WO2004039833A1 - Composition for the treatment of infection by flaviviridae viruses - Google Patents

Composition for the treatment of infection by flaviviridae viruses Download PDF

Info

Publication number
WO2004039833A1
WO2004039833A1 PCT/CA2003/001634 CA0301634W WO2004039833A1 WO 2004039833 A1 WO2004039833 A1 WO 2004039833A1 CA 0301634 W CA0301634 W CA 0301634W WO 2004039833 A1 WO2004039833 A1 WO 2004039833A1
Authority
WO
WIPO (PCT)
Prior art keywords
virus
flaviviridae
hcv
alkyl
formula
Prior art date
Application number
PCT/CA2003/001634
Other languages
French (fr)
Inventor
Daniel Lamarre
Lisette LAGACÉ
Original Assignee
Boehringer Ingelheim International Gmbh
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 Boehringer Ingelheim International Gmbh filed Critical Boehringer Ingelheim International Gmbh
Priority to EP03809665A priority Critical patent/EP1558633A1/en
Priority to EA200500610A priority patent/EA200500610A1/en
Priority to CA002498642A priority patent/CA2498642A1/en
Priority to BR0315781-4A priority patent/BR0315781A/en
Priority to JP2005501790A priority patent/JP2006517196A/en
Priority to MXPA05004604A priority patent/MXPA05004604A/en
Priority to AU2003275852A priority patent/AU2003275852A1/en
Publication of WO2004039833A1 publication Critical patent/WO2004039833A1/en
Priority to NO20052580A priority patent/NO20052580L/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/162Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from virus
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/05Dipeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/06Tripeptides
    • 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
    • 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
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/24011Flaviviridae
    • C12N2770/24211Hepacivirus, e.g. hepatitis C virus, hepatitis G virus
    • C12N2770/24222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • the present invention relates to compounds, compositions, use and method for the treatment of a Flaviviridae viral infection in a mammal. More particularly, the present invention relates to the use and a method of treatment comprising administration of a compound selected from a macrocyclic peptide family of compounds.
  • the Flaviviridae family of viruses are enveloped positive-stranded RNA viruses comprising a number of human pathogenic viruses such as viruses of the hepacivirus genus, including Hepatitis C, viruses of the flavivirus genus, including the Dengue Fever viruses, encephalitis viruses, West Nile viruses and Yellow Fever viruses, and viruses of the pestivirus genus, including the bovine viral diarrhea virus and border disease virus, both of which are animal pathogens.
  • the most newly discovered viruses, hepatitis G virus (HGV) and hepatitis GB virus (GBV-A, B, C) are also provisionally considered to be members of the Flaviviridae family belonging to a distinct genus.
  • Dengue viruses members of the family of Flaviviridae are transmitted by mosquitos. There are four serotypes that cause widespread human diseases, one of which causes dengue hemorrhagic fever, and about 40% of the population living in tropical and subtropical regions of the world is at risk for infection. Of the 1 million cases of hemorrhagic fever cases per year, about 5% are fatal. There is currently no effective vaccine or antiviral drug to protect against dengue diseases.
  • Pestiviruses such as bovine diarrhea virus (BVDV), classical swine fever virus (CSFV) and border disease virus (BDV) comprise a group of economically important animal pathogens affecting cattle, pigs and sheep. These positive-sense RNA viruses are classified as a separate genus in the family Flaviviridae.
  • BVDV bovine diarrhea virus
  • CSFV classical swine fever virus
  • BDV border disease virus
  • GB-viruses have been classified as members of the Flaviviridae family, but have not yet been assigned to a particular genus based on the analysis of genomic sequences. These RNA viruses, in addition to infecting humans, can cause acute resolving hepatitis in experimentally infected tamarins.
  • Viruses within the Flaviviridae family possess a number of similarities despite a relatively low overall sequence homology among its members.
  • the genome of these viruses is a small single-stranded RNA ( « 10 kilobases in length) having a single open reading frame (ORF).
  • the ORF encodes a polyprotein of about 3000 amino acids that contains structural proteins at its 5' end and non-structural (NS) proteins at its 3' end.
  • the polyprotein is proteolytically processed by both viral and host- encoded proteases into mature polypeptides, which for HCV, are as follows: NH 2 - ⁇ C- E1-E2-P7-NS2-NS3-NS4A-NS4B-NS5A-NS5B ⁇ -COOH.
  • the core protein or nucleocapsid (C) and the two envelope glycoproteins (E1 and E2) represent the constitutive structural proteins of the virions. These structural proteins are followed by the nonstructural (NS) proteins, which at least some are thought to be essential for viral RNA replication.
  • NS nonstructural
  • NS3 protein contains sequences with similarity to the serine protease and the nucleoside triphosphate-binding helicase of pestiviruses and flaviviruses. Analysis of sequence alignments had predicted the existence of a trypsin-like serine protease domain within the N-terminal region of flavi-, pesti-, hepaci- and GB- viruses. Sequence similarities between NS3 proteolytic domains of Flaviviridae viruses are well-established (Ryan MD et al. 1998).
  • the HCV NS3 protease domain shares a sequence similarity of about 77-90% among HCV genotypes and a sequence similarity of about 25-50% with other members of the Flaviviridae family.
  • the available atomic co-ordinates of the various crystallized HCV and Dengue NS3 proteases show an overall architecture that is characteristic of the trypsin-like fold (Kim et al. 1996, Love et al. 1996; Murthy et al. 1999).
  • Many studies have now firmly established that the N- terminal portion of the NS3 region encodes a serine protease that has a very specific and pivotal role in viral polyprotein processing within Flaviviridae.
  • polyprotein processing shows a requirement for the downstream NS4A protein.
  • the NS4A protein acts as a cofactor that enhances the NS3 protease cleavage efficiency (Lin et al., 1995; Kim et al., 1996; Stein uler et al., 1996).
  • this requirement for enhancing the NS3 protease activity is provided by the upstream NS2B protein.
  • the genomic organization and structure of GBV-B and HCV are similar despite the fact that the sequence homology between the polyprotein sequences of GBV-B and HCV is about 25 to 30%.
  • Flaviviridae family of viruses Given apparent similarities of viruses within the Flaviviridae family of viruses, it would be desirable to develop therapeutic agents effective against Flaviviridae viruses, and more particularly, effective against the pathogenic members of the Flaviviridae family.
  • an anti- Flaviviridae virus composition comprising a pharmaceutically acceptable carrier in combination with a compound of Formula (I):
  • A is selected from: C ⁇ to C 6 alkyl and C 3 to C 6 cycloalkyl; and B is selected from: phenyl or thiazolyl, both of which optionally substituted with a group selected from NH(R 1 ) and NH(CO)R 1 , wherein R 1 is C 1 to C 6 alkyl; R is OH or a sulfonamide derivative, or a pharmaceutically acceptable salt thereof.
  • the present invention provides a method for treating a mammal infected with a virus of the Flaviviridae family comprising administering to the infected mammal a pharmaceutical composition comprising a pharmaceutically acceptable carrier in combination with a therapeutically effective amount of a compound of Formula (I) as defined above.
  • the present invention provides a method of treating a mammal infected with a virus of the Flaviviridae family wherein a pharmaceutical composition comprising a pharmaceutically acceptable carrier in combination with a therapeutically effective amount of a compound of Formula (I) as defined above is co-administered with at least one additional agent selected from: an antiviral agent, an immunomodulatory agent, an HCV inhibitor, an HIV inhibitor, an HAV inhibitor and an HBV inhibitor; to the infected mammal.
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier in combination with a therapeutically effective amount of a compound of Formula (I) as defined above is co-administered with at least one additional agent selected from: an antiviral agent, an immunomodulatory agent, an HCV inhibitor, an HIV inhibitor, an HAV inhibitor and an HBV inhibitor; to the infected mammal.
  • the present invention provides a pharmaceutical composition for treating or preventing an infection of a mammal caused by a virus of the Flaviviridae family comprising a pharmaceutically acceptable carrier in combination with a therapeutically effective amount of a compound of Formula (I) and at least one additional agent selected from: an antiviral agent, an immunomodulatory agent, an HCV inhibitor, an HIV inhibitor, an HAV inhibitor and an HBV inhibitor.
  • a compound of Formula (I) as defined above for the manufacture of a medicament for the treatment of Flaviviridae viral infection.
  • an article of manufacture comprising packaging material contained within which is a composition effective to inhibit a virus of the Flaviviridae family and the packaging material comprises a label which indicates that the composition can be used to treat infection by a virus of the Flaviviridae family and, wherein said composition comprises a compound of Formula (I) as defined above.
  • the macrocyclic peptide family of compounds generally represented by Formula (I) as set out above are believed to interact at a conserved substrate binding site in the NS3 protease domain. Crystal structure studies confirm that the present macrocyclic compounds interact at the substrate-binding site within the HCV NS3 domain, a region that is functionally conserved among Flaviviridae viruses.
  • Tables 4-8 provide a sequence similarity comparison between members of the Flaviviridae family.
  • Figure 1 provides a comparison of polyproteins of viruses belonging to the Flaviviridae family (taken from: Ryan etal., 1998, J. Gen. Virology 79, 947-959);
  • Figure 2 is a sequence comparison of the NS3 protease domain between HCV genotypes and subtypes
  • Figures 3A-B are the IC 50 curves of a macrocyclic peptide compound according to
  • Figures 4A-B are the Dixon and Cornish-Bowden plots of the macrocyclic peptide of
  • Figures 5A-B are the Dixon and Cornish-Bowden plots of the macrocyclic peptide of
  • Figure 6 illustrates the inhibition of GBV-B replication by macrocyclic peptides according to Formula (I) in tamarin hepatocytes in culture;
  • Figure 7 graphically illustrates dose-dependent inhibition of GBV-B replication by the macrocyclic peptide III of Figure 6;
  • Figure 8 illustrates the 3-dimensional crystal structure of the HCV NS3-NS4A peptide structure complexed with a macrocyclic peptide according to Formula (I).
  • the term "Flaviviridae” as it is used herein to designate a viral family is meant to encompass viruses of the hepacivirus genus, such as Hepatitis C, viruses of the flavivirus genus, such as the Dengue Fever viruses, Encephalitis viruses, West Nile viruses and Yellow Fever viruses, and viruses of the pestivirus genus, such as the bovine viral diarrhea virus and border disease virus.
  • viruses of the hepacivirus genus such as Hepatitis C
  • viruses of the flavivirus genus such as the Dengue Fever viruses, Encephalitis viruses, West Nile viruses and Yellow Fever viruses
  • viruses of the pestivirus genus such as the bovine viral diarrhea virus and border disease virus.
  • Hepatitis G virus (HGV) and Hepatitis GB virus are also included in this viral family although the genus of these viruses has not yet been determined.
  • Flaviviridae family including for example, HCV 1a, HCV1b, HCV 2a-c, HCV 3a-b, HCV 4a, HCV 5 and HCV 6a,h,d & k, as well as GBV-A, B & C.
  • mammal as it is used herein is meant to encompass humans, as well as non-human mammals which are susceptible to infection by a Flaviviridae virus including domestic animals, such as cows, pigs, horses, dogs and cats, and sheep.
  • C 1-6 alkyl or “C C 6 " as used herein, either alone or in combination with another substituent, means acyclic, straight or branched chain alkyl substituents containing from one to six carbon atoms and includes, for example, methyl, ethyl, propyl, isopropyl, butyl, tert-bu ⁇ yl, hexyl, -methylethyl, 1-methylpropyl, 2-methylpropyl, 1 ,1-dimethylethyl.
  • C 3-6 cycloalkyl as used herein, either alone or in combination with another substituent, means a cycloalkyl substituent containing from three to six carbon atoms and includes cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • pharmaceutically acceptable salt means a salt of a compound of formula (I) which is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, generally water or oil-soluble or dispersible, and effective for their intended use.
  • pharmaceutically-acceptable acid addition salts and pharmaceutically- acceptable base addition salts. Lists of suitable salts are found in, e.g., S.M. Birge et al. J. Pharm. Sci., 1977, 66, pp. 1-19.
  • pharmaceutically-acceptable acid addition salt means those salts which retain the biological effectiveness and properties of the free bases and which are not biologically or otherwise undesirable, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, sulfamic acid, nitric acid, phosphoric acid, and the like, and organic acids such as acetic acid, trichloroacetic acid, trifluoroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 2-acetoxybenzoic acid, butyric acid, camphoric acid, camphorsulfonic acid, cinnamic acid, citric acid, digluconic acid, ethanesulfonic acid, glutamic acid, glycolic acid, glycerophosphoric acid, he isulfic acid, heptanoic acid, hexanoic acid, formic acid, fum
  • pharmaceutically-acceptable base addition salt means those salts which retain the biological effectiveness and properties of the free acids and which are not biologically or otherwise undesirable, formed with inorganic bases such as ammonia or hydroxide, carbonate, or bicarbonate of ammonium or a metal cation such as sodium, potassium, lithium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Particularly preferred are the ammonium, potassium, sodium, calcium, and magnesium salts.
  • Salts derived from pharmaceutically- acceptable organic nontoxic bases include salts of primary, secondary, and tertiary amines, quaternary amine compounds, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion-exchange resins, such as methylamine, dimethylamine, trimethylamine, ethylamine, diethylamine, triethylamine, isopropylamine, tripropylamine, tributylamine, ethanolamine, diethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N- ethylpiperidine, tetramethylammonium compounds, tetraethylammonium
  • N,N'-dibenzylethylenediamine, polyamine resins, and the like are particularly preferred organic nontoxic bases.
  • Particularly preferred organic nontoxic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine.
  • sulfonamide derivative means a sulfonamide group of the formula -NHS0 2 -R 2 wherein R 2 is -(C 1-8 )alkyl, -(C 3-7 )cycloalkyl or ⁇ -(C 1-6 )alkyl-(C 3- 6 )cycloalkyl ⁇ , which are all optionally substituted from 1 to 3 times with halo, cyano, nitro, 0-(C 1-6 )a)kyl, amido, amino or phenyl, or R 2 is C 6 or C 10 aryl which is optionally substituted from 1 to 3 times with halo, cyano, nitro, (C 1-6 )alkyl, 0-(C 1-6 )alkyl, amido, amino or phenyl.
  • antiviral agent means an agent (compound or biological) that is effective to inhibit the formation and/or replication of a virus in a mammal. This includes agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of a virus in a mammal.
  • Antiviral agents include, for example, ribavirin, amantadine, VX-497 (merimepodib, Vertex Pharmaceuticals), VX- 498 (Vertex Pharmaceuticals), Levovirin, Viramidine, Ceplene (maxamine), XTL-001 and XTL-002 (XTL Biopharmaceuticals).
  • immunomodulatory agent means those agents
  • Immunomodulatory agents include, for example, class I interferons (such as ⁇ -, ⁇ - and omega interferons, tau-interferons, consensus interferons and asialo-interferons), class II interferons (such as ⁇ -interferons) and pegylated interferons.
  • class I interferons such as ⁇ -, ⁇ - and omega interferons, tau-interferons, consensus interferons and asialo-interferons
  • class II interferons such as ⁇ -interferons
  • pegylated interferons pegylated interferons.
  • inhibitor of HCV NS3 protease means an agent (compound or biological) that is effective to inhibit the function of HCV NS3 protease in a mammal.
  • Inhibitors of HCV NS3 protease include, for example, those compounds described in WO 99/07733, WO 99/07734, WO 00/09558, WO
  • HCV inhibitor means an agent (compound or biological) that is effective to inhibit the formation and/or replication of HCV in a mammal. This includes agents that interfere with either host or HCV viral mechanisms necessary for the formation and/or replication of HCV in a mammal.
  • Inhibitors of HCV include, for example, agents that inhibit a target selected from: NS3 protease, NS3 helicase, HCV polymerase, NS2/3 protease or IRES.
  • Specific examples of inhibitors of HCV include ISIS-14803 (ISIS Pharmaceuticals).
  • inhibitor of HCV polymerase means an agent (compound or biological) that is effective to inhibit the function of an HCV polymerase in a mammal. This includes, for example, inhibitors of HCV NS5B polymerase. Inhibitors of HCV polymerase include non-nucleosides, for example, those compounds described in:
  • EP 1 256 628 A2 (Agouron).
  • other inhibitors of HCV polymerase also include nucleoside analogs, for example, those compounds described in:
  • WO 02/057287 A2 and WO 02/057425 A2 both Merck/lsis.
  • Specific examples of inhibitors of an HCV polymerase include JTK-002/003, JTK- 109 (Japan Tobacco), and NM283 (Idenix).
  • HIV inhibitor means an agents (compound or biological) that is effective to inhibit the formation and/or replication of HIV in a mammal. This includes agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of HIV in a mammal.
  • HIV inhibitors include, for example, nucleosidic inhibitors, non-nucleosidic inhibitors, protease inhibitors, fusion inhibitors and integrase inhibitors.
  • HAV inhibitor as used herein means an agent (compound or biological) that is effective to inhibit the formation and/or replication of HAV in a mammal. This includes agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of HAV in a mammal.
  • HAV inhibitors include Hepatitis A vaccines, for example, Havrix ® (GlaxoSmithKline), VAQTA ® (Merck) and Avaxim ® (Aventis Pasteur).
  • HBV inhibitor means an agent (compound or biological) that is effective to inhibit the formation and/or replication of HBV in a mammal. This includes agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of HBV in a mammal.
  • HBV inhibitors include, for example, agents that inhibit HBV viral DNA polymerase or HBV vaccines.
  • HBV inhibitors include Lamivudine (Epivir-HBV ® ), Adefovir Dipivoxil, Entecavir, FTC (Coviracil ® ), DAPD (DXG), L-FMAU (Clevudine ® ), AM365 (Amrad), Ldt (Telbivudine), monoval-LdC (Valtorcitabine), ACH-126,443 (L-Fd4C) (Achillion), MCC478 (Eli Lilly), Racivir (RCV), Fluoro-L and D nucleosides, Robustaflavone, ICN 2001-3 (ICN), Bam 205 (Novelos), XTL-001 (XTL), Imino-Sugars (Nonyl-DNJ) (Synergy), HepBzyme; and immunomodulator products such as: interferon alpha 2b, HE2000 (Hollis-Eden), Theradigm (E
  • class I interferon means an interferon selected from a group of interferons that all bind to receptor type I. This includes both naturally and synthetically produced class I interferons. Examples of class I interferons include ⁇ -, ⁇ -, ⁇ -, omega interferons, tau-interferons, consensus interferons, asialo-interferons.
  • class II interferon as used herein means an interferon selected from a group of interferons that all bind to receptor type II. Examples of class II interferons include ⁇ -interferons.
  • the term "therapeutically effective amount" as it is used herein with respect to compounds of Formula I means an amount of the compound which is effective to treat a Flaviviridae viral infection, i.e. to inhibit or at least reduce viral replication, while not being an amount that is toxic to a mammal being treated or an amount that may otherwise cause significant adverse effects in a mammal.
  • carrier is used to refer to compounds or mixtures of compounds for combination with the present therapeutic compounds which facilitate the administration thereof and/or enhance the function of the therapeutic compounds to inhibit a virus of the Flaviviridae family including, for example, diluents, excipients, adjuvants and vehicles. Stabilizers, colorants, flavorants, anti-microbial agents, and the like may also be combined with the therapeutic compound. Moreover, in some cases, the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form. Other suitable additives include those with which one of skill in the art would be familiar that function to improve the characteristics of the present combination while not adversely affecting its function.
  • composition effective to treat a mammal infected with a virus of the Flaviviridae family.
  • the composition comprises a pharmaceutically acceptable carrier in combination with a compound of Formula (I):
  • A is selected from: C-i to C 6 alkyl and C 3 to C 6 cycloalkyl; and B is selected from: phenyl orthiazolyl, both of which optionally substituted with a group selected from NH(R 1 ) and NH(CO)R ⁇ wherein R 1 is Cj to C 6 alkyl; R is OH or a sulfonamide derivative, or a pharmaceutically acceptable salt thereof.
  • a of Formula (I) is a branched C 4 to C 6 alkyl or C 4 to C 6 cycloalkyl group. In a preferred embodiment, A is cyclopentyl or tert-butyl.
  • B of Formula (I) is phenyl or a thiazole substituted at position 2 with NH(R 1 ) or NH(CO) R in which R is a C-, to C 4 alkyl.
  • B is a thiazole substituted at position 2 with NH(R 1 ) or NH(CO) R 1 in which R 1 is a C-i to C alkyl.
  • B is 4-thiazole substituted at position 2 with NH(CO)CH 3 or with NHCH(CH 3 ) 2 .
  • R of formula (I) is OH or a sulfonamide group of formula -NHS0 2 -R 2 wherein R 2 is -(C 1-6 )alkyl, -(C 3-6 )cycloalkyl, both optionally substituted 1 or 2 times with halo or phenyl, or R 2 is C 6 aryl optionally substituted from 1 or 2 times with halo or (C 1-6 )alkyl. More preferably, R is OH or a sulfonamide group wherein R 2 is methyl, cyclopropyl or phenyl.
  • the present invention is conducted with a compound of Formula (I) in which A is terf-butyl and B is phenyl as set out below in the formula of compound (II):
  • the present invention is conducted with a compound of Formula (I) in which A is tert-butyl and B is 4-thiazole substituted at its 2 position with NH(CO)CH 3 as set out below in the formula for compound (III):
  • the present invention is conducted with a compound of Formula (I) in which A is cyclopentyl and B is 4-thiazole substituted at its 2 position with NHCH(CH 3 ) 2 as set out below in the formula for compound (IV):
  • the present invention is conducted with a compound of Formula (I) in which A is cyclopentyl, B is 4-thiazole substituted at its 2 position with NHCH(CH 3 ) 2 , and R is a sulfonamide group wherein R is phenyl as set out below in the formula for compound (VI):
  • the present invention is conducted with a compound of Formula (I) in which A is cyclopentyl, B is 4-thiazole substituted at its 2 position with NHCH(CH 3 ) 2 , and R is a sulfonamide group wherein R 2 is methyl as set out below in the formula for compound (VII):
  • the present invention is conducted with a compound of Formula (I) in which A is cyclopentyl, B is 4-thiazole substituted at its 2 position with NHCH(CH 3 ) 2 , and R is a sulfonamide group wherein R 2 is cyclopropyl as set out below in the formula for compound (VIII):
  • a method for treating a mammal infected with a virus of the Flaviviridae family comprising administering to an infected mammal a pharmaceutical composition comprising a pharmaceutically acceptable carrier in combination with a therapeutically effective amount of a compound having Formula (I), (II), (III), (IV), (VI), (VII) or (VIII) as defined above.
  • a therapeutically effective amount of a compound of Formula (I), (II), (III), (IV), (VI), (VII) or (VIII) is administered to a mammal infected with a Flaviviridae virus.
  • a dosage of between about 0.01 and about 100mg/kg body weight per day, preferably between about 0.1 and about 50mg/kg body weight per day of the compound is administered to the infected mammal.
  • the method will involve administration of the compound from about 1 to about 5 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy.
  • Administration of the therapeutic compound in the treatment of Flaviviridae viral infection may be by any one of several routes including administration orally, parenterally or via an implanted reservoir.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, and intralesional injection or infusion techniques.
  • Oral administration or administration by injection are preferred.
  • Orally acceptable dosage forms include, but not limited to, capsules, tablets, and aqueous suspensions and solutions.
  • the compound of Formula (I), (II), (III), (IV), (VI), (VII) or (VIII) is administered in combination with one or more pharmaceutically acceptable carriers.
  • the nature of the carrier(s) will, of course, vary with the dosage form. Accordingly, in the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • injectable preparations including injectable aqueous or oleaginous suspensions, are formulated according to techniques known in the art using suitable dispersing or wetting agents such as Tween 80 and suspending agents.
  • the amount of the present therapeutic compound that is combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a typical preparation will contain from about 5% to about 95% therapeutic compound (w/w).
  • such preparations contain from about 20% to about 80% therapeutic compound.
  • a combination therapy is contemplated wherein the compound of Formula (I), (II), (III), (IV), (VI), (VII) or (VIII), or a pharmaceutically acceptable salt thereof, is co-administered with at least one additional agent selected from: an antiviral agent, an immunomodulatory agent, an HCV inhibitor, an HIV inhibitor, an HAV inhibitor, an HBV inhibitor, and a therapeutic effective to treat the symptoms of the viral infection.
  • additional agents may be combined with the compounds of this invention to create a single pharmaceutical dosage form. Alternatively these additional agents may be separately administered to the infected patient as part of a multiple dosage, form, for example, using a kit. Such additional agents may be administered to the patient prior to, concurrently with, or following the administration of the therapeutic compound of Formula (1), (II), (III), (IV), (VI), (VII) or (VIII), or a pharmaceutically acceptable salt thereof.
  • both the present compound and the additional therapeutic and/or prophylactic agents should be present at dosage levels of between about 10 to 100%, and more preferably between about 10 and 80% of the dosage normally administered in a monotherapy regimen.
  • an article of manufacture comprising packaging material contained within which is a composition effective to treat a mammal infected with a virus of the Flaviviridae family and the packaging material comprises a label which indicates that the composition can be used to treat infection by a virus of the Flaviviridae family, wherein said composition comprises a compound of Formula (I), (II), (111), (IV), (VI), (VII) or (VIII) as defined above.
  • the different embodiments of the present invention may be directed to distinct viruses, particularly viruses which are pathogenic in mammal.
  • the above-mentioned compounds of compositions can be used for the treatment of hepacivirus genus, such as Hepatitis C.
  • Preferred examples of HCV viruses are selected from genotypes: 1 , 2, 3, 4, 5, and 6. More preferred examples of HCV viruses are selected from genotypes: 2, 3, 4, 5, and 6.
  • HCV viruses are selected from subtypes: HCV 1 a, HCV1 b, HCV 2a-c, HCV 3a-b, HCV 4a, HCV 5 and HCV 6a,h,d & k. More preferably, HCV viruses are selected from subtypes: HCV 1a, HCV 2a-c, HCV 3a-b, HCV 4a, HCV 5 and HCV 6a,h,d & k.
  • the compounds of the invention may be directed against viruses of the flavivirus genus, such as the Dengue Fever viruses, Japanese Encephalitis viruses, West Nile viruses and Yellow Fever viruses.
  • the invention is directed at the treatment of viral disease in a human caused by Dengue virus.
  • the invention is directed at the treatment of viral disease in a human caused by Japanese encephalitis virus.
  • the invention is directed at the treatment of viral disease in a human caused by Yellow Fever virus.
  • the invention is directed at the treatment of viral disease in a human caused by West Nile virus.
  • the invention may be directed to the treatment of viral disease caused by viruses of the pestivirus genus, such as the bovine viral diarrhea virus (BVDV), classical swine fever virus (CSFV) and border disease virus (BDV).
  • viruses of the pestivirus genus such as the bovine viral diarrhea virus (BVDV), classical swine fever virus (CSFV) and border disease virus (BDV).
  • BVDV bovine viral diarrhea virus
  • CSFV classical swine fever virus
  • BDV border disease virus
  • the invention is directed at the treatment of viral diseases in cattle caused by BVDV.
  • the invention is directed at the treatment of viral diseases in pigs caused by CSFV.
  • the invention is directed at the treatment of viral diseases in sheep caused by BDV.
  • GB viruses can also be treated with composition of the present invention.
  • HBV Hepatitis G virus
  • Preferred examples are selected from: GBV-A, B & C.
  • the invention is directed at the treatment of viral disease in a human caused by GBV-C.
  • the invention is directed at the treatment of viral disease in a human caused by GBV-A.
  • the invention is directed at the treatment of viral disease in a human caused by HGV.
  • Embodiments of the present invention are exemplified by the following specific examples which are not to be construed as limiting.
  • Abu aminobutyric acid
  • DABCYL 4-((4-(dimethylamino)phenyl) azo)benzoic acid
  • DMEM Dulbecco's Modified Eagle Medium
  • DMSO dimethyl sulfoxide
  • EDANS 5- ((2-aminoethyl)amino)naphthalene-1-sulfonic acid
  • HPLC high performance liquid chromatography
  • IPTG isopropyl-b-D-thiogalactoside
  • LB Luria-Bertoni (as in LB broth)
  • Nva norvaline
  • PenStrep penicillin/streptomycin
  • PCR polymerase chain reaction
  • r.m.s. root mean square
  • RT-PCR real-time polymerase chain reaction
  • TCEP tris(2-carboxyethyl)phosphine hydrochloride.
  • HCV NS3/4A 1a and 1b For production of the HCV genotype 1 b NS3-NS4A heterodimer protein, a full-length HCV cDNA was cloned by RT-PCR using RNA extracted from the serum of an HCV genotype 1b infected individual (provided by Dr. Bernard Willems, H ⁇ pital St-Luc, Montreal, Canada).
  • the DNA region encoding the NS3-NS4A heterodimer protein was PCR-amplified (forward primer: 'CTCGGATCCGGCGCCCATCACGGCCTAC3' (SEQ ID No.1) ; reverse primer: 5'CTCTCTAGATCAGCACTCTTCCATTTCAT CGAA3') (SEQ ID No.2)) from the full-length HCV cDNA and subcloned into the pFastBacTM HTa baculovirus expression vector (Gibco/BRL).
  • HCV genotype 1a the DNA encoding NS3-NS4A heterodimer protein was PCR-amplified (forward primer: 5'CTCTCTAGATCAGCACTCTTCCATTTCATCGAACTC3' (SEQ ID No.3); reverse primer: 5OTCGGATCCGGCGCCCATCACGGCCTACTCCCAA3' (SEQ ID No.4)) from the HCV genotype 1a strain H77 (provided by ViroPharma Inc., Exton, PA, US) and subcloned as described above.
  • the cloning into the pFastBacTM HTa baculovirus expression vector generated a recombinant fusion protein containing an additional N-terminal 28 residues that comprised a hexahistidine tag and a rTEV protease cleavage site.
  • the Bac-to-BacTM baculovirus expression system (Gibco/BRL) was used to produce the recombinant baculovirus for protein expression.
  • His-tagged NS3-NS4A heterodimer protease was expressed by infecting Sf21 insect cells (lnvitrogen) at a density of 10 6 cells/mL with the recombinant baculovirus at a multiplicity of infection of 0.1-0.2 at 27°C.
  • the infected culture was harvested 48 to 64 h later by centrifugation at 4°C.
  • the cell pellet was homogenized in 50 mM sodium phosphate, pH 7.5, 40% glycerol (w/v), 2mM ⁇ -mercaptoethanol.
  • His-NS3- NS4A heterodimer protease was then extracted from the cell lysate with 1.5% NP- 40, 0.5% Triton X-100, 0.5M NaCI, and a DNase treatment. After ultracentrifugation (100,000xg for 30 min at 4°C), the soluble extract was diluted 4-fold in 50 mM sodium phosphate, pH 7.5, 0.5M NaCI and loaded on a Pharmacia Hi-Trap Ni +2 - chelating column.
  • the His-NS3-NS4A heterodimer protein was eluted using a 50 to 400 mM imidazole gradient prepared in 50 mM sodium phosphate, pH 7.5, 10% (w/v) glycerol, 0.1% NP-40, 0.5M NaCI.
  • Co-purification of mature NS3 and NS4A protein complex was verified by Western blot analysis of the purified proteins using an anti-NS3 and an anti-NS4A antiserum produced in-house.
  • the purified NS3 protease domain and the peptide H 2 N-PDREVLYREFDEMEEC-OH (SEQ ID No. 5 ) were used to immunize rabbits for the production of antisera specific to NS3 and NS4A respectively.
  • the NS3-NS4 protease genes of HCV genotypes 2b and 3a were amplified from RNA isolated from clinical samples of HCV-infected patients obtained from Dr. G. Steinmann (Germany) using RT-PCR.
  • the primers used for the RT- PCR were 5OTCGGATCCGGCTCCCATTACTGCTTAC3' (SEQ ID No. 6) as the forward and 5'GACGCGTCGACGCGGCCGCTCAGCACTCTTCCATTTCACTGAA3' (SEQ ID No.7) as the reverse primer for the NS3-NS4A of HCV genotype 2b and; 5OTCGGATCGGGCCCCGATCACAGCATACGCC3' (SEQ ID No.
  • Each primer contained a unique restriction site for subcloning the fragment in the bacterial expression vector pET11a.
  • the cloning into the vector generated a recombinant fusion protein containing an additional N-terminal 28 residues that comprised a hexahistidine tag and a rTEV protease cleavage site.
  • the recombinant plasmids were used to transform the bacterial strain BL21 DE3 pLysS for protein expression and recombinant protein expression was induced by the addition of IPTG.
  • the cells were collected by centrifugation at 4°C and the cell pellet lysed in a buffer containing 50mM sodium phosphate, 0.5M NaCI, 40% glycerol, 1.5% NP-40, 0.5% Triton X- 100 and the soluble protein fraction was passed through a 5ml HiTrap chelating affinity column as described above.
  • a poly(U)-Sepharose purification step can optionally be conducted in order to remove degradation products and contaminants.
  • the fluorogenic depsipeptide substrate used to assess inhibition of protease activity of the HCV 1 a, 1 b, 2b, 2a-c and 3a NS3-NS4A heterodimer protein was anthranilyl- DDlVPAbu[C(0)-0]-AMY(3-N0 2 )TW-OH (SEQ ID No.10). This substrate is cleaved between the aminobutyric (Abu) and the alanine residues.
  • the sequence DDIVPAbu-AMYTW (SEQ ID No.11) is derived from the sequence DDIVPC- SMSYTW (SEQ ID No.12) corresponding to the NS5A/NS5B natural cleavage site.
  • the introduction of the aminobutyric residue at position P1 resulted in a significant decrease of the N-terminal product inhibition while the deletion of the serine residue at position P3 improved the internal quenching efficiency.
  • the protease activity was assayed in 50 mM Tris-HCI, pH 8.0, 0.25M sodium citrate, 0.01% n-dodecyl- ⁇ -D- maltoside, 1 mM TCEP.
  • the assay was performed in a Microfluor®2 White U- Bottom Microtiter® plate. 5 ⁇ M of the substrate and various concentrations of the test compound were incubated with 1.5 nM of HCV 1a or 1b NS3-NS4A heterodimer protein for 45 minutes at 23°C under gentle agitation. The final DMSO content did not exceed 5.25%.
  • the reaction was terminated by the addition of a 1 M 2-[N- morpholinojethanesulfonic acid solution at pH 5.8.
  • the fluorescence was monitored using the BMG POLARstar Galaxy 96-well plate reader with an excitation filter of 320 nm, and an emission filter of 405 nm.
  • the inhibition constant (Ki) for compound (IV) was determined using a steady-state velocity method (Morrisson etal. 1985).
  • the protease activity was determined by monitoring the fluorescence change associated with the cleavage of the internally quenched fluorogenic substrate using a SLM- AMINCO ® 8100 spectrofluorometer (emission at 325 nm and excitation at 420 nm).
  • the cleavage reaction was monitored in the presence of 0.25 to 8 ⁇ M of substrate, 0.3 nM of HCV 1a or 1b NS3-NS4A heterodimer protein and various concentrations of the test compound IV in 50 mM Tris-HCI, pH 8.0, 0.25M sodium citrate, 0.01 % n- dodecyl- ⁇ -D-maltoside, 1 mM TCEP.
  • the steady-state analysis of inhibition of the HCV genotype 1a and 1b NS3-NS4A heterodimer proteases were performed using the Dixon and Cornish-Bowden graphical methods. In the Dixon graphical method, the reciprocal velocity 1/V is plotted against the test compound concentration at several substrate concentrations (S).
  • the ratio S/V is plotted against the test compound concentration at several S concentrations.
  • the points lie on a straight line at each S value.
  • the Dixon plot displays lines at different S values intersecting at a single point, while the Cornish-Bowden plot displays parallel lines at different S values.
  • the Kj was estimated by fitting the data to the equation describing competitive binding:
  • HCV genotype 1a, 1b, 2b, 2a-c and 3a NS3/NS4A heterodimer proteases was determined in the presence of compound (IV) using the in vitro enzymatic fluorogenic assay.
  • the IC 50 curves were analyzed individually by the SAS NLIN procedure.
  • For HCV 1a NS3-NS4A protease an average IC 50 value of 4.3 nM was obtained from the analysis of two batches of compound (IV).
  • An average IC 50 value of 3.4 nM was obtained from the analysis of two batches of compound (IV) in the presence of HCV 1 b NS3-NS4A protease.
  • Figures 3A and 3B illustrate the IC 50 curves of compound (IV) against HCV 1a and 1b NS3-NS4A proteases, respectively.
  • Compound (IV) was found to be a competitive test compound of HCV genotype 1a and 1b NS3-NS4A proteases following fitting of the data to the equation describing competitive binding with GraFit and also from the Dixon arid Cornish-Bowden graphical methods.
  • the Dixon plot showed that the lines at different S values were intersecting at a single point, while the Cornish- Bowden plot showed parallel lines at various S values.
  • FIG. 4 A Kj of 0.30 nM was obtained for compound (IV) from steady-state velocity analysis with the HCV genotype 1a NS3-NS4A protease, while a K f of 0.66 nM was obtained with the HCV genotype 1b NS3-NS4A protease, K
  • Figures 4 and 5 are the Dixon and Cornish-Bowden plots of compound (IV) against the HCV genotype 1a and 1b NS3-NS4A, proteases respectively.
  • HCV RNA replication was demonstrated in HCV 1a and 1b replicon-containing, Huh- 7-derived cell lines developed at Boehringer Ingelheim (Canada) Ltd., R & D (WO 02/052015). These cells were used to establish a sensitive HCV RNA replication cell-based assay for testing candidate test compounds.
  • Huh7 cells that stably maintain a subgenomic HCV replicon were established as previously described (Lohman et al. 1999; WO 02/052015). The cells were seeded into a 96 well cell culture cluster at 1 X 10 4 cells per well in DMEM complemented with 10% FBS, PenStrep (Life Technologies) and 1 ⁇ g/mL Geneticin. Cells were incubated in a 5% C0 2 incubator at 37 °C until addition of various concentrations of the test compound.
  • test compound was prepared for use in the assay as follows.
  • the test compound in 100% DMSO was added diluted in assay Medium for a final DMSO concentration of 0.5% and the solution was sonicated for 15 min and filtered through a 0.22 ⁇ M Millipore Filter Unit.
  • Serial dilutions of the test compounds were prepared using Assay Medium (containing 0.5% DMSO).
  • Cell culture medium was aspirated from the 96-well plate containing the cells and the appropriate dilution of test compound in assay medium was transferred to independent wells of the cell culture plate which was incubated at 37° with 5% C0 2 .
  • RT-PCR reverse transcription polymerase chain reaction
  • Comparison of the threshold cycles provides a highly sensitive measure of relative template concentration in different samples. Monitoring during early cycles, when PCR fidelity is at its highest, provides precise data for accurate quantification.
  • the relative template concentration can be converted to real numbers by using the standard curve of HCV with known number of copy.
  • compound (IV) showed a dose-dependent inhibition of HCV RNA replication with EC 50 s of 2.1 nM and 1 nM using HCV replicon-containing cells of genotype 1a and 1b, respectively. Cytotoxicity was not observed at concentrations higher than 1000 nM.
  • the full length GBV-B NS3/NS4A protease gene was isolated from infected tamarin serum. Total RNA was isolated from the serum using Qiagen ® viral RNA extraction kit according to standard protocol. The selection of primers for the reverse transcriptase and the PCR reactions were selected based on the published sequence (Muerhoff, A.S et al. 1995) and GeneBank accession number U22304 (forward: 5OGCATATGGCACCTTTTACGCTGCAGTGTC3' (SEQ ID No.14); reverse: 5'CGCGCGCTCGAGACACTCCTCCACGATTTCTTC3' (SEQ ID No.15)).
  • the amplified RT-PCR product was cloned into the polyhistidine tag-containing pET- 29 plasmid between the Ndel and Xhol sites in frame with the polyhistidine tag.
  • This construct produced a recombinant protein with a poly His tag at its C-terminus.
  • the plasmid was transformed into BL21 DE3 pLysS for protein expression.
  • the E. coli clone pGBV-B was grown in LB medium to a cell density (OD 600 ) of 0.6 at which time IPTG was added at a concentration of 0.2 mM. The induction was done at 23 C for a period of 4 hours.
  • the GBV-B NS3/NS4A-His protein was purified according to the procedure described by Zhong et al., 1999.. The soluble fraction was purified on a Pharmacia ® Hi-Trap Ni +2 -chelating affinity column using a 50 to 400 mM imidazole gradient. This step was followed by chromatography of the protein preparation on a Superdex ® 200 gel filtration column. The concentration of the protein preparation was determined by Bradford protein assay.
  • the depsipeptide substrate used to assess inhibition of the protease activity of GBV-
  • B NS3-NS4A heterodimer protein was Ac-DED(EDANS)EE-Abu[C(0)-0] ASK(DABCYL)-NH 2 (SEQ ID No. 16)
  • This substrate is cleaved between the aminobutyric (Abu) and the alanine residues and products of the reaction were analyzed by HPLC.
  • the protease activity was assayed in 50 mM Tris-HCI, pH 8.0, 0.25M sodium citrate, 0.01% n-dodecyl- ⁇ -D-maltoside, 1 mM TCEP.
  • the assay was performed in a Microfluor®2 White U-Bottom Microtiter® plate.
  • Hepatocytes isolated from uninfected tamarins were maintained in culture in defined medium for several days.
  • the cell culture model was as described by Beames et al. 2000.
  • Three days after plating the cells were infected with GBV-B-containing plasma. After viral adsorption, the virus was removed and the cells were incubated in the presence and absence of candidate test compounds at a concentration of 10 ⁇ M.
  • the cells were harvested 7 days post infection.
  • the levels of GBV-B RNA were quantitated from total cellular RNA by a real-time PCR assay using a primer-probe combination that recognized a portion of the GBV-B capsid gene (Beames etal. 2000).
  • proteases from other viruses of the flaviviridae family of viruses were obtained from a BLAST analysis (Altschul et al.1997) NCBI followed by the taxonomy report. The search for protease sequences was done using specific criteria that would retrieve all flaviviridae sequences with similarity to HCV protease excluding HCV sequences themselves. When possible an "NP_" sequence from NCBI was used to represent a particular group of related viruses since these "NP_" sequences are "Reference" sequences (RefSeq). Furthermore, when not known, the NS3 N-terminal and C-terminal ends for the proteases were determined from similarity with HCV NS3 protease.
  • Table 4 represents identities and similarities between several genotypes and subtypes of HCV. One can see that identities range between 70% and 89% whereas similarities range between 77% and 95% demonstrating that the NS3 protease is highly conserved among HCV. Also, from the amino acid sequence alignment of NS3 protease domain of various representative HCV genotypes ( Figure 2) and location of the conserved residues on the three dimensional structure ( Figure 8), one can see that the residues at the active site are highly conserved among HCV genotypes and subtypes.
  • Table 5 is an analysis of the similarity between GB viruses NS3 protease and HCV NS3 protease domains. Similarities range between 43 and 49%, whereas similarities between HCV and pestivirus NS3 domains range from 25 and 30% (Table 6). Similarities between HCV and dengue virus range also around 30% (Table 7), as well as the similarity between HCV and Flavivirus (Table 8). In contrast, Table 9 shows that the similarities between the NS3 protease domain of HCV and the protease of Human Cytomegalovirus (HCMV) revolves around 20%.
  • HCMV Human Cytomegalovirus
  • HCMV is a virus of the Herpes viridae and has a serine protease that has a different catalytic triad from Flaviviridae proteases.
  • the HCMV virus protease is therefore not considered to be similar to Flaviviridae NS3 proteases. This contrast will be illustrated further when comparison is made of the similarities between the amino acid in contact with our inhibitors in Flaviviridae and HCMV.
  • Example 5 Three-Dimensional Structural Crystal Studies
  • a crystal structure was obtained of the HCV NS3 protease complexed with an inhibitor, and the amino acids directly in contact with the inhibitor were analyzed in other Flaviviridae to assess degrees of similarity of these important contact points.
  • a co-crystal of the HCV NS3 protease-NS4A peptide complexed with macrocyclic compound (II) was obtained that diffracted X-rays to 2.75 A resolution.
  • the macrocyclic compound II was found at the NS3 protease active site and was clearly defined in the difference electron density. This structure reveals molecular details of how the test compound interacts with the active site and provides additional insight into the mechanism of inhibition of the HCV NS3 protease.
  • the structure of the NS3 protease complex with the test compound II ( Figure 8) better defines the binding site of the present substrate-based test compound series. From this structure of the co-complex, the residues directly in contact with the test compound i.e. within 3 A (H57, G137, S139, A156 and A157) are indicative of the active site, as predicted by the competitive mode of inhibition, and by the conservation at this site among representative sequences of HCV genotypes and various subtypes.
  • Viruses within the Flaviviridae family possess a number of similarities ( Figure 1 ) despite a relatively low overall sequence homology among its members (Tables 4-8).
  • the NS3 protein contains sequences with similarity to the protease and the nucleoside triphosphate-binding helicase of pestiviruses and flaviviruses.
  • the HCV NS3 protease domain shares a sequence similarity of about 77-95% among HCV genotypes ( Figure 1) and a sequence similarity of about 25-50% with other members of the Flaviviridae family (Tables 4-8).
  • BVDV-2 Bovine viral Diarrhea virus TABLE 7 - /> SIMILARITY OF HCV NS3 PROTEASE DOMAIN ( 181 AA) WITH MOSQUITO -BORNE
  • WVN West Nile virus
  • JEV Japanese encephalitis virus
  • MVEV Murray Valley encephalitis virus
  • HCV40_1 b Sequence of a HCV 1 b isolate that was used as reference for this analysis
  • n number of HCV sequences of all genotypes that was used in the study
  • GBV a The aa in bold are the residue found for GBV-B NS3 protease : % similarity for GBV-B

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Public Health (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Virology (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

Compositions, use , article of manufacture and method for the treatment of a mammal infected with a virus of the Flaviviridae family are provided comprising administration to the infected mammal of a compound having Formula (I), wherein A is selected from: C1 to C6 alkyl and C3 to C6 cycloalkyl; and B is selected from: phenyl or thiazolyl, both of which optionally substituted with a group selected from NH(R1) and NH(CO)R1, wherein R1 is C1 to C6 alkyl; R is OH or a sulfonamide derivative; or a pharmaceutically acceptable salt thereof.

Description

COMPOSITION FOR THE TREATMENT OF INFECTION BY FLAVIVIRIDAE
VIRUSES
FIELD OF THE INVENTION The present invention relates to compounds, compositions, use and method for the treatment of a Flaviviridae viral infection in a mammal. More particularly, the present invention relates to the use and a method of treatment comprising administration of a compound selected from a macrocyclic peptide family of compounds.
BACKGROUND OF THE INVENTION
The Flaviviridae family of viruses are enveloped positive-stranded RNA viruses comprising a number of human pathogenic viruses such as viruses of the hepacivirus genus, including Hepatitis C, viruses of the flavivirus genus, including the Dengue Fever viruses, encephalitis viruses, West Nile viruses and Yellow Fever viruses, and viruses of the pestivirus genus, including the bovine viral diarrhea virus and border disease virus, both of which are animal pathogens. The most newly discovered viruses, hepatitis G virus (HGV) and hepatitis GB virus (GBV-A, B, C), are also provisionally considered to be members of the Flaviviridae family belonging to a distinct genus.
Dengue viruses, members of the family of Flaviviridae are transmitted by mosquitos. There are four serotypes that cause widespread human diseases, one of which causes dengue hemorrhagic fever, and about 40% of the population living in tropical and subtropical regions of the world is at risk for infection. Of the 1 million cases of hemorrhagic fever cases per year, about 5% are fatal. There is currently no effective vaccine or antiviral drug to protect against dengue diseases.
Pestiviruses such as bovine diarrhea virus (BVDV), classical swine fever virus (CSFV) and border disease virus (BDV) comprise a group of economically important animal pathogens affecting cattle, pigs and sheep. These positive-sense RNA viruses are classified as a separate genus in the family Flaviviridae.
GB-viruses have been classified as members of the Flaviviridae family, but have not yet been assigned to a particular genus based on the analysis of genomic sequences. These RNA viruses, in addition to infecting humans, can cause acute resolving hepatitis in experimentally infected tamarins.
Viruses within the Flaviviridae family possess a number of similarities despite a relatively low overall sequence homology among its members. The genome of these viruses is a small single-stranded RNA (« 10 kilobases in length) having a single open reading frame (ORF). The ORF encodes a polyprotein of about 3000 amino acids that contains structural proteins at its 5' end and non-structural (NS) proteins at its 3' end. The polyprotein is proteolytically processed by both viral and host- encoded proteases into mature polypeptides, which for HCV, are as follows: NH2-{C- E1-E2-P7-NS2-NS3-NS4A-NS4B-NS5A-NS5B}-COOH. The core protein or nucleocapsid (C) and the two envelope glycoproteins (E1 and E2) represent the constitutive structural proteins of the virions. These structural proteins are followed by the nonstructural (NS) proteins, which at least some are thought to be essential for viral RNA replication.
Enzymatic activities have been ascribed to several of these NS proteins. In particular, the NS3 protein contains sequences with similarity to the serine protease and the nucleoside triphosphate-binding helicase of pestiviruses and flaviviruses. Analysis of sequence alignments had predicted the existence of a trypsin-like serine protease domain within the N-terminal region of flavi-, pesti-, hepaci- and GB- viruses. Sequence similarities between NS3 proteolytic domains of Flaviviridae viruses are well-established (Ryan MD et al. 1998). The HCV NS3 protease domain shares a sequence similarity of about 77-90% among HCV genotypes and a sequence similarity of about 25-50% with other members of the Flaviviridae family. With respect to the three dimensional structure, the available atomic co-ordinates of the various crystallized HCV and Dengue NS3 proteases show an overall architecture that is characteristic of the trypsin-like fold (Kim et al. 1996, Love et al. 1996; Murthy et al. 1999). Many studies have now firmly established that the N- terminal portion of the NS3 region encodes a serine protease that has a very specific and pivotal role in viral polyprotein processing within Flaviviridae. In hepacivirus, pestivirus and GB viruses, polyprotein processing shows a requirement for the downstream NS4A protein. The NS4A protein acts as a cofactor that enhances the NS3 protease cleavage efficiency (Lin et al., 1995; Kim et al., 1996; Stein uler et al., 1996). In flavivirus, this requirement for enhancing the NS3 protease activity is provided by the upstream NS2B protein. The genomic organization and structure of GBV-B and HCV are similar despite the fact that the sequence homology between the polyprotein sequences of GBV-B and HCV is about 25 to 30%.
Given apparent similarities of viruses within the Flaviviridae family of viruses, it would be desirable to develop therapeutic agents effective against Flaviviridae viruses, and more particularly, effective against the pathogenic members of the Flaviviridae family.
SUMMARY OF THE INVENTION Accordingly, in a first aspect of the present invention, there is provided an anti- Flaviviridae virus composition comprising a pharmaceutically acceptable carrier in combination with a compound of Formula (I):
Figure imgf000005_0001
Formula (I)
wherein,
A is selected from: C^ to C6 alkyl and C3 to C6 cycloalkyl; and B is selected from: phenyl or thiazolyl, both of which optionally substituted with a group selected from NH(R1) and NH(CO)R1, wherein R1 is C1 to C6 alkyl; R is OH or a sulfonamide derivative, or a pharmaceutically acceptable salt thereof.
In a second aspect, the present invention provides a method for treating a mammal infected with a virus of the Flaviviridae family comprising administering to the infected mammal a pharmaceutical composition comprising a pharmaceutically acceptable carrier in combination with a therapeutically effective amount of a compound of Formula (I) as defined above. In a third aspect, the present invention provides a method of treating a mammal infected with a virus of the Flaviviridae family wherein a pharmaceutical composition comprising a pharmaceutically acceptable carrier in combination with a therapeutically effective amount of a compound of Formula (I) as defined above is co-administered with at least one additional agent selected from: an antiviral agent, an immunomodulatory agent, an HCV inhibitor, an HIV inhibitor, an HAV inhibitor and an HBV inhibitor; to the infected mammal.
In a fourth aspect, the present invention provides a pharmaceutical composition for treating or preventing an infection of a mammal caused by a virus of the Flaviviridae family comprising a pharmaceutically acceptable carrier in combination with a therapeutically effective amount of a compound of Formula (I) and at least one additional agent selected from: an antiviral agent, an immunomodulatory agent, an HCV inhibitor, an HIV inhibitor, an HAV inhibitor and an HBV inhibitor.
In a fifth aspect of the present invention, there is provided the use of a compound of Formula (I) as defined above, for the manufacture of a medicament for the treatment of Flaviviridae viral infection.
In a sixth aspect of the present invention, there is provided an article of manufacture comprising packaging material contained within which is a composition effective to inhibit a virus of the Flaviviridae family and the packaging material comprises a label which indicates that the composition can be used to treat infection by a virus of the Flaviviridae family and, wherein said composition comprises a compound of Formula (I) as defined above.
While not wishing to be bound to any particular mode of action, the macrocyclic peptide family of compounds generally represented by Formula (I) as set out above are believed to interact at a conserved substrate binding site in the NS3 protease domain. Crystal structure studies confirm that the present macrocyclic compounds interact at the substrate-binding site within the HCV NS3 domain, a region that is functionally conserved among Flaviviridae viruses.
Other objects, advantages and features of the present invention will become more apparent upon reading the following non-restrictive description of the preferred embodiments with reference to the accompanying drawings and tables in which:
BRIEF DESCRIPTION OF TABLES Tables 4-8 provide a sequence similarity comparison between members of the Flaviviridae family.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 provides a comparison of polyproteins of viruses belonging to the Flaviviridae family (taken from: Ryan etal., 1998, J. Gen. Virology 79, 947-959);
Figure 2 is a sequence comparison of the NS3 protease domain between HCV genotypes and subtypes;
Figures 3A-B are the IC50 curves of a macrocyclic peptide compound according to
Formula (I) against HCV genotype 1a and 1b NS3-NS4A proteases, respectively; Figures 4A-B are the Dixon and Cornish-Bowden plots of the macrocyclic peptide of
Figure 3 against the HCV genotype 1a NS3-NS4A proteases;
Figures 5A-B are the Dixon and Cornish-Bowden plots of the macrocyclic peptide of
Figure 3 against the HCV genotype 1b NS3-NS4A proteases;
Figure 6 illustrates the inhibition of GBV-B replication by macrocyclic peptides according to Formula (I) in tamarin hepatocytes in culture;
Figure 7 graphically illustrates dose-dependent inhibition of GBV-B replication by the macrocyclic peptide III of Figure 6; and
Figure 8 illustrates the 3-dimensional crystal structure of the HCV NS3-NS4A peptide structure complexed with a macrocyclic peptide according to Formula (I).
DETAILED DESCRIPTION OF THE INVENTION
Definitions
Unless defined otherwise, the scientific and technological terms and nomenclature used herein have the same meaning as commonly understood by a person of ordinary skill in the art to which this invention pertains. Generally, the procedures for cell culture, infection, molecular biology methods and the like are common methods used in the art. Such standard techniques can be found in reference manuals such as for example Sambrook et al. (1989) and Ausubel et. al. (1994).
The term "Flaviviridae" as it is used herein to designate a viral family is meant to encompass viruses of the hepacivirus genus, such as Hepatitis C, viruses of the flavivirus genus, such as the Dengue Fever viruses, Encephalitis viruses, West Nile viruses and Yellow Fever viruses, and viruses of the pestivirus genus, such as the bovine viral diarrhea virus and border disease virus. Hepatitis G virus (HGV) and Hepatitis GB virus are also included in this viral family although the genus of these viruses has not yet been determined. Moreover, all subtypes and genotypes of the above-mentioned viruses are also encompassed within the Flaviviridae family, including for example, HCV 1a, HCV1b, HCV 2a-c, HCV 3a-b, HCV 4a, HCV 5 and HCV 6a,h,d & k, as well as GBV-A, B & C.
The term "mammal" as it is used herein is meant to encompass humans, as well as non-human mammals which are susceptible to infection by a Flaviviridae virus including domestic animals, such as cows, pigs, horses, dogs and cats, and sheep.
With respect to the compounds of Formula (I) administered in the treatment of
Flaviviridae infection, the term "C1-6 alkyl" or "C C6" as used herein, either alone or in combination with another substituent, means acyclic, straight or branched chain alkyl substituents containing from one to six carbon atoms and includes, for example, methyl, ethyl, propyl, isopropyl, butyl, tert-buϊyl, hexyl, -methylethyl, 1-methylpropyl, 2-methylpropyl, 1 ,1-dimethylethyl.
The term "C3-6 cycloalkyl" as used herein, either alone or in combination with another substituent, means a cycloalkyl substituent containing from three to six carbon atoms and includes cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
The term "pharmaceutically acceptable salt" means a salt of a compound of formula (I) which is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, generally water or oil-soluble or dispersible, and effective for their intended use. The term includes pharmaceutically-acceptable acid addition salts and pharmaceutically- acceptable base addition salts. Lists of suitable salts are found in, e.g., S.M. Birge et al. J. Pharm. Sci., 1977, 66, pp. 1-19.
The term "pharmaceutically-acceptable acid addition salt" means those salts which retain the biological effectiveness and properties of the free bases and which are not biologically or otherwise undesirable, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, sulfamic acid, nitric acid, phosphoric acid, and the like, and organic acids such as acetic acid, trichloroacetic acid, trifluoroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 2-acetoxybenzoic acid, butyric acid, camphoric acid, camphorsulfonic acid, cinnamic acid, citric acid, digluconic acid, ethanesulfonic acid, glutamic acid, glycolic acid, glycerophosphoric acid, he isulfic acid, heptanoic acid, hexanoic acid, formic acid, fumaric acid, 2- hydroxyethanesulfonic acid (isethionic acid), lactic acid, maleic acid, hydroxymaleic acid, malic acid, malonic acid, mandelic acid, mesitylenesulfonic acid, methanesulfonic acid, naphthalenesulfonic acid, nicotinic acid, 2- naphthalenesulfonic acid, oxalic acid, pamoic acid, pectinic acid, phenylacetic acid, 3-phenylpropionic acid, picric acid, pivalic acid, propionic acid, pyruvic acid, pyruvic acid, salicylic acid, stearic acid, succinic acid, sulfanilic acid, tartaric acid, p- toluenesulfonic acid, undecanoic acid, and the like.
The term "pharmaceutically-acceptable base addition salt" means those salts which retain the biological effectiveness and properties of the free acids and which are not biologically or otherwise undesirable, formed with inorganic bases such as ammonia or hydroxide, carbonate, or bicarbonate of ammonium or a metal cation such as sodium, potassium, lithium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Particularly preferred are the ammonium, potassium, sodium, calcium, and magnesium salts. Salts derived from pharmaceutically- acceptable organic nontoxic bases include salts of primary, secondary, and tertiary amines, quaternary amine compounds, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion-exchange resins, such as methylamine, dimethylamine, trimethylamine, ethylamine, diethylamine, triethylamine, isopropylamine, tripropylamine, tributylamine, ethanolamine, diethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N- ethylpiperidine, tetramethylammonium compounds, tetraethylammonium compounds, pyridine, N,N-diιmethylaniline, N-methylpiperidine, N-methylmorpholine, dicyclohexylamine, dibenzylamine, N,N-dibenzylphenethylamine, 1-ephenamine, . N,N'-dibenzylethylenediamine, polyamine resins, and the like. Particularly preferred organic nontoxic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine.
The term "sulfonamide derivative" as used herein means a sulfonamide group of the formula -NHS02-R2 wherein R2 is -(C1-8)alkyl, -(C3-7)cycloalkyl or {-(C1-6)alkyl-(C3- 6)cycloalkyl}, which are all optionally substituted from 1 to 3 times with halo, cyano, nitro, 0-(C1-6)a)kyl, amido, amino or phenyl, or R2 is C6 or C10 aryl which is optionally substituted from 1 to 3 times with halo, cyano, nitro, (C1-6)alkyl, 0-(C1-6)alkyl, amido, amino or phenyl.
The term "antiviral agent" as used herein means an agent (compound or biological) that is effective to inhibit the formation and/or replication of a virus in a mammal. This includes agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of a virus in a mammal. Antiviral agents include, for example, ribavirin, amantadine, VX-497 (merimepodib, Vertex Pharmaceuticals), VX- 498 (Vertex Pharmaceuticals), Levovirin, Viramidine, Ceplene (maxamine), XTL-001 and XTL-002 (XTL Biopharmaceuticals).
The term "immunomodulatory agent" as used herein means those agents
(compounds or biologicals) that are effective to enhance or potentiate the immune system response in a mammal. Immunomodulatory agents include, for example, class I interferons (such as α-, β- and omega interferons, tau-interferons, consensus interferons and asialo-interferons), class II interferons (such as γ-interferons) and pegylated interferons.
The term "inhibitor of HCV NS3 protease" as used herein means an agent (compound or biological) that is effective to inhibit the function of HCV NS3 protease in a mammal. Inhibitors of HCV NS3 protease include, for example, those compounds described in WO 99/07733, WO 99/07734, WO 00/09558, WO
00/09543 or WO 00/59929, WO 03/064416; WO 03/064455; WO 03/064456 and the Vertex pre-development candidate identified as VX-950.
The term "HCV inhibitor" as used herein means an agent (compound or biological) that is effective to inhibit the formation and/or replication of HCV in a mammal. This includes agents that interfere with either host or HCV viral mechanisms necessary for the formation and/or replication of HCV in a mammal. Inhibitors of HCV include, for example, agents that inhibit a target selected from: NS3 protease, NS3 helicase, HCV polymerase, NS2/3 protease or IRES. Specific examples of inhibitors of HCV include ISIS-14803 (ISIS Pharmaceuticals).
The term "inhibitor of HCV polymerase" as used herein means an agent (compound or biological) that is effective to inhibit the function of an HCV polymerase in a mammal. This includes, for example, inhibitors of HCV NS5B polymerase. Inhibitors of HCV polymerase include non-nucleosides, for example, those compounds described in:
WO 03/010140 (Boehringer Ingelheim),
WO 03/010141 (Boehringer Ingelheim),
WO 03/007945 (Boehringer Ingelheim), • WO 02/100846 A1 and WO 02/100851 A2 (both Shire),
WO 01/85172 A1 and WO 02/098424 A1 (both GSK),
WO 00/06529 and WO 02/06246 A1 (both Merck),
WO 01/47883 and WO 03/000254 (both Japan Tobacco) and
EP 1 256 628 A2 (Agouron). Furthermore other inhibitors of HCV polymerase also include nucleoside analogs, for example, those compounds described in:
• WO 01/90121 A2 (Idenix),
• WO 02/069903 A2 (Biocryst Pharmaceuticals Inc.), and
• WO 02/057287 A2 and WO 02/057425 A2 (both Merck/lsis). Specific examples of inhibitors of an HCV polymerase, include JTK-002/003, JTK- 109 (Japan Tobacco), and NM283 (Idenix).
The term "HIV inhibitor" as used herein means an agents (compound or biological) that is effective to inhibit the formation and/or replication of HIV in a mammal. This includes agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of HIV in a mammal. HIV inhibitors include, for example, nucleosidic inhibitors, non-nucleosidic inhibitors, protease inhibitors, fusion inhibitors and integrase inhibitors. The term "HAV inhibitor" as used herein means an agent (compound or biological) that is effective to inhibit the formation and/or replication of HAV in a mammal. This includes agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of HAV in a mammal. HAV inhibitors include Hepatitis A vaccines, for example, Havrix® (GlaxoSmithKline), VAQTA® (Merck) and Avaxim® (Aventis Pasteur).
The term "HBV inhibitor" as used herein means an agent (compound or biological) that is effective to inhibit the formation and/or replication of HBV in a mammal. This includes agents that interfere with either host or viral mechanisms necessary for the formation and/or replication of HBV in a mammal. HBV inhibitors include, for example, agents that inhibit HBV viral DNA polymerase or HBV vaccines. Specific examples of HBV inhibitors include Lamivudine (Epivir-HBV®), Adefovir Dipivoxil, Entecavir, FTC (Coviracil®), DAPD (DXG), L-FMAU (Clevudine®), AM365 (Amrad), Ldt (Telbivudine), monoval-LdC (Valtorcitabine), ACH-126,443 (L-Fd4C) (Achillion), MCC478 (Eli Lilly), Racivir (RCV), Fluoro-L and D nucleosides, Robustaflavone, ICN 2001-3 (ICN), Bam 205 (Novelos), XTL-001 (XTL), Imino-Sugars (Nonyl-DNJ) (Synergy), HepBzyme; and immunomodulator products such as: interferon alpha 2b, HE2000 (Hollis-Eden), Theradigm (Epimmune), EHT899 (Enzo Biochem), Thymosin alpha-1 (Zadaxin®), HBV DNA vaccine (PowderJect), HBV DNA vaccine (Jefferon Center), HBV antigen (OraGen), BayHep B® (Bayer), Nabi-HB® (Nabi) and Anti- hepatitis B (Cangene); and HBV vaccine products such as the following: Engerix B, Recombivax HB, GenHevac B, Hepacare, Bio-Hep B, TwinRix, Comvax, Hexavac.
The term "class I interferon" as used herein means an interferon selected from a group of interferons that all bind to receptor type I. This includes both naturally and synthetically produced class I interferons. Examples of class I interferons include α-, β-, δ-, omega interferons, tau-interferons, consensus interferons, asialo-interferons.
The term "class II interferon" as used herein means an interferon selected from a group of interferons that all bind to receptor type II. Examples of class II interferons include γ-interferons.
Moreover, the term "therapeutically effective amount" as it is used herein with respect to compounds of Formula I means an amount of the compound which is effective to treat a Flaviviridae viral infection, i.e. to inhibit or at least reduce viral replication, while not being an amount that is toxic to a mammal being treated or an amount that may otherwise cause significant adverse effects in a mammal.
The term "carrier" is used to refer to compounds or mixtures of compounds for combination with the present therapeutic compounds which facilitate the administration thereof and/or enhance the function of the therapeutic compounds to inhibit a virus of the Flaviviridae family including, for example, diluents, excipients, adjuvants and vehicles. Stabilizers, colorants, flavorants, anti-microbial agents, and the like may also be combined with the therapeutic compound. Moreover, in some cases, the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form. Other suitable additives include those with which one of skill in the art would be familiar that function to improve the characteristics of the present combination while not adversely affecting its function. Reference may be made to "Remington's Pharmaceutical Sciences", 17th Ed., Mack Publishing Company, Easton, Penn., 1985, for carriers that would be suitable for admixture with the present therapeutic compounds. The term "pharmaceutically acceptable" as it is used herein with respect to a carrier compound is meant to indicate that the carrier is suitable for administration to a mammal, i.e. is non-toxic and does not cause any adverse effect when used in amounts appropriate to function as a carrier.
Preferred Embodiments Anti-Flaviviridae virus composition
In a first embodiment of the present invention, there is provided a composition effective to treat a mammal infected with a virus of the Flaviviridae family. The composition comprises a pharmaceutically acceptable carrier in combination with a compound of Formula (I):
Figure imgf000014_0001
Formula (I) wherein,
A is selected from: C-i to C6 alkyl and C3 to C6 cycloalkyl; and B is selected from: phenyl orthiazolyl, both of which optionally substituted with a group selected from NH(R1) and NH(CO)R\ wherein R1 is Cj to C6 alkyl; R is OH or a sulfonamide derivative, or a pharmaceutically acceptable salt thereof.
In one embodiment, A of Formula (I) is a branched C4 to C6 alkyl or C4 to C6 cycloalkyl group. In a preferred embodiment, A is cyclopentyl or tert-butyl.
In another embodiment, B of Formula (I) is phenyl or a thiazole substituted at position 2 with NH(R1) or NH(CO) R in which R is a C-, to C4 alkyl. In a preferred embodiment, B is a thiazole substituted at position 2 with NH(R1) or NH(CO) R1 in which R1 is a C-i to C alkyl. More preferably, B is 4-thiazole substituted at position 2 with NH(CO)CH3 or with NHCH(CH3)2.
In a further preferred embodiment, R of formula (I) is OH or a sulfonamide group of formula -NHS02-R2 wherein R2 is -(C1-6)alkyl, -(C3-6)cycloalkyl, both optionally substituted 1 or 2 times with halo or phenyl, or R2 is C6 aryl optionally substituted from 1 or 2 times with halo or (C1-6)alkyl. More preferably, R is OH or a sulfonamide group wherein R2 is methyl, cyclopropyl or phenyl.
In a more preferred embodiment, the present invention is conducted with a compound of Formula (I) in which A is terf-butyl and B is phenyl as set out below in the formula of compound (II):
Figure imgf000015_0001
In another more preferred embodiment, the present invention is conducted with a compound of Formula (I) in which A is tert-butyl and B is 4-thiazole substituted at its 2 position with NH(CO)CH3 as set out below in the formula for compound (III):
Figure imgf000015_0002
In a most preferred embodiment, the present invention is conducted with a compound of Formula (I) in which A is cyclopentyl and B is 4-thiazole substituted at its 2 position with NHCH(CH3)2 as set out below in the formula for compound (IV):
Figure imgf000015_0003
In a most preferred embodiment, the present invention is conducted with a compound of Formula (I) in which A is cyclopentyl, B is 4-thiazole substituted at its 2 position with NHCH(CH3)2, and R is a sulfonamide group wherein R is phenyl as set out below in the formula for compound (VI):
Figure imgf000016_0001
In a most preferred embodiment, the present invention is conducted with a compound of Formula (I) in which A is cyclopentyl, B is 4-thiazole substituted at its 2 position with NHCH(CH3)2, and R is a sulfonamide group wherein R2 is methyl as set out below in the formula for compound (VII):
Figure imgf000016_0002
In a most preferred embodiment, the present invention is conducted with a compound of Formula (I) in which A is cyclopentyl, B is 4-thiazole substituted at its 2 position with NHCH(CH3)2, and R is a sulfonamide group wherein R2 is cyclopropyl as set out below in the formula for compound (VIII):
Figure imgf000016_0003
Method of Treatment
In a second aspect of the present invention, a method for treating a mammal infected with a virus of the Flaviviridae family is provided comprising administering to an infected mammal a pharmaceutical composition comprising a pharmaceutically acceptable carrier in combination with a therapeutically effective amount of a compound having Formula (I), (II), (III), (IV), (VI), (VII) or (VIII) as defined above.
In accordance with the method of the present invention, a therapeutically effective amount of a compound of Formula (I), (II), (III), (IV), (VI), (VII) or (VIII) is administered to a mammal infected with a Flaviviridae virus. To be therapeutically effective, a dosage of between about 0.01 and about 100mg/kg body weight per day, preferably between about 0.1 and about 50mg/kg body weight per day of the compound is administered to the infected mammal. Typically, the method will involve administration of the compound from about 1 to about 5 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy.
As one of skill in the art will appreciate, lower or higher doses than those recited above may be required. Specific dosage and treatment regimens will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health status, sex and diet of the infected mammal, the time of administration, the rate of excretion, the severity and course of the infection, the patient's disposition to the infection and the judgment of the treating physician. Generally, treatment is initiated with small dosages substantially less than the optimum dose of the peptide. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached. In general, the compound is most desirably administered at a concentration level that will generally afford a therapeutic effect without causing any harmful or deleterious side effects.
Administration of the therapeutic compound in the treatment of Flaviviridae viral infection may be by any one of several routes including administration orally, parenterally or via an implanted reservoir. The term "parenteral" as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, and intralesional injection or infusion techniques. Oral administration or administration by injection are preferred. Orally acceptable dosage forms include, but not limited to, capsules, tablets, and aqueous suspensions and solutions.
As set out above, the compound of Formula (I), (II), (III), (IV), (VI), (VII) or (VIII) is administered in combination with one or more pharmaceutically acceptable carriers. The nature of the carrier(s) will, of course, vary with the dosage form. Accordingly, in the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added. Injectable preparations including injectable aqueous or oleaginous suspensions, are formulated according to techniques known in the art using suitable dispersing or wetting agents such as Tween 80 and suspending agents.
The amount of the present therapeutic compound that is combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. A typical preparation will contain from about 5% to about 95% therapeutic compound (w/w). Preferably, such preparations contain from about 20% to about 80% therapeutic compound.
In another aspect, a combination therapy is contemplated wherein the compound of Formula (I), (II), (III), (IV), (VI), (VII) or (VIII), or a pharmaceutically acceptable salt thereof, is co-administered with at least one additional agent selected from: an antiviral agent, an immunomodulatory agent, an HCV inhibitor, an HIV inhibitor, an HAV inhibitor, an HBV inhibitor, and a therapeutic effective to treat the symptoms of the viral infection. Examples of such agents are well known to those of skill in the art. These additional agents may be combined with the compounds of this invention to create a single pharmaceutical dosage form. Alternatively these additional agents may be separately administered to the infected patient as part of a multiple dosage, form, for example, using a kit. Such additional agents may be administered to the patient prior to, concurrently with, or following the administration of the therapeutic compound of Formula (1), (II), (III), (IV), (VI), (VII) or (VIII), or a pharmaceutically acceptable salt thereof.
When a combination therapy is utilized, both the present compound and the additional therapeutic and/or prophylactic agents should be present at dosage levels of between about 10 to 100%, and more preferably between about 10 and 80% of the dosage normally administered in a monotherapy regimen.
Article of Manufacture In a further aspect of the present invention, there is provided an article of manufacture comprising packaging material contained within which is a composition effective to treat a mammal infected with a virus of the Flaviviridae family and the packaging material comprises a label which indicates that the composition can be used to treat infection by a virus of the Flaviviridae family, wherein said composition comprises a compound of Formula (I), (II), (111), (IV), (VI), (VII) or (VIII) as defined above.
Flaviviridae viruses
Particularly, the different embodiments of the present invention may be directed to distinct viruses, particularly viruses which are pathogenic in mammal. Preferably, the above-mentioned compounds of compositions can be used for the treatment of hepacivirus genus, such as Hepatitis C. Preferred examples of HCV viruses are selected from genotypes: 1 , 2, 3, 4, 5, and 6. More preferred examples of HCV viruses are selected from genotypes: 2, 3, 4, 5, and 6. Preferably, HCV viruses are selected from subtypes: HCV 1 a, HCV1 b, HCV 2a-c, HCV 3a-b, HCV 4a, HCV 5 and HCV 6a,h,d & k. More preferably, HCV viruses are selected from subtypes: HCV 1a, HCV 2a-c, HCV 3a-b, HCV 4a, HCV 5 and HCV 6a,h,d & k.
Alternatively, the compounds of the invention may be directed against viruses of the flavivirus genus, such as the Dengue Fever viruses, Japanese Encephalitis viruses, West Nile viruses and Yellow Fever viruses. Preferably, the invention is directed at the treatment of viral disease in a human caused by Dengue virus. Preferably, the invention is directed at the treatment of viral disease in a human caused by Japanese encephalitis virus. Alternatively, the invention is directed at the treatment of viral disease in a human caused by Yellow Fever virus. Alternatively, the invention is directed at the treatment of viral disease in a human caused by West Nile virus.
In addition, the invention may be directed to the treatment of viral disease caused by viruses of the pestivirus genus, such as the bovine viral diarrhea virus (BVDV), classical swine fever virus (CSFV) and border disease virus (BDV). Preferably, the invention is directed at the treatment of viral diseases in cattle caused by BVDV. Alternatively, the invention is directed at the treatment of viral diseases in pigs caused by CSFV. Alternatively, the invention is directed at the treatment of viral diseases in sheep caused by BDV.
Furthermore, GB viruses can also be treated with composition of the present invention. Included in this viral family are Hepatitis G virus (HGV) and Hepatitis GB virus. Preferred examples are selected from: GBV-A, B & C. Preferably, the invention is directed at the treatment of viral disease in a human caused by GBV-C. Alternatively, the invention is directed at the treatment of viral disease in a human caused by GBV-A. Alternatively, the invention is directed at the treatment of viral disease in a human caused by HGV.
, Embodiments of the present invention are exemplified by the following specific examples which are not to be construed as limiting.
EXAMPLES
Abbreviations used in the examples include:
Abu: aminobutyric acid; DABCYL: 4-((4-(dimethylamino)phenyl) azo)benzoic acid; DMEM: Dulbecco's Modified Eagle Medium; DMSO: dimethyl sulfoxide; EDANS: 5- ((2-aminoethyl)amino)naphthalene-1-sulfonic acid; HPLC: high performance liquid chromatography; IPTG: isopropyl-b-D-thiogalactoside; LB: Luria-Bertoni (as in LB broth); Nva: norvaline; PenStrep: penicillin/streptomycin; PCR: polymerase chain reaction; r.m.s.: root mean square; RT-PCR: real-time polymerase chain reaction; and TCEP: tris(2-carboxyethyl)phosphine hydrochloride.
Synthesis of Compounds of Formula (I)
Compounds of Formula (I) were prepared using the protocol outlined in detail in WO 00/059929, published October 12, 2000. In particular, reference is made to page 89, Example 34C for the preparation of compound (IV). Example 1 - Inhibition of HCV NS3-NS4A proteases of different genotypes by compounds If, III and IV
HCV NS3/4A 1a and 1b For production of the HCV genotype 1 b NS3-NS4A heterodimer protein, a full-length HCV cDNA was cloned by RT-PCR using RNA extracted from the serum of an HCV genotype 1b infected individual (provided by Dr. Bernard Willems, Hδpital St-Luc, Montreal, Canada). The DNA region encoding the NS3-NS4A heterodimer protein was PCR-amplified (forward primer: 'CTCGGATCCGGCGCCCATCACGGCCTAC3' (SEQ ID No.1) ; reverse primer: 5'CTCTCTAGATCAGCACTCTTCCATTTCAT CGAA3') (SEQ ID No.2)) from the full-length HCV cDNA and subcloned into the pFastBac™ HTa baculovirus expression vector (Gibco/BRL). For HCV genotype 1a, the DNA encoding NS3-NS4A heterodimer protein was PCR-amplified (forward primer: 5'CTCTCTAGATCAGCACTCTTCCATTTCATCGAACTC3' (SEQ ID No.3); reverse primer: 5OTCGGATCCGGCGCCCATCACGGCCTACTCCCAA3' (SEQ ID No.4)) from the HCV genotype 1a strain H77 (provided by ViroPharma Inc., Exton, PA, US) and subcloned as described above. The cloning into the pFastBac™ HTa baculovirus expression vector generated a recombinant fusion protein containing an additional N-terminal 28 residues that comprised a hexahistidine tag and a rTEV protease cleavage site. The Bac-to-Bac™ baculovirus expression system (Gibco/BRL) was used to produce the recombinant baculovirus for protein expression.
His-tagged NS3-NS4A heterodimer protease was expressed by infecting Sf21 insect cells (lnvitrogen) at a density of 106 cells/mL with the recombinant baculovirus at a multiplicity of infection of 0.1-0.2 at 27°C. The infected culture was harvested 48 to 64 h later by centrifugation at 4°C. The cell pellet was homogenized in 50 mM sodium phosphate, pH 7.5, 40% glycerol (w/v), 2mM β-mercaptoethanol. His-NS3- NS4A heterodimer protease was then extracted from the cell lysate with 1.5% NP- 40, 0.5% Triton X-100, 0.5M NaCI, and a DNase treatment. After ultracentrifugation (100,000xg for 30 min at 4°C), the soluble extract was diluted 4-fold in 50 mM sodium phosphate, pH 7.5, 0.5M NaCI and loaded on a Pharmacia Hi-Trap Ni+2- chelating column. The His-NS3-NS4A heterodimer protein was eluted using a 50 to 400 mM imidazole gradient prepared in 50 mM sodium phosphate, pH 7.5, 10% (w/v) glycerol, 0.1% NP-40, 0.5M NaCI. Co-purification of mature NS3 and NS4A protein complex was verified by Western blot analysis of the purified proteins using an anti-NS3 and an anti-NS4A antiserum produced in-house. The purified NS3 protease domain and the peptide H2N-PDREVLYREFDEMEEC-OH (SEQ ID No. 5 ) were used to immunize rabbits for the production of antisera specific to NS3 and NS4A respectively. The proper N-terminal amino acid of both proteins was confirmed by N-terminal amino acid sequencing (PE Biosystems 491/491 C Procise Sequencer). The purified enzymes were stored at -80°C in 50 mM sodium phosphate, pH 7.5, 10% (w/v) glycerol, 0.5 M NaCI, 0.25 M imidazole, 0.1% NP-40.
HCV NS3/4A 2b and 3a
The NS3-NS4 protease genes of HCV genotypes 2b and 3a were amplified from RNA isolated from clinical samples of HCV-infected patients obtained from Dr. G. Steinmann (Germany) using RT-PCR. The primers used for the RT- PCR were 5OTCGGATCCGGCTCCCATTACTGCTTAC3' (SEQ ID No. 6) as the forward and 5'GACGCGTCGACGCGGCCGCTCAGCACTCTTCCATTTCACTGAA3' (SEQ ID No.7) as the reverse primer for the NS3-NS4A of HCV genotype 2b and; 5OTCGGATCGGGCCCCGATCACAGCATACGCC3' (SEQ ID No. 8) as the forward and 5OACCGCTCGAGTCAGCATTCTTCCATCTCATCATATTGTTG3' (SEQ ID No. 9) as the reverse primer for HCV genotype 3a . Each primer contained a unique restriction site for subcloning the fragment in the bacterial expression vector pET11a. The cloning into the vector generated a recombinant fusion protein containing an additional N-terminal 28 residues that comprised a hexahistidine tag and a rTEV protease cleavage site. The recombinant plasmids were used to transform the bacterial strain BL21 DE3 pLysS for protein expression and recombinant protein expression was induced by the addition of IPTG. Following expression, the cells were collected by centrifugation at 4°C and the cell pellet lysed in a buffer containing 50mM sodium phosphate, 0.5M NaCI, 40% glycerol, 1.5% NP-40, 0.5% Triton X- 100 and the soluble protein fraction was passed through a 5ml HiTrap chelating affinity column as described above. A poly(U)-Sepharose purification step can optionally be conducted in order to remove degradation products and contaminants.
In Vitro HCV S3 Protease Assay
The fluorogenic depsipeptide substrate used to assess inhibition of protease activity of the HCV 1 a, 1 b, 2b, 2a-c and 3a NS3-NS4A heterodimer protein was anthranilyl- DDlVPAbu[C(0)-0]-AMY(3-N02)TW-OH (SEQ ID No.10). This substrate is cleaved between the aminobutyric (Abu) and the alanine residues. The sequence DDIVPAbu-AMYTW (SEQ ID No.11) is derived from the sequence DDIVPC- SMSYTW (SEQ ID No.12) corresponding to the NS5A/NS5B natural cleavage site. The introduction of the aminobutyric residue at position P1 resulted in a significant decrease of the N-terminal product inhibition while the deletion of the serine residue at position P3 improved the internal quenching efficiency. The protease activity was assayed in 50 mM Tris-HCI, pH 8.0, 0.25M sodium citrate, 0.01% n-dodecyl-β-D- maltoside, 1 mM TCEP. The assay was performed in a Microfluor®2 White U- Bottom Microtiter® plate. 5 μM of the substrate and various concentrations of the test compound were incubated with 1.5 nM of HCV 1a or 1b NS3-NS4A heterodimer protein for 45 minutes at 23°C under gentle agitation. The final DMSO content did not exceed 5.25%. The reaction was terminated by the addition of a 1 M 2-[N- morpholinojethanesulfonic acid solution at pH 5.8.
The fluorescence was monitored using the BMG POLARstar Galaxy 96-well plate reader with an excitation filter of 320 nm, and an emission filter of 405 nm. The level of inhibition (% inhibition) of each well containing test compound was calculated with the following equation (FU = fluorescence unit):
FU well - FU blank % • inhibition = 1 - * 100. FU • control - FU • blank
The calculated % inhibition values were then used to determine IC5o, slope factor (n) and maximum inhibition (lmax) by the non-linear regression routine NLIN procedure of SAS using the following equation:
% inhibition = I- X [inhmtθr]n .
[inhibitor]" + ICso"
Inhibition constant (Ki) determination and mode of inhibition The fluorogenic depsipeptide substrate anthranilyl-D(d)EIVP-Nva[C(0)-0] AMY(3- N02)TW-OH (SEQ ID No.13) was used to assess the mechanism of inhibition and the inhibition constants of the compound (IV) against HCV NS3-NS4A proteases. It was cleaved between the norvaline and the alanine residues. The substrate working solution was prepared in DMSO at the concentration of 200 μM from the substrate stock solution (2 mM in DMSO stored at -20°C). The final substrate concentration in the assay varied from 0.25 to 8 μM. The inhibition constant (Ki) for compound (IV) was determined using a steady-state velocity method (Morrisson etal. 1985). The protease activity was determined by monitoring the fluorescence change associated with the cleavage of the internally quenched fluorogenic substrate using a SLM- AMINCO® 8100 spectrofluorometer (emission at 325 nm and excitation at 420 nm). The cleavage reaction was monitored in the presence of 0.25 to 8 μM of substrate, 0.3 nM of HCV 1a or 1b NS3-NS4A heterodimer protein and various concentrations of the test compound IV in 50 mM Tris-HCI, pH 8.0, 0.25M sodium citrate, 0.01 % n- dodecyl-β-D-maltoside, 1 mM TCEP. The steady-state analysis of inhibition of the HCV genotype 1a and 1b NS3-NS4A heterodimer proteases were performed using the Dixon and Cornish-Bowden graphical methods. In the Dixon graphical method, the reciprocal velocity 1/V is plotted against the test compound concentration at several substrate concentrations (S). In the Cornish-Bowden graphical method, the ratio S/V is plotted against the test compound concentration at several S concentrations. For both methods, the points lie on a straight line at each S value. For a competitive test compound, the Dixon plot displays lines at different S values intersecting at a single point, while the Cornish-Bowden plot displays parallel lines at different S values. The Kj was estimated by fitting the data to the equation describing competitive binding:
Figure imgf000024_0001
Inhibition of HCV NS3 proteases
The activity of HCV genotype 1a, 1b, 2b, 2a-c and 3a NS3/NS4A heterodimer proteases was determined in the presence of compound (IV) using the in vitro enzymatic fluorogenic assay. The IC50 curves were analyzed individually by the SAS NLIN procedure. For HCV 1a NS3-NS4A protease, an average IC50 value of 4.3 nM was obtained from the analysis of two batches of compound (IV). An average IC50 value of 3.4 nM was obtained from the analysis of two batches of compound (IV) in the presence of HCV 1 b NS3-NS4A protease. TABLE 1
IC5o of different NS3 protease inhibitors in HCV of different genotypes
Figure imgf000025_0001
n/d= not done
Figures 3A and 3B illustrate the IC50 curves of compound (IV) against HCV 1a and 1b NS3-NS4A proteases, respectively.
Compound (IV) was found to be a competitive test compound of HCV genotype 1a and 1b NS3-NS4A proteases following fitting of the data to the equation describing competitive binding with GraFit and also from the Dixon arid Cornish-Bowden graphical methods. For both HCV genotype enzymes, the Dixon plot showed that the lines at different S values were intersecting at a single point, while the Cornish- Bowden plot showed parallel lines at various S values. A Kj of 0.30 nM was obtained for compound (IV) from steady-state velocity analysis with the HCV genotype 1a NS3-NS4A protease, while a Kf of 0.66 nM was obtained with the HCV genotype 1b NS3-NS4A protease, K|'s of 90 nM, 86 nM and 83 nM with HCV genotype 3a, 2a-c and 2b respectively. Figures 4 and 5 are the Dixon and Cornish-Bowden plots of compound (IV) against the HCV genotype 1a and 1b NS3-NS4A, proteases respectively.
Example 2 - Inhibition of HCV Replicon 1a and/or 1b
Cell-based HCV 1a and 1b RNA replicon assays
HCV RNA replication was demonstrated in HCV 1a and 1b replicon-containing, Huh- 7-derived cell lines developed at Boehringer Ingelheim (Canada) Ltd., R & D (WO 02/052015). These cells were used to establish a sensitive HCV RNA replication cell-based assay for testing candidate test compounds.
Huh7 cells that stably maintain a subgenomic HCV replicon were established as previously described (Lohman et al. 1999; WO 02/052015). The cells were seeded into a 96 well cell culture cluster at 1 X 104 cells per well in DMEM complemented with 10% FBS, PenStrep (Life Technologies) and 1μg/mL Geneticin. Cells were incubated in a 5% C02 incubator at 37 °C until addition of various concentrations of the test compound.
The test compound was prepared for use in the assay as follows. The test compound in 100% DMSO was added diluted in assay Medium for a final DMSO concentration of 0.5% and the solution was sonicated for 15 min and filtered through a 0.22μM Millipore Filter Unit. Serial dilutions of the test compounds were prepared using Assay Medium (containing 0.5% DMSO).
Cell culture medium was aspirated from the 96-well plate containing the cells and the appropriate dilution of test compound in assay medium was transferred to independent wells of the cell culture plate which was incubated at 37° with 5% C02.
Following a 3 day-incubation period, the cells were washed with PBS and total cellular RNA was extracted with the RNeasy Mini Kit® and Qiashredder® from Qiagen. RNA from each well was eluted in 50μL of H20. RNA was quantified by optical density at 260 nm on the Cary 1 E® UV-Visible spectrophotometer. The HCV RNA replicon copy number was evaluated by real-time RT-PCR with the Abi Prism® 7700 Sequence Detection System. The TAQMAN EZ® RT-PCR kit provides a system for the detection and analysis of RNA. Direct detection of the reverse transcription polymerase chain reaction (RT-PCR) product with no downstream processing was accomplished by monitoring the increase in fluorescence of a dye- labeled DNA probe. The nucleotide sequence of both primers and probe was located in the 5' region of the HCV genome. The replicon copy number was then evaluated using a standard curve made with known amounts of replicon copy (supplemented with 50 ng of wild type Huh-7 RNA) assayed in the same reaction mix. The following thermal cycle parameters were used for the RT-PCR reaction on the ABI Prism® 7700 Sequence Detection System. Conditions were optimized for • HCV detection. Quantification is based on the threshold cycle, where the amplification plot crosses a defined fluorescence threshold. Comparison of the threshold cycles provides a highly sensitive measure of relative template concentration in different samples. Monitoring during early cycles, when PCR fidelity is at its highest, provides precise data for accurate quantification. The relative template concentration can be converted to real numbers by using the standard curve of HCV with known number of copy.
ECfin Curve Fitting with NLIN Procedure of SAS
The level of inhibition (% inhibition) of each well containing test compound was calculated with the following equation (CN = HCV Replicon copy number):
CN • control - CN well
% • inhibition ■ : 100 . CN • control
The calculated % inhibition values were then used to determine EC50, slope factor (n) and maximum inhibition (lmaχ) by the non-linear regression routine NLIN procedure of SAS using the following equation:
% • inhibition =
[inhibitor]" + ECs "
TABLE 2 EC50 (nM) of different NS3 protease inhibitors in HCV of different genotypes
Figure imgf000027_0001
In HCV replicon cell-based assays, compound (IV) showed a dose-dependent inhibition of HCV RNA replication with EC50s of 2.1 nM and 1 nM using HCV replicon-containing cells of genotype 1a and 1b, respectively. Cytotoxicity was not observed at concentrations higher than 1000 nM. These results indicate that compounds II, III, IV, VI, VII and VIII are potent and specific inhibitors of cell-based HCV RNA replication. Compound Vis a compound from a different class but which is related in structure, and also active against NS3 protease but less potent. This compound will be used later on as a control to compare inhibitory activity levels.
Compounds VI, VII and VIII have the following structures:
Figure imgf000028_0001
compound (VI)
Figure imgf000028_0002
compound (VII)
Figure imgf000028_0003
compound (VIII)
Example 3 - Inhibition of GBV-B NS3/4A protease by compounds II, III and IV
Cloning of GBV-B NS3/NS4A protease
The full length GBV-B NS3/NS4A protease gene was isolated from infected tamarin serum. Total RNA was isolated from the serum using Qiagen® viral RNA extraction kit according to standard protocol. The selection of primers for the reverse transcriptase and the PCR reactions were selected based on the published sequence (Muerhoff, A.S et al. 1995) and GeneBank accession number U22304 (forward: 5OGCATATGGCACCTTTTACGCTGCAGTGTC3' (SEQ ID No.14); reverse: 5'CGCGCGCTCGAGACACTCCTCCACGATTTCTTC3' (SEQ ID No.15)).
The amplified RT-PCR product was cloned into the polyhistidine tag-containing pET- 29 plasmid between the Ndel and Xhol sites in frame with the polyhistidine tag. This construct produced a recombinant protein with a poly His tag at its C-terminus. The plasmid was transformed into BL21 DE3 pLysS for protein expression.
The E. coli clone pGBV-B was grown in LB medium to a cell density (OD600) of 0.6 at which time IPTG was added at a concentration of 0.2 mM. The induction was done at 23 C for a period of 4 hours. The GBV-B NS3/NS4A-His protein was purified according to the procedure described by Zhong et al., 1999.. The soluble fraction was purified on a Pharmacia® Hi-Trap Ni+2-chelating affinity column using a 50 to 400 mM imidazole gradient. This step was followed by chromatography of the protein preparation on a Superdex® 200 gel filtration column. The concentration of the protein preparation was determined by Bradford protein assay.
In vitro GBV-B NS3 protease assay
The depsipeptide substrate used to assess inhibition of the protease activity of GBV-
B NS3-NS4A heterodimer protein was Ac-DED(EDANS)EE-Abu[C(0)-0] ASK(DABCYL)-NH2 (SEQ ID No. 16) This substrate is cleaved between the aminobutyric (Abu) and the alanine residues and products of the reaction were analyzed by HPLC. The protease activity was assayed in 50 mM Tris-HCI, pH 8.0, 0.25M sodium citrate, 0.01% n-dodecyl-β-D-maltoside, 1 mM TCEP. The assay was performed in a Microfluor®2 White U-Bottom Microtiter® plate. 5 μM of the substrate and various concentrations of the test compounds were incubated with 0.4 nM of GBV-B NS3-NS4A heterodimer protein for 2 hours at 23°C under gentle agitation. The final DMSO content did not exceed 5.25%. The reaction was terminated by the addition of a 1 M 2-[N-morpholino]ethanesulfonic acid solution at pH 5.8. For quantification, the cleavage products and the substrate were separated by HPLC on a Perkin-Elmer® 3x3CR C8 column. The separation was accomplished by initially eluting at 3.5 mL/min with an aqueous solution containing 0.05% phosphoric acid and 3 mM SDS. A 0 to 45% acetonitrile linear gradient in 0.05% phosphoric acid was then applied for 10 minutes. The absorbance was monitored at 210 nm. The assay was conducted in the presence of the test compounds II, III, IV and V. The IC50 obtained for each test compound in the presence of GBV-B NS3-4A protein is indicated in Table 3.
TABLE 3
Figure imgf000030_0001
Compound Vis a compound from a different class which is related in structure and also active against NS3 protease but less active than compounds II, III and IV. This compound was included to demonstrate that the ranking of the activity of compounds II, III and IV was maintained between HCV and GBV-B.
Inhibition of GBV-B replication in tamarin hepatocvtes in culture
Hepatocytes isolated from uninfected tamarins were maintained in culture in defined medium for several days. The cell culture model was as described by Beames et al. 2000. Three days after plating, the cells were infected with GBV-B-containing plasma. After viral adsorption, the virus was removed and the cells were incubated in the presence and absence of candidate test compounds at a concentration of 10 μM. The cells were harvested 7 days post infection. The levels of GBV-B RNA were quantitated from total cellular RNA by a real-time PCR assay using a primer-probe combination that recognized a portion of the GBV-B capsid gene (Beames etal. 2000).
Despite lower IC50 obtained in the enzymatic assay, the results illustrated in Figure 6, show that more than a 3-log reduction in the viral RNA levels was observed when the cells were incubated in the presence of compound III and a 2-log reduction in the presence of compound V(when the results were expressed as g.e. (genome equivalents)/ μg RNA). The results obtained with this more representative viral replication assay demonstrate that these compounds are effective antivirals to reduce GBV-B virus production. Example 4 - Sequence Comparison of NS3 proteases within the Flaviviridae family
The sequences of proteases from other viruses of the flaviviridae family of viruses were obtained from a BLAST analysis (Altschul et al.1997) NCBI followed by the taxonomy report. The search for protease sequences was done using specific criteria that would retrieve all flaviviridae sequences with similarity to HCV protease excluding HCV sequences themselves. When possible an "NP_" sequence from NCBI was used to represent a particular group of related viruses since these "NP_" sequences are "Reference" sequences (RefSeq). Furthermore, when not known, the NS3 N-terminal and C-terminal ends for the proteases were determined from similarity with HCV NS3 protease.
Sequence alignments of proteases for the six HCV strains against the different groups of viruses were performed using ALIGNX (VectorNTI®) based on CLUSTALW (Thompson, JD, et al., 1994).
From these alignments, the percentage of similarity was calculated between all individual sequences and expressed in Tables 4-8. The similarity was based on the following grouping of amino acids; [ILVM], [FWY], [KR], [DE], [GA], [NQ], [ST].
Table 4 represents identities and similarities between several genotypes and subtypes of HCV. One can see that identities range between 70% and 89% whereas similarities range between 77% and 95% demonstrating that the NS3 protease is highly conserved among HCV. Also, from the amino acid sequence alignment of NS3 protease domain of various representative HCV genotypes (Figure 2) and location of the conserved residues on the three dimensional structure (Figure 8), one can see that the residues at the active site are highly conserved among HCV genotypes and subtypes.
Table 5 is an analysis of the similarity between GB viruses NS3 protease and HCV NS3 protease domains. Similarities range between 43 and 49%, whereas similarities between HCV and pestivirus NS3 domains range from 25 and 30% (Table 6). Similarities between HCV and dengue virus range also around 30% (Table 7), as well as the similarity between HCV and Flavivirus (Table 8). In contrast, Table 9 shows that the similarities between the NS3 protease domain of HCV and the protease of Human Cytomegalovirus (HCMV) revolves around 20%. HCMV is a virus of the Herpes viridae and has a serine protease that has a different catalytic triad from Flaviviridae proteases. The HCMV virus protease is therefore not considered to be similar to Flaviviridae NS3 proteases. This contrast will be illustrated further when comparison is made of the similarities between the amino acid in contact with our inhibitors in Flaviviridae and HCMV.
Example 5 - Three-Dimensional Structural Crystal Studies In order to analyze further the similarities between Flaviviridae, a crystal structure was obtained of the HCV NS3 protease complexed with an inhibitor, and the amino acids directly in contact with the inhibitor were analyzed in other Flaviviridae to assess degrees of similarity of these important contact points.
A co-crystal of the HCV NS3 protease-NS4A peptide complexed with macrocyclic compound (II) was obtained that diffracted X-rays to 2.75 A resolution. The macrocyclic compound II was found at the NS3 protease active site and was clearly defined in the difference electron density. This structure reveals molecular details of how the test compound interacts with the active site and provides additional insight into the mechanism of inhibition of the HCV NS3 protease.
The structure of the NS3 protease complex with the test compound II (Figure 8) better defines the binding site of the present substrate-based test compound series. From this structure of the co-complex, the residues directly in contact with the test compound i.e. within 3 A (H57, G137, S139, A156 and A157) are indicative of the active site, as predicted by the competitive mode of inhibition, and by the conservation at this site among representative sequences of HCV genotypes and various subtypes.
Other residues in contact within 4A of the test compound are listed in Table 10. Comparison analysis reveals that these amino acids are highly conserved among Flaviviridae ranging between 59% and 76%. Conservation of these residues among Flaviviridae is indicative that inhibitors of the HCV NS3 protease would also inhibit other members of the Flaviviridae family of viruses. In contrast, the level of similarity of the HCMV protease catalytic site and the contact points of Table 10 is in the same range as the similarity for the whole protease (-20%) once again illustrating the fact that other serine protease from non-Flaviviridae are not targeted by the compounds of formula (I).
Other structural studies performed between HCV and dengue virus protease domains have shown that all six strands of the C-terminal domain inhibitor binding site are strongly conserved and of comparable length. Superposition of 68 α carbon atoms of the C-terminal domain of Den2 protease (residues 87-167) and HCV including most of the residues in Table 10 (residues 69-189) yields a r.m.s. deviation of 0.9A. (Murthy et at. , 1999).
Discussion
Viruses within the Flaviviridae family possess a number of similarities (Figure 1 ) despite a relatively low overall sequence homology among its members (Tables 4-8). In particular, the NS3 protein contains sequences with similarity to the protease and the nucleoside triphosphate-binding helicase of pestiviruses and flaviviruses. The HCV NS3 protease domain shares a sequence similarity of about 77-95% among HCV genotypes (Figure 1) and a sequence similarity of about 25-50% with other members of the Flaviviridae family (Tables 4-8). In addition, the genomic organization and structure of GBV-B and HCV are similar despite the fact that the sequence homology between the polyprotein sequences of GBV-B and HCV is about 25 to 30%. This low similarity is however trumped by the fact that residues that come in contact with the inhibitor are highly conserved within the protease domain of Flaviviridae, a strong indication that this family of compounds would also bind to other Flaviviridae. The activity of compound IV obtained against GBV-B is a positive demonstration of that hypothesis.
With respect to the three dimensional structure, the available atomic co-ordinates of the various crystallized HCV and Dengue NS3 proteases showed an overall architecture that is characteristic of the trypsin-like fold. Many studies have now firmly established that the N-terminal portion of the NS3 region encodes a serine protease that has a very specific and pivotal role in viral polyprotein processing within Flaviviridae.
All results produced in the above experiments tend to support the contention that the compounds of formula (I) are active against members of the Flaviviridae family of viruses:
• 6 related compounds have been shown to be active against HCV 1 b and have similar activity against HCV 1 a (Table 2); • 3 of these compounds were also tested against HCV 3a, 2a-c and 2b and also found to be active (Table 1);
• these 3 same compounds were tested in the enzymatic assay against GBV-B and found to be active (Table 3);
• 1 of these compounds was tested in cell culture against replication of GBV-B in tamarin cells and found to be active (Figure 6);
• 1 of these compounds demonstrates a mode of binding that is situated in a region highly conserved between all members of Flaviviridae (Figure 8);
• studies have demonstrated strong functional and structural similarity between HCV and Dengue virus NS3 proteases.
Given all similarities of viruses within the Flaviviridae family of viruses, it seems highly likely that the compounds of Formula (I) are effective against members of the Flaviviridae family of viruses, and more particularly, effective against the pathogenic members of the Flaviviridae family.
REFERENCES
- Altschul S.F. et al. 1997, Nucleic Acids Res. 25, 3389-3402
- Ausubel et al. 1994, Current Protocols in Molecular Biology, Wiley, New York
- Beames, B. et al. 2000, J. Virol., 74, 11764-11772 - Butriewicz, N. et al. 2000, J. Virol., 74, 4291-4301
- Kim JL, et al. 1996, Cell;87:343-355.
- Lin, C. et al. 1995, J. Virol. 69, 4373-4380
- Lohmann etal. 1999, Science 285, 110
- Love R. A. et al. 1996 Cell 87, 331-342 - Morrison, J.F. and Stone, S.R. 1985, Mol. Cell. Biophys. 2, 347-368
- Muerhoff, A.S. etal. 1995 J. Virol.69, 5621-5630
- Murthy, H. M. et al. 1999, J. Biol. Chem. 274(9), 5573-5580
- Remington, 1995, The Science and Practice of Pharmacy
- Ryan, M. D. et al. 1998 J. Gen. Virol. 79, 947-959 - Sambrook et al. 1989, Molecular Cloning- A Laboratory Manual, Cold Spring Harbor Labs
- Scarselli, E., etal.1997, J. Virol.71, 4985-4989
- Steinkuler, C. etal.1996, J. Biol. Chem.271, 6367-6373.
- Thompson, et al.1994, Nucleic Acids Res.22: 4673- 4680 - Zhong, W. etal.1999, Virology 261, 216-226
TABLE 4 - IDENTITIES AND SIMILARITIES BETWEEN NS3 PROTEASE
ω 4-.
Figure imgf000036_0002
Sim arity score was o taine defining the fo lowing amino acids similarity: [ILVM], [FWY], [KR], [DE], £GA], [NQJ, [ST] domains (180aa) of the different HCV genotypes and subtypes
Figure imgf000036_0001
TABLE 5 - % SIMILARITY OF HCV NS3 PROTEASE DOMAIN ( 181 AA) WITH GB NS3
PROTEASES FROM THE FLAVIVIRIDAE FAMILY
Figure imgf000037_0001
TABLE 6 - /o SIMILARITY OF HCV NS3 PROTEASE DOMAIN ( 181 A) WITH PESTIVIRUSES
NS3 PROTEASES FROM THE FLAVIVIRIDAE FAMILY
Figure imgf000037_0002
BVDV-2= Bovine viral Diarrhea virus TABLE 7 - /> SIMILARITY OF HCV NS3 PROTEASE DOMAIN ( 181 AA) WITH MOSQUITO -BORNE
FLAVIVIRUS NS3 PROTEASES FROM THE FLAVIVIRIDAE FAMILY
Figure imgf000038_0001
TABLE 8 - /o SIMILARITY OF HCV NS3 PROTEASE DOMAIN (181 AA) WITH FLAVIVIRUS NS3 PROTEASES FROM THE FLAVIVIRIDAE FAMILY
O -4
Figure imgf000039_0002
WVN= West Nile virus
JEV= Japanese encephalitis virus
Kunjin= Kunjin virus
YFV= Yellow Fever virus
MVEV= Murray Valley encephalitis virus
Figure imgf000039_0001
TABLE 9 - % SIMILARITY OF HCV PROTEASE DOMAIN (181 AA) WITH HUMAN
CYTOMEGALOVIRUS PROTEASE DOMAIN (256 AA)
Figure imgf000040_0001
TABLE 10 - AMINO ACID DIFFERENCES IN THE NS3 PROTEASE INHIBITOR BINDING DOMAIN AMONG VIRUSES OF THE
(WITHIN 4 A OF THE INHIBITOR) FLAVIVIRIDAE FAMILY
Figure imgf000041_0001
HCV40_1 b : Sequence of a HCV 1 b isolate that was used as reference for this analysis n : number of HCV sequences of all genotypes that was used in the study
A„ : number of HCV sequences containing this modification
% Sim : % similarity
+ : Similarity according to Matrix [ILVM], [FWY], [KR], [DE], [GA], [NQ], [Sη
= : Similarities that are unique for the GBV-B member of the GBV group
GBVa : The aa in bold are the residue found for GBV-B NS3 protease : % similarity for GBV-B

Claims

WE CLAIM:
1. Use of a compound of Formula (I):
Figure imgf000042_0001
Formula (I) wherein A is selected from: C-i to C6 alkyl and C3 to C6 cycloalkyl; B is selected from: phenyl or thiazolyl, both of which optionally substituted with a group selected from NH(R1) and NH(CO)R1, wherein R1 is C, to C6 alkyl; and R is OH or a sulfonamide group of the formula -NHS02-R2 wherein R2 is -(C1-8)alkyl, -(C3- )cycloalkyl or {-(C1-6)alkyl-(C3.6)cycloalkyl}, which are all optionally substituted from 1 to 3 times with halo, cyano, nitro, 0-(C1-6)alkyl, amido, amino or phenyl, or R2 is C6 or C10 aryl which is optionally substituted from 1 to 3 times with halo, cyano, nitro, (C1-6)alkyl, 0-(C1-6)alkyl, amido, amino or phenyl; or a pharmaceutically acceptable salt thereof; for the preparation of a medicament for the treatment of Flaviviridae viral infection in a mammal.
2. The use according to claim 1 , wherein A of Formula (I) is a branched C4 to C6 alkyl or C to C6 cycloalkyl group; B of Formula (I) is phenyl or a thiazole substituted at position 2 with NH(R1) or NH(CO) R1 in which R1 is a C-j to C4 alkyl; and R is OH or a sulfonamide group of formula -NHS02-R2 wherein R2 is -(C1-6)alkyl, -(C3-6)cycloalkyl, both optionally substituted 1 or 2 times with halo or phenyl, or R2 is C6 aryl optionally substituted from 1 or 2 times with halo or (C1-6)alkyl.
3. The use according to claim 1 or 2, wherein A is cyclopentyl or terf-butyl; B is a thiazole substituted at position 2 with NH(R1) or NH(CO) R1 in which R is a Ci to C4 alkyl; and R is OH or a sulfonamide group wherein R2 is methyl, cyclopropyl or phenyl.
4. The use according to any one of claims 1 to 3, wherein said compound is a compound of formula (I) wherein R is OH.
5. The use according to any one of claim 1 to 3, wherein R is a sulfonamide group of formula -NHS02-R2 wherein R2 is -(C1-6)alkyl, -(C3.6)cycloalkyl, both optionally substituted 1 or 2 times with halo or phenyl, or R2 is C6 aryl optionally substituted from 1 or 2 times with halo or (C1-6)alkyl.
6. The use according to any one of claim 1 to 3, wherein A is cyclopentyl; B is a thiazole substituted at its 2 position with NHCH(CH3)2; and R is OH or a sulfonamide group wherein R2 is methyl, cyclopropyl or phenyl.
7. The use according to any one of claims 1 to 5, wherein said mammal is a human, and said Flaviviridae is selected from the group consisting of Yellow Fever virus, West Nile virus, Dengue fever virus, Japanese Encephalitis virus, GB virus A or C and Hepatitis G virus.
8. The use according to any one of claims 1 to 5, wherein said mammal is a cattle, and said Flaviviridae is selected from BVDV, border disease virus.
9. The use according to any one of claims 1 to 5, wherein said mammal is a pig, and said Flaviviridae is Classical Swine Fever Virus.
10. The use according to claim 5, wherein said mammal is a human, and said Flaviviridae is hepatitis C virus.
11. The use according to claim 6, wherein said mammal is a human, and said Flaviviridae is selected from the group consisting of Yellow Fever virus, West Nile virus, Dengue fever virus, Japanese Encephalitis virus, GB virus A or C and Hepatitis G virus.
12. The use, according to claim 6, wherein said mammal is a cattle, and said Flaviviridae is selected from BVDV, border disease virus.
13. The use according to claim 6, wherein said mammal is a pig, and said Flaviviridae is Classical Swine Fever Virus.
14. The use according to any one of claims 1 to 9, wherein said Flaviviridae virus comprises an NS3 protease comprising amino acid residues selected from: H57, G137, S139, A156 and A157.
15. An article of manufacture comprising packaging material contained within which is a composition effective to inhibit a virus of the Flaviviridae family and the packaging material comprises a label which indicates that the composition can be used to treat infection by a virus of the Flaviviridae family and, wherein said composition comprises a compound of Formula (I) as defined in claim 1.
PCT/CA2003/001634 2002-10-29 2003-10-24 Composition for the treatment of infection by flaviviridae viruses WO2004039833A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
EP03809665A EP1558633A1 (en) 2002-10-29 2003-10-24 COMPOSITION FOR THE TREATMENT OF INFECTION BY i FLAVIVIRIDAE /i VIRUSES
EA200500610A EA200500610A1 (en) 2002-10-29 2003-10-24 COMPOSITION FOR THE TREATMENT OF INFECTION CAUSED BY VIRUSES OF FAMILIES FLAVIVIRIDAE
CA002498642A CA2498642A1 (en) 2002-10-29 2003-10-24 Composition for the treatment of infection by flaviviridae viruses
BR0315781-4A BR0315781A (en) 2002-10-29 2003-10-24 Composition for treating infection caused by virus flaviviridae
JP2005501790A JP2006517196A (en) 2002-10-29 2003-10-24 Composition for treating Flaviviridae viral infection
MXPA05004604A MXPA05004604A (en) 2002-10-29 2003-10-24 Composition for the treatment of infection by flaviviridae.
AU2003275852A AU2003275852A1 (en) 2002-10-29 2003-10-24 Composition for the treatment of infection by flaviviridae viruses
NO20052580A NO20052580L (en) 2002-10-29 2005-05-27 Compounds for the treatment of infection by Flavivirida viruses

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US42190002P 2002-10-29 2002-10-29
US60/421,900 2002-10-29
US44276903P 2003-01-27 2003-01-27
US60/442,769 2003-01-27

Publications (1)

Publication Number Publication Date
WO2004039833A1 true WO2004039833A1 (en) 2004-05-13

Family

ID=32233460

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2003/001634 WO2004039833A1 (en) 2002-10-29 2003-10-24 Composition for the treatment of infection by flaviviridae viruses

Country Status (13)

Country Link
US (1) US20050159345A1 (en)
EP (1) EP1558633A1 (en)
JP (1) JP2006517196A (en)
KR (1) KR20050061592A (en)
AU (1) AU2003275852A1 (en)
BR (1) BR0315781A (en)
CA (1) CA2498642A1 (en)
EA (1) EA200500610A1 (en)
EC (1) ECSP055815A (en)
MX (1) MXPA05004604A (en)
NO (1) NO20052580L (en)
PL (1) PL376409A1 (en)
WO (1) WO2004039833A1 (en)

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1455809A2 (en) * 2001-12-20 2004-09-15 Bristol-Myers Squibb Company Inhibitors of hepatitis c virus
WO2007019674A1 (en) 2005-08-12 2007-02-22 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
US7368452B2 (en) 2003-04-18 2008-05-06 Enanta Pharmaceuticals, Inc. Quinoxalinyl macrocyclic hepatitis C serine protease inhibitors
WO2009076173A2 (en) 2007-12-05 2009-06-18 Enanta Pharmaceuticals, Inc. Fluorinated tripeptide hcv serine protease inhibitors
WO2009076747A1 (en) 2007-12-19 2009-06-25 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
US7608590B2 (en) 2004-01-30 2009-10-27 Medivir Ab HCV NS-3 serine protease inhibitors
WO2010021717A3 (en) * 2008-08-20 2010-04-15 Sequoia Pharmaceuticals, Inc. Hcv protease inhibitors
US7749961B2 (en) 2004-01-21 2010-07-06 Boehringer Ingelheim International Gmbh Macrocyclic peptides active against the hepatitis C virus
WO2010080874A1 (en) 2009-01-07 2010-07-15 Scynexis, Inc. Cyclosporine derivative for use in the treatment of hcv and hiv infection
US7816348B2 (en) 2006-02-03 2010-10-19 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
WO2010132163A1 (en) 2009-05-13 2010-11-18 Enanta Pharmaceuticals, Inc. Macrocyclic compounds as hepatitis c virus inhibitors
US7897622B2 (en) 2006-08-17 2011-03-01 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
EP2364984A1 (en) 2005-08-26 2011-09-14 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases
US8148399B2 (en) 2005-07-29 2012-04-03 Tibotec Pharmaceuticals Ltd. Macrocyclic inhibitors of hepatitis C virus
US8232246B2 (en) 2009-06-30 2012-07-31 Abbott Laboratories Anti-viral compounds
US8242140B2 (en) 2007-08-03 2012-08-14 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
US8314141B2 (en) 1996-10-18 2012-11-20 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases, particularly hepatitis C virus NS3 protease
US8420596B2 (en) 2008-09-11 2013-04-16 Abbott Laboratories Macrocyclic hepatitis C serine protease inhibitors
US8937041B2 (en) 2010-12-30 2015-01-20 Abbvie, Inc. Macrocyclic hepatitis C serine protease inhibitors
US8951964B2 (en) 2010-12-30 2015-02-10 Abbvie Inc. Phenanthridine macrocyclic hepatitis C serine protease inhibitors
US9333204B2 (en) 2014-01-03 2016-05-10 Abbvie Inc. Solid antiviral dosage forms
EP3020723A1 (en) 2010-09-21 2016-05-18 Enanta Pharmaceuticals, Inc. Macrocyclic proline derived hcv serine protease inhibitors
US10201584B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
USRE47334E1 (en) 2012-06-08 2019-04-02 Gilead Sciences, Inc. Macrocyclic inhibitors of flaviviridae viruses
US10246486B2 (en) 2012-06-08 2019-04-02 Gilead Sciences, Inc. Macrocyclic inhibitors of flaviviridae viruses
US10472392B2 (en) 2012-06-08 2019-11-12 Gilead Sciences, Inc. Macrocyclic inhibitors of flaviviridae viruses

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MXPA06003141A (en) * 2003-09-22 2006-06-05 Boehringer Ingelheim Int Macrocyclic peptides active against the hepatitis c virus.
BRPI0715714A2 (en) * 2006-08-25 2014-03-11 Wyeth Corp METHODS FOR REDUCING THE EMERGENCY FREQUENCY OF A RESISTANT TREATMENT HEPATITIS C VIRAL INFECTION TO DELAY THE EMERGENCY OF A TREATMENT RESISTANT HEPATITIS C VIRAL INFECTION TO REDUCE THE RESISTANCE LEVEL OF A VIRAL TREATMENT INFECTION TO REDUCE THE EMERGENCY OF AN HCV-796 RESISTANT HEPATITIS C VIRAL INFECTION, TO IDENTIFY AN INDIVIDUAL WITH A REDUCED PROBABILITY TO ANTI-HEPATITIS DIAGNOSTIC OR DIAGNOSTIC DIAGNOSIS MONITORING TREATMENT IN A PATIENT TO MONITOR THE TREATMENT COURSE OF HEPATITIS C VIRAL INFECTION IN A PATIENT TO PROGNOSTIC DEVELOPMENT OF A HEPATITIS C VIRAL INFECTION RESISTANT TO MONITOR THE INFECTION OF A HEPATITIS C PATIENT AND TO DIAGNOSE THE DEVELOPMENT OF A HEPATITIS VIRAL INFECTION OF TREATMENT IN A PATIENT YOU
EP2504343A4 (en) * 2009-11-24 2013-04-17 Boehringer Ingelheim Int Hepatitis c inhibitor compounds
JP2013511562A (en) * 2009-11-24 2013-04-04 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Hepatitis C inhibitor compound
KR101446049B1 (en) * 2013-01-14 2014-10-01 전남대학교산학협력단 Compositions for treatment or prevention of dengue virus related deseases
WO2021132626A1 (en) * 2019-12-27 2021-07-01 国立大学法人北海道大学 Therapeutic and/or preventive agent for classical swine fever

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000059929A1 (en) * 1999-04-06 2000-10-12 Boehringer Ingelheim (Canada) Ltd. Macrocyclic peptides active against the hepatitis c virus
WO2003053349A2 (en) * 2001-12-20 2003-07-03 Bristol-Myers Squibb Company Inhibitors of hepatitis c virus
WO2003064455A2 (en) * 2002-01-30 2003-08-07 Boehringer Ingelheim (Canada) Ltd. Macrocyclic peptides active against the hepatitis c virus
WO2003066103A1 (en) * 2002-02-07 2003-08-14 Boehringer Ingelheim Pharmaceuticals, Inc. Pharmaceutical compositions for hepatitis c viral protease inhibitors

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6608027B1 (en) * 1999-04-06 2003-08-19 Boehringer Ingelheim (Canada) Ltd Macrocyclic peptides active against the hepatitis C virus

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000059929A1 (en) * 1999-04-06 2000-10-12 Boehringer Ingelheim (Canada) Ltd. Macrocyclic peptides active against the hepatitis c virus
WO2003053349A2 (en) * 2001-12-20 2003-07-03 Bristol-Myers Squibb Company Inhibitors of hepatitis c virus
WO2003064455A2 (en) * 2002-01-30 2003-08-07 Boehringer Ingelheim (Canada) Ltd. Macrocyclic peptides active against the hepatitis c virus
WO2003066103A1 (en) * 2002-02-07 2003-08-14 Boehringer Ingelheim Pharmaceuticals, Inc. Pharmaceutical compositions for hepatitis c viral protease inhibitors

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BÜCHEN-OSMOND, C.: "00.026. Flaviridae", DATABASE UNIVERSAL VIRUS DATABASE, 2003, Columbia University, Oracle, AZ, USA, pages 1 - 3, XP002273068, Retrieved from the Internet <URL:www.ncbi.nlm.nih.gov/ICTVdb/ICTVdB/26000000.htm> [retrieved on 20040310] *
BÜCHEN-OSMOND, C.: "00.026.0.01. Flavivirus", DATABASE UNIVERSAL VIRUS DATABASE, 2003, Columbia University, Oracle, AZ, USA, pages 1 - 6, XP002274022, Retrieved from the Internet <URL:www.ncbi.nlm.nih.gov/ICTVdb/ICTVdB/00.026.0.01.htm> [retrieved on 20040310] *
BÜCHEN-OSMOND, C.: "00.026.0.02. Pestivirus", DATABASE UNIVERSAL VIRUS DATABASE, 2003, Columbia University, Oracle, AZ, USA, pages 1 - 4, XP002274023, Retrieved from the Internet <URL:www.ncbi.nlm.nih.gov/ICTVdb/ICTVdB/00.026.0.02.htm> [retrieved on 20040310] *
BÜCHEN-OSMOND, C.: "00.026.0.03. Hepacivirus", DATABASE UNIVERSAL VIRUS DATABASE, 2003, Columbia University, Oracle, AZ, USA, pages 1 - 4, XP002274024, Retrieved from the Internet <URL:www.ncbi.nlm.nih.gov/ICTVdb/ICTVdB/00.026.03.htm> [retrieved on 20040310] *

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8314141B2 (en) 1996-10-18 2012-11-20 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases, particularly hepatitis C virus NS3 protease
EP1455809A2 (en) * 2001-12-20 2004-09-15 Bristol-Myers Squibb Company Inhibitors of hepatitis c virus
EP1455809A4 (en) * 2001-12-20 2007-08-22 Bristol Myers Squibb Co Inhibitors of hepatitis c virus
US7368452B2 (en) 2003-04-18 2008-05-06 Enanta Pharmaceuticals, Inc. Quinoxalinyl macrocyclic hepatitis C serine protease inhibitors
US7749961B2 (en) 2004-01-21 2010-07-06 Boehringer Ingelheim International Gmbh Macrocyclic peptides active against the hepatitis C virus
US10172846B2 (en) 2004-01-30 2019-01-08 Medivir Ab HCV NS-3 serine protease inhibitors
US7608590B2 (en) 2004-01-30 2009-10-27 Medivir Ab HCV NS-3 serine protease inhibitors
US7671032B2 (en) 2004-01-30 2010-03-02 Medivir Ab HCV NS-3 serine protease inhibitors
US8349869B2 (en) 2005-07-29 2013-01-08 Tibotec Pharmaceuticals Ltd. Macrocylic inhibitors of hepatitis C virus
US9623022B2 (en) 2005-07-29 2017-04-18 Janssen Sciences Ireland Uc Macrocyclic inhibitors of hepatitis C virus
US8754106B2 (en) 2005-07-29 2014-06-17 Janssen R&D Ireland Macrocyclic inhibitors of hepatitis C virus
US9040562B2 (en) 2005-07-29 2015-05-26 Janssen R&D Ireland Macrocyclic inhibitors of hepatitis C virus
US9856265B2 (en) 2005-07-29 2018-01-02 Janssen Sciences Ireland Uc Macrocyclic inhibitors of hepatitis C virus
US9353103B2 (en) 2005-07-29 2016-05-31 Janssen Sciences Ireland Uc Macrocyclic inhibitors of hepatitis C virus
US8741926B2 (en) 2005-07-29 2014-06-03 Janssen R&D Ireland Macrocyclic inhibitors of hepatitis C virus
US8153800B2 (en) 2005-07-29 2012-04-10 Tibotec Pharmaceuticals Ltd. Macrocyclic inhibitors of hepatitis C virus
US8148399B2 (en) 2005-07-29 2012-04-03 Tibotec Pharmaceuticals Ltd. Macrocyclic inhibitors of hepatitis C virus
WO2007019674A1 (en) 2005-08-12 2007-02-22 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
EP2366704A1 (en) 2005-08-26 2011-09-21 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases
EP2364984A1 (en) 2005-08-26 2011-09-14 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases
US7816348B2 (en) 2006-02-03 2010-10-19 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
US7897622B2 (en) 2006-08-17 2011-03-01 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
US8242140B2 (en) 2007-08-03 2012-08-14 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
WO2009076173A2 (en) 2007-12-05 2009-06-18 Enanta Pharmaceuticals, Inc. Fluorinated tripeptide hcv serine protease inhibitors
US8476257B2 (en) 2007-12-19 2013-07-02 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
US8541402B2 (en) 2007-12-19 2013-09-24 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
WO2009076747A1 (en) 2007-12-19 2009-06-25 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
US8912182B2 (en) 2007-12-19 2014-12-16 Boehringer Ingelheim International Gmbh Viral polymerase inhibitors
WO2010021717A3 (en) * 2008-08-20 2010-04-15 Sequoia Pharmaceuticals, Inc. Hcv protease inhibitors
US8765667B2 (en) 2008-08-20 2014-07-01 Michael Eissenstat HCV protease inhibitors
US8420596B2 (en) 2008-09-11 2013-04-16 Abbott Laboratories Macrocyclic hepatitis C serine protease inhibitors
US8642538B2 (en) 2008-09-11 2014-02-04 Abbvie, Inc. Macrocyclic hepatitis C serine protease inhibitors
US9309279B2 (en) 2008-09-11 2016-04-12 Abbvie Inc. Macrocyclic hepatitis C serine protease inhibitors
WO2010080874A1 (en) 2009-01-07 2010-07-15 Scynexis, Inc. Cyclosporine derivative for use in the treatment of hcv and hiv infection
WO2010132163A1 (en) 2009-05-13 2010-11-18 Enanta Pharmaceuticals, Inc. Macrocyclic compounds as hepatitis c virus inhibitors
US8232246B2 (en) 2009-06-30 2012-07-31 Abbott Laboratories Anti-viral compounds
EP3020723A1 (en) 2010-09-21 2016-05-18 Enanta Pharmaceuticals, Inc. Macrocyclic proline derived hcv serine protease inhibitors
US8951964B2 (en) 2010-12-30 2015-02-10 Abbvie Inc. Phenanthridine macrocyclic hepatitis C serine protease inhibitors
US8937041B2 (en) 2010-12-30 2015-01-20 Abbvie, Inc. Macrocyclic hepatitis C serine protease inhibitors
US10201584B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
US10201541B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
USRE47334E1 (en) 2012-06-08 2019-04-02 Gilead Sciences, Inc. Macrocyclic inhibitors of flaviviridae viruses
US10246486B2 (en) 2012-06-08 2019-04-02 Gilead Sciences, Inc. Macrocyclic inhibitors of flaviviridae viruses
US10472392B2 (en) 2012-06-08 2019-11-12 Gilead Sciences, Inc. Macrocyclic inhibitors of flaviviridae viruses
US9744170B2 (en) 2014-01-03 2017-08-29 Abbvie Inc. Solid antiviral dosage forms
US9333204B2 (en) 2014-01-03 2016-05-10 Abbvie Inc. Solid antiviral dosage forms
US10105365B2 (en) 2014-01-03 2018-10-23 Abbvie Inc. Solid antiviral dosage forms

Also Published As

Publication number Publication date
KR20050061592A (en) 2005-06-22
NO20052580D0 (en) 2005-05-27
EA200500610A1 (en) 2005-10-27
JP2006517196A (en) 2006-07-20
NO20052580L (en) 2005-07-20
EP1558633A1 (en) 2005-08-03
BR0315781A (en) 2005-09-13
AU2003275852A1 (en) 2004-05-25
US20050159345A1 (en) 2005-07-21
PL376409A1 (en) 2005-12-27
MXPA05004604A (en) 2005-07-13
CA2498642A1 (en) 2004-05-13
ECSP055815A (en) 2005-08-11

Similar Documents

Publication Publication Date Title
US20050159345A1 (en) Composition for the treatment of infection by Flaviviridae viruses
ES2354282T3 (en) PEPTIDE ANALOGS INHIBITORS OF HEPATITIS C.
AU2003202348B2 (en) Heterocyclic tripeptides as hepatitis c inhibitors
AU2003202347B2 (en) Tripeptides having a hydroxyproline ether of a substituted quinoline for the inhibition of NS3 (Hepatitis C)
US7091184B2 (en) Hepatitis C inhibitor tri-peptides
JP4040578B2 (en) Macrocyclic peptide active against hepatitis C virus
US6642204B2 (en) Hepatitis C inhibitor tri-peptides
US20050075279A1 (en) Macrocyclic peptides active against the hepatitis C virus
AU2003202348A1 (en) Heterocyclic tripeptides as hepatitis c inhibitors
AU2003202347A1 (en) Tripeptides having a hydroxyproline ether of a substituted quinoline for the inhibition of NS3 (Hepatitis C)
ZA200100972B (en) Hepatituis C inhibitor peptides.
NO332056B1 (en) Hepatitis C inhibitor compounds, pharmaceutical compositions containing them as well as such compounds for the treatment of disease
CA2370400A1 (en) Hepatitis c inhibitor tri-peptides
CA2474156C (en) Tripeptides having a hydroxyproline ether of a substituted quinoline for the inhibition of ns3 (hepatitis c)
CA2474031C (en) Heterocyclic tripeptides as hepatitis c inhibitors
TW200412958A (en) Composition for the treatment of infection by Flaviviridae viruses

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2498642

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2003809665

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 200503192

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 1667/DELNP/2005

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 1-2005-500780

Country of ref document: PH

Ref document number: P-2005/0334

Country of ref document: YU

WWE Wipo information: entry into national phase

Ref document number: 168253

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2005501790

Country of ref document: JP

Ref document number: P20050380A

Country of ref document: HR

WWE Wipo information: entry into national phase

Ref document number: 376409

Country of ref document: PL

Ref document number: PA/a/2005/004604

Country of ref document: MX

Ref document number: 1020057007589

Country of ref document: KR

Ref document number: 20038A24361

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 200500610

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 540162

Country of ref document: NZ

Ref document number: 2003275852

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 05051554

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 20050196

Country of ref document: UZ

WWE Wipo information: entry into national phase

Ref document number: 1200500734

Country of ref document: VN

WWP Wipo information: published in national office

Ref document number: 1020057007589

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2003809665

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0315781

Country of ref document: BR

WWW Wipo information: withdrawn in national office

Ref document number: 2003809665

Country of ref document: EP