Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Cancer Gene Ther ; 13(8): 753-61, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16543916

ABSTRACT

Previous studies have shown that the human melanoma differentiation-associated gene-7 (mda-7)/interleukin-24 (IL-24) has tumor-suppressor activity in vitro and in vivo. Additionally, in vitro studies using human peripheral blood mononuclear cells indicate that mda-7/IL-24 has TH1 cytokine-like activity. However, the individual properties of mda-7/IL-24 have been previously examined separately. Thus, there is not a single study that has examined both, antitumor and proimmune properties of mda-7/IL-24. Furthermore, the tumor suppressive activity and the cytokine activity of mda-7/IL-24 have not been previously tested in an immunocompetent setting. We therefore in the present study evaluated the antitumor and immune properties of mda-7/IL-24 in a murine syngeneic tumor model. In vitro, adenovirus-mediated mda-7 gene (Ad-mda7) transfer to murine fibrosarcoma (UV2237m; MCA16) and normal (10T1/2) cells significantly inhibited growth (P=0.001) and induced apoptosis in tumor cells but not in normal cells. In vivo, intratumoral administration of Ad-mda7 resulted in significant inhibition of tumor growth (P<0.05), with a subset of mice showing complete tumor regression. We next evaluated the immune potentiation activity of Ad-mda7 in a cancer vaccine model. UV2237m cells transfected with Ad-mda7 and injected into syngeneic immunocompetent C3H mice were unable to grow; however, they did grow in immunocompromised nude mice. These tumor-free C3H mice, when challenged with parental tumor cells experienced no tumor growth, suggesting induction of systemic immunity. Moreover, splenocytes prepared from vaccinated C3H mice demonstrated higher proliferative activity and produced elevated levels of TH1 cytokines compared with those from control mice. An in vitro subset analysis of splenocytes from vaccinated mice demonstrated a significant increase in the CD3(+)CD8(+) but not the CD3(+)CD4(+) cell population (P=0.019). Thus Ad-mda7 treatment of syngeneic tumors induces tumor cell death and promotes immune activation, leading to anticancer immunity.


Subject(s)
Cancer Vaccines/immunology , Fibrosarcoma/therapy , Interleukins/immunology , Adenoviridae , Animals , Apoptosis/immunology , Cell Line , Cell Line, Tumor , Cell Proliferation , Cytokines/biosynthesis , Female , Fibrosarcoma/immunology , Genetic Therapy , Genetic Vectors , Immunocompetence , Injections, Intralesional , Interleukins/administration & dosage , Interleukins/genetics , Interleukins/therapeutic use , Mice , Mice, Inbred C3H , Spleen/cytology , Spleen/immunology , Th1 Cells/immunology , Transplantation, Isogeneic , Xenograft Model Antitumor Assays
2.
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
3.
Clin Cancer Res ; 6(11): 4192-200, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11106231

ABSTRACT

The presence of tumor-reactive CTLs in tumor infiltrates and in the peripheral blood of cancer patients demonstrates an immune response against tumors that apparently cannot control disease spread. This raises concerns as to whether amplification of this response may be useful during disease progression. Induction of tumor-reactive CTLs in healthy donors at risk, as well as in patients free of disease, may be therapeutically important, based on the hypothesis that CTLs that recognize tumors early may be more effective in containing their progression than CTLs that expand only when the disease progresses. To address the feasibility of priming cytolytic activity in healthy donors, we used the HER-2 peptide E75 (369-377) as an immunogen and autologous peripheral blood mononuclear cell-derived dendritic cells as antigen-presenting cells. We found that of 10 healthy donors tested, two responded at priming with E75 presented on autologous dendritic cells by induction of E75-specific CTL activity. Three other responders were identified after two additional restimulations. Of these five responders, three recognized E75 presented on the ovarian tumor line SKOV3.A2, as demonstrated by cold-target inhibition experiments. Induction of cytolytic activity at priming was enhanced in responders by tumor necrosis factor-alpha and interleukin 12 but not in the nonresponders. AlphaB7.1 monoclonal antibody added at priming enhanced induction of lytic activity in only one of the four nonresponding donors tested, suggesting that in the majority of donors, E75-precursor CTLs were not tolerized. Because of the possibility that disease may develop in nonresponders, strategies to improve the immunogenicity of tumor antigens for healthy donors may be required for development of cancer vaccines.


Subject(s)
Cytotoxicity, Immunologic , Peptide Fragments/immunology , Receptor, ErbB-2/immunology , B7-1 Antigen/physiology , CD28 Antigens/physiology , Dendritic Cells/physiology , Epitopes , Humans , Interleukin-12/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor-alpha/pharmacology
4.
Cancer Immunol Immunother ; 49(9): 459-68, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11092612

ABSTRACT

Immunization with tumor antigens induces cellular and humoral immune responses. These responses by T cells are specific for defined epitopes (determinants) in the molecule of the immunizing tumor antigen. Extension of such responses to self-antigens requires induction of autoimmunity to the tumor. As with systems of autoimmune disease, expression of T cell autoimmunity is charaterized by diversification of responses from the inducer determinant to other responder (cryptic) determinants. Since similar strategies may be useful for therapy of human cancers, we investigated whether the induction of response to a HER-2 peptide F7 (776-789) induces enhanced reactivity of other HER-2 peptides. We found that stimulation with F7 can expand a response to another epitope F13 (884-899) in both an ovarian cancer patient with progressive disease and a healthy donor who shared HLA-DR11. This response was characterized mainly by increased interferon gamma secretion, and proliferation, but was not observed with another donor who shared HLA-DR14 and HLA-DQ5 with the patient. Since repeated vaccination with the same epitope may lead to a decline of primary cell reactivity caused by apoptosis spreading the response to other epitopes, the tumor antigen may provide an approach for maintaining an inflammatory Th1 response during cancer vaccination.


Subject(s)
Peptides/metabolism , Receptor, ErbB-2/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Antigens, Neoplasm/metabolism , Antigens, Neoplasm/therapeutic use , Apoptosis , Case-Control Studies , Cell Division , Disease Progression , Enzyme-Linked Immunosorbent Assay , Epitopes , Female , HLA-DQ Antigens/metabolism , HLA-DR Antigens/metabolism , HLA-DR Serological Subtypes , Humans , Interferon-gamma/metabolism , Interleukin-2/pharmacology , Leukocytes, Mononuclear/metabolism , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/therapy , Peptides/immunology , Receptor, ErbB-2/immunology , Time Factors
5.
Biochemistry ; 39(33): 10319-29, 2000 Aug 22.
Article in English | MEDLINE | ID: mdl-10956021

ABSTRACT

Protein kinase C (PKC) isozymes are subject to inactivation by reactive oxygen species (ROS) through as yet undefined oxidative modifications of the isozyme structure. We previously reported that Cys-containing, Arg-rich peptide-substrate analogues spontaneously form disulfide-linked complexes with PKC isozymes, resulting in isozyme inactivation. This suggested that PKC might be inactivated by oxidant-induced S-glutathiolation, i.e., disulfide linkage of the endogenous molecule glutathione (GSH) to PKC. Protein S-glutathiolation is a reversible oxidative modification that has profound effects on the activity of certain enzymes and binding proteins. To directly examine whether PKC could be inactivated by S-glutathiolation, we used the thiol-specific oxidant diamide because its oxidant activity is restricted to induction of disulfide bridge formation. Diamide weakly inactivated purified recombinant cPKC-alpha, and this was markedly potentiated to nearly full inactivation by 100 microM GSH, which by itself was without effect on cPKC-alpha activity. Diamide inactivation of cPKC-alpha and its potentiation by GSH were both fully reversed by DTT. Likewise, GSH markedly potentiated diamide inactivation of a PKC isozyme mixture purified from rat brain (alpha, beta, gamma, epsilon, zeta) in a DTT-reversible manner. GSH potentiation of diamide-induced cPKC-alpha inactivation was associated with S-glutathiolation of the isozyme. cPKC-alpha S-glutathiolation was demonstrated by the DTT-reversible incorporation of [(35)S]GSH into the isozyme structure and by an associated change in the migration position of cPKC-alpha in nonreducing SDS-PAGE. Diamide treatment of NIH3T3 cells likewise induced potent, DTT-reversible inactivation of cPKC-alpha in association with [(35)S] S-thiolation of the isozyme. Taken together, the results indicate that PKC isozymes can be oxidatively inactivated by S-thiolation reactions involving endogenous thiols such as GSH.


Subject(s)
Glutathione/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , Protein Processing, Post-Translational , Animals , Brain/enzymology , Diamide/pharmacology , Dithiothreitol/pharmacology , Drug Synergism , Gene Expression Regulation, Enzymologic , Isoenzymes/drug effects , Oxidation-Reduction , Protein Kinase C/drug effects , Protein Kinase C-alpha , Rats , Rats, Sprague-Dawley
6.
Cancer Gene Ther ; 7(4): 530-6, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10811470

ABSTRACT

The immune responses of 10 patients with advanced non-small cell lung cancer receiving monthly intratumoral injections of a recombinant adenovirus containing human wild-type p53 (Ad-p53) to adenovirus and transgene antigens were studied. The predominate cellular and humoral immune responses as measured by lymphocyte proliferation and neutralizing antibody (Ab) formation were to adenovirus serotype 5 vector antigens, with increased responses in posttreatment samples. Consistent alterations in posttreatment cellular and humoral immune responses to p53 epitopes were not observed, and cytotoxic Abs to human lung cancer cells were not generated. Patients in this study had evidence of an antitumoral effect of this treatment with prolonged tumor stability or regression; however, neither Abs to p53 protein nor increased lymphocyte proliferative responses to wild-type or mutant p53 peptides have been consistently detected.


Subject(s)
Adenoviridae/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Genetic Therapy/methods , Lung Neoplasms/therapy , Tumor Suppressor Protein p53/immunology , Adenoviridae/genetics , Aged , Amino Acid Sequence , Antibody Formation , Carcinoma, Non-Small-Cell Lung/immunology , Cytotoxicity, Immunologic , Female , Gene Transfer Techniques , Genetic Vectors/immunology , Humans , Immunity, Cellular , Lung Neoplasms/immunology , Lymphocyte Activation , Male , Middle Aged , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/immunology , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
7.
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
8.
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
9.
J Immunother ; 23(1): 28-35, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10687135

ABSTRACT

Class I expression in context with T-cell receptor expression is crucial for peptide presentation and induction of CD8+ cytotoxic T lymphocytes (CTL). Presentation of class I bound peptides is dependent on transporter-associated proteins (TAP) expression and function. Tumor infiltrating lymphocytes from a patient with melanoma were isolated, expanded in vitro in the presence of interleukin-2, and tested for cytotoxicity against HLA-A2 positive, MART-1 positive autologous tumor cells, an HLA-A2-positive, MART-1 positive melanoma cell line (Mel-501), and HLA-A2-negative melanoma cells. Significant killing occurred against both A2-positive cell lines (63% and 65%, respectively), but not against the A2-negative line (18%) or A2-positive autologous tumor (1.5%). These CTL preferentially recognized the MART-1 peptide F119, 27-35, and gp100 peptide F125, 280-288, resulting in a 30% to 60% enhancement of lysis when autologous tumor or major histocompatibility complex class I "empty" T2 cells were pulsed with either peptide. To address whether the deficiency in autologous tumor recognition might be related to a deficiency in Ag presentation, we screened for the presence of TAP1 and TAP2 transcripts by polymerase chain reaction, Southern blotting, and scanning densitometry using sequence-specific primers and probes. Both TAP1 and TAP2 expression levels in the autologous tumor were minimal, yet were upregulated 7- to 18-fold, respectively, by interferon-gamma. Despite this increase, a similar increase in cytotoxicity did not occur. In short, deficiencies in TAP presentation may have functional significance for tumor escape from immunosurveillance and with respect to impending vaccine trials.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Abdominal Neoplasms/immunology , Antigens, Neoplasm/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Neoplasm Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP Binding Cassette Transporter, Subfamily B, Member 3 , Abdominal Neoplasms/secondary , Cytotoxicity, Immunologic , Female , Gene Expression , HLA-A2 Antigen/immunology , Humans , Interferon-gamma/immunology , Interferon-gamma/pharmacology , MART-1 Antigen , Major Histocompatibility Complex , Melanoma/pathology , Melanoma/secondary , Middle Aged , Neoplasm Metastasis , Peptides/immunology , Phenotype , T-Lymphocytes, Cytotoxic/drug effects , Tumor Cells, Cultured
10.
Semin Oncol ; 27(6 Suppl 11): 71-5; discussion 92-100, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11236031

ABSTRACT

Recently there has been a renewed interest in developing vaccines for use in cancer treatment. Part of this interest stems from a better understanding of the immune system, the identification of a number of T-cell-specific tumor antigens, more effective adjuvants, and the ability to construct more immunogenic molecules using recombinant DNA techniques. Studies from several laboratories have shown that breast cancer patients have preexisting immunity to the HER-2/neu oncoprotein receptor (HER-2) in the form of elevated antibody titers and T-cell immunity. Preclinical studies showed enhanced delayed-type hypersensitivity and cytotoxic T-lymphocyte precursors in spleens from animals immunized with several human leukocyte antigen class I and class II peptides derived from the HER-2 protein. Phase I trials of these peptides combined with the cytokine granulocyte-macrophage colony-stimulating factor as a part of therapy in patients with HER-2-positive cancers have shown minimal local toxicity, along with enhanced helper T-cell activity and antibody production in patients with minimal disease. Increases in cytotoxic T-lymphocyte precursor activity were less frequent, but in some cases could be enhanced when patient lymphocytes were incubated ex vivo with the proinflammatory cytokine interleukin-12. Other preclinical studies designed to enhance HER-2 peptide immunogenicity are in progress. Additional current and future clinical trials using HER-2-derived vaccines will be discussed.


Subject(s)
Cancer Vaccines , Receptor, ErbB-2 , Animals , Antigens, Neoplasm , Breast Neoplasms/therapy , Clinical Trials as Topic , Female , Humans , Ovarian Neoplasms/therapy , Peptide Fragments
11.
Biochemistry ; 38(40): 13244-51, 1999 Oct 05.
Article in English | MEDLINE | ID: mdl-10529197

ABSTRACT

Resveratrol, a polyphenolic natural product abundantly present in grape skins, is a candidate cancer chemopreventive agent that antagonizes each stage of carcinogenesis and inhibits protein kinase C (PKC), a key mediator of tumor promotion. While resveratrol has been shown to antagonize both isolated and cellular forms of PKC, the weak inhibitory potency observed against isolated PKC cannot account for the reported efficacy of the polyphenol against PKC in cells. In this report, we analyze the mechanism of PKC inhibition by resveratrol. Our results indicate that resveratrol has a broad range of inhibitory potencies against purified PKC that depend on the nature of the substrate and the cofactor dependence of the phosphotransferase reaction. Resveratrol weakly inhibited the Ca2+/phosphatidylserine-stimulated activity of a purified rat brain PKC isozyme mixture (IC(50) = 90 microM) by competition with ATP (K(i) = 55 microM). Consistent with the kinetic evidence for a catalytic domain-directed mechanism, resveratrol inhibited the lipid-dependent activity of PKC isozymes with divergent regulatory domains similarly, and it was even more effective in inhibiting a cofactor-independent catalytic domain fragment (CDF) of PKC generated by limited proteolysis. This suggested that regulatory features of PKC might impede resveratrol inhibition of the enzyme. To explore this, we examined the effects of resveratrol on PKC-catalyzed phosphorylation of the cofactor-independent substrate protamine sulfate, which is a polybasic protein that activates PKC by a novel mechanism. Resveratrol potently inhibited protamine sulfate phosphorylation (IC(50) = 10 microM) by a mechanism that entailed antagonism of the activation of PKC by protamine sulfate and did not involve competition with either substrate. On the basis of the presence of PKC isozymes at subcellular sites rich in polybasic proteins, it has been proposed that certain endogenous polybasic PKC substrates may activate PKC in cells by the same mechanism as protamine sulfate. Our results suggest that antagonism by resveratrol of the phosphorylation of cellular PKC substrates that resemble protamine sulfate in their interactions with PKC may contribute to the efficacy of resveratrol against PKC in cells.


Subject(s)
Arginine/metabolism , Enzyme Inhibitors/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Stilbenes/pharmacology , Animals , Brain , Catalysis/drug effects , Catalytic Domain/drug effects , Enzyme Activation/drug effects , Isoenzymes/antagonists & inhibitors , Kinetics , Phosphatidylserines/metabolism , Phosphorylation/drug effects , Protamines/metabolism , Rats , Resveratrol , Substrate Specificity/drug effects
12.
Cancer Immunol Immunother ; 48(7): 401-10, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10501854

ABSTRACT

Although natural killer (NK) cells have been described as non-MHC-restricted, new evidence suggests that NK activity can be either up- or down-regulated after interaction with the peptide-MHC-class-I complex expressed on target cells. However, the epitope(s) recognized by NK cells have remained ill-defined. We investigated NK cell recognition of synthetic peptides representing a portion of a self-protein encoded by the HER-2/neu (HER-2) proto-oncogene and presented by HLA-A2. HER-2 nonapeptides C85, E89, and E75 were found partially to protect T2 targets from lysis by freshly isolated and interleukin-2(IL-2)-activated NK cells (either HLA-A2(+) or A2(-)). This inhibition was not solely due to changes in the level of HLA-A2 expression or conformation of serological HLA-A2 epitopes. Using single-amino-acid variants at position 1 (P1) of two HER-2 peptides, we observed that protection of targets was dependent on the sequence and the side-chain. These results suggest similarities in the mechanism of target recognition by NK and T cells. This information may be important for understanding the mechanisms of tumor escape from immunosurveillance and could help explain the aggressiveness of HER-2-overexpressing tumor cells.


Subject(s)
Epitopes/immunology , HLA-A2 Antigen/immunology , Killer Cells, Natural/immunology , Receptor, ErbB-2/immunology , Cells, Cultured , Cytotoxicity Tests, Immunologic , Flow Cytometry , HLA-A2 Antigen/metabolism , Histocompatibility Antigens Class I/metabolism , Humans , Interleukin-2/pharmacology , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Mutagenesis, Site-Directed , Peptide Fragments/chemical synthesis , Peptide Fragments/immunology , Peptide Fragments/pharmacology , Proto-Oncogene Mas , Receptor, ErbB-2/genetics , Transfection
13.
Clin Cancer Res ; 5(12): 4214-23, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10632363

ABSTRACT

The immune system can be efficiently stimulated and targeted to specific antigens expressed exclusively or preferentially by experimental cancers. The foremost limitations to extending this vaccine technology to the prevalent epithelial-derived cancers are the lack of: (a) identified tumor-associated antigens recognized by cellular immunity; (b) antigens expressed on the majority of tumor cells during disease progression; and (c) immunogenic CTL epitopes. To date, only HER-2/neu has been shown to be the source of naturally occurring, MHC-restricted, CTL-recognized peptides in epithelial tumors. In this study, we demonstrate that the human high-affinity folate binding protein (FBP), which is a source of antigenic peptides recognized in ovarian cancer, is also recognized in breast cancer. Both immunodominant E39 (FBP, 191-199) and subdominant E41 (FBP, 245-253) epitopes are presented by HLA-A2 in these cancers. These peptides are efficient at amplifying the response of tumor-associated lymphocyte populations in terms of lytic function, enhanced proliferation, and specific IFN-gamma release. On a per cell basis, tumor-associated lymphocytes stimulated with the FBP peptides exhibit enhanced cytotoxicity not only against peptide-loaded targets but also against FBP-expressing epithelial tumors of different histologies. Furthermore, FBP peptides induced E39-specific CTLs and E39- and E41-specific IFN-gamma and IP-10 secretion in certain healthy donors. The broad distribution of FBP among >90% of ovarian and endometrial carcinomas, as well as 20-50% of breast, lung, colorectal, and renal cell carcinomas, along with pronounced differential overexpression in malignant tissues compared with the extremely limited expression in normal epithelium, suggests the exciting potential of a widely applicable FBP-based vaccine in epithelial cancers.


Subject(s)
Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Cancer Vaccines/immunology , Carrier Proteins/immunology , Cytotoxicity, Immunologic , Ovarian Neoplasms/immunology , Receptors, Cell Surface , T-Lymphocytes, Cytotoxic/immunology , Antigen Presentation , Antigens, Neoplasm/metabolism , Carrier Proteins/metabolism , Chemokine CXCL10 , Chemokines, CXC/biosynthesis , Female , Folate Receptors, GPI-Anchored , Folic Acid/immunology , Folic Acid/metabolism , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Peptides/immunology , T-Lymphocytes, Cytotoxic/metabolism
14.
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
15.
Ann Surg Oncol ; 5(8): 743-50, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9869522

ABSTRACT

BACKGROUND: Tumor-associated lymphocytes (TAL) isolated from ovarian cancer patients contain cytotoxic T lymphocytes (CTL) capable of recognizing specific HLA/peptide complexes on tumor cells leading to tumor cell lysis. Currently, HER2/neu, overexpressed in only 30% of breast and ovarian cancers, is the only known source of CTL-recognized peptides in epithelial cancers. Therefore, we have investigated peptides derived from folate binding protein (FBP), which is over-expressed in more than 90% of ovarian cancers and in the majority of other epithelial tumors. METHODS: TAL were isolated from the malignant ascites of four consecutive HLA-A2+ ovarian cancer patients and incubated in IL-2. Initial chromium-release assays were performed within 1 week. T2 cells, incubated with peptide, were used to reconstitute T cell epitopes. The FBP sequence was interrogated for HLA-A2 binding peptides, and five were synthesized (E37-41). RESULTS: Freshly cultured, unstimulated ovarian TAL recognize peptides derived from FBP. These peptides are presented in the context of HLA-A2, and are specifically recognized in a HLA class I-restricted fashion. TAL recognition of these reconstituted T cell epitopes is concentration dependent. Furthermore, the FBP peptides are shown by cold target inhibition studies to be naturally processed and presented antigens. CONCLUSIONS: FBP peptides are recognized by freshly isolated TAL from ovarian cancer patients, suggesting in vivo expression and sensitization. Because FBP is over-expressed 20-fold in most adenocarcinomas, these peptides may be used in a widely applicable peptide-based vaccine for epithelial tumors.


Subject(s)
Cancer Vaccines , Carrier Proteins/metabolism , Ovarian Neoplasms/immunology , Receptors, Cell Surface/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Carrier Proteins/chemistry , Carrier Proteins/immunology , Female , Folate Receptors, GPI-Anchored , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Humans , Peptides/chemical synthesis , Peptides/immunology , Peptides/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
16.
Cancer Res ; 58(21): 4790-3, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9809978

ABSTRACT

The emergence of drug resistance to chemotherapeutic agents is a major cause of treatment failure in cancer therapy. Therefore, much effort has been aimed at circumventing or reversing this undesired effect. Recently, we found that tumor cell lines selected for their multidrug-resistant phenotype can also exhibit increased levels of TAP mRNA and MHC class I proteins. This raised the question of whether drug-resistant tumors are more readily recognized by MHC-restricted CTLs. In this report, we show that five of five MHC class I+ tumor cell lines grown in medium containing Adriamycin developed into variants that expressed higher levels of MHC class I than did their corresponding parental cell lines. This was not observed with a MHC class I- cell line. No similar association was noted for changes in the expression of either HER-2 or intercellular adhesion molecule 1 protein. We also found that MHC class I+ drug-selected variants were more readily lysed by MHC-restricted, tumor-associated CTLs than were the drug-sensitive parental cell lines. When the drug-selected variants were cocultured with the same CTLs to eliminate tumor cells expressing higher levels of MHC-I (MHC-Ihi), the CTL-resistant tumor cells exhibited a drug sensitivity profile similar to that of the parental cell lines that were not exposed to Adriamycin. These findings suggest that certain chemotherapeutic drugs may increase the immunogenicity of some tumors, and that CTL immunotherapy may help reverse drug resistance.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cytotoxicity, Immunologic/drug effects , Doxorubicin/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , Drug Resistance, Neoplasm , Female , Histocompatibility Antigens Class I/analysis , Humans , Intercellular Adhesion Molecule-1/analysis , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
17.
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
18.
Clin Cancer Res ; 4(8): 2015-24, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9717833

ABSTRACT

Previous studies have characterized the reactivity of CD8+ CTLs with ovarian and breast cancer. There is little information about the antigens and epitopes recognized by CD4+ T cells in these patients. In this study, we analyzed the ability of T cells from peripheral blood mononuclear cells of breast cancer patients to recognize HER-2/neu (HER-2) peptides. We found that 13 of 18 patients responded by proliferation to at least one of the HER-2 peptides tested. Of these peptides, one designated G89 (HER-2: 777-789) was recognized by T cells from 10 patients. Seven of nine responding patients were HLA-DR4+, suggesting that this peptide is recognized preferentially in association with HLA-DR4. Analysis of the specificity and restriction of the cytokine responses to G89 by G89-stimulated T cells revealed that these cells secreted significantly higher levels of IFN-gamma than interleukin 4 and interleukin 10, suggesting priming for a Th0-T helper 1 response. The same pattern of cytokine responses was observed to the intracellular domain of HER-2 protein, suggesting that G89-stimulated T cells recognized epitopes of the HER-2 protein in association with HLA-DR4. Because HLA-DR4 is present in 25% of humans, characterization of MHC class II-restricted epitopes inducing Th0-T helper 1 responses may provide a basis for the development of multivalent HER-2-based vaccines against breast and ovarian cancer.


Subject(s)
Breast Neoplasms/blood , Cytokines/biosynthesis , Cytokines/metabolism , Leukocytes, Mononuclear/drug effects , Lymphocyte Activation/drug effects , Peptide Fragments/pharmacology , Receptor, ErbB-2/pharmacology , Amino Acid Sequence , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Division/drug effects , Female , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Molecular Sequence Data , Receptor, ErbB-2/biosynthesis , Sequence Homology, Amino Acid , Stimulation, Chemical , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
19.
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
20.
Anticancer Res ; 17(1A): 45-53, 1997.
Article in English | MEDLINE | ID: mdl-9066629

ABSTRACT

Identifying target antigens for tumor-reactive T cells is important for understanding the mechanisms of tumor escape and developing novel anticancer therapies. To date, mainly CTL responses from tumor infiltrating associated lymphocytes (TIL/TAL) to peptide antigens have been investigated in ovarian cancer. In the present study, the ability of self-peptides derived from HER-2/neu proto-oncogene product (HER-2) to stimulate proliferation of PBMC from healthy donors and ovarian cancer patients has been assessed. Peptide sequences from HER-2 containing anchors for major human MHC-class II molecules have been identified. These peptides induced proliferative and cytokine responses at higher frequency in healthy donors than ovarian cancer patients. Four HER-2 peptides corresponding to positions: 396-406, 474-487, 777-789, and 884-899 were able to stimulate proliferation of a larger number of healthy donors than three other distinct HER-2 peptides 449-464, 975-987 and 1086-1098. The pattern of responses of twenty five ovarian cancer patients was different from that in healthy donors. T cell lines were developed by stimulation with peptides from PBMC of an ovarian cancer patient who showed a stable response to all four HER-2 peptides for over six months. Each T cell line was different in its ability to secrete IFN-gamma and IL-10. These results demonstrate (a) that self-peptides from HER-2 can stimulate expansion of T cells in both healthy donors and ovarian cancer patients, and (b) the ability of different peptides to stimulate secretion of different cytokines from lymphocytes of ovarian cancer patients. These results may be important for understanding the mechanisms of tolerance and autoimmunity in human cancers.


Subject(s)
Lymphocyte Activation , Ovarian Neoplasms/immunology , Receptor, ErbB-2/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Cytokines/biosynthesis , Female , Humans , Molecular Sequence Data , Proto-Oncogene Mas
SELECTION OF CITATIONS
SEARCH DETAIL
...