Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Br J Cancer ; 112(12): 1895-903, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26010411

ABSTRACT

BACKGROUND: Identification and validation of a targeted therapy for triple-negative breast cancer (TNBC), that is, breast cancers negative for oestrogen receptors, progesterone receptors and HER2 amplification, is currently one of the most urgent problems in breast cancer treatment. EGFR is one of the best-validated driver genes for TNBC. EGFR is normally activated following the release of ligands such as TGFα, mediated by the two MMP-like proteases ADAM (a disintegrin and metalloproteinase)-10 and ADAM-17. The aim of this study was to investigate the antitumour effects of a monoclonal antibody against ADAM-17 on an in vitro model of TNBC. METHODS: We investigated an inhibitory cross-domain humanised monoclonal antibody targeting both the catalytic domain and the cysteine-rich domain of ADAM17-D1(A12) in the HCC1937 and HCC1143 cell lines. RESULTS: D1(A12) was found to significantly inhibit the release of TGFα, and to decrease downstream EGFR-dependent cell signalling. D1(A12) treatment reduced proliferation in two-dimensional clonogenic assays, as well as growth in three-dimensional culture. Furthermore, D1(A12) reduced invasion of HCC1937 cells and decreased migration of HCC1143 cells. Finally, D1(A12) enhanced cell death in HCC1143 cells. CONCLUSION: Our in vitro findings suggest that targeting ADAM-17 with D1(A12) may have anticancer activity in TNBC cells.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Antibodies, Monoclonal, Humanized/pharmacology , Triple Negative Breast Neoplasms/drug therapy , ADAM Proteins/immunology , ADAM17 Protein , Antibodies, Monoclonal, Humanized/immunology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Progression , Humans , Molecular Targeted Therapy , Signal Transduction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
2.
Clin Exp Immunol ; 181(1): 39-50, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25943872

ABSTRACT

Caspases are a group of proteolytic enzymes involved in the co-ordination of cellular processes, including cellular homeostasis, inflammation and apoptosis. Altered activity of caspases, particularly caspase-1, has been implicated in the development of intestinal diseases, such as inflammatory bowel disease (IBD) and colorectal cancer (CRC). However, the involvement of two related inflammatory caspase members, caspases-4 and -5, during intestinal homeostasis and disease has not yet been established. This study demonstrates that caspases-4 and -5 are involved in IBD-associated intestinal inflammation. Furthermore, we found a clear correlation between stromal caspase-4 and -5 expression levels, inflammation and disease activity in ulcerative colitis patients. Deregulated intestinal inflammation in IBD patients is associated with an increased risk of developing CRC. We found robust expression of caspases-4 and -5 within intestinal epithelial cells, exclusively within neoplastic tissue, of colorectal tumours. An examination of adjacent normal, inflamed and tumour tissue from patients with colitis-associated CRC confirmed that stromal expression of caspases-4 and -5 is increased in inflamed and dysplastic tissue, while epithelial expression is restricted to neoplastic tissue. In addition to identifying caspases-4 and -5 as potential targets for limiting intestinal inflammation, this study has identified epithelial-expressed caspases-4 and -5 as biomarkers with diagnostic and therapeutic potential in CRC.


Subject(s)
Caspases, Initiator/biosynthesis , Caspases/biosynthesis , Colitis, Ulcerative/pathology , Colorectal Neoplasms/pathology , Intestinal Mucosa/pathology , Adult , Aged , Biomarkers , Colitis, Ulcerative/diagnosis , Colorectal Neoplasms/diagnosis , Epithelial Cells/metabolism , Female , Humans , Inflammation/pathology , Intestinal Mucosa/cytology , Male , Middle Aged , Young Adult
3.
Ann Oncol ; 24(2): 362-369, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22967992

ABSTRACT

BACKGROUND: Validated targeted therapy is currently unavailable for patients with invasive breast cancer negative for oestrogen receptors, progesterone receptors and HER2 [i.e., those with triple-negative (TN) disease]. ADAM-17 is a protease involved in the activations of several ligands that bind to and promotes intracellular signalling from the EGFR/HER family of receptors. PATIENTS AND METHODS: Expression of ADAM-17 was measured in 86 triple-negative and 96 non-triple-negative breast cancers. The ADAM-17 specific inhibitor, PF-5480090 (TMI-002, Pfizer) was tested in a panel of breast cancer cell lines for effects on functional outputs. RESULTS: In this study we show using both Western blotting and immunohistochemistry that ADAM-17 is expressed at significantly higher levels in TN than non-TN breast cancers. Using a panel of breast cancer cell lines in culture, PF-5480090 was found to decrease release of the EGFR ligand, TGF-alpha, decrease levels of phosphorylated EGFR and block cell proliferation in a cell-type-dependent manner. Potentially important was the finding of a significant and moderately strong correlation between ADAM-17 activity and extent of proliferation inhibition by PF-5480090 (r = 0.809; p = 0.003; n = 11). Pretreatment of cell lines with PF-5480090 enhanced response to several different cytotoxic and anti-EGFR/HER agents. CONCLUSION: It is concluded that inhibition of ADAM-17, especially in combination with chemotherapy or anti-EGFR/HER inhibitors, may be a new approach for treating breast cancer, including patients with TN disease.


Subject(s)
ADAM Proteins/antagonists & inhibitors , ADAM Proteins/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , ErbB Receptors/metabolism , ADAM Proteins/biosynthesis , ADAM17 Protein , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Molecular Targeted Therapy , Phosphorylation/drug effects , RNA, Messenger/biosynthesis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Signal Transduction/drug effects , Transforming Growth Factor alpha/metabolism
4.
Br J Cancer ; 104(2): 338-44, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21119660

ABSTRACT

BACKGROUND: The eicosanoid signalling pathway promotes the progression of malignancies through the production of proliferative prostaglandins (PGs). Cytosolic phospholipase A(2)α (cPLA(2)α) activity provides the substrate for cyclooxygenase-dependent PG release, and we have previously found that cPLA(2)α expression correlated with EGFR/HER2 over-expression in a small number of breast cancer cell lines. METHODS: The importance of differential cPLA(2)α activity in clinical breast cancer was established by relating the expression of cPLA(2)α in tissue samples from breast cancer patients, and two microarray-based gene expression datasets to different clinicopathological and therapeutic parameters. RESULTS: High cPLA(2)α mRNA expression correlated with clinical parameters of poor prognosis, which are characteristic of highly invasive tumours of the HER2-positive and basal-like subtype, including low oestrogen receptor expression and high EGFR expression. High cPLA(2)α expression decreased overall survival in patients with luminal cancers, and correlated with a reduced effect of tamoxifen treatment. The cPLA(2)α expression was an independent predictive parameter of poor response to endocrine therapy in the first 5 years of follow-up. CONCLUSION: This study shows a role of cPLA(2)α in luminal breast cancer progression, in which the enzyme could represent a novel therapeutic target and a predictive marker.


Subject(s)
Breast Neoplasms/enzymology , Cytosol/enzymology , ErbB Receptors/metabolism , Phospholipases A2/metabolism , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cohort Studies , Female , Humans , Oligonucleotide Array Sequence Analysis , Phospholipases A2/genetics , Prognosis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
5.
Radiol Med ; 73(5): 390-3, 1987 May.
Article in Italian | MEDLINE | ID: mdl-3589012

ABSTRACT

We studied 19 consecutive subjects affected by effort angina using following tests: ecg stress test, stress 201-Tl scan, coronary arteriography; 201-Tl scan, coronary arteriography, hemodynamic, echo 2 D, ecgraphic monitoring during Dipyridamole test (D). Basing on coronary arteriography results we divided patients; in the group A (10 patients with significant stenoses greater than or equal to 50%) stress ecg and scintigraphy were positive in 9 patients; Dipyridamole test induced angor and ecgraphic changes in 5 patients and in 4 left ventricle wall motion disorders, 201-Tl scan was positive in all 9 patients tested. In the group B (9 subjects with no significant stenosis) ecgraphic changes were observed in 2 subjects and 201-Tl scan was positive in 6 subjects; D induced in 2 cases angor, in 1 case ecgraphic changes, in 1 case left ventricle wall motion disorders and the same 201-Tl defects in 6 previously individualized patients. In both groups we observed at coronary arteriography during D identical findings in comparison with the immediately before performed. In our experience D infusion is confirmed as provocative test of ischemia. The same ischemic pattern observed at stress and Dipyridamole scintigraphy in patients with no significant coronary stenosis suggests as pathogenetic mechanism the regional lack of dilatory reserve.


Subject(s)
Coronary Disease/diagnosis , Dipyridamole , Radioisotopes , Thallium , Adult , Aged , Coronary Disease/diagnostic imaging , Coronary Disease/physiopathology , Electrocardiography , Female , Humans , Male , Middle Aged , Monitoring, Physiologic , Radionuclide Imaging
SELECTION OF CITATIONS
SEARCH DETAIL