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.
Oncotarget ; 7(30): 48265-48279, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27374084

ABSTRACT

Current treatments for pancreatic ductal adenocarcinoma (PDA) are ineffective, making this the 4th leading cause of cancer deaths. Sunitinib is a broad-spectrum inhibitor of tyrosine kinase receptors mostly known for its anti-angiogenic effects. We tested the therapeutic effects of sunitinib in pancreatic cancer using the Ela-myc transgenic mouse model. We showed that Ela-myc pancreatic tumors express PDGFR and VEGFR in blood vessels and epithelial cells, rendering these tumors sensitive to sunitinib by more than only its anti-angiogenic activity. However, sunitinib treatment of Ela-myc mice with either early or advanced tumor progression had no impact on either survival or tumor burden. Further histopathological characterization of these tumors did not reveal differences in necrosis, cell differentiation, angiogenesis, apoptosis or proliferation. In stark contrast, in vitro sunitinib treatment of Ela-myc- derived cell lines showed high sensitivity to the drug, with increased apoptosis and reduced proliferation. Correspondingly, subcutaneous tumors generated from these cell lines completely regressed in vivo after sunitinib treatments. These data point at the pancreatic tumor microenvironment as the most likely barrier preventing sunitinib treatment efficiency in vivo. Combined treatments with drugs that disrupt tumor fibrosis may enhance sunitinib therapeutic effectiveness in pancreatic cancer treatment.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Indoles/pharmacology , Pancreatic Neoplasms/drug therapy , Pyrroles/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/blood supply , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Mice , Neovascularization, Pathologic/drug therapy , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Sunitinib
2.
Mol Cancer ; 14: 146, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26227809

ABSTRACT

BACKGROUND: Combined treatment of oncolytic adenoviruses with chemotherapeutic agents is foreseen as a therapeutic option for cancer. Here we have investigated the potential to use gemcitabine in combination with the oncolytic adenovirus AduPARE1A to treat pancreatic cancer and evaluate the underlying mechanism. METHODS: We treated pancreatic cancer cell lines BxPC-3 and PANC-1 with AduPARE1A and gemcitabine individually or in combination and analyzed cell viability, combination index, apoptosis and viral production. We also investigated the effects of the combination on tumor growth and mice survival in two xenograft models. Furthermore, we analyzed uPAR promoter activity from different uPAR-controlled adenovirus and studied NF-κB mediated effects. RESULTS: Synergistic cell killing from the combination AduPARE1A/Gemcitabine was observed in BxPC-3 and PANC-1 cells. Moreover, the combination treatment produced therapeutic benefits over either individual modality in two mouse models bearing orthotopic tumors, showing reduced tumor progression and significant prolonged mouse survival. Mechanistic studies showed that the synergistic cell death was not due to an increase in viral replication but occurred through an enhancement of apoptotic cell death. Gemcitabine stimulation increased the transcription of uPAR-controlled transgenes through the induction of NF-κB acting on the uPAR promoter. Interestingly, NF-κB gemcitabine-mediated induction of AduPAR adenoviruses interfered with the activation of NF-κB regulated genes, probably as a result of an intracellular competition for NF-κB DNA binding. Consequently, AduPARE1A infection sensitized cells to gemcitabine-induced apoptosis in the combined treatment. CONCLUSIONS: These data highlights the potential of the combination as a treatment modality for pancreatic cancer patients.


Subject(s)
Deoxycytidine/analogs & derivatives , NF-kappa B/metabolism , Oncolytic Virotherapy , Oncolytic Viruses , Pancreatic Neoplasms/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Adenoviridae , Animals , Apoptosis/drug effects , Cell Line, Tumor , Combined Modality Therapy , Deoxycytidine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Male , Mice , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Virus Replication/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
3.
J Control Release ; 192: 148-56, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25037019

ABSTRACT

Selective tumor targeting of oncolytic adenovirus at the level of cell entry remains a major challenge to improve efficacy and safety. Matrix metalloproteases (MMPs) are overexpressed in a variety of tumors and in particular in pancreatic cancer. In the current work, we have exploited the expression of MMPs together with the penetration capabilities of a TAT-like peptide to engineer tumor selective adenoviruses. We have generated adenoviruses containing CAR-binding ablated fibers further modified with a C-terminus TAT-like peptide linked to a blocking domain by an MMP-cleavable sequence. This linker resulted in a MMP-dependent cell transduction of the reporter MMP-activatable virus AdTATMMP and in efficient transduction of neoplastic cells and cancer-associated fibroblasts. Intravenous and intraductal administration of AdTATMMP into mice showed very low AdTATMMP activity in the normal pancreas, whereas increased transduction was observed in pancreatic tumors of transgenic Ela-myc mice. Intraductal administration of AdTATMMP into mice bearing orthotopic tumors led to a 25-fold increase in tumor targeting compared to the wild type fiber control. A replication competent adenovirus, Ad(RC)MMP, with the MMP-activatable fiber showed oncolytic efficacy and increased antitumor activity compared to Adwt in a pancreatic orthotopic model. Reduced local and distant metastases were observed in Ad(RC)MMP treated-mice. Moreover, no signs of pancreatic toxicity were detected. We conclude that MMP-activatable adenovirus may be beneficial for pancreatic cancer treatment.


Subject(s)
Adenoviridae/genetics , Matrix Metalloproteinases/genetics , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/therapy , Animals , Base Sequence , Cell Line, Tumor , Enzyme Activation , HEK293 Cells , Humans , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , NIH 3T3 Cells , Oncolytic Virotherapy/methods , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Transduction, Genetic
4.
Oncotarget ; 4(1): 94-105, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23328228

ABSTRACT

Gene-based anticancer therapies delivered by adenoviruses are limited by the poor viral distribution into the tumor. In the current work we have explored the feasibility of targeting pancreatic tumors through a loco-regional route. We have taken advantage of the ductal network in the pancreas to retrogradelly inject adenoviruses through the common bile duct in two different mouse models of pancreatic carcinogenesis: The transgenic Ela-myc mice that develop mixed neoplasms displaying both acinar-like and duct-like neoplastic cells affecting the whole pancreas; and mice bearing PANC-1 and BxPC-3 orthotopic xenografts that constitute a model of localized human neoplastic tumors. We studied tumor targeting and the anticancer effects of newly thymidine kinase-engineered adenoviruses both in vitro and in vivo, and conducted comparative studies between intraductal or intravenous administration. Our data indicate that the intraductal delivery of adenovirus efficiently targets pancreatic tumors in the two mouse models. The in vivo application of AduPARTKT plus ganciclovir (GCV) treatment induced tumor regression in Ela-myc mice. Moreover, the intraductal injection of ICOVIR15-TKT oncolytic adenoviruses significantly improved mean survival of mice bearing PANC-1 and BxPC-3 pancreatic xenografts from 30 to 52 days and from 20 to 68 days respectively (p less than 0.0001) when combined with GCV. Of notice, both AduPARTKT and ICOVIR15-TKT antitumoral responses were stronger by ductal viral application than intravenously, in line with the 38-fold increase in pancreas transduction observed upon ductal administration. In summary our data show that cytotoxic adenoviruses retrogradelly injected to the pancreas can be a feasible approach to treat localized pancreatic tumors.


Subject(s)
Ganciclovir/pharmacology , Genetic Therapy/methods , Pancreatic Neoplasms/therapy , Xenograft Model Antitumor Assays , Adenoviridae/genetics , Animals , Cell Line, Tumor , Common Bile Duct/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Kaplan-Meier Estimate , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Transgenic , Oncolytic Viruses/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Receptors, Urokinase Plasminogen Activator/genetics , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics
5.
Cell ; 151(7): 1595-607, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23260145

ABSTRACT

Most studies on TCF7L2 SNP variants in the pathogenesis of type 2 diabetes (T2D) focus on a role of the encoded transcription factor TCF4 in ß cells. Here, a mouse genetics approach shows that removal of TCF4 from ß cells does not affect their function, whereas manipulating TCF4 levels in the liver has major effects on metabolism. In Tcf7l2(-/-) mice, the immediate postnatal surge in liver metabolism does not occur. Consequently, pups die due to hypoglycemia. By combining chromatin immunoprecipitation with gene expression profiling, we identify a TCF4-controlled metabolic gene program that is acutely activated in the postnatal liver. In concordance, adult liver-specific Tcf7l2 knockout mice show reduced hepatic glucose production during fasting and display improved glucose homeostasis when maintained on high-fat diet. Furthermore, liver-specific TCF4 overexpression increases hepatic glucose production. These observations imply that TCF4 directly activates metabolic genes and that inhibition of Wnt signaling may be beneficial in metabolic disease.


Subject(s)
Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Glucose/metabolism , Liver/metabolism , Metabolic Networks and Pathways , Transcription Factor 7-Like 2 Protein/metabolism , Animals , Animals, Newborn , Diet, High-Fat , Fasting/metabolism , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Transcription Factor 7-Like 2 Protein/genetics , Transcriptional Activation
6.
Cancer Lett ; 317(1): 16-23, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22079741

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) therapies show limited success. Irreversible electroporation (IRE) is an innovative loco-regional therapy in which high-voltage pulses are applied to induce plasma membrane defects leading to cellular death. In the present study we evaluated the feasibility of IRE against PDAC. IRE treatment exhibited significant antitumor effects and prolonged survival in mice with orthotopic xenografts. Extensive tumor necrosis, reduced tumor cell proliferation and disruption of microvessels were observed at different days post-IRE. Animals had transient increases in transaminases, amylase and lipase enzymes that normalized at 24h post-IRE. These results suggest that IRE could be an effective treatment for locally advanced pancreatic tumors.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Electrochemotherapy , Pancreatic Neoplasms/therapy , Alanine Transaminase/blood , Amylases/blood , Animals , Aspartate Aminotransferases/blood , Biomarkers/blood , Blood Glucose/metabolism , Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/blood supply , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation , Electrochemotherapy/adverse effects , Feasibility Studies , Genes, Reporter , Humans , Lipase/blood , Luminescent Measurements , Male , Mice , Mice, Nude , Microvessels/pathology , Necrosis , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Time Factors , Transfection , Xenograft Model Antitumor Assays
7.
Cancers (Basel) ; 3(1): 368-95, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-24212620

ABSTRACT

The continuous identification of molecular changes deregulating critical pathways in pancreatic tumor cells provides us with a large number of novel candidates to engineer gene-targeted approaches for pancreatic cancer treatment. Targets-both protein coding and non-coding-are being exploited in gene therapy to influence the deregulated pathways to facilitate cytotoxicity, enhance the immune response or sensitize to current treatments. Delivery vehicles based on viral or non-viral systems as well as cellular vectors with tumor homing characteristics are a critical part of the design of gene therapy strategies. The different behavior of tumoral versus non-tumoral cells inspires vector engineering with the generation of tumor selective products that can prevent potential toxic-associated effects. In the current review, a detailed analysis of the different targets, the delivery vectors, the preclinical approaches and a descriptive update on the conducted clinical trials are presented. Moreover, future possibilities in pancreatic cancer treatment by gene therapy strategies are discussed.

8.
Neoplasia ; 11(6): 518-28, 4 p following 528, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19484141

ABSTRACT

Treatment options for pancreatic cancer have shown limited success mainly owing to poor selectivity for pancreatic tumor tissue and to a lack of activity in the tumor. In this study, we describe the ability of the urokinase-type plasminogen activator receptor (uPAR) promoter to efficiently and selectively target pancreatic tumors and metastases, which enables the successful management of pancreatic cancer. We have generated a replication-defective reporter adenovirus, AduPARLuc, and a conditionally replicating adenovirus, AduPARE1A, and we have studied the selectivity and antitumoral efficacy in pancreatic tumors and metastases. Toxicity was studied on intravascular delivery. We demonstrate that the uPAR promoter is highly active in pancreatic tumors but very weak in normal tissues. Tumor specificity is evidenced by a 100-fold increase in the tumor-to-liver ratio and by selective targeting of liver metastases (P < .001). Importantly, the AduPARE1A maintains the oncolytic activity of the wild-type virus, with reduced toxicity, and exhibits significant antitumoral activity (25% tumor eradication) and prolonged survival in pancreatic xenograft models (P < .0001). Furthermore, upon intravascular delivery, we demonstrate complete eradication of liver metastasis in 33% of mice, improving median survival (P = 5.43 x 10(-5)). The antitumoral selective activity of AduPARE1A shows the potential of uPAR promoter-based therapies in pancreatic cancer treatment.


Subject(s)
Adenoviridae/physiology , Liver Neoplasms, Experimental/therapy , Oncolytic Virotherapy/methods , Pancreatic Neoplasms/therapy , Receptors, Urokinase Plasminogen Activator/genetics , Adenoviridae/genetics , Adenoviridae/metabolism , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Cell Survival , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Liver/metabolism , Liver/pathology , Liver/virology , Liver Neoplasms, Experimental/secondary , Liver Neoplasms, Experimental/virology , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism , Oncolytic Viruses/physiology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/virology , Promoter Regions, Genetic/genetics , Tumor Burden , Xenograft Model Antitumor Assays
9.
FASEB J ; 23(5): 1366-75, 2009 May.
Article in English | MEDLINE | ID: mdl-19124560

ABSTRACT

Keratin 7 is expressed in simple epithelia but is expressed at low or undetectable levels in gastrointestinal epithelial cells. In the pancreas, it is present in ductal but not in acinar cells. K7 mRNA is overexpressed in pancreatic cancers. Here we use luciferase reporter assays to analyze the tissue-specific regulatory elements of murine keratin 7 (Krt7) promoter in vitro and in vivo. All elements required for appropriate cell and tissue specificity in reporter assays are present within the Krt7 -234 bp sequence. This fragment appears more selective to pancreatic ductal cells than the Krt19 promoter. GC-rich sequences corresponding to putative Sp1, AP-2 binding sites are essential for in vitro activity. Krt7-LacZ transgenic mice were generated to analyze in vivo activity. Sequences located 1.5 or 0.25 kb upstream of the transcription initiation site drive reporter expression to ductal, but not acinar, cells in transgenic mice. LacZ mRNA was detected in the pancreas as well as in additional epithelial tissues--such as the intestine and the lung--using both promoter constructs. An AdK7Luc adenovirus was generated to assess targeting selectivity in vivo by intravenous injection to immunocompetent mice and in a xenograft model of pancreatic cancer. The -0.25 kb region showed pancreatic selectivity, high activity in pancreatic cancers, and sustained transgene expression in xenografts. In conclusion, the krt7 promoter is useful to target pancreatic ductal adenocarcinoma cells in vitro and in vivo.


Subject(s)
Keratin-7/genetics , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/metabolism , Promoter Regions, Genetic , Transgenes , Adenoviridae/genetics , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Transgenic , Regulatory Sequences, Nucleic Acid/genetics , Sp1 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...