WO2011071535A2 - Compositions et procédés pour inhiber des facteurs cellulaires d'hôtes humains nécessaires pour la réplication du virus de la grippe - Google Patents

Compositions et procédés pour inhiber des facteurs cellulaires d'hôtes humains nécessaires pour la réplication du virus de la grippe Download PDF

Info

Publication number
WO2011071535A2
WO2011071535A2 PCT/US2010/003138 US2010003138W WO2011071535A2 WO 2011071535 A2 WO2011071535 A2 WO 2011071535A2 US 2010003138 W US2010003138 W US 2010003138W WO 2011071535 A2 WO2011071535 A2 WO 2011071535A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
influenza virus
host cell
human subject
fold
Prior art date
Application number
PCT/US2010/003138
Other languages
English (en)
Other versions
WO2011071535A3 (fr
Inventor
Megan Shaw
Silke Stertz
Adolfo Garcia-Sastre
Peter Palese
John Young
Renate Konig
Sumit Chanda
Original Assignee
Mount Sinai School Of Medicine Of New York University
Salk Institute For Biological Studies
Sanford-Burnham Medical Research Institute
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 Mount Sinai School Of Medicine Of New York University, Salk Institute For Biological Studies, Sanford-Burnham Medical Research Institute filed Critical Mount Sinai School Of Medicine Of New York University
Priority to US13/514,783 priority Critical patent/US9238815B2/en
Publication of WO2011071535A2 publication Critical patent/WO2011071535A2/fr
Publication of WO2011071535A3 publication Critical patent/WO2011071535A3/fr

Links

Classifications

    • 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
    • C12N15/1131Non-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 against viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/18Sulfonamides

Definitions

  • This application relates to the modulation of host cell factors required for influenza virus replication.
  • the application relates to compounds, including nucleic acid compounds (such as, e.g., small interfering RNAs (siRNAs)) and small molecules, that target human host cell factors involved in influenza virus replication, and the use of such compounds for modulating influenza virus replication and as antiviral agents.
  • the application also relates to methods of treating an influenza virus infection and methods of treating or preventing a symptom or disease associated with influenza virus infection, comprising administering to a subject a composition comprising a compound, such as a nucleic acid compound (e.g., an siRNA) or small molecule, that targets a human host cell factor involved in influenza virus replication.
  • a nucleic acid compound e.g., an siRNA
  • Influenza viruses are enveloped RNA viruses that belong to the family of
  • Influenza A and B viruses are considered to be major human pathogens and in a normal season they can cause between 3-5 million cases of severe illness and up to 500,000 deaths worldwide (World Health Organization, 2003).
  • Influenza A viruses can also cause pandemics such as those that occurred in 1918, 1957 and 1968. These outbreaks resulted in high mortality rates because of the lack of pre-existing immunity against the new virus strain.
  • rimantadine block the M2 ion channel of the virus and prevent the release of the viral genome into the host cell (Pinto and Lamb, 1995; Wharton et al., 1994). These drugs are effective if used prophylactically and if administered within 48 hours of infection but are not effective against influenza B viruses. However, the development of widespread resistance has precluded the use of adamantanes in recent influenza seasons (Bright et al. , 2006) and isolates of the H5N1 influenza virus have been shown to be resistant to these drugs due to mutations in M2 (Cheung et al, 2006).
  • NA neuraminidase
  • the present application is based, in part, on the discovery that influenza virus replication can be reduced by pharmacologically targeting human host cell factors required for viral replication.
  • Targeting host cell factors, rather than the viral factors required for influenza virus replication, may greatly reduce the emergence of viral resistance and expands the number of targets for antiviral intervention.
  • compositions including pharmaceutical compositions, comprising such compounds, and methods of using such compounds and compositions for modulating influenza virus replication.
  • the compounds and compositions comprising them reduce or inhibit influenza virus replication.
  • methods of using such compounds and compositions for reducing or inhibiting influenza virus replication the compounds modulate influenza virus replication by altering the expression (e.g., mRNA or protein) and/or activity of the human host cell factor involved in influenza virus replication.
  • the compounds reduce or inhibit influenza virus replication by reducing or inhibiting the expression (e.g., mRNA or protein) and/or activity of the human host cell factor involved in influenza virus replication.
  • the human host cell factor interacts with a component of the influenza virus.
  • the human host cell factor is required for influenza virus replication.
  • the compounds provided herein, and for use in the compositions and methods provided herein target human host cell factors involved in influenza virus replication and modulate influenza virus replication.
  • the compounds provided herein, and for use in the compositions and methods provided herein target human host cell factors involved in influenza virus replication and reduce or inhibit influenza virus replication.
  • the targeted human host cell factor may be required for influenza virus replication.
  • the targeted human host cell factor may be involved in or required for one or more of the following events of the influenza virus life cycle: entry; uncoating; nuclear import; viral RNA transcription; or viral RNA translation.
  • the targeted human host cell factor may be involved in or required for replication of more than one strain or sub-type of influenza.
  • the human host cell factor may be involved in or required for replication of an influenza A virus, an influenza B virus, and/or an influenza C virus.
  • the human host cell factor is involved in or required for replication of a human-origin, an avian-origin (e.g., H5N1), and/or a swine-origin (e.g., H1N1) influenza virus.
  • compositions and methods provided herein targets a component or regulator of, or factor that interacts with, one or more of the following categories of human host cell factors:
  • cytoskeleton cytoskeleton; ribonucleoprotein; spliceosome; ubiquitin/proteasome system; ribosome or other translation machinery; kinase; phosphatase; signaling (e.g., G-protein coupled receptors; signaling at the plasma membrane); mitochondrion or mitochondrial ribosome; plasminogen; stress response; v-ATPase; ion channel or other ion transport; nucleus;
  • a compound provided herein, and for use in the compositions and methods provided herein targets a component or regulator of, or a factor that interacts with, one or more of the following categories of human host cell factors: IP3-PKC pathway; COPI vesicles; endosomal uptake, maturation, acidification, and fusion; actin organization and function; PI3 -AKT pathway; endosomal recycling pathway; MAPK pathway; proteases; calcium/calmodulin system;
  • the compound reduces or inhibits the expression and/or activity of the human host cell factor.
  • the compounds provided herein, and for use in the compositions and methods provided herein reduce or inhibit influenza virus replication.
  • the compound is an agent that reduces or inhibits the expression (e.g., mR A or protein) and/or activity of a human host cell factor involved in influenza virus replication.
  • the human host cell factor is required for influenza virus replication.
  • the compound reduces or inhibits the interaction of a human host cell factor with a component of the influenza virus.
  • the compound reduces or inhibits one or more of the following events of the influenza viral life cycle: entry; uncoating; nuclear import; viral RNA transcription; or viral RNA translation.
  • the compound reduces or inhibits replication of more than one strain or sub-type of influenza.
  • the compound may reduce or inhibit replication of influenza virus A, an influenza B virus, and/or an influenza C virus.
  • the compound reduces or inhibits replication of a human-origin, an avian-origin (e.g., H5N1), and/or a swine-origin (e.g., H1N1) influenza virus.
  • the compound reduces or inhibits replication of an influenza virus and one or more other viruses.
  • the compound modulates the expression and/or activity of one or more of the human host cell factors listed in Table 3. In specific embodiments, the compound reduces or inhibits the expression and/or activity of one or more of the human host cell factors listed in Table 3.
  • the compound modulates the expression and/or activity of one or more of the human host cell factors listed in Table 7. In specific embodiments, the compound reduces or inhibits the expression and/or activity of one or more of the human host cell factors listed in Table 7.
  • the compound modulates the expression and/or activity of one or more of the human host cell factors listed in Table 9. In specific embodiments, the compound reduces or inhibits the expression and/or activity of one or more of the human host cell factors listed in Table 9.
  • the compound modulates the expression (e.g., mRNA or protein) and/or activity of one or more of the following human host cell factors: ACRC; AKAP13; AKT1; ANAPC2; ANPEP; ARCN1 ; BRWD3; CAD; CAMK2B; CANT1 ;
  • CBLL1 CD81 ; CHAF1A; CL 1 ; CLOCK; COPA; COPB1 ; COPB2; COPG; CSE1L;
  • CTSW CTSW; DTX2; DUSP3; EPHB2; EPS8L3; F13A1 ; FAM135A; FGFR2; FGFR4; FPR1, FRAP1 (mTOR); GABBR1 ; GRK6; GSK3B; HAND2; HIST3H3; HSP90AA1 ; IL1F9;
  • PRSS35 PSMD1; RAB1 IB; RBM5; RP1 1 -45B20.2; RPS10; RPS20; SF3A1 ; SNRPA1 ; STK31 ; STK39; STX10; SUM02; SUM04; TBK1 ; TEAD3; TNP03; TRPV2; TUBB;
  • the compound reduces or inhibits the expression and/or activity of one or more of the aforementioned human host cell factors.
  • the compound modulates the expression (e.g., mRNA or protein) and/or activity of one or more of the following human host cell factors: AKAP13; ARCN; BRWD3; CD81 ; COPG; CTSW; DUSP3; EPHB2; FAM135A; FGFR2; FGFR4; GABBR1 ; GSK3B; ITGA3; JA 2; MAP2K3; NEK6; RABl IB; or one or more of the v- ATPase subunits, ATP6V0B, ATP6V0C, ATP6V1A, ATP6V1B2, or ATP6AP1.
  • human host cell factors e.g., mRNA or protein
  • the compound modulates the expression (e.g., mRNA or protein) and/or activity of one or more of the following human host cell factors: CAMK2B; CSE1L; F13A1 ; KPNBl ; MAP3K12; PP1R14D; PRSS35; RPSIO; SF3A1 ; or SUM04.
  • CAMK2B CSE1L
  • F13A1 a human host cell factor
  • KPNBl e.g., MAP3K12
  • PP1R14D e.g., MAP3K12
  • PP1R14D e.g., PP1R14D
  • PRSS35 e.g., PRSS35
  • RPSIO SF3A1
  • SUM04 e.g., SUM04.
  • the compound modulates the expression (e.g., mRNA or protein) and/or activity of one or more of the following human host cell factors: ACRC; DTX2; EPS8L3; FPR1; MAP3K11 ; NUP214; PRPH2; RP1 1-45B20.2; STX10; SUM02; TRPV2; or TUBB.
  • the compound modulates the expression (e.g., mRNA or protein) and/or activity of one or more of the following human host cell factors: ANPEP; CAM2KB; FGFR4; FRAP1 (mTOR); GSK3B/CSNK1 G2; HSP90AA1 ; or TUBB.
  • the compound reduces or inhibits the expression and/or activity of one or more of the aforementioned human host cell factors.
  • the compound may be any compound described herein, known in the art, or yet to be discovered that targets one or more of the aforementioned categories of human host cell factors, a specific factor(s) in such a category, and/or one of the aforementioned human host cell factors. In certain embodiments, the compound is not toxic to the human host cell.
  • the compound does not target AKTl , ARCN1 , COPG, GRK6, HAND2, HIST3H3, an HSP90 (e.g., HSP90AA1), NUP153, RBM5, RPS10, RPS20, or a v-ATPase subunit.
  • the compound does not reduce or inhibit the expression and/or activity of AKTl, ARCN1, COPG, GRK6, HAND2, HIST3H3, an HSP90 (e.g., HSP90AA1), NUP153, RBM5, RPS10, RPS20, or a v-ATPase subunit.
  • the compound does not target AKAP13, CD81, CAMK2B, CSE1L, DUSP3, FGFR2, FGFR4, GSK3B, ITGA3, KPNBl, MAP2K3, or RABl IB. In certain embodiments, the compound does not reduce or inhibit the expression and/or activity of AKAP13, CD81, CAMK2B, CSE1L, DUSP3, FGFR2, FGFR4, GSK3B, ITGA3, KPNBl, MAP2K3, or RABl IB.
  • the compound is a nucleic acid compound.
  • the nucleic acid compound is an siRNA.
  • the nucleic acid compound has a sequence optimized for use as an siRNA, according to methods known in the art.
  • the nucleic acid compound is an antisense compound.
  • the nucleic acid compound is a modified oligonucleotide.
  • the nucleic acid compound is contained within a larger nucleic acid compound, such as a plasmid.
  • the nucleic acid compound comprises an oligonucleotide of 12 to 30 linked nucleosides, for example, 12 to 15, 15 to 20, 20 to 25, e.g., 21 or 25 nucleosides, or 26 to 30 linked nucleosides, which may be targeted to a nucleic acid encoding a human host cell factor involved in influenza virus replication.
  • the human host cell factor involved in influenza virus replication is a human host cell factor described supra. Any region of the human host cell factor gene or mRNA may be targeted as provided for herein and known to one of skill in the art.
  • the compound targets a nucleotide sequence selected from Table 1 (see also Table 9) (Section 7 below).
  • the nucleobases represented by a "U” (uracil) in a sequence in Table 1 may be replaced with thymine nucleobases (represented by a "T”).
  • the nucleobases represented by a "T” (thymine) in a sequence in Table 1 may be replaced with uracil nucleobases (represented by a "U”).
  • the nucleotide sequence "AAGTAGGGATAAATTACTCTA” in Table 1 may be replaced with the nucleotide sequence "AAGUAGGGAUAAAUUACUCUA.”
  • the nucleic acid compound targeting a sequence in Table 1 is an antisense compound.
  • the nucleic acid compound targeting a sequence in Table 1 is an siRNA.
  • the siRNA that targets one of the aforementioned human host cell factors or sequences is obtained from a commercially available source.
  • the siRNA can be from Qiagen (Druggable Set version 1 or 2), NM Set version 1 , XM Set version 1 , the kinome library from Invitrogen or the kinome library from IDT.
  • an siRNA duplex is created from a 21mer sequence in Table 1 as exemplified in the following example:
  • RNA ribonucleic acid
  • the siRNA duplexes based on the sequences in Table 1 that contain Us are created the same way, except that the sequence is already an RNA; i.e., the sequence in Table 1 containing Us correspond to host cell mRNA targets.
  • the siRNA compound comprises the sequence /5Phos/rGrGrCrUrArCrGrGrArCrCrArArGrUrUrArUrCrCrGrGCG. This sequence is the sense sequence for a 25mer siRNA duplex for use in accordance with the embodiments described herein.
  • the compound is a small molecule.
  • the small molecule is Betulinic acid (available from VWR International/Enzo Life Sciences Intl.); CCT018159 (4-(4-(2,3-Dihydro- 1 ,4-benzodioxin-6-yl)-5-methyl- 1 H-pyrazol-3-yl)-6- ethylresorcinol; available from Calbiochem); Diphyllin (available from Sigma; see Fig. 13a); the FGF VEGF receptor inhibitor 4-Hydroxy-3-benzimidazol-2-ylhydroquinolin-2-one; Hymenialdisine (available from Biomol International LP); KN-93 (available from
  • Podophyllotoxin Podophyllinic Acid Lactone
  • the compound is not CCT018159. In some embodiments, the compound is not Diphyllin.
  • compositions comprising a compound that targets one or more human host cell factors involved in influenza virus replication. Such compositions may be in a dose effective to modulate influenza virus replication. Such compositions may be in a dose effective to reduce or inhibit influenza virus replication.
  • Such compositions may be pharmaceutical compositions, and may additionally comprise a pharmaceutically acceptable carrier known in the art or described herein. Such pharmaceutical compositions may be in a dose effective to treat influenza virus infection or to reduce or inhibit a symptom or disease associated with influenza virus infection.
  • compositions and pharmaceutical compositions may include, by non-limiting example, (i) a compound that targets an aforementioned category of human host cell factor; (ii) a compound that targets a human host cell factor in such a category; (iii) a compound that targets an aforementioned human host cell factor; (iv) an aforementioned nucleic acid compound, such as an siRNA, optionally in an appropriate delivery vehicle; or (v) an aforementioned small molecule.
  • Such compositions may also include another active agent, for example, another compound that targets a human host cell factor involved in influenza virus replication described herein.
  • compositions including the pharmaceutical compositions, described herein contain the compound in an amount that is not significantly toxic to the cell, tissue, or subject for which it is intended.
  • Methods of testing toxicity include any method known in the art, for example, as described in Sections 5 and 6 infra.
  • a method for reducing or inhibiting replication of an influenza virus comprises: (a) infecting a cell with an influenza virus; and (b) contacting the cell with such a compound or composition in an amount sufficient to reduce or inhibit replication of the influenza virus. Also provided herein are methods for reducing or inhibiting influenza virus replication, comprising: (a) contacting a cell with such a compound or composition in an amount sufficient to reduce or inhibit replication of an influenza virus; and (b) infecting the cell with the influenza virus.
  • a compound or composition comprising the compound is considered to reduce or inhibit influenza virus replication if it reduces the amount of influenza virus replication as measured compared to a control, such as, for example, influenza virus replication in the absence of the compound or composition, or influenza virus replication in the presence of a negative control.
  • the compound or composition is contacted to a cell at risk for influenza virus infection.
  • Compounds for use in such methods may include, by non-limiting example, (i) a compound that targets an aforementioned category of human host cell factor; (ii) a compound that targets a human host cell factor in such a category; (iii) a compound that targets an aforementioned human host cell factor; (iv) an aforementioned nucleic acid compound, such as an siRNA, optionally in an appropriate delivery vehicle; or (v) an aforementioned small molecule.
  • a pharmaceutical composition comprising a compound, e.g., nucleic acid compound (e.g., siRNA) or small molecule, that targets one or more human host cell factors involved in influenza virus replication in an amount sufficient to reduce the influenza virus infection.
  • a compound e.g., nucleic acid compound (e.g., siRNA) or small molecule
  • the subject is a human.
  • Compounds for use in such methods may include, by non-limiting example, (i) a compound that targets an aforementioned category of human host cell factor; (ii) a compound that targets a human host cell factor in such a category; (iii) a compound that targets an aforementioned human host cell factor; (iv) an aforementioned nucleic acid compound, such as an siRNA, optionally in an appropriate delivery vehicle; or (v) an aforementioned small molecule.
  • a compound that targets an aforementioned category of human host cell factor a compound that targets a human host cell factor in such a category
  • a compound that targets an aforementioned human host cell factor e.g., an aforementioned nucleic acid compound, such as an siRNA, optionally in an appropriate delivery vehicle
  • an aforementioned small molecule e.g., siRNA, optionally in an appropriate delivery vehicle.
  • compositions comprising a compound, e.g., nucleic acid compound (e.g., siRNA) or small molecule, that targets one or more human host cell factors involved in influenza virus replication in an amount sufficient to reduce the symptom or disease associated with the influenza virus infection.
  • a compound e.g., nucleic acid compound (e.g., siRNA) or small molecule, that targets one or more human host cell factors involved in influenza virus replication in an amount sufficient to reduce the symptom or disease associated with the influenza virus infection.
  • the subject is infected with an influenza virus.
  • the subject is at risk for infection with an influenza virus.
  • the subject is a human.
  • Compounds for use in such methods may include, by non-limiting example, (i) a compound that targets an influenza virus.
  • aforementioned category of human host cell factor (ii) a compound that targets a human host cell factor in such a category; (iii) a compound that targets an aforementioned human host cell factor; (iv) an aforementioned nucleic acid compound, such as an siRNA, optionally in an appropriate delivery vehicle; or (v) an aforementioned small molecule.
  • Also provided herein are methods for preventing a symptom or disease associated with an influenza virus infection comprising administering to a subject in need thereof a composition comprising a compound, e.g., nucleic acid compound (e.g., siRNA) or small molecule, that targets one or more human host cell factors involved in influenza virus replication in an amount sufficient to prevent or reduce the symptom or disease associated with the influenza virus infection.
  • a compound e.g., nucleic acid compound (e.g., siRNA) or small molecule
  • the subject is infected with an influenza virus.
  • the subject is at risk for infection with an influenza virus.
  • the subject is a human.
  • Compounds for use in such methods may include, by non-limiting example, (i) a compound that targets an aforementioned category of human host cell factor; (ii) a compound that targets a human host cell factor in such a category; (iii) a compound that targets an aforementioned human host cell factor; (iv) an aforementioned nucleic acid compound, such as an siRNA, optionally in an appropriate delivery vehicle; or (v) an aforementioned small molecule.
  • the compounds, compositions, and pharmaceutical compositions are used in an amount that is not
  • Methods of testing toxicity include any method known in the art, for example, as described in Sections 5 and 6 infra.
  • the aforementioned methods may optionally comprise use of the compound that targets a human host cell factor involved in influenza virus replication in combination with one or more additional active agents.
  • additional active agents include, for example, one or more additional antiviral agents, e.g., an aforementioned compound that targets human host cell factors involved in influenza virus replication; an antibiotic; an immunomodulatory agent; or an agent used in the treatment or prophylaxis of one or more pulmonary diseases described herein (see, e.g., Section 5) or known in the art.
  • the subject is a human.
  • the influenza virus is an influenza A virus.
  • the influenza virus is an influenza B virus.
  • the influenza virus is an influenza C virus. Any type, subtype, or strain of influenza virus described herein or known in the art may be targeted in accordance with the embodiments described herein.
  • the influenza virus is of human origin.
  • the influenza virus is of avian origin (e.g. , H5N1).
  • the influenza virus is of swine origin (e.g., H1N1).
  • the compound or composition may have broad antiviral utility, e.g., it modulates replication of an influenza virus and one, or two, or three, or four, or five, or more additional viruses known in the art or yet to be discovered.
  • the term "2'-0-methoxyethyl” refers to an O-methoxy-ethyl modification of the 2' position of a furosyl ring.
  • a 2'- O-methoxyethyl modified sugar is a modified sugar.
  • the term "2'-0- methoxyethyl nucleotide” means a nucleotide comprising a 2'-0-methoxyethyl modified sugar moiety.
  • 5-methylcytosine means a cytosine modified with a methyl group attached to the 5' position.
  • a 5-methylcytosine is a modified nucleobase.
  • the term “antisense compound” means an oligomeric compound that is capable of undergoing hybridization to a target nucleic acid through hydrogen bonding.
  • the term “antisense inhibition” means reduction of target nucleic acid levels in the presence of an antisense compound complementary to the target nucleic acid compared to target nucleic acid levels in the absence of the antisense compound.
  • the term “antisense oligonucleotide” means a single-stranded oligonucleotide having a nucleobase sequence that permits hybridization to a corresponding region or segment of a target nucleic acid.
  • chimeric antisense compound means an antisense compound that has at least 2 chemically distinct regions, each position having a plurality of subunits.
  • bicyclic sugar means a furosyl ring modified by the bridging of two non-geminal ring atoms. A bicyclic sugar is a modified sugar.
  • bicyclic nucleic acid or “BNA” refers to a nucleoside or nucleotide wherein the furanose portion of the nucleoside includes a bridge connecting two carbon atoms on the furanose ring, thereby forming a bicyclic ring system.
  • cap structure or "terminal cap moiety” means chemical modifications, which have been incorporated at either terminus of an antisense compound.
  • complementarity means the capacity for pairing between nucleobases of a first nucleic acid and a second nucleic acid.
  • mismatch or “non-complementary nucleobase” means a nucleobase of first nucleic acid that is not capable of pairing with the corresponding nucleobase of a second or target nucleic acid.
  • the term "compound,” unless otherwise specified or apparent from the context, refers to any agent described herein that modulates, reduces, or inhibits influenza virus replication, including the compounds and structures provided herein or incorporated by reference herein, and solvates, hydrates, prodrugs, stereoisomers and pharmaceutically acceptable salts thereof.
  • RNA interference RNA interference
  • a compound is one of the compounds identified in Section 5 below.
  • a compound is purified.
  • a compound is purified.
  • an "effective amount" in the context of administering a treatment to a subject refers to the amount of a treatment which has a prophylactic and/or therapeutic effect(s).
  • an "effective amount" in the context of administration of a treatment to a subject refers to the amount of a treatment which is sufficient to achieve one, two, three, four, or more of the following effects: (i) reduce or ameliorate the severity of a viral infection or a symptom or disease associated therewith; (ii) reduce the duration of a viral infection or a symptom or disease associated therewith; (iii) reduce or prevent the progression of a viral infection or a symptom or disease associated therewith; (iv) cause regression of a viral infection or a symptom or disease associated therewith; (v) prevent the development or onset of a viral infection or a symptom or disease associated therewith; (vi) reduce or prevent the recurrence of a viral infection or a symptom or disease associated therewith; (vii)
  • hybridization means the annealing of complementary nucleic acid molecules.
  • complementary nucleic acid molecules include, but are not limited to, an antisense compound or oligonucleotide and a nucleic acid target or the paired strands of an siRNA molecule.
  • the term "specifically hybridizable” means when there is a sufficient degree of complementarity between an antisense compound and a target sequence to avoid non-specific binding of the antisense compound to non-target nucleic acid sequences under conditions in which specific binding is desired, i.e., under physiological conditions in the case of in vivo assays or therapeutic treatment, and under conditions in which assays are performed in the case of in vitro assays.
  • the term "in combination,” in the context of the administration of two or more treatments or therapies to a subject, refers to the use of more than one compound or composition, e.g., more than one prophylactic agent and/or therapeutic agent.
  • the two compounds may be formulated together in a single composition.
  • the use of the term “in combination” does not restrict the order in which therapies are administered to a subject with a viral infection.
  • a first therapy (e.g., a first prophylactic or therapeutic agent) can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 16 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 16 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapy to a subject with a viral infection.
  • a second therapy e.g., a first prophylactic or therapeutic agent
  • an infection means the invasion by, multiplication and/or presence of a virus in a cell, tissue, or subject.
  • an infection is an "active" infection, i.e., one in which the virus is replicating in a cell, tissue, or subject.
  • Such an infection may be characterized by the spread of the virus to other cells, tissues, organs, and/or subjects from the cells, tissues, organs, and/or subjects initially infected by the virus.
  • An infection may also be a latent infection, i.e., one in which the virus is not replicating.
  • an infection refers to the pathological state resulting from the presence of the virus in a cell, tissue, or subject, or by the invasion of a cell, tissue, or subject by the virus.
  • a compound that inhibits or reduces viral replication reduces viral replication by at least 1.5 fold, 2, fold, 3, fold, 4 fold, 5 fold, 6 fold, 7 fold, 8 fold, 9 fold, 10 fold, 15 fold, 20 fold, 25 fold, 30 fold, 35 fold, 40 fold, 45 fold, 50 fold, 100 fold, 500 fold, or 1000 fold relative to virus replication in the absence of compound or the presence of a negative control.
  • the compound reduces virus replication by at least 2 log relative to virus replication in the absence of compound or the presence of a negative control.
  • the compound reduces virus replication by 1.5 to 3 fold, 2 to 4 fold, 3 to 5 fold, 4 to 8 fold, 6 to 9 fold, 8 to 10 fold, 2 to 10 fold, 5 to 20 fold, 10 to 40 fold, 10 to 50 fold, 25 to 50 fold, 50 to 100 fold, 75 to 100 fold, 100 to 500 fold, 500 to 1000 fold, or 10 to 1000 fold.
  • the compound reduces the virus replication by approximately 2 logs or more, approximately 3 logs or more, approximately 4 logs or more, approximately 5 logs or more, or 2 to 10 logs or 2 to 5 logs relative to virus replication in the absence of compound or the presence of a negative control.
  • a decrease in viral replication is measured using an assay described in Section 5 or Section 6, infra. In some embodiments, a decrease in viral replication is screened for using a library of compounds. In one embodiment, a decrease in viral replication is measured by: (a) contacting a compound or a member of a library of compounds with a cell before (e.g., 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 16 hours, 24 hours or more before), concurrently and/or subsequent to (e.g., 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 16 hours, 24 hours or more after) infection with the virus; and (b) measuring virus replication.
  • the cells used in the assay should be susceptible to infection by the chosen virus and can be infected at different MOIs.
  • the effect of a compound on virus replication can be assessed by measuring virus replication at different times post-infection. For example, virus replication may be measured 6 hours, 12 hours, 16 hours, 24 hours, 48 hours or 72 hours post-infection, using any method known to one of skill in the art can be used measure virus replication.
  • a decrease in viral replication is assessed by measuring viral titer (as determined, e.g. , by plaque formation).
  • a decrease in viral replication is assessed by measuring the production of viral proteins (as determined, e.g., by Western blot analysis, ELISA or flow cytometry).
  • a decrease in viral replication is assessed by measuring the production of viral nucleic acids (as determined, e.g., by RT-PCR or Northern blot analysis) using techniques known to one of skill in the art. See Sections 5 and 6 below for more details of techniques for measuring viral replication.
  • viral replication is measured using a virus engineered to contain a reporter, such as the Renilla luciferase virus described in Section 6.
  • a compound is considered to decrease viral replication if it reduces the amount of viral replication as measured compared to a control, such as, for example, viral replication in the absence of the compound or viral replication in the presence of a negative control.
  • library in the context of compounds refers to a plurality of compounds.
  • a library can be a combinatorial library, e.g., a collection of compounds synthesized using combinatorial chemistry techniques, or a collection of unique chemicals with a low molecular weight (less than 1000 Daltons).
  • log refers to logio
  • a subject is administered one or more treatments to "manage" a disease so as to prevent the progression or worsening of the viral infection.
  • modified internucleoside linkage refers to a substitution or any change from a naturally occurring internucleoside linkage (i.e. a phosphodiester internucleoside bond).
  • naturally occurring internucleoside linkage means a 3' to 5' phosphodiester linkage.
  • modified nucleobase means any nucleobase other than adenine, cytosine, guanine, thymine, or uracil.
  • An "unmodified nucleobase” means the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U).
  • modified nucleotide means a nucleotide having, independently, a modified sugar moiety, modified internucleoside linkage, or modified nucleobase.
  • modified nucleoside means a nucleoside having, independently, a modified sugar moiety or modified nucleobase.
  • modified oligonucleotide means an oligonucleotide comprising a modified internucleoside linkage, a modified sugar, or a modified nucleobase.
  • modified sugar refers to a substitution or any change from a natural sugar.
  • motif means the pattern of unmodified and modified nucleosides in an antisense compound.
  • MOI multiplicity of infection
  • the MOI is determined by dividing the number of virus added (ml added x PFU) by the number of cells added (ml added x cells/ml).
  • natural sugar means a sugar found in DNA (2'-H) or RNA (2'-OH).
  • nucleic acid refers to a molecule composed of monomelic nucleotides.
  • a nucleic acid includes, but is not limited to, ribonucleic acids (RNA), deoxyribonucleic acids (DNA), single-stranded nucleic acids, double-stranded nucleic acids, small interfering ribonucleic acids (siRNA), and microRNAs (miRNA).
  • nucleobase means a heterocyclic moiety capable of pairing with a base of another nucleic acid.
  • nucleobase sequence means the order of contiguous nucleobases independent of any sugar, linkage, or nucleobase modification.
  • nucleoside means a nucleobase linked to a sugar.
  • nucleotide means a nucleoside having a phosphate group covalently linked to the sugar portion of the nucleoside.
  • oligomeric compound means a polymer of linked monomeric subunits which is capable of hybridizing to at least a region of a nucleic acid molecule.
  • oligonucleoside means an oligonucleotide in which the internucleoside linkages do not contain a phosphorus atom.
  • oligonucleotide means a polymer of linked nucleosides each of which can be modified or unmodified, independent one from another.
  • the term "pharmaceutically acceptable salt” refers to a salt of a compound prepared from a pharmaceutically acceptable acid or base including, but not limited to an inorganic acid, an inorganic base, an organic acid, or an organic base.
  • Suitable pharmaceutically acceptable base addition salts of the compounds include, but are not limited to metallic salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc or organic salts made from lysine, ⁇ , ⁇ '-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine.
  • Suitable acids include, but are not limited to, inorganic and organic acids such as acetic, alginic, anthranilic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethenesulfonic, formic, fumaric, furoic, galacturonic, gluconic, glucuronic, glutamic, glycolic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phenylacetic, phosphoric, propionic, salicylic, stearic, succinic, sulfanilic, sulfuric, tartaric, and p-toluenesulfonic acid.
  • inorganic and organic acids such as acetic, alginic, anthranilic, benzenesulfonic, benzoic, camphorsulfonic, citric,
  • the pharmaceutically acceptable salt is a hydrochloride or a mesylate salt.
  • Others are well-known in the art. See for example, Remington's Pharmaceutical Sciences, 18th eds., Mack
  • hydrate means a compound, or a pharmaceutically acceptable salt thereof, that further includes a
  • solvate means a compound, or a pharmaceutically acceptable salt thereof, that further includes a
  • phosphorothioate internucleoside linkage means a linkage between nucleosides where the phosphodiester bond is modified by replacing one of the non-bridging oxygen atoms with a sulfur atom.
  • a phosphorothioate linkage is a modified internucleoside linkage.
  • the terms "prevent,” “preventing” and “prevention” in the context of the administration of a treatment to a subject to prevent a viral infection or a symptom or disease associated with a viral infection refer to one or more of the following effects resulting from the administration of a treatment or a combination of treatments: (i) the inhibition of the development or onset of a viral infection and/or a symptom or disease associated therewith; (ii) the inhibition of the recurrence of a viral infection and/or a symptom or disease associated therewith; and.or (iii) delaying or forestalling the onset of a viral infection and/or a symptom or disease associated therewith.
  • prodrug means a compound derivative that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide the compound.
  • prodrugs include, but are not limited to, derivatives and metabolites of a compound that include biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues.
  • prodrugs of compounds with carboxyl functional groups are the lower alkyl esters of the carboxylic acid.
  • the carboxylic esters are conveniently formed by esterifying any of the carboxylic acid moieties present on the molecule.
  • Prodrugs can typically be prepared using well-known methods, such as those described by Burger's Medicinal Chemistry and Drug Discovery 6th ed. (Donald J. Abraham ed., 2001 , Wiley) and Design and Application of Prodrugs (H. Bundgaard ed., 1985,
  • prophylactic refers to use of an agent in the prevention of a viral infection or a symptom or disease associated therewith.
  • the prophylactic agent does not result in the complete prevention of the viral infection or symptom or disease associated therewith.
  • a prophylactic agent is an agent which is known to be useful to or has been or is currently being used to prevent or impede the onset and/or development of a viral infection or a symptom or disease associated therewith.
  • prophylactically effective amount refers to the amount of a treatment (e.g., with a prophylactic agent) which is sufficient to prevent a viral infection or a symptom or disease associated therewith in a subject.
  • a prophylactically effective amount is the amount of a compound that reduces the incidence of a viral infection in a subject.
  • the incidence of a viral infection in a subject is reduced by at least 2.5%, at least 5%, at least 10%, at least 15%, at least 25%, at least 35%, at least 45%, at least 50%, at least 75%, at least 85%, by at least 90%, at least 95%, or at least 99% in a subject administered a compound relative to a subject or group of subjects (e.g., two, three, five, ten or more subjects) not administered the compound.
  • the term "purified,” in the context of a compound that is chemically synthesized, refers to a compound that is substantially free of chemical precursors or other chemicals when chemically synthesized.
  • the compound is 60%, preferably 65%, 70%, 75%, 80%, 85%, 90%, or 99% free of other, different
  • cellular materials from the natural source such as but not limited to cell debris, cell wall materials, membranes, organelles, the bulk of the nucleic acids,
  • substantially free of natural source materials refers to preparations of a compound that has been separated from the material (e.g., cellular components of the cells) from which it is isolated.
  • a compound that is isolated includes preparations of a compound having less than about 30%, 20%, 10%), 5%, 2%, or 1% (by dry weight) of cellular materials and/or contaminating materials.
  • a "purified” or “isolated” nucleic acid sequence or nucleotide sequence such as an siRNA, miRNA, shRNA, or a vector construct for producing such a molecule, can be substantially free of other cellular material or culture medium when produced by recombinant techniques, or substantially free of chemical precursors when chemically synthesized.
  • an "isolated" nucleic acid sequence or nucleotide sequence is a nucleic acid sequence or nucleotide sequence that is recombinantly expressed in a heterologous cell.
  • replication refers to one or more, or all, of the stages of a viral life cycle which result in infection with or propagation of virus.
  • the steps of a viral life cycle include, but are not limited to, virus attachment to the host cell surface, penetration or entry of the host cell (e.g., through receptor mediated endocytosis or membrane fusion), uncoating (the process whereby the viral capsid is removed and degraded by viral enzymes or host enzymes thus releasing the viral genomic nucleic acid), genome replication, synthesis of viral messenger RNA (mRNA), viral protein synthesis, and assembly of viral ribonucleoprotein complexes for genome replication, assembly of virus particles, post-translational modification of the viral proteins, and release from the host cell by lysis or budding and acquisition of a phospholipid envelope which contains embedded viral glycoproteins.
  • the terms “replication,” “viral replication” and “virus replication” refer to the replication of the viral genome. In other embodiments, the terms “replication,” “viral replication” and “virus replication” refer to the synthesis of viral proteins. [0078] As used herein, the term “single-stranded oligonucleotide” means an oligonucleotide which is not hybridized to a complementary strand.
  • RNA interference refers to the process of sequence-specific post transcriptional gene silencing in mammals mediated by siRNAs (see, e.g., Fire et al, 1998, Nature 391, 806). Any nucleic acid compound or formulation that results in formation of an siRNA molecule may be used in accordance with the embodiments described herein. See, e.g., Section 5'below.
  • small molecule and “small molecular weight compound,” and analogous terms include, but are not limited to, peptides, peptidomimetics, amino acids, amino acid analogs, polynucleotides, polynucleotide analogs, nucleotides, nucleotide analogs, other organic and inorganic compounds (i.e., includi g heteroorganic and organometallic compounds) having a molecular weight less than about ⁇ ' ⁇ , ⁇ grams per mole, organic or inorganic compounds having a molecular weight less than about 5,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 1 ,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 500 grams per mole, organic or inorganic compounds having a molecular weight less than about 100 grams per mole, as well as solvates, hydrates, prodrugs, stereoisomers and pharmaceutically acceptable salts
  • the small molecule is an organic compound other than a peptide, peptidomimetic, amino acid, amino acid analog, polynucleotide, polynucleotide analog, nucleic acid, nucleotide or nucleotide analog.
  • stereoisomer or “stereomerically pure compound” means one stereoisomer of a compound, in the context of an organic or inorganic molecule, that is substantially free of other stereoisomers of that compound.
  • a stereomerically pure compound having one chiral center will be substantially free of the opposite enantiomer of the compound.
  • a stereomerically pure compound having two chiral centers will be substantially free of other diastereomers of the compound.
  • a typical stereomerically pure compound is characterized by an enantiomeric excess greater than about 60% of one stereoisomer of the compound over one or more other stereoisomers of the compound, greater than about 80% of one stereoisomer of the compound over one or more other stereoisomers of the compound, greater than about 90% of the compound over one ore more other stereoisomers of the compound, greater than about 94% of one stereoisomer of the compound over one or more other stereoisomers of the compound, or greater than about 97% of one stereoisomer of the compound over one or more other stereoisomers of the compound or greater than about 99% of one stereoisomer of the compound over one or more other stereoisomers of the compound.
  • the compounds can have chiral centers and can occur as racemates, individual enantiomers or diastereomers, and mixtures thereof. All such isomeric forms are included within the embodiments disclosed herein, including mixtures thereof.
  • Various compounds contain one or more chiral centers, and can exist as racemic mixtures of enantiomers, mixtures of diastereomers or enantiomerically or optically pure compounds.
  • the use of stereomencally pure forms of such compounds, as well as the use of mixtures of those forms are encompassed by the embodiments disclosed herein.
  • mixtures comprising equal or unequal amounts of the enantiomers of a particular compound may be used in methods and compositions disclosed herein.
  • These isomers may be asymmetrically synthesized or resolved using standard techniques such as chiral columns or chiral resolving agents. See, e.g., Jacques, J., et al, Enantiomers, Racemates and
  • compounds in the context of organic and inorganic molecules, can include E and Z isomers, or a mixture thereof, and cis and trans isomers or a mixture thereof.
  • compounds are isolated as either the E or Z isomer. In other embodiments, compounds are a mixture of the E and Z isomers.
  • the terms “subject” or “patient” are used interchangeably.
  • the term “subject” refers to an animal (e.g., bird, reptile, mammal), preferably a mammal including a non-primate (e.g., camel, donkey, zebra, cow, pig, horse, goat, sheep, cat, dog, rat, mouse) and a primate (e.g., a monkey, chimpanzee, human), and most preferably a human.
  • a non-primate e.g., camel, donkey, zebra, cow, pig, horse, goat, sheep, cat, dog, rat, mouse
  • a primate e.g., a monkey, chimpanzee, human
  • premature human infant refers to a human infant born at less than 37 weeks of gestational age.
  • human infant refers to a newborn to 1 year old year human.
  • human child refers to a human that is 1 year to 18 years old.
  • human adult refers to a human that is 18 years or older.
  • elderly human refers to a human 65 years or older.
  • a synergistic effect of a combination of treatments permits the use of lower dosages of one or more treatments and/or less frequent administration of said treatments to a subject with a viral infection or a disease or symptom associated therewith.
  • the ability to utilize lower dosages of treatments (e.g., the compounds described herein, or prophylactic or therapeutic agents) and/or to administer said treatments less frequently reduces the toxicity associated with the administration of said treatments to a subject without reducing the efficacy of said treatments in the prevention or treatment of a viral infection or a disease or symptom associated therewith.
  • a synergistic effect results in improved efficacy of treatments (e.g., prophylactic or therapeutic agents) in the prevention, management and/or treatment of a viral infection or a disease or symptom associated therewith.
  • a synergistic effect of a combination of treatments avoids or reduces adverse or unwanted side effects associated with the use of any single treatment.
  • targeted or “targeted to” means having a nucleobase sequence that will allow its hybridization to a target nucleic acid (e.g., human host cell factor required for influenza virus replication) to induce a desired effect.
  • a desired effect is a reduction in the amount of a target nucleic acid.
  • a desired effect is reduction of the expression (protein or mRNA) and/or activity of the target human host cell factor.
  • a desired effect is reduction of influenza virus replication.
  • targeting means the process of design and selection of a nucleic acid compound that will specifically hybridize to a target nucleic acid and induce a desired effect.
  • target human host cell factor means a nucleic acid capable of being targeted by such a nucleic acid compound.
  • target region means a portion of a target to which one or more nucleic acid compounds is targeted.
  • target segment refers to a smaller portion or sub-portion of a region within a target.
  • a target segment can be the sequence of nucleotides of a target nucleic acid to which a nucleic acid compound is targeted.
  • the terms “therapies” and “therapy” can refer to any protocol(s), method(s), compound(s), composition(s), formulation(s), inhibitor(s), and/or agent(s) that can be used in the prevention, treatment, management, or amelioration of a viral infection or a symptom associated therewith.
  • the terms “therapies” and “therapy” refer to biological therapy, supportive therapy, and/or other therapies useful in treatment, management, prevention, or amelioration of a viral infection or a symptom or disease associated therewith known to one of skill in the art.
  • a “therapeutically effective amount” refers to the amount of a treatment or therapy that is sufficient to treat, prevent, and/or manage a viral infection or a disease or symptom associated therewith.
  • a “therapeutically effective amount” is the amount of a compound that reduces the severity, the duration and/or the symptoms associated with a viral infection or a disease or symptom associated therewith in a subject.
  • a "therapeutically effective amount” is the amount of a compound that results in a reduction in viral titer by at least 1.5 logs, at least 2 logs, at least 3 logs, at least 4 logs, or at least 5 logs in a subject administered a compound relative to the viral titer in a subject or group of subjects (e.g., two, three, five, ten or more subjects) not administered a compound.
  • a "therapeutically effective amount” is the amount of a compound that results in a reduction in viral titer by 1.5 to 10 logs, 1.5 to 5 logs, 2 to 10 logs, 2 to 5 logs, or 2 to 4 logs in a subject administered a compound relative to the viral titer in a subject or group of subjects (e.g., two, three, five, ten or more subjects) not administered a compound.
  • a therapeutic agent refers to any agent(s) (e.g., a compound) that can be used in the prevention, treatment and/or management of a viral infection or a symptom or disease associated therewith.
  • a therapeutic agent is an agent that is known to be useful for, or has been or is currently being used for the prevention, treatment, and/or management of a viral infection or a symptom or disease associated therewith.
  • the terms “treat,” “treatment,” and “treating” refer, in the context of administration of a therapy to a subject to treat a viral infection, to a beneficial or therapeutic effect of a therapy or a combination of therapies.
  • the terms “treat,” “treatment,” and “treating” refer to administering a compound or composition described herein to effect an alteration or improvement of a disease, condition, or symptom associated therewith.
  • such terms refer to one, two, three, four, five or more of the following effects resulting from the administration of a therapy or treatment or a combination thereof: (i) the reduction or amelioration of the severity of a viral infection and/or a symptom or disease associated therewith; (ii) the reduction in the duration of a viral infection and/or a symptom or disease associated therewith; (iii) the regression of a viral infection and/or a symptom or disease associated therewith; (iv) the reduction of the titer of a virus; (v) the reduction in organ failure associated with a viral infection or a disease associated therewith; (vi) the reduction in hospitalization of a subject; (vii) the reduction in hospitalization length; (viii) the increase in the survival of a subject; (ix) the elimination of a virus infection or a symptom or disease associated therewith; (x) the reduction or inhibition of the progression of a viral infection and/or a symptom or disease associated therewith; (xi) the reduction or prevention
  • FIG. 1 A Genome-wide RNAi Screen for Influenza Virus Host Cellular Factors, (a) A schematic of the recombinant WSN-Ren virus showing the HA segment modified to express Renilla luciferase but maintaining the HA packaging sequences, (b) An arrayed genome- wide RNAi library (100,000 siRNAs targeting over 19,000 human genes) was transfected into A549 cells. Cells were subsequently infected with WSN-Ren and virus replication was monitored by measuring luciferase activities at the indicated times.
  • Fig. 2. Identification of Host Factors Involved in Influenza Virus Entry (a) Illustration of the screen progression from primary genome-wide analysis to the identification of factors involved in entry and post-entry steps in the virus life cycle. The number of confirmed genes and number of genes tested at each stage (in parentheses) are indicated, (b) The relative effects of gene depletion (2 siRNAs/gene) on infection of luciferase-encoding HIV particles pseudotyped with WSN, VSV or MMLV envelopes (right panel).
  • A/WSN/33 virus infection is shown (right panel). Significant effects (p ⁇ 0.01 based on Welch T-test) are seen at 180min with all genes and with CSEIL, PRSS35, F13A1 (p ⁇ 0.02) at 90min. Levels of virus replication (WT WSN), viral gene (NP/Ml) transcription and entry of WSN pseudotyped particles or Bla-Ml VLPs in cells lacking these factors are shown in the left panel. Values relative to negative controls (bottom row) are depicted in a continuum, (b) Confocal imaging of influenza virus NP protein localization at the indicated times following A/WSN/33 virus infection in cells depleted of CSEIL, CAMK2B and KPNBl .
  • Fig. 4 Infectivity - toxicity relationship curve.
  • siRNA siRNA known to inhibit influenza virus Renilla luciferase reporter activity
  • siRNA targeting Renilla- light gray dots an siRNA known to inhibit influenza virus Renilla luciferase reporter activity
  • a negative control siRNA black dots
  • the toxicity score for each of these are 1 and 0.4, respectively.
  • a decision boundary was established (light gray curve; see Methods in Section 6 infra); if an siRNA fell below the boundary, it was considered to be toxic. Otherwise, it was considered a true hit (p ⁇ 0.05).
  • Three siRNAs were tested in >4 replicates. Toxic control (dark gray dots) fell below the decision boundary. Positive control (light gray dots) fell above the decision boundary.
  • Fig. 5 Functional classification of influenza A virus-host cellular proteins. 177 of the 295 identified host proteins were classified into related functional groups revealing 1 1 highly overrepresented biological processes required for influenza virus replication. Host cellular genes are represented on the y-axis, and their inclusion in a primary functional category or secondary function category is indicated along the x-axis. Boundaries of gene clusters and biological processes are represented by gray lines. Enrichment scores for each functional class are also given. Functional classification and enrichment analysis was conducted using the Database for Annotation, Visualization and Integrated Discovery (DAVID) Bioinformatics Resource (Huang et al, 2009).
  • DAVID Annotation, Visualization and Integrated Discovery
  • Fig. 6 Small molecule inhibitors targeting identified host factors reduce influenza virus growth.
  • MDCK-HA cells were infected with WSN-Ren virus at an MOI of 0.03 in the presence of increasing concentrations of various inhibitors targeting specific host genes that were confirmed as host cellular factors for influenza virus entry.
  • DMSO control was set to a 100%.
  • Virus growth was assayed at 36 h post-infection and mean inhibition +/- standard deviation of triplicate samples are shown as grey bars. The concentrations for 50% inhibition (IC 50 ) and the respective target genes are indicated. Cell viability (toxicity) with increasing concentrations of each respective inhibitor was assessed in parallel experiments (black lines).
  • Small molecules targeting host factors are as follows: Sirolimus (Rapamycin) and FRAP1 (mTOR; GenelD 2475) (Terada et al, 1 92; Price et al., 1992; and Chung et al., 1992) ; HSP90 Inhibitor CCTOl 8159 (4-(4-(2,3-Dihydro-1,4-benzodioxin-6-yl)-5-methyl-1H- pyrazol-3-yl)-6-ethylresorcinol) and HSP90AA1 (GenelD 3320) (Hardcastle et al., 2005; Sharp et al., 2007; and Smith et al., 2006); Podophyllotoxin (Podophyllinic Acid Lactone) and TUBB (tubulin beta; Gene ID 203068 (Desbene at al., 2002); FGF/VEGF Receptor Inhibitor (4-Hydroxy-3-benzimidazol-2-yl
  • Fig. 7 The vATPase subunit, ATP6V0C, is a host gene involved in influenza virus entry,
  • Influenza virus VLPs carrying a Bla-Ml fusion protein were used to infect A549 cells pretransfected with the cognate siRNAs. The percentage of cells containing detectable cytoplasmic beta-lactamase activity is indicated. In cells transfected with a scrambled control siRNA, approximately 74% were infected by the VLPs as measured by cytoplasmic beta-lactamase activity (second panel). However, depletion of ATP6V0C resulted in reduced VLP entry (10.5%).
  • ATP6V0C-depleted cells was monitored by tracking the localization of influenza virus NP protein.
  • Cells were stained for NP and nuclei and analyzed by confocal microscopy. At 90 minutes post infection an inhibition of incoming RNP accumulation in the nucleus was observed and a delay in the appearance of newly synthesized NP both in the nucleus
  • MOI vATPases
  • the high-content imaging-based analysis was performed using the Opera (Perkin-Elmer, Waltham, MA), a fully automated microscope system. 384- well plates containing A549 cells were transfected with siRNAs targeting the indicating genes. 48h post infection cells were infected with A/WSN/33 virus and fixed at the indicated time points.
  • Fig. 11 CAMK2B inhibition in A549 cells impairs influenza virus growth.
  • A549 cells were infected with influenza A/WSN/33 virus in the presence of DMSO or 20 ⁇ KN-93. Viral growth was determined by plaque assay at 24h post infection. The mean viral titer +/- standard deviation of triplicate samples is shown. These data are consistent with the inhibition of influenza virus replication by KN-93 in MDCK cells (Figs. 3d and 3e).
  • Fig. 12 Additional effects of influenza virus host factors on VSV replication.
  • siRNA-transfected A549 cells were infected with VSV at a multiplicity of infection (MO I) of 0.01 at 48h post siRNA transfection.
  • MO I multiplicity of infection
  • At 36h post infection supernatants were harvested and virus titers were determined by plaque assay on Vero cells. The mean viral titer +/- standard deviation of triplicate samples is shown.
  • influenza virus replication can be modulated by pharmacologically targeting human host cell factors required for viral replication.
  • Targeting host cell factors rather than the viral factors required for influenza virus replication, may greatly reduce the emergence of viral resistance and expands the number of targets for antiviral intervention.
  • the embodiments provided herein are based in part on the discovery that compounds (including, e.g., nucleic acid compounds, such as siRNAs, and small molecules) that reduce or inhibit the expression or activity of specific classes of human host cell proteins reduce influenza virus replication and thus are useful as antiviral agents.
  • nucleic acid compounds e.g., siRNAs
  • small molecules that target human host cell factors involved in influenza virus replication
  • compositions including pharmaceutical compositions, comprising such compounds, and methods of using such compounds and compositions for modulating influenza virus replication
  • compositions comprising such compounds, and methods of using such compounds and compositions for reducing or inhibiting influenza virus replication, or for treating or preventing influenza virus infection, or a symptom associated therewith, in a subject in need thereof.
  • nucleic acid compounds e.g., siRNAs
  • small molecules that target human host cell factors involved in influenza virus replication.
  • the compound modulates influenza virus replication by altering the expression (e.g., mRNA or protein) and/or activity of the human host cell factor involved in influenza virus replication.
  • the compound reduces or inhibits influenza virus replication by reducing or inhibiting the expression (e.g., mRNA or protein) and/or activity of the human host cell factor involved in influenza virus replication.
  • the human host cell factor is required for influenza virus replication.
  • the human host cell factor interacts with a component of the influenza virus.
  • the interaction of the host cell factor with the component of the influenza virus is direct.
  • the host cell factor is not involved in the non-specific induction of an antiviral state, e.g., the cellular interferon system, recognition of double-stranded RNA, etc.
  • the human host cell factor does not interact with a component of the influenza virus.
  • the host cell factor is required for influenza virus replication in human cells but not in insect cells.
  • the compounds provided herein target human host cell factors involved in influenza virus replication and modulate influenza virus replication.
  • the compounds provided herein target human host cell factors involved in influenza virus replication and reduce or inhibit influenza virus replication.
  • the targeted human host cell factor may be required for influenza virus replication.
  • the targeted human host cell factor may be involved in or required for one or more of the following events of the influenza virus life cycle: entry; uncoating; nuclear import; viral RNA transcription; or viral RNA translation.
  • the human host cell factor is not involved in influenza virus entry.
  • the human host cell factor is not involved in the nuclear import stage.
  • the human host cell factor is not involved in influenza virus assembly, budding, or release from host cells.
  • the human host cell factor is required for replication of viruses whose entry into cells is low-pH-dependent.
  • the human host cell factor may be required for entry of such viruses into cells.
  • RNA replication and transcription may be measured by measuring the replication and transcription of reporter gene product from an influenza virus mini-genome reporter construct, using, e.g., the assays disclosed herein. Such assays permit the identification of inhibitors of the viral polymerase or inhibitors of cellular proteins that are involved in viral RNA replication, translation or RNA trafficking.
  • the compound does not have an inhibitory effect on the overall host cell replication machinery, or has only a slight inhibitory effect compared to the effect on viral replication, as monitored by assays such as, e.g., the expression of a Renilla luciferase reporter from a control plasmid (e.g., Section 6 below).
  • the inhibitors alter the kinetics of the viral cycle, e.g., the rate of viral replication or particle production is decreased.
  • the kinetic effect of a compound is measured by adding the compound to a cell at different times (e.g., before, concurrently with, or after) infection with a virus.
  • the targeted human host cell factor may be involved in or required for replication of more than one strain or sub-type of influenza.
  • the human host cell factor may be involved in or required for replication of an influenza A virus, an influenza B virus, and/or an influenza C virus.
  • the human host cell factor is involved in or required for replication of a human-origin, an avian-origin (e.g., H5N1), and/or a swine- origin (e.g., H1N1) influenza virus.
  • the human host cell factor is also required for replication of one or more other viruses, e.g., but not limited to, vesicular stomatitis virus (VSV).
  • VSV vesicular stomatitis virus
  • the human host cell factor is not required for replication (including, e.g., entry) of viruses whose entry into cells is pH-independent, such as, e.g., murine leukemia virus (MMLV).
  • MMLV murine leukemia virus
  • the human host cell factor is not required for replication (e.g., entry, genome replication, etc.) of one or more of human immunodeficiency virus (HIV), Dengue virus, hepatitis C virus (HCV), West Nile virus (WNV), or VSV.
  • the human host cell factor is uniquely required for influenza virus replication.
  • a compound provided herein targets a component or regulator of, or factor that interacts with, one or more of the following categories of human host cell factors: cytoskeleton; ribonucleoprotein; spliceosome; ubiquitin/proteasome system; ribosome or other translation machinery; kinase; phosphatase; signaling (e.g., G-protein coupled receptors; signaling at the plasma membrane); mitochondrion or mitochondrial ribosome; plasminogen; stress response; v-ATPase; ion channel or other ion transport;
  • human host cell factors cytoskeleton; ribonucleoprotein; spliceosome; ubiquitin/proteasome system; ribosome or other translation machinery; kinase; phosphatase; signaling (e.g., G-protein coupled receptors; signaling at the plasma membrane); mitochondrion or mitochondrial ribosome; plasminogen; stress response; v-ATPa
  • a compound provided herein, and for use in the compositions and methods provided herein targets a component or regulator of, or a factor that interacts with, one or more of the following categories of human host cell factors: IP3-PKC pathway; COPI vesicles;
  • endosomal uptake, maturation, acidification, and fusion actin organization and function
  • PI3 -AKT pathway endosomal recycling pathway
  • MAPK pathway proteases
  • the compound reduces or inhibits the expression and/or activity of a human host cell factor in one of the aforementioned categories.
  • the compound modulates the expression and/or activity of one or more of the human host cell factors listed in Table 3. In specific embodiments, the compound reduces or inhibits the expression and/or activity of one or more of the human host cell factors listed in Table 3.
  • the compound modulates the expression and/or activity of one or more of the human host cell factors listed in Table 7. In specific embodiments, the compound reduces or inhibits the expression and/or activity of one or more of the human host cell factors listed in Table 7. [00118] In some embodiments, the compound modulates the expression and/or activity of one or more of the human host cell factors listed in Table 9. In specific embodiments, the compound reduces or inhibits the expression and/or activity of one or more of the human host cell factors listed in Table 9.
  • the compound modulates the expression (e.g., mRNA or protein) and/or activity of one or more of the following human host cell factors: ACRC; AKAP13; AKT1 ; ANAPC2; ANPEP; ARCN1 ; BRWD3; CAD; CAMK2B; CANT1 ;
  • CBLL1 CD81 ; CHAF1A; CLK1; CLOCK; COPA; COPB1 ; COPB2; COPG; CSEIL;
  • CTSW CTSW; DTX2; DUSP3; EPHB2; EPS8L3; F13A1 ; F AMI 35 A; FGFR2; FGFR4; FPR1, FRAP1 (mTOR); GABBR1 ; GRK6; GSK3B; HAND2; HIST3H3; HSP90AA1 ; IL1F9;
  • PRSS35 PSMD1 ; RAB11B; RBM5; RP1 1-45B20.2; RPS10; RPS20; SF3A1 ; SNRPA1 ; STK31 ; STK39; STX10; SUM02; SUM04; TBK1 ; TEAD3; TNP03; TRPV2; TUBB;
  • the compound reduces or inhibits the expression and/or activity of one or more of the aforementioned human host cell factors.
  • the compound modulates the expression (e.g., mRNA or protein) and/or activity of one or more of the following human host cell factors: AKAP13; ARCN; BRWD3; CD81 ; COPG; CTSW; DUSP3; EPHB2; FAM135A; FGFR2; FGFR4; GABBR1 ; GSK3B; ITGA3; JAK2; MAP2K3; NEK6; RAB1 IB; or one or more of the v- ATPase subunits, ATP6V0B, ATP6V0C, ATP6V1A, ATP6V1B2, or ATP6AP1.
  • human host cell factors e.g., mRNA or protein
  • the compound modulates the expression (e.g., mRNA or protein) and/or activity of one or more of the following human host cell factors: CAMK2B; CSEIL; F13A1; KPNB1 ; MAP3K12; PP1R14D; PRSS35; RPS10; SF3A1 ; or SUM04.
  • CAMK2B CAMK2B
  • CSEIL e.g., CSEIL
  • F13A1 e.g., mRNA or protein
  • the compound modulates the expression (e.g., mRNA or protein) and/or activity of one or more of the following human host cell factors: ACRC; DTX2; EPS8L3; FPR1 ; MAP3K1 1 ; NUP214; PRPH2; RP1 1-45B20.2; STX10; SUM02; TRPV2; or TUBB.
  • the compound modulates the expression (e.g., mRNA or protein) and/or activity of one or more of the following human host cell factors: ANPEP; CAM2KB; FGFR4; FRAP1 (mTOR); GSK3B/CSNK1G2; HSP90AA1 ; or TUBB.
  • the compound reduces or inhibits the expression and/or activity of one or more of the aforementioned human host cell factors.
  • the compound may be any compound described herein, known in the art, or yet to be discovered that targets one or more of the aforementioned categories of human host cell factors, a specific factor(s) in such a category, and/or one of the aforementioned human host cell factors. In certain embodiments, the compound is not toxic to the human host cell.
  • the compound does not target AKT1 , ARCN1 , COPG, GRK6, HAND2, HIST3H3, an HSP90 (e.g., HSP90AA1), NUP153, RBM5, RPS10, RPS20, or a v-ATPase subunit.
  • the compound does not reduce or inhibit the expression and/or activity of AKT1 , ARCN1, COPG, GRK6, HAND2, HIST3H3, an HSP90 (e.g., HSP90AA1), NUP153, RBM5, RPS10, RPS20, or a v-ATPase subunit.
  • the compound does not target AKAP13, CD81, CAMK2B, CSE1L, DUSP3, FGFR2, FGFR4, GSK3B, ITGA3, KPNBl, MAP2K3, or RABl IB. In certain embodiments, the compound does not reduce or inhibit the expression and/or activity of AKAP13, CD81, CAMK2B, CSE1L, DUSP3, FGFR2, FGFR4, GSK3B, ITGA3, KPNBl, MAP2K3, or RABl IB.
  • the compound is an agent that reduces or inhibits the expression (e.g., mRNA or protein) and/or activity of a human host cell factor involved in influenza virus replication. In some embodiments, the compound is an agent that reduces or inhibits the expression (e.g., mRNA or protein) and/or activity of a human host cell factor required for influenza virus replication. In some embodiments, the compound reduces or inhibits the interaction of a human host cell factor with a component of the influenza virus. In some embodiments, the compound does not trigger a non-influenza-specific antiviral state. For example, in some embodiments, an siRNA compound does not induce a non-specific antiviral state, for example, it does not induce an interferon response. In some embodiments, the compound reduces or inhibits the interaction of a human host cell factor with a
  • the compound reduces or inhibits a direct interaction of a human host cell factor with a component of the influenza virus. In some embodiments, the compound reduces or inhibits influenza virus replication in human cells but not in insect cells. In some embodiments, the compound reduces or inhibits one or more of the following events of the influenza viral life cycle: entry; uncoating; nuclear import; viral RNA transcription; or viral RNA translation. In some embodiments, the compound does not reduce influenza virus entry. In some embodiments, the compound does not reduce the nuclear import stage. In some embodiments, the compound does not reduce or inhibit influenza virus assembly, budding, or release from host cells. In some embodiments, the compound reduces or inhibits replication of viruses whose entry into cells is low-pH- dependent. For example, the compound may reduce or inhibit entry of such viruses into cells.
  • the compound reduces or inhibits replication of more than one strain or sub-type of influenza.
  • the compound may reduce or inhibit replication of influenza virus A, an influenza B virus, and/or an influenza C virus.
  • the compound reduces or inhibits replication of a human-origin, an avian- origin (e.g., H5N1), and/or a swine-origin (e.g., H1N1) influenza virus.
  • a human-origin an avian- origin
  • a swine-origin e.g., H1N1 influenza virus.
  • the compound reduces or inhibits replication of another virus in addition to influenza virus such as, e.g., vesicular stomatitis virus (VSV).
  • influenza virus such as, e.g., vesicular stomatitis virus (VSV).
  • the compound does not reduce or inhibit replication (including, e.g., entry) of viruses whose entry into cells is pH-independent, such as, e.g., MMLV.
  • the compound does not reduce or inhibit replication (e.g., entry, genome replication, etc.) of one or more of HIV, Dengue virus, HCV, WNV, or VSV.
  • the compound reduces or inhibits influenza virus replication and not the replication of other viruses.
  • the compounds provided herein include compounds of any structure described herein or incorporated by reference herein, and solvates, hydrates, prodrugs, stereoisomers and pharmaceutically acceptable salts thereof.
  • Such compounds include, but are not limited to, nucleic acid molecules including, but not limited to, double-stranded or single-stranded DNA, or double-stranded or single-stranded RNA, antisense RNA, RNA interference (RNAi) molecules (e.g., small interfering RNA (siRNA), micro-RNA (miRNA), short hairpin RNA (shRNA), etc.), intron sequences, triple helix nucleic acid molecules and aptamers;
  • RNAi RNA interference
  • a compound is purified. In one embodiment, a compound is isolated.
  • the compound is a nucleic acid compound.
  • the nucleic acid compound may be any nucleic acid compound known in the art or described herein that is able to modulate the expression and/or activity of a human host cell factor described herein may.
  • the nucleic acid compound is an antisense compound.
  • the nucleic acid compound is an siRNA.
  • the nucleic acid compound has a sequence optimized for use as an siRNA, according to methods known in the art.
  • the nucleic acid compound is a modified oligonucleotide.
  • the nucleic acid compound comprises an oligonucleotide of 12 to 30 linked nucleosides, for example, 12 to 15, 15 to 20, 20 to 25, or 25 to 30 linked nucleosides, which may be targeted to a nucleic acid encoding a human host cell factor involved in influenza virus replication.
  • the antisense or siRNA compound reduces or inhibits the expression and/or activity of an aforementioned human host cell factor, or a factor in one of the aforementioned categories.
  • the compound targets a nucleotide sequence selected from Table 1 (see also Table 9). In certain embodiments, e.g., when targeting of a nucleotide sequence selected from Table 1 (see also Table 9). In certain embodiments, e.g., when targeting of a nucleotide sequence selected from Table 1 (see also Table 9). In certain embodiments, e.g., when targeting of a nucleotide sequence selected from Table 1 (see also Table 9). In certain embodiments, e.g., when targeting of a nucleotide sequence selected from Table 1 (see also Table 9). In certain embodiments, e.g., when targeting of a nucleotide sequence selected from Table 1 (see also Table 9). In certain embodiments, e.g., when targeting of a nucleotide sequence selected from Table 1 (see also Table 9). In certain embodiments, e.g., when targeting of a nucleotide sequence selected from Table 1 (see also Table 9). In certain embodiments, e.g
  • nucleobases represented by a “U” (uracil) in a sequence in Table 1 may be replaced with thymine nucleobases (represented by a "T”).
  • nucleobases represented by a "T” (thymine) in a sequence in Table 1 may be replaced with uracil nucleobases (represented by a "U”).
  • nucleotide sequence "AAGTAGGGATAAATTACTCTA" in Table 1 may be replaced with the nucleotide sequence "AAGUAGGGAUAAAUUACUCUA.”
  • the nucleic acid compound targeting a sequence in Table 1 is an antisense compound.
  • the nucleic acid compound targeting a sequence in Table 1 is an siRNA.
  • the siRNA that targets one. of the aforementioned human host cell factors or sequences is obtained from a commercially available source.
  • the siRNA can be from Qiagen (Draggable Set version 1 or 2), NM Set version 1 , XM Set version 1 , the kinome library from Invitrogen or the kinome library from IDT.
  • an siRNA duplex is created from a 21mer sequence in Table 1 as exemplified in the following example:
  • the first two are the antisense overhang, the sense overhang is always TT.
  • siRNA duplexes based on the sequences in Table 1 that contain Us are created the same way, except that the sequence is already and RNA; i.e., the sequence in Table 1 containing Us correspond to host cell mRNA targets.
  • the siRNA compound comprises the sequence
  • This sequence is the sense sequence for a 25mer siRNA duplex for use in accordance with the embodiments described herein.
  • Antisense compounds for use in the embodiments described herein include, but are not limited to, oligomeric compounds, oligonucleotides, oligonucleosides, oligonucleotide analogs, oligonucleotide mimetics, and antisense oligonucleotides.
  • Antisense compounds may target a nucleic acid, meaning that the antisense compound is capable of undergoing hybridization to a target nucleic acid through hydrogen bonding.
  • an antisense compound has a nucleobase sequence that, when written in the 5' to 3' direction, comprises the reverse complement of the target segment of a target nucleic acid to which it is targeted.
  • an antisense oligonucleotide has a nucleobase sequence that, when written in the 5' to 3' direction, comprises the reverse complement of the target segment of a target nucleic acid to which it is targeted.
  • an antisense compound targeted to a nucleic acid is 12 to 30 subunits in length.
  • antisense compounds are from 12 to 30 linked subunits.
  • the antisense compound is 8 to 80, 12 to 50, 15 to 30, 18 to 24, 19 to 22, or 20 linked subunits.
  • the antisense compounds are 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, or 80 linked subunits in length, or a range defined by any two of the above values.
  • the linked subunits are linked nucleobases, nucleosides, or nucleotides.
  • the antisense compound is an antisense oligonucleotide, and the linked subunits are nucleotides. Antisense compounds may also be shortened or lengthened, or have mismatches introduced, without eliminating their activity.
  • Antisense Compound Motifs [00137]
  • antisense compounds targeted to a nucleic acid have chemically modified subunits arranged in patterns, or motifs, to confer to the antisense compounds properties such as enhanced inhibitory activity, increased binding affinity for a target nucleic acid, or resistance to degradation by in vivo nucleases.
  • Chimeric antisense compounds typically contain at least one region modified so as to confer increased resistance to nuclease degradation, increased cellular uptake, increased binding affinity for the target nucleic acid, or increased inhibitory activity.
  • a second region of a chimeric antisense compound may optionally serve as a substrate for the cellular endonuclease RNaseH, which cleaves the RNA strand of an RNA:DNA duplex.
  • Antisense compounds having a gapmer motif are considered chimeric antisense compounds.
  • the term "gapmer” means an antisense compound in which an internal position having a plurality of nucleotides that supports RNaseH cleavage is positioned between external regions having one or more nucleotides that are chemically distinct from the nucleosides of the internal region.
  • a "gap segment” means the plurality of nucleotides that make up the internal region of a gapmer.
  • the antisense compound as a "wingmer” motif, having a wing-gap or gap-wing configuration, i.e. an X-Y or Y-Z configuration as described above for the gapmer configuration.
  • wingmer configurations for use herein include, but are not limited to, for example 5-10, 8-4, 4-12, 12-4, 3-14, 16-2, 18-1, 10-3, 2-10, 1-10 or 8-2.
  • a "wing segment” means the external region of a gapmer.
  • an antisense compound targeted to a nucleic acid has a gap-widened motif.
  • gap-widened means an antisense compound has a gap segment of 12 or more contiguous 2'-deoxyribonucleotides positioned between and immediately adjacent to 5' and 3' wing segments having from one to six nucleotides having modified sugar moieties.
  • the antisense compound comprises one or more chemically modified nucleosides.
  • the chemical modification comprises a 2' -sugar modification.
  • the chemical modification comprises a 2'-MOE sugar modification.
  • a target region of a human host cell factor involved in influenza virus replication is a structurally defined region of the nucleic acid.
  • a target region may encompass a 3' UTR, a 5' UTR, an exon, an intron, a coding region, a translation initiation region, translation termination region, or other defined nucleic acid region.
  • a target region may encompass the sequence from a 5' target site of one target segment within the target region to a 3' target site of another target segment within the target region.
  • Targeting includes determination of at least one target segment to which an antisense compound hybridizes, such that a desired effect occurs.
  • the desired effect is a reduction in mRNA target nucleic acid levels.
  • the desired effect is reduction of levels of protein encoded by the target nucleic acid or a phenotypic change associated with the target nucleic acid.
  • the reduction is 70% or greater, 75% or greater, 80% or greater, 85% or greater, 90% or greater, 95% or greater, or 100% at a concentration of 100 nM in T-24 cells.
  • a target region may contain one or more target segments. Multiple target segments within a target region may be overlapping. Alternatively, they may be non- overlapping. In certain embodiments, target segments within a target region are separated by no more than about 300 nucleotides. In other embodiments, target segments within a target region are separated by no more than about, 250, 200, 150, 100, 90, 80, 70, 60, 50, 40, 30, 20, or 10 nucleotides on the target nucleic acid. In certain embodiments, target segments within a target region are separated by no more than about 5 nucleotides on the target nucleic acid. In certain embodiments, target segments are contiguous.
  • Suitable target segments may be found within a 5' UTR, a coding region, a 3' UTR, an intron, or an exon.
  • Target segments containing a start codon or a stop codon are also suitable target segments.
  • a suitable target segment may specifically exclude a certain structurally defined region such as the start codon or stop codon.
  • the determination of suitable target segments may include a comparison of the sequence of a target nucleic acid to other sequences throughout the genome.
  • the BLAST algorithm may be used to identify regions of similarity amongst different nucleic acids. This comparison can prevent the selection of antisense compound sequences that may hybridize in a non-specific manner to sequences other than a selected target nucleic acid (i.e., non- target or off-target sequences).
  • phenotypic changes are indicative of inhibition of gene expression.
  • phenotypic changes may include a reduction in influenza virus replication, infection, or a symptom or disease associated therewith, as described herein infra.
  • hybridization occurs between an antisense compound disclosed herein and a target nucleic acid.
  • the most common mechanism of hybridization involves hydrogen bonding (e.g., Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding) between complementary nucleobases of the nucleic acid molecules.
  • Hybridization can occur under varying conditions. Stringent conditions are sequence-dependent and are determined by the nature and composition of the nucleic acid molecules to be hybridized. Methods of determining whether a sequence is specifically hybridizable to a target nucleic acid are well known in the art.
  • the antisense compounds provided herein are specifically hybridizable with a target nucleic acid.
  • An antisense compound and a target nucleic acid are complementary to each other when a sufficient number of nucleobases of the antisense compound can hydrogen bond with the corresponding nucleobases of the target nucleic acid, such that a desired effect will occur (e.g., antisense inhibition of a target nucleic acid).
  • a desired effect e.g., antisense inhibition of a target nucleic acid.
  • Non-complementary nucleobases between an antisense compound and a target nucleic acid may be tolerated provided that the antisense compound remains able to specifically hybridize to a target nucleic acid.
  • an antisense compound may hybridize over one or more segments of a target nucleic acid such that intervening or adjacent segments are not involved in the hybridization event (e.g., a loop structure, mismatch or hairpin structure).
  • the antisense compounds provided herein are at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% complementary to a target nucleic acid.
  • Percent complementarity of an antisense compound with a target nucleic acid can be determined using routine methods, e.g., using BLAST programs (basic local alignment search tools) and PowerBLAST programs known in the art (Altschul et al., J. Mol. Biol., 1990, 215, 403 410; Zhang and Madden, Genome Res., 1997, 7, 649 656). Percent homology, sequence identity or complementarity, can be determined by, for example, the Gap program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics
  • the antisense compounds provided herein are fully complementary (i.e., 100% complementary) to a target nucleic acid.
  • antisense compound may be fully complementary to a target nucleic acid, or a target region, or a target segment or target sequence thereof.
  • "fully complementary" means each nucleobase of an antisense compound is capable of precise base pairing with the
  • nucleic acid corresponding nucleobases of a target nucleic acid.
  • non-complementary nucleobase may be at the 5' end or 3' end of the antisense compound.
  • the non-complementary nucleobase or nucleobases may be at an internal position of the antisense compound.
  • two or more non- complementary nucleobases may be contiguous (i.e. linked) or noncontiguous.
  • non-complementary nucleobase is located in the wing segment of a gapmer antisense oligonucleotide.
  • antisense compounds up to 20 nucleobases in length comprise no more than 4, no more than 3, no more than 2 or no more than 1 non-complementary nucleobase(s) relative to a target nucleic acid.
  • antisense compounds up to 30 nucleobases in length comprise no more than 6, no more than 5, no more than 4, no more than 3, no more than 2 or no more than 1 non-complementary nucleobase(s) relative to a target nucleic acid.
  • the antisense compounds provided herein also include those which are complementary to a portion of a target nucleic acid.
  • portion refers to a defined number of contiguous (i.e. linked) nucleobases within a region or segment of a target nucleic acid.
  • a “portion” can also refer to a defined number of contiguous nucleobases of an antisense compound.
  • the antisense compounds are complementary to at least an 8 nucleobase portion of a target segment.
  • the antisense compounds are complementary to at least a 12 nucleobase portion of a target segment.
  • the antisense compounds are complementary to at least a 15 nucleobase portion of a target segment. Also contemplated are antisense compounds that are
  • nucleobase portion of a target segment complementary to at least a 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more nucleobase portion of a target segment, or a range defined by any two of these values.
  • the antisense compounds provided herein include those comprising a portion which consists of at least 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 contiguous nucleobases of the nucleobase sequence set forth in Table 1 supra or elsewhere herein, or incorporated by reference herein.
  • the antisense compounds are complementary to an equal-length portion of the nucleobase sequence.
  • the antisense compounds are at least 75%, 80%, 85%, 90%, 95%, or 100% (fully) complementary to the nucleobase sequence.
  • the antisense compounds provided herein may also have a defined percent identity to a particular nucleotide sequence.
  • an antisense compound is identical to the sequence disclosed herein if it has the same nucleobase pairing ability.
  • a RNA which contains uracil in place of thymidine in a disclosed DNA sequence would be considered identical to the DNA sequence since both uracil and thymidine pair with adenine.
  • Shortened and lengthened versions of the antisense compounds described herein as well as compounds having non-identical bases relative to the antisense compounds provided herein also are contemplated.
  • the non-identical bases may be adjacent to each other or dispersed throughout the antisense compound. Percent identity of an antisense compound is calculated according to the number of bases that have identical base pairing relative to the sequence to which it is being compared.
  • the antisense compounds are at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to one or more of the antisense compounds or sequences thereof, or a portion thereof, disclosed herein.
  • a nucleoside is a base-sugar combination.
  • the nucleobase (also known as base) portion of the nucleoside is normally a heterocyclic base moiety.
  • Nucleotides are nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside. For those nucleosides that include a pentofuranosyl sugar, the phosphate group can be linked to the 2', 3' or 5' hydroxyl moiety of the sugar.
  • Oligonucleotides are formed through the covalent linkage of adjacent nucleosides to one another, to form a linear polymeric oligonucleotide. Within the oligonucleotide structure, the phosphate groups are commonly referred to as forming the internucleoside linkages of the oligonucleotide.
  • Modifications to antisense compounds encompass substitutions or changes to internucleoside linkages, sugar moieties, or nucleobases. Modified antisense compounds are often preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for nucleic acid target, increased stability in the presence of nucleases, or increased inhibitory activity. [00164] Chemically modified nucleosides may also be employed to increase the binding affinity of a shortened or truncated antisense oligonucleotide for its target nucleic acid.
  • the naturally occurring intemucleoside linkage of RNA and DNA is a 3' to 5' phosphodiester linkage.
  • Antisense compounds having one or more modified, i.e. non- naturally occurring, intemucleoside linkages are often selected over antisense compounds having naturally occurring intemucleoside linkages because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for target nucleic acids, and increased stability in the presence of nucleases.
  • Oligonucleotides having modified intemucleoside linkages include
  • intemucleoside linkages that retain a phosphorus atom as well as intemucleoside linkages that do not have a phosphorus atom.
  • Representative phosphorus containing intemucleoside linkages include, but are not limited to, phosphodiesters, phosphotriesters,
  • antisense compounds targeted to a nucleic acid comprise one or more modified intemucleoside linkages.
  • the modified intemucleoside linkages are phosphorothioate linkages.
  • each intemucleoside linkage of an antisense compound is a phosphorothioate intemucleoside linkage.
  • Antisense compounds for use herein can optionally contain one or more nucleotides having modified sugar moieties.
  • Sugar modifications may impart nuclease stability, binding affinity or some other beneficial biological property to the antisense compounds.
  • the furanosyl sugar ring of a nucleoside can be modified in a number of ways including, but not limited to: addition of a substituent group, particularly at the 2' position; bridging of two non-geminal ring atoms to form a bicyclic nucleic acid (BNA); and substitution of an atom or group such as -S-, -N(R)- or -C(Ri)(R 2 ) for the ring oxygen at the 4'-position.
  • substituted sugars especially 2'- substituted sugars having a 2'-F, 2'-OCH 2 (2'-OMe) or a 2'-0(CH 2 ) 2 -OCH 3 (2'-0- methoxyethyl or 2'-MOE
  • Bicyclic modified sugars also include (6'S)-6'methyl BNA, Aminooxy (4'-CH2-0-N(R)-2') BNA, Oxyamino (4'-CH2-N(R)-0-2') BNA wherein, R is, independently, H, a protecting group, or CI -CI 2 alkyl.
  • Methods for the preparations of modified sugars are well known to those skilled in the art.
  • nucleobase moieties (natural, modified or a combination thereof) are maintained for hybridization with an appropriate nucleic acid target.
  • antisense compounds targeted to a nucleic acid comprise one or more nucleotides having modified sugar moieties.
  • the modified sugar moiety is 2'-MOE.
  • the 2'-MOE modified nucleotides are arranged in a gapmer motif.
  • nucleobases distinguishable from, yet functionally interchangeable with, naturally occurring or synthetic unmodified nucleobases.
  • Both natural and modified nucleobases are capable of participating in hydrogen bonding. Such nucleobase modifications may impart nuclease stability, binding affinity or some other beneficial biological property to antisense compounds.
  • Modified nucleobases include synthetic and natural nucleobases such as, for example, 5-methylcytosine (5-me-C). Certain nucleobase substitutions, including 5-methylcytosine substitutions, are particularly useful for increasing the binding affinity of an antisense compound for a target nucleic acid.
  • 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2°C (Sanghvi, Y.S., Crooke, S.T. and Lebleu, B., eds., Antisense Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278).
  • Additional unmodified nucleobases include 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl (-C ⁇ C-CH 3 ) uracil and cytosine and other alkynyl derivatives of pyrimidine bases, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8- substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other
  • 5- substituted uracils and cytosines 7-methylguanine and 7-methyladenine, 2-F-adenine, 2- amino-adenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3- deazaguanine and 3-deazaadenine.
  • Heterocyclic base moieties may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example 7-deaza-adenine, 7- deazaguanosine, 2-aminopyridine and 2-pyridone.
  • Nucleobases that are particularly useful for increasing the binding affinity of antisense compounds include 5-substituted pyrimidines,
  • antisense compounds targeted to a nucleic acid comprise one or more modified nucleobases.
  • gap- widened antisense oligonucleotides targeted to a nucleic acid comprise one or more modified nucleobases.
  • the modified nucleobase is 5-methylcytosine.
  • each cytosine is a 5-methylcytosine.
  • Antisense compounds may be covalently linked to one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the resulting antisense oligonucleotides.
  • Typical conjugate groups include cholesterol moieties and lipid moieties.
  • Additional conjugate groups include carbohydrates, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes.
  • Antisense compounds can also be modified to have one or more stabilizing groups that are generally attached to one or both termini of antisense compounds to enhance properties such as, for example, nuclease stability. Included in stabilizing groups are cap structures. These terminal modifications protect the antisense compound having terminal nucleic acid from exonuclease degradation, and can help in delivery or localization within a cell. The cap can be present at the 5'-terminus (5'-cap), or at the 3'-terminus (3'-cap), or can be present on both termini. Cap structures are well known in the art and include, for example, inverted deoxy abasic caps. Further 3' and 5'-stabilizing groups that can be used to cap one or both ends of an antisense compound to impart nuclease stability include those disclosed in WO 03/004602 published on January 16, 2003.
  • the nucleic acid compound for use in the embodiments described herein is an siR A compound.
  • siR A compound a compound that has emerged as one of the most efficient methods for inactivation of genes.
  • RNAi has emerged as one of the most efficient methods for inactivation of genes (Nature Reviews, 2002, v. 3, p. 737-47; Nature, 2002, v. 418, p. 244-51). As a method, it is based on the ability of dsRNA species to enter a specific protein complex, where it is then targeted to the complementary cellular RNA and specifically degrades it.
  • dsRNAs are digested into short (17-29 bp) interfering (also referred to as "inhibitor") RNAs (siRNAs) by type III RNAses (DICER, Drosha, etc) (Nature, 2001 , v. 409, p. 363-6; Nature, 2003, 425, p. 415-9). These fragments and complementary mRNA are recognized by the specific RISC protein complex. The whole process is culminated by endonuclease cleavage of target mRNA (Nature Reviews, 2002, v. 3, p. 737-47; Curr Opin Mol. Ther. 2003 June; 5(3):217-24). See also, e.g.
  • a compound provided herein is an siRNA compound.
  • siRNAs are double stranded nucleic acid molecules that when introduced into a cell, trigger RNA interference (RNAi).
  • RNAi RNA interference
  • Nonlimiting examples of targets for siRNA molecules which may be used in accordance with the embodiments described herein are provided in Table 1 infra.
  • the siRNA is long enough to induce RNAi but small enough to avoid inducing an immune response.
  • the siRNA compound may be generated, analyzed, and modified in accordance with the provisions of Section 5.1.1.
  • nucleic acids and nucleotide sequences that can be used for preparation of a double stranded nucleic acid molecule that reduces or inhibits expression of a human host cell factor described herein.
  • the double stranded nucleic acid is designed based on the nucleotide sequence of the target nucleic acid.
  • an appropriate siRNA can be designed and synthesized using techniques known in the art and described herein. See, e.g., Kazunori Taira, et al.: RNAi Jikken Protocol, Yodosha (2003); Elbashir S. M. et al.: Genes Dev.
  • RNA-directed RNA polymerase acts as a key catalyst.
  • a region downstream of an initiation codon maybe selected, in which the sequence AA(Ni 9-2 g)TT or AA(N 2 i -3 i) is searched for, and the GC content of this sequence is calculated.
  • a GC content of 50% is ideal; however, a sequence having a GC content of anywhere from at least 30% to 70% may be selected.
  • BLAST e.g. EST database of NCBI
  • a double stranded nucleic acid with the chosen sequence is introduced or expressed within the cell, and the amount of target mRNA is measured (e.g. Northern blot or RT-PCR methods) or the amount of target protein is measured (e.g. Western blot or fluorescent antibody method), or using an assay for the target's activity known to persons skilled in the art.
  • the double stranded nucleic acid comprises an antisense strand and a sense strand thereof.
  • the antisense strand comprises an antisense sequence of 18 to 29, preferably 19 to 25 nucleotides, which is completely complementary to a partial sequence of the oligonucleotide, and further, comprises 1 to 4 bases at the 3 '-end that protrude when annealed with the sense strand (overhang).
  • the sense strand ordinarily comprises a completely complementary sequence to the antisense strand and comprises 1 to 4 bases protruding at the 3' end (overhang).
  • one or more mutations or substitutions may be present in the sense strand.
  • the nucleic acid of the sense strand and the antisense strand may be RNA, DNA, or a mixture thereof.
  • the antisense strand sequence is RNA.
  • both the sense strand and the antisense strand are RNA.
  • the overhang portion may be formed with deoxyribonucleotides G, A, T, and C and/or ribonucleotides G, A, U, and C, but a deoxyribonucleotide T and a ribonucleotide U are preferable.
  • the number of overhang nucleotides is preferably 2 or 3, with 2 being preferable in some embodiments. Suitable examples include UU (RNA) and TT (DNA).
  • Methods for preparing the double stranded nucleic acid compounds for use as siRNA include, e.g., chemical synthesis, methods of in vitro synthesis, and methods of effecting expression within a cell using an expression vector (see, e.g. Takashi Morita, et al: Tanpakushitu Kakusan Kouso (Proteins, Nucleic Acids and Enzymes) Vol. 47 No. 14 p 1939-1945 (2002); Asako Sugimoto, Kagaku to Seibutsu
  • double stranded nucleic acid is prepared by annealing an artificially synthesized sense strand and antisense strand.
  • the resultant double stranded nucleic acid can be introduced into a cell using any suitable reagent known in the art, such as FuGENE6 (Roche) or Lipofectamine 2000 (Invitrogen).
  • FuGENE6 FuGENE6
  • Lipofectamine 2000 Invitrogen.
  • a double stranded siRNA is expressed by association with, e.g., a T7 promoter and T7 RNA
  • siRNA can be introduced into a cell by, e.g., lipofection methods using FuGENE6 (Roche).
  • Intracellular expression of siRNA can be effected using an siRNA expression vector.
  • a sense strand and an antisense strand may be simultaneously expressed from both ends by two kinds of promoters, from separate transcription units, or be expressing siRNA precursors which adopt a hairpin structure.
  • an expression vector for example, pSilencer siRNA Expression Vector (Ambion Inc.) can be used.
  • DNA-based vectors capable of generating siRNA within cells have also been developed and may be used in accordance with the embodiments described herein.
  • the method generally involves transcription of short hairpin RNAs that are efficiently processed to form siRNAs within cells.
  • siRNAs For methods on the delivery of siRNAs, see, for example, Shen et al (FEBS letters 539: 1 1 1-1 14 (2003)), Xia et al., Nature Biotechnology 20: 1006-1010 (2002), Reich et al., Molecular Vision 9: 210-216 (2003), Sorensen et al. (J. Mol. Biol. 327: 761-766 (2003), Lewis et al., Nature Genetics 32: 107-108 (2002) and Simeoni et al., Nucleic Acids Research 31, 1 1 : 2717-2724 (2003). siRNA has recently been successfully used for inhibition in primates; for further details see Tolentino et al., Retina 24(1) February 2004 pp 132-138.
  • the compound is a small molecule.
  • the small molecule is Betulinic acid (available from VWR Interaational/Enzo Life Sciences Intl.); CCT018159 (4-(4-(2,3-Dihydro- 1 ,4-benzodioxin-6-yl)-5-methyl- 1 H-pyrazol-3-yl)-6- ethylresorcinol; available from Calbiochem); Diphyllin (available from Sigma; see Fig. 13a); the FGF VEGF receptor inhibitor 4-Hydroxy-3-benzimidazol-2-ylhydroquinolin-2-one;
  • Hymenialdisine (available from Biomol International LP); KN-93 (available from
  • Podophyllotoxin Podophyllinic Acid Lactone
  • the compound is not CCT018159. In some embodiments, the compound is not Diphyllin.
  • any compound or library of compounds from any source can be tested for modulation, reduction or inhibition of influenza virus replication, or for use as antiviral agents, by targeting one or more of the classes of human host cell proteins or specific human host cell proteins described herein.
  • Such compounds include, but are not limited to, proteins, polypeptides, peptides, nucleic acids, including dominant negative mutants, ribozyme or triple helix molecules, antibodies
  • intrabodies include antibodies for intracellular use, referred to herein as intrabodies), small organic molecules, or inorganic molecules.
  • an antibody is used, for example, an intrabody.
  • Antibodies used include immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that specifically binds to one or more of the classes of human host cell proteins or specific human host cell proteins described herein.
  • Antibodies include, but are not limited to, monoclonal antibodies, multispecific antibodies, human antibodies, humanized antibodies, synthetic antibodies, chimeric antibodies, polyclonal antibodies, single domain antibodies, camelized antibodies, single-chain Fvs (scFv), single chain antibodies, Fab fragments, F(ab') fragments, disulfide-linked bispecific Fvs (sdFv), intrabodies, and anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id and anti-anti-Id antibodies to antibodies), and epitope-binding fragments of any of the above.
  • antibodies include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules.
  • Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG], IgG 2 , IgG 3 , IgG 4 , IgAi and IgA 2 ) or subclass.
  • the antibodies used are commercially or publicly available.
  • the antibodies described in this section can produced by any method well known in the art, e.g., as described in U.S. Patent Nos. 5,807,715, 6,331,415, and 6,818,216; U.S. Patent Application Publication Nos.
  • small molecular weight compounds are used.
  • the compound is in a form so that it can be delivered into a human host cell, preferably, in vivo.
  • the compounds are known inhibitors of the host cell proteins described herein. In some embodiments, the compounds are identified by screening for their ability to inhibit the classes of host cell proteins described herein, and are then tested for their ability to inhibit or reduce influenza virus replication.
  • nucleic acid compounds 5.2.1.1
  • the methods of treating cells with antisense compounds described herein may be modified appropriately for treatment with other nucleic acid compounds, such as siRNAs. With respect to siRNAs, see also Section 5.1.1.2 above and the references cited therein.
  • cell types used for such analyses are available from commercial vendors (e.g. American Type Culture Collection, Manassus, VA; Zen-Bio, Inc., Research Triangle Park, NC; Clonetics Corporation, Walkersville, MD) and cells are cultured according to the vendor's instructions using commercially available reagents (e.g. Invitrogen Life Technologies, Carlsbad, CA).
  • Illustrative cell types include, but are not limited to, Hep3B cells and primary hepatocytes.
  • cells are treated with antisense oligonucleotides when the cells reach approximately 60-80% confluency in culture.
  • One reagent commonly used to introduce antisense oligonucleotides into cultured cells includes the cationic lipid transfection reagent LIPOFECTIN® (Invitrogen, Carlsbad, CA). Antisense oligonucleotides are mixed with LIPOFECTIN® in OPTI-MEM® 1 (Invitrogen, Carlsbad, CA) to achieve the desired final concentration of antisense
  • oligonucleotide and a LIPOFECTIN® concentration typically ranges 2 to 12 ug/mL per 100 nM antisense oligonucleotide.
  • Another reagent used to introduce antisense is a LIPOFECTIN® concentration that typically ranges 2 to 12 ug/mL per 100 nM antisense oligonucleotide.
  • oligonucleotides into cultured cells includes LIPOFECTAMINE® (Invitrogen, Carlsbad, CA).
  • Antisense oligonucleotide is mixed with LIPOFECTAMINE® in OPTI-MEM® 1 reduced serum medium (Invitrogen, Carlsbad, CA) to achieve the desired concentration of antisense oligonucleotide and a LIPOFECTAMINE® concentration that typically ranges 2 to 12 ⁇ g/ ⁇ L per 100 nM antisense oligonucleotide.
  • Cells are treated with antisense oligonucleotides by routine methods. Cells are typically harvested 16-24 hours after antisense oligonucleotide treatment, at which time RNA or protein levels of target nucleic acids are measured by methods known in the art and described herein. In general, when treatments are performed in multiple replicates, the data are presented as the average of the replicate treatments.
  • the concentration of antisense oligonucleotide used varies from cell line to cell line. Methods to determine the optimal antisense oligonucleotide concentration for a particular cell line are well known in the art. Antisense oligonucleotides are typically used at concentrations ranging from 1 nM to 500 nM.
  • RNA Isolation can be performed on total cellular RNA or poly(A)+ mRNA.
  • RNA isolation is well known in the art. RNA is prepared using methods well known in the art, for example, using the TRIZOL® Reagent (Invitrogen, Carlsbad, CA) according to the manufacturer's recommended protocols.
  • Target nucleic acid levels can be quantitated by, e.g., Northern blot analysis, competitive polymerase chain reaction (PCR), or quantitative realtime PCR.
  • RNA analysis can be performed on total cellular RNA or poly(A)+ mRNA.
  • RNA isolation is well known in the art.
  • Northern blot analysis is also routine in the art.
  • Quantitative real-time PCR can be conveniently accomplished using the commercially available ABI PRISM® 7600, 7700, or 7900 Sequence Detection System, available from PE- Applied Biosystems, Foster City, CA and used according to manufacturer's instructions.
  • Quantitation of target RNA levels may be accomplished by quantitative real-time PCR using the ABI PRISM® 7600, 7700, or 7900 Sequence Detection System (PE-Applied Biosystems, Foster City, CA) according to manufacturer's instructions. Methods of quantitative real-time PCR are well known in the art.
  • RNA Prior to real-time PCR, the isolated RNA is subjected to a reverse transcriptase (RT) reaction, which produces complementary DNA (cDNA) that is then used as the substrate for the real-time PCR amplification.
  • RT reverse transcriptase
  • cDNA complementary DNA
  • the RT and real-time PCR reactions are performed sequentially in the same sample well.
  • RT and real-time PCR reagents are obtained from Invitrogen (Carlsbad, CA). RT, real-time-PCR reactions are carried out by methods well known to those skilled in the art.
  • Gene (or RNA) target quantities obtained by real time PCR are normalized using either the expression level of a gene whose expression is constant, such as cyclophilin A, or by quantifying total RNA using RIBOGREEN® (Invitrogen, Inc. Carlsbad, CA). Cyclophilin A expression is quantified by real time PCR, by being run simultaneously with the target, multiplexing, or separately. Total RNA is quantified using RIBOGREEN® RNA
  • RNA quantification reagent Invitrogen, Inc. Eugene, OR. Methods of RNA quantification by RIBOGREEN® are taught in Jones, L.J., et al, (Analytical Biochemistry, 1998, 265, 368- 374). A CYTOFLUOR® 4000 instrument (PE Applied Biosystems) is used to measure RIBOGREEN® fluorescence.
  • Probes and primers are designed to hybridize to a nucleic acid. Methods for designing real-time PCR probes and primers are well known in the art, and may include the use of software such as PRIMER EXPRESS® Software (Applied Biosystems, Foster City, CA).
  • Antisense inhibition of nucleic acids can be assessed by measuring protein levels. Protein levels can be evaluated or quantitated in a variety of ways well known in the art, such as immunoprecipitation, Western blot analysis (immunoblotting), enzyme-linked
  • ELISA immunosorbent assay
  • quantitative protein assays protein activity assays (for example, histone deacetylase activity), immunohistochemistry, immunocytochemistry or fluorescence-activated cell sorting (FACS).
  • FACS fluorescence-activated cell sorting
  • Antibodies directed to a target can be identified and obtained from a variety of sources, such as the MSRS catalog of antibodies (Aerie Corporation, Birmingham, MI), or can be prepared via conventional monoclonal or polyclonal antibody generation methods well known in the art.
  • administering results in reduction of expression (e.g., mRNA or protein levels) of the human host cell factor by at least 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 96, 97, 98, 99, or 100%, or a range defined by any two of these values.
  • expression e.g., mRNA or protein levels
  • Antisense compounds are tested in animals to assess their ability to inhibit expression of the target and produce the desired effect, such as reduction in influenza virus replication, reduction in influenza virus infection, and/or prevention or reduction of symptoms or disease associated with influenza virus infection, measurable by the methods provided herein.
  • the methods descrihbed herein for testing antisense compounds may be adapated for testing other nucleic acid compounds, such as siRNAs. With respect to siRNAs, see also Section 5.1.1.2 above and the references cited therein.
  • testing may be performed in normal animals, or in experimental influenza disease models known in the art and described below.
  • antisense oligonucleotides are formulated in a pharmaceutically acceptable diluent, such as phosphate- buffered saline.
  • Administration include any suitable route of administration, such as parenteral, intraperitoneal, intravenous, pulmonary, intranasally, topically, and subcutaneous.
  • RNA is isolated from a relevant tissue (e.g., lung tissue or other epithelial tissue) and changes in target nucleic acid expression are measured.
  • the effect of a compound on virus replication can be assessed by any assay known in the art.
  • assays may involve: (a) contacting a compound or a member of a library of compounds with a cell before (e.g., 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 16 hours, 24 hours or more before), concurrently and/or subsequent to (e.g., 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 16 hours, 24 hours or more after) infection with an influenza virus; and (b) measuring virus replication.
  • the cells can be infected at different MOIs and the effect of a compound on virus replication can be assessed.
  • the MOIs may be 0.001, 0.005, 0.01, 0.05, 0.1, 0.5, 1 , 2.5, or 5.
  • the effect of different concentrations of a compound on virus replication can also be assessed.
  • the cells or other substrate that contains cells (e.g., embryonated eggs) used in the assay should be susceptible to infection by the influenza virus.
  • the cells may be primary cells or established cell lines.
  • the following cells may be used in the assay for influenza virus replication: chicken cells (e.g., primary chick embryo cells or chick kidney cells), Vero cells, MDCK cells, human respiratory epithelial cells (e.g., A549 cells), calf kidney cells, and mink lung cells.
  • the cells used to assess the effect of a compound on virus replication are selected from the following cells or cell lines: MEF, 293T, Huh 7.5, Detroit, and human tracheobronchial epithelial (HTBE; primary lung cells) cells.
  • the cell or cell line is biologically relevant to virus infection.
  • Influenza virus replication can be measured at different times post-infection. For example, virus replication may be measured 6 hours, 12 hours, 16 hours, 24 hours, 48 hours or 72 hours post-infection. Any method known to one of skill in the art can be used measure virus replication. For example, viral replication may be assessed by measuring viral titer (as determined, e.g., by plaque formation), the production of viral proteins (as determined, e.g., by western blot analysis, ELISA or flow cytometry), or the production of viral nucleic acids (as determined, e.g., by RT-PCR or Northern blot analysis) using techniques known to one of skill in the art. See Sections 5.3.1.1-5.3.1.6 below for more details of techniques for measuring viral replication.
  • a compound is considered to inhibit (or reduce) influenza virus replication if the replication of the virus is decreased in the cell contacted with the compound relative to the replication of the virus in a cell contacted with a negative control (e.g., PBS or saline).
  • a negative control e.g., PBS or saline
  • a compound is considered to reduce or inhibit viral replication if it reduces the virus replication by at least 1.5 fold, 2 fold, 3 fold, 4 fold, 5 fold, 6 fold, 7 fold, 8 fold, 9 fold, 10 fold, 15 fold, 20 fold, 25 fold, 30 fold, 35 fold, 40 fold, 45 fold, 50 fold, 100 fold, 500 fold, or 1000 fold relative to virus replication in the absence of compound or the presence of a negative control.
  • a compound is considered to reduce or inhibit viral replication if it reduces the virus replication by 1.5 to 3 fold, 2 to 4 fold, 3 to 5 fold, 4 to 8 fold, 6 to 9 fold, 8 to 10 fold, 2 to 10 fold, 5 to 20 fold, 10 to 40 fold, 10 to 50 fold, 25 to 50 fold, 50 to 100 fold, 75 to 100 fold, 100 to 500 fold, 500 to 1000 fold, or 10 to 1000 fold.
  • a compound is considered to reduce or inhibit viral replication if it reduces the virus replication by approximately 2 logs or more, approximately 3 logs or more, approximately 4 logs or more, approximately 5 logs or more, or 2 to 10 logs or 2 to 5 logs relative to virus replication in the absence of compound or the presence of a negative control.
  • a compound is considered to reduce or inhibit viral replication if it reduces the replication of a viral genome by about at least 1.5 fold, 2, fold, 3 fold, 4 fold, 5 fold, 6 fold, 7 fold, 8 fold, 9 fold, 10 fold, 15 fold, 20 fold, 25 fold, 30 fold, 35 fold, 40 fold, 45 fold, 50 fold, 75 fold, 100 fold, 500 fold, or 1000 fold relative to replication of the viral genome in the absence of a compound or relative to a negative control in an assay described herein or others known to one of skill in the art.
  • a compound is considered to reduce or inhibit viral replication if it reduces the replication of a viral genome by about 1.5 to 3 fold, 2 to 4 fold, 3 to 5 fold, 4 to 8 fold, 6 to 9 fold, 8 to 10 fold, 2 to 10 fold, 5 to 20 fold, 10 to 40 fold, 10 to 50 fold, 25 to 50 fold, 50 to 100 fold, 75 to 100 fold, 100 to 500 fold, 500 to 1000 fold, or 10 to 1000 fold relative to replication of the viral genome in the absence of a compound or relative to a negative control in an assay described herein or others known to one of skill in the art.
  • a compound is considered to reduce or inhibit viral replication if it reduces the replication of a viral genome by at least 1 log, 1.5 logs, 2 logs, 2.5 logs, 3 logs, 3.5 logs, 4 logs, 4.5 logs, 5 logs or more relative to replication of the viral genome in the absence of a compound or relative to a negative control in an assay described herein or others known to one of skill in the art.
  • a compound is considered to reduce or inhibit viral replication if it reduces the synthesis of viral proteins by at least 1.5 fold, 2, fold, 3 fold, 4 fold, 5 fold, 6 fold, 7 fold, 8 fold, 9 fold, 10 fold, 15 fold, 20 fold, 25 fold, 30 fold, 35 fold, 40 fold, 45 fold, 50 fold, 75 fold, 100 fold, 500 fold, or 1000 fold relative to the synthesis of viral proteins in the absence of a compound or relative to a negative control in an assay described herein or others known to one of skill in the art in an assay described herein or others known to one of skill in the art.
  • a compound is considered to reduce or inhibit viral replication if it reduces the synthesis of viral proteins at least 1.5 to 3 fold, 2 to 4 fold, 3 to 5 fold, 4 to 8 fold, 6 to 9 fold, 8 to 10 fold, 2 to 10 fold, 5 to 20 fold, 10 to 40 fold, 10 to 50 fold, 25 to 50 fold, 50 to 100 fold, 75 to 100 fold, 100 to 500 fold, 500 to 1000 fold, or 10 to 1000 fold relative to the synthesis of viral proteins in the absence of a compound or relative to a negative control in an assay described herein or others known to one of skill in the art.
  • a compound is considered to reduce or inhibit viral replication if it reduces the synthesis of viral proteins approximately 1 log, 1.5 logs, 2 logs, 2.5 logs, 3 logs, 3.5 logs, 4 logs, 4.5 logs, 5 logs relative to the synthesis of viral proteins in the absence of a compound or relative to a negative control in an assay described herein or others known to one of skill in the art.
  • a compound is considered to reduce or inhibit viral replication if it results in 1.5 fold or more, 2 fold or more, 3 fold or more, 4 fold or more, 5 fold or more, 6 fold or more, 7 fold or more, 8 fold or more, 9 fold or more, 10 fold or more, 15 fold or more, 20 fold or more, 25 fold or more, 30 fold or more, 35 fold or more, 40 fold or more, 45 fold or more, 50 fold or more, 60 fold or more, 70 fold or more, 80 fold or more, 90 fold or more, or 100 fold or more reduction of viral yield per round of viral replication.
  • a compound results in about a 2 fold or more reduction of viral yield per round of viral replication.
  • a compound results in about a 10 fold or more reduction of viral yield per round of viral replication.
  • a compound is considered to reduce or inhibit viral replication if it reduces viral replication by at least 2 wells of hemagglutinin (HA) in a hemagglutination assay (see Section 5.2.1.7 below), which equals approximately a 75% reduction in viral titer.
  • HA hemagglutinin
  • a compound is considered to reduce or inhibit viral replication if it reduces viral titer by 50% or more, by 55% or more, by 60% or more, by 65% or more, by 70% or more, by 75% or more, by 80% or more, by 85% or more, by 90% or more, or by 95% or more.
  • the effect of a compound on the replication of an influenza A virus is determined.
  • the effect of a compound on the replication of an influenza B virus is determined.
  • the effect of a compound on the replication of an influenza C virus is determined.
  • the effect of a compound on the replication of a currently circulating influenza virus is determined.
  • the effect of a compound on replication of HlNl influenza virus is determined.
  • the effect of a compound on replication of H5N1 influenza virus is determined.
  • the effect of a compound on replication of an attenuated influenza virus is determined. In some embodiments, the effect of a compound on the replication of a naturally occurring strain, variant or mutant of an influenza virus, a mutagenized influenza virus, a reassortant influenza virus and/or a genetically engineered influenza virus can be assessed. In a specific embodiment, the effect of a compound on the replication of a vaccine strain of an influenza virus is determined.
  • a monolayer of the target mammalian cell line is infected with different amounts (e.g. , multiplicity of 3 plaque forming units (pfu) or 5 pfu) of influenza virus and subsequently cultured in the presence or absence of various dilutions of compounds (e.g., 0.1 ⁇ g/ml, 1 ⁇ g/ml, 5 ⁇ g/ml, or 10 g/ml).
  • Infected cultures are harvested 48 hours or 72 hours post infection and titered by standard plaque assays known in the art on the appropriate target cell line (e.g., Vero cells).
  • Flow cytometry can be utilized to detect expression of virus antigens in infected target cells cultured in the presence or absence of compounds (See, e.g., McSharry et al, Clinical Microbiology Rev., 1994, 7:576-604).
  • Non-limiting examples of viral antigens that can be detected on cell surfaces by flow cytometry include, but are not limited to HA of influenza.
  • intracellular viral antigens or viral nucleic acid can be detected by flow cytometry with techniques known in the art.
  • CPE is the morphological changes that cultured cells undergo upon being infected by most viruses. These morphological changes can be observed easily in unfixed, unstained cells by microscopy. Forms of CPE, which can vary depending on the virus, include, but are not limited to, rounding of the cells, appearance of inclusion bodies in the nucleus and/or cytoplasm of infected cells, and formation of syncytia, or polykaryocytes (large cytoplasmic masses that contain many nuclei).
  • the CPE assay can provide a measure of the effect of a compound on virus replication.
  • compounds are serially diluted (e.g. 1000, 500, 100, 50, 10, 1 ⁇ g/ml) and added to 3 wells containing a cell monolayer (preferably mammalian cells at 80-100% confluent) of a 96- well plate.
  • a cell monolayer preferably mammalian cells at 80-100% confluent
  • viruses are added and the plate sealed, incubated at 37°C for the standard time period required to induce near-maximal viral CPE (e.g. , approximately 48 to 120 hours, depending on the virus and multiplicity of infection).
  • CPE When assaying a compound for its potential activity, CPE is read microscopically after a known positive control drug (an antiviral) is evaluated in parallel with compounds in each test.
  • a positive control is ribavirin for influenza.
  • the data is expressed as 50% effective concentrations or approximated virus- inhibitory concentration, 50% endpoint (EC50) and cell-inhibitory concentration, 50% endpoint (IC50).
  • EC50 endpoint
  • IC50 cell-inhibitory concentration
  • SI General selectivity index
  • a compound has an SI of greater than 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 20, 21, 22, 23, 24, 25, 30, 35, 39, 40, 45, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 1,000, or 10,000.
  • a compound has an SI of greater than 10.
  • compounds with an SI of greater than 10 are further assessed in other in vitro and in vivo assays described herein or others known in the art to characterize safety and efficacy.
  • the NR Dye Uptake assay can be used to validate the CPE inhibition assay (See Section 5.3.1.3).
  • the same 96-well microplates used for the CPE inhibition assay can be used.
  • Neutral red is added to the medium, and cells not damaged by virus take up a greater amount of dye.
  • the percentage of uptake indicating viable cells is read on a microplate autoreader at dual wavelengths of 405 and 540 nm, with the difference taken to eliminate background. (See McManus et al, Appl. Environment. Microbiol. 31 :35-38, 1976).
  • An EC 50 is determined for samples with infected cells and contacted with compounds, and an IC 50 is determined for samples with uninfected cells contacted with compounds.
  • Lysed cells and supernatants from infected cultures such as those in the CPE inhibition assay (See Section 5.3.1.3) can be used to assay for virus yield (production of viral particles after the primary infection).
  • virus yield production of viral particles after the primary infection.
  • these supernatants are serially diluted and added onto monolayers of susceptible cells (e.g., Vero cells).
  • the virus is diluted into various concentrations and added to each well containing a monolayer of the target cells in triplicate.
  • the plates are then incubated for a period of time to achieve effective infection of the control sample (e.g., 1 hour with shaking every fifteen minutes).
  • an equal amount of 1% agarose is added to an equal volume of each compound dilution prepared in 2* concentration.
  • final compound concentrations between 0.03 g/ml to 100 ⁇ g/ml can be tested with a final agarose overlay concentration of 0.5%.
  • the drug agarose mixture is applied to each well in 2 ml volume and the plates are incubated for three days, after which the cells are stained with a 1.5% solution of neutral red. At the end of the 4-6 hour incubation period, the neutral red solution is aspirated, and plaques counted using a stereomicroscope. Alternatively, a final agarose concentration of 0.4% can be used. In other embodiments, the plates are incubated for more than three days with additional overlays being applied on day four and on day 8 when appropriate. In another embodiment, the overlay medium is liquid rather than semi-solid.
  • hemagglutination assay cells are contacted with a compound and are concurrently or subsequently infected with the virus (e.g., at an MOI of 1) and the virus is incubated under conditions to permit virus replication (e.g., 20-24 hours).
  • the compounds are preferably present throughout the course of infection.
  • Viral replication and release of viral particles is then determined by hem-agglutination assays using 0.5% chicken red blood cells.
  • a compound is considered to reduce or inhibit viral replication if it reduces viral replication by at least 2 wells of HA, which equals approximately a 75% reduction in viral titer.
  • a compound reduces viral titer in this assay by 50% or more, by 55% or more, by 60% or more, by 65% or more, by 70% or more, by 75% or more, by 80% or more, by 85% or more, by 90% or more, or by 95% or more.
  • compounds differentially affect the viability of uninfected cells and cells infected with virus.
  • the differential effect of a compound on the viability of virally infected and uninfected cells may be assessed using techniques known to one of skill in the art or described herein.
  • compounds are more toxic to cells infected with a virus than uninfected cells.
  • compounds are more toxic to cells infected with a virus than uninfected cells.
  • the compounds preferentially affect the viability of cells infected with a virus.
  • the compounds are not so cytotoxic that they are unsafe for administration to an animal or human subject.
  • cell proliferation can be assayed by measuring Bromodeoxyuridine (BrdU) incorporation (See, e.g., Hoshino et ah, 1986, Int. J. Cancer 38, 369; Campana et ah, 1988, J. Immunol. Meth. 107:79), (3H) thymidine incorporation (See, e.g., Chen, J., 1996, Oncogene 13: 1395-403; Jeoung, J., 1995, J. Biol. Chem.
  • PrdU Bromodeoxyuridine
  • RNA and mRNA and activity can be determined by any method well known in the art.
  • protein can be quantitated by known immunodiagnostic methods such as ELISA, Western blotting or immunoprecipitation using antibodies, including commercially available antibodies.
  • mRNA can be quantitated using methods that are well known and routine in the art, for example, using northern analysis, RNase protection, or polymerase chain reaction in connection with reverse transcription.
  • Cell viability can be assessed by using trypan-blue staining or other cell death or viability markers known in the art.
  • the level of cellular ATP is measured to determined cell viability.
  • cell viability is measured in three-day and seven-day periods using an assay standard in the art, such as the CellTiter-Glo Assay Kit (Promega) which measures levels of intracellular ATP. A reduction in cellular ATP is indicative of a cytotoxic effect.
  • cell viability can be measured in the neutral red uptake assay.
  • visual observation for morphological changes may include enlargement, granularity, cells with ragged edges, a filmy appearance, rounding, detachment from the surface of the well, or other changes.
  • the cells used in the cytotoxicity assay are animal cells, including primary cells and cell lines. In some embodiments, the cells are human cells. In certain embodiments, cytotoxicity is assessed in one or more of the following cell lines: U937, a human monocyte cell line; primary peripheral blood mononuclear cells (PBMC); Huh7, a human hepatoblastoma cell line; 293T, a human embryonic kidney cell line; or THP- 1, monocytic cells. In certain embodiments, cytotoxicity is assessed in one or more of the following cell lines: MDC , MEF, Huh 7.5, Detroit, or human tracheobronchial epithelial (HTBE) cells.
  • PBMC primary peripheral blood mononuclear cells
  • Huh7 a human hepatoblastoma cell line
  • 293T a human embryonic kidney cell line
  • THP- 1, monocytic cells THP- 1, monocytic cells.
  • cytotoxicity is assessed in one or more of the following cell lines
  • Compounds can be tested for in vivo toxicity in animal models.
  • animal models described herein and/or others known in the art, used to test the activities of compounds can also be used to determine the in vivo toxicity of these compounds.
  • animals are administered a range of concentrations of compounds. Subsequently, the animals are monitored over time for lethality, weight loss or failure to gain weight, and/or levels of serum markers that may be indicative of tissue damage (e.g., creatine phosphokinase level as an indicator of general tissue damage, level of glutamic oxalic acid transaminase or pyruvic acid transaminase as indicators for possible liver damage).
  • tissue damage e.g., creatine phosphokinase level as an indicator of general tissue damage, level of glutamic oxalic acid transaminase or pyruvic acid transaminase as indicators for possible liver damage.
  • These in vivo assays may also be adapted to test the toxicity of various
  • the toxicity and/or efficacy of a compound in accordance with the embodiments described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 5 o ED 50 .
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage of a compound identified in accordance with the embodiments described herein for use in humans.
  • the dosage of such agents lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 (i.e., the concentration of the test compound that achieves a half- maximal inhibition of symptoms) as determined in cell culture.
  • IC 50 i.e., the concentration of the test compound that achieves a half- maximal inhibition of symptoms
  • levels in plasma may be measured, for example, by high-performance liquid chromatography. Additional information concerning dosage determination is provided in Section 5.5.4, infra.
  • any technique known to one of skill in the art can be used to determine whether a compound has an apoptotic effect.
  • a fluorescence-based assay for caspase-3 activity can be used to detect whether a compound has a pro- or anti-apoptotic effect.
  • cells are seeded into 60 mm tissue culture treated dishes at 1.5xl0 6 cells per dish and allowed to incubate for 24 hours. After incubation, the medium is aspirated and the cells are washed with PBS. Fresh DMEM post-infection medium was added, containing compounds at the same concentrations as has been used for the viral infections.
  • cells are treated with any known inducer of apoptosis, for example, staurosporin at a concentration of 5 ⁇ . Cells are incubated for 6 hours. Subsequently, they are harvested, washed twice with PBS, lysed and incubated with the colorimetric substrate for an additional hour, at which time fluorescence is measured. An increase in fluorescence relative to a negative control or cells not treated with the compound indicates that the compound is pro-apoptotic.
  • any known inducer of apoptosis for example, staurosporin at a concentration of 5 ⁇ . Cells are incubated for 6 hours. Subsequently, they are harvested, washed twice with PBS, lysed and incubated with the colorimetric substrate for an additional hour, at which time fluorescence is measured. An increase in fluorescence relative to a negative control or cells not treated with the compound indicates that the compound is pro-apoptotic.
  • Compounds and compositions are preferably assayed in vivo for the desired therapeutic or prophylactic activity prior to use in humans.
  • in vivo assays can be used to determine whether it is preferable to administer a compound and/or another therapeutic agent.
  • the compound can be administered before the animal is infected with the virus.
  • a compound can be administered to the animal at the same time that the animal is infected with the virus.
  • the compound is administered after a viral infection in the animal.
  • a compound is administered to the animal at the same time that the animal is infected with the virus to treat and/or manage the viral infection.
  • the compound is administered to the animal more than one time.
  • Compounds can be tested for antiviral activity against virus in animal models systems including, but are not limited to, rats, mice, chicken, cows, monkeys, pigs, goats, sheep, dogs, rabbits, guinea pigs, etc.
  • compounds are tested in a mouse model system.
  • Such model systems are widely used and well-known to the skilled artisan.
  • Compounds can also be tested for replication enhancing activity toward virus replication in animal models systems including, but are not limited to, rats, mice, chicken, cows, monkeys, pigs, goats, sheep, dogs, rabbits, guinea pigs, etc.
  • compounds are tested in a mouse model system.
  • Such model systems are widely used and well-known to the skilled artisan.
  • Non-limiting examples of animal models for influenza virus are provided in Section 5.2.5.1 below.
  • Animals are infected with virus and concurrently or subsequently treated with a compound or placebo. Alternatively, animals are treated with a compound or placebo and subsequently infection with virus.
  • Samples obtained from these animals e.g., serum, urine, sputum, semen, saliva, plasma, or tissue sample
  • samples obtained from these animals can be tested for viral replication via well known methods in the art, e.g., those that measure altered viral titers (as determined, e.g., by plaque formation), the production of viral proteins (as determined, e.g., by Western blot, ELISA, or flow cytometry analysis) or the production of viral nucleic acids (as determined, e.g., by RT-PCR or northern blot analysis).
  • tissue samples are homogenized in phosphate-buffered saline (PBS), and dilutions of clarified homogenates are adsorbed for 1 hour at 37°C onto monolayers of cells (e.g., Vero, CEF or MDCK cells).
  • PBS phosphate-buffered saline
  • histopathologic evaluations are performed after infection, preferably evaluations of the organ(s) the virus is known to target for infection.
  • Virus immunohistochemistry can be performed using a viral-specific monoclonal antibody.
  • the effect of a compound on the virulence of a virus can also be determined using in vivo assays in which the titer of the virus in an infected subject administered a compound, the length of survival of an infected subject administered a compound, the immune response in an infected subject administered a compound, the number, duration and/or severity of the symptoms in an infected subject administered a compound, and/or the time period before onset of one or more symptoms in an infected subject administered a compound is assessed. Techniques known to one of skill in the art can be used to measure such effects.
  • influenza-infected mice administered to the influenza-infected mice include pneumonia-associated death, serum al- acid glycoprotein increase, animal weight, lung virus assayed by hemagglutinin, lung virus assayed by plaque assays, and histopathological change in the lung.
  • significance e.g., a P value of 0.05 or less.
  • Nasal turbinates and trachea may be examined for epithelial changes and subepithelial inflammation.
  • the lungs may be examined for bronchiolar epithelial changes and peribronchiolar inflammation in large, medium, and small or terminal bronchioles.
  • the alveoli are also evaluated for inflammatory changes.
  • the medium bronchioles are graded on a scale of 0 to 3+ as follows: 0 (normal: lined by medium to tall columnar epithelial cells with ciliated apical borders and basal pseudostratified nuclei; minimal inflammation); 1+
  • 2+ prominent changes in the epithelial layer ranging from attenuation to marked proliferation; cells disorganized and layer outline irregular at the luminal border
  • 3+ epitophelial layer markedly disrupted and disorganized with necrotic cells visible in the lumen; some bronchioles attenuated and others in marked reactive
  • the trachea is graded on a scale of 0 to 2.5+ as follows: 0 (normal: Lined by medium to tall columnar epithelial cells with ciliated apical border, nuclei basal and pseudostratified. Cytoplasm evident between apical border and nucleus. Occasional small focus with squamous cells); 1+ (focal squamous metaplasia of the epithelial layer); 2+ (diffuse squamous metaplasia of much of the epithelial layer, cilia may be evident focally); 2.5+ (diffuse squamous metaplasia with very few cilia evident).
  • Virus immunohistochemistry is performed using a viral-specific monoclonal antibody (e.g. NP-, N- or HN-specific monoclonal antibodies). Staining is graded 0 to 3+ as follows: 0 (no infected cells); 0.5+ (few infected cells); 1+ (few infected cells, as widely separated individual cells); 1.5+ (few infected cells, as widely separated singles and in small clusters); 2+ (moderate numbers of infected cells, usually affecting clusters of adjacent cells in portions of the epithelial layer lining bronchioles, or in small sublobular foci in alveoli); 3+ (numerous infected cells, affecting most of the epithelial layer in bronchioles, or widespread in large sublobular foci in alveoli).
  • a viral-specific monoclonal antibody e.g. NP-, N- or HN-specific monoclonal antibodies.
  • a compound that is a candidate for use in human subjects is assessed human subjects suffering from an influenza virus infection.
  • a candidate compound or a control compound is administered to the human subject, and the effect of a test compound on viral replication is determined by, e.g., analyzing the level of the virus or viral nucleic acids in a biological sample (e.g., serum or plasma).
  • a candidate compound that inhibits virus replication can be identified by
  • a decrease in viral replication can be detected by comparing the level of virus replication in a subject or group of subjects before and after the administration of a candidate compound.
  • the effect of a candidate compound on the severity of one or more symptoms associated with an influenza virus infection is assessed in a subject having an influenza virus infection.
  • a candidate compound or a control compound is administered to a human subject suffering from an influenza virus infection and the effect of the candidate compound on one or more symptoms of the virus infection is determined.
  • a candidate compound that reduces one or more symptoms can be identified by comparing the subjects treated with a control compound to the subjects treated with the candidate compound. Techniques known to physicians familiar with infectious diseases can be used to determine whether a candidate compound reduces one or more symptoms associated with the influenza virus infection.
  • compositions comprising a compound that targets one or more human host cell factors involved in influenza virus replication. Such compositions may be in a dose effective to modulate influenza virus replication. Such compositions may be in a dose effective to reduce or inhibit influenza virus replication.
  • Such compositions may be pharmaceutical compositions, and may additionally comprise a pharmaceutically acceptable carrier known in the art or described herein. Such pharmaceutical compositions may be in a dose effective to reduce or inhibit a symptom or disease associated with influenza virus infection.
  • compositions and pharmaceutical compositions may include, by non-limiting example, (i) a compound that targets an aforementioned category of human host cell factor; (ii) a compound that targets a human host cell factor in such a category; (iii) a compound that targets an aforementioned human host cell factor; (iv) an aforementioned nucleic acid compound, e.g., an siRNA; or (v) an aforementioned small molecule.
  • Such compositions may also include another active agent, for example, another compound that targets a human host cell factor involved in influenza virus replication described herein.
  • the compositions, including the pharmaceutical compositions, described herein contain the compound in an amount that is not significantly toxic to the cell, tissue, or subject for which it is intended. Methods of testing toxicity include any method known in the art, for example, as described in Sections 5.2.3 and 6 infra.
  • compositions comprising the compound and a carrier, excipient or diluent.
  • compositions comprising a compound and a pharmaceutically acceptable carrier, excipient, or diluent.
  • compositions comprising an effective amount of a compound and a pharmaceutically acceptable carrier, excipient, or diluent.
  • the pharmaceutical compositions comprise one or more of the compounds that reduce or inhibit influenza virus infection or replication described herein.
  • the pharmaceutical compositions are suitable for veterinary and/or human administration.
  • compositions provided herein can be in any form that allows for the composition to be administered to a subject, preferably a human.
  • the term "pharmaceutically acceptable carrier, excipient or diluent” means a carrier, excipient or diluent approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant (e.g., Freund's adjuvant (complete and incomplete)), excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a specific carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin.
  • compositions and dosage forms comprise one or more excipients.
  • Suitable excipients are well-known to those skilled in the art of pharmacy, and non limiting examples of suitable excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. Whether a particular excipient is suitable for incorporation into a pharmaceutical composition or dosage form depends on a variety of factors well known in the art including, but not limited to, the way in which the dosage form will be administered to a patient and the specific active ingredients in the dosage form.
  • the composition or single unit dosage form if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • Lactose free compositions can comprise excipients that are well known in the art and are listed, for example, in the U.S. Pharmacopeia (USP) SP (XXI)/NF (XVI).
  • USP U.S. Pharmacopeia
  • lactose free compositions comprise an active ingredient, a binder/filler, and a lubricant in pharmaceutically compatible and pharmaceutically acceptable amounts.
  • Specific lactose free dosage forms comprise a compound, microcrystalline cellulose, pre gelatinized starch, and magnesium stearate.
  • anhydrous pharmaceutical compositions and dosage forms comprising one or more compounds, since water can facilitate the degradation of some compounds.
  • water e.g., 5%
  • water is widely accepted in the pharmaceutical arts as a means of simulating long term storage in order to determine characteristics such as shelf life or the stability of formulations over time. See, e.g., Jens T. Carstensen, Drug Stability: Principles & Practice, 2d. Ed., Marcel Dekker, NY, NY, 1995, pp. 379 80.
  • water and heat accelerate the decomposition of some compounds.
  • the effect of water on a formulation can be of great significance since moisture and/or humidity are commonly encountered during manufacture, handling, packaging, storage, shipment, and use of formulations.
  • compositions and dosage forms provided herein can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • Compositions and dosage forms that comprise lactose and at least one compound that comprises a primary or secondary amine are preferably anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected.
  • anhydrous composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions are preferably packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e.g., vials), blister packs, and strip packs.
  • compositions and dosage forms that comprise one or more agents that reduce the rate by which a compound will decompose.
  • agents which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers.
  • compositions and single unit dosage forms can take the form of solutions, suspensions, emulsions, gels, lotions, or creams, tablets, pills, capsules, powders, sustained- release formulations and the like.
  • Oral formulations can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc.
  • Such compositions and dosage forms will contain a prophylactically or therapeutically effective amount of a compound preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
  • the formulation should suit the mode of administration.
  • the compositions or single unit dosage forms are sterile and in suitable form for administration to a subject, preferably an animal subject, more preferably a mammalian subject, and most preferably a human subject.
  • compositions provided herein are formulated to be compatible with the intended route of administration.
  • routes of administration include, but are not limited to, topical, parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), intranasal, transdermal (topical), transmucosal, intra-synovial and rectal administration.
  • the composition is formulated in accordance with routine procedures as a composition adapted for topical, intravenous, pulmonary , subcutaneous, intramuscular, oral, intranasal or topical administration to human beings.
  • a composition is formulated in accordance with routine procedures for subcutaneous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
  • dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic gelatin capsules; cachets; troches; lozenges; dispersions;
  • suppositories ointments; cataplasms (poultices); pastes; powders; dressings; creams or lotions; plasters; solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a patient, including suspensions (e.g., aqueous or non aqueous liquid suspensions, oil in water emulsions, or a water in oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a patient; and sterile solids (e.g., crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a patient.
  • suspensions e.g., aqueous or non aqueous liquid suspensions, oil in water emulsions, or a water in oil liquid emulsions
  • solutions elixirs
  • composition, shape, and type of dosage forms will typically vary depending on their use.
  • compositions provided herein are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • compositions provided herein that are suitable for oral
  • dosage forms such as, but are not limited to, tablets (e.g., chewable tablets), caplets, capsules, and liquids (e.g., flavored syrups).
  • dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton PA (1990).
  • Typical oral dosage forms provided herein are prepared by combining a compound in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques.
  • Excipients can take a wide variety of forms depending on the form of preparation desired for administration.
  • excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents.
  • excipients suitable for use in solid oral dosage forms include, but are not limited to, starches, sugars, micro crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.
  • tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or nonaqueous techniques. Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
  • a tablet can be prepared by compression or molding.
  • Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free flowing form such as powder or granules, optionally mixed with an excipient.
  • Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • excipients that can be used in oral dosage forms provided herein include, but are not limited to, binders, fillers, disintegrants, and lubricants.
  • Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre gelatinized starch, hydroxypropyl methyl cellulose, (e.g., Nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof.
  • fillers suitable for use in the pharmaceutical compositions and dosage forms provided herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre gelatinized starch, and mixtures thereof.
  • the binder or filler in pharmaceutical compositions provided herein is typically present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.
  • Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AVICEL PH 101, AVICEL PH 103 AVICEL RC 581, AVICEL PH 105 (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, PA), and mixtures thereof.
  • a specific binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC 581.
  • Suitable anhydrous or low moisture excipients or additives include AVICEL PH 103 and Starch 1500 LM.
  • Disintegrants are used in the compositions provided herein to provide tablets that disintegrate when exposed to an aqueous environment. Tablets that contain too much disintegrant may disintegrate in storage, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredients should be used to form solid oral dosage forms provided herein. The amount of disintegrant used varies based upon the type of formulation, and is readily discernible to those of ordinary skill in the art. Typical pharmaceutical compositions comprise from about 0.5 to about 15 weight percent of disintegrant, specifically from about 1 to about 5 weight percent of disintegrant.
  • Disintegrants that can be used in pharmaceutical compositions and dosage forms provided herein include, but are not limited to, agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, pre gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.
  • Lubricants that can be used in pharmaceutical compositions and dosage forms provided herein include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and mixtures thereof.
  • calcium stearate e.g., magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc
  • hydrogenated vegetable oil e.g., peanut oil, cottonseed oil
  • Additional lubricants include, for example, a syloid silica gel (AEROSIL 200, manufactured by W.R. Grace Co. of Baltimore, MD), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Piano, TX), CAB O SIL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, MA), and mixtures thereof. If used at all, lubricants are typically used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated. [00281]
  • a compound can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Patent Nos.: 3,845,770; 3,916,899; 3,536,809;
  • Such dosage forms can be used to provide slow or controlled release of one or more active ingredients using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions.
  • Suitable controlled release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active ingredients of the compositions described herein.
  • the embodiments described herein thus encompass single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled release.
  • controlled release pharmaceutical products have a common goal of improving drug therapy over that achieved by their noncontrolled counterparts.
  • the use of an optimally designed controlled release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time.
  • Advantages of controlled release formulations include extended activity of the drug, reduced dosage frequency, and increased patient compliance.
  • controlled release formulations can be used to affect the time of onset of action or other characteristics, such as blood levels of the drug, and can thus affect the occurrence of side (e.g., adverse) effects.
  • Controlled release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, temperature, enzymes, water, or other physiological conditions or agents.
  • Parenteral dosage forms can be administered to patients by various routes including, but not limited to, subcutaneous, intravenous (including bolus injection), intramuscular, and intraarterial. Because their administration typically bypasses patients' natural defenses against contaminants, parenteral dosage forms are preferably sterile or capable of being sterilized prior to administration to a patient. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions.
  • Suitable vehicles that can be used to provide parenteral dosage forms provided herein are well known to those skilled in the art. Examples include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
  • aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection
  • Agents that increase the solubility of one or more of the compounds provided herein can also be incorporated into the parenteral dosage forms provided herein.
  • Transdermal, topical, and mucosal dosage forms include, but are not limited to, ophthalmic solutions, sprays, aerosols, creams, lotions, ointments, gels, solutions, emulsions, suspensions, or other forms known to one of skill in the art. See, e.g., Remington's Pharmaceutical Sciences, 16th and 18th eds., Mack Publishing, Easton PA (1980 & 1990); and Introduction to Pharmaceutical Dosage Forms, 4th ed., Lea & Febiger, Philadelphia (1985). Dosage forms suitable for treating mucosal tissues within the oral cavity can be formulated as mouthwashes or as oral gels. Further, transdermal dosage forms include "reservoir type" or "matrix type” patches, which can be applied to the skin and worn for a specific period of time to permit the penetration of a desired amount of active ingredients.
  • Suitable excipients e.g., carriers and diluents
  • other materials that can be used to provide transdermal, topical, and mucosal dosage forms provided herein are well known to those skilled in the pharmaceutical arts, and depend on the particular tissue to which a given pharmaceutical composition or dosage form will be applied.
  • typical excipients include, but are not limited to, water, acetone, ethanol, ethylene glycol, propylene glycol, butane 1,3 diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof to form lotions, tinctures, creams, emulsions, gels or ointments, which are non toxic and pharmaceutically acceptable.
  • Moisturizers or humectants can also be added to pharmaceutical compositions and dosage forms if desired. Examples of such additional ingredients are well known in the art. See, e.g., Remington's Pharmaceutical Sciences, 16th and 18th eds., Mack Publishing, Easton PA (1980 & 1990).
  • penetration enhancers can be used to assist in delivering the active ingredients to the tissue.
  • Suitable penetration enhancers include, but are not limited to: acetone; various alcohols such as ethanol, oleyl, and tetrahydrofuryl; alkyl sulfoxides such as dimethyl sulfoxide; dimethyl acetamide; dimethyl formamide; polyethylene glycol; pyrrolidones such as
  • polyvinylpyrrolidone polyvinylpyrrolidone
  • ollidon grades Pieris, Polyvidone
  • urea various water soluble or insoluble sugar esters such as Tween 80 (polysorbate 80) and Span 60 (sorbitan monostearate).
  • the pH of a pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied may also be adjusted to improve delivery of one or more compounds.
  • the polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery.
  • Agents such as stearates can also be added to pharmaceutical compositions or dosage forms to advantageously alter the hydrophilicity or lipophilicity of one or more compounds so as to improve delivery.
  • stearates can serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, and as a delivery enhancing or penetration enhancing agent.
  • Different salts, hydrates or solvates of the compounds can be used to further adjust the properties of the resulting composition.
  • the compositions are in oral, injectable, or transdermal dosage forms. In one specific embodiment, the compositions are in oral dosage forms. In one specific embodiment, the compositions are in intranasal dosage forms. In another specific embodiment, the compositions are in the form of injectable dosage forms. In one specific embodiment, the compositions are in topical dosage forms. In another specific embodiment, the compositions are in the form of transdermal dosage forms.
  • nucleic acid molecules such as siRNAs
  • administration may be carried out by known methods, wherein a nucleic acid is introduced into a desired target cell in vitro or in vivo.
  • Commonly used gene transfer techniques include calcium phosphate, DEAE-dextran, electroporation and microinjection and viral methods (Graham, F. L. and van der Eb, A. J. (1973) Virol. 52, 456; McCutchan, J. H. and Pagano, J. S. (1968), J. Natl.
  • a composition may be in form of a solution, e.g. an injectable solution, a cream, ointment, tablet, suspension or the like.
  • the composition may be administered in any suitable way, e.g. by injection, by oral, topical, nasal, rectal application etc.
  • the carrier may be any suitable pharmaceutical carrier.
  • a carrier is used, which is capable of increasing the efficacy of the RNA molecules to enter the target-cells.
  • Suitable examples of such carriers are liposomes, particularly cationic liposomes.
  • a further preferred administration method is injection
  • kits for reducing or inhibiting influenza virus replication comprising contacting a cell infected with an influenza virus with a compound, or
  • a method for reducing or inhibiting replication of an influenza virus comprises: (a) infecting a cell with an influenza virus; and (b) contacting the cell with such a compound or composition in an amount sufficient to reduce or inhibit replication of the influenza virus. Also provided herein are methods for reducing or inhibiting influenza virus replication, comprising: (a) contacting a cell with such a compound or composition in an amount sufficient to reduce or inhibit replication of an influenza virus; and (b) infecting the cell with the influenza virus.
  • a compound or composition comprising the compound is considered to reduce or inhibit influenza virus replication if it reduces the amount of influenza virus replication as measured compared to a control, such as, for example, influenza virus replication in the absence of the compound or composition, or influenza virus replication in the presence of a negative control.
  • the compound or composition is contacted to a cell at risk for influenza virus infection.
  • Compounds for use in such methods may include, by non-limiting example, (i) a compound that targets an aforementioned category of human host cell factor; (ii) a compound that targets a human host cell factor in such a category; (iii) a compound that targets an aforementioned human host cell factor; (iv) an aforementioned siRNA; or (v) an
  • the cell is contacted with an influenza virus concurrently with the compound, or within, for example, 5 seconds, 15 seconds, 30 seconds, 1 minute, 5 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 6 hours, 12 hours, 16 hours or 24 hours, of each other.
  • a pharmaceutical composition comprising a compound, e.g., nucleic acid compound (e.g., siRNA) or small molecule, that targets one or more human host cell factors involved in influenza virus replication in an amount sufficient to reduce the influenza virus infection.
  • a compound e.g., nucleic acid compound (e.g., siRNA) or small molecule
  • the subject is a human.
  • Compounds for use in such methods may include, by non-limiting example, (i) a compound that targets an aforementioned category of human host cell factor; (ii) a compound that targets a human host cell factor in such a category; (iii) a compound that targets an aforementioned human host cell factor; (iv) an aforementioned siRNA; or (v) an
  • kits for treating a symptom or disease associated with an influenza virus infection comprising administering to a subject in need thereof a
  • compositions comprising a compound, e.g., nucleic acid compound (e.g., siRNA) or small molecule, that targets one or more human host cell factors involved in influenza virus replication in an amount sufficient to reduce the symptom or disease associated with the influenza virus infection.
  • a compound e.g., nucleic acid compound (e.g., siRNA) or small molecule, that targets one or more human host cell factors involved in influenza virus replication in an amount sufficient to reduce the symptom or disease associated with the influenza virus infection.
  • the subject is infected with an influenza virus.
  • the subject is at risk for infection with an influenza virus.
  • the subject is a human.
  • Compounds for use in such methods may include, by non-limiting example, (i) a compound that targets an influenza virus.
  • aforementioned category of human host cell factor (ii) a compound that targets a human host cell factor in such a category; (iii) a compound that targets an aforementioned human host cell factor; (iv) an aforementioned siRNA; or (v) an aforementioned small molecule.
  • Also provided herein are methods for preventing a symptom or disease associated with an influenza virus infection comprising administering to a subject in need thereof a composition comprising a compound, e.g., nucleic acid compound (e.g., siRNA) or small molecule, that targets one or more human host cell factors involved in influenza virus replication in an amount sufficient to prevent or reduce the symptom or disease associated with the influenza virus infection.
  • a compound e.g., nucleic acid compound (e.g., siRNA) or small molecule
  • the subject is infected with an influenza virus.
  • the subject is at risk for infection with an influenza virus.
  • the subject is a human.
  • Compounds for use in such methods may include, by non-limiting example, (i) a compound that targets an aforementioned category of human host cell factor; (ii) a compound that targets a human host cell factor in such a category; (iii) a compound that targets an aforementioned human host cell factor; (iv) an aforementioned siR A; or (v) an aforementioned small molecule.
  • the compounds, compositions, and pharmaceutical compositions used in an amount that is not significantly toxic to the cell, tissue, or subject for which it is intended are not significantly toxic to the cell, tissue, or subject for which it is intended.
  • Methods of testing toxicity include any method known in the art, for example, as described supra and in Section 6 below.
  • the aforementioned methods may optionally comprise use of the compound that targets a human host cell factor involved in influenza virus replication in combination with one or more additional active agents.
  • additional active agents include, for example, one or more additional antiviral agents, e.g., an aforementioned compound that targets human host cell factors involved in influenza virus replication; an antibiotic; an immunomodulatory agent; and an agent used in the treatment or prophylaxis of one or more pulmonary diseases described herein or known in the art.
  • the subject is a human.
  • the influenza virus is an influenza A virus.
  • the influenza virus is an influenza B virus.
  • the influenza virus is an influenza C virus. Any type, subtype, or strain of influenza virus described herein or known in the art may be targeted in accordance with the embodiments described herein.
  • the influenza virus is of human origin.
  • the influenza virus is of avian origin (e.g., H5N1).
  • the influenza virus is of swine origin (e.g., H1N1).
  • kits for preventing, treating and/or managing an influenza virus infection comprising administering to a subject in need thereof one or more compounds described herein.
  • a method of preventing, treating and/or managing an influenza virus infection comprising administering to a subject in need thereof a dose of a prophylactically or therapeutically effective amount of one or more compounds described herein or a composition (e.g., a pharmaceutical composition) comprising a compound described herein.
  • a compound or a composition described herein may be used as any line of therapy (e.g., a first, second, third, fourth or fifth line therapy) for an influenza virus infection.
  • the subject to be treated is severely ill.
  • the subject to be treated is unresponsive, or poorly responsive, to one or more previous antiviral therapies.
  • influenza virus infections to be treated in accordance with this aspect include one or more of an influenza A virus, influenza B virus, or influenza C virus.
  • influenza A virus is an H5N1 isolate.
  • influenza A virus is an H1N1 isolate.
  • influenza virus infects humans. In some embodiment, the influenza virus infects humans.
  • influenza virus is a naturally occurring strain, variant or mutant of an influenza virus, a mutagenized influenza virus, a reassortant influenza virus and/or a genetically engineered influenza virus.
  • a compound described herein is the only active ingredient administered to prevent, treat and/or manage an influenza virus infection.
  • the compound is the only active ingredient in a composition that is administered to prevent, treat and/or manage an influenza virus infection or symptom or disease associated therewith.
  • more than one such compound, or the compound together with another therapy, is administered in order to achieve a synergistic effect.
  • the compound specifically interferes with the replication of an influenza virus. In other embodiments, the compound interferes with the replication of influenza virus and one or more other viruses. In some embodiments, the compound reduces the viral replication of one type, subtype or strain of influenza virus more than another. For example, the compound may reduce the replication of an influenza A virus more than it reduces the replication of an influenza B virus, and vice versa.
  • the embodiments described herein encompass methods for preventing, treating, and/or managing an influenza virus infection for which no antiviral therapy is available.
  • the embodiments described herein also encompass methods for preventing, treating, and/or managing an influenza virus infection as an alternative to other conventional therapies.
  • the other therapies are currently being used, have been used or are known to be useful in the prevention, treatment and/or management of a viral infection.
  • Non-limiting examples of such therapies are provided below.
  • one or more compounds described herein are administered to a subject in combination with one or more therapies.
  • one or more compounds described herein are administered to a subject in combination with a supportive therapy, a pain relief therapy, or another therapy that does not have antiviral activity.
  • the therapy is a treatment of pulmonary disease.
  • the combination therapies can be administered sequentially or concurrently.
  • the combination therapies comprise an comprise a compound that targets a human host cell factor involved in influenza virus replication described and at least one other therapy which has the same mechanism of action.
  • the combination therapies described herein and at least one other therapy which has a different mechanism of action than the compound.
  • the combination therapies improve the prophylactic and/or therapeutic effect of a compound described herein by functioning together with the compound to have an additive or synergistic effect.
  • the combination therapies reduce the side effects associated with each therapy taken alone.
  • the prophylactic or therapeutic agents of the combination therapies can be administered to a subject in the same pharmaceutical composition.
  • the prophylactic or therapeutic agents of the combination therapies can be administered concurrently to a subject in separate pharmaceutical compositions.
  • the prophylactic or therapeutic agents may be administered to a subject by the same or different routes of administration.
  • a compound described herein, a composition comprising a compound described herein, or a combination therapy is administered to a subject suffering from an influenza virus infection.
  • a compound described herein, a composition comprising a compound described herein, or a combination therapy is administered to a subject suffering from an influenza virus infection.
  • a compound described herein, a composition comprising a compound described herein, or a combination therapy is administered to a subject that lives in a region where there has been or might be an outbreak with an influenza virus infection.
  • the influenza virus infection is an active infection.
  • influenza virus infection is chronic.
  • the compound, the composition comprising the compound or a combination therapy is administered to a mammal which is 0 to 6 months old, 6 to 12 months old, 1 to 5 years old, 5 to 10 years old, 10 to 15 years old, 15 to 20 years old, 20 to 25 years old, 25 to 30 years old, 30 to 35 years old, 35 to 40 years old, 40 to 45 years old, 45 to 50 years old, 50 to 55 years old, 55 to 60 years old, 60 to 65 years old, 65 to 70 years old, 70 to 75 years old, 75 to 80 years old, 80 to 85 years old, 85 to 90 years old, 90 to 95 years old or 95 to 100 years old.
  • the compound, a composition comprising the compound or a combination therapy is administered to a human at risk for an influenza virus infection.
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is
  • the subject is a human 0 to 6 months old, 6 to 12 months old, 1 to 5 years old, 5 to 10 years old, 5 to 12 years old, 10 to 15 years old, 15 to 20 years old, 13 to 19 years old, 20 to 25 years old, 25 to 30 years old, 20 to 65 years old, 30 to 35 years old, 35 to 40 years old, 40 to 45 years old, 45 to 50 years old, 50 to 55 years old, 55 to 60 years old, 60 to 65 years old, 65 to 70 years old, 70 to 75 years old, 75 to 80 years old, 80 to 85 years old, 85 to 90 years old, 90 to 95 years old or 95 to 100 years old.
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is administered to a pet, e.g., a dog or cat.
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is administered to a farm animal or livestock, e.g., pig, cow, horse, chicken, etc.
  • a compound described herein, a compound comprising a compound described herein or a combination therapy is administered to a bird, e.g., duck or chicken.
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is administered to a primate, preferably a human, or another mammal, such as a pig, cow, horse, sheep, goat, dog, cat and rodent, in an immunocompromised state or immunosuppressed state or at risk for becoming immunocompromised or immunosuppressed.
  • a compound, a composition comprising a compound described herein or a combination therapy is administered to a subject receiving or recovering from immunosuppressive therapy.
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is administered to a subject that has or is at risk of getting cancer, AIDS, another viral infection, or a bacterial infection.
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is administered to a subject that is, will or has undergone surgery, chemotherapy and/or radiation therapy.
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is administered to a subject that has, will have or had a tissue transplant.
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is administered to a subject that smokes, has asthma, emphysema, allergies, bronchitis, cystic fibrosis, pulmonary fibrosis, or another disease which makes the subject susceptible to an influenza virus infection.
  • the compound, a composition comprising the compound or a combination therapy is administered to a subject that lives or works at a nursing home, a group home (i.e., a home for 10 or more subjects), or a prison.
  • the compound, a composition comprising the compound or a combination therapy is administered to a subject that attends or works at a school (e.g., elementary school, middle school, junior high school, high school or university) or daycare.
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is administered to a subject that works in the healthcare area, such as a doctor or a nurse, or in a hospital.
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is administered to a subject that is pregnant or plans on becoming pregnant.
  • a patient is administered a compound described herein, a composition comprising a compound described herein or a combination therapy before any adverse effects or intolerance to therapies other than the compound develops.
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is administered to refractory patients.
  • a refractory patient is a patient refractory to a standard antiviral therapy.
  • a patient with a viral infection is refractory to a therapy when the infection has not significantly been eradicated and/or the symptoms have not been
  • a patient with a viral infection is refractory when viral replication has not decreased or has increased.
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is administered to a patient to prevent the onset or reoccurrence of an influenza virus infection in a patient at risk of developing such an infection.
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is administered to a patient who is susceptible to adverse reactions to conventional therapies.
  • a compound described herein, a composition comprising a compound described herein or a combination therapy to a patient who has proven refractory to therapies other than the compound, but are no longer on these therapies.
  • the patients being managed or treated in accordance with the methods described herein are patients already being treated with antibiotics, antivirals, antifungals, or other biological therapy/immunotherapy. Among these patients are refractory patients, patients who are too young for conventional therapies ⁇ and patients with reoccurring viral infections despite management or treatment with existing therapies.
  • the subject being administered a compound described herein, a composition comprising a compound described herein or a combination therapy has not received a therapy prior to the administration of the compound or composition or combination therapy.
  • a compound described herein, a composition comprising a compound described herein or a combination therapy is administered to a subject who has received a therapy prior to administration of the compound, composition or combination therapy.
  • the subject administered a compound described herein, a composition comprising a compound described herein or a combination therapy was refractory to a prior therapy or experienced adverse side effects to the prior therapy or the prior therapy was discontinued due to unacceptable levels of toxicity to the subject.
  • a compound described herein is preferably administered as a component of a composition that optionally comprises a pharmaceutically acceptable vehicle.
  • the composition can be administered orally, or by any other convenient route, for example, topically, by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal, and intestinal mucosa) and may be administered together with another biologically active agent. Administration can be systemic or local.
  • Various delivery systems are known, e.g., encapsulation in liposomes,
  • microparticles can be used to administer the compound and pharmaceutically acceptable salts thereof.
  • Methods of administration include but are not limited to parenteral, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, oral, sublingual, intranasal, intracerebral, intravaginal, transdermal, rectally, by inhalation, or topically, particularly to the ears, nose, eyes, or skin.
  • the mode of administration is left to the discretion of the practitioner. In most instances, administration will result in the release of a compound into the bloodstream.
  • a compound described herein may be desirable to administer a compound described herein locally. This may be achieved, for example, and not by way of limitation, by local infusion, topical application, e.g., in conjunction with a wound dressing, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent, or via perfusion in a fluorocarbon or synthetic pulmonary surfactant.
  • a compound is formulated as a suppository, with traditional binders and vehicles such as triglycerides.
  • the compound can be administered topically, ocularly, intranasally or by an inhaler or nebulizer.
  • the compound is delivered in a vesicle, in particular a liposome (See Langer, 1990, Science 249: 1527 1533; Treat et al, in Liposomes in the Therapy of Infectious Disease and Bacterial infection, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353 365 (1989); Lopez Berestein, ibid., pp. 317 327; See generally ibid.).
  • a liposome See Langer, 1990, Science 249: 1527 1533; Treat et al, in Liposomes in the Therapy of Infectious Disease and Bacterial infection, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353 365 (1989); Lopez Berestein, ibid., pp. 317 327; See generally ibid.).
  • the compound is delivered in a controlled release system (See, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 1 15 138 (1984)). Examples of controlled-release systems are discussed in the review by Langer, 1990, Science 249: 1527 1533 may be used.
  • a pump may be used (See Langer, supra; Sefton, 1987, CRC Crit. Ref. Biomed. Eng. 14:201 ; Buchwald et al, 1980, Surgery 88:507; Saudek et al, 1989, N. Engl. J. Med. 321 :574).
  • polymeric materials can be used (See Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, 1983, J. Macromol. Sci. Rev. Macromol. Chem. 23:61 ; See also Levy et al, 1985, Science 228:190; During et al, 1989, Ann. Neurol. 25:351; Howard et al, 1989, J. Neurosurg. 71 : 105).
  • a controlled-release system comprising the compound is placed in close proximity to the tissue infected with a virus to be prevented, treated and/or managed.
  • the close proximity of the controlled-release system to the infection may result in only a fraction of the dose of the compound required if it is systemically administered.
  • a compound described herein may be preferable to administer a compound described herein via the natural route of infection of the influenza virus against which the compound has antiviral activity.
  • Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent for use as a spray.
  • Therapeutic or prophylactic agents that can be used in combination with the compounds described herein for the prevention, treatment and/or management of influenza virus infection include, but are not limited to, small molecules, synthetic drugs, peptides (including cyclic peptides), polypeptides, proteins, nucleic acids (e.g., DNA and RNA nucleotides including, but not limited to, antisense nucleotide sequences, triple helices, RNAi, and nucleotide sequences encoding biologically active proteins, polypeptides or peptides), antibodies, synthetic or natural inorganic molecules, mimetic agents, and synthetic or natural organic molecules.
  • synthetic drugs peptides (including cyclic peptides), polypeptides, proteins, nucleic acids (e.g., DNA and RNA nucleotides including, but not limited to, antisense nucleotide sequences, triple helices, RNAi, and nucleotide sequences encoding biologically active proteins, polypeptides or peptid
  • agents include, but are not limited to, immunomodulatory agents (e.g., interferon), anti-inflammatory agents (e.g., adrenocorticoids, corticosteroids (e.g., beclomethasone, budesonide, flunisolide, fluticasone, triamcinolone, methylprednisolone, prednisolone, prednisone, hydrocortisone),
  • immunomodulatory agents e.g., interferon
  • anti-inflammatory agents e.g., adrenocorticoids, corticosteroids (e.g., beclomethasone, budesonide, flunisolide, fluticasone, triamcinolone, methylprednisolone, prednisolone, prednisone, hydrocortisone
  • anti-inflammatory agents e.g., adrenocorticoids, corticosteroids (e.g., beclomethasone, bude
  • glucocorticoids e.g., aspirin, ibuprofen, diclofenac, and COX-2 inhibitors
  • pain relievers e.g., aspirin, ibuprofen, diclofenac, and COX-2 inhibitors
  • leukotreine antagonists e.g., montelukast, methyl xanthines, zafirlukast, and zileuton
  • beta2-agonists e.g.
  • albuterol e.g., albuterol, biterol, fenoterol, isoetharie, metaproterenol, pirbuterol, salbutamol, terbutalin formoterol, salmeterol, and salbutamol terbutaline
  • anticholinergic agents e.g., ipratropium bromide and oxitropium bromide
  • sulphasalazine penicillamine, dapsone
  • antihistamines e.g., hydroxychloroquine
  • anti-viral agents e.g., nucleoside analogs (e.g., zidovudine, acyclovir, gangcyclovir, vidarabine, idoxuridine, trifluridine, and ribavirin), foscarnet, amantadine, rimantadine, saquinavir, indinavir, ritonavir, and AZT) and antibiotics (e
  • Any therapy which is known to be useful, or which has been used or is currently being used for the prevention, management, and/or treatment of an influenza virus infection or symptom or disease associated therewith can be used in combination with the compounds described herein in the compositions and methods described herein. See, e.g., Gilman et al, Goodman and Gilman's: The Pharmacological Basis of Therapeutics, 10th ed., McGraw-Hill, New York, 2001 ; The Merck Manual of Diagnosis and Therapy, Berkow, M.D. et al.
  • prophylactic or therapeutic agents which have been or are currently being used for preventing, treating and/or managing influenza virus infections.
  • Antiviral agents that can be used in combination with compounds described herein include, but are not limited to, non-nucleoside reverse transcriptase inhibitors, nucleoside reverse transcriptase inhibitors, protease inhibitors, and fusion inhibitors. In one
  • the antiviral agent is selected from the group consisting of amantadine, oseltamivir phosphate, rimantadine, and zanamivir.
  • the antiviral agent is a non-nucleoside reverse transcriptase inhibitor selected from the group consisting of delavirdine, efavirenz, and nevirapine.
  • the antiviral agent is a nucleoside reverse transcriptase inhibitor selected from the group consisting of abacavir, didanosine, emtricitabine, emtricitabine, lamivudine, stavudine, tenofovir DF, zalcitabine, and zidovudine.
  • the antiviral agent is a protease inhibitor selected from the group consisting of amprenavir, atazanavir, fosamprenav, indinavir, lopinavir, nelfinavir, ritonavir, and saquinavir.
  • the antiviral agent is a fusion inhibitor such as enfuvirtide.
  • Other examples of anti-viral agents include but are not limited to acemannan; acyclovir; acyclovir sodium; adefovir; alovudine; alvircept
  • cidofovir cipamfylline
  • cytarabine hydrochloride delavirdine mesylate
  • desciclovir delavirdine mesylate
  • ganciclovir ganciclovir sodium; idoxuridine; kethoxal; lamivudine; lobucavir; memotine hydrochloride; methisazone; nevirapine; oseltamivir phosphate (TAMIFLUTM); penciclovir; pirodavir; ribavirin; rimantadine hydrochloride (FLUMADINETM); saquinavir mesylate; somantadine hydrochloride; sorivudine; statolon; stavudine; tilorone hydrochloride;
  • valacyclovir hydrochloride vidarabine
  • vidarabine phosphate vidarabine sodium phosphate
  • viroxime zalcitabine
  • zanamivir RELENZATM
  • zidovudine and zinviroxime.
  • Antibacterial agents including antibiotics, that can be used in combination with the compounds described herein include, but are not limited to, aminoglycoside antibiotics, glycopeptides, amphenicol antibiotics, ansamycin antibiotics, cephalosporins, cephamycins oxazolidinones, penicillins, quinolones, streptogamins, tetracyclins, and analogs thereof.
  • antibiotics are administered in combination with the compound to prevent and/or treat a bacterial infection.
  • the compounds described herein are used in combination with other protein synthesis inhibitors, including but not limited to,
  • streptomycin streptomycin, neomycin, erythromycin, carbomycin, and spiramycin.
  • the antibacterial agent is selected from the group consisting of ampicillin, amoxicillin, ciprofloxacin, gentamycin, kanamycin, neomycin, penicillin G, streptomycin, sulfanilamide, and vancomycin.
  • the antibacterial agent is selected from the group consisting of azithromycin, cefonicid, cefotetan, cephalothin, cephamycin, chlortetracycline, clarithromycin, clindamycin, cycloserine, dalfopristin, doxycycline, erythromycin, linezolid, mupirocin, oxytetracycline, quinupristin, rifampin, spectinomycin, and trimethoprim.
  • antibacterial agents for use in combination with the compounds described herein include the following: aminoglycoside antibiotics (e.g., apramycin, arbekacin, bambermycins, butirosin, dibekacin, neomycin, neomycin, neomycin, aminoglycoside antibiotics (e.g., apramycin, arbekacin, bambermycins, butirosin, dibekacin, neomycin, neomycin,
  • amphenicol antibiotics e.g., azidamfenicol, chloramphenicol, florfenicol, and
  • thiamphenicol ansamycin antibiotics (e.g., rifamide and rifampin), carbacephems (e.g., loracarbef), carbapenems (e.g., biapenem and imipenem), cephalosporins (e.g., cefaclor, cefadroxil, cefamandole, cefatrizine, cefazedone, cefozopran, cefpimizole, cefpiramide, and cefpirome), cephamycins (e.g., cefbuperazone, cefmetazole, and cefminox), folic acid analogs (e.g., trimethoprim), glycopeptides (e.g., vancomycin), lincosamides (e.g.,
  • macrolides e.g., azithromycin, carbomycin, clarithomycin, dirithromycin, erythromycin, and erythromycin acistrate
  • monobactams e.g., aztreonam, carumonam, and tigemonam
  • nitrofurans e.g., furaltadone, and furazolium chloride
  • oxacephems e.g., flomoxef, and moxalactam
  • oxazolidinones e.g., linezolid
  • penicillins e.g., amdinocillin, amdinocillin pivoxil, amoxicillin, bacampicillin, benzylpenicillinic acid, benzylpenicillin sodium, epicillin, fenbenicillin, floxacillin, penamccillin, penethamate hydriodide, penicillin o benethamine, penicillin
  • Additional examples include cycloserine, mupirocin, tuberin amphomycin, bacitracin, capreomycin, colistin, enduracidin, enviomycin, and 2,4 diaminopyrimidines (e.g., brodimoprim).
  • the amount of a compound described herein, or the amount of a composition comprising a compound described herein, that will be effective in the prevention, treatment and/or management of an influenza virus infection can be determined by standard clinical techniques. In vitro or in vivo assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed will also depend, e.g., on the route of administration, the type of infection, and the seriousness of the infection, and should be decided according to the judgment of the practitioner and each patient's or subject's circumstances.
  • the dosage of a compound described herein is determined by extrapolating from the "no observed adverse effective level" (NOAEL), as determined in animal studies. This extrapolated dosage is useful in determining the maximum
  • the NOAELs can be extrapolated to determine human equivalent dosages (HED).
  • HED is extrapolated from a non-human animal dosage based on the doses that are normalized to body surface area (i.e., mg/m ).
  • the NOAELs are determined in mice, hamsters, rats, ferrets, guinea pigs, rabbits, dogs, primates, primates (monkeys, marmosets, squirrel monkeys, baboons), micropigs or minipigs.
  • a compound described herein or composition thereof is administered at a dose that is lower than the human equivalent dosage (HED) of the NOAEL over a period of 1 week, 2 weeks, 3 weeks, 1 month, 2 months, three months, four months, six months, nine months, 1 year, 2 years, 3 years, 4 years or more.
  • HED human equivalent dosage
  • a dosage regime for a human subject can be extrapolated from animal model studies using the dose at which 10% of the animals die (LDi 0 ).
  • the starting dose of a Phase I clinical trial is based on preclinical testing.
  • a standard measure of toxicity of a drug in preclinical testing is the percentage of animals that die because of treatment. It is well within the skill of the art to correlate the LDio in an animal study with the maximal-tolerated dose (MTD) in humans, adjusted for body surface area, as a basis to extrapolate a starting human dose.
  • MTD maximal-tolerated dose
  • the interrelationship of dosages for one animal model can be converted for use in another animal, including humans, using conversion factors (based on milligrams per meter squared of body surface) as described, e.g., in Freireich et al, Cancer Chemother. Rep., 1966, 50:219-244.
  • Body surface area may be approximately determined from height and weight of the patient. See, e.g., Scientific Tables, Geigy Pharmaceuticals, Ardley, N. Y., 1970, 537.
  • the adjustment for body surface area includes host factors such as, for example, surface area, weight, metabolism, tissue distribution, absorption rate, and excretion rate.
  • the route of administration, excipient usage, and the specific influenza virus or symptom thereof (and/or other disease) to target are also factors to consider.
  • the standard conservative starting dose is about 1/10 the murine LD 10 , although it may be even lower if other species (i.e., dogs) were more sensitive to the compound.
  • the - standard conservative starting dose is about 1/100, 1/95, 1/90, 1/85, 1/80, 1/75, 1/70, 1/65, 1/60, 1/55, 1/50, 1/45, 1/40, 1/35, 1/30, 1/25, 1/20, 1/15, 2/10, 3/10, 4/10, or 5/10 of the murine LDio-
  • a starting dose amount of a compound in a human is lower than the dose extrapolated from animal model studies.
  • a starting dose amount of a compound in a human is higher than the dose extrapolated from animal model studies. It is well within the skill of the art to start doses of the active composition at relatively low levels, and increase or decrease the dosage as necessary to achieve the desired effect with minimal toxicity.
  • Exemplary doses of compounds or compositions described herein include milligram or microgram amounts per kilogram of subject or sample weight (e.g., about 1 microgram per kilogram to about 500 milligrams per kilogram, about 5 micrograms per kilogram to about 100 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram).
  • a daily dose is at least 50 mg, 75 mg, 100 mg, 150 mg, 250 mg, 500 mg, 750 mg, or at least 1 g.
  • the dosage is a unit dose of 5 mg, preferably 10 mg, 50 mg, 100 mg, 150 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400 mg, 500 mg, 550 mg, 600 mg, 650 mg, 700 mg, 750 mg, 800 mg or more.
  • the dosage is a unit dose that ranges from about 5 mg to about 100 mg, about 100 mg to about 200 mg, about 150 mg to about 300 mg, about 150 mg to about 400 mg, 250 mg to about 500 mg, about 500 mg to about 800 mg, about 500 mg to about 1000 mg, or about 5 mg to about 1000 mg.
  • suitable dosage ranges for oral administration are about 0.001 milligram to about 500 milligrams of a compound, per kilogram body weight per day.
  • the oral dose is about 0.01 milligram to about 100 milligrams per kilogram body weight per day, about 0.1 milligram to about 75 milligrams per kilogram body weight per day or about 0.5 milligram to 5 milligrams per kilogram body weight per day.
  • the dosage amounts described herein refer to total amounts administered; that is, if more than one compound is administered, then, in some embodiments, the dosages correspond to the total amount administered.
  • oral compositions contain about 10% to about 95% of a compound described herein by weight.
  • Suitable dosage ranges for intravenous (i.v.) administration are about 0.01 milligram to about 100 milligrams per kilogram body weight per day, about 0.1 milligram to about 35 milligrams per kilogram body weight per day, and about 1 milligram to about 10 milligrams per kilogram body weight per day.
  • suitable dosage ranges for intranasal administration are about 0.01 pg kg body weight per day to about 1 mg/kg body weight per day.
  • Suppositories generally contain about 0.01 milligram to about 50 milligrams of a compound described herein per kilogram body weight per day and comprise active ingredient in the range of about 0.5% to about 10% by weight.
  • a subject is administered one or more doses of a prophylactically or therapeutically effective amount of a compound or a composition described herein, wherein the prophylactically or therapeutically effective amount is not the same for each dose.
  • a subject is administered a compound or a composition described herein in an amount effective to inhibit viral genome replication by at least 20% to 25%, preferably at least 25% to 30%, at least 30% to 35%, at least 35% to 40%, at least 40% to 45%, at least 45% to 50%, at least 50% to 55%, at least 55% to 60%, at least 60% to 65%, at least 65% to 70%, at least 70% to 75%, at least 75% to 80%, or up to at least 85% relative to a negative control as determined using an assay described herein or others known to one of skill in the art.
  • a subject is administered a compound or a composition in an amount effective to inhibit or reduce viral genome replication by at least 20% to 25%, preferably at least 25% to 30%, at least 30% to 35%, at least 35% to 40%, at least 40% to 45%, at least 45% to 50%, at least 50% to 55%, at least 55% to 60%, at least 60% to 65%, at least 65% to 70%, at least 70% to 75%, at least 75% to 80%, or up to at least 85% relative to a negative control as determined using an assay described herein or others known to one of skill in the art.
  • a subject is administered a compound or a composition in an amount effective to inhibit or reduce viral genome replication by at least 1.5 fold, 2 fold, 2.5 fold, 3 fold, 4 fold, 5 fold, 8 fold, 10 fold, 15 fold, 20 fold, or 2 to 5 fold, 2 to 10 fold, 5 to 10 fold, or 5 to 20 fold relative to a negative control as determined using an assay described herein or other known to one of skill in the art.
  • a subject is administered a compound or a composition described herein in an amount effective to inhibit or reduce viral protein synthesis by at least 20% to 25%, preferably at least 25% to 30%, at least 30% to 35%, at least 35% to 40%, at least 40% to 45%, at least 45% to 50%, at least 50% to 55%, at least 55% to 60%, at least 60% to 65%, at least 65% to 70%, at least 70% to 75%, at least 75% to 80%, or up to at least 85% relative to a negative control as determined using an assay described herein or others known to one of skill in the art.
  • a subject is administered a compound or a composition in an amount effective to inhibit or reduce viral protein synthesis by at least 20% to 25%, preferably at least 25% to 30%, at least 30% to 35%, at least 35% to 40%, at least 40% to 45%, at least 45% to 50%, at least 50% to 55%, at least 55% to 60%, at least 60% to 65%, at least 65% to 70%, at least 70% to 75%, at least 75% to 80%, or up to at least 85% relative to a negative control as determined using an assay described herein or others known to one of skill in the art.
  • a subject is administered a compound or a composition in an amount effective to inhibit or reduce viral protein synthesis by at least 1.5 fold, 2 fold, 2.5 fold, 3 fold, 4 fold, 5 fold, 8 fold, 10 fold, 15 fold, 20 fold, or 2 to 5 fold, 2 to 10 fold, 5 to 10 fold, or 5 to 20 fold relative to a negative control as determined using an assay described herein or others known to one of skill in the art.
  • a subject is administered a compound or a composition described herein in an amount effective to inhibit or reduce the spread of virus from a cell, tissue, or organ to another cell, tissue or organ by at least 20% to 25%, preferably at least 25% to 30%, at least 30% to 35%, at least 35% to 40%, at least 40% to 45%, at least 45% to 50%, at least 50% to 55%, at least 55% to 60%, at least 60% to 65%, at least 65% to 70%, at least 70% to 75%, at least 75% to 80%, or up to at least 85% relative to a negative control as determined using an assay described herein or others known to one of skill in the art.
  • a subject is administered a compound or a composition in an amount effective to inhibit or reduce the spread of virus from a cell, tissue or organ to another cell, tissue or organ by at least 1.5 fold, 2 fold, 2.5 fold, 3 fold, 4 fold, 5 fold, 8 fold, 10 fold, 15 fold, 20 fold, or 2 to 5 fold, 2 to 10 fold, 5 to 10 fold, or 5 to 20 fold relative to a negative control as determined using an assay described herein or others known to one of skill in the art.
  • a subject is administered a compound or a composition described herein in an amount effective to inhibit or reduce viral titer by at least 20% to 25%, preferably at least 25%» to 30%, at least 30% to 35%, at least 35% to 40%, at least 40% to 45%, at least 45% to 50%, at least 50% to 55%, at least 55% to 60%, at least 60% to 65%, at least 65% to 70%, at least 70% to 75%, at least 75% to 80%, or up to at least 85% relative to a negative control as determined using an assay described herein or others known to one of skill in the art.
  • a subject is administered a compound or a composition in an amount effective to inhibit or reduce viral titer by at least 1.5 fold, 2 fold, 2.5 fold, 3 fold, 4 fold, 5 fold, 8 fold, 10 fold, 15 fold, 20 fold, or 2 to 5 fold, 2 to 10 fold, 5 to 10 fold, or 5 to 20 fold relative to a negative control as determined using an assay described herein or others known to one of skill in the art.
  • a subject is administered a compound or a composition in an amount effective to inhibit or reduce viral titer by 1 log, 1.5 logs, 2 logs, 2.5 logs, 3 logs, 3.5 logs, 4 logs, 5 logs or more relative to a negative control as determined using an assay described herein or others known to one of skill in the art.
  • a subject is administered a compound or a composition described herein in an amount effective to inhibit or reduce viral replication by at least 20% to 25%, preferably at least 25% to 30%, at least 30% to 35%, at least 35% to 40%, at least 40% to 45%, at least 45% to 50%, at least 50% to 55%, at least 55% to 60%, at least 60% to 65%, at least 65% to 70%, at least 70% to 75%, at least 75% to 80%, or up to at least 85% relative to a negative control as determined using an assay described herein or others known to one of skill in the art.
  • a subject is administered a compound or a composition in an amount effective to inhibit or reduce viral replication by at least 1.5 fold, 2 fold, 2.5 fold, 3 fold, 4 fold, 5 fold, 8 fold, 10 fold, 15 fold, 20 fold, or 2 to 5 fold, 2 to 10 fold, 5 to 10 fold, or 5 to 20 fold relative to a negative control as determined using an assay described herein or others known to one of skill in the art.
  • a subject is administered a compound or a composition described herein in an amount effective to inhibit or reduce viral replication by 1 log, 1.5 logs, 2 logs, 2.5 logs, 3 logs, 3.5 logs, 4 logs, 5 logs or more relative to a negative control as determined using an assay described herein or others known to one of skill in the art.
  • a subject is administered a compound or a composition described herein in an amount effective to inhibit or reduce the ability of the virus to spread to other individuals by at least 20% to 25%, preferably at least 25% to 30%, at least 30% to 35%, at least 35% to 40%, at least 40% to 45%, at least 45% to 50%, at least 50% to 55%, at least 55% to 60%, at least 60% to 65%, at least 65% to 70%, at least 70% to 75%, at least 75% to 80%, or up to at least 85% relative to a negative control as determined using an assay described herein or others known to one of skill in the art.
  • a subject is administered a compound or a composition in an amount effective to inhibit or reduce the ability of the virus to spread to other cells, tissues or organs in the subject by at least 20% to 25%, preferably at least 25% to 30%, at least 30% to 35%, at least 35% to 40%, at least 40% to 45%, at least 45% to 50%, at least 50% to 55%, at least 55% to 60%, at least 60% to 65%, at least 65% to 70%, at least 70% to 75%, at least 75% to 80%, or up to at least 85% relative to a negative control as determined using an assay described herein or others known to one of skill in the art.
  • a dose of a compound or a composition described herein is administered to a subject every day, every other day, every couple of days, every third day, once a week, twice a week, three times a week, or once every two weeks.
  • two, three or four doses of a compound or a composition described herein is administered to a subject every day, every couple of days, every third day, once a week or once every two weeks.
  • a dose(s) of a compound or a composition is administered for 2 days, 3 days, 5 days, 7 days, 14 days, or 21 days.
  • a dose of a compound or a composition described herein is administered for 1 month, 1.5 months, 2 months, 2.5 months, 3 months, 4 months, 5 months, 6 months or more.
  • the dosages of prophylactic or therapeutic agents which have been or are currently used for the prevention, treatment and/or management of an influenza virus infection can be determined using references available to a clinician such as, e.g., the
  • dosages lower than those which have been or are currently being used to prevent, treat and/or manage the infection are utilized in combination with one or more compounds or compositions described herein.
  • safe ranges of doses can be readily determined using references available to clinicians, such as e.g., the
  • siRNA compositions and their subsequent administration is within the skill of those in the art.
  • a subject in need of such treatment is administered a nucleic acid compound in accordance with the embodiments described herein, commonly in a pharmaceutically acceptable carrier, in doses ranging from 0.01 ug to 100 g per kg of body weight depending on the age of the patient and the severity of the disease state being treated.
  • the treatment regimen may last for a period of time which will vary depending upon the nature of the particular disease, its severity and the overall condition of the patient, and may extend from once daily to once every 20 years.
  • the dosage of the compound may either be increased in the event the patient does not respond significantly to current dosage levels, or the dose may be decreased if an alleviation of the symptoms of the disease state is observed, or if the disease state has been ablated.
  • the dosages and regimens provided in Section 5.4.4 above may be adapted for the administration of nucleic acid compounds, such as, e.g., siRNAs.
  • one or more of the compounds described herein are added to cell culture media.
  • compounds that prove too toxic or are not used in subjects are added to cell culture-related products, such as media.
  • the compounds described herein as ingredients in disinfectants and soaps are also provided.
  • kits that can be used in the above methods.
  • the kit comprises a compound described herein contained in an appropriate package.
  • a kit further comprises a negative control and/or a positive control, in an appropriate package(s).
  • the kit further comprises an influenza virus.
  • the kit further comprises a reporter construct, in an appropriate package.
  • the kit contains instructions for use.
  • This example describes the identification of host cell proteins that reduce or inhibit the replication of influenza virus.
  • this example describes a genome- wide RNAi screen for siRNAs that inhibit host factors required for influenza virus replication in human cells, establishes particular classes of host proteins required for influenza virus replication, and demonstrates that siRNA and small molecule inhibition of these classes of proteins reduces influenza virus replication. See Konig et al., Nature, 2010, 463(7282):813- 817, which is incorporated by reference herein in its entirety
  • Renilla luciferase influenza virus The coding region for the viral hemagglutinin (HA) protein was replaced with that of Renilla luciferase and the packaging signals for the HA segment were incorporated, as previously described (Marsh et al., 2007).
  • HA hemagglutinin
  • WSN-Ren virus was generated by reverse genetics in the presence of complementing HA and amplified in HA-expressing MDCK cells (Marsh et al., 2007).
  • Genome-wide RNAi screen Genome-wide libraries comprising 98,737 synthetic siRNAs targeting 19,628 unique human genes were arrayed in 384- well plates (7ng/siRNA) such that each well contained either two (47,560 wells) or one (3,617 wells) unique and identifiable siRNA per gene.
  • the library matrix was introduced into A549 cells through a high throughput transfection process (Chanda et al., 2003; Aza-Blanc et al., 2003) and after 48h the cells were infected with WSN-Ren virus at a multiplicity of infection (MOI) of 0.5.
  • MOI multiplicity of infection
  • EnduRen Live Cell substrate (Promega) was added after 5 hours and relative luminescence for each well was analyzed on a plate reader (Viewlux) at 12h, 24h and 36h after infection.
  • a plate reader Viewlux
  • Cell-titer-glo Promega reagent was added 72h after siRNA transfection. The screens were run minimally in duplicate and analyzed using a scaling methodology that sets the positive control siRNA at an arbitrary value of 0.1 , and the negative control siRNAs at 1.
  • RNAi screen Additional information on RNAi screen.
  • a 384- well plate-based assay was optimized to identify siRNAs that influence infection of human A549 cells by WSN-Ren virus.
  • a toxicity assay was optimized to identify siRNAs that influence cell viability.
  • Genome-wide libraries comprising 98,737 synthetic siRNAs targeting 19,628 unique human genes in total were arrayed in 384- well plates (7ng/siRNA) such that each well contained either two (47,560 wells) or one (3617 wells) unique and identifiable siRNA per gene). On average, there were 3 wells/gene or 6 siRNAs/gene. Each plate also contained the positive and the negative control siRNAs as indicated above.
  • the library matrix was introduced into A549 cells through a high throughput transfection process (Chanda et al., 2003; Aza-Blanc et al., 2003).
  • lpmol siRNA was incubated at RT for 20 min with 50nl RNAimax in 20ul Optimem/well and then transfected into 1500 A549 cells in 10 ul DMEM supplemented with 10% FBS and antibiotics.
  • MOI multiplicity of infection
  • the cells were infected at a multiplicity of infection (MOI) of 0.5 in 10 ul serum-free DMEM containing 0.875 ug/ml final concentration Trypsin (Sigma). After 5 hours, EnduRen Life Cell substrate (Promega) was added at a final concentration of 10 uM in 10 ul serum-free DMEM.
  • Relative luminescence for each well was analyzed on a 384-well plate reader (Viewlux) at 12h, 24h and 36h after infection. The screen was run twice (in independent experiments) to generate duplicate results and statistically analyzed as described below. All steps were performed using a fully integrated high-throughput cellular genomics robotic system (GNF Systems; www. gnfsy stems . com) .
  • a scaling methodology was developed that sets the positive control (siRNA against luciferase) at an arbitrary value of 0.1, and the negative control siRNAs at 1. Luciferase activities of siRNAs targeting host factors are assigned a score based on the distribution of these values.
  • siRNAs targeting host factors were assigned a score based on the distribution of these values.
  • siRNA libraries The following commercially available siRNA libraries were used in this study: the whole-genome library from Qiagen (Druggable Set version 2 (approx. 7000 genes targeted by -28000 siRNA constructs), NM Set version 1 (approx. 10000 genes targeted by -42000 constructs) and XM Set version 1 (approx. 5300 genes targeted by -21000 constructs), the kinome library from Invitrogen (1287 siRNAs targeting 636 genes) and the kinome library from IDT (2176 siRNAs targeting 542 genes).
  • druggable genome library version 1 from Qiagen which is no longer commercially available, was used (approx. 5000 genes targeted by -10000 siRNAs). All target influenza virus host proteins can be found in Table 3.
  • siRNA Screening Data Analyses Screening data were normalized as previously described (Konig et al., 2007). The activity score for each gene in an assay was taken from the most potent siRNA per gene. Each screen was then analyzed using a Redundant siRNA Analysis (RSA) algorithm (Konig et al., 2007) and a p-value was assigned to each gene in a screen.
  • RSA Redundant siRNA Analysis
  • the RSA p-value represents the likelihood of the corresponding siRNA signal distribution to be generated by chance, i.e., the smaller the p-value, the higher the expected confirmation rate (Konig et al., 2007).
  • the minimum score of a gene among the 12h-, 24h-, and 36h- A549 assays was chosen as the A549 activity score for the gene.
  • a hit criterion of an activity score ⁇ 0.4 1 36 genes were defined as primary hits for the A549 influenza virus screen.
  • Ontology-based Pattern Identification Analysis of siRNA Profiles For all genes screened, a data matrix was constructed based on inhibition across several biological assays: three A549 assays were screened at 12h, 24h and 36h.
  • Ontology-based Pattern Identification OPI is an algorithm that has been previously successfully applied to either predict gene functions based on their expression patterns Zhou et al., 2005; Young et al., 2005), or prioritize genes based on their phenotypic patterns (Rines et al., 2008). Here the algorithm was applied to identify gene clusters that not only share similar inhibition patterns, but also show statistical enrichment in certain functional categories.
  • siRNAs for reconfirmation of 624 genes were individually re-arrayed such that each well of a 384- well plate contained a single siRNA (7 ng).
  • 43 scrambled negative controls Konig et al., 2007
  • the influenza virus infection assay was rerun as previously described. Additionally a parallel assay was run to assess potential cellular toxicity induced by the siRNA through addition of CellTiterGlo (Promega) detection reagent 72 hours after transfection.
  • Each siRNA was screened a minimum of three times for each readout, in at least two independent assay runs.
  • Toxicity filtering strategy For the toxic control siRNA - siRPS27a, titration data series were measured for both infectivity assays and toxicity assays in 5 replicates. To estimate experimental noise level, each data series was curve-fitted using the standard sigmoidal model as defined:
  • IC 50 is the siRNA concentration corresponding to the score representing 50% inhibition (around score 0.5)
  • slope is a negative value representing Hill slope
  • score is the measured normalized activity value.
  • Both data points and curve parameters were then linearly transformed so that all curves sit at a bottom value of 0 and a top value of 1. From the residue of the curve fitting, the intrinsic data noise in the experiment can be estimated as ⁇ and ⁇ / for the toxicity assay and the infectivity assay, respectively.
  • the five infectivity curves and five toxicity curves form 25 unbiased toxicity- infectivity (T-I) relationship pairs, i.e.,
  • the next step was to establish the infectivity confidence threshold for any given toxicity score, so that the boundary is below 95% of the possible infectivity scores produced by a random toxic siRNA. If an siRNA of interest produces an infectivity score below the established threshold (lower score means higher activity), the false positive p-value is below 0.05. This can be simulated by using the Gaussian noise term ⁇ ⁇ and ⁇ I in equation (2) and (3).
  • Fig. 4 shows the decision boundary as a curve.
  • a toxicity score is ⁇ 0.34
  • the infectivity score of an siRNA is highly likely to be affected by its toxicity, indicated by the flat horizontal line segment in the upper right corner of the plot.
  • toxicity scores get larger (weak toxicity) and the corresponding infectivity score is sufficiently low (above the curve), the effect of the siRNA on infectivity is most likely to be true.
  • Fig. 2a Genome-wide analysis was followed by bioinformatics analysis (see Methods), followed by reconfirmation analysis. After reconfirmation analysis, genes with at least 2 siRNAs that resulted in a 35% ( ⁇ 2 SD) or greater decrease in influenza virus reporter activity, without concomitant induction of cytotoxicity, were selected, (ii) WT influenza virus multi-cycle growth analyzed by hemagglutination assay: >4 fold reduction of wild-type influenza virus multi-cycle growth using at least 2 siRNAs targeting the same gene, (iii) Viral gene expression analyzed by quantitative RT-PCR of NP and Ml influenza protein: transfection of 1 or more siRNAs per gene resulting in a 35% or greater decrease in influenza virus NP and Ml RNA transcription, (iv) Functional assays employed to characterize several of the host factors (validated in the HA-assay) in entry and post
  • A549 cells, 293 T cells, Vero cells and MDCK cells were maintained in Dulbecco's minimal essential medium containing antibiotics and 10% fetal bovine serum at 37°C and 5% C0 2 .
  • Generation of and maintenance of MDCK cells expressing the HA protein of influenza A/WSN/33 virus was described previously (Marsh et al., 2007).
  • Influenza A virus A/WSN/33 and swine origin influenza A/Netherlands/602/2009 virus (SOIV) were grown in MDCK cells. Virus stocks were titered by plaque assay on MDCK cells. Vesicular stomatitis virus (VSV) was grown and titered in Vero cells. The WSN-Ren virus was grown and titered in MDCK-HA cells.
  • siRNA transfections of A549 cells A549 cells (passage 2-15) were transfected with siRNAs at a concentration of 30 nM in a reverse transfection procedure using
  • RNAiMAX (Invitrogen, Carlsbad, CA). Knockdown was allowed to proceed for 48h before cells were infected or tested in functional assays.
  • siRNA-transfected A549 cells were infected with either influenza A/WSN/33 virus or VSV (MOI of 0.01) or swine origin influenza
  • A/Netherlands/602/2009 virus (SOIV) (MOI of 1) at 48h post siRNA transfection. At 36h post infection supernatants were harvested and virus titers were determined by plaque assay on MDCK cells (for A/WSN/33 and A/Netherlands/602/2009) or on Vero cells for VSV.
  • A549 cells were transfected with siRNAs as described above. At 48h post transfection cells were infected with influenza A/WSN/33 virus at a multiplicity of infection (MOI) of 0.01. At 36h post infection supernatants were harvested and titered by hemagglutination assay (HA assay). In brief, two-fold serial dilutions of the supernatant were incubated with chicken red blood cells at a final concentration of 0.25% for 60 min on ice.
  • MOI multiplicity of infection
  • HA assay hemagglutination assay
  • A549 cells were reverse transfected with siRNAs using Lipofectamine RNAiMAX reagent (Invitrogen) in 96 well plates. Briefly, 2 pmol siRNAs were diluted in 20 ul of Opti-MEM (Invitrogen) and mixed with 200 nl of Lipofectamine RNAiMAX with 20 ul Opti-MEM for 20 min. A549 cells (2 xlO 5 cells/ml) in 60 ul DMEM containing 10% FBS were added to each well.
  • TPCK trypsin 50 ng; 0.9 ug/ml final
  • RNA samples were isolated using RNeasy 96 Total RNA Isolation kit (Qiagen).
  • QuantiTect Reverse Transcription Kit (Qiagen) was used in accordance to the manufacturer's protocol.
  • GAPDH was selected as the endogenous control gene and was amplified using sense primer sequence 5'- GAAGATGGTGATGGGATTTC-3' and antisense primer sequence 5'- GAAGGTGAAGGTCGGAGTC-3'. Primers for analyzing knock-down efficiency of siRNA treatment are shown in Table 2.
  • cDNA samples were amplified under standard thermal cycler protocol (50 °C for 2 minutes, 95 °C for 10 minutes, and 40 cycles of 95 °C for 15 seconds and 60 °C for 1 minute).
  • Relative expression level was calculated using the endogenous control GAPDH. Fold changes were calculated against the median of negative control siRNAs, scramble 177 (5 '-GGTAATTGCGCGTGC AACT-3 '), 1212 (5 '- ATCCGCGCG ATAGT ACGT A-3 '), 6105 (5 '-GTAAGCTCGTGCGACGT AT-3 '), siGL2 (Dharmacon), siGL3 (Dharmacon) and siGFP-22 (Qiagen).
  • Each value for relative expression levels in Tables 7 and 8 and Figures 2b and 3a represent the average of at least two independent results.
  • the relative expression levels in Table 11 are comprised of the average of four replicates.
  • Entry assays Pseudoparticles bearing different viral envelopes were used to elucidate genes involved in influenza virus entry. Specifically, siRNA-transfected A549 cells were incubated with an appropriate dilution of different pseudoparticles for one hour. The inoculum was removed, medium was added back and cells were incubated for 36h. Entry efficiency was measured as the amount of luciferase secreted into the supernatant (Renilla Luciferase Assay System, Promega, Madison, WI). A gene was considered a hit in the pseudoparticle assay if one siRNA reduced luciferase signal by at least 65% compared to a scrambled control siRNA and a second siRNA resulted in a reduction of at least 50%.
  • Pseudoparticles were generated by transfecting 293T cells with plasmids encoding (i) a minimal HIV provirus encoding the Gaussia luciferase reporter gene, (ii) HIV gag-pol, and (iii) a viral envelope protein (WSN-HA/NA, VSV-G or MMLV env) using FuGENE6 (Roche Applied Science, Indianapolis).
  • plasmids encoding (i) a minimal HIV provirus encoding the Gaussia luciferase reporter gene, (ii) HIV gag-pol, and (iii) a viral envelope protein (WSN-HA/NA, VSV-G or MMLV env) using FuGENE6 (Roche Applied Science, Indianapolis).
  • Each data point in Fig. 2b and Fig. 3 a represents the mean of at least 3 replicates.
  • the beta-lactamase-Ml (Bla-Ml) virus-like particle (VLP) assay was performed as follows: Bla-Ml VLPs contain WSN HA, NA, and a Bla-Ml fusion protein, which is packaged as a structural component into the VLP and released upon fusion with the target cell. siRNA-transfected A549 cells were incubated with the Bla-Ml VLPs and centrifuged at 1.5 k rpm, for 90 min at 4°C. The cells were then transferred to 37°C and incubated an additional 3-4 h. To detect beta-lactamase activity by flow cytometry, cells were detached and loaded with CCF2-AM substrate (Invitrogen).
  • Flow cytometry was performed at the Mount Sinai Flow Cytometry Shared Resource Facility on an LSRII flow cytometer (Becton Dickinson, Miami, FL). Samples were gated on live cells and analyzed for their cleavage of CCF2 using FlowJo 8.5.2 software.
  • Influenza mini-genome assay 293T cells were transfected with siRNAs as described for A549 cells. At 48h post transfection, cells were transfected with plasmids encoding the three polymerase subunits and the nucleoprotein of influenza virus A/WSN/33, the reporter construct pPOLI-Luc-RT, encoding firefly luciferase in the negative-sense orientation flanked by the noncoding regions of segment 8 of strain A/WSN/33 (Stertz et al., 2007) as well as the control reporter plasmid pRL-SV40-R wc (Promega, Madison, WI).
  • Fig. 9 Representative images were selected from the 10 images collected in the different controls (Fig. 9). Data shown in Fig. 3a were generated by background subtraction of all values using the scrambled negative control measurements at 0' as a baseline (negative values were set to 0.001), and then scaled such that the negative control 180' value equaled 1.
  • HSP90 Inhibitor CCT018159 (Calbiochem), Podophyllotoxin (MP Biomedicals), FGF/VEGF Receptor Tyrosine Kinase Inhibitor
  • Diphyllin was identified in a high-throughput screen of small molecular weight compounds as having influenza virus inhibitory activity.
  • the screen assay was described previously (Hoffmann et al., 2008) and diphyllin was identified from a library supplied by ChemDiv (San Diego, CA). The screen was performed at the National Screening Laboratory for the Regional Centers of Excellence in Biodefense (NSRB), Harvard Medical School, Boston.
  • Diphyllin and KN-93 were purchased from Sigma and Calbiochem, respectively and dissolved in DMSO. Cellular toxicity was determined by the CellTiterGlo assay
  • siRNA-transfected A549 cells were infected with either influenza A/WSN/33 virus or VSV at a multiplicity of infection (MOI) of 0.01 or swine origin influenza A/Netherlands/602/2009 virus (SOIV) at an MOI of 1 at 48h post siRNA transfection.
  • MOI multiplicity of infection
  • SOIV swine origin influenza A/Netherlands/602/2009 virus
  • At 36h post infection supernatants were harvested and virus titers were determined by plaque assay on MDC cells (for A/WSN/33 and A/Netherlands/602/2009) or on Vero cells for VSV.
  • Each sample in Fig. 3e is represented by at least 3 replicates.
  • RNAi screen with human lung epithelial (A549) cells was performed in order to characterize host cell factors involved in influenza virus . replication in human cells.
  • the coding region for the influenza A WSN/33 virus hemagglutinin (HA) protein was replaced with that of Renilla luciferase (Fig. la) (Marsh et al., 2007). As no HA is produced, this recombinant virus cannot complete its replication cycle.
  • the RNAi screen focused on the cellular requirements for viral entry, uncoating, nuclear import, and viral RNA transcription/translation, but was not expected to identify factors involved in virus assembly, budding or release.
  • FIG. lb An arrayed siRNA library targeting over 19,000 human genes was employed to transfect human A549 cells (Fig. lb). These cells were infected with the modified influenza virus (WSN-Ren), and luciferase readings were taken after 12, 24, and 36h. Data from two independent screens were analyzed using a Redundant siRNA Activity (RSA) and ontology- based analyses (see Methods supra; Konig et al., 2007). Using these methodologies, 295 cellular genes for which at least 2 siRNAs reduced viral infection by 35% or greater ( ⁇ 2 standard deviations from mean of negative controls), without a concomitant induction of significant cellular toxicity (Fig. 4 and Table 3), were confirmed. The majority of the factors identified through this analysis represent host genes that have not previously been implicated in mediating influenza virus replication.
  • RSA Redundant siRNA Activity
  • PP pseudotyped particle
  • WSN-PP infection was reduced in the presence of siRNAs targeting 23 of these genes, including CD81, FGFR4, GSK3B, MAP2K3 and the v-ATPase subunit ATP6V0C (Figs. 2a, 2b, and 7a-c; Table 9). These genes were also required for efficient VSV-G-PP (but not MMLV-PP) infection, suggesting a role in low-pH-dependent virus entry. Importantly, small molecule inhibitors of FGFR4, GSK3B, and v-ATPase activities attenuated replication of WSN virus, further highlighting their importance in influenza virus infection (Figs. 6 and 8a-e).
  • the COPI coat complex is made up of seven subunits. COPI association with endosomes is pH-dependent and coatomer complex is required for the formation of intermediate transport vesicles between the early and late endosomes (Whitney et al., 1995; Aniento et al., 1996). Consistent with this role, depletion of COPG and ARCN1 both blocked WSN-PP infection (Fig. 2b). The requirement for ARCN1 during the influenza virus entry step was further demonstrated using a more direct virus-like particle (VLP) assay (Fig. 2c) (Tscherne et al.), as well as immunolocalization studies (Fig. 2d).
  • VLP virus-like particle
  • NP influenza virus nucleoprotein
  • siRNA-depleted cells after infection with influenza A/WSN/33 virus was monitored (Figs. 3a and 9).
  • cells depleted of CSE1L, PRSS35, F13A1, SF3A1 , CAMK2B, KPNB1, and PPP1R14D showed a significant decrease (p ⁇ 0.01) of nuclear to cytoplasmic ratios of NP protein at 180 min.
  • F13A1 depleting these factors did not inhibit entry by WSN pseudotyped virus or ⁇ -lactamase (Bla)-Ml VLPs (Fig.
  • RNAi-mediated inhibition of CSE1L results in a decrease in nuclear vRNPs typically seen 90 min after infection with influenza virus (Fig. 3b) (Kutay et al., 1997).
  • CSE1L specifically inhibited influenza virus gene expression in a mini-genome replicon assay, indicating that CSE1L activity is required for the nuclear import of vRNPs as well as newly synthesized viral proteins (Fig. 3c; Table 10).
  • CaM kinase II beta CAMK2B
  • CaM kinase II beta CAMK2B
  • Influenza A virus is an RNA virus that encodes up to eleven proteins and this small coding capacity demands that the virus utilize the host cellular machinery for many aspects of its life cycle (Palese & Shaw, 2007).
  • genome-wide RNAi screening using an siRNA library was employed to identify 295 human host cell factors required for early-stage influenza virus replication. Within this group, those involved in kinase-regulated signaling, ubiquitination and phosphatase activity are the most highly enriched.
  • vATPase vacuolar ATPase
  • FGFR fibroblast growth factor receptor
  • GSK3 glycogen synthase kinase 3
  • 10 proteins were confirmed to be involved in post-entry steps of influenza virus replication. These include nuclear import components, proteases, and the calcium/calmodulin-dependent protein kinase (CaM kinase) II beta (CAMK2B). Growth of swine-origin H1N1 influenza virus was also found to be dependent on the identified host factors. Small molecule inhibitors of several of the identified human host cell factors, including vATPase and CAMK2B, were also found to antagonize influenza virus replication.
  • siRNA SCORE evidence score calculated based on siRNA activity
  • RSA_SCORE_LogP evidence score calculated based on Redundant siRNA Analysis (RSA) (see Methods section)
  • SCORE_Network _Direct, SCORE Network lndirect, SCORE MCODE binary evidence scores if respective genes are contained in Network based on direct or indirect interactions or MCODE (Molecular Complex Detection analysis) respectively (1 or 0);
  • SCORE GOEnrich evidence score calculated based on gene ontology enrichment analysis
  • SCORE KnownViralPartners Direct SCORE KnownViralPartners Indirect : Evidence score calculated based on direct and indirect interactions with influenza virus proteins respectively
  • SCORE Druglnformation binary evidence score on known drugs specific for respective gene; Calculations for each evidence score are described in Materials and
  • Table 7 Host proteins confirmed to be required for wild-type influenza virus growth and gene expression.
  • siRNAs The ability of the siRNAs to inhibit viral gene expression was examined using qRT-PCR on NP (column 10) and Ml (column 11) mRNA at 6h post-infection. Controls were set to the value of 1. An average value of column 10 and 11 is shown in column 12. A call for a confirmed gene (average value ⁇ 0.65) is indicated in column 13.
  • Table 9 Evaluation of host factors that regulate influenza virus entry.
  • a subset of siRNAs targeting factors shown to be required for efficient growth of wild-type influenza virus were evaluated for their effects on infection with pseudotyped lentivirus particles. Particles bearing envelopes derived from either influenza virus HA (WSN) (column 5), Vesicular stomatitis virus (VSV)-G protein (column 6) or Murine leukemia virus ( MLV) Envelope (Env) (column 7) were examined. Additionally, the effects of host factor depletion on entry of an influenza virus-like particle (VLP) was also assessed using a b- lactamase (Bla)-Ml assay (see Methods section) (column 8). Identified entry factors are marked with a Y in column 9; ost-entr factors are desi nated in column 10.
  • Table 10 Effects of host factor depletion on expression of an influenza virus mini- genome reporter.
  • 293T cells were transfected with siRNAs targeting the indicated genes and transfected again 48h later with an influenza virus mini-genome reporter construct encoding firefly luciferase and expression plasmids for NP, PBl, PB2, PA.
  • a Renilla luciferase expression construct under the control of an SV40 promoter was co- transfected. The percent firefly (column 5) and Renilla luciferase (column 6) expression relative to the control SCI is shown.
  • siRNA transfected A549 cells were analyzed 54h post transfection by quantitative RT-PCR for the expression levels of 12 host genes found to inhibit WSN and SOIV (swine origin influenza
  • transcriptome a microarray analysis using ontology-based pattern identification. Molecular and biochemical parasitology 143, 67-79 (2005).
  • influenza virus NEP (NS2 protein) mediates the nuclear export of viral ribonucleoproteins. EMBO J ll, 288-296 (1998). [00439] Hoffmann, H.H., Palese, P. & Shaw, M.L. Modulation of influenza virus replication by alteration of sodium ion transport and protein kinase C activity. Antiviral Res 80, 124-134 (2008).
  • NDV Newcastle disease virus
  • a method of inhibiting replication of an influenza virus in a human subject comprising administering to a human subject in need thereof an effective amount of a compound that reduces or inhibits the expression and/or activity of one or more of the following human host cell factors: AKAP13; ARCN; BRWD3; CD81 ; COPG; CTSW; DUSP3; EPHB2; F AMI 35 A; FGFR2; FGFR4; GABBR1 ; GSK3B; ITGA3; JAK2;
  • a method of inhibiting replication of an influenza virus in a human subject comprising administering to a human subject in need thereof an effective amount of a compound that reduces or inhibits the expression and/or activity of one or more of the following human host cell factors: CAMK2B; CSE1L; F13A1 ; KPNB1; MAP3K12;
  • a method of inhibiting replication of an influenza virus in a human subject comprising administering to a human subject in need thereof an effective amount of a compound that reduces or inhibits the expression and/or activity of one or more of the following human host cell factors: ACRC; DTX2; EPS8L3; FPRl ; MAP3K1 1 ; NUP214; PRPH2; RP1 1-45B20.2; STX10; SUM02; TRPV2; or TUBB.
  • a method of inhibiting replication of an influenza virus in a human subject comprising administering to a human subject in need thereof an effective amount of a compound that reduces or inhibits the expression and/or activity of one or more of the following human host cell factors: ANPEP; CAM2KB; FGFR4; FRAP1 (mTOR);
  • GSK3B/CSNK1G2 GSK3B/CSNK1G2; HSP90AA1 ; or TUBB.
  • a method of inhibiting replication of an influenza virus in a human subject comprising administering to a human subject in need thereof an effective amount of Betulinic acid, CCT018159, Diphyllin, KN-93, Podophyllotoxin, or Sirolimus.
  • a method of inhibiting replication of an influenza virus in a human subject comprising administering to a human subject in need thereof a nucleic acid compound that targets a sequence selected from the following:
  • nucleic acid compound is an siRNA comprising the sequence
  • a method of treating or managing an influenza virus infection, or a symptom or disease associated therewith, in a human subject comprising administering to a human subject in need thereof an effective amount of a compound that reduces or inhibits the expression and/or activity of one or more of the following human host cell factors: AKAP13; ARCN; BRWD3; CD81 ; COPG; CTSW; DUSP3; EPHB2; FAM135A; FGFR2; FGFR4; GABBR1 ; GSK3B; ITGA3; JAK2; MAP2K3; NEK6; RAB1 IB; or one or more of the v- ATPase subunits, ATP6V0B, ATP6V0C, ATP6V1A, ATP6V1B2, or ATP6AP1.
  • a method of treating or managing an influenza virus infection, or a symptom or disease associated therewith, in a human subject comprising administering to a human subject in need thereof an effective amount of a compound that reduces or inhibits the expression and/or activity of one or more of the following human host cell factors:
  • CAMK2B CSE1L; F13A1 ; KPNB1 ; MAP3K12; PP1R14D; PRSS35; RPS10; SF3A1 ; or SUM04.
  • a method of treating or managing an influenza virus infection, or a symptom or disease associated therewith, in a human subject comprising administering to a human subject in need thereof an effective amount of a compound that reduces or inhibits the expression and/or activity of one or more of the following human host cell factors: ACRC; DTX2; EPS8L3; FPR1 ; MAP3K1 1 ; NUP214; PRPH2; RP1 1-45B20.2; STX10; SUM02; TRPV2; or TUBB.
  • a method of treating or managing an influenza virus infection, or a symptom or disease associated therewith, in a human subject comprising administering to a human subject in need thereof an effective amount of a compound that reduces or inhibits the expression and/or activity of one or more of the following human host cell factors: ANPEP; CAM2KB; FGFR4; FRAP1 (mTOR); GSK3B/CSNK1G2; HSP90AA1; or TUBB.
  • a method of treating or managing an influenza virus infection, or a symptom or disease associated therewith, in a human subject comprising administering to a human subject in need thereof an effective amount of Betulinic acid, CCT018159, Diphyllin, KN-93, Podophyllotoxin, or Sirolimus.
  • a method of treating or managing an influenza virus infection, or a symptom or disease associated therewith, in a human subject comprising administering to a human subject in need thereof a nucleic acid compound that targets a sequence selected from the following: or wherein nucleic acid compound is an siRNA comprising the sequence
  • a method of preventing a symptom or disease associated with an influenza virus infection in a human subject comprising administering to a human subject in need thereof an effective amount of a compound that reduces or inhibits the expression and/or activity of one or more of the following human host cell factors: AKAP13; ARCN; BRWD3; CD81; COPG; CTSW; DUSP3; EPHB2; F AMI 35 A; FGFR2; FGFR4; GABBRl ; GSK3B; ITGA3; JAK2; MAP2K3; NEK6; RAB1 IB; or one or more of the v-ATPase subunits, ATP6V0B, ATP6V0C, ATP6V1A, ATP6V1B2, or ATP6AP1.
  • a method of preventing a symptom or disease associated with an influenza virus infection in a human subject comprising administering to a human subject in need thereof an effective amount of a compound that reduces or inhibits the expression and/or activity of one or more of the following human host cell factors: CAMK2B; CSEIL; F13A1; PNB1 ; MAP3K12; PP1R14D; PRSS35; RPS 10; SF3A1 ; or SUM04.
  • a method of preventing a symptom or disease associated with an influenza virus infection in a human subject comprising administering to a human subject in need thereof an effective amount of a compound that reduces or inhibits the expression and/or activity of one or more of the following human host cell factors: ACRC; DTX2; EPS8L3; FPR1; MAP3K11; NUP214; PRPH2; RP11-45B20.2; STX10; SUM02; TRPV2; or TUBB.
  • a method of preventing a symptom or disease associated with an influenza virus infection in a human subject comprising administering to a human subject in need thereof an effective amount of a compound that reduces or inhibits the expression and/or activity of one or more of the following human host cell factors: ANPEP; CAM2KB;
  • FGFR4 FGFR4
  • FRAP1 mTOR
  • GSK3B/CSNK1G2 GSK3B/CSNK1G2
  • HSP90AA1 HSP90AA1
  • TUBB TUBB
  • a method of preventing a symptom or disease associated with an influenza virus infection in a human subject comprising administering to a human subject in need thereof an effective amount of Betulinic acid, CCT018159, Diphyllin, KN-93,
  • Podophyllotoxin or Sirolimus.
  • a method of preventing a symptom or disease associated with an influenza virus infection in a human subject comprising administering to a human subject in need thereof a nucleic acid compound that targets a sequence selected from the following:
  • nucleic acid compound is an siRNA comprising the sequence
  • nucleic acid compound that targets the sequence is an siRNA.
  • influenza virus is an influenza A virus.
  • influenza A virus is an HlNl virus.

Abstract

Cette demande concerne la modulation de facteurs cellulaires de l'hôte nécessaires pour la réplication du virus de la grippe. La demande concerne des composés, notamment des composés d'acide nucléique (comme, par exemple, des petits ARN interférents (siARN)) et des petites molécules, qui ciblent les facteurs cellulaires des hôtes humains impliqués dans la réplication du virus de la grippe, et l'utilisation de ces composés pour moduler la réplication du virus de la grippe et comme agents antiviraux. La demande concerne également des procédés de traitement d'une infection par le virus de la grippe et des procédés de traitement ou de prévention d'un symptôme ou d'une maladie associé à une infection par le virus de la grippe, comprenant l'administration à un sujet d'une composition comprenant un composé, comme un composé d'acide nucléique (par exemple un siARN) ou une petite molécule, qui cible un facteur cellulaire d'hôte humain impliqué dans la réplication du virus de la grippe.
PCT/US2010/003138 2009-12-11 2010-12-10 Compositions et procédés pour inhiber des facteurs cellulaires d'hôtes humains nécessaires pour la réplication du virus de la grippe WO2011071535A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/514,783 US9238815B2 (en) 2009-12-11 2010-12-10 Compositions and methods for inhibiting human host factors required for influenza virus replication

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US28595109P 2009-12-11 2009-12-11
US61/285,951 2009-12-11

Publications (2)

Publication Number Publication Date
WO2011071535A2 true WO2011071535A2 (fr) 2011-06-16
WO2011071535A3 WO2011071535A3 (fr) 2011-08-04

Family

ID=44146099

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/003138 WO2011071535A2 (fr) 2009-12-11 2010-12-10 Compositions et procédés pour inhiber des facteurs cellulaires d'hôtes humains nécessaires pour la réplication du virus de la grippe

Country Status (2)

Country Link
US (1) US9238815B2 (fr)
WO (1) WO2011071535A2 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9238815B2 (en) 2009-12-11 2016-01-19 Icahn School of Medicine at Mounta Sinai Compositions and methods for inhibiting human host factors required for influenza virus replication

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105126080A (zh) * 2015-09-29 2015-12-09 武汉大学 锌指蛋白436(znf436)在治疗心肌肥厚中的应用
SG11201913654QA (en) * 2017-11-15 2020-01-30 10X Genomics Inc Functionalized gel beads
CN111363017B (zh) * 2018-12-25 2022-03-15 中国科学院微生物研究所 A型流感病毒核蛋白s69位突变序列及其突变体与应用
CN109771432B (zh) * 2019-03-13 2022-04-19 中国人民解放军军事科学院军事医学研究院 氨基葡萄糖及其衍生物作为抗病毒药物的应用
CN113502298A (zh) * 2021-06-21 2021-10-15 武汉轻工大学 非受体酪氨酸激酶tnk2蛋白在防治流感病毒感染中的应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008043561A2 (fr) * 2006-10-11 2008-04-17 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Cibles de la grippe
US20090068257A1 (en) * 2006-03-07 2009-03-12 Jean-Claude Leunis Betulonic and betulinic acid derivatives

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6063383A (en) * 1999-01-28 2000-05-16 Hsu; Wu-Ching Pharmaceutical suppository composites for fever and influenza and method of producing the composites
US8142819B2 (en) * 2002-10-21 2012-03-27 Metaproteomics, Llc Synergistic compositions that treat or inhibit pathological conditions associated with inflammatory response
US8222271B2 (en) * 2006-03-23 2012-07-17 Santen Pharmaceutical Co., Ltd. Formulations and methods for vascular permeability-related diseases or conditions
US8629283B2 (en) 2008-03-06 2014-01-14 Icahn School Of Medicine At Mount Sinai Compounds that modulate negative-sense, single-stranded RNA virus replication and uses thereof
US9238815B2 (en) 2009-12-11 2016-01-19 Icahn School of Medicine at Mounta Sinai Compositions and methods for inhibiting human host factors required for influenza virus replication
WO2011150413A1 (fr) 2010-05-28 2011-12-01 Mount Sinai School Of Medicine Composés antiviraux et leurs utilisations

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090068257A1 (en) * 2006-03-07 2009-03-12 Jean-Claude Leunis Betulonic and betulinic acid derivatives
WO2008043561A2 (fr) * 2006-10-11 2008-04-17 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Cibles de la grippe

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9238815B2 (en) 2009-12-11 2016-01-19 Icahn School of Medicine at Mounta Sinai Compositions and methods for inhibiting human host factors required for influenza virus replication

Also Published As

Publication number Publication date
US20130090367A1 (en) 2013-04-11
WO2011071535A3 (fr) 2011-08-04
US9238815B2 (en) 2016-01-19

Similar Documents

Publication Publication Date Title
US20180185374A1 (en) Inhibitors of mtor kinase as anti-viral agents
AU2008287542B2 (en) Treatment of viral infections by modulation of host cell metabolic pathways
Barik New treatments for influenza
US9168269B2 (en) Inhibitors of long and very long chain fatty acid metabolism as broad spectrum anti-virals
Ohol et al. Direct inhibition of cellular fatty acid synthase impairs replication of respiratory syncytial virus and other respiratory viruses
US8629283B2 (en) Compounds that modulate negative-sense, single-stranded RNA virus replication and uses thereof
WO2011071535A2 (fr) Compositions et procédés pour inhiber des facteurs cellulaires d'hôtes humains nécessaires pour la réplication du virus de la grippe
WO2012139028A2 (fr) Polythérapie antivirale
US20130090371A1 (en) Methods and compositions for inhibition of beta2-adrenergic receptor degradation
Zhang et al. p-STAT1 regulates the influenza A virus replication and inflammatory response in vitro and vivo
US20240116889A1 (en) Antiviral compounds and methods
US20210145795A1 (en) Soce facilitators for use in treating or preventing viral infections
US20140121237A1 (en) Methods for Inhibiting Virus Replication
US20110033498A1 (en) METHODS OF PREVENTING AND TREATING VIRAL INFECTIONS BY INHIBITING THE DelSGYLATION ACTIVITY OF OTU DOMAIN-CONTAINING VIRAL PROTEINS
Wu et al. The host-targeting compound peruvoside has a broad-spectrum antiviral activity against positive-sense RNA viruses
US20230002774A1 (en) Methods and compostions for inhibiting coronaviral replication
Muñoz-Torrero López-Ibarra Recent advances in pharmaceutical sciences I
WO2011079272A1 (fr) Composés pour la modulation du basculement entre réplication et transcription des génomes viraux d'orthomyxovirus et leurs méthodes d'utilisation
Tan Effects of Specific Host Factors and MicroRNAs During Infective and Regenerative Phases of Influenza Pneumonia
Class et al. Patent application title: TREATMENT OF VIRAL INFECTIONS BY MODULATION OF HOST CELL METABOLIC PATHWAYS Inventors: Josh Munger (Rochester, NY, US) Bryson Bennett (Metuchen, NJ, US) Thomas Shenk (Princeton, NJ, US) Thomas Shenk (Princeton, NJ, US) Joshua Rabinowitz (Princeton, NJ, US) Assignees: The Trustees of Princeton University
Müller et al. Journal of Antivirals & Antiretrovirals

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: 10836319

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2012543084

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: JP

WWE Wipo information: entry into national phase

Ref document number: 13514783

Country of ref document: US

122 Ep: pct application non-entry in european phase

Ref document number: 10836319

Country of ref document: EP

Kind code of ref document: A2