Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 97(12): 1655-63, 2007 Dec 17.
Article in English | MEDLINE | ID: mdl-18071348

ABSTRACT

D(b-/-)xbeta2 microglobulin (beta2m) null mice transgenic for a chimeric HLA-A2.1/D(b)-beta2m single chain (HHD mice) are an effective biological tool to evaluate the antitumour cytotoxic T-lymphocyte response of known major histocompatibility-restricted peptide tumour-associated antigens, and to screen for putative unknown novel peptides. We utilised HHD lymphocytes to identify immunodominant epitopes of colon carcinoma overexpressed genes. We screened with HHD-derived lymphocytes over 500 HLA-A2.1-restricted peptides derived from colon carcinoma overexpressed genes. This procedure culminated in the identification of seven immunogenic peptides, three of these were derived from the 'human 1-8D gene from interferon inducible gene' (1-8D). The 1-8D gene was shown to be overexpressed in fresh tumour samples. The three 1-8D peptides were both antigenic and immunogenic in the HHD mice. The peptides induce cytotoxic T lymphocytes that were able to kill a colon carcinoma cell line HCT/HHD, in vitro and retard its growth in vivo. One of the peptides shared by all the 1-8 gene family primed efficiently normal human cytotoxic T lymphocyte precursors. These results highlight the 1-8D gene and its homologues as putative immunodominant tumour-associated antigens of colon carcinoma.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Interferons/chemistry , Interferons/pharmacology , Membrane Proteins/genetics , Peptides/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Line, Tumor , Cross Reactions , Humans , Immunodominant Epitopes , Membrane Proteins/immunology , Mice , Mice, Transgenic
2.
Clin Exp Immunol ; 143(1): 139-49, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16367945

ABSTRACT

Due to the fact that many cellular proteins are extensively glycosylated, processing and presentation mechanisms are expected to produce a pool of major histocompatibility complex (MHC) class I-bound protein-derived peptides, part of which retain sugar moieties. The immunogenic properties of the presented glycosylated peptides in comparison to their non-glycosylated counterparts have not been determined clearly. We assessed the cellular immunogenicity of MUC1 (mucin)-derived peptides O-glycosylated with a Tn epitope (GalNAc) using HLA-A*0201 single chain (HHD)-transfected cell lines and transgenic mice. For part of the compounds Tn moiety did not interfere with the HLA-A*0201 binding. Moreover, part of the glycopeptides elicited effective cytotoxic responses, indicating recognition of the glycopeptide-HLA-A*0201 complex by the T cell receptor (TCR) and subsequent cytotoxic T lymphocyte (CTL) activation. The CTLs exhibited a substantial degree of cross-reactivity against target cells loaded with glycosylated and non-glycosylated forms of the same peptide. The studied (glyco)peptides showed cellular immunogenicity in both MUC1-HHD and HHD mice and induced effective lysis of (glyco)peptide-loaded target cells in CTL assays. However, the elicited CTLs did not induce selective lysis of human MUC1-expressing murine cell lines. Moreover, immunization with (glyco)peptide-loaded dendritic cells (DCs) did not induce significant immunotherapeutic effects. We conclude that Tn glycosylated MUC1-derived peptides can be presented by MHC class I molecules, and may be recognized by specific TCR molecules resulting in cytotoxic immune responses. However, the studied glycopeptides did not offer significant benefit as targets for cytotoxic immune response due apparently to (a) cross-reactivity of the elicited CTLs against the glycosylated and non-glycosylated forms of the same peptide and (b) low abundance of glycopeptides on tumour target cells.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/immunology , Carcinoma/therapy , HLA-A Antigens/immunology , Immunotherapy/methods , Mucin-1/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigen-Antibody Reactions , Antigens, Tumor-Associated, Carbohydrate/genetics , Carcinoma/immunology , Cell Line , Cytotoxicity Tests, Immunologic , Glycopeptides/immunology , HLA-A Antigens/genetics , Immunization , Mice , Mice, Transgenic , Mucin-1/genetics , Transfection/methods
3.
Br J Cancer ; 91(2): 398-407, 2004 Jul 19.
Article in English | MEDLINE | ID: mdl-15213716

ABSTRACT

Bladder carcinoma is the fourth most common cancer in men and the eighth most common cancer among women. Our study is aimed to characterise tumour-associated antigen peptides of transitional cell carcinoma of the bladder (TCC). A DNA micro-array-based differential display analysis of 10 000 genes was carried out, and MAGE-A8 gene expression was detected in the tumour, and not in the normal bladder. High occurrence of MAGE-A8 expression was observed in fresh tumour samples (17 out of 23) and TCC lines (four of eight). The MAGE-A8 protein sequence was screened for HLA-A2.1-binding motifs, six potential peptides were synthesised, and peptides binding to HLA-A2.1 were assured. Immunogenicity and antigenicity of the MAGE-A8 peptides were examined in the HHD system, murine class I MHC knockout mice, transgenic for HLA-A2.1. The MAGE-A8 peptide immunogenicity was examined in three modes of vaccination, delivered intranasally with cholera toxin, injected into the tail base with complete Freund's adjuvant (CFA), or presented directly as loaded onto cell surface HLA-A2.1 molecules. Two peptides, 8.1 and 8.3, induce CTL that kills the T24 TCC line in vitro, and prime human lymphocyte response of healthy donors. These results demonstrate the potential use of the MAGE-A8 peptides for specific immunotherapy of TCC.


Subject(s)
Antigens, Neoplasm/genetics , Carcinoma, Transitional Cell/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Oligopeptides/genetics , Urinary Bladder Neoplasms/genetics , Animals , Antigens, Neoplasm/immunology , Carcinoma, Transitional Cell/pathology , Cholera Toxin/administration & dosage , Cytotoxicity, Immunologic , Freund's Adjuvant , Gene Expression Profiling , HLA-A2 Antigen/immunology , Humans , Mice , Mice, Knockout , Mice, Transgenic , Neoplasm Proteins/immunology , Oligonucleotide Array Sequence Analysis , Oligopeptides/pharmacology , T-Lymphocytes/immunology , Urinary Bladder Neoplasms/pathology , Vaccination , beta 2-Microglobulin/genetics , beta 2-Microglobulin/physiology
4.
Br J Cancer ; 89(2): 314-9, 2003 Jul 21.
Article in English | MEDLINE | ID: mdl-12865923

ABSTRACT

Despite advances in the management of solid tumours, the development of metastases continues to be the most significant problem and cause of death for cancer patients. To define genetic determinants of pulmonary metastases, we have applied oligonucleotide microarrays to established murine models of highly metastatic D122 Lewis lung carcinoma and B16-F10.9 melanoma cell lines. These models are characterised by primary subcutaneous growth in C57BL/6J mice, a period of minimal residual disease and spontaneous pulmonary metastases. Microarray analysis defined seven genes, namely - arginase, brain natriuretic peptide (BNP), interleukin-1 alpha (IL-1 alpha), plasminogen activator inhibitor-2 (PAI-2), surfactant protein C (SP-C), uteroglobin (UG) and wnt-1-induced secreted protein-1 (WISP-1), which were consistently elevated in pulmonary metastases compared to the primary tumour of both D122 and B16-F10.9 models. Previous studies demonstrated that two of these seven genes, IL-1 alpha and PAI-2, are involved in the metastatic process. The results obtained by the microarrays were confirmed by real-time quantitative PCR, for three chosen genes - PAI-2, WISP-1 and UG. Our approach aimed to identify genes essential for the metastatic process in general and for pulmonary metastases specifically. Further research should address the precise role of these genes in the metastasising process to the lungs and test if they could be used as targets for future therapies.


Subject(s)
Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Gene Expression Regulation, Neoplastic , Growth Substances/biosynthesis , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Melanoma/genetics , Melanoma/pathology , Neoplasm Metastasis/genetics , Oncogene Proteins/biosynthesis , Animals , CCN Intercellular Signaling Proteins , Carrier Proteins/biosynthesis , Female , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Plasminogen Activator Inhibitor 2/biosynthesis , Polymerase Chain Reaction , Proto-Oncogene Proteins , Tumor Cells, Cultured , Uteroglobin/biosynthesis
5.
Clin Cancer Res ; 7(3 Suppl): 818s-821s, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11300478

ABSTRACT

The increasing ability to augment antitumor immunity in model systems has led to increased numbers of clinical trials. However, progress in detecting immune responses by patients against autologous tumors has been slow. Although a considerable number of tumor antigens, as well as peptides derived from them, and the MHC determinants together with which they are presented have been identified for melanoma, this is not so for the majority of solid tumors. Furthermore, tumor cells themselves are poor stimulators of immunity. Thus, approaches that do not depend upon defined antigens or using tumor cells as stimulators would be desirable. To attempt to measure immune responses in these situations, we tested whether total peptides, prepared from autologous tumor tissue, stimulated cytokine release by T cells. Peripheral blood mononuclear cells (PBMCs) were mixed with antigen-presenting cells (APCs), pulsed with tumor peptides, and tested in the ELISPOT assay for IFN-gamma secretion. Few spots were obtained when PBMCs were cultured with unpulsed APCs or in wells with peptide-pulsed APC alone. In contrast, a strong response was seen when PBMCs were cultured with APCs that had been pulsed with autologous total tumor peptides. This system should help to identify those immunotherapeutic approaches that induce responses against tumor cells in vivo. Because different cytokine profiles are associated with distinct arms of the immune response, testing in the ELISPOT assay may also help us understand the mechanisms responsible.


Subject(s)
Lymphocyte Activation , Neoplasms/metabolism , Peptides/chemistry , T-Lymphocytes/metabolism , Antigen-Presenting Cells/metabolism , Cells, Cultured , Colonic Neoplasms/metabolism , Cytokines/metabolism , Dose-Response Relationship, Drug , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/metabolism , Peptides/metabolism
6.
Immunol Lett ; 74(1): 27-34, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10996624

ABSTRACT

Cytotoxic T Lymphocytes (CTLs) recognize Major Histocompatibility Complex (MHC) class I in complex with peptides. Peptides derived from Tumor Associated Antigens (TAAs) are therefore targets for tumor rejection. A number of TAAs were identified in the last decade from human and murine tumors. Here we summarize the methods for TAA and peptide identification, the nature of TAA peptides and the making of antitumor vaccines.


Subject(s)
Cancer Vaccines/therapeutic use , Immunotherapy, Active , Neoplasms/immunology , Neoplasms/therapy , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use , Animals , Antibodies, Neoplasm/immunology , Antibody Specificity/immunology , Antigens, Neoplasm/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Cancer Vaccines/immunology , Humans
7.
J Immunol Methods ; 241(1-2): 61-8, 2000 Jul 31.
Article in English | MEDLINE | ID: mdl-10915849

ABSTRACT

Several issues remain to be resolved before the efficacy of various approaches to elicit anti-tumor immunity in patients can be evaluated. First, in vitro assays able to detect responses by T cells primed in vivo are needed. Second, a source of tumor antigen to stimulate patients' lymphocytes in vitro is required. The ELISPOT assay is attractive, because it can be performed with a small numbers of cells and requires only short-term culture in vitro. A source of tumor antigen is more problematic, since for most tumors, tumor-associated antigens (TAA) have not been identified and/or cloned. In this report we demonstrate that autologous antigen-presenting cells (APC) pulsed with total tumor peptides from autologous tumor tissue can stimulate IFNgamma release by patients' lymphocytes in the ELISPOT assay. Thus, this approach should be considered for monitoring immune responses in clinical immunotherapy trials.


Subject(s)
Antigens, Neoplasm/immunology , Enzyme-Linked Immunosorbent Assay/methods , Interferon-gamma/metabolism , Lymphocytes/immunology , Neoplasms/immunology , Peptides/immunology , Antigen Presentation , Antigen-Presenting Cells , Dose-Response Relationship, Drug , Humans , Immunotherapy , Neoplasms/therapy
8.
J Immunother ; 23(3): 344-52, 2000.
Article in English | MEDLINE | ID: mdl-10838663

ABSTRACT

Peptide vaccination of homozygous mice against syngeneic tumors using single major histocompatibility complex (MHC) class I-restricted cytotoxic T lymphocyte (CTL) epitopes elicits effective immune responses against metastatic growth. So far, single-peptide vaccination of patients against their autologous tumors seems to elicit less satisfactory results. In this study, the authors tried to determine whether effective anti-metastatic immunity requires the presentation of peptides restricted by the two parental class I major histocompatibility complex alleles in heterozygous hosts. The immune response against the H-2b-derived 3LL Lewis lung carcinoma was evaluated in heterozygous recombinant congenic F1 mice (Kk x K(b)) and (Kd x K(b)). Vaccination of such heterozygous animals with dendritic cells expressing the two parental H-2K alleles, pulsed with total tumor extract, elicited a potent anti-metastatic response. A comparable response was obtained after vaccination with tumor cells genetically modified to express the two class I alleles. In contrast, vaccination of the heterozygous mice with dendritic cells expressing only one of the parental F1 H-2K alleles or with tumors expressing only one H-2K allele failed to elicit effective immunity against tumor metastasis in recombinant congenic F1 mice. It appears, therefore, that to achieve effective anti-metastatic immunotherapy in heterozygous organisms, presentation of cytotoxic T lymphocyte epitopes restricted by the two parental class I alleles is required.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Epitopes/immunology , Histocompatibility Antigens Class I/immunology , Lung Neoplasms/therapy , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , Alleles , Animals , Cytotoxicity Tests, Immunologic , Flow Cytometry , Genes, MHC Class I , Heterozygote , Histocompatibility Antigens Class I/genetics , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Mice , Mice, Congenic , Neoplasm Metastasis/prevention & control , Tumor Cells, Cultured
9.
Int J Cancer ; 85(3): 391-7, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10652432

ABSTRACT

The MUC1 protein was found to be up-regulated in a spectrum of malignant tumors. T-cell responses to the MUC1 extracellular tandem repeat array (TRA) were observed in murine models as well as in breast-carcinoma patients. In the present study, we evaluated the anti-tumor potential of HLA-A2.1-motif-selected peptides from non-TRA domains of the molecule. Peptide immunogenicity was examined in the Db-/- x beta2 microglobulin (beta2m) null mice transgenic for a modified HLA-A2.1/Db-beta2 microglobulin single chain (HHD mice). Our results show the existence of 3 novel HLA-A2.1-restricted MUC1-derived cytotoxic T-lymphocyte (CTL) epitopes. These peptides are processed and presented by the HHD-transfected breast-tumor cell line MDA-MB-157. Moreover, CTL induced by these 3 peptides show higher lysis of target cells pulsed with breast-carcinoma-derived peptides than of targets pulsed with normal breast-tissue-derived peptides. These data suggest an important role for non-TRA MUC1-derived peptides as inducers of a MHC-restricted CTL reaction to a breast-carcinoma cell line and patient-derived tumor extracts.


Subject(s)
Breast Neoplasms/immunology , Cancer Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , Mucin-1/immunology , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology , beta 2-Microglobulin/immunology , Animals , Antigen Presentation , Breast Neoplasms/metabolism , Cancer Vaccines/chemistry , Cancer Vaccines/genetics , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/chemistry , Female , Fluorescence , Gene Expression Regulation, Neoplastic , H-2 Antigens/genetics , H-2 Antigens/immunology , Histocompatibility Antigen H-2D , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mucin-1/chemistry , Mucin-1/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptides/immunology , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Tumor Cells, Cultured , beta 2-Microglobulin/chemistry , beta 2-Microglobulin/genetics
10.
Int J Cancer ; 85(2): 236-42, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10629083

ABSTRACT

CTLs specific for tumor antigens play a major role in immunity against cancer. Improved binding affinity of putative TAA peptides could enhance the in vivo immunogenicity of these self-altered self- tumor antigens. We examined here the efficacy of tumor vaccines composed of an altered peptide ligand of MUT-1, designated MUT-D, which exhibited significantly higher class-I allele K(b) binding affinity than its native counterpart MUT-1. The peptide was loaded on antigen presenting cells composed of the C57BL/6-syngeneic fibroblast cell line BLK.CL4. These cells were treated with proteasome inhibitor in order to shut off the degradation of proteins and the subsequent loading of endogenous peptides onto MHC class-I molecules, thus allowing for the pulsing of these cells with the modified peptide MUT-D. Proteasome-inhibited and modified peptide-loaded fibroblasts induced a peptide-specific CTL that significantly delayed primary tumor progression and protected the pre-immunized mice against the development of lung metastasis following the surgical removal of the primary tumor. Genetic modification of the fibroblasts to express the immunostimulatory cytokine IL-2 did not improve the APC function of the modified cells, nor did it result in augmentation of the potency of the vaccine. Our results suggest that the proteasome-inhibited fibroblasts pulsed with modified, high binder tumor-associated antigen peptide are good antigen-presenting cells and represent an effective form of tumor vaccine.


Subject(s)
Cancer Vaccines/immunology , Fibroblasts/immunology , Lung Neoplasms/therapy , Oligopeptides/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Neoplasm/immunology , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Epitopes, T-Lymphocyte/immunology , Fibroblasts/enzymology , Immunotherapy , Interleukin-2/biosynthesis , Interleukin-2/genetics , Leupeptins/pharmacology , Lung Neoplasms/immunology , Male , Mice , Mice, Inbred C57BL , Multienzyme Complexes/metabolism , Neoplasm Metastasis/prevention & control , Neoplasm Transplantation , Proteasome Endopeptidase Complex
11.
J Immunother ; 23(6): 622-30, 2000.
Article in English | MEDLINE | ID: mdl-11186150

ABSTRACT

The development of a cell-free synthetic vaccine to induce an effective cytotoxic T lymphocyte response is an important challenge in T-cell--mediated immunity. Because standard vaccinations with nominal epitopes were found to be only partially effective in vivo, the authors suggest an alternative strategy: the delivery of epitopes directly to the cell cytosol in a proteasome bypass mechanism of processing. Two model peptides, the presentation level on the cell surface of which can be directly assessed, were conjugated via a cross-linker to an internalization peptide derived from an antennapedia homeobox protein. The linker was designed to undergo spontaneous hydrolysis, after which the epitope is subsequently released. The conjugates were shown to enter RMA and P815 cells, where the epitopes were released mainly in cytosol and endogenously loaded on the major histocompatibility complex class I molecules to be presented on the cell surface. Concomitant inhibition of proteasome activity by MG132 significantly increased the presentation level of both model peptides, indicating proteasome-independent processing. This phenomenon was exploited to enhance the immunogenicity of the conjugates. Conjugates were emulsified with MG132 in incomplete Freund's adjuvant and injected into mouse footpads. Analysis of the draining lymph nodes indicated an increase in the percentage of both CD4+ and CD8+ lymphocytes. In vitro cytolytic assays implied significant, albeit moderate, priming only when the proteasome inhibitor was administered with the conjugate. This approach may be useful for the development of efficient synthetic cell-free vaccines.


Subject(s)
Antigen Presentation , Cysteine Endopeptidases/metabolism , Epitopes/immunology , Histocompatibility Antigens Class I/immunology , Lipids , Multienzyme Complexes/metabolism , Nuclear Proteins , Transcription Factors , Vaccines, Subunit/pharmacology , Animals , Antennapedia Homeodomain Protein , Antigen Presentation/drug effects , Biological Transport , Cell Line , Cell Membrane/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Cytosol/drug effects , Cytosol/immunology , Cytotoxicity Tests, Immunologic , Epitopes/metabolism , Female , Freund's Adjuvant/pharmacology , Histocompatibility Antigens Class I/metabolism , Homeodomain Proteins/metabolism , Leupeptins/pharmacology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Count , Mice , Mice, Inbred DBA , Multienzyme Complexes/antagonists & inhibitors , Oligopeptides/immunology , Oligopeptides/metabolism , Proteasome Endopeptidase Complex , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Subunit/metabolism
12.
Immunol Lett ; 70(1): 21-8, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10541048

ABSTRACT

CTL induction by immunization with synthetic peptide epitopes has been shown to inhibit tumor growth and its metastatic spread. Ex vivo pulsing of peptides on MHC class I-bearing cells such as RMA-S cells or professional APCs elicits an effective CTL response. Since the stability of the MHC-peptide complex is strongly correlated with the overall immunogenecity, we compared the effect of immunization with low affinity, high affinity, and irreversibly bound MHC peptides in the context of immunotherapy of metastasis. MUT1, a tumor-associated antigen peptide that was isolated from 3LL Lewis lung carcinoma, is a low H-2Kb binder. MUT1 was modified into a high binder by changing positions 3, 5, and 8 to the favorable anchor residues. In addition, we introduced a photo-active chemical moiety, which can bind irreversibly to MHC upon illumination. These peptides, loaded onto RMA-S, were used to immunize mice against the 3LL tumor. Vaccination via the covalent conjugation of the low binder peptide was found to increase the CTL response measured against MUT1 loaded cells and against H-2Kb transfected D122 cells relative to the native MUT1 peptide. However, the photo cross-linking of the high affinity peptide to the MHC did not significantly improve the induction of specific CTL. The level of CTL activity was elevated to the same extent by either cross-linking the peptide to the MHC or by modifying it into a high-binder peptide. The protective capacity of all the peptide-based vaccines against D122 metastatic spread to the lungs was found to be comparable. These results indicate that augmentation of the affinity of a TAA peptide to the RMA-S surface MHC molecules, by conversion to a high-affinity mimotope or by photo-conjugation, can significantly enhance the immune response. There seems to be, however, a ceiling beyond which increase in the peptide-binding affinity does not lead to a corresponding enhancement of the overall immunogenicity of the peptide.


Subject(s)
Cancer Vaccines/immunology , H-2 Antigens/immunology , Peptides/immunology , Vaccines, Synthetic/immunology , Animals , Cancer Vaccines/chemistry , Cell Transplantation , Epitopes/immunology , Mice , Mice, Inbred C57BL , Molecular Mimicry , Peptides/chemistry , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured , Vaccines, Synthetic/chemistry
13.
Eur J Immunol ; 29(10): 3295-301, 1999 10.
Article in English | MEDLINE | ID: mdl-10540341

ABSTRACT

Epitope spreading is a process whereby epitopes distinct from and non-cross-reactive with an inducing epitope become targets of an evolving immune response. This phenomenon has been associated most notably with the progression of naturally occurring or experimentally induced chronic autoimmune diseases. We have investigated the potential occurrence of epitope spreading in the context of antitumor cytotoxic T cell (CTL) responses using chicken ovalbumin (OVA) as a model antigen. Our results indicate that following rejection of OVA-expressing EG.7 tumor cells effectuated by a CTL response which is induced against the MHC class I-restricted immunodominant epitope OVA257-264, there occurs intramolecular diversification of the CTL response to two additional OVA-derived epitopes, OVA176-183 and OVA55-62, as well as intermolecular spreading to other endogenous tumor-derived determinants. It seems that CTL-mediated tumor cell destruction in vivo favors cross-presentation of additional epitopes with the consequent activation of additional tumor-reactive lymphocytes. The process of epitope spreading in that context has obvious important implications for the design of antigen-specific antitumor immunotherapies.


Subject(s)
Cancer Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , Graft Rejection/immunology , H-2 Antigens/immunology , Immunodominant Epitopes/immunology , Neoplasms, Experimental/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cancer Vaccines/genetics , Chickens , Female , Mice , Mice, Inbred C57BL , Ovalbumin/immunology
14.
J Interferon Cytokine Res ; 19(12): 1391-401, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10638708

ABSTRACT

Development of cytokine gene-modified autologous tumor vaccines must take into account the strictly paracrine physiology of cytokines whose expression at the tumor microenvironment is important for the successful induction of tumor-specific immunity. In this study, we investigated the efficacy of a tumor vaccine composed of inactivated autologous cells transfected with two plasmid vectors encoding a mutant membrane-bound murine granulocyte-macrophage colony-stimulating factor (MuGM-CSF) and murine interferon-gamma (MuIFN-gamma). Expression of both cytokines as cell surface ligands on the highly metastatic D122 clone of Lewis lung carcinoma led to abrogation of their tumorigenicity and metastatic phenotype. More importantly, vaccination with irradiated tumor cells expressing the membrane-bound GM-CSF and IFN-gamma induced a cytotoxic T lymphocyte (CTL) response that protected syngeneic mice against a subsequent challenge with D122 cells as a primary tumor in preimmunized mice as well as against lung metastasis developing after surgical removal of the primary tumor in naive mice. Autologous cells expressing the membrane-bound GM-CSF and IFN-gamma exhibited comparable efficacy as an antimetastatic vaccine to a vaccine composed of transfectants expressing wild-type secreted cytokine molecules. These results indicate that membrane-bound cytokines can cause enhanced immunogenicity when transfected into tumor cells for the induction of antitumor immunity.


Subject(s)
Cancer Vaccines/pharmacology , Cytokines/genetics , Animals , Base Sequence , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Membrane/immunology , DNA Primers/genetics , Female , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Interferon-gamma/genetics , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Male , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Recombinant Proteins , T-Lymphocytes, Cytotoxic/immunology , Transfection , Transplantation, Autologous
15.
Clin Exp Metastasis ; 16(7): 623-32, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9932609

ABSTRACT

Interleukin 12 (IL-12) is a disulfide-linked heterodimer molecule produced predominantly by professional antigen presenting cells. It promotes the induction of sundry biological effects with significant relevance to antitumor immunity, such as enhancing a T(H)1 helper response, an in vivo antiangiogenic effect, induction of adhesion molecules that assist in lymphocyte homing to sites of tumor growth, and a direct stimulatory effect on both T-cells and NK cells. We tested the efficacy of an antimetastatic vaccine composed of autologous murine D122 cells transfected with both subunits of IL-12 cDNA to express biologically-active IL-12 molecule. Expression of IL-12 by D122 cells significantly reduced their tumorigenicity and metastatic potential in immunocompetent syngeneic hosts. Furthermore, vaccination of mice with 2 x 10(6) irradiated IL-12-transfected D122 cells engendered a protective CTL response which rejected a subsequent challenge with parental D122 cells and eradicated lung micrometastasis in animals whose primary tumors have been surgically removed. The antitumor effects of IL-12 were mediated primarily by its ability to induce gammaIFN expression in vivo. CD8+ T-cells as well as NK cells were crucial in the execution of the antitumor effects of IL-12. These results suggest that autologous tumor cells expressing IL-12 by gene transfer are a potent antitumor vaccine able to induce a systemic immune response against poorly immunogenic and spontaneously metastatic tumors.


Subject(s)
Cancer Vaccines/therapeutic use , Carcinoma, Lewis Lung/prevention & control , Genetic Therapy/methods , Interferon-gamma/genetics , Interleukin-12/genetics , Neoplasm Metastasis/prevention & control , Animals , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/secondary , Cell Division , Cells, Cultured , Immunotherapy/methods , Interleukin-12/immunology , Killer Cells, Natural/immunology , Mice , Plasmids/genetics , Polymerase Chain Reaction , Th1 Cells/drug effects , Transfection , Tumor Cells, Cultured
16.
Nature ; 390(6656): 180-4, 1997 Nov 13.
Article in English | MEDLINE | ID: mdl-9367156

ABSTRACT

DAP kinase is a new type of calcium/calmodulin-dependent enzyme that phosphorylates serine/threonine residues on proteins. Its structure contains ankyrin repeats and the 'death' domain, and it is associated with the cell cytoskeleton. The gene encoding DAP kinase was initially isolated as a positive mediator of apoptosis induced by interferon-gamma, by using a strategy of functional cloning. We have now tested whether this gene has tumour-suppressive activity. We found that lung carcinoma clones, characterized by their highly aggressive metastatic behaviour and originating from two independent murine lung tumours, did not express DAP kinase, in contrast to their low-metastatic counterparts. Restoration of DAP kinase to physiological levels in high-metastatic Lewis carcinoma cells suppressed their ability to form lung metastases after intravenous injection into syngeneic mice, and delayed local tumour growth in a foreign 'microenvironment' Conversely, in vivo selection of rare lung lesions following injection into syngeneic mice of low-metastatic Lewis carcinoma cells or of DAP kinase transfectants, was associated with loss of DAP kinase expression. In situ TUNEL staining of tumour sections revealed that DAP kinase expression from the transgene raised the incidence of apoptosis in vivo. DAP-kinase transfectants also showed increased sensitivity in vitro to apoptotic stimuli, of the sort encountered by metastasizing cells at different stages of malignancy. We propose that loss of DAP kinase expression provides a unique mechanism that links suppression of apoptosis to metastasis.


Subject(s)
Apoptosis/genetics , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Genes, Tumor Suppressor , Neoplasm Metastasis/genetics , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Death-Associated Protein Kinases , Female , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
17.
Oncogene ; 15(13): 1545-54, 1997 Sep 25.
Article in English | MEDLINE | ID: mdl-9380406

ABSTRACT

Functional PDGFalpha receptors are selectively expressed on highly lung-metastasizing clones of the 3LL Lewis lung carcinoma, but not on low-mestastatic clones. The highly metastatic clones are also growth induced in vitro by PDGF and lung conditioned medium. To investigate whether modification of PDGFalpha receptor expression or function can affect metastatic capability, we transfected cells of a low-metastatic 3LL clone with a full length PDGFalpha receptor gene and cells of a highly-metastatic clone with a truncated kinase domain PDGFalpha receptor gene. Introduction of the full length PDGFalpha receptor conferred upon low-metastatic cells the ability to grow in vitro in the presence of PDGF-AA and to colonize the lung in experimental and spontaneous metastases assays. Conversely, introduction of a truncated version of the PDGFalpha receptor into highly metastatic cells reduced their metastatic load to control levels. Accordingly, their responses to PDGF-AA, including growth stimulation and receptor autophosphorylation, were reduced. These results demonstrate that PDGFalpha receptor expression and function can control the capacity of tumor cells to generate metastases in the lung. The response of this receptor to lung-derived PDGF-like factors may define a paracrine mode of metastatic cell growth in the target organ.


Subject(s)
Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Neoplasm Metastasis , Receptors, Platelet-Derived Growth Factor/metabolism , Animals , Cell Division/drug effects , Mice , Mice, Inbred C57BL , Phosphorylation , Receptor, Platelet-Derived Growth Factor alpha , Receptors, Platelet-Derived Growth Factor/genetics , Transfection , Tumor Cells, Cultured
18.
Oncogene ; 15(5): 569-77, 1997 Jul 31.
Article in English | MEDLINE | ID: mdl-9247310

ABSTRACT

Interleukin-6 (IL-6) inhibits the growth of melanocytes and of early stage melanoma cells, but not that of advanced melanoma cells. The in vitro IL-6 response can be restored in the highly metastatic melanoma B16-F10.9 by addition of recombinant soluble IL-6 receptor alpha-chain (sIL-6R). The F10.9 cells then undergo irreversible growth-arrest and show increased adherence with changes from epithelioid to spindleoid morphology. The sIL-6R is required for IL-6 to induce a sustained activation of the various Stat transcription factors which bind to specific IL-6 inducible enhancers. The sIL-6R and IL-6 combination causes an increase in the level of the anti-oncogenic transcription factor IRF-1 protein and DNA-binding, which remain elevated for 24 h. The promoter activity of the anti-oncogenic p21/Waf-1/Cip-1 gene is induced and accumulation of the p21 protein is observed. These results illustrate the potent agonist activity of sIL-6R on molecular pathways which could mediate the growth-arrest and differentiation of the metastatic melanoma cells. Previously observed antimetastatic effects of IL-6 therapy in mice bearing F10.9 tumors may be at least partly due to direct growth inhibition and differentiation elicited by sIL-6R present in biological fluids.


Subject(s)
Antigens, CD/metabolism , Interleukin-6/pharmacology , Melanoma/pathology , Receptors, Interleukin/metabolism , Animals , Antigens, CD/chemistry , Antigens, CD/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Division/drug effects , Cell Division/genetics , Cricetinae , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/drug effects , Cyclins/genetics , Cyclins/metabolism , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Interferon Regulatory Factor-1 , Melanoma/drug therapy , Melanoma/metabolism , Mice , Phosphoproteins/drug effects , Phosphoproteins/metabolism , Receptors, Interleukin/chemistry , Receptors, Interleukin/genetics , Receptors, Interleukin-6 , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , STAT1 Transcription Factor , Solubility , Trans-Activators/drug effects , Trans-Activators/genetics , Trans-Activators/metabolism , Tumor Cells, Cultured
19.
Oncogene ; 14(2): 145-55, 1997 Jan 16.
Article in English | MEDLINE | ID: mdl-9010216

ABSTRACT

Cellular functions of tumor suppressor proteins can be mediated by protein-protein interactions. Using p53 as a probe to screen an expression library, a cDNA encoding a 250 kDa protein was isolated. Recombinant forms of this protein, designated PACT, bind to wild type p53 while two different mutations abolish this interaction. PACT protein can also interfere with p53 specific DNA binding. PACT contains a serine/arginine (SR) rich region and a C' terminal lysine rich domain. The 250 kDa PACT protein can be precipitated from cell lysates by a method specific for SR proteins. snRNPs can be co-immunoprecipitated from cells with anti-PACT antibodies. These antibodies stain cell nuclei in a speckled pattern reminiscent of the distribution of known splicing factors. Recently, RBQ1, a truncated human homologue of PACT was identified by virtue of Rb binding. We show that RBQ1 is truncated as a result of a possible mutational event. PACT can interact with both cellular Rb and p53.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA/metabolism , RNA-Binding Proteins , Testis/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Alternative Splicing , Animals , Base Sequence , Cloning, Molecular , Humans , Male , Mice , Molecular Sequence Data , Mutation , Retinoblastoma Protein/metabolism , Ribonucleoproteins, Small Nuclear/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...