Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Gene Ther ; 11(8): 658-67, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14973547

RESUMO

Specific activation of apoptosis in tumor cells offers a promising approach for cancer therapy. Induction of apoptosis leads to activation of specific proteases. Two major pathways for caspase activation in mammalian cells have been described. One apoptotic pathway involves members of the tumor necrosis factor family of cytokine receptors (eg death receptor 5 (DR5)). The other pathway is controlled by the Bcl-2 family of proteins. The purpose of this study was to investigate whether increased apoptosis occurs in human glioma cells following infection with a recombinant adenoviral vector encoding the human Bax gene under the control of human vascular endothelial growth factor (VEGF) promoter element (AdVEGFBax) in combination with an anti-human DR5 monoclonal antibody (TRA-8). Specific overexpression of exogenous Bax protein induced apoptosis and cell death in glioma cell lines, through activation of both caspase-8 and -9, leading to activation of downstream caspase-3. The relative sensitivity to AdVEGFBax for the glioma cell lines was U251MG>U373MG>U87MG>D54MG. The recently characterized TRA-8 monoclonal antibody induces apoptosis of most TRAIL-sensitive tumor cells by specific binding to DR5 receptors on the cellular membrane. TRA-8 induced rapid apoptosis and cell death in glioma cells, but did not demonstrate detectable cytotoxicity of primary normal human astrocytes. The efficiency of TRA-8-induced apoptosis was variable in different glioma cell lines. The relative sensitivity to TRA-8 was U373MG>U87MG>U251MG>D54MG. The combination of TRA-8 treatment and overexpression of Bax overcame TRA-8 resistance of glioma cells in vitro. Cell viability of U251MG cells was 71.1% for TRA-8 (100 ng/ml) alone, 75.9% for AdVEGFBax (5 MOI) alone and 41.1% for their combination as measured by MTS assay. Similar enhanced apoptosis results were obtained for the other glioma cell lines. In vivo studies demonstrated that the combined treatment significantly (P<0.05) suppressed the growth of U251MG xenografts and produced 60% complete tumor regressions without recurrence. These data suggest that the combination of TRA-8 treatment with specific overexpression of Bax using AdVEGFBax may be an effective approach for the treatment of human malignant gliomas.


Assuntos
Adenoviridae/genética , Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Glioma/terapia , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas Proto-Oncogênicas/genética , Animais , Anticorpos Monoclonais/farmacologia , Apoptose , Western Blotting/métodos , Neoplasias Encefálicas/metabolismo , Caspases/metabolismo , Morte Celular , Linhagem Celular Tumoral , Terapia Combinada , Fragmentação do DNA , Feminino , Citometria de Fluxo , Expressão Gênica , Glioma/metabolismo , Humanos , Camundongos , Camundongos Nus , Regiões Promotoras Genéticas , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/análise , Receptores do Fator de Necrose Tumoral/imunologia , Fator A de Crescimento do Endotélio Vascular/análise , Fator A de Crescimento do Endotélio Vascular/genética , Proteína X Associada a bcl-2
2.
J Clin Oncol ; 18(21 Suppl): 47S-53S, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11060327

RESUMO

PURPOSE: Epidermal growth factor receptor (EGFr) is overexpressed in a majority of head and neck squamous cell carcinomas, and this overexpression is associated with a poor prognosis. Therefore, EGFr has become the target of investigations aimed at disabling the receptor to determine whether this process leads to improved tumor kill with conventional treatment. MATERIALS AND METHODS: C225 is an anti-EGFr monoclonal antibody that inhibits receptor activity by blocking the ligand binding site. A panel of human head and neck squamous cell carcinoma cell lines was used to study the combination of C225 and radiation. RESULTS: It was determined that the combination of C225 (5 microgram/mL) delivered simultaneously with radiation (3 Gy) resulted in a greater decrement in cellular proliferation than either treatment alone. This reduction in proliferation correlated with reduced EGFr tyrosine phosphorylation and a reduction in phosphorylated signal transducer and activator of transcription-3 (STAT-3) protein (known to protect cells from apoptosis). Also, the decrement in proliferation correlated with increased apoptotic events, thereby indirectly linking C225/radiation-induced regulation of STAT-3 protein to apoptosis. CONCLUSION: This preclinical work serves as important support for the ongoing clinical investigation of C225 and radiotherapy for patients with head and neck carcinomas. The initial results of these clinical studies have been promising.


Assuntos
Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Carcinoma de Células Escamosas/terapia , Receptores ErbB/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/terapia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/radioterapia , Ensaios Clínicos como Assunto , Terapia Combinada , Receptores ErbB/imunologia , Neoplasias de Cabeça e Pescoço/diagnóstico por imagem , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Radiografia , Células Tumorais Cultivadas
4.
Mol Ther ; 1(6): 545-54, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10933979

RESUMO

Overexpression of proapoptotic Bax favors death in cells resistant to ionizing radiation. We hypothesized that expression of Bax via adenoviral-mediated gene delivery could sensitize radiation-refractory cells to radiotherapy. An inducible Bax recombinant adenovirus (Ad/Bax) had been generated using the Cre/loxp system. Human ovarian cancer cell lines and primary, patient-derived cancer cells from ascites were irradiated and infected with the Ad/Bax and an expression-inducing vector, Ad/Cre. Cell death was evaluated by crystal violet staining, fluorescence-activated cell sorter analysis of Annexin V, and colony formation assay (cell lines only). To further characterize the mechanism of death, cell morphology was examined by nuclear staining with Hoechst 33258. Lastly, to evaluate the capacity of the combined treatment to inhibit tumor growth, mice were injected subcutaneously with ovarian cancer cells exposed to Bax, radiation therapy (RT), or both, and tumor size was measured periodically. Infection of the cancer cell lines and primary cells with both Ad/Bax and Ad/Cre significantly enhanced sensitivity to ionizing radiation, achieving high levels of cell killing in short-term assays. In addition, the combination of Bax and radiotherapy reduced the survival fraction of cell lines 2 logs in standard colony-forming assays. Investigation into the involved mechanism suggests that Bax-mediated radiosensitization occurs through both apoptosis and necrosis pathways. Further, mice subcutaneously injected with ovarian tumor cells previously treated with radiation, or with radiation and irrelevant viruses, consistently developed tumor nodules. In addition, approximately 80% of injections were followed by tumor formation after treatment with Ad/Bax and Ad/Cre alone. In contrast, tumor formation was completely inhibited after combined treatment with Ad/Bax and Ad/Cre and radiation. Augmentation of the effect of radiotherapy on human ovarian cancer cells and primary cancer cells from patients via a recombinant adenovirus encoding Bax is feasible.


Assuntos
Adenoviridae/genética , Apoptose/genética , Neoplasias Ovarianas/terapia , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas Proto-Oncogênicas/genética , Proteínas Virais , Animais , Terapia Combinada , Feminino , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos , Humanos , Integrases/genética , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/radioterapia , Tolerância a Radiação/genética , Transplante Heterólogo , Células Tumorais Cultivadas , Proteína X Associada a bcl-2
5.
Gene Ther ; 7(12): 1019-26, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10871750

RESUMO

Radiosensitization of human gastrointestinal tumors by 5-fluorouracil (5-FU) has been studied in vitro and clinically in human cancer therapy trials. The bacterial enzyme cytosine deaminase (CD) converts the nontoxic prodrug 5-fluorocytosine (5-FC) into 5-FU. Human colon cancer cells stably expressing CD have been shown by other investigators to be sensitized to radiation following treatment with 5-FC. We previously used an adenoviral vector under control of the cytomegalovirus promoter (AdCMVCD) encoding the CD gene in combination with 5-FC and a single fraction of radiation exposure to enhance cytotoxicity to human cholangiocarcinoma cells in vitro and in vivo. The purpose of this study was to determine whether AdCMVCD infection and 5-FC with multiple fraction low-dose radiotherapy results in enhanced cytotoxicity. In the present study, we utilized AdCMVCD and 5-FC with single fraction radiotherapy to demonstrate enhanced cytotoxicity to WiDr human colon carcinoma cells in vitro. Additionally, we tested this gene therapy/prodrug treatment strategy employing a fractionated radiation dosing schema in animal models of WiDr colon carcinoma and SK-ChA-1 cholangiocarcinoma. A prolonged WiDr tumor regrowth delay was obtained with AdCMVCD infection in combination with systemic delivery of 5-FC and fractionated external beam radiation therapy compared with control animals treated without radiation, without 5-FC, or without AdCMVCD. The results of treatment with AdCMVCD + 5-FC + radiation therapy to cholangiocarcinoma xenografts were equivalent to those obtained with systemic 5-FU administration + radiation. Thus, the use of AdCMVCD can be effectively combined with clinically relevant 5-FC and radiation administration schemes to achieve enhanced tumor cell killing and increased control of established tumors of human gastrointestinal malignancies.


Assuntos
Neoplasias dos Ductos Biliares/radioterapia , Ductos Biliares Intra-Hepáticos , Colangiocarcinoma/radioterapia , Neoplasias Colorretais/radioterapia , Terapia Genética/métodos , Radiossensibilizantes , Adenoviridae/genética , Animais , Neoplasias dos Ductos Biliares/patologia , Sobrevivência Celular/efeitos da radiação , Colangiocarcinoma/patologia , Neoplasias Colorretais/patologia , Citosina Desaminase , Relação Dose-Resposta à Radiação , Flucitosina/uso terapêutico , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Nucleosídeo Desaminases/genética , Pró-Fármacos/uso terapêutico , Tolerância a Radiação , Transplante Heterólogo , Células Tumorais Cultivadas
6.
Cancer Gene Ther ; 6(3): 209-19, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10359206

RESUMO

Radioimmunotherapy is limited by a variety of factors, including poor tumor penetration of monoclonal antibodies and low levels of intratumoral antigen expression. To address these limitations, a gene therapy strategy was devised to genetically induce tumor cells to express enhanced levels of membrane receptors with high affinity for a radiolabeled peptide. We designated this approach as genetic radioisotope targeting strategy. To this end, an adenoviral vector (AdCMVGRPr) encoding the murine gastrin-releasing peptide receptor (GRPr) was used to achieve a high level of binding of radiolabeled bombesin (BBN). To achieve genetic induction of membrane GRPr specifically to tumor cells, we constructed two adenoviral vectors encoding the GRPr gene under the control of the tumor-specific regulatory elements, DF3 (AdDF3GRPr) or erbB-2 (AderbGRPr). We investigated the binding of [125I]BBN to the GRPr following infection with AdDF3GRPr and AderbGRPr in a panel of human breast, pancreatic, and cholangiocarcinoma tumor cell lines. [125I]BBN binding and GRPr expression increased with increasing multiplicities of infection of AdCMVGRPr in all of the cell lines tested. Breast cancer cell lines expressing erbB-2 showed significant GRPr expression using AderbGRPr. A similar result was observed in breast and cholangiocarcinoma cells infected with AdDF3GRPr expressing MUC1 as detected by immunohistochemistry but was not seen in the pancreatic cell lines tested. Thus, adenoviral vectors with tissue-specific promoter elements can be used to achieve a selective expression of membrane receptors that can be targeted with a radiolabeled peptide. The use of such a transcriptional targeting approach may restrict gene expression to tumors and limit the radiation dose deposited in normal tissues in vivo.


Assuntos
Adenoviridae/genética , Antígenos de Neoplasias/genética , Técnicas de Transferência de Genes , Proteínas Oncogênicas v-erbB/genética , Regiões Promotoras Genéticas , Animais , Fibroblastos/metabolismo , Vetores Genéticos , Humanos , Queratinas/análise , Camundongos , Modelos Genéticos , Mucina-1/análise , Receptores da Bombesina/genética , Células Tumorais Cultivadas
7.
Cancer Biother Radiopharm ; 14(6): 451-63, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10850332

RESUMO

BACKGROUND: Monoclonal antibodies (mAb) to epidermal growth factor receptor (EGFr) inhibit tumor cell proliferation and enhance cytotoxicity of chemotherapeutic agents. The purpose of this study was to investigate the interaction of the anti-EGFr antibody C225 combined with radiotherapy (RT) on EGFr expressing A431 human epidermoid cancer cells. METHODS: Cell proliferation, apoptosis, EGFr expression and phosphorylation, and clonogenic survival were assayed in vitro. A431 tumor growth inhibition and immunohistochemistry analysis of EGFr expression and apoptosis were assessed in vivo. RESULTS: C225 plus RT produced greater inhibition of A431 cell proliferation than C225 or RT alone which was corroborated by enhanced apoptosis. Similar clonogenic survival occurred following the addition of C225 to RT, although colonies were smaller in the presence of C225. C225 produced inhibition of EGF-induced phosphorylation of EGFr without concurrent down-regulation of surface receptor, which was not altered by RT. Combined treatment of mice bearing tumors demonstrated enhancement of complete regressions, reduction in time to tumor size doubling, and prolongation of survival. Significant apoptosis occurred in xenograft tumors treated with C225 with or without RT. CONCLUSIONS: These data demonstrate an interaction between C225 and RT. C225-mediated apoptosis and inhibition of EGFr phosphorylation may be critical in the interaction. Studies to define the precise influence of combined modality treatment on the EGFr signal transduction cascade need to be pursued. The combination of growth factor receptor antibodies and RT has potential application in clinical oncology.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/radioterapia , Animais , Anticorpos Monoclonais/toxicidade , Anticorpos Monoclonais Humanizados , Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Carcinoma de Células Escamosas/patologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Cetuximab , Terapia Combinada , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Humanos , Camundongos , Camundongos Nus , Fosforilação , Transplante Heterólogo , Células Tumorais Cultivadas
8.
J Gastrointest Surg ; 2(3): 283-91, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9841986

RESUMO

Cholangiocarcinoma is a malignancy that is resistant to current therapy. We applied the toxin gene therapy strategy of cytosine deaminase conversion of the nontoxic producing 5-fluorocytosine to 5-fluorouracil combined with radiotherapy to cholangiocarcinoma. The transduction efficiency of SK-ChA-1 cholangiocarcinoma cells was determined by fluorescence-activated cell-sorting analysis following infection with recombinant adenovirus AdCMVLacZ, which encodes thc gene for Beta-galactosidase. To evaluate cytosine deaminase-mediated conversion of 5-fluorocytosine to 5-fluorouracil and subsequent cytotoxicity, SK-ChA-1 cells were infected with the recombinant adenovirus AdCMVCD, which encodes cytosine deaminase, and exposed to 5-fluorocytosine for 6 to 8 days. Additive cytotoxicity of radiation therapy was evaluated by cobalt-60 exposure following AdCMVCD infection and 5-fluorocytosine treatment. SK-ChA-1 cells were transduced (98.4%) by AdCMVLacZ at 100 plaque-forming units per cell. Following infection with AdCMVCD and exposure to 5 to 100 microgram/ml of 5-fluorocytosine, 20% to 64% of SK-ChA-1 cells were killed. A combination of radiation and cytosine deaminase/5-fluorocytosine therapy resulted in enhanced cell killing (83.5% to 91.5%). Cholangiocarcinoma cells were transduced by recombinant adenoviral vectors and were killed by cytosine deaminase-mediated production of 5-fluorouracil. Enhanced cytotoxicity was seen with the addition of external beam radiation. These results provide a foundation for multimodality therapy for human cholangiocarcinoma that combines gene therapy technology with radiation therapy.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Neoplasias dos Ductos Biliares/terapia , Colangiocarcinoma/terapia , Flucitosina/metabolismo , Fluoruracila/uso terapêutico , Terapia Genética , Nucleosídeo Desaminases/farmacologia , Pró-Fármacos/uso terapêutico , Adenoviridae , Antimetabólitos Antineoplásicos/farmacologia , Neoplasias dos Ductos Biliares/radioterapia , Colangiocarcinoma/radioterapia , Terapia Combinada , Citosina Desaminase , Fluoruracila/farmacologia , Terapia Genética/métodos , Vetores Genéticos , Humanos , Radioterapia de Alta Energia , Células Tumorais Cultivadas
9.
Int J Radiat Oncol Biol Phys ; 42(4): 817-22, 1998 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9845103

RESUMO

PURPOSE: The erbB-2 receptor is overexpressed in several human cancers, including ovarian, prostate, and breast. We have developed plasmid and adenoviral vectors expressing an anti-erbB-2 single chain antibody (sFv), directed to the endoplasmic reticulum (ER) of target cells, that is cytotoxic to tumor cells overexpressing erbB-2 through induction of apoptosis. The anti-erbB-2 sFv also sensitizes erbB-2 overexpressing cells to the cytotoxic effects of cisplatin. On this basis, we hypothesized that human ovarian cancer cells expressing anti-erbB-2 sFv with downregulated erbB-2 product, p185erbB-2, also would be sensitized to ionizing radiation. Therefore, we designed experiments to test the ability of the anti-erbB-2 sFv to radiosensitize human ovarian cancer cells in vitro and in vivo. METHODS AND MATERIALS: To test our hypothesis, we established subcutaneous (s.c.) tumors in the flanks of nude mice with SKOV3.ip1 human ovarian cancer cells and SKOV3 cells stably expressing the ER directed anti-erbB-2 sFv (SKOV3/pGT21). The tumors were treated with 10 Gy 60Co, or received no radiation. We then determined the regression rate, delay in regrowth, and time to tumor doubling of the tumors treated with radiation in the transfected group and controls. In addition, SKOV3.ip1 and SKOV3/pGT21 tumors were dissected from the irradiated animals and assayed for differences in p185erbB-2 expression at 12 weeks after irradiation by immunohistochemistry. Further, in vitro clonogenic survival assays were performed on the parental SKOV3.ip1 and SKOV3/pGT21 cell lines. RESULTS: A statistical analysis of the combined data was done for two in vivo experiments. The analysis of the combined data showed that animals with irradiated tumor SKOV3/pGT21 had a significantly higher regression rate (p = 0.0055), longer delay in regrowth (p = 0.0001) and time to tumor doubling (p = 0.0004), than those animals with tumor SKOV3.ip1 that received radiation. We observed a similar significant effect for the same parameters in the unirradiated tumor SKOV3/pGT21 compared to unirradiated tumor SKOV3.ip1. Immunohistochemical analysis of the SKOV3/pGT21 tumor cells demonstrated focal accumulation of p185erbB-2 in scattered clumps of cells and less p185erbB-2 membrane expression than cells of SKOV3.ip1 tumors. However, SKOV3.ip1 and SKOV3/pGT21 cells had similar in vitro sensitivity to radiation. CONCLUSIONS: These data support the hypothesis that tumors with reduced p185erbB-2 expression mediated by the anti-erbB-2 sFv are rendered more susceptible in vivo to the cytotoxic effects of ionizing radiation than tumors that maintain their normal expression of p185erbB-2. However, a similar effect was not observed with the same tumor cells in vitro. Thus, as has been described by others (1, 2), in vitro and in vivo results do not always correlate. Therefore, appropriate assays to assess clinical relevance need to be determined for each particular system studied.


Assuntos
Anticorpos/uso terapêutico , Proteínas de Neoplasias/imunologia , Neoplasias Ovarianas/radioterapia , Tolerância a Radiação/genética , Receptor ErbB-2/imunologia , Animais , Anticorpos/genética , Feminino , Vetores Genéticos , Humanos , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Transplante de Neoplasias , Neoplasias Ovarianas/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Transfecção , Transplante Heterólogo , Ensaio Tumoral de Célula-Tronco
10.
Cancer Res ; 57(19): 4325-32, 1997 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9331094

RESUMO

Cholangiocarcinoma is a virtually incurable tumor, resistant to current surgical, chemotherapy, and radiotherapy interventions. We applied the gene therapy strategy of toxin gene conversion of nontoxic prodrug to chemotherapeutic drug in combination with radiation therapy to the treatment of cholangiocarcinoma. In this regard, 5-fluorouracil (5-FU) is an accepted radiosensitizing and chemotherapeutic agent presently used in cancer therapy. The Escherichia coli enzyme cytosine deaminase (CD) converts the prodrug 5-fluorocytosine (5-FC) to 5-FU. Therefore, our goal was to express the CD gene in the human cholangiocarcinoma cell line, SK-ChA-1, assess the cytotoxicity of intracellular production of 5-FU, and determine any enhanced cell killing by the addition of external beam radiation. The susceptibility of SK-ChA-1 cells to recombinant adenoviral infection was determined by fluorescence-activated cell sorting analysis. We used the recombinant adenoviral vector AdCMVLacZ, encoding the E. coli beta-galactosidase reporter gene under control of the human cytomegalovirus (CMV) promoter, to infect SK-ChA-1 and HeLa cells at 10 and 100 plaque forming units (pfu)/cell, followed by FACS analysis. To evaluate CD-mediated conversion of 5-FC to 5-FU and subsequent cytotoxicity, SK-ChA-1 cells were infected with the recombinant adenovirus AdCMVCD, which encodes CD. Cells were then plated in 96-well microtiter plates and exposed to varying concentrations of 5-FC. Cell proliferation assays (tetrazolium salt conversion to formazan colorimetric assay) were performed beginning 2-8 days after plating. We evaluated the effects of external beam radiation using a single 8 Gy 60Co dose to AdCMVCD infected cells, with prior exposure to 5-FC for 2-3 days. MTS assays were performed following radiation treatment. Radiation dose-response analysis, via clonogenic assay, was used as a more sensitive assay to confirm the interaction of the treatment conditions. s.c. SK-ChA-1 tumors in athymic nude mice were established, which then received three intratumoral injections of 1 x 10(9) pfu AdCMVCD. Mice received i.p. injections of 400 mg/kg of 5-FC twice daily for 7 days beginning the day of initial AdCMVCD injection (day -2). The radiation treatment group received 10 Gy of 60Co exposure to their tumor on day 0. SK-ChA-1 cells were efficiently transduced (48.7 and 99.2%) by 10 and 100 pfu/cell of AdCMVLacZ, respectively. From 37.9 to 84.4% of SK-ChA-1 cells were killed following infection with 10 pfu/cell AdCMVCD and 8 days of exposure to various concentrations of 5-FC (5, 10, 30, 50, and 100 microg/ml). Higher 5-FC concentrations and longer duration of exposure resulted in greater cell killing. Radiation treatment (8 Gy) enhanced cell killing by greater than 70% when combined with 10 or 20 microg/ml of 5-FC. Radiation dose-response analysis with clonogenic assay confirmed enhanced SK-ChA-1 cell cytotoxicity as a result of radiation treatment following AdCMVCD infection and 5-FC exposure, with radiobiological parameters alpha = 0.44 and D0 = 0.96. Combined treatment of SK-ChA-1 tumors with AdCMVCD, 5-FC, and radiation in animals resulted in significantly greater survival, time to tumor regrowth, and doubling time compared to the nonradiation treatment group (P = 0.03, 0.015, and 0.002, respectively). Significantly greater change in tumor size, smaller ratio of final tumor size to original tumor size, and smaller final tumor size were observed in the radiation treatment group compared to the no radiation treatment group (P = 0.02, 0.03, and 0.03, respectively). Human cholangiocarcinoma cells were transduced with a recombinant adenovirus in vitro at high efficiency and were susceptible to CD-mediated intracellular 5-FU production. Radiobiological survival curve parameters confirmed an interactive cytotoxic effect when viral infection and prodrug therapy were combined with external beam radiation exposure. (ABSTRACT TRUNCATED)


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Neoplasias dos Ductos Biliares/terapia , Ductos Biliares Intra-Hepáticos , Colangiocarcinoma/terapia , Fluoruracila/uso terapêutico , Terapia Genética , Nucleosídeo Desaminases/uso terapêutico , Pró-Fármacos/farmacocinética , Radiossensibilizantes/uso terapêutico , Adenoviridae/genética , Animais , Antimetabólitos Antineoplásicos/farmacologia , Neoplasias dos Ductos Biliares/radioterapia , Ductos Biliares Intra-Hepáticos/efeitos dos fármacos , Ductos Biliares Intra-Hepáticos/efeitos da radiação , Colangiocarcinoma/radioterapia , Terapia Combinada , Citomegalovirus/genética , Citosina Desaminase , Fluoruracila/farmacologia , Raios gama , Vetores Genéticos/genética , Humanos , Camundongos , Camundongos Nus , Nucleosídeo Desaminases/genética , Pró-Fármacos/metabolismo , Regiões Promotoras Genéticas , Radiossensibilizantes/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Transplante Heterólogo
11.
J Nucl Med ; 38(8): 1221-9, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9255155

RESUMO

UNLABELLED: The gastrin releasing peptide receptor (GRPr) has a high affinity for the 14 amino acid bombesin peptide. For this analysis, [125I]-Tyr4-bombesin was compared with [125I]-mIP-bombesin (a seven amino acid bombesin analog) for in vitro binding and internalization into tumor cells and for tumor localization in vivo. Also, a recombinant adenoviral vector (AdCMVGRPr) was used for gene transfer to induce the expression of GRPr in human ovarian cancer cells for binding and tumor localization with these radiolabeled peptides. METHODS: [125I]-mIP-bombesin was synthesized and compared with [125I]-Tyr4-bombesin in internalization assays using BNR-11 cells (mouse fibroblast cells stably transfected with GRPr) over a 24-hr period. In vitro binding assays used BNR-11, and A427, HeLa and SKOV3.ip1 human cancer cells, which were either uninfected or infected with AdCMVGRPr. Biodistribution studies were performed in normal BALB/c mice and in athymic nude mice bearing orthotopic SKOV3.ip1 ovarian cancer tumors. The SKOV3.ip1 tumors were induced to express GRPr with the AdCMVGRPr adenoviral vector. RESULTS: Internalization assays showed that [125I]-Tyr4-bombesin was rapidly internalized and catabolized at 37 degrees C with approximately 10% of the radioactivity remaining intracellularly at 4 hr, compared with approximately 30% with [125I]-mIP-bombesin. HeLa, A427 and SKOV3.ip1 cells were all induced to express levels of GRPr that were higher than those seen with the positive control BNR-11 cells. Normal mice showed a lower level of radioactivity in both the blood and thyroid for [125I]-mIP-bombesin [0.26% +/- 0.10% injected dose per gram (ID/g) and 0.24% +/- 0.05% ID] than for [125I]-Tyr4-bombesin (3.5% +/- 1.6% ID/g and 5.2% +/- 4.4% ID) at 4 hr postinjection. Mice bearing intraperitoneal (i.p.) SKOV3.ip1 tumors and given AdCMVGRPr i.p. 5 days after tumor cell inoculation followed by [125I]-mIP-bombesin i.p. at day 7 showed 16.5% +/- 4.8% ID/g in tumor compared with 5.9% +/- 3.0% ID/g with [125I]-Tyr4-bombesin at 4 hr postinjection. Tumor bearing mice given saline or a control adenovirus expressing the beta-galactosidase (LacZ) gene showed significantly lower tumor uptake values of both bombesin peptides. CONCLUSION: Internalization assays showed that [125I]-mIP-bombesin has favorable characteristics compared with [125I]-Tyr4-bombesin with regards to cellular internalization and retention. The results demonstrate successful in vitro and in vivo transduction of human tumor cells with a recombinant adenoviral vector-expressing GRPr. Additionally, tumors transduced in vivo to express GRPr demonstrated significantly greater localization of [125I]-mIP-bombesin when compared with [125I]-Tyr4-bombesin.


Assuntos
Bombesina , Radioisótopos do Iodo , Neoplasias Ovarianas/diagnóstico por imagem , Fragmentos de Peptídeos , Receptores da Bombesina/biossíntese , Adenoviridae , Animais , Bombesina/análogos & derivados , Bombesina/farmacocinética , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/metabolismo , Fragmentos de Peptídeos/farmacocinética , Cintilografia , Distribuição Tecidual , Células Tumorais Cultivadas
12.
Radiat Res ; 147(2): 166-71, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9008208

RESUMO

The genetic defect responsible for hypersensitivity of Chinese hamster ovary (CHO) irs-20 cells to ionizing radiation was found to be recessive in nature and could be complemented to produce wild-type radiosensitivity in irs-20/human hybrids. The radiosensitivities of six hybrid clones were determined based on their colony-forming ability under continuous irradiation at 6 cGy/h. A parallel cytogenetic analysis revealed a concordance between the presence or absence of human chromosome 8 and the resistant or sensitive phenotype. Confirming evidence was obtained using human chromosome 8-specific PCR primers. Positive amplification was obtained in hybrids with wild-type radiosensitivity, while no amplification was obtained in sensitive hybrids. Complementation analysis between radiosensitive CHO irs-20 and murine scid cell lines was carried out to determine whether the defects leading to their ionizing radiation hypersensitivity could be corrected by genetic complementation in the hybrids. Complementation did not occur. A transient V(D)J recombination assay after the introduction of the RAG1 and RAG2 genes indicated that the V(D)J recombination ability of the CHO irs-20 cells was about 10% of that for the CHO wild-type cells for signal join formation with an 80% joining fidelity and only 3% of the parental level for coding join formation. These data show that murine scid and irs-20 mutant hamster cells fall into the same complementation group and show similar defects in V(D)J recombination.


Assuntos
Células CHO/efeitos da radiação , Cromossomos Humanos Par 8/genética , Cricetulus/genética , Proteínas de Homeodomínio , Células Híbridas/efeitos da radiação , Camundongos SCID/genética , Tolerância a Radiação/genética , Recombinação Genética , Imunodeficiência Combinada Severa/genética , Animais , Mapeamento Cromossômico , Ensaio de Unidades Formadoras de Colônias , Cricetinae , DNA Nucleotidiltransferases/metabolismo , Reparo do DNA/genética , Proteínas de Ligação a DNA , Rearranjo Gênico , Genes Recessivos , Teste de Complementação Genética , Humanos , Células Híbridas/enzimologia , Camundongos , Proteínas Nucleares , Proteínas/genética , Proteínas/fisiologia , Proteínas Recombinantes de Fusão/metabolismo , Imunodeficiência Combinada Severa/enzimologia , Transfecção , VDJ Recombinases
13.
Gene Ther ; 3(12): 1042-68, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8986431

RESUMO

This review presents an overview and discussion of the potential synergistic strategies of radiation therapy and gene transfer for treating neoplastic disease. Topics discussed include radiation-inducible promoters coupled to genes which produce proteins that are cytotoxic or enhance radiosensitivity, employment of molecular chemotherapy approaches in conjunction with radiation therapy, and genetic induction of radiosensitization through modification of DNA repair, signal transduction, and cell cycle control genes. Additional topics discussed relate to gene transfer augmentation of radioimmunotherapy of cancer. Specifically, gene transfer methods to genetically induce tumor cells to express enhanced levels of cell surface antigens and receptors to increase radiolabeled antibody and peptide targeting and thus increase their therapeutic effect, selection of radionuclides for therapeutic ligand labeling, and computer simulation of genetic tumor-specific delivery of radiolabeled ligands are proposed.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética , Neoplasias/radioterapia , Animais , Terapia Combinada , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/terapia
15.
Int J Radiat Biol ; 65(5): 571-82, 1994 May.
Artigo em Inglês | MEDLINE | ID: mdl-7910196

RESUMO

The ionizing radiation-sensitive mutant of CHO cells, irs-20, showed a defect in the rate and extent of rejoining of gamma-ray-induced DNA double-strand breaks (dsbs) compared with the parental CHO cells as measured using pulsed-field gel electrophoresis. Virtually all rejoined in wild-type cells but some 20-30% failed to rejoin in the mutant cells during a 5-h period after irradiation. An increased level of chromosome-type aberrations per unit dose was seen in irs-20 cells compared with wild-type cells irradiated during the G1 phase. For the irs-20 cells, about half the dose was required to produce the same chromosome-type aberration frequency. Chromatid-type aberrations were induced in G1-irradiated irs-20 cells at frequencies nearly the same as for chromosome types. For the parental wild-type CHO 10B2 cells, only chromosome types were seen. The distribution of aberrations among cells was not significantly different from Poisson for wild-type cells, but this was not the case for irs-20 cells where the overdispersion was highly significant. The mutant irs-20 cells displayed a much greater cell cycle delay per unit dose (about five-fold) in reaching mitosis after irradiation in G1 than the more radioresistant wild-type parental cells.


Assuntos
Células CHO/fisiologia , Aberrações Cromossômicas , Dano ao DNA , DNA/efeitos da radiação , Mitose/efeitos da radiação , Mutação , Animais , Células CHO/citologia , Células CHO/efeitos da radiação , Ciclo Celular/efeitos da radiação , Cricetinae , Raios gama , Fenótipo , Tolerância a Radiação
16.
Mutat Res ; 323(1-2): 47-52, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7508566

RESUMO

The mouse lymphoma (L5178Y) cell mutant M10 is defective in rejoining DNA double-strand breaks and is hypersensitive to ionizing radiation. The introduction of human chromosome 5 into M10 cells by microcell mediated chromosome transfer complemented the ionizing-radiation hypersensitivity defect of this cell line. The presence of chromosome 5 in the microcell hybrids was shown using PCR with chromosome-specific primers and fluorescence in situ hybridization. From this data we conclude that the gene that corrects the radiation hypersensitivity of M10 cells is located on chromosome 5 and tentatively assigned to the 5q14 to 5pter region. We designate this gene XRCC4L.


Assuntos
Cromossomos Humanos Par 5 , Teste de Complementação Genética , Mutação , Animais , Reparo do DNA/genética , Humanos , Hibridização in Situ Fluorescente , Camundongos , Reação em Cadeia da Polimerase , Tolerância a Radiação , Células Tumorais Cultivadas
17.
Radiat Res ; 136(2): 250-4, 1993 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8248482

RESUMO

An ionizing radiation-sensitive mutant of CHO cells, irs-20, irradiated in plateau-phase cultures showed a complete lack of a dose-rate effect, i.e., there was no reduction in cell killing per unit dose as the dose rate was lowered from 0.75 Gy/min to 0.06 Gy/h. No measurable repair of potentially lethal damage or sublethal damage was evident for irs-20 cells in the G1 phase of the cell cycle. During S phase, irs-20 cells showed repair of sublethal damage similar to that for the wild-type cells.


Assuntos
Células CHO/efeitos da radiação , Tolerância a Radiação , Animais , Sobrevivência Celular/efeitos da radiação , Cricetinae , Cricetulus , Relação Dose-Resposta à Radiação , Mutação , Radiação Ionizante
18.
Radiat Res ; 136(2): 241-9, 1993 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7504309

RESUMO

We have isolated a stable mutant of CHO cells, designated irs-20, that is hypersensitive to ionizing radiation. The selection system was designed to select for mutants unable to proliferate at the low dose rate of 0.06 Gy per hour, a dose rate that has little influence on the effect of radiation on the cell cycle of wild-type cells during 1- to 2-week exposures. The irs-20 mutant cells irradiated continuously at 0.06 Gy/h showed a cell cycle redistribution, with an increasing G2 + M-phase fraction, but underwent approximately four doublings before cell population growth was completely inhibited. Dose rates three to four times higher were required to produce similar perturbation in the cell cycle of wild-type cells. Asynchronous log-phase irs-20 cells were approximately twofold more sensitive than the parental CHO cells as measured by comparing the doses required to reduce survival to 10%. The survival response of synchronized irs-20 cells after a single radiation dose of 3.8 Gy at different times during the cell cycle was qualitatively similar to the pattern for wild-type CHO cells for an approximately isosurvival dose of 7.4 Gy. The irs-20 cells were hypersensitive to the "radiomimetic" drug bleomycin but showed the wild-type sensitivity to ethyl methane sulfonate, ultraviolet light (254 nm) and mitomycin C. The irs-20 mutant cell has maintained its phenotype for over 1 year in continuous culture, indicating that the defect is genetically stable. The karyotype of the mutant cells is not different from that of its parent. Further evidence of stability is that clonal lines derived from cells surviving high radiation doses also had the irs-20 phenotype, and treatments with 5-azacytidine sufficient to cause high reversion (approximately 2 x 10(-1)) to proline independence resulted in no measurable reversion to wild-type radiosensitivity.


Assuntos
Células CHO/efeitos da radiação , Separação Celular/métodos , Tolerância a Radiação , Animais , Azacitidina/farmacologia , Ciclo Celular , Sobrevivência Celular/efeitos da radiação , Cricetinae , Cricetulus , Cariotipagem , Mutação , Radiação Ionizante
19.
Int J Radiat Biol ; 64(2): 225-30, 1993 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8103547

RESUMO

Recently, it has been hypothesized (Iliakis and Seaner 1990) that DNA double-strand break (dsb) repair proficiency is a prerequisite for heat radiosensitization on the basis of the finding that the radiosensitive and dsb-repair-deficient mutant xrs-5 cell line shows no significant heat-induced radiosensitization (not even for severe heat doses), whereas their wildtype counterpart (CHO) did show such an effect. In the current study, the extent of hyperthermic radiosensitization in a new gamma-radiation-sensitive cell line, irs-20, recently isolated by Stackhouse and Bedford (1991) and a heat-sensitive mutant hs-36 (Harvey and Bedford 1988) was compared with the radiosensitization of their mutual parent CHO 10B12 cell line. The irs-20 and CHO 10B12 cells have comparable heat (43.5 degrees C) sensitivities, whereas hs-36 and CHO 10B12 show a similar sensitivity to gamma- and X-rays. Radiosensitization due to pre-exposure to 43.5 degrees C heating of plateau phase cultures was found for all three cell lines, even after relatively mild heat treatment killing < 20% of cells. Experiments using CHEF electrophoresis confirmed the dsb repair deficiency of the irs-20 cells (Stackhouse and Bedford 1992) and showed that heat inhibited dsb repair in all three cells lines. These data indicate that DNA repair deficiency (overall dsb repair) per se does not imply an absence of the ability for heat radiosensitization.


Assuntos
Células CHO/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Reparo do DNA , Animais , Cricetinae , Dano ao DNA , Eletroforese , Raios gama , Temperatura Alta , Mutação , Doses de Radiação , Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...