Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
2.
Cancer Cell ; 33(6): 1017-1032.e7, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29894689

ABSTRACT

A significant proportion of cancer patients do not respond to immune checkpoint blockade. To better understand the molecular mechanisms underlying these treatments, we explored the role of CD4+Foxp3- T cells expressing PD-1 (4PD1hi) and observed that 4PD1hi accumulate intratumorally as a function of tumor burden. Interestingly, CTLA-4 blockade promotes intratumoral and peripheral 4PD1hi increases in a dose-dependent manner, while combination with PD-1 blockade mitigates this effect and improves anti-tumor activity. We found that lack of effective 4PD1hi reduction after anti-PD-1 correlates with poor prognosis. Mechanistically, we provide evidence that mouse and human circulating and intra-tumor 4PD1hi inhibit T cell functions in a PD-1/PD-L1 dependent fashion and resemble follicular helper T cell (TFH)-like cells. Accordingly, anti-CTLA-4 activity is improved in TFH deficient mice.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/immunology , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Antibodies/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/genetics , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
3.
Adv Cancer Res ; 128: 1-68, 2015.
Article in English | MEDLINE | ID: mdl-26216629

ABSTRACT

Using the immune system to control cancer has been investigated for over a century. Yet it is only over the last several years that therapeutic agents acting directly on the immune system have demonstrated improved overall survival for cancer patients in phase III clinical trials. Furthermore, it appears that some patients treated with such agents have been cured of metastatic cancer. This has led to increased interest and acceleration in the rate of progress in cancer immunotherapy. Most of the current immunotherapeutic success in cancer treatment is based on the use of immune-modulating antibodies targeting critical checkpoints (CTLA-4 and PD-1/PD-L1). Several other immune-modulating molecules targeting inhibitory or stimulatory pathways are being developed. The combined use of these medicines is the subject of intense investigation and holds important promise. Combination regimens include those that incorporate targeted therapies that act on growth signaling pathways, as well as standard chemotherapy and radiation therapy. In fact, these standard therapies have intrinsic immune-modulating properties that can support antitumor immunity. In the years ahead, adoptive T-cell therapy will also be an important part of treatment for some cancer patients. Other areas which are regaining interest are the use of oncolytic viruses that immunize patients against their own tumors and the use of vaccines against tumor antigens. Immunotherapy has demonstrated unprecedented durability in controlling multiple types of cancer and we expect its use to continue expanding rapidly.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , Humans , Signal Transduction/drug effects , T-Lymphocytes/drug effects
4.
J Immunother Cancer ; 2: 7, 2014.
Article in English | MEDLINE | ID: mdl-24855562

ABSTRACT

With the success of ipilimumab and promise of programmed death-1 pathway-targeted agents, the field of tumor immunotherapy is expanding rapidly. Newer targets for clinical development include select members of the tumor necrosis factor receptor (TNFR) family. Agonist antibodies to these co-stimulatory molecules target both T and B cells, modulating T-cell activation and enhancing immune responses. In vitro and in vivo preclinical data have provided the basis for continued development of 4-1BB, OX40, glucocorticoid-induced TNFR-related gene, herpes virus entry mediator, and CD27 as potential therapies for patients with cancer. In this review, we summarize the immune response to tumors, consider preclinical and early clinical data on select TNFR family members, discuss potential translational challenges and suggest possible combination therapies with the aim of inducing durable antitumor responses.

5.
Cancer Immunol Res ; 2(5): 448-58, 2014 May.
Article in English | MEDLINE | ID: mdl-24795357

ABSTRACT

Induction of potent immune responses to self-antigens remains a major challenge in tumor immunology. We have shown that a vaccine based on alphavirus replicon particles (VRP) activates strong cellular and humoral immunity to tyrosinase-related protein-2 (TRP2) melanoma antigen, providing prophylactic and therapeutic effects in stringent mouse models. Here, we report that the immunogenicity and efficacy of this vaccine is increased in combination with either antagonist anti-CTL antigen-4 (CTLA-4) or agonist anti-glucocorticoid-induced TNF family-related gene (GITR) immunomodulatory monoclonal antibodies (mAb). In the challenging therapeutic setting, VRP-TRP2 plus anti-GITR or anti-CTLA-4 mAb induced complete tumor regression in 90% and 50% of mice, respectively. These mAbs had similar adjuvant effects in priming an adaptive immune response against the vaccine-encoded antigen, augmenting, respectively, approximately 4- and 2-fold the TRP2-specific CD8(+) T-cell response and circulating Abs, compared with the vaccine alone. Furthermore, while both mAbs increased the frequency of tumor-infiltrating CD8(+) T cells, anti-CTLA-4 mAb also increased the quantity of intratumor CD4(+)Foxp3(-) T cells expressing the negative costimulatory molecule programmed death-1 (PD-1). Concurrent GITR expression on these cells suggests that they might be controlled by anti-GITR mAbs, thus potentially explaining their differential accumulation under the two treatment conditions. These findings indicate that combining immunomodulatory mAbs with alphavirus-based anticancer vaccines can provide therapeutic antitumor immune responses in a stringent mouse model, suggesting potential utility in clinical trials. They also indicate that tumor-infiltrating CD4(+)Foxp3(-)PD-1(+) T cells may affect the outcome of immunomodulatory treatments.


Subject(s)
Alphavirus/genetics , Antibodies, Monoclonal/pharmacology , Immunologic Factors/pharmacology , Melanoma, Experimental/immunology , Vaccines, Virus-Like Particle/immunology , Animals , CTLA-4 Antigen/antagonists & inhibitors , Disease Models, Animal , Female , Glucocorticoid-Induced TNFR-Related Protein/antagonists & inhibitors , Immunoglobulin G/immunology , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/therapy , Mice , Phenotype , Replicon , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Cytotoxic/drug effects , Vaccines, Virus-Like Particle/administration & dosage
6.
Clin Cancer Res ; 19(5): 1044-53, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23460535

ABSTRACT

Therapy for cancer can be achieved by artificially stimulating antitumor T and natural killer (NK) lymphocytes with agonist monoclonal antibodies (mAb). T and NK cells express several members of the TNF receptor (TNFR) family specialized in delivering a costimulatory signal on their surface. Engagement of these receptors is typically associated with proliferation, elevated effector functions, resistance to apoptosis, and differentiation into memory cells. These receptors lack any intrinsic enzymatic activity and their signal transduction relies on associations with TNFR-associated factor (TRAF) adaptor proteins. Stimulation of CD137 (4-1BB), CD134 (OX40), and glucocorticoid-induced TNFR (GITR; CD357) promotes impressive tumor-rejecting immunity in a variety of murine tumor models. The mechanisms of action depend on a complex interplay of CTL, T-helper cells, regulatory T cells, dendritic cells, and vascular endothelium in tumors. Agonist mAbs specific for CD137 have shown signs of objective clinical activity in patients with metastatic melanoma, whereas anti-OX40 and anti-GITR mAbs have entered clinical trials. Preclinical evidence suggests that engaging TNFR members would be particularly active with conventional cancer therapies and additional immunotherapeutic approaches. Indeed, T-cell responses elicited to tumor antigens by means of immunogenic tumor cell death are amplified by these immunostimulatory agonist mAbs. Furthermore, anti-CD137 mAbs have been shown to enhance NK-mediated cytotoxicity elicited by rituximab and trastuzumab. Combinations with other immunomodulatory mAb that block T-cell checkpoint blockade receptors such as CTLA-4 and PD-1 are also promising.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunotherapy , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Neoplasms/therapy , Receptors, Tumor Necrosis Factor/agonists , Animals , Humans , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Neoplasms/immunology , Neoplasms/metabolism , Receptors, Tumor Necrosis Factor/immunology
7.
Cancer Immunol Res ; 1(4): 235-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24396833

ABSTRACT

CD4(+) T cells provide help to enhance and sustain cytotoxic CD8(+) T cell responses. A direct lytic role for this cell population in mouse models further supports the use of tumor-reactive CD4(+) T cells for cancer immunotherapy. CTLA-4 blockade has been shown to expand antigen-specific cytotoxic CD4(+) T cells in mouse models. We took advantage of spontaneous immunity to the NY-ESO-1 cancer-testis antigen to investigate quantitative and qualitative changes in antigen-specific CD4(+) T cell responses after ipilimumab (anti-CTLA-4 monoclonal antibody) treatment in advanced melanoma patients. Four NY-ESO-1 seropositive melanoma patients were chosen upon the availability of suitable blood specimens for characterizing the functions of NY-ESO-1 antigen-specific CD4(+) T cell response by enzyme-linked immunospot (ELISPOT), intracellular cytokine staining (ICS) and cytotoxicity assays. Multiple NY-ESO-1 antigen-specific CD4(+) T cell responses with Th1 dominance were induced or enhanced after ipilimumab treatment in peripheral blood in all four patients. NY-ESO-1 antigen-specific CD4(+) T cell lines established from all 4 patients after ipilimumab treatment recognized naturally processed NY-ESO-1 protein in antigen-presenting cells, expressed master transcription factor Eomesodermin (Eomes) and secreted perforin and Granzyme B. Finally, we demonstrated that these NY-ESO-1 antigen-specific CD4(+) T cell lines directly lysed autologous melanoma cell lines expressing NY-ESO-1 in an MHC class II restricted manner. Our results show that antigen specific cytotoxic CD4(+) T cell responses are induced after ipilimumab therapy in human cancer patients. Ipilimumab may induce the expression of lytic granules on antigen specific cytotoxic CD4(+) T cells via Eomes, revealing a novel consequence of immunologic checkpoint blockade.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD4-Positive T-Lymphocytes/immunology , Melanoma/drug therapy , T-Lymphocytes, Cytotoxic/immunology , Adult , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , CTLA-4 Antigen/immunology , Cell Line, Tumor , Female , Granzymes/immunology , Humans , Immunotherapy , Ipilimumab , Lymphocyte Activation , Male , Membrane Proteins/immunology , Middle Aged , Perforin/immunology , T-Box Domain Proteins/immunology
8.
J Exp Med ; 209(11): 2113-26, 2012 Oct 22.
Article in English | MEDLINE | ID: mdl-23008334

ABSTRACT

Harnessing the adaptive immune response to treat malignancy is now a clinical reality. Several strategies are used to treat melanoma; however, very few result in a complete response. CD4(+) T cells are important and potent mediators of anti-tumor immunity and adoptive transfer of specific CD4(+) T cells can promote tumor regression in mice and patients. OX40, a costimulatory molecule expressed primarily on activated CD4(+) T cells, promotes and enhances anti-tumor immunity with limited success on large tumors in mice. We show that OX40 engagement, in the context of chemotherapy-induced lymphopenia, induces a novel CD4(+) T cell population characterized by the expression of the master regulator eomesodermin that leads to both terminal differentiation and central memory phenotype, with concomitant secretion of Th1 and Th2 cytokines. This subpopulation of CD4(+) T cells eradicates very advanced melanomas in mice, and an analogous population of human tumor-specific CD4(+) T cells can kill melanoma in an in vitro system. The potency of the therapy extends to support a bystander killing effect of antigen loss variants. Our results show that these uniquely programmed effector CD4(+) T cells have a distinctive phenotype with increased tumoricidal capability and support the use of immune modulation in reprogramming the phenotype of CD4(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Immunologic Memory/immunology , Melanoma, Experimental/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cell Line, Tumor , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Cytokines/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Humans , Immunotherapy, Adoptive , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxidoreductases/genetics , Oxidoreductases/immunology , Oxidoreductases/metabolism , RNA Interference , Receptors, OX40/immunology , Receptors, OX40/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
9.
Cancer Res ; 72(4): 876-86, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22174368

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of cells that accumulate during tumor formation, facilitate immune escape, and enable tumor progression. MDSCs are important contributors to the development of an immunosuppressive tumor microenvironment that blocks the action of cytotoxic antitumor T effector cells. Heterogeneity in these cells poses a significant barrier to studying the in vivo contributions of individual MDSC subtypes. Herein, we show that granulocyte-macrophage colony stimulating factor, a cytokine critical for the numeric and functional development of MDSC populations, promotes expansion of a monocyte-derived MDSC population characterized by expression of CD11b and the chemokine receptor CCR2. Using a toxin-mediated ablation strategy to target CCR2-expressing cells, we show that these monocytic MDSCs regulate entry of activated CD8 T cells into the tumor site, thereby limiting the efficacy of immunotherapy. Our results argue that therapeutic targeting of monocytic MDSCs would enhance outcomes in immunotherapy.


Subject(s)
Immune Tolerance , Melanoma/immunology , Myeloid Cells/immunology , Receptors, CCR2/metabolism , Tumor Escape/immunology , Tumor Microenvironment/immunology , Animals , CD11b Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Female , Granulocyte-Macrophage Colony-Stimulating Factor , Immune Tolerance/immunology , Lymphocyte Activation , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/metabolism
10.
PLoS One ; 5(9)2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20844763

ABSTRACT

BACKGROUND: Malignant melanoma is the deadliest form of skin cancer and is refractory to conventional chemotherapy and radiotherapy. Therefore alternative approaches to treat this disease, such as immunotherapy, are needed. Melanoma vaccine design has mainly focused on targeting CD8+ T cells. Activation of effector CD8+ T cells has been achieved in patients, but provided limited clinical benefit, due to immune-escape mechanisms established by advanced tumors. We have previously shown that alphavirus-based virus-like replicon particles (VRP) simultaneously activate strong cellular and humoral immunity against the weakly immunogenic melanoma differentiation antigen (MDA) tyrosinase. Here we further investigate the antitumor effect and the immune mechanisms of VRP encoding different MDAs. METHODOLOGY/PRINCIPAL FINDINGS: VRP encoding different MDAs were screened for their ability to prevent the growth of the B16 mouse transplantable melanoma. The immunologic mechanisms of efficacy were investigated for the most effective vaccine identified, focusing on CD8+ T cells and humoral responses. To this end, ex vivo immune assays and transgenic mice lacking specific immune effector functions were used. The studies identified a potent therapeutic VRP vaccine, encoding tyrosinase related protein 2 (TRP-2), which provided a durable anti-tumor effect. The efficacy of VRP-TRP2 relies on a novel immune mechanism of action requiring the activation of both IgG and CD8+ T cell effector responses, and depends on signaling through activating Fcγ receptors. CONCLUSIONS/SIGNIFICANCE: This study identifies a VRP-based vaccine able to elicit humoral immunity against TRP-2, which plays a role in melanoma immunotherapy and synergizes with tumor-specific CD8+ T cell responses. These findings will aid in the rational design of future immunotherapy clinical trials.


Subject(s)
Alphavirus/genetics , Immunity, Cellular , Immunity, Humoral , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/therapeutic use , Melanoma/therapy , Replicon , Alphavirus/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Female , Gene Expression , Genetic Vectors/genetics , Genetic Vectors/physiology , Humans , Immunotherapy , Intramolecular Oxidoreductases/immunology , Melanoma/immunology , Melanoma/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Methods Mol Biol ; 651: 131-55, 2010.
Article in English | MEDLINE | ID: mdl-20686965

ABSTRACT

The use of gene constructs for DNA immunization offers several potential advantages over other commonly used vaccine approaches: (1) full-length cDNA provides multiple potential class I and class II epitopes, thus bypassing limitations of MHC restriction; (2) bacterial plasmid DNA contains immunogenic unmethylated CpG motifs (immunostimulatory sequences) that may act as a potent immunological adjuvant; and (3) DNA is relatively simple to purify in large quantities. The cDNA encoding the antigen of interest is cloned into a bacterial expression plasmid with a constitutively active promoter and this plasmid is injected into the skin or muscle where it is taken up by professional antigen-presenting cells, particularly dendritic cells, either through direct transfection or cross-priming. One can further enhance or modulate the immune response through co-delivery of DNA encoding cytokines or chemokines, including cytokine-Fc fusion molecules. The latter use molecular techniques to fuse a cytokine to the Fc portion of IgG1, creating a chimeric molecule with functional activity. In the present chapter, we will outline the approach to develop cytokine-Fc fusion genes as molecular adjuvants and will use GM-CSF as an example.


Subject(s)
Adjuvants, Immunologic/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunoglobulin G/genetics , Molecular Biology/methods , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Vaccines, DNA/therapeutic use , Animals , Cell Line , Cloning, Molecular , Dendritic Cells/cytology , Dendritic Cells/immunology , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Immunoglobulin G/therapeutic use , Mice , Transfection , Vaccines, DNA/genetics
12.
Blood ; 115(22): 4384-92, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20154220

ABSTRACT

Cyclophosphamide (CTX), a commonly used chemotherapeutic agent can enhance immune responses. The ability of CTX to promote the proliferation of effector T cells and abrogate the function of regulatory T cells (Tregs) has been described. In this study, we examined the effects of CTX treatment on dendritic cell (DC) subsets and the subsequent outcome on the effector and suppressive arms of adaptive immunity. In secondary lymphoid tissues, tissue-derived migratory DCs (migratory DCs), lymphoid tissue-resident DCs (resident DCs), and plasmacytoid DCs (pDCs) are well described. CTX has profound and selective cytotoxic effects on CD8(+) resident DCs, but not skin-derived migratory DCs or pDCs in lymph nodes (LNs) and spleen, causing an imbalance among these DC subsets. CTX treatment increases the potency of DCs in antigen presentation and cytokine secretion, and partially inhibits the suppressor activity of Tregs. Adoptive transfer of CD8(+) DCs can reconstitute this population in regional draining LNs and abrogate the immune-enhancing effects of CTX in vivo. These findings demonstrate that CTX may improve immune responses by preferentially depleting CD8(+) lymphoid-resident DCs, which leads to diminished Treg suppression and enhanced effector T-cell function in vivo.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cyclophosphamide/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Lymphoid Tissue/drug effects , Lymphoid Tissue/immunology , Adoptive Transfer , Animals , Antigen Presentation/drug effects , CD8 Antigens/metabolism , Dendritic Cells/classification , Female , In Vitro Techniques , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Culture Test, Mixed , Lymphoid Tissue/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Models, Immunological , Spleen/cytology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes, Regulatory/immunology
13.
J Clin Invest ; 120(1): 343-56, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19955659

ABSTRACT

Thymic graft-versus-host disease (tGVHD) can contribute to profound T cell deficiency and repertoire restriction after allogeneic BM transplantation (allo-BMT). However, the cellular mechanisms of tGVHD and interactions between donor alloreactive T cells and thymic tissues remain poorly defined. Using clinically relevant murine allo-BMT models, we show here that even minimal numbers of donor alloreactive T cells, which caused mild nonlethal systemic graft-versus-host disease, were sufficient to damage the thymus, delay T lineage reconstitution, and compromise donor peripheral T cell function. Furthermore, to mediate tGVHD, donor alloreactive T cells required trafficking molecules, including CCR9, L selectin, P selectin glycoprotein ligand-1, the integrin subunits alphaE and beta7, CCR2, and CXCR3, and costimulatory/inhibitory molecules, including Ox40 and carcinoembryonic antigen-associated cell adhesion molecule 1. We found that radiation in BMT conditioning regimens upregulated expression of the death receptors Fas and death receptor 5 (DR5) on thymic stromal cells (especially epithelium), while decreasing expression of the antiapoptotic regulator cellular caspase-8-like inhibitory protein. Donor alloreactive T cells used the cognate proteins FasL and TNF-related apoptosis-inducing ligand (TRAIL) (but not TNF or perforin) to mediate tGVHD, thereby damaging thymic stromal cells, cytoarchitecture, and function. Strategies that interfere with Fas/FasL and TRAIL/DR5 interactions may therefore represent a means to attenuate tGVHD and improve T cell reconstitution in allo-BMT recipients.


Subject(s)
Bone Marrow Transplantation/immunology , Fas Ligand Protein/physiology , Graft vs Host Disease/etiology , TNF-Related Apoptosis-Inducing Ligand/physiology , Thymus Gland/immunology , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/analysis , Cell Movement , Graft vs Host Disease/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, OX40/physiology , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/physiology , Stromal Cells/physiology , T-Lymphocytes/immunology , Thymus Gland/cytology , Transplantation, Homologous
14.
J Immunol ; 183(8): 4853-7, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19786544

ABSTRACT

We report that OX40 stimulation drives all lineages of CD4 T cell development, including regulatory T cells (Tregs), and the plasticity of the response is dependant on local cytokines. In TGF-beta1-treated cultures, an OX40 agonist increased IFN-gamma and IL-4 production and diverted T cells from the Treg lineage. However, cytokine blockade in the context of OX40 stimulation promoted enhanced Treg accumulation. This observation was evident in naive mice, as OX40 engagement enhanced Treg proliferation and accumulation in vivo. Lastly, OX40 agonist administration influenced experimental autoimmune encephalomyelitis disease severity in opposing directions, depending on the timing of administration. Given during Ag priming, the OX40 agonist drove Treg expansion and inhibited disease, whereas given later it enhanced T cell effector cytokine production in the CNS and exacerbated disease. Hence, OX40 signaling can augment the accumulation of all CD4 T cell lineages; however, its accentuation of immune responses may have vastly different biologic outcomes depending upon the local cytokine milieu.


Subject(s)
Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Receptors, OX40/agonists , T-Lymphocytes, Regulatory/drug effects , Adoptive Transfer , Animals , Antibodies/administration & dosage , Antibodies/immunology , CD28 Antigens/drug effects , CD28 Antigens/immunology , CD28 Antigens/metabolism , CD3 Complex/drug effects , CD3 Complex/immunology , CD3 Complex/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Interleukin-4/antagonists & inhibitors , Interleukin-4/immunology , Interleukin-4/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, OX40/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/pharmacology
15.
J Exp Med ; 206(5): 1103-16, 2009 May 11.
Article in English | MEDLINE | ID: mdl-19414558

ABSTRACT

Expansion and recruitment of CD4(+) Foxp3(+) regulatory T (T reg) cells are mechanisms used by growing tumors to evade immune elimination. In addition to expansion of effector T cells, successful therapeutic interventions may require reduction of T reg cells within the tumor microenvironment. We report that the combined use of the alkylating agent cyclophosphamide (CTX) and an agonist antibody targeting the co-stimulatory receptor OX40 (OX86) provides potent antitumor immunity capable of regressing established, poorly immunogenic B16 melanoma tumors. CTX administration resulted in tumor antigen release, which after OX86 treatment significantly enhanced the antitumor T cell response. We demonstrated that T reg cells are an important cellular target of the combination therapy. Paradoxically, the combination therapy led to an expansion of T reg cells in the periphery. In the tumor, however, the combination therapy induced a profound T reg cell depletion that was accompanied by an influx of effector CD8(+) T cells leading to a favorable T effector/T reg cell ratio. Closer examination revealed that diminished intratumoral T reg cell levels resulted from hyperactivation and T reg cell-specific apoptosis. Thus, we propose that CTX and OX40 engagement represents a novel and rational chemoimmunotherapy.


Subject(s)
Melanoma, Experimental/drug therapy , Receptors, OX40/genetics , T-Lymphocytes, Regulatory/immunology , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Apoptosis , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cyclophosphamide/therapeutic use , Drug Therapy, Combination , Immunotherapy/methods , Lymphocyte Depletion , Melanoma, Experimental/immunology , Mice , T-Lymphocytes, Regulatory/drug effects
16.
J Exp Med ; 206(4): 849-66, 2009 Apr 13.
Article in English | MEDLINE | ID: mdl-19332877

ABSTRACT

A primary goal of cancer immunotherapy is to improve the naturally occurring, but weak, immune response to tumors. Ineffective responses to cancer vaccines may be caused, in part, by low numbers of self-reactive lymphocytes surviving negative selection. Here, we estimated the frequency of CD8(+) T cells recognizing a self-antigen to be <0.0001% ( approximately 1 in 1 million CD8(+) T cells), which is so low as to preclude a strong immune response in some mice. Supplementing this repertoire with naive antigen-specific cells increased vaccine-elicited tumor immunity and autoimmunity, but a threshold was reached whereby the transfer of increased numbers of antigen-specific cells impaired functional benefit, most likely because of intraclonal competition in the irradiated host. We show that cells primed at precursor frequencies below this competitive threshold proliferate more, acquire polyfunctionality, and eradicate tumors more effectively. This work demonstrates the functional relevance of CD8(+) T cell precursor frequency to tumor immunity and autoimmunity. Transferring optimized numbers of naive tumor-specific T cells, followed by in vivo activation, is a new approach that can be applied to human cancer immunotherapy. Further, precursor frequency as an isolated variable can be exploited to augment efficacy of clinical vaccine strategies designed to activate any antigen-specific CD8(+) T cells.


Subject(s)
Autoantigens/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , Animals , Autoantigens/therapeutic use , Autoimmunity , CD8-Positive T-Lymphocytes/radiation effects , Cell Division , Humans , Melanoma/immunology , Melanoma, Experimental/immunology , Mice , Neoplasms/pathology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Skin Neoplasms/immunology , Spleen/immunology
17.
Clin Cancer Res ; 12(18): 5511-9, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-17000687

ABSTRACT

PURPOSE: Plasmid DNAs encoding cytokines enhance immune responses to vaccination in models of infectious diseases and cancer. We compared DNA adjuvants for their ability to enhance immunity against a poorly immunogenic self-antigen expressed by cancer. EXPERIMENTAL DESIGN: DNAs encoding cytokines that affect T cells [interleukin (IL)-2, IL-12, IL-15, IL-18, IL-21, and the chemokine CCL21] and antigen-presenting cells [granulocyte macrophage colony-stimulating factor (GM-CSF)] were compared in mouse models as adjuvants to enhance CD8+ T-cell responses and tumor immunity. A DNA vaccine against a self-antigen, gp100, expressed by melanoma was used in combination with DNA encoding cytokines and cytokines fused to the Fc domain of mouse IgG1 (Ig). RESULTS: We found that (a) cytokine DNAs generally increased CD8+ T-cell responses against gp100; (b) ligation to Fc domains further enhanced T-cell responses; (c) adjuvant effects were sensitive to timing of DNA injection; (d) the most efficacious individual adjuvants for improving tumor-free survival were IL-12/Ig, IL-15/Ig, IL-21/Ig, GM-CSF/Ig, and CCL21; and (e) combinations of IL-2/Ig+IL-12/Ig, IL-2/Ig+IL-15/Ig, IL-12/Ig+IL-15/Ig, and IL-12/Ig+IL-21/Ig were most active; and (f) increased adjuvanticity of cytokine/Ig fusion DNAs was not related to higher tissue levels or greater stability. CONCLUSIONS: These observations support the potential of cytokine DNA adjuvants for immunization against self-antigens expressed by cancer, the importance of timing, and the enhancement of immune responses by Fc domains through mechanisms unrelated to increased half-life.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cytokines/genetics , Immunotherapy, Active/methods , Neoplasms/immunology , Vaccines, DNA/therapeutic use , Acute-Phase Reaction/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Animals , Autoantigens/immunology , CD8 Antigens/metabolism , Cytokines/immunology , Graft Rejection/immunology , Hypopigmentation/immunology , Immunoglobulin Fc Fragments/genetics , Immunotherapy, Active/adverse effects , Immunotherapy, Active/statistics & numerical data , Interleukin-12/immunology , Mice , Mice, Inbred C57BL , Plasmids/chemistry , Plasmids/immunology , Survival Analysis , T-Lymphocytes/immunology , Time Factors , Vaccines, DNA/adverse effects , Vaccines, DNA/chemistry , Vaccines, DNA/immunology , Xenograft Model Antitumor Assays/methods
18.
J Immunol ; 176(11): 6434-42, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16709800

ABSTRACT

Glucocorticoid-induced TNF receptor family related protein (GITR) is present on many different cell types. Previous studies have shown that in vivo administration of an anti-GITR agonist mAb (DTA-1) inhibits regulatory T cells (Treg)-dependent suppression and enhances T cell responses. In this study, we show that administration of DTA-1 induces >85% tumor rejection in mice challenged with B16 melanoma. Rejection requires CD4+, CD8+, and NK1.1+ cells and is dependent on IFN-gamma and Fas ligand and independent of perforin. Depletion of Treg via anti-CD25 treatment does not induce B16 rejection, whereas 100% of the mice depleted of CD25+ cells and treated with DTA-1 reject tumors, indicating a predominant role of GITR on effector T cell costimulation rather than on Treg modulation. T cells isolated from DTA-1-treated mice challenged with B16 are specific against B16 and several melanoma differentiation Ags. These mice develop memory against B16, and a small proportion of them develop mild hypopigmentation. Consistent with previous studies showing that GITR stimulation increases Treg proliferation in vitro, we found in our model that GITR stimulation expanded the absolute number of FoxP3+ cells in vivo. Thus, we conclude that overall, GITR stimulation overcomes self-tolerance/ignorance and enhances T cell-mediated antitumor activity with minimal autoimmunity.


Subject(s)
Adjuvants, Immunologic/genetics , Antigens, Differentiation/immunology , Gene Expression Regulation, Neoplastic/immunology , Immune Tolerance/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/prevention & control , Neoplasm Proteins/immunology , Receptors, Nerve Growth Factor/genetics , Receptors, Tumor Necrosis Factor/genetics , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/physiology , Adoptive Transfer , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm , Cell Line, Tumor , Disease Progression , Dose-Response Relationship, Immunologic , Female , Glucocorticoid-Induced TNFR-Related Protein , Graft Rejection , Killer Cells, Natural/immunology , Lymphocyte Depletion , Melanoma, Experimental/genetics , Melanoma-Specific Antigens , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Receptors, Nerve Growth Factor/agonists , Receptors, Nerve Growth Factor/immunology , Receptors, Nerve Growth Factor/metabolism , Receptors, Tumor Necrosis Factor/agonists , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , T-Lymphocyte Subsets/immunology , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...