Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 8(8): e70594, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23967078

RESUMO

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.


Assuntos
Citosina Desaminase/genética , Flucitosina/metabolismo , Genes Transgênicos Suicidas/genética , Parvovirus H-1/genética , Terapia Viral Oncolítica/métodos , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citosina Desaminase/metabolismo , DNA Recombinante/genética , Feminino , Parvovirus H-1/fisiologia , Humanos , Camundongos , NF-kappa B/metabolismo , Terapia Neoadjuvante , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/virologia , Fosfatidilinositol 3-Quinases/metabolismo , Pró-Fármacos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Leveduras/enzimologia
2.
Nanomedicine ; 7(6): 841-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21419876

RESUMO

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.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antimetabólitos Antineoplásicos/uso terapêutico , Desoxicitidina/análogos & derivados , Nanopartículas/química , Neoplasias Pancreáticas/tratamento farmacológico , Esqualeno/uso terapêutico , Adenocarcinoma/patologia , Animais , Antimetabólitos Antineoplásicos/administração & dosagem , Antimetabólitos Antineoplásicos/química , Apoptose/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Desoxicitidina/administração & dosagem , Desoxicitidina/química , Desoxicitidina/uso terapêutico , Feminino , Humanos , Camundongos , Camundongos Nus , Nanomedicina , Pâncreas/efeitos dos fármacos , Pâncreas/patologia , Neoplasias Pancreáticas/patologia , Esqualeno/administração & dosagem , Esqualeno/química , Gencitabina
3.
Neoplasia ; 11(7): 637-50, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19568409

RESUMO

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.


Assuntos
Antimetabólitos Antineoplásicos/administração & dosagem , Desoxicitidina/análogos & derivados , Terapia Genética/métodos , Neoplasias Pancreáticas/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Adenoviridae/genética , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Desoxicitidina/administração & dosagem , Desoxicitidina Quinase/genética , Desoxicitidina Quinase/uso terapêutico , Ensaio de Imunoadsorção Enzimática , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Vetores Genéticos , Humanos , Camundongos , Camundongos Nus , Núcleosídeo-Fosfato Quinase/genética , Núcleosídeo-Fosfato Quinase/uso terapêutico , Pró-Fármacos/administração & dosagem , Interferência de RNA , RNA Interferente Pequeno , Proteínas Recombinantes de Fusão/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleosídeo Difosfato Redutase/genética , Timidilato Sintase/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
4.
Clin Cancer Res ; 15(2): 511-9, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19147756

RESUMO

UNLABELLED: Pancreatic carcinoma is a gastrointestinal malignancy with poor prognosis. Treatment with gemcitabine, the most potent chemotherapeutic against this cancer up to date, is not curative, and resistance may appear. Complementary treatment with an oncolytic virus, such as the rat parvovirus H-1PV, which is infectious but nonpathogenic in humans, emerges as an innovative option. PURPOSE: To prove that combining gemcitabine and H-1PV in a model of pancreatic carcinoma may reduce the dosage of the toxic drug and/or improve the overall anticancer effect. EXPERIMENTAL DESIGN: Pancreatic tumors were implanted orthotopically in Lewis rats or subcutaneously in nude mice and treated with gemcitabine, H-1PV, or both according to different regimens. Tumor size was monitored by micro-computed tomography, whereas bone marrow, liver, and kidney functions were monitored by measuring clinically relevant markers. Human pancreatic cell lines and gemcitabine-resistant derivatives were tested in vitro for sensitivity to H-1PV infection with or without gemcitabine. RESULTS: In vitro studies proved that combining gemcitabine with H-1PV resulted in synergistic cytotoxic effects and achieved an up to 15-fold reduction in the 50% effective concentration of the drug, with drug-resistant cells remaining sensitive to virus killing. Toxicologic screening showed that H-1PV had an excellent safety profile when applied alone or in combination with gemcitabine. The benefits of applying H-1PV as a second-line treatment after gemcitabine included reduction of tumor growth, prolonged survival of the animals, and absence of metastases on CT-scans. CONCLUSION: In addition to their potential use as monotherapy for pancreatic cancer, parvoviruses can be best combined with gemcitabine in a two-step protocol.


Assuntos
Carcinoma/tratamento farmacológico , Desoxicitidina/análogos & derivados , Vírus Oncolíticos/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Parvovirus/metabolismo , Animais , Antimetabólitos Antineoplásicos/farmacologia , Carcinoma/terapia , Linhagem Celular Tumoral , Desoxicitidina/farmacologia , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Pancreáticas/terapia , Ratos , Ratos Endogâmicos Lew , Gencitabina
5.
Mol Ther ; 16(2): 252-60, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17984977

RESUMO

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.


Assuntos
Desoxicitidina/análogos & derivados , Terapia Genética/métodos , Neoplasias Pancreáticas/terapia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Telomerase/genética , Proteína X Associada a bcl-2/genética , Adenoviridae/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Apoptose/fisiologia , Linhagem Celular Tumoral , Desoxicitidina/farmacologia , Modelos Animais de Doenças , Feminino , Vetores Genéticos/genética , Humanos , Camundongos , Modelos Genéticos , Regiões Promotoras Genéticas/genética , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Transcrição Gênica , Proteína X Associada a bcl-2/fisiologia , Gencitabina
6.
Int J Oncol ; 30(6): 1397-406, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17487360

RESUMO

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.


Assuntos
Adenocarcinoma/terapia , Antineoplásicos/farmacologia , Genes Transgênicos Suicidas , Terapia Genética/métodos , Neoplasias Pancreáticas/terapia , Purina-Núcleosídeo Fosforilase/genética , Vidarabina/análogos & derivados , Animais , Linhagem Celular Tumoral , Eletroquimioterapia , Eletroporação , Técnicas de Transferência de Genes , Genes Bacterianos , Humanos , Neoplasias Experimentais/terapia , Pró-Fármacos/farmacologia , RNA Mensageiro/análise , Ratos , Ratos Endogâmicos Lew , Vidarabina/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Cancer Sci ; 98(7): 1128-36, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17489984

RESUMO

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.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Desoxicitidina/análogos & derivados , Inativação Gênica , Genes ras , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , RNA Interferente Pequeno/genética , Animais , Carcinoma/tratamento farmacológico , Carcinoma/genética , Carcinoma/patologia , Linhagem Celular Tumoral , Desoxicitidina/uso terapêutico , Humanos , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/patologia , Plasmídeos , RNA Mensageiro/genética , Transcrição Gênica , Transfecção , Transplante Heterólogo , Gencitabina
8.
J Gene Med ; 7(5): 672-80, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15580629

RESUMO

BACKGROUND: Pancreatic cancer remains a rapidly fatal disease. Suicide gene therapy has been shown to be an effective tool for pancreatic tumor cell destruction, but a cell-specific gene delivery is required to limit host toxicity. The objective of this study was both to design recombinant vectors in which the suicide gene E. coli purine nucleoside phosphorylase (ePNP) is under the control of either CEA or MUC1 promoter sequences and to investigate on experimental pancreatic carcinomas the selective killing effects of the conditional ePNP/prodrug (MePdR) system. METHODS: Transcriptional activities of CEA and MUC1 promoter sequences were analyzed using luciferase reporter gene constructions. Thereafter, recombinant vectors expressing ePNP under control of the most promising pCEA and pMUC1 sequences were designed and used to establish stable tumor cell transfectants from two human pancreatic cell lines, respectively tumor-marker positive (BxPc3) or negative (Panc-1), then applied for in vitro and in vivo experiments. RESULTS: Transient experiments indicated that CEA and MUC1 promoter sequences confer specificity while preserving high transcriptional activities. The MePdR treatment induced a high in vitro cytotoxicity on the sole CEA- and MUC1-producing cell lines (i.e. BxPc3-CEA and -MUC1/ePNP). In the same way, prodrug treatment induced a significant tumor regression on the sole tumor-marker-positive BxPc3 xenografts, whilst the Panc1-CEA and -MUC1/ePNP tumors were not affected. CONCLUSIONS: These data confirm and extend the antitumor efficacy of the ePNP/MePdR killing system and demonstrate the feasibility of the transcriptional targeting strategy under tumor marker promoter control and thereby a preferential killing of CEA- and MUC1-producing pancreatic tumor cells. Thus, efficient in vivo gene delivery and transcriptional targeting constitute the major future clinical challenge for a selective pancreatic cancer suicide gene strategy.


Assuntos
Escherichia coli/enzimologia , Genes Transgênicos Suicidas , Terapia Genética , Neoplasias Pancreáticas/terapia , Pró-Fármacos/uso terapêutico , Regiões Promotoras Genéticas/genética , Purina-Núcleosídeo Fosforilase/genética , Animais , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/metabolismo , Proliferação de Células , Feminino , Humanos , Luciferases/metabolismo , Camundongos , Camundongos Nus , Mucina-1/genética , Mucina-1/metabolismo , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/genética , Nucleosídeos de Purina/uso terapêutico , Purina-Núcleosídeo Fosforilase/metabolismo , Transcrição Gênica , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Pancreas ; 28(2): E54-64, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15028961

RESUMO

OBJECTIVE: Recent advances in diagnostics, staging, and therapy for pancreatic cancer have not resulted in significant improvements in long-term survival, and development of new approaches is particularly urgent. The use of prodrug-activating genes is a possible approach for cancer gene therapy. The aim of this study was to evaluate the efficacy of Escherichia coli purine nucleoside phosphorylase (ePNP) on pancreatic tumors. ePNP activates the prodrug 6-methylpurine deoxyribose (MePdR) into methyl purine (MeP), which is highly toxic to dividing and nondividing cells. METHODS: A recombinant pCAG-ePNP vector was constructed and used to establish pancreatic cancer cells expressing constitutively ePNP (ePNP+). The ePNP/MePdR system effects were tested in vitro on HA-RPC (rat) and BxPC3 (human) pancreatic cancer cell lines and then in vivo on tumors established in nude mice with BxPC3 ePNP+ cells. RESULTS: MePdR treatment of ePNP+ tumor cells induced cytotoxic and antiproliferative effects in a concentration-dependent manner with a 100% cell death since 5 x 10 mol/L. Bystander effect was strong in vitro as more than 50% of tumor cells were killed by MePdR with only 1%-2% of ePNP+ cells. In vivo, tumor growth was completely abolished with a prodrug treatment initiated 2 days after tumor cell inoculation, and mice remained tumor free. In addition, even if MePdR treatment was applied to large tumors, tumors significantly regressed. CONCLUSION: These preliminary results support the therapeutic potential of the MePdR/ePNP system, which induces a highly cytotoxic effect with a potent bystander effect on pancreatic tumors.


Assuntos
Adenocarcinoma/terapia , Antineoplásicos/uso terapêutico , Genes Transgênicos Suicidas , Terapia Genética , Neoplasias Pancreáticas/terapia , Pró-Fármacos/uso terapêutico , Nucleosídeos de Purina/uso terapêutico , Purina-Núcleosídeo Fosforilase/genética , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Animais , Apoptose , Efeito Espectador , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Terapia Combinada , Escherichia coli/enzimologia , Feminino , Junções Comunicantes/efeitos dos fármacos , Vetores Genéticos , Humanos , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Ensaio Tumoral de Célula-Tronco , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Gene Ther ; 11(1): 16-27, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14681723

RESUMO

Peritoneal dissemination is a common end-stage complication of pancreatic cancer for which novel therapeutic modalities are actively investigated, as there is no current effective therapy. Thus, we evaluated, in a mouse model of pancreatic peritoneal carcinomatosis, the therapeutic potential of a novel nonviral gene therapy approach consisting of bis-guanidinium-tren-cholesterol (BGTC)-mediated lipofection of a combined suicide gene system. Human BxPC-3 pancreatic cells secreting the carcinoembryonic antigen (CEA) tumor marker were injected into the peritoneal cavity of nude mice. After 8 days, intraperitoneal (i.p.) lipofection was performed using BGTC/DOPE cationic liposomes complexed with plasmids encoding the two prodrug-activating enzymes Herpes Simplex Virus thymidine kinase and Escherichia coli cytosine deaminase, the latter being expressed from a bicistronic cassette also encoding E. coli uracil phosphoribosyltransferase. Administration of the lipoplexes was followed by treatment with the corresponding prodrugs ganciclovir and 5-fluorocytosine. The results presented herein demonstrate that BGTC/DOPE liposomes can efficiently mediate gene transfection into peritoneal tumor nodules. Indeed, HSV-TK mRNA was detected in tumor nodule tissues by semiquantitative reverse transcription-polymerase chain reaction analysis. In addition, green fluorescent protein (GFP) fluorescence and X-gal staining were observed in the peritoneal tumor foci following lipofection of the corresponding EGFP and LacZ reporter genes. These expression analyses also showed that transgene expression lasted for about 2 weeks and was preferential for the tumor nodules, this tumor preference being in good agreement with the absence of obvious treatment-related toxicity. Most importantly, mice receiving the full treatment scheme (BGTC liposomes, suicide genes and prodrugs) had significantly lower serum CEA levels than those of the various control groups, a finding indicating that peritoneal carcinomatosis progression was strongly reduced in these mice. In conclusion, our results demonstrate the therapeutic efficiency of BGTC-mediated i.p. lipofection of a combined suicide gene system in a mouse peritoneal carcinomatosis model and suggest that BGTC-based prodrug-activating gene therapy approaches may constitute a potential treatment modality for patients with peritoneal carcinomatosis and minimal residual disease.


Assuntos
Colesterol/análogos & derivados , Genes Transgênicos Suicidas/genética , Terapia Genética/métodos , Guanidinas , Lipossomos/administração & dosagem , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Animais , Biópsia , Antígeno Carcinoembrionário/análise , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , Progressão da Doença , Escherichia coli , Feminino , Flucitosina/farmacologia , Flucitosina/uso terapêutico , Ganciclovir/farmacologia , Ganciclovir/uso terapêutico , Humanos , Lipossomos/química , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Cavidade Peritoneal/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Simplexvirus/enzimologia , Simplexvirus/genética , Timidina Quinase/genética , Timidina Quinase/metabolismo , Transfecção
11.
Mol Ther ; 7(6): 765-73, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12788650

RESUMO

Whole-body imaging of green fluorescent protein (GFP) can be used to test the efficiency of gene carriers for in vivo transduction. The aim of the current study was to determine the sensitivity and the accuracy of a GFP imaging procedure by in vivo investigation of GFP-expressing tumor cells. An improved method of whole-body GFP imaging made use of a laser excitation source and band-pass filters matched specifically to GFP and constitutive tissue fluorescence emission bands. Processing of the primary GFP fluorescence images acquired by the CCD camera subtracted background tissue autofluorescence. Our approach achieved 100% sensitivity and specificity for in vivo detection of 10%-transfected BxPc3 pancreatic tumor after subcutaneous grafting or orthotopical implantation in the pancreas of nude mice. It also detected less transfected tumors (i.e., 1 to 5%) but with a loss in sensitivity (50% of cases). The system was employed over a 5-week period to monitor the persistence of GFP expression in 10%-transfected BxPc3 tumors orthotopically implanted in the pancreas of two nude mice, allowing the direct visualization of tumor progression and spread. In facilitating the temporal-spatial follow-up of GFP expression in vivo, the optimized laser-induced fluorescence imaging device can support preclinical investigations of vectors for therapeutic gene transduction through regular, harmless, real-time monitoring of theirin vivo transductional efficacy and persistence.


Assuntos
Diagnóstico por Imagem/métodos , Proteínas Luminescentes , Neoplasias Pancreáticas/diagnóstico , Animais , Modelos Animais de Doenças , Estudos de Viabilidade , Feminino , Proteínas de Fluorescência Verde , Humanos , Lasers , Proteínas Luminescentes/genética , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Pancreáticas/genética , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Espectrometria de Fluorescência , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...