Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Clin Transl Oncol ; 21(6): 721-728, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30374838

ABSTRACT

BACKGROUND: Advanced non-small cell lung cancer (NSCLC) has remained challenging to treat effectively. This study aimed to investigate the clinical effects and safety of immunotherapy with dendritic cells and cytokine-induced killer cells (DC-CIK) administered with chemotherapy (CT) in this malignancy. METHODS: We have developed a new clinical trial design termed as the prospective patient's preference-based study (PPPS). Consecutive patients (n = 135) with advanced NSCLC were treated with DC-CIK administered with CT or mono-therapy (CT or DC-CIK alone). RESULTS: For all the patients, the median PFS was 5.7 months and the median OS was 17.5 months. The 1-year PFS and OS rates were 29.4% and 58.2%, respectively. The 1-year PFS and OS rates for DC-CIK plus CT were significantly higher than that in the group of patients who received DC-CIK alone and CT alone (P < 0.05). The number of adoptively infused DC-CIK cells was associated with clinical efficacy. After adjusting for competing risk factors, DC-CIK combined with CT and infused number of CIKs remained independent predictors of PFS and OS. Phenotypic analysis of peripheral blood mononuclear cells showed that CD8+CD28+, and CD8+CD28- T cells, changed significantly in all groups (P < 0.01). The CD3+ T cells increased in the chemotherapy plus immunotherapy and the immunotherapy alone group (P < 0.01), while CD3-CD16+CD56 T cells decreased in the chemotherapy plus immunotherapy and the immunotherapy alone group (P < 0.01). CONCLUSIONS: DC-CIK combined with chemotherapy administration resulted in numerically superior PFS and OS compared with monotherapy in advanced NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Cytokine-Induced Killer Cells/transplantation , Dendritic Cells/transplantation , Immunotherapy, Adoptive , Lung Neoplasms/therapy , Patient Preference , T-Lymphocytes/transplantation , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Cytokine-Induced Killer Cells/immunology , Dendritic Cells/immunology , Female , Follow-Up Studies , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Male , Middle Aged , Prognosis , Prospective Studies , Survival Rate , T-Lymphocytes/immunology
2.
Cancer Gene Ther ; 19(1): 30-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21997230

ABSTRACT

CD4+CD25+regulatory T cells (T(reg)) impair anti-tumor and anti-viral immunity. As there are higher T(reg) levels in cancer patients compared with healthy individuals, there is considerable interest in eliminating them or altering their function as part of cancer or viral immunotherapy strategies. The scurfin transcriptional regulator encoded by the member of the forkhead winged helix protein family (FOXP3) is critical for maintaining the functions of T(reg). We hypothesized that targeting FOXP3 expression with a novel arginine-rich, cell-penetrating, peptide-conjugated phosphorodiamidate morpholino (PPMO) based antisense would eliminate T(reg) and enhance the induction of effector T-cell responses. We observed that the PPMO was taken up by activated T cells in vitro and could downregulate FOXP3 expression, which otherwise increases during antigen-specific T-cell activation. Generation of antigen-specific T cells in response to peptide stimulation was enhanced by pre-treatment of peripheral blood mononuclear cells with the FOXP3-targeted PPMO. In summary, modulation of T(reg) levels using the FOXP3 PPMO antisense-based genomic strategy has the potential to optimize immunotherapy strategies in cancer and viral immunotherapy.


Subject(s)
Forkhead Transcription Factors/genetics , Morpholinos/pharmacology , Oligonucleotides, Antisense/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Animals , Case-Control Studies , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/immunology , Humans , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Morpholinos/genetics , Morpholinos/pharmacokinetics , Neoplasms/blood , Neoplasms/immunology , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacokinetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , T-Lymphocytes, Regulatory/metabolism
3.
Clin Exp Immunol ; 163(3): 324-32, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21175594

ABSTRACT

Selection of suitable antigens is critical for the development of cancer vaccines. Most desirable are over-expressed cell surface proteins that may serve as targets for both antibodies and T cells, thus maximizing a concerted immune response. Towards this goal, we characterized the relevance of tumour necrosis factor-α-converting enzyme (ADAM17) for such targeted therapeutics. ADAM17 is one of the several metalloproteinases that play a key role in epidermal growth factor receptor (EGFR) signalling and has recently emerged as a new therapeutic target in several tumour types. In the present study, we analysed the expression profile of ADAM17 in a variety of normal and cancer cells of human origin and found that this protein is over-expressed on the surface of several types of cancer cells compared to the normal counterparts. Furthermore, we analysed the presentation of a human leucocyte antigen (HLA)-A2-restricted epitope from ADAM17 protein to specific T cells established from normal donors as well as ovarian cancer patients. Our analysis revealed that the HLA-A2-restricted epitope is processed efficiently and presented by various cancer cells and not by normal cells. Tumour-specific T cell activation results in the secretion of both interferon-γ and granzyme B that can be blocked by HLA-A2 specific antibodies. Collectively, our data present evidence that ADAM17 can be a potential target antigen to devise novel immunotherapeutic strategies against ovarian, breast and prostate cancer.


Subject(s)
ADAM Proteins/immunology , Breast Neoplasms/immunology , Histocompatibility Antigens Class I/immunology , Immunotherapy , Ovarian Neoplasms/immunology , Prostatic Neoplasms/immunology , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM17 Protein , Antigen Presentation/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cell Line, Tumor , Epitopes, T-Lymphocyte/immunology , Female , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/physiology , Granzymes/metabolism , HLA-A2 Antigen/immunology , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/therapy , Peptide Fragments/immunology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/therapy , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
4.
Br J Cancer ; 102(1): 124-33, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-19953093

ABSTRACT

BACKGROUND: Novel technologies to redirect T-cell killing against cancer cells are emerging. We hypothesised that metastatic human colorectal cancer (CRC) previously treated with conventional chemotherapy would be sensitive to T-cell killing mediated by carcinoembryonic antigen (CEA)/CD3-bispecific T-cell-engaging BiTE antibody (MEDI-565). METHODS: We analysed proliferation and lysis of CEA-positive (CEA+) CRC specimens that had survived previous systemic chemotherapy and biologic therapy to determine whether they could be killed by patient T cells engaged by MEDI-565 in vitro. RESULTS: At low concentrations (0.1-1 ng ml(-1)), MEDI-565+ T cells caused reduced proliferation and enhanced apoptosis of CEA+ human CRC specimens. High levels of soluble CEA did not impair killing by redirected T cells and there was no increase in resistance to T-cell killing despite multiple rounds of exposure. CONCLUSIONS: This study shows for the first time that metastatic CRC specimens derived from patients previously treated with conventional chemotherapy can be lysed by patient T cells. Clinical testing of cancer immunotherapies, such as MEDI-565 that result in exposure of tumours to large numbers of T cells, is warranted.


Subject(s)
Adenocarcinoma/secondary , Antibodies, Bispecific/immunology , CD3 Complex/immunology , Carcinoembryonic Antigen/immunology , Colorectal Neoplasms/immunology , Liver Neoplasms/secondary , T-Lymphocytes, Cytotoxic/immunology , Adenocarcinoma/drug therapy , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , Antibodies, Bispecific/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Drug Resistance, Neoplasm , Fas Ligand Protein/physiology , Granzymes/physiology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Organoplatinum Compounds/pharmacology , Oxaliplatin , fas Receptor/physiology
5.
Expert Opin Biol Ther ; 1(2): 153-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11727526

ABSTRACT

Clinically effective cancer immunotherapy has been sought for more than 100 years and has been recently applied most successfully in strategies that passively deliver immune effectors such as monoclonal antibodies (anti-CD20 for lymphoma and anti-HER2/neu for breast cancer), donor lymphocyte infusions in chronic myelongenous leukemia and non-myeloablative allogeneic peripheral blood progenitor transplants for renal cell carcinoma. There is mounting enthusiasm for strategies employing active stimulation of antitumour immune responses. These include vaccines based on tumour antigen proteins and peptides, autologous, allogeneic or gene-modified tumour cells, dendritic cells and antigen-encoding viral vector constructs. Indeed, randomised Phase III clinical trials of autologous tumour cell vaccines for colorectal cancer demonstrated an improvement in disease free survival and a trend toward improved overall survival [1]. Despite these preliminary successes, it is clear that the many strategies under development cannot all be evaluated for survival benefit in large clinical trials that require many years, patients and resources to complete. This highlights the need to develop intermediate markers to help prioritise which agents to test in prospective randomised Phase III trials.


Subject(s)
Biomarkers/analysis , Immunotherapy , Monitoring, Immunologic/methods , Neoplasms/immunology , Neoplasms/therapy , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Clinical Trials, Phase III as Topic , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Humans , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Treatment Outcome
6.
Int J Cancer ; 94(6): 834-41, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11745486

ABSTRACT

B16F10 melanoma is a tumor derived from C57BL/6 mice that has been found to be poorly immunogenic and highly aggressive. Here we have shown that vaccination of mice with irradiated B16F10 cells followed by treatment with a combination of staphylococcal enterotoxins A and B (SEA/SEB) leads to significant and specific protection against subsequent challenge with viable B16F10 cells (at least 25-fold greater than a lethal dose). Also, 75% of mice surviving over 150 days remained tumor-free after rechallenge with viable B16F10 cells, evidence of the development of strong immunologic memory. Additional studies showed increases in CD4(+) and CD8(+) T-cell populations, cytotoxic T-lymphocyte activity and interferon-gamma production, all of which may contribute to enhanced survival. Furthermore, failure to produce protection in either CD4(-/-) or CD8(-/-) T-cell knockout mice is evidence that CD4(+) and CD8(+) T cells play an essential role in induction of immunity. These results show that superantigen administration subsequent to vaccination with inactivated tumor cells results in protective antitumor immunity. Thus, prophylactic vaccination against cancer is a feasible method for arming the immune system prior to the incidence of cancer.


Subject(s)
Cancer Vaccines/immunology , Enterotoxins/pharmacology , Melanoma, Experimental/immunology , Neoplasm Proteins/immunology , Superantigens/pharmacology , Animals , Antigens, Neoplasm , Female , Interferon-gamma/pharmacology , Melanoma-Specific Antigens , Mice , Mice, Inbred C57BL , Vaccination
7.
Crit Rev Immunol ; 21(1-3): 287-97, 2001.
Article in English | MEDLINE | ID: mdl-11642610

ABSTRACT

Successful application of active immunotherapy to the treatment of cancer will require stimulation of potent antigen-specific T-cell responses. It is not known how numerous or how potent these T cells must be in order to abrogate tumors, but the levels of immunity needed to control chronic viral infections may provide estimates for comparison. Evaluation of the efficacy of a vaccine strategy in attaining these levels of immunity will depend on the use of assays that create a picture of T-cell number and function that correlates with clinical outcomes. We discuss the currently available in vivo and in vitro T-cell assays and their relevance for detecting therapeutic levels of T-cell activity. We also propose a strategy for efficiently evaluating the immunologic efficacy of cancer vaccines so that the most promising candidates can be brought more rapidly into definitive clinical trials.


Subject(s)
Cancer Vaccines/immunology , T-Lymphocytes/immunology , Cytokines/analysis , Cytokines/genetics , Cytotoxicity Tests, Immunologic , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Hypersensitivity, Delayed , Lymphocyte Activation , RNA, Messenger/analysis
8.
Clin Cancer Res ; 7(5): 1127-35, 2001 May.
Article in English | MEDLINE | ID: mdl-11350875

ABSTRACT

Numerous cancer immunotherapy strategies are currently being tested in clinical trials. Although clinical efficacy will be the final test of these approaches, the long and complicated developmental pathway for these agents necessitates evaluating immunological responses as intermediate markers of the most likely candidates for success. This has emphasized the need for assays that accurately detect and quantitate T cell-mediated, antigen-specific immune responses. This review evaluates the currently used in vivo and in vitro methods of assessing T-cell number and function, including delayed-type hypersensitivity, tetramer analysis, ELISPOT, flow cytometry-based analysis of cytokine expression, and PCR-based detection of T-cell receptor gene usage or cytokine production. We provide examples of how each has been used to monitor recent clinical trials and a discussion of how well each correlates with clinical outcome.


Subject(s)
Immunotherapy, Active , Neoplasms/therapy , Cytokines/genetics , Cytokines/metabolism , Humans , Immunity, Active , Immunity, Cellular , Neoplasms/immunology , Phenotype , Reverse Transcriptase Polymerase Chain Reaction
9.
Exp Biol Med (Maywood) ; 226(3): 164-76, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11361034

ABSTRACT

Increasing evidence suggests that superantigens play a role in immune-mediated diseases. Superantigens are potent activators of CD4+ T cells, causing rapid and massive proliferation of cells and cytokine production. This characteristic of superantigens can be exploited in diseases where strong immunologic responses are required, such as in the B16F10 animal model of melanoma. Superantigen administration is able to significantly enhance ineffective anti-tumor immune responses, resulting in potent and long-lived protective anti-tumor immunity. However, superantigens are more well-known for the role they play in diseases. Studies using an animal model for neurologic demyelinating diseases such as multiple sclerosis show that superantigens can induce severe relapses and activate autoreactive T cells not involved in the initial bout of disease. This may also involve epitope spreading of disease. Superantigens have also been implicated in acute diseases such as food poisoning and TSS, and in chronic diseases such as psoriasis and rheumatoid arthritis. Viral superantigens are also involved in the disease process, including superantigens derived from human immunodeficiency virus and mouse mammary tumor virus. Finally, immunotherapies that ameliorate the role played by superantigens in disease are discussed.


Subject(s)
Superantigens/immunology , Animals , Humans
10.
Curr Opin Mol Ther ; 3(1): 45-52, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11249731

ABSTRACT

Many clinical trials are testing the feasibility of stimulating the immune system to treat cancer. Although the efficacy of this approach will ultimately be determined by clinically relevant endpoints, detection of the magnitude and activity of the immune response is an important intermediate point in the development of these strategies. Assays that predict clinically relevant endpoints are particularly desirable for helping to determine which strategies should ultimately be tested in larger randomized clinical trials. In this review, we will discuss these cellular immunological assays and the current status of their role in clinical trials of immunotherapy.


Subject(s)
Immunity, Cellular , Immunoassay/methods , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Animals , Clinical Trials as Topic , Complementarity Determining Regions , Cytokines/genetics , Cytokines/metabolism , Humans , Hypersensitivity, Delayed , In Vitro Techniques , Lymphocyte Activation , Peptides/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/immunology
11.
Surgery ; 129(3): 248-54, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11231452

ABSTRACT

The evaluation of cancer immunotherapy is predicated on the hypothesis that markers of tumor antigen-specific T-cell immunity will cone-late with clinical efficacy. Establishing which candidate vaccines should enter large-scale clinical trials will necessitate optimal application of immunologic monitoring assays. Evidence suggests that available techniques are adequate for the direct detection of clinically significant antigen-specific T-cell responses from tissue specimens. To achieve this goal, it is important to have an understanding of individual methods and their limitations, to include appropriate control antigens in the monitoring strategy, and to incorporate statistical considerations into the design and analysis of such studies.


Subject(s)
Cancer Vaccines/immunology , Immunity, Cellular , Epitopes , Humans , T-Lymphocytes/immunology
12.
J Clin Oncol ; 18(23): 3883-93, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11099317

ABSTRACT

PURPOSE: To evaluate preoperative dendritic cell (DC) mobilization and tumor infiltration after administration of Flt3 ligand (Flt3L) to patients with metastatic colon cancer. PATIENTS AND METHODS: Twelve patients with colon cancer metastatic to the liver or lung received Flt3L (20 microg/kg/d subcutaneously for 14 days for one to three cycles at monthly intervals) before attempted metastasectomy. The number and phenotype of DCs mobilized into peripheral-blood mononuclear cells (PBMCs) were evaluated by flow cytometry. After surgical resection, metastatic tumor tissue was evaluated for DC infiltration. In vivo immune responses to recall antigens were measured. RESULTS: After Flt3L administration, on average, the total number of leukocytes in the peripheral blood increased from 5.9 +/- 1.0 x 10(3)/mm(3) to 11.2 +/- 3.8 x 10(3)/mm(3) (mean +/- SD, P: =. 0001). The percentage of CD11c(+)CD14(-) DCs in PBMCs increased from 2.4% +/- 1.8% to 8.8% +/- 4.7% (P: =.004). Delayed-type hypersensitivity (DTH) responses to recall antigens (CANDIDA:, mumps, and tetanus) showed marginally significant increases in reactivity after Flt3L administration (P: =.06, P: =.03, and P: =.08, respectively). An increase in the number of DCs was observed at the periphery of the tumors of patients who received Flt3L compared with those of patients who had not. CONCLUSION: Flt3L is capable of mobilizing DCs into the peripheral blood of patients with metastatic colon cancer and may be associated with increases in DC infiltration in the peritumoral regions. Flt3L mobilization is associated with a trend toward increased DTH responses to recall antigens in vivo. The use of Flt3L to increase circulating DCs for cancer immunotherapy should be considered.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Colonic Neoplasms/immunology , Dendritic Cells/immunology , Immunotherapy, Active/methods , Membrane Proteins/immunology , Antigens/immunology , Blood Cell Count , Colonic Neoplasms/drug therapy , Colonic Neoplasms/surgery , Female , Humans , Hypersensitivity, Delayed/immunology , Immunophenotyping , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lymphocytes, Tumor-Infiltrating/immunology , Male , Membrane Proteins/adverse effects , Membrane Proteins/therapeutic use , Middle Aged , T-Lymphocytes/immunology
13.
Blood ; 96(10): 3499-504, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11071647

ABSTRACT

Dendritic cells (DCs) may arise from multiple lineages and progress through a series of intermediate stages until fully mature, at which time they are capable of optimal antigen presentation and T-cell activation. High cell surface expression of CD83 is presumed to correlate with full maturation of DCs, and a number of agents have been shown to increase CD83 expression on DCs. We hypothesized that interleukin 12 (IL-12) expression would be a more accurate marker of functionally mature DCs capable of activating antigen-specific T cells. We used combinations of signaling through CD40, using CD40 ligand trimer (CD40L), and interferon gamma to demonstrate that CD83 expression is necessary but not sufficient for optimal production of IL-12 by DCs. Phenotypically mature DCs could be induced to produce high levels of IL-12 p70 only when provided 2 simultaneous stimulatory signals. By intracellular cytokine detection, we determined that only a subset of cells that express high levels of CD80 and CD83 generate large amounts of IL-12. DCs matured with both signals are superior to DCs stimulated with the individual agents in activating antigen-specific T cell in vitro. These findings have important implications regarding the identification, characterization, and clinical application of functionally mature DCs.


Subject(s)
CD40 Ligand/pharmacology , Dendritic Cells/drug effects , Interferon-gamma/pharmacology , Interleukin-12/metabolism , Adjuvants, Immunologic/metabolism , Antigen Presentation/drug effects , Antigens, CD , B7-1 Antigen/metabolism , B7-1 Antigen/physiology , Biomarkers , Cell Culture Techniques , Cell Differentiation , Cytotoxicity Tests, Immunologic , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Fluorescent Antibody Technique, Direct , Humans , Immunoglobulins/metabolism , Immunoglobulins/physiology , Immunophenotyping , Interleukin-12/immunology , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/physiology , Monocytes/cytology , Protein Subunits , CD83 Antigen
14.
Cancer Res ; 60(14): 3904-8, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10919667

ABSTRACT

Interferons (IFNs) are known to possess potent antitumor properties. Previous studies have indicated that IFNs are capable of modulating the expression of various tumor suppressor genes and oncogenes. In this study, we looked at the effect of IFN-gamma on the neu/HER-2 proto-oncogene in the DU145, LNCaP, and PC-3 prostate cancer cell lines. IFN-gamma inhibited cell proliferation in both DU145 and PC-3 cells in a dose-dependent manner, whereas no inhibition of proliferation was seen in LNCaP cells. Correspondingly, IFN-gamma treatment of DU145 and PC-3 cells resulted in an increased production of the cyclin-dependent kinase inhibitor p21(WAF1), whereas no increase in p21(WAF1) was seen in LNCaP cells. In addition, IFN-gamma induced phosphorylation of signal transducer and activator of transcription (STAT) 1 in DU145 and PC-3 cells, but not in LNCaP cells. Consistent with these findings, we found that IFN-gamma treatment of DU145 and PC-3 cells caused a reduction in neu/HER-2 expression, with no change seen in the LNCaP cell line. Transfection and overexpression of the transcriptional coactivator p300 in PC-3 cells suppressed the reduction in neu/HER-2 expression after IFN-gamma treatment, suggesting a role for p300 in neu/HER-2 expression. The antiproliferative activity and p21(WAF1) production of these cells after IFN-gamma treatment were found to be reduced as well. We propose that the down-regulation of neu/HER-2 by IFN-gamma occurs via the interaction of phosphorylated STAT1 with p300 because IFN-gamma activities requiring phosphorylated STAT1 are reduced in cells overexpressing p300. These findings suggest that neu/HER-2 may play a role in the growth of some prostate cancers and that IFN-gamma may suppress such cancers by down-regulation of neu/HER-2.


Subject(s)
Down-Regulation , Interferon-gamma/genetics , Interferon-gamma/metabolism , Prostatic Neoplasms/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Binding Sites , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Humans , Immunoblotting , Male , Nuclear Proteins/metabolism , Phosphorylation , Plasmids , Precipitin Tests , Prostatic Neoplasms/genetics , Proto-Oncogene Mas , STAT1 Transcription Factor , Trans-Activators/metabolism , Transcription, Genetic , Transfection , Tumor Cells, Cultured , Up-Regulation
15.
J Interferon Cytokine Res ; 19(12): 1351-61, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10638704

ABSTRACT

Interferons (IFN) inhibit the growth of tumor cells by blocking the progression of their cell cycle. Recently, we showed that this cell cycle inhibition correlates with the ability of IFN to upregulate the cyclin-dependent kinase inhibitor p21(WAF1). This, however, is not proof of a causal relationship. Using p21(WAF1)-deficient cells derived from the HCT116 colon adenocarcinoma cell line, we now show that p21(WAF1) is indeed responsible for the antiproliferative effects of the type II IFN, IFN-gamma. IFN-gamma upregulated p21(WAF1) expression in a p53-independent manner, decreased cyclin-dependent kinase 2 activity, and inhibited entry into the S phase of the cell cycle in p21+/+ but not in p21-/- HCT116 cells. We additionally found that the lack of p21(WAF1) expression resulted in an increase in the ability of IFN-gamma to induce apoptosis, as reflected by an earlier induction of DNA fragmentation and caspase 3 activity in p21-/- cell. Our results indicate that p21(WAF1) expression is necessary for IFN-gamma-mediated cell cycle inhibition and suppression of IFN-gamma-induced apoptosis.


Subject(s)
Apoptosis/drug effects , CDC2-CDC28 Kinases , Cell Cycle/drug effects , Cyclins/biosynthesis , Interferon-gamma/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Cyclins/deficiency , Cyclins/genetics , DNA Fragmentation/drug effects , Enzyme Inhibitors/pharmacology , Gene Targeting , Genes, p53 , Humans , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins , Tumor Cells, Cultured
16.
Int J Cancer ; 77(1): 138-45, 1998 Jul 03.
Article in English | MEDLINE | ID: mdl-9639405

ABSTRACT

Type I and type II interferons (IFNs) are known to exert antitumor effects on a variety of tissues and cell types. We have previously shown that the type I IFN IFN alpha induces the expression of the cyclin-dependent kinase inhibitor p21WAF1 and inhibits the cell cycle of the human prostate adenocarcinoma cell line, DU145, that carries mutations in the tumor suppressor gene products p53 and pRB. We now show that the type II IFN IFN gamma similarly induces the expression of p21WAF1 and inhibits the cell cycle of DU145 cells. In addition, we show that while both IFNs exert antiproliferative activity, only IFN gamma induced phenotypic changes in these cells that accompanied the antiproliferative effect. For example, IFN gamma, but not IFN alpha, caused a significant reduction in epidermal growth factor receptor expression as well as an increase in the adhesion molecules intercellular adhesion molecule-1 and integrin alpha3. These phenotypic changes in DU145 cells are suggestive of the acquisition of a non-tumorigenic state. Consistent with these findings, IFN gamma showed a significantly lower invasive ability in in vitro assays using invasion chambers. Thus, IFN gamma inhibits both the cell cycle and the metastatic potential of DU145 cells independent of the p53 and RB status, and our data describe a mechanism for mediating the antitumor capabilities of IFN gamma that bypasses tumor suppressor genes like p53.


Subject(s)
Cell Cycle/drug effects , Cyclins/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Interferon-gamma/pharmacology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21 , Enzyme Induction/drug effects , Humans , Interferon-alpha/pharmacology , Male , Neoplasm Invasiveness , Prostatic Neoplasms/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured
17.
Oncogene ; 16(14): 1885-90, 1998 Apr 09.
Article in English | MEDLINE | ID: mdl-9583686

ABSTRACT

We show, in this study, that type I IFN induction of the cyclin-dependent kinase (cdk) inhibitor p21WAF1 in the human Burkitt lymphoma B cell-line Daudi and ensuing cell cycle arrest correlate with the terminal differentiation of these cells, and is ultimately followed by apoptosis and cell death. The expression of p21WAF1 paralleled the onset of G1 arrest and the reduction of surface IgM expression which was used as a marker of the differentiation response, and the IFN treated cells acquired a typical plasma cell-like morphology. The type II IFN IFNgamma, which does not inhibit the growth of Daudi cells, did not induce the expression of p21WAF1, nor affect the expression of surface IgM. The induction of p21WAF1 which paralleled the inhibition of the phosphorylation of the retinoblastoma protein, pRB, was preceded by the strong reduction in c-myc levels. We propose that the coupled down-regulation of c-myc and induction of p21WAF1 may be crucial to the induction of differentiation and G1 arrest in Daudi cells by type I IFN. Growth arrest and differentiation was followed by apoptosis and cell death, and was accompanied by the induction of the activity of the apoptotic ICE-family protease CPP32. G1 arrest and differentiation followed by apoptotic cell death are characteristics of terminal differentiation. Thus, our data suggest that the induction of p21WAF1 and G1 arrest mediated by type I IFN in Daudi cells is part of terminal differentiation response in these cells, highlighting a role for type I IFN as B cell terminal differentiation factors.


Subject(s)
Apoptosis/drug effects , B-Lymphocytes/pathology , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/biosynthesis , G1 Phase/drug effects , Interferon Type I/pharmacology , Apoptosis/genetics , B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , Burkitt Lymphoma , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/physiology , G1 Phase/genetics , Humans , Tumor Cells, Cultured
18.
Oncogene ; 17(23): 2973-9, 1998 Dec 10.
Article in English | MEDLINE | ID: mdl-9881699

ABSTRACT

Glioblastoma is a highly aggressive form of brain cancer characterized by uncontrolled cell growth resulting from a loss of cell cycle regulation. In this study we determined the antiproliferative effects of interferon gamma (IFNgamma) on the glioblastoma cell lines T98G, SNB-19 and U-373, focusing on the ability of IFNgamma to increase levels of p21WAF1/CIP1, an important negative regulator of cell cycle events. IFNgamma was found to inhibit the growth of all cell lines, with inhibition ranging from 82.2% to 45.4%. Flow cytometry analysis showed that IFNgamma treatment caused a cell cycle delay in the G1 or S phases. The strength of this delay varied, correlating with the degree by which IFNgamma inhibited proliferation of each cell line. IFNgamma treatment increased the production of the cyclin dependent kinase inhibitor (CKI) p21WAF1/ CIP1 in all cell lines, the level and kinetics of production of which correlated with the degree and stage of inhibition of cellular proliferation. Further, immunoprecipitation of p21WAF1/CIP1 in complexes of p21WAF1/CIP1/cyclin-dependent kinase 2 (cdk2)/cyclin showed that the amount of p21WAF1/CIP1 in the complexes and the inhibition of cdk2-cyclin kinase activity correlated with the level of p21WAF1/CIP1 produced in the cells by IFNgamma. These results show that IFNgamma has significant antiproliferative effects on the glioblastoma cell lines and suggest that p21WAF1/CIP1 plays a role in mediating these effects.


Subject(s)
CDC2-CDC28 Kinases , Cyclins/metabolism , Growth Inhibitors/metabolism , Interferon-gamma/metabolism , Cell Cycle/drug effects , Cell Division/drug effects , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/metabolism , Glioblastoma , Growth Inhibitors/pharmacology , Humans , Interferon-gamma/pharmacology , Protein Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured
19.
Oncogene ; 14(10): 1165-70, 1997 Mar 13.
Article in English | MEDLINE | ID: mdl-9121765

ABSTRACT

Prostate cancer, like other types of cancer, is associated with the loss of cell cycle control, resulting in unregulated growth of cells. We report here on the inhibitory effects of interferon alpha (IFN alpha) on the cell cycle of prostate cancer cells, using the human prostate carcinoma cell line DU145 that has mutations in the tumor suppressor genes pRB, p53 and KAI1. IFN alpha inhibited growth and colony formation of DU145 cells and analysis by flow cytometry suggests that IFN alpha inhibited the progression of these cancer cells from the G1 through S phase of the cell cycle. IFN alpha treatment of DU145 cells reduced cyclin dependent kinase 2 (cdk2) activity. In particular, cyclin E dependent cdk2 activity was inhibited by IFN alpha treatment. IFN alpha treatment, however, did not affect the amount of cdk2 bound to cyclin E. Consistent with this data, IFN alpha was able to induce expression of the kinase inhibitor p21 in DU145 cells. Furthermore, IFN treatment increased the amounts of p21 complexed with cdk2 in these cells. These data support a role for p21 in mediating the antiproliferative action of IFN alpha. The induction of p21 and its growth inhibitory effects in DU145 cells appears independent of p53, pRB and KAI1 status.


Subject(s)
Antineoplastic Agents/pharmacology , CDC2-CDC28 Kinases , Cyclins/biosynthesis , Interferon-alpha/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/therapy , Cell Cycle/drug effects , Cell Division/drug effects , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/drug effects , Cyclin-Dependent Kinases/metabolism , Flow Cytometry , Genes, Tumor Suppressor , Humans , Male , Mutation , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism
20.
Biochem Biophys Res Commun ; 223(3): 565-71, 1996 Jun 25.
Article in English | MEDLINE | ID: mdl-8687436

ABSTRACT

We have previously shown that sequence 121-149 of the staphylococcal enterotoxin superantigen SEA plays an important role in superantigen function. A synthetic peptide of this region, SEA(121-149), blocks SEA binding to class II MHC molecules and induces interleukin-1 and tumor necrosis factor production in monocytes. In this study, we further emphasize the structural and functional significance of this region of SEA by showing that the SEA(121-149) peptide induces T cell proliferation in a manner similar to that of SEA. SEA(121-149) reacted with antibodies produced to SEA, and the SEA(121-149) specific antibodies neutralized SEA mitogenic activity. A tetrameric form of SEA(121-149) showed increased binding to antibodies and enhanced T cell activation, consistent with the greater avidity associated with increased valency. These data suggest that the internal domain of SEA corresponding to residues 121-149 plays an important role in superantigen activity.


Subject(s)
Enterotoxins/pharmacology , Epitopes/pharmacology , Superantigens/pharmacology , T-Lymphocytes/immunology , Adult , Amino Acid Sequence , Animals , Antibodies/pharmacology , Enterotoxins/chemistry , Epitopes/chemistry , Humans , Lymphocyte Activation , Mice , Mice, Inbred Strains , Molecular Sequence Data , Neutralization Tests , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Spleen/immunology , Superantigens/chemistry , T-Lymphocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...