Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Blood ; 114(9): 1776-83, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19561320

ABSTRACT

Naive and memory CD8(+) T cells can undergo programmed activation and expansion in response to a short T-cell receptor stimulus, but the extent to which in vitro programming can qualitatively substitute for an in vivo antigen stimulation remains unknown. We show that self-/tumor-reactive effector memory CD8(+) T cells (T(EM)) programmed in vitro either with peptide-pulsed antigen-presenting cells or plate-bound anti-CD3/anti-CD28 embark on a highly stereotyped response of in vivo clonal expansion and tumor destruction nearly identical to that of vaccine-stimulated T(EM) cells. This programmed response was associated with an interval of antigen-independent interferon-gamma (IFN-gamma) release that facilitated the dynamic expression of the major histocompatibility complex class I restriction element H-2D(b) on responding tumor cells, leading to recognition and subsequent tumor lysis. Delaying cell transfer for more than 24 hours after stimulation or infusion of cells deficient in IFN-gamma entirely abrogated the benefit of the programmed response, whereas transfer of cells unable to respond to IFN-gamma had no detriment to antitumor immunity. These findings extend the phenomenon of a programmable effector response to memory CD8(+) T cells and have major implications for the design of current adoptive-cell transfer trials.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Cancer Vaccines/metabolism , Animals , CD28 Antigens/biosynthesis , CD3 Complex/biosynthesis , Cell Separation , Flow Cytometry , Genes, MHC Class I , In Vitro Techniques , Interferon-gamma/metabolism , Major Histocompatibility Complex , Melanoma, Experimental , Mice , Mice, Transgenic , Neoplasms/immunology , Phenotype
2.
Vaccine ; 24(24): 5110-8, 2006 Jun 12.
Article in English | MEDLINE | ID: mdl-16725231

ABSTRACT

The immunogenicity and efficacy of nucleic acid vaccines can be greatly enhanced when antigen production is under the control of an alphaviral replicase enzyme. However, replicase-mediated mRNA overproduction does not necessarily result in enhanced antigen level. Instead, the strong adaptive immune response of alphavirus replicon-based vectors is due to their production of double-stranded RNA (dsRNA) intermediates, which trigger innate immunity. Because viral infections are known to trigger innate immune responses that lead to the rapid production of Type I Interferons (IFNs), namely IFN-alpha and IFN-beta, we investigated the role of Type I IFNs in the enhanced immunogenicity of replicase-based DNA vaccines. In vitro, cells transfected with replicase-based plasmids produce significantly more Type I IFNs than cells transfected with a conventional DNA plasmid. In vivo, replicase-based DNA vaccines yield stronger humoral responses in the absence of Type I IFN signaling but the lack of this signaling pathway in IFN-alphabeta receptor-/- (knockout) mice abolishes T cell mediated efficacy against tumors of both conventional and alphavirus replicase-based DNA vaccines. Moreover, the co-delivery of an IFNalpha-encoding plasmid significantly improved the efficacy of a weakly immunogenic conventional plasmid. These results suggest a central role for Type I IFNs in the mechanism of replicase-based DNA vaccines and indicate that vaccines can be enhanced by enabling their capacity to triggering innate anti-viral defense pathways.


Subject(s)
Alphavirus/enzymology , Interferon Type I/physiology , Melanoma, Experimental/prevention & control , Membrane Glycoproteins/genetics , Oxidoreductases/genetics , Replicon , Vaccines, DNA/immunology , Animals , Immunization , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Oxidoreductases/immunology , Plasmids
3.
Cancer Res ; 66(2): 1132-8, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16424050

ABSTRACT

It has been hypothesized that rapidly dividing tumor cells can outpace adoptively transferred antitumor lymphocytes when tumors are large. However, this hypothesis is at odds with clinical observations indicating that bulky tumors can be destroyed by small numbers of adoptively transferred antitumor T cells. We sought to measure the relative growth rates of T cells and tumor cells in a model using transgenic CD8(+) T cells specific for the gp100(25-33) H-2D(b) epitope (called pmel-1) to treat large, well-established s.c. B16 melanoma. We tested the effect of the immunization using an altered peptide ligand vaccine alone or in combination with interleukin-2 (IL-2) by analyzing the kinetics of T-cell expansion using direct enumeration. We found that pmel-1 T cells proliferated explosively during a 5-day period following transfer. Calculations from net changes in population suggest that, at the peak of cell division, pmel-1 T cells divide at a rate of 5.3 hours per cell division, which was much faster than B16 tumor cells during optimal growth (24.9 hours per cell division). These results clearly indicate that the notion of a kinetic "race" between the tumor and the lymphocyte is no contest when adoptively transferred cells are stimulated with immunization and IL-2. When appropriately stimulated, tumor-reactive T-cell expansion can far exceed the growth of even an aggressively growing mouse tumor.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Immunotherapy, Adoptive , Melanoma/pathology , Skin Neoplasms/pathology , Animals , Cancer Vaccines/immunology , Immunization , Interleukin-2/physiology , Kinetics , Ligands , Mice , Mice, Inbred C57BL , Mice, Transgenic
4.
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
5.
J Immunol ; 173(12): 7209-16, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15585842

ABSTRACT

It has been suggested that antitumor T cells specifically traffic to the tumor site, where they effect tumor destruction. To test whether tumor-reactive CD8(+) T cells specifically home to tumor, we assessed the trafficking of gp100-specific pmel-1 cells to large, vascularized tumors that express or do not express the target Ag. Activation of tumor-specific CD8(+) pmel-1 T cells with IL-2 and vaccination with an altered peptide ligand caused regression of gp100-positive tumors (B16), but not gp100-negative tumors (methylcholanthrene 205), implanted on opposing flanks of the same mouse. Surprisingly, we found approximately equal and very large numbers of pmel-1 T cells (>25% of all lymphocytes) infiltrating both Ag-positive and Ag-negative tumors. We also found evidence of massive infiltration and proliferation of activated antitumor pmel-1 cells in a variety of peripheral tissues, including lymph nodes, liver, spleen, and lungs, but not peripheral blood. Most importantly, evidence for T cell function, as measured by production of IFN-gamma, release of perforin, and activation of caspase-3 in target cells, was confined to Ag-expressing tumor. We thus conclude that CD8(+) T cell-mediated destruction of tumor is the result of specific T cell triggering at the tumor site. The ability to induce ubiquitous homing and specific tumor destruction may be important in the case of noninflammatory metastatic tumor foci.


Subject(s)
CD8-Positive T-Lymphocytes/transplantation , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Movement/immunology , Cytotoxicity, Immunologic , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/transplantation , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cytotoxicity, Immunologic/genetics , Fibrosarcoma/pathology , Fibrosarcoma/prevention & control , Fibrosarcoma/therapy , Immunotherapy, Adoptive/methods , Lymphocyte Activation/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Melanoma, Experimental/therapy , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Proteins/administration & dosage , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Transplantation/immunology , Neoplasm Transplantation/methods , Neoplasm Transplantation/pathology , Organ Specificity/genetics , Organ Specificity/immunology , gp100 Melanoma Antigen
6.
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
7.
Vaccine ; 22(11-12): 1537-44, 2004 Mar 29.
Article in English | MEDLINE | ID: mdl-15063579

ABSTRACT

Alphaviral replicons can increase the efficacy and immunogenicity of naked nucleic acid vaccines. To study the impact of apoptosis on this increased effectiveness, we co-delivered an anti-apoptotic gene (Bcl-X(L)) with the melanocyte/melanoma differentiation antigen TRP-1. Although cells co-transfected with Bcl-X(L) lived longer, produced more antigen and elicited increased antibody production in vivo, co-delivery of pro-survival Bcl-X(L) with antigen significantly reduced the ability of the replicase-based vaccine to protect against an aggressive tumor challenge. These data show for the first time that the induction of apoptotic cell death of transfected cells in vivo is required for the increased effectiveness of replicase-based vaccines. Our findings also provide an explanation for the paradoxical observation that replicase-based DNA vaccines are much more immunogenic than conventional constructs despite reduced antigen production.


Subject(s)
Apoptosis/immunology , Apoptosis/physiology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/analysis , Antibodies, Viral/biosynthesis , Cancer Vaccines/immunology , Cell Line , Cell Survival/drug effects , Cricetinae , DNA/genetics , DNA/immunology , Flow Cytometry , Melanoma/immunology , Mice , Mice, Inbred C57BL , Plasmids/genetics , Plasmids/immunology , Proto-Oncogene Proteins c-bcl-2/immunology , Replicon , Sindbis Virus/enzymology , Sindbis Virus/immunology , Transfection , bcl-X Protein
8.
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
9.
Nat Med ; 9(1): 33-9, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12496961

ABSTRACT

Cancer vaccines targeting 'self' antigens that are expressed at consistently high levels by tumor cells are potentially useful in immunotherapy, but immunological tolerance may block their function. Here, we describe a novel, naked DNA vaccine encoding an alphavirus replicon (self-replicating mRNA) and the self/tumor antigen tyrosinase-related protein-1. Unlike conventional DNA vaccines, this vaccine can break tolerance and provide immunity to melanoma. The vaccine mediates production of double-stranded RNA, as evidenced by the autophosphorylation of dsRNA-dependent protein kinase R (PKR). Double-stranded RNA is critical to vaccine function because both the immunogenicity and the anti-tumor activity of the vaccine are blocked in mice deficient for the RNase L enzyme, a key component of the 2',5'-linked oligoadenylate synthetase antiviral pathway involved in double-stranded RNA recognition. This study shows for the first time that alphaviral replicon-encoding DNA vaccines activate innate immune pathways known to drive antiviral immune responses, and points the way to strategies for improving the efficacy of immunization with naked DNA.


Subject(s)
Alphavirus/genetics , Alphavirus/immunology , Cancer Vaccines/immunology , Immune Tolerance , Melanoma/immunology , Membrane Glycoproteins , Oxidoreductases , Proteins/immunology , Vaccines, DNA/immunology , Animals , Cancer Vaccines/genetics , Gene Transfer Techniques , Genes, Reporter , Humans , Immunization , Immunotherapy , Melanoma/therapy , Mice , Mice, Inbred C57BL , Plasmids/genetics , Plasmids/immunology , Plasmids/metabolism , Proteins/genetics , Proteins/metabolism , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Replicon/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Cells, Cultured , Vaccines, DNA/genetics , Vaccines, DNA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...