Compounds with anti-influenza activity: present and future of strategies for the optimal treatment and management of influenza. Part II: Future compounds against influenza virus

R Gasparini, D Amicizia, P L Lai, N L Bragazzi, D Panatto, R Gasparini, D Amicizia, P L Lai, N L Bragazzi, D Panatto

Abstract

In the first part of this overview, we described the life cycle of the influenza virus and the pharmacological action of the currently available drugs. This second part provides an overview of the molecular mechanisms and targets of still-experimental drugs for the treatment and management of influenza. Briefly, we can distinguish between compounds with anti-influenza activity that target influenza virus proteins or genes, and molecules that target host components that are essential for viral replication and propagation. These latter compounds have been developed quite recently. Among the first group, we will focus especially on hemagglutinin, M2 channel and neuraminidase inhibitors. The second group of compounds may pave the way for personalized treatment and influenza management. Combination therapies are also discussed. In recent decades, few antiviral molecules against influenza virus infections have been available; this has conditioned their use during human and animal outbreaks. Indeed, during seasonal and pandemic outbreaks, antiviral drugs have usually been administered in mono-therapy and, sometimes, in an uncontrolled manner to farm animals. This has led to the emergence of viral strains displaying resistance, especially to compounds of the amantadane family. For this reason, it is particularly important to develop new antiviral drugs against influenza viruses. Indeed, although vaccination is the most powerful means of mitigating the effects of influenza epidemics, antiviral drugs can be very useful, particularly in delaying the spread of new pandemic viruses, thereby enabling manufacturers to prepare large quantities of pandemic vaccine. In addition, antiviral drugs are particularly valuable in complicated cases of influenza, especially in hospitalized patients. To write this overview, we mined various databases, including Embase, PubChem, DrugBank and Chemical Abstracts Service, and patent repositories.

Figures

Fig. 1.
Fig. 1.
The attack points of several antiviral drugs are shown, with a particular focus on future potential compounds and strategies against influenza virus. Abbreviations: HA: hemagglutinin; M: Matrix protein; M1: Matrix type 1 protein; M2: Matrix type 2 protein; MTOC: microtubule- organizing center; NA: neuraminidase; NAIs: neuraminidase inhibitors; NEP: nuclear export protein; REs: recycling endosomes; RNA: ribonucleic acid; RNP: ribonucleoprotein; siRNA: short interfering RNA; TBHQ: Tert-butyl-hydroquinone.

References

    1. Gasparini R, Amicizia D, Lai PL, et al. Compounds with antiinfluenza activity: present and future of strategies for the optimal treatment and management of influenza. Part I: influenza life-cycle and currently available drugs. J Prev Med Hyg. 2014;55:69–85.
    1. Müller KH, Kakkola L, Nagaraj AS, et al. Emerging cellular targets for influenza antiviral agents. Trends Pharmacol Sci. 2012;33:89–99.
    1. Planz O. Development of cellular signaling pathway inhibitors as new antivirals against influenza. Antiviral Res. 2013;98:457–468.
    1. Embase. Accessible at embase (last accessed: 16/07/2014).
    1. PubChem. Accessible at (last accessed: 16/07/2014).
    1. PubChem. Accessible at (last accessed: 16/09/2014).
    1. DrugBank. Accessible at (last accessed: 16/07/2014).
    1. Chemical Abstracts Service. Accessible at (last accessed: 16/07/2014).
    1. Clinical trials registries. Accessible at (last accessed: 16/07/2014).
    1. Eyer L, Hruska K. Antiviral agents targeting the influenza virus: a review and publication analysis. Veterinarni Medicina. 2013;58:113–185.
    1. CIRI-IT. Accessible at (last accessed: 16/09/2014).
    1. Pu JY, He L, Wu SY, et al. Anti-virus research of triterpenoids in licorice. Bing Du Xue Bao. 2013;29:673–679.
    1. Jia W, Wang C, Wang Y, et al. Qualitative and quantitative analysis of the major constituents in chinese medical preparation Lianhua-Qingwen capsule by UPLC-DAD-QTOF-MS. Scientific World Journal. 2015;2015:731–765.
    1. Utsunomiya T, Kobayashi M, Pollard RB, et al. Glycyrrhizin, an active component of licorice roots, reduces morbidity and mortality of mice infected with lethal doses of influenza virus. Antimicrob Agents Chemother. 1997;41:551–556.
    1. Fiore C, Eisenhut M, Krausse R, et al. Antiviral effects of Glycyrrhiza species. Phytother Res. 2008;22:141–148.
    1. Harada S. The broad anti-viral agent glycyrrhizin directly modulates the fluidity of plasma membrane and HIV-1 envelope. Biochem J. 2005;392:191–199.
    1. Smirnov VS, Garshinina AV, Guseva VM, et al. The anti-viral activity of the complex glycyrrhizic acid-alpha-glutamyl-tryptophan against experimental lethal influenza infection in white mice caused by oseltamivir-resistant strain of the virus. Vopr Virusol. 2013;58:19–26.
    1. Smirnov VS, Zarubaev VV, Anfimov PM, et al. Effect of a combination of glutamyl-tryptophan and glycyrrhizic acid on the course of acute infection caused by influenza (H3H2) virus in mice. Vopr Virusol. 2012;57:23–27.
    1. Wolkerstorfer A, Kurz H, Bachhofner N, et al. Glycyrrhizin inhibits influenza A virus uptake into the cell. Antiviral Res. 2009;83:171–178.
    1. Michaelis M, Geiler J, Naczk P, et al. Glycyrrhizin inhibits highly pathogenic H5N1 influenza A virus-induced pro-inflammatory cytokine and chemokine expression in human macrophages. Med Microbiol Immunol. 2010;199:291–297.
    1. Michaelis M, Geiler J, Naczk P, et al. Glycyrrhizin exerts antioxidative effects in H5N1 influenza A virus-infected cells and inhibits virus replication and pro-inflammatory gene expression. PLoS One. 2011;6:e19705–e19705.
    1. Moisy D, Avilov SV, Jacob Y, et al. HMGB1 protein binds to influenza virus nucleoprotein and promotes viral replication. J Virol. 2012;86:9122–9133.
    1. Stanetty C, Wolkerstorfer A, Amer H, et al. Synthesis and antiviral activities of spacer-linked 1-thioglucuronide analogues of glycyrrhizin. Beilstein J Org Chem. 2012;8:705–711.
    1. Pompei R, Paghi L, Ingianni A, et al. Glycyrrhizic acid inhibits influenza virus growth in embryonated eggs. Microbiologica. 1983;6:247–250.
    1. Scherließ R, Ajmera A, Dennis M, et al. Induction of protective immunity against H1N1 influenza A(H1N1)pdm09 with spraydried and electron-beam sterilised vaccines in non-human primates. Vaccine. 2014;32:2231–2240.
    1. Song G, Yang S, Zhang W, et al. Discovery of the first series of small molecule H5N1 entry inhibitors. J Med Chem. 2009;52:7368–7371.
    1. Song W, Si L, Ji S, et al. Uralsaponins M-Y, antiviral triterpenoid saponins from the roots of Glycyrrhiza uralensis. J Nat Prod. 2014;77:1632–1643.
    1. Song X, Chen J, Sakwiwatkul K, et al. Enhancement of immune responses to influenza vaccine (H3N2) by ginsenoside Re. Int Immunopharmacol. 2010;10:351–356.
    1. Barr IG, Mitchell GF. ISCOMs (immunostimulating complexes): the first decade. Immunol Cell Biol. 1996;74:8–25.
    1. Liu H, Patil HP, Vries-Idema J, et al. Enhancement of the immunogenicity and protective efficacy of a mucosal influenza subunit vaccine by the saponin adjuvant GPI-0100. PLoS One. 2012;7:e52135–e52135.
    1. Liu H, Vries-Idema J, Ter Veer W, et al. Influenza virosomes supplemented with GPI-0100 adjuvant: a potent vaccine formulation for antigen dose sparing. Med Microbiol Immunol. 2014;203:47–55.
    1. Zhai L, Li Y, Wang W, et al. Enhancement of humoral immune responses to inactivated Newcastle disease and avian influenza vaccines by oral administration of ginseng stem-and-leaf saponins in chickens. Poult Sci. 2011;90:1955–1959.
    1. Sun H, He S, Shi M. Adjuvant-active fraction from Albizia julibrissin saponins improves immune responses by inducing cytokine and chemokine at the site of injection. Int Immunopharmacol. 2014;22:346–355.
    1. Kazakova OB, Giniyatullina GV, Yamansarov EY, et al. Betulin and ursolic acid synthetic derivatives as inhibitors of Papilloma virus. Bioorg Med Chem Lett. 2010;20:4088–4090.
    1. Kazakova OB, Medvedeva NI, Baĭkova IP, et al. Synthesis of triterpenoid acylates - an effective reproduction inhibitors of influenza A (H1N1) and papilloma viruses. Bioorg Khim. 2010;36:841–848.
    1. Flekhter OB, Medvedeva NI, Kukovinets OS, et al. Synthesis and antiviral activity of lupane triterpenoids with modified cycle E. Bioorg Khim. 2007;33:629–634.
    1. Baltina LA, Flekhter OB, Nigmatullina LR, et al. Lupane triterpenes and derivatives with antiviral activity. Bioorg Med Chem Lett. 2003;13:3549–3552.
    1. Grishko VV, Galaiko NV, Tolmacheva IA, et al. Functionalization, cyclization and antiviral activity of A-secotriterpenoids. Eur J Med Chem. 2014;83:601–608.
    1. Krumbiegel M, Dimitrov DS, Puri A, et al. Dextran sulfate inhibits fusion of influenza virus and cells expressing influenza hemagglutinin with red blood cells. Biochim Biophys Acta. 1992;1110:158–164.
    1. Herrmann A, Korte T, Arnold K, et al. The influence of dextran sulfate on influenza A virus fusion with erythrocyte membranes. Antiviral Res. 1992;19:295–311.
    1. Lüscher-Mattli M, Glück R, Kempf C, et al. A comparative study of the effect of dextran sulfate on the fusion and the in vitro replication of influenza A and B, Semliki Forest, vesicular stomatitis, rabies, Sendai, and mumps virus. Arch Virol. 1993;130:317–326.
    1. Yamada H, Moriishi E, Haredy AM, et al. Influenza virus neuraminidase contributes to the dextran sulfate-dependent suppressive replication of some influenza A virus strains. Antiviral Res. 2012;96:344–352.
    1. Yamada H, Nagao C, Haredy AM, et al. Dextran sulfate-resistant A/Puerto Rico/8/34 influenza virus is associated with the emergence of specific mutations in the neuraminidase glycoprotein. Antiviral Res. 2014;111:69–77.
    1. Shkurupy VA, Potapova OV, Sharkova TV, et al. Experimental study of the efficiency of oxidized dextran for prevention of influenza A/H5N1. Bull Exp Biol Med. 2014;158:112–114.
    1. Shkurupy VA, Potapova OV, Sharkova TV, et al. Effects of Preventive Administration of Oxidized Dextran on Liver Injury and Reparative Regeneration in Mice Infected with Influenza A/ H5N1 Virus. Bull Exp Biol Med. 2015;158:483–488.
    1. Potapova OV, Shkurupiy VA, Sharkova TV, et al. Preventive efficacy of oxidized dextran and pathomorphological processes in mouse lungs in avian influenza A/H5N1. Bull Exp Biol Med. 2011;150:707–710.
    1. Clercq E. Highlights in the development of new antiviral agents. Mini Rev Med Chem. 2002;2:163–175.
    1. Bond S, Draffan AG, Fenner JE, et al. 1,2,3,9b-Tetrahydro-5Himidazo, 1-a]isoindol-5-ones as a new class of respiratory syncytial virus (RSV) fusion inhibitors. Part 2: identification of BTA9881 as a preclinical candidate. Bioorg Med Chem Lett. 2015;25:976–981.
    1. Weisman LE. Respiratory syncytial virus (RSV) prevention and treatment: past, present, and future. Cardiovasc Hematol Agents Med Chem. 2009;7:223–233.
    1. Guinea R, Carrasco L. Concanamycin A blocks influenza virus entry into cells under acidic conditions. FEBS Lett. 1994;349:327–330.
    1. Guinea R, Carrasco L. Concanamycin A: a powerful inhibitor of enveloped animal-virus entry into cells. Biochem Biophys Res Commun. 1994;201:1270–1278.
    1. Guinea R, Carrasco L. Requirement for vacuolar proton-ATPase activity during entry of influenza virus into cells. J Virol. 1995;69:2306–2312.
    1. Müller KH, Kainov DE, El Bakkouri K, et al. The proton translocation domain of cellular vacuolar ATPase provides a target for the treatment of influenza A virus infections. Br J Pharmacol. 2011;164:344–357.
    1. Yeganeh B, Ghavami S, Kroeker AL, et al. Suppression of influenza A virus replication in human lung epithelial cells by noncytotoxic concentrations bafilomycin A1. Am J Physiol Lung Cell Mol Physiol. 2015;308:L270–L286.
    1. Ochiai H, Sakai S, Hirabayashi T, et al. Inhibitory effect of bafilomycin A1, a specific inhibitor of vacuolar-type proton pump, on the growth of influenza A and B viruses in MDCK cells. Antiviral Res. 1995;27:425–430.
    1. Bimbo LM, Denisova OV, Mäkilä E, et al. Inhibition of influenza A virus infection in vitro by saliphenylhalamide-loaded porous silicon nanoparticles. ACS Nano. 2013;7:6884–6893.
    1. Paton NI, Lee L, Xu Y, et al. Chloroquine for influenza prevention: a randomised, double-blind, placebo controlled trial. Lancet Infect Dis. 2011;11:677–683.
    1. Savarino A. Use of chloroquine in viral diseases. Lancet Infect Dis. 2011;11:653–654.
    1. Ooi EE, Chew JS, Loh JP, et al. In vitro inhibition of human influenza A virus replication by chloroquine. Virol J. 2006;3:39–39.
    1. Trani L, Savarino A, Campitelli L, et al. Different pH requirements are associated with divergent inhibitory effects of chloroquine on human and avian influenza A viruses. Virol J. 2007;4:39–39.
    1. Vigerust DJ, McCullers JA. Chloroquine is effective against influenza A virus in vitro but not in vivo. Influenza Other Respir Viruses. 2007;1:189–192.
    1. Yan Y, Zou Z, Sun Y, et al. Anti-malaria drug chloroquine is highly effective in treating avian influenza A H5N1 virus infection in an animal model. Cell Res. 2013;23:300–302.
    1. Garulli B, Mario G, Sciaraffia E, et al. Enhancement of T cell-mediated immune responses to whole inactivated influenza virus by chloroquine treatment in vivo. Vaccine. 2013;31:1717–1724.
    1. Wu L, Dai J, Zhao X, et al. Chloroquine enhances replication of influenza A virus A/WSN/33 (H1N1) in dose-, time-, and MOIdependent manners in human lung epithelial cells A549. J Med Virol. 2015 in press.
    1. Clercq E. A Cutting-edge view on the current state of antiviral drug development. Med Res Rev. 2013;33:1249–1277.
    1. Blaising J, Lévy PL, Gondeau C, et al. Silibinin inhibits hepatitis C virus entry into hepatocytes by hindering clathrin-dependent trafficking. Cell Microbiol. 2013;15:1866–1882.
    1. Dai JP, Wu LQ, Li R, et al. Identification of 23-(s)-2-amino- 3-phenylpropanoyl-silybin as an antiviral agent for influenza A virus infection in vitro and in vivo. Antimicrob Agents Chemother. 2013;57:4433–4443.
    1. Gazák R, Purchartová K, Marhol P, et al. Antioxidant and antiviral activities of silybin fatty acid conjugates. Eur J Med Chem. 2010;45:1059–1067.
    1. Garozzo A, Timpanaro R, Bisignano B, et al. In vitro antiviral activity of Melaleuca alternifolia essential oil. Lett Appl Microbiol. 2009;49:806–808.
    1. Garozzo A, Timpanaro R, Stivala A, et al. Activity of Melaleuca alternifolia (tea tree) oil on Influenza virus A/PR/8: study on the mechanism of action. Antiviral Res. 2011;89:83–88.
    1. Li X, Duan S, Chu C, et al. Melaleuca alternifolia concentrate inhibits in vitro entry of influenza virus into host cells. Molecules. 2013;18:9550–9566.
    1. Mantil E, Daly G, Avis TJ. Effect of tea tree (Melaleuca alternifolia) oil as a natural antimicrobial agent in lipophilic formulations. Can J Microbiol. 2015;61:82–88.
    1. He J, Qi WB, Wang L, et al. Amaryllidaceae alkaloids inhibit nuclear-to-cytoplasmic export of ribonucleoprotein (RNP) complex of highly pathogenic avian influenza virus H5N1. Influenza Other Respir Viruses. 2013;7:922–931.
    1. Aggarwal BB, Deb L, Prasad S. Curcumin differs from tetrahydrocurcumin for molecular targets, signaling pathways and cellular responses. Molecules. 2014;20:185–205.
    1. Chen TY, Chen DY, Wen HW, et al. Inhibition of enveloped viruses infectivity by curcumin. PLoS One. 2013;8:e62482–e62482.
    1. Shuto T. Regulation of expression, function, and inflammatory responses of innate immune receptor Toll-like receptor-2 (TLR2) during inflammatory responses against infection. Yakugaku Zasshi. 2013;133:1401–1409.
    1. Kim K, Kim KH, Kim HY, et al. Curcumin inhibits hepatitis C virus replication via suppressing the Akt-SREBP-1 pathway. FEBS Lett. 2010;584:707–712.
    1. Rajput N, Naeem M, Ali S, et al. The effect of dietary supplementation with the natural carotenoids curcumin and lutein on broiler pigmentation and immunity. Poult Sci. 2013;92:1177–1185.
    1. Ou JL, Mizushina Y, Wang SY, et al. Structure-activity relationship analysis of curcumin analogues on anti-influenza virus activity. FEBS J. 2013;280:5829–5840.
    1. Haasbach E, Hartmayer C, Hettler A, et al. Antiviral activity of Ladania067, an extract from wild black currant leaves against influenza A virus in vitro and in vivo. Front Microbiol. 2014;5:171–171.
    1. Ehrhardt C, Dudek SE, Holzberg M, et al. A plant extract of Ribes nigrum folium possesses anti-influenza virus activity in vitro and in vivo by preventing virus entry to host cells. PLoS One. 2013;8:e63657–e63657.
    1. Yokomizo K, Miyamoto Y, Nagao K, et al. Fattiviracin A1, a novel antiviral agent produced by Streptomyces microflavus strain No. 2445. II. Biological properties. J Antibiot (Tokyo) 1998;51:1035–1039.
    1. Habib ES, Yokomizo K, Nagao K, et al. Antiviral activity of fattiviracin FV-8 against human immunodeficiency virus type 1 (HIV-1) Biosci Biotechnol Biochem. 2001;65:683–685.
    1. Tanaka T, Ikeda T, Kaku M, et al. A new lignan glycoside and phenylethanoid glycosides from Strobilanthes cusia BREMEK. Chem Pharm Bull (Tokyo) 2004;52:1242–1245.
    1. Uyeda M. Metabolites produced by actinomycetes-antiviral antibiotics and enzyme inhibitors. Yakugaku Zasshi. 2004;124:469–479.
    1. Liao Q, Qian Z, Liu R, et al. Germacrone inhibits early stages of influenza virus infection. Antiviral Res. 2013;100:578–588.
    1. Denisova OV, Söderholm S, Virtanen S, et al. Akt inhibitor MK2206 prevents influenza pH1N1 virus infection in vitro. Antimicrob Agents Chemother. 2014;58:3689–3696.
    1. Hirata N, Suizu F, Matsuda-Lennikov M, et al. Inhibition of Akt kinase activity suppresses entryand replication of influenza virus. Biochem Biophys Res Commun. 2014;450:891–898.
    1. Hsieh CF, Lo CW, Liu CH, et al. Mechanism by which ma-xingshi- gan-tang inhibits the entry of influenza virus. J Ethnopharmacol. 2012;143:57–67.
    1. Murray JL, McDonald NJ, Sheng J, et al. Inhibition of influenza A virus replication by antagonism of a PI3K-AKT-mTOR pathway member identified by gene-trap insertional mutagenesis. Antivir Chem Chemother. 2012;22:205–215.
    1. Chan RW, Chan MC, Wong AC, et al. DAS181 inhibits H5N1 influenza virus infection of human lung tissues. Antimicrob Agents Chemother. 2009;53:3935–3941.
    1. Triana-Baltzer GB, Gubareva LV, Nicholls JM, et al. Novel pandemic influenza A(H1N1) viruses are potently inhibited by DAS181, a sialidase fusion protein. PLoS One. 2009;4:e7788–e7788.
    1. Triana-Baltzer GB, Gubareva LV, Klimov AI, et al. Inhibition of neuraminidase inhibitor-resistant influenza virus by DAS181, a novel sialidase fusion protein. PLoS One. 2009;4:e7838–e7838.
    1. Triana-Baltzer GB, Babizki M, Chan MC, et al. DAS181, a sialidase fusion protein, protects human airway epithelium against influenza virus infection: an in vitro pharmacodynamic analysis. J Antimicrob Chemother. 2010;65:275–284.
    1. Moss RB, Hansen C, Sanders RL, et al. A phase II study of DAS181, a novel host directed antiviral for the treatment of influenza infection. J Infect Dis. 2012;206:1844–1851.
    1. Ison MG. Expanding the armamentarium against respiratory viral infections: DAS181. J Infect Dis. 2012;206:1806–1808.
    1. Zhang H. DAS181 and H5N1 virus infection. J Infect Dis. 2009;199:1250–1250. author reply 1250-1.
    1. Moscona A, Porotto M, Palmer S, et al. A recombinant sialidase fusion protein effectively inhibits human parainfluenza viral infection in vitro and in vivo. J Infect Dis. 2010;202:234–241.
    1. Jones BG, Hayden RT, Hurwitz JL. Inhibition of primary clinical isolates of human parainfluenza virus by DAS181 in cell culture and in a cotton rat model. Antiviral Res. 2013;100:562–566.
    1. Malakhov MP, Aschenbrenner LM, Smee DF, et al. Sialidase fusion protein as a novel broad-spectrum inhibitor of influenza virus infection. Antimicrob Agents Chemother. 2006;50:1470–1479.
    1. Hayden F. Developing new antiviral agents for influenza treatment: what does the future hold? Clin Infect Dis. 2009;48(Suppl 1):S3–S13.
    1. Belser JA, Lu X, Szretter KJ, et al. DAS181, a novel sialidase fusion protein, protects mice from lethal avian influenza H5N1 virus infection. J Infect Dis. 2007;196:1493–1499.
    1. Marjuki H, Mishin VP, Chesnokov AP, et al. An investigational antiviral drug, DAS181, effectively inhibits replication of zoonotic influenza A virus subtype H7N9 and protects mice from lethality. J Infect Dis. 2014;210:435–440.
    1. Zhu L, Li Y, Li S, et al. Inhibition of influenza A virus (H1N1) fusion by benzenesulfonamide derivatives targeting viral hemagglutinin. PLoS One. 2011;6:e29120–e29120.
    1. Hsieh HP, Hsu JT. Strategies of development of antiviral agents directed against influenza virus replication. Curr Pharm Des. 2007;13:3531–3542.
    1. Shigeta S. Current status of research and development for anti-influenza virus drugs--chemotherapy for influenza. Nihon Rinsho. 1997;55:2758–2764.
    1. Luo G, Torri A, Harte WE, et al. Molecular mechanism underlying the action of a novel fusion inhibitor of influenza A virus. J Virol. 1997;71:4062–4070.
    1. Vichkanova SA, Oĭfa AI, Goriunova LV. Antiviral properties of gossypol in experimental influenza pneumonia. Antibiotiki. 1970;15:1071–1073.
    1. Krylov VF. Treatment of patients with influenza. Ter Arkh. 1975;47:49–55.
    1. Yang J, Zhang F, Li J, et al. Synthesis and antiviral activities of novel gossypol derivatives. Bioorg Med Chem Lett. 2012;22:1415–1420.
    1. Yang J, Chen G, Li LL, et al. Synthesis and anti-H5N1 activity of chiral gossypol derivatives and its analogs implicated by a viral entry blocking mechanism. Bioorg Med Chem Lett. 2013;23:2619–2623.
    1. Jones JC, Turpin EA, Bultmann H, et al. Inhibition of influenza virus infection by a novel antiviral peptide that targets viral attachment to cells. J Virol. 2006;80:11960–11967.
    1. Altmann SE, Brandt CR, Jahrling PB, et al. Antiviral activity of the EB peptide against zoonotic poxviruses. Virol J. 2012;9:6–6.
    1. Nicol MQ, Ligertwood Y, Bacon MN, et al. A novel family of peptides with potent activity against influenza A viruses. J Gen Virol. 2012;93:980–986.
    1. Rajik M, Jahanshiri F, Omar AR, et al. Identification and characterisation of a novel anti-viral peptide against avian influenza virus H9N2. Virol J. 2009;6:74–74.
    1. Matsubara T. Potential of peptides as inhibitors and mimotopes: selection of carbohydrate-mimetic peptides from phage display libraries. J Nucleic Acids. 2012;2012:740982–740982.
    1. Selman L, Hansen S. Structure and function of collectin liver 1 (CL-L1) and collectin 11 (CL-11, CL-K1) Immunobiology. 2012;217:851–863.
    1. Ling MT, Tu W, Han Y, et al. Mannose-binding lectin contributes to deleterious inflammatory response in pandemic H1N1 and avian H9N2 infection. J Infect Dis. 2012;205:44–53.
    1. Dec M, Wernicki A. Conglutinin, CL-43 and CL-46-three bovine collectins. Pol J Vet Sci. 2006;9:265–275.
    1. Kawai T, Kase T, Suzuki Y, et al. Anti-influenza A virus activities of mannan-binding lectins and bovine conglutinin. J Vet Med Sci. 2007;69:221–224.
    1. Malhotra R, Haurum JS, Thiel S, et al. Binding of human collectins (SP-A and MBP) to influenza virus. Biochem J. 1994;304:455–461.
    1. Hartshorn KL, Sastry K, Brown D, et al. Conglutinin acts as an opsonin for influenza A viruses. J Immunol. 1993;151:6265–6273.
    1. Verma A, White M, Vathipadiekal V, et al. Human H-ficolin inhibits replication of seasonal and pandemic influenza A viruses. J Immunol. 2012;189:2478–2487.
    1. Pan Q, Chen H, Wang F, et al. L-ficolin binds to the glycoproteins hemagglutinin and neuraminidase and inhibits influenza A virus infection both in vitro and in vivo. J Innate Immun. 2012;4:312–324.
    1. Chang WC, Hartshorn KL, White MR, et al. Recombinant chimeric lectins consisting of mannose-binding lectin and L-ficolin are potent inhibitors of influenza A virus compared with mannose- binding lectin. Biochem Pharmacol. 2011;81:388–395.
    1. Gordts SC, Renders M, Férir G, et al. NICTABA and UDA, two GlcNAc-binding lectins with unique antiviral activity profiles. J Antimicrob Chemother. 2015 in press.
    1. Pustylnikov S, Sagar D, Jain P, et al. Targeting the C-type lectins- mediated host-pathogen interactions with dextran. J Pharm Pharm Sci. 2014;17:371–392.
    1. Morita M, Kuba K, Ichikawa A, et al. The lipid mediator protectin D1 inhibits influenza virus replication and improves severe influenza. Cell. 2013;153:112–125.
    1. Zhang C, Xu Y, Jia L, et al. A new therapeutic strategy for lung tissue injury induced by influenza with CR2 targeting complement inhibitor. Virol J. 2010;7:30–30.
    1. Imai Y. Role of omega-3 PUFA-derived mediators, the protectins, in influenza virus infection. Biochim Biophys Acta. 2015;1851:496–502.
    1. Takahashi M, Mori S, Shigeta S, et al. Role of MBL-associated serine protease (MASP) on activation of the lectin complement pathway. Adv Exp Med Biol. 2007;598:93–104.
    1. Smee DF, Bailey KW, Wong MH, et al. Treatment of influenza A (H1N1) virus infections in mice and ferrets with cyanovirin- N. Antiviral Res. 2008;80:266–271.
    1. O'Keefe BR, Smee DF, Turpin JA, et al. Potent anti-influenza activity of cyanovirin-N and interactions with viral hemagglutinin. Antimicrob Agents Chemother. 2003;47:2518–2525.
    1. Miyamoto D, Hasegawa S, Sriwilaijaroen N, et al. Clarithromycin inhibits progeny virus production from human influenza virus-infected host cells. Biol Pharm Bull. 2008;31:217–222.
    1. Yamaya M, Shinya K, Hatachi Y, et al. Clarithromycin inhibits type a seasonal influenza virus infection in human airway epithelial cells. J Pharmacol Exp Ther. 2010;333:81–90.
    1. Ghendon Y, Markushin S, Heider H, et al. Haemagglutinin of influenza A virus is a target for the antiviral effect of Norakin. J Gen Virol. 1986;67:1115–1122.
    1. Ott S, Wunderli-Allenspach H. Effect of the virostatic Norakin (triperiden) on influenza virus activities. Antiviral Res. 1994;24:37–42.
    1. Presber HW, Schroeder C, Hegenscheid B, et al. Antiviral activity of Norakin (triperiden) and related anticholinergic antiparkinsonism drugs. Acta Virol. 1984;28:501–507.
    1. Heider H, Markushin S, Schroeder C, Ghendon Y. The influence of Norakin on the reproduction of influenza A and B viruses. Arch Virol. 1985;86:283–290.
    1. Schroeder C, Heider H, Hegenscheid B, et al. The anticholinergic anti-Parkinson drug Norakin selectively inhibits influenza virus replication. Antiviral Res. 1985;(Suppl 1):95–99.
    1. Prösch S, Heider H, Schroeder C, et al. Mutations in the hemagglutinin gene associated with influenza virus resistance to norakin. Arch Virol. 1988;102:125–129.
    1. Prösch S, Heider H, Schroeder C, et al. Mapping mutations in influenza A virus resistant to norakin. FEBS Lett. 1990;267:19–21.
    1. Klimov AI, Markushin SG, Prösch S, et al. Relation between drug resistance and antigenicity among norakin-resistant mutants of influenza A (fowl plague) virus. Arch Virol. 1992;124:147–155.
    1. Markushin SG, Ginzburg VP, Khaĭder AM, et al. Factors that cause a change in the antigenic structure of of the influenza virus hemagglutinin. Vopr Virusol. 1992;37:196–199.
    1. Oka M, Ishiwata Y, Iwata N, et al. Synthesis and anti-influenza virus activity of tricyclic compounds with a unique amine moiety. Chem Pharm Bull (Tokyo) 2001;49:379–383.
    1. Rossignol JF. Nitazoxanide. A first-in-class broad-spectrum antiviral agent. Antiviral Res. 2014 in press.
    1. Ashiru O, Howe JD, Butters TD. Nitazoxanide, an antiviral thiazolide, depletes ATP-sensitive intracellular Ca(2+) stores. Virology. 2014;462-463:135–148.
    1. Belardo G, Cenciarelli O, La Frazia S, et al. Synergistic effect of nitazoxanide with neuraminidase inhibitors against influenza A viruses in vitro. Antimicrob Agents Chemother. 2015;59:1061–1069.
    1. Täubel J, Lorch U, Rossignol JF, et al. Analyzing the relationship of QT interval and exposure to nitazoxanide, a prospective candidate for influenza antiviral therapy – A formal TQT study. J Clin Pharmacol. 2014;54:987–994.
    1. Haffizulla J, Hartman A, Hoppers M, et al. Effect of nitazoxanide in adults and adolescents with acute uncomplicated influenza: a double-blind, randomised, placebo-controlled, phase 2b/3 trial. Lancet Infect Dis. 2014;14:609–618.
    1. Ashton LV, Callan RL, Rao S, et al. In vitro susceptibility of canine influenza A (H3N8) virus to nitazoxanide and tizoxanide. Vet Med Int. 2010;2010
    1. Ulyanova V, Vershinina V, Ilinskaya O. Barnase and binase: twins with distinct fates. FEBS J. 2011;278:3633–3643.
    1. Shah Mahmud R, Ilinskaya ON. Antiviral Activity of Binase against the Pandemic Influenza A (H1N1) Virus. Acta Naturae. 2013;5:44–51.
    1. Sato Y, Hirayama M, Morimoto K, et al. High mannose-binding lectin with preference for the cluster of alpha1-2-mannose from the green alga Boodlea coacta is a potent entry inhibitor of HIV-1 and influenza viruses. J Biol Chem. 2011;286:19446–19458.
    1. Savov VM, Galabov AS, Tantcheva LP, et al. Effects of rutin and quercetin on monooxygenase activities in experimental influenza virus infection. Exp Toxicol Pathol. 2006;58:59–64.
    1. Davis JM, Murphy EA, McClellan JL, et al. Quercetin reduces susceptibility to influenza infection following stressful exercise. Am J Physiol Regul Integr Comp Physiol. 2008;295:R505–R509.
    1. Choi HJ, Song JH, Park KS, et al. Inhibitory effects of quercetin 3-rhamnoside on influenza A virus replication. Eur J Pharm Sci. 2009;37:329–333.
    1. Kumar P, Khanna M, Srivastava V, et al. Effect of quercetin supplementation on lung antioxidants after experimental influenza virus infection. Exp Lung Res. 2005;31:449–459.
    1. Raju TA, Lakshmi AN, Anand T, et al. Protective effects of quercetin during influenza virus-induced oxidative stress. Asia Pac J Clin Nutr. 2000;9:314–317.
    1. Friel H, Lederman H. A nutritional supplement formula for influenza A (H5N1) infection in humans. Med Hypotheses. 2006;67:578–587.
    1. Eşanu V, Prahoveanu E, Crişan I, et al. The effect of an aqueous propolis extract, of rutin and of a rutin-quercetin mixture on experimental influenza virus infection in mice. Virologie. 1981;32:213–215.
    1. Chang SS, Huang HJ, Chen CY. Two birds with one stone? Possible dual-targeting H1N1 inhibitors from traditional Chinese medicine. PLoS Comput Biol. 2011;7:e1002315–e1002315.
    1. Chang TT, Sun MF, Chen HY, et al. Screening from the world�s largest TCM database against H1N1 virus. J Biomol Struct Dyn. 2011;28:773–786.
    1. Nakayama M, Suzuki K, Toda M, et al. Inhibition of the infectivity of influenza virus by tea polyphenols. Antiviral Res. 1993;21:289–299.
    1. Yang ZF, Bai LP, Huang WB, et al. Comparison of in vitro antiviral activity of tea polyphenols against influenza A and B viruses and structure-activity relationship analysis. Fitoterapia. 2014;93:47–53.
    1. Zu M, Yang F, Zhou W, et al. In vitro anti-influenza virus and anti-inflammatory activities of theaflavin derivatives. Antiviral Res. 2012;94:217–224.
    1. Hayden FG, Aoki FY. Amantadine, rimatadine, and related agents. In: Barriere SL, editor. Antimicrobial Therapy and Vaccines. Baltimore: Williams & Williams; 1999. pp. 1344–1365.
    1. Wang C, Takeuchi K, Pinto LH, et al. Ion channel activity of influenza A virus M2 protein: characterization of the amantadine block. J Virol. 1993:675585–675594.
    1. Ruigrok RW, Hirst EM, Hay AJ. The specific inhibition of influenza A virus maturation by amantadine: an electron microscopic examination. J Gen Virol. 1991;72:191–194.
    1. Sheu TG, Fry AM, Garten RJ, et al. Dual resistance to adamantanes and oseltamivir among seasonal influenza A(H1N1) viruses: 2008-2010. J Infect Dis. 2011;203:13–17.
    1. Smirnova TD, Danilenko DM, Eropkin MIu, et al. Influence of rimantadine, ribavirine and triazavirine on influenza A virus replication in human monolayer and lymphoblastoid cell lines. Antibiot Khimioter. 2011;56:11–16.
    1. Karpenko I, Deev S, Kiselev O, et al. Antiviral properties, metabolism, and pharmacokinetics of a novel azolo-1,2,4-triazinederived inhibitor of influenza A and B virus replication. Antimicrob Agents Chemother. 2010;54:2017–2022.
    1. Tanner JA, Zheng BJ, Zhou J, et al. The adamantane-derived bananins are potent inhibitors of the helicase activities and replication of SARS coronavirus. Chem Biol. 2005;12:303–311.
    1. Moorthy NS, Poongavanam V, Pratheepa V. Viral M2 ion channel protein: a promising target for anti-influenza drug discovery. Mini Rev Med Chem. 2014;14:819–830.
    1. Rey-Carrizo M, Torres E, Ma C, et al. 3-Azatetracyclo. 2.1.1(5,8).0(1,5)]undecane derivatives: from wild-type inhibitors of the M2 ion channel of influenza A virus to derivatives with potent activity against the V27A mutant. J Med Chem. 2013;56:9265–9274.
    1. Wang J, Wu Y, Ma C, et al. Structure and inhibition of the drugresistant S31N mutant of the M2 ion channel of influenza A virus. Proc Natl Acad Sci U S A. 2013;110:1315–1320.
    1. Wang J, Ma C, Wang J, et al. Discovery of novel dual inhibitors of the wild-type and the most prevalent drug-resistant mutant, S31N, of the M2 proton channel from influenza A virus. J Med Chem. 2013;56:2804–2812.
    1. Gasparini R, Lai PL, Casabona F, et al. Do the omeprazole family compounds exert a protective effect against influenza-like illness? BMC Infect Dis. 2014;14:297–297.
    1. Bozdaganyan M, Orekhov P, Bragazzi NL, et al. Docking and molecular dynamics (MD) simulations in potential drugs discovery: an application to influenza virus M2 protein. Am J Biochem Biotechnol. 2014;10:180–188.
    1. 3C9J. The Crystal structure of Transmembrane domain of M2 protein and Amantadine complex. Accesible at .
    1. Long J, Wright E, Molesti E, et al. Antiviral therapies against Ebola and other emerging viral diseases using existing medicines that block virus entry. F1000Research. 2015;4:30–30.
    1. Bachrach U, Don S. Inactivation of influenza and Newcastle disease viruses by oxidized spermine. Isr J Med Sci. 1970;6:435–437.
    1. Bachrach U. Antiviral activity of oxidized polyamines. Amino Acids. 2007;33:267–272.
    1. Lin TI, Heider H, Schroeder C. Different modes of inhibition by adamantane amine derivatives and natural polyamines of the functionally reconstituted influenza virus M2 proton channel protein. J Gen Virol. 1997;78:767–774.
    1. Even-Or O, Samira S, Rochlin E, et al. Immunogenicity, protective efficacy and mechanism of novel CCS adjuvanted influenza vaccine. Vaccine. 2010;28:6527–6541.
    1. Even-Or O, Joseph A, Itskovitz-Cooper N, et al. A new intranasal influenza vaccine based on a novel polycationic lipid-ceramide carbamoyl-spermine (CCS). II. Studies in mice and ferrets and mechanism of adjuvanticity. Vaccine. 2011;29:2474–2486.
    1. Fytas C, Kolocouris A, Fytas G, et al. Influence of an additional amino group on the potency of aminoadamantanes against influenza virus A. II - Synthesis of spiropiperazines and in vitro activity against influenza A H3N2 virus. Bioorg Chem. 2010;38:247–251.
    1. Zhao X, Jie Y, Rosenberg MR, et al. Design and synthesis of pinanamine derivatives as anti-influenza A M2 ion channel inhibitors. Antiviral Res. 2012;96:91–99.
    1. Serkedjieva J, Manolova N, Bankova V. Anti-influenza virus effect of some propolis constituents and their analogues (esters of substituted cinnamic acids) J Nat Prod. 1992;55:294–302.
    1. Kesel AJ. Synthesis of novel test compounds for antiviral chemotherapy of severe acute respiratory syndrome (SARS) Curr Med Chem. 2005;12:2095–2162.
    1. Iwai A, Shiozaki T, Miyazaki T. Relevance of signaling molecules for apoptosis induction on influenza A virus replication. Biochem Biophys Res Commun. 2013;441:531–537.
    1. Jurgeit A, McDowell R, Moese S, et al. Niclosamide is a proton carrier and targets acidic endosomes with broad antiviral effects. PLoS Pathog. 2012;8:e1002976–e1002976.
    1. Krátký M, Vinšová J. Antiviral activity of substituted salicylanilides – a review. Mini Rev Med Chem. 2011;11:956–967.
    1. Russell RJ, Kerry PS, Stevens DJ, et al. Structure of influenza hemagglutinin in complex with an inhibitor of membrane fusion. Proc Natl Acad Sci U S A. 2008;105:17736–17741.
    1. Hosoya M, Matsuyama S, Baba M, et al. Effects of protease inhibitors on replication of various myxoviruses. Antimicrob Agents Chemother. 1992;36:1432–1436.
    1. Bahgat MM, Blazejewska P, Schughart K. Inhibition of lung serine proteases in mice: a potentially new approach to control influenza infection. Virol J. 2011;8:27–27.
    1. Hamilton BS, Chung C, Cyphers SY, et al. Inhibition of influenza virus infection and hemagglutinin cleavage by the protease inhibitor HAI-2. Biochem Biophys Res Commun. 2014;450:1070–1075.
    1. Lee MG, Kim KH, Park KY, et al. Evaluation of anti-influenza effects of camostat in mice infected with non-adapted human influenza viruses. Arch Virol. 1996;141:1979–1989.
    1. Puzis LE, Lozitsky VP. Action of epsilon-aminocaproic acid on the proteolysis system during experimental influenza in mice. Acta Virol. 1988;32:515–521.
    1. Tashiro M, Klenk HD, Rott R. Inhibitory effect of a protease inhibitor, leupeptin, on the development of influenza pneumonia, mediated by concomitant bacteria. J Gen Virol. 1987;68:2039–2041.
    1. Zhirnov OP, Klenk HD, Wright PF. Aprotinin and similar protease inhibitors as drugs against influenza. Antiviral Res. 2011;92:27–36.
    1. Zhou Y, Wu C, Zhao L, et al. Exploring the early stages of the pH-induced conformational change of influenza hemagglutinin. Proteins. 2014;82:2412–2428.
    1. Leikina E, Delanoe-Ayari H, Melikov K, et al. Carbohydratebinding molecules inhibit viral fusion and entry by crosslinking membrane glycoproteins. Nat Immunol. 2005;6:995–1001.
    1. Streeter DG, Witkowski JT, Khare GP, et al. Mechanism of action of 1-β-D-ribofuranosyl-1,2,4-triazole-3-carboxamide (Virazole), a new broad-spectrum antiviral agent. Proc Natl Acad Sci U S A. 1973;70:1174–1178.
    1. Crotty S, Cameron C, Andino R. Ribavirin's antiviral mechanism of action: lethal mutagenesis? J Mol Med (Berl) 2002;80:86–95.
    1. Chan-Tack KM, Murray JS, Birnkrant DB. Use of ribavirin to treat influenza. N Engl J Med. 2009;361:1713–1714.
    1. Gangemi JD, Nachtigal M, Barnhart D, et al. Therapeutic efficacy of liposome-encapsulated ribavirin and muramyl tripeptide in experimental infection with influenza or herpes simplex virus. J Infect Dis. 1987;155:510–517.
    1. Smee DF, Hurst BL, Day CW, et al. Influenza Virus H1N1 inhibition by serine protease inhibitor (serpin) antithrombin III. Int Trends Immun. 2014;2:83–86.
    1. Stoller JK, Lacbawan FL, Aboussouan LS. Alpha-1 Antitrypsin Deficiency. In: Pagon RA, Adam MP, Ardinger HH, et al., editors. GeneReviews ® [Internet] Seattle: University of Washington; 1993-2014. 2006 Oct 27 [updated 2014 May 01]
    1. Campos MA, Alazemi S, Zhang G, et al. Influenza vaccination in subjects with alpha1-antitrypsin deficiency. Chest. 2008;133:49–55.
    1. Yagi S, Ono J, Yoshimoto J, et al. Development of anti-influenza virus drugs I: improvement of oral absorption and in vivo anti-influenza activity of Stachyflin and its derivatives. Pharm Res. 1999;16:1041–1046.
    1. Yoshimoto J, Yagi S, Ono J, et al. Development of anti-influenza drugs: II. Improvement of oral and intranasal absorption and the anti-influenza activity of stachyflin derivatives. J Pharm Pharmacol. 2000;52:1247–1255.
    1. Minagawa K, Kouzuki S, Yoshimoto J, et al. Stachyflin and acetylstachyflin, novel anti-influenza A virus substances, produced by Stachybotrys sp. RF-7260. I. Isolation, structure elucidation and biological activities. J Antibiot (Tokyo) 2002;55:155–164.
    1. Minagawa K, Kouzuki S, Kamigauchi T. Stachyflin and acetylstachyflin, novel anti-influenza A virus substances, produced by Stachybotrys sp. RF-7260. II. Synthesis and preliminary structure- activity relationships of stachyflin derivatives. J Antibiot (Tokyo) 2002;55:165–171.
    1. Motohashi Y, Igarashi M, Okamatsu M, et al. Antiviral activity of stachyflin on influenza A viruses of different hemagglutinin subtypes. Virol J. 2013;10:118–118.
    1. Watanabe K, Sakurai J, Abe H, et al. Total synthesis of (+)-stachyflin: a potential anti-influenza A virus agent. Chem Commun (Camb) 2010;46:4055–4057.
    1. Nakatani M, Nakamura M, Suzuki A, et al. A new strategy toward the total synthesis of stachyflin, a potent anti-influenza A virus agent: concise route to the tetracyclic core structure. Org Lett. 2002;4:4483–4486.
    1. Combrink KD, Gulgeze HB, Yu KL, et al. Salicylamide inhibitors of influenza virus fusion. Bioorg Med Chem Lett. 2000;10:1649–1652.
    1. Zhu L, Li Y, Li S, et al. Inhibition of influenza A virus (H1N1) fusion by benzenesulfonamide derivatives targeting viral hemagglutinin. PLoS One. 2011;6:e29120–e29120.
    1. Yu KL, Torri AF, Luo G, et al. Structure-activity relationships for a series of thiobenzamide influenza fusion inhibitors derived from 1,3,3-trimethyl-5-hydroxy-cyclohexylmethylamine. Bioorg Med Chem Lett. 2002;12:3379–3382.
    1. Yuan S. Drugs to cure avian influenza infection-multiple ways to prevent cell death. Cell Death Dis. 2013;4:e835–e835.
    1. Ye M, Zheng JB, Yu KJ, et al. Effects of high dose ulinastatin treatment in patients with severe pneumonia complicating influenza A H1N1 infection. Zhongguo Wei Zhong Bing Ji Jiu Yi Xue. 2011;23:48–49.
    1. Naganuma A, Mizuma H, Doi I, et al. A case of acute respiratory distress syndrome induced by fulminant influenza A (H3 N2) pneumonia. Nihon Kokyuki Gakkai Zasshi. 2000;38:783–787.
    1. Munakata M, Kato R, Yokoyama H, et al. Combined therapy with hypothermia and anticytokine agents in influenza A encephalopathy. Brain Dev. 2000;22:373–377.
    1. Leng YX, Yang SG, Song YH, et al. Ulinastatin for acute lung injury and acute respiratory distress syndrome: A systematic review and meta-analysis. World J Crit Care Med. 2014;3:34–41.
    1. Ketscher L, Hannß R, Morales DJ, et al. Selective inactivation of USP18 isopeptidase activity in vivo enhances ISG15 conjugation and viral resistance. Proc Natl Acad Sci U S A. 2015;112:1577–1582.
    1. Uchida Y, Watanabe C, Takemae N, et al. Identification of host genes linked with the survivability of chickens infected with recombinant viruses possessing H5N1 surface antigens from a highly pathogenic avian influenza virus. J Virol. 2012;86:2686–2695.
    1. Liu AL, Li YF, Qi W, et al. Comparative analysis of selected innate immune-related genes following infection of immortal DF-1 cells with highly pathogenic (H5N1) and low pathogenic (H9N2) avian influenza viruses. Virus Genes. 2015 in press.
    1. Loregian A, Mercorelli B, Nannetti G, et al. Antiviral strategies against influenza virus: towards new therapeutic approaches. Cell Mol Life Sci. 2014 in press.
    1. Bauman JD, Patel D, Baker SF, et al. Crystallographic fragment screening and structure-based optimization yields a new class of influenza endonuclease inhibitors. ACS Chem Biol. 2013;8:2501–2508.
    1. Sheppard S. Moroxydine: the story of a mislaid antiviral. Acta Derm Venereol Suppl (Stockh) 1994;183:1–9.
    1. Mertens T, Eggers HJ. Moroxydine. Dtsch Med Wochenschr. 1980;105:184–184.
    1. Dreyfus P. Treatment of influenzal infections by a moroxydine derivative. Sem Ther. 1966;42:51–52.
    1. Furuta Y, Takahashi K, Fukuda Y, et al. In vitro and in vivo activities of anti-influenza virus compound T-705. Antimicrob Agents Chemother. 2002;46:977–981.
    1. Caroline AL, Powell DS, Bethel LM, et al. Broad spectrum antiviral activity of favipiravir (T-705): protection from highly lethal inhalational Rift Valley Fever. PLoS Negl Trop Dis. 2014;8:e2790–e2790.
    1. Oestereich L, Rieger T, Neumann M, et al. Evaluation of antiviral efficacy of ribavirin, arbidol, and T-705 (favipiravir) in a mouse model for Crimean-Congo hemorrhagic fever. PLoS Negl Trop Dis. 2014;8:e2804–e2804.
    1. Smither SJ, Eastaugh LS, Steward JA, et al. Post-exposure efficacy of oral T-705 (Favipiravir) against inhalational Ebola virus infection in a mouse model. Antiviral Res. 2014;104:153–155.
    1. Safronetz D, Falzarano D, Scott DP, et al. Antiviral efficacy of favipiravir against two prominent etiological agents of hantavirus pulmonary syndrome. Antimicrob Agents Chemother. 2013;57:4673–4680.
    1. Iwai Y, Murakami K, Gomi Y, et al. Anti-influenza activity of marchantins, macrocyclic bisbibenzyls contained in liverworts. PLoS One. 2011;6:e19825–e19825.
    1. Shaw ML, Klumpp K. Successes and challenges in the antiviral field. Curr Opin Virol. 2013;3:483–486.
    1. Loregian A, Coen DM. Selective anti-cytomegalovirus compounds discovered by screening for inhibitors of subunit interactions of the viral polymerase. Chem Biol. 2006;13:191–200.
    1. Fukuoka M, Minakuchi M, Kawaguchi A, et al. Structure-based discovery of anti-influenza virus A compounds among medicines. Biochim Biophys Acta. 2012;1820:90–95.
    1. Sugiyama K, Obayashi E, Kawaguchi A, et al. Structural insight into the essential PB1-PB2 subunit contact of the influenza virus RNA polymerase. EMBO J. 2009;28:1803–1811.
    1. Chase G, Wunderlich K, Reuther P, et al. Identification of influenza virus inhibitors which disrupt of viral polymerase proteinprotein interactions. Methods. 2011;55:188–191.
    1. Li C, Ba Q, Wu A, et al. A peptide derived from the C-terminus of PB1 inhibits influenza virus replication by interfering with viral polymerase assembly. FEBS J. 2013;280:1139–1149.
    1. Nasser EH, Judd AK, Sanchez A, et al. Antiviral activity of influenza virus M1 zinc finger peptides. J Virol. 1996;70:8639–8644.
    1. Li L, Chang S, Xiang J, et al. Screen anti-influenza lead compounds that target the PA(C) subunit of H5N1 viral RNA polymerase. PLoS One. 2012;7:e35234–e35234.
    1. Clark MP, Ledeboer MW, Davies I, et al. Discovery of a novel, first-in-class, orally bioavailable azaindole inhibitor (VX-787) of influenza PB2. J Med Chem. 2014;57:6668–6678.
    1. Pagano M, Castagnolo D, Bernardini M, et al. The fight against the influenza A virus H1N1: synthesis, molecular modeling, and biological evaluation of benzofurazan derivatives as viral RNA polymerase inhibitors. Chem Med Chem. 2014;9:129–150.
    1. Lepri S, Nannetti G, Muratore G, et al. Optimization of smallmolecule inhibitors of influenza virus polymerase: from thiophene- 3-carboxamide to polyamido scaffolds. J Med Chem. 2014;57:4337–4350.
    1. Gao J, Luo X, Li Y, et al. Synthesis and biological evaluation of 2-oxo-pyrazine-3-carboxamide-yl nucleoside analogues and their epimers as inhibitors of influenza A viruses. Chem Biol Drug Des. 2014 in press.
    1. Dierkes R, Warnking K, Liedmann S, et al. The Rac1 inhibitor NSC23766 exerts anti-influenza virus properties by affecting the viral polymerase complex activity. PLoS One. 2014;9:e88520–e88520.
    1. Elton D, Simpson-Holley M, Archer K, et al. Interaction of the influenza virus nucleoprotein with the cellular CRM1-mediated nuclear export pathway. J Virol. 2001;75:408–419.
    1. Perwitasari O, Johnson S, Yan X, et al. Verdinexor, a novel selective inhibitor of nuclear export, reduces influenza a virus replication in vitro and in vivo. J Virol. 2014;88:10228–10243.
    1. Amorim MJ, Kao RY, Digard P. Nucleozin targets cytoplasmic trafficking of viral ribonucleoprotein-Rab11 complexes in influenza A virus infection. J Virol. 2013;87:4694–4703.
    1. Su CY, Cheng TJ, Lin MI, et al. High-throughput identification of compounds targeting influenza RNA-dependent RNA polymerase activity. Proc Natl Acad Sci USA. 2010;107:19151–19156.
    1. Jiang H, Xu Y, Li L, et al. Inhibition of influenza virus replication by constrained peptides targeting nucleoprotein. Antivir Chem Chemother. 2011;22:119–130.
    1. Verhelst J, Parthoens E, Schepens B, et al. Interferon-inducible protein Mx1 inhibits influenza virus by interfering with functional viral ribonucleoprotein complex assembly. J Virol. 2012;86:13445–13455.
    1. Cianci C, Gerritz SW, Deminie C, et al. Influenza nucleoprotein: promising target for antiviral chemotherapy. Antivir Chem Chemother. 2012;23:77–91.
    1. Pons M. Effect of actinomycin D on the replication of influenza virus and influenza virus RNA. Virology. 1967;33:150–154.
    1. Vogel U, Scholtissek C. Inhibition of the intracellular transport of influenza viral RNA by actinomycin D. Arch Virol. 1995;140:1715–1723.
    1. Pons MW. The inhibition of influenza virus RNA synthesis by actinomycin D and cycloheximide. Virology. 1973;51:120–128.
    1. Lejal N, Tarus B, Bouguyon E, et al. Structure-based discovery of the novel antiviral properties of naproxen against the nucleoprotein of influenza A virus. Antimicrob Agents Chemother. 2013;57:2231–2242.
    1. Zarubaev VV, Beliaevskaia SV, Sirotkin AK, et al. In vitro and in vivo effects of ingavirin on the ultrastructure and infectivity of influenza virus. Vopr Virusol. 2011;56:21–25.
    1. Zarubaev VV, Garshinina AV, Kalinina NA, et al. Activity of Ingavirin (6 -(1H-Imidazol-4-yl)ethylamino]-5-oxohexanoic acid) against human respiratory viruses in vivo experiments. Pharmaceuticals. 2011;4:1518–1534.
    1. Semenova NP, Prokudina EN, Livov DK, et al. Effect of the antiviral drug Ingaviruin on intracellular transformations and import into the nucleus of influenza A virus nucleocapsid protein. Vopr Virusol. 2010;55:17–20.
    1. Loginova SIa, Borisevich SV, Shkliaeva OM, et al. Prophylactic and therapeutic efficacies of Ingavirin, a novel Russian chemotherapeutic, with respect to influenza pathogen A (H5N1) Antibiot Khimioter. 2010;55:10–12.
    1. Shishkina LN, Nebol'sin VE, Skarnovich MO, et al. In vivo efficacy of Ingavirin against pandemic A (H1N1/09)v influenza virus. Antibiot Khimioter. 2010;55:32–35.
    1. Kolobukhina LV, Merkulova LN, Shchelkanov MI, et al. Efficacy of ingavirin in adults with influenza. Ter Arkh. 2009;81:51–54.
    1. Galegov GA, Andronova VL, Nebol'sin VE. Antiviral effect of Ingavirin against seasonal influenza virus A/H1N1 in MDCK cell culture. Antibiot Khimioter. 2009;54:19–22.
    1. Loginova SIa, Borisevich SV, Maksimov VA, et al. Investigation of prophylactic activity of Ingavirin, a new Russian drug, against grippe A virus (H3N2) Antibiot Khimioter. 2008;53:19–21.
    1. Shul'diakov AA, Liapina EP, Kuznetsov VI, et al. Current principles in the chemoprophylaxis of acute respiratory viral infections. Ter Arkh. 2013;85:27–33.
    1. Isaeva EI, Nebol'sin VE, Kozulina IS, et al. In vitro investigation of the antiviral activity of Ingavirin against human metapneumovirus. Vopr Virusol. 2012;57:34–38.
    1. Gubareva LV, Kaiser L, Hayden FG. Influenza virus neuraminidase inhibitors. Lancet. 2000;355:827–835.
    1. Feng E, Ye D, Li J, et al. Recent advances in neuraminidase inhibitor development as anti-influenza drugs. ChemMedChem. 2012;7:1527–1536.
    1. Verma RP, Hansch C. A QSAR study on influenza neuraminidase inhibitors. Bioorg Med Chem. 2006;14:982–996.
    1. Barroso L, Treanor J, Gubareva L, et al. Efficacy and tolerability of the oral neuraminidase inhibitor peramivir in experimental human influenza: randomized, controlled trials for prophylaxis and treatment. Antivir Ther. 2005;10:901–910.
    1. Birnkrant D, Cox E. The Emergency Use Authorization of peramivir for treatment of 2009 H1N1 influenza. N Engl J Med. 2009;361:2204–2207.
    1. Koyama K, Ogura Y, Nakai D, et al. Identification of bioactivating enzymes involved in the hydrolysis of laninamivir octanoate, a long-acting neuraminidase inhibitor, in human pulmonary tissue. Drug Metab Dispos. 2014;42:1031–1038.
    1. Weight AK, Haldar J, Alvarez de Cienfuegos L, et al. Attaching zanamivir to a polymer markedly enhances its activity against drug-resistant strains of influenza a virus. J Pharm Sci. 2011;100:831–835.
    1. Hayden FG, Cote KM, Douglas RG., Jr Plaque inhibition assay for drug susceptibility testing of influenza viruses. Antimicrob Agents Chemother. 1980;17:865–870.
    1. Jedrzejas MJ, Singh S, Brouillette WJ, et al. Structures of aromatic inhibitors of influenza virus neuraminidase. Biochemistry. 1995;34:3144–3151.
    1. Li Y, Silamkoti A, Kolavi G, et al. Pyrrolidinobenzoic acid inhibitors of influenza virus neuraminidase: the hydrophobic side chain influences type A subtype selectivity. Bioorg Med Chem. 2012;20:4582–4589.
    1. Kim CU, Lew W, Williams MA, et al. Structure-activity relationship studies of novel carbocyclic influenza neuraminidase inhibitors. J Med Chem. 1998;41:2451–2460.
    1. Kati WM, Saldivar AS, Mohamadi F, et al. GS4071 is a slowbinding inhibitor of influenza neuraminidase from both A and B strains. Biochem Biophys Res Commun. 1998;244:408–413.
    1. Kim CU, Lew W, Williams MA, et al. Influenza neuraminidase inhibitors possessing a novel hydrophobic interaction in the enzyme active site: design, synthesis, and structural analysis of carbocyclic sialic acid analogues with potent anti-influenza activity. J Am Chem Soc. 1997;119:681–690.
    1. Jang YJ, Achary R, Lee HW, et al. Synthesis and anti-influenza virus activity of 4-oxo- or thioxo-4,5-dihydrofuro,4-c]pyridin- 3(1H)-ones. Antiviral Res. 2014;107:66–75.
    1. Lou J, Yang X, Rao Z, et al. Design and synthesis of 6-oxo- 1,4,5,6-tetrahydropyrimidine-5-carboxylate derivatives as neuraminidase inhibitors. Eur J Med Chem. 2014;83:466–473.
    1. Li J, Zhang D, Zhu X, et al. Studies on synthesis and structureactivity relationship (SAR) of derivatives of a new natural product from marine fungi as inhibitors of influenza virus neuraminidase. Mar Drugs. 2011;9:1887–1901.
    1. Ding Y, Dou J, Teng Z, et al. Antiviral activity of baicalin against influenza A (H1N1/H3N2) virus in cell culture and in mice and its inhibition of neuraminidase. Arch Virol. 2014;159:3269–3278.
    1. Nayak MK, Agrawal AS, Bose S, et al. Antiviral activity of baicalin against influenza virus H1N1-pdm09 is due to modulation of NS1-mediated cellular innate immune responses. J Antimicrob Chemother. 2014;69:1298–1310.
    1. Wan Q, Wang H, Han X, et al. Baicalin inhibits TLR7/MYD88 signaling pathway activation to suppress lung inflammation in mice infected with influenza A virus. Biomed Rep. 2014;2:437–441.
    1. Xu G, Dou J, Zhang L, et al. Inhibitory effects of baicalein on the influenza virus in vivo is determined by baicalin in the serum. Biol Pharm Bull. 2010;33:238–243.
    1. Nagai T, Suzuki Y, Tomimori T, et al. Antiviral activity of plant flavonoid, 5,7,4'-trihydroxy-8-methoxyflavone, from the roots of Scutellaria baicalensis against influenza A (H3N2) and B viruses. Biol Pharm Bull. 1995;18:295–299.
    1. Nagai T, Miyaichi Y, Tomimori T, et al. In vivo anti-influenza virus activity of plant flavonoids possessing inhibitory activity for influenza virus sialidase. Antiviral Res. 1992;19:207–217.
    1. Hale BG, Randall RE, Ortìn J, et al. The multifunctional NS1 protein of influenza A viruses. J Gen Virol. 2008;89:2359–2376.
    1. Zhirnov OP, Konakova TE, Wolff T, et al. NS1 protein of influenza A virus down-regulates apoptosis. J Virol. 2002;76:1617–1625.
    1. Kong JQ, Shen JH, Huang Y, et al. Development of a yeast twohybrid screen for selection of A/H1N1 influenza NS1 non-structural protein and human CPSF30 protein interaction inhibitors. Yao Xue Xue Bao. 2010;45:388–394.
    1. Twu KY, Noah DL, Rao P, et al. The CPSF30 binding site on the NS1A protein of influenza A virus is a potential antiviral target. J Virol. 2006;80:3957–3965.
    1. Jablonski JJ, Basu D, Engel DA, et al. Design, synthesis, and evaluation of novel small molecule inhibitors of the influenza virus protein NS1. Bioorg Med Chem. 2012;20:487–497.
    1. Mata MA, Satterly N, Versteeg GA, et al. Chemical inhibition of RNA viruses reveals REDD1 as a host defense factor. Nat Chem Biol. 2011;7:712–719.
    1. Mahy BW, Cox NJ, Armstrong SJ, et al. Multiplication of influenza virus in the presence of cordycepin, an inhibitor of cellular RNA synthesis. Nat New Biol. 1973;243:172–174.
    1. Kurokawa M, Koyama AH, Yasuoka S, et al. Influenza virus overcomes apoptosis by rapid multiplication. Int J Mol Med. 1999;3:527–530.
    1. Zhirnov OP, Klenk HD. Control of apoptosis in influenza virusinfected cells by up-regulation of Akt and p53 signaling. Apoptosis. 2007;12:1419–1432.
    1. Palese P, Shaw ML. Orthomyxoviridae: the viruses and their replication. In: Knipe DM, Holey PM, editors. Fields Virology. 5th Edition. Vol. 2. Philadelphia: Lippincott Williams & Wilkins; 2007. pp. 1647–1689.
    1. Wurzer WJ, Planz O, Ehrhardt C, et al. Caspase 3 activation is essential for efficient influenza virus propagation. EMBO J. 2003;22:2717–2728.
    1. Hinshaw VS, Olsen CW, Dybdahl-Sissoko N, et al. Apoptosis: a mechanism of cell killing by influenza A and B viruses. J Virol. 1994;68:3667–3673.
    1. Jaworska J, Coulombe F, Downey J, et al. NLRX1 prevents mitochondrial induced apoptosis and enhances macrophage antiviral immunity by interacting with influenza virus PB1-F2 protein. Proc Natl Acad Sci USA. 2014;111:E2110–E2119.
    1. Furman D, Jojic V, Kidd B, et al. Apoptosis and other immune biomarkers predict influenza vaccine responsiveness. Mol Syst Biol. 2014;10:750–750.
    1. Feldman T, Kabaleeswaran V, Jang SB, et al. A class of allosteric caspase inhibitors identified by high-throughput screening. Mol Cell. 2012;47:585–595.
    1. Dai J, Wang G, Li W, et al. High-throughput screening for antiinfluenza A virus drugs and study of the mechanism of procyanidin on influenza A virus-induced autophagy. J Biomol Screen. 2012;17:605–617.
    1. Muniruzzaman S, Pan YT, Zeng Y, et al. Inhibition of glycoprotein processing by L-fructose and L-xylulose. Glycobiology. 1996;6:795–803.
    1. Hussain S, Miller JL, Harvey DJ, et al. Strain-specific antiviral activity of iminosugars against human influenza A viruses. J Antimicrob Chemother. 2014 in press.
    1. Oguin TH, 3rd, Sharma S, Stuart AD, et al. Phospholipase D facilitates efficient entry of influenza virus, allowing escape from innate immune inhibition. J Biol Chem. 2014;289:25405–25417.
    1. Husain M, Cheung CY. Histone deacetylase 6 inhibits influenza A virus release by downregulating the trafficking of viral components to the plasma membrane via its substrate, acetylated microtubules. J Virol. 2014;88:11229–11239.
    1. Hamamoto I, Harazaki K, Inase N, et al. Cyclosporin A inhibits the propagation of influenza virus by interfering with a late event in the virus life cycle. Jpn J Infect Dis. 2013;66:276–283.
    1. Hsieh CF, Yen HR, Liu CH, et al. Ching-fang-pai-tu-san inhibits the release of influenza virus. J Ethnopharmacol. 2012;144:533–544.
    1. Buffinton GD, Christen S, Peterhans E, et al. Oxidative stress in lungs of mice infected with influenza A virus. Free Radic Res Commun. 1992;16:99–110.
    1. Drago L, Nicola L, Ossola F, et al. In vitro antiviral activity of resveratrol against respiratory viruses. J Chemother. 2008;20:393–394.
    1. Palamara AT, Nencioni L, Aquilano K, et al. Inhibition of influenza A virus replication by resveratrol. J Infect Dis. 2005;191:1719–1729.
    1. Li C, Fang JS, Lian WW, et al. In vitro antiviral effects and 3D QSAR Study of resveratrol derivatives as potent inhibitors of influenza H1N1 neuraminidase. Chem Biol Drug Des. 2015;85:427–438.
    1. Furuya A, Uozaki M, Yamasaki H, et al. Antiviral effects of ascorbic and dehydroascorbic acids in vitro. Int J Mol Med. 2008;22:541–545.
    1. Uozaki M, Ikeda K, Tsujimoto K, et al. Antiviral effects of dehydroascorbic acid. Exp Ther Med. 2010;1:983–986.
    1. Khare D, Godbole NM, Pawar SD, et al. Calcitriol [1, 25[OH]2 D3] pre- and post-treatment suppresses inflammatory response to influenza A (H1N1) infection in human lung A549 epithelial cells. Eur J Nutr. 2013;52:1405–1415.
    1. Goldstein MR, Mascitelli L, Pezzetta F. Pandemic influenza A (H1N1): mandatory vitamin D supplementation? Med Hypotheses. 2010;74:756–756.
    1. Fedson DS. Pandemic influenza: a potential role for statins in treatment and prophylaxis. Clin. Infect. Dis. 2006;43:199–205.
    1. Fedson DS. Treating influenza with statins and other immunomodulatory agents. Antiviral Res. 2013;99:417–435.
    1. Mehrbod P, Hair-Bejo M, Tengku Ibrahim TA, et al. Simvastatin modulates cellular components in influenza A virus-infected cells. Int J Mol Med. 2014;34:61–73.
    1. Glück B, Schmidtke M, Walther M, et al. Simvastatin treatment showed no prophylactic effect in influenza virus-infected mice. J Med Virol. 2013;85:1978–1982.
    1. Magulick JP, Frei CR, Ali SK, et al. The effect of statin therapy on the incidence of infections: a retrospective cohort analysis. Am J Med Sci. 2014;347:211–216.
    1. Cho WK, Weeratunga P, Lee BH, et al. Epimedium koreanum Nakai displays broad spectrum of antiviral activity in vitro and in vivo by inducing cellular antiviral state. Viruses. 2015;7:352–377.
    1. Leibbrandt A, Meier C, König-Schuster M, et al. Iota-carrageenan is a potent inhibitor of influenza A virus infection. PLoS One. 2010;5:e14320–e14320.
    1. Koenighofer M, Lion T, Bodenteich A, et al. Carrageenan nasal spray in virus confirmed common cold: individual patient data analysis of two randomized controlled trials. Multidiscip Respir Med. 2014;9:57–57.
    1. Romantsov MG, Golofeevskiĭ SV. Cycloferon efficacy in the treatment of acute respiratory tract viral infection and influenza during the morbidity outbreak in 2009-201. Antibiot Khimioter. 2010;55:30–35.
    1. Romantsov MG, Ershov FI, Kovalenko AL, et al. The therapeutic efficacy of cycloferon and the pharmacological activity of interferon inducers. Ter Arkh. 2014;86:83–88.
    1. Sukhinin VP, Zarubaev VV, Platonov VG, et al. Protective effect of cycloferon in experimental influenza. Vopr Virusol. 2000;45:26–30.
    1. Tazulakhova EB, Parshina OV, Guseva TS, et al. Russian experience in screening, analysis, and clinical application of novel interferon inducers. J Interferon Cytokine Res. 2001;21:65–73.
    1. Ye S, Lowther S, Stambas J. Inhibition of Reactive Oxygen Species Production Ameliorates Inflammation Induced by Influenza A Viruses via Upregulation of SOCS1 and SOCS3. J Virol. 2015;89:2672–2683.
    1. Selemidis S, Seow HJ, Broughton BR, et al. Nox1 oxidase suppresses influenza a virus-induced lung inflammation and oxidative stress. PLoS One. 2013;8:e60792–e60792.
    1. Vlahos R, Selemidis S. NADPH oxidases as novel pharmacologic targets against influenza A virus infection. Mol Pharmacol. 2014;86:747–759.
    1. Sharma G, Sharma DC, Fen LH, et al. Reduction of influenza virus-induced lung inflammation and mortality in animals treated with a phosophodisestrase-4 inhibitor and a selective serotonin reuptake inhibitor. Emerging Microbes & Infections. 2013;2:e54–e54.
    1. Marsolais D, Hahm B, Walsh KB, et al. A critical role for the sphingosine analog AAL-R in dampening the cytokine response during influenza virus infection. Proc Natl Acad Sci USA. 2009;106:1560–1565.
    1. Sugamata R, Sugawara A, Nagao T, et al. Leucomycin A3, a 16-membered macrolide antibiotic, inhibits influenza A virus infection and disease progression. J Antibiot (Tokyo) 2014;67:213–222.
    1. BG-777. Accessible at .
    1. Mbawuike I, Zang Y, Couch RB. Humoral and cell-mediated immune responses of humans to inactivated influenza vaccine with or without QS21 adjuvant. Vaccine. 2007;25:3263–3269.
    1. Panatto D, Amicizia D, Lai PL, et al. Utility of thymosin alpha-1 (Zadaxin) as a co-adjuvant in influenza vaccines: a review. J Prev Med Hyg. 2011;52:111–115.
    1. Carraro G, Naso A, Montomoli E, et al. Thymosin-alpha 1 (Zadaxin) enhances the immunogenicity of an adjuvated pandemic H1N1v influenza vaccine (Focetria) in hemodialyzed patients: a pilot study. Vaccine. 2012;30:1170–1180.
    1. Gravenstein S, Duthie EH, Miller BA, et al. Augmentation of influenza antibody response in elderly men by thymosin alpha one. A double-blind placebo-controlled clinical study. J Am Geriatr Soc. 1989;37:1–8.
    1. Chioato A, Noseda E, Felix SD, et al. Influenza and meningococcal vaccinations are effective in healthy subjects treated with the interleukin-1 beta-blocking antibody canakinumab: results of an open-label, parallel group, randomized, singlecenter study. Clin Vaccine Immunol. 2010;17:1952–1957.
    1. Jefferson T, Jones MA, Doshi P, et al. Neuraminidase inhibitors for preventing and treating influenza in healthy adults and children. Cochrane Database Syst Rev. 2014;4:CD008965–CD008965.
    1. CDC , author. Influenza antiviral medications: summary for clinicians (current for the 2013-14 influenza season) Document available at: . Accessed on 24th July 2014.
    1. WHO , author. Global Alert and Response (GAR) antiviral drugs for pandemic (H1N1) 2009: definitions and use. Document available at: . Accessed on 24th July 2014).
    1. Budd A, Alleva L, Alsharifi M, et al. Increased survival after gemfibrozil treatment of severe mouse influenza. Antimicrob Agents Chemother. 2007;51:2965–2968.
    1. Lin KL, Sweeney S, Kang BD, et al. CCR2-antagonist prophylaxis reduces pulmonary immune pathology and markedly improves survival during influenza infection. J Immunol. 2011;186:508–515.
    1. Zheng BJ, Chan KW, Lin YP, et al. Delayed antiviral plus immunomodulator treatment still reduces mortality in mice infected by high inoculum of influenza A/H5N1 virus. Proc Natl Acad Sci USA. 2008;105:8091–8096.
    1. Ghezzi P, Ungheri D. Synergistic combination of N-acetylcysteine and ribavirin to protect from lethal influenza viral infection in a mouse model. Int J Immunopathol Pharmacol. 2004;17:99–102.
    1. Ottolini M, Blanco J, Porter D, et al. Combination anti-inflammatory and antiviral therapy of influenza in a cotton rat model. Pediatr Pulmonol. 2003;36:290–294.
    1. Khanna M, Saxena L, Rajput R, et al. Gene silencing: a therapeutic approach to combat influenza virus infections. Future Microbiol. 2015;10:131–140.
    1. Wichadakul D, Mhuantong W, Jongkaewwattana A, et al. A computational tool for the design of live attenuated virus vaccine based on microRNA-mediated gene silencing. BMC Genomics. 2012;13(Suppl 7):S15–S15.
    1. Buggele WA, Johnson KE, Horvath CM. Influenza A virus infection of human respiratory cells induces primary microRNA expression. J Biol Chem. 2012;287:31027–31040.
    1. Langlois RA, Albrecht RA, Kimble B, et al. MicroRNA-based strategy to mitigate the risk of gain-of-function influenza studies. Nat Biotechnol. 2013;31:844–847.
    1. Zhang H, Li Z, Li Y, et al. A computational method for predicting regulation of human microRNAs on the influenza virus genome. BMC Syst Biol. 2013;7(Suppl 2):S3–S3.
    1. Li Y, Chan EY, Li J, et al. MicroRNA expression and virulence in pandemic influenza virus-infected mice. J Virol. 2010;84:3023–3032.
    1. Betáková T, Svančarová P. Role and application of RNA interference in replication of influenza viruses. Acta Virol. 2013;57:97–104.
    1. Seth S, Templin MV, Severson G, et al. A potential therapeutic for pandemic influenza using RNA interference. Methods Mol Biol. 2010;623:397–422.
    1. Ge Q, Filip L, Bai A, et al. Inhibition of influenza virus production in virus-infected mice by RNA interference. Proc Natl Acad Sci USA. 2004;101:8676–8681.
    1. Winterling C, Koch M, Koeppel M, et al. Evidence for a crucial role of a host non-coding RNA in influenza A virus replication. RNA Biol. 2014;11:66–75.
    1. Ouyang J, Zhu X, Chen Y, et al. NRAV, a long noncoding RNA, modulates antiviral responses through suppression of interferon-stimulated gene transcription. Cell Host Microbe. 2014;16:616–626.
    1. Xiang DX, Chen Q, Pang L, et al. Inhibitory effects of silver nanoparticles on H1N1 influenza A virus in vitro. J Virol Methods. 2011;178:137–142.
    1. Xiang D, Zheng Y, Duan W, et al. Inhibition of A/Human/ Hubei/3/2005 (H3N2) influenza virus infection by silver nanoparticles in vitro and in vivo. Int J Nanomedicine. 2013;8:4103–4113.
    1. McKenzie Z, Kendall M, Mackay RM, et al. Nanoparticles modulate surfactant protein A and D mediated protection against influenza A infection in vitro. Philos Trans R Soc Lond B Biol Sci. 2015;370:20140049–20140049.
    1. Torrecilla J, Rodríguez-Gascón A, Solinís MÁ, et al. Lipid nanoparticles as carriers for RNAi against viral infections: current status and future perspectives. Biomed Res Int. 2014;2014:161794–161794.
    1. Okamoto S, Yoshii H, Akagi T, et al. Influenza hemagglutinin vaccine with poly(gamma-glutamic acid) nanoparticles enhances the protection against influenza virus infection through both humoral and cell-mediated immunity. Vaccine. 2007;25:8270–8278.
    1. Shoji M, Takahashi E, Hatakeyama D, et al. Anti-influenza activity of c60 fullerene derivatives. PLoS One. 2013;8:e66337–e66337.
    1. Amidi M, Romeijn SG, Verhoef JC, et al. N-trimethyl chitosan (TMC) nanoparticles loaded with influenza subunit antigen for intranasal vaccination: biological properties and immunogenicity in a mouse model. Vaccine. 2007;25:144–153.
    1. Sawaengsak C, Mori Y, Yamanishi K, et al. Chitosan nanoparticle encapsulated hemagglutinin-split influenza virus mucosal vaccine. AAPS PharmSciTech. 2014;15:317–325.
    1. Sawaengsak C, Mori Y, Yamanishi K, et al. Intranasal chitosan- DNA vaccines that protect across influenza virus subtypes. Int J Pharm. 2014;473:113–125.
    1. Sanpui P, Zheng X, Loeb JC, et al. Single-walled carbon nanotubes increase pandemic influenza A H1N1 virus infectivity of lung epithelial cells. Part Fibre Toxicol. 2014;11:66–66.
    1. Govorkova EA, McCullers JA. Therapeutics against influenza. Curr Top Microbiol Immunol. 2013;370:273–300.
    1. Govorkova EA, Webster RG. Combination chemotherapy for influenza. Viruses. 2010;2:1510–1529.
    1. Prabakaran M, Prabhu N, He F, et al. Combination therapy using chimeric monoclonal antibodies protects mice from lethal H5N1 infection and prevents formation of escape mutants. PLoS One. 2009;4:e5672–e5672.
    1. Bastos LF, Coelho MM. Drug repositioning: playing dirty to kill pain. CNS Drugs. 2014;28:45–61.
    1. Wilkinson GF, Pritchard K. In vitro screening for drug repositioning. J Biomol Screen. 2015;20:167–179.
    1. Heldt FS, Frensing T, Pflugmacher A, et al. Multiscale modeling of influenza A virus infection supports the development of direct-acting antivirals. PLoS Comput Biol. 2013;9:e1003372–e1003372.
    1. Smith SB, Dampier W, Tozeren A, et al. Identification of common biological pathways and drug targets across multiple respiratory viruses based on human host gene expression analysis. PLoS One. 2012;7:e33174–e33174.
    1. Law GL, Tisoncik-Go J, Korth MJ, et al. Drug repurposing: a better approach for infectious disease drug discovery? Curr Opin Immunol. 2013;25:588–592.
    1. Sharma D, Priyadarshini P, Vrati S. Unraveling the web of viroinformatics: computational tools and databases in virus research. J Virol. 2015;89:1489–1501.
    1. Bao S, Zhou X, Zhang L, et al. Prioritizing genes responsible for host resistance to influenza using network approaches. BMC Genomics. 2013;14:816–816.
    1. Li Z, Zhou H, Lu Y, et al. A critical role for immune system response in mediating anti-influenza drug synergies assessed by mechanistic modeling. CPT Pharmacometrics Syst Pharmacol. 2014;3:e135–e135.
    1. Frank D. One world, one health, one medicine. Can Vet J. 2008;49:1063–1065.
    1. Travis DA, Sriramarao P, Cardona C, et al. One medicine one science: a framework for exploring challenges at the intersection of animals, humans, and the environment. Ann N Y Acad Sci. 2014;1334:26–44.

Source: PubMed

3
Iratkozz fel