Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 19(19): 5390-401, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23948973

ABSTRACT

PURPOSE: Mutations in receptor tyrosine kinase (RTK) genes can confer resistance to receptor-targeted therapies. A T798M mutation in the HER2 oncogene has been shown to confer resistance to the tyrosine kinase inhibitor (TKI) lapatinib. We studied the mechanisms of HER2-T798M-induced resistance to identify potential strategies to overcome that resistance. EXPERIMENTAL DESIGN: HER2-T798M was stably expressed in BT474 and MCF10A cells. Mutant cells and xenografts were evaluated for effects of the mutation on proliferation, signaling, and tumor growth after treatment with combinations of inhibitors targeting the EGFR/HER2/HER3/PI3K axis. RESULTS: A low 3% allelic frequency of the T798M mutant shifted 10-fold the IC50 of lapatinib. In mutant-expressing cells, lapatinib did not block basal phosphorylation of HER2, HER3, AKT, and ERK1/2. In vitro kinase assays showed increased autocatalytic activity of HER2-T798M. HER3 association with PI3K p85 was increased in mutant-expressing cells. BT474-T798M cells were also resistant to the HER2 antibody trastuzumab. These cells were sensitive to the pan-PI3K inhibitors BKM120 and XL147 and the irreversible HER2/EGFR TKI afatinib but not the MEK1/2 inhibitor CI-1040, suggesting continued dependence of the mutant cells on ErbB receptors and downstream PI3K signaling. BT474-T798M cells showed increased expression of the EGFR ligands EGF, TGFα, amphiregulin, and HB-EGF. Addition of the EGFR neutralizing antibody cetuximab or lapatinib restored trastuzumab sensitivity of BT474-T798M cells and xenografts, suggesting that increased EGFR ligand production was causally associated with drug resistance. CONCLUSIONS: Simultaneous blockade of HER2 and EGFR should be an effective treatment strategy against HER2 gene-amplified breast cancer cells harboring T798M mutant alleles.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , ErbB Receptors/metabolism , Gene Expression , Mutation , Receptor, ErbB-2/genetics , Amino Acid Substitution , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Catalysis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Codon , Disease Models, Animal , ErbB Receptors/antagonists & inhibitors , Female , Gene Amplification , Humans , Ligands , Mice , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Xenograft Model Antitumor Assays
2.
Cancer Res ; 71(5): 1871-82, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21324925

ABSTRACT

In breast cancer cells with HER2 gene amplification, HER2 receptors exist on the cell surface as monomers, homodimers, and heterodimers with EGFR/HER3. The therapeutic antibody trastuzumab, an approved therapy for HER2(+) breast cancer, cannot block ligand-induced HER2 heterodimers, suggesting it cannot effectively inhibit HER2 signaling. Hence, HER2 oligomeric states may predict the odds of a clinical response to trastuzumab in HER2-driven tumors. To test this hypothesis, we generated nontransformed human MCF10A mammary epithelial cells stably expressing a chimeric HER2-FKBP molecule that could be conditionally induced to homodimerize by adding the FKBP ligand AP1510, or instead induced to heterodimerize with EGFR or HER3 by adding the heterodimer ligands EGF/TGFα or heregulin. AP1510, EGF, and heregulin each induced growth of MCF10A cells expressing HER2-FKBP. Trastuzumab inhibited homodimer-mediated but not heterodimer-mediated cell growth. In contrast, the HER2 antibody pertuzumab, which blocks HER2 heterodimerization, inhibited growth induced by heregulin but not AP1510. Lastly, the HER2/EGFR tyrosine kinase inhibitor lapatinib blocked both homodimer- and heterodimer-induced growth. AP1510 triggered phosphorylation of Erk1/2 but not AKT, whereas trastuzumab inhibited AP1510-induced Erk1/2 phosphorylation and Shc-HER2 homodimer binding, but not TGFα-induced AKT phosphorylation. Consistent with these observations, high levels of HER2 homodimers correlated with longer time to progression following trastuzumab therapy in a cohort of patients with HER2-overexpressing breast cancer. Together, our findings confirm the notion that HER2 oligomeric states regulate HER2 signaling, also arguing that trastuzumab sensitivity of homodimers may reflect their inability to activate the PI3K (phosphoinositide 3-kinase)/AKT pathway. A clinical implication of our results is that high levels of HER2 homodimers may predict a positive response to trastuzumab.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Protein Multimerization/physiology , Receptor, ErbB-2/metabolism , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized , Breast Neoplasms/chemistry , Breast Neoplasms/drug therapy , Cells, Cultured , Female , Gene Expression , Gene Expression Profiling , Humans , Immunoblotting , Immunoprecipitation , Lapatinib , Middle Aged , Oligonucleotide Array Sequence Analysis , Quinazolines/pharmacology , Receptor, ErbB-2/chemistry , Signal Transduction/drug effects , Signal Transduction/physiology , Trastuzumab
3.
J Clin Oncol ; 29(2): 166-73, 2011 Jan 10.
Article in English | MEDLINE | ID: mdl-21135276

ABSTRACT

PURPOSE: Phosphatase and tensin homolog (PTEN) loss or activating mutations of phosphoinositol-3 (PI3) kinase (PIK3CA) may be associated with trastuzumab resistance. Trastuzumab, the humanized human epidermal growth factor receptor 2 (HER2) monoclonal antibody, and lapatinib, an epidermal growth factor receptor/HER2 tyrosine kinase inhibitor, are both established treatments for HER2-overexpressing breast cancers. Understanding of the cellular response to HER2-targeted therapies is needed to tailor treatments and to identify patients less likely to benefit. METHODS: We evaluated the effect of trastuzumab or lapatinib in three HER2-overexpressing cell lines. We confirmed the in vitro observations in two neoadjuvant clinical trials in patients with HER2 overexpression; 35 patients received trastuzumab as a single agent for the first 3 weeks, then docetaxel every 3 weeks for 12 weeks (trastuzumab regimen), whereas 49 patients received lapatinib as a single agent for 6 weeks, followed by trastuzumab/docetaxel for 12 weeks before primary surgery (lapatinib regimen). Apoptosis, Ki67, p-MAPK, p-AKT, and PTEN were assessed by immunohistochemistry. Genomic DNA was sequenced for PIK3CA mutations. RESULTS: Under low PTEN conditions, in vitro data indicate that lapatinib alone and in combination with trastuzumab was effective in decreasing p-MAPK and p-AKT levels, whereas trastuzumab was ineffective. In the clinical trials, we confirmed that low PTEN or activating mutation in PIK3CA conferred resistance to the trastuzumab regimen (P = .015), whereas low PTEN tumors were associated with a high pathologic complete response rate (P = .007). CONCLUSION: Activation of PI3 kinase pathway is associated with trastuzumab resistance, whereas low PTEN predicted for response to lapatinib. These observations support clinical trials with the combination of both agents.


Subject(s)
Antibodies, Monoclonal/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Quinazolines/pharmacology , Receptor, ErbB-2/biosynthesis , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/genetics , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Clinical Trials, Phase II as Topic , Docetaxel , Enzyme Activation , Female , Humans , Lapatinib , Middle Aged , Mutation , Neoadjuvant Therapy , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Quinazolines/administration & dosage , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Taxoids/administration & dosage , Transfection , Trastuzumab
4.
Clin Cancer Res ; 15(23): 7266-76, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19934303

ABSTRACT

PURPOSE: A significant fraction of HER2-overexpressing breast cancers exhibit resistance to the HER2 antibody trastuzumab. Hyperactivity of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway confers trastuzumab resistance, and mammalian target of rapamycin (mTOR) is a major downstream effector of PI3K/AKT. Therefore, we examined whether mTOR inhibitors synergize with trastuzumab. EXPERIMENTAL DESIGN: Immunocompetent mice bearing HER2(+) mammary tumors were treated with trastuzumab, the mTOR inhibitor rapamycin, or the combination. Mice were imaged for tumor cell death using an optical Annexin-V probe and with [(18)F]FDG positron emission tomography. The signaling and growth effects of the mTOR inhibitor RAD001 on HER2(+) cells treated with trastuzumab or lapatinib were evaluated. RESULTS: Treatment of mice with trastuzumab plus rapamycin was more effective than single-agent treatments, inducing complete regression of 26 of 26 tumors. The combination induced tumor cell death (Annexin-V binding) and inhibited FDG uptake. Rapamycin inhibited mTOR and tumor cell proliferation as determined by phosphorylated S6 and Ki-67 immunohistochemistry, respectively. In culture, the combination of RAD001 plus trastuzumab inhibited cell growth more effectively than either drug alone. Trastuzumab partially decreased PI3K but not mTOR activity. Knockdown of TSC2 resulted in HER2-independent activation of mTOR and dampened the response to trastuzumab and lapatinib. Treatment with the HER2 inhibitor lapatinib decreased phosphorylated S6 and growth in TSC2-expressing cells but not in TSC2-knockdown cells. CONCLUSIONS: Inhibition of PI3K and mTOR are required for the growth-inhibitory effect of HER2 antagonists. These findings collectively support the combined use of trastuzumab and mTOR inhibitors for the treatment of HER2(+) breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, ErbB-2/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , Female , Humans , Mice , Mice, SCID , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , TOR Serine-Threonine Kinases , Trastuzumab
5.
Cancer Res ; 69(22): 8797-806, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19887618

ABSTRACT

Tumor-microenvironment interactions are increasingly recognized to influence tumor progression. To understand the competitive dynamics of tumor cells in diverse microenvironments, we experimentally parameterized a hybrid discrete-continuum mathematical model with phenotypic trait data from a set of related mammary cell lines with normal, transformed, or tumorigenic properties. Surprisingly, in a resource-rich microenvironment, with few limitations on proliferation or migration, transformed (but not tumorigenic) cells were most successful and outcompeted other cell types in heterogeneous tumor simulations. Conversely, constrained microenvironments with limitations on space and/or growth factors gave a selective advantage to phenotypes derived from tumorigenic cell lines. Analysis of the relative performance of each phenotype in constrained versus unconstrained microenvironments revealed that, although all cell types grew more slowly in resource-constrained microenvironments, the most aggressive cells were least affected by microenvironmental constraints. A game theory model testing the relationship between microenvironment resource availability and competitive cellular dynamics supports the concept that microenvironmental independence is an advantageous cellular trait in resource-limited microenvironments.


Subject(s)
Breast Neoplasms/pathology , Models, Theoretical , Cell Adhesion/physiology , Cell Communication/physiology , Cell Line, Tumor , Disease Progression , Extracellular Matrix/metabolism , Female , Game Theory , Humans
6.
Cancer Res ; 69(10): 4192-201, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19435893

ABSTRACT

Knockdown of the tumor suppressor phosphatase Phosphatase and tensin homologue deleted on chromosome 10 (PTEN) with shRNA in three estrogen receptor (ER)-positive breast cancer cell lines resulted in increased phosphatidylinositol-3 kinase (PI3K) and AKT activities, resistance to tamoxifen and fulvestrant, and hormone-independent growth. PTEN knockdown induced the up-regulation of ER transcriptional activity in MCF-7 cells but decreased ER protein levels and transcriptional activity in T47D and MDA-361 cells. Tamoxifen and fulvestrant treatment inhibited estradiol-induced ER transcriptional activity in all shPTEN cell lines but did not abrogate the increased cell proliferation induced by PTEN knockdown. PTEN knockdown increased basal and ligand-induced activation of the insulin-like growth factor-I (IGF-I) and ErbB3 receptor tyrosine kinases, and prolonged the association of the p85 PI3K subunit with the IGF-I receptor (IGF-IR) effector insulin receptor substrate-1 and with ErbB3, implicating PTEN in the modulation of signaling upstream of PI3K. Consistent with these data, PTEN levels inversely correlated with levels of tyrosine-phosphorylated IGF-IR in tissue lysate arrays of primary breast cancers. Inhibition of IGF-IR and/or ErbB2-mediated activation of ErbB3 with tyrosine kinase inhibitors restored hormone dependence and the growth inhibitory effect of tamoxifen and fulvestrant on shPTEN cells, suggesting that cotargeting both ER and receptor tyrosine kinase pathways holds promise for the treatment of patients with ER+, PTEN-deficient breast cancers.


Subject(s)
Breast Neoplasms/genetics , Chromosome Deletion , Chromosomes, Human, Pair 19 , Drug Resistance, Neoplasm , Estrogen Receptor Modulators/therapeutic use , PTEN Phosphohydrolase/deficiency , Receptor, ErbB-3/physiology , Receptor, IGF Type 1/physiology , Breast Neoplasms/pathology , Cell Division , Cell Line, Tumor , Female , Genes, Reporter , Humans , Receptor, ErbB-2/physiology , Transcription, Genetic
7.
J Biol Chem ; 282(8): 5661-9, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17204482

ABSTRACT

Using two-dimensional difference gel electrophoresis, we identified the tumor suppressor gene maspin as a transforming growth factor beta (TGFbeta) target gene in human mammary epithelial cells. TGFbeta up-regulatesMaspin expression both at the RNA and protein levels. This up-regulation required Smad2/3 function and intact p53-binding elements in the Maspin promoter. DNA affinity immunoblot and chromatin immunoprecipitation revealed the presence of both Smads and p53 at the Maspin promoter in TGFbeta-treated cells, suggesting that both transcription factors cooperate to induce Maspin transcription. TGFbeta did not activate Maspin-luciferase reporter in p53-mutant MDA-MB-231 breast cancer cells, which exhibit methylation of the endogenous Maspin promoter. Expression of ectopic p53, however, restored ligand-induced association of Smad2/3 with a transfected Maspin promoter. Stable transfection of Maspin inhibited basal and TGFbeta-stimulated MDA-MB-231 cell motility. Finally, knockdown of endogenous Maspin in p53 wild-type MCF10A/HER2 cells enhanced basal and TGFbeta-stimulated motility. Taken together, these data support cooperation between the p53 and TGFbeta tumor suppressor pathways in the induction of Maspin expression, thus leading to inhibition of cell migration.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Human/metabolism , Serpins/biosynthesis , Signal Transduction/drug effects , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Epithelial Cells/cytology , Genes, Tumor Suppressor , Humans , Mammary Glands, Human/cytology , Promoter Regions, Genetic/physiology , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Up-Regulation/physiology
8.
Cancer Cell ; 10(1): 25-38, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16843263

ABSTRACT

HER2/Neu gene mutations have been identified in lung cancer. Expression of a HER2 mutant containing a G776(YVMA) insertion in exon 20 was more potent than wild-type HER2 in associating with and activating signal transducers, phosphorylating EGFR, and inducing survival, invasiveness, and tumorigenicity. HER2(YVMA) transphosphorylated kinase-dead EGFR(K721R) and EGFR(WT) in the presence of EGFR tyrosine kinase inhibitors (TKIs). Knockdown of mutant HER2 in H1781 lung cancer cells increased apoptosis and restored sensitivity to EGFR TKIs. The HER2 inhibitors lapatinib, trastuzumab, and CI-1033 inhibited growth of H1781 cells and cells expressing exogenous HER2(YVMA). These data suggest that (1) HER2(YVMA) activates cellular substrates more potently than HER2(WT); and (2) cancer cells expressing this mutation remain sensitive to HER2-targeted therapies but insensitive to EGFR TKIs.


Subject(s)
Drug Resistance, Neoplasm/genetics , ErbB Receptors/metabolism , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Female , Gefitinib , Humans , Mice , Mice, Nude , Models, Biological , Morpholines/pharmacology , Phosphorylation/drug effects , Quinazolines/pharmacology , RNA, Small Interfering/genetics , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Transforming Growth Factor alpha/pharmacology , Trastuzumab , Xenograft Model Antitumor Assays
9.
EMBO J ; 23(22): 4506-16, 2004 Nov 10.
Article in English | MEDLINE | ID: mdl-15510220

ABSTRACT

Wild-type alpha-synuclein, a protein of unknown function, has received much attention because of its involvement in a series of diseases that are known as synucleinopathies. We find that long-lasting potentiation of synaptic transmission between cultured hippocampal neurons is accompanied by an increase in the number of alpha-synuclein clusters. Conversely, suppression of alpha-synuclein expression through antisense nucleotide and knockout techniques blocks the potentiation, as well as the glutamate-induced increase in presynaptic functional bouton number. Consistent with these findings, alpha-synuclein introduction into the presynaptic neuron of a pair of monosynaptically connected cells causes a rapid and long-lasting enhancement of synaptic transmission, and rescues the block of potentiation in alpha-synuclein null mouse cultures. Also, we report that the application of nitric oxide (NO) increases the number of alpha-synuclein clusters, and inhibitors of NO-synthase block this increase, supporting the hypothesis that NO is involved in the enhancement of the number of alpha-synuclein clusters. Thus, alpha-synuclein is involved in synaptic plasticity by augmenting transmitter release from the presynaptic terminal.


Subject(s)
Nerve Tissue Proteins/metabolism , Synaptic Transmission/physiology , Animals , Animals, Newborn , Blotting, Western , Cells, Cultured , Electrophysiology , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , Exocytosis/drug effects , Hippocampus/cytology , Immunohistochemistry , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/drug effects , Neurons/ultrastructure , Nitric Oxide/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/drug effects , Oligonucleotides, Antisense/pharmacology , Patch-Clamp Techniques , Rats , Recombinant Fusion Proteins/metabolism , Synapses/metabolism , Synaptic Transmission/drug effects , Synucleins , alpha-Synuclein
SELECTION OF CITATIONS
SEARCH DETAIL
...