Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 488
Filter
1.
ESMO Open ; 7(6): 100636, 2022 12.
Article in English | MEDLINE | ID: mdl-36423363

ABSTRACT

BACKGROUND: Immune checkpoint inhibition is an established treatment in programmed death-ligand 1 (PD-L1)-positive metastatic triple-negative (TN) breast cancer (BC). However, the immune landscape of breast cancer brain metastasis (BCBM) remains poorly defined. MATERIALS AND METHODS: The tumour-infiltrating lymphocytes (TILs) and the messenger RNA (mRNA) levels of 770 immune-related genes (NanoString™, nCounter™ Immuno-oncology IO360) were assessed in primary BCs and BCBMs. The prognostic role of ARG2 transcripts and protein expression in primary BCs and its association with outcome was determined. RESULTS: There was a significant reduction of TILs in the BCBMs in comparison to primary BCs. 11.5% of BCs presented a high immune infiltrate (hot), 46.2% were altered (immunosuppressed/excluded) and 34.6% were cold (no/low immune infiltrate). 3.8% of BCBMs were hot, 23.1% altered and 73.1% cold. One hundred and twelve immune-related genes including PD-L1 and CTLA4 were decreased in BCBM compared to the primary BCs (false discovery rate <0.01, log2 fold-change >1.5). These genes are involved in matrix remodelling and metastasis, cytokine-chemokine signalling, lymphoid compartment, antigen presentation and immune cell adhesion and migration. Immuno-modulators such as PD-L1 (CD274), CTLA4, TIGIT and CD276 (B7H3) were decreased in BCBMs. However, PD-L1 and CTLA4 expression was significantly higher in TN BCBMs (P = 0.01), with CTLA4 expression also high in human epidermal growth factor receptor 2-positive (P < 0.01) compared to estrogen receptor-positive BCBMs. ARG2 was one of four genes up-regulated in BCBMs. High ARG2 mRNA expression in primary BCs was associated with worse distant metastasis-free survival (P = 0.038), while ARG2 protein expression was associated with worse breast-brain metastasis-free (P = 0.027) and overall survival (P = 0.019). High transcript levels of ARG2 correlated to low levels of cytotoxic and T cells in both BC and BCBM (P < 0.01). CONCLUSION: This study highlights the immunological differences between primary BCs and BCBMs and the potential importance of ARG2 expression in T-cell depletion and clinical outcome.


Subject(s)
Arginase , Brain Neoplasms , Breast Neoplasms , T-Lymphocytes , Tumor Microenvironment , Female , Humans , B7 Antigens/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CTLA-4 Antigen/genetics , Arginase/genetics , Arginase/metabolism , Brain Neoplasms/secondary
3.
Oncogene ; 34(29): 3871-80, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25284587

ABSTRACT

Breast cancer resistance to endocrine therapies such as tamoxifen and aromatase inhibitors is a significant clinical problem. Steroid receptor coactivator-1 (SRC-1), a coregulatory protein of the oestrogen receptor (ER), has previously been shown to have a significant role in the progression of breast cancer. The chromatin protein high mobility group box 2 (HMGB2) was identified as an SRC-1 interacting protein in the endocrine-resistant setting. We investigated the expression of HMGB2 in a cohort of 1068 breast cancer patients and found an association with increased disease-free survival time in patients treated with endocrine therapy. However, it was also verified that HMGB2 expression could be switched on in endocrine-resistant tumours from breast cancer patients. To explore the function of this poorly characterized protein, we performed HMGB2 ChIPseq and found distinct binding patterns between the two contexts. In the resistant setting, the HMGB2, SRC-1 and ER complex are enriched at promoter regions of target genes, with bioinformatic analysis indicating a switch in binding partners between the sensitive and resistant phenotypes. Integration of binding and gene expression data reveals a concise set of target genes of this complex including the RNA helicase DDX18. Modulation of DDX18 directly affects growth of tamoxifen-resistant cells, suggesting that it may be a critical downstream effector of the HMGB2:ER complex. This study defines HMGB2 interactions with the ER complex at specific target genes in the tamoxifen-resistant setting.


Subject(s)
Breast Neoplasms/metabolism , DEAD-box RNA Helicases/metabolism , HMGB2 Protein/metabolism , Receptors, Estrogen/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/genetics , DEAD-box RNA Helicases/genetics , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HMGB2 Protein/genetics , Humans , Kaplan-Meier Estimate , MCF-7 Cells , Mice, Inbred BALB C , Mice, SCID , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Receptors, Estrogen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Xenograft Model Antitumor Assays
4.
Oncogene ; 34(4): 525-30, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-24469058

ABSTRACT

Treatment with tyrosine kinase inhibitors (TKIs) including trastuzumab has revolutionized the management of HER2-positive breast cancer. Recent evaluation of clinical trial data suggests that a subset of HER2/ER double-positive cancers may not receive significant benefit from the TKI therapy. Here we investigate the cross talk between HER2 and ER in breast cancer and monitor the effect of trastuzumab on the tyrosine kinase effector transcription factor Myc. In HER2-positive breast cancer patients treated with neoadjuvant trastuzumab, steroid receptor-negative status (ER and PR negative) of pre-treatment biopsies predicted pathological complete response (pCR) (n=31 patients, P=0.0486), whereas elevated Myc protein inversely associated with pCR (P=0.0446). Liquid chromatography mass spectrometry identified the corepressor SMRT as a novel Myc-interacting protein. Trastuzumab treatment enhanced Myc-SMRT interactions in HER2-overexpressing breast cancer cells (LCC1) and inhibited expression of the Myc target gene survivin. In HER2-low, ER-positive steroid-dominant cells (MCF7), trastuzumab therapy repressed Myc-SMRT interactions and upregulated survivin expression. Trastuzumab treatment induced ER-CBP interactions, enhanced ER transcriptional activity and upregulated expression of the ER target gene pS2. The absence of pS2 expression in pre-treatment biopsies predicted pCR to neoadjuvant trastuzumab in breast cancer patients (n=25, P=0.0089) and pS2 expression associated with residual cancer burden (P=0.0196). Furthermore, metastatic tissues from patients who had failed trastuzumab therapy were pS2 positive. In HER2-overexpressing cells, trastuzumab treatment can repress Myc transcriptional activity and clinical response is favorable. However, with co-expression of the steroid pathway, this inhibition is lost and response to treatment is often poor.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Receptor Cross-Talk/physiology , Receptor, ErbB-2/physiology , Receptors, Estrogen/physiology , Breast Neoplasms/chemistry , Female , Humans , MCF-7 Cells , Nuclear Receptor Co-Repressor 2/physiology , Proto-Oncogene Proteins c-myc/physiology , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Trastuzumab , Trefoil Factor-1 , Tumor Suppressor Proteins/analysis
5.
Cell Death Dis ; 5: e1213, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24810049

ABSTRACT

CD40-induced signalling through ligation with its natural ligand (CD40L/CD154) is dependent on recruitment of TRAF molecules to the cytoplasmic domain of the receptor. Here, we applied the yeast two-hybrid system to examine whether other proteins can interact with CD40. Fas-Associated Factor 1(FAF1) was isolated from a HeLa cDNA library using the CD40 cytoplasmic tail (216-278 aa) as a bait construct. FAF1 was able to interact with CD40 both in vitro and in vivo. The FAF1 N-terminal domain was sufficient to bind CD40 and required the TRAF6-binding domain within the cytoplasmic tail of CD40 for binding. CD40 ligation induced FAF1 expression in an NFκB-dependent manner. Knockdown of FAF1 prolonged CD40-induced NFκB, whereas overexpression of FAF1 suppressed CD40-induced NFκB activity and this required interaction of FAF1 with the CD40 receptor via its FID domain. Thus, we report a novel role for FAF1in regulating CD40-induced NFκB activation via a negative feedback loop. Loss of FAF1 function in certain human malignancies may contribute to oncogenesis through unchecked NFκB activation, and further understanding of this process may provide a biomarker of NFκB-targeted therapies for such malignancies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , CD40 Antigens/metabolism , NF-kappa B/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins , CD40 Antigens/genetics , Feedback, Physiological , HEK293 Cells , HeLa Cells , Humans , Mutation , Protein Binding , Protein Interaction Domains and Motifs , RNA Interference , TNF Receptor-Associated Factor 6/metabolism , Time Factors , Transfection , Two-Hybrid System Techniques
6.
Oncogene ; 32(22): 2799-804, 2013 May 30.
Article in English | MEDLINE | ID: mdl-22751120

ABSTRACT

Junctional adhesion molecule-A (JAM-A) is a membranous cell-cell adhesion protein involved in tight-junction formation in epithelial and endothelial cells. Its overexpression in breast tumors has recently been linked with increased risk of metastasis. We sought to identify if JAM-A overexpression was associated with specific subtypes of breast cancer as defined by the expression of human epidermal growth factor receptor-2 (HER2), estrogen receptor (ER) and progesterone receptor. To this end, JAM-A immunohistochemistry was performed in two breast cancer tissue microarrays. In parallel, cross-talk between JAM-A, HER2 and ER was examined in several breast cell lines, using complementary genetic and pharmacological approaches. High JAM-A expression correlated significantly with HER2 protein expression, ER negativity, lower patient age, high-grade breast cancers, and aggressive luminal B, HER2 and basal subtypes of breast cancer. JAM-A and HER2 were co-expressed at high levels in vitro in SKBR3, UACC-812, UACC-893 and MCF7-HER2 cells. Knockdown or functional antagonism of HER2 did not alter JAM-A expression in any cell line tested. Interestingly, however, JAM-A knockdown decreased HER2 and ER-α expression, resulting in reduced levels of phospho-(active) AKT without an effect on the extracellular signal-related kinase phosphorylation. The downstream effects of JAM-A knockdown on HER2 and phospho-AKT were partially reversed upon treatment with the proteasomal inhibitor MG132. We conclude that JAM-A is co-expressed with HER2 and associates with aggressive breast cancer phenotypes. Furthermore, we speculate that JAM-A may regulate HER2 proteasomal degradation and activity, potentially offering a promise as a therapeutic target in HER2-positive breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Cell Adhesion Molecules/metabolism , Receptor, ErbB-2/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Adult , Aged , Aged, 80 and over , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Leupeptins/pharmacology , MCF-7 Cells , Middle Aged , Phosphorylation , Proteolysis , Proto-Oncogene Proteins c-akt/biosynthesis , RNA Interference , RNA, Small Interfering , Receptor, ErbB-2/genetics , Receptors, Cell Surface/genetics , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Tissue Array Analysis
7.
Br J Cancer ; 105(1): 118-23, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21654685

ABSTRACT

BACKGROUND: Cutaneous melanoma is an aggressive disease. S100beta is an established biomarker of disease progression; however, the mechanism of its regulation in melanoma is undefined. METHODS: Expression of HOXC11 and SRC-1 was examined by immunohistochemistry and immunofluorescence. Molecular and cellular techniques were used to investigate regulation of S100beta, including, western blot, qPCR, ChIP and migration assays. RESULTS: Expression levels of the transcription factor HOXC11 and its coactivator SRC-1 were significantly elevated in malignant melanoma in comparison with benign nevi (P<0.001 and P=0.017, respectively, n=80), and expression of HOXC11 and SRC-1 in the malignant tissue associated with each other (P<0.001). HOXC11 recruitment to the promoter of S100beta was observed in the primary melanoma cell line SKMel28. S100beta expression was found to be dependant on both HOXC11 and SRC-1. Treatment with the Src/Abl inhibitor, dasatinib, reduced HOXC11-SRC-1 interaction and prevented recruitment of HOXC11 to the S100beta promoter. Dasatinib inhibited both mRNA and protein levels of S100beta and reduced migration of the metastatic cell line MeWo. CONCLUSION: We have defined a signalling mechanism regulating S100beta in melanoma, which can be modulated by dasatinib. Profiling patients for expression of key markers of this network has the potential to increase the efficacy of dasatinib treatment.


Subject(s)
Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Melanoma/drug therapy , Melanoma/metabolism , Nerve Growth Factors/genetics , Nuclear Receptor Coactivator 1/metabolism , Pyrimidines/pharmacology , S100 Proteins/genetics , Thiazoles/pharmacology , Blotting, Western , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Dasatinib , Fluorescent Antibody Technique , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Immunoenzyme Techniques , Immunoprecipitation , Melanoma/genetics , Nerve Growth Factors/metabolism , Nuclear Receptor Coactivator 1/genetics , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Tumor Cells, Cultured
8.
Br J Cancer ; 104(9): 1500-4, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21487403

ABSTRACT

BACKGROUND: To prove a causal link between an epigenetic change and an environmental or behavioural risk factor for a given disease, it is first necessary to show that the onset of exposure precedes the first detection of that epigenetic change in subjects who are still free of disease. METHODS: Towards this end, a cohort of women aged 15-19 years, recruited soon after they first had sexual intercourse, were used to provide sequential observations on the relationship between cigarette smoking and the detection in cervical cytological samples of methylated forms of CDKN2A (p16) using nested methylation-specific polymerase chain reaction. RESULTS: Among women who remained cytologically normal and who tested negative for human papillomavirus DNA in cervical smears during follow-up, those who first started to smoke during follow-up had an increased risk of acquiring CDKN2A methylation compared with never-smokers (odds ratio=3.67; 95% confidence interval 1.09-12.33; P=0.04). CONCLUSION: Smoking initiation is associated with the appearance of methylated forms of CDKN2A.


Subject(s)
Cervix Uteri/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Methylation , Epigenesis, Genetic , Smoking/adverse effects , Adolescent , Alphapapillomavirus , Cohort Studies , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Female , Humans , Logistic Models , Longitudinal Studies , Odds Ratio , Papillomavirus Infections/complications , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Polymerase Chain Reaction/methods , Risk Factors , Smoking/genetics , Surveys and Questionnaires , Tumor Virus Infections/complications , Tumor Virus Infections/pathology , Tumor Virus Infections/virology , Uterine Cervical Neoplasms/etiology , Vaginal Smears , Young Adult , Uterine Cervical Dysplasia/etiology
9.
Gene Ther ; 18(6): 584-93, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21307889

ABSTRACT

This trial tested a dendritic cell (DC) therapeutic cancer vaccine in which antigen is loaded using a novel non-viral transfection method enabling the uptake of plasmid DNA condensed with a cationic peptide. Proof of principle required the demonstration of diverse T lymphocyte responses following vaccination, including multiple reactivities restricted through both major histocompatibility complex (MHC) class I and II. Patients with advanced melanoma were offered four cycles of vaccination with autologous DC expressing melan A and gp100. Disease response was measured using Response Evaluation Criteria in Solid Tumours. Circulating MHC class I- and II-restricted responses were measured against peptide and whole antigen targets using interferon-γ ELIspot and enzyme-linked immunosorbent assay assays, respectively. Responses were analyzed across the trial population and presented descriptively for some individuals. Twenty-five patients received at least one cycle. Vaccination was well tolerated. Three patients had reduction in disease volume. Across the trial population, vaccination resulted in an expansion of effector responses to both antigens, to the human leukocyte antigen A2-restricted modified epitope, melan A ELAGIGILTV, and to a panel of MHC class I- and II-restricted epitopes. Vaccination with mature DC non-virally transfected with DNA encoding antigen had biological effect causing tumour regression and inducing diverse T lymphocyte responses.


Subject(s)
Dendritic Cells/immunology , MART-1 Antigen/genetics , Melanoma/therapy , Vaccines, DNA/therapeutic use , gp100 Melanoma Antigen/genetics , Adult , Aged , Cancer Vaccines/therapeutic use , Female , Humans , Male , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , T-Lymphocytes/immunology , Transfection
10.
Clin Microbiol Infect ; 15(11): 982-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19874382

ABSTRACT

Subsequent to its discovery over 45 years ago, Epstein-Barr virus (EBV) has been associated with numerous human carcinomas. Approximately 95% of the world's population sustain an asymptomatic life-long EBV infection. EBV persists in the memory B cell pool of normal healthy individuals and any disruption of this interaction results in virus-associated B cell tumours. The association of EBV with epithelial cell tumours, specifically nasopharyngeal carcinoma and EBV-positive gastric carcinoma, is less clear and is currently considered to be a consequence of the aberrant establishment of virus latency in epithelial cells displaying pre-malignant genetic changes. Although the precise role of EBV in the carcinogenic process is currently poorly understood, the presence of the virus in all tumour cells provides opportunities for the development of novel therapeutic and diagnostic approaches. The study of EBV and its role in carcinomas continues to provide insights into the carcinogenic process that are relevant to a broader understanding of tumour pathogenesis and to the development of targeted cancer therapies.


Subject(s)
Burkitt Lymphoma/virology , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/pathogenicity , Lymphoma, B-Cell/virology , Nasopharyngeal Neoplasms/virology , Humans
11.
Oncogene ; 28(44): 3903-14, 2009 Nov 05.
Article in English | MEDLINE | ID: mdl-19718044

ABSTRACT

Although frequently expressed in Epstein-Barr virus (EBV)-positive malignancies, the role that latent membrane protein 2A and 2B (LMP2A and LMP2B) have in the oncogenic process remains obscure. Here we show a novel function for these proteins in epithelial cells, namely, their ability to modulate signalling from type I/II interferon receptors (IFNRs). We show that LMP2A- and LMP2B-expressing epithelial cells show decreased responsiveness to interferon (IFN)alpha and IFNgamma, as assessed by STAT1 phosphorylation, ISGF3 and GAF-mediated binding to IFN-stimulated response element and IFNgamma-activated factor sequence elements and luciferase reporter activation. Transcriptional profiling highlighted the extent of this modulation, with both viral proteins impacting 'globally' on IFN-stimulated gene expression. Although not affecting the levels of cell-surface IFNRs, LMP2A and LMP2B accelerated the turnover of IFNRs through processes requiring endosome acidification. This function may form part of EBV's strategy to limit anti-viral responses and define a novel function for LMP2A and LMP2B in modulating signalling from receptors that participate in innate immune responses.


Subject(s)
Epithelial Cells/metabolism , Epstein-Barr Virus Infections/metabolism , Herpesvirus 4, Human/metabolism , Oncogene Proteins, Viral/metabolism , Receptors, Interferon/metabolism , Viral Matrix Proteins/metabolism , Cell Line , Endosomes/immunology , Endosomes/metabolism , Epithelial Cells/immunology , Epithelial Cells/virology , Epstein-Barr Virus Infections/immunology , Gene Expression Regulation/immunology , Herpesvirus 4, Human/immunology , Humans , Immunity, Innate , Interferon-Stimulated Gene Factor 3, gamma Subunit/immunology , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferon-alpha/immunology , Interferon-alpha/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Oncogene Proteins, Viral/immunology , Receptors, Interferon/immunology , Response Elements/immunology , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , Signal Transduction/immunology , Viral Matrix Proteins/immunology
12.
J Pathol ; 216(1): 83-92, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18566961

ABSTRACT

Although the latent membrane protein-1 (LMP1) of the Epstein-Barr virus (EBV) is believed to be important for the transformation of germinal centre (GC) B cells, the precise contribution of this viral oncogene to lymphoma development is poorly understood. In this study, we used a non-viral vector-based method to express LMP1 in primary human GC B cells. Gene expression profiling revealed that LMP1 induced in GC B cells transcriptional changes characteristic of Hodgkin's lymphoma cell lines. Strikingly, LMP1 down-regulated the expression of B-cell-specific genes including B-cell receptor components such as CD79A, CD79B, CD19, CD20, CD22, and BLNK. LMP1 also induced the expression of ID2, a negative regulator of B-cell differentiation. Our data suggest that in EBV-positive cases, LMP1 is likely to be a major contributor to the altered transcriptional pattern characteristic of Hodgkin/Reed-Sternberg cells, including the loss of B-cell identity.


Subject(s)
B-Lymphocytes/metabolism , Hodgkin Disease/genetics , Reed-Sternberg Cells/metabolism , Viral Matrix Proteins/physiology , B-Lymphocytes/virology , Hodgkin Disease/virology , Humans , Phenotype , Reed-Sternberg Cells/virology , Tissue Array Analysis/methods , Tumor Cells, Cultured
13.
Oncogene ; 27(21): 3021-31, 2008 May 08.
Article in English | MEDLINE | ID: mdl-18059336

ABSTRACT

Associations between p160 coactivator proteins and endocrine resistance have been described. Though thought to primarily interact with steroid receptors, the p160 proteins can also interact with non-nuclear receptor transcription factors including the MAP kinase effector proteins Ets. Here, we observed that in breast cancer cells resistant and insensitive to endocrine treatment, the growth factor EGF induced Ets-2 but not Ets-1 transcriptional regulation of the oncogene myc. Ets-2 regulation of myc was found to be reliant on the p160 proteins SRC-1 and SRC-3. In support of these molecular observations, strong associations were observed between the transcription factor, Ets-2 and its coactivator SRC-1 (P<0.01) and the target gene myc (P<0.0001) in a cohort of breast cancer patients with locally advanced disease. Expression of Ets-2, SRC-1 and c-Myc individually all associated with reduced disease-free survival (P<0.001, P<0.001 and P=0.002 respectively). There was no association between SRC-3 and disease-free survival (P=0.707). SRC-1 can utilize MAP kinase effector transcription factor Ets-2 to regulate the production of the oncogene myc. These signalling mechanisms may be important in the development of steroid resistant/independent breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/physiopathology , Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Proto-Oncogene Protein c-ets-2/metabolism , Proto-Oncogene Proteins c-myc/physiology , Tamoxifen/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , DNA-Binding Proteins , Female , Humans , Protein Binding , RNA-Binding Proteins , Transcription Factors
14.
Prostate Cancer Prostatic Dis ; 11(2): 187-93, 2008.
Article in English | MEDLINE | ID: mdl-17726452

ABSTRACT

The nitroreductase (NR)/CB1954 enzyme prodrug system has given promising results in pre-clinical studies and is currently being assessed in phase I and II clinical trials in prostate cancer. Enhanced cell killing by apparent immune-mediated mechanisms has been shown in pancreatic and colorectal cancer models, by co-expressing murine granulocyte macrophage colony-stimulating factor (GM-CSF) with NR in a single replication deficient adenoviral vector. This consists of the CMV immediate early promotor driving expression of NR, with an internal ribosome entry site (IRES) and the gene for murine GM-CSF (mGM-CSF). To examine if similar enhancement of tumour cell killing could be produced in prostate cancer, the TRAMP model was chosen. Results illustrate that the combination of suicide gene therapy using NR and CB1954, with cytokine stimulation with mGM-CSF gives an improved response compared with either modality alone. The mechanism of this improved response is however likely to be non-immune based as it lacks a memory effect.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Agents/therapeutic use , Aziridines/therapeutic use , Escherichia coli Proteins/metabolism , Genes, Transgenic, Suicide , Genetic Therapy , Genetic Vectors/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Immunotherapy , Nitroreductases/metabolism , Prodrugs/therapeutic use , Prostatic Neoplasms/therapy , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenoviridae/genetics , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Aziridines/pharmacokinetics , Aziridines/toxicity , Biotransformation , Combined Modality Therapy , Defective Viruses/genetics , Escherichia coli Proteins/genetics , Genes, Synthetic , Genetic Therapy/methods , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunotherapy/methods , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Nitroreductases/genetics , Prodrugs/pharmacokinetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Recombinant Proteins , Specific Pathogen-Free Organisms
15.
Oncogene ; 26(28): 4135-47, 2007 Jun 14.
Article in English | MEDLINE | ID: mdl-17486072

ABSTRACT

The Epstein-Barr virus (EBV)-encoded EBNA1 protein is expressed in all virus-associated tumors where it plays an essential role in the maintenance, replication and transcription of the EBV genome. Transcriptional profiling of EBNA1-expressing carcinoma cells demonstrated that EBNA1 also influences the expression of a range of cellular genes including those involved in translation, transcription and cell signaling. Of particular interest was the ability of EBNA1 to enhance expression of STAT1 and sensitize cells to interferon-induced STAT1 activation with resultant enhancement of major histocompatibility complex expression. A negative effect of EBNA1 on the expression of TGFbeta1-responsive betaig-h3 and PAI-1 genes was confirmed at the protein level in EBV-infected carcinoma cells. This effect resulted from the ability of EBNA1 to repress TGFbeta1-induced transcription via a reduction in the interaction of SMAD2 with SMAD4. More detailed analysis revealed that EBNA1 induces a lower steady-state level of SMAD2 protein as a consequence of increased protein turnover. These data show that EBNA1 can influence cellular gene transcription resulting in effects that may contribute to the development of EBV-associated tumors.


Subject(s)
Epstein-Barr Virus Nuclear Antigens/physiology , Herpesvirus 4, Human/physiology , STAT1 Transcription Factor/metabolism , Signal Transduction/physiology , Transcription, Genetic/physiology , Transforming Growth Factor beta/metabolism , Cell Line, Tumor , Humans , Reverse Transcriptase Polymerase Chain Reaction
16.
Br J Cancer ; 95(9): 1202-11, 2006 Nov 06.
Article in English | MEDLINE | ID: mdl-17024127

ABSTRACT

We used SEREX technology to identify novel tumour-associated antigens in patients with primary hepatocellular carcinoma and found serological responses to the polycomb group (PcG) protein BMI-1, which is overexpressed in a range of different tumour types. Further studies identified T-cell responses to both BMI-1 and another PcG protein, EZH2, in cancer patients and at relatively lower levels in some normal donors. We next identified several CD8+ T-cell epitopes derived from BMI-1 and EZH2 and demonstrated that EZH2-derived peptides elicited more significant interferon-gamma (IFN-gamma) release than BMI-1-derived peptides. That CD8(+) T cells were responsible for the observed responses was confirmed for EZH2 by both IFN-gamma capture assays and tetramer staining using an HLA-A0201-restricted, EZH2-derived YMSCSFLFNL (aa 666-674) epitope. The ability of YMSCSFLFNL (aa 666-674) to stimulate the in vitro expansion of specific T cells from peripheral blood lymphocytes was greatly enhanced when the CD25(+) T-cell population was depleted. EZH2-specific cytotoxic T lymphocyte clones specific for two HLA-A0201 epitopes were generated and found to recognise endogenously processed EZH2 in both HLA-matched fibroblasts and tumour cell lines. Given the widespread overexpression of PcG proteins in cancer and their critical role in oncogenesis, these data suggest that they may be useful targets for cancer immunotherapy.


Subject(s)
DNA-Binding Proteins/genetics , Neoplasms/pathology , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Amino Acid Sequence , Antigens, Neoplasm/analysis , Antigens, Neoplasm/genetics , Cell Line, Tumor , Cells, Cultured , Cytotoxicity, Immunologic/immunology , DNA-Binding Proteins/analysis , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Interferon-gamma/biosynthesis , Interleukin-2 Receptor alpha Subunit/analysis , Leukocytes, Mononuclear/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/analysis , Polycomb Repressive Complex 1 , Polycomb Repressive Complex 2 , Proto-Oncogene Proteins/analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/analysis
17.
Br J Cancer ; 95(10): 1404-9, 2006 Nov 20.
Article in English | MEDLINE | ID: mdl-17060941

ABSTRACT

Overexpression of HER2 is associated with an adverse prognosis in breast cancer. Despite this, the mechanism of its transcriptional regulation remains poorly understood. PEA3, a MAP kinase (MAPK)-activated member of the Ets transcription factor family has been implicated in the transcriptional regulation of HER2. The direction of its modulation remains controversial. We assessed relative levels of PEA3 expression and DNA binding in primary breast cultures derived from patient tumours (n=18) in the presence of an activated MAPK pathway using Western blotting and shift analysis. Expression of PEA3 in breast tumours from patients of known HER2 status (n=107) was examined by immunohistochemistry. In primary breast cancer cell cultures, growth factors induced interaction between PEA3 and its DNA response element. Upregulation of PEA3 expression in the presence of growth factors associated with HER2 positivity and axillary lymph node metastasis (P=0.034 and 0.049, respectively). PEA3 expression in breast cancer tissue associated with reduced disease-free survival (P<0.001), Grade III tumours (P<0.0001) and axillary lymph node metastasis (P=0.026). Co-expression of PEA3 and HER2 significantly associated with rate of recurrence compared to patients who expressed HER2 alone (P=0.0039). These data support a positive role for PEA3 in HER2-mediated oncogenesis in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Receptor, ErbB-2/genetics , Transcription Factors/metabolism , Axilla/pathology , Biomarkers, Tumor/metabolism , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Disease Progression , Electrophoretic Mobility Shift Assay , Epidermal Growth Factor/pharmacology , Female , Fibroblast Growth Factor 2/pharmacology , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Middle Aged , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Promoter Regions, Genetic , Receptor, ErbB-2/metabolism , Survival Rate , Transcription, Genetic , Tumor Cells, Cultured/drug effects , raf Kinases/metabolism
18.
Gene Ther ; 12(15): 1187-97, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15800660

ABSTRACT

Virus-directed enzyme prodrug therapy utilizing the bacterial enzyme nitroreductase delivered by a replication-defective adenovirus vector to activate the prodrug CB1954 is a promising strategy currently undergoing clinical trials in patients with a range of cancers. Similarly, selectively replicating oncolytic adenoviruses are entering clinical trials. An understanding of interactions between vector and target cell are critical to the development of these strategies. We demonstrate that adenovirus vectors activate cellular pathways that promote cell survival in an NF-kappaB-dependent manner, and consequently have a negative effect on the efficacy of cell killing induced by cancer gene therapy strategies. This provides a potential therapeutic target to enhance the cytotoxicity of these approaches.


Subject(s)
Genetic Therapy/methods , NF-kappa B/metabolism , Neoplasms/therapy , Oncolytic Virotherapy/methods , Signal Transduction , Adenoviridae/genetics , Aziridines/administration & dosage , Cell Line, Tumor , Enzyme Activation , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Neoplasms/metabolism , Neoplasms/virology , Nitroreductases/genetics , Nitroreductases/metabolism , Oncolytic Viruses/genetics , Prodrugs/administration & dosage , Virus Replication
19.
J Mol Endocrinol ; 33(3): 663-77, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15591026

ABSTRACT

Human pituitary tumor-transforming gene (PTTG), known also as securin, is a multifunctional protein implicated in the control of mitosis and the pathogenesis of thyroid, colon, oesophageal and other tumour types. Critical to PTTG function is a C-terminal double PXXP motif, forming a putative SH3-interacting domain and housing the gene's sole reported phosphorylation site. The exact role of phosphorylation and PXXP structure in the modulation of PTTG action in vitro remains poorly understood. We therefore examined the mitotic, transformation, proliferation and transactivation function of the C-terminal PXXP motifs of human PTTG. Live-cell imaging studies using an EGFP-PTTG construct indicated that PTTG's regulation of mitosis is retained regardless of phosphorylation status. Colony-formation assays demonstrated that phosphorylation of PTTG may act as a potent inhibitor of cell transformation. In proliferation assays, NIH-3T3 cells stable transfected and overexpressing mutations preventing PTTG phosphorylation (Phos-) showed significantly increased [3H]thymidine incorporation compared with WT, whereas mutants mimicking constitutive phosphorylation of PTTG (Phos+) exhibited reduced cell proliferation. We demonstrated that PTTG transactivation of FGF-2 in primary thyroid and PTTG-null cell lines was not affected by PTTG phosphorylation but was prevented by a mutant disrupting the PXXP motifs (SH3-). Taken together, our data suggest that PXXP structure and phosphorylation are likely to exert independent and critical influences upon PTTG's diverse actions in vitro.


Subject(s)
Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Amino Acid Motifs , Animals , Cell Proliferation/drug effects , Cell Transformation, Neoplastic , Cells, Cultured , Fibroblast Growth Factor 2/pharmacology , Humans , Mice , Mutation/genetics , Neoplasm Proteins/genetics , Phosphorylation/drug effects , Securin
20.
Infection ; 32(5): 257-70, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15624889

ABSTRACT

Nontuberculous mycobacteria (NTM) are important environmental pathogens that can cause a broad spectrum of diseases. The last few years brought several changes in this expanding field: The number of infections that can be associated with specific species as well as the number of new species as etiological agents has exploded due to the development of new diagnostic tools. The incidence of disseminated Mycobacterium avium complex (MAC) infections in HIV patients is decreasing with more potent anti-HIV treatments, while the rate of pulmonary NTM infection and disease as well as the prevalence of Buruli ulcer, a chronic, necrotizing, progressive ulcerous disease caused by Mycobacterium ulcerans, is increasing. The disease manifestations depend on the interaction between the specific mycobacterial pathogen and the host's immune system. This article presents an update of the epidemiology, diagnosis, and treatment of NTM-associated pulmonary disease, lymphadenitis, skin and soft tissue disease, skeletal infection and foreign body- and catheter-related NTM infections.


Subject(s)
Mycobacterium Infections, Nontuberculous , Anti-Bacterial Agents/therapeutic use , Humans , Mycobacterium Infections, Nontuberculous/diagnosis , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...