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.
J Hepatol ; 78(4): 717-730, 2023 04.
Article in English | MEDLINE | ID: mdl-36634821

ABSTRACT

BACKGROUND & AIMS: We recently developed a heterologous therapeutic vaccination scheme (TherVacB) comprising a particulate protein prime followed by a modified vaccinia-virus Ankara (MVA)-vector boost for the treatment of HBV. However, the key determinants required to overcome HBV-specific immune tolerance remain unclear. Herein, we aimed to study new combination adjuvants and unravel factors that are essential for the antiviral efficacy of TherVacB. METHODS: Recombinant hepatitis B surface and core antigen (HBsAg and HBcAg) particles were formulated with different liposome- or oil-in-water emulsion-based combination adjuvants containing saponin QS21 and monophosphoryl lipid A; these formulations were compared to STING-agonist c-di-AMP and conventional aluminium hydroxide formulations. Immunogenicity and the antiviral effects of protein antigen formulations and the MVA-vector boost within TherVacB were evaluated in adeno-associated virus-HBV-infected and HBV-transgenic mice. RESULTS: Combination adjuvant formulations preserved HBsAg and HBcAg integrity for ≥12 weeks, promoted human and mouse dendritic cell activation and, within TherVacB, elicited robust HBV-specific antibody and T-cell responses in wild-type and HBV-carrier mice. Combination adjuvants that prime a balanced HBV-specific type 1 and 2 T helper response induced high-titer anti-HBs antibodies, cytotoxic T-cell responses and long-term control of HBV. In the absence of an MVA-vector boost or following selective CD8 T-cell depletion, HBsAg still declined (mediated mainly by anti-HBs antibodies) but HBV replication was not controlled. Selective CD4 T-cell depletion during the priming phase of TherVacB resulted in a complete loss of vaccine-induced immune responses and its therapeutic antiviral effect in mice. CONCLUSIONS: Our results identify CD4 T-cell activation during the priming phase of TherVacB as a key determinant of HBV-specific antibody and CD8 T-cell responses. IMPACT AND IMPLICATIONS: Therapeutic vaccination is a potentially curative treatment option for chronic hepatitis B. However, it remains unclear which factors are essential for breaking immune tolerance in HBV carriers and determining successful outcomes. Our study provides the first direct evidence that efficient priming of HBV-specific CD4 T cells determines the success of therapeutic hepatitis B vaccination in two preclinical HBV-carrier mouse models. Applying an optimal formulation of HBV antigens that activates CD4 and CD8 T cells during prime immunization provided the foundation for an antiviral effect of therapeutic vaccination, while depletion of CD4 T cells led to a complete loss of vaccine-induced antiviral efficacy. Boosting CD8 T cells was important to finally control HBV in these mouse models. Our findings provide important insights into the rational design of therapeutic vaccines for the cure of chronic hepatitis B.


Subject(s)
Hepatitis B Vaccines , Hepatitis B, Chronic , Mice , Humans , Animals , Hepatitis B virus , Hepatitis B Surface Antigens , Hepatitis B Core Antigens , CD4-Positive T-Lymphocytes , Immunization , Vaccination/methods , Hepatitis B Antibodies , CD8-Positive T-Lymphocytes , Mice, Transgenic , Adjuvants, Immunologic , Antiviral Agents
2.
Vaccine ; 34(13): 1566-1574, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26874325

ABSTRACT

BACKGROUND: Plasmodium falciparum MSP2 is a blood stage protein that is associated with protection against malaria. It was shown that the MSP2 dimorphic (D) and constant (C) regions were well recognized by immune human antibodies, and were characterized by major conserved epitopes in different endemic areas and age groups. These Abs recognized merozoite-derived proteins in WB and IFA. Here, the goal was to determine in mice the immunogenicity of the two allelic MSP2 D and C domains formulated with different adjuvants, for their possible use in future clinical studies. METHOD: Female A/J, C3H, and ICR mice were immunized subcutaneously 3 times at 3-week interval with a mixture of allelic and conserved MSP2 long synthetic peptides formulated with different adjuvants. One week after the third injection, sera from each group were obtained and stored at -20°C for subsequent testing. RESULTS: Both domains of the two MSP2 families are immunogenic and the fine specificity and intensity of the Ab responses are dependent on mouse strains and adjuvants. The major epitopes were restricted to the 20-mer peptide sequences comprising the last 8aa of D and first 12aa of C of the two allelic families and the first 20aa of the C region, this for most strains and adjuvants. Strong immune responses were associated with GLA-SE adjuvant and its combination with other TLR agonists (CpG or GDQ) compared to alhydrogel and Montanide. Further, the elicited Abs were also capable of recognizing Plasmodium-derived MSP2 and inhibiting parasite growth in ADCI. CONCLUSION: The data provide a valuable opportunity to evaluate in mice different adjuvant and antigen formulations of a candidate vaccine containing both MSP2 D and C fragments. The formulations with GLA-SE seem to be a promising option to be compared with the alhydrogel one in human clinical trials.


Subject(s)
Adjuvants, Immunologic/chemistry , Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Protozoan Proteins/immunology , Amino Acid Sequence , Animals , Cells, Cultured , Epitope Mapping , Epitopes/immunology , Female , Glucosides/chemistry , Humans , Immunoglobulin G/blood , Lipid A/chemistry , Mice, Inbred C3H , Mice, Inbred ICR , Molecular Sequence Data , Monocytes/parasitology , Plasmodium falciparum/immunology , Toll-Like Receptors/agonists , Vaccines, Synthetic/immunology
3.
Malar J ; 13: 510, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25526742

ABSTRACT

BACKGROUND: Two long synthetic peptides representing the dimorphic and constant C-terminal domains of the two allelic families of Plasmodium falciparum merozoite surface proteins 2 are considered promising malaria vaccine candidates. The aim of the current study is to characterize the immune response (epitope mapping) in naturally exposed individuals and relate immune responses to the risk of clinical malaria. METHODS: To optimize their construction, the fine specificity of human serum antibodies from donors of different age, sex and living in four distinct endemic regions was determined in ELISA by using overlapping 20 mer peptides covering the two domains. Immune purified antibodies were used in Western blot and immunofluorescence assay to recognize native parasite derivate proteins. RESULTS: Immunodominant epitopes were characterized, and their distribution was similar irrespective of geographic origin, age group and gender. Acquisition of a 3D7 family and constant region-specific immune response and antibody avidity maturation occur early in life while a longer period is needed for the corresponding FC27 family response. In addition, the antibody response to individual epitopes within the 3D7 family-specific region contributes to protection from malaria infection with different statistical weight. It is also illustrated that affinity-purified antibodies against the dimorphic or constant regions recognized homologous and heterologous parasites in immunofluorescence and homologous and heterologous MSP2 and other polypeptides in Western blot. CONCLUSION: Data from this current study may contribute to a development of MSP2 vaccine candidates based on conserved and dimorphic regions thus bypassing the complexity of vaccine development related to the polymorphism of full-length MSP2.


Subject(s)
Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Epitope Mapping , Epitopes/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Adolescent , Adult , Blotting, Western , Child , Child, Preschool , Conserved Sequence/immunology , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Humans , Infant , Male , Middle Aged , Young Adult
4.
PLoS One ; 7(10): e46112, 2012.
Article in English | MEDLINE | ID: mdl-23056243

ABSTRACT

In a genome-wide screen for alpha-helical coiled coil motifs aiming at structurally defined vaccine candidates we identified PFF0165c. This protein is exported in the trophozoite stage and was named accordingly Trophozoite exported protein 1 (Tex1). In an extensive preclinical evaluation of its coiled coil peptides Tex1 was identified as promising novel malaria vaccine candidate providing the rational for a comprehensive cell biological characterization of Tex1. Antibodies generated against an intrinsically unstructured N-terminal region of Tex1 and against a coiled coil domain were used to investigate cytological localization, solubility and expression profile. Co-localization experiments revealed that Tex1 is exported across the parasitophorous vacuole membrane and located to Maurer's clefts. Change in location is accompanied by a change in solubility: from a soluble state within the parasite to a membrane-associated state after export to Maurer's clefts. No classical export motifs such as PEXEL, signal sequence/anchor or transmembrane domain was identified for Tex1.


Subject(s)
Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Erythrocytes/parasitology , Malaria/parasitology , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , Antigens, Protozoan/genetics , Base Sequence , Blotting, Western , Brefeldin A/pharmacology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cytosol/drug effects , Cytosol/metabolism , Erythrocytes/drug effects , Erythrocytes/metabolism , Gene Expression , Malaria/metabolism , Malaria Vaccines/immunology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Microscopy, Confocal , Molecular Sequence Data , Organelles/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Protein Transport/drug effects , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Vacuoles/metabolism
5.
Vaccine ; 29(40): 7090-9, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21803099

ABSTRACT

A new strategy for the rapid identification of new malaria antigens based on protein structural motifs was previously described. We identified and evaluated the malaria vaccine potential of fragments of several malaria antigens containing α-helical coiled coil protein motifs. By taking advantage of the relatively short size of these structural fragments, we constructed different poly-epitopes in which 3 or 4 of these segments were joined together via a non-immunogenic linker. Only peptides that are targets of human antibodies with anti-parasite in vitro biological activities were incorporated. One of the constructs, P181, was well recognized by sera and peripheral blood mononuclear cells (PBMC) of adults living in malaria-endemic areas. Affinity purified antigen-specific human antibodies and sera from P181-immunized mice recognised native proteins on malaria-infected erythrocytes in both immunofluorescence and western blot assays. In addition, specific antibodies inhibited parasite development in an antibody dependent cellular inhibition (ADCI) assay. Naturally induced antigen-specific human antibodies were at high titers and associated with clinical protection from malaria in longitudinal follow-up studies in Senegal.


Subject(s)
Antigens, Protozoan/chemistry , Antigens, Protozoan/immunology , Epitopes/chemistry , Epitopes/immunology , Malaria Vaccines/chemistry , Malaria Vaccines/immunology , Animals , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Follow-Up Studies , Humans , Immune Sera/immunology , Longitudinal Studies , Malaria/immunology , Malaria/prevention & control , Mice , Mice, Inbred C3H , Mice, Inbred ICR , Peptides/chemistry , Peptides/immunology , Plasmodium falciparum/immunology , Protein Structure, Secondary , Senegal , Structure-Activity Relationship , T-Lymphocytes/immunology
6.
Vaccine ; 27(20): 2653-61, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19428875

ABSTRACT

Merozoite surface protein 2 (MSP2) is a promising vaccine candidate against Plasmodium falciparum blood stages. A recombinant 3D7 form of MSP2 was a subunit of Combination B, a blood stage vaccine tested in the field in Papua New Guinea. A selective effect in favour of the allelic family not represented by the vaccine argued for a MSP2 vaccine consisting of both dimorphic variants. An alternative approach to recombinant manufacture of vaccines is the production of long synthetic peptides (LSP). LSP exceeding a length of well over 100 amino acids can now be routinely synthesized. Synthetic production of vaccine antigens cuts the often time-consuming steps of protein expression and purification short. This considerably reduces the time for a candidate to reach the phase of clinical trials. Here we present the evaluation of two long synthetic peptides representing both allelic families of MSP2 as potential vaccine candidates. The constructs were well recognized by human immune sera from different locations and different age groups. Furthermore, peptide-specific antibodies in human immune sera were associated with protection from clinical malaria. The synthetic fragments share major antigenic properties with native MSP2. Immunization of mice with these antigens yielded high titre antibody responses and monoclonal antibodies recognized parasite-derived MSP2. Our results justify taking these candidate poly-peptides into further vaccine development.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/chemical synthesis , Malaria Vaccines/immunology , Protozoan Proteins/chemical synthesis , Protozoan Proteins/immunology , Adult , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Protozoan/blood , Child, Preschool , Female , Humans , Infant , Malaria, Falciparum/immunology , Male , Mice , Molecular Sequence Data , Plasmodium falciparum/immunology , Sequence Alignment , Vaccines, Synthetic/immunology , Young Adult
7.
Vaccine ; 27(8): 1266-71, 2009 Feb 18.
Article in English | MEDLINE | ID: mdl-19111883

ABSTRACT

Needle-free procedures are very attractive ways to deliver vaccines because they diminish the risk of contamination and may reduce local reactions, pain or pain fear especially in young children with a consequence of increasing the vaccination coverage for the whole population. For this purpose, the possible development of a mucosal malaria vaccine was investigated. Intranasal immunization was performed in BALB/c mice using a well-studied Plasmodium berghei model antigen derived from the circumsporozoite protein with the modified heat-labile toxin of Escherichia coli (LTK63), which is devoid of any enzymatic activity compared to the wild type form. Here, we show that intranasal administration of the two compounds activates the T and B cell immune response locally and systemically. In addition, a total protection of mice is obtained upon a challenge with live sporozoites.


Subject(s)
Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Bacterial Toxins/pharmacology , Enterotoxins/pharmacology , Escherichia coli Proteins/pharmacology , Malaria Vaccines/immunology , Malaria/prevention & control , Plasmodium berghei/immunology , Protozoan Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Animals , B-Lymphocytes/immunology , Bacterial Toxins/administration & dosage , Enterotoxins/administration & dosage , Escherichia coli/genetics , Escherichia coli Proteins/administration & dosage , Female , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Mice , Mice, Inbred BALB C , Plasmodium berghei/genetics , Protozoan Proteins/administration & dosage , Protozoan Proteins/genetics , T-Lymphocytes/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
8.
PLoS One ; 2(7): e645, 2007 Jul 25.
Article in English | MEDLINE | ID: mdl-17653272

ABSTRACT

To identify malaria antigens for vaccine development, we selected alpha-helical coiled coil domains of proteins predicted to be present in the parasite erythrocytic stage. The corresponding synthetic peptides are expected to mimic structurally "native" epitopes. Indeed the 95 chemically synthesized peptides were all specifically recognized by human immune sera, though at various prevalence. Peptide specific antibodies were obtained both by affinity-purification from malaria immune sera and by immunization of mice. These antibodies did not show significant cross reactions, i.e., they were specific for the original peptide, reacted with native parasite proteins in infected erythrocytes and several were active in inhibiting in vitro parasite growth. Circular dichroism studies indicated that the selected peptides assumed partial or high alpha-helical content. Thus, we demonstrate that the bioinformatics/chemical synthesis approach described here can lead to the rapid identification of molecules which target biologically active antibodies, thus identifying suitable vaccine candidates. This strategy can be, in principle, extended to vaccine discovery in a wide range of other pathogens.


Subject(s)
Malaria Vaccines/chemistry , Malaria Vaccines/pharmacology , Plasmodium/genetics , Protozoan Proteins/chemistry , Animals , Antibodies, Protozoan/chemistry , Antibodies, Protozoan/genetics , Antibodies, Protozoan/immunology , Circular Dichroism , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique, Indirect/methods , Genome , Humans , Malaria Vaccines/genetics , Mice , Mice, Inbred Strains , Peptide Fragments/chemistry , Peptide Fragments/immunology , Peptides/chemical synthesis , Peptides/chemistry , Protein Conformation , Protozoan Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...