Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 112(35): 10890-5, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26283346

RESUMEN

Emerging strains of influenza represent a significant public health threat with potential pandemic consequences. Of particular concern are the recently emerged H7N9 strains which cause pneumonia with acute respiratory distress syndrome. Estimates are that nearly 80% of hospitalized patients with H7N9 have received intensive care unit support. VIS410, a human antibody, targets a unique conserved epitope on influenza A. We evaluated the efficacy of VIS410 for neutralization of group 2 influenza strains, including H3N2 and H7N9 strains in vitro and in vivo. VIS410, administered at 50 mg/kg, protected DBA mice infected with A/Anhui/2013 (H7N9), resulting in significant survival benefit upon single-dose (-24 h) or double-dose (-12 h, +48 h) administration (P < 0.001). A single dose of VIS410 at 50 mg/kg (-12 h) combined with oseltamivir at 50 mg/kg (-12 h, twice daily for 7 d) in C57BL/6 mice infected with A/Shanghai 2/2013 (H7N9) resulted in significant decreased lung viral load (P = 0.002) and decreased lung cytokine responses for nine of the 11 cytokines measured. Based on these results, we find that VIS410 may be effective either as monotherapy or combined with antivirals in treating H7N9 disease, as well as disease from other influenza strains.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Subtipo H7N9 del Virus de la Influenza A/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Anticuerpos ampliamente neutralizantes , Humanos , Gripe Humana/terapia , Ratones , Ratones Endogámicos
2.
J Virol ; 88(3): 1447-60, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24198411

RESUMEN

Influenza viruses are a major public health threat worldwide, and options for antiviral therapy are limited by the emergence of drug-resistant virus strains. The influenza virus glycoprotein hemagglutinin (HA) plays critical roles in the early stage of virus infection, including receptor binding and membrane fusion, making it a potential target for the development of anti-influenza drugs. Using pseudotype virus-based high-throughput screens, we have identified several new small molecules capable of inhibiting influenza virus entry. We prioritized two novel inhibitors, MBX2329 and MBX2546, with aminoalkyl phenol ether and sulfonamide scaffolds, respectively, that specifically inhibit HA-mediated viral entry. The two compounds (i) are potent (50% inhibitory concentration [IC50] of 0.3 to 5.9 µM); (ii) are selective (50% cytotoxicity concentration [CC(50)] of >100 µM), with selectivity index (SI) values of >20 to 200 for different influenza virus strains; (iii) inhibit a wide spectrum of influenza A viruses, which includes the 2009 pandemic influenza virus A/H1N1/2009, highly pathogenic avian influenza (HPAI) virus A/H5N1, and oseltamivir-resistant A/H1N1 strains; (iv) exhibit large volumes of synergy with oseltamivir (36 and 331 µM(2) % at 95% confidence); and (v) have chemically tractable structures. Mechanism-of-action studies suggest that both MBX2329 and MBX2546 bind to HA in a nonoverlapping manner. Additional results from HA-mediated hemolysis of chicken red blood cells (cRBCs), competition assays with monoclonal antibody (MAb) C179, and mutational analysis suggest that the compounds bind in the stem region of the HA trimer and inhibit HA-mediated fusion. Therefore, MBX2329 and MBX2546 represent new starting points for chemical optimization and have the potential to provide valuable future therapeutic options and research tools to study the HA-mediated entry process.


Asunto(s)
Antivirales/farmacología , Hemaglutininas Virales/metabolismo , Virus de la Influenza A/efectos de los fármacos , Gripe Aviar/virología , Gripe Humana/virología , Enfermedades de las Aves de Corral/virología , Bibliotecas de Moléculas Pequeñas/farmacología , Internalización del Virus/efectos de los fármacos , Animales , Antivirales/química , Línea Celular , Pollos , Hemaglutininas Virales/genética , Humanos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/fisiología , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/fisiología , Virus de la Influenza A/genética , Virus de la Influenza A/fisiología , Bibliotecas de Moléculas Pequeñas/química
3.
Arch Virol ; 159(6): 1279-91, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24311151

RESUMEN

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.


Asunto(s)
Amidas/farmacología , Antivirales/farmacología , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Pirazinas/farmacología , Animales , Línea Celular , Perros , Farmacorresistencia Viral , Humanos , Subtipo H1N1 del Virus de la Influenza A/aislamiento & purificación , Gripe Humana/virología , Pruebas de Sensibilidad Microbiana , Oseltamivir/farmacología
4.
Virus Genes ; 47(1): 10-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23686695

RESUMEN

The human parainfluenza virus type 3 (HPIV3) phosphoprotein (P) gene is unusual as it contains an editing site where nontemplated ribonucleotide residues can be inserted. This RNA editing can lead to the expression of the viral P, PD, putative W, and theoretical V protein from a single gene. Although the HPIV3 PD protein has been detected, its function and those of the W and V proteins are poorly understood. Therefore, we first used reverse genetics techniques to construct and rescue a recombinant (r)HPIV3 clone with a polyhistidine sequence at the 5' end of the P gene for tagged protein detection. Western blot analysis demonstrated the presence of the P, PD, and W proteins, but no V protein was detected. Then, we functionally studied the D domain of the PD protein by constructing two rHPIV3 knockout clones that are deficient in the expression of the D domain. Results from growth kinetic studies with infected MA-104 and A596 cells showed that viral replication of the two knockout viruses (rHPIV3-ΔES and rHPIV3-ΔD) was comparable to that of the parental virus in both cell lines. However, viral mRNA transcription and genomic replication was significantly reduced. Furthermore, cytokine/chemokine profiles of A549 cells infected with either knockout virus were unchanged or showed lower levels compared to those from cells infected with the parental virus. These data suggest that the D domain of the PD protein may play a luxury role in HPIV3 RNA synthesis and may also be involved in disrupting the expression of beta interferon.


Asunto(s)
Interferón beta/genética , Virus de la Parainfluenza 3 Humana/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , ARN Viral/genética , Infecciones por Respirovirus/genética , Proteínas Virales/química , Proteínas Virales/metabolismo , Línea Celular , Regulación hacia Abajo , Humanos , Interferón beta/inmunología , Virus de la Parainfluenza 3 Humana/química , Virus de la Parainfluenza 3 Humana/genética , Fosfoproteínas/genética , Estructura Terciaria de Proteína , ARN Viral/metabolismo , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/virología , Eliminación de Secuencia , Proteínas Virales/genética
5.
Heliyon ; 8(7): e09887, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35821966

RESUMEN

Galahad™ is a proanthocyanidin complexed with polysaccharides that inactivates viruses and indicates potential for an innovative approach to making protective vaccines. The polysaccharide portion of Galahad™ consists mainly of arabinan and arabinogalactan. In a seven-day toxicity study in rats, it was not toxic even when tested undiluted. Galahad™ inactivated a wide range of DNA and RNA viruses including adenoviruses, corona viruses such as SARS-CoV-2, and influenza viruses. Electron microscopy studies showed that exposure to Galahad™ caused extensive clumping of virions followed by lack of detection of virions after longer periods of exposure. Based on the viral inactivation data, the hypotheses tested is that Galahad™ inactivation of virus can be used to formulate a protective inactivated virus vaccine. To evaluate this hypothesis, infectious influenza A virus (H5N1, Duck/MN/1525/81) with a titer of 105.7 CCID50/0.1 ml was exposed for 10 min to Galahad™. This treatment caused the infectious virus titer to be reduced to below detectable limits. The Galahad™ -inactivated influenza preparation without adjuvant or preservative was given to BALB/c mice using a variety of routes of administration and dosing regimens. The most protective route of administration and dosing regimen was when mice were given the vaccine twice intranasally, the second dose coming 14 days after the primary vaccine dose. All the mice receiving this vaccine regimen survived the virus challenge while only 20% of the mice receiving placebo survived. This suggests that a Galahad™-inactivated influenza virus vaccine can elicit a protective immune response even without the use of an adjuvant. This technology should be investigated further for its potential to make effective human vaccines.

6.
J Virol ; 84(5): 2511-21, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20032190

RESUMEN

Viruses of the family Coronaviridae have recently emerged through zoonotic transmission to become serious human pathogens. The pathogenic agent responsible for severe acute respiratory syndrome (SARS), the SARS coronavirus (SARS-CoV), is a member of this large family of positive-strand RNA viruses that cause a spectrum of disease in humans, other mammals, and birds. Since the publicized outbreaks of SARS in China and Canada in 2002-2003, significant efforts successfully identified the causative agent, host cell receptor(s), and many of the pathogenic mechanisms underlying SARS. With this greater understanding of SARS-CoV biology, many researchers have sought to identify agents for the treatment of SARS. Here we report the utility of the potent antiviral protein griffithsin (GRFT) in the prevention of SARS-CoV infection both in vitro and in vivo. We also show that GRFT specifically binds to the SARS-CoV spike glycoprotein and inhibits viral entry. In addition, we report the activity of GRFT against a variety of additional coronaviruses that infect humans, other mammals, and birds. Finally, we show that GRFT treatment has a positive effect on morbidity and mortality in a lethal infection model using a mouse-adapted SARS-CoV and also specifically inhibits deleterious aspects of the host immunological response to SARS infection in mammals.


Asunto(s)
Proteínas Algáceas , Antivirales , Infecciones por Coronaviridae/tratamiento farmacológico , Coronaviridae/efectos de los fármacos , Lectinas , Proteínas Algáceas/farmacología , Proteínas Algáceas/uso terapéutico , Secuencia de Aminoácidos , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Calorimetría , Línea Celular , Coronaviridae/genética , Coronaviridae/inmunología , Coronaviridae/patogenicidad , Infecciones por Coronaviridae/inmunología , Infecciones por Coronaviridae/mortalidad , Infecciones por Coronaviridae/prevención & control , Citocinas/inmunología , Femenino , Humanos , Lectinas/farmacología , Lectinas/uso terapéutico , Pulmón/patología , Pulmón/virología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Datos de Secuencia Molecular , Lectinas de Plantas , Unión Proteica , Conformación Proteica , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Glicoproteína de la Espiga del Coronavirus , Proteínas del Envoltorio Viral/metabolismo , Zoonosis
7.
Bioorg Med Chem Lett ; 20(8): 2601-4, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20231094

RESUMEN

Enantiomerically pure cyclopentyl cytosine [(-)-carbodine 1] was synthesized from d-ribose and evaluated for its anti-influenza activity in vitro in comparison to the (+)-carbodine, (+/-)-carbodine and ribavirin. (-)-Carbodine 1 exhibited potent antiviral activity against various strains of influenza A and B viruses.


Asunto(s)
Antivirales/síntesis química , Antivirales/farmacología , Citidina/análogos & derivados , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Animales , Antivirales/química , Línea Celular , Citidina/síntesis química , Citidina/química , Citidina/farmacología , Perros , Pase Seriado , Estereoisomerismo
8.
Sci Rep ; 9(1): 7484, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-31097731

RESUMEN

Development of antiviral drug resistance is a continuous concern for viruses with high mutation rates such as influenza. The use of antiviral drugs targeting host proteins required for viral replication is less likely to result in the selection of resistant viruses than treating with direct-acting antivirals. The iminosugar UV-4B is a host-targeted glucomimetic that inhibits endoplasmic reticulum α-glucosidase I and II enzymes resulting in improper glycosylation and misfolding of viral glycoproteins. UV-4B has broad-spectrum antiviral activity against diverse viruses including dengue and influenza. To examine the ability of influenza virus to develop resistance against UV-4B, mouse-adapted influenza virus was passaged in mice in the presence or absence of UV-4B and virus isolated from lungs was used to infect the next cohort of mice, for five successive passages. Deep sequencing was performed to identify changes in the viral genome during passaging in the presence or absence of UV-4B. Relatively few minor variants were identified within each virus and the ratio of nonsynonymous to synonymous (dN/dS) substitutions of minor variants confirmed no apparent positive selection following sustained exposure to UV-4B. Three substitutions (one synonymous in PB2, one nonsynonymous in M and PA each) were specifically enriched (>3%) in UV-4B-treated groups at passage five. Recombinant viruses containing each individual or combinations of these nonsynonymous mutations remained sensitive to UV-4B treatment in mice. Overall, these data provide evidence that there is a high genetic barrier to the generation and selection of escape mutants following exposure to host-targeted iminosugar antivirals.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Viral/genética , Inhibidores de Glicósido Hidrolasas/farmacología , Virus de la Influenza A/genética , Infecciones por Orthomyxoviridae/virología , Animales , Femenino , Genoma Viral , Virus de la Influenza A/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Mutación , Recombinación Genética , Selección Genética
9.
Biochem Biophys Res Commun ; 371(1): 110-3, 2008 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-18406349

RESUMEN

The primary targets for SARS-CoV infection are the epithelial cells in the respiratory and intestinal tract. The angiotensin-converting enzyme 2 (ACE-2) has been identified as a functional receptor for SARS-CoV. ACE-2 has been shown to be expressed at the apical domain of polarized Calu-3 cells. In this report, interferon alfacon 1 was examined for inhibitory activities against SARS-CoV on human lung carcinoma epithelial Calu-3 cell line and the other three African green monkey kidney epithelial cell lines. Interferon alfacon 1 demonstrated significant antiviral activity in neutral red uptake assay and virus yield reduction assay. The data might provide an important insight into the mechanism of pathogenesis of SARS-CoV allowing further development of antiviral therapies for treating SARS infections.


Asunto(s)
Antivirales/farmacología , Bronquios/virología , Interferón Tipo I/farmacología , Mucosa Respiratoria/virología , Síndrome Respiratorio Agudo Grave/virología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Antivirales/uso terapéutico , Línea Celular , Línea Celular Tumoral , Humanos , Interferón Tipo I/uso terapéutico , Interferón-alfa , Proteínas Recombinantes , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico
10.
Antiviral Res ; 79(2): 105-13, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18423639

RESUMEN

Phenothiazine and derivatives were tested for inhibition of SARS-CoV replication. Phenothiazine slightly inhibited SARS-CoV replication in a neutral red (NR) uptake assay. Adding a propylamino group to give promazine reduced virus yields (VYR assay) with an EC(90)=8.3+/-2.8 microM, but without selectivity. Various substitutions in the basic phenothiazine structure did not promote efficacy. Phenazine ethosulfate was the most potent compound by VYR assay (EC(90)=6.1+/-4.3 microM). All compounds were toxic (IC(50)=6.6-74.5 microM) except for phenoxathiin (IC(50)=858+/-208 microM) and 10-(alpha-diethylamino-propionyl) phenothiazine.HCl (IC(50)=195+/-71.2 microM). Consequently, none were selective inhibitors of SARS-CoV replication (SI values <1-3.3 microM). These data portended the poor efficacy of promazine in a SARS-CoV mouse lung replication model. Intraperitoneal treatment with promazine using a prophylactic (-4h)/therapeutic regimen of 1, 10, or 50mg/(kg day) did not reduce virus lung titers at day 3, yet prolonged virus replication to 14 days. Similar therapeutic promazine doses were not efficacious. Thus, promazine did not affect SARS-CoV replication in vitro or in vivo, nor were any other phenothiazines efficacious in reducing virus replication. Therefore, treating SARS infections with compounds like promazine is not warranted.


Asunto(s)
Antivirales/farmacología , Fenotiazinas/farmacología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Replicación Viral , Animales , Antivirales/administración & dosificación , Antivirales/uso terapéutico , Antivirales/toxicidad , Supervivencia Celular , Quimioprevención , Chlorocebus aethiops , Citocinas/análisis , Femenino , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Estructura Molecular , Rojo Neutro/metabolismo , Fenotiazinas/administración & dosificación , Fenotiazinas/uso terapéutico , Fenotiazinas/toxicidad , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/prevención & control , Células Vero
11.
Antivir Chem Chemother ; 19(1): 15-24, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18610554

RESUMEN

BACKGROUND: The antipoxviral activities and phosphorylation of N-methanocarbathymidine ([N]-MCT) and four 5-halo-2'-deoxyuridines, namely 5-fluoro-(FdU), 5-chloro-(CldU), 5-bromo-(BrdU), and 5-iodo-(IdU) derivatives, were explored. METHODS: Antiviral activities and nucleoside metabolism were determined in C127I mouse, LLC-MK2 monkey, and A549 human cells infected with thymidine-kinase-containing and -deficient (TK+ and TK-) vaccinia (WR strain) viruses. RESULTS: The antiviral potencies of CldU, BrdU and IdU were increased 16-26-fold in LLC-MK2 cells infected with TK+ compared with TK- virus infections, but enhancement of activity was much less in the other cell lines. (N)-MCT was nearly equally active against TK+ and TK- viruses in the three cell lines. Antiviral activity of FdU was associated with cytotoxicity. Uninfected and infected cells metabolized compounds to mono-, di- and triphosphates. The thymidine, BrdU and IdU triphosphate levels were higher in C127I and LLC-MK2 cells infected with TK+ than with TK- virus. (N)-MCT monophosphate levels were much higher in TK+ virus-infected cells, but without corresponding increases in (N)-MCT triphosphate. Furthermore, TK+ virus infections did not appreciably alter (N)-MCT triphosphate levels in other mouse (L929), monkey (MA-104 and Vero) and human cell lines (A549). Antiviral potency of the compounds was greater in C127I than in LLC-MK2 cells, yet lower intracellular triphosphate levels were found in C127I cells. CONCLUSION: We conclude that viral TK plays an important role in increasing the antiviral potencies of these compounds in some cell lines, but minimally in others. These findings may have implications in treating infected animals with compounds that are dependent upon poxvirus TK for their activation, because viral TK activity may vary greatly due to cell type.


Asunto(s)
Antivirales/farmacología , Desoxiuridina/farmacología , Timidina/análogos & derivados , Virus Vaccinia/efectos de los fármacos , Vaccinia/tratamiento farmacológico , Animales , Antivirales/metabolismo , Bromodesoxiuridina/metabolismo , Bromodesoxiuridina/farmacología , Línea Celular Tumoral , Chlorocebus aethiops , Desoxiuridina/análogos & derivados , Desoxiuridina/metabolismo , Floxuridina/metabolismo , Floxuridina/farmacología , Humanos , Idoxuridina/metabolismo , Idoxuridina/farmacología , Ratones , Fosforilación , Timidina/metabolismo , Timidina/farmacología , Timidina Quinasa/metabolismo , Células Vero , Ensayo de Placa Viral
12.
Chemotherapy ; 54(3): 176-80, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18560223

RESUMEN

BACKGROUND: The type 1 interferons (INF-alpha and INF-beta) are potent antiviral agents. Albumin-INF-alpha and albumin-INF-beta are novel recombinant proteins consisting of IFN-alpha or IFN-beta genetically fused to human albumin. METHODS: The in vitro antiviral activity of albumin-IFN-alpha was evaluated against representative bioterrorism viral agents and the severe acute respiratory syndrome virus. Antiviral activity was assessed using inhibition of cytopathic effect and neutral red staining. RESULTS: EC(50) values for albumin-IFN-alpha ranged from <0.1 ng/ml for Punta Toro virus to 65 ng/ml for Venezuelan equine encephalitis virus in the neutral red assay. Albumin-IFN-beta showed 75- and 360-fold greater in vitro activity than albumin-IFN-alpha against Ebola virus and severe acute respiratory syndrome, respectively. CONCLUSION: Further evaluation of these long-acting albumin-IFN fusion proteins as prophylactic or therapeutic agents against these viral agents of bioterrorism in relevant primate models is warranted.


Asunto(s)
Antivirales/farmacología , Bioterrorismo , Interferón Tipo I/farmacología , Interferón-alfa/farmacología , Interferón beta/farmacología , Proteínas Recombinantes de Fusión/farmacología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Animales , Línea Celular , Chlorocebus aethiops , Ebolavirus/efectos de los fármacos , Ebolavirus/fisiología , Proteínas Recombinantes , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Replicación Viral/efectos de los fármacos
13.
Antiviral Res ; 158: 122-126, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30096340

RESUMEN

On September 22, 2008, a physician on Prince of Wales Island, Alaska, notified the Alaska Department of Health and Social Services (ADHSS) of an unusually high number of adult patients with recently diagnosed pneumonia (n = 10), including three persons who required hospitalization and one who died. ADHSS and CDC conducted an investigation to determine the cause and distribution of the outbreak, identify risk factors for hospitalization, and implement control measures. This report summarizes the results of that investigation, which found that the outbreak was caused by adenovirus 14 (Ad14), an emerging adenovirus serotype in the United States that is associated with a higher rate of severe illness compared with other adenoviruses. Among the 46 cases identified in the outbreak from September 1 through October 27, 2008, the most frequently observed characteristics included the following: male (70%), Alaska Native (61%), underlying pulmonary disease (44%), aged > or = 65 years (26%), and current smoker (48%). Patients aged > or = 65 years had a fivefold increased risk for hospitalization. The most commonly reported symptoms were cough (100%), shortness of breath (87%), and fever (74%). Of the 11 hospitalized patients, three required intensive care, and one required mechanical ventilation. One death was reported. Ad14 isolates obtained during the outbreak were identical genetically to those in recent community-acquired outbreaks in the United States which suggests the emergence of a new, and possibly more virulent Ad14 variant. Clinicians should consider Ad14 infection in the differential diagnosis for patients with community-acquired pneumonia, particularly when unexplained clusters of severe respiratory infections are detected.


Asunto(s)
Adenovirus Humanos/efectos de los fármacos , Ésteres/farmacología , Nucleósidos/farmacología , Serogrupo , Células A549 , Infecciones por Adenovirus Humanos/epidemiología , Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/patogenicidad , Anciano , Brotes de Enfermedades , Femenino , Fiebre , Humanos , Masculino , Nucleósidos/análogos & derivados , Análisis de Regresión , Infecciones del Sistema Respiratorio/virología , Estados Unidos
14.
Antivir Chem Chemother ; 18(3): 125-32, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17626596

RESUMEN

Avian influenza H5N1 infections can cause severe, lethal human infections. Whether influenza A virus treatments effectively ameliorate avian influenza H5N1 human infections is uncertain. The research objective was to evaluate the efficacy of novel zinc and other metallo-ion formulations in two influenza A mouse models. Mice infected with influenza A/Duck/MN/1525/81 (H5N1) virus were treated orally 48 h before virus exposure and then twice daily for 13 days with ZnAL42. The optimal dosing regimen for ZnAL42 was achieved at 17.28 mg/kg 48 h prior to virus exposure, twice daily for 7 days. The survival rate was 80% compared with 10% in the untreated control group and a 100% survival rate with ribavirin (75 mg/kg/day, twice a day for 5 days, beginning 4 h before virus exposure). ZnAL42 treatment significantly lessened the decline in arterial oxygen saturation (SaO2; P < 0.001). This regimen was also well tolerated by the mice. Manganese and selenium formulations were not inhibitory to virus replication when given therapeutically. Mice were also infected with influenza A/NWS/33 (H1N1) virus and were treated 48 h before virus exposure with three dosages of ZnAL42 (8.64, 1.46 or 0.24 mg/kg/day). Treatment was by oral gavage twice daily for 13 days. The highest dose of ZnAL42 was significantly inhibitory to the virus infection as seen by prevention of deaths and lessening of decline in SaO2. The data suggest that the prophylactic use of ZnAL42 is effective against avian influenza H5N1 or H1N1 virus infection in mice and should be further explored as an option for treating human influenza virus infections.


Asunto(s)
Compuestos de Aluminio/farmacología , Antivirales/farmacología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/prevención & control , Compuestos de Zinc/farmacología , Administración Oral , Animales , Antivirales/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Humanos , Manganeso/farmacología , Ratones , Ratones Endogámicos BALB C , Oxígeno/sangre , Ribavirina/administración & dosificación , Ribavirina/farmacología , Selenio/farmacología , Organismos Libres de Patógenos Específicos , Análisis de Supervivencia
15.
Antivir Chem Chemother ; 18(5): 301-5, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18046963

RESUMEN

We designed and synthesized novel 2,3-disubstituted quinazolin-4(3H)-ones by microwave technique and characterized them by spectral analysis. Synthesized compounds were screened for cytotoxicity and for antiviral activity against influenza A (H1N1, H3N2 and H5N1), severe acute respiratory syndrome corona, dengue, yellow fever, Venezuelan equine encephalitis (VEE), Rift Valley fever, and Tacaribe viruses in cell culture. A neutral red uptake assay was used to determine 50% virus-inhibitory concentrations (EC50) of test compounds and their 50% cytotoxicity concentration (CC50) in uninfected Madin-Darby canine kidney, Vero, and Vero 76 cells; selectivity indices (ratio of CC50 to EC50) were derived from the data. The compound 4-(6,8-dibromo-4-oxo-2-phenyl quinazolin-3(4H)-yl)-N-(4,5-dimethyloxazol-2yl) benzenesulphonamide 15 inhibited the replication of avian influenza (H5N1) virus (EC50 = 8.4 microg/ml, CC50 > 100 microg/ml, SI > 11.9) as did 4-(6-bromo-4oxo-2phenylquinazolin-3(4H)-yl) benzene]sulphonamide 5 (EC50 = 3 microg/ml, CC50 = 32 microg/ml, SI = 11). Compound 5 was also moderately active against VEE and Tacaribe viruses. The methodology described in this report is applicable for rapid synthesis of many compounds with potential antiviral properties.


Asunto(s)
Antivirales/farmacología , Quinazolinonas/farmacología , Sulfonamidas/farmacología , Virus/efectos de los fármacos , Animales , Antivirales/síntesis química , Antivirales/efectos de la radiación , Chlorocebus aethiops , Perros , Pruebas de Sensibilidad Microbiana , Microondas , Estructura Molecular , Quinazolinonas/síntesis química , Quinazolinonas/efectos de la radiación , Sulfonamidas/síntesis química , Sulfonamidas/efectos de la radiación , Células Vero , Replicación Viral/efectos de los fármacos
16.
Antiviral Res ; 139: 1-12, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27956136

RESUMEN

Hiltonol®, (Poly IC:LC), a potent immunomodulator, is a synthetic, double-stranded polyriboinosinic-polyribocytidylic acid (poly IC) stabilized with Poly-L-lysine and carboxymethyl cellulose (LC). Hiltonol® was tested for efficacy in a lethal SARS-CoV-infected BALB/c mouse model. Hiltonol® at 5, 1, 0.5 or 0.25 mg/kg/day by intranasal (i.n.) route resulted in significant survival benefit when administered at selected times 24 h prior to challenge with a lethal dose of mouse-adapted severe acute respiratory syndrome coronavirus (SARS-CoV). The infected BALB/c mice receiving the Hiltonol® treatments were also significantly effective in protecting mice against weight loss due to infection (p < 0.001). Groups of 20 mice were dosed with Hiltonol® at 2.5 or 0.75 mg/kg by intranasal instillation 7, 14, and 21 days before virus exposure and a second dose was given 24 h later, prophylactic Hiltonol® treatments (2.5 mg/kg/day) were completely protective in preventing death, and in causing significant reduction in lung hemorrhage scores, lung weights and lung virus titers. Hiltonol® was also effective as a therapeutic when give up to 8 h post virus exposure; 100% of the-infected mice were protected against death when Hiltonol® was administered at 5 mg/kg/day 8 h after infection. Our data suggest that Hiltonol® treatment of SARS-CoV infection in mice leads to substantial prophylactic and therapeutic effects and could be used for treatment of other virus disease such as those caused by MERS-CoV a related coronavirus. These properties might be therapeutically advantageous if Hiltonol® is considered for possible clinical use.


Asunto(s)
Carboximetilcelulosa de Sodio/análogos & derivados , Inmunomodulación , Inductores de Interferón/uso terapéutico , Poli I-C/uso terapéutico , Polilisina/análogos & derivados , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/prevención & control , Adyuvantes Inmunológicos , Administración Intranasal , Animales , Carboximetilcelulosa de Sodio/administración & dosificación , Carboximetilcelulosa de Sodio/uso terapéutico , Modelos Animales de Enfermedad , Inductores de Interferón/administración & dosificación , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Poli I-C/administración & dosificación , Polilisina/administración & dosificación , Polilisina/uso terapéutico , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Síndrome Respiratorio Agudo Grave/mortalidad , Síndrome Respiratorio Agudo Grave/virología , Análisis de Supervivencia
17.
ACS Infect Dis ; 3(5): 330-335, 2017 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-28301927

RESUMEN

Influenza A virus envelop protein hemagglutinin (HA) plays important roles in viral entry. We previously have reported that MBX2546, a novel influenza A virus inhibitor, binds to HA and inhibits HA-mediated membrane fusion. In this report, we show that (i) both binding and stabilization of HA by MBX2546 are required for the inhibition of viral infection and (ii) the binding of HA by MBX2546 represses the low-pH-induced conformational change in the HA, which is a prerequisite for membrane fusion. Mutations in MBX2546-resistant influenza A/PR/8/34 (H1N1) viruses are mapped in the HA stem region near the amino terminus of HA2. Finally, we have modeled the binding site of MBX2546 using molecular dynamics and find that the resulting structure is in good agreement with our results. Together, these studies underscore the importance of the HA stem loop region as a potential target for therapeutic intervention.


Asunto(s)
Acetanilidas/química , Antivirales/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Subtipo H1N1 del Virus de la Influenza A/química , Fusión de Membrana/efectos de los fármacos , Sulfonamidas/química , Secuencias de Aminoácidos , Animales , Sitios de Unión , Bioensayo , Perros , Farmacorresistencia Viral/fisiología , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Concentración de Iones de Hidrógeno , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Cinética , Células de Riñón Canino Madin Darby , Simulación de Dinámica Molecular , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
18.
Curr Pharm Des ; 12(11): 1379-90, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16611122

RESUMEN

Picornaviruses are important human pathogens causing severe morbidity and some mortality with the potential to cause worldwide crippling disease. Currently, there are few treatments for many of the viruses in the Picornaviridae, For rhinoviruses, there are no approved treatments, although ruprintrivir looks promising in clinical trials and pyridazinyl oxime ethers may prove useful. Poliovirus treatments are needed to supplement the World Health Organization's polio eradication plan in order to treat infections caused by reversion of the attenuated vaccine virus and to supplement vaccine coverage control in polio endemic areas. However, no promising compounds for treatment of poliovirus have been developed due to the efficacy of the vaccines in use. Broad-spectrum inhibitors developed for other picornavirus may be useful for poliovirus infections. Coxsackievirus infections in children and in infants are being treated with pleconaril with some efficacy in reducing mortality and improving recovery, albeit the treatment is often on a compassionate use basis. There are no therapies for echovirus infections. Very little drug discovery research is being done to develop inhibitors for echovirus infections, probably due to the broad-spectrum inhibition exhibited by capsid binding agents and protease inhibitors discovered for treatment of other picornaviruses. For example, pyridazinyl oxime ethers are inhibitory to most echoviruses. Treatments for enterovirus infections are also limited, although in a small clinical trial, milrinone seemed to reduce mortality and improve recovery from EV71-induced pulmonary edema. Thus, these results strongly emphasize the need for the development of potent and nontoxic compounds for the treatment of picornavirus infections.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Picornaviridae/tratamiento farmacológico , Animales , Infecciones por Coxsackievirus/tratamiento farmacológico , Infecciones por Echovirus/tratamiento farmacológico , Infecciones por Enterovirus/tratamiento farmacológico , Humanos , Picornaviridae/genética , Inhibidores de Proteasas/uso terapéutico , ARN Viral/biosíntesis
19.
Antiviral Res ; 71(2-3): 379-90, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16806515

RESUMEN

Respiratory syncytial virus (RSV) infections remain a significant public health problem throughout the world, although recently developed and clinically approved anti-RSV antibodies administered prophylactically to at-risk populations appear to have significantly affected the disease development. Much effort has been expended to develop effective anti-RSV therapies, using both in vitro assay systems and mouse, cotton rat, and primate models, with several products now in various stages of clinical study. Several products are also being considered for the treatment of clinical symptoms of RSV. In this review, updates on the status of the approved anti-RSV antibodies, ribavirin, and recent results of studies with potential new anti-RSV compounds are summarized and discussed.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Antivirales/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Animales , Antivirales/química , Antivirales/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/inmunología , Proteínas Virales/efectos adversos
20.
Antiviral Res ; 71(1): 53-63, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16621037

RESUMEN

Because of the conflicting data concerning the SARS-CoV inhibitory efficacy of ribavirin, an inosine monophosphate (IMP) dehydrogenase inhibitor, studies were done to evaluate the efficacy of ribavirin and other IMP dehydrogenase inhibitors (5-ethynyl-1-beta-D-ribofuranosylimidazole-4-carboxamide (EICAR), mizoribine, and mycophenolic acid) in preventing viral replication in the lungs of BALB/c mice, a replication model for severe acute respiratory syndrome (SARS) infections (Subbarao, K., McAuliffe, J., Vogel, L., Fahle, G., Fischer, S., Tatti, K., Packard, M., Shieh, W.J., Zaki, S., Murphy, B., 2004. Prior infection and passive transfer of neutralizing antibody prevent replication of severe acute respiratory syndrome coronavirus (SARS-CoV) in the respiratory tract of mice. J. Virol. 78, 3572-3577). Ribavirin given at 75 mg/kg 4 h prior to virus exposure and then given twice daily for 3 days beginning at day 0 was found to increase virus lung titers and extend the length of time that virus could be detected in the lungs of mice. Other IMP dehydrogenase inhibitors administered near maximum tolerated doses using the same dosing regimen as for ribavirin were found to slightly enhance virus replication in the lungs. In addition, ribavirin treatment seemed also to promote the production of pro-inflammatory cytokines 4 days after cessation of treatment, although after 3 days of treatment ribavirin inhibited pro-inflammatory cytokine production in infected mice, significantly reducing the levels of the cytokines IL-1alpha, interleukin-5 (IL-5), monocyte chemotactic protein-1 (MCP-1), and granulocyte-macrophage colony stimulating factor (GM-CSF). These findings suggest that ribavirin may actually contribute to the pathogenesis of SARS-CoV by prolonging and/or enhancing viral replication in the lungs. By not inhibiting viral replication in the lungs of infected mice, ribavirin treatment may have provided a continual source of stimulation for the inflammatory response thought to contribute to the pathogenesis of the infection. Our data do not support the use of ribavirin or other IMP dehydrogenase inhibitors for treating SARS infections in humans.


Asunto(s)
Antivirales/farmacología , IMP Deshidrogenasa/antagonistas & inhibidores , Ribavirina/farmacología , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/virología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Replicación Viral/efectos de los fármacos , Animales , Células CACO-2 , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Citocinas/metabolismo , Efecto Citopatogénico Viral/efectos de los fármacos , Femenino , Humanos , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Ácido Micofenólico/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Ribonucleósidos/farmacología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/enzimología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Organismos Libres de Patógenos Específicos , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA