Skip to main content
Influenza and Other Respiratory Viruses logoLink to Influenza and Other Respiratory Viruses
. 2012 Dec 28;7(Suppl 1):63–75. doi: 10.1111/irv.12045

Newer influenza antivirals, biotherapeutics and combinations

Frederick G Hayden 1,2
PMCID: PMC5978626  PMID: 23279899

Abstract

Please cite this paper as: Hayden FG. (2012) Newer Influenza Antivirals, Biotherapeutics and Combinations. Influenza and Other Respiratory Viruses 7(Suppl. 1), 63–75.

This summary provides an overview of investigational antiviral agents for influenza and of future directions for development of influenza therapeutics. While progress in developing clinically useful antiviral agents for influenza has been generally slow, especially with respect to seriously ill and high‐risk patients, important clinical studies of intravenous neuraminidase inhibitors, antibodies and drug combinations are currently in progress. The current decade offers the promise of developing small molecular weight inhibitors with novel mechanisms of action, including host‐directed therapies, new biotherapeutics and drug combinations, that should provide more effective antiviral therapies and help mitigate the problem of antiviral resistance. Immunomodulatory interventions also offer promise but need to be based on better understanding of influenza pathogenesis, particularly in seriously ill patients. The development of combination interventions, immunomodulators and host‐directed therapies presents unique clinical trial design and regulatory hurdles that remain to be addressed.

Keywords: Antibodies, antivirals, combinations, influenza, immunomodulators, neuraminidase inhibitors

Introduction

The purpose of this summary is to provide an overview of where we stand with respect to investigational antiviral agents for influenza and of future directions for development of influenza therapeutics. This commentary is based on a presentation at the first isirv Antiviral Group meeting in November 2011 and focuses primarily on clinical studies. It updates previous reviews 1 and summaries based on presentations by the author at international meetings in 2006, 2 , 3 2008, 4 and 2010. 5 In addition, a number of review articles that provide more detailed consideration of pre‐clinical and clinical aspects of influenza antiviral development have been published recently by others. 6 , 7 , 8 , 9 , 10 , 11 , 12

When considered historically, the development of the first class of influenza antivirals, the aminoadamantanes (amantadine, rimantadine), dates to nearly five decades ago in the 1960s. Studies on ribavirin administered by various routes and on intranasal interferons followed in the 1970s and 1980s, respectively, but did not lead to approval for influenza in most countries. Developmental work on the second class of currently available agents, the neuraminidase inhibitors (NAIs) (zanamivir, oseltamivir), took place during the 1990s, but overall progress on developing clinically useful antivirals for influenza has been slow. In addition, global circulation of influenza A(H3N2) viruses resistant to the aminoadamantanes and of seasonal A(H1N1) viruses resistant to oseltamivir, 13 , 14 as well as instances of oseltamivir resistance among the 2009 pandemic A(H1N1) viruses, 15 are reminders of the very limited size of our current influenza antiviral armamentarium.

However, recent pre‐clinical studies have identified interesting inhibitors of influenza virus replication, and several of these have just entered or are expected to enter into clinical development. Table 1 contains a representative list of those which have shown activity either in animal models of influenza or in some cases in infected humans. In addition to the viral targets of inhibitors with proven clinical utility (i.e. M2, NA), a variety of targets and approaches have been identified that could potentially be used for developing new inhibitors. At meetings organised by the National Institute of Allergy and Infectious Diseases in 2009 16 and 2011, 17 investigators provided detailed updates on many of these approaches. Substantial data have also emerged in regard to using dual or multiple inhibitors in combination, including one modality combining three available agents, to increase effectiveness and manage the problem of anti‐viral resistance. With adequate funding and agreement on feasible clinical study pathways to address regulatory concerns, 18 , 19 the current decade offers the promise of progress in developing agents with novel mechanisms of action and of drug combinations that provide more effective therapies.

Table 1.

 Representative investigational anti‐influenza agents and biotherapeutics with antiviral activity in animal models and/or humans

NA inhibitors (NAIs)
 Peramivir (IV)*, zanamivir (IV)*, oseltamivir (IV)
 A‐315675 (oral)(120, 121)
Long‐acting NAIs (LANIs)
 Laninamivir (topical)*
 ZNV dimers (topical)*
Conjugated sialidase
 DAS181 (topical)*
Protease inhibitors
 Aprotinin (topical, IV) (122)
HA inhibitors and viral binding agents
 Peptides‐ FluPep (topical) (123), Entry Blocker (topical) (124), HB80/36 (70), Flufirvitide (72, 73)
 Arbidol (oral) (125, 126)
 Cyanovirin‐N (topical) (127)
 Iota‐carrageenan (topical) (128)
 Pentraxin PTX3 (IP) (129)
 Polymer bound 6′ sialyl‐N‐acetyllactosamine (topical)(130)
 CYSTUS052 (topical) (131)
 Recombinant human galectin‐1 (topical)(132)
Polymerase inhibitors
 Ribavirin (oral, IV, inhaled)(3)
 Favipiravir/T‐705 (oral)*
 Viramidine (oral) (133)
 Antisense oligonucleotides (IV, topical) (134, 135)
M gene
 Antisense oligonucleotide (AVI‐7100) (topical, IV)*
NP inhibitors
 Nucleozin (IP) (136, 137)
 Antisense oligonucleotides (IV) (93, 138)
Interferons (139, 140, 141, 142, 143)
 IFN inducers‐ poly‐ICLC (topical) (144, 145); (107), nitazoxanide (PO)*
 RIG‐I activator (5′PPP‐RNA) (IV) (146)
Antibodies to viral proteins
 Convalescent plasma, hyperimmune globulin*
 Anti‐HA, M2e, NA, NP*
Other topical agents
 Cationic airway lining modulators (iCALM‐ topical)(16, 147)
 Surfactant nano‐emulsions (topical) (148)
 SOFA‐HDV ribozymes targeting M, NS, NP (149)
 Defective interfering particles (244 DI RNA in a cloned A/PR/8/34) (150)

*See text for discussion of selected agents and additional references.

IN, intranasal; IP, intraperitoneal; IV, intravenous; SC, subcutaneous.

Antivirals in current clinical development

As shown in Table 2, there is a relatively short list of anti‐influenza agents in advanced clinical development and a focus on NAIs, including three being developed for intravenous (IV) administration. Intravenous peramivir and the inhaled long‐acting NAI laninamivir are already approved in Japan, and peramivir also in South Korea. In addition, several novel agents that retain activity against influenza viruses resistant to the currently available classes of drug are also under clinical study.

Table 2.

 Selected influenza antiviral agents in advanced clinical development

Agent Viral target Sponsor Route Development phase
Zanamivir NA GSK IV Phase 3
Peramivir NA Biocryst, Shionogi IV Phase 3*
Oseltamivir NA Roche IV Phase 3
Laninamivir (CS‐8958) NA Biota, Daiichi‐Sankyo Inhaled Phase 3*
Favipiravir (T‐705) Polymerase Toyama Oral Phase 2‐3
DAS181 HA receptor Nexbio Inhaled Phase 1‐2
Nitazoxanide Possibly HA; IFN inducer Romark Oral Phase 2

*Licensed in Japan.

licensed in South Korea.

Neuraminidase inhibitors

A medical need for parenteral antivirals in treating severe influenza has been recognised for many years. Intravenous administration of NAIs like zanamivir and peramivir can guarantee a rapid delivery of high‐plasma drug levels in a reliable fashion. Indeed, the maximum plasma concentrations following IV zanamivir or peramivir are approximately 50‐fold higher than those observed with double‐dose (150 mg) oseltamivir, although the plasma AUC and minimum concentrations are closer. 20 Whether these pharmacologic differences will translate into greater antiviral activity, less frequent resistance emergence, and improved clinical outcomes remains to be determined.

While the available NAIs have inhibitory activity against influenza A and B viruses, their antiviral spectra and cross‐resistance patterns vary by agent as they bind differently within the active site of the enzyme. 11 , 13 In general, zanamivir and laninamivir have similar profiles of susceptibility. For example, the H275Y mutation confers high‐level resistance to oseltamivir carboxylate and reduced susceptibility to peramivir in N1‐containing viruses but does not substantially diminish susceptibility to zanamivir and laninamivir. 21 Although peramivir has been reported to inhibit a laboratory strain of influenza A(H1N1) with H275Y, 22 this particular mutation has emerged during in vitro passage with peramivir and also during its therapeutic use in an immunocompromised patient. 23 Furthermore, IV peramivir has not shown antiviral effects in treating infections due to oseltamivir‐resistant A(H1N1)pdm09 infections. 24 Consequently, peramivir would not be reliable in treating such resistant variants, especially in immunocompromised hosts.

In adults with uncomplicated influenza, single IV doses of peramivir (300 or 600 mg) were superior to placebo 25 and comparable to a 5‐day course of oseltamivir, 26 but IV peramivir was no better than oseltamivir in treating adults infected with oseltamivir‐resistant seasonal A(H1N1) virus harbouring the H275Y mutation. Peramivir in daily IV doses (200 or 400 mg once daily for 5 days) was comparable to oseltamivir in hospitalised adults and did not select for resistance, 27 but a once‐daily dose of 300 mg appeared less effective than one of 600 mg. 28 Peramivir was used on both compassionate use and Emergency Use Authorization bases in the United States for treating severe pandemic 2009 A(H1N1) illness, 29 and controlled studies in hospitalised patients are in progress.

Intravenous zanamivir was used extensively on a compassionate use basis during the 2009 pandemic, particularly for treating suspected or proven oseltamivir resistance, 30 , 31 , 32 and a phase III trial is currently in progress to compare IV zanamivir to oral oseltamivir in hospitalised patients. In a small, phase II study, 33 hospitalised patients with high frequencies of severe illness (40% requiring mechanical ventilation), co‐morbidities and prior oseltamivir therapy were initiated on IV zanamivir at a median of 5 days after symptom onset when they still had, despite oseltamivir treatment, high levels of viral RNA in nasopharyngeal samples. Zanamivir in this setting was temporally associated with median viral RNA load reductions of nearly two log10 over the subsequent 4–5 days of administration. It remains to be determined whether even more rapid and profound anti‐viral inhibition might be possible with combinations of antivirals.

Inhalation of the NAI laninamivir prodrug (termed CS‐8958) provides prolonged duration of antiviral activity in animal models 34 and prolonged presence of laninamivir in humans. 35 Laninamivir has an antiviral spectrum similar to zanamivir 21 and was found to be superior to oseltamivir in treating children infected with oseltamivir‐resistant seasonal A(H1N1) virus. 36 Single inhaled doses of laninamivir (20 mg or 40 mg) were comparable to 5 days of oseltamivir in adults, 37 although for unclear reasons it was not superior in treating adults infected with oseltamivir‐resistant seasonal A(H1N1) virus. Inhaled dimers of zanamivir are also in early clinical development. 38 , 39

Conjugated sialidase

DAS181 is a novel fusion construct that includes the catalytic domain from Actinomyces viscosus sialidase linked with an epithelium‐anchoring domain of human amphiregulin. 40 This sialidase removes both the human‐like α2,6‐ and avian‐like α2,3‐linked sialic acids from cellular receptors, and hence, this agent has a broad range of activity for influenza viruses, including those resistant to the amino‐adamantanes and NAIs. Resistance has been difficult to select during in vitro passage and appears low‐level (3‐ to 18‐fold reductions in susceptibility). 41 When administered topically, DAS181 shows inhibitory activity in animal models, including infections due to avian A(H5N1) and A(H1N1)pdm09 viruses. 42 , 43 DAS181 is also inhibitory for parainfluenza viruses in vitro and in the cotton rat model 44 ; inhaled DAS181 has been given on compassionate use basis to hematopoietic stem cell and lung transplant patients with severe PIV infection with apparent benefit. 45 , 46

In a phase II randomised, controlled trial (RCT) of this agent for treating uncomplicated influenza, 47 264 previously healthy adults with acute influenza were randomised to receive treatment with a single 10‐mg inhalation of DAS181, once‐daily inhalations for 3 days or placebo in a double‐blinded fashion. Throat gargle virus titres, the primary virologic end point, showed significantly greater declines between the day of enrolment and the following day in the active groups compared with placebo. This accelerated clearance of pharyngeal virus continued to day 5 in the group that received DAS181 treatment over 3 days but was not seen with a single administration. This trial showed an encouraging antiviral effect, although this was not associated with greater improvement in symptom resolution. The reasons for this apparent discrepancy remain to be clarified but may relate to the relatively mild influenza illness in these patients. More work needs to be done to assess the tolerability and efficacy of different topical formulations of this novel host‐directed inhibitor for potential influenza management.

Favipiravir

Favipiravir, previously designated T‐705, also has a unique mechanism of antiviral action, so that it has inhibitory activity against both NAI‐ and aminoadamantane‐resistant viruses. 48 , 49 After undergoing intracellular metabolism (ribosylation and phosphorylation), so that it has a nucleoside‐like configuration, the triphosphate inhibits influenza RNA polymerase. 50 In vitro favipiravir is active against all influenza types (A, B, C) at relatively low concentrations (0·01–0·5 ug/ml), and higher concentrations also show activity against some other RNA viruses. 50 Oral favipiravir is active in murine models of influenza, including lethal A(H5N1), 49 and shows synergistic interactions with oseltamivir. 51 Favipiravir‐resistant variants have not been reported to date.

In a phase II randomised, double‐blind controlled trial in Japan, oral favipiravir (600 mg BID twice daily for 1 day followed by 600 mg daily for 4 days) gave a similar mean time to illness alleviation when compared to oseltamivir (approximately 50 hours in both groups), whereas a lower favipiravir dose was less effective. 52 Pharmacokinetic studies have shown that there is a need for both initial loading doses and dose adjustments based on weight and perhaps ethnicity. A large phase III treatment study of ambulatory patients with uncomplicated influenza has been conducted in Japan and other Asian countries. The time courses of resolution of virus detection in the upper respiratory tract, based on titres of infectious virus, were comparable in the favipiravir (1200 mg once followed by 400 mg on day 1 and then 400 mg BID for 4 days) and oseltamivir groups. 52 A phase II placebo‐controlled RCT treatment study in adults aged 55–80 years (favipiravir doses of 1000 mg BID on 1 day and then 400 mg BID for 4 days versus 1200 mg BID on 1 day and then 800 mg BID for 4 days) is in progress in the USA and other countries. While the clinical efficacy and safety data remain to be published from these studies, the available data show that favipiravir exerts anti‐viral effects in humans. These proof‐of‐concept findings confirm that influenza viral polymerase is an important target for antiviral development.

Nitazoxanide

Nitazoxanide, an oral antiparasitic agent, has interesting immunomodulatory effects, including up‐regulation of various interferon and interferon‐inducible genes. In addition, it has been reported to exert a specific influenza inhibitory effect related to blockade of HA maturation. 53 A recent phase II RCT compared two different doses of nitazoxanide (300 or 600 mg twice daily for 5 days) to placebo in ambulatory patients with suspected influenza. 54 Among 257 influenza‐infected persons, the time to alleviation of symptoms, similar to the end point that was used in the pivotal NAI trials, was shorter by about 20 hours in the high‐dose group compared with placebo. This study also found evidence for an antiviral effect with an approximate 1 log10 reduction in treatment day 1 virus titres in the high‐dose group compared with placebo. The 300‐mg dose groups showed intermediate effects. These interesting results need confirmation, but given the extensive safety record of this drug and its unique mechanisms of action, it might be particularly interesting for use in combination with other antiviral agents.

Antibodies

Interest in antibody therapies of influenza has been stimulated in part by observations from the use of convalescent blood products as therapy in pneumonia patients during the 1918 pandemic. 55 Although these studies were not RCTs and used various forms of blood products, a retrospective analysis found a very dramatic reduction in overall mortality (crude case‐fatality, 16% in treated versus 37% in controls), particularly if the products were administered within 4 days of a pneumonia diagnosis (19% compared to 59% with delayed treatment). 55 More recent anecdotal reports of administering convalescent plasma for treatment of severe avian A(H5N1) 56 and 2009 pandemic A(H1N1) 57 illness have also indicated benefit. A case–control study of convalescent 2009 pandemic A(H1N1) plasma, selected to have relatively high neutralising antibody titres, compared outcomes in 20 patients given plasma and 70 controls, all of whom were critically ill in intensive care (94% receiving mechanical ventilation) and already receiving oseltamivir therapy. 57 The crude case‐fatality was much lower with convalescent plasma compared with no treatment (20% versus 55%), and there was also a suggestion of some acceleration of virus clearance Because of these promising observations, a RCT is now being mounted through the NIAID to determine whether addition of convalescent plasma adds to oseltamivir therapy in seriously ill hospitalised patients.

This area has also received increased interest because of the identification of conserved epitopes on the stem region of the influenza haemagglutin (HA). The 16 HA subtypes can be divided into two phylogenetic groups, designated group 1 (containing H1, H2, H5, H9, and others) and group 2 (containing H3, H7, and others). Hetero‐subtypic, neutralising monoclonal human antibodies that are therapeutically active after passive transfer in mice and ferrets have been identified for both group 1 58 , 59 , 60 , 61 , 62 and more recently group 2 HAs. 63 In one case, an antibody that recognises HA subtypes in both groups to variable extent has been reported. 64 These antibodies target conserved sites on the stem region and prevent the conformational changes in HA needed for membrane fusion during replication. 59 , 60 Several of these antibodies have gone into initial clinical studies or are about to so. These broad spectrum neutralising anti‐HA monoclonal antibodies and possibly ones directed to other relatively conserved epitopes on M2e, 65 , 66 which appear to mediate cellular cytotoxicity and require intact Fc receptors, 67 and possibly NA 68 or NP, 69 offer the interesting prospect of combination therapies with small molecular weight inhibitors and activity against influenza viruses resistant to NAIs and/or aminoadamantanes.

In addition, the identification of highly conserved regions in the HA stalk has led to the identification of peptides that are able to bind potently and inhibit the fusogenic activity of HA. 70 , 71 One of these peptides, a 16‐mer called flufirvitide, has progressed into initial clinical development. 72 , 73

Antiviral combinations

The fact that combinations of influenza antivirals offer the possibilities of enhanced potency and reduced resistance emergence, as well as potential dose‐sparing, is a well‐established concept. Work in this field started over 40 years ago with an amantadine and interferon combination. 74 About 25 years ago, the first triple drug combination including interferon, rimantadine and ribavirin was described. 75 Subsequent pre‐clinical studies have indicated that if an influenza A virus is aminoadamantane‐susceptible, synergistic interactions in vitro and increased survival in murine models of influenza, including A(H5N1), are observed when the aminoadamantane is combined with a NAI or ribavirin. 76 , 77 , 78 If a virus is aminoadamantane resistant, no consistent benefit has been found in using the aminoadamantane in combination with oseltamivir or ribavirin. Ribavirin and oseltamivir show primarily additive interactions in vitro and in murine models of A(H5N1), 78 , 79 , 80 whereas favipiravir and NAIs show dose‐related additive to synergistic effects for influenza A viruses in vitro and on survival in mice. 51 Combinations of oseltamivir and zanamivir showed concentration‐related additive to antagonistic antiviral effects for A(H1N1)pdm09 viruses in vitro, 81 whereas combinations of oseltamivir and peramivir showed primarily additive activities in vitro and in mice. 82 These reports did not describe possible effects in preventing resistance emergence, although such a benefit was seen with aminoadamantane and oseltamivir combinations for a range of aminoadamantane‐susceptible influenza A viruses. 76

An increasing number of human studies have been done to assess influenza antiviral combinations, most often examining possible pharmacokinetic interactions and tolerability with currently available agents (e.g. oral oseltamivir + amantadine, oral oseltamivir + favipiravir, IV peramivir + oral rimantadine, IV peramivir + oral oseltamivir, IV zanamivir + oral oseltamivir). 83 , 84 , 85 In general, these combinations appear to be adequately tolerated without important pharmacokinetic interactions. However, the number of combinations that have been tested for efficacy in humans in controlled trials is much more limited. One placebo‐controlled trial of nebulised zanamivir in hospitalised influenza A‐infected patients, all of whom were given rimantadine, was under‐enrolled but found interesting trends towards faster cough resolution and lesser risk of rimantadine resistance emergence. 86 In contrast, a recent double‐blind, placebo‐controlled RCT highlighted the potential for antagonism with dual NAI use, when it found slower virologic and clinical responses in those given combined therapy with oseltamivir and inhaled zanamivir compared with oseltamivir alone in uncomplicated influenza. 87 Consequently, combinations of zanamivir and oseltamivir need further evaluation before being used in clinical practice. As indicated previously, a controlled study of convalescent 2009 pandemic A(H1N1) plasma combined with oseltamivir therapy is ongoing in hospitalised patients under NIAID sponsorship.

One triple drug regimen with three available agents (amantadine, ribavirin, oseltamivir) showed synergistic activity in vitro against not only influenza A viruses that are susceptible 88 but also those resistant to the amantadine or oseltamivir at baseline, including A(H1N1)pdm09 virus. 81 This triple regimen, termed TCAD, was more inhibitory than any of the dual combinations and was also more effective at preventing resistance emergence during in vitro passage. 89 Murine model studies indicated that amantadine contributes to the activity of TCAD and also enhances the activity of oseltamivir in a dual combination, in increasing survival following infection by amantadine‐resistant A(H1N1)pdm09 virus, 90 although the mechanisms have not been clarified. TCAD has been studied in a small cohort of highly immunocompromised patients with influenza at the Fred Hutchinson Cancer Centre in Seattle [Janet Englund, presented at ICAR, April 2010]. Those who received the triple regimen did not show the emergence of new resistance mutations, and the regimen was reasonably well‐tolerated over 10 days and provided the target blood levels of the individual drugs. A retrospective Korean study of critically ill adults with influenza A(H1N1)pdm09 infection suggested trends towards lower 14‐day (17% versus 35%; P = 0·08) and 90‐day (46% versus 59%; P = 0·23) mortality in TCAD recipients compared with those receiving oseltamivir monotherapy. 91 A RCT trial sponsored by NIAID comparing TCAD to oseltamivir monotherapy for ambulatory high‐risk patients is in progress.

There are a number of possibilities with regard to future combinations of antivirals and of antivirals combined with biotherapeutics including nitazoxanide and therapeutic antibodies, as well as immunomodulators. Combining antivirals with different mechanisms of action, for example, a polymerase inhibitor‐like favipiravir with a NAI, would be especially interesting for treating more severe forms of influenza or infections in immunocompromised hosts. A large number of potential immunomodulatory agents have been proposed for adjunctive influenza treatment, many of which have shown activity in animal models (Table 3). For example, one recent report looked at a strategy of targeting sphingosine‐1‐ phosphate (S1P) receptors with a sphingosine analog, designated AAL‐R, to inhibit various pro‐inflammatory cytokine and chemokine responses. 92 In a murine model of A(H1N1)pdm09 infection, intratracheal application of AAL‐R alone had a beneficial effect (survival increased to 82% compared to 21% in vehicle control and to 50% with oseltamivir), and when combined with oseltamivir, 96% survival was observed.

Table 3.

 Examples of potential adjunctive influenza treatments tested in animal models or used in humans

Proposed agent Comment/Influenza model system
Glucocorticoids IN reduced inflammation in cotton rats (151), but systemic delivery ineffective for A(H5N1) in mice (152); strong observational evidence for harmful effects in severe human influenza (see text)
Statins Oral rosuvastatin ineffective in murine model (153), but combined statin/caffeine IN or oral inhibited viral replication (154). Reduced mortality reported in hospitalised influenza patients on prior therapy (see text)
Gemfibrizol IP therapy increased survival in mice (155)
Pioglitazone PPAR‐γ agonist beneficial for A(H5N1) in mice by decreasing tipDC trafficking to lung (156). Pre‐treatment with pioglitazone or rosiglitazone reduced influenza mortality in one murine model (157). AICAR (aminoimidazole carboxamide ribonucleotide), an activator of AMP‐activated protein kinase (AMPK) that stimulates PPARs, is also active in mice (157).
PF‐04178903 Prophylactic SC delivery of CCR2 blocker increased survival and decreased inflammatory markers in mice (158)
AAL‐R Topical sphingosine‐1‐phosphate receptor agonist active in mice alone and in combination with oseltamivir (92)
Cyclo‐oxygenase inhibitors Cox‐2 inhibitor (celecoxib) beneficial with NAI for treating A(H5N1) in mice but ineffective alone (159). Pre‐treatment with Cox‐1 inhibitor (SC‐560) associated with hypothermia, weight loss, and increased mortality in mice (160)
N‐acetyl‐cysteine+ Dose‐related protection alone and with antivirals in mice (161); case report of possible benefit (162)
Chloroquine Ineffective in mice and ferrets (163), and in oral prophylaxis RCT in humans (164)
Bacterial lysate Prophylactic topical delivery increased survival and decreased virus titres in mice (106)
Erythromycin IP delivery increased survival and modulated immune measures in mice (165)
Ketotifen Oral mast cell degranulation inhibitor reduced inflammatory mediators in H5N1‐infected mice and was highly protective in combination with oseltamivir (166)
Pamidronate Increased survival and antiviral effects in humanised mice (167)
Allopurinol Oral allopurinol, an inhibitor of xanthine oxidase, and IV superoxide dismutase, an oxygen radical scavenger, reduced mortality in mice (168)
Cocaine Modest antiviral effect after IP dosing in mice (169)
CpG oligonucleotides Topical TLR‐9 agonist protective in mice (144)
3M‐011 IN TLR7/8 agonist active in mice (170)
Lactobacilus pentosus and plantarum IN delivery of strain S‐PT84 protective against influenza A challenge in mice (171). Oral prophylaxis with killed strains showed dose‐related immunostimulatory effects, reduced lung virus titres, and increased survival in mice(172, 173)
TJS‐064 Oral traditional Chinese herbal therapy active in mice (174)
Maxingshigan‐Yinqiaosan Oral traditional Chinese therapy comparable to oseltamivir in fever resolution in RCT in uncomplicated influenza A(H1N1)pdm09 (175)
Nitric oxide inhalation No antiviral or beneficial clinical effects in mice (176)
Echinacea extract Oral use reduced weight loss and inflammation measures in mice (177)
PUL‐042 Inhaled oligodeoxynucleotide and lipoprotein immunotherapeutic that protects against various pathogens in mice (17)
Clara cell protein CC10 Topical dosing of recombinant human CC10 showed antiviral effects in cotton rats (17)
Gabexate mesilate Protease inhibitor that reduces cytokine responses in mice after IP dosing (178)
Green tea (catechins) Oral catechins and theanine prophylaxis RCT showed possible reduced influenza infections (179)
Chitin microparticles IN prophylaxis increased A(H5N1) survival in mice (180)
Isoquercetin Plant‐derived polyphenolic with antiviral effects in mice after IP delivery (181)
Cannabis Oral Δ9‐tetrahydrocannabinol increased virus loads and decreased inflammatory responses in mice (182)
Resveratrol Antiviral effects and increased survival of mice after IP delivery (183)
Slit2N IV treatment reduces endothelial hyper‐permeability and mortality after H5N1 infection in mice (184).

IN, intranasal; IP, intraperitoneal; SC, subcutaneous; IV, intravenous.

Future directions

The following section provides a highly selected commentary on novel approaches for developing more effective influenza therapeutics. The reader is referred to the many recent reviews regarding compounds in pre‐clinical development for established and alternative (e.g. polymerase, nucleoprotein) targets. 6 , 7 , 8 , 9 , 10 , 12 , 16 , 17

RNA inhibition

There have been a number of interesting preclinical reports regarding antisense strategies and the use of siRNAs for treating influenza 93 and other respiratory viruses. 94 One that has moved forward clinically is AVI‐7100, a phosphorodiamidate morpholino oligomer containing three modified linkages (PMOplus) that is designed to interfere with the translation of both the M1 and M2 mRNAs of influenza A virus (AVI Biopharma Inc, Bothell, WA, USA). These two proteins are products of splice variants from the same genome segment and share the same translation initiation start site which is targeted by this oligomer. The unique backbone structure allows for better delivery of the antisense oligomer to infected cells, and the molecule has been shown to have good activity against influenza A viruses in both cell culture and in animal model studies. In a ferret model of oseltamivir‐resistant A(H1N1)pdm09 virus infection, this antisense molecule given either intraperitoneally or intranasally was associated with significant antiviral effects in terms of reduced nasal and BAL virus loads and lesser illness. 95 Intravenous administration of this molecule is now being examined in a dose‐ranging phase 1 study. Depending on subsequent findings, this agent might eventually be an option for use in combination with other parenteral agents in more seriously ill patients.

Cellular targets

Another area of active investigation is the interaction between influenza virus and various host cell factors, at both the RNA and protein levels, to identify host cellular pathways essential for virus replication that might be amenable to inhibition, as a basis for treatment of acute infection. 17 This approach of host‐directed therapies has also been promoted because of the very low likelihood of resistance emergence and its potential applicability to multiple respiratory viruses.

Influenza infection results in the activation of various intracellular signalling responses, some of which the virus uses to ensure efficient replication. Two particular pathways have been established as suitable targets for inhibition of virus replication in murine models: the Raf/MEK/ERK mitogenic kinase cascade (involved in nuclear export of viral RNPs) and the IKK/NF‐κB module, the activation of which affects both several steps in replication and host innate immune responses. 12 Topical application of acetylsalicylic acid (aspirin), an inhibitor of IKK2, showed antiviral effects in mice, 96 although systemic aspirin was associated with increased mortality in several influenza animal models. 97

Recently, multiple groups using different RNAi genome‐wide screening systems have published on the complexity of these interactions and identified possible targets in influenza, as well as several candidate inhibitors. 98 , 99 , 100 , 101 Integrated analysis of five screens determined that 85 cellular factors were identified in two or more of the influenza virus screens, of which 50 were considered to have druggable properties and 34 were also needed for influenza replication in vitro. 102 , 103 In particular, these analyses found that the vATPase and COPI complexes, the ribosomal mRNA splicing and nuclear trafficking machinery, and kinase‐regulated signalling are all required for efficient replication of influenza A virus. Such cross‐comparisons at the pathway level rather than the gene level reveal more common features that might provide potential targets for antiviral drug development, either influenza‐specific or broader in spectrum. 104 Of course, even in the context of short‐term inhibitor administration, the targeting of cellular functions raises important tolerability concerns that will require careful safety studies in key patient populations.

Adjunctive therapies

Another area of considerable interest has been adjunctive treatments for influenza, primarily those directed against excessive pro‐inflammatory host responses to infection. 105 Animal model studies have identified a wide range of agents with apparent beneficial effects (Table 3), but there are few for which clinical data have been developed. Depending on the particular model, agents with either pro‐ 106 or anti‐inflammatory effects (Table 3) have been reported as showing benefit. This in part relates to the complexity of host responses leading to acute lung injury and differences among model systems. 107

The use of currently available drugs with immunomodulatory activity, well‐characterised safety profiles, and low production costs has been promoted as a possible treatment strategy. 108 Some epidemiologic studies have reported substantial mortality benefits in patients taking statins who were subsequently hospitalised for influenza 109 or pneumonia, 110 but the results are not consistent across studies. 111 The possible benefit of starting such drugs at the time of influenza onset or hospitalisation have not been reported to date, although one ICU‐based, open‐label RCT suggested reduced risks of ventilator‐associated pneumonia and mortality associated with the addition of pravastatin therapy in patients requiring mechanical ventilation. 112 In contrast, while systemic glucocorticoids have been frequently used for treating influenza pneumonia and associated ARDS, studies from the 2009 A(H1N1) pandemic have found that glucocorticoids in such patients were associated with prolongation of virus replication, increases in secondary bacterial and fungal infections and higher rates of mortality in ICU patients. 113 , 114 , 115 , 116 , 117 Consequently, one needs to be very cautious in terms of the particular immunomodulatory intervention, its therapeutic potency and its timing of use in relation to the type and course of respiratory illness. For example, studies of severe or fatal influenza viral pneumonia during seasonal 118 and pandemic 2009 119 outbreaks have found evidence for deficiency in interferon responses that appear key to controlling virus replication. Consequently, the possibility of using immunomodulatory interventions will need to consider the particular target population and goal of either suppressing adverse host responses or supplementing deficient ones, such that clinical trials will be challenging.

Summary

In conclusion, it is clear that medical needs exist for more effective therapies for severe influenza, particularly in those who are hospitalised and in immunocompromised hosts. Considerable progress has been made in the clinical development of intravenous NAIs and to an increasing extent other novel antivirals and biotherapeutics for influenza management. In addition to optimisation of dosing regimens of existing drugs, combination therapies offer great promise going forward. Selective immunomodulatory interventions, in conjunction with antivirals to control replication, are another promising area for investigation, but the particular type(s) and timing of intervention need to be based on a better understanding of disease pathogenesis. Detailed pathogenesis studies to improve understanding of the relationships between virologic measures, biomarkers and clinical outcomes are needed, as are strategies for linking these findings to inform improved therapeutic monitoring approaches, particularly in seriously ill patients. In addition, the study of combination interventions, immunomodulators and host‐directed therapies presents unique regulatory hurdles, 18 , 19 and the pathways to efficient study and eventual marketing of such interventions require clarification.

Conflicts of interest

From 2008 to present, Dr. Hayden has been an unpaid consultant to multiple companies engaged in the development or marketing of influenza antivirals, including AVI Biopharma, Biocryst, Crucell, GSK, Nexbio, Roche, Romark, Toyama and Visterra whose agents are included in this review. Since 2008 to present, the University of Virginia has received funding from the Wellcome Trust for his part‐time work as influenza research coordinator at the Trust and from SAIC on behalf of the NIAID for his work as consultant the Southeast Asia Infectious Diseases Clinical Research Network. From 2008 to 2011, the University also received honoraria for his participation in the Neuraminidase Inhibitor Susceptibility Network which received funding from Roche and GSK.

Acknowledgements

I would like to thank Dr. Janet Englund, Seattle Children’s Hospital and colleagues from AVI Biopharma, Nexbio, Romark, and Toyama for their willingness to share unpublished data and Lisa Cook for her assistance in manuscript preparation.

References

  • 1. Hayden FG. Antivirals for influenza: historical perspectives and lessons learned. Antiviral Res 2006; 71:372–378. [DOI] [PubMed] [Google Scholar]
  • 2. Ong AK, Hayden FG, John F. Enders lecture 2006: antivirals for influenza. J Infect Dis 2007; 196:181–190. [DOI] [PubMed] [Google Scholar]
  • 3. Fischer W, Hayden F. Chapter 18. Antivirals for influenza: novel agents and approaches; in Georgiev V, Western K, McGowan J, (eds): National Institute of Allergy and Infectious Diseases, NIH. Totowa NJ: Frontiers in Research Humana Press; 2008:179–192. [Google Scholar]
  • 4. Hayden F. Developing new antiviral agents for influenza treatment: what does the future hold? Clin Infect Dis 2009; 48(Suppl 1):S3–S13. [DOI] [PubMed] [Google Scholar]
  • 5. Hayden FG. Influenza antivirals: challenges and future directions. Influenza Other Respir Viruses 2011; 5(Suppl. 1):20–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Krug RM, Aramini JM. Emerging antiviral targets for influenza A virus. Trends Pharmacol Sci 2009; 30:269–277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Beigel J, Bray M. Current and future antiviral therapy of severe seasonal and avian influenza. Antiviral Res 2008; 78:91–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Hsieh HP, Hsu JT. Strategies of development of antiviral agents directed against influenza virus replication. Curr Pharm Des 2007; 13:3531–3542. [DOI] [PubMed] [Google Scholar]
  • 9. Saladino R, Barontini M, Crucianelli M, Nencioni L, Sgarbanti R, Palamara AT. Current advances in anti‐influenza therapy. Curr Med Chem 2010; 17:2101–2140. [DOI] [PubMed] [Google Scholar]
  • 10. Boltz DA, Aldridge JR Jr, Webster RG, Govorkova EA. Drugs in development for influenza. Drugs 2010; 70:1349–1362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Ison MG. Antivirals and resistance: influenza virus. Curr Opin Virol 2011; 1:563–573. [DOI] [PubMed] [Google Scholar]
  • 12. Ludwig S. Disruption of virus‐host cell interactions and cell signaling pathways as an anti‐viral approach against influenza virus infections. Biol Chem 2011; 392:837–847. [DOI] [PubMed] [Google Scholar]
  • 13. Hayden FG. Antiviral resistance in influenza viruses: clinical and epidemiological aspects in: Mayers DL, (ed.): Antimicrobial Drug Resistance. New York, New York: Humana Press; 2009: 1011–1033. [Google Scholar]
  • 14. Hayden FG, de Jong MD. Emerging influenza antiviral resistance threats. J Infect Dis 2011; 1:6–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Hurt AC, Chotpitayasunondh T, Cox NJ et al. Antiviral resistance during the 2009 influenza A H1N1 pandemic: public health, laboratory, and clinical perspectives. Lancet Infect Dis 2011; 12:240–248. [DOI] [PubMed] [Google Scholar]
  • 16. NIAID Influenza Antiviral Development Workshop: New Generation. Available from: http://www.niaid.nih.gov/topics/Flu/Documents/fluantiviral09.pdf (Accessed on 26 March 2009).
  • 17. NIAID 2011 Influenza Antiviral Research Pipeline Workshop. Available from: http://www.niaid.nih.gov/about/organization/dmid/meetings/Documents/fluantiviralwkshop2011.pdf (Accessed on 23 March 2011).
  • 18. Guidance for industry influenza: developing drugs for treatment and/or prophylaxis. Available from: http://www.fda.gov/Drugs/guidanceComplianceRegulatoryInformation/Guidances/default.htm (Acce‐ssed on 1 April 2011).
  • 19. Ison MG, MD dJ, Gilligan KJ et al. End points for testing influenza antiviral treatments for patients at high risk of severe and life‐threatening disease. J Infect Dis 2010; 201:1654–1662. [DOI] [PubMed] [Google Scholar]
  • 20. Writing Committee of the WHO Consultation on Clinical Aspects of Pandemic (H1N1) 2009 Influenza . Clinical aspects of pandemic 2009 influenza A (H1N1) virus infection. N Engl J Med 2010;362:1708–1719. (See Supplementary Materials) [DOI] [PubMed] [Google Scholar]
  • 21. Yamashita M, Tomozawa T, Kakuta M, Tokumitsu A, Nasu H, Kubo S. CS‐8958, a prodrug of the new neuraminidase inhibitor R‐125489, shows long‐acting anti‐influenza virus activity. Antimicrob Agents Chemother 2009; 53:186–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Abed Y, Simon P, Boivin G. Prophylactic activity of intramuscular peramivir in mice infected with a recombinant influenza A/WSN/33 (H1N1) virus containing the H274Y neuraminidase mutation. Antimicrob Agents Chemother 2010; 54:2819–2822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Renaud C, Pergam SA, Polyak C et al. Early emergence of an H275Y mutation in a hematopoietic cell transplant recipient treated with intravenous peramivir. Transpl Infect Dis 2010; 12:513–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Memoli MJ, Hrabal RJ, Hassantoufighi A, Eichelberger MC, Taubenberger JK. Rapid selection of oseltamivir‐ and peramivir‐resistant pandemic H1N1 virus during therapy in 2 immunocompromised hosts. Clin Infect Dis 2010; 50:1252–1255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Kohno S, Kida H, Mizuguchi M, Shimada J. Efficacy and safety of intravenous peramivir for treatment of seasonal influenza virus infection. Antimicrob Agents Chemother 2010; 54:4568–4574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Kohno S, Yen MY, Cheong HJ et al. Phase III randomized, double‐blind study comparing single‐dose intravenous peramivir with oral oseltamivir in patients with seasonal influenza virus infection. Antimicrob Agents Chemother 2011; 55:5267–5276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Ison M, Hui D, Flynt A, Collis P, Alexander J, Hernandez E. No evidence of resistance in influenza viruses after 5‐day IV peramivir therapy in hospitalized patients. ICAAC 2011, V‐1541. 2011.
  • 28. Kohno S, Kida H, Mizuguchi M et al. Intravenous peramivir for treatment of influenza A and B virus infection in high‐risk patients. Antimicrob Agents Chemother 2011; 55:2803–2812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Hernandez JE, Adiga R, Armstrong R et al. Clinical experience in adults and children treated with intravenous peramivir for 2009 influenza A (H1N1) under an Emergency IND program in the United States. Clin Infect Dis 2011; 52:695–706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Fraaij PL, van der Vries E, Beersma MF et al. Evaluation of the anti‐viral response to zanamivir administered intravenously for treatment of critically ill patients with pandemic influenza A (H1N1) infection. J Infect Dis 2011; 204:777–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Gaur AH, Bagga B, Barman S et al. Intravenous zanamivir for oseltamivir‐resistant 2009 H1N1 influenza. N Engl J Med 2010; 362:88–89. [DOI] [PubMed] [Google Scholar]
  • 32. Dulek DE, Williams JV, Creech CB et al. Use of intravenous zanamivir after development of oseltamivir resistance in a critically Ill immunosuppressed child infected with 2009 pandemic influenza A (H1N1) virus. Clin Infect Dis 2010; 50:1493–1496. [DOI] [PubMed] [Google Scholar]
  • 33. Yates PJ, Man CY, Zhao H. Interim virological analysis of a prospective single arm phase II study of intravenous zanamivir for the treatment of hospitalized patients with influenza A/H1N1 2009 infection [abstract P‐160]. In: Program and abstracts of the International Society for Influenza and other Respiratory Virus Diseases Hong Kong SAR, China: Options for the Control of Influenza VII, 2010:139. Options for the Control of Influenza VII Abstract Book. 9‐3‐0010.
  • 34. Kubo S, Tomozawa T, Kakuta M, Tokumitsu A, Yamashita M. Laninamivir prodrug CS‐8958, a long‐acting neuraminidase inhibitor, shows superior anti‐influenza virus activity after a single administration. Antimicrob Agents Chemother 2010; 54:1256–1264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Ishizuka H, Yoshiba S, Okabe H, Yoshihara K. Clinical pharmacokinetics of laninamivir, a novel long‐acting neuraminidase inhibitor, after single and multiple inhaled doses of its prodrug, CS‐8958, in healthy male volunteers. J Clin Pharmacol 2010; 50:1319–1329. [DOI] [PubMed] [Google Scholar]
  • 36. Sugaya N, Ohashi Y. Long‐acting neuraminidase inhibitor laninamivir octanoate (CS‐8958) versus oseltamivir as treatment for children with influenza virus infection. Antimicrob Agents Chemother 2010; 54:2575–2582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Watanabe A, Chang SC, Kim MJ, Chu DW, Ohashi Y. Long‐acting neuraminidase inhibitor laninamivir octanoate versus oseltamivir for treatment of influenza: a double‐blind, randomized, noninferiority clinical trial. Clin Infect Dis 2010; 51:1167–1175. [DOI] [PubMed] [Google Scholar]
  • 38. Macdonald SJ, Watson KG, Cameron R et al. Potent and long‐acting dimeric inhibitors of influenza virus neuraminidase are effective at a once‐weekly dosing regimen. Antimicrob Agents Chemother 2004; 48:4542–4549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Macdonald SJ, Cameron R, Demaine DA et al. Dimeric zanamivir conjugates with various linking groups are potent, long‐lasting inhibitors of influenza neuraminidase including H5N1 avian influenza. J Med Chem 2005; 48:2964–2971. [DOI] [PubMed] [Google Scholar]
  • 40. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Triana‐Baltzer GB, Sanders RL, Hedlund M et al. Phenotypic and genotypic characterization of influenza virus mutants selected with the sialidase fusion protein DAS181. J Antimicrob Chemother 2011; 66:15–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. 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. [DOI] [PubMed] [Google Scholar]
  • 43. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Chen YB, Driscoll JP, McAfee SL et al. Treatment of parainfluenza 3 infection with DAS181 in a patient after allogeneic stem cell transplantation. Clin Infect Dis 2011; 53:e77–e80. [DOI] [PubMed] [Google Scholar]
  • 46. Guzman‐Suarez BB, Buckley MW, Gilmore ET et al. Clinical potential of DAS181 for treatment of parainfluenza‐3 infections in transplant recipients. Transpl Infect Dis 2012. doi: 10.1111/j.1399-3062.2012.00718.x. [DOI] [PubMed] [Google Scholar]
  • 47. Moss R, Steigbigel R, Wurtman D, Hansen C, Tranel K. A Phase 2A dose‐ranging clinical trial of DAS181 for treatment of influenza in healthy adults. Presented at the 51st Interscience Conference on Antimicrobial Agents and Chemotherapy (ICAAC) Abstract V‐4052. 9‐17‐2011.
  • 48. Sleeman K, Mishin VP, Deyde VM, Furuta Y, Klimov AI, Gubareva LV. In vitro antiviral activity of favipiravir (T‐705) against drug‐resistant influenza and 2009 A(H1N1) viruses. Antimicrob Agents Chemother 2010; 54:2517–2524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Kiso M, Takahashi K, Sakai‐Tagawa Y et al. T‐705 (favipiravir) activity against lethal H5N1 influenza A viruses. Proc Natl Acad Sci U S A 2010; 107:882–887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Furuta Y, Takahashi K, Shiraki K et al. T‐705 (favipiravir) and related compounds: novel broad‐spectrum inhibitors of RNA viral infections. Antiviral Res 2009; 82:95–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Smee DF, Hurst BL, Wong MH et al. Effects of the combination of favipiravir (T‐705) and oseltamivir on influenza A virus infections in mice. Antimicrob Agents Chemother 2010; 54:126–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Kobayashi O, Kashiwagi S, Iwamoto A. Clinical effectiveness and safety of favipiravir, a novel anti‐influenza drug with a selective inhibition activity against viral RNA polymerase. Presented at the 51st Interscience Conference on Antimicrobial Agents and Chemotherapy (ICAAC) Chicago, V‐405. 9‐17‐2011.
  • 53. Rossignol JF, La FS, Chiappa L, Ciucci A, Santoro MG. Thiazolides, a new class of anti‐influenza molecules targeting viral hemagglutinin at the post‐translational level. J Biol Chem 2009; 284:29798–29808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Rossignol JF, Samudrals S, Hoppers M, Haffizulla J. A randomized, double‐blind, placebo (PCB) controlled study of nitazoxanide (NTZ) in adults and adolescents with acute uncomplicated influenza. Presented at the IDSA Annual Meeting, Boston. LB‐35. 10‐22‐2011.
  • 55. Luke TC, Kilbane EM, Jackson JL, Hoffman SL. Meta‐analysis: convalescent blood products for Spanish influenza pneumonia: a future H5N1 treatment? Ann Intern Med 2006; 145:599–609. [DOI] [PubMed] [Google Scholar]
  • 56. Zhou B, Zhong N, Guan Y. Treatment with convalescent plasma for influenza A (H5N1) infection. N Engl J Med 2007; 357:1450–1451. [DOI] [PubMed] [Google Scholar]
  • 57. Hung IF, To KK, Lee CK et al. Convalescent plasma treatment reduced mortality in patients with severe pandemic influenza A (H1N1) 2009 virus infection. Clin Infect Dis 2011; 52:447–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Friesen RH, Koudstaal W, Koldijk MH et al. New class of monoclonal antibodies against severe influenza: prophylactic and therapeutic efficacy in ferrets. PLoS One 2010; 5:e9106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Sui J, Hwang WC, Perez S et al. Structural and functional bases for broad‐spectrum neutralization of avian and human influenza A viruses. Nat Struct Mol Biol 2009; 16:265–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Ekiert DC, Bhabha G, Elsliger MA et al. Antibody recognition of a highly conserved influenza virus epitope. Science 2009; 324:246–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Simmons CP, Bernasconi NL, Suguitan AL et al. Prophylactic and therapeutic efficacy of human monoclonal antibodies against H5N1 influenza. PLoS Med 2007; 4:e178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Throsby M, van denBE, Jongeneelen M et al. Heterosubtypic neutralizing monoclonal antibodies cross‐protective against H5N1 and H1N1 recovered from human IgM+ memory B cells. PLoS One 2008; 3:e3942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Ekiert DC, Friesen RH, Bhabha G et al. A highly conserved neutralizing epitope on group 2 influenza A viruses. Science 2011; 333:843–850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Corti D, Voss J, Gamblin SJ et al. A neutralizing antibody selected from plasma cells that binds to group 1 and group 2 influenza A hemagglutinins. Science 2011; 333:850–856. [DOI] [PubMed] [Google Scholar]
  • 65. Wang R, Song A, Levin J et al. Therapeutic potential of a fully human monoclonal antibody against influenza A virus M2 protein. Antiviral Res 2008; 80:168–177. [DOI] [PubMed] [Google Scholar]
  • 66. Grandea AG III, Olsen OA, Cox TC et al. Human antibodies reveal a protective epitope that is highly conserved among human and nonhuman influenza A viruses. Proc Natl Acad Sci U S A 2010; 107:12658–12663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. El BK, Descamps F, De FM et al. Universal vaccine based on ectodomain of matrix protein 2 of influenza A: Fc receptors and alveolar macrophages mediate protection. J Immunol 2011; 186:1022–1031. [DOI] [PubMed] [Google Scholar]
  • 68. Sandbulte MR, Jimenez GS, Boon AC, Smith LR, Treanor JJ, Webby RJ. Cross‐reactive neuraminidase antibodies afford partial protection against H5N1 in mice and are present in unexposed humans. PLoS Med 2007; 4:e59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Carragher DM, Kaminski DA, Moquin A, Hartson L, Randall TD. A novel role for non‐neutralizing antibodies against nucleoprotein in facilitating resistance to influenza virus. J Immunol 2008; 181:4168–4176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Fleishman SJ, Whitehead TA, Ekiert DC et al. Computational design of proteins targeting the conserved stem region of influenza hemagglutinin. Science 2011; 332:816–821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Whitehead TA, Chevalier A, Song Y et al. Optimization of affinity, specificity and function of designed influenza inhibitors using deep sequencing. Nat Biotechnol 2012; 30:543–548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Badani H, Garry R, Wilson R, Wimley C. Mechanism and action of flufirvitide, a peptide inhibitor of influenza virus infection. Biophysical 2011; 100:216a. [Google Scholar]
  • 73. Autoimmune Technologies . LLC 12 A.D. August 3. http://autoimmune.com/Flufirvitide.html .
  • 74. Lavrov SV, Eremkina EI, Orlova TG, Galegov GA, Soloviev VD, Zhdanov VM. Combined inhibition of influenza virus reproduction in cell culture using interferon and amantadine. Nature 1968; 217:856–857. [DOI] [PubMed] [Google Scholar]
  • 75. Hayden FG, Schlepushkin AN, Pushkarskaya NL. Combined interferon‐alpha 2, rimantadine hydrochloride, and ribavirin inhibition of influenza virus replication in vitro. Antimicrob Agents Chemother 1984; 25:53–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Ilyushina NA, Bovin NV, Webster RG, Govorkova EA. Combination chemotherapy, a potential strategy for reducing the emergence of drug‐resistant influenza A variants. Antiviral Res 2006; 70:121–131. [DOI] [PubMed] [Google Scholar]
  • 77. Ilyushina NA, Hoffmann E, Salomon R, Webster RG, Govorkova EA. Amantadine‐oseltamivir combination therapy for H5N1 influenza virus infection in mice. Antivir Ther 2007; 12:363–370. [PubMed] [Google Scholar]
  • 78. Smee DF, Hurst BL, Wong MH, Bailey KW, Morrey JD. Effects of double combinations of amantadine, oseltamivir, and ribavirin on influenza A (H5N1) virus infections in cell culture and in mice. Antimicrob Agents Chemother 2009; 53:2120–2128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Smee DF, Wong MH, Bailey KW, Sidwell RW. Activities of oseltamivir and ribavirin used alone and in combination against infections in mice with recent isolates of influenza A (H1N1) and B viruses. Antivir Chem Chemother 2006; 17:185–192. [DOI] [PubMed] [Google Scholar]
  • 80. Ilyushina NA, Hay A, Yilmaz N, Boon AC, Webster RG, Govorkova EA. Oseltamivir‐ribavirin combination therapy for highly pathogenic H5N1 influenza virus infection in mice. Antimicrob Agents Chemother 2008; 52:3889–3897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Nguyen JT, Hoopes JD, Le MH et al. Triple combination of amantadine, ribavirin, and oseltamivir is highly active and synergistic against drug resistant influenza virus strains in vitro. PLoS One 2010; 5:e9332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Smee DF, Hurst BL, Wong MH et al. Combinations of oseltamivir and peramivir for the treatment of influenza A (H1N1) virus infections in cell culture and in mice. Antiviral Res 2010; 88:38–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Atiee G, Lasseter K, Baughman S et al. Absence of pharmacokinetic interaction between intravenous peramivir and oral oseltamivir or rimantadine in humans. J Clin Pharmacol 2011; 52:1410–1419. [DOI] [PubMed] [Google Scholar]
  • 84. Morrison D, Roy S, Rayner C et al. A randomized, crossover study to evaluate the pharmacokinetics of amantadine and oseltamivir administered alone and in combination. PLoS One 2007; 2:e1305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Pukrittayakamee S, Jittamala P, Stepniewska K et al. An open‐label crossover study to evaluate potential pharmacokinetic interactions between oral oseltamivir and intravenous zanamivir in healthy thai adults. Antimicrob Agents Chemother 2011; 55:4050–4057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Ison MG, Gnann JW Jr, Nagy‐Agren S et al. Safety and efficacy of nebulized zanamivir in hospitalized patients with serious influenza. Antivir Ther 2003; 8:183–190. [PubMed] [Google Scholar]
  • 87. Duval X, van derWS, Blanchon T et al. Efficacy of oseltamivir‐zanamivir combination compared to each monotherapy for seasonal influenza: a randomized placebo‐controlled trial. PLoS Med 2010; 7:e1000362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Nguyen JT, Hoopes JD, Smee DF et al. Triple combination of oseltamivir, amantadine, and ribavirin displays synergistic activity against multiple influenza virus strains in vitro. Antimicrob Agents Chemother 2009; 53:4115–4126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Hoopes JD, Driebe EM, Kelley E et al. Triple combination antiviral drug (TCAD) composed of amantadine, oseltamivir, and ribavirin impedes the selection of drug‐resistant influenza A virus. PLoS One 2011; 6:e29778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Nguyen JT, Smee DF, Barnard DL et al. Efficacy of combined therapy with amantadine, oseltamivir, and ribavirin in vivo against susceptible and amantadine‐resistant influenza A viruses. PLoS One 2012; 7:e31006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Kim WY, Young SG, Huh JW et al. Triple‐combination antiviral drug for pandemic H1N1 influenza virus infection in critically ill patients on mechanical ventilation. Antimicrob Agents Chemother 2011; 55:5703–5709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Walsh KB, Teijaro JR, Wilker PR et al. Suppression of cytokine storm with a sphingosine analog provides protection against pathogenic influenza virus. Proc Natl Acad Sci U S A 2011; 108:12018–12023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Zhou H, Jin M, Yu Z et al. Effective small interfering RNAs targeting matrix and nucleocapsid protein gene inhibit influenza A virus replication in cells and mice. Antiviral Res 2007; 76:186–193. [DOI] [PubMed] [Google Scholar]
  • 94. Devincenzo JP. The promise, pitfalls and progress of RNA‐interference‐based antiviral therapy for respiratory viruses. Antivir Ther 2012;17 (1 Pt B):213–225. [DOI] [PubMed] [Google Scholar]
  • 95. Iversen PL, Mourich DV, Voss T. AVI‐7100 is effective in oseltamivir resistant H1N1 infected ferrets. Presented at the 51st Interscience Conference on Antimicrobial Agents and Chemotherapy (ICAAC) Chicago 9‐17‐2011, Abstract F1‐13725a.
  • 96. Mazur I, Wurzer WJ, Ehrhardt C et al. Acetylsalicylic acid (ASA) blocks influenza virus propagation via its NF‐kappaB‐inhibiting activity. Cell Microbiol 2007; 9:1683–1694. [DOI] [PubMed] [Google Scholar]
  • 97. Eyers S, Weatherall M, Shirtcliffe P, Perrin K, Beasley R. The effect on mortality of antipyretics in the treatment of influenza infection: systematic review and meta‐analysis. J R Soc Med 2010; 103:403–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Hao L, Sakurai A, Watanabe T et al. Drosophila RNAi screen identifies host genes important for influenza virus replication. Nature 2008; 454:890–893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Karlas A, Machuy N, Shin Y et al. Genome‐wide RNAi screen identifies human host factors crucial for influenza virus replication. Nature 2010; 463:818–822. [DOI] [PubMed] [Google Scholar]
  • 100. Brass AL, Huang IC, Benita Y et al. The IFITM proteins mediate cellular resistance to influenza A H1N1 virus, West Nile virus, and dengue virus. Cell 2009; 139:1243–1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Josset L, Textoris J, Loriod B et al. Gene expression signature‐based screening identifies new broadly effective influenza A antivirals. PLoS One 2010; 5:pii:e13169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Stertz S, Shaw ML. Uncovering the global host cell requirements for influenza virus replication via RNAi screening. Microbes Infect 2011; 13:516–525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Shaw ML. The host interactome of influenza virus presents new potential targets for antiviral drugs. Rev Med Virol 2011; 21:358–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Smith SB, Dampier W, Tozeren A, Brown JR, Magid‐Slav M. Identification of common biological pathways and drug targets across multiple respiratory viruses based on human host gene expression analysis. PLoS One 2012; 7:e33174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Fedson DS. Confronting the next influenza pandemic with anti‐inflammatory and immunomodulatory agents: why they are needed and how they might work. Influenza Other Respi Viruses 2009; 3:129–142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Tuvim MJ, Evans SE, Clement CG, Dickey BF, Gilbert BE. Augmented lung inflammation protects against influenza A pneumonia. PLoS One 2009; 4:e4176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Howard WA, Peiris M, Hayden FG. Report of the ‘mechanisms of lung injury and immunomodulator interventions in influenza’ workshop, 21 March 2010, Ventura, California, USA. Influenza Other Respi Viruses 2011; 5:453–475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Fedson DS. Confronting an influenza pandemic with inexpensive generic agents: can it be done?. Lancet Infect Dis 2008; 8:571–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Vandermeer ML, Thomas AR, Kamimoto L et al. Association between use of statins and mortality among patients hospitalized with laboratory‐confirmed influenza virus infections: a multistate study. J Infect Dis 2012; 205:13–19. [DOI] [PubMed] [Google Scholar]
  • 110. Douglas I, Evans S, Smeeth L. Effect of statin treatment on short term mortality after pneumonia episode: cohort study. BMJ 2011; 342:d1642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Brett SJ, Myles P, Lim WS et al. Pre‐admission statin use and in‐hospital severity of 2009 pandemic influenza A(H1N1) disease. PLoS One 2011; 6:e18120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Makris D, Manoulakas E, Komnos A et al. Effect of pravastatin on the frequency of ventilator‐associated pneumonia and on intensive care unit mortality: open‐label, randomized study. Crit Care Med 2011; 39:2440–2446. [DOI] [PubMed] [Google Scholar]
  • 113. Xi X, Xu Y, Jiang L, Li A, Duan J, Du B. Hospitalized adult patients with 2009 influenza A(H1N1) in Beijing, China: risk factors for hospital mortality. BMC Infect Dis 2010; 10:256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Martin‐Loeches I, Lisboa T, Rhodes A et al. Use of early corticosteroid therapy on ICU admission in patients affected by severe pandemic (H1N1)v influenza A infection. Intensive Care Med 2011; 37:272–283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Lat A, Bhadelia N, Miko B, Furuya EY, Thompson GR III. Invasive aspergillosis after pandemic (H1N1) 2009. Emerg Infect Dis 2010; 16:971–973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Brun‐Buisson C, Richard JC, Mercat A, Thiebaut AC, Brochard L. Early corticosteroids in severe influenza A/H1N1 pneumonia and acute respiratory distress syndrome. Am J Respir Crit Care Med 2011; 183:1200–1206. [DOI] [PubMed] [Google Scholar]
  • 117. Kim SH, Hong SB, Yun SC et al. Corticosteroid treatment in critically ill patients with pandemic influenza A/H1N1 2009 infection: analytic strategy using propensity scores. Am J Respir Crit Care Med 2011; 183:1207–1214. [DOI] [PubMed] [Google Scholar]
  • 118. Baron S, ISAACS A. Absence of interferon in lungs fom fatal cases of influenza. Br Med J 1962; 1:18–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Agrati C, Gioia C, Lalle E et al. Association of profoundly impaired immune competence in H1N1v‐infected patients with a severe or fatal clinical course. J Infect Dis 2010; 202:681–689. [DOI] [PubMed] [Google Scholar]
  • 120. Ison MG, Mishin VP, Braciale TJ, Hayden FG, Gubareva LV. Comparative activities of oseltamivir and A‐322278 in immunocompetent and immunocompromised murine models of influenza virus infection. J Infect Dis 2006; 193:765–772. [DOI] [PubMed] [Google Scholar]
  • 121. Abed Y, Nehme B, Baz M, Boivin G. Activity of the neuraminidase inhibitor A‐315675 against oseltamivir‐resistant influenza neuraminidases of N1 and N2 subtypes. Antiviral Res 2008; 77:163–166. [DOI] [PubMed] [Google Scholar]
  • 122. Zhirnov OP, Klenk HD, Wright PF. Aprotinin and similar protease inhibitors as drugs against influenza. Antiviral Res 2011; 92:27–36. [DOI] [PubMed] [Google Scholar]
  • 123. Nicol MQ, Ligertwood Y, Bacon MN, Dutia BM, Nash AA. A novel family of peptides with potent activity against influenza A viruses. J Gen Virol 2012;93 (Pt 5):980–986. [DOI] [PubMed] [Google Scholar]
  • 124. Jones JC, Turpin EA, Bultmann H, Brandt CR, Schultz‐Cherry S. Inhibition of influenza virus infection by a novel antiviral peptide that targets viral attachment to cells. J Virol 2006; 80:11960–11967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Shi L, Xiong H, He J et al. Antiviral activity of arbidol against influenza A virus, respiratory syncytial virus, rhinovirus, coxsackie virus and adenovirus in vitro and in vivo. Arch Virol 2007; 152:1447–1455. [DOI] [PubMed] [Google Scholar]
  • 126. Boriskin YS, Leneva IA, Pecheur EI, Polyak SJ. Arbidol: a broad‐spectrum antiviral compound that blocks viral fusion. Curr Med Chem 2008; 15:997–1005. [DOI] [PubMed] [Google Scholar]
  • 127. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Leibbrandt A, Meier C, Konig‐Schuster M et al. Iota‐carrageenan is a potent inhibitor of influenza A virus infection. PLoS One 2010; 5:e14320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Reading PC, Bozza S, Gilbertson B et al. Antiviral activity of the long chain pentraxin PTX3 against influenza viruses. J Immunol 2008; 180:3391–3398. [DOI] [PubMed] [Google Scholar]
  • 130. Gambaryan AS, Boravleva EY, Matrosovich TY et al. Polymer‐bound 6′ sialyl‐N‐acetyllactosamine protects mice infected by influenza virus. Antiviral Res 2005; 68:116–123. [DOI] [PubMed] [Google Scholar]
  • 131. Droebner K, Ehrhardt C, Poetter A, Ludwig S, Planz O. CYSTUS052, a polyphenol‐rich plant extract, exerts anti‐influenza virus activity in mice. Antiviral Res 2007; 76:1–10. [DOI] [PubMed] [Google Scholar]
  • 132. Mei‐Lin Yang M‐L, Chen Y‐H, Wang S‐W et al. Galectin‐1 binds to influenza virus and ameliorates influenza virus pathogenesis. J Virology 2011; 85:10010–10020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Sidwell RW, Bailey KW, Wong MH, Barnard DL, Smee DF. In vitro and in vivo influenza virus‐inhibitory effects of viramidine. Antiviral Res 2005; 68:10–17. [DOI] [PubMed] [Google Scholar]
  • 134. Zhang T, Zhao PS, Zhang W et al. Antisense oligonucleotide inhibits avian influenza virus H5N1 replication by single chain antibody delivery system. Vaccine 2011; 29:1558–1564. [DOI] [PubMed] [Google Scholar]
  • 135. Mizuta T, Fujiwara M, Abe T et al. Inhibitory effects of an antisense oligonucleotide in an experimentally infected mouse model of influenza A virus. Biochem Biophys Res Commun 2000; 279:158–161. [DOI] [PubMed] [Google Scholar]
  • 136. Su CY, Cheng TJ, Lin MI et al. High‐throughput identification of compounds targeting influenza RNA‐dependent RNA polymerase activity. Proc Natl Acad Sci U S A 2010; 107:19151–19156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Kao RY, Yang D, Lau LS et al. Identification of influenza A nucleoprotein as an antiviral target. Nat Biotechnol 2010; 28:600–605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Zhang T, Wang TC, Zhao PS et al. Antisense oligonucleotides targeting the RNA binding region of the NP gene inhibit replication of highly pathogenic avian influenza virus H5N1. Int Immunopharmacol 2011; 11:2057–2061. [DOI] [PubMed] [Google Scholar]
  • 139. Szretter KJ, Gangappa S, Belser JA et al. Early control of H5N1 influenza virus replication by the type I interferon response in mice. J Virol 2009; 83:5825–5834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Kugel D, Kochs G, Obojes K et al. Intranasal administration of alpha interferon reduces seasonal influenza A virus morbidity in ferrets. J Virol 2009; 83:3843–3851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Weiss ID, Wald O, Wald H et al. IFN‐gamma treatment at early stages of influenza virus infection protects mice from death in a NK cell‐dependent manner. J Interferon Cytokine Res 2010; 30:439–449. [DOI] [PubMed] [Google Scholar]
  • 142. Xu C, Song X, Fu L et al. Antiviral potential of exogenous human omega interferon to inhibit pandemic 2009 A (H1N1) influenza virus. Viral Immunol 2011; 24:369–374. [DOI] [PubMed] [Google Scholar]
  • 143. Van HN, Belser JA, Szretter KJ et al. Pathogenesis of 1918 pandemic and H5N1 influenza virus infections in a guinea pig model: antiviral potential of exogenous alpha interferon to reduce virus shedding. J Virol 2009; 83:2851–2861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Wong JP, Christopher ME, Viswanathan S et al. Activation of toll‐like receptor signaling pathway for protection against influenza virus infection. Vaccine 2009; 27:3481–3483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Wong JP, Christopher ME, Salazar AM, Dale RM, Sun LQ, Wang M. Nucleic acid‐based antiviral drugs against seasonal and avian influenza viruses. Vaccine 2007; 25:3175–3178. [DOI] [PubMed] [Google Scholar]
  • 146. Ranjan P, Jayashankar L, Deyde V et al. 5′PPP‐RNA induced RIG‐I activation inhibits drug‐resistant avian H5N1 as well as 1918 and 2009 pandemic influenza virus replication. Virol J 2010; 7:102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Hava DL, Griel LC, DeHaan WH, Hubeau C, Kenyon J. Inhaled cationic airway lining modulator (iCALM) therapy, A novel aerosol treatment for respiratory infections reduces clinical symptoms and transmission of influenza A infection. Am J Respir Crit Care Med 2010; 181:A6846. [Google Scholar]
  • 148. Donovan BW, Reuter JD, Cao Z, Myc A, Johnson KJ, Baker JR Jr. Prevention of murine influenza A virus pneumonitis by surfactant nano‐emulsions. Antivir Chem Chemother 2000; 11:41–49. [DOI] [PubMed] [Google Scholar]
  • 149. Motard J, Rouxel R, Paun A, Von Messling V, Bisaillon M, Perreault JP. A novel ribozyme‐based prophylaxis inhibits influenza A virus replication and protects from severe disease. PLoS One 2011; 6:e27327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Dimmock NJ, Dove BK, Meng B et al. Comparison of the protection of ferrets against pandemic 2009 influenza A virus (H1N1) by 244 DI influenza virus and oseltamivir. Antiviral Research 2012; http://dx.doi.org/10.1016/j.antiviral.2012.09.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Ottolini M, Blanco J, Porter D, Peterson L, Curtis S, Prince G. Combination anti‐inflammatory and antiviral therapy of influenza in a cotton rat model. Pediatr Pulmonol 2003; 36:290–294. [DOI] [PubMed] [Google Scholar]
  • 152. Xu T, Qiao J, Zhao L et al. Effect of dexamethasone on acute respiratory distress syndrome induced by the H5N1 virus in mice. Eur Respir J 2009; 33:852–860. [DOI] [PubMed] [Google Scholar]
  • 153. Radigan KA, Urich D, Misharin AV et al. The effect of rosuvastatin in a murine model of influenza A infection. PLoS One 2012; 7:e35788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Liu Z, Guo Z, Wang G et al. Evaluation of the efficacy and safety of a statin/caffeine combination against H5N1, H3N2 and H1N1 virus infection in BALB/c mice. Eur J Pharm Sci 2009; 38:215–223. [DOI] [PubMed] [Google Scholar]
  • 155. Budd A, Alleva L, Alsharifi M et al. Increased survival after gemfibrozil treatment of severe mouse influenza. Antimicrob Agents Chemother 2007; 51:2965–2968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Aldridge JR Jr, Moseley CE, Boltz DA et al. TNF/iNOS‐producing dendritic cells are the necessary evil of lethal influenza virus infection. Proc Natl Acad Sci U S A 2009; 106:5306–5311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Moseley CE, Webster RG, Aldridge JR. Peroxisome proliferator‐activated receptor and AMP‐activated protein kinase agonists protect against lethal influenza virus challenge in mice. Influenza and Other Respiratory Viruses 2010; 4:307–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Lin KL, Sweeney S, Kang BD, Ramsburg E, Gunn MD. CCR2‐antagonist prophylaxis reduces pulmonary immune pathology and markedly improves survival during influenza infection. J Immunol 2011; 186:508–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. 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 U S A 2008; 105:8091–8096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Carey MA, Bradbury JA, Rebolloso YD, Graves JP, Zeldin DC, Germolec DR. Pharmacologic inhibition of COX‐1 and COX‐2 in influenza A viral infection in mice. PLoS One 2010; 5:e11610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Garozzo A, Tempera G, Ungheri D, Timpanaro R, Castro A. N‐acetylcysteine synergizes with oseltamivir in protecting mice from lethal influenza infection. Int J Immunopathol Pharmacol 2007; 20:349–354. [DOI] [PubMed] [Google Scholar]
  • 162. Lai KY, Ng WY, Osburga ChanPK, Wong KF, Cheng F. High‐dose N‐acetylcysteine therapy for novel H1N1 influenza pneumonia. Ann Intern Med 2010; 152:687–688. [DOI] [PubMed] [Google Scholar]
  • 163. Vigerust DJ, McCullers JA. Chloroquine is effective against influenza A virus in vitro but not in vivo. Influenza Other Respi Viruses 2007; 1:189–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. 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. [DOI] [PubMed] [Google Scholar]
  • 165. Sato K, Suga M, Akaike T et al. Therapeutic effect of erythromycin on influenza virus‐induced lung injury in mice. Am J Respir Crit Care Med 1998;157 (3 Pt 1):853–857. [DOI] [PubMed] [Google Scholar]
  • 166. Hu Y, Jin Y, Han D et al. Mast cell‐induced lung injury in mice infected with H5N1 influenza virus. J Virol 2012; 86:3347–3356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Tu W, Zheng J, Liu Y et al. The aminobisphosphonate pamidronate controls influenza pathogenesis by expanding a gammadelta T cell population in humanized mice. J Exp Med 2011; 208:1511–1522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Akaike T, Ando M, Oda T et al. Dependence on O2‐ generation by xanthine oxidase of pathogenesis of influenza virus infection in mice. J Clin Invest 1990; 85:739–745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Grattendick K, Lefkowitz DL, Lefkowitz SS. Inhibition of influenza virus replication by cocaine. Int J Immunopharmacol 2000; 22:105–111. [DOI] [PubMed] [Google Scholar]
  • 170. Hammerbeck DM, Burleson GR, Schuller CJ et al. Administration of a dual toll‐like receptor 7 and toll‐like receptor 8 agonist protects against influenza in rats. Antiviral Res 2007; 73:1–11. [DOI] [PubMed] [Google Scholar]
  • 171. Izumo T, Maekawa T, Ida M et al. Effect of intranasal administration of Lactobacillus pentosus S‐PT84 on influenza virus infection in mice. Int Immunopharmacol 2010; 10:1101–1106. [DOI] [PubMed] [Google Scholar]
  • 172. Kobayashi N, Saito T, Uematsu T et al. Oral administration of heat‐killed Lactobacillus pentosus strain b240 augments protection against influenza virus infection in mice. Int Immunopharmacol 2011; 11:199–203. [DOI] [PubMed] [Google Scholar]
  • 173. Maeda N, Nakamura R, Hirose Y et al. Oral administration of heat‐killed Lactobacillus plantarum L‐137 enhances protection against influenza virus infection by stimulation of type I interferon production in mice. Int Immunopharmacol 2009; 9:1122–1125. [DOI] [PubMed] [Google Scholar]
  • 174. Ball MA, Utsunomiya T, Ikemoto K, Kobayashi M, Pollard RB, Suzuki F. The antiviral effect of keishi‐ni‐eppi‐ichi‐to, a traditional Chinese herbal medicine, on influenza A2(H2N2) virus infection in mice. Experientia 1994; 50:774–779. [DOI] [PubMed] [Google Scholar]
  • 175. Wang C, Cao B, Liu QQ et al. Oseltamivir compared with the Chinese traditional therapy maxingshigan‐yinqiaosan in the treatment of H1N1 influenza: a randomized trial. Ann Intern Med 2011; 155:217–225. [DOI] [PubMed] [Google Scholar]
  • 176. Darwish I, Miller C, Kain KC, Liles WC. Inhaled nitric oxide therapy fails to improve outcome in experimental severe influenza. Int J Med Sci 2012; 9:157–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Fusco D, Liu X, Savage C et al. Echinacea purpurea aerial extract alters course of influenza infection in mice. Vaccine 2010; 28:3956–3962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Kosai K, Seki M, Yanagihara K et al. Gabexate mesilate suppresses influenza pneumonia in mice through inhibition of cytokines. J Int Med Res 2008; 36:322–328. [DOI] [PubMed] [Google Scholar]
  • 179. Matsumoto K, Yamada H, Takuma N, Niino H, Sagesaka YM. Effects of green tea catechins and theanine on preventing influenza infection among healthcare workers: a randomized controlled trial. BMC Complement Altern Med 2011; 11:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Ichinohe T, Nagata N, Strong P et al. Prophylactic effects of chitin microparticles on highly pathogenic H5N1 influenza virus. J Med Virol 2007; 79:811–819. [DOI] [PubMed] [Google Scholar]
  • 181. Kim Y, Narayanan S, Chang KO. Inhibition of influenza virus replication by plant‐derived isoquercetin. Antiviral Res 2010; 88:227–235. [DOI] [PubMed] [Google Scholar]
  • 182. Buchweitz JP, Karmaus PW, Harkema JR, Williams KJ, Kaminski NE. Modulation of airway responses to influenza A/PR/8/34 by Delta9‐tetrahydrocannabinol in C57BL/6 mice. J Pharmacol Exp Ther 2007; 323:675–683. [DOI] [PubMed] [Google Scholar]
  • 183. Palamara AT, Nencioni L, Aquilano K et al. Inhibition of influenza A virus replication by resveratrol. J Infect Dis 2005; 191:1719–1729. [DOI] [PubMed] [Google Scholar]
  • 184. London NR, Zhu W, Bozza FA et al. Targeting Robo4‐Dependent Slit Signaling to Survive the Cytokine Storm in Sepsis and Influenza Science. Translational Med 2010; 2(23):23ra19. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Influenza and Other Respiratory Viruses are provided here courtesy of Wiley

RESOURCES