Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 66(2): 1105-13, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16424047

ABSTRACT

Recent studies suggest that immunotherapy targeting specific tumor-associated antigens (TAAs) may be beneficial in cancer patients. However, most of these TAAs are tumor type specific and heterogeneous among patients, thus limiting their applications. Here, we describe the de novo induction of a cancer/testis antigen (CTA) for immunotherapy of tumors of various histologies. The murine CTA P1A, normally expressed only in a few tumor lines, could be induced de novo in all P1A-negative cancer lines of eight histologic origins in vitro and in various murine xenografts by systemic administration of 5-aza-2'-deoxycytidine. The induction of P1A expression correlated strongly with demethylation of the CpG island in the promoter region of this gene. The induced antigen was processed and presented properly for recognition by H-2L(d)-restricted P1A-specific CTLs. The combination of a demethylating agent and adoptive transfer of P1A-specific CTL effectively treated lung metastases in syngeneic mice challenged with P1A-negative 4T1 mammary carcinoma cells. These data show a novel strategy of combined chemoimmunotherapy of cancer targeting a CTA induced de novo in a broad range of tumor histologies, and support further evaluation of chromatin-remodeling agents for human cancer therapy.


Subject(s)
Antigens, Neoplasm/biosynthesis , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Immunotherapy, Adoptive , Animals , Antigens, Neoplasm/physiology , Azacitidine/pharmacology , Chromatin/metabolism , CpG Islands , DNA Methylation , Decitabine , Gene Expression Profiling , Humans , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/therapy , Mice , Neoplasms/immunology , Neoplasms/therapy , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Cells, Cultured
2.
J Exp Med ; 202(7): 907-12, 2005 Oct 03.
Article in English | MEDLINE | ID: mdl-16203864

ABSTRACT

Depletion of immune elements before adoptive cell transfer (ACT) can dramatically improve the antitumor efficacy of transferred CD8+ T cells, but the specific mechanisms that contribute to this enhanced immunity remain poorly defined. Elimination of CD4+CD25+ regulatory T (T reg) cells has been proposed as a key mechanism by which lymphodepletion augments ACT-based immunotherapy. We found that even in the genetic absence of T reg cells, a nonmyeloablative regimen substantially augmented CD8+ T cell reactivity to self-tissue and tumor. Surprisingly, enhanced antitumor efficacy and autoimmunity was caused by increased function rather than increased numbers of tumor-reactive T cells, as would be expected by homeostatic mechanisms. The gammaC cytokines IL-7 and IL-15 were required for augmenting T cell functionality and antitumor activity. Removal of gammaC cytokine-responsive endogenous cells using antibody or genetic means resulted in the enhanced antitumor responses similar to those seen after nonmyeloablative conditioning. These data indicate that lymphodepletion removes endogenous cellular elements that act as sinks for cytokines that are capable of augmenting the activity of self/tumor-reactive CD8+ T cells. Thus, the restricted availability of homeostatic cytokines can be a contributing factor to peripheral tolerance, as well as a limiting resource for the effectiveness of tumor-specific T cells.


Subject(s)
Adoptive Transfer/methods , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Lymphopenia/immunology , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line, Tumor , Female , Mice , Mice, Inbred C57BL , Vaccination , Whole-Body Irradiation
3.
J Leukoc Biol ; 76(2): 333-7, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15155774

ABSTRACT

Immunotherapy using adoptive cell transfer is a promising approach that can result in the regression of bulky, invasive cancer in some patients. However, currently available therapies remain less successful than desired. To study the mechanisms of action and possible improvements in cell-transfer therapies, we use a murine model system with analogous components to the treatment of patients. T cell receptor transgenic CD8+ T cells (pmel-1) specifically recognizing the melanocyte differentiation antigen gp100 are adoptively transferred into lympho-depleted mice bearing large, established, 14-day subcutaneous B16 melanoma (0.5-1 cm in diameter) on the day of treatment. Adoptive cell transfer in combination with interleukin interleukin-2 or interleukin-15 cytokine administration and vaccination using an altered form of the target antigen, gp100, can result in the complete and durable regression of large tumor burdens. Complete responders frequently develop autoimmunity with vitiligo at the former tumor site that often spreads to involve the whole coat. These findings have important implications for the design of immunotherapy trials in humans.


Subject(s)
Adoptive Transfer , Disease Models, Animal , Immunotherapy , Melanoma/therapy , Neoplasm Metastasis/therapy , Animals , Melanoma/immunology , Mice , Neoplasm Metastasis/immunology , T-Lymphocytes/immunology
4.
Proc Natl Acad Sci U S A ; 101(7): 1969-74, 2004 Feb 17.
Article in English | MEDLINE | ID: mdl-14762166

ABSTRACT

IL-15 and IL-2 possess similar properties, including the ability to induce T cell proliferation. However, whereas IL-2 can promote apoptosis and limit CD8(+) memory T cell survival and proliferation, IL-15 helps maintain a memory CD8(+) T cell population and can inhibit apoptosis. We sought to determine whether IL-15 could enhance the in vivo function of tumor/self-reactive CD8(+) T cells by using a T cell receptor transgenic mouse (pmel-1) whose CD8(+) T cells recognize an epitope derived from the self/melanoma antigen gp100. By removing endogenous IL-15 by using tumor-bearing IL-15 knockout hosts or supplementing IL-15 by means of exogenous administration, as a component of culture media or as a transgene expressed by adoptively transferred T cells, we demonstrate that IL-15 can improve the in vivo antitumor activity of adoptively transferred CD8(+) T cells. These results provide several avenues for improving adoptive immunotherapy of cancer in patients.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interleukin-15/immunology , Melanoma, Experimental/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cells, Cultured , Female , Humans , Immunotherapy , Interleukin-15/genetics , Interleukin-15/pharmacology , Interleukin-2/immunology , Interleukin-2/pharmacology , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasm Proteins/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , gp100 Melanoma Antigen
5.
Cancer Res ; 63(18): 5957-61, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-14522922

ABSTRACT

Interleukin (IL)-1 is a pleiotropic inflammatory cytokine that promotes angiogenesis and enhances tumor growth and metastases. We evaluated the effects of IL-1 receptor antagonist (IL-1ra) on tumor growth and metastases in human melanoma xenografts. We selected two human melanoma lines (SMEL and PMEL) with differential (high versus low, respectively) constitutive production of IL-1 by ELISA. The IL-1ra gene was isolated from monocyte RNA by PCR and retrovirally transduced into SMEL and PMEL. In vitro cell proliferation was evaluated using a WST-1 assay. Athymic nude mice received s.c. or i.v. injection with parental, vector-transduced, or IL-1ra-transduced melanoma cells, and tumor growth, lung metastases, and histology were characterized. IL-1 was produced by SMEL in vitro and ex vivo (117 and 67 pg/ml/10(6) cells/24 h, respectively), but not by PMEL (15 and 0 pg/ml/10(6) cells/24 h, respectively). Neither made IL-1ra natively. Gene-transduced cell lines secreted >1000 pg/ml/10(6) cells/24 h of IL-1ra by ELISA. In vitro proliferation of each parental cell line was comparable to the proliferation rate of each transduced cell line. IL-1ra-transduced SMEL (SMEL/IL-1ra) showed significantly slower tumor growth compared with null-transduced and parental cell lines (P < 0.001, ANOVA-Bonferroni/Dunn). There was no difference in growth rates between PMEL and IL-1ra-transduced PMEL (PMEL/IL-1ra). A mixing study of SMEL and SMEL/IL-1ra showed significant inhibition of tumor growth at various ratios (P < 0.001, ANOVA-Bonferroni/Dunn). There were significantly fewer lung metastases with SMEL/IL-1ra versus SMEL (P < 0.002). IL-1ra decreases in vivo growth and metastatic potential of a human melanoma xenograft that constitutively secretes IL-1. This effect may be exploitable using clinically available IL-1ra for the treatment of human cancers.


Subject(s)
Genetic Therapy/methods , Melanoma/therapy , Sialoglycoproteins/physiology , Animals , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/metabolism , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Nude , Sialoglycoproteins/biosynthesis , Sialoglycoproteins/genetics , Transduction, Genetic , Transfection , Xenograft Model Antitumor Assays
6.
J Exp Med ; 198(4): 569-80, 2003 Aug 18.
Article in English | MEDLINE | ID: mdl-12925674

ABSTRACT

Many tumor-associated antigens are derived from nonmutated "self" proteins. T cells infiltrating tumor deposits recognize self-antigens presented by tumor cells and can be expanded in vivo with vaccination. These T cells exist in a functionally tolerant state, as they rarely result in tumor eradication. We found that tumor growth and lethality were unchanged in mice even after adoptive transfer of large numbers of T cells specific for an MHC class I-restricted epitope of the self/tumor antigen gp100. We sought to develop new strategies that would reverse the functionally tolerant state of self/tumor antigen-reactive T cells and enable the destruction of large (with products of perpendicular diameters of >50 mm2), subcutaneous, unmanipulated, poorly immunogenic B16 tumors that were established for up to 14 d before the start of treatment. We have defined three elements that are all strictly necessary to induce tumor regression in this model: (a) adoptive transfer of tumor-specific T cells; (b) T cell stimulation through antigen-specific vaccination with an altered peptide ligand, rather than the native self-peptide; and (c) coadministration of a T cell growth and activation factor. Cells, vaccination, or cyto-kine given alone or any two in combination were insufficient to induce tumor destruction. Autoimmune vitiligo was observed in mice cured of their disease. These findings illustrate that adoptive transfer of T cells and IL-2 can augment the function of a cancer vaccine. Furthermore, these data represent the first demonstration of complete cures of large, established, poorly immunogenic, unmanipulated solid tumors using T cells specific for a true self/tumor antigen and form the basis for a new approach to the treatment of patients with cancer.


Subject(s)
Autoimmunity , CD8-Positive T-Lymphocytes/immunology , Immune Tolerance , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Membrane Glycoproteins/immunology , Neoplasm Proteins/immunology , Adoptive Transfer , Animals , Histocompatibility Antigens Class I , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-2/metabolism , Major Histocompatibility Complex , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Self Tolerance , Survival Rate , Vaccination , gp100 Melanoma Antigen
7.
J Immunother ; 25(3): 218-25, 2002.
Article in English | MEDLINE | ID: mdl-12000863

ABSTRACT

Salmonella typhimurium genetically modified at the purI and msbB genes to increase dependence on adenine and decrease stimulation of tumor necrosis factor-alpha production were injected intravenously into C57BL/6 mice bearing subcutaneous tumor or lung metastases. Decreased tumor growth and prolonged survival were seen in some, but not all of nine transplantable tumors. Salmonella increased in number in the tumor and reached levels 10,000 times higher than in the normal liver reservoir of these bacteria. Histologic studies revealed Salmonella growth in areas of the tumor although, in all cases, a viable rim of tumor survived and ultimately resulted in progressive tumor growth in all mice. These studies demonstrate that Salmonella can localize to transplantable murine tumors and partially inhibit tumor growth; however, additional modifications of the bacteria may be necessary if this approach is to develop into an effective treatment for patients with cancer.


Subject(s)
Neoplasms, Experimental/therapy , Salmonella typhimurium/immunology , Animals , Injections, Intravenous , Liver/microbiology , Liver/pathology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/microbiology , Neoplasms, Experimental/pathology
8.
J Immunol ; 168(1): 338-47, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11751979

ABSTRACT

Tumor Ag-specific vaccines used for cancer immunotherapy can generate specific CD8 responses detectable in PBMCs and in tumor-infiltrating lymphocytes. However, human studies have shown that detection of a systemic vaccine-induced response does not necessarily correlate with the occasional instances of tumor rejection. Because this discrepancy might partially be attributable to the genetic heterogeneity of human cancers, as well as to the immunosuppressive effects of previous treatments, we turned to a mouse model in which these variables could be controlled to determine whether a relationship exists between the strength of vaccine-induced immune responses and tumor rejection. We challenged mice with the beta-galactosidase (beta-gal)-expressing tumor cells, C25.F6, vaccinated them with beta-gal-carrying viral vectors, and used quantitative RT-PCR to measure the vaccine-induced immune response of splenocytes directly ex vivo. We found that the strength of the response increased with increasing doses of beta-gal-carrying vector and/or upon boosting with a heterologous beta-gal-carrying virus. Most importantly, we found that the strength of the detected immune response against this foreign Ag strongly correlated with reduction in the number of lung metastases. The results from this mouse model have major implications for the implementation of tumor vaccines in humans.


Subject(s)
Cancer Vaccines/immunology , Neoplasms, Experimental/prevention & control , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Cells, Cultured , Cytokines/biosynthesis , Cytokines/genetics , Female , Immunization, Secondary , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Polymerase Chain Reaction/methods , RNA, Messenger/biosynthesis , Spleen/immunology , Transduction, Genetic , Tumor Cells, Cultured , beta-Galactosidase/genetics , beta-Galactosidase/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...