Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.068
Filtrer
1.
Fish Shellfish Immunol ; 152: 109803, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39096980

RÉSUMÉ

Nervous necrosis virus (NNV) capsid protein plays an important role in producing viral particles without any genetic elements. Thus, NNV is a promising candidate for vaccine development and is widely used for constructing vaccines, including DNA, recombinant proteins, and virus-like particles (VLPs). Our study aimed to investigate the potential of NNV capsid protein (NNV) and NNV capsid protein fused to enhanced green fluorescent protein (NNV-EGFP) through VLP formation and whether their application can induce specific antibody responses against certain antigens. We focused on producing DNA and recombinant protein vaccines consisting of the genes for NNV, EGFP, and NNV-EGFP. The approach using NNV-EGFP allowed NNV to act as a carrier or inducer while EGFP was incorporated as part of the capsid protein, thereby enhancing the immune response. In vitro studies demonstrated that all DNA vaccines expressed in HINAE cells resulted in varying protein expression levels, with particularly low levels observed for pNNV and pNNV-EGFP. Consequently, structural proteins derived from HINAE cells could not be observed using transmission electron microscopy (TEM). In contrast, recombinant proteins of NNV and NNV-EGFP were expressed through the Escherichia coli expression system. TEM revealed that rNNV was assembled into VLPs with an approximate size of 30 nm, whereas rNNV-EGFP presented particles ranging from 10 nm to 50 nm in size. For the vaccination test, DNA vaccination marginally induced specific antibody responses in Japanese flounder compared to unvaccinated fish. Meanwhile, NNV and NNV-EGFP recombinant vaccines enhanced a greater anti-NNV antibody response than the others, whereas antibody responses against EGFP were also marginal. These results indicate that NNV capsid protein-based antigens, presenting as particles, play an important role in eliciting a specific anti-NNV antibody response and have the potential to improve fish immune responses.


Sujet(s)
Protéines de capside , Maladies des poissons , Nodaviridae , Vaccins antiviraux , Animaux , Nodaviridae/immunologie , Protéines de capside/immunologie , Protéines de capside/génétique , Maladies des poissons/immunologie , Maladies des poissons/prévention et contrôle , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/immunologie , Infections à virus à ARN/médecine vétérinaire , Infections à virus à ARN/immunologie , Infections à virus à ARN/prévention et contrôle , Vaccins à ADN/immunologie , Vaccins à ADN/administration et posologie , Développement de vaccin , Vaccins à pseudo-particules virales/immunologie , Vaccins à pseudo-particules virales/administration et posologie
2.
mSphere ; 9(8): e0028324, 2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39087764

RÉSUMÉ

In 2009, a novel swine-origin H1N1 virus emerged, causing a pandemic. The virus, known as H1N1pdm09, quickly displaced the circulating H1 lineage and became the dominant seasonal influenza A virus subtype infecting humans. Human-to-swine spillovers of the H1N1pdm09 have occurred frequently, and each occurrence has led to sustained transmission of the human-origin H1N1pdm09 within swine populations. In the present study, we developed a lipid nanoparticle-based DNA vaccine (LNP-DNA) containing the hemagglutinin gene of a swine-origin H1N1pdm09. In pigs, this LNP-DNA vaccine induced a robust antibody response after a single intramuscular immunization and protected the pigs against challenge infection with the homologous swine-origin H1N1pdm09 virus. In a mouse model, the LNP-DNA vaccine induced antibody and T-cell responses and protected mice against lethal challenge with a mouse-adapted human-origin H1N1pdm09 virus. These findings demonstrate the potential of the LNP-DNA vaccine to protect against both swine- and human-origin H1N1pdm09 viruses. IMPORTANCE: Swine influenza A virus (IAV) is widespread and causes significant economic losses to the swine industry. Moreover, bidirectional transmission of IAV between swine and humans commonly occurs. Once introduced into the swine population, human-origin IAV often reassorts with endemic swine IAV, resulting in reassortant viruses. Thus, it is imperative to develop a vaccine that is not only effective against IAV strains endemic in swine but also capable of preventing the spillover of human-origin IAV. In this study, we developed a lipid nanoparticle-encapsulated DNA plasmid vaccine (LNP-DNA) that demonstrates efficacy against both swine- and human-origin H1N1 viruses. The LNP-DNA vaccines are non-infectious and non-viable, meeting the criteria to serve as a vaccine platform for rapidly updating vaccines. Collectively, this LNP-DNA vaccine approach holds great potential for alleviating the impact of IAV on the swine industry and preventing the emergence of reassortant IAV strains.


Sujet(s)
Anticorps antiviraux , Sous-type H1N1 du virus de la grippe A , Vaccins antigrippaux , Grippe humaine , Nanoparticules , Infections à Orthomyxoviridae , Maladies des porcs , Vaccins à ADN , Animaux , Sous-type H1N1 du virus de la grippe A/immunologie , Sous-type H1N1 du virus de la grippe A/génétique , Vaccins à ADN/immunologie , Vaccins à ADN/administration et posologie , Suidae , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Vaccins antigrippaux/génétique , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/médecine vétérinaire , Nanoparticules/administration et posologie , Humains , Souris , Maladies des porcs/prévention et contrôle , Maladies des porcs/virologie , Maladies des porcs/immunologie , Anticorps antiviraux/sang , Grippe humaine/prévention et contrôle , Grippe humaine/immunologie , Grippe humaine/virologie , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Femelle , Souris de lignée BALB C , Liposomes/administration et posologie
3.
EBioMedicine ; 106: 105269, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39111250

RÉSUMÉ

BACKGROUND: Influenza viruses pose a persistent threat to global public health, necessitating the development of innovative and broadly effective vaccines. METHODS: This study focuses on a multiepitope vaccine (MEV) designed to provide broad-spectrum protection against different influenza viruses. The MEV, containing 19 B-cell linear epitopes, 7 CD4+ T cells, and 11 CD8+ T cells epitopes identified through enzyme-linked immunospot assay (ELISPOT) in influenza viruses infected mice, was administered through a regimen of two doses of DNA vaccine followed by one dose of a protein vaccine in C57BL/6 female mice. FINDINGS: Upon lethal challenge with both seasonal circulating strains (H1N1, H3N2, BV, and BY) and historical strains (H1N1-PR8 and H3N2-X31), MEV demonstrated substantial protection against different influenza seasonal strains, with partial efficacy against historical strains. Notably, the increased germinal centre B cells and antibody-secreting cells, along with robust T cell immune responses, highlighted the comprehensive immune defence elicited by MEV. Elevated hemagglutinin inhibition antibody was also observed against seasonal circulating and historical strains. Additionally, mice vaccinated with MEV exhibited significantly lower counts of inflammatory cells in the lungs compared to negative control groups. INTERPRETATION: Our results demonstrated the efficacy of a broad-spectrum MEV against influenza viruses in mice. Conducting long-term studies to evaluate the durability of MEV-induced immune responses and explore its potential application in diverse populations will offer valuable insights for the continued advancement of this promising vaccine. FUNDING: Funding bodies are described in the Acknowledgments section.


Sujet(s)
Déterminants antigéniques des lymphocytes B , Virus influenza B , Vaccins antigrippaux , Infections à Orthomyxoviridae , Animaux , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Souris , Virus influenza B/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Femelle , Déterminants antigéniques des lymphocytes B/immunologie , Virus de la grippe A/immunologie , Anticorps antiviraux/immunologie , Déterminants antigéniques des lymphocytes T/immunologie , Modèles animaux de maladie humaine , Souris de lignée C57BL , Vaccins à ADN/immunologie , Vaccins à ADN/administration et posologie , Saisons , Sous-type H3N2 du virus de la grippe A/immunologie , Humains
4.
Hum Vaccin Immunother ; 20(1): 2384189, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-39171509

RÉSUMÉ

Modified Vaccinia Ankara Bavarian Nordic (MVA-BN) as a smallpox and mpox vaccine has been approved in its liquid-frozen (LF) formulation in the US, Canada, and EU. A freeze-dried (FD) formulation may offer additional benefits, such as a longer shelf life and reduced dependence on cold chain storage and transport. In a phase 2 clinical trial, 651 vaccinia-naïve participants were vaccinated with two doses of MVA-BN LF or FD, 4 weeks apart. The objectives were to compare MVA-BN FD with LF in terms of vaccine-induced immune responses, safety, and reactogenicity. Non-inferiority of the immune response was assessed by the 95% CI of the geometric mean ratios. Both formulations induced robust vaccinia-specific humoral and cellular immune responses. At peak humoral responses (Week 6), geometric means of total antibody titers were 1096 (95% CI 1013, 1186) from the FD group and 877 (95% CI 804, 956) from the LF group, achieving the primary endpoint of non-inferiority of MVA-BN FD compared to MVA-BN LF. At peak cellular responses (Week 2), geometric means of T cell spot forming units were 449 (95% CI 341, 590) from the FD group and 316 (95% CI 234, 427) from the LF group. Both formulations of MVA-BN were well tolerated, with similar unsolicited AEs and solicited systemic reactions in both groups but slightly more local reactions in the FD group. No vaccine-related serious adverse events (SAEs) or vaccine-related AE of special interest were reported. The FD formulation of MVA-BN was shown to be equivalent to MVA-BN LF.


Sujet(s)
Anticorps antiviraux , Lyophilisation , Vaccin antivariolique , Humains , Vaccin antivariolique/immunologie , Vaccin antivariolique/effets indésirables , Vaccin antivariolique/administration et posologie , Femelle , Mâle , Adulte , Jeune adulte , Anticorps antiviraux/sang , Adulte d'âge moyen , Vaccins à ADN/immunologie , Vaccins à ADN/administration et posologie , Vaccins à ADN/effets indésirables , Immunité humorale , Immunité cellulaire , Adolescent , Variole/prévention et contrôle , Variole/immunologie , Congélation , Vaccins atténués
5.
Nat Commun ; 15(1): 6874, 2024 Aug 11.
Article de Anglais | MEDLINE | ID: mdl-39128904

RÉSUMÉ

Lymphoplasmacytic lymphoma (LPL) is an incurable low-grade lymphoma with no standard therapy. Nine asymptomatic patients treated with a first-in-human, neoantigen DNA vaccine experienced no dose limiting toxicities (primary endpoint, NCT01209871). All patients achieve stable disease or better, with one minor response, and median time to progression of 72+ months. Post-vaccine single-cell transcriptomics reveal dichotomous antitumor responses, with reduced tumor B-cells (tracked by unique B cell receptor) and their survival pathways, but no change in clonal plasma cells. Downregulation of human leukocyte antigen (HLA) class II molecules and paradoxical upregulation of insulin-like growth factor (IGF) by the latter suggest resistance mechanisms. Vaccine therapy activates and expands bone marrow T-cell clonotypes, and functional neoantigen-specific responses (secondary endpoint), but not co-inhibitory pathways or Treg, and reduces protumoral signaling by myeloid cells, suggesting favorable perturbation of the tumor immune microenvironment. Future strategies may require combinations of vaccines with agents targeting plasma cell subpopulations, or blockade of IGF-1 signaling or myeloid cell checkpoints.


Sujet(s)
Vaccins anticancéreux , Macroglobulinémie de Waldenström , Humains , Mâle , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Vaccins anticancéreux/administration et posologie , Adulte d'âge moyen , Femelle , Macroglobulinémie de Waldenström/immunologie , Macroglobulinémie de Waldenström/thérapie , Macroglobulinémie de Waldenström/génétique , Sujet âgé , Microenvironnement tumoral/immunologie , Médecine de précision/méthodes , Vaccins à ADN/immunologie , Vaccins à ADN/usage thérapeutique , Antigènes néoplasiques/immunologie , Lymphocytes B/immunologie
6.
Fish Shellfish Immunol ; 152: 109773, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39019124

RÉSUMÉ

Fish nocardiosis is a chronic disease mainly caused by Nocardia seriolae, which occurs in a variety of economically cultured freshwater and marine fish. Studies have shown that DNA vaccine is an effective treatment to protect fish from bacterial infection. In our previous experiment, an in vivo-induced gene of N. seriolae, encoding phosphoketolase (PK) family protein, was identified by in vivo-induced antigen technology. In the present study, the antigenic gene encoding PK family protein was analyzed by bioinformatics and further inserted into the eukaryotic expression vector pcDNA3.1-myc-his-A for DNA vaccine development. The immunological effects of pcDNA-PK DNA vaccine were assessed in hybrid snakehead (Channa maculata ♀ × Channa argus ♂), showing induction in several serum enzyme activity parameters (including LZM, SOD, ACP and AKP), increasing in specific-antibody IgM levels, as well as up-regulation in six immune-related genes (CD4, CD8α, TNFα, IL-1ß, MHCIα and MHCIIα). Moreover, an immune-protection with a relative survival rate was provided at 53.82 % following artificial challenge with N. seriolae in vaccinated fish in comparison to the control group. In summary, these results indicate that pcDNA-PK DNA vaccine could boost strong immune responses in hybrid snakehead and show preferably protective efficacy against N. seriolae, which may be applied in aquaculture to control fish nocardiosis.


Sujet(s)
Vaccins antibactériens , Maladies des poissons , Infections à Nocardia , Nocardia , Vaccins à ADN , Animaux , Nocardia/immunologie , Infections à Nocardia/médecine vétérinaire , Infections à Nocardia/immunologie , Infections à Nocardia/prévention et contrôle , Maladies des poissons/immunologie , Maladies des poissons/prévention et contrôle , Vaccins à ADN/immunologie , Vaccins antibactériens/immunologie , Aldehyde-lyases/génétique , Aldehyde-lyases/immunologie , Poissons/immunologie , Protéines bactériennes/immunologie , Protéines bactériennes/génétique
7.
Cancer Immunol Immunother ; 73(9): 178, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38954031

RÉSUMÉ

Intracranial tumors present a significant therapeutic challenge due to their physiological location. Immunotherapy presents an attractive method for targeting these intracranial tumors due to relatively low toxicity and tumor specificity. Here we show that SCIB1, a TRP-2 and gp100 directed ImmunoBody® DNA vaccine, generates a strong TRP-2 specific immune response, as demonstrated by the high number of TRP2-specific IFNγ spots produced and the detection of a significant number of pentamer positive T cells in the spleen of vaccinated mice. Furthermore, vaccine-induced T cells were able to recognize and kill B16HHDII/DR1 cells after a short in vitro culture. Having found that glioblastoma multiforme (GBM) expresses significant levels of PD-L1 and IDO1, with PD-L1 correlating with poorer survival in patients with the mesenchymal subtype of GBM, we decided to combine SCIB1 ImmunoBody® with PD-1 immune checkpoint blockade to treat mice harboring intracranial tumors expressing TRP-2 and gp100. Time-to-death was significantly prolonged, and this correlated with increased CD4+ and CD8+ T cell infiltration in the tissue microenvironment (TME). However, in addition to PD-L1 and IDO, the GBM TME was found to contain a significant number of immunoregulatory T (Treg) cell-associated transcripts, and the presence of such cells is likely to significantly affect clinical outcome unless also tackled.


Sujet(s)
Tumeurs du cerveau , Vaccins anticancéreux , Inhibiteurs de points de contrôle immunitaires , Récepteur-1 de mort cellulaire programmée , Vaccins à ADN , Animaux , Femelle , Humains , Souris , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/thérapie , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Lignée cellulaire tumorale , Glioblastome/immunologie , Glioblastome/thérapie , Glioblastome/traitement médicamenteux , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Immunothérapie/méthodes , Intramolecular oxidoreductases , Souris de lignée C57BL , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Vaccins à ADN/immunologie , Vaccins à ADN/usage thérapeutique , Mâle , Enfant , Adulte d'âge moyen
8.
Curr Microbiol ; 81(9): 279, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-39031239

RÉSUMÉ

Recent advancements in in vitro transcribed mRNA (IVT-mRNA) vaccine manufacturing have attracted considerable interest as advanced methods for combating viral infections. The respiratory mucosa is a primary target for pathogen attack, but traditional intramuscular vaccines are not effective in generating protective ion mucosal surfaces. Mucosal immunization can induce both systemic and mucosal immunity by effectively eliminating microorganisms before their growth and development. However, there are several biological and physical obstacles to the administration of genetic payloads, such as IVT-mRNA and DNA, to the pulmonary and nasal mucosa. Nucleic acid vaccine nanocarriers should effectively protect and load genetic payloads to overcome barriers i.e., biological and physical, at the mucosal sites. This may aid in the transfection of specific antigens, epithelial cells, and incorporation of adjuvants. In this review, we address strategies for delivering genetic payloads, such as nucleic acid vaccines, that have been studied in the past and their potential applications.


Sujet(s)
Immunité muqueuse , Nanoparticules , Vaccination , Humains , Animaux , Vaccination/méthodes , Vaccins à ADN/immunologie , Vaccins à ADN/administration et posologie , Vaccins à base d'acide nucléique/immunologie , Vaccins à base d'acide nucléique/génétique , Vaccins à base d'acide nucléique/administration et posologie
9.
Front Immunol ; 15: 1433185, 2024.
Article de Anglais | MEDLINE | ID: mdl-39081320

RÉSUMÉ

Tumor related angiogenesis is an attractive target in cancer therapeutic research due to its crucial role in tumor growth, invasion, and metastasis. Different agents were developed aiming to inhibit this process; however they had limited success. Cancer vaccines could be a promising tool in anti-cancer/anti-angiogenic therapy. Cancer vaccines aim to initiate an immune response against cancer cells upon presentation of tumor antigens which hopefully will result in the eradication of disease and prevention of its recurrence by inducing an efficient and long-lasting immune response. Different vaccine constructs have been developed to achieve this and they could include either protein-based or nucleic acid-based vaccines. Nucleic acid vaccines are simple and relatively easy to produce, with high efficiency and safety, thus prompting a high interest in the field. Different DNA vaccines have been developed to target crucial regulators of tumor angiogenesis. Most of them were successful in pre-clinical studies, mostly when used in combination with other therapeutics, but had limited success in the clinic. Apparently, different tumor evasion mechanisms and reduced immunogenicity still limit the potential of these vaccines and there is plenty of room for improvement. Nowadays, mRNA cancer vaccines are making remarkable progress due to improvements in the manufacturing technology and represent a powerful potential alternative. Apart from their efficiency, mRNA vaccines are simple and cheap to produce, can encompass multiple targets simultaneously, and can be quickly transferred from bench to bedside. mRNA vaccines have already accomplished amazing results in cancer clinical trials, thus ensuring a bright future in the field, although no anti-angiogenic mRNA vaccines have been described yet. This review aims to describe recent advances in anti-angiogenic DNA vaccine therapy and to provide perspectives for use of revolutionary approaches such are mRNA vaccines for anti-angiogenic treatments.


Sujet(s)
Vaccins anticancéreux , Tumeurs , Néovascularisation pathologique , Vaccins à ARNm , Humains , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/usage thérapeutique , Tumeurs/immunologie , Tumeurs/thérapie , Néovascularisation pathologique/immunologie , Animaux , Vaccins à ADN/immunologie , Antigènes néoplasiques/immunologie ,
10.
Vaccine ; 42(21): 126140, 2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39033079

RÉSUMÉ

Transmission-blocking vaccines interrupting malaria transmission within mosquitoes represent an ideal public health tool to eliminate malaria at the population level. Plasmodium falciparum and P. vivax account for more than 90% of the global malaria burden, co-endemic in many regions of the world. P25 and P48/45 are two leading candidates for both species and have shown promising transmission-blocking activity in preclinical and clinical studies. However, neither of these target antigens as individual vaccines has induced complete transmission inhibition in mosquitoes. In this study, we assessed immunogenicity of combination vaccines based on P25 and P48/45 using a DNA vaccine platform to broaden vaccine specificity against P. falciparum and P. vivax. Individual DNA vaccines encoding Pvs25, Pfs25, Pvs48/45 and Pfs48/45, as well as various combinations including (Pvs25 + Pvs48/45), (Pfs25 + Pfs48/45), (Pvs25 + Pfs25), and (Pvs48/45 + Pfs48/45), were evaluated in mice using in vivo electroporation. Potent antibody responses were induced in mice immunized with individual and combination DNA vaccines, and specific antibody responses were not compromised when combinations of DNA vaccines were evaluated against individual DNA vaccines. The anti-Pvs25 IgG from individual and combination groups revealed concentration-dependent transmission-reducing activity (TRA) in direct membrane feeding assays (DMFA) using blood from P. vivax-infected donors in Brazil and independently in ex vivo MFA using Pvs25-transgenic P. berghei. Similarly, anti-Pfs25 and anti-Pfs48/45 IgGs from mice immunized with Pfs25 and Pfs48/45 DNA vaccines individually and in various combinations revealed antibody dose-dependent TRA in standard membrane feeding assays (SMFA) using culture-derived P. falciparum gametocytes. However, antibodies induced by immunization with Pvs48/45 DNA vaccines were ineffective in DMFA and require further vaccine construct optimization, considering the possibility of induction of both transmission-blocking and transmission-enhancing antibodies revealed by competition ELISA. These studies provide a rationale for combining multiple antigens to simultaneously target transmission of malaria caused by P. falciparum and P. vivax.


Sujet(s)
Anticorps antiprotozoaires , Vaccins contre le paludisme , Paludisme à Plasmodium falciparum , Paludisme à Plasmodium vivax , Plasmodium falciparum , Plasmodium vivax , Vaccins à ADN , Vaccins contre le paludisme/immunologie , Vaccins contre le paludisme/administration et posologie , Animaux , Paludisme à Plasmodium falciparum/prévention et contrôle , Paludisme à Plasmodium falciparum/transmission , Paludisme à Plasmodium falciparum/immunologie , Plasmodium falciparum/immunologie , Plasmodium falciparum/génétique , Plasmodium vivax/immunologie , Plasmodium vivax/génétique , Paludisme à Plasmodium vivax/prévention et contrôle , Paludisme à Plasmodium vivax/transmission , Paludisme à Plasmodium vivax/immunologie , Souris , Vaccins à ADN/immunologie , Vaccins à ADN/administration et posologie , Anticorps antiprotozoaires/immunologie , Anticorps antiprotozoaires/sang , Femelle , Vaccins combinés/immunologie , Vaccins combinés/administration et posologie , Antigènes de protozoaire/immunologie , Antigènes de protozoaire/génétique , Protéines de protozoaire/immunologie , Protéines de protozoaire/génétique , Souris de lignée BALB C , Humains
11.
Acta Trop ; 257: 107302, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38959992

RÉSUMÉ

Toxoplasma gondii is an important protozoan pathogen, which can cause severe diseases in the newborns and immunocompromised individuals. Developing an effective vaccine against Toxoplasma infection is a critically important global health priority. Immunofluorescence staining analysis revealed that TgSAG2 and TgSRS2 are membrane associated and displayed on the surface of the parasite. Immunizations with pBud-SAG2, pBud-SRS2 and pBud-SAG2-SRS2 DNA vaccines significantly increased the production of specific IgG antibodies. Immunization with pBud-SAG2-SRS2 elicited cellular immune response with higher concentrations of IFN-γ and IL-4 compared to the control group. Antigen-specific lymphocyte proliferations in the pBud-SRS2 and pBud-SAG2-SRS2 groups were significantly higher compared to that in the control group. Furthermore, 30 % of mice immunized with pBud-SAG2-SRS2 survived after the challenge infection with virulent T. gondii RH tachyzoites. This study revealed that immunization with pBud-SAG2-SRS2 induced potent immune responses, and has the potential as a promising vaccine candidate for the control of T. gondii infection.


Sujet(s)
Anticorps antiprotozoaires , Antigènes de protozoaire , Immunoglobuline G , Protéines de protozoaire , Vaccins antiprotozoaires , Toxoplasma , Toxoplasmose animale , Vaccins à ADN , Animaux , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Vaccins à ADN/administration et posologie , Antigènes de protozoaire/immunologie , Antigènes de protozoaire/génétique , Protéines de protozoaire/immunologie , Protéines de protozoaire/génétique , Toxoplasma/immunologie , Toxoplasma/génétique , Anticorps antiprotozoaires/sang , Vaccins antiprotozoaires/immunologie , Vaccins antiprotozoaires/administration et posologie , Vaccins antiprotozoaires/génétique , Souris , Immunoglobuline G/sang , Femelle , Toxoplasmose animale/prévention et contrôle , Toxoplasmose animale/immunologie , Souris de lignée BALB C , Interféron gamma/immunologie , Modèles animaux de maladie humaine , Prolifération cellulaire , Interleukine-4/immunologie , Analyse de survie
12.
Microbiol Res ; 287: 127837, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39059097

RÉSUMÉ

Klebsiella pneumoniae, a prominent nosocomial pathogen, poses a critical global health threat due to its multidrug-resistant (MDR) and hypervirulent strains. This comprehensive review focuses into the complex approaches undertaken in the development of vaccines against K. pneumoniae. Traditional methods, such as whole-cell and ribosomal-based vaccines, are compared with modern strategies, including DNA and mRNA vaccines, and extracellular vesicles (EVs), among others. Each method presents unique advantages and challenges, emphasising the complexity of developing an effective vaccine against this pathogen. Significant advancements in computational tools and artificial intelligence (AI) have revolutionised antigen identification and vaccine design, enhancing the precision and efficiency of developing multiepitope-based vaccines. The review also highlights the potential of glycomics and immunoinformatics in identifying key antigenic components and elucidating immune evasion mechanisms employed by K. pneumoniae. Despite progress, challenges remain in ensuring the safety, efficacy, and manufacturability of these vaccines. Notably, EVs demonstrate promise due to their intrinsic adjuvant properties and ability to elicit robust immune responses, although concerns regarding inflammation and antigen variability persist. This review provides a critical overview of the current landscape of K. pneumoniae vaccine development, stressing the need for continued innovation and interdisciplinary collaboration to address this pressing public health issue. The integration of advanced computational methods and AI holds the potential to accelerate the development of effective immunotherapies, paving the way for novel vaccines against MDR K. pneumoniae.


Sujet(s)
Vaccins antibactériens , Infections à Klebsiella , Klebsiella pneumoniae , Développement de vaccin , Klebsiella pneumoniae/immunologie , Klebsiella pneumoniae/génétique , Humains , Vaccins antibactériens/immunologie , Infections à Klebsiella/prévention et contrôle , Infections à Klebsiella/immunologie , Infections à Klebsiella/microbiologie , Animaux , Vaccins à ADN/immunologie , Intelligence artificielle , Vésicules extracellulaires/immunologie , Antigènes bactériens/immunologie
13.
Egypt J Immunol ; 31(3): 95-112, 2024 07.
Article de Anglais | MEDLINE | ID: mdl-38995715

RÉSUMÉ

In this study, we aimed to evaluate the immunogenic profile of a chimeric DNA-based hepatitis C virus (HCV) vaccine candidate encoding the full-length viral core-E1-E2 (HCV-CE) fragment. The vaccine candidate was designed to uniformly express the HCV genotype 4 core-E1-E2 protein. The recombinant HCV-CE protein was bacterially expressed in C41 (DE3) cells, and then BALB/c mice were immunized with different combinations of DNA/DNA or DNA/protein prime/boost immunizations. The proper construction of our vaccine candidate was confirmed by specific amplification of the encoded fragments and basic local alignment search tool (BLAST) results of the nucleotide sequence, which revealed a high degree of similarity with several HCV serotypes/genotypes. The platform for bacterial expression was optimized to maximize the yield of the purified recombinant HCV-CE protein. The recombinant protein showed high specific antigenicity against the sera of HCV-infected patients according to the ELISA and western blot results. The predicted B- and T-cell epitopes showed high antigenic and interferon-γ (IFN-γ) induction potential, in addition to cross-genotype conservation and population coverage. The mice antisera further demonstrated a remarkable ability to capture 100% of the native viral antigens circulating in the sera of HCV patients, with no cross-reactivity detected in control sera. In conclusion, the proposed HCV vaccination strategy demonstrated promising potential regarding its safety, immunogenicity, and population coverage.


Sujet(s)
Hepacivirus , Hépatite C , Souris de lignée BALB C , Vaccins à ADN , Vaccins contre les hépatites virales , Animaux , Hepacivirus/immunologie , Hepacivirus/génétique , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Souris , Vaccins contre les hépatites virales/immunologie , Hépatite C/prévention et contrôle , Hépatite C/immunologie , Humains , Immunogénicité des vaccins/immunologie , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/génétique , Protéines du core viral/immunologie , Protéines du core viral/génétique , Femelle , Anticorps de l'hépatite C/immunologie , Anticorps de l'hépatite C/sang
14.
Int J Mol Sci ; 25(13)2024 Jun 23.
Article de Anglais | MEDLINE | ID: mdl-39000000

RÉSUMÉ

Somatostatin (SS) plays crucial regulatory roles in animal growth and reproduction by affecting the synthesis and secretion of growth hormone (GH). However, the mechanism by which SS regulates growth and development in goats is still unclear. In order to investigate the regulatory networks of the hypothalamus and pituitary in goats affected by SS DNA vaccines, in this study, we used a previously established oral attenuated Salmonella typhimurium SS DNA vaccine, X9241 (ptCS/2SS-asd), to treat wethers. We analyzed the protein changes in hypothalamic and pituitary tissues using a TMT-based proteomics approach. Additionally, we examined the metabolic profiles of the serum of control and immunized wethers through untargeted metabolomics using liquid chromatography-mass spectrometry (LC-MS). Key signaling pathways were identified based on differentially expressed metabolites (DEMs) and differentially expressed proteins (DEPs). Furthermore, the effect of critical DEPs on signaling pathways was confirmed through Western blotting (WB) experiments, which elucidated the mechanism of active SS immunization in wethers. A proteomics analysis revealed that the expression of 58 proteins in the hypothalamus and 124 in the pituitary gland was significantly altered following SS vaccine treatment (fold change > 1.2 or < 0.83, p < 0.05). In the hypothalamus, many DEPs were associated with gene ontology (GO) terms related to neuronal signaling. In contrast, most DEPs were associated with metabolic pathways. In the pituitary gland, the DEPs were largely related to immune and nutrient metabolism functions, with significant enrichment in KEGG pathways, particularly those involving the metabolic pathway, sphingolipid signaling, and the cGMP-PKG signaling pathway. A metabolomic analysis further showed that active SS immunization in wethers led to significant alterations in seven serum metabolites. Notably, the sphingolipid signaling pathway, secondary bile acid synthesis, sphingolipid metabolism, and lysine synthesis were significantly disrupted. SS vaccines induced marked changes in hypothalamic-pituitary proteins in wethers, facilitating alterations in their growth processes. This study not only provides insights into the mechanism of the SS gene in regulating GH secretion in wethers but also establishes a basis for hormone immunoregulation technology to enhance livestock production performance.


Sujet(s)
Capra , Hypothalamus , Hypophyse , Protéomique , Somatostatine , Vaccins à ADN , Animaux , Somatostatine/métabolisme , Protéomique/méthodes , Hypothalamus/métabolisme , Vaccins à ADN/immunologie , Hypophyse/métabolisme , Métabolomique/méthodes , Transduction du signal , Métabolome
15.
Biomaterials ; 311: 122647, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38878479

RÉSUMÉ

DNA technology has emerged as a promising route to accelerated manufacture of sequence agnostic vaccines. For activity, DNA vaccines must be protected and delivered to the correct antigen presenting cells. However, the physicochemical properties of the vector must be carefully tuned to enhance interaction with immune cells and generate sufficient immune response for disease protection. In this study, we have engineered a range of polymer-based nanocarriers based on the poly(beta-amino ester) (PBAE) polycation platform to investigate the role that surface poly(ethylene glycol) (PEG) density has on pDNA encapsulation, formulation properties and gene transfectability both in vitro and in vivo. We achieved this by synthesising a non-PEGylated and PEGylated PBAE and produced formulations containing these PBAEs, and mixed polyplexes to tune surface PEG density. All polymers and co-formulations produced small polyplex nanoparticles with almost complete encapsulation of the cargo in all cases. Despite high gene transfection in HEK293T cells, only the fully PEGylated and mixed formulations displayed significantly higher expression of the reporter gene than the negative control in dendritic cells. Further in vivo studies with a bivalent SARS-CoV-2 pDNA vaccine revealed that only the mixed formulation led to strong antigen specific T-cell responses, however this did not translate into the presence of serum antibodies indicating the need for further studies into improving immunisation with polymer delivery systems.


Sujet(s)
Vaccins contre la COVID-19 , Polyéthylène glycols , SARS-CoV-2 , Lymphocytes T , Vaccins à ADN , Humains , Vaccins à ADN/immunologie , Polyéthylène glycols/composition chimique , SARS-CoV-2/immunologie , Lymphocytes T/immunologie , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/composition chimique , Cellules HEK293 , Animaux , COVID-19/prévention et contrôle , COVID-19/immunologie , Polymères/composition chimique , Cellules dendritiques/immunologie , Souris , Femelle , Nanoparticules/composition chimique
16.
Front Immunol ; 15: 1347926, 2024.
Article de Anglais | MEDLINE | ID: mdl-38903517

RÉSUMÉ

Introduction: The HVTN 105 vaccine clinical trial tested four combinations of two immunogens - the DNA vaccine DNA-HIV-PT123, and the protein vaccine AIDSVAX B/E. All combinations induced substantial antibody and CD4+ T cell responses in many participants. We have now re-examined the intracellular cytokine staining flow cytometry data using the high-resolution SWIFT clustering algorithm, which is very effective for enumerating rare populations such as antigen-responsive T cells, and also determined correlations between the antibody and T cell responses. Methods: Flow cytometry samples across all the analysis batches were registered using the swiftReg registration tool, which reduces batch variation without compromising biological variation. Registered data were clustered using the SWIFT algorithm, and cluster template competition was used to identify clusters of antigen-responsive T cells and to separate these from constitutive cytokine producing cell clusters. Results: Registration strongly reduced batch variation among batches analyzed across several months. This in-depth clustering analysis identified a greater proportion of responders than the original analysis. A subset of antigen-responsive clusters producing IL-21 was identified. The cytokine patterns in each vaccine group were related to the type of vaccine - protein antigens tended to induce more cells producing IL-2 but not IFN-γ, whereas DNA vaccines tended to induce more IL-2+ IFN-γ+ CD4 T cells. Several significant correlations were identified between specific antibody responses and antigen-responsive T cell clusters. The best correlations were not necessarily observed with the strongest antibody or T cell responses. Conclusion: In the complex HVTN105 dataset, alternative analysis methods increased sensitivity of the detection of antigen-specific T cells; increased the number of identified vaccine responders; identified a small IL-21-producing T cell population; and demonstrated significant correlations between specific T cell populations and serum antibody responses. Multiple analysis strategies may be valuable for extracting the most information from large, complex studies.


Sujet(s)
Vaccins contre le SIDA , Lymphocytes T CD4+ , Cytokines , Cytométrie en flux , Infections à VIH , Humains , Vaccins contre le SIDA/immunologie , Lymphocytes T CD4+/immunologie , Cytométrie en flux/méthodes , Analyse de regroupements , Infections à VIH/immunologie , Infections à VIH/virologie , Cytokines/métabolisme , Cytokines/immunologie , Immunité humorale , Anticorps anti-VIH/immunologie , Anticorps anti-VIH/sang , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Vaccins à ADN/immunologie , Interleukines/immunologie
17.
J Acquir Immune Defic Syndr ; 96(4): 350-360, 2024 08 01.
Article de Anglais | MEDLINE | ID: mdl-38916429

RÉSUMÉ

BACKGROUND: An effective vaccine is required to end the HIV pandemic. We evaluated the safety and immunogenicity of a DNA (DNA-HIV-PT123) vaccine with low- or high-dose bivalent (TV1.C and 1086.C glycoprotein 120) subtype C envelope protein combinations, adjuvanted with MF59 or AS01B. METHODS: HIV Vaccine Trials Network (HVTN)108 was a randomized, placebo-controlled, double-blind, phase 1/2a trial conducted in the United States and South Africa. HIV-negative adults were randomly assigned to 1 of 7 intervention arms or placebo to assess DNA prime with DNA/protein/adjuvant boosts, DNA/protein/adjuvant co-administration, and low-dose protein/adjuvant regimens. HVTN111 trial participants who received an identical regimen were also included. Outcomes included safety and immunogenicity 2 weeks and 6 months after final vaccination. RESULTS: From June 2016 to July 2018, 400 participants were enrolled (N = 334 HVTN108, N = 66 HVTN111); 370 received vaccine and 30 received placebo. There were 48 grade 3 and 3 grade 4 reactogenicity events among 39/400 (9.8%) participants, and 32 mild/moderate-related adverse events in 23/400 (5.8%) participants. All intervention groups demonstrated high IgG response rates (>89%) and high magnitudes to HIV-1 Env gp120 and gp140 proteins; response rates for AS01B-adjuvanted groups approached 100%. V1V2 IgG magnitude, Fc-mediated functions, IgG3 Env response rates, and CD4+ T-cell response magnitudes and rates were higher in the AS01B-adjuvanted groups. The AS01B-adjuvanted low-dose protein elicited greater IgG responses than the higher protein dose. CONCLUSIONS: The vaccine regimens were generally well tolerated. Co-administration of DNA with AS01B-adjuvanted bivalent Env gp120 elicited the strongest humoral responses; AS01B-adjuvanted regimens elicited stronger CD4+ T-cell responses, justifying further evaluation.ClinicalTrials.gov registration: NCT02915016, registered 26 September 2016.


Sujet(s)
Vaccins contre le SIDA , Adjuvants immunologiques , Anticorps anti-VIH , Protéine d'enveloppe gp120 du VIH , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Polysorbates , Squalène , Vaccins à ADN , Humains , Vaccins contre le SIDA/immunologie , Vaccins contre le SIDA/administration et posologie , Vaccins contre le SIDA/effets indésirables , Vaccins à ADN/immunologie , Vaccins à ADN/administration et posologie , Vaccins à ADN/effets indésirables , Femelle , Mâle , Adulte , Squalène/administration et posologie , Polysorbates/administration et posologie , Protéine d'enveloppe gp120 du VIH/immunologie , Adjuvants immunologiques/administration et posologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Infections à VIH/immunologie , Infections à VIH/prévention et contrôle , Anticorps anti-VIH/sang , Méthode en double aveugle , Adulte d'âge moyen , Jeune adulte , Adjuvants vaccinaux/administration et posologie , République d'Afrique du Sud , Immunogénicité des vaccins , Adolescent , États-Unis
18.
Proc Natl Acad Sci U S A ; 121(25): e2322264121, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38865265

RÉSUMÉ

Despite the tremendous clinical potential of nucleic acid-based vaccines, their efficacy to induce therapeutic immune response has been limited by the lack of efficient local gene delivery techniques in the human body. In this study, we develop a hydrogel-based organic electronic device (µEPO) for both transdermal delivery of nucleic acids and in vivo microarrayed cell electroporation, which is specifically oriented toward one-step transfection of DNAs in subcutaneous antigen-presenting cells (APCs) for cancer immunotherapy. The µEPO device contains an array of microneedle-shaped electrodes with pre-encapsulated dry DNAs. Upon a pressurized contact with skin tissue, the electrodes are rehydrated, electrically triggered to release DNAs, and then electroporate nearby cells, which can achieve in vivo transfection of more than 50% of the cells in the epidermal and upper dermal layer. As a proof-of-concept, the µEPO technique is employed to facilitate transdermal delivery of neoantigen genes to activate antigen-specific immune response for enhanced cancer immunotherapy based on a DNA vaccination strategy. In an ovalbumin (OVA) cancer vaccine model, we show that high-efficiency transdermal transfection of APCs with OVA-DNAs induces robust cellular and humoral immune responses, including antigen presentation and generation of IFN-γ+ cytotoxic T lymphocytes with a more than 10-fold dose sparing over existing intramuscular injection (IM) approach, and effectively inhibits tumor growth in rodent animals.


Sujet(s)
Électroporation , Immunothérapie , Vaccins à ADN , Animaux , Vaccins à ADN/administration et posologie , Vaccins à ADN/immunologie , Électroporation/méthodes , Souris , Immunothérapie/méthodes , Administration par voie cutanée , Tumeurs/thérapie , Tumeurs/immunologie , Vaccins anticancéreux/immunologie , Vaccins anticancéreux/administration et posologie , Ovalbumine/immunologie , Ovalbumine/administration et posologie , Cellules présentatrices d'antigène/immunologie , Femelle , Souris de lignée C57BL , Humains , Vaccination/méthodes
19.
Sci Rep ; 14(1): 13865, 2024 06 15.
Article de Anglais | MEDLINE | ID: mdl-38879684

RÉSUMÉ

Severe acute respiratory syndrome coronavirus 2 had devastating consequences for human health. Despite the introduction of several vaccines, COVID-19 continues to pose a serious health risk due to emerging variants of concern. DNA vaccines gained importance during the pandemic due to their advantages such as induction of both arms of immune response, rapid development, stability, and safety profiles. Here, we report the immunogenicity and protective efficacy of a DNA vaccine encoding spike protein with D614G mutation (named pcoSpikeD614G) and define a large-scale production process. According to the in vitro studies, pcoSpikeD614G expressed abundant spike protein in HEK293T cells. After the administration of pcoSpikeD614G to BALB/c mice through intramuscular (IM) route and intradermal route using an electroporation device (ID + EP), it induced high level of anti-S1 IgG and neutralizing antibodies (P < 0.0001), strong Th1-biased immune response as shown by IgG2a polarization (P < 0.01), increase in IFN-γ levels (P < 0.01), and increment in the ratio of IFN-γ secreting CD4+ (3.78-10.19%) and CD8+ (5.24-12.51%) T cells. Challenging K18-hACE2 transgenic mice showed that pcoSpikeD614G administered through IM and ID + EP routes conferred 90-100% protection and there was no sign of pneumonia. Subsequently, pcoSpikeD614G was evaluated as a promising DNA vaccine candidate and scale-up studies were performed. Accordingly, a large-scale production process was described, including a 36 h fermentation process of E. coli DH5α cells containing pcoSpikeD614G resulting in a wet cell weight of 242 g/L and a three-step chromatography for purification of the pcoSpikeD614G DNA vaccine.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Vaccins contre la COVID-19 , COVID-19 , Souris de lignée BALB C , Mutation , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Vaccins à ADN , Vaccins à ADN/immunologie , Vaccins à ADN/administration et posologie , Vaccins à ADN/génétique , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Animaux , Humains , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Souris , COVID-19/prévention et contrôle , COVID-19/immunologie , Cellules HEK293 , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Femelle , Immunogénicité des vaccins , Immunoglobuline G/sang , Immunoglobuline G/immunologie
20.
ACS Biomater Sci Eng ; 10(7): 4374-4387, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38869358

RÉSUMÉ

DNA vaccines represent an innovative approach for the immunization of diverse diseases. However, their clinical trial outcomes are constrained by suboptimal transfection efficiency and immunogenicity. In this work, we present a universal methodology involving the codelivery of Toll-like receptor 7/8 agonists (TLR7/8a) and antigen gene using TLR7/8a-conjugated peptide-coated poly(ß-amino ester) (PBAE) nanoparticles (NPs) to augment delivery efficiency and immune response. Peptide-TLR7/8a-coated PBAE NPs exhibit advantageous biophysical attributes, encompassing diminutive particle dimensions, nearly neutral ζ potential, and stability in the physiological environment. This synergistic approach not only ameliorates the stability of plasmid DNA (pDNA) and gene delivery efficacy but also facilitates subsequent antigen production. Furthermore, under optimal formulation conditions, the TLR7/8a-conjugated peptide coated PBAE NPs exhibit a potent capacity to induce robust immune responses. Collectively, this nanoparticulate gene delivery system demonstrates heightened transfection efficacy, stability, biodegradability, immunostimulatory effect, and low toxicity, making it a promising platform for the clinical advancement of DNA vaccines.


Sujet(s)
Nanoparticules , Peptides , Récepteur de type Toll-7 , Récepteur de type Toll-8 , Vaccins à ADN , Vaccins à ADN/immunologie , Vaccins à ADN/administration et posologie , Récepteur de type Toll-8/immunologie , Récepteur de type Toll-8/agonistes , Récepteur de type Toll-7/agonistes , Récepteur de type Toll-7/immunologie , Animaux , Nanoparticules/composition chimique , Peptides/composition chimique , Peptides/immunologie , Humains , Souris , Femelle , Polymères/composition chimique , Plasmides/génétique , Plasmides/immunologie , Souris de lignée C57BL
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE