Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 265
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Gene Ther ; 16(5): 605-19, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19262615

RESUMO

The woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) is widely used in retroviral gene transfer vectors. However, this element contains an open-reading frame (ORF) encoding a truncated peptide of the woodchuck hepatitis virus X protein (WHX). Because we are developing a lentiviral vector for the gene therapy of Wiskott-Aldrich syndrome (WAS), we evaluated whether the WPRE was needed in the gene transfer cassette and tested the possibility of replacing it with a mutated derivative. The transcriptional activity of the WPRE was undetectable in the context of the lentiviral vector but the element was capable of translating a polypeptide. This capability was abrogated by mutating the WHX ORF translation start. The WPRE was required to express high levels of the transgene and for that, the native form or mutated derivatives functioned equivalently. The vector using a WAS gene promoter and the mut6 WPRE induced long-term expression of the WAS transgene in vivo, correcting cytoskeletal defects, thymocyte and B-cell numbers and improved the colitis of WAS-null mice. By providing additional evidence of efficacy of this WAS lentiviral vector with improved safety features, our results validate a mutated WPRE, which should be useful in future gene therapy applications.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/genética , Vírus da Hepatite B da Marmota/genética , Transativadores/genética , Síndrome de Wiskott-Aldrich/terapia , Animais , Linhagem Celular , Colite/patologia , Colite/terapia , Regulação da Expressão Gênica , Humanos , Lentivirus/genética , Camundongos , Mutação , Fases de Leitura Aberta/genética , Plasmídeos/genética , Transativadores/biossíntese , Transdução Genética , Transgenes , Síndrome de Wiskott-Aldrich/patologia
2.
Carcinogenesis ; 29(11): 2236-42, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18753414

RESUMO

Plasminogen activator inhibitor-1 is known to play a paradoxical positive role in tumor angiogenesis, but its contribution to metastatic spread remains unclear. We studied the impact of plasminogen activator inhibitor (PAI)-1 deficiency in a transgenic mouse model of ocular tumors originating from retinal epithelial cells and leading to brain metastasis (TRP-1/SV40 Tag mice). PAI-1 deficiency did not affect primary tumor growth or vascularization, but was associated with a smaller number of brain metastases. Brain metastases were found to be differentially distributed between the two genotypes. PAI-1-deficient mice displayed mostly secondary foci expanding from local optic nerve infiltration, whereas wild-type animals displayed more disseminated nodules in the scissura and meningeal spaces. SuperArray GEarray analyses aimed at detecting molecules potentially compensating for PAI-1 deficiency demonstrated an increase in fibroblast growth factor-1 (FGF-1) gene expression in primary tumors, which was confirmed by reverse transcription-polymerase chain reaction and western blotting. Our data provide the first evidence of a key role for PAI-1 in a spontaneous model of metastasis and suggest that angiogenic factors, such as FGF-1, may be important for primary tumor growth and may compensate for the absence of PAI-1. They identify PAI-1 and FGF-1 as important targets for combined antitumor strategies.


Assuntos
Neoplasias Encefálicas/prevenção & controle , Neoplasias Encefálicas/secundário , Neoplasias Oculares/patologia , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Animais , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Oculares/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Inibidor 1 de Ativador de Plasminogênio/genética , Epitélio Pigmentado da Retina/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
3.
Gene Ther ; 15(21): 1436-45, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18548116

RESUMO

Given as a prophylactic treatment, a single muscle electrogene transfer of plasmid coding canstatin fused to human serum albumin (CanHSA), slowed down the development of two xenografted human carcinomas from mammary (MDA-MB-231) and prostate origin (PC-3) in nude mice and delayed lung metastatic spreading of B16F10 melanoma cells in syngenic mice. No effect was observed with unfused canstatin. The long lasting circulating blood level of CanHSA (20 ng ml(-1)) resulted in a profound disorganization of the tumor blood vessel network. However, when used as a curative treatment, on well-established tumors, CanHSA electrogenetherapy was ineffective in reducing tumor growth. As radiation is known to increase the alpha v beta3 and alpha v beta5 integrins, which are canstatin receptors, to extend the use of CanHSA electrogenetherapy, as a curative treatment, we explored the combination of CanHSA and ionizing radiation. We demonstrated a better efficacy (P=0.01) of the bitherapy over irradiation alone, as a result of strong vessel disorganization and dramatic increase of tumor cells apoptosis. This extremely simple virus free curative protocol could open the door to potential clinical applications, especially for prostate cancer that often develops radioresistance.


Assuntos
Neoplasias da Mama/terapia , Colágeno Tipo IV/genética , Terapia Genética/métodos , Fragmentos de Peptídeos/genética , Neoplasias da Próstata/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Apoptose , Neoplasias da Mama/radioterapia , Linhagem Celular Tumoral , Colágeno Tipo IV/sangue , Terapia Combinada , Eletroporação , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Músculo Esquelético/metabolismo , Fragmentos de Peptídeos/sangue , Neoplasias da Próstata/radioterapia , Dosagem Radioterapêutica , Proteínas Recombinantes de Fusão/genética , Albumina Sérica/genética , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Oncogene ; 25(22): 3150-9, 2006 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-16501609

RESUMO

In vivo neuroblastoma (NB) xenograft model, resistant to the DNA-topoisomerase I inhibitor irinotecan (CPT-11), has been established to study resistance mechanisms acquired in a therapeutic setting. Common mechanisms of resistance were not involved in this resistance. Thus, we compared the gene expression profiles of sensitive, resistant, and reverted tumors using cDNA expression arrays. Expression of selected transcripts was confirmed by quantitative real-time PCR. We found that pleiotrophin (PTN), a heparin-binding growth factor, was the only gene significantly affected: PTN gene expression was downregulated in all resistant tumors (8-14-fold) as compared to sensitive tumors, and was increased (2-4-fold) in all reverted tumors as compared to resistant tumors. PTN thus appeared to be a likely candidate gene associated with resistance to CPT-11 in this in vivo model. To investigate the direct implication of PTN in NB, we transfected two NB cell lines with RNA interferences in order to silence PTN. PTN failed to demonstrate implication in resistance to CPT-11 in vitro but could influence sensitivity to CPT-11 exclusively through an in vivo mechanism. Indeed, vasculature was significantly enhanced in resistant NB xenografts compared to sensitive and reverted xenografts, and we suggest that PTN is acting in our resistant in vivo NB model as an angiostatic factor.


Assuntos
Camptotecina/análogos & derivados , Proteínas de Transporte/genética , Citocinas/genética , Resistencia a Medicamentos Antineoplásicos , Neovascularização Patológica/etiologia , Neuroblastoma/tratamento farmacológico , Animais , Camptotecina/uso terapêutico , Proteínas de Transporte/antagonistas & inibidores , Pré-Escolar , Citocinas/antagonistas & inibidores , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Irinotecano , Masculino , Camundongos , Neuroblastoma/irrigação sanguínea , Neuroblastoma/genética , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Inibidores da Topoisomerase I , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Clin Invest ; 99(4): 608-17, 1997 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-9045862

RESUMO

A serious limitation in the use of the DNA-cleaving, antitumoral-antibiotic, bleomycin during chemotherapy is pulmonary toxicity. Lung injury induced by bleomycin is characterized by an increased deposition of interstitial extracellular matrix proteins in the alveolar wall that compromises respiratory function. Several drugs have been tested in animal models to prevent the pulmonary toxicity of bleomycin, but have not led to a useful clinical treatment because of their adverse effects on other tissues. We have shown that transgenic mice expressing Streptoalloteichus hindustanus (Sh) ble bleomycin resistance protein in pulmonary epithelial cells in the lungs are protected against bleomycin-induced toxicity in lungs. In the present study, we used intranasal administration by adenovirus-mediated gene transfer of the bleomycin resistance Sh ble gene to mouse lung for prevention of bleomycin-induced pulmonary fibrosis. We constructed recombinant adenoviruses Ad.CMVble and Ad.RSVble harboring the bleomycin resistance Sh ble gene under the control of the cytomegalovirus early promoter and the Rous sarcoma virus early promoter, respectively. Transgene expression was detected in epithelia of conducting airways and alveolar septa by immunostaining with a rabbit polyclonal antibody directed against the bleomycin resistance protein and persisted for the duration of drug treatment; i.e., up to 17 d. No toxic effect was seen in adenovirus-treated mice. Pretreatment of mice with Ad.CMVble or Ad.RSVble completely prevented collagen deposition 42-133 d after bleomycin treatment, as measured by lung OH-proline content. Histologic studies indicated that there was little or no lung injury in the adenovirus/bleomycin-treated mice compared with the bleomycin-treated mice. These observations may lead to new approaches for the prevention of bleomycin-induced pulmonary fibrosis.


Assuntos
Acetiltransferases , Adenovírus Humanos/fisiologia , Bleomicina , Técnicas de Transferência de Genes , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/prevenção & controle , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/farmacologia , Brônquios/química , Brônquios/enzimologia , Resistência Microbiana a Medicamentos/genética , Epitélio/química , Epitélio/enzimologia , Feminino , Vetores Genéticos , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Prolina/efeitos dos fármacos , Fibrose Pulmonar/patologia , Streptomyces/genética , beta-Galactosidase/genética
6.
J Clin Invest ; 100(9): 2218-26, 1997 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9410899

RESUMO

Animal studies indicate that the use of replication-deficient adenovirus for human gene therapy is limited by host antivector immune responses that result in transient recombinant protein expression and blocking of gene transfer when rechallenged. Therefore, we have examined immune responses to an adenoviral vector and to the beta-galactosidase protein in four patients with lung cancer given a single intratumor injection of 10(9) plaque-forming units of recombinant adenovirus. The beta-galactosidase protein was expressed in day-8 tumor biopsies from all patients at variable levels. Recombinant virus DNA was detected by PCR in day-30 and day-60 tumor biopsies from all patients except patient 1. A high level of neutralizing antiadenovirus antibodies was detected in patient 1 before Ad-beta-gal injection whereas it was low (patient 3) or undetectable in the other two patients. All patients developed potent CD4 type 1 helper T cell (Th1) responses to adenoviral particles which increased gradually over time after injection. Antiadenovirus cytotoxic T lymphocyte responses were consistently boosted in the two patients examined (patients 3 and 4). Sustained production of anti-beta-galactosidase IgG was observed in all patients except patient 1. Consistent with anti-beta-gal antibody production, all patients except patient 1 developed intense, dose-dependent Th1 responses to soluble beta-galactosidase which increased over time. Strong beta-galactosidase-specific cytotoxic T lymphocyte responses were detected in patients 2, 3, and 4. Our results clearly show that despite the intensity of antiadenovirus responses, transgene protein expression was sufficient to induce strong and prolonged immunity in three patients. Recombinant adenovirus injected directly into the tumor is a highly efficient vector for immunizing patients against the transgene protein.


Assuntos
Neoplasias Pulmonares/terapia , Adenoviridae/genética , Anticorpos Antivirais/biossíntese , Citotoxicidade Imunológica , DNA Viral/análise , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Interferon gama/biossíntese , Interleucina-2/biossíntese , Ativação Linfocitária , Linfócitos T Citotóxicos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Fatores de Tempo , beta-Galactosidase/genética
7.
Cancer Gene Ther ; 14(3): 251-61, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17082795

RESUMO

Different antiangiogenic and antimetastatic recombinant adenoviruses were tested in a transgenic mouse model of metastatic ocular cancer (TRP1/SV40 Tag transgenic mice), which is a highly aggressive tumor, developed from the pigmented epithelium of the retina. These vectors, encoding amino-terminal fragments of urokinase plasminogen activator (ATF), angiostatin Kringles (K1-3), endostatin (ES) and canstatin (Can) coupled to human serum albumin (HSA) were injected to assess their metastatic and antiangiogenic activities in our model. Compared to AdCO1 control group, AdATF-HSA did not significantly reduce metastatic growth. In contrast, mice treated with AdK1-3-HSA, AdES-HSA and AdCan-HSA displayed significantly smaller metastases (1.19+/-1.19, 0.87+/-1.5, 0.43+/-0.56 vs controls 4.04+/-5.12 mm3). Moreover, a stronger inhibition of metastatic growth was obtained with AdCan-HSA than with AdK1-3-HSA (P=0.04). Median survival was improved by 4 weeks. A close correlation was observed between the effects of these viruses on metastatic growth and their capacity to inhibit tumor angiogenesis. Our study indicates that systemic antiangiogenic factors production by recombinant adenoviruses, particularly Can, might represent an effective way of delaying metastatic growth via inhibition of angiogenesis.


Assuntos
Inibidores da Angiogênese/genética , Neoplasias Encefálicas/terapia , Neoplasias Oculares/terapia , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/genética , Albumina Sérica/genética , Fatores Ativadores da Transcrição/genética , Adenoviridae/genética , Inibidores da Angiogênese/uso terapêutico , Angiostatinas/genética , Animais , Proteínas Sanguíneas/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/secundário , Colágeno Tipo IV/genética , Endostatinas/genética , Neoplasias Oculares/genética , Neoplasias Oculares/patologia , Humanos , Camundongos , Camundongos Nus , Camundongos Transgênicos , Neovascularização Patológica , Fragmentos de Peptídeos/genética , Taxa de Sobrevida , Células Tumorais Cultivadas , Ativador de Plasminogênio Tipo Uroquinase/genética
8.
Technol Cancer Res Treat ; 6(4): 301-6, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17668937

RESUMO

Immune cell recruitment during the treatment of sarcoma tumors in mice with irreversible electroporation was studied by immunohistochemistry. Irreversible electroporation is a non-thermal tissue ablation technique in which certain short duration electrical fields are used to permanently permeabilize the cell membrane, presumably through the formation of nanoscale defects in the membrane. Employing irreversible electroporation parameters known to completely ablate the tumors without thermal effects we did not find infiltration of immune cells probably because of the destruction of infiltration routes. We confirm here that immune response is not instrumental in irreversible electroporation efficacy, and we propose that irreversible electroporation may be, therefore, a treatment modality of interest to immunodepressed cancer patients.


Assuntos
Eletroporação/métodos , Sarcoma Experimental/imunologia , Sarcoma Experimental/cirurgia , Animais , Antígenos CD/análise , Linhagem Celular Tumoral , Feminino , Imuno-Histoquímica , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Subpopulações de Linfócitos T/imunologia
9.
Stem Cell Res ; 23: 154-157, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28925363

RESUMO

Multiple Endocrine Neoplasia Type 2B (MEN2B) is a cancer-predisposing syndrome that affects patients with germline RET mutations. The clinical spectrum of the syndrome includes medullary thyroid carcinoma (MTC) and pheochromocytoma. Currently, there is no satisfactory model recapitulating all the features of the disease especially at the level of stem cells. We generated induced pluripotent stem cells (iPSCs) from a patient with RET mutation at codon 918 who developed pheochromocytoma and MTC. These iPSC had normal karyotype, harboured the RETM918T mutation and expressed pluripotency hallmarks. A comprehensive pathological assessment of teratoma was performed after injection in immunodeficient mice.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Pluripotentes Induzidas/patologia , Neoplasia Endócrina Múltipla Tipo 2b/genética , Neoplasia Endócrina Múltipla Tipo 2b/patologia , Mutação/genética , Proteínas Proto-Oncogênicas c-ret/genética , Linhagem Celular , Humanos , Fatores de Risco , Adulto Jovem
10.
Hum Gene Ther ; 17(10): 1019-26, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17007568

RESUMO

Overexpression of RhoA in cancer indicates a poor prognosis, because of increased tumor cell proliferation and invasion and tumor angiogenesis. We showed previously that anti-RhoA small interfering RNA (siRNA) inhibited aggressive breast cancer more effectively than conventional blockers of Rho-mediated signaling pathways. This study reports the efficacy and lack of toxicity of intravenously administered encapsulated anti-RhoA siRNA in chitosan-coated polyisohexylcyanoacrylate (PIHCA) nanoparticles in xenografted aggressive breast cancers (MDA-MB-231). The siRNA was administered every 3 days at a dose of 150 or 1500 microg/kg body weight in nude mice. This treatment inhibited the growth of tumors by 90% in the 150-microg group and by even more in the 1500-microg group. Necrotic areas were observed in tumors from animals treated with anti-RhoA siRNA at 1500 microg/kg, resulting from angiogenesis inhibition. In addition, this therapy was found to be devoid of toxic effects, as evidenced by similarities between control and treated animals for the following parameters: body weight gain; biochemical markers of hepatic, renal, and pancreatic function; and macroscopic appearance of organs after 30 days of treatment. Because of its efficacy and the absence of toxicity, it is suggested that this strategy of anti-RhoA siRNA holds significant promise for the treatment of aggressive cancers.


Assuntos
Neoplasias da Mama/terapia , Quitosana/administração & dosagem , Bombas de Infusão , Transplante de Neoplasias/normas , RNA Interferente Pequeno/administração & dosagem , Proteína rhoA de Ligação ao GTP/genética , Animais , Neoplasias da Mama/irrigação sanguínea , Linhagem Celular Tumoral , Quitosana/uso terapêutico , Quitosana/toxicidade , Humanos , Camundongos , Nanopartículas/administração & dosagem , Nanopartículas/uso terapêutico , Nanopartículas/toxicidade , Transplante de Neoplasias/métodos , Neoplasias/fisiopatologia , Neovascularização Patológica/terapia , RNA Interferente Pequeno/uso terapêutico , RNA Interferente Pequeno/toxicidade
11.
J Natl Cancer Inst ; 88(24): 1857-63, 1996 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-8961977

RESUMO

BACKGROUND: Despite vigorous efforts at curbing tobacco consumption and aggressive combined-modality treatment programs, both the incidence of and the mortality from lung cancer have remained virtually unchanged in the last 10 years. More effective innovative therapies are clearly needed. The direct transfer into tumor cells of tumor suppressor genes or toxic gene products that specifically promote tumor cell death and spare nonmalignant cells is a potentially novel anticancer treatment approach that should be investigated. PURPOSE: On the basis of compelling preclinical data, we initiated a phase I study involving six patients with inoperable lung cancer and an endobronchial lesion accessible by bronchoscopy. Our purpose was to evaluate the feasibility, tolerance, and clinical, biologic, and immunologic effects of the intratumoral administration of a recombinant, replication-deficient adenovirus (rAd.RSV beta-gal), using the Rous sarcoma virus promoter to drive transcription of the Escherichia coli lacZ marker gene that encodes for the bacterial enzyme beta-galactosidase (beta-gal). METHODS: From June 1994 through April 1995, six patients (five males and one female) were enrolled in the trial. A single dose of recombinant virus suspension containing 10(7) or 10(8) plaque-forming units (PFU) was injected intratumorally into two successive cohorts of three patients. Eligible patients received concomitant chemotherapy. Patients were kept under isolation conditions from 3 days before the injection was given until virus excretion was undetectable. Biopsy specimens of the tumor and surrounding mucosa were collected on the 8th day and at 1, 2, and 3 months after injection. They were analyzed by cell culture, polymerase chain reaction (PCR), and beta-gal expression for the presence of recombinant adenovirus. So that the risk of virus recombination or complementation could be minimized, wildtype adenovirus carriers among the hospital staff (identified by PCR) were excluded from contact with patients who were potentially excreting recombinant virus. RESULTS: beta-gal was expressed in tumor biopsy specimens of three patients (one who received the 10(7) PFU dose level and two who received 10(8)). Bronchoalveolar lavage specimens collected immediately after injection were positive for recombinant adenovirus when analyzed in culture and by PCR. All biologic fluids were negative for recombinant virus as judged by PCR after day 12, with the exception of bronchoalveolar lavage specimens (positive PCR up to 90 days in two of three patients treated with 10(8) PFU). The blood samples obtained from the three patients treated with 10(8) PFU showed positive PCR results immediately after virus injection. Patients were kept in isolation for a median of 17 days. The most common toxic effects were moderate bleeding (occurring in two patients) during bronchoscopy and fever (seen in four patients). Endoscopic and clinically objective antitumor responses were seen in four patients, including one patient who showed a complete response by pathologic evaluation. The median survival for the patients was 12.5 months (range, 3-16+ months). Throughout the study, hospital staff remained negative for recombinant adenovirus infection. CONCLUSIONS: This ongoing phase I study has demonstrated that a recombinant adenovirus-mediated marker gene, such as rAd.RSV beta-gal, can be safely introduced into humans and that the gene product is expressed by lung tumor cells of the host.


Assuntos
Neoplasias Brônquicas/terapia , Carcinoma/terapia , Terapia Genética/métodos , Neoplasias Pulmonares/terapia , beta-Galactosidase/genética , Adenoviridae , Neoplasias Brônquicas/enzimologia , Líquido da Lavagem Broncoalveolar , Broncoscopia , Carcinoma/enzimologia , Estudos de Viabilidade , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Neoplasias Pulmonares/enzimologia , Reação em Cadeia da Polimerase
12.
Cancer Res ; 60(13): 3484-92, 2000 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10910060

RESUMO

The Na+/I- symporter (NIS) present in the membranes of thyroid cells is responsible for the capacity of the thyroid to concentrate iodide. This allows treatment of thyroid cancers with 131I. We propose to enlarge this therapeutic strategy to nonthyroid tumors by using an adenoviral vector to deliver the NIS gene into the tumor cells. We constructed a recombinant adenovirus encoding the rat NIS gene under the control of the cytomegalovirus promoter (AdNIS). Infection of SiHa cells (human cervix tumor cells) with AdNIS resulted in perchlorate-sensitive 125I uptake by these cells to a level 125-225 times higher than that in noninfected cells. Similar results were obtained for other human tumor cell lines, including MCF7 and T-47D (mammary gland), DU 145 and PC-3 (prostate), A549 (lung), and HT-29 (colon), demonstrating that the AdNIS vector can function in tumor cells of various origins. In addition, AdNIS-infected tumor cells were selectively killed by exposure to 131I, as revealed by clonogenic assays. To assess the efficiency of this cancer gene therapy strategy in vivo, we injected the AdNIS vector in human tumors (SiHa or MCF7 cells) established s.c. in nude mice. Immunohistological analysis confirmed the expression of the NIS protein in the tumor. Three days after intratumoral injection, AdNIS-treated tumors could specifically accumulate 125I or 123I, as revealed by kinetics and imaging experiments. A quantitative analysis demonstrated that the uptake in AdNIS-injected tumors was 4-25 times higher than that in nontreated tumors. On average, 11% of the total amount of injected 125I could be recovered per gram of AdNIS-treated tumor tissue. Altogether, these data indicate that AdNIS is very efficient in triggering significant iodide uptake by a tumor, outlining the potential of this novel cancer gene therapy approach for a targeted radiotherapy.


Assuntos
Proteínas de Transporte/genética , Terapia Genética/métodos , Iodetos/farmacocinética , Radioisótopos do Iodo/farmacocinética , Proteínas de Membrana/genética , Radioterapia/métodos , Simportadores , Glândula Tireoide/metabolismo , Neoplasias do Colo do Útero/patologia , Adenoviridae , Animais , Transporte Biológico , Neoplasias da Mama , Proteínas de Transporte/metabolismo , Neoplasias do Colo , Feminino , Vetores Genéticos , Humanos , Neoplasias Pulmonares , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Neoplasias da Próstata , Cintilografia , Ratos , Distribuição Tecidual , Transfecção/métodos , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/diagnóstico por imagem
13.
Stem Cell Res ; 17(2): 235-237, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27879209

RESUMO

Chronic myeloid leukemia (CML) is a clonal malignancy initiated by the occurrence of a t (9;22) translocation, generating Ph1 chromosome and BCR-ABL oncogene in a primitive hematopoietic stem cell (HSC). The resistance of HSC to targeted therapies using tyrosine kinase inhibitors remains a major obstacle towards the cure. We have generated an iPSC line from a patient with CML using leukemic CD34+ cells cryopreserved at diagnosis. Ph1+ CML cells were reprogrammed by non-integrative viral transduction. These iPSCs harboured Ph1 chromosome and expressed pluripotency hallmarks as well as BCR-ABL. Teratoma assays revealed normal differentiation after injection in immunodeficient mice.


Assuntos
Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Leucócitos Mononucleares/citologia , Adolescente , Animais , Antígenos CD34/metabolismo , Diferenciação Celular , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/transplante , Cariótipo , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Leucócitos Mononucleares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Teratoma/metabolismo , Teratoma/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transplante Heterólogo , Células Tumorais Cultivadas
14.
Stem Cell Res ; 17(1): 154-157, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27558615

RESUMO

Multiple Endocrine Neoplasia Type 2A (MEN2A) is a cancer-predisposing syndrome that affects patients with germline RET mutations. The clinical spectrum of the syndrome includes medullary thyroid carcinoma (MTC), pheochromocytoma, hyperparathyroidism and cutaneous lichen amyloidosis (CLA) and/or Hirschsprung disease in some variants. Currently, there is no satisfactory animal model recapitulating all the features of the disease especially at the level of stem cells. We generated induced pluripotent stem cells (iPSCs) from a patient with RET mutation at codon 634 who developed pheochromocytoma and MTC. RETC634Y-mutated cells were reprogrammed by non-integrative viral transduction. These iPSCs had normal karyotype, harboured the RETC634Y mutation and expressed pluripotency hallmarks as well as RET. A comprehensive pathological assessment of teratoma was performed after injection in immunodeficient mice.

15.
Oncogene ; 34(24): 3207-13, 2015 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-25065593

RESUMO

Osteosarcoma is the most prevalent primary pediatric cancer-related bone disease. These tumors frequently develop resistance to chemotherapy and are highly metastatic, leading to poor outcome. Thus, there is a need for new therapeutic strategies that can prevent cell dissemination. We previously showed that CYR61/CCN1 expression in osteosarcoma cells is correlated to aggressiveness both in vitro and in vivo in mouse models, as well as in patients. In this study, we found that CYR61 is a critical contributor to the vascularization of primary tumor. We demonstrate that silencing CYR61, using lentiviral transduction, leads to a significant reduction in expression level of pro-angiogenic markers such as VEGF, FGF2, PECAM and angiopoietins concomitantly to an increased expression of major anti-angiogenic markers such as thrombospondin-1 and SPARC. Matrix metalloproteinase-2 family member expression, a key pathway in osteosarcoma metastatic capacity was also downregulated when CYR61 was downregulated in osteosarcoma cells. Using a metastatic murine model, we show that CYR61 silencing in osteosarcoma cells results in reduced tumor vasculature and slows tumor growth compared with control. We also find that microvessel density correlates with lung metastasis occurrence and that CYR61 silencing in osteosarcoma cells limits the number of metastases. Taken together, our data indicate that CYR61 silencing can blunt the malignant behavior of osteosarcoma tumor cells by limiting primary tumor growth and dissemination process.


Assuntos
Neoplasias Ósseas/irrigação sanguínea , Proteína Rica em Cisteína 61/genética , Neovascularização Patológica/genética , Osteossarcoma/irrigação sanguínea , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Proliferação de Células/genética , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos Transgênicos , Invasividade Neoplásica , Metástase Neoplásica , Osteossarcoma/genética , Osteossarcoma/patologia , Interferência de RNA , Células Tumorais Cultivadas
16.
Hum Gene Ther ; 9(13): 1919-28, 1998 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9741430

RESUMO

Adenoviruses are attractive vectors for gene transfer into cardiac muscle. However, their promiscuous tissue tropism, which leads to an ectopic expression of the transgene, is a considerable limitation. To restrict expression to cardiomyocytes, we have constructed two recombinant adenoviruses (Ad-MLC2-250betagal and Ad-MLC2-2100betagal) containing the beta-galactosidase reporter gene under the control of the 250- or 2100-bp rat ventricle-specific cardiac myosin light chain-2v promoter (MLC-2v). Our in vitro and in vivo data have evidenced that the 2100-bp promoter allows stronger beta-galactosidase activity than the 250-bp promoter and that the deleted promoter allows a weak beta-galactosidase expression in skeletal muscle-derived cells in vitro. In contrast to the in vitro results, the highly deleted MLC-2v promoter of 250 pb conserved its heart specificity in in ovo and in vivo when introduced into the adenovirus genome, indicating that the specificity of this promoter is neither altered by the inverted terminal repeat nor by the enhancer of the Ela promoter, both of which located in the 5' flanking region of the promoter. Systemic injections of both recombinant adenoviruses into chicken embryos showed beta-galactosidase expression mainly in the right ventricle of the heart. We have confirmed the cardiac specificity of both promoters in mammalian species after injection of both recombinant adenoviruses into the heart of adult rats in vivo. The comparison of both promoters in vitro and in vivo has shown that the 250-bp MLC-2v promoter is 80% less active than the 2100-bp MLC-2v promoter and has enabled us to conclude that the MLC-2v promoter of 2100 bp is the most appropriate for efficient expression of a reporter gene or a therapeutic cardiac gene (e.g., SERCA2a or minidystrophin gene).


Assuntos
Adenoviridae/genética , Miosinas Cardíacas , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Ventrículos do Coração/metabolismo , Cadeias Leves de Miosina/genética , Proteínas E1A de Adenovirus/genética , Animais , Células Cultivadas , Embrião de Galinha , DNA Viral/análise , Elementos Facilitadores Genéticos/genética , Feminino , Regulação da Expressão Gênica/genética , Genes Reporter/genética , Músculo Esquelético/metabolismo , Miocárdio/citologia , Especificidade de Órgãos , Regiões Promotoras Genéticas/genética , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Sequências Repetidas Terminais/genética , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
17.
Hum Gene Ther ; 12(5): 515-26, 2001 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-11268284

RESUMO

TIMP-2 is a natural matrix metalloproteinase (MMP) inhibitor that prevents the degradation of extracellular matrix proteins. It abolishes the hydrolytic activity of all activated members of the metalloproteinase family and in particular that of MT1-MMP, MMP-2, and MMP-9, which are selective for type IV collagenolysis. Since MMPs have been implicated in both cancer progression and angiogenesis, we generated a recombinant adenovirus to deliver human TIMP-2 (AdTIMP-2) and evaluated its anticancer efficiency in three murine models. Our results demonstrated that overexpression in vitro of TIMP-2 inhibited the invasion of both tumor and endothelial cells without affecting cell proliferation. Its in vivo efficiency has been evaluated in murine lung cancer LLC, and colon cancer C51 in syngeneic mice as well as in human breast cancer MDA-MB231 in athymic mice. Preinfection of tumor cells by AdTIMP-2 resulted in an inhibition of tumor establishment in more than 50% of mice in LLC and C51 models and in 100% mice in the MDA-MB231 model. A single local injection of AdTIMP-2 into preestablished tumors of these three types significantly reduced tumor growth rates by 60--80% and tumor-associated angiogenesis index by 25--75%. Lung metastasis of LLC tumor was inhibited by >90%. In addition, AdTIMP-2-treated mice showed a significantly prolonged survival in all the cancer models tested. These data demonstrate the potential of adenovirus-mediated TIMP-2 therapy in cancer treatment.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Inibidor Tecidual de Metaloproteinase-2/genética , Animais , Apoptose , Western Blotting , Colágeno/metabolismo , Meios de Cultivo Condicionados/farmacologia , DNA Complementar/metabolismo , Combinação de Medicamentos , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imuno-Histoquímica , Laminina/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Metástase Neoplásica , Neovascularização Patológica , Proteoglicanas/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas
18.
Hum Gene Ther ; 10(18): 3045-53, 1999 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-10609663

RESUMO

Systemic administration of Ad5-based recombinant adenovirus leads to preferential transduction of the liver. Using this property, we have assessed the potential of venous viral injection to deliver a recombinant antiangiogenic adenovirus to treat cancer dissemination and improve survival. The results demonstrate that venous injection of adenovirus AdmATF, which encodes a secretable mouse ATF (amino-terminal fragment of urokinase) known to inhibit angiogenesis, suppressed angiogenesis induced by colon cancer metastasis growth in mice liver and improved survival. Nude mice were injected intravenously with 5 X 10(9) PFU of AdmATF and subsequently challenged after a 3-day interval by intrasplenically injected human colon carcinoma cells (LS174T, 3 x 10(6)) that home to liver. Microscopic inspection revealed that, within the AdmATF-pretreated mice (n = 8), the size and number of liver-metastasized nodules on day 30 were remarkably reduced (80% in number, p < 0.05) compared with control mice (n = 7) pretreated in parallel with a control adenovirus. Metastatic growth-related liver weight gain was also inhibited up to 90%. AdmATF-specific capability that offers liver resistance to the apparition and growth of liver metastasis was shown to correlate with the inhibition of peritumoral and intratumoral angiogenesis (reduced by 79%, p < 0.01 as shown by anti-vWF immunostaining of liver sections) and a twofold increase in tumor necrotic area and an eightfold increase in apoptotic tumor cell number. This protective effect was still observed when the mice were challenged 10 days after venous AdmATF injection (visible metastasis nodules: 6.3+/-3.1, n = 7 for control mice versus 2.7+/-2.9, n = 10 for treated mice, p < 0.05). More importantly, the mean survival has been prolonged from 45.1 days (n = 9) to 83.3 days (n = 10, p < 0.05). Altogether, the high efficacy, although transient, in this experimental mice model strongly advocates the plausibility of transforming the liver into a dissemination resistant organ by antiangiogenic gene therapy through systemic delivery approach.


Assuntos
Adenoviridae/genética , Neoplasias Hepáticas Experimentais/prevenção & controle , Neoplasias Hepáticas Experimentais/secundário , Neovascularização Patológica/genética , Fragmentos de Peptídeos/administração & dosagem , Ativador de Plasminogênio Tipo Uroquinase/administração & dosagem , Animais , Feminino , Imuno-Histoquímica , Neoplasias Hepáticas Experimentais/irrigação sanguínea , Camundongos , Camundongos Nus , Fragmentos de Peptídeos/genética , Análise de Sobrevida , Ativador de Plasminogênio Tipo Uroquinase/genética
19.
Cancer Gene Ther ; 8(10): 759-70, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11687899

RESUMO

The loss of BRCA1 function appears as an essential step in breast and ovarian epithelial cells oncogenesis and is consistent with the concept that BRCA1 acts as a tumor suppressor gene. However, the mechanism underlying this activity is not understood. In 1996, a retroviral vector was used for BRCA1 delivery to demonstrate that the transfer of BRCA1 inhibits breast and ovarian cancer cell growth. Since this early observation, the tumor growth inhibitory activity of BRCA1 in vivo has not been further documented. Here we re-address this issue and report experiments designed to evaluate the potential of adenovirus-mediated BRCA1 delivery to suppress the growth of cells with various status of endogenous BRCA1 in comparison with p53 and p21. Delivery of wild-type BRCA1 by an adenovirus vector in breast and ovarian tumor cells, decreases in vitro proliferation and tumorigenicity. Similarly, in vivo administration of BRCA1 provokes tumor growth retardation or regression comparable to that obtained with p53 or p21. The antitumor effect of BRCA1 is not observed upon transfer of a mutant lacking the 542 C-terminal residues. The p53- or p21-mediated antiproliferative activities are likely to bear on their capacity to induce apoptosis and/or interfere with cell cycle checkpoint. By contrast, the data presented here show that neither of these mechanisms can account for the BRCA1-mediated antitumor activity and suggest the activation of an alternative route.


Assuntos
Proteína BRCA1/genética , Ciclinas/genética , Genes Supressores de Tumor/fisiologia , Neoplasias Mamárias Animais/terapia , Neoplasias Ovarianas/terapia , Proteína Supressora de Tumor p53/genética , Adenoviridae/genética , Animais , Apoptose , Proteína BRCA1/metabolismo , Ciclo Celular , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Feminino , Terapia Genética/métodos , Humanos , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Nus , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Deleção de Sequência , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
20.
Clin Liver Dis ; 3(4): 869-81, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11291256

RESUMO

In patients with chronic hepatitis C who have a sustained virologic response to IFN therapy, there is a dramatic effect on the natural history of the disease, with ALT levels becoming normal, histologic activity improving or disappearing, and the progression of fibrosis slowing. A sustained virologic response 6 months after the end of treatment is predictive of a sustained remission 4 years later. From these results, a long-term survival benefit is expected from IFN treatment in patients with an intermediate or rapid rate of fibrosis. For patients with chronic hepatitis C who do not experience a sustained eradication of virus, there is evidence that IFN treatment significantly reduces the viral load and serum ALT level, improves histologic activity, and blocks fibrosis progression, in comparison with the natural history of this disease. Therefore, patients who still have a detectable level of HCV RNA should no longer be considered nonresponders to IFN therapy. Although the number of randomized trials is [figure: see text] small, cumulative data suggest that IFN therapy can reduce the incidence of and the mortality from hepatocellular carcinoma in patients with cirrhosis.


Assuntos
Antivirais/uso terapêutico , Carcinoma Hepatocelular/mortalidade , Hepatite C Crônica/tratamento farmacológico , Interferons/uso terapêutico , Cirrose Hepática/complicações , Neoplasias Hepáticas/mortalidade , Alanina Transaminase/sangue , Hepatite C Crônica/complicações , Hepatite C Crônica/virologia , Humanos , RNA Viral/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA