Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Oncogene ; 35(42): 5552-5564, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27086929

ABSTRACT

High-level amplification of the protein phosphatase PPM1D (WIP1) is present in a subset of medulloblastomas (MBs) that have an expression profile consistent with active Sonic Hedgehog (SHH) signaling. We found that WIP1 overexpression increased expression of Shh target genes and cell proliferation in response to Shh stimulation in NIH3T3 and cerebellar granule neuron precursor cells in a p53-independent manner. Thus, we developed a mouse in which WIP1 is expressed in the developing brain under control of the Neurod2 promoter (ND2:WIP1). The external granule layer (EGL) in early postnatal ND2:WIP1 mice exhibited increased proliferation and expression of Shh downstream targets. MB incidence increased and survival decreased when ND2:WIP1 mice were crossed with an Shh-activated MB mouse model. Conversely, Wip1 knockout significantly suppressed MB formation in two independent mouse models of Shh-activated MB. Furthermore, Wip1 knockdown or treatment with a WIP1 inhibitor suppressed the effects of Shh stimulation and potentiated the growth inhibitory effects of SHH pathway-inhibiting drugs in Shh-activated MB cells in vitro. This suggests an important cross-talk between SHH and WIP1 pathways that accelerates tumorigenesis and supports WIP1 inhibition as a potential treatment strategy for MB.


Subject(s)
Cerebellar Neoplasms/metabolism , Hedgehog Proteins/metabolism , Medulloblastoma/metabolism , Neural Stem Cells/metabolism , Protein Phosphatase 2C/metabolism , Signal Transduction , Animals , Biomarkers , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Gene Knockdown Techniques , Humans , Mice , Mice, Transgenic , NIH 3T3 Cells , Protein Phosphatase 2C/genetics , Tumor Suppressor Protein p53/metabolism
2.
Curr Med Chem ; 21(4): 501-10, 2014.
Article in English | MEDLINE | ID: mdl-24251561

ABSTRACT

Intrinsic or acquired resistance to the HER2-targeted therapy trastuzumab is a clinical concern in the treatment of patients with HER2-over-expressing metastatic breast cancers. We demonstrate here that multiple models of intrinsic and acquired resistance exhibit increased phosphorylation of p38 MAPK. Kinase inhibition of p38 rescued trastuzumab sensitivity in cells with acquired resistance. In addition, knockdown of p38 increased sensitivity to trastuzumab in an intrinsically resistant cell line. We previously reported that expression of growth differentiation factor 15 (GDF15) is increased in trastuzumab-resistant HER2-overexpressing breast cancer cells. In this study, we found that exogenous GDF15 or stable overexpression of GDF15 stimulated p38 phosphorylation in HER2-positive cells, suggesting a possible mechanism by which p38 is activated in resistant cells.GDF15 stable clones showed significantly increased invasiveness, which was rescued by p38 kinase inhibition, suggesting that p38 plays a role in the pro-invasive phenotype conferred by GDF15. Importantly, immunohistochemical analysis of a breast tumor tissue array indicated a significant (p=0.0053) correlation between HER2 and phosphorylated p38 specifically in GDF15-positive tissues. Our results suggest that p38 signaling drives trastuzumab resistance and invasiveness in HER2-overexpressing breast cancer. Upstream growth factor signals that have previously been implicated in trastuzumab resistance, such as GDF15, may contribute to the increased phosphorylation of p38 found in resistant cells.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Receptor, ErbB-2/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Immunohistochemistry , Phosphorylation , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/metabolism , Trastuzumab , p38 Mitogen-Activated Protein Kinases/genetics
3.
Curr Med Chem ; 20(19): 2486-99, 2013.
Article in English | MEDLINE | ID: mdl-23531216

ABSTRACT

The standard targeted therapy for HER2-overexpressing breast cancer is the HER2 monoclonal antibody, trastuzumab. Although effective, many patients eventually develop trastuzumab resistance. The dual EGFR/HER2 small molecule tyrosine kinase inhibitor lapatinib is approved for use in trastuzumab-refractory metastatic HER2-positive breast cancer. However, lapatinib resistance is a problem as most patients with trastuzumab-refractory disease do not benefit from lapatinib. Understanding the mechanisms underlying lapatinib resistance may ultimately facilitate development of new therapeutic strategies for HER2-overexpressing breast cancer. Our current results indicate that MEK inhibition increases lapatinib-mediated cytotoxicity in resistant HER2-overexpressing breast cancer cells. We genetically and pharmacologically blocked MEK/ERK signaling and evaluated lapatinib response by trypan blue exclusion, anchorage-independent growth assays, flow cytometric cell cycle and apoptosis analysis, and in tumor xenografts. Combined MEK inhibition and lapatinib treatment reduced phosphorylated ERK more than single agent treatment. In addition, Western blots, immunofluorescence, and immunohistochemistry demonstrated that the combination of MEK inhibitor plus lapatinib reduced nuclear expression of the MEK/ERK downstream proto-oncogene FOXM1. Genetic knockdown of MEK was tested for the ability to increase lapatinib-mediated cell cycle arrest or apoptosis in JIMT-1 and MDA361 cells. Finally, xenograft studies demonstrated that combined pharmacological inhibition of MEK plus lapatinib suppressed tumor growth and reduced expression of FOXM1 in HER2-overexpressing breast cancers that are resistant to trastuzumab and lapatinib. Our results suggest that FoxM1 contributes to lapatinib resistance downstream of MEK signaling, and supports further study of pharmacological MEK inhibition to improve response to lapatinib in HER2-overexpressing trastuzumab-resistant breast cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast/drug effects , Forkhead Transcription Factors/metabolism , MAP Kinase Signaling System/drug effects , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Breast/metabolism , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Forkhead Box Protein M1 , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lapatinib , Mice , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Mas , Quinazolines/pharmacology
4.
Curr Med Chem ; 19(7): 1065-75, 2012.
Article in English | MEDLINE | ID: mdl-22229414

ABSTRACT

Approximately 20-30% of breast cancers show increased expression of the HER2 receptor tyrosine kinase. Trastuzumab (Herceptin) is a clinically approved anti-HER2 monoclonal antibody. Many patients with HER2-overexpressing metastatic breast cancer respond to trastuzumab; however, a subset display primary drug resistance. In addition, many patients who initially respond to trastuzumab ultimately develop disease progression. Multiple molecular mechanisms contributing to trastuzumab resistance have been proposed in the literature. These mechanisms include cross-signaling from related HER/erbB receptors and compensatory signaling from receptors outside of the HER/erbB family, including receptors for insulin-like growth factor-I, vascular endothelial growth factor, and transforming growth factor beta. The major downstream signaling pathway activated by HER2 cross-talk is PI3K/mTOR, and a potential integrator of receptor cross-talk is Src-focal adhesion kinase (FAK) signaling. PI3K, Src, and FAK have independently been implicated in trastuzumab resistance. In this review, we will discuss pharmacological inhibition of HER2 cross-talk as a strategy to treat trastuzumab-refractory HER2-overexpresssing breast cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Breast Neoplasms/drug therapy , Drug Design , Drug Resistance, Neoplasm/drug effects , Receptor Cross-Talk/drug effects , Receptor, ErbB-2/metabolism , Antineoplastic Agents/therapeutic use , Chemistry, Pharmaceutical , Dimerization , Female , Humans , Receptor, ErbB-3/metabolism , Trastuzumab
5.
Curr Pharmacogenomics Person Med ; 7(4): 263-274, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-20300449

ABSTRACT

HER2 is over-expressed in approximately 25% to 30% of human metastatic breast cancers, primarily due to gene amplification. There are currently two HER2-targeted therapies approved for clinical use, the monoclonal HER2 antibody trastuzumab and the EGFR/HER2 dual tyrosine kinase inhibitor lapatinib. Although both agents show clinical benefit in a subset of patients with metastatic breast cancer, many patients with HER2-over-expressing metastatic breast tumors do not respond to these agents. Furthermore, those who do show an initial response generally demonstrate disease progression, on average in less than one year. It has become clear that HER2 expression status alone does not adequately predict response to HER2-targeted therapy. Identification and clinical validation of molecular predictors of response to trastuzumab and lapatinib is critical for further personalizing treatment and improving clinical benefit for patients whose tumors over-express HER2. In this review, we discuss published data describing potential predictors of response or resistance to trastuzumab and lapatinib. While a discussion of the preclinical work is provided, the emphasis is placed on potential predictors that have been studied in clinical specimens such as tumor tissue or serum obtained from patients treated with HER2-targeted therapy. The present analysis and synthesis of the available literature therefore contribute towards an emerging knowledgebase to personalize breast cancer treatment taking into factors including but beyond HER2 expression.

6.
Ann Oncol ; 19(2): 315-20, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17804473

ABSTRACT

BACKGROUND: AKT phosphorylation is a critical step in the activation of growth factor receptors and can mediate tumor resistance to anthracyclines. We evaluated the expression patterns and predictive value of phosphorylated AKT (pAKT) in breast cancer tissues. PATIENTS AND METHODS: pAKT expression was assessed by immunohistochemistry in 823 tumors from patients with early breast cancer enrolled in two randomized trials. The distribution of pAKT expression was correlated with HER2 and epidermal growth factor receptor (EGFR) expression. The predictive value of pAKT for the efficacy of adjuvant chemotherapy was determined by test for interaction. RESULTS: pAKT, EGFR, and HER2 were expressed in 119 of 781 (15%), 118 of 758 (16%), and 99 of 775 (13%) assessable tumors. Staining was positive for pAKT in 28 of 99 (28%) and 90 of 676 (13%) HER2+ and HER2- tumors (P < 0.001). pAKT was expressed in 15 of 94 (16%) and 75 of 563 (13%) HER2-/EGFR+ and HER2-/EGFR- tumors, respectively (P = 0.49). A positive staining for pAKT did not correlate with prognosis (P = 0.94), and did not predict the resistance to anthracyclines (test for interaction, P = 0.70). CONCLUSIONS: AKT phosphorylation is associated with HER2 expression but not EGFR expression in patients with early breast cancer. pAKT is not predictive for the efficacy of anthracycline-based adjuvant chemotherapy.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Adult , Age Factors , Aged , Breast Neoplasms/therapy , Chi-Square Distribution , Cohort Studies , Combined Modality Therapy , Disease-Free Survival , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Middle Aged , Neoplasm Staging , Predictive Value of Tests , Probability , Proportional Hazards Models , Proto-Oncogene Proteins c-akt/genetics , Randomized Controlled Trials as Topic , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Risk Assessment , Survival Analysis , Time Factors , Treatment Outcome
7.
Oncogene ; 26(25): 3637-43, 2007 May 28.
Article in English | MEDLINE | ID: mdl-17530017

ABSTRACT

The human epidermal growth factor receptor 2 (HER2) tyrosine kinase receptor is overexpressed in approximately 20-30% of human breast cancers, and is associated with reduced survival. Hence, numerous therapeutic strategies have been tested for their ability to target the HER2 protein. The humanized monoclonal antibody trastuzumab (Herceptin) was the first HER2-targeted agent approved for clinical use in breast cancer patients. Response rates to single-agent trastuzumab range from 12 to 34% for metastatic breast cancer (MBC), and significant improvements in survival rates are achieved in patients with early-stage HER2-overexpressing breast cancer in the adjuvant setting. Despite its initial efficacy, acquired resistance to trastuzumab develops in a majority of patients with MBC, and a large subset never responds, demonstrating primary resistance. Molecular mechanisms of trastuzumab antineoplastic activity and potential mechanisms contributing to its resistance will be discussed in this review. Novel agents that may enhance trastuzumab efficacy will also be discussed.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Drug Resistance, Neoplasm , Humans , Immunotherapy , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Signal Transduction , Trastuzumab
8.
Cancer Chemother Pharmacol ; 43(1): 73-9, 1999.
Article in English | MEDLINE | ID: mdl-9923544

ABSTRACT

PURPOSE: We have previously reported preferential repair of DNA interstrand crosslinks in the 4-hydroperoxycyclophosphamide-resistant human medulloblastoma cell line D-283 Med (4-HCR). We now report further studies that explored the potential mechanisms underlying this repair. METHODS: Limiting dilution assays and Western, Southern, and Northern blots were used to compare specific differences between D-283 Med (4-HCR) and its parental line D-283 Med. RESULTS: D-283 Med (4-HCR) was cross-resistant to melphalan and 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU), with O6-alkylguanine-DNA alkyltransferase (AGT) levels of 466+/-164 fmol/mg protein; AGT levels in the parental line, D-283 Med, were 76+/-96 fmol/mg. The increase in AGT activity was not a result of gene amplification. Depleting AGT with O6-benzylguanine partially restored sensitivity to BCNU. Both cell lines were deficient in the human mismatch protein MutLalpha. ERCC4 mRNA and poly(ADP-ribose) polymerase levels were similar in both cell lines, and ERCC1 mRNA levels were 2- to 2.5-fold lower in D-283 Med (4-HCR). Topoisomerase I levels were 2- to 2.5-fold higher in D-283 Med compared with D-283 Med (4-HCR). CONCLUSION: These results, while illustrating the multiple differences between D-283 Med and D-283 Med (4-HCR), do not explain the enhanced DNA interstrand crosslink repair seen in D-283 Med (4-HCR).


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Cerebellar Neoplasms/pathology , DNA Repair/drug effects , DNA, Neoplasm , Endonucleases , Medulloblastoma/pathology , Blotting, Northern , Blotting, Southern , Blotting, Western , Carmustine/pharmacology , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/biosynthesis , Drug Resistance, Neoplasm , Humans , Indicator Dilution Techniques , O(6)-Methylguanine-DNA Methyltransferase/analysis , Poly(ADP-ribose) Polymerases/biosynthesis , Protein Biosynthesis , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...