Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Breast Cancer Res ; 11(4): R53, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19635153

RESUMO

INTRODUCTION: The sodium iodide symporter (NIS) directs the uptake and concentration of iodide in thyroid cells. This in turn allows radioiodine imaging and therapy for thyroid cancer. To extend the use of NIS-mediated radioiodine therapy to other types of cancer, we successfully transferred and expressed the sodium-iodide symporter (NIS) gene in prostate, colon, and breast cancer cells both in vivo and in vitro by using non-replicating adenoviral vectors. METHODS: To improve virotherapy efficiency, we developed a conditionally replicating adenovirus (CRAd) in which the transcriptional cassette RSV promoter-human NIScDNA-bGH polyA was also inserted at the E3 region. The E1a gene is driven by the tumor-specific promoter MUC-1 in the CRAd Ad5AMUCH_RSV-NIS. RESULTS: In vitro infection of the MUC-1-positive breast cell line T47D resulted in virus replication, cytolysis, and release of infective viral particles. Conversely, the MUC-1-negative breast cancer cell line MDA-MB-231 was refractory to the viral cytopathic effect and did not support viral replication. The data indicate that Ad5AMUCH_RSV-NIS activity is stringently restricted to MUC-1-positive cancer cells. Radioiodine uptake was readily measurable in T47 cells infected with Ad5AMUCH_RSV-NIS 24 hours after infection, thus confirming NIS expression before viral-induced cell death. CONCLUSIONS: This construct may allow multimodal therapy, combining virotherapy with radioiodine therapy to be developed as a novel treatment for breast and other MUC1-overexpressing cancers.


Assuntos
Adenoviridae/genética , Neoplasias da Mama/terapia , Vírus Defeituosos/genética , Genes Sintéticos , Vetores Genéticos/genética , Radioisótopos do Iodo/uso terapêutico , Mucina-1/genética , Simportadores/genética , Adenoviridae/fisiologia , Proteínas E1A de Adenovirus/biossíntese , Proteínas E1A de Adenovirus/genética , Animais , Neoplasias da Mama/patologia , Neoplasias da Mama/radioterapia , Linhagem Celular Tumoral , Terapia Combinada , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Efeito Citopatogênico Viral , Vírus Defeituosos/fisiologia , Feminino , Vetores Genéticos/uso terapêutico , Humanos , Radioisótopos do Iodo/farmacocinética , Camundongos , Camundongos Nus , Regiões Promotoras Genéticas/genética , Receptores Virais/fisiologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/fisiologia , Simportadores/biossíntese , Replicação Viral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Hum Gene Ther ; 18(10): 916-24, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17931047

RESUMO

In contrast to follicular cell-derived thyroid cancer, medullary thyroid cancer (MTC) remains difficult to treat because of its unresponsiveness to radioiodine therapy, or to conventional chemo- and radiotherapy. We therefore examined the feasibility of radioiodine therapy of MTC after human sodium iodide symporter (hNIS) gene transfer, using the tumor-specific carcinoembryonic antigen (CEA) promoter for transcriptional targeting. NIS gene transfer was performed in vivo in human MTC cell (TT) xenografts, using adenoviral vectors carrying the NIS gene linked to the cytomegalovirus promoter (Ad5-CMV-NIS) or a CEA promoter fragment (Ad5-CEA-NIS). Functional NIS expression was confirmed by immunostaining as well as in vivo (123)I gamma-camera imaging followed by application of a therapeutic (131)I dose. TT cell xenografts in nude mice injected intratumorally with Ad5-CEA-NIS accumulated 7.5 +/- 1.2% ID/g (percentage injected dose per gram tumor tissue; 5 x 10(8) PFU) and 12 +/- 2.95% ID/g (1 x 10(9) PFU) with an average biological half-life of 6.1 +/- 0.8 and 23.6 +/- 3.7 hr, respectively, as compared with accumulation of 8.4 +/- 0.9% ID/g with a biological half-life of 12 +/- 8 hr after application of Ad5-CMV-NIS (5 x 10(8) PFU). After Ad5-CEA-NIS-mediated NIS gene transfer in TT cell xenografts administration of a therapeutic dose of 111 MBq (3 mCi) of (131)I resulted in a significant reduction of tumor growth associated with significantly lower calcitonin serum levels in treated mice as well as improved survival. We conclude that a therapeutic effect of (131)I was demonstrated in vivo in MTC cell xenografts after adenovirus-mediated induction of tumor-specific iodide accumulation by CEA promoter-directed hNIS expression.


Assuntos
Carcinoma Medular/radioterapia , Terapia Genética , Radioisótopos do Iodo/uso terapêutico , Simportadores/genética , Neoplasias da Glândula Tireoide/radioterapia , Adenoviridae/genética , Animais , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/metabolismo , Carcinoma Medular/genética , Linhagem Celular Tumoral , Vetores Genéticos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Regiões Promotoras Genéticas , Simportadores/metabolismo , Neoplasias da Glândula Tireoide/genética , Transdução Genética
3.
Hum Gene Ther ; 17(6): 661-8, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16776574

RESUMO

Pancreatic cancer is the fourth leading cause of cancer death in the United States. It is highly aggressive with no uniformly effective chemotherapy available for metastatic disease. The sodium-iodide symporter (NIS) is a transmembrane protein responsible for uptake of iodide into cells. The presence of NIS in thyroid cells permits diagnostic imaging and therapy of thyroid tumors, using radioiodide. Previous studies from this laboratory reported mucin-1 (MUC1)-driven expression of NIS in cancer cells. MUC1 overexpression has also been reported in 90% of pancreatic tumors. In this study Ad5/MUC1/NIS was used to infect pancreatic cancer cells both in vitro and in vivo, to investigate the potential for radioiodide imaging and ablation of this disease. In vitro studies revealed a 43-fold increase in iodide uptake in NIS-transduced cells compared with controls. In vivo imaging revealed effective iodide uptake and retention at the site of NIS-transduced tumors, with optimal uptake (13% of injected dose) observed 5 hr after iodide administration. Intravenous delivery was performed to investigate potential hepatotoxicity of the construct in the event of virus leakage. Intravenous injection of Ad5/CMV/NIS resulted in robust iodide uptake throughout mouse liver, whereas no uptake was detected in the liver of animals given Ad5/MUC1/NIS intravenously. Administration of therapeutic doses of 131I resulted in significant regression of NIS-transduced tumors, with a mean 50% reduction in volume within 10 weeks of therapy (p<0.0001). The ability to target NIS expression to pancreatic cancer, which has such limited treatment options, may be highly beneficial and warrants further investigation.


Assuntos
Adenoviridae/genética , Terapia Genética , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/terapia , Simportadores/genética , Animais , Linhagem Celular Tumoral , Vetores Genéticos/genética , Humanos , Radioisótopos do Iodo/farmacocinética , Radioisótopos do Iodo/uso terapêutico , Masculino , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/metabolismo , Cintilografia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Simportadores/metabolismo , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Biochem Biophys Res Commun ; 337(1): 22-9, 2005 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-16171777

RESUMO

A recombinant measles virus (MV) expressing the sodium iodide symporter (NIS) is being considered for therapy of advanced multiple myeloma. Auger electrons selectively damage cells in which the isotope decays. We hypothesized that the Auger electron emitting isotope 125I can be used to control viral proliferation. MV was engineered to express both carcinoembryonic antigen and NIS (MV-NICE). Cells were infected with MV-NICE and exposed to 125I with appropriate controls. MV-NICE replication in vitro is inhibited by the selective uptake of 125I by cells expressing NIS. Auger electron damage is partly mediated by free radicals and abrogated by glutathione. In myeloma xenografts, control of MV-NICE with 125I was not possible under the conditions of the experiment. MV-NICE does not replicate faster in the presence of radiation. Auger electron emitting isotopes effectively stop propagation of MV vectors expressing NIS in vitro. Additional work is necessary to translate these observations in vivo.


Assuntos
Elétrons , Vetores Genéticos/efeitos da radiação , Radioisótopos do Iodo , Vírus do Sarampo/genética , Mieloma Múltiplo/terapia , Animais , Anticorpos Antivirais/sangue , Antígeno Carcinoembrionário/genética , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Radicais Livres/metabolismo , Vírus do Sarampo/imunologia , Vírus do Sarampo/efeitos da radiação , Camundongos , Mieloma Múltiplo/imunologia , Simportadores/genética , Células Vero , Replicação Viral/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Mol Ther ; 12(5): 835-41, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16054438

RESUMO

The sodium-iodide symporter (NIS) is primarily a thyroid protein, providing for the accumulation of iodide for biosynthesis of thyroid hormones. Native NIS expression has made possible the use of radioactive iodide to image and treat thyroid disease successfully. The current study, using adult male beagle dogs, was carried out in preparation for a Phase I clinical trial of adenovirus-mediated NIS gene (approved symbol SLC5A5) therapy for prostate cancer. Direct intraprostatic injection of virus (Ad5/CMV/NS) was followed by iv injection of 3 mCi 123I and serial image acquisition. The dogs were then given a therapeutic dose of 131I (116 mCi/m2) and observed for 7 days. SPECT/CT fusion imaging revealed clear images of the NIS-transduced prostates. Dosimetry calculations revealed an average absorbed dose to the prostate of 23 +/- 42 cGy/mCi 131I, with acceptably low radiation doses to other organs. This study demonstrated the successful introduction of localized NIS expression in the prostate gland of dogs, with no vector-related toxicity observed. None of the animals experienced any surgical complications, and serum chemistry panels showed no significant change following therapy. The results presented provide further evidence of the safety and efficacy of NIS as a therapeutic gene and support translation of this work into the clinical setting.


Assuntos
Adenoviridae/genética , Terapia Genética , Neoplasias da Próstata/terapia , Simportadores/uso terapêutico , Animais , Modelos Animais de Doenças , Cães , Radioisótopos do Iodo/uso terapêutico , Masculino , Neoplasias da Próstata/diagnóstico por imagem , Radiometria , Cintilografia , Compostos Radiofarmacêuticos/uso terapêutico , Simportadores/genética , Transdução Genética , Transfecção
6.
Clin Cancer Res ; 11(4): 1483-9, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15746050

RESUMO

PURPOSE: Expression of the sodium iodide symporter (NIS) in the thyroid gland provides for effective imaging and treatment of thyroid cancer using radiolabeled iodide. Transfer of NIS into other tumors would expand the utility of this treatment to tumors of nonthyroid origin. MUC1 is a transmembrane glycoprotein that is overexpressed in many tumor types, including breast, pancreatic, and ovarian. The aim of this study was to create a construct containing NIS under the control of the MUC1 promoter to target expression specifically to MUC1-positive breast cancer cells. EXPERIMENTAL DESIGN: A replication-deficient adenoviral construct was created containing the MUC1 promoter followed by the human NIS gene. Iodide uptake assays, Western blot, and immunohistochemistry were used to confirm NIS expression and function. Breast cancer xenografts in mice were infected with Ad5/MUC1/NIS and then imaged and treated using radioiodide. RESULTS: A 58-fold increase in iodide uptake was observed in infected MUC1-positive T47D cells with no significant increase observed in MUC1-negative MDA-MB-231 cells or in cells infected with the control virus. The in vivo study yielded clear images of Ad/MUC1/NIS-infected tumor xenografts using (123)I. Administration of a therapeutic dose of (131)I resulted in an 83% reduction in tumor volume, whereas control tumors continued to increase in size (P < 0.01). CONCLUSIONS: These results show that the MUC1 promoter is capable of directing efficient and selective expression of the NIS gene in MUC1-positive breast tumor cells. This could potentially have applications for both imaging and therapy in a range of MUC1-positive tumor types.


Assuntos
Neoplasias da Mama/terapia , Mucina-1/genética , Regiões Promotoras Genéticas/genética , Simportadores/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Adenoviridae/genética , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Regulação da Expressão Gênica , Vetores Genéticos/genética , Humanos , Imuno-Histoquímica , Radioisótopos do Iodo/farmacocinética , Radioisótopos do Iodo/uso terapêutico , Camundongos , Camundongos Nus , Simportadores/genética , Fatores de Tempo , Transfecção , Resultado do Tratamento
7.
Biochem Biophys Res Commun ; 325(1): 157-66, 2004 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-15522214

RESUMO

The thyroidal sodium iodide symporter (NIS) in combination with various radioactive isotopes has shown promise as a therapeutic gene in various tumor models. Therapy depends on adequate retention of the isotope in the tumor. We hypothesized that in the absence of iodide organification, isotope trapping is a dynamic process either due to slow efflux or re-uptake of the isotope by cells expressing NIS. Iodide efflux is slower in ARH-77 and K-562 cells expressing NIS compared to a thyroid cell line. Isotope retention half times varied linearly with the number of cells expressing NIS. With sufficient NIS expression, iodide efflux is a zero-order process. Efflux kinetics in the presence or absence of perchlorate also supports the hypothesis that iodide re-uptake occurs and contributes to the retention of the isotope in tumor cells. Iodide organification was insignificant. In vivo studies in tumors composed of mixed cell populations confirmed these observations.


Assuntos
Iodetos/farmacocinética , Neoplasias/metabolismo , Simportadores/farmacocinética , Animais , Feminino , Humanos , Radioisótopos do Iodo/farmacocinética , Células K562 , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neoplasias/patologia , Percloratos/farmacocinética , Ratos
8.
Cancer Res ; 63(22): 7840-4, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14633711

RESUMO

Prostate cancer is one of the most promising candidates for sodium iodide symporter (NIS)-mediated gene therapy. Adenovirus-mediated expression of NIS that is driven by prostate-specific promoters induces generous radioiodine accumulation in prostate cancer cells that may be used for therapy with (131)I. We have recently developed a replication-deficient adenovirus carrying the human NIS cDNA linked to a composite probasin promoter, ARR(2)PB, aiming toward specific expression of the human NIS gene (h-NIS) in prostate tissue for targeted radioactive iodide therapy of prostate cancer (Ad-ARR(2)PB/hNIS). The ability of Ad-ARR(2)PB/hNIS to cause NIS expression in tumor cells was characterized by iodide uptake assay and compared with Ad-CMV/hNIS in which the h-NIS expression is driven by the cytomegalovirus (CMV) promoter. Androgen-dependent prostate cancer cell lines (LNCaP) and non-prostate origin tumor cell lines (SNU449, MCF-7, HCT116, OVCAR-3, and Panc-1) were infected with the viral constructs, and perchlorate-sensitive (125)I uptake and NIS protein expression were measured. Ad-ARR(2)PB/hNIS-infected LNCaP cells showed androgen-dependent and perchlorate-sensitive iodide uptake. Iodide accumulation in LNCaP cells infected with Ad-ARR(2)PB/hNIS, followed by incubation with synthetic androgen, was 5.3-fold increased compared with those coincubated with perchlorate (15,184 +/- 1,173 cpm versus 2,837 +/- 187 cpm). Ad-ARR(2)PB/hNIS-infected LNCaP cells revealed a 3.2-fold increase of iodide accumulation compared with those infected with Ad-CMV/hNIS (multiplicity of infection = 30). Iodide uptake in a panel of non-prostate tumor cell lines infected with Ad-ARR(2)PB/hNIS was no more than 2,500 cpm, demonstrating the tissue specificity of this construct. These results indicate that Ad-ARR(2)PB/hNIS can be used to achieve high-magnitude and tissue-specific expression of h-NIS in prostate tissue and is a promising candidate for cancer gene therapy of prostate cancer.


Assuntos
Proteína de Ligação a Androgênios/genética , Terapia Genética/métodos , Radioisótopos do Iodo/uso terapêutico , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/radioterapia , Iodeto de Sódio/farmacocinética , Simportadores/biossíntese , Adenovírus Humanos/genética , Western Blotting , Linhagem Celular Tumoral , Humanos , Radioisótopos do Iodo/farmacocinética , Masculino , Regiões Promotoras Genéticas , Neoplasias da Próstata/genética , Simportadores/genética , Simportadores/metabolismo
9.
Blood ; 102(2): 489-96, 2003 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12649158

RESUMO

Multiple myeloma is a disseminated neoplasm of terminally differentiated plasma cells that is incurable with currently available therapies. Although the disease is radiosensitive, external beam radiation leads to significant toxicity due to sensitive end-organ damage. Thus, genetic approaches for therapy are required. We hypothesized that the incorporation of immunoglobulin promoter and enhancer elements in a self-inactivating (SIN) lentiviral vector should lead to specific and high-level transgene expression in myeloma cells. A SIN lentivector with enhanced green fluorescent protein (EGFP) expression under the control of a minimal immunoglobulin promoter as well as the Kappa light chain intronic and 3' enhancers transduced myeloma cell lines with high efficiency (30%-90%). EGFP was expressed at a high level in myeloma cells but silent in all nonmyeloma cell lines tested compared with the cytomegalovirus (CMV) promoter/enhancer. Transduction of myeloma cells with the targeted vector coding for the human sodiumiodide symporter (hNIS) led to hNIS expression by these cells allowing them to concentrate radioiodine up to 18-fold compared with controls. Tumor xenografts in severe combined immunodeficiency mice expressing hNIS could be imaged using iodine-123 (123I) and shown to retain iodide for up to 48 hours. These tumor xenografts were completely eradicated by a single dose of the therapeutic isotope iodine-131 (131I) without evidence of recurrence up to 5 months after therapy. We conclude that lentivectors can be transcriptionally targeted for myeloma cells and the use of hNIS as a therapeutic gene for myeloma in combination with 131I needs further exploration.


Assuntos
Vírus Defeituosos/genética , Elementos Facilitadores Genéticos , Vetores Genéticos/uso terapêutico , Radioisótopos do Iodo/uso terapêutico , Lentivirus/genética , Mieloma Múltiplo/radioterapia , Proteínas do Mieloma/genética , Regiões Promotoras Genéticas , Compostos Radiofarmacêuticos/uso terapêutico , Simportadores/genética , Animais , Citomegalovirus/genética , Sistemas de Liberação de Medicamentos , Genes de Imunoglobulinas , Genes Reporter , Genes Sintéticos , Vetores Genéticos/genética , Proteínas de Fluorescência Verde , Humanos , Cadeias kappa de Imunoglobulina/genética , Íntrons/genética , Radioisótopos do Iodo/farmacocinética , Proteínas Luminescentes/genética , Camundongos , Camundongos SCID , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Compostos Radiofarmacêuticos/farmacocinética , Proteínas Recombinantes de Fusão/fisiologia , Simportadores/fisiologia , Transcrição Gênica , Transdução Genética , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/transplante , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...