Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Chonnam Medical Journal ; : 1-7, 2015.
Article in English | WPRIM | ID: wpr-133657

ABSTRACT

Although the introduction of stem cell transplantation and novel agents has improved survival, multiple myeloma (MM) is still difficult to cure. Alternative approaches are clearly needed to prolong the survival of patients with MM. Dendritic cell (DC) therapy is a very promising tool immunologically in MM. We developed a method to generate potent DCs with increased Th1 polarization and migration ability for inducing strong myeloma-specific cytotoxic T lymphocytes. In this review, we discuss how the efficacy of cancer immunotherapy using DCs can be improved in MM.


Subject(s)
Humans , Dendritic Cells , Immunotherapy , Multiple Myeloma , Stem Cell Transplantation , T-Lymphocytes, Cytotoxic
2.
Chonnam Medical Journal ; : 1-7, 2015.
Article in English | WPRIM | ID: wpr-133656

ABSTRACT

Although the introduction of stem cell transplantation and novel agents has improved survival, multiple myeloma (MM) is still difficult to cure. Alternative approaches are clearly needed to prolong the survival of patients with MM. Dendritic cell (DC) therapy is a very promising tool immunologically in MM. We developed a method to generate potent DCs with increased Th1 polarization and migration ability for inducing strong myeloma-specific cytotoxic T lymphocytes. In this review, we discuss how the efficacy of cancer immunotherapy using DCs can be improved in MM.


Subject(s)
Humans , Dendritic Cells , Immunotherapy , Multiple Myeloma , Stem Cell Transplantation , T-Lymphocytes, Cytotoxic
3.
Chonnam Medical Journal ; : 1-7, 2015.
Article in English | WPRIM | ID: wpr-788307

ABSTRACT

Although the introduction of stem cell transplantation and novel agents has improved survival, multiple myeloma (MM) is still difficult to cure. Alternative approaches are clearly needed to prolong the survival of patients with MM. Dendritic cell (DC) therapy is a very promising tool immunologically in MM. We developed a method to generate potent DCs with increased Th1 polarization and migration ability for inducing strong myeloma-specific cytotoxic T lymphocytes. In this review, we discuss how the efficacy of cancer immunotherapy using DCs can be improved in MM.


Subject(s)
Humans , Dendritic Cells , Immunotherapy , Multiple Myeloma , Stem Cell Transplantation , T-Lymphocytes, Cytotoxic
4.
Journal of Korean Medical Science ; : 237-246, 2013.
Article in English | WPRIM | ID: wpr-25352

ABSTRACT

SUV39H1 is a histone 3 lysine 9 (H3K9)-specific methyltransferase that is important for heterochromatin formation and the regulation of gene expression. Chaetocin specifically inhibits SUV39H1, resulted in H3K9 methylation reduction as well as reactivation of silenced genes in cancer cells. Histone deacetylase (HDAC) inhibitors inhibit deacetylases and accumulate high levels of acetylation lead to cell cycle arrest and apoptosis. In this study, we demonstrated that treatment with chaetocin enhanced apoptosis in human leukemia HL60, KG1, Kasumi, K562, and THP1 cells. In addition, chaetocin induced the expression of cyclin-dependent kinase inhibitor 2B (p15), E-cadherin (CDH1) and frizzled family receptor 9 (FZD9) through depletion of SUV39H1 and reduced H3K9 methylation in their promoters. Co-treatment with chaetocin and HDAC inhibitor trichostatin A (TSA) dramatically increased apoptosis and produced greater activation of genes. Furthermore, this combined treatment significantly increased loss of SUV39H1 and reduced histone H3K9 trimethylation responses accompanied by increased acetylation. Importantly, co-treatment with chaetocin and TSA produced potent antileukemic effects in leukemia cells derived from patients. These in vitro findings suggest that combination therapy with SUV39H1 and HDAC inhibitors may be of potential value in the treatment of leukemia.


Subject(s)
Adolescent , Adult , Aged , Humans , Male , Middle Aged , Young Adult , Acetylation/drug effects , Apoptosis/drug effects , Cadherins/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p15/metabolism , DNA Methylation/drug effects , Enzyme Inhibitors/therapeutic use , Frizzled Receptors/metabolism , Gene Expression Regulation/drug effects , HL-60 Cells , Histone Deacetylase Inhibitors/therapeutic use , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histones/genetics , Hydroxamic Acids/therapeutic use , K562 Cells , Leukemia/drug therapy , Leukemia, Myeloid, Acute/genetics , Piperazines/therapeutic use , Promoter Regions, Genetic
5.
Korean Journal of Hematology ; : 17-27, 2012.
Article in English | WPRIM | ID: wpr-720221

ABSTRACT

Cellular therapy with dendritic cells (DCs) is emerging as a useful immunotherapeutic tool to treat multiple myeloma (MM). DC-based idiotype vaccination was recently suggested to induce idiotype-specific immune responses in MM patients. However, the clinical results so far have been largely disappointing, and the clinical effectiveness of such vaccinations in MM still needs to be demonstrated. DC-based therapies against MM may need to be boosted with other sources of tumor-associated antigens, and potent DCs should be recruited to increase the effectiveness of treatment. DCs with both high migratory capacity and high cytokine production are very important for effective DC-based cancer vaccination in order to induce high numbers of Th1-type CD4+ T cells and CD8+ cytotoxic T lymphocytes. The tumor microenvironment is also important in the regulation of tumor cell growth, proliferation, and the development of therapeutic resistance after treatment. In this review, we discuss how the efficacy of DC vaccination in MM can be improved. In addition, novel treatment strategies that target not only myeloma cells but also the tumor microenvironment are urgently needed to improve treatment outcomes.


Subject(s)
Humans , Dendritic Cells , Immunotherapy , Multiple Myeloma , T-Lymphocytes , T-Lymphocytes, Cytotoxic , Tumor Microenvironment , Vaccination
6.
Experimental & Molecular Medicine ; : 60-67, 2012.
Article in English | WPRIM | ID: wpr-211717

ABSTRACT

Anterior gradient-2 (AGR2) promotes tumor growth, cell migration, and cellular transformation, and is one of the specific mRNA markers for circulating tumor cells in patients with gastrointestinal cancer. We investigated the feasibility of AGR2 as a potent antigen for tumor immunotherapy against colorectal cancer (CRC) cells using dendritic cells (DCs) transduced with a recombinant adenovirus harboring the AGR2 gene (AdAGR2). DCs transduced with a recombinant adenovirus encoding the AGR2 gene (AdAGR2/DCs) were characterized. These genetically-modified DCs expressed AGR2 mRNA as well as AGR2 protein at a multiplicity of infection of 1,000 without any significant alterations in DC viability and cytokine secretion (IL-10 and IL-12p70) compared with unmodified DCs as a control. In addition, AdAGR2 transduction did not impair DC maturation, but enhanced expression of HLA-DR, CD80, and CD86. AdAGR2/DCs augmented the number of IFN-gamma-secreting T-cells and elicited potent AGR2-specific cytotoxic T lymphocytes capable of lysing AGR2-expressing CRC cell lines. These results suggest that AGR2 act as a potentially important antigen for immunotherapy against CRC in clinical applications.


Subject(s)
Humans , Adenoviridae , Antigen Presentation/genetics , Antigens, Neoplasm/immunology , Carcinoma/therapy , Cell Line, Tumor , Colorectal Neoplasms/therapy , Cytotoxicity, Immunologic/genetics , Dendritic Cells/immunology , Immunotherapy, Adoptive , Interferon-gamma/metabolism , Lymphocyte Activation/genetics , Proteins/genetics , T-Lymphocytes, Cytotoxic/immunology , Transduction, Genetic , Transgenes/genetics , Biomarkers, Tumor/immunology
7.
Experimental & Molecular Medicine ; : 407-419, 2010.
Article in English | WPRIM | ID: wpr-27762

ABSTRACT

Dendritic cells (DCs) play a role in natural killer (NK) cell activation, while NK cells are also able to activate and mature DCs. Toll-like receptors (TLRs) on the surface of DCs and NK cells induce the maturation and activation of these cells when engaged with their cognate ligand. We investigated to generate potent DCs by maturation with NK cells in the presence of TLR agonist in vitro and tested the efficacy of these DC vaccinations in mouse colon cancer model. The optimal ratios of DCs versus NK cells were 1:1 to 1:2. Immature DCs were mature with NK cells in the presence of lipopolysaccharide, which is TLR4 agonist, and further addition of IL-2 induced phenotypically and functionally mature bone marrow-derived DCs. These potent DCs exhibited not only high expression of several costimulatory molecules and high production of IL-12p40 and IL-12p70, but also high allogeneic T cells stimulatory capacity, and the induction of the high activities to generate tumor-specific CTLs. Consistently, vaccination with these DCs efficiently inhibited CT-26 tumor growth in mouse colon cancer model when compared to other vaccination strategies. Interestingly, combination therapy of these DC-based vaccines and with low-dose cyclophosphamide showed dramatic inhibition effects of tumor growth. These results suggest that the DCs maturated with NK cells in the presence of TLR agonist are potent inducer of antitumor immune responses in mouse model and may provide a new source of DC-based vaccines for the development of immunotherapy against colon cancer.


Subject(s)
Animals , Female , Mice , Cancer Vaccines/immunology , Carcinoma/immunology , Cell Line, Tumor , Cells, Cultured , Colonic Neoplasms/immunology , Dendritic Cells/drug effects , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Lipopolysaccharides/pharmacology , Mice, Inbred BALB C , Toll-Like Receptor 4/agonists , Toll-Like Receptors/agonists
8.
Cancer Research and Treatment ; : 175-180, 2007.
Article in English | WPRIM | ID: wpr-127959

ABSTRACT

PURPOSE: Calcium ionophore (CI) is used to generate dendritic cells (DCs) from progenitor cells, monocytes, or leukemic cells. The aim of this study was to determine the optimal dose of CI and the appropriate length of cell culture required for acute myeloid leukemia (AML) cells and to evaluate the limitations associated with CI. MATERIALS AND METHODS: To generate leukemic DCs, leukemic cells (4 x 10(6) cells) from six AML patients were cultured with various concentrations of CI and/or IL-4 for 1, 2 or 3 days. RESULTS: Potent leukemic DCs were successfully generated from all AML patients, with an average number of 1.2 x 10(6) cells produced in the presence of CI (270 ng/ml) for 2 days. Several surface molecules were clearly upregulated in AML cells supplemented with CI and IL-4, but not CD11c. Leukemic DCs cultured with CI had a higher allogeneic T cell stimulatory capacity than untreated AML cells, but the addition of IL-4 did not augment the MLR activity of these cells. AML cells cultured with CI in the presence or absence of IL-4 showed increased levels of apoptosis in comparison to primary cultures of AML cells. CONCLUSION: Although CI appears to be advantageous in terms of time and cost effectiveness, the results of the present study suggest that the marked induction of apoptosis by CI limits its application to the generation of DCs from AML cells.


Subject(s)
Humans , Apoptosis , Calcium , Cell Culture Techniques , Cost-Benefit Analysis , Dendritic Cells , Interleukin-4 , Leukemia, Myeloid, Acute , Monocytes , Stem Cells
9.
Korean Journal of Hematology ; : 186-193, 2006.
Article in Korean | WPRIM | ID: wpr-720722

ABSTRACT

BACKGROUND: In multiple myeloma (MM), the idiotype (ID) determinant of the paraprotein has been used for immunotherapy using dendritic cells (DCs). However, ID-specific immune responses showed limited clinical responses after the Id vaccination. Therefore, an alternative approach using DCs pulsed with other tumor antigens is required. METHODS: We investigated the possibility of immunotherapy for MM using myeloma cell line-specific cytotoxic T lymphocytes (CTLs), that were stimulated in vitro by monocyte-derived DCs pulsed with the myeloma cell line ysates. CD14+ cells isolated from the peripheral blood of HLA-A0201+ healthy donors were cultured in the presence of GM-CSF and IL-4. On day 6, the immature DCs were pulsed with the myeloma cell line lysates (IM-9: HLA0201+ and ARH-77: HLA0201+), and then maturation of DCs was induced by the addition of TNF- alpha for 2 days. CTL lines were generated by a 2 time stimulation with DCs to the autologous CD3+ T cells. RESULTS: DCs pulsed with myeloma cell lysates showed the production of IL-12p70, but less than that of unpulsed DCs. CTLs lines stimulated with the DCs pulsing, for the myeloma cell line lysates, showed potent cytotoxic activities against autologous target cells, but not against HLA-A2-cell lines (RPMI-8226). Mature DCs pulsed with the myeloma cell line lysates showed a higher stimulatory capacity for autologous CTL when compared with mature non-pulsed DCs. CONCLUSION: These results suggest that DCs pulsed with the myeloma cell line lysates can generate potent myeloma cell line-specific CTLs for the myeloma cell-based immunotherapeutic approach in MM.


Subject(s)
Humans , Antigens, Neoplasm , Cell Line , Dendritic Cells , Granulocyte-Macrophage Colony-Stimulating Factor , Immunotherapy , Interleukin-4 , Multiple Myeloma , T-Lymphocytes , T-Lymphocytes, Cytotoxic , Tissue Donors , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL