Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Korean Circ J ; 54(2): 63-75, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38111184

RESUMEN

BACKGROUND AND OBJECTIVES: Evidence regarding the efficacy and safety of intracardiac echocardiography (ICE) for guidance during transcatheter aortic valve replacement (TAVR) is limited. This study aimed to compare the clinical efficacy and safety of ICE versus transesophageal echocardiography (TEE) for guiding TAVR. METHODS: This prospective cohort study included patients who underwent TAVR from August 18, 2015, to June 31, 2021. Eligible patients were stratified by echocardiographic modality (ICE or TEE) and anesthesia mode (monitored anesthesia care [MAC] or general anesthesia [GA]). Primary outcome was the 1-year composite of all-cause mortality, rehospitalization for cardiovascular cause, or stroke, according to the Valve Academic Research Consortium-3 (VARC-3) definition. Propensity score matching was performed, and study outcomes were analyzed for the matched cohorts. RESULTS: Of the 359 eligible patients, 120 patients were matched for the ICE-MAC and TEE-GA groups, respectively. The incidence of primary outcome was similar between matched groups (18.3% vs. 20.0%; adjusted hazard ratio, 0.94; 95% confidence interval [CI], 0.53-1.68; p=0.843). ICE-MAC and TEE-GA also had similar incidences of moderate-to-severe paravalvular regurgitation (PVR) (4.2% vs. 5.0%; adjusted odds ratio, 0.83; 95% CI, 0.23-2.82; p=0.758), new permanent pacemaker implantation, and VARC-3 types 2-4 bleeding. CONCLUSIONS: ICE was comparable to TEE for guidance during TAVR for the composite clinical efficacy outcome, with similar incidences of moderate-to-severe PVR, new permanent pacemaker implantation, and major bleeding. These results suggest that ICE could be a safe and effective alternative echocardiographic modality to TEE for guiding TAVR.

2.
Gynecol Oncol ; 130(3): 579-87, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23721800

RESUMEN

OBJECTIVES: While most women with ovarian cancer will achieve complete remission after treatment, the majority will relapse within two years, highlighting the need for novel therapies. Cancer stem cells (CSC) have been identified in ovarian cancer and most other carcinomas as a small population of cells that can self-renew. CSC are more chemoresistant and radio-resistant than the bulk tumor cells; it is likely that CSC are responsible for relapse, the major problem in cancer treatment. CD133 has emerged as one of the most promising markers for CSC in ovarian cancer. The hypothesis driving this study is that despite their low numbers in ovarian cancer tumors, CSC can be eradicated using CD133 targeted therapy and tumor growth can be inhibited. METHODS: Ovarian cancer cell lines were evaluated using flow cytometry for expression of CD133. In vitro viability studies with an anti-CD133 targeted toxin were performed on one of the cell lines, NIH:OVCAR5. The drug was tested in vivo using a stably transfected luciferase-expressing NIH:OVCAR5 subline in nude mice, so that tumor growth could be monitored by digital imaging in real time. RESULTS: Ovarian cancer cell lines showed 5.6% to 16.0% CD133 expression. dCD133KDEL inhibited the in vitro growth of NIH:OVCAR5 cells. Despite low numbers of CD133-expressing cells in the tumor population, intraperitoneal drug therapy caused a selective decrease in tumor progression in intraperitoneal NIH:OVCAR5-luc tumors. CONCLUSIONS: Directly targeting CSC that are a major cause of drug resistant tumor relapse with an anti-CD133 targeted toxin shows promise for ovarian cancer therapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD/metabolismo , Biomarcadores de Tumor/metabolismo , Citotoxinas/uso terapéutico , Glicoproteínas/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Péptidos/metabolismo , Antígeno AC133 , Animales , Anticuerpos Monoclonales/farmacología , Antígenos CD/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citotoxinas/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Glicoproteínas/inmunología , Humanos , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Células Madre Neoplásicas/metabolismo , Neoplasias Ováricas/metabolismo , Péptidos/inmunología
3.
Cancer Biother Radiopharm ; 28(4): 274-82, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23611188

RESUMEN

A heterodimeric bispecific biological recombinant drug was synthesized by splicing DNA fragments from two fully humanized single-chain variable-fragment (scFV) antibody fragments forming a novel drug simultaneously recognizing the CD16 natural killer (NK) cell marker and the cancer marker epithelial cell adhesion molecule (EpCAM). The drug precipitously enhanced the killing of human carcinomas of the prostate, breast, colon, head, and neck even at very low effector:target ratios. The drug EpCAM16 rendered even nonactivated NK cell-proficient killers and activated them to kill via degranulation and cytokine production. Studies show that bispecific antibodies can be used to induce proficient killing of the carcinoma targets that ordinarily are resistant to NK-mediated killing. Apparently, the innate immune system can be effectively recruited to kill cancer cells using the bispecific antibody platform and EpCAM targeting.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Carcinoma/inmunología , Degranulación de la Célula , Inmunidad Innata/efectos de los fármacos , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Proteínas Recombinantes de Fusión/farmacología , Anticuerpos de Cadena Única/farmacología , Formación de Anticuerpos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Neoplasias de la Mama/inmunología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Línea Celular Tumoral , Supervivencia Celular , Neoplasias del Colon/inmunología , Molécula de Adhesión Celular Epitelial , Neoplasias de Cabeza y Cuello/inmunología , Humanos , Interferón gamma/metabolismo , Masculino , Neoplasias de la Próstata/inmunología , Receptores de IgG/genética , Receptores de IgG/inmunología , Proteínas Recombinantes de Fusión/genética , Anticuerpos de Cadena Única/genética
4.
Drug Deliv Transl Res ; 3(2): 195-204, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25787984

RESUMEN

CD133 expression enriches for tumor-initiating cells and is a negative prognostic factor in numerous cancers. We previously developed an immunotoxin against CD133 by fusing a gene fragment encoding the scFv portion of an anti-CD133 antibody to a gene fragment encoding deimmunized PE38KDEL. The resulting fusion protein, dCD133KDEL, demonstrated potent antitumor activity following intratumoral delivery into head neck cell carcinoma xenografts. However, the efficacy against other tumors and the tolerability of systemic administration remained unclear. The purpose of this study was to evaluate the tolerability and efficacy of dCD133KDEL in a systemic human breast carcinoma model. Time course viability studies showed that dCD133KDEL selectively inhibited MDA-MB-231 ductal breast carcinoma cells that contained a minority CD133(+) subpopulation, implicating CD133(+) cells as a source for self-renewal within this cell line. Furthermore, systemic administration of dCD133KDEL caused regression or inhibition of tumor growth in mice bearing an intrasplenic MDA-MB-231 tumor challenge as a model for metastatic disease. In the same model, combined therapy with dCD133KDEL and another immunotoxin designed to target the bulk tumor mass was the most effective therapy, supporting the idea that such combination therapies might better address tumor heterogeneity. dCD133KDEL shows promise as a therapeutic agent and as a biologic tool to study cancer stem cells.

5.
Oral Oncol ; 48(12): 1202-7, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22818892

RESUMEN

OBJECTIVES: To investigate the efficacy of the bispecific targeted toxin, dEGFATFKDEL, on head and neck carcinoma cell lines in vitro and in vivo. MATERIALS AND METHODS: A deimmunized bispecific anti-cancer agent was constructed to simultaneously target both the overexpressed EGF receptor on carcinomas and the urokinase receptor (uPAR), that is found on the endothelial cells of the neovasculature within tumors. Flow cytometry assays were performed to determine the level of EGFR expressed on a variety of carcinoma lines. These lines were then tested in tritiated leucine incorporation assays to determine the efficacy of dEGFATFKDEL. Human vein endothelial primary cells were also tested to determine the effectiveness of the ATF portion of the molecule that binds uPAR. Furthermore, mouse studies were performed to determine whether dEGFATFKDEL was effective at inhibiting tumor growth in vivo. RESULTS: UMSCC-11B and NA, two head and neck squamous cell carcinomas, highly expressed EGFR. Both the carcinoma lines and the human vein endothelial cells were inhibited at sub-nanomolar concentrations by dEGFATFKDEL. The tumor studies showed that the tumors treated with dEGFATFKDEL were significantly inhibited whereas the negative control and untreated tumors progressed. In a separate in vivo study involving another carcinoma line, MDA-MB-231, the effectiveness of dEGFATFKDEL was confirmed. No toxicity was seen at the doses used in either of these mouse studies. CONCLUSIONS: This bispecific agent is effective in a mouse model of head and neck squamous cell carcinoma. Further study of this reagent for use in the treatment of carcinomas is warranted.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Modelos Animales de Enfermedad , Receptores ErbB/efectos de los fármacos , Neoplasias de Cabeza y Cuello/metabolismo , Oligopéptidos/farmacología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/efectos de los fármacos , Animales , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Receptores ErbB/metabolismo , Citometría de Flujo , Neoplasias de Cabeza y Cuello/patología , Humanos , Masculino , Ratones , Ratones Desnudos , Oligopéptidos/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo
6.
Pancreas ; 41(5): 789-96, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22258068

RESUMEN

OBJECTIVE: The objective was to test a bispecific ligand-directed toxin (BLT), with reduced immunogenicity for enhanced efficacy in targeting orthotopic pancreatic cancer in vivo. METHOD: A new BLT was created in which both human epidermal growth factor (EGF) and interleukin 4 cytokines were cloned onto the same single chain molecule with deimmunized pseudomonas exotoxin (dEGF4KDEL). Key amino acids dictating B-cell generation of neutralizing antitoxin antibodies were mutated. Bioassays were used to determine whether mutation reduced potency, and enzyme-linked immunosorbent assay studies were performed to determine whether antitoxin antibodies were reduced. A genetically altered luciferase MIA PaCa-2 xenograft model was used to image in real time and determine effects on systemic malignant human cancer. Bispecific ligand-directed toxins targeting B cells were used as specificity controls. RESULTS: Deimmunized EGF4KDEL was significantly effective after systemic injection against established orthotopic MIA PaCa-2 pancreatic cancer and selectively prevented metastasis. Mutagenesis significantly reduced antitoxin levels in vivo with no apparent activity loss in vitro. The drug was effective against 3 human pancreatic cancer lines in vitro, MIA PaCa-2, SW1990, and S2VP10. CONCLUSIONS: Despite the metastatic nature of the MIA PaCa-2 orthotopic tumor xenografted in nude mice, high percentages of tumors responded to extended dEGFKDEL treatment resulting in significant anticancer effects and disease-free survivors.


Asunto(s)
Antineoplásicos/farmacología , Inmunotoxinas/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , Secuencia de Aminoácidos , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacología , Antineoplásicos/inmunología , Línea Celular Tumoral , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/inmunología , Exotoxinas/genética , Exotoxinas/inmunología , Humanos , Inmunotoxinas/genética , Inmunotoxinas/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Ligandos , Masculino , Ratones , Ratones Desnudos , Mutación , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Pseudomonas/genética , Pseudomonas/inmunología , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología
7.
J Neuroimmune Pharmacol ; 7(2): 436-43, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21858458

RESUMEN

Opioids significantly alter functional responses of lymphocytes following activation. The opiate Morphine, alters the Th1 to Th2 response and modulates functional responses such as cytolytic activity and T-cell proliferation. Although there has been extensive research involving morphine's effects on lymphocytes, little is known about the effects morphine has on lymphocyte trafficking. The objective of the study was to use in vivo bioluminescent imaging to determine morphine's effect on the trafficking pattern of splenocytes systemically and into the CNS either in a naïve state or following a neuroinflammatory stimulus. A neuroinflammatory response was induced by intracerebrally administering a DNA IFN-γ DNA plasmid into morphine-dependent or placebo wildtype mice. Mice with or without a neurostimulus received adoptively transferred firefly luciferase transgenic splenocytes and imaged. Morphine dependence significantly altered the inherent ability of splenocytes to traffic into the spleen, and lead to non-directed chaotic trafficking throughout the animal, including into the CNS. The morphine-mediated effects on trafficking were blocked by the antagonist naltrexone. Morphine dependence intensified splenocyte infiltration into the CNS following neuroinflammation induced by IFN-γ gene transfer. The study precented determined that morphine severely altered the ability of non-activated splenocytes to home to the spleen, inducing extrasplenic trafficking thoughout the animal. In addition to altering the ability of naive splenocyte to traffic to the spleen, this study demonstrated that morphine profoundly exacerbated lymphocyte infiltration into the CNS following a neurostimulus.


Asunto(s)
Encéfalo/inmunología , Quimiotaxis de Leucocito/efectos de los fármacos , Linfocitos/efectos de los fármacos , Morfina/farmacología , Narcóticos/farmacología , Bazo/efectos de los fármacos , Traslado Adoptivo , Animales , Inflamación/inducido químicamente , Interferón gamma/inmunología , Linfocitos/citología , Linfocitos/inmunología , Ratones , Ratones Transgénicos , Plásmidos , Bazo/citología , Bazo/inmunología
8.
Mol Cancer Ther ; 10(10): 1829-38, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21862685

RESUMEN

A novel anticancer agent was constructed by fusing a gene encoding the scFV that targets both glycosylated and unglycosylated forms of CD133 to a gene fragment encoding deimmunized PE38KDEL. The resulting fusion protein, dCD133KDEL, was studied to determine its ability to bind and kill tumor-initiating cells in vitro and in vivo. The anti-CD133 scFV selectively bound HEK293 cells transfected with the CD133 receptor gene. Time course viability studies showed that dCD133KDEL selectively inhibited NA-SCC and UMSCC-11B, 2 head and neck squamous cell carcinomas that contain a CD133 expressing subpopulation. Importantly, the drug did not inhibit the viability of hematopoietic lineages measured by long-term culture-initiating cell and colony-forming assays from sorted human CD34+ progenitor cells. In addition to in vitro studies, in vivo tumor initiation experiments confirmed that CD133-sorted cells implanted into the flanks of nude mice grew faster and larger than unsorted cells. In contrast, cells that were pretreated with dCD133KDEL before implantation showed the slowest and lowest incidence of tumors. Furthermore, UMSCC-11B-luc tumors treated with multiple intratumoral injections of dCD133KDEL showed marked growth inhibition, leading to complete degradation of the tumors that was not observed with an irrelevant control-targeted toxin. Experiments in immunocompetent mice showed that toxin deimmunization resulted in a 90% reduction in circulating antitoxin levels. These studies show that dCD133KDEL is a novel anticancer agent effective at inhibiting cell proliferation, tumor initiation, and eliminating established tumors by targeting the CD133 subpopulation. This agent shows significant promise for potential development as a clinically useful therapy.


Asunto(s)
Antígenos CD/inmunología , Carcinoma de Células Escamosas/tratamiento farmacológico , Glicoproteínas/inmunología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/patología , Inmunotoxinas/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Péptidos/inmunología , Proteínas Recombinantes de Fusión/farmacología , Antígeno AC133 , ADP Ribosa Transferasas/genética , ADP Ribosa Transferasas/farmacocinética , ADP Ribosa Transferasas/farmacología , Animales , Antígenos CD/biosíntesis , Antígenos CD/genética , Toxinas Bacterianas/genética , Toxinas Bacterianas/farmacocinética , Toxinas Bacterianas/farmacología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Exotoxinas/genética , Exotoxinas/farmacocinética , Exotoxinas/farmacología , Femenino , Glicoproteínas/biosíntesis , Glicoproteínas/genética , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Fragmentos de Inmunoglobulinas/genética , Fragmentos de Inmunoglobulinas/farmacología , Inmunotoxinas/genética , Inmunotoxinas/farmacocinética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Péptidos/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacocinética , Transfección , Factores de Virulencia/genética , Factores de Virulencia/farmacocinética , Factores de Virulencia/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Exotoxina A de Pseudomonas aeruginosa
9.
J Neurosurg ; 114(6): 1662-71, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21294620

RESUMEN

OBJECT: The authors of this study aimed to genetically design a bispecific targeted toxin that would simultaneously target overexpressed markers on glioma as well as the tumor vasculature, to mutate certain amino acids to reduce the immunogenicity of this new drug, and to determine whether the drug was able to effectively reduce aggressive human brain tumors in a rat xenograft model via a novel hollow fiber (HF) catheter delivery system. METHODS: A new bispecific ligand-directed toxin (BLT) was created in which 2 human cytokines-epidermal growth factor ([EGF], targeting overexpressed EGF receptor) and amino acid terminal fragment ([ATF], targeting urokinase plasminogen activator receptor)-were cloned onto the same single-chain molecule with truncated Pseudomonas exotoxin with a terminal lysyl-aspartyl-glutamyl-leucine (KDEL) sequence. Site-specific mutagenesis was used to mutate amino acids in 7 key epitopic toxin regions that dictate the B cell generation of neutralizing antitoxin antibodies to deimmunize the drug, now called "EGFATFKDEL 7mut." Bioassays were used to determine whether mutation reduced the drug's potency, and enzyme-linked immunosorbent assay studies were performed to determine whether antitoxin antibodies were decreased. Aggressive brain tumors were intracranially established in nude rats by using human U87 glioma genetically marked with a firefly luciferase reporter gene (U87-luc), and the rats were stereotactically treated with 2 intracranial injections of deimmunized EGFATFKDEL via convection-enhanced delivery (CED). Drug was administered through a novel HF catheter to reduce drug backflow upon delivery. RESULTS: In vitro, EGFATFKDEL 7mut selectively killed the human glioblastoma cell line U87-luc as well as cultured human endothelial cells in the form of the human umbilical vein endothelial cells. Deimmunization did not reduce drug activity. In vivo, when rats with brain tumors were intracranially treated with drug via CED and a novel HF catheter to reduce backflow, there were significant tumor reductions in 2 experiments (p < 0.01). Some rats survived with a tumor-free status until 130 days post-tumor inoculation. An irrelevant BLT control did not protect establishing specificity. The maximal tolerated dose of EGFATFKDEL 7mut was established at 2 µg/injection or 8.0 µg/kg, and data indicated that this dose was nontoxic. Antitoxin antibodies were reduced by at least 90%. CONCLUSIONS: First, data indicated that the BLT framework is effective for simultaneously targeting glioma and its neovasculature. Second, in the rodent CED studies, newly developed HF catheters that limit backflow are effective for drug delivery. Third, by mutating critical amino acids, the authors reduced the threat of the interference of neutralizing antibodies that are generated against the drug. The authors' experiments addressed some of the most urgent limitations in the targeted toxin field.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Factor de Crecimiento Epidérmico/uso terapéutico , Glioblastoma/tratamiento farmacológico , Inmunotoxinas/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Receptores del Activador de Plasminógeno Tipo Uroquinasa/uso terapéutico , Animales , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Diseño de Fármacos , Ensayo de Inmunoadsorción Enzimática , Glioblastoma/irrigación sanguínea , Glioblastoma/patología , Humanos , Mutagénesis Sitio-Dirigida , Trasplante de Neoplasias , Ratas , Ratas Desnudas , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Neurooncol ; 103(2): 255-66, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20830604

RESUMEN

A bispecific ligand-directed toxin (BLT), called EGFATFKDEL, consisting of human epidermal growth factor, a fragment of urokinase, and truncated pseudomonas exotoxin (PE38) was assembled in order to target human glioblastoma. Immunogenicity was reduced by mutating seven immunodominant B-cell epitopes on the PE38 molecule to create a new agent, EGFATFKDEL 7mut. In vitro, the drug selectively killed several human glioblastoma cell lines. EGFATFKDEL is our first BLT designed to simultaneously target EGFR on solid tumors and uPAR on the tumor neovasculature. In vitro assays revealed that the agent is effective against glioblastoma cell lines as well as human umbilical vein endothelial cells (HUVEC). Additionally, the bispecific drug displayed enhanced binding to overexpressed epidermal growth factor receptor and urokinase receptor when compared to similar monospecific drugs, EGFKDEL and ATFKDEL. In vivo, an aggressive human glioblastoma cell line was genetically marked with a firefly luciferase reporter gene and administered to the flanks of nude mice. Treatment with intratumoral injections of EGFATFKDEL 7mut eradicated small tumors in over half of the treated mice, which survived with tumor free status at least 100 days post tumor inoculation. ATFKDEL, which primarily targets the tumor neovasculature, prevented tumor growth but did not result in tumor-free mice in most cases. Specificity was shown by treating with an irrelevant BLT control which did not protect mice. Finally, immunization experiments in immunocompetent mice revealed significantly reduced anti-toxin production in EGFATFKDEL 7mut treated groups. Thus, EGFATFKDEL 7mut is an effective drug for glioblastoma therapy in this murine model and warrants further study.


Asunto(s)
ADP Ribosa Transferasas/farmacología , Antineoplásicos/farmacología , Toxinas Bacterianas/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Receptores ErbB , Exotoxinas/farmacología , Glioblastoma/tratamiento farmacológico , Activador de Plasminógeno de Tipo Uroquinasa , Factores de Virulencia/farmacología , Animales , Neoplasias Encefálicas/metabolismo , Células Endoteliales/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Citometría de Flujo , Glioblastoma/metabolismo , Humanos , Ligandos , Ratones , Ratones Desnudos , Fragmentos de Péptidos/síntesis química , Proteínas Recombinantes de Fusión/síntesis química , Proteínas Recombinantes de Fusión/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Exotoxina A de Pseudomonas aeruginosa
11.
Mol Cancer Ther ; 9(6): 1872-83, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20530709

RESUMEN

A drug of high potency and reduced immunogenicity is needed to develop a targeted biological drug that when injected systemically can penetrate to malignant B cells. Therefore, a novel deimmunized bispecific ligand-directed toxin targeted by dual high-affinity single-chain Fvs (scFv) spliced to PE38 with a KDEL COOH-terminus was genetically engineered. The aims were to reduce toxin immunogenicity using mutagenesis, measure the ability of mutated drug to elicit antitoxin antibody responses, and show that mutated drug was effective against systemic B-cell lymphoma in vivo. Both human anti-CD22 scFv and anti-CD19 scFv were cloned onto the same single-chain molecule with truncated pseudomonas exotoxin (PE38) to create the drug. Site-specific mutagenesis was used to mutate amino acids in seven key epitopic toxin regions that dictate B-cell generation of neutralizing antitoxin antibodies. Bioassays were used to determine whether mutation reduced potency, and ELISAs were done to determine whether antitoxin antibodies were reduced. Finally, a powerful genetically altered luciferase xenograft model was used that could be imaged in real time to determine the effect on systemic malignant human B-cell lymphoma, Raji-luc. Patient B-lineage acute lymphoblastic leukemia, B-cell chronic lymphocytic leukemia, and B lymphoma were high in CD22 and CD19 expression. 2219KDEL7mut was significantly effective against systemic Raji-luc in mice and prevented metastatic spread. Mutagenesis reduced neutralizing antitoxin antibodies by approximately 80% with no apparent loss in in vitro or in vivo activity. Because 2219KDEL7mut immunogenicity was significantly reduced and the drug was highly effective in vivo, we can now give multiple drug treatments with targeted toxins in future clinical trials.


Asunto(s)
ADP Ribosa Transferasas/inmunología , Anticuerpos Biespecíficos/inmunología , Antígenos CD19/metabolismo , Linfocitos B/patología , Toxinas Bacterianas/inmunología , Bioingeniería/métodos , Exotoxinas/inmunología , Metástasis de la Neoplasia/patología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Factores de Virulencia/inmunología , ADP Ribosa Transferasas/farmacología , Animales , Anticuerpos Biespecíficos/farmacología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Toxinas Bacterianas/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Exotoxinas/farmacología , Humanos , Ratones , Ratones SCID , Modelos Moleculares , Mutación/genética , Factores de Virulencia/farmacología , Exotoxina A de Pseudomonas aeruginosa
12.
Pancreas ; 39(6): 913-22, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20182395

RESUMEN

OBJECTIVE: Biologicals targeting epidermal growth factor (EGF) and interleukin 13 receptors not only react with overexpressed markers on cancer cells but also react with receptors on normal cells. Because we developed novel bispecific ligand-directed toxins synthesized by cloning EGF and interleukin 13 on the same molecule with toxin, our objective was to determine whether we could block normal receptors while still targeting receptors overexpressed on cancer cells, thereby decreasing toxicity while maintaining efficacy. METHODS: A method, toxicity blocking (ToxBloc), was developed in which a bolus intraperitoneal dose of recombinant EGF13 (without toxin) was given to mice approximately 15 to 20 minutes before DTEGF13. Experiments were then performed to determine whether the maximal tolerated dose (MTD) was reduced and whether we were still able to eliminate progression of aggressive human, metastatic, pancreatic cancer induced by orthotopic injection (OT) in nude mice. RESULTS: ToxBloc permitted us to safely exceed the DTEGF13 maximal tolerated dose by 15-fold. This approach permitted repetitive high dosing with the bispecific ligand-directed toxin resulting in tumor regression (P < 0.01). Tumor effects were documented using a tumor imaging model in which OT tumor growth was monitored noninvasively in real time. ToxBloc was selective because other bispecific peptides did not block. CONCLUSIONS: ToxBloc represents a new method of drug delivery and a potential solution to the problem of toxicity.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Neoplasias Pancreáticas/tratamiento farmacológico , Proteínas Recombinantes de Fusión/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Toxina Diftérica/genética , Toxina Diftérica/metabolismo , Relación Dosis-Respuesta a Droga , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Interleucina-13/genética , Interleucina-13/metabolismo , Masculino , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/patología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Resultado del Tratamiento
13.
Clin Cancer Res ; 15(19): 6137-47, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19789305

RESUMEN

PURPOSE: To develop a targeted biological drug that when systemically injected can penetrate to metastatic breast cancer tumors, one needs a drug of high potency and reduced immunogenicity. Thus, we bioengineered a novel bispecific ligand-directed toxin (BLT) targeted by dual high-affinity cytokines with a PE(38)KDEL COOH terminus. Our purpose was to reduce toxin immunogenicity using mutagenesis, measure the ability of mutated drug to elicit B-cell antitoxin antibody responses, and show that mutated drug was effective against systemic breast cancer in vivo. EXPERIMENTAL DESIGN: A new BLT was created in which both human epidermal growth factor (EGF) and interleukin 4 cytokines were cloned onto the same single-chain molecule with truncated Pseudomonas exotoxin (PE(38)). Site-specific mutagenesis was used to mutate amino acids in seven key epitopic toxin regions that dictate B-cell generation of neutralizing antitoxin antibodies. Bioassays were used to determine whether mutation reduced potency, and ELISA studies were done to determine whether antitoxin antibodies were reduced. Finally, a genetically altered luciferase xenograft model was used; this model could be imaged in real time to determine the effect on the systemic malignant human breast cancer MDA-MB-231. RESULTS: EGF4KDEL 7mut was significantly effective against established systemic human breast cancer and prevented metastatic spread. Mutagenesis reduced immunogenicity by approximately 90% with no apparent loss in in vitro or in vivo activity. CONCLUSIONS: Because EGF4KDEL 7mut was highly effective even when we waited 26 days to begin therapy and because immunogenicity was significantly reduced, we can now give multiple drug treatments for chemotherapy-refractory breast cancer in clinical trials.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Carcinoma/tratamiento farmacológico , Factor de Crecimiento Epidérmico/uso terapéutico , Receptores de Interleucina-4/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Carcinoma/inmunología , Carcinoma/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/inmunología , Factor de Crecimiento Epidérmico/administración & dosificación , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/inmunología , Femenino , Humanos , Inmunotoxinas/genética , Inmunotoxinas/inmunología , Inmunotoxinas/uso terapéutico , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutagénesis Sitio-Dirigida , Metástasis de la Neoplasia , Receptores de Interleucina-4/administración & dosificación , Receptores de Interleucina-4/genética , Receptores de Interleucina-4/inmunología , Proteínas Recombinantes de Fusión/inmunología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Neurooncol ; 95(3): 331-342, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19517064

RESUMEN

A bispecific ligand-directed toxin (BLT) consisting of human interleukin-13, epithelial growth factor, and the first 389 amino acids of diphtheria toxin was assembled in order to target human glioblastoma. In vitro, DTEGF13 selectively killed the human glioblastoma cell line U87-luc as well as other human glioblastomas. DTEGF13 fulfilled the requirement of a successful BLT by having greater activity than either of its monospecific counterparts or their mixture proving it necessary to have both ligands on the same single chain molecule. Aggressive brain tumors established intracranially (IC) in nude rats with U87 glioma genetically marked with a firefly luciferase reporter gene were treated with two injections of DTEGF13 using convection enhanced delivery resulting in tumor eradication in 50% of the rats which survived with tumor free status at least 110 days post tumor inoculation. An irrelevant BLT control did not protect establishing specificity. The bispecific DTEGF13 MTD dose was measured at 2 microg/injection or 0.5 microg/kg and toxicity studies indicated safety in this dose. Combination of monospecific DTEGF and DTIL13 did not inhibit tumor growth. ELISA assay indicated that anti-DT antibodies were not generated in normal immunocompetent rats given identical intracranial DTEGF13 therapy. Thus, DTEGF13 is safe and efficacious as an alternative drug for glioblastoma therapy and warrants further study.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Glioblastoma/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/toxicidad , Animales , Especificidad de Anticuerpos , Neoplasias Encefálicas/patología , División Celular/efectos de los fármacos , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Genes Reporteros , Glioblastoma/patología , Humanos , Inmunoglobulina G/sangre , Ligandos , Hígado/efectos de los fármacos , Luciferasas/genética , Trasplante de Neoplasias , Ratas , Ratas Desnudas , Proteínas Recombinantes de Fusión/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Stem Cells Dev ; 17(1): 173-84, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18271701

RESUMEN

The concept of cancer stem cells suggests that there are malignant stem-like cells within a tumor that are responsible for tumor renewal and resistance to cytotoxic therapies. Studies have identified glioma stem-like cells that extrude Hoechst 33342 dye, representing a double-negative "side population" (SP) thought to be selectively resistant to drug therapy. A CD133+ stem cell-like subpopulation has been isolated from a human glioma that was enriched for tumor-initiating cells. It is unknown whether CD133+ cells with similar phenotype persist in established glioma cell lines, or if CD133 is a marker of glioma stem-like cells in rodents. We investigated whether CD133+ and SP cells existed in the GL261 cell line, a syngeneic mouse glioma model that is widely used for preclinical and translational research. Intracerebral injection of less than 100 CD133+ GL261 cells formed tumors, whereas it required 10,000 CD133(-) cells to initiate a tumor. CD133+ GL261 cells expressed nestin, formed tumor spheres with high frequency, and differentiated into glial and neuronal-like cells. Similar to GL261, seven human glioma cell lines analyzed also contained a rare CD133+ population. Surprisingly, we found that CD133+ GL261 cells did not reside in the SP, nor did the majority ( approximately 94%) of CD133+ human glioma cells. These results demonstrate that the expression of CD133 in murine glioma cells is associated with enhanced tumorigenicity and a stem-like phenotype. This study also reveals a previously unrecognized level of heterogeneity in glioma cell lines, exposing several populations of cells that have characteristics of cancer stem cells.


Asunto(s)
Antígenos CD , Glioma/patología , Glicoproteínas , Células Madre Neoplásicas/patología , Péptidos , Antígeno AC133 , Animales , Diferenciación Celular , Línea Celular Tumoral , Humanos , Ratones , Trasplante de Neoplasias , Neuroglía , Neuronas
16.
J Neurooncol ; 87(1): 51-61, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18084721

RESUMEN

To improve activity of a recombinant IL-13 cytotoxin (CT) comprised of IL-13 spliced to truncated diphtheria toxin (DT(390)), epidermal growth factor (EGF) was added to the same single chain protein. This new recombinant bispecific CT, called DTEGF13, enhanced the killing potency against the human glioblastoma lines, U87MG (0.015 nM) and U118MG (0.02 nM). A similar enhancement was observed against the lung carcinoma cell line, Calu-3 (0.0018 nM). Enhanced activity could not be explained by an increased number of cytokines available for binding since a combination of monospecific DTEGF and DTIL13 did not cause the same enhanced activity. Enhanced activity was dependent on the presence of both cytokines on the same single chain molecule and killing was receptor specific since target receptor negative leukemia cells were unaffected by the highly selective DTEGF13 and cytotoxicity could be blocked with anti-EGFR and anti-IL-13 antibodies. In a xenograft flank tumor model, intratumoral injection of DTEGF13, but not monospecific DTEGF or DTIL13, significantly inhibited the growth of established U87 tumors in nude mice (P < 0.04). In this model, the human EGF and IL-13 components of DTEGF13 are reactive with mouse EGFR and IL-13R, respectively. These studies show that a new co-targeting agent that simultaneously recognizes EGFR and IL-13R is more effective than its monospecific counterparts and that DTEGF13 has therapeutic advantages for glioblastoma.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Receptores ErbB/metabolismo , Glioblastoma/tratamiento farmacológico , Interleucina-13/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Animales , Antineoplásicos/síntesis química , Línea Celular Tumoral , Clonación Molecular , Toxina Diftérica/farmacología , Humanos , Ratones , Ratones Desnudos , Proteínas Recombinantes de Fusión/síntesis química , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Immunother ; 30(8): 789-97, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18049330

RESUMEN

Dendritic cell (DC) vaccines have shown antitumor activity in experimental glioma models and in human glioma patients. The typical approach has been to generate the vaccine ex vivo, by pulsing DCs with tumor lysate or peptides, then administering the DCs back into the patient. This process requires significant expertise and expenses in DC generation. Immature DCs which present antigens to T cells in the absence of appropriate costimulatory signals can lead to induction of immune tolerance. Recent studies have shown that coadministration of toll-like receptor 9 agonists, CpG oligodeoxynucleotides, can promote DC vaccines to break immune tolerance to tumor antigens. We investigated the therapeutic efficacy of in vivo DC activation, by directly administering glioma cell lysate with CpG oligodeoxynucleotides (CpG/lysate), in glioma-bearing mice. Subcutaneous vaccination with CpG/lysate induced a significant increase (P<0.05) in the number of total T cells and activated DCs in lymph nodes draining the vaccination site as compared to mice treated with CpG or tumor lysate alone. Mice vaccinated with CpG/lysate exhibited over 2 times greater median survival than mice in the control groups (P<0.05). Up to 55% of mice vaccinated with CpG/lysate were rendered tumor-free as assessed by survival and bioluminescent imaging. Splenocytes taken from mice vaccinated with CpG/lysate elaborated significantly more IFN-gamma production and displayed greater tumor cell lysis activity compared with the control groups (P<0.05). These results suggest direct vaccination with CpG/lysate provides an alternative and effective approach to induce host antitumor immunity and warrants clinical investigation in the immunotherapy of cancer.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Glioblastoma/terapia , Oligodesoxirribonucleótidos/inmunología , Vacunación/métodos , Adyuvantes Inmunológicos/uso terapéutico , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Recuento de Células , Extractos Celulares/inmunología , Línea Celular Tumoral , Técnicas de Cocultivo , Pruebas Inmunológicas de Citotoxicidad , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Glioblastoma/inmunología , Glioblastoma/patología , Interferón gamma/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Bazo/citología , Bazo/inmunología , Análisis de Supervivencia , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/inmunología , Resultado del Tratamiento
18.
J Neurosurg ; 107(3): 568-77, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17886557

RESUMEN

OBJECT: A hollow fiber catheter was developed to improve the distribution of drugs administered via direct infusion into the central nervous system (CNS). It is a porous catheter that significantly increases the surface area of brain tissue into which a drug is infused. METHODS: Dye was infused into the mouse brain through convection-enhanced delivery (CED) using a 28-gauge needle compared with a 3-mm-long hollow fiber catheter. To determine whether a hollow fiber catheter could increase the distribution of gene therapy vectors, a recombinant adenovirus expressing the firefly luciferase reporter was injected into the mouse striatum. Gene expression was monitored using in vivo bioluminescent imaging. To assess the distribution of gene transfer, an adenovirus expressing green fluorescent protein was injected into the striatum using a hollow fiber catheter or a needle. RESULTS: Hollow fiber catheter-mediated infusion increased the volume of brain tissue labeled with dye by 2.7 times relative to needle-mediated infusion. In vivo imaging revealed that catheter-mediated infusion of adenovirus resulted in gene expression that was 10-times greater than that mediated by a needle. The catheter appreciably increased the area of brain transduced with adenovirus relative to a needle, affecting a significant portion of the injected hemisphere. CONCLUSIONS: The miniature hollow fiber catheter used in this study significantly increased the distribution of dye and adenoviral-mediated gene transfer in the mouse brain compared with the levels reached using a 28-gauge needle. Compared with standard single-port clinical catheters, the hollow fiber catheter has the advantage of millions of nanoscale pores to increase surface area and bulk flow in the CNS. Extending the scale of the hollow fiber catheter for the large mammalian brain shows promise in increasing the distribution and efficacy of gene therapy and drug therapy using CED.


Asunto(s)
Adenoviridae , Encéfalo/metabolismo , Cateterismo , Técnicas de Transferencia de Gen/instrumentación , Vectores Genéticos/farmacocinética , Adenoviridae/enzimología , Adenoviridae/genética , Animales , Materiales Biocompatibles , Colorantes/administración & dosificación , Colorantes/farmacocinética , Convección , Diseño de Equipo , Azul de Evans/administración & dosificación , Azul de Evans/farmacocinética , Vectores Genéticos/administración & dosificación , Infusiones Parenterales/instrumentación , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos CBA , Polímeros , Sulfonas
19.
J Neurosurg ; 107(1): 136-44, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17639883

RESUMEN

OBJECT: The purpose of this study was to evaluate the gene transfer capability and tolerability of plasmid DNA/polyethylenimine (PEI) complexes in comparison with adenovirus and naked plasmid DNA in the canine brain. METHODS: Plasmid or adenoviral vectors encoding firefly luciferase were injected directly into the cerebral parenchyma of five adult dogs at varying doses and volumes. Serial physical and neurological examinations, as well as blood and cerebrospinal fluid (CSF) analyses, were conducted before and after the surgery for 3 days. Three days after gene delivery, a luciferase activity assay and immunofluorescence analysis were used to test the brain tissue for gene expression. RESULTS: Injection into the brain parenchyma resulted in gene transfer throughout the cerebrum with every vector tested. Luciferase expression was highest when adenovirus vectors were used. Injection of plasmid DNA/PEI complexes and naked DNA resulted in similar levels of luciferase expression, which were on average 0.5 to 1.5% of the expression achieved with adenovirus vectors. Immunofluorescent microscopy analysis revealed that plasmid DNA/PEI complexes transduced mainly neurons, whereas adenovirus transduced mainly astrocytes. No significant acute side effects or neurological complications were observed in any of the dogs. Mononuclear cell counts significantly increased in the CSF after adenovirus injection and modestly increased after injection of plasmid DNA/PEI complexes, suggesting that a mild, acute inflammatory response occurred in the central nervous system (CNS). CONCLUSIONS: Compared with rodent models that are limited by very small brains, the dog is an excellent preclinical model in which to assess the distribution and safety of emerging gene transfer technologies. In this study, short-term gene transfer was evaluated as a prelude to long-term expression and safety studies. The authors conclude that the viral and nonviral vectors tested were well tolerated and effective at mediating gene transfer throughout a large portion of the canine brain. The nonviral plasmid vectors were less effective than adenovirus, yet they still achieved appreciable gene expression levels. Due to reduced gene transfer efficiency relative to viral vectors, nonviral vectors may be most useful when the expressed protein is secreted or exerts a bystander effect. Nonviral vectors offer an alternative means to genetically modify cells within the CNS of large mammals.


Asunto(s)
Adenovirus Caninos/genética , Técnicas de Transferencia de Gen/instrumentación , Terapia Genética/instrumentación , Plásmidos/genética , Animales , Astrocitos/citología , Astrocitos/virología , Análisis Químico de la Sangre , Encéfalo/citología , Encéfalo/enzimología , Encéfalo/virología , Neoplasias Encefálicas/terapia , Enfermedades Virales del Sistema Nervioso Central/genética , Enfermedades Virales del Sistema Nervioso Central/patología , Enfermedades Virales del Sistema Nervioso Central/virología , Perros , Estudios de Factibilidad , Vectores Genéticos/genética , Glioma/terapia , Inflamación/patología , Inflamación/virología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Luciferasas de Luciérnaga/genética , Luciferasas de Luciérnaga/metabolismo , Microscopía Fluorescente , Neuronas/citología , Neuronas/metabolismo , Neuronas/virología , Plásmidos/fisiología , Polietileneimina/uso terapéutico , Transducción Genética/métodos , Vacunas de ADN/genética
20.
Mol Ther ; 12(5): 778-88, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16150649

RESUMEN

Glioblastoma is a fatal brain tumor that becomes highly vascularized by secreting proangiogenic factors and depends on continued angiogenesis to increase in size. Consequently, a successful antiangiogenic therapy should provide long-term inhibition of tumor-induced angiogenesis, suggesting long-term gene transfer as a therapeutic strategy. In this study a soluble vascular endothelial growth factor receptor (sFlt-1) and an angiostatin-endostatin fusion gene (statin-AE) were codelivered to human glioblastoma xenografts by nonviral gene transfer using the Sleeping Beauty (SB) transposon. In subcutaneously implanted xenografts, co-injection of both transgenes showed marked anti-tumor activity as demonstrated by reduction of tumor vessel density, inhibition or abolition of glioma growth, and increase in animal survival (P = 0.003). Using luciferase-stable engrafted intracranial gliomas, the anti-tumor effect of convection-enhanced delivery of plasmid DNA into the tumor was assessed by luciferase in vivo imaging. Sustained tumor regression of intracranial gliomas was achieved only when statin-AE and sFlt-1 transposons were coadministered with SB-transposase-encoding DNA to facilitate long-term expression. We show that SB can be used to increase animal survival significantly (P = 0.008) by combinatorial antiangiogenic gene transfer in an intracranial glioma model.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias Encefálicas/terapia , Elementos Transponibles de ADN , Terapia Genética , Vectores Genéticos , Glioblastoma/terapia , Inhibidores de la Angiogénesis/genética , Angiostatinas/genética , Animales , Neoplasias Encefálicas/genética , Endostatinas/genética , Expresión Génica , Técnicas de Transferencia de Gen , Glioblastoma/genética , Humanos , Luciferasas/análisis , Ratones , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Plásmidos/genética , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Trasplante Heterólogo , Transposasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...