Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Clin Microbiol ; 59(3)2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-33303562

RESUMO

As the coronavirus disease 2019 (COVID-19) pandemic second wave is emerging, it is of the upmost importance to screen the population immunity in order to keep track of infected individuals. Consequently, immunoassays for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with high specificity and positive predictive values are needed to obtain an accurate epidemiological picture. As more data accumulate about the immune responses and the kinetics of neutralizing-antibody (nAb) production in SARS-CoV-2-infected individuals, new applications are forecast for serological assays such as nAb activity prediction in convalescent-phase plasma from recovered patients. This multicenter study, involving six hospital centers, determined the baseline clinical performances, reproducibility, and nAb level correlations of 10 commercially available immunoassays. In addition, three lateral-flow chromatography assays were evaluated, as these devices can be used in logistically challenged areas. All assays were evaluated using the same patient panels in duplicate, thus enabling accurate comparison of the tests. Seven immunoassays examined in this study were shown to have excellent specificity (98 to 100%) and good to excellent positive predictive values (82 to 100%) when used in a low (5%)-seroprevalence setting. We observed sensitivities as low as 74% and as high as 95% at ≥15 days after symptom onset. The determination of optimized cutoff values through receiver operating characteristic (ROC) curve analyses had a significant impact on the diagnostic resolution of several enzyme immunoassays by increasing the sensitivity significantly without a large trade-off in specificity. We found that spike-based immunoassays seem to be better correlates of nAb activity. Finally, the results reported here will add to the general knowledge of the interlaboratory reproducibility of clinical performance parameters of immunoassays and provide new evidence about nAb activity prediction.


Assuntos
Anticorpos Neutralizantes/análise , Anticorpos Antivirais/análise , COVID-19/diagnóstico , Ensaios de Triagem em Larga Escala/normas , COVID-19/imunologia , Humanos , Laboratórios , Reprodutibilidade dos Testes , SARS-CoV-2 , Sensibilidade e Especificidade , Estudos Soroepidemiológicos
3.
Ont Health Technol Assess Ser ; 12(16): 1-132, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23074426

RESUMO

BACKGROUND: Minimal research has been conducted into the potential patient safety issues related to administering multiple intravenous (IV) infusions to a single patient. Previous research has highlighted that there are a number of related safety risks. In Phase 1a of this study, an analysis of 2 national incident-reporting databases (Institute for Safe Medical Practices Canada and United States Food and Drug Administration MAUDE) found that a high percentage of incidents associated with the administration of multiple IV infusions resulted in patient harm. OBJECTIVES: The primary objectives of Phase 1b of this study were to identify safety issues with the potential to cause patient harm stemming from the administration of multiple IV infusions; and to identify how nurses are being educated on key principles required to safely administer multiple IV infusions. DATA SOURCES AND REVIEW METHODS: A field study was conducted at 12 hospital clinical units (sites) across Ontario, and telephone interviews were conducted with program coordinators or instructors from both the Ontario baccalaureate nursing degree programs and the Ontario postgraduate Critical Care Nursing Certificate programs. Data were analyzed using Rasmussen's 1997 Risk Management Framework and a Health Care Failure Modes and Effects Analysis. RESULTS: Twenty-two primary patient safety issues were identified with the potential to directly cause patient harm. Seventeen of these (critical issues) were categorized into 6 themes. A cause-consequence tree was established to outline all possible contributing factors for each critical issue. Clinical recommendations were identified for immediate distribution to, and implementation by, Ontario hospitals. Future investigation efforts were planned for Phase 2 of the study. LIMITATIONS: This exploratory field study identifies the potential for errors, but does not describe the direct observation of such errors, except in a few cases where errors were observed. Not all issues are known in advance, and the frequency of errors is too low to be observed in the time allotted and with the limited sample of observations. CONCLUSIONS: The administration of multiple IV infusions to a single patient is a complex task with many potential associated patient safety risks. Improvements to infusion and infusion-related technology, education standards, clinical best practice guidelines, hospital policies, and unit work practices are required to reduce the risk potential. This report makes several recommendations to Ontario hospitals so that they can develop an awareness of the issues highlighted in this report and minimize some of the risks. Further investigation of mitigating strategies is required and will be undertaken in Phase 2 of this research.


Assuntos
Infusões Intravenosas/efeitos adversos , Segurança do Paciente , Avaliação da Tecnologia Biomédica , Humanos , Infusões Intravenosas/enfermagem , Ontário , Medição de Risco , Gestão da Segurança
4.
Br J Cancer ; 104(6): 989-99, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21326240

RESUMO

BACKGROUND: Epithelial ovarian cancer (EOC) cells are prone to metastasise throughout the peritoneal cavity. The epithelial-to-mesenchymal transition (EMT) is a necessary step towards metastatic tumour progression. CA125/MUC16 mucin is a high-molecular-weight glycoprotein overexpressed in the majority of serous carcinomas, suggesting a possible role in the pathogenesis of these cancers. METHODS: The role of CA125/MUC16 in EMT was investigated using single-chain antibody-mediated knockdown of cell surface CA125/MUC16 in overexpressing EOC NIH:OVCAR3 cells. RESULTS: CA125/MUC16 knockdown was associated with morphological alterations along with decreased surface expression of epithelial markers (E-cadherin, cytokeratin-18) and increased expression of mesenchymal markers (N-cadherin, vimentin). Co-immunoprecipitation experiments revealed that CA125/MUC16 binds to E-cadherin and ß-catenin complexes. The in vitro studies showed disruption of cell-cell junctions, enhanced motility, migration and invasiveness in CA125/MUC16 knockdown cells. Enhanced epidermal growth factor receptor (EGFR) activation was observed in CA125/MUC16 knockdown cells along with increased Akt and ERK1/2 phosphorylation, which are downstream effectors of EGFR, and increased MMP-2 and MMP-9 expression and activities. Epidermal growth factor receptor inhibition strongly inhibited the motility of CA125/MUC16 knockdown cells. CONCLUSIONS: Our findings suggest that CA125/MUC16 plays a role in EMT, presumably through its interaction with E-cadherin and ß-catenin complexes and by modulating EGFR and its downstream signalling pathway in NIH:OVCAR3 cells.


Assuntos
Antígeno Ca-125/metabolismo , Carcinoma/metabolismo , Transição Epitelial-Mesenquimal , Receptores ErbB/metabolismo , Proteínas de Membrana/metabolismo , Neoplasias Ovarianas/metabolismo , Antígenos de Superfície/metabolismo , Antígenos de Superfície/fisiologia , Antígeno Ca-125/fisiologia , Caderinas/metabolismo , Carcinoma/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Regulação para Baixo/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Receptores ErbB/fisiologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/fisiologia , Neoplasias Ovarianas/patologia , Ligação Proteica/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , beta Catenina/metabolismo
5.
Oncogene ; 29(40): 5523-36, 2010 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-20661217

RESUMO

Epithelial ovarian cancer (EOC) cells often show increased activity of the PI3K/Akt pathway. In addition, we have previously shown that EOC ascites induce Akt activation in the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-sensitive EOC cell line, CaOV3, leading to TRAIL-mediated apoptosis inhibition. In this study, we investigated the role of Akt in intrinsic resistance to TRAIL, which is common in EOC cells. We report that Akt activation reduces the sensitivity of EOC cells to TRAIL. TRAIL-resistant SKOV3ip1 and COV2 cells were sensitized to TRAIL-induced apoptosis by PI3K or Akt inhibitors although inhibition of PI3K/Akt signaling pathway did not interfere with the recruitment and processing of caspase-8 to the death-inducing signaling complex. Conversely, overexpression of Akt1 in TRAIL-sensitive cells promoted resistance to TRAIL. Although the fact that TRAIL-induced caspase-8 activation was observed in both sensitive and resistant cell lines, Bid cleavage occurred only in sensitive cells or in SKOV3ip1 cells treated with LY294002. Bid expression was low in resistant cells and Akt activation downregulated its expression. Depletion of Bid by siRNA in OVCAR3 cells was associated with a decrease in TRAIL-mediated apoptosis. Overexpression of Bid only in SKOV3ip1 cells enhanced TRAIL-induced apoptosis. Simultaneous blockade of Akt pathway further increased TRAIL-induced apoptosis. Thus, Akt acts upstream of mitochondria and inhibits TRAIL-induced apoptosis by decreasing Bid protein levels and possibly inhibiting its cleavage.


Assuntos
Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/biossíntese , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Ovarianas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/genética , Western Blotting , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Feminino , Expressão Gênica , Humanos , Imunoprecipitação , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Proteínas Proto-Oncogênicas c-akt/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
6.
Oncogene ; 29(24): 3519-31, 2010 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-20400979

RESUMO

Interactions between ovarian cancer cells and the surrounding tumor microenvironment are not well characterized. We have earlier shown that ovarian cancer ascites induces Akt activation and protect tumor cells from TRAIL-induced apoptosis. Here, we investigated the mechanism by which ascites activates Akt. The ability of ovarian cancer ascites to activate Akt and inhibit TRAIL-induced cell death and caspase activity was decreased by heat inactivation, but was retained in ascites fractions >5 kDa. The survival promoting activity of ascites was not affected by inhibitors of growth factor receptor including epidermal growth factor receptor (EGFR), VEGFR, FGFR, Her2/neu, and IGF-R1. However, this activity was inhibited by an alphavbeta5 integrin-blocking antibody, but not by blocking antibodies against alphavbeta3, beta1, or beta3 integrins. alphavbeta5 integrin-blocking antibodies also inhibited ascites-induced Akt phosphorylation and c-FLIPs up-regulation. Ovarian cancer ascites induced a rapid phosphorylation of focal adhesion kinase (FAK), which closely correlated with the phosphorylation of Akt overtime. FAK phosphorylation was strongly inhibited by alphavbeta5 integrin-blocking antibodies. Depletion of FAK content by RNA interference was also associated with inhibition of ascites-mediated Akt activation and survival. These results suggest that ovarian cancer ascites induces FAK and Akt activation in an alphavbeta5 integrin-dependent pathway, which confers protection from TRAIL-induced cell death and caspase activation.


Assuntos
Ascite/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Vitronectina/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ascite/patologia , Morte Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática , Receptores ErbB/antagonistas & inibidores , Feminino , Proteína-Tirosina Quinases de Adesão Focal/deficiência , Proteína-Tirosina Quinases de Adesão Focal/genética , Técnicas de Silenciamento de Genes , Temperatura Alta , Humanos , Peso Molecular , Neoplasias Ovarianas/enzimologia , Fosforilação
7.
Int J Gynecol Cancer ; 18(4): 670-6, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17868340

RESUMO

TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) is a potent inducer of apoptosis but many ovarian cancer cells display intrinsic resistance to TRAIL. The molecular determinants regulating TRAIL sensitivity in these resistant tumor cells are still incompletely understood. We observed that cell detachment enhances TRAIL-induced apoptosis in two TRAIL-resistant ovarian cancer cell lines. This process was accompanied by an increase of caspase activation, which could be blocked by caspase-8 inhibitor IETD. Cell detachment inhibited Akt phosphorylation. Phosphatidylinositol 3-kinase inhibition by LY294002 also enhanced TRAIL-induced apoptosis. Further decreased Akt activity by LY294002 in detached cells translated to increased cell death after TRAIL treatment. Our data indicate that cell detachment enhances TRAIL-induced killing by decreasing Akt activity in TRAIL-resistant ovarian carcinoma cells and suggest that Akt inhibition primes TRAIL-resistant cells to TRAIL-induced apoptosis.


Assuntos
Carcinoma/tratamento farmacológico , Comunicação Celular/fisiologia , Resistencia a Medicamentos Antineoplásicos , Proteína Oncogênica v-akt/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Fosfatidilinositol 3-Quinases/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinoma/metabolismo , Carcinoma/patologia , Adesão Celular/fisiologia , Comunicação Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cromonas/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Feminino , Humanos , Morfolinas/farmacologia , Células Neoplásicas Circulantes/efeitos dos fármacos , Células Neoplásicas Circulantes/metabolismo , Proteína Oncogênica v-akt/fisiologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fosfatidilinositol 3-Quinases/fisiologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Células Tumorais Cultivadas
8.
Cytotherapy ; 5(6): 509-22, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14660047

RESUMO

BACKGROUND: Resistance to conventional chemotherapy is a major clinical problem for ovarian cancer, and long-term survival for patients with advanced-stage disease is rare. Other therapeutic strategies, such as gene therapy, have been explored but several limitations exist, which include low viral vector transduction efficiency, host immune response to the vector, and vector toxicity. METHODS: We developed a cell-based therapy that exploits human mesothelial cells to deliver anticancer modalities for treatment of ovarian cancer. As a proof of concept, we genetically engineered mesothelial with the herpes simplex virus thymidine kinase/ganciclovir (HSVTK/GCV) system to deliver cytotoxicity to human ovarian cancer cells. This system is well characterized, and has been widely used in different gene-therapy based strategies. RESULTS: Our results demonstrate that HSVTK-modified mesothelial cells are sensitive to GCV killing in vitro and support the HSVTK bystander effect. Engineered mesothelial cells can deliver the HSVTK bystander effect to human ovarian cancer cell-lines, as well as to primary ovarian cancer cells. A significant reduction of tumor growth and prolongation of survival in s.c. and i.p. xenograft mouse models of ovarian cancer are obtained with as little as 1% of HSVTK-expressing mesothelial cells. Intraperitoneal administration of HSVTK-expressing mesothelial cells in an established mouse model of ovarian cancer results in a statistically significant prolonged survival of treated animals. Importantly, distribution studies showed that mesothelial cells localize preferentially to tumor sites. DISCUSSION: Our study demonstrates the feasibility of using a mesothelial cell-based therapy for treatment of ovarian cancer, and suggests that this strategy should be further explored.


Assuntos
Efeito Espectador , Células Epiteliais/transplante , Neoplasias Ovarianas/terapia , Simplexvirus/enzimologia , Timidina Quinase/genética , Adenoviridae/genética , Animais , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Transplante de Células , Técnicas de Cocultura , Conexina 43/análise , Conexina 43/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Citometria de Fluxo , Ganciclovir/farmacologia , Terapia Genética/métodos , Vetores Genéticos/genética , Proteínas de Fluorescência Verde , Histocitoquímica , Humanos , Injeções Intraperitoneais , Injeções Subcutâneas , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Camundongos , Camundongos Nus , Camundongos SCID , Timidina Quinase/metabolismo , Transfecção , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , beta-Galactosidase/análise , beta-Galactosidase/genética
9.
Curr Gene Ther ; 1(4): 317-24, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12109059

RESUMO

A better understanding of the molecular events responsible for the development of drug resistance in cancer cells has emerged in recent years. It is now established that tumor cells can acquire drug resistance by alterations of pathways involved in the regulation of apoptosis and that failure to activate this pathway in cancer cells may confer resistance to chemotherapy. This resistance to drug-induced apoptosis is likely to play an important role in tumors that are refractory to chemotherapy. The identification of points in the apoptotic pathway at which dysregulation occurs opens up new therapeutic opportunities in situations where conventional cytotoxic chemotherapy approaches fail. Although these gene therapy-based strategies are still in their infancy they will likely lead to more effective treatments for human cancers. This review will focus on gene therapy strategies developed to specifically target the apoptotic pathway and how these strategies can affect the sensitivity of tumor cells to chemotherapy.


Assuntos
Apoptose/genética , Resistencia a Medicamentos Antineoplásicos/genética , Terapia Genética , Neoplasias/genética , Neoplasias/terapia , Anticorpos/metabolismo , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Oligonucleotídeos Antissenso/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Catalítico/metabolismo
10.
Cancer Gene Ther ; 7(1): 151-9, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10678368

RESUMO

Ablation of c-Myb function might be an effective approach for the therapy of chronic myelogenous leukemia or other c-myb-dependent malignancies. To this end, we have previously used an intracellular anti-c-Myb single-chain antibody (sFv) to achieve the functional knockout of the c-Myb oncoprotein. In this study, we have employed a tetracycline-inducible system to control the expression of the sFv. A nuclear-localizing form of an anti-c-Myb sFv was cloned into a tet-regulated plasmid vector. Using a transient expression system in COS-1 cells, we observed that doxycycline (Dox) induced expression of the sFv in a dose-dependent manner, and that the sFv was localized mainly in the nucleus. The Dox-induced anti-c-Myb sFv also inhibited the transactivating activity of c-Myb in a dose-dependent manner. We subsequently confirmed the Dox-induced expression of the sFv in the leukemia cell line K562. Proliferation of the target leukemia cells was also inhibited. These results suggest that the anti-c-Myb sFv may represent a viable method for gene therapy of c-myb-dependent hematopoietic malignancies.


Assuntos
Anticorpos/imunologia , Leucemia/terapia , Proteínas Proto-Oncogênicas c-myb/imunologia , Animais , Anticorpos/genética , Anticorpos/farmacologia , Células COS , Divisão Celular/efeitos dos fármacos , Doxiciclina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Terapia Genética , Humanos , Células K562 , Leucemia/imunologia , Leucemia/patologia , Proteínas Proto-Oncogênicas c-myb/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Transfecção
11.
Clin Cancer Res ; 5(1): 43-50, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9918201

RESUMO

In this study, we report that an interleukin-6 (IL-6)-inducible E1A-substituting activity can be exploited for the production of infectious adenoviral particles during infection with the E1A-deleted adenovirus (Ad) Ad5dl312. The basal level of complementation can be increased by 1.5 log by induction of the HepG2 cells with recombinant human IL-6. Additionally, the IL-6-inducible E1A-substituting activity can complement E1A deletion in other cancer cell lines to render them Ad producer cells on induction with recombinant human IL-6, although the efficiency of complementation varies between cell lines. Ad5dl312 can replicate in, produce cytotoxic effect, and kill human tumor cells without addition of exogenous IL-6 in the context of tumor cells possessing an IL-6 autocrine arc, such as ovarian tumor cells. In contrast, normal human mesothelial cells isolated from normal human peritoneum lining do not support replication of Ad5dl312, even in the presence of exogenous IL-6. These results suggest that Ad5dl312 could be used as a cytotoxic agent to selectively kill tumor cells responsive to or possessing an IL-6 autocrine arc.


Assuntos
Adenoviridae/fisiologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/terapia , Interleucina-6/farmacologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Replicação Viral/genética , Adenoviridae/genética , Proteínas E1A de Adenovirus/biossíntese , Proteínas E1A de Adenovirus/genética , Carcinoma Hepatocelular/virologia , Deleção de Genes , Terapia Genética/métodos , Humanos , Neoplasias Hepáticas/virologia , Células Tumorais Cultivadas
12.
Can J Infect Dis ; 10(4): 307-12, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22346390

RESUMO

Despite significant advances in the treatment of human immunodeficiency virus (HIV) infection in the past 10 years, it remains an incurable disease. The inability of traditional drug-based therapies to inhibit HIV replication effectively for extended periods of time has stimulated intense research to develop novel approaches for this disease. Current understanding of HIV molecular biology and pathogenesis has opened the way for the development of gene therapy strategies for HIV infections. In this context, a number of intracellular immunization-based strategies have been evaluated, and some of them have reached the stage of phase I/II human clinical trials. These strategies include the use of single-chain antibodies, capsid-targeted viral inactivation, transdominant negative mutants, ribozymes, antisense oligonucleotides and RNA decoys. While a number of issues remain to be studied before intracellular immunization can be applied to the treatment of HIV infections, the significant progress already made in this field is likely to lead to clinical applications.

13.
Biochem Biophys Res Commun ; 252(3): 686-90, 1998 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-9837767

RESUMO

Subgroups of the B cell malignancies are known to be associated with Epstein-Barr virus (EBV) infection, especially in immunocompromised patients. These are fatal and refractory to conventional antineoplastic therapy. B cells are usually post-mitotic cells and even mitogen activated or transformed B cells have shown relative resistance against viral mediated gene transfer. To address this issue, we employed a replication-defective herpes simplex virus-1 (HSV-1) to mediate gene transfer into EBV-transformed B cells. The virus expresses the herpes simplex virus thymidine kinase (HSV-TK) and the E. coli lacZ reporter genes and is designated T0Z.1. We used the lymphoblastoid cell line SWEIG as a model for human EBV-related B cell malignancy. This cell line was established by in vitro EBV infection of primary human peripheral blood mononuclear cells. When SWEIG cells were infected with T0Z.1, X-gal staining revealed lacZ expression in more than 20% cells even at multiplicity of infection (MOI) as low as 1 and the expression persisted for at least one week. Ganciclovir (GCV) administration after T0Z.1 infection effectively decreased the number of the infected tumor cells in a dose-responsive manner. Viral toxicity was analyzed by cell proliferation assay (MTS assay) and found to be little even at 10 MOI infection. Three MOI of the virus yielded maximum antineoplastic effect and more than 50% tumor cells were killed by HSV-TK/GCV. These results suggest the potential utility of replication-defective HSV-1 for the treatment of EBV-related B cell malignancies.


Assuntos
Linfócitos B/virologia , Terapia Genética/métodos , Vetores Genéticos , Herpesvirus Humano 1/genética , Herpesvirus Humano 4/genética , Linfoma de Células B/terapia , Timidina Quinase/genética , Replicação Viral , Antivirais/uso terapêutico , Divisão Celular/efeitos dos fármacos , Transformação Celular Viral , Relação Dose-Resposta a Droga , Ganciclovir/administração & dosagem , Ganciclovir/uso terapêutico , Técnicas de Transferência de Genes , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 4/patogenicidade , Humanos , Linfoma de Células B/virologia , Replicação Viral/genética
14.
Biochem Biophys Res Commun ; 251(1): 124-30, 1998 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-9790918

RESUMO

Aberrant expression of the c-myb proto-oncogene is a key factor in the development of the neoplastic phenotype in a variety of contexts. On this basis, it has been proposed that ablation of c-myb function might be an effective approach for therapy. To this end, we have employed an intracellular single-chain antibody (sFv) approach to achieve the functional knock-out of the c-Myb onco-protein. We derived an anti-c-Myb sFv, which was configured into eukaryotic expression plasmids. We confirmed the expression of the cytoplasmic and nuclear forms of the sFvs in the correct subcellular compartments by immunofluorescent staining. Importantly, the anti-c-Myb sFvs strongly inhibited the transactivation activity of c-Myb. Furthermore, cytotoxic effect of the sFv was observed only in the c-Myb positive cell line K562. These results suggest that anti-c-Myb sFv is a valuable tool for understanding the molecular mechanisms of c-myb induced transformation. In addition, this approach may have potential utility in the gene therapy for c-myb-dependent malignant diseases.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Região Variável de Imunoglobulina/fisiologia , Líquido Intracelular/imunologia , Proteínas Proto-Oncogênicas/imunologia , Transativadores/imunologia , Sequência de Aminoácidos , Animais , Células COS , Células Clonais , Proteínas de Ligação a DNA/genética , Vetores Genéticos/imunologia , Vetores Genéticos/metabolismo , Humanos , Região Variável de Imunoglobulina/biossíntese , Região Variável de Imunoglobulina/isolamento & purificação , Células K562 , Dados de Sequência Molecular , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-myb , Fatores de Tempo , Transativadores/antagonistas & inibidores , Transativadores/fisiologia
15.
Clin Cancer Res ; 4(10): 2455-61, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9796978

RESUMO

A number of preclinical and human clinical gene therapy trials using adenoviral vectors have shown that the number of viral particles necessary to give adequate levels of gene transfer can be associated with significant vector-related toxicity. In an effort to reduce the number of adenoviral particles required for a given level of gene transfer, we sought to redirect adenoviral infection via a receptor that is highly expressed on the target cells. By using basic fibroblast growth factor (FGF2) as the targeting ligand, adenovirus-mediated gene transfer to the human ovarian cancer cell line SKOV3.ip1 was significantly enhanced, permitting the transduction of a greater number of target cells to be achieved by a given dose of virus. In a murine model of human ovarian carcinoma, an FGF2-redirected adenoviral vector carrying the gene for herpes simplex virus thymidine kinase (AdCMVHSV-TK) was shown to result in a significant prolongation of survival compared with the same number of particles of unmodified AdCMVHSV-TK. In addition, equivalent survival rates were achieved with a 10-fold lower dose of the FGF2-redirected AdCMVHSV-TK compared with the unmodified vector. To our knowledge, this is the first report demonstrating that strategies to enhance the efficiency of in vivo transduction of adenoviral vectors will be of clinical utility.


Assuntos
Adenoviridae/genética , Fator 2 de Crescimento de Fibroblastos/farmacologia , Terapia Genética , Neoplasias Ovarianas/terapia , Simplexvirus/enzimologia , Timidina Quinase/genética , Animais , Feminino , Humanos , Camundongos , Camundongos SCID , Neoplasias Ovarianas/mortalidade , Taxa de Sobrevida , Células Tumorais Cultivadas
16.
Cancer Res ; 58(10): 2134-40, 1998 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-9605757

RESUMO

Extensive experimental evidence suggests that Bcl-2 promotes cell survival by preventing the onset of apoptosis induced by a variety of stimuli. In addition, Bcl-2 expression has been correlated with resistance and poor response to chemotherapy in a number of cell types. Therefore, this protein represents a logical target for gene therapy strategies designed to achieve selective gene product ablation. In this study, we have developed an approach based upon intracellular expression of single-chain antibodies (sFvs) to achieve modulation of Bcl-2 protein levels in target cells. Using a transient expression system, we show that this intracellular anti-Bcl-2 sFv mediates specific reduction of Bcl-2 levels. This effect significantly enhances drug-mediated cytotoxicity in Bcl-2-overexpressing tumor cells, whereas transfection of the anti-Bcl-2 sFv did not affect the growth rate of the tumor cell lines. This method thus represents a novel and efficient way to selectively abrogate the activity of Bcl-2.


Assuntos
Anticorpos Monoclonais , Neoplasias da Mama/metabolismo , Proteínas de Neoplasias/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Anticorpos Monoclonais/genética , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Células HeLa/metabolismo , Humanos , Masculino , Proteínas de Neoplasias/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transfecção , Células Tumorais Cultivadas
17.
Gene Ther ; 5(9): 1171-9, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9930317

RESUMO

Epstein-Barr virus (EBV) causes lymphoproliferative diseases in immunocompromised patients and is associated with endemic Burkitt lymphoma, nasopharyngeal carcinoma and some cases of Hodgkin disease. The latent membrane protein 1 (LMP1) of EBV is a transmembrane protein that is essential for the transformation of B lymphocytes. LMP1-mediated up-regulation of Bcl-2 is thought to be an important element in this process. As an approach to explore novel treatments for EBV-associated lymphomas, we constructed a single-chain antibody (sFv) directed against LMP1 to achieve functional inhibition of this oncoprotein in EBV-transformed B lymphocytes. We demonstrated that intracellular expression of an endoplasmic reticulum (ER)-targeted form of this sFv markedly reduced LMP1 protein levels. We also observed a decrease in intracellular level of this protein which correlated with a marked reduction of Bcl-2 expression in EBV-transformed B lymphocytes. We further demonstrated that anti-LMP1 sFv-mediated reduction of Bcl-2 correlated with increased sensitivity of these cells to drug-induced cell death. Therefore, these data suggest that an anti-LMP1 sFv used in combination with conventional chemotherapy may be useful for gene therapy of EBV-associated lymphomas in immunocompromised patients.


Assuntos
Anticorpos Antivirais/uso terapêutico , Antígenos Virais/imunologia , Linfócitos B/virologia , Ativação Linfocitária , Transtornos Linfoproliferativos/terapia , Proteínas da Matriz Viral/imunologia , Anticorpos Antivirais/genética , Antígenos Virais/metabolismo , Linfócitos B/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Transformada , Inibidores Enzimáticos/farmacologia , Expressão Gênica , Genes bcl-2 , Engenharia Genética , Terapia Genética/métodos , Células HeLa , Herpesvirus Humano 4 , Humanos , Líquido Intracelular/metabolismo , Líquido Intracelular/virologia , Transtornos Linfoproliferativos/imunologia , Transtornos Linfoproliferativos/virologia , Microscopia de Fluorescência , Estaurosporina/farmacologia , Proteínas da Matriz Viral/metabolismo
18.
Gene Ther ; 4(12): 1284-8, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9472551

RESUMO

AIDS-related malignancies (ARM) include AIDS-defining cancers such as Kaposi's sarcoma, non-Hodgkin's lymphoma and cervical carcinoma. In addition, certain other malignancies are also increased with human immunodeficiency virus (HIV) infection. New antiretroviral agents and better prophylaxis and treatment of HIV-related opportunistic infections are prolonging the lives of HIV-infected individuals. There will thus likely be a continued rise in the incidence and prevalence of ARM in the long term, even if effective antiretroviral and other AIDS-related therapies reduce their appearance in the short term. There are presently no curative regimens for the common ARM, with the possible exception of some lymphomas. Survival is shortened by most, and treatment is often toxic and poorly tolerated. Gene therapies may thus offer a useful adjunct to conventional treatment strategies for selected ARM. Although some gene therapy strategies may work well in the HIV setting, the chronic viral infection, immunodeficient status of the host, the tendency for HIV-infected individuals to have altered drug metabolism and an increased rate of adverse drug reactions will likely present special challenges. This review summarizes the state-of-the-art in the fledgling field of gene therapy for ARM, and explores areas for future research.


Assuntos
Terapia Genética/métodos , Linfoma Relacionado a AIDS/terapia , Linfoma não Hodgkin/terapia , Sarcoma de Kaposi/terapia , Neoplasias do Colo do Útero/terapia , Síndrome da Imunodeficiência Adquirida/complicações , Feminino , Humanos , Linfoma não Hodgkin/complicações , Masculino , Neoplasias do Colo do Útero/complicações
19.
J Virol ; 61(3): 840-4, 1987 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-3027409

RESUMO

RmI is a circular chimera containing 1.03 copies of polyomavirus DNA and 1,628 base pairs of mouse DNA, joined through direct and inverted repeat sequences. It is excised from the chromosome of a transformed cell via a site-specific recombination event that is dependent on the activation of the viral gene coding for large T antigen. RmI is shown here to be highly infectious for normal mouse cells. This infectivity reflects the ability of RmI to effectively yield unit-length viral DNA via intramolecular recombination. The effectiveness with which infectious viral DNA is produced from RmI is consistent with the idea that the underlying recombination event is site specific, rather than homologous or illegitimate.


Assuntos
Polyomavirus/genética , Replicon , Animais , Sequência de Bases , Transformação Celular Viral , DNA Recombinante , DNA Viral/genética , Camundongos , Recombinação Genética , Replicação Viral
20.
J Virol ; 61(3): 845-50, 1987 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-3027410

RESUMO

RmI, a circular chimera made of the polyomavirus (Py) genome with an insertion of mouse DNA (Ins), effectively undergoes intramolecular recombination in normal mouse cells, as indicated by the conversion of cloned RmI (RmIc) into unit-length Py DNA in transfected cultures. To follow the fate of the cellular component of RmI after recombination, the origin of simian virus 40 (SV40) DNA was inserted into the Ins region of RmIc, generating a new molecular species designated SV-RmIc. The recombination of SV-RmIc in simian cells synthesizing SV40 large T antigen gave rise to a molecule containing the SV40 origin, the reciprocal of unit-length Py DNA. However, SV-RmIc failed to yield unit-length Py DNA in murine cells unless Py large T antigen was provided in trans. In murine cells synthesizing SV40 large T antigen, the only detectable product from SV-RmIc contained only Py sequences, but was heterogeneous in size. These results and others also reported here strongly suggest that Py large T antigen plays a direct role in the resolution of RmI in murine cells.


Assuntos
Antígenos Virais de Tumores/genética , Polyomavirus/genética , Recombinação Genética , Animais , Linhagem Celular , Chlorocebus aethiops , Replicação do DNA , DNA Recombinante , DNA Viral/genética , Camundongos , Replicon
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...