Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.078
Filtrar
1.
BMC Biotechnol ; 24(1): 71, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39350162

RESUMO

BACKGROUND: Human papillomavirus type 16 (HPV-16) infection is strongly associated with considerable parts of cervical, neck, and head cancers. Performed investigations have had moderate clinical success, so research to reach an efficient vaccine has been of great interest. In the present study, the immunization potential of a newly designed HPV-16 construct was evaluated in a mouse model. RESULTS: Initially, a construct containing HPV-16 mutant (m) E6/E7 fusion gene was designed and antigen produced in two platforms (i.e., DNA vaccine and recombinant protein). Subsequently, the immunogenicity of these platforms was investigated in five mice) C57BL/6 (groups based on several administration strategies. Three mice groups were immunized recombinant protein, DNA vaccine, and a combination of them, and two other groups were negative controls. The peripheral blood mononuclear cells (PBMCs) proliferation, Interleukin-5 (IL-5) and interferon-γ (IFN-γ) cytokines, IgG1 and IgG2a antibody levels were measured. After two weeks, TC-1 tumor cells were injected into all mice groups, and subsequently further analysis of tumor growth and metastasis and mice survival were performed according to the schedule. Overall, the results obtained from in vitro immunology and tumor cells challenging assays indicated the potential of the mE6/E7 construct as an HPV16 therapeutic vaccine candidate. The results demonstrated a significant increase in IFN-γ cytokine (P value < 0.05) in the Protein/Protein (D) and DNA/Protein (E) groups. This finding was in agreement with in vivo assays. Control groups show a 10.5-fold increase (P value < 0.001) and (C) DNA/DNA group shows a 2.5-fold increase (P value < 0.01) in tumor growth compared to D and E groups. Also, a significant increase in survival of D and E (P value < 0.001) and C (P value < 0.01) groups were observed. CONCLUSIONS: So, according to the findings, the recombinant protein could induce stronger protection compared to the DNA vaccine form. Protein/Protein and DNA/Protein are promising administration strategies for presenting this construct to develop an HPV-16 therapeutic vaccine candidate.


Assuntos
Papillomavirus Humano 16 , Camundongos Endogâmicos C57BL , Proteínas Oncogênicas Virais , Proteínas E7 de Papillomavirus , Vacinas contra Papillomavirus , Proteínas Repressoras , Vacinas de DNA , Animais , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/imunologia , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/imunologia , Camundongos , Proteínas Repressoras/genética , Proteínas Repressoras/imunologia , Vacinas de DNA/imunologia , Vacinas de DNA/genética , Vacinas de DNA/administração & dosagem , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/imunologia , Vacinas contra Papillomavirus/imunologia , Vacinas contra Papillomavirus/genética , Vacinas contra Papillomavirus/administração & dosagem , Feminino , Infecções por Papillomavirus/prevenção & controle , Infecções por Papillomavirus/virologia , Infecções por Papillomavirus/imunologia , Modelos Animais de Doenças , Humanos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia
2.
Cancer Res ; 84(19): 3173-3188, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39350665

RESUMO

Memory T cells play a key role in immune protection against cancer. Vaccine-induced tissue-resident memory T (TRM) cells in the lung have been shown to protect against lung metastasis. Identifying the source of lung TRM cells can help to improve strategies, preventing tumor metastasis. Here, we found that a prime-boost vaccination approach using intramuscular DNA vaccine priming, followed by intranasal live-attenuated influenza-vectored vaccine (LAIV) boosting induced higher frequencies of lung CD8+ TRM cells compared with other vaccination regimens. Vaccine-induced lung CD8+ TRM cells, but not circulating memory T cells, conferred significant protection against metastatic melanoma and mesothelioma. Central memory T (TCM) cells induced by the DNA vaccination were major precursors of lung TRM cells established after the intranasal LAIV boost. Single-cell RNA sequencing analysis indicated that transcriptional reprogramming of TCM cells for differentiation into TRM cells in the lungs started as early as day 2 post the LAIV boost. Intranasal LAIV altered the mucosal microenvironment to recruit TCM cells via CXCR3-dependent chemotaxis and induced CD8+ TRM-associated transcriptional programs. These results identified TCM cells as the source of vaccine-induced CD8+ TRM cells that protect against lung metastasis. Significance: Prime-boost vaccination shapes the mucosal microenvironment and reprograms central memory T cells to generate lung resident memory T cells that protect against lung metastasis, providing insights for the optimization of vaccine strategies.


Assuntos
Linfócitos T CD8-Positivos , Vacinas Anticâncer , Memória Imunológica , Neoplasias Pulmonares , Células T de Memória , Animais , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/patologia , Camundongos , Células T de Memória/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/administração & dosagem , Camundongos Endogâmicos C57BL , Vacinas de DNA/imunologia , Vacinas de DNA/administração & dosagem , Imunização Secundária/métodos , Vacinação/métodos , Feminino , Humanos , Administração Intranasal , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/administração & dosagem , Pulmão/imunologia , Pulmão/patologia
3.
Cancer Immunol Immunother ; 73(12): 248, 2024 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-39358555

RESUMO

BACKGROUND: Tumor-reactive T cells play a crucial role in anti-tumor responses, but T cells induced by DNA vaccination are time-consuming processes and exhibit limited anti-tumor efficacy. Therefore, we evaluated the anti-tumor effectiveness of reactive T cells elicited by electroporation (EP)-mediated DNA vaccine targeting epidermal growth factor receptor variant III (pEGFRvIII plasmid), in conjunction with adoptive cell therapy (ACT), involving the transfer of lymphocytes from a pEGFRvIII EP-vaccinated healthy donor. METHODS: The validation of the established pEGFRvIII plasmid and EGFRvIII-positive cell model was confirmed through immunofluorescence and western blot analysis. Flow cytometry and cytotoxicity assays were performed to evaluate the functionality of antigen-specific reactive T cells induced by EP-mediated pEGFRvIII vaccines, ACT, or their combination. The anti-tumor effectiveness of EP-mediated pEGFRvIII vaccines alone or combined with ACT was evaluated in the B16F10-EGFRvIII tumor model. RESULTS: EP-mediated pEGFRvIII vaccines elicited serum antibodies and a robust cellular immune response in both healthy and tumor-bearing mice. However, this response only marginally inhibited early-stage tumor growth in established tumor models. EP-mediated pEGFRvIII vaccination followed by adoptive transfer of lymphocytes from vaccinated healthy donors led to notable anti-tumor efficacy, attributed to the synergistic action of antigen-specific CD4+ Th1 cells supplemented by ACT and antigen-specific CD8+ T cells elicited by the EP-mediated DNA vaccination. CONCLUSIONS: Our preclinical studies results demonstrate an enhanced anti-tumor efficacy of EP-mediated DNA vaccination boosted with adoptively transferred, vaccinated healthy donor-derived allogeneic lymphocytes.


Assuntos
Vacinas Anticâncer , Eletroporação , Vacinas de DNA , Animais , Vacinas de DNA/imunologia , Eletroporação/métodos , Camundongos , Vacinas Anticâncer/imunologia , Camundongos Endogâmicos C57BL , Imunoterapia Adotiva/métodos , Feminino , Humanos , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Modelos Animais de Doenças , Linhagem Celular Tumoral , Células Alógenas/imunologia , Receptores ErbB/imunologia
4.
Front Immunol ; 15: 1445209, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39346922

RESUMO

Introduction: Epstein-Barr virus (EBV) is an oncogenic human herpesvirus associated with ~350,000 cases of lymphoid and epithelial malignancies every year, and is etiologically linked to infectious mononucleosis and multiple sclerosis. Despite four decades of research, no EBV vaccine candidate has yet reached licensure. Most previous vaccine attempts focused on a single viral entry glycoprotein, gp350, but recent data from clinical and pre-clinical studies, and the elucidation of viral entry mechanisms, support the inclusion of multiple entry glycoproteins in EBV vaccine design. Methods: Here we generated a modified vaccinia Ankara (MVA)-vectored EBV vaccine, MVA-EBV5-2, that targets five EBV entry glycoproteins, gp350, gB, and the gp42gHgL complex. We characterized the genetic and translational stability of the vaccine, followed by immunogenicity assessment in BALB/c mice and rhesus lymphocryptovirus-negative rhesus macaques as compared to a gp350-based MVA vaccine. Finally, we assessed the efficacy of MVA-EBV5-2-immune rhesus serum at preventing EBV infection in human CD34+ hematopoietic stem cell-reconstituted NSG mice, under two EBV challenge doses. Results: The MVA-EBV5-2 vaccine was genetically and translationally stable over 10 viral passages as shown by genetic and protein expression analysis, and when administered to female and male BALB/c mice, elicited serum EBV-specific IgG of both IgG1 and IgG2a subtypes with neutralizing activity in vitro. In Raji B cells, this neutralizing activity outperformed that of serum from mice immunized with a monovalent MVA-vectored gp350 vaccine. Similarly, MVA-EBV5-2 elicited EBV-specific IgG in rhesus macaques that were detected in both serum and saliva of immunized animals, with serum antibodies demonstrating neutralizing activity in vitro that outperformed serum from MVA-gp350-immunized macaques. Finally, pre-treatment with serum from MVA-EBV5-2-immunized macaques resulted in fewer EBV-infected mice in the two challenge experiments than pretreatment with serum from pre-immune macaques or macaques immunized with the monovalent gp350-based vaccine. Discussion: These results support the inclusion of multiple entry glycoproteins in EBV vaccine design and position our vaccine as a strong candidate for clinical translation.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Infecções por Vírus Epstein-Barr , Herpesvirus Humano 4 , Macaca mulatta , Animais , Humanos , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/prevenção & controle , Camundongos , Herpesvirus Humano 4/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Camundongos Endogâmicos BALB C , Vacinas de DNA/imunologia , Feminino , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Vetores Genéticos/genética , Vaccinia virus/imunologia , Vaccinia virus/genética
5.
Viruses ; 16(9)2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39339952

RESUMO

Herpes simplex virus (HSV) has coevolved with Homo sapiens for over 100,000 years, maintaining a tenacious presence by establishing lifelong, incurable infections in over half the human population. As of 2024, an effective prophylactic or therapeutic vaccine for HSV remains elusive. In this review, we independently screened PubMed, EMBASE, Medline, and Google Scholar for clinically relevant articles on HSV vaccines. We identified 12 vaccines from our literature review and found promising candidates across various classes, including subunit vaccines, live vaccines, DNA vaccines, and mRNA vaccines. Notably, several vaccines-SL-V20, HF10, VC2, and mRNA-1608-have shown promising preclinical results, suggesting that an effective HSV vaccine may be within reach. Additionally, several other vaccines such as GEN-003 (a subunit vaccine from Genocea), HerpV (a subunit vaccine from Agenus), 0ΔNLS/RVx201 (a live-attenuated replication-competent vaccine from Rational Vaccines), HSV 529 (a replication-defective vaccine from Sanofi Pasteur), and COR-1 (a DNA-based vaccine from Anteris Technologies) have demonstrated potential in clinical trials. However, GEN-003 and HerpV have not advanced further despite promising results. Continued progress with these candidates brings us closer to a significant breakthrough in preventing and treating HSV infections.


Assuntos
Vacinas contra o Vírus do Herpes Simples , Herpes Simples , Simplexvirus , Vacinação , Humanos , Herpes Simples/prevenção & controle , Herpes Simples/imunologia , Herpes Simples/virologia , Vacinas contra o Vírus do Herpes Simples/imunologia , Vacinas contra o Vírus do Herpes Simples/administração & dosagem , Vacinas contra o Vírus do Herpes Simples/genética , Animais , Simplexvirus/genética , Simplexvirus/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de DNA/imunologia , Vacinas Atenuadas/imunologia , Erradicação de Doenças
6.
Front Immunol ; 15: 1455730, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39234239

RESUMO

Introduction: Various COVID-19 vaccine trials have shown that vaccines can successfully prevent symptomatic cases of COVID-19 and death. Head-to-head comparisons help to better understand the immune response characteristics of different COVID-19 vaccines in humans. Methods: We randomly selected 20 participants from each of five ongoing Phase II trials of COVID-19 vaccines. Here, SARS-CoV 2-specific immune responses to DNA vaccine (INO-4800), mRNA vaccine (BNT162b2), Adenovirus-vectored vaccine (CONVIDECIA), Protein subunit vaccine (Recombinant COVID- 19 Vaccine (Sf9 Cells)), Inactivated Vaccine (KCONVAC) were examined longitudinally in healthy adults between Jan 15, 2021 and July 5, 2021 for 6 months. RBD-IgG titres were detected by ELISA, neutralising antibody titer were detected by pseudoviral neutralization and immune cell response were detected by flow cytometry. Results: At the first visit (V1), 100% of individuals who received the BNT162b2, CONVIDECIA, or KCONVAC vaccines experienced seroconversion of neutralizing and binding antibodies in the serum. Except for the Recombinant COVID-19 Vaccine (Sf9 Cells) vaccine having the highest neutralizing antibody GMT at the second visit (although there was no statistically significant difference in geometric mean titers between V1 and V2), the rest of the vaccines had the highest levels of binding antibodies and neutralizing antibodies at V1. The neutralizing antibodies GMT of all vaccines showed a significant decrease at V3 compared to V1. The neutralizing antibody GMT against the omicron variant of all vaccines at V1 showed a significant decrease compared to the wild strain. We observed statistically significant differences in Tcm cells and RBD-specific memory B cells among various vaccines. Discussion: BNT162b2 (mRNA vaccine) exhibits the highest antibody levels among the five vaccines evaluated, regardless of whether the target is the wild-type virus or its variants. However, its cellular immune response may be weaker compared to CONVIDECIA (adenovirus type 5 vector vaccine).


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Vacinas contra COVID-19 , COVID-19 , Imunidade Celular , Imunidade Humoral , SARS-CoV-2 , Humanos , Vacinas contra COVID-19/imunologia , Adulto , COVID-19/imunologia , COVID-19/prevenção & controle , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , SARS-CoV-2/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Masculino , Feminino , China , Pessoa de Meia-Idade , Adulto Jovem , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de DNA/imunologia , Vacina BNT162/imunologia , Imunogenicidade da Vacina , Vacinas de Produtos Inativados/imunologia
7.
Sci Rep ; 14(1): 21742, 2024 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-39289418

RESUMO

Vaccine manufacturing fosters the prevention, control, and eradication of infectious diseases. Recombinant DNA and in vitro (IVT) mRNA vaccine manufacturing technologies were enforced to combat the recent pandemic. Despite the impact of these technologies, there exists no scientific announcement that compares them. Digital Shadows are employed in this study to simulate each technology, investigating root cause deviations, technical merits, and liabilities, evaluating cost scenarios. Under this lens we provide an unbiased, advanced comparative technoeconomic study, one that determines which of these manufacturing platforms are suited for the two types of vaccines considered (monoclonal antibodies or antigens). We find recombinant DNA technology to exhibit higher Profitability Index due to lower capital and starting material requirements, pertaining to lower Minimum Selling Price per Dose values, delivering products of established quality. However, the potency of the mRNA, the streamlined and scalable synthetic processes involved and the raw material availability, facilitate faster market penetration and product flexibility, constituting these vaccines preferable whenever short product development cycles become a necessity.


Assuntos
RNA Mensageiro , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Humanos , DNA Recombinante/genética , Vacinas/imunologia , Vacinas de DNA/imunologia , Vacinas de DNA/genética , Anticorpos Monoclonais/imunologia , Desenvolvimento de Vacinas
8.
J Immunotoxicol ; 21(1): 2400624, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39319829

RESUMO

Infections caused by the influenza virus lead to both epidemic and pandemic outbreaks in humans and animals. Owing to their rapid production, safety, and stability, DNA vaccines represent a promising avenue for eliciting immunity and thwarting viral infections. While DNA vaccines have demonstrated substantial efficacy in murine models, their effectiveness in larger animals remains subdued. This limitation may be addressed by augmenting the immunogenicity of DNA-based vaccines. In the investigation here, protein expression was enhanced via codon optimization and then mouse cytotoxic T-lymphocyte antigen 4 (CTLA-4) was harnessed as a modulatory adjunct to bind directly to antigen-presenting cells. Further, the study evaluated the immunogenicity of two variants of the hemagglutinin (HA) antigen, i.e. the full-length and the C-terminal deletion versions. The study findings revealed that the codon-optimized HA gene (pcHA) led to increased protein synthesis, as evidenced by elevated mRNA levels. Codon optimization also significantly bolstered both cellular and humoral immune responses. In cytokine assays, all plasmid constructs, particularly pCTLA4-cHA, induced robust interferon (IFN)-γ production, while interleukin (IL)-4 levels remained uniformly non-significant. Mice immunized with pcHA displayed an augmented presence of IFNγ+ T-cells, underscoring the enhanced potency of the codon-optimized HA vaccine. Contrarily, CTLA-4-fused DNA vaccines did not significantly amplify the immune response.


Assuntos
Antígeno CTLA-4 , Códon , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Vacinas contra Influenza , Infecções por Orthomyxoviridae , Vacinas de DNA , Animais , Vacinas de DNA/imunologia , Vacinas de DNA/genética , Camundongos , Antígeno CTLA-4/genética , Antígeno CTLA-4/imunologia , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/administração & dosagem , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Códon/genética , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Humanos , Feminino , Camundongos Endogâmicos BALB C , Modelos Animais de Doenças , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Vírus da Influenza A Subtipo H1N1/imunologia
9.
Front Immunol ; 15: 1420304, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39267752

RESUMO

Despite the decrease in mortality and morbidity due to SARS-CoV-2 infection, the incidence of infections due to Omicron subvariants of SARS-CoV-2 remains high. The mutations acquired by these subvariants, mainly concentrated in the receptor-binding domain (RBD), have caused a shift in infectivity and transmissibility, leading to a loss of effectiveness of the first authorized COVID-19 vaccines, among other reasons, by neutralizing antibody evasion. Hence, the generation of new vaccine candidates adapted to Omicron subvariants is of special interest in an effort to overcome this immune evasion. Here, an optimized COVID-19 vaccine candidate, termed MVA-S(3P_BA.1), was developed using a modified vaccinia virus Ankara (MVA) vector expressing a full-length prefusion-stabilized SARS-CoV-2 spike (S) protein from the Omicron BA.1 variant. The immunogenicity and efficacy induced by MVA-S(3P_BA.1) were evaluated in mice in a head-to-head comparison with the previously generated vaccine candidates MVA-S(3P) and MVA-S(3Pbeta), which express prefusion-stabilized S proteins from Wuhan strain and Beta variant, respectively, and with a bivalent vaccine candidate composed of a combination of MVA-S(3P) and MVA-S(3P_BA.1). The results showed that all four vaccine candidates elicited, after a single intramuscular dose, protection of transgenic K18-hACE2 mice challenged with SARS-CoV-2 Omicron BA.1, reducing viral loads, histopathological lesions, and levels of proinflammatory cytokines in the lungs. They also elicited anti-S IgG and neutralizing antibodies against various Omicron subvariants, with MVA-S(3P_BA.1) and the bivalent vaccine candidate inducing higher titers. Additionally, an intranasal immunization in C57BL/6 mice with all four vaccine candidates induced systemic and mucosal S-specific CD4+ and CD8+ T-cell and humoral immune responses, and the bivalent vaccine candidate induced broader immune responses, eliciting antibodies against the ancestral Wuhan strain and different Omicron subvariants. These results highlight the use of MVA as a potent and adaptable vaccine vector against new emerging SARS-CoV-2 variants, as well as the promising feature of combining multivalent MVA vaccine candidates.


Assuntos
Anticorpos Antivirais , Vacinas contra COVID-19 , COVID-19 , Imunidade Celular , Imunidade Humoral , Camundongos Transgênicos , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Animais , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/genética , SARS-CoV-2/imunologia , Vacinas contra COVID-19/imunologia , COVID-19/prevenção & controle , COVID-19/imunologia , Camundongos , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Humanos , Enzima de Conversão de Angiotensina 2/imunologia , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Feminino , Vacinas de DNA/imunologia , Vaccinia virus/imunologia , Vaccinia virus/genética , Imunogenicidade da Vacina
10.
Nanomedicine (Lond) ; 19(21-22): 1779-1799, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39140594

RESUMO

Aim: To develop a trivalent DNA vaccine candidate encapsulated in Chitosan-TPP nanoparticles against hand foot and mouth disease (HFMD) and assess its immunogenicity in mice.Materials & methods: Trivalent plasmid carrying the VP1 and VP2 genes of EV-A71, VP1 gene of CV-A16 was encapsulated in Chitosan-TPP nanoparticles through ionic gelation. In vitro characterization and in vivo immunization studies of the CS-TPP-NPs (pIRES-VP121) were performed.Results: Mice administered with CS-TPP NPs (pIRES-VP121) intramuscularly were observed to have the highest IFN-γ response. Sera from mice immunized with the naked pDNA and CS-TPP-NPs (pIRES-VP121) demonstrated good viral clearance against wild-type EV-A71 and CV-A16 in RD cells.Conclusion: CS-TPP-NPs (pIRES-VP121) could serve as a prototype for future development of multivalent HFMD DNA vaccine candidates.


[Box: see text].


Assuntos
Quitosana , Enterovirus Humano A , Doença de Mão, Pé e Boca , Nanopartículas , Vacinas de DNA , Animais , Vacinas de DNA/imunologia , Vacinas de DNA/administração & dosagem , Quitosana/química , Nanopartículas/química , Camundongos , Enterovirus Humano A/imunologia , Doença de Mão, Pé e Boca/prevenção & controle , Doença de Mão, Pé e Boca/imunologia , Camundongos Endogâmicos BALB C , Feminino , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Humanos , Plasmídeos , Interferon gama/metabolismo , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/química , Polifosfatos
11.
JCI Insight ; 9(18)2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39088271

RESUMO

BACKGROUNDAn HIV-1 DNA vaccine composed of 7 highly conserved, structurally important elements (conserved elements, CE) of p24Gag was tested in a phase I randomized, double-blind clinical trial (HVTN 119, NCT03181789) in people without HIV. DNA vaccination of CE prime/CE+p55Gag boost was compared with p55Gag.METHODSTwo groups (n = 25) received 4 DNA vaccinations (CE/CE+p55Gag or p55Gag) by intramuscular injection/electroporation, including IL-12 DNA adjuvant. The placebo group (n = 6) received saline. Participants were followed for safety and tolerability. Immunogenicity was assessed for T cell and antibody responses.RESULTSBoth regimens were safe and generally well tolerated. The p24CE vaccine was immunogenic and significantly boosted by CE+p55Gag (64% CD4+, P = 0.037; 42% CD8+, P = 0.004). CE+p55Gag induced responses to 5 of 7 CE, compared with only 2 CE by p55Gag DNA, with a higher response to CE5 in 30% of individuals (P = 0.006). CE+p55Gag induced significantly higher CD4+ CE T cell breadth (0.68 vs. 0.22 CE; P = 0.029) and a strong trend for overall T cell breadth (1.14 vs. 0.52 CE; P = 0.051). Both groups developed high cellular and humoral responses. p24CE vaccine-induced CD4+ CE T cell responses correlated (P = 0.007) with p24Gag antibody responses.CONCLUSIONThe CE/CE+p55Gag DNA vaccine induced T cell responses to conserved regions in p24Gag, increasing breadth and epitope recognition throughout p55Gag compared with p55Gag DNA. Vaccines focusing immune responses by priming responses to highly conserved regions could be part of a comprehensive HIV vaccine strategy.TRIAL REGISTRATIONClinical Trials.gov NCT03181789FUNDINGHVTN, NIAID/NIH.


Assuntos
Vacinas contra a AIDS , Infecções por HIV , HIV-1 , Vacinas de DNA , Produtos do Gene gag do Vírus da Imunodeficiência Humana , Humanos , Vacinas de DNA/imunologia , Vacinas de DNA/administração & dosagem , HIV-1/imunologia , Vacinas contra a AIDS/imunologia , Vacinas contra a AIDS/administração & dosagem , Feminino , Adulto , Masculino , Método Duplo-Cego , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Pessoa de Meia-Idade , Adulto Jovem , Linfócitos T/imunologia , Anticorpos Anti-HIV/imunologia , Vacinação/métodos , Imunogenicidade da Vacina , Linfócitos T CD4-Positivos/imunologia
12.
Antiviral Res ; 230: 105978, 2024 10.
Artigo em Inglês | MEDLINE | ID: mdl-39117282

RESUMO

Seasonal influenza is an annually severe crisis for global public health, and an ideal influenza vaccine is expected to provide broad protection against constantly drifted strains. Compared to highly flexible hemagglutinin (HA), increasing data have demonstrated that neuraminidase (NA) might be a potential target against influenza variants. In the present study, a series of genetic algorithm-based mosaic NA were designed, and then cloned into recombinant DNA and replication-defective Vesicular Stomatitis Virus (VSV) vector as a novel influenza vaccine candidate. Our Results showed that DNA prime/VSV boost strategy elicited a robust NA-specific Th1-dominated immune response, but the traditional inactivated influenza vaccine elicited a Th2-dominated immune response. More importantly, the superior NA-specific immunity induced by our strategy could confer both a full protection against lethal homologous influenza challenge and a partial protection against heterologous influenza infection. These findings will provide insights on designing NA-based universal vaccine strategy against influenza variants.


Assuntos
Vacinas contra Influenza , Neuraminidase , Infecções por Orthomyxoviridae , Neuraminidase/imunologia , Neuraminidase/genética , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/genética , Animais , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Camundongos , Linfócitos T/imunologia , Camundongos Endogâmicos BALB C , Feminino , Humanos , Influenza Humana/prevenção & controle , Influenza Humana/imunologia , Influenza Humana/virologia , Vacinas de DNA/imunologia , Vacinas de DNA/genética , Células Th1/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue
13.
Nanomedicine (Lond) ; 19(24): 2027-2043, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39110059

RESUMO

The post COVID-19 pandemic era has emerged with more efficient vaccines, all based on genetic materials. However, to expand the use of nucleic components as vaccines, a new generation of nanosystems particularly constructed to increase RNA/DNA stability, half-life and facilitate administration are still required. This review highlights novel developments in mRNA and pDNA vaccines formulated into nanostructures exclusively composed by biopolymeric materials. Recent advances suggest that a new generation of vaccines may arise by adapting the structural features of biopolymers with the effectiveness of nucleic acids. The advantages offered by biopolymers, such as increased stability and targeting ability may cause a revolution in the immunization field for offering promptly adaptable and effective formulations for worldwide distribution.


[Box: see text].


Assuntos
COVID-19 , SARS-CoV-2 , Vacinas de DNA , Vacinas de DNA/imunologia , Vacinas de DNA/química , Vacinas de DNA/administração & dosagem , Humanos , Biopolímeros/química , COVID-19/prevenção & controle , SARS-CoV-2/imunologia , Nanoestruturas/química , Vacinas contra COVID-19/química , Vacinas contra COVID-19/imunologia , Vacinas de mRNA , Animais
14.
Expert Rev Vaccines ; 23(1): 796-811, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39188013

RESUMO

INTRODUCTION: Global outbreaks involving mpox clade IIb began in mid-2022. Today, clade IIb and clade I outbreaks continue. Reliable mpox vaccines can prevent serious mpox disease and death. AREAS COVERED: Globally, two vaccines hold mpox indications, regardless of mpox viral clade: MVA-BN (Bavarian Nordic) and LC16m8 (KM Biologics). This review summarizes the human and pivotal animal data establishing safety and efficacy for MVA-BN and LC16m8, including real-world evidence gathered during mpox outbreaks from 2022 through 2024. EXPERT OPINION: Some regulatory decisions for MVA-BN and LC16m8 followed pathways based on surrogate outcomes, including lethal-challenge studies in nonhuman primates, among other atypical aspects. Nonetheless, MVA-BN and LC16m8 hold unencumbered registration in multiple countries. Effectiveness of MVA-BN as primary preventive vaccination (PPV) in humans against clade IIb mpox is clear from real-world studies; effectiveness of LC16m8 against clade IIb is likely from surrogate endpoints. Effectiveness of MVA-BN and LC16m8 as PPV against more-lethal clade I is likely, based on animal-challenge studies with multiple orthopoxvirus species and other studies. Both vaccines have solid safety records. MVA-BN's replication incompetence favors adoption, whereas LC16m8 has more pediatric data. Additional real-world evidence, in additional geographic settings and special populations (e.g. pregnancy, immune suppression, atopic dermatitis), is needed.


Situation Mpox outbreaks spread globally in 2022, hospitalizing many people. Many recent mpox cases in Africa occur in children. Two vaccines, known as MVA-BN and LC16m8, can help prevent mpox.MVA-BN MVA-BN protects animals from lethal doses of mpox and similar viruses. During outbreaks, MVA-BN lowered the chance of mpox disease by 62% to 85%. In people already exposed to mpox, MVA-BN reduced disease risk by 20%. MVA-BN may help reduce how serious mpox cases are, even if this vaccine does not block infection fully. MVA-BN cannot grow inside the body, making it very safe, even in children. Side effects include pain, redness, swelling, and itching. Some people feel muscle pain, headache, fatigue, nausea, or chills after vaccination. Several million people have received MVA-BN so far, including thousands of people living with HIV.LC16m8 LC16m8 protects animals from lethal doses of mpox and similar viruses. There are not much data about LC16m8 used during mpox outbreaks. LC16m8 contains a weakened virus. Side effects include fever, fatigue, redness, swollen lymph nodes, and itching. Vaccine virus can spread to other parts of the body. Over 90,000 people have received LC16m8 so far. No significant safety signals were found after these doses, including 50,000 children. People who are immunosuppressed, have certain skin diseases, or are pregnant should not be given LC16m8.Mpox vaccine recommendations Health officials recommend mpox vaccine for people at risk, including children.


Assuntos
Mpox , Vacinas Virais , Animais , Humanos , Surtos de Doenças/prevenção & controle , Vacinação/métodos , Eficácia de Vacinas , Vacinas de DNA/imunologia , Vacinas de DNA/administração & dosagem , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Mpox/prevenção & controle
15.
PLoS Negl Trop Dis ; 18(8): e0012411, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39207951

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is a newly identified tick-borne viral hemorrhagic fever caused by Dabie Banda virus (DBV). The virus was first discovered in eastern China in 2009 and is now considered an infectious disease with a mortality rate ranging from 6.3% to 30%. The best strategy for controlling SFTS is to develop effective vaccines. However, no approved vaccines are currently available to prevent this disease, despite the number of extensive and in-depth studies conducted on DBV in the past few years. This review focuses on the structure of DBV and the induced host immune responses which are the fundamental factors in vaccine development, and thoroughly summarizes the current research progress on DBV vaccines. The developing DBV vaccines include protein subunit vaccines, live attenuated vaccines, recombinant virus vector vaccines, and DNA vaccines. At present, almost all candidate vaccines for DBV are in the laboratory development or preclinical stages. There remain challenges in successfully developing clinically approved DBV vaccines.


Assuntos
Vacinas Virais , Humanos , Vacinas Virais/imunologia , Animais , Vacinas Atenuadas/imunologia , Desenvolvimento de Vacinas , Febre Grave com Síndrome de Trombocitopenia/prevenção & controle , Febre Grave com Síndrome de Trombocitopenia/imunologia , Phlebovirus/imunologia , Phlebovirus/genética , Vacinas de DNA/imunologia , Vacinas de Subunidades Antigênicas/imunologia
16.
Hum Vaccin Immunother ; 20(1): 2384189, 2024 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-39171509

RESUMO

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.


Assuntos
Anticorpos Antivirais , Liofilização , Vacina Antivariólica , Humanos , Vacina Antivariólica/imunologia , Vacina Antivariólica/efeitos adversos , Vacina Antivariólica/administração & dosagem , Feminino , Masculino , Adulto , Adulto Jovem , Anticorpos Antivirais/sangue , Pessoa de Meia-Idade , Vacinas de DNA/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/efeitos adversos , Imunidade Humoral , Imunidade Celular , Adolescente , Varíola/prevenção & controle , Varíola/imunologia , Congelamento , Vacinas Atenuadas
17.
Hum Vaccin Immunother ; 20(1): 2395680, 2024 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-39208856

RESUMO

We have previously reported two single-agent phase I trials, evaluating the dose or schedule, of a DNA vaccine (pTVG-HP) encoding prostatic acid phosphatase (PAP) administered with GM-CSF as the adjuvant. These were in patients with PSA-recurrent, radiographically non-metastatic, prostate cancer (PCa). We report here the long-term safety and overall survival of these patients. Specifically, 22 patients with non-metastatic, castration-sensitive PCa (nmCSPC) were treated with pTVG-HP, 100-1500 µg, administered over 12 weeks and followed for 15 y. 17 patients with non-metastatic castration-resistant PCa (nmCRPC) were treated with 100 µg pTVG-HP with different schedules of administration over 1 y and followed for 5 y. No adverse events were detected in long-term follow-up from either trial that were deemed possibly related to vaccination. Patients with nmCSPC had a median overall survival of 12.3 y, with 5/22 (23%) alive at 15 y. 8/22 (36%) died due to prostate cancer with a median survival of 11.0 y, and 9/22 (41%) died of other causes. Patients with nmCRPC had a median overall survival of 4.5 y, with 8/17 (47%) alive at 5 y. The presence of T-cells specific for the PAP target antigen was detectable in 6/10 (60%) individuals with nmCSPC, and 3/5 (60%) individuals with nmCRPC, many years after immunization. The detection of immune responses to the vaccine target years after immunization suggests durable immunity can be elicited in patients using a DNA vaccine encoding a tumor-associated antigen.Trial Registration: NCT00582140 and NCT00849121.


Assuntos
Vacinas Anticâncer , Antígeno Prostático Específico , Neoplasias da Próstata , Vacinas de DNA , Humanos , Masculino , Vacinas de DNA/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/efeitos adversos , Antígeno Prostático Específico/imunologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/administração & dosagem , Idoso , Seguimentos , Neoplasias da Próstata/imunologia , Pessoa de Meia-Idade , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/efeitos adversos , Resultado do Tratamento , Idoso de 80 Anos ou mais , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Recidiva Local de Neoplasia , Análise de Sobrevida , Fosfatase Ácida , Proteínas Tirosina Fosfatases/imunologia
18.
Fish Shellfish Immunol ; 153: 109841, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39173984

RESUMO

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.


Assuntos
Bass , Infecções por Vírus de DNA , Doenças dos Peixes , Vacinas de DNA , Vacinas Virais , Animais , Vacinas de DNA/imunologia , Vacinas de DNA/administração & dosagem , Doenças dos Peixes/prevenção & controle , Doenças dos Peixes/imunologia , Bass/imunologia , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Infecções por Vírus de DNA/veterinária , Infecções por Vírus de DNA/prevenção & controle , Infecções por Vírus de DNA/imunologia , Vacinas de Produtos Inativados/imunologia , Vacinas de Produtos Inativados/administração & dosagem , Imunidade Humoral , Ranavirus/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Imunidade Celular
19.
Microb Pathog ; 195: 106871, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39163919

RESUMO

The H9N2 avian influenza virus (AIV) is spreading worldwide. Presence of H9N2 virus tends to increase the chances of infection with other pathogens which can lead to more serious economic losses. In a previous study, a regulated delayed lysis Salmonella vector was used to deliver a DNA vaccine named pYL233 encoding M1 protein, mosaic HA protein and chicken GM-CSF adjuvant. To further increase its efficiency, chitosan as a natural adjuvant was applied in this study. The purified plasmid pYL233 was coated with chitosan to form a DNA containing nanoparticles (named CS233) by ionic gel method and immunized by intranasal boost immunization in birds primed by oral administration with Salmonella strain. The CS233 DNA nanoparticle has a particle size of about 150 nm, with an encapsulation efficiency of 93.2 ± 0.12 % which protected the DNA plasmid from DNase I digestion and could be stable for a period of time at 37°. After intranasal boost immunization, the CS233 immunized chickens elicited higher antibody response, elevated CD4+ T cells and CD8+ T cells activation and increased T-lymphocyte proliferation, as well as increased productions of IL-4 and IFN-γ. After challenge, chickens immunized with CS233 resulted in the lowest levels of pulmonary virus titer and viral shedding as compared to the other challenge groups. The results showed that the combination of intranasal immunization with chitosan-coated DNA vaccine and oral immunization with regulatory delayed lytic Salmonella strain could enhance the immune response and able to provide protection against H9N2 challenge.


Assuntos
Administração Intranasal , Anticorpos Antivirais , Galinhas , Quitosana , Imunidade Celular , Vírus da Influenza A Subtipo H9N2 , Vacinas contra Influenza , Influenza Aviária , Plasmídeos , Vacinas de DNA , Eliminação de Partículas Virais , Animais , Vírus da Influenza A Subtipo H9N2/imunologia , Vírus da Influenza A Subtipo H9N2/genética , Vacinas de DNA/imunologia , Vacinas de DNA/administração & dosagem , Influenza Aviária/prevenção & controle , Influenza Aviária/imunologia , Galinhas/imunologia , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/administração & dosagem , Anticorpos Antivirais/sangue , Plasmídeos/genética , Nanopartículas , Imunização Secundária , Linfócitos T CD8-Positivos/imunologia , Adjuvantes Imunológicos/administração & dosagem , Interferon gama , Interleucina-4 , Adjuvantes de Vacinas , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/virologia , Linfócitos T CD4-Positivos/imunologia , Salmonella/imunologia , Salmonella/genética
20.
Int Immunopharmacol ; 141: 112956, 2024 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-39168022

RESUMO

DNA vaccines are prospective for their efficient manufacturing process, but their immunogenicity is limited as they cannot efficiently induce CD8+ T cell responses. A promising approach is to induce cross-presentation by targeting antigens to DCs. Flt3L can expand the number of type 1 conventional DCs and thereby improve cross-presentation. In this study, we first constructed a DNA vaccine expressing soluble PD1 and found that the therapeutic effect of targeting DCs with only the sPD1 vaccine was limited. When combined the vaccine with Flt3L, the anti-tumor effect was significantly enhanced. Considering the complexity of tumors and that a single method may not be able to activate a large number of effective CD8+ T cells, we combined different drugs and the vaccine with Flt3L based on the characteristics of different tumors. In 4T1 model, we reduced Tregs through cyclophosphamide. In Panc02 model, we increased activated DCs by using aCD40. Both strategies triggered strong CD8+ T cell responses and significantly improved the therapeutic effect. Our study provides important support for the clinical exploration of DC-targeted DNA vaccines in combination with Flt3L.


Assuntos
Linfócitos T CD8-Positivos , Vacinas Anticâncer , Células Dendríticas , Proteínas de Membrana , Camundongos Endogâmicos BALB C , Receptor de Morte Celular Programada 1 , Vacinas de DNA , Vacinas de DNA/imunologia , Animais , Células Dendríticas/imunologia , Receptor de Morte Celular Programada 1/imunologia , Vacinas Anticâncer/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteínas de Membrana/imunologia , Proteínas de Membrana/genética , Feminino , Camundongos , Linhagem Celular Tumoral , Humanos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...