Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Emerg Infect Dis ; 30(7): 1410-1415, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38916572

RESUMEN

Since May 2023, a novel combination of neuraminidase mutations, I223V + S247N, has been detected in influenza A(H1N1)pdm09 viruses collected in countries spanning 5 continents, mostly in Europe (67/101). The viruses belong to 2 phylogenetically distinct groups and display ≈13-fold reduced inhibition by oseltamivir while retaining normal susceptibility to other antiviral drugs.


Asunto(s)
Antivirales , Farmacorresistencia Viral , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana , Neuraminidasa , Oseltamivir , Filogenia , Oseltamivir/farmacología , Oseltamivir/uso terapéutico , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/genética , Humanos , Antivirales/farmacología , Antivirales/uso terapéutico , Gripe Humana/virología , Gripe Humana/tratamiento farmacológico , Gripe Humana/epidemiología , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/genética , Farmacorresistencia Viral/genética , Mutación
2.
Antimicrob Agents Chemother ; 68(5): e0172723, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38587392

RESUMEN

Antiviral susceptibility of influenza viruses was assessed using a high-content imaging-based neutralization test. Cap-dependent endonuclease inhibitors, baloxavir and AV5116, were superior to AV5115 against type A viruses, and AV5116 was most effective against PA mutants tested. However, these three inhibitors displayed comparable activity (EC50 8-22 nM) against type C viruses from six lineages. Banana lectin and a monoclonal antibody, YA3, targeting the hemagglutinin-esterase protein effectively neutralized some, but not all, type C viruses.


Asunto(s)
Antivirales , Dibenzotiepinas , Triazinas , Antivirales/farmacología , Humanos , Triazinas/farmacología , Dibenzotiepinas/farmacología , Gammainfluenzavirus/efectos de los fármacos , Gammainfluenzavirus/genética , Morfolinas/farmacología , Piridonas/farmacología , Animales , Anticuerpos Monoclonales/farmacología , Células de Riñón Canino Madin Darby , Perros , Ciclopropanos/farmacología , Virus de la Influenza A/efectos de los fármacos , Pruebas de Neutralización , Piridinas/farmacología
3.
Emerg Infect Dis ; 27(7): 1953-1957, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34152954

RESUMEN

Four cases of oseltamivir-resistant influenza A(H1N1)pdm09 virus infection were detected among inhabitants of a border detention center in Texas, USA. Hemagglutinin of these viruses belongs to 6B.1A5A-156K subclade, which may enable viral escape from preexisting immunity. Our finding highlights the necessity to monitor both drug resistance and antigenic drift of circulating viruses.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Gripe Humana , Antivirales/uso terapéutico , Farmacorresistencia Viral , Hemaglutininas , Humanos , Gripe Humana/tratamiento farmacológico , Neuraminidasa , Oseltamivir/uso terapéutico , Texas , Proteínas Virales
4.
J Infect Dis ; 221(3): 367-371, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31541547

RESUMEN

Susceptibility of influenza A viruses to baloxavir can be affected by changes at amino acid residue 38 in the polymerase acidic (PA) protein. Information on replicative fitness of PA-I38-substituted viruses remains sparse. We demonstrated that substitutions I38L/M/S/T not only had a differential effect on baloxavir susceptibility (9- to 116-fold) but also on in vitro replicative fitness. Although I38L conferred undiminished growth, other substitutions led to mild attenuation. In a ferret model, control viruses outcompeted those carrying I38M or I38T substitutions, although their advantage was limited. These findings offer insights into the attributes of baloxavir-resistant viruses needed for informed risk assessment.


Asunto(s)
Antivirales/uso terapéutico , Farmacorresistencia Viral/genética , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/genética , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Oxazinas/uso terapéutico , Piridinas/uso terapéutico , Tiepinas/uso terapéutico , Triazinas/uso terapéutico , Replicación Viral/genética , Sustitución de Aminoácidos , Animales , Dibenzotiepinas , Modelos Animales de Enfermedad , Perros , Hurones , Secuenciación de Nucleótidos de Alto Rendimiento , Células de Riñón Canino Madin Darby , Masculino , Pruebas de Sensibilidad Microbiana , Morfolinas , Infecciones por Orthomyxoviridae/virología , Piridonas , ARN Polimerasa Dependiente del ARN/genética , Estaciones del Año , Resultado del Tratamiento , Proteínas Virales/genética
5.
Emerg Infect Dis ; 25(10): 1969-1972, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31287050
6.
Euro Surveill ; 24(3)2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30670144

RESUMEN

The anti-influenza therapeutic baloxavir targets cap-dependent endonuclease activity of polymerase acidic (PA) protein. We monitored baloxavir susceptibility in the United States with next generation sequencing analysis supplemented by phenotypic one-cycle infection assay. Analysis of PA sequences of 6,891 influenza A and B viruses collected during 2016/17 and 2017/18 seasons showed amino acid substitutions: I38L (two A(H1N1)pdm09 viruses), E23G (two A(H1N1)pdm09 viruses) and I38M (one A(H3N2) virus); conferring 4-10-fold reduced susceptibility to baloxavir.


Asunto(s)
Sustitución de Aminoácidos/efectos de los fármacos , Antivirales/farmacología , Farmacorresistencia Viral/genética , Inhibidores Enzimáticos/farmacología , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza B/efectos de los fármacos , Gripe Humana/tratamiento farmacológico , Oxazinas/farmacología , Piridinas/farmacología , Tiepinas/farmacología , Triazinas/farmacología , Sustitución de Aminoácidos/genética , Antivirales/uso terapéutico , Dibenzotiepinas , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza B/genética , Virus de la Influenza B/aislamiento & purificación , Gripe Humana/epidemiología , Gripe Humana/virología , Pruebas de Sensibilidad Microbiana , Morfolinas , Piridonas , Estaciones del Año , Vigilancia de Guardia , Estados Unidos , Proteínas Virales/genética
7.
J Infect Dis ; 216(suppl_4): S566-S574, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28934455

RESUMEN

Background: Neuraminidase (NA) inhibitors are the recommended antiviral medications for influenza treatment. However, their therapeutic efficacy can be compromised by NA changes that emerge naturally and/or following antiviral treatment. Knowledge of which molecular changes confer drug resistance of influenza A(H7N9) viruses (group 2NA) remains sparse. Methods: Fourteen amino acid substitutions were introduced into the NA of A/Shanghai/2/2013(H7N9). Recombinant N9 (recN9) proteins were expressed in a baculovirus system in insect cells and tested using the Centers for Disease Control and Prevention standardized NA inhibition (NI) assay with oseltamivir, zanamivir, peramivir, and laninamivir. The wild-type N9 crystal structure was determined in complex with oseltamivir, zanamivir, or sialic acid, and structural analysis was performed. Results: All substitutions conferred either reduced or highly reduced inhibition by at least 1 NA inhibitor; half of them caused reduced inhibition or highly reduced inhibition by all NA inhibitors. R292K conferred the highest increase in oseltamivir half-maximal inhibitory concentration (IC50), and E119D conferred the highest zanamivir IC50. Unlike N2 (another group 2NA), H274Y conferred highly reduced inhibition by oseltamivir. Additionally, R152K, a naturally occurring variation at the NA catalytic residue of A(H7N9) viruses, conferred reduced inhibition by laninamivir. Conclusions: The recNA method is a valuable tool for assessing the effect of NA changes on drug susceptibility of emerging influenza viruses.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Viral Múltiple/genética , Subtipo H7N9 del Virus de la Influenza A/efectos de los fármacos , Neuraminidasa/antagonistas & inhibidores , Proteínas Virales/antagonistas & inhibidores , Ácidos Carbocíclicos , Ciclopentanos/farmacología , Bases de Datos Genéticas , Inhibidores Enzimáticos/farmacología , Guanidinas/farmacología , Humanos , Subtipo H7N9 del Virus de la Influenza A/genética , Gripe Humana/tratamiento farmacológico , Concentración 50 Inhibidora , Neuraminidasa/genética , Oseltamivir/farmacología , Conformación Proteica , Piranos , Proteínas Recombinantes/genética , Ácidos Siálicos , Proteínas Virales/genética , Zanamivir/análogos & derivados , Zanamivir/farmacología
8.
Clin Infect Dis ; 65(10): 1644-1651, 2017 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-29020179

RESUMEN

BACKGROUND: Antigenically drifted A(H3N2) viruses circulated extensively during the 2014-2015 influenza season. Vaccine effectiveness (VE) was low and not significant among outpatients but in a hospitalized population was 43%. At least one study paradoxically observed increased A(H3N2) infection among those vaccinated 3 consecutive years. METHODS: We followed a cohort of 1341 individuals from 340 households. VE against laboratory-confirmed influenza was estimated. Hemagglutination-inhibition and neuraminidase-inhibition antibody titers were determined in subjects ≥13 years. RESULTS: Influenza A(H3N2) was identified in 166 (12%) individuals and B(Yamagata) in 34 (2%). VE against A(H3N2) was -3% (95% confidence interval [CI]: -55%, 32%) and similarly ineffective between age groups; increased risk of infection was not observed among those vaccinated in 2 or 3 previous years. VE against influenza B(Yamagata) was 57% (95% CI: -3%, 82%) but only significantly protective in children <9 years (87% [95% CI: 43%, 97%]). Less than 20% of older children and adults had ≥4-fold antibody titer rise against influenza A(H3N2) and B antigens following vaccination; responses were surprisingly similar for antigens included in the vaccine and those similar to circulating viruses. Antibody against A/Hong Kong/4801/14, similar to circulating 2014-2015 A(H3N2) viruses and included in the 2016-2017 vaccine, did not significantly predict protection. CONCLUSIONS: Absence of VE against A(H3N2) was consistent with circulation of antigenically drifted viruses; however, generally limited antibody response following vaccination is concerning even in the context of antigenic mismatch. Although 2014-2015 vaccines were not effective in preventing A(H3N2) infection, no increased susceptibility was detected among the repeatedly vaccinated.


Asunto(s)
Anticuerpos Antivirales/sangre , Subtipo H3N2 del Virus de la Influenza A/inmunología , Virus de la Influenza B/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Adolescente , Adulto , Niño , Estudios de Cohortes , Femenino , Humanos , Gripe Humana/epidemiología , Masculino , Persona de Mediana Edad , Adulto Joven
9.
Emerg Infect Dis ; 23(4): 686-690, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28322707

RESUMEN

In February 2016, three influenza B/Victoria/2/87 lineage viruses exhibiting 4- to 158-fold reduced inhibition by neuraminidase inhibitors were detected in Laos. These viruses had an H134N substitution in the neuraminidase and replicated efficiently in vitro and in ferrets. Current antiviral drugs may be ineffective in controlling infections caused by viruses harboring this mutation.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Viral/genética , Virus de la Influenza B/efectos de los fármacos , Gripe Humana/epidemiología , Gripe Humana/virología , Neuraminidasa/genética , Adolescente , Adulto , Anciano , Sustitución de Aminoácidos , Niño , Preescolar , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Viral de la Expresión Génica , Humanos , Lactante , Virus de la Influenza B/genética , Laos/epidemiología , Masculino , Persona de Mediana Edad , Adulto Joven
10.
Anal Chem ; 89(5): 3130-3137, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28192976

RESUMEN

Mounting evidence suggests that neuraminidase's functionality extends beyond its classical role in influenza virus infection and that antineuraminidase antibodies offer protective immunity. Therefore, a renewed interest in the development of neuraminidase (NA)-specific methods to characterize the glycoprotein and evaluate potential advantages for NA standardization in influenza vaccines has emerged. NA displays sialidase activity by cleaving off the terminal N-acetylneuraminic acid on α-2,3 or α-2,6 sialic acid containing receptors of host cells. The type and distribution of these sialic acid containing receptors is considered to be an important factor in transmission efficiency of influenza viruses between and among host species. Changes in hemagglutinin (HA) binding and NA specificity in reassortant viruses may be related to the emergence of new and potentially dangerous strains of influenza. Current methods to investigate neuraminidase activity use small derivatized sugars that are poor models for natural glycoprotein receptors and do not provide information on the linkage specificity. Here, a novel approach for rapid and accurate quantification of influenza neuraminidase activity is achieved utilizing ultra-high performance liquid chromatography (UPLC) and isotope dilution mass spectrometry (IDMS). Direct LC-MS/MS quantification of NA-released sialic acid provides precise measurement of influenza neuraminidase activity over a range of substrates. The method provides exceptional sensitivity and specificity with a limit of detection of 0.38 µM for sialic acid and the capacity to obtain accurate measurements of specific enzyme activity preference toward α-2,3-sialyllactose linkages, α-2,6-sialyllactose linkages, or whole glycosylated proteins such as fetuin.


Asunto(s)
Cromatografía Líquida de Alta Presión , Subtipo H1N1 del Virus de la Influenza A/enzimología , Neuraminidasa/metabolismo , Espectrometría de Masas en Tándem , Proteínas Virales/metabolismo , Isótopos de Carbono/química , Humanos , Vacunas contra la Influenza/análisis , Vacunas contra la Influenza/metabolismo , Cinética , Lactosa/análogos & derivados , Lactosa/análisis , Especificidad por Sustrato
11.
J Clin Microbiol ; 55(1): 145-154, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27795346

RESUMEN

The rapid evolution of influenza A(H3N2) viruses necessitates close monitoring of their antigenic properties so the emergence and spread of antigenic drift variants can be rapidly identified. Changes in hemagglutinin (HA) acquired by contemporary A(H3N2) viruses hinder antigenic characterization by traditional methods, thus complicating vaccine strain selection. Sequence-based approaches have been used to infer virus antigenicity; however, they are time consuming and mid-throughput. To facilitate virological surveillance and epidemiological studies, we developed and validated a pyrosequencing approach that enables identification of six HA clades of contemporary A(H3N2) viruses. The identification scheme of viruses of the H3 clades 3C.2, 3C.2a, 3C.2b, 3C.3, 3C.3a, and 3C.3b is based on the interrogation of five single nucleotide polymorphisms (SNPs) within three neighboring HA regions, namely 412 to 431, 465 to 481, and 559 to 571. Two bioinformatics tools, IdentiFire (Qiagen) and FireComb (developed in-house), were utilized to expedite pyrosequencing data analysis. The assay's analytical sensitivity was 10 focus forming units, and respiratory specimens with threshold cycle (CT) values of <34 typically produced good quality pyrograms. When applied to 120 A(H3N2) virus isolates and 27 respiratory specimens, the assay displayed 100% agreement with clades determined by HA sequencing coupled with phylogenetics. The multi-SNP analysis described here was readily adopted by another laboratory with pyrosequencing capabilities. The implementation of this approach enhanced the findings from virological surveillance and epidemiological studies between 2013 and 2016, which examined more than 3,000 A(H3N2) viruses.


Asunto(s)
Flujo Genético , Técnicas de Genotipaje/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Subtipo H3N2 del Virus de la Influenza A/clasificación , Subtipo H3N2 del Virus de la Influenza A/genética , Gripe Humana/virología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Subtipo H3N2 del Virus de la Influenza A/aislamiento & purificación , Polimorfismo de Nucleótido Simple , Sensibilidad y Especificidad
12.
J Virol ; 90(12): 5770-5784, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27053557

RESUMEN

UNLABELLED: During 2014, a subclade 2.3.4.4 highly pathogenic avian influenza (HPAI) A(H5N8) virus caused poultry outbreaks around the world. In late 2014/early 2015, the virus was detected in wild birds in Canada and the United States, and these viruses also gave rise to reassortant progeny, composed of viral RNA segments (vRNAs) from both Eurasian and North American lineages. In particular, viruses were found with N1, N2, and N8 neuraminidase vRNAs, and these are collectively referred to as H5Nx viruses. In the United States, more than 48 million domestic birds have been affected. Here we present a detailed structural and biochemical analysis of the surface antigens of H5N1, H5N2, and H5N8 viruses in addition to those of a recent human H5N6 virus. Our results with recombinant hemagglutinin reveal that these viruses have a strict avian receptor binding preference, while recombinantly expressed neuraminidases are sensitive to FDA-approved and investigational antivirals. Although H5Nx viruses currently pose a low risk to humans, it is important to maintain surveillance of these circulating viruses and to continually assess future changes that may increase their pandemic potential. IMPORTANCE: The H5Nx viruses emerging in North America, Europe, and Asia pose a great public health concern. Here we report a molecular and structural study of the major surface proteins of several H5Nx influenza viruses. Our results improve the understanding of these new viruses and provide important information on their receptor preferences and susceptibilities to antivirals, which are central to pandemic risk assessment.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H5N1 del Virus de la Influenza A/química , Subtipo H5N2 del Virus de la Influenza A/química , Subtipo H5N8 del Virus de la Influenza A/química , Neuraminidasa/química , Neuraminidasa/metabolismo , Animales , Animales Salvajes/virología , Asia/epidemiología , Canadá/epidemiología , Brotes de Enfermedades , Europa (Continente)/epidemiología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Subtipo H5N1 del Virus de la Influenza A/enzimología , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N2 del Virus de la Influenza A/enzimología , Subtipo H5N2 del Virus de la Influenza A/genética , Subtipo H5N8 del Virus de la Influenza A/enzimología , Subtipo H5N8 del Virus de la Influenza A/genética , Gripe Aviar/epidemiología , Gripe Aviar/virología , Gripe Humana/virología , Neuraminidasa/genética , América del Norte/epidemiología , Filogenia , Aves de Corral , Virus Reordenados
13.
J Virol ; 90(23): 10446-10458, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27630240

RESUMEN

The pandemic threat posed by emerging zoonotic influenza A viruses necessitates development of antiviral agents effective against various antigenic subtypes. Human monoclonal antibody (hMAb) targeting the hemagglutinin (HA) stalk offers a promising approach to control influenza virus infections. Here, we investigated the ability of the hMAb 81.39a to inhibit in vitro replication of human and zoonotic viruses, representing 16 HA subtypes. The majority of viruses were effectively neutralized by 81.39a at a 50% effective concentration (EC50) of <0.01 to 4.9 µg/ml. Among group 2 HA viruses tested, a single A(H7N9) virus was not neutralized at 50 µg/ml; it contained HA2-Asp19Gly, an amino acid position previously associated with resistance to neutralization by the group 2 HA-neutralizing MAb CR8020. Notably, among group 1 HA viruses, H11-H13 and H16 subtypes were not neutralized at 50 µg/ml; they shared the substitution HA2-Asp19Asn/Ala. Conversely, H9 viruses harboring HA2-Asp19Ala were fully susceptible to neutralization. Therefore, amino acid variance at HA2-Asp19 has subtype-specific adverse effects on in vitro neutralization. Mice given a single injection (15 or 45 mg/kg of body weight) at 24 or 48 h after infection with recently emerged A(H5N2), A(H5N8), A(H6N1), or A(H7N9) viruses were protected from mortality and showed drastically reduced lung viral titers. Furthermore, 81.39a protected mice infected with A(H7N9) harboring HA2-Asp19Gly, although the antiviral effect was lessened. A(H1N1)pdm09-infected ferrets receiving a single dose (25 mg/kg) had reduced viral titers and showed less lung tissue injury, despite 24- to 72-h-delayed treatment. Taken together, this study provides experimental evidence for the therapeutic potential of 81.39a against diverse influenza A viruses. IMPORTANCE: Zoonotic influenza viruses, such as A(H5N1) and A(H7N9) subtypes, have caused severe disease and deaths in humans, raising public health concerns. Development of novel anti-influenza therapeutics with a broad spectrum of activity against various subtypes is necessary to mitigate disease severity. Here, we demonstrate that the hemagglutinin (HA) stalk-targeting human monoclonal antibody 81.39a effectively neutralized the majority of influenza A viruses tested, representing 16 HA subtypes. Furthermore, delayed treatment with 81.39a significantly suppressed virus replication in the lungs, prevented dramatic body weight loss, and increased survival rates of mice infected with A(H5Nx), A(H6N1), or A(H7N9) viruses. When tested in ferrets, delayed 81.39a treatment reduced viral titers, particularly in the lower respiratory tract, and substantially alleviated disease symptoms associated with severe A(H1N1)pdm09 influenza. Collectively, our data demonstrated the effectiveness of 81.39a against both seasonal and emerging influenza A viruses.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Virus de la Influenza A/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Variación Antigénica/genética , Variación Antigénica/inmunología , Femenino , Hurones , Glicoproteínas Hemaglutininas del Virus de la Influenza/clasificación , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Técnicas In Vitro , Virus de la Influenza A/clasificación , Virus de la Influenza A/genética , Gripe Humana/inmunología , Gripe Humana/virología , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/terapia , Infecciones por Orthomyxoviridae/virología , Resultado del Tratamiento
14.
Euro Surveill ; 22(18)2017 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-28494845

RESUMEN

A new rapid assay for detecting oseltamivir resistance in influenza virus, iART, was used to test 149 clinical specimens. Results were obtained for 132, with iART indicating 41 as 'resistant'. For these, sequence analysis found known and suspected markers of oseltamivir resistance, while no such markers were detected for the remaining 91 samples. Viruses isolated from the 41 specimens showed reduced or highly reduced inhibition by neuraminidase inhibition assay. iART may facilitate broader antiviral resistance testing.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Viral , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Neuraminidasa/antagonistas & inhibidores , Oseltamivir/farmacología , Antivirales/uso terapéutico , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/aislamiento & purificación , Gripe Humana , Pruebas de Sensibilidad Microbiana/métodos , Neuraminidasa/genética , Neuraminidasa/metabolismo , Neuraminidasa/uso terapéutico , Oseltamivir/uso terapéutico
15.
J Infect Dis ; 214(7): 1010-9, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27190176

RESUMEN

BACKGROUND: During the 2014-2015 US influenza season, expanded genetic characterization of circulating influenza A(H3N2) viruses was used to assess the impact of the genetic variability of influenza A(H3N2) viruses on influenza vaccine effectiveness (VE). METHODS: A novel pyrosequencing assay was used to determine genetic group, based on hemagglutinin (HA) gene sequences, of influenza A(H3N2) viruses from patients enrolled at US Influenza Vaccine Effectiveness Network sites. VE was estimated using a test-negative design comparing vaccination among patients infected with influenza A(H3N2) viruses and uninfected patients. RESULTS: Among 9710 enrollees, 1868 (19%) tested positive for influenza A(H3N2) virus; genetic characterization of 1397 viruses showed that 1134 (81%) belonged to 1 HA genetic group (3C.2a) of antigenically drifted influenza A(H3N2) viruses. Effectiveness of 2014-2015 influenza vaccination varied by influenza A(H3N2) virus genetic group from 1% (95% confidence interval [CI], -14% to 14%) against illness caused by antigenically drifted influenza A(H3N2) virus group 3C.2a viruses versus 44% (95% CI, 16%-63%) against illness caused by vaccine-like influenza A(H3N2) virus group 3C.3b viruses. CONCLUSIONS: Effectiveness of 2014-2015 influenza vaccination varied by genetic group of influenza A(H3N2) virus. Changes in HA genes related to antigenic drift were associated with reduced VE.


Asunto(s)
Variación Genética , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Gripe Humana/virología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Preescolar , Femenino , Flujo Genético , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Subtipo H3N2 del Virus de la Influenza A/clasificación , Vacunas contra la Influenza/administración & dosificación , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Estados Unidos , Adulto Joven
16.
J Virol ; 89(5): 2801-12, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25540377

RESUMEN

UNLABELLED: In late 2011, an A(H3N8) influenza virus infection resulted in the deaths of 162 New England harbor seals. Virus sequence analysis and virus receptor binding studies highlighted potential markers responsible for mammalian adaptation and a mixed receptor binding preference (S. J. Anthony, J. A. St Leger, K. Pugliares, H. S. Ip, J. M. Chan, Z. W. Carpenter, I. Navarrete-Macias, M. Sanchez-Leon, J. T. Saliki, J. Pedersen, W. Karesh, P. Daszak, R. Rabadan, T. Rowles, W. I. Lipkin, MBio 3:e00166-00112, 2012, http://dx.doi.org/10.1128/mBio.00166-12). Here, we present a detailed structural and biochemical analysis of the surface antigens of the virus. Results obtained with recombinant proteins for both the hemagglutinin and neuraminidase indicate a true avian receptor binding preference. Although the detection of this virus in new species highlights an increased potential for cross-species transmission, our results indicate that the A(H3N8) virus currently poses a low risk to humans. IMPORTANCE: Cross-species transmission of zoonotic influenza viruses increases public health concerns. Here, we report a molecular and structural study of the major surface proteins from an A(H3N8) influenza virus isolated from New England harbor seals. The results improve our understanding of these viruses as they evolve and provide important information to aid ongoing risk assessment analyses as these zoonotic influenza viruses continue to circulate and adapt to new hosts.


Asunto(s)
Antígenos Virales/metabolismo , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H3N8 del Virus de la Influenza A/fisiología , Neuraminidasa/metabolismo , Infecciones por Orthomyxoviridae/veterinaria , Phoca/virología , Proteínas Virales/metabolismo , Acoplamiento Viral , Secuencia de Aminoácidos , Animales , Antígenos Virales/química , Cristalografía por Rayos X , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Subtipo H3N8 del Virus de la Influenza A/química , Subtipo H3N8 del Virus de la Influenza A/aislamiento & purificación , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Datos de Secuencia Molecular , Neuraminidasa/química , New England , Infecciones por Orthomyxoviridae/virología , Polisacáridos/análisis , Unión Proteica , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Proteínas Virales/química
17.
J Virol ; 89(10): 5419-26, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25740997

RESUMEN

UNLABELLED: Human infections by avian influenza A(H7N9) virus entail substantial morbidity and mortality. Treatment of infected patients with the neuraminidase (NA) inhibitor oseltamivir was associated with emergence of viruses carrying NA substitutions. In the NA inhibition (NI) assay, R292K conferred highly reduced inhibition by oseltamivir, while E119V and I222K each caused reduced inhibition. To facilitate establishment of laboratory correlates of clinically relevant resistance, experiments were conducted in ferrets infected with virus carrying wild-type or variant NA genes recovered from the A/Taiwan/1/2013 isolate. Oseltamivir treatment (5 or 25 mg/kg of body weight/dose) was given 4 h postinfection, followed by twice-daily treatment for 5 days. Treatment of ferrets infected with wild-type virus resulted in a modest dose-dependent reduction (0.7 to 1.5 log10 50% tissue culture infectious dose [TCID50]) in nasal wash viral titers and inflammation response. Conversely, treatment failed to significantly inhibit the replication of R292K or E119V virus. A small reduction of viral titers was detected on day 5 in ferrets infected with the I222K virus. The propensity for oseltamivir resistance emergence was assessed in oseltamivir-treated animals infected with wild-type virus; emergence of R292K virus was detected in 3 of 6 ferrets within 5 to 7 days postinfection. Collectively, we demonstrate that R292K, E119V, and I222K reduced the inhibitory activity of oseltamivir, not only in the NI assay, but also in infected ferrets, judged particularly by viral loads in nasal washes, and may signal the need for alternative therapeutics. Thus, these clinical outcomes measured in the ferret model may correlate with clinically relevant oseltamivir resistance in humans. IMPORTANCE: This report provides more evidence for using the ferret model to assess the susceptibility of influenza A(H7N9) viruses to oseltamivir, the most prescribed anti-influenza virus drug. The information gained can be used to assist in the establishment of laboratory correlates of human disease and drug therapy. The rapid emergence of viruses with R292K in treated ferrets correlates well with the multiple reports on this NA variant in treated human patients. Our findings highlight the importance of the discovery and characterization of new antiviral drugs with different mechanisms of action and the use of combination treatment strategies against emerging viruses with pandemic potential, such as avian H7N9 virus, particularly against those carrying drug resistance markers.


Asunto(s)
Antivirales/farmacología , Subtipo H7N9 del Virus de la Influenza A/efectos de los fármacos , Subtipo H7N9 del Virus de la Influenza A/genética , Gripe Humana/tratamiento farmacológico , Gripe Humana/virología , Neuraminidasa/genética , Oseltamivir/farmacología , Animales , Modelos Animales de Enfermedad , Farmacorresistencia Viral/genética , Inhibidores Enzimáticos/farmacología , Hurones , Genes Virales , Humanos , Subtipo H7N9 del Virus de la Influenza A/fisiología , Masculino , Mutación , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/virología , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/virología , Replicación Viral/efectos de los fármacos
19.
J Infect Dis ; 212(8): 1209-13, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25943200

RESUMEN

Prolonged treatment of an immunocompromised child with oseltamivir and zanamivir for A(H1N1)pdm09 virus infection led to the emergence of viruses carrying H275Y and/or E119G in the neuraminidase (NA). When phenotypically evaluated by NA inhibition, the dual H275Y-E119G substitution caused highly reduced inhibition by 4 NA inhibitors: oseltamivir, zanamivir, peramivir, and laninamivir.


Asunto(s)
Antivirales/uso terapéutico , Farmacorresistencia Viral/genética , Inhibidores Enzimáticos/uso terapéutico , Subtipo H1N1 del Virus de la Influenza A/genética , Gripe Humana/virología , Neuraminidasa/genética , Ácidos Carbocíclicos , Sustitución de Aminoácidos , Ciclopentanos/uso terapéutico , Guanidinas/uso terapéutico , Humanos , Huésped Inmunocomprometido , Lactante , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Gripe Humana/tratamiento farmacológico , Masculino , Mutación Missense , Oseltamivir/uso terapéutico , Piranos , Ácidos Siálicos , Proteínas Virales/genética , Zanamivir/análogos & derivados , Zanamivir/uso terapéutico
20.
J Infect Dis ; 211(2): 249-57, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25124927

RESUMEN

BACKGROUND: Patients contracting influenza A(H7N9) infection often developed severe disease causing respiratory failure. Neuraminidase (NA) inhibitors (NAIs) are the primary option for treatment, but information on drug-resistance markers for influenza A(H7N9) is limited. METHODS: Four NA variants of A/Taiwan/1/2013(H7N9) virus containing a single substitution (NA-E119V, NA-I222K, NA-I222R, or NA-R292K) recovered from an oseltamivir-treated patient were tested for NAI susceptibility in vitro; their replicative fitness was evaluated in cell culture, mice, and ferrets. RESULTS: NA-R292K led to highly reduced inhibition by oseltamivir and peramivir, while NA-E119V, NA-I222K, and NA-I222R caused reduced inhibition by oseltamivir. Mice infected with any virus showed severe clinical signs with high mortality rates. NA-I222K virus was the most virulent in mice, whereas virus lacking NA change (NA-WT) and NA-R292K virus seemed the least virulent. Sequence analysis suggests that PB2-S714N increased virulence of NA-I222K virus in mice; NS1-K126R, alone or in combination with PB2-V227M, produced contrasting effects in NA-WT and NA-R292K viruses. In ferrets, all viruses replicated to high titers in the upper respiratory tract but produced only mild illness. NA-R292K virus, showed reduced replicative fitness in this animal model. CONCLUSIONS: Our data highlight challenges in assessment of the replicative fitness of H7N9 NA variants that emerged in NAI-treated patients.


Asunto(s)
Antivirales/uso terapéutico , Farmacorresistencia Viral , Subtipo H7N9 del Virus de la Influenza A/efectos de los fármacos , Gripe Humana/tratamiento farmacológico , Gripe Humana/virología , Oseltamivir/uso terapéutico , Animales , Modelos Animales de Enfermedad , Hurones , Humanos , Subtipo H7N9 del Virus de la Influenza A/enzimología , Subtipo H7N9 del Virus de la Influenza A/genética , Subtipo H7N9 del Virus de la Influenza A/aislamiento & purificación , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Proteínas Mutantes/genética , Mutación Missense , Neuraminidasa/genética , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Proteínas Virales/genética , Cultivo de Virus , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA