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.
Vaccine ; 19(32): 4595-602, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11535306

ABSTRACT

Two chimpanzees were vaccinated intramuscularly against malaria using plasmid DNA expressing the pre-erythrocytic antigens thrombospondin related adhesion protein (PfTRAP) and liver stage specific antigen-1 (PfLSA-1) of Plasmodium falciparum together with GM-CSF protein. A recombinant modified vaccinia virus Ankara (MVA) expressing PfTRAP was injected intramuscularly 6 weeks later to boost the immune response. This sequence of antigen delivery induced a specific and long-lasting T cell and antibody response to PfTRAP as detected by ELISPOT assay and ELISA. Antibody responses were detected after four DNA injections, and were boosted by injection of recombinant MVA expressing PfTRAP. Interferon-gamma secreting antigen-specific T cells were detected in both animals, but only after boosting with recombinant MVA. By screening a panel of PfTRAP-derived peptides, an epitope was identified that was recognized by cytotoxic T lymphocytes in one of the chimpanzees studied. T cells specific for this epitope were present in PBMCs and liver-infiltrating lymphocytes at a frequency of between 1 in 200 and 1 in 500. The high immunogenicity of this prime-boost regimen in chimpanzees supports further assessment of this delivery strategy for the induction of protection against P. falciparum malaria in humans.


Subject(s)
Antigens, Protozoan/immunology , Immunization Schedule , Immunization, Secondary , Malaria Vaccines/administration & dosage , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/administration & dosage , Vaccinia virus/genetics , Animals , Antibodies, Protozoan/biosynthesis , Antigens, Protozoan/genetics , COS Cells , Chick Embryo , Chlorocebus aethiops , DNA, Protozoan/genetics , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Fibroblasts/virology , Genetic Vectors/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunity, Cellular , Leukocytes, Mononuclear/immunology , Malaria Vaccines/immunology , Male , Pan troglodytes , Protozoan Proteins/genetics , Recombinant Proteins/pharmacology , Transfection , Vaccines, DNA/immunology
2.
Vaccine ; 18(7-8): 623-32, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10547421

ABSTRACT

In influenza and malaria, CD8+ T cells play an important role in protective immunity in mice. An immunization strategy consisting of DNA priming followed by boosting with recombinant modified vaccinia virus Ankara (MVA) induces complete protection, associated with high levels of CD8+ T cells, against Plasmodium berghei sporozoite challenge in mice. Intradermal delivery of DNA with a gene gun requires smaller amounts of DNA than intramuscular injection, in order to induce similar levels of immune responses. The present study compares both routes for the induction of specific CD8+ T cell responses and protection using different prime-boost immunization regimes in the influenza and the malaria models. In the DNA/MVA regime, equally high CD8+ T cell responses and levels of protection are achieved using ten times less DNA when delivered with a gene gun compared to intramuscular injection.


Subject(s)
Biolistics , CD8-Positive T-Lymphocytes/immunology , Influenza Vaccines/administration & dosage , Malaria Vaccines/administration & dosage , Malaria/prevention & control , Orthomyxoviridae Infections/prevention & control , Vaccines, DNA/administration & dosage , Vaccinia virus/genetics , Animals , DNA, Protozoan/administration & dosage , DNA, Protozoan/genetics , DNA, Viral/administration & dosage , DNA, Viral/genetics , Female , Immunization, Secondary , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Malaria/immunology , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Plasmodium berghei/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccinia virus/immunology
3.
Immunol Rev ; 170: 29-38, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10566139

ABSTRACT

One of the current challenges in vaccine design is the development of antigen delivery systems or vaccination strategies that induce high protective levels of CD8+ T cells. These cells are crucial for protection against certain tumours and intracellular pathogens such as the liver-stage parasite of malaria. A liver-stage malaria vaccine should therefore include CD8+ T-cell-inducing components. This review provides an overview of prime-boost immunisation strategies that result in protective CD8+ T-cell responses against malaria with an emphasis on work from our laboratory. Possible mechanisms explaining why heterologous prime-boost strategies, in particular boosting with replication-impaired recombinant poxviruses, are so effective are discussed.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunization, Secondary/methods , Adjuvants, Immunologic/administration & dosage , Animals , Epitopes/administration & dosage , Genetic Vectors , Humans , Malaria/immunology , Malaria/prevention & control , Malaria Vaccines/administration & dosage , Mice , Primates , Vaccines, DNA/administration & dosage
4.
Biol Chem ; 380(3): 299-303, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10223332

ABSTRACT

Three types of vaccine, all expressing the same antigen from Plasmodium berghei, or a CD8+ T cell epitope from that antigen, were compared for their ability to induce CD8+ T cell responses in mice. Higher levels of lysis and numbers of IFN-gamma secreting T cells were primed with Ty virus-like particles and Modified Vaccinia Virus Ankara (MVA) than with DNA vaccines, but none of the vaccines were able to protect immunised mice from infectious challenge even after repeated doses. However, when the immune response was primed with one type of vaccine (Ty-VLPs or DNA) and boosted with another (MVA) complete protection against infection was achieved. Protection correlated with very high levels of IFN-gamma secreting T cells and lysis. This method of vaccination uses delivery systems and routes that can be used in humans and could provide a generally applicable regime for the induction of high levels of CD8+ T cells.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria/prevention & control , Plasmodium berghei/immunology , Protozoan Proteins/immunology , Vaccines, DNA/immunology , Vaccinia virus/immunology , Animals , Female , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/immunology , Retroelements , Virion/immunology
5.
Nat Med ; 5(5): 565-71, 1999 May.
Article in English | MEDLINE | ID: mdl-10229235

ABSTRACT

Variation in epitopes of infectious pathogens inhibits various effector functions of T lymphocytes through antagonism of the T-cell receptor. However, a more powerful strategy for immune evasion would be to prevent the induction of T-cell responses. We report here mutual 'interference' with the priming of human T-cell responses by a pair of naturally occurring variants of a malaria cytotoxic T-cell epitope. Interference with priming also occurs in vivo for a murine malaria T-cell epitope. Reshaping of the T-cell repertoire by such immune interference during naive T-cell induction may provide a general mechanism for observed patterns of immunodominance and persistence by many polymorphic pathogens.


Subject(s)
Antigenic Variation , Antigens, Protozoan/immunology , Lymphocyte Activation , Malaria, Falciparum/immunology , T-Lymphocytes/immunology , Antigen Presentation , Epitopes , Humans , Ligands , Peptide Fragments/immunology , Protozoan Proteins/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic
6.
Eur J Immunol ; 28(12): 4345-55, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9862371

ABSTRACT

The desirability of inducing cytotoxic T cell responses to defined epitopes in humans has led to the development of a variety of recombinant delivery systems. Recombinant protein particles derived from a yeast retrotransposon (Ty) and the modified Ankara vaccinia (MVA) virus can deliver large epitope strings or even whole proteins. Both have previously been administered safely in humans. Immunization with recombinant Ty and MVA containing a single Plasmodium berghei class I-binding epitope provided 95% sterile protection against malaria in mice. The sequence of immunization, Ty followed by MVA, was critical to elicit high levels of IFN-gamma-producing cells and protection. The reciprocal sequence (MVA/TY) or homologous boosting was not protective. Both constructs (Ty and MVA) contain the H-2Kd-restricted pb9 CTL epitope from the circumsporozoite protein of P. berghei among a string of 8-15 human P. falciparum-derived CTL epitopes restricted through 7 common HLA alleles as well as widely recognized CD4 T cell epitopes. Thus, the novel recombinant Ty/MVA prime/boost combination with these constructs provides a safe alternative for evaluation for human vaccination against P. falciparum malaria.


Subject(s)
Antigens, Protozoan/immunology , Drug Delivery Systems , Histocompatibility Antigens Class I/immunology , Malaria Vaccines , Malaria/immunology , Plasmodium berghei/immunology , T-Lymphocytes/immunology , Animals , Cytotoxicity, Immunologic , Humans , Malaria/prevention & control , Mice , Recombinant Proteins/genetics , Recombinant Proteins/immunology
7.
Nat Med ; 4(4): 397-402, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9546783

ABSTRACT

Immunization with irradiated sporozoites can protect against malaria infection and intensive efforts are aimed at reproducing this effect with subunit vaccines. A particular sequence of subunit immunization with pre-erythrocytic antigens of Plasmodium berghei, consisting of single dose priming with plasmid DNA followed by a single boost with a recombinant modified vaccinia virus Ankara (MVA) expressing the same antigen, induced unprecedented complete protection against P. berghei sporozoite challenge in two strains of mice. Protection was associated with very high levels of splenic peptide-specific interferon-gamma-secreting CD8+ T cells and was abrogated when the order of immunization was reversed. DNA priming followed by MVA boosting may provide a general immunization regime for induction of high levels of CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunization, Secondary , Malaria Vaccines , Malaria/immunology , Plasmodium berghei/immunology , Vaccines, DNA , Vaccinia virus/immunology , Animals , Anopheles/parasitology , Cells, Cultured , Chick Embryo , Cytotoxicity, Immunologic , Female , Humans , Interferon-gamma/biosynthesis , Interleukin-2/pharmacology , Lymphocyte Activation/drug effects , Malaria/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmids , Plasmodium berghei/growth & development , Plasmodium berghei/isolation & purification , Species Specificity , Spleen/immunology
8.
Vaccine ; 16(5): 439-45, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9491498

ABSTRACT

Human immunodeficiency virus (HIV) vaccine candidates were previously constructed as a string of cytotoxic T lymphocyte (CTL) epitopes delivered and expressed using DNA and modified virus Ankara (MVA; an attenuated vaccinia virus) vectors. These vaccines were shown to induce interferon (IFN)-gamma-producing and cytolytic CD8+ T cells after a single vaccine administration. In the course of this work, immunization protocols were sought which would improve the levels of induced HIV-specific T cells. It was found that previous immunological exposure to MVA reduced the efficiency of subsequent priming and boosting using the same vaccine vehicle. However, a combined regime whereby the animals were first primed with the DNA vaccine and then boosted with MVA was the most potent protocol for the induction of both interferon-gamma-producing and cytolytic T cells against two CTL epitopes simultaneously. The general applicability of this novel vaccination method for induction of major histocompatibility complex class I-restricted T cells is discussed.


Subject(s)
DNA, Viral , Histocompatibility Antigens Class I/immunology , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccinia virus/immunology , Viral Vaccines , Animals , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Epitopes , Female , Immunization, Secondary , Interferon Inducers , Interferon-gamma/biosynthesis , Mice , Mice, Inbred BALB C , Vaccines, Attenuated
SELECTION OF CITATIONS
SEARCH DETAIL