Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 6.797
1.
Can Vet J ; 65(6): 581-586, 2024 Jun.
Article En | MEDLINE | ID: mdl-38827595

Objective: Bovine respiratory disease (BRD) and overall postweaning treatment rates were compared among 3 groups of calves either differentially primed and boosted with commercially available bovine coronavirus (BCoV) vaccine or not vaccinated against BCoV. Animals: Commercial heifer and steer beef calves born in April and May 2022. Procedure: In June 2022, calves were randomly enrolled into 3 treatment groups. Those in 2 groups [V1 (n = 160) and V2 (n = 160)] were administered a mucosal priming dose of 1 of 2 commercial BCoV vaccines; those in the 3rd group [CTL (n = 151)] were unvaccinated against BCoV. The V1 and V2 groups were boosted by intramuscular injection pre-weaning with the same vaccine used for priming. Weaning occurred 3 wk after the last preweaning processing day. Ranch staff used a BRD case definition provided by their herd veterinarian to identify, treat, and record treatments for 45 d post-weaning. Results: Postweaning BRD treatment rates for V1, V2, and CTL were 7%, 9%, and 14%, respectively. The CTL calves had 2.2× greater odds of receiving treatment for BRD than V1 calves. There were no differences in odds of treatment between CTL and V2 calves or V1 and V2 calves. Conclusion: In a herd with previously diagnosed BCoV BRD cases, prime-boost vaccination of calves is associated with a difference in odds of BRD treatment post-weaning compared to not vaccinating calves against BCoV. Clinical relevance: Prime-boost vaccination with commercial BCoV vaccine may be an important management tool for herds with known BCoV BRD outbreaks.


Comparaison des taux de traitement des maladies respiratoires bovines après le sevrage entre des veaux de boucherie témoins non vaccinés et des veaux vaccinés amorce-rappel de manière variable à l'aide de vaccins contre le coronavirus bovin commercialement disponibles. Objectif: La maladie respiratoire bovine (BRD) et les taux globaux de traitement post-sevrage ont été comparés parmi 3 groupes de veaux soit vaccinés de manière différentielle et avec un rappel avec le vaccin contre le coronavirus bovin (BCoV) disponible commercialement, soit non vaccinés contre le BCoV. Animaux: Génisses et veaux de boucherie commerciaux nés en avril et mai 2022. Procédure: En juin 2022, les veaux ont été randomisés lors du recrutement dans 3 groupes de traitement. Ceux des 2 groupes [V1 (n = 160) et V2 (n = 160)] ont reçu une dose d'amorce par voie muqueuse de l'un des deux vaccins commerciaux BCoV; ceux du 3ème groupe [CTL (n = 151)] n'étaient pas vaccinés contre le BCoV. Les groupes V1 et V2 ont eu un rappel par injection intramusculaire avant le sevrage avec le même vaccin que celui utilisé pour l'amorçage. Le sevrage a eu lieu 3 semaines après le dernier jour de conditionnement pré-sevrage. Le personnel du ranch a utilisé une définition de cas de BRD fournie par le vétérinaire de leur troupeau pour identifier, traiter et enregistrer les traitements pendant 45 jours après le sevrage. Résultats: Les taux de traitement BRD post-sevrage pour V1, V2 et CTL étaient respectivement de 7 %, 9 % et 14 %. Les veaux CTL avaient 2,2 fois plus de chances de recevoir un traitement contre la BRD que les veaux V1. Il n'y avait aucune différence dans les probabilités de traitement entre les veaux CTL et V2 ou entre les veaux V1 et V2. Conclusion: Dans un troupeau avec des cas de BRD causés par le BCoV déjà diagnostiqués, la vaccination amorce-rappel des veaux est associée à une différence de probabilité de traitement par le BRD après le sevrage par rapport à la nonvaccination des veaux contre le BCoV. Pertinence clinique: La vaccination amorce-rappel avec le vaccin commercial BCoV peut être un outil de gestion important pour les troupeaux présentant des foyers connus de BCoV BRD.(Traduit par Dr Serge Messier).


Coronavirus, Bovine , Viral Vaccines , Animals , Cattle , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Coronavirus, Bovine/immunology , Male , Female , Coronavirus Infections/veterinary , Coronavirus Infections/prevention & control , Weaning , Vaccination/veterinary , Bovine Respiratory Disease Complex/prevention & control
2.
Vaccine ; 42(17): 3674-3683, 2024 Jun 20.
Article En | MEDLINE | ID: mdl-38749821

The Zika virus (ZIKV) is considered a public health problem worldwide due to its association with the development of microcephaly and the Guillain-Barré syndrome. Currently, there is no specific treatment or vaccine approved to combat this disease, and thus, developing safe and effective vaccines is a relevant goal. In this study, a multi-epitope protein called rpZDIII was designed based on a series of ZIKV antigenic sequences, a bacterial carrier, and linkers. The analysis of the predicted 3D structure of the rpZDIII chimeric antigen was performed on the AlphaFold 2 server, and it was produced in E. coli and purified from inclusion bodies, followed by solubilization and refolding processes. The yield achieved for rpZDIII was 11 mg/L in terms of pure soluble recombinant protein per liter of fermentation. rpZDIII was deemed immunogenic since it induced serum IgG and IgM responses in mice upon subcutaneous immunization in a three-dose scheme. Moreover, sera from mice immunized with rpZDIII showed neutralizing activity against ZIKV. Therefore, this study reveals rpZDIII as a promising immunogen for the development of a rationally designed multi-epitope vaccine against ZIKV, and completion of its preclinical evaluation is guaranteed.


Antibodies, Neutralizing , Antibodies, Viral , Antigens, Viral , Zika Virus Infection , Zika Virus , Animals , Zika Virus/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Mice , Antibodies, Viral/immunology , Antibodies, Viral/blood , Zika Virus Infection/prevention & control , Zika Virus Infection/immunology , Antigens, Viral/immunology , Antigens, Viral/genetics , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Epitopes/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Female , Escherichia coli/genetics , Escherichia coli/metabolism , Immunoglobulin M/immunology , Immunoglobulin M/blood , Mice, Inbred BALB C
3.
Methods Mol Biol ; 2786: 89-133, 2024.
Article En | MEDLINE | ID: mdl-38814391

While mRNA vaccines have shown their worth, they have the same failing as inactivated vaccines, namely they have limited half-life, are non-replicating, and therefore limited to the size of the vaccine payload for the amount of material translated. New advances averting these problems are combining replicon RNA (RepRNA) technology with nanotechnology. RepRNA are large self-replicating RNA molecules (typically 12-15 kb) derived from viral genomes defective in at least one essential structural protein gene. They provide sustained antigen production, effectively increasing vaccine antigen payloads over time, without the risk of producing infectious progeny. The major limitations with RepRNA are RNase-sensitivity and inefficient uptake by dendritic cells (DCs), which need to be overcome for efficacious RNA-based vaccine design. We employed biodegradable delivery vehicles to protect the RepRNA and promote DC delivery. Condensing RepRNA with polyethylenimine (PEI) and encapsulating RepRNA into novel Coatsome-replicon vehicles are two approaches that have proven effective for delivery to DCs and induction of immune responses in vivo.


Dendritic Cells , Genome, Viral , Pestivirus , RNA, Viral , Replicon , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , RNA, Viral/genetics , Pestivirus/genetics , Pestivirus/immunology , Replicon/genetics , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Vaccines/administration & dosage , Mice , Polyethyleneimine/chemistry , mRNA Vaccines , Vaccines, Synthetic/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/administration & dosage
4.
Front Immunol ; 15: 1384417, 2024.
Article En | MEDLINE | ID: mdl-38726013

Nipah virus (NiV) poses a significant threat to human and livestock populations across South and Southeast Asia. Vaccines are required to reduce the risk and impact of spillover infection events. Pigs can act as an intermediate amplifying host for NiV and, separately, provide a preclinical model for evaluating human vaccine candidate immunogenicity. The aim of this study was therefore to evaluate the immunogenicity of an mRNA vectored NiV vaccine candidate in pigs. Pigs were immunized twice with 100 µg nucleoside-modified mRNA vaccine encoding soluble G glycoprotein from the Malaysia strain of NiV, formulated in lipid nanoparticles. Potent antigen-binding and virus neutralizing antibodies were detected in serum following the booster immunization. Antibody responses effectively neutralized both the Malaysia and Bangladesh strains of NiV but showed limited neutralization of the related (about 80% amino acid sequence identity for G) Hendra virus. Antibodies were also capable of neutralizing NiV glycoprotein mediated cell-cell fusion. NiV G-specific T cell cytokine responses were also measurable following the booster immunization with evidence for induction of both CD4 and CD8 T cell responses. These data support the further evaluation of mRNA vectored NiV G as a vaccine for both pigs and humans.


Antibodies, Neutralizing , Antibodies, Viral , Henipavirus Infections , Nipah Virus , Viral Vaccines , Animals , Nipah Virus/immunology , Nipah Virus/genetics , Swine , Henipavirus Infections/prevention & control , Henipavirus Infections/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Antibodies, Viral/blood , Antibodies, Viral/immunology , Swine Diseases/immunology , Swine Diseases/prevention & control , Swine Diseases/virology , RNA, Messenger/genetics , RNA, Messenger/immunology , Immunogenicity, Vaccine , Immunization, Secondary , Cytokines/immunology , Vaccines, Synthetic/immunology , Liposomes , Nanoparticles
5.
Hum Vaccin Immunother ; 20(1): 2346390, 2024 Dec 31.
Article En | MEDLINE | ID: mdl-38691025

Middle East respiratory coronavirus (MERS-CoV) is a newly emergent, highly pathogenic coronavirus that is associated with 34% mortality rate. MERS-CoV remains listed as priority pathogen by the WHO. Since its discovery in 2012 and despite the efforts to develop coronaviruses vaccines to fight against SARS-CoV-2, there are currently no MERS-CoV vaccine that has been approved. Therefore, there is high demand to continue on the development of prophylactic vaccines against MERS-CoV. Current advancements in vaccine developments can be adapted for the development of improved MERS-CoV vaccines candidates. Nucleic acid-based vaccines, including pDNA and mRNA, are relatively new class of vaccine platforms. In this work, we developed pDNA and mRNA vaccine candidates expressing S.FL gene of MERS-CoV. Further, we synthesized a silane functionalized hierarchical aluminosilicate to encapsulate each vaccine candidates. We tested the nucleic acid vaccine candidates in mice and evaluated humoral antibodies response. Interestingly, we determined that the non-encapsulated, codon optimized S.FL pDNA vaccine candidate elicited the highest level of antibody responses against S.FL and S1 of MERS-CoV. Encapsulation of mRNA with nanoporous aluminosilicate increased the humoral antibody responses, whereas encapsulation of pDNA did not. These findings suggests that MERS-CoV S.FL pDNA vaccine candidate induced the highest level of humoral responses. This study will enhance further optimization of nanosilica as potential carrier for mRNA vaccines. In conclusion, this study suggests MERS-CoV pDNA vaccine candidate as a suitable vaccine platform for further pivotal preclinical testings.


Antibodies, Viral , Coronavirus Infections , Middle East Respiratory Syndrome Coronavirus , Nanoparticles , Silicon Dioxide , Vaccines, DNA , Viral Vaccines , Animals , Vaccines, DNA/immunology , Vaccines, DNA/genetics , Vaccines, DNA/administration & dosage , Middle East Respiratory Syndrome Coronavirus/immunology , Middle East Respiratory Syndrome Coronavirus/genetics , Mice , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Vaccines/administration & dosage , Antibodies, Viral/immunology , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Silicon Dioxide/chemistry , Mice, Inbred BALB C , Female , Humans , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Vaccine Development
6.
Euro Surveill ; 29(18)2024 May.
Article En | MEDLINE | ID: mdl-38699900

BackgroundTick-borne encephalitis (TBE) is a severe, vaccine-preventable viral infection of the central nervous system. Symptoms are generally milder in children and adolescents than in adults, though severe disease does occur. A better understanding of the disease burden and duration of vaccine-mediated protection is important for vaccination recommendations.AimTo estimate TBE vaccination coverage, disease severity and vaccine effectiveness (VE) among individuals aged 0-17 years in Switzerland.MethodsVaccination coverage between 2005 and 2022 was estimated using the Swiss National Vaccination Coverage Survey (SNVCS), a nationwide, repeated cross-sectional study assessing vaccine uptake. Incidence and severity of TBE between 2005 and 2022 were determined using data from the Swiss disease surveillance system and VE was calculated using a case-control analysis, matching TBE cases with SNVCS controls.ResultsOver the study period, vaccination coverage increased substantially, from 4.8% (95% confidence interval (CI): 4.1-5.5%) to 50.1% (95% CI: 48.3-52.0%). Reported clinical symptoms in TBE cases were similar irrespective of age. Neurological involvement was less likely in incompletely (1-2 doses) and completely (≥ 3 doses) vaccinated cases compared with unvaccinated ones. For incomplete vaccination, VE was 66.2% (95% CI: 42.3-80.2), whereas VE for complete vaccination was 90.8% (95% CI: 87.7-96.4). Vaccine effectiveness remained high, 83.9% (95% CI: 69.0-91.7) up to 10 years since last vaccination.ConclusionsEven children younger than 5 years can experience severe TBE. Incomplete and complete vaccination protect against neurological manifestations of the disease. Complete vaccination offers durable protection up to 10 years against TBE.


Encephalitis, Tick-Borne , Vaccination Coverage , Vaccination , Viral Vaccines , Humans , Encephalitis, Tick-Borne/prevention & control , Encephalitis, Tick-Borne/epidemiology , Adolescent , Case-Control Studies , Switzerland/epidemiology , Child , Cross-Sectional Studies , Male , Female , Child, Preschool , Infant , Vaccination/statistics & numerical data , Vaccination Coverage/statistics & numerical data , Viral Vaccines/administration & dosage , Incidence , Vaccine Efficacy/statistics & numerical data , Encephalitis Viruses, Tick-Borne/immunology , Infant, Newborn , Population Surveillance
7.
Viral Immunol ; 37(4): 216-219, 2024 05.
Article En | MEDLINE | ID: mdl-38717823

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.


Vaccine Efficacy , Vaccines, Attenuated , Humans , Vaccines, Attenuated/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Poxviridae Infections/prevention & control , Poxviridae Infections/immunology , Vaccinia virus/immunology , Vaccinia virus/genetics , Vaccination , Injections, Subcutaneous , Injections, Intradermal , Viral Vaccines/adverse effects , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Orthopoxvirus/immunology , Orthopoxvirus/genetics , Child
8.
Antiviral Res ; 226: 105900, 2024 Jun.
Article En | MEDLINE | ID: mdl-38705200

BACKGROUND & AIMS: The spread of foot-and-mouth disease virus (FMDV) through aerosol droplets among cloven-hoofed ungulates in close contact is a major obstacle for successful animal husbandry. Therefore, the development of suitable mucosal vaccines, especially nasal vaccines, to block the virus at the initial site of infection is crucial. PATIENTS AND METHODS: Here, we constructed eukaryotic expression plasmids containing the T and B-cell epitopes (pTB) of FMDV in tandem with the molecular mucosal adjuvant Fms-like tyrosine kinase receptor 3 ligand (Flt3 ligand, FL) (pTB-FL). Then, the constructed plasmid was electrostatically attached to mannose-modified chitosan-coated poly(lactic-co-glycolic) acid (PLGA) nanospheres (MCS-PLGA-NPs) to obtain an active nasal vaccine targeting the mannose-receptor on the surface of antigen-presenting cells (APCs). RESULTS: The MCS-PLGA-NPs loaded with pTB-FL not only induced a local mucosal immune response, but also induced a systemic immune response in mice. More importantly, the nasal vaccine afforded an 80% protection rate against a highly virulent FMDV strain (AF72) when it was subcutaneously injected into the soles of the feet of guinea pigs. CONCLUSIONS: The nasal vaccine prepared in this study can effectively induce a cross-protective immune response against the challenge with FMDV of same serotype in animals and is promising as a potential FMDV vaccine.


Administration, Intranasal , Chitosan , Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Nanospheres , Polylactic Acid-Polyglycolic Acid Copolymer , Viral Vaccines , Animals , Chitosan/chemistry , Chitosan/administration & dosage , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease Virus/genetics , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease/immunology , Mice , Nanospheres/chemistry , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Mice, Inbred BALB C , Antibodies, Viral/blood , Antibodies, Viral/immunology , Female , Nucleic Acids/administration & dosage , Immunity, Mucosal , Drug Delivery Systems
9.
Front Immunol ; 15: 1373656, 2024.
Article En | MEDLINE | ID: mdl-38742108

African swine fever virus (ASFV) is one of the most complex viruses. ASFV is a serious threat to the global swine industry because no commercial vaccines against this virus are currently available except in Vietnam. Moreover, ASFV is highly stable in the environment and can survive in water, feed, and aerosols for a long time. ASFV is transmitted through the digestive and respiratory tract. Mucosal immunity is the first line of defense against ASFV. Saccharomyces cerevisiae (SC), which has been certified by the U.S. Food and Drug Administration and has a generally recognized as safe status in the food industry, was used for oral immunization in this study. ASFV antigens were effectively expressed in recombinant SC strains with high DNA copy numbers and stable growth though surface display technology and chromosome engineering (δ-integration). The recombinant SC strains containing eight ASFV antigens-KP177R, E183L, E199L, CP204L, E248R, EP402R, B602L, and B646L- induced strong humoral and mucosal immune responses in mice. There was no antigenic competition, and these antigens induced Th1 and Th2 cellular immune responses. Therefore, the oral immunization strategy using recombinant SC strains containing multiple ASFV antigens demonstrate potential for future testing in swine, including challenge studies to evaluate its efficacy as a vaccine against ASFV.


African Swine Fever Virus , African Swine Fever , Antigens, Viral , Immunization , Saccharomyces cerevisiae , Viral Vaccines , Animals , African Swine Fever Virus/immunology , African Swine Fever Virus/genetics , Saccharomyces cerevisiae/immunology , Saccharomyces cerevisiae/genetics , Administration, Oral , Mice , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Antigens, Viral/immunology , African Swine Fever/immunology , African Swine Fever/prevention & control , Swine , Immunity, Mucosal , Antibodies, Viral/blood , Antibodies, Viral/immunology , Mice, Inbred BALB C , Female , Immunity, Humoral
10.
Vaccine ; 42(13): 3180-3189, 2024 May 10.
Article En | MEDLINE | ID: mdl-38614954

BACKGROUND: Tick-borne encephalitis (TBE) virus infects the central nervous system and may lead to severe neurological complications or death. This study assessed immunogenicity, safety, and tolerability of TBE vaccine in Japanese participants 1 year of age and older. METHODS: This phase 3, multicenter, single-arm, open-label study was conducted in Japanese adult (≥ 16 years) and pediatric (1-< 16 years) populations. Participants received a single 0.5-mL (adult) or 0.25-mL (pediatric) dose of TBE vaccine at each of 3 visits. The primary endpoint was the proportion of participants who were seropositive (neutralization test [NT] titer ≥ 1:10) 4 weeks after Dose 3. Secondary and exploratory endpoints included NT seropositivity rates 4 weeks after Dose 2, immunoglobulin G (IgG) seropositivity 4 weeks after Doses 2 and 3, NT geometric mean titers (GMTs), IgG geometric mean concentrations (GMCs), and geometric mean fold rises. Primary safety endpoints were frequencies of local reactions, systemic events, adverse events (AEs), and serious AEs. RESULTS: Among 100 adult and 65 pediatric participants, 99.0 % and 100.0 % completed the study, respectively. NT seropositivity was achieved in 98.0 % adult and 100.0 % pediatric participants after Dose 3; seropositivity after Dose 2 was 93.0 % and 92.3 %, respectively. In both age groups, IgG seropositivity was ≥ 90.0 % and ≥ 96.0 % after Doses 2 and 3, respectively; GMTs and GMCs were highest 4 weeks after Dose 3. Reactogenicity events were generally mild to moderate in severity and short-lived. AEs were reported by 15.0 % (adult) and 43.1 % (pediatric) of participants. No life-threatening AEs, AEs leading to discontinuation, immediate AEs, related AEs, or deaths were reported. No serious AEs were considered related to TBE vaccine. CONCLUSIONS: TBE vaccine elicited robust immune responses in Japanese participants 1 year of age and older. The 3-dose regimen was safe and well tolerated, and findings were consistent with the known safety profile of this TBE vaccine. CLINICALTRIALS: gov: NCT04648241.


Antibodies, Viral , Encephalitis Viruses, Tick-Borne , Encephalitis, Tick-Borne , Immunoglobulin G , Viral Vaccines , Humans , Male , Female , Encephalitis, Tick-Borne/prevention & control , Encephalitis, Tick-Borne/immunology , Antibodies, Viral/blood , Adult , Child , Child, Preschool , Adolescent , Infant , Immunoglobulin G/blood , Young Adult , Encephalitis Viruses, Tick-Borne/immunology , Middle Aged , Japan , Viral Vaccines/immunology , Viral Vaccines/adverse effects , Viral Vaccines/administration & dosage , Immunogenicity, Vaccine , Healthy Volunteers , Aged , Antibodies, Neutralizing/blood , Neutralization Tests , East Asian People
11.
Avian Dis ; 68(1): 10-17, 2024 Mar.
Article En | MEDLINE | ID: mdl-38687102

The relationship between passive immunity and the development of false layer syndrome (FLS) and its associated lesions was investigated in this study by comparing the long-term reproductive effects of an infectious bronchitis virus (IBV) DMV/1639 wild-type strain and the GA08 vaccine in birds with and without maternal antibodies. There was a clear protective effect provided by maternal antibodies against both the early vaccination and challenge. It was also observed that vaccination at an early age, in the absence of maternal antibodies, can induce reproductive issues, such as reduced egg production and FLS-associated lesions (e.g., cystic oviduct and egg yolk coelomitis). This might indicate that maternal antibodies and the timing of IBV infection are more important in the generation of FLS than the IBV strain type.


Mitigación del síndrome de la falsa ponedora mediante anticuerpos maternos contra el virus de la bronquitis infecciosa. En este estudio se investigó la relación entre la inmunidad pasiva y el desarrollo del síndrome de la falsa ponedora (FLS) y sus lesiones asociadas comparando los efectos reproductivos a largo plazo de una cepa de tipo silvestre DMV/1639 del virus de la bronquitis infecciosa (IBV) y la cepa vacunal GA08, en aves con y sin anticuerpos maternos. Hubo un claro efecto protector proporcionado por los anticuerpos maternos tanto contra la vacunación temprana como contra el desafío. También se observó que la vacunación a una edad temprana, en ausencia de anticuerpos maternos, puede inducir problemas reproductivos, como una reducción de la producción de huevo y lesiones asociadas al síndrome de la falsa ponedora (p. ej., oviducto quístico y celomitis de yema de huevo). Esto podría indicar que los anticuerpos maternos y el momento de la infección por el virus de la bronquitis infecciosa son más importantes en la generación del síndrome de la falsa ponedora que el tipo de cepa del virus de la bronquitis infecciosa.


Antibodies, Viral , Chickens , Coronavirus Infections , Infectious bronchitis virus , Poultry Diseases , Infectious bronchitis virus/immunology , Animals , Poultry Diseases/virology , Poultry Diseases/immunology , Female , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Infections/immunology , Immunity, Maternally-Acquired , Viral Vaccines/immunology , Viral Vaccines/administration & dosage
12.
Emerg Infect Dis ; 30(5): 1004-1008, 2024 May.
Article En | MEDLINE | ID: mdl-38666640

We evaluated the in vitro effects of lyophilization for 2 vesicular stomatitis virus-based vaccines by using 3 stabilizing formulations and demonstrated protective immunity of lyophilized/reconstituted vaccine in guinea pigs. Lyophilization increased stability of the vaccines, but specific vesicular stomatitis virus-based vaccines will each require extensive analysis to optimize stabilizing formulations.


Disease Models, Animal , Freeze Drying , Vesicular Stomatitis , Viral Vaccines , Animals , Guinea Pigs , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Vesicular Stomatitis/immunology , Vesicular Stomatitis/prevention & control , Vesicular Stomatitis/virology , Vesiculovirus/immunology , Vesiculovirus/genetics , Antibodies, Viral/immunology , Antibodies, Viral/blood , Vaccine Efficacy , Vesicular stomatitis Indiana virus/immunology
13.
J Gen Virol ; 105(4)2024 Apr.
Article En | MEDLINE | ID: mdl-38656455

Porcine epidemic diarrhea (PED) is a serious disease in piglets that leads to high mortality. An effective measure that provides higher IgA levels in the intestine and milk is required to decrease losses. Porcine epidemic diarrhea virus (PEDV) was dissolved in calcium alginate (Alg) and combined with chitosan (CS) via electrostatic interactions between cationic chitosan and anionic alginate to create a porous gel (Alg-CS+PEDV). The gel was used to immunize mice orally or in combination with subcutaneous injections of inactivated PEDV vaccine. At 12 and 24 days after immunization, levels of IgA and IgG in Alg-CS+PEDV were higher than with normal PEDV oral administration. At 24 days after immunization, the concentration of IFN-γ in Alg-CS+PEDV was higher than with normal PEDV oral administration. Furthermore, oral administration combining subcutaneous immunization induced higher levels of IgG and IgA than oral administration alone. Our study provides a new method for the preparation and administration of oral vaccines to achieve enhanced mucosal immunity against PEDV.


Alginates , Antibodies, Viral , Chitosan , Immunity, Mucosal , Immunoglobulin A , Immunoglobulin G , Porcine epidemic diarrhea virus , Viral Vaccines , Animals , Administration, Oral , Porcine epidemic diarrhea virus/immunology , Alginates/administration & dosage , Chitosan/administration & dosage , Mice , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Antibodies, Viral/immunology , Immunoglobulin A/immunology , Immunoglobulin G/blood , Swine , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Swine Diseases/immunology , Swine Diseases/prevention & control , Swine Diseases/virology , Female , Gels/administration & dosage , Mice, Inbred BALB C , Interferon-gamma/immunology , Glucuronic Acid/administration & dosage , Hexuronic Acids/administration & dosage
14.
J Virol ; 98(5): e0176223, 2024 May 14.
Article En | MEDLINE | ID: mdl-38563762

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged at the end of 2019 and is responsible for the largest human pandemic in 100 years. Thirty-four vaccines are currently approved for use worldwide, and approximately 67% of the world population has received a complete primary series of one, yet countries are dealing with new waves of infections, variant viruses continue to emerge, and breakthrough infections are frequent secondary to waning immunity. Here, we evaluate a measles virus (MV)-vectored vaccine expressing a stabilized prefusion SARS-CoV-2 spike (S) protein (MV-ATU3-S2PΔF2A; V591) with demonstrated immunogenicity in mouse models (see companion article [J. Brunet, Z. Choucha, M. Gransagne, H. Tabbal, M.-W. Ku et al., J Virol 98:e01693-23, 2024, https://doi.org/10.1128/jvi.01693-23]) in an established African green monkey model of disease. Animals were vaccinated with V591 or the control vaccine (an equivalent MV-vectored vaccine with an irrelevant antigen) intramuscularly using a prime/boost schedule, followed by challenge with an early pandemic isolate of SARS-CoV-2 at 56 days post-vaccination. Pre-challenge, only V591-vaccinated animals developed S-specific antibodies that had virus-neutralizing activity as well as S-specific T cells. Following the challenge, V591-vaccinated animals had lower infectious virus and viral (v) RNA loads in mucosal secretions and stopped shedding virus in these secretions earlier. vRNA loads were lower in these animals in respiratory and gastrointestinal tract tissues at necropsy. This correlated with a lower disease burden in the lungs as quantified by PET/CT at early and late time points post-challenge and by pathological analysis at necropsy.IMPORTANCESevere acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the largest human pandemic in 100 years. Even though vaccines are currently available, countries are dealing with new waves of infections, variant viruses continue to emerge, breakthrough infections are frequent, and vaccine hesitancy persists. This study uses a safe and effective measles vaccine as a platform for vaccination against SARS-CoV-2. The candidate vaccine was used to vaccinate African green monkeys (AGMs). All vaccinated AGMs developed robust antigen-specific immune responses. After challenge, these AGMs produced less virus in mucosal secretions, for a shorter period, and had a reduced disease burden in the lungs compared to control animals. At necropsy, lower levels of viral RNA were detected in tissue samples from vaccinated animals, and the lungs of these animals lacked the histologic hallmarks of SARS-CoV-2 disease observed exclusively in the control AGMs.


COVID-19 Vaccines , COVID-19 , Measles virus , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Chlorocebus aethiops , SARS-CoV-2/immunology , SARS-CoV-2/genetics , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , Measles virus/immunology , Measles virus/genetics , COVID-19 Vaccines/immunology , Humans , Antibodies, Viral/immunology , Antibodies, Viral/blood , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Genetic Vectors , Vero Cells , Pandemics/prevention & control , Female , Betacoronavirus/immunology , Betacoronavirus/genetics , Pneumonia, Viral/prevention & control , Pneumonia, Viral/virology , Pneumonia, Viral/immunology , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Coronavirus Infections/virology , Coronavirus Infections/veterinary , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Vaccines/administration & dosage , Disease Models, Animal
16.
Prev Vet Med ; 227: 106197, 2024 Jun.
Article En | MEDLINE | ID: mdl-38613943

The use of virus-neutralizing (VN) and nonstructural protein (NSP) antibody tests in a serosurveillance program for foot-and-mouth disease (FMD) can identify pig herds that are adequately vaccinated, with a high percentage of pigs with VN positive antibody titers; these tests can also help identify pigs with NSP-positivity that have previously been or are currently infected even in vaccinated herds. To identify infected herds and manage infection, the combination of VN and NSP antibody tests was used in Taiwan's serosurveillance program implemented simultaneously with the compulsory FMD vaccination program. The result was the eradication of FMD: Taiwan was recognized by the World Organization for Animal Health as an FMD-free country without vaccination in 2020. Evaluation of the compulsory vaccination program incorporated in the FMD control program in Taiwan revealed that the vaccine quality was satisfactory and the vaccination program was effective during the period of compulsory vaccination (2010-2017). Sound immunological coverage was achieved, with 89.1% of pigs having VN antibody titers exceeding 1:16 in 2016. This level of immunological coverage would be expected to substantially reduce or prevent FMD transmission, which was borne out by the results of the NSP tests. We identified farms having positive NSP reactors (very low annual prevalence) before the cessation of FMD vaccination in July 2018; however, detailed serological and clinical investigations of pigs of all ages in suspect herds demonstrated that no farms were harboring infected animals after the second half of 2013. Thus, the results revealed no evidence of FMD circulation in the field, and Taiwan regained FMD-free status.


Antibodies, Neutralizing , Antibodies, Viral , Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Swine Diseases , Viral Nonstructural Proteins , Animals , Foot-and-Mouth Disease/epidemiology , Foot-and-Mouth Disease/prevention & control , Taiwan/epidemiology , Swine , Swine Diseases/epidemiology , Swine Diseases/prevention & control , Swine Diseases/virology , Viral Nonstructural Proteins/immunology , Seroepidemiologic Studies , Antibodies, Viral/blood , Antibodies, Neutralizing/blood , Foot-and-Mouth Disease Virus/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Vaccination/veterinary
17.
Viruses ; 16(4)2024 03 30.
Article En | MEDLINE | ID: mdl-38675881

Rabbit hemorrhagic disease virus 2 (RHDV2) emerged in the United States in 2018 and has spread in both domestic and wild rabbits nationwide. The virus has a high mortality rate and can spread rapidly once introduced in a rabbit population. Vaccination against RHDV2 provides the best protection against disease and should be considered by all rabbit owners. Here, we investigate the duration of immunity provided by vaccination with the Medgene Platform conditionally licensed commercial vaccine 6 months following the initial series. Rabbits received either the vaccination or a placebo and were challenged with RHDV2 6 months later. All vaccinated rabbits survived challenge whereas 18/19 non-vaccinated controls succumbed to infection within 10 or fewer days post-challenge. These results demonstrate lasting immunity following vaccination with the Medgene RHDV2 vaccine.


Baculoviridae , Caliciviridae Infections , Hemorrhagic Disease Virus, Rabbit , Vaccination , Vaccines, Synthetic , Viral Vaccines , Animals , Hemorrhagic Disease Virus, Rabbit/immunology , Hemorrhagic Disease Virus, Rabbit/genetics , Rabbits , Caliciviridae Infections/prevention & control , Caliciviridae Infections/immunology , Caliciviridae Infections/virology , Caliciviridae Infections/veterinary , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Baculoviridae/genetics , Baculoviridae/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology
18.
Viruses ; 16(4)2024 03 30.
Article En | MEDLINE | ID: mdl-38675887

PRRS is a viral disease that profoundly impacts the global swine industry, causing significant economic losses. The development of a novel and effective vaccine is crucial to halt the rapid transmission of this virus. There have been several vaccination attempts against PRRSV using both traditional and alternative vaccine design development approaches. Unfortunately, there is no currently available vaccine that can completely control this disease. Thus, our study aimed to develop an mRNA vaccine using the antigens expressed by single or fused PRRSV structural proteins. In this study, the nucleotide sequence of the immunogenic mRNA was determined by considering the antigenicity of structural proteins and the stability of spatial structure. Purified GP5 protein served as the detection antigen in the immunological evaluation. Furthermore, cellular mRNA expression was detected by immunofluorescence and western blotting. In a mice experiment, the Ab titer in serum and the activation of spleen lymphocytes triggered by the antigen were detected by ELISA and ICS, respectively. Our findings demonstrated that both mRNA vaccines can significantly stimulate cellular and humoral immune responses. More specifically, the GP5-mRNA exhibited an immunological response that was similar to that of the commercially available vaccine when administered in high doses. To conclude, our vaccine may show promising results against the wild-type virus in a natural host.


Antibodies, Viral , Immunity, Cellular , Immunity, Humoral , Mice, Inbred BALB C , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Viral Envelope Proteins , Viral Vaccines , mRNA Vaccines , Animals , Porcine respiratory and reproductive syndrome virus/immunology , Porcine respiratory and reproductive syndrome virus/genetics , Mice , Porcine Reproductive and Respiratory Syndrome/prevention & control , Porcine Reproductive and Respiratory Syndrome/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Swine , Female , Viral Structural Proteins/immunology , Viral Structural Proteins/genetics , RNA, Messenger/genetics
19.
Viruses ; 16(4)2024 03 31.
Article En | MEDLINE | ID: mdl-38675892

Canine distemper virus (CDV) can cause fatal infections in giant pandas. Vaccination is crucial to prevent CDV infection in giant pandas. In this study, two bacterium-like particle vaccines F3-GEM and H4-GEM displaying the trimeric F protein or tetrameric H protein of CDV were constructed based on the Gram-positive enhanced-matrix protein anchor (GEM-PA) surface display system. Electron microscopy and Western blot results revealed that the F or H protein was successfully anchored on the surface of GEM particles. Furthermore, one more bacterium-like particle vaccine F3 and H4-GEM was also designed, a mixture consisting of F3-GEM and H4-GEM at a ratio of 1:1. To evaluate the effect of the three vaccines, mice were immunized with F3-GEM, H4-GEM or F3 and H4-GEM. It was found that the level of IgG-specific antibodies and neutralizing antibodies in the F3 and H4-GEM group was higher than the other two groups. Additionally, F3 and H4-GEM also increased the secretion of Th1-related and Th2-related cytokines. Moreover, F3 and H4-GEM induce IgG and neutralizing antibodies' response in dogs. Conclusions: In summary, F3 and H4-GEM can provoke better immune responses to CDV in mice and dogs. The bacterium-like particle vaccine F3 and H4-GEM might be a potential vaccine candidate for giant pandas against CDV infection.


Antibodies, Neutralizing , Antibodies, Viral , Distemper Virus, Canine , Distemper , Viral Vaccines , Animals , Distemper Virus, Canine/immunology , Dogs , Mice , Distemper/prevention & control , Distemper/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Female , Immunoglobulin G/blood , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/administration & dosage , Viral Envelope Proteins/immunology , Viral Envelope Proteins/genetics , Mice, Inbred BALB C , Cytokines/metabolism , Vaccination
20.
Vet Microbiol ; 293: 110088, 2024 Jun.
Article En | MEDLINE | ID: mdl-38640639

Orf virus (ORFV), a member of the genus Parapoxvirus, possesses an excellent immune activation capability, which makes it a promising immunomodulation agent. In this study, we evaluated ORFV as a novel adjuvant to enhance the immune response of mice to a subunit vaccine using porcine circovirus type 2 (PCV2) capsid (Cap) protein as a model. Our results showed that both inactivated and live attenuated ORFV activated mouse bone marrow-derived dendritic cells and increased expression of immune-related cytokines interleukin (IL)-1ß, IL-6, and TNF-α. Enhanced humoral and cellular immune responses were induced in mice immunized with PCV2 Cap protein combined with inactivated or live attenuated ORFV adjuvant compared with the aluminum adjuvant. Increased secretion of Th1 and Th2 cytokines by splenic lymphocytes in immunized mice further indicated that the ORFV adjuvant promoted a mixed Th1/Th2 immune response. Moreover, addition of the ORFV adjuvant to the PCV2 subunit vaccine significantly reduced the viral load in the spleen and lungs of PCV2-challenged mice and prevented pathological changes in lungs. This study demonstrates that ORFV enhances the immunogenicity of a PCV2 subunit vaccine by improving the adaptive immune response, suggesting the potential application of ORFV as a novel adjuvant.


Adjuvants, Immunologic , Circoviridae Infections , Circovirus , Cytokines , Orf virus , Vaccines, Subunit , Viral Vaccines , Animals , Circovirus/immunology , Mice , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Circoviridae Infections/prevention & control , Circoviridae Infections/veterinary , Circoviridae Infections/immunology , Circoviridae Infections/virology , Adjuvants, Immunologic/administration & dosage , Cytokines/immunology , Orf virus/immunology , Capsid Proteins/immunology , Female , Immunity, Cellular , Dendritic Cells/immunology , Viral Load , Antibodies, Viral/blood , Immunity, Humoral , Swine , Adjuvants, Vaccine , Mice, Inbred BALB C , Th1 Cells/immunology
...