Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Immunol ; 195(7): 3482-9, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26320256

ABSTRACT

Maintaining antitumor immunity remains a persistent impediment to cancer immunotherapy. We and others have previously reported that high-avidity CD8(+) T cells are more susceptible to tolerance induction in the tumor microenvironment. In the present study, we used a novel model where T cells derived from two independent TCR transgenic mouse lines recognize the same melanoma antigenic epitope but differ in their avidity. We tested whether providing CD4(+) T cell help would improve T cell responsiveness as a function of effector T cell avidity. Interestingly, delivery of CD4(+) T cell help during in vitro priming of CD8(+) T cells improved cytokine secretion and lytic capacity of high-avidity T cells, but not low-avidity T cells. Consistent with this observation, copriming with CD4(+) T cells improved antitumor immunity mediated by higher avidity, melanoma-specific CD8(+) T cells, but not T cells with similar specificity but lower avidity. Enhanced tumor immunity was associated with improved CD8(+) T cell expansion and reduced tolerization, and it was dependent on presentation of both CD4(+) and CD8(+) T cell epitopes by the same dendritic cell population. Our findings demonstrate that CD4(+) T cell help preferentially augments high-avidity CD8(+) T cells and provide important insight for understanding the requirements to elicit and maintain durable tumor immunity.


Subject(s)
Antibody Affinity/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Immune Tolerance/immunology , Animals , Antibody Affinity/genetics , Antigens, Neoplasm/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , Lymphocyte Activation/immunology , Melanoma/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic
2.
Mol Ther ; 22(1): 206-18, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24196579

ABSTRACT

Despite clear evidence of immunogenicity, cancer vaccines only provide a modest clinical benefit. To evaluate the mechanisms that limit tumor regression following vaccination, we have investigated the weak efficacy of a highly immunogenic experimental vaccine using a murine melanoma model. We discovered that the tumor adapts rapidly to the immune attack instigated by tumor-specific CD8+ T cells in the first few days following vaccination, resulting in the upregulation of a complex set of biological networks, including multiple immunosuppressive processes. This rapid adaptation acts to prevent sustained local immune attack, despite continued infiltration by increasing numbers of tumor-specific T cells. Combining vaccination with adoptive transfer of tumor-specific T cells produced complete regression of the treated tumors but did not prevent the adaptive immunosuppression. In fact, the adaptive immunosuppressive pathways were more highly induced in regressing tumors, commensurate with the enhanced level of immune attack. Examination of tumor infiltrating T-cell functionality revealed that the adaptive immunosuppression leads to a progressive loss in T-cell function, even in tumors that are regressing. These novel observations that T cells produced by therapeutic intervention can instigate a rapid adaptive immunosuppressive response within the tumor have important implications for clinical implementation of immunotherapies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Adaptive Immunity/genetics , Adenoviruses, Human/genetics , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/immunology , Female , Genetic Vectors/genetics , Genetic Vectors/immunology , Immunomodulation/genetics , Immunomodulation/immunology , Immunotherapy, Adoptive , Interferon-gamma/immunology , Interferon-gamma/metabolism , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Neoplasm Recurrence, Local , Neoplasms/genetics , Neoplasms/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Burden/genetics , Tumor Burden/immunology , Vaccination , Vaccines, Synthetic
3.
Nucleic Acids Res ; 40(8): 3689-703, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22210864

ABSTRACT

We previously reported that miR-1 is among the most consistently down-regulated miRs in primary human prostate tumors. In this follow-up study, we further corroborated this finding in an independent data set and made the novel observation that miR-1 expression is further reduced in distant metastasis and is a candidate predictor of disease recurrence. Moreover, we performed in vitro experiments to explore the tumor suppressor function of miR-1. Cell-based assays showed that miR-1 is epigenetically silenced in human prostate cancer. Overexpression of miR-1 in these cells led to growth inhibition and down-regulation of genes in pathways regulating cell cycle progression, mitosis, DNA replication/repair and actin dynamics. This observation was further corroborated with protein expression analysis and 3'-UTR-based reporter assays, indicating that genes in these pathways are either direct or indirect targets of miR-1. A gene set enrichment analysis revealed that the miR-1-mediated tumor suppressor effects are globally similar to those of histone deacetylase inhibitors. Lastly, we obtained preliminary evidence that miR-1 alters the cellular organization of F-actin and inhibits tumor cell invasion and filipodia formation. In conclusion, our findings indicate that miR-1 acts as a tumor suppressor in prostate cancer by influencing multiple cancer-related processes and by inhibiting cell proliferation and motility.


Subject(s)
Biomarkers, Tumor/metabolism , MicroRNAs/metabolism , Prostatic Neoplasms/genetics , Biomarkers, Tumor/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA Repair/genetics , Epigenesis, Genetic , Genes, Tumor Suppressor , Histone Deacetylase Inhibitors/pharmacology , Humans , Male , MicroRNAs/genetics , Mitosis , Neoplasm Metastasis , Neoplasm Recurrence, Local/diagnosis , Prognosis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
4.
Cancer Immunol Immunother ; 61(2): 289-293, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22237887

ABSTRACT

Immune suppression remains a consistent obstacle to successful anti-tumor immune responses. As tumors develop, they create a microenvironment that not only supports tumor growth and metastasis but also reduces potential adaptive immunity to tumor antigens. Among the many components of this tumor microenvironment is a population of dendritic cells which exert profound immune suppressive effects on T cells. In this review, we discuss our recent findings related to these tumor-associated dendritic cells and how targeting them may serve to generate more durable anti-tumor immune responses.


Subject(s)
Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Immunotherapy , Neoplasms/therapy , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Humans , Immune Tolerance , Immunosuppression Therapy , Neoplasms/immunology , Tumor Escape , Tumor Microenvironment/immunology
5.
J Immunol ; 185(1): 174-82, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20525892

ABSTRACT

Our previous study showed that TNFR2 is preferentially expressed by CD4(+)FoxP3(+) regulatory T cells (Tregs), and expression of this receptor identified maximally suppressive Tregs. TNFR2 is also expressed by a small fraction of CD4(+)FoxP3(-) conventional T cells (Tconvs) in normal mice, and its expression is upregulated by T cell activation. This raises questions about the role of TNFR2 signaling in the function of Tconv cells. In this study, by using FoxP3/gfp knock-in mice, we showed that TNFR2 signaling did not induce FoxP3(-) CD4 cells to become suppressive. Ki-67, a marker of proliferation, was concomitantly expressed with TNFR2 by CD4 cells, independent of forkhead box P3 expression, in normal mice and Lewis lung carcinoma-bearing mice. TNFR2 is associated with greater suppressive functions when expressed by Tregs and is associated with greater resistance to suppression when expressed by Tconv cells. In mice bearing 4T1 breast tumor or Lewis lung carcinoma, intratumoral Tconv cells expressing elevated levels of TNFR2 acquired the capacity to resist suppression by lymph node-derived Tregs. However, they remained susceptible to inhibition by more suppressive tumor-infiltrating Tregs, which expressed higher levels of TNFR2. Our data indicate that TNFR2 also costimulates Tconv cells. However, intratumoral Tregs expressing more TNFR2 are able to overcome the greater resistance to suppression of intratumoral Tconv cells, resulting in a dominant immunosuppressive tumor environment.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Forkhead Transcription Factors , Immunity, Innate , Lymphocyte Activation/immunology , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/physiology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/prevention & control , Cell Line, Tumor , Cells, Cultured , Clonal Anergy/genetics , Clonal Anergy/immunology , Coculture Techniques , Female , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/deficiency , Gene Knock-In Techniques , Immunity, Innate/genetics , Lymphocyte Activation/genetics , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor, Type II/genetics , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
7.
J Immunol ; 183(8): 4848-52, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19801511

ABSTRACT

We previously reported that naive, tumor-specific CD8(+) (TcR-I) T cells transferred into prostate tumor-bearing mice traffic to the prostate where they become tolerized. We now report that TcR-I cells suppress the proliferation of naive T cells. This suppression is mediated at least in part by secreted factors, and the suppressive activity can be blocked by Abs directed against TGF-beta. We further report that TcR-I cells must infiltrate the prostate to acquire suppressive activity. Delivery of tumor-specific CD4(+) T cells prevents the conversion of TcR-I cells into suppressor cells. Taken together, our findings may have critical implications for sustaining T cell responsiveness during immunotherapy, as the development of suppressor cells in the tumor microenvironment may eliminate the potency of T cells primed in the periphery or delivered during adoptive immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Prostatic Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Animals , Antibodies/immunology , CD8-Positive T-Lymphocytes/transplantation , Immune Tolerance , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/transplantation , Male , Mice , Mice, Transgenic , Prostatic Neoplasms/therapy , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism
8.
J Immunol ; 182(7): 4328-38, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19299733

ABSTRACT

IL-27 exerts antitumor activity in murine orthotopic neuroblastoma, but only partial antitumor effect in disseminated disease. This study demonstrates that combined treatment with IL-2 and IL-27 induces potent antitumor activity in disseminated neuroblastoma metastasis. Complete durable tumor regression was achieved in 90% of mice bearing metastatic TBJ-IL-27 tumors treated with IL-2 compared with only 40% of mice bearing TBJ-IL-27 tumors alone and 0% of mice bearing TBJ-FLAG tumors with or without IL-2 treatment. Comparable antitumor effects were achieved by IL-27 protein produced upon hydrodynamic IL-27 plasmid DNA delivery when combined with IL-2. Although delivery of IL-27 alone, or in combination with IL-2, mediated pronounced regression of neuroblastoma metastases in the liver, combined delivery of IL-27 and IL-2 was far more effective than IL-27 alone against bone marrow metastases. Combined exposure to IL-27 produced by tumor and IL-2 synergistically enhances the generation of tumor-specific CTL reactivity. Potentiation of CTL reactivity by IL-27 occurs via mechanisms that appear to be engaged during both the initial sensitization and effector phase. Potent immunologic memory responses are generated in mice cured of their disseminated disease by combined delivery of IL-27 and IL-2, and depletion of CD8(+) ablates the antitumor efficacy of this combination. Moreover, IL-27 delivery can inhibit the expansion of CD4(+)CD25(+)Foxp3(+) regulatory and IL-17-expressing CD4(+) cells that are otherwise observed among tumor-infiltrating lymphocytes from mice treated with IL-2. These studies demonstrate that IL-27 and IL-2 synergistically induce complete tumor regression and long-term survival in mice bearing widely metastatic neuroblastoma tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/immunology , Interleukin-2/immunology , Interleukins/immunology , Lymphocyte Activation/drug effects , Neuroblastoma/immunology , T-Lymphocytes, Cytotoxic/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bone Marrow Neoplasms/drug therapy , Bone Marrow Neoplasms/secondary , Drug Synergism , Flow Cytometry , Interferon-gamma/immunology , Interleukin-2/administration & dosage , Interleukins/administration & dosage , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Neuroblastoma/drug therapy , Neuroblastoma/secondary , T-Lymphocytes, Cytotoxic/immunology
9.
Elife ; 102021 04 30.
Article in English | MEDLINE | ID: mdl-33929324

ABSTRACT

Self-specific CD8+T cells can escape clonal deletion, but the properties and capabilities of such cells in a physiological setting are unclear. We characterized polyclonal CD8+ T cells specific for the melanocyte antigen tyrosinase-related protein 2 (Trp2) in mice expressing or lacking this enzyme (due to deficiency in Dct, which encodes Trp2). Phenotypic and gene expression profiles of pre-immune Trp2/Kb-specific cells were similar; the size of this population was only slightly reduced in wild-type (WT) compared to Dct-deficient (Dct-/-) mice. Despite comparable initial responses to Trp2 immunization, WT Trp2/Kb-specific cells showed blunted expansion and less readily differentiated into a CD25+proliferative population. Functional self-tolerance clearly emerged when assessing immunopathology: adoptively transferred WT Trp2/Kb-specific cells mediated vitiligo much less efficiently. Hence, CD8+ T cell self-specificity is poorly predicted by precursor frequency, phenotype, or even initial responsiveness, while deficient activation-induced CD25 expression and other gene expression characteristics may help to identify functionally tolerant cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Self Tolerance , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Female , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Male , Mice , Mice, Inbred C57BL , Vitiligo/immunology
10.
Blood ; 112(8): 3283-92, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18664628

ABSTRACT

Interleukin-7 (IL-7), a cytokine produced by stromal cells, is required for thymic development and peripheral homeostasis of most major subsets of T cells. We examined whether regulatory T (Treg) cells also required the IL-7 pathway by analyzing IL-7Ralpha(-/-) mice. We observed a striking reduction in cells with the Treg surface phenotype (CD4, CD25, GITR (glucocorticoid-induced tumor necrosis factor [TNF]-like receptor), CD45RB, CD62L, CD103) or intracellular markers (cytotoxic T-lymphocyte-associated antigen-4, CTLA-4, and forkhead box transcription factor 3, Foxp3). Foxp3 transcripts were virtually absent in IL-7Ralpha(-/-) lymphoid tissues, and no Treg cell suppressive activity could be detected. There are 2 known ligands for IL-7Ralpha: IL-7 itself and thymic stromal lymphopoietin (TSLP). Surprisingly, mice deficient in IL-7 or the other chain of the TSLP receptor (TSLPR) developed relatively normal numbers of Treg cells. Combined deletion of IL-7 and TSLP receptor greatly reduced Treg cell development in the thymus but was not required for survival of mature peripheral Treg cells. We conclude that Treg cells, like other T cells, require signals from the IL-7 receptor, but unlike other T cells, do not require IL-7 itself because of at least partially overlapping actions of IL-7 and TSLP for development of Treg cells.


Subject(s)
Cytokines/metabolism , Interleukin-7/metabolism , Receptors, Interleukin-7/metabolism , T-Lymphocytes, Regulatory/cytology , Animals , Cell Membrane/metabolism , Cell Separation , Female , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/cytology , Thymic Stromal Lymphopoietin
12.
J Leukoc Biol ; 84(4): 973-80, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18653463

ABSTRACT

IL-2 is essential for CD4+CD25+forkhead box P3+ (FoxP3+) naturally occurring regulatory T cell (Treg) homeostasis and activation. Binding of IL-2 to its receptor leads to phosphorylation of STAT5, and binding of phosphorylated STAT5 to the foxp3 promoter increases foxp3 transcription, resulting in elevated levels of FoxP3 protein in Tregs. Transcriptional regulation by the elevated levels of FoxP3 is thought to be essential for the strong suppressor function seen in activated Tregs. IL-2 belongs to a family cytokines, which all depend on the common gamma-receptor chain (gammac). Given the well-documented effects of IL-2 on Treg function, the effect of other IL-2 family cytokines (IL-7, -15, and -21) on Tregs was examined. We observed that IL-7 and IL-15 induce STAT5 phosphorylation and up-regulation of FoxP3 in Tregs. STAT5 activation correlated with enhanced viability. However, only in the presence of IL-2 did Tregs acquire potent suppressor function. This finding is surprising, as IL-15 as well as IL-2 use the same IL-2R betac and gammac for signaling. In contrast, IL-21 activated STAT3 but did not activate STAT5 and had no effect on Treg viability, activation, or function. We therefore conclude that phosphorylation of STAT5, mediated through the IL-2Rgamma, promotes Treg survival in a resting and activated state. However, activation of STAT5 alone in conjunction with TCR signaling is not sufficient for the induction of potent suppressor function in Tregs, as IL-7 and IL-15 are not capable of inducing potent Treg suppressor function.


Subject(s)
Cytokines/pharmacology , Interleukin-2/pharmacology , Lymphocyte Activation/drug effects , T-Lymphocytes, Regulatory/immunology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Crosses, Genetic , Forkhead Transcription Factors/drug effects , Gene Expression Regulation/drug effects , Interleukin-2 Receptor alpha Subunit/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Receptors, Interleukin-15/genetics , Receptors, Interleukin-21/genetics , Receptors, Interleukin-7/genetics , Spleen/immunology , T-Lymphocytes, Regulatory/drug effects , Transcription, Genetic/drug effects
13.
PLoS One ; 13(5): e0197356, 2018.
Article in English | MEDLINE | ID: mdl-29771971

ABSTRACT

Transplacental viral and parasitic infections have been shown to initiate an innate response in the mammalian embryo by increasing the expression of pro-inflammatory cytokines such as interferon-gamma (Ifng). However, the developmental consequences of an activated innate immunity and, in particular, the effects of induction of Ifng expression independent of infection have been largely overlooked. Here, we demonstrate in vivo that the conditional overexpression of Ifng in metanephric mesenchymal (MM) progenitors results in renal agenesis or hypoplasia. Cell death was observed in and around the MM region of E10.5-11.5 mutants where Ifng was constitutively expressed during early kidney development and resulted in a retardation of branching morphogenesis. Furthermore, isolated mutant or normal Ifng-treated metanephroi replicated this phenotype in culture, demonstrating the inherent nature of the aberrant morphogenesis. The expression of renal progenitor marker Sall1 was significantly decreased in the MM of mutant kidneys, suggesting that a reduction in Sall1 may be the cause of cell death in the MM during early kidney development and that, in turn, retards UB branching in the mutants. Therefore, the aberrant induction of Ifng expression, as part of an innate immune response, may contribute to renal agenesis or hypoplasia during early metanephric development by regulating the MM progenitor population.


Subject(s)
Interferon-gamma/metabolism , Kidney/abnormalities , Kidney/embryology , Mesenchymal Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Death/physiology , Gene Expression Regulation, Developmental , Interferon-gamma/genetics , Kidney/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Organogenesis/physiology , Tissue Culture Techniques
14.
Adv Exp Med Biol ; 601: 357-68, 2007.
Article in English | MEDLINE | ID: mdl-17713024

ABSTRACT

It is widely recognized that the immune system plays a role in cancer progression and that some tumors are inherently immunogenic. The identification of tumor-associated antigens (TAAs) has stimulated research focused on immunotherapies to mediate the regression of established tumors. Cancer-specific immunity has traditionally been aimed at activating CD8+ cytotoxic T lymphocytes (CTLs) directed against major histocompatibility complex (MHC) class I-binding peptide epitopes. Other approaches utilize T cell adoptive therapy where autologous, tumor-specific T cells propagated in vitro are transferred back into recipients. However, these strategies have met with limited success in part due to the regulatory mechanisms of T cell tolerance, which poses a considerable challenge to cancer immunotherapy. Our laboratory utilizes the TRansgenic Adenocarcinoma of the Mouse Prostate (TRAMP) model, a murine model of prostate cancer, to study mechanisms of T cell tolerization to tumor antigens. We previously demonstrated that upon encounter with their cognate antigen in the tumor microenvironment, naive T cell become tolerized. Our ongoing studies are testing whether provision of CD4+ T cells can enhance tumor immunity by preventing CD8+ T cell tolerance. A greater understanding of the interaction between various tumor-specific T cell subsets will facilitate the design of novel approaches to stimulate a more potent antitumor immune response.


Subject(s)
Immune System/physiology , Immune Tolerance , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Animals , Antigens, Neoplasm/chemistry , Epitopes/chemistry , Humans , Major Histocompatibility Complex , Male , Mice , Models, Biological , Prostatic Neoplasms/immunology
15.
Cancer Res ; 76(5): 1055-1065, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26719530

ABSTRACT

Smokers develop metastatic prostate cancer more frequently than nonsmokers, suggesting that a tobacco-derived factor is driving metastatic progression. To identify smoking-induced alterations in human prostate cancer, we analyzed gene and protein expression patterns in tumors collected from current, past, and never smokers. By this route, we elucidated a distinct pattern of molecular alterations characterized by an immune and inflammation signature in tumors from current smokers that were either attenuated or absent in past and never smokers. Specifically, this signature included elevated immunoglobulin expression by tumor-infiltrating B cells, NF-κB activation, and increased chemokine expression. In an alternate approach to characterize smoking-induced oncogenic alterations, we also explored the effects of nicotine in human prostate cancer cells and prostate cancer-prone TRAMP mice. These investigations showed that nicotine increased glutamine consumption and invasiveness of cancer cells in vitro and accelerated metastatic progression in tumor-bearing TRAMP mice. Overall, our findings suggest that nicotine is sufficient to induce a phenotype resembling the epidemiology of smoking-associated prostate cancer progression, illuminating a novel candidate driver underlying metastatic prostate cancer in current smokers.


Subject(s)
Inflammation/metabolism , Prostatic Neoplasms/immunology , Smoking/adverse effects , Transcriptome , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Immunoglobulins/genetics , Interleukin-8/blood , Male , Mice , NF-kappa B/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Nicotine/pharmacology , Prostatic Neoplasms/etiology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism
16.
BioDrugs ; 17(2): 131-8, 2003.
Article in English | MEDLINE | ID: mdl-12641491

ABSTRACT

The absence of curative therapies for advanced or recurrent forms of prostate cancer has prompted a vigorous search for novel treatment strategies. Immunotherapy encompasses one particularly promising systemic approach to the treatment of prostate cancer. Immune-based strategies for treating prostate cancer have recently been facilitated by the identification of a number of prostate tissue/tumour antigens that can be targeted, either by antibody or T cells, to promote prostate tumour cell injury or death. These same prostate antigens can also be used for the construction of vaccines to induce prostate-specific T cell-mediated immunity. Greater insight into specific mechanisms that govern antigen-specific T cell activation has brought with it a number of innovative methods to induce and enhance T cell-mediated responses against prostate tumours. For instance, autologous dendritic cells loaded with prostate antigens have proved useful to induce prostate-specific T cell activation. Similarly, in vivo manipulations of T cell costimulatory pathway receptors can greatly facilitate tumour-specific T cell activation and potentiate T cell-mediated responses against a number of malignancies, including prostate cancer. For example, blocking T cell cytotoxic lymphocyte-associated antigen 4 (CTLA-4) receptor binding to its ligand prevents the down-regulation of T cell responses and can even potentiate T cell antitumoural immunity in mouse models of prostate cancer. Androgen ablation (AA) may induce prostate tumour/tissue-specific T cell mediated inflammation and, as such, a phase II trial is currently in progress to ascertain whether CTLA-4 blockade can enhance AA-induced treatment responses in patients with advanced prostate cancer. Nevertheless, further basic and clinical investigation is still required to establish immunotherapy as a true prostate cancer treatment option.


Subject(s)
Immunotherapy , Prostatic Neoplasms/therapy , Androgens/immunology , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Cancer Vaccines/therapeutic use , Cytokines/therapeutic use , Dendritic Cells/immunology , Humans , Male , Prostatic Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology
17.
Methods Mol Med ; 102: 421-7, 2004.
Article in English | MEDLINE | ID: mdl-15286398

ABSTRACT

Generating an antitumor immune response can be thought of as eliciting an immune response to cells derived from self-tissue. As such, tumor immunity may result in autoimmunity. Melanoma patients undergoing immunotherapy often develop a form of autoimmune depigmentation referred to as vitiligo, in which T cells with antigenic specificity for pigmentation antigens destroy normal melanocytes. The models described in this chapter can be used to study immunity to melanoma antigens. These models employ a well-characterized pigmentation antigen relevant to melanoma and a common transplantable murine melanoma cell line. As more sophisticated approaches to cancer therapy are developed, models such as these may be key in understanding how immunity to self-antigens can be manipulated to elicit tumor immunity.


Subject(s)
Antigens, Neoplasm/administration & dosage , Autoimmunity , Hypopigmentation/etiology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Animals , Autoantigens , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Humans , Hypopigmentation/immunology , Hypopigmentation/pathology , Immunodominant Epitopes/administration & dosage , Intramolecular Oxidoreductases/administration & dosage , Intramolecular Oxidoreductases/immunology , Melanoma, Experimental/therapy , Mice , Peptide Fragments/administration & dosage , Peptide Fragments/immunology
18.
Cancer Microenviron ; 7(1-2): 1-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24357332

ABSTRACT

A potent T cell response is an important component of durable anti-tumor immunity. The quality of the T cell response can, in-part, be measured by the avidity of the T cell for its tumor antigen-expressing target. While convention suggests that raising the avidity of the responding T cells may make for a more potent anti-tumor immune response, the threshold for effective tumor immunity remains unclear, as do some of the adverse effects of an inappropriately high avidity response. In this review, we discuss the relationship between T cell avidity and anti-tumor immunity, considering both experimental model systems as well as human clinical trials.

19.
Cancer Res ; 73(2): 595-604, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23204239

ABSTRACT

One obstacle in eliciting potent antitumor immune responses is the induction of tolerance to tumor antigens. TCR(lo) mice bearing a TCR transgene specific for the melanoma antigen tyrosinase-related protein-2 (TRP-2, Dct) harbor T cells that maintain tumor antigen responsiveness but lack the ability to control melanoma outgrowth. We used this model to determine whether higher avidity T cells could control tumor growth without becoming tolerized. As a part of the current study, we developed a second TRP-2-specific TCR transgenic mouse line (TCR(hi)) that bears higher avidity T cells and spontaneously developed autoimmune depigmentation. In contrast to TCR(lo) T cells, which were ignorant of tumor-derived antigen, TCR(hi) T cells initially delayed subcutaneous B16 melanoma tumor growth. However, persistence in the tumor microenvironment resulted in reduced IFN-γ production and CD107a (Lamp1) mobilization, hallmarks of T-cell tolerization. IFN-γ expression by TCR(hi) T cells was critical for upregulation of MHC-I on tumor cells and control of tumor growth. Blockade of PD-1 signals prevented T-cell tolerization and restored tumor immunity. Depletion of tumor-associated dendritic cells (TADC) reduced tolerization of TCR(hi) T cells and enhanced their antitumor activity. In addition, TADCs tolerized TCR(hi) T cells but not TCR(lo) T cells in vitro. Our findings show that T-cell avidity is a critical determinant of not only tumor control but also susceptibility to tolerization in the tumor microenvironment. For this reason, care should be exercised when considering T-cell avidity in designing cancer immunotherapeutics.


Subject(s)
Immune Tolerance , Intramolecular Oxidoreductases/immunology , Melanoma, Experimental/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Antigens, Neoplasm , Dendritic Cells/immunology , Female , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Microenvironment
20.
Oncoimmunology ; 1(2): 252-254, 2012 03 01.
Article in English | MEDLINE | ID: mdl-22720261

ABSTRACT

Recent findings demonstrate that dendritic cells in prostate tumors induce immune tolerance in tumor antigen-specific CD8(+) T cells. We propose that DC tolerogenicity can be regulated by expression of Foxo3; silencing Foxo3 expression enhances anti-tumor immune responses and renders FOXO3 a potential target for immunotherapy.

SELECTION OF CITATIONS
SEARCH DETAIL