Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.539
Filtrar
1.
BMC Biotechnol ; 24(1): 71, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39350162

RESUMEN

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.


Asunto(s)
Papillomavirus Humano 16 , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales , Proteínas E7 de Papillomavirus , Vacunas contra Papillomavirus , Proteínas Represoras , Vacunas de ADN , Animales , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/inmunología , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/inmunología , Ratones , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Vacunas de ADN/inmunología , Vacunas de ADN/genética , Vacunas de ADN/administración & dosificación , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/inmunología , Vacunas contra Papillomavirus/inmunología , Vacunas contra Papillomavirus/genética , Vacunas contra Papillomavirus/administración & dosificación , Femenino , Infecciones por Papillomavirus/prevención & control , Infecciones por Papillomavirus/virología , Infecciones por Papillomavirus/inmunología , Modelos Animales de Enfermedad , Humanos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología
2.
Cancer Res ; 84(19): 3173-3188, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39350665

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos , Vacunas contra el Cáncer , Memoria Inmunológica , Neoplasias Pulmonares , Células T de Memoria , Animales , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/patología , Ratones , Células T de Memoria/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/administración & dosificación , Ratones Endogámicos C57BL , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Inmunización Secundaria/métodos , Vacunación/métodos , Femenino , Humanos , Administración Intranasal , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Pulmón/inmunología , Pulmón/patología
3.
Front Immunol ; 15: 1457327, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39421744

RESUMEN

Introduction: To comprehensively identify and provide an overview of in vivo or clinical studies of nucleic acids (NA)-based vaccines against TB we included human or animal studies of NA vaccines for the prevention or treatment of TB and excluded in vitro or in silico research, studies of microorganisms other than M. tuberculosis, reviews, letters, and low-yield reports. Methods: We searched PubMed, Scopus, Embase, selected Web of Science and ProQuest databases, Google Scholar, eLIBRARY.RU, PROSPERO, OSF Registries, Cochrane CENTRAL, EU Clinical Trials Register, clinicaltrials.gov, and others through WHO International Clinical Trials Registry Platform Search Portal, AVMA and CABI databases, bioRxiv, medRxiv, and others through OSF Preprint Archive Search. We searched the same sources and Google for vaccine names (GX-70) and scanned reviews for references. Data on antigenic composition, delivery systems, adjuvants, and vaccine efficacy were charted and summarized descriptively. Results: A total of 18,157 records were identified, of which 968 were assessed for eligibility. No clinical studies were identified. 365 reports of 345 animal studies were included in the review. 342 (99.1%) studies involved DNA vaccines, and the remaining three focused on mRNA vaccines. 285 (82.6%) studies used single-antigen vaccines, while 48 (13.9%) used multiple antigens or combinations with adjuvants. Only 12 (3.5%) studies involved multiepitope vaccines. The most frequently used antigens were immunodominant secretory antigens (Ag85A, Ag85B, ESAT6), heat shock proteins, and cell wall proteins. Most studies delivered naked plasmid DNA intramuscularly without additional adjuvants. Only 4 of 17 studies comparing NA vaccines to BCG after M. tuberculosis challenge demonstrated superior protection in terms of bacterial load reduction. Some vaccine variants showed better efficacy compared to BCG. Systematic review registration: https://osf.io/, identifier F7P9G.


Asunto(s)
Vacunas contra la Tuberculosis , Tuberculosis , Vacunas de ADN , Vacunas de ARNm , Humanos , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Animales , Vacunas contra la Tuberculosis/inmunología , Vacunas contra la Tuberculosis/administración & dosificación , Tuberculosis/prevención & control , Tuberculosis/inmunología , Vacunas de ARNm/inmunología , Mycobacterium tuberculosis/inmunología , Eficacia de las Vacunas
4.
Microb Pathog ; 195: 106871, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39163919

RESUMEN

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.


Asunto(s)
Administración Intranasal , Anticuerpos Antivirales , Pollos , Quitosano , Inmunidad Celular , Subtipo H9N2 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Aviar , Plásmidos , Vacunas de ADN , Esparcimiento de Virus , Animales , Subtipo H9N2 del Virus de la Influenza A/inmunología , Subtipo H9N2 del Virus de la Influenza A/genética , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Gripe Aviar/prevención & control , Gripe Aviar/inmunología , Pollos/inmunología , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Anticuerpos Antivirales/sangre , Plásmidos/genética , Nanopartículas , Inmunización Secundaria , Linfocitos T CD8-positivos/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Interferón gamma , Interleucina-4 , Adyuvantes de Vacunas , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Linfocitos T CD4-Positivos/inmunología , Salmonella/inmunología , Salmonella/genética
5.
Nanomedicine (Lond) ; 19(21-22): 1779-1799, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39140594

RESUMEN

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].


Asunto(s)
Quitosano , Enterovirus Humano A , Enfermedad de Boca, Mano y Pie , Nanopartículas , Vacunas de ADN , Animales , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Quitosano/química , Nanopartículas/química , Ratones , Enterovirus Humano A/inmunología , Enfermedad de Boca, Mano y Pie/prevención & control , Enfermedad de Boca, Mano y Pie/inmunología , Ratones Endogámicos BALB C , Femenino , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Humanos , Plásmidos , Interferón gamma/metabolismo , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/química , Polifosfatos
6.
Fish Shellfish Immunol ; 153: 109841, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39173984

RESUMEN

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


Asunto(s)
Lubina , Infecciones por Virus ADN , Enfermedades de los Peces , Vacunas de ADN , Vacunas Virales , Animales , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Enfermedades de los Peces/prevención & control , Enfermedades de los Peces/inmunología , Lubina/inmunología , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Infecciones por Virus ADN/veterinaria , Infecciones por Virus ADN/prevención & control , Infecciones por Virus ADN/inmunología , Vacunas de Productos Inactivados/inmunología , Vacunas de Productos Inactivados/administración & dosificación , Inmunidad Humoral , Ranavirus/inmunología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Inmunidad Celular
7.
Hum Vaccin Immunother ; 20(1): 2395680, 2024 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-39208856

RESUMEN

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.


Asunto(s)
Vacunas contra el Cáncer , Antígeno Prostático Específico , Neoplasias de la Próstata , Vacunas de ADN , Humanos , Masculino , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/efectos adversos , Antígeno Prostático Específico/inmunología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/administración & dosificación , Anciano , Estudios de Seguimiento , Neoplasias de la Próstata/inmunología , Persona de Mediana Edad , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/efectos adversos , Resultado del Tratamiento , Anciano de 80 o más Años , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Recurrencia Local de Neoplasia , Análisis de Supervivencia , Fosfatasa Ácida , Proteínas Tirosina Fosfatasas/inmunología
8.
Fish Shellfish Immunol ; 152: 109803, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39096980

RESUMEN

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.


Asunto(s)
Proteínas de la Cápside , Enfermedades de los Peces , Nodaviridae , Vacunas Virales , Animales , Nodaviridae/inmunología , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/genética , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/prevención & control , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/inmunología , Infecciones por Virus ARN/veterinaria , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/prevención & control , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Desarrollo de Vacunas , Vacunas de Partículas Similares a Virus/inmunología , Vacunas de Partículas Similares a Virus/administración & dosificación
9.
JCI Insight ; 9(18)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39088271

RESUMEN

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.


Asunto(s)
Vacunas contra el SIDA , Infecciones por VIH , VIH-1 , Vacunas de ADN , Productos del Gen gag del Virus de la Inmunodeficiencia Humana , Humanos , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , VIH-1/inmunología , Vacunas contra el SIDA/inmunología , Vacunas contra el SIDA/administración & dosificación , Femenino , Adulto , Masculino , Método Doble Ciego , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Persona de Mediana Edad , Adulto Joven , Linfocitos T/inmunología , Anticuerpos Anti-VIH/inmunología , Vacunación/métodos , Inmunogenicidad Vacunal , Linfocitos T CD4-Positivos/inmunología
10.
Expert Rev Vaccines ; 23(1): 796-811, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39188013

RESUMEN

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.


Asunto(s)
Mpox , Vacunas Virales , Animales , Humanos , Brotes de Enfermedades/prevención & control , Vacunación/métodos , Eficacia de las Vacunas , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Mpox/prevención & control
11.
Hum Vaccin Immunother ; 20(1): 2384189, 2024 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-39171509

RESUMEN

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.


Asunto(s)
Anticuerpos Antivirales , Liofilización , Vacuna contra Viruela , Humanos , Vacuna contra Viruela/inmunología , Vacuna contra Viruela/efectos adversos , Vacuna contra Viruela/administración & dosificación , Femenino , Masculino , Adulto , Adulto Joven , Anticuerpos Antivirales/sangre , Persona de Mediana Edad , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/efectos adversos , Inmunidad Humoral , Inmunidad Celular , Adolescente , Viruela/prevención & control , Viruela/inmunología , Congelación , Vacunas Atenuadas
12.
mSphere ; 9(8): e0028324, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39087764

RESUMEN

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.


Asunto(s)
Anticuerpos Antivirales , Subtipo H1N1 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Nanopartículas , Infecciones por Orthomyxoviridae , Enfermedades de los Porcinos , Vacunas de ADN , Animales , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/genética , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Porcinos , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/veterinaria , Nanopartículas/administración & dosificación , Humanos , Ratones , Enfermedades de los Porcinos/prevención & control , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/inmunología , Anticuerpos Antivirales/sangre , Gripe Humana/prevención & control , Gripe Humana/inmunología , Gripe Humana/virología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Femenino , Ratones Endogámicos BALB C , Liposomas/administración & dosificación
13.
EBioMedicine ; 106: 105269, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39111250

RESUMEN

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.


Asunto(s)
Epítopos de Linfocito B , Virus de la Influenza B , Vacunas contra la Influenza , Infecciones por Orthomyxoviridae , Animales , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Ratones , Virus de la Influenza B/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/inmunología , Femenino , Epítopos de Linfocito B/inmunología , Virus de la Influenza A/inmunología , Anticuerpos Antivirales/inmunología , Epítopos de Linfocito T/inmunología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Estaciones del Año , Subtipo H3N2 del Virus de la Influenza A/inmunología , Humanos
14.
Nanomedicine (Lond) ; 19(24): 2027-2043, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39110059

RESUMEN

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].


Asunto(s)
COVID-19 , SARS-CoV-2 , Vacunas de ADN , Vacunas de ADN/inmunología , Vacunas de ADN/química , Vacunas de ADN/administración & dosificación , Humanos , Biopolímeros/química , COVID-19/prevención & control , SARS-CoV-2/inmunología , Nanoestructuras/química , Vacunas contra la COVID-19/química , Vacunas contra la COVID-19/inmunología , Vacunas de ARNm , Animales
15.
Vaccine ; 42(21): 126140, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39033079

RESUMEN

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.


Asunto(s)
Anticuerpos Antiprotozoarios , Vacunas contra la Malaria , Malaria Falciparum , Malaria Vivax , Plasmodium falciparum , Plasmodium vivax , Vacunas de ADN , Vacunas contra la Malaria/inmunología , Vacunas contra la Malaria/administración & dosificación , Animales , Malaria Falciparum/prevención & control , Malaria Falciparum/transmisión , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Plasmodium falciparum/genética , Plasmodium vivax/inmunología , Plasmodium vivax/genética , Malaria Vivax/prevención & control , Malaria Vivax/transmisión , Malaria Vivax/inmunología , Ratones , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Anticuerpos Antiprotozoarios/inmunología , Anticuerpos Antiprotozoarios/sangre , Femenino , Vacunas Combinadas/inmunología , Vacunas Combinadas/administración & dosificación , Antígenos de Protozoos/inmunología , Antígenos de Protozoos/genética , Proteínas Protozoarias/inmunología , Proteínas Protozoarias/genética , Ratones Endogámicos BALB C , Humanos
16.
Curr Microbiol ; 81(9): 279, 2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-39031239

RESUMEN

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.


Asunto(s)
Inmunidad Mucosa , Nanopartículas , Vacunación , Humanos , Animales , Vacunación/métodos , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Vacunación Basada en Ácidos Nucleicos/inmunología , Vacunación Basada en Ácidos Nucleicos/genética , Vacunación Basada en Ácidos Nucleicos/administración & dosificación
17.
J Fish Dis ; 47(11): e14002, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39075840

RESUMEN

Globally, streptococcal disease caused by Streptococcus agalactiae is known for its high mortality rate, which severely limits the development of the tilapia breeding industry. As a third-generation vaccine, DNA vaccines have shown great application prospects in the prevention and control of aquatic diseases, but their low immunogenicity limits their development. The combination of DNA vaccines and molecular adjuvants proved to be an effective method for inducing protective immunity. This study constructed recombinant plasmids encoding tilapia HSP70 and IL-1ß genes (pcHSP70 and pcIL-1ß) to verify their effectiveness as molecular adjuvants for S. agalactiae DNA vaccine (pcSIP) in the immunized tilapia model. The results revealed that serum-specific IgM production, enzyme activities, and immune-related gene expression in tilapia immunized with pcSIP plus pcHSP70 or pcIL-1ß were significantly higher than those in tilapia immunized with pcSIP alone. It is worth noting that combination with molecular adjuvants improved the immune protection of DNA vaccines, with a relative percentage survival (RPS) of 51.72% (pcSIP plus pcHSP70) and 44.83% (pcSIP plus pcIL-1ß), respectively, compared with that of pcSIP alone (24.14%). Thus, our study indicated that HSP70 and IL-1ß in tilapia are promising molecular adjuvants of the DNA vaccine in controlling S. agalactiae infection.


Asunto(s)
Adyuvantes Inmunológicos , Cíclidos , Enfermedades de los Peces , Proteínas HSP70 de Choque Térmico , Interleucina-1beta , Infecciones Estreptocócicas , Streptococcus agalactiae , Vacunas de ADN , Animales , Enfermedades de los Peces/prevención & control , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/microbiología , Streptococcus agalactiae/inmunología , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Infecciones Estreptocócicas/prevención & control , Infecciones Estreptocócicas/veterinaria , Infecciones Estreptocócicas/inmunología , Cíclidos/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/inmunología , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Vacunas Estreptocócicas/inmunología , Vacunas Estreptocócicas/administración & dosificación
18.
Acta Trop ; 257: 107302, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38959992

RESUMEN

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.


Asunto(s)
Anticuerpos Antiprotozoarios , Antígenos de Protozoos , Inmunoglobulina G , Proteínas Protozoarias , Vacunas Antiprotozoos , Toxoplasma , Toxoplasmosis Animal , Vacunas de ADN , Animales , Vacunas de ADN/inmunología , Vacunas de ADN/genética , Vacunas de ADN/administración & dosificación , Antígenos de Protozoos/inmunología , Antígenos de Protozoos/genética , Proteínas Protozoarias/inmunología , Proteínas Protozoarias/genética , Toxoplasma/inmunología , Toxoplasma/genética , Anticuerpos Antiprotozoarios/sangre , Vacunas Antiprotozoos/inmunología , Vacunas Antiprotozoos/administración & dosificación , Vacunas Antiprotozoos/genética , Ratones , Inmunoglobulina G/sangre , Femenino , Toxoplasmosis Animal/prevención & control , Toxoplasmosis Animal/inmunología , Ratones Endogámicos BALB C , Interferón gamma/inmunología , Modelos Animales de Enfermedad , Proliferación Celular , Interleucina-4/inmunología , Análisis de Supervivencia
19.
Vaccine ; 42(24): 126065, 2024 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-38880692

RESUMEN

Toxoplasma gondii is an obligate intracellular parasite that can infect a variety of mammals including humans and causes toxoplasmosis. Unfortunately, a protective and safe vaccine against toxoplasmosis hasn't been developed yet. In this study, we developed a DNA vaccine encoding the SRS13 protein and immunized BALB/c mice thrice with pVAX1-SRS13 through the intramuscular route (IM) or intradermally using an electroporation device (ID + EP). The immunogenicity of pVAX1-SRS13 was analyzed by ELISA, Western blot, cytokine ELISA, and flow cytometry. The protective efficacy of the pVAX1-SRS13 was investigated by challenging mice orally with T. gondii PRU strain tissue cysts. The results revealed that pVAX1-SRS13 administered through IM or ID + EP routes induced high level of anti-SRS13 IgG antibody responses (P = 0.0037 and P < 0.0001). The IFN-γ level elicited by the pVAX1-SRS13 (ID + EP) was significantly higher compared to the control group (P = 0.00159). In mice administered with pVAX1-SRS13 (ID + EP), CD8+ cells secreting IFN-γ was significantly higher compared to pVAX1-SRS13 (IM) (P = 0.0035) and the control group (P = 0.0068). Mice vaccinated with the SRS13 DNA vaccine did not induce significant IL-4 level. Moreover, a significant reduction in the number of tissue cysts and the load of T. gondii DNA was detected in brains of mice administered with pVAX1-SRS13 through ID + EP and IM routes compared to controls. In conclusion, the SRS13 DNA vaccine was found to be highly immunogenic and confers strong protection against chronic toxoplasmosis.


Asunto(s)
Anticuerpos Antiprotozoarios , Electroporación , Ratones Endogámicos BALB C , Proteínas Protozoarias , Vacunas Antiprotozoos , Toxoplasma , Vacunas de ADN , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Animales , Toxoplasma/inmunología , Toxoplasma/genética , Anticuerpos Antiprotozoarios/sangre , Anticuerpos Antiprotozoarios/inmunología , Electroporación/métodos , Vacunas Antiprotozoos/inmunología , Vacunas Antiprotozoos/administración & dosificación , Proteínas Protozoarias/inmunología , Proteínas Protozoarias/genética , Ratones , Femenino , Toxoplasmosis Animal/prevención & control , Toxoplasmosis Animal/inmunología , Inmunoglobulina G/sangre , Toxoplasmosis/prevención & control , Toxoplasmosis/inmunología , Antígenos de Protozoos/inmunología , Antígenos de Protozoos/genética , Interferón gamma/inmunología , Linfocitos T CD8-positivos/inmunología
20.
J Acquir Immune Defic Syndr ; 96(4): 350-360, 2024 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-38916429

RESUMEN

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.


Asunto(s)
Vacunas contra el SIDA , Adyuvantes Inmunológicos , Anticuerpos Anti-VIH , Proteína gp120 de Envoltorio del VIH , Infecciones por VIH , VIH-1 , Polisorbatos , Escualeno , Vacunas de ADN , Humanos , Vacunas contra el SIDA/inmunología , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/efectos adversos , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/efectos adversos , Femenino , Masculino , Adulto , Escualeno/administración & dosificación , Polisorbatos/administración & dosificación , Proteína gp120 de Envoltorio del VIH/inmunología , Adyuvantes Inmunológicos/administración & dosificación , VIH-1/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Anticuerpos Anti-VIH/sangre , Método Doble Ciego , Persona de Mediana Edad , Adulto Joven , Adyuvantes de Vacunas/administración & dosificación , Sudáfrica , Inmunogenicidad Vacunal , Adolescente , Estados Unidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...