Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Vaccines (Basel) ; 12(5)2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38793733

RESUMO

COVID-19 vaccination strategies, including heterologous prime-boost regimens and additional booster doses, aim to optimize immune responses. However, seroepidemiological studies on immune responses to different COVID-19 vaccine types and schedules remain limited. This study investigated antibody levels following homologous and heterologous prime-and-boost COVID-19 vaccination in Bangladesh. In a cohort of 606 participants who received first/second/booster doses of vaccines (AstraZeneca, Moderna, Pfizer-BioNTech, and Sinopharm), anti-spike IgG and anti-nucleocapsid IgG levels were measured. Antibody titer variations with respect to age, gender, intervals between doses, and prior infection status were analyzed. mRNA vaccines elicited the highest antibody levels after homologous and heterologous boosting. The AstraZeneca booster resulted in a sharp titer decline rate of ~0.04 units per day. Second or booster vaccine doses significantly increased antibody levels, especially in males (p < 0.05). Older age correlated with higher titers, likely reflecting previous infection, which was further confirmed by the elevation of anti-nucleocapsid IgG levels. About 95.5% of non-Sinopharm recipients were anti-nucleocapsid IgG positive, suggesting prior exposure exceeding self-reported infections (12.5%). mRNA and heterologous COVID-19 boosting enhances humoral immunity over homologous prime-boost vector/inactivated vaccination. However, waning immunity merits further investigation across vaccine platforms.

2.
EBioMedicine ; 90: 104523, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36933409

RESUMO

BACKGROUND: The tick-borne bunyavirus, Crimean-Congo Haemorrhagic Fever virus (CCHFV), can cause severe febrile illness in humans and has a wide geographic range that continues to expand due to tick migration. Currently, there are no licensed vaccines against CCHFV for widespread usage. METHODS: In this study, we describe the preclinical assessment of a chimpanzee adenoviral vectored vaccine (ChAdOx2 CCHF) which encodes the glycoprotein precursor (GPC) from CCHFV. FINDINGS: We demonstrate here that vaccination with ChAdOx2 CCHF induces both a humoral and cellular immune response in mice and 100% protection in a lethal CCHF challenge model. Delivery of the adenoviral vaccine in a heterologous vaccine regimen with a Modified Vaccinia Ankara vaccine (MVA CCHF) induces the highest levels of CCHFV-specific cell-mediated and antibody responses in mice. Histopathological examination and viral load analysis of the tissues of ChAdOx2 CCHF immunised mice reveals an absence of both microscopic changes and viral antigen associated with CCHF infection, further demonstrating protection against disease. INTERPRETATION: There is the continued need for an effective vaccine against CCHFV to protect humans from lethal haemorrhagic disease. Our findings support further development of the ChAd platform expressing the CCHFV GPC to seek an effective vaccine against CCHFV. FUNDING: This research was supported by funding from the Biotechnology and Biological Sciences Research Council (UKRI-BBSRC) [BB/R019991/1 and BB/T008784/1].


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo , Febre Hemorrágica da Crimeia , Vacinas Virais , Humanos , Animais , Camundongos , Febre Hemorrágica da Crimeia/prevenção & controle , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Vacinação , Vetores Genéticos/genética , Vaccinia virus
3.
Nanomedicine ; 48: 102648, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36584738

RESUMO

Recent advances in bioinformatics and nanotechnology offer great opportunities for personalized cancer vaccine development. However, the timely identification of neoantigens and unsatisfactory efficacy of therapeutic cancer vaccines remain two obstacles for clinical transformation. We propose a "prime and boost" strategy to facilitate neoantigen-based immunotherapy. To prime the immune system, we first constructed personalized liposomes with cancer cell membranes and adjuvant R848 to provide immunostimulatory efficacy and time for identifying tumor antigens. Liposomes loaded with personalized neopeptides and adjuvants were used to boost the immune response. In vitro experiments verified potent immune responses, including macrophage polarization, dendritic cell maturation, and T lymphocyte activation. In vivo B16F10 and TC-1 cancer model were used to investigate efficient tumor growth suppression. Liposomal vaccines with neopeptides could stimulate human dendritic cells and T lymphocytes in vitro. These results demonstrate that the "prime and boost" strategy provides simple, quick, and efficient personalized vaccines for cancer therapy.


Assuntos
Vacinas Anticâncer , Neoplasias , Humanos , Lipossomos , Neoplasias/terapia , Linfócitos T , Antígenos de Neoplasias , Adjuvantes Imunológicos/farmacologia , Membrana Celular , Imunoterapia/métodos
4.
Front Immunol ; 13: 940715, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36177016

RESUMO

The world has responded to the COVID-19 pandemic with unprecedented speed and vigor in the mass vaccination campaigns, targeted to reduce COVID-19 severity and mortality, reduce the pressure on the healthcare system, re-open society, and reduction in disease mortality and morbidity. Here we review the preclinical and clinical development of BBV152, a whole virus inactivated vaccine and an important tool in the fight to control this pandemic. BBV152, formulated with a TLR7/8 agonist adjuvant generates a Th1-biased immune response that induces high neutralization efficacy against different SARS-CoV-2 variants of concern and robust long-term memory B- and T-cell responses. With seroconversion rates as high as 98.3% in vaccinated individuals, BBV152 shows 77.8% and 93.4% protection from symptomatic COVID-19 disease and severe symptomatic COVID-19 disease respectively. Studies in pediatric populations show superior immunogenicity (geometric mean titer ratio of 1.76 compared to an adult) with a seroconversion rate of >95%. The reactogenicity and safety profiles were comparable across all pediatric age groups between 2-18 yrs. as in adults. Like most approved vaccines, the BBV152 booster given 6 months after full vaccination, reverses a waning immunity, restores the neutralization efficacy, and shows synergy in a heterologous prime-boost study with about 3-fold or 300% increase in neutralization titers against multiple SARS-CoV-2 variants of concern. Based on the interim Phase III data, BBV152 received full authorization for adults and emergency use authorization for children from ages 6 to 18 years in India. It is also licensed for emergency use in 14 countries globally. Over 313 million vaccine doses have already been administered in India alone by April 18th, 2022.


Assuntos
COVID-19 , SARS-CoV-2 , Adjuvantes Imunológicos , Adolescente , Adulto , COVID-19/prevenção & controle , Vacinas contra COVID-19/efeitos adversos , Criança , Pré-Escolar , Humanos , Pandemias/prevenção & controle , Receptor 7 Toll-Like , Desenvolvimento de Vacinas , Vacinas de Produtos Inativados/efeitos adversos
5.
EBioMedicine ; 80: 104036, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35500538

RESUMO

BACKGROUND: Schistosomiasis is an underestimated neglected tropical disease which affects over 236.6 million people worldwide. According to the CDC, the impact of this disease is second to only malaria as the most devastating parasitic infection. Affected individuals manifest chronic pathology due to egg granuloma formation, destroying the liver over time. The only FDA approved drug, praziquantel, does not protect individuals from reinfection, highlighting the need for a prophylactic vaccine. Schistosoma mansoni Cathepsin B (SmCB) is a parasitic gut peptidase necessary for helminth growth and maturation and confers protection as a vaccine target for intestinal schistosomiasis. METHODS: An SmCB expressing human adenovirus serotype 5 (AdSmCB) was constructed and delivered intramuscularly to female C57BL/6 mice in a heterologous prime and boost vaccine with recombinant protein. Vaccine induced immunity was described and subsequent protection from parasite infection was assessed by analysing parasite burden and liver pathology. FINDINGS: Substantially higher humoral and cell-mediated immune responses, consisting of IgG2c, Th1 effectors, and polyfunctional CD4+ T cells, were induced by the heterologous administration of AdSmCB when compared to the other regimens. Though immune responses favoured Th1 immunity, Th2 responses provided by SmCB protein boosts were maintained. This mixed Th1/Th2 immune response resulted in significant protection from S. mansoni infection comparable to other vaccine formulations which are in clinical trials. Schistosomiasis associated liver pathology was also prevented in a murine model. INTERPRETATION: Our study provides missing preclinical data supporting the use of adenoviral vectoring in vaccines for S. mansoni infection. Our vaccination method significantly reduces parasite burden and its associated liver pathology - both of which are critical considerations for this helminth vaccine. FUNDING: This work was supported by the Canadian Institutes of Health Research, R. Howard Webster Foundation, and the Foundation of the McGill University Health Centre.


Assuntos
Vacinas contra Adenovirus , Esquistossomose mansoni , Esquistossomose , Vacinas , Adenoviridae/genética , Animais , Anticorpos Anti-Helmínticos , Antígenos de Helmintos , Canadá , Catepsina B/genética , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Schistosoma mansoni/genética , Esquistossomose mansoni/prevenção & controle
6.
Lett Appl Microbiol ; 69(4): 258-263, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31278766

RESUMO

Porcine epidemic diarrhoea virus (PEDV) causes severe diarrhoea in neonatal suckling piglets with a high mortality. Maternal vaccines that can induce lactogenic immunity to protect suckling piglets via colostrums and milk are pivotal for the prevention and control of PEDV infection in neonatal suckling piglets. In this study, a group of pregnant sows were first orally immunized with coated PEDV-loaded microspheres and boosted with killed PEDV vaccines (heterologous prime-boost). It has been detected that the levels of PEDV-specific antibodies (IgG and IgA) in their sera and milks were higher than other negative groups (P < 0·001 or P < 0·05). Furthermore, it has been proved by the neutralization assay that the induced antibodies could significantly inhibit virus infection as compared to other negative groups (P < 0·01 or P < 0·05). Importantly, after PEDV challenge, more than 90% of the suckling piglets delivered by the sows in the heterologous prime-boost group were completely protected. Overall, the results show that 'heterologous prime-boost' form is an efficient and effective way to provide protection for suckling piglets against PEDV through lactogenic immunity. SIGNIFICANCE AND IMPACT OF THE STUDY: As a widespread swine pathogen, PEDV affects the swine industry enormously. It causes enteritis in swine of all ages and is often fatal in neonatal piglets. Our data show that pregnant sows were immunized with 'coated PEDV-loaded microspheres + killed PEDV vaccines' (heterologous prime-boost immunization) could protect more than 90% suckling piglets delivered by the sows against the virus. These findings provide a new model of developing safe and effective immunizations for newborn animals against established and emerging enteric infections.


Assuntos
Anticorpos Antivirais/sangue , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/veterinária , Vírus da Diarreia Epidêmica Suína/imunologia , Doenças dos Suínos/prevenção & controle , Animais , Infecções por Coronavirus/virologia , Diarreia/prevenção & controle , Diarreia/veterinária , Diarreia/virologia , Feminino , Leite , Gravidez , Suínos , Doenças dos Suínos/virologia , Vacinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA