Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.349
Filtrar
1.
Nano Lett ; 24(42): 13118-13125, 2024 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-39365010

RESUMEN

Peptide vaccines induce specific neutralizing antibodies and are effective in disease prevention and treatment. However, peptide antigens have a low immunogenicity and are unstable, requiring efficient vaccine carriers to enhance their immunogenicity. Here, we develop a tobacco mosaic virus (TMV)-based peptide vaccine for transdermal immunization using a tip-loaded dissolving microneedle (MN) patch. TMV is decorated with the model peptide antigen PEP3. The prepared TMV-PEP3 promotes dendritic cell maturation and induces dendritic cells to overexpress MHC II, costimulatory factors, and pro-inflammatory factors. By encapsulation of TMV-PEP3 in the tips of a trehalose MN, TMV-PEP3 can be delivered by MN and significantly promote local immune cell infiltration. In vivo studies show that both subcutaneous injection and MN administration of TMV-PEP3 increase the production of anti-PEP3 IgG antibodies and the harvested serum can induce complement-dependent cytotoxicity. This work provides a promising strategy for constructing efficient and health-care-friendly peptide vaccines.


Asunto(s)
Administración Cutánea , Células Dendríticas , Agujas , Virus del Mosaico del Tabaco , Vacunas de Subunidad , Animales , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Ratones , Virus del Mosaico del Tabaco/inmunología , Virus del Mosaico del Tabaco/química , Células Dendríticas/inmunología , Inmunización , Péptidos/química , Péptidos/inmunología , Vacunas de Subunidades Proteicas
2.
Front Immunol ; 15: 1441944, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39381003

RESUMEN

Introduction: Development of an effective vaccine against tuberculosis is a critical step towards reducing the global burden of disease. A therapeutic vaccine might also reduce the high rate of TB recurrence and help address the challenges of drug-resistant strains. ID93+GLA-SE is a candidate subunit vaccine that will soon be evaluated in a phase 2b efficacy trial for prevention of recurrent TB among patients undergoing TB treatment. ID93+GLA-SE vaccination was shown to elicit robust CD4+ T cell and IgG antibody responses among recently treated TB patients in the TBVPX-203 Phase 2a study (NCT02465216), but the mechanisms underlying these responses are not well understood. Methods: In this study we used specimens from TBVPX-203 participants to describe the changes in peripheral blood gene expression that occur after ID93+GLA-SE vaccination. Results: Analyses revealed several distinct modules of co-varying genes that were either up- or down-regulated after vaccination, including genes associated with innate immune pathways at 3 days post-vaccination and genes associated with lymphocyte expansion and B cell activation at 7 days post-vaccination. Notably, the regulation of these gene modules was affected by the dose schedule and by participant sex, and early innate gene signatures were correlated with the ID93-specific CD4+ T cell response. Discussion: The results provide insight into the complex interplay of the innate and adaptive arms of the immune system in developing responses to vaccination with ID93+GLA-SE and demonstrate how dosing and schedule can affect vaccine responses.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Vacunas contra la Tuberculosis , Vacunación , Humanos , Femenino , Masculino , Vacunas contra la Tuberculosis/inmunología , Vacunas contra la Tuberculosis/administración & dosificación , Adulto , Tuberculosis/prevención & control , Tuberculosis/inmunología , Mycobacterium tuberculosis/inmunología , Linfocitos T CD4-Positivos/inmunología , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación
3.
Avian Dis ; 68(3): 183-191, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39400212

RESUMEN

Fowl cholera, caused by Pasteurella multocida infection, poses challenges for prevention because of its many serotypes. Bacterins are currently widely used for vaccination against fowl cholera, but protection is limited to homologous strains. Live attenuated vaccines of P. multocida provide some heterologous protection, but side effects are considerable. More recently, protein-based antigens are promising subunit vaccines when their low immunogenicity has been addressed with effective adjuvants. Bacterial flagellin has been widely considered a promising adjuvant for vaccines. In this study, we tested the adjutancy of flagellin in a subunit vaccine against P. multocida in a mice and chicken models. For vaccine formulation, the antigen fPlpE (P. multocida liporotein E) was combined with fFliC (Salmonella Typhimurium flagellin). The recombinant proteins of fPlpE and fFliC were successfully expressed using the Escherichia coli system as the expected sizes of 55 kDa and 70 kDa, respectively. The fFliC elicited strong expression levels of proinflammatory cytokine (IL-1ß, IL-8, and IL-6) when stimulated in native chicken peripheral blood mononuclear cells. Immunization of mice and chickens with the subunit vaccines containing fFliC accelerated the antibody response. In the challenge tests, fFliC increased vaccine protective efficacy against the heterologous strain P. multocida A1 and highly virulent strain Chu01 in mice and chickens, respectively. These data indicated potential possibilities of using fFliC as an immunostimulant adjuvant in developing a subunit vaccine against fowl cholera.


Nota de Investigación- La flagelina aumenta la inmunogenicidad de una vacuna subunitaria que contiene lipoprote'ina E de Pasteurella multocida. La enfermedad del cólera aviar, causada por la infección por Pasteurella multocida, representa desaf'ios para la prevención debido a sus numerosos serotipos. Actualmente, las bacterinas se utilizan ampliamente para la vacunación contra el cólera aviar, pero la protección se limita a cepas homólogas. Las vacunas vivas atenuadas de P. multocida proporcionan cierta protección heteróloga, pero los efectos secundarios son considerables. Recientemente, el uso de ant'igenos basados en prote'inas parece ser prometedor como vacunas subunitarias cuando se ha abordado el problema de su baja inmunogenicidad con adyuvantes eficaces. La flagelina bacteriana se ha considerado ampliamente como un adyuvante prometedor para las vacunas. En este estudio, se analizó la capacidad como adyuvante de la flagelina en una vacuna de subunitaria contra P. multocida en modelos de ratones y pollos. Para la formulación de la vacuna, el ant'igeno fPlpE (liporote'ina E de P. multocida) se combinó con fFliC (flagelina de Salmonella Typhimurium). Las prote'inas recombinantes de fPlpE y fFliC se expresaron con éxito utilizando el sistema de E. coli con los tamaños esperados de 55 kDa y 70 kDa, respectivamente. El fFliC indujo fuertes niveles de expresión de citoquinas proinflamatorias (IL-1ß, IL-8 e IL-6) cuando se estimuló en células mononucleares de sangre periférica de pollos nativos. La inmunización de ratones y pollos con las vacunas subunitarias que contienen fFliC aceleró la respuesta de anticuerpos. En las pruebas de desaf'io, la fFliC aumentó la eficacia protectora de la vacuna contra la cepa heteróloga de P. multocida A1 y la cepa altamente virulenta Chu01 en ratones y pollos, respectivamente. Estos datos indicaron posibilidades potenciales de utilizar fFliC como adyuvante inmunoestimulante en el desarrollo de una vacuna subunitaria contra el cólera aviar.


Asunto(s)
Vacunas Bacterianas , Pollos , Flagelina , Infecciones por Pasteurella , Pasteurella multocida , Enfermedades de las Aves de Corral , Vacunas de Subunidad , Pasteurella multocida/inmunología , Animales , Flagelina/inmunología , Infecciones por Pasteurella/veterinaria , Infecciones por Pasteurella/prevención & control , Infecciones por Pasteurella/inmunología , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/administración & dosificación , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Ratones , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/microbiología , Lipoproteínas/inmunología , Lipoproteínas/genética , Femenino , Ratones Endogámicos BALB C , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología
4.
Eur J Pharm Biopharm ; 204: 114528, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39383977

RESUMEN

In recent decades, protein-based therapy has garnered valid attention for treating infectious diseases, genetic disorders, cancer, and other clinical requirements. However, preserving protein-based drugs against degradation and denaturation during processing, storage, and delivery poses a formidable challenge. Herein, we designed a novel fluoroamphiphiles polymer to deliver protein. Two different formulations of nanoparticles, cross-linked (CNP) and micelle (MNP) polymer, were prepared rationally by disulfide cross-linked and thin-film hydration techniques, respectively. The size, zeta potential, and morphology of both formulations were characterized and the delivery efficacy of both in vitro and in vivo was also assessed. The in vitro findings demonstrated that both formulations effectively facilitated protein delivery into various cell lines. Moreover, in vivo experiments revealed that intramuscular administration of the two formulations loaded with a SARS-CoV-2 recombinant receptor-binding domain (RBD) vaccine induced robust antibody responses in mice without adding another adjuvant. These results highlight the potential use of our carrier system as a safe and effective platform for the in vivo delivery of subunit vaccines.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Ratones Endogámicos BALB C , Micelas , Nanopartículas , SARS-CoV-2 , Vacunas de Subunidad , Animales , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/administración & dosificación , Vacunas contra la COVID-19/química , Ratones , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , COVID-19/prevención & control , COVID-19/inmunología , Humanos , SARS-CoV-2/inmunología , SARS-CoV-2/efectos de los fármacos , Nanopartículas/química , Femenino , Polímeros/química , Anticuerpos Antivirales/inmunología , Portadores de Fármacos/química
5.
Int J Nanomedicine ; 19: 9757-9770, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39318604

RESUMEN

Background: The global prevalence of Dengue virus (DENV) infection poses a significant health risk, urging the need for effective vaccinations. Peptide vaccines, known for their capacity to induce comprehensive immunity against multiple virus serotypes, offer promise due to their stability, safety, and design flexibility. Spherical nucleic acid (SNA), particularly those with gold nanoparticle cores, present an attractive avenue for enhancing peptide vaccine efficacy due to their modularity and immunomodulatory properties. Methods: The spherical nucleic acid-TBB (SNA-TBB), a novel nanovaccine construct, was fabricated through the co-functionalization process of SNA with epitope peptide, targeting all four serotypes of the DENV. This innovative approach aims to enhance immunogenicity and provide broad-spectrum protection against DENV infections. The physicochemical properties of SNA-TBB were characterized using dynamic light scattering, zeta potential measurement, and transmission electron microscopy. In vitro assessments included endocytosis studies, cytotoxicity evaluation, bone marrow-dendritic cells (BMDCs) maturation and activation analysis, cytokine detection, RNA sequencing, and transcript level analysis in BMDCs. In vivo immunization studies in mice involved evaluating IgG antibody titers, serum protection against DENV infection and safety assessment of nanovaccines. Results: SNA-TBB demonstrated successful synthesis, enhanced endocytosis, and favorable physicochemical properties. In vitro assessments revealed no cytotoxicity and promoted BMDCs maturation. Cytokine analyses exhibited heightened IL-12p70, TNF-α, and IL-1ß levels. Transcriptomic analysis highlighted genes linked to BMDCs maturation and immune responses. In vivo studies immunization with SNA-TBB resulted in elevated antigen-specific IgG antibody levels and conferred protection against DENV infection in neonatal mice. Evaluation of in vivo safety showed no signs of adverse effects in vital organs. Conclusion: The study demonstrates the successful development of SNA-TBB as a promising nanovaccine platform against DENV infection and highlights the potential of SNA-based peptide vaccines as a strategy for developing safe and effective antiviral immunotherapy.


Asunto(s)
Células Dendríticas , Vacunas contra el Dengue , Virus del Dengue , Dengue , Animales , Virus del Dengue/inmunología , Ratones , Dengue/prevención & control , Dengue/inmunología , Vacunas contra el Dengue/inmunología , Vacunas contra el Dengue/química , Vacunas contra el Dengue/administración & dosificación , Células Dendríticas/inmunología , Presentación de Antígeno , Péptidos/química , Péptidos/farmacología , Péptidos/inmunología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/química , Vacunas de Subunidad/administración & dosificación , Humanos , Nanopartículas del Metal/química , Oro/química , Femenino , Citocinas/metabolismo , Ácidos Nucleicos/química , Ácidos Nucleicos/inmunología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre
6.
Sci Immunol ; 9(99): eadl3755, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39303017

RESUMEN

Prolonging exposure to subunit vaccines during the primary immune response enhances humoral immunity. Escalating-dose immunization (EDI), administering vaccines every other day in an increasing pattern over 2 weeks, is particularly effective but challenging to implement clinically. Here, using an HIV Env trimer/saponin adjuvant vaccine, we explored simplified EDI regimens and found that a two-shot regimen administering 20% of the vaccine followed by the remaining 80% of the dose 7 days later increased TFH responses 6-fold, antigen-specific germinal center (GC) B cells 10-fold, and serum antibody titers 10-fold compared with bolus immunization. Computational modeling of TFH priming and the GC response suggested that enhanced activation/antigen loading on dendritic cells and increased capture of antigen delivered in the second dose by follicular dendritic cells contribute to these effects, predictions we verified experimentally. These results suggest that a two-shot priming approach can be used to substantially enhance responses to subunit vaccines.


Asunto(s)
Centro Germinal , Inmunidad Humoral , Centro Germinal/inmunología , Animales , Inmunidad Humoral/inmunología , Ratones , Femenino , Vacunas contra el SIDA/inmunología , Vacunas contra el SIDA/administración & dosificación , Ratones Endogámicos C57BL , Linfocitos B/inmunología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación
7.
Sci Rep ; 14(1): 20579, 2024 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-39242614

RESUMEN

During COVID-19 pandemic, cases of postvaccination infections and restored SARS-CoV-2 virus have increased after full vaccination, which might be contributed to by immune surveillance escape or virus rebound. Here, artificial linear 9-mer human leucocyte antigen (HLA)-restricted UC peptides were designed based on the well-conserved S2 region of the SARS-CoV-2 spike protein regardless of rapid mutation and glycosylation hindrance. The UC peptides were characterized for its effect on immune molecules and cells by HLA-tetramer refolding assay for HLA-binding ability, by HLA-tetramer specific T cell assay for engaged cytotoxic T lymphocytes (CTLs) involvement, by HLA-dextramer T cell assay for B cell activation, by intracellular cytokine release assay for polarization of immune response, Th1 or Th2. The specific lysis activity assay of T cells was performed for direct activation of cytotoxic T lymphocytes by UC peptides. Mice were immunized for immunogenicity of UC peptides in vivo and immunized sera was assay for complement cytotoxicity assay. Results appeared that through the engagement of UC peptides and immune molecules, HLA-I and II, that CTLs elicited cytotoxic activity by recognizing SARS-CoV-2 spike-bearing cells and preferably secreting Th1 cytokines. The UC peptides also showed immunogenicity and generated a specific antibody in mice by both intramuscular injection and oral delivery without adjuvant formulation. In conclusion, a T-cell vaccine could provide long-lasting protection against SARS-CoV-2 either during reinfection or during SARS-CoV-2 rebound. Due to its ability to eradicate SARS-CoV-2 virus-infected cells, a COVID-19 T-cell vaccine might provide a solution to lower COVID-19 severity and long COVID-19.


Asunto(s)
Linfocitos B , Vacunas contra la COVID-19 , COVID-19 , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Linfocitos T Citotóxicos , Vacunas de Subunidad , Glicoproteína de la Espiga del Coronavirus/inmunología , Animales , Humanos , Ratones , SARS-CoV-2/inmunología , Vacunas contra la COVID-19/inmunología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Linfocitos B/inmunología , Linfocitos T Citotóxicos/inmunología , COVID-19/inmunología , COVID-19/prevención & control , Femenino , Antígenos HLA/inmunología , Ratones Endogámicos BALB C , Vacunas de Subunidades Proteicas
8.
Poult Sci ; 103(10): 104148, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39142031

RESUMEN

Avian pathogenic Escherichia coli (APEC) is a notable pathogen that frequently leads to avian colibacillosis, posing a substantial risk to both the poultry industry and public health. The commercial vaccines against avian colibacillosis are primarily inactivated vaccines, but their effectiveness is limited to specific serotypes. Recent advances have highlighted bacterial membrane vesicles (MV) as a promising candidate in vaccine research. How to produce bacterial MVs vaccines on a large scale is a significant challenge for the industrialization of MVs. The msbB gene encodes an acyltransferase and has been implicated in altering the acylation pattern of lipid A, leading to a decrease in lipid A content in lipopolysaccharides (LPS). Here, we evaluated the immunoprotective efficacy of MVs derived from the LPS low-expressed APEC strain FY26ΔmsbB, which was an APEC mutant strain with a deletion of the msbB gene. The nitrogen cavitation technique was employed to extract APEC MVs, with results indicating a significant increase in MVs yield compared to that obtained under natural culture. The immunization effectiveness was assessed, revealing that FY26ΔmsbB MVs elicited an antibody response of laying hens and facilitated bacterial clearance. Protective efficacy studies demonstrated that immunization with FY26ΔmsbB MVs conferred the immune protection in chickens challenged with the wild-type APEC strain FY26. Notably, LPS low-carried MVs recovered from the mutant FY26ΔmsbB also displayed cross-protective capabilities, and effectively safeguarding against infections caused by O1, O7, O45, O78, and O101 serotypes virulent APEC strains. These findings suggest that MVs generated from the LPS low-expressed APEC strain FY26ΔmsbB represent a novel and empirically validated subunit vaccine for the prevention and control of infections by various APEC serotypes.


Asunto(s)
Pollos , Infecciones por Escherichia coli , Vacunas contra Escherichia coli , Escherichia coli , Enfermedades de las Aves de Corral , Vacunas de Subunidad , Animales , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/inmunología , Infecciones por Escherichia coli/veterinaria , Infecciones por Escherichia coli/prevención & control , Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Vacunas contra Escherichia coli/inmunología , Vacunas contra Escherichia coli/administración & dosificación , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Femenino , Protección Cruzada
9.
Vaccine ; 42(24): 126254, 2024 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-39213981

RESUMEN

Lawsonia intracellularis is the causative agent of ileitis in swine that manifests as slower weight gain, mild or hemorrhagic diarrhea and/or death in severe cases. As an economically important swine pathogen, development of effective vaccines is important to the swine industry. In developing a subunit vaccine with three recombinant antigens - FliC, GroEL and YopN - we wanted to identify a formulation that would produce robust immune responses that reduce disease parameters associated with Lawsonia intracellularis infection. We formulated these three antigens with four adjuvants: Montanide ISA 660 VG, Montanide Gel 02 PR, Montanide IMS 1313 VG NST, and Montanide ISA 61 VG in an immunogenicity study. Groups vaccinated with formulations including Montanide ISA 660 VG or Montanide ISA 61 VG had significantly more robust immune responses than groups vaccinated with formulations including Montanide Gel 02 PR or Montanide IMS 1313 VG NST. In the challenge study, animals vaccinated with these antigens and Montanide ISA 61 VG had reduced lesion scores, reduced lesion lengths, and increased average daily gain, but no reduction in shedding relative to the control animals. This work shows that this vaccine formulation should be considered for future study in a field and performance trial.


Asunto(s)
Infecciones por Desulfovibrionaceae , Lawsonia (Bacteria) , Enfermedades de los Porcinos , Vacunas de Subunidad , Animales , Porcinos , Lawsonia (Bacteria)/inmunología , Enfermedades de los Porcinos/prevención & control , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/microbiología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Infecciones por Desulfovibrionaceae/prevención & control , Infecciones por Desulfovibrionaceae/inmunología , Infecciones por Desulfovibrionaceae/veterinaria , Vacunación/métodos , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/administración & dosificación , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Emulsiones , Derrame de Bacterias
10.
Eur J Pharm Biopharm ; 203: 114437, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39122053

RESUMEN

Tuberculosis (TB) has been and still is a global emergency for centuries. Prevention of disease through vaccination would have a major impact on disease prevalence, but the only available current vaccine, BCG, has insufficient impact. In this article, a novel subunit vaccine against TB was developed, using the Ag85B-ESAT6-Rv2034 fusion antigen, two adjuvants - CpG and MPLA, and a cationic pH-sensitive liposome as a delivery system, representing a new TB vaccine delivery strategy not previously reported for TB. In vitro in human dendritic cells (DCs), the adjuvanted formulation induced a significant increase in the production of (innate) cytokines and chemokines compared to the liposome without additional adjuvants. In vivo, the new vaccine administrated subcutaneously significantly reduced Mycobacterium tuberculosis (Mtb) bacterial load in the lungs and spleens of mice, significantly outperforming results from mice vaccinated with the antigen mixed with adjuvants without liposomes. In-depth analysis underpinned the vaccine's effectiveness in terms of its capacity to induce polyfunctional CD4+ and CD8+ T-cell responses, both considered essential for controlling Mtb infection. Also noteworthy was the differential abundance of various CD69+ B-cell subpopulations, which included IL17-A-producing B-cells. The vaccine stimulated robust antigen-specific antibody titers, further extending its potential as a novel protective agent against TB.


Asunto(s)
Adyuvantes Inmunológicos , Liposomas , Mycobacterium tuberculosis , Vacunas contra la Tuberculosis , Tuberculosis , Vacunas de Subunidad , Animales , Vacunas contra la Tuberculosis/administración & dosificación , Vacunas contra la Tuberculosis/inmunología , Mycobacterium tuberculosis/inmunología , Ratones , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Humanos , Femenino , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Concentración de Iones de Hidrógeno , Tuberculosis/prevención & control , Tuberculosis/inmunología , Ratones Endogámicos C57BL , Células Dendríticas/inmunología , Cationes , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/administración & dosificación , Citocinas/metabolismo , Linfocitos T CD8-positivos/inmunología , Pulmón/inmunología , Pulmón/microbiología , Linfocitos T CD4-Positivos/inmunología , Carga Bacteriana
11.
Vaccine ; 42(25): 126213, 2024 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-39138071

RESUMEN

Zika virus (ZIKV) infection remains a global public health problem. After the "Public Health Emergencies of International Concern" declared in February 2016, the incidence of new infections by this pathogen has been decreasing in many areas. However, there is still a likely risk that ZIKV will spread to more countries. To date, there is no vaccine or antiviral drug available to prevent or treat Zika virus infection. In the Zika vaccine development, those based on protein subunits are attractive as a non-replicable platform due to their potentially enhanced safety profile to be used in all populations. However, these vaccines frequently require multiple doses and adjuvants to achieve protective immunity. In this study we show the immunological evaluation of new formulations of the recombinant protein ZEC, which combines regions of domain III of the envelope and the capsid from ZIKV. Two nucleotide-based adjuvants were used to enhance the immunity elicited by the vaccine candidate ZEC. ODN 39M or c-di-AMP was incorporated as immunomodulator into the formulations combined with aluminum hydroxide. Following immunizations in immunocompetent BALB/c mice, the formulations stimulated high IgG antibodies. Although the IgG subtypes suggested a predominantly Th1-biased immune response by the formulation including the ODN 39M, cellular immune responses measured by IFNγ secretion from spleen cells after in vitro stimulations were induced by both immunomodulators. These results demonstrate the capacity of both immunomodulators to enhance the immunogenicity of the recombinant subunit ZEC as a vaccine candidate against ZIKV.


Asunto(s)
Adyuvantes Inmunológicos , Anticuerpos Antivirales , Ratones Endogámicos BALB C , Vacunas de Subunidad , Vacunas Sintéticas , Infección por el Virus Zika , Virus Zika , Animales , Virus Zika/inmunología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Infección por el Virus Zika/prevención & control , Infección por el Virus Zika/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Ratones , Femenino , Adyuvantes Inmunológicos/administración & dosificación , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunogenicidad Vacunal , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Adyuvantes de Vacunas , Inmunidad Celular , Proteínas del Envoltorio Viral/inmunología , Proteínas de la Cápside/inmunología , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/inmunología
12.
Vaccine ; 42(22): 126223, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39151232

RESUMEN

Objectives We evaluated the safety, immunogenicity and efficacy of Abdala, a protein subunit vaccine for 2019 coronavirus disease (COVID-19), in children and adolescents. Methods A phase 2, open-label, single-arm clinical trial was carried out. Subjects aged 3 to 18 years were eligible. Abdala vaccine was administered intramuscularly at 0-14-28 days. The main endpoints were safety and the immunobridging analysis with a non-inferiority design, to infer the efficacy of the vaccine in paediatric population based on the comparison of neutralizing antibodies (NAb) to SARS-CoV-2, with adults (19-21 years). The trial is registered with the Cuban Public Registry of Clinical Trials, RPCEC00000390. Results From September 13th to September 17th, 2021, 703 participants were included in the context of a predominantly SARS-CoV-2 Delta variant circulation. The number of individuals who experienced adverse reactions was 264/703 (37·6%). Adverse reactions were mostly mild and occurred at the injection site, which resolved within the first 24-48 h. There were no reports of severe adverse events. For the non-inferiority comparison of 297 children (3-11 years) with 297 adults, the geometric mean (GMT) ratio of SARS-CoV-2 NAb was 0·87 (95% CI 0·69-1·08) and 1·07 (0·82-1·39) in the same comparison for 203 adolescents (12-18 years) and 203 adults. For both age groups, the lower limit of GMT was higher than 0·67. The differences in seroresponse rates of Nab for children were 1% (-2%, 4%) and -3% (-7%, 1%) for adolescents, higher than -10% in both age groups. Conclusions The Abdala vaccine was safe and immunogenic in a paediatric population aged 3-18 years, with inferred efficacy based on non-inferior analysis. The vaccine is very suitable to fit into massive vaccination strategies, considering the advantages of using the same vaccine strength (RBD 50 µg) and schedule of administration for both adults and children, as well as the easy storage and handling conditions at 2-8 °C.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Vacunas contra la COVID-19 , COVID-19 , SARS-CoV-2 , Vacunas de Subunidad , Humanos , Adolescente , Niño , Vacunas contra la COVID-19/efectos adversos , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/administración & dosificación , COVID-19/prevención & control , COVID-19/inmunología , Femenino , Masculino , Preescolar , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , SARS-CoV-2/inmunología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/efectos adversos , Vacunas de Subunidad/administración & dosificación , Eficacia de las Vacunas , Inmunogenicidad Vacunal , Adulto Joven
13.
Viruses ; 16(8)2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39205245

RESUMEN

Neonates are more susceptible to influenza virus infection than adults, resulting in increased morbidity and mortality and delayed clearance of the virus. Generating effective CD8+ T cell responses may be important for improving vaccination outcomes in vulnerable populations, but neonatal T cells frequently respond differently than adult cells. We sought to understand CD8+ T cell specificity and immunodominance during neonatal influenza infection and how any differences from the adult hierarchy might impact peptide vaccine effectiveness. Neonatal C57BL/6 mice displayed an altered CD8+ T cell immunodominance hierarchy during influenza infection, preferentially responding to an epitope in the influenza protein PA rather than the co-dominant adult response to NP and PA. Heterosubtypic infections in mice first infected as pups also displayed altered immunodominance and reduced protection compared to mice first infected as adults. Adoptive transfer of influenza-infected bone-marrow-derived dendritic cells promoted an NP-specific CD8+ T cell response in influenza-virus-infected pups and increased viral clearance. Finally, pups responded to PA (224-233), but not NP (366-374) during peptide vaccination. PA (224-233)-vaccinated mice were not protected during viral challenge. Epitope usage should be considered when designing vaccines that target T cells when the intended patient population includes infants and adults.


Asunto(s)
Animales Recién Nacidos , Linfocitos T CD8-positivos , Epítopos Inmunodominantes , Vacunas contra la Influenza , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae , Animales , Linfocitos T CD8-positivos/inmunología , Ratones , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Epítopos Inmunodominantes/inmunología , Vacunación , Femenino , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Epítopos de Linfocito T/inmunología
15.
ACS Infect Dis ; 10(9): 3419-3429, 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39196071

RESUMEN

Despite concerted efforts to tackle the COVID-19 pandemic, the persistent transmission of SARS-CoV-2 demands continued research into novel vaccination strategies to combat the virus. In light of this, intranasally administered peptide vaccines, particularly those conjugated to an immune adjuvant to afford so-called "self-adjuvanted vaccines", remain underexplored. Here, we describe the synthesis and immunological evaluation of self-adjuvanting peptide vaccines derived from epitopes of the spike glycoprotein of SARS-CoV-2 covalently fused to the potent adjuvant, Pam2Cys, that targets toll-like receptor 2 (TLR2). When administered intranasally, these vaccines elicited a strong antigen-specific CD4+ and CD8+ T-cell response in the lungs as well as high titers of IgG and IgA specific to the native spike protein of SARS-CoV-2. Unfortunately, serum and lung fluid from mice immunized with these vaccines failed to inhibit viral entry in spike-expressing pseudovirus assays. Following this, we designed and synthesized fusion vaccines composed of the T-cell epitope discovered in this work, covalently fused to epitopes of the receptor-binding domain of the spike protein reported to be neutralizing. While antibodies elicited against these fusion vaccines were not neutralizing, the T-cell epitope retained its ability to stimulate strong antigen-specific CD4+ lymphocyte responses within the lungs. Given the Spike(883-909) region is still completely conserved in SARS-CoV-2 variants of concern and variants of interest, we envision the self-adjuvanting vaccine platform reported here may inform future vaccine efforts.


Asunto(s)
Adyuvantes Inmunológicos , Administración Intranasal , Anticuerpos Antivirales , Vacunas contra la COVID-19 , COVID-19 , Lipopéptidos , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Animales , SARS-CoV-2/inmunología , Ratones , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/administración & dosificación , Glicoproteína de la Espiga del Coronavirus/inmunología , COVID-19/prevención & control , COVID-19/inmunología , Lipopéptidos/inmunología , Lipopéptidos/administración & dosificación , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Femenino , Humanos , Ratones Endogámicos BALB C , Adyuvantes de Vacunas/administración & dosificación , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Inmunidad Celular , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD4-Positivos/inmunología
16.
Brain Behav Immun ; 122: 185-201, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39142420

RESUMEN

Amyloid-ß (Aß) and hyperphosphorylated tau protein are targets for Alzheimer's Disease (AD) immunotherapies, which are generally focused on single epitopes within Aß or tau. However, due to the complexity of both Aß and tau in AD pathogenesis, a multipronged approach simultaneously targeting multiple epitopes of both proteins could overcome limitations of monotherapies. Herein, we propose an active AD immunotherapy based on a nanoparticle vaccine comprising two Aß peptides (1-14 and pyroglutamate pE3-14) and three tau peptides (centered on phosphorylated pT181, pT217 and pS396/404). These correspond to both soluble and aggregated targets and are displayed on the surface of immunogenic liposomes in an orientation that maintains reactivity with epitope-specific monoclonal antibodies. Intramuscular immunization of mice with individual epitopes resulted in minimally cross-reactive antibody induction, while simultaneous co-display of 5 antigens ("5-plex") induced antibodies against all epitopes without immune interference. Post-immune sera recognized plaques and neurofibrillary tangles from human AD brain tissue. Vaccine administration to 3xTg-AD mice using a prophylactic dosing schedule inhibited tau and amyloid pathologies and resulted in improved cognitive function. Immunization was well tolerated and did not induce antigen-specific cellular responses or persistent inflammatory responses in the peripheral or central nervous system. Antibody levels could be reversed by halting monthly vaccinations. Altogether, these results indicate that active immune therapies based on nanoparticle formulations of multiple Aß and tau epitopes warrant further study for treating early-stage AD.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Modelos Animales de Enfermedad , Ratones Transgénicos , Proteínas tau , Animales , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/prevención & control , Proteínas tau/inmunología , Proteínas tau/metabolismo , Péptidos beta-Amiloides/inmunología , Péptidos beta-Amiloides/metabolismo , Ratones , Humanos , Vacunas contra el Alzheimer/inmunología , Vacunas contra el Alzheimer/administración & dosificación , Encéfalo/metabolismo , Femenino , Epítopos/inmunología , Nanopartículas , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Anticuerpos , Vacunas de Subunidades Proteicas
17.
Microbiol Spectr ; 12(10): e0105424, 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39189763

RESUMEN

Yersinia pestis has a broad host range and has caused lethal bubonic and pneumonic plague in humans. With the emergence of multiple resistant strains and the potential for biothreat use, there is an urgent need for new therapeutic strategies that can protect populations from natural or deliberate infection. Targeting F1 has been proven to be the main strategy for developing vaccines and therapeutic antibodies, but data on anti-F1 antibodies, especially in humans, are scarce. To date, three human anti-F1 monoclonal antibodies (m252, αF1Ig2, and αF1Ig8) from naive populations have been reported. Here, we constructed an antibody library from vaccinees immunized with the plague subunit vaccine IIa by phage display. The genetic basis, epitopes, and biological functions of the obtained mAbs were assessed and evaluated in plague-challenged mice. Three human mAbs, namely, F3, F19, and F23, were identified. Their biolayer responses were 0.4, 0.6, and 0.6 nm, respectively. The dissociation constants (KD) of the F1 antigen were 1 pM, 0.165 nM, and 1 pM, respectively. Although derived from distinct Ab lineages, that is, VH3-30-D3-10-JH4 (F3&F23) and VH3-43-D6-19-JH4 (F19), these mAbs share similar binding sites in F1 with some overlap with αF1Ig8 but are distinct from αF1Ig2. Each of them provided a significant protective effect for Balb/c mice against a 100 median lethal dose (MLD) challenge of a virulent Y. pestis strain when administered at a dose of 100 µg. No synergistic or antagonistic effects were observed among them. These mAbs are novel and excellent candidates for further drug development and use in clinical practice.IMPORTANCEIn this study, we identified three human monoclonal antibodies with a high affinity to F1 protein of Yersinia pestis. We discovered that they have relatively lower somatic hypermutations compared with antibodies, m252, αF1Ig2, and αF1Ig8, derived from the naive library reported previously. We also observed that these mAbs share similar binding sites in F1 with some overlapping with αF1Ig8 but distinct from that of αF1Ig2. Furthermore, each of them could provide complete protection for mice against a lethal dose of Yersinia pestis challenge. Our data provided new insights into the anti-F1 Ab repertories and their associated epitopes during vaccination in humans. The findings support the additional novel protective human anti-F1Abs for potential therapeutics against plaque.


Asunto(s)
Anticuerpos Antibacterianos , Anticuerpos Monoclonales , Ratones Endogámicos BALB C , Vacuna contra la Peste , Peste , Yersinia pestis , Peste/prevención & control , Peste/inmunología , Yersinia pestis/inmunología , Animales , Humanos , Ratones , Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/administración & dosificación , Vacuna contra la Peste/inmunología , Vacuna contra la Peste/administración & dosificación , Femenino , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/genética , Epítopos/inmunología , Vacunación , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación
18.
Fish Shellfish Immunol ; 153: 109841, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39173984

RESUMEN

Largemouth bass virus (LMBV) infections has resulted in high mortality and economic losses to the global largemouth bass industry and has seriously restricted the healthy development of the bass aquaculture industry. There are currently no antiviral therapies available for the control of this disease. In this study, we developed three types of vaccine against LMBV; whole virus inactivated vaccine (I), a subunit vaccine composed of the major viral capsid protein MCP (S) as well as an MCP DNA vaccine(D), These were employed using differing immunization and booster strategies spaced 2 weeks apart as follows: II, SS, DD and DS. We found that all vaccine groups induced humoral and cellular immune responses and protected largemouth bass from a lethal LMBV challenge to varying degrees and DD produced the best overall effect. Specifically, the levels of specific IgM in serum in all immunized groups were elevated and significantly higher than those in the control group. Moreover, the expression of humoral immunity (CD4 and IgM) and cellular immunity (MHCI-α) as well as cytokines (IL-1ß) was increased, and the activity of immunity-related enzymes ACP, AKP, LZM, and T-SOD in the serum was significantly enhanced. In addition, the relative percent survival of fish following an LMBV lethal challenge 4 weeks after the initial immunizations were high for each group: DD(89.5 %),DS(63.2 %),SS(50 %) and II (44.7 %). These results indicated that the MCP DNA vaccine is the most suitable and promising vaccine candidate for the effective control of LMBV disease.


Asunto(s)
Lubina , Infecciones por Virus ADN , Enfermedades de los Peces , Vacunas de ADN , Vacunas Virales , Animales , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Enfermedades de los Peces/prevención & control , Enfermedades de los Peces/inmunología , Lubina/inmunología , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Infecciones por Virus ADN/veterinaria , Infecciones por Virus ADN/prevención & control , Infecciones por Virus ADN/inmunología , Vacunas de Productos Inactivados/inmunología , Vacunas de Productos Inactivados/administración & dosificación , Inmunidad Humoral , Ranavirus/inmunología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Inmunidad Celular
19.
Int J Nanomedicine ; 19: 8029-8042, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39130684

RESUMEN

Purpose: Heterologous immunization using different vaccine platforms has been demonstrated as an efficient strategy to enhance antigen-specific immune responses. In this study, we performed a head-to-head comparison of both humoral and cellular immune response induced by different prime-boost immunization regimens of mRNA vaccine and adjuvanted protein subunit vaccine against varicella-zoster virus (VZV) in middle-aged mice, aiming to get a better understanding of the influence of vaccination schedule on immune response. Methods: VZV glycoprotein (gE) mRNA was synthesized and encapsulated into SM-102-based lipid nanoparticles (LNPs). VZV-primed middle-aged C57BL/6 mice were then subjected to homologous and heterologous prime-boost immunization strategies using VZV gE mRNA vaccine (RNA-gE) and protein subunit vaccine (PS-gE). The antigen-specific antibodies were evaluated using enzyme-linked immunosorbent assay (ELISA) analysis. Additionally, cell-mediated immunity (CMI) was detected using ELISPOT assay and flow cytometry. Besides, in vivo safety profiles were also evaluated and compared. Results: The mRNA-loaded lipid nanoparticles had a hydrodynamic diameter of approximately 130 nm and a polydispersity index of 0.156. Total IgG antibody levels exhibited no significant differences among different immunization strategies. However, mice received 2×RNA-gE or RNA-gE>PS-gE showed a lower IgG1/IgG2c ratio than those received 2×PS-gE and PS-gE> RNA-gE. The CMI response induced by 2×RNA-gE or RNA-gE>PS-gE was significantly stronger than that induced by 2×PS-gE and PS-gE> RNA-gE. The safety evaluation indicated that both mRNA vaccine and protein vaccine induced a transient body weight loss in mice. Furthermore, the protein vaccine produced a notable inflammatory response at the injection sites, while the mRNA vaccine showed no observable inflammation. Conclusion: The heterologous prime-boost strategy has demonstrated that an mRNA-primed immunization regimen can induce a better cell-mediated immune response than a protein subunit-primed regimen in middle-aged mice. These findings provide valuable insights into the design and optimization of VZV vaccines with the potentials to broaden varicella vaccination strategies in the future.


Asunto(s)
Adyuvantes Inmunológicos , Inmunidad Celular , Ratones Endogámicos C57BL , Nanopartículas , Vacunas de Subunidad , Animales , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Nanopartículas/química , Adyuvantes Inmunológicos/administración & dosificación , Femenino , Vacunas de ARNm , Ratones , Herpesvirus Humano 3/inmunología , Anticuerpos Antivirales/sangre , Inmunización Secundaria/métodos , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/administración & dosificación , Vacuna contra el Herpes Zóster/inmunología , Vacuna contra el Herpes Zóster/administración & dosificación , Liposomas
20.
Hum Vaccin Immunother ; 20(1): 2396710, 2024 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-39193781

RESUMEN

Human papillomavirus (HPV) has been linked to the development of various cancers, including head and neck, cervical, vaginal, penile, and anal cancers. The development of therapeutic vaccines against HPV-positive tumors is crucial for protecting individuals already infected with HPV, preventing tumor progression, and effectively treating the disease. The HPV therapeutic peptide-based vaccines demonstrate specificity and safety advantages by targeting specific epitopes while minimizing the risk of allergic or autoimmune reactions. However, HPV therapeutic peptide-based vaccines typically lack immunogenicity and frequently fail to induce effective immune responses. Therefore, there is a need for more effective approaches to improve the immunogenicity of HPV peptide-based vaccines. Here, we review relevant research and possible uses for increasing the immunogenicity and therapeutic efficacy of HPV peptide-based vaccines through combined therapy and improved delivery strategies. Additional research is necessary to validate the application of combination therapy and delivery strategy modifications as standard treatment approaches for HPV therapeutic peptide-based vaccines.


Asunto(s)
Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Vacunas de Subunidad , Humanos , Vacunas contra Papillomavirus/administración & dosificación , Vacunas contra Papillomavirus/inmunología , Infecciones por Papillomavirus/prevención & control , Infecciones por Papillomavirus/terapia , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Terapia Combinada/métodos , Papillomaviridae/inmunología , Inmunogenicidad Vacunal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...