ABSTRACT
BACKGROUND: In response to the coronavirus disease 2019 (Covid-19) pandemic, Brazil authorised the Astra Zeneca/Fiocruz vaccine in January 2021. As the Delta variant emerged in May 2021, interval between vaccine doses was adjusted. By September 2021, the Brazilian National Immunisation Program recommended a booster dose for individuals over 70, and later expanded the recommendation to all adults. OBJECTIVES: Assess the equivalence of IgG antibody response against the Covid-19 S protein before and approximately 28 days after the third dose of a Covid-19 recombinant vaccine. Two groups received initial two doses with intervals of eight and 12 weeks. METHODS: This is a phase IV clinical study, uncontrolled, non-randomised. The study proposes calculating the ratio of geometric means titres (GMT) 28 days after the third dose, with a target ratio of confidence interval (CI) between 0.77 and 1.3. FINDINGS: In the primary endpoint, there was no equivalence between the eight- and 12-week intervals with a slight variation favouring the eight-week group. Post-third dose, both groups showed increases titres at 28 days, three months, six months and 12 months. Both groups responded similarly to Delta and Omicron BA.1, with a more significant increase for Delta. MAIN CONCLUSIONS: The study showed strong and consistent immune response in all age groups receiving the Covid-19 recombinant vaccine. Third dose elicited an increase in GMT by at least three times aligned with Ministry of Health strategies emphasising Bio-Manguinhos crucial role in pandemic control in the country.
Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunization Schedule , Immunization, Secondary , Immunogenicity, Vaccine , Immunoglobulin G , SARS-CoV-2 , Vaccines, Synthetic , Humans , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , COVID-19/immunology , Male , Adult , Female , Middle Aged , SARS-CoV-2/immunology , Antibodies, Viral/blood , Immunoglobulin G/blood , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Young Adult , Aged , Brazil , Adolescent , Time FactorsABSTRACT
Acute respiratory infections are the leading cause of death and illness in children under 5 years old and represent a significant burden in older adults. Primarily caused by viruses infecting the lower respiratory tract, symptoms include cough, congestion, and low-grade fever, potentially leading to bronchiolitis and pneumonia. Messenger ribonucleic acid (mRNA)-based vaccines are biopharmaceutical formulations that employ mRNA molecules to induce specific immune responses, facilitating the expression of viral or bacterial antigens and promoting immunization against infectious diseases. Notably, this technology had significant relevance during the COVID-19 pandemic, as these formulations helped to limit SARS-CoV-2 virus infections, hospitalizations, and deaths. Importantly, mRNA vaccines promise to be implemented as new alternatives for fighting other respiratory viruses, such as influenza, human respiratory syncytial virus, and human metapneumovirus. This review article analyzes mRNA-based vaccines' main contributions, perspectives, challenges, and implications against respiratory viruses.
Subject(s)
Respiratory Tract Infections , mRNA Vaccines , Humans , Respiratory Tract Infections/prevention & control , Respiratory Tract Infections/virology , Respiratory Tract Infections/immunology , Vaccine Development , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , Animals , COVID-19 Vaccines/immunology , RNA, Messenger/genetics , RNA, Messenger/immunologyABSTRACT
INTRODUCTION: Producing commercial bacterins/toxoids against Clostridium spp. is laborious and hazardous. Conversely, developing prototype vaccines using purified recombinant toxoids, though safe and effective, is both laborious and costly for application in production animals. OBJECTIVE: Considering that inactivated recombinant Escherichiacoli (bacterin) is a simple, cost-effective, and to be safe solution, we evaluated, for the first time, a pentavalent formulation of recombinant bacterins containing the alpha, beta, and epsilon toxins of Clostridiumperfringens and C and D neurotoxins of Clostridiumbotulinum in sheep. METHODS: Subcutaneously, 18 Texel sheep received two doses (200 µg of each antigen) of recombinant bacterin (n = 7) or purified recombinant antigens (n = 6) on days 0 and 28, while the control group (n = 5) did not receive an immunization. Sera samples from days 0 (before the 1st dose), 28 (before the 2nd dose), and 56, 84, and 112 were used for measuring IgG (indirect ELISA) and neutralizing antibodies (mouse serum neutralization). RESULTS: Both formulations induced significant levels of IgG against all five toxins (p < 0.05) up to day 112, with peaks at days 28 and 56 post-immunization. The expected booster effect occurred only for the botulinum toxins. The neutralizing antibody titers were satisfactory against ETX (≥2 IU/ml for both formulations) and BoNT-D [5 IU/ml (bacterin) and 10 IU/ml (purified)]. CONCLUSION: While adjustments are required, the recombinant bacterin platform holds great potential for polyvalent vaccines due to its straightforward, safe, and cost-effective production, establishing it as a user-friendly technology for the veterinary immunobiological industry.
Subject(s)
Antibodies, Bacterial , Antibodies, Neutralizing , Bacterial Vaccines , Botulism , Enterotoxemia , Animals , Botulism/prevention & control , Botulism/veterinary , Botulism/immunology , Sheep , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Antibodies, Bacterial/blood , Enterotoxemia/prevention & control , Enterotoxemia/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Sheep Diseases/prevention & control , Sheep Diseases/immunology , Sheep Diseases/microbiology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Immunoglobulin G/blood , Escherichia coli/genetics , Recombinant Proteins/immunology , Recombinant Proteins/genetics , FemaleABSTRACT
INTRODUCTION: Clostridioides difficile is the main cause of antibiotic-associated diarrhea in humans and is a major enteropathogen in several animal species. In newborn piglets, colonic lesions caused by C. difficile A and B toxins (TcdA and TcdB, respectively) cause diarrhea and significant production losses. OBJECTIVE: The present study aimed to develop two recombinant vaccines from immunogenic C-terminal fragments of TcdA and TcdB and evaluate the immune response in rabbits and in breeding sows. Two vaccines were produced: bivalent (rAB), consisting of recombinant fragments of TcdA and TcdB, and chimeric (rQAB), corresponding to the synthesis of the same fragments in a single protein. Groups of rabbits were inoculated with 10 or 50 µg of proteins adjuvanted with aluminum or 0.85 % sterile saline in a final volume of 1 mL/dose. Anti-TcdA and anti-TcdB IgG antibodies were detected in rabbits and sows immunized with both rAB and rQAB vaccines by ELISA. The vaccinated sows were inoculated intramuscularly with 20 µg/dose using a prime-boost approach. RESULTS: Different antibody titers (p ≤ 0.05) were observed among the vaccinated groups of sows (rAB and rQAB) and control. Additionally, newborn piglets from vaccinated sows were also positive for anti-TcdA and anti-TcdB IgGs, in contrast to control piglets (p ≤ 0.05). Immunization of sows with the rQAB vaccine conferred higher anti-TcdA and anti-TcdB responses in piglets, suggesting the superiority of this compound over rAB. CONCLUSION: The synthesized recombinant proteins were capable of inducing antibody titers against C. difficile toxins A and B in sows, and were passively transferred to piglets through colostrum.
Subject(s)
Animals, Newborn , Antibodies, Bacterial , Bacterial Toxins , Bacterial Vaccines , Clostridioides difficile , Clostridium Infections , Swine Diseases , Vaccines, Synthetic , Animals , Female , Swine , Rabbits , Clostridium Infections/prevention & control , Clostridium Infections/veterinary , Clostridium Infections/immunology , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Pregnancy , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Clostridioides difficile/immunology , Clostridioides difficile/genetics , Antibodies, Bacterial/blood , Bacterial Toxins/immunology , Bacterial Toxins/genetics , Swine Diseases/prevention & control , Swine Diseases/immunology , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Enterotoxins/immunology , Enterotoxins/geneticsABSTRACT
SARS-CoV-2 is the causative virus of COVID-19, which has been responsible for millions of deaths worldwide since its discovery. After its emergence, several variants have been identified that challenge the efficacy of the available vaccines. Previously, we generated and evaluated a vaccine based on a recombinant Bacillus Calmette-Guérin (rBCG) expressing the nucleoprotein (N) of SARS-CoV-2 (rBCG-N-SARS-CoV-2). This protein is a highly immunogenic antigen and well conserved among variants. Here, we tested the administration of this vaccine with recombinant N and viral Spike proteins (S), or Receptor Binding Domain (RBD-Omicron variant), plus a booster with the recombinant proteins only, as a novel and effective strategy to protect against SARS-CoV-2 variants. METHODS: BALB/c mice were immunized with rBCG-N-SARS-CoV-2 and recombinant SARS-CoV-2 proteins in Alum adjuvant, followed by a booster with recombinant proteins to assess the safety and virus-specific cellular and humoral immune responses against SARS-CoV-2 antigens. RESULTS: Immunization with rBCG-N-SARS-CoV-2 + recombinant proteins as a vaccine was safe and promoted the activation of CD4+ and CD8+ T cells that recognize SARS-CoV-2 N, S, and RBD antigens. These cells were able to secrete cytokines with an antiviral profile. This immunization strategy also induced robust titers of specific antibodies against N, S, and RBD and neutralizing antibodies of SARS-CoV-2. CONCLUSIONS: Co-administration of the rBCG-N-SARS-CoV-2 vaccine with recombinant SARS-CoV-2 proteins could be an effective alternative to control particular SARS-CoV-2 variants. Due to its safety and capacity to induce virus-specific immune responses, we believe the rBCG-N-SARS-CoV-2 + Proteins vaccine could be an attractive candidate to protect against this virus, especially in newborns.
Subject(s)
Antibodies, Viral , BCG Vaccine , COVID-19 Vaccines , COVID-19 , Mice, Inbred BALB C , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , Mice , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Antibodies, Viral/blood , Antibodies, Viral/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , COVID-19/prevention & control , COVID-19/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , BCG Vaccine/immunology , BCG Vaccine/administration & dosage , BCG Vaccine/genetics , Female , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Immunization, Secondary , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Immunity, Humoral , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Coronavirus Nucleocapsid Proteins/immunology , Coronavirus Nucleocapsid Proteins/genetics , CD8-Positive T-Lymphocytes/immunology , Phosphoproteins/immunology , Phosphoproteins/genetics , Adjuvants, Immunologic/administration & dosage , Immunity, CellularABSTRACT
Multivalent live-attenuated or inactivated vaccines are often used to control the bovine viral diarrhea disease (BVD). Still, they retain inherent disadvantages and do not provide the expected protection. This study developed a new vaccine prototype, including the external segment of the E2 viral protein from five different subgenotypes selected after a massive screening. The E2 proteins of every subgenotype (1aE2, 1bE2, 1cE2, 1dE2, and 1eE2) were produced in mammalian cells and purified by IMAC. An equimolar mixture of E2 proteins formulated in an oil-in-water adjuvant made up the vaccine candidate, inducing a high humoral response at 50, 100, and 150 µg doses in sheep. A similar immune response was observed in bovines at 50 µg. The cellular response showed a significant increase in the transcript levels of relevant Th1 cytokines, while those corresponding to the Th2 cytokine IL-4 and the negative control were similar. High levels of neutralizing antibodies against the subgenotype BVDV1a demonstrated the effectiveness of our vaccine candidate, similar to that observed in the sera of animals vaccinated with the commercial vaccine. These results suggest that our vaccine prototype could become an effective recombinant vaccine against the BVD.
Subject(s)
Antibodies, Viral , Bovine Virus Diarrhea-Mucosal Disease , Vaccines, Subunit , Vaccines, Synthetic , Viral Vaccines , Animals , Cattle , Viral Vaccines/immunology , Vaccines, Subunit/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Vaccines, Synthetic/immunology , Bovine Virus Diarrhea-Mucosal Disease/prevention & control , Bovine Virus Diarrhea-Mucosal Disease/immunology , Bovine Virus Diarrhea-Mucosal Disease/virology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Sheep , Viral Envelope Proteins/immunology , Viral Envelope Proteins/genetics , Cytokines/metabolism , Diarrhea Viruses, Bovine Viral/immunology , Diarrhea Viruses, Bovine Viral/genetics , Diarrhea Virus 1, Bovine Viral/immunology , Diarrhea Virus 1, Bovine Viral/geneticsABSTRACT
Vaccination is the best strategy to control Paratuberculosis (PTB), which is a significant disease in cattle and sheep. Previously we showed the humoral and cellular immune response induced by a novel vaccine candidate against PTB based on the Argentinian Mycobacterium avium subspecies paratuberculosis (Map) 6611 strain. To improve 6611 immunogenicity and efficacy, we evaluated this vaccine candidate in mice with two different adjuvants and a heterologous boost with a recombinant modified vaccinia Ankara virus (MVA) expressing the antigen 85A (MVA85A). We observed that boosting with MVA85A did not improve total IgG or specific isotypes in serum induced by one or two doses of 6611 formulated with incomplete Freund's adjuvant (IFA). However, when 6611 was formulated with ISA201 adjuvant, MVA85A boost enhanced the production of IFNγ, Th1/Th17 cytokines (IL-2, TNF, IL-17A) and IL-6, IL-4 and IL-10. Also, this group showed the highest levels of IgG2b and IgG3 isotypes, both important for better protection against Map infection in the murine model. Finally, the heterologous scheme elicited the highest levels of protection after Map challenge (lowest CFU count and liver lesion score). In conclusion, our results encourage further evaluation of 6611 strain + ISA201 prime and MVA85A boost in bovines.
Subject(s)
Adjuvants, Immunologic , Antibodies, Bacterial , Cytokines , Disease Models, Animal , Immunization, Secondary , Immunoglobulin G , Mycobacterium avium subsp. paratuberculosis , Paratuberculosis , Animals , Mycobacterium avium subsp. paratuberculosis/immunology , Immunization, Secondary/methods , Mice , Paratuberculosis/prevention & control , Paratuberculosis/immunology , Immunoglobulin G/blood , Cytokines/metabolism , Female , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Adjuvants, Immunologic/administration & dosage , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Mice, Inbred BALB C , Vaccinia virus/immunology , Vaccinia virus/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Immunity, Cellular/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Freund's Adjuvant/administration & dosage , Freund's Adjuvant/immunologyABSTRACT
Probiotic microorganisms can stimulate an immune response and increase the efficiency of vaccines. For example, Bacillus toyonensis is a nonpathogenic, Gram-positive bacterium that has been used as a probiotic in animal supplementation. It induces immunomodulatory effects and increases the vaccine response in several species. This study aimed to evaluate the effect of B. toyonensis supplementation on the modulation of the immune response in horses vaccinated with recombinant Clostridium tetani toxin. Twenty horses were vaccinated twice, with an interval of 21 days between doses, and equally divided into two groups: the first group was supplemented orally for 42 days with feed containing viable spores of B. toyonensis (1 × 108) mixed with molasses (40 ml), starting 7 days before the first vaccination; the second (control) group received only feed mixed with molasses, starting 7 days before the first vaccination. Serum samples were collected to evaluate the humoral immune response using an in-house indirect enzyme-linked immunosorbent assay (ELISA), and peripheral blood mononuclear cells (PBMCs) were collected to evaluate cytokine transcription (qPCR). For the specific IgG-anti-rTENT titer, the supplemented group had ELISA values that were four times higher than those of the control group (p < 0.05). The supplemented group also showed higher ELISA values for the IgGa and IgGT sub-isotypes compared to the control group. In PBMCs stimulated with B. toyonensis, relative cytokine transcription of the supplemented group showed 15-, 8-, 7-, and 6-fold increases for IL1, TNFα, IL10 and IL4, respectively. When stimulated with a vaccine antigen, the supplemented group showed 1.6-, 1.8-, and 0.5-fold increases in IL1, TNFα, and IL4, respectively, compared to the control group. Horses supplemented with B. toyonensis had a significantly improved vaccine immune response compared to those in the control group, which suggests a promising approach for improving vaccine efficacy with probiotics.
Subject(s)
Bacillus , Horse Diseases , Probiotics , Animals , Horses/immunology , Bacillus/immunology , Probiotics/administration & dosage , Probiotics/pharmacology , Horse Diseases/prevention & control , Horse Diseases/immunology , Horse Diseases/microbiology , Tetanus/prevention & control , Tetanus/immunology , Tetanus Toxoid/immunology , Tetanus Toxoid/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Male , Animal Feed , Female , Diet/veterinary , Cytokines/metabolismABSTRACT
Neosporosis is the major infectious cause of abortion and reproductive losses in cattle worldwide; however, there are no available vaccines or drugs to control this disease. Recently, a dual (positive and negative) DIVA-like (Differentiation of Infected from Vaccinated Animals) vaccine was evaluated in a pregnant mouse model of neosporosis, showing promising immunogenic and protective results. The current report aimed to study the safety, the dose-dependent immunogenicity and the dual DIVA-like character of a recombinant subunit vaccine composed of the major surface antigen from Neospora caninum (rNcSAG1) and the carrier/adjuvant Heat shock protein 81.2 from Arabidopsis thaliana (rAtHsp81.2) in cattle. Healthy heifers were separated and assigned to experimental groups A-F and subcutaneously immunized with 2 doses of vaccine formulations 30 days apart as follows: A (n = 4): 50 µg rNcSAG1 + 150 µg rAtHsp81.2; B (n = 4): 200 µg rNcSAG1 + 600 µg rAtHsp81.2; C (n = 4): 500 µg rNcSAG1 + 1,500 µg rAtHsp81.2; D (n = 3): 150 µg rAtHsp81.2; E (n = 3):1,500 µg rAtHsp81.2, and F (n = 3) 2 ml of sterile PBS. The immunization of heifers with the different vaccine or adjuvant doses (groups A-E) was demonstrated to be safe and did not modify the mean value of the evaluated serum biomarkers of metabolic function (GOT/ASP, GPT/ALT, UREA, Glucose and total proteins). The kinetics and magnitude of the immune responses were dose-dependent. The higher dose of the vaccine formulation (group C) stimulated a broad and potent humoral and cellular immune response, characterized by an IgG1/IgG2 isotype profile and IFN-γ secretion. In addition, this was the first time that dual DIVA-like character of a vaccine against neosporosis was demonstrated, allowing us to differentiate vaccinated from infected heifers by two different DIVA compliant test approaches. These results encourage us to evaluate its protective efficacy in infected pregnant cattle in the future.
Subject(s)
Cattle Diseases , Coccidiosis , Neospora , Protozoan Vaccines , Vaccines, Synthetic , Animals , Cattle , Coccidiosis/prevention & control , Coccidiosis/veterinary , Coccidiosis/immunology , Cattle Diseases/prevention & control , Cattle Diseases/immunology , Cattle Diseases/parasitology , Neospora/immunology , Female , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Protozoan Vaccines/immunology , Protozoan Vaccines/administration & dosage , Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Protozoan Proteins/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Immunogenicity, Vaccine , PregnancyABSTRACT
COVID-19 manifests as a milder disease in children than adults, but the underlying mechanisms are not fully characterized. Here we assess the difference in cellular or humoral immune responses of pediatric and adult COVID-19 patients to see if these factors contribute to the severity dichotomy. Children's non-specific immune profile is dominated by naive lymphocytes and HLA-DRhighCX3CR1low dendritic cells; meanwhile, children show strong specific antibody and T cell responses for viral structural proteins, with their T cell responses differing from adults by having weaker CD8+TNF+ T cells responses to S peptide pool but stronger responses to N and M peptide pools. Finally, viral mRNA is more abundant in pediatric patients. Our data thus support a scenario in which SARS-CoV-2 infected children contribute to transmission yet are less susceptible to COVID-19 symptoms due to strong and differential responses to the virus.
Subject(s)
Antibodies, Viral/immunology , COVID-19/immunology , Immunity, Humoral , RNA, Viral , SARS-CoV-2/genetics , Vaccines, Synthetic/immunology , Adolescent , Adult , Aged , Antibodies, Viral/blood , Brazil , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes , COVID-19/prevention & control , COVID-19 Vaccines , Child , Child, Preschool , Cytokines/blood , Female , Humans , Immunity, Innate , Male , Middle Aged , RNA, Messenger , Spike Glycoprotein, Coronavirus/immunology , T-Lymphocytes , Viral Structural Proteins/immunology , Young Adult , mRNA VaccinesSubject(s)
COVID-19 Vaccines/administration & dosage , COVID-19/virology , SARS-CoV-2/isolation & purification , Antigens, Viral/genetics , Antigens, Viral/immunology , COVID-19/immunology , COVID-19/prevention & control , COVID-19/transmission , COVID-19 Vaccines/immunology , Humans , Mutation , SARS-CoV-2/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , mRNA VaccinesABSTRACT
Massive vaccination offers great promise for halting the global COVID-19 pandemic. However, the limited supply and uneven vaccine distribution create an urgent need to optimize vaccination strategies. We evaluate SARS-CoV-2-specific antibody responses after Sputnik V vaccination of healthcare workers in Argentina, measuring IgG anti-spike titers and neutralizing capacity after one and two doses in a cohort of naive or previously infected volunteers. By 21 days after receiving the first dose of the vaccine, 94% of naive participants develop spike-specific IgG antibodies. A single Sputnik V dose elicits higher antibody levels and virus-neutralizing capacity in previously infected individuals than in naive ones receiving the full two-dose schedule. The high seroconversion rate after a single dose in naive participants suggests a benefit of delaying administration of the second dose to increase the number of people vaccinated. The data presented provide information for guiding public health decisions in light of the current global health emergency.
Subject(s)
COVID-19 Vaccines/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , Vaccines, Synthetic/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Argentina/epidemiology , COVID-19/immunology , Chlorocebus aethiops , HEK293 Cells , Health Personnel , Humans , Pandemics , SARS-CoV-2/pathogenicity , Seroconversion , Spike Glycoprotein, Coronavirus/immunology , Vaccination , Vaccines , Vero CellsABSTRACT
mRNA vaccines against SARS-CoV-2 are remarkably effective. Limited information exists about the incidence of adverse events following immunization (AEFI) with their use. We conducted a prospective observational study including data from 704,003 first-doses recipients; 6536 AEFI were reported, of whom 65.1% had at least one neurologic AEFI (non-serious 99.6%). Thirty-three serious events were reported; 17 (51.5%) were neurologic (observed frequency, 2.4/100,000 doses). At the time of writing this report, 16/17 cases had been discharged without deaths. Our data suggest that the BNT162b2 mRNA COVID-19 vaccine is safe; its individual and societal benefits outweigh the low percentage of serious neurologic AEFI. This information should help to dissipate hesitancy towards this new vaccine platform.
Subject(s)
COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/immunology , COVID-19/prevention & control , Nervous System Diseases/etiology , SARS-CoV-2 , Adult , BNT162 Vaccine , COVID-19/epidemiology , Cohort Studies , Female , Humans , Male , Mexico/epidemiology , Middle Aged , Nervous System Diseases/epidemiology , Prospective Studies , Vaccines, Synthetic/immunology , mRNA VaccinesSubject(s)
Humans , COVID-19 Vaccines/administration & dosage , SARS-CoV-2/isolation & purification , COVID-19/virology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , COVID-19 Vaccines/immunology , SARS-CoV-2/genetics , COVID-19/immunology , COVID-19/prevention & control , COVID-19/transmission , Mutation , Antigens, Viral/genetics , Antigens, Viral/immunologyABSTRACT
Entamoeba histolytica is an intestinal parasite that causes dysentery and amebic liver abscess. E. histolytica has the capability to invade host tissue by union of virulence factor Gal/GalNAc lectin; this molecule induces an adherence-inhibitory antibody response as well as to protect against amebic liver abscess (ALA). The present work showed the effect of the immunization with PEΔIII-LC3-KDEL3 recombinant protein. In vitro, this candidate vaccine inhibited adherence of E. histolytica trophozoites to HepG2 cell monolayer, avoiding the cytolysis, and in a hamster model, we observed a vaccine-induced protection against the damage to tissue liver and the inhibition of uncontrolled inflammation. PEΔIII-LC3-KDEL3 reduced the expression of TNF-α, IL-1ß, and NF-κB in all immunized groups at 4- and 7-day postinfection. The levels of IL-10, FOXP3, and IFN-γ were elevated at 7 days. The immunohistochemistry assay confirmed this result, revealing an elevated quantity of +IFN-γ cells in the liver tissue. ALA formation in hamsters immunized was minimal, and few trophozoites were identified. Hence, immunization with PEΔIII-LC3-KDEL3 herein prevented invasive amebiasis, avoided an acute proinflammatory response, and activated a protective response within a short time. Finally, this recombinant protein induced an increase of serum IgG.
Subject(s)
Entamoeba histolytica/immunology , Liver Abscess, Amebic/prevention & control , Protozoan Proteins/administration & dosage , Protozoan Vaccines/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Animals , Antibodies, Protozoan/blood , Disease Models, Animal , Entamoeba histolytica/genetics , Humans , Immunogenicity, Vaccine , Lectins/genetics , Lectins/immunology , Liver/immunology , Liver/parasitology , Liver/pathology , Liver Abscess, Amebic/blood , Liver Abscess, Amebic/parasitology , Liver Abscess, Amebic/pathology , Male , Mesocricetus , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Protozoan Vaccines/genetics , Protozoan Vaccines/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunologyABSTRACT
H65, a fusion protein of three pairs of ESX-secreted antigens of Mycobacterium tuberculosis and Mycobacterium bovis, formulated with the liposomal adjuvant CAF01 has been shown to confer protection against M. tuberculosis infection in mice. In this study, we evaluated the impact of combining BCG with H65 + CAF01 immunization in a M. bovis mouse model of infection. We found that a BCG-H65 + CAF01/ H65 + CAF01 prime-boost scheme induced higher protection than BCG and H65 + CAF01 alone. Altogether, H65 antigen formulated in liposomal adjuvant improved the BCG-induced immune protection, thus making this vaccine strategy a promising tool to control bovine tuberculosis.
Subject(s)
BCG Vaccine/immunology , Mycobacterium bovis/immunology , Tuberculosis, Bovine/prevention & control , Adjuvants, Immunologic/pharmacology , Animals , Cattle , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , Mycobacterium tuberculosis/immunology , Tuberculosis, Bovine/immunology , Vaccines, Subunit/immunology , Vaccines, Synthetic/immunologyABSTRACT
BACKGROUND: In recent years, there has been great interest in developing molecular adjuvants based on antisense oligonucleotides (ASOs) targeting immunosuppressor pathways with inhibitory effects on regulatory T cells (Tregs) to improve immunogenicity and vaccine efficacy. We aim to evaluate the immunostimulating effect of 2'OMe phosphorothioated Foxp3-targeted ASO in an antifungal adjuvanted recombinant vaccine. METHODS: The uptake kinetics of Foxp3 ASO, its cytotoxicity and its ability to deplete Tregs were evaluated in murine splenocytes in vitro. Groups of mice were vaccinated with recombinant enolase (Eno) of Sporothix schenckii in Montanide Gel 01 adjuvant alone or in combination with either 1 µg or 8 µg of Foxp3 ASO. The titers of antigen-specific antibody in serum samples from vaccinated mice (male C57BL/6) were determined by ELISA (enzyme-linked immunosorbent assay). Cultured splenocytes from each group were activated in vitro with Eno and the levels of IFN-γ and IL-12 were also measured by ELISA. The results showed that the anti-Eno antibody titer was significantly higher upon addition of 8 µM Foxp3 ASO in the vaccine formulation compared to the standard vaccine without ASO. In vitro and in vivo experiments suggest that Foxp3 ASO enhances specific immune responses by means of Treg depletion during vaccination. CONCLUSION: Foxp3 ASO significantly enhances immune responses against co-delivered adjuvanted recombinant Eno vaccine and it has the potential to improve vaccine immunogenicity.
Subject(s)
Forkhead Transcription Factors/genetics , Gene Silencing , Immunogenicity, Vaccine , Oligonucleotides, Antisense/chemistry , Sporothrix/immunology , Vaccines, Synthetic/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Pharmaceutic , Animals , Immune System , Interferon-gamma/metabolism , Interleukin-12 Subunit p35/metabolism , Kinetics , Male , Mice , Mice, Inbred C57BL , Spleen/cytology , T-Lymphocytes, Regulatory/metabolismABSTRACT
AIM: The aim of the present study was to examine the vaccine immune response in ewes supplemented with Bacillus toyonensis BCT-7112T during a period of 5-day supplementation before vaccination against a recombinant Clostridium perfringens epsilon toxin (rETX). METHODS AND RESULTS: Ewes were vaccinated with 200 µg of rETX adjuvanted with 10% aluminium hydroxide. The treat group was orally supplemented with B. toyonensis BCT-7112T (3 × 108 viable spores) for 5 days prior to the first and second vaccination. Ewes supplemented with B. toyonensis BCT-7112T showed higher neutralizing antibody titres than the non-supplemented ewes (P < 0·05), with an increase in serum levels for total IgG anti-rETX by 3·2-fold (P < 0·0001), and for both IgG isotypes IgG1 and IgG2 by 2·1-fold and 2·3-fold (P < 0·01), respectively, compared with the control group. The peripheral blood mononuclear cells of ewes in the supplemented group had a higher (P < 0·05) cytokine mRNA transcription levels for IL-2 (6·4-fold increase), IFN-γ (2·9-fold increase) and transcription factor Bcl6 (2·3-fold increase) compared with the control group. CONCLUSION: We conclude that a 5 days of supplementation with B. toyonensis BCT-7112T prior vaccination is sufficient to significantly improve the humoral immune response of ewes against C. perfringens recombinant ETX vaccine. SIGNIFICANCE AND IMPACT OF THE STUDY: These findings open a new perspective in the utilization of B. toyonensis BCT-7112T as an immunomodulator since a 5 days period of probiotic supplementation is sufficient to improve the vaccine immune response.
Subject(s)
Bacillus , Bacterial Toxins/immunology , Bacterial Vaccines/immunology , Probiotics , Sheep/immunology , Animals , Bacterial Toxins/genetics , Bacterial Vaccines/genetics , Female , Immunity, Humoral , Immunoglobulin G/blood , Immunomodulation , Leukocytes, Mononuclear/immunology , Vaccination , Vaccines, Synthetic/immunologySubject(s)
Humans , Animals , Female , Adult , Middle Aged , Young Adult , Pneumonia, Viral/prevention & control , Vaccines, Synthetic/immunology , Coronavirus Infections/prevention & control , Pneumonia, Viral/immunology , Vaccines, Synthetic/adverse effects , Randomized Controlled Trials as Topic , Immunization, Secondary , Clinical Trials, Phase III as Topic , Coronavirus Infections/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Injections, Intramuscular , Moscow , Antibodies, Viral/bloodABSTRACT
Background: Our previous work has demonstrated the benefits of transcutaneous immunization in targeting Langerhans cells and preferentially inducing CD8 T-cell responses. Methods: In this randomized phase Ib clinical trial including 20 HIV uninfected volunteers, we compared the safety and immunogenicity of the MVA recombinant vaccine expressing HIV-B antigen (MVA-B) by transcutaneous and intramuscular routes. We hypothesized that the quality of innate and adaptive immunity differs according to the route of immunization and explored the quality of the vector vaccine-induced immune responses. We also investigated the early blood transcriptome and serum cytokine levels to identify innate events correlated with the strength and quality of adaptive immunity. Results: We demonstrate that MVA-B vaccine is safe by both routes, but that the quality and intensity of both innate and adaptive immunity differ significantly. Transcutaneous vaccination promoted CD8 responses in the absence of antibodies and slightly affected gene expression, involving mainly genes associated with metabolic pathways. Intramuscular vaccination, on the other hand, drove robust changes in the expression of genes involved in IL-6 and interferon signalling pathways, mainly those associated with humoral responses, and also some levels of CD8 response. Conclusion: Thus, vaccine delivery route perturbs early innate responses that shape the quality of adaptive immunity. Clinical Trial Registration: http://ClinicalTrials.gov, identifier PER-073-13.