WO2013021344A1 - Imidazole derivatives as antiviral agents - Google Patents

Imidazole derivatives as antiviral agents Download PDF

Info

Publication number
WO2013021344A1
WO2013021344A1 PCT/IB2012/054020 IB2012054020W WO2013021344A1 WO 2013021344 A1 WO2013021344 A1 WO 2013021344A1 IB 2012054020 W IB2012054020 W IB 2012054020W WO 2013021344 A1 WO2013021344 A1 WO 2013021344A1
Authority
WO
WIPO (PCT)
Prior art keywords
imidazol
amino
methyl
alkyl
cyclopentyl
Prior art date
Application number
PCT/IB2012/054020
Other languages
French (fr)
Inventor
Vidya Ramdas
Advait Arun JOSHI
Moloy Manoj BANERJEE
Amit Kumar DAS
Deepak Sahebrao WALKE
Venkata P. Palle
Rajender Kumar Kamboj
Original Assignee
Lupin Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Lupin Limited filed Critical Lupin Limited
Publication of WO2013021344A1 publication Critical patent/WO2013021344A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems

Definitions

  • the present invention is related to novel compounds of the general formula I,
  • HCV Persistent hepatitis C virus
  • HCV chronic infection is often asymptomatic with latent periods lasting for decades before manifestation by which time extensive liver damage has occurred.
  • HCV is spread primarily by unscreened blood transfusions and use of contaminated needles and syringes; the highest risk groups are intravenous drug users and people who received blood transfusions (mainly haemophiliacs) before 1990 when screening for HCV was introduced.
  • Factors that have been reported to influence the rate of HCV disease progression include age (increasing age is associated with more rapid progression), gender (males have more rapid disease progression than females), alcohol consumption (associated with an increased rate of disease progression), HIV co-infection (associated with a markedly increased rate of disease progression), and fatty liver.
  • HCV exists as many closely related viral sequences, termed as quasi-species, in the infected individual, making specific pharmaceutical targeting of HCV proteins challenging due to the rapid evolution of escape mutants. It is increasingly evident that a broad collection of specific, pan genotypic anti- viral drugs targeting multiple essential viral functions, in addition to the current viral therapies will be required for effective global control of HCV.
  • the present invention provides a pharmaceutical composition, containing the compound of the general formula (I) as defined herein, its tautomeric forms, its stereoisomers, its analogs, its prodrugs, its isotopes, its N-oxides, its metabolites, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates, or its co-crystals in combination with the usual pharmaceutically employed carriers, diluents and the like are useful for the treatment of HCV infection.
  • HCV is a member of the Flaviviridae family of enveloped, positive stranded RNA viruses belonging to the genus Hepacivirus.
  • the genome is a single ⁇ 9.6kb strand of RNA and consists of one open reading frame that encodes for a polyprotein of -3000 amino acids flanked by untranslated regions at both 5' and 3' ends.
  • This precursor polyprotein is then processed by viral and cellular proteases to yield 10 separate mature viral proteins critical for replication and assembly of progeny viral particles.
  • the organization of the structural and non-structural proteins in the HCV polyprotein is as follows: C-E1 -E2-P7-NS2-NS3- NS4a-NS4b-NS5a-NS5b.
  • NS2 is a zinc dependent metalloproteinase that functions in conjunction with a part of NS3 protein.
  • NS3 protein has two catalytic activities associated with it: a serine protease at the N-terminal end which requires NS4A as a cofactor, and an ATPase dependent helicase activity at the C-terminal end.
  • NS5A is a membrane anchored phosphoprotein that is present in basally phosphorylated (56kDa) and hyperphosphorylated (58kDa) forms.
  • NS5A plays an important role in replication and infectivity of HCV.
  • the NS5B protein encodes an RNA dependent RNA polymerase activity, key to the generation of progeny viruses. While the pathology of HCV infection mainly affects the liver, the virus is found in other cell types in the body including peripheral blood lymphocytes [Thomson BJ et al., Clin Microbial Infect. 2005, 1J., 86-94; Moriishi K et al., Antivir. Chem. Chemother. 2003, 14, 285-297].
  • Characterization of the replicase machinery required for HCV RNA synthesis has defined the protease/helicase NS3 protein, the NS4A cofactor, the NS4B integral membrane protein, the NS5A protein and the RNA dependent RNA polymerase NS5B as being its essential components.
  • Ring 'A' is a saturated carbocycle, which may be a monocyclic system or may be a fused carbocycle or may be a bridged carbocycle, the said ring A may contain 5 to 10 carbons; Ring 'D' is selected from 5 to 10 membered carbocycle and 5 to 10 membered
  • the ring 'D' may be monocyclic, fused bicyclic, bridged bicyclic or spiro bicyclic;
  • Y is selected from -CH(R 2a )- and -N(R 2 )-;
  • R 5 , R 6 , R 7 , R 8 , R 9 and R 10 are independently selected as hydrogen, or R 5 and R 6 together, R 7 and R 10 together, or R 8 and R 9 together independently along with the carbon atoms to which they are attached forming 5 to 8 membered substituted- or unsubstituted- carbocycle, 5 to 8 membered substituted- or unsubstituted- heterocycle, 6 membered substituted- or unsubstituted- aryl, or 5 to 6 membered substituted- or unsubstituted- heteroaryl; with a proviso that the compound of formula I must have at least one cyclic system formed out of either R 5 and R 6 , R 8 and R 9 , or R 7 and R 10 ; also provided that R 7 and R 10 take part in formation of cyclic system only when n is 0;
  • R 11 and R 12 are independently selected from a group consisting of hydrogen, halogen, substituted- or unsubstituted- Ci- 6 alkyl, R 13b O-, and (R 13 )(R 14 )N-; wherein, R 13 , R 14 , R 14a and R 15 are independently selected from hydrogen, substituted- or unsubstituted- Ci -6 alkyl, substituted- or unsubstituted- aryl, substituted- or unsubstituted- heteroaryl, substituted- or unsubstituted- cycloalkyl, and substituted- or unsubstituted- heterocyclyl; R 13a is selected from substituted- or unsubstituted- Ci -6 alkyl, perhaloalkyl, substituted- or unsubstituted- aryl, substituted- or unsubstituted- heteroaryl, substituted- or unsubstituted- cycloalkyl, and substituted- or unsubstitute
  • R 13b is selected from hydrogen, substituted- or unsubstituted- Ci -6 alkyl, perhaloalkyl, substituted- or unsubstituted- aryl, substituted- or unsubstituted- heteroaryl, substituted- or unsubstituted- cycloalkyl, and substituted- or unsubstituted- heterocyclyl;
  • R 16 is selected from hydrogen or substituted- or unsubstituted alkyl group
  • R a , R b , R c and R d are independently selected from hydrogen, halogen, substituted- or unsubstituted- Ci -6 alkyl, substituted- or unsubstituted- aryl, substituted- or unsubstituted- heteroaryl, substituted- or unsubstituted- cycloalkyl, and substituted- or unsubstituted- heterocyclyl, or R a , R b , R c and R d together with the carbon atom(s) to which they are attached forming substituted- or unsubstituted- carbocycle, substituted- or unsubstituted- heterocycle; m is an integer ranging between 0 to 2, selected independently at each occurrence; n is an integer ranging between 0 and 2; 'alkyl' may be substituted with 1 to 4 substituents selected from the group consisting of halogen, oxo, Ci- 6 alkyl, haloalkyl,
  • R 17 , R 18 and R 19 are independently selected from hydrogen and alkyl;
  • R 17a is selected from alkyl and perhaloalkyl;
  • R 17b is selected from the group consisting of hydrogen, alkyl, and perhaloalkyl
  • any sub-range or individual number of carbon atoms falling within the indicated range also can be used.
  • One of the embodiments of the present invention is compound of formula (I) as described above, wherein, ring A is particularly selected as cyclopentane;
  • Y is particularly selected as - N(R 2 )-;
  • m is particularly selected as 0 at all the occurrences
  • R 3 is particularly selected from NH and O;
  • n is particularly selected as 0;
  • R 5 and R 6 are independently selected from hydrogen and halogen, or R 5 and R 6 together with the carbon atoms to which they are attached form a six membered carbocycle;
  • R 7 and R 10 are particularly selected as hydrogen, or R 7 and R 10 together with the carbon atoms to which they are attached form a five or six membered carbocycle, the said carbocycle is unsubstituted or substituted with one or two alkyl groups;
  • R 9 and R 8 are particularly selected as hydrogen, or R 9 and R 8 together with the carbon atoms to which they are attached form a six or seven membered carbocycle, or R 9 and R 8 together with the carbon atoms to which they are attached form a seven membered heterocycle containing one heteroatom;
  • R 11 and R 12 are particularly selected as hydrogen; In any of the embodiments of the invention described above, ring D is particularly selected as
  • ring A is particularly selected
  • m is particularly selected as 0 at all the occurrences
  • R 3 is particularly selected from NH and O
  • n is particularly selected as 0,
  • R 5 and R 6 are independently selected from hydrogen and halogen, or R 5 and R 6 together with the carbon atoms to which they are attached form a six membered carbocycle
  • R 7 and R 10 are particularly selected as hydrogen, or R 7 and R 10 together with the carbon atoms to which they are attached form a five or six membered carbocycle, the said carbocycle is unsubstituted or substituted with one or two alkyl groups
  • R 9 and R 8 are particularly selected as hydrogen, or R 9 and R 8 together with the carbon atoms to which they are attached form a six or seven membered carbocycle, or R 9 and R 8 together with the carbon
  • alkyl means a straight chain or branched hydrocarbon containing from 1 to 20 carbon atoms. Preferably the alkyl chain may contain 1 to 10 carbon atoms. More preferably alkyl chain may contain up to 6 carbon atoms. Representative examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, and n- hexyl.
  • haloalkyl used herein means an alkyl group as defined hereinabove wherein at least one of the hydrogen atoms of the said alkyl group is substituted with halogen.
  • the haloalkyl group is exemplified by monofluoromethyl, 1 ,2-dichloroethyl and the like.
  • perhaloalkyl means an alkyl group as defined hereinabove wherein all the hydrogen atoms of the said alkyl group are substituted with halogen.
  • the perhaloalkyl group is exemplified by trifluoromethyl, pentafluoroethyl and the like.
  • cycloalkyi means a monocyclic, bicyclic, or tricyclic non- aromatic ring system containing from 3 to 14 carbon atoms, preferably monocyclic cycloalkyi ring containing 3 to 6 carbon atoms.
  • monocyclic ring systems include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • Bicyclic ring systems are also exemplified by a bridged monocyclic ring system in which two non-adjacent carbon atoms of the monocyclic ring are linked by an alkylene bridge.
  • bicyclic ring systems include, but are not limited to, bicyclo[3.1 .1 ]heptane, bicyclo[2.2.1 ]heptane, bicyclo[2.2.2]octane, bicyclo[3.2.2]nonane, bicyclo[3.3.1 ]nonane, and bicyclo[4.2.1 ]nonane, bicyclo[3.3.2]decane, bicyclo[3.1 .0]hexane, bicyclo[410]heptane, bicyclo[3.2.0]heptanes, octahydro-1 H-indene.
  • Tricyclic ring systems are also exemplified by a bicyclic ring system in which two non- adjacent carbon atoms of the bicyclic ring are linked by a bond or an alkylene bridge.
  • Representative examples of tricyclic-ring systems include, but are not limited to, tricyclo[3.3.1 .0 3 7 ]nonane and tricyclo[3.3.1 .1 3 7 ]decane (adamantane).
  • cycloalkyl also include spiro systems wherein one of the ring is annulated on a single carbon atom such ring systems are exemplified by spiro[2.5]octane, spiro[4.5]decane, spiro[bicyclo[4.1 .0]heptane-2,1 '-cyclopentane], hexahydro-2'H-spiro[cyclopropane-1 ,1 '- pentalene].
  • cycloalkenyl as used herein, means a cycloalkyl group as defined above containing at least one double bond.
  • carrier means a cyclic system made up of carbon atoms, which includes cycloalkyl, cycloalkenyl and aryl.
  • aryl refers to a monovalent monocyclic, bicyclic or tricyclic aromatic hydrocarbon ring system.
  • aryl groups include phenyl, naphthyl, anthracenyl, fluorenyl, indenyl, azulenyl, and the like.
  • Aryl group also include partially saturated bicyclic and tricyclic aromatic hydrocarbons such as tetrahydro-naphthalene.
  • the said aryl group also includes aryl rings fused with heteroaryl or heterocyclic rings such as 2,3-dihydro- benzo[1 ,4]dioxin-6-yl, 2,3-dihydro-benzo[1 ,4]dioxin-5-yl, 2,3-dihydro-benzofuran-5-yl, 2,3- dihydro-benzofuran-4-yl, 2,3-dihydro-benzofuran-6-yl, 2,3-dihydro-benzofuran-6-yl, 2,3- dihydro-1 H-indol-5-yl, 2,3-dihydro-1 H-indol-4-yl, 2,3-dihydro-1 H-indol-6-yl, 2,3-dihydro-1 H- indol-7-yl, benzo[1 ,3]dioxol-4-yl, benzo[1 ,3]dioxol-5-yl
  • substituents selected independently from the group consisting of halogen, nitro, cyano, hydroxy, alkyl, perhaloalkyl, alkyl-O-,
  • heteroaryl refers to a 5-14 membered monocyclic, bicyclic, or tricyclic ring system having 1 -4 ring heteroatoms selected from O, N, or S, and the remainder ring atoms being carbon (with appropriate hydrogen atoms unless otherwise indicated), wherein at least one ring in the ring system is aromatic.
  • Heteroaryl groups may be optionally substituted with one or more substituents. In one embodiment, 0, 1 , 2, 3, or 4 atoms of each ring of a heteroaryl group may be substituted by a substituent.
  • heteroaryl groups include pyridyi, 1 -oxo-pyridyl, furanyi, thienyi, pyrroiyl, oxazolyi, oxadiazolyl, imidazolyl, thiazolyl, isoxazolyl, quinolinyl, pyrazolyl, isothiazolyl, pyridazinyl, pyrimidinyi, pyrazinyl, triazinyl. triazolyl.
  • thiadiazolyl isoquinolinyl, benzoxazolyl, benzofuranyl, indolizinyl, imidazopyridyl, tetrazolyi, benzimidazolyi, benzothiazolyl. benzothiadiazolyl, benzoxadiazolyl, indolyl. azaindolyl, imidazopyridyl. quinazolinyl.
  • substituents selected independently from the group consisting of halogen, nitro, cyano, hydroxy, alkyl, perhaloalkyl, alkyl
  • heterocycle or “heterocyclic” as used herein, means a 'cycloalkyl' group wherein one or more of the carbon atoms replaced by -0-, -S-, -S(0 2 )-, -S(O)-, -N(R m )-, - Si(R m )R n -, wherein, R m and R n are independently selected from hydrogen, alkyl, aryl, heteroaryl, cycloalkyi, and heterocyclyl.
  • the heterocycle may be connected to the parent molecular moiety through any carbon atom or any nitrogen atom contained within the heterocycle.
  • monocyclic heterocycle include, but are not limited to, azetidinyl, azepanyl, aziridinyl, diazepanyl, 1 ,3-dioxanyl, 1 ,3-dioxolanyl, 1 ,3- dithiolanyl, 1 ,3-dithianyl, imidazolinyl, imidazolidinyl, isothiazolinyl, isothiazolidinyl, isoxazolinyl, isoxazolidinyl, morpholinyl, oxadiazolinyl.
  • oxadiazolidinyl oxazolinyl, oxazolidinyl, piperazinyl, piperidinyl, pyranyl, pyrazolinyl, pyrazolidinyl.
  • thiopyranyl, and trithianyl thiopyranyl, and trithianyl.
  • bicyclic heterocycle include, but are not limited to 1 ,3-benzodioxolyl, 1 ,3-benzodithiolyl, 2,3-dihydro-1 ,4-benzodioxinyl, 2,3- dihydro-1 -benzofuranyl, 2,3-dihydro-1 -benzothienyl, 2,3-dihydro-1 H-indolyl and 1 ,2,3,4- tetrahydroquinolinyl.
  • the term heterocycle also include bridged heterocyclic systems such as azabicyclo[3.2.1 ]octane, azabicyclo[3.3.1 ]nonane and the like.
  • oxo attached to carbon forms a carbonyl
  • oxo substituted on cyclohexane forms a cyclohexanone
  • 'annulated' means the ring system under consideration is either annulated with another ring at a carbon atom of the cyclic system or across a bond of the cyclic system as in the case of fused or spiro ring systems.
  • bridged' means the ring system under consideration contain an alkylene bridge having 1 to 4 methylene units joining two non adjuscent ring atoms.
  • the compounds of general formula I where all the symbols are as defined earlier can be prepared by methods given in Schemes given below or example. Representative procedures are shown below, however; the disclosure should not be construed to limit the scope of the invention arriving at compound of formula I as disclosed hereinabove.
  • Cyclization of 4 using NH 4 OAc could lead to the formation of 5 which may be deprotected and coupled with (S)-2-((methoxycarbonyl)amino)-3-methylbutanoic acid using amide coupling reagents like carbodiimides, phosphonium salts, uronium salts, carbonyldiimidazole in presence of a mild base like triethylamine, 4-methylmorpholine, diisopropylethylamine in a solvent like DMF, 1 ,4-dioxane, THF, dichloromethane leading to the formation of 6.
  • de-protected form of 5 can be coupled to similar moieties by methods known in the literature to furnish compounds of formula I.
  • oxime 35 On treatment with isoamyl nitrite in an appropriate solvent, oxime 35 might be generated from 34 which upon reaction with Boc-L-prolinal under basic conditions may lead to formation of 36. Treatment of 36 with triethyl phosphite might lead to the formation of either 37 or 38. Pd-catalyzed C-C coupling of either 37 or 38 with 25 followed by de-protection and coupling with (S)-2- ((methoxycarbonyl)amino)-3-methylbutanoic acid might lead to the formation of 39 or 40.
  • Symmetrical compounds could be prepared by coupling 42 (generated from 34) with the boronate/boronic acid of 42 through the use of Pd catalyzed C-C coupling reactions to yield 44 or compounds of formula I.
  • a further embodiment of the present invention includes pharmaceutical compositions comprising any single compound, a combination of two or more compounds delineated herein, or a pharmaceutically acceptable salt thereof, with a pharmaceutically acceptable carrier or excipient.
  • a further embodiment of the present invention is a pharmaceutical composition
  • a pharmaceutical composition comprising any single compound or a combination of two or more compounds delineated herein, or a pharmaceutically acceptable salt thereof, in combination with one or more agents known in the art, with a pharmaceutically acceptable carrier or excipient.
  • compounds of the present invention can be administered as the sole active pharmaceutical agent, or used in combination with one or more agents to treat or prevent hepatitis C infections or the symptoms associated with HCV infection.
  • Other agents to be administered in combination with a compound or combination of compounds of the present invention include therapies for diseases caused by HCV infection that suppresses HCV viral replication by direct or indirect mechanisms.
  • agents include, but not limited to, host immune modulators (for example, interferon-alpha, pegylated interferon-alpha, consensus interferon, interferon-beta, interferon-gamma, CpG oligonucleotides and the like); antiviral compounds that inhibit host cellular functions such as inosine monophosphate dehydrogenase (for example, ribavirin and the like); cytokines that modulate immune function (for example, interleukin 2, interleukin 6, and interleukin 12); a compound that enhances the development of type 1 helper T cell response; interfering RNA; anti-sense RNA; vaccines comprising HCV antigens or antigen adjuvant combinations directed against HCV; agents that interact with host cellular components to block viral protein synthesis by inhibiting the internal ribosome entry site (IRES) initiated translation step of HCV viral replication or to block viral particle maturation and release with agents targeted toward the viroporin family of membrane proteins such as, for example
  • compositions of the present invention may further comprise inhibitor(s) of other targets in the HCV life cycle, including, but not limited to, helicase, polymerase, metalloprotease, NS4A protein, NS5A protein, and internal ribosome entry site (IRES).
  • inhibitor(s) of other targets in the HCV life cycle including, but not limited to, helicase, polymerase, metalloprotease, NS4A protein, NS5A protein, and internal ribosome entry site (IRES).
  • one embodiment of the present invention is directed to a method for treating or preventing an infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment one or more agents selected from the group consisting of a host immune modulator and a second or more antiviral agents, or a combination thereof, with a therapeutically effective amount of a compound or combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof.
  • Examples of the host immune modulator are, but not limited to, interferon-alpha, pegylated-interferon-alpha, interferon-beta, interferon-gamma, a cytokine, a vaccine, and a vaccine comprising an antigen and an adjuvant, and said second antiviral agent inhibits replication of HCV either by inhibiting host cellular functions associated with viral replication or by targeting proteins of the viral genome.
  • Example of the RNA-containing virus includes, but not limited to, hepatitis C virus (HCV).
  • a further embodiment of the present invention is directed to a method of treating or preventing infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment an agent or combination of agents that treat or alleviate symptoms of HCV infection including cirrhosis and inflammation of the liver, with a therapeutically effective amount of a compound or combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof.
  • RNA-containing virus includes, but not limited to, hepatitis C virus (HCV).
  • Yet another embodiment of the present invention provides a method of treating or preventing infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment one or more agents that treat patients for disease caused by hepatitis B (HBV) infection, with a therapeutically effective amount of a compound or a combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof.
  • An agent that treats patients for disease caused by hepatitis B (HBV) infection may be for example, but not limited thereto, L-deoxythymidine, adefovir, lamivudine or tenfovir, or any combination thereof.
  • Example of the RNA-containing virus includes, but not limited to, hepatitis C virus (HCV).
  • a further embodiment of the present invention provides a method of treating or preventing infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment one or more agents that treat patients for disease caused by human immunodeficiency virus (HIV) infection, with a therapeutically effective amount of a compound or a combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof.
  • HIV human immunodeficiency virus
  • the agent that treats patients for disease caused by human immunodeficiency virus (H IV) infection may include, but is not limited thereto, ritonavir, lopinavir, indinavir, nelfmavir, saquinavir, amprenavir, atazanavir, tipranavir, TMC-1 14, fosamprenavir, zidovudine, lamivudine, didanosine, stavudine, tenofovir, zalcitabine, abacavir, efavirenz, nevirapine, delavirdine, TMC-125, L-870812, S-1360, enfuvirtide (T- 20) or T-1249, or any combination thereof.
  • RNA-containing virus includes, but not limited to, hepatitis C virus (HCV). It can occur that a patient may be co-infected with hepatitis C virus and one or more other viruses, including but not limited to human immunodeficiency virus (HIV), hepatitis A virus (HAV) and hepatitis B virus (HBV). Thus also contemplated is combination therapy to treat such co-infections by co-administering a compound according to the present invention with at least one of an HIV inhibitor, an HAV inhibitor and an HBV inhibitor.
  • HCV human immunodeficiency virus
  • HAV hepatitis A virus
  • HBV hepatitis B virus
  • the present invention provides the use of a compound or a combination of compounds of the invention, or a therapeutically acceptable salt thereof, and one or more agents selected from the group consisting of a host immune modulator and a second or more antiviral agents, or a combination thereof, to prepare a medicament for the treatment of an infection caused by an RNA-containing virus in a patient, particularly hepatitis C virus.
  • host immune modulators include, but are not limited to, interferon- alpha, pegylated-interferon-alpha, interferon-beta, interferon-gamma, a cytokine, a vaccine, and a vaccine comprising an antigen and an adjuvant, and said second antiviral agent inhibits replication of HCV either by inhibiting host cellular functions associated with viral replication or by targeting proteins of the viral genome.
  • combination of compound or compounds of the present invention, together with one or more agents as defined herein above can be employed in pure form or, where such forms exist, in pharmaceutically acceptable salt thereof.
  • combination of therapeutic agents can be administered as a pharmaceutical composition containing a therapeutically effective amount of the compound or combination of compounds of interest, or their pharmaceutically acceptable salt thereof, in combination with one or more agents as defined hereinabove, and a pharmaceutically acceptable carrier.
  • Such pharmaceutical compositions can be used for inhibiting the replication of an RNA-containing virus, particularly Hepatitis C virus (HCV), by contacting said virus with said pharmaceutical composition.
  • such compositions are useful for the treatment or prevention of an infection caused by an RNA-containing virus, particularly Hepatitis C virus (HCV).
  • a still further embodiment of the invention is directed to a method of treating or preventing infection caused by an RNA-containing virus, particularly a hepatitis C virus (HCV), comprising administering to a patient in need of such treatment a pharmaceutical composition comprising a compound or combination of compounds of the invention or a pharmaceutically acceptable salt thereof, and one or more agents as defined hereinabove, with a pharmaceutically acceptable carrier.
  • an RNA-containing virus particularly a hepatitis C virus (HCV)
  • HCV hepatitis C virus
  • the therapeutic agents When administered as a combination, the therapeutic agents can be formulated as separate compositions which are given at the same time or within a predetermined period of time, or the therapeutic agents can be given as a single unit dosage form.
  • Antiviral agents contemplated for use in such combination therapy include agents (compounds or biologicals) that are effective to inhibit the formation and/or replication of a virus in a mammal, including but not limited to agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of a virus in a mammal.
  • agents can be selected from another anti-HCV agent; an HIV inhibitor; an HAV inhibitor; and an HBV inhibitor.
  • agents to be administered in combination with a compound of the present invention include a cytochrome P450 monooxygenase inhibitor, which is expected to inhibit metabolism of the compounds of the invention. Therefore, the cytochrome P450 monooxygenase inhibitor would be in an amount effective to inhibit metabolism of the compounds of the present invention. Accordingly, the CYP inhibitor is administered in an amount such that the bioavailability of the compounds of the present invention is increased in comparison to the bioavailability in the absence of the CYP inhibitor.
  • room temperature used in the specification denotes any temperature ranging between about 20 °C to about 40 °C, except and otherwise it is specifically mentioned in the specification.
  • the intermediates and the compounds of the present invention may obtained in pure form in a manner known per se, for example, by distilling off the solvent in vacuum and re- crystallizing the residue obtained from a suitable solvent, such as pentane, diethyl ether, isopropyl ether, chloroform, dichloromethane, ethyl acetate, acetone or their combinations or subjecting it to one of the purification methods, such as column chromatography (e.g., flash chromatography) on a suitable support material such as alumina or silica gel using eluent such as dichloromethane, ethyl acetate, hexane, methanol, acetone and their combinations.
  • a suitable solvent such as pentane, diethyl ether, isopropyl ether, chloroform, dichloromethane, ethyl acetate, acetone or their combinations
  • the purification methods such as column chromatography (e.g., flash
  • Salts of compound of formula I can be obtained by dissolving the compound in a suitable solvent, for example in a chlorinated hydrocarbon, such as methyl chloride or chloroform or a low molecular weight aliphatic alcohol, for example, ethanol or isopropanol, which was then treated with the desired acid or base as described in Berge S.M. et al. "Pharmaceutical Salts, a review article in Journal of Pharmaceutical sciences volume 66, page 1 -19 (1977)" and in handbook of pharmaceutical salts properties, selection, and use by P.H.Einrich Stahland Camille G.wermuth, Wiley- VCH (2002).
  • a suitable solvent for example in a chlorinated hydrocarbon, such as methyl chloride or chloroform or a low molecular weight aliphatic alcohol, for example, ethanol or isopropanol
  • salts can also be found in Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing Company, Easton, PA, 1990, p. 1445, and Journal of Pharmaceutical Science, 66, 2-19 (1977).
  • they can be a salt of an alkali metal (e.g., sodium or potassium), alkaline earth metal (e.g., calcium), or ammonium of salt.
  • the compound of the invention or a composition thereof can potentially be administered as a pharmaceutically acceptable acid-addition, base neutralized or addition salt, formed by reaction with inorganic acids, such as hydrochloric acid, hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, and phosphoric acid, and organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid, malonic acid, succinic acid, maleic acid, and fumaric acid, or by reaction with an inorganic base, such as sodium hydroxide, potassium hydroxide.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, and phosphoric acid
  • organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid, mal
  • the conversion to a salt is accomplished by treatment of the base compound with at least a stoichiometric amount of an appropriate acid.
  • the free base is dissolved in an inert organic solvent such as diethyl ether, ethyl acetate, chloroform, ethanol, methanol, and the like, and the acid is added in a similar solvent.
  • the mixture is maintained at a suitable temperature (e.g., between 0 °C and 50 °C).
  • the resulting salt precipitates spontaneously or can be brought out of solution with a less polar solvent.
  • the stereoisomers of the compounds of formula I of the present invention may be prepared by stereospecific syntheses or resolution of the achiral compound using an optically active amine, acid or complex forming agent, and separating the diastereomeric salt/complex by fractional crystallization or by column chromatography.
  • prodrug denotes a derivative of a compound, which derivative, when administered to warm-blooded animals, e.g. humans, is converted into the compound (drug).
  • the enzymatic and/or chemical hydrolytic cleavage of the compounds of the present invention occurs in such a manner that the proven drug form (parent carboxylic acid drug) is released, and the moiety or moieties split off remain nontoxic or are metabolized so that nontoxic metabolic products are produced.
  • a carboxylic acid group can be esterified, e.g., with a methyl group or ethyl group to yield an ester.
  • an ester is administered to a subject, the ester is cleaved, enzymatically or non- enzymatically, reductively, oxidatively, or hydrolytically, to reveal the anionic group.
  • An anionic group can be esterified with moieties (e.g., acyloxymethyl esters) which are cleaved to reveal an intermediate compound which subsequently decomposes to yield the active compound.
  • the prodrugs can be prepared in situ during the isolation and purification of the compounds, or by separately reacting the purified compound with a suitable derivatizing agent.
  • hydroxy groups can be converted into esters via treatment with a carboxylic acid in the presence of a catalyst.
  • cleavable alcohol prodrug moieties include substituted or unsubstituted, branched or unbranched lower alkyl ester moieties, e.g., ethyl esters, di-lower alkylamino lower-alkyl esters, e.g., dimethylaminoethyl ester, acylamino lower alkyl esters, acyloxy lower alkyl esters (e.g., pivaloyloxymethyl ester), aryl esters, e.g., phenyl ester, aryl-lower alkyl esters, e.g., benzyl ester, optionally substituted, e.g., with methyl, halo, or methoxy substituents aryl and aryl- lower alkyl esters, amides, lower-alkyl amides, di-lwer alkyl amides, and hydroxy amides.
  • lower alkyl ester moieties
  • the present invention further provides a pharmaceutical composition, containing the compounds of the general formula (I) as defined above, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates and its co-crystals in combination with the usual pharmaceutically acceptable carriers, diluents and the like.
  • the pharmaceutically acceptable carrier is preferably one that is chemically inert to the compound of the invention and one that has no detrimental side effects or toxicity under the conditions of use.
  • Such pharmaceutically acceptable carriers preferably include saline (e.g., 0.9% saline), Cremophor EL (which is a derivative of castor oil and ethylene oxide available from Sigma Chemical Co., St.
  • a preferred pharmaceutical carrier is polyethylene glycol, such as PEG 400, and particularly a composition comprising 40% PEG 400 and 60% water or saline. The choice of carrier will be determined in part by the particular compound chosen, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of the pharmaceutical composition of the present invention.
  • formulations for oral, aerosol, parenteral, subcutaneous, intravenous, intraarterial, intramuscular, interperitoneal, rectal, and vaginal administration are merely exemplary and are in no way limiting.
  • compositions for parenteral administration that comprise a solution of the compound of the invention dissolved or suspended in an acceptable carrier suitable for parenteral administration, including aqueous and non-aqueous, isotonic sterile injection solutions.
  • compositions include solutions containing anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
  • the compound can be administered in a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol, isopropanol (for example in topical applications), or hexadecyl alcohol, glycols, such as propylene glycol or polyethylene glycol, dimethylsulfoxide, glycerol ketals, such as 2,2-dimethyl-1 ,3-dioxolane- 4-methanol, ethers, such as poly(ethyleneglycol) 400, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or carb
  • Oils useful in parenteral formulations include petroleum, animal, vegetable, and synthetic oils. Specific examples of oils useful in such formulations include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral oil. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.
  • Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts
  • suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylene polypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-3-aminopropionates, and 2-alkyl-imidazoline quaternary ammonium salts, and (e) mixtures thereof.
  • the parenteral formulations typically will contain from about 0.5% or less to about 25% or more by weight of a compound of the invention in solution. Preservatives and buffers can be used. In order to minimize or eliminate irritation at the site of injection, such compositions can contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations will typically range from about 5% to about 15% by weight. Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
  • HLB hydrophile-lipophile balance
  • parenteral formulations can be presented in unit-dose or multi-dose sealed containers, such as ampoules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use.
  • sterile liquid excipient for example, water
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets.
  • Topical formulations including those that are useful for transdermal drug release, are well known to those of skill in the art and are suitable in the context of the present invention for application to skin.
  • Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of a compound of the invention dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets, tablets, lozenges, and troches, each containing a pre-determined amount of the compound of the invention, as solids or granules; (c) powders; (d) suspensions in an appropriate liquid; and (e) suitable emulsions.
  • Liquid formulations can include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent.
  • diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent.
  • Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and cornstarch.
  • Tablet forms can include one or more of lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible excipients.
  • Lozenge forms can comprise the compound ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising a compound of the invention in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the compound of the invention, such excipients as are known in the art.
  • a flavor usually sucrose and acacia or tragacanth
  • pastilles comprising a compound of the invention in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the compound of the invention, such excipients as are known in the art.
  • An compound of the present invention can be made into aerosol formulations to be administered via inhalation.
  • a compound or epimer of the invention is preferably supplied in finely divided form along with a surfactant and propellant.
  • Typical percentages of the compounds of the invention can be about 0.01 % to about 20% by weight, preferably about 1 % to about 10% by weight.
  • the surfactant must, of course, be nontoxic, and preferably soluble in the propellant.
  • Such surfactants are the esters or partial esters of fatty acids containing from 6 to 22 carbon atoms, such as caproic, octanoic, lauric, palmitic, stearic, linoleic, linolenic, olesteric and oleic acids with an aliphatic polyhydric alcohol or its cyclic anhydride.
  • Mixed esters such as mixed or natural glycerides can be employed.
  • the surfactant can constitute from about 0.1 % to about 20% by weight of the composition, preferably from about 0.25% to about 5%.
  • the balance of the composition is ordinarily propellant.
  • a carrier can also be included as desired, e.g., lecithin, for intranasal delivery.
  • aerosol formulations can be placed into acceptable pressurized propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also can be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer. Such spray formulations can be used to spray mucosa.
  • acceptable pressurized propellants such as dichlorodifluoromethane, propane, nitrogen, and the like.
  • non-pressured preparations such as in a nebulizer or an atomizer.
  • Such spray formulations can be used to spray mucosa.
  • the compound of the invention can be made into suppositories by mixing with a variety of bases, such as emulsifying bases or water-soluble bases.
  • bases such as emulsifying bases or water-soluble bases.
  • Formulations suitable for vaginal administration can be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the compound ingredient, such carriers as are known in the art to be appropriate.
  • the concentration of the compound in the pharmaceutical formulations can vary, e.g., from less than about 1 % to about 10%, to as much as 20% to 50% or more by weight, and can be selected primarily by fluid volumes, and viscosities, in accordance with the particular mode of administration selected.
  • a typical pharmaceutical composition for intravenous infusion could be made up to contain 250 ml of sterile Ringer's solution, and 100 mg of at least one compound of the invention.
  • Actual methods for preparing parenterally administrable compounds of the invention will be known or apparent to those skilled in the art and are described in more detail in, for example, Remington's Pharmaceutical Science (17 th ed., Mack Publishing Company, Easton, PA, 1985). It will be appreciated by one of ordinary skill in the art that, in addition to the aforedescribed pharmaceutical compositions, the compound of the invention can be formulated as inclusion complexes, such as cyclodextrin inclusion complexes, or liposomes.
  • Liposomes can serve to target a compound of the invention to a particular tissue, such as lymphoid tissue or cancerous hepatic cells. Liposomes can also be used to increase the half-life of a compound of the invention. Many methods are available for preparing liposomes, as described in, for example, Szoka et al., Ann. Rev. Biophys. Bioeng., 9, 467 (1980) and U.S. Patents 4,235,871 , 4,501 ,728, 4,837,028, and 5,019,369.
  • the present invention also provides a pharmaceutical composition, containing the compounds of the general formula (I) as defined above, its tautomeric forms, its stereoisomers, its analogs, its prodrugs, its isotopes, its metabolites, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates and its co- crystals in combination with the usual pharmaceutically employed carriers, diluents and the like, and for use in any of the methods described herein.
  • the compounds of the invention can be administered in a dose sufficient to treat the disease, condition or disorder.
  • doses are known in the art (see, for example, the Physicians' Desk Reference (2004)).
  • the compounds can be administered using techniques such as those described in, for example, Wasserman et al., Cancer, 36, pp. 1258-1268 (1975) and Physicians' Desk Reference, 58th ed., Thomson PDR (2004).
  • Suitable doses and dosage regimens can be determined by conventional range-finding techniques known to those of ordinary skill in the art. Generally, treatment is initiated with smaller dosages that are less than the optimum dose of the compound of the present invention. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached.
  • the present method can involve the administration of about 0.1 ⁇ g to about 50 mg of at least one compound of the invention per kg body weight of the individual. For a 70 kg patient, dosages of from about 10 ⁇ g to about 200 mg of the compound of the invention would be more commonly used, depending on a patient's physiological response.
  • the dose of the pharmaceutically active agent(s) described herein for methods of treating or preventing a disease or condition as described above can be about 0.001 to about 1 mg/kg body weight of the subject per day, for example, about 0.001 mg, 0.002 mg, 0.005 mg, 0.010 mg, 0.015 mg, 0.020 mg, 0.025 mg, 0.050 mg, 0.075 mg, 0.1 mg, 0.15 mg, 0.2 mg, 0.25 mg, 0.5 mg, 0.75 mg, or 1 mg/kg body weight per day.
  • the dose of the pharmaceutically active agent(s) described herein for the described methods can be about 1 to about 1000 mg/kg body weight of the subject being treated per day, for example, about 1 mg, 2 mg, 5 mg, 10 mg, 15 mg, 0.020 mg, 25 mg, 50 mg, 75 mg, 100 mg, 150 mg, 200 mg, 250 mg, 500 mg, 750 mg, or 1000 mg/kg body weight per day.
  • the present invention provides methods of treating, preventing, ameliorating, and/or inhibiting a hepatitis C virus infection comprising administering a compound of formula (I) or a salt thereof.
  • the compounds of the present invention can inhibit multiple genotypes of HCV.
  • compound of the present invention are active against the 1 a, 1 b, 2a, 2b, 3a, 4a and 5a genotypes.
  • the terms “treat,” “prevent,” “ameliorate,” and “inhibit,” as well as words stemming therefrom, as used herein, do not necessarily imply 100% or complete treatment, prevention, amelioration, or inhibition. Rather, there are varying degrees of treatment, prevention, amelioration, and inhibition of which one of ordinary skill in the art recognizes as having a potential benefit or therapeutic effect.
  • the inventive methods can provide any amount of any level of treatment, prevention, amelioration, or inhibition of the disorder in a mammal.
  • a disorder, including symptoms or conditions thereof may be reduced by, for example, 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, or 10%.
  • the treatment, prevention, amelioration, or inhibition provided by the inventive method can include treatment, prevention, amelioration, or inhibition of one or more conditions or symptoms of the disorder, e.g., cancer.
  • "treatment,” “prevention,” “amelioration,” or “inhibition” can encompass delaying the onset of the disorder, or a symptom or condition thereof.
  • the term subject includes an "animal” which in turn includes a mammal such as, without limitation, the order Rodentia, such as mice, and the order Lagomorpha, such as rabbits.
  • the mammals are from the order Carnivora, including Felines (cats) and Canines (dogs). It is more preferred that the mammals are from the order Artiodactyla, including Bovines (cows) and Swine (pigs) or of the order Perssodactyla, including Equines (horses). It is most preferred that the mammals are of the order Primates, Ceboids, or Simoids (monkeys) or of the order Anthropoids (humans and apes). An especially preferred mammal is the human.
  • viral infection refers to the introduction of a virus into cells or tissues, e.g., hepatitis C virus (HCV). In general, the introduction of a virus is also associated with replication. Viral infection may be determined by measuring virus antibody titer in samples of a biological fluid, such as blood, using, e.g., enzyme immunoassay. Other suitable diagnostic methods include molecular based techniques, such as RT-PCR, direct hybrid capture assay, nucleic acid sequence based amplification, and the like. A virus may infect an organ, e.g., liver, and cause disease, e.g., hepatitis, cirrhosis, chronic liver disease and hepatocellular carcinoma.
  • HCV hepatitis C virus
  • immune modulator refers to any substance meant to alter the working of the humoral or cellular immune system of a subject.
  • immune modulators include inhibitors of mast cell-mediated inflammation, interferons, interleukins, prostaglandins, steroids, cortico-steroids, colony-stimulating factors, chemotactic factors, etc.
  • ACN Acetonitrile. Boc: tert-butyloxycarbonyl. Cbz: Carbobenzyloxy.
  • CDCI 3 Deuterochloroform.
  • DCM dichloromethane.
  • DDQ 2,3-Dichloro-5,6-dicyano-p- benzoquinone.
  • DIPEA N, N-diisopropylethylamine.
  • DME 1 , 2-dimethoxyethane.
  • DMF N, N-dimethylformamide.
  • DMSO-c 6 Dimethyl sulfoxide-d 6 .
  • EDCI A/-(3-Dimethylaminopropyl)- /V-ethylcarbodiimide.
  • EtOAc Ethyl acetate.
  • HATU 0-(7-Azabenzotriazol-1 -yl)-A/,A/,A/',/V- tetramethyluronium hexafluorophosphate.
  • HCI hydrochloric acid.
  • HOBt 1 - Hydroxybenzotriazole hydrate.
  • HPLC High Performance Liquid Chromatography.
  • KOAc potassium acetate.
  • MeOD Methanol-d 4 .
  • Na 2 S04 sodium sulphate.
  • NaHC0 3 sodium bicarbonate.
  • NCS N-chlorosuccinimide.
  • PCy 3 Tricyclohexylphosphine.
  • Pd(PPh 3 ) 4 Tetrakis(triphenylphosphine)palladium(0).
  • Pd-C Palladium on charcoal.
  • PdCI 2 (dppf) [1 ,1 '- Bis(diphenylphosphino)ferrocene]dichloropalladium(ll).
  • PdCl 2 (dppf)-CH 2 CI 2 [1 ,1 '- Bis(diphenylphosphino)ferrocene]dichloropalladium(ll), complex with dichloromethane.
  • pet-ether petroleum ether.
  • TFA trifluoroacetic acid.
  • THF Tetrahydrofuran
  • Example 1 Preparation of methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (mixture of trans isomers) (Compound 1):
  • Step 1 2-(4-bromophenyl)-2-oxoethyl-2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylate (trans isomers) (1 a):
  • Step 2 tert-butyl (2-(5-(4-bromophenyl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (trans isomers)(1 b):
  • Step 3 tert-butyl (2-(5-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)carbamate (trans isomers) (1 c):
  • Step 4 (2S)-tert-butyl 2-(7-(4-(2-(2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H-imidazol- 5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (trans isomers) (1 d):
  • Step 5 methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (mixture of trans isomers) (Compound 1 ):
  • Example 2 methyl ((2S)-1-((2-(5-(4-(2-((S)-1-((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate (mixture of cis isomers) (Compound 2):
  • Step 1 2-(4-bromophenyl)-2-oxoethyl-2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylate (cis isomers) (2a):
  • Step 2 tert-butyl (2-(5-(4-bromophenyl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (cis isomers) (2b):
  • Step 3 tert-butyl (2-(5-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)carbamate (cis isomers) (2c):
  • Step 4 (2S)-tert-butyl 2-(8-(4-(2-(2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H-imidazol- 5-yl)phenyl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-2-yl)pyrrolidine-1 - carboxylate (2d):
  • Step 5 methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (mixture of cis isomers): (Compound 2)
  • Example 3 Preparation of methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (mixture of cis isomers) (Compound 3):
  • Step 1 (2S)-tert-butyl 2-(7-(4-(2-(2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H-imidazol- 5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (cis isomers) (3a):
  • Step 2 methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (mixture of cis isomers): (Compound 3)
  • Step 1 (1 S,2R)-2-(4-bromophenyl)-2-oxoethyl-2-((tertbutoxycarbonyl)amino) cyclopentanecarboxylate (4a):
  • Step 2 tert-butyl ((1 R,2S)-2-(4-(4-bromophenyl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (4b):
  • Step 3 tert-butyl ((1 R,2S)-2-(5-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)carbamate (4c):
  • Step 4 (S)-tert-butyl 2-(7-(4-(2-((1 S,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (4d):
  • Step 5 methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 4).
  • Example 5 Preparation of methyl ((S)-1-(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 , 4,5,6- tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Example 5):
  • Step 1 (S)-tert-butyl 2-(8-(4-(2-((1 S,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate (5a):
  • Step 2 methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 5):
  • Step 1 (S)-tert-butyl 2-(8-(4-(2-((1 S,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-2-yl)pyrrol ⁇ carboxylate (6a):
  • Step 2 methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 6):
  • Example 7 methyl ((S)-1-(((1 S,2R)-2-(5-(4-(2-((S)-1-((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 7):
  • Step 1 (1 R,2S)-2-(4-bromophenyl)-2-oxoethyl-2-((tertbutoxycarbonyl) amino)cyclopentanecarboxylate (7a):
  • Step 2 tert-butyl- ((1 S,2R)-2-(4-(4-bromophenyl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (7b):
  • Step 3 tert-butyl ((1 S,2R)-2-(5-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)carbamate (7c):
  • Step 4 (S)-tert-butyl 2-(8-(4-(2-((1 R,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-2-yl)pyrrolidine-1 - carboxylate (7d):
  • Step 5 methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 7):
  • Example 8 Preparation of methyl ((S)-1-(((1S,2R)-2-(5-(4-(2-((S)-1 -((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 , 4,5,6- tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 8):
  • Step 1 (S)-tert-butyl 2-(8-(4-(2-((1 R,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate (8a):
  • Step 2 methyl ((S)-1-(((1S,2R)-2-(5-(4-(2-((S)-1-((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8- yl)phenyl)-1H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 8):
  • Example 9 Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1, 4,5,6- tetrahydrobenzo[3,4]cyclohepta[1,2-d]imidazol-8-yl)phenyl)-1H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 9):
  • Step 1 (1S,2S)-2-(4-bromophenyl)-2-oxoethyl-2-((tertbutoxycarbonyl)amino) cyclopentanecarboxylate (9a):
  • Step 3 tert-butyl ((1 S,2S)-2-(5-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)carbamate (9c):
  • Step 4 (S)-tert-butyl 2-(8-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate (9d):
  • Step 5 methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 9):
  • Step 1 (S)-tert-butyl 2-(7-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (10a)
  • Step 2 methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 10):
  • Example 11 Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- benzo[2,3]oxepino[4,5-d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)- 3-methyl-1-oxobutan-2-yl)carbamate (Compound 11 ):
  • Step 1 (S)-tert-butyl 2-(8-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-ben ⁇
  • Step 2 methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 1 1 ):
  • Example 12 Preparation of methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 , 4,5,6- tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 12):
  • Step 1 (1 R, 2R)-2-(4-bromophenyl)-2-oxoethyl-2-((tert-butoxycarbonyl)amino) cyclopentane carboxylate (12a):
  • Step 2 tert-butyl ((1 R,2R)-2-(4-(4-bromophenyl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (12b):
  • Step 3 tert-butyl ((1 R,2R)-2-(5-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)carbamate (12c):
  • Step 4 (S)-tert-butyl 2-(8-(4-(2-((1 R,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate (12d):
  • Step 5 methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 12):
  • Example 13 Preparation of methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- benzo[2,3]oxepino[4,5-d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)- 3-methyl-1-oxobutan-2-yl)carbamate (Compound 13):
  • Step 1 (S)-tert-butyl 2-(8-(4-(2-((1 R,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-2-yl)pyrrolidine-1 - carboxylate (13a):
  • Step 2 methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 13):
  • Example 14 Preparation of methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (Compound 14):
  • Step 1 (S)-tert-butyl 2-(7-(4-(2-((1 R,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (14a):
  • Step 2 methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 14):
  • Example 15 Preparation of methyl ((S)-1-(((1S,2R)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (Compound 15):
  • Step 1 (S)-tert-butyl 2-(7-(4-(2-((1 R,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (15a):
  • Step 2 methyl ((S)-1-(((1S,2R)-2-(5-(4-(2-((S)-1-((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 15):
  • Example 16 Preparation of methyl ((S)-1-(((1 R,2R)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1H-naphtho[1,2- d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2- yl)carbamate (Compound 16):
  • Step 2 (S)-tert-butyl 2-(7-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-3H-naphtho[1 ,2- d]imidazol-2-yl)pyrrolidine-1 -carboxylate (16b):
  • Step 3 (S)-tert-butyl 2-(7-(4-(2-((1 R,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (16c):
  • Step 4 methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 16):
  • Example 17 Preparation of methyl ((S)-1-(((1S,2R)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2- d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate (Compound 17):
  • Step 1 (S)-tert-butyl 2-(7-(4-(2-((1 R,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (17a):
  • Step 2 methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 17):
  • Example 18 Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2- d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate (Compound 18):
  • Step 1 (S)-tert-butyl 2-(7-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (18a):
  • Step 2 methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 18):
  • Example 19 Preparation of methyl ((S)-1-(((1S,2R)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2- d]imidazol-7-yl)phenyl)-4-chloro-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (Compound 19):
  • Step 1 tert-butyl ((1 S,2R)-2-(5-(4-bromophenyl)-4-chloro-1 H-imidazol-2- yl)cyclopentyl)carbamate (19a):
  • Step 2 (S)-tert-butyl 2-(7-(4-(2-((1 R,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-4- chloro-1 H-imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate
  • Step 3 methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-4-chloro-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 19).
  • Example 20 Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2- d]imidazol-7-yl)phenyl)-4-chloro-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (Compound 20):
  • Step 1 tert-butyl ((1 S,2S)-2-(5-(4-bromophenyl)-4-chloro-1 H-imidazol-2- yl)cyclopentyl)carbamate (20a):
  • Step 2 (S)-tert-butyl 2-(7-(4-(2-((1S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-4- chloro-1 H-imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (20b):
  • Step 3 methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-4-chloro-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 20):
  • reaction mixture was poured into water (100 mL) and the compound was extracted with ethyl acetate (100 mL ⁇ 2), dried over sodium sulphate, concentrated under reduced pressure and purified by flash column chromatography (45-55% Ethylacetate/Hexane) to get the title compound as side product, orange colored solid (0.7 g, 23 % yield) along with (S)-tert-butyl 2-(8-bromo-1 , 4,5,6- tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate as the major compound.
  • Step 2 (S)-tert-butyl 2-(8-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-5,6-dihydro-4H- benzo[3,4]cyclohepta[1 ,2-d]oxazol-2-yl)pyrrolidine-1 -carboxylate (21 b):
  • Step 3 (S)-tert-butyl 2-(8-(4-(2-((1S,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1H- imidazol-5-yl)phenyl)-5,6-dihydro-4H-benzo[3,4]cyclohepta[1,2-d]oxazol-2-yl)pyrrolidine-1- carboxylate (21c):
  • Step 4 methyl ((S)-1-(((1R,2S)-2-(5-(4-(2-((S)-1-((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-5,6-dihydro-4H-benzo[3,4]cyclohepta[1,2-d]oxazol-8- yl)phenyl)-1H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 21):
  • Example 22 Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5- dihydrobenzo[2,3]oxepino[4,5-d]oxazol-8-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 22):
  • Step 1 (S)-tert-butyl 2-(8-bromo-4,5-dihydrobenzo[2,3]oxepino[4,5-d]oxazol-2- yl)pyrrolidine-1 -carboxylate (22a):
  • Step 2 (S)-tert-butyl 2-(8-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-4,5- dihydrobenzo[2,3]oxepino[4,5-d]oxazol-2-yl)pyrrolidine-1 -carboxylate (22b):
  • Step 3 (S)-tert-butyl 2-(8-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydrobenzo[2,3]oxepino[4,5-d]oxazol-2-yl)pyrrolidine-1 - carboxylate (22c):
  • Step 4 methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydrobenzo[2,3]oxepino[4,5-d]oxazol-8-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 22):
  • Example 23 Preparation of methyl ((S)-1-(((1 R,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5- dihydrobenzo[2,3]oxepino[4,5-d]oxazol-8-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 23):
  • Step 1 (S)-tert-butyl 2-(8-(4-(2-((1 S,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydrobenzo[2,3]oxepino[4,5-d]oxazol-2-yl)pyrrolidine-1 - carboxylate (23a):
  • Step 2 methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydrobenzo[2,3]oxepino[4,5-d]oxazol-8-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 23):
  • Example 24 Preparation of methyl ((S)-1-(((1S,2S)-2-(7-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)- 4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (Compound 24):
  • Step 1 (1 S,2S)-6-bromo-1 -oxo-1 ,2,3,4-tetrahydronaphthalen-2-yl 2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylate (24a):
  • Step 2 tert-butyl ((1 S,2S)-2-(7-bromo-4,5-dihydro-3H-naphtho[1 ,2-d]imidazol-2- yl)cyclopentyl)carbamate (24b):
  • Step 3 (S)-tert-butyl 2-(5-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-4,5- dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)pyrrolidine-1 -carboxylate (24c).
  • Step 4 methyl ((S)-1 -(((1 S,2S)-2-(7-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2- d]imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 24):
  • Step 2 tert-butyl ((1 S,2R)-2-(7-bromo-4,5-dihydro-3H-naphtho[1 ,2-d]imidazol-2- yl)cyclopentyl)carbamate (25b):
  • Step 3 (S)-tert-butyl 2-(5-(4-(2-((1 R,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-4,5- dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)pyrrolidine-1 -carboxylate (25c):
  • Title compound was synthesized by following the procedure as described in Step 4 of Example 1 , using the intermediates (25b) from the previous step and (S)-tert-butyl 2-(5-(4- (4,4,5, 5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)-1 H-imidazol-2-yl)pyrrolidine-1 - carboxylate [synthesized using procedures described in ref. WO2010/99527A1 ] as starting materials.
  • Step 4 methyl ((S)-1 -(((1 S,2R)-2-(7-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2- d]imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 25):
  • Step 1 (S)-tert-butyl 2-(5-(4-(2-((1 R,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)pyrrolidine-1 -carboxylate (26a):
  • Step 2 methyl ((S)-1 -(((1 S,2R)-2-(7-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol-2- yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 26):
  • Step 1 (S)-tert-butyl 2-(5-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)pyrrolidine-1 -carboxylate (27a):
  • Step 2 methyl ((S)-1 -(((1 S,2S)-2-(7-(4-(2-((S)-1 -((S)-2-amino-3-methylbutanoyl)pyrrolidin- 2-yl)-1 H-imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol-2-yl)cyclopentyl)amino)-3- methyl-1 -oxobutan-2-yl)carbamate (Compound 27):
  • Example 28 Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)(ethyl)amino)-3- methyl-1 -oxobutan-2-yl)carbamate (Compound 28): Step 1 : (1 S,2S)-ethyl-2-(ethylamino)cyclopentanecarboxylate (28a):
  • Step 2 (1 S,2S)-ethyl 2-((tert-butoxycarbonyl)(ethyl)amino)cyclopentanecarboxylate (28b):
  • Step 3 (1 S,2S)-2-((tert-butoxycarbonyl)(ethyl)amino)cyclopentanecarboxylic acid (28c):
  • Step 4 (1 S, 2S)-2-(4-bromophenyl)-2-oxoethyl 2-((tert-butoxycarbonyl)(ethyl) amino) cyclopentane carboxylate (28d):
  • Step 5 tert-butyl ((1 S,2S)-2-(5-(4-bromophenyl)-1 H-imidazol-2- yl)cyclopentyl)(ethyl)carbamate (28e):
  • Step 6 (S)-tert-butyl 2-(7-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)(ethyl)amino) cyclopentyl)-1 H-imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate (28f):
  • Step 7 methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)(ethyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 28):
  • Step 2 (1 S, 2S)-ethyl 2-((tert-butoxycarbonyl)(methyl)amino) cyclopentanecarboxylate (29b):
  • Step 4 (1 S,2S)-2-(4-bromophenyl)-2-oxoethyl 2-((tert-butoxycarbonyl)(methyl) amino)cyclopentanecarboxylate (29d) :
  • Step 5 tert-butyl ((1 S,2S)-2-(5-(4-bromophenyl)-1 H-imidazol-2- yl)cyclopentyl)(methyl)carbamate (29e):
  • Step 6 (S)-tert-butyl 2-(7-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)(methyl)amino) cyclopentyl)-1 H-imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate (29f):
  • Step 7 methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)(methyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 29):
  • Example 30 Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(7-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)-9H- fluoren-2-yl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate (Compound 30): Step 1 : 1 -(7-bromo-9H-fluoren-2-yl)ethanone (30a):
  • Step 4 (1 S,2S)-2-(7-bromo-9H-fluoren-2-yl)-2-oxoethyl-2-((tert-butoxycarbonyl) amino)cyclopentanecarboxylate (30d) :
  • Step 5 tert-butyl ((1 S,2S)-2-(5-(7-bromo-9H-fluoren-2-yl)-1 H-imidazol-2- yl)cyclopentyl)carbamate (30e):
  • Step 6 tert-butyl ((1 S,2S)-2-(5-(7-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-9H-fluoren- 2-yl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (30f):
  • Step 7 (S)-tert-butyl 2-(5-(7-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)-9H-fluoren-2-yl)-1 -((2-(trimethylsilyl)ethoxy)methyl)-1 H-imidazol-2- yl)pyrrolidine-1 -carboxylate (30g):
  • Step 8 methyl ((S)-1 -(((1 S,2S)-2-(5-(7-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)-9H-fluoren-2-yl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 30):
  • Example 31 Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(7-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)-9,9- dimethyl-9H-fluoren-2-yl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1-oxobutan- 2-yl)carbamate (Compound 31 ):
  • Step 1 1 -(7-bromo-9,9-dimethyl-9H-fluoren-2-yl)ethanone (31 a):
  • Step 2 2,2-dibromo-1 -(7-bromo-9,9-dimethyl-9H-fluoren-2-yl)ethanone
  • Step 3 2-bromo-1 -(7-bromo-9,9-dimethyl-9H-fluoren-2-yl)ethanone (31 c): Title compound was synthesized by following the procedure as described in Step 3 of Example 30, using intermediate (31b) from the previous step.
  • Step 4 (1S,2S)-2-(7-bromo-9,9-dimethyl-9H-fluoren-2-yl)-2-oxoethyl 2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylate (31d):
  • Step 5 tert-butyl ((1S,2S)-2-(5-(7-bromo-9,9-dimethyl-9H-fluoren-2-yl)-1 H-imidazol-2- yl)cyclopentyl)carbamate (31 e):
  • Step 6 tert-butyl ((1S,2S)-2-(5-(9,9-dimethyl-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)-9H-f luoren-2-yl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (31 f)
  • Step 7 (S)-tert-butyl 2-(5-(7-(2-((1S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)-9,9-dimethyl-9H-fluoren-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H- imidazol-2-yl)pyrrolidine-1-carboxylate (31 g):
  • Step 8 methyl ((S)-1-(((1S,2S)-2-(5-(7-(2-((S)-1-((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)-9,9-dimethyl-9H-fluoren-2-yl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 31 ):
  • Example 32 Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(7-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5- yl)phenanthren-2-yl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2- yl)carbamate (Compound 32):
  • Step 1 1 -(7-bromophenanthren-2-yl)ethanone (32a):
  • Step 4 (1S,2S)-2-(7-bromophenanthren-2-yl)-2-oxoethyl 2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylate (32d):
  • Step 5 tert-butyl ((1S,2S)-2-(5-(7-bromophenanthren-2-yl)-1 H-imidazol-2- yl)cyclopentyl)carbamate (32e): Title compound was synthesized by following the procedure as described in Step 5 of Example 30, using intermediate (32d) from the previous step.
  • Step 6 tert-butyl ((1 S,2S)-2-(5-(7-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenanthren-2-yl)-1 H-i )
  • Step 7 (S)-tert-butyl 2-(5-(7-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenanthren-2-yl)-1 -((2-(trimethylsilyl)ethoxy)methyl)-1 H-imidazol-2- yl)pyrrolidine-1 -carboxyla
  • Step 8 methyl ((S)-1 -(((1 S,2S)-2-(5-(7-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenanthren-2-yl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 32):
  • the Huh7.5/ Con1/SG-Neo(l)hRluc2aUb cell line persistently expressing a bicistronic genotype 1 b replicon in Huh 7.5 cells was obtained from Apath LLC. This cell line was used to test inhibition of genotype 1 b replicon levels by test compound using Renilla luciferase enzyme activity readout as a measure of viral replication efficiency. Briefly, 7000 cells were seeded in 96 well black clear bottom plates and allowed to adhere overnight. The next day each compound was added in triplicate to the cells at the desired concentration with a final DMSO concentration of 0.5%.
  • Table 1 shows EC50 values, for inhibition of genotype 1 b replicon, of the compounds of the invention.
  • Group A compounds exhibited EC50 value between 1 pM to 999 pM
  • Group B exhibited EC50 value between 1 nM to 999 nM
  • Goup C exhibited EC50 value of more than 1 ⁇ .

Abstract

Compounds of the general formula (I) their tautomeric forms, their stereoisomers, their analogs, their prodrugs, their isotopes, their N-oxides, their metabolites, their pharmaceutically acceptable salts, polymorphs, solvates, optical isomers, clathrates, co-crystals, combinations with suitable medicament, pharmaceutical compositions containing them, methods of making of the above compounds, and their use as antiviral candidate, more specifically as anti-HCV are disclosed.

Description

IMIDAZOLE DERIVATIVES AS ANTIVIRAL AGENTS
FIELD OF THE INVENTION:
The present invention is related to novel compounds of the general formula I,
Figure imgf000002_0001
their tautomeric forms, their stereoisomers, their analogs, their prodrugs, their isotopes, their N-oxides, their metabolites, their pharmaceutically acceptable salts, polymorphs, solvates, optical isomers, clathrates, co-crystals, combinations with suitable medicament, pharmaceutical compositions containing them, methods of making of the above compounds , and their use as antiviral candidate, more specifically as anti-HCV. BACKGROUND OF THE INVENTION
Persistent hepatitis C virus (HCV) infection is a major health problem globally affecting -3% of the world population and is an important contributor to chronic liver disease culminating with liver cirrhosis, hepatocellular carcinoma and liver failure [Szabo E,Lotz G, et al., Pathol. Oncol. Res. 2003, 9, 215-221 ; Hoofnagle JH., Hepatology 1997, 26 15S-20]. An estimated 170 million chronic carriers worldwide are at risk of developing liver disease. In the United States alone ~3 million are chronically infected with HCV and the number of HCV related deaths is increasing significantly over the years [Barnes E., WHO factsheet 2010. Available at: http://www.who.int/vaccine_research/diseases/viral_cancers/en/index2.html]. Clinically, chronic infection is often asymptomatic with latent periods lasting for decades before manifestation by which time extensive liver damage has occurred. HCV is spread primarily by unscreened blood transfusions and use of contaminated needles and syringes; the highest risk groups are intravenous drug users and people who received blood transfusions (mainly haemophiliacs) before 1990 when screening for HCV was introduced. Factors that have been reported to influence the rate of HCV disease progression include age (increasing age is associated with more rapid progression), gender (males have more rapid disease progression than females), alcohol consumption (associated with an increased rate of disease progression), HIV co-infection (associated with a markedly increased rate of disease progression), and fatty liver.
Despite significant efforts, no vaccine exists for HCV and until a year ago the standard therapy for HCV was a combination of pegylated interferon (PEG-IFN) a and weight based ribavarin (RBV), which was inadequate for majority of the patients and therapy associated side effects such as pancytopenia, flu-like symptoms or depression were commonly observed leading to early treatment discontinuation [Fried MW, et al., N Engl J Med. 2002, 347, 975-982]. The approval of two direct acting agents (DAA) i.e. 1 st generation protease inhibitors, Incivek and Victrelis in May 201 1 ushered in the era of specifically targeted HCV therapy[Jesudian AB, Gambarin-Gelwan M and Jacobson IM., Gastroenterology Hepatol. 2012, 8, 91 -101 .
The combination of above mentioned DAAs, Peg-IFN and RBV (triple therapy) substantially increases the rate of sustained virologic response in treatment nal've and experienced patients. However, a number of issue restrict the usage of these drugs - i) complex treatment algorithms issued by the regulatory bodies; ii) they are restricted to genotype 1 ; iii) low barrier to resistance mutations and iv) increased cost of therapy leading to limited access to care. Hence, there exists a need for alternative therapeutic strategies that ensure broader genotype coverage, better efficacy, better tolerance and limited selection of resistant HCV variants. The sequence diversity of HCV is complex with the virus organized into 6 distinct genotypes and over 100 subtypes. Additionally, HCV exists as many closely related viral sequences, termed as quasi-species, in the infected individual, making specific pharmaceutical targeting of HCV proteins challenging due to the rapid evolution of escape mutants. It is increasingly evident that a broad collection of specific, pan genotypic anti- viral drugs targeting multiple essential viral functions, in addition to the current viral therapies will be required for effective global control of HCV.
SUMMARY OF THE INVENTION
According to one aspect of the present invention there is provided a novel compound of the general formula (I), its tautomeric forms, its stereoisomers, its analogs, its prodrugs, its isotopes, its N-oxides, its metabolites, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates, or its co-crystals,
Figure imgf000004_0001
(I) wherein, R1a, R1 to R12, m, n, Y, ring D and ring A are as defined hereinbelow. In another aspect, the present invention provides a pharmaceutical composition, containing the compound of the general formula (I) as defined herein, its tautomeric forms, its stereoisomers, its analogs, its prodrugs, its isotopes, its N-oxides, its metabolites, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates, or its co-crystals in combination with the usual pharmaceutically employed carriers, diluents and the like are useful for the treatment of HCV infection.
DESCRIPTION OF THE INVENTION
HCV is a member of the Flaviviridae family of enveloped, positive stranded RNA viruses belonging to the genus Hepacivirus. The genome is a single ~9.6kb strand of RNA and consists of one open reading frame that encodes for a polyprotein of -3000 amino acids flanked by untranslated regions at both 5' and 3' ends. This precursor polyprotein is then processed by viral and cellular proteases to yield 10 separate mature viral proteins critical for replication and assembly of progeny viral particles. The organization of the structural and non-structural proteins in the HCV polyprotein is as follows: C-E1 -E2-P7-NS2-NS3- NS4a-NS4b-NS5a-NS5b. The three structural proteins C, E1 and E2 are involved in packaging of the virus and the infectivity cycle. The function of the p7 protein is unknown. Of the non-structural proteins, NS2 is a zinc dependent metalloproteinase that functions in conjunction with a part of NS3 protein. NS3 protein has two catalytic activities associated with it: a serine protease at the N-terminal end which requires NS4A as a cofactor, and an ATPase dependent helicase activity at the C-terminal end. NS5A is a membrane anchored phosphoprotein that is present in basally phosphorylated (56kDa) and hyperphosphorylated (58kDa) forms. Its precise role has not been determined but it has been shown to play a role in RNA binding, multiple host protein interactions, and interferon resistance. Additionally recent evidence suggests that NS5A plays an important role in replication and infectivity of HCV. The NS5B protein encodes an RNA dependent RNA polymerase activity, key to the generation of progeny viruses. While the pathology of HCV infection mainly affects the liver, the virus is found in other cell types in the body including peripheral blood lymphocytes [Thomson BJ et al., Clin Microbial Infect. 2005, 1J., 86-94; Moriishi K et al., Antivir. Chem. Chemother. 2003, 14, 285-297]. Characterization of the replicase machinery required for HCV RNA synthesis has defined the protease/helicase NS3 protein, the NS4A cofactor, the NS4B integral membrane protein, the NS5A protein and the RNA dependent RNA polymerase NS5B as being its essential components.
Hence, one of the aspects of the present invention is provision of novel compounds of the general formula I,
Figure imgf000005_0001
(I) their tautomeric forms, their isomers, their isotopes, their metabolites, their prodrugs, their pharmaceutically acceptable salts, pharmaceutical compositions containing them, methods of making of the above compounds, and their use as antiviral compounds; wherein,
Ring 'A' is a saturated carbocycle, which may be a monocyclic system or may be a fused carbocycle or may be a bridged carbocycle, the said ring A may contain 5 to 10 carbons; Ring 'D' is selected from 5 to 10 membered carbocycle and 5 to 10 membered
heterocycle, the ring 'D' may be monocyclic, fused bicyclic, bridged bicyclic or spiro bicyclic;
Y is selected from -CH(R2a)- and -N(R2)-;
R1 is selected from the group consisting of R13aC(=0)N(R14)-, R13aOC(=0)N(R14)-, R13(R14)N-, R13(R14)NC(=0)N(R15)-, R13aS02N(R14)-,
R13aOC(=0)N(R14)CRa(Rb)C(=0)N(R15)-, R13aOC(=0)N(R14)CRa(Rb)C(Rc)(Rd)C(=0)N(R15)-,
R13(R14a)NC(=0)N(R14)CRa(Rb)C(=0)N(R15)-, and R13(R14a)NC(=0)N(R14)CRa(Rb)C(Rc)(Rd)C(=0)N(R15)-;
R1a is selected independently at each occurrence from the group consisting of halogen, substituted- or unsubstituted- Ci-6 alkyl, R13aC(=0)-, R13bO-, R13aOC(=0)-, R13aC(=0)0-, and R13(R14)NC(=0)-,
R2 is selected independently at each occurrence from the group consisting of substituted- or unsubstituted- Ci-6 alkyl, R13aC(=0)-, R13aS02-, R13aOC(=0)-, R13(R14)NC(=0)-,
R13aOC(=0)N(R14)C(Ra)(Rb)C(=0)-, R13aOC(=0)N(R14)C(Ra)(Rb)C(Rc)(Rd)C(=0)-,
R13(R14)NC(=0)N(R15)C(Ra)(Rb)C(=0)-, R13(R14)NC(=0)N(R15)C(Ra)(Rb)C(Rc)(Rd)C(=0)-, R13S02N(R14)C(Ra)(Rb)C(=0)-, R13S02N(R14)C(Ra)(Rb)C(Rc)(Rd)C(=0)-, and
R13aOC(=0)N(R14)C(Ra)(Rb)S02-;
R2a is selected independently at each occurrence from the group consisting of R13aC(=0)N(R14)-, R13aOC(=0)N(R14)-, R13(R14)N-, R13(R14)NC(=0)N(R15)-, R13aS02N(R14)-, R13aOC(=0)N(R14)C(Ra)(Rb)C(=0)N(R15)-, R13aOC(=0)N(R14)C(Ra)(Rb)C(Rc)(Rd)C(=0)N(R15)-,
R13(R14a)NC(=0)N(R14)C(Ra)(Rb)C(=0)N(R15)-, and R13(R14a)NC(=0)N(R14)CRa(Rb)C(Rc)(Rd)C(=0)N(R15)-;
R3 is independently selected from O and N(R16); R4 is selected independently at each occurrence from CRe(Rf), O and N(R14); such that, when n = 2 and R4 is selected as CRe(Rf) for both the occurrences, two Res together can form a bond to form a alkenylene linkage or two Res and two Rfs together can form bonds to form alkynylene linkage;
R5, R6, R7, R8, R9 and R10 are independently selected as hydrogen, or R5 and R6 together, R7 and R10 together, or R8 and R9 together independently along with the carbon atoms to which they are attached forming 5 to 8 membered substituted- or unsubstituted- carbocycle, 5 to 8 membered substituted- or unsubstituted- heterocycle, 6 membered substituted- or unsubstituted- aryl, or 5 to 6 membered substituted- or unsubstituted- heteroaryl; with a proviso that the compound of formula I must have at least one cyclic system formed out of either R5 and R6, R8 and R9, or R7 and R10; also provided that R7 and R10 take part in formation of cyclic system only when n is 0;
R11 and R12 are independently selected from a group consisting of hydrogen, halogen, substituted- or unsubstituted- Ci-6 alkyl, R13bO-, and (R13)(R14)N-; wherein, R13, R14, R14a and R15 are independently selected from hydrogen, substituted- or unsubstituted- Ci-6 alkyl, substituted- or unsubstituted- aryl, substituted- or unsubstituted- heteroaryl, substituted- or unsubstituted- cycloalkyl, and substituted- or unsubstituted- heterocyclyl; R13a is selected from substituted- or unsubstituted- Ci-6 alkyl, perhaloalkyl, substituted- or unsubstituted- aryl, substituted- or unsubstituted- heteroaryl, substituted- or unsubstituted- cycloalkyl, and substituted- or unsubstituted- heterocyclyl;
R13b is selected from hydrogen, substituted- or unsubstituted- Ci-6 alkyl, perhaloalkyl, substituted- or unsubstituted- aryl, substituted- or unsubstituted- heteroaryl, substituted- or unsubstituted- cycloalkyl, and substituted- or unsubstituted- heterocyclyl;
R16 is selected from hydrogen or substituted- or unsubstituted alkyl group;
Ra, Rb, Rc and Rd, are independently selected from hydrogen, halogen, substituted- or unsubstituted- Ci-6 alkyl, substituted- or unsubstituted- aryl, substituted- or unsubstituted- heteroaryl, substituted- or unsubstituted- cycloalkyl, and substituted- or unsubstituted- heterocyclyl, or Ra, Rb, Rc and Rd together with the carbon atom(s) to which they are attached forming substituted- or unsubstituted- carbocycle, substituted- or unsubstituted- heterocycle; m is an integer ranging between 0 to 2, selected independently at each occurrence; n is an integer ranging between 0 and 2; 'alkyl' may be substituted with 1 to 4 substituents selected from the group consisting of halogen, oxo, Ci-6 alkyl, haloalkyl, aryl, heteroaryl, cycloalkyl, heterocyclyl, R17aC(=0)-, R17aS02-, R17bO-, R17aOC(=0)-, R17aC(=0)0-, (R17)(R18)NC(=0)-, (R17a)C(=0)N(R18)-, (R17)(R18)N-, (R17)(R18)NC(=0)N(R19)-, and R17aS02N(R18)-; 'cycloalkyl', 'cycloalkenyl' and 'carbocycle' may be substituted with 1 to 2 substituents selected from the group consisting of halogen, oxo, Ci-6 alkyl, haloalkyl, R17aC(=0)-, R17aS02-, R17bO-, R17aOC(=0)-, R17aC(=0)0-, (R17)(R18)NC(=0)-, (R17a)C(=0)N(R18)-, (R17)(R18)N-, (R17)(R18)NC(=0)N(R19)-, and R17aS02N(R18)-; 'aryl' may be substituted with 1 to 2 substituents selected from the group consisting of halogen, nitro, cyano, hydroxy, alkyl, perhaloalkyl, alkyl-O-, perhaloalkyl-O-, alkyl(H)N-, alkyl(alkyl)N-, H2N-, alkyl-S02-, alkyl-C(=0)(H)N-, alkyl-C(=0)(alkyl)N-, alkyl(H)NC(=0)-, alkyl(alkyl)NC(=0)-, H2NC(=0)-, alkyl(H)NS02-, alkyl(alkyl)NS02-, and H2NS02-;
'heteroaryl' may be substituted with 1 to 2 substituents selected from the group consisting of halogen, nitro, cyano, hydroxy, alkyl, perhaloalkyl, alkyl-O-, perhaloalkyl-O-, alkyl-S02-, H2N-, alkyl(H)N-, alkyl(alkyl)N-, alkyl-C(=0)(H)N-, alkyl-C(=0)(alkyl)N-, NH2C(=0)-, alkyl(H)NC(=0)-, alkyl(alkyl)NC(=0)-, NH2S02-, alkyl(H)NS02-, and alkyl(alkyl)NS02-; ring carbon(s) of 'heterocycle' may be substituted with 1 to 2 substituents selected from the group consisting of halogen, nitro, cyano, oxo, alkyl, R17bO-, R17aOC(=0)-, R17aC(=0)0-, R17(H)NC(=0)-, R17(alkyl)NC(=0)-, R17(H)N-, R17(alkyl)N-, R17aC(=0)(H)N-, R17(H)NC(=0)(H)N-, and R17(alkyl)NC(=0)(H)N-; the substituents on ring nitrogen(s) of 'heterocycle' are selected from the group consisting of alkyl, R17aS02-, R17aC(=0)-, R17aOC(=0)-, R17(H)NC(=0)-, and R17(alkyl)NC(=0);
R17, R18 and R19 are independently selected from hydrogen and alkyl; R17a is selected from alkyl and perhaloalkyl;
R17b is selected from the group consisting of hydrogen, alkyl, and perhaloalkyl;
Whenever a range of the number of atoms in a structure is indicated (e.g., a Ci-i2, Ci-8, Ci-
6, or Ci-4 alkyl, alkylamino, etc.), it is specifically contemplated that any sub-range or individual number of carbon atoms falling within the indicated range also can be used. Thus, for instance, the recitation of a range of 1 -8 carbon atoms (e.g., CrC8), 1 -6 carbon atoms (e.g., CrC6), 1 -4 carbon atoms (e.g., CrC4), 1 -3 carbon atoms (e.g., CrC3), or 2-8 carbon atoms (e.g., C2-C8) as used with respect to any chemical group (e.g., alkyl, alkylamino, etc.) referenced herein encompasses and specifically describes 1 , 2, 3, 4, 5, 6,
7, 8, 9, 10, 1 1 , and/or 12 carbon atoms, as appropriate, as well as any sub-range thereof (e.g., 1 -2 carbon atoms, 1 -3 carbon atoms, 1 -4 carbon atoms, 1 -5 carbon atoms, 1 -6 carbon atoms, 1 -7 carbon atoms, 1 -8 carbon atoms, 1 -9 carbon atoms, 1 -10 carbon atoms, 1 -1 1 carbon atoms, 1 -12 carbon atoms, 2-3 carbon atoms, 2-4 carbon atoms, 2-5 carbon atoms, 2-6 carbon atoms, 2-7 carbon atoms, 2-8 carbon atoms, 2-9 carbon atoms, 2-10 carbon atoms, 2-1 1 carbon atoms, 2-12 carbon atoms, 3-4 carbon atoms, 3-5 carbon atoms, 3-6 carbon atoms, 3-7 carbon atoms, 3-8 carbon atoms, 3-9 carbon atoms, 3-10 carbon atoms, 3-1 1 carbon atoms, 3-12 carbon atoms, 4-5 carbon atoms, 4-6 carbon atoms, 4-7 carbon atoms, 4-8 carbon atoms, 4-9 carbon atoms, 4-10 carbon atoms, 4-1 1 carbon atoms, and/or 4-12 carbon atoms, etc., as appropriate).
One of the embodiments of the present invention is compound of formula (I) as described above, wherein, ring A is particularly selected as cyclopentane;
In any of the embodiments of the invention described above, Y is particularly selected as - N(R2)-; In any of the embodiments of the invention described above, R1 is particularly selected as R13aOC(=0)N(R14)CRa(Rb)C(=0)N(R15)-;
In any of the embodiments of the invention described above, m is particularly selected as 0 at all the occurrences;
In any of the embodiments of the invention described above, R2 is particularly selected as R13aOC(=0)N(R14)CRa(Rb)C(=0)-;
In any of the embodiments of the invention described above, R3 is particularly selected from NH and O;
In any of the embodiments of the invention described above, n is particularly selected as 0;
In any of the embodiments of the invention described above, R5 and R6 are independently selected from hydrogen and halogen, or R5 and R6 together with the carbon atoms to which they are attached form a six membered carbocycle;
In any of the embodiments of the invention described above, R7 and R10 are particularly selected as hydrogen, or R7 and R10 together with the carbon atoms to which they are attached form a five or six membered carbocycle, the said carbocycle is unsubstituted or substituted with one or two alkyl groups;
In any of the embodiments of the invention described above, R9 and R8 are particularly selected as hydrogen, or R9 and R8 together with the carbon atoms to which they are attached form a six or seven membered carbocycle, or R9 and R8 together with the carbon atoms to which they are attached form a seven membered heterocycle containing one heteroatom;
In any of the embodiments of the invention described above, R11 and R12 are particularly selected as hydrogen; In any of the embodiments of the invention described above, ring D is particularly selected as
Figure imgf000010_0001
In any of the embodiments of the invention described above, ring A is particularly selected
Figure imgf000010_0002
as cyclopentane, ring D is particularly selected as , R1 is particularly selected as R13aOC(=0)N(R14)CRa(Rb)C(=0)N(R15)-, m is particularly selected as 0 at all the occurrences, R3 is particularly selected from NH and O, n is particularly selected as 0, R5 and R6 are independently selected from hydrogen and halogen, or R5 and R6 together with the carbon atoms to which they are attached form a six membered carbocycle, R7 and R10 are particularly selected as hydrogen, or R7 and R10 together with the carbon atoms to which they are attached form a five or six membered carbocycle, the said carbocycle is unsubstituted or substituted with one or two alkyl groups, R9 and R8 are particularly selected as hydrogen, or R9 and R8 together with the carbon atoms to which they are attached form a six or seven membered carbocycle, or R9 and R8 together with the carbon atoms to which they are attached form a seven membered heterocycle containing one heteroatom, R11 and R12 are particularly selected as hydrogen, such that at least one cyclic system is formed out of either R5 and R6, R8 and R9, or R7 and R10; General terms used in formula can be defined as follows; however, the meaning stated should not be interpreted as limiting the scope of the term per se.
The term "alkyl", as used herein, means a straight chain or branched hydrocarbon containing from 1 to 20 carbon atoms. Preferably the alkyl chain may contain 1 to 10 carbon atoms. More preferably alkyl chain may contain up to 6 carbon atoms. Representative examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, and n- hexyl.
'Alkyl' as defined hereinabove may be substituted with one or more substituents selected independently from the group comprising of halogen, oxo, Ci -6 alkyl, haloalkyl, aryl, heteroaryl, cycloalkyi, heterocyclyl, R17aC(=0)-, R17aS02-, R17bO-, R17aOC(=0)-, R17aC(=0)0-, (R17)(R18)NC(=0)-, (R17a)C(=0)N(R18)-, (R17)(R18)N-, (R17)(R18)NC(=0)N(R19)-, and R17aS02N(R18)-; wherein, R17, R18 and R19 are independently selected from hydrogen and alkyl, R17a is selected from alkyl and perhaloalkyl, R17b is selected from the group consisting of hydrogen, alkyl, and perhaloalkyl.
The term "haloalkyl" used herein means an alkyl group as defined hereinabove wherein at least one of the hydrogen atoms of the said alkyl group is substituted with halogen. The haloalkyl group is exemplified by monofluoromethyl, 1 ,2-dichloroethyl and the like. The term "perhaloalkyl" means an alkyl group as defined hereinabove wherein all the hydrogen atoms of the said alkyl group are substituted with halogen. The perhaloalkyl group is exemplified by trifluoromethyl, pentafluoroethyl and the like.
The term "cycloalkyi" as used herein, means a monocyclic, bicyclic, or tricyclic non- aromatic ring system containing from 3 to 14 carbon atoms, preferably monocyclic cycloalkyi ring containing 3 to 6 carbon atoms. Examples of monocyclic ring systems include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Bicyclic ring systems are also exemplified by a bridged monocyclic ring system in which two non-adjacent carbon atoms of the monocyclic ring are linked by an alkylene bridge. Representative examples of bicyclic ring systems include, but are not limited to, bicyclo[3.1 .1 ]heptane, bicyclo[2.2.1 ]heptane, bicyclo[2.2.2]octane, bicyclo[3.2.2]nonane, bicyclo[3.3.1 ]nonane, and bicyclo[4.2.1 ]nonane, bicyclo[3.3.2]decane, bicyclo[3.1 .0]hexane, bicyclo[410]heptane, bicyclo[3.2.0]heptanes, octahydro-1 H-indene. Tricyclic ring systems are also exemplified by a bicyclic ring system in which two non- adjacent carbon atoms of the bicyclic ring are linked by a bond or an alkylene bridge. Representative examples of tricyclic-ring systems include, but are not limited to, tricyclo[3.3.1 .03 7]nonane and tricyclo[3.3.1 .13 7]decane (adamantane). The term cycloalkyl also include spiro systems wherein one of the ring is annulated on a single carbon atom such ring systems are exemplified by spiro[2.5]octane, spiro[4.5]decane, spiro[bicyclo[4.1 .0]heptane-2,1 '-cyclopentane], hexahydro-2'H-spiro[cyclopropane-1 ,1 '- pentalene].
The term "cycloalkenyl" as used herein, means a cycloalkyl group as defined above containing at least one double bond.
The term "carbocycle" as used herein, means a cyclic system made up of carbon atoms, which includes cycloalkyl, cycloalkenyl and aryl.
Cycloalkyl, cycloalkenyl and carbocycle as defined hereinabove may be substituted with one or more substituents selected independently from the group comprising of halogen, oxo, Ci-6 alkyl, haloalkyl, R17aC(=0)-, R17aS02-, R17bO-, R17aOC(=0)-, R17aC(=0)0-, (R17)(R18)NC(=0)-, (R17a)C(=0)N(R18)-, (R17)(R18)N-, (R17)(R18)NC(=0)N(R19)-, and R17aS02N(R18)-; wherein, R17, R18 and R19 are independently selected from hydrogen and alkyl, R17a is selected from alkyl and perhaloalkyl, R17b is selected from the group consisting of hydrogen, alkyl, and perhaloalkyl.
The term "aryl" refers to a monovalent monocyclic, bicyclic or tricyclic aromatic hydrocarbon ring system. Examples of aryl groups include phenyl, naphthyl, anthracenyl, fluorenyl, indenyl, azulenyl, and the like. Aryl group also include partially saturated bicyclic and tricyclic aromatic hydrocarbons such as tetrahydro-naphthalene. The said aryl group also includes aryl rings fused with heteroaryl or heterocyclic rings such as 2,3-dihydro- benzo[1 ,4]dioxin-6-yl, 2,3-dihydro-benzo[1 ,4]dioxin-5-yl, 2,3-dihydro-benzofuran-5-yl, 2,3- dihydro-benzofuran-4-yl, 2,3-dihydro-benzofuran-6-yl, 2,3-dihydro-benzofuran-6-yl, 2,3- dihydro-1 H-indol-5-yl, 2,3-dihydro-1 H-indol-4-yl, 2,3-dihydro-1 H-indol-6-yl, 2,3-dihydro-1 H- indol-7-yl, benzo[1 ,3]dioxol-4-yl, benzo[1 ,3]dioxol-5-yl, 1 ,2,3,4-tetrahydroquinolinyl, 1 ,2,3,4-tetrahydroisoquinolinyl, 2,3-dihydrobenzothien-4-yl, 2-oxoindolin-5-yl. Aryl as defined hereinabove may be substituted with one or more substituents selected independently from the group consisting of halogen, nitro, cyano, hydroxy, alkyl, perhaloalkyl, alkyl-O-, perhaloalkyl-O-, alkyl(H)N-, alkyl(alkyl)N-, H2N-, alkyl-S02-, alkyl- C(=0)(H)N-, alkyl-C(=0)(alkyl)N-, alkyl(H)NC(=0)-, alkyl(alkyl)NC(=0)-, H2NC(=0)-, alkyl(H)NS02-, alkyl(alkyl)NS02-, and H2NS02-.
The term "heteroaryl" refers to a 5-14 membered monocyclic, bicyclic, or tricyclic ring system having 1 -4 ring heteroatoms selected from O, N, or S, and the remainder ring atoms being carbon (with appropriate hydrogen atoms unless otherwise indicated), wherein at least one ring in the ring system is aromatic. Heteroaryl groups may be optionally substituted with one or more substituents. In one embodiment, 0, 1 , 2, 3, or 4 atoms of each ring of a heteroaryl group may be substituted by a substituent. Exampies of heteroaryl groups include pyridyi, 1 -oxo-pyridyl, furanyi, thienyi, pyrroiyl, oxazolyi, oxadiazolyl, imidazolyl, thiazolyl, isoxazolyl, quinolinyl, pyrazolyl, isothiazolyl, pyridazinyl, pyrimidinyi, pyrazinyl, triazinyl. triazolyl. thiadiazolyl, isoquinolinyl, benzoxazolyl, benzofuranyl, indolizinyl, imidazopyridyl, tetrazolyi, benzimidazolyi, benzothiazolyl. benzothiadiazolyl, benzoxadiazolyl, indolyl. azaindolyl, imidazopyridyl. quinazolinyl. purinyl, pyrrolo[2,3]pyrimidinyi, pyrazoio[3,4]pyrimidinyl, and benzo(b)thienyl, 2,3- thiadiazolyl, 1 H-pyrazoio[5,1 -c]-1 ,2,4-triazoiyi, pyrrolo[3,4-d]-1 ,2,3-triazolyL cyciopentairiazolyL 3H-pyrroio[3,4-c] isoxazolyl and the like.
Heteroaryl as defined hereinabove may be substituted with one or more substituents selected independently from the group consisting of halogen, nitro, cyano, hydroxy, alkyl, perhaloalkyl, alkyl-O-, perhaloalkyl-O-, alkyl-S02-, H2N-, alkyl(H)N-, alkyl(alkyl)N-, alkyl- C(=0)(H)N-, alkyl-C(=0)(alkyl)N-, NH2C(=0)-, alkyl(H)NC(=0)-, alkyl(alkyl)NC(=0)-, NH2S02-, alkyl(H)NS02-, and alkyl(alkyl)NS02-.
The term "heterocycle" or "heterocyclic" as used herein, means a 'cycloalkyl' group wherein one or more of the carbon atoms replaced by -0-, -S-, -S(02)-, -S(O)-, -N(Rm)-, - Si(Rm)Rn-, wherein, Rm and Rn are independently selected from hydrogen, alkyl, aryl, heteroaryl, cycloalkyi, and heterocyclyl. The heterocycle may be connected to the parent molecular moiety through any carbon atom or any nitrogen atom contained within the heterocycle. Representative examples of monocyclic heterocycle include, but are not limited to, azetidinyl, azepanyl, aziridinyl, diazepanyl, 1 ,3-dioxanyl, 1 ,3-dioxolanyl, 1 ,3- dithiolanyl, 1 ,3-dithianyl, imidazolinyl, imidazolidinyl, isothiazolinyl, isothiazolidinyl, isoxazolinyl, isoxazolidinyl, morpholinyl, oxadiazolinyl. oxadiazolidinyl, oxazolinyl, oxazolidinyl, piperazinyl, piperidinyl, pyranyl, pyrazolinyl, pyrazolidinyl. pyrrolinyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, thiadiazolinyl, thiadiazolidinyl, thiazolinyl, thiazolidinyl, thiomorpholinyl, 1 .1 -dioxidothiomorpholinyl (thiomorpholine sulfone). thiopyranyl, and trithianyl. Representative examples of bicyclic heterocycle include, but are not limited to 1 ,3-benzodioxolyl, 1 ,3-benzodithiolyl, 2,3-dihydro-1 ,4-benzodioxinyl, 2,3- dihydro-1 -benzofuranyl, 2,3-dihydro-1 -benzothienyl, 2,3-dihydro-1 H-indolyl and 1 ,2,3,4- tetrahydroquinolinyl. The term heterocycle also include bridged heterocyclic systems such as azabicyclo[3.2.1 ]octane, azabicyclo[3.3.1 ]nonane and the like.
Heterocyclyl group may be substituted on ring carbons with one or more substituents selected independently from the group consisting of halogen, nitro, cyano, oxo, alkyl, R17bO-, R17aOC(=0)-, R17aC(=0)0-, R17(H)NC(=0)-, R17(alkyl)NC(=0)-, R17(H)N-, R17(alkyl)N-, R17aC(=0)(H)N-, R17(H)NC(=0)(H)N-, and R17(alkyl)NC(=0)(H)N-; wherein, R17 is selected from hydrogen and alkyl, R17a is selected from alkyl and perhaloalkyl, R17b is selected from the group consisting of hydrogen, alkyl, and perhaloalkyl.
Heterocyclyl group may further be substituted on ring nitrogen(s) with substituents selected from the group comprising of alkyl, R17aS02-, R17aC(=0)-, R17aOC(=0)-, R17(H)NC(=0)-, and R17(alkyl)NC(=0); wherein, R17 is selected from hydrogen and alkyl, R17a is selected from alkyl and perhaloalkyl.
The term Oxo' means a divalent oxygen (=0) attached to the parent group. For example oxo attached to carbon forms a carbonyl, oxo substituted on cyclohexane forms a cyclohexanone, and the like. The term 'annulated' means the ring system under consideration is either annulated with another ring at a carbon atom of the cyclic system or across a bond of the cyclic system as in the case of fused or spiro ring systems.
The term 'bridged' means the ring system under consideration contain an alkylene bridge having 1 to 4 methylene units joining two non adjuscent ring atoms.
A compound its stereoisomers, racemates, pharmaceutically acceptable salt thereof as described hereinabove wherein the compound of general formula I is selected from:
1 . methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)- 1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
2. methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate.
3. methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)- 1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
4. methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)- 1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
5. methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate.
6. methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate.
7. methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate.
methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2 yl)carbamate.
methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2 yl)carbamate.
methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)- 1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate.
methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2 yl)carbamate.
methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate.
methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)- 1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)- 1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate. methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol 2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol 2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol 2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-4-chloro-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-4-chloro-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-5,6-dihydro-4H-benzo[3,4]cyclohepta[1 ,2-d]oxazol-8 yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate.
methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydrobenzo[2,3]oxepino[4,5-d]oxazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate.
methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydrobenzo[2,3]oxepino[4,5-d]oxazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate.
methyl ((S)-1 -(((1 S,2S)-2-(7-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate.
methyl ((S)-1 -(((1 S,2R)-2-(7-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate.
26. methyl ((S)-1 -(((1 S,2R)-2-(7-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol- 2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
27. methyl ((S)-1 -(((1 S,2S)-2-(7-(4-(2-((S)-1 -((S)-2-amino-3-methylbutanoyl)pyrrolidin-2- yl)-1 H-imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol-2-yl)cyclopentyl)amino)-3- methyl-1 -oxobutan-2-yl)carbamate.
28. methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)- 1 H-imidazol-2-yl)cyclopentyl)(ethyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
29. methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)- 1 H-imidazol-2-yl)cyclopentyl)(methyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
30. methyl ((S)-1 -(((1 S,2S)-2-(5-(7-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)-9H-fluoren-2-yl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
31. methyl ((S)-1 -(((1 S,2S)-2-(5-(7-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)-9,9-dimethyl-9H-fluoren-2-yl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
32. methyl ((S)-1 -(((1 S,2S)-2-(5-(7-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenanthren-2-yl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
According to a feature of the present invention, the compounds of general formula I where all the symbols are as defined earlier, can be prepared by methods given in Schemes given below or example. Representative procedures are shown below, however; the disclosure should not be construed to limit the scope of the invention arriving at compound of formula I as disclosed hereinabove.
Scheme 1 : Halogenation of synthon 1 (could be prepared by methods well known in the art, Tetrahedron, 2000, 56, 5225-5240) may be achieved leading to the formation of 2 which could be coupled to the protected (P = Boc, Cbz) β-aminocyclopentane carboxylic acid 3 (individual isomers RR, SS, RS and SR could be made by methods known in the literature, Tet. Lett., 2002, 43, 5401 -5404; J. Am. Chem. Soc, 1999, 121 , 7574-7581 ; Tet. Asymm., 2008, 19, 2796-2803) using an appropriate base like triethylamine, 4- methylmorpholine, diisopropylethylamine and solvents like DMF, 1 ,4-dioxane, THF, acetonitrile to yield 4. Cyclization of 4 using NH4OAc could lead to the formation of 5 which may be deprotected and coupled with (S)-2-((methoxycarbonyl)amino)-3-methylbutanoic acid using amide coupling reagents like carbodiimides, phosphonium salts, uronium salts, carbonyldiimidazole in presence of a mild base like triethylamine, 4-methylmorpholine, diisopropylethylamine in a solvent like DMF, 1 ,4-dioxane, THF, dichloromethane leading to the formation of 6. Alternatively, de-protected form of 5 can be coupled to similar moieties by methods known in the literature to furnish compounds of formula I.
Figure imgf000019_0001
Scheme 1 :
Scheme 2: Reduction of 7, could be prepared by methods known in the literature, using either of Pd/C, Pt02 or Raney Ni, Fe/HCI, tin(ll) chloride, titanium(ll l) chloride or zinc/AcOH could lead to the formation of 8. Diazotization followed by Sandmeyer reaction of 8 might lead to the formation of 9 which on acidic/basic hydrolysis might result in formation of 10. One carbon homologation followed by halogenations might lead to formation of 11 from 10. Intermediate 11 could further be O-alkylated with 3 in presence of a suitable base and solvent and cyclized using NH4OAc to yield 13. Pd-catalyzed C-C coupling of 13 with 14 (ref WO2009102318 A1 ) might lead to the formation of 15 which on de-protection and coupling with (S)-2-((methoxycarbonyl)amino)-3-methylbutanoic acid or similar moieties might lead to the synthesis of 16 or compounds of formula I.
Figure imgf000020_0001
Scheme 2
Scheme 3: Diazotization followed by Sandmeyer reaction on synthon 17 may lead to the formation of 18. Synthesis of 19 may be accomplished by halogenation of 18 with suitable halogenating agents. O-alkylation of 19 with Boc-L-proline using an appropriate base like triethylamine, 4-methylmorpholine, diisopropylethylamine and solvent like DMF, 1 ,4- dioxane, THF, acetonitrile might lead to the formation of 20. Cyclization of intermediate 20 with NH4OAc might lead to the formation of tricyclic system 21. Synthon 25 might be synthesized from 22 wherein 22 could be O-alkylated with 3 followed by cyclization using NH4OAC and finally generation of boronate/boronic acid via Pd-catalysis. Synthons 21 and
25 could be linked through C-C coupling method using Pd catalysts to obtain 26. Synthesis of the compound 27 or compounds of formula I may be accomplished by de-protection of
26 and coupling the de-protected amine to (S)-2-((methoxycarbonyl)amino)-3- methylbutanoic acid or similar moieties using a suitable combination of coupling reagents, bases and solvents.
Figure imgf000021_0001
Scheme 3
Scheme 4: In another process, Wittig reaction on m-halo substituted benzaldehyde 28 using the appropriate base and solvent might lead to the formation of 29 which on reduction with Pt-based reagents amongst others may lead to the formation of acid 30 which under acidic conditions may lead to the formation of 34. Alternatively, 34 could also be synthesized from the phenol 31 which on alkylation might lead to the formation of 32 which on acidic/basic hydrolysis could lead to the formation of the acid 33. Cyclization of 33 under acidic conditions might lead to the formation of 34. (J. Med. Chem., 2000, 43, 2049-2063; ibid, 2005, 48, 7351 -7362; WO2009102633 A1 ). On treatment with isoamyl nitrite in an appropriate solvent, oxime 35 might be generated from 34 which upon reaction with Boc-L-prolinal under basic conditions may lead to formation of 36. Treatment of 36 with triethyl phosphite might lead to the formation of either 37 or 38. Pd-catalyzed C-C coupling of either 37 or 38 with 25 followed by de-protection and coupling with (S)-2- ((methoxycarbonyl)amino)-3-methylbutanoic acid might lead to the formation of 39 or 40.
Symmetrical compounds could be prepared by coupling 42 (generated from 34) with the boronate/boronic acid of 42 through the use of Pd catalyzed C-C coupling reactions to yield 44 or compounds of formula I.
Figure imgf000022_0001
Figure imgf000022_0002
Scheme 4
Scheme 5: Acetophenone 45, which is readily available or prepared from suitable reagents, could be protected with a suitable protecting group to get 46 which could be then halogenated and O-alkylated to possibly yield 48. Cyclization of 48 might be accomplished using ammonium acetate to yield 49 which upon de-protection of benzylic hydroxyl group followed by alkylation under basic conditions with either synthon 37 or 38 might lead to the synthesis of 50. De-protection of 50 under acidic conditions and sequential coupling with (S)-2-((methoxycarbonyl)amino)-3-methylbutanoic acid or similar moieties might lead to the formation of 51 or compounds of formula I.
Figure imgf000023_0001
Compounds of the present invention were prepared using synthetic schemes provided below:
Compounds 1 to 3 were prepared by following the route provided in Scheme I
Figure imgf000024_0001
Compounds 4 to 15 were prepared by following the route provided in Scheme II
ntitative ative
Figure imgf000025_0001
Compounds 16 to 20 were prepared by following vided in Scheme III
Figure imgf000025_0002
Figure imgf000025_0003
Compounds 21 to 23 were prepared by following the route provided in Scheme IV
Figure imgf000026_0001
Compounds 24 to 27 were prepared by following the route provided in Scheme V
Figure imgf000026_0002
Compounds 28 and 29 were prepared by following the route provided in Scheme VI
Figure imgf000027_0001
Compounds 30 to 32 were prepared by following the route provided in Scheme VII
Figure imgf000028_0001
Scheme VII A further embodiment of the present invention includes pharmaceutical compositions comprising any single compound, a combination of two or more compounds delineated herein, or a pharmaceutically acceptable salt thereof, with a pharmaceutically acceptable carrier or excipient.
Yet a further embodiment of the present invention is a pharmaceutical composition comprising any single compound or a combination of two or more compounds delineated herein, or a pharmaceutically acceptable salt thereof, in combination with one or more agents known in the art, with a pharmaceutically acceptable carrier or excipient. It will be further appreciated that compounds of the present invention can be administered as the sole active pharmaceutical agent, or used in combination with one or more agents to treat or prevent hepatitis C infections or the symptoms associated with HCV infection. Other agents to be administered in combination with a compound or combination of compounds of the present invention include therapies for diseases caused by HCV infection that suppresses HCV viral replication by direct or indirect mechanisms. These agents include, but not limited to, host immune modulators (for example, interferon-alpha, pegylated interferon-alpha, consensus interferon, interferon-beta, interferon-gamma, CpG oligonucleotides and the like); antiviral compounds that inhibit host cellular functions such as inosine monophosphate dehydrogenase (for example, ribavirin and the like); cytokines that modulate immune function (for example, interleukin 2, interleukin 6, and interleukin 12); a compound that enhances the development of type 1 helper T cell response; interfering RNA; anti-sense RNA; vaccines comprising HCV antigens or antigen adjuvant combinations directed against HCV; agents that interact with host cellular components to block viral protein synthesis by inhibiting the internal ribosome entry site (IRES) initiated translation step of HCV viral replication or to block viral particle maturation and release with agents targeted toward the viroporin family of membrane proteins such as, for example, HCV P7 and the like; and any agent or combination of agents that inhibit the replication of HCV by targeting other proteins of the viral genome involved in the viral replication and/or interfere with the function of other viral targets, such as inhibitors of NS3/NS4A protease, NS3 helicase, NS5B polymerase, NS4A protein and NS5A protein. According to yet another embodiment, the pharmaceutical compositions of the present invention may further comprise inhibitor(s) of other targets in the HCV life cycle, including, but not limited to, helicase, polymerase, metalloprotease, NS4A protein, NS5A protein, and internal ribosome entry site (IRES).
Accordingly, one embodiment of the present invention is directed to a method for treating or preventing an infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment one or more agents selected from the group consisting of a host immune modulator and a second or more antiviral agents, or a combination thereof, with a therapeutically effective amount of a compound or combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof. Examples of the host immune modulator are, but not limited to, interferon-alpha, pegylated-interferon-alpha, interferon-beta, interferon-gamma, a cytokine, a vaccine, and a vaccine comprising an antigen and an adjuvant, and said second antiviral agent inhibits replication of HCV either by inhibiting host cellular functions associated with viral replication or by targeting proteins of the viral genome. Example of the RNA-containing virus includes, but not limited to, hepatitis C virus (HCV).
A further embodiment of the present invention is directed to a method of treating or preventing infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment an agent or combination of agents that treat or alleviate symptoms of HCV infection including cirrhosis and inflammation of the liver, with a therapeutically effective amount of a compound or combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof. Example of the RNA- containing virus includes, but not limited to, hepatitis C virus (HCV).
Yet another embodiment of the present invention provides a method of treating or preventing infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment one or more agents that treat patients for disease caused by hepatitis B (HBV) infection, with a therapeutically effective amount of a compound or a combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof. An agent that treats patients for disease caused by hepatitis B (HBV) infection may be for example, but not limited thereto, L-deoxythymidine, adefovir, lamivudine or tenfovir, or any combination thereof. Example of the RNA-containing virus includes, but not limited to, hepatitis C virus (HCV). A further embodiment of the present invention provides a method of treating or preventing infection caused by an RNA-containing virus comprising co-administering to a patient in need of such treatment one or more agents that treat patients for disease caused by human immunodeficiency virus (HIV) infection, with a therapeutically effective amount of a compound or a combination of compounds of the present invention, or a pharmaceutically acceptable salt thereof. The agent that treats patients for disease caused by human immunodeficiency virus (H IV) infection may include, but is not limited thereto, ritonavir, lopinavir, indinavir, nelfmavir, saquinavir, amprenavir, atazanavir, tipranavir, TMC-1 14, fosamprenavir, zidovudine, lamivudine, didanosine, stavudine, tenofovir, zalcitabine, abacavir, efavirenz, nevirapine, delavirdine, TMC-125, L-870812, S-1360, enfuvirtide (T- 20) or T-1249, or any combination thereof. Example of the RNA-containing virus includes, but not limited to, hepatitis C virus (HCV). It can occur that a patient may be co-infected with hepatitis C virus and one or more other viruses, including but not limited to human immunodeficiency virus (HIV), hepatitis A virus (HAV) and hepatitis B virus (HBV). Thus also contemplated is combination therapy to treat such co-infections by co-administering a compound according to the present invention with at least one of an HIV inhibitor, an HAV inhibitor and an HBV inhibitor.
In addition, the present invention provides the use of a compound or a combination of compounds of the invention, or a therapeutically acceptable salt thereof, and one or more agents selected from the group consisting of a host immune modulator and a second or more antiviral agents, or a combination thereof, to prepare a medicament for the treatment of an infection caused by an RNA-containing virus in a patient, particularly hepatitis C virus. Examples of the host immune modulators include, but are not limited to, interferon- alpha, pegylated-interferon-alpha, interferon-beta, interferon-gamma, a cytokine, a vaccine, and a vaccine comprising an antigen and an adjuvant, and said second antiviral agent inhibits replication of HCV either by inhibiting host cellular functions associated with viral replication or by targeting proteins of the viral genome.
When used in the above or other treatments, combination of compound or compounds of the present invention, together with one or more agents as defined herein above, can be employed in pure form or, where such forms exist, in pharmaceutically acceptable salt thereof. Alternatively, such combination of therapeutic agents can be administered as a pharmaceutical composition containing a therapeutically effective amount of the compound or combination of compounds of interest, or their pharmaceutically acceptable salt thereof, in combination with one or more agents as defined hereinabove, and a pharmaceutically acceptable carrier. Such pharmaceutical compositions can be used for inhibiting the replication of an RNA-containing virus, particularly Hepatitis C virus (HCV), by contacting said virus with said pharmaceutical composition. In addition, such compositions are useful for the treatment or prevention of an infection caused by an RNA-containing virus, particularly Hepatitis C virus (HCV).
Hence, a still further embodiment of the invention is directed to a method of treating or preventing infection caused by an RNA-containing virus, particularly a hepatitis C virus (HCV), comprising administering to a patient in need of such treatment a pharmaceutical composition comprising a compound or combination of compounds of the invention or a pharmaceutically acceptable salt thereof, and one or more agents as defined hereinabove, with a pharmaceutically acceptable carrier.
When administered as a combination, the therapeutic agents can be formulated as separate compositions which are given at the same time or within a predetermined period of time, or the therapeutic agents can be given as a single unit dosage form.
Antiviral agents contemplated for use in such combination therapy include agents (compounds or biologicals) that are effective to inhibit the formation and/or replication of a virus in a mammal, including but not limited to agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of a virus in a mammal. Such agents can be selected from another anti-HCV agent; an HIV inhibitor; an HAV inhibitor; and an HBV inhibitor.
Other agents to be administered in combination with a compound of the present invention include a cytochrome P450 monooxygenase inhibitor, which is expected to inhibit metabolism of the compounds of the invention. Therefore, the cytochrome P450 monooxygenase inhibitor would be in an amount effective to inhibit metabolism of the compounds of the present invention. Accordingly, the CYP inhibitor is administered in an amount such that the bioavailability of the compounds of the present invention is increased in comparison to the bioavailability in the absence of the CYP inhibitor.
The term 'room temperature' used in the specification denotes any temperature ranging between about 20 °C to about 40 °C, except and otherwise it is specifically mentioned in the specification.
The intermediates and the compounds of the present invention may obtained in pure form in a manner known per se, for example, by distilling off the solvent in vacuum and re- crystallizing the residue obtained from a suitable solvent, such as pentane, diethyl ether, isopropyl ether, chloroform, dichloromethane, ethyl acetate, acetone or their combinations or subjecting it to one of the purification methods, such as column chromatography (e.g., flash chromatography) on a suitable support material such as alumina or silica gel using eluent such as dichloromethane, ethyl acetate, hexane, methanol, acetone and their combinations. Preparative LC-MS method is also used for the purification of molecules described herein.
Salts of compound of formula I can be obtained by dissolving the compound in a suitable solvent, for example in a chlorinated hydrocarbon, such as methyl chloride or chloroform or a low molecular weight aliphatic alcohol, for example, ethanol or isopropanol, which was then treated with the desired acid or base as described in Berge S.M. et al. "Pharmaceutical Salts, a review article in Journal of Pharmaceutical sciences volume 66, page 1 -19 (1977)" and in handbook of pharmaceutical salts properties, selection, and use by P.H.Einrich Stahland Camille G.wermuth, Wiley- VCH (2002). Lists of suitable salts can also be found in Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing Company, Easton, PA, 1990, p. 1445, and Journal of Pharmaceutical Science, 66, 2-19 (1977). For example, they can be a salt of an alkali metal (e.g., sodium or potassium), alkaline earth metal (e.g., calcium), or ammonium of salt.
The compound of the invention or a composition thereof can potentially be administered as a pharmaceutically acceptable acid-addition, base neutralized or addition salt, formed by reaction with inorganic acids, such as hydrochloric acid, hydrobromic acid, perchloric acid, nitric acid, thiocyanic acid, sulfuric acid, and phosphoric acid, and organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid, malonic acid, succinic acid, maleic acid, and fumaric acid, or by reaction with an inorganic base, such as sodium hydroxide, potassium hydroxide. The conversion to a salt is accomplished by treatment of the base compound with at least a stoichiometric amount of an appropriate acid. Typically, the free base is dissolved in an inert organic solvent such as diethyl ether, ethyl acetate, chloroform, ethanol, methanol, and the like, and the acid is added in a similar solvent. The mixture is maintained at a suitable temperature (e.g., between 0 °C and 50 °C). The resulting salt precipitates spontaneously or can be brought out of solution with a less polar solvent. The stereoisomers of the compounds of formula I of the present invention may be prepared by stereospecific syntheses or resolution of the achiral compound using an optically active amine, acid or complex forming agent, and separating the diastereomeric salt/complex by fractional crystallization or by column chromatography. The term "prodrug" denotes a derivative of a compound, which derivative, when administered to warm-blooded animals, e.g. humans, is converted into the compound (drug). The enzymatic and/or chemical hydrolytic cleavage of the compounds of the present invention occurs in such a manner that the proven drug form (parent carboxylic acid drug) is released, and the moiety or moieties split off remain nontoxic or are metabolized so that nontoxic metabolic products are produced. For example, a carboxylic acid group can be esterified, e.g., with a methyl group or ethyl group to yield an ester. When an ester is administered to a subject, the ester is cleaved, enzymatically or non- enzymatically, reductively, oxidatively, or hydrolytically, to reveal the anionic group. An anionic group can be esterified with moieties (e.g., acyloxymethyl esters) which are cleaved to reveal an intermediate compound which subsequently decomposes to yield the active compound.
The prodrugs can be prepared in situ during the isolation and purification of the compounds, or by separately reacting the purified compound with a suitable derivatizing agent. For example, hydroxy groups can be converted into esters via treatment with a carboxylic acid in the presence of a catalyst. Examples of cleavable alcohol prodrug moieties include substituted or unsubstituted, branched or unbranched lower alkyl ester moieties, e.g., ethyl esters, di-lower alkylamino lower-alkyl esters, e.g., dimethylaminoethyl ester, acylamino lower alkyl esters, acyloxy lower alkyl esters (e.g., pivaloyloxymethyl ester), aryl esters, e.g., phenyl ester, aryl-lower alkyl esters, e.g., benzyl ester, optionally substituted, e.g., with methyl, halo, or methoxy substituents aryl and aryl- lower alkyl esters, amides, lower-alkyl amides, di-lwer alkyl amides, and hydroxy amides. Thus the present invention further provides a pharmaceutical composition, containing the compounds of the general formula (I) as defined above, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates and its co-crystals in combination with the usual pharmaceutically acceptable carriers, diluents and the like.
The pharmaceutically acceptable carrier (or excipient) is preferably one that is chemically inert to the compound of the invention and one that has no detrimental side effects or toxicity under the conditions of use. Such pharmaceutically acceptable carriers preferably include saline (e.g., 0.9% saline), Cremophor EL (which is a derivative of castor oil and ethylene oxide available from Sigma Chemical Co., St. Louis, MO) (e.g., 5% Cremophor EL/5% ethanol/90% saline, 10% Cremophor EL/90% saline, or 50% Cremophor EL/50% ethanol), propylene glycol (e.g., 40% propylene glycol/10% ethanol/50% water), polyethylene glycol (e.g., 40% PEG 400/60% saline), and alcohol (e.g., 40% ethanol/60% water). A preferred pharmaceutical carrier is polyethylene glycol, such as PEG 400, and particularly a composition comprising 40% PEG 400 and 60% water or saline. The choice of carrier will be determined in part by the particular compound chosen, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of the pharmaceutical composition of the present invention.
The following formulations for oral, aerosol, parenteral, subcutaneous, intravenous, intraarterial, intramuscular, interperitoneal, rectal, and vaginal administration are merely exemplary and are in no way limiting.
The pharmaceutical compositions can be administered parenterally, e.g., intravenously, intraarterially, subcutaneously, intradermal^, intrathecal^, or intramuscularly. Thus, the invention provides compositions for parenteral administration that comprise a solution of the compound of the invention dissolved or suspended in an acceptable carrier suitable for parenteral administration, including aqueous and non-aqueous, isotonic sterile injection solutions.
Overall, the requirements for effective pharmaceutical carriers for parenteral compositions are well known to those of ordinary skill in the art. See Pharmaceutics and Pharmacy Practice, J.B. Lippincott Company, Philadelphia, PA, Banker and Chalmers, eds., pages 238-250 (1982), and ASHP Handbook on Injectable Drugs, Toissel, 4th ed., pages 622- 630 (1986). Such compositions include solutions containing anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. The compound can be administered in a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol, isopropanol (for example in topical applications), or hexadecyl alcohol, glycols, such as propylene glycol or polyethylene glycol, dimethylsulfoxide, glycerol ketals, such as 2,2-dimethyl-1 ,3-dioxolane- 4-methanol, ethers, such as poly(ethyleneglycol) 400, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose, or emulsifying agents and other pharmaceutical adjuvants. Oils useful in parenteral formulations include petroleum, animal, vegetable, and synthetic oils. Specific examples of oils useful in such formulations include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral oil. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.
Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts, and suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylene polypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-3-aminopropionates, and 2-alkyl-imidazoline quaternary ammonium salts, and (e) mixtures thereof. The parenteral formulations typically will contain from about 0.5% or less to about 25% or more by weight of a compound of the invention in solution. Preservatives and buffers can be used. In order to minimize or eliminate irritation at the site of injection, such compositions can contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations will typically range from about 5% to about 15% by weight. Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol. The parenteral formulations can be presented in unit-dose or multi-dose sealed containers, such as ampoules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets.
Topical formulations, including those that are useful for transdermal drug release, are well known to those of skill in the art and are suitable in the context of the present invention for application to skin.
Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of a compound of the invention dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets, tablets, lozenges, and troches, each containing a pre-determined amount of the compound of the invention, as solids or granules; (c) powders; (d) suspensions in an appropriate liquid; and (e) suitable emulsions. Liquid formulations can include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent. Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and cornstarch. Tablet forms can include one or more of lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible excipients. Lozenge forms can comprise the compound ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising a compound of the invention in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the compound of the invention, such excipients as are known in the art.
An compound of the present invention, alone or in combination with other suitable components, can be made into aerosol formulations to be administered via inhalation. A compound or epimer of the invention is preferably supplied in finely divided form along with a surfactant and propellant. Typical percentages of the compounds of the invention can be about 0.01 % to about 20% by weight, preferably about 1 % to about 10% by weight. The surfactant must, of course, be nontoxic, and preferably soluble in the propellant. Representative of such surfactants are the esters or partial esters of fatty acids containing from 6 to 22 carbon atoms, such as caproic, octanoic, lauric, palmitic, stearic, linoleic, linolenic, olesteric and oleic acids with an aliphatic polyhydric alcohol or its cyclic anhydride. Mixed esters, such as mixed or natural glycerides can be employed. The surfactant can constitute from about 0.1 % to about 20% by weight of the composition, preferably from about 0.25% to about 5%. The balance of the composition is ordinarily propellant. A carrier can also be included as desired, e.g., lecithin, for intranasal delivery. These aerosol formulations can be placed into acceptable pressurized propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also can be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer. Such spray formulations can be used to spray mucosa.
Additionally, the compound of the invention can be made into suppositories by mixing with a variety of bases, such as emulsifying bases or water-soluble bases. Formulations suitable for vaginal administration can be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the compound ingredient, such carriers as are known in the art to be appropriate.
The concentration of the compound in the pharmaceutical formulations can vary, e.g., from less than about 1 % to about 10%, to as much as 20% to 50% or more by weight, and can be selected primarily by fluid volumes, and viscosities, in accordance with the particular mode of administration selected.
For example, a typical pharmaceutical composition for intravenous infusion could be made up to contain 250 ml of sterile Ringer's solution, and 100 mg of at least one compound of the invention. Actual methods for preparing parenterally administrable compounds of the invention will be known or apparent to those skilled in the art and are described in more detail in, for example, Remington's Pharmaceutical Science (17th ed., Mack Publishing Company, Easton, PA, 1985). It will be appreciated by one of ordinary skill in the art that, in addition to the aforedescribed pharmaceutical compositions, the compound of the invention can be formulated as inclusion complexes, such as cyclodextrin inclusion complexes, or liposomes. Liposomes can serve to target a compound of the invention to a particular tissue, such as lymphoid tissue or cancerous hepatic cells. Liposomes can also be used to increase the half-life of a compound of the invention. Many methods are available for preparing liposomes, as described in, for example, Szoka et al., Ann. Rev. Biophys. Bioeng., 9, 467 (1980) and U.S. Patents 4,235,871 , 4,501 ,728, 4,837,028, and 5,019,369.
The present invention also provides a pharmaceutical composition, containing the compounds of the general formula (I) as defined above, its tautomeric forms, its stereoisomers, its analogs, its prodrugs, its isotopes, its metabolites, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates and its co- crystals in combination with the usual pharmaceutically employed carriers, diluents and the like, and for use in any of the methods described herein.
The compounds of the invention can be administered in a dose sufficient to treat the disease, condition or disorder. Such doses are known in the art (see, for example, the Physicians' Desk Reference (2004)). The compounds can be administered using techniques such as those described in, for example, Wasserman et al., Cancer, 36, pp. 1258-1268 (1975) and Physicians' Desk Reference, 58th ed., Thomson PDR (2004).
Suitable doses and dosage regimens can be determined by conventional range-finding techniques known to those of ordinary skill in the art. Generally, treatment is initiated with smaller dosages that are less than the optimum dose of the compound of the present invention. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached. The present method can involve the administration of about 0.1 μg to about 50 mg of at least one compound of the invention per kg body weight of the individual. For a 70 kg patient, dosages of from about 10 μg to about 200 mg of the compound of the invention would be more commonly used, depending on a patient's physiological response. By way of example and not intending to limit the invention, the dose of the pharmaceutically active agent(s) described herein for methods of treating or preventing a disease or condition as described above can be about 0.001 to about 1 mg/kg body weight of the subject per day, for example, about 0.001 mg, 0.002 mg, 0.005 mg, 0.010 mg, 0.015 mg, 0.020 mg, 0.025 mg, 0.050 mg, 0.075 mg, 0.1 mg, 0.15 mg, 0.2 mg, 0.25 mg, 0.5 mg, 0.75 mg, or 1 mg/kg body weight per day. The dose of the pharmaceutically active agent(s) described herein for the described methods can be about 1 to about 1000 mg/kg body weight of the subject being treated per day, for example, about 1 mg, 2 mg, 5 mg, 10 mg, 15 mg, 0.020 mg, 25 mg, 50 mg, 75 mg, 100 mg, 150 mg, 200 mg, 250 mg, 500 mg, 750 mg, or 1000 mg/kg body weight per day.
In accordance with embodiments, the present invention provides methods of treating, preventing, ameliorating, and/or inhibiting a hepatitis C virus infection comprising administering a compound of formula (I) or a salt thereof.
It should also be understood that the compounds of the present invention can inhibit multiple genotypes of HCV. In one of the embodiment compound of the present invention are active against the 1 a, 1 b, 2a, 2b, 3a, 4a and 5a genotypes. The terms "treat," "prevent," "ameliorate," and "inhibit," as well as words stemming therefrom, as used herein, do not necessarily imply 100% or complete treatment, prevention, amelioration, or inhibition. Rather, there are varying degrees of treatment, prevention, amelioration, and inhibition of which one of ordinary skill in the art recognizes as having a potential benefit or therapeutic effect. In this respect, the inventive methods can provide any amount of any level of treatment, prevention, amelioration, or inhibition of the disorder in a mammal. For example, a disorder, including symptoms or conditions thereof, may be reduced by, for example, 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, or 10%. Furthermore, the treatment, prevention, amelioration, or inhibition provided by the inventive method can include treatment, prevention, amelioration, or inhibition of one or more conditions or symptoms of the disorder, e.g., cancer. Also, for purposes herein, "treatment," "prevention," "amelioration," or "inhibition" can encompass delaying the onset of the disorder, or a symptom or condition thereof. In accordance with the invention, the term subject includes an "animal" which in turn includes a mammal such as, without limitation, the order Rodentia, such as mice, and the order Lagomorpha, such as rabbits. It is preferred that the mammals are from the order Carnivora, including Felines (cats) and Canines (dogs). It is more preferred that the mammals are from the order Artiodactyla, including Bovines (cows) and Swine (pigs) or of the order Perssodactyla, including Equines (horses). It is most preferred that the mammals are of the order Primates, Ceboids, or Simoids (monkeys) or of the order Anthropoids (humans and apes). An especially preferred mammal is the human.
The term "viral infection" refers to the introduction of a virus into cells or tissues, e.g., hepatitis C virus (HCV). In general, the introduction of a virus is also associated with replication. Viral infection may be determined by measuring virus antibody titer in samples of a biological fluid, such as blood, using, e.g., enzyme immunoassay. Other suitable diagnostic methods include molecular based techniques, such as RT-PCR, direct hybrid capture assay, nucleic acid sequence based amplification, and the like. A virus may infect an organ, e.g., liver, and cause disease, e.g., hepatitis, cirrhosis, chronic liver disease and hepatocellular carcinoma.
The term "immune modulator" refers to any substance meant to alter the working of the humoral or cellular immune system of a subject. Such immune modulators include inhibitors of mast cell-mediated inflammation, interferons, interleukins, prostaglandins, steroids, cortico-steroids, colony-stimulating factors, chemotactic factors, etc.
Following are the abbreviations used and meaning thereof in the specification:
ACN: Acetonitrile. Boc: tert-butyloxycarbonyl. Cbz: Carbobenzyloxy. CDCI3: Deuterochloroform. DCM: dichloromethane. DDQ: 2,3-Dichloro-5,6-dicyano-p- benzoquinone. DIPEA: N, N-diisopropylethylamine. DME: 1 , 2-dimethoxyethane. DMF: N, N-dimethylformamide. DMSO-c6: Dimethyl sulfoxide-d6. EDCI: A/-(3-Dimethylaminopropyl)- /V-ethylcarbodiimide. EtOAc: Ethyl acetate. HATU: 0-(7-Azabenzotriazol-1 -yl)-A/,A/,A/',/V- tetramethyluronium hexafluorophosphate. HCI: hydrochloric acid. HOBt: 1 - Hydroxybenzotriazole hydrate. HPLC - High Performance Liquid Chromatography. KOAc: potassium acetate. MeOD: Methanol-d4. Na2S04: sodium sulphate. NaHC03: sodium bicarbonate. NCS: N-chlorosuccinimide. PCy3: Tricyclohexylphosphine. Pd(PPh3)4: Tetrakis(triphenylphosphine)palladium(0). Pd-C: Palladium on charcoal. PdCI2(dppf): [1 ,1 '- Bis(diphenylphosphino)ferrocene]dichloropalladium(ll). PdCl2(dppf)-CH2CI2: [1 ,1 '- Bis(diphenylphosphino)ferrocene]dichloropalladium(ll), complex with dichloromethane. pet-ether: petroleum ether. TFA: trifluoroacetic acid. THF: Tetrahydrofuran
The following examples are provided to further illustrate the present invention and therefore should not be construed in any way to limit the scope of the present invention. All 1 HNMR spectra were determined in the solvents indicated and chemical shifts are reported in δ units downfield from the internal standard tetramethylsilane (TMS) and interproton coupling constants are reported in Hertz (Hz). In case of mixture of the isomers, the peak values given are for the dominant isomer (rotamer/tautomer).
Example 1 : Preparation of methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (mixture of trans isomers) (Compound 1):
Step 1 : 2-(4-bromophenyl)-2-oxoethyl-2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylate (trans isomers) (1 a):
Figure imgf000042_0001
HN-Boc
To a stirred solution of frans-2-((tert-butoxycarbonyl)amino)cyclopentanecarboxylic acid (0.8 g, 3.49 mmol) [synthesized as described in J. Org. Chem., 2001 , 66 (16), 5629-5632] and 2-bromo-1 -(4-bromophenyl)ethanone (0.97 g, 3.49 mmol) in acetonitrile (50 mL) DIPEA (0.61 mL, 3.49 mmol) was added and the reaction was stirred at rt for 2 h. The contents were concentrated under reduced pressure and water (100 mL) was added and extracted with ethyl acetate (2 χ 50mL). The combined organic layers were washed with brine, dried over anhydrous Na2S04 and concentrated under reduced pressure to get pale yellow colored oily liquid (1 .3 g, 87 %) that was used in the next step without further purification. 1 H-NMR (400 MHz, CDCI3), δ 7.81 -7.79 (m, 2H), 7.67-7.65 (m, 2H), 5.61 (bs, 1 H), 5.42-5.20 (m, 2H), 4.30-4.22 (m, 1 H), 3.19-3.14 (m, 1 H), 2.15-1 .65 (m, 6H), 1 .47 (s, 9H); m/z 426.30 (M++1 ).
Step 2: tert-butyl (2-(5-(4-bromophenyl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (trans isomers)(1 b):
Figure imgf000043_0001
To the stirred solution of 2-(4-bromophenyl)-2-oxoethyl-2-((tert-butoxycarbonyl)amino) cyclopentanecarboxylate (1 .3 g, 3.05 mmol) from the previous step in toluene (20 ml_), ammonium acetate (4.70 g, 61 .0 mmol) was added and the contents were heated at 1 10 eC for 15 h. The reaction mixture was poured into 100 mL water and extracted with ethyl acetate (2 χ 50 mL). The combined organic layers were washed with brine, dried over anhydrous Na2S04 and purified by flash column chromatography (45% EtOAc/hexane) to get orange colored solid (1 .0 g, 81 %). 1 H-NMR (400 MHz, CDCI3), δ 7.57-7.47 (m, 4H), 7.21 (s, 1 H), 5.14-4.95 (m, 1 H), 4.35-4.15 (m, 1 H), 3.39-3.15 (m, 1 H), 2.14-1 .65 (m, 6H), 1 .43 (s, 9H); m/z 406.1 1 (M++1 ).
Step 3: tert-butyl (2-(5-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)carbamate (trans isomers) (1 c):
Figure imgf000043_0002
A mixture of tert-butyl (2-(5-(4-bromophenyl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (1 g, 2.46 mmol) from the previous step, potassium acetate (0.68 g, 6.90 mmol) and 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1 ,3,2-dioxaborolane) (1 .3 g, 4.92 mmol) in dioxane (10 mL) was de-gassed with nitrogen for 30 min after which Pd(PPh3)4 (0.08 g, 0.07 mmol) was added and the reaction mixture was irradiated with microwaves at 120 eC for 40 min. The contents were taken up in EtOAc (20 mL) and passed through a celite bed. The celite bed was thoroughly rinsed with EtOAc (20 mL). The combined organic layers were washed with brine, dried over sodium sulphate and purified by flash chromatography (50% ethylacetate/hexane) to yield a pale yellow coloured solid (0.7 g, 62%). 1 H-NMR (400 MHz, CDCI3), δ 7.79-7.77 (d, J = 8.0 Hz, 2H), 7.61 -7.59 (d, J = 8.0 Hz, 2H), 7.32 (s, 1 H), 5.20-5.10 (m, 1 H), 4.30-4.25 (m, 1 H), 3.39-3.30 (m, 1 H), 2.14-1 .60 (m, 6H), 1 .36 (s, 12H), 1 .30 (s, 9H); m/z 454.28 (M++1 ).
Step 4: (2S)-tert-butyl 2-(7-(4-(2-(2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H-imidazol- 5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (trans isomers) (1 d):
Figure imgf000044_0001
The solution of (S)-tert-butyl 2-(7-bromo-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate (0.2 g, 0.48 mmol) [synthesized as reported in ref. WO2009/102633] and tert-butyl (2-(5-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (0.26 g, 0.57 mmol) [from the previous step] in a mixture of DME-H20 (3:1 , 4 mL) was degassed using nitrogen gas for 15 min. Potassium carbonate (0.33 g, 2.40 mmol) and Pd(PPh3)4 (0.055 g, 0.048 mmol) were added and irradiated using microwaves at 120 SC for 40 min. The contents were taken up in ethyl acetate and washed with water. The organic layer was dried over Na2S04 and concentrated under reduced pressure to obtain the crude compound which was purified by flash chromatography (80% EtOAc/hexane) to get a pale yellow solid (0.16 g, 50%). 1 H- NMR (400 MHz, CDCI3), δ 7.85-7.40 (m, 7H), 7.12-6.90 (m, 1 H), 5.07-4.86 (m, 2H), 4.40- 4.20 (m, 2H), 3.50-3.33 (m, 3H), 3.15-2.80 (m, 4H), 2.45-1 .80 (m, 8H), 1 .60-1.51 (m, 18H); m/z 665.40 (M++1 ). Step 5: methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (mixture of trans isomers) (Compound 1 ):
Figure imgf000045_0001
(2S)-tert-butyl 2-(7-(4-(2-(2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H-imidazol-5- yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (0.1 g, 0.15 mmol) [from previous step] was dissolved in DCM (4 mL) and TFA (0.2 mL) was added to the reaction mixture at 5-10 eC. After completion of addition, the mixture was stirred at rt for 2 h. The mixture was evaporated to dryness and triturated with diethyl ether to obtain the product as a TFA salt which was used in the next step without any further purification. (S)-2-((methoxycarbonyl)amino)-3-methylbutanoic acid (0.05 g, 0.30 mmol) [synthesized as reported in ref. WO2010/132538 A1 ] was added to acetonitrile (10 mL) followed by EDCI hydrochloride (0.06 g, 0.30 mmol), HOBt (0.05 g, 0.30 mmol) and DIPEA (0.18 mL, 0.9 mmol) and stirred for 20 min. Subsequently crude TFA salt from previous step (0.070 g) was added and stirred overnight at rt. The reaction mixture was evaporated to dryness and the required amide was precipitated out by adding water (20 mL) and filtered. The pale yellow solid was purified by preparative HPLC (0.023 g, 20 %). 1 H NMR (DMSO-Gfe, 400 MHz), δ 12.10-1 1 .55 (m, 2H), 7.82-7.29 (m, 9H), 7.05-6.60 (m, 2H), 5.10- 5.05 (m, 1 H), 4.40-4.32 (m, 2H), 4.1 1 -4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51 -3.45 (s, 6H), 3.02-2.67 (m, 4H), 2.25-1 .65 (m, 10H), 0.99-0.51 (m, 12H); m/z 779.50 (M++1 ). m.p.: 148- 150 eC. Example 2: methyl ((2S)-1-((2-(5-(4-(2-((S)-1-((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate (mixture of cis isomers) (Compound 2):
Step 1 : 2-(4-bromophenyl)-2-oxoethyl-2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylate (cis isomers) (2a):
Figure imgf000046_0001
Title compound was synthesized by following the procedure as described in Step 1 of Example 1 using c/'s-2-((tert-butoxycarbonyl)amino)cyclopentanecarboxylic acid [synthesized as described in Tetrahedron: Asymmetry, 2008, 19, 2796-2803]. 1 H-NMR (400 MHz, CDCI3), δ 7.85-7.79 (m, 2H), 7.70-7.65 (m, 2H), 5.65 (bs, 1 H), 5.35-5.20 (m, 2H), 4.35-4.22 (m, 1 H), 3.25-3.14 (m, 1 H), 2.20-1.65 (m, 6H), 1 .47 (s, 9H); m/z 426.30 (M++1 ).
Step 2: tert-butyl (2-(5-(4-bromophenyl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (cis isomers) (2b):
Figure imgf000046_0002
Title compound was synthesized by following the procedure as described in Step 2 of Example 1 using intermediate (2a) from the previous step. 1 H-NMR (400 MHz, CDCI3), δ 7.60-7.45 (m, 4H), 7.23 (s, 1 H), 5.20-4.95 (m, 1 H), 4.37-4.15 (m, 1 H), 3.40-3.17 (m, 1 H), 2.20-1 .70 (m, 6H), 1 .50 (s, 9H); m/z 406.1 1 (M++1 ). Step 3: tert-butyl (2-(5-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)carbamate (cis isomers) (2c):
Figure imgf000047_0001
Title compound was synthesized by following the procedure as described in Step 3 of Example 1 using intermediate (2b) from the previous step. 1 H-NMR (400 MHz, CDCI3), δ 7.79-7.77 (m, 2H), 7.70-7.67 (m, 2H), 7.30 (s, 1 H), 5.20-5.10 (m, 1 H), 4.30-4.25 (m, 1 H), 3.39-3.32 (m, 1 H), 2.14-1 .60 (m, 6H), 1 .36 (s, 12H), 1 .30 (s, 9H); m/z 454.20 (M++1 ).
Step 4: (2S)-tert-butyl 2-(8-(4-(2-(2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H-imidazol- 5-yl)phenyl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-2-yl)pyrrolidine-1 - carboxylate (2d):
Figure imgf000047_0002
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using intermediate (2c) from the previous step and (S)-tert-butyl 2-(8-bromo- 3,4,5, 6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate
[synthesized as described in WO2009/102633]. 1 H-NMR (400 MHz, CDCI3), δ 7.83-7.45 (m, 8H), 5.07-4.86 (m, 2H), 4.40-4.20 (m, 2H), 3.50-3.43 (m, 3H), 3.10-2.80 (m, 4H), 2.45- 1 .80 (m, 10H), 1 .60-1 .51 (m, 18H), m/z 679.40 (M++1 ).
Step 5: methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (mixture of cis isomers): (Compound 2)
Figure imgf000048_0001
Title compound was synthesized by following the procedure as described in Step 5 of Example 1 using intermediate (2d) from the previous step. 1 H NMR (DMSO-c6, 400 MHz), δ 1 1 .88-1 1 .55 (m, 2H), 8.15-8.00 (m, 2H), 7.82-7.29 (m, 9H), 5.15-5.02 (m, 1 H), 4.40-4.32 (m, 2H), 4.1 1 -4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51 (s, 6H), 2.94-2.80 (m, 4H), 2.14-1 .65 (m, 10H), 0.97-0.59 (m, 14H); m/z 793.50 (M++1 ). m.p.: 155-157 eC.
Example 3: Preparation of methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (mixture of cis isomers) (Compound 3):
Step 1 : (2S)-tert-butyl 2-(7-(4-(2-(2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H-imidazol- 5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (cis isomers) (3a):
Figure imgf000048_0002
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 , using intermediate 2c from Example 2, Step 3 and (S)-tert-butyl 2-(7-bromo- 4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate [synthesized as described in WO2009/102633]. 1 H-NMR (400 MHz, CDCI3), δ 7.85-7.40 (m, 7H), 7.12-6.90 (m, 1 H), 5.07-4.86 (m, 2H), 4.40-4.20 (m, 2H), 3.50-3.33 (m, 3H), 3.15-2.80 (m, 4H), 2.45- 1 .80 (m, 8H), 1 .60-1 .51 (m, 18H); m/z 665.40 (M++1 ). Step 2: methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (mixture of cis isomers): (Compound 3)
Figure imgf000049_0001
Title compound was synthesized by following the procedure as described in Step 5 of Example 1 using intermediate (3a) from the previous step. 1 H NMR (DMSO-c6, 400 MHz), δ 1 1 .88-1 1 .55 (m, 2H), 7.88-7.28 (m, 9 H), 7.02-6.87 (m, 2H), 5.15-5.02 (m, 1 H), 4.40-4.32 (m, 2H), 4.1 1 -4.07 (m, 2H), 3.87-3.81 (m, 2H), 3.51 -3.45 (m, 6H), 3.02-2.67 (m, 4H), 2.20- 1 .55 (m, 12H), 1 .10-0.55 (m, 12H); m/z 779.50 (M++1 ). m.p.: 140 eC.
Example 4: Preparation of methyl ((S)-1-(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (Compound 4):
Step 1 : (1 S,2R)-2-(4-bromophenyl)-2-oxoethyl-2-((tertbutoxycarbonyl)amino) cyclopentanecarboxylate (4a):
Figure imgf000049_0002
Title compound was synthesized by following the procedure as described in Step 1 of Example 1 using 2-bromo-1 -(4-bromophenyl)ethanone and (1 S,2R)-2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylic acid [synthesized as described in Tetrahedron: Asymmetry, 2008, 19, 2796-2803] as starting materials. 1 H-NMR (400 MHz, CDCI3) δ 7.79 (d, J = 8 Hz, 2H), 7.66 (d, J = 8 Hz, 2H), 5.60 (bs, 1 H), 5.40-5.20 (m, 2H), 4.35-4.30 (m, 1 H), 3.20-3.15 (m, 1 H), 2.20-1 .60 (m, 6H), 1 .40 (s, 9H). LC-MS: m/z 326.0 [(M++1 ) - 100] (value observed for the de-Boc product).
Step 2: tert-butyl ((1 R,2S)-2-(4-(4-bromophenyl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (4b):
Figure imgf000050_0001
Title compound was synthesized by following the procedure as described in Step 2 of Example 1 using the intermediate (4a) from the previous step. 1 H-NMR (400 MHz, CDCI3), δ 7.60-7.56 (m, 2H), 7.49-7.45 (m, 2H), 7.21 (s, 1 H), 5.0-4.95 (m, 1 H), 4.35-4.30 (m, 1 H), 3.40-3.35 (m, 1 H), 2.40-1 .60 (m, 6H), 1 .48/1 .33 (s, 9H); m/z 406.10 (M++1 ).
Step 3: tert-butyl ((1 R,2S)-2-(5-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)carbamate (4c):
Figure imgf000050_0002
Title compound was synthesized by following the procedure as described in Step 3 of Example 1 using the intermediate (4b) from the previous step. 1 H-NMR (400 MHz, CDCI3), δ 7.79-7.77 (m, 2H), 7.70-7.67 (m, 2H), 7.30 (s, 1 H), 5.20-5.10 (m, 1 H), 4.30-4.25 (m, 1 H), 3.39-3.32 (m, 1 H), 2.14-1 .60 (m, 6H), 1 .36 (s, 12H), 1 .30 (s, 9H); m/z 454.20 (M++1 ).
Step 4: (S)-tert-butyl 2-(7-(4-(2-((1 S,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (4d):
Figure imgf000050_0003
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediate 4c and and (S)-tert-butyl 2-(7-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate [synthesized as described in WO2009102633]. 1 H-NMR (400 MHz, CDCI3) δ 7.83-7.45 (m, 7H), 7.20-7.10 (m, 1 H), 5.07-4.86 (m, 2H), 4.40-4.20 (m, 2H), 3.50-3.43 (m, 3H), 3.15-3.05 (m, 2H), 2.97-2.80 (m, 2H), 2.45-1 .80 (m, 8H), 1 .60-1 .51 (m, 18H), m/z 665.40 (M++1 ).
Step 5: methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 4).
Figure imgf000051_0001
Title compound was synthesized by following the procedure as described in Step 5 of Example 1 using the intermediate (4d) from previous step. 1 H NMR (DMSO-afe, 400 MHz), δ 12.10-1 1 .55 (m, 2H), 7.82-7.29 (m, 9H), 7.05-6.60 (m, 2H), 5.10-5.05 (m, 1 H), 4.40-4.32 (m, 2H), 4.1 1 -4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51 -3.45 (s, 6H), 3.02-2.67 (m, 4H), 2.20- 1 .65 (m, 10H), 0.99-0.51 (m, 12H), m/z 779.50 (M++1 ). m.p.: 162-165 eC.
Example 5: Preparation of methyl ((S)-1-(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 , 4,5,6- tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Example 5):
Step 1 : (S)-tert-butyl 2-(8-(4-(2-((1 S,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate (5a):
Figure imgf000052_0001
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediates from Step 2 of Example 4 (4b) and (S)-tert-butyl 2-(8- (4,4,5, 5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2- d]imidazol-2-yl)pyrrolidine-1 -carboxylate [synthesized as described in WO2009102633]. 1 H-NMR (400 MHz, CDCI3) δ 7.83-7.45 (m, 8H), 5.07-4.86 (m, 2H), 4.40-4.20 (m, 2H), 3.50-3.43 (m, 3H), 3.10-2.80 (m, 4H), 2.45-1 .80 (m, 10H), 1 .60-1 .51 (m, 18H), m/z 679.40 (M++1 ).
Step 2: methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 5):
Figure imgf000052_0002
Title compound was synthesized by following the procedure as described in Step 5 of Example 1 using intermediate (5a) from the previous step. 1 H NMR (DMSO-c6, 400 MHz), δ 1 1 .88-1 1 .55 (m, 2H), 8.04-7.29 (m, 1 1 H), 5.15-5.02 (m, 1 H), 4.40-4.32 (m, 2H), 4.1 1 - 4.07 (m, 2H), 3.87-3.70 (m, 4H), 3.51 -3.45 (m, 6H), 2.94-2.80 (m, 4H), 2.20-1.65 (m, 12H), 0.97-0.71 (m, 12H), m/z 793.50 (M++1 ). m.p. : 165-167 eC. Example 6: Preparation of methyl ((S)-1-(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- benzo[2,3]oxepino[4,5-d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)- 3-methyl-1-oxobutan-2-yl)carbamate (Compound 6):
Step 1 : (S)-tert-butyl 2-(8-(4-(2-((1 S,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-2-yl)pyrrol^ carboxylate (6a):
Figure imgf000053_0001
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediates from Step 2 of Example 4 (4b) and (S)-tert-butyl 2-(8- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5- d]imidazol-2-yl)pyrrolidine-1 -carboxylate [synthesized using procedures described in WO2009102633]. 1 H-NMR (400 MHz, CDCI3) δ 1 1 .00 (bs, 1 H), 10.30 (bs, 1 H), 7.83-7.40 (m, 8H), 5.07-4.90 (m, 2H), 4.40-4.20 (m, 3H), 3.50-3.43 (m, 3H), 3.25-3.10 (m, 2H), 2.95- 2.90 (m, 1 H), 2.45-1 .72 (m, 8H), 1 .65-1 .51 (m, 18H), m/z 681 .40 (M++1 ).
Step 2: methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 6):
Figure imgf000053_0002
Title compound was synthesized by following the procedure as described in Step 5 of Example 1 using HATU, DMF and the intermediate (6a) from previous step. 1 H NMR (DMSO-Gfe, 400 MHz), δ 12.00-1 1 .70 (m, 2H), 8.14-6.90 (m, 1 1 H), 5.05-5.02 (m, 1 H), 4.40- 4.10 (m, 4H), 3.90-3.70 (m, 4H), 3.60 (s, 6H), 3.15-3.00 (m, 2H), 2.14-1.55 (m, 12H), 0.97- 0.71 (m, 12H), m/z 795.50 (M++1 ). m.p. : 191 eC.
Example 7: methyl ((S)-1-(((1 S,2R)-2-(5-(4-(2-((S)-1-((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 7):
Step 1 : (1 R,2S)-2-(4-bromophenyl)-2-oxoethyl-2-((tertbutoxycarbonyl) amino)cyclopentanecarboxylate (7a):
Figure imgf000054_0001
Title compound was synthesized by following the procedure as described in Step 1 of Example 1 using 2-bromo-1 -(4-bromophenyl)ethanone and (1 R,2S)-2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylic acid [synthesized by following the procedures described in Tetrahedron: Asymmetry, 2008, 19, 2796-2803]. 1 H-NMR (400 MHz, CDCI3) δ 7.80 (d, J = 8 Hz, 2H), 7.67 (d, J = 8 Hz, 2H), 5.6 (bs, 1 H), 5.40-5.20 (m, 2H), 4.35-4.30 (m, 1 H), 3.19-3.15 (m, 1 H), 2.20-1 .60 (m, 6H), 1 .40 (s, 9H); m/z 326.2 [(M++1 ) - 100] (value observed for the de-Boc product).
Step 2: tert-butyl- ((1 S,2R)-2-(4-(4-bromophenyl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (7b):
Figure imgf000054_0002
Title compound was synthesized by following the procedure as described in Step 2 of Example 1 , using the intermediate (7a) from the previous step. 1 H-NMR (400 MHz, CDCI3) δ 7.60-7.56 (m, 2H), 7.49-7.47 (m, 2H), 7.20 (s, 1 H), 5.05-4.95 (m, 1 H), 4.40-4.31 (m, 1 H), 3.40-3.35 (m, 1 H), 2.40-1 .60 (m, 6H), 1 .33/1 .26 (s, 9H); m/z 406.0 (M++1 ).
Step 3: tert-butyl ((1 S,2R)-2-(5-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)carbamate (7c):
Figure imgf000055_0001
Title compound was synthesized by following the procedure as described in Step 3 of Example 1 using the intermediate (7b) from the previous step. 1 H-NMR (400 MHz, CDCI3) δ 7.85-7.78 (m, 2H), 7.65-7.60 (m, 2H), 7.30 (s, 1 H), 5.15-5.10 (m, 1 H), 4.35-4.25 (m, 1 H), 3.45-3.30 (m, 1 H), 2.19-1 .60 (m, 6H), 1 .40 (s, 12H), 1 .35 (s, 9H); m/z 454.28 (M++1 ).
Step 4: (S)-tert-butyl 2-(8-(4-(2-((1 R,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-2-yl)pyrrolidine-1 - carboxylate (7d):
Figure imgf000055_0002
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediate 7c and (S)-tert-butyl 2-(8-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-2-yl)pyrrolidine-1 - carboxylate [synthesized as described in WO2009102633]. 1 H-NMR (400 MHz, CDCI3) δ 7.78-7.48 (m, 8H), 5.07-4.86 (m, 2H), 4.40-4.20 (m, 2H), 3.70-3.43 (m, 3H), 3.05-2.90 (m, 4H), 2.45-1 .90 (m, 10H), 1 .50-1 .31 (m, 18H), m/z 681 .40 (M++1 ). Step 5: methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 7):
Figure imgf000056_0001
Title compound was synthesized by following the procedure as described in Step 2 of Example 6 using the intermediate (7d) from the previous step. 1 H NMR (DMSO-c6, 400 MHz), δ 12.10-1 1 .80 (m, 2H), 8.14-6.90 (m, 1 1 H), 5.05-5.02 (m, 1 H), 4.40-4.10 (m, 4H), 3.90-3.70 (m, 4H), 3.60 (s, 6H), 3.15-3.00 (m, 2H), 2.14-1 .95 (m, 12H), 0.97-0.71 (m, 12H), m/z 795.30 (M++1 ). m.p.: 218 eC.
Example 8: Preparation of methyl ((S)-1-(((1S,2R)-2-(5-(4-(2-((S)-1 -((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 , 4,5,6- tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 8):
Step 1 : (S)-tert-butyl 2-(8-(4-(2-((1 R,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate (8a):
Figure imgf000056_0002
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediates from Step 2 Example 7 (7b) and (S)-tert-butyl 2-(8- (4,4,5, 5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2- d]imidazol-2-yl)pyrrolidine-1 -carboxylate [synthesized as described in WO2009102633]. 1H-NMR (400 MHz, CDCI3) δ 11.00 (bs, 1H), 10.20 (bs, 1H), 7.83-7.45 (m, 8H), 5.07-4.86 (m, 2H), 4.40-4.20 (m, 2H), 3.50-3.43 (m, 3H), 3.10-2.80 (m, 4H), 2.45-1.80 (m, 10H), 1.60-1.51 (m, 18H), m/z 679.40 (M++1).
Step 2: methyl ((S)-1-(((1S,2R)-2-(5-(4-(2-((S)-1-((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8- yl)phenyl)-1H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 8):
Figure imgf000057_0001
Title compound was synthesized by following the procedure as described in Step 5 of Example 1 using the intermediate (8a) from the previous step. 1H NMR (DMSO-c6, 400 MHz), δ 11.88-11.55 (m, 2H), 8.31-6.99 (m, 11 H), 5.15-5.02 (m, 1H), 4.40-4.32 (m, 2H), 4.11-4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51-3.45 (s, 6H), 2.94-2.80 (m , 4H), 2.10-1.40 (m, 12H), 0.95-0.54 (m, 12H), m/z 793.50 (M++1). m.p.: 162-164 eC.
Example 9: Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1, 4,5,6- tetrahydrobenzo[3,4]cyclohepta[1,2-d]imidazol-8-yl)phenyl)-1H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 9):
Step 1 : (1S,2S)-2-(4-bromophenyl)-2-oxoethyl-2-((tertbutoxycarbonyl)amino) cyclopentanecarboxylate (9a):
Figure imgf000057_0002
Title compound was synthesized by following the procedure as described in Step 1 of Example 1 using 2-bromo-1 -(4-bromophenyl)ethanone and (1 S,2S)-2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylic acid [synthesized as described in J. Org. Chem., 2001 , 66 (16), 5629-5632]. 1 H NMR (DMSO-afe, 400 MHz), δ 7.90-7.87 (m, 2H), 7.78-7.76 (m, 2H), 7.06-7.03 (m, 1 H), 5.45-5.30 (m, 2H), 4.05-3.95 (m, 1 H), 2.80-2.75 (m, 1 H), 1 .90-1 .40 (m, 6H), 1 .35 (s, 9H); m/z 326.0 [(M++1 ) - 100] (value observed for the de- Boc product). tert-butyl ((1 S,2S)-2-(4-(4-bromophenyl)-1 H-imidazol-2-yl)cyclopentyl)carbamate
Figure imgf000058_0001
Title compound was synthesized by following the procedure as described in Step 2 of Example 1 using the intermediate (9a) from the previous step. 1 H-NMR (DMSO-afe, 400 MHz), δ 7.68 (d, J = 8.0 Hz, 2H), 7.57-7.54 (m, 1 H), 7.48 (d, J = 8.0 Hz, 2H), 6.95 (d, J = 8.0 Hz, 1 H), 4.05-3.90 (m, 1 H), 3.10-3.02 (m, 1 H), 2.10-1 .40 (m, 6H), 1 .35 (s, 9H); m/z 406.0 (M++1 ).
Step 3: tert-butyl ((1 S,2S)-2-(5-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)carbamate (9c):
Figure imgf000058_0002
Title compound was synthesized by following the procedure as described in Step 3 of Example 1 using the intermediate (9b) from the previous step. 1 H-NMR (400 MHz, CDCI3), δ 7.85-7.78 (m, 2H), 7.65-7.60 (m, 2H), 7.30 (s, 1 H), 5.30-5.10 (m, 1 H), 4.30-4.20 (m, 1 H), 3.45-3.30 (m, 1 H), 2.25-1 .60 (m, 6H), 1 .40 (s, 12H), 1 .35 (s, 9H); m/z 454.28 (M++1 ). Step 4: (S)-tert-butyl 2-(8-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate (9d):
Figure imgf000059_0001
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediates 9c and (S)-tert-butyl 2-(8-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-2-yl)pyrrolidine- 1 -carboxylate [synthesized as described in WO2009102633]. 1 H-NMR (400 MHz, CDCI3) δ 1 1 .00 (bs, 1 H), 10.50 (bs, 1 H), 7.83-7.45 (m, 8H), 4.95-4.86 (m, 2H), 4.25-4.15 (m, 2H), 3.50-3.43 (m, 3H), 3.20-2.80 (m, 4H), 2.45-1 .80 (m, 10H), 1 .60-1 .51 (m, 18H), m/z 679.40 (M++1 ).
Step 5: methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 9):
Figure imgf000059_0002
Title compound was synthesized by following the procedure as described in Step 5 of Example 1 using the intermediate (9d) from the previous step. 1 H NMR (DMSO-afe, 400 MHz), δ 1 1 .88-1 1 .55 (m, 2H), 8.31 -6.99 (m, 1 1 H), 5.15-5.02 (m, 1 H), 4.40-4.32 (m, 2H), 4.1 1 -4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51 -3.45 (s, 6H), 2.94-2.80 (m , 4H), 2.14-1 .50 (m, 12H), 0.97-0.70 (m, 12H), m/z 793.50 (M++1 ). m.p.: 147 eC. Example 10: Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (Compound 10):
Step 1 : (S)-tert-butyl 2-(7-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (10a)
Figure imgf000060_0001
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediates from Step 2 of Example 9 (9b) and (S)-tert-butyl 2-(7- (4,4,5, 5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate [synthesized as described in WO2009102633]. m/z 665.3 (M++1 ).
Step 2: methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 10):
Figure imgf000060_0002
Title compound was synthesized by following the procedure as described in Step 5 of Example 1 using the intermediate (10a) from the previous step. 1 H NMR (DMSO-afe, 400 MHz), δ 12.10-1 1 .55 (m, 2H), 8.08-6.96 (m, 1 1 H), 5.10-5.05 (m, 1 H), 4.40-4.32 (m, 2H), 4.1 1 -4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51 -3.45 (s, 6H), 3.02-2.67 (m, 4H), 2.20-1 .45 (m, 10H), 0.97-0.68 (m, 12H), m/z 779.50 (M++1 ). m.p.: 181 eC. Example 11 : Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- benzo[2,3]oxepino[4,5-d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)- 3-methyl-1-oxobutan-2-yl)carbamate (Compound 11 ):
Step 1 : (S)-tert-butyl 2-(8-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-ben^
carboxylate (1 1 a):
Figure imgf000061_0001
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediates from Step 2 of Example 9 (9b) and (S)-tert-butyl 2-(8- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5- d]imidazol-2-yl)pyrrolidine-1 -carboxylate [synthesized as described in WO2009102633]. 1 H-NMR (400 MHz, CDCI3) δ 1 1.00 (bs, 1 H), 10.30 (bs, 1 H), 7.90-7.30 (m, 8H), 5.07-4.90 (m, 2H), 4.40-4.10 (m, 3H), 3.50-3.43 (m, 2H), 3.25-3.10 (m , 3H), 2.95-2.90 (m, 1 H), 2.45- 1 .72 (m, 8H), 1 .65-1 .51 (m, 18H), m/z 681 .40 (M++1 ).
Step 2: methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 1 1 ):
Figure imgf000061_0002
Title compound was synthesized by following the procedure as described in Step 2 of Example 6 using the intermediate (1 1 a) from the previous step. 1 H NMR (DMSO-afe, 400 MHz), δ 12.10-1 1 .80 (m, 2H), 8.14-7.00 (m, 1 1 H), 5.05-5.02 (m, 1 H), 4.40-4.10 (m, 5H), 3.90-3.70 (m, 3H), 3.55-3.50 (m, 6H), 3.15-3.10 (m, 2H), 2.14-1 .95 (m, 12H), 0.97-0.71 (m, 12H), m/z 795.40 (M++1 ). m.p.: 172-174 eC.
Example 12: Preparation of methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 , 4,5,6- tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 12):
Step 1 : (1 R, 2R)-2-(4-bromophenyl)-2-oxoethyl-2-((tert-butoxycarbonyl)amino) cyclopentane carboxylate (12a):
Figure imgf000062_0001
Title compound was synthesized by following the procedure as described in Step 1 of Example 1 using 2-bromo-1 -(4-bromophenyl)ethanone and (1 R,2R)-2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylic acid [synthesized by following the procedures provided in J. Org. Chem., 2001 , 66 (16), 5629-5632]. 1 H-NMR (400 MHz, CDCI3), δ 7.79 (d, J = 8 Hz, 2H), 7.66 (d, J = 8 Hz, 2H), 5.40-5.20 (m, 2H), 4.25-4.18 (m, 1 H), 2.85-2.80 (m, 1 H), 2.20-2.00 (m, 3H), 1 .82-1 .76 (m, 2H), 1 .60-1 .50 (m, 1 H), 1 .40 (s, 9H). m/z 326.0 [(M++1 ) - 100] (value observed for the de-Boc product).
Step 2: tert-butyl ((1 R,2R)-2-(4-(4-bromophenyl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (12b):
Figure imgf000062_0002
Title compound was synthesized by following the procedure as described in Step 2 of Example 1 using intermediate (12a) from the previous step. 1 H-NMR (400 MHz, CDCI3), δ 7.60-7.56 (m, 2H), 7.49-7.47 (m, 2H), 7.26 (s, 1 H), 5.00-4.90 (m, 1 H), 4.17-4.10 (m, 1 H), 3.17-3.12 (m, 1 H), 2.38-2.10 (m, 4H), 1 .80-1 .55 (m, 2H), 1 .50 (s, 9H); m/z 405.85 (M++1 ). Step 3: tert-butyl ((1 R,2R)-2-(5-(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)carbamate (12c):
Figure imgf000063_0001
Title compound was synthesized by following the procedure as described in Step 3 of Example 1 using intermediate (12b) from the previous step. 1 H-NMR (400 MHz, CDCI3), δ 7.85-7.78 (m, 2H), 7.70-7.60 (m, 2H), 7.35 (s, 1 H), 5.25-5.10 (m, 1 H), 4.30-4.25 (m, 1 H), 3.45-3.30 (m, 1 H), 2.19-1 .60 (m, 6H), 1 .45 (s, 12H), 1 .35 (s, 9H); m/z 454.28 (M++1 ).
Step 4: (S)-tert-butyl 2-(8-(4-(2-((1 R,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate (12d):
Figure imgf000063_0002
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediates 12c and (S)-tert-butyl 2-(8-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-2-yl)pyrrolidine- 1 -carboxylate [synthesized as described in WO2009102633]. 1 H-NMR (400 MHz, CDCI3) δ 7.83-7.30 (m, 8H), 5.07-4.86 (m, 2H), 4.25-4.15 (m, 2H), 3.50-3.43 (m, 2H), 3.20-2.90 (m, 5H), 2.45-1 .70 (m, 10H), 1 .60-1 .51 (m, 18H), m/z 679.40 (M++1 ).
Step 5: methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 12):
Figure imgf000064_0001
Title compound was synthesized by following the procedure as described in Step 5 of Example 1 using the intermediate (12d) from the previous step. 1 H NMR (DMSO-afe, 400 MHz), δ 1 1 .88-1 1 .55 (m, 2H), 8.20-8.02 (m, 2H), 7.78-7.29 (m, 8H), 7.08-7.05 (m, 1 H), 5.15-5.02 (m, 1 H), 4.40-4.32 (m, 2H), 4.1 1 -4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51 -3.45 (s, 6H), 2.97-2.94 (m ,4H), 2.14-1 .55 (m, 12H), 0.97-0.79 (m, 12H), m/z 793.50 (M++1 ). m.p.: 157-162 eC.
Example 13: Preparation of methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- benzo[2,3]oxepino[4,5-d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)- 3-methyl-1-oxobutan-2-yl)carbamate (Compound 13):
Step 1 : (S)-tert-butyl 2-(8-(4-(2-((1 R,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-2-yl)pyrrolidine-1 - carboxylate (13a):
Figure imgf000064_0002
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediates from Step 2 of Example 12 (12b) and (S)-tert-butyl 2- (8-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5- d]imidazol-2-yl)pyrrolidine-1 -carboxylate [synthesized following the procedures provided in WO2009102633]. 1 H-NMR (400 MHz, CDCI3) δ 7.83-7.40 (m, 8H), 5.07-4.90 (m, 2H), 4.40-4.20 (m, 3H), 3.50-3.43 (m, 2H), 3.25-3.10 (m, 3H), 2.95-2.90 (m, 1 H), 2.45-1 .72 (m, 8H), 1 .65-1 .51 (m, 18H), m/z 681 .40 (M++1 ). Step 2: methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5-d]imidazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 13):
Figure imgf000065_0001
Title compound was synthesized by following the procedure as described in Step 2 of Example 6 using the intermediate (13a) from the previous step. 1 H NMR (DMSO-afe, 400 MHz), δ 12.10-1 1 .80 (m, 2H), 8.14-7.00 (m, 1 1 H), 5.05-5.02 (m, 1 H), 4.40-4.10 (m, 5H), 3.90-3.70 (m, 3H), 3.55-3.50 (m, 6H), 3.15-3.10 (m, 2H), 2.14-1 .95 (m, 12H), 0.97-0.71 (m, 12H), m/z 795.40 (M++1 ). m.p.:162-165 eC.
Example 14: Preparation of methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (Compound 14):
Step 1 : (S)-tert-butyl 2-(7-(4-(2-((1 R,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (14a):
Figure imgf000065_0002
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediates from Step 2 of Example 12 (12b) and (S)-tert-butyl 2- (7-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate [synthesized following the procedures described in WO2009102633]. 1 H-NMR (400 MHz, CDCI3) δ 7.83-7.35 (m, 8H), 5.07-4.86 (m, 2H), 4.20-4.10 (m, 2H), 3.50-3.43 (m, 3H), 3.19-3.10 (m , 2H), 2.97-2.80 (m, 2H), 2.45-1 .80 (m, 8H), 1 .60-1 .51 (m, 18H), m/z 665.40 (M++1 ).
Step 2: methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 14):
Figure imgf000066_0001
Title compound was synthesized by following the procedure as described in Step 5 of Example 1 using the intermediate (14a) from the previous step. 1 H NMR (DMSO-afe, 400 MHz), δ 12.10-1 1 .55 (m, 2H), 8.20-8.14 (m, 1 H), 7.80-6.96 (m, 10H), 5.10-5.05 (m, 1 H), 4.40-4.32 (m, 2H), 4.1 1 -4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51 -3.45 (s, 6H), δ 3.02-2.67 (m, 4H), 2.20-1 .45 (m, 10H), 0.99-0.75 (m, 12H), m/z 779.50 (M++1 ). m.p.:142-145 eC.
Example 15: Preparation of methyl ((S)-1-(((1S,2R)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (Compound 15):
Step 1 : (S)-tert-butyl 2-(7-(4-(2-((1 R,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (15a):
Figure imgf000066_0002
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediates from Step 2 of Example 7 (7b) and (S)-tert-butyl 2-(7- (4,4,5, 5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2- yl)pyrrolidine-1-carboxylate [synthesized by following procedures provided in WO2009102633].1H-NMR (400 MHz, CDCI3) δ 11.00 (bs, 1H), 10.50 (bs, 1H), 7.83-7.45 (m, 7H), 7.20-7.10 (m, 1H), 5.07-4.86 (m, 2H), 4.40-4.20 (m, 2H), 3.50-3.43 (m, 3H), 3.29- 3.26 (m, 2H), 2.97-2.80 (m, 2H), 2.45-1.80 (m, 8H), 1.60-1.51 (m, 18H), m/z 665.40 (M++1).
Step 2: methyl ((S)-1-(((1S,2R)-2-(5-(4-(2-((S)-1-((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 15):
Figure imgf000067_0001
Title compound was synthesized by following the procedure as described in Step 5 of Example 1 using the intermediate (15a) from the previous step.1H NMR (DMSO-afe, 400 MHz), δ 12.10-11.55 (m, 2H), 8.08-6.96 (m, 11H), 5.10-5.05 (m, 1H), 4.40-4.32 (m, 2H), 4.11-4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51-3.45 (s, 6H), 3.02-2.67 (m ,4H), 2.14-1.50 (m, 10H), 0.99-0.54 (m, 12H), m/z 779.30 (M++1). m.p.:163 eC.
Example 16: Preparation of methyl ((S)-1-(((1 R,2R)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1H-naphtho[1,2- d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2- yl)carbamate (Compound 16):
Step 1: (S)-tert-butyl 2-(7-bromo-3H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1-carboxylate (16a):
Figure imgf000067_0002
DDQ (0.41 g, 1 .79 mmol) was added to the solution of (S)-tert-butyl 2-(7-bromo-4,5- dihydro-3H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate [synthesized by following the procedures provided in WO2009102633] (0.5 g, 1 .19 mmol) in benzene (10 mL) and stirred at 80 eC for 3 h. Benzene was evaporated under reduced pressure and the residue was purified by column chromatography (60% EtOAc/hexane) to afford the title compound (0.3 g, 60%). 1 H-NMR (400 MHz, CDCI3):-5 8.05-8.03 (m, 1 H), 7.62-7.50 (m, 4H), 5.24- 5.23 (m, 1 H), 3.61 -3.50 (m, 1 H), 3.15-3.10 (m, 1 H), 2.29-2.19 (m, 2H), 2.09-2.01 (m, 2H), 1 .54-1 .34 (m, 9H), m/z 416.09 (M++1 ).
Step 2: (S)-tert-butyl 2-(7-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-3H-naphtho[1 ,2- d]imidazol-2-yl)pyrrolidine-1 -carboxylate (16b):
Figure imgf000068_0001
In a 5 - 20 mL microwave vial, 1 ,4-Dioxane (15 mL) was purged with nitrogen for 10 min and (S)-tert-butyl 2-(7-bromo-3H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (16a) (0.4 g, 0.96 mmol), bis(pinacolato)diboron (0.49 g, 1 .92 mmol), PCy3 (0.03 g, 0.1 mmol), KOAc (0.38 g, 3.84 mmol) were added and purged with nitrogen. Finally, PdCI2(dppf)-CH2Cl2 adduct (0.08 g, 0.1 mmol) was added & purged with nitrogen and irradiated with microwaves at 1 15 eC for 45 min. The reaction mixture was poured into water and extracted with ethyl acetate. The organic layer was washed with saturated brine solution, dried over anhydrous sodium sulphate and purified by column chromatography (70% EtOAc/hexane) to afford the title compound (0.3 g, 67%). 1 H-NMR (400 MHz, CDCI3) δ 8.46 (s, 1 H), 7.97-7.95 (m, 1 H),7.71 -7.47 (m, 3H), 5.23-5.22 (br s,1 H), 3.48-3.41 (m,1 H), 3.20-3.23 (m, 1 H), 2.26-2.19 (m, 2H), 2.61 -2.41 (m, 2H), 1 .41 (s, 9H),1 .30-1 .22 (m,12H), m/z 464.26 (M++1 ). Step 3: (S)-tert-butyl 2-(7-(4-(2-((1 R,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (16c):
Figure imgf000069_0001
The title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediates (16b) from the previous step and from Step 2 of Example 12. 1 H-NMR (400 MHz, CDCI3) δ 8.15-7.32 (m, 9H), 7.20-7.10 (m, 1 H), 5.40-5.10 (m, 2H), 4.40-4.20 (m, 1 H), 3.55-3.50 (m, 2H), 3.35-3.10 (m, 2H), 2.45-1 .80 (m, 8H), 1 .60- 1 .51 (m, 18H), m/z 663.40 (M++1 ).
Step 4: methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 16):
Figure imgf000069_0002
Title compound was synthesized by following the procedure as described in Step 5 of Example 1 using the intermediate (16c) from the previous step. 1H NMR (DMSO-afe, 400 MHz), δ 13.02-1 1 .85 (m, 2H), 8.50-8.30 (m, 2H), 8.10-7.65 (m, 8H), 7.37-7.25 (m, 1 H), 7.10-6.90 (m, 1 H), 5.30-5.28 (m, 1 H), 4.40-4.32 (m, 2H), 4.1 1 -4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51 -3.45 (s, 6H), 2.20-1 .50 (m, 10H), 0.99-0.81 (m, 12H), m/z 777.30 (M++1 ). m.p.:158-161 eC.
Example 17: Preparation of methyl ((S)-1-(((1S,2R)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2- d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate (Compound 17):
Step 1 : (S)-tert-butyl 2-(7-(4-(2-((1 R,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (17a):
Figure imgf000070_0001
The title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediates from the Step 2 of Example 16 (16b) and Step 2 of Example 7 (7b). 1H-NMR (400 MHz, CDCI3) δ 7.83-7.52 (m, 10H), 5.30-5.20 (m, 2H), 4.35- 4.25 (m, 2H), 3.55-3.35 (m, 2H), 3.29-3.26 (m, 1 H), 2.60-1 .72 (m, 8H), 1 .65-1.51 (m, 18H), m/z 663.40 (M++1 ).
Step 2: methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 17):
Figure imgf000070_0002
Title compound was synthesized by following the procedure as described in Step 2 of Example 6 using the intermediate (17a) from the previous step. 1 H NMR (DMSO-afe, 400 MHz), δ 13.02-1 1 .85 (m, 2H), 8.50-8.30 (m, 2H), 8.10-7.65 (m, 8H), 7.37-7.25 (m, 1 H), 7.10-6.90 (m, 1 H), 5.30-5.28 (m, 1 H), 4.40-4.32 (m, 2H), 4.1 1 -4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51 -3.45 (s, 6H), 2.14-1 .65 (m, 10H), 0.99-0.51 (m, 12H), m/z 777.30 (M++1 ). m.p.:210 eC.
Example 18: Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2- d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate (Compound 18):
Step 1 : (S)-tert-butyl 2-(7-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (18a):
Figure imgf000071_0001
The title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediates from the Step 2 of Example 16 (16b) and from Step 2 of Example 9 (9b). 1H-NMR (400 MHz, CDCI3) δ 7.83-7.52 (m, 10H), 5.30-5.20 (m, 2H), 4.35-4.25 (m, 2H), 3.55-3.35 (m, 2H), 3.29-3.26 (m, 1 H), 2.60-1 .72 (m, 8H), 1 .65-1 .51 (m, 18H), m/z 663.40 (M++1 ).
Step 2: methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 18):
Figure imgf000071_0002
Title compound was synthesized by following the procedure as described in Step 5 of Example 1 using the intermediate (18a) from the previous step. 1 H NMR (DMSO-afe, 400 MHz), δ 13.02-1 1 .85 (m, 2H), 8.45-7.06 (m, 13H), 5.30-5.25 (m, 1 H), 4.44-4.32 (m, 2H), 4.1 1 -4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51 -3.45 (s, 6H), 3.12-3.10 (m ,1 H), 2.14-1 .50 (m, 9H), 0.99-0.51 (m, 12H), m/z 777.30 (M++1 ). m.p.:171 eC.
Example 19: Preparation of methyl ((S)-1-(((1S,2R)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2- d]imidazol-7-yl)phenyl)-4-chloro-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (Compound 19):
Step 1 : tert-butyl ((1 S,2R)-2-(5-(4-bromophenyl)-4-chloro-1 H-imidazol-2- yl)cyclopentyl)carbamate (19a):
Figure imgf000072_0001
To the stirred solution of tert-butyl ((1 S,2R)-2-(5-(4-bromophenyl)-1 H-imidazol-2- yl)cyclopentyl)carbamate (0.15 g, 0.369 mmol) [7b] in DMF (2 mL) was added NCS (0.074 g, 0.55 mmol) at room temperature and the reaction was warmed to 50 °C and stirred for 20 min. The contents were diluted by water and extracted by ethyl acetate. The organic layer was separated, dried over sodium sulphate, evaporated under reduced pressure to get crude product which was used in the next step without further purification (0.1 g, 62%). 1 H-NMR (400 MHz, CDCI3) δ 7.58-7.51 (m, 4H), 4.75-4.70 (m, 1 H), 4.40-4.30 (m, 1 H), 3.35-3.25 (m, 1 H), 2.40-1 .60 (m, 6H), 1 .50/1 .30 (s, 9H); m/z 440.10 (M++1 ).
Step 2: (S)-tert-butyl 2-(7-(4-(2-((1 R,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-4- chloro-1 H-imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate
(19b):
Figure imgf000072_0002
The title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediates (19a) from the previous step and intermediate from Step 2 of Example 16 (16b). 1 H-NMR (400 MHz, CDCI3) δ 8.20-7.52 (m, 9H), 5.40-5.30 (m, 1 H), 4.83-4.75 (m, 1 H), 4.40-4.35 (m, 2H), 3.55-3.35 (m, 2H), 3.30-3.26 (m, 1 H), 2.45-1 .72 (m, 8H), 1 .55-1 .31 (m, 18H), m/z 697.40 (M++1 ).
Step 3: methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-4-chloro-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 19).
Figure imgf000073_0001
Title compound was synthesized by following the procedure as described in Step 2 of Example 6 using the intermediate (19b) from the previous step. 1 H NMR (DMSO-afe, 400 MHz), δ 13.10-12.30 (m, 2H), 8.50-8.30 (m, 2H), 8.10-7.65 (m, 7H), 7.35-7.30 (m, 1 H), 6.82-6.80 (m, 1 H), 5.45-5.30 (m, 1 H), 4.55-4.32 (m, 2H), 4.1 1 -4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51 -3.45 (s, 6H), 2.40-1 .65 (m, 10H), 0.99-0.51 (m, 12H), m/z 81 1 .40 (M++1 ). m.p.:255-257 eC.
Example 20: Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2- d]imidazol-7-yl)phenyl)-4-chloro-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (Compound 20):
Step 1 : tert-butyl ((1 S,2S)-2-(5-(4-bromophenyl)-4-chloro-1 H-imidazol-2- yl)cyclopentyl)carbamate (20a):
Figure imgf000073_0002
To the stirred solution of tert-butyl ((1 S,2S)-2-(5-(4-bromophenyl)-1 H-imidazol-2- yl)cyclopentyl)carbamate (0.15 g, 0.369 mmol) (9b) in DMF (2 mL) was added NCS (0.074 g, 0.55 mmol) at room temperature and the reaction was warmed to 50 °C and stirred for 20 min. The contents were diluted by water and extracted by ethyl acetate. The organic layer was separated, dried over sodium sulphate, evaporated under reduced pressure to get crude product which was used in the next step without further purification (0.1 g, 62%). 1 H-NMR (400 MHz, CDCI3) δ 12.07 (bs, 1 H), 7.69-7.67 (m 2H), 7.56-7.51 (m, 2H), 4.90- 4.89 (m, 1H), 3.10-3.00 (m, 1H), 2.36-2.33 (m, 1H), 2.21-2.18 (m, 2H), 1.83-1.78 (m, 3H), 1.57-1.53 (m, 9H). m/z 440.04 (M++1).
Step 2: (S)-tert-butyl 2-(7-(4-(2-((1S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-4- chloro-1 H-imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate (20b):
Figure imgf000074_0001
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediate (20a) from the previous step and Step 2 of Example 16 (16b). m/z 697.26 (M++1).
Step 3: methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-4-chloro-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 20):
Figure imgf000074_0002
Title compound was synthesized by following the procedure as described in Step 2 of Example 6 using the intermediate (20b) from the previous step. 1H NMR- (400 MHz, MeOD), δ 8.46-8.43 (m, 1H), 8.30-8.25 (m, 1H), 8.10 (s, 1H), 7.96-7.94 (m, 1H), 7.88-7.87 (m, 3H), 7.83-7.80 (m, 1H), 7.73-7.67 (m, 1H), 5.38-5.37 (m, 1H), 4.50-4.49 (m, 1H), 4.30- 4.29 (m ,1H), 4.15-4.10 (m, 1H), 4.01-4.00 (m, 1H), 3.7 (s, 6H), 3.32-3.30 (m, 2H), 3.10- 3.02 (m, 1H), 2.60-2.50 (m, 1H), 2.40-2.15 (m, 6H), 2.10-1.85 (m, 7H), 1.80-1.71 (m, 2H). 1.10-0.95 (m, 6H), 0.91-0.82 (m, 6H); m/z811.4 (M++1). m.p.:185-187 eC. Example 21 : Preparation of methyl ((S)-1-(((1 R,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-5,6-dihydro-4H- benzo[3,4]cyclohepta[1 ,2-d]oxazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)- 3-methyl-1-oxobutan-2-yl)carbamate (Compound 21 ): Step 1 : (S)-tert-butyl 2-(8-bromo-5,6-dihydro-4H-benzo[3,4]cyclohepta[1 ,2-d]oxazol-2- yl)pyrrolidine-1 -carboxylate (21 a):
Figure imgf000075_0001
To the stirred solution of (2S)-2-(2-bromo-5-oxo-6,7,8,9-tetrahydro-5H-benzo[7]annulen-6- yl) 1 -tert-butyl pyrrolidine- 1 ,2-dicarboxylate (3.2 g, 7.04 mmol) [synthesized using procedures described in WO2009/102633] in xylene (25 mL), ammonium acetate (10.86 g, 141 mmol) was added at room temperature and the reaction mass was heated at 130 eC for 15 h. The reaction mixture was poured into water (100 mL) and the compound was extracted with ethyl acetate (100 mL χ 2), dried over sodium sulphate, concentrated under reduced pressure and purified by flash column chromatography (45-55% Ethylacetate/Hexane) to get the title compound as side product, orange colored solid (0.7 g, 23 % yield) along with (S)-tert-butyl 2-(8-bromo-1 , 4,5,6- tetrahydrobenzo[3,4]cyclohepta[1 ,2-d]imidazol-2-yl)pyrrolidine-1 -carboxylate as the major compound. 1 H NMR (CDCI3, 400 MHz): δ 7.52-7.50 (m, 1 H), 7.37-7.28 (m, 2H), 4.88-4.85 (m, 1 H), 3.60-3.40 (m, 2H), 2.95-2.80 (m, 4H), 2.40-1 .90 (m, 6H), 1 .45/1 .25 (s, 9H); m/z 433.05 (M++1 )
Step 2: (S)-tert-butyl 2-(8-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-5,6-dihydro-4H- benzo[3,4]cyclohepta[1 ,2-d]oxazol-2-yl)pyrrolidine-1 -carboxylate (21 b):
Figure imgf000075_0002
Title compound was synthesized by following the procedure as described in Step 3 of Example 1 using intermediate (21a) from the previous step.1H NMR (CDCI3, 400 MHz): δ 7.80-7.60 (m, 2H), 7.56-7.53 (m, 1H), 4.90-4.80 (m, 1H), 3.80-3.50 (m, 2H), 2.95-2.80 (m, 4H), 2.40-1.90 (m, 6H), 1.48/1.44 (s, 9H), 1.25 (m, 12H); m/z 480.90 (M++1)
Step 3: (S)-tert-butyl 2-(8-(4-(2-((1S,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1H- imidazol-5-yl)phenyl)-5,6-dihydro-4H-benzo[3,4]cyclohepta[1,2-d]oxazol-2-yl)pyrrolidine-1- carboxylate (21c):
Figure imgf000076_0001
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediate from Step 2 of Example 4 (4b) and the boronate intermediate (21b) from the previous step.1H-NMR (400 MHz, CDCI3) δ 7.78-7.45 (m, 8H), 5.07-4.86 (m, 2H), 4.40-4.20 (m, 2H), 3.70-3.43 (m, 3H), 3.05-2.90 (m, 4H), 2.45-1.90 (m, 10H), 1.50-1.31 (m, 18H), m/z 680.40 (M++1 ).
Step 4: methyl ((S)-1-(((1R,2S)-2-(5-(4-(2-((S)-1-((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-5,6-dihydro-4H-benzo[3,4]cyclohepta[1,2-d]oxazol-8- yl)phenyl)-1H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 21):
Figure imgf000076_0002
Title compound was synthesized by following the procedure as described in Step 5 of
Example 1 using the intermediate (21c) from the previous step.1H NMR (DMSO-afe, 400
MHz), δ 11.85-11.70 (m, 1H), 7.82-7.03 (m, 11H), 5.15-5.02 (m, 1H), 4.44-4.32 (m, 2H), 4.1 1 -4.10 (m, 2H), 3.90-3.75 (m, 4H), 3.51 -3.45 (s, 6H), 2.94-2.80 (m, 4H), 2.24-1 .55 (m, 12H), 0.97-0.71 (m, 12H), m/z 794.50 (M++1 ). m.p.:138-141 eC.
Example 22: Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5- dihydrobenzo[2,3]oxepino[4,5-d]oxazol-8-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 22):
Step 1 : (S)-tert-butyl 2-(8-bromo-4,5-dihydrobenzo[2,3]oxepino[4,5-d]oxazol-2- yl)pyrrolidine-1 -carboxylate (22a):
Figure imgf000077_0001
Title compound was synthesized by following the procedure described in Step 1 of Example 21 using (2S)-2-(8-bromo-5-oxo-2,3,4,5-tetrahydrobenzo[b]oxepin-4-yl) 1 -tert- butyl pyrrolidine- 1 ,2-dicarboxylate [synthesized as described in WO2009/102633] as the starting material. 1 H NMR (DMSO-afe, 400 MHz): δ 7.57-7.53 (m, 1 H), 7.38-7.35 (m, 1 H), 7.30-7.28 (m, 1 H), 4.90-4.82 (m, 1 H), 4.30-4.26 (m, 2H), 3.50-3.45 (m, 1 H), 3.00-2.95 (m, 2H), 2.40-2.20 (m, 1 H), 2.05-1 .90 (m, 4H), 1 .38/1 .15 (s, 9H); m/z 435.00 (M++1 )
Step 2: (S)-tert-butyl 2-(8-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-4,5- dihydrobenzo[2,3]oxepino[4,5-d]oxazol-2-yl)pyrrolidine-1 -carboxylate (22b):
Figure imgf000077_0002
Title compound was synthesized by following the procedure as described in Step 3 of Example 1 using intermediate (22a) from the previous step. 1 H NMR (CDCI3, 400 MHz): δ 7.66-7.63 (m, 1 H), 7.56-7.53 (m, 2H), 5.07-4.90 (m, 1 H), 4.29-4.21 (m, 2H), 3.70-3.60 (m, 1 H), 3.15-3.05 (m, 2H), 2.40-1 .70 (m, 5H), 1 .48/1 .44 (s, 9H), 1.25 (m, 12H); m/z 482.98
(M++1 )
Step 3: (S)-tert-butyl 2-(8-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydrobenzo[2,3]oxepino[4,5-d]oxazol-2-yl)pyrrolidine-1 - carboxylate (22c):
Figure imgf000078_0001
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using the intermediates from Step 2 of Example 9 (9b) and the boronate (22b) from the previous step. 1 H-NMR (400 MHz, CDCI3) δ 7.83-7.35 (m, 8H), 5.07-4.90 (m, 2H), 4.40-4.20 (m, 3H), 3.75-3.40 (m, 4H), 3.25-3.10 (m, 2H), 2.45-1 .72 (m, 8H), 1 .65-1 .51 (m, 18H), m/z 682.40 (M++1 ).
Step 4: methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydrobenzo[2,3]oxepino[4,5-d]oxazol-8-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 22):
Figure imgf000078_0002
Title compound was synthesized by following the procedure as described in Step 2 of Example 6 using the intermediate (22c) from the previous step. 1H NMR (DMSO-afe, 400 MHz), δ 12.10-1 1 .80 (m, 1 H), 8.14-6.90 (m, 1 1 H), 5.15-5.10 (m, 1 H), 4.40-4.30 (m, 4H), 4.15-4.10 (m, 1 H), 3.95-3.70 (m, 3H), 3.55-3.50 (m, 6H), 3.10-2.95 (m, 2H), 2.14-1 .95 (m, 12H), 0.97-0.71 (m, 12H), m/z 796.50 (M++1 ). m.p.:220-222 eC. Example 23: Preparation of methyl ((S)-1-(((1 R,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5- dihydrobenzo[2,3]oxepino[4,5-d]oxazol-8-yl)phenyl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (Compound 23):
Step 1 : (S)-tert-butyl 2-(8-(4-(2-((1 S,2R)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenyl)-4,5-dihydrobenzo[2,3]oxepino[4,5-d]oxazol-2-yl)pyrrolidine-1 - carboxylate (23a):
Figure imgf000079_0001
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 , using the intermediates from Step 2 of Example 4 (4b) and the intermediate from step 2 of Example 22 (22b). 1 H-NMR (400 MHz, CDCI3) δ 7.83-7.35 (m, 8H), 5.07- 4.90 (m, 2H), 4.40-4.20 (m, 3H), 3.75-3.40 (m, 4H), 3.25-3.10 (m, 2H), 2.45-1 .72 (m, 8H), 1 .65-1 .51 (m, 18H), m/z 682.40 (M++1 ).
Step 2: methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydrobenzo[2,3]oxepino[4,5-d]oxazol-8-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 23):
Figure imgf000079_0002
Title compound was synthesized by following the procedure as described in Step 2 of Example 6 using the intermediate (23a) from the previous step. 1 H NMR (DMSO-afe, 400 MHz), δ 12.10-1 1 .80 (m, 1 H), 8.14-6.90 (m, 1 1 H), 5.15-5.10 (m, 1 H), 4.40-4.30 (m, 4H), 4.15-4.10 (m, 1 H), 3.95-3.70 (m, 3H), 3.55-3.50 (m, 6H), 3.10-2.95 (m,2H), 2.14-1 .95 (m, 12H), 0.97-0.71 (m, 12H), m/z 796.50 (M++1 ). m.p.:166-168 eC. Example 24: Preparation of methyl ((S)-1-(((1S,2S)-2-(7-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)- 4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (Compound 24):
Step 1 : (1 S,2S)-6-bromo-1 -oxo-1 ,2,3,4-tetrahydronaphthalen-2-yl 2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylate (24a):
Figure imgf000080_0001
Title compound was synthesized by following the procedure as described in Step 1 of Example 1 using 2,6-dibromo-3,4-dihydronaphthalen-1 (2H)-one [synthesized using procedures described in WO2010/1 17635A1 ] and (1 S,2S)-2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylic acid [synthesized using procedures described in J. Org. Chem., 2001 , 66 (16), 5629-5632] as starting materials. 1 H-NMR (400 MHz, CDCI3) δ 8.09-8.07 (m, 1 H), 7.85-7.81 (m, 1 H), 7.55-7.50 (m, 2H), 5.56-5.50 (m, 1 H), 4.30-4.05 (m, 2H), 3.30-3.05 (m, 4H), 2.45-1 .60 (m, 6H), 1 .40 (s, 9H); m/z 352.10 [(M++1 ) - 100] (value observed for the de-Boc product).
Step 2: tert-butyl ((1 S,2S)-2-(7-bromo-4,5-dihydro-3H-naphtho[1 ,2-d]imidazol-2- yl)cyclopentyl)carbamate (24b):
Figure imgf000080_0002
Title compound was synthesized by following the procedure as described in Step 2 of Example 1 , using the intermediate (24a) from the previous step. 1 H-NMR (400 MHz, CDCI3) δ 7.34-7.30 (m, 3H), 4.90-4.87 (m, 1 H), 4.19-4.12 (m, 1 H), 3.15-3.13 (m, 1 H), 3.05- 2.85 (m, 4H), 2.45-2.10 (m, 2H), 1 .90-1 .60 (m, 4H), 1 .50 (s, 9H); m/z 432.10 (M++1 ). Step 3: (S)-tert-butyl 2-(5-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-4,5- dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)pyrrolidine-1 -carboxylate (24c).
Figure imgf000081_0001
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 , using the intermediates (24b) from the previous step and (S)-tert-butyl 2-(5-(4- (4,4,5, 5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)-1 H-imidazol-2-yl)pyrrolidine-1 - carboxylate [synthesized using procedures described in ref. WO2010/99527A1 ] as starting materials. 1 H-NMR (400 MHz, CDCI3) δ 1 1 .00 (bs, 1 H), 10.50 (bs, 1 H), 7.85-7.40 (m, 7H), 7.20-7.10 (m, 1 H), 5.07-4.86 (m, 2H), 4.40-4.20 (m, 2H), 3.50-3.43 (m, 2H), 3.17-3.09 (m , 3H), 2.93-2.89 (m, 2H), 2.45-1 .80 (m, 8H), 1 .60-1 .51 (m, 18H); m/z 665.40 (M++1 ).
Step 4: methyl ((S)-1 -(((1 S,2S)-2-(7-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2- d]imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 24):
Figure imgf000081_0002
Title compound was synthesized by following the procedure as described in Step 5 of Example 1 using the intermediate (24c) from the previous step. 1H NMR (DMSO-afe, 400 MHz), δ 12.10-1 1 .55 (m, 2H), 8.13-7.46 (m, 9H), 7.31 -7.29 (m, 1 H), 7.04-7.01 (m, 1 H), 5.10-5.05 (m, 1 H), 4.40-4.32 (m, 2H), 4.1 1 -4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51 -3.45 (s, 6H), 3.02-2.67 (m ,4H), 2.14-1 .55 (m, 10H), 0.99-0.51 (m, 12H), m/z 779.30 (M++1 ). m.p.:181 -183 eC. Example 25: Preparation of methyl ((S)-1-(((1S,2R)-2-(7-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)- 4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2-yl)cyclopentyl)amino)-3-methyl-1- oxobutan-2-yl)carbamate (Compound 25): Step 1 : (1 R,2S)-6-bromo-1 -oxo-1 ,2,3,4-tetrahydronaphthalen-2-yl 2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylate (25a):
Figure imgf000082_0001
Title compound was synthesized by following the procedure as described in Step 1 of Example 1 , using 2,6-dibromo-3,4-dihydronaphthalen-1 (2H)-one [synthesized using procedures described in WO2010/1 17635A1 ] and (1 R,2S)-2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylic acid [synthesized using procedures described in Tetrahedron: Asymmetry, 2008, 19, 2796-2803] as starting materials. 1H- NMR (400 MHz, CDCI3) δ 8.09-8.07 (m, 1 H), 7.85-7.81 (m, 1 H), 7.55-7.50 (m, 2H), 5.56- 5.50 (m, 1 H), 4.30-4.05 (m, 2H), 3.30-3.05 (m, 4H), 2.45-1 .60 (m, 6H), 1 .40 (s, 9H); m/z 352.10 [(M++1 ) - 100] (value observed for the de-Boc product).
Step 2: tert-butyl ((1 S,2R)-2-(7-bromo-4,5-dihydro-3H-naphtho[1 ,2-d]imidazol-2- yl)cyclopentyl)carbamate (25b):
Figure imgf000082_0002
Title compound was synthesized by following the procedure as described in Step 2 of Example 1 , using the intermediate (25a) from the previous step. 1 H-NMR (400 MHz, CDCI3) δ 7.34-7.30 (m, 3H), 5.19-5.16 (m, 1 H), 4.90-4.87 (m, 1 H), 4.40-4.30 (m, 1 H), 3.36- 3.31 (m, 1 H), 3.05-2.80 (m, 4H), 2.45-2.10 (m, 2H), 1 .90-1 .60 (m, 4H), 1 .50/1 .43 (s, 9H); m/z 431 .85 (M++1 ). Step 3: (S)-tert-butyl 2-(5-(4-(2-((1 R,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-4,5- dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)pyrrolidine-1 -carboxylate (25c):
Figure imgf000083_0001
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 , using the intermediates (25b) from the previous step and (S)-tert-butyl 2-(5-(4- (4,4,5, 5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)-1 H-imidazol-2-yl)pyrrolidine-1 - carboxylate [synthesized using procedures described in ref. WO2010/99527A1 ] as starting materials. 1 H-NMR (400 MHz, CDCI3) δ 1 1 .00 (bs, 1 H), 10.50 (bs, 1 H), 7.85-7.40 (m, 7H), 7.20-7.10 (m, 1 H), 5.07-4.86 (m, 2H), 4.40-4.20 (m, 2H), 3.50-3.43 (m, 2H), 3.17-3.09 (m, 3H), 2.93-2.89 (m, 2H), 2.45-1 .80 (m, 8H), 1 .60-1 .51 (m, 18H), m/z 665.40 (M++1 ).
Step 4: methyl ((S)-1 -(((1 S,2R)-2-(7-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2- d]imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 25):
Figure imgf000083_0002
Title compound was synthesized following the procedure as described in Step 2 of Example 6 using the intermediate (25c) from the previous step. 1H NMR (DMSO-afe, 400 MHz), δ 12.10-1 1 .55 (m, 2H), 8.13-7.46 (m, 8H), 7.41 -7.29 (m, 2H), 7.04-6.90 (m, 1 H), 5.10-5.05 (m, 1 H), 4.40-4.32 (m, 2H), 4.1 1 -4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51 -3.45 (s, 6H), 3.02-2.67 (m ,4H), 2.14-1 .50 (m, 10H), 0.99-0.51 (m, 12H), m/z 779.30 (M++1 ). m.p.: 224 eC. Example 26: Preparation of methyl ((S)-1-(((1S,2R)-2-(7-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)- 1 H-naphtho[1 ,2-d]imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate (Compound 26):
Step 1 : (S)-tert-butyl 2-(5-(4-(2-((1 R,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)pyrrolidine-1 -carboxylate (26a):
Figure imgf000084_0001
Title compound was synthesized by following the procedure as described in Step 1 of Example 16, using the intermediate from Step 3 of Example 25 (25c). 1 H-NMR (400 MHz, CDCI3) δ 8.10-7.45 (m, 9H), 7.26 (s, 1 H), 5.15-5.03 (m, 2H), 4.55-4.35 (m, 1 H), 3.75- 3.35 (m, 2H), 3.25-3.15 (m, 2H), 2.55-1 .60 (m, 8H), 1 .51 -1 .30 (m, 18H), m/z 663.40 (M++1 ).
Step 2: methyl ((S)-1 -(((1 S,2R)-2-(7-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol-2- yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 26):
Figure imgf000084_0002
Title compound was synthesized by following the procedure as described in Step 2 of Example 6 using the intermediate (26a) from the previous step. 1 H NMR (DMSO-afe, 400 MHz), δ 12.10-1 1 .55 (m, 2H), 8.60-7.50 (m, 1 1 H), δ 7.31 -7.29 (m, 1 H), 7.10-6.85 (m, 1 H), 5.10-5.05 (m, 1 H), 4.55-4.40 (m, 2H), 4.1 1 -4.07 (m, 2H), 3.87-3.81 (m, 4H), 3.51 -3.45 (s, 6H), 2.40-1 .50 (m, 10H), 0.99-0.51 (m, 12H), m/z 777.30 (M++1 ). m.p. :238-240 eC. Example 27: Preparation of methyl ((S)-1-(((1S,2S)-2-(7-(4-(2-((S)-1-((S)-2-amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol- 2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 27):
Step 1 : (S)-tert-butyl 2-(5-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)pyrrolidine-1 -carboxylate (27a):
Figure imgf000085_0001
Title compound was synthesized by following the procedure as described in Step 1 of Example 16, using the intermediate from Step 3 of Example 24 (24c) as starting material. 1 H-NMR (400 MHz, CDCI3) δ 8.10-7.45 (m, 9H), 7.26 (s, 1 H), 5.15-5.03 (m, 2H), 4.60-4.35 (m, 1 H), 3.75- 3.35 (m, 2H), 3.25-3.15 (m, 2H), 2.55-1 .60 (m, 8H), 1 .51 -1 .30 (m, 18H), m/z 663.40 (M++1 ).
Step 2: methyl ((S)-1 -(((1 S,2S)-2-(7-(4-(2-((S)-1 -((S)-2-amino-3-methylbutanoyl)pyrrolidin- 2-yl)-1 H-imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2-d]imidazol-2-yl)cyclopentyl)amino)-3- methyl-1 -oxobutan-2-yl)carbamate (Compound 27):
Figure imgf000085_0002
Title compound was synthesized by following the procedure as described in Step 2 of Example 6 using the intermediate (27a) from the previous step. 1H NMR (400 MHz, MeOD), δ 8.55-8.47 (m, 2H), 8.20-7.75 (m, 8H), 7.42-7.30 (m, 2H), 5.30-5.27 (m, 2H), 4.65-4.60 (m, 2H), 4.27-4.15 (m, 2H), 3.95-3.85 (m, 3H), 3.70-3.65 (m, 6H), 2.70-1 .95 (m, 10H), 0.99-0.80 (m, 12H), m/z 777.30 (M++1 ). Example 28: Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)(ethyl)amino)-3- methyl-1 -oxobutan-2-yl)carbamate (Compound 28): Step 1 : (1 S,2S)-ethyl-2-(ethylamino)cyclopentanecarboxylate (28a):
Figure imgf000086_0001
To a solution of (1 S,2S)-ethyl 2-aminocyclopentanecarboxylate (0.2 g, 1 .27 mmol) [synthesized using procedures described in J. Org. Chem., 2001 , 66 (16), 5629-5632] in THF (10 mL), acetic acid (0.07 mL, 1 .27 mmol) and acetaldehyde (0.07 mL, 1 .27 mmol) were added and stirred for 1 h at room temperature after which sodium triacetoxyborohydride (0.4 g, 1 .91 mmol) was added and stirred at room temperature for 16 h. After completion, the mixture was added to saturated NaHC03 solution and extracted with EtOAc (2 x 35 mL). The organic layer was washed with brine, dried over Na2S04 and concentrated under reduced pressure to get colorless oil (0.14 g, 59%) which was taken to next step without any purification. 1 H-NMR (400 MHz, CDCI3), δ 4.20-4.10 (m, 2H), 3.56 (q, J = 7.2 Hz, 1 H), 2.72 (q, J = 7.2 Hz , 1 H), 2.62-2.50 (m, 2H), 1 .94-1 .50 (m, 6H), 1 .28- 1 .23 (t, J = 7.2 Hz , 3H), 1 .05-1.02 (t, J = 7.2 Hz , 3H); m/z 186 (M++1 ).
Step 2: (1 S,2S)-ethyl 2-((tert-butoxycarbonyl)(ethyl)amino)cyclopentanecarboxylate (28b):
Figure imgf000086_0002
To a stirred solution of (1 S, 2S)-ethyl-2-(ethylamino)cyclopentanecarboxylate (28a) (0.22 g, 1 .19 mmol) in 1 ,4-Dioxane (10 mL) was added a solution of NaHC03 (0.35 g, 4.16 mmol) in water (10 mL) till the solution turned basic. To this Boc anhydride (0.33 mL, 1 .43 mmol) was added. The resultant mixture was stirred at room temperature for 16 h. The reaction mixture was poured into water and extracted with ethyl acetate (2 x 1 5 ml_). The combined organic layers were dried over sodium sulfate and concentrated under reduced pressure to get white solid (0.2 g, 59%) which was used in the next step without any further purification. 1 H-NMR (400 MHz, CDCI3), δ 4.25 (bs, 1 H), 4.16-4.1 1 (m, 1 H), 3.20- 3.19 (m, 2H), 2.93 (q, J = 7.2 Hz, 2H), 2.05-1 .70 (m, 6H), 1 .47-1.44 (m, 9H), 1 .26 (t, J = 7.2 Hz, 3H), 1 .12 (t, J = 7.2 Hz, 3H); m/z 285.2 (M+).
Step 3: (1 S,2S)-2-((tert-butoxycarbonyl)(ethyl)amino)cyclopentanecarboxylic acid (28c):
Figure imgf000087_0001
To a stirred solution of (1 S, 2S)-ethyl-2-((tert- butoxycarbonyl)(ethyl)amino)cyclopentanecarboxylate (28b) (1 .3 g, 4.56 mmol) in Tetrahydrofuran (10 ml_), Ethanol (10 mL) and Water (10 mL) lithium hydroxide (0.44 g, 18.22 mmol) was added. The reaction mixture was stirred at room temperature for 4 h. The reaction mixture was concentrated under reduced pressure and diluted with water and extracted with ethyl acetate (2 χ 35 mL). The aqueous layer was acidified by 1 N HCI. and extracted with ethyl acetate (2 x 1 0 mL) and the combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure to yield a white solid (0.7 g, 59%) which was used in the next step without any purification. 1 H-NMR (400 MHz, CDCI3), δ 4.33-4.29 (m, 1 H), 3.28-3.18 (m, 1 H), 3.00-2.90 (m, 2H), 2.00-1 .65 (m, 6H), 1 .47 (s, 9H), 1 .13 (t, J = 7.2 Hz, 3H); m/z 257.18 (M+).
Step 4: (1 S, 2S)-2-(4-bromophenyl)-2-oxoethyl 2-((tert-butoxycarbonyl)(ethyl) amino) cyclopentane carboxylate (28d):
Figure imgf000087_0002
To a stirred solution of (1 S,2S)-2-((tert- butoxycarbonyl)(ethyl)amino)cyclopentanecarboxylic acid (28c) (0.7 g, 2.72 mmol) in ACN (20 mL), 2-bromo-1 -(4-bromophenyl)ethanone (0.76 g, 2.72 mmol) and DIPEA (0.35 g, 2.72 mmol) were added and stirred for 3 h at room temperature. The contents were concentrated under reduced pressure and water (50 mL) was added to it and extracted with DCM (20 mL χ 2). The combined organic layers were washed with brine, dried over Na2S04 and concentrated under reduced pressure to get white foam (0.9 g, 73%) that was used in the next step without any further purification, m/z 354 [(M+) - 100].
Step 5: tert-butyl ((1 S,2S)-2-(5-(4-bromophenyl)-1 H-imidazol-2- yl)cyclopentyl)(ethyl)carbamate (28e):
Figure imgf000088_0001
To a stirred solution of (1 S,2S)-2-(4-bromophenyl)-2-oxoethyl-2-((tert- butoxycarbonyl)(ethyl)amino)cyclopentanecarboxylate (28d) (0.900 g, 1 .98 mmol) in toluene (12 mL) ammonium acetate (4.58 g, 59.4 mmol) was added and heated for 18 hr at 1 10 eC. The mixture was added to water (100mL) and extracted with DCM (70 mL χ 2). The combined organic layers were washed with brine and dried over Na2S04, concentrated under reduced pressure and purified by flash chromatography using 5% EtOAc / pet-ether to get yellow solid (0.22 g, 26%). 1 H-NMR (400 MHz, CDCI3): δ 7.70- 7.60 (m, 1 H), 7.48-7.46 (m, 3H), 7.16 (bs, 1 H), 4.90-4.80 (m, 1 H), 3.30-3.20 (m, 2H), 3.10-3.00 (m, 1 H), 2.50-2.40 (m, 1 H), 2.00-1 .70 (m, 5H), 1 .50 (s, 9H), 1 .18-1 .13 (m, 3H); m/z 434.98 (M++1 ).
Step 6: (S)-tert-butyl 2-(7-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)(ethyl)amino) cyclopentyl)-1 H-imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate (28f):
Figure imgf000089_0001
Title compound was synthesized by following the procedure as described in Step 4 of Example 1 using intermediate (28e) from the previous step and (S)-tert-butyl 2-(7-(4,4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate [synthesized using procedures described in WO2009/102633]. 1 H-NMR (400 MHz, CDCI3): δ 1 1 .00 (bs, 1 H), 10.50 (bs, 1 H), 7.83-7.52 (m, 8H), 5.07-4.86 (m, 2H), 3.50-3.43 (m, 3H), 3.2-3.26 (m, 2H), 3.08-3.03 (m, 2H), 2.97-2.90 (m, 2H), 2.50- 2.40 (m, 2H), 2.25-1 .72 (m, 8H), 1 .60-1 .51 (m, 18H), 1 .20-1 .15 (m, 3H), m/z 693.50 (M++1 ). Step 7: methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)(ethyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 28):
Figure imgf000089_0002
Title compound was synthesized by following the procedure as described in Step 5 (Example 1 ) using intermediate (28f) from the previous step. 1 H NMR (DMSO-afe, 400 MHz), δ 1 1 .80-1 1 .75 (bs, 2H), 8.14-7.92 (m, 6H), 7.34-7.31 (m, 1 H), 6.99-6.96 (m, 1 H), 5.17-4.99 (m, 4H), 4.20-3.85 (m, 5H), 3.55-3.50 (m , 6H), 3.15-2.85 (m, 4H), 2.14-1 .50 (m, 12H), 0.99-0.51 (m, 15H), m/z 807.50 (M++1 ). m.p.: 193-195 eC. Example 29: Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(4-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)(methyl)amino)-3- methyl-1 -oxobutan-2-yl)carbamate (Compound 29): Step 1 : (1 S, 2S)-ethyl 2-(methylamino)cyclopentanecarboxylate (29a):
Figure imgf000090_0001
To a solution of (1 S,2S)-ethyl 2-aminocyclopentanecarboxylate (2 g, 12.72 mmol) in ethanol (30 mL), formaldehyde solution (37% v/v, 0.385 mL, 14 mmol) was added and stirred for 1 h. Then, 10% Pd-C (2.03 g, 19.1 mmol) was added to the mixture and stirred for 2 h under H2 atmosphere. After completion of reaction, the mixture was filtered through a pad of celite. The celite bed was washed with ethanol (20 mL). The combined organic layers were evaporated under reduced pressure and the residue (1 .4 g, 64%) was taken to next step without any purification. 1 H-NMR (400 MHz, CDCI3), δ 4.20-4.10 (m, 2H), 3.07 (q, J = 7.6 Hz, 1 H), 2.70 (q, J = 7.2 Hz, 1 H), 2.26 (s, 3H), 1 .94-1 .50 (m, 6H), 1 .27 (t, J = 7.2 Hz, 3H); m/z 171 .23 (M+)
Step 2: (1 S, 2S)-ethyl 2-((tert-butoxycarbonyl)(methyl)amino) cyclopentanecarboxylate (29b):
Figure imgf000090_0002
Title compound was synthesized by following the procedure as described in Step 2 of Example 28, using intermediate (29a) from the previous step. 1 H-NMR (400 MHz, CDCI3), δ 4.60-4.50 (m, 1 H), 4.19-4.10 (m, 2H), 2.56 (q, J = 7.2 Hz, 1 H), 2.05-1 .70 (m, 6H), 3.27 (s, 3H), 1 .47-1 .44 (m, 9H), 1 .25 (t, J = 7.2 Hz, 3H); m/z 271 .29 (M+) Step 3: (1 S, 2S)-2-((tert-butoxycarbonyl)(methyl)amino)cyclopentanecarboxylic acid (29c):
Figure imgf000091_0001
Title compound was synthesized by following the procedure as described in Step 3 of Example 28, using intermediate 29b as starting material. 1 H-NMR (400 MHz, CDCI3), δ 4.61 -4.59 (m, 1 H), 3.28-3.18 (m, 1 H), 2.81 (s, 3H), 2.00-1 .65 (m, 6H), 1 .47 (s, 9H); m/z 244.08 (M++1 ).
Step 4: (1 S,2S)-2-(4-bromophenyl)-2-oxoethyl 2-((tert-butoxycarbonyl)(methyl) amino)cyclopentanecarboxylate (29d) :
Figure imgf000091_0002
Title compound was synthesized by following the procedure as described in Step 4 of Example 28, using intermediate 29c as starting material, m/z 439.79 (M+)
Step 5: tert-butyl ((1 S,2S)-2-(5-(4-bromophenyl)-1 H-imidazol-2- yl)cyclopentyl)(methyl)carbamate (29e):
Figure imgf000091_0003
Title compound was synthesized by following the procedure as described in Step 5 of Example 28, using intermediate 29d as starting material. 1 H-NMR (400 MHz, CDCI3), δ 7.60-7.45 (m, 4H), 7.20 (bs, 1 H), 4.90-4.88 (m, 1 H), 3.30-3.23 (m, 1 H), 2.80 (s, 3H), 2.50- 2.40 (m, 1 H), 2.00-1 .70 (m, 5H), 1 .50 (s, 9H); m/z 420.20 (M++1 ).
Step 6: (S)-tert-butyl 2-(7-(4-(2-((1 S,2S)-2-((tert-butoxycarbonyl)(methyl)amino) cyclopentyl)-1 H-imidazol-5-yl)phenyl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate (29f):
Figure imgf000092_0001
Title compound was synthesized by following the procedure as described in Step 6 of Example 28, using intermediate (29e) from the previous step and (S)-tert-butyl 2-(7- (4,4,5, 5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-2- yl)pyrrolidine-1 -carboxylate [synthesized using procedures described in WO2009/102633]. 1 H-NMR (400 MHz, CDCI3), δ 1 1 .00 (bs, 1 H), 10.50 (bs, 1 H), 7.83-7.52 (m, 8H), 5.07-4.86 (m, 2H), 3.50-3.43 (m, 3H), 3.20-3.10 (m, 2H), 2.97-2.90 (m, 2H), 2.85 (s, 3H), 2.50-2.40 (m, 2H), 2.25-1 .72 (m, 8H), 1 .60-1 .51 (m, 18H), m/z 679.50 (M++1 ).
Step 7: methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)(methyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 29):
Figure imgf000092_0002
Title compound was synthesized by following the procedure as described in Step 7 of Example 28, using intermediate 29f as starting material. 1 H-NMR (400 MHz, DMSO-c6) δ 1 1 .80-1 1 .75 (bs, 2H), 8.14-7.92 (m, 6H), 7.34-7.31 (m, 1 H), 6.99-6.96 (m, 1 H), 5.17-4.99 (m, 2H), 4.20-3.85 (m, 4H), 3.55-3.50 (m, 6H), 3.15-2.85 (m, 5H), 2.14-1.50 (m, 12H), 0.99-0.51 (m, 15H), m/z 793.50 (M++1 ). m.p. : 162-155 eC. Example 30: Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(7-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)-9H- fluoren-2-yl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate (Compound 30): Step 1 : 1 -(7-bromo-9H-fluoren-2-yl)ethanone (30a):
Figure imgf000093_0001
To a degassed solution of 2,7-dibromo-9H-fluorene (3.0 g, 9.3 mmol) and tributyl(1 - ethoxyvinyl)stannane (3.3 g, 9.3 mmol) in 1 ,4-Dioxane (20 mL) was added Pd(PPh3)4 (0.56 g, 0.463 mmol) and PdCI2(dppf) (0.34 g, 0.46 mmol) and the contents were heated at 80 eC for 5 h. The reaction mixture was cooled to room temperature and water was added and the contents were extracted with EtOAc (2 χ 20 mL). The organic layer was washed with brine, dried over sodium sulphate and concentrated under reduced pressure to yield 2-bromo-7-(1 -ethoxyvinyl)-9,9 dimethyl-9H-fluorene as an orange liquid. To the above product THF (6 mL) and 2M HCI (10 mL) was added and the reaction was stirred at rt for 2 h. The contents were evaporated under reduced pressure, the residue was dissolved in water and extracted with EtOAc (2 χ 20 mL). The organic contents were washed with brine, dried over sodium sulphate and purified by flash chromatography (3% EtOAc/Hexane) to yield a white colored solid (0.90g, 33.9%).
1 H-NMR (400 MHz, CDCI3): δ 8.15 (bs, 1 H), 8.04-8.01 (m, 1 H), 7.83-7.81 (m, 1 H), 7.74- 7.70 (m, 2H), 7.57-7.55 (m, 1 H), 3.96 (s, 2H), 2.67 (s, 3H); m/z 286.07 (M+).
Step 2: 2,2-dibromo-1 -(7-bromo- -fluoren-2-yl)ethanone (30b):
Figure imgf000093_0002
To a solution of 1 -(7-bromo-9H-fluoren-2-yl)ethanone (30a) (0.4 g, 1 .4 mmol) in chloroform (15 mL) was added bromine (0.07 mL, 1 .4 mmol) at 0 eC under nitrogen atmosphere. The cooling bath was removed and the reaction mixture was warmed to rt and stirred for 2 h. Water was added to the reaction mixture and the organic contents were washed with sodium bisulphite solution, brine, dried over sodium sulphate and evaporated under reduced pressure to yield a yellow colored solid that was used in the next step without further purification (0.550, 89%). 1 H-NMR (400 MHz, CDCI3) δ 8.27 (bs, 1 H), 8.18-8.14 (m, 1 H), 8.09-8.04 (m, 1 H), 7.88-7.80 (m, 1 H), 7.77-7.72 (m, 2H), 7.62-7.59 (m, 1 H), 4.10-3.99 (m, 2H).
Step 3: 2-bromo-1 -(7-bromo-9H-fluoren-2-yl)ethanone 30c):
Figure imgf000094_0001
To a solution of 2-bromo-1 -(7-bromo-9H-fluoren-2-yl)ethanone (30b) (0.35 g, 0.96 mmol) in THF (3 mL) at 0 eC was added triethylamine (0.24 ml_, 1 .69 mmol) and diethyl phosphite (0.22 mL, 1 .69 mmol). The reaction was gradually warmed to room temperature and the mixture was stirred for 1 .5 h after which excess THF was removed under reduced pressure. The crude was triturated with n-pentane to get a white solid (0.350g, 85%) that was used in the next step without further purification. 1 H NMR (DMSO-afe, 400 MHz), δ 8.29 (bs, 1 H), 8.22-8.14 (m, 2H), 8.06-8.03 (m, 1 H), 7.90-7.87 (m, 1 H), 7.67-7.65 (m, 1 H), 4.97 (s, 2H), 4.03 (s, 2H); m/z 386 (M++23).
Step 4: (1 S,2S)-2-(7-bromo-9H-fluoren-2-yl)-2-oxoethyl-2-((tert-butoxycarbonyl) amino)cyclopentanecarboxylate (30d) :
Figure imgf000094_0002
Title compound was synthesized by following the procedure as described in Step 1 of Example 1 using intermediate (30c) from the previous step and (1 S,2S)-2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylic acid [synthesized using procedures described in J. Org. Chem., 2001 , 66 (16), 5629-5632] as starting materials. 1 H-NMR (400 MHz, CDCI3): δ 8.1 1 -8.10 (m, 1 H), 7.98-7.96 (m, 1 H), 7.86-7.83 (m, 1 H), 7.75-7.71 (m, 2H), 7.58-7.56 (m, 1 H), 5.53-5.31 (m, 2H), 3.97 (s, 2H), 3.90-3.85 (m, 1 H), 2.80-2.75 (m, 1 H), 2.20-2.10 (m, 4H), 1 .90-1 .80 (m, 2H), 1 .45 (s, 9H); m/z 414.10 [(M++1 ) - 100] (value observed for the de-Boc product). Step 5: tert-butyl ((1 S,2S)-2-(5-(7-bromo-9H-fluoren-2-yl)-1 H-imidazol-2- yl)cyclopentyl)carbamate (30e):
Figure imgf000095_0001
Title compound was synthesized by following the procedure as described in Step 2 of Example 1 using intermediate (30d) from the previous step. 1 H-NMR (400 MHz, CDCI3): δ 7.88 (bs, 1 H), 7.76-7.61 (m, 4H), 7.51 -7.48 (m, 1 H), 7.33 (s, 1 H), 4.93-4.91 (m, 1 H), 4.22- 4.18 (s, 1 H), 3.91 (s, 2H), 3.20-3.18 (m, 1 H), 2.50-2.20 (m, 4H), 1 .90-1 .80 (m, 2H), 1.50 (s, 9H); m/z 493.99 (M++1 ).
Step 6: tert-butyl ((1 S,2S)-2-(5-(7-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-9H-fluoren- 2-yl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (30f):
Figure imgf000095_0002
Boc
Title compound was synthesized by following the procedure as described in Step 3 of Example 1 using intermediate (30e) from the previous step. 1 H-NMR (400 MHz, CDCI3): δ 8.00 (bs, 1 H), 7.85-7.77 (m, 3H), 7.71 -7.66 (m, 1 H), 7.58-7.47 (m, 1 H), 7.36-7.32 (m, 1 H), 4.89-4.87 (m, 1 H), 4.20-4.15 (m, 1 H), 3.93 (s, 2H), 3.27-3.20 (m, 1 H), 2.40-1 .60 (m, 6H), 1 .40 (s, 9H), 1 .34 (s, 12H); m/z 542.12 (M++1 ).
Step 7: (S)-tert-butyl 2-(5-(7-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)-9H-fluoren-2-yl)-1 -((2-(trimethylsilyl)ethoxy)methyl)-1 H-imidazol-2- yl)pyrrolidine-1 -carboxylate (30g):
Figure imgf000095_0003
Title compound was synthesized following the procedure as described in Step 4 of Example 1 using the intermediates (30f) from the previous step and (S)-tert-butyl 2-(5- bromo-1 -((2-(trimethylsilyl)ethoxy)methyl)-1 H-imidazol-2-yl)pyrrolidine-1 -carboxylate
[synthesized using procedures described in ref. WO2010/096302 A1] as starting materials maintaining a temperature of 125 eC for 35 min. 1 H-NMR (400 MHz, CDCI3), δ 12.00 (bs, 1 H), 1 1 .50 (bs, 1 H), 8.05-7.05 (m, 8H), 5.70-5.35 (m, 2H), 5.25-5.05 (m, 1 H), 4.95-4.80 (m, 2H), 4.30-4.10 (m, 1 H), 4.05-3.90 (m, 2H), 3.75-3.30 (m, 4H), 3.25-3.10 (m, 1 H), 2.45- 1 .72 (m, 8H), 1 .65-1 .51 (m, 18H), 0.95-0.80 (m, 2H), 0.01 (s, 9H); m/z 781 .2 (M++1 ).
Step 8: methyl ((S)-1 -(((1 S,2S)-2-(5-(7-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)-9H-fluoren-2-yl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 30):
Figure imgf000096_0001
To a stirred solution of (S)-tert-butyl 2-(5-(7-(2-((1 S,2S)-2-((tert- butoxycarbonyl)amino)cyclopentyl)-1 H-imidazol-5-yl)-9H-fluoren-2-yl)-1 -((2-
(trimethylsilyl)ethoxy)methyl)-1 H-imidazol-2-yl)pyrrolidine-1 -carboxylate (30g) (0.06 g, 0.07 mmol) in DCM (4 mL) was added TFA (0.143 ml_, 1 .854 mmol). The resultant mixture was stirred at 45 eC for 18 h. All the volatiles were evaporated under reduced pressure. The residue was washed with diethyl ether to get white solid [m/z 451 .20 (M++1 )]. The solid so obtained (0.030 g, 0.063 mmol) was taken up in DMF (2 mL) and (S)-2- ((methoxycarbonyl)amino)-3-methylbutanoic acid (0.024 g 0.13 mmol), DIPEA (0.06 mL, 0.3 mmol) and HATU (0.052 g, 0.13 mmol) was added and the resultant mixture was stirred at room temperature for 16 h under nitrogen atmosphere. Ice cold water was added in the reaction mixture and the off white colored precipitate was filtered off, washed with water, dried under high vacuum and purified by preparative HPLC to yield the title compound as a pale yellow solid (0.02 g, 25%). 1 H NMR (DMSO-c6, 400 MHz): δ 1 1 .80 (bs, 2H), 8.20-8.16 (m, 1 H), 7.93-7.50 (m, 8H), 7.33-7.31 (m, 1 H), 7.09-7.07 (m, 1 H), 5.10- 5.05 (m, 1 H), 4.40-4.32 (m, 1 H), 4.10-3.85 (m, 6H), 3.55-3.45 (m, 7H), 3.20-3.15 (m, 2H), 2.20-1 .50 (m, 10H), 0.99-0.51 (m, 12H), m/z 765.50 (M++1 ). m.p.: 176-178 eC.
Example 31 : Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(7-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)-9,9- dimethyl-9H-fluoren-2-yl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1-oxobutan- 2-yl)carbamate (Compound 31 ):
Step 1 : 1 -(7-bromo-9,9-dimethyl-9H-fluoren-2-yl)ethanone (31 a):
Figure imgf000097_0001
Title compound was synthesized by following the procedure as described in Step 1 of Example 30, using 2,7-dibromo-9,9-dimethyl-9H-fluorene as the starting material. 1 H-NMR (400 MHz, CDCI3), δ 8.07-8.05 (m, 1 H), 7.99-7.97 (m, 1 H), 7.82-7.77 (m, 1 H), 7.66 (d, J = 8 Hz, 1 H), 7.61 (d, J = 1 .6 Hz, 1 H), 7.53-7.48 (m, 1 H), 2.67 (s, 3H), 1 .53 (s, 6H); m/z 316.96 (M++1 ).
Step 2: 2,2-dibromo-1 -(7-bromo-9,9-dimethyl-9H-fluoren-2-yl)ethanone
Figure imgf000097_0002
Title compound was synthesized by following the procedure as described in Step 2 of Example 30 using intermediate (31 a) from the previous step. 1 H-NMR (400 MHz, CDCI3), δ 8.08-8.07 (m, 1 H), 7.99-7.97 (m, 1 H), 7.85-7.79 (m, 1 H), 7.66 (d, J = 8 Hz, 1 H), 7.63-7.61 (m, 1 H), 7.53-7.48 (m, 1 H), 6.77 (s, 1 H), 1 .53 (s, 6H); m/z 470.5 (M++1 ).
Step 3: 2-bromo-1 -(7-bromo-9,9-dimethyl-9H-fluoren-2-yl)ethanone (31 c):
Figure imgf000097_0003
Title compound was synthesized by following the procedure as described in Step 3 of Example 30, using intermediate (31b) from the previous step.1H-NMR (400 MHz, CDCI3), δ 8.07-8.05 (m, 1H), 7.99-7.97 (m, 1H), 7.82-7.77 (m, 1H), 7.66 (d, J= 8 Hz, 1H), 7.61 (d, J= 1.6 Hz, 1H), 7.53-7.48 (m, 1H), 4.15 (s, 2H), 1.57 (s, 6H); m/z 394.9 (M++1). Step 4: (1S,2S)-2-(7-bromo-9,9-dimethyl-9H-fluoren-2-yl)-2-oxoethyl 2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylate (31d):
Figure imgf000098_0001
Title compound was synthesized by following the procedure as described in Step 4 of Example 30, using intermediate (31c) from the previous step and (1S,2S)-2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylic acid [synthesized using procedures described in J. Org. Chem., 2001, 66 (16), 5629-5632] as starting materials.1H-NMR (400 MHz, CDCI3), δ 8.0 (s, 1H), 7.94-7.92 (m, 1H), 7.78 (d, J = 8 Hz, 1H), 7.66 (d, J = 8 Hz, 1H), 7.62 (s, 1H), 7.58-7.53 (m, 1H), 5.59-5.53 (m, 1H), 5.40-5.36 (m, 1H), 4.95 (bs, 1H), 4.16 (s, 1H), 2.86-2.84 (m, 1H), 2.12-2.06 (m, 4H),1.80-1.75 (m, 2H), 1.54 (s, 6H), 1.29 (s, 9H); m/z 542.5 (M++1).
Step 5: tert-butyl ((1S,2S)-2-(5-(7-bromo-9,9-dimethyl-9H-fluoren-2-yl)-1 H-imidazol-2- yl)cyclopentyl)carbamate (31 e):
Figure imgf000098_0002
Title compound was synthesized by following the procedure as described in Step 5 of Example 30, using intermediate (31 d) from the previous step.1H-NMR (400 MHz, CDCI3), δ 7.76 (s, 1H), 7.69-7.67 (m, 2H), 7.59-7.55 (m, 2H), 7.50 (d, J = 8 Hz, 1H), 7.32 (s, 1H), 4.92 (bs, 1H), 4.23-4.21 (m, 1H), 3.25-3.21 (m, 1H), 2.48-2.45 (m, 1H), 2.25-1.60 (m, 5H), 1.54 (s, 6H), 1.29 (s, 9H); m/z 524.1 (M++1). Step 6: tert-butyl ((1S,2S)-2-(5-(9,9-dimethyl-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)-9H-f luoren-2-yl)-1 H-imidazol-2-yl)cyclopentyl)carbamate (31 f)
Figure imgf000099_0001
Title compound was synthesized by following procedure as described in Step 6 of Example 30, using intermediate (31 e) from the previous step.1H-NMR (400 MHz, CDCI3), δ 7.89 (s, 1H), 7.84 (m, 2H), 7.76 (m, 1H), 7.77 (s, 1H), 7.51 (m, 1H), 7.49 (s, 1H), 4.24 (m, 1H), 4.04 (m, 1H), 3.27 (m, 1H), 2.17-1.69 (m, 6H), 1.56 (s, 6H), 1.44 (s, 9H), 1.34 (s, 12H); m/z 570.2 (M++1).
Step 7: (S)-tert-butyl 2-(5-(7-(2-((1S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)-9,9-dimethyl-9H-fluoren-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H- imidazol-2-yl)pyrrolidine-1-carboxylate (31 g):
Figure imgf000099_0002
Title compound was synthesized by following the procedure as described in Step 7 of Example 30, using intermediate (31 f) from the previous step.1H-NMR (400 MHz, CDCI3), δ 7.85 (s, 1H), 7.80-7.78 (m, 2H), 7.60-7.58 (m, 1H), 7.51 (s, 1H), 7.25-7.22 (m, 1H), 7.19 (d, J= 8 Hz, 1H), 7.01-6.99 (m, 1H), 5.33 (s, 2H), 5.22-5.21 (m, 1H), 4.90-4.88 (m, 1H), 4.25- 4.24 (m, 1H), 3.66-3.64 (m, 1H), 3.50-3.13 (m, 4H), 2.39-2.06 (m, 5H), 1.95-1.81 (m, 5H), 1.56 (s, 6H), 1.44 (s, 18H), 0.89-0.84 (m, 2H), 0.00 (s, 9H); m/z 809.5 (M++1 ).
Step 8: methyl ((S)-1-(((1S,2S)-2-(5-(7-(2-((S)-1-((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)-9,9-dimethyl-9H-fluoren-2-yl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 31 ):
Figure imgf000100_0001
Title compound was synthesized by following the procedure as described in Step 8 of Example 30, using intermediate from the previous step. 1 H-NMR (400 MHz, CDCI3), δ 8.30-7.57 (m, 5H), 7.53 (s, 1 H), 7.42 (s, 1 H), 6.86 (s, 1 H), 5.39-5.31 (m, 3H), 4.49-4.46 (m, 2H), 4.04-4.02 (m, 1 H), 3.84 (s, 6H), 3.20-3.18 (m, 2H), 3.15-3.12 (m, 1 H), 2.34-1 .75 (m, 12H), 1 .66 (s, 6H), 1 .06-0.89 (m, 12H); m/z 793.5 (M++1 ). m.p.: 157-158 eC.
Example 32: Preparation of methyl ((S)-1-(((1S,2S)-2-(5-(7-(2-((S)-1-((S)-2- (methoxycarbonyl)amino-3-methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5- yl)phenanthren-2-yl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1-oxobutan-2- yl)carbamate (Compound 32):
Step 1 : 1 -(7-bromophenanthren-2-yl)ethanone (32a):
Figure imgf000100_0002
Title compound was synthesized by following the procedure as described in Step 1 of Example 30, using 2,7-dibromophenanthrene [Synthesized according to procedure reported in WO 2010132601 A1 ] as starting material. 1 H-NMR (400 MHz, CDCI3): δ 8.74 (d, J = 9 Hz, 1 H), 8.60 (d, J = 9 Hz, 1 H), 8.55 (s, 1 H), 8.16 (d, J = 8.8 Hz, 1 H), 8.02 (s, 1 H), 7.75 (d, J = 8.8 Hz, 1 H), 7.58-7.51 (m, 2H), 2.56 (s, 3H); LCMS: m/z = 299.0 (M+).
Step 2: 2,2-dibromo-1 -(7-bromophenanthren-2-yl)ethanone (32b):
Figure imgf000100_0003
Title compound was synthesized by following the procedure as described in Step 2 of Example 30, using intermediate (32a) from the previous step.1H-NMR (400 MHz, CDCI3): δ 8.74 (d, J= 8.8 Hz, 1H), 8.60 (d, J= 9.6 Hz, 1H), 8.55 (s, 1H), 7.90 (d, J= 8.8 Hz, 1H), 7.81 (s, 1H), 7.78 (d, J= 9.6 Hz, 1H), 7.45 (s, 2H), 6.83 (s, 1H).
Step 3: 2-bromo-1-(7-bromophe c)
Figure imgf000101_0001
Title compound was synthesized by following the procedure as described in Step 3 of Example 30, using intermediate (32b) from the previous step.1H-NMR (400 MHz, CDCI3): δ 8.74 (d, J = 8.8 Hz, 1 H), 8.60 (d, J = 8.4 Hz, 1 H), 8.52 (s, 1 H), 8.26 (d, J = 8.8 Hz, 1 H), 8.09 (s, 1H), 7.85 (d, J= 8.4 Hz, 1H), 7.58-7.51 (m, 2H), 4.89 (s, 2H).
Step 4: (1S,2S)-2-(7-bromophenanthren-2-yl)-2-oxoethyl 2-((tert- butoxycarbonyl)amino)cyclopentanecarboxylate (32d):
Figure imgf000101_0002
Title compound was synthesized by following the procedure as described in Step 4 of Example 30, using intermediate (32c) from the previous step.1H-NMR (400 MHz, CDCI3): δ 8.62-8.49 (m, 2H), 8.28 (s, 1H), 7.81-7.28 (m, 4H), 7.42-7.31 (m, 1H), 5.52-5.37 (m, 2H), 4.96 (bs, 1H), 4.32-4.27 (m,1H), 2.92-2.86 (m, 1H), 2.32-1.76 (m, 6H), 1.38 (s, 9H). m/z 527.5 (M++1).
Step 5: tert-butyl ((1S,2S)-2-(5-(7-bromophenanthren-2-yl)-1 H-imidazol-2- yl)cyclopentyl)carbamate (32e):
Figure imgf000101_0003
Title compound was synthesized by following the procedure as described in Step 5 of Example 30, using intermediate (32d) from the previous step. 1 H-NMR (400 MHz, CDCI3): δ 8.63-8.51 (m, 2H), 8.28 (s, 1 H), 7.81 -7.26 (m, 4H), 7.42-7.31 (m, 2H), 4.98-4.96 (bs, 1 H), 4.29-4.22 (m, 1 H), 3.29-3.209 (m, 1 H), 2.88-1 .61 (m, 6H), 1 .38 (s, 9H). m/z 507.5 (M++1 ).
Step 6: tert-butyl ((1 S,2S)-2-(5-(7-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenanthren-2-yl)-1 H-i )
Figure imgf000102_0001
Title compound was synthesized by following the procedure as described in Step 6 of Example 30, using intermediate (32e) from the previous step. 1 H-NMR (400 MHz, CDCI3): δ 8.71 -8.64 (m, 2H), 8.37 (s, 1 H), 8.02 (s, 1 H), 7.80-7.56 (m, 2H), 7.45-7.35 (m, 2H), 4.29- 4.21 (m, 1 H), 3.29-3.19 (m, 1 H), 2.87-1 .71 (m, 6H), 1 .38 (s, 9H), 1 .27 (s, 12H), m/z 553.7 (M++1 ).
Step 7: (S)-tert-butyl 2-(5-(7-(2-((1 S,2S)-2-((tert-butoxycarbonyl)amino)cyclopentyl)-1 H- imidazol-5-yl)phenanthren-2-yl)-1 -((2-(trimethylsilyl)ethoxy)methyl)-1 H-imidazol-2- yl)pyrrolidine-1 -carboxyla
Figure imgf000102_0002
Title compound was synthesized by following procedure as described in Step 7 of Example 30, using intermediate (32f) from the previous step, m/z 795.3 (M++1 ).
Step 8: methyl ((S)-1 -(((1 S,2S)-2-(5-(7-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenanthren-2-yl)-1 H-imidazol-2- yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate (Compound 32):
Figure imgf000103_0001
Title compound was synthesized by following the procedure as described in Step 8 of Example 30, using intermediate (32g) from the previous step. 10.99-10.86 (m, 1 H), 10.61 - 10.56 (m, 1 H), 8.65-8.58 (m, 2H), 8.28 (s, 1 H), 8.12-8.09 (m, 1 H), 7.78-7.71 (m, 4H), 7.57- 7.49 (m, 1 H), 7.41 -7.38 (m, 1 H), 5.49-5.41 (m, 1 H), 4.37-4.29 (m, 2H), 3.99-3.86 (m, 2H), 3.84-3.78 (m, 1 H), 3.68 (s, 6H), 3.21 -3.18 (m, 2H), 2.67-2.14 (m, 5H), 1 .86-1 .63 (m, 7H), 0.91 (d, 12H), m/z 777.2 (M++1 ), m.p.: 153-155 eC.
Example 33: Biological Activity
Anti-viral activity of the compounds of the invention was monitored using an HCV replicon assay. The Huh7.5/ Con1/SG-Neo(l)hRluc2aUb cell line persistently expressing a bicistronic genotype 1 b replicon in Huh 7.5 cells was obtained from Apath LLC. This cell line was used to test inhibition of genotype 1 b replicon levels by test compound using Renilla luciferase enzyme activity readout as a measure of viral replication efficiency. Briefly, 7000 cells were seeded in 96 well black clear bottom plates and allowed to adhere overnight. The next day each compound was added in triplicate to the cells at the desired concentration with a final DMSO concentration of 0.5%. Cells in media alone and cells incubated without drug with 0.5% DMSO served as controls. The plates were incubated for 72h at 37°C prior to running the luciferase assay. Enzyme activity was measured using Renilla-Glo Luciferase Assay kit from Promega as per the manufacturer's instructions. The following equation was used to generate the percent inhibition value for each test concentration.
Average Control (cells alone +0.5% DMSO) - Average compound value(cells + drug)
% Inhibition = X 100
Average Control (cells alone+0.5% DMSO) The EC5o value was determined using GraphPad Prism and the following equation:
Y=Bottom + (Top-Bottom)/(1 +10A((LoglC5o-X)*HillSlope))
EC50 values/% inhibitions of compounds were determined 2-3 times in the replicon assay.
Following table 1 shows EC50 values, for inhibition of genotype 1 b replicon, of the compounds of the invention. Group A compounds exhibited EC50 value between 1 pM to 999 pM, Group B exhibited EC50 value between 1 nM to 999 nM, and Goup C exhibited EC50 value of more than 1 μΜ.
Table 1 :
Figure imgf000104_0001

Claims

1. A compound of formula I, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its sulfoxides, its N- oxides, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its cl
Figure imgf000105_0001
wherein, in the compound of formula I,
Ring 'A' is a saturated carbocycle, which may be a monocyclic system or may be a fused carbocycle or may be a bridged carbocycle, the said ring A may contain 5 to
10 carbons;
Ring 'D' is selected from 5 to 10 membered carbocycle and 5 to 10 membered heterocycle, the ring 'D' may be monocyclic, fused bicyclic, bridged bicyclic or spiro bicyclic; Y is selected from -CH(R2a)- and -N(R2)-;
R1 is selected from the group consisting of R13aC(=0)N(R14)-, R13aOC(=0)N(R14)-, R13(R14)N-, R13(R14)NC(=0)N(R15)-, R13aS02N(R14)-,
R13aOC(=0)N(R14)CRa(Rb)C(=0)N(R15)-,
R13aOC(=0)N(R14)CRa(Rb)C(Rc)(Rd)C(=0)N(R15)-,
R13(R14a)NC(=0)N(R14)CRa(Rb)C(=0)N(R15)-, and
R13(R14a)NC(=0)N(R14)CRa(Rb)C(Rc)(Rd)C(=0)N(R15)-;
R1a is selected independently at each occurrence from the group consisting of halogen, substituted- or unsubstituted- Ci-6 alkyl, R13aC(=0)-, R13bO-, R13aOC(=0)-, R13aC(=0)0-, and R13(R14)NC(=0)-, R2 is selected independently at each occurrence from the group consisting of substituted- or unsubstituted- Ci-6 alkyl, R13aC(=0)-, R13aS02-, R13aOC(=0)-, R13(R14)NC(=0)-, R13aOC(=0)N(R14)C(Ra)(Rb)C(=0)-,
l R13aOC(=0)N(R14)C(Ra)(Rb)C(Rc)(Rd)C(=0)-,
R13(R14)NC(=0)N(R15)C(Ra)(Rb)C(=0)-,
R13(R14)NC(=0)N(R15)C(Ra)(Rb)C(Rc)(Rd)C(=0)-, R13S02N(R14)C(Ra)(Rb)C(=0)-, R13S02N(R14)C(Ra)(Rb)C(Rc)(Rd)C(=0)-, and R13aOC(=0)N(R14)C(Ra)(Rb)S02-;
R2a is selected independently at each occurrence from the group consisting of R13aC(=0)N(R14)-, R13aOC(=0)N(R14)-, R13(R14)N-, R13(R14)NC(=0)N(R15)-, R13aS02N(R14)-, R13aOC(=0)N(R14)C(Ra)(Rb)C(=0)N(R15)-, R13aOC(=0)N(R14)C(Ra)(Rb)C(Rc)(Rd)C(=0)N(R15)-,
R13(R14a)NC(=0)N(R14)C(Ra)(Rb)C(=0)N(R15)-, and R13(R14a)NC(=0)N(R14)CRa(Rb)C(Rc)(Rd)C(=0)N(R15)-;
R3 is independently selected from O and N(R16);
R4 is selected independently at each occurrence from CRe(Rf), O and N(R14); such that, when n = 2 and R4 is selected as CRe(Rf) for both the occurrences, two Res together can form a bond to form a alkenylene linkage or two Res and two Rfs together can form bonds to form alkynylene linkage;
R5, R6, R7, R8, R9 and R10 are independently selected as hydrogen, or R5 and R6 together, R7 and R10 together, or R8 and R9 together independently along with the carbon atoms to which they are attached forming 5 to 8 membered substituted- or unsubstituted- carbocycle, 5 to 8 membered substituted- or unsubstituted- heterocycle, 6 membered substituted- or unsubstituted- aryl, or 5 to 6 membered substituted- or unsubstituted- heteroaryl; with a proviso that the compound of formula I must have at least one cyclic system formed out of either R5 and R6, R8 and R9, or R7 and R10; also provided that R7 and R10 take part in formation of cyclic system only when n is 0;
R11 and R12 are independently selected from a group consisting of hydrogen, halogen, substituted- or unsubstituted- Ci-6 alkyl, R13bO-, and (R13)(R14)N-; wherein, R13, R14, R14a and R15 are independently selected from hydrogen, substituted- or unsubstituted- Ci-6 alkyl, substituted- or unsubstituted- aryl, substituted- or unsubstituted- heteroaryl, substituted- or unsubstituted- cycloalkyl, and substituted- or unsubstituted- heterocyclyl;
2 R13a is selected from substituted- or unsubstituted- Ci-6 alkyl, perhaloalkyl, substituted- or unsubstituted- aryl, substituted- or unsubstituted- heteroaryl, substituted- or unsubstituted- cycloalkyi, and substituted- or unsubstituted- heterocyclyl;
R13b is selected from hydrogen, substituted- or unsubstituted- Ci-6 alkyl, perhaloalkyl, substituted- or unsubstituted- aryl, substituted- or unsubstituted- heteroaryl, substituted- or unsubstituted- cycloalkyi, and substituted- or unsubstituted- heterocyclyl;
R16 is selected from hydrogen or substituted- or unsubstituted alkyl group;
Ra, Rb, Rc and Rd, are independently selected from hydrogen, halogen, substituted- or unsubstituted- Ci-6 alkyl, substituted- or unsubstituted- aryl, substituted- or unsubstituted- heteroaryl, substituted- or unsubstituted- cycloalkyi, and substituted- or unsubstituted- heterocyclyl, or Ra, Rb, Rc and Rd together with the carbon atom(s) to which they are attached forming substituted- or unsubstituted- carbocycle, substituted- or unsubstituted- heterocycle; m is an integer ranging between 0 to 2, selected independently at each occurrence; n is an integer ranging between 0 and 2;
'alkyl' may be substituted with 1 to 4 substituents selected from the group consisting of halogen, oxo, Ci-6 alkyl, haloalkyl, aryl, heteroaryl, cycloalkyi, and heterocyclyl, R17aC(=0)-, R17aS02-, R17bO-, R17aOC(=0)-, R17aC(=0)0-, (R17)(R18)NC(=0)-, (R17a)C(=0)N(R18)-, (R17)(R18)N-, (R17)(R18)NC(=0)N(R19)-, and R17aS02N(R18)-;
'cycloalkyi', 'cycloalkenyl' and 'carbocycle' may be substituted with 1 to 2 substituents selected from the group consisting of halogen, oxo, Ci-6 alkyl, haloalkyl, R17aC(=0)-, R17aS02-, R17bO-, R17aOC(=0)-, R17aC(=0)0-, (R17)(R18)NC(=0)-, (R17a)C(=0)N(R18)-, (R17)(R18)N-, (R17)(R18)NC(=0)N(R19)-, and R17aS02N(R18)-;
'aryl' may be substituted with 1 to 2 substituents selected from the group consisting of halogen, nitro, cyano, hydroxy, alkyl, perhaloalkyl, alkyl-O-, perhaloalkyl-O-, alkyl(H)N-, alkyl(alkyl)N-, H2N-, alkyl-S02-, alkyl-C(=0)(H)N-, alkyl-C(=0)(alkyl)N-, alkyl(H)NC(=0)-, alkyl(alkyl)NC(=0)-, H2NC(=0)-, alkyl(H)NS02-, alkyl(alkyl)NS02-,
Figure imgf000108_0001
'heteroaryl' may be substituted with 1 to 2 substituents selected from the group consisting of halogen, nitro, cyano, hydroxy, alkyl, perhaloalkyl, alkyl-O-, perhaloalkyl-O-, alkyl-S02-, H2N-, alkyl(H)N-, alkyl(alkyl)N-, alkyl-C(=0)(H)N-, alkyl- C(=0)(alkyl)N-, NH2C(=0)-, alkyl(H)NC(=0)-, alkyl(alkyl)NC(=0)-, NH2S02-, alkyl(H)NS02-, and alkyl(alkyl)NS02-; ring carbon(s) of 'heterocycle' may be substituted with 1 to 2 substituents selected from the group consisting of halogen, nitro, cyano, oxo, alkyl, R17bO-, R17aOC(=0)-, R17aC(=0)0-, R17(H)NC(=0)-, R17(alkyl)NC(=0)-, R17(H)N-, R17(alkyl)N-, R17aC(=0)(H)N-, R17(H)NC(=0)(H)N-, and R17(alkyl)NC(=0)(H)N-; the substituents on ring nitrogen(s) of 'heterocycle' are selected from the group consisting of alkyl, R17aS02-, R17aC(=0)-, R17aOC(=0)-, R17(H)NC(=0)-, and R17(alkyl)NC(=0);
R17, R18 and R19 are independently selected from hydrogen and alkyl;
R17a is selected from alkyl and perhaloalkyl;
R17b is selected from the group consisting of hydrogen, alkyl, and perhaloalkyl.
2. The compound of formula I, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its sulfoxides, its N-oxides, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates or its co-crystals, as claimed in claim 1 , wherein ring A is selected as cyclopentane.
The compound of formula I, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its sulfoxides, its N-oxides, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates or its co-crystals, as claimed in claim 1 or 2, wherein ring Y is selected as -N(R2)-.
The compound of formula I, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its sulfoxides, its
4 N-oxides, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates or its co-crystals, as claimed in any one of claims 1 -3, wherein R1 is selected as R13aOC(=0)N(R14)CRa(Rb)C(=0)N(R15)-.
5. The compound of formula I, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its sulfoxides, its N-oxides, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates or its co-crystals, as claimed in any one of claims 1 -4, wherein m is selected as 0 at all the occurrences.
6. The compound of formula I, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its sulfoxides, its N-oxides, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates or its co-crystals, as claimed in any one of claims 1 -5, wherein R2 is selected as R13aOC(=0)N(R14)CRa(Rb)C(=0)-.
7. The compound of formula I, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its sulfoxides, its N-oxides, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates or its co-crystals, as claimed in any one of claims 1 -6, wherein R3 is selected from NH and O.
8. The compound of formula I, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its sulfoxides, its N-oxides, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates or its co-crystals, as claimed in any one of claims 1 -7, wherein n is selected as 0.
9. The compound of formula I, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its sulfoxides, its N-oxides, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates or its co-crystals, as claimed in any one of claims 1 -8, wherein R5 and R6 are independently selected from hydrogen and halogen, or R5 and R6 together with the carbon atoms to which they are attached form a six membered carbocycle.
10. The compound of formula I, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its sulfoxides, its N-oxides, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates or its co-crystals, as claimed in any one of claims 1 -9, wherein R7 and R10 are selected as hydrogen, or R7 and R10 together with the carbon atoms to which they are attached form a five or six membered carbocycle, the said carbocycle is unsubstituted or substituted with one or two alkyl groups.
11. The compound of formula I, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its sulfoxides, its N-oxides, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates or its co-crystals, as claimed in any one of claims 1 -
10, wherein R9 and R8 are selected as hydrogen, or R9 and R8 together with the carbon atoms to which they are attached form a six or seven membered carbocycle, or R9 and R8 together with the carbon atoms to which they are attached form a seven membered heterocycle containing one heteroatom.
12. The compound of formula I, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its sulfoxides, its N-oxides, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates or its co-crystals, as claimed in any one of claims 1 -
1 1 , wherein R11 and R12 are selected as hydrogen.
13. The compound of formula I, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its sulfoxides, its N-oxides, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates or its co-crystals, as claimed in any one of claims 1 -
12, wherein ring D is selected as
Figure imgf000111_0001
The compound of formula I, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its sulfoxides, its N-oxides, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates or its co-crystals, as claimed in any on -
13, wherein ring A is selected as cyclopentane, ring D is selected as
Figure imgf000111_0002
R1 is selected as R13aOC(=0)N(R14)CRa(Rb)C(=0)N(R15)-, m is selected as 0 at all the occurrences, R3 is selected from NH and O, n is selected as 0, R5 and R6 are independently selected from hydrogen and halogen, or R5 and R6 together with the carbon atoms to which they are attached form a six membered carbocycle, R7 and R10 are selected as hydrogen, or R7 and R10 together with the carbon atoms to which they are attached form a five or six membered carbocycle, the said carbocycle is unsubstituted or substituted with one or two alkyl groups, R9 and R8 are selected as hydrogen, or R9 and R8 together with the carbon atoms to which they are attached form a six or seven membered carbocycle, or R9 and R8 together with the carbon atoms to which they are attached form a seven membered heterocycle containing one heteroatom, R11 and R12 are selected as hydrogen; such that at least one cyclic system is formed out of R5 and R6, R8 and R9, or R7 and R10.
The compound of formula I, its tautomeric forms, its stereoisomers, its analogues, its prodrugs, its isotopically substituted analogues, its metabolites, its sulfoxides, its N-oxides, its pharmaceutically acceptable salts, its polymorphs, its solvates, its optical isomers, its clathrates or its co-crystals, as claimed in any one of claims 1 -
14, wherein the compound is selected from the group consisting of:
methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate; methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 - oxobutan-2-yl)carbamate; methyl ((2S)-1 -((2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate; methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate; methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 - oxobutan-2-yl)carbamate; methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 - oxobutan-2-yl)carbamate; methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 - oxobutan-2-yl)carbamate; methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 - oxobutan-2-yl)carbamate;
8 methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 - oxobutan-2-yl)carbamate; methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate; methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 - oxobutan-2-yl)carbamate; methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 ,4,5,6-tetrahydrobenzo[3,4]cyclohepta[1 ,2- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 - oxobutan-2-yl)carbamate; methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-benzo[2,3]oxepino[4,5- d]imidazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 - oxobutan-2-yl)carbamate; methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate; methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate; methyl ((S)-1 -(((1 R,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate; methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate; methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate; methyl ((S)-1 -(((1 S,2R)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-4- chloro-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate; methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-naphtho[1 ,2-d]imidazol-7-yl)phenyl)-4- chloro-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate; methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-5,6-dihydro-4H-benzo[3,4]cyclohepta[1 ,2- d]oxazol-8-yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 - oxobutan-2-yl)carbamate; methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydrobenzo[2,3]oxepino[4,5-d]oxazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate;
10 methyl ((S)-1 -(((1 R,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydrobenzo[2,3]oxepino[4,5-d]oxazol-8- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate; methyl ((S)-1 -(((1 S,2S)-2-(7-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate; methyl ((S)-1 -(((1 S,2R)-2-(7-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)-4,5-dihydro-1 H- naphtho[1 ,2-d]imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate; methyl ((S)-1 -(((1 S,2R)-2-(7-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2- d]imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate; methyl ((S)-1 -(((1 S,2S)-2-(7-(4-(2-((S)-1 -((S)-2-amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenyl)-1 H-naphtho[1 ,2- d]imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate; methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)(ethyl)amino)-3-methyl-1 -oxobutan-2- yl)carbamate; methyl ((S)-1 -(((1 S,2S)-2-(5-(4-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-4,5-dihydro-1 H-naphtho[1 ,2-d]imidazol-7- yl)phenyl)-1 H-imidazol-2-yl)cyclopentyl)(methyl)amino)-3-methyl-1 -oxobutan- 2-yl)carbamate;
11 methyl ((S)-1 -(((1 S,2S)-2-(5-(7-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)-9H-fluoren-2-yl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate; methyl ((S)-1 -(((1 S,2S)-2-(5-(7-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)-9,9-dimethyl-9H-fluoren-2- yl)-1 H-imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate; and methyl ((S)-1 -(((1 S,2S)-2-(5-(7-(2-((S)-1 -((S)-2-(methoxycarbonyl)amino-3- methylbutanoyl)pyrrolidin-2-yl)-1 H-imidazol-5-yl)phenanthren-2-yl)-1 H- imidazol-2-yl)cyclopentyl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate.
16. A pharmaceutical composition comprising a compound or a combination of compounds according to any one of claims 1 -15 or a pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable carrier or excipient.
17. A method of inhibiting the replication of an RNA-containing virus comprising contacting said virus with a therapeutically effective amount of a compound or combination of compounds of any one of claims 1 -15, or a pharmaceutically acceptable salt thereof.
18. A method of treating or preventing infection caused by an RNA-containing virus comprising administering to a patient in need of such treatment a therapeutically effective amount of a compound or combination of compounds of any one of claims 1 -15, or a pharmaceutically acceptable salt thereof.
19. The method of claim 18, wherein the RNA-containing virus is hepatitis C virus.
20. The method of claim 18, further comprising the step of co-administering one or more agents selected from the group consisting of a host immune modulator and an antiviral agent, or a combination thereof.
21. The method of claim 20, wherein the host immune modulator is selected from the group consisting of interferon-alpha, pegylated-interferon-alpha, interferon-beta, interferon-gamma, consensus interferon, a cytokine, and a vaccine.
22. The method of claim 20, wherein the antiviral agent inhibits replication of HCV by inhibiting host cellular functions associated with viral replication.
12
23. The method of claim 20, wherein the antiviral agent inhibits the replication of HCV by targeting proteins of the viral genome.
24. The method of claim 20, wherein said antiviral agent is an inhibitor of a HCV viral protein, a replication process or a combination thereof, wherein said targeting protein or replication process is selected from the group consisting of helicase, protease, polymerase, metalloprotease, NS4A, NS4B, NS5A, assembly, entry, and IRES.
25. The method of claim 18, further comprising the step of co-administering an agent or combination of agents that treat or alleviate symptoms of HCV infection selected from cirrhosis and inflammation of the liver.
26. The method of claim 18, further comprising the step of co-administering one or more agents that treat patients for disease caused by hepatitis B (HBV) infection.
27. The method of claim 18, further comprising the step of co-administering one or more agents that treat patients for disease caused by human immunodeficiency virus (HIV) infection.
28. The pharmaceutical composition of claim 16, further comprising an agent selected from interferon, pegylated interferon, ribavirin, amantadine, an HCV protease inhibitor, an HCV polymerase inhibitor, an HCV helicase inhibitor, or an internal ribosome entry site inhibitor.
29. The composition of claim 16, further comprising a cytochrome P450 monooxygenase inhibitor or a pharmaceutically acceptable salt thereof.
30. A method of treating hepatitis C infection in a subject in need thereof comprising coadministering to said subject a cytochrome P450 monooxygenase inhibitor or a pharmaceutically acceptable salt thereof, and a compound of any one of claims 1 - 15 or a pharmaceutically acceptable salt thereof
13
PCT/IB2012/054020 2011-08-08 2012-08-07 Imidazole derivatives as antiviral agents WO2013021344A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN1046KO2011 2011-08-08
IN1046/KOL/2011 2011-08-08

Publications (1)

Publication Number Publication Date
WO2013021344A1 true WO2013021344A1 (en) 2013-02-14

Family

ID=46845794

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2012/054020 WO2013021344A1 (en) 2011-08-08 2012-08-07 Imidazole derivatives as antiviral agents

Country Status (1)

Country Link
WO (1) WO2013021344A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104513147A (en) * 2014-11-20 2015-04-15 上海众强药业有限公司 Preparation method of fluorene ethyl ketone derivative
US9326973B2 (en) 2012-01-13 2016-05-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9717712B2 (en) 2013-07-02 2017-08-01 Bristol-Myers Squibb Company Combinations comprising tricyclohexadecahexaene derivatives for use in the treatment of hepatitis C virus
US9770439B2 (en) 2013-07-02 2017-09-26 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9775831B2 (en) 2013-07-17 2017-10-03 Bristol-Myers Squibb Company Combinations comprising biphenyl derivatives for use in the treatment of HCV
US10617675B2 (en) 2015-08-06 2020-04-14 Bristol-Myers Squibb Company Hepatitis C virus inhibitors

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5019369A (en) 1984-10-22 1991-05-28 Vestar, Inc. Method of targeting tumors in humans
WO2009102633A1 (en) 2008-02-13 2009-08-20 Bristol-Myers Squibb Company Hepatitis c virus inhibitors
WO2009102318A1 (en) 2008-02-12 2009-08-20 Bristol-Myers Squibb Company Hepatitis c virus inhibitors
WO2009102568A1 (en) * 2008-02-13 2009-08-20 Bristol-Myers Squibb Company Conformationally restricted biphenyl derivatives for use as hepatitis c virus inhibitors
WO2010096302A1 (en) 2009-02-17 2010-08-26 Bristol-Myers Squibb Company Hepatitis c virus inhibitors
WO2010099527A1 (en) 2009-02-27 2010-09-02 Enanta Pharmaceuticals, Inc. Hepatitis c virus inhibitors
WO2010117635A1 (en) 2009-03-30 2010-10-14 Bristol-Myers Squibb Company Hepatitis c virus inhibitors
WO2010132601A1 (en) 2009-05-13 2010-11-18 Gilead Sciences, Inc. Antiviral compounds
WO2010132538A1 (en) 2009-05-12 2010-11-18 Schering Corporation Fused tricyclic aryl compounds useful for the treatment of viral diseases

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US5019369A (en) 1984-10-22 1991-05-28 Vestar, Inc. Method of targeting tumors in humans
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
WO2009102318A1 (en) 2008-02-12 2009-08-20 Bristol-Myers Squibb Company Hepatitis c virus inhibitors
WO2009102633A1 (en) 2008-02-13 2009-08-20 Bristol-Myers Squibb Company Hepatitis c virus inhibitors
WO2009102568A1 (en) * 2008-02-13 2009-08-20 Bristol-Myers Squibb Company Conformationally restricted biphenyl derivatives for use as hepatitis c virus inhibitors
WO2010096302A1 (en) 2009-02-17 2010-08-26 Bristol-Myers Squibb Company Hepatitis c virus inhibitors
WO2010099527A1 (en) 2009-02-27 2010-09-02 Enanta Pharmaceuticals, Inc. Hepatitis c virus inhibitors
WO2010117635A1 (en) 2009-03-30 2010-10-14 Bristol-Myers Squibb Company Hepatitis c virus inhibitors
WO2010132538A1 (en) 2009-05-12 2010-11-18 Schering Corporation Fused tricyclic aryl compounds useful for the treatment of viral diseases
WO2010132601A1 (en) 2009-05-13 2010-11-18 Gilead Sciences, Inc. Antiviral compounds

Non-Patent Citations (26)

* Cited by examiner, † Cited by third party
Title
"Pharmaceutics and Pharmacy Practice", 1982, J.B. LIPPINCOTT COMPANY, pages: 238 - 250
"Physicians' Desk Reference", 2004
"Remington's Pharmaceutical Science", 1985, MACK PUBLISHING COMPANY
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING COMPANY, pages: 1445
BARNES E., WHO FACTSHEET, 2010, Retrieved from the Internet <URL:http://www.who.int/vaccine_research/diseases/virai_cancers/en/index2.htmi>
BERGE S.M. ET AL.: "Pharmaceutical Salts", JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
FRIED MW ET AL., N ENGL J MED., vol. 347, 2002, pages 975 - 982
HOOFNAGLE JH., HEPATOLOGY, vol. 26, 1997, pages 15S - 20
J. AM. CHEM. SOC., vol. 121, 1999, pages 7574 - 7581
J. MED. CHEM., vol. 43, 2000, pages 2049 - 2063
J. MED. CHEM., vol. 48, 2005, pages 7351 - 7362
J. ORG. CHEM., vol. 66, no. 16, 2001, pages 5629 - 5632
JESUDIAN AB; GAMBARIN-GELWAN M; JACOBSON IM., GASTROENTEROLOGY HEPATOL., vol. 8, 2012, pages 91 - 101
JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 2 - 19
MORIISHI K ET AL., ANTIVIR. CHEM. CHEMOTHER., vol. 14, 2003, pages 285 - 297
P.H.EINRICH; STAHLAND; CAMILLE; G.WERMUTH: "handbook of pharmaceutical salts properties, selection, and use", 2002, WILEY- VCH
PHYSICIANS' DESK REFERENCE, 2004
SZABO E; LOTZ G ET AL., PATHOL. ONCOL. RES., vol. 9, 2003, pages 215 - 221
SZOKA ET AL., ANN. REV. BIOPHYS. BIOENG., vol. 9, 1980, pages 467
TET. ASYMM., vol. 19, 2008, pages 2796 - 2803
TET. LETT., vol. 43, 2002, pages 5401 - 5404
TETRAHEDRON, vol. 56, 2000, pages 5225 - 5240
TETRAHEDRON: ASYMMETRY, vol. 19, 2008, pages 2796 - 2803
THOMSON BJ ET AL., CLIN MICROBIAL INFECT., vol. 11, 2005, pages 86 - 94
TOISSEL: "ASHP Handbook on Injectable Drugs", 1986, pages: 622 - 630
WASSERMAN ET AL., CANCER, vol. 36, 1975, pages 1258 - 1268

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9326973B2 (en) 2012-01-13 2016-05-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9717712B2 (en) 2013-07-02 2017-08-01 Bristol-Myers Squibb Company Combinations comprising tricyclohexadecahexaene derivatives for use in the treatment of hepatitis C virus
US9770439B2 (en) 2013-07-02 2017-09-26 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9775831B2 (en) 2013-07-17 2017-10-03 Bristol-Myers Squibb Company Combinations comprising biphenyl derivatives for use in the treatment of HCV
CN104513147A (en) * 2014-11-20 2015-04-15 上海众强药业有限公司 Preparation method of fluorene ethyl ketone derivative
US10617675B2 (en) 2015-08-06 2020-04-14 Bristol-Myers Squibb Company Hepatitis C virus inhibitors

Similar Documents

Publication Publication Date Title
AU2013217229B2 (en) Antiviral compounds with a heterotricycle moiety
WO2013021337A1 (en) Antiviral compounds with a fused tricyclic ring
US8785487B2 (en) Hepatitis C virus inhibitors
WO2013021344A1 (en) Imidazole derivatives as antiviral agents
WO2012021704A1 (en) Hepatitis c virus inhibitors
EP2545051B1 (en) Compounds for the treatment of hepatitis c
KR20140048848A (en) Hepatitis c virus inhibitors
US8933110B2 (en) Hepatitis C virus inhibitors
EP2768517A2 (en) Hepatitis c virus inhibitors
KR20140037114A (en) Hepatitis c virus inhibitors
WO2013030750A1 (en) Antiviral compounds
CN106008552B (en) Benzo [ e ] pyrazolo [1,5-c ] [1,3] oxazine compound and application thereof
AU2020322598A1 (en) Dihydropyrimidine derivatives and uses thereof in the treatment of HBV infection or of HBV-induced diseases
CN105985355B (en) Fused tricyclic hepatitis virus inhibitor and application thereof
AU2020321067A1 (en) Dihydropyrimidine derivatives and uses thereof in the treatment of HBV infection or of HBV-induced diseases
EP3604295A1 (en) Silicon-containing compound for resistance to hepatitis c virus infection

Legal Events

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

Ref document number: 12758902

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12758902

Country of ref document: EP

Kind code of ref document: A1