Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Int Rev Cytol ; 257: 181-212, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17280898

RESUMEN

Hypoxia is an integral characteristic of the tumor microenvironment, primarily due to the microvascular defects that accompany the accelerated neoplastic growth. The presence of tumor hypoxic areas correlates with negative outcome after radiotherapy, chemotherapy, and surgery, as hypoxia not only provides an environment directly facilitating chemo- and radio-resistance, but also encourages the evolution of phenotypic changes inducing permanent resistance to treatment and metastatic spread. Therefore, successful treatment of hypoxic cells has the potential to not only improve local control but also impact overall patient survival. Specific and selective targeting of hypoxic tumor areas can be achieved at all three steps of a gene therapy treatment: delivery of the therapeutic gene to the tumor, regulation of gene expression, and therapeutic efficacy. In this review the latest developments and innovations in hypoxia-targeted gene therapy are discussed. In particular, approaches such as hypoxia-conditionally replicating viruses, cellular vehicles, and gene therapy means to disrupt the hypoxia-inducible factor (HIF) signaling are outlined.


Asunto(s)
Terapia Genética , Neoplasias/patología , Neoplasias/terapia , Animales , Hipoxia de la Célula , Terapia Combinada , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias/genética
2.
Int J Radiat Oncol Biol Phys ; 69(5): 1553-62, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17920784

RESUMEN

PURPOSE: To investigate the effects of radiation on the endothelial cytoskeleton and endothelial monolayer permeability and to evaluate associated signaling pathways, which could reveal potential mechanisms of known vascular effects of radiation. METHODS AND MATERIALS: Cultured endothelial cells were X-ray irradiated, and actin filaments, microtubules, intermediate filaments, and vascular endothelial (VE)-cadherin junctions were examined by immunofluorescence. Permeability was determined by the passage of fluorescent dextran through cell monolayers. Signal transduction pathways were analyzed using RhoA, Rho kinase, and stress-activated protein kinase-p38 (SAPK2/p38) inhibitors by guanosine triphosphate-RhoA activation assay and transfection with RhoAT19N. The levels of junction protein expression and phosphorylation of myosin light chain and SAPK2/p38 were assessed by Western blotting. The radiation effects on cell death were verified by clonogenic assays. RESULTS: Radiation induced rapid and persistent actin stress fiber formation and redistribution of VE-cadherin junctions in microvascular, but not umbilical vein endothelial cells, and microtubules and intermediate filaments remained unaffected. Radiation also caused a rapid and persistent increase in microvascular permeability. RhoA-guanosine triphosphatase and Rho kinase were activated by radiation and caused phosphorylation of downstream myosin light chain and the observed cytoskeletal and permeability changes. SAPK2/p38 was activated by radiation but did not influence either the cytoskeleton or permeability. CONCLUSION: This study is the first to show rapid activation of the RhoA/Rho kinase by radiation in endothelial cells and has demonstrated a link between this pathway and cytoskeletal remodeling and permeability. The results also suggest that the RhoA pathway might be a useful target for modulating the permeability and other effects of radiation for therapeutic gain.


Asunto(s)
Citoesqueleto/efectos de la radiación , Células Endoteliales/efectos de la radiación , Actinas/efectos de la radiación , Cadherinas/efectos de la radiación , Muerte Celular/efectos de la radiación , Células Cultivadas , Dextranos/farmacocinética , Células Endoteliales/ultraestructura , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Endotelio Vascular/efectos de la radiación , Humanos , Filamentos Intermedios/efectos de la radiación , Microtúbulos/efectos de la radiación , Proteína Quinasa 11 Activada por Mitógenos/fisiología , Permeabilidad , Tolerancia a Radiación , Transducción de Señal/fisiología , Transducción de Señal/efectos de la radiación , Proteína de Unión al GTP rhoA/fisiología
3.
Cancer Gene Ther ; 12(7): 655-62, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15818381

RESUMEN

Combining gene therapy with radiotherapy and chemotherapy holds potential to increase the efficacy of cancer treatment, while minimizing side effects. We tested the responsiveness of synthetic gene promoters containing CArG elements from the Early Growth Response 1 (Egr1) gene after neutron irradiation, doxorubicin and cisplatin. Human MCF-7 breast adenocarcinoma and U373-MG glioblastoma cells were transfected with plasmids containing CArG promoters controlling the expression of the green fluorescent protein (GFP). Exposing the cells to neutrons, doxorubicin or cisplatin resulted in a significant induction of transgene expression. Therapeutic advantage was demonstrated by replacing the reporter with the herpes simplex virus thymidine kinase (HSVtk), able to convert the prodrug ganciclovir (GCV) into a cytotoxin. A 1.3 Gy neutron dose caused 49% growth inhibition in MCF-7 cells, which increased to 63% in irradiated CArG-HSVtk-transfectants treated with GCV. Exposure to 0.5 microM cisplatin or 0.01 microM doxorubicin induced a growth inhibition of 25-30% in MCF-7 cells. In the presence of GCV, this value increased to 65-70% in cells transfected with the CArG promoter constructs driving the expression of HSVtk. These data indicate that combining CArG-mediated HSVtk/GCV suicide gene therapy with radio- and chemotherapy can enhance antitumor toxicity, and validates future in vivo investigations.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/terapia , Neoplasias de la Mama/terapia , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/efectos de los fármacos , Terapia Genética/métodos , Vectores Genéticos , Proteínas Inmediatas-Precoces/genética , Factores de Transcripción/genética , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/radioterapia , Adenocarcinoma/terapia , Antivirales/uso terapéutico , Apoptosis , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/radioterapia , Cisplatino/farmacología , Terapia Combinada , Doxorrubicina/farmacología , Proteína 1 de la Respuesta de Crecimiento Precoz , Ganciclovir/metabolismo , Técnicas de Transferencia de Gen , Glioblastoma/tratamiento farmacológico , Glioblastoma/radioterapia , Glioblastoma/terapia , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Neutrones , Regiones Promotoras Genéticas , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Elementos de Respuesta/genética , Simplexvirus/enzimología , Simplexvirus/genética , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Células Tumorales Cultivadas , Dedos de Zinc
4.
Int J Radiat Oncol Biol Phys ; 62(1): 213-22, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15850924

RESUMEN

PURPOSE: Tumor hypoxia is unequivocally linked to poor radiotherapy outcome. This study aimed to identify enhancer sequences that respond maximally to a combination of radiation and hypoxia for use in genetic radiotherapy approaches. METHODS AND MATERIALS: The influence of radiation (5 Gy) and hypoxia (1% O2) on reporter-gene expression driven by hypoxia (HRE) and radiation (Egr-1) responsive elements was evaluated in tumor cells grown as monolayers or multicellular spheroids. Hypoxia-inducible factor-1alpha (HIF-1alpha) and HIF-2alpha protein expression was monitored in parallel. RESULTS: Of the sequences tested, an HRE from the phosphoglycerate kinase-1 gene (PGK-18[5+]) was maximally induced in response to hypoxia plus radiation in all 5 cell lines tested. The additional radiation treatment afforded a significant increase in the induction of PGK-18[5+] compared with hypoxia alone in 3 cell lines. HIF-1alpha/2alpha were induced by radiation but combined hypoxia/radiation treatment did not yield a further increase. The dual responsive nature of HREs was maintained when spheroids were irradiated after delivery of HRE constructs in a replication-deficient adenovirus. CONCLUSIONS: Hypoxia-responsive enhancer element sequences are dually responsive to combined radiation and hypoxic treatment. Their use in genetic radiotherapy in vivo could maximize expression in the most radio-resistant population at the time of radiation and also exploit microenvironmental changes after radiotherapy to yield additional switch-on.


Asunto(s)
Hipoxia de la Célula/genética , Regulación de la Expresión Génica , Fosfoglicerato Quinasa/genética , Tolerancia a Radiación/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Subunidad alfa del Factor 1 Inducible por Hipoxia , Fosfoglicerato Quinasa/metabolismo , Regiones Promotoras Genéticas , Tolerancia a Radiación/fisiología , Esferoides Celulares , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Cancer Gene Ther ; 11(7): 508-13, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15143345

RESUMEN

Horseradish peroxidase has previously been shown to catalyze the conversion of indole-3-acetic acid (IAA) to a potent cytotoxin in a gene therapy setting. A three-dimensional spheroid model composed of a human head and neck carcinoma cell line, has been used to mimic the tumor microenvironment, such as regions of hypoxia. Exposure of intact spheroids to 0.05-5 mM concentrations of IAA and the halogenated indole, 5-bromoindole-3-acetic acid (5Br-IAA), resulted in decreased cell survival, and demonstrates that this combination is effective under tumor-simulated conditions. In addition, 5Br-IAA, displayed selectivity for spheroids with a large hypoxic fraction following short exposure times.


Asunto(s)
Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Peroxidasa de Rábano Silvestre/metabolismo , Ácidos Indolacéticos/metabolismo , Ácidos Indolacéticos/farmacología , Modelos Biológicos , Técnicas de Cultivo de Célula , Hipoxia de la Célula/fisiología , Supervivencia Celular/efectos de los fármacos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Peroxidasa de Rábano Silvestre/genética , Humanos , Ácidos Indolacéticos/uso terapéutico , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Factores de Tiempo , Transfección , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
6.
Curr Pharm Des ; 9(26): 2131-54, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14529410

RESUMEN

Improvements in the radiotherapeutic management of solid tumors through the concurrent use of gene therapy is a realistic possibility. Of the broad array of candidate genes that have been evaluated, those encoding prodrug-activating enzymes are particularly appealing since they directly complement ongoing clinical chemoradiation regimes. Gene-Directed Enzyme-Prodrug Therapy (GDEPT) only requires a fraction of the target cells to be genetically modified, providing that the resultant cytotoxic prodrug metabolites redistribute efficiently (the bystander effect). This transfer of cytotoxicity to neighboring non-targeted cancer cells is central to the success of any gene therapy strategy, irrespective of the therapeutic gene employed. In the context of genetic chemoradiotherapy, efficient prodrug metabolite diffusion will be a prerequisite for efficient radiosensitization. Some, but not all GDEPT approaches have been analysed in combination with radiotherapy. Examples of prodrugs of clinically established chemotherapeutic agents currently used in conjunction with radiotherapy include: 5-fluorocytosine (5FC), cyclophosphamide (CPA), irinotecan (CPT-11), gemcitabine (dFdC), capecitabine, mitomycin C (MMC) and AQ4N. Other GDEPT paradigms, such as ganciclovir (GCV) and Herpes Simplex thymidine kinase (HSV-tk), dinitrobenzamide (DNB) mustard or aziridinyl analogs and the E. coli nitroreductase (NTR), CMDA or ZP2767P with Pseudomonas aeruginosa carboxypeptidase G2 (CPG2), and indole-3-acetic acid (IAA) activated by horseradish peroxidase (HRP) have no clinically established chemotherapeutic counterpart. Each prodrug is discussed in this review in the context of GDEPT, with a particular attention to translational research and clinical utility in combination with radiotherapy.


Asunto(s)
Terapia Genética/métodos , Neoplasias/terapia , Profármacos/uso terapéutico , Animales , Terapia Combinada , Enzimas/genética , Enzimas/metabolismo , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/radioterapia , Profármacos/química , Fármacos Sensibilizantes a Radiaciones , Radioterapia Adyuvante
7.
Front Biosci ; 7: d1516-24, 2002 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12045003

RESUMEN

Gene therapy for cancer treatment represents a promising approach that has shown selectivity and efficacy in experimental systems as well as clinical trials. Some major problems remain to be solved before this strategy becomes routinely adopted in the clinic, one of the main challenges being the improvement of gene delivery. Namely, the development of DNA vectors characterized by maximum efficiency and minimal toxicity will define the success of gene therapy and its chances of being accepted by public and clinicians. A number of issues need to be considered. The "magic" vector should be targeted, protected from degradation and immune attack, and safe for the recipient and the environment. Moreover, it should express the therapeutic gene for as long as required, in an appropriately regulated fashion. Vehicles such as retroviruses, adenoviruses and liposomes have been adopted in clinical studies, with varying results. New therapeutic modalities are also being explored in order to overcome the limitation of poor gene transfer and patient toxicity, including bacteria, adeno-associated and herpes simplex viruses, lentiviruses, cationic polymer-DNA complexes and electroporation. Some of the delivery systems tested in preclinical and clinical models are reviewed in this article, with particular attention to the targeting of the tumor environment.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Neoplasias/terapia , Animales , Técnicas de Transferencia de Gen/efectos adversos , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Vectores Genéticos/uso terapéutico , Humanos
8.
Biochem Pharmacol ; 63(2): 265-72, 2002 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11841802

RESUMEN

Indole-3-acetic acid and some derivatives are oxidized by horseradish peroxidase, forming a radical-cation that rapidly fragments (eliminating CO(2)) to form cytotoxic products. No toxicity is seen when either indole-3-acetic acid or horseradish peroxidase is incubated alone at concentrations that together form potent cytotoxins. Unexpectedly, 5-fluoroindole-3-acetic acid, which is oxidized by horseradish peroxidase compound I 10-fold more slowly than indole-3-acetic acid, is much more cytotoxic towards V79 hamster fibroblasts in the presence of peroxidase than the unsubstituted indole. The fluorinated prodrug/peroxidase combination also shows potent cytotoxic activity in human and rodent tumor cell lines. Cytotoxicity is thought to arise in part from the formation of 3-methylene-2-oxindole (or analogues) that can conjugate with thiols and probably DNA or other biological nucleophiles. Levels of the fluorinated prodrug in the murine carcinoma NT after intraperitoneal administration of 50 mg/kg were about 200 microM. Although these were 4-5-fold lower than plasma levels (which reached 1mM), the integrated area under the concentration/time curve in tumors over 2 hr was approximately 20 mM min, almost double the exposure needed to achieve approximately 90-99% cell kill in human MCF7 breast or HT29 colon tumor cell lines and CaNT murine cells in vitro, although the human bladder T24 carcinoma cell line was more resistant. The high cytotoxicity of 5-fluoroindole-3-acetic acid after oxidative activation suggests its further evaluation as a prodrug for targeted cancer therapy involving antibody-, polymer-, or gene-directed delivery of horseradish peroxidase or similar activating enzymes.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Indolacéticos/farmacología , Peroxidasas/metabolismo , Profármacos/farmacología , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Femenino , Flúor/metabolismo , Glutatión/metabolismo , Células HT29 , Humanos , Ácidos Indolacéticos/metabolismo , Ácidos Indolacéticos/farmacocinética , Ratones , Ratones Endogámicos CBA , Neoplasias Experimentales/metabolismo , Oxidación-Reducción , Profármacos/metabolismo , Profármacos/farmacocinética
9.
PLoS One ; 9(8): e104015, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25119572

RESUMEN

Vascular endothelial growth factor-A (VEGF) is produced by most cancer cells as multiple isoforms, which display distinct biological activities. VEGF plays an undisputed role in tumour growth, vascularisation and metastasis; nevertheless the functions of individual isoforms in these processes remain poorly understood. We investigated the effects of three main murine isoforms (VEGF188, 164 and 120) on tumour cell behaviour, using a panel of fibrosarcoma cells we developed that express them individually under endogenous promoter control. Fibrosarcomas expressing only VEGF188 (fs188) or wild type controls (fswt) were typically mesenchymal, formed ruffles and displayed strong matrix-binding activity. VEGF164- and VEGF120-producing cells (fs164 and fs120 respectively) were less typically mesenchymal, lacked ruffles but formed abundant cell-cell contacts. On 3D collagen, fs188 cells remained mesenchymal while fs164 and fs120 cells adopted rounded/amoeboid and a mix of rounded and elongated morphologies respectively. Consistent with their mesenchymal characteristics, fs188 cells migrated significantly faster than fs164 or fs120 cells on 2D surfaces while contractility inhibitors accelerated fs164 and fs120 cell migration. VEGF164/VEGF120 expression correlated with faster proliferation rates and lower levels of spontaneous apoptosis than VEGF188 expression. Nevertheless, VEGF188 was associated with constitutively active/phosphorylated AKT, ERK1/2 and Stat3 proteins. Differences in proliferation rates and apoptosis could be explained by defective signalling downstream of pAKT to FOXO and GSK3 in fs188 and fswt cells, which also correlated with p27/p21 cyclin-dependent kinase inhibitor over-expression. All cells expressed tyrosine kinase VEGF receptors, but these were not active/activatable suggesting that inherent differences between the cell lines are governed by endogenous VEGF isoform expression through complex interactions that are independent of tyrosine kinase receptor activation. VEGF isoforms are emerging as potential biomarkers for anti-VEGF therapies. Our results reveal novel roles of individual isoforms associated with cancer growth and metastasis and highlight the importance of understanding their diverse actions.


Asunto(s)
Movimiento Celular/genética , Proliferación Celular/genética , Fibrosarcoma/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis , Carcinogénesis/genética , Adhesión Celular , Línea Celular Tumoral , Supervivencia Celular/genética , Fibrosarcoma/patología , Regulación Neoplásica de la Expresión Génica , Ratones , Metástasis de la Neoplasia/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/química
11.
Cancer Res ; 68(7): 2301-11, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18381437

RESUMEN

Tubulin-binding vascular-disrupting agents (VDA) are currently in clinical trials for cancer therapy but the factors that influence tumor susceptibility to these agents are poorly understood. We evaluated the consequences of modifying tumor vascular morphology and function on vascular and therapeutic response to combretastatin-A4 3-O-phosphate (CA-4-P), which was chosen as a model VDA. Mouse fibrosarcoma cell lines that are capable of expressing all vascular endothelial growth factor (VEGF) isoforms (control) or only single isoforms of VEGF (VEGF120, VEGF164, or VEGF188) were developed under endogenous VEGF promoter control. Once tumors were established, VEGF isoform expression did not affect growth or blood flow rate. However, VEGF188 was uniquely associated with tumor vascular maturity, resistance to hemorrhage, and resistance to CA-4-P. Pericyte staining was much greater in VEGF188 and control tumors than in VEGF120 and VEGF164 tumors. Vascular volume was highest in VEGF120 and control tumors (CD31 staining) but total vascular length was highest in VEGF188 tumors, reflecting very narrow vessels forming complex vascular networks. I.v. administered 40 kDa FITC-dextran leaked slowly from the vasculature of VEGF188 tumors compared with VEGF120 tumors. Intravital microscopy measurements of vascular length and RBC velocity showed that CA-4-P produced significantly more vascular damage in VEGF120 and VEGF164 tumors than in VEGF188 and control tumors. Importantly, this translated into a similar differential in therapeutic response, as determined by tumor growth delay. Results imply differences in signaling pathways between VEGF isoforms and suggest that VEGF isoforms might be useful in vascular-disrupting cancer therapy to predict tumor susceptibility to VDAs.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Fibrosarcoma/irrigación sanguínea , Fibrosarcoma/tratamiento farmacológico , Estilbenos/farmacología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Animales , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibrosarcoma/metabolismo , Inmunohistoquímica , Ratones , Ratones SCID , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Isoformas de Proteínas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Factor A de Crecimiento Endotelial Vascular/genética
12.
Virology ; 349(1): 121-33, 2006 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-16464484

RESUMEN

To facilitate a more efficient radiation and chemotherapy of mammary tumours, synthetic enhancer elements responsive to hypoxia and ionizing radiation were coupled to the mammary-specific minimal promoter of the murine whey acidic protein (WAP) encoding gene. The modified WAP promoter was introduced into a retroviral promoter conversion (ProCon) vector. Expression of a transduced reporter gene in response to hypoxia and radiation was analysed in stably infected mammary cancer cell lines and an up to 9-fold increase in gene expression demonstrated in comparison to the respective basic vector. Expression analyses in vitro, moreover, demonstrated a widely preserved mammary cell-specific promoter activity. For in vivo analyses, xenograft tumours consisting of infected human mammary adenocarcinoma cells were established in SCID/beige mice. Immunohistochemical analyses demonstrated a hypoxia-specific, markedly increased WAP promoter-driven expression in these tumours. Thus, this retroviral vector will facilitate a targeted gene therapeutic approach exploiting the unique environmental condition in solid tumours.


Asunto(s)
Neoplasias de la Mama/terapia , Hipoxia de la Célula , Terapia Genética/métodos , Vectores Genéticos , Proteínas de la Leche/genética , Regiones Promotoras Genéticas , Retroviridae , Animales , Línea Celular , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Humanos , Inmunohistoquímica , Ratones , Ratones SCID , Microscopía Fluorescente , Trasplante de Neoplasias , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de la radiación , Sensibilidad y Especificidad , Trasplante Heterólogo , Rayos X
13.
Cancer Metastasis Rev ; 23(3-4): 269-76, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15197328

RESUMEN

Radiotherapy remains one of the primary treatment modalities for most malignancies. Biologically based improvements in the scheduling of conventional radiotherapy and treatment planning, innovations like conformal radiotherapy and intensity-modulated radiation therapy have considerably improved the targeting and effectiveness of radiation for treatment of solid tumors. These new radiotherapy technologies are also promising means of focusing the activation of anti-tumor gene therapy systems, as an approach to further improve radiotherapeutic treatment, particularly for tumors refractive to current therapies. Gene therapy vectors that express therapeutic genes following irradiation have been produced. Delivery of such vectors to the tumor allows temporal and spatial expression of the transgenes within the radiation field. Hypoxia is a physiological characteristic of solid tumors and an independent prognostic marker for poor radiation treatment outcome. Nevertheless, hypoxia has been exploited to drive therapeutic gene expression from gene therapy vectors delivered to solid tumors exhibiting significant areas of low oxygen tension. Radiation and hypoxia inducible gene therapy systems rely on the activation of gene promoters containing specific responsive elements. Recent studies have shown the potential to combine these elements, permitting either or both stimuli to drive therapeutic gene expression. Furthermore, transgene expression can be amplified and sustained using novel 'signal feedback' or recombination systems. Such innovations allow promising new strategies to improve radiation treatment outcome, particularly where tumor hypoxia is a predominant issue.


Asunto(s)
Hipoxia de la Célula/genética , Terapia Genética/métodos , Neoplasias/genética , Neoplasias/radioterapia , Animales , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/metabolismo , Neoplasias/terapia , Oxígeno/metabolismo
14.
Am J Pathol ; 165(4): 1401-11, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15466404

RESUMEN

The tubulin-binding agent combretastatin A-4-phosphate (CA-4-P), rapidly disrupts the vascular network of tumors leading to secondary tumor cell death. In vitro, CA-4-P destabilizes microtubules and causes endothelial cell death. In this study we analyze the mechanisms by which CA-4-P induces the death of proliferating endothelial cells. We demonstrate that at >/=7.5 nmol/L, CA-4-P damages mitotic spindles, arrests cells at metaphase, and leads to the death of mitotic cells with characteristic G(2)/M DNA content. Mitotic arrest was associated with elevated levels of cyclin B1 protein and p34(cdc2) activity. Inhibition of p34(cdc2) activity by purvalanol A caused mitotic-arrested cells to rapidly exit mitosis, suggesting that sustained p34(cdc2) activity was responsible for metaphase arrest. Pharmacological prevention of entry into mitosis protected cells from undergoing cell death, further establishing the link between mitosis and cell death induction by CA-4-P. CA-4-P-mediated cell death shared characteristics of apoptosis but was independent of caspase activation suggesting the involvement of a non-caspase pathway(s). These data suggest that induction of apoptosis in endothelial cells by CA-4-P is associated with prolonged mitotic arrest. Therefore, by activating cell death pathways, CA-4-P, in addition to being an effective anti-vascular agent, may also interfere with regrowth of blood vessels in the tumor.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Mitosis/efectos de los fármacos , Estilbenos/farmacología , Apoptosis/fisiología , Western Blotting , Proteína Quinasa CDC2/efectos de los fármacos , Proteína Quinasa CDC2/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos
15.
J Cell Physiol ; 197(3): 312-25, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14566961

RESUMEN

Tumor hypoxia has long been recognized as a critical issue in oncology. Resistance of hypoxic areas has been shown to affect treatment outcome after radiation, chemotherapy, and surgery in a number of tumor sites. Two main strategies to overcome tumor hypoxia are to increase the delivery of oxygen (or oxygen-mimetic drugs), and exploiting this unique environmental condition of solid tumors for targeted therapy. The first strategy includes hyperbaric oxygen breathing, the administration of carbogen and nicotinamide, and the delivery of chemical radiosensitizers. In contrast, bioreductive drugs and hypoxia-targeted suicide gene therapy aim at activating cytotoxic agents at the tumor site, while sparing normal tissue from damage. The cellular machinery responds to hypoxia by activating the expression of genes involved in angiogenesis, anaerobic metabolism, vascular permeability, and inflammation. In most cases, transcription is initiated by the binding of the transcription factor hypoxia-inducible factor (HIF) to hypoxia responsive elements (HREs). Hypoxia-targeting for gene therapy has been achieved by utilizing promoters containing HREs, to induce selective and efficient transgene activation at the tumor site. Hypoxia-targeted delivery and prodrug activation may add additional levels of selectivity to the treatment. In this article, the latest developments of cancer gene therapy of the hypoxic environment are discussed, with particular attention to combined protocols with ionizing radiation. Ultimately, it is proposed that by adopting specific transgene activation and molecular amplification systems, resistant hypoxic tumor tissues may be effectively targeted with gene therapy.


Asunto(s)
Terapia Genética/métodos , Hipoxia/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Factores de Transcripción , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Terapia Genética/tendencias , Humanos , Hipoxia/tratamiento farmacológico , Hipoxia/genética , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Radioterapia Adyuvante/métodos , Radioterapia Adyuvante/tendencias , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Activación Transcripcional/efectos de la radiación
16.
J Cell Biochem ; 87(2): 221-32, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12244574

RESUMEN

We have previously proposed the horseradish peroxidase (HRP) and the non-toxic plant hormone indole-3-acetic acid (IAA) as a novel system for gene-directed enzyme/prodrug therapy (GDEPT). The cytotoxic potential of HRP/IAA GDEPT and the induction of a bystander effect were demonstrated in vitro under normoxic as well as hypoxic tumour conditions. To date, the chemical agents and the cellular targets involved in HRP/IAA-mediated toxicity have not been identified. In the present work, some of the molecular and morphological features of the cells treated with HRP/IAA gene therapy were analysed. Human T24 bladder carcinoma cells transiently transfected with the HRP cDNA and exposed to the prodrug IAA showed chromatin condensation, formation of apoptotic bodies, DNA fragmentation, and Annexin V binding. Similar effects were observed when the cells were incubated with the apoptotic agent cisplatin. Caspases appeared to be involved as effectors in HRP/IAA-mediated apoptosis, since treatment with a general caspase inhibitor decreased the fraction of cells with micronuclei (MN) by 30%, with fragmented DNA by 50%, and with condensed chromatin by 60%. However, very little degradation of one of the downstream targets of caspase-3, PARP, could be detected, and apoptosis alone did not appear to account for the killing levels measured with a clonogenic assay. The effect of HRP/IAA treatment on cell cycle progression was also investigated, and a rapid cytostatic effect, equally affecting all phases of the division cycle, was observed.


Asunto(s)
Terapia Genética/efectos adversos , Peroxidasa de Rábano Silvestre/genética , Ácidos Indolacéticos/farmacología , Profármacos/farmacología , Anexina A5/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/ultraestructura , Cromatina/metabolismo , Terapia Combinada , Fragmentación del ADN/efectos de los fármacos , ADN Complementario/metabolismo , Fase G1/efectos de los fármacos , Terapia Genética/métodos , Peroxidasa de Rábano Silvestre/metabolismo , Humanos , Ácidos Indolacéticos/farmacocinética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Profármacos/farmacocinética , Factores de Tiempo , Transfección , Células Tumorales Cultivadas/metabolismo , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA