Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
Immunity ; 38(4): 729-41, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23562161

ABSTRACT

The therapeutic efficacy of anthracyclines relies on antitumor immune responses elicited by dying cancer cells. How chemotherapy-induced cell death leads to efficient antigen presentation to T cells, however, remains a conundrum. We found that intratumoral CD11c(+)CD11b(+)Ly6C(hi) cells, which displayed some characteristics of inflammatory dendritic cells and included granulomonocytic precursors, were crucial for anthracycline-induced anticancer immune responses. ATP released by dying cancer cells recruited myeloid cells into tumors and stimulated the local differentiation of CD11c(+)CD11b(+)Ly6C(hi) cells. Such cells efficiently engulfed tumor antigens in situ and presented them to T lymphocytes, thus vaccinating mice, upon adoptive transfer, against a challenge with cancer cells. Manipulations preventing tumor infiltration by CD11c(+)CD11b(+)Ly6C(hi) cells, such as the local overexpression of ectonucleotidases, the blockade of purinergic receptors, or the neutralization of CD11b, abolished the immune system-dependent antitumor activity of anthracyclines. Our results identify a subset of tumor-infiltrating leukocytes as therapy-relevant antigen-presenting cells.


Subject(s)
Anthracyclines/administration & dosage , Antigen-Presenting Cells/immunology , Antineoplastic Agents/administration & dosage , Dendritic Cells/immunology , Neoplasms, Experimental/immunology , Adoptive Transfer , Animals , Anthracyclines/adverse effects , Antigens, Ly/metabolism , Antigens, Neoplasm/immunology , Antineoplastic Agents/adverse effects , Apoptosis , CD11b Antigen/metabolism , CD11c Antigen/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Granulocyte Precursor Cells/immunology , Immunity, Cellular , Mice , Mice, Inbred C57BL , Monocyte-Macrophage Precursor Cells/immunology , Neoplasms, Experimental/drug therapy , Nucleotidases/metabolism , Receptors, Purinergic/metabolism
2.
Blood ; 120(15): 3019-29, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-22932803

ABSTRACT

Immunomodulators are effective in controlling hematologic malignancy by initiating or reactivating host antitumor immunity to otherwise poorly immunogenic and immune suppressive cancers. We aimed to boost antitumor immunity in B-cell lymphoma by developing a tumor cell vaccine incorporating α-galactosylceramide (α-GalCer) that targets the immune adjuvant properties of NKT cells. In the Eµ-myc transgenic mouse model, single therapeutic vaccination of irradiated, α-GalCer-loaded autologous tumor cells was sufficient to significantly inhibit growth of established tumors and prolong survival. Vaccine-induced antilymphoma immunity required NKT cells, NK cells, and CD8 T cells, and early IL-12-dependent production of IFN-γ. CD4 T cells, gamma/delta T cells, and IL-18 were not critical. Vaccine treatment induced a large systemic spike of IFN-γ and transient peripheral expansion of both NKT cells and NK cells, the major sources of IFN-γ. Furthermore, this vaccine approach was assessed in several other hematopoietic tumor models and was also therapeutically effective against AML-ETO9a acute myeloid leukemia. Replacing α-GalCer with ß-mannosylceramide resulted in prolonged protection against Eµ-myc lymphoma. Overall, our results demonstrate a potent immune adjuvant effect of NKT cell ligands in therapeutic anticancer vaccination against oncogene-driven lymphomas, and this work supports clinical investigation of NKT cell-based immunotherapy in patients with hematologic malignancies.


Subject(s)
Cancer Vaccines/therapeutic use , Galactosylceramides/administration & dosage , Genes, myc/genetics , Immunotherapy , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/prevention & control , Natural Killer T-Cells/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Cytotoxicity, Immunologic/immunology , Female , Flow Cytometry , Genes, T-Cell Receptor delta/physiology , Humans , Interferon-gamma/metabolism , Interleukin-12/physiology , Interleukin-18/physiology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Lymphoma, B-Cell/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology , Vaccination
3.
Immunol Cell Biol ; 91(1): 105-14, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23090488

ABSTRACT

The natural killer T (NKT) cell ligand, alpha-galactosylceramide (α-GalCer), represents a potential adjuvant to boost immunotherapeutic vaccination strategies against poorly immunogenic cancers. The objective of this study was to assess the therapeutic potential of an α-GalCer-loaded tumor-cell vaccine against solid tumors in mice and to enhance the effectiveness of this approach by removing immune suppression associated with the activity of Foxp3(+) regulatory T cells (Tregs). In the B16F10 melanoma model, we show that single vaccination with irradiated, α-GalCer-loaded tumor cells resulted in suppression of established subcutaneous (s.c.) B16F10 tumor growth, which was mediated by NKT cell-dependent IFN-γ production and enhanced in the absence of IL-17 A. Selective depletion of Foxp3(+) Tregs in transgenic DEpletion of REGulatory T cells (DEREG) mice led to significant inhibition of B16F10 tumor growth and enhanced survival of mice receiving vaccination. Short-term elimination of Foxp3(+) Tregs (<7 days) was sufficient to boost vaccine-induced immunity. Enhanced antitumor activity with combination therapy was associated with an increase in systemic NK cell and effector CD8(+) T-cell activation and IFN-γ production, as well as infiltration of effector CD8(+) T cells into the tumor. Overall, these findings demonstrate that transient depletion of Foxp3(+) Tregs constitutes a highly effective strategy to improve the therapeutic efficacy of anticancer vaccination with NKT cell adjuvants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cancer Vaccines/immunology , Forkhead Transcription Factors , Galactosylceramides/pharmacology , Melanoma, Experimental/therapy , Natural Killer T-Cells/immunology , Vaccination , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/pharmacology , Cell Line, Tumor , Galactosylceramides/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-17 , Lymphocyte Depletion , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Knockout , T-Lymphocytes, Regulatory
SELECTION OF CITATIONS
SEARCH DETAIL