Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Viruses ; 15(12)2023 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-38140576

RESUMEN

Marburg virus (MARV) causes severe disease and high mortality in humans. The objective of this study was to characterize disease manifestations and pathogenesis in cynomolgus macaques exposed to MARV. The results of this natural history study may be used to identify features of MARV disease useful in defining the ideal treatment initiation time for subsequent evaluations of investigational therapeutics using this model. Twelve cynomolgus macaques were exposed to a target dose of 1000 plaque-forming units MARV by the intramuscular route, and six control animals were mock-exposed. The primary endpoint of this study was survival to Day 28 post-inoculation (PI). Anesthesia events were minimized with the use of central venous catheters for periodic blood collection, and temperature and activity were continuously monitored by telemetry. All mock-exposed animals remained healthy for the duration of the study. All 12 MARV-exposed animals (100%) became infected, developed illness, and succumbed on Days 8-10 PI. On Day 4 PI, 11 of the 12 MARV-exposed animals had statistically significant temperature elevations over baseline. Clinically observable signs of MARV disease first appeared on Day 5 PI, when 6 of the 12 animals exhibited reduced responsiveness. Ultimately, systemic inflammation, coagulopathy, and direct cytopathic effects of MARV all contributed to multiorgan dysfunction, organ failure, and death or euthanasia of all MARV-exposed animals. Manifestations of MARV disease, including fever, systemic viremia, lymphocytolysis, coagulopathy, and hepatocellular damage, could be used as triggers for initiation of treatment in future therapeutic efficacy studies.


Asunto(s)
Enfermedad del Virus de Marburg , Marburgvirus , Humanos , Animales , Macaca fascicularis , Viremia , Hígado
2.
J Infect Dis ; 224(4): 632-642, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-33367826

RESUMEN

BACKGROUND: Ebola virus disease (EVD) supportive care strategies are largely guided by retrospective observational research. This study investigated the effect of EVD supportive care algorithms on duration of survival in a controlled nonhuman primate (NHP) model. METHODS: Fourteen rhesus macaques were challenged intramuscularly with a target dose of Ebola virus (1000 plaque-forming units; Kikwit). NHPs were allocated to intensive care unit (ICU)-like algorithms (n = 7), intravenous fluids plus levofloxacin (n = 2), or a control group (n = 5). The primary outcome measure was duration of survival, and secondary outcomes included changes in clinical laboratory values. RESULTS: Duration of survival was not significantly different between the pooled ICU-like algorithm and control groups (8.2 vs 6.9 days of survival; hazard ratio; 0.50; P = .25). Norepinephrine was effective in transiently maintaining baseline blood pressure. NHPs treated with ICU-like algorithms had delayed onset of liver and kidney injury. CONCLUSIONS: While an obvious survival difference was not observed with ICU-like care, clinical observations from this model may aid in EVD supportive care NHP model refinement.


Asunto(s)
Cuidados Críticos , Fiebre Hemorrágica Ebola , Unidades de Cuidados Intensivos , Animales , Modelos Animales de Enfermedad , Ebolavirus , Fiebre Hemorrágica Ebola/terapia , Macaca mulatta , Primates , Estudios Retrospectivos
3.
Emerg Infect Dis ; 23(8): 1316-1324, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28726603

RESUMEN

The Ebola virus (EBOV) outbreak in West Africa during 2013-2016 demonstrated the need to improve Ebola virus disease (EVD) diagnostics and standards of care. This retrospective study compared laboratory values and clinical features of 3 nonhuman primate models of lethal EVD to assess associations with improved survival time. In addition, the study identified laboratory values useful as predictors of survival, surrogates for EBOV viral loads, and triggers for initiation of therapeutic interventions in these nonhuman primate models. Furthermore, the data support that, in nonhuman primates, the Makona strain of EBOV may be less virulent than the Kikwit strain of EBOV. The applicability of these findings as potential diagnostic and management tools for EVD in humans warrants further investigation.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola/epidemiología , Fiebre Hemorrágica Ebola/virología , Animales , Biomarcadores , Fiebre Hemorrágica Ebola/mortalidad , Fiebre Hemorrágica Ebola/transmisión , Humanos , Estimación de Kaplan-Meier , Primates , ARN Viral , Curva ROC , Estudios Retrospectivos , Carga Viral
4.
Viruses ; 8(4): 94, 2016 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-27070636

RESUMEN

Marburg virus (MARV) was the first filovirus to be identified following an outbreak of viral hemorrhagic fever disease in Marburg, Germany in 1967. Due to several factors inherent to filoviruses, they are considered a potential bioweapon that could be disseminated via an aerosol route. Previous studies demonstrated that MARV virus-like particles (VLPs) containing the glycoprotein (GP), matrix protein VP40 and nucleoprotein (NP) generated using a baculovirus/insect cell expression system could protect macaques from subcutaneous (SQ) challenge with multiple species of marburgviruses. In the current study, the protective efficacy of the MARV VLPs in conjunction with two different adjuvants: QS-21, a saponin derivative, and poly I:C against homologous aerosol challenge was assessed in cynomolgus macaques. Antibody responses against the GP antigen were equivalent in all groups receiving MARV VLPs irrespective of the adjuvant; adjuvant only-vaccinated macaques did not demonstrate appreciable antibody responses. All macaques were subsequently challenged with lethal doses of MARV via aerosol or SQ as a positive control. All MARV VLP-vaccinated macaques survived either aerosol or SQ challenge while animals administered adjuvant only exhibited clinical signs and lesions consistent with MARV disease and were euthanized after meeting the predetermined criteria. Therefore, MARV VLPs induce IgG antibodies recognizing MARV GP and VP40 and protect cynomolgus macaques from an otherwise lethal aerosol exposure with MARV.


Asunto(s)
Enfermedad del Virus de Marburg/prevención & control , Marburgvirus/inmunología , Vacunación , Vacunas de Partículas Similares a Virus/inmunología , Animales , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Inmunoglobulina G/inmunología , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Macaca fascicularis , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/virología , Bazo/inmunología , Bazo/metabolismo , Bazo/patología , Factores de Tiempo , Resultado del Tratamiento , Vacunas de Partículas Similares a Virus/administración & dosificación , Viremia/virología
5.
PLoS One ; 10(3): e0118881, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25793502

RESUMEN

Filoviruses cause hemorrhagic fever resulting in significant morbidity and mortality in humans. Several vaccine platforms that include multiple virus-vectored approaches and virus-like particles (VLPs) have shown efficacy in nonhuman primates. Previous studies have shown protection of cynomolgus macaques against homologous infection for Ebola virus (EBOV) and Marburg virus (MARV) following a three-dose vaccine regimen of EBOV or MARV VLPs, as well as heterologous protection against Ravn Virus (RAVV) following vaccination with MARV VLPs. The objectives of the current studies were to determine the minimum number of vaccine doses required for protection (using EBOV as the test system) and then demonstrate protection against Sudan virus (SUDV) and Taï Forest virus (TAFV). Using the EBOV nonhuman primate model, we show that one or two doses of VLP vaccine can confer protection from lethal infection. VLPs containing the SUDV glycoprotein, nucleoprotein and VP40 matrix protein provide complete protection against lethal SUDV infection in macaques. Finally, we demonstrate protective efficacy mediated by EBOV, but not SUDV, VLPs against TAFV; this is the first demonstration of complete cross-filovirus protection using a single component heterologous vaccine within the Ebolavirus genus. Along with our previous results, this observation provides strong evidence that it will be possible to develop and administer a broad-spectrum VLP-based vaccine that will protect against multiple filoviruses by combining only three EBOV, SUDV and MARV components.


Asunto(s)
Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Virión/inmunología , Animales , Protección Cruzada/inmunología , Fiebre Hemorrágica Ebola/sangre , Fiebre Hemorrágica Ebola/virología , Inmunidad Humoral , Macaca/sangre , Macaca/inmunología , Macaca/virología , Vacunación , Vacunas Virales/inmunología
6.
Viral Immunol ; 28(1): 62-70, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25514232

RESUMEN

Filoviruses are causative agents of hemorrhagic fever, and to date no effective vaccine or therapeutic has been approved to combat infection. Filovirus glycoprotein (GP) is the critical immunogenic component of filovirus vaccines, eliciting high levels of antibody after successful vaccination. Previous work has shown that protection against both Ebola virus (EBOV) and Marburg virus (MARV) can be achieved by vaccinating with a mixture of virus-like particles (VLPs) expressing either EBOV GP or MARV GP. In this study, the potential for eliciting effective immune responses against EBOV, Sudan virus, and MARV with a single GP construct was tested. Trimeric hybrid GPs were produced that expressed the sequence of Marburg GP2 in conjunction with a hybrid GP1 composed EBOV and Sudan virus GP sequences. VLPs expressing these constructs, along with EBOV VP40, provided comparable protection against MARV challenge, resulting in 75 or 100% protection. Protection from EBOV challenge differed depending upon the hybrid used, however, with one conferring 75% protection and one conferring no protection. By comparing the overall antibody titers and the neutralizing antibody titers specific for each virus, it is shown that higher antibody responses were elicited by the C terminal region of GP1 than by the N terminal region, and this correlated with protection. These data collectively suggest that GP2 and the C terminal region of GP1 are highly immunogenic, and they advance progress toward the development of a pan-filovirus vaccine.


Asunto(s)
Protección Cruzada , Ebolavirus/inmunología , Marburgvirus/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Antígenos Virales/inmunología , Ebolavirus/genética , Femenino , Cobayas , Fiebre Hemorrágica Ebola/prevención & control , Enfermedad del Virus de Marburg/prevención & control , Marburgvirus/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Análisis de Supervivencia , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/genética , Proteínas del Envoltorio Viral/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Virosomas/genética , Virosomas/inmunología
7.
Viruses ; 6(11): 4666-82, 2014 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-25421892

RESUMEN

Multiple products are being developed for use against filoviral infections. Efficacy for these products will likely be demonstrated in nonhuman primate models of filoviral disease to satisfy licensure requirements under the Animal Rule, or to supplement human data. Typically, the endpoint for efficacy assessment will be survival following challenge; however, there exists no standardized approach for assessing the health or euthanasia criteria for filovirus-exposed nonhuman primates. Consideration of objective criteria is important to (a) ensure test subjects are euthanized without unnecessary distress; (b) enhance the likelihood that animals exhibiting mild or moderate signs of disease are not prematurely euthanized; (c) minimize the occurrence of spontaneous deaths and loss of end-stage samples; (d) enhance the reproducibility of experiments between different researchers; and (e) provide a defensible rationale for euthanasia decisions that withstands regulatory scrutiny. Historic records were compiled for 58 surviving and non-surviving monkeys exposed to Ebola virus at the US Army Medical Research Institute of Infectious Diseases. Clinical pathology parameters were statistically analyzed and those exhibiting predicative value for survival are reported. These findings may be useful for standardization of objective euthanasia assessments in rhesus monkeys exposed to Ebola virus and may serve as a useful approach for other standardization efforts.


Asunto(s)
Eutanasia Animal , Haplorrinos , Fiebre Hemorrágica Ebola/patología , Enfermedades de los Primates/patología , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Fiebre Hemorrágica Ebola/terapia , Enfermedades de los Primates/terapia , Análisis de Supervivencia
8.
PLoS One ; 9(2): e89735, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586996

RESUMEN

Identifying safe and effective adjuvants is critical for the advanced development of protein-based vaccines. Pattern recognition receptor (PRR) agonists are increasingly being explored as potential adjuvants, but there is concern that the efficacy of these molecules may be dependent on potentially dangerous levels of non-specific immune activation. The filovirus virus-like particle (VLP) vaccine protects mice, guinea pigs, and nonhuman primates from viral challenge. In this study, we explored the impact of a stabilized dsRNA mimic, polyICLC, on VLP vaccination of C57BL/6 mice and Hartley guinea pigs. We show that at dose levels as low as 100 ng, the adjuvant increased the efficacy of the vaccine in mice. Antigen-specific, polyfunctional CD4 and CD8 T cell responses and antibody responses increased significantly upon inclusion of adjuvant. To determine whether the efficacy of polyICLC correlated with systemic immune activation, we examined serum cytokine levels and cellular activation in the draining lymph node. PolyICLC administration was associated with increases in TNFα, IL6, MCP1, MIP1α, KC, and MIP1ß levels in the periphery and with the activation of dendritic cells (DCs), NK cells, and B cells. However, this activation resolved within 24 to 72 hours at efficacious adjuvant dose levels. These studies are the first to examine the polyICLC-induced enhancement of antigen-specific immune responses in the context of non-specific immune activation, and they provide a framework from which to consider adjuvant dose levels.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Carboximetilcelulosa de Sodio/análogos & derivados , Poli I-C/farmacología , Polilisina/análogos & derivados , Receptores Toll-Like/agonistas , Vacunas de Partículas Similares a Virus/farmacología , Animales , Carboximetilcelulosa de Sodio/farmacología , Citocinas/sangre , Ebolavirus/efectos de los fármacos , Ebolavirus/inmunología , Cobayas , Fiebre Hemorrágica Ebola/prevención & control , Ratones Endogámicos C57BL , Polilisina/farmacología
9.
J Virol ; 88(2): 811-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24198421

RESUMEN

Sin Nombre virus (SNV) is a rodent-borne hantavirus that causes hantavirus pulmonary syndrome (HPS) predominantly in North America. SNV infection of immunocompetent hamsters results in an asymptomatic infection; the only lethal disease model for a pathogenic hantavirus is Andes virus (ANDV) infection of Syrian hamsters. Efforts to create a lethal SNV disease model in hamsters by repeatedly passaging virus through the hamster have demonstrated increased dissemination of the virus but no signs of disease. In this study, we demonstrate that immunosuppression of hamsters through the administration of a combination of dexamethasone and cyclophosphamide, followed by infection with SNV, results in a vascular leak syndrome that accurately mimics both HPS disease in humans and ANDV infection of hamsters. Immunosuppressed hamsters infected with SNV have a mean number of days to death of 13 and display clinical signs associated with HPS, including pulmonary edema. Viral antigen was widely detectable throughout the pulmonary endothelium. Histologic analysis of lung sections showed marked inflammation and edema within the alveolar septa of SNV-infected hamsters, results which are similar to what is exhibited by hamsters infected with ANDV. Importantly, SNV-specific neutralizing polyclonal antibody administered 5 days after SNV infection conferred significant protection against disease. This experiment not only demonstrated that the disease was caused by SNV, it also demonstrated the utility of this animal model for testing candidate medical countermeasures. This is the first report of lethal disease caused by SNV in an adult small-animal model.


Asunto(s)
Modelos Animales de Enfermedad , Síndrome Pulmonar por Hantavirus/inmunología , Síndrome Pulmonar por Hantavirus/virología , Mesocricetus , Virus Sin Nombre/fisiología , Animales , Anticuerpos Antivirales/uso terapéutico , Cricetinae , Ciclofosfamida/administración & dosificación , Dexametasona/administración & dosificación , Femenino , Síndrome Pulmonar por Hantavirus/tratamiento farmacológico , Síndrome Pulmonar por Hantavirus/mortalidad , Síndrome Pulmonar por Hantavirus/patología , Humanos , Terapia de Inmunosupresión , Inmunosupresores/administración & dosificación
10.
PLoS One ; 8(4): e60579, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23577127

RESUMEN

BACKGROUND: The rapid development of effective medical countermeasures against potential biological threat agents is vital. Repurposing existing drugs that may have unanticipated activities as potential countermeasures is one way to meet this important goal, since currently approved drugs already have well-established safety and pharmacokinetic profiles in patients, as well as manufacturing and distribution networks. Therefore, approved drugs could rapidly be made available for a new indication in an emergency. METHODOLOGY/PRINCIPAL FINDINGS: A large systematic effort to determine whether existing drugs can be used against high containment bacterial and viral pathogens is described. We assembled and screened 1012 FDA-approved drugs for off-label broad-spectrum efficacy against Bacillus anthracis; Francisella tularensis; Coxiella burnetii; and Ebola, Marburg, and Lassa fever viruses using in vitro cell culture assays. We found a variety of hits against two or more of these biological threat pathogens, which were validated in secondary assays. As expected, antibiotic compounds were highly active against bacterial agents, but we did not identify any non-antibiotic compounds with broad-spectrum antibacterial activity. Lomefloxacin and erythromycin were found to be the most potent compounds in vivo protecting mice against Bacillus anthracis challenge. While multiple virus-specific inhibitors were identified, the most noteworthy antiviral compound identified was chloroquine, which disrupted entry and replication of two or more viruses in vitro and protected mice against Ebola virus challenge in vivo. CONCLUSIONS/SIGNIFICANCE: The feasibility of repurposing existing drugs to face novel threats is demonstrated and this represents the first effort to apply this approach to high containment bacteria and viruses.


Asunto(s)
Antibacterianos/farmacología , Antivirales/farmacología , Armas Biológicas , Aprobación de Drogas , Evaluación Preclínica de Medicamentos/métodos , United States Food and Drug Administration , Animales , Línea Celular , Femenino , Humanos , Masculino , Ratones , Estados Unidos
11.
Viruses ; 4(11): 2806-30, 2012 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-23202506

RESUMEN

There are no currently approved treatments for filovirus infections. In this study we report the discovery process which led to the development of antisense Phosphorodiamidate Morpholino Oligomers (PMOs) AVI-6002 (composed of AVI-7357 and AVI-7539) and AVI-6003 (composed of AVI-7287 and AVI-7288) targeting Ebola virus and Marburg virus respectively. The discovery process involved identification of optimal transcript binding sites for PMO based RNA-therapeutics followed by screening for effective viral gene target in mouse and guinea pig models utilizing adapted viral isolates. An evolution of chemical modifications were tested, beginning with simple Phosphorodiamidate Morpholino Oligomers (PMO) transitioning to cell penetrating peptide conjugated PMOs (PPMO) and ending with PMOplus containing a limited number of positively charged linkages in the PMO structure. The initial lead compounds were combinations of two agents targeting separate genes. In the final analysis, a single agent for treatment of each virus was selected, AVI-7537 targeting the VP24 gene of Ebola virus and AVI-7288 targeting NP of Marburg virus, and are now progressing into late stage clinical development as the optimal therapeutic candidates.


Asunto(s)
Antivirales/administración & dosificación , Ebolavirus/genética , Fiebre Hemorrágica Ebola/terapia , Enfermedad del Virus de Marburg/terapia , Marburgvirus/genética , Morfolinos/administración & dosificación , Oligodesoxirribonucleótidos Antisentido/administración & dosificación , Animales , Antivirales/química , Secuencia de Bases , Ebolavirus/metabolismo , Genes Virales , Cobayas , Fiebre Hemorrágica Ebola/mortalidad , Fiebre Hemorrágica Ebola/virología , Enfermedad del Virus de Marburg/mortalidad , Enfermedad del Virus de Marburg/virología , Marburgvirus/metabolismo , Ratones , Morfolinos/química , Oligodesoxirribonucleótidos Antisentido/química , Primates , Biosíntesis de Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo
12.
J Virol ; 85(8): 3905-17, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21289122

RESUMEN

Marburgvirus (MARV) infections are generally lethal in humans and nonhuman primates but require in vivo lethal mouse variant selection by the serial transfer (passage) of the nonlethal virus into naïve mice to propagate a lethal infection. The passage of progenitor (wild-type) MARV or Ravn virus (RAVV) from infected scid BALB/c mouse liver homogenates into immunocompetent BALB/c mice results in the selection of lethal mouse viruses from within the quasispecies sufficient to establish lethality in immunocompetent mice. Genomic analysis in conjunction with the passage history of each mutation detailed the altered primary and secondary structures of the viral genomic RNA throughout the process. Key findings included the following: (i) a VP40:D184N mutation previously identified in the lethal guinea pig MARV genome was the first mutation to occur during the passage of both the MARV and RAVV variants; (ii) there was biased hypermutagenesis in the RAVV variant genome; (iii) there were two identical mutations in lethal mouse MARV and RAVV variants, VP40:Y19H in the PPPY motif and VP40:D184N in a loop structure between the two VP40 domains; (iv) the passage of wild-type MARV and RAVV in mice resulted in the selection of viral variants from among the quasispecies with different genotypes than those of the wild-type viruses; and (v) a lethal mouse RAVV variant had different tissue tropisms distinct from those of its wild-type virus. These studies provide insights into how marburgviruses manipulate the host for enzymes, metabolites, translation regulators, and effectors of the innate immune response to serve as potential viral countermeasures.


Asunto(s)
Evolución Molecular , Genoma Viral , Enfermedad del Virus de Marburg/virología , Marburgvirus/genética , Marburgvirus/patogenicidad , Enfermedades de los Roedores/virología , Selección Genética , Sustitución de Aminoácidos/genética , Animales , Análisis Mutacional de ADN , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Mutación Missense , ARN Viral/genética , Enfermedades de los Roedores/mortalidad , Pase Seriado , Análisis de Supervivencia , Tropismo Viral
13.
Antiviral Res ; 66(1): 35-8, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15781130

RESUMEN

Serum from wild alligators was collected and tested for antibiotic activity against three enveloped viruses using cell-based assays. Alligator serum demonstrated antiviral activities against human immunodeficiency virus type 1 (HIV-1; IC50=0.9%), West Nile virus (WNV; IC50=4.3%), and Herpes simplex virus type 1 (HSV-1; IC50=3.4%). The inhibitory concentration (IC50) is defined as the concentration of serum that inhibits 50% of viral activity. The antiviral effects of the alligator serum were difficult to evaluate at high concentrations due to the inherent toxicity to the mammalian cells used to assay viral activities. The TC50 (serum concentration that reduces cell viability to 50%) values for the serum in the HIV-1, WNV, and HSV-1 assays were 32.8, 36.3 and 39.1%, respectively. Heat-treated serum (56 degrees C, 30 min) displayed IC50 values of >50, 9.8 and 14.9% for HIV-1, WNV and HSV-1 viruses, respectively. In addition, the TC50 values using heat-treated serum were substantially elevated for all three assays, relative to untreated serum (47.3 to >50%). Alligator serum complement activity has been shown to be heat labile under these conditions. HIV-1 antiviral action was heat-sensitive, and thus possibly due to the action of serum complement, while the anti-WNV and anti-HSV-1 activities were not heat labile and thus probably not complement mediated.


Asunto(s)
Caimanes y Cocodrilos/sangre , Antivirales/farmacología , Virus ARN/efectos de los fármacos , Suero/inmunología , Caimanes y Cocodrilos/inmunología , Animales , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Suero/química , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...