Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Eur J Immunol ; 48(10): 1739-1749, 2018 10.
Article in English | MEDLINE | ID: mdl-30138536

ABSTRACT

RELT (tumor necrosis factor receptor superfamily member 19-like, TNFRSF19L) is a TNFR superfamily member that is primarily expressed in immune cells and lymphoid tissues, but whose immunological function is not well-defined. Here, we show that RELT is expressed by naive T cells and DCs, and their activation or maturation decreases RELT expression. Using RELT knockout (RELT-/- ) mice, we demonstrate that RELT deficiency selectively promotes the homeostatic proliferation of CD4+ T cells but not CD8+ T cells, and enhances anti-tumor CD8+ T-cell responses. We also demonstrate, using an adoptive transfer model in which RELT is knocked-out in either the transferred transgenic CD8+ T cells or the recipient melanoma-bearing mice, that RELT on multiple immune cells limits the hyper-response of tumor-specific CD8+ T cells. Hyper-responsiveness of RELT-deficient T cells was induced by promoting their proliferation. Taken together, our findings suggest that RELT acts as a negative regulator that controls the early phase of T-cell activation probably by promoting T-cell apoptosis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Lymphocyte Activation , Receptors, Tumor Necrosis Factor/genetics , Adoptive Transfer , Animals , Apoptosis , CD8-Positive T-Lymphocytes/immunology , Melanoma/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Nat Commun ; 9(1): 468, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29391449

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy is an effective method for treating specific cancers. CARs are normally designed to recognize antigens, which are highly expressed on malignant cells but not on T cells. However, when T cells are engineered with CARs that recognize antigens expressed on the T cell surface, CAR T cells exhibit effector function on other T cells, which results in fratricide, or killing of neighboring T cells. Here, using human leukocyte antigen-DR (HLA-DR)-targeted CAR T cells, we show that weak affinity between CAR and HLA-DR reduces fratricide and induces sustained CAR downregulation, which consequently tunes the avidity of CAR T cells, leading to desensitization. We further demonstrate that desensitized CAR T cells selectively kill Epstein-Barr virus-transformed B cells with enhanced HLA-DR expression, while sparing normal B cells. Our study supports an avidity-tuning strategy that permits sensing of antigen levels by CAR T cells.


Subject(s)
B-Lymphocytes/cytology , Cell- and Tissue-Based Therapy/methods , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/physiology , Animals , Antibodies, Monoclonal/genetics , B-Lymphocytes/virology , Cell Death , Female , HLA-DR Antigens/metabolism , HLA-DRB1 Chains/metabolism , Herpesvirus 4, Human/pathogenicity , Humans , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , T-Lymphocytes/cytology
3.
Cell Mol Immunol ; 14(9): 748-757, 2017 Sep.
Article in English | MEDLINE | ID: mdl-26972770

ABSTRACT

4-1BB (CD137) is a strong enhancer of the proliferation of CD8+ T cells. Since these cells require increased production of energy and biomass to support their proliferation, we hypothesized that 4-1BB signaling activated glucose and fatty acid metabolism. We found that treatment with agonistic anti-4-1BB mAb promoted the proliferation of CD8+ T cells in vitro, increasing their size and granularity. Studies with a glycolysis inhibitor and a fatty acid oxidation inhibitor revealed that CD8+ T cell proliferation required both glucose and fatty acid metabolism. Anti-4-1BB treatment increased glucose transporter 1 expression and activated the liver kinase B1 (LKB1)-AMP-activated protein kinase (AMPK)-acetyl-CoA carboxylase (ACC) signaling pathway, which may be responsible for activating the metabolism of glucose and fatty acids. We also examined whether blocking glucose or fatty acid metabolism affected cell cycle progression and the anti-apoptotic effect of 4-1BB signaling. The increase of anti-apoptotic factors and cyclins in response to anti-4-1BB treatment was completely prevented by treating CD8+ T cells with the fatty acid oxidation inhibitor, etomoxir, but not with the glycolysis inhibitor, 2-deoxy-D-glucose. We conclude that anti-4-1BB treatment activates glucose and fatty acid metabolism thus supporting the increased demand for energy and biomass, and that fatty acid metabolism plays a crucial role in enhancing the cell cycle progression of anti-CD3-activated CD8+ T cells in vitro and the anti-apoptotic effects of 4-1BB signaling on these cells.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Fatty Acids/metabolism , Glucose/metabolism , Signal Transduction , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Apoptosis , Calcium/metabolism , Cell Cycle , Cell Proliferation , Cell Separation , Cell Size , Lipid Metabolism , Mice, Inbred C57BL , Oxidation-Reduction
4.
Cancer Lett ; 382(2): 186-194, 2016 11 28.
Article in English | MEDLINE | ID: mdl-27609067

ABSTRACT

Although considerable effort has been expended in identifying definitive markers for cancer stem cells (CSCs) or cancer-initiating cells (CICs), the phenotypic plasticity of these cells obviates simple characterization using cell surface markers. We hypothesized that these cells could be characterized by their metabolic properties because they are in a quiescent state with low energy needs. We examined whether cancer cells differ in mitochondrial membrane potential (Δψm) when they are under stress. The Δψm of B16-F10 melanoma cells increased when they were exposed in vitro to serum starvation and chemotherapeutic agents, but not when exposed to hypoxia. Such TMREhigh cells were also present in tumor tissue. They primarily used glucose and/or lactate, and were superior to TMRElow B16-F10 cells in their ability to drive tumor growth. These findings suggest that CSCs or CICs could be identified in heterogeneous melanoma populations by measuring Δψm.


Subject(s)
Cell Proliferation , Melanoma, Experimental/metabolism , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Neoplastic Stem Cells/metabolism , Skin Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Survival , Cisplatin/pharmacology , Energy Metabolism , Female , Glucose/metabolism , Lactic Acid/metabolism , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Phenotype , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Time Factors , Tumor Burden , Tumor Microenvironment
5.
Autophagy ; 12(9): 1647-59, 2016 09.
Article in English | MEDLINE | ID: mdl-27440002

ABSTRACT

VSIG4/CRIg (V-set and immunoglobulin domain containing 4) is a transmembrane receptor of the immunoglobulin superfamily that is expressed specifically on macrophages and mature dendritic cells. VSIG4 signaling accelerates phagocytosis of C3-opsonized bacteria, thereby efficiently clearing pathogens within macrophages. We found that VSIG4 signaling triggered by C3-opsonized Listeria (opLM) or by agonistic anti-VSIG4 monoclonal antibody (mAb) induced macrophages to form autophagosomes. VSIG4-induced autophagosomes were selectively colocalized with the intracellular LM while starvation-induced autophagosomes were not. Consistent with these results, the frequency of autophagosomes induced by infection with opLM was lower in VSIG4-deficient bone marrow-derived macrophages (BMDMs) than in WT BMDMs. Furthermore, when VSIG4 molecules were overexpressed in HeLa cells, which are non-macrophage cells, VSIG4 triggering led to efficient uptake of LM, autophagosome formation, and killing of the infected LM. These findings suggest that VSIG4 signaling not only promotes rapid phagocytosis and killing of C3-opsonized intracellular bacteria, as previously reported, but also induces autophagosome formation, eliminating the LM that have escaped from phagosomes. We conclude that VSIG4 signaling provides an anti-immune evasion mechanism that prevents the outgrowth of intracellular bacteria in macrophages.


Subject(s)
Autophagy , Bacterial Infections/immunology , Macrophages/metabolism , Receptors, Complement/metabolism , Animals , Antibodies, Monoclonal/immunology , Autophagosomes/metabolism , Autophagy-Related Protein 5/genetics , Bone Marrow Cells/cytology , Cell Line , HeLa Cells , Humans , Immunoglobulin G/immunology , Listeria/metabolism , Macrophages/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/immunology , Phagosomes/metabolism , Signal Transduction
6.
J Immunol ; 195(10): 4721-9, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26423152

ABSTRACT

The glucocorticoid-induced TNFR family-related protein (GITR, TNFRSF18, CD357) is expressed on effector and regulatory T (Treg) cells. Previous studies demonstrated that GITR triggering by anti-GITR mAb enhanced T and B cell-mediated immune responses. GITR-deficient T cells, however, also proliferate more than normal T cells, and this effect is unexplained. Because the activities of mAbs are controlled by their Fc regions, the true effect of GITR signaling needs to be determined by examining its interaction with authentic ligand. Therefore, we generated a pentamerized form of the GITRL extracellular domain (pGITRL) for ligation to GITR and compared its effect on T cells with that of anti-GITR mAb. The pGITRL was more effective than anti-GITR mAb in enhancing the proliferation of effector and regulatory cells in vitro and in vivo. Nonetheless, the growth of MC38 adenocarcinoma cells in vivo was only suppressed for initial 15 d by pGITRL, whereas it was suppressed indefinitely by anti-GITR mAb. Detailed analysis revealed that pGITRL induced extensive proliferation of Foxp3(+)CD4(+) Treg cells and led to the accumulation of activated Treg cells in tumor tissue and draining lymph nodes. Because GITR signaling could not neutralize the suppressive activity of activated Treg cells, pGITRL seems to lose its adjuvant effect when sufficient activated Treg cells have accumulated in the lymph nodes and tumor tissue. Indeed, the antitumor effects of pGITRL were markedly enhanced by depleting CD4(+) cells. These results suggest that GITR signaling has stimulatory effects on effector T cells and inhibitory effects through Treg cells.


Subject(s)
Antibodies, Monoclonal/immunology , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factors/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Forkhead Transcription Factors/metabolism , Glucocorticoid-Induced TNFR-Related Protein/immunology , Humans , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Protein Binding , Signal Transduction/immunology , Tumor Necrosis Factors/genetics
7.
PLoS One ; 10(5): e0126765, 2015.
Article in English | MEDLINE | ID: mdl-25962156

ABSTRACT

4-1BB (CD137), a member of the tumor necrosis factor receptor superfamily (TNFRSF), is primarily expressed on activated T cells and is known to enhance proliferation of T cells, prevent activation-induced cell death, and promote memory formation of CD8+ T cells. In particular, it is well acknowledged that 4-1BB triggering preferentially enhances the expansion of CD8+ T cells rather than CD4+ T cells, but the underlying mechanism remains unclear. Here we found that 4-1BB triggering markedly increased IL-2Rα (CD25) and IL-2 expressions of CD8+ T cells but minimally for CD4+ T cells. Proliferation of CD8+ T cells was moderately enhanced by direct 4-1BB triggering in the absence of signaling through IL-2Rα/IL-2 interactions, but further promoted in the presence of IL-2Rα/IL-2 interactions. Among the TNFRSF members including OX40, GITR, CD30, and CD27, 4-1BB was superior in the ability to induce IL-2Rα expression on CD8+ T cells. When the primary and secondary expansions of CD8+ T cells in vivo were examined by adoptively transferring OVA-specific CD8+ T cells along with the treatment with agonistic anti-4-1BB and/or antagonistic anti-CD25 F(ab')2 mAb, 4-1BB triggering enhanced both primary and secondary expansion of CD8+ T cells in vivo, and the 4-1BB effects were moderately suppressed in primary expansion while completely abolished in secondary expansion of OVA-specific CD8+ T cells by blocking IL-2Rα. These results suggest that 4-1BB-mediated increases of IL-2Rα and IL-2 prolong the effects of transient TCR- and 4-1BB-mediated signaling in CD8+ T cells, and that 4-1BB triggering preferentially enhances the expansion of CD8+ T cells through the amplification of autocrine IL-2/IL-2R signaling loop.


Subject(s)
Autocrine Communication , CD8-Positive T-Lymphocytes/metabolism , Interleukin-2/metabolism , Receptors, Interleukin-2/metabolism , Signal Transduction , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Lymphocyte Activation , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin-2/genetics , Signal Transduction/drug effects
8.
J Immunol ; 194(4): 1580-90, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25601928

ABSTRACT

4-1BB signals are considered positive regulators of T cell responses against viruses and tumors, but recent studies suggest that they have more complex roles in modulating T cell responses. Although dual roles of 4-1BB signaling in T cell responses have been suggested, the underlying mechanisms are still not fully understood. In this study, we tested whether 4-1BB expression affected T cell responses differently when expressed in myeloid versus lymphoid cells in vivo. By assessing the proliferation of 4-1BB(+/+) and 4-1BB(-/-) T cells in lymphocyte-deficient RAG2(-/-) and RAG2(-/-)4-1BB(-/-) mice, we were able to compare the effects on T cell responses of 4-1BB expression on myeloid versus T cells. Surprisingly, adoptively transferred T cells were more responsive in tumor-bearing RAG2(-/-)4-1BB(-/-) mice than in RAG2(-/-) mice, and this enhanced T cell proliferation was further enhanced if the T cells were 4-1BB deficient. Dendritic cells (DCs) rather than NK or tissue cells were the myeloid lineage cells primarily responsible for the enhanced T cell proliferation. However, individual 4-1BB(-/-) DCs were less effective in T cell priming in vivo than 4-1BB(+/+) DCs; instead, more DCs in the secondary lymphoid organs of RAG2(-/-)4-1BB(-/-) mice appeared to induce the enhanced T cell proliferation by producing and transpresenting more IL-15. Therefore, we conclude that in vivo 4-1BB signaling of myeloid cells negatively regulates peripheral T cell responses by limiting the differentiation of DCs and their accumulation in secondary lymphoid organs.


Subject(s)
4-1BB Ligand/immunology , Cell Proliferation , Interleukin-15/immunology , Lymphocyte Activation/immunology , Myeloid Cells/immunology , T-Lymphocytes/immunology , 4-1BB Ligand/deficiency , Adoptive Transfer , Animals , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interleukin-15/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
9.
J Immunother ; 37(4): 225-36, 2014 May.
Article in English | MEDLINE | ID: mdl-24714356

ABSTRACT

Adoptive T-cell therapy is a promising approach to the immunotherapy of cancer, but for it to be a general cancer therapy a simple and standardized procedure for producing tumor-specific CD8 T cells is needed. On the basis of a unique property of 4-1BB (CD137), the selective expression on activated T cells, we have developed a simple and practical protocol to produce antigen-specific CD8 T cells from peripheral blood mononuclear cells. We have proved the feasibility of this procedure by isolating and expanding cytomegalovirus-specific CD8 T cells, and applied the procedure to produce Epstein-Barr virus (EBV)-specific CD8 T cells. By using this procedure, we could readily produce 10-10 antigen-specific CD8 T cells from 30 to 50 mL of blood in about 4 weeks. Moreover, our protocol allowed us to produce, from solid cancer patients, CD8 T cells that were specific for self/tumor antigens such as human telomerase reverse transcriptase (hTERT). It is interesting to note that, we were unable to amplify hTERT-specific CD8 T cells from healthy donors. Our protocol can be readily translated into cGMP-compliant production and is currently being used to produce EBV-specific CD8 T cells for phase I clinical trial. We believe that our method will provide a practical and effective option for adoptive T-cell therapy in the clinic.


Subject(s)
Antigens, Neoplasm/immunology , Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive , Neoplasms/immunology , Neoplasms/therapy , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Animals , Antigens, Neoplasm/chemistry , Autoantigens/genetics , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Separation/methods , Cytotoxicity, Immunologic , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Humans , Immunophenotyping , Lymphocyte Activation/immunology , Mice , Peptides/chemistry , Peptides/immunology , Phenotype , Phosphoproteins/immunology , T-Cell Antigen Receptor Specificity/immunology , Telomerase/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Viral Matrix Proteins/immunology
10.
J Immunol ; 187(3): 1120-8, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21715692

ABSTRACT

Agonistic anti-4-1BB Ab is known to ameliorate experimental autoimmune encephalomyelitis. 4-1BB triggering typically leads to the expansion of CD8(+) T cells, which produce abundant IFN-γ, and this in turn results in IDO-dependent suppression of autoimmune responses. However, because neutralization of IFN-γ or depletion of CD8(+) T cell only partially abrogates the effect of 4-1BB triggering, we sought to identify an additional mechanism of 4-1BB-triggered suppression of autoimmune responses using IFN-γ- or IFN-γR-deficient mice. 4-1BB triggering inhibited the generation of Th17 cells that is responsible for experimental autoimmune encephalomyelitis induction and progression, and increased Foxp3(+)CD4(+) regulatory T (Treg) cells, particularly among CD4(+) T cells. This was not due to a direct effect of 4-1BB signaling on CD4(+) T cell differentiation: 4-1BB signaling not only reduced Th17 cells and increased Treg cells in wild-type mice, which could be due to IFN-γ production by the CD8(+) T cells, but also did so in IFN-γ-deficient mice, in that case by downregulating IL-6 production. These results show that although secondary suppressive mechanisms evoked by 4-1BB triggering are usually masked by the strong effects of IFN-γ, 4-1BB signaling seems to modulate autoimmune responses by a number of mechanisms, and modulation of the Th17 versus Treg cell balance is one of those mechanisms.


Subject(s)
Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Tumor Necrosis Factor Receptor Superfamily, Member 9/physiology , Amino Acid Sequence , Animals , CD4 Lymphocyte Count , Cell Differentiation/genetics , Cells, Cultured , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/pathology , Interferon-gamma/deficiency , Interferon-gamma/metabolism , Interferon-gamma/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Receptors, Interferon/physiology , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Interferon gamma Receptor
11.
J Immunol ; 185(3): 1404-11, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20610645

ABSTRACT

Stimulation of 4-1BB (CD137) was shown to produce strong anticancer effects in vivo. In contrast, 4-1BB-deficient (4-1BB(-/-)) B6 mice are remarkably resistant to tumor growth. We set out to determine the mechanisms involved in these seemingly contradictory observations. We found that the therapeutic effects of 4-1BB triggering were mainly dependent on CD8(+) T cells and partially on NK cells, whereas CD8(+) T and NK cells were equally needed to suppress tumor growth in 4-1BB(-/-) mice. Cellular analysis showed that the frequency and number of NK cells in the spleen and bone marrow were decreased by 4-1BB triggering but were increased in the absence of 4-1BB signaling in tumor-challenged mice. The 4-1BB-mediated downregulation of NK cell development was primarily dependent on IFN-gamma, which was produced by peripheral CD8(+) T and NK cells. The suppression of NK cell development by 4-1BB-mediated IFN-gamma production occurred in the bone marrow. As 4-1BB signaling increased in the periphery, more CD8(+) T cells but fewer NK cells contributed to the antitumor immunity. As 4-1BB signaling decreased, more NK cells participated in the antitumor immunity. We conclude that 4-1BB signaling results in a shift of the dominant type of immune cell in antitumor immunity from the innate NK cell to the adaptive CD8(+) T cell and that the level of IFN-gamma is critical for this 4-1BB-mediated shift.


Subject(s)
Cell Differentiation/immunology , Down-Regulation/immunology , Interferon-gamma/physiology , Killer Cells, Natural/immunology , Signal Transduction/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/physiology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma/prevention & control , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/genetics , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/prevention & control , Down-Regulation/genetics , Female , Immunity, Innate/genetics , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Interferon-gamma/deficiency , Interferon-gamma/genetics , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Kidney Neoplasms/prevention & control , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Thymoma/immunology , Thymoma/pathology , Thymoma/prevention & control , Tumor Necrosis Factor Receptor Superfamily, Member 9/deficiency , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
12.
Mol Cancer Ther ; 8(2): 469-78, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19190115

ABSTRACT

Chemotherapy can precondition for immunotherapy by creating an environment for homeostatic lymphoproliferation and eliminating some of the suppressive immune networks. We found that combination therapy with anti-4-1BB and cyclophosphamide (CTX) produced synergistic anticancer effects in the poorly immunogenic B16 melanoma model in mice. The antitumor effect of the combination therapy depended mainly on CD8(+) T cells, the 4-1BB-dependent expansion and differentiation of which into IFN-gamma-producing CD11c(+)CD8(+) T cells was enhanced by CTX. Anti-4-1BB induced a rapid repopulation of T and B cells from CTX-mediated lymphopenia. Anti-4-1BB protected naïve T cells from CTX and promoted proliferation of memory/effector and memory T cells. The combination treatment produced approximately 60- and 2.2-fold more CTLs per tumor-associated antigen compared with CTX or anti-4-1BB alone, respectively. This indicates that anti-4-1BB promoted a preferential expansion of tumor-specific CD8(+) T cells among the repopulated lymphocytes following CTX-mediated lymphopenia. CTX treatment enhanced 4-1BB expression on CD4 and CD8 T cells, and CTX alone or in combination with anti-4-1BB effectively suppressed peripheral regulatory T cells. Our results indicate that anti-4-1BB and CTX can be practical partners in cancer therapy because CTX creates an environment in which anti-4-1BB actively promotes the differentiation and expansion of tumor-specific CTLs.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclophosphamide/therapeutic use , Melanoma/drug therapy , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Antineoplastic Agents/pharmacology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , Cyclophosphamide/pharmacology , Drug Synergism , Epitopes , Forkhead Transcription Factors/metabolism , Immunologic Memory/drug effects , Lymphopenia/drug therapy , Melanoma/immunology , Mice , T-Lymphocytes, Regulatory/drug effects , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...