Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Antimicrob Chemother ; 76(1): 189-198, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33035324

ABSTRACT

BACKGROUND: Baloxavir acid, the active form of the orally available prodrug baloxavir marboxil, is a novel cap-dependent endonuclease inhibitor of influenza virus. Baloxavir marboxil has been shown to rapidly reduce virus titres compared with oseltamivir in clinical studies. OBJECTIVES: We investigated the relationship between pharmacokinetic (PK) parameters and antiviral activity of baloxavir acid based on virus titre reduction in lungs of infected mice. METHODS: BALB/c mice infected with a sub-lethal dose of influenza A(H1N1), A(H1N1)pdm09, A(H3N2) or type B virus were treated on day 5 with oral baloxavir marboxil (0.5-50 mg/kg q12h), subcutaneous baloxavir acid (0.25-8 mg/kg/day), oseltamivir phosphate (5 or 50 eq mg/kg q12h) or other antivirals for 1 day. Lung virus titres were assessed 24 h after initial antiviral dosing. PK testing was performed at up to 24 h post-dosing of baloxavir marboxil or baloxavir acid in A/WSN/33-infected mice and the PK/pharmacodynamic (PD) relationship was evaluated for baloxavir acid. RESULTS: Oral baloxavir marboxil administration showed dose-dependent virus titre reductions in lungs of mice infected with the different types/subtypes of influenza viruses 24 h post-dosing. Baloxavir marboxil at 15 mg/kg q12h resulted in ≥100-fold and ≥10-fold reductions in influenza A and B virus titres, respectively, compared with oseltamivir phosphate. PK/PD analysis showed that the plasma concentration at the end of the dosing interval (Cτ) or the plasma concentration at 24 h after initial dosing (C24) was the PK parameter predicting the virus titres at 24 h post-dosing of baloxavir acid. CONCLUSIONS: PK/PD analysis of baloxavir acid based on virus titre reduction in this mouse model could be helpful in predicting and maximizing virological outcomes in clinical settings.


Subject(s)
Dibenzothiepins , Influenza A Virus, H1N1 Subtype , Influenza, Human , Animals , Antiviral Agents/therapeutic use , Dibenzothiepins/therapeutic use , Disease Models, Animal , Endonucleases , Humans , Influenza A Virus, H3N2 Subtype , Influenza, Human/drug therapy , Mice , Mice, Inbred BALB C , Morpholines/therapeutic use , Oxazines , Pyridones , Triazines
2.
Influenza Other Respir Viruses ; 14(6): 710-719, 2020 11.
Article in English | MEDLINE | ID: mdl-32533654

ABSTRACT

BACKGROUND: Baloxavir marboxil (BXM), the oral prodrug of baloxavir acid (BXA), greatly reduces virus titers as well as influenza symptoms of uncomplicated influenza in patients. OBJECTIVES: To investigate the pharmacokinetic profiles of BXA and its efficacy against influenza A virus infection in ferrets. METHODS: Ferrets were dosed orally with BXM (10 and 30 mg/kg twice daily for 1 day), oseltamivir phosphate (OSP) (5 mg/kg twice daily for 2 days) or vehicle to measure the antiviral effects of BXM and OSP. The pharmacokinetic parameters of BXA was determined after single oral dosing of BXM. RESULTS: The maximum plasma concentrations of BXA were observed at 1.50 and 2.00 hours with the two BXM doses, which then declined with an elimination half-life of 6.91 and 4.44 hours, respectively. BXM at both doses remained detectable in the plasma in ferrets, which may be due to higher stability in liver microsomes. BXM (10 and 30 mg/kg twice daily) treatment at Day 1 post-infection (p.i.) reduced virus titers by ≥3 log10 of the 50% tissue culture infective doses by Day 2, which was significantly different compared with vehicle or OSP. Body temperature drops over time were significantly greater with BXM than with vehicle or OSP. Significant reduction in virus titers was also demonstrated when BXM was administrated after symptom onset at Day 2 p.i. compared with vehicle and OSP, although body temperature changes largely overlapped between Day 2 and Day 4. CONCLUSIONS: The results highlight the rapid antiviral action of BXM with post-exposure prophylaxis or therapeutic dosing in ferrets and offer support for further research on prevention of influenza virus infection and transmission.


Subject(s)
Antiviral Agents/therapeutic use , Dibenzothiepins/therapeutic use , Influenza A virus/drug effects , Morpholines/therapeutic use , Orthomyxoviridae Infections/drug therapy , Pyridones/therapeutic use , Triazines/therapeutic use , Animals , Antiviral Agents/pharmacokinetics , Body Temperature/drug effects , Dibenzothiepins/pharmacokinetics , Ferrets , Humans , Influenza, Human/drug therapy , Influenza, Human/metabolism , Microsomes/metabolism , Morpholines/pharmacokinetics , Orthomyxoviridae Infections/metabolism , Oseltamivir/pharmacokinetics , Oseltamivir/therapeutic use , Pyridones/pharmacokinetics , Triazines/pharmacokinetics , Viral Load/drug effects
3.
Sci Rep ; 9(1): 11891, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31417163

ABSTRACT

Neuraminidase inhibitors (NAIs) complement influenza virus infection management by helping to clear virus, alleviate symptoms, and reduce transmission. In a previous randomised study, we examined the effect of 4 NAIs on virus clearance and influenza symptoms in Japanese paediatric patients. In this second analysis, we examined the effects of NAI treatment on antibody responses and virus clearance, and the relationships between antibody responses and patients' infection histories (previous infection; asymptomatic infection via household members of same virus type/subtype; vaccination), and between infection histories and viral kinetics. Haemagglutination inhibition (HI) antibody responses produced HI titres ≥40 by Day 14 of NAI treatment, in parallel with virus clearance (trend test P = 0.001). Comparing patients with and without influenza infection histories (directly or asymptomatic infection via household members) showed that infection history had a marked positive effect on HI antibody responses in patients vaccinated before the current influenza season (before enrolment). Current virus clearance was significantly faster in patients previously infected with the same virus type/subtype than in those not previously infected, and clearance pattern depended on the NAI. Assessment of anti-influenza effects of antiviral drugs and vaccines should consider virus and antibody dynamics in response to vaccination and natural infection histories.


Subject(s)
Antibodies, Viral/immunology , Antiviral Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Influenza A virus/drug effects , Influenza A virus/immunology , Influenza, Human/drug therapy , Influenza, Human/immunology , Neuraminidase/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Antibodies, Viral/blood , Antiviral Agents/pharmacology , Child , Drug Therapy, Combination , Enzyme Inhibitors/pharmacology , Host-Pathogen Interactions/immunology , Humans , Influenza A virus/enzymology , Influenza, Human/mortality , Influenza, Human/virology , Prognosis , Treatment Outcome
4.
PLoS One ; 14(5): e0217307, 2019.
Article in English | MEDLINE | ID: mdl-31107922

ABSTRACT

Baloxavir marboxil (BXM) is an orally available small molecule inhibitor of cap-dependent endonuclease (CEN), an essential enzyme in the initiation of mRNA synthesis of influenza viruses. In the present study, we evaluated the efficacy of BXM against influenza virus infection in mouse models. Single-day oral administration of BXM completely prevented mortality due to infection with influenza A and B virus in mice. Moreover, 5-day repeated administration of BXM was more effective for reducing mortality and body weight loss in mice infected with influenza A virus than oseltamivir phosphate (OSP), even when the treatment was delayed up to 96 hours post infection (p.i.). Notably, administration of BXM, starting at 72 hours p.i. led to significant decrease in virus titers of >2-log10 reduction compared to the vehicle control within 24 hours after administration. Virus reduction in the lung was significantly greater than that observed with OSP. In addition, profound and sustained reduction of virus titer was observed in the immunocompromised mouse model without emergence of variants possessing treatment-emergent amino acid substitutions in the target protein. In our immunocompetent and immunocompromised mouse models, delayed treatment with BXM resulted in rapid and potent reduction in infectious virus titer and prevention of signs of influenza infection, suggesting that BXM could extend the therapeutic window for patients with influenza virus infection regardless of the host immune status.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Orthomyxoviridae/drug effects , Oxazines/pharmacology , Pyridines/pharmacology , Thiepins/pharmacology , Triazines/pharmacology , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Dibenzothiepins , Disease Models, Animal , Drug Administration Schedule , Enzyme Inhibitors/administration & dosage , Female , Host Microbial Interactions/drug effects , Host Microbial Interactions/immunology , Humans , Immunocompetence , Immunocompromised Host , Influenza A virus/drug effects , Influenza A virus/physiology , Influenza B virus/drug effects , Influenza B virus/physiology , Influenza, Human/drug therapy , Influenza, Human/immunology , Influenza, Human/virology , Mice , Mice, Inbred BALB C , Morpholines , Orthomyxoviridae/physiology , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Oseltamivir/pharmacology , Oxazines/administration & dosage , Pyridines/administration & dosage , Pyridones , Thiepins/administration & dosage , Triazines/administration & dosage , Virus Replication/drug effects
5.
J Antimicrob Chemother ; 74(3): 654-662, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30476172

ABSTRACT

OBJECTIVES: Baloxavir marboxil (formerly S-033188) is a first-in-class, orally available, cap-dependent endonuclease inhibitor licensed in Japan and the USA for the treatment of influenza virus infection. We evaluated the efficacy of delayed oral treatment with baloxavir marboxil in combination with a neuraminidase inhibitor in a mouse model of lethal influenza virus infection. METHODS: The inhibitory potency of baloxavir acid (the active form of baloxavir marboxil) in combination with neuraminidase inhibitors was tested in vitro. The therapeutic effects of baloxavir marboxil and oseltamivir phosphate, or combinations thereof, were evaluated in mice lethally infected with influenza virus A/PR/8/34; treatments started 96 h post-infection. RESULTS: Combinations of baloxavir acid and neuraminidase inhibitor exhibited synergistic potency against viral replication by means of inhibition of cytopathic effects in vitro. In mice, baloxavir marboxil monotherapy (15 or 50 mg/kg twice daily) significantly and dose-dependently reduced virus titre 24 h after administration and completely prevented mortality, whereas oseltamivir phosphate treatments were not as effective. In this model, a suboptimal dose of baloxavir marboxil (0.5 mg/kg twice daily) in combination with oseltamivir phosphate provided additional efficacy compared with monotherapy in terms of virus-induced mortality, elevation of cytokine/chemokine levels and pathological changes in the lung. CONCLUSIONS: Baloxavir marboxil monotherapy with 96 h-delayed oral dosing achieved drastic reductions in virus titre, inflammatory response and mortality in a mouse model. Combination treatment with baloxavir acid and oseltamivir acid in vitro and baloxavir marboxil and oseltamivir phosphate in mice produced synergistic responses against influenza virus infections, suggesting that treating humans with the combination may be beneficial.


Subject(s)
Antiviral Agents/administration & dosage , Influenza A virus/drug effects , Orthomyxoviridae Infections/drug therapy , Oseltamivir/administration & dosage , Oxazines/administration & dosage , Pyridines/administration & dosage , Thiepins/administration & dosage , Triazines/administration & dosage , Administration, Oral , Animals , Dibenzothiepins , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Mice, Inbred BALB C , Morpholines , Orthomyxoviridae Infections/pathology , Pyridones , Survival Analysis , Treatment Outcome , United States
6.
Antiviral Res ; 160: 109-117, 2018 12.
Article in English | MEDLINE | ID: mdl-30316915

ABSTRACT

Cap-dependent endonuclease (CEN) resides in the PA subunit of the influenza virus and mediates the critical "cap-snatching" step of viral RNA transcription, which is considered to be a promising anti-influenza target. Here, we describe in vitro characterization of a novel CEN inhibitor, baloxavir acid (BXA), the active form of baloxavir marboxil (BXM). BXA inhibits viral RNA transcription via selective inhibition of CEN activity in enzymatic assays, and inhibits viral replication in infected cells without cytotoxicity in cytopathic effect assays. The antiviral activity of BXA is also confirmed in yield reduction assays with seasonal type A and B viruses, including neuraminidase inhibitor-resistant strains. Furthermore, BXA shows broad potency against various subtypes of influenza A viruses (H1N2, H5N1, H5N2, H5N6, H7N9 and H9N2). Additionally, serial passages of the viruses in the presence of BXA result in isolation of PA/I38T variants with reduced BXA susceptibility. Phenotypic and genotypic analyses with reverse genetics demonstrate the mechanism of BXA action via CEN inhibition in infected cells. These results reveal the in vitro characteristics of BXA and support clinical use of BXM to treat influenza.


Subject(s)
Antiviral Agents/pharmacology , Endonucleases/antagonists & inhibitors , Influenza A virus/drug effects , Influenza B virus/drug effects , Oxazines/pharmacology , Pyridines/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Thiepins/pharmacology , Triazines/pharmacology , Viral Proteins/antagonists & inhibitors , Cytopathogenic Effect, Viral , DNA Mutational Analysis , Dibenzothiepins , Drug Resistance, Viral , Endonucleases/genetics , Influenza A virus/enzymology , Influenza A virus/growth & development , Influenza B virus/enzymology , Influenza B virus/growth & development , Microbial Sensitivity Tests , Morpholines , Mutation, Missense , Pyridones , RNA-Dependent RNA Polymerase/genetics , Reverse Genetics , Serial Passage , Transcription, Genetic/drug effects , Viral Proteins/genetics , Virus Replication/drug effects
7.
Article in English | MEDLINE | ID: mdl-29530860

ABSTRACT

Human norovirus (HuNoV) is a major cause of nonbacterial gastroenteritis worldwide, yet despite its impact on society, vaccines and antivirals are currently lacking. A HuNoV replicon system has been widely applied to the evaluation of antiviral compounds and has thus accelerated the process of drug discovery against HuNoV infection. Rupintrivir, an irreversible inhibitor of the human rhinovirus 3C protease, has been reported to inhibit the replication of the Norwalk virus replicon via the inhibition of the norovirus protease. Here we report, for the first time, the generation of rupintrivir-resistant human Norwalk virus replicon cells in vitro Sequence analysis revealed that these replicon cells contained amino acid substitutions of alanine 105 to valine (A105V) and isoleucine 109 to valine (I109V) in the viral protease NS6. The application of a cell-based fluorescence resonance energy transfer (FRET) assay for protease activity demonstrated that these substitutions were involved in the enhanced resistance to rupintrivir. Furthermore, we validated the effect of these mutations using reverse genetics in murine norovirus (MNV), demonstrating that a recombinant MNV strain with a single I109V substitution in the protease also showed reduced susceptibility to rupintrivir. In summary, using a combination of different approaches, we have demonstrated that, under the correct conditions, mutations in the norovirus protease that lead to the generation of resistant mutants can rapidly occur.


Subject(s)
Antiviral Agents/pharmacology , Isoxazoles/pharmacology , Norwalk virus/drug effects , Pyrrolidinones/pharmacology , 3C Viral Proteases , Amino Acid Sequence , Cell Line, Tumor , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Fluorescence Resonance Energy Transfer , Humans , Mutation , Norwalk virus/genetics , Phenylalanine/analogs & derivatives , Valine/analogs & derivatives , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects , Virus Replication/genetics
8.
Antiviral Res ; 117: 52-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25752738

ABSTRACT

INTRODUCTION: Influenza virus infection increases susceptibility to bacterial infection and mortality in humans. Although the efficacy of approved intravenous peramivir, a neuraminidase (NA) inhibitor, against influenza virus infection has been reported, its efficacy against bacterial co-infection, which occurs during the period of viral shedding, was not fully investigated. To further understand the significance of treatment with peramivir, we assessed the efficacy of peramivir against a bacterial co-infection model in mice caused by clinically isolated influenza A(H1N1)pdm09 virus and Streptococcus pneumoniae. METHODS: Mice were infected with influenza A(H1N1)pdm09. Peramivir was intravenously administered after the viral infection. At 2days post viral infection, the mice were infected with S. pneumoniae. Peramivir efficacy was measured by the survival rates and viral titers, bacterial titers, or proinflammatory cytokine concentrations in lung homogenates. RESULTS: Peramivir treatment reduced the mortality of mice infected with influenza virus and S. pneumoniae. The survival rate in the peramivir-treated group was significantly higher than that in the oseltamivir-treated group. Viral titers and proinflammatory cytokine responses in the peramivir-treated group were significantly lower than those in the oseltamivir-treated group until at 2days post viral infection. Bacterial titer was significantly lower in the peramivir-treated group than in the oseltamivir-treated group at 4days post viral infection. CONCLUSION: These results demonstrated that peramivir inhibits viral replication, consequently leading to bacterial clearance and prevention of mortality during severe murine bacterial co-infection, which occurs during the period of viral shedding, with the efficacy of peramivir being superior to that of oseltamivir.


Subject(s)
Antiviral Agents/administration & dosage , Coinfection/drug therapy , Cyclopentanes/administration & dosage , Guanidines/administration & dosage , Influenza A Virus, H1N1 Subtype/drug effects , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/drug therapy , Pneumococcal Infections/complications , Streptococcus pneumoniae/drug effects , Acids, Carbocyclic , Administration, Intravenous , Animals , Bacterial Load/drug effects , Coinfection/mortality , Cytokines/immunology , Disease Models, Animal , Dogs , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/mortality , Oseltamivir/administration & dosage , Pneumococcal Infections/mortality , Viral Load/drug effects , Virus Replication/drug effects
9.
J Immunol ; 194(6): 2673-82, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25681338

ABSTRACT

Cyclodextrins are commonly used as a safe excipient to enhance the solubility and bioavailability of hydrophobic pharmaceutical agents. Their efficacies and mechanisms as drug-delivery systems have been investigated for decades, but their immunological properties have not been examined. In this study, we reprofiled hydroxypropyl-ß-cyclodextrin (HP-ß-CD) as a vaccine adjuvant and found that it acts as a potent and unique adjuvant. HP-ß-CD triggered the innate immune response at the injection site, was trapped by MARCO(+) macrophages, increased Ag uptake by dendritic cells, and facilitated the generation of T follicular helper cells in the draining lymph nodes. It significantly enhanced Ag-specific Th2 and IgG Ab responses as potently as did the conventional adjuvant, aluminum salt (alum), whereas its ability to induce Ag-specific IgE was less than that of alum. At the injection site, HP-ß-CD induced the temporary release of host dsDNA, a damage-associated molecular pattern. DNase-treated mice, MyD88-deficient mice, and TBK1-deficient mice showed significantly reduced Ab responses after immunization with this adjuvant. Finally, we demonstrated that HP-ß-CD-adjuvanted influenza hemagglutinin split vaccine protected against a lethal challenge with a clinically isolated pandemic H1N1 influenza virus, and the adjuvant effect of HP-ß-CD was demonstrated in cynomolgus macaques. Our results suggest that HP-ß-CD acts as a potent MyD88- and TBK1-dependent T follicular helper cell adjuvant and is readily applicable to various vaccines.


Subject(s)
Antigens/immunology , Inflammation/immunology , T-Lymphocytes, Helper-Inducer/immunology , Th2 Cells/immunology , beta-Cyclodextrins/immunology , 2-Hydroxypropyl-beta-cyclodextrin , Adjuvants, Immunologic/administration & dosage , Animals , Antibody Formation/drug effects , Antibody Formation/genetics , Antibody Formation/immunology , Antigens/administration & dosage , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Inflammation/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/metabolism , Macaca fascicularis , Male , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence, Multiphoton , Oligonucleotide Array Sequence Analysis , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Spleen/drug effects , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/metabolism , Th2 Cells/drug effects , Th2 Cells/metabolism , Transcriptome/drug effects , Transcriptome/immunology , beta-Cyclodextrins/administration & dosage
10.
Antivir Ther ; 20(1): 11-9, 2015.
Article in English | MEDLINE | ID: mdl-24517996

ABSTRACT

BACKGROUND: Pneumococcal pneumonia often occurs secondary to influenza infection and accounts for a large proportion of the morbidity and mortality associated with seasonal and pandemic influenza outbreaks. Peramivir is a novel, intravenous neuraminidase inhibitor that exhibits potent antiviral activity against influenza A and B viruses. We investigated the efficacy of peramivir for modulating the severity of secondary pneumococcal pneumonia. METHODS: CBA/JNCrlj mice, infected with influenza virus and superinfected with Streptococcus pneumoniae, were treated with either intravenous peramivir (single or multiple doses of 60 mg/kg/day) or oral oseltamivir at doses of 10 or 40 mg/kg/day in divided doses. The survival rate, viable bacterial count and virus titre in the lungs, as well as cytokine/chemokine concentration and histopathological findings were compared between both groups. RESULTS: The median duration of survival of coinfected mice was significantly prolonged by treatment with multiple doses of peramivir, relative to mice treated with oseltamivir at either dose. Viable bacterial counts and virus titres in the lungs were significantly reduced by intravenous peramivir treatment compared with no treatment or oral oseltamivir treatment. The production of inflammatory cytokines/chemokines was also significantly suppressed by multiple dosing of peramivir compared with oseltamivir. Increased survival appeared to be mediated by decreased inflammation, manifested as lower levels of inflammatory cells and proinflammatory cytokines in the lungs and less severe histopathological findings. The lungs of mice treated with multiple doses of peramivir showed mild inflammatory changes compared to oseltamivir. CONCLUSIONS: This study demonstrated that a multiple-dose regimen of intravenous peramivir was more efficacious than a single peramivir dose or multiple doses of oseltamivir for improving outcomes in pneumococcal pneumonia following influenza virus infection in mice.


Subject(s)
Antiviral Agents/pharmacology , Cyclopentanes/pharmacology , Guanidines/pharmacology , Orthomyxoviridae Infections/drug therapy , Oseltamivir/pharmacology , Pneumonia, Pneumococcal/mortality , Acids, Carbocyclic , Administration, Intravenous , Administration, Oral , Animals , Chemokine CXCL2/biosynthesis , Chemokine CXCL2/immunology , Chemokines/biosynthesis , Chemokines/immunology , Coinfection , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Lung/drug effects , Lung/immunology , Lung/microbiology , Lung/virology , Male , Mice , Mice, Inbred CBA , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/microbiology , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/immunology , Survival Analysis , Treatment Outcome , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
11.
Antiviral Res ; 109: 110-5, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24997412

ABSTRACT

The purpose of this study was to investigate the relationship between pharmacokinetic (PK) parameters of intravenous (IV) peramivir and in vivo antiviral activity pharmacodynamic (PD) outcomes in a mouse model of influenza virus infection. Peramivir was administrated to mice in three dosing schedules; once, twice and four times after infection of A/WS/33 (H1N1). The survival rate at day 14 after virus infection was employed as the antiviral activity outcome for analysis. The relationship between day 14 survival and PK parameters, including area under the concentration-time curve (AUC), maximum concentration (Cmax) and time that drug concentration exceeds IC95 (T(>IC95)), was estimated using a logistic regression model, and model fitness was evaluated by calculation of the Akaike information criterion (AIC) index. The AIC indices of AUC, Cmax and T(>IC95) were about 114, 151 and 124, respectively. The AIC of AUC and T(>IC95) were smaller than that of Cmax. Therefore, both AUC and T(>IC95) were the PK parameters that correlated best with the antiviral activity of peramivir IV against influenza virus infection in mice.


Subject(s)
Antiviral Agents/pharmacokinetics , Cyclopentanes/pharmacokinetics , Guanidines/pharmacokinetics , Influenza, Human/drug therapy , Acids, Carbocyclic , Animals , Antiviral Agents/administration & dosage , Cyclopentanes/administration & dosage , Disease Models, Animal , Female , Guanidines/administration & dosage , Humans , Influenza, Human/virology , Mice , Mice, Inbred BALB C
12.
PLoS Pathog ; 10(6): e1004192, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24945244

ABSTRACT

Highly pathogenic avian influenza (HPAI) viruses of the H5N1 subtype often cause severe pneumonia and multiple organ failure in humans, with reported case fatality rates of more than 60%. To develop a clinical antibody therapy, we generated a human-mouse chimeric monoclonal antibody (MAb) ch61 that showed strong neutralizing activity against H5N1 HPAI viruses isolated from humans and evaluated its protective potential in mouse and nonhuman primate models of H5N1 HPAI virus infections. Passive immunization with MAb ch61 one day before or after challenge with a lethal dose of the virus completely protected mice, and partial protection was achieved when mice were treated 3 days after the challenge. In a cynomolgus macaque model, reduced viral loads and partial protection against lethal infection were observed in macaques treated with MAb ch61 intravenously one and three days after challenge. Protective effects were also noted in macaques under immunosuppression. Though mutant viruses escaping from neutralization by MAb ch61 were recovered from macaques treated with this MAb alone, combined treatment with MAb ch61 and peramivir reduced the emergence of escape mutants. Our results indicate that antibody therapy might be beneficial in reducing viral loads and delaying disease progression during H5N1 HPAI virus infection in clinical cases and combined treatment with other antiviral compounds should improve the protective effects of antibody therapy against H5N1 HPAI virus infection.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Immunization, Passive/methods , Influenza A Virus, H5N1 Subtype/immunology , Orthomyxoviridae Infections/therapy , Acids, Carbocyclic , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antiviral Agents/therapeutic use , Cell Line , Cyclopentanes/therapeutic use , Dogs , Drug Therapy, Combination , Female , Guanidines/therapeutic use , Immunocompromised Host/immunology , Influenza A Virus, H5N1 Subtype/isolation & purification , Interleukin-6/blood , Lung/pathology , Lung/virology , Macaca fascicularis , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Models, Animal , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Viral Load/immunology
13.
Antimicrob Agents Chemother ; 58(8): 4795-803, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24913156

ABSTRACT

Highly pathogenic avian influenza A (H5N1) viruses cause severe and often fatal disease in humans. We evaluated the efficacy of repeated intravenous dosing of the neuraminidase inhibitor peramivir against highly pathogenic avian influenza virus A/Vietnam/UT3040/2004 (H5N1) infection in cynomolgus macaques. Repeated dosing of peramivir (30 mg/kg/day once a day for 5 days) starting immediately after infection significantly reduced viral titers in the upper respiratory tract, body weight loss, and cytokine production and resulted in a significant body temperature reduction in infected macaques compared with that of macaques administered a vehicle (P < 0.05). Repeated administration of peramivir starting at 24 h after infection also resulted in a reduction in viral titers and a reduction in the period of virus detection in the upper respiratory tract, although the body temperature change was not statistically significant. The macaque model used in the present study demonstrated that inhibition of viral replication at an early time point after infection by repeated intravenous treatment with peramivir is critical for reduction of the production of cytokines, i.e., interleukin-6 (IL-6), tumor necrosis factor α, gamma interferon, monocyte chemotactic protein 1, and IL-12p40, resulting in amelioration of symptoms caused by highly pathogenic avian influenza virus infection.


Subject(s)
Antiviral Agents/pharmacology , Cyclopentanes/pharmacology , Guanidines/pharmacology , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/pathogenicity , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/veterinary , Acids, Carbocyclic , Administration, Intravenous , Animals , Body Temperature/drug effects , Body Weight/drug effects , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/biosynthesis , Drug Administration Schedule , Female , Influenza A Virus, H5N1 Subtype/physiology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interleukin-12 Subunit p40/antagonists & inhibitors , Interleukin-12 Subunit p40/biosynthesis , Interleukin-6/antagonists & inhibitors , Interleukin-6/biosynthesis , Macaca fascicularis , Orthomyxoviridae Infections/physiopathology , Orthomyxoviridae Infections/virology , Time Factors , Treatment Outcome , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Virulence , Virus Replication/drug effects
14.
Vaccine ; 32(25): 3004-9, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24721532

ABSTRACT

BACKGROUND: Synthetic hemozoin (sHZ, also known as ß-hematin) from monomeric heme is a particle adjuvant which activates antigen-presenting cells (APCs), such as dendritic cells and macrophages, and enhances humoral immune responses to several antigens, including ovalbumin, human serum albumin, and serine repeat antigen 36 of Plasmodium falciparum. In the present study, we evaluated the adjuvanticity and pyrogenicity of sHZ as an adjuvant for seasonal trivalent hemagglutinin split vaccine (SV) for humans using the experimental ferret model. METHOD: Ferrets were twice immunized with trivalent SV, SV with sHZ (SV/sHZ) or Fluad, composed of trivalent SV with MF59. Serum hemagglutination inhibition (HI) titers against three viral hemagglutinin (HA) antigens were measured at every week after the immunization. The pyrogenicity of SV/sHZ was examined by monitoring the body temperature of the immunized ferrets. To evaluate the protective efficacy of SV/sHZ, the immunized ferrets were challenged with influenza virus B infection, followed by measurement of viral titers in the nasal cavity and body temperature. RESULTS: sHZ enhanced HI titers against three viral HA antigens in a dose-dependent manner, to an extent comparable to that of Fluad. The highest dose of sHZ (800 µg) immunized with SV conferred sterile protection against infection with heterologous Influenza B virus, without causing any pyrogenic reaction such as high fever. CONCLUSION: In the present study, sHZ enhanced the protective efficacy of SV against influenza infection without inducing pyrogenic reaction, suggesting sHZ to be a promising adjuvant candidate for human SV.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemeproteins/administration & dosage , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Viral/blood , Body Temperature , Female , Ferrets , Hemagglutination Inhibition Tests , Influenza B virus
15.
PLoS One ; 8(12): e82740, 2013.
Article in English | MEDLINE | ID: mdl-24376571

ABSTRACT

H5N1 highly pathogenic avian influenza virus (HPAIV) infection has been reported in poultry and humans with expanding clade designations. Therefore, a vaccine that induces immunity against a broad spectrum of H5N1 viruses is preferable for pandemic preparedness. We established a second H5N1 vaccine candidate, A/duck/Hokkaido/Vac-3/2007 (Vac-3), in our virus library and examined the efficacy of inactivated whole particles of this strain against two clades of H5N1 HPAIV strains that caused severe morbidity in cynomolgus macaques. Virus propagation in vaccinated macaques infected with either of the H5N1 HPAIV strains was prevented compared with that in unvaccinated macaques. This vaccine also prevented propagation of a pandemic (H1N1) 2009 virus in macaques. In the vaccinated macaques, neutralization activity, which was mainly shown by anti-hemagglutinin antibody, against H5N1 HPAIVs in plasma was detected, but that against H1N1 virus was not detected. However, neuraminidase inhibition activity in plasma and T-lymphocyte responses in lymph nodes against H1N1 virus were detected. Therefore, cross-clade and heterosubtypic protective immunity in macaques consisted of humoral and cellular immunity induced by vaccination with Vac-3.


Subject(s)
Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza Vaccines/immunology , Influenza, Human/epidemiology , Macaca fascicularis/virology , Orthomyxoviridae Infections/prevention & control , Vaccines, Inactivated/immunology , Animals , Antibodies, Viral/immunology , Antibody Formation/immunology , Birds/virology , Body Temperature , Female , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Macaca fascicularis/immunology , Neuraminidase/antagonists & inhibitors , Neuraminidase/metabolism , Neutralization Tests , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Pandemics , T-Lymphocytes/immunology , Vaccination , Viral Load
16.
PLoS One ; 8(9): e75910, 2013.
Article in English | MEDLINE | ID: mdl-24086663

ABSTRACT

Pandemic (H1N1) 2009 influenza virus spread throughout the world since most people did not have immunity against the virus. In the post pandemic phase when many humans might possess immunity against the pandemic virus, one of the concerns is infection in immunocompromised people. Therefore, we used an immunosuppressed macaque model to examine pathogenicity of the pandemic (H1N1) 2009 virus under an immunocompromised condition. The virus in nasal samples of immunosuppressed macaques infected with the pandemic (H1N1) 2009 virus was detected longer after infection than was the virus in nasal samples of immunocompetent macaques. As expected, not only virus amounts but also virus propagation sites in the immunosuppressed macaques were larger than those in lungs of the immunocompetent macaques when they were infected with the pandemic virus. Immunosuppressed macaques possessed low levels of immune cells producing cytokines and chemokines, but levels of inflammatory cytokines/chemokine interleukin (IL)-6, IL-18, and monocyte chemotactic protein (MCP)-1 in lungs of the immunosuppressed macaques were higher than those in lungs of the immunocompetent macaques, though the differences were not statistically significant. Therefore, under an immunosuppressive condition, the pandemic influenza (H1N1) 2009 virus might cause more severe morbidity with high cytokine/chemokine production by the host innate immune system than that seen in macaques under the immunocompetent condition.


Subject(s)
Immunocompromised Host/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Animals , Cyclophosphamide/pharmacology , Cyclosporine/pharmacology , Cytokines/immunology , Immunohistochemistry , Immunosuppression Therapy/methods , Lung/immunology , Macaca fascicularis , Virus Replication/physiology
17.
Antimicrob Agents Chemother ; 57(5): 2286-94, 2013 May.
Article in English | MEDLINE | ID: mdl-23478960

ABSTRACT

The efficacy of intravenous peramivir against influenza A (H1N1) 2009 virus infection was evaluated in mice in which the immune system was suppressed by cyclophosphamide (CP) treatment. The mortality rate of the vehicle control group was 100%, and the mice lost 20% of their body weight on average by day 13 postinfection (p.i.). Repeated administration of peramivir (40 mg/kg of body weight once a day, given intravenously for 20 days), starting at 1 h p.i., significantly reduced mortality, body weight loss, viral titers, and cytokine production in infected mice compared with results for administration of vehicle (P < 0.01). In addition, repeated administration of peramivir, starting at 24 h, 48 h, or 72 h p.i., also resulted in increases in survival rates and reduction of viral titers in the lungs (P < 0.01). The mean days to death (MDD) of the vehicle group was 14.5 days, while in the groups treated with peramivir starting at 24 h, 48 h, and 72 h p.i., the MDDs were >23.0, 20.9, and 21.8 days, respectively. In comparison, repeated administration of oseltamivir phosphate (5 mg/kg twice a day, given orally for 20 days), starting at 24 h, 48 h, and 72 h p.i., also significantly prevented body weight loss, whereas no significant differences in mortality rates and viral titers in the lungs were observed compared with results for the vehicle group. These data indicated that repeated administration of peramivir was effective in promoting the survival and reducing virus replication in immunosuppressed mice infected with influenza A (H1N1) 2009 virus.


Subject(s)
Antiviral Agents/pharmacology , Cyclopentanes/pharmacology , Guanidines/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Lung/drug effects , Orthomyxoviridae Infections/drug therapy , Oseltamivir/analogs & derivatives , Phosphorous Acids/pharmacology , Acids, Carbocyclic , Animals , Body Weight/drug effects , Cyclophosphamide/pharmacology , Drug Administration Schedule , Female , Immunosuppressive Agents/pharmacology , Influenza A Virus, H1N1 Subtype/growth & development , Injections, Intravenous , Lung/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Oseltamivir/pharmacology , Survival Analysis , Treatment Outcome
18.
Antimicrob Agents Chemother ; 55(11): 4961-70, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21844317

ABSTRACT

We evaluated the efficacy of a single intravenous dose peramivir for treatment of influenza B virus infection in ferrets and cynomolgus macaques in the present study. A single dose of peramivir (60 mg/kg of body weight) given to ferrets on 1 day postinfection with influenza B virus significantly reduced median area under the curve (AUC) virus titers (peramivir, 8.3 log(10) 50% tissue culture infective doses [TCID(50)s] · day/ml; control, 10.7 log(10) TCID(50)s · day/ml; P < 0.0001). Furthermore, nasal virus titers on day 2 postinfection in ferrets receiving a single injection of peramivir (30 mg/kg) and AUCs of the body temperature increase in ferrets receiving a single injection of peramivir (30 and 60 mg/kg) were lower than those in ferrets administered oral oseltamivir phosphate (30 and 60 mg/kg/day twice daily for 3 days). In macaques infected with influenza B virus, viral titers in the nasal swab fluid on days 2 and 3 postinfection and body temperature after a single injection of peramivir (30 mg/kg) were lower than those after oral administration of oseltamivir phosphate (30 mg/kg/day for 5 days). The two animal models used in the present study demonstrated that inhibition of viral replication at the early time point after infection was critical in reduction of AUCs of virus titers and interleukin-6 production, resulting in amelioration of symptoms. Our results shown in animal models suggest that the early treatment with a single intravenous injection of peramivir is clinically recommended to reduce symptoms effectively in influenza B virus infection.


Subject(s)
Cyclopentanes/therapeutic use , Ferrets/virology , Guanidines/therapeutic use , Influenza B virus/drug effects , Influenza B virus/pathogenicity , Macaca/virology , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Acids, Carbocyclic , Animals , Cyclopentanes/administration & dosage , Guanidines/administration & dosage , Injections, Intravenous
19.
Virology ; 407(2): 178-84, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-20822788

ABSTRACT

Pathogenicity of influenza B virus was examined in cynomolgus macaques to establish a macaque model suitable for vaccine and antiviral drug development. We prepared influenza B viruses for inoculation with minimal passages after isolation from patients. Macaques inoculated with influenza B virus showed higher body temperature than that before infection for 6 to 12 days. Virus was detected in nasal, tracheal, and bronchial samples until 6 days after inoculation followed by an increase in neutralizing antibody. High levels of IL-6 and TNF-α in nasal swabs from the infected macaques were correlated with fever. Symptoms and duration of the viral replication would be sufficient to evaluate efficacy of vaccines and antiviral agents. In addition, measurement of immune responses including antibody and cytokine production would provide an immunological rationale in efficacy of vaccines and antiviral agents. The results suggest that cynomolgus macaques are appropriate model animals for research of influenza B virus.


Subject(s)
Disease Models, Animal , Influenza B virus/pathogenicity , Influenza, Human/physiopathology , Macaca fascicularis , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cytokines/metabolism , Humans , Influenza B virus/immunology , Influenza B virus/isolation & purification , Influenza, Human/immunology , Influenza, Human/virology , Virus Replication
20.
Hum Immunol ; 71(2): 123-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19891998

ABSTRACT

Most escape mutations have been identified on cytotoxic T lymphocyte (CTL) epitopes presented by Caucasian or African human leukocyte antigen (HLA) class I alleles, whereas a limited number of studies have identified the escape mutations on epitopes presented by Asian alleles. HLA-B54 is a common HLA allele in Asian countries. We recently identified five HLA-B*5401-restricted HIV-1-specific CTL epitopes. We here investigated escape mutations in these CTL epitopes in Japanese HIV-1-infected individuals. The frequency of substitution from Glu (E) to Asp (D) at position 7 (FV9-7D) in the Pol 154-162 (FV9) epitope was significantly higher in HLA-B*5401(+) HIV-infected individuals than in HLA-B*5401(-) individuals, whereas substitutions that were significantly higher in HLA-B*5401(+) individuals than in HLA-B*5401(-) individuals were not found in the other four epitopes. FV9-specific CTLs showed reduced killing activity against target cells pulsed with the FV9-7D mutant peptide and failed to kill those infected with the FV9-7D mutant virus, strongly suggesting that FV9-7D is an escape mutant. Furthermore, longitudinal sequence analysis of the FV9 epitope in two HLA-B*5401(+) individuals revealed that the sequence had changed from the wild type to the FV9-7D during the clinical course. Taken together, these results indicate that the FV9-7D escape mutant had been selected by FV9-specific CTLs among chronically HIV-1-infected HLA-B*5401(+) individuals.


Subject(s)
Epitopes, T-Lymphocyte/genetics , HIV Infections/genetics , HIV Infections/immunology , HLA-B Antigens/genetics , T-Lymphocytes, Cytotoxic/immunology , Chronic Disease , Epitopes, T-Lymphocyte/immunology , HIV-1/genetics , HIV-1/immunology , HLA-B Antigens/immunology , Humans , Mutation , T-Lymphocytes, Cytotoxic/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...