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










Publication year range
1.
Arch Virol ; 169(2): 29, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38216710

ABSTRACT

Genetic reassortment of avian, swine, and human influenza A viruses (IAVs) poses potential pandemic risks. Surveillance is important for influenza pandemic preparedness, but the susceptibility of zoonotic IAVs to the cap-dependent endonuclease inhibitor baloxavir acid (BXA) has not been thoroughly researched. Although an amino acid substitution at position 38 in the polymerase acidic protein (PA/I38) in seasonal IAVs reduces BXA susceptibility, PA polymorphisms at position 38 are rarely seen in zoonotic IAVs. Here, we examined the impact of PA/I38 substitutions on the BXA susceptibility of recombinant A(H5N1) viruses. PA mutants that harbored I38T, F, and M were 48.2-, 24.0-, and 15.5-fold less susceptible, respectively, to BXA than wild-type A(H5N1) but were susceptible to the neuraminidase inhibitor oseltamivir acid and the RNA polymerase inhibitor favipiravir. PA mutants exhibited significantly impaired replicative fitness in Madin-Darby canine kidney cells at 24 h postinfection. In addition, in order to investigate new genetic markers for BXA susceptibility, we screened geographically and temporally distinct IAVs isolated worldwide from birds and pigs. The results showed that BXA exhibited antiviral activity against avian and swine viruses with similar levels to seasonal isolates. All viruses tested in the study lacked the PA/I38 substitution and were susceptible to BXA. Isolates harboring amino acid polymorphisms at positions 20, 24, and 37, which have been implicated in the binding of BXA to the PA endonuclease domain, were also susceptible to BXA. These results suggest that monitoring of the PA/I38 substitution in animal-derived influenza viruses is important for preparedness against zoonotic influenza virus outbreaks.


Subject(s)
Dibenzothiepins , Influenza A Virus, H5N1 Subtype , Influenza A virus , Influenza, Human , Morpholines , Orthomyxoviridae , Pyridones , Thiepins , Triazines , Animals , Dogs , Humans , Swine , Influenza A virus/genetics , Oxazines/pharmacology , Pyridines/pharmacology , Pyridines/therapeutic use , Influenza A Virus, H5N1 Subtype/genetics , Thiepins/pharmacology , Thiepins/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Orthomyxoviridae/genetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Amino Acid Substitution , Endonucleases/genetics , Drug Resistance, Viral/genetics
2.
Viruses ; 15(11)2023 Nov 16.
Article in English | MEDLINE | ID: mdl-38005940

ABSTRACT

Influenza remains a worldwide health concern. Antiviral drugs are considered as one of the useful options for its prevention as a complementary measure to vaccination. Baloxavir acid selectively inhibits the cap-dependent endonuclease of influenza viruses and exhibits marked viral titre reduction in patients. Here, we describe the prophylactic potency of baloxavir acid against lethal infection with influenza A and B viruses in mice. BALB/c mice were subcutaneously administered once with baloxavir acid suspension, or orally administered once daily for 10 days with oseltamivir phosphate solution at human relevant doses. Next, the mice were intranasally inoculated with A/PR/8/34 (H1N1) or B/Hong Kong/5/72 strain at 24 to 96 h after the initial dosing. Prophylactic treatment with the antiviral drugs significantly reduced the lung viral titres and prolonged survival time. In particular, baloxavir acid showed a greater suppressive effect on lung viral titres compared to oseltamivir phosphate. In this model, baloxavir acid maintained significant prophylactic effects against influenza A and B virus infections when the plasma concentration at the time of infection was at least 0.88 and 3.58 ng/mL, respectively. The significant prophylactic efficacy observed in our mouse model suggests the potential utility of baloxavir marboxil for prophylaxis against influenza in humans.


Subject(s)
Herpesvirus 1, Cercopithecine , Influenza A Virus, H1N1 Subtype , Influenza, Human , Thiepins , Humans , Animals , Mice , Influenza, Human/drug therapy , Influenza, Human/prevention & control , Oseltamivir/pharmacology , Oseltamivir/therapeutic use , Oxazines/therapeutic use , Pyridines/therapeutic use , Thiepins/pharmacology , Thiepins/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Mice, Inbred BALB C , Phosphates
3.
Viruses ; 15(5)2023 05 11.
Article in English | MEDLINE | ID: mdl-37243240

ABSTRACT

Although the prevalence of polymerase acidic (PA)/I38T strains of influenza virus with reduced susceptibility to baloxavir acid is low, there is a possibility of emergence under selective pressure. Furthermore, the virus may be transmitted between humans. We investigated the in vivo efficacy of baloxavir acid and oseltamivir phosphate against influenza A subtypes H1N1, H1N1pdm09, and H3N2, with PA/I38T substitution, at doses simulating human plasma concentrations. A pharmacokinetic/pharmacodynamic analysis was performed to strengthen the validity of the findings and the applicability in a clinical setting. Although the antiviral effect of baloxavir acid was attenuated in mice infected with PA/I38T-substituted viral strains compared with the wild type (WT), baloxavir acid significantly reduced virus titers at higher-but clinically relevant-doses. The virus titer reduction with baloxavir acid (30 mg/kg subcutaneous single dose) was comparable to that of oseltamivir phosphate (5 mg/kg orally twice daily) against H1N1 and H1N1pdm09 PA/I38T strains in mice, as well as the H3N2 PA/I38T strain in hamsters. Baloxavir acid demonstrated an antiviral effect against PA/I38T-substituted strains, at day 6, with no further viral rebound. In conclusion, baloxavir acid demonstrated dose-dependent antiviral effects comparable to that of oseltamivir phosphate, even though the degree of lung virus titer reduction was diminished in animal models infected with PA/I38T-substituted strains.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A virus , Influenza, Human , Thiepins , Humans , Animals , Mice , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Oseltamivir/pharmacology , Oseltamivir/therapeutic use , Oxazines/pharmacology , Pyridines/pharmacology , Influenza A Virus, H3N2 Subtype , Thiepins/pharmacology , Thiepins/therapeutic use , Drug Resistance, Viral , Nucleotidyltransferases , Phosphates
4.
Proc Natl Acad Sci U S A ; 119(36): e2206104119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037386

ABSTRACT

Viral hemorrhagic fevers caused by members of the order Bunyavirales comprise endemic and emerging human infections that are significant public health concerns. Despite the disease severity, there are few therapeutic options available, and therefore effective antiviral drugs are urgently needed to reduce disease burdens. Bunyaviruses, like influenza viruses (IFVs), possess a cap-dependent endonuclease (CEN) that mediates the critical cap-snatching step of viral RNA transcription. We screened compounds from our CEN inhibitor (CENi) library and identified specific structural compounds that are 100 to 1,000 times more active in vitro than ribavirin against bunyaviruses, including Lassa virus, lymphocytic choriomeningitis virus (LCMV), and Junin virus. To investigate their inhibitory mechanism of action, drug-resistant viruses were selected in culture. Whole-genome sequencing revealed that amino acid substitutions in the CEN region of drug-resistant viruses were located in similar positions as those of the CEN α3-helix loop of IFVs derived under drug selection. Thus, our studies suggest that CENi compounds inhibit both bunyavirus and IFV replication in a mechanistically similar manner. Structural analysis revealed that the side chain of the carboxyl group at the seventh position of the main structure of the compound was essential for the high antiviral activity against bunyaviruses. In LCMV-infected mice, the compounds significantly decreased blood viral load, suppressed symptoms such as thrombocytopenia and hepatic dysfunction, and improved survival rates. These data suggest a potential broad-spectrum clinical utility of CENis for the treatment of both severe influenza and hemorrhagic diseases caused by bunyaviruses.


Subject(s)
Antiviral Agents , Endonucleases , Orthobunyavirus , Animals , Antiviral Agents/pharmacology , Drug Evaluation, Preclinical , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Endonucleases/antagonists & inhibitors , Humans , Mice , Orthobunyavirus/drug effects , Orthobunyavirus/genetics , Orthobunyavirus/metabolism , Virus Replication/drug effects
5.
Viruses ; 14(1)2022 01 08.
Article in English | MEDLINE | ID: mdl-35062315

ABSTRACT

Human infections caused by the H5 highly pathogenic avian influenza virus (HPAIV) sporadically threaten public health. The susceptibility of HPAIVs to baloxavir acid (BXA), a new class of inhibitors for the influenza virus cap-dependent endonuclease, has been confirmed in vitro, but it has not yet been fully characterized. Here, the efficacy of BXA against HPAIVs, including recent H5N8 variants, was assessed in vitro. The antiviral efficacy of baloxavir marboxil (BXM) in H5N1 virus-infected mice was also investigated. BXA exhibited similar in vitro activities against H5N1, H5N6, and H5N8 variants tested in comparison with seasonal and other zoonotic strains. Compared with oseltamivir phosphate (OSP), BXM monotherapy in mice infected with the H5N1 HPAIV clinical isolate, the A/Hong Kong/483/1997 strain, also caused a significant reduction in viral titers in the lungs, brains, and kidneys, thereby preventing acute lung inflammation and reducing mortality. Furthermore, compared with BXM or OSP monotherapy, combination treatments with BXM and OSP using a 48-h delayed treatment model showed a more potent effect on viral replication in the organs, accompanied by improved survival. In conclusion, BXM has a potent antiviral efficacy against H5 HPAIV infections.


Subject(s)
Dibenzothiepins/pharmacology , Influenza A virus/drug effects , Morpholines/pharmacology , Orthomyxoviridae Infections/drug therapy , Pyridones/pharmacology , Triazines/pharmacology , A549 Cells , Animals , Antiviral Agents/pharmacology , Chemokines/metabolism , Cytokines/metabolism , Drug Therapy, Combination , Female , Humans , Influenza A Virus, H5N1 Subtype/drug effects , Lung/pathology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Oseltamivir/pharmacology , Pneumonia/drug therapy , Sequence Analysis , Virus Replication/drug effects
6.
J Infect Chemother ; 27(8): 1223-1229, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34045119

ABSTRACT

INTRODUCTION: Baloxavir marboxil is an oral anti-influenza drug with demonstrated safety and efficacy in pediatric patients when a 2% granules formulation is administered at 1 mg/kg. This study assessed safety, effectiveness, and pharmacokinetics of a higher dose (2 mg/kg) of baloxavir marboxil 2% granules in pediatric patients weighing <20 kg. METHODS: This multicenter, open-label, noncontrolled study was conducted at 15 sites in Japan (January 2019-March 2020; JapicCTI-194577). Patients aged <12 years with confirmed influenza received a single oral dose of baloxavir marboxil at 2 mg/kg if body weight was <10 kg or 20 mg if ≥ 10 to <20 kg. Safety, pharmacokinetics, effectiveness (time to illness alleviation [TTIA] of influenza; time to resolution of fever; virus titer), and polymerase acidic protein (PA) substituted viruses were assessed over 22 days. RESULTS: 45 patients, all aged ≤6 years, were enrolled. Adverse events were reported in 24 (53.3%) patients, most commonly nasopharyngitis, diarrhea, and upper respiratory tract infection. Median (95% confidence interval [CI]) TTIA was 37.8 (27.5-46.7) hours; median (95% CI) time to resolution of fever was 22.0 (20.2-28.6) hours. A >4 log decrease in mean viral titer occurred at day 2 and a subsequent temporary 1-2 log increase in patients with influenza A(H3N2) and B. Treatment-emergent PA/I38X-substituted virus was detected in 16/39 (41.0%) patients, but no prolonged TTIA or time to resolution of fever was associated with its presence. CONCLUSIONS: Baloxavir granules administered at 2 mg/kg in children <20 kg were well tolerated, with symptom alleviation similar to 1 mg/kg.


Subject(s)
Dibenzothiepins , Influenza, Human , Antiviral Agents/adverse effects , Child , Dibenzothiepins/therapeutic use , Humans , Influenza A Virus, H3N2 Subtype , Influenza, Human/drug therapy , Japan , Morpholines/therapeutic use , Oxazines , Pyridones/therapeutic use , Triazines
7.
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
8.
Bioorg Med Chem Lett ; 30(22): 127547, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32927030

ABSTRACT

This work describes a set of discovery research studies of an influenza cap-dependent endonuclease (CEN) inhibitor with a carbamoyl pyridone bicycle (CAB) scaffold. Using influenza CEN inhibitory activity, antiviral activity and pharmacokinetic (PK) parameters as indices, structure activity relationships (SAR) studies were performed at the N-1 and N-3 positions on the CAB scaffold, which is a unique template to bind two metals. The hydrophobic substituent at the N-1 position is extremely important for CEN inhibitory activity and antiviral activity, and dihydrodibenzothiepine is the most promising pharmacophore. The compound (S)-13i showed potent virus titer reduction over oseltamivir phosphate in an in vivo mouse model. The CAB compound described herein served as the lead compound of baloxavir marboxil with a tricyclic scaffold, which was approved in Japan and the USA in 2018.


Subject(s)
Antiviral Agents/pharmacology , Endonucleases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Orthomyxoviridae/drug effects , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Endonucleases/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Hydrophobic and Hydrophilic Interactions , Microbial Sensitivity Tests , Molecular Structure , Orthomyxoviridae/enzymology , Structure-Activity Relationship
9.
N Engl J Med ; 383(4): 309-320, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32640124

ABSTRACT

BACKGROUND: Baloxavir marboxil (baloxavir) is a polymerase acidic protein (PA) endonuclease inhibitor with clinical efficacy in the treatment of uncomplicated influenza, including in outpatients at increased risk for complications. The postexposure prophylactic efficacy of baloxavir in the household setting is unclear. METHODS: We conducted a multicenter, double-blind, randomized, placebo-controlled trial to evaluate the postexposure prophylactic efficacy of baloxavir in household contacts of index patients with confirmed influenza during the 2018-2019 season in Japan. The participants were assigned in a 1:1 ratio to receive either a single dose of baloxavir or placebo. The primary end point was clinical influenza, as confirmed by reverse-transcriptase-polymerase-chain-reaction testing, over a period of 10 days. The occurrence of baloxavir-selected PA substitutions associated with reduced susceptibility was assessed. RESULTS: A total of 752 household contacts of 545 index patients were randomly assigned to receive baloxavir or placebo. Among the index patients, 95.6% had influenza A virus infection, 73.6% were younger than 12 years of age, and 52.7% received baloxavir. Among the participants who could be evaluated (374 in the baloxavir group and 375 in the placebo group), the percentage in whom clinical influenza developed was significantly lower in the baloxavir group than in the placebo group (1.9% vs. 13.6%) (adjusted risk ratio, 0.14; 95% confidence interval [CI], 0.06 to 0.30; P<0.001). Baloxavir was effective in high-risk, pediatric, and unvaccinated subgroups of participants. The risk of influenza infection, regardless of symptoms, was lower with baloxavir than with placebo (adjusted risk ratio, 0.43; 95% CI, 0.32 to 0.58). The incidence of adverse events was similar in the two groups (22.2% in the baloxavir group and 20.5% in the placebo group). In the baloxavir group, the viral PA substitutions I38T/M or E23K were detected in 10 (2.7%) and 5 (1.3%) participants, respectively. No transmission of these variants from baloxavir-treated index patients to participants in the placebo group was detected; however, several instances of transmission to participants in the baloxavir group could not be ruled out. CONCLUSIONS: Single-dose baloxavir showed significant postexposure prophylactic efficacy in preventing influenza in household contacts of patients with influenza. (Funded by Shionogi; Japan Primary Registries Network number, JapicCTI-184180.).


Subject(s)
Antiviral Agents/therapeutic use , Disease Transmission, Infectious/prevention & control , Influenza A virus , Influenza, Human/prevention & control , Oxazines/therapeutic use , Pyridines/therapeutic use , Thiepins/therapeutic use , Triazines/therapeutic use , Administration, Oral , Adolescent , Adult , Aged , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Child , Child, Preschool , Dibenzothiepins , Double-Blind Method , Endonucleases/antagonists & inhibitors , Family , Female , Humans , Influenza A virus/genetics , Influenza A virus/isolation & purification , Influenza, Human/transmission , Influenza, Human/virology , Intention to Treat Analysis , Male , Middle Aged , Morpholines , Oxazines/administration & dosage , Oxazines/adverse effects , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridones , Reverse Transcriptase Polymerase Chain Reaction , Thiepins/administration & dosage , Thiepins/adverse effects , Triazines/administration & dosage , Triazines/adverse effects
10.
Influenza Other Respir Viruses ; 14(3): 353-357, 2020 05.
Article in English | MEDLINE | ID: mdl-31994309

ABSTRACT

Baloxavir marboxil (BXM) demonstrated a rapid and profound decline in infectious viral titer 1 day after BXM administration. Rapid reduction in virus titer is a characteristic of BXM. There may be a possibility that drug carryover effects have impacts on the observed antiviral effects due to the poor correlation that was observed between viral titer reduction and alleviation of influenza symptoms. Here, we report possible carryover effects of baloxavir acid (BXA), an active form of BXM, on infectious titer testing. Our findings indicate that there is little impact of BXA carryover on the infectious titer testing.


Subject(s)
Antiviral Agents/administration & dosage , Dibenzothiepins/administration & dosage , Influenza, Human/drug therapy , Morpholines/administration & dosage , Nasopharynx/virology , Orthomyxoviridae/drug effects , Pharynx/virology , Pyridones/administration & dosage , Triazines/administration & dosage , Humans , Influenza, Human/diagnosis , Influenza, Human/virology , Orthomyxoviridae/genetics , Orthomyxoviridae/growth & development , Orthomyxoviridae/physiology
11.
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
12.
Sci Rep ; 9(1): 3466, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30837531

ABSTRACT

Human infections with avian-origin influenza A(H7N9) virus represent a serious threat to global health; however, treatment options are limited. Here, we show the inhibitory effects of baloxavir acid (BXA) and its prodrug baloxavir marboxil (BXM), a first-in-class cap-dependent endonuclease inhibitor, against A(H7N9), in vitro and in vivo. In cell culture, BXA at four nanomolar concentration achieved a 1.5-2.8 log reduction in virus titers of A(H7N9), including the NA-R292K mutant virus and highly pathogenic avian influenza viruses, whereas NA inhibitors or favipiravir required approximately 20-fold or higher concentrations to achieve the same levels of reduction. A(H7N9)-specific amino acid polymorphism at position 37, implicated in BXA binding to the PA endonuclease domain, did not impact on BXA susceptibility. In mice, oral administration of BXM at 5 and 50 mg/kg twice a day for 5 days completely protected from a lethal A/Anhui/1/2013 (H7N9) challenge, and reduced virus titers more than 2-3 log in the lungs. Furthermore, the potent therapeutic effects of BXM in mice were still observed when a higher virus dose was administered or treatment was delayed up to 48 hours post infection. These findings support further investigation of BXM for A(H7N9) treatment in humans.


Subject(s)
Antiviral Agents/pharmacology , Endonucleases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Influenza A Virus, H7N9 Subtype/drug effects , Influenza A Virus, H7N9 Subtype/enzymology , Oxazines/pharmacology , Pyridines/pharmacology , Thiepins/pharmacology , Triazines/pharmacology , Virus Replication/drug effects , Animals , Cytokines/biosynthesis , Dibenzothiepins , Disease Models, Animal , Ducks , Humans , Inflammation Mediators/metabolism , Influenza in Birds/drug therapy , Influenza in Birds/virology , Influenza, Human/drug therapy , Influenza, Human/metabolism , Influenza, Human/virology , Mice , Morpholines , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Pyridones , Viral Load
13.
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
14.
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
15.
Sci Rep ; 8(1): 9633, 2018 06 25.
Article in English | MEDLINE | ID: mdl-29941893

ABSTRACT

Baloxavir acid (BXA), derived from the prodrug baloxavir marboxil (BXM), potently and selectively inhibits the cap-dependent endonuclease within the polymerase PA subunit of influenza A and B viruses. In clinical trials, single doses of BXM profoundly decrease viral titers as well as alleviating influenza symptoms. Here, we characterize the impact on BXA susceptibility and replicative capacity of variant viruses detected in the post-treatment monitoring of the clinical studies. We find that the PA I38T substitution is a major pathway for reduced susceptibility to BXA, with 30- to 50-fold and 7-fold EC50 changes in A and B viruses, respectively. The viruses harboring the I38T substitution show severely impaired replicative fitness in cells, and correspondingly reduced endonuclease activity in vitro. Co-crystal structures of wild-type and I38T influenza A and B endonucleases bound to BXA show that the mutation reduces van der Waals contacts with the inhibitor. A reduced affinity to the I38T mutant is supported by the lower stability of the BXA-bound endonuclease. These mechanistic insights provide markers for future surveillance of treated populations.


Subject(s)
Amino Acid Substitution , Endonucleases/antagonists & inhibitors , Endonucleases/genetics , Enzyme Inhibitors/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/enzymology , Oxazines/pharmacology , Pyridines/pharmacology , Thiepins/pharmacology , Triazines/pharmacology , Animals , Child , Clinical Trials, Phase I as Topic , Dibenzothiepins , Dogs , Drug Resistance, Viral/genetics , Endonucleases/chemistry , Endonucleases/metabolism , Enzyme Inhibitors/metabolism , Enzyme Stability , Humans , Influenza A Virus, H1N1 Subtype/genetics , Madin Darby Canine Kidney Cells , Molecular Docking Simulation , Morpholines , Mutation , Oxazines/metabolism , Pyridines/metabolism , Pyridones , Temperature , Thiepins/metabolism , Triazines/metabolism
16.
Antiviral Res ; 139: 41-48, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28012921

ABSTRACT

High morbidity and mortality associated with human cases of highly pathogenic avian influenza (HPAI) viruses, including H5N1 influenza virus, have been reported. The purpose of the present study was to evaluate the antiviral effects of peramivir against HPAI viruses. In neuraminidase (NA) inhibition and virus replication inhibition assays, peramivir showed strong inhibitory activity against H5N1, H7N1 and H7N7 HPAI viruses with sub-nanomolar activity in enzyme assays. In H5N1 viruses containing the NA H275Y mutation, the antiviral activity of peramivir against the variant was lower than that against the wild-type. Evaluation of the in vivo antiviral activity showed that a single intravenous treatment of peramivir (10 mg/kg) prevented lethality in mice infected with wild-type H5N1 virus and also following infection with H5N1 virus with the H275Y mutation after a 5 day administration of peramivir (30 mg/kg). Furthermore, mice injected with peramivir showed low viral titers and low levels of proinflammatory cytokines in the lungs. These results suggest that peramivir has therapeutic activity against HPAI viruses even if the virus harbors the NA H275Y mutation.


Subject(s)
Antiviral Agents/therapeutic use , Cyclopentanes/therapeutic use , Guanidines/therapeutic use , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/genetics , Neuraminidase/genetics , Orthomyxoviridae Infections/drug therapy , Acids, Carbocyclic , Animals , Antiviral Agents/administration & dosage , Cyclopentanes/administration & dosage , Cytokines/immunology , Disease Models, Animal , Guanidines/administration & dosage , Humans , Influenza A Virus, H5N1 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H7N1 Subtype/drug effects , Influenza A Virus, H7N1 Subtype/enzymology , Influenza A Virus, H7N7 Subtype/drug effects , Influenza A Virus, H7N7 Subtype/enzymology , Influenza, Human/drug therapy , Lung/immunology , Lung/virology , Mice , Mutation , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae Infections/virology , Viral Load/drug effects , Virus Replication/drug effects
17.
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
18.
Virol J ; 10: 118, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23587221

ABSTRACT

BACKGROUND: The hemagglutinin (HA) of influenza viruses is a possible target for antiviral drugs because of its key roles in the initiation of infection. Although it was found that a natural compound, Stachyflin, inhibited the growth of H1 and H2 but not H3 influenza viruses in MDCK cells, inhibitory activity of the compound has not been assessed against H4-H16 influenza viruses and the precise mechanism of inhibition has not been clarified. METHODS: Inhibitory activity of Stachyflin against H4-H16 influenza viruses, as well as H1-H3 viruses was examined in MDCK cells. To identify factors responsible for the susceptibility of the viruses to this compound, Stachyflin-resistant viruses were selected in MDCK cells and used for computer docking simulation. RESULTS: It was found that in addition to antiviral activity of Stachyflin against influenza viruses of H1 and H2 subtypes, it inhibited replication of viruses of H5 and H6 subtypes, as well as A(H1N1)pdm09 virus in MDCK cells. Stachyflin also inhibited the virus growth in the lungs of mice infected with A/WSN/1933 (H1N1) and A/chicken/Ibaraki/1/2005 (H5N2). Substitution of amino acid residues was found on the HA2 subunit of Stachyflin-resistant viruses. Docking simulation indicated that D37, K51, T107, and K121 are responsible for construction of the cavity for the binding of the compound. In addition, 3-dimensional structure of the cavity of the HA of Stachyflin-susceptible virus strains was different from that of insusceptible virus strains. CONCLUSION: Antiviral activity of Stachyflin was found against A(H1N1)pdm09, H5, and H6 viruses, and identified a potential binding pocket for Stachyflin on the HA. The present results should provide us with useful information for the development of HA inhibitors with more effective and broader spectrum.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/drug effects , Sesquiterpenes/pharmacology , Animals , Antiviral Agents/metabolism , Antiviral Agents/therapeutic use , Dogs , Drug Resistance, Viral , Female , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A virus/physiology , Lung/virology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Molecular Dynamics Simulation , Mutant Proteins/genetics , Mutation, Missense , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Protein Binding , Protein Conformation , Sesquiterpenes/metabolism , Sesquiterpenes/therapeutic use , Virus Replication/drug effects
19.
Chem Asian J ; 6(4): 1048-56, 2011 Apr 04.
Article in English | MEDLINE | ID: mdl-21305698

ABSTRACT

A potent inhibitor for Vibrio cholerae neuraminidase (VCNA) was developed by using a novel two-step strategy, a target amino acid validation using mechanism-based labeling information, and a potent inhibitor search using a focused library. The labeling information suggested the hidden dynamics of a loop structure of VCNA, which can be a potential target of the novel inhibitor. A focused library composed of 187 compounds was prepared from a 9-azide derivative of 2,3-dehydro-N-acetylneuraminic acid (DANA) to interrupt the function of the loop of the labeled residues. Inhibitor 3 c showed potent inhibition properties and was the strongest inhibitor with FANA, a N-trifluoroacetyl derivative of DANA. Validation studies of the inhibitor with a detergent and a Lineweaver-Burk plot suggested that the 9-substitution group would interact hydrophobically with the target loop moiety, adding a noncompetitive inhibition property to the DANA skeleton. This information enabled us to design compound 4 having the combined structure of 3 c and FANA. Compound 4 showed the most potent inhibition (K(i) =73 nM, mixed inhibition) of VCNA with high selectivity among the tested viral, bacterial, and mammal neuraminidases.


Subject(s)
Enzyme Inhibitors/chemistry , Neuraminidase/antagonists & inhibitors , Catalytic Domain , Drug Design , Enzyme Inhibitors/pharmacology , Neuraminidase/metabolism , Protein Structure, Tertiary , Sialic Acids/chemistry , Structure-Activity Relationship , Vibrio cholerae/enzymology
20.
Antiviral Res ; 81(2): 141-6, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19027039

ABSTRACT

Passage of HIV-1 in the presence of integrase inhibitors (INIs) generates resistant viruses that have mutations in the integrase region. Integrase-resistant mutations Q148K and Q148R were identified as primary mutations with the passage of HIV-1 IIIB in the presence of INIs S-1360 or S/GSK-364735, respectively. Secondary amino acid substitutions E138K or G140S were observed when passage with INI was continued. The role of these mutations was investigated with molecular clones. Relative to Q148K alone, Q148K/E138K had 2- and >6-fold increases in resistance to S-1360 and S/GSK-364735, respectively, and the double mutant had slightly better infectivity and replication kinetics. In contrast, Q148K/G140S and Q148R/E138K had nearly equivalent or slightly reduced fold resistance to the INI compared with their respective Q148 primary mutants, and had increases in infectivity and replication kinetics. Recovery of these surrogates of viral fitness coincided with the recovery of integration efficiency of viral DNA into the host cell chromosome for these double mutants. These data show that recovery of viral integration efficiency can be an important factor for the emergence and maintenance of INI-resistant mutations.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Integrase/metabolism , HIV-1/drug effects , HIV-1/physiology , Mutation, Missense , Virus Replication/drug effects , Furans/pharmacology , HIV Integrase/genetics , HIV-1/genetics , Humans , Jurkat Cells , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL