Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Biol Blood Marrow Transplant ; 10(8): 524-33, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15282530

ABSTRACT

We demonstrated previously that tumor lysate-pulsed dendritic cells (TP-DC) could mediate a specific and long-lasting antitumor immune response against a weakly immunogenic breast tumor during early lymphoid reconstitution. The purpose of this study was to examine the potential therapeutic efficacy of bone marrow transplants from TP-DC-vaccinated donors. In 2 aggressive metastatic models, bone marrow transplantation with donor bone marrow cells from TP-DC-immunized mice mediated a tumor-specific immune response in the recipient, and this caused regressions of preexisting tumor metastases. After vaccination with TP-DC, donors harbored increased numbers of both activated CD4+ and CD8+ T-cell populations in the bone marrow. Adoptive transfer of T cells purified from the bone marrow of TP-DC-vaccinated mice led to a reduction in preestablished lung metastases, whereas depletion of T cells from bone marrow abolished this effect. By using T cells derived from the bone marrow of TP-DC-vaccinated major histocompatibility complex class I and class II knockout mice, the effector cells required for the observed antitumor effect were determined to be major histocompatibility complex class I-restricted CD8+ T cells. Additionally, the tumor burden in TP-DC-immunized transplant recipients could be reduced further by repetitive TP-DC immunizations after bone marrow transplantation. Collectively, these results demonstrate an important therapeutic role of bone marrow from TP-DC-immunized donors and raise the potential for this approach in patients with advanced cancer.


Subject(s)
Adoptive Transfer , Antigens, Neoplasm/immunology , Bone Marrow Transplantation/immunology , Dendritic Cells/immunology , Animals , Antigens, CD/analysis , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Transplantation/methods , Coculture Techniques , Dendritic Cells/transplantation , Female , Flow Cytometry , Genes, MHC Class I/genetics , Genes, MHC Class I/immunology , Immunophenotyping , Interferon-gamma/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lymphocyte Activation/immunology , Lymphocyte Depletion , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vaccination
2.
Proc Natl Acad Sci U S A ; 99(2): 931-6, 2002 Jan 22.
Article in English | MEDLINE | ID: mdl-11792864

ABSTRACT

Dendritic cells (DC) can serve to immunize the newborn immune system to foreign antigen. In a lymphopenic environment, naive T cells undergoing homeostasis-driven proliferation can acquire increased sensitivity to antigen stimulation. Here, we evaluated the capacity of DC to effectively prime the host immune system to elicit antitumor effects in the setting of early lymphoid reconstitution after bone marrow transplantation (BMT). Indeed, bone marrow-derived, cytokine-driven DC pulsed with whole tumor lysates (TP-DC) could, early on, prime a specific and long-lasting antitumor immune response, which mediated the rejection of a lethal challenge of a weakly immunogenic breast tumor. In the therapeutic setting, TP-DC could also inhibit the growth of preexisting breast tumor metastases by repetitive immunizations initiated early after BMT. Spleen T cells obtained from mice immunized with TP-DC early after BMT showed a substantial increase in tumor-specific IFN-gamma production. Our findings demonstrate that it is possible to promote effective antitumor immunity in a defined lymphopenic environment through DC-based immunization.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/transplantation , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/therapy , T-Lymphocytes/immunology , Animals , Bone Marrow Transplantation , Female , Immunotherapy, Adoptive , Interferon-gamma/biosynthesis , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation, Homologous , Transplantation, Isogeneic
SELECTION OF CITATIONS
SEARCH DETAIL
...