WO2011003071A1 - Procédés de modulation d'une infection par le virus de l'hépatite c - Google Patents

Procédés de modulation d'une infection par le virus de l'hépatite c Download PDF

Info

Publication number
WO2011003071A1
WO2011003071A1 PCT/US2010/040918 US2010040918W WO2011003071A1 WO 2011003071 A1 WO2011003071 A1 WO 2011003071A1 US 2010040918 W US2010040918 W US 2010040918W WO 2011003071 A1 WO2011003071 A1 WO 2011003071A1
Authority
WO
WIPO (PCT)
Prior art keywords
host factor
nucleic acid
compound
acid molecule
encoded
Prior art date
Application number
PCT/US2010/040918
Other languages
English (en)
Inventor
T. Jake Liang
Qisheng Li
Abraham L. Brass
Stephen Elledge
Original Assignee
The United States Of America, As Represented By The Secretary, Department Of Health & Human Services
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 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services filed Critical The United States Of America, As Represented By The Secretary, Department Of Health & Human Services
Publication of WO2011003071A1 publication Critical patent/WO2011003071A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/04Nitro compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.

Definitions

  • the present invention relates to methods of identifying inhibitors or enhancers of hepatitis C virus infection, and the use of such inhibitors in protecting patients from hepatitis C virus infection.
  • Hepatitis C virus is an important cause of liver disease worldwide. The virus causes a chronic infection resulting in progressive liver damage. Hepatitis C virus (HCV) enters hepatocytes via interactions between the viral envelope proteins, El and E2, and four known host receptors, CD81, Claudin-1, SBR-I and occludin (Ploss, Nature 457, 2009, pp 882-886). Subsequent to entry, host ribosomes bind to the internal ribosomal entry site (IRES) of the HCV genome, and translate viral polyproteins on the rough endoplasmic reticulum (ER) (Moradpour (2007) Nat Rev Microbiol 5, 453-463).
  • IRS internal ribosomal entry site
  • Host and viral proteases process the polyprotein into both structural (core, and envelope proteins, El and E2) and nonstructural proteins (p7, NS2-3, NS3, NS4A, NS4B, NS5A, and NS5B, (Lindenbach (2005) Nature 436, 933-938; WoIk (2008) J Virol 82, 10519-10531.).
  • Oligomerization of NS4B distorts the host ER into membranous webs, which house HCV replication complexes (RCs), within which the RNA-dependent RNA polymerase, NS5B, transcribes viral genomic RNAs (Moradpour (2007) Nat Rev Microbiol 5, 453-463.).
  • RCs HCV replication complexes
  • Progeny viral genomes generated in the RCs are translocated to lipid droplet-containing organelles and assemble into virions, which then traffic to the cell surface for release (Miyanari (2007) Nat Cell Biol 9, 1089-1097
  • HCV replication has benefited from the use of the replicon system, consisting of sub-genomic or whole genome viral RNAs stably expressed in permissive tissue culture cells.
  • the replicon system consisting of sub-genomic or whole genome viral RNAs stably expressed in permissive tissue culture cells.
  • several recent functional genomic screens using either partial or genome-wide siRNA libraries, have identified a number of host factors involved in HCV replication (Tai (2009) Cell Host Microbe 5, 298-307; Supekova (2008) J Biol Chem 283, 29-36; Ng (2007) Hepatology 45, 1413-1421.).
  • the replicon system is unable to address the role of host factors in the entire HCV lifecycle.
  • HCVcc The infection-competent HCV cell culture system (HCVcc) recapitulates the complete HCV lifecycle, thereby permitting a greater range of host- viral interactions to be studied (Wakita (2005) Nat Med 11, 791-796; Zhong (2005) Proc Natl Acad Sd U S A 102, 9294-9299; Lindenbach (2005) Science 309, 623-626; Tellinghuisen (2007) J Virol 81, 8853-8867.)); see also PCT International Publication WO 2007/013882 A2, published Feb 1, 2007.
  • Two efforts have employed the HCVcc system using limited candidate gene siRNA screens of either 65 or 140 curated targets. (Randall (2007) Proc Natl Acad Sd USA 104, 12884-12889; Berger (2009) PNAS.).
  • the present invention identifies, for the first time, numerous host factors that are involved in modulating hepatitis C infection. Because these host factors are involved in modulating viral infection, they represent new targets that can be used to identify compounds capable of inhibiting or enhancing viral infection. Thus the present invention relates to methods of using host factors of the present invention to identify compounds that modulate viruses of the present invention. The invention further relates to compounds that interact with host factors of the present invention, or biological pathways comprising host factors of the present invention, in such a way as to inhibit or enhance viral infection. The present invention also relates to methods to inhibit or enhance viral infection using inhibitory or enhancement compounds, and methods to protect a patient from viral infection using such compounds.
  • One embodiment of the present invention is a method to identify a compound that modulates viral infection, the method comprising assaying a candidate compound for the ability to interact with a host factor of the present invention in such a manner as to modulate viral infection.
  • the virus is hepatitis C virus.
  • One embodiment of the present invention is a method to identify a compound that modulates viral infection, the method comprising; (a) contacting a candidate compound with at least one isolated host factor under conditions suitable for formation of a complex between said compound and said host factor; and,
  • Another embodiment of the present invention is a method to identify a compound that modulates viral infection, the method comprising;
  • Still another embodiment of the present invention is a method to identify a compound hat modulates viral infection, which comprises:
  • an inhibitory compound of the present invention modulates a pathway comprising a host factor of the present invention by interacting with said host factor, thereby inhibiting viral infection.
  • an enhancing compound of the present invention modulates a pathway comprising a host factor of the present invention by interacting with said host factor, hereby enhancing viral infection.
  • Another embodiment of the present invention is an inhibitory compound for inhibiting nfection by a virus of the present invention, wherein the inhibitory compound interacts with a host factor of the present invention, thereby inhibiting the viral infection.
  • an inhibitory compound of the present invention modulates a pathway comprising a host factor of the present invention by interacting with said host factor, thereby nhibiting viral infection
  • an enhancing compound of the present nvention modulates a pathway comprising a host factor of the present invention by nteracting with said host factor, thereby enhancing viral infection.
  • One embodiment of the present invention is a method to inhibit viral infection, the method comprising contacting a cell infected with the virus with an inhibitory compound that nteracts with a host factor of the present invention in such a manner as to inhibit viral nfection.
  • Another embodiment of the present invention is a method to enhance viral nfection, the method comprising contacting an infected cell with an enhancing compound hat interacts with a host factor in such a manner as to increase the amount of virus produced.
  • the cell is infected with hepatitis C virus.
  • Another embodiment is a method to protect a patient from virus infection, comprising administering to a patient an inhibitory compound that modulates a pathway comprising a host factor of the present invention in such a manner as to inhibit viral infection.
  • the embodiments include a method to inhibit viral infection, wherein such a method comprises contacting a cell with an inhibitory compound that interacts with a host factor encoded by a nucleic acid molecule listed in Table 5 or with a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1, thereby inhibiting viral infection.
  • such a method comprises contacting a patient with such an inhibitory compound.
  • Such a patient can be infected with such a virus.
  • such a virus is a Flavi virus, such as a hepatitis C virus.
  • the embodiments include a method to inhibit viral infection, wherein such a method comprises contacting a cell with an inhibitory compound that modulates a pathway comprising a host factor encoded by a nucleic acid molecule listed in Table 5 or a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1, therebynhibiting viral infection.
  • such a method comprises contacting a patient with such an inhibitory compound.
  • Such a patient can be infected with such a virus.
  • such a virus is a Flavivirus, such as a hepatitis C virus.
  • the embodiments include a method to protect a patient from hepatitis C virus nfection, wherein such a method comprises administering an inhibitory compound that nteracts with a host factor encoded by a nucleic acid molecule listed in Table 5 or with a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1.
  • a method can provide prophylactic protection.
  • such a method can provide therapeutic protection.
  • the embodiments include a method to protect a patient from hepatitis C virus nfection, wherein such a method comprises administering an inhibitory compound that modulates a pathway comprising a host factor encoded by a nucleic acid molecule listed in Table 5 or a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1.
  • a method can provide prophylactic protection.
  • such a method can provide therapeutic protection.
  • the embodiments include a method to enhance viral infection, wherein such a method comprises contacting a cell with an enhancing compound that interacts with a host factor encoded by a nucleic acid molecule listed in Table 5 or with a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1, thereby enhancing viral infection.
  • a virus is a Flavivirus, such as a hepatitis C virus.
  • the embodiments include a method to enhance viral infection, wherein such a method comprises contacting a cell with an enhancing compound that modulates a pathway comprising a host factor encoded by a nucleic acid molecule listed in Table 5 or a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1 , thereby enhancing viral infection.
  • a virus is a Flavivirus, such as a hepatitis C virus.
  • the embodiments include an inhibitory compound that interacts with a host factor encoded by a nucleic acid molecule listed in Table 5 or with a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1, thereby inhibiting viral infection.
  • the embodiments include an inhibitory compound that modulates a pathway comprising a host factor encoded by a nucleic acid molecule listed in Table 5 or a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1, therebynhibiting viral infection.
  • a virus is a Flavivirus, such as a hepatitis C virus.
  • the embodiments include an enhancing compound that interacts with a host factor encoded by a nucleic acid molecule listed in Table 5 or with a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1, thereby enhancing viral nfection.
  • the embodiments include an enhancing compound that modulates a pathway comprising a host factor encoded by a nucleic acid molecule listed in Table 5 or a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1 , thereby enhancing viral infection.
  • a virus is a Flavivirus, such as a hepatitis C virus.
  • the embodiments include a method to identify a compound that inhibits viral nfection, wherein such a method comprises assaying a candidate compound for the ability to nteract with a host factor in such a manner as to inhibit such an infection, wherein such a host factor is encoded by a nucleic acid molecule listed in Table 5 or is encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1.
  • a virus is a Flavivirus, such as a hepatitis C virus.
  • the embodiments include an inhibitory compound identified by such a method.
  • the embodiments include a method to identify a compound that inhibits viral infection, wherein such a method comprises: (a) combining a candidate compound with a host factor encoded by a nucleic acid molecule listed in Table 5 or with a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1 under conditions suitable for such a candidate compound to interact with such a host factor; (b) assaying for the presence of such an interaction; and (c) determining if such an interaction results in a decrease in the amount of virus obtained upon infection of a cell with such a virus; wherein a decrease in the amount of virus indicates that such a compound inhibits viral infection.
  • a virus is a Flavivirus, such as a hepatitis C virus.
  • embodiments include an inhibitory compound identified by such a method.
  • the embodiments include a method to identify a compound that inhibits viral infection, wherein such a method comprises assaying a candidate compound for the ability to modulate a pathway comprising a host factor encoded by a nucleic acid molecule listed in Table 5 or a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1, wherein the ability to modulate such a pathway identifies a compound that inhibits viral infection.
  • a virus is a Flavivirus, such as a hepatitis C virus.
  • the embodiments include an inhibitory compound identified by such a method.
  • the embodiments include a method to identify a compound that enhances viral infection, wherein such a method comprises assaying a candidate compound for the ability to interact with a host factor in such a manner as to enhance such an infection, wherein such a host factor is encoded by a nucleic acid molecule listed in Table 5 or is encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1.
  • a virus is a Flavivirus, such as a hepatitis C virus.
  • the embodiments include a method to identify a compound that enhances viral infection, wherein such a method comprises: (a) combining a candidate compound with a host factor encoded by a nucleic acid molecule listed in Table 5 or with a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1 under conditions suitable for such a candidate compound to interact with such a host factor; (b) assaying for the presence of such an interaction; and (c) determining if such an interaction results in an increase in the amount of virus obtained upon infection of a cell with such a virus; wherein an increase in the amount of virus indicates that such a compound enhances viral infection.
  • a virus is a Flavivirus, such as a hepatitis C virus.
  • embodiments include an inhibitory compound identified by such a method.
  • the embodiments include a method to identify a compound that enhances viral infection, wherein such a method comprises assaying a candidate compound for the ability to modulate a pathway comprising a host factor encoded by a nucleic acid molecule listed in Table 5 or a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1, wherein the ability to modulate such a pathway identifies a compound that enhances viral infection.
  • a virus is a Flavivirus, such as a hepatitis C virus.
  • the embodiments include an inhibitory compound identified by such a method.
  • the embodiments include a host factor encoded by a nucleic acid molecule listed in Table 2.
  • the embodiments include a host factor encoded by a nucleic acid molecule listed in Table 11, Table 12, Table 13 or Table 14.
  • the embodiments include a host factor encoded by a nucleic acid molecule comprising SEQ ID NO:79.
  • the embodiments include a host factor comprising amino acid sequence SEQ ID NO:80.
  • the embodiments include one of more inhibitory compounds listed in Table 3.
  • the embodiments include one or more of the following inhibitory compounds: colchicine, cytochalasin, EIPA, rottlerin, UO 126 or wortmannin.
  • the embodiments include one or more of the following inhibitory compounds: brefeldin A or golgicide A.
  • the embodiments include an inhibitory compound comprising a small interfering RNA (siRNA).
  • siRNA small interfering RNA
  • Fig. 6 (A) Effect of siRNA silencing of expression of various NF-k ⁇ pathway-related factors on HCV RNA levels. (B) Relative knock-down efficiency of various host factors by the siRNAs.
  • Fig. 7 (A) Effect of expression of IKK ⁇ dominant negative mutant (HA-IKKaKA) on HCV RNA levels. (B) Western blot illustrating levels of HA-IKKaKA.
  • Fig. 8 Effect of siRNA silencing of IKK ⁇ expression on (A) intracellular HCV RNA levels and (B) extracellular HCV RNA levels.
  • C Relative silencing efficiency of IKK ⁇ expression as measured by quantitative PCR.
  • Fig. 9 (A) Effect of siRNA silencing of IKK ⁇ expression on (A) viral entry of an HCV pseudovirus and (B) RNA replication.
  • Fig 10 Effect of (A) rotterlin, (B) colchicines, (C) EIPA, (D) wortmannin, (E) cytochalasin, r (F) U0126 on HCV entry.
  • Fig. 11 Effect of (A) rottlerin or (B) EIPA on HCV RNA levels.
  • Fig. 12 Effect of (A) golgicide or (B) brefeldin on HCV RNA levels.
  • a entity or “an” entity refers to one or more of that entity.
  • a host factor refers to one or more host factors.
  • the terms “a”, “an”, “one or more” and “at least one” can be used interchangeably.
  • the terms “comprising”, “including” and “having” can be used interchangeably.
  • the present invention generally relates to the discovery that numerous host factors are involved in modulating (e.g., inhibiting or enhancing) hepatitis C virus (HCV) infection.
  • the present invention relates to methods of using host factors of the present invention to identify compounds that modulate viral infection.
  • Also encompassed are compounds that interact with host factors of the present invention, and the use of such compounds to modulate infection by viruses of the present invention.
  • the instant invention is novel since, prior to the inventors' surprising discovery, it was not appreciated that host factors disclosed herein were involved in modulating viral infection.
  • host factors of the present invention represent new tools for identifying compounds that can modulate viral infection.
  • a host factor of the present invention is a molecule produced by the host cell that modulates HCV infection.
  • Host factors are biological molecules, examples of which include proteins, peptides, lipids, nucleic acid molecules, carbohydrates, lipoproteins, glycoproteins, metabolic products, and the like.
  • a host factor modulates HCV infection if, when the host factor-related activity is altered (e.g., reduced or eliminated, or increased) the amount of virus is decreased or increased.
  • host factors can be proviral or antiviral, and the amount of virus produced during an infection is influenced by interactions between viral components, proviral host factors and antiviral host factors.
  • a proviral host factor is a molecule that positively influences virus production. Consequently when the amount or activity of a proviral host factor is reduced or eliminated, the amount of virus produced is decreased.
  • 3-hydroxy-3-methylgluttaryl CoA reductase is known to be required for replication of HCV and thus is a proviral host factor (Ye, PNAS, Dec. 23, 2003, ppl5865-15870).
  • An antiviral host factor is a molecule that negatively influences virus production.
  • an antiviral host factor is interleukin 7 receptor ⁇ .
  • Host factors of the present invention can be identified by those skilled in the art using the screening methods disclosed herein, e.g., siRNA screening.
  • host factors of the present invention or the nucleic acid molecules encoding such host factors are listed in one or more of the Tables included in this disclosure. These nucleic acid molecules are characterized in various ways appreciated by those skilled in the art, e.g., Entrez Gene Symbol, Entrez Gene ID, Genbank Accession Number, Gene Name, SEQ ID NO: (or SEQ NO).
  • host factors of the present invention are encoded by the nucleic acid molecules listed in Table 1 , with their corresponding nucleic acid sequence SEQ ID NO:s.
  • Preferred host factors are those encoded by nucleic acid molecules listed in Table 2.
  • host factors are described by their amino acid sequences as listed in Table 1.
  • Certain host factors are encoded by nucleic acid molecules listed in Table 5.
  • Certain host factors are encoded by a nucleic acid molecule listed in Table 5 or are encoded by a nucleic acid molecule having a SEQ ID NO listed in Table 1. Certain host factors involved in viral entry are encoded by nucleic acid molecules listed in Table 11. Certain host factors involved in viral IRES-mediated translation are encoded by nucleic acid molecules listed in Table 12. Certain host factors involved in viral replication are encoded by nucleic acid molecules listed in Table 13. Certain host factors involved in viral trafficking, assembly and/or release are encoded by nucleic acid molecules listed in Table 14. In some embodiments, host factors are encoded by nucleic acid molecules listed in Table 11, Table 12, Table 13 or Table 14. hi certain embodiments, a host factor is encoded by a nucleic acid molecule comprising SEQ ID NO:79. In certain embodiments, a host factor comprises amino acid sequence SEQ ID NO:80.
  • viral infection refers to the ability of a virus to carry out all steps in the viral life cycle, resulting in the production of infectious particles.
  • a life cycle comprises a variety of steps including, for example, attachment, uncoating,ranscription, translation, protein processing, replication of nucleic acid molecules, assembly of viral particles, intracellular transport of viral particles, budding, release and the like.
  • Other teps may also be included depending on the virus.
  • One embodiment of the present invention is a method to identify a compound that modulates viral infection, the method comprising assaying a candidate compound for the ability to interact with a host factor of the present invention in such a manner as to modulate viral infection.
  • the virus is hepatitis C virus.
  • the method identifies a compound that inhibits viral infection by assaying a candidate compound for the ability to interact with a host factor of the present invention in such a manner as to inhibit viral infection.
  • the method dentifies a compound that enhances viral infection by assaying a candidate compound for the ability to interact with a host factor of the present invention in such a manner as to enhance viral infection.
  • the phrases modulate viral infection, modulate infection by a virus of he present invention, modulate viral infectivity, modulate viral propagation, and the like refers to increasing or decreasing the amount of virus present in an infected cell or patient relative to the amount of virus present in a cell or patient that has not been treated using the disclosed methods or compounds. Also encompassed is the ability to prevent viral infection. It should be appreciated that the terms amount and concentration can be used
  • An amount of virus can also be referred to as a titer. It is also understood by those of skill in the art that the amount of virus can refer to the total number of viral particles, or it can refer to the number of viral particles that are infectious, i.e. capable of carrying out the viral life cycle, including the ability to effect another cycle of infectious particle formation. For example, in a given population of virus particles, some or all of the particles may be unable to carry out a specific step in its life cycle (e.g., attachment or entry) due to a deficiency in a molecule needed to perform that step. While the number of particles in the population may be large, the number of infectious particles could be small to none.
  • the amount of virus determined by counting virus particles may differ from that determined by measuring functional virus in, for example, a plaque assay. Accordingly methods of the present invention can be used to identify compounds that affect the total number of viral particles produced, as well as the number of infectious viral particles produced. Appropriate methods of determining the amount of virus are understood by those skilled in the art and nclude, but are not limited to, directly counting virus particles, titering virus in cell culture e.g., plaque assay), measuring the amount of viral protein(s), measuring the amount of viral nucleic acids, or measuring the amount of a reporter protein, e.g., luciferase.
  • Modulation of viral infection can result in a partial reduction in the amount of virus, or it can result in complete elimination of virus from a cell or patient or in prevention of viral nfection.
  • the amount of virus is reduced by at east 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at east 70%, at least 80%, at least 90%, or at least 95%.
  • the amount of virus is reduced by a factor of at least 10, at least 100, at least 1000, or at least 10,000.
  • the viral infection is completely inhibited (i.e., there are no infectious particles).
  • the amount of virus is increased by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95%.
  • the amount of virus is ncreased by a factor of at least 10, at least 100, at least 1000, or at least 10,000.
  • a candidate compound is any compound which may have the ability o modulate viral infection by interacting with a host factor of the present invention, but that has not yet been tested for such activity.
  • Candidate compounds encompassed by the present nvention include, but are not limited to, proteins, peptides, antibodies, lipids, nucleic acid molecules, small interfering RNA' s (siRNA's), carbohydrates, sugars, lipoproteins, small molecule compounds and combinations thereof.
  • Such compounds can be isolated from nature (e.g., isolated from organisms) or they can be produced in a laboratory (e.g., recombinantly or synthetically).
  • compounds that are combinations of natural and synthetic molecules. Methods to isolate or produce recombinant or synthetic candidate compounds are known to those skilled in the art.
  • an inhibitory compound of the present invention is any molecule that inhibits (e.g., reduces, eliminates or prevents) a virus of the present invention by interacting with a pro viral host factor of the present invention to reduce the host factor's amount or activity, or by interacting with an antiviral host factor of the present invention to increase that host factor's amount or activity.
  • Inhibitory compounds of the present invention also encompass any molecule that inhibits a virus of the present invention by interacting with a pathway comprising a host factor of the present invention in such a manner as to inhibit viral infection.
  • an enhancing compound of the present invention is any molecule that enhances infection of a virus of the present invention by interacting with an antiviral host factor of the present invention to reduce that host factor's amount or activity, or by interacting with a pro viral host factor of the present invention to increase that host factor's amount or activity.
  • Enhancing compounds of the present invention also encompass any molecule that enhances infection of a virus of the present invention by interacting with a pathway comprising a host factor of the present invention in such a manner as to enhance viral infection.
  • a virus of the present invention is any virus, the infectivity of which is modulated by a host factor of the present invention (i.e., a host factor that modulates HCV infection), or a pathway comprising a host factor of the present invention.
  • a host factor of the present invention i.e., a host factor that modulates HCV infection
  • a pathway comprising a host factor of the present invention e.g., a virus that modulates HCV infection
  • viruses include those in a family selected from the group consisting of adenoviridae, herpesviridae, pappillomaviridae, polyomaviridae, poxviridae, parvoviridae, hepadnaviridae, retroviridae, arenaviridae, bornaviridae, bunyaviridae, filoviridae, orthomyxoviridae, paramyxoviridae, rhabdoviridae, arteriviridae, astroviridae, caliciviridae, coronaviridae, flaviviridae, hepatitis-E-like viruses, picornaviridae, and togaviridae that require a host factor of the present invention.
  • a preferred virus is one from the Flaviviridae family of viruses, which includes the genera Flavivirus, Pestisvirus, and Hepacivirus.
  • Suitable Flavi viruses for practicing the instant invention include Gadget's Gully virus, Kadam virus, Kyasanur Forrest disease virus, Langat virus, Omsk hemorrhagic fever virus, Tick-borne encephalitis virus, Louping ill virus, Aroa virus, Dengue viruses 1-4, Kedougou virus, Cacipacore virus, Koutango virus, Murray Valley encephalitis virus, St.
  • Louis encephalitis virus Usutu virus, West Nile virus, Yaounde virus, Kokobera virus group, Kokobera virus, Bagaza virus, Ilheus virus, Israel turkey meningoencephalomyelitis virus, Ntaya virus, Tembusu virus, Zika virus, Banzi virus, Bouboui virus,Edge Hill virus, Jugra virus, Saboya virus, Sepik virus, Kenya S virus, Wesselsbron virus, Yellow fever virus, Entebbe bat virus, Yokose virus, acea virus, Cowbone Ridge virus, Jutiapa virus, Modoc virus, Sal Vieja virus, San Perlita virus, Bukalasa bat virus, Carey Island virus, Dakar bat virus, Montana myotis leukoencephalitis virus, Phnom Penh bat virus, Rio Bravo virus, hepatitis C virus, e.g., hepatitis C virus genotypes 1-6, and GB virus A and B.
  • a preferred virus is a member of the Hepacvirus gen
  • the term interact indicates that the candidate compound and the host factor come into sufficient physical proximity so that the compound affects the host factor in such a manner as to inhibit or enhance viral infection.
  • Preferred compounds are those that selectively interact with a host factor of the present invention.
  • the terms selectively, selective, specifically, and the like indicate the compound has a greater affinity for the host factor than it does for molecules unrelated to the host factor.
  • One type of interaction is a binding interaction. In such an interaction the compound associates with the host factor to form a complex.
  • An example of complex formation is the association of an antigen with an antibody or a drug with its target. According to the present invention, association of a compound with a host factor can be reversible or non-reversible.
  • Kd dissociation constant
  • Formation of a complex may or may not result in modification of the host factor.
  • Methods of measuring and analyzing interactions, including binding interactions, between a compound and a host factor are known by those of skill in the art. hi some instances, the interaction between a compound and a host factor can result in modification of the host factor. In these instances, the host factor and the compound may or may not form a complex.
  • a compound may have proteolytic activity, and may cleave the host factor thereby reducing the amount of host factor present, or affecting any activity possessed by the host factor.
  • host factor modifications include, but are not limited to, cleavage, phosphorylation, myristylation, ligation, and the like. Suitable techniques for assaying candidate compounds for their ability to interact with host factors of the present invention are known to those skilled in the art. Such assays can be in vitro or in vivo assays. Examples of useful assays include, but are not limited to, an enzyme-linked immunoassay, a competitive enzyme-linked immunoassay, a
  • radioimmunoassay a fluorescence immunoassay, a chemiluminescent assay, a lateral flow assay, a flow-through assay, an agglutination assay, a particulate-based assay (e.g., using particulates such as, but not limited to, magnetic particles or plastic polymers, such as latex or polystyrene beads), an immunoprecipitation assay, an immunoblot assay (e.g., a western blot), a phosphorescence assay, a flow-through assay, a chromatography assay, a polyacrylamide gel electrophoresis (PAGe)-based assay, a surface plasmon resonance assay, a spectrophotometry assay, a particulate-based assay, and an electronic sensory assay.
  • PAGe polyacrylamide gel electrophoresis
  • an assay can be performed in cells in culture or it can be performed in a whole animal. Assays can be designed to give qualitative, quantitative or semi-quantitative results, depending on how they are used and the type of result that is desired.
  • One embodiment of the present invention is a method to identify a compound that modulates viral infection, the method comprising;
  • a complex if present; wherein the presence of said complex indicates that the test compound modulates viral infection.
  • Suitable methods for detecting the formation of a complex between a candidate compound and a host factor have been disclosed herein.
  • a compound can be immobilized on a substrate and the immobilized compound then brought into contact with a host factor under conditions suitable for formation of a complex.
  • the presence of a complex can be detected using a detectable marker.
  • detectable markers include, but are not limited to, a radioactive marker, a colorimetric marker, a fluorescent marker, and a chemiluminescent marker.
  • the detectable marker is directly linked (e.g., covalently) to the host factor.
  • the complex can be detected by using a detection molecule that binds to the host factor, and that is labeled with a detectable marker.
  • a detection molecule is any molecule that binds to the host factor, and can be used detect its presence. Examples of such molecules include, but are not limited to, antibodies, ligand, and nucleic acid molecules.
  • the host factor can be immobilized on a substrate and a candidate compound introduced under conditions suitable for formation of a complex. Similar methodology to that described above can then be used to detect binding of the candidate compound to the immobilized host factor. In one
  • the Kd of a complex formed by a host factor of the present invention and a compound that selectively interacts with the host factor is at least 10 -4 , 10 -5 , 10 '6 , 10 '7 , 10 '8 , 10 '9 , 10 -10 , 10 -11 or 10 -12 .
  • binding of a host factor to a compound that selectively interacts with the host factor is irreversible.
  • a compound forms a covalent bond with the host factor.
  • a host factor is encoded by a nucleic acid molecule having a SEQ ID NO. listed in Table 1.
  • a host factor is encoded by a nucleic acid molecule listed in Table 2. Ln certain embodiments, a host factor is encoded by a nucleic acid molecule listed in Table 5. hi certain embodiments, a host factor is encoded by a nucleic acid molecule listed in Table 5 or encoded by a nucleic acid molecule having a SEQ ID NO listed in Table 1. In certain embodiments, a host factor is encoded by a nucleic acid molecule listed in Table 11, Table 12, Table 13 or Table 14. In certain embodiments, a host factor is encoded by a nucleic acid molecule listed in Table 11. In certain embodiments, a host factor is encoded by a nucleic acid molecule listed in Table 12.
  • a host factor is encoded by a nucleic acid molecule listed in Table 13.
  • a host factor is encoded by a nucleic acid molecule listed in Table 14.
  • a host factor is encoded by a nucleic acid molecule having SEQ ID NO:79.
  • a host factor comprises amino acid sequence SEQ ID NO:80.
  • Another embodiment of the present invention is a method to identify a compound that modulates viral infection, the method comprising; (a) contacting a candidate compound with at least one isolated host factor under conditions suitable for allowing an interaction between the compound and the at least one host factor; and
  • the step of determining includes determining if the host factor has been altered by, for example, enzymatic cleavage, ligation, esterification, polymerization, phosphorylation, myristylation, acetylation, deacetylation, alkylation, glutamylation, hydroxylation, sulfation, or combinations thereof. Other types of modifications known by those skilled in the art are also encompassed.
  • a host factor is encoded by a nucleic acid molecule having a SEQ ID NO. listedn Table 1.
  • the host factor is encoded by a nucleic acid moleculeisted in Table 2.
  • a host factor is encoded by a nucleic acid molecule isted in Table 5. In certain embodiments, a host factor is encoded by a nucleic acid molecule isted in Table 5 or encoded by a nucleic acid molecule having a SEQ ID NO listed in Table
  • a host factor is encoded by a nucleic acid molecule listed in Table
  • a host factor is encoded by a nucleic acid molecule listed in Table 11. In certain embodiments, a host factor is encoded by a nucleic acid molecule listed in Table 12. In certain embodiments, a host factor is encoded by a nucleic acid molecule listed in Table 13. In certain embodiments, a host factor is encoded by a nucleic acid molecule listed in Table 14. In certain embodiments, a host factor is encoded by a nucleic acid molecule having SEQ ID NO:79. In certain embodiments, a host factor comprises amino acid sequence SEQ ID NO:80.
  • a host factor can possess activity, such as enzymatic activity.
  • one host factor identified using the methods disclosed herein is encoded by the AGP AT3 gene, which encodes a protein having O-acetyltransferase activity.
  • interaction with a compound can increase, decrease, or completely eliminate, the enzymatic activity of the host factor.
  • compounds that modulate viral infection are identified by contacting a host factor with a compound, and measuring the level of host factor-related activity. In such a method, the level of host factor-related activity is compared to the level of host factor-related activity measured in the absence of the compound.
  • Suitable assays for measuring host factor- related activity can be determined by those skilled in the art, and depend on the type of activity being measured.
  • candidate compounds that form a complex with a host factor, or that modify a host factor can be further assayed for their ability to modulate viral infection.
  • Such assays can be conducted in cells in culture or they can be conducted using whole animals. For example, the amount of virus produced by cells in culture in the presence and absence of a compound that interacts with a host factor can be compared.
  • a candidate compound that interacts with a host factor can be administered to an animal infected with a virus, and the amount of virus produced determined.
  • One embodiment of the present invention is a method to identify a compound that modulates viral infection, which comprises (a) combining a candidate compound with a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1 under conditions suitable for such candidate compound to interact with such host factor, (b) assaying for the presence of such interaction between such candidate compound and such host factor and (c) determining if such an interaction decreases or increases viral infectivity, wherein a decrease or increase in viral infectivity indicates that the compound modulates viral infection.
  • One embodiment is a method to identify a compound that modulates viral infection, which comprises (a) combining a candidate compound with a host factor encoded by a nucleic acid molecule listed in Table 5 or with a host factor encoded by a nucleic acid molecule having a SEQ ID NO: listed in Table 1 under conditions suitable for such candidate compound to interact with such host factor, (b) assaying for the presence of such interaction between such candidate compound and such host factor and (c) determining if such an interaction decreases or increases viral infectivity, wherein a decrease or increase in viral infectivity indicates that the compound modulates viral infection.
  • One embodiment is a method to identify a compound that modulates viral infection, which comprises (a) combining a candidate compound with a host factor encoded by a nucleic acid molecule listed in Table 5 under conditions suitable for such candidate compound to interact with such host factor, (b) assaying for the presence of such interaction between such candidate compound and such host factor and (c) determining if such an interaction decreases or increases viral infectivity, wherein a decrease or increase in viral infectivity indicates that the compound modulates viral infection.
  • viral infectivity is decreased, indicating that the compound inhibits viral infection.
  • viral infectivity is increased, indicating that the compound enhances viral infection.
  • Viral infectivity can be measured by determining the amount of virus, as described herein.
  • the interaction can be a binding interaction in which a complex is formed between the candidate compound and host factor or it can be an interaction in which the host factor is modified.
  • a host factor of the present invention is known to interact with existing compounds (e.g., drugs) used to treat non-HCV related disease. Some of these compounds have already been shown to modulate HCV infection.
  • the cholesterol-lowering drug simvastatin which is known to reduce the activity of the host factor diacylglycerol-2-acetyltransferase (Waterman, Diabetes (2002);51:1708-13), has been shown to inhibit HCV propagation.
  • simvastatin which is known to reduce the activity of the host factor diacylglycerol-2-acetyltransferase (Waterman, Diabetes (2002);51:1708-13)
  • the potential of many existing compounds to modulate HCV infection was not appreciated since, prior to the present invention, it was not known that the targets of these compounds (i.e., host factors that interact with these compounds) were involved in modulating HCV infection.
  • one embodiment of the present invention is a method to inhibit viral infection by contacting a cell infected with the virus or susceptible to the virus with a compound selected from Table 3.
  • Table 3 provides examples of host factors of the present invention, and pathways comprising such host factors, that appear to have some association with existing drugs used to treat non-HCV related disease.
  • one or more host factors can interact with each other as part of (or as an entire) pathway, and thus assays of the present invention can comprise more than one host factor. Therefore, the present invention also discloses pathways that modulate viral infectivity.
  • one embodiment of the present invention is a method to identify a compound that modulates viral infection, the method comprising assaying a candidate compound for the ability to modulate a pathway comprising a host factor of the present invention in such a manner as to modulate viral infection.
  • the virus is a hepatitis C virus.
  • the method identifies a compound that inhibits viral infection by assaying a candidate compound for the ability to modulate a pathway comprising a host factor of the present invention in such a manner as to inhibit viral infection.
  • the method identifies a compound that enhances viral infection by assaying a candidate compound for the ability to modulate a pathway comprising a host factor of the present invention in such a manner as to enhance viral infection.
  • the term pathway refers to a sequence of biochemical molecules and/or related activities (e.g., reactions) that result in the production of an end product.
  • the end product can be a biological molecule, such as a protein, or it can be an activity, such as an enzymatic activity (e.g., a phosphorylation event).
  • the glycolytic pathway consists of several enzymatic proteins, the end product of which is pyruvate.
  • the rumor growth factor beta (TGF-beta) signaling pathway comprises several proteins that interact in a sequential manner, resulting in regulation of gene expression.
  • a pathway of the present invention comprises at least one host factor of the present invention. It is appreciated by those of skill in the art that the end result of one pathway can be an activity that effects, or modulates, another pathway.
  • the interaction of biological pathways is referred to in the art as a network. Examples of pathways and networks of the present invention are shown in Figures 2, 3 and 4. Because of the inter-related nature of biological pathways, it will be appreciated by those of skill in the art that modulation of one pathway frequently results in modulation of one or more other pathways.
  • methods and compounds of the present invention can modulate viral infection by modulating a pathway that directly affects viral infection, or a pathway that indirectly affects viral infection.
  • a pathway that directly modulates viral infection is one in which the host factors that comprise the pathway directly interact with components of the virus.
  • a pathway that indirectly modulates viral infection is one in which the end result of the pathway does not directly interact with components of the virus, but affects another pathway that directly modulates viral infection.
  • to modulate a pathway refers to changing, varying, or altering the activity of that pathway by altering the amount of a host factor associated with the pathway, or the level of activity (e.g., enzymatic activity) resulting from a host factor or a pathway.
  • modulation can refer to an increase (e.g., up-regulation) or a decrease (e.g., down-regulation) in the amount of a host factor or activity.
  • the end result of modulating a host factor or pathway of the present invention is modulation of viral infection. In one embodiment, such viral infection is preferably inhibited. Table S3 lists pathways of the present invention.
  • the present invention also includes inhibitory compounds and enhancing compounds identified by the methods disclosed herein.
  • one embodiment of the present invention is an inhibitory compound for inhibiting infection by a virus of the present invention, wherein the inhibitory compound interacts with a host factor of the present invention, thereby inhibiting the viral infection.
  • an inhibitory compound can inhibit infection either by decreasing the amount or activity of a proviral host factor or by increasing the mount or activity of an antiviral host factor.
  • a further embodiment of the present invention s an inhibitory compound for inhibiting infection by a virus of the present invention, whereinhe inhibitory compound modulates a pathway comprising a host factor of the present nvention, thereby inhibiting viral infection.
  • an inhibitory compound cannhibit infection either by decreasing the activity of a pathway required for viral infection, or by increasing the activity of a pathway that inhibits viral infection.
  • Another embodiment of he present invention is an enhancing compound for enhancing infection by a virus of the present invention, wherein the enhancing compound interacts with a host factor of the present nvention, thereby enhancing the viral infection.
  • an enhancing compound can enhance infection either by increasing the amount or activity of a proviral host factor or by decreasing the amount or activity of an antiviral host factor.
  • a further embodiment of the present invention is an enhancing compound for enhancing infection by a virus of the present nvention, wherein the enhancing compound modulates a pathway comprising a host factor of he present invention, thereby enhancing viral infection.
  • an enhancing compound can enhance infection either by increasing the activity of a pathway required for viral infection, or by decreasing the activity of a pathway that inhibits viral infection.
  • useful compounds can be isolated from a natural source or they can be synthetic. Examples of useful inhibitory and enhancing compounds include, but are not imited to, proteins, peptides, antibodies, lipids, lipoproteins, nucleic acid molecules (e.g., microRNA and small interfering RNAs), an organic molecule, a synthetic molecule, small molecule compounds, and combinations thereof.
  • an inhibitory compound is an antibody that binds a host factor encoded by a nucleic acid molecule having a SEQ ID NO. listed in Table 1 in such a manner as to inhibit viral infection.
  • an enhancing compound is an antibody that binds a host factor encoded by a nucleic acid molecule having a SEQ ID NO. listed in Table 1 in such a manner as to enhance viral infection.
  • an inhibitory compound is an antibody that binds a host factor encoded by a nucleic acid molecule listed in Table 5 in such a manner as to inhibit viral infection.
  • an enhancing compound is an antibody that binds a host factor encoded by a nucleic acid molecule listed in Table 5 in such a manner as to enhance viral infection.
  • siRNAs small interfering RNA
  • RISC RNA-induced silencing complex
  • siRNAs useful for practicing the instant invention are listed in Table 4.
  • siRNAs can be proviral or antiviral, depending on the host factor they target.
  • a siRNA interacts with the same host factor as that targeted by a siRNA listed in Table 4 in such a manner as to inhibit viral infection.
  • the siRNA's can interact with the host factor at the same site, at overlapping sites, or at completely different sites.
  • an inhibitory compound of the present invention modulates a pathway comprising a host factor of the present invention by interacting with said host factor, thereby inhibiting viral infection.
  • an enhancing compound of the present invention modulates a pathway comprising a host factor of the present invention by interacting with said host factor, thereby enhancing viral infection.
  • Preferred host factors with which inhibitory compounds or enhancing compounds can interact are those encoded by a nucleic acid molecule listed in Table 1.
  • host factors with which inhibitory compounds or enhancing compounds can interact are those encoded by a nucleic acid molecule listed in Table 5.
  • host factors with which inhibitory compounds or enhancing compounds can interact are those encoded by a nucleic acid molecule listed in Table 1 or in Table 5.
  • the compound binds to the host factor with a Kd of at least 10 -4 , 10 -5 , 10 -6 , 10 '7 , 10 -8 , 10 '9 , 10 '10 , 10 -11 or 10 -12 .
  • the binding is irreversible.
  • the compound modifies the host factor thereby modulating its activity.
  • a compound that interacts with a host factor of the present invention reduces the amount of virus produced by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or at least 95%.
  • the amount of virus is reduced by a factor of at least 10, at least 100, at least 1000, or at least 10,000.
  • the viral infection is completely inhibited.
  • one embodiment of the present invention is a method to inhibit viral infection, the method comprising contacting a cell infected with the virus with an inhibitory compound that interacts with a host factor of the present invention in such a manner as to inhibit viral nfection.
  • the cell is infected with hepatitis C virus. In some circumstances it may be desirable to increase the amount of virus produced during annfection.
  • one embodiment of the present invention is a method to enhance viral infection, the method comprising contacting annfected cell with an enhancing compound that interacts with a host factor in such a manner as to increase the amount of virus produced.
  • the cell is infected with hepatitis C virus.
  • the term contacting refers to bringing the compound and the cell into proximity so that the compound is capable of interacting with a host factor of the present nvention.
  • Such contacting can be achieved by introducing the compound to the cell when he cell is in a tissue culture environment, or it can be achieved when the cell is present in a whole body. Consequently contacting the compound with the infected cell can be achieved hrough introducing the compound into a patient, for example, through an oral medication, an njection or other route of administration.
  • the compound can interact with and remain on outside of the cell, or it can enter the cell and interact with a host factor within the cell.
  • the end result of such contact is modulation of a pathway comprising a host factor of the present invention.
  • One embodiment of the present invention is a method to inhibit viral infection, the method comprising contacting a cell with an inhibitory compound that modulates a pathway comprising a host factor of the present invention in such a manner as to inhibit viral infection.
  • Another embodiment of the present invention is a method to enhance viral infection, the method comprising contacting a cell with an enhancing compound that modulates a pathway comprising a host factor of the present invention in such a manner as to enhance viral infection.
  • Suitable host factors are those encoded by a nucleic acid molecule having a SEQ ID NO. listed in Table 1.
  • Preferred host factors are those encoded by nucleic acid molecules listed in Table 2.
  • a host factor is encoded by a nucleic acid molecule listed in Table 5.
  • a host factor is encoded by a nucleic acid molecule listed in Table 5 or encoded by a nucleic acid molecule having a SEQ ID NO listed in Table 1.
  • a host factor is encoded by a nucleic acid molecule listed in Table .11, Table 12, Table 13 or Table 14.
  • a host factor is encoded by a nucleic acid molecule listed in Table 11.
  • a host factor is encoded by a nucleic acid molecule listed in Table 12.
  • a host factor is encoded by a nucleic acid molecule listed in Table 13.
  • a host factor is encoded by a nucleic acid molecule listed in Table 14.
  • a host factor s encoded by a nucleic acid molecule having SEQ ID NO:79.
  • a host factor comprises amino acid sequence SEQ ID NO: 80.
  • an inhibitory compound of the present invention is useful for nhibiting infection by a virus of the present invention. Consequently such a compound can be used to protect a patient from viral infection, such as hepatitis C infection. As used herein, he term protect refers to prophylactic as well as therapeutic use.
  • one embodiment of he present invention is a method to prevent viral infection in a patient capable of being nfected by a virus of the present invention by administering a pharmaceutical composition comprising an inhibitory compound that interacts with a host factor of the present invention n such a manner as to inhibit viral infection.
  • Another embodiment is a method to reduce the amount of virus in a patient infected with a virus, said method comprising administering a pharmaceutical composition comprising an inhibitory compound that interacts with a host factor of the present invention.
  • the patient is infected with Flavivirus.
  • the patient is nfected with hepatitis C virus.
  • Another embodiment is a method to protect a patient from virus infection, comprising administering to a patient an inhibitory compound that modulates a pathway comprising a host factor of the present invention in such a manner as to inhibit viral infection.
  • Suitable host factors are those encoded by a nucleic acid molecule having a SEQ ID NO. listed in Table 1.
  • a preferred host factor is one encoded by a nucleic acid molecule listed in Table 2.
  • a host factor is encoded by a nucleic acid molecule listed in Table 5.
  • a host factor is encoded by a nucleic acid molecule listed in Table 5 or encoded by a nucleic acid molecule having a SEQ ID NO listed in Table 1.
  • a host factor is encoded by a nucleic acid molecule listed in Table 11, Table 12, Table 13 or Table 14. In certain embodiments, a host factor is encoded by a nucleic acid molecule listed in Table 11. In certain embodiments, a host factor is encoded by a nucleic acid molecule listed in Table 12. In certain embodiments, a host factor is encoded by a nucleic acid molecule listed in Table 13. In certain embodiments, a host factor is encoded by a nucleic acid molecule listed in Table 14. In certain embodiments, a host factor is encoded by a nucleic acid molecule having SEQ ID NO:79. In certain embodiments, a host factor omprises amino acid sequence SEQ ID NO:80. In a preferred embodiment, the patient isnfected with hepatitis C virus.
  • inhibitory compounds include, but are not limited to, inhibitory ompounds that inhibit viral entry, viral translation, viral replication or viral trafficking, assembly or release. In certain embodiments, an inhibitory compound inhibits viral entry. In ertain embodiments, an inhibitory compound inhibits viral translation. In certain embodiments, an inhibitory compound inhibits viral replication. In certain embodiments, an nhibitory compound inhibits viral trafficking, assembly or release. Examples of preferred nhibitory compounds are listed in Table 3. Table 3. Drugs and Small Molecule Inhibitors that Affect Pathways and Host Factors of the Present Invention
  • T) the product of the gene is the direct drug target
  • a preferred inhibitory compound is a siRNA, such as those listed in Table 4.
  • an inhibitory compound can be colchicine, cytochalasin, EIPA, rottlerin, UO 126, wortmannin or a mixture of two or more of such compounds, hi certain embodiments, an inhibitory compound can be brefeldin A, golgicide A or a mixture thereof. LQ certain embodiments, an inhibitory compound can be colchicine. In certain embodiments, an inhibitory compound can be, cytochalasin. In certain embodiments, an inhibitory compound can be EIPA. In certain embodiments, an inhibitory compound can be rottlerin. In certain embodiments, an inhibitory compound can be UO 126. In certain embodiments, an inhibitory compound can be wortmannin. hi certain embodiments, an inhibitory compound can be brefeldin A. In certain embodiments, an inhibitory compound can be golgicide A.
  • a pharmaceutical composition comprises an inhibitory compound that interacts with a host factor of the present invention and a pharmaceutically acceptable vehicle, such as a diluent, carrier, excipient, adjuvant or a combination thereof. Selection of such a vehicle is known to those skilled in the art.
  • a pharmaceutically acceptable vehicle such as a diluent, carrier, excipient, adjuvant or a combination thereof. Selection of such a vehicle is known to those skilled in the art.
  • the term "patient” refers to an animal infected with a virus of the present invention. The animal can be a human or a non-human animal. A preferred animal o treat is a mammal.
  • a compound of the present invention, or a pharmaceutical composition thereof can be administered to a patient by a variety of routes, including, but limited to, by injection (e.g., ntravenous, intramuscular, subcutaneous, intrathecal, intraperitoneal), by inhalation, by oral e.g., in a pill, tablet, capsule, powder, syrup, solution, suspension, thin film, dispersion or emulsion.), transdermal, transmucosal, pulmonary, buccal, intranasal, sublingual,
  • ntracerebral intravaginal rectal or topical administration or by any other convenient method known to those of skill in the art.
  • the amount of a compound of the present invention and/or a pharmaceutical composition thereof that will be effective can be determined by standard clinical techniques known in the art. Such an amount is dependent on, among other factors, the patient being reated, including, but not limited to the weight, age, and condition of the patient, the ntended effect of the compound, the manner of administration and the judgment of the prescribing physician.
  • a compound of the present invention can be administered alone or in combination with one or more other pharmaceutical agents, ncluding other compounds of the present invention.
  • a compound can be administered or applied per se or as pharmaceutical compositions.
  • the specific pharmaceutical composition depends on the desired mode of administration, as is well known to the skilled artisan.
  • a patient is protected using a composition comprising an inhibitory compound that inhibits viral infection by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or at least 95%.
  • the amount of virus is reduced by a factor of at least 10, 100, 1000, or 10,000. In one embodiment the viral infection is completely inhibited.
  • kits to identify a compound that modulates viral infection One embodiment is a kit to identify an inhibitory compound, the kit comprising a means for determining if a candidate compound is capable of inhibiting viral infection by interacting with a host factor of the present invention.
  • Another embodiment is a kit to identify an enhancing compound, the kit comprising a means for determining if a candidate compound is capable of enhancing viral infection by interacting with a host factor of the present invention.
  • Kits can also comprise means for identifying both inhibitory compounds and enhancing compounds. Suitable and preferred means are disclosed herein.
  • a kit can also comprise host factors of the present invention.
  • the kit can also contain associated reagents and components, such as, but not limited to, buffers, labels, containers, inserts, tubings, vials, syringes and the like.
  • amino acids can be denoted by either the tandard 3 -letter or 1 -letter code.
  • RNAi screen To identify host factors required for HCV infection, a high-throughput RNAi- based screen was undertaken on an arrayed library targeting 19,470 genes (Dharmacon iARRAY siRNA Library (Human Genome, G-005000-05, Thermo Fisher Scientific, Lafayette, CO, the remaining 1,651 pools of the original 21,121 gene library were not creened because the vast majority have been retired due to revised refseq annotation).
  • siRNAs were transiently transfected into the Huh 7.5.1 cells at a 50 nM final concentration, using a reverse transfection protocol employing 0.44% Oligofectamine (Invitrogen, Carlsbad, CA) in a 384-well format (384 well, black plastic, clear bottomed assay plate, Corning 3712). The Oligofectamine was diluted in Opti-MEM (Invitrogen) and allowed to incubate ten minutes. The lipid solution was then aliquoted into the wells (9 ul/well) using a Wellmate liquid handing robot (Thermo Fisher Matrix).
  • the plates were spun down at 1,000 RPM and the arrayed siRNAs were added robotically, 1.5 ⁇ L of a 1 ⁇ M stock solution per well. After a twenty minute incubation, 800 Huh 7.5.1 cells were added per well, in 20 ⁇ L of Dulbecco's modified minimal essential media (DMEM, Invitrogen), supplemented with 15% fetal bovine serum (FBS, Invitrogen). The plates were next spun at 1000 RPM and then placed in a tissue culture incubator at 37 0 C and 5% CO2. The subsequent day, 5 ⁇ L of fresh complete media (10% FBS) was added to the outer two wells at the plate margins to decrease edge effects.
  • DMEM Dulbecco's modified minimal essential media
  • FBS fetal bovine serum
  • the "part one" cells were then fixed with 4% Formalin, permeabilized with 0.3% Triton-XIOO (Sigma) in Dulbecco's Phosphate Buffered Saline (D-PBS, nvitrogen) containing 3% FBS and 3% bovine serum albumin Fraction V (BSA, Sigma Aldrich), then stained for HCV core, using purified anti-HCV core monoclonal antibody produced from the anti-HCV core 6G7 hybridoma cells generously provided by Drs. Harry Greenberg and Xiaosong He, Stanford University) diluted in D-PBS with 1% BSA.
  • D-PBS Dulbecco's Phosphate Buffered Saline
  • BSA bovine serum albumin Fraction V
  • the cells were then incubated with an Alexa 488 goat anti-mouse secondary at 1:1,000 in D-PBS with 1% BSA (Al 1001, Invitrogen), and stained for DNA with Hoechst 33342 at 1 :5,000-10,000 Invitrogen). Each step was followed by two washes with 30 ⁇ L D-PBS. The cells were then maged on an automated Image Express Micro (IXM) microscope (Molecular Devices) at 4X magnification, using two wavelengths, 488 nm to detect HCV infected cells expressing core, and 350 nm for nuclear DNA bound by Hoechst 33342.
  • IXM Image Express Micro
  • HCV RNA was extracted with QIAamp Viral RNA Mini Kit (Qiagen, Valencia, CA) from 140 ⁇ L of culture medium, or with RNeasy Mini Kit (Qiagen) from whole cell lysate. Copy numbers of HCV RNA were determined by quantitative PCR with the probe and primers listed below, using the TaqMan EZ RT-PCR CORE REAGENTS (Applied Biosystems) on an ABI 7500 Real Time PCR System (Applied Biosystem).
  • PCR parameters consisted of 1 cycle of 50°C X 2 min, then 60 °C X 30 min, and 95°C X 3:30 min, followed by 50 cycles of PCR at 95 °C X 0 s, and 62 °C X 1 min.
  • the relative amount of HCV RNA was normalized to the internal ontrol Human 18S rRNA (Applied Biosystems).
  • HCV TaqMan probe used was 6-FAM CTGCGGAACCGGTGAGTACACTAMRA (IDT, Coralville, IA).
  • HCV primer sequences were 5' CGGGAGAGCCATAGTGG, and 3' AGTACCACAAGGCCTTTCG.
  • RNA was prepared from whole cell lysate with RNeasy Mini Kit according to manufacturer's instructions.
  • Complementary DNA cDNA was synthesized from total RNA with First Strand cDNA Synthesis Kit (Roche, ndianapolis, IN).
  • the mRNA expression levels of CD81 and ApoE were quantified by quantitative PCR using a TaqMan Gene Express Master Mix (Applied Biosystems) on an ABI 7500 Real Time PCR System. PCR parameters were 1 cycle of 50°C X 2 min, then 95°C X 10 min, followed by 50 cycles of PCR at 95 °C X 15 s, and 60 °C X 1 min.
  • RNA screen Part one: siRNAs were transiently transfected into the Huh 7.5.1 cells at a 50 nM final concentration, using 0.44% Oligofectamine (Invitrogen, Carlsbad, CA) in a 384- well format (384 well, black plastic, clear bottomed assay plate, Corning 3712).
  • siRNA pools were classified as hits (decreased infection) if the average of the riplicate plates showed that the percentage of core positive cells was less than 50% of the plate mean, and cell number was not less than 50% of the mean of the plate. Pools which ncreased infection by greater than 150% of the plate mean were also selected as hits increased infection).
  • Preferred host factors identified by the disclosed screens are listed in Table 2. Table 2. Preferred Host Factors
  • siRNA pools were considered validated if two or more of the individual oligos scored (50% or less infected cells (decreased infection) or 150% or greater infected cells (increased infection)) as compared to the negative control wells on the plate, in either both part one and two or part two alone, and the cell number was not less than 40% of the average of the negative control wells on the plate.
  • the percent of infected cells relative to controls, as well as the normalized cell numbers for each of the individual genes that confirmed with two or more siRNAs is provided in Table Sl. Visual spot inspections of control images were done throughout the screen to confirm the accuracy of the automated imaging and cell scoring systems.
  • Huh 7.5.1 cells were grown in DMEM supplemented with 10% FBS.
  • siRNA screen hits were classified into molecular function and biological process categories according to the Panther classification system, and statistical enrichment of hese categories were assessed for the set of hit genes relative to their representation in the global et of genes examined in the siRNA screen. P-values were computed using the hypergeometric distribution, implemented in the R statistical computing language.
  • Networks were generated with data extracted from interactome screens and urated literature entries in HPRD (Keshava (2009) Nucleic Acids Res 37, D767-772.) and proteome-wide mapping efforts from the I-MAP project examining HCV-host protein nteractions (de Chassey (2008) MoI Syst Biol 4, 230.). Network constructions were implemented n the Perl programming language and incorporate graph theoretical representations by abstracting gene products as nodes and interactions as edges.
  • This creen utilizes the HCVcc system, which consists of the infectious JFH-I genotype 2a virus and he Huh 7.5.1 human hepatocellular cell line (Brass (2008) Science 319, 921-926.).
  • Huh 7.5.1 cells were transfected with small siRNAs for 72 hours, and then challenged with JFH-I virus. After 48 h the cells were stained and imaged for expression of the HCV core protein as a marker * for productive viral infection (Fig IA).
  • Fig IA productive viral infection
  • genes functioning in either anti-viral responses or safeguarding the cells against the stress of infection may be detected in this screen.
  • the screen was optimized using siRNAs against the host proteins, CD81 (part one) and ApoE (part two).
  • ApoE a host protein involved in lipoprotein biosysnthesis, has been shown to e required for infectious particle formation (Hao (2008) Nature 454, 890-893; Huang (2007) Proc Natl Acad Sci U S A 104, 5848-5853; Chang (2007) J Virol 81, 13783-13793.).
  • siRNAs gainst CD81 resulted in inhibition of part one infection (5-6 fold, Fig IA, SlA, B).
  • siRNAs argeting ApoE did not affect infection in part one, but did inhibit HCV infection in art two by 2-3 fold, in keeping with its previously assigned role in infectious particle formation Fig IA, SlA, B).
  • the levels of infection were verified using quantitative RT-PCR and found to e directly proportional to the levels determined microscopically (Fig Sl C, D).
  • the image-based latform was then used for a genetic screen with a commercially available siRNA whole-genome brary (Dharmacon siGenome, 19,470 pools of four siRNAs per gene).
  • NUAK/SNARK an AMPK- elated kinase functioning in the cellular stress response, was one of nine host factors found by Ng et. al. to be necessary for HCV replicon function in a siRNA screen targeting -4,000 genes Ng (2007) Hepatology 45, 1413-1421; Kuga (2008) Biochem Biophys Res Commun 317, 1062- 066.).
  • ribonucleoprotein complex omponents e.g. RBM22, SRRM2, UBA52
  • transcription factors e.g. BRFl, E2F2, RCC5, FOXE3, MLXIPL, SMAD5, SMAD6, TRRAP, WWTRl.
  • CypA is the likely target of the anti-HCV non-immunosuppressive cyclosporin analogue, Debio-025, thus demonstrating that functional genomics can identify high-yield leads for host-directed anti-viral therapies (HDAVs) (Flisiak (2008) Hepatology 47, 817-826.).
  • HDAVs host-directed anti-viral therapies
  • TCOFl physically associates with nucleolar proteins, and is the gene responsible for Treacher Collins Syndrome, a severe congenital craniofacial developmental disease (Sakai D (2009) Int J Biochem Cell Biol 41, 1229-1232.).
  • Knockout mice deficient in TCOFl display ribosome biogenesis deficiencies suggesting that cells depleted in TCOFl may be unable to meet the increased demand for protein production required by HCV (Sakai (2009) Int J Biochem Cell Biol 41, 1229-1232.).
  • Cherry et al. demonstrated that the IRES-mediated transcription of the picornavirus, Drosophila C, was dependent on the levels of specific ribosomal sub-units (Cherry (2005) Genes Dev 19, 445-452.). Decreased levels of nucleolar factors may adversely impact HCV IRES function by altering ribosomal subunit levels or modifications.
  • the Ccr4-Not complex is a global regulator of gene expression that functions in transcription and polyadenylation (Collart (2004) Prog Nucleic Acid Res MoI Biol 77, 289-322.). Mutation of each of the components of the orthologous yeast Ccr4-Not complex has shown that the each subunit regulates the expression of a largely unique set of genes (Azzouz (2009) RNA 15, 377- 383; Cui (2008) MoI Genet Genomics 279, 323-337.). Therefore, the components identified in this screen may control distinct sets of HCV host factors.
  • I-MAP HCV infection mapping project
  • HCV 3 WNV and dengue virus belong to the Flaviviridae family, whose members include the human pathogens Japanese encephalitis virus and yellow fever virus (Murray (2008) Nat Rev Microbiol 6, 699-708; Lindenbach (2003) Adv Virus Res 59, 23-61.).
  • Rab9p40 functions in the movement of late endosomes to the trans Golgi network (TGN), and along with its interaction partners, Rab9 and PIKfyve, is required for the particle assembly of multiple enveloped viruses (HFV, Marburg, Ebola and measles viruses), though the mechanism remains poorly understood (Murray (2008) Nat Rev Microbiol 6, 699-708; Murray (2005) J Virol 79, 11742-11751; Shisheva (2008) Cell Biol Int 32, 591-604.). This study, and that of Tai et al., now add HCV to the list of Rab9p40-dependent viruses.
  • TGN trans Golgi network
  • siRNA Library and Functional Genomic Screens Genes identified by the genome- wide siRNA screen in Example 1 served as a starting point for defining the comprehensive interactions between HCV and host.
  • the life cycle of HCV infection broadly encompasses viral entry, intracellular trafficking, viral RNA translation and replication, and virion assembly and release. Each of these steps engages in a complex set of host cell machineries.
  • High-throughput screens with various in vitro HCV model systems were undertaken using a custom-designed arrayed library targeting these 232 genes (Dharmacon ON-TARGETplus SMARTpool siRNA library).
  • ON- TARGETplus pools were used to significantly reduce off-target effects while maintaining siRNA potency, and hence provide a highly effective tool for validation and functional stratification following primary screen with the siGENOME SMARTpools.
  • siRNAs were transiently transfected into the Huh7.5.1 cells at a final concentration of 50 nM, using a transfection protocol employing 0.44% Oligofectamine (Invitrogen) in a 96-well format. Briefly, this protocol is as follows: Oligofectamine was diluted in Opti-MEM (Invitrogen) and incubated for 10 min before being distributed into wells using a Wellmate liquid handling robot (Thermo Fisher Matrix). Following distribution, the plates were spun down at 1,000 RPM and the arrayed siRNAs added.
  • Oligofectamine was diluted in Opti-MEM (Invitrogen) and incubated for 10 min before being distributed into wells using a Wellmate liquid handling robot (Thermo Fisher Matrix). Following distribution, the plates were spun down at 1,000 RPM and the arrayed siRNAs added.
  • the plates were incubated for 20 minutes, after which 4,500 Huh 7.5.1 cells, in 60 ⁇ L Dulbecco's modified minimal essential media (DMEM, Invitrogen) supplemented with 15% FBS (FBS, Invitrogen), were added to each well.
  • DMEM Dulbecco's modified minimal essential media
  • FBS FBS, Invitrogen
  • the plates were spun at 1,000 RPM and then placed in a humidified tissue culture incubator at 37°C and 5% CO 2 . After 72 h of incubation, to allow for siRNA-mediated gene knockdown, the medium was removed and the cells were infected with the JFH-I genotype 2a virus, at an MOI of 0.5 in 100 niL DMEM with 10% FBS.
  • the cells were then incubated with an Alexa Fluor 488 goat anti-mouse secondary (A-11001, Invitrogen) at 1:1,000 in D-PBS with 1% BSA, and stained for DNA with Hoechst 33342 at 1:5,000-10,000 (Invitrogen). Each step was followed by two washes with D-PBS.
  • the cells were then imaged on an automated Image Express Micro (IXM) microscope (Molecular Devices) at 4x magnification, using two wavelengths: 488 nm to detect HCV infected cells expressing core, and 350 nm for nuclear DNA bound by Hoechst 33342. Images were then analyzed using the Metamorph Cell Scoring software program (Molecular Devices Inc.) to determine the total cells per well, and the percentage of core positive cells in each well (percent infected).
  • IXM Image Express Micro
  • the 80 ml of medium removed from the infected cells in part A were replica plated onto a new 96-well plate, which contained 10,000 Huh 7.5.1 cells per well plated out 24 h before viral supernatant addition. At 48 h post-infection, the cells were fixed, stained, and imaged using the staining and imaging protocol described in part A.
  • HCV RNA Quantification Infected cells were prepared as described above in parts A and B. The level of extracellular and intracellular HCV RNA was determined as follows: Total RNA was extracted with QIAamp Viral RNA Mini Kit (Qiagen) from 140 mL culture medium, or with RNeasy Mini Kit (Qiagen) from whole-cell lysate. Copy numbers of HCV RNA were determined by quantitative PCR with the probe and primers listed below, using the TaqMan EZ RT-PCR CORE REAGENTS (Applied Biosystems) on an ABI 7500 Real Time PCR System (Applied Biosystems).
  • PCR parameters consisted of 1 cycle of 50°C for 2 min, then 60°C for 30 min, and 95°C for 3:30 min, followed by 50 cycles of PCR at 95°C for 20 s, and 62°C for 1 min.
  • the relative amount of HCV RNA was normalized to the internal control Human 18S rRNA (Applied Biosystems).
  • HCV TaqMan probe used was 6-FAM
  • HCV primer sequences were 5' CGGGAGAGCCATAGTGG, and 3' AGTACCAC AAGGCCTTTCG.
  • siRNA pools were classified as proviral or antiviral hits if the average normalized HCV RNA levels decreased or increased by more than 1.5 -fold comparing with non-targeting control siRNA and the P value was less than 0.05. The results of these experiments are shown in Table 7.
  • HCVpp HCV pseudoparticles
  • VSVpp vesicular stomatitis virus pseudoparticles
  • Virus entry was assessed two days after infection by measuring the firefly luciferase reporter activities in cell lysate using the Bright GIo Luciferase assay system (Promega. This screen identified eight HCV-specific entry factors and 15 common factors for the entry of HCV and VSV. The results of these experiments are shown in Table 8 & 11.
  • HCV subgenomic replicon assay Huh 7.5.1 cells were transfected with siRNA using the protocol described in part A. After 48 hours, SGR-Luc- JFH-I subgenomic replicon RNA, which contains the luciferase gene, was transiently transfected into the Huh7.5.1 cells. After 72 hours, cells were lysed directly in 96-well plates and Renilla luciferase reporter activities were measured using Renilla Luciferase assay system (Promega) to determine levels of HCV RNA replication.
  • siRNA pools were classified as proviral or ICT antiviral hits if the average normalized HCV subgenomic replicon activities were decreased or increased for more than 1.5 -fold comparing with non-targeting control siRNA and the P value was less than 0.05.
  • 52 human genes were identified to be important for HCV RNA replication. These genes are listed in Table 9. As shown in Table 13, 17 genes restrict HCV infection at the RNA replication level.
  • HCV IRES-luciferase assay was used to identify host factors associated with HCV IRES-mediated translation. Briefly, Huh7.5.1 cells were transfected with siRNA as described in part A. After 48 hours, the transfected cells were transiently transfected with pHCV-CLX-CMV WT RNA. Cell lysate was prepared at 24 h following HCV RNA transfection, and assayed for firefly luciferase reporter activities using Luciferase assay system (Promega). siRNA pools were classified as pro viral or antiviral hits if the average normalized firefly luciferase activities were decreased or increased for more than 1.5 -fold comparing with non-targeting control siRNA and the P value was less than 0.05. These results are shown in Table 10. Table 10. Host Factors Modulating HCV Translation
  • HCVcc HCV cell culture system
  • RNAi-based mammalian genetics A major concern of RNAi-based mammalian genetics is false negatives, stemming in part from inefficient targeting or nonspecific toxicity.
  • bioinformatics meta-analyses after the selection of our validated gene set, which integrated several HCV host factor data sets and disclosed statistically enriched complexes and pathways. By performing hypothesis-driven follow- up studies of these highlighted pathways, we were able to identify 22 additional host factors that were not previously identified by our primary screen. These host factors are listed in Table 16.
  • IKK ⁇ was a strongly positive hit as a proviral factor.
  • IKK ⁇ is an IKB kinase that plays a predominant role in non-canonical NFKB activation pathway.
  • Innate immunity mediated by activation of NF- ⁇ B is a fundamental early signaling step of host antiviral defense.
  • This Example demonstrates an NF- ⁇ B-independent function of IKK ⁇ in hepatitis C virus infection and propagation.
  • siRNA screen was performed as described in Example 1. Briefly, siRNA's for the IKK ⁇ gene were transiently transfected into Huh 7.5.1 cells and after 72 hours, the cells infected with hepatitis C virus. AT 48 hours post-infection, 80 ml of media were removed and used for "late-stage" experiments. After removal of the media, the infected cells were fixed and stained with anti-HCV core 6G7 monoclonal antibody, as described in Example 1. The results of this experiment are shown in Figure 5(A). The data show that siRNA silencing of IKK ⁇ production resulted in a marked decrease of HCV production in Huh7.5 cells.
  • Huh7.5.1 cells were transfected with siRNA's targeting these specific factors, and then infected with HCV, as described in Example 1. Intracellular and extracellular HCV RNA levels were then determined at 48 hours post-infection, using the protocol described in Example 1. The results of this experiment, which are shown in Figure 6, demonstrate that siRNA silencing of IKK ⁇ , and two IKK ⁇ upstream activators, NF- ⁇ B inducing kinase (NIK) and lymphotoxin- ⁇ (LTB), resulted in a marked decrease of HCV RNA production in Huh 7.5. cells.
  • NIK NF- ⁇ B inducing kinase
  • LTB lymphotoxin- ⁇
  • NS5B but not other HCV proteins interacts specifically with and activates IKK ⁇ .
  • the activated IKK ⁇ translocates to the nucleus and induces a CREB- binding protein (CBP)-mediated transcriptional program that includes SREBPl, a critical transcriptional factor controlling lipid metabolism and biogenesis of lipid droplets.
  • CBP CREB- binding protein
  • SREBPl was identified as a proviral factor in the genome- wide siRNA screen and siRNA knock-down of SREBPl displayed the same phenotype as IKK ⁇ silencing in HCV infection.
  • Our study demonstrates that HCV has evolved a mechanism to exploit the host innate defense for its own advantage.
  • Huh7.5.1 cells were transiently transfected with a pool of siRNAs, or individual siRNAs, against IKK ⁇ , and then infected with HCV. At 48 hours post-infection, the levels of intracellular and extracellular HCV RNA were measured using quantitative PCR, as described in Example 2(C). CD81, a major receptor of HCV, was used as a positive control for the early stage of the HCV lifecycle. ApoE, which is required for infectious viral particle formation, was the positive control for the late stage. The results are shown in Figure 8.
  • HCV pseudovirus and replicon assays described in Example 2, parts D and E, respectively, were used to measure the effect of IKK ⁇ silencing on viral entry and replication.
  • Positive siRNA controls of CD81 and PIK4CA were used for the HCVpp and HCV replicon assays, respectively.
  • This Example demonstrates the ability of certain compounds to inhibit HCV entry.
  • Huh7.5.1 cells were infected with HCVpp in increasing concentrations of compounds as indicated, and assayed for luciferase activities 48 h later. The results, which are shown in Figure 10, show that HCV entry was inhibited by these compounds, i.e., rotterlin, colchicines, EIPA, wormannin, cytochalasin, and UO 126.
  • Huh7.5.1 cells were infected with HCVcc in the presence of varying concentrations of the Rottlerin and EIPA. After 4 h of infection, the culture medium was removed, the cells washed to remove the inoculating virus, fresh medium added and the cells incubated for 48 hours. At the end of the incubation period, HCV RNA levels in the culture supernatant were determined as described in Example 2(C). The results, which are shown in Figure 11, indicate that both drugs inhibited HCV infection.
  • HCV replication involves formation of membranous webs, which are sites of RNA replication.
  • the present work identified several ADP-ribosylation factors and related proteins as proviral factors.
  • Brefeldin A and golgicide A are compounds that have been shown to interfere with the functions of these factors in membrane-associated mechanisms of secretory and endocytic pathways.
  • Huh7.5.1 cells were infected with HCVcc in the presence of increasing concentrations of the compounds for 4 h and the culture supernatant was then removed with washing to remove the inoculating virus. Infected cells were treated with the same concentrations of the compounds for an additional 48 h and the culture supernatant was harvested for HCV RNA determination. The results, which are shown in Figure 12, indicate that both drugs inhibit HCV infection.
  • the present work enabled the reconstruction of an in-depth network map of cellular pathways and machineries that are associated with the complete life cycle of HCV (summarized in Table 1).
  • a comprehensive investigation of HCV-host interactions yields critical insights into HCV pathogenesis and valuable targets for prophylactic and therapeutic interventions.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biotechnology (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Public Health (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne la découverte selon laquelle de nombreux facteurs produits par la cellule hôte modulent (c'est-à-dire, augmentent ou diminuent) l'infection par le virus de l'hépatite C (VHC). De tels facteurs endogènes peuvent être des facteurs endogènes proviraux ou des facteurs endogènes antiviraux. La présente invention concerne également un procédé d'utilisation de ces facteurs endogènes pour identifier des composés modulant une infection virale. L'invention concerne également des composés inhibiteurs ainsi que des composés amplificateurs qui modulent une infection virale. De tels composés inhibiteurs ou amplificateurs agissent sur les facteurs endogènes de la présente invention, ou sur les voies biochimiques comprenant de tels facteurs endogènes. L'invention concerne également des procédés d'inhibition ou d'amplification d'une infection virale, utilisant de tels composés inhibiteurs ou de tels composés amplificateurs, respectivement, ainsi que des procédés de protection d'un patient d'une infection virale par l'administration de tels composés inhibiteurs au patient.
PCT/US2010/040918 2009-07-02 2010-07-02 Procédés de modulation d'une infection par le virus de l'hépatite c WO2011003071A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US22283309P 2009-07-02 2009-07-02
US61/222,833 2009-07-02
US22849609P 2009-07-24 2009-07-24
US61/228,496 2009-07-24

Publications (1)

Publication Number Publication Date
WO2011003071A1 true WO2011003071A1 (fr) 2011-01-06

Family

ID=43411479

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/040918 WO2011003071A1 (fr) 2009-07-02 2010-07-02 Procédés de modulation d'une infection par le virus de l'hépatite c

Country Status (1)

Country Link
WO (1) WO2011003071A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020218577A1 (fr) * 2019-04-26 2020-10-29 ネオファーマジャパン株式会社 Agent thérapeutique pour une infection à flavivirus
CN113769093A (zh) * 2021-09-09 2021-12-10 中国人民解放军海军军医大学 古蛋白1在制备预防或治疗乙型脑炎病毒感染药物中的应用
JP2022514657A (ja) * 2018-12-21 2022-02-14 アイオニス・ファーマシューティカルズ・インコーポレイテッド Hsd17b13発現のモジュレータ
EP3873488A4 (fr) * 2018-10-31 2022-08-03 The University of Sydney Compositions et méthodes de traitement d'infections virales

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050107386A1 (en) * 2003-11-19 2005-05-19 Rama Krishna Narla Methods of treating diseases and disorders by targeting multiple kinases
US20080044413A1 (en) * 2005-12-21 2008-02-21 Hammond Scott A EphA2 BiTE molecules and uses thereof
US20080161324A1 (en) * 2006-09-14 2008-07-03 Johansen Lisa M Compositions and methods for treatment of viral diseases

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050107386A1 (en) * 2003-11-19 2005-05-19 Rama Krishna Narla Methods of treating diseases and disorders by targeting multiple kinases
US20080044413A1 (en) * 2005-12-21 2008-02-21 Hammond Scott A EphA2 BiTE molecules and uses thereof
US20080161324A1 (en) * 2006-09-14 2008-07-03 Johansen Lisa M Compositions and methods for treatment of viral diseases

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3873488A4 (fr) * 2018-10-31 2022-08-03 The University of Sydney Compositions et méthodes de traitement d'infections virales
JP2022514657A (ja) * 2018-12-21 2022-02-14 アイオニス・ファーマシューティカルズ・インコーポレイテッド Hsd17b13発現のモジュレータ
JP7394137B2 (ja) 2018-12-21 2023-12-07 アイオニス・ファーマシューティカルズ・インコーポレイテッド Hsd17b13発現のモジュレータ
WO2020218577A1 (fr) * 2019-04-26 2020-10-29 ネオファーマジャパン株式会社 Agent thérapeutique pour une infection à flavivirus
CN113769093A (zh) * 2021-09-09 2021-12-10 中国人民解放军海军军医大学 古蛋白1在制备预防或治疗乙型脑炎病毒感染药物中的应用
CN113769093B (zh) * 2021-09-09 2023-01-17 中国人民解放军海军军医大学 古蛋白1在制备预防或治疗乙型脑炎病毒感染药物中的应用

Similar Documents

Publication Publication Date Title
Lee et al. The SARS-CoV-2 RNA Interactome
Suzuki et al. Characterization of RyDEN (C19orf66) as an interferon-stimulated cellular inhibitor against dengue virus replication
Arita et al. Oxysterol-binding protein family I is the target of minor enviroxime-like compounds
Li et al. Cellular microRNA networks regulate host dependency of hepatitis C virus infection
Ramage et al. A combined proteomics/genomics approach links hepatitis C virus infection with nonsense-mediated mRNA decay
Ariumi et al. Hepatitis C virus hijacks P-body and stress granule components around lipid droplets
Garaigorta et al. Hepatitis C virus (HCV) induces formation of stress granules whose proteins regulate HCV RNA replication and virus assembly and egress
Carbajo-Lozoya et al. Replication of human coronaviruses SARS-CoV, HCoV-NL63 and HCoV-229E is inhibited by the drug FK506
Raychoudhuri et al. ISG56 and IFITM1 proteins inhibit hepatitis C virus replication
Poenisch et al. Identification of HNRNPK as regulator of hepatitis C virus particle production
Ishida et al. Alterations in microRNA expression profile in HCV-infected hepatoma cells: involvement of miR-491 in regulation of HCV replication via the PI3 kinase/Akt pathway
Katoh et al. Heterogeneous nuclear ribonucleoprotein A2 participates in the replication of Japanese encephalitis virus through an interaction with viral proteins and RNA
Gaither et al. Multiple cyclophilins involved in different cellular pathways mediate HCV replication
Fusco et al. A genetic screen identifies interferon-α effector genes required to suppress hepatitis C virus replication
Li et al. Lyn kinase regulates egress of flaviviruses in autophagosome-derived organelles
Colpitts et al. Use of a tandem affinity purification assay to detect interactions between West Nile and dengue viral proteins and proteins of the mosquito vector
Murillo et al. Identification of host cell factors associated with astrovirus replication in Caco-2 cells
Li et al. The yin and yang of hepatitis C: synthesis and decay of hepatitis C virus RNA
Dechtawewat et al. Role of human heterogeneous nuclear ribonucleoprotein C1/C2 in dengue virus replication
Guo et al. A conserved inhibitory mechanism of a lycorine derivative against enterovirus and hepatitis C virus
Shi et al. Inhibition of hepatitis C virus production by aptamers against the core protein
Wang et al. Multiple functions of heterogeneous nuclear ribonucleoproteins in the positive single-stranded RNA virus life cycle
Nag et al. Suppression of viral RNA binding and the assembly of infectious hepatitis C virus particles in vitro by cyclophilin inhibitors
Giovannoni et al. Cellular promyelocytic leukemia protein is an important dengue virus restriction factor
Yi et al. Affinity purification of the hepatitis C virus replicase identifies valosin-containing protein, a member of the ATPases associated with diverse cellular activities family, as an active virus replication modulator

Legal Events

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

Ref document number: 10794818

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10794818

Country of ref document: EP

Kind code of ref document: A1