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.
PLoS One ; 8(8): e70594, 2013.
Article in English | MEDLINE | ID: mdl-23967078

ABSTRACT

Peritoneal carcinomatosis is common in advanced pancreatic cancer. Despite current standard treatment, patients with this disease until recently were considered incurable. Cancer gene therapy using oncolytic viruses have generated much interest over the past few years. Here, we investigated a new gene directed enzyme prodrug therapy (GDEPT) approach for an oncosuppressive virotherapy strategy using parvovirus H1 (PV-H1) which preferentially replicates and kills malignant cells. Although, PV-H1 is not potent enough to destroy tumors, it represents an attractive vector for cancer gene therapy. We therefore sought to determine whether the suicide gene/prodrug system, yCD/5-FC could be rationally combined to PV-H1 augmenting its intrinsic oncolytic activity for pancreatic cancer prevention and treatment. We showed that the engineered recombinant parvovirus rPVH1-yCD with 5-FC treatment increased significantly the intrinsic cytotoxic effect and resulted in potent induction of apoptosis and tumor growth inhibition in chemosensitive and chemoresistant cells. Additionally, the suicide gene-expressing PV-H1 infection reduced significantly the constitutive activities of NFκB and Akt/PI3K. Combination of their pharmacological inhibitors (MG132 and LY294002) with rPVH1-yCD/5-FC resulted in substantial increase of antitumor activity. In vivo, high and sustained expression of NS1 and yCD was observed in the disseminated tumor nodules and absent in normal tissues. Treatment of mice bearing intraperitoneal pancreatic carcinomatosis with rPVH1-yCD/5-FC resulted in a drastic inhibition of tumor cell spreading and subsequent increase in long-term survival. Together, the presented data show the improved oncolytic activity of wPV-H1 by yCD/5-FC and thus provides valuable effective and promising virotherapy strategy for prevention of tumor recurrence and treatment. In the light of this study, the suicide gene parvovirotherapy approach represents a new weapon in the war against pancreatic cancer. Moreover, these preliminary accomplishments are opening new field for future development of new combined targeted therapies to have a meaningful impact on advanced cancer.


Subject(s)
Cytosine Deaminase/genetics , Flucytosine/metabolism , Genes, Transgenic, Suicide/genetics , H-1 parvovirus/genetics , Oncolytic Virotherapy/methods , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cytosine Deaminase/metabolism , DNA, Recombinant/genetics , Female , H-1 parvovirus/physiology , Humans , Mice , NF-kappa B/metabolism , Neoadjuvant Therapy , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/virology , Phosphatidylinositol 3-Kinases/metabolism , Prodrugs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Yeasts/enzymology
2.
Nanomedicine ; 7(6): 841-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21419876

ABSTRACT

Development of chemoresistance and rapid inactivation of gemcitabine (Gem), the standard therapy for advanced pancreatic cancer, are responsible of the major therapeutic failures. To overcome the above drawbacks we designed a novel nanomedicine strategy for Gem nanoparticle (NP) formulation based on squalene conjugation. The purpose was to investigate the antitumor efficacy of gemcitabine-squalene (SQ-Gem) NPs on chemoresistant and chemosensitive pancreatic adenocarcinoma models. Cell viability and apoptosis assays showed that SQ-Gem NPs displayed higher antiproliferative and cytotoxic effects, particularly in chemoresistant Panc1 tumor cells. In in vivo studies, compared to native Gem, SQ-Gem NPs decreased significantly the tumor growth, prevented tumor cell invasion, and prolonged the survival time of mice bearing orthotopic pancreatic tumors. These results correlate with a greater reduction of Ki-67 and induction of apoptosis. These findings demonstrate the feasibility of utilizing SQ-Gem NPs to make tumor cells more sensitive to Gem and thus provide an efficient new therapeutic alternative for pancreatic adenocarcinoma. FROM THE CLINICAL EDITOR: Pancreatic malignancies represent some of the most notoriously treatment resistant cancer varieties. This paper discusses a novel and promising nanotechnology-based treatment approach, currently at the basic science stage.


Subject(s)
Adenocarcinoma/drug therapy , Antimetabolites, Antineoplastic/therapeutic use , Deoxycytidine/analogs & derivatives , Nanoparticles/chemistry , Pancreatic Neoplasms/drug therapy , Squalene/therapeutic use , Adenocarcinoma/pathology , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/chemistry , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/chemistry , Deoxycytidine/therapeutic use , Female , Humans , Mice , Mice, Nude , Nanomedicine , Pancreas/drug effects , Pancreas/pathology , Pancreatic Neoplasms/pathology , Squalene/administration & dosage , Squalene/chemistry , Gemcitabine
3.
Neoplasia ; 11(7): 637-50, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19568409

ABSTRACT

Gemcitabine is a first-line agent for advanced pancreatic cancer therapy. However, its efficacy is often limited by its poor intracellular metabolism and chemoresistance. To exert its antitumor activity, gemcitabine requires to be converted to its active triphosphate form. Thus, our aim was to improve gemcitabine activation using gene-directed enzyme prodrug therapy based on gemcitabine association with the deoxycytidine kinase::uridine monophosphate kinase fusion gene (dCK::UMK) and small interference RNA directed against ribonucleotide reductase (RRM2) and thymidylate synthase (TS). In vitro, cytotoxicity was assessed by 3-[4,5-dimethylthiazol-2-yl]-3,5-diphenyl tetrazolium bromide and [(3)H]thymidine assays. Apoptosis-related gene expression and activity were analyzed by reverse transcription-polymerase chain reaction, Western blot, and ELISA. For in vivo studies, the treatment efficacy was evaluated on subcutaneous and orthotopic pancreatic tumor models. Our data indicated that cell exposure to gemcitabine induced a down-regulation of dCK expression and up-regulation of TS and RR expression in Panc1-resistant cells when compared with BxPc3- and HA-hpc2-sensitive cells. The combination of TS/RRM2 small interference RNA with Ad-dCK::UMK induced a 40-fold decrease of gemcitabine IC(50) in Panc1 cells. This strong sensitization was associated to apoptosis induction with a remarkable increase in TRAIL expression and a diminution of gemcitabine-induced nuclear factor-kappaB activity. In vivo, the gemcitabine-based tritherapy strongly reduced tumor volumes and significantly prolonged mice survival. Moreover, we observed an obvious increase of apoptosis and decrease of cell proliferation in tumors receiving the tritherapy regimens. Together, these findings suggest that simultaneous TS/RRM2-gene silencing and dCK::UMK gene overexpression markedly improved gemcitabine's therapeutic activity. Clearly, this combined strategy warrants further investigation.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Deoxycytidine/analogs & derivatives , Genetic Therapy/methods , Pancreatic Neoplasms/therapy , Recombinant Fusion Proteins/therapeutic use , Adenoviridae/genetics , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine Kinase/genetics , Deoxycytidine Kinase/therapeutic use , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression , Gene Expression Profiling , Genetic Vectors , Humans , Mice , Mice, Nude , Nucleoside-Phosphate Kinase/genetics , Nucleoside-Phosphate Kinase/therapeutic use , Prodrugs/administration & dosage , RNA Interference , RNA, Small Interfering , Recombinant Fusion Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoside Diphosphate Reductase/genetics , Thymidylate Synthase/genetics , Xenograft Model Antitumor Assays , Gemcitabine
4.
Mol Ther ; 16(2): 252-60, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17984977

ABSTRACT

Currently, gemcitabine is approved as the first-line therapy for patients with locally advanced or metastatic pancreatic cancer. Unfortunately, because of pre-existing or acquired chemoresistance of most of the tumor cells, gemcitabine has failed to significantly improve the outcome for pancreatic carcinoma patients. The present study explored the possibility of sensitizing pancreatic cancer to gemcitabine chemotherapy by combining the chemotherapy with the proapoptotic genes Bax and TNF-related apoptosis-inducing ligand (TRAIL). We designed two tetracycline-inducible recombinant adenoviruses using the human telomerase reverse transcriptase (hTERT) promoter for transcriptional apoptogene targeting. Our data showed that treatment with the adenoviral systems resulted in high-level expression of Bax and TRAIL genes directly related to apoptosis induction, leading to a significant sensitization of resistant pancreatic tumor cells. Furthermore, treatment with Bax and TRAIL adenoviruses plus a suboptimal dose of gemcitabine resulted in significant tumor regression and prolongation of the experimental animal';s life, in contrast to the weak retardation in tumor growth observed when gemcitabine alone was used. Additionally, using an orthotopic tumor model, we showed the usefulness of a non-invasive whole-body optical imaging for real-time evaluation of therapeutic efficacy. Together, these findings suggest that hTERT-targeted proapoptotic gene expression in combination with gemcitabine may be a potential therapeutic strategy for treatment of pancreatic adenocarcinoma.


Subject(s)
Deoxycytidine/analogs & derivatives , Genetic Therapy/methods , Pancreatic Neoplasms/therapy , TNF-Related Apoptosis-Inducing Ligand/genetics , Telomerase/genetics , bcl-2-Associated X Protein/genetics , Adenoviridae/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Cell Line, Tumor , Deoxycytidine/pharmacology , Disease Models, Animal , Female , Genetic Vectors/genetics , Humans , Mice , Models, Genetic , Promoter Regions, Genetic/genetics , TNF-Related Apoptosis-Inducing Ligand/physiology , Transcription, Genetic , bcl-2-Associated X Protein/physiology , Gemcitabine
5.
Int J Oncol ; 30(6): 1397-406, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17487360

ABSTRACT

The aim of this study was to demonstrate the potential of electrogene therapy with the bacterial purine nucleoside phosphorylase gene (ePNP), on pancreatic carcinoma (PC) large tumors. The in vivo electroporation (EP) conditions and efficacy were investigated on both subcutaneous xenografts of human PC cells in immunocompromised mice and orthotopic intrapancreatic grafts of rat PC cells in syngenic rats. After intratumoral injection of naked plasmid DNA, EP was performed using a two-needle array with 25-msec pulses and either a 300 V/cm field strength for subcutaneous or a 500 V/cm field strength for orthotopic PC, parameters providing the best electrotransfer as reflected by the measurements of both luciferase activity and ePNP mRNA. As expected, tumors developed sensitivity to prodrug treatment (6-methylpurine deoxyribose or fludarabine phosphate). We observed both significant inhibition of tumor growth and extended survival of treated mice. In fact, after prodrug treatment, PC growth in the subcutaneous model was delayed by 50-70% for ePNP-expressing tumors. In an orthotopic pancreatic tumor model, the animal survival was significantly prolonged after ePNP electrogene transfer followed by fludarabine treatment, with one animal out of 10 being tumor-free 6 months thereafter. The current study demonstrates for the first time on PC the in vivo feasibility of electrogene transfer and its therapeutic efficiency using the suicide gene/prodrug system, ePNP/fludarabine. These findings suggest that electrogene therapy strategy must be considered for pancreatic cancer treatment, particularly at advanced stages of the disease.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Agents/pharmacology , Genes, Transgenic, Suicide , Genetic Therapy/methods , Pancreatic Neoplasms/therapy , Purine-Nucleoside Phosphorylase/genetics , Vidarabine/analogs & derivatives , Animals , Cell Line, Tumor , Electrochemotherapy , Electroporation , Gene Transfer Techniques , Genes, Bacterial , Humans , Neoplasms, Experimental/therapy , Prodrugs/pharmacology , RNA, Messenger/analysis , Rats , Rats, Inbred Lew , Vidarabine/pharmacology , Xenograft Model Antitumor Assays
6.
Cancer Sci ; 98(7): 1128-36, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17489984

ABSTRACT

Pancreatic adenocarcinoma remains a fatal disease characterized by rapid tumor progression, high metastatic potential and profound chemoresistance. Gemcitabine is the current standard chemotherapy for advanced pancreatic cancer, but it is still far from optimal and novel therapeutic strategies are needed urgently. Mutations in the k-ras gene have been found in more than 90% of pancreatic cancers and are believed to play a key role in this malignancy. Thus, the goal of this study was to investigate the impact of k-ras oncogene silencing on pancreatic tumor growth. Additionally, we examined whether combining k-ras small interfering RNA (siRNA) with gemcitabine has therapeutic potential for pancreatic cancer. The treatment of tumor cell cultures with the corresponding k-ras siRNA resulted in a significant inhibition of k-ras endogenous expression and cell proliferation. In vivo, tumor xenografts were significantly reduced with k-ras siRNA(GAT) delivered by electroporation. Moreover, combined treatment with pSsik-ras(GAT) plus gemcitabine resulted in strong growth inhibition of orthotopic pancreatic tumors. Survival rate was significantly prolonged and the mean tumor volume was dramatically reduced in mice receiving the combined treatment compared with single agents. Collectively, these findings show that targeting mutant k-ras through specific siRNA might be effective for k-ras oncogene silencing and tumor growth inhibition. The improvement of gemcitabine-based chemotherapy suggests that this strategy might be used therapeutically against human pancreatic cancer to potentiate the effects of conventional therapy.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Deoxycytidine/analogs & derivatives , Gene Silencing , Genes, ras , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , RNA, Small Interfering/genetics , Animals , Carcinoma/drug therapy , Carcinoma/genetics , Carcinoma/pathology , Cell Line, Tumor , Deoxycytidine/therapeutic use , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Plasmids , RNA, Messenger/genetics , Transcription, Genetic , Transfection , Transplantation, Heterologous , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...