Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Cell ; 163(3): 746-58, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26496612

ABSTRACT

A key effector route of the Sugar Code involves lectins that exert crucial regulatory controls by targeting distinct cellular glycans. We demonstrate that a single amino-acid substitution in a banana lectin, replacing histidine 84 with a threonine, significantly reduces its mitogenicity, while preserving its broad-spectrum antiviral potency. X-ray crystallography, NMR spectroscopy, and glycocluster assays reveal that loss of mitogenicity is strongly correlated with loss of pi-pi stacking between aromatic amino acids H84 and Y83, which removes a wall separating two carbohydrate binding sites, thus diminishing multivalent interactions. On the other hand, monovalent interactions and antiviral activity are preserved by retaining other wild-type conformational features and possibly through unique contacts involving the T84 side chain. Through such fine-tuning, target selection and downstream effects of a lectin can be modulated so as to knock down one activity, while preserving another, thus providing tools for therapeutics and for understanding the Sugar Code.


Subject(s)
Plant Lectins/chemistry , Plant Lectins/genetics , Anti-HIV Agents/chemistry , Carbohydrate Sequence , Genetic Engineering , Mitogens/chemistry , Models, Molecular , Molecular Dynamics Simulation , Musa/chemistry
2.
J Virol ; 96(15): e0083322, 2022 08 10.
Article in English | MEDLINE | ID: mdl-35852353

ABSTRACT

Human enterovirus D68 (EV-D68) is a globally reemerging respiratory pathogen that is associated with the development of acute flaccid myelitis (AFM) in children. Currently, there are no approved vaccines or treatments for EV-D68 infection, and there is a paucity of data related to the virus and host-specific factors that predict disease severity and progression to the neurologic syndrome. EV-D68 infection of various animal models has served as an important platform for characterization and comparison of disease pathogenesis between historic and contemporary isolates. Still, there are significant gaps in our knowledge of EV-D68 pathogenesis that constrain the development and evaluation of targeted vaccines and antiviral therapies. Continued refinement and characterization of animal models that faithfully reproduce key elements of EV-D68 infection and disease is essential for ensuring public health preparedness for future EV-D68 outbreaks.


Subject(s)
Central Nervous System Viral Diseases , Enterovirus D, Human , Enterovirus Infections , Models, Animal , Myelitis , Animals , Antiviral Agents , Central Nervous System Viral Diseases/complications , Central Nervous System Viral Diseases/virology , Child , Disease Outbreaks , Disease Progression , Enterovirus D, Human/pathogenicity , Enterovirus D, Human/physiology , Enterovirus Infections/complications , Humans , Myelitis/complications , Myelitis/virology , Viral Vaccines
3.
Proc Natl Acad Sci U S A ; 117(4): 2122-2132, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31932446

ABSTRACT

There is a strong need for a new broad-spectrum antiinfluenza therapeutic, as vaccination and existing treatments are only moderately effective. We previously engineered a lectin, H84T banana lectin (H84T), to retain broad-spectrum activity against multiple influenza strains, including pandemic and avian, while largely eliminating the potentially harmful mitogenicity of the parent compound. The amino acid mutation at position 84 from histidine to threonine minimizes the mitogenicity of the wild-type lectin while maintaining antiinfluenza activity in vitro. We now report that in a lethal mouse model H84T is indeed nonmitogenic, and both early and delayed therapeutic administration of H84T intraperitoneally are highly protective, as is H84T administered subcutaneously. Mechanistically, attachment, which we anticipated to be inhibited by H84T, was only somewhat decreased by the lectin. Instead, H84T is internalized into the late endosomal/lysosomal compartment and inhibits virus-endosome fusion. These studies reveal that H84T is efficacious against influenza virus in vivo, and that the loss of mitogenicity seen previously in tissue culture is also seen in vivo, underscoring the potential utility of H84T as a broad-spectrum antiinfluenza agent.


Subject(s)
Antiviral Agents/administration & dosage , Influenza, Human/drug therapy , Lectins/administration & dosage , Lectins/genetics , Musa/genetics , Plant Proteins/administration & dosage , Plant Proteins/genetics , Virus Internalization/drug effects , Animals , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/physiology , Influenza, Human/virology , Male , Mice , Musa/chemistry , Musa/metabolism , Mutation , Protein Engineering
4.
Antimicrob Agents Chemother ; 59(1): 520-6, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25385098

ABSTRACT

The treatment of progressive vaccinia in individuals has involved antiviral drugs, such as cidofovir (CDV), brincidofovir, and/or tecovirimat, combined with vaccinia immune globulin (VIG). VIG is costly, and its supply is limited, so sparing the use of VIG during treatment is an important objective. VIG sparing was modeled in immunosuppressed mice by maximizing the treatment benefits of CDV combined with VIG to determine the effective treatments that delayed the time to death, reduced cutaneous lesion severity, and/or decreased tissue viral titers. SKH-1 hairless mice immunosuppressed with cyclophosphamide and hairless SCID mice (SHO strain) were infected cutaneously with vaccinia virus. Monotherapy, dual combinations (CDV plus VIG), or triple therapy (topical CDV, parenteral CDV, and VIG) were initiated 2 days postinfection and were given every 3 to 4 days through day 11. The efficacy assessment included survival rate, cutaneous lesion severity, and viral titers. Delays in the time to death and the reduction in lesion severity occurred in the following order of efficacy: triple therapy had greater efficacy than double combinations (CDV plus VIG or topical plus parenteral CDV), which had greater efficacy than VIG alone. Parenteral administration of CDV or VIG was necessary to suppress virus titers in internal organs (liver, lung, and spleen). The skin viral titers were significantly reduced by triple therapy only. The greatest efficacy was achieved by triple therapy. In humans, this regimen should translate to a faster cure rate, thus sparing the amount of VIG used for treatment.


Subject(s)
Antiviral Agents/pharmacology , Cytosine/analogs & derivatives , Immunoglobulins/pharmacology , Organophosphonates/pharmacology , Vaccinia/drug therapy , Administration, Topical , Animals , Antiviral Agents/administration & dosage , Cidofovir , Cytosine/administration & dosage , Cytosine/pharmacology , Disease Models, Animal , Drug Therapy, Combination , Female , Immunoglobulins/administration & dosage , Infusions, Parenteral , Mice, Hairless , Mice, SCID , Organophosphonates/administration & dosage , Skin Diseases, Infectious/drug therapy , Skin Diseases, Infectious/virology , Treatment Outcome , Vaccinia/immunology , Vaccinia/virology
5.
J Antimicrob Chemother ; 69(8): 2164-74, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24777908

ABSTRACT

OBJECTIVES: Emerging drug resistance to antiviral therapies is an increasing challenge for the treatment of influenza virus infections. One new antiviral compound, BTA938, a dimeric derivative of the viral neuraminidase inhibitor zanamivir, contains a 14-carbon linker bridging two zanamivir moieties. In these studies, we evaluated antiviral efficacy in cell cultures infected with influenza virus and in mouse models of lethal influenza using H1N1pdm09, H3N2 and oseltamivir-resistant (H275Y) viruses. METHODS: In vitro activity was evaluated against 22 strains of influenza virus. Additionally, in vivo studies compared the efficacy of BTA938 or zanamivir after intranasal treatment. We also tested the hypothesis of a dual mode of action for BTA938 using scanning electron microscopy (SEM). RESULTS: BTA938 inhibited the viruses at nanomolar concentrations in vitro with a median 50% effective concentration value of 0.5 nM. In mouse models, the dimer provided ∼10-fold greater protection than zanamivir. The data also showed that a single low dose (3 mg/kg) protected 100% of mice from an otherwise lethal oseltamivir-resistant (H275Y) influenza virus infection. Remarkably, a single prophylactic treatment (10 mg/kg) administered 7 days before the challenge protected 70% of mice and when administered 1 or 3 days before the challenge it protected 90% of mice. Additionally, SEM provides evidence that the increased antiviral potency may be mediated by an enhanced aggregation of virus on the cell surface. CONCLUSIONS: In vitro and in vivo experiments showed the high antiviral activity of BTA938 for the treatment of influenza virus infections. Moreover, we demonstrated that a single dose of BTA938 is sufficient for prophylactic and therapeutic protection in mouse models.


Subject(s)
Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Orthomyxoviridae Infections/drug therapy , Zanamivir/analogs & derivatives , Zanamivir/pharmacology , Animals , Antiviral Agents/pharmacology , Dogs , Drug Combinations , Drug Resistance, Viral , Enzyme Inhibitors/pharmacology , Female , Influenza A Virus, H1N1 Subtype/classification , Influenza A Virus, H3N2 Subtype/classification , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae Infections/virology , Oseltamivir/pharmacology
6.
Arch Virol ; 159(6): 1279-91, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24311151

ABSTRACT

Few anti-influenza drugs are licensed in the United States for the prevention and therapy of influenza A and B virus infections. This shortage, coupled with continuously emerging drug resistance, as detected through a global surveillance network, seriously limits our anti-influenza armamentarium. Combination therapy appears to offer several advantages over traditional monotherapy in not only delaying development of resistance but also potentially enhancing single antiviral activity. In the present study, we evaluated the antiviral drug susceptibilities of fourteen pandemic influenza A (H1N1) virus isolates in MDCK cells. In addition, we evaluated favipiravir (T-705), an investigational drug with a broad antiviral spectrum and a unique mode of action, alone and in dual combination with the neuraminidase inhibitors (NAIs) oseltamivir, peramivir, or zanamivir, against oseltamivir-sensitive pandemic influenza A/California/07/2009 (H1N1) and oseltamivir-resistant A/Hong Kong/2369/2009 (H1N1) virus. Mean inhibitory values showed that the tested virus isolates remained sensitive to commonly used antiviral drugs, with the exception of the Hong Kong virus isolate. Drug dose-response curves confirmed complete drug resistance to oseltamivir, partial sensitivity to peramivir, and retained susceptibility to zanamivir and favipiravir against the A/Hong Kong/2369/2009 virus. Three-dimensional analysis of drug interactions using the MacSynergy(TM) II program indicated an overall synergistic interaction when favipiravir was combined with the NAIs against the oseltamivir-sensitive influenza virus, and an additive effect against the oseltamivir-resistant virus. Although the clinical relevance of these drug combinations remains to be evaluated, results obtained from this study support the use of combination therapy with favipiravir and NAIs for treatment of human influenza virus infections.


Subject(s)
Amides/pharmacology , Antiviral Agents/pharmacology , Drug Synergism , Enzyme Inhibitors/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Pyrazines/pharmacology , Animals , Cell Line , Dogs , Drug Resistance, Viral , Humans , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza, Human/virology , Microbial Sensitivity Tests , Oseltamivir/pharmacology
7.
Vaccine ; 41(38): 5592-5602, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37532610

ABSTRACT

There is a major unmet need for strategies to improve the immunogenicity and effectiveness of pandemic influenza vaccines, particularly in poor responder populations such as neonates. Recombinant protein approaches to pandemic influenza offer advantages over more traditional inactivated virus approaches, as they are free of problems such as egg adaptation or need for high level biosecurity containment for manufacture. However, a weakness of recombinant proteins is their low immunogenicity. We asked whether the use of an inulin polysaccharide adjuvant (Advax) alone or combined with a TLR9 agonist (CpG55.2) would enhance the immunogenicity and protection of a recombinant hemagglutinin vaccine against H7N9 influenza (rH7HA), including in neonatal mice. Advax adjuvant induced predominantly IgG1 responses against H7HA, whereas Advax-CpG55.2 adjuvant also induced IgG2a, IgG2b and IgG3 responses, consistent with the TLR9 agonist component inducing a Th1 bias. Advax-CpG55.2 adjuvanted rH7HA induced high serum neutralizing antibody titers in adult mice. In newborns it similarly overcame immune hypo-responsiveness and enhanced serum anti-rH7HA IgG levels in 7-day-old BALB/C and C57BL/6 mice. Immunized adult mice were protected against a lethal H7N9 virus challenge. When formulated with Advax-CpG55.2 adjuvant, greater protection was seen with rH7HA than with inactivated H7 whole virus antigen. Advax-CpG55.2 adjuvanted rH7HA represents a promising influenza vaccine platform for further development.


Subject(s)
Influenza A Virus, H7N9 Subtype , Influenza Vaccines , Influenza, Human , Orthomyxoviridae Infections , Animals , Mice , Humans , Animals, Newborn , Hemagglutinins , Toll-Like Receptor 9 , Antibodies, Viral , Mice, Inbred BALB C , Mice, Inbred C57BL , Adjuvants, Immunologic , Vaccines, Synthetic , Recombinant Proteins , Orthomyxoviridae Infections/prevention & control
8.
Virology ; 580: 62-72, 2023 03.
Article in English | MEDLINE | ID: mdl-36780728

ABSTRACT

Enterovirus A71 can cause serious neurological disease in young children. Animal models for EV-A71 are needed to evaluate potential antiviral therapies. Existing models have limitations, including lack of lethality or crucial disease signs. Here we report the development of an EV-A71 model in 28-day-old mice. Virus was serially passaged until it produced consistent lethality and rear-limb paralysis. Onset of disease occurred between days 6-9 post-infection, with mortality following weight loss and neurological signs on days 9-14. In addition, a single administration of human intravenous immunoglobulin at doses of 200, 400 and 800 mg/kg at 4h post-infection was evaluated in the model. Protection from weight loss, neurological signs, and mortality (between 50 and 89%) were observed at doses of 400 mg/kg or greater. Based on these results, IVIG was selected for use as a positive control in this acute model, and suggest that IVIG is a potential therapeutic for EV-A71 infections.


Subject(s)
Enterovirus A, Human , Enterovirus Infections , Enterovirus , Nervous System Diseases , Child , Humans , Mice , Animals , Child, Preschool , Immunoglobulins, Intravenous/therapeutic use , Disease Models, Animal
9.
ACS Med Chem Lett ; 14(4): 506-513, 2023 Apr 13.
Article in English | MEDLINE | ID: mdl-37077387

ABSTRACT

We report for the first time the antiviral activities of two iminovirs (antiviral imino-C-nucleosides) 1 and 2, structurally related to galidesivir (Immucillin A, BCX4430). An iminovir containing the 4-aminopyrrolo[2,1-f][1,2,4-triazine] nucleobase found in remdesivir exhibited submicromolar inhibition of multiple strains of influenza A and B viruses, as well as members of the Bunyavirales order. We also report the first syntheses of ProTide prodrugs of iminovir monophosphates, which unexpectedly displayed poorer viral inhibition than their parent nucleosides in vitro. An efficient synthesis of the 4-aminopyrrolo[2,1-f][1,2,4-triazine]-containing iminovir 2 was developed to enable preliminary in vivo studies, wherein it displayed significant toxicity in BALB/c mice and limited protection against influenza. Further modification of this anti-influenza iminovir will therefore be required to improve its therapeutic value.

10.
Antiviral Res ; 214: 105605, 2023 06.
Article in English | MEDLINE | ID: mdl-37068595

ABSTRACT

This study compared disease progression of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) in three different models of golden hamsters: aged (≈60 weeks old) wild-type (WT), young (6 weeks old) WT, and adult (14-22 weeks old) hamsters expressing the human-angiotensin-converting enzyme 2 (hACE2) receptor. After intranasal (IN) exposure to the SARS-CoV-2 Washington isolate (WA01/2020), 2-deoxy-2-[fluorine-18]fluoro-D-glucose positron emission tomography with computed tomography (18F-FDG PET/CT) was used to monitor disease progression in near real time and animals were euthanized at pre-determined time points to directly compare imaging findings with other disease parameters associated with coronavirus disease 2019 (COVID-19). Consistent with histopathology, 18F-FDG-PET/CT demonstrated that aged WT hamsters exposed to 105 plaque forming units (PFU) developed more severe and protracted pneumonia than young WT hamsters exposed to the same (or lower) dose or hACE2 hamsters exposed to a uniformly lethal dose of virus. Specifically, aged WT hamsters presented with a severe interstitial pneumonia through 8 d post-exposure (PE), while pulmonary regeneration was observed in young WT hamsters at that time. hACE2 hamsters exposed to 100 or 10 PFU virus presented with a minimal to mild hemorrhagic pneumonia but succumbed to SARS-CoV-2-related meningoencephalitis by 6 d PE, suggesting that this model might allow assessment of SARS-CoV-2 infection on the central nervous system (CNS). Our group is the first to use (18F-FDG) PET/CT to differentiate respiratory disease severity ranging from mild to severe in three COVID-19 hamster models. The non-invasive, serial measure of disease progression provided by PET/CT makes it a valuable tool for animal model characterization.


Subject(s)
COVID-19 , Pneumonia , Humans , Animals , Cricetinae , COVID-19/diagnostic imaging , SARS-CoV-2 , Fluorodeoxyglucose F18 , Positron Emission Tomography Computed Tomography/methods , Angiotensin-Converting Enzyme 2 , Positron-Emission Tomography , Mesocricetus , Disease Progression
11.
Antimicrob Agents Chemother ; 56(12): 6328-33, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23027194

ABSTRACT

Compounds lacking oral activity may be delivered intranasally to treat influenza virus infections in mice. However, intranasal treatments greatly enhance the virulence of such virus infections. This can be partially compensated for by giving reduced virus challenge doses. These can be 100- to 1,000-fold lower than infections without such treatment and still cause equivalent mortality. We found that intranasal liquid treatments facilitate virus production (probably through enhanced virus spread) and that lung pneumonia was delayed by only 2 days relative to a 1,000-fold higher virus challenge dose not accompanied by intranasal treatments. In one study, zanamivir was 90 to 100% effective at 10 mg/kg/day by oral, intraperitoneal, and intramuscular routes against influenza A/California/04/2009 (H1N1) virus in mice. However, the same compound administered intranasally at 20 mg/kg/day for 5 days gave no protection from death although the time to death was significantly delayed. A related compound, Neu5Ac2en (N-acetyl-2,3-dehydro-2-deoxyneuraminic acid), was ineffective at 100 mg/kg/day. Intranasal zanamivir and Neu5Ac2en were 70 to 100% protective against influenza A/NWS/33 (H1N1) virus infections at 0.1 to 10 and 30 to 100 mg/kg/day, respectively. Somewhat more difficult to treat was A/Victoria/3/75 virus that required 10 mg/kg/day of zanamivir to achieve full protection. These results illustrate that treatment of influenza virus infections by the intranasal route requires consideration of both virus challenge dose and virus strain in order to avoid compromising the effectiveness of a potentially useful antiviral agent. In addition, the intranasal treatments were shown to facilitate virus replication and promote lung pathology.


Subject(s)
Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Influenza, Human/complications , Administration, Intranasal , Animals , Antiviral Agents/therapeutic use , Dose-Response Relationship, Drug , Female , Humans , Influenza A Virus, H1N1 Subtype , Influenza, Human/drug therapy , Influenza, Human/pathology , Lung/virology , Mice , Mice, Inbred BALB C , N-Acetylneuraminic Acid/analogs & derivatives , N-Acetylneuraminic Acid/therapeutic use , Virus Replication/drug effects , Zanamivir/administration & dosage , Zanamivir/therapeutic use
12.
Vaccine ; 39(38): 5410-5421, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34391593

ABSTRACT

Traditional bolus vaccine administration leads to rapid clearance of vaccine from lymphoid tissue. However, there is increasing evidence suggesting that the kinetics of antigen delivery can impact immune responses to vaccines, particularly when tailored to mimic natural infections. Here, we present the specific enhancements sustained release immunization confers to seasonal influenza vaccine, including the magnitude, durability, and breadth of humoral responses. To achieve sustained vaccine delivery kinetics, we have developed a microneedle array patch (MIMIX), with silk fibroin-formulated vaccine tips designed to embed in the dermis after a short application to the skin and release antigen over 1-2 weeks, mimicking the time course of a natural influenza infection. In a preclinical murine model, a single influenza vaccine administration via MIMIX led to faster seroconversion, response-equivalence to prime-boost bolus immunization, higher HAI titers against drifted influenza strains, and improved protective efficacy upon lethal influenza challenge when compared with intramuscular injection. These results highlight infection mimicry, achieved through sustained release silk microneedles, as a powerful approach to improve existing seasonal influenza vaccines, while also suggesting the broader potential of this platform technology to enable more efficacious next-generation vaccines and vaccine combinations.


Subject(s)
Influenza Vaccines , Influenza, Human , Animals , Humans , Immunogenicity, Vaccine , Influenza, Human/prevention & control , Mice , Needles , Silk
13.
Sci Rep ; 11(1): 20595, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663881

ABSTRACT

The delivery of safe, visible wavelengths of light can be an effective, pathogen-agnostic, countermeasure that would expand the current portfolio of SARS-CoV-2 intervention strategies beyond the conventional approaches of vaccine, antibody, and antiviral therapeutics. Employing custom biological light units, that incorporate optically engineered light-emitting diode (LED) arrays, we harnessed monochromatic wavelengths of light for uniform delivery across biological surfaces. We demonstrated that primary 3D human tracheal/bronchial-derived epithelial tissues tolerated high doses of a narrow spectral band of visible light centered at a peak wavelength of 425 nm. We extended these studies to Vero E6 cells to understand how light may influence the viability of a mammalian cell line conventionally used for assaying SARS-CoV-2. The exposure of single-cell monolayers of Vero E6 cells to similar doses of 425 nm blue light resulted in viabilities that were dependent on dose and cell density. Doses of 425 nm blue light that are well-tolerated by Vero E6 cells also inhibited infection and replication of cell-associated SARS-CoV-2 by > 99% 24 h post-infection after a single five-minute light exposure. Moreover, the 425 nm blue light inactivated cell-free betacoronaviruses including SARS-CoV-1, MERS-CoV, and SARS-CoV-2 up to 99.99% in a dose-dependent manner. Importantly, clinically applicable doses of 425 nm blue light dramatically inhibited SARS-CoV-2 infection and replication in primary human 3D tracheal/bronchial tissue. Safe doses of visible light should be considered part of the strategic portfolio for the development of SARS-CoV-2 therapeutic countermeasures to mitigate coronavirus disease 2019 (COVID-19).


Subject(s)
COVID-19 Drug Treatment , COVID-19/prevention & control , Light , SARS-CoV-2 , Trachea/radiation effects , Virus Replication/radiation effects , Adult , Animals , Antiviral Agents/pharmacology , Bronchi , Calibration , Cell-Free System , Chlorocebus aethiops , Epithelium/pathology , Female , Humans , Respiratory Mucosa/radiation effects , Trachea/virology , Vero Cells
14.
Antimicrob Agents Chemother ; 54(1): 126-33, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19901093

ABSTRACT

Favipiravir (T-705 [6-fluoro-3-hydroxy-2-pyrazinecarboxamide]) and oseltamivir were combined to treat influenza virus A/NWS/33 (H1N1), A/Victoria/3/75 (H3N2), and A/Duck/MN/1525/81 (H5N1) infections. T-705 alone inhibited viruses in cell culture at 1.4 to 4.3 microM. Oseltamivir inhibited these three viruses in cells at 3.7, 0.02, and 0.16 microM and in neuraminidase assays at 0.94, 0.46, and 2.31 nM, respectively. Oral treatments were given twice daily to mice for 5 to 7 days starting, generally, 24 h after infection. Survival resulting from 5 days of oseltamivir treatment (0.1 and 0.3 mg/kg/day) was significantly better in combination with 20 mg/kg of body weight/day of T-705 against the H1N1 infection. Treatment of the H3N2 infection required 50 mg/kg/day of oseltamivir for 7 days to achieve 60% protection; 25 mg/kg/day was ineffective. T-705 was >or=70% protective at 50 to 100 mg/kg/day but inactive at 25 mg/kg/day. The combination of inhibitors (25 mg/kg/day each) increased survival to 90%. The H5N1 infection was not benefited by treatment with oseltamivir (

Subject(s)
Amides/therapeutic use , Antiviral Agents/therapeutic use , Influenza A virus , Orthomyxoviridae Infections/drug therapy , Oseltamivir/therapeutic use , Pyrazines/therapeutic use , Animals , Cell Line , Drug Combinations , Drug Interactions , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/enzymology , Influenza A virus/drug effects , Influenza A virus/enzymology , Mice , Mice, Inbred BALB C , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae Infections/virology
15.
Antiviral Res ; 180: 104753, 2020 08.
Article in English | MEDLINE | ID: mdl-32114033

ABSTRACT

Seasonal influenza causes significant morbidity and mortality around the world each year, even with the use of vaccines and antivirals. There is a need for more effective treatments for severe and hospitalized cases of influenza. In this study, we have tested the efficacy of a human plasma-derived IgG product (FLU-IGIV) against seasonal influenza in mouse and ferret models of influenza infection. FLU-IGIV successfully protected mice (100% survival) against lethal influenza infection. Also, the survival rate observed with FLU-IGIV treatment was better than the survival rate observed with oseltamivir (60% survival). FLU-IGIV significantly reduced the viral load in the lungs compared to placebo (PBS) in ferrets infected with influenza A/California/07/2009 (H1N1pdm09) virus. Overall, these studies demonstrate the efficacy of human plasma-derived FLU-IGIV in relevant animal models of influenza virus infection.


Subject(s)
Antibodies, Viral/therapeutic use , Antiviral Agents/therapeutic use , Immunoglobulins, Intravenous/therapeutic use , Orthomyxoviridae Infections/therapy , Animals , Antiviral Agents/pharmacokinetics , Dose-Response Relationship, Immunologic , Female , Ferrets/virology , Humans , Immunoglobulins, Intravenous/pharmacokinetics , Influenza A Virus, H1N1 Subtype , Mice , Mice, Inbred BALB C , Pandemics , Viral Load/drug effects
16.
Virology ; 526: 146-154, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30390563

ABSTRACT

Enterovirus D68 (EV-D68) is unique among enteroviruses because of the ability to cause severe respiratory disease as well as neurological disease. We developed separate models of respiratory and neurological disease following EV-D68 infection in AG129 mice that respond to antiviral treatment with guanidine. In four-week-old mice infected intranasally, EV-D68 replicates to high titers in lung tissue increasing the proinflammatory cytokines MCP-1 and IL-6. The respiratory infection also produces an acute viremia. In 10-day-old mice infected intraperitoneally, EV-D68 causes a neurological disease with weight-loss, paralysis, and mortality. In our respiratory model, treatment with guanidine provides a two-log reduction in lung virus titers, reduces MCP-1 and IL-6, and prevents histological lesions in the lungs. Importantly, viremia is prevented by early treatment with guanidine. In our neurological model, guanidine treatment protects mice from weight-loss, paralysis, and mortality. These results demonstrate the utility of these models for evaluation of antiviral therapies for EV-D68 infection.


Subject(s)
Antiviral Agents/therapeutic use , Disease Models, Animal , Enterovirus D, Human , Enterovirus Infections/drug therapy , Guanidine/therapeutic use , Nervous System Diseases/drug therapy , Respiratory Tract Infections/drug therapy , Animals , Cytokines/metabolism , Enterovirus Infections/pathology , Enterovirus Infections/physiopathology , Enterovirus Infections/virology , Female , Humans , Lung/metabolism , Lung/pathology , Lung/physiopathology , Lung/virology , Male , Mice , Nervous System Diseases/pathology , Nervous System Diseases/physiopathology , Nervous System Diseases/virology , Respiratory Tract Infections/pathology , Respiratory Tract Infections/physiopathology , Respiratory Tract Infections/virology , Treatment Outcome , Viral Load/drug effects , Viremia/prevention & control
17.
Antiviral Res ; 162: 61-70, 2019 02.
Article in English | MEDLINE | ID: mdl-30521834

ABSTRACT

Enterovirus D68 (EV-D68) is a non-polio enterovirus that affects the respiratory system and can cause serious complications, especially in children and older people with weakened immune systems. As an emerging virus, there are no current antiviral therapies or vaccines available. Our goal was to develop a mouse model of human EV-D68 infection that mimicked the disease observed in humans and could be used for evaluation of experimental therapeutics. This is the first report of a respiratory disease model for EV-D68 infection in mice. We adapted the virus by 30 serial passages in AG129 mice, which are deficient in IFN- α/ß and -γ receptors. Despite a lack of weight loss or mortality in mice, lung function measured by plethysmography, showed an increase in enhanced pause (Penh) on days 6 and 7 post-infection. In addition, as virus adapted to mice, virus titer in the lungs increased 50-fold, and the pro-inflammatory cytokines MCP-1 and RANTES increased 15-fold and 2-fold in the lung, respectively. In addition, a time course of mouse-adapted EV-D68 infection was determined in lung, blood, liver, kidney, spleen, leg muscle, spinal cord and brain. Virus in the lung replicated rapidly after intranasal inoculation of adapted virus, 106 CCID50/mL by 4 h and 108.3 CCID50/mL by 24 h. Virus then spread to the blood and other tissues, including spinal cord and brain. This mouse model for EV-D68 infection includes enhanced pause (Penh) as an indicator of morbidity, and viremia, virus titers and proinflammatory cytokines in the lung, and lung histopathology as indicators of disease. Our mouse-adapted virus has a similar antiviral profile to the original isolate as well as another respiratory picornavirus, rhinovirus-14. This model will be valuable in evaluating experimental therapies in the future.


Subject(s)
Disease Models, Animal , Enterovirus Infections/immunology , Enterovirus Infections/virology , Lung/virology , Respiratory Tract Infections/virology , Animals , Antiviral Agents/therapeutic use , Chemokines/immunology , Cytokines/immunology , Enterovirus D, Human , Enterovirus Infections/drug therapy , Female , Male , Mice , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/immunology , Viral Load , Viremia
18.
Viruses ; 10(1)2018 01 12.
Article in English | MEDLINE | ID: mdl-29329211

ABSTRACT

Enterovirus D68 (EV-D68) caused a large outbreak in the summer and fall of 2014 in the United States. It causes serious respiratory disease, but causation of associated paralysis is controversial, because the virus is not routinely identified in cerebrospinal fluid. To establish clinical correlates with human disease, we evaluated EV-D68 infection in non-lethal paralysis mouse models. Ten-day-old mice lacking interferon responses were injected intraperitoneally with the virus. Paralysis developed in hindlimbs. After six weeks of paralysis, the motor neurons were depleted due to viral infection. Hindlimb muscles were also infected and degenerating. Even at the earliest stage of paralysis, muscles were still infected and were degenerating, in addition to presence of virus in the spinal cord. To model natural respiratory infection, five-day-old mice were infected intranasally with EV-D68. Two of the four infected mice developed forelimb paralysis. The affected limbs had muscle disease, but no spinal cord infection was detected. The unique contributions of this study are that EV-D68 causes paralysis in mice, and that causation by muscle disease, with or without spinal cord disease, may help to resolve the controversy that the virus can cause paralysis, even if it cannot be identified in cerebrospinal fluid.


Subject(s)
Enterovirus D, Human/pathogenicity , Enterovirus Infections/physiopathology , Myelitis/virology , Myositis/virology , Paralysis/etiology , Animals , Enterovirus Infections/virology , Male , Mice , Motor Neurons/virology , Muscular Atrophy/physiopathology , Muscular Atrophy/virology , Myelitis/physiopathology , Myositis/physiopathology , Paralysis/virology , Receptor, Interferon alpha-beta/deficiency , Receptors, Interferon/deficiency , Spinal Cord/virology , Interferon gamma Receptor
19.
Vaccine ; 35(35 Pt B): 4569-4577, 2017 08 16.
Article in English | MEDLINE | ID: mdl-28716554

ABSTRACT

The emergence of pandemic influenza strains, particularly the reemergence of the swine-derived influenza A (H1N1) in 2009, is reaffirmation that influenza viruses are very adaptable and influenza remains as a significant global public health treat. As recommended by the World Health Organization (WHO), the use of adjuvants is an attractive approach to improve vaccine efficacy and allow dose-sparing during an influenza emergency. In this study, we utilized CaPtivate Pharmaceutical's proprietary calcium phosphate nanoparticles (CaPNP) vaccine adjuvant and delivery platform to formulate an inactivated whole virus influenza A/CA/04/2009 (H1N1pdm) vaccine as a potential dose-sparing strategy. We evaluated the relative immunogenicity and the efficacy of the formulation in BALB/c mice following single intramuscularly administration of three different doses (0.3, 1, or 3µg based on HA content) of the vaccine in comparison to non-adjuvanted or alum-adjuvant vaccines. We showed that, addition of CaPNP in vaccine elicited significantly higher hemagglutination inhibition (HAI), virus neutralization (VN), and IgG antibody titers, at all dose levels, relative to the non-adjuvanted vaccine. In addition, the vaccine containing CaPNP provided equal protection with 1/3rd of the antigen dose as compared to the non-adjuvanted or alum-adjuvanted vaccines. Our data provided support to earlier studies indicating that CaPNP is an attractive vaccine adjuvant and delivery system and should play an important role in the development of safe and efficacious dose-sparing vaccines. Our findings also warrant further investigation to validate CaPNP's capacity as an alternative adjuvant to the ones currently licensed for influenza/pandemic influenza vaccination.


Subject(s)
Adjuvants, Immunologic , Calcium Phosphates/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Nanoparticles , Orthomyxoviridae Infections/prevention & control , Aluminum Hydroxide/immunology , Animals , Antibodies, Viral/blood , Dose-Response Relationship, Immunologic , Female , Hemagglutination Inhibition Tests , Humans , Immunogenicity, Vaccine , Immunoglobulin G/blood , Influenza, Human/prevention & control , Mice , Nanoparticles/chemistry , Neutralization Tests , Off-Label Use , Pandemics/prevention & control , Vaccination , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/chemistry
20.
Antiviral Res ; 145: 184-196, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28774800

ABSTRACT

The 30th International Conference on Antiviral Research (ICAR) was held in Atlanta, GA, USA from May 18 to 21, 2017. This report provides an account of award lectures, invited keynote addresses and oral presentations during the meeting. The 2017 Gertrude Elion Memorial Lecture Award by Michael Sofia highlighted one of the most important accomplishments in recent drug discovery in antiviral research, the identification of the hepatitis C virus direct-acting antiviral sofosbuvir and new alternatives to combat hepatitis B virus (HBV) infection. The Antonín Holý Lecture Award by David Chu on medicinal chemistry provided an overview of early developments of nucleoside analogs for the treatment of HIV and varicella zoster virus infection and how this knowledge serves to develop new drugs targeting HBV. Priscilla Yang gave the first ISAR Women in Science lecture. She reported on pharmacological validation of new antiviral targets for dengue, Zika and other flaviviruses. The William Prusoff Young Investigator Lecture Award by Maaike Everts described the Alabama Drug Discovery Alliance and the Antiviral Drug Discovery and Development Consortium, and how they are helping to accelerate the development of new antivirals. The 30th ICAR was a success in promoting new discoveries in antiviral drug development and research. The 31st ICAR will be held in Porto, Portugal, June 11-15, 2018.


Subject(s)
Antiviral Agents , Chemistry, Pharmaceutical , Drug Discovery , Dengue/drug therapy , Hepatitis B/drug therapy , Humans , Zika Virus Infection/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL