Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
JCI Insight ; 6(20)2021 10 22.
Article in English | MEDLINE | ID: mdl-34676831

ABSTRACT

Only a subset of cancer patients responds to checkpoint blockade inhibition in the clinic. Strategies to overcome resistance are promising areas of investigation. Targeting glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR) has shown efficacy in preclinical models, but GITR engagement is ineffective in controlling advanced, poorly immunogenic tumors, such as B16 melanoma, and has not yielded benefit in clinical trials. The alkylating agent cyclophosphamide (CTX) depletes regulatory T cells (Tregs), expands tumor-specific effector T cells (Teffs) via homeostatic proliferation, and induces immunogenic cell death. GITR agonism has an inhibitory effect on Tregs and activates Teffs. We therefore hypothesized that CTX and GITR agonism would promote effective antitumor immunity. Here we show that the combination of CTX and GITR agonism controlled tumor growth in clinically relevant mouse models. Mechanistically, we show that the combination therapy caused tumor cell death, clonal expansion of highly active CD8+ T cells, and depletion of Tregs by activation-induced cell death. Control of tumor growth was associated with the presence of an expanded population of highly activated, tumor-infiltrating, oligoclonal CD8+ T cells that led to a diminished TCR repertoire. Our studies show that the combination of CTX and GITR agonism is a rational chemoimmunotherapeutic approach that warrants further clinical investigation.


Subject(s)
Cyclophosphamide/therapeutic use , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Immunosuppressive Agents/therapeutic use , Immunotherapy/methods , T-Lymphocytes, Regulatory/immunology , Animals , Cyclophosphamide/pharmacology , Humans , Immunosuppressive Agents/pharmacology , Mice
2.
Sci Signal ; 10(494)2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28851824

ABSTRACT

Regulatory T cells (Tregs) suppress antitumor immunity by inhibiting the killing of tumor cells by antigen-specific CD8+ T cells. To better understand the mechanisms involved, we used ex vivo three-dimensional collagen-fibrin gel cultures of dissociated B16 melanoma tumors. This system recapitulated the in vivo suppression of antimelanoma immunity, rendering the dissociated tumor cells resistant to killing by cocultured activated, antigen-specific T cells. Immunosuppression was not observed when tumors excised from Treg-depleted mice were cultured in this system. Experiments with neutralizing antibodies showed that blocking transforming growth factor-ß (TGF-ß) also prevented immunosuppression. Immunosuppression depended on cell-cell contact or cellular proximity because soluble factors from the collagen-fibrin gel cultures did not inhibit tumor cell killing by T cells. Moreover, intravital, two-photon microscopy showed that tumor-specific Pmel-1 effector T cells physically interacted with tumor-resident Tregs in mice. Tregs isolated from B16 tumors alone were sufficient to suppress CD8+ T cell-mediated killing, which depended on surface-bound TGF-ß on the Tregs Immunosuppression of CD8+ T cells correlated with a decrease in the abundance of the cytolytic protein granzyme B and an increase in the cell surface amount of the immune checkpoint receptor programmed cell death protein 1 (PD-1). These findings suggest that contact between Tregs and antitumor T cells in the tumor microenvironment inhibits antimelanoma immunity in a TGF-ß-dependent manner and highlight potential ways to inhibit intratumoral Tregs therapeutically.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunosuppression Therapy , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Communication , Cell Line, Tumor , Coculture Techniques , Female , Granzymes/metabolism , Immunity, Cellular , Mice , Mice, Inbred C57BL , Mice, Transgenic , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes, Regulatory/metabolism
3.
PLoS One ; 8(12): e82496, 2013.
Article in English | MEDLINE | ID: mdl-24349298

ABSTRACT

The development of successful cancer vaccines is contingent on the ability to induce effective and persistent anti-tumor immunity against self-antigens that do not typically elicit immune responses. In this study, we examine the effects of a non-myeloablative dose of total body irradiation on the ability of tumor-naïve mice to respond to DNA vaccines against melanoma. We demonstrate that irradiation followed by lymphocyte infusion results in a dramatic increase in responsiveness to tumor vaccination, with augmentation of T cell responses to tumor antigens and tumor eradication. In irradiated mice, infused CD8(+) T cells expand in an environment that is relatively depleted in regulatory T cells, and this correlates with improved CD8(+) T cell functionality. We also observe an increase in the frequency of dendritic cells displaying an activated phenotype within lymphoid organs in the first 24 hours after irradiation. Intriguingly, both the relative decrease in regulatory T cells and increase in activated dendritic cells correspond with a brief window of augmented responsiveness to immunization. After this 24 hour window, the numbers of dendritic cells decline, as does the ability of mice to respond to immunizations. When immunizations are initiated within the period of augmented dendritic cell activation, mice develop anti-tumor responses that show increased durability as well as magnitude, and this approach leads to improved survival in experiments with mice bearing established tumors as well as in a spontaneous melanoma model. We conclude that irradiation can produce potent immune adjuvant effects independent of its ability to induce tumor ablation, and that the timing of immunization and lymphocyte infusion in the irradiated host are crucial for generating optimal anti-tumor immunity. Clinical strategies using these approaches must therefore optimize such parameters, as the correct timing of infusion and vaccination may mean the difference between an ineffective treatment and successful tumor eradication.


Subject(s)
Cancer Vaccines/immunology , Neoplasms/immunology , Neoplasms/therapy , Adoptive Transfer , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Autoantigens/genetics , Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Dendritic Cells/immunology , Disease Models, Animal , Female , Humans , Immunization , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Melanoma, Experimental , Mice , Mice, Transgenic , Neoplasms/genetics , T-Cell Antigen Receptor Specificity , T-Lymphocytes, Regulatory/immunology , Time Factors , Treatment Outcome , Vaccines, DNA/immunology , Whole-Body Irradiation
4.
PLoS One ; 8(6): e66264, 2013.
Article in English | MEDLINE | ID: mdl-23805207

ABSTRACT

One of the key questions about genomic alterations in cancer is whether they are functional in the sense of contributing to the selective advantage of tumor cells. The frequency with which an alteration occurs might reflect its ability to increase cancer cell growth, or alternatively, enhanced instability of a locus may increase the frequency with which it is found to be aberrant in tumors, regardless of oncogenic impact. Here we've addressed this on a genome-wide scale for cancer-associated focal deletions, which are known to pinpoint both tumor suppressor genes (tumor suppressors) and unstable loci. Based on DNA copy number analysis of over one-thousand human cancers representing ten different tumor types, we observed five loci with focal deletion frequencies above 5%, including the A2BP1 gene at 16p13.3 and the MACROD2 gene at 20p12.1. However, neither RNA expression nor functional studies support a tumor suppressor role for either gene. Further analyses suggest instead that these are sites of increased genomic instability and that they resemble common fragile sites (CFS). Genome-wide analysis revealed properties of CFS-like recurrent deletions that distinguish them from deletions affecting tumor suppressor genes, including their isolation at specific loci away from other genomic deletion sites, a considerably smaller deletion size, and dispersal throughout the affected locus rather than assembly at a common site of overlap. Additionally, CFS-like deletions have less impact on gene expression and are enriched in cell lines compared to primary tumors. We show that loci affected by CFS-like deletions are often distinct from known common fragile sites. Indeed, we find that each tumor tissue type has its own spectrum of CFS-like deletions, and that colon cancers have many more CFS-like deletions than other tumor types. We present simple rules that can pinpoint focal deletions that are not CFS-like and more likely to affect functional tumor suppressors.


Subject(s)
Genome/genetics , Neoplasms/genetics , Sequence Deletion , Animals , Cell Line, Tumor , Chromosome Fragile Sites/genetics , Chromosome Mapping , Chromosomes/genetics , Chromosomes/metabolism , Comparative Genomic Hybridization , DNA Repair Enzymes/genetics , Humans , Hydrolases/genetics , Mice , Neoplasms/physiopathology , RNA Splicing Factors/genetics , Real-Time Polymerase Chain Reaction , Transplantation, Heterologous
5.
J Immunother Cancer ; 1: 20, 2013.
Article in English | MEDLINE | ID: mdl-24829756

ABSTRACT

BACKGROUND: Prior studies show that intramuscular injection and particle-mediated epidermal delivery of xenogeneic melanosomal antigens (tyrosinase or Tyr, gp100) induce CD8(+) T cell responses to the syngeneic protein. To further define the optimal vaccination strategy, we conducted a phase I study of in vivo electroporation (EP) of a murine Tyr DNA vaccine (pINGmuTyr) in malignant melanoma patients. METHODS: Human leukocyte antigen (HLA)-A1, A2, A24 or B35 stage IIb-IV melanoma patients received up to five doses of the mouse tyrosinase DNA vaccine by EP every three weeks at dose levels of 0.2 mg, 0.5 mg, or 1.5 mg per injection. Peripheral blood mononuclear cells (PBMC) were collected, cultured with a peptide pool containing eight HLA class I-restricted Tyr-specific T-cell epitopes, and analyzed by HLA-A*0101-restricted tetramers and intracellular cytokine staining (ICS). RESULTS: Twenty-four patients received ≥1 dose of the pINGmuTyr vaccine; PBMCs from 21 patients who completed all five doses were available for Tyr immune assays. The only common toxicity was grade 1 injection site reaction. Six of 15 patients (40%) in the 1.5 mg dose cohort developed Tyr-reactive CD8(+) T cell responses following stimulation, defined as a ≥3 standard deviation increase in baseline reactivity by tetramer or ICS assays. No Tyr-reactive CD8(+) T cell response was detected in the 0.2 mg and 0.5 mg dose cohort patients. Epitope spreading of CD8(+) T cell response to NY-ESO-1 was observed in one patient with vitiligo. One patient subsequently received ipilimumab and developed an enhanced Tyr-reactive response with polyfunctional cytokine profile. After a median follow-up of 40.9 months, median survival has not been reached. CONCLUSIONS: A regimen of five immunizations with pINGmuTyr administered by EP was found to be safe and resulted in Tyr-reactive immune responses in six of 15 patients at 1.5 mg dose cohort. TRIAL REGISTRATION: ClinicalTrials.gov NCT00471133.

6.
Cancer Immunol Res ; 1(5): 320-31, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24416730

ABSTRACT

Ligation of GITR (glucocorticoid-induced tumor necrosis factor (TNF) receptor-related gene, or TNFRSF18) by agonist antibody has recently entered into early phase clinical trials for the treatment of advanced malignancies. Although the ability of GITR modulation to induce tumor regression is well-documented in preclinical studies, the underlying mechanisms of action, particularly its effects on CD4(+)foxp3(+) regulatory T cells (Treg), have not been fully elucidated. We have previously demonstrated that GITR ligation in vivo by agonist antibody DTA-1 causes a >50% reduction of intra-tumor Treg with down modulation of Foxp3 expression. Here we show that the loss of Foxp3 is tumor-dependent. Adoptively-transferred Foxp3(+)Treg from tumor-bearing animals lose Foxp3 expression in the host when treated with DTA-1, whereas Treg from naïve mice maintain Foxp3 expression. GITR ligation also alters the expression of various transcription factors and cytokines important for Treg function. Complete Foxp3 loss in intra-tumor Treg correlates with a dramatic decrease in Helios expression and is associated with the upregulation of transcription factors T-Bet and Eomes. Changes in Helios correspond with a reduction in IL-10 and an increase in IFNγ expression in DTA-1-treated Treg. Together, these data show that GITR agonist antibody alters Treg lineage stability inducing an inflammatory effector T cell phenotype. The resultant loss of lineage stability causes Treg to lose their intra-tumor immune suppressive function, making the tumor susceptible to killing by tumor-specific effector CD8(+) T cells.


Subject(s)
Glucocorticoid-Induced TNFR-Related Protein/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line, Tumor , Glucocorticoid-Induced TNFR-Related Protein/agonists , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Immune Tolerance , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Regulatory/metabolism
7.
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
8.
PLoS One ; 7(5): e36823, 2012.
Article in English | MEDLINE | ID: mdl-22606294

ABSTRACT

Plasmacytoid dendritic cells (pDCs) play important roles in antiviral innate immunity by producing type I interferon (IFN). In this study, we assess the immune responses of primary human pDCs to two poxviruses, vaccinia and myxoma virus. Vaccinia, an orthopoxvirus, was used for immunization against smallpox, a contagious human disease with high mortality. Myxoma virus, a Leporipoxvirus, causes lethal disease in rabbits, but is non-pathogenic in humans. We report that myxoma virus infection of human pDCs induces IFN-α and TNF production, whereas vaccinia infection does not. Co-infection of pDCs with myxoma virus plus vaccinia blocks myxoma induction effects. We find that heat-inactivated vaccinia (Heat-VAC; by incubating the virus at 55°C for 1 h) gains the ability to induce IFN-α and TNF in primary human pDCs. Induction of IFN-α in pDCs by myxoma virus or Heat-VAC is blocked by chloroquine, which inhibits endosomal acidification required for TLR7/9 signaling, and by inhibitors of cellular kinases PI3K and Akt. Using purified pDCs from genetic knockout mice, we demonstrate that Heat-VAC-induced type I IFN production in pDCs requires the endosomal RNA sensor TLR7 and its adaptor MyD88, transcription factor IRF7 and the type I IFN feedback loop mediated by IFNAR1. These results indicate that (i) vaccinia virus, but not myxoma virus, expresses inhibitor(s) of the poxvirus sensing pathway(s) in pDCs; and (ii) Heat-VAC infection fails to produce inhibitor(s) but rather produces novel activator(s), likely viral RNA transcripts that are sensed by the TLR7/MyD88 pathway. Using vaccinia gene deletion mutants, we show that the Z-DNA/RNA binding domain at the N-terminus of the vaccinia immunomodulatory E3 protein is an antagonist of the innate immune response of human pDCs to poxvirus infection and TLR agonists. The myxoma virus ortholog of vaccinia E3 (M029) lacks the N-terminal Z-DNA/RNA binding domain, which might contribute to the immunostimulating properties of myxoma virus.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , Immunity, Innate , RNA-Binding Proteins/immunology , Vaccinia virus/immunology , Viral Proteins/immunology , Animals , Chloroquine/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Down-Regulation , Humans , Interferon-alpha/biosynthesis , Membrane Glycoproteins/metabolism , Mice , Myeloid Differentiation Factor 88/metabolism , Myxoma virus/genetics , Myxoma virus/immunology , Myxoma virus/pathogenicity , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Rabbits , Toll-Like Receptor 7/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Vaccinia virus/genetics , Vaccinia virus/pathogenicity , Viral Proteins/chemistry , Viral Proteins/genetics
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.
J Virol ; 85(20): 10814-25, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21835795

ABSTRACT

Poxviruses are large DNA viruses that replicate in the cytoplasm of infected cells. Myxoma virus is a rabbit poxvirus that belongs to the Leporipoxvirus genus. It causes a lethal disease called myxomatosis in European rabbits but cannot sustain any detectable infection in nonlagomorphs. Vaccinia virus is a prototypal orthopoxvirus that was used as a vaccine to eradicate smallpox. Myxoma virus is nonpathogenic in mice, whereas systemic infection with vaccinia virus can be lethal even in immunocompetent mice. Plasmacytoid dendritic cells (pDCs) are potent type I interferon (IFN)-producing cells that play important roles in antiviral innate immunity. How poxviruses are sensed by pDCs to induce type I IFN production is not well understood. Here we report that infection of primary murine pDCs with myxoma virus, but not with vaccinia virus, induces IFN-α, IFN-ß, tumor necrosis factor (TNF), and interleukin-12p70 (IL-12p70) production. Using pDCs derived from genetic knockout mice, we show that the myxoma virus-induced innate immune response requires the endosomal DNA sensor TLR9 and its adaptor MyD88, transcription factors IRF5 and IRF7, and the type I IFN positive-feedback loop mediated by IFNAR1. It is independent of the cytoplasmic RNA sensing pathway mediated by the mitochondrial adaptor molecule MAVS, the TLR3 adaptor TRIF, or the transcription factor IRF3. Using pharmacological inhibitors, we demonstrate that myxoma virus-induced type I IFN and IL-12p70 production in murine pDCs is also dependent on phosphatidylinositol 3-kinase (PI3K) and Akt. Furthermore, our results reveal that the N-terminal Z-DNA/RNA binding domain of vaccinia virulence factor E3, which is missing in the orthologous M029 protein expressed by myxoma virus, plays an inhibitory role in poxvirus sensing and innate cytokine production by murine pDCs.


Subject(s)
Dendritic Cells/immunology , Interferon Regulatory Factor-7/immunology , Interferon Regulatory Factors/immunology , Interferon Type I/metabolism , Myeloid Differentiation Factor 88/immunology , Myxoma virus/immunology , Toll-Like Receptor 9/immunology , Animals , Cells, Cultured , Female , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factors/metabolism , Interleukin-12/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Interferon alpha-beta/immunology , Receptor, Interferon alpha-beta/metabolism , Toll-Like Receptor 9/metabolism , Vaccinia virus/immunology
11.
PLoS One ; 6(6): e21214, 2011.
Article in English | MEDLINE | ID: mdl-21731676

ABSTRACT

Determining how tumor immunity is regulated requires understanding the extent to which the anti-tumor immune response "functions" in vivo without therapeutic intervention. To better understand this question, we developed advanced multimodal reflectance confocal/two photon fluorescence intra-vital imaging techniques to use in combination with traditional ex vivo analysis of tumor specific T cells. By transferring small numbers of melanoma-specific CD8+ T cells (Pmel-1), in an attempt to mimic physiologic conditions, we found that B16 tumor growth alone was sufficient to induce naive Pmel-1 T cell proliferation and acquisition of effector phenotype. Tumor -primed Pmel-1 T cells, are capable of killing target cells in the periphery and secrete IFNγ, but are unable to mediate tumor regression. Within the tumor, Pmel-1 T cells have highly confined mobility, displaying long term interactions with tumor cells. In contrast, adoptively transferred non tumor-specific OT-I T cells show neither confined mobility, nor long term interaction with B16 tumor cells, suggesting that intra-tumor recognition of cognate self antigen by Pmel-1 T cells occurs during tumor growth. Together, these data indicate that lack of anti-tumor efficacy is not solely due to ignorance of self antigen in the tumor microenvironment but rather to active immunosuppressive influences preventing a protective immune response.


Subject(s)
Antigens, Neoplasm/immunology , Autoantigens/immunology , Disease Progression , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Microscopy, Confocal/methods , Adoptive Transfer , Animals , Cell Communication , Cell Proliferation , Epitopes/immunology , Kinetics , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Time Factors
12.
Clin Cancer Res ; 17(7): 1984-97, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21355077

ABSTRACT

PURPOSE: We compared the efficacy of human Langerhans cells (LC) as tumor immunogens in vivo with monocyte-derived dendritic cells (moDC) and investigated how interleukin 15 (IL15) supports optimal DC-stimulated antitumor immunity. EXPERIMENTAL DESIGN: American Joint Committee on Cancer stage III/IV melanoma patients participated in this first clinical trial comparing melanoma peptide-pulsed LC with moDC vaccines (NCT00700167, www.ClinicalTrials.gov). Correlative studies evaluated mechanisms mediating IL15 support of DC-stimulated antitumor immunity. RESULTS: Both DC vaccines were safe and immunogenic for melanoma antigens. LC-based vaccines stimulated significantly greater tyrosinase-HLA-A*0201 tetramer reactivity than the moDC-based vaccines. The two DC subtypes were otherwise statistically comparable, in contrast to extensive prior data in vitro showing LC superiority. LCs synthesize much more IL15 than moDCs and stimulate significantly more antigen-specific lymphocytes with a cytolytic IFN-γ profile even without exogenous IL15. When supplemented by low-dose IL15, instead of IL2, moDCs stimulate 5 to 6 logs more tumor antigen-specific effector memory T cells (T(EMRA)) over 3 to 4 weeks in vitro. IL2 and IL15 can be synergistic in moDC stimulation of cytolytic T cells. IL15 promotes T-cell expression of the antiapoptotic bcl-2 and inhibits candidate regulatory T-cell (Treg) expansion after DC stimulation, countering two effects of IL2 that do not foster tumor immunity. CONCLUSIONS: MoDC-based vaccines will require exogenous IL15 to achieve clinical efficacy. Alternatively, LCs can couple the endogenous production of IL15 with potent T-cell stimulatory activity. Optimization of full-length tumor antigen expression for processing into multiple immunogenic peptides for presentation by both class I and II MHC therefore merits emphasis to support more effective antitumor immunity stimulated by LCs.


Subject(s)
Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Immunotherapy, Active , Interleukin-15/metabolism , Langerhans Cells/immunology , Melanoma/therapy , T-Lymphocytes, Cytotoxic/immunology , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/immunology , Apoptosis/drug effects , CD3 Complex/metabolism , CD8 Antigens/metabolism , Cancer Vaccines/adverse effects , Dendritic Cells/drug effects , Drug Synergism , Erythema/etiology , Female , HLA-A Antigens/metabolism , HLA-A2 Antigen , Humans , Interferon-gamma/metabolism , Interleukin-15/immunology , Interleukin-15/pharmacology , Interleukin-2/immunology , Interleukin-2/pharmacology , Langerhans Cells/drug effects , Lymphocyte Activation , Male , Melanoma/immunology , Middle Aged , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/physiology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/physiology
13.
Vaccine ; 29(20): 3646-54, 2011 May 09.
Article in English | MEDLINE | ID: mdl-21435405

ABSTRACT

HER2/neu is an oncogene amplified and over-expressed in 20-30% of breast adenocarcinomas. Treatment with the humanized monoclonal antibody trastuzumab has shown efficacy in combination with cytotoxic agents, although resistance occurs over time. Novel approaches are needed to further increase antibody efficacy. In this study, we provide evidence in a mouse breast cancer therapeutic tumor model that the combination of active immunization with a modified HER2/neu DNA vaccine and passive infusion of an anti-HER2/neu monoclonal antibody leads to significant regression of established tumors. Our data indicate that combination therapy with a HER2/neu DNA vaccine and trastuzumab may have clinical activity in breast cancer patients.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cancer Vaccines/immunology , Epitopes/immunology , Mammary Neoplasms, Experimental/therapy , Receptor, ErbB-2/immunology , Adenocarcinoma/immunology , Adenocarcinoma/therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/immunology , Cancer Vaccines/administration & dosage , Female , Immunotherapy , Lymphocytes, Tumor-Infiltrating/immunology , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Mutagenesis, Site-Directed , Trastuzumab , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
14.
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
15.
Clin Cancer Res ; 16(15): 4057-65, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20647477

ABSTRACT

PURPOSE: Prior studies show that i.m. injection of xenogeneic orthologues of melanosomal antigens (tyrosinase, gp100) induces CD8(+) T-cell responses to the syngeneic protein. To further define the optimal vaccination strategy, we conducted a pilot clinical trial comparing i.m. injection with particle-mediated epidermal delivery (PMED). EXPERIMENTAL DESIGN: Human leukocyte antigen (HLA)-A*0201(+) disease-free melanoma patients were randomized to the PMED or i.m. arm, receiving eight vaccinations over 4 months. Patients received 4 microg or 2,000 microg per injection, respectively, of mouse gp100 DNA. Peripheral blood mononuclear cells were collected, cultured with gp100 peptides, and analyzed by tetramer and intracellular cytokine staining for responses to HLA-A*0201-restricted gp100 epitopes [gp100(209-217) (ITDQVPFSV) and gp100(280-288) (YLEPGPVTA)]. RESULTS: Twenty-seven patients with stage IIB-IV melanoma were analyzable for immune response. The only common toxicity was grade 1 injection site reaction in nine patients with no intergroup difference, and one dose-limiting toxicity of acute hypersensitivity occurred in a PMED patient with undiagnosed gold allergy. Four of 27 patients produced gp100 tetramer(+)CD8(+) T cells, all carrying the CCR7(lo)CD45RA(lo) effector-memory phenotype. Five of 27 patients generated IFN-gamma(+)CD8(+) T cells, one who was also tetramer-positive. Overall, vaccination induced a response in 30% of patients, which was not significantly associated with study arm or clinical outcome. However, the PMED group showed a trend toward increased IFN-gamma(+)CD8(+) T-cell generation (P = 0.07). CONCLUSION: A comparable efficacy and safety profile was shown between the i.m. and PMED arms, despite a significantly decreased dose of DNA used for PMED injection.


Subject(s)
Biolistics , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Melanoma/therapy , Membrane Glycoproteins/administration & dosage , Administration, Intranasal , Adult , Aged , Animals , Antigens, Heterophile/administration & dosage , Antigens, Heterophile/adverse effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/adverse effects , DNA/administration & dosage , DNA/adverse effects , DNA/immunology , Female , HLA-A Antigens , HLA-A2 Antigen , Humans , Kaplan-Meier Estimate , Male , Melanoma/mortality , Melanoma/pathology , Membrane Glycoproteins/adverse effects , Membrane Glycoproteins/immunology , Mice , Middle Aged , Neoplasm Staging , Peptide Fragments , Peptides , Pilot Projects , Vaccines, DNA/administration & dosage , Vaccines, DNA/adverse effects , Vaccines, DNA/immunology , gp100 Melanoma Antigen
16.
PLoS One ; 5(5): e10436, 2010 May 03.
Article in English | MEDLINE | ID: mdl-20454651

ABSTRACT

In vivo GITR ligation has previously been shown to augment T-cell-mediated anti-tumor immunity, yet the underlying mechanisms of this activity, particularly its in vivo effects on CD4+ foxp3+ regulatory T cells (Tregs), have not been fully elucidated. In order to translate this immunotherapeutic approach to the clinic it is important gain better understanding of its mechanism(s) of action. Utilizing the agonist anti-GITR monoclonal antibody DTA-1, we found that in vivo GITR ligation modulates regulatory T cells (Tregs) directly during induction of melanoma tumor immunity. As a monotherapy, DTA-1 induced regression of small established B16 melanoma tumors. Although DTA-1 did not alter systemic Treg frequencies nor abrogate the intrinsic suppressive activity of Tregs within the tumor-draining lymph node, intra-tumor Treg accumulation was significantly impaired. This resulted in a greater Teff:Treg ratio and enhanced tumor-specific CD8+ T-cell activity. The decreased intra-tumor Treg accumulation was due both to impaired infiltration, coupled with DTA-1-induced loss of foxp3 expression in intra-tumor Tregs. Histological analysis of B16 tumors grown in Foxp3-GFP mice showed that the majority of GFP+ cells had lost Foxp3 expression. These "unstable" Tregs were absent in IgG-treated tumors and in DTA-1 treated TDLN, demonstrating a tumor-specific effect. Impairment of Treg infiltration was lost if Tregs were GITR(-/-), and the protective effects of DTA-1 were reduced in reconstituted RAG1(-/-) mice if either the Treg or Teff subset were GITR-negative and absent if both were negative. Our results demonstrate that DTA-1 modulates both Teffs and Tregs during effective tumor treatment. The data suggest that DTA-1 prevents intra-tumor Treg accumulation by altering their stability, and as a result of the loss of foxp3 expression, may modify their intra-tumor suppressive capacity. These findings provide further support for the continued development of agonist anti-GITR mAbs as an immunotherapeutic strategy for cancer.


Subject(s)
Antibodies, Monoclonal/immunology , Immunity/immunology , Melanoma/immunology , Receptors, Nerve Growth Factor/agonists , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/agonists , Receptors, Tumor Necrosis Factor/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation , Forkhead Transcription Factors/metabolism , Glucocorticoid-Induced TNFR-Related Protein , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Up-Regulation/immunology
17.
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
18.
Cytotherapy ; 11(7): 912-22, 2009.
Article in English | MEDLINE | ID: mdl-19903103

ABSTRACT

BACKGROUND AIMS: Monitoring cellular immune responses is one prerequisite for the rational development of cancer vaccines. METHODS: We describe an extensive effort to optimize and validate quantitatively an in vitro T-cell culture method by determining the phenotype and function of both CD4(+) and CD8(+) T cells, including measurement of the phenotype markers CCR7, CD45RA, CD28 and CD27 and the functional markers interferon (IFN)-gamma, interleukin (IL)-2, macrophage inflammatory protein (MIP)-1beta, tumor necrosis factor (TNF)-alpha and CD107a. RESULTS: Autologous peripheral blood mononuclear cells (PBMC) were potent stimulators that expanded antigen (Ag)-specific CD8(+) T cells during short-term culture with the addition of IL-2 and IL-15 cytokines. Polyfunctional Ag-specific CD4(+) and CD8(+) T cells were detectable using this method. CONCLUSIONS: Our culture system represents a robust human T-cell culture protocol that permits phenotypic, quantitative and qualitative evaluation of vaccine-induced CD4(+) and CD8(+) T-cell responses.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Culture Techniques , Neoplasms/immunology , Antigens/immunology , Antigens, CD/metabolism , Antigens, Differentiation/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines , Cells, Cultured , Cytokines/metabolism , Feasibility Studies , Humans , Neoplasms/therapy
19.
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
20.
Cancer Immun ; 9: 5, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19496531

ABSTRACT

A differentiation antigen commonly expressed on melanoma cells, gp100 is the target of infiltrating T cells. We conducted a phase I randomized cross-over trial of melanoma patients with either xenogeneic (mouse) or human gp100 plasmid DNA injected intramuscularly at three dosages (100, 500 or 1,500 microg) every three weeks for three doses. After the first three injections, patients were then immunized three times with gp100 from the other species. Peripheral blood samples were analyzed at various time points following 10-day culture with gp100 peptides using multi-parametric flow cytometry. A total of 19 patients were enrolled, with 18 assessable for immune function and survival. 14 (74%) were male, with a median age of 56 years (range, 20-82). All patients had no evidence of disease; 10 (53%) had stage III disease, 3 each (16%) had stage IIB and IV disease, 2 (11%) had choroidal and 1 (5%) had anal mucosal involvement. With a median follow-up of 30 months, median progression-free survival (PFS) is 44 months. Median survival is not reached. There was no grade 3/4 toxicity; the most common grade 1/2 toxicity was an injection site reaction in 12 patients (63%, all grade 1). Five patients developed CD8+ cells binding gp100(280-288) HLA-A2-restricted tetramer. One patient had an increase in CD8+ IFN-gamma+ cells. This xenogeneic immunization strategy was safe and associated with minimal toxicity. There was also evidence of immune response.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Melanoma/immunology , Membrane Glycoproteins/immunology , Skin Neoplasms/immunology , T-Lymphocytes/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antigens, Neoplasm/genetics , Cancer Vaccines/adverse effects , Disease-Free Survival , Follow-Up Studies , HLA-A2 Antigen , Humans , Injections, Intramuscular , Lymphocyte Activation , Male , Melanoma/therapy , Membrane Glycoproteins/genetics , Mice , Middle Aged , Neoplasm Staging , Peptide Fragments , Peptides , Skin Neoplasms/therapy , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Vaccines, DNA/administration & dosage , Vaccines, DNA/adverse effects , gp100 Melanoma Antigen
SELECTION OF CITATIONS
SEARCH DETAIL
...