Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cancer Chemother Pharmacol ; 83(1): 97-105, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30368585

RESUMEN

PURPOSE: The primary objective of this study was to determine the recommended dose of the vascular disrupting agent, BNC105P, in combination with gemcitabine and carboplatin in patients with ovarian cancer in first or second relapse with a minimum 4 month progression-free interval after last platinum. METHODS: Patients received carboplatin AUC4 on day 1 in combination with escalating doses of 800 or 1000 mg/m2 gemcitabine on days 1 and 8 and escalating doses of 12 or 16 mg/m2 BNC105P on days 2 and 9 every 21 days for a maximum for six cycles. Maintenance treatment with 16 mg/m2 BNC105P treatment continued for a maximum of six additional cycles. Patients were followed for safety and anti-tumor activity. RESULTS: Fifteen patients were enrolled in the study. Adverse events were most commonly of hematological origin. Dose-limiting toxicities (thrombocytopenia and neutropenia) occurred in two patients at the dose level of 800 mg/m2 gemcitabine, carboplatin AUC4 and 16 mg/m2 BNC105P. No dose-limiting toxicities were observed at a dose level of gemcitabine 1000 mg/m2, carboplatin AUC4 and BNC105P 12 mg/m2. BNC105P as a single agent was well tolerated at a dose of 16 mg/m2 in maintenance treatment. Ten patients (67%) achieved a complete or partial response according to CA125 and/or RECIST response criteria, four of 13 (31%) responded by RECIST alone. The median progression-free survival was 5.9 months. CONCLUSIONS: We have established that BNC105P 12 mg/m2 with gemcitabine 1000 mg/m2 and carboplatin AUC4 is the recommended dose level and has an acceptable toxicity profile. Further exploration of BNC105P in the ovarian cancer setting is planned.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Cistadenocarcinoma Seroso/tratamiento farmacológico , Neoplasias Endometriales/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Adenocarcinoma/secundario , Adulto , Anciano , Benzofuranos/administración & dosificación , Carboplatino/administración & dosificación , Cistadenocarcinoma Seroso/secundario , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Neoplasias Endometriales/secundario , Femenino , Estudios de Seguimiento , Humanos , Dosis Máxima Tolerada , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Organofosfatos/administración & dosificación , Neoplasias Ováricas/patología , Pronóstico , Tasa de Supervivencia , Gemcitabina
2.
J Clin Invest ; 109(4): 533-40, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11854326

RESUMEN

The congenital polycystic kidney (cpk) mutation is the most extensively characterized mouse model of polycystic kidney disease (PKD). The renal cystic disease is fully expressed in homozygotes and is strikingly similar to human autosomal recessive PKD (ARPKD), whereas genetic background modulates the penetrance of the corresponding defect in the developing biliary tree. We now describe the positional cloning, mutation analysis, and expression of a novel gene that is disrupted in cpk mice. The cpk gene is expressed primarily in the kidney and liver and encodes a hydrophilic, 145-amino acid protein, which we term cystin. When expressed exogenously in polarized renal epithelial cells, cystin is detected in cilia, and its expression overlaps with polaris, another PKD-related protein. We therefore propose that the single epithelial cilium is important in the functional differentiation of polarized epithelia and that ciliary dysfunction underlies the PKD phenotype in cpk mice.


Asunto(s)
Proteínas de la Membrana/genética , Mutación , Riñón Poliquístico Autosómico Dominante/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Mapeo Cromosómico , Cilios/metabolismo , Clonación Molecular , ADN Complementario/genética , Modelos Animales de Enfermedad , Expresión Génica , Homocigoto , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Ratones Mutantes , Datos de Secuencia Molecular , Riñón Poliquístico Autosómico Dominante/etiología , ARN Mensajero/genética , ARN Mensajero/metabolismo
3.
Mol Cancer Res ; 4(9): 655-65, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16966434

RESUMEN

The transcriptional repressor CBFA2T3 is a putative breast tumor suppressor. To define the role of CBFA2T3, we used a segment of this protein as bait in a yeast two-hybrid screen and identified a novel uncharacterized protein, ZNF652. In general, primary tumors and cancer cell lines showed lower expression of ZNF652 than normal tissues. Together with the location of this gene on the long arm of chromosome 17q, a region of frequent loss of heterozygosity in cancer, these results suggest a possible role of ZNF652 in tumorigenesis. In silico analysis of this protein revealed that it contains multiple classic zinc finger domains that are predicted to bind DNA. Coimmunoprecipitation assays showed that ZNF652 strongly interacts with CBFA2T3 and this interaction occurs through the COOH-terminal 109 amino acids of ZNF652. In contrast, there was a weak interaction of ZNF652 with CBFA2T1 and CBFA2T2, the other two members of this ETO family. Transcriptional reporter assays further confirmed the strength and selectivity of the ZNF652-CBFA2T3 interaction. The transcriptional repression of growth factor independent-1 (GFI-1), a previously characterized ETO effector zinc finger protein, was shown to be enhanced by CBFA2T1, but to a lesser extent by CBFA2T2 and CBFA2T3. We therefore suggest that each of the various gene effector zinc finger proteins may specifically interact with one or more of the ETO proteins to generate a defined range of transcriptional repressor complexes.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas de Unión al ADN/genética , Fosfoproteínas/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Dedos de Zinc/fisiología , Secuencia de Aminoácidos , Animales , Neoplasias de la Mama/metabolismo , Proteínas de Unión al ADN/metabolismo , Genes Supresores de Tumor , Humanos , Ratones , Datos de Secuencia Molecular , Fosfoproteínas/biosíntesis , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteína 1 Compañera de Translocación de RUNX1 , Conejos , Ratas , Proteínas Represoras/biosíntesis , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/metabolismo , Técnicas del Sistema de Dos Híbridos , Dedos de Zinc/genética
4.
Cancer Res ; 62(16): 4599-604, 2002 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12183414

RESUMEN

Numerous cytogenetic and molecular studies of breast cancer have identified frequent loss of heterozygosity (LOH) of the long arm of human chromosome 16. On the basis of these data, the likely locations of breast cancer tumor suppressor genes are bands 16q22.1 and 16q24.3. We have mapped the CBFA2T3 (MTG16) gene, previously cloned as a fusion partner of the AML1 protein from a rare (16;21) leukemia translocation, to the 16q24.3 breast cancer LOH region. The expression of CBFA2T3 was significantly reduced in a number of breast cancer cell lines and in primary breast tumors, including early ductal carcinomas in situ, when compared with nontransformed breast epithelial cell lines and normal breast tissue. Reintroduction of CBFA2T3 into different breast tumor derived cell lines with decreased expression of this gene reduced colony growth on plastic and in soft agar. CBFA2T3 was shown to function as a transcriptional repressor when tethered to the GAL4 DNA-binding domain in a reporter gene assay and, therefore, has the potential to be a transcriptional repressor in normal breast epithelial cells. Taken together, these findings suggest that CBFA2T3 is a likely candidate for the breast cancer tumor suppressor gene that is the target for the frequent 16q24 LOH in breast neoplasms.


Asunto(s)
Neoplasias de la Mama/genética , Cromosomas Humanos Par 16/genética , Genes Supresores de Tumor , Pérdida de Heterocigocidad , Fosfoproteínas , Proteínas/genética , Proteínas Supresoras de Tumor , División Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Biosíntesis de Proteínas , Proteínas/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Proteínas Represoras , Células Tumorales Cultivadas
5.
Clin Cancer Res ; 21(15): 3420-7, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25788492

RESUMEN

PURPOSE: BNC105P inhibits tubulin polymerization, and preclinical studies suggest possible synergy with everolimus. In this phase I/II study, efficacy and safety of the combination were explored in patients with metastatic renal cell carcinoma (mRCC). EXPERIMENTAL DESIGN: A phase I study in patients with clear cell mRCC and any prior number of therapies was conducted using a classical 3 + 3 design to evaluate standard doses of everolimus with increasing doses of BNC105P. At the recommended phase II dose (RP2D), patients with clear cell mRCC and one to two prior therapies (including ≥ 1 VEGF-TKI) were randomized to BNC105P with everolimus (arm A) or everolimus alone (arm B). The primary endpoint of the study was 6-month progression-free survival (6MPFS). Secondary endpoints included response rate, PFS, overall survival, and exploratory biomarker analyses. RESULTS: In the phase I study (N = 15), a dose of BNC105P at 16 mg/m(2) with everolimus at 10 mg daily was identified as the RP2D. In the phase II study, 139 patients were randomized, with 69 and 67 evaluable patients in arms A and B, respectively. 6MPFS was similar in the treatment arms (arm A: 33.82% vs. arm B: 30.30%, P = 0.66) and no difference in median PFS was observed (arm A: 4.7 mos vs. arm B: 4.1 mos; P = 0.49). Changes in matrix metalloproteinase-9, stem cell factor, sex hormone-binding globulin, and serum amyloid A protein were associated with clinical outcome with BNC105P. CONCLUSIONS: Although the primary endpoint was not met in an unselected population, correlative studies suggest several biomarkers that warrant further prospective evaluation.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Benzofuranos/administración & dosificación , Carcinoma de Células Renales/tratamiento farmacológico , Everolimus/administración & dosificación , Organofosfatos/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Benzofuranos/efectos adversos , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Supervivencia sin Enfermedad , Everolimus/efectos adversos , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Metaloproteinasa 9 de la Matriz/biosíntesis , Persona de Mediana Edad , Organofosfatos/efectos adversos , Globulina de Unión a Hormona Sexual/biosíntesis , Resultado del Tratamiento
6.
Mol Cancer ; 3: 22, 2004 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-15301688

RESUMEN

BACKGROUND: The CBFA2T3 locus located on the human chromosome region 16q24.3 is frequently deleted in breast tumors. CBFA2T3 gene expression levels are aberrant in breast tumor cell lines and the CBFA2T3B isoform is a potential tumor suppressor gene. In the absence of identified mutations to further support a role for this gene in tumorigenesis, we explored whether the CBFA2T3B promoter region is aberrantly methylated and whether this correlates with expression. RESULTS: Aberrant hypo and hypermethylation of the CBFA2T3B promoter was detected in breast tumor cell lines and primary breast tumor samples relative to methylation index interquartile ranges in normal breast counterpart and normal whole blood samples. A statistically significant inverse correlation between aberrant CBFA2T3B promoter methylation and gene expression was established. CONCLUSION: CBFA2T3B is a potential breast tumor suppressor gene affected by aberrant promoter methylation and gene expression. The methylation levels were quantitated using a second-round real-time methylation-specific PCR assay. The detection of both hypo and hypermethylation is a technicality regarding the methylation methodology.


Asunto(s)
Neoplasias de la Mama/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , ADN de Neoplasias/química , ADN de Neoplasias/genética , Regulación de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Metilación , Fosfoproteínas/química , Subunidades de Proteína/genética , Proteínas Represoras/química , Análisis de Secuencia de ADN/métodos , Sulfitos/metabolismo , Proteínas Supresoras de Tumor/química
7.
Cancer Biol Ther ; 15(11): 1552-60, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25482941

RESUMEN

BNC105 is a tubulin targeting compound that selectively disrupts vasculature within solid tumors. The severe tumor hypoxia and necrosis that ensues translates to short term tumor growth inhibition. We sought to identify the molecular and cellular events activated following BNC105 treatment that drives tumor recovery. We investigated tumor adaptation to BNC105-induced hypoxia in animal models of breast and renal cancer. HIF-1α and GLUT-1 were found to be strongly upregulated by BNC105 as was the VEGF signaling axis. Phosphorylation of mTOR, 4E-BP-1 and elF2α were upregulated, consistent with increased protein synthesis and increased expression of VEGF-A. We sought to investigate the potential therapeutic utility of combining BNC105 with agents targeting VEGF and mTOR signaling. Bevacizumab and pazopanib target the VEGF axis and have been approved for first line use in renal cancer. Everolimus targets mTOR and is currently approved in second line therapy of renal and particular breast cancers. We combined these agents with BNC105 to explore the effects on tumor vasculature, tumor growth inhibition and animal survival. Bevacizumab hindered tumor vascular recovery following BNC105 treatment leading to greater tumor growth inhibition in a breast cancer model. Consistent with this, addition of BNC105 to pazopanib treatment resulted in a significant increase in survival in an orthotopic renal cancer model. Combination treatment of BNC105 with everolimus also increased tumor growth inhibition. BNC105 is currently being evaluated in a randomized phase II clinical trial in combination with everolimus in renal cancer.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anisoles/farmacología , Antineoplásicos/farmacología , Benzofuranos/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias Renales/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Anisoles/administración & dosificación , Antineoplásicos/administración & dosificación , Benzofuranos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Humanos , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/mortalidad , Neoplasias Renales/patología , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Lung Cancer ; 81(3): 422-427, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23787063

RESUMEN

BNC105P is a tubulin polymerisation inhibitor that selectively disrupts tumour vasculature and suppresses cancer cell proliferation. This agent has exhibited preclinical and phase I activity in Malignant Pleural Mesothelioma (MPM). This phase II, single arm trial investigated the efficacy and safety of BNC105P as second line therapy in MPM. Participants had progressive MPM after first line pemetrexed/platinum chemotherapy, ECOG PS 0-1, adequate organ function, and measurable disease. BNC105P 16 mg/m(2) was administered intravenously on day 1 and 8 every 21 days until progression or undue toxicity. The primary endpoint was centrally reviewed objective response rate (RR). Tumour response was assessed every two cycles using modified RECIST. 30 patients were enrolled in 10 months, predominantly male (90%), ECOG PS 1 (77%), epithelioid histology (67%), and non-metastatic disease (67%). All patients received at least one dose of study drug, with a median of 2 cycles. No significant haematologic, biochemical, or cardiac adverse events (AEs) were observed. Grade 3 or 4 AEs occurred in 10 patients (33%). There were 2 deaths on study: 1 cardiorespiratory, the other to pneumonia. We observed 1 partial response (3%); 13 patients had stable disease (43%). Median progression free survival was 1.5 months (95% CI 1.4-2.4); median overall survival was 8.2 months (95% CI 3.8-11.9). BNC105P was safe and tolerable. The sole response was insufficient to warrant further research as a single agent.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzofuranos/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Mesotelioma/tratamiento farmacológico , Mesotelioma/patología , Organofosfatos/uso terapéutico , Neoplasias Pleurales/tratamiento farmacológico , Neoplasias Pleurales/patología , Moduladores de Tubulina/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Benzofuranos/administración & dosificación , Benzofuranos/efectos adversos , Biomarcadores/sangre , Biomarcadores/metabolismo , Femenino , Proteínas Ligadas a GPI/sangre , Proteínas Ligadas a GPI/metabolismo , Humanos , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/mortalidad , Masculino , Mesotelina , Mesotelioma/diagnóstico por imagen , Mesotelioma/mortalidad , Mesotelioma Maligno , Persona de Mediana Edad , Estadificación de Neoplasias , Organofosfatos/administración & dosificación , Organofosfatos/efectos adversos , Neoplasias Pleurales/diagnóstico por imagen , Neoplasias Pleurales/mortalidad , Radiografía , Resultado del Tratamiento , Moduladores de Tubulina/administración & dosificación , Moduladores de Tubulina/efectos adversos , Carga Tumoral
9.
Clin Cancer Res ; 17(15): 5152-60, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21690571

RESUMEN

PURPOSE: To determine the recommended phase II dose and evaluate the safety and toxicity profile and pharmacokinetic (PK) and pharmacodynamic (PD) effects of BNC105P, an inhibitor of tubulin polymerization that has vascular disrupting and antiproliferative effects. EXPERIMENTAL DESIGN: BNC105P was administered as a 10-minute infusion on days 1 and 8 of a 21-day cycle in a first-in-human phase I study. A dynamic accelerated dose titration method was used for dose escalation. Plasma concentrations of BNC105P (phosphate prodrug) and BNC105 (active agent) were determined. PD assessments were carried out using dynamic contrast enhanced (DCE)-MRI and analysis of a blood-borne biomarker. RESULTS: Twenty-one subjects with advanced solid tumors were enrolled on 6 dose levels (range: 2.1-18.9 mg/m(2)). The recommended dose level was 16 mg/m(2) and was well tolerated. BNC105P (prodrug) rapidly converted to BNC105 with a half-life of 0.13 hours. Plasma concentrations of BNC105 generally increased in proportion to dose with a half-life of 0.57 hours. Pharmacodymanically active plasma levels were obtained with a dose dependant reduction in the levels of polymerized tubulin (on-target action) being observed in PBMCs. DCE-MRI also indicated blood flow changes in the tumor lesions of a number of subjects. CONCLUSIONS: BNC105P has a favorable toxicity profile at the recommended dose of 16 mg/m(2) and is associated with PD changes consistent with its known mechanism of action. Phase II studies in renal cancer and mesothelioma have commenced.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzofuranos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Organofosfatos/uso terapéutico , Profármacos/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Anisoles/análisis , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Benzofuranos/efectos adversos , Benzofuranos/análisis , Benzofuranos/farmacocinética , Benzofuranos/farmacología , Biomarcadores/análisis , Fármacos Cardiovasculares/uso terapéutico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/metabolismo , Organofosfatos/efectos adversos , Organofosfatos/farmacocinética , Organofosfatos/farmacología , Tubulina (Proteína)/sangre , Tubulina (Proteína)/metabolismo
10.
J Med Chem ; 54(17): 6014-27, 2011 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-21774499

RESUMEN

A structure-activity relationship (SAR) guided design of novel tubulin polymerization inhibitors has resulted in a series of benzo[b]furans with exceptional potency toward cancer cells and activated endothelial cells. The potency of early lead compounds has been substantially improved through the synergistic effect of introducing a conformational bias and additional hydrogen bond donor to the pharmacophore. Screening of a focused library of potent tubulin polymerization inhibitors for selectivity against cancer cells and activated endothelial cells over quiescent endothelial cells has afforded 7-hydroxy-6-methoxy-2-methyl-3-(3,4,5-trimethoxybenzoyl)benzo[b]furan (BNC105, 8) as a potent and selective antiproliferative. Because of poor solubility, 8 is administered as its disodium phosphate ester prodrug 9 (BNC105P), which is rapidly cleaved in vivo to return the active 8. 9 exhibits both superior vascular disrupting and tumor growth inhibitory properties compared with the benchmark agent combretastatin A-4 disodium phosphate 5 (CA4P).


Asunto(s)
Anisoles/farmacología , Aorta/efectos de los fármacos , Benzofuranos/farmacología , Proliferación Celular/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Moduladores de Tubulina/farmacología , Anisoles/síntesis química , Anisoles/química , Aorta/citología , Benzofuranos/síntesis química , Benzofuranos/química , Células Cultivadas , Endotelio Vascular/citología , Humanos , Relación Estructura-Actividad , Moduladores de Tubulina/síntesis química , Moduladores de Tubulina/química
11.
Mol Cancer Ther ; 9(6): 1562-73, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20515948

RESUMEN

Vascular disruption agents (VDA) cause occlusion of tumor vasculature, resulting in hypoxia-driven tumor cell necrosis. Tumor vascular disruption is a therapeutic strategy of great potential; however, VDAs currently under development display a narrow therapeutic margin, with cardiovascular toxicity posing a dose-limiting obstacle. Discovery of new VDAs, which display a wider therapeutic margin, may allow attainment of improved clinical outcomes. To identify such compounds, we used an in vitro selectivity screening approach that exploits the fact that tumor endothelial cells are in a constant state of activation and angiogenesis and do not undergo senescence. Our effort yielded the compound BNC105. This compound acts as a tubulin polymerization inhibitor and displays 80-fold higher potency against endothelial cells that are actively proliferating or are engaged in the formation of in vitro capillaries compared with nonproliferating endothelial cells or endothelium found in stable capillaries. This selectivity was not observed with CA4, a VDA currently under evaluation in phase III clinical trials. BNC105 is more potent and offers a wider therapeutic window. CA4 produces 90% vascular disruption at its no observed adverse event level (NOAEL), whereas BNC105 causes 95% vascular disruption at 1/8th of its NOAEL. Tissue distribution analysis of BNC105 in tumor-bearing mice showed that while the drug is cleared from all tissues 24 hours after administration, it is still present at high concentrations within the solid tumor mass. Furthermore, BNC105 treatment causes tumor regressions with complete tumor clearance in 20% of treated animals.


Asunto(s)
Anisoles/uso terapéutico , Antineoplásicos/uso terapéutico , Benzofuranos/uso terapéutico , Neoplasias/irrigación sanguínea , Neovascularización Patológica/tratamiento farmacológico , Moduladores de Tubulina/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Anisoles/química , Anisoles/metabolismo , Anisoles/farmacocinética , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Benzofuranos/química , Benzofuranos/metabolismo , Benzofuranos/farmacocinética , Capilares/efectos de los fármacos , Línea Celular , Permeabilidad de la Membrana Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Extensiones de la Superficie Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Inducción de Remisión , Resultado del Tratamiento , Moduladores de Tubulina/química , Moduladores de Tubulina/metabolismo , Moduladores de Tubulina/farmacocinética
12.
Expert Opin Ther Targets ; 9(2): 211-5, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15934910

RESUMEN

The formation of blood vessels is a key process in the progression of solid tumours, providing the means for tumour growth and metastasis. A number of drugs are currently being developed to exploit inhibition of angiogenesis in the therapy of cancer. An even greater number of genes that are regulated in models of in vitro angiogenesis have been identified. These genes present potential drug targets for the development of novel, more efficient, drugs that will enable the judicious design of drug cocktails that may be able to account for the many different cancer pathologies and their drug resistance properties. Dealing with the validation of hundreds of potential angiogenesis drug targets requires the utilisation of experimental technology platforms that enable concomitant and dynamic target selection filtering and validation. Such platforms should act as a funnel-like medium-to-low throughput processes that enable the sequential short-listing of hundreds of candidates culminating in the selection of only a small number of well-validated targets that are manageable by drug screening regimes.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de la Angiogénesis/normas , Animales , Antineoplásicos/normas , Sistemas de Liberación de Medicamentos/normas , Humanos , Neoplasias/fisiopatología , Neovascularización Patológica/fisiopatología , Reproducibilidad de los Resultados
13.
Proc Natl Acad Sci U S A ; 101(33): 12212-7, 2004 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-15302923

RESUMEN

Angiogenesis is a major therapeutic target. Ideal drug targets are genes expressed only in endothelial cells (ECs) or only during the angiogenic process. Here, we describe a gene, p73RhoGAP (p73), that has both of these properties. By using a PCR-based subtraction-hybridization approach to clone cDNAs from ECs undergoing capillary-tube formation, we identified a RhoGAP member, p73. p73 displays GTPase activity to Rho but not to Rac or Cdc42. Knockdown of p73 protein, achieved by adenovirus delivery of p73 antisense and by small interfering RNA into ECs, demonstrated the importance of this protein in EC function. Under such conditions, EC migration, proliferation, and capillary-tube formation were inhibited. Furthermore, angiogenesis in vivo was also inhibited by antisense p73. A mutant R82A alteration achieved a similar phenotype in vitro to the antisense, demonstrating the importance of the GTPase-activating protein activity to p73 function. Expression profiling of p73 shows that it is vascular cell-selective, being highly expressed in ECs and smooth-muscle cells but not in other cell types. Finally, we show that the mRNA of p73 is up-regulated in an angiogenic milieu with little or no regulation seen under nonangiogenic conditions. p73, a vascular cell-specific GTPase-activating protein, is an important modulator of angiogenesis and displays many of features that make it worthy of being a drug target.


Asunto(s)
Proteínas Activadoras de GTPasa/fisiología , Neovascularización Fisiológica , Secuencia de Bases , Células Cultivadas , Clonación Molecular , Endotelio Vascular/crecimiento & desarrollo , Endotelio Vascular/fisiología , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/aislamiento & purificación , Regulación de la Expresión Génica , Humanos , Mutagénesis Sitio-Dirigida , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
14.
J Hum Genet ; 49(6): 308-311, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15148587

RESUMEN

The 16p13.3 breakpoints of two de novo translocations of chromosome 16, t(1;16) and t(14;16), were shown by initial mapping studies to have physically adjacent breakpoints. The translocations were ascertained in patients with abnormal phenotypes characterized by predominant epilepsy in one patient and mental retardation in the other. Distamycin/DAPI banding showed that the chromosome 1 breakpoint of the t(1;16) was in the pericentric heterochromatin therefore restricting potential gene disruption to the 16p13.3 breakpoint. The breakpoints of the two translocations were localized to a region of 3.5 and 115 kb respectively and were approximately 900 kb apart. The mapping was confirmed by fluorescence in situ hybridization (FISH) of clones that spanned the breakpoints to metaphase spreads derived from the patients. The mapping data showed both translocations disrupted the ataxin-2-binding protein 1 ( A2BP1) gene that encompasses a large genomic region of 1.7 Mb. A2BP1 encodes a protein that is known to interact with the spinocerebellar ataxia type 2 ( SCA2) protein. It is proposed that disruption of the A2BP1 gene is a cause of the abnormal phenotype of the two patients. Ninety-six patients with sporadic epilepsy and 96 female patients with mental retardation were screened by SSCP for potential mutations of A2BP1. No mutations were found, suggesting that disruption of the A2BP1 gene is not a common cause of sporadic epilepsy or mental retardation.


Asunto(s)
Cromosomas Humanos Par 16 , Epilepsia/genética , Discapacidad Intelectual/genética , Proteínas de Unión al ARN/genética , Translocación Genética , Adolescente , Ataxinas , Preescolar , Mapeo Cromosómico , Exones , Femenino , Humanos , Hibridación Fluorescente in Situ , Intrones , Masculino , Modelos Genéticos , Mutación , Proteínas del Tejido Nervioso , Fenotipo , Mapeo Físico de Cromosoma , Polimorfismo Conformacional Retorcido-Simple , Proteínas/genética , Factores de Empalme de ARN
15.
Genomics ; 80(3): 303-10, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12213200

RESUMEN

Loss of heterozygosity (LOH) of chromosome 16q24.3 is a common genetic alteration observed in invasive ductal and lobular breast carcinomas. We constructed a physical map and generated genomic DNA sequence data spanning 2.4 Mb in this region. Detailed in silico and in vitro analyses of the genomic sequence data enabled the identification of 104 genes. It was hypothesized that tumor-suppressor genes would exhibit marked mRNA expression variability in a panel of breast cancer cell lines as a result of downregulation due to mutation or hypermethylation. We examined the mRNA expression profiles of the genes identified at 16q24.3 in normal breast, a normal breast epithelial cell line, and several breast cancer cell lines exhibiting 16q24.3 LOH. Three of the genes, CYBA, Hs.7970, and CBFA2T3, exhibited variability ten times higher than the baseline. The possible role of these genes as tumor suppressors is discussed.


Asunto(s)
Neoplasias de la Mama/genética , Cromosomas Humanos Par 16 , Genes Supresores de Tumor , Pérdida de Heterocigocidad , Femenino , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Mapeo Físico de Cromosoma , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA