Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 23(22): 6833-6845, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28855356

ABSTRACT

Purpose: The transcription factor brachyury has been shown in preclinical studies to be a driver of the epithelial-to-mesenchymal transition (EMT) and resistance to therapy of human tumor cells. This study describes the characterization of a Modified Vaccinia Ankara (MVA) vector-based vaccine expressing the transgenes for brachyury and three human costimulatory molecules (B7.1, ICAM-1, and LFA-3, designated TRICOM) and a phase I study with this vaccine.Experimental Design: Human dendritic cells (DC) were infected with MVA-brachyury-TRICOM to define their ability to activate brachyury-specific T cells. A dose-escalation phase I study (NCT02179515) was conducted in advanced cancer patients (n = 38) to define safety and to identify brachyury-specific T-cell responses.Results: MVA-brachyury-TRICOM-infected human DCs activated CD8+ and CD4+ T cells specific against the self-antigen brachyury in vitro No dose-limiting toxicities were observed due to vaccine in cancer patients at any of the three dose levels. One transient grade 3 adverse event (AE) possibly related to vaccine (diarrhea) resolved without intervention and did not recur with subsequent vaccine. All other AEs related to vaccine were transient and ≤grade 2. Brachyury-specific T-cell responses were observed at all dose levels and in most patients.Conclusions: The MVA-brachyury-TRICOM vaccine directed against a transcription factor known to mediate EMT can be administered safely in patients with advanced cancer and can activate brachyury-specific T cells in vitro and in patients. Further studies of this vaccine in combination therapies are warranted and planned. Clin Cancer Res; 23(22); 6833-45. ©2017 AACR.


Subject(s)
Cancer Vaccines/immunology , Fetal Proteins/immunology , Genetic Vectors , Neoplasms/immunology , Neoplasms/therapy , T-Box Domain Proteins/immunology , Vaccinia virus , Adult , Aged , Aged, 80 and over , B7-1 Antigen/genetics , Biomarkers, Tumor , CD58 Antigens/genetics , Cancer Vaccines/genetics , Dendritic Cells/immunology , Dendritic Cells/metabolism , Fetal Proteins/genetics , Genetic Vectors/genetics , Humans , Intercellular Adhesion Molecule-1/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Middle Aged , Neoplasms/genetics , Neoplasms/mortality , T-Box Domain Proteins/genetics , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transgenes , Treatment Outcome , Vaccinia virus/genetics
2.
Vaccine ; 35(19): 2605-2611, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28389098

ABSTRACT

Human papillomavirus (HPV) is associated with the etiology of cervical carcinoma, head and neck squamous cell carcinoma, and several other cancer types. Vaccines directed against HPV virus-like particles and coat proteins have been extremely successful in the prevention of cervical cancer through the activation of host HPV-specific antibody responses; however, HPV-associated cancers remain a major public health problem. The development of a therapeutic vaccine will require the generation of T-cell responses directed against early HPV proteins (E6/E7) expressed in HPV-infected tumor cells. Clinical studies using various vaccine platforms have demonstrated that both HPV-specific human T cells can be generated and patient benefit can be achieved. However, no HPV therapeutic vaccine has been approved by the Food and Drug Administration to date. One method of enhancing the potential efficacy of a therapeutic vaccine is the generation of agonist epitopes. We report the first description of enhancer cytotoxic T lymphocyte agonist epitopes for HPV E6 and E7. While the in silico algorithm revealed six epitopes with potentially improved binding to human leukocyte antigen-A2 allele (HLA-A2)-Class I, 5/6 demonstrated enhanced binding to HLA-Class I in cell-based assays and only 3/6 had a greater ability to activate HPV-specific T cells which could lyse tumor cells expressing native HPV, compared to their native epitope counterparts. These agonist epitopes have potential for use in a range of HPV therapeutic vaccine platforms and for use in HPV-specific adoptive T- or natural killer-cell platforms.


Subject(s)
Epitope Mapping , Epitopes, T-Lymphocyte/immunology , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/immunology , Repressor Proteins/immunology , Female , Histocompatibility Antigens Class I/metabolism , Humans , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Protein Binding , Repressor Proteins/metabolism
3.
Oncotarget ; 7(27): 42031-42044, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27248176

ABSTRACT

A signaling pathway that is frequently deregulated in human carcinomas and has been explored as a therapeutic target involves the activation of the epidermal growth factor receptor (EGFR). Inhibition of EGFR via the small molecule inhibitors erlotinib and gefitinib commonly results in tumor resistance, even in patients with EGFR-mutant tumors that initially show substantial clinical responses. This study was designed to broaden our understanding of the molecular mechanisms of acquired resistance to erlotinib in lung cancer cells bearing wild type or mutated EGFR. We report here that generation of erlotinib-resistant lung cancer cells in vitro resulted in a phenotypic alteration reminiscent of an epithelial-mesenchymal transition (EMT) concomitant with a robust upregulation of the IL-8/IL-8R axis. Our results also demonstrate that upregulation of p38 MAPK signaling is responsible for the enhanced IL-8 secretion in the erlotinib-resistant tumor cells. Blockade of IL-8 signaling effectively reduced mesenchymal features of the resistant cells and also markedly enhanced their susceptibility to erlotinib. These results provide a rationale for the development of new therapeutic approaches involving blockade of IL-8 signaling for the management of acquired resistance to EGFR inhibition in patients with lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Drug Resistance, Neoplasm , Erlotinib Hydrochloride/therapeutic use , Interleukin-8/metabolism , Lung Neoplasms/metabolism , A549 Cells , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Epithelial-Mesenchymal Transition , ErbB Receptors/metabolism , Female , Gefitinib , Humans , Lung Neoplasms/drug therapy , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Phenotype , Quinazolines/therapeutic use , Signal Transduction , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Oncotarget ; 7(25): 37762-37772, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27192116

ABSTRACT

Epacadostat is a novel inhibitor of indoleamine-2,3-dioxygenase-1 (IDO1) that suppresses systemic tryptophan catabolism and is currently being evaluated in ongoing clinical trials. We investigated the effects of epacadostat on (a) human dendritic cells (DCs) with respect to maturation and ability to activate human tumor antigen-specific cytotoxic T-cell (CTL) lines, and subsequent T-cell lysis of tumor cells, (b) human regulatory T cells (Tregs), and (c) human peripheral blood mononuclear cells (PBMCs) in vitro. Simultaneous treatment with epacadostat and IFN-γ plus lipopolysaccharide (LPS) did not change the phenotype of matured human DCs, and as expected decreased the tryptophan breakdown and kynurenine production. Peptide-specific T-cell lines stimulated with DCs pulsed with peptide produced significantly more IFN-γ, TNFα, GM-CSF and IL-8 if the DCs were treated with epacadostat. These T cells also displayed higher levels of tumor cell lysis on a per cell basis. Epacadostat also significantly decreased Treg proliferation induced by IDO production from IFN-γ plus LPS matured human DCs, although the Treg phenotype did not change. Multicolor flow cytometry was performed on human PBMCs treated with epacadostat; analysis of 123 discrete immune cell subsets revealed no changes in major immune cell types, an increase in activated CD83+ conventional DCs, and a decrease in immature activated Tim3+ NK cells. These studies show for the first time several effects of epacadostat on human DCs, and subsequent effects on CTL and Tregs, and provide a rationale as to how epacadostat could potentially increase the efficacy of immunotherapeutics, including cancer vaccines.


Subject(s)
Dendritic Cells/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Oximes/pharmacology , Sulfonamides/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Adult , Aged , Cancer Vaccines/pharmacology , Dendritic Cells/drug effects , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Healthy Volunteers , Humans , Interferon-gamma/metabolism , Kynurenine/chemistry , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/chemistry , Lymphocyte Activation/immunology , Male , Middle Aged , Peptides/chemistry , Phenotype , T-Lymphocytes, Cytotoxic/drug effects , Tryptophan/chemistry , Young Adult
5.
Oncotarget ; 6(7): 4853-62, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25605015

ABSTRACT

The embryonic transcription factor brachyury is overexpressed in a variety of human tumors, including lung, breast, colon and prostate carcinomas, chordomas and hemangioblastomas. In human carcinoma cells, overexpression of brachyury associates with the occurrence of the phenomenon of epithelial-mesenchymal transition (EMT), acquisition of metastatic propensity and resistance to a variety of anti-cancer therapeutics. Brachyury is preferentially expressed in human tumors vs. normal adult tissues, and high levels of this molecule associate with poor prognosis in patients with lung, colon and prostate carcinomas, and in breast cancer patients treated with adjuvant tamoxifen. Brachyury is immunogenic in humans and vaccines against this novel oncotarget are currently undergoing clinical investigation. While our group and others have employed various anti-brachyury antibodies to interrogate the above findings, we report here on the development and thorough characterization of a novel rabbit monoclonal antibody (MAb 54-1) that reacts with distinct high affinity and specificity with human brachyury. MAb 54-1 was successfully used in ELISA, western blot, immunofluorescence and immunohistochemistry assays to evaluate expression of brachyury in various human tumor cell lines and tissues. We propose the use of this antibody to assist in research studies of EMT and in prognostic studies for a range of human tumors.


Subject(s)
Antibodies, Monoclonal/chemistry , Fetal Proteins/biosynthesis , Lung Neoplasms/metabolism , T-Box Domain Proteins/biosynthesis , Animals , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/analysis , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/immunology , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Fetal Proteins/analysis , Fetal Proteins/immunology , Humans , Immunohistochemistry , Lung Neoplasms/pathology , Prognosis , Rabbits , T-Box Domain Proteins/analysis , T-Box Domain Proteins/immunology
6.
Exp Cell Res ; 331(1): 1-10, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25499972

ABSTRACT

We have previously demonstrated that the anti-apoptotic protein BAD is expressed in normal human breast tissue and shown that BAD inhibits expression of cyclin D1 to delay cell-cycle progression in breast cancer cells. Herein, expression of proteins in breast tissues was studied by immunohistochemistry and results were analyzed statistically to obtain semi-quantitative data. Biochemical and functional changes in BAD-overexpressing MCF7 breast cancer cells were evaluated using PCR, reporter assays, western blotting, ELISA and extracellular matrix invasion assays. Compared to normal tissues, Grade II breast cancers expressed low total/phosphorylated forms of BAD in both cytoplasmic and nuclear compartments. BAD overexpression decreased the expression of ß-catenin, Sp1, and phosphorylation of STATs. BAD inhibited Ras/MEK/ERK and JNK signaling pathways, without affecting the p38 signaling pathway. Expression of the metastasis-related proteins, MMP10, VEGF, SNAIL, CXCR4, E-cadherin and TlMP2 was regulated by BAD with concomitant inhibition of extracellular matrix invasion. Inhibition of BAD by siRNA increased invasion and Akt/p-Akt levels. Clinical data and the results herein suggest that in addition to the effect on apoptosis, BAD conveys anti-metastatic effects and is a valuable prognostic marker in breast cancer.


Subject(s)
Cell Movement , Down-Regulation , bcl-Associated Death Protein/metabolism , Blotting, Western , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Epithelial-Mesenchymal Transition , Female , Flow Cytometry , Humans , Immunoenzyme Techniques , MCF-7 Cells , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Tumor Cells, Cultured , bcl-Associated Death Protein/antagonists & inhibitors , beta Catenin/genetics , beta Catenin/metabolism
7.
J Natl Cancer Inst ; 106(5)2014 May 09.
Article in English | MEDLINE | ID: mdl-24815864

ABSTRACT

BACKGROUND: The epithelial-mesenchymal transition (EMT) has been implicated as an important process in tumor cell invasion, metastasis, and drug resistance. The transcription factor brachyury has recently been described as a driver of EMT of human carcinoma cells. METHODS: Brachyury mRNA and protein expression was analyzed in human breast carcinomas and benign tissues. The role of brachyury in breast tumor prognosis and drug resistance and the ability of brachyury-specific T cells to lyse human breast carcinoma cells were also evaluated. Kaplan-Meier analyses were used to evaluate the association between brachyury expression and survival. All statistical tests were two-sided. RESULTS: The level of brachyury expression in breast cancer cells was positively associated with their ability to invade the extracellular matrix, efficiently form mammospheres in vitro, and resist the cytotoxic effect of docetaxel. A comparison of survival among breast cancer patients treated with tamoxifen in the adjuvant setting who had tumors with high vs low brachyury mRNA expression demonstrated that high expression of brachyury is associated as an independent variable with higher risk of recurrence (hazard ratio [HR] = 7.5; 95% confidence interval [CI] = 2.4 to 23.5; P = 5.14×10(-4)) and distant metastasis (HR = 15.2; 95% CI = 3.5 to 66.3; P = 3.01×10(-4)). We also demonstrated that brachyury-specific T cells can lyse human breast carcinoma cells. CONCLUSIONS: The studies reported here provide the rationale for the use of a vaccine targeting brachyury for the therapy of human breast cancer, either as a monotherapy or in combination therapies.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Fetal Proteins/biosynthesis , T-Box Domain Proteins/biosynthesis , Adult , Aged , Aged, 80 and over , Breast Neoplasms/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Fetal Proteins/genetics , Formaldehyde , Humans , Immunohistochemistry , Middle Aged , Neoplasm Invasiveness , Paraffin Embedding , Prognosis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , T-Box Domain Proteins/genetics , Tissue Fixation
8.
Cancer Res ; 74(9): 2510-9, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24626094

ABSTRACT

Aberrant expression of the T-box transcription factor brachyury in human carcinomas drives the phenomenon of epithelial-mesenchymal transition (EMT), a phenotypic modulation that facilitates tumor dissemination and resistance to conventional therapies, including chemotherapy and radiotherapy. By generating isogenic cancer cell lines with various levels of brachyury expression, we demonstrate that high levels of brachyury also significantly reduce the susceptibility of cancer cells to lysis by both antigen-specific T cells and natural killer cells. Our results indicated that resistance of brachyury-high tumor cells to immune-mediated attack was due to inefficient caspase-dependent apoptosis, manifested as inefficient nuclear lamin degradation in the presence of activated effector caspases. We correlated this phenomenon with loss of cell-cycle-dependent kinase 1 (CDK1), which mediates lamin phosphorylation. In support of a causal connection, pretreatment of tumor cells with a specific inhibitor of WEE1, a negative regulator kinase of CDK1, could counter the defective apoptosis of tumor cells expressing high levels of brachyury. Thus, our findings suggested that reconstituting CDK1 activity to threshold levels may be sufficient to restore immunosurveillance of mesenchymal-like cancer cells that have escaped previous immune detection or eradication.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Proteins/metabolism , Cytotoxicity, Immunologic/drug effects , Epithelial-Mesenchymal Transition/drug effects , Nuclear Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Animals , Apoptosis , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Fetal Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Nuclear Proteins/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidinones , T-Box Domain Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/physiology , Tumor Escape
9.
Oncoimmunology ; 3(1): e27220, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24575384

ABSTRACT

Targeting the epithelial-to-mesenchymal transition (EMT) is emerging as a novel intervention against tumor progression and metastatic dissemination, as well as the resistance to chemo- and radiotherapy displayed by multiple carcinomas. We have recently developed an immunotherapeutic approach to target a major driver of EMT, the T-box transcription factor T (also known as brachyury). This therapeutic paradigm is currently being tested in patients with advanced carcinomas in the context of a Phase I clinical trial.

10.
Oncotarget ; 4(10): 1777-90, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24125763

ABSTRACT

The embryonic T-box transcription factor brachyury is aberrantly expressed in a range of human tumors. Previous studies have demonstrated that brachyury is a driver of the epithelial-mesenchymal transition (EMT), a process associated with cancer progression. Brachyury expression in human tumor cells enhances tumor invasiveness in vitro and metastasis in vivo, and induces resistance to various conventional therapeutics including chemotherapy and radiation. These characteristics, and the selective expression of brachyury for a range of human tumor types vs. normal adult tissues, make brachyury an attractive tumor target. Due to its intracellular localization and the "undruggable" character of transcription factors, available options to target brachyury are currently limited. Here we report on the development and characterization of an immunological platform for the efficient targeting of brachyury-positive tumors consisting of a heat-killed, recombinant Saccharomyces cerevisiae (yeast)-brachyury vector-based vaccine (designated as GI-6301) that expresses the full-length human brachyury protein. We demonstrate that human dendritic cells treated with recombinant yeast-brachyury can activate and expand brachyury-specific CD4+ and CD8+ T cells in vitro that, in turn, can effectively lyse human tumor cells expressing the brachyury protein. Vaccination of mice with recombinant yeast-brachyury is also shown here to elicit brachyury-specific CD4+ and CD8+ T-cell responses, and to induce anti-tumor immunity in the absence of toxicity. Based on these results, a Phase I clinical trial of GI-6301 is currently ongoing in patients with advanced tumors; to our knowledge, this is the first vaccine platform aimed at targeting a driver of tumor EMT that has successfully reached the clinical stage.


Subject(s)
Cancer Vaccines/pharmacology , Epithelial-Mesenchymal Transition/immunology , Fetal Proteins/immunology , T-Box Domain Proteins/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/immunology , Wound Healing/immunology
11.
Mol Cancer Ther ; 12(9): 1805-15, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23783250

ABSTRACT

The epithelial-mesenchymal transition (EMT) is a process associated with the metastasis of solid tumors as well as with the acquisition of resistance to standard anticancer modalities. A major initiator of EMT in carcinoma cells is TGF-ß, which has been shown to induce the expression of several transcription factors ultimately responsible for initiating and maintaining the EMT program. We have previously identified Brachyury, a T-box transcription factor, as an inducer of mesenchymal features in human carcinoma cells. In this study, a potential link between Brachyury and TGF-ß signaling has been investigated. The results show for the first time that Brachyury expression is enhanced during TGF-ß1-induced EMT in various human cancer cell lines, and that a positive feedback loop is established between Brachyury and TGF-ß1 in mesenchymal-like tumor cells. In this context, Brachyury overexpression is shown to promote upregulation of TGF-ß1 at the mRNA and protein levels, an effect mediated by activation of the TGF-ß1 promoter in the presence of high levels of Brachyury. Furthermore, inhibition of TGF-ß1 signaling by a small-molecule inhibitor of TGF-ß receptor type I decreases Brachyury expression, induces a mesenchymal-to-epithelial transition, and renders cancer cells more susceptible to chemotherapy. This study thus has implications for the future development of clinical trials using TGF-ß inhibitors in combination with other anticancer agents.


Subject(s)
Epithelial-Mesenchymal Transition , Fetal Proteins/metabolism , T-Box Domain Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Antineoplastic Combined Chemotherapy Protocols , Cell Line, Tumor , Cisplatin/therapeutic use , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Feedback, Physiological , Fetal Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Neoplasms/metabolism , Phenotype , Pteridines/pharmacology , Pteridines/therapeutic use , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Signal Transduction/drug effects , T-Box Domain Proteins/genetics , Transforming Growth Factor beta1/antagonists & inhibitors , Vinblastine/analogs & derivatives , Vinblastine/therapeutic use , Vinorelbine
12.
Future Oncol ; 8(6): 713-22, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22764769

ABSTRACT

The phenomenon of epithelial-mesenchymal transition (EMT) has gained attention in the field of cancer biology for its potential contribution to the progression of carcinomas. Tumor EMT is a phenotypic switch that promotes the acquisition of a fibroblastoid-like morphology by epithelial tumor cells, resulting in enhanced tumor cell motility and invasiveness, increased metastatic propensity and resistance to chemotherapy, radiation and certain small-molecule-targeted therapies. Tumor cells undergoing EMT are also known to increase the secretion of specific factors, including cytokines, chemokines and growth factors, which could play an important role in tumor progression. This review summarizes the current knowledge on the secretory properties of epithelial tumor cells that have undergone an EMT, with an emphasis on the potential role of the IL-8-IL-8 receptor axis on the induction and/or maintenance of tumor EMT and its ability to remodel the tumor microenvironment.


Subject(s)
Epithelial-Mesenchymal Transition , Interleukin-8/metabolism , Tumor Microenvironment , Animals , Autocrine Communication , Humans , Receptors, Interleukin-8/metabolism
13.
Clin Cancer Res ; 18(14): 3868-79, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22611028

ABSTRACT

PURPOSE: The epithelial-mesenchymal transition (EMT) is emerging as a critical factor for the progression and metastasis of carcinomas, as well as drug resistance. The T-box transcription factor Brachyury has been recently characterized as a driver of EMT in human carcinoma cells. The purpose of this study was to characterize Brachyury as a potential target for lung cancer therapy. EXPERIMENTAL DESIGN: The expression of Brachyury was evaluated by PCR and by immunohistochemistry in human lung tumors and adult normal tissues. Brachyury gene copy number and promoter methylation status were analyzed in tumor tissues with various levels of Brachyury expression. Lung carcinoma cells' susceptibility to T-cell lysis and EGF receptor (EGFR) kinase inhibition were also evaluated relative to the levels of Brachyury. RESULTS: Our results showed Brachyury protein expression in 41% of primary lung carcinomas, including 48% of adenocarcinomas and 25% of squamous cell carcinomas. With the exception of normal testis and some thyroid tissues, the majority of normal tissues evaluated in this study were negative for the expression of Brachyury protein. Brachyury-specific T cells could lyse Brachyury-positive tumors and the level of Brachyury corresponded to resistance of tumor cells to EGFR kinase inhibition. CONCLUSION: We hypothesize that the elimination of Brachyury-positive tumor cells may be able to prevent and/or diminish tumor dissemination and the establishment of metastases. The ability of Brachyury-specific T-cell lines to lyse Brachyury-positive tumor cells, in vitro, supports the development of Brachyury-based immunotherapeutic approaches for the treatment of lung cancer.


Subject(s)
Epithelial-Mesenchymal Transition , Fetal Proteins , Lung Neoplasms , Neoplasm Invasiveness/genetics , T-Box Domain Proteins , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Disease Progression , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Fetal Proteins/genetics , Fetal Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplasm Metastasis , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
14.
Semin Oncol ; 39(3): 358-66, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22595058

ABSTRACT

The epithelial-mesenchymal transition (EMT) is thought to be a critical step along the metastasis of carcinomas. In addition to gaining motility and invasiveness, tumor cells that undergo EMT also acquire increased resistance to many traditional cancer treatment modalities, including chemotherapy and radiation. As such, EMT has become an attractive, potentially targetable process for therapeutic interventions against tumor metastasis. The process of EMT is driven by a group of transcription factors designated as EMT transcription factors, such as Snail, Slug, Twist, and the recently identified T-box family member, Brachyury. In an attempt to determine which of these drivers of EMT is more amenable to targeted therapies and, in particular, T-cell-mediated immunotherapeutic approaches, we have examined their relative expression levels in a range of human and murine normal tissues, cancer cell lines, and human tumor biopsies. Our results demonstrated that Brachyury is a molecule with a highly restricted human tumor expression pattern. We also demonstrated that Brachyury is immunogenic and that Brachyury-specific CD8(+) T cells expanded in vitro are able to lyse Brachyury-positive tumor cells. We thus propose Brachyury as an attractive target for vaccination strategies designed to specifically target tumor cells undergoing EMT.


Subject(s)
Cancer Vaccines/immunology , Carcinoma/immunology , Epithelial-Mesenchymal Transition/immunology , Fetal Proteins/immunology , T-Box Domain Proteins/immunology , Animals , Cancer Vaccines/genetics , Cancer Vaccines/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Fetal Proteins/genetics , Fetal Proteins/metabolism , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/immunology , Phenotype , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
15.
Cancer Res ; 71(15): 5296-306, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21653678

ABSTRACT

The switch of tumor cells from an epithelial to a mesenchymal-like phenotype [designated as epithelial-to-mesenchymal transition (EMT)] is known to induce tumor cell motility and invasiveness, therefore promoting metastasis of solid carcinomas. Although multiple studies have focused on elucidating the signaling events that initiate this phenotypic switch, there has been so far no characterization of the pattern of soluble mediators released by tumor cells undergoing EMT, and the potential impact that this phenotypic switch could have on the remodeling of the tumor microenvironment. Here we show that induction of EMT in human carcinoma cells via overexpression of the transcription factor Brachyury is associated with enhanced secretion of multiple cytokines, chemokines, and angiogenic factors and, in particular, with the induction of the IL-8/IL-8R axis. Our results also indicate the essential role of interleukin 8 (IL-8) signaling for the acquisition and/or maintenance of the mesenchymal and invasive features of Brachyury-overexpressing tumor cells and show that IL-8 secreted by tumor cells undergoing EMT could potentiate tumor progression by inducing adjacent epithelial tumor cells into EMT. Altogether, our results emphasize the potential role of EMT in the modulation of the tumor microenvironment via secretion of multiple soluble mediators and suggest that IL-8 signaling blockade may provide a means of targeting mesenchymal-like, invasive tumor cells.


Subject(s)
Carcinoma/pathology , Epithelial-Mesenchymal Transition/physiology , Interleukin-8/physiology , Neoplasm Proteins/physiology , Tumor Microenvironment/physiology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Bystander Effect , Carcinoma/metabolism , Cell Line, Tumor/metabolism , Cell Line, Tumor/pathology , Cell Movement , Chemokines/metabolism , Culture Media, Conditioned/pharmacology , Culture Media, Serum-Free , Cytokines/metabolism , Fetal Proteins/antagonists & inhibitors , Fetal Proteins/biosynthesis , Fetal Proteins/genetics , Fetal Proteins/physiology , Fibronectins/biosynthesis , Fibronectins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Invasiveness , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , RNA, Small Interfering/pharmacology , Receptors, Interleukin-8/biosynthesis , Receptors, Interleukin-8/genetics , Recombinant Fusion Proteins/antagonists & inhibitors , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/genetics , T-Box Domain Proteins/physiology
16.
Exp Biol Med (Maywood) ; 236(5): 537-45, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21427233

ABSTRACT

The switch of carcinoma cells from an epithelial to a mesenchymal-like phenotype, via a process designated 'epithelial-to-mesenchymal transition (EMT),' has been recognized as a relevant step in the metastasis of solid tumors. Additionally, this phenotypic switch of carcinoma cells has been associated with the acquisition of tumor resistance mechanisms that reduce the antitumor effects of radiation, chemotherapy and some small-molecule-targeted therapies. As multiple signaling pathways and transcriptional regulators that play a role in this phenotypic switch are being identified, novel strategies can be designed to specifically target tumor cells with this metastatic and resistant phenotype. In particular, this review focuses on the potential use of cancer vaccine strategies to target tumor cells that exhibit a mesenchymal-like phenotype, with an emphasis on the characterization of a novel tumor antigen, Brachyury, which we have identified as a critical regulator of EMT in human cancer cells.


Subject(s)
Carcinoma/pathology , Mesoderm/pathology , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Humans , Phenotype , Transcription Factors/metabolism
17.
J Clin Invest ; 120(2): 533-44, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20071775

ABSTRACT

Metastatic disease is responsible for the majority of human cancer deaths. Understanding the molecular mechanisms of metastasis is a major step in designing effective cancer therapeutics. Here we show that the T-box transcription factor Brachyury induces in tumor cells epithelial-mesenchymal transition (EMT), an important step in the progression of primary tumors toward metastasis. Overexpression of Brachyury in human carcinoma cells induced changes characteristic of EMT, including upregulation of mesenchymal markers, downregulation of epithelial markers, and an increase in cell migration and invasion. Brachyury overexpression also repressed E-cadherin transcription, an effect partially mediated by Slug. Conversely, inhibition of Brachyury resulted in downregulation of mesenchymal markers and loss of cell migration and invasion and diminished the ability of human tumor cells to form lung metastases in a xenograft model. Furthermore, we found Brachyury to be overexpressed in various human tumor tissues and tumor cell lines compared with normal tissues. We also determined that the percentage of human lung tumor tissues positive for Brachyury expression increased with the stage of the tumor, indicating a potential association between Brachyury and tumor progression. The selective expression of Brachyury in tumor cells and its role in EMT and cancer progression suggest that Brachyury may be an attractive target for antitumor therapies.


Subject(s)
Epithelial Cells/pathology , Fetal Proteins/physiology , Mesoderm/pathology , T-Box Domain Proteins/physiology , Cadherins/genetics , Cell Division , Cell Movement , DNA, Complementary/genetics , Disease Progression , Epithelial Cells/physiology , Fetal Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Mesoderm/physiology , Neoplasm Invasiveness , Neoplasm Metastasis/pathology , Neoplasms/genetics , Neoplasms/pathology , Phenotype , Polymerase Chain Reaction , T-Box Domain Proteins/genetics
18.
Clin Cancer Res ; 13(8): 2471-8, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17438107

ABSTRACT

PURPOSE: Identification of tumor antigens is essential in advancing immune-based therapeutic interventions in cancer. Particularly attractive targets are those molecules that are selectively expressed by malignant cells and that are also essential for tumor progression. EXPERIMENTAL DESIGN AND RESULTS: We have used a computer-based differential display analysis tool for mining of expressed sequence tag clusters in the human Unigene database and identified Brachyury as a novel tumor antigen. Brachyury, a member of the T-box transcription factor family, is a key player in mesoderm specification during embryonic development. Moreover, transcription factors that control mesoderm have been implicated in the epithelial-mesenchymal transition (EMT), which has been postulated to be a key step during tumor progression to metastasis. Reverse transcription-PCR analysis validated the in silico predictions and showed Brachyury expression in tumors of the small intestine, stomach, kidney, bladder, uterus, ovary, and testis, as well as in cell lines derived from lung, colon, and prostate carcinomas, but not in the vast majority of the normal tissues tested. An HLA-A0201 epitope of human Brachyury was identified that was able to expand T lymphocytes from blood of cancer patients and normal donors with the ability to lyse Brachyury-expressing tumor cells. CONCLUSIONS: To our knowledge, this is the first demonstration that (a) a T-box transcription factor and (b) a molecule implicated in mesodermal development, i.e., EMT, can be a potential target for human T-cell-mediated cancer immunotherapy.


Subject(s)
Fetal Proteins/genetics , Fetal Proteins/immunology , Immunotherapy/methods , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , Carcinoma , Cell Line, Tumor , DNA Primers , DNA, Complementary , Epithelial Cells/physiology , Expressed Sequence Tags , HLA-A2 Antigen/metabolism , Humans , Mesoderm/physiology , Peptide Fragments/chemistry , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction
19.
J Biol Chem ; 278(42): 41355-66, 2003 Oct 17.
Article in English | MEDLINE | ID: mdl-12904306

ABSTRACT

The cyclin-dependent kinase (CDK) inhibitor p27Kip1 plays a key role in growth and development of the mammary epithelium and in breast cancer. p27Kip1 levels are regulated through ubiquitin/proteasome-mediated proteolysis, promoted by CDK2 and the F box protein Skp2 at the G1/S transition, and independent of Skp2 in mid-G1. We investigated the respective roles of Skp2 and subcellular localization of p27Kip1 in down-regulation of p27Kip1 induced in MCF-7 cells by estrogens. 17beta-Estradiol treatment increased Skp2 expression in MCF-7 cells; however, this increase was prevented by G1 blockade mediated by p16Ink4a or the CDK inhibitor roscovitine, whereas down-regulation of p27Kip1 was maintained. Exogenous Skp2 prevented growth arrest of MCF-7 cells by antiestrogen, coinciding with decreased p27Kip1 expression. Under conditions of G1 blockade, p27Kip1 was stabilized by inhibition of CRM1-dependent nuclear export with leptomycin B or by mutation of p27Kip1 (Ser10 --> Ala; S10A) interfering with CRM1/p27Kip1 interaction. Antisense Skp2 oligonucleotides and a dominant-interfering Cul-1(1-452) mutant prevented down-regulation of p27Kip1S10A, whereas Skp2 overexpression elicited its destruction in mitogen-deprived cells. Active mediators of the extracellular signal-regulated kinase (ERK) pathway including Raf-1caax induced cytoplasmic localization of p27Kip1 in antiestrogen-treated cells and prevented accumulation of p27Kip1 in these cells independent of Skp2 expression and coinciding with ERK activation. Genetic or chemical blockade of the ERK pathway prevented down-regulation and cytoplasmic localization of p27Kip1 in response to estrogen. Our studies indicate that estrogens elicit down-regulation of p27Kip1 in MCF-7 cells through Skp2-dependent and -independent mechanisms that depend upon subcellular localization of p27Kip1 and require the participation of mediators of the Ras/Raf-1/ERK signaling pathway.


Subject(s)
Active Transport, Cell Nucleus , Breast Neoplasms/metabolism , Cell Cycle Proteins/biosynthesis , Cell Nucleus/metabolism , Estrogens/metabolism , Mitogen-Activated Protein Kinases/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Tumor Suppressor Proteins/biosynthesis , Blotting, Western , Cell Division , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27 , Cytoplasm/metabolism , Down-Regulation , Enzyme Activation , Fatty Acids, Unsaturated/pharmacology , Flow Cytometry , G1 Phase , Gene Expression Regulation, Enzymologic , Genetic Vectors , Humans , MAP Kinase Signaling System , Models, Biological , Mutation , Oligonucleotides, Antisense/pharmacology , Phosphorylation , Signal Transduction , Time Factors , Transfection
20.
Reprod Biol Endocrinol ; 1: 46, 2003 Jun 03.
Article in English | MEDLINE | ID: mdl-12816543

ABSTRACT

Distinct luteinizing hormone receptor (LHR) protein variants exist due to the posttranslational modifications. Besides ovaries, LHR immunoreactivity (LHRI) was also found in other tissues, such as the brain, fallopian tube, endometrium, trophoblast and resident tissue macrophages. The 3B5 mouse monoclonal antibody was raised against purified rat LHR. In rat, porcine and human ovaries, the 3B5 identified six distinct LHR bands migrating at approximately 92, 80, 68, 59, 52 and 48 kDa. Characteristic LHRI was detected in rat, human and porcine corpora lutea. During cellular differentiation, subcellular LHR distribution changed from none to granular cytoplasmic, perinuclear, surface, nuclear and no staining. There were also differences in vascular LHR expression--lack of LHRI in ovarian vessels and strong staining of vessels in other tissues investigated. In normal human term placentae, villous LHRI was associated with blood sinusoids and cytotrophoblast cells, and rarely detected in trophoblastic syncytium. In all abnormal placentae, the LHRI of sinusoids was absent, and syncytium showed either enhanced (immature placental phenotypes) or no LHRI (aged placental phenotype). LHRI in human brain was identified in microglial cells (CD68+ resident macrophages). Protein extracts from human vaginal wall and levator ani muscle and fascia showed strong approximately 92 and 68 kDa species, and LHRI was detected in smooth muscle cells, fibroblasts, resident macrophages and nuclei of skeletal muscle fibers. Our observations indicate that, in contrast to the theory on the role of vascular hormone receptors in preferential pick up of circulating hormones, there is no need to enhance selective pick up rather only prevent LH/CG transport to inappropriate sites. Abnormal placental LHR expression may play a role in the development of abnormal pregnancy. Expression of LHR in the pelvic floor compartments suggests that high LH levels in postmenopausal women may contribute to the pelvic floor relaxation and increased incidence of pelvic floor disorders. Since chorionic gonadotropin increases secretion of a variety of cytokines by monocytes, and induces their inflammatory reaction and phagocytic activity, high LH levels in aging individuals may also activate microglia (mononuclear phagocyte system in the central nervous system) and contribute to the development of Alzheimer's disease and other inflammation-mediated neurodegenerative diseases.


Subject(s)
Brain Chemistry , Placenta/chemistry , Rats/physiology , Receptors, LH/physiology , Swine/physiology , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Animals , Blood Vessels/chemistry , Chorionic Gonadotropin/physiology , Corpus Luteum/chemistry , Female , Genitalia, Female/blood supply , Genitalia, Female/chemistry , Humans , Immunoenzyme Techniques , Luteinizing Hormone/physiology , Microglia/chemistry , Molecular Weight , Muscle Proteins/analysis , Muscle Proteins/physiology , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/physiology , Organ Specificity , Ovary/blood supply , Ovary/chemistry , Pelvic Floor , Placenta/blood supply , Placenta Diseases/etiology , Placenta Diseases/metabolism , Pregnancy , Pregnancy Complications/etiology , Pregnancy Complications/metabolism , Protein Isoforms/chemistry , Protein Isoforms/physiology , Protein Processing, Post-Translational , Receptors, LH/analysis , Receptors, LH/chemistry , Subcellular Fractions/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...