Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 73(1): 450-8, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23288917

ABSTRACT

Senescence, a terminal cell proliferation arrest, can be triggered by oncogenes. Oncogene-induced senescence is classically considered a tumor defense barrier. However, several findings show that, under certain circumstances, senescent cells may favor tumor progression because of their secretory phenotype. Here, we show that the expression in different breast epithelial cell lines of p95HER2, a constitutively active fragment of the tyrosine kinase receptor HER2, results in either increased proliferation or senescence. In senescent cells, p95HER2 elicits a secretome enriched in proteases, cytokines, and growth factors. This secretory phenotype is not a mere consequence of the senescence status and requires continuous HER2 signaling to be maintained. Underscoring the functional relevance of the p95HER2-induced senescence secretome, we show that p95HER2-induced senescent cells promote metastasis in vivo in a non-cell-autonomous manner.


Subject(s)
Breast Neoplasms/pathology , Cellular Senescence/physiology , Receptor, ErbB-2/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Breast Neoplasms/metabolism , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Transplantation, Heterologous
2.
Cancer Res ; 70(21): 8537-46, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20978202

ABSTRACT

Current classification of breast cancers depends in great part on the expression of human epidermal growth factor receptor 2 (HER2), a cell surface tyrosine kinase receptor, and estrogen receptor (ER), the nuclear receptor for estrogen. In addition to reliable biomarkers, these receptors are targets of effective and widely used antitumor drugs. During malignant progression, HER2 and ER can establish an intricate cross-talk. In some cases, HER2 overexpression leads to the downregulation of ER and undermining of anti-ER therapies. A subgroup of HER2-positive breast cancer patients with poor prognosis expresses a heterogeneous collection of HER2 carboxy-terminal fragments (CTF) collectively known as p95HER2. One of these fragments, 611-CTF, is oncogenic in a variety of preclinical models. However, because of the lack of an appropriate tool to specifically analyze its levels in the clinical setting, the value of 611-CTF as a biomarker has not been established yet. Here, we show that 611-CTF induces resistance to antiestrogen therapy and a more pronounced down-modulation of ER than that induced by full-length HER2. To validate this effect in breast cancer samples, we developed specific anti-611-CTF antibodies. With these antibodies, we showed that, whereas the frequency of ER positivity in HER2-positive/611-CTF-negative tumors (72.6%) is similar to that reported for HER2-negative tumors (70-80%), the number of ER-positive tumors in the 611-CTF-positive subgroup is very low (31.2%). These results reveal a mechanism of ER regulation mediated by HER2, which suggests a new strategy to improve responses to endocrine therapy in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms, Hormone-Dependent/metabolism , Receptor, ErbB-2/physiology , Tamoxifen/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Antineoplastic Agents, Hormonal/therapeutic use , Apoptosis , Blotting, Western , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Drug Resistance, Neoplasm , Female , Humans , Immunoenzyme Techniques , Immunoprecipitation , In Situ Hybridization, Fluorescence , Mice , Mice, Nude , Peptide Fragments/immunology , Protein Structure, Tertiary , RNA, Messenger/genetics , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Clin Cancer Res ; 16(9): 2688-95, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20406840

ABSTRACT

PURPOSE: A subgroup of human epidermal growth factor receptor 2 (HER2)-overexpressing breast tumors coexpresses p95HER2, a truncated HER2 receptor that retains a highly functional HER2 kinase domain but lacks the extracellular domain and results in intrinsic trastuzumab resistance. We hypothesized that lapatinib, a HER2 tyrosine kinase inhibitor, would be active in these tumors. We have studied the correlation between p95HER2 expression and response to lapatinib, both in preclinical models and in the clinical setting. EXPERIMENTAL DESIGN: Two different p95HER2 animal models were used for preclinical studies. Expression of p95HER2 was analyzed in HER2-overexpressing breast primary tumors from a first-line lapatinib monotherapy study (EGF20009) and a second-line lapatinib in combination with capecitabine study (EGF100151). p95HER2 expression was correlated with overall response rate (complete + partial response), clinical benefit rate (complete response + partial response + stable disease > or =24 wk), and progression-free survival using logistic regression and Cox proportional hazard models. RESULTS: Lapatinib inhibited tumor growth and the HER2 downstream signaling of p95HER2-expressing tumors. A total of 68 and 156 tumors from studies EGF20009 and EGF100151 were evaluable, respectively, for p95HER2 detection. The percentage of p95HER2-positive patients was 20.5% in the EGF20009 study and 28.5% in the EGF100151 study. In both studies, there was no statistically significant difference in progression-free survival, clinical benefit rate, and overall response rate between p95HER2-positive and p95HER2-negative tumors. CONCLUSIONS: Lapatinib as a monotherapy or in combination with capecitabine seems to be equally effective in patients with p95HER2-positive and p95HER2-negative HER2-positive breast tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , ErbB Receptors/metabolism , Quinazolines/therapeutic use , Receptor, ErbB-2/metabolism , 3T3 Cells , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Capecitabine , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Female , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Humans , Immunoblotting , In Situ Hybridization, Fluorescence , Lapatinib , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Quinazolines/administration & dosage , Randomized Controlled Trials as Topic , Receptor, ErbB-2/genetics , Survival Analysis , Treatment Outcome , Xenograft Model Antitumor Assays
4.
J Biol Chem ; 284(37): 25302-13, 2009 Sep 11.
Article in English | MEDLINE | ID: mdl-19589785

ABSTRACT

A group of breast cancer patients with a higher probability of developing metastasis expresses a series of carboxyl-terminal fragments (CTFs) of the tyrosine kinase receptor HER2. One of these fragments, 611-CTF, is a hyperactive form of HER2 that constitutively establishes homodimers maintained by disulfide bonds, making it an excellent model to study overactivation of HER2 during tumor progression and metastasis. Here we show that expression of 611-CTF increases cell motility in a variety of assays. Since cell motility is frequently regulated by phosphorylation/dephosphorylation, we looked for phosphoproteins mediating the effect of 611-CTF using two alternative proteomic approaches, stable isotope labeling with amino acids in cell culture and difference gel electrophoresis, and found that the latter is particularly well suited to detect changes in multiphosphorylated proteins. The difference gel electrophoresis screening identified cortactin, a cytoskeleton-binding protein involved in the regulation of cell migration, as a phosphoprotein probably regulated by 611-CTF. This result was validated by characterizing cortactin in cells expressing this HER2 fragment. Finally, we showed that the knockdown of cortactin impairs 611-CTF-induced cell migration. These results suggest that cortactin is a target of 611-CTF involved in the regulation of cell migration and, thus, in the metastatic behavior of breast tumors expressing this CTF.


Subject(s)
Cortactin/chemistry , Gene Expression Regulation, Neoplastic , Receptor, ErbB-2/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Humans , Microscopy, Fluorescence , Models, Biological , Peptides/chemistry , Phosphoproteins/chemistry , Phosphorylation , Protein Structure, Tertiary , Signal Transduction
5.
Mol Cell Biol ; 29(12): 3319-31, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19364815

ABSTRACT

HER2 is a tyrosine kinase receptor causally involved in cancer. A subgroup of breast cancer patients with particularly poor clinical outcomes expresses a heterogeneous collection of HER2 carboxy-terminal fragments (CTFs). However, since the CTFs lack the extracellular domain that drives dimerization and subsequent activation of full-length HER2, they are in principle expected to be inactive. Here we show that at low expression levels one of these fragments, 611-CTF, activated multiple signaling pathways because of its unanticipated ability to constitutively homodimerize. A transcriptomic analysis revealed that 611-CTF specifically controlled the expression of genes that we found to be correlated with poor prognosis in breast cancer. Among the 611-CTF-regulated genes were several that have previously been linked to metastasis, including those for MET, EPHA2, matrix metalloproteinase 1, interleukin 11, angiopoietin-like 4, and different integrins. It is thought that transgenic mice overexpressing HER2 in the mammary glands develop tumors only after acquisition of activating mutations in the transgene. In contrast, we show that expression of 611-CTF led to development of aggressive and invasive mammary tumors without the need for mutations. These results demonstrate that 611-CTF is a potent oncogene capable of promoting mammary tumor progression and metastasis.


Subject(s)
Breast Neoplasms/etiology , Peptide Fragments/physiology , Receptor, ErbB-2/physiology , Adenocarcinoma/etiology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Amino Acid Sequence , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/secondary , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Models, Biological , Molecular Sequence Data , Peptide Chain Initiation, Translational , Peptide Fragments/chemistry , Peptide Fragments/genetics , Prognosis , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/genetics , Signal Transduction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...