Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biologicals ; 73: 1-7, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34489162

RESUMEN

BACKGROUND: In the pre-clinical phase, SARS-CoV-2 vaccines were tested in animal models, including exposure trials, to investigate protection against SARS-CoV-2. These studies paved the way for clinical development. The objective of our review was to provide an overview of published animal exposure results, focussing on the capacity of vaccines to reduce/prevent viral shedding. METHOD: Using Medline, we retrieved eighteen papers on eight different vaccine platforms in four animal models. Data were extracted on presence/absence of viral RNA in nose, throat, or lungs, and neutralizing antibody levels in the blood. RESULTS: All vaccines showed a tendency of reduced viral load after exposure. Particularly nasal swab results are likely to give an indication about the impact on virus excretion in the environment. Similarly, the reduction or prevention of viral replication in the bronchoalveolar environment might be related with disease prevention, explaining the high efficacy in clinical trials. DISCUSSION: Although it remains difficult to compare the results directly, the potential for a strong reduction of transmission was shown, indicating that the animal models predicted what is observed in the field after large scale human vaccination. This merits further attention for standardization of exposure experiments, with the intention to speed up future vaccine development.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Vacunas contra la COVID-19 , COVID-19 , SARS-CoV-2/inmunología , Vacunación , Animales , COVID-19/inmunología , COVID-19/prevención & control , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/farmacología , Humanos
2.
Antimicrob Agents Chemother ; 60(8): 4620-9, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27185803

RESUMEN

The clinical impact of infections with respiratory viruses belonging to the family Paramyxoviridae argues for the development of antiviral therapies with broad-spectrum activity. Favipiravir (T-705) has demonstrated potent antiviral activity against multiple RNA virus families and is presently in clinical evaluation for the treatment of influenza. Here we demonstrate in vitro activity of T-705 against the paramyxoviruses human metapneumovirus (HMPV), respiratory syncytial virus, human parainfluenza virus, measles virus, Newcastle disease virus, and avian metapneumovirus. In addition, we demonstrate activity against HMPV in hamsters. T-705 treatment inhibited replication of all paramyxoviruses tested in vitro, with 90% effective concentration (EC90) values of 8 to 40 µM. Treatment of HMPV-challenged hamsters with T-705 at 200 mg/kg of body weight/day resulted in 100% protection from infection of the lungs. In all treated and challenged animals, viral RNA remained detectable in the respiratory tract. The observation that T-705 treatment had a significant effect on infectious viral titers, with a limited effect on viral genome titers, is in agreement with its proposed mode of action of viral mutagenesis. However, next-generation sequencing of viral genomes isolated from treated and challenged hamsters did not reveal (hyper)mutation. Polymerase activity assays revealed a specific effect of T-705 on the activity of the HMPV polymerase. With the reported antiviral activity of T-705 against a broad range of RNA virus families, this small molecule is a promising broad-range antiviral drug candidate for limiting the viral burden of paramyxoviruses and for evaluation for treatment of infections with (re)emerging viruses, such as the henipaviruses.


Asunto(s)
Amidas/farmacología , Antivirales/farmacología , Metapneumovirus/efectos de los fármacos , Infecciones por Paramyxoviridae/tratamiento farmacológico , Pirazinas/farmacología , Animales , Anticuerpos Antivirales/inmunología , Chlorocebus aethiops , Cricetinae , Células HEK293 , Humanos , Pulmón/virología , Mesocricetus , Virus Sincitiales Respiratorios/efectos de los fármacos , Células Vero , Replicación Viral/efectos de los fármacos
3.
J Viral Hepat ; 23(7): 512-21, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26891712

RESUMEN

Hepatitis E virus (HEV) is the causative agent of hepatitis E in humans and a member of the genus Orthohepevirus in the family Hepeviridae. Infection usually leads to acute hepatitis that can become fulminant, particularly among pregnant women and in patients with preexisting liver disease, or may evolve to a chronic state, especially in immunosuppressed individuals. HEV has been shown to produce a range of extra-hepatic manifestations including aplastic anaemia, acute thyroiditis, glomerulonephritis as well as neurological disorders such as Guillain-Barré syndrome, neuralgic amyotrophy and encephalitis. The pathogenesis of these neurological injuries remains largely unknown, and it is also uncertain whether or not HEV can directly infect neuronal cells. In this study, we investigated whether HEV is capable of completing the viral life cycle in human neuronal-derived cell lines such as neuroepithelioma (SK-N-MC), desmoplastic cerebellar medulloblastoma (DAOY), glioblastoma multiforme (DBTRG), glioblastoma astrocytoma (U-373 MG) and oligodendrocytic (M03.13) cells. Following transfection of these cells with HEV Gaussia luciferase reporter virus, all tested cell lines supported HEV RNA replication. Furthermore, extra- and intracellular viral capsid was detected by an HEV antigen ELISA as a marker for virus assembly and release. Permissiveness for HEV cell entry could be demonstrated for the oligodendrocytic cell line M03.13. In conclusion, these results indicate that HEV tropism is not restricted to the liver and HEV can potentially complete the full viral life cycle in neuronal-derived tissues explaining neurologic disorders during HEV infection.


Asunto(s)
Virus de la Hepatitis E/fisiología , Neuroglía/virología , Neuronas/virología , Tropismo Viral , Replicación Viral , Línea Celular , Humanos
4.
J Antimicrob Chemother ; 70(1): 190-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25228588

RESUMEN

OBJECTIVES: Norovirus outbreaks of acute gastroenteritis are highly prevalent, extensive and can disturb the functioning of health institutions, leading to the closure of hospital wards and causing life-threatening infections in long-term care facilities. There is no vaccine available; hence there is a pressing need for antivirals for the treatment (in immunodeficient patients) and prophylaxis of norovirus infections. We explored in a mouse model whether an inhibitor of norovirus replication can prevent/reduce transmission of the virus. METHODS: We reported recently that the viral polymerase inhibitor 2'-C-methylcytidine (2CMC) efficiently protects against murine norovirus (MNV)-induced diarrhoea and mortality in mice. Here, we established an MNV-transmission model, determined the 50% infectious dose and assessed the ability of an antiviral molecule to prevent or reduce transmission of (murine) norovirus when given either to the infected (seeder) mice or to the uninfected (sentinel) mice. RESULTS: A robust norovirus transmission model was established using genogroup V (murine) norovirus in AG129 mice. The 50% infectious dose was determined to be ∼ 270 CCID50 (50% cell culture infectious dose). Treatment of infected mice with 2CMC reduced viral shedding and markedly reduced transmission to uninfected sentinels. Also, prophylactic treatment of sentinels with 2CMC resulted in protection against infection with MNV. CONCLUSIONS: These findings constitute an important first step towards developing an efficient prophylaxis for the control of norovirus outbreaks.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Caliciviridae/prevención & control , Quimioprevención/métodos , Citidina/análogos & derivados , Norovirus/efectos de los fármacos , Animales , Citidina/uso terapéutico , Diarrea/prevención & control , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Análisis de Supervivencia
5.
Antimicrob Agents Chemother ; 58(8): 4675-81, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24890597

RESUMEN

Potent and safe inhibitors of norovirus replication are needed for the treatment and prophylaxis of norovirus infections. We here report that the in vitro anti-norovirus activity of the protease inhibitor rupintrivir is extended to murine noroviruses and that rupintrivir clears human cells from their Norwalk replicon after only two passages of antiviral pressure. In addition, we demonstrate that rupintrivir inhibits the human norovirus (genogroup II [GII]) protease and further explain the inhibitory effect of the molecule by means of molecular modeling on the basis of the crystal structure of the Norwalk virus protease. The combination of rupintrivir with the RNA-dependent RNA polymerase inhibitors 2'-C-methylcytidine and favipiravir (T-705) resulted in a merely additive antiviral effect. The fact that rupintrivir is active against noroviruses belonging to genogroup I (Norwalk virus), genogroup V (murine norovirus), and the recombinant 3C-like protease of a GII norovirus suggests that the drug exerts cross-genotypic anti-norovirus activity and will thus most likely be effective against the clinically relevant human norovirus strains. The design of antiviral molecules targeting the norovirus protease could be a valuable approach for the treatment and/or prophylaxis of norovirus infections.


Asunto(s)
Antivirales/farmacología , Cisteína Endopeptidasas/metabolismo , Isoxazoles/farmacología , Virus Norwalk/efectos de los fármacos , Pirrolidinonas/farmacología , Proteínas Virales/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Amidas/química , Amidas/farmacología , Antivirales/química , Línea Celular , Cisteína Endopeptidasas/química , Citidina/análogos & derivados , Citidina/química , Citidina/farmacología , Combinación de Medicamentos , Sinergismo Farmacológico , Hepatocitos/efectos de los fármacos , Hepatocitos/virología , Humanos , Isoxazoles/química , Simulación del Acoplamiento Molecular , Virus Norwalk/enzimología , Virus Norwalk/genética , Papaína/antagonistas & inhibidores , Papaína/química , Papaína/metabolismo , Fenilalanina/análogos & derivados , Pirazinas/química , Pirazinas/farmacología , Pirrolidinonas/química , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/metabolismo , Replicón , Valina/análogos & derivados , Proteínas Virales/química , Proteínas Virales/metabolismo
6.
Biochem Biophys Res Commun ; 424(4): 777-80, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22809499

RESUMEN

Human noroviruses are the primary cause of foodborne gastroenteritis. Potent and safe inhibitors are needed for the treatment/prophylaxis of norovirus infections. We demonstrate that Favipiravir [T-705, a drug in advanced clinical development for the treatment of infections with the influenza virus] inhibits in vitro murine norovirus replication. Time-of-drug addition studies reveal that T-705 exerts its activity at a time-point that coincides with onset of viral RNA synthesis, which is in line with the viral polymerase as the presumed target.


Asunto(s)
Amidas/farmacología , Norovirus/efectos de los fármacos , Pirazinas/farmacología , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Ratones , ARN Viral/antagonistas & inhibidores , ARN Viral/biosíntesis
7.
Biochem Biophys Res Commun ; 427(4): 796-800, 2012 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-23063849

RESUMEN

We here report on the activity of 2'-C-methylcytidine (2CMC) [a nucleoside polymerase inhibitor of the hepatitis C virus (HCV)] on the in vitro replication of (murine) norovirus (MNV). 2CMC inhibits (i) virus-induced CPE formation, (ii) viral RNA synthesis and (iii) infectious progeny formation with EC(50) values of ∼2µM. 2CMC acts at a time-point that coincides with the onset of viral RNA synthesis. Even following 30 passages of selective pressure no MNV-resistant virus was selected, which is in line with the high barrier to resistance of the nucleoside analogue for HCV. When combined with the broad-spectrum RNA virus inhibitor ribavirin, a marked antagonistic activity was observed indicating that these molecules should not be combined for the treatment of norovirus infections. Our results suggest that 2'-C-methyl nucleoside analogues should be further explored for the treatment and prophylaxis of norovirus infections.


Asunto(s)
Antivirales/farmacología , Citidina/análogos & derivados , Norovirus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Citidina/farmacología , Farmacorresistencia Viral/genética , Ratones , Norovirus/genética , Norovirus/fisiología , Ribavirina/farmacología
8.
J Viral Hepat ; 18(7): e175-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21692930

RESUMEN

Primary or secondary failure of adefovir dipivoxil (ADV) therapy of chronic hepatitis B is not infrequent. The reasons for suboptimal responses are not well defined. In HIV and hepatitis C virus infection, failure of antiviral drug therapy has been linked with low blood drug levels. We have studied 20 well-defined patients with chronic hepatitis B who were treated with ADV for drug and virus kinetics. Importantly, neither Cmax levels (mean 26 ng/mL, range 14-59 ng/mL) nor the time to maximal drug levels (mean 4 h, range 2-8 h) differed between patients showing a complete virological response to adefovir (n = 10), patients with secondary treatment failure (n = 7) and patients with suboptimal primary response (hepatitis B virus-DNA >10,000 IU/mL after 6 months of treatment; n = 3). Thus, adefovir treatment failure is unlikely to be due to an inability to mount sufficient drug levels in the blood.


Asunto(s)
Adenina/análogos & derivados , Antivirales/sangre , Hepatitis B Crónica/sangre , Hepatitis B Crónica/tratamiento farmacológico , Organofosfonatos/sangre , Adenina/administración & dosificación , Adenina/sangre , Adenina/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Antivirales/uso terapéutico , Cromatografía Líquida de Alta Presión , Femenino , Antígenos e de la Hepatitis B/análisis , Antígenos e de la Hepatitis B/inmunología , Humanos , Masculino , Persona de Mediana Edad , Organofosfonatos/administración & dosificación , Organofosfonatos/uso terapéutico , Insuficiencia del Tratamiento
9.
Nat Commun ; 12(1): 4117, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34226537

RESUMEN

Epidemiological and clinical reports indicate that SARS-CoV-2 virulence hinges upon the triggering of an aberrant host immune response, more so than on direct virus-induced cellular damage. To elucidate the immunopathology underlying COVID-19 severity, we perform cytokine and multiplex immune profiling in COVID-19 patients. We show that hypercytokinemia in COVID-19 differs from the interferon-gamma-driven cytokine storm in macrophage activation syndrome, and is more pronounced in critical versus mild-moderate COVID-19. Systems modelling of cytokine levels paired with deep-immune profiling shows that classical monocytes drive this hyper-inflammatory phenotype and that a reduction in T-lymphocytes correlates with disease severity, with CD8+ cells being disproportionately affected. Antigen presenting machinery expression is also reduced in critical disease. Furthermore, we report that neutrophils contribute to disease severity and local tissue damage by amplification of hypercytokinemia and the formation of neutrophil extracellular traps. Together our findings suggest a myeloid-driven immunopathology, in which hyperactivated neutrophils and an ineffective adaptive immune system act as mediators of COVID-19 disease severity.


Asunto(s)
COVID-19/complicaciones , COVID-19/inmunología , Síndrome de Liberación de Citoquinas/complicaciones , Monocitos/patología , Activación Neutrófila , Anciano , Células Presentadoras de Antígenos/inmunología , COVID-19/sangre , COVID-19/virología , Estudios de Casos y Controles , Síndrome de Liberación de Citoquinas/sangre , Síndrome de Liberación de Citoquinas/patología , Síndrome de Liberación de Citoquinas/virología , Citocinas/sangre , Trampas Extracelulares/metabolismo , Femenino , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Inmunofenotipificación , Masculino , Persona de Mediana Edad , SARS-CoV-2/fisiología , Índice de Severidad de la Enfermedad
10.
Handb Exp Pharmacol ; (189): 53-84, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19048197

RESUMEN

Nucleoside or nucleotide analogue inhibitors of viral replication almost act as chain terminators during DNA (DNA- and retroviruses) or RNA (RNA viruses) synthesis. Following intracellular phosphorylation, by viral and/or cellular kinases, the 5'-triphosphate metabolites (or 2'-diphosphate metabolites in the case of acyclic nucleoside phosphonate analogues) compete with the natural substrate in the DNA or RNA polymerization reaction. Obligatory chain terminators (e.g., acyclovir) do not offer the 3'-hydroxyl function at the riboside moiety of the molecule. Nucleoside analogues that possess a hydroxyl function at a position equivalent of the 3'-hydroxyl position may act as chain terminators if this hydroxyl group is conformationally constrained (e.g., ganciclovir) or sterically hindered to enter into a phosphodiester linkage with the incoming nucleotide. In case that the 3'-hydroxylgroup is correctly positioned, chain elongation may be hampered through steric hindrance from neighboring substituents (e.g., 2'-C-methyl or 4'-azido nucleoside inhibitors of HCV replication). Here, we review the molecular mechanism of action and the clinical applications of the nucleosides and nucleotides acting as chain terminators. A further discussion of clinical applications in combination therapy can be found in Chap. 12.


Asunto(s)
Antivirales/farmacología , ADN Viral/efectos de los fármacos , ARN Viral/efectos de los fármacos , Animales , Antivirales/síntesis química , Hepacivirus/efectos de los fármacos , Virus de la Hepatitis B/efectos de los fármacos , Humanos , Nucleósidos/síntesis química , Nucleósidos/farmacología
11.
Antimicrob Agents Chemother ; 52(11): 4064-8, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18765685

RESUMEN

Caprine herpesvirus 1 (CpHV-1) infection in goats induces genital vesicular-ulcerative lesions that strictly resemble those produced by human herpesvirus 2 in humans. In previous studies, the potent inhibition of CpHV-1 by cidofovir was demonstrated. Cidofovir antiherpetic activity was evaluated in goats infected experimentally by the vaginal route with CpHV-1 and then treated locally at different times after infection. The administration of 1% cidofovir cream onto vaginal mucosa was able to prevent the onset of genital lesions and to decrease significantly the titers of the virus shed by the infected animals, notably in the groups treated shortly after infection (24 and 48 h). The efficacy of cidofovir against caprine herpesvirus infection was higher when the treatment was started shortly after infection than when lesions were already present and advanced. Herpesvirus genital infection of goats is a useful animal model to study the activity of antiviral drugs against human herpesvirus infections.


Asunto(s)
Antivirales/uso terapéutico , Citosina/análogos & derivados , Enfermedades de los Genitales Femeninos/veterinaria , Enfermedades de las Cabras/tratamiento farmacológico , Infecciones por Herpesviridae/veterinaria , Organofosfonatos/uso terapéutico , Varicellovirus , Animales , Antivirales/administración & dosificación , Secuencia de Bases , Cidofovir , Citosina/administración & dosificación , Citosina/uso terapéutico , Cartilla de ADN/genética , ADN Viral/genética , Modelos Animales de Enfermedad , Femenino , Enfermedades de los Genitales Femeninos/tratamiento farmacológico , Enfermedades de las Cabras/virología , Cabras , Herpes Genital/tratamiento farmacológico , Infecciones por Herpesviridae/tratamiento farmacológico , Infecciones por Herpesviridae/virología , Humanos , Organofosfonatos/administración & dosificación , Especificidad de la Especie , Cremas, Espumas y Geles Vaginales , Varicellovirus/efectos de los fármacos , Varicellovirus/genética , Varicellovirus/aislamiento & purificación
12.
Antiviral Res ; 77(2): 114-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17997169

RESUMEN

Selective inhibitors of the replication of the classical swine fever virus (CSFV) may have the potential to control the spread of the infection in an epidemic situation. We here report that 5-[(4-bromophenyl)methyl]-2-phenyl-5H-imidazo[4,5-c]pyridine (BPIP) is a highly potent inhibitor of the in vitro replication of CSFV. The compound resulted in a dose-dependent antiviral effect in PK(15) cells with a 50% effective concentration (EC(50)) for the inhibition of CSFV Alfort(187) (subgroup 1.1) of 1.6+/-0.4 microM and for CSFV Wingene (subgroup 2.3) 0.8+/-0.2 microM. Drug-resistant virus was selected by serial passage of the virus in increasing drug-concentration. The BPIP-resistant virus (EC(50): 24+/-4.0 microM) proved cross-resistant with VP32947 [3-[((2-dipropylamino)ethyl)thio]-5H-1,2,4-triazino[5,6-b]indole], an unrelated earlier reported selective inhibitor of pestivirus replication. BPIP-resistant CSFV carried a T259S mutation in NS5B, encoding the RNA-dependent RNA-polymerase (RdRp). This mutation is located near F224, a residue known to play a crucial role in the antiviral activity of BPIP against bovine viral diarrhoea virus (BVDV). The T259S mutation was introduced in a computational model of the BVDV RdRp. Molecular docking of BPIP in the BVDV polymerase suggests that T259S may have a negative impact on the stacking interaction between the imidazo[4,5-c]pyridine ring system of BPIP and F224.


Asunto(s)
Antivirales/farmacología , Virus de la Fiebre Porcina Clásica/efectos de los fármacos , Imidazoles/farmacología , Piridinas/farmacología , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Sustitución de Aminoácidos , Animales , Antivirales/química , Línea Celular , Virus de la Fiebre Porcina Clásica/fisiología , Virus de la Diarrea Viral Bovina/efectos de los fármacos , Farmacorresistencia Viral/efectos de los fármacos , Imagenología Tridimensional , Imidazoles/química , Concentración 50 Inhibidora , Modelos Moleculares , Estructura Molecular , Reacción en Cadena de la Polimerasa , Piridinas/química , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/genética , Análisis de Secuencia de ADN , Relación Estructura-Actividad
13.
Antiviral Res ; 78(1): 37-46, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18083241

RESUMEN

Life-threatening RNA viruses emerge regularly, and often in an unpredictable manner. Yet, the very few drugs available against known RNA viruses have sometimes required decades of research for development. Can we generate preparedness for outbreaks of the, as yet, unknown viruses? The VIZIER (VIral enZymes InvolvEd in Replication) (http://www.vizier-europe.org/) project has been set-up to develop the scientific foundations for countering this challenge to society. VIZIER studies the most conserved viral enzymes (that of the replication machinery, or replicases) that constitute attractive targets for drug-design. The aim of VIZIER is to determine as many replicase crystal structures as possible from a carefully selected list of viruses in order to comprehensively cover the diversity of the RNA virus universe, and generate critical knowledge that could be efficiently utilized to jump-start research on any emerging RNA virus. VIZIER is a multidisciplinary project involving (i) bioinformatics to define functional domains, (ii) viral genomics to increase the number of characterized viral genomes and prepare defined targets, (iii) proteomics to express, purify, and characterize targets, (iv) structural biology to solve their crystal structures, and (v) pre-lead discovery to propose active scaffolds of antiviral molecules.


Asunto(s)
Antivirales/farmacología , Biología Computacional , Cristalografía , Diseño de Fármacos , Genómica , Proteómica , Virus ARN/efectos de los fármacos , ARN Polimerasa Dependiente del ARN , Replicación Viral/efectos de los fármacos , Antivirales/química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Cooperación Internacional , Modelos Moleculares , Virus ARN/enzimología , Virus ARN/patogenicidad , Virus ARN/fisiología , ARN Viral/biosíntesis , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo
14.
Vet Microbiol ; 122(1-2): 43-51, 2007 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-17276631

RESUMEN

Equine herpesvirus 1 (EHV-1) is an important equine pathogen that causes respiratory disease, abortion, neonatal death and paralysis. Although vaccines are available, they are not fully protective and outbreaks of disease may occur in vaccinated herds. Therefore, there is an urgent need for effective antiviral treatment. For three abortigenic (94P247, 97P70 and 99P96) and three neuropathogenic isolates (97P82, 99P136 and 03P37), the effect of acyclovir, ganciclovir, cidofovir, adefovir, 9-(2-phosphonylmethoxyethyl)-2,6-diaminopurine (PMEDAP) and foscarnet on plaque number was studied. Additionally, for isolate 97P70, the effect on plaque size was investigated. Ganciclovir was most potent in reducing plaque number, followed by PMEDAP and acyclovir. Adefovir and cidofovir were less effective and foscarnet was the least effective compound. There were no differences detected for acyclovir, ganciclovir, adefovir and PMEDAP between the abortigenic and neuropathogenic isolates. One abortigenic isolate (99P96) was more susceptible to cidofovir and two neuropathogenic isolates (99P136 and 03P37) were less susceptible to foscarnet. For isolate 97P70, all compounds resulted in a significant reduction of plaque size. The most remarkable effect was observed for cidofovir. It was 40-fold more effective in reducing plaque size than in reducing plaque number. In conclusion, ganciclovir was the most potent compound and therefore, may be a valuable candidate for the treatment of EHV-1 infections in horses. The antiviral effect of foscarnet on plaque number was highly dependent on the viral isolate tested. Therefore, it is no valuable antiviral for the treatment of herpesvirus-infections. Cidofovir, although less effective in reducing plaque number, had a strong effect on plaque size.


Asunto(s)
Aciclovir/farmacología , Adenina/análogos & derivados , Citosina/análogos & derivados , Foscarnet/farmacología , Ganciclovir/farmacología , Herpesvirus Équido 1/efectos de los fármacos , Organofosfonatos/farmacología , Adenina/farmacología , Animales , Antivirales/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cidofovir , Citosina/farmacología , Relación Dosis-Respuesta a Droga , Farmacorresistencia Viral , Células Epiteliales/efectos de los fármacos , Caballos , Pulmón/citología
15.
Glob Health Action ; 10(1): 1398485, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29235414

RESUMEN

The ongoing Zika virus (ZIKV) outbreak in Latin America, the Caribbean, and the Pacific Islands has underlined the need for a coordinated research network across the whole region that can respond rapidly to address the current knowledge gaps in Zika and enhance research preparedness beyond Zika. The European Union under its Horizon 2020 Research and Innovation Programme awarded three research consortia to respond to this need. Here we present the ZikaPLAN (Zika Preparedness Latin American Network) consortium. ZikaPLAN combines the strengths of 25 partners in Latin America, North America, Africa, Asia, and various centers in Europe. We will conduct clinical studies to estimate the risk and further define the full spectrum and risk factors of congenital Zika virus syndrome (including neurodevelopmental milestones in the first 3 years of life), delineate neurological complications associated with ZIKV due to direct neuroinvasion and immune-mediated responses in older children and adults, and strengthen surveillance for birth defects and Guillain-Barré Syndrome. Laboratory-based research to unravel neurotropism and investigate the role of sexual transmission, determinants of severe disease, and viral fitness will underpin the clinical studies. Social messaging and engagement with affected communities, as well as development of wearable repellent technologies against Aedes mosquitoes will enhance the impact. Burden of disease studies, data-driven vector control, and vaccine modeling as well as risk assessments on geographic spread of ZIKV will form the foundation for evidence-informed policies. While addressing the research gaps around ZIKV, we will engage in capacity building in laboratory and clinical research, collaborate with existing and new networks to share knowledge, and work with international organizations to tackle regulatory and other bottlenecks and refine research priorities. In this way, we can leverage the ZIKV response toward building a long-term emerging infectious diseases response capacity in the region to address future challenges.


Asunto(s)
Control de Mosquitos/métodos , Infección por el Virus Zika/epidemiología , Infección por el Virus Zika/prevención & control , Aedes/virología , Animales , Investigación Biomédica/organización & administración , Creación de Capacidad , Niño , Conducta Cooperativa , Brotes de Enfermedades , Humanos , Relaciones Interinstitucionales , América Latina/epidemiología , Mosquitos Vectores , Vigilancia en Salud Pública , Medición de Riesgo , Factores de Riesgo
16.
Cancer Res ; 59(10): 2376-83, 1999 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-10344747

RESUMEN

Hemangiomas represent the most frequent tumors of infancy. However, the pathogenesis of these tumors is still largely unknown, and current treatment of juvenile hemangiomas remains unsatisfactory. Here we present a novel animal model to study proliferating hemangiomas and to evaluate the effect of angiostatic compounds on their growth. Intraperitoneal (i.p.) infection of 4-day-old rats with murine polyomavirus resulted in the development of multiple cutaneous, intramuscular (i.m.), and cerebral hemangiomas with 100% frequency. Histological examination of the brain revealed the formation of immature lesions as soon as 4 days postinfection (p.i.). The subsequent exponential growth of the hemangiomas, both in number and size, was associated with severe hemorrhage and anemia. The cerebral, cutaneous, and i.m. lesions consisted of blood-filled cysts, histologically similar to human cavernous hemangiomas and stained positive for proliferating cell nuclear antigen, urokinase-type plasminogen activator, and vascular endothelial growth factor. Mature cerebral hemangiomas also expressed von Willebrand factor. Cerebral lesions caused death of the untreated animals within 19.2 +/- 1.1 days p.i. Remarkably fewer and smaller hemangiomas developed in animals that had been treated s.c. with the angiogenesis inhibitor TNP-470. Accordingly, TNP-470 (50 mg/kg), administered twice a week from 3 days p.i., significantly delayed tumor-associated mortality [mean day of death, 28.2 +/- 3.3 (P < 0.001)]. Even if therapy was initiated when cerebral hemangiomas were already macroscopically visible (i.e., 9 days p.i.), a significant delay in hemangioma-associated mortality was observed. Also, the IFN-inducer polyinosinic-polycytidylic acid caused a delay of 9 days (P < 0.005) in tumor-associated mortality when administered i.p. at 5 mg/kg, twice a week, starting at day 3 p.i. The model described here may be useful for investigating (a) the angiogenic mechanism(s) underlying hemangioma progression; and (b) the effect of anti-angiogenic compounds on vascular tumor growth.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/prevención & control , Transformación Celular Viral , Modelos Animales de Enfermedad , Hemangioma/prevención & control , Neovascularización Patológica/tratamiento farmacológico , Infecciones por Polyomavirus/tratamiento farmacológico , Poliomavirus/patogenicidad , Sesquiterpenos/uso terapéutico , Infecciones Tumorales por Virus/tratamiento farmacológico , Animales , Animales Recién Nacidos , Peso Corporal/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/virología , Ciclohexanos , Progresión de la Enfermedad , Endotelio Vascular/efectos de los fármacos , Femenino , Hemangioma/tratamiento farmacológico , Hemangioma/etiología , Hemangioma/virología , Humanos , Lactante , Recién Nacido , Inyecciones Subcutáneas , Inductores de Interferón/uso terapéutico , Masculino , O-(Cloroacetilcarbamoil) Fumagilol , Poli I-C/uso terapéutico , Ratas , Factores Sexuales , Carga Viral
17.
Cancer Res ; 58(12): 2562-7, 1998 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-9635579

RESUMEN

The acyclic nucleoside phosphonate analogue cidofovir elicited a marked protection against hemangioma growth in newborn rats that had been infected i.p. with a high titer of murine polyomavirus. Untreated, infected rats developed cutaneous, i.m., and cerebral hemangiomas associated with severe hemorrhage and anemia leading to death within 3 weeks postinfection (p.i.). s.c. treatment with cidofovir at 25 mg/kg, once a week, resulted in a complete suppression of hemangioma development and associated mortality when treatment was initiated at 3 days p.i. (100% survival compared with 0% for the untreated animals). Cidofovir still afforded 40% survival and a significant delay in tumor-associated mortality when treatment was started at a time at which cerebral hemangiomas were already macroscopically visible (i.e., 9 days p.i.). Infectious virus or viral DNA was undetectable in the brain at different times p.i. as assessed by means of (a) a DNA-DNA hybridization assay and (b) titration of the brain for infectious virus content, indicating that there was no viral replication in murine polyomavirus-infected rats. Moreover, a semiquantitative PCR for viral protein 1 DNA revealed that the amount of viral protein 1 DNA declined with time after infection to become virtually undetectable at 18 days p.i. Therefore, an antitumor or antiangiogenic effect, rather than inhibition of viral replication, may be the reason for the inhibitory activity of cidofovir in this model. Cidofovir may thus be further explored for the treatment of vascular tumors and, in particular, life-threatening juvenile hemangiomas.


Asunto(s)
Antivirales/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Citosina/análogos & derivados , Hemangioma/tratamiento farmacológico , Organofosfonatos , Compuestos Organofosforados/uso terapéutico , Poliomavirus/efectos de los fármacos , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Peso Corporal/efectos de los fármacos , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/virología , Cidofovir , Citosina/uso terapéutico , Hemangioma/mortalidad , Hemangioma/virología , Incidencia , Ratas , Ratas Wistar , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/virología , Análisis de Supervivencia
18.
Cancer Res ; 58(3): 384-8, 1998 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-9458076

RESUMEN

The effect of the antiviral agent (S)-1-(3-hydroxy-2-phosphonylmethoxypropyl) cytosine (cidofovir) on the EBV-associated tumor nasopharyngeal carcinoma (NPC) was evaluated in NPC xenografts in athymic mice. Intratumoral injection arrested tumor growth within 1 week, and by 4 weeks, tumors regressed to 8-75% (39 +/- 33%) of the original size, whereas control tumors injected with PBS grew to 282 +/- 25% of the original size. Ganciclovir slowed but did not arrest or cause regression of tumor growth. A striking antitumor effect was also produced by systemic administration; at 4 weeks, tumors were 79 +/- 49% of the original size, compared with 635 +/- 91% for the controls. Widespread apoptosis was detected after treatment for 2-6 days in C15 as well as two other NPC xenografts, C17 and C18; the latter NPCs have mutations in the p53 gene. These data indicate that cidofovir induces rapid cell death through apoptosis in EBV-transformed epithelial cells.


Asunto(s)
Antivirales/uso terapéutico , Carcinoma/tratamiento farmacológico , Citosina/análogos & derivados , Infecciones por Herpesviridae/tratamiento farmacológico , Herpesvirus Humano 4/efectos de los fármacos , Neoplasias Nasofaríngeas/tratamiento farmacológico , Organofosfonatos , Compuestos Organofosforados/uso terapéutico , Infecciones Tumorales por Virus/tratamiento farmacológico , Adolescente , Animales , Apoptosis/efectos de los fármacos , Carcinoma/genética , Carcinoma/patología , Carcinoma/virología , Cidofovir , Citosina/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Ganciclovir/farmacología , Ganciclovir/uso terapéutico , Genes p53 , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Herpesvirus Humano 4/patogenicidad , Humanos , Inyecciones Intralesiones , Ratones , Ratones Desnudos , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patología , Neoplasias Nasofaríngeas/virología , Trasplante de Neoplasias , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/virología
19.
Cancer Res ; 61(13): 5057-64, 2001 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-11431341

RESUMEN

Cidofovir [(S)-HPMPC; (S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine] is an antiviral drug that has been approved for the treatment of cytomegalovirus retinitis in AIDS patients. Cidofovir also possesses potent inhibitory activity against various human papillomavirus-induced tumors in animal models and patients. In addition, cidofovir inhibits the development of murine polyomavirus-induced hemangiomas in rats by an as-yet-uncharacterized, antivirus-independent mechanism. Here we report the inhibitory effect of cidofovir on the development of virus-independent vascular tumors originated by basic fibroblast growth factor (FGF2)-overexpressing endothelial cells (FGF2-T-MAE cells). In vitro, cidofovir was cytostatic to FGF2-T-MAE cells at a 50% cytostatic concentration of 6.7 microg/ml. Cidofovir concentrations >25 microg/ml resulted in cytotoxicity because of induction of apoptosis. Cidofovir did not affect FGF2-T-MAE cell sprouting in three-dimensional fibrin gel and morphogenesis on Matrigel at noncytotoxic concentrations. In vivo, cidofovir (100 microg/egg) completely suppressed hemangioma formation on the chick chorioallantoic membrane (CAM) induced by intra-allantoic injection of FGF2-T-MAE cells, without affecting the formation of normal CAM vessels. Accordingly, cidofovir applied locally at 200 microg/disc, reduced neovascularization on the CAM by only 35%. Intratumoral or systemic administration of cidofovir caused a significant inhibition of the growth of s.c., i.p., or intracerebral FGF2-T-MAE xenografts in nude mice and severe combined immunodeficient mice. Drug-induced apoptosis was observed in FGF2-T-MAE tumors as soon as 2 days after the beginning of treatment. In conclusion, cidofovir appears to inhibit the growth of endothelium-derived tumors via induction of apoptosis without exerting a direct antiangiogenic activity.


Asunto(s)
Antineoplásicos/farmacología , Citosina/farmacología , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Hemangioendotelioma/tratamiento farmacológico , Organofosfonatos , Compuestos Organofosforados/farmacología , Animales , Antivirales/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/tratamiento farmacológico , División Celular/efectos de los fármacos , Embrión de Pollo , Cidofovir , Citosina/análogos & derivados , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Femenino , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/fisiología , Hemangioendotelioma/irrigación sanguínea , Hemangioendotelioma/patología , Ratones , Ratones Desnudos , Ratones SCID , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Fisiológica/efectos de los fármacos , Transfección , Células Tumorales Cultivadas
20.
Transbound Emerg Dis ; 63(2): e205-12, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25164494

RESUMEN

An antiviral containment strategy for foot-and-mouth disease (FMD) outbreaks could support or replace current contingency plans in case of an outbreak in Europe and could spare many healthy animals from being pre-emptively culled. Recently, substantial progress has been made towards the development of small molecule drugs that inhibit FMD virus (FMDV) replication in vitro. For the initial in vivo evaluation of antiviral lead molecules, a refined FMDV-infection model in guinea pigs (GP) is herewith described. This GP model was validated by demonstrating the antiviral effect of T-1105 (an influenza virus inhibitor with reported activity against FMDV). Sixteen animals were orally administered with T-1105 twice daily (400 mg/kg/day) for five consecutive days and inoculated intraplantarly with 100 GPID50 of the GP-adapted FMDV strain O1 Manisa 1 h after the first administration. The efficacy of T-1105 was compared with that of prophylactic vaccination with a highly potent double-oil emulsion-inactivated O1 Manisa vaccine. Ten animals received a single, full (2 ml) cattle vaccine dose and were inoculated 3 weeks later. Fourteen T-1105-treated and all vaccinated GP were completely protected from generalization of vesicular lesions. At 2 dpi, viral RNA was detected in serum of 9/16 T-1105-treated and of 6/10 vaccinated animals. At 4 dpi, viral RNA was detected in serum, organs and oral swabs of half of the T-1105-treated animals and only in the serum of 1/10 of the vaccinated animals. Mean viral RNA levels in serum and organs of T-1105-treated and vaccinated animals were reduced compared to untreated controls (P < 0.01). T-1105 conferred a substantial clinical and virological protection against infection with O1 Manisa, similar to the protection afforded by vaccination. These results validate the suitability of the enhanced GP model for the purpose of initial evaluation of inhibitors of FMDV replication and illustrate the potential of selective inhibitors of viral replication to control FMD outbreaks.


Asunto(s)
Antivirales/uso terapéutico , Fiebre Aftosa/tratamiento farmacológico , Pirazinas/uso terapéutico , Animales , Anticuerpos Antivirales/sangre , Modelos Animales de Enfermedad , Europa (Continente) , Fiebre Aftosa/prevención & control , Virus de la Fiebre Aftosa/aislamiento & purificación , Cobayas , ARN Viral/sangre , Vacunación/veterinaria , Vacunas Virales/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA