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 ; 78(1): 246-255, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29180468

ABSTRACT

Activation of the receptor tyrosine kinase Axl is associated with poor outcomes in pancreatic cancer (PDAC), where it coordinately mediates immune evasion and drug resistance. Here, we demonstrate that the selective Axl kinase inhibitor BGB324 targets the tumor-immune interface to blunt the aggressive traits of PDAC cells in vitro and enhance gemcitibine efficacy in vivo Axl signaling stimulates the TBK1-NFκB pathway and innate immune suppression in the tumor microenvironment. In tumor cells, BGB324 treatment drove epithelial differentiation, expression of nucleoside transporters affecting gemcitabine response, and an immune stimulatory microenvironment. Our results establish a preclinical mechanistic rationale for the clinical development of Axl inhibitors to improve the treatment of PDAC patients.Significance: These results establish a preclinical mechanistic rationale for the clinical development of AXL inhibitors to improve the treatment of PDAC patients. Cancer Res; 78(1); 246-55. ©2017 AACR.


Subject(s)
Benzocycloheptenes/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Triazoles/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzocycloheptenes/administration & dosage , Carcinoma, Pancreatic Ductal/immunology , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Female , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Pancreatic Neoplasms/immunology , Triazoles/administration & dosage , Xenograft Model Antitumor Assays , Gemcitabine , Axl Receptor Tyrosine Kinase
2.
J Biol Chem ; 291(42): 22244-22252, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27531748

ABSTRACT

The deposition of extracellular matrix (ECM) is a defining feature of pancreatic ductal adenocarcinoma (PDA), where ECM signaling can promote cancer cell survival and epithelial plasticity programs. However, ECM signaling can also limit PDA tumor growth by producing cytotoxic levels of reactive oxygen species. For example, excess fibronectin stimulation of α5ß1 integrin on stromal cells in PDA results in reduced angiogenesis and increased tumor cell apoptosis because of oxidative stress. Fibulin-5 (Fbln5) is a matricellular protein that blocks fibronectin-integrin interaction and thus directly limits ECM-driven reactive oxygen species production and supports PDA progression. Compared with normal pancreatic tissue, Fbln5 is expressed abundantly in the stroma of PDA; however, the mechanisms underlying the stimulation of Fbln5 expression in PDA are undefined. Using in vitro and in vivo approaches, we report that hypoxia triggers Fbln5 expression in a TGF-ß- and PI3K-dependent manner. Pharmacologic inhibition of TGF-ß receptor, PI3K, or protein kinase B (AKT) was found to block hypoxia-induced Fbln5 expression in mouse embryonic fibroblasts and 3T3 fibroblasts. Moreover, tumor-associated fibroblasts from mouse PDA were also responsive to TGF-ß receptor and PI3K/AKT inhibition with regard to suppression of Fbln5. In genetically engineered mouse models of PDA, therapy-induced hypoxia elevated Fbln5 expression, whereas pharmacologic inhibition of TGF-ß signaling reduced Fbln5 expression. These findings offer insight into the signaling axis that induces Fbln5 expression in PDA and a potential strategy to block its production.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Extracellular Matrix Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Neoplasms, Experimental/metabolism , Pancreatic Neoplasms/metabolism , Recombinant Proteins/biosynthesis , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Cell Hypoxia , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Integrin alpha5beta1/metabolism , Mice , Mice, Mutant Strains , NIH 3T3 Cells , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
3.
Cancer Lett ; 381(1): 252-8, 2016 10 10.
Article in English | MEDLINE | ID: mdl-26742464

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is a highly metastatic disease that resists most current therapies. A defining characteristic of PDA is an intense fibrotic response that promotes tumor cell invasion and chemoresistance. Efforts to understand the complex relationship between the tumor and its extracellular network and to therapeutically perturb tumor-stroma interactions are ongoing. Fibronectin (FN), a provisional matrix protein abundant in PDA stroma but not normal tissues, supports metastatic spread and chemoresistance of this deadly disease. FN also supports angiogenesis, which is required for even hypovascular tumors such as PDA to develop and progress. Targeting components of the tumor stroma, such as FN, can effectively reduce tumor growth and spread while also enhancing delivery of chemotherapy. Here, we review the molecular mechanisms by which FN drives angiogenesis, metastasis and chemoresistance in PDA. In light of these new findings, we also discuss therapeutic strategies to inhibit FN signaling.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Extracellular Matrix/metabolism , Fibronectins/metabolism , Pancreatic Neoplasms/metabolism , Signal Transduction , Tumor Microenvironment , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/secondary , Cell Movement , Drug Resistance, Neoplasm , Extracellular Matrix/drug effects , Extracellular Matrix/pathology , Fibrosis , Humans , Molecular Targeted Therapy , Neoplasm Invasiveness , Neovascularization, Pathologic , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Signal Transduction/drug effects
4.
Cancer Res ; 75(23): 5058-69, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26577699

ABSTRACT

Elevated oxidative stress is an aberration seen in many solid tumors, and exploiting this biochemical difference has the potential to enhance the efficacy of anticancer agents. Homeostasis of reactive oxygen species (ROS) is important for normal cell function, but excessive production of ROS can result in cellular toxicity, and therefore ROS levels must be balanced finely. Here, we highlight the relationship between the extracellular matrix and ROS production by reporting a novel function of the matricellular protein Fibulin-5 (Fbln5). We used genetically engineered mouse models of pancreatic ductal adenocarcinoma (PDAC) and found that mutation of the integrin-binding domain of Fbln5 led to decreased tumor growth, increased survival, and enhanced chemoresponse to standard PDAC therapies. Through mechanistic investigations, we found that improved survival was due to increased levels of oxidative stress in Fbln5-mutant tumors. Furthermore, loss of the Fbln5-integrin interaction augmented fibronectin signaling, driving integrin-induced ROS production in a 5-lipooxygenase-dependent manner. These data indicate that Fbln5 promotes PDAC progression by functioning as a molecular rheostat that modulates cell-ECM interactions to reduce ROS production, and thus tip the balance in favor of tumor cell survival and treatment-refractory disease.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Extracellular Matrix Proteins/metabolism , Pancreatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Extracellular Matrix Proteins/biosynthesis , Humans , Mice , Mice, Transgenic , Oxidative Stress/physiology , Pancreatic Neoplasms/pathology , Recombinant Proteins/biosynthesis , Tumor Microenvironment/physiology
5.
Cancer Res ; 74(18): 4996-5007, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25060520

ABSTRACT

Elevated levels of TGFß are a negative prognostic indicator for patients diagnosed with pancreatic cancer; as a result, the TGFß pathway is an attractive target for therapy. However, clinical application of pharmacologic inhibition of TGFß remains challenging because TGFß has tumor suppressor functions in many epithelial malignancies, including pancreatic cancer. In fact, direct neutralization of TGFß promotes tumor progression of genetic murine models of pancreatic cancer. Here, we report that neutralizing the activity of murine TGFß receptor 2 using a monoclonal antibody (2G8) has potent antimetastatic activity in orthotopic human tumor xenografts, syngeneic tumors, and a genetic model of pancreatic cancer. 2G8 reduced activated fibroblasts, collagen deposition, microvessel density, and vascular function. These stromal-specific changes resulted in tumor cell epithelial differentiation and a potent reduction in metastases. We conclude that TGFß signaling within stromal cells participates directly in tumor cell phenotype and pancreatic cancer progression. Thus, strategies that inhibit TGFß-dependent effector functions of stromal cells could be efficacious for the therapy of pancreatic tumors. Cancer Res; 74(18); 4996-5007. ©2014 AACR.


Subject(s)
Antibodies, Monoclonal/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Animals , Antimetabolites, Antineoplastic/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Humans , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Neoplasm Metastasis , Pancreatic Neoplasms/metabolism , Random Allocation , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...