Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
EMBO Mol Med ; 10(12)2018 12.
Article in English | MEDLINE | ID: mdl-30467127

ABSTRACT

PARP-1 holds major functions on chromatin, DNA damage repair and transcriptional regulation, both of which are relevant in the context of cancer. Here, unbiased transcriptional profiling revealed the downstream transcriptional profile of PARP-1 enzymatic activity. Further investigation of the PARP-1-regulated transcriptome and secondary strategies for assessing PARP-1 activity in patient tissues revealed that PARP-1 activity was unexpectedly enriched as a function of disease progression and was associated with poor outcome independent of DNA double-strand breaks, suggesting that enhanced PARP-1 activity may promote aggressive phenotypes. Mechanistic investigation revealed that active PARP-1 served to enhance E2F1 transcription factor activity, and specifically promoted E2F1-mediated induction of DNA repair factors involved in homologous recombination (HR). Conversely, PARP-1 inhibition reduced HR factor availability and thus acted to induce or enhance "BRCA-ness". These observations bring new understanding of PARP-1 function in cancer and have significant ramifications on predicting PARP-1 inhibitor function in the clinical setting.


Subject(s)
DNA Repair , E2F1 Transcription Factor/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Prostatic Neoplasms/pathology , Animals , Cell Line , Disease Progression , Gene Expression Profiling , Homologous Recombination , Humans , Immunohistochemistry , Male , Mice, Inbred BALB C , Tissue Array Analysis
2.
Clin Cancer Res ; 22(1): 107-21, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26272062

ABSTRACT

PURPOSE: We performed parallel investigations in cabozantinib-treated patients in a phase II trial and simultaneously in patient-derived xenograft (PDX) models to better understand the roles of MET and VEGFR2 as targets for prostate cancer therapy. EXPERIMENTAL DESIGN: In the clinical trial, radiographic imaging and serum markers were examined, as well as molecular markers in tumors from bone biopsies. In mice harboring PDX intrafemurally or subcutaneously, cabozantinib effects on tumor growth, MET, PDX in which MET was silenced, VEGFR2, bone turnover, angiogenesis, and resistance were examined. RESULTS: In responsive patients and PDX, islets of viable pMET-positive tumor cells persisted, which rapidly regrew after drug withdrawal. Knockdown of MET in PDX did not affect tumor growth in mice nor did it affect cabozantinib-induced growth inhibition but did lead to induction of FGFR1. Inhibition of VEGFR2 and MET in endothelial cells reduced the vasculature, leading to necrosis. However, each islet of viable cells surrounded a VEGFR2-negative vessel. Reduction of bone turnover was observed in both cohorts. CONCLUSIONS: Our studies demonstrate that MET in tumor cells is not a persistent therapeutic target for metastatic castrate-resistant prostate cancer (CRPC), but inhibition of VEGFR2 and MET in endothelial cells and direct effects on osteoblasts are responsible for cabozantinib-induced tumor inhibition. However, vascular heterogeneity represents one source of primary therapy resistance, whereas induction of FGFR1 in tumor cells suggests a potential mechanism of acquired resistance. Thus, integrated cross-species investigations demonstrate the power of combining preclinical models with clinical trials to understand mechanisms of activity and resistance of investigational agents.


Subject(s)
Anilides/pharmacology , Antineoplastic Agents/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Pyridines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Anilides/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Bone Neoplasms/diagnosis , Bone Neoplasms/secondary , Cell Line, Tumor , Clinical Trials, Phase II as Topic , Disease Models, Animal , Drug Resistance, Neoplasm , Gene Knockdown Techniques , Humans , Male , Mice , Multicenter Studies as Topic , Neoplasm Staging , Phosphorylation , Positron-Emission Tomography , Prostatic Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridines/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction/drug effects , Treatment Outcome , Tumor Burden/drug effects , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Xenograft Model Antitumor Assays
3.
Oncotarget ; 6(30): 29161-77, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26313360

ABSTRACT

While several new therapies are FDA-approved for bone-metastatic prostate cancer (PCa), patient survival has only improved marginally. Here, we report that chitosan nanoparticle-mediated delivery of miR-34a, a tumor suppressive microRNA that downregulates multiple gene products involved in PCa progression and metastasis, inhibited prostate tumor growth and preserved bone integrity in a xenograft model representative of established PCa bone metastasis. Expression of miR-34a induced apoptosis in PCa cells, and, in accord with downregulation of targets associated with PCa growth, including MET and Axl and c-Myc, also induced a form of non-canonical autophagy that is independent of Beclin-1, ATG4, ATG5 and ATG7. MiR-34a-induced autophagy is anti-proliferative in prostate cancer cells, as blocking apoptosis still resulted in growth inhibition of tumor cells. Thus, combined effects of autophagy and apoptosis are responsible for miR-34a-mediated prostate tumor growth inhibition, and have translational impact, as this non-canonical form of autophagy is tumor inhibitory. Together, these results provide a new understanding of the biological effects of miR-34a and highlight the clinical potential for miR-34a delivery as a treatment for bone metastatic prostate cancer.


Subject(s)
Autophagy , Bone Neoplasms/prevention & control , Chitosan/chemistry , Gene Transfer Techniques , Genetic Therapy/methods , MicroRNAs/genetics , Nanoparticles , Prostatic Neoplasms/therapy , Animals , Apoptosis , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Nude , MicroRNAs/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA Interference , Signal Transduction , Time Factors , Transfection , Tumor Burden , X-Ray Microtomography , Xenograft Model Antitumor Assays
4.
Sci Rep ; 3: 2687, 2013.
Article in English | MEDLINE | ID: mdl-24045437

ABSTRACT

The epithelial-mesenchymal transition (EMT) imparts metastatic competence on otherwise non-metastatic cancer cells through decreased inter-cellular adhesions, increased migratory capacity, stem cell properties and anoikis and chemotherapy resistance. In this study, we profiled changes in microRNA expression during EMT in conjunction with changes in DNA methylation at microRNA promoters to discover essential mediators of EMT-imparted stemness properties. MicroRNA-203 (miR-203) expression is repressed following EMT induced by multiple different stimuli and in established claudin-low cell lines as well as the CD44hi/CD24lo stem cell-enriched fraction. Expression of miR-203 in mesenchymal cells compromises migratory and invasive capacity in vitro, and tumor initiation and metastasis in vivo. Unexpectedly, miR-203 expression affects the sphere-forming capacity of neighboring cells by indirectly enhancing expression of DKK1, a secreted inhibitor of Wnt signaling and stemness resulting in suppression of ß-catenin protein levels. Our data suggest that restoring miR-203 expression levels may inhibit metastasis and combat deregulated Wnt signaling.


Subject(s)
Epigenesis, Genetic , Epithelial-Mesenchymal Transition/genetics , Gene Silencing , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Animals , Cell Differentiation , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , CpG Islands , DNA Methylation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mice , MicroRNAs/metabolism , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Paracrine Communication , Promoter Regions, Genetic , beta Catenin/metabolism
5.
Int J Cancer ; 133(7): 1536-46, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23526299

ABSTRACT

The receptor tyrosine kinase, MET, has been implicated in tumorigenesis and metastasis of many solid tumors, by multiple mechanisms, including cross talk with epidermal growth factor receptor. In this study, we examined the role of insulin-like growth factor receptor-1 (IGF-1R) signaling in MET activation, focusing on prostate cancer cells. Stimulation of the prostate cancer cell line PC3 with IGF-1 induces a delayed phosphorylation of MET at multiple sites (indicative of full activation), reaching a maximum 18 hr after IGF-1 addition. MET activation does not require the sole MET ligand hepatocyte growth factor (HGF), but does require transcription to occur. Furthermore, direct injection of IGF-1 is sufficient to induce MET activation in vivo, in a PC3 xenograft model. Pharmacologic or genetic inhibition of the tyrosine kinase, Src, abolishes MET phosphorylation, and expression of activated Src is sufficient to induce Met phosphorylation in the absence of IGF-1 stimulation. Activated MET is essential for IGF-1-mediated increased migration of PC3 cells, demonstrating an important biologic effect of IGF-1-mediated MET activation. Finally, we demonstrate that IGF-1-induced delayed MET activation occurs in multiple cell lines which express both the receptors, suggesting that IGF-1R-mediated MET activation may contribute to tumorigenic properties of multiple cancer types when both growth factor receptors are expressed. The results further suggest that MET may be activated by multiple receptor tyrosine kinase receptors, and dual targeting of these receptors may be important therapeutically.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptor, IGF Type 1/metabolism , src-Family Kinases/metabolism , Animals , Cell Line, Tumor , Cell Movement , Enzyme Activation , Hepatocyte Growth Factor/metabolism , Humans , Insulin-Like Growth Factor I/pharmacology , Integrins/metabolism , Ligands , Male , Mice , Neoplasm Transplantation , Phosphorylation , RNA Interference , RNA, Small Interfering , Receptor, IGF Type 1/genetics , Signal Transduction/drug effects , Transcription, Genetic , Xenograft Model Antitumor Assays , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
6.
Expert Opin Investig Drugs ; 20(12): 1677-84, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22035268

ABSTRACT

INTRODUCTION: An increasing number of basic, translational and clinical studies demonstrate the importance of the protein tyrosine kinase receptor, c-Met, in the progression of prostate cancer. c-Met is overexpressed in primary prostate cancers, further increased in expression in bone metastases and is associated with the development of castrate-resistant disease. Because of its importance as a target, c-Met inhibitors have reached clinical trial for advanced, castrate-resistant prostate cancer. AREAS COVERED: In this review, altered expression of c-Met and hepatocyte growth factor in prostate tumors and the microenvironment and how they contribute to growth and invasion of prostate cancer cells is described. Next, preclinical studies providing the support for use of c-Met inhibitors are discussed. Finally, early promising results from c-Met inhibitors in clinical trial, and future prospects for c-Met inhibitors in the treatment of advanced stage prostate cancer, are discussed. EXPERT OPINION: An emerging theme in treating metastatic prostate cancer is the requirement to target both the epithelial and stromal compartments. Results from clinical trials suggest that inhibitors of c-Met that block stromal-mediated c-Met activation in prostate tumors may be important therapeutic agents in at least a subset of patients with metastatic prostate cancer. However, as many of the inhibitors have multiple targets, the efficacy of targeting c-Met alone remains to be determined.


Subject(s)
Hepatocyte Growth Factor/metabolism , Molecular Targeted Therapy , Prostate/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-met/metabolism , Clinical Trials as Topic , Disease Progression , Drug Design , Hepatocyte Growth Factor/pharmacology , Humans , Male , Prostate/pathology , Prostate/physiopathology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/physiopathology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...