation. Mol Cell Biol 21: 4097-4109.

  • 89. Mitchell, S. A., E. C. Brown, M. J. Coldwell, R. J. Jackson, and A. E. Willis. 2001. Protein factor requirements of the Apaf-1 internal ribosome entry segment: roles of polypyrimidine tract binding protein and upstream of N-ras. Mol Cell Biol 21:3364-3374.

  • 90. Moriguchi, e. al. 1992. Chem Pharm Bull 40:127-130.

  • 91. Nanbru, C., I. Lafon, S. Audigier, M. C. Gensac, S. Vagner, G. Huez, and A. C. Prats. 2003. Alternative translation of the proto-oncogene c-myc by an internal ribosome entry site. J Biol Chem 272:32061-32066.

  • 92. Niepmann, M., A. Petersen, K. Meyer, and E. Beck. 1997. Functional involvement of polypyrimidine tract-binding protein in translation initiation complexes with the internal ribosome entry site of foot-and-mouth disease virus. J Virol 71:8330-8339.

  • 93. Odreman-Macchioli, F., F. E. Baralle, and E. Buratti. 2001. Mutational analysis of the different bulge regions of hepatitis C virus domain II and their influence on internal ribosome entry site translational ability. J Biol Chem 276:41648-41655.

  • 94. Odreman-Macchioli, F. E., S. G. Tisminetzky, M. Zotti, F. E. Baralle, and E. Buratti. 2000. Influence of correct secondary and tertiary RNA folding on the binding of cellular factors to the HCV IRES. Nucleic Acids Res 28:875-885.

  • 95. Ohlmann, T., M. Lopez-Lastra, and J. L. Darlix. 2000. An internal ribosome entry segment promotes translation of the simian immunodeficiency virus genomic RNA. J Biol Chem 275:11899-11906.

  • 96. Pain V M. 1996. Initiation of protein synthesis in eukaryotic cells. Eur J Biochem 236:747-771.

  • 97. Pelletier, J. and N. Sonenberg . 1988. Internal initiation of translation of eukaryotic mRNA directed by a sequence derived from poliovirus RNA. Nature 334:320-325.

  • 98. Pelletier, J. and N. Sonenberg . 1989. Internal binding of eucaryotic ribosomes on poliovirus RNA: translation in HeLa cell extracts. J Virol 63:441-444.

  • 99. Pestova, T. V., S. I. Borukhov, and C. U. Hellen. 1998. Eukaryotic ribosomes require initiation factors 1 and 1A to locate initiation codons. Nature 394:854-859.

  • 100. Pestova, T. V., I. N. Shatsky, S. P. Fletcher, R. J. Jackson, and C. U. Hellen. 1998. A prokaryotic-like mode of cytoplasmic eukaryotic ribosome binding to the initiation codon during internal translation initiation of hepatitis C and classical swine fever virus RNAs. Genes Dev 12: 67-83.

  • 101. Pestova, T. V., I. N. Shatsky, and C. U. Hellen. 1996. Functional dissection of eukaryotic initiation factor 4F: the 4A subunit and the central domain of the 4G subunit are sufficient to mediate internal entry of 43S preinitiation complexes. Mol Cell Biol 16:6870-6878.

  • 102. Peytou, V., R. Condom, N. Patino, R. Guedj, A. M. Aubertin, N. Gelus, C. Bailly, R. Terreux, and D. Cabrol_Bass. 1999. Synthesis and antiviral activity of ethidium-arginine conjugates directed against the TAR RNA of HIV-1. Journal of Medicinal Chemistry 42:4042-53.

  • 103. Pietschmann, T., V. Lohmann, A. Kaul, N. Krieger, G. Rinck, G. Rutter, D. Strand, and R. Bartenschlager. 2002. Persistent and transient replication of full-length hepatitis C virus genomes in cell culture. J Virol 76:4008-4021.

  • 104. Pietschmann, T., V. Lohmann, G. Rutter, K. Kurpanek, and R. Bartenschlager. 2001. Characterization of cell lines carrying self-replicating hepatitis C virus RNAs. J Virol 75:1252-1264.

  • 105. Poole, T. L., C. Wang, R. A. Popp, L. N. Potgieter, A. Siddiqui, and M. S. Collett. 1995. Pestivirus translation initiation occurs by internal ribosome entry. Virology 206:750-754.

  • 106. Pringle, C. 1999. Virus taxonomy—1999. The universal system of virus taxonomy, updated to include the new proposals ratified by the International Committee on Taxonomy of Viruses during 1998. Arch Virol 144:421-429.

  • 107. Psaridi, L., U. Georgopoulou, A. Varaklioti, and P. Mavromara. 1999. Mutational analysis of a conserved tetraloop in the 5′ untranslated region of hepatitis C virus identifies a novel RNA element essential for the internal ribosome entry site function. FEBS Lett 453:49-53.

  • 108. Reynolds, J. E., A. Kaminski, A. R. Carroll, B. E. Clarke, D. J. Rowlands, and R. J. Jackson. 1996. Internal initiation of translation of hepatitis C virus RNA: the ribosome entry site is at the authentic initiation codon. RNA 2:867-878.

  • 109. Reynolds, J. E., A. Kaminski, H. J. Kettinen, K. Grace, B. E. Clarke, A. R. Carroll, D. J. Rowlands, and R. J. Jackson. 1995. Unique features of internal initiation of hepatitis C virus RNA translation. EMBO J. 14: 6010-6020.

  • 110. Rijnbrand R, Bredenbeek P; van der Straaten T, Whetter L, Inchauspe G, Lemon S, and Spaan W. 1995. Almost the entire 5′ non-translated region of hepatitis C virus is required for cap-independent translation. FEBS Lett 365:115-119.

  • 111. Rijnbrand R C and Lemon S M. 2000. Internal ribosome entry site-mediated translation in hepatitis C virus replication. Curr Top. Microbiol Immunol. 242:85-116.

  • 112. Rijnbrand, R., P. J. Bredenbeek, P. C. Haasnoot, J. S. Kieft, W. J. Spaan, and S. M. Lemon. 2001. The influence of downstream protein-coding sequence on internal ribosome entry on hepatitis C virus and other flavivirus RNAs. RNA 7:585-597.

  • 113. Rijnbrand, R. C., T. E. Abbink, P. C. Haasnoot, W. J. Spaan, and P. J. Bredenbeek. 1996. The influence of AUG codons in the hepatitis C virus 5′ nontranslated region on translation and mapping of the translation initiation window. Virology 226 :47-56.

  • 114. Sachs, A. B., P. Sarnow, and M. W. Hentze. 1997. Starting at the beginning, middle, and end: translation initiation in eukaryotes. Cell 89:831-838.

  • 115. Saito I, Miyamura T, Ohbayashi A, Harada H, Katayama T, Kikuchi S, Watanabe Y, Koi S, Onji M, Ohta Y, Choo Q, Houghton M, and Kuo G. 2003. Hepatitis C virus infection is associated with the development of hepatocellular carcinoma. Proc Natl Acad Sci U.S. A 87:6547-6549.

  • 116. Schultz, D. E., M. Honda, L. E. Whetter, K. L. McKnight, and S. M. Lemon. 1996. Mutations within the 5′ nontranslated RNA of cell culture-adapted hepatitis A virus which enhance cap-independent translation in cultured African green monkey kidney cells. J Virol 70:1041-1049.

  • 117. Shimazaki, T., M. Honda, S. Kaneko, and K. Kobayashi. 2002. Inhibition of internal ribosomal entry site-directed translation of HCV by recombinant IFN-alpha correlates with a reduced La protein. Hepatology 35:199-208.

  • 118. Simmonds, P. 2003. Variability of hepatitis C virus. Hepatology 21:570-583.

  • 119. Sinha, R., P. Yang, S. Kodali, Y. Xiong, R. M. Kim, P. R. Griffin, H. R. Onishi, J. Kohler, L. L. Silver, and K. Chapman. 2001. Direct interaction of a vancomycin derivative with bacterial enzymes involved in cell wall biosynthesis. Chem Biol 8:1095-1106.

  • 120. Sizova, D. V., V. G. Kolupaeva, T. V. Pestova, I. N. Shatsky, and C. U. Hellen. 1998. Specific interaction of eukaryotic translation initiation factor 3 with the 5′ nontranslated regions of hepatitis C virus and classical swine fever virus RNAs. J Virol 72:4775-4782.

  • 121. Smith. 1994. Eur J Drug Metab Pharm 3:193-199.

  • 122. Smith, D. B., J. Mellor, L. M. Jarvis, F. Davidson, J. Kolberg, M. Urdea, P. L. Yap, and P. Simmonds. 1995. Variation of the hepatitis C virus 5′ non-coding region: implications for secondary structure, virus detection and typing. The International HCV Collaborative Study Group. J Gen Virol 76 (Pt 7):1749-1761.

  • 123. Sonenberg N, Mathews M B, and Hershey J W B. 2000. Translational control of gene expression. Cold Spring Harbor. Cold Spring Harbor Laboratory Press, New York.

  • 124. Spahn, C. M., J. S. Kieft, R. A. Grassucci, P. A. Penczek, K. Zhou, J. A. Doudna, and J. Frank. 2001. Hepatitis C virus IRES RNA-induced changes in the conformation of the 40s ribosomal subunit. Science 291:1959-1962.

  • 125. Spatzenegger, M. and W. Jaeger. 1995. Clinical importance of hepatic cytochrome P450 in drug metabolism. Drug Metab Rev 27:397-417.

  • 126. Subkhankulova, T., S. A. Mitchell, and A. E. Willis. 2001. Internal ribosome entry segment-mediated initiation of c-Myc protein synthesis following genotoxic stress. Biochem J 359:183-192.

  • 127. Tang, S., A. J. Collier, and R. M. Elliott. 1999. Alterations to both the primary and predicted secondary structure of stem-loop IIIc of the hepatitis C virus 1b 5′ untranslated region (5′UTR) lead to mutants severely defective in translation which cannot be complemented in trans by the wild-type 5′UTR sequence. J Virol 73:2359-2364.

  • 128. Thiel, V. and S. G. Siddell. 1994. Internal ribosome entry in the coding region of murine hepatitis virus mRNA 5. J Gen Virol. 75 (Pt 11):3041-3046.

  • 129. Tsukiyama-Kohara, K., N. Iizuka, M. Kohara, and A. Nomoto. 1992. Internal ribosome entry site within hepatitis C virus RNA. J Virol 66:1476-1483.

  • 130. Vagner, S., M. C. Gensac, A. Maret, F. Bayard, F. Amalric, H. Prats, and A. C. Prats. 1995. Alternative translation of human fibroblast growth factor 2 mRNA occurs by internal entry of ribosomes. Mol Cell Biol 15:35-44.

  • 131. Varaklioti A, Georgopoulou U, Kakkanas A, Psaridi L, Serwe M, Caselmann W H, and Mavromara P. 1998. Mutational analysis of two unstructured domains of the 5, untranslated region of HCV RNA. Biochem Biophys. Res Commun. 253:678-685.

  • 132. Wang, C., S. Y. Le, N. Ali, and A. Siddiqui. 1995. An RNA pseudoknot is an essential structural element of the internal ribosome entry site located within the hepatitis C virus 5′ noncoding region. RNA 1:526-537.

  • 133. Wang, C., P. Sarnow, and A. Siddiqui. 1993. Translation of human hepatitis C virus RNA in cultured cells is mediated by an internal ribosome-binding mechanism. J Virol 67:3338-3344.

  • 134. Wang, C., P. Sarnow, and A. Siddiqui. 1994. A conserved helical element is essential for internal initiation of translation of hepatitis C virus RNA. J Virol 68:7301-7307.

  • 135. Wang, S. M., S. C. Fears, L. Zhang, J. J. Chen, and J. D. Rowley. 2000. Screening poly(dA/dT)-cDNAs for gene identification. Proceedings of the National Academy of Sciences of the United States of America 97:4162-7.

  • 136. Wang, T. H., R. C. Rijnbrand, and S. M. Lemon. 2000. Core protein-coding sequence, but not core protein, modulates the efficiency of cap-independent translation directed by the internal ribosome entry site of hepatitis C virus. J Virol 74:11347-11358.

  • 137. Wimmer, E., C. U. Hellen, and X. Cao. 1993. Genetics of poliovirus. Annu Rev Genet 27:353-436.

  • 138. Wong, J. B., T. Poynard, M. H. Ling, J. K. Albrecht, and S. G. Pauker. 2000. Cost-effectiveness of 24 or 48 weeks of interferon alpha-2b alone or with ribavirin as initial treatment of chronic hepatitis C. International Hepatitis Interventional Therapy Group. Am. J. Gastroenterol. 95:1524-1530.

  • 139. Zhao, W. D. and E. Wimmer. 2001. Genetic analysis of a poliovirus/hepatitis C virus chimera: new structure for domain II of the internal ribosomal entry site of hepatitis C virus. J Virol 75:3719-3730.

  • 140. Zhao, W. D., E. Wimmer, and F. C. Lahser. 1999. Poliovirus/Hepatitis C virus (internal ribosomal entry site-core) chimeric viruses: improved growth properties through modification of a proteolytic cleavage site and requirement for core RNA sequences but not for core-related polypeptides. Journal of Virology 73:1546-54.


Claims
  • 1. A compound of Formula (I):
  • 2. A compound of Formula (I):
  • 3. The compound of claim 1, wherein Z is selected from the group consisting of -cyclopropyl; -cyclobutyl; -cyclopentyl; -cyclohexyl; -cyclopropylmethyl; -cyclobutylmethyl; -cyclopentylmethyl; -hydrogen; —C1 to C6 alkyl optionally substituted with:—C1 to C6 alkoxy, -one or more halogen substituents, or -aryl; —C2 to C6 alkenyl; and -aryl optionally substituted with C1 to C6 alkoxy.
  • 4. The compound of claim 1, wherein R is hydrogen.
  • 5. The compound of claim 1, wherein R1 is selected from the group consisting of -cyclopropyl; -cyclobutyl; -cyclopentyl; -cyclohexyl; -hydrogen; -halogen; -nitro; -5 or 6 membered heterocycle; —C1 to C6 alkoxy optionally substituted with: -aryl; -aryl optionally substituted with C1 to C6 alkoxy.
  • 6. The compound of claim 1, wherein R2 is selected from the group consisting of -nitro; -hydrogen; -halogen; -hydroxy; —C1 to C6 alkyl, optionally substituted with one or more halogen substituents; -cyclopropyl; -cyclobutyl; -cyclopentyl; -cyclohexyl; —C1 to C6 alkoxy optionally substituted with: -one or more halogen substituents, —OCORx, where Rx is as defined above, -di-C1 to C6 alkyl-amino optionally substituted with C1 to C6 alkoxy, -5 or 6 membered heterocycle optionally substituted with C1 to C6 alkyl, or -5 or 6 membered heteroaryl; -amide; —NHSO2Rx, where Rx is as defined above; —NHSO2cyclopropyl; —NHSO2cyclobutyl; —NHSO2cyclopentyl; or —NHSO2cyclohexyl.
  • 7. The compound of claim 1, wherein R3 is hydrogen.
  • 8. A compound selected from the group consisting of:
  • 9. A pharmaceutical composition comprising a compound of claim 1 or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients.
  • 10. A pharmaceutical composition comprising a compound of claim 2 or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients.
  • 11. A pharmaceutical composition comprising a compound of claim 8 or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients.
  • 12. A compound of Formula (I):
  • 13. The compound of claim 12, wherein Y is:-aryl substituted with one or more of the following: —C1 to C6 alkoxy-nitro,—C1 to C6 alkyl-halogen;andZ is:—C1 to C6 alkyl optionally substituted with: —C1 to C6 alkoxy,-one or more halogen substituents, or-aryl.
  • 14. A pharmaceutical composition comprising a compound of claim 12 or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients.
Priority Claims (1)
Number Date Country Kind
PCT/US2005/024881 Jul 2005 US national
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of each of U.S. Provisional Application No. 60/587,487, filed Jul. 14, 2004, U.S. Provisional Application No. 60/634,979, filed Dec. 13, 2004, U.S. Provisional Application No. 60/645,586, filed Jan. 24, 2005, U.S. Provisional Application No. 60/665,349, filed Mar. 28, 2005, and U.S. Provisional Application No. 60/675,440, filed Apr. 28, 2005, all of which applications are incorporated herein by reference in their entireties. This application corresponds to International Application PCT/US2005/024881, filed Jul. 14, 2005, which application is herein incorporated by reference in its entirety.

GOVERNMENT SUPPORT

The present invention was made with U.S. Government support under DHHS Grant No. 5R44 AI054029-03. The U.S. Government has certain rights in the invention.

US Referenced Citations (73)
Number Name Date Kind
4788206 Guthrie et al. Nov 1988 A
4874756 Mertens et al. Oct 1989 A
5072003 Behrend et al. Dec 1991 A
5190942 Poss Mar 1993 A
5215980 Jones Jun 1993 A
5217996 Ksander Jun 1993 A
5354759 Oku et al. Oct 1994 A
5369120 Woodruff Nov 1994 A
5413999 Vacca et al. May 1995 A
5474994 Leonardi et al. Dec 1995 A
5527819 Williams et al. Jun 1996 A
5559127 Hartman et al. Sep 1996 A
5605896 Leonardi et al. Feb 1997 A
5633388 Diana et al. May 1997 A
5639906 London et al. Jun 1997 A
5681954 Yamamoto et al. Oct 1997 A
5693643 Gilbert et al. Dec 1997 A
5714496 Brown et al. Feb 1998 A
5880137 Miller et al. Mar 1999 A
5922898 Miller et al. Jul 1999 A
5958086 Adam et al. Sep 1999 A
5977090 Slusher et al. Nov 1999 A
5985910 Miller et al. Nov 1999 A
5994378 Matsuo et al. Nov 1999 A
6030785 Katze et al. Feb 2000 A
6057093 Han et al. May 2000 A
6124311 Chandrasekhar et al. Sep 2000 A
6132966 Draper Oct 2000 A
6194599 Miller et al. Feb 2001 B1
6221902 Malamas et al. Apr 2001 B1
6326392 Gast et al. Dec 2001 B1
6335445 Chabrier de Lassauniere et al. Jan 2002 B1
6358992 Pamukcu et al. Mar 2002 B1
6376529 Tang et al. Apr 2002 B1
6380166 Miller et al. Apr 2002 B1
6384022 Jackson et al. May 2002 B1
6555555 Konishi et al. Apr 2003 B1
6589570 Thyagarajan Jul 2003 B1
6589954 Mavunkel et al. Jul 2003 B1
6685931 Grint et al. Feb 2004 B1
6690975 Yamamoto et al. Feb 2004 B2
6974870 Cywin et al. Dec 2005 B2
20020055651 Moran et al. May 2002 A1
20020091116 Zhu et al. Jul 2002 A1
20020099054 Conner et al. Jul 2002 A1
20020099080 Gagliardi et al. Jul 2002 A1
20020103210 Furuya et al. Aug 2002 A1
20020143022 Pamukeu et al. Oct 2002 A1
20020169101 Gonzalez et al. Nov 2002 A1
20020169107 Rajagopalan et al. Nov 2002 A1
20030004119 Ganguly et al. Jan 2003 A1
20030050320 Hashimoto et al. Mar 2003 A1
20030078420 Chabrier de Lassauniere et al. Apr 2003 A1
20030096825 Wang et al. May 2003 A1
20030176433 Beaulieu et al. Sep 2003 A1
20030176697 Overman et al. Sep 2003 A1
20030199689 Nazare et al. Oct 2003 A1
20030220377 Chesworth Nov 2003 A1
20030232866 Watterson et al. Dec 2003 A1
20030236391 Klunk et al. Dec 2003 A1
20040044059 Pinney et al. Mar 2004 A1
20040059131 Dell et al. Mar 2004 A1
20040067996 Sheppeck Apr 2004 A1
20040180945 Artico et al. Sep 2004 A1
20040186125 Poupart et al. Sep 2004 A1
20050026969 Cheng et al. Feb 2005 A1
20050075242 Holtcamp et al. Apr 2005 A1
20050075384 Sheppeck et al. Apr 2005 A1
20050085529 Brown et al. Apr 2005 A1
20050113283 Solow-Cordero et al. May 2005 A1
20050119318 Hudyma et al. Jun 2005 A1
20050123560 Sinnott Jun 2005 A1
20050227291 Kinsella Oct 2005 A1
Foreign Referenced Citations (270)
Number Date Country
2099060 Dec 1993 CA
258014 Jun 1988 DE
258015 Jun 1988 DE
258016 Jun 1988 DE
0 406 734 Jan 1991 EP
0 414 386 Feb 1991 EP
0 427 225 May 1991 EP
0 430 186 Jun 1991 EP
0 436 199 Jul 1991 EP
0 471 372 Feb 1992 EP
470665 Feb 1992 EP
0 480 204 Apr 1992 EP
0 488 532 Jun 1992 EP
0 548 798 Jun 1993 EP
0 553 682 Aug 1993 EP
0 556 949 Aug 1993 EP
0 558 245 Sep 1993 EP
0 502 424 Jan 1994 EP
0 617 968 Oct 1994 EP
0 622 356 Nov 1994 EP
0 624 584 Nov 1994 EP
0 628 559 Dec 1994 EP
0 630 895 Dec 1994 EP
0 697 172 Feb 1996 EP
0 708 091 Apr 1996 EP
0 716 855 Jun 1996 EP
0 802 183 Oct 1997 EP
0 802 184 Oct 1997 EP
1 118 323 Jul 2001 EP
1 120 114 Aug 2001 EP
1 125 582 Aug 2001 EP
1 149 579 Oct 2001 EP
1 177 787 Feb 2002 EP
1 192 945 Apr 2002 EP
1 199 069 Apr 2002 EP
1 226 823 Jul 2002 EP
1 314 733 May 2003 EP
1 457 485 Sep 2004 EP
1 532 980 May 2005 EP
1 574 502 Sep 2005 EP
2 282 808 Apr 1995 GB
2 292 149 Feb 1996 GB
2002 105081 Apr 2002 JP
2005-194198 Jul 2005 JP
WO 9215579 Sep 1992 WO
9314758 Aug 1993 WO
WO 9318030 Sep 1993 WO
WO 9318765 Sep 1993 WO
WO 9318766 Sep 1993 WO
WO 9319067 Sep 1993 WO
WO 9404153 Mar 1994 WO
WO 9404535 Mar 1994 WO
WO 9408583 Apr 1994 WO
WO 9408962 Apr 1994 WO
WO 9411378 May 1994 WO
WO 9414435 Jul 1994 WO
WO 9414438 Jul 1994 WO
WO 9414763 Jul 1994 WO
WO 9414771 Jul 1994 WO
WO 9426746 Nov 1994 WO
WO 9502583 Jan 1995 WO
WO 9507910 Mar 1995 WO
WO 9514003 May 1995 WO
WO 9532710 Dec 1995 WO
WO 9533720 Dec 1995 WO
WO 9610012 Apr 1996 WO
9616054 May 1996 WO
WO 9626207 Aug 1996 WO
WO 9632379 Oct 1996 WO
WO 9640650 Dec 1996 WO
WO 9641800 Dec 1996 WO
WO 9714419 Apr 1997 WO
WO 9745410 Apr 1997 WO
WO 9813044 Apr 1998 WO
WO 9822457 May 1998 WO
WO 9825883 Jun 1998 WO
9848797 Nov 1998 WO
WO 9906836 Feb 1999 WO
WO 9911634 Mar 1999 WO
WO 9913714 Mar 1999 WO
WO 9918096 Apr 1999 WO
9924027 May 1999 WO
WO 9923072 May 1999 WO
WO 9926946 Jun 1999 WO
WO 9933849 Jul 1999 WO
WO9932482 Jul 1999 WO
9943651 Sep 1999 WO
WO 9950237 Oct 1999 WO
9958520 Nov 1999 WO
9959581 Nov 1999 WO
9959969 Nov 1999 WO
9964415 Dec 1999 WO
WO 9961426 Dec 1999 WO
WO 9964035 Dec 1999 WO
WO 0015645 Mar 2000 WO
WO 0028991 May 2000 WO
WO 0029384 May 2000 WO
WO 0035886 Jun 2000 WO
WO 0043393 Jul 2000 WO
WO 0061586 Oct 2000 WO
WO 0073269 Dec 2000 WO
0119839 Mar 2001 WO
0121609 Mar 2001 WO
WO 0119798 Mar 2001 WO
WO 0121589 Mar 2001 WO
0123353 Apr 2001 WO
WO 0123390 Apr 2001 WO
0144182 Jun 2001 WO
WO 0147883 Jul 2001 WO
0155136 Aug 2001 WO
0155137 Aug 2001 WO
0155138 Aug 2001 WO
0155139 Aug 2001 WO
0155144 Aug 2001 WO
WO 0155111 Aug 2001 WO
WO 0158859 Aug 2001 WO
WO 0164678 Sep 2001 WO
WO 0168585 Sep 2001 WO
WO 0174773 Oct 2001 WO
0190105 Nov 2001 WO
WO 0183451 Nov 2001 WO
WO 0185687 Nov 2001 WO
0203975 Jan 2002 WO
0203976 Jan 2002 WO
0203977 Jan 2002 WO
0203986 Jan 2002 WO
0203987 Jan 2002 WO
0203988 Jan 2002 WO
0203989 Jan 2002 WO
0203990 Jan 2002 WO
0203991 Jan 2002 WO
0203992 Jan 2002 WO
0204418 Jan 2002 WO
WO 0200651 Jan 2002 WO
WO 0206226 Jan 2002 WO
0213802 Feb 2002 WO
0216353 Feb 2002 WO
WO 0216333 Feb 2002 WO
0230358 Apr 2002 WO
WO 0226696 Apr 2002 WO
WO 0226703 Apr 2002 WO
WO 0230879 Apr 2002 WO
0236562 May 2002 WO
0242292 May 2002 WO
WO 0236203 May 2002 WO
WO 0236580 May 2002 WO
WO 0248099 Jun 2002 WO
WO 02051805 Jul 2002 WO
WO 02053534 Jul 2002 WO
WO 02055496 Jul 2002 WO
02059088 Aug 2002 WO
02059120 Aug 2002 WO
WO 02060374 Aug 2002 WO
WO 02060447 Aug 2002 WO
WO 02066477 Aug 2002 WO
02072549 Sep 2002 WO
WO 02070462 Sep 2002 WO
WO 02070469 Sep 2002 WO
WO 02070510 Sep 2002 WO
WO 02074742 Sep 2002 WO
WO 02076926 Oct 2002 WO
WO 02083134 Oct 2002 WO
02089811 Nov 2002 WO
WO 02053565 Nov 2002 WO
WO 02096426 Dec 2002 WO
WO 02098424 Dec 2002 WO
WO 03000254 Jan 2003 WO
WO 03000690 Jan 2003 WO
WO 03004458 Jan 2003 WO
WO 03005025 Jan 2003 WO
WO 03006447 Jan 2003 WO
WO 03010140 Feb 2003 WO
WO 03010141 Feb 2003 WO
WO 03022214 Mar 2003 WO
WO 03048101 Jun 2003 WO
WO 03053359 Jul 2003 WO
WO 03053368 Jul 2003 WO
WO 03053938 Jul 2003 WO
WO 03053941 Jul 2003 WO
WO 03055447 Jul 2003 WO
WO 03059269 Jul 2003 WO
WO 03062392 Jul 2003 WO
WO 03064539 Aug 2003 WO
WO 03066629 Aug 2003 WO
WO 03074047 Sep 2003 WO
WO 03082265 Oct 2003 WO
WO 03087092 Oct 2003 WO
03091211 Nov 2003 WO
WO 03097036 Nov 2003 WO
2005058315 Dec 2003 WO
WO 03099276 Dec 2003 WO
WO 2004003103 Jan 2004 WO
2004014912 Feb 2004 WO
WO 2004012736 Feb 2004 WO
WO 2004013135 Feb 2004 WO
WO 2004022057 Mar 2004 WO
WO 2004024060 Mar 2004 WO
WO 2004024655 Mar 2004 WO
WO 2004024896 Mar 2004 WO
WO 2004030630 Apr 2004 WO
WO 2004035047 Apr 2004 WO
WO 2004035522 Apr 2004 WO
WO 2004035525 Apr 2004 WO
WO 2004035571 Apr 2004 WO
2004037788 May 2004 WO
2004041256 May 2004 WO
WO 2004037791 May 2004 WO
WO 2004041781 May 2004 WO
WO 2004050035 Jun 2004 WO
WO 2004064759 Aug 2004 WO
WO 2004064925 Aug 2004 WO
WO 2004065367 Aug 2004 WO
2004082638 Sep 2004 WO
WO 2004074447 Sep 2004 WO
WO 2004083195 Sep 2004 WO
WO 2004087714 Oct 2004 WO
WO 2004093871 Nov 2004 WO
WO 2004093912 Nov 2004 WO
WO 2004094409 Nov 2004 WO
WO 2004096210 Nov 2004 WO
WO 2004099168 Nov 2004 WO
WO 2004099170 Nov 2004 WO
WO 2004099171 Nov 2004 WO
WO 2004099192 Nov 2004 WO
WO 2004099239 Nov 2004 WO
WO 2004111056 Dec 2004 WO
WO 2005003086 Jan 2005 WO
WO 2005003131 Jan 2005 WO
2005009389 Feb 2005 WO
2005013976 Feb 2005 WO
2005013977 Feb 2005 WO
2005014000 Feb 2005 WO
2005014045 Feb 2005 WO
WO 2005013950 Feb 2005 WO
WO 2005014543 Feb 2005 WO
WO 2005016862 Feb 2005 WO
2005021505 Mar 2005 WO
WO 2005018531 Mar 2005 WO
WO 2005020899 Mar 2005 WO
WO 2005020921 Mar 2005 WO
WO 2005028502 Mar 2005 WO
WO 2005034941 Apr 2005 WO
WO 2005034943 Apr 2005 WO
WO 2005039489 May 2005 WO
WO 2005042018 May 2005 WO
WO 2005055940 Jun 2005 WO
WO 2005061519 Jul 2005 WO
WO 2005062676 Jul 2005 WO
WO 2005066180 Jul 2005 WO
WO 2005072132 Aug 2005 WO
WO 2005076861 Aug 2005 WO
WO 2005077122 Aug 2005 WO
WO 2005077969 Aug 2005 WO
2005090282 Sep 2005 WO
WO 2005080335 Sep 2005 WO
WO 2005080388 Sep 2005 WO
WO 2005082895 Sep 2005 WO
WO 2005082905 Sep 2005 WO
WO 2005086754 Sep 2005 WO
WO 2005092855 Oct 2005 WO
2005107747 Nov 2005 WO
2005112519 Nov 2005 WO
2005113529 Dec 2005 WO
2006019831 Feb 2006 WO
2006024699 Mar 2006 WO
2006041874 Apr 2006 WO
2006049013 May 2006 WO
2006050236 May 2006 WO
2006057354 Jun 2006 WO
2006083458 Aug 2006 WO
Related Publications (2)
Number Date Country
20060223863 A1 Oct 2006 US
20080096928 A9 Apr 2008 US
Provisional Applications (5)
Number Date Country
60587487 Jul 2004 US
60634979 Dec 2004 US
60645586 Jan 2005 US
60665349 Mar 2005 US
60675440 Apr 2005 US