Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Mol Carcinog ; 56(3): 1189-1196, 2017 03.
Article in English | MEDLINE | ID: mdl-27649287

ABSTRACT

Ephrin B2 is variably expressed on tumor cells and its blockade has been shown to inhibit angiogenesis in animal models of pancreatic, colorectal, lung and head, and neck squamous cell carcinomas. However, the implications of ephrinB2 expression in cancer patients have remained elusive. In this study, we analyzed the cancer genome atlas (TCGA) for ephrinB2 expression. We report significant correlations between EFNB2 expression, overall survival and disease-free survival in head and neck squamous cell carcinoma (HNSCC, n = 519), pancreatic adenocarcinoma (n = 186), and bladder urothelial carcinoma (n = 410). In HNSCC patients, high-EFNB2 mRNA expression was associated with tumor HPV negativity, oral cavity location, alcohol intake, higher TP53 mutation, and EGFR amplification. EphrinB2 overexpression also correlated with worse response to chemotherapy and radiotherapy. The therapeutic potential of blocking ephrinB2 was validated in HNSCC patient-derived tumor xenografts and showed significant improvement in survival and tumor growth delay. Our data shows that ephrinB2 overexpression can serve as a critical biomarker for patient prognosis and response to therapy. These results should guide design of future clinical trials exploring EphrinB2 inhibition in cancer patients. © 2016 Wiley Periodicals, Inc.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Squamous Cell/drug therapy , Ephrin-B2/genetics , Head and Neck Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Up-Regulation , Urinary Bladder Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Ephrin-B2/antagonists & inhibitors , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Male , Mice , Middle Aged , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Prognosis , Squamous Cell Carcinoma of Head and Neck , Survival Analysis , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
2.
Int J Cancer ; 138(1): 195-205, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26152787

ABSTRACT

Dysregulation of the Notch1 receptor has been shown to facilitate the development and progression of colorectal cancer (CRC) and has been identified as an independent predictor of disease progression and worse survival. Although mutations in the NOTCH1 receptor have not been described in CRC, we have previously discovered a NOTCH1 gene copy number gain in a portion of CRC tumor samples. Here, we demonstrated that a NOTCH1 gene copy number gain is significantly associated with worse survival and a high percentage of gene duplication in a cohort of patients with advanced CRC. In our CRC patient-derived tumor xenograft (PDTX) model, tumors harboring a NOTCH1 gain exhibited significant elevation of the Notch1 receptor, JAG1 ligand and cleaved Notch1 activity. In addition, a significant association was identified between a gain in NOTCH1 gene copy number and sensitivity to a Notch1-targeting antibody. These findings suggest that patients with metastatic CRC that harbor a gain in NOTCH1 gene copy number have worse survival and that targeting this patient population with a Notch1 antibody may yield improved outcomes.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , DNA Copy Number Variations , Gene Dosage , Receptor, Notch1/genetics , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers, Tumor , Calcium-Binding Proteins/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Female , Gene Duplication , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Male , Membrane Proteins/metabolism , Mice , Neoplasm Metastasis , Prognosis , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/metabolism , Serrate-Jagged Proteins , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
3.
Int J Cancer ; 136(8): 1967-75, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25242168

ABSTRACT

Antiangiogenic therapy is commonly used for the treatment of colorectal cancer (CRC). Although patients derive some clinical benefit, treatment resistance inevitably occurs. The MET signaling pathway has been proposed to be a major contributor of resistance to antiangiogenic therapy. MET is upregulated in response to vascular endothelial growth factor pathway inhibition and plays an essential role in tumorigenesis and progression of tumors. In this study, we set out to determine the efficacy of cabozantinib in a preclinical CRC patient-derived tumor xenograft model. We demonstrate potent inhibitory effects on tumor growth in 80% of tumors treated. The greatest antitumor effects were observed in tumors that possess a mutation in the PIK3CA gene. The underlying antitumor mechanisms of cabozantinib consisted of inhibition of angiogenesis and Akt activation and significantly decreased expression of genes involved in the PI3K pathway. These findings support further evaluation of cabozantinib in patients with CRC. PIK3CA mutation as a predictive biomarker of sensitivity is intriguing and warrants further elucidation. A clinical trial of cabozantinib in refractory metastatic CRC is being activated.


Subject(s)
Anilides/pharmacology , Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Adult , Aged , Angiogenesis Inhibitors/pharmacology , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Colorectal Neoplasms/metabolism , Female , HCT116 Cells , Humans , Mice , Mice, Nude , Middle Aged , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays/methods
4.
Lancet Oncol ; 13(8): 773-81, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22805291

ABSTRACT

BACKGROUND: Inhibition of MEK stops cell proliferation and induces apoptosis; therefore, this enzyme is a key anticancer target. Trametinib is a selective, orally administered MEK1/MEK2 inhibitor. We aimed to define the maximum tolerated dose and recommended phase 2 dose of trametinib and to assess its safety, pharmacokinetics, pharmacodynamics, and response rate in individuals with advanced solid tumours. METHODS: We undertook a multicentre phase 1 study in patients with advanced solid tumours and adequate organ function. The study was in three parts: dose escalation to define the maximum tolerated dose; identification of the recommended phase 2 dose; and assessment of pharmacodynamic changes. Intermittent and continuous dosing regimens were analysed. Blood samples and tumour biopsy specimens were taken to assess pharmacokinetic and pharmacodynamic changes. Adverse events were defined with common toxicity criteria, and tumour response was measured by Response Evaluation Criteria In Solid Tumors. This study is registered with ClinicalTrials.gov, number NCT00687622. FINDINGS: We enrolled 206 patients (median age 58·5 years, range 19-92). Dose-limiting toxic effects included rash (n=2), diarrhoea (n=1), and central serous retinopathy (n=2). The most common treatment-related adverse events were rash or dermatitis acneiform (n=165; 80%) and diarrhoea (87; 42%), most of which were grade 1 and 2. The maximum tolerated dose was 3 mg once daily and the recommended phase 2 dose was 2 mg a day. The effective half-life of trametinib was about 4 days. At the recommended phase 2 dose, the exposure profile of the drug showed low interpatient variability and a small peak:trough ratio of 1·81. Furthermore, mean concentrations in plasma were greater than the preclinical target concentration throughout the dosing interval. Pathway inhibition and clinical activity were seen, with 21 (10%) objective responses recorded. INTERPRETATION: The recommended phase 2 dose of 2 mg trametinib once a day is tolerable, with manageable side-effects. Trametinib's inhibition of the expected target and clinical activity warrants its further development as a monotherapy and in combination. FUNDING: GlaxoSmithKline.


Subject(s)
Antineoplastic Agents/administration & dosage , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Biopsy , Drug Administration Schedule , Drug Monitoring , Female , Half-Life , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Male , Maximum Tolerated Dose , Metabolic Clearance Rate , Middle Aged , Molecular Targeted Therapy , Neoplasms/enzymology , Neoplasms/pathology , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Pyridones/adverse effects , Pyridones/pharmacokinetics , Pyrimidinones/adverse effects , Pyrimidinones/pharmacokinetics , Treatment Outcome , United States , Young Adult
5.
Clin Cancer Res ; 12(23): 7099-107, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17145834

ABSTRACT

PURPOSE: Standard treatments have modest effect against pancreatic cancer, and current research focuses on agents targeting molecular pathways involved in tumor growth and angiogenesis. This study investigated the interactions between ZD6474, an inhibitor of tyrosine kinase activities of vascular endothelial growth factor receptor-2 and epidermal growth factor receptor (EGFR), gemcitabine, and ionizing radiation in human pancreatic cancer cells and analyzed the molecular mechanisms underlying this combination. EXPERIMENTAL DESIGN: ZD6474, ionizing radiation, and gemcitabine, alone or in combination, were given in vitro to MIA PaCa-2, PANC-1, and Capan-1 cells and in vivo to MIA PaCa-2 tumor xenografts. The effects of treatments were studied by the evaluation of cytotoxicity, apoptosis, cell cycle, EGFR and Akt phosphorylation, modulation of gene expression of enzymes related to gemcitabine activity (deoxycytidine kinase and ribonucleotide reductase), as well as vascular endothelial growth factor immunohistochemistry and microvessel count. RESULTS: In vitro, ZD6474 dose dependently inhibited cell growth, induced apoptosis, and synergistically enhanced the cytotoxic activity of gemcitabine and ionizing radiation. Moreover, ZD6474 inhibited phosphorylation of EGFR and Akt and triggered cell apoptosis. PCR analysis showed that ZD6474 increased the ratio between gene expression of deoxycytidine kinase and ribonucleotide reductase. In vivo, ZD6474 showed significant antitumor activity alone and in combination with radiotherapy and gemcitabine, and the combination of all three modalities enhanced MIA PaCA-2 tumor growth inhibition compared with gemcitabine alone. CONCLUSIONS: ZD6474 decreases EGFR and Akt phosphorylation, enhances apoptosis, favorably modulates gene expression in cancer cells, and acts synergistically with gemcitabine and radiotherapy to inhibit tumor growth. These findings support the investigation of this combination in the clinical setting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxycytidine/analogs & derivatives , ErbB Receptors/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy , Piperidines/pharmacology , Quinazolines/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Combined Modality Therapy , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Deoxycytidine Kinase/genetics , Deoxycytidine Kinase/radiation effects , Disease Models, Animal , Dose-Response Relationship, Radiation , Drug Screening Assays, Antitumor , Drug Synergism , Female , Gene Expression Profiling , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/diagnosis , Phosphorylation , Piperidines/therapeutic use , Polymerase Chain Reaction , Predictive Value of Tests , Quinazolines/therapeutic use , Radiation, Ionizing , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction/drug effects , Transplantation, Heterologous , Gemcitabine
6.
J Clin Oncol ; 35(3): 352-360, 2017 Jan 20.
Article in English | MEDLINE | ID: mdl-27870570

ABSTRACT

Purpose Adenoid cystic carcinomas (ACCs) represent a heterogeneous group of chemotherapy refractory tumors, with a subset demonstrating an aggressive phenotype. We investigated the molecular underpinnings of this phenotype and assessed the Notch1 pathway as a potential therapeutic target. Methods We genotyped 102 ACCs that had available pathologic and clinical data. Notch1 activation was assessed by immunohistochemistry for Notch1 intracellular domain. Luciferase reporter assays were used to confirm Notch1 target gene expression in vitro. The Notch1 inhibitor brontictuzumab was tested in patient-derived xenografts from patients with ACC and in a patient with ACC who was enrolled in a phase I study. Results NOTCH1 mutations occurred predominantly (14 of 15 patients) in the negative regulatory region and Pro-Glu-Ser-Thr-rich domains, the same two hotspots seen in T-cell acute lymphoblastic leukemias, and led to pathway activation in vitro. NOTCH1-mutant tumors demonstrated significantly higher levels of Notch1 pathway activation than wild-type tumors on the basis of Notch1 intracellular domain staining ( P = .004). NOTCH1 mutations define a distinct aggressive ACC subgroup with a significantly higher likelihood of solid subtype ( P < .001), advanced-stage disease at diagnosis ( P = .02), higher rate of liver and bone metastasis ( P ≤ .02), shorter relapse-free survival (median, 13 v 34 months; P = .01), and shorter overall survival (median 30 v 122 months; P = .001) when compared with NOTCH1 wild-type tumors. Significant tumor growth inhibition with brontictuzumab was observed exclusively in the ACC patient-derived xenograft model that harbored a NOTCH1 activating mutation. Furthermore, an index patient with NOTCH1-mutant ACC had a partial response to brontictuzumab. Conclusion NOTCH1 mutations define a distinct disease phenotype characterized by solid histology, liver and bone metastasis, poor prognosis, and potential responsiveness to Notch1 inhibitors. Clinical studies targeting Notch1 in a genotype-defined ACC subgroup are warranted.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Carcinoma, Adenoid Cystic/genetics , Liver Neoplasms/genetics , Mutation , Receptor, Notch1/genetics , Salivary Gland Neoplasms/genetics , Adult , Aged , Animals , Bone Neoplasms/drug therapy , Bone Neoplasms/mortality , Bone Neoplasms/secondary , Carcinoma, Adenoid Cystic/drug therapy , Carcinoma, Adenoid Cystic/mortality , Carcinoma, Adenoid Cystic/secondary , DNA Mutational Analysis , Disease-Free Survival , Female , Genetic Predisposition to Disease , HEK293 Cells , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Liver Neoplasms/drug therapy , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Male , Mice, Nude , Middle Aged , Molecular Targeted Therapy , Phenotype , Predictive Value of Tests , Proportional Hazards Models , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/metabolism , Retrospective Studies , Risk Factors , Salivary Gland Neoplasms/drug therapy , Salivary Gland Neoplasms/mortality , Salivary Gland Neoplasms/pathology , Time Factors , Transfection , Treatment Outcome , Xenograft Model Antitumor Assays
7.
J Clin Oncol ; 31(9): 1231-8, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23358974

ABSTRACT

The ubiquitin proteasome system (UPS) regulates the ubiquitination, and thus degradation and turnover, of many proteins vital to cellular regulation and function. The UPS comprises a sequential series of enzymatic processes using four key enzyme families: E1 (ubiquitin-activating enzymes), E2 (ubiquitin-carrier proteins), E3 (ubiquitin-protein ligases), and E4 (ubiquitin chain assembly factors). Because the UPS is a crucial regulator of the cell cycle, and abnormal cell-cycle control can lead to oncogenesis, aberrancies within the UPS pathway can result in a malignant cellular phenotype and thus has become an attractive target for novel anticancer agents. This article will provide an overall review of the mechanics of the UPS, describe aberrancies leading to cancer, and give an overview of current drug therapies selectively targeting the UPS.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic , Proteasome Endopeptidase Complex/physiology , Ubiquitin-Activating Enzymes/physiology , Ubiquitin-Conjugating Enzymes/physiology , Ubiquitin-Protein Ligases/physiology , Antineoplastic Agents/therapeutic use , Cyclin-Dependent Kinase Inhibitor p27/physiology , DNA-Binding Proteins/physiology , Humans , Male , Mutation , Oncogene Proteins, Viral/physiology , Prostatic Neoplasms/genetics , Proteasome Endopeptidase Complex/drug effects , Proto-Oncogene Proteins c-mdm2/physiology , Tumor Suppressor Protein p53/physiology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics
8.
J Clin Oncol ; 27(33): 5660-9, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19858397

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer mortality, despite significant improvements in diagnostic imaging and operative mortality rates. The 5-year survival rate remains less than 5% because of microscopic or gross metastatic disease at time of diagnosis. The Clinical Trials Planning Meeting in pancreatic cancer was convened by the National Cancer Institute's Gastrointestinal Cancer Steering Committee to discuss the integration of basic and clinical knowledge in the design of clinical trials in PDAC. Major emphasis was placed on the enhancement of research to identify and validate the relevant targets and molecular pathways in PDAC, cancer stem cells, and the microenvironment. Emphasis was also placed on developing rational combinations of targeted agents and the development of predictive biomarkers to assist selection of patient subsets. The development of preclinical tumor models that are better predictive of human PDAC must be supported with wider availability to the research community. Phase III clinical trials should be implemented only if there is a meaningful clinical signal of efficacy and safety in the phase II setting. The emphasis must therefore be on performing well-designed phase II studies with uniform sets of basic entry and evaluation criteria with survival as a primary endpoint. Patients with either metastatic or locally advanced PDAC must be studied separately.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/therapy , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/therapy , Animals , Carcinoma, Pancreatic Ductal/diagnosis , Clinical Trials, Phase III as Topic , Combined Modality Therapy , Disease Models, Animal , Disease-Free Survival , Drug Delivery Systems , Female , Humans , Male , Mice , National Cancer Institute (U.S.) , Neoplasm Invasiveness , Neoplasm Staging , Pancreatectomy/methods , Pancreatic Neoplasms/diagnosis , Practice Guidelines as Topic , Prognosis , Risk Assessment , Survival Analysis , United States
SELECTION OF CITATIONS
SEARCH DETAIL