Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cell Biochem ; 217(1-2): 21-33, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11269662

ABSTRACT

We investigated the ubiquitination and degradation of a tumor antigen, the HER-2/neu (HER-2) protooncogene product which is overexpressed in epithelial cancers. HER-2 degradation was investigated in the ovarian tumor line, SKOV3.A2, that constitutively overexpressed long-life HER-2. We used as agonist geldanamycin (GA), which initiated downmodulation of HER-2 from the cell surface. HER-2 was polyubiquitinated and degraded faster in the presence than in the absence of GA. GA did not decrease HLA-A2 expression. Presentation of the immunodominant cytotoxic T lymphocyte (CTL) epitope, E75 (369-377) from SKOV.A2 was inhibited by proteasome inhibitors, such as LLnL but was enhanced by cysteine protease inhibitors such as E64, indicating that both the proteasome and cysteine proteases are involved in epitope formation but have different effects. Enhanced tumor recognition was not an immediate or early effect of GA treatment, but was evident after 20 h of GA treatment. In contrast, 20 h GA treatment did not increase tumor sensitivity to LAK cell lysis. Twenty hour GA-treated SKOV3.A2 cells expressed an unstable HER-2 protein synthesized in the presence of GA, of faster electrophoretic mobility than control HER-2. This suggested that the newly synthesized HER-2 in the presence of GA was the main source of epitopes recognized by CTL. Twenty hour GA-treated SKOV3.A2 cells were better inducers of CTL activity directed to a number of HER-2 CTL epitopes, in peripheral blood mononuclear cells compared with control untreated SKOV3.A2 cells. Thus, induction of HER-2 protein instability enhanced the sensitivity of tumor for CTL lysis. Increased HER-2 CTL epitopes presentation may have implications for overcoming the poor immuno-genicity of human tumors, and design of epitope precursors for cancer vaccination.


Subject(s)
Ovarian Neoplasms/immunology , Receptor, ErbB-2/metabolism , T-Lymphocytes, Cytotoxic/immunology , Antigen Presentation , Benzoquinones , Cysteine Endopeptidases/metabolism , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/immunology , Female , HLA-A2 Antigen/metabolism , Humans , Immunodominant Epitopes/immunology , Lactams, Macrocyclic , Multienzyme Complexes/metabolism , Ovarian Neoplasms/metabolism , Proteasome Endopeptidase Complex , Quinones/pharmacology , Receptor, ErbB-2/immunology , Tumor Cells, Cultured , Ubiquitins/metabolism
2.
J Interferon Cytokine Res ; 20(4): 391-401, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10805374

ABSTRACT

CXC chemokines play an important role in recruitment of T cells to the site of activation and regulation of angiogenesis. CXC chemokines are secreted by T cells stimulated with cytokines or by established cytotoxic T lymphocyte (CTL) lines at recognition of conventional antigen (Ag), but the activation requirements and the relationship of interferon-gamma (IFN-gamma) inducible protein (IP-10) secretion with IFN-gamma induction in lymphocytes are still unclear. We studied the induction of IP-10 from nonadherent peripheral blood mononuclear cells (PBMC) by IFN-gamma, interleukin-12 (IL-12), and the HER-2 peptide E75, which forms a CTL-defined antigen. We found that IFN-gamma alone was a weak inducer of IP-10 in these cells, whereas IL-12 was a significantly stronger inducer of IP-10. In the presence of IL-12, the tumor peptide E75 (HER-2, 369-377) was a stronger inducer of IP-10 than was IL-12 alone. E75 and its variants mutated at position 5 could also induce IP-10 in the absence of exogenous IL-12 or IFN-gamma. IP-10 induction by E75 required HLA-A2 presentation and B7-CD28 interactions and was partially inhibited by blocking of CD40-CD40L interactions. These results indicate that presentation of tumor peptides to peripheral T cells can induce a fast chemokine response, which in its early phase may be higher than the IFN-gamma response. This shows that the IP-10 response was independent of any early-phase IFN-gamma response in peripheral T cells. This may be important for understanding the regulation of the balance between chemoattractant chemokines (CC) and CXC chemokines by tumor Ag and may have implications for understanding the mechanisms of polarization of T cells and conditioning of antigen-presenting cells (APC) by tumor antigens.


Subject(s)
Breast Neoplasms/immunology , Chemokines, CXC/metabolism , Leukocytes, Mononuclear/metabolism , Peptide Fragments/physiology , Receptor, ErbB-2/physiology , CD40 Ligand , Cell Adhesion/immunology , Chemokine CXCL10 , Chemokines, CXC/biosynthesis , Chemokines, CXC/blood , HLA-A2 Antigen/metabolism , Humans , Interferon-gamma/pharmacology , Interleukin-12/pharmacology , Kinetics , Membrane Glycoproteins/physiology , Plastics
3.
Oncol Rep ; 7(3): 455-66, 2000.
Article in English | MEDLINE | ID: mdl-10767352

ABSTRACT

We investigated the ability of HER-2 peptide E75, which maps an immunodominant CTL epitope for ovarian and breast tumor-associated lymphocytes (TAL), to activate effector functions in freshly isolated CD8+ cells from healthy individuals. IFN-gamma was rapidly induced by E75 within 20-24 h, in five of six healthy donors, in the presence of IL-12 and was detectable as early as 6 h. The IFN-gamma levels were Ag-concentration dependent. Similar results were obtained with peptides mapping CTL epitopes from two other tumor Ag: folate binding protein (FBP) and amino-enhancer of split of Notch (AES). IFN-gamma was also detected, from freshly isolated, unstimulated PBMC in response to HLA-A2 matched tumors + IL-12 but not of IL-12 alone. The major source of IFN-gamma were CD45RO+ CD8+ cells. Induction of IFN-gamma and IL-2 from CD8+ cells and of IL-12 from dendritic cells (DC) by CD8+ cells reactive with E75 mirrored their induction by the influenza matrix peptide (M1: 58-66) in the same individual. Responses to M1 are used to define the presence of activated memory cells in healthy individuals. Compared to M1 responses E75 recognition induced 2-4-fold lower levels of IL-12 from the same APC and IFN-gamma and IL-2 from the same CD8+ cells. At lower Ag concentrations the endogenous IL-12 induced by E75-reactive CD8+ cells did not reach the threshold required to co-stimulate for IFN-gamma. alphaB7.1 synergized with E75 in increasing the overall levels of IL-2 induced within 24 h. The presence of tumor Ag-reactive activated CD8+ cells in healthy individuals may improve our understanding of the mechanisms of immunosurveillance and regulation of immune responses by tumors.


Subject(s)
Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Receptor, ErbB-2/immunology , Breast Neoplasms/blood , CD8-Positive T-Lymphocytes/drug effects , Cells, Cultured , Female , Histocompatibility Antigens Class I/blood , Histocompatibility Testing , Humans , Interferon-gamma/biosynthesis , Ovarian Neoplasms , Peptide Fragments/immunology , Peptide Fragments/pharmacology , Receptor, ErbB-2/chemistry , Reference Values , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
4.
Anticancer Res ; 19(4B): 2907-16, 1999.
Article in English | MEDLINE | ID: mdl-10652572

ABSTRACT

Tumor associated lymphocytes (TAL) isolated from malignant ascites cultured in media containing interleukin-2 show antitumor responses. These antitumor responses are mediated by cytotoxic T lymphocytes (CTL) which recognize antigen in the context of MHC molecules using T cell receptors. CD8+ CTL recognize peptide epitopes processed from cellular proteins in the context of MHC class I molecules. These peptides have a restricted length of 8-11 amino acids. The folate binding protein (FBP) is overexpressed in over 90% of ovarian and 20-50% of breast cancers. We recently found that FBP is the source of antigenic peptides recognized by a number of these CTL-TAL. This indicated that FBP peptides are antigenic in vivo for ovarian and breast CTL-TAL. To define FBP immunogenicity, a peptide defining the epitope E39 (FBP, 191-199) was presented by PMBC derived dendritic cells (DC) from healthy donors isolated by the CD14 method to ovarian and breast CTL-TAL. Stimulation of ovarian and breast CTL-TAL by E39 pulsed DC (DC-E39), in the presence of IL-2, rapidly enhanced or induced E39 specific CTL activity. This E39-responder population consisted of cells expressing TCR V beta 9, V beta 13, and V beta 17 families, based on the increase in the percentages of these families in DC-E39 versus DC-NP stimulated TAL. Characterization of immunogenic tumor antigens and of cytokine requirements for induction of functional antitumor effectors may be important for future cancer vaccine developments.


Subject(s)
Breast Neoplasms/immunology , Carrier Proteins/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Ovarian Neoplasms/immunology , Peptide Fragments/metabolism , Receptors, Cell Surface , T-Lymphocytes, Cytotoxic/immunology , Carrier Proteins/chemistry , Cells, Cultured , Female , Folate Receptors, GPI-Anchored , Humans , Immunotherapy, Adoptive , Lymphocyte Activation , Receptors, Antigen, T-Cell, alpha-beta/immunology
5.
J Interferon Cytokine Res ; 18(7): 529-36, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9712369

ABSTRACT

In the present study, we isolated tumor-infiltrating lymphocytes (TIL) from 21 primary solid tumors and tumor-associated lymphocytes (TAL) from 9 malignant effusions, respectively, of breast cancer patients. Significant proliferation and expansion of T cells was observed in 23 of 30 distinct samples. TIL were isolated from primary tumors by either enzymatic digestion or mechanical disruption. The TIL cultures were initiated using OKT3 mAb in the presence of moderate concentrations (25-50 U/ml) of IL-2, followed by 100 U/ml of tumor necrosis factor (TNF)-alpha. TAL were not stimulated with OKT3 mAb, but all were successfully expanded in culture in the presence of IL-2 alone or together with TNF-alpha. Seven of nine distinct TAL grew in culture as predominantly CD4+ lines. In contrast, only 14 of 21 (66%) of primary breast TIL expanded in culture and were predominantly of CD8+ phenotype. Autologous tumor lysis was observed in seven of eight cases tested. Only one of the four TIL tested and one of the four TAL tested preferentially lysed autologous tumor. HER-2 peptide E75 (369-377) was recognized by two TIL lines of the five primary TIL tested and three of the four TAL tested. This suggests that E75 may be recognized by primary breast tumors. This may be of interest in developing vaccine strategies for therapeutic management of breast cancer.


Subject(s)
Antigen-Antibody Reactions , Breast Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/pathology , Breast Neoplasms/immunology , Cell Division/immunology , Epitopes , Female , Humans , Immunophenotyping , Lymphocytes, Tumor-Infiltrating/immunology , Pleural Effusion, Malignant/immunology , Pleural Effusion, Malignant/pathology , Receptor, ErbB-2/immunology , Tumor Cells, Cultured
6.
Mol Immunol ; 35(17): 1121-33, 1998 Dec.
Article in English | MEDLINE | ID: mdl-10395201

ABSTRACT

In this study we investigated recognition by ovarian tumor associated lymphocyte (OVTAL), and breast tumor associated lymphocytes (BRTAL), of peptides corresponding to the sequence 125-135 of the Aminoenhancer of split (AES) protein. Three of these peptides designated as G75:AES1/2 (128-135), G60: AES1/2 (127-137) and G61: AES1/2 (125-133) correspond to the wildtype AES sequence, while the fourth G76:GPLTPLPV, AES1/2 (128-135) corresponds to a variant sequence of the peptide G75 with the N-terminal Leu substituted to glycine. These sequences were chosen for study because mass-spectrometric analysis (MS) of a CTL active HPLC peptide fraction eluted from immunoaffinity precipitated HLA-A2 molecule, revealed: (a) the presence of an ion with a mass-to-charge ratio (m/z) of 793 which was more abundant than other ions of similar masses; (b) the tentatively reconstituted sequence of the ion 793 matched the sequence of peptide G76. We found that AES peptides G75 (128-135) and G76 (128-135) (L128G) reconstituted CTL recognition at concentrations ranging between 200-500 nM. These concentrations are lower than concentrations reported to activate effector function of CTL recognizing other epithelial tumor Ag. Furthermore, analysis with cloned CD8+ T cells indicated that G75 and G76 were not cross-reactive specificities, suggesting a key role for the N-terminal residues of the variant peptide in dictating specificities. Since the AES proteins are part of a set of transcriptional repressors encoded by the Enhancer of split [E(spl)] genes, and since these repressors are activated to suppress cell differentiation in response to Notch receptors signalling, the AES peptides may represent a novel class of self-antigens that deserve further consideration as tumor Ag in epithelial cancers.


Subject(s)
Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Membrane Proteins/immunology , Ovarian Neoplasms/immunology , Proteins , Repressor Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Co-Repressor Proteins , Epitopes , Female , Humans , Mass Spectrometry , Peptide Fragments/immunology , Receptors, Notch , Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...