Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters










Publication year range
1.
Science ; 378(6622): 899-904, 2022 11 25.
Article in English | MEDLINE | ID: mdl-36423275

ABSTRACT

Seasonal influenza vaccines offer little protection against pandemic influenza virus strains. It is difficult to create effective prepandemic vaccines because it is uncertain which influenza virus subtype will cause the next pandemic. In this work, we developed a nucleoside-modified messenger RNA (mRNA)-lipid nanoparticle vaccine encoding hemagglutinin antigens from all 20 known influenza A virus subtypes and influenza B virus lineages. This multivalent vaccine elicited high levels of cross-reactive and subtype-specific antibodies in mice and ferrets that reacted to all 20 encoded antigens. Vaccination protected mice and ferrets challenged with matched and mismatched viral strains, and this protection was at least partially dependent on antibodies. Our studies indicate that mRNA vaccines can provide protection against antigenically variable viruses by simultaneously inducing antibodies against multiple antigens.


Subject(s)
Influenza A virus , Influenza B virus , Orthomyxoviridae Infections , Vaccines, Combined , Vaccines, Synthetic , mRNA Vaccines , Animals , Mice , Ferrets , Nucleosides/chemistry , Nucleosides/genetics , Orthomyxoviridae Infections/prevention & control , Vaccines, Combined/genetics , Vaccines, Combined/immunology , mRNA Vaccines/genetics , mRNA Vaccines/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Influenza A virus/immunology , Influenza B virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Cross Reactions
2.
J Virol ; 96(7): e0216121, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35297660

ABSTRACT

Vaccine-induced protective T cell immunity is necessary for HIV-1 functional cure. We previously reported that rhesus PD1-Gag-based DNA vaccination sustained simian-human immunodeficiency virus (SHIV) suppression by inducing effector-memory CD8+ T cells. Here, we investigated a human PD1-Gag-based DNA vaccine, namely, ICVAX, for clinical translation. PD1-based dendritic cell targeting and mosaic antigenic designs were combined to generate the ICVAX by fusing the human soluble PD1 domain with a bivalent HIV-1 Gag-p41 mosaic antigen. The mosaic antigen was cross-reactive with patients infected with B, CRF07/08_BC, and CRF01_AE variants. In mice, ICVAX elicited stronger, broader, and more polyfunctional T cell responses than mosaic Gag-p41 alone, and suppressed EcoHIV infection more efficiently. In macaques, ICVAX elicited polyfunctional effector-memory T cell responses that targeted multiple nonoverlapping epitopes of the Gag-p41 antigen. Furthermore, ICVAX manufactured following good manufacturing practices proved potent immunogenicity in macaques after biannual homologous vaccination, warranting clinical evaluation of ICVAX as an immunotherapy against HIV-1. IMPORTANCE This study presents that ICVAX, a PD1-based DNA vaccine against HIV-1, could induce broad and polyfunctional T cell responses against different HIV-1 subtypes. ICVAX encodes a recombinant antigen consisting of the human soluble PD1 domain fused with two mosaic Gag-p41 antigens. The mosaic antigens cover more than 500 HIV-1 strains circulating in China including the subtypes B/B', CRF01_AE, and CRF07/08_BC. In mice, ICVAX elicited stronger, broader, and more polyfunctional T cell responses, with better EcoHIV suppression than the nontargeting mosaic Gag-p41 DNA vaccine. Moreover, both lab-generated and GMP-grade ICVAX also elicited strong polyfunctional effector-memory T cell responses in rhesus macaques with good immunogenicity against multiple nonoverlapping epitopes of the Gag-p41 antigen. This study therefore highlights the great potential to translate the PD1-based DNA vaccine approach into clinical use, and opens up new avenues for alternative HIV-1 vaccine design for HIV-1 preventive and functional cure.


Subject(s)
HIV Infections , HIV-1 , Vaccines, Combined , Vaccines, DNA , Viral Vaccines , AIDS Vaccines/immunology , Animals , Antigens, Viral , CD48 Antigen , CD8-Positive T-Lymphocytes , Epitopes/immunology , Gene Products, gag/genetics , Gene Products, gag/immunology , HIV Infections/prevention & control , HIV-1/genetics , Humans , Macaca mulatta , Memory T Cells , Mice , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology
3.
Front Immunol ; 12: 736360, 2021.
Article in English | MEDLINE | ID: mdl-34671354

ABSTRACT

Bacterial infections cause huge losses in aquaculture and a wide range of health issues in humans. A vaccine is the most economical, efficient, and environment-friendly agent for protecting hosts against bacterial infections. This study aimed to identify broad, cross-protective antigens from the extracellular secretory proteome of the marine bacterium Vibrio alginolyticus. Of the 69 predicted extracellular secretory proteins in its genome, 16 were randomly selected for gene cloning to construct DNA vaccines, which were used to immunize zebrafish (Danio rerio). The innate immune response genes were also investigated. Among the 16 DNA vaccines, 3 (AT730_21605, AT730_22220, and AT730_22910) were protective against V. alginolyticus infection with 47-66.7% increased survival compared to the control, while other vaccines had lower or no protective effects. Furthermore, AT730_22220, AT730_22910, and AT730_21605 also exhibited cross-immune protective effects against Pseudomonas fluorescens and/or Aeromonas hydrophila infection. Mechanisms for cross-protective ability was explored based on conserved epitopes, innate immune responses, and antibody neutralizing ability. These results indicate that AT730_21605, AT730_22220, and AT730_22910 are potential polyvalent vaccine candidates against bacterial infections. Additionally, our results suggest that the extracellular secretory proteome is an antigen pool that can be used for the identification of cross-protective immunogens.


Subject(s)
Antigens, Bacterial/pharmacology , Bacterial Infections/prevention & control , Bacterial Vaccines/pharmacology , Vaccine Development , Vaccines, Combined/pharmacology , Vibrio alginolyticus/immunology , Animals , Antibodies, Bacterial/metabolism , Antibodies, Neutralizing/metabolism , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Infections/immunology , Bacterial Infections/microbiology , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Cross Reactions , Disease Models, Animal , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/prevention & control , Immunity, Innate/drug effects , Immunization , Immunogenicity, Vaccine , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas Infections/prevention & control , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Vibrio Infections/immunology , Vibrio Infections/microbiology , Vibrio Infections/prevention & control , Vibrio alginolyticus/genetics , Zebrafish
4.
Front Immunol ; 12: 692937, 2021.
Article in English | MEDLINE | ID: mdl-34497604

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) kills thousands of people worldwide every day, thus necessitating rapid development of countermeasures. Immunoinformatics analyses carried out here in search of immunodominant regions in recently identified SARS-CoV-2 unannotated open reading frames (uORFs) have identified eight linear B-cell, one conformational B-cell, 10 CD4+ T-cell, and 12 CD8+ T-cell promising epitopes. Among them, ORF9b B-cell and T-cell epitopes are the most promising followed by M.ext and ORF3c epitopes. ORF9b40-48 (CD8+ T-cell epitope) is found to be highly immunogenic and antigenic with the highest allele coverage. Furthermore, it has overlap with four potent CD4+ T-cell epitopes. Structure-based B-cell epitope prediction has identified ORF9b61-68 to be immunodominant, which partially overlaps with one of the linear B-cell epitopes (ORF9b65-69). ORF3c CD4+ T-cell epitopes (ORF3c2-16, ORF3c3-17, and ORF3c4-18) and linear B-cell epitope (ORF3c14-22) have also been identified as the candidate epitopes. Similarly, M.ext and 7a.iORF1 (overlap with M and ORF7a) proteins have promising immunogenic regions. By considering the level of antigen expression, four ORF9b and five M.ext epitopes are finally shortlisted as potent epitopes. Mutation analysis has further revealed that the shortlisted potent uORF epitopes are resistant to recurrent mutations. Additionally, four N-protein (expressed by canonical ORF) epitopes are found to be potent. Thus, SARS-CoV-2 uORF B-cell and T-cell epitopes identified here along with canonical ORF epitopes may aid in the design of a promising epitope-based polyvalent vaccine (when connected through appropriate linkers) against SARS-CoV-2. Such a vaccine can act as a bulwark against SARS-CoV-2, especially in the scenario of emergence of variants with recurring mutations in the spike protein.


Subject(s)
Antigens, Viral/immunology , COVID-19 Vaccines/immunology , COVID-19/prevention & control , Coronavirus Nucleocapsid Proteins/immunology , SARS-CoV-2/immunology , Amino Acid Sequence/genetics , Antigens, Viral/genetics , COVID-19/immunology , COVID-19/virology , COVID-19 Vaccines/genetics , COVID-19 Vaccines/therapeutic use , Computational Biology , Coronavirus Nucleocapsid Proteins/genetics , Drug Design , Epitope Mapping , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Humans , Open Reading Frames/genetics , Open Reading Frames/immunology , SARS-CoV-2/genetics , Sequence Analysis, Protein , Vaccines, Combined/genetics , Vaccines, Combined/immunology
5.
Commun Biol ; 4(1): 597, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34011948

ABSTRACT

The COVID-19 pandemic continues to wreak havoc as worldwide SARS-CoV-2 infection, hospitalization, and death rates climb unabated. Effective vaccines remain the most promising approach to counter SARS-CoV-2. Yet, while promising results are emerging from COVID-19 vaccine trials, the need for multiple doses and the challenges associated with the widespread distribution and administration of vaccines remain concerns. Here, we engineered the coat protein of the MS2 bacteriophage and generated nanoparticles displaying multiple copies of the SARS-CoV-2 spike (S) protein. The use of these nanoparticles as vaccines generated high neutralizing antibody titers and protected Syrian hamsters from a challenge with SARS-CoV-2 after a single immunization with no infectious virus detected in the lungs. This nanoparticle-based vaccine platform thus provides protection after a single immunization and may be broadly applicable for protecting against SARS-CoV-2 and future pathogens with pandemic potential.


Subject(s)
COVID-19 Vaccines/administration & dosage , COVID-19/immunology , COVID-19/prevention & control , Pandemics , SARS-CoV-2 , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , COVID-19 Vaccines/genetics , COVID-19 Vaccines/immunology , Drug Delivery Systems , Female , Humans , Immunization/methods , Levivirus/genetics , Levivirus/immunology , Mesocricetus , Microscopy, Electron, Transmission , Models, Animal , Nanoparticles/administration & dosage , Nanoparticles/ultrastructure , Nanotechnology , Pandemics/prevention & control , Protein Engineering , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/administration & dosage , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Combined/administration & dosage , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/immunology
6.
Vet Microbiol ; 257: 109076, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33957572

ABSTRACT

The Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne hemorrhagic zoonotic disease, which is potentially fatal in human with mortality rates ranging from 16.2%-32%. The rabies virus (RABV) LBNSE vector expressing foreign antigens have shown considerable promise as vaccines against viral diseases, which is effective and safe. In the present study, we generated a recombinant RABV rLBNSE-Gn expressing a SFTSV glycoprotein Gn by reverse genetic technology to control rabies and SFTS in animals. An extra insertion of Gn gene did not impact replication of the recombinant virus rLBNSE-Gn in NA and BHK-21 cells compared to the parent rLBNSE strain. The SFTSV Gn gene together with RABV N and G genes were efficiently expressed in rLBNSE-infected Vero cells by immunostaining and immune blots. A single dose of 107 FFU of the rLBNSE-Gn intramuscularly inoculated in BALB/c mice induced rapid and robust humoral responses against both RABV and SFTSV without any signs of disease or weight loss. Compared to the rLBNSE and DMEM groups, the extra Gn expression contributed to the recruitments and/or activations of the dendritic cells and B cells from inguinal lymph nodes of BALB/c mice vaccinated with rLBNSE-Gn. The protective efficacy of rLBNSE-Gn against SFTSV in C57BL/6 mice was evaluated, and the virus loading in the spleens reduced to 10 TCID50/mg at 7 days post SFTSV infections, which indicated that the rLBNSE-Gn conferred efficacious protective immune responses from SFTSV in C57BL/6 mice. All the mice immunization with rLBNSE-Gn and rLBNSE survived after a lethal RABV challenge, suggesting a 100 % protection from RABV. Therefore, the rLBNSE-Gn would be a promising bivalent candidate vaccine against SFTS and rabies in animals.


Subject(s)
Antibodies, Viral/blood , Genetic Vectors , Phlebovirus/immunology , Rabies virus/genetics , Rabies/prevention & control , Severe Fever with Thrombocytopenia Syndrome/prevention & control , Viral Vaccines/immunology , Animals , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phlebovirus/genetics , Rabies Vaccines/administration & dosage , Rabies Vaccines/immunology , Rabies virus/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccines, Combined/administration & dosage , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
7.
Parasit Vectors ; 14(1): 241, 2021 May 07.
Article in English | MEDLINE | ID: mdl-33962671

ABSTRACT

BACKGROUND: Transmission-blocking vaccine (TBV) is a promising strategy for malaria elimination. It is hypothesized that mixing or fusing two antigens targeting different stages of sexual development may provide higher transmission-blocking activity than these antigens used individually. METHODS: A chimeric protein composed of fragments of Pbg37 and PSOP25 was designed and expressed the recombinant protein in Escherichia coli Rosetta-gami B (DE3). After immunizing mice with individual recombinant proteins Pbg37 and PSOP25, mixed proteins (Pbg37+PSOP25), or the fusion protein (Pbg37-PSOP25), the antibody titers of individual sera were analyzed by ELISA. IFA and Western blot were performed to test the reactivity of the antisera with the native proteins in the parasite. The transmission-blocking activity of the different immunization schemes was assessed using in vitro and in vivo assays. RESULTS: When Pbg37 and PSOP25 were co-administered in a mixture or as a fusion protein, they elicited similar antibody responses in mice as single antigens without causing immunological interference with each other. Antibodies against the mixed or fused antigens recognized the target proteins in the gametocyte, gamete, zygote, and ookinete stages. The mixed proteins or the fusion protein induced antibodies with significantly stronger transmission-reducing activities in vitro and in vivo than individual antigens. CONCLUSIONS: There was no immunological interference between Pbg37 and PSOP25. The bivalent vaccines, which expand the portion of the sexual development during which the transmission-blocking antibodies act, produced significantly stronger transmission-reducing activities than single antigens. Altogether, these data provide the theoretical basis for the development of combination TBVs targeting different sexual stages.


Subject(s)
Malaria Vaccines/administration & dosage , Malaria/prevention & control , Plasmodium berghei/growth & development , Plasmodium berghei/immunology , Protozoan Proteins/administration & dosage , Vaccines, Combined/administration & dosage , Animals , Antibodies, Protozoan/blood , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Humans , Immunization , Malaria/blood , Malaria/parasitology , Malaria/transmission , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Mice , Mice, Inbred BALB C , Plasmodium berghei/genetics , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Vaccines, Combined/genetics , Vaccines, Combined/immunology
8.
Infect Immun ; 89(7): e0010621, 2021 06 16.
Article in English | MEDLINE | ID: mdl-33875477

ABSTRACT

There are no vaccines licensed for enterotoxigenic Escherichia coli (ETEC), a leading cause of diarrhea for children in developing countries and international travelers. Virulence heterogeneity among strains and difficulties identifying safe antigens for protective antibodies against STa, a potent but poorly immunogenic heat-stable toxin which plays a key role in ETEC diarrhea, are challenges in ETEC vaccine development. To overcome these challenges, we applied a toxoid fusion strategy and a novel epitope- and structure-based multiepitope fusion antigen (MEFA) vaccinology platform to construct two chimeric multivalent proteins, toxoid fusion 3xSTaN12S-mnLTR192G/L211A and adhesin CFA/I/II/IV MEFA, and demonstrated that the proteins induced protective antibodies against STa and heat-labile toxin (LT) produced by all ETEC strains or the seven most important ETEC adhesins (CFA/I and CS1 to CS6) expressed by the ETEC strains causing 60 to 70% of diarrheal cases and moderate to severe cases. Combining two proteins, we prepared a protein-based multivalent ETEC vaccine, MecVax. MecVax was broadly immunogenic; mice and pigs intramuscularly immunized with MecVax developed no apparent adverse effects but had robust antibody responses to the target toxins and adhesins. Importantly, MecVax-induced antibodies were broadly protective, demonstrated by significant adherence inhibition against E. coli bacteria producing any of the seven adhesins and neutralization of STa and cholera toxin (CT) enterotoxicity. Moreover, MecVax protected against watery diarrhea and provided over 70% and 90% protection against any diarrhea from an STa-positive or an LT-positive ETEC strain in a pig challenge model. These results indicated that MecVax induces broadly protective antibodies and prevents diarrhea preclinically, signifying that MecVax is potentially an effective injectable vaccine for ETEC. IMPORTANCE Enterotoxigenic Escherichia coli (ETEC) bacteria are a top cause of children's diarrhea and travelers' diarrhea and are responsible for over 220 million diarrheal cases and more than 100,000 deaths annually. A safe and effective ETEC vaccine can significantly improve public health, particularly in developing countries. Data from this preclinical study showed that MecVax induces broadly protective antiadhesin and antitoxin antibodies, becoming the first ETEC vaccine candidate to induce protective antibodies inhibiting adherence of the seven most important ETEC adhesins and neutralizing the enterotoxicity of not only LT but also STa toxin. More importantly, MecVax is shown to protect against clinical diarrhea from STa-positive or LT-positive ETEC infection in a pig challenge model, recording protection from antibodies induced by the protein-based, injectable, subunit vaccine MecVax against ETEC diarrhea and perhaps the possibility of intramuscularly administered protein vaccines for protection against intestinal mucosal infection.


Subject(s)
Diarrhea/microbiology , Diarrhea/prevention & control , Enterotoxigenic Escherichia coli/immunology , Escherichia coli Infections/prevention & control , Escherichia coli Vaccines/immunology , Animals , Antibodies, Bacterial/immunology , Antibodies, Neutralizing/immunology , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Diarrhea/immunology , Disease Models, Animal , Epitopes/immunology , Escherichia coli Proteins/immunology , Escherichia coli Vaccines/administration & dosage , Escherichia coli Vaccines/adverse effects , Mice , Recombinant Fusion Proteins/immunology , Swine , Vaccines, Combined/genetics , Vaccines, Combined/immunology
9.
Int J Mol Sci ; 22(6)2021 Mar 23.
Article in English | MEDLINE | ID: mdl-33807097

ABSTRACT

Enteric fever is a major global healthcare issue caused largely by Salmonella enterica serovars Typhi and Paratyphi A. The objective of this study was to develop a novel, bivalent oral vaccine capable of protecting against both serovars. Our approach centred on genetically engineering the attenuated S. Typhi ZH9 strain, which has an excellent safety record in clinical trials, to introduce two S. Paratyphi A immunogenic elements: flagellin H:a and lipopolysaccharide (LPS) O:2. We first replaced the native S. Typhi fliC gene encoding flagellin with the highly homologous fliC gene from S. Paratyphi A using Xer-cise technology. Next, we replaced the S. Typhi rfbE gene encoding tyvelose epimerase with a spacer sequence to enable the sustained expression of O:2 LPS and prevent its conversion to O:9 through tyvelose epimerase activity. The resulting new strain, ZH9PA, incorporated these two genetic changes and exhibited comparable growth kinetics to the parental ZH9 strain. A formulation containing both ZH9 and ZH9PA strains together constitutes a new bivalent vaccine candidate that targets both S. Typhi and S. Paratyphi A antigens to address a major global healthcare gap for enteric fever prophylaxis. This vaccine is now being tested in a Phase I clinical trial (NCT04349553).


Subject(s)
Bioengineering , Salmonella Vaccines/immunology , Salmonella typhi/immunology , Typhoid Fever/prevention & control , Vaccines, Combined/immunology , Administration, Oral , Animals , Disease Models, Animal , Female , Flagellin/immunology , Genetic Vectors/genetics , Humans , Immunogenicity, Vaccine , Lipopolysaccharides/immunology , Mice , Salmonella Vaccines/administration & dosage , Salmonella Vaccines/genetics , Salmonella typhi/genetics , Vaccines, Combined/administration & dosage , Vaccines, Combined/genetics
10.
J Immunother Cancer ; 9(3)2021 03.
Article in English | MEDLINE | ID: mdl-33762322

ABSTRACT

BACKGROUND: Antitumor vaccines targeting tumor-associated antigens (TAAs) can generate antitumor immune response. A novel vaccine platform using adenovirus 5 (Ad5) vectors [E1-, E2b-] targeting three TAAs-prostate-specific antigen (PSA), brachyury, and MUC-1-has been developed. Both brachyury and the C-terminus of MUC-1 are overexpressed in metastatic castration-resistant prostate cancer (mCRPC) and have been shown to play an important role in resistance to chemotherapy, epithelial-mesenchymal transition, and metastasis. The transgenes for PSA, brachyury, and MUC-1 all contain epitope modifications for the expression of CD8+ T-cell enhancer agonist epitopes. We report here the first-in-human trial of this vaccine platform. METHODS: Patients with mCRPC were given concurrently three vaccines targeting PSA, brachyury, and MUC-1 at 5×1011 viral particles (VP) each, subcutaneously every 3 weeks for a maximum of three doses (dose de-escalation cohort), followed by a booster vaccine every 8 weeks for 1 year (dose-expansion cohort only). The primary objective was to determine the safety and the recommended phase II dose. Immune assays and clinical responses were evaluated. RESULTS: Eighteen patients with mCRPC were enrolled between July 2018 and September 2019 and received at least one vaccination. Median PSA was 25.58 ng/mL (range, 0.65-1006 ng/mL). The vaccine was tolerable and safe, and no grade >3 treatment-related adverse events or dose-limiting toxicities (DLTs) were observed. One patient had a partial response, while five patients had confirmed PSA decline and five had stable disease for >6 months. Median progression-free survival was 22 weeks (95% CI: 19.1 to 34). Seventeen (100%) of 17 patients mounted T-cell responses to at least one TAA, whereras 8 (47%) of 17 patients mounted immune responses to all three TAAs. Multifunctional T-cell responses to PSA, MUC-1, and brachyury were also detected after vaccination in the majority of the patients. CONCLUSIONS: Ad5 PSA/MUC-1/brachyury vaccine is well tolerated. The primary end points were met and there were no DLTs. The recommended phase II dose is 5×1011 VP. The vaccine demonstrated clinical activity, including one partial response and confirmed PSA responses in five patients. Three patients with prolonged PSA responses received palliative radiation therapy. Further research is needed to evaluate the clinical benefit and immunogenicity of this vaccine in combination with other immuno-oncology agents and/or palliative radiation therapy. TRIAL REGISTRATION NUMBER: NCT03481816.


Subject(s)
Adenoviridae/immunology , Cancer Vaccines/therapeutic use , Fetal Proteins/immunology , Kallikreins/immunology , Mucin-1/immunology , Prostate-Specific Antigen/immunology , Prostatic Neoplasms, Castration-Resistant/therapy , T-Box Domain Proteins/immunology , Vaccines, Combined/therapeutic use , Adenoviridae/genetics , Aged , Aged, 80 and over , Cancer Vaccines/adverse effects , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Fetal Proteins/genetics , Genetic Vectors , Humans , Kallikreins/genetics , Male , Middle Aged , Mucin-1/genetics , Progression-Free Survival , Prostate-Specific Antigen/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/immunology , T-Box Domain Proteins/genetics , Time Factors , Vaccination , Vaccine Efficacy , Vaccines, Combined/adverse effects , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Viral Vaccines
11.
Curr Opin Virol ; 43: 79-87, 2020 08.
Article in English | MEDLINE | ID: mdl-33164790

ABSTRACT

Dengue is the most important arboviral disease world-wide with an estimated 400 million annual infections. Dengvaxia™ is a live attenuated tetravalent vaccine recently licensed for dengue seropositive individuals aged 9-45 years. There is great need for a dengue vaccine that could be given to dengue-naïve individuals and very young children. To that end, the U.S. NIH developed a live attenuated tetravalent dengue vaccine using an iterative approach evaluating the safety, infectivity, and immunogenicity of different candidates. This approach identified poor candidates who were then discarded from further evaluation. Each of the components of the tetravalent vaccine formulation is able to replicate to very low titer, inducing a homotypic immune response to each. The immune response elicited by the tetravalent vaccine is balanced, without immunodominance of one component. The vaccine was licensed by several manufacturers for development, including the Instituto Butantan which initiated a Phase 3 efficacy trial.


Subject(s)
Dengue Vaccines/immunology , Dengue Virus/immunology , Dengue/prevention & control , Drug Development , Vaccines, Attenuated/immunology , Dengue/history , Dengue/immunology , Dengue/virology , Dengue Vaccines/administration & dosage , Dengue Vaccines/genetics , Dengue Vaccines/history , Dengue Virus/genetics , Drug Development/history , History, 20th Century , History, 21st Century , Humans , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/history , Vaccines, Combined/administration & dosage , Vaccines, Combined/genetics , Vaccines, Combined/history , Vaccines, Combined/immunology
12.
Front Immunol ; 11: 988, 2020.
Article in English | MEDLINE | ID: mdl-32595634

ABSTRACT

To counteract the deadly pathogens, i.e., Y. pestis, Y. enetrocolitica, and Y. pseudotuberculosis, we prepared a recombinant DNA construct lcrV-hsp70 encoding the bivalent fusion protein LcrV-HSP70. The lcrV gene of Y. pestis and hsp70 domain II DNA fragment of M. tuberculosis were amplified by PCR. The lcrV amplicon was first ligated in the pET vector using NcoI and BamHI restriction sites. Just downstream to the lcrV gene, the hsp70 domain II was ligated using BamHI and Hind III restriction sites. The in-frame and the orientation of cloned lcrV-hsp70 were checked by restriction analysis and nucleotide sequencing. The recombinant bivalent fusion protein LcrV-HSP70 was expressed in E. coli and purified by affinity chromatography. The vaccine potential of LcrV-HSP70 fusion protein was evaluated in formulation with alum. BALB/c mice were vaccinated, and the humoral and cellular immune responses were studied. The fusion protein LcrV-HSP70 induced a strong and significant humoral immune response in comparison to control animals. We also observed a significant difference in the expression levels of IFN-γ and TNF-α in LcrV-HSP70-immunized mice in comparison to control, HSP70, and LcrV groups. To test the protective efficacy of the LcrV-HSP70 fusion protein against plague and Yersiniosis, the vaccinated mice were challenged with Y. pestis, Y. enterocolitica, and Y. pseudotuberculosis separately. The bivalent fusion protein LcrV-HSP70 imparted 100% protection against the plague. In the case of Yersiniosis, on day 2 post challenge, there was a significant reduction in the number of CFU of Y. enterocolitica and Y. pseudotuberculosis in the blood (CFU/ml) and the spleen (CFU/g) of vaccinated animals in comparison to the LcrV, HSP70, and control group animals.


Subject(s)
Antigens, Bacterial/administration & dosage , Bacterial Proteins/administration & dosage , Bacterial Vaccines/administration & dosage , HSP70 Heat-Shock Proteins/administration & dosage , Immunogenicity, Vaccine , Pore Forming Cytotoxic Proteins/administration & dosage , Vaccination , Vaccines, Combined/administration & dosage , Yersinia Infections/prevention & control , Yersinia/immunology , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Load , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Biomarkers/blood , Cytokines/blood , Female , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/immunology , Immunity, Cellular , Immunity, Humoral , Mice, Inbred BALB C , Plague , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Yersinia/genetics , Yersinia/pathogenicity , Yersinia Infections/immunology , Yersinia Infections/microbiology
13.
J Immunother Cancer ; 8(1)2020 06.
Article in English | MEDLINE | ID: mdl-32561639

ABSTRACT

BACKGROUND: Stress-induced post-translational modifications occur during autophagy and can result in generation of new epitopes and immune recognition. One such modification is the conversion of arginine to citrulline by peptidylarginine deiminase enzymes. METHODS: We used Human leukocyte antigen (HLA) transgenic mouse models to assess the immunogenicity of citrullinated peptide vaccine by cytokine Enzyme linked immunosorbant spot (ELISpot) assay. Vaccine efficacy was assessed in tumor therapy studies using HLA-matched B16 melanoma and ID8 ovarian models expressing either constitutive or interferon-gamma (IFNγ) inducible Major Histocompatibility Complex (MHC) class II (MHC-II) as represented by most human tumors. To determine the importance of CD4 T cells in tumor therapy, we analyzed the immune cell infiltrate into murine tumors using flow cytometry and performed therapy studies in the presence of CD4 and CD8 T cell depletion. We assessed the T cell repertoire to citrullinated peptides in ovarian cancer patients and healthy donors using flow cytometry. RESULTS: The combination of citrullinated vimentin and enolase peptides (Modi-1) stimulated strong CD4 T cell responses in mice. Responses resulted in a potent anti-tumor therapy against established tumors and generated immunological memory which protected against tumor rechallenge. Depletion of CD4, but not CD8 T cells, abrogated the primary anti-tumor response as well as the memory response to tumor rechallenge. This was further reinforced by successful tumor regression being associated with an increase in tumor-infiltrating CD4 T cells and a reduction in tumor-associated myeloid suppressor cells. The anti-tumor response also relied on direct CD4 T cell recognition as only tumors expressing MHC-II were rejected. A comparison of different Toll-like receptor (TLR)-stimulating adjuvants showed that Modi-1 induced strong Th1 responses when combined with granulocyte-macrophage colony-stimulating factor (GMCSF), TLR9/TLR4, TLR9, TLR3, TLR1/2 and TLR7 agonists. Direct linkage of the TLR1/2 agonist to the peptides allowed the vaccine dose to be reduced by 10-fold to 100-fold without loss of anti-tumor activity. Furthermore, a CD4 Th1 response to the citrullinated peptides was seen in ovarian cancer patients. CONCLUSIONS: Modi-1 citrullinated peptide vaccine induces potent CD4-mediated anti-tumor responses in mouse models and a CD4 T cell repertoire is present in ovarian cancer patients to the citrullinated peptides suggesting that Modi-1 could be an effective vaccine for ovarian cancer patients.


Subject(s)
Cancer Vaccines/immunology , Melanoma, Experimental/therapy , Phosphopyruvate Hydratase/immunology , Vimentin/immunology , Adjuvants, Immunologic/administration & dosage , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cell Line, Tumor , Citrullination/immunology , Female , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Immunogenicity, Vaccine , Interferon-gamma/immunology , Lymphocyte Depletion , Male , Melanoma, Experimental/immunology , Mice , Mice, Transgenic , Phosphopyruvate Hydratase/genetics , Vaccines, Combined/administration & dosage , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vimentin/genetics
14.
Vaccine ; 38(14): 2949-2959, 2020 03 23.
Article in English | MEDLINE | ID: mdl-32111526

ABSTRACT

Junin (JUNV) and Machupo (MACV), two mammalian arenaviruses placed on the 2018 WHO watch list, are prevalent in South America causing Argentine and Bolivian hemorrhagic fevers (AHF and BHF), respectively. The live attenuated JUNV vaccine, Candid #1, significantly reduced the incidence of AHF. Vaccination induces neutralizing antibody (nAb) responses which effectively target GP1 (the viral attachment glycoprotein) pocket which accepts the tyrosine residue of the cellular receptor, human transferrin receptor 1 (TfR1). In spite of close genetic relationships between JUNV and MACV, variability in the GP1 receptor binding site (e.g., MACV GP1 loop 10) results in poor MACV neutralization by Candid #1-induced nAbs. Candid #1 is not recommended for vaccination of children younger than 15 years old (a growing "at risk" group), pregnant women, or other immunocompromised individuals. Candid #1's primary reliance on limited missense mutations for attenuation, genetic heterogeneity, and potential stability concerns complicate approval of this vaccine in the US. To address these issues, we applied alphavirus RNA replicon vector technology based on the human Venezuelan equine encephalitis vaccine (VEEV) TC-83 to generate replication restricted virus-like-particles vectors (VLPVs) simultaneously expressing cellular glycoprotein precursors (GPC) of both viruses, JUNV and MACV. Resulting JV&MV VLPVs were found safe and immunogenic in guinea pigs. Immunization with VLPVs induced humoral responses which correlated with complete protection against lethal disease after challenge with pathogenic strains of JUNV (Romero) and MACV (Carvallo).


Subject(s)
Alphavirus , Hemorrhagic Fever, American , Replicon , Viral Vaccines/immunology , Alphavirus/genetics , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Arenaviruses, New World , Guinea Pigs , Hemorrhagic Fever, American/prevention & control , Immunity, Humoral , Junin virus , RNA , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Viral Vaccines/genetics
15.
Vaccine ; 38(8): 2005-2015, 2020 02 18.
Article in English | MEDLINE | ID: mdl-31982262

ABSTRACT

Dengue virus (DENV) is a Flavivirus estimated to cause 390 million infections/year. Currently, there is no anti-viral specific treatment for dengue, and efficient DENV vector control is still unfeasible. Here, we designed and produced chimeric proteins containing potential immunogenic epitopes from the four DENV serotypes in an attempt to further compose safer, balanced tetravalent dengue vaccines. For this, South American DENV isolate sequences were downloaded from the NCBI/Virus Variation/Dengue virus databases and intraserotype-aligned to generate four consensuses. Four homologous DENV sequences were retrieved using BLAST and then interserotype-aligned. In parallel, sequences were subjected to linear B epitope prediction analysis. Regions of the envelope and NS1 proteins that are highly homologous among the four DENV serotypes, non-conserved antigenic regions and the most antigenic epitopes found in the C, prM, E and NS1 DENV proteins were used to construct 11 chimeric peptides. Genes encoding the chimeric proteins were commercially synthesized, and proteins were expressed, purified by affinity chromatography and further subjected to ELISA assays using sera from individuals infected with DENVs 1, 2, 3 or 4. As a proof-of-concept, the chimeric EnvEpII protein was selected to immunize BALB/c and C57BL/6 mice strains. The immunization with EnvEpII protein associated with aluminum induced an increased number of T CD4+ and CD8+ cells, high production of IgG1 and IgG2 antibodies, and increased levels of IL-2 and IL-17 cytokines, in both mouse strains. Because the EnvEpII protein associated with aluminum induced an efficient cellular response by stimulating the production of IL-2, IL-4, IL-17 and induced a robust humoral response in mice, we conclude that it resembles an efficient specific response against DENV infection. Although further experiments are required, our results indicate that epitope selection by bioinformatic tools is efficient to create recombinant proteins that can be used as candidates for the development of vaccines against infectious diseases.


Subject(s)
Dengue Vaccines , Dengue , Recombinant Fusion Proteins/immunology , Viral Proteins/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cytokines/immunology , Dengue/prevention & control , Dengue Vaccines/genetics , Dengue Virus/genetics , Dengue Virus/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Fusion Proteins/genetics , T-Lymphocytes/immunology , Vaccines, Combined/genetics , Viral Proteins/genetics
16.
Vaccine ; 38(7): 1690-1699, 2020 02 11.
Article in English | MEDLINE | ID: mdl-31937412

ABSTRACT

Newcastle disease virus (NDV) has been used as a vector in the development of vaccines and gene delivery. In the present study, we generated the thermostable recombinant NDV (rNDV) expressing the different forms of hemagglutinin (HA) of highly pathogenic avian influenza virus (HPAIV) H5N1 based on the full-length cDNA clone of thermostable TS09-C strain. The recombinant thermostable Newcastle disease viruses, rTS-HA, rTS-HA1 and rTS-tPAs/HA1, expressed the HA, HA1 or modified HA1 protein with the tissue plasminogen activator signal sequence (tPAs), respectively. The rNDVs displayed similar thermostability, growth kinetics and pathogenicity compared with the parental TS09-C virus. The tPAs facilitated the expression and secretion of HA1 protein in cells infected with rNDV. Animal studies demonstrated that immunization with rNDVs elicited effective H5N1- and NDV-specific antibody responses and conferred immune protection against lethal H5N1 and NDV challenges in chickens and mice. Importantly, vaccination of rTS-tPAs/HA1 resulted in enhanced protective immunity in chickens and mice. Our study thus provides a novel thermostable NDV-vectored vaccine candidate expressing a soluble form of a heterologous viral protein, which will greatly aid the poultry industry in developing countries.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines , Influenza in Birds , Newcastle Disease , Animals , Antibodies, Viral/blood , Chickens/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H5N1 Subtype , Influenza Vaccines/genetics , Influenza in Birds/prevention & control , Mice , Newcastle Disease/prevention & control , Newcastle disease virus , Tissue Plasminogen Activator/genetics , Vaccines, Combined/genetics
17.
Lancet Infect Dis ; 18(11): 1260-1268, 2018 11.
Article in English | MEDLINE | ID: mdl-30266329

ABSTRACT

BACKGROUND: The immunogenicity of acellular pertussis vaccines and persistence of immunity after vaccination might be improved by using genetically inactivated pertussis toxin (PTgen) instead of chemically inactivated pertussis toxin (PTchem) because of the preservation of conformational epitopes. We assessed the safety and immunogenicity of two vaccines containing PTgen 1 year after vaccination. METHODS: We did a phase 2/3 non-inferiority, randomised, controlled trial involving 450 adolescents (age 12-17 years) enrolled between July 6, 2015, and Aug 20, 2015. Participants were randomised 1:1:1 to receive one dose of vaccine containing PTgen and filamentous haemagglutinin (FHA) either in a monovalent formulation (aP[PTgen/FHA]) or in a combined formulation with tetanus and reduced-dose diphtheria toxoids (TdaP[PTgen/FHA]) or to receive a commercial vaccine containing reduced-dose PTchem (Tdap) as a comparator. We report a secondary trial outcome, namely antibody persistence 1 year after vaccination, assessed per protocol in 150 randomly preselected participants (50 per group). Seroconversion was defined as antibody titres at least four times greater than at baseline. Safety was assessed in all trial participants. This study is registered in the Thai Clinical Trial Registry, number TCTR20150703002. FINDINGS: Between June 5, 2016, and Aug 9, 2016, 442 (98%) of 450 enrolled participants attended a 1-year follow-up visit. After 1 year, persistent seroconversion for pertussis toxin neutralising antibodies was seen in 38 (76%, 95% CI 64-88) participants in the aP(PTgen/FHA) group and 41 (81%, 70-92) in the TdaP(PTgen/FHA) group, but in only four (8%, 1-16) in the Tdap comparator group. Seroconversion rates for IgG antibodies against pertussis toxin and FHA were also greater in the aP(PTgen/FHA) group (82%, 95% CI 71-93 and 64%, 51-77, respectively) and TdaP(PTgen/FHA) group (75%, 63-87 and 56%, 42-70, respectively) than in the Tdap group (4%, 0-9, p<0·0001, and 28%, 16-41, p=0·0007, respectively). 13 serious adverse events were reported in 12 participants and all were judged to be unrelated to the study vaccines. Five pregnancies were reported during follow-up, none of which had any maternal or neonatal complications. INTERPRETATION: A monovalent and a combined recombinant acellular pertussis vaccine containing PTgen induced antibody responses that were greater and sustained for longer than those achieved with the Tdap comparator vaccine. New recombinant pertussis vaccines containing PTgen might offer new opportunities to limit pertussis resurgence and can be widely used, including in pregnant women. FUNDING: BioNet-Asia.


Subject(s)
Antibodies, Bacterial/blood , Antitoxins/blood , Pertussis Toxin/immunology , Pertussis Vaccine/immunology , Adolescent , Asia , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Humans , Male , Pertussis Toxin/genetics , Pertussis Vaccine/administration & dosage , Pertussis Vaccine/adverse effects , Pertussis Vaccine/genetics , Seroconversion , Single-Blind Method , Time Factors , Vaccines, Acellular/administration & dosage , Vaccines, Acellular/adverse effects , Vaccines, Acellular/genetics , Vaccines, Acellular/immunology , Vaccines, Combined/administration & dosage , Vaccines, Combined/adverse effects , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
18.
Viruses ; 9(9)2017 09 15.
Article in English | MEDLINE | ID: mdl-28914805

ABSTRACT

Hepatitis A virus (HAV) and hepatitis E virus (HEV) are causative agents of acute viral hepatitis transmitted via the fecal-oral route. Both viruses place a heavy burden on the public health and economy of developing countries. To test the possibility that HAV could be used as an expression vector for the development of a combination vaccine against hepatitis A and E infections, recombinant HAV-HEp148 was created as a vector to express an HEV neutralization epitope (HEp148) located at aa 459-606 of the HEV capsid protein. The recombinant virus expressed the HEp148 protein in a partially dimerized state in HAV-susceptible cells. Immunization with the HAV-HEp148 virus induced a strong HAV- and HEV-specific immune response in mice. Thus, the present study demonstrates a novel approach to the development of a combined hepatitis A and E vaccine.


Subject(s)
Epitopes/immunology , Hepatitis A virus/genetics , Hepatitis A virus/immunology , Hepatitis Antibodies/biosynthesis , Hepatitis E virus/immunology , Viral Hepatitis Vaccines/immunology , Animals , Capsid Proteins/genetics , Capsid Proteins/immunology , Genetic Vectors , Hepatitis A/immunology , Hepatitis A/virology , Hepatitis Antibodies/immunology , Hepatitis E/immunology , Hepatitis E/virology , Hepatitis E virus/genetics , Mice , Neutralization Tests , Vaccination , Vaccines, Combined/administration & dosage , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Viral Hepatitis Vaccines/administration & dosage , Viral Hepatitis Vaccines/genetics
19.
Vaccine ; 35(20): 2728-2735, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28408118

ABSTRACT

Chicken pox and hand, foot and mouth disease (HFMD) are two major infectious diseases that mainly affect infants and children, causing significant morbidity annually. Varicella-zoster virus (VZV) and enterovirus 71 (EV71), respectively, are the principal epidemic pathogens causing these two diseases. To investigate the possibility of developing a novel combined vaccine to prevent chicken pox and HFMD, we constructed three chimeric virus-like particles (VLPs) (termed HBc-V/1/2, HBc-2/V/1 and HBc-1/2/V) based on the hepatitis B core antigen (HBc) carrier that display epitopes derived from VZV-gE, EV71-VP1, and EV71-VP2 in a varied tandem manner. The chimeric HBc can self-assemble into VLPs with these three epitopes displayed on the surface of particles. Epitope-specific antibody characterization suggested that HBc-V/1/2 elicits a balanced antibody response toward these three epitopes, and no immune interference was observed between the three epitopes. Importantly, the anti-HBc-V/1/2 sera could simultaneously neutralize VZV and EV71 and cross-neutralize coxsackievirus A16 (CVA16), another major pathogen causing HFMD. Moreover, the anti-HBc-V/1/2 sera protected neonatal mice from lethal challenge of EV71 and CVA16. Collectively, our study not only demonstrated that HBc-V/1/2 is a promising candidate combined vaccine for HFMD and Chicken pox but also provides a novel strategy for the design of combined vaccines.


Subject(s)
Chickenpox/prevention & control , Enterovirus A, Human/immunology , Epitopes/immunology , Hand, Foot and Mouth Disease/prevention & control , Herpesvirus 3, Human/immunology , Vaccines, Virus-Like Particle/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Drug Carriers , Enterovirus A, Human/genetics , Epitopes/genetics , Female , Hepatitis B Core Antigens/genetics , Herpesvirus 3, Human/genetics , Mice, Inbred BALB C , Vaccines, Combined/administration & dosage , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
20.
Retrovirology ; 14(1): 8, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28166802

ABSTRACT

BACKGROUND: In the Friend retrovirus mouse model we developed potent adenovirus-based vaccines that were designed to induce either strong Friend virus GagL85-93-specific CD8+ T cell or antibody responses, respectively. To optimize the immunization outcome we evaluated vaccination strategies using combinations of these vaccines. RESULTS: While the vaccines on their own confer strong protection from a subsequent Friend virus challenge, the simple combination of the vaccines for the establishment of an optimized immunization protocol did not result in a further improvement of vaccine effectivity. We demonstrate that the co-immunization with GagL85-93/leader-gag encoding vectors together with envelope-encoding vectors abrogates the induction of GagL85-93-specific CD8+ T cells, and in successive immunization protocols the immunization with the GagL85-93/leader-gag encoding vector had to precede the immunization with an envelope encoding vector for the efficient induction of GagL85-93-specific CD8+ T cells. Importantly, the antibody response to envelope was in fact enhanced when the mice were adenovirus-experienced from a prior immunization, highlighting the expedience of this approach. CONCLUSIONS: To circumvent the immunosuppressive effect of envelope on immune responses to simultaneously or subsequently administered immunogens, we developed a two immunizations-based vaccination protocol that induces strong immune responses and confers robust protection of highly Friend virus-susceptible mice from a lethal Friend virus challenge.


Subject(s)
Adenoviridae/genetics , Retroviridae Infections/immunology , Retroviridae Infections/prevention & control , Vaccination/methods , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Adenoviridae/immunology , Animals , Antibodies, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Friend murine leukemia virus/genetics , Friend murine leukemia virus/immunology , Genetic Vectors , HEK293 Cells , Humans , Mice , Retroviridae Infections/virology , Vaccines, Combined/administration & dosage , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Vaccines, Synthetic/immunology , Viral Vaccines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...