Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(6): 995-1007.e18, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35303429

RESUMEN

Several ebolaviruses cause outbreaks of severe disease. Vaccines and monoclonal antibody cocktails are available to treat Ebola virus (EBOV) infections, but not Sudan virus (SUDV) or other ebolaviruses. Current cocktails contain antibodies that cross-react with the secreted soluble glycoprotein (sGP) that absorbs virus-neutralizing antibodies. By sorting memory B cells from EBOV infection survivors, we isolated two broadly reactive anti-GP monoclonal antibodies, 1C3 and 1C11, that potently neutralize, protect rodents from disease, and lack sGP cross-reactivity. Both antibodies recognize quaternary epitopes in trimeric ebolavirus GP. 1C11 bridges adjacent protomers via the fusion loop. 1C3 has a tripartite epitope in the center of the trimer apex. One 1C3 antigen-binding fragment anchors simultaneously to the three receptor-binding sites in the GP trimer, and separate 1C3 paratope regions interact differently with identical residues on the three protomers. A cocktail of both antibodies completely protected nonhuman primates from EBOV and SUDV infections, indicating their potential clinical value.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Ebolavirus , Fiebre Hemorrágica Ebola , Animales , Epítopos , Glicoproteínas/química , Subunidades de Proteína
2.
Cell ; 184(22): 5593-5607.e18, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34715022

RESUMEN

Ebolaviruses cause a severe and often fatal illness with the potential for global spread. Monoclonal antibody-based treatments that have become available recently have a narrow therapeutic spectrum and are ineffective against ebolaviruses other than Ebola virus (EBOV), including medically important Bundibugyo (BDBV) and Sudan (SUDV) viruses. Here, we report the development of a therapeutic cocktail comprising two broadly neutralizing human antibodies, rEBOV-515 and rEBOV-442, that recognize non-overlapping sites on the ebolavirus glycoprotein (GP). Antibodies in the cocktail exhibited synergistic neutralizing activity, resisted viral escape, and possessed differing requirements for their Fc-regions for optimal in vivo activities. The cocktail protected non-human primates from ebolavirus disease caused by EBOV, BDBV, or SUDV with high therapeutic effectiveness. High-resolution structures of the cocktail antibodies in complex with GP revealed the molecular determinants for neutralization breadth and potency. This study provides advanced preclinical data to support clinical development of this cocktail for pan-ebolavirus therapy.


Asunto(s)
Anticuerpos Antivirales/inmunología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Sitios de Unión , Línea Celular , Microscopía por Crioelectrón , Ebolavirus/ultraestructura , Epítopos/inmunología , Femenino , Glicoproteínas/química , Glicoproteínas/inmunología , Fiebre Hemorrágica Ebola/virología , Humanos , Concentración de Iones de Hidrógeno , Ratones Endogámicos BALB C , Modelos Moleculares , Primates , Receptores Fc/metabolismo , Proteínas Recombinantes/inmunología , Viremia/inmunología
3.
Nat Immunol ; 22(1): 86-98, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33235385

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for an unprecedented global pandemic of COVID-19. Animal models are urgently needed to study the pathogenesis of COVID-19 and to screen vaccines and treatments. We show that African green monkeys (AGMs) support robust SARS-CoV-2 replication and develop pronounced respiratory disease, which may more accurately reflect human COVID-19 cases than other nonhuman primate species. SARS-CoV-2 was detected in mucosal samples, including rectal swabs, as late as 15 days after exposure. Marked inflammation and coagulopathy in blood and tissues were prominent features. Transcriptome analysis demonstrated stimulation of interferon and interleukin-6 pathways in bronchoalveolar lavage samples and repression of natural killer cell- and T cell-associated transcripts in peripheral blood. Despite a slight waning in antibody titers after primary challenge, enhanced antibody and cellular responses contributed to rapid clearance after re-challenge with an identical strain. These data support the utility of AGM for studying COVID-19 pathogenesis and testing medical countermeasures.


Asunto(s)
COVID-19/inmunología , Modelos Animales de Enfermedad , Reinfección/inmunología , SARS-CoV-2/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Antivirales/inmunología , COVID-19/epidemiología , COVID-19/virología , Chlorocebus aethiops , Epidemias/prevención & control , Expresión Génica/genética , Expresión Génica/inmunología , Perfilación de la Expresión Génica , Humanos , Interferones/genética , Interferones/inmunología , Interferones/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Reinfección/virología , SARS-CoV-2/fisiología , Linfocitos T/metabolismo , Linfocitos T/virología
4.
Proc Natl Acad Sci U S A ; 120(34): e2304876120, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37590417

RESUMEN

There are no approved treatments for Lassa fever (LF), which is responsible for thousands of deaths each year in West Africa. A major challenge in developing effective medical countermeasures against LF is the high diversity of circulating Lassa virus (LASV) strains with four recognized lineages and four proposed lineages. The recent resurgence of LASV in Nigeria caused by genetically distinct strains underscores this concern. Two LASV lineages (II and III) are dominant in Nigeria. Here, we show that combinations of two or three pan-lineage neutralizing human monoclonal antibodies (8.9F, 12.1F, 37.D) known as Arevirumab-2 or Arevirumab-3 can protect up to 100% of cynomolgus macaques against challenge with both lineage II and III LASV isolates when treatment is initiated at advanced stages of disease on day 8 after LASV exposure. This work demonstrates that it may be possible to develop postexposure interventions that can broadly protect against most strains of LASV.


Asunto(s)
Fiebre de Lassa , Virus Lassa , Animales , Humanos , Fiebre de Lassa/prevención & control , África Occidental , Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Macaca fascicularis
5.
Proc Natl Acad Sci U S A ; 119(12): e2200065119, 2022 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-35286211

RESUMEN

SignificanceConcern has increased about the pandemic potential of Nipah virus (NiV). Similar to SARS-CoV-2, NiV is an RNA virus that is transmitted by respiratory droplets. There are currently no NiV vaccines licensed for human use. While several preventive vaccines have shown promise in protecting animals against lethal NiV disease, most studies have assessed protection 1 mo after vaccination. However, in order to contain and control outbreaks, vaccines that can rapidly confer protection in days rather than months are needed. Here, we show that a recombinant vesicular stomatitis virus vector expressing the NiV glycoprotein can completely protect monkeys vaccinated 7 d prior to NiV exposure and 67% of animals vaccinated 3 d before NiV challenge.


Asunto(s)
Infecciones por Henipavirus/veterinaria , Virus Nipah/inmunología , Enfermedades de los Primates/prevención & control , Vacunas Sintéticas/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales/inmunología , Biomarcadores , Vectores Genéticos , Estimación de Kaplan-Meier , Pruebas de Neutralización , Evaluación de Resultado en la Atención de Salud , Enfermedades de los Primates/diagnóstico , Enfermedades de los Primates/mortalidad , Enfermedades de los Primates/virología , Vacunación , Carga Viral
6.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33836604

RESUMEN

The COVID-19 pandemic has reemphasized the need to identify safe and scalable therapeutics to slow or reverse symptoms of disease caused by newly emerging and reemerging viral pathogens. Recent clinical successes of monoclonal antibodies (mAbs) in therapy for viral infections demonstrate that mAbs offer a solution for these emerging biothreats. We have explored this with respect to Junin virus (JUNV), an arenavirus classified as a category A high-priority agent and the causative agent of Argentine hemorrhagic fever (AHF). There are currently no Food and Drug Administration-approved drugs available for preventing or treating AHF, although immune plasma from convalescent patients is used routinely to treat active infections. However, immune plasma is severely limited in quantity, highly variable in quality, and poses significant safety risks including the transmission of transfusion-borne diseases. mAbs offer a highly specific and consistently potent alternative to immune plasma that can be manufactured at large scale. We previously described a chimeric mAb, cJ199, that provided protection in a guinea pig model of AHF. To adapt this mAb to a format more suitable for clinical use, we humanized the mAb (hu199) and evaluated it in a cynomolgus monkey model of AHF with two JUNV isolates, Romero and Espindola. While untreated control animals experienced 100% lethality, all animals treated with hu199 at 6 d postinoculation (dpi) survived, and 50% of animals treated at 8 dpi survived. mAbs like hu199 may offer a safer, scalable, and more reproducible alternative to immune plasma for rare viral diseases that have epidemic potential.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Antivirales/farmacología , Fiebre Hemorrágica Americana/prevención & control , Virus Junin/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Cobayas , Fiebre Hemorrágica Americana/sangre , Humanos , Macaca fascicularis
7.
J Infect Dis ; 228(Suppl 7): S559-S570, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37610176

RESUMEN

BACKGROUND: Marburg virus (MARV) has caused numerous sporadic outbreaks of severe hemorrhagic fever in humans. Human case fatality rates of Marburg virus disease (MVD) outbreaks range from 20% to 90%. Viral genotypes of MARV can differ by over 20%, suggesting variable virulence between lineages may accompany this genetic divergence. Comparison of existing animal models of MVD employing different strains of MARV support differences in virulence across MARV genetic lineages; however, there are few systematic comparisons in models that recapitulate human disease available. METHODS: We compared features of disease pathogenesis in uniformly lethal hamster models of MVD made possible through serial adaptation in rodents. RESULTS: No further adaptation from a previously reported guinea pig-adapted (GPA) isolate of MARV-Angola was necessary to achieve uniform lethality in hamsters. Three passages of GPA MARV-Ci67 resulted in uniform lethality, where 4 passages of a GPA Ravn virus was 75% lethal. Hamster-adapted MARV-Ci67 demonstrated delayed time to death, protracted weight loss, lower viral burden, and slower histologic alteration compared to GPA MARV-Angola. CONCLUSIONS: These data suggest isolate-dependent virulence differences are maintained even after serial adaptation in rodents and may serve to guide choice of variant and model used for development of vaccines or therapeutics for MVD.


Asunto(s)
Enfermedad del Virus de Marburg , Marburgvirus , Cricetinae , Humanos , Cobayas , Animales , Mesocricetus , Virulencia , Angola
8.
J Infect Dis ; 228(Suppl 7): S712-S720, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37290053

RESUMEN

BACKGROUND: The filovirus Bundibugyo virus (BDBV) causes severe disease with a mortality rate of approximately 20%-51%. The only licensed filovirus vaccine in the United States, Ervebo, consists of a recombinant vesicular stomatitis virus (rVSV) vector that expresses Ebola virus (EBOV) glycoprotein (GP). Ervebo was shown to rapidly protect against fatal Ebola disease in clinical trials; however, the vaccine is only indicated against EBOV. Recent outbreaks of other filoviruses underscore the need for additional vaccine candidates, particularly for BDBV infections. METHODS: To examine whether the rVSV vaccine candidate rVSVΔG/BDBV-GP could provide therapeutic protection against BDBV, we inoculated seven cynomolgus macaques with 1000 plaque-forming units of BDBV, administering rVSVΔG/BDBV-GP vaccine to 6 of them 20-23 minutes after infection. RESULTS: Five of the treated animals survived infection (83%) compared to an expected natural survival rate of 21% in this macaque model. All treated animals showed an early circulating immune response, while the untreated animal did not. Surviving animals showed evidence of both GP-specific IgM and IgG production, while animals that succumbed did not produce significant IgG. CONCLUSIONS: This small, proof-of-concept study demonstrated early treatment with rVSVΔG/BDBV-GP provides a survival benefit in this nonhuman primate model of BDBV infection, perhaps through earlier initiation of adaptive immunity.


Asunto(s)
Vacunas contra el Virus del Ébola , Ebolavirus , Fiebre Hemorrágica Ebola , Estomatitis Vesicular , Vacunas Virales , Animales , Estomatitis Vesicular/prevención & control , Anticuerpos Antivirales , Vesiculovirus/genética , Glicoproteínas/genética , Macaca fascicularis , Inmunoglobulina G
9.
J Infect Dis ; 228(Suppl 7): S571-S581, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37348509

RESUMEN

BACKGROUND: The primary route of infection by Ebola virus (EBOV) is through contact of mucosal surfaces. Few studies have explored infection of nonhuman primates (NHPs) via the oral mucosa, which is a probable portal of natural infection in humans. METHODS: To further characterize the pathogenesis of EBOV infection via the oral exposure route, we challenged cohorts of cynomolgus monkeys with low doses of EBOV variant Makona. RESULTS: Infection with 100 or 50 PFU of EBOV Makona via the oral route resulted in 50% and 83% lethality, respectively. Animals that progressed to fatal disease exhibited lymphopenia, marked coagulopathy, high viral loads, and increased levels of serum markers of inflammation and hepatic/renal injury. Survival in these cohorts was associated with milder fluctuations in leukocyte populations, lack of coagulopathy, and reduced or absent serum markers of inflammation and/or hepatic/renal function. Surprisingly, 2 surviving animals from the 100- and 50-PFU cohorts developed transient low-level viremia in the absence of other clinical signs of disease. Conversely, all animals in the 10 PFU cohort remained disease free and survived to the study end point. CONCLUSIONS: Our observations highlight the susceptibility of NHPs, and by extension, likely humans, to relatively low doses of EBOV via the oral route.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Humanos , Animales , Modelos Animales de Enfermedad , Viremia , Macaca fascicularis , Biomarcadores
10.
J Infect Dis ; 228(Suppl 7): S604-S616, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37145930

RESUMEN

BACKGROUND: Highly pathogenic filoviruses such as Ebola virus (EBOV) hold capacity for delivery by artificial aerosols, and thus potential for intentional misuse. Previous studies have shown that high doses of EBOV delivered by small-particle aerosol cause uniform lethality in nonhuman primates (NHPs), whereas only a few small studies have assessed lower doses in NHPs. METHODS: To further characterize the pathogenesis of EBOV infection via small-particle aerosol, we challenged cohorts of cynomolgus monkeys with low doses of EBOV variant Makona, which may help define risks associated with small particle aerosol exposures. RESULTS: Despite using challenge doses orders of magnitude lower than previous studies, infection via this route was uniformly lethal across all cohorts. Time to death was delayed in a dose-dependent manner between aerosol-challenged cohorts, as well as in comparison to animals challenged via the intramuscular route. Here, we describe the observed clinical and pathological details including serum biomarkers, viral burden, and histopathological changes leading to death. CONCLUSIONS: Our observations in this model highlight the striking susceptibility of NHPs, and likely humans, via small-particle aerosol exposure to EBOV and emphasize the need for further development of diagnostics and postexposure prophylactics in the event of intentional release via deployment of an aerosol-producing device.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Humanos , Animales , Macaca fascicularis , Aerosoles , Carga Viral
11.
Nature ; 520(7549): 688-691, 2015 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-25853476

RESUMEN

The family Filoviridae contains three genera, Ebolavirus (EBOV), Marburg virus, and Cuevavirus. Some members of the EBOV genus, including Zaire ebolavirus (ZEBOV), can cause lethal haemorrhagic fever in humans. During 2014 an unprecedented ZEBOV outbreak occurred in West Africa and is still ongoing, resulting in over 10,000 deaths, and causing global concern of uncontrolled disease. To meet this challenge a rapid-acting vaccine is needed. Many vaccine approaches have shown promise in being able to protect nonhuman primates against ZEBOV. In response to the current ZEBOV outbreak several of these vaccines have been fast tracked for human use. However, it is not known whether any of these vaccines can provide protection against the new outbreak Makona strain of ZEBOV. One of these approaches is a first-generation recombinant vesicular stomatitis virus (rVSV)-based vaccine expressing the ZEBOV glycoprotein (GP) (rVSV/ZEBOV). To address safety concerns associated with this vector, we developed two candidate, further-attenuated rVSV/ZEBOV vaccines. Both attenuated vaccines produced an approximately tenfold lower vaccine-associated viraemia compared to the first-generation vaccine and both provided complete, single-dose protection of macaques from lethal challenge with the Makona outbreak strain of ZEBOV.


Asunto(s)
Vacunas contra el Virus del Ébola/administración & dosificación , Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Fiebre Hemorrágica Ebola/virología , Vacunas Atenuadas/inmunología , Vesiculovirus/genética , África Occidental/epidemiología , Animales , Anticuerpos Antivirales/inmunología , República Democrática del Congo/epidemiología , Vacunas contra el Virus del Ébola/genética , Ebolavirus/clasificación , Femenino , Vectores Genéticos/genética , Fiebre Hemorrágica Ebola/inmunología , Humanos , Inmunoglobulina G/inmunología , Cinética , Macaca fascicularis , Masculino , Análisis de Supervivencia , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vesiculovirus/crecimiento & desarrollo
12.
Nature ; 521(7552): 362-5, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-25901685

RESUMEN

The current outbreak of Ebola virus in West Africa is unprecedented, causing more cases and fatalities than all previous outbreaks combined, and has yet to be controlled. Several post-exposure interventions have been employed under compassionate use to treat patients repatriated to Europe and the United States. However, the in vivo efficacy of these interventions against the new outbreak strain of Ebola virus is unknown. Here we show that lipid-nanoparticle-encapsulated short interfering RNAs (siRNAs) rapidly adapted to target the Makona outbreak strain of Ebola virus are able to protect 100% of rhesus monkeys against lethal challenge when treatment was initiated at 3 days after exposure while animals were viraemic and clinically ill. Although all infected animals showed evidence of advanced disease including abnormal haematology, blood chemistry and coagulopathy, siRNA-treated animals had milder clinical features and fully recovered, while the untreated control animals succumbed to the disease. These results represent the first, to our knowledge, successful demonstration of therapeutic anti-Ebola virus efficacy against the new outbreak strain in nonhuman primates and highlight the rapid development of lipid-nanoparticle-delivered siRNA as a countermeasure against this highly lethal human disease.


Asunto(s)
Ebolavirus/efectos de los fármacos , Ebolavirus/genética , Fiebre Hemorrágica Ebola/terapia , Fiebre Hemorrágica Ebola/virología , Nanopartículas/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/uso terapéutico , Animales , Secuencia de Bases , Modelos Animales de Enfermedad , Ebolavirus/clasificación , Femenino , Fiebre Hemorrágica Ebola/patología , Fiebre Hemorrágica Ebola/prevención & control , Humanos , Macaca mulatta/virología , Masculino , ARN Interferente Pequeño/farmacología , Análisis de Supervivencia , Factores de Tiempo , Resultado del Tratamiento , Carga Viral/efectos de los fármacos
13.
J Infect Dis ; 221(Suppl 4): S431-S435, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-31665351

RESUMEN

The high case-fatality rates and potential for use as a biological weapon make Nipah virus (NiV) a significant public health concern. Previous studies assessing the pathogenic potential of NiV delivered by the aerosol route in African green monkeys (AGMs) used the Malaysia strain (NiVM), which has caused lower instances of respiratory illness and person-to-person transmission during human outbreaks than the Bangladesh strain (NiVB). Accordingly, we developed a small particle aerosol model of NiVB infection in AGMs. Consistent with other mucosal AGM models of NiVB infection, we achieved uniform lethality and disease pathogenesis reflective of that observed in humans.


Asunto(s)
Infecciones por Henipavirus/virología , Virus Nipah/clasificación , Virus Nipah/fisiología , Aerosoles , Animales , Infecciones por Henipavirus/patología
14.
J Infect Dis ; 221(Suppl 4): S414-S418, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-31665362

RESUMEN

Due to the difficulty in conducting clinical trials for vaccines and treatments against Nipah virus (NiV), licensure will likely require animal models, most importantly non-human primates (NHPs). The NHP models of infection have primarily relied on intratracheal instillation or small particle aerosolization of NiV. However, neither of these routes adequately models natural mucosal exposure to NiV. To develop a more natural NHP model, we challenged African green monkeys with the Bangladesh strain of NiV by the intranasal route using the laryngeal mask airway (LMA) mucosal atomization device (MAD). LMA MAD exposure resulted in uniformly lethal disease that accurately reflected the human condition.


Asunto(s)
Chlorocebus aethiops , Modelos Animales de Enfermedad , Infecciones por Henipavirus/virología , Virus Nipah , Administración Intranasal , Aerosoles , Animales , Femenino , Infecciones por Henipavirus/mortalidad , Masculino , Carga Viral , Tropismo Viral
15.
J Infect Dis ; 221(Suppl 4): S436-S447, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32022850

RESUMEN

BACKGROUND: The henipaviruses, Hendra virus (HeV) and Nipah virus (NiV), are capable of causing severe and often lethal respiratory and/or neurologic disease in animals and humans. Given the sporadic nature of henipavirus outbreaks, licensure of vaccines and therapeutics for human use will likely require demonstration of efficacy in animal models that faithfully reproduce the human condition. Currently, the African green monkey (AGM) best mimics human henipavirus-induced disease. METHODS: The pathogenic potential of HeV and both strains of NiV (Malaysia, Bangladesh) was assessed in cynomolgus monkeys and compared with henipavirus-infected historical control AGMs. Multiplex gene and protein expression assays were used to compare host responses. RESULTS: In contrast to AGMs, in which henipaviruses cause severe and usually lethal disease, HeV and NiVs caused only mild or asymptomatic infections in macaques. All henipaviruses replicated in macaques with similar kinetics as in AGMs. Infection in macaques was associated with activation and predicted recruitment of cytotoxic CD8+ T cells, Th1 cells, IgM+ B cells, and plasma cells. Conversely, fatal outcome in AGMs was associated with aberrant innate immune signaling, complement dysregulation, Th2 skewing, and increased secretion of MCP-1. CONCLUSION: The restriction factors identified in macaques can be harnessed for development of effective countermeasures against henipavirus disease.


Asunto(s)
Virus Hendra , Infecciones por Henipavirus/veterinaria , Inmunidad Celular , Inmunidad Humoral , Macaca fascicularis , Virus Nipah , Animales , Infecciones por Henipavirus/virología , Masculino , Enfermedades de los Monos/inmunología , Enfermedades de los Monos/virología , Carga Viral , Tropismo Viral
16.
Virol J ; 17(1): 125, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32811514

RESUMEN

We recently reported the development of the first African green monkey (AGM) model for COVID-19 based on a combined liquid intranasal (i.n.) and intratracheal (i.t.) exposure to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here, we followed up on this work by assessing an i.n. particle only route of exposure using the LMA mucosal atomization device (MAD). Six AGMs were infected with SARS-CoV-2; three animals were euthanized near the peak stage of virus replication (day 5) and three animals were euthanized during the early convalescence period (day 34). All six AGMs supported robust SARS-CoV-2 replication and developed respiratory disease. Evidence of coagulation dysfunction as noted by a transient increases in aPTT and circulating levels of fibrinogen was observed in all AGMs. The level of SARS-CoV-2 replication and lung pathology was not quite as pronounced as previously reported with AGMs exposed by the combined i.n. and i.t. routes; however, SARS-CoV-2 RNA was detected in nasal swabs of some animals as late as day 15 and rectal swabs as late as day 28 after virus challenge. Of particular importance to this study, all three AGMs that were followed until the early convalescence stage of COVID-19 showed substantial lung pathology at necropsy as evidenced by multifocal chronic interstitial pneumonia and increased collagen deposition in alveolar walls despite the absence of detectable SARS-CoV-2 in any of the lungs of these animals. These findings are consistent with human COVID-19 further demonstrating that the AGM faithfully reproduces the human condition.


Asunto(s)
Betacoronavirus/patogenicidad , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Neumonía Viral/patología , Neumonía Viral/virología , Animales , Betacoronavirus/inmunología , COVID-19 , Chlorocebus aethiops , Convalecencia , Infecciones por Coronavirus/sangre , Modelos Animales de Enfermedad , Femenino , Lesión Pulmonar/patología , Lesión Pulmonar/virología , Mucosa Nasal/virología , Pandemias , Neumonía Viral/sangre , SARS-CoV-2 , Seroconversión , Carga Viral , Esparcimiento de Virus
17.
Emerg Infect Dis ; 25(6): 1144-1152, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31107231

RESUMEN

Nipah virus (NiV) is a zoonotic pathogen that causes high case-fatality rates (CFRs) in humans. Two NiV strains have caused outbreaks: the Malaysia strain (NiVM), discovered in 1998-1999 in Malaysia and Singapore (≈40% CFR); and the Bangladesh strain (NiVB), discovered in Bangladesh and India in 2001 (≈80% CFR). Recently, NiVB in African green monkeys resulted in a more severe and lethal disease than NiVM. No NiV vaccines or treatments are licensed for human use. We assessed replication-restricted single-injection recombinant vesicular stomatitis vaccine NiV vaccine vectors expressing the NiV glycoproteins against NiVB challenge in African green monkeys. All vaccinated animals survived to the study endpoint without signs of NiV disease; all showed development of NiV F Ig, NiV G IgG, or both, as well as neutralizing antibody titers. These data show protective efficacy against a stringent and relevant NiVB model of human infection.


Asunto(s)
Chlorocebus aethiops , Infecciones por Henipavirus , Virus Nipah , Vesiculovirus , Vacunas Virales , Zoonosis , Animales , Femenino , Masculino , Infecciones por Henipavirus/mortalidad , Infecciones por Henipavirus/prevención & control , Infecciones por Henipavirus/veterinaria , Infecciones por Henipavirus/virología , Inmunidad Humoral , Enfermedades de los Monos/patología , Enfermedades de los Monos/virología , Vesiculovirus/inmunología , Carga Viral , Vacunas Virales/inmunología
18.
J Virol ; 92(3)2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29142131

RESUMEN

Previous studies demonstrated that a single intramuscular (i.m.) dose of an attenuated recombinant vesicular stomatitis virus (rVSV) vector (VesiculoVax vector platform; rVSV-N4CT1) expressing the glycoprotein (GP) from the Mayinga strain of Zaire ebolavirus (EBOV) protected nonhuman primates (NHPs) from lethal challenge with EBOV strains Kikwit and Makona. Here, we studied the immunogenicities of an expanded range of attenuated rVSV vectors expressing filovirus GP in mice. Based on data from those studies, an optimal attenuated trivalent rVSV vector formulation was identified that included rVSV vectors expressing EBOV, Sudan ebolavirus (SUDV), and the Angola strain of Marburg marburgvirus (MARV) GPs. NHPs were vaccinated with a single dose of the trivalent formulation, followed by lethal challenge 28 days later with each of the three corresponding filoviruses. At day 14 postvaccination, a serum IgG response specific for all three GPs was detected in all the vaccinated macaques. A modest and balanced cell-mediated immune response specific for each GP was also detected in a majority of the vaccinated macaques. No matter the level of total GP-specific immune response detected postvaccination, all the vaccinated macaques were protected from disease and death following lethal challenge with each of the three filoviruses. These findings indicate that vaccination with a single dose of attenuated rVSV-N4CT1 vectors each expressing a single filovirus GP may provide protection against the filoviruses most commonly responsible for outbreaks of hemorrhagic fever in sub-Saharan Africa.IMPORTANCE The West African Ebola virus Zaire outbreak in 2013 showed that the disease was not only a regional concern, but a worldwide problem, and highlighted the need for a safe and efficacious vaccine to be administered to the populace. However, other endemic pathogens, like Ebola virus Sudan and Marburg, also pose an important health risk to the public and therefore require development of a vaccine prior to the occurrence of an outbreak. The significance of our research was the development of a blended trivalent filovirus vaccine that elicited a balanced immune response when administered as a single dose and provided complete protection against a lethal challenge with all three filovirus pathogens.


Asunto(s)
Ebolavirus/metabolismo , Glicoproteínas/metabolismo , Fiebre Hemorrágica Ebola/prevención & control , Enfermedad del Virus de Marburg/prevención & control , Marburgvirus/metabolismo , Vesiculovirus/genética , Vacunas Virales/administración & dosificación , Animales , Anticuerpos Antivirales/metabolismo , Ebolavirus/inmunología , Glicoproteínas/genética , Glicoproteínas/inmunología , Fiebre Hemorrágica Ebola/inmunología , Inmunoglobulina G/metabolismo , Inyecciones Intramusculares , Macaca fascicularis , Enfermedad del Virus de Marburg/inmunología , Marburgvirus/inmunología , Ratones , Vacunación , Vacunas Atenuadas , Vacunas Sintéticas , Vesiculovirus/metabolismo , Proteínas Virales/genética , Proteínas Virales/inmunología , Proteínas Virales/metabolismo , Vacunas Virales/inmunología
19.
Proc Natl Acad Sci U S A ; 113(16): 4458-63, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27044104

RESUMEN

Countermeasures against potential biothreat agents remain important to US Homeland Security, and many of these pharmaceuticals could have dual use in the improvement of global public health. Junin virus, the causative agent of Argentine hemorrhagic fever (AHF), is an arenavirus identified as a category A high-priority agent. There are no Food and Drug Administration (FDA) approved drugs available for preventing or treating AHF, and the current treatment option is limited to administration of immune plasma. Whereas immune plasma demonstrates the feasibility of passive immunotherapy, it is limited in quantity, variable in quality, and poses safety risks such as transmission of transfusion-borne diseases. In an effort to develop a monoclonal antibody (mAb)-based alternative to plasma, three previously described neutralizing murine mAbs were expressed as mouse-human chimeric antibodies and evaluated in the guinea pig model of AHF. These mAbs provided 100% protection against lethal challenge when administered 2 d after infection (dpi), and one of them (J199) was capable of providing 100% protection when treatment was initiated 6 dpi and 92% protection when initiated 7 dpi. The efficacy of J199 is superior to that previously described for all other evaluated drugs, and its high potency suggests that mAbs like J199 offer an economical alternative to immune plasma and an effective dual use (bioterrorism/public health) therapeutic.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Antivirales/farmacología , Fiebre Hemorrágica Americana/tratamiento farmacológico , Fiebre Hemorrágica Americana/inmunología , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/genética , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Cobayas , Humanos , Virus Junin , Ratones , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacología
20.
J Infect Dis ; 218(suppl_5): S582-S587, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29939296

RESUMEN

A recombinant vesicular stomatitis virus (rVSV) expressing the Marburg virus (MARV) Musoke variant glycoprotein fully protects macaques against 2 MARV variants and Ravn virus as a preventive vaccine and MARV variant Musoke as a postexposure treatment. To evaluate postexposure efficacy against the most pathogenic MARV variant, Angola, we engineered rVSVs expressing homologous Angola glycoprotein. Macaques were challenged with high or low doses of variant Angola and treated 20-30 minutes after exposure. A total of 25% and 60%-75% of treated macaques survived the high-dose and low-dose challenges, respectively. The more rapid disease progression of variant Angola versus variant Musoke may account for the incomplete protection observed.


Asunto(s)
Vectores Genéticos , Enfermedad del Virus de Marburg/prevención & control , Marburgvirus/inmunología , Virus de la Estomatitis Vesicular Indiana/genética , Vacunas Virales/inmunología , Animales , Femenino , Macaca mulatta , Masculino , Vacunas Sintéticas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA