Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Cancer Gene Ther ; 16(4): 310-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18949016

ABSTRACT

The delivery of stimulatory signals to dendritic cells (DCs) in the tumor microenvironment could be an effective means to break tumor-induced tolerance. The work presented here evaluates the immunostimulatory properties of pathogen-associated molecular patterns (PAMPs), microbial molecules which bind Toll-like receptors and deliver activating signals to immune cells, when expressed in tumor cells using adenoviral (Ad) vectors. In vitro, transduction of A549 tumor cells with Ad vectors expressing either flagellin from Listeria monocytogenes or P40 protein from Klebsiella pneumoniae induced the maturation of human monocyte-derived DCs in co-cultures. In mixed lymphocyte reactions (MLRs), Ad-flagellin and Ad-P40 transduction of tumor cells stimulated lymphocyte proliferation and the secretion of IFN-gamma. In vivo, these vectors were used either as stand-alone immunoadjuvants injected intratumorally or as vaccine adjuvants combined with a tumor antigen-expressing vector. When Ad-PAMPs were administered intratumorally to mice bearing subcutaneous syngeneic B16F0-CAR (cocksackie-adenovirus receptor) melanomas, tumor progression was transiently inhibited by Ad-P40. In a therapeutic vaccine setting, the combination of Ad-MUC1 and Ad-PAMP vectors injected subcutaneously delayed the growth of implanted RenCa-MUC1 tumors and improved tumor rejection when compared with vaccination with Ad-MUC1 alone. These results suggest that Ad-PAMPs could be effective immunoadjuvants for cancer immunotherapy.


Subject(s)
Adenoviridae/genetics , Bacterial Outer Membrane Proteins/immunology , Genetic Therapy , HN Protein/immunology , Immunotherapy , Neoplasms/therapy , Animals , Bacterial Outer Membrane Proteins/biosynthesis , Bacterial Outer Membrane Proteins/genetics , Cell Line, Tumor , Coculture Techniques , Cytokines/biosynthesis , Dendritic Cells/pathology , Dendritic Cells/physiology , Female , Gene Transfer Techniques , Genetic Vectors , HN Protein/biosynthesis , HN Protein/genetics , Humans , Lymphocyte Activation , Mice , Neoplasms/genetics , Neoplasms/immunology , Newcastle disease virus/genetics
2.
Gene Ther ; 15(20): 1361-71, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18480846

ABSTRACT

We have generated a thymidine kinase gene-deleted vaccinia virus (VV) (Copenhagen strain) that expressed the fusion suicide gene FCU1 derived from the yeast cytosine deaminase and uracil phosphoribosyltransferase genes. Intratumoral inoculation of this thymidine kinase gene-deleted VV encoding FCU1 (VV-FCU1) in the presence of systemically administered prodrug 5-fluorocytosine (5-FC) produced statistically significant reductions in the growth of subcutaneous human colon cancer in nude mice compared with thymidine kinase gene-deleted VV treatments or with control 5-fluorouracil alone. A limitation of prodrug therapies has often been the requirement for the direct injection of the virus into relatively large, accessible tumors. Here we demonstrate vector targeting of tumors growing subcutaneously following systemic administration of VV-FCU1. More importantly we also demonstrate that the systemic injection of VV-FCU1 in nude mice bearing orthotopic liver metastasis of a human colon cancer, with concomitant administration of 5-FC, leads to substantial tumor growth retardation. In conclusion, the insertion of the fusion FCU1 suicide gene potentiates the oncolytic efficiency of the thymidine kinase gene-deleted VV and represents a potentially efficient means for gene therapy of distant metastasis from colon and other cancers.


Subject(s)
Colorectal Neoplasms/therapy , Genes, Transgenic, Suicide , Genetic Therapy/methods , Oncolytic Virotherapy/methods , Transduction, Genetic/methods , Vaccinia virus/genetics , Animals , Antineoplastic Agents/therapeutic use , Cytosine Deaminase/genetics , Flucytosine/therapeutic use , Gene Targeting , Genetic Vectors/administration & dosage , Humans , Injections , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Mice , Mice, Nude , Neoplasm Transplantation , Pentosyltransferases/genetics , Prodrugs/therapeutic use , Transplantation, Heterologous , Virus Replication
3.
Cancer Gene Ther ; 15(1): 18-28, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17992203

ABSTRACT

Modified vaccinia virus Ankara (MVA) has been used successfully to express various antigens for the development of vaccines. Here we show that MVA can also be used as an efficient vector for the transfer of suicide genes to cancer cells. We have generated a new and highly potent suicide gene, FCU1, which encodes a fusion protein derived from the yeast cytosine deaminase and uracil phosphoribosyltransferase genes. We now describe the therapeutic benefit of using MVA to deliver and express the FCU1 gene in cancer cells. MVA-mediated transfer of the FCU1 gene to various human tumor cells results in the production of a bifunctional intracellular enzyme, such that exposure to the prodrug 5-FC suppresses the growth of the tumor cells both in vitro and in vivo. Moreover, we report a more potent tumor growth delay at lower doses of 5-FC using MVA-FCU1 in comparison to adenovirus encoding FCU1. Prolonged therapeutic levels of cytotoxic 5-FU were detected in tumors in mice treated with both MVA-FCU1 and 5-FC while no detectable 5-FU was found in the circulation. This original combination between MVA and FCU1 represents a potentially safe and attractive therapeutic option to test in man.


Subject(s)
Cytosine Deaminase/genetics , Genes, Transgenic, Suicide/genetics , Pentosyltransferases/genetics , Recombinant Fusion Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Vaccinia virus/genetics , Adenoviridae , Animals , Antimetabolites/pharmacology , Cell Line, Tumor , Chick Embryo , Cytosine Deaminase/biosynthesis , Flucytosine/pharmacology , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/therapy , Pentosyltransferases/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Transduction, Genetic , Vaccinia virus/enzymology
4.
Vaccine ; 25(42): 7339-53, 2007 Oct 16.
Article in English | MEDLINE | ID: mdl-17875349

ABSTRACT

We designed and evaluated in HLA-class I transgenic mouse models a hepatitis C virus (HCV) T cell-based MVA vectored vaccine expressing three viral antigens known to be targets of potent CD8+- and CD4+-mediated responses. An accelerated (3 week-based) vaccination induced specific CD8+ T cells harboring two effector functions (cytolytic activity - both in vitro and in vivo- and production of IFNgamma) as well as specific CD4+ T cells recognizing all three vaccine antigens. Responses were long lasting (6 months), boostable by a fourth MVA vaccination and in vivo cross-reactive as demonstrated in a surrogate Listeria-based challenge assay. This candidate vaccine has now moved into clinical trials.


Subject(s)
Hepacivirus/immunology , Immunization Schedule , T-Lymphocytes/immunology , Viral Hepatitis Vaccines/administration & dosage , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/immunology , Cross Reactions , Genetic Vectors , HLA-A2 Antigen/genetics , HLA-B7 Antigen/genetics , Hepacivirus/genetics , Hepatitis C Antigens/genetics , Humans , Immunization, Secondary , Listeria monocytogenes/genetics , Listeria monocytogenes/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Viral Hepatitis Vaccines/genetics , Viral Hepatitis Vaccines/immunology
5.
Cancer Immunol Immunother ; 50(8): 397-407, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11726134

ABSTRACT

The epithelial mucin MUC1 is considered an opportune target antigen for cancer immunotherapy, as it is over-expressed and exhibits aberrant glycosylation in malignant cells. Because dendritic cells (DC) are powerful initiators of immune responses, efforts have focused on tumor antigen-bearing DC as potent cancer vaccines. In this study we have characterized the transduction of monocyte-derived DC with a highly attenuated vaccinia virus vector [modified vaccinia Ankara (MVA)] encoding human MUC1 and the immunostimulatory cytokine IL-2. Analysis of transduced DC cultures generated from a number of donors revealed MUC1 expression in the range of 27-54% of the cells and a co-regulated secretion of bioactive IL-2. As shown by FACS analysis with MUCI-specific antibodies, the MVA-MUC1/IL-2-transduced DC predominantly expressed the fully processed glycoform of MUC1, typical of that displayed by normal epithelia. Over a 3-day period after transduction, transgene expression declined concurrent with an increase in MVA-induced cytopathic effects. The transduced DC stimulated allogeneic lymphocyte proliferation, indicating that DC immunostimulatory function is not impaired by vector transduction. In the presence of IL-2, MVA-transduced DC were able to enhance autologous lymphocyte proliferation. Also, vector expression was analyzed in DC cultures treated with TNF-alpha, a known DC maturation factor. As indicated by the up-regulation of several DC maturation markers, neither virus infection nor transgene expression influenced the maturation capacity of the cells. The MVA-MUC1/IL-2 vector effectively transduced both immature and TNF-alpha-matured DC. Overall, our results are encouraging for the clinical application of MVA-MUC1/IL-2-transduced DC.


Subject(s)
Antigen Presentation , Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Genetic Therapy , Interleukin-2/immunology , Mucin-1/immunology , Antigens, Neoplasm/genetics , Apoptosis , Blood Donors , Cells, Cultured/metabolism , Dendritic Cells/metabolism , Dose-Response Relationship, Immunologic , Genetic Vectors/genetics , Glycosylation , Humans , Interleukin-2/genetics , Kinetics , Leukocytes, Mononuclear/cytology , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Mucin-1/genetics , Mucin-1/metabolism , Protein Processing, Post-Translational , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Transfection , Transgenes , Tumor Necrosis Factor-alpha/pharmacology , Vaccinia virus/genetics
6.
Gene Ther ; 7(17): 1447-57, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11001364

ABSTRACT

Intramuscular administration of plasmid expressing full-length human dystrophin in dystrophin-deficient adult mdx mice resulted in humoral and weak specific T cell responses against the human dystrophin protein. Following plasmid injection, human dystrophin was detected in the injected muscles at 7 days, but decreased thereafter. Anti-dystrophin antibodies were found 21 days following plasmid injection, which coincided with transient myositis. This immune rejection prevented the mice from expressing human dystrophin after a second plasmid injection. No anti-DNA antibodies were found. Anti-dystrophin antibodies were seen in a smaller proportion of plasmid-injected dystrophin-competent C57BL/10 mice, suggesting that the immune rejection of dystrophin may be explained partially by species differences in the dystrophin protein.


Subject(s)
Antibodies/analysis , Dystrophin/genetics , Genetic Therapy/adverse effects , Muscular Dystrophy, Duchenne/therapy , Plasmids/administration & dosage , T-Lymphocytes/immunology , Animals , Blotting, Western/methods , Dystrophin/analysis , Dystrophin/immunology , Enzyme-Linked Immunosorbent Assay/methods , Genetic Therapy/methods , Humans , Immunohistochemistry/methods , Injections, Intramuscular , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/chemistry , Muscle, Skeletal/immunology , Muscular Dystrophy, Duchenne/immunology , Species Specificity
7.
J Immunother ; 23(5): 570-80, 2000.
Article in English | MEDLINE | ID: mdl-11001550

ABSTRACT

Polymorphic epithelial mucin, encoded by the MUC1 gene, is present at the apical surface of glandular epithelial cells. It is over-expressed and aberrantly glycosylated in most breast tumors, resulting in an antigenically distinct molecule and a potential target for immunotherapy. This transmembrane protein, when produced by tumor cells, is often cleaved into the circulation, where it is detectable as a tumor marker (CA 15.3) by various antibodies, allowing for early detection of recurrences and evaluation of treatment efficacy. The objective of the current study was to examine the clinical and environmental safety and immunogenicity of a live recombinant vaccinia virus expressing the human MUC1 and IL2 genes (VV TG5058), referred to here as TG1031. The study was an open-label phase 1 and 2 trial in nine patients with advanced inoperable breast cancer recurrences to the chest wall. The patients were vaccinated intramuscularly with a single dose of TG1031; three patients were treated at each of three progressive dose levels ranging from 5x10(5) to 5x10(7) plaque-forming units. A boost injection at their original dose level was administered in patients responding immunologically, clinically, or both. Vaccination resulted in no significant clinical adverse effects, and there was no environmental contamination by live TG1031. All patients had been vaccinated as children, and patients treated at the highest dose level mounted a significant anti-vaccinia antibody response. None of the nine patients had a significant increase in MUC1-specific antibody titers after one single injection, whereas five patients had a detectable increase in vaccinia virus antibody titers. Peripheral blood mononuclear cells of one patient at the intermediate dose level showed a proliferative response to in vitro culture with vaccinia virus, with a stimulation index of 6. A second patient treated at the intermediate dose level had a stimulation index of 7 to MUC1 peptide and of 14 after a boost injection. This patient had a concomitant decrease in carcinoembryonic antigen serum levels and remained clinically stable for 10 weeks. Evidence of MUC1-specific cytotoxic T lymphocytes was detected in two patients. Immunohistochemical analysis revealed an increase in T memory cells (CD45RO) in tumor biopsies after vaccination. The absence of serious adverse events, together with the documentation of immune stimulations in vivo, warrant the further use of TG1031 in immunotherapy trials of breast cancer.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/therapy , Immunotherapy/methods , Recombinant Fusion Proteins/therapeutic use , Vaccinia virus/immunology , Viral Fusion Proteins/therapeutic use , Adult , Aged , Breast Neoplasms/pathology , Cancer Vaccines/administration & dosage , Cytokines/metabolism , DNA, Complementary/metabolism , DNA, Viral/metabolism , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Humans , Immunity, Cellular , Interleukin-2/genetics , Interleukin-2/immunology , Lymphatic Metastasis , Middle Aged , Mucin-1/immunology , Mucin-1/metabolism , Recombinant Fusion Proteins/immunology , Treatment Outcome , Viral Fusion Proteins/immunology
8.
Hum Gene Ther ; 11(10): 1417-28, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10910139

ABSTRACT

Antigen-specific recognition and subsequent destruction of tumor cells is the goal of vaccine-based immunotherapy of cancer. Often, however, tumor antigen-specific cytotoxic T lymphocytes (CTLs) are either not available or in a state of anergy. In addition, MHCI expression on tumor cells is often downregulated. Either or both of these situations can allow tumor growth to proceed unchecked by CTL control. We have shown previously that tumor antigen-specific monoclonal antibodies can be expressed in vaccinia virus and that activated macrophages infected with this virus acquire the ability to kill tumor cells expressing that antigen. Here we show that a membrane-anchored form of the scFv portion of the MUC1 tumor antigen-specific monoclonal antibody, SM3, can be expressed on activated macrophages with the highly attenuated poxvirus, modified vaccinia Ankara (MVA), as a gene transfer vector. Cells infected with the MVA-scFv construct were shown to express the membrane-bound scFv by Western blot and FACS analysis. That cells expressing the membrane-anchored scFv specifically bind antigen was shown by FACS and by BIAcore analysis. GM-CSF-activated macrophages were infected with the construct and shown to recognize specifically MUC1-expressing tumor cells as measured by IL-12 release. Furthermore, activated macrophages expressing the membrane-bound scFv specifically lyse target cells expressing the MUC1 antigen but not cells that do not express MUC1.


Subject(s)
Antibodies/immunology , Genetic Vectors , Macrophages/cytology , Macrophages/metabolism , Neoplasms/therapy , Animals , Base Sequence , Biosensing Techniques , Blotting, Western , Cell Death , Cell Separation , Chick Embryo , DNA, Complementary/metabolism , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-12/metabolism , Mice , Models, Genetic , Molecular Sequence Data , Mucin-1/genetics , Mucin-1/immunology , Mucins/genetics , Mucins/immunology , Peptides/genetics , Peptides/immunology , Phenotype , Poxviridae/genetics , Time Factors , Transfection , Tumor Cells, Cultured , Vaccinia virus/genetics
9.
Virology ; 273(1): 9-15, 2000 Jul 20.
Article in English | MEDLINE | ID: mdl-10891402

ABSTRACT

Modified vaccinia virus Ankara (MVA) is a highly attenuated virus strain that may be useful as a vaccine vector. Ultrastructural examination of purified MVA showed that most of the viral particles are enveloped in contrast to the Copenhagen strain (COP). In CsCl gradients, the majority of the MVA particles displayed a light buoyant density characteristic of the enveloped form. Consistent with these results, MVA particles were poorly labeled with antibodies against the surface of intracellular mature virus but strongly labeled with antibodies against an envelope antigen. Furthermore, MVA was more resistant than the COP strain to neutralization by mouse anti-COP antibodies. These results suggest that the MVA strain may be particularly suitable for the engineering of envelope proteins and that MVA may be able to resist the humoral immunity displayed by previously vaccinated individuals.


Subject(s)
Vaccinia virus/classification , Vaccinia virus/ultrastructure , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Assembly , Animals , Antibodies, Viral/immunology , Cells, Cultured , Centrifugation, Density Gradient , Chick Embryo , Cricetinae , Fibroblasts , Mice , Microscopy, Electron , Molecular Conformation , Neutralization Tests , Vaccines, Attenuated/chemistry , Vaccinia virus/chemistry , Vaccinia virus/physiology , Viral Envelope Proteins/immunology
10.
Cancer Immunol Immunother ; 48(10): 588-94, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10630311

ABSTRACT

Analyses of MUC1-specific cytotoxic T cell precursor (CTLp) frequencies were performed in mice immunized with three different MUC1 vaccine immunotherapeutic agents. Mice were immunized with either a fusion protein comprising MUC1 and glutathione S-transferase (MUC1-GST), MUC1-GST fusion protein coupled to mannan (MFP) or with a recombinant vaccinia virus expressing both MUC1 and interleukin-2. Mouse strain variations in immune responsiveness have been observed with these vaccines. We have constructed mice transgenic for the human MUC1 gene to study MUC1-specific immune responses and the risk of auto-immunity following MUC1 immunization. Transgenic mice immunized with MUC1 were observed to be partially tolerant in that the MUC1-specific antibody response is lower than that observed in syngeneic but non-transgenic mice. However, a significant MUC1-specific CTLp response to all three vaccines was observed, indicating the ability to overcome T cell, but to a lesser extent B cell, tolerance to MUC1 in these mice. Histological analysis indicates no evidence of auto-immunity to the cells expressing the human MUC1 molecule. These results suggest that it is possible to generate an immune response to a cancer-related antigen without damage to normal tissues expressing the antigen.


Subject(s)
Cancer Vaccines/immunology , Mucin-1/immunology , Animals , Antibodies, Neoplasm/blood , Humans , Immune Tolerance , Mice , Mice, Transgenic , Mucin-1/genetics , Neoplasms, Glandular and Epithelial/therapy , T-Lymphocytes, Cytotoxic/immunology , Vaccination
11.
Cell Mol Biol (Noisy-le-grand) ; 40 Suppl 1: 49-59, 1994.
Article in English | MEDLINE | ID: mdl-7950862

ABSTRACT

Breast cancer is considered as the major cause of mortality by cancer for women. Even if chemotherapy, radiotherapy and surgery have improved the life expectancy of patients bearing tumours, breast cancer is responsible for the death of 42,000 women per year in USA and 25,000 women in France. In this context, cancer vaccines may add an attractive alternative therapeutic strategy to the current existing treatments. We describe here the construction of recombinant vaccinia viruses co-expressing a tumour associated antigen (MUC 1) and an "adjuvant" cytokine, which have potential applications in the active immunotherapy of breast cancer. Indeed, recombinant vaccinia viruses have been extensively used during the past decade to induce a protective response against a whole variety of pathogens, and has proven to be of great value in the elicitation of a cellular immune response leading to the rejection of tumour grafts in mouse models.


Subject(s)
Antigens, Neoplasm/genetics , Breast Neoplasms/therapy , Immunotherapy/methods , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mucins/genetics , Mucins/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology , Animals , Base Sequence , Breast Neoplasms/immunology , Combined Modality Therapy , Cytokines/genetics , DNA, Complementary/genetics , Female , Genetic Vectors , Humans , Mice , Molecular Sequence Data , Mucin-1 , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Repetitive Sequences, Nucleic Acid
12.
J Immunother Emphasis Tumor Immunol ; 14(2): 136-43, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8280702

ABSTRACT

MUC1 is a mucin found on the apical surfaces of some normal mammalian mucin-secreting cells. It is characterized by heavy glycosylation and a 20-amino-acid tandem repeat segment. In most cases of human breast adenocarcinoma, this antigen is overexpressed. Moreover, abnormal glycosylation exposes a novel peptide epitope within the tandem repeat, such that antibodies to this epitope can distinguish normal from malignant adenocarcinomatous breast tissue. We have constructed a vaccinia virus (VV) that carries the cDNA for the MUC1 antigen. Murine and human cells infected with this virus express the MUC1 molecule, with three to four tandem repeats per molecule and with the tumor-associated epitopes exposed. Mice immunized with this virus produce antibodies that recognize MUC1 outside the tandem repeat, within the tandem repeat, and within the tumor-associated protein core epitope. Tumorigenic P815 (DBA) and 3T3 (BALB/c) cells have been transfected with MUC1. Thirty percent of DBA mice immunized with VV-MUC1 are protected from growth of P815-MUC1 tumors when implanted with 10(5) cells. Immunized BALB/c mice show a late development of transfected 3T3 tumor cells. Immunized mice show a moderate MUC1-specific IgG titer, but it cannot be correlated with subsequent tumor rejection. No evidence for a MUC1-specific cytotoxic T lymphocyte response has been found after immunization with VV-MUC1.


Subject(s)
Antigens, Neoplasm , Antigens, Viral/immunology , Immunization , Mucins/immunology , Neoplasms, Experimental/immunology , Vaccinia virus/immunology , Animals , Antibody Formation , Cell Division/immunology , Disease Models, Animal , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Neoplasm Transplantation
13.
Parasite Immunol ; 13(5): 473-90, 1991 Sep.
Article in English | MEDLINE | ID: mdl-1956696

ABSTRACT

A member of the glutathione S-transferase family, Sm28GST has previously demonstrated a good ability to protect rodents against experimental infection with Schistosoma mansoni. In order to evaluate its efficacy in a model closer to man, two different protocols of immunization with recombinant Sm28GST were tested on baboons in a large-scale trial. Three injections in the presence of aluminium hydroxide as adjuvant resulted in a significant 38% reduction in the adult worm burden together with a trend for a lower percentage of inflammatory tissue in the liver. Individual levels of protection, ranging from 0 to 80%, underlined the heterogeneity of the immune response to this purified molecule in outbred primates. On the other hand, two injections of Sm28GST in the presence of aluminium hydroxide and Bordetella pertussis reduced female schistosome fecundity by 33%, with a more pronounced effect (66%) on faecal egg output; there was also a trend, in this protocol, for decrease of the mean granuloma surface in the liver. Individual anti-Sm28GST IgG antibodies were apparently unrelated to levels of immunity, but there was partial evidence that cytophilic IgE might play a role in the immune mechanisms affecting worm viability, but not fecundity. In the mouse model, Sm28GST vaccination resulted in a lower hatching ability of tissue eggs recovered from immunized mice whereas passive transfer of specific anti-Sm28GST T-lymphocytes, one day before infection, significantly reduced the number of eggs in the liver of mice. We propose that different protocols of immunization with a recombinant molecule can impede Schistosoma mansoni worm viability and fecundity, but can also affect miracidium physiology, with important consequences for disease transmission and granuloma-derived pathology.


Subject(s)
Glutathione Transferase , Schistosoma mansoni/growth & development , Schistosomiasis mansoni/prevention & control , Vaccines, Synthetic , Animals , Antibodies, Helminth/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Survival , Female , Male , Mice , Mice, Inbred BALB C , Papio , Parasite Egg Count , Schistosoma mansoni/immunology , Schistosoma mansoni/isolation & purification , Schistosomiasis mansoni/immunology , T-Lymphocytes/immunology
14.
J Clin Microbiol ; 28(9): 1918-24, 1990 Sep.
Article in English | MEDLINE | ID: mdl-2121788

ABSTRACT

The 28-kilodalton antigen of Schistosoma mansoni has been previously described as having importance as the basis for a potential vaccine. The P28 recombinant molecule and three peptides derived from its primary sequence, namely the 24-43, 115-131, and 140-153 peptides, have been tested to evaluate the humoral responses of Kenyan school children previously classified as susceptible or resistant to reinfection after chemotherapy. We report here that the P28 molecule and two of the peptides studied (peptides 115-131 and 140-153) can be used for detecting specific immunoglobulin G (IgG), IgE, and IgA antibodies. Moreover, the IgG4 response of the susceptible population was significantly greater than that of the resistant group, whereas no differences between the two populations were noticed in total IgG anti-P28 antibodies. This suggested that IgG4 could play a role in the lack of immunity of susceptible patients. A strong IgG3 response restricted to the 140-153 peptide was observed but did not discriminate between the resistant and susceptible populations. In contrast, a marked increase in the IgA response to the 140-153 peptide epitope(s) in sera of the resistant population was noticed. Taken together, these results suggest that the P28 antigen and two of the three peptides selected could give predictive information about the development of the disease or the efficiency of vaccination with P28 as the immunogen.


Subject(s)
Antibodies, Helminth/biosynthesis , Antigens, Helminth , Helminth Proteins , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Adolescent , Animals , Child , Glutathione Transferase/immunology , Humans , Immunoglobulin E/biosynthesis , Immunoglobulin G/biosynthesis , Immunoglobulin G/classification , Peptides/chemical synthesis , Peptides/immunology , Recombinant Proteins/immunology , Schistosomiasis mansoni/prevention & control , Vaccines, Synthetic/immunology
15.
Mol Biochem Parasitol ; 41(1): 35-44, 1990 Jun.
Article in English | MEDLINE | ID: mdl-2385266

ABSTRACT

A Schistosoma mansoni cDNA library was constructed from the mRNA of adult worms in the expression vector lambda gt11 and screened with a rabbit antiserum raised against the 26-kDa S. mansoni glutathione S-transferase isoforms (Sm GST 26). Two clones were selected and the nucleotide sequences deduced. The predicted amino acid sequence, specified by these cDNAs, shows strong homology with a Schistosoma japonicum 26 kDa glutathione S-transferase and a lower level of homology with mammalian glutathione S-transferase class mu isoenzymes (EC 2.5.1.18). No significant homology score was found with a 28-kDa S. mansoni glutathione S-transferase (Sm GST 28). A study of the tissue distribution of the cloned Sm GST 26 by immunoelectron microscopy shows similarities to Sm GST 28 in that they are present in the tegument and in subtegumentary parenchymal cells. However, a major difference exists in the protonephridial region in which Sm GST 26 is present in the cytoplasmic digitations localized in the apical chamber delineated by the flame cell body, suggesting that Sm GST 26 may be actively excreted by adult worms.


Subject(s)
Glutathione Transferase/analysis , Schistosoma mansoni/enzymology , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Cloning, Molecular , Electrophoresis, Gel, Two-Dimensional , Glutathione Transferase/genetics , Immune Sera , Immunohistochemistry , Microscopy, Electron , Molecular Sequence Data , Schistosoma mansoni/genetics , Sequence Homology, Nucleic Acid
16.
Yeast ; 5(6): 497-507, 1989.
Article in English | MEDLINE | ID: mdl-2694678

ABSTRACT

Strains of Saccharomyces cerevisiae expressing P28-I, an antigen inducing protection against schistosomiasis, have been constructed. Transformants containing a very high copy number of a P28-I expression vector were selected by genetic complementation involving deficient LEU2 or URA3 alleles carried by plasmids. Using the ura3 fur1 auto-selection system, constitutive and stable expression of P28-I could be obtained in cultures grown in rich medium. The accumulation of the foreign protein exceeds 25% of total yeast proteins when estimated by Coomassie Brilliant Blue staining of SDS-PAGE. Moreover, P28-I which was located intracellularly was soluble and biologically active.


Subject(s)
Antigens, Helminth/biosynthesis , Saccharomyces cerevisiae/metabolism , Schistosoma mansoni/immunology , Animals , Antigens, Helminth/genetics , Base Sequence , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Glutathione Transferase/isolation & purification , Glutathione Transferase/metabolism , Plasmids , Recombinant Proteins/biosynthesis , Transformation, Genetic
17.
J Immunol ; 142(12): 4416-21, 1989 Jun 15.
Article in English | MEDLINE | ID: mdl-2723435

ABSTRACT

In hypereosinophilic patients, eosinophil heterogeneity has been assessed mainly according to morphologic and biologic criteria. In order to investigate the molecular basis of such heterogeneity, biochemical analysis was performed on various eosinophil subpopulations fractionated on metrizamide gradients. Whole cell extracts from purified eosinophils disrupted with a nonionic (NP-40) detergent were successively analyzed by SDS-PAGE and two-dimensional electrophoresis (isoelectric focusing or nonequilibrium pH gradient electrophoresis in the first dimension). Hypodense eosinophils that sediment in the lightest density gradients (18 to 22% metrizamide solution) differed from other purified eosinophils (intermediate and normodense eosinophils respectively collected in 22 to 23% and 23 to 25% metrizamide solutions). Comparative analysis of protein patterns on both monodimensional and bidimensional electrophoresis showed that a basic protein of Mr 51 kDa, present on normodense or intermediate eosinophils, was poorly detected in the case of hypodense eosinophils. In contrast, two other proteins with apparent Mr of about 23 kDa and 41 kDa were exclusively or predominantly identified in these latter cell fractions. Immunochemical analysis with polyclonal antibodies against eosinophil basic proteins and enzymatic assays revealed that the 51-kDa polypeptide could be related to an eosinophil peroxidase-like molecule. In addition, the two proteins detected only in hypodense eosinophils might be related to proteins newly synthesized by in vivo activated eosinophils. Our results suggest that variations in protein expression might represent a good marker of in vivo activation.


Subject(s)
Blood Proteins/metabolism , Eosinophils/classification , Blood Proteins/isolation & purification , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Eosinophil Peroxidase , Eosinophilia/blood , Eosinophils/enzymology , Eosinophils/physiology , Humans , Leukocyte Count , Peroxidases/blood
18.
J Immunol ; 142(4): 1342-50, 1989 Feb 15.
Article in English | MEDLINE | ID: mdl-2492579

ABSTRACT

The P28-1 Ag induces a strong protective immunity toward Schistosoma mansoni infection in various experimental models. T lymphocytes of mice immunized with the recombinant P28-1 Ag were stimulated in vitro by schistosome Ag of different development stages and by three P28-1 Ag-derived synthetic peptides. The most significant stimulation was achieved with the 24-43 peptide. The use of two fragments of this peptide showed that the P28-1 T lymphocyte specificity concerned essentially the NH2 terminal sequence of the 24-43 peptide. Moreover, T lymphocytes specific for the 24-43 peptide were stimulated by both schistosome Ag and the recombinant P28-1 protein. The passive transfer of (Th + Ts) lymphocytes recovered from P28-1 Ag-immunized mice increased the IgG response to P28-1 and its peptides during infection but did not protect against a challenge infection, such as the passive transfer of anti-P28-1 sera. In contrast, P28-1 specific Th cell lines maintained in culture for 2 mo, passively transferred a strong protection (50%) to infected mice. Supernatants of P28-1-specific T cells obtained after stimulation with the corresponding Ag, were able to confer cytotoxic properties to platelets and macrophages. The presence of IFN-gamma for the cytotoxicity mediated by platelets and macrophage activating factor for the cytotoxicity mediated by macrophages in these supernatants is in a large part responsible for the parasite killing observed. Finally, a preliminary immunogenetic approach with H-2 congenic mice on BALB background showed that the P28-1 Ag T cell response was under the control of the MHC and that the H-2b haplotype determined a low response to P28-1 Ag and its peptides while H-2d and k haplotypes determined high responders.


Subject(s)
Antigens, Helminth/administration & dosage , Schistosomiasis mansoni/prevention & control , Vaccines, Synthetic/administration & dosage , Vaccines/administration & dosage , Animals , Antibodies, Helminth/administration & dosage , Antigens, Helminth/analysis , Antigens, Helminth/immunology , Blood Platelets/immunology , Cell-Free System , Cytotoxicity, Immunologic , Immune Sera/administration & dosage , Immunization, Passive , Lymphocyte Activation , Macrophages/immunology , Mice , Mice, Inbred BALB C , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Vaccines, Synthetic/analysis , Vaccines, Synthetic/immunology
19.
J Immunol ; 141(5): 1687-94, 1988 Sep 01.
Article in English | MEDLINE | ID: mdl-2457624

ABSTRACT

The Schistosoma mansoni P28 molecule is an Ag inducing protective immunity in various experimental models. Three synthetic peptides, derived from the primary sequence of the recombinant P28 and comprising amino acids 24-43, 115-131, and 140-153, respectively, were synthesized according to their hydrophilicity, mobility, and accessibility profiles. The presence of B and T lymphocyte epitopes in these peptides has been examined in the rat model. The results showed that the 24-43 and the 115-131 peptides contained major epitopes for IgG but not for IgE. Moreover, the 24-43 peptide-specific IgG produced after injecting either the recombinant P28 Ag or the 24-43 peptide coupled to tetanus toxoid was essentially of the IgG2a subclass and to a lesser extent of the IgG1 subclass, whereas no IgG2c was detected. These 24-43 peptide-specific antibodies were cytotoxic in vitro for schistosomula in the presence of eosinophils as effector cells. The 24-43 and the 140-153 peptides contained major targets of T lymphocytes specific for the recombinant P28 Ag. T cell lines specific for the 24-43 peptide have been prepared. These cells proliferated in vitro when stimulated with various S. mansoni crude antigenic preparations or with the recombinant P28 Ag. Moreover, their passive transfer to rats immunized with the P28 Ag led to a significant increase in specific IgE without modifying the IgG response.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , Antigens, Helminth/analysis , Helminth Proteins , Peptides/immunology , Schistosomiasis mansoni/immunology , Animals , Antibody-Dependent Cell Cytotoxicity , Antigens, Helminth/immunology , Cell Line , Disease Models, Animal , Eosinophils/immunology , Epitopes/analysis , Immunization, Passive , Immunoglobulin G/physiology , Lymphocyte Activation , Male , Peptides/analysis , Rats , Rats, Inbred F344 , T-Lymphocytes/immunology
20.
EMBO J ; 7(2): 465-72, 1988 Feb.
Article in English | MEDLINE | ID: mdl-3284744

ABSTRACT

A protective Mr28K antigen of Schistosoma mansoni, expressed from its cDNA, has been purified in a single step and shown to possess glutathione (GSH) transferase activity as predicted from sequence homologies with two mammalian GSH transferase multigene families. It is notable for its high 1-chloro-2,4-dinitrobenzene GSH transferase and linoleic acid hydroperoxide GSH peroxidase activities. The major GSH transferase of S. mansoni has been purified and its subunit is identical to this Mr28K antigen by criteria of Mr, immunochemistry, substrate specificity and peptide sequence analysis. In the parasite, the antigen is present in the tegument, protonephridial cells and subtegumental parenchymal cells. No significant immunological cross-reactivity between the S.mansoni and mammalian (human and rat) GSH transferases was observed.


Subject(s)
Antigens, Helminth/genetics , Cloning, Molecular , Glutathione Transferase/genetics , Schistosoma mansoni/genetics , Amino Acid Sequence , Animals , Antigens, Helminth/immunology , Cross Reactions , Escherichia coli/genetics , Glutathione Transferase/immunology , Glutathione Transferase/metabolism , Humans , Macromolecular Substances , Microscopy, Electron , Molecular Sequence Data , Molecular Weight , Saccharomyces cerevisiae/genetics , Schistosoma mansoni/enzymology , Schistosoma mansoni/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...