Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 491
Filter
1.
Antiviral Res ; 228: 105938, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38897317

ABSTRACT

We compared the duration of fever in children infected with A(H1N1)pdm09, A(H3N2), or influenza B viruses following treatment with baloxavir marboxil (baloxavir) or neuraminidase inhibitors (NAIs) (oseltamivir, zanamivir, or laninamivir). This observational study was conducted at 10 outpatient clinics across 9 prefectures in Japan during the 2012-2013 and 2019-2020 influenza seasons. Patients with influenza rapid antigen test positive were treated with one of four anti-influenza drugs. The type/subtype of influenza viruses were identified from MDCK or MDCK SIAT1 cell-grown samples using two-step real-time PCR. Daily self-reported body temperature after treatment were used to evaluate the duration of fever by treatment group and various underlying factors. Among 1742 patients <19 years old analyzed, 452 (26.0%) were A(H1N1)pdm09, 827 (48.0%) A(H3N2), and 463 (26.0%) influenza B virus infections. Among fours treatment groups, baloxavir showed a shorter median duration of fever compared to oseltamivir in univariate analysis for A(H1N1)pdm09 virus infections (baloxavir, 22.0 h versus oseltamivir, 26.7 h, P < 0.05; laninamivir, 25.5 h, and zanamivir, 25.0 h). However, this difference was not significant in multivariable analyses. For A(H3N2) virus infections, there were no statistically significant differences observed (20.3, 21.0, 22.0, and 19.0 h) uni- and multivariable analyses. For influenza B, baloxavir shortened the fever duration by approximately 15 h than NAIs (20.3, 35.0, 34.3, and 34.1 h), as supported by uni- and multivariable analyses. Baloxavir seems to have comparable clinical effectiveness with NAIs on influenza A but can be more effective for treating pediatric influenza B virus infections than NAIs.


Subject(s)
Antiviral Agents , Dibenzothiepins , Fever , Guanidines , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Influenza B virus , Influenza, Human , Morpholines , Oseltamivir , Pyrans , Pyridones , Sialic Acids , Triazines , Zanamivir , Humans , Influenza, Human/drug therapy , Influenza, Human/virology , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Influenza B virus/drug effects , Influenza B virus/genetics , Child , Zanamivir/therapeutic use , Zanamivir/analogs & derivatives , Zanamivir/pharmacology , Triazines/therapeutic use , Triazines/pharmacology , Guanidines/therapeutic use , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H1N1 Subtype/drug effects , Pyridones/therapeutic use , Dibenzothiepins/therapeutic use , Japan , Female , Male , Child, Preschool , Oseltamivir/therapeutic use , Fever/drug therapy , Fever/virology , Adolescent , Morpholines/therapeutic use , Infant , Seasons , Thiepins/therapeutic use , Thiepins/pharmacology , Oxazines/therapeutic use , Time Factors , Benzoxazines/therapeutic use
2.
Antiviral Res ; 227: 105918, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38795911

ABSTRACT

The most widely used class of antivirals available for Influenza treatment are the neuraminidase inhibitors (NAI) Oseltamivir and Zanamivir. However, amino acid (AA) substitutions in the neuraminidase may cause reduced inhibition or high antiviral resistance. In Mexico, the current state of knowledge about NAI susceptibility is scarce, in this study we report the results of 14 years of Influenza surveillance by phenotypic and genotypic methods. A total of 255 isolates were assessed with the NAI assay, including Influenza A(H1N1)pdm09, A(H3N2) and Influenza B (IBV). Furthermore, 827 sequences contained in the GISAID platform were analyzed in search of relevant mutations.Overall, five isolates showed highly reduced inhibition or reduced inhibition to Oseltamivir, and two showed reduced inhibition to Zanamivir in the NAI assays. Additionally, five A(H1N1)pdm09 sequences from the GISAID possessed AA substitutions associated to reduced inhibition to Oseltamivir and none to Zanamivir. Oseltamivir resistant A(H1N1)pdm09 harbored the H275Y mutation. No genetic mutations were identified in Influenza A(H3N2) and IBV. Overall, these results show that in Mexico the rate of NAI resistance is low (0.6%), but it is essential to continue the Influenza surveillance in order to understand the drug susceptibility of circulating strains.


Subject(s)
Antiviral Agents , Drug Resistance, Viral , Influenza B virus , Influenza, Human , Neuraminidase , Oseltamivir , Zanamivir , Drug Resistance, Viral/genetics , Antiviral Agents/pharmacology , Mexico/epidemiology , Humans , Influenza B virus/drug effects , Influenza B virus/genetics , Influenza, Human/virology , Influenza, Human/drug therapy , Influenza, Human/epidemiology , Oseltamivir/pharmacology , Zanamivir/pharmacology , Neuraminidase/genetics , Neuraminidase/antagonists & inhibitors , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Mutation , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/genetics , Adult , Influenza A virus/drug effects , Influenza A virus/genetics , Adolescent , Child , Amino Acid Substitution , Young Adult , Middle Aged , Female , Child, Preschool , Genotype , Male , Aged , Microbial Sensitivity Tests , Viral Proteins/genetics
3.
Chem Biol Drug Des ; 99(3): 398-415, 2022 03.
Article in English | MEDLINE | ID: mdl-34873848

ABSTRACT

In previous investigations, we identified a class of 1,3,4-thiadiazole derivatives with antiviral activity. N-{3-(Methylsulfanyl)-1-[5-(phenylamino)-1,3,4-thiadiazole-2-yl]propyl}benzamide emerged as a relevant lead compound for designing novel influenza A virus inhibitors. In the present study, we elaborated on this initial lead by performing chemical synthesis and antiviral evaluation of a series of structural analogues. During this research, thirteen novel 1,3,4-thiadiazole derivatives were synthesized by the cyclization of the corresponding thiosemicarbazides as synthetic precursors. The structures and the purities of the synthesized compounds were confirmed through chromatographic and spectral data. Four L-methionine-based 1,3,4-thiadiazole derivatives displayed activity against influenza A virus, the two best compounds being 24 carrying a 5-(4-chlorophenylamino)-1,3,4-thiadiazole moiety and 30 possessing a 5-(benzoylamino)-1,3,4-thiadiazole structure [antiviral EC50 against influenza A/H3N2 virus: 4.8 and 7.4 µM, respectively]. The 1,3,4-thiadiazole derivatives were inactive against influenza B virus and a wide panel of unrelated DNA and RNA viruses. Compound 24 represents a new class of selective influenza A virus inhibitors acting during the virus entry process, as evidenced by our findings in a time-of-addition assay. Molecular descriptors and in silico prediction of ADMET properties of the active compounds were calculated. According to in silico ADMET and drug similarity studies, active compounds have been estimated to be good candidates for oral administration with no apparent toxicity considerations.


Subject(s)
Antiviral Agents/chemical synthesis , Methionine/chemistry , Thiadiazoles/chemistry , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Drug Design , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/physiology , Influenza B virus/drug effects , Influenza B virus/physiology , Structure-Activity Relationship , Thiadiazoles/chemical synthesis , Thiadiazoles/pharmacology , Virus Internalization/drug effects
4.
J Med Chem ; 65(1): 369-385, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34905383

ABSTRACT

Influenza viruses are responsible for contagious respiratory illnesses in humans and cause seasonal epidemics and occasional pandemics worldwide. Previously, we identified a quinolinone derivative PA-49, which inhibited the influenza virus RNA-dependent RNA polymerase (RdRp) by targeting PA-PB1 interaction. This paper reports the structure optimization of PA-49, which resulted in the identification of 3-((dibenzylamino)methyl)quinolinone derivatives with more potent anti-influenza virus activity. During the optimization, the hit compound 89, which was more active than PA-49, was identified. Further optimization and scaffold hopping of 89 led to the most potent compounds 100 and a 1,8-naphthyridinone derivative 118, respectively. We conclusively determined that compounds 100 and 118 suppressed the replication of influenza virus and exhibited anti-influenza virus activity against both influenza virus types A and B in the range of 50% effective concentration (EC50) = 0.061-0.226 µM with low toxicity (50% cytotoxic concentration (CC50) >10 µM).


Subject(s)
Antiviral Agents/pharmacology , DNA-Directed RNA Polymerases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Orthomyxoviridae/drug effects , Orthomyxoviridae/enzymology , Animals , Antiviral Agents/chemistry , Antiviral Agents/toxicity , Cell Line , Dogs , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , Humans , Influenza A virus/drug effects , Influenza B virus/drug effects , Madin Darby Canine Kidney Cells , Models, Molecular , Molecular Docking Simulation , Structure-Activity Relationship
5.
Nutrients ; 13(11)2021 Oct 30.
Article in English | MEDLINE | ID: mdl-34836165

ABSTRACT

It is difficult to match annual vaccines against the exact influenza strain that is spreading in any given flu season. Owing to the emergence of drug-resistant viral strains, new approaches for treating influenza are needed. Euglena gracilis (hereinafter Euglena), microalga, used as functional foods and supplements, have been shown to alleviate symptoms of influenza virus infection in mice. However, the mechanism underlying the inhibitory action of microalgae against the influenza virus is unknown. Here, we aimed to study the antiviral activity of Euglena extract against the influenza virus and the underlying action mechanism using Madin-Darby canine kidney (MDCK) cells. Euglena extract strongly inhibited infection by all influenza virus strains examined, including those resistant to the anti-influenza drugs oseltamivir and amantadine. A time-of-addition assay revealed that Euglena extract did not affect the cycle of virus replication, and cell pretreatment or prolonged treatment of infected cells reduced the virus titer. Thus, Euglena extract may activate the host cell defense mechanisms, rather than directly acting on the influenza virus. Moreover, various minerals, mainly zinc, in Euglena extract were found to be involved in the antiviral activity of the extract. In conclusion, Euglena extract could be a potent agent for preventing and treating influenza.


Subject(s)
Antiviral Agents , Complex Mixtures/pharmacology , Euglena , Influenza A virus/growth & development , Influenza B virus/growth & development , Animals , Dogs , Euglena/chemistry , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A virus/drug effects , Influenza B virus/drug effects , Madin Darby Canine Kidney Cells , Virus Replication/drug effects , Zinc/analysis , Zinc Acetate/pharmacology
6.
Antiviral Res ; 196: 105208, 2021 12.
Article in English | MEDLINE | ID: mdl-34793841

ABSTRACT

To suppress serious influenza infections in persons showing insufficient protection from the vaccines, antiviral drugs are of vital importance. There is a need for novel agents with broad activity against influenza A (IAV) and B (IBV) viruses and with targets that differ from those of the current antivirals. We here report a new small molecule influenza virus inhibitor referred to as CPD A (chemical name: N-(pyridin-3-yl)thiophene-2-carboxamide). In an influenza virus minigenome assay, this non-nucleoside compound inhibited RNA synthesis of IAV and IBV with EC50 values of 2.3 µM and 2.6 µM, respectively. Robust in vitro activity was noted against a broad panel of IAV (H1N1 and H3N2) and IBV strains, with a median EC50 value of 0.20 µM, which is 185-fold below the 50% cytotoxic concentration. The action point in the viral replication cycle was located between 1 and 5 h p.i., showing a similar profile as ribavirin. Like this nucleoside analogue, CPD A was shown to cause strong depletion of the cellular GTP pool and, accordingly, its antiviral activity was antagonized when this pool was restored with exogenous guanosine. This aligns with the observed inhibition in a cell-based IMP dehydrogenase (IMPDH) assay, which seems to require metabolic activation of CPD A since no direct inhibition was seen in an enzymatic IMPDH assay. The combination of CPD A with ribavirin, another IMPDH inhibitor, proved strongly synergistic. To conclude, we established CPD A as a new inhibitor of influenza A and B virus replication and RNA synthesis, and support the potential of IMPDH inhibitors for influenza therapy with acceptable safety profile.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , Influenza A virus/drug effects , Influenza B virus/drug effects , Ribavirin/pharmacology , Cell Line , Drug Synergism , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A virus/classification , Influenza, Human/drug therapy
7.
PLoS One ; 16(10): e0259129, 2021.
Article in English | MEDLINE | ID: mdl-34714852

ABSTRACT

Influenza viruses cause respiratory tract infections and substantial health concerns. Infection may result in mild to severe respiratory disease associated with morbidity and some mortality. Several anti-influenza drugs are available, but these agents target viral components and are susceptible to drug resistance. There is a need for new antiviral drug strategies that include repurposing of clinically approved drugs. Drugs that target cellular machinery necessary for influenza virus replication can provide a means for inhibiting influenza virus replication. We used RNA interference screening to identify key host cell genes required for influenza replication, and then FDA-approved drugs that could be repurposed for targeting host genes. We examined the effects of Clopidogrel and Triamterene to inhibit A/WSN/33 (EC50 5.84 uM and 31.48 uM, respectively), A/CA/04/09 (EC50 6.432 uM and 3.32 uM, respectively), and B/Yamagata/16/1988 (EC50 0.28 uM and 0.11 uM, respectively) replication. Clopidogrel and Triamterene provide a druggable approach to influenza treatment across multiple strains and subtypes.


Subject(s)
Antiviral Agents/pharmacology , Clopidogrel/pharmacology , Influenza A virus/drug effects , Influenza B virus/drug effects , Influenza, Human/drug therapy , Triamterene/pharmacology , A549 Cells , Animals , Dogs , Drug Repositioning , Humans , Madin Darby Canine Kidney Cells , Virus Replication/drug effects
8.
Sci Rep ; 11(1): 9427, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33941825

ABSTRACT

Influenza viruses cause significant morbidity and mortality worldwide. Long-term or frequent use of approved anti-influenza agents has resulted in drug-resistant strains, thereby necessitating the discovery of new drugs. In this study, we found aprotinin, a serine protease inhibitor, as an anti-influenza candidate through screening of compound libraries. Aprotinin has been previously reported to show inhibitory effects on a few influenza A virus (IAV) subtypes (e.g., seasonal H1N1 and H3N2). However, because there were no reports of its inhibitory effects on the other types of influenza viruses, we investigated the inhibitory effects of aprotinin in vitro on a wide range of influenza viruses, including avian and oseltamivir-resistant influenza virus strains. Our cell-based assay showed that aprotinin had inhibitory effects on seasonal human IAVs (H1N1 and H3N2 subtypes), avian IAVs (H5N2, H6N5, and H9N2 subtypes), an oseltamivir-resistant IAV, and a currently circulating influenza B virus. We have also confirmed its activity in mice infected with a lethal dose of influenza virus, showing a significant increase in survival rate. Our findings suggest that aprotinin has the capacity to inhibit a wide range of influenza virus subtypes and should be considered for development as a therapeutic agent against influenza.


Subject(s)
Antiviral Agents/pharmacology , Aprotinin/pharmacology , Drug Evaluation, Preclinical , Orthomyxoviridae Infections/drug therapy , Serine Proteinase Inhibitors/pharmacology , Animals , Cell Line , Dogs , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H5N2 Subtype/drug effects , Influenza A Virus, H5N2 Subtype/growth & development , Influenza A Virus, H9N2 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/growth & development , Influenza B virus/drug effects , Influenza B virus/growth & development , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL
9.
Int J Mol Sci ; 22(5)2021 Feb 27.
Article in English | MEDLINE | ID: mdl-33673431

ABSTRACT

In this study, we have introduced newly synthesized substituted benzothiazole based berberine derivatives that have been analyzed for their in vitro and in silico biological properties. The activity towards various kinds of influenza virus strains by employing the cytopathic effect (CPE) and sulforhodamine B (SRB) assay. Several berberine-benzothiazole derivatives (BBDs), such as BBD1, BBD3, BBD4, BBD5, BBD7, and BBD11, demonstrated interesting anti-influenza virus activity on influenza A viruses (A/PR/8/34, A/Vic/3/75) and influenza B viral (B/Lee/40, and B/Maryland/1/59) strain, respectively. Furthermore, by testing neuraminidase activity (NA) with the neuraminidase assay kit, it was identified that BBD7 has potent neuraminidase activity. The molecular docking analysis further suggests that the BBD1-BBD14 compounds' antiviral activity may be because of interaction with residues of NA, and the same as in oseltamivir.


Subject(s)
Benzothiazoles/pharmacology , Berberine/pharmacology , Molecular Docking Simulation , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae Infections/drug therapy , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Benzothiazoles/therapeutic use , Berberine/analogs & derivatives , Berberine/therapeutic use , Cell Line , Cytopathogenic Effect, Viral , Dogs , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Influenza A virus/drug effects , Influenza A virus/enzymology , Influenza B virus/drug effects , Influenza B virus/enzymology , Orthomyxoviridae Infections/enzymology , Viral Proteins/antagonists & inhibitors
10.
Sci Rep ; 11(1): 4554, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33654128

ABSTRACT

While vaccines remain the best tool for preventing influenza virus infections, they have demonstrated low to moderate effectiveness in recent years. Seasonal influenza vaccines typically consist of wild-type influenza A and B viruses that are limited in their ability to elicit protective immune responses against co-circulating influenza virus variant strains. Improved influenza virus vaccines need to elicit protective immune responses against multiple influenza virus drift variants within each season. Broadly reactive vaccine candidates potentially provide a solution to this problem, but their efficacy may begin to wane as influenza viruses naturally mutate through processes that mediates drift. Thus, it is necessary to develop a method that commercial vaccine manufacturers can use to update broadly reactive vaccine antigens to better protect against future and currently circulating viral variants. Building upon the COBRA technology, nine next-generation H3N2 influenza hemagglutinin (HA) vaccines were designed using a next generation algorithm and design methodology. These next-generation broadly reactive COBRA H3 HA vaccines were superior to wild-type HA vaccines at eliciting antibodies with high HAI activity against a panel of historical and co-circulating H3N2 influenza viruses isolated over the last 15 years, as well as the ability to neutralize future emerging H3N2 isolates.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Vaccines, Virus-Like Particle/therapeutic use , Hemagglutination Inhibition Tests , Humans , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza A virus/drug effects , Influenza A virus/pathogenicity , Influenza B virus/drug effects , Influenza B virus/pathogenicity , Influenza Vaccines/genetics , Influenza Vaccines/therapeutic use , Influenza, Human/immunology , Influenza, Human/virology , Seasons , Vaccines, Virus-Like Particle/immunology
11.
J Biol Chem ; 296: 100486, 2021.
Article in English | MEDLINE | ID: mdl-33647314

ABSTRACT

Baloxavir marboxil (BXM) is an FDA-approved antiviral prodrug for the treatment of influenza A and B infection and postexposure prophylaxis. The active form, baloxavir acid (BXA), targets the cap-snatching endonuclease (PA) of the influenza virus polymerase complex. The nuclease activity delivers the primer for transcription, and previous reports have shown that BXA blocks the nuclease activity with high potency. However, biochemical studies on the mechanism of action are lacking. Structural data have shown that BXA chelates the two divalent metal ions at the active site, like inhibitors of the human immunodeficiency virus type 1 (HIV-1) integrase or ribonuclease (RNase) H. Here we studied the mechanisms underlying the high potency of BXA and how the I38T mutation confers resistance to the drug. Enzyme kinetics with the recombinant heterotrimeric enzyme (FluB-ht) revealed characteristics of a tight binding inhibitor. The apparent inhibitor constant (Kiapp) is 12 nM, while the I38T mutation increased Kiapp by ∼18-fold. Order-of-addition experiments show that a preformed complex of FluB-ht, Mg2+ ions and BXA is required to observe inhibition, which is consistent with active site binding. Conversely, a preformed complex of FluB-ht and RNA substrate prevents BXA from accessing the active site. Unlike integrase inhibitors that interact with the DNA substrate, BXA behaves like RNase H inhibitors that compete with the nucleic acid at the active site. The collective data support the conclusion that BXA is a tight binding inhibitor and the I38T mutation diminishes these properties.


Subject(s)
Dibenzothiepins/pharmacology , Endonucleases/antagonists & inhibitors , Influenza B virus/drug effects , Influenza, Human/drug therapy , Influenza, Human/virology , Morpholines/pharmacology , Pyridones/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Triazines/pharmacology , Virus Replication/drug effects , Antiviral Agents/pharmacology , Catalytic Domain , Endonucleases/metabolism , Humans , Influenza B virus/enzymology , Influenza B virus/isolation & purification , Influenza, Human/enzymology , RNA-Dependent RNA Polymerase/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism
12.
J Med Virol ; 93(6): 3465-3472, 2021 06.
Article in English | MEDLINE | ID: mdl-32827314

ABSTRACT

Influenza A virus (IAV) causes great morbidity and mortality worldwide every year. However, there are only a limited number of drugs clinically available against IAV infection. Further, emergence of drug-resistant strains can render those drugs ineffective. Thus there is an unmet medical need to develop new anti-influenza agents. In this study, we show that punicalagin from plants possesses strong anti-influenza activity with a low micromolar IC50 value in tissue culture. Using a battery of bioassays such as single-cycle replication assay, neuraminidase (NA) inhibition assay, and virus yield reduction assay, we demonstrate that the primary mechanism of action (MOA) of punicalagin is the NA-mediated viral release. Moreover, punicalagin can inhibit replication of different strains of influenza A and B viruses, including oseltamivir-resistant virus (NA/H274Y), indicating that punicalagin is a broad spectrum antiviral against both IAV and IBV. Further, although punicalagin targets NA like oseltamivir, it has a different MOA. These results suggest that punicalagin is an influenza NA inhibitor that may be further developed as a novel antiviral against influenza viruses.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Hydrolyzable Tannins/pharmacology , Influenza A virus/drug effects , Influenza B virus/drug effects , Neuraminidase/antagonists & inhibitors , Plant Extracts/pharmacology , Animals , Dogs , Influenza A virus/enzymology , Inhibitory Concentration 50 , Madin Darby Canine Kidney Cells , Virus Replication/drug effects
13.
Antiviral Res ; 185: 104970, 2021 01.
Article in English | MEDLINE | ID: mdl-33159999

ABSTRACT

Influenza B viruses cause significant morbidity and mortality, particularly in children, but the awareness of their impact is often less than influenza A viruses partly due to their lack of pandemic potential. Here, we summarise the biology, epidemiology and disease burden of influenza B, and review existing data on available antivirals for its management. There has long been uncertainty surrounding the clinical efficacy of neuraminidase inhibitors (NAIs) for influenza B treatment. In this article, we bring together the existing data on NAIs and discuss these alongside recent large randomised controlled trial data for the new polymerase inhibitor baloxavir in high-risk influenza B patients. Finally, we offer considerations for the clinical management of influenza B, with a focus on children and high-risk patients where disease burden is highest.


Subject(s)
Cost of Illness , Disease Management , Influenza B virus/pathogenicity , Influenza, Human/prevention & control , Antiviral Agents/therapeutic use , Child , Clinical Trials, Phase III as Topic , Dibenzothiepins/pharmacology , Dibenzothiepins/therapeutic use , Drug Resistance, Viral , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Influenza B virus/drug effects , Influenza, Human/drug therapy , Morpholines/pharmacology , Morpholines/therapeutic use , Pandemics/prevention & control , Pyridones/pharmacology , Pyridones/therapeutic use , Triazines/pharmacology , Triazines/therapeutic use
15.
Rev Med Virol ; 31(3): e2175, 2021 05.
Article in English | MEDLINE | ID: mdl-32975358

ABSTRACT

Neuraminidase inhibitors (NAIs), that currently include oseltamivir (Tamiflu® ), zanamivir (Relenza® ), peramivir (Rapivab® ) and laninamivir (Inavir® ), constitute an important class of antivirals recommended against seasonal influenza A and B infections. NAIs target the surface NA protein whose sialidase activity is responsible for virion release from infected cells. Because of their pivotal role in the transcription/translation process, the polymerase acidic (PA) and polymerase basic 1 and 2 (PB1 and PB2, respectively) internal proteins also constitute targets of interest for the development of additional anti-influenza agents. Baloxavir marboxil (BXM), an inhibitor of the cap-dependent endonuclease activity of the influenza PA protein, was approved in the United States and Japan in 2018. Baloxavir acid (BXA), the active compound of BXM, demonstrated a potent in vitro activity against different types/subtypes of influenza viruses including seasonal influenza A/B strains as well as avian influenza A viruses with a pandemic potential. A single oral dose of BXM provided virological and clinical benefits that were respectively superior or equal to those displayed by the standard (5 days, twice daily) oseltamivir regimen. Nevertheless, BXM-resistant variants have emerged at relatively high rates in BXM-treated children and adults. Consequently, there is a need to study the fitness (virulence and transmissibility) characteristics of mutants with a high potential to emerge as such variants can compromise the clinical usefulness of BXM. The purpose of this manuscript is to review the fitness properties of influenza A and B isolates harbouring mutations of reduced susceptibility to BXA.


Subject(s)
Antiviral Agents/pharmacology , Dibenzothiepins/pharmacology , Influenza A virus/drug effects , Influenza B virus/drug effects , Influenza, Human/drug therapy , Morpholines/pharmacology , Pyridones/pharmacology , Triazines/pharmacology , Drug Resistance, Viral , Humans , Influenza A virus/isolation & purification
16.
Nat Commun ; 11(1): 5597, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33154358

ABSTRACT

Seasonal influenza epidemics lead to 3-5 million severe infections and 290,000-650,000 annual global deaths. With deaths from the 1918 influenza pandemic estimated at >50,000,000 and future pandemics anticipated, the need for a potent influenza treatment is critical. In this study, we design and synthesize a bifunctional small molecule by conjugating the neuraminidase inhibitor, zanamivir, with the highly immunogenic hapten, dinitrophenyl (DNP), which specifically targets the surface of free virus and viral-infected cells. We show that this leads to simultaneous inhibition of virus release, and immune-mediated elimination of both free virus and virus-infected cells. Intranasal or intraperitoneal administration of a single dose of drug to mice infected with 100x MLD50 virus is shown to eradicate advanced infections from representative strains of both influenza A and B viruses. Since treatments of severe infections remain effective up to three days post lethal inoculation, our approach may successfully treat infections refractory to current therapies.


Subject(s)
Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Immunotherapy/methods , Orthomyxoviridae Infections/drug therapy , 2,4-Dinitrophenol/administration & dosage , 2,4-Dinitrophenol/chemistry , 2,4-Dinitrophenol/immunology , Administration, Intranasal , Animals , Antibodies/administration & dosage , Antibodies/immunology , Antiviral Agents/chemistry , Cell Line , Cytotoxicity, Immunologic/drug effects , Drug Delivery Systems , Humans , Influenza A virus/drug effects , Influenza A virus/enzymology , Influenza A virus/physiology , Influenza B virus/drug effects , Influenza B virus/enzymology , Influenza B virus/physiology , Infusions, Parenteral , Mice , Mice, Inbred BALB C , Neuraminidase/antagonists & inhibitors , Neuraminidase/metabolism , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Protein Binding , Treatment Outcome , Virus Release/drug effects , Zanamivir/administration & dosage , Zanamivir/chemistry , Zanamivir/pharmacology
17.
Antiviral Res ; 182: 104895, 2020 10.
Article in English | MEDLINE | ID: mdl-32750469

ABSTRACT

With the introduction of the influenza specific neuraminidase inhibitors (NAIs) in 1999, there were concerns about the emergence and spread of resistant viruses in the community setting. Surveillance and testing of community isolates for their susceptibility to the NAIs was initially carried out by the Neuraminidase Inhibitor Susceptibility Network (NISN) and has subsequently been taken on by the global WHO influenza network laboratories. During the NISN surveillance, we identified two Yamagata lineage influenza B viruses with amino acid substitutions of H134Y (B/Auckland/2/2001) or W438R (B/Yokohama/12/2005) which had slightly elevated IC50 values for zanamivir and/or oseltamivir, but not sufficiently to be characterized as mild outliers at the time. As it has now been well demonstrated that mixed populations can mask the true magnitude of resistance of a mutant, we re-examined both of these isolates by plaque purification to see if the true susceptibilities were being masked due to mixed populations. Results confirmed that the B/Auckland isolate contained both wild type and H134Y mutant populations, with mutant IC50 values > 250 nM for both oseltamivir and peramivir in the enzyme inhibition assay. The B/Yokohama isolate also contained both wild type and W438R mutant populations, the latter now demonstrating IC50 values > 400 nM for zanamivir, oseltamivir and peramivir. In addition, plaque purification of the B/Yokohama isolate identified viruses with other single neuraminidase substitutions H134Y, H134R, H431R, or T436P. H134R and H431R viruses had IC50 values > 400 nM and >250 nM respectively against all three NAIs. All changes conferred much greater resistance to peramivir than to zanamivir, and less to oseltamivir, and affected the kinetics of binding and dissociation of the NAIs. Most affected affinity (Km) for the MUNANA substrate, but some had decreased while others had increased affinity. Despite resistance in the enzyme assay, no reduced susceptibility was seen in plaque reduction assays in MDCK cells for any of the mutant viruses. None of these substitutions was in the active site. Modelling suggests that these substitutions affect the 150 and 430-loop regions described for influenza A NAs, suggesting they may also be important for substrate and inhibitor binding for influenza B NAs.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral/genetics , Enzyme Inhibitors/pharmacology , Influenza B virus/drug effects , Neuraminidase/antagonists & inhibitors , Neuraminidase/genetics , Amino Acid Substitution , Animals , Antiviral Agents/classification , Dogs , Enzyme Inhibitors/classification , Humans , Influenza B virus/genetics , Influenza, Human/virology , Inhibitory Concentration 50 , Madin Darby Canine Kidney Cells , Viral Proteins/genetics
18.
Drugs ; 80(11): 1109-1118, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32601915

ABSTRACT

Baloxavir marboxil (Xofluza®; hereafter referred to as baloxavir), the prodrug of baloxavir acid, is a first-in-class, small molecule inhibitor of the polymerase acidic (PA) protein subunit of the influenza virus polymerase complex. Baloxavir (after conversion to baloxavir acid) acts to block influenza virus replication by inhibiting the cap-dependent endonuclease activity of the PA protein. Taken orally as a single dose, baloxavir is approved in the USA for the treatment of acute uncomplicated influenza in patients ≥ 12 years of age who have been symptomatic for ≤ 48 h. Data from randomized, double-blind, placebo- and oseltamivir-controlled phase III trials have shown that baloxavir is efficacious in improving influenza symptoms both in otherwise healthy adolescents and adults and in those at high risk of influenza complications, displaying similar efficacy to that of oseltamivir. Furthermore, there is evidence that baloxavir can reduce influenza viral load more rapidly than oseltamivir. Baloxavir has activity against influenza A and B viruses (including strains resistant to neuraminidase inhibitors) and is well tolerated. Evidence of the emergence and likely human-to-human transmission of variant viruses with reduced susceptibility to baloxavir highlights the importance of monitoring and surveillance for changes in influenza virus drug susceptibility patterns. However, currently available evidence suggests that baloxavir, with the benefits of a single oral dose regimen, provides a useful alternative to neuraminidase inhibitors for the treatment of acute uncomplicated influenza in adolescents and adults.


Subject(s)
Dibenzothiepins/pharmacology , Influenza, Human/drug therapy , Morpholines/pharmacology , Pyridones/pharmacology , Triazines/pharmacology , Antiviral Agents/pharmacology , Humans , Influenza A virus/drug effects , Influenza B virus/drug effects , Prodrugs/pharmacology , Treatment Outcome
19.
Viruses ; 12(5)2020 05 18.
Article in English | MEDLINE | ID: mdl-32443510

ABSTRACT

Influenza virus infections can lead to viral pneumonia and acute respiratory distress syndrome in severe cases, causing significant morbidity and mortality and posing a great threat to human health. Because of the diversity of influenza virus strains and drug resistance to the current direct antiviral agents, there have been no effective drugs as yet to cure all patients infected by influenza viruses. Natural products from plants contain compounds with diverse structures that have the potential to interact with multiple host and virus factors. In this study, we identified the ethanol extract of Caesalpinia decapetala (Roth) Alston (EEC) as an inhibitor against the replication of a panel of influenza A and B viruses both on human pulmonary epithelial A549 and human monocytic U937 cells. The animal study revealed that EEC administration reduces the weight loss and improves the survival rate of mice infected with lethal influenza virus. Also, EEC treatment attenuated lung injury and reduced virus titer significantly. In conclusion, we showed that EEC has antiviral activity both in vitro and in vivo, suggesting that the plant C. decapetala has the potential to be further developed as a resource of new anti-influenza drugs.


Subject(s)
Antiviral Agents/administration & dosage , Caesalpinia/chemistry , Orthomyxoviridae Infections/drug therapy , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Animals , Antiviral Agents/pharmacology , Cell Line , Cell Survival/drug effects , Dogs , Ethanol/chemistry , Female , Humans , Influenza A virus/drug effects , Influenza B virus/drug effects , Mice , Mice, Inbred BALB C , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Viral Load/drug effects , Virus Replication/drug effects
20.
Viruses ; 12(4)2020 04 21.
Article in English | MEDLINE | ID: mdl-32326238

ABSTRACT

Every year, influenza B viruses (IBVs) contribute to annual illness, and infection can lead to serious respiratory disease among humans. More attention is needed in several areas, such as increasing virulence or pathogenicity of circulating B viruses and developing vaccines against current influenza. Since preclinical trials of anti-influenza drugs are mainly conducted in mice, we developed an appropriate infection model, using an antigenically-relevant IBV strain, for furtherance of anti-influenza drug testing and influenza vaccine protective efficacy analysis. A Victoria lineage (clade 1A) IBV was serially passaged 17 times in BALB/c mice, and adaptive amino acid substitutions were found in hemagglutinin (HA) (T214I) and neuraminidase (NA) (D432N). By electron microscopy, spherical and elliptical IBV forms were noted. Light microscopy showed that mouse-adapted IBVs caused influenza pneumonia on day 6 post inoculation. We evaluated the illness pathogenicity, viral load, and histopathological features of mouse-adapted IBVs and estimated anti-influenza drugs and vaccine efficiency in vitro and in vivo. Assessment of an investigational anti-influenza drug (oseltamivir ethoxysuccinate) and an influenza vaccine (Ultrix®, SPBNIIVS, Saint Petersburg, Russia) showed effectiveness against the mouse-adapted influenza B virus.


Subject(s)
Adaptation, Biological , Host-Pathogen Interactions , Influenza B virus/physiology , Orthomyxoviridae Infections/virology , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Disease Models, Animal , Genome, Viral , Influenza B virus/drug effects , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Male , Mice , Microbial Sensitivity Tests , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/prevention & control , RNA, Viral , Viral Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...