Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Antimicrob Agents Chemother ; 59(4): 2086-93, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25624323

RESUMEN

Dengue virus (DENV) is the most prevalent mosquito-borne viral pathogen in humans. Currently, there is no clinically approved vaccine or antiviral for DENV. Combination therapy is a common practice in antiviral treatment and a potential approach to search for new treatments for infectious pathogens. In this study, we performed a combination treatment in cell culture by using three distinct classes of inhibitors, including ribavirin (a guanosine analog with several antiviral mechanisms), brequinar (a pyrimidine biosynthesis inhibitor), and INX-08189 (a guanosine analog). The compound pairs were evaluated for antiviral activity by use of a DENV-2 luciferase replicon assay. Our result indicated that the combination of ribavirin and INX-08189 exhibited strong antiviral synergy. This result suggests that synergy can be achieved with compound pairs in which one compound suppresses the synthesis of the nucleoside for which the other compound is a corresponding nucleoside analog. In addition, we found that treatment of cells with brequinar alone could activate interferon-stimulated response elements (ISREs); furthermore, brequinar and NITD-982 (another pyrimidine biosynthesis inhibitor) potentiated interferon-induced ISRE activation. Compared to treatment with brequinar, treatment of cells with ribavirin alone could also induce ISRE activation, but to a lesser extent; however, when cells were cotreated with ribavirin and beta interferon, ribavirin did not augment the interferon-induced ISRE activation.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Nucleósidos/antagonistas & inhibidores , Nucleósidos/farmacología , Replicación Viral/efectos de los fármacos , Línea Celular , Combinación de Medicamentos , Sinergismo Farmacológico , Células HEK293 , Humanos , Inductores de Interferón/farmacología , Interferón beta/farmacología , Nucleósidos/biosíntesis , Oxidorreductasas/antagonistas & inhibidores , Ribavirina/farmacología
2.
J Virol ; 85(21): 11183-95, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21865382

RESUMEN

We report a novel inhibitor that selectively suppresses dengue virus (DENV) by targeting viral NS4B protein. The inhibitor was identified by screening a 1.8-million-compound library using a luciferase replicon of DENV serotype 2 (DENV-2). The compound specifically inhibits all four serotypes of DENV (50% effective concentration [EC(50)], 1 to 4 µM; and 50% cytotoxic concentration [CC(50)], >40 µM), but it does not inhibit closely related flaviviruses (West Nile virus and yellow fever virus) or nonflaviviruses (Western equine encephalomyelitis virus, Chikungunya virus, and vesicular stomatitis virus). A mode-of-action study suggested that the compound inhibits viral RNA synthesis. Replicons resistant to the inhibitor were selected in cell culture. Sequencing of the resistant replicons revealed two mutations (P104L and A119T) in the viral NS4B protein. Genetic analysis, using DENV-2 replicon and recombinant viruses, demonstrated that each of the two NS4B mutations alone confers partial resistance and double mutations confer additive resistance to the inhibitor in mammalian cells. In addition, we found that a replication defect caused by a lethal NS4B mutation could be partially rescued through trans complementation. The ability to complement NS4B in trans affected drug sensitivity when a single cell was coinfected with drug-sensitive and drug-resistant viruses. Mechanistically, NS4B was previously shown to interact with the viral NS3 helicase domain; one of the two NS4B mutations recovered in our resistance analysis-P104L-abolished the NS3-NS4B interaction (I. Umareddy, A. Chao, A. Sampath, F. Gu, and S. G. Vasudevan, J. Gen. Virol. 87:2605-2614, 2006). Collectively, the results suggest that the identified inhibitor targets the DENV NS4B protein, leading to a defect in viral RNA synthesis.


Asunto(s)
Antivirales/metabolismo , Virus del Dengue/efectos de los fármacos , Virus del Dengue/crecimiento & desarrollo , Proteínas no Estructurales Virales/antagonistas & inhibidores , Factores de Virulencia/antagonistas & inhibidores , Animales , Antivirales/aislamiento & purificación , Línea Celular , Análisis Mutacional de ADN , Evaluación Preclínica de Medicamentos , Farmacorresistencia Viral , Humanos , Pruebas de Sensibilidad Microbiana , ARN Viral/biosíntesis , Proteínas no Estructurales Virales/genética , Factores de Virulencia/genética
3.
J Virol ; 85(13): 6548-56, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21507975

RESUMEN

Viral replication relies on the host to supply nucleosides. Host enzymes involved in nucleoside biosynthesis are potential targets for antiviral development. Ribavirin (a known antiviral drug) is such an inhibitor that suppresses guanine biosynthesis; depletion of the intracellular GTP pool was shown to be the major mechanism to inhibit flavivirus. Along similar lines, inhibitors of the pyrimidine biosynthesis pathway could be targeted for potential antiviral development. Here we report on a novel antiviral compound (NITD-982) that inhibits host dihydroorotate dehydrogenase (DHODH), an enzyme required for pyrimidine biosynthesis. The inhibitor was identified through screening 1.8 million compounds using a dengue virus (DENV) infection assay. The compound contains an isoxazole-pyrazole core structure, and it inhibited DENV with a 50% effective concentration (EC(50)) of 2.4 nM and a 50% cytotoxic concentration (CC(50)) of >5 µM. NITD-982 has a broad antiviral spectrum, inhibiting both flaviviruses and nonflaviviruses with nanomolar EC(90)s. We also show that (i) the compound inhibited the enzymatic activity of recombinant DHODH, (ii) an NITD-982 analogue directly bound to the DHODH protein, (iii) supplementing the culture medium with uridine reversed the compound-mediated antiviral activity, and (iv) DENV type 2 (DENV-2) variants resistant to brequinar (a known DHODH inhibitor) were cross resistant to NITD-982. Collectively, the results demonstrate that the compound inhibits DENV through depleting the intracellular pyrimidine pool. In contrast to the in vitro potency, the compound did not show any efficacy in the DENV-AG129 mouse model. The lack of in vivo efficacy is likely due to the exogenous uptake of pyrimidine from the diet or to a high plasma protein-binding activity of the current compound.


Asunto(s)
Antivirales/farmacología , Antivirales/uso terapéutico , Virus del Dengue/efectos de los fármacos , Dengue/tratamiento farmacológico , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/antagonistas & inhibidores , Pirimidinas/antagonistas & inhibidores , Animales , Antivirales/química , Antivirales/farmacocinética , Chlorocebus aethiops , Efecto Citopatogénico Viral/efectos de los fármacos , Dengue/virología , Virus del Dengue/enzimología , Virus del Dengue/patogenicidad , Virus del Dengue/fisiología , Dihidroorotato Deshidrogenasa , Modelos Animales de Enfermedad , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Pirimidinas/biosíntesis , Sigmodontinae , Resultado del Tratamiento , Células Vero , Replicación Viral/efectos de los fármacos
4.
Bioorg Med Chem ; 20(20): 6123-33, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22985963

RESUMEN

A series of new pyrimido[4,5-b]indole ribonucleosides bearing phenyl or hetaryl group at position 4 has been prepared by selective Pd-catalyzed cross-coupling reactions of the corresponding protected 4,6-dichloropyrimido[4,5-b]indole ribonucleoside with (het)arylboronic acids or stannanes followed by deprotection. Further cross-couplings under harsher conditions and employing X-Phos ligand proceeded at the position 6 leading to 4,6-disubstituted pyrimido[4,5-b]indole ribonucleosides. Some of these compounds displayed antiviral activity against Dengue virus.


Asunto(s)
Antivirales/síntesis química , Indoles/química , Pirimidinas/química , Ribonucleósidos/síntesis química , Antivirales/farmacología , Antivirales/toxicidad , Catálisis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Virus del Dengue/efectos de los fármacos , Virus del Dengue/metabolismo , Células Hep G2 , Hepacivirus/efectos de los fármacos , Hepacivirus/metabolismo , Humanos , Paladio , Ribonucleósidos/farmacología , Ribonucleósidos/toxicidad , Replicación Viral/efectos de los fármacos
5.
Proc Natl Acad Sci U S A ; 106(48): 20435-9, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19918064

RESUMEN

Dengue virus (DENV), a mosquito-borne flavivirus, is a major public health threat. The virus poses risk to 2.5 billion people worldwide and causes 50 to 100 million human infections each year. Neither a vaccine nor an antiviral therapy is currently available for prevention and treatment of DENV infection. Here, we report a previously undescribed adenosine analog, NITD008, that potently inhibits DENV both in vitro and in vivo. In addition to the 4 serotypes of DENV, NITD008 inhibits other flaviviruses, including West Nile virus, yellow fever virus, and Powassan virus. The compound also suppresses hepatitis C virus, but it does not inhibit nonflaviviruses, such as Western equine encephalitis virus and vesicular stomatitis virus. A triphosphate form of NITD008 directly inhibits the RNA-dependent RNA polymerase activity of DENV, indicating that the compound functions as a chain terminator during viral RNA synthesis. NITD008 has good in vivo pharmacokinetic properties and is biologically available through oral administration. Treatment of DENV-infected mice with NITD008 suppressed peak viremia, reduced cytokine elevation, and completely prevented the infected mice from death. No observed adverse effect level (NOAEL) was achieved when rats were orally dosed with NITD008 at 50 mg/kg daily for 1 week. However, NOAEL could not be accomplished when rats and dogs were dosed daily for 2 weeks. Nevertheless, our results have proved the concept that a nucleoside inhibitor could be developed for potential treatment of flavivirus infections.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/metabolismo , Dengue/tratamiento farmacológico , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Viremia/tratamiento farmacológico , Adenosina/química , Animales , Antivirales/farmacocinética , Antivirales/uso terapéutico , Chlorocebus aethiops , Perros , Ensayo de Inmunoadsorción Enzimática , Femenino , Masculino , Ratones , Estructura Molecular , Nivel sin Efectos Adversos Observados , Ratas , Células Vero
6.
Antimicrob Agents Chemother ; 55(9): 4072-80, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21730119

RESUMEN

We describe a novel translation inhibitor that has anti-dengue virus (DENV) activity in vitro and in vivo. The inhibitor was identified through a high-throughput screening using a DENV infection assay. The compound contains a benzomorphan core structure. Mode-of-action analysis indicated that the compound inhibits protein translation in a viral RNA sequence-independent manner. Analysis of the stereochemistry demonstrated that only one enantiomer of the racemic compound inhibits viral RNA translation. Medicinal chemistry was performed to eliminate a metabolically labile glucuronidation site of the compound to improve its in vivo stability. Pharmacokinetic analysis showed that upon a single subcutaneous dosing of 25 mg/kg of body weight in mice, plasma levels of the compound reached a C(max) (maximum plasma drug concentration) above the protein-binding-adjusted 90% effective concentration (EC(90)) value of 0.96 µM. In agreement with the in vivo pharmacokinetic results, treatment of DENV-infected mice with 25 mg/kg of compound once per day reduced peak viremia by about 40-fold. However, mice treated with 75 mg/kg of compound per day exhibited adverse effects. Collectively, our results demonstrate that the benzomorphan compounds inhibit DENV through suppression of RNA translation. The therapeutic window of the current compounds needs to be improved for further development.


Asunto(s)
Antivirales/farmacología , Antivirales/farmacocinética , Virus del Dengue/efectos de los fármacos , Animales , Antivirales/efectos adversos , Antivirales/química , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Cromatografía Líquida de Alta Presión , Cricetinae , Virus del Dengue/genética , Femenino , Humanos , Ratones , Estructura Molecular , ARN Viral/genética , Ratas , Células Vero
7.
Antimicrob Agents Chemother ; 54(9): 3686-95, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20606073

RESUMEN

Brequinar is an inhibitor of dihydroorotate dehydrogenase, an enzyme that is required for de novo pyrimidine biosynthesis. Here we report that brequinar has activity against a broad spectrum of viruses. The compound not only inhibits flaviviruses (dengue virus, West Nile virus, yellow fever virus, and Powassan virus) but also suppresses a plus-strand RNA alphavirus (Western equine encephalitis virus) and a negative-strand RNA rhabdovirus (vesicular stomatitis virus). Using dengue virus serotype 2 (DENV-2) as a model, we found that brequinar suppressed the viral infection cycle mainly at the step of RNA synthesis. Supplementing the culture medium with pyrimidines (cytidine or uridine) but not purines (adenine or guanine) could be used to reverse the inhibitory effect of the compound. Continuous culturing of DENV-2 in the presence of brequinar generated viruses that were partially resistant to the inhibitor. Sequencing of the resistant viruses revealed two amino acid mutations: one mutation (M260V) located at a helix in the domain II of the viral envelope protein and another mutation (E802Q) located at the priming loop of the nonstructural protein 5 (NS5) polymerase domain. Functional analysis of the mutations suggests that the NS5 mutation exerts resistance through enhancement of polymerase activity. The envelope protein mutation reduced the efficiency of virion assembly/release; however, the mutant virus became less sensitive to brequinar inhibition at the step of virion assembly/release. Taken together, the results indicate that (i) brequinar blocks DENV RNA synthesis through depletion of intracellular pyrimidine pools and (ii) the compound may also exert its antiviral activity through inhibition of virion assembly/release.


Asunto(s)
Antivirales/farmacología , Compuestos de Bifenilo/farmacología , Virus del Dengue/efectos de los fármacos , Animales , Chlorocebus aethiops , Virus del Dengue/genética , Farmacorresistencia Viral , Pirimidinas/farmacología , ARN Viral/genética , Células Vero
8.
Antimicrob Agents Chemother ; 54(8): 3255-61, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20516277

RESUMEN

Dengue virus (DENV) is the most prevalent mosquito-borne viral pathogen that infects humans. Neither a vaccine nor an antiviral therapy is currently available for DENV. Here, we report an adenosine nucleoside prodrug that potently inhibits DENV replication both in cell culture and in a DENV mouse model. NITD449 (2'-C-acetylene-7-deaza-7-carbamoyladenosine) was initially identified as a parental compound that inhibits all four serotypes of DENV with low cytotoxicity. However, in vivo pharmacokinetic studies indicated that NITD449 had a low level of exposure in plasma when dosed orally. To increase the oral bioavailability, we covalently linked isobutyric acids to the 3'- and 5'-hydroxyl groups of ribose via ester linkage to NITD449, leading to the prodrug NITD203 (3',5'-O-diisobutyryl-2'-C-acetylene-7-deaza-7-carbamoyl-adenosin). Pharmacokinetic analysis showed that upon oral dosing of the prodrug, NITD203 was readily converted to NITD449, resulting in improved exposure of the parental compound in plasma in both mouse and rat. In DENV-infected AG129 mice, oral dosing of the prodrug at 25 mg/kg of body weight reduced peak viremia by 30-fold. Antiviral spectrum analysis showed that NITD203 inhibited various flaviviruses (DENV, yellow fever virus, and West Nile virus) and hepatitis C virus but not Chikungunya virus (an alphavirus). Mode-of-action analysis, using a luciferase-reporting replicon, indicated that NITD203 inhibited DENV RNA synthesis. Although NITD203 exhibited potent in vitro and in vivo efficacies, the compound could not reach a satisfactory no-observable-adverse-effect level (NOAEL) in a 2-week in vivo toxicity study. Nevertheless, our results demonstrate that a prodrug approach using a nucleoside analog could potentially be developed for flavivirus antiviral therapy.


Asunto(s)
Adenosina/análogos & derivados , Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Dengue/tratamiento farmacológico , Profármacos/farmacología , Replicación Viral/efectos de los fármacos , Adenosina/farmacocinética , Adenosina/farmacología , Adenosina/uso terapéutico , Animales , Antivirales/química , Antivirales/farmacocinética , Antivirales/uso terapéutico , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Dengue/virología , Células Epiteliales/virología , Ésteres/química , Humanos , Ratones , Profármacos/química , Profármacos/farmacocinética , Profármacos/uso terapéutico , Ratas , Células Vero
9.
Antimicrob Agents Chemother ; 54(7): 2932-9, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20457821

RESUMEN

We recently reported that (2R,3R,4R,5R)-2-(4-amino-pyrrolo[2,3-d]pyrimidin-7-yl)-3-ethynyl-5-hydroxy-methyl-tetrahydro-furan-3,4-diol is a potent inhibitor of dengue virus (DENV), with 50% effective concentration (EC(50)) and cytotoxic concentration (CC(50)) values of 0.7 microM and >100 microM, respectively. Here we describe the synthesis, structure-activity relationship, and antiviral characterization of the inhibitor. In an AG129 mouse model, a single-dose treatment of DENV-infected mice with the compound suppressed peak viremia and completely prevented death. Mode-of-action analysis using a DENV replicon indicated that the compound blocks viral RNA synthesis. Recombinant adenosine kinase could convert the compound to a monophosphate form. Suppression of host adenosine kinase, using a specific inhibitor (iodotubercidin) or small interfering RNA (siRNA), abolished or reduced the compound's antiviral activity in cell culture. Studies of rats showed that (14)C-labeled compound was converted to mono-, di-, and triphosphate metabolites in vivo. Collectively, the results suggest that this adenosine inhibitor is phosphorylated to an active (triphosphate) form which functions as a chain terminator for viral RNA synthesis.


Asunto(s)
Adenosina/farmacología , Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Virus del Dengue/genética , ARN Viral/genética , Adenosina/química , Adenosina/metabolismo , Adenosina Quinasa/genética , Adenosina Quinasa/metabolismo , Animales , Antivirales/química , Antivirales/metabolismo , Línea Celular , Línea Celular Tumoral , Humanos , Masculino , Fosforilación , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño , Ratas , Ratas Wistar
10.
Antimicrob Agents Chemother ; 53(5): 1823-31, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19223625

RESUMEN

The incidence of dengue fever epidemics has increased dramatically over the last few decades. However, no vaccine or antiviral therapies are available. Therefore, the need for safe and effective antiviral drugs has become imperative. The entry of dengue virus into a host cell is mediated by its major envelope (E) protein. The crystal structure of the E protein reveals a hydrophobic pocket that is presumably important for low-pH-mediated membrane fusion. High-throughput docking with this hydrophobic pocket was performed, and hits were evaluated in cell-based assays. Compound 6 was identified as one of the inhibitors and had an average 50% effective concentration of 119 nM against dengue virus serotype 2 in a human cell line. Mechanism-of-action studies demonstrated that compound 6 acts at an early stage during dengue virus infection. It arrests dengue virus in vesicles that colocalize with endocytosed dextran and inhibits NS3 expression. The inhibitors described in this report can serve as molecular probes for the study of the entry of flavivirus into host cells.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/patogenicidad , Bibliotecas de Moléculas Pequeñas , Internalización del Virus/efectos de los fármacos , Animales , Antivirales/química , Sitios de Unión , Línea Celular , Cricetinae , Virus del Dengue/efectos de los fármacos , Virus del Dengue/crecimiento & desarrollo , Humanos , Modelos Moleculares , Relación Estructura-Actividad , Proteínas del Envoltorio Viral/antagonistas & inhibidores
11.
ACS Med Chem Lett ; 6(3): 344-8, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25878766

RESUMEN

Spiropyrazolopyridone 1 was identified, as a novel dengue virus (DENV) inhibitor, from a DENV serotype 2 (DENV-2) high-throughput phenotypic screen. As a general trend within this chemical class, chiral resolution of the racemate revealed that R enantiomer was significantly more potent than the S. Cell-based lead optimization of the spiropyrazolopyridones focusing on improving the physicochemical properties is described. As a result, an optimal compound 14a, with balanced in vitro potency and pharmacokinetic profile, achieved about 1.9 log viremia reduction at 3 × 50 mg/kg (bid) or 3 × 100 mg/kg (QD) oral doses in the dengue in vivo mouse efficacy model.

12.
Antiviral Res ; 91(1): 11-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21575658

RESUMEN

To facilitate dengue virus (DENV) drug discovery, we developed a stable luciferase reporter DENV-2. A renilla luciferase gene was engineered into the capsid-coding region of an infectious cDNA clone of DENV-2. Transfection of BHK-21 cells with the cDNA clone-derived RNA generated high titers (>10(6)PFU/ml) of luciferase reporter DENV-2. The reporter virus was infectious to a variety of cells, producing robust luciferase signals. Compared with wild-type virus, the reporter virus replicated slower in both mammalian Vero and mosquito C6/36 cells. To examine the stability of the reporter virus, we continuously passaged the virus on Vero cells for five rounds. All passaged viruses stably maintained the luciferase gene, demonstrating the stability of the reporter virus. Furthermore, we found that the passaged virus accumulated a mutation (T108M) in viral NS4B gene that could enhance viral RNA replication in a cell-type specific manner. Using the reporter virus, we developed a HTS assay in a 384-well format. The HTS assay was validated with known DENV inhibitors and showed a robust Z' factor of 0.79. The Luc-DENV-2 HTS assay allows screening for inhibitors of all steps of the viral life cycle. The reporter virus will also be a useful tool for studying DENV replication and pathogenesis.


Asunto(s)
Virus del Dengue/genética , Genes Reporteros , Ensayos Analíticos de Alto Rendimiento/métodos , Luciferasas/genética , Animales , Antivirales/farmacología , Cápside/química , Línea Celular , Chlorocebus aethiops , Cricetinae , Culicidae , Virus del Dengue/efectos de los fármacos , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Farmacorresistencia Viral , Ingeniería Genética , Mutación , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas no Estructurales Virales/genética , Replicación Viral/genética
13.
J Infect Dis ; 195(5): 665-74, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17262707

RESUMEN

Dengue fever is an emerging arboviral disease for which no vaccine or antiviral treatment exists and that causes thousands of fatalities each year. To develop an in vivo test system for antidengue drugs, AG129 mice, which are deficient for the interferon- alpha / beta and - gamma receptors, were injected with unadapted dengue virus, resulting in a dose-dependent transient viremia lasting several days and peaking on day 3 after infection. Additionally, nonstructural protein 1, increased levels of proinflammatory cytokines, and neutralizing IgM and IgG antibodies were found, and mice had splenomegaly. Oral administration of the antiviral compounds 7-deaza-2'-C-methyl-adenosine, N-nonyl-deoxynojirimycin, or 6-O-butanoyl castanospermine significantly reduced viremia in a dose-dependent manner, even after delayed treatment, leading to a reduction of splenomegaly and proinflammatory cytokine levels. The results validate this dengue viremia mouse model as a suitable system for testing antidengue drugs and indicate that antiviral treatment during the acute phase of dengue fever can reduce the severity of the disease.


Asunto(s)
Antivirales/uso terapéutico , Dengue/sangre , Dengue/tratamiento farmacológico , Modelos Animales de Enfermedad , Viremia , Replicación Viral/efectos de los fármacos , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacología , Animales , Antivirales/farmacología , Dengue/inmunología , Relación Dosis-Respuesta a Droga , Indolizinas/farmacología , Ratones , Ribavirina/farmacología , Ribavirina/uso terapéutico , Factores de Tiempo , Tubercidina/análogos & derivados , Tubercidina/uso terapéutico , Viremia/tratamiento farmacológico , Viremia/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA