Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 429
Filtrar
1.
Angew Chem Int Ed Engl ; 61(41): e202209556, 2022 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-35950629

RESUMO

Acinetobacter baumannii is a opportunistic bacterial pathogen responsible for serious nosocomial infections that is becoming increasingly resistant against antibiotics. Capsular polysaccharides (CPS) that cover A. baumannii are a major virulence factor that play an important role in pathogenesis, are used to assign serotypes and provide the basis for vaccine development. Synthetic oligosaccharides resembling the CPS of A. baumannii 17978 were printed onto microarray slides and used to screen sera from patients infected with A. baumannii as well as a monoclonal mouse antibody (mAb C8). A synthetic oligosaccharide emerged from glycan array screening as lead for the development of a vaccine against A. baumannii 17978. Tetrasaccharide 20 is a key epitope for recognition by an antibody and is a vaccine lead.


Assuntos
Infecções por Acinetobacter , Acinetobacter baumannii , Glicoconjugados , Vacinas Sintéticas , Infecções por Acinetobacter/prevenção & controle , Acinetobacter baumannii/efeitos dos fármacos , Animais , Antibacterianos/farmacologia , Epitopos , Glicoconjugados/farmacologia , Humanos , Camundongos , Oligossacarídeos , Polissacarídeos , Vacinas Sintéticas/farmacologia , Fatores de Virulência
2.
Signal Transduct Target Ther ; 7(1): 69, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-35241645

RESUMO

Emerging SARS-CoV-2 variants and the gradually decreasing neutralizing antibodies over time post vaccination have led to an increase in incidents of breakthrough infection across the world. To investigate the potential protective effect of the recombinant protein subunit COVID-19 vaccine targeting receptor-binding domain (RBD) (PS-RBD) and whole inactivated virus particle vaccine (IV) against the variant strains, in this study, rhesus macaques were immunized with PS-RBD or IV vaccine, followed by a Beta variant (B.1.351) challenge. Although neutralizing activity against the Beta variant was reduced compared with that against the prototype, the decreased viral load in both upper and lower respiratory tracts, milder pathological changes, and downregulated inflammatory cytokine levels in lung tissues after challenge demonstrated that PS-RBD and IV still provided effective protection against the Beta variant in the macaque model. Furthermore, PS-RBD-induced macaque sera possessed general binding and neutralizing activity to Alpha, Beta, Delta, and Omicron variants in our study, though the neutralizing antibody (NAb) titers declined by varying degrees, demonstrating potential protection of PS-RBD against current circulating variants of concern (VOCs). Interestingly, although the IV vaccine-induced extremely low neutralizing antibody titers against the Beta variant, it still showed reduction for viral load and significantly alleviated pathological change. Other correlates of vaccine-induced protection (CoP) like antibody-dependent cellular cytotoxicity (ADCC) and immune memory were both confirmed to be existing in IV vaccinated group and possibly be involved in the protective mechanism.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Vacinas contra COVID-19 , COVID-19 , Imunogenicidade da Vacina , SARS-CoV-2/imunologia , Animais , COVID-19/imunologia , COVID-19/prevenção & controle , Vacinas contra COVID-19/imunologia , Vacinas contra COVID-19/farmacologia , Humanos , Macaca mulatta , Vacinas de Produtos Inativados/imunologia , Vacinas de Produtos Inativados/farmacologia , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/farmacologia
3.
JCI Insight ; 7(5)2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35104245

RESUMO

Benchmarks for protective immunity from infection or severe disease after SARS-CoV-2 vaccination are still being defined. Here, we characterized virus neutralizing and ELISA antibody levels, cellular immune responses, and viral variants in 4 separate groups: healthy controls (HCs) weeks (early) or months (late) following vaccination in comparison with symptomatic patients with SARS-CoV-2 after partial or full mRNA vaccination. During the period of the study, most symptomatic breakthrough infections were caused by the SARS-CoV-2 Alpha variant. Neutralizing antibody levels in the HCs were sustained over time against the vaccine parent virus but decreased against the Alpha variant, whereas IgG titers and T cell responses against the parent virus and Alpha variant declined over time. Both partially and fully vaccinated patients with symptomatic infections had lower virus neutralizing antibody levels against the parent virus than the HCs, similar IgG antibody titers, and similar virus-specific T cell responses measured by IFN-γ. Compared with HCs, neutralization activity against the Alpha variant was lower in the partially vaccinated infected patients and tended to be lower in the fully vaccinated infected patients. In this cohort of breakthrough infections, parent virus neutralization was the superior predictor of breakthrough infections with the Alpha variant of SARS-CoV-2.


Assuntos
Imunidade Adaptativa , Anticorpos Antivirais/imunologia , Vacinas contra COVID-19/farmacologia , COVID-19/virologia , SARS-CoV-2/imunologia , Vacinação/métodos , Vacinas Sintéticas/farmacologia , Vacinas de mRNA/farmacologia , Adulto , Idoso , COVID-19/epidemiologia , COVID-19/prevenção & controle , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Pandemias , Vigilância da População , Estudos Retrospectivos , Estados Unidos/epidemiologia , Adulto Jovem
5.
Front Immunol ; 12: 730116, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745099

RESUMO

Klebsiella pneumoniae found in the normal flora of the human oral and intestinal tract mainly causes hospital-acquired infections but can also cause community-acquired infections. To date, most clinical trials of vaccines against K. pneumoniae have ended in failure. Furthermore, no single conserved protein has been identified as an antigen candidate to accelerate vaccine development. In this study, we identified five outer membrane proteins of K. pneumoniae, namely, Kpn_Omp001, Kpn_Omp002, Kpn_Omp003, Kpn_Omp004, and Kpn_Omp005, by using reliable second-generation proteomics and bioinformatics. Mice vaccinated with these five KOMPs elicited significantly higher antigen-specific IgG, IgG1, and IgG2a. However, only Kpn_Omp001, Kpn_Omp002, and Kpn_Omp005 were able to induce a protective immune response with two K. pneumoniae infection models. These protective effects were accompanied by the involvement of different immune responses induced by KOMPs, which included KOMPs-specific IFN-γ-, IL4-, and IL17A-mediated immune responses. These findings indicate that Kpn_Omp001, Kpn_Omp002, and Kpn_Omp005 are three potential Th1, Th2, and Th17 candidate antigens, which could be developed into multivalent and serotype-independent vaccines against K. pneumoniae infection.


Assuntos
Proteínas da Membrana Bacteriana Externa/farmacologia , Vacinas Bacterianas/farmacologia , Infecções por Klebsiella/prevenção & controle , Klebsiella pneumoniae/imunologia , Desenvolvimento de Vacinas , Animais , Carga Bacteriana , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Modelos Animais de Doenças , Células HL-60 , Humanos , Imunogenicidade da Vacina , Infecções por Klebsiella/imunologia , Infecções por Klebsiella/microbiologia , Klebsiella pneumoniae/genética , Camundongos Endogâmicos BALB C , Fagócitos/imunologia , Fagócitos/microbiologia , Fagocitose , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Linfócitos T/imunologia , Linfócitos T/microbiologia , Vacinação , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/farmacologia
6.
Elife ; 102021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34636722

RESUMO

While mRNA vaccines are proving highly efficacious against SARS-CoV-2, it is important to determine how booster doses and prior infection influence the immune defense they elicit, and whether they protect against variants. Focusing on the T cell response, we conducted a longitudinal study of infection-naïve and COVID-19 convalescent donors before vaccination and after their first and second vaccine doses, using a high-parameter CyTOF analysis to phenotype their SARS-CoV-2-specific T cells. Vaccine-elicited spike-specific T cells responded similarly to stimulation by spike epitopes from the ancestral, B.1.1.7 and B.1.351 variant strains, both in terms of cell numbers and phenotypes. In infection-naïve individuals, the second dose boosted the quantity and altered the phenotypic properties of SARS-CoV-2-specific T cells, while in convalescents the second dose changed neither. Spike-specific T cells from convalescent vaccinees differed strikingly from those of infection-naïve vaccinees, with phenotypic features suggesting superior long-term persistence and ability to home to the respiratory tract including the nasopharynx. These results provide reassurance that vaccine-elicited T cells respond robustly to emerging viral variants, confirm that convalescents may not need a second vaccine dose, and suggest that vaccinated convalescents may have more persistent nasopharynx-homing SARS-CoV-2-specific T cells compared to their infection-naïve counterparts.


Vaccination is one of the best ways to prevent severe COVID-19. Two doses of mRNA vaccine protect against serious illness caused by the coronavirus SARS-CoV-2. They do this, in part, by encouraging the immune system to make specialised proteins known as antibodies that recognise the virus. Most of the vaccine research so far has focussed on these antibodies, but they are only one part of the immune response. Vaccines also activate immune cells called T cells. These cells have two main roles, coordinating the immune response and killing cells infected with viruses. It is likely that they play a key role in preventing severe COVID-19. There are many kinds of T cells, each with a different role. Currently, the identity and characteristics of the T cells that protect against COVID-19 is unclear. Different types of T cells have unique proteins on their surface. Examining these proteins can reveal details about how the T cells work, which part of the virus they recognise, and which part of the body they protect. A tool called cytometry by time of flight allows researchers to measure these proteins, one cell at a time. Using this technique, Neidleman, Luo et al. investigated T cells from 11 people before vaccination and after their first and second doses. Five people had never had COVID-19 before, and six had already recovered from COVID-19. Neidleman, Luo et al. found that the T cells recognizing SARS-CoV-2 in the two groups differed. In people who had never had COVID-19 before, the second dose of vaccine improved the quality and quantity of the T cells. The same was not true for people who had already recovered from COVID-19. However, although their T cells did not improve further after a second vaccine dose, they did show signs that they might offer more protection overall. The proteins on the cells suggest that they might last longer, and that they might specifically protect the nose, throat and lungs. Neidleman, Luo et al. also found that, for both groups, T cells activated by vaccination responded in the same way to different variants of the virus. This work highlights the importance of getting both vaccine doses for people who have never had COVID-19. It also suggests that vaccination in people who have had COVID-19 may generate better T cells. Larger studies could show whether these patterns remain true across the wider population. If so, it is possible that delivering vaccines to the nose or throat could boost immunity by mimicking natural infection. This might encourage T cells to make the surface proteins that allow them to home to these areas.


Assuntos
Vacinas contra COVID-19/farmacologia , COVID-19/imunologia , SARS-CoV-2/imunologia , Linfócitos T/efeitos dos fármacos , Vacinas Sintéticas/farmacologia , Adulto , Idoso , COVID-19/prevenção & controle , COVID-19/virologia , Feminino , Humanos , Imunização Secundária , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinação , Adulto Jovem , Vacinas de mRNA
7.
J Immunother Cancer ; 9(9)2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34479925

RESUMO

BACKGROUND: MVA-BN-brachyury-TRICOM is a recombinant vector-based therapeutic cancer vaccine designed to induce an immune response against brachyury. Brachyury, a transcription factor overexpressed in advanced cancers, has been associated with treatment resistance, epithelial-to-mesenchymal transition, and metastatic potential. MVA-BN-brachyury-TRICOM has demonstrated immunogenicity and safety in previous clinical trials of subcutaneously administered vaccine. Preclinical studies have suggested that intravenous administration of therapeutic vaccines can induce superior CD8+ T cell responses, higher levels of systemic cytokine release, and stronger natural killer cell activation and proliferation. This is the first-in-human study of the intravenous administration of MVA-BN-brachyury-TRICOM. METHODS: Between January 2020 and March 2021, 13 patients were treated on a phase 1, open-label, 3+3 design, dose-escalation study at the National Institutes of Health Clinical Center. The study population was adults with advanced solid tumors and was enriched for chordoma, a rare sarcoma of the notochord that overexpresses brachyury. Vaccine was administered intravenously at three DLs on days 1, 22, and 43. Blood samples were taken to assess drug pharmacokinetics and immune activation. Imaging was conducted at baseline, 1 month, and 3 months post-treatment. The primary endpoint was safety and tolerability as determined by the frequency of dose-limiting toxicities; a secondary endpoint was determination of the recommended phase 2 dose. RESULTS: No dose-limiting toxicities were observed and no serious adverse events were attributed to the vaccine. Vaccine-related toxicities were consistent with class profile (ie, influenza-like symptoms). Cytokine release syndrome up to grade 2 was observed with no adverse outcomes. Dose-effect trend was observed for fever, chills/rigor, and hypotension. Efficacy analysis of objective response rate per RECIST 1.1 at the end of study showed one patient with a partial response, four with stable disease, and eight with progressive disease. Three patients with stable disease experienced clinical benefit in the form of improvement in pain. Immune correlatives showed T cell activation against brachyury and other tumor-associated cascade antigens. CONCLUSIONS: Intravenous administration of MVA-BN-brachyury-TRICOM vaccine was safe and tolerable. Maximum tolerated dose was not reached. The maximum administered dose was 109 infectious units every 3 weeks for three doses. This dose was selected as the recommended phase 2 dose. TRIAL REGISTRATION NUMBER: NCT04134312.


Assuntos
Administração Intravenosa/métodos , Vacinas Anticâncer/uso terapêutico , Proteínas Fetais/uso terapêutico , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Proteínas com Domínio T/uso terapêutico , Vacinas Anticâncer/farmacologia , Feminino , Proteínas Fetais/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas com Domínio T/farmacologia , Vacinas Sintéticas/farmacologia , Vacinas Sintéticas/uso terapêutico
8.
Nat Rev Drug Discov ; 20(11): 817-838, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34433919

RESUMO

Over the past several decades, messenger RNA (mRNA) vaccines have progressed from a scepticism-inducing idea to clinical reality. In 2020, the COVID-19 pandemic catalysed the most rapid vaccine development in history, with mRNA vaccines at the forefront of those efforts. Although it is now clear that mRNA vaccines can rapidly and safely protect patients from infectious disease, additional research is required to optimize mRNA design, intracellular delivery and applications beyond SARS-CoV-2 prophylaxis. In this Review, we describe the technologies that underlie mRNA vaccines, with an emphasis on lipid nanoparticles and other non-viral delivery vehicles. We also overview the pipeline of mRNA vaccines against various infectious disease pathogens and discuss key questions for the future application of this breakthrough vaccine platform.


Assuntos
COVID-19/prevenção & controle , Controle de Doenças Transmissíveis , Vacinas Sintéticas , COVID-19/epidemiologia , Ensaios Clínicos como Assunto , Controle de Doenças Transmissíveis/métodos , Controle de Doenças Transmissíveis/tendências , Desenho de Fármacos , Desenvolvimento de Medicamentos/métodos , Humanos , RNA Mensageiro/genética , SARS-CoV-2 , Vacinas Sintéticas/classificação , Vacinas Sintéticas/farmacologia , Vacinas de mRNA
9.
Biomed Pharmacother ; 142: 111953, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34343897

RESUMO

Currently, there are over 230 different COVID-19 vaccines under development around the world. At least three decades of scientific development in RNA biology, immunology, structural biology, genetic engineering, chemical modification, and nanoparticle technologies allowed the accelerated development of fully synthetic messenger RNA (mRNA)-based vaccines within less than a year since the first report of a SARS-CoV-2 infection. mRNA-based vaccines have been shown to elicit broadly protective immune responses, with the added advantage of being amenable to rapid and flexible manufacturing processes. This review recapitulates current advances in engineering the first two SARS-CoV-2-spike-encoding nucleoside-modified mRNA vaccines, highlighting the strategies followed to potentiate their effectiveness and safety, thus facilitating an agile response to the current COVID-19 pandemic.


Assuntos
Engenharia Biomédica , Vacinas contra COVID-19 , COVID-19 , Desenvolvimento de Medicamentos/métodos , Descoberta de Drogas/métodos , SARS-CoV-2 , Vacina de mRNA-1273 contra 2019-nCoV , Engenharia Biomédica/métodos , Engenharia Biomédica/tendências , COVID-19/prevenção & controle , COVID-19/virologia , Vacinas contra COVID-19/classificação , Vacinas contra COVID-19/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Humanos , Imunogenicidade da Vacina , Lipossomos/farmacologia , Nanopartículas , Nucleosídeos/farmacologia , Nucleosídeos/fisiologia , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas Sintéticas/farmacologia
11.
J Cancer Res Clin Oncol ; 147(11): 3255-3268, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34291357

RESUMO

PURPOSE: Mutation-specific T-cell response to epithelial cancers and T-cell-based immunotherapy has been successfully used to treat several human solid cancers. We aimed to investigate the anti-tumour effect of neo-antigen-reactive T(NRT) cells induced by RNA mutanome vaccine, which may serve as a feasible and effective therapeutic approach for lung cancer. METHODS: We predicted candidate neo-antigens according to the mutant gene analysis by sequencing the mouse Lewis cells and C57BL/6 mouse tail tissue. RNA vaccine was prepared with the neo-antigens as the template. We assessed antitumor efficacy, cytokine secretion and pathological changes after adoptive transfer of NRT cells in vitro and vivo experiments. RESULTS: We identified 10 non-synonymous somatic mutations and successfully generated NRT cells. The percentage of T-cell activation proportion was increased from 0.072% in conventional T cells to 9.96% in NRT cells. Interferon-γ secretion augmented from 17.8 to 24.2% as well. As an in vivo model, adoptive NRT cell infusion could promote active T-cell infiltration into the tumour tissue and could delay tumour progression. CONCLUSION: NRT cells induced by RNA mutanome vaccine exert a significant anti-tumour effect in mouse lung cancer, and adoptive NRT cell therapy might be considered a feasible, effective therapeutic approach for lung cancer.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/farmacologia , Imunoterapia Adotiva/métodos , Neoplasias Pulmonares/terapia , Linfócitos T/imunologia , Animais , Antígenos de Neoplasias/genética , Vacinas Anticâncer/imunologia , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/terapia , Linhagem Celular Tumoral , Feminino , Interferon gama/imunologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação Puntual , Distribuição Aleatória , Linfócitos T/transplante , Fator de Necrose Tumoral alfa/imunologia , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/farmacologia , Vacinas de mRNA
12.
Front Immunol ; 12: 670578, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34084170

RESUMO

Life-threatening, invasive fungal infections (IFIs) cause over 1.5 million deaths worldwide and are a major public health concern with high mortality rates even with medical treatment. Infections with the opportunistic fungal pathogen, Aspergillus fumigatus are among the most common. Despite the growing clinical need, there are no licensed vaccines for IFIs. Here we evaluated the immunogenicity and protective efficacy of an A. fumigatus recombinant protein vaccine candidate, AF.KEX1, in experimental murine models of drug-induced immunosuppression. Immunization of healthy mice with AF.KEX1 and adjuvant induced a robust immune response. Following AF.KEX1 or sham immunization, mice were immunosuppressed by treatment with either cortisone acetate or hydrocortisone and the calcineurin inhibitor, tacrolimus. To test vaccine efficacy, immunosuppressed mice were intranasally challenged with A. fumigatus conidia (Af293) and weight and body temperature were monitored for 10 days. At study termination, organism burden in the lungs was evaluated by quantitative PCR and Gomori's methanamine silver staining. In both models of immunosuppression, AF.KEX1 vaccinated mice experienced decreased rates of mortality and significantly lower lung organism burden compared to non-vaccinated controls. The lung fungal burden was inversely correlated with the peak anti-AF.KEX1 IgG titer achieved following vaccination. These studies provide the basis for further evaluation of a novel vaccine strategy to protect individuals at risk of invasive aspergillosis due to immunosuppressive treatments.


Assuntos
Vacinas Fúngicas/imunologia , Vacinas Fúngicas/farmacologia , Hospedeiro Imunocomprometido/imunologia , Aspergilose Pulmonar Invasiva/imunologia , Infecções Oportunistas/imunologia , Animais , Aspergillus fumigatus/imunologia , Modelos Animais de Doenças , Camundongos , Vacinas Sintéticas/farmacologia
13.
Int J Biol Macromol ; 183: 2162-2173, 2021 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-34102236

RESUMO

Effective controls on viral infections rely on the continuous development in vaccine technology. Nanoparticle (NP) antigens are highly immunogenic based on their unique physicochemical properties, making them molecular scaffolds to present soluble vaccine antigens. Here, viral targets (113-354 aas) were genetically fused to N terminal of mi3, a protein that self-assembles into nanoparticles composed of 60 subunits. With transmission electron microscopy, it was confirmed that target-mi3 fusion proteins which have insertions of up to 354 aas in N terminal form intact NPs. Moreover, viral targets are surface-displayed on NPs as indicated in dynamic light scattering. NPs exhibit perfect stability after long-term storage at room temperature. Moreover, SP-E2-mi3 NPs enhance antigen uptake and maturation in dendritic cells (DCs) via up-regulating marker molecules and immunostimulatory cytokines. Importantly, in a mouse model, SP-E2-mi3 nanovaccines against Classical swine fever virus (CSFV) remarkably improved CSFV-specific neutralizing antibodies (NAbs) and cellular immunity related cytokines (IFN-γ and IL-4) as compared to monomeric E2. Specially, improved NAb response with more than tenfold increase in NAb titer against both CSFV Shimen and HZ-08 strains indicated better cross-protection against different genotypes. Collectively, this structure-based, self-assembling NP provides an attractive platform to improve the potency of subunit vaccine for emerging pathogens.


Assuntos
Antígenos Virais/farmacologia , Vírus da Febre Suína Clássica/imunologia , Peste Suína Clássica/prevenção & controle , Imunogenicidade da Vacina , Nanopartículas , Vacinas Virais/farmacologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Antígenos Virais/genética , Antígenos Virais/imunologia , Células Cultivadas , Peste Suína Clássica/sangue , Peste Suína Clássica/imunologia , Peste Suína Clássica/virologia , Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Estabilidade de Medicamentos , Feminino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Suínos , Temperatura , Vacinas de Subunidades/imunologia , Vacinas de Subunidades/farmacologia , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/farmacologia , Vacinas Virais/imunologia
14.
Protein Pept Lett ; 28(10): 1138-1147, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34132177

RESUMO

BACKGROUND: Brucellosis is a zoonotic disease that causes serious economic losses due to factors, such as miscarriages and decreased milk yield in animals. Existing live vaccines have some disadvantages, so effective vaccines need to be developed with new technological approaches. OBJECTIVE: The primary objectives of this study were the expression and purification of recombinant Omp25 fusion protein from B. abortus, and the evaluation of the effect of the Omp25 protein on cell viability and inflammatory response. METHODS: The omp25 gene region was amplified by a polymerase chain reaction and cloned into a Pet102/D-TOPO expression vector. The protein expression was carried out using the prokaryotic expression system. The recombinant Omp25 protein was purified with affinity chromatography followed by GPC (Gel Permeation Chromatography). The MTS assay and cytokine-release measurements were carried out to evaluate cell viability and inflammatory response, respectively. RESULTS: It was determined that doses of the recombinant Omp25 protein greater than 0.1 µg/mL are toxic to RAW cells. Doses of 1 µg/mL and lower significantly increased inflammation due to Nitric Oxide (NO) levels. ELISA results showed that IFN-γ was produced in stimulated RAW 264.7 cells at a dose that did not affect the viability (0.05 µg/mL). However, IL-12, which is known to have a dual role in the activation of macrophages, did not show a statistically significant difference at the same dose. CONCLUSION: Studies on cell viability and Th1-related cytokine release suggest Omp25 protein to be a promising candidate molecule for vaccine development.


Assuntos
Brucella abortus/genética , Brucelose/tratamento farmacológico , Proteínas de Membrana/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Vacinas Sintéticas/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Escherichia coli/química , Escherichia coli/genética , Humanos , Imunogenicidade da Vacina , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Óxido Nítrico/metabolismo , Células RAW 264.7 , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Desenvolvimento de Vacinas , Vacinas Sintéticas/química , Vacinas Sintéticas/genética
15.
Chem Rev ; 121(7): 3598-3626, 2021 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-33794090

RESUMO

The glycocalyx, a thick layer of carbohydrates, surrounds the cell wall of most bacterial and parasitic pathogens. Recognition of these unique glycans by the human immune system results in destruction of the invaders. To elicit a protective immune response, polysaccharides either isolated from the bacterial cell surface or conjugated with a carrier protein, for T-cell help, are administered. Conjugate vaccines based on isolated carbohydrates currently protect millions of people against Streptococcus pneumoniae, Haemophilus influenzae type b, and Neisseria meningitides infections. Active pharmaceutical ingredients (APIs) are increasingly discovered by medicinal chemistry and synthetic in origin, rather than isolated from natural sources. Converting vaccines from biologicals to pharmaceuticals requires a fundamental understanding of how the human immune system recognizes carbohydrates and could now be realized. To illustrate the chemistry-based approach to vaccine discovery, I summarize efforts focusing on synthetic glycan-based medicinal chemistry to understand the mammalian antiglycan immune response and define glycan epitopes for novel synthetic glycoconjugate vaccines against Streptococcus pneumoniae, Clostridium difficile, Klebsiella pneumoniae, and other bacteria. The chemical tools described here help us gain fundamental insights into how the human system recognizes carbohydrates and drive the discovery of carbohydrate vaccines.


Assuntos
Infecções Bacterianas/prevenção & controle , Glicocálix/química , Polissacarídeos/química , Vacinas Conjugadas/química , Vacinas Sintéticas/química , Animais , Clostridioides difficile , Glicoconjugados/química , Humanos , Klebsiella pneumoniae , Bibliotecas de Moléculas Pequenas/química , Streptococcus pneumoniae , Relação Estrutura-Atividade , Vacinas Conjugadas/farmacologia , Vacinas Sintéticas/farmacologia
16.
Front Immunol ; 12: 653092, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33815413

RESUMO

Q fever is caused by the obligate intracellular bacterium, Coxiella burnetii, a designated potential agent of bioterrorism because of its route of transmission, resistance to disinfectants, and low infectious dose. The only vaccine licensed for human use is Q-VAX® (Seqirus, licensed in Australia), a formalin-inactivated whole-cell vaccine, which produces severe local and systemic reactogenic responses in previously sensitized individuals. Accordingly, the U.S. Food and Drug Administration and other regulatory bodies around the world, have been reluctant to approve Q-VAX for widespread use. To obviate these adverse reactions, we prepared recombinant protein subunit vaccine candidates containing purified CBU1910, CBU0307, CBU0545, CBU0612, CBU0891, and CBU1398 proteins and TLR triagonist adjuvants. TLR triagonist adjuvants combine different TLR agonists to enhance immune responses to vaccine antigens. We tested both the protective efficacy and reactogenicity of our vaccine candidates in Hartley guinea pigs using intratracheal infection with live C. burnetii. While all of our candidates showed varying degrees of protection during challenge, local reactogenic responses were significantly reduced for one of our vaccine candidates when compared with a formalin-inactivated whole-cell vaccine. Our findings show that subunit vaccines combined with novel TLR triagonist adjuvants can generate protective immunity to C. burnetii infection while reducing reactogenic responses.


Assuntos
Adjuvantes Imunológicos/farmacologia , Vacinas Bacterianas/farmacologia , Coxiella burnetii/imunologia , Febre Q/prevenção & controle , Receptores Toll-Like/antagonistas & inibidores , Adjuvantes Imunológicos/uso terapêutico , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/farmacologia , Antígenos de Bactérias/uso terapêutico , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Vacinas Bacterianas/genética , Vacinas Bacterianas/uso terapêutico , Modelos Animais de Doenças , Cobaias , Humanos , Imunogenicidade da Vacina , Febre Q/imunologia , Febre Q/microbiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Vacinas de Subunidades/genética , Vacinas de Subunidades/farmacologia , Vacinas de Subunidades/uso terapêutico , Vacinas Sintéticas/genética , Vacinas Sintéticas/farmacologia , Vacinas Sintéticas/uso terapêutico
18.
Angew Chem Int Ed Engl ; 60(26): 14679-14692, 2021 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-33852172

RESUMO

Streptococcus suis bacteria are one of the most serious health problems for pigs and an emerging zoonotic agent in humans working in the swine industry. S. suis bacteria express capsular polysaccharides (CPS) a major bacterial virulence factor that define the serotypes. Oligosaccharides resembling the CPS of S. suis serotypes 2, 3, 9, and 14 have been synthesized, glycans related to serotypes 2 and 9 were placed on glycan array surfaces to screen blood from infected pigs. Lead antigens for the development of semi-synthetic S. suis serotypes 2 and 9 glycoconjugate veterinary vaccines were identified in this way.


Assuntos
Antibacterianos/farmacologia , Antígenos/imunologia , Glicoconjugados/farmacologia , Polissacarídeos Bacterianos/imunologia , Streptococcus suis/efeitos dos fármacos , Vacinas Sintéticas/farmacologia , Antibacterianos/química , Antibacterianos/imunologia , Antígenos/química , Descoberta de Drogas , Glicoconjugados/química , Glicoconjugados/imunologia , Polissacarídeos Bacterianos/química , Vacinas Sintéticas/química , Vacinas Sintéticas/imunologia
19.
Hum Gene Ther ; 32(11-12): 541-562, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33858231

RESUMO

Severe acute respiratory syndrome (SARS) is a newly emerging infectious disease (COVID-19) caused by the novel coronavirus SARS-coronavirus 2 (CoV-2). To combat the devastating spread of SARS-CoV-2, extraordinary efforts from numerous laboratories have focused on the development of effective and safe vaccines. Traditional live-attenuated or inactivated viral vaccines are not recommended for immunocompromised patients as the attenuated virus can still cause disease via phenotypic or genotypic reversion. Subunit vaccines require repeated dosing and adjuvant use to be effective, and DNA vaccines exhibit lower immune responses. mRNA vaccines can be highly unstable under physiological conditions. On the contrary, naturally antigenic viral vectors with well-characterized structure and safety profile serve as among the most effective gene carriers to provoke immune response via heterologous gene transfer. Viral vector-based vaccines induce both an effective cellular immune response and a humoral immune response owing to their natural adjuvant properties via transduction of immune cells. Consequently, viral vectored vaccines carrying the SARS-CoV-2 spike protein have recently been generated and successfully used to activate cytotoxic T cells and develop a neutralizing antibody response. Recent progress in SARS-CoV-2 vaccines, with an emphasis on gene therapy viral vector-based vaccine development, is discussed in this review.


Assuntos
Vacinas contra COVID-19/farmacologia , Vetores Genéticos , Vacinas Atenuadas/farmacologia , Vacinas Sintéticas/farmacologia , Proteínas Estruturais Virais/química , Adenoviridae/genética , Terapia Genética/métodos , Vetores Genéticos/química , Vetores Genéticos/genética , Humanos , Lentivirus/genética , SARS-CoV-2/genética , Vacinas de DNA/farmacologia , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo , Vacinas de mRNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...