WO2013036994A1 - Compounds for the treatment of hcv - Google Patents

Compounds for the treatment of hcv Download PDF

Info

Publication number
WO2013036994A1
WO2013036994A1 PCT/AU2012/001091 AU2012001091W WO2013036994A1 WO 2013036994 A1 WO2013036994 A1 WO 2013036994A1 AU 2012001091 W AU2012001091 W AU 2012001091W WO 2013036994 A1 WO2013036994 A1 WO 2013036994A1
Authority
WO
WIPO (PCT)
Prior art keywords
cyclohexyl
imidazo
methyl
optionally substituted
amino
Prior art date
Application number
PCT/AU2012/001091
Other languages
French (fr)
Inventor
Barbara Frey
Richard Hufton
Michael Harding
Alistair George Draffan
Original Assignee
Biota Scientific Management Pty Ltd
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 Biota Scientific Management Pty Ltd filed Critical Biota Scientific Management Pty Ltd
Publication of WO2013036994A1 publication Critical patent/WO2013036994A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41881,3-Diazoles condensed with other heterocyclic ring systems, e.g. biotin, sorbinil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • 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
    • C07D498/12Heterocyclic 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 three hetero rings
    • C07D498/14Ortho-condensed systems

Definitions

  • the present invention relates to viral polymerase inhibitors, in particular inhibitors of viral polymerases within the Flaviviridae family such as hepatitis C virus (HCV), processes for their preparation and their use in the treatment of Flaviviridae viral infections such as Hepatitis C virus (HCV) infections.
  • HCV hepatitis C virus
  • the Flaviviridae are a group of positive single-stranded RNA viruses with a genome size from 9-15 kb.
  • the Flaviviridae consist of various genera including: Hepaciviruses (this genus contains only one species, the Hepatitis C virus (HCV), which is composed of many genotypes and subtypes); Flaviviruses (this genus includes the Dengue virus, Japanese Tick-Borne and the Yellow Fever virus and there are some additional Flaviviruses that are unclassified) and Pestiviruses (this genus includes three serotypes of bovine viral diarrhoea virus, but no known human pathogens).
  • HCV Hepatitis C virus
  • Hepatitis C virus is a major cause of viral hepatitis and has infected more than 200 million people worldwide.
  • Hepatitis C virus has a positive-strand RNA genome enclosed in a nucleocapsid and lipid envelope.
  • the HCV genome is approximately 9.6 kb in length and encodes a polyprotein of about 3,000 amino acids.
  • genotypes 1 a and 1 b account for about 75 % of cases, and genotypes 2 and 3 for 10-20 % of cases. Significant differences are observed in the geographic distribution of HCV genotypes. For example, in Europe genotypes 2 and 3 comprise up to one half of cases whereas genotype 3 is thought to dominate in India.
  • varied genotype are observed in the geographic distribution of HCV genotypes.
  • HCV chronic bloodborne infection
  • HCV pegylated interferon
  • SVR sustained viral response
  • patients with HCV genotype 1 remain the most difficult to treat, with SVR rates of approximately 40% after 48 weeks of therapy.
  • combination peg-IFN/ribavirin therapy is limited by serious side effects, including fatigue, influenza-like symptoms, depression and suicide with peg-I FN, and haemolytic anaemia with ribavirin.
  • peg-IFN/ribavirin therapy is contra-indicated in patients who have depression, anaemia, HCV-related decompensated cirrhosis, alcohol/substance abuse and autoimmune disorders or who are pregnant.
  • telaprevir Very short-acting antiviral
  • boceprevir boceprevir
  • Both drugs are protease inhibitors and are approved for the treatment of chronic HCV genotype 1 infection in combination with peg-IFN and ribavirin.
  • Pivotal phase 3 trials demonstrated that the addition of telaprevir or boceprevir to peg-IFN/RBV therapy achieved shortened durations of therapy and potent viral suppression, with SVR rates approaching 75% in genotype 1 treatment-naive patients and 30% to 85% in treatment-experienced patients.
  • telaprevir is associated with an increased incidence of rash and anaemia
  • boceprevir is associated with anaemia and dysgeusia.
  • Triple therapy with telaprevir or boceprevir and peg-IFN/ribavirin remains unsuitable for those intolerant to or with contraindications to peg-I FN/ribavirin therapy.
  • the HCV genome possesses structural (core) and non-structural (NS2, NS3, NS4A, NS4B, NS5A and NS5B) proteins.
  • the non-structural proteins are involved in viral genomic replication, with the initial synthesis of RNA carried out by NS5B RNA dependent RNA polymerase.
  • the NS5B protein is a key target for anti-HCV therapy, as it is essential for HCV replication and has no human host equivalent. This protein has been well characterised and is a validated target for drug discovery.
  • HCV therapy is also anticipated to evolve towards oral multidrug therapy, in which combinations of different DAA drugs with complementary mechanisms of action serve to increase viral suppression and delay or prevent the emergence of resistance.
  • HCV agents particularly with targeted mechanisms of action such as NS5B inhibitors.
  • HCV agents with cross-genotypic activity against genotypes 1 , 2 and 3.
  • the inventors have found a new class of antiviral compounds, more particularly NS5B polymerase inhibitors, for the treatment of HCV infections.
  • Compounds of the present invention are therefore considered to be useful in treating and preventing hepatitis C infections when used on their own or in combination with one or more other antiviral agents such as ribavirin, an antiviral nucleoside, polymerase inhibitor, protease inhibitor and/or inhibitor of viral entry, assembly or egress.
  • the combination may also additionally comprise at least one immunomodulatory agent for example an interferon or interferon derivative and/or an inhibitor of inosine-5'-monophosphate dehydrogenase (IMPDH).
  • IMPDH inosine-5'-monophosphate dehydrogenase
  • compounds of the invention will be efficacious in combination with at least one other DAA with a different mechanism of action and a complementary resistance profile (for example an NS5A inhibitor, a nucleoside or nucleotide NS5B inhibitor or a NS3/4A protease inhibitor) thereby offering an alternative treatment regime for patients not eligible for or treatable with the recently approved triple combination therapy.
  • a complementary resistance profile for example an NS5A inhibitor, a nucleoside or nucleotide NS5B inhibitor or a NS3/4A protease inhibitor
  • R 1 is H or R a or R 1 together with R 2 and the atoms to which they are attached form an optionally substituted 6-8 membered carbocyclic ring which may be optionally interrupted by one or more heteroatoms independently selected from O, N and S;
  • R 2 is H or R b or R 2 together with Ri joins to form an optionally substituted 6-8 membered carbocyclic ring which may be optionally interrupted by one or more heteroatoms independently selected from O, N and S;
  • R 3 is an optionally substituted C 3 - 8 cycloalkyl group
  • Xi, X 2 and X 3 are each independently selected from N, CH, C-R c and C-A-R 4 provided that at least one of Xi , X 2 and X 3 is C-A-R 4 ;
  • A is a covalent bond, -CONH-C(R 5 )(R 6 )-B- or -CONCi-aalkyl-CiRsXReJ-B-;
  • R 4 is selected from H, C0 2 H, optionally substituted C0 2 Ci_ 6 alkyl, optionally substituted aryl, optionally substituted arylCi_ 6 alkyl, optionally substituted arylC 2 . 6 alkenyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted heterocyclylC ⁇ alkyl, optionally substituted heteroarylC ⁇ alkyl, optionally substituted heterocyclylC 2 - 6 alkenyl and optionally substituted heteroarylC 2 - 6 alkenyl;
  • R 5 and R 6 are each independently selected from H, optionally substituted C ⁇ alkyl, optionally substituted aryl, optionally substituted C ⁇ alkylaryl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted C 1 . 6 alkylheterocyclyl and optionally substituted C ⁇ alkylheteroaryl or R 5 and R 6 together with the carbon atom to which they are attached form an optionally substituted C 3 . 6 cycloalkyl group;
  • B is a covalent bond or -CONH-R7- where R 7 is selected from optionally substituted aryl, optionally substituted arylCi -6 alkyl, optionally substituted arylC 2 . 6 alkenyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted
  • heterocyclylC ⁇ alkyl optionally substituted heteroarylC ⁇ alkyl, optionally substituted heterocyclylC 2 - 6 alkenyl and optionally substituted heteroarylC 2 - 6 alkenyl;
  • Z 1 is C or N
  • Z 2 , Z 3 , Z 4 and Z 5 are each independently selected from N, CH and C-R d ;
  • each R 8 is independently selected from H and C 1 . 6 alkyl and each alkyl, alkenyl, alkynyl, cycloalkyi, aryl, heterocyclyl and heteroaryl in the definition of R a , R b , R c and R d may be optionally substituted;
  • each heterocyclyl or heteroaryl has 1 , 2, 3 or 4 heteroatoms independently selected from O, S and N.
  • C-A-R 4 is not C-H.
  • ⁇ , X' 2 and X' 3 are each independently selected from N, CH, C-R c , C-A-R' 4 provided that at least one of ⁇ , X' 2 and X' 3 is C-A-R' 4 wherein R' 4 is C0 2 H or COaC ⁇ alkyl; and
  • Ri, R2, R3, R5, R6, R7, C-R c , Zi, Z 2 , Z 3 , Z 4 and Z 5 are as defined above.
  • the compounds of formula (I) are inhibitors of HCV.
  • the compounds of formula (I) inhibit RNA synthesis by the RNA dependent RNA polymerase of HCV (the NS5B protein encoded by HCV).
  • NS5B polymerase inhibitors have been clinically validated as potential antiviral agents for the treatment of HCV infection.
  • a pharmaceutical agent comprising the compound of formula (I) defined above and pharmaceutically acceptable salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
  • the pharmaceutical agent may be an antiviral agent.
  • a viral polymerase inhibitor in particular a HCV polymerase inhibitor such as a NS5B polymerase inhibitor comprising the compound of formula (I) defined above and pharmaceutically acceptable salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
  • the compound of formula (I ) and pharmaceutically acceptable salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof may be administered in the form of a pharmaceutical composition together with a pharmaceutically acceptable carrier.
  • a pharmaceutical composition comprising the compound of formula (I) and pharmaceutically acceptable salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition additionally comprises a therapeutically effective amount of one or more antiviral agents such as at least one other anti-HCV agent.
  • a method for the treatment of a Flaviviridae viral infection such as a HCV infection which comprises administering an effective amount of the compound of formula (I) and pharmaceutically acceptable salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof or the pharmaceutical composition defined above to a subject diagnosed with, suffering from or at risk of developing said viral infection.
  • a method of inhibiting the RNA-dependent RNA polymerase activity of the enzyme NS5B, encoded by HCV comprising exposing the enzyme NS5B to an effective amount of the compound of formula (I) defined above and pharmaceutically acceptable salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
  • a method of inhibiting HCV replication comprising exposing a cell infected with HCV to an effective amount of the compound of formula (I) defined above and pharmaceutically acceptable salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
  • the present invention relates to compounds of formula (I) which inhibit viral polymerases and are useful in the treatment of Flaviviridae viral infections, particularly, hepatitis C (HCV).
  • HCV hepatitis C
  • the present invention relates to compounds of formula I, salts, N-oxides, racemates, enantiomers and isomers thereof as defined above.
  • R 1 is H or d- 6 alkyl, preferably methyl, ethyl or propyl, more preferably H or methyl and R 2 is H or R b , preferably H.
  • R 1 together with R 2 joins to form an optionally substituted 6-8 membered carbocyclic ring which may be optionally interrupted by one or more heteroatoms independently selected from O, N and S.
  • R 1 together with R 2 joins to form a 7- membered carbocycle.
  • R 2 together are a divalent linking moiety selected from the group consisting of -CH 2 CH 2 CH 2 -; -OCH 2 CH 2 -; -CH 2 OCH 2 -; - CH 2 CH 2 0-; -NCH 2 CH 2 -; -CH 2 NCH 2 -; and -CH 2 CH 2 N-, thereby joining to form a 7- membered carbocyclic ring together with the atoms to which R 1 and R 2 are attached the carbocyclic ring being optionally interrupted by O or N, preferably O.
  • R 2 together are a divalent linking moiety of formula - CH 2 CH 2 0-.
  • R 3 is an optionally substituted C 3 . 6 cycloalkyl group, preferably cyclohexyl.
  • A is a covalent bond and R 4 is C0 2 H or optionally substituted C0 2 Ci_ 6 alkyl.
  • X 2 and X 3 are each independently selected from N, CH, C-R c or C-A-R 4 provided that at least one of Xi , X 2 and X 3 is C-A-R 4 and A is a covalent bond and R 4 is C0 2 H or optionally substituted C0 2 C 1 . 6 alkyl.
  • X 1 is C-A-R 4 ;
  • X 2 and X 3 are each independently selected from N, CH or C-R c preferably N or CH.
  • X 2 is C-A-R 4 and X 1 and X 3 are each independently selected from N, CH or C-R c preferably N or CH.
  • X 3 is C-A-R 4 and X 1 and X 2 are each independently selected from N, CH or C-R c preferably N or CH.
  • R 4 is selected from H, C0 2 H and optionally substituted CO ⁇ . 6 alkyl.
  • R 4 is C0 2 H or CC ⁇ C ⁇ al yl, preferably C0 2 H or CO ⁇ . 3 alkyl, more preferably R 4 is selected from C0 2 H, C0 2 CH 3 and C0 2 CH 2 CH 3 .
  • A is a covalent bond
  • A is -CONH-C(R 5 )(R 6 )-B- wherein R 5 and R 6 are each independently selected from H, C ⁇ alkyl (preferably C ⁇ alkyl, more preferably methyl), C ⁇ 6 alkylaryl (preferably optionally substituted benzyl); C ⁇ alkylheterocyclyl and Ci_
  • alkylheteroaryl preferably CH 2 heteroaryl, wherein the heteroaryl is preferably a 5-, 6- or 9- membered heteroaryl, more preferably a 5-, 6- or 9-membered N-containing heteroaryl
  • R 5 and R 6 together with the carbon atom to which they are attached form a cyclobutyl group.
  • B is a covalent bond
  • B is -CONHR 7 - wherein R 7 is selected from aryl (preferably phenyl), arylC ⁇ alkyl (preferably phenylC ⁇ alkyl) and arylC 2 - 6 alkenyl (preferably phenylC 2 _ 3 alkenyl).
  • R 7 is phenylethenyl.
  • the compound of formula (I) is a compound of formula (la)
  • Z 3 is CH or C-R d and Ri , R 2 , R 3 , R d , X 2 and X 3 are as previously defined and salts, A/-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
  • R d is selected from the group consisting of halo, OH, d_ 3 alkoxy or d_ 3 alkoxyaryl wherein the aryl is unsubstituted or substituted with heterocyclyl (preferably 5 or 6 membered heterocyclyl containing N such as morpholine or pyrrolidinone).
  • heterocyclyl preferably 5 or 6 membered heterocyclyl containing N such as morpholine or pyrrolidinone.
  • F, OH, OCH 3 and optionally substituted benzyloxy including unsubstituted benzyloxy and 2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1 -yl)benzyloxy are particularly preferred R d groups when Z 3 is C-R d .
  • R ⁇ R 2 , R3, Z 3 ⁇ , X 2 and X 3 are as previously defined and salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
  • A, R 2 , R3, R4, Z ⁇ X ⁇ , X2 and X 3 are as previously defined provided that A- R 4 is not H (i.e. where A is a covalent bond and R 4 is H) and salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof
  • C h alky refers to optionally substituted straight chain or branched chain hydrocarbon groups having from 1 to 6 carbon atoms. Examples include methyl (Me), ethyl (Et), propyl (Pr), isopropyl (/-Pr), butyl (Bu), isobutyl (/-Bu), sec-butyl (s-Bu), tert-butyl (f-Bu), pentyl, neopentyl, hexyl and the like. Unless the context requires otherwise, the term “d. 6 alkyl” also encompasses alkyl groups containing one less hydrogen atom such that the group is attached via two positions i.e. divalent. and "C 1 .
  • alkyl including methyl, ethyl, propyl, /sopropyl, n-butyl, / ' so-butyl, sec-butyl and ferf-butyl are preferred with methyl being particularly preferred.
  • C 2 - 6 alkenyl refers to optionally substituted straight chain or branched chain hydrocarbon groups having at least one double bond of either E or Z stereochemistry where applicable and 2 to 6 carbon atoms. Examples include vinyl, 1 -propenyl, 1- and 2- butenyl and 2-methyl-2-propenyl. Unless the context requires otherwise, the term "C 2 .
  • alkenyl also encompasses alkenyl groups containing one less hydrogen atom such that the group is attached via two positions i.e. divalent.
  • C 2 - 4 alkenyl and “C 2 _ 3 alkenyl” including ethenyl, propenyl and butenyl are preferred with ethenyl being particularly preferred.
  • C 2 - 6 alkynyl refers to optionally substituted straight chain or branched chain hydrocarbon groups having at least one triple bond and 2 to 6 carbon atoms.
  • C 2 - 6 alkynyl also encompasses alkynyl groups containing one less hydrogen atom such that the group is attached via two positions i.e. divalent.
  • C 2 . 3 alkynyl is preferred.
  • C 3 . 8 cycloalkyl refers to non-aromatic cyclic groups having from 3 to 8 carbon atoms, including cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl. It will be understood that cycloalkyl groups may be saturated such as cyclohexyl or unsaturated such as cyclohexenyl.
  • C 3 . 6 cycloalkyl such as cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl are preferred.
  • hydroxy and "hydroxyl” refer to the group -OH.
  • Ci_ 6 alkoxy refers to an alkyl group as defined above covalently bound via an O linkage containing 1 to 6 carbon atoms, such as methoxy, ethoxy, propoxy, isoproxy, butoxy, tert-butoxy and pentoxy.
  • C 1 . 4 alkoxy and “C ⁇ alkoxy” including methoxy, ethoxy, propoxy and butoxy are preferred with methoxy being particularly preferred.
  • Ci_ 6 alkylhalo refers to a Ci_ 6 alkyl which is substituted with one or more halogens. d. 3 alkylhalo groups are preferred, such as for example, -CHF 2 and -CF 3 .
  • Ci_ 6 alkoxylhalo refers to a C ⁇ alkoxyl which is substituted with one or more halogens. d- 3 alkoxylhalo groups are preferred, such as for example, -OCHF 2 and - OCF 3 .
  • carboxylate or “carboxyl” refers to the group -COO " or -COOH.
  • esters refers to a carboxyl group having the hydrogen replaced with, for example a C ⁇ alkyl group (“carboxylCi. 6 alkyl” or “alkylester”), an aryl or aralkyl group (“arylester” or “aralkylester”) and so on.
  • C0 2 Ci_ 3 alkyl groups are preferred, such as for example, methylester (CO 2 Me), ethylester (C0 2 Et) and propylester (C0 2 Pr) and includes reverse esters thereof (e.g. -OCOMe, -OCOEt and -OCOPr).
  • cyano refers to the group -CN.
  • nitro refers to the group -N0 2 .
  • amino refers to the group -NH 2 .
  • substituted amino or “secondary amino” refers to an amino group having a hydrogen replaced with, for example a C ⁇ alkyl group (“C ⁇ alkylamino”), an aryl or aralkyl group (“arylamino", “aralkylamino”) and so on.
  • C ⁇ alkylamino groups are preferred, such as for example, methylamino (NHMe), ethylamino (NHEt) and propylam ino (NHPr).
  • disubstituted amino or "tertiary amino” refers to an amino group having the two hydrogens replaced with, for example a C ⁇ alkyl group, which may be the same or different (“dialkylamino"), an aryl and alkyl group (“aryl(alkyl)amino”) and so on.
  • D ' ⁇ (C ⁇ .
  • 3 alkyl)amino groups are preferred, such as for example, dimethylamino (NMe 2 ), diethylamino (NEt 2 ), dipropylamino (NPr 2 ) and variations thereof (e.g. N(Me)(Et) and so on).
  • NMe 2 dimethylamino
  • NEt 2 diethylamino
  • NPr 2 dipropylamino
  • variations thereof e.g. N(Me)(Et) and so on.
  • substituted acyl or “ketone” refers to an acyl group having a hydrogen replaced with, for example a C ⁇ alkyl group ("Ci. 6 alkylacyl” or “alkylketone” or “ketoalkyl”), an aryl group (“arylketone”), an aralkyl group (“aralkylketone) and so on. Ci_ 3 alkylacyl groups are preferred.
  • aminoacyl refers to the group -NHC(0)H.
  • substituted amido or “substituted amide” refers to an amido group having a hydrogen replaced with, for example a C ⁇ alkyl group ("Ci_ 6 alkylamido” or “Ci_ 6 alkylam ide”), an aryl (“arylamido”), aralkyl group (“aralkylamido”) and so on.
  • C ⁇ alkylamide groups are preferred, such as for example, methylamide (-C(O)NHMe), ethylamide (- C(O)NHEt) and propylamide (-C(O)NHPr) and includes reverse amides thereof (e.g. - NHMeC(O)-, -NHEtC(O)- and -NHPrC(O)-).
  • disubstituted amido or “disubstituted amide” refers to an amido group having the two hydrogens replaced with, for example a Ci_ 6 alkyl group ("d C ⁇ ealky amido” or “diiC ⁇ ealkylJamide”), an aralkyl and alkyl group ("alkyl(aralkyl)am ido") and so on.
  • Di(d. 3 alkyl)amide groups are preferred, such as for example, dimethylamide (-C(0)NMe 2 ), diethylamide (-C(0)NEt 2 ) and dipropylamide ((-C(0)NPr 2 ) and variations thereof (e.g. - C(0)N(Me)Et and so on) and includes reverse amides thereof.
  • thiol refers to the group -SH.
  • Ci_ 6 alkylthio refers to a thiol group having the hydrogen replaced with a Ci- 6 alkyl group.
  • Ci_ 3 alkylthio groups are preferred, such as for example, thiolmethyl, thiolethyl and thiolpropyl.
  • substituted sulfinyl or “sulfoxide” refers to a sulfinyl group having the hydrogen replaced with, for example a C ⁇ alkyl group ("C ⁇ alkylsulfinyl” or "Ci_
  • alkylsulfoxide an aryl (“arylsulfinyl”), an aralkyl (“aralkyl sulfinyl”) and so on.
  • d_ 3 alkylsulf inyl groups are preferred, such as for example, -SOmethyl, -SOethyl and - SOpropyl.
  • sulfonyl refers to the group -S0 2 H.
  • substituted sulfonyl refers to a sulfonyl group having the hydrogen replaced with, for example a C ⁇ alkyl group (“sulfonylC ⁇ ealkyl”), an aryl (“arylsulfonyl”), an aralkyl (“aralkylsulfonyl”) and so on.
  • SulfonylC ⁇ alkyl groups are preferred, such as for example, -S0 2 Me, -S0 2 Et and -S0 2 Pr.
  • sulfonylamido or "sulfonamide” refers to the group -S0 2 NH 2 .
  • substituted sulfonamido or “substituted sulphonamide” refers to an sulfonylamido group having a hydrogen replaced with, for example a C ⁇ alkyl group
  • SulfonylamidoCi. 6 alkyl an aryl (“arylsulfonamide”), aralkyl (“aralkylsulfonamide”) and so on.
  • SulfonylamidoC ⁇ alkyl groups are preferred, such as for example, -S0 2 NHMe, - S0 2 NHEt and -S0 2 NHPr and includes reverse sulfonamides thereof (e.g. -NHS0 2 Me, - NHS0 2 Et and -NHS0 2 Pr).
  • disubstituted sufonamido or “disubstituted sulphonamide” refers to an sulfonylamido group having the two hydrogens replaced with, for example a C ⁇ alkyl group, which may be the same or different ("sulfonylamidodi(C 1 . 6 alkyl)"), an aralkyl and alkyl group ("sulfonamido(aralkyl)alkyl”) and so on.
  • Sulfonylamidod C ⁇ alkyl) groups are preferred, such as for example, -S0 2 NMe 2 , -S0 2 NEt 2 and -S0 2 NPr 2 and variations thereof (e.g. - S0 2 N(Me)Et and so on) and includes reserve sulfonamides thereof.
  • sulfate refers to the group OS(0) 2 OH and includes groups having the hydrogen replaced with, for example a C 1 . 6 alkyl group ("alkylsulfates”), an aryl (“arylsulfate”), an aralkyl (“aralkylsulfate”) and so on. ( ⁇ sulfates are preferred, such as for example, OS(0) 2 OMe, OS(0) 2 OEt and OS(0) 2 OPr.
  • sulfonate refers to the group S0 3 H and includes groups having the hydrogen replaced with, for example a C 1 . 6 alkyl group (“alkylsulfonate”), an aryl
  • arylsulfonate an aralkyl
  • aralkylsulfonate an aralkyl
  • aryl refers to a carbocyclic (non-heterocyclic) aromatic ring or mono-, bi- or tri-cyclic ring system.
  • the aromatic ring or ring system is generally composed of 6 to 10 carbon atoms.
  • aryl groups include but are not limited to phenyl, biphenyl, naphthyl and tetrahydronaphthyl. 6-membered aryls such as phenyl are preferred.
  • alkylaryl refers to C ⁇ alkylaryl such as benzyl.
  • alkoxyaryl refers to Ci_
  • 6alkyloxyaryl such as benzyloxy.
  • heterocyclyl refers to a moiety obtained by removing a hydrogen atom from a ring atom of a heterocyclic compound which moiety has from 3 to 10 ring atoms (unless otherwise specified), of which 1 , 2, 3 or 4 are ring heteroatoms each heteroatom being independently selected from O, S and N.
  • the prefixes 3-, 4-, 5-, 6-, 7-, 8-, 9- and 10- membered denote the number of ring atoms, or range of ring atoms, whether carbon atoms or heteroatoms.
  • the term "3-10 membered heterocylyl”, as used herein, pertains to a heterocyclyl group having 3, 4, 5, 6, 7, 8, 9 or 10 ring atoms.
  • heterocylyl groups include 5- 6-membered monocyclic heterocyclyls and 9-10 membered fused bicyclic heterocyclyls.
  • Examples of monocyclic heterocyclyl groups include, but are not limited to, those containing one nitrogen atom such as aziridine (3-membered ring), azetidine (4-membered ring), pyrrolidine (tetrahydropyrrole), pyrroline (e.g., 3-pyrroline, 2,5-dihydropyrrole), 2H- pyrrole or 3H-pyrrole (isopyrrole, isoazole) or pyrrolidinone (5-membered rings) , piperidine, dihydropyridine, tetrahydropyridine (6-membered rings), and azepine (7-membered ring); those containing two nitrogen atoms such as imidazoline, pyrazolidine (diazolidine), imidazoline, pyrazoline (dihydropyrazole) (5-membered rings), piperazine (6-membered ring); those containing one oxygen atom such as oxirane (3-membered
  • Heterocyclyls also encompass aromatic heterocyclyls and non-aromatic heterocyclyls. Such groups may be substituted or unsubstituted.
  • aromatic heterocyclyl may be used interchangeably with the term “heteroaromatic” or the term “heteroaryl” or “hetaryl”.
  • the heteroatoms in the aromatic heterocyclyl group may be independently selected from N, S and O.
  • Heteroaryl is used herein to denote a heterocyclic group having aromatic character and embraces aromatic monocyclic ring systems and polycyclic (e.g. bicyclic) ring systems containing one or more aromatic rings.
  • aromatic heterocyclyl also encompasses pseudoaromatic heterocyclyls.
  • aromatic heterocyclyl refers to a ring system which is not strictly aromatic, but which is stabilized by means of delocalization of electrons and behaves in a similar manner to aromatic rings.
  • aromatic heterocyclyl therefore covers polycyclic ring systems in which all of the fused rings are aromatic as well as ring systems where one or more rings are non-aromatic, provided that at least one ring is aromatic. In polycyclic systems containing both aromatic and non-aromatic rings fused together, the group may be attached to another moiety by the aromatic ring or by a non- aromatic ring.
  • heteroaryl groups are monocyclic and bicyclic groups containing from five to ten ring members.
  • the heteroaryl group can be, for example, a five membered or six membered monocyclic ring or a bicyclic structure formed from fused five and six membered rings or two fused six membered rings or two fused five membered rings.
  • Each ring may contain up to about four heteroatoms typically selected from nitrogen, sulphur and oxygen.
  • the heteroaryl ring will contain up to 4 heteroatoms, more typically up to 3 heteroatoms, more usually up to 2, for example a single heteroatom.
  • the heteroaryl ring contains at least one ring nitrogen atom.
  • the nitrogen atoms in the heteroaryl rings can be basic, as in the case of an imidazole or pyridine, or essentially non-basic as in the case of an indole or pyrrole nitrogen.
  • the number of basic nitrogen atoms present in the heteroaryl group, including any amino group substituents of the ring, will be less than five.
  • Aromatic heterocyclyl groups may be 5-membered or 6-membered mono-cyclic aromatic ring systems.
  • 5-membered monocyclic heteroaryl groups include but are not limited to furanyl, thienyl, pyrrolyl, oxazolyl, oxadiazolyl (including 1 ,2,3 and 1 ,2,4 oxadiazolyls and furazanyl i.e.
  • thiazolyl isoxazolyl, isothiazolyl, pyrazolyl, imidazolyl, triazolyl (including 1 ,2,3, 1 ,2,4 and 1 ,3,4 triazolyls), oxatriazolyl, tetrazolyl, thiadiazolyl (including 1 ,2,3 and 1 ,3,4 thiadiazolyls) and the like.
  • 6-membered monocyclic heteroaryl groups include but are not limited to pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, pyranyl, oxazinyl, dioxinyl, thiazinyl, thiadiazinyl and the like.
  • 6-membered aromatic heterocyclyls containing nitrogen include pyridyl (1 nitrogen), pyrazinyl, pyrimidinyl and pyridazinyl (2 nitrogens).
  • Aromatic heterocyclyl groups may also be bicyclic or polycyclic heteroaromatic ring systems such as fused ring systems (including purine, pteridinyl, napthyridinyl, 1 H thieno[2,3-c]pyrazolyl, thieno[2,3-b]furyl and the like) or linked ring systems (such as oligothiophene, polypyrrole and the like).
  • fused ring systems including purine, pteridinyl, napthyridinyl, 1 H thieno[2,3-c]pyrazolyl, thieno[2,3-b]furyl and the like
  • linked ring systems such as oligothiophene, polypyrrole and the like.
  • Fused ring systems may also include aromatic 5- membered or 6-membered heterocyclyls fused to carbocyclic aromatic rings such as phenyl, napthyl, indenyl, azulenyl, fluorenyl, anthracenyl and the like, such as 5-membered aromatic heterocyclyls containing nitrogen fused to phenyl rings, 5-membered aromatic heterocyclyls containing 1 or 2 nitrogens fused to phenyl ring.
  • aromatic 5- membered or 6-membered heterocyclyls fused to carbocyclic aromatic rings such as phenyl, napthyl, indenyl, azulenyl, fluorenyl, anthracenyl and the like, such as 5-membered aromatic heterocyclyls containing nitrogen fused to phenyl rings, 5-membered aromatic heterocyclyls containing 1 or 2 nitrogens fused to phenyl ring.
  • a bicyclic heteroaryl group may be, for example, a group selected from: a) a benzene ring fused to a 5- or 6-membered ring containing 1 , 2 or 3 ring heteroatoms; b) a pyridine ring fused to a 5- or 6-membered ring containing 1 , 2 or 3 ring heteroatoms; c) a pyrimidine ring fused to a 5- or 6-membered ring containing 1 or 2 ring heteroatoms; d) a pyrrole ring fused to a 5- or 6-membered ring containing 1 , 2 or 3 ring heteroatoms; e) a pyrazole ring fused to a 5- or 6-membered ring containing 1 or 2 ring heteroatoms; f) an imidazole ring fused to a 5- or 6-membered ring containing 1 or 2 ring heteroatoms; g) an oxazole ring fused to a 5-
  • bicyclic heteroaryl groups containing a five membered ring fused to another five membered ring include but are not limited to imidazothiazole (e.g. imidazo[2, 1-b]thiazole) and imidazoimidazole (e.g. imidazo[1 ,2-a]imidazole).
  • imidazothiazole e.g. imidazo[2, 1-b]thiazole
  • imidazoimidazole e.g. imidazo[1 ,2-a]imidazole
  • bicyclic heteroaryl groups containing a six membered ring fused to a five membered ring include but are not limited to benzofuran, benzothiophene, benzimidazole, benzoxazole, isobenzoxazole, benzisoxazole, benzothiazole,
  • benzisothiazole isobenzofuran, indole, isoindole, indolizine, indoline, isoindoline, purine (e.g., adenine, guanine), indazole, pyrazolopyrimidine (e.g. pyrazolo[1 ,5-a]pyrimidine), benzodioxole and pyrazolopyridine (e.g. pyrazolo[1 ,5-a]pyridine) groups.
  • a further example of a six membered ring fused to a five membered ring is a pyrrolopyridine group such as a pyrrolo[2,3-b]pyridine group.
  • bicyclic heteroaryl groups containing two fused six membered rings include but are not limited to quinoline, isoquinoline, chroman, thiochroman, chromene, isochromene, isochroman, benzodioxan, quinolizine, benzoxazine, benzodiazine, pyridopyridine, quinoxaline, quinazoline, cinnoline, phthalazine, naphthyridine and pteridine groups.
  • heteroaryl groups containing an aromatic ring and a non-aromatic ring include tetrahydronaphthalene, tetrahydroisoquinoline, tetrahydroquinoline,
  • aromatic heterocyclyls fused to carbocyclic aromatic rings may therefore include but are not limited to benzothiophenyl, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzimidazolyl, indazolyl, benzoxazolyl, benzisoxazolyl, isobenzoxazoyi, benzothiazolyl, benzisothiazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, quinazolinyl, cinnolinyl, benzotriazinyl, phthalazinyl, carbolinyl and the like.
  • non-aromatic heterocyclyl encompasses optionally substituted saturated and unsaturated rings which contain at least one heteroatom selected from the group consisting of N, S and O.
  • Non-aromatic heterocyclyls may be 3-7 membered mono-cyclic rings.
  • Examples of 5-membered non-aromatic heterocyclyl rings include 2H-pyrrolyl, 1 - pyrrolinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrrolidinyl, 1 -pyrrolidinyl, 2-pyrrolidinyl, 3-pyrrolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, pyrazolinyl, 2-pyrazolinyl, 3-pyrazolinyl, pyrazolidinyl, 2-pyrazolidinyl, 3-pyrazolidinyl, imidazolidinyl, 3-dioxalanyl, thiazolidinyl, isoxazolidinyl, 2-imidazolinyl and the like.
  • 6-membered non-aromatic heterocyclyls include piperidinyl, piperidinonyl, pyranyl, dihyrdopyranyl, tetrahydropyranyl, 2H pyranyl, 4H pyranyl, thianyl, thianyl oxide, thianyl dioxide, piperazinyl, diozanyl, 1 ,4-dioxinyl, 1 ,4-dithianyl, 1 ,3,5- triozalanyl, 1 ,3,5-trithianyl, 1 ,4-morpholinyl, thiomorpholinyl, 1 ,4-oxathianyl, triazinyl, 1 ,4- thiazinyl and the like.
  • Examples of 7-membered non-aromatic heterocyclyls include azepanyl, oxepanyl, thiepanyl and the like.
  • Non-aromatic heterocyclyl rings may also be bicyclic heterocyclyl rings such as linked ring systems (for example uridinyl and the like) or fused ring systems.
  • Fused ring systems include non-aromatic 5-membered, 6-membered or 7-membered heterocyclyls fused to carbocyclic aromatic rings such as phenyl, napthyl, indenyl, azulenyl, fluorenyl, anthracenyl and the like.
  • Examples of non-aromatic 5-membered, 6-membered or 7- membered heterocyclyls fused to carbocyclic aromatic rings include indolinyl,
  • halo refers to fluoro, chloro, bromo or iodo.
  • the term "optionally substituted” or “optional substituent” as used herein refers to a group which may or may not be further substituted with 1 , 2, 3, 4 or more groups, preferably 1 , 2 or 3, more preferably 1 or 2 groups selected from the group consisting of Chalky!, C 2 - 6 alkenyl, C 2 - 6 alkynyl, C 3 .
  • suitable derivatives of aromatic heterocyclyls containing nitrogen include W-oxides thereof.
  • the compounds of the invention may also be prepared as salts which are pharmaceutically acceptable, but it will be appreciated that non-pharmaceutically acceptable salts also fall within the scope of the present invention, since these are useful as
  • pharmaceutically acceptable salts include salts of pharmaceutically acceptable cations such as sodium , potassium, lithium, calcium, magnesium, ammonium and alkylamimonium; acid addition salts of pharmaceutically acceptable inorganic acids such as hydrochloric, orthophosphoric, sulfuric, phosphoric, nitric, carbonic, boric, sulfamic and hydrobromic acids; or salts of pharmaceutically acceptable organic acids such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumaric, citric, lactic, mucic, gluconic, benzoic, succinic, oxalic, phenylacetic, methanesulfonic, trihalomethanesulfonic, toluenesulfonic,
  • Salts of amine groups may also comprise quaternary ammonium salts in which the amino nitrogen atom carries a suitable organic group such as an alkyl, alkenyl, alkynyl or aralkyl moiety.
  • the salts may be formed by conventional means, such as by reacting the free base form of the compound with one or more equivalents of the appropriate acid.
  • a reference to a pharmaceutically acceptable salt includes the solvent addition forms or crystal forms thereof, particularly solvates or polymorphs.
  • Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and may be formed during the process of crystallization with pharmaceutically acceptable solvents such as water, alcohols such as methanol, ethanol or isopropyl alcohol, DMSO, acetonitrile, dimethyl formamide (DMF) and the like with the solvate forming part of the crystal lattice by either non-covalent binding or by occupying a hole in the crystal lattice. Hydrates are formed when the solvent is water, alcoholates are formed when the solvent is alcohol.
  • Solvates of the compounds of the present invention can be conveniently prepared or formed during the processes described herein.
  • the compounds of the present invention can exist in unsolvated as well as solvated forms.
  • the solvated forms are considered equivalent to the unsolvated forms for the purposes of the compounds and methods provided herein.
  • the compounds of the present invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • the solvated forms of the compounds of the present invention are also considered to be disclosed herein.
  • compounds of formula (I) may possess a chiral centre and may therefore exist as an isomer such as a racemate or an R- or S- enantiomer.
  • the compounds may therefore be used as a purified enantiomer or diastereomer, or as a mixture of any ratio thereof.
  • the isomers may be separated conventionally by chromatographic methods or using a resolving agent. Alternatively the individual isomers may be prepared by asymmetric synthesis using chiral intermediates. Where the compound has a carbon- carbon double bond, it may occur in Z- or E- form and all isomeric forms of the compounds being included in the present invention.
  • This invention also encompasses prodrugs of the compounds of formula (I).
  • pro-drug is used herein in its broadest sense to include those compounds which are converted in vivo to the compound of formula (I).
  • Use of the prodrug strategy optimises the delivery of the drug to its site of action.
  • the compounds of formula (I) having carboxylic esters may act as prodrug moieties with respect to their corresponding carboxylic acid analogues.
  • RNA dependent RNA polymerase of HCV RNA dependent RNA polymerase of HCV
  • HCV NS5B polymerase which is the viral RNA-dependent RNA polymerase (RdRp) that is responsible for viral replications.
  • RdRp viral RNA-dependent RNA polymerase
  • HCV NS5B protein is released from a polyprotein and is involved in the synthesis of double-stranded RNA from a single-stranded viral RNA genome. It is believed that the replication and/or reproduction of HCV virus may be inhibited or prevented through the inhibition of NS5B polymerase and suppress or prevent the formation of the double- stranded HCV RNA.
  • the compounds of formula (I) act by specific inhibition of NS5B polymerase, the compounds may be tested for the lack of inhibitory activity in an assay measuring the activity of an RNA-dependent RNA polymerase other than HCV polymerase or in a DNA dependent RNA polymerase assay.
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition may further comprise or be administered in combination with one or more other antiviral agents such as Ribavirin (Copegus® or Rebetol®), an antiviral nucleoside inhibitor of NS5B polymerase (such as 4-amino-7-(2-C-methyl- -D-ribofuranosyl)-pyrrolo[2, 1-f][1 ,2,4]triazine; PSI-7977; PSI-938; RG7128 or mericitabine; IDX-184; INX-189 and other such agents that may be developed) a non-nucleoside inhibitor of NS5B polymerase (such as GS-9190 or tegobuvir; PF-868554 or filibuvir; VX-222; IDX-375; ABT-072; ABT-333; ANA-598 or setrobuvir;
  • the composition may also additionally comprise at least one immunomodulatory agent for example an interferon or interferon derivative such as interferon alpha 2B (such as Intron® A interferon available from Schering Corp., Kenilworth, N.J.), pegylated interferon alpha 2A (such as Pegasys® available from Hoffmann-LaRoche, Nutley, N.J.), pegylated interferon alpha 2B (such as Peg-lntron® available from Schering Corp.
  • interferon alpha 2B such as Intron® A interferon available from Schering Corp., Kenilworth, N.J.
  • pegylated interferon alpha 2A such as Pegasys® available from Hoffmann-LaRoche, Nutley, N.J.
  • pegylated interferon alpha 2B such as Peg-lntron® available from Schering Corp.
  • the other antiviral agent is Ribavarin optionally in combination with peg/I FN.
  • the other antiviral agent is an NS5B inhibitor, more particularly a nucleoside inhibitor such as the bicyclic nucleosides and nucleotides of the general formula described in WO2010/002877, for example, 4-amino-7-(2-C-methyl-3-D- ribofuranosyl)-pyrrolo[2, 1-f][1 ,2,4]triazine.
  • the other antiviral agent is an NS3/4A protease inhibitor such as telaprevir (VX-950) or Incivek®; boceprevir or Victrelis®; BI-201335; TMC-435; RG- 7227 or danoprevir; MK-7009 or vaniprevir; GS-9451 ; GS-9256; BMS-650032; ACH-1625; ACH-2684; MK-5172; ABT-450; IDX-320; SCH-900518, particularly telaprevir (VX-950).
  • VX-950 NS3/4A protease inhibitor
  • the other antiviral agent is an NS5A inhibitor such as BMS-790052 (daclatasvir); GS-5885; ABT-267; PPI-461 ; ACH-2928; GSK2336805, particularly BMS-790052 (daclatasvir).
  • composition is intended to include the formulation of an active ingredient with conventional carriers and excipients, and also with encapsulating materials as the carrier, to give a capsule in which the active ingredient (with or without other carriers) is surrounded by the encapsulation carrier.
  • Any carrier must be “pharmaceutically acceptable” meaning that it is compatible with the other ingredients of the composition and is not deleterious to a subject.
  • compositions of the present invention may contain other therapeutic agents as described above, and may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavours, etc.) according to techniques such as those well known in the art of pharmaceutical formulation (See, for example, Remington: The Science and Practice of Pharmacy, 21st Ed. , 2005, Lippincott Williams & Wilkins).
  • the pharmaceutical composition includes those suitable for oral, rectal, nasal, topical (including buccal and sub-lingual), vaginal or parenteral (including intramuscular, sub-cutaneous and intravenous) administration or in a form suitable for administration by inhalation or insufflation.
  • compositions and unit dosages thereof may thus be placed into the form of pharmaceutical compositions and unit dosages thereof, and in such form may be employed as solids, such as tablets or filled capsules, or liquids such as solutions, suspensions, emulsions, elixirs, or capsules filled with the same, all for oral use, in the form of suppositories for rectal administration ; or in the form of sterile injectable solutions for parenteral (including subcutaneous) use.
  • Such pharmaceutical compositions and unit dosage forms thereof may comprise conventional ingredients in conventional proportions, with or without additional active compounds or principles, and such unit dosage forms may contain any suitable effective amount of the active ingredient commensurate with the intended daily dosage range to be employed.
  • pharmaceutically acceptable carriers can be either solid or liquid.
  • Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispensable granules.
  • a solid carrier can be one or more substances which may also act as diluents, flavouring agents, solubilisers, lubricants, suspending agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material.
  • Suitable carriers are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium
  • carboxymethylcellulose a low melting wax, cocoa butter, and the like.
  • preparation is intended to include the formulation of the active compound with
  • encapsulating material as carrier providing a capsule in which the active component, with or without carriers, is surrounded by a carrier, which is thus in association with it.
  • a carrier which is thus in association with it.
  • cachets and lozenges are included. Tablets, powders, capsules, pills, cachets, and lozenges can be used as solid forms suitable for oral administration.
  • Liquid form preparations include solutions, suspensions, and emulsions, for example, water or water-propylene glycol solutions.
  • parenteral injection liquid preparations can be formulated as solutions in aqueous polyethylene glycol solution.
  • Sterile liquid form compositions include sterile solutions, suspensions, emulsions, syrups and elixirs.
  • the active ingredient can be dissolved or suspended in a
  • pharmaceutically acceptable carrier such as sterile water, sterile organic solvent or a mixture of both.
  • compositions according to the present invention may thus be formulated for parenteral administration (e. g. by injection, for example bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi-dose containers with an added preservative.
  • the compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulation agents such as suspending, stabilising and/or dispersing agents.
  • the active ingredient may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilisation from solution, for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use.
  • compositions suitable for injectable use include sterile injectable solutions or dispersions, and sterile powders for the extemporaneous preparation of sterile injectable solutions. They should be stable under the conditions of manufacture and storage and may be preserved against oxidation and the contaminating action of microorganisms such as bacteria or fungi.
  • the solvent or dispersion medium for the injectable solution or dispersion may contain any of the conventional solvent or carrier systems for the compounds, and may contain, for example, water, ethanol, polyol (for example, glycerol, propylene glycol and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • polyol for example, glycerol, propylene glycol and liquid polyethylene glycol, and the like
  • compositions suitable for injectable use may be delivered by any appropriate route including intravenous, intramuscular, intracerebral, intrathecal, epidural injection or infusion.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various other ingredients such as these enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilised active ingredient into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • preferred methods of preparation are vacuum drying or freeze-drying of a previously sterile-filtered solution of the active ingredient plus any additional desired ingredients.
  • the active ingredients When the active ingredients are suitably protected they may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsule, or it may be compressed into tablets, or it may be incorporated directly with the food of the diet.
  • the active compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • the amount of active compound in therapeutically useful compositions should be sufficient that a suitable dosage will be obtained.
  • the tablets, troches, pills, capsules and the like may also contain the components as listed hereafter: a binder such as gum, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such a sucrose, lactose or saccharin may be added or a flavouring agent such as peppermint, oil of wintergreen, or cherry flavouring.
  • a binder such as gum, acacia, corn starch or gelatin
  • excipients such as dicalcium phosphate
  • a disintegrating agent such as corn starch, potato starch, alginic acid and the like
  • a lubricant such as magnesium stearate
  • a sweetening agent such as sucrose, lactose or saccharin may be added or a flavouring agent such as peppermint, oil of winter
  • tablets, pills, or capsules may be coated with shellac, sugar or both.
  • a syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavouring such as cherry or orange flavour.
  • any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed.
  • the active compound (s) may be incorporated into sustained-release preparations and formulations, including those that allow specific delivery of the active peptide to specific regions of the gut.
  • Aqueous solutions suitable for oral use can be prepared by dissolving the active component in water and adding suitable colorants, flavours, stabilising and thickening agents, as desired.
  • Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, or other well known suspending agents.
  • Pharmaceutically acceptable carriers and/or diluents include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like.
  • solid form preparations that are intended to be converted, shortly before use, to liquid form preparations for oral administration.
  • liquid forms include solutions, suspensions, and emulsions.
  • These preparations may contain, in addition to the active component, colorants, flavours, stabilisers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilising agents, and the like.
  • the compounds according to the invention may be formulated as ointments, creams or lotions, or as a transdermal patch.
  • Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilising agents, dispersing agents, suspending agents, thickening agents, or colouring agents.
  • Formulations suitable for topical administration in the mouth include lozenges comprising active agent in a flavoured base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Solutions or suspensions are applied directly to the nasal cavity by conventional means, for example with a dropper, pipette or spray.
  • the formulations may be provided in single or multidose form. In the latter case of a dropper or pipette, this may be achieved by the patient administering an appropriate, predetermined volume of the solution or suspension.
  • the compounds according to the invention may be encapsulated with cyclodextrins, or formulated with other agents expected to enhance delivery and retention in the nasal mucosa.
  • Administration to the respiratory tract may also be achieved by means of an aerosol formulation in which the active ingredient is provided in a pressurised pack with a suitable propellant such as a chlorofluorocarbon (CFC) for example dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane, carbon dioxide, or other suitable gas.
  • a suitable propellant such as a chlorofluorocarbon (CFC) for example dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane, carbon dioxide, or other suitable gas.
  • CFC chlorofluorocarbon
  • the aerosol may conveniently also contain a surfactant such as lecithin.
  • a surfactant such as lecithin.
  • the dose of drug may be controlled by provision of a metered valve.
  • the active ingredients may be provided in the form of a dry powder, for example a powder mix of the compound in a suitable powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP).
  • a powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP).
  • PVP polyvinylpyrrolidone
  • the powder carrier will form a gel in the nasal cavity.
  • the powder composition may be presented in unit dose form for example in capsules or cartridges of, e. g. gelatin, or blister packs from which the powder may be administered by means of an inhaler.
  • the compound In formulations intended for administration to the respiratory tract, including intranasal formulations, the compound will generally have a small particle size for example of the order of 5 to 10 microns or less. Such a particle size may be obtained by means known in the art, for example by micronisation.
  • formulations adapted to give sustained release of the active ingredient may be employed.
  • the pharmaceutical preparations are preferably in unit dosage forms.
  • the preparation is subdivided into unit doses containing appropriate quantities of the active component.
  • the unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules.
  • the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms are dictated by and directly dependent on (a) the unique characteristics of the active material and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active material for the treatment of a HCV viral infection in living subjects having a diseased condition in which bodily health is impaired.
  • the invention also includes the compounds in the absence of carrier where the compounds are in unit dosage form.
  • compositions comprising compounds of the invention formulated for oral delivery either alone or in combination with another HCV antiviral agent are particularly preferred.
  • the compounds of formula (I) may be used in the treatment of a Flaviviridae viral infection such as a HCV infection.
  • the term "treatment" means affecting a subject, tissue or cell to obtain a desired pharmacological and/or physiological effect and includes: (a) inhibiting the viral infection, i.e. arresting its development or further development; (b) relieving or ameliorating the effects of the viral infection, i.e. cause regression of the effects of the viral infection; (c) reducing the incidence or the viral infection or (d) preventing the infection from occurring in a subject, tissue or cell predisposed to the viral infection disease or at risk thereof, but has not yet been diagnosed with a protective pharmacological and/or physiological effect so that the viral infection does not develop or occur in the subject, tissue or cell.
  • the prevention of hepatitis C means, for example, administration of a
  • subject refers to any animal, in particular mammals such as humans having a disease or condition which requires treatment with the compound of formula (I).
  • administering refers to providing the compound or pharmaceutical composition of the invention to a subject suffering from or at risk of the diseases or conditions to be treated or prevented.
  • 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-PC , 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
  • Flaviviridae virus refers to a virus of the family Flaviviridae, which family includes the Hepacivirus Flavivirus and Pestivirus or hepatitis C-like virus genera.
  • a representative species of the genus of hepatitis C-like viruses is hepatitis C virus.
  • terapéuticaally effective amount refers to the amount of the compound of formula (I) that will elicit the biological or medical response of a subject, tissue or cell that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • an appropriate dosage level will generally be about 0.01 to 500 mg per kg subject body weight per day which can be administered in single or multiple doses.
  • the dosage level will be about 0.1 to about 250 mg/kg per day; more preferably about 0.5 to about 100 mg/kg per day.
  • a suitable dosage level may be about 0.01 to 250 mg/kg per day, about 0.05 to 100 mg/kg per day, or about 0.1 to 50 mg/kg per day. Within this range the dosage may be 0.05 to 0.5, 0.5 to 5 or 5 to 50 mg/kg per day.
  • the dosage may be selected, for example to any dose within any of these ranges, for therapeutic efficacy and/or symptomatic adjustment of the dosage to the subject to be treated
  • the dosage forms and levels may be formulated for either concurrent, sequential or separate administration or a combination thereof.
  • each moiety having a substitutable hydrogen such as for example, alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, cycloalkenyl, heterocyclyl, aryl and heteroaryl, in each occurrence as described in the general schemes and methods which follow may be optionally substituted.
  • substitutable hydrogen such as for example, alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, cycloalkenyl, heterocyclyl, aryl and heteroaryl, in each occurrence as described in the general schemes and methods which follow may be optionally substituted.
  • the particular examples which are described herein may undergo further functionalisation using methods known in the art, for example, compounds comprising amino groups or acid groups may undergo reduction amination or amide couplings respectively to form further examples of compounds of the invention.
  • compounds of formula (II) are prepared by heating a suitable amino- substituted heterocyclyl with a suitable alpha-halo ketone in the presence of a solvent, such as DMF, and a suitable base, such as potassium carbonate.
  • a solvent such as DMF
  • a suitable base such as potassium carbonate
  • intermediates of formula (II) containing carboxylic ester groups i.e. C0 2 C 1 . 6 alkyl (such as C0 2 CH 3 , C0 2 CH 2 CH 3 and C0 2 CH 2 CH 2 CH 3 ) may be converted to their corresponding carboxylic acids using various conditions (e.g. treatment with aq. NaOH in methanol/THF) which may then be coupled with any suitable primary or secondary amides using a variety of standard peptide coupling techniques to make the corresponding amides (e.g. the use of a peptide coupling agent such as HATU, HBTU, EDCI etc. with the appropriate solvent and base).
  • a peptide coupling agent such as HATU, HBTU, EDCI etc.
  • a process for producing a compound of formula (I) when B is -CONH-R7- comprising the step of reacting a compound of formula (II) with an amino precursor of general formula NH 2 R 7 under amide coupling conditions.
  • Suitable amide coupling conditions will be understood by those skilled in the art and have been described in many references such as Advanced Organic Chemistry - 4 th Edition, March J., John Wiley & Sons Inc, New York, 1992 (see also 5 th Edition, John Wiley & Sons, New York 2001 and 6 th Edition, John Wiley & Sons, New Jersey, 2007).
  • compounds of formula (II) have also demonstrated antiviral activity. Accordingly, compounds of formula (II) will be understood to be process intermediates and/or final compounds as the case may be.
  • Preparative HPLC was carried out using either a Gilson 322 pump with a Gilson 215 liquid handler and a HP1 100 PDA detector or an Agilent 1200 Series mass detected preparative LCMS using a Varian XRs C-18 100 x 21 .2 mm column. Unless otherwise specified, the HPLC systems employed Phenomenex C8(2) columns using either acetonitrile or acetonitrile containing 0.06% TFA in water or water containing 0.1 % TFA.
  • BSA Bovine serum albumin
  • DIPEA N,N-Diisopropylethylamine
  • DMEM Dulbecco's modified eagle medium
  • GTP Guanosine triphosphate
  • HATU 2-(7-Aza-1 H-benzotriazole-1 -yl)-1 , 1 ,3,3-tetramethyluronium hexafluorophosphate
  • HBTU 0-(Benzotriazol-1 -yl)-/V,/V,/V',A/-tetramethyluronium hexafluorophosphate
  • LHMDS Lithium hexamethyldisilazide
  • Step a To a solution of 1-(4-fluorophenyl)ethanone (5.00 g; 36.20 mmol) in THF (80 mL) at - 78°C was added a 1 M THF solution of LHMDS in THF (40 mmol). After 30 minutes the cyclohexanone (3.90 g; 40 mmol) in THF (80mL) was added and the reaction mixture was stirred at that temperature for 30 minutes and then quenched upon addition of an aqueous solution of sodium bircarbonate. Ethyl acetate (200 mL) was added and the organic layer was washed with water (2 x 100 mL) then brine (100 mL).
  • Step b To a solution of 1-(4-fluorophenyl)-2-(1 -hydroxycyclohexyl)ethanone (350 mg; 1 .48 mmol) in DCM (4 mL) was added triethylsilane (450 mg; 3.90 mmol) followed by TFA (2 mL) and the reaction mixture was stirred for 16 hours at ambient temperature. The reaction mixture was partitioned between EtOAc (75 mL) and water (100 mL). The aqueous layer was extracted with EtOAc (3 x 60 mL) and the combined organics were washed with brine (75 mL), dried (MgS0 4 ) and concentrated to leave a brown oil (4100 mg).
  • the crude reaction material was purified by automated flash column chromatography (Biotage-SP4) on silica (80g) eluting with EtOAc and hexane to leave 2-cyclohexyl-1 -(4-fluorophenyl)ethanone (2.20 g) as a colourless oil.
  • Step c To a solution of 2-cyclohexyl-1 -(4-fluorophenyl)ethanone (2.50 g; 1 1 .35 mmol) in dioxane/diethyl ether (1 : 1 ; 100 mL) was added bromine (0.64 mL; 12 mmol) and the reaction mixture was stirred at ambient temperature. After 2 hours the reaction mixture was partitioned between EtOAc (125ml) and water (125 ml) and the aqueous layer was separated and extracted with EtOAc (3 x 30 ml). The combined organics were washed with brine (35 mL). dried (MgS0 4 ) and concentrated to leave 2-bromo-2-cyclohexyl-1 -(4- fluorophenyl)ethanone (2.79 g) as a colourless oil.
  • Step d To a solution of methyl 5-amino-1 H-pyrazole-3-carboxylate (75 mg; 0.53 mmol) in DMF (2 mL) was added the 2-bromo-2-cyclohexyl-1-(4-fluorophenyl)ethanone ( 170 mg; 0.58 mmol) followed by potassium carbonate (263 mg; 2.66 mmol) and the reaction mixture was stirred at 75°C. After 2 hours water (25 mL) was added and the aqueous layer was extracted with EtOAc (3 x 30 mL). The combined organics were washed with brine (25 mL), dried (MgS0 4 ) and concentrated to leave a brown oil (89 mg).
  • Step e To a solution of methyl 3-cyclohexyl-2-(4-fluorophenyl)-1 H-imidazo[1 ,2-b]pyrazole-6- carboxylate (i) (3 mg) in MeOH/THF (1 : 1 ; 1 mL) was added a 1 M aqueous solution of lithium hydroxide (0.09 mmol) and the reaction was heated at 60°C for 3 hrs. A 1 M aqueous solution of citric acid (25 ml) was added and the aqueous layer was extracted with EtOAc (3 x 30 mL). The combined organics were washed with brine (25 mL), dried (MgS0 4 ) and concentrated to leave a brown oil (4 mg). Purification by reverse phase LCMS:
  • Step a To a solution of methyl 3-cyclohexyl-2-(4-fluorophenyl)-1 H-imidazo[1 ,2-b]pyrazole-6- carboxylate (i) (10 mg; 0.03 mmol) in DMF (1 mL) was added iodomethane (0.06 mmol) followed by potassium carbonate (29 mg; 0.29 mmol). After 16 hours H 2 0 (25 ml) was added and the aqueous layer was extracted with EtOAc (3 x 30 mL). The combined organics were washed with brine (25 mL), dried (MgS0 4 ) and concentrated to leave a brown oil (12 mg).
  • Step b To a solution of methyl 3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2- b]pyrazole-6-carboxylate (iii) (5 mg; 0.01 mmol) in MeOH/THF (1 : 1 ; 2 mL) was added a 1 M aqueous solution of lithium hydroxide (0.1 mmol). The reaction was heated at 60°C for 2 hrs.
  • Step a To a solution of ethyl 5-amino-1 H-pyrazole-4-carboxylate (30 mg; 0.19 mmol) in DMF (2 ml_) was added 2-bromo-2-cyclohexyl-1-(4-fluorophenyl)ethanone (64 mg; 0.21 mmol) followed by potassium carbonate (96 mg; 0.97 mmol) and the reaction was stirred at ambient temperature for 4 hours. H 2 0 (25 ml) was added and the aqueous layer was extracted with EtOAc (3 x 30 ml_) and the combined organics were washed with brine (25 ml_), dried (MgS0 4 ) and concentrated to leave a brown oil (85 mg).
  • Step b To a solution of ethyl 3-cyclohexyl-2-(4-fluorophenyl)-1 H-imidazo[1 ,2-b]pyrazole-7- carboxylate (v) (75 mg; 0.21 mmol) in DMF (5 mL) was added iodomethane (60 mg; 0.42 mmol) followed by potassium carbonate (105 mg; 1 .06 mmol). After 16 hours H 2 0 (25 ml) was added and the aqueous layer was extracted with EtOAc (3 x 30 mL). The combined organics were washed with brine (25 mL), dried (MgS0 4 ) and concentrated to leave a brown oil (65 mg).
  • Step c To a solution of ethyl 3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2- b]pyrazole-7-carboxylate (vi) (20 mg; 0.05 mmol) in MeOH/THF (1 :1 ; 2 mL) was added a 1 M aqueous solution of lithium hydroxide (1 .08 mmol). The reaction was heated at 60 °C for 6 hrs. 1 M HCI was added until ⁇ pH 3 and the aqueous layer was extracted with EtOAc (3 x 30 mL). The combined organics were washed with brine (25 mL), dried (MgS0 4 ) and concentrated to leave a brown oil (15 mg). Purification by reverse phase LCMS:
  • Step a A 1 M solution of zinc chloride in diethyl ether (39.5 mL) was added to 140 mL anhydrous ether under an atmosphere of argon at 0°C. To this was added
  • Step b To a solution of 2-cyclohexyl-1-phenylethanone (3470 mg) in dioxane/diethyl ether (1 : 1 , 20 mL) was added at rt bromine (930 ⁇ _) in portions over 40 min (progress monitored by 1 H NMR), and the brown mixture was stirred at ambient temperature for another 30 min. Work-up: the reaction mixture was partitioned between EtOAc (100 ml) and NaHC0 3 (100 ml). The aqueous layer was separated and extracted with EtOAc (100 ml).
  • Step c To a solution of ethyl 5-amino-1 /-/-pyrazole-4-carboxylate (120 mg) in anhydrous DMF (5 mL) was added under an atmosphere of argon 2-bromo-2-cyclohexyl-1 - phenylethanone (185 mg) followed by potassium carbonate (320 mg), and the reaction mixture was stirred at 100 °C for 2 hrs and at rt for 17 hrs. After this period iodomethane (50 pL) was added and the reaction mixture was stirred at rt for 20 min.
  • Step d A solution of ethyl 3-cyclohexyl-1 -methyl-2-phenyl-1 H-imidazo[1 ,2-b]pyrazole-7- carboxylate (ix) (60 mg) in 1 M aquous KOH/1 ,4-dioxane (6.4 mL, 1 : 1 ) was heated at reflux for 2 hrs. The reaction mixture was cooled to 0°C, and adjusted to pH 7 with 1 M HCI and satd NaHC0 3 solution, extracted with EtOAc (2x 20 mL), the combined organic layers were washed with brine (10 mL), dried (MgS0 4 ), filtered and evaporated to drynes to afford an oil that solidified on standing.
  • Step a To a mixture of 63 mL water, 7 mL 7 N NaOH and 60 mL 1 ,2-dichloroethane was added 1 -(2-hydroxyphenyl)ethanone, 1 100 mg tetrabutylammonium bromide and 1 -bromo-2- chloroethane (4.5 mL), and the reaction mixture was stirred vigorously at rt for 96 hrs, then heated at reflux for 16 hrs.
  • Step b To a solution of 1-[2-(2-chloroethoxy)phenyl]ethanone (4200 mg) in THF (90 mL) at - 78 C was added under an atmosphere of argon a 1 M THF solution of LiHMDS (25 mL). After 30 minutes cyclohexanone (2520 ⁇ ) in THF (10 mL) was added and the reaction mixture was stirred at -78 °C for 1 .5 hrs.
  • Step c To a solution of 1-[2-(2-chloroethoxy)phenyl]-2-(1 -hydroxycyclohexyl)ethanone (4910 mg) in anhydrous dichloromethane (100 mL, argon) was added under an atmosphere of argon triethylsilane (27 mL) followed by trifluoroacetic acid (1.5 mL) and the reaction mixture was stirred at ambient temperature for 2.5 days. Progress of the reaction was monitored by TLC. After ca 90% conversion to the product, the reaction mixture was quenched with satd. NaHC0 3 solution (100 mL), followed by extraction with DCM (2x50 mL). The combined organic layers were dried (MgS0 4 ), filtered and evaporated to dryness.
  • Step d To a solution of 1-[2-(2-chloroethoxy)phenyl]-2-cyclohexylethanone (1000 mg) in dioxane/diethyl ether (1 : 1 , 60 mL) was added bromine (180 pL), and the brown mixture was stirred at ambient temperature for 30 min. The progress of the reaction was followed by LCMS. After 30 min reaction time, more bromine (9 pL) was added and stirring was continued for 30 min. The reaction mixture was partitioned between EtOAc (150 ml) and NaHC0 3 (50 ml). The aqueous layer was separated and extracted with EtOAc (50 ml).
  • Step e To a solution of methyl 5-amino-1 H-pyrazole-3-carboxylate hydrochloride (270 mg) in anhydrous DMF (18 mL) under an atmosphere of argon was added sodium hydride (60% in mineral oil, 130 mg) at 0 °C. The reaction mixture was stirred at this temperature for 45 min followed by addition of a solution of 2-bromo-1 -[2-(2-chloroethoxy)phenyl]-2- cyclohexylethanone (500 mg) in anhydrous DMF (18 mL). The reaction mixture was stirred at rt for 21 hrs, followed by heating at 100 °C for 1 hr. The progress of the reaction was monitored by LCMS.
  • Reaction mixture was acidified with glacial acetic acid, concentrated on the rotavap (60 °C), the residue was partitioned between water and EtOAc (200 mL, 1 : 1 ), the aqueous layer was extracted with EtOAc (100 mL), the combined organic layers were dried (MgS0 4 ), filtered and evaporated to dryness to leave a brown oil.
  • Step f A solution of methyl 13-cyclohexyl-6,7-dihydropyrazolo[5',1':2,3]imidazo[1,5- c][1,4]benzoxazepine-10-carboxylate (xi) (40 mg) in 1M aquous KOH/1 ,4-dioxane (6 mL, 1:1) was heated at reflux for 15 min. Reaction mixture was cooled to 0°C, and adjusted to pH 5 with 1 M HCI, then extracted with EtOAc (2x 20 mL), the combined organic layers were washed with brine (10 mL), dried (MgS0 4 ), filtered and evaporated to dryness: 31.3 mg (81 % yield) oil.
  • Step a Resorcinol (2370 mg) and boron trifluoride etherate (4.50 mL) were combined in a RBF under an atmosphere of argon (suspension) and stirred at rt. Cyclohexylacetyl chloride (3.00 mL) was added over a period of 30 min and the reaction mixture was stirred at rt o/n. The reaction mixture was added dropwise to 200 mL 10% aqueous NaOAc solution, stirred at rt for 30 min, then extracted into ether (2x100 mL). The combined organic layers were dried (MgS0 4 ), filtered and evaporated to dryness.
  • Step b To a solution of 2-cyclohexyl-1-(2,4-dihydroxyphenyl)ethanone in anhydrous acetonitrile (100 mL) was added under an atmosphere of argon potassium carbonate (4.20 g) and benzyl bromide ( 2.40 mL) and the reaction mixture was stirred at rt for 17 hrs. After this period diethyl ether (300 mL) was added and the mixture was filtered. The filter cake was washed with diethyl ether, and the filtrate was evaporated to dryness to afford 6.15 g of a colourless solid.
  • Step c To a solution of 1-[4-(benzyloxy)-2-hydroxyphenyl]-2-cyclohexylethanone (3000 mg) in 1 ,2-dichloroethane (70 mL) was added 1-bromo-2-chloroethane (1200 ⁇ ),
  • Step d To a solution of 1-[4-(benzyloxy)-2-(2-chloroethoxy)phenyl]-2-cyclohexylethanone (2850 mg) in dioxane/diethyl ether (1 : 1 , 160 mL) was added bromine (450 ⁇ ) dropwise within 2 min, and the mixture was stirred at ambient temperature. The reaction was monitored by LCMS. More bromine (2 x 10 ⁇ ) was added in 10 min intervals to reach complete conversion of 1-[4-(benzyloxy)-2-(2-chloroethoxy)phenyl]-2-cyclohexylethanone.
  • Step e To a solution of methyl 5-amino-1 A7-pyrazole-3-carboxylate hydrochloride (350 mg) in anhydrous DMF (15 ml_) was added under an atmosphere of argon sodium hydride (60% in mineral oil, 175 mg) at 0 °C. The reaction mixture was stirred at this temperature for 20 min followed by addition of a solution of 1-[4-(benzyloxy)-2-(2-chloroethoxy)phenyl]-2-bromo- 2-cyclohexylethanone (810 mg) in anhydrous DMF (30 ml_). The progress of the reaction was monitored by LCMS.
  • reaction mixture was stirred at rt for 17 hrs, then heated at 100 °C for 1 hr. After this period the reaction mixture was cooled to rt, acidified with glacial acetic acid and concentrated on the rotavap (60 °C). The residue was partitioned between water and EtOAc (100 ml_, 1 : 1 ), the aqueous layer was extracted with EtOAc (50 mL), the combined organic layers were washed with brine (20 mL), dried (MgS0 4 ), filtered and evaporated to dryness to leave a brown oil.
  • Step f To a solution of methyl 3-(benzyloxy)-13-cyclohexyl-6,7- dihydropyrazolo[5',1 ':2,3]imidazo[1 ,5-c/][1 ,4]benzoxazepine-10-carboxylate (xiii) (15 mg) in methanol (12 ml_) was added palladium on carbon (10%, 3 mg) and the mixture was stirred under a hydrogen atmosphere (4 L gasbag) for 65 min. Work-up: reaction mixture was flushed with argon and filtered through filter aid (Hiflo medium) with methanol. The filtrate was evaporated to dryness.
  • Step a To a solution of (xiv) (10 mg) in anhydrous DMF (1 mL) was added at 0 °C and under under an atmosphere of argon NaH (1 .3 mg). The mixture was stirred at this temperature for 30 min. A solution of 1-[3-(chloromethyl)-4-(morpholin-4-yl)phenyl]pyrrolidin-
  • Step b A solution of (xviii) (6 mg) in 1 M aqueous KOH/1 ,4-dioxane (3 mL, 1 : 1 ) was heated at 40 °C for 30 min, then stirred at rt for 2 hrs. The reaction mixture was cooled to 0°C, and adjusted to pH 5 with 1 M HCI, and extracted with EtOAc (2x 10 mL). The combined organic layers were washed with brine (5 mL), dried (MgS0 4 ), filtered and evaporated to dryness to afford a solid. The crude material was purified on reverse phase silica by gradient elution (C18; 0-100% acetonitrile/H 2 0) and freeze-dried to afford 2.5 mg (43% yield) 13-cyclohexyl-
  • Step a To a solution of 3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-6- carboxylic acid (iv) (5.0 mg; 0.01 mmol) in DMF (1 mL) at ambient temperature was added ethyl (2E)-3-(4- ⁇ [(1 -aminocyclobutyl)carbonyl] amino ⁇ phenyl)prop-2-enoate hydrochloride (0.02 mmol) followed by HATU (16.7 mg; 0.04 mmol) and DIPEA (5.7 mg; 0.04 mmol).
  • Step b To a solution of ethyl (E)-3-[4-[[1-[[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl- imidazo[1 ,2-b]pyrazole-6-carbonyl]amino]cyclobutanecarbonyl]amino] phenyl]prop-2-enoate (1 ) (4 mg; 0.007 mmol) in MeOH/THF (1 : 1 ; 2 mL) was added a 1 M aqueous solution of lithium hydroxide (0.14 mmol) and the reaction mixture was heated at 60°C for 2 hrs.
  • Step a To a solution of 3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-7- carboxylic acid (vii) (3 mg; 0.01 mmol) in DCM (1 mL) was added ethyl (E)-3-[4-[(1- aminocyclobutanecarbonyl)amino]phenyl]prop-2-enoate (3.8 mg; 0.01 mmol) followed by HATU (10 mg; 0.03 mmol) and DIPEA (3.4 mg; 0.03 mmol).
  • Step b To a solution of ethyl (E)-3-[4-[[1-[[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl- imidazo[1 ,2-b]pyrazole-7-carbonyl]amino]cyclobutanecarbonyl] amino]phenyl]prop-2-enoate (4) (5 mg; 0.01 mmol) in MeOH/THF (1 : 1 ; 2 mL) was added a 1 M aqueous solution of lithium hydroxide (0.16 mmol) and the reaction was heated at 60°C for 3 hrs.
  • Step a To a solution of compound 3-cyclohexyl-1 -methyl-2-phenyl-1 H-imidazo[1 ,2- 6]pyrazole-7-carboxylic acid (x) (10 mg) and ethyl (2E)-3-(4- ⁇ [(1- aminocyclobutyl)carbonyl]amino ⁇ phenyl)prop-2-enoate hydrochloride (15 mg) in dry DMF (500 was added under an atmosphere of argon DIPEA (20 pL) followed by HATU (17.6 mg). The reaction mixture was stirred at rt for 16 hrs. Work-up: EtOAc (15 mL) was added and this mixture was washed with brine (5 mL) and concentrated.
  • Step b A suspension of compound (6) (4.5 mg) in 1 ,4-dioxane/isopropanol/0.6 M aqueous LiOH (0.1 mL/0.1 mL/0.08 mL) was stirred at 60-85 °C for 35 min. Work-up: the reaction mixture was cooled in ice/water, acidified with 1 M HCI and concentrated.
  • Step a To a solution of 13-cyclohexyl-6J-dihydropyrazolo[5',1':2,3]imidazo[1 ,5- c][1,4]benzoxazepine-10-carboxylic acid (xii) (10 mg) and ethyl 1- aminocyclobutanecarboxylate hydrochloride (5.7 mg) in dry DMF (2 mL) under an atmosphere of argon was added DIPEA (16 pL) followed by HATU (13 mg). The reaction mixture was stirred at rt for 17 hrs.
  • Step b A solution of compound (8) (9 mg) in 1 M aquous KOH/1 ,4-dioxane (6 mL, 1 : 1 ) was heated at reflux for 15 min, cooled to 0°C, and adjusted to pH 5 with 1 M HCI and evaporated to dryness.
  • Step c To a solution of compound (9) (7 mg) and ethyl (2E)-3-(4-aminophenyl)prop-2- enoate (3.5 mg) in dry DMF (2 mL) under under an atmosphere of argon was added DIPEA (10 pL) followed by HATU (7.5 mg). The reaction mixture was stirred at rt for 20 hrs. Workup: EtOAc (15 mL) was added and the mixture was washed with brine (5 mL), dried (MgS0 4 ), filtered and concentrated.
  • Step d A suspension of 9 mg compound (10) in 1 ,4-dioxane/isopropanol/0.5 M aqueous LiOH (0.4 mL/0.4 mL/0.32 mL) was stirred in a flask under an atmosphere of argon at 65 °C for 60 min. Work-up: reaction mixture was cooled in ice/water, acidified with 1 M HCI (pH 5) and concentrated.
  • Step a To a solution of 3-cyclohexyl-2-(4-fluorophenyl)-1-methyl-imidazo[1,2-b]pyrazole-6- carboxylic acid (iv) (15 mg; 0.04 mmol) in DCM (1 mL) at ambient temperature was added methyl (2S)-2-amino-3-(4-hydroxyphenyl)propanoate hydrochloride (15 mg; 0.07 mmol), HATU (50 mg; 0.13 mmol) and DIPEA (17 mg; 0.13mmol). After 16 hours a saturated aqueous solution of NaHC0 3 (15 ml) was added and the aqueous layer was extracted with EtOAc (3x 15 mL).
  • Step b To a solution of crude methyl (2S)-2-[[3-cyclohexyl-2-(4-fluorophenyl)-1-methyl- imidazo[1,2-b]pyrazole-6-carbonyl]amino]-3-(4-hydroxyphenyl)propanoate (12) (8 mg) in MeOH/THF (1:1; 2 mL) was added a 1M aqueous solution of lithium hydroxide (0.31 mmol) and the reaction was heated at 60°C for 2 hrs.1M HCI ( aq) was added until ⁇ pH3 and the aqueous layer was extracted with EtOAc (3x 15 mL) and the combined organics were washed with brine (15 mL), dried (MgS0 4 ) and concentrated to leave a brown oil (3 mg).
  • Step a To a solution of 3-cyclohexyl-2-(4-fluorophenyl)-1-methyl-imidazo[1 ,2-b]pyrazole-6- carboxylic acid (iv) (15 mg; 0.04 mmol) in DCM (1 mL) was added methyl (2S)-2-amino-3- (1H-indol-3-yl)propanoate hydrochloride (17 mg; 0.07 mmol) followed by HATU (33 mg; 0.09 mmol) and DIPEA (23 mg; 0.18 mmol). After 2 hours a saturated aqueous solution of NaHC0 3 (15 ml) was added and the aqueous layer was extracted with EtOAc (3x15 mL). The combined organics were washed with brine (10 mL), dried (MgS0 4 ) and concentrated to leave a brown oil (17 mg). Purification of a 4 mg portion by reverse phase LCMS:
  • Step b To a solution of crude methyl N- ⁇ [3-cyclohexyl-2-(4-fluorophenyl)-1-methyl-1H- imidazo[1,2-b]pyrazol-6-yl]carbonyl ⁇ -L-tryptophanate (14) (10 mg) in MeOH/THF (1:1; 2mL) was added a 1M aqueous solution of lithium hydroxide (0.37 mmol).
  • the in vitro and in vivo antiviral activity of the compounds of the invention may be determined using the following protocols.
  • HCV polymerase reactions were carried out using a modified method of Howe et ai , Antimicrobial Agents and Chemotherapy 2004 48(12): 4813-4821.
  • Reactions contained 50 nM NS5bA21 , 20 mM Tris-HCI pH 7.5, 5 rtiM MgCI 2 , 5 mM MnCI 2 , 3 mM DTT, 0.05% BSA, 0.2 U/pL RNasin, 10 ⁇ g mL Poly(rC) template, GTP (at Km) and 0.05 pCi/pL 33 P-GTP in a total reaction volume of 50 pL.
  • a genotype 1 b subgenomic replicon cell line based on Blight et al., Science 2000 290: 1972-1974, modified to express a Renilla luciferase reporter gene was used to assess antiviral activity of test compounds.
  • Cell cultures were maintained in a sub-confluent state in DMEM with glutamine, 10% heat-inactivated foetal bovine serum and Geneticin.
  • Table 2 provides EC 50 values for selected carboxylic esters of the invention.
  • Table 2 EC 50 and IC 50 values for selected compounds of the invention
  • Cytotoxicity of compounds against genotype 1 b subgenomic replicon cells were assessed using the MTT assay (Watanabe et al. , Journal of Virological Methods 1994 48:257-265). Plates were prepared as described for the HCV eplicon assay and cytotoxicity of the test article was evaluated after three days. MTT was added to assay plates followed by three hour incubation at 37 ° C. Wells were aspirated to dryness and the formazan dye dissolved by the addition of 100% isopropanol. Absorbencies were read in a spectrophotometer at two wavelengths (540 nm and 690 nm). The compound concentration that reduced cell viability by 50% (CC 50 ) is calculated using non-linear regression.
  • HCV replicon assays and HCV polymerase assays for genotypes 1 b, 1 a, 2a and 3a to determine the cross-genotype HCV activity of a compound are conducted using essentially the same methods as described above.
  • a genotype 1 b (Con 1 ) subgenomic replicon cell line based on Blight et al. , Science 2000 290: 1972-1974, modified to express a Renilla luciferase reporter gene is used to assess synergy of test compounds.
  • Cell cultures were maintained in a sub-confluent state in DMEM with glutamine, 10% heat-inactivated foetal bovine serum (FBS) and G418 (Geneticin®).
  • Results are analysed and levels of synergy assessed via generation of 3D synergy plots using MacSynergyTM II (Prichard, M. N., K. R. Aseltine, and C. Shipman, Jr. 1993. MacSynergy II. Version 1.0. User's manual. University of Michigan, Ann Arbor.).
  • This method is therefore useful in determining synergistic combinations of an inhibitor of the invention and an HCV inhibitor targeting a different viral protein, or with a different mechanism of inhibiting the NS5B polymerase (for example an NS5A inhibitor, a nucleoside or nucleotide NS5B inhibitor or a NS3/4A protease inhibitor)

Abstract

The present invention relates to viral polymerase inhibitors of formula (I), salts, solvates, hydrates, racemates, enatiomers and isomers thereof, in particular inhibitors of viral polymerases within the Flaviviridae family such as hepatitis C virus (HCV), processes for their preparation and their use in the treatment of Flaviviridae viral infection such as Hepatitis C virus (HCV) infections.

Description

COMPOUNDS FOR THE TREATMENT OF HCV
TECHNICAL FIELD
The present invention relates to viral polymerase inhibitors, in particular inhibitors of viral polymerases within the Flaviviridae family such as hepatitis C virus (HCV), processes for their preparation and their use in the treatment of Flaviviridae viral infections such as Hepatitis C virus (HCV) infections.
BACKGROUND
The Flaviviridae are a group of positive single-stranded RNA viruses with a genome size from 9-15 kb. The Flaviviridae consist of various genera including: Hepaciviruses (this genus contains only one species, the Hepatitis C virus (HCV), which is composed of many genotypes and subtypes); Flaviviruses (this genus includes the Dengue virus, Japanese Tick-Borne and the Yellow Fever virus and there are some additional Flaviviruses that are unclassified) and Pestiviruses (this genus includes three serotypes of bovine viral diarrhoea virus, but no known human pathogens).
Hepatitis C virus (HCV) is a major cause of viral hepatitis and has infected more than 200 million people worldwide. Hepatitis C virus has a positive-strand RNA genome enclosed in a nucleocapsid and lipid envelope. The HCV genome is approximately 9.6 kb in length and encodes a polyprotein of about 3,000 amino acids. There are at least six major genotypes, which have different geographic distributions. In the United States (US), for example, genotypes 1 a and 1 b account for about 75 % of cases, and genotypes 2 and 3 for 10-20 % of cases. Significant differences are observed in the geographic distribution of HCV genotypes. For example, in Europe genotypes 2 and 3 comprise up to one half of cases whereas genotype 3 is thought to dominate in India. In addition, varied genotype
distributions can be observed between countries in a particular region as well as in different areas of a given nation. In the US, HCV is the most common chronic bloodborne infection, affecting approximately 3.2 million persons. After infection with HCV, approximately 75-85% of people develop chronic infection, whilst 60-70% develop chronic liver disease. Of these, 5-20% go on to develop cirrhosis over a period of 20-30 years, and, finally, 1-5% succumb to the consequences of chronic infection (liver cancer/cirrhosis).
Until recently, the only treatment option for HCV was 24 or 48 weeks of combination therapy consisting of weekly injections of pegylated interferon (peg-IFN) and oral ribavirin for 24 or 48 weeks. The best treatment response is seen in patients with HCV genotypes 2 and 3, in whom sustained viral response (SVR) rates of approximately 80% can be achieved with 24 weeks of therapy. Patients with HCV genotype 1 remain the most difficult to treat, with SVR rates of approximately 40% after 48 weeks of therapy. In addition to the low response rates, combination peg-IFN/ribavirin therapy is limited by serious side effects, including fatigue, influenza-like symptoms, depression and suicide with peg-I FN, and haemolytic anaemia with ribavirin. Furthermore, peg-IFN/ribavirin therapy is contra-indicated in patients who have depression, anaemia, HCV-related decompensated cirrhosis, alcohol/substance abuse and autoimmune disorders or who are pregnant.
New treatment options for HCV became available in May 201 1 with the US launch of the first direct-acting antiviral (DAA) HCV drugs, telaprevir (Vertex Pharmaceuticals) and boceprevir (Merck). Both drugs are protease inhibitors and are approved for the treatment of chronic HCV genotype 1 infection in combination with peg-IFN and ribavirin. Pivotal phase 3 trials demonstrated that the addition of telaprevir or boceprevir to peg-IFN/RBV therapy achieved shortened durations of therapy and potent viral suppression, with SVR rates approaching 75% in genotype 1 treatment-naive patients and 30% to 85% in treatment-experienced patients.
However, addition of a third drug to the treatment regimen has resulted in increased adverse events. Telaprevir is associated with an increased incidence of rash and anaemia, while boceprevir is associated with anaemia and dysgeusia. Triple therapy with telaprevir or boceprevir and peg-IFN/ribavirin remains unsuitable for those intolerant to or with contraindications to peg-I FN/ribavirin therapy.
Due to the limited tolerability, efficacy, side effects, genotype coverage and concern over the emergence of resistance there is an ongoing need to find alternative agents for the treatment of HCV. The majority of compounds that are currently in development have a limited spectrum of activity against the various HCV genotypes and, in many cases, are only active against HCV genotypes 1 b and/or 1 a.
The HCV genome possesses structural (core) and non-structural (NS2, NS3, NS4A, NS4B, NS5A and NS5B) proteins. The non-structural proteins are involved in viral genomic replication, with the initial synthesis of RNA carried out by NS5B RNA dependent RNA polymerase. The NS5B protein is a key target for anti-HCV therapy, as it is essential for HCV replication and has no human host equivalent. This protein has been well characterised and is a validated target for drug discovery.
HCV therapy is also anticipated to evolve towards oral multidrug therapy, in which combinations of different DAA drugs with complementary mechanisms of action serve to increase viral suppression and delay or prevent the emergence of resistance.
Accordingly, there remains on ongoing need for HCV agents, particularly with targeted mechanisms of action such as NS5B inhibitors. There is also an unmet need for HCV agents with cross-genotypic activity against genotypes 1 , 2 and 3. SUMMARY
The inventors have found a new class of antiviral compounds, more particularly NS5B polymerase inhibitors, for the treatment of HCV infections.
Compounds of the present invention are therefore considered to be useful in treating and preventing hepatitis C infections when used on their own or in combination with one or more other antiviral agents such as ribavirin, an antiviral nucleoside, polymerase inhibitor, protease inhibitor and/or inhibitor of viral entry, assembly or egress. The combination may also additionally comprise at least one immunomodulatory agent for example an interferon or interferon derivative and/or an inhibitor of inosine-5'-monophosphate dehydrogenase (IMPDH).
It is also believed that compounds of the invention will be efficacious in combination with at least one other DAA with a different mechanism of action and a complementary resistance profile (for example an NS5A inhibitor, a nucleoside or nucleotide NS5B inhibitor or a NS3/4A protease inhibitor) thereby offering an alternative treatment regime for patients not eligible for or treatable with the recently approved triple combination therapy.
According to a first aspect there is provided a compound of formula (I), salts, N- oxides, solvates, hydrates, racemates, enantiomers and isomers thereof:
Figure imgf000004_0001
(I)
wherein
1 is H or Ra or R1 together with R2 and the atoms to which they are attached form an optionally substituted 6-8 membered carbocyclic ring which may be optionally interrupted by one or more heteroatoms independently selected from O, N and S;
R2 is H or Rb or R2 together with Ri joins to form an optionally substituted 6-8 membered carbocyclic ring which may be optionally interrupted by one or more heteroatoms independently selected from O, N and S;
R3 is an optionally substituted C3-8cycloalkyl group;
Xi, X2 and X3 are each independently selected from N, CH, C-Rc and C-A-R4 provided that at least one of Xi , X2 and X3 is C-A-R4;
A is a covalent bond, -CONH-C(R5)(R6)-B- or -CONCi-aalkyl-CiRsXReJ-B-;
R4 is selected from H, C02H, optionally substituted C02Ci_6alkyl, optionally substituted aryl, optionally substituted arylCi_6alkyl, optionally substituted arylC2.6alkenyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted heterocyclylC^alkyl, optionally substituted heteroarylC^alkyl, optionally substituted heterocyclylC2-6alkenyl and optionally substituted heteroarylC2-6alkenyl;
R5 and R6 are each independently selected from H, optionally substituted C^alkyl, optionally substituted aryl, optionally substituted C^alkylaryl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted C1.6alkylheterocyclyl and optionally substituted C^alkylheteroaryl or R5 and R6 together with the carbon atom to which they are attached form an optionally substituted C3.6cycloalkyl group;
B is a covalent bond or -CONH-R7- where R7 is selected from optionally substituted aryl, optionally substituted arylCi-6alkyl, optionally substituted arylC2.6alkenyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted
heterocyclylC^alkyl, optionally substituted heteroarylC^alkyl, optionally substituted heterocyclylC2-6alkenyl and optionally substituted heteroarylC2-6alkenyl;
Z1 is C or N;
Z2, Z3, Z4 and Z5 are each independently selected from N, CH and C-Rd;
Ra, Rb, Rc and Rd are each independently selected from C^alkyl, C2.6alkenyl, C2. 6alkynyl, C3-8cycloalkyl, aryl, Ci_6alkylaryl, C^alkoxyaryl, heterocyclyl, heteroaryl, halo, Ci_ 6alkylhalo, OH, Ci-6alkylOH, Ci_6alkoxy, Ci_6alkoxyhalo, oxy (C=0), C02R8, OCOR8, C(=0)R8, CN, N02, N(R8)2, NR8COR8, CONR8, SR8, thio (=S), SC^d-ealkyl, S02NR8R8, NR8S02R8;
wherein each R8 is independently selected from H and C1.6alkyl and each alkyl, alkenyl, alkynyl, cycloalkyi, aryl, heterocyclyl and heteroaryl in the definition of Ra, Rb, Rc and Rd may be optionally substituted;
and further wherein each heterocyclyl or heteroaryl has 1 , 2, 3 or 4 heteroatoms independently selected from O, S and N.
In a preferred embodiment, C-A-R4 is not C-H.
In a second aspect there is provided a process for producing a compound of general formula (I) wherein A is -CONH-C(R5)(R6)-B- and B is CONH-R7- comprising the step of reacting a compound of formula (II) with NH2R7 under amidation conditions
Figure imgf000005_0001
(ll) wherein ΧΊ , X'2 and X'3 are each independently selected from N, CH, C-Rc, C-A-R'4 provided that at least one of ΧΊ , X'2 and X'3 is C-A-R'4 wherein R'4 is C02H or COaC^alkyl; and
Ri, R2, R3, R5, R6, R7, C-Rc, Zi, Z2, Z3, Z4 and Z5 are as defined above.
The compounds of formula (I) are inhibitors of HCV. In particular, the compounds of formula (I) inhibit RNA synthesis by the RNA dependent RNA polymerase of HCV (the NS5B protein encoded by HCV). NS5B polymerase inhibitors have been clinically validated as potential antiviral agents for the treatment of HCV infection.
I n a third aspect, there is provided a pharmaceutical agent comprising the compound of formula (I) defined above and pharmaceutically acceptable salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
There is also provided use of the compound of formula (I) defined above and pharmaceutically acceptable salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof as a pharmaceutical agent.
There is further provided the compound of formula (I) defined above and pharmaceutically acceptable salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof for use as a pharmaceutical agent.
The pharmaceutical agent may be an antiviral agent.
I n a fourth aspect, there is provided a viral polymerase inhibitor in particular a HCV polymerase inhibitor such as a NS5B polymerase inhibitor comprising the compound of formula (I) defined above and pharmaceutically acceptable salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
There is also provided use of the compound of formula (I) defined above and pharmaceutically acceptable salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof as a viral polymerase inhibitor in particular a HCV polymerase inhibitor such as a NS5B polymerase inhibitor.
There is further provided the compound of formula (I) defined above and pharmaceutically acceptable salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof for use as a viral polymerase inhibitor in particular a HCV polymerase inhibitor such as a NS5B polymerase inhibitor.
The compound of formula (I ) and pharmaceutically acceptable salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof may be administered in the form of a pharmaceutical composition together with a pharmaceutically acceptable carrier.
I n a fifth aspect, there is provided a pharmaceutical composition comprising the compound of formula (I) and pharmaceutically acceptable salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof and a pharmaceutically acceptable carrier.
According to one embodiment, the pharmaceutical composition additionally comprises a therapeutically effective amount of one or more antiviral agents such as at least one other anti-HCV agent.
In a sixth aspect, there is provided a method for the treatment of a Flaviviridae viral infection such as a HCV infection which comprises administering an effective amount of the compound of formula (I) and pharmaceutically acceptable salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof or the pharmaceutical composition defined above to a subject diagnosed with, suffering from or at risk of developing said viral infection.
There is also provided use of the compound of formula (I) and pharmaceutically acceptable salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof or the pharmaceutical composition as defined above in the manufacture of a medicament for use in the treatment of a Flaviviridae viral infection such as a HCV infection.
There is further provided use of the compound of formula (I) and pharmaceutically acceptable salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof or the pharmaceutical composition as defined above in the treatment of a Flaviviridae viral infection such as a HCV infection.
There is still further provided the compound of the formula (I) and pharmaceutically acceptable salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof or the pharmaceutical composition defined above for use in the treatment of a Flaviviridae viral infection such as a HCV infection.
In a seventh aspect, there is provided a method of inhibiting the RNA-dependent RNA polymerase activity of the enzyme NS5B, encoded by HCV, comprising exposing the enzyme NS5B to an effective amount of the compound of formula (I) defined above and pharmaceutically acceptable salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
In an eighth aspect, there is provided a method of inhibiting HCV replication comprising exposing a cell infected with HCV to an effective amount of the compound of formula (I) defined above and pharmaceutically acceptable salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
DETAILED DESCRIPTION
The present invention relates to compounds of formula (I) which inhibit viral polymerases and are useful in the treatment of Flaviviridae viral infections, particularly, hepatitis C (HCV). Compounds
The present invention relates to compounds of formula I, salts, N-oxides, racemates, enantiomers and isomers thereof as defined above.
In one embodiment R1 is H or d-6alkyl, preferably methyl, ethyl or propyl, more preferably H or methyl and R2 is H or Rb, preferably H.
In another embodiment R1 together with R2 joins to form an optionally substituted 6-8 membered carbocyclic ring which may be optionally interrupted by one or more heteroatoms independently selected from O, N and S. Preferably R1 together with R2 joins to form a 7- membered carbocycle. In a particular embodiment and R2 together are a divalent linking moiety selected from the group consisting of -CH2CH2CH2-; -OCH2CH2-; -CH2OCH2-; - CH2CH20-; -NCH2CH2-; -CH2NCH2-; and -CH2CH2N-, thereby joining to form a 7- membered carbocyclic ring together with the atoms to which R1 and R2 are attached the carbocyclic ring being optionally interrupted by O or N, preferably O. In a particularly preferred embodiment and R2 together are a divalent linking moiety of formula - CH2CH20-.,
In another embodiment R3 is an optionally substituted C3.6cycloalkyl group, preferably cyclohexyl.
In one embodiment A is a covalent bond and R4 is C02H or optionally substituted C02Ci_6alkyl.
In one embodiment X2 and X3 are each independently selected from N, CH, C-Rc or C-A-R4 provided that at least one of Xi , X2 and X3 is C-A-R4 and A is a covalent bond and R4 is C02H or optionally substituted C02C1.6alkyl.
In one embodiment X1 is C-A-R4; X2 and X3 are each independently selected from N, CH or C-Rc preferably N or CH.
In another embodiment X2 is C-A-R4 and X1 and X3 are each independently selected from N, CH or C-Rc preferably N or CH.
In yet another embodiment X3 is C-A-R4 and X1 and X2 are each independently selected from N, CH or C-Rc preferably N or CH.
In one embodiment R4 is selected from H, C02H and optionally substituted CO^. 6alkyl. In a further embodiment R4 is C02H or CC^C^al yl, preferably C02H or CO^. 3alkyl, more preferably R4 is selected from C02H, C02CH3 and C02CH2CH3.
In one embodiment A is a covalent bond.
In another embodiment A is -CONH-C(R5)(R6)-B- wherein R5 and R6 are each independently selected from H, C^alkyl (preferably C^alkyl, more preferably methyl), C^ 6alkylaryl (preferably optionally substituted benzyl); C^alkylheterocyclyl and Ci_
6alkylheteroaryl (preferably CH2heteroaryl, wherein the heteroaryl is preferably a 5-, 6- or 9- membered heteroaryl, more preferably a 5-, 6- or 9-membered N-containing heteroaryl) or R5 and R6 together with the carbon atom to which they are attached form a cyclobutyl group.
In one embodiment B is a covalent bond.
In another embodiment B is -CONHR7- wherein R7 is selected from aryl (preferably phenyl), arylC^alkyl (preferably phenylC^alkyl) and arylC2-6alkenyl (preferably phenylC2_ 3alkenyl). In a particularly preferred embodiment R7 is phenylethenyl.
In one embodiment the compound of formula (I) is a compound of formula (la)
Figure imgf000009_0001
(la)
wherein Z3 is CH or C-Rd and Ri , R2, R3, Rd, X2 and X3 are as previously defined and salts, A/-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof. In a preferred embodiment when Z3 is C-Rd then Rd is selected from the group consisting of d- 3alkyl (preferably methyl), aryl (preferably phenyl), alkylaryl (preferably benzyl), d- 3alkoxyaryl (preferably benzyloxy), heterocyclyl, heteroaryl (preferably 5-membered or 6- membered heteroaryl), halo (preferably F), haloC^alkyl (preferably CHF2 and CF3), OH, d- 3alkoxy (preferably OCH3), halod-salkoxy (preferably OCHF2 and OCF3), oxy (C=0), carboxy (preferably C02H and C02d-3alkyl preferably C02methyl or C02ethyl), CN, N02, NH2, d-6alkylamino (preferably NHd salky)! and diC1.6alkylamino (preferably N(d.3alkyl)2), wherein each alkyl, aryl, heterocyclyl and heteroaryl may be further optionally substituted.
In a particularly preferred embodiment Rd is selected from the group consisting of halo, OH, d_3alkoxy or d_3alkoxyaryl wherein the aryl is unsubstituted or substituted with heterocyclyl (preferably 5 or 6 membered heterocyclyl containing N such as morpholine or pyrrolidinone). F, OH, OCH3 and optionally substituted benzyloxy including unsubstituted benzyloxy and 2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1 -yl)benzyloxy are particularly preferred Rd groups when Z3 is C-Rd.
In one embodiment of formula (la), there is provided compounds selected from the group of compounds of formula (la-i) to (la-vi):
Figure imgf000009_0002
(la-i) (la-ii) (la-iii)
Figure imgf000010_0001
(la-iv) (la-v) (la-vi)
wherein R^ R2, R3, Z3 ^ , X2 and X3 are as previously defined and salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
I n another embodiment the compounds of formulae (la) are compounds of formula
(la-i) and (la-vi) selected from the group consisting of compounds of formulae:
Figure imgf000010_0002
wherein A, R2, R3, R4, Z^ X^ , X2 and X3 are as previously defined provided that A- R4 is not H (i.e. where A is a covalent bond and R4 is H) and salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof
I n one embodiment there is provided a compound selected from the group consisting of
methyl 3-cyclohexyl-2-(4-fluorophenyl)-1 H-imidazo[1 ,2-b]pyrazole-6-carboxylate (i);
3-cyclohexyl-2-(4-fluorophenyl)-1 H-imidazo[1 ,2-b]pyrazole-6-carboxylic acid (ii); methyl 3- cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-6-carboxylate (iii);
3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazole-6-carboxylic acid (iv); ethyl 3-cyclohexyl-2-(4-fluorophenyl)-1 H-imidazo[1 ,2-b]pyrazole-7-carboxylate (v); ethyl 3- cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-7-carboxylate (vi);
3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazole-7-carboxylic acid (vii);
3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazole (viii);
ethyl 3-cyclohexyl-1 -methyl-2-phenyl-1 H-im idazo[1 ,2-b]pyrazole-7-carboxylate (ix);
3-cyclohexyl-1 -methyl-2-phenyl-1 H-imidazo[1 ,2-b]pyrazole-7-carboxylic acid (x);
methyl 13-cyclohexyl-6,7-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10- carboxylate (xi); 13-cyclohexyl-6J-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10-carboxylic acid (xii);
methyl 3-(benzyloxy)-13-cyclohexyl-6J-dihydropyrazolo[5', 1 ':2,3] imidazo[1 ,5- d][1 ,4]benzoxazepine-10-carboxylate (xiii);
methyl 13-cyclohexyl-3-hydroxy-6,7-dihydropyrazolo[5',1 ':2,3] imidazo[1 ,5- d][1 ,4]benzoxazepine-10-carboxylate (xiv);
3-(benzyloxy)-13-cyclohexyl-6J-dihydropyrazolo[5',1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine- 10-carboxylic acid (xv);
13-cyclohexyl-3-hydroxy-6J-dihydropyrazolo[5',1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10- carboxylic acid (xvi);
ethyl 3-(benzyloxy)-13-cyclohexyl-6J-dihydroimidazo[2', 1 ':2,3]imidazo [1 ,5- d][1 ,4]benzoxazepine-10-carboxylate (xvii);
methyl 13-cyclohexyl-3-{[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1-yl)benzyl]oxy}-6J- dihydropyrazolo[5',1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10-carboxylate (xviii);
13-cyclohexyl-3-{[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1 -yl)benzyl]oxy}-6,7- dihydropyrazolo[5',1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10-carboxylic acid (xix);
3-(benzyloxy)-13-cyclohexyl-6J-dihydroimidazo[2', 1 ':2,3]imidazo [1 ,5-d][1 ,4]benzoxazepine- 10-carboxylic acid (xx);
ethyl 13-cyclohexyl-3-hydroxy-6J-dihydroimidazo[2', 1 ':2,3]imidazo[1 ,5- d][1 ,4]benzoxazepine-10-carboxylate (xxi);
ethyl 13-cyclohexyl-3-{[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1-yl)benzyl]oxy}-6J- dihydroimidazo[2', 1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10-carboxylate (xxii);
13-cyclohexyl-3-{[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1 -yl)benzyl]oxy}-6,7- dihydroimidazo[2', 1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10-carboxylic acid (xxiii);
ethyl (2E)-3-[4-({[1 -({[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazol-6- yl]carbonyl}amino)cyclobutyl]carbonyl} amino)phenyl]prop-2-enoate (1 );
(E)-3-[4-[[1 -[[3-cyclohexyl-2-(4-fluorophenyl)-1-methyl-imidazo[1 ,2-b]pyrazole-6- carbonyl]amino]cyclobutanecarbonyl] amino]phenyl]prop-2-enoic acid (2);
methyl (E)-3-[4-[[1-[[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-6- carbonyl]amino] cyclobutanecarbonyl]amino]phenyl]prop-2-enoate (3);
ethyl (E)-3-[4-[[1 -[[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-7- carbonyl]amino]cyclobutanecarbonyl] amino]phenyl]prop-2-enoate (4); (2E)-3-[4-({[1 -({[3- cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazol-7- yl]carbonyl}amino)cyclobutyl]carbonyl}amino)phenyl]prop-2-enoic acid (5);
ethyl (2E)-3-(4-{[(1 -{[(3-cyclohexyl-1 -methyl-2-phenyl-1 H-imidazo[1 ,2-b]pyrazol-7- yl)carbonyl]amino}cyclobutyl) carbonyl]amino}phenyl)prop-2-enoate (6); (2E)-3-(4-{[(1-{[(3-cyclohexyl-1 -methyl-2-phenyl-1 H-imidazo[1 ,2-b]pyrazol-7- yl)carbonyl]amino}cyclobutyl) carbonyl]amino}phenyl)prop-2-enoic acid (7);
ethyl 1 -{[(13-cyclohexyl-6J-dihydropyrazolo[5', 1':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepin-10- yl)carbonyl]amino}cyclobutanecarboxylate (8);
1 -{[(13-cyclohexyl-6,7-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepin-10- yl)carbonyl]amino}cyclobutanecarboxylic acid (9);
ethyl (2E)-3-(4-{[(1 -{[(13-cyclohexyl-6J-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5- d][1 ^]benzoxazepin-10-yl)carbonyl]amino}cyclobutyl)carbonyl]amino}phenyl)prop-2-enoate
(10);
(2E)-3-(4-{[(1-{[(13-cyclohexyl-6J-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5- d][1 ^]benzoxazepin-10-yl)carbonyl]amino}cyclobutyl)carbonyl]amino}phenyl)prop-2-enoic acid (11 );
methyl (2S)-2-[[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-6- carbonyl]amino]-3-(4-hydroxyphenyl)propanoate (12);
N-{[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazol-6-yl]carbonyl}-L- tyrosine (13);
methyl N-{[3-cyclohexyl-2-(4-fluorophenyl)-1-methyl-1 H-imidazo[1 ,2-b]pyrazol-6-yl]carbonyl}- L-tryptophanate (14);
N-{[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazol-6-yl]carbonyl}-L- tryptophan (15); and
3-cyclohexyl-2-(4-fluorophenyl)-N-[2-(1 H-indol-3-yl)ethyl]-1 -methyl-1 H-imidazo[1 ,2- b]pyrazole-7-carboxamide (16);
and salts, A/-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
The term "Chalky!" refers to optionally substituted straight chain or branched chain hydrocarbon groups having from 1 to 6 carbon atoms. Examples include methyl (Me), ethyl (Et), propyl (Pr), isopropyl (/-Pr), butyl (Bu), isobutyl (/-Bu), sec-butyl (s-Bu), tert-butyl (f-Bu), pentyl, neopentyl, hexyl and the like. Unless the context requires otherwise, the term "d. 6alkyl" also encompasses alkyl groups containing one less hydrogen atom such that the group is attached via two positions i.e. divalent.
Figure imgf000012_0001
and "C1.3alkyl" including methyl, ethyl, propyl, /sopropyl, n-butyl, /'so-butyl, sec-butyl and ferf-butyl are preferred with methyl being particularly preferred.
The term "C2-6alkenyl" refers to optionally substituted straight chain or branched chain hydrocarbon groups having at least one double bond of either E or Z stereochemistry where applicable and 2 to 6 carbon atoms. Examples include vinyl, 1 -propenyl, 1- and 2- butenyl and 2-methyl-2-propenyl. Unless the context requires otherwise, the term "C2.
6alkenyl" also encompasses alkenyl groups containing one less hydrogen atom such that the group is attached via two positions i.e. divalent. "C2-4alkenyl" and "C2_3alkenyl" including ethenyl, propenyl and butenyl are preferred with ethenyl being particularly preferred.
The term "C2-6alkynyl" refers to optionally substituted straight chain or branched chain hydrocarbon groups having at least one triple bond and 2 to 6 carbon atoms.
Examples include ethynyl, 1 -propynyl, 1 - and 2-butynyl, 2-methyl-2-propynyl, 2-pentynyl, 3- pentynyl, 4-pentynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl and 5-hexynyl and the like. Unless the context indicates otherwise, the term "C2-6alkynyl" also encompasses alkynyl groups containing one less hydrogen atom such that the group is attached via two positions i.e. divalent. C2.3alkynyl is preferred.
The term "C3.8cycloalkyl" refers to non-aromatic cyclic groups having from 3 to 8 carbon atoms, including cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl. It will be understood that cycloalkyl groups may be saturated such as cyclohexyl or unsaturated such as cyclohexenyl. C3.6cycloalkyl such as cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl are preferred.
The terms "hydroxy" and "hydroxyl" refer to the group -OH.
The term "oxo" refers to the group =0.
The term "Ci_6alkoxy" refers to an alkyl group as defined above covalently bound via an O linkage containing 1 to 6 carbon atoms, such as methoxy, ethoxy, propoxy, isoproxy, butoxy, tert-butoxy and pentoxy. "C1.4alkoxy" and "C^alkoxy" including methoxy, ethoxy, propoxy and butoxy are preferred with methoxy being particularly preferred.
The term "Ci_6alkylhalo" refers to a Ci_6alkyl which is substituted with one or more halogens. d.3alkylhalo groups are preferred, such as for example, -CHF2 and -CF3.
The term "Ci_6alkoxylhalo" refers to a C^alkoxyl which is substituted with one or more halogens. d-3alkoxylhalo groups are preferred, such as for example, -OCHF2 and - OCF3.
The term "carboxylate" or "carboxyl" refers to the group -COO" or -COOH.
The term "ester" refers to a carboxyl group having the hydrogen replaced with, for example a C^alkyl group ("carboxylCi.6alkyl" or "alkylester"), an aryl or aralkyl group ("arylester" or "aralkylester") and so on. C02Ci_3alkyl groups are preferred, such as for example, methylester (CO 2Me), ethylester (C02Et) and propylester (C02Pr) and includes reverse esters thereof (e.g. -OCOMe, -OCOEt and -OCOPr).
The term "cyano" refers to the group -CN.
The term "nitro" refers to the group -N02.
The term "amino" refers to the group -NH2.
The term "substituted amino" or "secondary amino" refers to an amino group having a hydrogen replaced with, for example a C^alkyl group ("C^alkylamino"), an aryl or aralkyl group ("arylamino", "aralkylamino") and so on. C^alkylamino groups are preferred, such as for example, methylamino (NHMe), ethylamino (NHEt) and propylam ino (NHPr).
The term "disubstituted amino" or "tertiary amino" refers to an amino group having the two hydrogens replaced with, for example a C^alkyl group, which may be the same or different ("dialkylamino"), an aryl and alkyl group ("aryl(alkyl)amino") and so on. D'\(C†.
3alkyl)amino groups are preferred, such as for example, dimethylamino (NMe2), diethylamino (NEt2), dipropylamino (NPr2) and variations thereof (e.g. N(Me)(Et) and so on).
The term "acyl" or "aldehyde" refers to the group -C(=0)H.
The term "substituted acyl" or "ketone" refers to an acyl group having a hydrogen replaced with, for example a C^alkyl group ("Ci.6alkylacyl" or "alkylketone" or "ketoalkyl"), an aryl group ("arylketone"), an aralkyl group ("aralkylketone) and so on. Ci_3alkylacyl groups are preferred.
The term "amido" or "amide" refers to the group -C(0)NH2.
The term "aminoacyl" refers to the group -NHC(0)H.
The term "substituted amido" or "substituted amide" refers to an amido group having a hydrogen replaced with, for example a C^alkyl group ("Ci_6alkylamido" or "Ci_ 6alkylam ide"), an aryl ("arylamido"), aralkyl group ("aralkylamido") and so on. C^alkylamide groups are preferred, such as for example, methylamide (-C(O)NHMe), ethylamide (- C(O)NHEt) and propylamide (-C(O)NHPr) and includes reverse amides thereof (e.g. - NHMeC(O)-, -NHEtC(O)- and -NHPrC(O)-).
The term "disubstituted amido" or "disubstituted amide" refers to an amido group having the two hydrogens replaced with, for example a Ci_6alkyl group ("d C^ealky amido" or "diiC^ealkylJamide"), an aralkyl and alkyl group ("alkyl(aralkyl)am ido") and so on. Di(d. 3alkyl)amide groups are preferred, such as for example, dimethylamide (-C(0)NMe2), diethylamide (-C(0)NEt2) and dipropylamide ((-C(0)NPr2) and variations thereof (e.g. - C(0)N(Me)Et and so on) and includes reverse amides thereof.
The term "thiol" refers to the group -SH.
The term "Ci_6alkylthio" refers to a thiol group having the hydrogen replaced with a Ci-6alkyl group. Ci_3alkylthio groups are preferred, such as for example, thiolmethyl, thiolethyl and thiolpropyl.
The term "thioxo" refers to the group =S.
The term "sulfinyl" refers to the group -S(=0)H.
The term "substituted sulfinyl" or "sulfoxide" refers to a sulfinyl group having the hydrogen replaced with, for example a C^alkyl group ("C^alkylsulfinyl" or "Ci_
6alkylsulfoxide"), an aryl ("arylsulfinyl"), an aralkyl ("aralkyl sulfinyl") and so on. d_ 3alkylsulf inyl groups are preferred, such as for example, -SOmethyl, -SOethyl and - SOpropyl.
The term "sulfonyl" refers to the group -S02H.
The term "substituted sulfonyl" refers to a sulfonyl group having the hydrogen replaced with, for example a C^alkyl group ("sulfonylC^ealkyl"), an aryl ("arylsulfonyl"), an aralkyl ("aralkylsulfonyl") and so on. SulfonylC^alkyl groups are preferred, such as for example, -S02Me, -S02Et and -S02Pr.
The term "sulfonylamido" or "sulfonamide" refers to the group -S02NH2.
The term "substituted sulfonamido" or "substituted sulphonamide" refers to an sulfonylamido group having a hydrogen replaced with, for example a C^alkyl group
("sulfonylamidoCi.6alkyl"), an aryl ("arylsulfonamide"), aralkyl ("aralkylsulfonamide") and so on. SulfonylamidoC^alkyl groups are preferred, such as for example, -S02NHMe, - S02NHEt and -S02NHPr and includes reverse sulfonamides thereof (e.g. -NHS02Me, - NHS02Et and -NHS02Pr).
The term "disubstituted sufonamido" or "disubstituted sulphonamide" refers to an sulfonylamido group having the two hydrogens replaced with, for example a C^alkyl group, which may be the same or different ("sulfonylamidodi(C1.6alkyl)"), an aralkyl and alkyl group ("sulfonamido(aralkyl)alkyl") and so on. Sulfonylamidod C^alkyl) groups are preferred, such as for example, -S02NMe2, -S02NEt2 and -S02NPr2 and variations thereof (e.g. - S02N(Me)Et and so on) and includes reserve sulfonamides thereof.
The term "sulfate" refers to the group OS(0)2OH and includes groups having the hydrogen replaced with, for example a C1.6alkyl group ("alkylsulfates"), an aryl ("arylsulfate"), an aralkyl ("aralkylsulfate") and so on. (^sulfates are preferred, such as for example, OS(0)2OMe, OS(0)2OEt and OS(0)2OPr.
The term "sulfonate" refers to the group S03H and includes groups having the hydrogen replaced with, for example a C1.6alkyl group ("alkylsulfonate"), an aryl
("arylsulfonate"), an aralkyl ("aralkylsulfonate") and so on. C 1.3s u If o nates are preferred, such as for example, S03Me, S03Et and S03Pr.
The term "aryl" refers to a carbocyclic (non-heterocyclic) aromatic ring or mono-, bi- or tri-cyclic ring system. The aromatic ring or ring system is generally composed of 6 to 10 carbon atoms. Examples of aryl groups include but are not limited to phenyl, biphenyl, naphthyl and tetrahydronaphthyl. 6-membered aryls such as phenyl are preferred. The term "alkylaryl" refers to C^alkylaryl such as benzyl. The term "alkoxyaryl" refers to Ci_
6alkyloxyaryl such as benzyloxy.
The term "heterocyclyl" refers to a moiety obtained by removing a hydrogen atom from a ring atom of a heterocyclic compound which moiety has from 3 to 10 ring atoms (unless otherwise specified), of which 1 , 2, 3 or 4 are ring heteroatoms each heteroatom being independently selected from O, S and N.
In this context, the prefixes 3-, 4-, 5-, 6-, 7-, 8-, 9- and 10- membered denote the number of ring atoms, or range of ring atoms, whether carbon atoms or heteroatoms. For example, the term "3-10 membered heterocylyl", as used herein, pertains to a heterocyclyl group having 3, 4, 5, 6, 7, 8, 9 or 10 ring atoms. Examples of heterocylyl groups include 5- 6-membered monocyclic heterocyclyls and 9-10 membered fused bicyclic heterocyclyls.
Examples of monocyclic heterocyclyl groups include, but are not limited to, those containing one nitrogen atom such as aziridine (3-membered ring), azetidine (4-membered ring), pyrrolidine (tetrahydropyrrole), pyrroline (e.g., 3-pyrroline, 2,5-dihydropyrrole), 2H- pyrrole or 3H-pyrrole (isopyrrole, isoazole) or pyrrolidinone (5-membered rings) , piperidine, dihydropyridine, tetrahydropyridine (6-membered rings), and azepine (7-membered ring); those containing two nitrogen atoms such as imidazoline, pyrazolidine (diazolidine), imidazoline, pyrazoline (dihydropyrazole) (5-membered rings), piperazine (6-membered ring); those containing one oxygen atom such as oxirane (3-membered ring), oxetane (4- membered ring), oxolane (tetrahydrofuran), oxole (dihydrofuran) (5-membered rings), oxane (tetrahydropyran), dihydropyran, pyran (6-membered rings), oxepin (7-membered ring); those containing two oxygen atoms such as dioxolane (5-membered ring), dioxane (6- membered ring), and dioxepane (7-membered ring); those containing three oxygen atoms such as trioxane (6-membered ring); those containing one sulfur atom such as thiirane (3- membered ring), thietane (4-membered ring), thiolane (tetrahydrothiophene) (5-membered ring), thiane (tetrahyd roth io pyran) (6-membered ring), thiepane (7-membered ring); those containing one nitrogen and one oxygen atom such as tetrahydrooxazole, dihydrooxazole, tetrahydroisoxazole, dihydroisoxazole (5-membered rings), morpholine, tetrahydrooxazine, dihydrooxazine, oxazine (6-membered rings); those containing one nitrogen and one sulfur atom such as thiazoline, thiazolidine (5-membered rings), thiomorpholine (6-membered ring); those containing two nitrogen and one oxygen atom such as oxadiazine (6-membered ring); those containing one oxygen and one sulfur such as: oxathiole (5-membered ring) and oxathiane (thioxane) (6-membered ring); and those containing one nitrogen, one oxygen and one sulfur atom such as oxathiazine (6-membered ring).
Heterocyclyls also encompass aromatic heterocyclyls and non-aromatic heterocyclyls. Such groups may be substituted or unsubstituted.
The term "aromatic heterocyclyl" may be used interchangeably with the term "heteroaromatic" or the term "heteroaryl" or "hetaryl". The heteroatoms in the aromatic heterocyclyl group may be independently selected from N, S and O. "Heteroaryl" is used herein to denote a heterocyclic group having aromatic character and embraces aromatic monocyclic ring systems and polycyclic (e.g. bicyclic) ring systems containing one or more aromatic rings. The term aromatic heterocyclyl also encompasses pseudoaromatic heterocyclyls. The term "pseudoaromatic" refers to a ring system which is not strictly aromatic, but which is stabilized by means of delocalization of electrons and behaves in a similar manner to aromatic rings. The term aromatic heterocyclyl therefore covers polycyclic ring systems in which all of the fused rings are aromatic as well as ring systems where one or more rings are non-aromatic, provided that at least one ring is aromatic. In polycyclic systems containing both aromatic and non-aromatic rings fused together, the group may be attached to another moiety by the aromatic ring or by a non- aromatic ring.
Examples of heteroaryl groups are monocyclic and bicyclic groups containing from five to ten ring members. The heteroaryl group can be, for example, a five membered or six membered monocyclic ring or a bicyclic structure formed from fused five and six membered rings or two fused six membered rings or two fused five membered rings. Each ring may contain up to about four heteroatoms typically selected from nitrogen, sulphur and oxygen. The heteroaryl ring will contain up to 4 heteroatoms, more typically up to 3 heteroatoms, more usually up to 2, for example a single heteroatom. In one embodiment, the heteroaryl ring contains at least one ring nitrogen atom. The nitrogen atoms in the heteroaryl rings can be basic, as in the case of an imidazole or pyridine, or essentially non-basic as in the case of an indole or pyrrole nitrogen. In general the number of basic nitrogen atoms present in the heteroaryl group, including any amino group substituents of the ring, will be less than five.
Aromatic heterocyclyl groups may be 5-membered or 6-membered mono-cyclic aromatic ring systems.
Examples of 5-membered monocyclic heteroaryl groups include but are not limited to furanyl, thienyl, pyrrolyl, oxazolyl, oxadiazolyl (including 1 ,2,3 and 1 ,2,4 oxadiazolyls and furazanyl i.e. 1 ,2,5-oxadiazolyl), thiazolyl, isoxazolyl, isothiazolyl, pyrazolyl, imidazolyl, triazolyl (including 1 ,2,3, 1 ,2,4 and 1 ,3,4 triazolyls), oxatriazolyl, tetrazolyl, thiadiazolyl (including 1 ,2,3 and 1 ,3,4 thiadiazolyls) and the like.
Examples of 6-membered monocyclic heteroaryl groups include but are not limited to pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, pyranyl, oxazinyl, dioxinyl, thiazinyl, thiadiazinyl and the like. Examples of 6-membered aromatic heterocyclyls containing nitrogen include pyridyl (1 nitrogen), pyrazinyl, pyrimidinyl and pyridazinyl (2 nitrogens).
Aromatic heterocyclyl groups may also be bicyclic or polycyclic heteroaromatic ring systems such as fused ring systems (including purine, pteridinyl, napthyridinyl, 1 H thieno[2,3-c]pyrazolyl, thieno[2,3-b]furyl and the like) or linked ring systems (such as oligothiophene, polypyrrole and the like). Fused ring systems may also include aromatic 5- membered or 6-membered heterocyclyls fused to carbocyclic aromatic rings such as phenyl, napthyl, indenyl, azulenyl, fluorenyl, anthracenyl and the like, such as 5-membered aromatic heterocyclyls containing nitrogen fused to phenyl rings, 5-membered aromatic heterocyclyls containing 1 or 2 nitrogens fused to phenyl ring.
A bicyclic heteroaryl group may be, for example, a group selected from: a) a benzene ring fused to a 5- or 6-membered ring containing 1 , 2 or 3 ring heteroatoms; b) a pyridine ring fused to a 5- or 6-membered ring containing 1 , 2 or 3 ring heteroatoms; c) a pyrimidine ring fused to a 5- or 6-membered ring containing 1 or 2 ring heteroatoms; d) a pyrrole ring fused to a 5- or 6-membered ring containing 1 , 2 or 3 ring heteroatoms; e) a pyrazole ring fused to a 5- or 6-membered ring containing 1 or 2 ring heteroatoms; f) an imidazole ring fused to a 5- or 6-membered ring containing 1 or 2 ring heteroatoms; g) an oxazole ring fused to a 5- or 6-membered ring containing 1 or 2 ring heteroatoms; h) an isoxazole ring fused to a 5- or 6-membered ring containing 1 or 2 ring heteroatoms; i) a thiazole ring fused to a 5- or 6-membered ring containing 1 or 2 ring heteroatoms; j) an isothiazole ring fused to a 5- or 6-membered ring containing 1 or 2 ring heteroatoms; k) a thiophene ring fused to a 5- or 6-membered ring containing 1 , 2 or 3 ring heteroatoms; I) a furan ring fused to a 5- or 6- membered ring containing 1 , 2 or 3 ring heteroatoms; m) a cyclohexyl ring fused to a 5- or 6- membered ring containing 1 , 2 or 3 ring heteroatoms; and n) a cyclopentyl ring fused to a 5- or 6-membered ring containing 1 , 2 or 3 ring heteroatoms.
Particular examples of bicyclic heteroaryl groups containing a five membered ring fused to another five membered ring include but are not limited to imidazothiazole (e.g. imidazo[2, 1-b]thiazole) and imidazoimidazole (e.g. imidazo[1 ,2-a]imidazole).
Particular examples of bicyclic heteroaryl groups containing a six membered ring fused to a five membered ring include but are not limited to benzofuran, benzothiophene, benzimidazole, benzoxazole, isobenzoxazole, benzisoxazole, benzothiazole,
benzisothiazole, isobenzofuran, indole, isoindole, indolizine, indoline, isoindoline, purine (e.g., adenine, guanine), indazole, pyrazolopyrimidine (e.g. pyrazolo[1 ,5-a]pyrimidine), benzodioxole and pyrazolopyridine (e.g. pyrazolo[1 ,5-a]pyridine) groups. A further example of a six membered ring fused to a five membered ring is a pyrrolopyridine group such as a pyrrolo[2,3-b]pyridine group.
Particular examples of bicyclic heteroaryl groups containing two fused six membered rings include but are not limited to quinoline, isoquinoline, chroman, thiochroman, chromene, isochromene, isochroman, benzodioxan, quinolizine, benzoxazine, benzodiazine, pyridopyridine, quinoxaline, quinazoline, cinnoline, phthalazine, naphthyridine and pteridine groups. Examples of heteroaryl groups containing an aromatic ring and a non-aromatic ring include tetrahydronaphthalene, tetrahydroisoquinoline, tetrahydroquinoline,
dihydrobenzothiophene, dihydrobenzofuran, 2,3-dihydro- benzo[1 ,4]dioxine,
benzo[1 ,3]dioxole, 4,5,6,7-tetrahydrobenzofuran, indoiine, isoindoline and indane groups.
Examples of aromatic heterocyclyls fused to carbocyclic aromatic rings may therefore include but are not limited to benzothiophenyl, indolyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzimidazolyl, indazolyl, benzoxazolyl, benzisoxazolyl, isobenzoxazoyi, benzothiazolyl, benzisothiazolyl, quinolinyl, isoquinolinyl, quinoxalinyl, quinazolinyl, cinnolinyl, benzotriazinyl, phthalazinyl, carbolinyl and the like.
The term "non-aromatic heterocyclyl" encompasses optionally substituted saturated and unsaturated rings which contain at least one heteroatom selected from the group consisting of N, S and O.
Non-aromatic heterocyclyls may be 3-7 membered mono-cyclic rings.
Examples of 5-membered non-aromatic heterocyclyl rings include 2H-pyrrolyl, 1 - pyrrolinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrrolidinyl, 1 -pyrrolidinyl, 2-pyrrolidinyl, 3-pyrrolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, pyrazolinyl, 2-pyrazolinyl, 3-pyrazolinyl, pyrazolidinyl, 2-pyrazolidinyl, 3-pyrazolidinyl, imidazolidinyl, 3-dioxalanyl, thiazolidinyl, isoxazolidinyl, 2-imidazolinyl and the like.
Examples of 6-membered non-aromatic heterocyclyls include piperidinyl, piperidinonyl, pyranyl, dihyrdopyranyl, tetrahydropyranyl, 2H pyranyl, 4H pyranyl, thianyl, thianyl oxide, thianyl dioxide, piperazinyl, diozanyl, 1 ,4-dioxinyl, 1 ,4-dithianyl, 1 ,3,5- triozalanyl, 1 ,3,5-trithianyl, 1 ,4-morpholinyl, thiomorpholinyl, 1 ,4-oxathianyl, triazinyl, 1 ,4- thiazinyl and the like.
Examples of 7-membered non-aromatic heterocyclyls include azepanyl, oxepanyl, thiepanyl and the like.
Non-aromatic heterocyclyl rings may also be bicyclic heterocyclyl rings such as linked ring systems (for example uridinyl and the like) or fused ring systems. Fused ring systems include non-aromatic 5-membered, 6-membered or 7-membered heterocyclyls fused to carbocyclic aromatic rings such as phenyl, napthyl, indenyl, azulenyl, fluorenyl, anthracenyl and the like. Examples of non-aromatic 5-membered, 6-membered or 7- membered heterocyclyls fused to carbocyclic aromatic rings include indolinyl,
benzodiazepinyl, benzazepinyl, dihydrobenzofuranyl and the like.
The term "halo" refers to fluoro, chloro, bromo or iodo.
Unless otherwise defined, the term "optionally substituted" or "optional substituent" as used herein refers to a group which may or may not be further substituted with 1 , 2, 3, 4 or more groups, preferably 1 , 2 or 3, more preferably 1 or 2 groups selected from the group consisting of Chalky!, C2-6alkenyl, C2-6alkynyl, C3.8cycloalkyl, hydroxyl, oxo, C^alkoxy, aryloxy, C^alkoxyaryl, halo, C^alkylhalo, C^alkoxyhalo, carboxyl, esters, cyano, nitro, am ino, substituted amino, disubstituted amino, acyl, ketones, amides, aminoacyl, substituted am ides, disubstituted amides, thiol, alkylthio, thioxo, sulfates, sulfonates, sulfinyl, substituted sulfinyl, sulfonyl, substituted sulfonyl, sulfonylam ides, substituted sulfonamides, disubstituted sulfonamides, aryl, arCi-ealkyl, heterocyclyl and heteroaryl wherein each alkyl, alkenyl, alkynyl, cycloalkyl, aryl and heterocyclyl and groups containing them may be further optionally substituted. Optional substituents in the case of heterocycles containing N may also include but are not limited to C1.6alkyl i.e. N-C^a\Wy\, more preferably methyl particularly N-methyl.
It will be understood that suitable derivatives of aromatic heterocyclyls containing nitrogen include W-oxides thereof.
The compounds of the invention may also be prepared as salts which are pharmaceutically acceptable, but it will be appreciated that non-pharmaceutically acceptable salts also fall within the scope of the present invention, since these are useful as
intermediates in the preparation of pharmaceutically acceptable salts. Examples of pharmaceutically acceptable salts include salts of pharmaceutically acceptable cations such as sodium , potassium, lithium, calcium, magnesium, ammonium and alkylamimonium; acid addition salts of pharmaceutically acceptable inorganic acids such as hydrochloric, orthophosphoric, sulfuric, phosphoric, nitric, carbonic, boric, sulfamic and hydrobromic acids; or salts of pharmaceutically acceptable organic acids such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumaric, citric, lactic, mucic, gluconic, benzoic, succinic, oxalic, phenylacetic, methanesulfonic, trihalomethanesulfonic, toluenesulfonic,
benzenesulfonic, isethionic, salicylic, sulphanilic, aspartic, glutamic, edetic, stearic, palmitic, oleic, lauric, pantothenic, tannic, ascorbic, valeric and orotic acids. Salts of amine groups may also comprise quaternary ammonium salts in which the amino nitrogen atom carries a suitable organic group such as an alkyl, alkenyl, alkynyl or aralkyl moiety.
The salts may be formed by conventional means, such as by reacting the free base form of the compound with one or more equivalents of the appropriate acid.
It should be understood that a reference to a pharmaceutically acceptable salt includes the solvent addition forms or crystal forms thereof, particularly solvates or polymorphs. Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and may be formed during the process of crystallization with pharmaceutically acceptable solvents such as water, alcohols such as methanol, ethanol or isopropyl alcohol, DMSO, acetonitrile, dimethyl formamide (DMF) and the like with the solvate forming part of the crystal lattice by either non-covalent binding or by occupying a hole in the crystal lattice. Hydrates are formed when the solvent is water, alcoholates are formed when the solvent is alcohol. Solvates of the compounds of the present invention can be conveniently prepared or formed during the processes described herein. In addition, the compounds of the present invention can exist in unsolvated as well as solvated forms. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the compounds and methods provided herein.
Additionally, the compounds of the present invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms of the compounds of the present invention are also considered to be disclosed herein.
It will be understood that compounds of formula (I) may possess a chiral centre and may therefore exist as an isomer such as a racemate or an R- or S- enantiomer. The compounds may therefore be used as a purified enantiomer or diastereomer, or as a mixture of any ratio thereof. The isomers may be separated conventionally by chromatographic methods or using a resolving agent. Alternatively the individual isomers may be prepared by asymmetric synthesis using chiral intermediates. Where the compound has a carbon- carbon double bond, it may occur in Z- or E- form and all isomeric forms of the compounds being included in the present invention.
This invention also encompasses prodrugs of the compounds of formula (I).
The term "pro-drug" is used herein in its broadest sense to include those compounds which are converted in vivo to the compound of formula (I). Use of the prodrug strategy optimises the delivery of the drug to its site of action. Without wishing to be bound by theory the compounds of formula (I) having carboxylic esters may act as prodrug moieties with respect to their corresponding carboxylic acid analogues.
Viral Polymerase Inhibition
The ability of the compounds of formula I to inhibit RNA synthesis by the RNA dependent RNA polymerase of HCV (NS5B) can be demonstrated by any assay capable of measuring RNA dependent RNA polymerase activity. A suitable assay is described in the examples.
While the invention is described with particular reference to compounds having inhibitory activity against a HCV NS5B polymerase, it will be understood that other polymerases can, if desired, be substituted in whole or in part for the HCV polymerase herein described. For example, one microbial polymerase target is HCV NS5B polymerase which is the viral RNA-dependent RNA polymerase (RdRp) that is responsible for viral replications. HCV NS5B protein, is released from a polyprotein and is involved in the synthesis of double-stranded RNA from a single-stranded viral RNA genome. It is believed that the replication and/or reproduction of HCV virus may be inhibited or prevented through the inhibition of NS5B polymerase and suppress or prevent the formation of the double- stranded HCV RNA.
To demonstrate that the compounds of formula (I) act by specific inhibition of NS5B polymerase, the compounds may be tested for the lack of inhibitory activity in an assay measuring the activity of an RNA-dependent RNA polymerase other than HCV polymerase or in a DNA dependent RNA polymerase assay.
Pharmaceutical Compositions
The invention also provides a pharmaceutical composition comprising a compound of formula (I) and a pharmaceutically acceptable carrier.The pharmaceutical composition may further comprise or be administered in combination with one or more other antiviral agents such as Ribavirin (Copegus® or Rebetol®), an antiviral nucleoside inhibitor of NS5B polymerase (such as 4-amino-7-(2-C-methyl- -D-ribofuranosyl)-pyrrolo[2, 1-f][1 ,2,4]triazine; PSI-7977; PSI-938; RG7128 or mericitabine; IDX-184; INX-189 and other such agents that may be developed) a non-nucleoside inhibitor of NS5B polymerase (such as GS-9190 or tegobuvir; PF-868554 or filibuvir; VX-222; IDX-375; ABT-072; ABT-333; ANA-598 or setrobuvir; BI207127; JTK-853; GS-9669; and other such agents that may be developed), a NS3/4A protease inhibitor (such as telaprevir or Incivek®; boceprevir or Victrelis®; Bl- 201335; TMC-435; RG-7227 or danoprevir; MK-7009 or vaniprevir; GS-9451 ; GS-9256; BMS-650032; ACH-1625; ACH-2684; MK-5172; ABT-450; IDX-320; SCH-900518 and other such agents that may be developed), an NS5A inhibitor (such as BMS-790052 (daclatasvir); GS-5885; ABT-267; PPI-461 ; ACH-2928; GSK2336805 and other such agents that may be developed) and/or inhibitor of viral entry, assembly or egress. The composition may also additionally comprise at least one immunomodulatory agent for example an interferon or interferon derivative such as interferon alpha 2B (such as Intron® A interferon available from Schering Corp., Kenilworth, N.J.), pegylated interferon alpha 2A (such as Pegasys® available from Hoffmann-LaRoche, Nutley, N.J.), pegylated interferon alpha 2B (such as Peg-lntron® available from Schering Corp. , Kenilworth, N.J.), consensus interferon (such as interferon alphacon-1 , or Infergen® available from Valeant Pharmaceuticals, Costa Mesa, CA.), interferon alpha 2A, recombinant interferon alpha 2A (such as Roferon® available from Hoffmann-LaRoche, Nutley, N.J.), or lymphoblastoid interferon tau, and/or an inhibitor of inosine-5'-monophosphate dehydrogenase (IMPDH) and other large or small molecules known to modulate host immune responses.
Accordingly, in one embodiment of the pharmaceutical composition, the other antiviral agent is Ribavarin optionally in combination with peg/I FN. In another embodiment, the other antiviral agent is an NS5B inhibitor, more particularly a nucleoside inhibitor such as the bicyclic nucleosides and nucleotides of the general formula described in WO2010/002877, for example, 4-amino-7-(2-C-methyl-3-D- ribofuranosyl)-pyrrolo[2, 1-f][1 ,2,4]triazine.
In yet another embodiment, the other antiviral agent is an NS3/4A protease inhibitor such as telaprevir (VX-950) or Incivek®; boceprevir or Victrelis®; BI-201335; TMC-435; RG- 7227 or danoprevir; MK-7009 or vaniprevir; GS-9451 ; GS-9256; BMS-650032; ACH-1625; ACH-2684; MK-5172; ABT-450; IDX-320; SCH-900518, particularly telaprevir (VX-950).
In still another embodiment, the other antiviral agent is an NS5A inhibitor such as BMS-790052 (daclatasvir); GS-5885; ABT-267; PPI-461 ; ACH-2928; GSK2336805, particularly BMS-790052 (daclatasvir).
It will be understood that combined administration of the compounds of the invention with the other antiviral agent may be concurrent, sequential or separate administration.
The term "composition" is intended to include the formulation of an active ingredient with conventional carriers and excipients, and also with encapsulating materials as the carrier, to give a capsule in which the active ingredient (with or without other carriers) is surrounded by the encapsulation carrier. Any carrier must be "pharmaceutically acceptable" meaning that it is compatible with the other ingredients of the composition and is not deleterious to a subject. The compositions of the present invention may contain other therapeutic agents as described above, and may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavours, etc.) according to techniques such as those well known in the art of pharmaceutical formulation (See, for example, Remington: The Science and Practice of Pharmacy, 21st Ed. , 2005, Lippincott Williams & Wilkins).
The pharmaceutical composition includes those suitable for oral, rectal, nasal, topical (including buccal and sub-lingual), vaginal or parenteral (including intramuscular, sub-cutaneous and intravenous) administration or in a form suitable for administration by inhalation or insufflation.
The compounds of the invention, together with a conventional adjuvant, carrier, or diluent, may thus be placed into the form of pharmaceutical compositions and unit dosages thereof, and in such form may be employed as solids, such as tablets or filled capsules, or liquids such as solutions, suspensions, emulsions, elixirs, or capsules filled with the same, all for oral use, in the form of suppositories for rectal administration ; or in the form of sterile injectable solutions for parenteral (including subcutaneous) use. Such pharmaceutical compositions and unit dosage forms thereof may comprise conventional ingredients in conventional proportions, with or without additional active compounds or principles, and such unit dosage forms may contain any suitable effective amount of the active ingredient commensurate with the intended daily dosage range to be employed.
For preparing pharmaceutical compositions from the compounds of the present invention, pharmaceutically acceptable carriers can be either solid or liquid. Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispensable granules. A solid carrier can be one or more substances which may also act as diluents, flavouring agents, solubilisers, lubricants, suspending agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material.
Suitable carriers are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium
carboxymethylcellulose, a low melting wax, cocoa butter, and the like. The term
"preparation" is intended to include the formulation of the active compound with
encapsulating material as carrier providing a capsule in which the active component, with or without carriers, is surrounded by a carrier, which is thus in association with it. Similarly, cachets and lozenges are included. Tablets, powders, capsules, pills, cachets, and lozenges can be used as solid forms suitable for oral administration.
Liquid form preparations include solutions, suspensions, and emulsions, for example, water or water-propylene glycol solutions. For example, parenteral injection liquid preparations can be formulated as solutions in aqueous polyethylene glycol solution.
Sterile liquid form compositions include sterile solutions, suspensions, emulsions, syrups and elixirs. The active ingredient can be dissolved or suspended in a
pharmaceutically acceptable carrier, such as sterile water, sterile organic solvent or a mixture of both.
The compositions according to the present invention may thus be formulated for parenteral administration (e. g. by injection, for example bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi-dose containers with an added preservative. The compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulation agents such as suspending, stabilising and/or dispersing agents. Alternatively, the active ingredient may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilisation from solution, for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use. Pharmaceutical forms suitable for injectable use include sterile injectable solutions or dispersions, and sterile powders for the extemporaneous preparation of sterile injectable solutions. They should be stable under the conditions of manufacture and storage and may be preserved against oxidation and the contaminating action of microorganisms such as bacteria or fungi.
The solvent or dispersion medium for the injectable solution or dispersion may contain any of the conventional solvent or carrier systems for the compounds, and may contain, for example, water, ethanol, polyol (for example, glycerol, propylene glycol and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
Pharmaceutical forms suitable for injectable use may be delivered by any appropriate route including intravenous, intramuscular, intracerebral, intrathecal, epidural injection or infusion.
Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various other ingredients such as these enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilised active ingredient into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, preferred methods of preparation are vacuum drying or freeze-drying of a previously sterile-filtered solution of the active ingredient plus any additional desired ingredients.
When the active ingredients are suitably protected they may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsule, or it may be compressed into tablets, or it may be incorporated directly with the food of the diet. For oral therapeutic administration, the active compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
The amount of active compound in therapeutically useful compositions should be sufficient that a suitable dosage will be obtained.
The tablets, troches, pills, capsules and the like may also contain the components as listed hereafter: a binder such as gum, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such a sucrose, lactose or saccharin may be added or a flavouring agent such as peppermint, oil of wintergreen, or cherry flavouring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules may be coated with shellac, sugar or both. A syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavouring such as cherry or orange flavour. Of course, any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed. In addition, the active compound (s) may be incorporated into sustained-release preparations and formulations, including those that allow specific delivery of the active peptide to specific regions of the gut.
Aqueous solutions suitable for oral use can be prepared by dissolving the active component in water and adding suitable colorants, flavours, stabilising and thickening agents, as desired. Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, or other well known suspending agents.
Pharmaceutically acceptable carriers and/or diluents include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like.
Also included are solid form preparations that are intended to be converted, shortly before use, to liquid form preparations for oral administration. Such liquid forms include solutions, suspensions, and emulsions. These preparations may contain, in addition to the active component, colorants, flavours, stabilisers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilising agents, and the like.
For topical administration to the epidermis the compounds according to the invention may be formulated as ointments, creams or lotions, or as a transdermal patch. Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents. Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilising agents, dispersing agents, suspending agents, thickening agents, or colouring agents.
Formulations suitable for topical administration in the mouth include lozenges comprising active agent in a flavoured base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier. Solutions or suspensions are applied directly to the nasal cavity by conventional means, for example with a dropper, pipette or spray. The formulations may be provided in single or multidose form. In the latter case of a dropper or pipette, this may be achieved by the patient administering an appropriate, predetermined volume of the solution or suspension.
In the case of a spray, this may be achieved for example by means of a metering atomising spray pump. To improve nasal delivery and retention the compounds according to the invention may be encapsulated with cyclodextrins, or formulated with other agents expected to enhance delivery and retention in the nasal mucosa.
Administration to the respiratory tract may also be achieved by means of an aerosol formulation in which the active ingredient is provided in a pressurised pack with a suitable propellant such as a chlorofluorocarbon (CFC) for example dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane, carbon dioxide, or other suitable gas.
The aerosol may conveniently also contain a surfactant such as lecithin. The dose of drug may be controlled by provision of a metered valve.
Alternatively the active ingredients may be provided in the form of a dry powder, for example a powder mix of the compound in a suitable powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP). Conveniently the powder carrier will form a gel in the nasal cavity. The powder composition may be presented in unit dose form for example in capsules or cartridges of, e. g. gelatin, or blister packs from which the powder may be administered by means of an inhaler.
In formulations intended for administration to the respiratory tract, including intranasal formulations, the compound will generally have a small particle size for example of the order of 5 to 10 microns or less. Such a particle size may be obtained by means known in the art, for example by micronisation.
When desired, formulations adapted to give sustained release of the active ingredient may be employed.
The pharmaceutical preparations are preferably in unit dosage forms. In such form, the preparation is subdivided into unit doses containing appropriate quantities of the active component. The unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules. Also, the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms are dictated by and directly dependent on (a) the unique characteristics of the active material and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active material for the treatment of a HCV viral infection in living subjects having a diseased condition in which bodily health is impaired.
The invention also includes the compounds in the absence of carrier where the compounds are in unit dosage form.
Compositions comprising compounds of the invention formulated for oral delivery either alone or in combination with another HCV antiviral agent are particularly preferred. Methods of treatment
The compounds of formula (I) may be used in the treatment of a Flaviviridae viral infection such as a HCV infection.
Generally, the term "treatment" means affecting a subject, tissue or cell to obtain a desired pharmacological and/or physiological effect and includes: (a) inhibiting the viral infection, i.e. arresting its development or further development; (b) relieving or ameliorating the effects of the viral infection, i.e. cause regression of the effects of the viral infection; (c) reducing the incidence or the viral infection or (d) preventing the infection from occurring in a subject, tissue or cell predisposed to the viral infection disease or at risk thereof, but has not yet been diagnosed with a protective pharmacological and/or physiological effect so that the viral infection does not develop or occur in the subject, tissue or cell.
The prevention of hepatitis C means, for example, administration of a
pharmaceutical agent to a subject found to carry a HCV by a test and the like but without a symptom of infection, or to a subject who shows an improved disease state of hepatitis after a treatment of hepatitis C, but who still carries a HCV and is associated with a risk of recurrence of hepatitis.
The term "subject" as used herein refers to any animal, in particular mammals such as humans having a disease or condition which requires treatment with the compound of formula (I).
The term "administering" refers to providing the compound or pharmaceutical composition of the invention to a subject suffering from or at risk of the diseases or conditions to be treated or prevented.
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-PC , 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 "Flaviviridae virus" refers to a virus of the family Flaviviridae, which family includes the Hepacivirus Flavivirus and Pestivirus or hepatitis C-like virus genera. A representative species of the genus of hepatitis C-like viruses is hepatitis C virus.
Dosages
The term "therapeutically effective amount" refers to the amount of the compound of formula (I) that will elicit the biological or medical response of a subject, tissue or cell that is being sought by the researcher, veterinarian, medical doctor or other clinician.
In the prevention or treatment of HCV infections or diseases an appropriate dosage level will generally be about 0.01 to 500 mg per kg subject body weight per day which can be administered in single or multiple doses. Preferably, the dosage level will be about 0.1 to about 250 mg/kg per day; more preferably about 0.5 to about 100 mg/kg per day. A suitable dosage level may be about 0.01 to 250 mg/kg per day, about 0.05 to 100 mg/kg per day, or about 0.1 to 50 mg/kg per day. Within this range the dosage may be 0.05 to 0.5, 0.5 to 5 or 5 to 50 mg/kg per day. The dosage may be selected, for example to any dose within any of these ranges, for therapeutic efficacy and/or symptomatic adjustment of the dosage to the subject to be treated
It will be understood that the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the subject undergoing therapy.
It will further be understood that when the compounds of the invention are to be administered in combination with one or more HCV antiviral agents the dosage forms and levels may be formulated for either concurrent, sequential or separate administration or a combination thereof.
Methods of preparation
It will be understood that unless otherwise defined each moiety having a substitutable hydrogen such as for example, alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, cycloalkenyl, heterocyclyl, aryl and heteroaryl, in each occurrence as described in the general schemes and methods which follow may be optionally substituted. It will also be understood that the particular examples which are described herein may undergo further functionalisation using methods known in the art, for example, compounds comprising amino groups or acid groups may undergo reduction amination or amide couplings respectively to form further examples of compounds of the invention.
Compounds of formula (I) may be generally synthesised via a common synthetic intermediate of general formula (II):
Figure imgf000030_0001
wherein ΧΊ , X'2 X'3, Ri , R2, R3, Zi , Z2, Z3, Z4 and Z5 are as defined above.
Generally, compounds of formula (II) are prepared by heating a suitable amino- substituted heterocyclyl with a suitable alpha-halo ketone in the presence of a solvent, such as DMF, and a suitable base, such as potassium carbonate.
Accordingly, in one embodiment there is provided a process for preparing a compound of formula (I) via an intermediate of general formula (II).
It will be recognised by those skilled in the art that intermediates of formula (II) containing carboxylic ester groups, i.e. C02C1.6alkyl (such as C02CH3, C02CH2CH3 and C02CH2CH2CH3) may be converted to their corresponding carboxylic acids using various conditions (e.g. treatment with aq. NaOH in methanol/THF) which may then be coupled with any suitable primary or secondary amides using a variety of standard peptide coupling techniques to make the corresponding amides (e.g. the use of a peptide coupling agent such as HATU, HBTU, EDCI etc. with the appropriate solvent and base). It will be understood that potential reactive groups under amide coupling conditions may be suitably protected during the course of the reaction and subsequently deprotected. Compounds of formula (I I) containing carboxylic esters and acid groups may therefore be used to access a diversity of compounds of formula (I) with side chains via amide coupling. Side chains comprising a carboxylic acid or ester moiety may also undergo further amide coupling thereby providing multistep access to a diversity of compounds with extended side chains.
Accordingly, in one embodiment there is provided a process for producing a compound of formula (I) when B is -CONH-R7- comprising the step of reacting a compound of formula (II) with an amino precursor of general formula NH2R7 under amide coupling conditions. Suitable amide coupling conditions will be understood by those skilled in the art and have been described in many references such as Advanced Organic Chemistry - 4th Edition, March J., John Wiley & Sons Inc, New York, 1992 (see also 5th Edition, John Wiley & Sons, New York 2001 and 6th Edition, John Wiley & Sons, New Jersey, 2007).
In some instances, the compounds of formula (II) have also demonstrated antiviral activity. Accordingly, compounds of formula (II) will be understood to be process intermediates and/or final compounds as the case may be.
EXAMPLES
The invention will now be described with reference to the following non-limiting examples.
Synthetic Methods
1 H NMR spectra were recorded on either a Bruker Avance DRX 400, AC 200 or AM 300 spectrometer. Spectra were recorded in CDCI3, acetone-c/6, CD3OD (MeOD) or DMSO-ck using the residual solvent peak as a reference. Chemical shifts are reported on the δ scale in parts per million (ppm) using the following conventions to assign the multiplicity: s (singlet), d (doublet), t (triplet), q (quartet), qt (quintet), m (multiplet) and prefixed br (broad). Mass spectra (ESI) were recorded on either a Micromass Platform QMS or Thermo Finnigan LCQ Advantage spectrometer. Flash chromatography was performed on 40-63pm silica gel 60 (Merck No. 9385). Automated flash chromatography was performed either on a Combi- Flash™ purification system using Combi-Flash™ silica gel columns or on a Biotage SP4 purification system using either GraceResolv™ silica gel cartridges, Grace Reveleris™ C-18 reverse phase silica gel cartridges or Biotage SNAP™ C-18 reverse phase silica gel cartridges. Preparative HPLC was carried out using either a Gilson 322 pump with a Gilson 215 liquid handler and a HP1 100 PDA detector or an Agilent 1200 Series mass detected preparative LCMS using a Varian XRs C-18 100 x 21 .2 mm column. Unless otherwise specified, the HPLC systems employed Phenomenex C8(2) columns using either acetonitrile or acetonitrile containing 0.06% TFA in water or water containing 0.1 % TFA.
During the reactions a number of the moieties may need to be protected. Suitable protecting groups are well known in industry and have been described in many references such as "Greene's Protective Groups in Organic Synthesis" Fourth Edition, Peter G.M. Wuts and Theodora W. Greene, Wiley-lnterscience, 2007 .
The abbreviations used in the Examples are as follows unless indicated otherwise:
BSA: Bovine serum albumin
DCM: Dichloromethane
DIPEA: N,N-Diisopropylethylamine
DMEM: Dulbecco's modified eagle medium
DMF: dimethylformamide
DMSO: dimethylsulfoxide DTT: DL-dithiothreitol
EDCI: A/-(3-Dimethylaminopropyl)-/V'-ethylcarbodiimide hydrochloride
EDTA: Ethylenediaminetetraacetic acid
ESI: electrospray ionisation
GTP: Guanosine triphosphate
HATU: 2-(7-Aza-1 H-benzotriazole-1 -yl)-1 , 1 ,3,3-tetramethyluronium hexafluorophosphate HBTU: 0-(Benzotriazol-1 -yl)-/V,/V,/V',A/-tetramethyluronium hexafluorophosphate
HPLC: high performance liquid chromatography
LCMS: liquid chromatography coupled mass spectrometry
LHMDS: Lithium hexamethyldisilazide
MS: mass spectrometry
NM : nuclear magnetic resonance
RBF: round bottomed flask
RT: room temperature
THF: tetrahydrofuran
TLC: thin-layer chromatography
Example syntheses of compounds of formulae (I) and (II) where A is a covalent bond
Synthesis of methyl 3-cyclohexyl-2-(4-fluorophenyl)-1H-imidazo[ 1, 2-b]pyrazole-6- carboxylate (i) and 3-cyclohexyl-2-(4-fluoroDhenyl)-1H-imidazo[1,2-blDyrazole-6-carboxylic acid (ii)
Figure imgf000032_0001
0) (ii)
Step a: To a solution of 1-(4-fluorophenyl)ethanone (5.00 g; 36.20 mmol) in THF (80 mL) at - 78°C was added a 1 M THF solution of LHMDS in THF (40 mmol). After 30 minutes the cyclohexanone (3.90 g; 40 mmol) in THF (80mL) was added and the reaction mixture was stirred at that temperature for 30 minutes and then quenched upon addition of an aqueous solution of sodium bircarbonate. Ethyl acetate (200 mL) was added and the organic layer was washed with water (2 x 100 mL) then brine (100 mL). The organic layer was dried (Na2S04) and concentrated to leave a pale brown oil (9.55 g). The crude reaction material was purified by automated flash column chromatography (Biotage-SP4) on silica (40g) eluting with EtOAc/Hexane to afford 1 -(4-fluorophenyl)-2-(1 -hydroxycyclohexyl)ethanone (5.60 g) as a colourless oil. Step b: To a solution of 1-(4-fluorophenyl)-2-(1 -hydroxycyclohexyl)ethanone (350 mg; 1 .48 mmol) in DCM (4 mL) was added triethylsilane (450 mg; 3.90 mmol) followed by TFA (2 mL) and the reaction mixture was stirred for 16 hours at ambient temperature. The reaction mixture was partitioned between EtOAc (75 mL) and water (100 mL). The aqueous layer was extracted with EtOAc (3 x 60 mL) and the combined organics were washed with brine (75 mL), dried (MgS04) and concentrated to leave a brown oil (4100 mg). The crude reaction material was purified by automated flash column chromatography (Biotage-SP4) on silica (80g) eluting with EtOAc and hexane to leave 2-cyclohexyl-1 -(4-fluorophenyl)ethanone (2.20 g) as a colourless oil.
Step c: To a solution of 2-cyclohexyl-1 -(4-fluorophenyl)ethanone (2.50 g; 1 1 .35 mmol) in dioxane/diethyl ether (1 : 1 ; 100 mL) was added bromine (0.64 mL; 12 mmol) and the reaction mixture was stirred at ambient temperature. After 2 hours the reaction mixture was partitioned between EtOAc (125ml) and water (125 ml) and the aqueous layer was separated and extracted with EtOAc (3 x 30 ml). The combined organics were washed with brine (35 mL). dried (MgS04) and concentrated to leave 2-bromo-2-cyclohexyl-1 -(4- fluorophenyl)ethanone (2.79 g) as a colourless oil.
Step d: To a solution of methyl 5-amino-1 H-pyrazole-3-carboxylate (75 mg; 0.53 mmol) in DMF (2 mL) was added the 2-bromo-2-cyclohexyl-1-(4-fluorophenyl)ethanone ( 170 mg; 0.58 mmol) followed by potassium carbonate (263 mg; 2.66 mmol) and the reaction mixture was stirred at 75°C. After 2 hours water (25 mL) was added and the aqueous layer was extracted with EtOAc (3 x 30 mL). The combined organics were washed with brine (25 mL), dried (MgS04) and concentrated to leave a brown oil (89 mg). The residue was redissolved in EtOH ( 2 mL) and 2M sulphuric acid (0.5 mL) was added and the reaction mixture was stirred at 70 °C. After 2 hours NaHC03 (25 ml) was added and the aqueous layer was extracted with EtOAc (3 x 30 mL). The combined organics were washed with brine (25 mL), dried (MgS04) and concentrated to leave a brown oil (75 mg). Purification by reverse phase LCMS: Acetonitrile/water (0.1 % formic acid) afforded methyl 3-cyclohexyl-2-(4-fluorophenyl)- 1 H-imidazo[1 ,2-b]pyrazole-6-carboxylate (i) (8.5 mg) as an amorphous white solid. ESI-MS m/z calculated for [M+H]+: 342.16; found: 342.09.
Step e: To a solution of methyl 3-cyclohexyl-2-(4-fluorophenyl)-1 H-imidazo[1 ,2-b]pyrazole-6- carboxylate (i) (3 mg) in MeOH/THF (1 : 1 ; 1 mL) was added a 1 M aqueous solution of lithium hydroxide (0.09 mmol) and the reaction was heated at 60°C for 3 hrs. A 1 M aqueous solution of citric acid (25 ml) was added and the aqueous layer was extracted with EtOAc (3 x 30 mL). The combined organics were washed with brine (25 mL), dried (MgS04) and concentrated to leave a brown oil (4 mg). Purification by reverse phase LCMS:
Acetonitrile/water (0.1 % formic acid) afforded 3-cyclohexyl-2-(4-fluorophenyl)-1 H- imidazo[1 ,2-b]pyrazole-6-carboxylic acid (ii) (0.81 mg) as an amorphous white solid. ESI-MS m/z calculated for [M+H]+: 328.15; found: 328.08. 1H NM (400 MHz, CD3CN) δ 9.22 (s, 1 H), 7.59 - 7.50 (m, 2H), 7.30 - 7.21 (m, 2H), 6.22 (s, 1 H), 3.00 - 2.89 (m, 1 H), 2.01 - 1 .97 (m, 2H), 1 .88 - 1.61 (m, 5H), 1.42 - 1 .32 (m, 3H).
Synthesis of methyl 3-cvclohexyl-2-(4-fluoroDhenyl)-1-methyl-imidazo[ 1, 2-bl yrazole-6- carboxylate (iii) and 3-cvclohexyl-2-(4-fluoroDhenyl)-1-methyl-1H-imidazo[1,2-bloyrazole-6- carboxylic aci
Figure imgf000034_0001
II IV
Step a: To a solution of methyl 3-cyclohexyl-2-(4-fluorophenyl)-1 H-imidazo[1 ,2-b]pyrazole-6- carboxylate (i) (10 mg; 0.03 mmol) in DMF (1 mL) was added iodomethane (0.06 mmol) followed by potassium carbonate (29 mg; 0.29 mmol). After 16 hours H20 (25 ml) was added and the aqueous layer was extracted with EtOAc (3 x 30 mL). The combined organics were washed with brine (25 mL), dried (MgS04) and concentrated to leave a brown oil (12 mg). Purification by reverse phase LCMS: Acetonitrile/water (0.1 % formic acid) afforded methyl 3-cyclohexyl-2-(4-fluorophenyl)-1-methyl-imidazo[1 ,2-b]pyrazole-6-carboxylate (iii) (5.6 mg) as an amorphous white solid. ESI-MS m/z calculated for [M+H]+: 356.18; found: 356.09.
1 H NMR (400 MHz, CD3CN) δ 7.51 - 7.43 (m, 2H), 7.34 - 7.24 (m, 2H), 6.28 (s, 1 H), 3.86 (s, 3H), 3.37 (s, 3H), 2.71 (tt, J = 12.2, 3.4 Hz, 1 H), 2.10 - 1 .98 (m, 2H), 1.84 - 1 .64 (m, 5H), 1 .36 - 1 .24 (m, 3H).
Step b: To a solution of methyl 3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2- b]pyrazole-6-carboxylate (iii) (5 mg; 0.01 mmol) in MeOH/THF (1 : 1 ; 2 mL) was added a 1 M aqueous solution of lithium hydroxide (0.1 mmol). The reaction was heated at 60°C for 2 hrs. 1 M HCI (aq) was added until ~pH 3 and the aqueous layer was extracted with EtOAc (3 x 30 mL) and the combined organics were washed with brine (25 mL), dried (MgS04) and concentrated to leave a brown oil (4 mg). Purification by reverse phase LCMS:
Acetonitrile/water (0.1 % formic acid) afforded 3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H- imidazo[1 ,2-b]pyrazole-6-carboxylic acid (iv) (2.88 mg) as an amorphous white solid ESI-MS m/z calculated for [M+H]+: 342.16; found: 342.07. 1H NMR (400 MHz, CD3CN) δ 7.49 - 7.45 (m, 2H), 7.32 - 7.24 (m, 2H), 6.27 (s, 1 H), 3.37 (s, 3H), 2.76 - 2.65 (m, 1 H), 2.13 - 2.07 (m, 2H), 1.83 - 1.66 (m, 5H), 1.29 (s, 3H). Synthesis of ethyl 3-cvclohexyl-2-(4-fluoroDhenyl)-1H-imidazo[ 1, 2-bl yrazole-7-carboxylate (v); ethyl 3-cvclohexyl-2-(4-fluoroDhenyl)-1-methyl-imidazo[ 1, 2-bloyrazole-l-carboxylate (vi); 3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-blpyrazole-7-carboxylic acid (vii); and 3-cvclohex l-2-(4-fluoroohenyl)-1 -methyl-1 H-imidazof 1, 2-blpyrazole (viii)
Figure imgf000035_0001
(vii) (viii)
Step a: To a solution of ethyl 5-amino-1 H-pyrazole-4-carboxylate (30 mg; 0.19 mmol) in DMF (2 ml_) was added 2-bromo-2-cyclohexyl-1-(4-fluorophenyl)ethanone (64 mg; 0.21 mmol) followed by potassium carbonate (96 mg; 0.97 mmol) and the reaction was stirred at ambient temperature for 4 hours. H20 (25 ml) was added and the aqueous layer was extracted with EtOAc (3 x 30 ml_) and the combined organics were washed with brine (25 ml_), dried (MgS04) and concentrated to leave a brown oil (85 mg). The residue was redissolved in EtOH (2 imL) and 2M sulphuric acid (1 mL) was added and the reaction mixture was stirred at 65°C. After 2 hours NaHC03 (25 ml) was added and the aqueous layer was extracted with EtOAc (3 x 30 mL) and the combined organics were washed with brine (25 mL), dried (MgS04) and concentrated to leave ethyl 3-cyclohexyl-2-(4- fluorophenyl)-1 H-imidazo[1 ,2-b]pyrazole-7-carboxylate (v) as a brown oil (65 mg). The crude reaction material was used directly without purification. ESI-MS m/z calculated for [M+H]+: 356.18; found: 356.12.
Step b: To a solution of ethyl 3-cyclohexyl-2-(4-fluorophenyl)-1 H-imidazo[1 ,2-b]pyrazole-7- carboxylate (v) (75 mg; 0.21 mmol) in DMF (5 mL) was added iodomethane (60 mg; 0.42 mmol) followed by potassium carbonate (105 mg; 1 .06 mmol). After 16 hours H20 (25 ml) was added and the aqueous layer was extracted with EtOAc (3 x 30 mL). The combined organics were washed with brine (25 mL), dried (MgS04) and concentrated to leave a brown oil (65 mg). Purification by reverse phase LCMS: Acetonitrile/water (0.1 % formic acid) afforded ethyl 3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-7-carboxylate (vi) (15 mg) as an amorphous white solid. ESI-MS m/z calculated for [M+H]+: 370.19; found: 370.21 .
Step c: To a solution of ethyl 3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2- b]pyrazole-7-carboxylate (vi) (20 mg; 0.05 mmol) in MeOH/THF (1 :1 ; 2 mL) was added a 1 M aqueous solution of lithium hydroxide (1 .08 mmol). The reaction was heated at 60 °C for 6 hrs. 1 M HCI was added until ~pH 3 and the aqueous layer was extracted with EtOAc (3 x 30 mL). The combined organics were washed with brine (25 mL), dried (MgS04) and concentrated to leave a brown oil (15 mg). Purification by reverse phase LCMS:
Acetonitrile/water (0.1 % formic acid) afforded 3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H- imidazo[1 ,2-b]pyrazole-7-carboxylic acid (vii) (1.23 mg) as an amorphous white solid. ESI- MS m/z calculated for [M+H]+: 342.16; found: 342.1 1. 1H NMR (400 MHz, CD3CN) δ 7.94 (s, 1 H), 7.50 - 7.43 (m, 2H), 7.30 - 7.24 (m, 2H), 3.72 (s, 3H), 2.71 - 2.61 (m, 1 H), 2.07 - 1 .98 (m, 2H), 1 .79 - 1.64 (m, 5H), 1.26 (t, J = 9.3 Hz, 3H) and 3-cyclohexyl-2-(4-fluorophenyl)-1 - methyl-1 H-imidazo[1 ,2-b]pyrazole (viii) (0.5 mg) as an amorphous white solid. ESI-MS m/z calculated for [M+H]+: 298.17; found: 298.23. 1 H NMR (400 MHz, CD3CN) δ 7.52 (d, J = 2.2 Hz, 1 H), 7.48 - 7.43 (m, 2H), 7.29 - 7.23 (m, 2H), 5.66 (d, J = 2.2 Hz, 1 H), 3.34 (s, 3H), 2.67 (tt, J = 12.3, 3.5 Hz, 1 H), 2.12 - 2.08 (m, 2H), 1 .81 - 1.65 (m, 5H), 1 .28 (s, 3H).
Synthesis of ethyl 3-cyclohexyl-1 -methyl-2-phenyl-1 H-imidazo[1 ,2-blpyrazole-7-carboxylate (ix) and 3-cvclohexyl-1-methyl-2-Dhenyl-1H-imidazo[1,2-blDyrazole-7-carboxylic acid (x)
Figure imgf000036_0001
Step a: A 1 M solution of zinc chloride in diethyl ether (39.5 mL) was added to 140 mL anhydrous ether under an atmosphere of argon at 0°C. To this was added
cyclohexylmagnesium chloride (2M solution in ether, 19.5 mL). The resulting suspension was stirred vigorously at 0°C for 20 minutes. To this reaction mixture was added a suspension of 2-chloroacetophenone (4000 mg) and copper acetylacetonate (320 mg) in anhydrous THF (60 mL) at rt via a syringe. The reaction mixture was stirred at rt for 19.5 hrs. Work-up: the reaction mixture was diluted with ethyl acetate (800 mL) and washed with satd ammonium chloride solution. The aqueous solution was back-extracted with EtOAc (2x 100 mL), the combined organic phases were dried (Na2S04/MgS04), filtered and evaporated to dryness. Purification of the residue on normal phase silica by gradient elution (1-50% EtOAc in hexane) afforded 2-cyclohexyl-1-phenylethanone (3470 mg, 66% yield). ESI-MS m/z calculated for [M+H]+: 203.15; found: 203.06. 1 H NMR (400 MHz, CDCI3) δ 7.97 - 7.93 (m, 2H), 7.58 - 7.52 (m, 1 H), 7.48 - 7.43 (m, 2H), 2.82 (d, J = 6.8 Hz, 2H), 2.04 - 1.91 (m, 1 H), 1 .81 - 1 .61 (m, 5H), 1.35 - 1.23 (m, 2H), 1 .22 - 1.09 (m, 1 H), 1.08 - 0.96 (m, 2H).
Step b: To a solution of 2-cyclohexyl-1-phenylethanone (3470 mg) in dioxane/diethyl ether (1 : 1 , 20 mL) was added at rt bromine (930 μΙ_) in portions over 40 min (progress monitored by 1H NMR), and the brown mixture was stirred at ambient temperature for another 30 min. Work-up: the reaction mixture was partitioned between EtOAc (100 ml) and NaHC03 (100 ml). The aqueous layer was separated and extracted with EtOAc (100 ml). The combined organic layers were washed with brine (100 mL), dried (Na2S04/MgS04) and concentrated to afford 2-bromo-2-cyclohexyl-1 -phenylethanone (4770 mg, 99% yield) as a pale brown oil. 1 H NMR (400 MHz, CDCI3) δ 8.02 - 7.98 (m, 2H), 7.62 - 7.57 (m, 1 H), 7.51 - 7.46 (m, 2H), 4.94 (d, J = 9.3 Hz, 1 H), 2.27 - 2.1 1 (m, 2H), 1 .87 - 1.78 (m, 1 H), 1 .75 - 1.63 (m, 3H), 1.40 - 0.93 (m, 5H).
Step c: To a solution of ethyl 5-amino-1 /-/-pyrazole-4-carboxylate (120 mg) in anhydrous DMF (5 mL) was added under an atmosphere of argon 2-bromo-2-cyclohexyl-1 - phenylethanone (185 mg) followed by potassium carbonate (320 mg), and the reaction mixture was stirred at 100 °C for 2 hrs and at rt for 17 hrs. After this period iodomethane (50 pL) was added and the reaction mixture was stirred at rt for 20 min. Water (50 mL) was added and the aqueous layer was extracted with EtOAc (2x50 mL), and the combined organic extracts were washed with brine (20 mL), dried (MgS04), filtered and evaporated to dryness. Purification of the residue on normal phase silica by gradient elution (20-50% EtOAc in hexane) afforded ethyl 3-cyclohexyl-1 -methyl-2-phenyl-1 H-imidazo[1 ,2-b]pyrazole- 7-carboxylate (ix) as an oil (75 mg, 32% yield). ESI-MS m/z calculated for [M+H]+: 352.20; found: 352.19. 1 H NMR (400 MHz, CDCI3) δ 8.04 (s, 1 H), 7.54 - 7.47 (m, 3H), 7.36 - 7.33 (m, 2H), 4.29 (q, J = 7.1 Hz, 2H), 3.78 (s, 3H), 2.72 (tt, J = 12.2, 3.3 Hz, 1 H), 2.09 - 1.96 (m, 2H), 1.81 - 1.63 (m, 5H), 1.36 (t, J = 7.1 Hz, 3H), 1 .34 - 1 .19 (m, 3H).
Step d: A solution of ethyl 3-cyclohexyl-1 -methyl-2-phenyl-1 H-imidazo[1 ,2-b]pyrazole-7- carboxylate (ix) (60 mg) in 1 M aquous KOH/1 ,4-dioxane (6.4 mL, 1 : 1 ) was heated at reflux for 2 hrs. The reaction mixture was cooled to 0°C, and adjusted to pH 7 with 1 M HCI and satd NaHC03 solution, extracted with EtOAc (2x 20 mL), the combined organic layers were washed with brine (10 mL), dried (MgS04), filtered and evaporated to drynes to afford an oil that solidified on standing. Purification of this material on normal phase silica by gradient elution (33-100% EtOAc in hexane) afforded 3-cyclohexyl-1-methyl-2-phenyl-1 H- imidazo[1 ,2-b]pyrazole-7-carboxylic acid (x) as a white solid (31 mg, 56% yield). ESI-MS m/z calculated for [M+H]+: 324.16; found: 324.16. 1H-NMR (400 MHz, CDCI3) δ 8.12 (s, 1 H), 7.53-7.50 (m, 3H), 7.36-7.34 (m, 2H), 3.78 (s, 3H), 2.72 (tt, J 3.1 Hz, J 12.3 Hz, 1 H), 2.08- 1 .98 (m, 2H), 1.80-1 .65 (m, 5H), 1 .37-1.21 (m, 3H).
Synthesis of 13-cyclohexyl-6, 7-dihvdropyrazolo[5', 1 ':2, 31imidazof 1, 5-dtf 1, 4]benzoxazepine- 10-carboxylic acid (xii)
Figure imgf000038_0001
(xi) (xii)
Lit. (NM data): K.-S. Huang, S.-R. Li, Y.-F. Wang, Y.-L. Lin, Y.-H. Chen, T.-W. Tsai, C.-H. Yang, E.-C. Wang, J. Chin. Chem. Soc. 2005, 52, 159-167.
Step a: To a mixture of 63 mL water, 7 mL 7 N NaOH and 60 mL 1 ,2-dichloroethane was added 1 -(2-hydroxyphenyl)ethanone, 1 100 mg tetrabutylammonium bromide and 1 -bromo-2- chloroethane (4.5 mL), and the reaction mixture was stirred vigorously at rt for 96 hrs, then heated at reflux for 16 hrs. The reaction mixture was cooled to rt, the organic layer was separated, the aqueous layer was extracted with DCM (20 mL), the combined organic layers were washed with 1 M HCI (20 mL), then brine to neutral pH, dried (Na2S04/MgS04), filtered and concentrated. Purification of the residue on silica gel by gradient elution (10-50% EtOAc in hexane) afforded 3800 mg (52% yield) 1-[2-(2-chloroethoxy)phenyl]ethanone as a white solid. ESI-MS m/z calculated for [M+H]+: 199.05; found: 199.02.
Step b: To a solution of 1-[2-(2-chloroethoxy)phenyl]ethanone (4200 mg) in THF (90 mL) at - 78 C was added under an atmosphere of argon a 1 M THF solution of LiHMDS (25 mL). After 30 minutes cyclohexanone (2520 μί) in THF (10 mL) was added and the reaction mixture was stirred at -78 °C for 1 .5 hrs. The reaction mixture was quenched into saturated ammonium chloride solution (300 mL), extracted with EtOAc (2x100 mL), the combined organic layer were washed with brine (1 x100 mL), and dried (MgS04), filtered and evaporated to dryness to leave a solid: 6270 mg (100%). Purification on silica gel by gradient elution (5-50% EtOAc in hexane) afforded 1 -[2-(2-chloroethoxy)phenyl]-2-(1- hydroxycyclohexyl)ethanone (4910 mg, 78% yield) as a colourless solid. ESI-MS m/z calculated for [M-H20+H]+: 279.12; found: 278.89. 1 H-NMR (400 MHz, CDCI3) δ 7.86 (dd, J 1 .8 Hz, J 7.8 Hz, 1 H), 7.46 (ddd, J 1 .8 Hz, J 7.4 Hz, , J 8.3 Hz 1 H), 7.05 (dt, J 0.9 Hz, J 7.6 Hz 1 H), 6.90 (br d, J 8.3 Hz, 1 H), 4.35-4.33 (m, 2H), 4.03 (br s, 1 H), 3.90-3.88 (m, 2H), 3.26 (s, 2H), 1.76-1.66 (m, 4H), 1 .59-1 .52 (m, 1 H), 1.49-1 .40 (m, 4H), 1 .32-1.23 (m, 1 H).
Step c: To a solution of 1-[2-(2-chloroethoxy)phenyl]-2-(1 -hydroxycyclohexyl)ethanone (4910 mg) in anhydrous dichloromethane (100 mL, argon) was added under an atmosphere of argon triethylsilane (27 mL) followed by trifluoroacetic acid (1.5 mL) and the reaction mixture was stirred at ambient temperature for 2.5 days. Progress of the reaction was monitored by TLC. After ca 90% conversion to the product, the reaction mixture was quenched with satd. NaHC03 solution (100 mL), followed by extraction with DCM (2x50 mL). The combined organic layers were dried (MgS04), filtered and evaporated to dryness.
Purification of the residue (6300 mg) on silica gel by gradient elution (5-50% EtOAc in hexane) afforded 2.50 g (54%) 1-[2-(2-chloroethoxy)phenyl]-2-cyclohexyl-ethanone, purity 95%. ESI-MS m/z calculated for [M+H]+: 281 .13; found: 281.09. 1H-NMR (400 MHz, CDCI3) δ 7.61 (dd, J 1 .8 Hz, J 7.6 Hz, 1 H), 7.42 (ddd, J 1.8 Hz, J 7.4 Hz, J 8.3 Hz, 1 H), 7.02 (dt, J 0.9 Hz, J 7.5 Hz, 1 H), 6.90 (br d, J 8.3 Hz, 1 H), 4.34-4.27 (m, 2H), 3.90-3.81 (m, 2H), 2.92 (d, J 6.8 Hz, 2H), 1.99-1 .88 (m, 1 H), 1.75-1.61 (m, 5H), 1.32-1.09 (m, 3H), 1 .03-0.93 (m, 2H).
Step d: To a solution of 1-[2-(2-chloroethoxy)phenyl]-2-cyclohexylethanone (1000 mg) in dioxane/diethyl ether (1 : 1 , 60 mL) was added bromine (180 pL), and the brown mixture was stirred at ambient temperature for 30 min. The progress of the reaction was followed by LCMS. After 30 min reaction time, more bromine (9 pL) was added and stirring was continued for 30 min. The reaction mixture was partitioned between EtOAc (150 ml) and NaHC03 (50 ml). The aqueous layer was separated and extracted with EtOAc (50 ml). The combined organic layers were washed with brine (50 mL), dried (Na2S04/MgS04) and concentrated to leave 2-bromo-1 -[2-(2-chloroethoxy)phenyl]-2-cyclohexylethanone as a pale yellow solid: 1.157 mg (90% yield, ca 95% purity). ESI-MS m/z calculated for [M+H]+: 359.03/361.02/363.02; found: 359.02/360.99/363.00. 1H-NMR (400 MHz, CDCI3) δ 7.72 (dd, J 1 .8 Hz, J 7.7 Hz, 1 H), 7.47 (ddd, J 1.8 Hz, J 7.4 Hz, J 8.4 Hz, 1 H), 7.06 (dt, J 0.9 Hz, J 7.6 Hz, 1 H), 6.90 (br d, J 8.4 Hz, 1 H), 5.39 (d, J 8.4 Hz, 1 H), 4.36 (br t, J 5.4 Hz, 2H), 3.96-3.84 (m, 2H), 2.14-2.06 (m, 2H), 1.80-1.76 (m, 2H), 1.72-1 .63 (m, 2H), 1.36-1.07 (m, 5H).
Step e: To a solution of methyl 5-amino-1 H-pyrazole-3-carboxylate hydrochloride (270 mg) in anhydrous DMF (18 mL) under an atmosphere of argon was added sodium hydride (60% in mineral oil, 130 mg) at 0 °C. The reaction mixture was stirred at this temperature for 45 min followed by addition of a solution of 2-bromo-1 -[2-(2-chloroethoxy)phenyl]-2- cyclohexylethanone (500 mg) in anhydrous DMF (18 mL). The reaction mixture was stirred at rt for 21 hrs, followed by heating at 100 °C for 1 hr. The progress of the reaction was monitored by LCMS. Reaction mixture was acidified with glacial acetic acid, concentrated on the rotavap (60 °C), the residue was partitioned between water and EtOAc (200 mL, 1 : 1 ), the aqueous layer was extracted with EtOAc (100 mL), the combined organic layers were dried (MgS04), filtered and evaporated to dryness to leave a brown oil. Purification on silica gel by gradient elution (20-50% EtOAc in hexane) afforded 81 mg (15% yield) methyl 13- cyclohexyl-6,7-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5-c/][1 ,4]benzoxazepine-10-carboxylate (xi) as a white solid. ESI-MS m/z calculated for [M+H]+: 366.18; found: 366.12.1H-NMR (400 MHz, CDCI3) δ 7.44-7.38 (m, 2H), 7.29 (dd, J = 1.0 Hz, J = 7.5 Hz, 1 H), 7.21 (d, J = 8.0 Hz, 1H), 6.27 (s, 1H), 4.47 (t, J 5.7 Hz, 2H), 4.04 (t, J 5.7 Hz, 2H), 3.95 (s, 3H), 3.07-2.99 (m, 1H), 2.42-2.31 (m, 2H), 1.88-1.81 (m, 4H), 1.74-1.70 (m, 1H), 1.51-1.31 (m, 3H).
Step f: A solution of methyl 13-cyclohexyl-6,7-dihydropyrazolo[5',1':2,3]imidazo[1,5- c][1,4]benzoxazepine-10-carboxylate (xi) (40 mg) in 1M aquous KOH/1 ,4-dioxane (6 mL, 1:1) was heated at reflux for 15 min. Reaction mixture was cooled to 0°C, and adjusted to pH 5 with 1 M HCI, then extracted with EtOAc (2x 20 mL), the combined organic layers were washed with brine (10 mL), dried (MgS04), filtered and evaporated to dryness: 31.3 mg (81 % yield) oil. Purification on silica gel by gradient elution (0-20% MeOH in EtOAc) and subsequent freeze drying afforded 31 mg (81% yield) 13-cyclohexyl-6J- dihydropyrazolo[5',1':2,3]imidazo[1,5-c/][1,4]benzoxazepine-10-carboxylic acid (xii) as white solid. ESI-MS m/z calculated for [M+H]+: 352.16; found: 352.14.1H-NMR (400 MHz, c4- methanol) δ 7.48-7.44 (m, 2H), 7.34-7.30 (m, 1H), 7.23 (brd, J = 8.1 Hz, 1H), 6.35 (s, 1H), 4.44 (t, J = 5.8 Hz, 2H), 4.16 (t, J = 5.8 Hz, 2H), 3.07-2.99 (m, 1H), 2.44-2.33 (m, 2H), 1.90- 1.87 (m, 2H), 1.79-1.71 (m, 3H), 1.56-1.29 (m, 3H).
Synthesis of methyl 3-(benzyloxy)-13-cyclohexyl-6, 7-dihydropyrazolof5', 1 '.2,31 imidazof 1, 5- dl[1,4lbenzoxazeDine-10-carboxylate (xiii) and methyl 13-cvclohexyl-3-hydroxy-6,7- dih ydroD yrazolo[5', 1 '.2, 3/ imidazof 1,5-dlf1,4 Ibenzoxazeoine- 10-carboxylate (xiv)
Figure imgf000040_0001
(xiii) (xiv)
Step a: Resorcinol (2370 mg) and boron trifluoride etherate (4.50 mL) were combined in a RBF under an atmosphere of argon (suspension) and stirred at rt. Cyclohexylacetyl chloride (3.00 mL) was added over a period of 30 min and the reaction mixture was stirred at rt o/n. The reaction mixture was added dropwise to 200 mL 10% aqueous NaOAc solution, stirred at rt for 30 min, then extracted into ether (2x100 mL). The combined organic layers were dried (MgS04), filtered and evaporated to dryness. Residual acetic acid was removed by co- evaporation with toluene (3 x 100 mL). The residue was subjected to purification on normal phase silica by gradient elution (20-50% EtOAc in hexane) to afford 4.6 g (quantitative yield) 2-cyclohexyl-1-(2,4-dihydroxyphenyl)ethanone. ESI-MS m/z calculated for [M+H]+: 235.13; found: 235.09.1H-NMR (400 MHz, CDCI3)512.95 (s, 1H), 7.67-7.64 (m, 1H), 6.39-6.37 (m, 2H), 5.74 (brs, 1H), 2.74 (d, J6.9 Hz, 2H), 1.98-1.87 (m, 1H), 1.78-1.63 (m, 5H), 1.34-1.10 (m, 3H), 1.08-0.98 (m, 2H). Step b: To a solution of 2-cyclohexyl-1-(2,4-dihydroxyphenyl)ethanone in anhydrous acetonitrile (100 mL) was added under an atmosphere of argon potassium carbonate (4.20 g) and benzyl bromide ( 2.40 mL) and the reaction mixture was stirred at rt for 17 hrs. After this period diethyl ether (300 mL) was added and the mixture was filtered. The filter cake was washed with diethyl ether, and the filtrate was evaporated to dryness to afford 6.15 g of a colourless solid. Purification on normal phase silica by gradient elution (1-5% EtOAc in hexane) afforded 5.79 g (91 % yield) product 1-[4-(benzyloxy)-2-hydroxyphenyl]-2- cyclohexylethanone as colourless solid. ESI-MS m/z calculated for [M+H]+: 325.18; found: 325.15. H NM (400 MHz, CDCI3) δ 12.98 (s, 1 H), 7.67 - 7.64 (m, 1 H), 7.44 - 7.32 (m, 5H), 6.53 - 6.49 (m, 2H), 5.09 (s, 2H), 2.75 (d, J = 6.9 Hz, 2H), 2.00 - 1 .87 (m, 1 H), 1 .79 - 1 .62 (m, 5H), 1.38 - 1.09 (m, 3H), 1.1 1 - 0.97 (m, 2H).
Step c: To a solution of 1-[4-(benzyloxy)-2-hydroxyphenyl]-2-cyclohexylethanone (3000 mg) in 1 ,2-dichloroethane (70 mL) was added 1-bromo-2-chloroethane (1200 μί),
tetrabutylammonium bromide (300 mg), water (70 mL) and 7 N NaOH (1800 μί). The reaction mixture was heated at reflux. The progress of the reaction was monitored by TLC (EtOAc-hexane 1 :4). More 1 -bromo-2-chloroethane (1.2 mL) and 7N NaOH (1 .8 mL) was added after 19 hrs and the reaction mixture was heated at reflux for another 6.5 hrs. Another batch of 1-bromo-2-chloroethane (1 .2 mL) and 7N NaOH (1.8 mL) was added after this period and reaction mixture was heated at reflux for another 24 hrs to reach near completion. Work-up: the organic layer was separated, the aqueous layer was extracted with DCM (2x100 mL), the combined organic layers were dried (MgS04), filtered and
concentrated to afford a clear syrup (4.85 g). Purification of the residue on normal phase silica by gradient elution (5-20% EtOAc in hexane) afforded 2950 mg (82% yield) product 1 - [4-(benzyloxy)-2-(2-chloroethoxy)phenyl]-2-cyclohexylethanone. ESI-MS m/z calculated for [M+H]+: 387.17; found: 387.17. 1H-NMR (400 MHz, CDCI3) δ 7.75 (d, J = 8.7 Hz, 1 H), 7.44- 7.33 (m, 5H), 6.63 (dd, J = 8.7 Hz, 2.2 Hz, 1 H), 6.48 (d, J = 2.2 Hz, 1 H), 5.10 (s, 2H), 4.27 (dd, J = 5.6 Hz, 5.4 Hz, 2H), 3.85 (dd, J = 5.6 Hz, 5.4 Hz, 2H), 2.89 (d, J = 6.8 Hz, 2H), 1 .99-1.88 (m, 1 H), 1 .74-1 .62 (m, 5H), 1 .31-1.09 (m, 3H), 1 .03-0.94 (m, 2H).
Step d: To a solution of 1-[4-(benzyloxy)-2-(2-chloroethoxy)phenyl]-2-cyclohexylethanone (2850 mg) in dioxane/diethyl ether (1 : 1 , 160 mL) was added bromine (450 μί) dropwise within 2 min, and the mixture was stirred at ambient temperature. The reaction was monitored by LCMS. More bromine (2 x 10 μί) was added in 10 min intervals to reach complete conversion of 1-[4-(benzyloxy)-2-(2-chloroethoxy)phenyl]-2-cyclohexylethanone. Work-up 20 min after last addition: the reaction mixture was partitioned between EtOAc (400 ml) and NaHC03 (400 ml). The aqueous layer was separated and extracted with EtOAc (100 ml). The combined organic layers were washed with brine (100 mL), dried (MgS04) and concentrated to leave a colourless solid: 341 1 mg 1 -[4-(benzyloxy)-2-(2- chloroethoxy)phenyl]-2-bromo-2-cyclohexylethanone that contained ca 1 1 % bis-bromo product.
Purification of 1.2 g of this material on normal phase silica by gradient elution (5-10-20% EtOAc in hexane) afforded 810 mg (24%) pure 1-[4-(benzyloxy)-2-(2-chloroethoxy)phenyl]-2- bromo-2-cyclohexylethanone (purity >95%).
The remainder of the crude material was recrystallised from heptane-EtOAc ca 20: 1 to afford a colourless solid: 2250 mg (56%) 1-[4-(benzyloxy)-2-(2-chloroethoxy)phenyl]-2-bromo-2- cyclohexylethanone (purity 89%). Combined yield of 1-[4-(benzyloxy)-2-(2- chloroethoxy)phenyl]-2-bromo-2-cyclohexylethanone: 3060 mg (80%). ESI-MS m/z calculated for [M+H]+: 465.09/467.09; found: 465.07/467.03. 1 H NMR (400 MHz, CDCI3) δ
7.83 (d, J = 8.8 Hz, 1 H), 7.53 - 7.31 (m, 5H), 6.66 (dd, J = 8.8, 2.2 Hz, 1 H), 6.48 (d, J = 2.2 Hz, 1 H), 5.43 (d, J = 8.7 Hz, 1 H), 5.1 1 (s, 2H), 4.42 - 4.15 (m, 2H), 4.08 - 3.81 (m, 2H), 2.21 - 2.05 (m, 2H), 1.88 - 1 .60 (m, 4H), 1 .42 - 0.99 (m, 5H).
Step e: To a solution of methyl 5-amino-1 A7-pyrazole-3-carboxylate hydrochloride (350 mg) in anhydrous DMF (15 ml_) was added under an atmosphere of argon sodium hydride (60% in mineral oil, 175 mg) at 0 °C. The reaction mixture was stirred at this temperature for 20 min followed by addition of a solution of 1-[4-(benzyloxy)-2-(2-chloroethoxy)phenyl]-2-bromo- 2-cyclohexylethanone (810 mg) in anhydrous DMF (30 ml_). The progress of the reaction was monitored by LCMS. The reaction mixture was stirred at rt for 17 hrs, then heated at 100 °C for 1 hr. After this period the reaction mixture was cooled to rt, acidified with glacial acetic acid and concentrated on the rotavap (60 °C). The residue was partitioned between water and EtOAc (100 ml_, 1 : 1 ), the aqueous layer was extracted with EtOAc (50 mL), the combined organic layers were washed with brine (20 mL), dried (MgS04), filtered and evaporated to dryness to leave a brown oil. Purification on normal phase silica by gradient elution (20-33-50% EtOAc in hexane) followed by purification on reverse phase silica by gradient elution (C18 0-100% acetonitrile in water) afforded after freeze-drying 81.5 mg (10% yield) compound methyl 3-(benzyloxy)-13-cyclohexyl-6,7- dihydropyrazolo[5',1 ':2,3]imidazo[1 ,5-c/][1 ,4]benzoxazepine-10-carboxylate (xiii)
as white solid. ESI-MS m/z calculated for [M+H]+: 472.23; found: 472.24. 1 H NMR (400
MHz, CDCI3) δ 7.47 - 7.33 (m, 5H), 7.29 (d, J = 8.5 Hz, 1 H), 6.90 (dd, J = 8.5, 2.6 Hz, 1 H),
6.84 (d, J = 2.5 Hz, 1 H), 6.26 (s, 1 H), 5.1 1 (s, 2H), 4.45 (t, J = 5.6 Hz, 2H), 4.07 (t, J = 5.6 Hz, 2H), 3.94 (s, 3H), 3.05 - 2.95 (m, 1 H), 2.41 - 2.28 (m, 2H), 1 .89 - 1.69 (m, 5H), 1.51 - 1 .28 (m, 3H).
Step f: To a solution of methyl 3-(benzyloxy)-13-cyclohexyl-6,7- dihydropyrazolo[5',1 ':2,3]imidazo[1 ,5-c/][1 ,4]benzoxazepine-10-carboxylate (xiii) (15 mg) in methanol (12 ml_) was added palladium on carbon (10%, 3 mg) and the mixture was stirred under a hydrogen atmosphere (4 L gasbag) for 65 min. Work-up: reaction mixture was flushed with argon and filtered through filter aid (Hiflo medium) with methanol. The filtrate was evaporated to dryness. Purification on normal phase silica by gradient elution (20-50% EtOAc in hexane) afforded 8.2 mg (66% yield) methyl 13-cyclohexyl-3- hydroxy-6,7-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5-c ][1 ,4]benzoxazepine-10-carboxylate (xiv) as white solid. ESI-MS m/z calculated for [M+H]+: 382.1 1 ; found: 382.18. 1 H NMR (400 MHz, de-acetone) δ 8.92 (s, 1 H), 7.31 (d, J = 8.4 Hz, 1 H), 6.81 (dd, J = 8.4, 2.5 Hz, 1 H), 6.70 (d, J = 2.5 Hz, 1 H), 6.29 (s, 1 H), 4.47 (t, J = 5.7 Hz, 2H), 4.24 (t, J = 5.6 Hz, 2H), 3.83 (s, 3H), 3.00 (tt, J = 12.3, 3.4 Hz, 1 H), 2.48 - 2.36 (m, 2H), 1.89 - 1 .70 (m, 5H), 1 .47 - 1 .32 (m, 3H).
Synthesis of 3-(benzyloxy)-13-cvclohexyl-6, 7-dihydroDyrazolo[5', 1 ':2,3limidazof 1, 5- dlf1,4lbenzoxazepine-10-carbox lic acid (xy)
Figure imgf000043_0001
A solution of methyl 3-(benzyloxy)-13-cyclohexyl-6,7-dihydropyrazolo[5', 1 ':2,3] imidazo[1 ,5- c ][1 ,4]benzoxazepine-10-carboxylate (xiii) (7 mg) in 1 M aqueous KOH/1 ,4-dioxane (2 imL, 1 :1 ) was heated at reflux for 5 min. The reaction mixture was cooled to 0°C, adjusted to pH 5 with 1 M HCI, and extracted with EtOAc (2x 10 mL). The combined organic layers were washed with brine (5 mL), dried (MgS04), filtered and evaporated to dryness to afford a solid. The crude material was purified on reverse phase silica by gradient elution (C18, 0- 100% acetonitrile/H20) to afford after freeze-drying 5.3 mg (81 % yield) 3-(benzyloxy)-13- cyclohexyl-6,7-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5-c/][1 ,4]benzoxazepine-10-carboxylic acid (xv) as a white solid. ESI-MS m/z calculated for [M+H]+: 458.21 ; found: 458.14. 1H NMR (400 MHz, c/6-DMSO) δ 7.50 - 7.46 (m, 2H), 7.45 - 7.39 (m, 2H), 7.36 (dt, J = 5.1 , 2.1 Hz, 1 H), 7.33 (d, J = 8.6 Hz, 1 H), 7.00 (dd, J = 8.6, 2.6 Hz, 1 H), 6.90 (d, J = 2.6 Hz, 1 H), 6.27 (s, 1 H), 5.16 (s, 2H), 4.41 (t, J = 5.6 Hz, 2H), 4.18 (t, J = 5.5 Hz, 2H), 2.87 (br t, J = 12.1 Hz, 1 H), 2.34 - 2.21 (m, 2H), 1 .85 - 1 .64 (m, 5H), 1 .41 - 1 .24 (m, 3H). Synthesis of 13-cvclohexyl-3-hvdroxy-6, 7-dihvdrooyrazoloi5', 1 '.2, 3limidazo[ 1, 5- d][1 lbenzoxazepine-10-carboxylic acid (xyi)
Figure imgf000044_0001
(xvi)
A solution of methyl 13-cyclohexyl-3-hydroxy-6J-dihydropyrazolo[5', 1 ':2,3] imidazo[1 ,5- c ][1 ,4]benzoxazepine-10-carboxylate (xiv) (5 mg) in 1 M aqueous KOH/1 ,4-dioxane (2 imL, 1 :1 ) was heated at 40 °C for 25 min. The reaction mixture was cooled to 0°C, adjusted to pH 6 with 1 M HCI, and extracted with EtOAc (2x 10 mL). The combined organic layers were washed with brine (5 mL), dried (MgS04), filtered and evaporated to dryness to afford a solid. The crude material was purified on reverse phase silica by gradient elution (C18; 0- 100% acetonitrile/H20) to afford after freeze-drying 1 .5 mg (31 % yield) 13-cyclohexyl-3- hydroxy-6,7-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5-c ][1 ,4]benzoxazepine-10-carboxylic acid
(xvi) as a white solid. ESI-MS m/z calculated for [M+H]+: 368.16; found: 368.10. 1 H NMR (400 MHz, c/4-methanol) δ 7.24 (d, J = 8.4 Hz, 1 H), 6.74 (dd, J = 8.4, 2.5 Hz, 1 H), 6.64 (d, J = 2.4 Hz, 1 H), 6.30 (s, 1 H), 4.40 (t, J = 5.6 Hz, 2H), 4.14 (t, J = 5.6 Hz, 2H), 3.03 - 2.94 (m, 1 H), 2.43 - 2.29 (m, 2H), 1.93 - 1.83 (m, 2H), 1.81 - 1 .66 (m, 3H), 1 .58 - 1 .32 (m, 3H). Synthesis of ethyl 3-(benzyloxy)-13-cvclohexyl-6, 7-dihydroimidazo[2', 1 ':2, 31imidazo [1,5- d}[1, 41benzoxazepine- 10-carboxylate (xyii)
Figure imgf000044_0002
(xvii)
Compound (xvii) was synthesised analogously to compound (xiii) but starting with ethyl 2- amino-1 /-/-imidazole-5-carboxylate (100 mg, 0.645 mmol), which was reacted in DMF with 1 .25 eq NaH (10 min at 0 °C) followed by 0.93 eq 1 -[4-(benzyloxy)-2-(2- chloroethoxy)phenyl]-2-bromo-2-cyclohexylethanone (18 h rt and 4.5 h at 100 °C). Yield of product ethyl 3-(benzyloxy)-13-cyclohexyl-6,7-dihydroimidazo[2', 1 ':2,3] imidazo[1 ,5- d][1 ,4]benzoxazepine-10-carboxylate (xvii) 29 mg (10%). ESI-MS m/z calculated for [M+H]+: 486.24; found: 486.19. 1H NMR (400 MHz, CDCI3) δ 7.85 (s, 1 H), 7.49 - 7.31 (m, 5H), 7.21 (d, J = 8.5 Hz, 1 H), 6.87 (dd, J = 8.5, 2.6 Hz, 1 H), 6.83 (d, J = 2.5 Hz, 1 H), 5.10 (s, 2H), 4.48 - 4.38 (m, 4H), 4.24 (t, J = 5.6 Hz, 2H), 2.96 - 2.84 (m, 1 H), 1 .96 - 1.73 (m, 7H), 1.41 (t, J = 7.1 Hz, 3H), 1 .45 - 1.30 (m, 3H).
Synthesis of methyl 13-cyclohexyl-3-{[2-( morpholin-4- yl)-5- ( 2-oxop yrrolidin- 1 - vDbenzylloxy}-
6, 7-dih ydroD yrazolo[5', 1 ': 2, 3limidazo[ 1,5-dlf1.41benzoxaze ine- 10-carboxylate (xyiii) and
13-cvclohexyl-3-{[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1-yl)benzylloxy}-6, 7-
Figure imgf000045_0001
Step a: To a solution of (xiv) (10 mg) in anhydrous DMF (1 mL) was added at 0 °C and under under an atmosphere of argon NaH (1 .3 mg). The mixture was stirred at this temperature for 30 min. A solution of 1-[3-(chloromethyl)-4-(morpholin-4-yl)phenyl]pyrrolidin-
2- one (10 mg) in anhydrous DMF (1 mL) was added and the reaction mixture was stirred at rt for 1 hr and at 60 °C for 1 hr. The reaction mixture was cooled to rt and poured into satd NaHC03 solution (5 mL), extracted with EtOAc (2x5 mL), the combined organic extracts were washed with satd brine (2 mL), dried (MgS04), filtered and evaporated to dryness. Purification on normal phase silica by gradient elution (20-100% EtOAc in hexane) and freeze-drying afforded methyl 13-cyclohexyl-3-{[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1- yl)benzyl]oxy}-6,7-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5-c/][1 ,4]benzoxazepine-10- carboxylate (xviii) (9 mg, 54% yield) as a solid. ESI-MS m/z calculated for [M+H]+: 640.31 ; found: 640.23. 1 H NMR (400 MHz, CDCI3) δ 7.74 (d, J = 2.6 Hz, 1 H), 7.62 (dd, J = 8.7, 2.7 Hz, 1 H), 7.29 - 7.26 (m, 1 H), 7.19 (d, J = 8.7 Hz, 1 H), 6.90 - 6.86 (m, 2H), 6.26 (s, 1 H), 5.20 (s, 2H), 4.45 (t, J = 5.6 Hz, 2H), 4.07 (t, J = 5.6 Hz, 2H), 3.94 (s, 3H), 3.88 - 3.83 (m, 6H), 3.04 - 2.93 (m, 5H), 2.60 (t, J = 8.1 Hz, 2H), 2.41 - 2.28 (m, 2H), 2.21 - 2.12 (m, 2H), 1 .89 - 1 .68 (m, 5H), 1.51 - 1.30 (m, 3H).
Step b: A solution of (xviii) (6 mg) in 1 M aqueous KOH/1 ,4-dioxane (3 mL, 1 : 1 ) was heated at 40 °C for 30 min, then stirred at rt for 2 hrs. The reaction mixture was cooled to 0°C, and adjusted to pH 5 with 1 M HCI, and extracted with EtOAc (2x 10 mL). The combined organic layers were washed with brine (5 mL), dried (MgS04), filtered and evaporated to dryness to afford a solid. The crude material was purified on reverse phase silica by gradient elution (C18; 0-100% acetonitrile/H20) and freeze-dried to afford 2.5 mg (43% yield) 13-cyclohexyl-
3- {[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1 -yl)benzyl]oxy}-6,7- dihydropyrazolo[5',1 ':2,3]imidazo[1 ,5-c/][1 ,4]benzoxazepine-10-carboxylic acid (xix) as a white solid. ESI-MS m/z calculated for [M+H]+: 626.30; found: 626.25. 1 H NMR (400 MHz, c/4-methanol) δ 7.78 (d, J = 2.6 Hz, 1 H), 7.55 (dd, J = 8.7, 2.7 Hz, 1 H), 7.32 (d, J = 8.4 Hz, 1 H), 7.25 (d, J = 8.7 Hz, 1 H), 6.96 - 6.90 (m , 2H), 6.31 (s, 1 H), 5.25 (s, 2H), 4.41 (t, J = 5.6 Hz, 2H), 4.14 (t, J = 5.6 Hz, 2H), 3.90 (t, J = 7.1 Hz, 2H), 3.86 - 3.82 (m , 4H), 2.98 - 2.95 (m , 5H), 2.58 (t, J = 8.1 Hz, 2H), 2.42 - 2.29 (m , 2H), 2.21 - 2.12 (m , 2H), 1 .90 - 1 .83 (m, 2H), 1 .78 - 1 .68 (m , 3H), 1 .57 - 1 .32 (m, 3H).
Synthesis of 3-(benzyloxy)-13-cyclohexyl-6, 7-dihydroimidazo[2', 1 ':2, 31imidazo [ 1, 5- dli1, 4lbenzoxazeDine-10-carbox lic acid (xx)
Figure imgf000046_0001
Com pound (xx) was synthesised analogously to compound (xv) but starting with ethyl 3- (benzyloxy)-13-cyclohexyl-6,7-dihydroimidazo[2', 1 ':2,3] imidazo[1 ,5-d][1 ,4]benzoxazepine- 10-carboxylate (xvii) (22 mg, 0.0453 mmol), which was reacted in 1 M aqueous KOH/1 ,4- dioxane (10 mL, 1 : 1 ) at reflux for 5 min. Yield of product 3-(benzyloxy)-13-cyclohexyl-6,7- dihydroimidazo[2', 1 ':2,3] imidazo[1 ,5-d][1 ,4]benzoxazepine-10-carboxylic acid (xx) after additional purification by recrystallisation from EtOAc: 9.2 mg (44%). ESI-MS m/z calculated for [M+H]+: 458.2; found: 458.2. 1 H NMR (400 MHz, DMSO) δ 8.06 (s, 1 H), 7.50 - 7.29 (m, 6H), 6.99 (dd, J = 8.5, 2.4 Hz, 1 H), 6.91 (d, J = 2.4 Hz, 1 H), 5.16 (s, 2H), 4.40 (t, J = 5.4 Hz, 2H), 4.14 (t, J = 5.2 Hz, 2H), 2.91 - 2.77 (m, 1 H), 1 .98 - 1 .60 (m , 7H), 1 .51 - 1.25 (m , 3H); signal for C02H not observed.
Synthesis of ethyl 13-cvclohexyl-3-hydroxy-6, 7-dihvdroimidazo[2', 1 ':2, 3limidazo[1, 5- d][1, 4]benzoxazepine- 10-carboxylate (xxi)
Figure imgf000046_0002
Com pound (xxi) was synthesised analogously to compound (xiv) but starting with ethyl 3- (benzyloxy)-13-cyclohexyl-6,7-dihydroimidazo[2', 1 ':2,3] imidazo[1 ,5-d][1 ,4]benzoxazepine- 10-carboxylate (xvii) (50 mg, 0.103 mmol), which was reacted in ethanol (40 mL) with hydrogen and palladium on carbon (10%, 10 mg) for 65 min. Yield of product ethyl 13- cyclohexyl-3-hydroxy-6,7-dihydroimidazo[2', 1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10- carboxylate (xxi): 21 mg (52%). ESI-MS m/z calculated for [M+H]+: 396.19; found: 396.15. 1 H NMR (400 MHz, CDCI3 + 5% CD3OD) δ 7.82 (s, 1 H), 7.11 (d, J = 8.4 Hz, 1 H), 6.72 (dd, J = 8.3, 2.5 Hz, 1 H), 6.67 (d, J = 2.4 Hz, 1 H), 4.45 - 4.33 (m, 4H), 4.19 (t, J = 5.6 Hz, 2H), 2.94 - 2.83 (m, 1 H), 1.91 - 1.73 (m, 7H), 1 .39 (t, J = 7.1 Hz, 3H), 1.42 - 1 .26 (m, 3H); signal for OH not observed.
Synthesis of ethyl 13-cvclohexyl-3-{[2-(morDholin-4-yl)-5-(2-oxoDyrrolidin-1-yl)benzylloxy}- 6, 7-dihydroimidazo[2', 1 ':2, 31imidazo[ 1, 5-dli 1 Albenzoxazepine-10-carboxylate (xxii)
Figure imgf000047_0001
Compound (xxii) was synthesised analogously to compound (xviii) but starting with ethyl 13-cyclohexyl-3-hydroxy-6J-dihydroimidazo[2', 1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10- carboxylate (xxi) (8 mg, 0.0202 mmol), which was reacted in DMF (2 mL) with NaH (60% in mineral oil, 1.0 mg), followed by a solution of 1-[3-(chloromethyl)-4-(morpholin-4- yl)phenyl]pyrrolidin-2-one (16 mg) in anhydrous DMF (0.9 mL). Yield of product ethyl 13- cyclohexyl-3-{[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1 -yl)benzyl]oxy}-6,7- dihydroimidazo[2', 1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10-carboxylate (xxii) after additional purification on reverse phase silica (C18, 0-100% ACN/H20): 5.96 mg (45%). ESI- MS m/z calculated for [M+H]+: 654.33; found: 654.31 . 1 H NMR (400 MHz, CDCI3) δ 7.85 (s,
1 H), 7.73 (d, J = 2.6 Hz, 1 H), 7.62 (dd, J = 8.7, 2.7 Hz, 1 H), 7.20 (d, J = 2.5 Hz, 1 H), 7.18 (d, J = 2.8 Hz, 1 H), 6.91 - 6.83 (m, 2H), 5.19 (s, 2H), 4.48 - 4.38 (m, 4H), 4.25 (t, J = 5.6 Hz, 2H), 3.90 - 3.81 (m, 6H), 2.98 - 2.92 (m, 4H), 2.98 - 2.84 (m, 1 H), 2.60 (t, J = 8.1 Hz, 2H), 2.22 - 2.1 1 (m, 2H), 1.95 - 1.75 (m, 7H), 1 .42 (t, J = 7.1 Hz, 3H), 1.44 - 1 .30 (m, 3H).
Synthesis of 13-cvclohexyl-3-{i2-(mor holin-4-yl)-5-(2-oxoDyrrolidin-1-yl)benzylloxy}-6, 7- dihvdroimidazof2', 1 ':2,3limidazof1,5-dlf1,4lbenzoxazeDine-10-carboxylic acid (xxiii)
Figure imgf000047_0002
Compound (xxiii) was synthesised analogously to compound (xix) but starting with ethyl 13- cyclohexyl-3-{[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1 -yl)benzyl]oxy}-6,7- dihydroimidazo[2', 1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10-carboxylate (xxii) (7.7 mg, 0.012 mmol), which was reacted in 1 M aqueous KOH/1 ,4-dioxane (10 mL, 1 :1 ) at 40 °C for 2.5 hrs. Yield of product 13-cyclohexyl-3-{[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1- yl)benzyl]oxy}-6,7-dihydroimidazo[2', 1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10-carboxylic acid (xxiii): 2.5 mg (33%). ESI-MS m/z calculated for [M+H]+: 626.30; found: 626.27. 1H NM (400 MHz, DMSO) δ 7.90 (br s, 1 H), 7.82 (d, J = 2.6 Hz, 1 H), 7.56 (dd, J = 8.8, 2.7 Hz, 1 H), 7.29 (d, J = 9.2 Hz, 1 H), 7.22 (d, J = 8.8 Hz, 1 H), 7.00 - 6.94 (m, 2H), 5.20 (s, 2H), 4.39 (t, J = 5.5 Hz, 2H), 4.17 - 4.1 1 (m, 2H), 3.81 (t, J = 7.0 Hz, 2H), 3.77 - 3.73 (m, 4H), 2.91 - 2.86 (m, 4H), 2.86 - 2.78 (m, 1 H), 2.46 (partially obscured t, J = 8.3 Hz, 2H), 2.10 - 2.00 (m, 2H), 1.96 - 1.84 (m, 2H), 1.83 - 1.63 (m, 5H), 1.50 - 1 .26 (m, 3H); signal for CO2H not observed.
Example syntheses of compounds of formulae (I) and (II) via amide coupling
Synthesis of ethyl (2E)-3 4-({[1-({[3-cvclohexyl-2-(4-fluoroDhenyl)-1-methyl-M 2- blDyrazol-6-yllcarbonyl}amino)cvclobutyllcarbonyl} amino)DhenyllDroD-2-enoate ( 1 ); (E)-3-[4- Π 1-[[3-cvclohexyl-2-(4-fluoroDhenyl)-1-methyl-imidazo[ 1, 2-bloyrazole-6- carbonyllaminolcvclobutanecarbonyll aminolDhenyllDroD-2-enoic acid (2) and methyl (E)-3- \4-\\ 1-[[3-cvclohexyl-2-(4-fluoroDhenyl)- 1-methyl-imidazoi 1, 2-bl yrazole-6-carbonyllaminol cvclobutanecarbonyllaminolphenyllprop-2-enoate (3)
Figure imgf000048_0001
Step a: To a solution of 3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-6- carboxylic acid (iv) (5.0 mg; 0.01 mmol) in DMF (1 mL) at ambient temperature was added ethyl (2E)-3-(4-{[(1 -aminocyclobutyl)carbonyl] amino}phenyl)prop-2-enoate hydrochloride (0.02 mmol) followed by HATU (16.7 mg; 0.04 mmol) and DIPEA (5.7 mg; 0.04 mmol). After 16 hours a saturated aqueous solution of NaHC03 (15 ml) was added and the aqueous layer was extracted with EtOAc (3 x 15 mL). The combined organics were washed with brine (10 mL), dried (MgS04) and concentrated to leave a brown oil (8 mg). Purification by reverse phase LCMS: Acetonitrile/water (0.1 % formic acid) afforded ethyl (2E)-3-[4-({[1-({[3- cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazol-6- yl]carbonyl}amino)cyclobutyl]carbonyl}amino)phenyl]prop-2-enoate (1 ) (3.8 mg) as an amorphous white solid. ESI-MS m/z calculated for [M+H]+: 612.30; found: 61 1.95. 1H NMR (400 MHz, CD3CN) δ 9.19 (s, 1 H), 7.91 (s, 1 H), 7.68 - 7.58 (m, 5H), 7.53 - 7.48 (m, 2H), 7.35 - 7.29 (m, 2H), 6.44 (d, J = 16.0 Hz, 1 H), 6.24 (s, 1 H), 4.24 (q, J = 7.1 Hz, 2H), 3.40 (s, 3H), 2.92 - 2.83 (m, 2H), 2.80 - 2.70 (m, 1 H), 2.47 - 2.36 (m, 2H), 2.30 - 2.04 (m, 4H), 1.91 - 1 .69 (m, 5H), 1.45 - 1 .33 (m, 3H), 1 .32 (t, J = 7.1 Hz, 3H).
Step b: To a solution of ethyl (E)-3-[4-[[1-[[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl- imidazo[1 ,2-b]pyrazole-6-carbonyl]amino]cyclobutanecarbonyl]amino] phenyl]prop-2-enoate (1 ) (4 mg; 0.007 mmol) in MeOH/THF (1 : 1 ; 2 mL) was added a 1 M aqueous solution of lithium hydroxide (0.14 mmol) and the reaction mixture was heated at 60°C for 2 hrs. 1 M HCI was added until ~ pH3 and the aqueous layer was extracted with EtOAc (3 x 15 mL) and the combined organics were washed with brine (15 mL), dried (MgS04) and concentrated to leave a brown oil (4 mg). Purification by reverse phase LCMS: Acetonitrile/water (0.1 % formic acid) afforded (E)-3-[4-[[1 -[[3-cyclohexyl-2-(4-fluorophenyl)-1-methyl-imidazo[1 ,2- b]pyrazole-6-carbonyl]amino] cyclobutanecarbonyl]amino]phenyl]prop-2-enoic acid (2) (1 .21 mg) as an amorphous white solid ESI-MS m/z calculated for [M+H]+: 584.27; found: 583.95. 1 H NMR (400 MHz, CD3CN) 5 9.15 (s, 1 H), 7.88 (s, 1 H), 7.64 - 7.53 (m, 5H), 7.50 - 7.44 (m, 2H), 7.32 - 7.25 (m, 2H), 6.37 (d, J = 16.0 Hz, 1 H), 6.20 (s, 1 H), 3.36 (s, 3H), 2.89 - 2.78 (m, 2H), 2.76 - 2.66 (m, 1 H), 2.44 - 2.32 (m, 2H), 2.07 - 1 .99 (m, 4H), 1 .86 - 1.68 (m, 5H), 1 .43 - 1 .25 (m, 3H); and methyl (E)-3-[4-[[1-[[3-cyclohexyl-2-(4-fluorophenyl)-1-methyl- imidazo[1 ,2-b]pyrazole-6-carbonyl]amino] cyclobutanecarbonyl]amino]phenyl]prop-2-enoate (3) (0.64 mg) as an amorphous white solid. ESI-MS m/z calculated for [M+H]+: 598.28; found: 597.97. 1 H NMR (400 MHz, CD3CN) δ 9.16 (s, 1 H), 7.88 (s, 1 H), 7.65 - 7.54 (m, 5H), 7.51 - 7.43 (m, 2H), 7.32 - 7.25 (m, 2H), 6.42 (d, J = 16.0 Hz, 1 H), 6.20 (s, 1 H), 3.73 (s, 3H), 3.36 (s, 3H), 2.87 - 2.79 (m, 2H), 2.76 - 2.66 (m, 1 H), 2.42 - 2.31 (m, 2H), 2.12 - 2.00 (m, 4H), 1 .84 - 1.68 (m, 5H), 1.46 - 1 .25 (m, 3H).
Synthesis of ethyl (E)-3-i4-ii1-ii3-cvclohexyl-2-(4-fluorophenyl)-1-methyl-imidazoi1,2- blDyrazole-7-carbonyllaminolcyclobutanecarbonvH aminolDhenyllDroD-2-enoate (4) and (2E)-3-[4-({[1-({f3-cvclohexyl-2-(4-fluorophenyl)-1-methyl-M
Figure imgf000050_0001
Step a: To a solution of 3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-7- carboxylic acid (vii) (3 mg; 0.01 mmol) in DCM (1 mL) was added ethyl (E)-3-[4-[(1- aminocyclobutanecarbonyl)amino]phenyl]prop-2-enoate (3.8 mg; 0.01 mmol) followed by HATU (10 mg; 0.03 mmol) and DIPEA (3.4 mg; 0.03 mmol). After 2 hours a saturated aqueous solution of NaHC03 (15 ml) was added and the aqueous layer was extracted with EtOAc (3 x 15 mL). The combined organics were washed with brine (10 mL), dried (MgS04) and concentrated to leave ethyl (E)-3-[4-[[1-[[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl- imidazo[1 ,2-b]pyrazole-7-carbonyl]amino]cyclobutanecarbonyl]amino]phenyl]prop-2-enoate (4)
as a brown oil (5 mg). Crude material was used directly without purification.
ESI-MS m/z calculated for [M+H]+: 612.30; found: 612.12.
Step b: To a solution of ethyl (E)-3-[4-[[1-[[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl- imidazo[1 ,2-b]pyrazole-7-carbonyl]amino]cyclobutanecarbonyl] amino]phenyl]prop-2-enoate (4) (5 mg; 0.01 mmol) in MeOH/THF (1 : 1 ; 2 mL) was added a 1 M aqueous solution of lithium hydroxide (0.16 mmol) and the reaction was heated at 60°C for 3 hrs. 1 M HCI was added until ~pH 3 and the aqueous layer was extracted with EtOAc (3 x 30 mL). The combined organics were washed with brine (25 mL), dried (MgS04) and concentrated to leave a brown oil (4 mg). Purification by reverse phase LCMS: Acetonitrile/water (0.1 % formic acid) afforded (2E)-3-[4-({[1-({[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazol- 7-yl]carbonyl}amino)cyclobutyl] carbonyl}amino)phenyl]prop-2-enoic acid (5) (2.17 mg) as an amorphous white solid. ESI-MS m/z calculated for [M+H]+: 584.27; found: 584.12. 1H NMR (400 MHz, CD3CN) δ 9.24 (s, 1 H), 8.10 (s, 1 H), 7.67 - 7.55 (m, 5H), 7.51 - 7.46 (m, 2H), 7.32 - 7.27 (m, 2H), 7.19 (s, 1 H), 6.39 (d, J = 16.0 Hz, 1 H), 3.76 (s, 3H), 2.86 - 2.75 (m, 2H), 2.73 - 2.65 (m, 1 H), 2.39 - 2.30 (m, 2H), 2.10 - 2.02 (m, 4H), 1 .84 - 1 .68 (m, 5H), 1.35 - 1 .26 (m, 3H). Synthesis of ethyl (2E)-3-(4-{[(1-{[(3-cvclohexyl-1-methyl-2-phenyl-1H-im^ 7-yl)carbonyllamino}cvclobutyl) carbonyllamino}Dhenyl)oroD-2-enoate (6) and (2E)-3-(4-{f(1- {[(3-cyclohexyl-1-methyl-2-phenyl-1H-imidazo[1,2-blpyrazol-7-yl)carbonyllamino}cyclob carbonyllamino}Dhenyl)DroD-2-enoic acid (7)
Figure imgf000051_0001
Step a: To a solution of compound 3-cyclohexyl-1 -methyl-2-phenyl-1 H-imidazo[1 ,2- 6]pyrazole-7-carboxylic acid (x) (10 mg) and ethyl (2E)-3-(4-{[(1- aminocyclobutyl)carbonyl]amino}phenyl)prop-2-enoate hydrochloride (15 mg) in dry DMF (500 was added under an atmosphere of argon DIPEA (20 pL) followed by HATU (17.6 mg). The reaction mixture was stirred at rt for 16 hrs. Work-up: EtOAc (15 mL) was added and this mixture was washed with brine (5 mL) and concentrated. Repeated purification on normal phase silica by gradient elution (20-100% EtOAc in hexane) and subsequent freeze drying afforded compound (6) (4 mg, 22 % yield) as a white solid. ESI-MS m/z calculated for [M+H]+: 594.30; found: 594.13. 1H-NMR (400 MHz, CDCI3) δ 10.26 (br s, 1 H), 7.85 (s, 1 H), 7.63-7.44 (m, 8H), 7.37-7.34 (m, 2H), 6.33 (d, J 16.0 Hz, 1 H), 6.27 (br s, 1 H), 4.25 (q, J 7.1 Hz, 2H), 3.86 (s, 3H), 2.95-2.89 (m, 2H), 2.76-2.68 (m, 1 H), 2.37-2.30 (m, 2H), 2.09-1.96 (m, 4H), 1.79-1.66 (m, 5H), 1.32 (t, J 7.1 Hz, 3H), 1.34-1 .24 (m, 3H).
Step b: A suspension of compound (6) (4.5 mg) in 1 ,4-dioxane/isopropanol/0.6 M aqueous LiOH (0.1 mL/0.1 mL/0.08 mL) was stirred at 60-85 °C for 35 min. Work-up: the reaction mixture was cooled in ice/water, acidified with 1 M HCI and concentrated. The crude material was purified on reverse phase silica by gradient elution (C18; 0-100% acetonitrile/H20) to afford after freeze drying (2£)-3-(4-{[(1-{[(3-cyclohexyl-1-methyl-2-phenyl-1 /-/-imidazo[1 ,2- 0]pyrazol-7-yl)carbonyl]amino}cyclobutyl)carbonyl]amino}phenyl)prop-2-enoic acid (7) (1.9 mg, 45% yield) as a white solid. ESI-MS m/z calculated for [M+H]+: 566.27; found: 566.13. 1 H-NMR (400 MHz, c/6-DMSO) δ 9.74 (br s, 1 H), 8.44 (br s, 1 H), 8.29 (s, 1 H), 7.66-7.64 (m, 2H), 7.56-7.50 (m, 5H), 7.45-7.43 (m, 2H), 7.37 (br d, J 15.9 Hz, 1 H), 6.36 (d, J 15.9 Hz, 1 H), 3.72 (s, 3H), 2.68-2.58 (m, 3H), 2.34-2.24 (m, 2H), 2.07-1.79 (m, 4H), 1.73-1.66 (m, 5H), 1.23-1.19 (m, 3H). Synthesis of ethyl 1-U( 13-cvclohexyl-6, 7-dihvdroDyrazolo[5 1 ':2, 3limidazo[1 , 5- dlf 1, 4lbenzoxazeDin-10-yl)carbonyllamino}cvclobutanecarboxylate (8); 1-{[(13-cvclohexyl- 6, 7-dihydropyrazolo[5', 1 '.2, 3limidazof 1, 5-dlf 1 ,4lbenzoxazepin-10- vDcarbonyllaminoicvclobutanecarboxylic acid (9); ethyl (2E)-3-(4-{[(1-{[(13-cyclohexyl-6,7- dihydroDyrazolo[5', 1 ':2,3limidazof 1, 5-dlf1 Albenzoxaze in- 10- yl)carbonyllamino}cvclobutyl)carbonyllamino}Dhenyl) roD-2-enoate (10); (2E)-3-(4-{f(1-{f(13- cvclohexyl-6,7-dihvdroDyrazoloi5',1':2,3limidazof1,5-dli1,4lbenzoxazeDin-10- l)carbonyllamino}cvclobutyl)carbonyllamino}Dhenyl) roD-2-enoic acid (11)
Figure imgf000052_0001
Step a: To a solution of 13-cyclohexyl-6J-dihydropyrazolo[5',1':2,3]imidazo[1 ,5- c][1,4]benzoxazepine-10-carboxylic acid (xii) (10 mg) and ethyl 1- aminocyclobutanecarboxylate hydrochloride (5.7 mg) in dry DMF (2 mL) under an atmosphere of argon was added DIPEA (16 pL) followed by HATU (13 mg). The reaction mixture was stirred at rt for 17 hrs. Work-up: EtOAc (15 mL) was added and the mixture was washed with brine (5 mL), dried (MgS04), filtered and concentrated. The crude material was purified on normal phase silica by gradient elution (20-100% EtOAc in hexane) to afford ethyl 1-{[(13-cyclohexyl-6J-dihydropyrazolo[5',1':2,3]imidazo[1,5-d][1,4]benzoxazepin-10- yl)carbonyl]amino}cyclobutanecarboxylate (8) as an oil (9 mg, 66 % yield). ESI-MS m/z calculated for [M+H]+: 477.25; found: 477.19.1H NMR (400 MHz, CDCI3) δ 7.62 (s, 1H),
7.43-7.37 (m, 1H), 7.35 (dd, J= 7.7, 1.7 Hz, 1H), 7.26 (td, J = 7.4, 1.2 Hz, 1H), 7.20 (dd, J = 8.0, 1.1 Hz, 1H), 6.22 (s, 1H), 4.46 (t, J = 5.7 Hz, 2H), 4.27 (q, J = 7.1 Hz, 2H), 4.07 (t, J = 5.7 Hz, 2H), 2.98 (tt, J= 12.3, 3.3 Hz, 1H), 2.81 -2.69 (m, 2H), 2.68-2.57 (m, 2H), 2.47- 2.32 (m, 2H), 2.20-2.05 (m, 2H), 1.96- 1.74 (m, 5H), 1.50- 1.34 (m, 3H), 1.31 (t, J = 7.1 Hz, 3H). Step b: A solution of compound (8) (9 mg) in 1 M aquous KOH/1 ,4-dioxane (6 mL, 1 : 1 ) was heated at reflux for 15 min, cooled to 0°C, and adjusted to pH 5 with 1 M HCI and evaporated to dryness. The residue was purified on reverse phase silica by gradient elution (C18; 0-100% acetonitrile/H20) to afford after freeze-drying 1 -{[(13-cyclohexyl-6,7- dihydropyrazolo[5',1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepin-10- yl)carbonyl]amino}cyclobutanecarboxylic acid (9) as a white solid (7 mg, 83% yield). ESI-MS m/z calculated for [M+H]+: 449.22; found: 449.19. 1H NMR (400 MHz, CDCI3) δ 7.78 (s, 1 H), 7.36 (td, J = 7.9, 1 .7 Hz, 1 H), 7.30 (dd, J = 7.6, 1.6 Hz, 1 H), 7.22 (td, J = 7.5, 1.1 Hz, 1 H), 7.15 (dd, J = 8.0, 1.0 Hz, 1 H), 6.29 (s, 1 H), 4.36 (t, J = 5.6 Hz, 2H), 4.00 (t, J = 5.5 Hz, 2H), 2.97 - 2.87 (m, 1 H), 2.77 - 2.68 (m, 2H), 2.37 - 2.24 (m, 4H), 2.01 - 1.90 (m, 2H), 1.88 - 1 .78 (m, 2H), 1.78 - 1.66 (m, 3H), 1.43 - 1 .24 (m, 3H). Signal for C02H and remaining water in the sample appeared at 4.43 ppm (br s).
Step c: To a solution of compound (9) (7 mg) and ethyl (2E)-3-(4-aminophenyl)prop-2- enoate (3.5 mg) in dry DMF (2 mL) under under an atmosphere of argon was added DIPEA (10 pL) followed by HATU (7.5 mg). The reaction mixture was stirred at rt for 20 hrs. Workup: EtOAc (15 mL) was added and the mixture was washed with brine (5 mL), dried (MgS04), filtered and concentrated. The crude material was purified on reverse phase silica by gradient elution (C18; 0-100% acetonitrile/H20) and freeze-dried to afford ethyl (2E)-3-(4- {[(1-{[(13-cyclohexyl-6J-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5-c/][1 ,4]benzoxazepin-10- yl)carbonyl]amino}cyclobutyl)carbonyl]amino}phenyl)prop-2-enoate (10) (8 mg, 82% yield) as a white solid. ESI-MS m/z calculated for [M+H]+: 622.31 ; found: 622.05. 1H NMR (400 MHz, CDCI3) 5 10.06 (s, 1 H), 7.67 - 7.59 (m, 3H), 7.54 (s, 1 H), 7.47 (d, J = 8.6 Hz, 2H), 7.44 - 7.39 (m, 1 H), 7.35 (dd, J = 7.7, 1.7 Hz, 1 H), 7.29 (dd, J = 7.5, 1.1 Hz, 1 H), 7.21 (dd, J = 8.0, 1.0 Hz, 1 H), 6.35 (d, J = 16.0 Hz, 1 H), 6.27 (s, 1 H), 4.47 (t, J = 5.6 Hz, 2H), 4.25 (q, J = 7.1 Hz, 2H), 4.09 (t, J = 5.7 Hz, 2H), 3.02 - 2.93 (m, 3H), 2.50 - 2.40 (m, 2H), 2.39 - 2.28 (m, 2H), 2.16 - 1.99 (m, 2H), 1.93 - 1 .85 (m, 2H), 1.83 - 1 .74 (m, 3H), 1 .45 - 1.35 (m, 3H), 1 .32 (t, J = 7.1 Hz, 3H).
Step d: A suspension of 9 mg compound (10) in 1 ,4-dioxane/isopropanol/0.5 M aqueous LiOH (0.4 mL/0.4 mL/0.32 mL) was stirred in a flask under an atmosphere of argon at 65 °C for 60 min. Work-up: reaction mixture was cooled in ice/water, acidified with 1 M HCI (pH 5) and concentrated. Repeated purification on reverse phase silica by gradient elution (C18; 0- 100% acetonitrile/H20) and freeze-drying afforded (2E)-3-(4-{[(1 -{[(13-cyclohexyl-6,7- dihydropyrazolo[5',1 ':2,3]imidazo[1 ,5-c/][1 ,4]benzoxazepin-10- yl)carbonyl]amino}cyclobutyl)carbonyl]amino}phenyl)prop-2-enoic acid (11 ) (1.80 mg, 35% yield) as a white solid. ESI-MS m/z calculated for [M+H]+: 594.27; found: 594.02. 1H NMR (400 MHz, c/4-methanol) δ 7.60 - 7.56 (m, 2H), 7.53 - 7.44 (m, 4H), 7.37 - 7.30 (m, 2H), 7.23 (dd, J = 8.5, 1.2 Hz, 1 H), 6.44 (d, J= 15.9 Hz, 1 H), 6.29 (s, 1 H), 4.44 (t, J = 5.7 Hz, 2H), 4.16 (t, J = 5.7 Hz, 2H), 3.11 - 3.02 (m, 1H), 2.94 - 2.85 (m, 2H), 2.50 - 2.37 (m, 4H), 2.19- 1.99 (m, 2H), 1.98- 1.77 (m, 5H), 1.57- 1.39 (m, 3H).
Synthesis of methyl (2S)-2-[[3-cvclohexyl-2-(4-fluoroDhenyl)-1-methyl-imidazo[1, 2- blDyrazole-6-carbonyllaminol-3-(4-hydroxyDhenyl)DroDanoate (12); and N-{[3-cvclohexyl-2- -fluorophenyl)- 1-methyl-1H-imidazo[ 1,2-blDyrazol-6-yllcarbonyl}-L-tyrosine (13)
Figure imgf000054_0001
Step a: To a solution of 3-cyclohexyl-2-(4-fluorophenyl)-1-methyl-imidazo[1,2-b]pyrazole-6- carboxylic acid (iv) (15 mg; 0.04 mmol) in DCM (1 mL) at ambient temperature was added methyl (2S)-2-amino-3-(4-hydroxyphenyl)propanoate hydrochloride (15 mg; 0.07 mmol), HATU (50 mg; 0.13 mmol) and DIPEA (17 mg; 0.13mmol). After 16 hours a saturated aqueous solution of NaHC03 (15 ml) was added and the aqueous layer was extracted with EtOAc (3x 15 mL). The combined organics were washed with brine (10 mL), dried (MgS04) and concentrated to leave crude methyl (2S)-2-[[3-cyclohexyl-2-(4-fluorophenyl)-1-methyl- imidazo[1 ,2-b]pyrazole-6-carbonyl]amino]-3-(4-hydroxyphenyl)propanoate (12) as a brown oil (13 mg). Used directly in next step. ESI-MS m/z calculated for [M+H]+: 519.24; found: 519.12.
Step b: To a solution of crude methyl (2S)-2-[[3-cyclohexyl-2-(4-fluorophenyl)-1-methyl- imidazo[1,2-b]pyrazole-6-carbonyl]amino]-3-(4-hydroxyphenyl)propanoate (12) (8 mg) in MeOH/THF (1:1; 2 mL) was added a 1M aqueous solution of lithium hydroxide (0.31 mmol) and the reaction was heated at 60°C for 2 hrs.1M HCI (aq) was added until ~ pH3 and the aqueous layer was extracted with EtOAc (3x 15 mL) and the combined organics were washed with brine (15 mL), dried (MgS04) and concentrated to leave a brown oil (3 mg). Purification by reverse phase LCMS: Acetonitrile/water (0.1% formic acid) afforded N-{[3- cyclohexyl-2-(4-fluorophenyl)-1-methyl-1H-imidazo[1 ,2-b]pyrazol-6-yl]carbonyl}-L-tyrosine
(13) (2.32 mg) as an amorphous white solid. ESI-MS m/z calculated for [M+H]+: 505.22; found: 505.17.
1H NMR (400 MHz, CD3CN) δ 7.49 - 7.43 (m, 2H), 7.39 (d, J = 7.7 Hz, 1H), 7.31 - 7.24 (m, 2H), 7.14-7.10 (m, 2H), 6.90 (s, 1H), 6.75-6.70 (m, 2H), 6.15 (s, 1H), 4.73 (td, J = 7.6, 5.3 Hz, 1H), 3.35 (s, 3H), 3.18 (dd, J = 14.0, 5.3 Hz, 1H), 3.10 (dd, J = 14.0, 7.5 Hz, 1H), 2.72-2.63 (m, 1H), 2.12-2.07 (m, 2H), 1.88- 1.67 (m, 5H), 1.39- 1.23 (m, 3H). Synthesis of methyl N-{[3-cvclohexyl-2-(4-fluoroDhenyl)-1-methyl-1H-imidazo[ 1, 2-blpyrazol- 6-yllcarbonyl}-L-trvDtoDhanate (14) and N-{[3-cvclohexyl-2-(4-fluorophenyl)-1-methyl-1H- imidazof 1, 2-blpyrazol-6-yllcarbonyl}-L-tryptophan (15)
Figure imgf000055_0001
Step a: To a solution of 3-cyclohexyl-2-(4-fluorophenyl)-1-methyl-imidazo[1 ,2-b]pyrazole-6- carboxylic acid (iv) (15 mg; 0.04 mmol) in DCM (1 mL) was added methyl (2S)-2-amino-3- (1H-indol-3-yl)propanoate hydrochloride (17 mg; 0.07 mmol) followed by HATU (33 mg; 0.09 mmol) and DIPEA (23 mg; 0.18 mmol). After 2 hours a saturated aqueous solution of NaHC03 (15 ml) was added and the aqueous layer was extracted with EtOAc (3x15 mL). The combined organics were washed with brine (10 mL), dried (MgS04) and concentrated to leave a brown oil (17 mg). Purification of a 4 mg portion by reverse phase LCMS:
Acetonitrile/water (0.1% formic acid) afforded methyl N-{[3-cyclohexyl-2-(4-fluorophenyl)-1- methyl-1H-imidazo[1,2-b]pyrazol-6-yl]carbonyl}-L-tryptophanate (14) (0.85 mg) as an amorphous white solid. ESI-MS m/z calculated for [M+H]+: 542.26; found: 542.19.
1H NMR (400 MHz, CD3CN) δ 9.20 (s, 1H), 7.59 (d, J = 7.9 Hz, 1H), 7.48-7.38 (m, 4H), 7.30-7.24 (m, 2H), 7.16-7.11 (m, 2H), 7.06-7.01 (m, 1H), 6.14 (s, 1H), 4.92 (dt, J = 8.0, 6.1 Hz, 1H), 3.70 (s, 3H), 3.39 (s, 1H), 3.37 (s, 1H), 3.35 (s, 3H), 2.70 - 2.61 (m, 1H), 2.07- 2.00 (m, 2H), 1.87 - 1.63 (m, 5H), 1.36 - 1.20 (m, 3H).
Step b: To a solution of crude methyl N-{[3-cyclohexyl-2-(4-fluorophenyl)-1-methyl-1H- imidazo[1,2-b]pyrazol-6-yl]carbonyl}-L-tryptophanate (14) (10 mg) in MeOH/THF (1:1; 2mL) was added a 1M aqueous solution of lithium hydroxide (0.37 mmol). The reaction was heated at 60°C for 2 hrs.1 M HCI was added until ~ pH 3 and the aqueous layer was extracted with EtOAc (3 x 30 mL) and the combined organics were washed with brine (25 mL), dried (MgS04) and concentrated to leave a brown oil (8 mg). Purification by reverse phase LCMS: Acetonitrile/water (0.1% formic acid) afforded N-{[3-cyclohexyl-2-(4- fluorophenyl)-1-methyl-1H-imidazo[1,2-b]pyrazol-6-yl]carbonyl}-L-tryptophan (15) (3.2 mg) as an amorphous white solid. ESI-MS m/z calculated for [M+H]+: 528.24; found: 528.18.1H NMR (400 MHz, CD3CN) δ 9.20 (s, 1H), 7.63 (d, J = 7.9 Hz, 1H), 7.49 - 7.37 (m, 4H), 7.29 - 7.24 (m, 2H), 7.18 (d, J = 2.4 Hz, 1H), 7.15-7.10 (m, 1H), 7.03 (ddd, J = 8.0, 7.1, 1.0 Hz, 1H), 6.14 (s, 1H), 4.87 (td, J = 7.2, 5.3 Hz, 1H), 3.47 - 3.35 (m, 2H), 3.34 (s, 3H), 2.65 (tt, J = 12.3, 3.4 Hz, 1H), 2.12-2.02 (m, 2H), 1.87-1.63 (m, 5H), 1.35-1.18 (m, 3H). Synthesis of 3-cvclohexyl-2-(4-fluoroDhenyl)-N-(2-(1H-indol-3-yl)ethyl^
Figure imgf000056_0001
To a solution of 3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-7- carboxylic acid (vii) (3 mg) in DCM (1 mL) was added tryptamine (2 mg; 0.01 mmol) followed by HATU (10 mg; 0.03 mmol) and DIPEA (3.4 mg; 0.03 mmol). After 2 hours a saturated aqueous solution of NaHC03 (15 ml) was added and the aqueous layer was extracted with EtOAc (3 x 15 mL). The combined organics were washed with brine (10 mL), dried (MgS04) and concentrated to leave a brown oil (8 mg). Purification by reverse phase LCMS: Acetonitrile/water (0.1 % formic acid) afforded 3-cyclohexyl-2-(4-fluorophenyl)-N-[2- (1 H-indol-3-yl)ethyl]-1 -methyl-1 H-imidazo[1 ,2-b]pyrazole-7-carboxamide (16) (1 .98 mg) as an amorphous white solid.
ESI-MS m/z calculated for [M+H]+: 484.25; found: 484.16. 1 H NMR (400 MHz, CD3CN) δ 9.08 (s, 1 H), 7.77 (s, 1 H), 7.63 (d, J = 7.6 Hz, 1 H), 7.49 - 7.43 (m, 2H), 7.39 (d, J = 8.1 Hz, 1 H), 7.30 - 7.23 (m, 2H), 7.16 - 7.10 (m, 2H), 7.07 - 7.02 (m, 1 H), 6.61 - 6.57 (m, 1 H), 3.76 (s, 3H), 3.64 - 3.53 (m, 2H), 2.99 (t, J = 7.1 Hz, 2H), 2.65 (tt, J = 12.2, 3.4 Hz, 1 H), 2.08 - 1 .97 (m, 2H), 1.82 - 1.63 (m, 5H), 1.35 - 1 .18 (m, 3H).
Biological Data
The in vitro and in vivo antiviral activity of the compounds of the invention may be determined using the following protocols.
HCV Polymerase Inhibition Assay
HCV polymerase reactions were carried out using a modified method of Howe et ai , Antimicrobial Agents and Chemotherapy 2004 48(12): 4813-4821. Reactions contained 50 nM NS5bA21 , 20 mM Tris-HCI pH 7.5, 5 rtiM MgCI2, 5 mM MnCI2, 3 mM DTT, 0.05% BSA, 0.2 U/pL RNasin, 10 μg mL Poly(rC) template, GTP (at Km) and 0.05 pCi/pL 33P-GTP in a total reaction volume of 50 pL. Compounds were tested in a three fold dilutions series for example starting from 50 μΜ. Reactions were initiated with the addition of GTP and terminated after 1 hour with 50 pL ice cold 0.2 M EDTA. Terminated reactions were transferred to DEAE 96-well filter plates, unincorporated nucleotides washed from the filters and 50 pL scintillation fluid added prior to reading on a scintillation counter. The compound concentration that reduced 33P-GTP incorporation 50% (IC50) was calculated using nonlinear regression. Table 1 provides IC50 values for selected compounds of the invention. The majority of these compounds were determined to have IC50 values less than 10 μΜ.
Table 1 : IC50 values for selected compounds of the invention
Figure imgf000057_0001
, enzoxazep ne- -car oxy c ac ethyl 13-cyclohexyl-3-hydroxy-6J-dihydroimidazo[2', 1 ':2,3]imidazo[1 ,5- 10.9 d][1 ,4]benzoxazepine-10-carboxylate
ethyl 13-cyclohexyl-3-{[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1- 0.192 yl)benzyl]oxy}-6J-dihydroimidazo[2', 1 ':2,3]imidazo[1 ,5- dlf 1 ,4lbenzoxazepine-10-carboxylate
13-cyclohexyl-3-{[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1 -yl)benzyl]oxy}- 0.021 6J-dihydroimidazo[2', 1 ':2,3]imidazo[1 ,5-d][1 ,4]berizoxazepine-10- carboxylic acid
HCV replicon assays
A genotype 1 b subgenomic replicon cell line based on Blight et al., Science 2000 290: 1972-1974, modified to express a Renilla luciferase reporter gene was used to assess antiviral activity of test compounds. Cell cultures were maintained in a sub-confluent state in DMEM with glutamine, 10% heat-inactivated foetal bovine serum and Geneticin.
For assay, cells were seeded at a concentration of 7000 cells/well into 96 well tissue culture trays in culture media lacking Geneticin. Compounds were tested in a three fold dilutions series for example starting from 50 μΜ. After 72 hours incubation at 37°C and 5% C02, Renilla luciferase activity was quantified. The compound concentration that reduced luciferase activity by 50% (EC50) was calculated using non-linear regression.
Table 2 provides EC50 values for selected carboxylic esters of the invention.
Although these compounds did not demonstrate significant activity in the HCV polymerase enzyme assay (IC50 > 50 μΜ) they were shown to be active in the cell based HCV replicon assay. Without wishing to be bound by theory, these results may indicate that compounds of formula (I) having carboxylic esters are acting as prodrug moieties with respect to their corresponding carboxylic acid analogues. The corresponding carboxylic acid analogues were shown to be active in the HCV polymerase enzyme assay (see results provided above in Table 1 ).
Table 2: EC50 and IC50 values for selected compounds of the invention
Figure imgf000058_0001
Cytotoxicity analysis
Cytotoxicity of compounds against genotype 1 b subgenomic replicon cells were assessed using the MTT assay (Watanabe et al. , Journal of Virological Methods 1994 48:257-265). Plates were prepared as described for the HCV eplicon assay and cytotoxicity of the test article was evaluated after three days. MTT was added to assay plates followed by three hour incubation at 37°C. Wells were aspirated to dryness and the formazan dye dissolved by the addition of 100% isopropanol. Absorbencies were read in a spectrophotometer at two wavelengths (540 nm and 690 nm). The compound concentration that reduced cell viability by 50% (CC50) is calculated using non-linear regression.
Cross-genotype HCV activity
HCV replicon assays and HCV polymerase assays for genotypes 1 b, 1 a, 2a and 3a to determine the cross-genotype HCV activity of a compound are conducted using essentially the same methods as described above.
Combination studies in Replicon Cells
A genotype 1 b (Con 1 ) subgenomic replicon cell line based on Blight et al. , Science 2000 290: 1972-1974, modified to express a Renilla luciferase reporter gene is used to assess synergy of test compounds. Cell cultures were maintained in a sub-confluent state in DMEM with glutamine, 10% heat-inactivated foetal bovine serum (FBS) and G418 (Geneticin®).
For assay, cells were seeded at a density of 7000 cells/well into 96 well tissue culture trays in culture media lacking G418. The compound concentration that reduced luciferase activity by 50% (EC50) is determined independently for each compound and used to set the range of concentrations for the combination experiments. Each compound is tested singly and in combination using 3-fold serial dilutions above and below the EC50. The ratio of 2 compounds when tested remains fixed across the titration range. Cytotoxicity of individual compounds is assessed independently and the titration range is below the compound concentration that reduces cell viability by 50% (CC50). After 72 hours incubation at 37°C and 5% C02, Renilla luciferase activity is quantified via the Promega Renilla Luciferase Assay System.
Results are analysed and levels of synergy assessed via generation of 3D synergy plots using MacSynergy™ II (Prichard, M. N., K. R. Aseltine, and C. Shipman, Jr. 1993. MacSynergy II. Version 1.0. User's manual. University of Michigan, Ann Arbor.). This method is therefore useful in determining synergistic combinations of an inhibitor of the invention and an HCV inhibitor targeting a different viral protein, or with a different mechanism of inhibiting the NS5B polymerase (for example an NS5A inhibitor, a nucleoside or nucleotide NS5B inhibitor or a NS3/4A protease inhibitor)
Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.
The reference in this specification to any prior publication, or information derived from it, or to any matter which is known, is not, and should not be taken as an acknowledgement or admission or any form of suggestion that that prior publication, or information derived from it, or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates.

Claims

1 . A compound of formula (I), salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof:
Figure imgf000061_0001
(I)
wherein
Ri is H or Ra or R1 together with R2 and the atoms to which they are attached form an optionally substituted 6-8 membered carbocyclic ring which may be optionally interrupted by one or more heteroatoms independently selected from O, N and S;
R2 is H or Rb or R2 together with Ri joins to form an optionally substituted 6-8 membered carbocyclic ring which may be optionally interrupted by one or more heteroatoms independently selected from O, N and S;
R3 is an optionally substituted C3-8cycloalkyl group;
XL X2 and X3 are each independently selected from N, CH, C-RC and C-A-R4 provided that at least one of X2 and X3 is C-A-R4;
A is a covalent bond, -CONH-C(R5)(R6)-B- or -CONCi-aalkyl-CiRsXReJ-B-;
R4 is selected from H, C02H, optionally substituted C02Ci_6alkyl, optionally substituted aryl, optionally substituted arylCi_6alkyl, optionally substituted arylC2.6alkenyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted heterocyclylC^alkyl, optionally substituted heteroarylC^alkyl, optionally substituted heterocyclylC2.6alkenyl and optionally substituted heteroarylC2.6alkenyl;
R5 and R6 are each independently selected from H, optionally substituted C^alkyl, optionally substituted aryl, optionally substituted C^alkylaryl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted C1.6alkylheterocyclyl and optionally substituted C^alkylheteroaryl or R5 and R6 together with the carbon atom to which they are attached form an optionally substituted C3.6cycloalkyl group;
B is a covalent bond or -CONH-R7- where R7 is selected from optionally substituted aryl, optionally substituted arylCi-6alkyl, optionally substituted arylC2.6alkenyl, optionally substituted heterocyclyl, optionally substituted heteroaryl, optionally substituted
heterocyclylC^alkyl, optionally substituted heteroarylC^alkyl, optionally substituted heterocyclylC2.6alkenyl and optionally substituted heteroarylC2.6alkenyl; Z1 is C or N;
Z2, Z3, Z4 and Z5 are each independently selected from N, CH and C-Rd;
Ra, Rb, Rc and Rd are each independently selected from C^alkyl, d ealkenyl, C2. 6alkynyl, C3-8cycloalkyl, aryl, Ci_6alkylaryl, d ealkoxyaryl, heterocyclyl, heteroaryl, halo, d- 6alkylhalo, OH, d-ealkylOH, d.6alkoxy, d-ealkoxyhalo, oxy (C=0), C02R8, OCOR8, C(=0)R8, CN, N02, N(R8)2, NR8COR8, CONR8, SR8, thio (=S), S02d-6alkyl, S02NR8R8, NR8S02R8;
wherein each R8 is independently selected from H and C1.6alkyl and each alkyl, alkenyl, alkynyl, cycloalkyi, aryl, heterocyclyl and heteroaryl in the definition of Ra, Rb, Rc and Rd may be optionally substituted;
and further wherein each heterocyclyl or heteroaryl has 1 , 2, 3 or 4 heteroatoms independently selected from O, S and N.
2. The compound according to claim 1 wherein C-A-R4 is not C-H.
3. The compound according to claim 1 or claim 2 wherein R1 is H or Chalky!.
4. The compound according to claim 1 or claim 2 wherein R1 together with R2 joins to form an optionally substituted 6-8 membered carbocyclic ring which may be optionally interrupted by one or more heteroatoms independently selected from O, N and S.
5. The compound according to any one of claims 1 to 4 wherein R3 is an optionally substituted C3-6cycloalkyl group.
6. The compound according to any one of claims 1 to 5 wherein R4 is selected from H, C02H and optionally substituted C02d-6alkyl.
7. The compound according to any one of claims 1 to 6 wherein A is a covalent bond
8. The compound according to any one of claims 1 to 6 wherein A is -CONH- C(R5)(R6)-B- wherein R5 and R6 are each independently selected from H, Chalky!, d-
6alkylaryl, d_6alkylheterocyclyl and C1.6alkylheteroaryl or R5 and R6 together with the carbon atom to which they are attached form a cyclobutyl group.
9. The compound according to any one of claims 1 to 8 wherein B is a covalent bond
10. The compound according to any one of claims 1 to 8 wherein B is -CONHR7- wherein R7 is selected from aryl, aryld_6alkyl and arylC2.6alkenyl.
1 1. The compound according to claim 1 of formula (la)
Figure imgf000063_0001
(la)
wherein Z3 is CH or C-Rd and R2, R3, Rd, Χι , X2 and X3 are defined according to any one of claims 1 to 10;
and salts, /V-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
12. The compound of formula (la) according to claim 1 1 selected from the group of compounds of formula (la-i) to (la-vi):
Figure imgf000063_0002
(la-iv) (la-v) (la-vi)
wherein Ri , R2, R3, ∑3, Xi, X2 and X3 are defined according to any one of claims 1 to 10; and salts, /V-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof
13. The compound of formula (la-i) and (la-vi) according to claim 12 selected from the group consisting of compounds of formulae:
Figure imgf000064_0001
wherein A, R2, R3, R4, Z^ X^ , X2 and X3 are defined according to any one of claims 1 to 10 provided that A-R4 is not H;
and salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
14. A compound according to claim 1 selected from the group consisting of methyl 3-cyclohexyl-2-(4-fluorophenyl)-1 H-imidazo[1 ,2-b]pyrazole-6-carboxylate (i);
3-cyclohexyl-2-(4-fluorophenyl)-1 H-imidazo[1 ,2-b]pyrazole-6-carboxylic acid (ii); methyl 3- cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-6-carboxylate (iii);
3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazole-6-carboxylic acid (iv); ethyl 3-cyclohexyl-2-(4-fluorophenyl)-1 H-imidazo[1 ,2-b]pyrazole-7-carboxylate (v); ethyl 3- cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-7-carboxylate (vi);
3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazole-7-carboxylic acid (vii); 3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazole (viii);
ethyl 3-cyclohexyl-1 -methyl-2-phenyl-1 H-imidazo[1 ,2-b]pyrazole-7-carboxylate (ix);
3-cyclohexyl-1 -methyl-2-phenyl-1 H-imidazo[1 ,2-b]pyrazole-7-carboxylic acid (x);
methyl 13-cyclohexyl-6,7-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10- carboxylate (xi);
13-cyclohexyl-6,7-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10-carboxylic acid (xii);
methyl 3-(benzyloxy)-13-cyclohexyl-6,7-dihydropyrazolo[5', 1 ':2,3] imidazo[1 ,5- d][1 ,4]benzoxazepine-10-carboxylate (xiii);
methyl 13-cyclohexyl-3-hydroxy-6,7-dihydropyrazolo[5',1 ':2,3] imidazo[1 ,5- d][1 ,4]benzoxazepine-10-carboxylate (xiv);
3-(benzyloxy)-13-cyclohexyl-6,7-dihydropyrazolo[5',1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine- 10-carboxylic acid (xv);
13-cyclohexyl-3-hydroxy-6J-dihydropyrazolo[5',1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10- carboxylic acid (xvi); ethyl 3-(benzyloxy)-13-cyclohexyl-6J-dihydroimidazo[2', 1 ':2,3]imidazo [1 ,5- d][1 ,4]benzoxazepine-10-carboxylate (xvii);
methyl 13-cyclohexyl-3-{[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1-yl)benzyl]oxy}-6J- dihydropyrazolo[5',1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10-carboxylate (xviii);
13-cyclohexyl-3-{[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1 -yl)benzyl]oxy}-6,7- dihydropyrazolo[5',1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10-carboxylic acid (xix);
3-(benzyloxy)-13-cyclohexyl-6J-dihydroimidazo[2', 1 ':2,3]imidazo [1 ,5-d][1 ,4]benzoxazepine- 10-carboxylic acid (xx);
ethyl 13-cyclohexyl-3-hydroxy-6J-dihydroimidazo[2', 1 ':2,3]imidazo[1 ,5- d][1 ,4]benzoxazepine-10-carboxylate (xxi);
ethyl 13-cyclohexyl-3-{[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1-yl)benzyl]oxy}-6J- dihydroimidazo[2', 1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10-carboxylate (xxii);
13-cyclohexyl-3-{[2-(morpholin-4-yl)-5-(2-oxopyrrolidin-1 -yl)benzyl]oxy}-6,7- dihydroimidazo[2', 1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepine-10-carboxylic acid (xxiii);
ethyl (2E)-3-[4-({[1 -({[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazol-6- yl]carbonyl}amino)cyclobutyl]carbonyl} amino)phenyl]prop-2-enoate (1 );
(E)-3-[4-[[1 -[[3-cyclohexyl-2-(4-fluorophenyl)-1-methyl-imidazo[1 ,2-b]pyrazole-6- carbonyl]amino]cyclobutanecarbonyl] amino]phenyl]prop-2-enoic acid (2);
methyl (E)-3-[4-[[1-[[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-6- carbonyl]amino] cyclobutanecarbonyl]amino]phenyl]prop-2-enoate (3);
ethyl (E)-3-[4-[[1 -[[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-7- carbonyl]amino]cyclobutanecarbonyl] amino]phenyl]prop-2-enoate (4); (2E)-3-[4-({[1 -({[3- cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazol-7- yl]carbonyl}amino)cyclobutyl]carbonyl}amino)phenyl]prop-2-enoic acid (5);
ethyl (2E)-3-(4-{[(1 -{[(3-cyclohexyl-1 -methyl-2-phenyl-1 H-imidazo[1 ,2-b]pyrazol-7- yl)carbonyl]amino}cyclobutyl) carbonyl]amino}phenyl)prop-2-enoate (6);
(2E)-3-(4-{[(1-{[(3-cyclohexyl-1 -methyl-2-phenyl-1 H-imidazo[1 ,2-b]pyrazol-7- yl)carbonyl]amino}cyclobutyl) carbonyl]amino}phenyl)prop-2-enoic acid (7);
ethyl 1 -{[(13-cyclohexyl-6J-dihydropyrazolo[5', 1':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepin-10- yl)carbonyl]amino}cyclobutanecarboxylate (8);
1 -{[(13-cyclohexyl-6,7-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5-d][1 ,4]benzoxazepin-10- yl)carbonyl]amino}cyclobutanecarboxylic acid (9);
ethyl (2E)-3-(4-{[(1 -{[(13-cyclohexyl-6J-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5- d][1 ,4]benzoxazepin-10-yl)carbonyl]amino}cyclobutyl)carbonyl]amino}phenyl)prop-2-enoate (10); (2E)-3-(4-{[(1-{[(13-cyclohexyl-6J-dihydropyrazolo[5', 1 ':2,3]imidazo[1 ,5- d][1 ,4]benzoxazepin-10-yl)carbonyl]amino}cyclobutyl)carbonyl]amino}phenyl)prop-2-enoic acid (11 );
methyl (2S)-2-[[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-imidazo[1 ,2-b]pyrazole-6- carbonyl]amino]-3-(4-hydroxyphenyl)propanoate (12);
N-{[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazol-6-yl]carbonyl}-L- tyrosine (13);
methyl N-{[3-cyclohexyl-2-(4-fluorophenyl)-1-methyl-1 H-imidazo[1 ,2-b]pyrazol-6-yl]carbonyl}- L-tryptophanate (14);
N-{[3-cyclohexyl-2-(4-fluorophenyl)-1 -methyl-1 H-imidazo[1 ,2-b]pyrazol-6-yl]carbonyl}-L- tryptophan (15); and
3-cyclohexyl-2-(4-fluorophenyl)-N-[2-(1 H-indol-3-yl)ethyl]-1 -methyl-1 H-imidazo[1 ,2- b]pyrazole-7-carboxamide (16);
and salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
15. A process for producing a compound as defined according to any one of claims 1 to 14 wherein A is -CONH-C( 5)( 6)-B- and B is -CONH-R7- comprising the step of reacting a compound of formula (II) with NH2R7 under amidation conditions
Figure imgf000066_0001
(II)
wherein ΧΊ , X'2 and X'3 are each independently selected from N, CH, C-Rc, C-A-R'4 and at least one of ΧΊ , X'2 and X'3 is C-A-R'4 wherein R'4 is C02H or C02Ci_6alkyl; and
Ri , R, R3, R5, R6, R7, C-Rc, Zi , Z2, Z3, Z4 and Z5 are defined according to any one of claims 1 to 10.
16. A pharmaceutical agent comprising the compound as defined according to any one of claims 1 to 14 and pharmaceutically acceptable salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
17. A HCV viral polymerase inhibitor comprising the compound as defined according to any one of claims 1 to 14 and pharmaceutically acceptable salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
18. A pharmaceutical composition comprising the compound as defined according to any one of claims 1 to 14 and pharmaceutically acceptable salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof and a pharmaceutically acceptable carrier and optionally one or more antiviral agents.
19. A method for the treatment of a Flaviviridae viral infection which comprises administering an effective amount of the compound as defined according to any one of claims 1 to 14 and pharmaceutically acceptable salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof, the pharmaceutical agent according to claim 16 or the pharmaceutical composition according to claim 18 to a subject diagnosed with, suffering from or at risk of developing said viral infection.
20. The method of treatment according to claim 19 wherein the Flaviviridae viral infection is a Hepatitis C virus (HCV) infection.
21. A method of inhibiting the RNA-dependent RNA polymerase activity of the enzyme NS5B, encoded by HCV, comprising exposing the enzyme NS5B to an effective amount of the compound as defined according to any one of claims 1 to 14 and
pharmaceutically acceptable salts, N-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
22. A method of inhibiting HCV replication comprising exposing a cell infected with HCV to an effective amount of the compound as defined according to any one of claims 1 to 14 and pharmaceutically acceptable salts, W-oxides, solvates, hydrates, racemates, enantiomers and isomers thereof.
PCT/AU2012/001091 2011-09-16 2012-09-13 Compounds for the treatment of hcv WO2013036994A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161535666P 2011-09-16 2011-09-16
US61/535,666 2011-09-16

Publications (1)

Publication Number Publication Date
WO2013036994A1 true WO2013036994A1 (en) 2013-03-21

Family

ID=47882443

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2012/001091 WO2013036994A1 (en) 2011-09-16 2012-09-13 Compounds for the treatment of hcv

Country Status (1)

Country Link
WO (1) WO2013036994A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9717731B2 (en) 2012-11-02 2017-08-01 Pharmacyclics Llc TEC family kinase inhibitor adjuvant therapy
US9814721B2 (en) 2010-06-03 2017-11-14 Pharmacyclics Llc Use of inhibitors of bruton'S tyrosine kinase (BTK)
US9885086B2 (en) 2014-03-20 2018-02-06 Pharmacyclics Llc Phospholipase C gamma 2 and resistance associated mutations
US10316040B2 (en) 2015-10-16 2019-06-11 Eisai R&D Management Co., Ltd. EP4 antagonists
US10954567B2 (en) 2012-07-24 2021-03-23 Pharmacyclics Llc Mutations associated with resistance to inhibitors of Bruton's Tyrosine Kinase (BTK)
CN113039186A (en) * 2018-11-02 2021-06-25 艾库里斯有限及两合公司 Urea 6, 7-dihydro-4H-pyrazolo [1,5-a ] pyrazine active agents against hepatitis b virus HBV

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008141385A1 (en) * 2007-05-21 2008-11-27 Biota Scientific Management Pty Ltd Viral polymerase inhibitors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008141385A1 (en) * 2007-05-21 2008-11-27 Biota Scientific Management Pty Ltd Viral polymerase inhibitors

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10653696B2 (en) 2010-06-03 2020-05-19 Pharmacyclics Llc Use of inhibitors of bruton's tyrosine kinase (BTK)
US10004746B2 (en) 2010-06-03 2018-06-26 Pharmacyclics Llc Use of inhibitors of Bruton's tyrosine kinase (Btk)
US10016435B2 (en) 2010-06-03 2018-07-10 Pharmacyclics Llc Use of inhibitors of Bruton's tyrosine kinase (Btk)
US9814721B2 (en) 2010-06-03 2017-11-14 Pharmacyclics Llc Use of inhibitors of bruton'S tyrosine kinase (BTK)
US10478439B2 (en) 2010-06-03 2019-11-19 Pharmacyclics Llc Use of inhibitors of bruton's tyrosine kinase (Btk)
US10004745B2 (en) 2010-06-03 2018-06-26 Pharmacyclics Llc Use of inhibitors of Bruton'S tyrosine kinase (Btk)
US10751342B2 (en) 2010-06-03 2020-08-25 Pharmacyclics Llc Use of inhibitors of Bruton's tyrosine kinase (Btk)
US11672803B2 (en) 2010-06-03 2023-06-13 Pharmacyclics Llc Use of inhibitors of Brutons tyrosine kinase (Btk)
US10954567B2 (en) 2012-07-24 2021-03-23 Pharmacyclics Llc Mutations associated with resistance to inhibitors of Bruton's Tyrosine Kinase (BTK)
US9717731B2 (en) 2012-11-02 2017-08-01 Pharmacyclics Llc TEC family kinase inhibitor adjuvant therapy
US9885086B2 (en) 2014-03-20 2018-02-06 Pharmacyclics Llc Phospholipase C gamma 2 and resistance associated mutations
US10316040B2 (en) 2015-10-16 2019-06-11 Eisai R&D Management Co., Ltd. EP4 antagonists
US11434246B2 (en) 2015-10-16 2022-09-06 Eisai R&D Management Co., Ltd. EP4 antagonists
US10941148B2 (en) 2015-10-16 2021-03-09 Eisai R&D Management Co., Ltd. EP4 antagonists
CN113039186A (en) * 2018-11-02 2021-06-25 艾库里斯有限及两合公司 Urea 6, 7-dihydro-4H-pyrazolo [1,5-a ] pyrazine active agents against hepatitis b virus HBV
JP2022512870A (en) * 2018-11-02 2022-02-07 アイクリス ゲゼルシャフト ミット ベシュレンクテル ハフツング ウント コンパニー コマンディトゲゼルシャフト Novel urea 6,7-dihydro-4H-pyrazolo [1,5-A] pyrazine active against hepatitis B virus (HBV)

Similar Documents

Publication Publication Date Title
CN105001225B (en) The heterocyclic modulators of lipid synthesis
WO2011153588A1 (en) Viral polymerase inhibitors
EP3860989B1 (en) Fused pyrrolines which act as ubiquitin-specific protease 30 (usp30) inhibitors
WO2013036994A1 (en) Compounds for the treatment of hcv
AU2014234906B2 (en) Cycloalkyl nitrile pyrazolo pyridones as Janus kinase inhibitors
AU2015339196A1 (en) Substituted tetrahydropyrans and method of use
BRPI0918174A2 (en) compound that has activity against the hepatitis c virus (hcv), its composition and its use
WO2008141385A1 (en) Viral polymerase inhibitors
WO2016174079A1 (en) Rsv antiviral pyrazolo- and triazolo-pyrimidine compounds
CN110177551B (en) Heterocyclic modulators of lipid synthesis
EP4001279A1 (en) Novel spiropyrrolidine derived antiviral agents
EP3426243A1 (en) 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
US8546389B2 (en) Viral polymerase inhibitors
US8785478B2 (en) Viral polymerase inhibitors
MX2010013630A (en) Tricyclic indole derivatives and methods of use thereof.
KR20230142706A (en) oxytocin receptor modulator
CN106008552B (en) Benzo [ e ] pyrazolo [1,5-c ] [1,3] oxazine compound and application thereof
WO2013075173A1 (en) Compounds for the treatment of hcv
WO2023003610A1 (en) Novel spiropyrrolidine derived antiviral agents
AU2021290652A1 (en) Amido compounds
CN105985355B (en) Fused tricyclic hepatitis virus inhibitor and application thereof

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: 12832384

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: 12832384

Country of ref document: EP

Kind code of ref document: A1