Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Oncogene ; 34(41): 5295-301, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-25619840

RESUMO

Oncolytic adenoviruses, such as Delta-24-RGD (Δ24RGD), are replication-competent viruses that are genetically engineered to induce selective cancer cell lysis. In cancer cells, Δ24RGD induces massive autophagy, which is required for efficient cell lysis and adenoviral spread. Understanding the cellular mechanisms underlying the regulation of autophagy in cells treated with oncolytic adenoviruses may provide new avenues to improve the therapeutic effect. In this work, we showed that cancer cells infected with Δ24RGDundergo autophagy despite the concurrent activation of the AKT/mTOR pathway. Moreover, adenovirus replication induced sustained activation of JNK proteins in vitro. ERK1/2 phosphorylation remained unchanged during adenoviral infection, suggesting specificity of JNK activation. Using genetic ablation and pharmacological inactivation of JNK, we unequivocally demonstrated that cells infected with Δ24RGD required JNK activation. Thus, genetic co-ablation of JNK1 and JNK2 genes or inhibition of JNK kinase function rendered Δ24RGD-treated cells resistant to autophagy. Accordingly, JNK activation induced phosphorylation of Bcl-2 and prevented the formation of Bcl-2/Beclin 1 autophagy suppressor complexes. Using an orthotopic model of human glioma xenograft, we showed that treatment with Δ24RGD induced phosphorylation and nuclear translocation of JNK, as well as phosphorylation of Bcl-2. Collectively, our data identified JNK proteins as an essential mechanistic link between Δ24RGD infection and autophagy in cancer cells. Activation of JNK without inactivation of the AKT/mTOR pathway constitutes a distinct molecular signature of autophagy regulation that differentiates Δ24RGD adenovirus from the mechanism used by other oncolytic viruses to induce autophagy and provides a new rationale for the combination of oncolytic viruses and chemotherapy.


Assuntos
Adenoviridae/fisiologia , Autofagia , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Vírus Oncolíticos/fisiologia , Linhagem Celular , Humanos , Terapia Viral Oncolítica , Transdução de Sinais
2.
Br J Cancer ; 108(10): 2005-12, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-23632480

RESUMO

BACKGROUND: Current evidence indicates that a stem cell-like sub-population within malignant glioblastomas, that overexpress members of the adenosine triphosphate-binding cassette (ABC) family transporters, is responsible for multidrug resistance and tumour relapse. Eradication of the brain tumour stem cell (BTSC) compartment is therefore essential to achieve a stable and long-lasting remission. METHODS: Melatonin actions were analysed by viability cell assays, flow cytometry, quantitative PCR for mRNA expression, western blot for protein expression and quantitative and qualitative promoter methylation methods. RESULTS: Combinations of melatonin and chemotherapeutic drugs (including temozolomide, current treatment for malignant gliomas) have a synergistic toxic effect on BTSCs and A172 malignant glioma cells. This effect is correlated with a downregulation of the expression and function of the ABC transporter ABCG2/BCRP. Melatonin increased the methylation levels of the ABCG2/BCRP promoter and the effects on ABCG2/BCRP expression and function were prevented by preincubation with a DNA methyltransferase inhibitor. CONCLUSION: Our results point out a possible relationship between the downregulation of ABCG2/BCRP function and the synergistic toxic effect of melatonin and chemotherapeutic drugs. Melatonin could be a promising candidate to overcome multidrug resistance in the treatment of glioblastomas, and thus improve the efficiency of current therapies.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Neoplasias Encefálicas/patologia , Metilação de DNA/efeitos dos fármacos , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioblastoma/patologia , Melatonina/farmacologia , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/efeitos dos fármacos , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/fisiologia , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Metilação de DNA/fisiologia , Avaliação Pré-Clínica de Medicamentos , Resistência a Múltiplos Medicamentos/genética , Resistencia a Medicamentos Antineoplásicos/genética , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/genética , Humanos , Melatonina/administração & dosagem , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/fisiologia , Regiões Promotoras Genéticas/efeitos dos fármacos
3.
Cancer Gene Ther ; 17(4): 289-97, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19876078

RESUMO

The fiber-modified adenoviral vector Delta-24-RGD (D24RGD) offers vast therapeutic potential. Direct injection of D24RGD has been used to successfully target ovarian tumors in mice. However, systemic toxicity, especially in the liver, profoundly limits the efficacy of direct viral vector delivery. Mesenchymal stem cells (MSC) have the ability to function as a vector for targeted gene therapy because of their preferential engraftment into solid tumors and participation in tumor stroma formation. We show that MSC-guided delivery of D24RGD is specific and efficient and reduces the overall systemic toxicity in mice to negligible levels compared with D24RGD alone. In our model, we found efficient targeted delivery of MSC-D24RGD to both breast and ovarian cell lines. Furthermore, immunohistochemical staining for adenoviral hexon protein confirmed negligible levels of systemic toxicity in mice that were administered MSC-D24RGD compared with those that were administered D24RGD. These data suggest that delivery of D24RGD through MSC not only increases the targeted delivery efficiency, but also reduces the systemic exposure of the virus, thereby reducing overall systemic toxicity to the host and ultimately enhancing its value as an anti-tumor therapeutic candidate.


Assuntos
Adenoviridae/genética , Vetores Genéticos/uso terapêutico , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/virologia , Terapia Viral Oncolítica , Replicação Viral , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/terapia , Neoplasias da Mama/virologia , Linhagem Celular Tumoral , Feminino , Humanos , Técnicas Imunoenzimáticas , Melanoma Experimental/genética , Melanoma Experimental/terapia , Melanoma Experimental/virologia , Camundongos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/terapia , Neoplasias Ovarianas/virologia , Taxa de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Oncogene ; 28(24): 2358-63, 2009 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-19421150

RESUMO

Resistance and relapse are still primary causes that result in poor effectiveness of chemotherapy in malignant gliomas. Therefore, development of new therapeutic strategies requires the identification of key molecular pathways regulating chemoresistance. We previously found that abnormal high expression of the Tie2 receptor in gliomas was associated with tumor malignancy. Here, we studied the role of Tie2 activation in drug resistance by testing the cytotoxicity of several chemotherapeutic drugs in a panel of human glioma cell lines and brain tumor stem cells and found that Tie2 activation was significantly related to chemoresistance. The essential role of Tie2 in this phenotype was illustrated by silencing Tie2 using specific siRNA, and the subsequent abrogation of the angiopoietin 1 (Ang1)-mediated chemoresistance. Using quantitative real-time PCR and functional drug efflux studies, we observed that Tie2 activation resulted in increased expression of ATP-binding cassette (ABC) transporters. Consistent with these results, downmodulation of ABCG2 or ABCC2 resulted in the inability of Tie2 activation to induce a chemoresistant phenotype. Our results indicate that Tie2 activation may be important in modifying the evolution of gliomas during conventional chemotherapy regimens, and open new avenues for the search of more effective therapies to avoid the inevitable brain tumor recurrence.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Receptor TIE-2/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Antineoplásicos/farmacologia , Western Blotting , Camptotecina/análogos & derivados , Camptotecina/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/farmacologia , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Glioma/metabolismo , Glioma/patologia , Humanos , Concentração Inibidora 50 , Irinotecano , Mitoxantrona/farmacologia , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , RNA Interferente Pequeno/genética , Receptor TIE-2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Regulação para Cima
5.
Cancer Gene Ther ; 16(3): 256-65, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18927600

RESUMO

The fact that glioblastomas, which are one of the most devastating cancers, frequently express the Delta-EGFR (epithelial growth factor receptor) also called mutant variant III of EGFR (EGFRvIII) suggests that this cancer cell-specific receptor might serve as an ideal target for cancer therapy. To assess its potential as such a target, we constructed an oncolytic adenovirus with Retargeted Infectivity Via EGFR (Delta-24-RIVER) on the backbone of Delta-24. This new oncolytic adenovirus targets, as Delta-24 does, the disrupted Rb pathway in cancer cells; in addition, this adenovirus has also been retargeted through the abrogation of CAR binding (Y477A mutation in adenoviral fiber protein) and insertion of an EGFRvIII-specific binding peptide in the HI loop of the fiber protein. As compared with Delta-24, Delta-24-RIVER induced EGFRvIII-selective cytotoxicity in U-87 MG isogenic cell lines and in tetracycline-inducible EGFRVIII expressing U-251 MG cells. Accordingly, by tittering the viral progeny and examining fiber protein expression in the above cells, we showed that the replication of this new construct also correlated with EGFRvIII expression. Consistently, immunohistochemistry staining of the adenoviral capsid protein hexon in the virus-treated tumors revealed that the virus replicated more efficiently in EGFRvIII-expressing U-87 MG.DeltaEGFR xenografts than in the tumors grown from U-87 MG cells. Importantly, treatment with Delta-24-RIVER prolonged the survival of animals with intracranial xenografts derived from U-87 MG.DeltaEGFR cells. Therefore, our results constitute the first proof of the direct targeting of a cancer-specific receptor using an oncolytic adenovirus.


Assuntos
Adenovírus Humanos/fisiologia , Neoplasias Encefálicas/terapia , Receptores ErbB/antagonistas & inibidores , Vetores Genéticos/uso terapêutico , Glioblastoma/terapia , Proteínas de Neoplasias/antagonistas & inibidores , Terapia Viral Oncolítica , Adenovírus Humanos/genética , Sequência de Aminoácidos , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral/transplante , Éxons/genética , Genes do Retinoblastoma , Genes erbB-1 , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Deleção de Sequência , Replicação Viral
6.
Histol Histopathol ; 23(6): 773-80, 2008 06.
Artigo em Inglês | MEDLINE | ID: mdl-18366015

RESUMO

The tyrosine kinase receptor Tie2 was initially identified as a specific vascular growth factor that governed several properties of endothelial cells under both physiological and pathological conditions. It was subsequently found that angiopoietins, the natural ligands of Tie2, modulate Tie2-dependent signaling, which in turn regulates the survival and apoptosis of endothelial cells, controls vascular permeability, and regulates the capillary sprouting that occurs during normal angiogenesis such as through development and ovarian remodeling. Tie2 also seems to play a crucial role in several vascular abnormalities, such as familial venous malformations. Beyond its critical role in angiogenesis, Tie2 also appears to maintain the long-term population and quiescent status of hematopoietic stem cells in the bone marrow stem cell niche. In cancer, Tie2 was originally found to be overexpressed in tumoral vessels. More recently, our laboratory and others have found that Tie2 is also expressed outside the vascular compartment in several types of cancer, including leukemia and solid neoplasms such as gastric tumors, breast tumors, and gliomas. The role of Tie2 in these tumoral cells is currently being explored. In this regard, our group reported the importance of Tie2-expressing glioma cells in their adhesion to the tumoral microenvironment. Because cancer may be considered as a complex organ with several cellular lineages coexisting in the same tumor, the expression of Tie2 by different tumoral compartments makes this cellular receptor an attractive target for cancer therapy.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias/irrigação sanguínea , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica/fisiologia , Receptor TIE-2/fisiologia , Animais , Humanos , Transdução de Sinais
7.
Oncogene ; 27(9): 1310-4, 2008 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-17704802

RESUMO

Angiogenesis is thought to depend on a perfectly coordinated balance between endogenous-positive and negative regulatory factors. Of these factors, the vascular endothelial growth factor (VEGF) and angiopoietins (Angs) seem to play an essential role. Recently, we reported the expression of the Ang-natural receptor, Tie2, in neoplastic astrocytic cells within gliomas. Because of the VEGF/Ang2 functional partnership together with the presence of Tie2 in gliomas, we hypothesized a role of Ang2 on the modulation of VEGF levels in these tumors. We examined the effect of Ang2 on VEGF expression in a panel of glioma cells, which showed that Ang2 inhibited VEGF expression at both mRNA and protein levels in Tie2-expressing cells, but not in Tie2-negative cells. VEGF promoter analysis showed that Ang2 regulated VEGF expression at the transcriptional level in relation to a decrease in HIF-1alpha expression and HIF-DNA-binding activity. Tie2 silencing by siRNA rescued the Ang2-mediated downmodulation of VEGF, suggesting an essential role for Tie2 in this regulatory loop. To our knowledge, this is the first report on the role of Ang2/Tie2 in the regulation of HIF-1alpha/VEGF expression, providing additional evidence of the intrinsic coordination that occurs among these factors during angiogenesis.


Assuntos
Angiopoietina-2/fisiologia , Glioma/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/biossíntese , Linhagem Celular Tumoral , DNA de Neoplasias/metabolismo , Glioma/genética , Humanos , Ligantes , Ligação Proteica/genética , Receptor TIE-2/biossíntese , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética
8.
Cancer Gene Ther ; 14(8): 756-61, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17557108

RESUMO

Novel therapies are clearly needed for gliomas, and the combination of oncolytic vectors with chemotherapy possesses a significant hope for the treatment of this malignancy. In addition, combination with chemotherapy allows for lower virus doses to achieve anticancer effect, thus resulting in lower undesirable toxicities due to viral proteins. In this work, we sought to determine whether combination of an oncolytic adenovirus ICOVIR-5, with RAD001 or temozolomide (TMZ) could result in enhanced anti-glioma effect in vivo. We assessed the in vitro cytotoxic effect and replication properties of ICOVIR-5 in combination with RAD001 or TMZ in U87 MG glioma cell line by MTT and TCID(50), respectively. Our data showed that in vitro treatment with RAD001 or TMZ not only interfered with adenovirus replication but, in addition, enhanced its oncolytic properties. To evaluate the in vivo anticancer effect, athymic mice bearing glioma xenografts (5 x 10(5) U87 MG cells/animal) received a single intratumoral injection of ICOVIR-5 (10(7) PFU/animal). RAD001 was given as a regimen of 5 mg/kg 5 days per week until the end of the experiment and TMZ was administered for 5 days at 7.5 mg/kg/mice. Of significance, combination of ICOVIR-5 with RAD001 or TMZ showed a potent anti-glioma effect in vivo, resulting in a dramatic extension of the median animal survival and in 20-40% animals becoming free of disease beyond 90 days.


Assuntos
Adenoviridae , Antineoplásicos Alquilantes/farmacologia , Dacarbazina/análogos & derivados , Glioma/tratamento farmacológico , Terapia Viral Oncolítica , Sirolimo/análogos & derivados , Animais , Linhagem Celular Tumoral , Dacarbazina/farmacologia , Everolimo , Glioma/terapia , Humanos , Camundongos , Camundongos Nus , Neoplasias Experimentais/tratamento farmacológico , Sirolimo/farmacologia , Temozolomida
9.
Cancer Res ; 61(18): 6693-7, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11559537

RESUMO

Strong evidence exists to support the tenet that activation of E2F transcription factors, via alterations in the p16-cyclin D-Rb pathway, is a key event in the malignant progression of most human malignant gliomas. The oncogenic ability of E2F has been related to the E2F-mediated up-regulation of several proteins that positively regulate cell proliferation. However, E2F may indirectly enhance proliferation by activating antiapoptotic molecules. In this work, we sought to ascertain whether E2F-1-mediated events involve the up-regulation of the antiapoptotic molecule Bcl-2. Western blot analyses showed up-regulation of Bcl-2 but not of Bcl-x(L) by 24 h after the transfer of E2F-1. Northern blot studies showed that transfer of E2F-1 also up-regulated Bcl-2 RNA. In support of these findings and the concept that E2F-1 has a direct effect in the induction of Bcl-2, we found a putative E2F binding site within the Bcl-2 sequence. Subsequent gel-mobility shift and supershift experiments involving the CTCCGCGC site in the bcl-2 promoter showed that E2F-1 bound Bcl-2. Transactivation experiments consistently showed that ectopic E2F-1 activated responsive elements located in the -1448/-1441 region in the P1 promoter region of the bcl-2 gene. As expected, other members of the E2F family of transcription factors such as E2F-2 and E2F-4 also transactivated the bcl-2 promoter. Our results demonstrate that E2F-1 modulates the expression of the antiapoptotic molecule Bcl-2 and suggest that up-regulation of Bcl-2 may favor the oncogenic role of E2F-1 and other members of the E2F family of transcription factors.


Assuntos
Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Glioma/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Fatores de Transcrição/genética , Ativação Transcricional , Sítios de Ligação , Ciclo Celular/fisiologia , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Fator de Transcrição E2F2 , Fator de Transcrição E2F4 , Regulação Neoplásica da Expressão Gênica , Técnicas de Transferência de Genes , Genes bcl-2/genética , Glioma/metabolismo , Humanos , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Fatores de Transcrição/biossíntese , Regulação para Cima
10.
Int J Oncol ; 19(2): 359-65, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11445852

RESUMO

The therapeutic efficacy of standard cancer treatments such as chemotherapy may be improved if they are combined with gene-therapy. Less than 30% of patients with glioblastoma multiforme respond to adjuvant chemotherapy. Actively dividing cells are generally more sensitive to chemotherapy than are non-dividing cells. To determine whether forced cell-cycle progression selectively sensitizes tumor cells to alkylating agents, we examined the effects of overexpressing the E2F-1 protein (a positive regulator of cell-cycle progression) on the sensitivity of two malignant human glioma cell lines, U-251 MG and D-54 MG, to BCNU and temozolomide. Treating these cells with 20-35 microM BCNU or 20-30 microM temozolomide resulted in 50% growth inhibition (IC50) within 4 or 6 days, respectively. By contrast, cells that were first induced to overexpress E2F-1 protein by infection with an adenoviral vector had IC50s that were 37-50% lower. Conversely, transferring the cyclin-dependent kinase inhibitors p16 and p21 to the cells, also by adenoviral infection, produced 3 to 4-fold increases in chemoresistance. Cell-cycle analyses showed that the combination of E2F-1 overexpression and treatment with BCNU or temozolomide increased the proportion of cells in S phase, but the combination of p16 or p21 overexpression and drug treatment reduced the proportion of cells in S phase. These observations suggest that overexpression of genes that positively control cell-cycle progression may be useful for increasing the sensitivity of glioma cells to alkylating agents.


Assuntos
Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Dacarbazina/farmacologia , Glioma/patologia , Fatores de Transcrição/genética , Adenoviridae/genética , Antineoplásicos Alquilantes/farmacologia , Carmustina/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/efeitos dos fármacos , Ciclinas/metabolismo , Dacarbazina/análogos & derivados , Relação Dose-Resposta a Droga , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Técnicas de Transferência de Genes , Glioma/metabolismo , Glioma/terapia , Humanos , Concentração Inibidora 50 , Sensibilidade e Especificidade , Temozolomida , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
11.
J Neurooncol ; 51(3): 277-87, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11407598

RESUMO

Current therapies for glioblastoma multiforme are ineffective. Therefore, novel therapies that target specific differences between normal and malignant cells are urgently needed. Abnormalities of cell-cycle related genes are a common feature of cancer in general and astrocytic tumors in particular. The role of these proteins is to help to regulate cell proliferation, differentiation and apoptosis. Restoring wild-type activity of critical regulators of the cell cycle to astrocytic tumors generally results in modification of the growth properties, and often the viability, of the cancer cells. Transfer of p53 induces growth arrest and, more importantly, apoptosis. Restoration of the Rb pathway results in either reversible growth arrest or senescence. Expression of E2F-1 induces transient increase of proliferation followed by massive apoptosis. Overexpression of MMAC/PTEN arrests cell cycle progression in G1 and promotes anoikis. Current knowledge of the functions of these cell-cycle controllers can be used to design small peptides and drugs to induce cell-cycle related anti-cancer effect. Inactivation of the p53 and Rb pathways in cancer cells is also being used to engineer mutant viruses that are able to replicate exclusively in cancer cells.


Assuntos
Astrocitoma/patologia , Proteínas de Ciclo Celular , Ciclo Celular/genética , Proteínas de Ligação a DNA , Técnicas de Transferência de Genes , Proteínas Supressoras de Tumor , Animais , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Genes p53 , Humanos , Família Multigênica , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/genética , Proteína do Retinoblastoma/genética , Fatores de Transcrição/genética
12.
Neurologia ; 16(3): 122-7, 2001 Mar.
Artigo em Espanhol | MEDLINE | ID: mdl-11333782

RESUMO

The transfer of genetic material as a therapy (gene therapy) is one of the experimental treatments being considered in patients with brain tumors resistant to any conventional treatment. Several clinical trials have proved that the intratumoral administration of genes is fairly safe for patients, however the anti-tumor effect of these strategies remains suboptimal. One of the main problems in cancer gene therapy is the failure of current vectors to achieve enough tumor transduction in a suficient number of cells. This is even true for vectors derived from viruses with high infectivity ability such as adenovirus. For that reason, current strategies explore the use of adenoviruses able to replicate and spread throughout the tumor. The local, intratumoral injection of adenovirus is an especially suitable strategy for gliomas because these tumors, although infiltrative, rarely metastasize. Two approaches have been used to generate tumor-selective replicative adenoviruses: use of tumor-specific promoters to regulate the expression of viral genes, and the deletion of the viral functions required for the activation of the cell cycle. Since normal cells surrounding giomas are quiescent, the second strategy is particularly attractive to develop new treatments for brain tumors.


Assuntos
Adenoviridae , Neoplasias Encefálicas/terapia , Terapia Genética/tendências , Adenoviridae/genética , DNA Viral/genética , Previsões , Terapia Genética/métodos , Humanos , Sequências Reguladoras de Ácido Nucleico/genética , Proteína do Retinoblastoma/genética , Proteína Supressora de Tumor p53/genética
13.
Br J Cancer ; 84(9): 1252-7, 2001 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-11336478

RESUMO

Increased expression of VEGF in several types of tumours has been shown to correlate with poor prognosis. We used a replication-deficient adenoviral vector containing antisense VEGF cDNA (Ad5CMV-alphaVEGF) to down-regulate VEGF expression and increase the efficiency of delivery of the antisense sequence in the human breast cancer cell line MDA231-MB. Transfection of these cells with Ad5CMV-alphaVEGF in vitro reduced secreted levels of VEGF protein without affecting cell growth. Moreover, injection of the Ad5CMV-alphaVEGF vector into intramammary xenografts of these cells established in nude mice inhibited tumour growth and reduced the amount of VEGF protein and the density of microvessels in those tumours relative to tumours treated with the control vector Ad5(dl312). Our results showed that antisense VEGF(165)cDNA was efficiently delivered in vivo via an adenoviral vector and that this treatment significantly inhibited the growth of established experimental breast tumours. The Ad5CMV-alphaVEGF vector may be useful in targeting the tumour vasculature in the treatment of breast cancer.


Assuntos
Elementos Antissenso (Genética) , Neoplasias da Mama/patologia , Fatores de Crescimento Endotelial/genética , Terapia Genética , Linfocinas/genética , Neovascularização Patológica , Adenoviridae/fisiologia , Inibidores da Angiogênese/farmacologia , Neoplasias da Mama/irrigação sanguínea , Fatores de Crescimento Endotelial/farmacologia , Feminino , Vetores Genéticos , Humanos , Linfocinas/farmacologia , Transfecção , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
14.
Neurología (Barc., Ed. impr.) ; 16(3): 122-127, mar. 2001.
Artigo em Es | IBECS | ID: ibc-3324

RESUMO

La transferencia de material genético con fines terapéuticos (terapia génica) es uno de los tratamientos experimentales a considerar en pacientes con tumores cerebrales incurables mediante tratamientos convencionales. Aunque diferentes estudios clínicos han demostrado que la administración intratumoral de genes no produce una toxicidad intolerable para el paciente, todavía no se ha conseguido una eficacia antitumoral significativa. Uno de los principales problemas de la terapia génica del cáncer es la imposibilidad de los vectores actuales para introducir el gen terapéutico en un número suficiente de células del tumor, incluso utilizando vectores derivados de virus de alta infectividad, como el adenovirus.Para solventar esta dificultad se están utilizando virus, y sus vectores derivados, con capacidad de replicarse y extenderse a través del tumor. Las características infiltrantes pero raramente metastásicas y multifocales de los tumores cerebrales ofrecen una oportunidad única para utilizar este tipo de terapia con inyección local intratumoral. Para conseguir adenovirus capaces de replicarse en células cancerosas pero no en células normales se han utilizado principalmente dos estrategias: la regulación de la expresión de genes virales con promotores específicos de tumor y la deleción de funciones virales necesarias para activar el ciclo celular. El estado quiescente de las células normales que rodean a los gliomas hacen que esta última estrategia sea especialmente atractiva para este tipo de tumores. (AU)


Assuntos
Humanos , Adenoviridae , Terapia Genética , Proteína do Retinoblastoma , DNA Viral , Previsões , Proteína Supressora de Tumor p53 , Sequências Reguladoras de Ácido Nucleico , Neoplasias Encefálicas
15.
Int J Oncol ; 18(2): 343-7, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11172602

RESUMO

Gliomas are highly resistant to conventional treatment. Improved knowledge of the molecular defects of glioma cells offers new avenues for the development of gene therapy strategies. Transfer of the p53 gene has proven effective in suppressing proliferation in human glioma cell lines. However, several human glioma cell lines are resistant to p53-induced cell death. The E2F family of transcription factors are pivotal for the regulation of cell-cycle and cell-death related genes in gliomas. In the present study, we sought a more effective strategy for glioma treatment by examining the therapeutic potential of the simultaneous transfer of p53 and E2F-2 to gliomas. Trypan blue cell viability assays and flow cytometric cell-cycle analysis demonstrated that the transfer of both p53 and E2F-2 induced cell death in D-54 MG, a p53-resistant glioma cell line. In addition, transfer of E2F-2 did not interfere with the apoptotic properties of exogenous wild-type p53 in U-251 MG cells. Finally, the expression of E2F-2 in D-54 MG cells suppressed the expression of the apoptotic molecule mdm-2 induced by exogenous p53 in these cells. These results show that co-expression of E2F-2 and p53 enhances the anti-cancer effect of p53 in gliomas.


Assuntos
Apoptose/fisiologia , Genes p53/fisiologia , Glioblastoma/metabolismo , Glioma/metabolismo , Proteínas Nucleares , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Actinas/metabolismo , Adenoviridae , Fator de Transcrição E2F2 , Glioblastoma/terapia , Glioma/terapia , Humanos , Proteínas Proto-Oncogênicas c-mdm2 , Fatores de Transcrição/genética , Células Tumorais Cultivadas/metabolismo
16.
Oncogene ; 19(1): 2-12, 2000 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-10644974

RESUMO

Effective anti cancer strategies necessitate the use of agents that target tumor cells rather than normal tissues. In this study, we constructed a tumor-selective adenovirus, Delta24, that carries a 24-bp deletion in the E1A region responsible for binding Rb protein. Immunoprecipitation analyses verified that this deletion rendered Delta24 unable to bind the Rb protein. However, titration experiments in 293 cells demonstrated that the Delta24 adenovirus could replicate in and lyse cancer cells with great efficiency. Lysis of most human glioma cells was observed within 10 - 14 days after infection with Delta24 at 10 PFU/cell. In vivo, a single dose of the Delta24 virus induced a 66.3% inhibition (P<0.005) and multiple injections, an 83.8% inhibition (P<0.01) of tumor growth in nude mice. However, normal fibroblasts or cancer cells with restored Rb activity were resistant to the Delta24 adenovirus. These results suggest that the E1A-mutant Delta24 adenovirus may be clinically and therapeutically useful against gliomas and possibly other cancers with disrupted Rb pathway.


Assuntos
Adenoviridae/genética , Terapia Genética , Glioma/terapia , Proteína do Retinoblastoma/fisiologia , Animais , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação , Fase S , Células Tumorais Cultivadas , Replicação Viral
17.
Exp Cell Res ; 252(1): 1-12, 1999 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-10502394

RESUMO

Currently, most of the approved clinical gene therapy protocols involve cancer patients and several of the therapies are designed to treat brain tumors. Two factors promoting the use of gene therapy for gliomas are the failure and toxicity of conventional therapies and the identification of the genetic abnormalities that contribute to the malignancy of gliomas. During the malignant progression of astrocitic tumors several tumor suppressor genes are inactivated, and numerous growth factors and oncogenes are overexpressed progressively. Thus, theoretically, brain tumors could be treated by targeting their fundamental molecular defects, provided the gene-drug can be delivered to a sufficient number of malignant cells. However, gene therapy strategies have not been abundantly successful clinically, in part because the delivery systems are still imperfect. In the first part of this brief review we will discuss the most common targets for gene therapy in brain tumors. In the second part, we will review the evolution of adenoviruses as gene vehicles. In addition, we will examine the role of recombinant mutant oncolytic adenoviruses as anticancer tools. From the results to date it is clear that gene therapy strategies for brain tumors are quite promising but more critical research is required, mainly in the vector field, if the strategies are to achieve their true potential in ameliorating patients with gliomas.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioma/terapia , Adenovírus Humanos/genética , Adenovírus Humanos/fisiologia , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/genética , Ensaios Clínicos como Assunto , Genes Supressores de Tumor , Vetores Genéticos , Glioblastoma/genética , Glioblastoma/terapia , Glioma/genética , Vírus Auxiliares/genética , Humanos , Síndrome , Replicação Viral
18.
Arch Neurol ; 56(4): 445-8, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10199333

RESUMO

Cancer is a disease of a series of genes. Thus, theoretically, brain tumors could be treated by targeting their fundamental molecular defects. Currently, most of the approved clinical protocols for gene therapy involve cancer patients. Several of these protocols are designed to improve the treatment of brain tumors. In this brief report, we analyze the rationale, advantages, and disadvantages of a series of gene therapy approaches against brain tumors that include transfer of tumor suppressor genes and cell-cycle modulators; suicide or prodrug strategies; immunogene therapy; antiangiogenesis; and oncolytic virus therapy. In summary, in this review, we highlight the translational advances in molecular medicine that broaden our battery of therapies for patients with brain tumors.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Proteínas de Transporte , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Terapia Genética/métodos , Glioma/genética , Glioma/terapia , Proteínas Supressoras de Tumor , Neoplasias Encefálicas/fisiopatologia , Ciclo Celular/genética , Fatores de Transcrição E2F , Genes Supressores de Tumor/genética , Genes p53 , Glioma/fisiopatologia , Humanos , Neovascularização Patológica/genética , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/genética , Pró-Fármacos/uso terapêutico , Proteína 1 de Ligação ao Retinoblastoma , Fator de Transcrição DP1 , Fatores de Transcrição/genética , Vírus/genética
19.
Cancer Res ; 59(4): 895-900, 1999 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-10029081

RESUMO

Presently, there is no effective treatment for glioblastoma, the most malignant and common brain tumor. Angiogenic factors are potentially optimal targets for therapeutic strategies because they are essential for tumor growth and progression. In this study, we sought a strategy for efficiently delivering an antisense cDNA molecule of the vascular endothelial growth factor (VEGF) to glioma cells. The recombinant adenoviral vector Ad5CMV-alphaVEGF carried the coding sequence of wild-type VEGF165 cDNA in an antisense orientation. Infection of U-87 MG malignant glioma cells with the Ad5CMV-alphaVEGF resulted in reduction of the level of the endogenous VEGF mRNA and drastically decreased the production of the targeted secretory form of the VEGF protein. Treatment of s.c. human glioma tumors established in nude mice with intralesional injection of Ad5CMV-alphaVEGF inhibited tumor growth. Taken together, these findings indicate that the efficient down-regulation of the VEGF produced by tumoral cells using antisense strategies has an antitumor effect in vivo. This is the first time that an adenoviral vector is used to transfer antisense VEGF sequence into glioma cells in an animal model, and our results suggest that this system may have clinical and therapeutic utility.


Assuntos
DNA Antissenso/farmacologia , Fatores de Crescimento Endotelial/genética , Terapia Genética , Glioma/terapia , Linfocinas/genética , Neovascularização Patológica/terapia , Adenoviridae/genética , Animais , Regulação para Baixo , Glioma/irrigação sanguínea , Humanos , Camundongos , Camundongos Nus , RNA Mensageiro/análise , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
20.
Int J Mol Med ; 3(1): 81-5, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9864390

RESUMO

Current therapy for glioma is suboptimal. The transfer of apoptosis genes to tumors constitutes one of the most promising strategies for cancer gene therapy. We have previously shown that massive apoptosis occurs when wild-type p53 or E2F-1 expression is induced in glioma. However, the mechanism of action and the efficiency in inducing apoptosis of these two proteins are not similar. Adenovirus-mediated p53 gene transfer is ineffective in causing apoptosis in glioma cells that retain wild-type p53 genotype or overexpress the p21 protein. The p16/Rb/E2F pathway is the most frequent target of genetic alterations in gliomas, and therefore constitutes a suitable target for gene therapy strategies. However, the transfer of either the p16 or Rb gene to glioma cells results in cytostatic effect. The E2F-1 protein is able to induce generalized apoptosis in gliomas independently of the p53, p16 or Rb status. In addition, p21- or p16-mediated growth arrest did not protect glioma cells from E2F-1-mediated apoptosis. The apoptotic molecule bax is induced in p53-mediated apoptosis, but bax is not induced in E2F-1-mediated apoptosis in glioma cells. Careful selection of patients may be necessary before designing therapeutic strategies using either p53 or E2F-1 as a therapeutic tools for glioma patients.


Assuntos
Proteínas de Transporte , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Terapia Genética , Glioma/genética , Apoptose/genética , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Regulação Neoplásica da Expressão Gênica , Glioma/terapia , Humanos , Proteína 1 de Ligação ao Retinoblastoma , Fator de Transcrição DP1 , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...