Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 13(1): 18902, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37919366

ABSTRACT

Throughout the COVID-19 pandemic, several variants of concern (VoC) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have evolved, affecting the efficacy of the approved COVID-19 vaccines. To address the need for vaccines that induce strong and persistent cross-reactive neutralizing antibodies and T cell responses, we developed a prophylactic SARS-CoV-2 vaccine candidate based on our easily and rapidly adaptable plasmid DNA vaccine platform. The vaccine candidate, referred to here as VB2129, encodes a protein homodimer consisting of the receptor binding domain (RBD) from lineage B.1.351 (Beta) of SARS-CoV-2, a VoC with a severe immune profile, linked to a targeting unit (human LD78ß/CCL3L1) that binds chemokine receptors on antigen-presenting cells (APCs) and a dimerization unit (derived from the hinge and CH3 exons of human IgG3). Immunogenicity studies in mice demonstrated that the APC-targeted vaccine induced strong antibody responses to both homologous Beta RBD and heterologous RBDs derived from Wuhan, Alpha, Gamma, Delta, and Omicron BA.1 variants, as well as cross-neutralizing antibodies against these VoC. Overall, preclinical data justify the exploration of VB2129 as a potential booster vaccine that induces broader antibody- and T cell-based protection against current and future SARS-CoV-2 VoC.


Subject(s)
COVID-19 , Cancer Vaccines , Vaccines, DNA , Animals , Humans , Mice , COVID-19 Vaccines , SARS-CoV-2 , Pandemics , COVID-19/prevention & control , T-Lymphocytes , Antigen-Presenting Cells , Broadly Neutralizing Antibodies , DNA , Immunoglobulin G , Antibodies, Neutralizing , Antibodies, Viral
2.
J Immunol ; 197(9): 3575-3585, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27671110

ABSTRACT

It has been difficult to translate promising results from DNA vaccination in mice to larger animals and humans. Previously, DNA vaccines encoding proteins that target Ag to MHC class II (MHC-II) molecules on APCs have been shown to induce rapid, enhanced, and long-lasting Ag-specific Ab titers in mice. In this study, we describe two novel DNA vaccines that as proteins target HLA class II (HLA-II) molecules. These vaccine proteins cross-react with MHC-II molecules in several species of larger mammals. When tested in ferrets and pigs, a single DNA delivery with low doses of the HLA-II-targeted vaccines resulted in rapid and increased Ab responses. Importantly, painless intradermal jet delivery of DNA was as effective as delivery by needle injection followed by electroporation. As an indication that the vaccines could also be useful for human application, HLA-II-targeted vaccine proteins were found to increase human CD4+ T cell responses by a factor of ×103 in vitro. Thus, targeting of Ag to MHC-II molecules may represent an attractive strategy for increasing efficacy of DNA vaccines in larger animals and humans.


Subject(s)
Antibody Formation , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Vaccines, DNA/immunology , Animals , Antigens/metabolism , Cell Proliferation , Cells, Cultured , Cross Reactions , Ferrets , HLA Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Lymphocyte Activation , Mice , Swine , Vaccination
3.
Front Immunol ; 7: 321, 2016.
Article in English | MEDLINE | ID: mdl-27602032

ABSTRACT

Current influenza vaccines are effective but imperfect, failing to cover against emerging strains of virus and requiring seasonal administration to protect against new strains. A key step to improving influenza vaccines is to improve our understanding of vaccine-induced protection. While it is clear that antibodies play a protective role, vaccine-induced CD8(+) T cells can improve protection. To further explore the role of CD8(+) T cells, we used a DNA vaccine that encodes antigen dimerized to an immune cell targeting module. Immunizing CB6F1 mice with the DNA vaccine in a heterologous prime-boost regime with the seasonal protein vaccine improved the resolution of influenza disease compared with protein alone. This improved disease resolution was dependent on CD8(+) T cells. However, DNA vaccine regimes that induced CD8(+) T cells alone were not protective and did not boost the protection provided by protein. The MHC-targeting module used was an anti-I-E(d) single chain antibody specific to the BALB/c strain of mice. To test the role of MHC targeting, we compared the response between BALB/c, C57BL/6 mice, and an F1 cross of the two strains (CB6F1). BALB/c mice were protected, C57BL/6 were not, and the F1 had an intermediate phenotype; showing that the targeting of antigen is important in the response. Based on these findings, and in agreement with other studies using different vaccines, we conclude that, in addition to antibody, inducing a protective CD8 response is important in future influenza vaccines.

4.
J Immunol ; 191(6): 3221-31, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23956431

ABSTRACT

New influenza A viruses with pandemic potential periodically emerge due to viral genomic reassortment. In the face of pandemic threats, production of conventional egg-based vaccines is time consuming and of limited capacity. We have developed in this study a novel DNA vaccine in which viral hemagglutinin (HA) is bivalently targeted to MHC class II (MHC II) molecules on APCs. Following DNA vaccination, transfected cells secreted vaccine proteins that bound MHC II on APCs and initiated adaptive immune responses. A single DNA immunization induced within 8 d protective levels of strain-specific Abs and also cross-reactive T cells. During the Mexican flu pandemic, a targeted DNA vaccine (HA from A/California/07/2009) was generated within 3 wk after the HA sequences were published online. These results suggest that MHC II-targeted DNA vaccines could play a role in situations of pandemic threats. The vaccine principle should be extendable to other infectious diseases.


Subject(s)
Hemagglutinins/immunology , Histocompatibility Antigens Class II/immunology , Influenza Vaccines/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Viral/immunology , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Pandemics , Reverse Transcriptase Polymerase Chain Reaction , Spain/epidemiology , Transfection
5.
Eur J Haematol ; 86(5): 385-95, 2011 May.
Article in English | MEDLINE | ID: mdl-21332794

ABSTRACT

OBJECTIVES: A homodimeric fusion DNA vaccine targeting idiotype (Id) to antigen-presenting cells (APC) induced robust tumor protection in a mouse model of multiple myeloma (MM). Similar Id vaccine molecules were generated for four patients with MM with three main objectives: (i) do the vaccine molecules induce bona fide anti-Id immune responses in mice? (ii) does targeting of the vaccine molecules to APC enhance immune responses? (iii) can anti-Id antibodies, generated as by-product in vaccinated mice, be used to establish sensitive assays for complete remission (CR) prior to patient vaccination? METHODS: Chimeric vaccine molecules targeting patient Id to mouse major histocompatibility complex (MHC) class II molecules were genetically constructed for four patients with MM. RESULTS: DNA vaccination of mice with chimeric vaccines targeting patient Id to mouse MHC class II molecules elicited antibodies specific for the patient's myeloma protein. Targeting MHC class II greatly enhanced anti-Id responses. Mouse anti-Id antibodies were used to establish myeloma protein-specific enzyme-linked immunosorbent assays (ELISAs) that were between 75 and 1500 times more sensitive than conventional serum protein electrophoresis and immunofixation. CONCLUSIONS: These results pave the way for testing targeted DNA Id vaccines in patients in CR. Id- and patient-specific ELISA could be established affording evaluation of CR depth beyond current serological methods.


Subject(s)
Cancer Vaccines/pharmacology , Multiple Myeloma/therapy , Animals , Antibodies, Anti-Idiotypic/genetics , Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibody Specificity , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Gene Rearrangement, B-Lymphocyte, Light Chain , Histocompatibility Antigens Class II/immunology , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Myeloma Proteins/analysis , Myeloma Proteins/immunology , Plasmacytoma/genetics , Plasmacytoma/immunology , Plasmacytoma/therapy , Remission Induction , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, DNA/pharmacology
6.
Blood ; 110(6): 1797-805, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17540847

ABSTRACT

V regions of monoclonal Ig express an exquisite B-cell tumor-specific antigen called idiotype (Id). Id is a weak antigen and it is important to improve immunogenicity of Id vaccines. Chemokine receptors are expressed on antigen-presenting cells (APCs) and are promising targets for Id vaccines. Here we compare monomeric and dimeric forms of MIP-1alpha and RANTES that target Id to APCs in a mouse B lymphoma (A20) and a multiple myeloma model (MOPC315). MIP-1alpha was more potent than RANTES. The dimeric proteins were more potent than monomeric equivalents in short-term assays. When delivered in vivo by intramuscular injection of plasmids followed by electroporation, dimeric proteins efficiently primed APCs in draining lymph nodes for activation and proliferation of Id-specific CD4(+) T cells. Good anti-Id antibody responses were obtained, and mice immunized only once were 60% to 80% protected in both tumor models. CD8(+) T cells contributed to the protection. Antibody responses and tumor protection were reduced when the human Ig hinge = C(H)3 dimerization motif was replaced with syngeneic mouse counterparts, indicating that tumor-protective responses were dependent on xenogeneic sequences. The results suggest that bivalency and foreign sequences combine to increase the efficiency of chemokine-Id DNA vaccines.


Subject(s)
Antigen-Presenting Cells/immunology , Chemokine CCL5/immunology , Chemokines/immunology , Immunoglobulin Idiotypes/immunology , Macrophage Inflammatory Proteins/immunology , Vaccines, DNA/immunology , Animals , Chemokine CCL3 , Chemokine CCL4 , Chemokine CCL5/genetics , Dimerization , Electroporation , Flow Cytometry , Histocompatibility Antigens Class II/immunology , Humans , Immunoglobulin Idiotypes/genetics , Injections, Intramuscular , Lymphoma, B-Cell/immunology , Macrophage Inflammatory Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, SCID , Mice, Transgenic , Models, Immunological , Multiple Myeloma/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/prevention & control , Plasmids , Recombinant Fusion Proteins/immunology , Time Factors , Vaccination , Vaccines, DNA/chemistry
7.
Eur J Immunol ; 35(11): 3142-52, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16184515

ABSTRACT

In order to prevent or ameliorate autoimmune disease, it would be desirable to induce central tolerance to peripheral self-antigens. We have investigated whether recombinant antibodies (Ab) that deliver T cell epitopes to antigen-presenting cells (APC) in the thymus can be used to induce thymocyte deletion. Troybodies are recombinant Ab with V regions specific for APC surface molecules that have T cell epitopes genetically introduced in their C domains. When MHC class II-specific Troybodies with the lambda2(315)T cell epitope were injected into lambda2(315)-specific TCR transgenic mice, a profound deletion of (CD4+)8+ thymocytes was observed. MHC class II-specific Troybodies were 10-100-fold more efficient than non-targeting peptide Ab, and 500-fold more efficient than synthetic peptide at inducing deletion. Similar findings were observed when MHC class II-specific Troybodies with the OVA(323-339) T cell epitope were injected into OVA-specific TCR transgenic mice. Although deletion was transient after a single injection, newborn mice repeatedly injected with MHC class II-specific Troybodies for 4 weeks, had reduced antigen-specific T cells in peripheral lymphoid tissues and reduced T cell responses. These experiments suggest that Troybodies constructed to target specifically thymic APC could be useful tools for induction and maintenance of central T cell tolerance in autoimmune diseases.


Subject(s)
Antibodies , Clonal Deletion/immunology , Epitopes, T-Lymphocyte/biosynthesis , Immune Tolerance , Peptides/metabolism , Thymus Gland/metabolism , Animals , Antibodies/genetics , Antibodies/metabolism , Antigen Presentation/genetics , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Histocompatibility Antigens Class II/immunology , Immune Tolerance/genetics , Lymphocyte Count , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Ovalbumin/metabolism , Peptide Fragments/metabolism , Peptides/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Fc/immunology , Recombinant Proteins , T-Lymphocytes/immunology , Thymus Gland/immunology
8.
Eur J Immunol ; 35(6): 1786-94, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15864781

ABSTRACT

Due to somatic recombination and hypermutation, Ig variable heavy (V(H)) and light (V(L)) regions contain unique immunogenic determinants, idiotopes (Id), which can stimulate T cells. To address the relevance of this in a human disease, monoclonal IgG (mAb)-secreting B cell clones were established from the cerebrospinal fluid (CSF) of two patients with multiple sclerosis (MS). HLA-DR-restricted CD4(+) T cell lines and clones from CSF of both patients specifically recognized autologous CSF mAb. The CSF T cell clones produced IFN-gamma; some also produced TNF-alpha, IL-10 and IL-5. V(H) and V(L) on the monoclonal IgG derived from CSF B cells expressed amino acid replacements due to somatic mutations. A T cell epitope was mapped to a V(H) framework region, where an amino acid replacement was critical for the T cell recognition. The finding of Id-specific T cells and Id-bearing B cells in the CSF indicates that they coexist within the diseased organ, and provide a basis for the study of Id-driven T-B cell collaboration in a human autoimmune disease.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin G/cerebrospinal fluid , Immunoglobulin Idiotypes/immunology , Multiple Sclerosis/immunology , T-Lymphocytes/immunology , Adult , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Cytokines/biosynthesis , Epitopes, T-Lymphocyte , Female , Humans , Lymphocyte Activation , Molecular Sequence Data , Multiple Sclerosis/cerebrospinal fluid
SELECTION OF CITATIONS
SEARCH DETAIL
...