Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 289(12): 8493-507, 2014 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-24474690

RESUMEN

Targeting effector molecules to tumor cells is a promising mode of action for cancer therapy and diagnostics. Binding proteins with high affinity and specificity for a tumor target that carry effector molecules such as toxins, cytokines, or radiolabels to their intended site of action are required for these applications. In order to yield high tumor accumulation while maintaining low levels in healthy tissues and blood, the half-life of such conjugates needs to be in an optimal range. Scaffold-based binding molecules are small proteins with high affinity and short systemic circulation. Due to their low molecular complexity, they are well suited for combination with effector molecules as well as half-life extension technologies yielding therapeutics with half-lives adapted to the specific therapy. We have identified ubiquitin as an ideal scaffold protein due to its outstanding biophysical and biochemical properties. Based on a dimeric ubiquitin library, high affinity and specific binding molecules, so-called Affilin® molecules, have been selected against the extradomain B of fibronectin, a target almost exclusively expressed in tumor tissues. Extradomain B-binding molecules feature high thermal and serum stability as well as strong in vitro target binding and in vivo tumor accumulation. Application of several half-life extension technologies results in molecules of largely unaffected affinity but significantly prolonged in vivo half-life and tumor retention. Our results demonstrate the utility of ubiquitin as a scaffold for the generation of high affinity binders in a modular fashion, which can be combined with effector molecules and half-life extension technologies.


Asunto(s)
Fibronectinas/metabolismo , Neoplasias/metabolismo , Ubiquitina/metabolismo , Animales , Línea Celular , Humanos , Ratones , Modelos Moleculares , Biblioteca de Péptidos , Unión Proteica , Ingeniería de Proteínas , Estructura Terciaria de Proteína , Ubiquitina/química , Ubiquitina/genética , Ubiquitina/farmacocinética
2.
Nat Med ; 12(2): 235-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16462802

RESUMEN

The angiopoietins Ang-1 and Ang-2 have been identified as ligands of the receptor tyrosine kinase Tie-2 (refs. 1,2). Paracrine Ang-1-mediated activation of Tie-2 acts as a regulator of vessel maturation and vascular quiescence. In turn, the antagonistic ligand Ang-2 acts by an autocrine mechanism and is stored in endothelial Weibel-Palade bodies from where it can be rapidly released upon stimulation. The rapid release of Ang-2 implies functions of the angiopoietin-Tie system beyond its established role during vascular morphogenesis as a regulator of rapid vascular responses. Here we show that mice deficient in Ang-2 (encoded by the gene Angpt2) cannot elicit an inflammatory response in thioglycollate-induced or Staphylococcus aureus-induced peritonitis, or in the dorsal skinfold chamber model. Recombinant Ang-2 restores the inflammation defect in Angpt2(-/-) mice. Intravital microscopy showed normal TNF-alpha-induced leukocyte rolling in the vasculature of Angpt2(-/-)mice, but rolling cells did not firmly adhere to activated endothelium. Cellular experiments showed that Ang-2 promotes adhesion by sensitizing endothelial cells toward TNF-alpha and modulating TNF-alpha-induced expression of endothelial cell adhesion molecules. Together, these findings identify Ang-2 as an autocrine regulator of endothelial cell inflammatory responses. Ang-2 thereby acts as a switch of vascular responsiveness exerting a permissive role for the activities of proinflammatory cytokines.


Asunto(s)
Angiopoyetina 2/fisiología , Inflamación/etiología , Factor de Necrosis Tumoral alfa/fisiología , Angiopoyetina 1/fisiología , Angiopoyetina 2/deficiencia , Angiopoyetina 2/genética , Animales , Citocinas/fisiología , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Humanos , Inflamación/patología , Inflamación/fisiopatología , Mediadores de Inflamación/fisiología , Ratones , Ratones Noqueados , Modelos Biológicos , Neovascularización Patológica , Transducción de Señal
3.
J Biol Chem ; 285(31): 23842-9, 2010 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-20519501

RESUMEN

The angiopoietins (Ang-1 and Ang-2) have been identified as agonistic and antagonistic ligands of the endothelial receptor tyrosine kinase Tie2, respectively. Both ligands have been demonstrated to induce translocation of Tie2 to cell-cell junctions. However, only Ang-1 induces Tie2-dependent Akt activation and subsequent survival signaling and endothelial quiescence. Ang-2 interferes negatively with Ang-1/Tie2 signaling, thereby antagonizing the Ang-1/Tie2 axis. Here, we show that both Ang-1 and Ang-2 recruit beta3 integrins to Tie2. This co-localization is most prominent in cell-cell junctions. However, only Ang-2 stimulation resulted in complex formation among Tie2, alphavbeta3 integrin, and focal adhesion kinase as evidenced by co-immunoprecipitation experiments. Focal adhesion kinase was phosphorylated in the FAT domain at Ser(910) upon Ang-2 stimulation and the adaptor proteins p130Cas and talin dissociated from alphavbeta3 integrin. The alphavbeta3 integrin was internalized, ubiquitinylated, and gated toward lysosomes. Taken together, the experiments define Tie2/alphavbeta3 integrin association-induced integrin internalization and degradation as mechanistic consequences of endothelial Ang-2 stimulation.


Asunto(s)
Angiopoyetina 2/metabolismo , Células Endoteliales/citología , Integrina alfaVbeta3/metabolismo , Comunicación Celular , Línea Celular , Movimiento Celular , Endotelio Vascular/citología , Humanos , Inmunoprecipitación , Lisosomas/metabolismo , Modelos Biológicos , Fosforilación , Receptor TIE-2/metabolismo , Transducción de Señal , Ubiquitina/química
4.
J Clin Oncol ; 39(2): 145-154, 2021 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-33301375

RESUMEN

PURPOSE: A first-in-human study was performed with MP0250, a DARPin drug candidate. MP0250 specifically inhibits both vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) with the aim of disrupting the tumor microenvironment. PATIENTS AND METHODS: A multicenter, open-label, repeated-dose, phase I study was conducted to assess the safety, tolerability, and pharmacokinetics of MP0250 in 45 patients with advanced solid tumors. In the dose-escalation part, 24 patients received MP0250 as a 3-hour infusion once every 2 weeks at five different dose levels (0.5-12 mg/kg). Once the maximum tolerated dose (MTD) was established, 21 patients were treated with a 1-hour infusion (n = 13, 8 mg/kg, once every 2 weeks and n = 8, 12 mg/kg, once every 3 weeks) of MP0250 in the dose confirmation cohorts. RESULTS: In the dose-escalation cohort, patients treated with 12 mg/kg MP0250 once every 2 weeks experienced dose-limiting toxicities. Therefore, MTD was 8 mg/kg once every 2 weeks or 12 mg/kg once every 3 weeks. The most common adverse events (AEs) were hypertension (69%), proteinuria (51%), and diarrhea and nausea (both 36%); hypoalbuminemia was reported in 24% of patients. Most AEs were consistent with inhibition of the VEGF and HGF pathways. Exposure was dose-proportional and sustained throughout the dosing period for all patients (up to 15 months). The half-life was about 2 weeks. Signs of single-agent antitumor activity were observed: 1 unconfirmed partial response with a time to progression of 23 weeks and 24 patients with stable disease, with the longest duration of 72 weeks and a median duration of 18 weeks. CONCLUSION: MP0250 is a first-in-class DARPin drug candidate with suitable tolerability and appropriate pharmacokinetic properties for further development in combination with other anticancer therapies.


Asunto(s)
Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Proteínas Recombinantes de Fusión/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Adulto , Anciano , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Estudios de Cohortes , Relación Dosis-Respuesta a Droga , Femenino , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/farmacocinética , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adulto Joven
5.
J Proteome Res ; 9(4): 1913-22, 2010 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-20085282

RESUMEN

The cancer cell secretome has emerged as an attractive subproteome for discovery of candidate blood-based biomarkers. To choose the best performing workflow, we assessed the performance of three first-dimension separation strategies prior to nanoLC-MS/MS analysis: (1) 1D gel electrophoresis (1DGE), (2) peptide SCX chromatography, and (3) tC2 protein reversed phase chromatography. 1DGE using 4-12% gradient gels outperformed the SCX and tC2 methods with respect to number of identified proteins (1092 vs 979 and 580, respectively), reproducibility of protein identification (80% vs 70% and 72%, respectively, assessed in biological N = 3). Reproducibility of protein quantitation based on spectral counting was similar for all 3 methods (CV: 26% vs 24% and 24%, respectively). As a proof-of-concept of secretome proteomics for blood-based biomarker discovery, the gradient 1DGE workflow was subsequently applied to identify IGF1R-signaling related proteins in the secretome of mouse embryonic fibroblasts transformed with human IGF1R (MEF/Toff/IGF1R). VEGF and osteopontin were differentially detected by LC-MS/MS and verified in secretomes by ELISA. Follow-up in serum of mice bearing MEF/Toff/IGF1R-induced tumors showed an increase of osteopontin levels paralleling tumor growth, and reduction in the serum of mice in which IGF1R expression was shut off and tumor regressed.


Asunto(s)
Biomarcadores de Tumor/sangre , Cromatografía en Gel/métodos , Cromatografía de Fase Inversa/métodos , Electroforesis en Gel de Poliacrilamida/métodos , Proteoma/metabolismo , Proteómica/métodos , Animales , Línea Celular , Ratones , Trasplante de Neoplasias , Osteopontina/metabolismo , Receptor IGF Tipo 1/metabolismo , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
J Immunol Res ; 2020: 7375947, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32832572

RESUMEN

PD-1/PD-L1 blockade has revolutionized the field of immunooncology. Despite the relative success, the response rate to anti-PD-1 therapy requires further improvements. Our aim was to explore the enhancement of T-cell function by using novel PD-1-blocking proteins and compare with clinically approved monoclonal antibodies (mAbs). We isolated T-cells from the ascites and tumor of 17 patients with advanced epithelial ovarian cancer (EOC) and analyzed the effects using the mAbs nivolumab and pembrolizumab and two novel engineered ankyrin repeat proteins (DARPin® proteins). PD-1 blockade with either mAb or DARPin® molecule significantly increased the release of IFN-γ, granzyme B, IL-2, and TNF-α, demonstrating successful reinvigoration. The monovalent DARPin® protein was less effective compared to its bivalent equivalent, demonstrating that bivalency brings an additional benefit to PD-1 blockade. Overall, we found a higher fold increase of lymphokine secretion in response to the PD-1 blockade by tumor-derived T-cells; however, the absolute amounts were significantly lower compared to the release from ascites-derived T-cells. Our results demonstrate that PD-1 blockade can only partially reinvigorate functionally suppressed T-cells from EOC patients. This warrants further investigation preferably in combination with other therapeutics. The study provides an early pilot proof-of-concept for the potential use of DARPin® proteins as eligible alternative scaffold proteins to block PD-1.


Asunto(s)
Anticuerpos Bloqueadores/farmacología , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunomodulación/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Biomarcadores de Tumor , Carcinoma Epitelial de Ovario/inmunología , Carcinoma Epitelial de Ovario/metabolismo , Carcinoma Epitelial de Ovario/patología , Estudios de Casos y Controles , Línea Celular Tumoral , Femenino , Humanos , Clasificación del Tumor , Estadificación de Neoplasias , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo
7.
J Mol Biol ; 372(1): 172-85, 2007 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-17628592

RESUMEN

The concept of novel binding proteins as an alternative to antibodies has undergone rapid development and is now ready for practical use in a wide range of applications. Alternative binding proteins, based on suitable scaffolds with desirable properties, are selected from combinatorial libraries in vitro. Here, we describe an approach using a beta-sheet of human gamma-B-crystallin to generate a universal binding site through randomization of eight solvent-exposed amino acid residues selected according to structural and sequence analyses. Specific variants, so-called Affilin, have been isolated from a phage display library against a variety of targets that differ considerably in size and structure. The isolated Affilin variants can be produced in Escherichia coli as soluble proteins and have a high level of thermodynamic stability. The crystal structures of the human wild-type gamma-B-crystallin and a selected Affilin variant have been determined to 1.7 A and 2.0 A resolution, respectively. Comparison of the two molecules indicates that the human gamma-B-crystallin tolerates amino acid exchanges with no major structural change. We conclude that the intrinsically stable and easily expressed gamma-B-crystallin provides a suitable framework for the generation of novel binding molecules.


Asunto(s)
Proteínas Portadoras/síntesis química , Proteínas Portadoras/aislamiento & purificación , Ingeniería de Proteínas/métodos , gamma-Cristalinas/química , Animales , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Bovinos , Estradiol/metabolismo , Estudios de Factibilidad , Humanos , Inmunoglobulina G/metabolismo , Modelos Moleculares , Factor de Crecimiento Nervioso/metabolismo , Biblioteca de Péptidos , Unión Proteica , Precursores de Proteínas/metabolismo , Estructura Secundaria de Proteína , Especificidad por Sustrato , Testosterona/metabolismo
8.
Oncotarget ; 9(17): 13366-13381, 2018 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-29568363

RESUMEN

The investigational drug MP0250 is a multi-specific DARPin® molecule that simultaneously binds and neutralizes VEGF and HGF with high specificity and affinity. Here we studied the antiangiogenic effects of the MP0250 in multiple myeloma (MM). In endothelial cells (EC) isolated from bone marrow (BM) of MM patients (MMEC) MP0250 reduces VEGFR2 and cMet phosphorylation and affects their downstream signaling cascades. MP0250 influences the secretory profile of MMEC and inhibits their in vitro angiogenic activities (spontaneous and chemotactic migration, adhesion, spreading and capillarogenesis). Compared to anti-VEGF or anti-HGF neutralizing mAbs, MP0250 strongly reduces capillary network formation and vessel-sprouting in a Matrigel angiogenesis assay. MP0250 potentiates the effect of bortezomib in the same in vitro setting. It significantly reduces the number of newly formed vessels in the choriollantoic membrane assay (CAM) and the Matrigel plug assay. In the syngeneic 5T33MM tumor model, MP0250 decreases the microvessel density (MVD) and the combination MP0250/bortezomib lowers the percentage of idiotype positive cells and the serum levels of M-protein. Overall results define MP0250 as a strong antiangiogenic agent with potential as a novel combination drug for treatment of MM patients.

9.
J Leukoc Biol ; 80(4): 719-26, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16864601

RESUMEN

Vascular receptor tyrosine kinases (RTK) have been identified as critical regulatory signaling molecules of developmental and adult vascular morphogenic processes [vascular endothelial growth factor (VEGF) receptors=sprouting; EphB receptors=assembly; Tie2 receptor=maturation and quiescence]. It is intriguing that the same molecules that control the growth of blood and lymphatic vessels play critical roles in the adult to regulate maintenance functions related to vascular homeostasis. VEGF is among the most potent inducers of vascular permeability. The second vascular RTK system, the interaction of paracrine-acting Angiopoietin-1 with its cognate receptor Tie2, acts as an endothelial maintenance and survival-mediating molecular system, which stabilizes the vessel wall and controls endothelial cell quiescence. The third vascular RTK system, the interaction of Eph receptors with their Eph family receptor-interacting protein (ephrin) ligands, transduces positional guidance cues on outgrowing vascular sprouts, which are critical for proper arteriovenous assembly and establishment of blood flow. As such, Eph-ephrin interactions act as an important regulator of cell-cell interactions, exerting propulsive and repulsive functions on neighboring cells and mediating adhesive functions. This review summarizes recent findings related to the roles of the Angiopoietin-Tie and the Eph-ephrin systems as regulators of cell trafficking in the vascular system. The recognition of vascular homeostatic functions of vascular RTKs marks an important change of paradigm in the field of angiogenesis research as it relates angiogenesis-inducing molecules to vascular maintenance functions in the adult. This may also broaden the scope of vascular RTK-targeted therapies.


Asunto(s)
Angiopoyetinas/fisiología , Comunicación Celular/fisiología , Efrinas/fisiología , Receptor TIE-2/fisiología , Receptores de la Familia Eph/fisiología , Animales , Movimiento Celular/fisiología , Humanos , Neovascularización Fisiológica/fisiología , Transducción de Señal/fisiología
10.
Oncotarget ; 8(58): 98371-98383, 2017 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-29228696

RESUMEN

BACKGROUND: The VEGF/VEGFR and the HGF/cMET pathways are key mediators of the interplay of tumor cells and their microenvironment. However, inhibition of VEGF has been shown to produce only limited clinical benefit and inhibition of the activation of cMET by HGF has not translated into clinical benefit in pivotal trials. MP0250, a DARPin® molecule that specifically inhibits both VEGF and HGF has been developed to explore the clinical potential of dual inhibition of these pathways. RESULTS: MP0250 binding to VEGF and HGF inhibited downstream signalling through VEGFR2 and cMET resulting in inhibition of proliferation of VEGF- and HGF-dependent cells. Antitumor activity was demonstrated in VEGF- and HGF-dependent xenograft and syngeneic models with activity superior to that of individual VEGF- and HGF-blocking DARPin® molecules. Combination therapy studies showed potentiation of the antitumor activity of chemotherapy and immunotherapy agents, including an anti-PD1 antibody. MATERIALS AND METHODS: Potency of MP0250 was assessed in cellular models and in a variety of xenograft models as monotherapy or in combination with standard-of-care drugs. CONCLUSIONS: Dual inhibition of VEGF and HGF by MP0250 produced powerful single agent and combination antitumor activity. This, together with increasing understanding of the role of the HGF/cMET pathway in resistance to VEGF (and other agents), supports testing of MP0250 in the clinic.

12.
MAbs ; 9(8): 1262-1269, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29035637

RESUMEN

MP0250 is a multi-domain drug candidate currently being tested in clinical trials for the treatment of cancer. It comprises one anti-vascular endothelial growth factor-A (VEGF-A), one anti-hepatocyte growth factor (HGF), and two anti-human serum albumin (HSA) DARPin® domains within a single polypeptide chain. While there is first clinical validation of a single-domain DARPin® drug candidate, little is known about DARPin® drug candidates comprising multiple domains. Here, we show that MP0250 can be expressed at 15 g/L in soluble form in E. coli high cell-density fermentation, it is stable in soluble/frozen formulation for 2 years as assessed by reverse phase HPLC, it has picomolar potency in inhibiting VEGF-A and HGF in ELISA and cellular assays, and its domains are simultaneously active as shown by surface plasmon resonance. The inclusion of HSA-binding DARPin® domains leads to a favorable pharmacokinetic profile in mouse and cynomolgus monkey, with terminal half-lives of ∼ 30 hours in mouse and ∼ 5 days in cynomolgus monkey. MP0250 is thus a highly potent drug candidate that could be particularly useful in oncology. Beyond MP0250, the properties of MP0250 indicate that multi-domain DARPin® proteins can be valuable next-generation drug candidates.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos/inmunología , Antineoplásicos/inmunología , Proteínas Recombinantes de Fusión/inmunología , Administración Intravenosa , Animales , Repetición de Anquirina/genética , Repetición de Anquirina/inmunología , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacocinética , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Diseño de Fármacos , Femenino , Semivida , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/inmunología , Humanos , Infusiones Intravenosas , Macaca fascicularis , Masculino , Ratones Endogámicos BALB C , Unión Proteica/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacocinética , Albúmina Sérica Humana/genética , Albúmina Sérica Humana/inmunología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/inmunología
13.
Nucleic Acids Res ; 30(11): 2290-8, 2002 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12034815

RESUMEN

TFIIS is a transcription elongation factor for RNA polymerase II (pol II), which can suppress ribonucleotide misincorporation. We reconstituted transcription complexes in a highly purified pol II system on adenovirus Major-Late promoter constructs. We noted that these complexes have a high propensity for read-through upon GTP omission. Read-through occurred during the early stages at all registers analyzed. Addition of TFIIS reversed read-through of productive elongation complexes, which indicated that it was due to misincorporation. However, before register 13 transcription complexes were insensitive to TFIIS. These findings are discussed with respect to the structural models for pol II and we propose that TFIIS action is linked to the RNA:DNA hybrid.


Asunto(s)
ARN Polimerasa II/química , ARN Polimerasa II/metabolismo , Procesamiento Postranscripcional del ARN , Factores Generales de Transcripción , Factores de Transcripción/metabolismo , Transcripción Genética , Factores de Elongación Transcripcional , Adenoviridae/genética , Secuencia de Bases , ADN/genética , ADN/metabolismo , Humanos , Cinética , Sustancias Macromoleculares , Mutación , Ácidos Nucleicos Heterodúplex/biosíntesis , Ácidos Nucleicos Heterodúplex/genética , Ácidos Nucleicos Heterodúplex/metabolismo , Regiones Promotoras Genéticas/genética , ARN/biosíntesis , ARN/genética , ARN/metabolismo , Moldes Genéticos
14.
Arterioscler Thromb Vasc Biol ; 24(10): 1803-9, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15284088

RESUMEN

OBJECTIVE: Angiopoietin-2 (Ang-2) is a non-signal transducing ligand of the endothelial receptor tyrosine kinase Tie-2. Ang-2 is produced by endothelial cells and acts as an autocrine regulator mediating vascular destabilization by inhibiting Angiopoietin-1-mediated Tie-2 activation. To examine the transcriptional regulation of Ang-2, we studied the Ang-2 promoter in endothelial cells and nonendothelial cells. METHODS AND RESULTS: The human Ang-2 promoter contains a 585-bp region around the transcriptional start site (-109 to +476) that is sufficient to control endothelial cell-specific and cytokine-dependent Ang-2 expression. Strong repressor elements of Ang-2-promoter activity are located in the 5'-region of the promoter and in the first intron. The Ets family transcription factors Ets-1 and Elf-1 act as strong enhancers of endothelial cell Ang-2-promoter activity. Ets-binding sites -4 and -7 act as positive regulators, whereas Ets-binding site -3 acts as negative regulator. Demethylation experiments revealed that the Ang-2 gene (in contrast to the Tie-2 gene) is not controlled by imprinting. CONCLUSIONS: The data determine unique positive and negative regulatory mechanisms of endothelial cell Ang-2 expression and provide further evidence for the critical role of Ang-2 as a key autocrine regulator of vascular stability and responsiveness.


Asunto(s)
Angiopoyetina 2/biosíntesis , Células Endoteliales/química , Células Endoteliales/metabolismo , Regiones Promotoras Genéticas/fisiología , Región de Flanqueo 5'/genética , Angiopoyetina 2/genética , Animales , Aorta/citología , Secuencia de Bases/genética , Bovinos , Línea Celular , Línea Celular Tumoral , Clonación Molecular/métodos , Citocinas/fisiología , ADN/genética , Proteínas de Unión al ADN/fisiología , Endotelio Vascular/química , Endotelio Vascular/metabolismo , Impresión Genómica/genética , Humanos , Riñón/química , Riñón/embriología , Riñón/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares , Proteína Proto-Oncogénica c-ets-1 , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-ets , Factores de Transcripción/fisiología , Sitio de Iniciación de la Transcripción
15.
Curr Eye Res ; 30(4): 249-57, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15823917

RESUMEN

PURPOSE: The purpose of this study was to develop pharmacological therapeutic alternatives for ischemia-induced proliferative retinopathy. METHODS: C57BL/6J mice were placed in 76% oxygen on postnatal day 7 (P7) for 5 days. On P12 recombinant, chimeric vascular endothelial growth factor (sVEGF-R2) or sTie2 was injected intravitreally in one eye. The fellow eye received a control injection. On P17, retinal wholemounts were prepared after perfusion with fluorescein-dextran to quantify the retinopathy. RESULTS: A single intravitreal injection of sVEGF-R2 reduced pathologic vascular changes significantly (p = 0.02). No significant effect was observed after intravitreal application of sTie2 (p = 0.07), although Ang-2 was upregulated in control animals without treatment as neovascularization developed and Ang-1 was constantly transcribed (ratio PCR). CONCLUSIONS: sVEGF-R2 interferes with VEGF signaling via VEGF-R2 receptor. Thus, local application of soluble receptors for angiogenic factors is a possible therapy for proliferative retinopathy. Receptors with a wide range of ligands might prove more useful for local application than those binding few or antagonistic ligands.


Asunto(s)
Angiopoyetina 1/análogos & derivados , Receptor TIE-2/administración & dosificación , Neovascularización Retiniana/prevención & control , Receptor 2 de Factores de Crecimiento Endotelial Vascular/administración & dosificación , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Angiopoyetina 2/genética , Angiopoyetina 2/metabolismo , Animales , Dextranos , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Angiografía con Fluoresceína , Fluoresceínas , Inyecciones , Ratones , Ratones Endogámicos C57BL , Oxígeno/toxicidad , ARN Mensajero/metabolismo , Receptor TIE-2/metabolismo , Receptores Fc , Proteínas Recombinantes de Fusión , Neovascularización Retiniana/inducido químicamente , Neovascularización Retiniana/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Cuerpo Vítreo
16.
PLoS One ; 8(8): e70459, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23940579

RESUMEN

Genetic experiments (loss-of-function and gain-of-function) have established the role of Angiopoietin/Tie ligand/receptor tyrosine kinase system as a regulator of vessel maturation and quiescence. Angiopoietin-2 (Ang-2) acts on Tie2-expressing resting endothelial cells as an antagonistic ligand to negatively interfere with the vessel stabilizing effects of constitutive Ang-1/Tie-2 signaling. Ang-2 thereby controls the vascular response to inflammation-inducing as well as angiogenesis-inducing cytokines. This study was aimed at assessing the role of Ang-2 as an autocrine (i.e. endothelial-derived) regulator of rapid vascular responses (within minutes) caused by permeability-inducing agents. Employing two independent in vivo assays to quantitatively assess vascular leakage (tracheal microsphere assay, 1-5 min and Miles assay, 20 min), the immediate vascular response to histamine, bradykinin and VEGF was analyzed in Ang-2-deficient (Ang-2(-/-)) mice. In comparison to the wild type control mice, the Ang2(-/-) mice demonstrated a significantly attenuated response. The Ang-2(-/-) phenotype was rescued by systemic administration (paracrine) of an adenovirus encoding Ang-2. Furthermore, cytokine-induced intracellular calcium influx was impaired in Ang-2(-/-) endothelioma cells, consistent with reduced phospholipase activation in vivo. Additionally, recombinant human Ang-2 (rhAng-2) alone was unable to induce vascular leakage. In summary, we report here in a definite genetic setting that Ang-2 is critical for multiple vascular permeability-inducing cytokines.


Asunto(s)
Angiopoyetina 2/metabolismo , Citocinas/farmacología , Angiopoyetina 2/genética , Animales , Western Blotting , Bradiquinina/genética , Bradiquinina/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/genética , Células Cultivadas , Femenino , Células Endoteliales de la Vena Umbilical Humana , Pulmón/citología , Pulmón/metabolismo , Ratones , Ratones Noqueados , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Am J Ophthalmol ; 155(4): 697-704, 704.e1-2, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23218689

RESUMEN

PURPOSE: To evaluate the safety and bioactivity of MP0112, a designed ankyrin repeat protein (DARPin) that specifically binds vascular endothelial growth factor (VEGF) in patients with diabetic macular edema (DME). DARPins are a novel class of proteins selected for specific, high-affinity binding to a target protein. DESIGN: Phase I/II, open-label, multicenter dose-escalation trial. METHODS: After a single intravitreal injection of MP0112, the main outcomes were safety assessments, aqueous MP0112 levels, change in best-corrected visual acuity (BCVA), and foveal thickness measured by optical coherence tomography. Six cohorts were planned, but only 3 were enrolled (0.04, 0.15, 0.4 mg), because a maximally tolerated dose of 1.0 mg was identified in a parallel age-related macular degeneration trial. RESULTS: Median aqueous concentration of MP0112 was 555 nM 1 week and >10 nM in 3 of 4 patients 12 weeks post injection of 0.4 mg. Median BCVA improvement at week 12 was 4, 6, and 10 letters in cohorts 1, 2, and 3. Ocular inflammation was observed in 11 patients (61%) and was severe in 1. High-resolution chromatography separated proinflammatory impurities from MP0112, resulting in a new formulation. CONCLUSIONS: A single intraocular injection of 0.4 mg MP0112 resulted in levels above the half-maximal inhibitory concentration and neutralization of VEGF in aqueous humor for 8-12 weeks. Despite inflammation in several patients, there was prolonged edema reduction and improvement in vision in several patients. The source of the inflammation was eliminated from a new preparation that is being tested in an ongoing clinical trial.


Asunto(s)
Repetición de Anquirina , Retinopatía Diabética/tratamiento farmacológico , Edema Macular/tratamiento farmacológico , Péptidos/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/metabolismo , Anciano , Anciano de 80 o más Años , Humor Acuoso/metabolismo , Disponibilidad Biológica , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Retinopatía Diabética/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Fóvea Central/patología , Humanos , Inyecciones Intravítreas , Edema Macular/metabolismo , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Péptidos/efectos adversos , Péptidos/farmacocinética , Unión Proteica , Tomografía de Coherencia Óptica , Agudeza Visual/fisiología
18.
Anticancer Res ; 32(7): 2399-406, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22753696

RESUMEN

BACKGROUND/AIM: Sequential treatment with targeted agents is standard of care for patients with metastatic renal cell carcinoma (mRCC). However, clinical data directly comparing treatment outcomes with a mammalian target of rapamycin inhibitor or a vascular endothelial growth factor-targeted agent in the second-line setting are lacking. We evaluated sequential treatment in a syngeneic, orthotopic mouse model of mRCC. MATERIALS AND METHODS: BALB/c mice were orthotopically implanted with murine RCC (RENCA) cells expressing luciferase and randomized to vehicle, sunitinib, sunitinib followed by sorafenib, or sunitinib followed by everolimus. Tumor growth and metastases were assessed by in vivo (whole body) and ex vivo (primary tumor, lung, liver) luciferase activity and necropsies, performed on day 20 or 46 for vehicle and treatment groups, respectively. RESULTS: Sunitinib followed by everolimus was associated with reduced luciferase activity and primary tumor weight and volume compared with sunitinib, and sunitinib followed by sorafenib. CONCLUSION: Sequential therapy with sunitinib followed by everolimus demonstrated significant antitumor and anti-metastatic effects.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Células Renales/tratamiento farmacológico , Neoplasias Renales/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Esquema de Medicación , Everolimus , Femenino , Indoles/administración & dosificación , Indoles/efectos adversos , Neoplasias Renales/enzimología , Neoplasias Renales/patología , Luciferasas/biosíntesis , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes/métodos , Ratones , Ratones Endogámicos BALB C , Pirroles/administración & dosificación , Pirroles/efectos adversos , Distribución Aleatoria , Sirolimus/administración & dosificación , Sirolimus/efectos adversos , Sirolimus/análogos & derivados , Sunitinib
19.
Cardiovasc Res ; 94(3): 510-8, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22505659

RESUMEN

AIMS: Adequate endothelial cell stimulation is a prerequisite for the adaptive remodelling of macro- and microvessels. A pivotal autocrine mechanism following endothelial cell activation is the release of angiopoietin-2 (Ang-2), which subsequently antagonizes the binding of Ang-1 to the Tie-2 receptor, thus sensitizing the endothelial cells to pro-angiogenic and/or pro-inflammatory stimuli. Based on the observation that hypertension in mice reduces the abundance of Ang-2 stored in arterial endothelial cells, this study was aimed at testing the hypothesis that an increase in wall stress (WS) or stretch-a hallmark of hypertension-is sufficient to release Ang-2 from endothelial cells. METHODS AND RESULTS: In fact, stretching of isolated perfused mouse arteries or human cultured endothelial cells rapidly elicited an increased release of Ang-2. In the cultured endothelial cells, this was preceded by a transient rise in intracellular free calcium, abrogated through calcium chelation and accompanied by a decrease in Tie-2 phosphorylation. Interestingly, Ang-1 abolished the stretch-induced release of Ang-2 from both cultured and native endothelial cells through inhibiting the stretch-dependent mobilization of intracellular calcium. CONCLUSION: Collectively, these results indicate that increased WS or stretch facilitates the release of Ang-2 from endothelial cell Weibel-Palade bodies, and that Ang-1 can block this by attenuating the stretch-mediated rise in intracellular calcium.


Asunto(s)
Angiopoyetina 1/farmacología , Angiopoyetina 2/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Hipertensión/fisiopatología , Animales , Calcio/metabolismo , Células Cultivadas , Endotelio Vascular/citología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor TIE-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA