Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Immunother ; 23(1): 168-76, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10687150

ABSTRACT

Immunization with tumor-specific-associated antigen--pulsed dendritic cells has proved to be efficacious in various animal models and is being evaluated for the treatment of cancer in humans. Use of dendritic cells pulsed with specific peptides or transfected with tumor-associated antigen genes has been a focused area of investigation for inducing potent tumor and viral immune responses. In this study, the authors demonstrate transgene expression, including the lacZ and MART-1 genes, in dendritic cells infected with adenoviral constructs. These transiently transduced dendritic cells, derived from melanoma patients' monocytes cultured with granulocyte-macrophage colony-stimulating factor and interleukin-4, express the transgene and can stimulate patients' CD8+ T cells to elicit an antitumor immune response comparable to dendritic cells loaded with a defined peptide. These cytotoxic T lymphocytes were able to recognize both known and unknown tumor-associated antigen epitopes and exhibited cytolytic activity against HLA-matched tumor cells expressing the antigen. The ability to induce tumor-specific cytotoxic T lymphocytes in vitro using gene-modified dendritic cells that transiently express tumor-associated antigens demonstrates the potential use of these antigen-presenting cells for developing in vivo cancer vaccines.


Subject(s)
Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Melanoma/immunology , Neoplasm Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Adenoviruses, Human , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Cells, Cultured , Dendritic Cells/cytology , Gene Expression , Genetic Vectors , Humans , Immunotherapy, Adoptive/methods , MART-1 Antigen , Melanoma/blood , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Peptides/genetics , Peptides/immunology , Transgenes , Tumor Cells, Cultured
2.
J Immunother ; 22(3): 229-36, 1999 May.
Article in English | MEDLINE | ID: mdl-10335482

ABSTRACT

Immunization with tumor-associated antigen pulsed dendritic cells (DC) has been shown to elicit both protective and therapeutic antitumor immunity in a variety of animal models and is currently being investigated for the treatment of cancer patients in clinical trials. In this study we show that DC can be generated from peripheral blood mononuclear cells of healthy donors as well as breast and melanoma cancer patients using granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-13 (IL-13) and that these DC have many of the same characteristics as DC differentiated using GM-CSF and IL-4. The DC generated in GM-CSF and IL-13 are CD14- and express high levels of the cell surface markers CD86, HLA-DR, and CD58, as do DC generated in GM-CSF and IL-4. The purity and yield of both DC populations are not significantly different. Furthermore, both populations of DC are effective at presentation of alloantigen as determined in a mixed lymphocyte response, and both are able to process and present soluble tetanus toxoid antigen to CD4+ T cells. Because we are interested in the generation of DC for antigen-specific cytotoxic T lymphocyte (CTL) generation, we compared the ability of peptide-pulsed DC differentiated in GM-CSF and IL-4 versus GM-CSF and IL-13 for the generation of influenza and MART-1 specific CTL. Both populations of DC induced CD3+ CD8+ CD4- and CD56- CTL, which could lyse the appropriate targets in an antigen-specific manner. Finally, both GM-CSF and IL-4 DC and GM-CSF and IL-13 DC yielded similar beta galactosidase expression levels after transduction with recombinant adenovirus containing the LacZ gene. These results suggest that DC generated in GM-CSF and IL-13 may be useful for immunotherapy and gene therapy protocols.


Subject(s)
Dendritic Cells/immunology , Genetic Therapy , Interleukin-13/pharmacology , Interleukin-4/pharmacology , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm , Breast Neoplasms/blood , Breast Neoplasms/immunology , Cell Differentiation , Dendritic Cells/cytology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunophenotyping , Immunotherapy , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Lymphocyte Culture Test, Mixed , MART-1 Antigen , Melanoma/blood , Melanoma/immunology , Neoplasm Proteins/immunology
3.
Cancer Gene Ther ; 5(4): 236-46, 1998.
Article in English | MEDLINE | ID: mdl-9694075

ABSTRACT

Immunization with specific tumor-associated antigen (Ag) (TAA)-pulsed dendritic cells (DC) has proven to be efficacious in a variety of animal models and is being investigated for the treatment of cancer patients. Use of DC pulsed with specific peptides or transfected with TAA genes has been a focused area of investigation for the induction of potent tumor and viral immune responses. In this study we demonstrate transgene expression, including expression of the MART-1 gene, in DC transfected with plasmid DNA and cationic liposome complexes. These transiently transfected DC, derived from healthy donor monocytes cultured with granulocyte macrophage colony-stimulating factor and interleukin-4, express the transgene and can stimulate naive CD8+ T cells to elicit an antitumor immune response. These cytotoxic T lymphocytes (CTL) were capable of recognizing both known and unknown TAA epitopes and were able to exhibit cytolytic activity against human histocompatibility leukocyte Ag-matched tumor cells expressing the Ag. In addition to their cytolytic function, the CTL displayed an oligoclonal T-cell receptor repertoire, indicating that the presented Ag induced alterations in the T-cell population. The ability to induce tumor-specific CTL in vitro using gene-modified DC transiently expressing TAAs demonstrates the potential use of these Ag-presenting cells to generate future in vivo cancer vaccine strategies.


Subject(s)
Carcinoembryonic Antigen/genetics , Dendritic Cells/physiology , Neoplasm Proteins/genetics , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinoembryonic Antigen/metabolism , Cells, Cultured , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dependovirus/genetics , Genes, MHC Class I , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interleukin-4/pharmacology , Liposomes , MART-1 Antigen , Major Histocompatibility Complex , Monocytes , Neoplasm Proteins/metabolism , Receptors, Antigen, T-Cell/drug effects , Receptors, Antigen, T-Cell/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection , Transgenes
4.
J Immunother ; 21(1): 17-26, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9456432

ABSTRACT

The human carcinoembryonic antigen (CEA), which is expressed in several cancer types is a potential target for antigen-specific immunotherapy. In this study, we show that dendritic cells (DC) pulsed with an HLA class I restricted CEA cytotoxic T lymphocyte (CTL) peptide epitope can stimulate T cells to kill CEA peptide loaded T2 target cells as well as CEA expressing tumor lines in the presence of interleukin-7 (IL-7) in an HLA-restricted manner. This has been demonstrated for carcinoma patients as well as healthy donors. The DC-CEA + IL-7 stimulated cultures contained predominantly CD3+CD8+CD56- cells indicative of MHC class I restricted CTL. In addition, DC-CEA + IL-7 stimulated cells showed higher levels of CD69 expression compared with cells stimulated with IL-7 alone, implying an activated phenotype. When the T-cell receptor (TCR) from CTL cultures stimulated with DC-CEA + IL-7 was analyzed, an oligoclonal pattern of expression was found for certain V beta subfamilies compared with the polyclonal patterns shown by IL-7 or phytohemagglutinin stimulated T cells from the same donors. This TCR restriction appeared to be maintained and enhanced after additional rounds of restimulation with DC-CEA + IL-7. The association between cytotoxicity and TCR restriction suggests that TCR analysis may be useful as an in vitro indicator to monitor alterations in the T-cell population in response to antigen-specific immunotherapies.


Subject(s)
Carcinoembryonic Antigen/immunology , Dendritic Cells/immunology , Interleukin-7/pharmacology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Antigen-Presenting Cells , Flow Cytometry , HLA-A2 Antigen/immunology , Histocompatibility Antigens Class I/immunology , Humans , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/immunology , Interleukin-2/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/immunology , Phytohemagglutinins/pharmacology , Receptors, Antigen, T-Cell/analysis , Tumor Cells, Cultured
5.
Adv Exp Med Biol ; 417: 519-24, 1997.
Article in English | MEDLINE | ID: mdl-9286413

ABSTRACT

Antigen specific cytotoxic T lymphocytes (CTL) are being studied for their potential immunotherapeutic benefit in the treatment of cancer. Carcinoembryonic antigen (CEA) is an oncofetal protein best known for its overexpression in the majority of colorectal, gastric, pancreatic, non small cell lung, and breast carcinomas. We are using dendritic cells (DC) pulsed with the CEA CTL peptide epitope to generate CEA specific CTL. DC from HLA A2+ donors were isolated by culturing plastic adherent PBMC in GMCSF and IL4 for 7 days. As expected these DC expressed the relevant cell surface molecules including HLA DR, CD58, CD80, and CD86. The DC were stripped of their endogenous peptides, pulsed with the A2 restricted CEA peptide, irradiated and used to stimulate autologous CD8+ T cells in the presence of IL7. Using this approach we have been able to generate CEA specific CTL from the PBMC of breast and pancreatic carcinoma patients as well as HLA A2+ tumor cells expressing the CEA antigen. This data is being used to support a phase I active immunotherapy clinical protocol using DC pulsed with CEA peptide to treat patients with metastatic malignancies expressing CEA.


Subject(s)
Carcinoembryonic Antigen/immunology , Dendritic Cells/immunology , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Antigen Presentation , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , HLA-A2 Antigen/metabolism , Humans , In Vitro Techniques , Interleukin-4/pharmacology , Leukocytes, Mononuclear/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy
7.
J Autoimmun ; 8(2): 209-20, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7612149

ABSTRACT

Insulin dependent diabetes mellitus (IDDM) in the non-obese diabetic (NOD) mouse is the result of a cellular mediated autoimmune event that destroys pancreatic islet beta cells. This destruction is characterized by a progressive lymphocytic infiltration into the islets as well as circulating autoantibodies and T cells reactive with islet antigens. To gain a better understanding of the cells responsible for islet destruction we isolated lymphocytes from the islets of prediabetic NOD mice and conducted a comparative phenotypic analysis with the analogous subpopulations of lymphocytes isolated from peripheral blood and lymph node (LN) of the same mice. CD3+ cells were analysed for T cell receptor (TcR); cell bearing gamma delta TcR were consistently observed at a higher frequency in the infiltrating T cells than in the periphery. Lymphocytes were also characterized for the expression of CD4 and CD8 T cell markers and, within each population, for the expression of activation markers (CD25, CD69) and adhesion markers (CD51, CD54, CD11b, CD49e, L-selectin). Significantly increased levels of CD4+CD8+ double-positive and CD4-CD8- double-negative T cell populations were observed in the infiltrating lymphocytes as compared with peripheral lymphocytes. In addition, within both CD4 and CD8 subpopulations isolated from islet infiltrates, CD11b+ and CD49e+ cells were increased with respect to the same subset of cells isolated from the periphery. In contrast, the level of cells that expressed L-selectin was significantly higher in the periphery for both CD4+ and CD8+ cells than for infiltrating cells. These data describe the phenotype of islet reactive T cells in the NOD mouse and identify possible targets for therapeutic intervention.


Subject(s)
Autoimmune Diseases/immunology , Cell Adhesion Molecules/analysis , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Islets of Langerhans/pathology , Lymphocyte Activation , Lymphocyte Subsets/pathology , Mice, Inbred NOD/immunology , Receptors, Antigen, T-Cell/analysis , Animals , Antigens, CD/analysis , Antigens, Differentiation, B-Lymphocyte/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , Autoimmune Diseases/blood , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Biomarkers , Cell Movement , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Female , Immunity, Cellular , Islets of Langerhans/immunology , Lymphocyte Count , Lymphocyte Subsets/immunology , Mice
8.
Transplantation ; 56(3): 633-8, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8212161

ABSTRACT

Diabetic (B6) (IE-) mice treated with a depleting regimen of anti-CD4 monoclonal antibody at the time of transplantation with A/J (IEK) islets of Langerhans showed indefinite acceptance of their islet allograft, as evidenced by persistent normoglycemia. To address the mechanisms involved in such anti-CD4 induced transplantation tolerance we studied potentially IE-reactive V beta 11+ T cells from the tolerant allografted mice. Following complete repopulation of the CD4+ cells, both the CD4+V beta 11+ and CD8+V beta 11+ T cell subsets of the transplanted mice were unresponsive to anti-V beta 11 specific crosslinking. In contrast, lymphocytes tested within the first ten days following transplant were responsive to anti-V beta 11 specific crosslinking; this response decreased as a function of time and reached background levels by day 120 posttransplant. Sorting experiments indicated that the response of lymphocytes to anti-V beta 11 specific crosslinking seen during the initial 120 days posttransplant was confined to the peripheral CD8+ cells; the repopulating CD4+V beta 11+ T cells were unresponsive. In addition, administration of r-IL-2 at the time of transplantation induced rejection in anti-CD4-treated animals, again indicating that the peripheral CD8+ cells could respond shortly after transplant if provided with appropriate help. The decreasing response of CD8+ T cells from transplanted animals to anti-V beta 11 stimulation was inversely correlated with the rate of migration of cells from the thymus to the periphery, implying that new thymic migrant V beta 11+ cells, both CD4+ and CD8+, were rendered anergic upon encountering peripheral alloantigen. These data suggest the possibility that recent thymic migrants are rendered anergic upon encountering antigen in the periphery, a simple model to serve as a "fail-safe" mechanism to prevent autoreactivity.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD4 Antigens/immunology , T-Lymphocytes/immunology , Animals , CD8 Antigens/analysis , Cell Movement , Cell Separation , Clonal Anergy/immunology , Graft Rejection/chemically induced , Graft Rejection/etiology , Interleukin-2/pharmacology , Islets of Langerhans Transplantation/immunology , Kinetics , Mice , Mice, Inbred A , Mice, Inbred C57BL , Recombinant Proteins/pharmacology , Thymus Gland/cytology
10.
Transplantation ; 55(1): 133-9, 1993 Jan.
Article in English | MEDLINE | ID: mdl-8420037

ABSTRACT

Monoclonal antibodies directed against different T cell subpopulations have been used in several rodent models of transplantation to induce long-term unresponsiveness to allografts by a variety of mechanisms. To investigate whether different mechanisms may be operative when different regimens of mAb therapy are used, we studied the effects of various combinations of anti-T-cell antibody treatment on the induction of tolerance in a mouse islet allograft model. Anti-CD4 mAb alone, anti-CD8 mAb alone, anti-CD4 mAb plus anti-CD8 mAb, and anti-Thy1.2 mAb alone were given at the time of engraftment. Only the anti-CD4 mAb and the anti-CD4 mAb plus anti-CD8 mAb regimens were successful in inducing permanent unresponsiveness to islet allografts. We have previously shown that anti-CD4 mAb alone induces permanent unresponsiveness to islet allografts by a mechanism of clonal anergy, as demonstrated by unresponsiveness of potentially alloreactive T cells to anti-T-cell receptor-specific cross-linking. Interestingly, the potentially alloreactive T cell subsets of recipient mice (V beta 5+ and V beta 11+) made unresponsive to islet allografts by anti-CD4 mAb plus anti-CD8 mAb therapy were not found to be anergic using the same assay. Differences between the repopulation kinetics of CD8+ T cells of anti-CD4 mAb plus anti-CD8 mAb treated recipient mice, which accepted islet allografts, and anti-Thy1.2 treated recipient mice, which rejected islet allografts despite similar levels of initial T cell depletion, suggest that unresponsiveness to alloantigen may have been induced in anti-CD4 mAb plus anti-CD8 mAb treated recipients by clearance of donor passenger leukocytes during prolonged CD8+ T cell depletion.


Subject(s)
Antibodies, Monoclonal/immunology , Antilymphocyte Serum/immunology , CD4 Antigens/immunology , CD8 Antigens/immunology , Graft Rejection/immunology , Graft Survival/immunology , Immune Tolerance/immunology , Islets of Langerhans Transplantation/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Antigenic Modulation/immunology , Mice , T-Lymphocytes/cytology , Transplantation, Homologous
12.
Diabetes ; 40(11): 1430-4, 1991 Nov.
Article in English | MEDLINE | ID: mdl-1834499

ABSTRACT

We studied the effects of anti-CD4 treatment of diabetic ACI rats on the induction of tolerance to allogeneic (Lewis) islet allografts. When given as a 4-day treatment regimen, OX38, a mouse anti-rat CD4 antibody, caused depletion of greater than 80% of CD4+ cells from the peripheral blood of treated rats. After induction of diabetes (a single high-dose bolus of streptozocin) and 3 days after the initiation of anti-CD4 immunotherapy, recipient ACI rats were transplanted with fully allogeneic (Lewis) islets of Langerhans via the portal circulation. These transplanted islets were capable of returning the anti-CD4-treated ACI recipients to normoglycemia, which was maintained indefinitely in the absence of further immunosuppression. In contrast, treatment of recipient rats with OX8, an anti-CD8 monoclonal antibody (MoAb), induced only a slight prolongation of graft survival (less than or equal to 30 days). Further characterization of the cellular requirements for the induction of long-term transplantation survival revealed that successful pretransplantation anti-CD4 therapy could be ablated by the coincident treatment of recipient rats with depleting levels of anti-CD8 MoAb. These data point to the necessity of a regulator CD8+ cell in the induction of anti-CD4-mediated transplantation survival in this rat model of islet transplantation.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD4 Antigens/immunology , CD8 Antigens/immunology , Graft Survival/immunology , Islets of Langerhans Transplantation/immunology , Animals , Antibodies, Monoclonal/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Experimental/therapy , Disease Models, Animal , Histocompatibility Antigens Class I/immunology , Immunosuppression Therapy , Immunotherapy , Male , Rats , Rats, Inbred ACI , Rats, Inbred Lew , Streptozocin , T-Lymphocytes/immunology , Transplantation, Homologous
13.
J Exp Med ; 173(2): 491-4, 1991 Feb 01.
Article in English | MEDLINE | ID: mdl-1899105

ABSTRACT

Depletion of CD4+ cells using anti-CD4 monoclonal antibodies leads to allograft tolerance. Here we show that anti-CD4-mediated tolerance to pancreatic islets of Langerhans transplanted from an A/J (IEk) donor to a diabetic C57B1/6 (B6) (IE-) recipient occurs in the absence of clonal deletion of the potentially IE-reactive V beta 11+ T cells. Instead, a state of clonal anergy is induced in both the CD4+V beta 11+ and CD8+V beta 11+ T cell subsets. This clonal anergy can be partially overcome in vitro by the addition of recombinant interleukin 2.


Subject(s)
CD4 Antigens/immunology , Immune Tolerance/immunology , Islets of Langerhans Transplantation/immunology , Animals , Antibodies, Monoclonal , Antigens, Differentiation, T-Lymphocyte/immunology , CD8 Antigens , Graft Survival/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred A , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/immunology , T-Lymphocyte Subsets/immunology
14.
J Immunol ; 144(12): 4587-92, 1990 Jun 15.
Article in English | MEDLINE | ID: mdl-1972161

ABSTRACT

A family of rat-mouse chimeric anti-murine CD4 antibodies was used to study the mechanisms of anti-CD4-mediated depletion and immunotherapy. The chimeric antibodies retain identical affinity and specificity as the therapeutically effective prototype antibody, rat GK1.5, but are of different mouse isotypes. GK1.5 gamma 1, GK1.5 gamma 2a, and GK1.5 gamma 2b are significantly more effective at CD4+ cell depletion than rat GK1.5 when low doses of antibody are administered. In contrast, no depletion is seen with GK1.5 gamma 3 at any dose. Depletion of CD4+ cells in vivo is not correlated with either the ability of the antibody to mediate C-dependent cytotoxicity or antibody-dependent cell-mediated cytotoxicity in vitro, implying that additional antibody-mediated cytotoxic mechanisms occur in vivo. The chimeric antibodies were used to investigate the mechanism of GK1.5-mediated immunotherapy in a prototypic model of T cell-mediated autoimmunity, experimental allergic encephalomyelitis. Mice treated with a single dose of 100 micrograms of either GK1.5, GK1.5 gamma 1, or GK1.5 gamma 2a showed significant recovery within 72 h. In contrast, mice treated with 100 micrograms of GK1.5 gamma 3 showed only marginal improvement within the first 72 h and regressed within 5 days of treatment initiation. These data suggest that anti-CD4-mediated immunotherapy of murine experimental allergic encephalomyelitis is correlated with depletion of CD4+ cells.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Animals , Antibody-Dependent Cell Cytotoxicity , Dose-Response Relationship, Immunologic , Immunotherapy , Macrophages/immunology , Mice , Rats , Recombinant Proteins
15.
J Immunol ; 142(6): 2018-23, 1989 Mar 15.
Article in English | MEDLINE | ID: mdl-2466082

ABSTRACT

GK1.5, a rat anti-mouse CD4 mAb, is effective in the treatment of several autoimmune syndromes, induces tolerance to co-administered Ag, and prolongs allograft survival. We have constructed a family of molecules with GK1.5 V regions and mouse gamma 1, gamma 2a, gamma 2b, or gamma 3 constant regions to investigate the mechanisms underlying the effectiveness of GK1.5. The rat-mouse chimeric antibodies are specific for murine CD4 and have identical binding curves as rat GK1.5 on CD4+ T cells. The chimeric GK1.5 gamma 2a, GK1.5 gamma 2b, and GK1.5 gamma 3 antibodies are more efficient than rat GK1.5 at C-mediated cytotoxicity. This is attributed to the enhanced capacity of the chimeric antibodies, compared to rat GK1.5, to lyse CD4+ cells with a low cell surface Ag density. This observation may have important implications for therapy.


Subject(s)
Antibodies, Monoclonal/physiology , Antigens, Differentiation, T-Lymphocyte/immunology , Antilymphocyte Serum/physiology , Cytotoxicity, Immunologic , Recombinant Proteins/physiology , T-Lymphocytes/immunology , Animals , Cell Line , Complement System Proteins , Epitopes/analysis , Leukocyte Count , Mice , Rats , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...