Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Free Radic Biol Med ; 147: 231-241, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31883467

ABSTRACT

High fidelity and effective adaptive changes of the cell and tissue metabolism to changing environments require strict coordination of numerous biological processes. Multicellular organisms developed sophisticated signaling systems of monitoring and responding to these different contexts. Among these systems, oxygenated lipids play a significant role realized via a variety of re-programming mechanisms. Some of them are enacted as a part of pro-survival pathways that eliminate harmful or unnecessary molecules or organelles by a variety of degradation/hydrolytic reactions or specialized autophageal processes. When these "partial" intracellular measures are insufficient, the programs of cells death are triggered with the aim to remove irreparably damaged members of the multicellular community. These regulated cell death mechanisms are believed to heavily rely on signaling by a highly diversified group of molecules, oxygenated phospholipids (PLox). Out of thousands of detectable individual PLox species, redox phospholipidomics deciphered several specific molecules that seem to be diagnostic of specialized death programs. Oxygenated cardiolipins (CLs) and phosphatidylethanolamines (PEs) have been identified as predictive biomarkers of apoptosis and ferroptosis, respectively. This has led to decoding of the enzymatic mechanisms of their formation involving mitochondrial oxidation of CLs by cytochrome c and endoplasmic reticulum-associated oxidation of PE by lipoxygenases. Understanding of the specific biochemical radical-mediated mechanisms of these oxidative reactions opens new avenues for the design and search of highly specific regulators of cell death programs. This review emphasizes the usefulness of such selective lipid peroxidation mechanisms in contrast to the concept of random poorly controlled free radical reactions as instruments of non-specific damage of cells and their membranes. Detailed analysis of two specific examples of phospholipid oxidative signaling in apoptosis and ferroptosis along with their molecular mechanisms and roles in reprogramming has been presented.


Subject(s)
Ferroptosis , Phospholipids , Apoptosis , Cell Death , Oxidation-Reduction
2.
Gene Ther ; 9(5): 345-52, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11938454

ABSTRACT

P53-based immunization is an attractive approach to cancer immunotherapy due to the accumulation of p53 protein in tumor, but not in normal cells. However, it was not known whether immune response against self-protein (p53) could be generated in vivo. Mouse dendritic cells (DCs) were transduced with adenoviral construct containing murine full-length wild-type p53 (Ad-p53). Repeated immunizations with these cells protected 60% of mice against challenge with MethA sarcoma cells bearing point mutations in p53 gene. Activation of DCs via ligation of CD40 significantly improved the results of immunization: all mice were protected against MethA sarcoma. The treatment of MethA tumor-bearing mice with activated Ad-p53-transduced DCs showed complete tumor rejection in four out of six mice. The specificity of antitumor immune response was confirmed by CTL assay. The analysis of phenotype and function of DCs demonstrated that the effect of CD40 ligation on these cells was enhanced by their infection with Ad-p53. The level of neutralizing anti-adenovirus antibody was moderately elevated in these mice. No signs of autoimmune reaction were evident during detailed pathological evaluation of treated mice. These data demonstrate that activated Ad-p53-infected DCs are able to break tolerance to this protein and can be used in immunotherapy of cancer.


Subject(s)
Cancer Vaccines/administration & dosage , Dendritic Cells/metabolism , Genes, p53 , Genetic Therapy/methods , Sarcoma, Experimental/therapy , Adenoviridae/genetics , Animals , Cytotoxicity Tests, Immunologic , Female , Genetic Vectors/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Sarcoma, Experimental/immunology , Transduction, Genetic
3.
Int J Cancer ; 94(6): 825-33, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11745485

ABSTRACT

We investigated a new approach to the treatment of advanced stage cancer, a combination of apoptosis-inducing therapy and dendritic cell (DC) administration. MethA sarcoma and C3 tumor containing defined tumor-specific antigens in form of peptides' epitopes were selected as experimental mouse models. Sites of established subcutaneous tumors were gamma-irradiated with 10 Gy 3-5 times with 4-5 day interval. DCs generated from bone marrow of syngeneic mice were injected i.v. and s.c. after each irradiation. A large number of cell tracker-labeled DCs accumulated at the site of the irradiated tumor after s.c. injections. This effect was not observed in non-irradiated tumors. Almost all of these DCs bound GFP-labeled MethA cells in tumor tissue. Interferon-gamma production by splenocytes in response to the tumor-specific MHC class I matched peptides was determined by ELISPOT assays. The combination of gamma-irradiation and DC administration, but not each of the treatments alone resulted in a significant increase in T cell response to the specific, but not to the control peptides. An increased proportion of tumor peptide-specific CD8(+)-cells was found only in that group of mice using staining with tetramers. DCs with defective presentation of antigen via MHC class I or MHC class II pathways were unable to induce peptide-specific T cell response. The combination of gamma-irradiation and DC administration but not each of the treatments alone resulted in a dramatic antitumor effect. A substantial proportion of mice completely rejected their tumors (80% in case of MethA sarcoma and 40% in case of C3 tumor). In the rest of the mice, tumor growth was notably suppressed or completely blocked. These data suggest that the combination of apoptosis-inducing therapy and DC administration may be an attractive approach to the treatment of advanced cancer.


Subject(s)
Apoptosis , Dendritic Cells/immunology , Neoplasms, Experimental/therapy , Animals , Antigen Presentation , Combined Modality Therapy , Female , Gamma Rays , Interferon-gamma/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology
4.
J Immunol ; 167(8): 4458-67, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11591772

ABSTRACT

We investigated the interaction between two elements critical for differentiation of hemopoietic cells, the Notch-1 receptor and the transcription factor NF-kappaB. These factors were studied in hemopoietic progenitor cells (HPC) using Notch-1 antisense transgenic (Notch-AS-Tg) mice. DNA binding of NF-kappaB as well as its ability to activate transcription was strongly decreased in HPC from Notch-AS-Tg mice. NF-kappaB-driven transcriptional activity was completely restored after transduction of the cells with retroviral constructs containing activated Notch-1 gene. HPC from Notch-AS-Tg mice have decreased levels of several members of the NF-kappaB family, p65, p50, RelB, and c-Rel and this is due to down-regulation of the gene expression. To investigate functional consequences of decreased NF-kappaB activity in transgenic mice, we studied LPS-induced proliferation of B cells and GM-CSF-dependent differentiation of dendritic cells from HPC. These two processes are known to be closely dependent on NF-kappaB. B cells from Notch-AS-Tg mice had almost 3-fold lower response to LPS than B cells isolated from control mice. Differentiation of dendritic cells was significantly affected in Notch-AS-Tg mice. However, it was restored by transduction of activated Notch-1 into HPC. Taken together, these data indicate that in HPC NF-kappaB activity is regulated by Notch-1 via transcriptional control of NF-kappaB.


Subject(s)
Hematopoietic Stem Cells/metabolism , Membrane Proteins/metabolism , NF-kappa B/genetics , Receptors, Cell Surface , Transcription Factors , Animals , Antisense Elements (Genetics) , B-Lymphocytes/immunology , Cell Differentiation , Dendritic Cells/cytology , Gene Expression Regulation , Lipopolysaccharides/immunology , Membrane Proteins/genetics , Mice , Mice, Transgenic , Protein Subunits , Receptor, Notch1 , Transcription, Genetic
5.
J Virol ; 75(16): 7621-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11462034

ABSTRACT

A novel technology combining replication- and integration-defective human immunodeficiency virus type 1 (HIV-1) vectors with genetically modified dendritic cells was developed in order to induce T-cell immunity. We introduced the vector into dendritic cells as a plasmid DNA using polyethylenimine as the gene delivery system, thereby circumventing the problem of obtaining viral vector expression in the absence of integration. Genetically modified dendritic cells (GMDC) presented viral epitopes efficiently, secreted interleukin 12, and primed both CD4(+) and CD8(+) HIV-specific T cells capable of producing gamma interferon and exerting potent HIV-1-specific cytotoxicity in vitro. In nonhuman primates, subcutaneously injected GMDC migrated into the draining lymph node at an unprecedentedly high rate and expressed the plasmid DNA. The animals presented a vigorous HIV-specific effector cytotoxic-T-lymphocyte (CTL) response as early as 3 weeks after a single immunization, which later developed into a memory CTL response. Interestingly, antibodies did not accompany these CTL responses, indicating that GMDC can induce a pure Th1 type of immune response. Successful induction of a broad and long-lasting HIV-specific cellular immunity is expected to control virus replication in infected individuals.


Subject(s)
Antigen Presentation/genetics , Dendritic Cells/immunology , HIV Infections/immunology , HIV-1/immunology , Immunity, Cellular/genetics , T-Lymphocytes/immunology , Animals , Cell Communication/genetics , Cell Communication/immunology , Genetic Vectors , HIV Antigens/genetics , HIV Antigens/immunology , HIV Infections/genetics , Humans , Plasmids
6.
J Immunol ; 166(9): 5398-406, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11313376

ABSTRACT

The mechanism of tumor-associated T cell dysfunction remains an unresolved problem of tumor immunology. Development of T cell defects in tumor-bearing hosts are often associated with increased production of immature myeloid cells. In tumor-bearing mice, these immature myeloid cells are represented by a population of Gr-1(+) cells. In this study we investigated an effect of these cells on T cell function. Gr-1(+) cells were isolated from MethA sarcoma or C3 tumor-bearing mice using cell sorting. These Gr-1(+) cells expressed myeloid cell marker CD11b and MHC class I molecules, but they lacked expression of MHC class II molecules. Tumor-induced Gr-1(+) cells did not affect T cell responses to Con A and to a peptide presented by MHC class II. In sharp contrast, Gr-1(+) cells completely blocked T cell response to a peptide presented by MHC class I in vitro and in vivo. Block of the specific MHC class I molecules on the surface of Gr-1(+) cells completely abrogated the observed effects of these cells. Thus, immature myeloid cells specifically inhibited CD8-mediated Ag-specific T cell response, but not CD4-mediated T cell response. Differentiation of Gr-1(+) cells in the presence of growth factors and all-trans retinoic acid completely eliminated inhibitory potential of these cells. This may suggest a new approach to cancer treatment.


Subject(s)
Immune Tolerance/immunology , Myeloid Cells/immunology , Sarcoma, Experimental/immunology , Sarcoma, Experimental/pathology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Separation , Cells, Cultured , Female , Histocompatibility Antigens Class I/biosynthesis , Injections, Subcutaneous , Lymphocyte Activation/immunology , Methylcholanthrene , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/metabolism , Neoplasm Transplantation , Sarcoma, Experimental/chemically induced , Spleen/cytology , Spleen/immunology
7.
Hum Immunol ; 62(4): 332-41, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11295465

ABSTRACT

CD34(+) progenitor cells have previously been shown to be mobilized in patients with squamous cell carcinoma of the head and neck (HNSCC). The present study showed that these CD34(+) cells inhibit the capacity of intratumoral lymphoid cells to become activated in response to stimulation through the TCR/CD3 complex. The mechanisms that could lead to the accumulation of CD34(+) cells within the tumor tissue were assessed. This was accomplished through in vitro studies that determined if HNSCC produce soluble factors that chemoattract CD34(+) cells. The migration of cord blood CD34(+) cells, which were used as a readily available source of progenitor cells, was stimulated by products derived from HNSCC explants and primary HNSCC cultures. This stimulated migration was due to chemotaxis because it was dependent on an increasing gradient of HNSCC-derived products. CD34(+) cells that were isolated from the peripheral blood of HNSCC patients were similarly chemoattracted to the HNSCC-derived products. The majority of the chemotactic activity produced by HNSCC could be attributed to vascular endothelial cell growth factor (VEGF). These studies indicate that HNSCC can chemoattract immune inhibitory CD34(+) progenitor cells through their production of VEGF.


Subject(s)
Antigens, CD34 , Carcinoma, Squamous Cell/metabolism , Cell Movement/physiology , Chemotaxis/physiology , Endothelial Growth Factors/metabolism , Head and Neck Neoplasms/metabolism , Hematopoietic Stem Cells/physiology , Lymphokines/metabolism , Carcinoma, Squamous Cell/pathology , Fetal Blood/cytology , Head and Neck Neoplasms/pathology , Humans , Immunosuppression Therapy , Lymphocytes/immunology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
8.
Cancer Res ; 61(5): 2015-21, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11280761

ABSTRACT

Vascular endothelial growth factor (VEGF) inhibits of the activation of transcription factor nuclear factor-kappaB (NF-kappaB) in hematopoietic progenitor cells (HPCs), and this is associated with alterations in the development of multiple lineages of hematopoietic cells and defective immune induction in tumor-bearing animals. Antibodies to VEGF have been shown to abrogate this effect. The mechanism by which VEGF antagonizes the induction of NF-kappaB was investigated in this study. Using supershift electrophoretic mobility shift analysis, we found that although tumor necrosis factor alpha (TNF-alpha) induced the nuclear translocation and DNA binding of p65-containing complexes, VEGF alone induced nuclear translocation and DNA binding of the complexes containing RelB. These results were confirmed by immunofluorescence confocal microscopy. VEGF effectively blocked TNF-alpha-induced NF-kappaB activation in HPCs from RelB-/- mice, however, similar to the effect observed in HPCs obtained from RelB+/- and RelB+/+ mice. This suggests that RelB is not required for VEGF to inhibit NF-kappaB activation. However, although TNF-alpha induced rapid activation of IkappaB kinase (IKK) as expected, this activity was substantially reduced in the presence of VEGF. This decreased IKK activation correlated with the inhibition of IkappaB alpha phosphorylation and degradation of IkappaB alpha and IkappaB epsilon in HPCs. VEGF alone, however, did not have any effect on phosphorylation of IkappaB alpha or degradation of IkappaB alpha and other inhibitory molecules IkappaB beta, IkappaB epsilon, or Bcl-3. SU5416, a potent inhibitor of the VEGF receptor I (VEGFR1) and VEGFR2 receptor tyrosine kinases, did not abolish the inhibitory effect of VEGF, indicating that the VEGF effect is mediated by a mechanism unrelated to VEGFR1 or VEGFR2 tyrosine kinase activity. Thus, VEGF appears to inhibit TNF-alpha-induced NF-kappaB activation by VEGFR kinase-independent inhibition of IKK. Therapeutic strategies aimed at overcoming VEGF-mediated defects in immune induction in tumor-bearing hosts will need to target this kinase-independent pathway.


Subject(s)
Endothelial Growth Factors/pharmacology , Hematopoietic Stem Cells/drug effects , Lymphokines/pharmacology , NF-kappa B/antagonists & inhibitors , Animals , Cell Nucleus/metabolism , DNA/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiology , Enzyme Activation/drug effects , Female , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/physiology , Humans , I-kappa B Kinase , I-kappa B Proteins/antagonists & inhibitors , I-kappa B Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/physiology , Rats , Rats, Wistar , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Transcription Factor RelB , Transcription Factors/physiology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
9.
Clin Cancer Res ; 7(1): 127-35, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11205900

ABSTRACT

Accumulation of wild-type or mutant p53 protein occurs in approximately 50% of human malignancies. This overexpression may generate antigenic epitopes recognized by CTLs. Because normal cells have undetectable levels of p53, these CTLs are likely to be tumor specific. Here, for the first time, we test the hypothesis that full-length wild-type p53 protein can be used for generation of an immune response against tumor cells with p53 overexpression. T cells obtained from nine HLA-A2-positive cancer patients and three HLA-A2-positive healthy individuals were stimulated twice with dendritic cells (DCs) transduced with an adenovirus wild-type p53 (Ad-p53) construct. Significant cytotoxicity was detected against HLA-A2-positive tumor cells with accumulation of mutant or wild-type p53 but not against HLA-A2-positive tumor cells with normal (undetectable) levels of p53 or against HLA-A2-negative tumor cells. This response was specific and mediated by CD8+ CTLs. These CTLs recognized HLA-A2-positive tumor cells expressing normal levels of p53 protein after their transduction with Ad-p53 but not with control adenovirus. Stimulation of T cells with Ad-p53-transduced DCs resulted in generation of CTLs specific for p53-derived peptide. These data demonstrate that DCs transduced with the wild-type p53 gene were able to induce a specific antitumor immune response. This offers a new promising approach to immunotherapy of cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/blood , Dendritic Cells/immunology , Head and Neck Neoplasms/blood , Lung Neoplasms/blood , T-Lymphocytes, Cytotoxic/immunology , Tumor Suppressor Protein p53/immunology , Adenoviridae/genetics , Adult , Aged , Animals , CD8 Antigens/immunology , Female , Gene Expression , HLA-A2 Antigen/metabolism , Humans , Immunoenzyme Techniques , Immunotherapy , Macroglobulins/immunology , Macroglobulins/metabolism , Male , Mice , Middle Aged , Peptide Fragments/immunology , Peptide Fragments/metabolism , Tumor Cells, Cultured/drug effects , Tumor Suppressor Protein p53/biosynthesis
10.
J Immunol ; 166(1): 678-89, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11123353

ABSTRACT

Defective dendritic cell (DC) function caused by abnormal differentiation of these cells is an important mechanism of tumor escape from immune system control. Previously, we have demonstrated that the number and function of DC were dramatically reduced in cancer patients. This effect was closely associated with accumulation of immature cells (ImC) in peripheral blood. In this study, we investigated the nature and functional role of those ImC. Using flow cytometry, electron microscopy, colony formation assays, and cell differentiation in the presence of different cell growth factors, we have determined that the population of ImC is composed of a small percentage (<2%) of hemopoietic progenitor cells, with all other cells being represented by MHC class I-positive myeloid cells. About one-third of ImC were immature macrophages and DC, and the remaining cells were immature myeloid cells at earlier stages of differentiation. These cells were differentiated into mature DC in the presence of 1 microM all-trans-retinoic acid. Removal of ImC from DC fractions completely restored the ability of the DC to stimulate allogeneic T cells. In two different experimental systems ImC inhibited Ag-specific T cell responses. Thus, immature myeloid cells generated in large numbers in cancer patients are able to directly inhibit Ag-specific T cell responses. This may represent a new mechanism of immune suppression in cancer and may suggest a new approach to cancer treatment.


Subject(s)
Immune Tolerance , Myeloid Cells/immunology , Myeloid Cells/pathology , Neoplasms/immunology , Neoplasms/pathology , Adult , Aged , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cytokines/pharmacology , Dendritic Cells/immunology , Dendritic Cells/pathology , Growth Substances/pharmacology , Humans , Immunophenotyping , Leukocyte Count , Middle Aged , Monocytes/immunology , Monocytes/pathology , Myeloid Cells/drug effects , Myeloid Cells/ultrastructure , Neoplasms/ultrastructure , Tretinoin/pharmacology , Tumor Cells, Cultured
11.
Methods Mol Med ; 64: 3-7, 2001.
Article in English | MEDLINE | ID: mdl-21374244

ABSTRACT

Lymph nodes are the primary sites of T-cell stimulation by dendritic cells (DC). After contact with antigens, DCs migrate to draining lymph nodes from the skin and other tissues (1-3). Investigation of the morphology and function of lymph node DCs may provide important information about the role of these cells in normal and pathological conditions. Therefore, lymph nodes are popular sites for the isolation of dendritic cells. Dendritic cells isolated from lymph nodes represent interdigitating" DCs that are localized in T-dependent regions of lymph nodes. DCs represent about 1% of the total population of lymph node cells. Therefore, in order to perform almost any functional tests, the DC fraction should be enriched. The most practical way to enrich the DC fraction is to use a density gradient. Several gradients-metrizamide (4), Nycodenz (5), and Percoll (6)-have successfully been used for enrichment of DCs obtained from different sources. When isolating DC from lymph nodes, density gradient separation produces a population of DC with a purity of 40-50%. Most contaminating cells are lymphocytes with a small fraction (usually less than 5%) of macrophages. The choice of lymph nodes is dependent on the purpose of the experiment. The most commonly used lymph nodes are axillary, inguinal, and popliteal. DCs can be further enriched using monoclonal antibodies and flow cytometric cell sorting, magnetic beads separation, panning, or cytotoxic elimination with complement. All these methods are based on the negative selection of DCs using anti-T, anti-B, and anti-macrophage antibodies. Since the first step of isolation involves gradient centrifugation, granulocyte contamination is negligible and further purification steps do not require use of anti-granulo-cyte antibodies.

12.
Clin Cancer Res ; 6(5): 1755-66, 2000 May.
Article in English | MEDLINE | ID: mdl-10815894

ABSTRACT

Defective dendritic cell (DC) function has been described previously in cancer patients and tumor-bearing mice. It can be an important factor in the escape of tumors from immune system control. However, the mechanism and clinical significance of this phenomenon remain unclear. Here, 93 patients with breast, head and neck, and lung cancer were investigated. The function of peripheral blood and tumor draining lymph node DCs was equally impaired in cancer patients, consistent with a systemic rather than a local effect of tumor on DCs. The number of DCs was dramatically reduced in the peripheral blood of cancer patients. This decrease was associated with the accumulation of cells lacking markers of mature hematopoietic cells. The presence of these immature cells was closely associated with the stage and duration of the disease. Surgical removal of tumor resulted in partial reversal of the observed effects. The presence of immature cells in the peripheral blood of cancer patients was closely associated with an increased plasma level of vascular endothelial growth factor but not interleukin 6, granulocyte macrophage colony-stimulating factor, macrophage colony-stimulating factor, interleukin 10, or transforming growth factor-beta and was decreased in lung cancer patients receiving therapy with antivascular endothelial growth factor antibodies. These data indicate that defective DC function in cancer patients is the result of decreased numbers of competent DCs and the accumulation of immature cells. This effect may have significant clinical implications.


Subject(s)
Cell Differentiation , Dendritic Cells/pathology , Neoplasms/pathology , Adult , Aged , Antibodies/adverse effects , Antineoplastic Agents/therapeutic use , Carboplatin/administration & dosage , Cell Count , Cytokines/blood , Endothelial Growth Factors/immunology , Flow Cytometry , Growth Substances/blood , Humans , Lymph Nodes/pathology , Lymphokines/immunology , Middle Aged , Neoplasm Staging , Neoplasms/drug therapy , Neoplasms/physiopathology , Paclitaxel/administration & dosage , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
13.
Clin Cancer Res ; 5(10): 2963-70, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10537366

ABSTRACT

Inadequate function of dendritic cells (DCs) in tumor-bearing hosts is one mechanism of tumor escape from immune system control and may compromise the efficacy of cancer immunotherapy. Vascular endothelial growth factor (VEGF), produced by most tumors, not only plays an important role in tumor angiogenesis but also can inhibit the maturation of DCs from hematopoietic progenitors. Here, we investigate a novel combination of antiangiogenic and immunotherapy based on this dual role of VEGF. Two s.c. mouse tumor models were used: D459 cells, expressing mutant human p53; and MethA sarcoma with point mutations in the endogenous murine p53 gene. Therapy with anti-mouse VEGF antibody (10 microg i.p. twice a week over 4 weeks) was initiated when tumors became palpable. Treatment of established tumors with anti-VEGF antibody alone did not affect the rate of tumor growth. However, anti-VEGF antibody significantly improved the number and function of lymph node and spleen DCs in these tumor-bearing animals. To investigate the possible effects of this antibody on the immunotherapy of established tumors, tumor-bearing mice were immunized with DCs pulsed with the corresponding mutation-specific p53 peptides, together with injections of anti-VEGF antibody. Therapy with peptide-pulsed DCs alone resulted in considerable slowing of tumor growth but only during the period of treatment, and tumor growth resumed after the end of the therapy. Combined treatment with peptide-pulsed DCs and anti-VEGF antibody resulted in a prolonged and much more pronounced antitumor effect. This effect was associated with the induction of significant anti-p53 CTL responses only in this group of mice. These data suggest that inhibition of VEGF may be a valuable adjuvant in the immunotherapy of cancer.


Subject(s)
Antibodies/therapeutic use , Dendritic Cells/immunology , Endothelial Growth Factors/antagonists & inhibitors , Lymphokines/antagonists & inhibitors , Neoplasms, Experimental/therapy , Amino Acid Sequence , Animals , Female , Humans , Immunization , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Molecular Sequence Data , Neoplasms, Experimental/immunology , T-Lymphocytes, Cytotoxic/immunology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
J Immunol ; 163(6): 3260-8, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10477595

ABSTRACT

The cytokine FLT3 ligand (FL) enhances dendritic cell (DC) generation and has therefore been proposed as a means to boost antitumor immunity. Vascular endothelial growth factor (VEGF) is produced by a large percentage of tumors and is required for development of tumor neovasculature. We previously showed that VEGF decreases DC production and function in vivo. In this study, we tested the hypothesis that VEGF regulates FL effects on DC generation. In seven experiments, four groups of mice were treated with PBS, VEGF alone (100 ng/h), FL alone (10 microgram/day), or with the combination of FL and VEGF. VEGF and PBS were administered continuously for 14 days via s.c. pumps. FL was given s.c. daily for 9 days, beginning on day 4. Tissues were collected and the number, phenotype, and function of lymph node, splenic, and thymic DCs were analyzed on day 14. As expected, treatment with FL resulted in a marked increase in the number of lymph node and spleen DCs and a smaller increase in thymic DC. Pretreatment of mice with VEGF inhibited these FL effects in lymph nodes and thymus by about 50%, whereas spleen DC numbers were undiminished by VEGF. VEGF treatment in vivo also inhibited the ability of FL to increase the number of hemopoietic precursor cells and the level of maturity exhibited by DC derived from these hemopoietic precursor cells in vitro. VEGF inhibited FL-inducible activation of transcription factor NF-kappaB. These data suggest that VEGF interferes with the ability of FL to promote dendritic cell differentiation from bone marrow progenitor cells in mice and therefore may decrease the therapeutic efficacy of FL in settings where increased numbers of DCs might provide clinical benefits.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/immunology , Endothelial Growth Factors/administration & dosage , Lymphokines/administration & dosage , Membrane Proteins/administration & dosage , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Division/drug effects , Cell Division/immunology , Cells, Cultured , Dendritic Cells/drug effects , Endothelial Growth Factors/physiology , Female , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Infusion Pumps , Injections, Subcutaneous , Ligands , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphokines/physiology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Spleen/cytology , Spleen/immunology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
15.
Clin Exp Immunol ; 117(2): 244-51, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10444254

ABSTRACT

In this study we have tested the concept of using wild-type p53 gene for immunotherapy of cancer. Dendritic cells (DC) were transduced with a human wild-type p53 containing recombinant adenovirus (Ad-p53). About a half of DC transduced with this virus expressed p53 protein by FACS analysis 48 h after infection. Mice immunized twice with Ad-p53 DC developed substantial cytotoxic T lymphocyte (CTL) responses against tumour cells expressing wild-type and different mutant human and murine p53 genes. Very low CTL responses were observed against target cells infected with control adenovirus (Ad-c). Immunization with Ad-p53 provided complete tumour protection in 85% of mice challenged with tumour cells expressing human mutant p53 and in 72.7% of mice challenged with tumour cells with murine mutant p53. Treatment with Ad-p53-transduced DC significantly slowed the growth of established tumours. Thus, DC transduced with wild-type p53 may be a promising new tool for the immunotherapy of cancer.


Subject(s)
Antineoplastic Agents/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Genes, p53/immunology , Transfection/immunology , Adenoviruses, Human/genetics , Adenoviruses, Human/immunology , Animals , Antineoplastic Agents/pharmacology , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Dendritic Cells/transplantation , Dose-Response Relationship, Immunologic , Female , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/immunology , Humans , Immunotherapy, Adoptive/methods , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Neoplasm Transplantation , Sarcoma, Experimental/genetics , Sarcoma, Experimental/immunology , Sarcoma, Experimental/therapy , Tumor Cells, Cultured
16.
Clin Exp Metastasis ; 17(10): 881-8, 1999.
Article in English | MEDLINE | ID: mdl-11089887

ABSTRACT

Patients and animals with GM-CSF-producing tumors have an increased number of mobilized CD34+ progenitor cells within their peripheral blood and tumor tissue. These CD34+ cells are inhibitory to the activity of intratumoral T-cells. The present study used the murine Lewis lung carcinoma (LLC) model to assess mechanisms that could lead to the accumulation of CD34+ cells within the tumor tissue. In vitro analyses showed that LLC tumor explants released chemoattractants for normal femoral CD34+ cells. The LLC tumor cells contributed to the production of this activity since CD34+ cell chemoattractants were also released by cultured LLC cells. Antibody neutralization studies showed that most, although not all, of the chemotactic activity that was produced by LLC cells could be attributed to VEGF. In vivo studies with fluorescent-tagged CD34+ cells showed their accumulation within the tumor tissue, but not within the lungs, spleen or bone marrow, suggesting a selective accumulation within the tumor. Whether or not VEGF could chemoattract CD34+ cells in vivo was measured with a VEGF-containing Matrigel plug assay. Infusion of fluorescent-tagged CD34+ cells into mice after the plugs became vascularized revealed the accumulation of fluorescent-tagged cells within the plugs. However, these CD34+ cells failed to accumulate within the VEGF-containing Matrigel plugs when they were infused together with neutralizing anti-VEGF antibody. Through a combination of in vitro and in vivo analyses, the LLC cells were shown to be capable of chemoattracting CD34+ cells, with most of the tumor-derived chemotactic activity being due to tumor release of VEGF.


Subject(s)
Antigens, CD34/metabolism , Endothelial Growth Factors/metabolism , Hematopoietic Stem Cells/metabolism , Lymphokines/metabolism , Animals , Antibodies/pharmacology , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Endothelial Growth Factors/immunology , Hematopoietic Stem Cells/pathology , Lymphokines/immunology , Mice , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
Blood ; 92(11): 4150-66, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9834220

ABSTRACT

Defective function of dendritic cells (DC) in cancer has been recently described and may represent one of the mechanisms of tumor evasion from immune system control. We have previously shown in vitro that vascular endothelial growth factor (VEGF), produced by almost all tumors, is one of the tumor-derived factors responsible for the defective function of these cells. In this study, we investigated whether in vivo infusion of recombinant VEGF could reproduce the observed DC dysfunction. Continuous VEGF infusion, at rates as low as 50 ng/h (resulting in serum VEGF concentrations of 120 to 160 pg/mL), resulted in a dramatic inhibition of dendritic cell development, associated with an increase in the production of B cells and immature Gr-1(+) myeloid cells. Infusion of VEGF was associated with inhibition of the activity of the transcription factor NF-kappaB in bone marrow progenitor cells. Experiments in vitro showed that VEGF itself, and not factors released by VEGF-activated endothelial cells, affected polypotent stem cells resulting in the observed abnormal hematopoiesis. These data suggest that VEGF, at pathologically relevant concentrations in vivo, may exert effects on pluripotent stem cells that result in blocked DC development as well as affect many other hematopoietic lineages.


Subject(s)
Dendritic Cells/cytology , Endothelial Growth Factors/pharmacology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Lymphokines/pharmacology , Animals , Blood Cell Count , Cell Differentiation/drug effects , Dendritic Cells/drug effects , Dendritic Cells/physiology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , NF-kappa B/physiology , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
18.
J Immunol ; 161(9): 4842-51, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9794417

ABSTRACT

Langerhans cells (LC), the APCs in the skin, serve as a model for investigation of dendritic cell (DC) function in tissues. DC play a crucial role in the generation of antitumor immune responses. In this study, we investigated the effect of the presence of tumor in vivo on the ability of LC to take up Ag, migrate to draining lymph nodes, and stimulate primary T cell responses. In two animal models, these functions were substantially inhibited. This effect was not restricted to LC located in the skin near a tumor but was also seen at sites distant from the tumor. The duration of tumor exposure, and not its ultimate size, were found to be important, suggesting that tumors could be inhibiting the maturation of LC rather than directly inhibiting their function. Model experiments with radiation chimeras supported this hypothesis. To investigate the potential role of vascular endothelial growth factor (VEGF) in these effects we used anti-VEGF-neutralizing Ab to treat animals bearing tumors. Treatment with the Ab at a dose of 10 microg i.p. per mouse, twice a week for 4 wk, significantly improved the number and function of LC as measured by their ability to migrate to lymph nodes and stimulate primary T cell responses, even at doses that do not affect the growth of these established poorly immunogenic tumors. Thus, inhibition of VEGF signaling may improve DC function in tumor-bearing hosts and possibly serve to improve the efficacy of cancer immunotherapy.


Subject(s)
Endothelial Growth Factors/physiology , Hematopoiesis , Langerhans Cells/pathology , Lymphokines/physiology , Sarcoma, Experimental/immunology , Skin/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antigen Presentation , Antigens, Neoplasm/immunology , Bone Marrow/pathology , Cell Count , Cell Movement , Chemotactic Factors/physiology , Dendritic Cells/immunology , Dendritic Cells/pathology , Endothelial Growth Factors/antagonists & inhibitors , Female , Fluorescein-5-isothiocyanate/pharmacokinetics , Fluorescent Dyes/pharmacokinetics , Immunization, Passive , Immunologic Deficiency Syndromes/etiology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphokines/antagonists & inhibitors , Mice , Mice, Inbred A , Mice, Inbred BALB C , Neoplasm Transplantation , Radiation Chimera , Sarcoma, Experimental/pathology , Sarcoma, Experimental/therapy , Skin/pathology , Specific Pathogen-Free Organisms , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
19.
J Immunol ; 160(3): 1224-32, 1998 Feb 01.
Article in English | MEDLINE | ID: mdl-9570538

ABSTRACT

Vascular endothelial growth factor (VEGF), produced by almost all tumor cells, affects the ability of hemopoietic progenitor cells (HPC) to differentiate into functional dendritic cells (DC) during the early stages of their maturation. In this study we demonstrate specific binding of VEGF to HPC. This binding was efficiently competed by placenta growth factor (PIGF), a ligand reportedly specific for the Flt-1 receptor. The number of binding sites for VEGF decreased during DC maturation in vitro associated with decreased levels of mRNA for Flt-1. VEGF significantly inhibited nuclear factor-kappa B (NF-kappa B)-dependent activation of reporter gene transcription during the first 24 h in culture. The presence of VEGF significantly decreased the specific DNA binding of NF-kappa B as early as 30 min after induction with TNF-alpha. This was followed on days 7 to 10 by decreases in the mRNA for RelB and c-Rel, two subunits of NF-kappa B. Blockade of NF-kappa B activity in HPC at early stages of differentiation with an adenovirus expressing a dominant I kappa B inhibitor of NF-kappa B reproduced the pattern of effects observed with VEGF. Thus, NF-kappa B plays an important role in maturation of HPCs to DC, and VEGF activation of the Flt-1 receptor is able to block the activation of NF-kappa B in this system. Blockade of NF-kappa B activation in HPCs by tumor-derived factors may therefore be a mechanism by which tumor cells can directly down-modulate the ability of the immune system to generate effective antitumor immune responses.


Subject(s)
Dendritic Cells/cytology , Endothelial Growth Factors/physiology , Hematopoietic Stem Cells/metabolism , I-kappa B Proteins , Lymphokines/physiology , NF-kappa B/antagonists & inhibitors , Antigens, CD34/analysis , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell-Free System/immunology , Cells, Cultured , DNA-Binding Proteins/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Endothelial Growth Factors/metabolism , Fetal Blood , Hematopoietic Stem Cells/drug effects , Humans , Lymphokines/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Binding/immunology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Signal Transduction/immunology , Transcription, Genetic/drug effects , Transcription, Genetic/immunology , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
20.
Clin Cancer Res ; 3(3): 483-90, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9815709

ABSTRACT

We evaluated T-cell responses to mitogens and to defined antigens in breast cancer patients. Significant defects in responses to tetanus toxoid and influenza virus were observed in patients with advanced-stage breast cancer. To define whether these defects were associated with a defect in antigen presentation [dendritic cells (DCs)] or effector function (T cells), these cells were studied separately. Purified DCs from 32 patients with breast cancer demonstrated a significantly decreased ability to stimulate control allogeneic T cells, but stimulation of patient T cells with either control allogeneic DCs or immobilized anti-CD3 antibody resulted in normal T-cell responses, even in patients with stage IV tumors. These data suggest that reduced DC function could be one of the major causes of the observed defect in cellular immunity in patients with advanced breast cancer. We then tested whether stem cells from these patients could give rise to functional DCs after in vitro growth with granulocyte/macrophage colony-stimulating factor and interleukin 4. Normal levels of control allogeneic and tetanus toxoid-dependent T-cell proliferation were observed when DCs obtained from precursors were used as stimulators. Those cells also induced substantially higher levels of influenza virus-specific CTL responses than mature DCs from the peripheral blood of these patients, although responses did not quite reach control values. Thus, defective T-cell function in patients with advanced breast cancer can be overcome by stimulation with DCs generated from precursors, suggesting that these cells may better serve as autologous antigen carriers for cancer immunotherapy than mature peripheral blood DCs.


Subject(s)
Breast Neoplasms/immunology , Dendritic Cells/immunology , T-Lymphocytes/immunology , Breast Neoplasms/blood , Breast Neoplasms/pathology , Female , HLA-D Antigens/analysis , Histocompatibility Antigens Class I/analysis , Humans , Lymphocyte Activation , Neoplasm Staging
SELECTION OF CITATIONS
SEARCH DETAIL
...