Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Immunol Immunother ; 65(5): 601-11, 2016 May.
Article in English | MEDLINE | ID: mdl-27011014

ABSTRACT

Adoptive cell transfer (ACT) melanoma immunotherapy typically employs acutely activated effector CD8+ T cells for their ability to rapidly recognize and clear antigen. We have previously observed that effector CD8+ T cells are highly susceptible to melanoma-induced suppression, whereas memory CD8+ T cells are not. Although memory T cells have been presumed to be potentially advantageous for ACT, the kinetics of local and systemic T cell responses after effector and memory ACT have not been compared. B16F10 melanoma cells stably transfected to express very low levels of the lymphocytic choriomeningitis virus (LCMV) peptide GP33 (B16GP33) were inoculated into syngeneic C57BL/6 mice. Equal numbers of bona fide naïve, effector, or memory phenotype GP33-specific CD8+ T cells were adoptively transferred into mice 1 day after B16GP33 inoculation. The efficacy of ACT immunotherapy was kinetically assessed using serial tumor measurements and flow cytometric analyses of local and systemic CD8+ T cell responses. Control of B16GP33 tumor growth, persistence of adoptively transferred CD8+ cells, intratumoral infiltration of CD8+ T cells, and systemic CD8+ T cell responsiveness to GP33 were strongest after ACT of memory CD8+ T cells. Following surgical tumor resection and melanoma tumor challenge, only mice receiving memory T cell-based ACT immunotherapy exhibited durable tumor-specific immunity. These findings demonstrate how the use of non-expanded memory CD8+ T cells may enhance ACT immunotherapeutic efficacy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , T-Lymphocytes/transplantation , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , Antigens, Viral/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/immunology , Female , Flow Cytometry , Glycoproteins/genetics , Glycoproteins/immunology , Glycoproteins/metabolism , Immunologic Memory/immunology , Melanoma, Experimental/genetics , Mice, Inbred C57BL , Peptide Fragments/genetics , Peptide Fragments/immunology , Peptide Fragments/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Burden/genetics , Tumor Burden/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism
2.
J Immunother ; 38(2): 54-61, 2015.
Article in English | MEDLINE | ID: mdl-25658614

ABSTRACT

Immunotherapeutic approaches to the treatment of advanced melanoma have relied on strategies that augment the responsiveness of endogenous tumor-specific T-cell populations [eg, cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) blockade-mediated checkpoint inhibition] or introduce exogenously prepared tumor-specific T-cell populations [eg, adoptive cell transfer (ACT)]. Although both approaches have shown considerable promise, response rates to these therapies remain suboptimal. We hypothesized that a combinatorial approach to immunotherapy using both CTLA-4 blockade and nonlymphodepletional ACT could offer additive therapeutic benefit. C57BL/6 mice were inoculated with syngeneic B16F10 melanoma tumors transfected to express low levels of the lymphocytic choriomeningitis virus peptide GP33 (B16GP33), and treated with no immunotherapy, CTLA-4 blockade, ACT, or combination immunotherapy of CTLA-4 blockade with ACT. Combination immunotherapy resulted in optimal control of B16GP33 melanoma tumors. Combination immunotherapy promoted a stronger local immune response reflected by enhanced tumor-infiltrating lymphocyte populations, and a stronger systemic immune responses reflected by more potent tumor antigen-specific T-cell activity in splenocytes. In addition, whereas both CTLA-4 blockade and combination immunotherapy were able to promote long-term immunity against B16GP33 tumors, only combination immunotherapy was capable of promoting immunity against parental B16F10 tumors as well. Our findings suggest that a combinatorial approach using CTLA-4 blockade with nonlymphodepletional ACT may promote additive endogenous and exogenous T-cell activities that enable greater therapeutic efficacy in the treatment of melanoma.


Subject(s)
Antibodies, Monoclonal/administration & dosage , CTLA-4 Antigen/antagonists & inhibitors , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/immunology , Skin Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antigens, Viral/genetics , Antigens, Viral/metabolism , CTLA-4 Antigen/immunology , Combined Modality Therapy , Disease Models, Animal , Female , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Immunity, Cellular , Immunity, Humoral , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Peptide Fragments/genetics , Peptide Fragments/metabolism , Skin Neoplasms/genetics , Transgenes/genetics , Tumor Burden , Viral Proteins/genetics , Viral Proteins/metabolism
3.
Cancer Immunol Immunother ; 62(1): 149-59, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22865267

ABSTRACT

We have previously observed that in vivo exposure to growing melanoma tumors fundamentally alters activated T cell homeostasis by suppressing the ability of naïve T cells to undergo antigen-driven proliferative expansion. We hypothesized that exposure of T cells in later stages of differentiation to melanoma would have similar suppressive consequences. C57BL/6 mice were inoculated with media or syngeneic B16F10 melanoma tumors 8 or 60 days after infection with lymphocytic choriomeningitis virus (LCMV), and splenic populations of LCMV-specific T cells were quantified using flow cytometry 18 days after tumor inoculation. Inoculation with melanoma on post-infection day 8 potentiated the contraction of previously activated T cells. This enhanced contraction was associated with increased apoptotic susceptibility among T cells from tumor-bearing mice. In contrast, inoculation with melanoma on post-infection day 60 did not affect the ability of previously established memory T cells to maintain themselves in stable numbers. In addition, the ability of previously established memory T cells to respond to LCMV challenge was unaffected by melanoma. Following adoptive transfer into melanoma-bearing mice, tumor-specific memory T cells were significantly more effective at controlling melanoma growth than equivalent numbers of tumor-specific effector T cells. These observations suggest that memory T cells are uniquely resistant to suppressive influences exerted by melanoma on activated T cell homeostasis; these findings may have implications for T cell-based cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Melanoma, Experimental/immunology , Animals , Apoptosis/immunology , Disease Models, Animal , Female , Flow Cytometry , Immunotherapy, Adoptive , Male , Mice , Mice, Inbred C57BL
4.
Cell Immunol ; 271(1): 104-9, 2011.
Article in English | MEDLINE | ID: mdl-21741629

ABSTRACT

We have observed that in vivo interaction between melanoma and resting T cells promotes suppression of antigen-driven proliferative T cell expansion. We hypothesized that this suppression would affect tumor antigen-specific T cell populations more potently than tumor-unrelated T cell populations. A B16F10 cell line was stably transfected to express low levels of the lymphocytic choriomeningitis virus (LCMV) glycoprotein GP33 (B16GP33). Mice bearing B16F10 or B16GP33 tumors were infected with LCMV, and proliferative expansion of LCMV epitope-specific T cell populations was quantified. In vitro and in vivo assays confirmed low levels of antigenic GP33 expression by B16GP33 tumors. Suppressed expansion of GP33-specific T cells was equivalent between mice bearing B16F10 and B16GP33 tumors. These observations suggest that the ability of growing melanoma tumors to impair antigen-driven proliferative expansion of activated T cells is global and not antigen-specific, and provide further insight into the influence of cancer on activated T cell homeostasis.


Subject(s)
Antigens, Neoplasm/immunology , Cell Proliferation , Melanoma, Experimental/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , Cell Line, Tumor , Female , Flow Cytometry , Glycoproteins/genetics , Glycoproteins/immunology , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Peptide Fragments/genetics , Peptide Fragments/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Time Factors , Tumor Burden/immunology , Viral Proteins/genetics , Viral Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...