Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.286
Filtrar
1.
Viral Immunol ; 37(4): 216-219, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38717823

RESUMEN

In May 2022, mpox began to spread worldwide, posing a serious threat to human public health. Modified Vaccinia Ankara-Bavaria Nordic (MVA-BN) is a live attenuated orthopoxvirus vaccine that has been authorized by the U.S. Food and Drug Administration as the vaccine of choice for the prevention of mpox. In this study, we conducted a meta-analysis of all currently published literature on the efficacy and safety of the MVA-BN vaccine in the real world, showing that the MVA-BN vaccine is effective and safe, with efficacy of up to 75% with a single dose and up to 80% with a two-dose vaccine. Meanwhile, we found that subcutaneous injection has lower local and systemic adverse events than intradermal injection, regardless of single- or two-dose vaccination, and subcutaneous injection is better tolerated in children, the elderly, or people with underlying medical conditions. These results have important reference value for clinical practice.


Asunto(s)
Eficacia de las Vacunas , Vacunas Atenuadas , Humanos , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Infecciones por Poxviridae/prevención & control , Infecciones por Poxviridae/inmunología , Virus Vaccinia/inmunología , Virus Vaccinia/genética , Vacunación , Inyecciones Subcutáneas , Inyecciones Intradérmicas , Vacunas Virales/efectos adversos , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Orthopoxvirus/inmunología , Orthopoxvirus/genética , Niño
2.
Vet Q ; 44(1): 1-12, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38726839

RESUMEN

Duck plague (DP) is an acute, contagious and fatal disease, caused by duck enteritis virus (DEV), with worldwide distribution causing several outbreaks and posing severe economic losses. The present study was carried out with a goal of development of a live attenuated cell culture based DP vaccine using an Indian strain of DEV and evaluation of its safety, efficacy along with complete genome analysis. The live attenuated DP vaccine (DPvac/IVRI-19) was developed by serial propagation of a virulent isolate of DEV (DEV/India/IVRI-2016) in the chicken embryo fibroblast (CEF) primary cell culture. Adaptation of DEV in CEF cell culture was indicated by more rapid appearance of cytopathic effects (CPE) and gradual increase of virus titre, which reached up to 107.5 TCID50/mL after 41 passages. The safety, immunogenicity and efficacy of the vaccine were determined by immunization trials in ducklings. The DPvac/IVRI-19 was found to be avirulent and completely safe in the ducklings. Further, the vaccine induced both humoral and cell mediated immune responses and afforded 100% protection against the virulent DEV challenge. A comparison of the whole genome of DPvac/IVRI-19 (MZ911871) and DEV/India/IVRI-2016 (MZ824102) revealed significant number of mutations, which might be associated with viral attenuation. Phylogenetic tree of DEV/India/IVRI-2016 revealed its evolutionary relationship with other DEV isolates, but it formed a separate cluster with certain unique mutations. Thus, with the proven safety and 100% efficacy, the DPvac/IVRI-19 is suitable for large scale production with precisely pure form of vaccine and has potential utility at national and global levels.


Asunto(s)
Patos , Fibroblastos , Mardivirus , Enfermedades de las Aves de Corral , Vacunas Atenuadas , Vacunas Virales , Animales , Vacunas Atenuadas/inmunología , Patos/virología , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/virología , Fibroblastos/virología , Embrión de Pollo , Vacunas Virales/inmunología , Mardivirus/inmunología , Mardivirus/patogenicidad , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/prevención & control , Infecciones por Herpesviridae/virología , India
3.
JCI Insight ; 9(9)2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38716733

RESUMEN

Vaccination of malaria-naive volunteers with a high dose of Plasmodium falciparum sporozoites chemoattenuated by chloroquine (CQ) (PfSPZ-CVac [CQ]) has previously demonstrated full protection against controlled human malaria infection (CHMI). However, lower doses of PfSPZ-CVac [CQ] resulted in incomplete protection. This provides the opportunity to understand the immune mechanisms needed for better vaccine-induced protection by comparing individuals who were protected with those not protected. Using mass cytometry, we characterized immune cell composition and responses of malaria-naive European volunteers who received either lower doses of PfSPZ-CVac [CQ], resulting in 50% protection irrespective of the dose, or a placebo vaccination, with everyone becoming infected following CHMI. Clusters of CD4+ and γδ T cells associated with protection were identified, consistent with their known role in malaria immunity. Additionally, EMRA CD8+ T cells and CD56+CD8+ T cell clusters were associated with protection. In a cohort from a malaria-endemic area in Gabon, these CD8+ T cell clusters were also associated with parasitemia control in individuals with lifelong exposure to malaria. Upon stimulation with P. falciparum-infected erythrocytes, CD4+, γδ, and EMRA CD8+ T cells produced IFN-γ and/or TNF, indicating their ability to mediate responses that eliminate malaria parasites.


Asunto(s)
Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Vacunas contra la Malaria , Malaria Falciparum , Plasmodium falciparum , Esporozoítos , Humanos , Vacunas contra la Malaria/inmunología , Vacunas contra la Malaria/administración & dosificación , Malaria Falciparum/inmunología , Malaria Falciparum/prevención & control , Plasmodium falciparum/inmunología , Linfocitos T CD8-positivos/inmunología , Adulto , Esporozoítos/inmunología , Masculino , Linfocitos T CD4-Positivos/inmunología , Cloroquina/uso terapéutico , Cloroquina/farmacología , Femenino , Adulto Joven , Gabón , Vacunación/métodos , Antimaláricos/uso terapéutico , Antimaláricos/administración & dosificación , Europa (Continente) , Parasitemia/inmunología , Adolescente , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/administración & dosificación , Pueblo Europeo
5.
Front Immunol ; 15: 1367253, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38646533

RESUMEN

Bovine respiratory disease (BRD) is one of the most common diseases in the cattle industry worldwide; it is caused by multiple bacterial or viral coinfections, of which Mycoplasma bovis (M. bovis) and bovine herpesvirus type 1 (BoHV-1) are the most notable pathogens. Although live vaccines have demonstrated better efficacy against BRD induced by both pathogens, there are no combined live and marker vaccines. Therefore, we developed an attenuated and marker M. bovis-BoHV-1 combined vaccine based on the M. bovis HB150 and BoHV-1 gG-/tk- strain previously constructed in our lab and evaluated in rabbits. This study aimed to further evaluate its safety and protective efficacy in cattle using different antigen ratios. After immunization, all vaccinated cattle had a normal rectal temperature and mental status without respiratory symptoms. CD4+, CD8+, and CD19+ cells significantly increased in immunized cattle and induced higher humoral and cellular immune responses, and the expression of key cytokines such as IL-4, IL-12, TNF-α, and IFN-γ can be promoted after vaccination. The 1.0 × 108 CFU of M. bovis HB150 and 1.0 × 106 TCID50 BoHV-1 gG-/tk- combined strain elicited the most antibodies while significantly increasing IgG and cellular immunity after challenge. In conclusion, the M. bovis HB150 and BoHV-1 gG-/tk- combined strain was clinically safe and protective in calves; the mix of 1.0 × 108 CFU of M. bovis HB150 and 1.0 × 106 TCID50 BoHV-1 gG-/tk- strain was most promising due to its low amount of shedding and highest humoral and cellular immune responses compared with others. This study introduces an M. bovis-BoHV-1 combined vaccine for application in the cattle industry.


Asunto(s)
Herpesvirus Bovino 1 , Mycoplasma bovis , Vacunas Atenuadas , Vacunas Combinadas , Animales , Bovinos , Herpesvirus Bovino 1/inmunología , Vacunas Combinadas/inmunología , Vacunas Combinadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/administración & dosificación , Mycoplasma bovis/inmunología , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/efectos adversos , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/efectos adversos , Citocinas/metabolismo , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Infecciones por Mycoplasma/prevención & control , Infecciones por Mycoplasma/veterinaria , Infecciones por Mycoplasma/inmunología , Vacunas Marcadoras/inmunología , Vacunas Marcadoras/administración & dosificación , Vacunación/veterinaria , Eficacia de las Vacunas , Inmunidad Humoral , Complejo Respiratorio Bovino/prevención & control , Complejo Respiratorio Bovino/inmunología , Complejo Respiratorio Bovino/virología
6.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(2): 441-446, 2024 Mar 20.
Artículo en Chino | MEDLINE | ID: mdl-38645870

RESUMEN

Objective: To study the immunoadjuvant effects of chitosan oligosaccharide (COS), including the immune activation and the triggering of lysosomal escape, and to explore whether COS can be used as an adjuvant for attenuated live bacteria vector vaccines. Methods: 1) Mouse macrophages RAW264.7 cells were cultured with COS at 0 mg/mL (the control group) and 0.1-4 mg/mL for 24 h and the effect on cell viability was measured by CCK8 assay. Mouse macrophages RAW264.7 were treated with COS at 0 (the control group), 1, 2, and 4 mg/mL for 24 h. Then, the mRNA expression levels of the cytokines, including IFN-γ, IL-10, TGF-ß, and TLR4, were determined by RT-qPCR assay. 2) RAW264.7 cells were treated with 1 mL of PBS containing different components, including calcein at 50 µg/mL, COS at 2 mg/mL, and bafilomycin A1, an inhibitor, at 1 µmol/mL, for culturing. The cells were divided into the Calcein group, Calcein+COS group, and Calcein+COS+Bafilomycin A1 group accordingly. Laser scanning confocal microscopy was used to observe the phagocytosis and the intracellular fluorescence distribution of calcein, a fluorescent dye, in RAW264.7 cells in the presence or absence of COS intervention to determine whether COS was able to trigger lysosomal escape. 3) LM∆E6E7 and LI∆E6E7, the attenuated Listeria vector candidate therapeutic vaccines for cervical cancer, were encapsulated with COS at the mass concentrations of 0.5 mg/mL, 1 mg/mL, 2 mg/mL , 4 mg/mL, and 8 mg/mL. Then, the changes in zeta potential were measured to select the concentration of COS that successfully encapsulated the bacteria. Phagocytosis of the vaccine strains by RAW264.7 cells was measured before and after LM∆E6E7 and LI∆E6E7 were coated with COS at 2 mg/mL. Results: 1) CCK8 assays showed that, compared with the findings for the control group, the intervention of RAW264.7 cells with COS at different concentrations for 24 h was not toxic to the cells and promoted cell proliferation, with the difference being statistically significant (P<0.05). According to the RT-qPCR results, compared with those of the control group, the COS intervention up-regulated the mRNA levels of TLR4 and IFN-γ in RAW264.7 cells, while it inhibited the mRNA expression levels of TGF-ß and IL-10, with the most prominent effect being observed in the 4 mg/mL COS group (P<0.05). 2) Laser scanning confocal microscopy revealed that the amount of fluorescent dye released from lysosomes into the cells was greater in the Calcein+COS group than that in the Calcein group. In other words, a greater amount of fluorescent dye was released from lysosomes into the cells under COS intervention. Furthermore, this process could be blocked by bafilomycin A1. 3) The zeta potential results showed that COS could successfully encapsulate the surface of bacteria when its mass concentration reached 2 mg/mL. Before and after the vaccine strain was encapsulated by COS, the phagocytosis of LM∆E6E7 by RAW264.7 cells was 5.70% and 22.00%, respectively, showing statistically significant differences (P<0.05); the phagocytosis of LI∆E6E7 by RAW264.7 cells was 1.55% and 6.12%, respectively, showing statistically significant differences (P<0.05). Conclusion: COS has the effect of activating the immune response of macrophages and triggering lysosomal escape. The candidates strains of coated live attenuated bacterial vector vaccines can promote the phagocytosis of bacteria by macrophages. Further research is warranted to develop COS into an adjuvant for bacterial vector vaccine.


Asunto(s)
Adyuvantes Inmunológicos , Vacunas Bacterianas , Quitosano , Oligosacáridos , Animales , Ratones , Células RAW 264.7 , Oligosacáridos/farmacología , Adyuvantes Inmunológicos/farmacología , Vacunas Bacterianas/inmunología , Macrófagos/metabolismo , Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Vacunas Atenuadas/inmunología , Citocinas/metabolismo , Supervivencia Celular/efectos de los fármacos
7.
Hum Vaccin Immunother ; 20(1): 2341456, 2024 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-38650460

RESUMEN

Few papers focus their attention on VZV vaccination effectiveness among people living with HIV (PLWH). Flanking the live attenuated vaccine (VZL) available, a newly recombinant vaccine (RZV) was recently introduced and approved for HZ prevention among adults. PLWH represents a population on which a particular attention should be applied, in order to guarantee the vaccine efficacy and safety. We performed a literature search in USNLM, PubMed, PubMed Central, PMC and Cochrane Library. From all the publications found eligible, data were extracted and processed per population, vaccine type, immunogenicity and ADRs. The review of the 13 included studies shows that both RZV and VZL are immunogenic and have an acceptable safety profile in adults and children living with HIV. However, given the lack of research available about vaccine efficacy in preventing VZV and HZ in PLWH, additional studies need to be performed, in order to achieve a full completeness of data.


Asunto(s)
Infecciones por VIH , Vacuna contra el Herpes Zóster , Herpes Zóster , Vacunas Atenuadas , Vacunas Sintéticas , Humanos , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/administración & dosificación , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Vacuna contra el Herpes Zóster/inmunología , Vacuna contra el Herpes Zóster/efectos adversos , Vacuna contra el Herpes Zóster/administración & dosificación , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/administración & dosificación , Herpes Zóster/prevención & control , Herpes Zóster/inmunología , Vacunas de Productos Inactivados/inmunología , Vacunas de Productos Inactivados/efectos adversos , Vacunas de Productos Inactivados/administración & dosificación , Inmunogenicidad Vacunal , Eficacia de las Vacunas , Herpesvirus Humano 3/inmunología , Adulto , Niño , Vacunación , Vacuna contra la Varicela/inmunología , Vacuna contra la Varicela/administración & dosificación , Vacuna contra la Varicela/efectos adversos
8.
Int J Mol Sci ; 25(8)2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38673913

RESUMEN

Shigellosis is a severe gastrointestinal disease that annually affects approximately 270 million individuals globally. It has particularly high morbidity and mortality in low-income regions; however, it is not confined to these regions and occurs in high-income nations when conditions allow. The ill effects of shigellosis are at their highest in children ages 2 to 5, with survivors often exhibiting impaired growth due to infection-induced malnutrition. The escalating threat of antibiotic resistance further amplifies shigellosis as a serious public health concern. This review explores Shigella pathology, with a primary focus on the status of Shigella vaccine candidates. These candidates include killed whole-cells, live attenuated organisms, LPS-based, and subunit vaccines. The strengths and weaknesses of each vaccination strategy are considered. The discussion includes potential Shigella immunogens, such as LPS, conserved T3SS proteins, outer membrane proteins, diverse animal models used in Shigella vaccine research, and innovative vaccine development approaches. Additionally, this review addresses ongoing challenges that necessitate action toward advancing effective Shigella prevention and control measures.


Asunto(s)
Disentería Bacilar , Vacunas contra la Shigella , Shigella , Humanos , Vacunas contra la Shigella/inmunología , Vacunas contra la Shigella/administración & dosificación , Disentería Bacilar/prevención & control , Disentería Bacilar/inmunología , Animales , Shigella/inmunología , Shigella/patogenicidad , Vacunas de Subunidad/inmunología , Desarrollo de Vacunas , Vacunas Atenuadas/inmunología
9.
Eur J Immunol ; 54(5): e2250133, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38571392

RESUMEN

Live-attenuated yellow fever vaccine (YF17D) was developed in the 1930s as the first ever empirically derived human vaccine. Ninety years later, it is still a benchmark for vaccines made today. YF17D triggers a particularly broad and polyfunctional response engaging multiple arms of innate, humoral and cellular immunity. This unique immunogenicity translates into an extraordinary vaccine efficacy and outstanding longevity of protection, possibly by single-dose immunization. More recently, progress in molecular virology and synthetic biology allowed engineering of YF17D as a powerful vector and promising platform for the development of novel recombinant live vaccines, including two licensed vaccines against Japanese encephalitis and dengue, even in paediatric use. Likewise, numerous chimeric and transgenic preclinical candidates have been described. These include prophylactic vaccines against emerging viral infections (e.g. Lassa, Zika and SARS-CoV-2) and parasitic diseases (e.g. malaria), as well as therapeutic applications targeting persistent infections (e.g. HIV and chronic hepatitis), and cancer. Efforts to overcome historical safety concerns and manufacturing challenges are ongoing and pave the way for wider use of YF17D-based vaccines. In this review, we summarize recent insights regarding YF17D as vaccine platform, and how YF17D-based vaccines may complement as well as differentiate from other emerging modalities in response to unmet medical needs and for pandemic preparedness.


Asunto(s)
Vacunas Atenuadas , Vacuna contra la Fiebre Amarilla , Virus de la Fiebre Amarilla , Humanos , Vacuna contra la Fiebre Amarilla/inmunología , Virus de la Fiebre Amarilla/inmunología , Vacunas Atenuadas/inmunología , Animales , Fiebre Amarilla/prevención & control , Fiebre Amarilla/inmunología , Vacunación/métodos
10.
Nat Commun ; 15(1): 3553, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38670948

RESUMEN

Immunization via the respiratory route is predicted to increase the effectiveness of a SARS-CoV-2 vaccine. Here, we evaluate the immunogenicity and protective efficacy of one or two doses of a live-attenuated murine pneumonia virus vector expressing SARS-CoV-2 prefusion-stabilized spike protein (MPV/S-2P), delivered intranasally/intratracheally to male rhesus macaques. A single dose of MPV/S-2P is highly immunogenic, and a second dose increases the magnitude and breadth of the mucosal and systemic anti-S antibody responses and increases levels of dimeric anti-S IgA in the airways. MPV/S-2P also induces S-specific CD4+ and CD8+ T-cells in the airways that differentiate into large populations of tissue-resident memory cells within a month after the boost. One dose induces substantial protection against SARS-CoV-2 challenge, and two doses of MPV/S-2P are fully protective against SARS-CoV-2 challenge virus replication in the airways. A prime/boost immunization with a mucosally-administered live-attenuated MPV vector could thus be highly effective in preventing SARS-CoV-2 infection and replication.


Asunto(s)
Anticuerpos Antivirales , Vacunas contra la COVID-19 , COVID-19 , Inmunización Secundaria , Macaca mulatta , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Animales , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/administración & dosificación , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , COVID-19/prevención & control , COVID-19/inmunología , COVID-19/virología , Masculino , Anticuerpos Antivirales/inmunología , Ratones , Linfocitos T CD8-positivos/inmunología , Vectores Genéticos/inmunología , Vectores Genéticos/genética , Anticuerpos Neutralizantes/inmunología , Administración Intranasal , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/administración & dosificación , Inmunoglobulina A/inmunología , Linfocitos T CD4-Positivos/inmunología , Humanos
11.
Avian Dis ; 68(1): 18-24, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38687103

RESUMEN

The application of live attenuated Salmonella Typhimurium vaccines has significantly helped control Salmonella in poultry products. Because the U.S. Department of Agriculture-Food Safety Inspection Service (USDA-FSIS) scores all Salmonella as positive, regardless of serovar, attenuated vaccine strains that are identified at processing contribute negatively toward Salmonella performance standards. This study was designed to determine the incidence of a live attenuated Salmonella serovar Typhimurium vaccine identified in broiler products by FSIS and to develop a PCR assay for screening of isolates. Salmonella Typhimurium short-read sequences from broiler samples uploaded to the National Center for Biotechnology Information (NCBI) Pathogen Detection database by the USDA-FSIS from 2016 to 2022 were downloaded and assembled. These were analyzed using the Basic Local Alignment Search Tool (BLAST) with a sequence unique to field strains, followed by a sequence unique to the vaccine strain. The PCR assays were developed against field and vaccine strains by targeting transposition events in the crp and cya genes and validated by screening Salmonella serovar Typhimurium isolates. Between 2016 and 2022, 1708 Salmonella Typhimurium isolates of chicken origin were found in the NCBI Pathogen Detection database, corresponding to 7.99% of all Salmonella identified. Of these, 104 (5.97%) were identified as the vaccine strain. The PCR assay differentiated field strains from the vaccine strain when applied to isolates and was also able to detect the vaccine strain from DNA isolated from mixed serovar overnight Salmonella enrichment cultures. Live attenuated Salmonella vaccines are a critical preharvest tool for Salmonella control and are widely used in industry. With forthcoming regulations that will likely focus on Salmonella Typhimurium, along with other serovars, there is a need to distinguish between isolates belonging to the vaccine strain and those that are responsible for causing human illness.


Detección in silico y por PCR de una cepa vacunal viva atenuada de Salmonella Typhimurium. La aplicación de vacunas vivas atenuadas contra Salmonella Typhimurium ha ayudado significativamente a controlar Salmonella en productos avícolas. Debido a que el Servicio de Inspección de Seguridad Alimentaria del Departamento de Agricultura de los Estados Unidos. (USDA-FSIS) califica todas las Salmonella como positivas, independientemente del serovar. Las cepas atenuadas de la vacuna que se identifican en el procesamiento contribuyen negativamente a los estándares de desempeño de Salmonella. Este estudio fue diseñado para determinar la incidencia de una vacuna viva atenuada de Salmonella serovar Typhimurium identificada en productos de pollo de engorde por el FSIS y para desarrollar un ensayo de PCR para la detección de aislados. Se recolectaron y ensamblaron secuencias de lectura corta de Salmonella Typhimurium de muestras de pollos de engorde introducidas en la plataforma de detección de patógenos del Centro Nacional de Información Biotecnológica (NCBI) por el USDA-FSIS entre los años 2016 al 2022. Estos se analizaron utilizando la herramienta de búsqueda de alineación local básica con una secuencia exclusiva para las cepas de campo, seguida de una secuencia exclusiva para la cepa vacunal. Los ensayos de PCR se desarrollaron contra cepas de campo y vacunales centrándose en eventos de transposición en los genes crp y cya y se validaron mediante la detección de aislados de Salmonella serovar Typhimurium. Entre 2016 y 2022, se encontraron 1708 aislados de Salmonella Typhimurium de origen avícola en el sistema de detección de patógenos del NCBI, lo que corresponde al 7.99 % de todas las Salmonellas identificadas. De ellas, 104 (5.97%) fueron identificadas como cepa vacunal. El ensayo de PCR diferenció las cepas de campo de la cepa de la vacuna cuando se aplicó a los aislados y también fue capaz de detectar la cepa de la vacuna a partir del ADN aislado de cultivos de enriquecimiento por toda la noche de Salmonella con serovares mixtos. Las vacunas vivas atenuadas contra Salmonella son una herramienta fundamental para el control de Salmonella y se utilizan ampliamente en la industria. Con las próximas regulaciones que probablemente se centrarán en Salmonella Typhimurium, junto con otros serovares, es necesario distinguir entre los aislados que pertenecen a la cepa vacunal y los que son responsables de causar enfermedades humanas.


Asunto(s)
Pollos , Reacción en Cadena de la Polimerasa , Enfermedades de las Aves de Corral , Salmonelosis Animal , Vacunas contra la Salmonella , Salmonella typhimurium , Vacunas Atenuadas , Salmonella typhimurium/genética , Salmonella typhimurium/aislamiento & purificación , Vacunas Atenuadas/inmunología , Animales , Vacunas contra la Salmonella/inmunología , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/microbiología , Salmonelosis Animal/prevención & control , Salmonelosis Animal/microbiología , Reacción en Cadena de la Polimerasa/veterinaria , Simulación por Computador
12.
PLoS Negl Trop Dis ; 18(4): e0012120, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38648230

RESUMEN

Chikungunya fever virus (CHIKV) is a mosquito-borne alphavirus that causes wide-spread human infections and epidemics in Asia, Africa and recently, in the Americas. CHIKV is considered a priority pathogen by CEPI and WHO. Despite recent approval of a live-attenuated CHIKV vaccine, development of additional vaccines is warranted due to the worldwide outbreaks of CHIKV. Previously, we developed immunization DNA (iDNA) plasmid capable of launching live-attenuated CHIKV vaccine in vivo. Here we report the use of CHIKV iDNA plasmid to prepare a novel, live-attenuated CHIKV vaccine V5040 with rearranged RNA genome. In V5040, genomic RNA was rearranged to encode capsid gene downstream from the glycoprotein genes. Attenuated mutations derived from experimental CHIKV 181/25 vaccine were also engineered into E2 gene of V5040. The DNA copy of rearranged CHIKV genomic RNA with attenuated mutations was cloned into iDNA plasmid pMG5040 downstream from the CMV promoter. After transfection in vitro, pMG5040 launched replication of V5040 virus with rearranged genome and attenuating E2 mutations. Furthermore, V5040 virus was evaluated in experimental murine models for general safety and immunogenicity. Vaccination with V5040 virus subcutaneously resulted in elicitation of CHIKV-specific, virus-neutralizing antibodies. The results warrant further evaluation of V5040 virus with rearranged genome as a novel live-attenuated vaccine for CHIKV.


Asunto(s)
Anticuerpos Antivirales , Fiebre Chikungunya , Virus Chikungunya , Genoma Viral , Vacunas Atenuadas , Vacunas Virales , Replicación Viral , Animales , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/genética , Vacunas Atenuadas/administración & dosificación , Ratones , Virus Chikungunya/genética , Virus Chikungunya/inmunología , Vacunas Virales/inmunología , Vacunas Virales/genética , Vacunas Virales/administración & dosificación , Fiebre Chikungunya/prevención & control , Fiebre Chikungunya/inmunología , Fiebre Chikungunya/virología , Anticuerpos Antivirales/sangre , Femenino , Humanos , Chlorocebus aethiops , Anticuerpos Neutralizantes/sangre , Células Vero , Ratones Endogámicos BALB C
13.
Vaccine ; 42(13): 3166-3171, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38677792

RESUMEN

BACKGROUND: Immunisation against herpes zoster is recommended for adults aged ≥ 50 years. Two vaccines, a live attenuated (ZVL, Zostavax®) and an adjuvant recombinant subunit (HZ/su, Shingrix®), are available in Australia. Immunisation guidelines are shifting their recommendations towards HZ/su because of higher efficacy in preventing herpes zoster and associated complications. However, there are limited post-marketing data comparing the safety profiles of these vaccines. METHODS: Data from SmartVax, an active surveillance system for monitoring adverse events following immunisation (AEFIs) utilised by > 450 clinics throughout Australia, were analysed. Data from patients aged ≥ 50 years, who received ZVL or HZ/su, from 1 June 2021 to 31 May 2022, at clinics that utilised SmartVax were included. The proportion of records where patients who reported any, local, and systemic AEFIs after receiving ZVL or HZ/su were compared using multivariable logistic regression models. RESULTS: Data from 10,392 immunisation records (n = 8341 ZVL; n = 2051 HZ/su) were included. The proportion of AEFIs reported was higher with HZ/su (41.9 % [any], 33.8 % [local], 25.2 % [systemic]) than with ZVL (8.7 % [any], 6.2 % [local], 3.5 % [systemic]). After controlling for demographic variables, HZ/su presented a 6-fold increase in the odds (OR 6.44; 95 %CI: 5.57-7.46) of a reported AEFI compared to ZVL. Only 59 (0.6 %) of vaccinations lead to medical attention being sought due to an AEFI. CONCLUSIONS: While rates of AEFIs was higher with HZ/su than ZVL, most AEFIs were mild and did not require medical attention. Our findings support the change in vaccine recommendations and the use of HZ/su in immunisation programs.


Asunto(s)
Vacuna contra el Herpes Zóster , Herpes Zóster , Vigilancia de Productos Comercializados , Humanos , Vacuna contra el Herpes Zóster/efectos adversos , Vacuna contra el Herpes Zóster/administración & dosificación , Vacuna contra el Herpes Zóster/inmunología , Australia/epidemiología , Herpes Zóster/prevención & control , Herpes Zóster/epidemiología , Masculino , Femenino , Persona de Mediana Edad , Anciano , Vacunación/efectos adversos , Anciano de 80 o más Años , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Herpesvirus Humano 3/inmunología , Sistemas de Registro de Reacción Adversa a Medicamentos/estadística & datos numéricos
14.
Am J Trop Med Hyg ; 110(5): 892-901, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38531102

RESUMEN

Malaria eradication efforts prioritize safe and efficient vaccination strategies, although none with high-level efficacy against malaria infection are yet available. Among several vaccine candidates, Sanaria® PfSPZ Vaccine and Sanaria PfSPZ-CVac are, respectively, live radiation- and chemo-attenuated sporozoite vaccines designed to prevent infection with Plasmodium falciparum, the leading cause of malaria-related morbidity and mortality. We are conducting a randomized normal saline placebo-controlled trial called IDSPZV1 that will analyze the safety, tolerability, immunogenicity, and efficacy of PfSPZ Vaccine and PfSPZ-CVac administered pre-deployment to malaria-naive Indonesian soldiers assigned to temporary duties in a high malaria transmission area. We describe the manifold challenges of enrolling and immunizing 345 soldier participants at their home base in western Indonesia before their nearly 6,000-km voyage to eastern Indonesia, where they are being monitored for incident P. falciparum and Plasmodium vivax malaria cases during 9 months of exposure. The unique regulatory, ethical, and operational complexities of this trial demonstrate the importance of thorough planning, frequent communication, and close follow-up with stakeholders. Effective engagement with the military community and the ability to adapt to unanticipated events have proven key to the success of this trial.


Asunto(s)
Vacunas contra la Malaria , Malaria Falciparum , Malaria Vivax , Personal Militar , Plasmodium falciparum , Esporozoítos , Vacunas Atenuadas , Humanos , Vacunas contra la Malaria/inmunología , Vacunas contra la Malaria/uso terapéutico , Vacunas contra la Malaria/administración & dosificación , Indonesia/epidemiología , Malaria Falciparum/prevención & control , Malaria Falciparum/epidemiología , Esporozoítos/inmunología , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/uso terapéutico , Plasmodium falciparum/inmunología , Malaria Vivax/prevención & control , Malaria Vivax/epidemiología , Masculino , Adulto , Adulto Joven , Plasmodium vivax/inmunología , Femenino
15.
Am J Vet Res ; 85(5)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38457927

RESUMEN

OBJECTIVE: Compare immune responses induced by 2 commercial intranasal (IN) modified-live viral (MLV) vaccines given individually or coadministered and evaluate prevention of infection and lung pathology following bovine herpesvirus-1 (BHV-1) challenge. ANIMALS: 36 male Holstein calves (ages, 5 to 12 days). METHODS: In a randomized complete block design, each calf received an IN injection of either vaccine diluent (Placebo), an MLV vaccine containing bovine herpesvirus-1 (BHV-1; N3), bovine coronavirus vaccine (BC), or both N3 and BC (BC + N3) with a booster 4 weeks later. Nasal secretions and blood were collected weekly. Three weeks after the booster, the calves were challenged with BHV-1, sampled for virus shedding, and euthanized 10 days later to quantify lung pathology. The study period was September 7, 2020, to April 6, 2021. RESULTS: Calves were seropositive for BHV-1 and BC before vaccination. No significant difference in BC-specific serum immunoglobin G and nasal immunoglobin A antibody responses in the BC versus BC + N3 group or BHV-1-specific serum immunoglobin G and nasal immunoglobin A antibody responses in the N3 versus BC + N3 group. Cytokine responses to BHV-1 and BC did not differ among groups. BHV-1 shedding after challenge was significantly reduced in N3 groups versus Placebo and BC. There was a significant reduction in lung pathology in the N3 + BC group versus Placebo. CLINICAL RELEVANCE: This study provides evidence an MLV vaccine containing BHV-1 and an MLV BC vaccine can be coadministered to neonatal calves without significantly altering immune responses to the 2 viruses or compromising the prevention of BHV-1 respiratory disease. Calves receiving the BC + N3 vaccine had a significant reduction in lung pathology after BHV-1 aerosol challenge.


Asunto(s)
Administración Intranasal , Animales Recién Nacidos , Enfermedades de los Bovinos , Infecciones por Coronavirus , Coronavirus Bovino , Infecciones por Herpesviridae , Herpesvirus Bovino 1 , Vacunas Atenuadas , Vacunas Virales , Animales , Bovinos , Herpesvirus Bovino 1/inmunología , Administración Intranasal/veterinaria , Masculino , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Coronavirus Bovino/inmunología , Enfermedades de los Bovinos/prevención & control , Enfermedades de los Bovinos/virología , Enfermedades de los Bovinos/inmunología , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/prevención & control , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Rinotraqueítis Infecciosa Bovina/prevención & control , Rinotraqueítis Infecciosa Bovina/inmunología , Esparcimiento de Virus , Anticuerpos Antivirales/sangre , Distribución Aleatoria
16.
J Virol ; 97(12): e0119323, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-37971221

RESUMEN

IMPORTANCE: Coronaviruses are important pathogens of humans and animals, and vaccine developments against them are imperative. Due to the ability to induce broad and prolonged protective immunity and the convenient administration routes, live attenuated vaccines (LAVs) are promising arms for controlling the deadly coronavirus infections. However, potential recombination events between vaccine and field strains raise a safety concern for LAVs. The porcine epidemic diarrhea virus (PEDV) remodeled TRS (RMT) mutant generated in this study replicated efficiently in both cell culture and in pigs and retained protective immunogenicity against PEDV challenge in pigs. Furthermore, the RMT PEDV was resistant to recombination and genetically stable. Therefore, RMT PEDV can be further optimized as a backbone for the development of safe LAVs.


Asunto(s)
Infecciones por Coronavirus , Virus de la Diarrea Epidémica Porcina , Recombinación Genética , Enfermedades de los Porcinos , Porcinos , Vacunas Atenuadas , Vacunas Virales , Animales , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/veterinaria , Virus de la Diarrea Epidémica Porcina/genética , Virus de la Diarrea Epidémica Porcina/crecimiento & desarrollo , Virus de la Diarrea Epidémica Porcina/inmunología , Porcinos/inmunología , Porcinos/virología , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/prevención & control , Enfermedades de los Porcinos/virología , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Virales/genética , Vacunas Virales/inmunología , Replicación Viral , Células Cultivadas , Mutación
17.
J Virol ; 97(11): e0079523, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37902401

RESUMEN

IMPORTANCE: African swine fever virus (ASFV), the only known DNA arbovirus, is the causative agent of African swine fever (ASF), an acutely contagious disease in pigs. ASF has recently become a crisis in the pig industry in recent years, but there are no commercially available vaccines. Studying the immune evasion mechanisms of ASFV proteins is important for the understanding the pathogenesis of ASFV and essential information for the development of an effective live-attenuated ASFV vaccines. Here, we identified ASFV B175L, previously uncharacterized proteins that inhibit type I interferon signaling by targeting STING and 2'3'-cGAMP. The conserved B175L-zf-FCS motif specifically interacted with both cGAMP and the R238 and Y240 amino acids of STING. Consequently, this interaction interferes with the interaction of cGAMP and STING, thereby inhibiting downstream signaling of IFN-mediated antiviral responses. This novel mechanism of B175L opens a new avenue as one of the ASFV virulent genes that can contribute to the advancement of ASFV live-attenuated vaccines.


Asunto(s)
Virus de la Fiebre Porcina Africana , Fiebre Porcina Africana , Interferón Tipo I , Proteínas de la Membrana , Nucleótidos Cíclicos , Porcinos , Proteínas Virales , Animales , Fiebre Porcina Africana/inmunología , Fiebre Porcina Africana/virología , Virus de la Fiebre Porcina Africana/química , Virus de la Fiebre Porcina Africana/genética , Virus de la Fiebre Porcina Africana/inmunología , Virus de la Fiebre Porcina Africana/patogenicidad , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/inmunología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Nucleótidos Cíclicos/antagonistas & inhibidores , Nucleótidos Cíclicos/metabolismo , Porcinos/inmunología , Porcinos/virología , Vacunas Atenuadas/inmunología , Proteínas Virales/metabolismo , Vacunas Virales/inmunología , Interacciones Microbiota-Huesped
18.
J Virol ; 97(10): e0070423, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37768081

RESUMEN

IMPORTANCE: African swine fever (ASF) caused by ASF virus (ASFV) is a highly contagious and acute hemorrhagic viral disease in domestic pigs. Until now, no effective commercial vaccine and antiviral drugs are available for ASF control. Here, we generated a new live-attenuated vaccine candidate (ASFV-ΔH240R-Δ7R) by deleting H240R and MGF505-7R genes from the highly pathogenic ASFV HLJ/18 genome. Piglets immunized with ASFV-ΔH240R-Δ7R were safe without any ASF-related signs and produced specific antibodies against p30. Challenged with a virulent ASFV HLJ/18, the piglets immunized with high-dose group (105 HAD50) exhibited 100% protection without clinical symptoms, showing that low levels of virus replication with no observed pathogenicity by postmortem and histological analysis. Overall, our results provided a new strategy by designing live-attenuated vaccine candidate, resulting in protection against ASFV infection.


Asunto(s)
Virus de la Fiebre Porcina Africana , Eliminación de Gen , Genes Virales , Vacunas Atenuadas , Vacunas Virales , Animales , Fiebre Porcina Africana/inmunología , Fiebre Porcina Africana/prevención & control , Fiebre Porcina Africana/virología , Virus de la Fiebre Porcina Africana/clasificación , Virus de la Fiebre Porcina Africana/inmunología , Virus de la Fiebre Porcina Africana/patogenicidad , Sus scrofa/virología , Vacunas Atenuadas/inmunología , Proteínas Virales/genética , Vacunas Virales/genética , Vacunas Virales/inmunología , Virulencia , Replicación Viral , Genes Virales/genética
19.
Front Immunol ; 14: 1201973, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37600784

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) remains a leading cause of economic loss in pig farming worldwide. Existing commercial vaccines, all based on modified live or inactivated PRRSV, fail to provide effective immunity against the highly diverse circulating strains of both PRRSV-1 and PRRSV-2. Therefore, there is an urgent need to develop more effective and broadly active PRRSV vaccines. In the absence of neutralizing antibodies, T cells are thought to play a central role in controlling PRRSV infection. Herpesvirus-based vectors are novel vaccine platforms capable of inducing high levels of T cells against encoded heterologous antigens. Therefore, the aim of this study was to assess the immunogenicity and efficacy of an attenuated herpesvirus-based vector (bovine herpesvirus-4; BoHV-4) expressing a fusion protein comprising two well-characterized PRRSV-1 T-cell antigens (M and NSP5). Prime-boost immunization of pigs with BoHV-4 expressing the M and NSP5 fusion protein (vector designated BoHV-4-M-NSP5) induced strong IFN-γ responses, as assessed by ELISpot assays of peripheral blood mononuclear cells (PBMC) stimulated with a pool of peptides representing PRRSV-1 M and NSP5. The responses were closely mirrored by spontaneous IFN-γ release from unstimulated cells, albeit at lower levels. A lower frequency of M and NSP5 specific IFN-γ responding cells was induced following a single dose of BoHV-4-M-NSP5 vector. Restimulation using M and NSP5 peptides from PRRSV-2 demonstrated a high level of cross-reactivity. Vaccination with BoHV-4-M-NSP5 did not affect viral loads in either the blood or lungs following challenge with the two heterologous PRRSV-1 strains. However, the BoHV-4-M-NSP5 prime-boost vaccination showed a marked trend toward reduced lung pathology following PRRSV-1 challenge. The limited effect of T cells on PRRSV-1 viral load was further examined by analyzing local and circulating T-cell responses using intracellular cytokine staining and proliferation assays. The results from this study suggest that vaccine-primed T-cell responses may have helped in the control of PRRSV-1 associated tissue damage, but had a minimal, if any, effect on controlling PRRSV-1 viral loads. Together, these results indicate that future efforts to develop effective PRRSV vaccines should focus on achieving a balanced T-cell and antibody response.


Asunto(s)
Vacunas contra Herpesvirus , Inmunogenicidad Vacunal , Síndrome Respiratorio y de la Reproducción Porcina , Virus del Síndrome Respiratorio y Reproductivo Porcino , Proteínas de la Matriz Viral , Proteínas no Estructurales Virales , Vacunas contra Herpesvirus/inmunología , Vacunas Atenuadas/inmunología , Linfocitos T/inmunología , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Vectores Genéticos , Síndrome Respiratorio y de la Reproducción Porcina/inmunología , Síndrome Respiratorio y de la Reproducción Porcina/prevención & control , Animales , Porcinos , Proteínas de la Matriz Viral/inmunología
20.
Nature ; 619(7968): 135-142, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37316671

RESUMEN

Vaccination with Sabin, a live attenuated oral polio vaccine (OPV), results in robust intestinal and humoral immunity and has been key to controlling poliomyelitis. As with any RNA virus, OPV evolves rapidly to lose attenuating determinants critical to the reacquisition of virulence1-3 resulting in vaccine-derived, virulent poliovirus variants. Circulation of these variants within underimmunized populations leads to further evolution of circulating, vaccine-derived poliovirus with higher transmission capacity, representing a significant risk of polio re-emergence. A new type 2 OPV (nOPV2), with promising clinical data on genetic stability and immunogenicity, recently received authorization from the World Health Organization for use in response to circulating, vaccine-derived poliovirus outbreaks. Here we report the development of two additional live attenuated vaccine candidates against type 1 and 3 polioviruses. The candidates were generated by replacing the capsid coding region of nOPV2 with that from Sabin 1 or 3. These chimeric viruses show growth phenotypes similar to nOPV2 and immunogenicity comparable to their parental Sabin strains, but are more attenuated. Our experiments in mice and deep sequencing analysis confirmed that the candidates remain attenuated and preserve all the documented nOPV2 characteristics concerning genetic stability following accelerated virus evolution. Importantly, these vaccine candidates are highly immunogenic in mice as monovalent and multivalent formulations and may contribute to poliovirus eradication.


Asunto(s)
Poliomielitis , Vacuna Antipolio Oral , Poliovirus , Vacunas Atenuadas , Animales , Ratones , Modelos Animales de Enfermedad , Poliomielitis/inmunología , Poliomielitis/prevención & control , Poliomielitis/virología , Poliovirus/clasificación , Poliovirus/genética , Poliovirus/inmunología , Vacuna Antipolio Oral/química , Vacuna Antipolio Oral/genética , Vacuna Antipolio Oral/inmunología , Vacunas Atenuadas/química , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Erradicación de la Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA