Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Cell Biochem ; 123(3): 532-542, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34935169

RESUMEN

Selenium (Se) is incorporated into the body via the selenocysteine (Sec) biosynthesis pathway, which is critical in the synthesis of selenoproteins, such as glutathione peroxidases and thioredoxin reductases. Selenoproteins, which play a key role in several biological processes, including ferroptosis, drug resistance, endoplasmic reticulum stress, and epigenetic processes, are guided by Se uptake. In this review, we critically analyze the molecular mechanisms of Se metabolism and its potential as a therapeutic target for cancer. Sec insertion sequence binding protein 2 (SECISBP2), which is a positive regulator for the expression of selenoproteins, would be a novel prognostic predictor and an alternate target for cancer. We highlight strategies that attempt to develop a novel Se metabolism-based approach to uncover a new metabolic drug target for cancer therapy. Moreover, we expect extensive clinical use of SECISBP2 as a specific biomarker in cancer therapy in the near future. Of note, scientists face additional challenges in conducting successful research, including investigations on anticancer peptides to target SECISBP2 intracellular protein.


Asunto(s)
Neoplasias , Selenio , Proteínas Portadoras/metabolismo , Humanos , Redes y Vías Metabólicas , Neoplasias/tratamiento farmacológico , Selenio/metabolismo , Selenio/uso terapéutico , Selenoproteínas/química , Selenoproteínas/metabolismo , Reductasa de Tiorredoxina-Disulfuro/metabolismo
2.
Transl Res ; 229: 100-114, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33164812

RESUMEN

Abundant intraperitoneal (IP) accumulation of extracellular mucus in patients with appendiceal mucinous carcinoma peritonei (MCP) causes compressive organ dysfunction and prevents delivery of chemotherapeutic drugs to cancer cells. We hypothesized that reducing extracellular mucus would decrease tumor-related symptoms and improve chemotherapeutic effect in patient-derived models of MCP. Mucolysis was achieved using a combination of bromelain (BRO) and N-acetylcysteine (NAC). Ex vivo experiments of mucolysis and chemotherapeutic drug delivery/effect were conducted with MCP and non-MCP tissue explants. In vivo experiments were performed in mouse and rat patient-derived xenograft (PDX) models of early and late (advanced) MCP. MCP tumor explants were less chemosensitive than non-MCP explants. Chronic IP administration of BRO + NAC in a mouse PDX model of early MCP and a rat PDX model of late (advanced) MCP converted solid mucinous tumors into mucinous ascites (mucolysis) that could be drained via a percutaneous catheter (rat model only), significantly reduced solid mucinous tumor growth and improved the efficacy of chemotherapeutic drugs. Combination of BRO + NAC efficiently lyses extracellular mucus in clinically relevant models of MCP. Conversion of solid mucinous tumors into mucinous ascites decreases tumor bulk and allows for minimally invasive drainage of liquified tumors. Lysis of extracellular mucus removes the protective mucinous coating surrounding cancer cells and improves chemotherapeutic drug delivery/efficacy in cancer cells. Our data provide a preclinical rationale for the clinical evaluation of BRO + NAC as a therapeutic strategy for MCP.


Asunto(s)
Adenocarcinoma Mucinoso/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias del Apéndice/tratamiento farmacológico , Moco/efectos de los fármacos , Neoplasias Peritoneales/tratamiento farmacológico , Acetilcisteína/administración & dosificación , Acetilcisteína/farmacología , Adenocarcinoma Mucinoso/patología , Animales , Neoplasias del Apéndice/patología , Bromelaínas/administración & dosificación , Bromelaínas/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Ratones Desnudos , Neoplasias Peritoneales/patología , Ratas Desnudas , Técnicas de Cultivo de Tejidos/métodos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Transl Res ; 169: 19-30.e1, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26589109

RESUMEN

Excessive accumulation of mucin 2 (MUC2; a gel-forming secreted mucin) protein in the peritoneal cavity is the major cause of morbidity and mortality in pseudomyxoma peritonei (PMP). Hypoxia (hypoxia-inducible factor-1α; HIF-1α) has been shown to regulate the expression of similar mucins (eg, MUC5AC). We hypothesized that hypoxia (HIF-1α) drives MUC2 expression in PMP and is therefore a novel target to reduce mucinous tumor growth. The regulation of MUC2 by 2% hypoxia (HIF-1α) was evaluated in MUC2-secreting LS174T cells. The effect of BAY 87-2243, an inhibitor of HIF-1α, on MUC2 expression and mucinous tumor growth was evaluated in LS174T cells, PMP explant tissue, and in a unique intraperitoneal murine xenograft model of PMP. In vitro exposure of LS174T cells to hypoxia increased MUC2 messenger RNA (mRNA) and protein expression and increased HIF-1α binding to the MUC2 promoter. Hypoxia-mediated MUC2 protein overexpression was downregulated by transfected HIF-1α small interfering RNA (siRNA) compared with scrambled siRNA in LS174T cells. BAY 87-2243 inhibited hypoxia-induced MUC2 mRNA and protein expression in LS174T cells and PMP explant tissue. In a murine xenograft model of PMP, chronic oral therapy with BAY 87-2243 inhibited mucinous tumor growth and MUC2, HIF-1α expression in the tumor tissue. Our data suggest that hypoxia (HIF-1α) induces MUC2 promoter activity to increase MUC2 expression. HIF-1α inhibition decreases MUC2 production and mucinous tumor growth, providing a preclinical rationale for the use of HIF-1α inhibitors to treat patients with PMP.


Asunto(s)
Hipoxia de la Célula , Mucina 2/biosíntesis , Seudomixoma Peritoneal/terapia , Animales , Línea Celular Tumoral , Xenoinjertos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Seudomixoma Peritoneal/metabolismo , ARN Interferente Pequeño/genética
4.
Apoptosis ; 19(11): 1603-15, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25156145

RESUMEN

In this study, we attempted to develop a multimodality approach using chemotherapeutic agent mitomycin C, biologic agent tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/Apo-2L), and mild hyperthermia to treat colon cancer. For this study, human colon cancer LS174T, LS180, HCT116 and CX-1 cells were infected with secretory TRAIL-armed adenovirus (Ad.TRAIL) and treated with chemotherapeutic agent mitomycin C and hyperthermia. The combinatorial treatment caused a synergistic induction of apoptosis which was mediated through an increase in caspase activation. The combinational treatment promoted the JNK-Bcl-xL-Bak pathway which transmitted the synergistic effect through the mitochondria-dependent apoptotic pathway. JNK signaling led to Bcl-xL phosphorylation at serine 62, dissociation of Bak from Bcl-xL, oligomerization of Bak, alteration of mitochondrial membrane potential, and subsequent cytochrome c release. Overexpression of dominant-negative mutant of Bcl-xL (S62A), but not dominant-positive mutant of Bcl-xL (S62D), suppressed the synergistic death effect. Interestingly, Beclin-1 was dissociated from Bcl-xL and overexpression of dominant-negative mutant of Bcl-xL (S62A), but not dominant-positive mutant of Bcl-xL (S62D), suppressed dissociation of Beclin-1 from Bcl-xL. A combinatorial treatment of mitomycin C, Ad.TRAIL and hyperthermia induced Beclin-1 cleavage, but the Beclin-1 cleavage was abolished in Beclin-1 double mutant (D133A/D146A) knock-in HCT116 cells, suppressing the apoptosis induced by the combination therapy. We believe that this study supports the application of the multimodality approach to colon cancer therapy.


Asunto(s)
Adenoviridae/genética , Antibióticos Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Neoplasias del Colon/patología , Hipertermia Inducida , Proteínas de la Membrana/metabolismo , Mitomicina/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteína bcl-X/metabolismo , Beclina-1 , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Citocromos c/metabolismo , Vectores Genéticos , Humanos , MAP Quinasa Quinasa 4/metabolismo , Mitocondrias/metabolismo , Multimerización de Proteína , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo
5.
PLoS One ; 8(8): e73654, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24013390

RESUMEN

Colorectal cancer is the third leading cause of cancer-related mortality in the world--the main cause of death from colorectal cancer is hepatic metastases, which can be treated with isolated hepatic perfusion (IHP). Searching for the most clinically relevant approaches for treating colorectal metastatic disease by isolated hepatic perfusion (IHP), we developed the application of oxaliplatin concomitantly with hyperthermia and humanized death receptor 4 (DR4) antibody mapatumumab (Mapa), and investigated the molecular mechanisms of this multimodality treatment in human colon cancer cell lines CX-1 and HCT116 as well as human colon cancer stem cells Tu-12, Tu-21 and Tu-22. We showed here, in this study, that the synergistic effect of the multimodality treatment-induced apoptosis was caspase dependent and activated death signaling via both the extrinsic apoptotic pathway and the intrinsic pathway. Death signaling was activated by c-Jun N-terminal kinase (JNK) signaling which led to Bcl-xL phosphorylation at serine 62, decreasing the anti-apoptotic activity of Bcl-xL, which contributed to the intrinsic pathway. The downregulation of cellular FLICE inhibitory protein long isoform (c-FLIPL) in the extrinsic pathway was accomplished through ubiquitination at lysine residue (K) 195 and protein synthesis inhibition. Overexpression of c-FLIPL mutant (K195R) and Bcl-xL mutant (S62A) completely abrogated the synergistic effect. The successful outcome of this study supports the application of multimodality strategy to patients with colorectal hepatic metastases who fail to respond to standard chemoradiotherapy that predominantly targets the mitochondrial apoptotic pathway.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Apoptosis , Neoplasias del Colon/metabolismo , Neoplasias del Colon/terapia , Hipertermia Inducida , Compuestos Organoplatinos/farmacología , Anticuerpos Monoclonales Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica , Línea Celular Tumoral , Neoplasias del Colon/patología , Sinergismo Farmacológico , Humanos , Mitocondrias/metabolismo , Mitocondrias/patología , Metástasis de la Neoplasia , Compuestos Organoplatinos/agonistas , Oxaliplatino
6.
Mol Cancer Res ; 10(12): 1567-79, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23051936

RESUMEN

Colorectal cancer is the third leading cause of cancer-related mortality in the world. The main cause of death because of colorectal cancer is hepatic metastases, which can be treated using isolated hepatic perfusion (IHP), allowing treatment of colorectal metastasis with various methods. In this study, we present a novel potent multimodality strategy comprising humanized death receptor 4 (DR4) antibody mapatumumab in combination with oxaliplatin and hyperthermia to treat human colon cancer cells. Oxaliplatin and hyperthermia sensitized colon cancer cells to mapatumumab in the mitochondrial-dependent apoptotic pathway and increased reactive oxygen species (ROS) production, leading to Bcl-xL phosphorylation at serine 62 in a c-jun-NH2-kinase (JNK)-dependent manner. Overexpression of Bcl-xL reduced the efficacy of the multimodality treatment, whereas phosphorylation of Bcl-xL decreased its antiapoptotic activity. The multimodality treatment dissociated Bcl-xL from Bax, allowing Bax oligomerization to induce cytochrome c release from mitochondria. In addition, the multimodality treatment significantly inhibited colorectal cancer xenografts' tumor growth. The successful outcome of this study will support the application of multimodality strategy to colorectal hepatic metastases.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Neoplasias del Colon/terapia , Hipertermia Inducida/métodos , Compuestos Organoplatinos/farmacología , Proteína bcl-X/metabolismo , Animales , Anticuerpos Monoclonales Humanizados , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Terapia Combinada , Citocromos c/metabolismo , Sinergismo Farmacológico , Células HCT116 , Humanos , MAP Quinasa Quinasa 4/metabolismo , Masculino , Ratones , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Oxaliplatino , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Proteína X Asociada a bcl-2/metabolismo
7.
J Cell Biochem ; 113(5): 1547-58, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22174016

RESUMEN

Colorectal cancer is the third leading cause of cancer-related mortality in the world; death usually results from uncontrolled metastatic disease. Previously, we developed a novel strategy of TNF-related apoptosis-inducing ligand (Apo2L/TRAIL) in combination with hyperthermia to treat hepatic colorectal metastases. However, previous studies suggest a potential hepatocyte cytotoxicity with TRAIL. Unlike TRAIL, anti-human TRAIL receptor antibody induces apoptosis without hepatocyte toxicity. In this study, we evaluated the anti-tumor efficacy of humanized anti-death receptor 4 (DR4) antibody mapatumumab (Mapa) by comparing it with TRAIL in combination with hyperthermia. TRAIL, which binds to both DR4 and death receptor 5 (DR5), was approximately tenfold more effective than Mapa in inducing apoptosis. However, hyperthermia enhances apoptosis induced by either agent. We observed that the synergistic effect was mediated through elevation of reactive oxygen species, c-Jun N-terminal kinase activation, Bax oligomerization, and translocalization to the mitochondria, loss of mitochondrial membrane potential, release of cytochrome c to cytosol, activation of caspases, and increase in poly(ADP-ribose) polymerase cleavage. We believe that the successful outcome of this study will support the application of Mapa in combination with hyperthermia to colorectal hepatic metastases.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Anticuerpos Monoclonales Humanizados , Apoptosis , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Terapia Combinada , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Hipertermia Inducida , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/terapia , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteína X Asociada a bcl-2/deficiencia , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
8.
J Cell Biochem ; 112(1): 118-27, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21053278

RESUMEN

The aim of this study was to investigate the effect of garlic constituent diallyl trisulfide (DATS) on the cell-death signaling pathway in a human breast cell line (MDA-MB-231). We observed that DATS (10-100 µM) treatment resulted in dose- and time-dependent cytotoxicity. Treatment of MDA-MB-231 cells with a cytotoxicity inducing concentration of DATS (50-80 µM) resulted in an increase in the intracellular level of reactive oxygen species (ROS). Data from assay with MitoSOX(TM) Red reagent suggest that mitochondria are the main source of ROS generation during DATS treatment. DATS-induced oxidative stress was detected through glutaredoxin (GRX), a redox-sensing molecule, and subsequently GRX was dissociated from apoptosis signal-regulating kinase 1 (ASK1). Dissociation of GRX from ASK1 resulted in the activation of ASK1. ASK1 activated a downstream signal transduction JNK (c-Jun N-terminal kinase)-Bim pathway. SP600125, a JNK inhibitor, inhibited DATS-induced Bim phosphorylation and protected cells from DATS-induced cytotoxicity. Our results indicate that the cytotoxicity caused by DATS is mediated by the generation of ROS and subsequent activation of the ASK1-JNK-Bim signal transduction pathway in human breast carcinoma MDA-MB-231 cells.


Asunto(s)
Compuestos Alílicos/toxicidad , Antineoplásicos/toxicidad , Antioxidantes/toxicidad , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Sulfuros/toxicidad , Compuestos Alílicos/farmacología , Antineoplásicos/farmacología , Antioxidantes/farmacología , Apoptosis , Proteína 11 Similar a Bcl2 , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ajo/química , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MAP Quinasa Quinasa Quinasa 5/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Sulfuros/farmacología
9.
J Cell Biochem ; 110(5): 1073-81, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20544795

RESUMEN

Approximately 25% of patients with colorectal cancer develop metastases to the liver, and surgery is currently the best treatment available. But there are several patients who are unresectable, and isolated hepatic perfusion (IHP) offers a different approach in helping to treat these patients. IHP is a method used for isolating the liver and delivering high doses of chemotherapeutic agents. The efficacy of IHP has been improved by combining hyperthermia not only with chemotherapeutics but with other deliverable agents such as tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). In this study, we used human colorectal cancer CX-1 cells and treated them with hyperthermia and TRAIL, causing cytotoxicity. We were able to demonstrate that the numbers of live cells were significantly reduced with hyperthermia and 10 ng/ml of TRAIL combined. We also showed that the effect of hyperthermia on TRAIL in our studies was enhancement of the apoptotic pathway by the promotion of JNK and Bim(EL) activity as well as PARP cleavage. We have also used our CX-1 cells to generate tumors in Balb/c nude mice. With intratumoral injections of TRAIL combined with hyperthermia at 42 degrees C, we were able to show a delayed onset of tumor growth in our xenograft model.


Asunto(s)
Neoplasias Colorrectales/prevención & control , Hipertermia Inducida , Transducción de Señal/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antracenos/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Terapia Combinada , Calor , Humanos , Immunoblotting , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Proto-Oncogénicas/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Carga Tumoral/efectos de los fármacos
10.
Int J Hyperthermia ; 22(8): 713-28, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17391000

RESUMEN

PURPOSE: This study investigated whether hyperthermia can enhance TRAIL-induced apoptotic death. METHODS: Human prostate adenocarcinoma DU-145, human pancreatic carcinoma MIA PaCa-2 and BxPC-3, human colon fibroblast CCD-33Co and rat prostate endothelial YPEN-1 cells were treated with various concentrations of TRAIL (0-200 ngml(-1)) with hyperthermia (40-42 degrees C). RESULTS: It was observed in human cancer cells, but not in normal cells, that TRAIL induced apoptotic death and also that hyperthermia (40-42 degrees C) promoted TRAIL-induced apoptotic death. Enhancement of TRAIL-mediated apoptosis by hyperthermia was detected by an increase in PARP cleavage, the hallmark feature of apoptosis, as well as by activation of caspases. There were no significant changes in the intra-cellular levels of death receptors (DRs), decoy receptors (DcRs) and anti-apoptotic proteins. Interestingly, data from in vitro enzyme kinetics assay demonstrated that hyperthermia promoted caspase enzyme activity. CONCLUSIONS: These data suggest that cancer cells are more susceptible to TRAIL in the condition of hyperthermia (40-42 degrees C). The promotion of caspase enzyme activity by hyperthermia may be responsible for enhancement of TRAIL-induced apoptotic death.


Asunto(s)
Apoptosis/fisiología , Hipertermia Inducida , Proteínas Recombinantes/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Animales , Terapia Biológica , Caspasa 1/metabolismo , Línea Celular Tumoral , Terapia Combinada/métodos , Humanos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ratas , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/fisiología
11.
Cancer Gene Ther ; 9(3): 267-74, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11896443

RESUMEN

We have previously developed a recombinant adenovirus containing a fusion gene of Escherichia coli cytosine deaminase (CD) and herpes simplex virus type 1 thymidine kinase (HSV-1 TK) controlled by a cytomegalovirus (CMV) enhancer-promoter. This replication-incompetent adenovirus effectively transduced the CD-TK gene into human prostate adenocarcinoma DU-145 or PC-3 cells. Interestingly, heat shock at 41 degrees C for 4 hours elevated the level of CD-TK by approximately 5- to 20-fold at a multiplicity of infection (MOI) of 1. Heat-enhanced expression of CD-TK promoted cytotoxicity by 23-, 9-, or 47-fold in the presence of 50 microg/mL ganciclovir (GCV), 500 microg/mL 5-fluorocytosine (5-FC), or 50 microg/mL GCV+500 microg/mL 5-FC, respectively, at an MOI of 1. Moreover, there was an increase in radiosensitivity when adenovirus-infected cells were heated at 41 degrees C for 4 hours followed by irradiation in the presence of the prodrugs. Virus+heat+1 microg/mL GCV treatment increased radiosensitivity by a dose-modifying factor (DMF) of 2.2, whereas virus+heat+10 microg/mL 5-FC exposure resulted in a DMF of 2.3. Radiosensitization was clearly enhanced as a result of combined prodrug exposure (DMF=4.4). Our results suggest that the efficiency in expression of suicide genes from an adenoviral vector used for cytotoxic anticancer therapy could be improved by combining heat treatment with radiation therapy.


Asunto(s)
Adenocarcinoma/terapia , Adenoviridae/genética , Antivirales/uso terapéutico , Vectores Genéticos , Hipertermia Inducida , Neoplasias de la Próstata/terapia , Adenocarcinoma/genética , Supervivencia Celular , Citosina Desaminasa , Sinergismo Farmacológico , Flucitosina/uso terapéutico , Ganciclovir/uso terapéutico , Expresión Génica , Técnicas de Transferencia de Gen , Herpesvirus Humano 1/enzimología , Humanos , Masculino , Nucleósido Desaminasas/genética , Neoplasias de la Próstata/genética , Radioterapia , Proteínas Recombinantes de Fusión/genética , Timidina Quinasa/genética , Timidina Quinasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA