Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Immun Inflamm Dis ; 5(3): 346-354, 2017 09.
Article in English | MEDLINE | ID: mdl-28508570

ABSTRACT

INTRODUCTION: While most transcripts arising from the human T Cell Receptor locus reflect fully rearranged genes, several germline transcripts have been identified. We describe a new germline transcript arising from the human TCRB locus. METHODS: cDNA sequencing, promoter, and gene expression analyses were used to characterize the new transcript. RESULTS: The new germline transcript encoded by the human TCRB locus consists of a new exon of 103 bp, which we named TRBX1 (X1), spliced with the first exon of gene segments Cß1 or Cß2. X1 is located upstream of gene segment Dß1 and is therefore deleted from a V-DJ rearranged TCRB locus. The X1-Cß transcripts do not appear to code for a protein. We define their transcription start and minimal promoter. These transcripts are found in populations of mature T lymphocytes from blood or tissues and in T cell clones with a monoallelic TCRB rearrangement. In immature thymocytes, they are already detectable in CD1a- CD34+ CD4- CD8- cells, therefore before completion of the TCRB rearrangements. CONCLUSIONS: The X1 promoter appears to be the ortholog of the mouse pre-Dß1 promoter (PDß1). Like PDß1, its activation is regulated by Eß in T cells and might facilitate the TCRB rearrangement process by contributing to the accessibility of the Dß1 locus.


Subject(s)
Genes, T-Cell Receptor beta , Genetic Loci , Promoter Regions, Genetic , RNA, Messenger/genetics , Transcription, Genetic , Animals , Humans , Mice , RNA, Messenger/biosynthesis
2.
Proc Natl Acad Sci U S A ; 111(9): 3502-7, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24550491

ABSTRACT

Female mice of inbred strain CBA do not reject syngeneic male skin grafts even though they mount a T-cell response against the male-specific HY antigen. We show that local immunostimulation performed by injecting cytokines and Toll-like receptor ligands in close vicinity to the graft causes rejection. We feel that this approach should be tested in tumor-bearing human patients in combination with antitumor vaccination. Relief of intratumor immunosuppression may increase considerably the fraction of patients who respond to vaccination directed against tumor antigens recognized by T cells.


Subject(s)
Disease Models, Animal , Graft Rejection/chemically induced , Immune Tolerance/immunology , Immunization/methods , Neoplasms/therapy , Adoptive Transfer , Animals , Cytokines/adverse effects , Cytokines/immunology , DNA Primers/genetics , Female , Immunization/adverse effects , Male , Mice , Mice, Inbred CBA , Neoplasms/immunology , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors , Skin Transplantation/methods
3.
PLoS One ; 8(9): e76186, 2013.
Article in English | MEDLINE | ID: mdl-24098777

ABSTRACT

GARP is a transmembrane protein present on stimulated human regulatory T lymphocytes (Tregs), but not on other T lymphocytes (Th cells). It presents the latent form of TGF-ß1 on the Treg surface. We report here that GARP favors the cleavage of the pro-TGF-ß1 precursor and increases the amount of secreted latent TGF-ß1. Stimulated Tregs, which naturally express GARP, and Th cells transfected with GARP secrete a previously unknown form of latent TGF-ß1 that is disulfide-linked to GARP. These GARP/TGF-ß1 complexes are possibly shed from the T cell surface. Secretion of GARP/TGF-ß1 complexes was not observed with transfected 293 cells and may thus be restricted to the T cell lineage. We conclude that in stimulated human Tregs, GARP not only displays latent TGF-ß1 at the cell surface, but also increases its secretion by forming soluble disulfide-linked complexes. Moreover, we identified six microRNAs (miRNAs) that are expressed at lower levels in Treg than in Th clones and that target a short region of the GARP 3' UTR. In transfected Th cells, the presence of this region decreased GARP levels, cleavage of pro-TGF-ß1, and secretion of latent TGF-ß1.


Subject(s)
Membrane Proteins/immunology , MicroRNAs/metabolism , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Immunoprecipitation , Luciferases , Membrane Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/metabolism
4.
Cancer Res ; 71(4): 1253-62, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21216894

ABSTRACT

A core challenge in cancer immunotherapy is to understand the basis for efficacious vaccine responses in human patients. In previous work we identified a melanoma patient who displayed a low-level antivaccine cytolytic T-cell (CTL) response in blood with tumor regression after vaccination with melanoma antigens (MAGE). Using a genetic approach including T-cell receptor ß (TCRß) cDNA libraries, we found very few antivaccine CTLs in regressing metastases. However, a far greater number of TCRß sequences were found with several of these corresponding to CTL clones specific for nonvaccine tumor antigens, suggesting that antigen spreading was occurring in regressing metastases. In this study, we found another TCR belonging to tumor-specific CTL enriched in regressing metastases and detectable in blood only after vaccination. We used the TCRß sequence to detect and clone the desired T cells from tumor-infiltrating lymphocytes isolated from the patient. This CD8 clone specifically lysed autologous melanoma cells and displayed HLA-A2 restriction. Its target antigen was identified as the mitochondrial enzyme caseinolytic protease. The target antigen gene was mutated in the tumor, resulting in production of a neoantigen. Melanoma cell lysis by the CTL was increased by IFN-γ treatment due to preferential processing of the antigenic peptide by the immunoproteasome. These results argue that tumor rejection effectors in the patient were indeed CTL responding to nonvaccine tumor-specific antigens, further supporting our hypothesis. Among such antigens, the mutated antigen we found is the only antigen against which no T cells could be detected before vaccination. We propose that antigen spreading of an antitumor T-cell response to truly tumor-specific antigens contributes decisively to tumor regression.


Subject(s)
Antigens, Neoplasm/metabolism , Cancer Vaccines/therapeutic use , Melanoma/therapy , Neoplasm Proteins/immunology , Skin Neoplasms/therapy , Amino Acid Sequence , Antigens, Neoplasm/immunology , Base Sequence , Cancer Vaccines/pharmacology , Cells, Cultured , Humans , K562 Cells , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Melanoma-Specific Antigens , Models, Biological , Molecular Sequence Data , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Remission Induction , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology , Tissue Distribution/immunology , Tissue Distribution/physiology , Tumor Burden
5.
J Immunol ; 180(5): 3585-93, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18292586

ABSTRACT

We previously characterized the CTL response of a melanoma patient who experienced tumor regression following vaccination with an ALVAC virus coding for a MAGE-A3 Ag. Whereas anti-vaccine CTL were rare in the blood and inside metastases of this patient, anti-tumor CTL recognizing other tumor Ags, mainly MAGE-C2, were 100 times more frequent in the blood and considerably enriched in metastases following vaccination. In this study we report the analysis of the CTL response of a second melanoma patient who showed a mixed tumor response after vaccination with dendritic cells pulsed with two MAGE-A3 antigenic peptides presented, respectively, by HLA-A1 and HLA-DP4. Anti-MAGE-3.A1 CD8 and anti-MAGE-3.DP4 CD4 T cells became detectable in the blood after vaccination at a frequency of approximately 10(-5) among the CD8 or CD4 T cells, respectively, and they were slightly enriched in slowly progressing metastases. Additional anti-tumor CTL were present in the blood at a frequency of 2x10(-4) among the CD8 T cells and, among these, an anti-MAGE-C2 CTL clone was detected only following vaccination and was enriched by >1,000-fold in metastases relative to the blood. The striking similarity of these results with our previous observations further supports the hypothesis that the induction of a few anti-vaccine T cells may prime or restimulate additional anti-tumor T cell clones that are mainly responsible for the tumor regression.


Subject(s)
Antigens, Neoplasm/therapeutic use , Dendritic Cells/immunology , Dendritic Cells/transplantation , Immunotherapy, Adoptive/methods , Melanoma/immunology , Melanoma/therapy , Neoplasm Proteins/therapeutic use , Peptide Fragments/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cell Differentiation/immunology , Cell Line, Transformed , Cytotoxicity Tests, Immunologic , Dendritic Cells/metabolism , Humans , K562 Cells , Lymphocyte Count , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Melanoma/pathology , Melanoma/secondary , Molecular Sequence Data , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/secondary , Neoplasm Recurrence, Local/therapy , Peptide Fragments/immunology , Peptide Fragments/metabolism , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/secondary , Skin Neoplasms/therapy , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology , Tumor Cells, Cultured
6.
Cancer Immunol Immunother ; 56(6): 807-18, 2007 Jun.
Article in English | MEDLINE | ID: mdl-16988823

ABSTRACT

Because of the high frequency of HLA-DP4 in the Caucasian population, we have selectively delineated HLA-DP4 restricted T cell epitopes in the MAGE-A tumor antigens. We identified 12 good binders to HLA-DP4 and investigated the capacity of the seven best binders to induce in vitro specific CD4+ T cell lines from HLA-DP4 healthy donors. We found that the MAGE-A1 90-104 peptide exhibited a high and constant frequency of CD4+ T cell precursors in all the six tested donors. The MAGE-A1 268-282 peptide was found immunogenic in only two donors but with a high precursor frequency. The MAGE-A12 127-141 peptide was T cell stimulating in six different donors and induced fewer T cell lines. The peptide-specific T cell lines were stimulated by DC loaded with the lysates of cells transfected with MAGE-A1 or MAGE-A12, or loaded with the recombinant protein. We also show that the immunoreactivity of CD4+ T cell epitopes restricted to the same HLA II molecule may vary from one individual to another, as a result of inter-individual variations in the CD4+ T cell repertoire.


Subject(s)
Antigens, Neoplasm/immunology , Epitopes, T-Lymphocyte/immunology , HLA-DP Antigens/immunology , Neoplasm Proteins/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , COS Cells , Chlorocebus aethiops , HLA-DP beta-Chains , Humans , Lymphocyte Activation/immunology , Melanoma-Specific Antigens
7.
Cancer Immunol Immunother ; 55(6): 644-52, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16187088

ABSTRACT

Antigens encoded by genes of the LAGE family, including LAGE-1 and NY-ESO-1, are of interest for cancer immunotherapy because they are tumor-specific and shared by tumors of different histological types. Several clinical trials are in progress with NY-ESO-1 peptides, protein, recombinant poxviruses, and dendritic cells pulsed with peptides. In this study, CD8 T lymphocytes from an individual without cancer were stimulated with dendritic cells infected with a recombinant avian poxvirus encoding a complete LAGE-1 protein. A CTL clone was isolated that recognized a new LAGE-1 peptide, ELVRRILSR, which corresponds to position 103-111 of the protein sequence. It is presented by HLA-A6801 molecules. When tumor cells expressing LAGE-1 were transfected with HLA-A68, they were lysed by the CTL clone, indicating that the peptide is processed in tumor cells. These results indicate that the LAGE-1.A68 peptide can be used for antitumoral vaccination. We observed also that specific T cells could be detected in a blood sample with a high sensitivity by using an A68/LAGE-1 fluorescent multimer.


Subject(s)
Antigens, Neoplasm/immunology , Membrane Proteins/immunology , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Antigens, Surface , COS Cells , Chlorocebus aethiops , HLA Antigens/immunology , Humans , Melanoma/immunology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Peptides/genetics , Polymerase Chain Reaction , Transfection
8.
Int J Cancer ; 117(4): 596-604, 2005 Nov 20.
Article in English | MEDLINE | ID: mdl-15945101

ABSTRACT

The purpose of this phase 1/2 study was to evaluate toxicity, tumor evolution and immunologic response following administration of a fixed dose of a recombinant MAGE-3 protein by subcutaneous and intradermal routes in the absence of immunologic adjuvant. Thirty-two patients with detectable metastatic melanoma expressing gene MAGE-3 were included and 30 received at least one injection with a fixed dose of a ProtD-MAGE-3 fusion protein. The immunization schedule included 6 intradermal and subcutaneous injections at 3-week intervals. Afterward, patients without major tumor progression who required other treatments received additional vaccinations at increasing time intervals. The vaccine was generally well tolerated. Among the 26 patients who received at least 4 vaccinations, we observed 1 partial response and 4 mixed responses. For these 5 responding patients, time to progression varied from 3.5 to 51+ months. An anti-MAGE-3 CD4 T-lymphocyte response was detected in 1 out of the 5 responding patients. The majority of patients had no anti-MAGE-3 antibody response. The clinical and immunologic responses generated by the vaccine are rather limited. Nevertheless, given the potential antitumor efficacy and the very mild toxicity of vaccinations, further studies combining MAGE proteins and/or peptides with potent immunologic adjuvants are warranted, not only in metastatic melanoma, but also in the adjuvant setting.


Subject(s)
Antigens, Neoplasm/administration & dosage , Melanoma/drug therapy , Neoplasm Proteins/administration & dosage , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/therapeutic use , CD4-Positive T-Lymphocytes/immunology , Disease Progression , Female , Humans , Injections, Subcutaneous , Male , Melanoma/immunology , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Neoplasm Proteins/adverse effects , Neoplasm Proteins/therapeutic use , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Survival Analysis
9.
J Exp Med ; 201(2): 241-8, 2005 Jan 17.
Article in English | MEDLINE | ID: mdl-15657293

ABSTRACT

After vaccination of melanoma patients with MAGE antigens, we observed that even in the few patients showing tumor regression, the frequency of anti-vaccine T cells in the blood was often either undetectable or <10(-5) of CD8 T cells. This frequency being arguably too low for these cells to be sole effectors of rejection, we reexamined the contribution of T cells recognizing other tumor antigens. The presence of such antitumor T cells in melanoma patients has been widely reported. To begin assessing their contribution to vaccine-induced rejection, we evaluated their blood frequency in five vaccinated patients. The antitumor cytotoxic T lymphocyte (CTL) precursors ranged from 10(-4) to 3 x 10(-3), which is 10-10,000 times higher than the anti-vaccine CTL in the same patient. High frequencies were also observed before vaccination. In a patient showing nearly complete regression after vaccination with a MAGE-3 antigen, we observed a remarkably focused antitumoral response. A majority of CTL precursors (CTLp's) recognized antigens encoded by MAGE-C2, another cancer-germline gene. Others recognized gp100 antigens. CTLp's recognizing MAGE-C2 and gp100 antigens were already present before vaccination, but new clonotypes appeared afterwards. These results suggest that a spontaneous antitumor T cell response, which has become ineffective, can be reawakened by vaccination and contribute to tumor rejection. This notion is reinforced by the frequencies of anti-vaccine and antitumor CTLs observed inside metastases, as presented by Lurquin et al. (Lurquin, C., B. Lethe, V. Corbiere, I. Theate, N. van Baren, P.G. Coulie, and T. Boon. 2004. J. Exp. Med. 201:249-257).


Subject(s)
Antigens, Neoplasm/therapeutic use , Cancer Vaccines/therapeutic use , Melanoma/blood , Melanoma/therapy , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cell Count , Cytotoxicity, Immunologic , Humans , Melanoma/immunology , Membrane Glycoproteins/immunology , Neoplasm Proteins/immunology , Neoplasm Proteins/therapeutic use , T-Lymphocytes, Cytotoxic/pathology , Treatment Outcome , gp100 Melanoma Antigen
10.
J Exp Med ; 201(2): 249-57, 2005 Jan 17.
Article in English | MEDLINE | ID: mdl-15657294

ABSTRACT

Melanoma patients have high frequencies of T cells directed against antigens of their tumor. The frequency of these antitumor T cells in the blood is usually well above that of the anti-vaccine T cells observed after vaccination with tumor antigens. In a patient vaccinated with a MAGE-3 antigen presented by HLA-A1, we measured the frequencies of anti-vaccine and antitumor T cells in several metastases to evaluate their respective potential contribution to tumor rejection. The frequency of anti-MAGE-3.A1 T cells was 1.5 x 10(-5) of CD8 T cells in an invaded lymph node, sixfold higher than in the blood. An antitumor cytotoxic T lymphocyte (CTL) recognizing a MAGE-C2 antigen showed a much higher enrichment with a frequency of approximately 10%, 1,000 times higher than its blood frequency. Several other antitumor T clonotypes had frequencies >1%. Similar findings were made on a regressing cutaneous metastasis. Thus, antitumor T cells were approximately 10,000 times more frequent than anti-vaccine T cells inside metastases, representing the majority of T cells present there. This suggests that the anti-vaccine CTLs are not the effectors that kill the bulk of the tumor cells, but that their interaction with the tumor generates conditions enabling the stimulation of large numbers of antitumor CTLs that proceed to destroy the tumor cells. Naive T cells appear to be stimulated in the course of this process as new antitumor clonotypes arise after vaccination.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Melanoma/immunology , Neoplasm Metastasis/immunology , Neoplasm Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm/therapeutic use , Cancer Vaccines/therapeutic use , Humans , Neoplasm Proteins/therapeutic use
11.
J Immunol ; 171(9): 4898-904, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14568971

ABSTRACT

We have analyzed the T cell responses of HLA-A1 metastatic melanoma patients with detectable disease, following vaccination with a recombinant ALVAC virus, which bears short MAGE-1 and MAGE-3 sequences coding for antigenic peptides presented by HLA-A1. To evaluate the anti-MAGE CTL responses, we resorted to antigenic stimulation of blood lymphocytes under limiting dilution conditions, followed by tetramer analysis and cloning of the tetramer-positive cells. The clones were tested for their specific lytic ability and their TCR sequences were obtained. Four patients who showed tumor regression were analyzed, and an anti-MAGE-3.A1 CTL response was observed in three of these patients. Postvaccination frequencies of anti-MAGE-3.A1 CTL were 3 x 10(-6), 3 x 10(-3), and 3 x 10(-7) of the blood CD8 T cells, respectively. These three responses were monoclonal. No anti-MAGE-1.A1 CTL response was observed. These results indicate that, like peptide immunization, ALVAC immunization produces monoclonal responses. They also suggest that low-level antivaccine CTL responses can initiate a tumor regression process. Taken together, our analysis of anti-MAGE-3.A1 T cell responses following peptide or ALVAC vaccination shows a degree of correlation between CTL response and tumor regression, but firm conclusions will require larger numbers.


Subject(s)
Antigens, Neoplasm/immunology , Canarypox virus/immunology , Cancer Vaccines/immunology , Melanoma/immunology , Melanoma/prevention & control , Neoplasm Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/genetics , Canarypox virus/genetics , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Clone Cells , Female , Humans , Injections, Intradermal , Injections, Subcutaneous , Lymphatic Metastasis/immunology , Lymphatic Metastasis/prevention & control , Melanoma/secondary , Neoplasm Proteins/administration & dosage , Neoplasm Proteins/genetics , Polymerase Chain Reaction , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/blood , T-Lymphocytes, Cytotoxic/metabolism , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...