Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Curr Issues Mol Biol ; 46(4): 3278-3293, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38666935

RESUMEN

Protein S (PROS1) is a vitamin K-dependent anticoagulant factor, which also acts as an agonist for the TYRO3, AXL, and MERTK (TAM) tyrosine kinase receptors. PROS1 is produced by the endothelium which also expresses TAM receptors, but little is known about its effects on vascular function and permeability. Transwell permeability assays as well as Western blotting and immunostaining analysis were used to monitor the possible effects of PROS1 on both endothelial cell permeability and on the phosphorylation state of specific signaling proteins. We show that human PROS1, at its circulating concentrations, substantially increases both the basal and VEGFA-induced permeability of endothelial cell (EC) monolayers. PROS1 induces p38 MAPK (Mitogen Activated Protein Kinase), Rho/ROCK (Rho-associated protein kinase) pathway activation, and actin filament remodeling, as well as substantial changes in Vascular Endothelial Cadherin (VEC) distribution and its phosphorylation on Ser665 and Tyr685. It also mediates c-Src and PAK-1 (p21-activated kinase 1) phosphorylation on Tyr416 and Ser144, respectively. Exposure of EC to human PROS1 induces VEC internalization as well as its cleavage into a released fragment of 100 kDa and an intracellular fragment of 35 kDa. Using anti-TAM neutralizing antibodies, we demonstrate that PROS1-induced VEC and c-Src phosphorylation are mediated by both the MERTK and TYRO3 receptors but do not involve the AXL receptor. MERTK and TYRO3 receptors are also responsible for mediating PROS1-induced MLC (Myosin Light Chain) phosphorylation on a site targeted by the Rho/ROCK pathway. Our report provides evidence for the activation of the c-Src/VEC and Rho/ROCK/MLC pathways by PROS1 for the first time and points to a new role for PROS1 as an endogenous vascular permeabilizing factor.

2.
Pharm Res ; 35(4): 90, 2018 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-29520495

RESUMEN

PURPOSE: This work explores synthesis of novel cholesterol derivative for the preparation of cationic liposomes and its interaction with Paclitaxel (PTX) within liposome membrane using molecular dynamic (MD) simulation and in-vitro studies. METHODS: Cholesteryl Arginine Ethylester (CAE) was synthesized and characterized. Cationic liposomes were prepared using Soy PC (SPC) at a molar ratio of 77.5:15:7.5 of SPC/CAE/PTX. Conventional liposomes were composed of SPC/cholesterol/PTX (92:5:3 M ratio). The interaction between paclitaxel, ligand and the membrane was studied using 10 ns MD simulation. The interactions were studied using Differential Scanning Calorimetry (DSC) and Small Angle Neutron Scattering analysis. The efficacy of liposomes was evaluated by MTT assay and endothelial cell migration assay on different cell lines. The safety of the ligand was determined using the Comet Assay. RESULTS: The cationic liposomes improved loading efficiency and stability compared to conventional liposomes. The increased PTX loading could be attributed to the hydrogen bond between CAE and PTX and deeper penetration of PTX in the bilayer. The DSC study suggested that inclusion of CAE in the DPPC bilayer eliminates Tg. SANS data showed that CAE has more pronounced membrane thickening effect as compared to cholesterol. The cationic liposomes showed slightly improved cytotoxicity in three different cell lines and improved endothelial cell migration inhibition compared to conventional liposomes. Furthermore, the COMET assay showed that CAE alone does not show any genotoxicity. CONCLUSIONS: The novel cationic ligand (CAE) retains paclitaxel within the phospholipid bilayer and helps in improved drug loading and physical stability. Graphical Abstract ᅟ.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Colesterol/análogos & derivados , Composición de Medicamentos/métodos , Diseño de Fármacos , Paclitaxel/administración & dosificación , Animales , Antineoplásicos Fitogénicos/química , Cationes/química , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Colesterol/química , Ensayos de Selección de Medicamentos Antitumorales , Estabilidad de Medicamentos , Células Endoteliales , Humanos , Liposomas , Ratones , Simulación de Dinámica Molecular , Paclitaxel/química
3.
Am J Hematol ; 90(7): 608-17, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25808486

RESUMEN

Thrombocytopenia and thromboembolism(s) may develop in heparin immune-mediated thrombocytopenia (HIT) patients after reexposure to heparin. At the Onassis Cardiac Surgery Center, 530 out of 17,000 patients requiring heart surgery over an 11-year period underwent preoperative HIT assessment by ELISA and a three-point heparin-induced platelet aggregation assay (HIPAG). The screening identified 110 patients with HIT-reactive antibodies, out of which 46 were also thrombocytopenic (true HIT). Cardiac surgery was performed in HIT-positive patients under heparin anticoagulation and iloprost infusion. A control group of 118 HIT-negative patients received heparin but no iloprost during surgery. For the first 20 patients, the dose of iloprost diminishing the HIPAG test to ≤5% was determined prior to surgery by in vitro titration using the patients' own plasma and donor platelets. In parallel, the iloprost "target dose" was also established for each patient intraoperatively, but before heparin administration. Iloprost was infused initially at 3 ng/kg/mL and further adjusted intraoperatively, until ex vivo aggregation reached ≤5%. As a close correlation was observed between the "target dose" identified before surgery and that established intraoperatively, the remaining 90 patients were administered iloprost starting at the presurgery identified "target dose." This process significantly reduced the number of intraoperative HIPAG reassessments needed to determine the iloprost target dose, and reduced surgical time, while maintaining similar primary clinical outcomes to controls. Therefore, infusion of iloprost throughout surgery, under continuous titration, allows cardiac surgery to be undertaken safely using heparin, while avoiding life-threatening iloprost-induced hypotension in patients diagnosed with HIT-reactive antibodies or true HIT.


Asunto(s)
Anticuerpos/sangre , Fármacos Cardiovasculares/uso terapéutico , Iloprost/uso terapéutico , Trombocitopenia/patología , Tromboembolia/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Anticoagulantes/efectos adversos , Aneurisma de la Aorta/patología , Aneurisma de la Aorta/cirugía , Plaquetas/efectos de los fármacos , Plaquetas/inmunología , Plaquetas/patología , Anuloplastia de la Válvula Cardíaca/métodos , Puente de Arteria Coronaria/métodos , Esquema de Medicación , Monitoreo de Drogas , Femenino , Heparina/efectos adversos , Humanos , Masculino , Persona de Mediana Edad , Atención Perioperativa/métodos , Agregación Plaquetaria/efectos de los fármacos , Recuento de Plaquetas , Trombocitopenia/inducido químicamente , Trombocitopenia/inmunología , Tromboembolia/inmunología , Tromboembolia/patología , Resultado del Tratamiento
4.
Blood ; 120(25): 5073-83, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23065156

RESUMEN

Protein S is a vitamin K-dependent glycoprotein, which, besides its anticoagulant function, acts as an agonist for the tyrosine kinase receptors Tyro3, Axl, and Mer. The endothelium expresses Tyro3, Axl, and Mer and produces protein S. The interaction of protein S with endothelial cells and particularly its effects on angiogenesis have not yet been analyzed. Here we show that human protein S, at circulating concentrations, inhibited vascular endothelial growth factor (VEGF) receptor 2-dependent vascularization of Matrigel plugs in vivo and the capacity of endothelial cells to form capillary-like networks in vitro as well as VEGF-A-induced endothelial migration and proliferation. Furthermore, protein S inhibited VEGF-A-induced endothelial VEGFR2 phosphorylation and activation of mitogen-activated kinase-Erk1/2 and Akt. Protein S activated the tyrosine phosphatase SHP2, and the SHP2 inhibitor NSC 87877 reversed the observed inhibition of VEGF-A-induced endothelial proliferation. Using siRNA directed against Tyro3, Axl, and Mer, we demonstrate that protein S-mediated SHP2 activation and inhibition of VEGF-A-stimulated proliferation were mediated by Mer. Our report provides the first evidence for the existence of a protein S/Mer/SHP2 axis, which inhibits VEGFR2 signaling, regulates endothelial function, and points to a role for protein S as an endogenous angiogenesis inhibitor.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Neovascularización Fisiológica , Proteína S/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Proliferación Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Activación Enzimática , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Proteína S/administración & dosificación , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/genética , Proteínas Tirosina Quinasas Receptoras/genética , Tirosina Quinasa c-Mer
5.
Nat Rev Cancer ; 5(6): 423-35, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15928673

RESUMEN

Low-molecular-weight vascular-disrupting agents (VDAs) cause a pronounced shutdown in blood flow to solid tumours, resulting in extensive tumour-cell necrosis, while they leave the blood flow in normal tissues relatively intact. The largest group of VDAs is the tubulin-binding combretastatins, several of which are now being tested in clinical trials. DMXAA (5,6-dimethylxanthenone-4-acetic acid) - one of a structurally distinct group of drugs - is also being tested in clinical trials. A full understanding of the action of these and other VDAs will provide insights into mechanisms that control tumour blood flow and will be the basis for the development of new therapeutic drugs for targeting the established tumour vasculature for therapy.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/uso terapéutico , Bibencilos/farmacología , Bibencilos/uso terapéutico , Permeabilidad Capilar , Ensayos Clínicos como Asunto , Células Endoteliales/fisiología , Humanos , Estilbenos/farmacología , Estilbenos/uso terapéutico , Xantonas/farmacología , Xantonas/uso terapéutico
6.
Int J Cancer ; 133(11): 2563-76, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23712501

RESUMEN

Antiangiogenic therapy based on blocking the actions of vascular endothelial growth factor-A (VEGF) can lead to "normalization" of blood vessels in both animal and human tumors. Differential expression of VEGF isoforms affects tumor vascular maturity, which could influence the normalization process and response to subsequent treatment. Fibrosarcoma cells expressing only VEGF120 or VEGF188 isoforms were implanted either subcutaneously (s.c.) or in dorsal skin-fold "window" chambers in SCID mice. VEGF120 was associated with vascular fragility and hemorrhage. Tumor-bearing mice were treated with repeat doses of SU5416, an indolinone receptor tyrosine kinase inhibitor with activity against VEGFR-2 and proven preclinical ability to induce tumor vascular normalization. SU5416 reduced vascularization in s.c. implants of both VEGF120 and VEGF188 tumors. However, in the window chamber, SU5416 treatment increased red cell velocity in VEGF120 (representing vascular normalization) but not VEGF188 tumors. SU5416 treatment had no effect on growth or necrosis levels in either tumor type but tended to counteract the increase in interstitial fluid pressure seen with growth of VEGF120 tumors. SU5416 pretreatment resulted in the normally fragile blood vessels in VEGF120-expressing tumors becoming resistant to the vascular damaging effects of the tubulin-binding vascular disrupting agent (VDA), combretastatin A4 3-O-phosphate (CA4P). Thus, vascular normalization induced by antiangiogenic treatment can reduce the efficacy of subsequent VDA treatment. Expression of VEGF120 made tumors particularly susceptible to vascular normalization by SU5416, which in turn made them resistant to CA4P. Therefore, VEGF isoform expression may be useful for predicting response to both antiangiogenic and vascular-disrupting therapy.


Asunto(s)
Fibrosarcoma/genética , Isoformas de Proteínas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Fibrosarcoma/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Indoles/farmacología , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Isoformas de Proteínas/genética , Pirroles/farmacología , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
7.
Proc Natl Acad Sci U S A ; 107(9): 3966-71, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20150514

RESUMEN

This study describes the previously unreported intrinsic capacity of poly-L-lysine (PLL) sixth generation (G(6)) dendrimer molecules to exhibit systemic antiangiogenic activity that could lead to solid tumor growth arrest. The PLL-dendrimer-inhibited tubule formation of SVEC4-10 murine endothelial cells and neovascularization in the chick embryo chick chorioallantoic membrane (CAM) assay. Intravenous administration of the PLL-dendrimer molecules into C57BL/6 mice inhibited vascularisation in Matrigel plugs implanted subcutaneously. Antiangiogenic activity was further evidenced using intravital microscopy of tumors grown within dorsal skinfold window chambers. Reduced vascularization of P22 rat sarcoma implanted in the dorsal window chamber of SCID mice was observed following tail vein administration (i.v.) of the PLL dendrimers. Also, the in vivo toxicological profile of the PLL-dendrimer molecules was shown to be safe at the dose regime studied. The antiangiogenic activity of the PLL dendrimer was further shown to be associated with significant suppression of B16F10 solid tumor volume and delayed tumor growth. Enhanced apoptosis/necrosis within tumors of PLL-dendrimer-treated animals only and reduction in the number of CD31 positive cells were observed in comparison to protamine treatment. This study suggests that PLL-dendrimer molecules can exhibit a systemic antiangiogenic activity that may be used for therapy of solid tumors, and in combination with their capacity to carry other therapeutic or diagnostic agents may potentially offer capabilities for the design of theranostic systems.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , División Celular/efectos de los fármacos , Dendrímeros , Neoplasias Experimentales/patología , Polilisina/farmacología , Animales , Cationes , Ratones , Ratones Endogámicos C57BL , Ratones SCID
8.
Biosensors (Basel) ; 13(7)2023 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-37504117

RESUMEN

We present a critical review of the current progress in wearable insulin biosensors. For over 40 years, glucose biosensors have been used for diabetes management. Measurement of blood glucose is an indirect method for calculating the insulin administration dosage, which is critical for insulin-dependent diabetic patients. Research and development efforts aiming towards continuous-insulin-monitoring biosensors in combination with existing glucose biosensors are expected to offer a more accurate estimation of insulin sensitivity, regulate insulin dosage and facilitate progress towards development of a reliable artificial pancreas, as an ultimate goal in diabetes management and personalised medicine. Conventional laboratory analytical techniques for insulin detection are expensive and time-consuming and lack a real-time monitoring capability. On the other hand, biosensors offer point-of-care testing, continuous monitoring, miniaturisation, high specificity and sensitivity, rapid response time, ease of use and low costs. Current research, future developments and challenges in insulin biosensor technology are reviewed and assessed. Different insulin biosensor categories such as aptamer-based, molecularly imprinted polymer (MIP)-based, label-free and other types are presented among the latest developments in the field. This multidisciplinary field requires engagement between scientists, engineers, clinicians and industry for addressing the challenges for a commercial, reliable, real-time-monitoring wearable insulin biosensor.


Asunto(s)
Técnicas Biosensibles , Diabetes Mellitus , Dispositivos Electrónicos Vestibles , Humanos , Insulina/uso terapéutico , Técnicas Biosensibles/métodos , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/tratamiento farmacológico , Glucosa
9.
Int J Cancer ; 129(8): 1979-89, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21154772

RESUMEN

Vascular-targeted therapeutics are increasingly used in the clinic. However, less is known about the direct response of tumor cells to these agents. We have developed a combretastatin-A-4-phosphate (CA4P) resistant variant of SW1222 human colorectal carcinoma cells to examine the relative importance of vascular versus tumor cell targeting in the ultimate treatment response. SW1222(Res) cells were generated through exposure of wild-type cells (SW1222(WT) ) to increasing CA4P concentrations in vitro. Increased resistance was confirmed through analyses of cell viability, apoptosis and multidrug-resistance (MDR) protein expression. In vivo, comparative studies examined tumor cell necrosis, apoptosis, vessel morphology and functional vascular end-points following treatment with CA4P (single 100 mg/kg dose). Tumor response to repeated CA4P dosing (50 mg/kg/day, 5 days/week for 2 weeks) was examined through growth measurement, and ultimate tumor cell survival was studied by ex vivo clonogenic assay. In vitro, SW1222(Res) cells showed reduced CA4P sensitivity, enhanced MDR protein expression and a reduced apoptotic index. In vivo, CA4P induced significantly lower apoptotic cell death in SW1222(Res) versus SW1222(WT) tumors indicating maintenance of resistance characteristics. However, CA4P-induced tumor necrosis was equivalent in both lines. Similarly, rapid CA4P-mediated vessel disruption and blood flow shut-down were observed in both lines. Cell surviving fraction was comparable in the two tumor types following single dose CA4P and SW1222(Res) tumors were at least as sensitive as SW1222(WT) tumors to repeated dosing. Despite tumor cell resistance to CA4P, SW1222(Res) response in vivo was not impaired, strongly supporting the view that vascular damage dominates the therapeutic response to this agent.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/tratamiento farmacológico , Estilbenos/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/análisis , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones , Ratones SCID , Neovascularización Patológica/tratamiento farmacológico , Flujo Sanguíneo Regional/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Trends Endocrinol Metab ; 31(6): 398-409, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32396842

RESUMEN

In this opinion article we critically assess evidence for the existence of a family of antiangiogenic vascular endothelial growth factor (Vegfaxxxb) transcripts, arising from the use of a phylogenetically conserved alternative distal splice site within exon 8 of the VEGFA gene. We explain that prior evidence for Vegfaxxxb transcripts in tissues rests heavily upon flawed RT-PCR methodologies, with the extensive use of 5'-tailing in primer design being the main issue. Furthermore, our analysis of large RNA-seq data sets (human and ovine) fails to identify a single Vegfaxxxb transcript. Therefore, we challenge the very existence of Vegfaxxxb transcripts, which further questions the physiological relevance of studies based on the use of 'anti-VEGFAxxxb' antibodies. Our analysis has implications for the proposed therapeutic use of isoform-specific anti-VEGFA strategies for treating cancer and retinopathies.


Asunto(s)
Empalme Alternativo , Inhibidores de la Angiogénesis , Reacción en Cadena de la Polimerasa/normas , Análisis de Secuencia de ARN/normas , Factor A de Crecimiento Endotelial Vascular , Empalme Alternativo/genética , Humanos , Isoformas de Proteínas , Factor A de Crecimiento Endotelial Vascular/genética
11.
Sci Rep ; 10(1): 9926, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32555222

RESUMEN

Combretastatin A-4 phosphate (CA4P) is a microtubule-disrupting tumour-selective vascular disrupting agent (VDA). CA4P activates the actin-regulating RhoA-GTPase/ ROCK pathway, which is required for full vascular disruption. While hypoxia renders tumours resistant to many conventional therapies, little is known about its influence on VDA activity. Here, we found that active RhoA and ROCK effector phospho-myosin light chain (pMLC) were downregulated in endothelial cells by severe hypoxia. CA4P failed to activate RhoA/ROCK/pMLC but its activity was restored upon reoxygenation. Hypoxia also inhibited CA4P-mediated actinomyosin contractility, VE-cadherin junction disruption and permeability rise. Glucose withdrawal downregulated pMLC, and coupled with hypoxia, reduced pMLC faster and more profoundly than hypoxia alone. Concurrent inhibition of glycolysis (2-deoxy-D-glucose, 2DG) and mitochondrial respiration (rotenone) caused profound actin filament loss, blocked RhoA/ROCK signalling and rendered microtubules  CA4P-resistant. Withdrawal of the metabolism inhibitors restored the cytoskeleton and CA4P activity. The AMP-activated kinase AMPK was investigated as a potential mediator of pMLC downregulation. Pharmacological AMPK activators that generate AMP, unlike allosteric activators, downregulated pMLC but only when combined with 2DG and/or rotenone. Altogether, our results suggest that Rho/ROCK and actinomyosin contractility are regulated by AMP/ATP levels independently of AMPK, and point to hypoxia/energy depletion as potential modifiers of CA4P response.


Asunto(s)
Endotelio Vascular/patología , Hipoxia/fisiopatología , Neovascularización Patológica/patología , Estilbenos/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Actinas/metabolismo , Antineoplásicos Fitogénicos/farmacología , Permeabilidad de la Membrana Celular , Endotelio Vascular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neovascularización Patológica/inducido químicamente , Transducción de Señal , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
12.
Int J Exp Pathol ; 90(3): 284-94, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19563611

RESUMEN

Vascular disrupting agents (VDAs) are a relatively new group of 'vascular targeting' agents that exhibit selective activity against established tumour vascular networks, causing severe interruption of tumour blood flow and necrosis to the tumour mass. Microtubule depolymerizing agents form by far the largest group of small molecular weight VDAs many of which, including lead compound disodium combretastatin A-4 3-O-phosphate (CA-4-P), are under clinical development for cancer. Although distinct from the angiogenesis inhibitors, VDAs can also interfere with angiogenesis and therefore constitute a potential group of novel drugs for the treatment of pathological conditions characterized by excessive angiogenesis, in addition to cancer. The endothelial cytoskeleton is the primary cellular target of this family of drugs, and some progress in understanding the molecular and signalling mechanisms associated with their endothelial disrupting activity has been made in the last few years. Susceptibility of tumour vessels to VDA damage is ascribed to their immature pericyte-defective nature, although the exact molecular mechanisms involved have not been clearly defined. Despite causing profound damage to tumours, VDAs fail to halt tumour growth unless used together with conventional treatments. This failure is attributed to resistance mechanisms, primarily associated with cells that remain viable within the tumour rim, and enhanced angiogenesis. The focus is now to understand mechanisms of susceptibility and resistance to identify novel molecular targets and develop strategies that are more effective.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , Moduladores de Tubulina/uso terapéutico , Antineoplásicos Fitogénicos/uso terapéutico , Bibencilos/uso terapéutico , Resistencia a Antineoplásicos , Humanos , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico
13.
Br J Radiol ; 92(1093): 20180405, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30160184

RESUMEN

The development of blood vessels by the process of angiogenesis underpins the growth and metastasis of many tumour types. Various angiogenesis inhibitors targeted against vascular endothelial growth factor A (VEGF-A) and its receptors have entered the clinic more than a decade ago. However, despite substantial clinical improvements, their overall efficacy proved to be significantly lower than many of the pre-clinical studies had predicted. Antiangiogenic agents have been combined with chemotherapy, radiotherapy and more recently immunotherapy in many pre-clinical and clinical studies in an effort to improve their efficacy. To date, only their use alongside chemotherapy is approved as part of standard treatment protocols. Most pre-clinical studies have reported improved tumour control from the addition of antiangiogenic therapies to radiotherapy and progress has been made in unravelling the complex mechanisms through which VEGF inhibition potentiates radiotherapy responses. However, the efficacy of this combination is variable, and many questions still remain as to how best to administer the two modalities to achieve optimal response and minimal toxicity. One important limiting factor is that, unlike some other targeted therapies, antiangiogenic agents are not administered to selected patient populations, since biomarkers for identifying responders have not yet been established. Here, we outline VEGF biology and review current approaches that aim to identify biomarkers for stratifying patients for treatment with angiogenesis inhibitors. We also discuss current progress in elucidating mechanisms of interaction between radiotherapy and VEGF inhibitors. Ongoing clinical trials will determine whether these combinations will ultimately improve treatment outcomes for cancer patients.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Neoplasias/irrigación sanguínea , Neoplasias/terapia , Neovascularización Patológica/prevención & control , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Animales , Bevacizumab/administración & dosificación , Terapia Combinada/métodos , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Masculino , Pronóstico , Radioterapia/métodos , Resultado del Tratamiento , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de la radiación , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
ChemMedChem ; 13(24): 2618-2626, 2018 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-30281922

RESUMEN

The combretastatins have attracted significant interest as small-molecule therapies for cancer due to their ability to function as vascular disrupting agents. We have successfully prepared a range of combretastatin analogues that are based on a novel sydnone heterocycle core, and their potential as tubulin binders has been assessed in vitro and in vivo. The most potent candidate was found to disrupt microtubules and affect cellular morphology at sub-micromolar levels. Moreover, it was found to bind reversibly to tubulin and significantly increase endothelial cell monolayer permeability, in a similar manner to combretastatin A4. Surprisingly, the compound did not exhibit efficacy in vivo, possibly due to rapid metabolism.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Estilbenos/farmacología , Sidnonas/farmacología , Inhibidores de la Angiogénesis/química , Animales , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones SCID , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Unión Proteica , Estilbenos/química , Relación Estructura-Actividad , Sidnonas/química , Tubulina (Proteína)/metabolismo
15.
Int J Radiat Oncol Biol Phys ; 69(5): 1553-62, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17920784

RESUMEN

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


Asunto(s)
Citoesqueleto/efectos de la radiación , Células Endoteliales/efectos de la radiación , Actinas/efectos de la radiación , Cadherinas/efectos de la radiación , Muerte Celular/efectos de la radiación , Células Cultivadas , Dextranos/farmacocinética , Células Endoteliales/ultraestructura , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Endotelio Vascular/efectos de la radiación , Humanos , Filamentos Intermedios/efectos de la radiación , Microtúbulos/efectos de la radiación , Proteína Quinasa 11 Activada por Mitógenos/fisiología , Permeabilidad , Tolerancia a Radiación , Transducción de Señal/fisiología , Transducción de Señal/efectos de la radiación , Proteína de Unión al GTP rhoA/fisiología
16.
Mol Cancer Ther ; 5(12): 3145-52, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17172418

RESUMEN

Electroporation-based therapies, such as electrochemotherapy and electrogene therapy, result in the disruption of blood vessel networks in vivo and cause changes in blood flow and vascular permeability. The effects of electroporation on the cytoskeleton of cultured primary endothelial cells and on endothelial monolayer permeability were investigated to elucidate possible mechanisms involved. Human umbilical vein endothelial cells (HUVECs) were electroporated in situ and then immunofluorescence staining for filamentous actin, beta-tubulin, vimentin, and VE-cadherin as well as Western blotting analysis of levels of phosphorylated myosin light chain and cytoskeletal proteins were performed. Endothelial permeability was determined by monitoring the passage of FITC-coupled dextran through endothelial monolayers. Exposure of endothelial cells to electric pulses resulted in a profound disruption of microfilament and microtubule cytoskeletal networks, loss of contractility, and loss of vascular endothelial cadherin from cell-to-cell junctions immediately after electroporation. These effects were voltage dependent and reversible because cytoskeletal structures recovered within 60 min of electroporation with up to 40 V, without any significant loss of cell viability. The cytoskeletal effects of electroporation were paralleled by a rapid increase in endothelial monolayer permeability. These results suggest that the remodeling of the endothelial cytoskeleton and changes in endothelial barrier function could contribute to the vascular disrupting actions of electroporation-based therapies and provide an insight into putative mechanisms responsible for the observed increase in permeability and cessation of blood flow in vivo.


Asunto(s)
Citoesqueleto/fisiología , Electroporación/métodos , Células Endoteliales/fisiología , Neoplasias/terapia , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Antígenos CD/metabolismo , Cadherinas/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Células Endoteliales/citología , Humanos , Filamentos Intermedios/metabolismo , Microtúbulos/metabolismo , Miosinas/metabolismo , Neoplasias/metabolismo
17.
Oncotarget ; 8(56): 95648-95661, 2017 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-29221156

RESUMEN

Solid tumours vary in sensitivity to the vascular disrupting agent combretastatin A-4 3-O-phosphate (CA4P), but underlying factors are poorly understood. The signaling sphingolipid, sphingosine-1-phosphate (S1P), promotes vascular barrier integrity by promoting assembly of VE-cadherin/ß-catenin complexes. We tested the hypothesis that tumour pre-treatment with S1P would render tumours less susceptible to CA4P. S1P (1µM) pretreatment attenuated an increase in endothelial cell (HUVEC) monolayer permeability induced by 10µM CA4P. Intravenously administered S1P (8mg/kg/hr for 20 minutes then 2mg/kg/hr for 40 minutes), reduced CA4P-induced (30mg/kg) blood flow shut-down in fibrosarcoma tumours in SCID mice (n≥7 per group), as measured by tumour retention of an intravenously administered fluorescent lectin. A trend towards in vivo protection was also found using laser Doppler flowmetry. Immunohistochemical staining of tumours ex vivo revealed disrupted patterns of VE-cadherin in vasculature of mice treated with CA4P, which were decreased by pretreatment with S1P. S1P treatment also stabilized N-cadherin junctions between endothelial cells and smooth muscle cells in culture, and stabilized tubulin filaments in HUVEC monolayers. We conclude that the rapid shutdown of tumour microvasculature by CA4P is due in part to disruption of adherens junctions and that S1P has a protective effect on both adherens junctions and the endothelial cell cytoskeleton.

18.
Cancer Res ; 77(10): 2633-2646, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28377452

RESUMEN

Elevated plasma concentrations of soluble VEGFA isoforms are associated with poor prognosis in parallel with improved response to treatment with the anti-VEGFA antibody bevacizumab. To uncover the underlying mechanism to these observations, we administered anti-VEGFA therapy to mice bearing luminescent mouse fibrosarcomas expressing single VEGFA isoforms or their wild-type counterparts expressing all isoforms (fs120, fs164, fs188, or fsWT). Expression of the more soluble isoforms conferred an advantage for lung metastasis from subcutaneous tumors (fs120/164 vs. fs188/WT); fs120 cells also produced more lung colonies than fs188 cells when injected intravenously. Metastasis from subcutaneous fs120 tumors was more sensitive than fs188 to treatment with the anti-VEGFA antibody B20-4.1.1. Despite elevated plasma levels of VEGFA in fs120 tumor-bearing mice and a dependence on VEGF receptor 1 activity for metastasis to the lung, B20-4.1.1 did not affect survival in the lung on intravenous injection. B20-4.1.1 inhibited subcutaneous tumor growth and decreased vascular density in both fs120 and fs188 tumors. However, migration of fs120, but not fs188 cells, in vitro was inhibited by B20-4.1.1. The greater survival of fs120 cells in the lung was associated with VEGFR1-dependent accumulation of CD11b-positive myeloid cells and higher expression of the VEGFR1 ligand, PlGF2, by the fs120 cells in vitro and in the plasma and lungs of fs120 tumor-bearing mice. We conclude that soluble VEGFA isoform expression increases fibrosarcoma metastasis through multiple mechanisms that vary in their sensitivity to anti-VEGF/VEGFR inhibition, with VEGFA-targeted therapy suppressing metastasis through effects on the primary tumor rather than the metastatic site. Cancer Res; 77(10); 2633-46. ©2017 AACR.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Sarcoma/genética , Sarcoma/patología , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Biomarcadores , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Ratones Transgénicos , Metástasis de la Neoplasia , Isoformas de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Sarcoma/tratamiento farmacológico , Sarcoma/mortalidad , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
BMC Cancer ; 6: 280, 2006 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-17156434

RESUMEN

BACKGROUND: A functional vascular network is essential for the survival, growth and spread of solid tumours, making blood vessels a key target for therapeutic strategies. Combretastatin A-4 phosphate (CA-4-P) is a tubulin-depolymerising agent in Phase II clinical trials as a vascular disrupting agent. Not much is known of the molecular effect of CA-4-P under tumour conditions. The tumour microenvironment differs markedly from that in normal tissue, specifically with respect to oxygenation (hypoxia). Gene regulation under tumour conditions is governed by hypoxia inducible factor 1 (HIF-1), controlling angiogenic and metastatic pathways. METHODS: We investigated the effect of CA-4-P on factors of the upstream and downstream signalling pathway of HIF-1 in vitro. RESULTS: CA-4-P treatment under hypoxia tended to reduce HIF-1 accumulation in a concentration-dependent manner, an effect which was more prominent in endothelial cells than in cancer cell lines. Conversely, CA-4-P increased HIF-1 accumulation under aerobic conditions in vitro. At these concentrations of CA-4-P under aerobic conditions, nuclear factor kappaB was activated via the small GTPase RhoA, and expression of the HIF-1 downstream angiogenic effector gene, vascular endothelial growth factor (VEGF-A), was increased. CONCLUSION: Our findings advance the understanding of signal transduction pathways involved in the actions of the anti-vascular agent CA-4-P.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor 1 Inducible por Hipoxia/efectos de los fármacos , Factor 1 Inducible por Hipoxia/metabolismo , Estilbenos/farmacología , Carcinoma/patología , Neoplasias del Colon/patología , Relación Dosis-Respuesta a Droga , Humanos , Transducción de Señal , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/patología
20.
J Med Chem ; 59(20): 9473-9488, 2016 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-27690431

RESUMEN

The combretastatins are an important class of tubulin-binding agents. Of this family, a number of compounds are potent tumor vascular disrupting agents (VDAs) and have shown promise in the clinic for cancer therapy. We have developed a modular synthetic route to combretastatin analogs based on a pyrazole core through highly regioselective alkyne cycloaddition reactions of sydnones. These compounds show modest to high potency against human umbilical vein endothelial cell proliferation. Moreover, evidence is presented that these novel VDAs have the same mode of action as CA4P and bind reversibly to ß-tubulin, believed to be a key feature in avoiding toxicity. The most active compound from in vitro studies was taken forward to an in vivo model and instigated an increase in tumor cell necrosis.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos Fitogénicos/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Pirazoles/farmacología , Estilbenos/farmacología , Sidnonas/química , Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/química , Animales , Antineoplásicos Fitogénicos/síntesis química , Antineoplásicos Fitogénicos/química , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Ratones , Ratones SCID , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Pirazoles/química , Estilbenos/síntesis química , Estilbenos/química , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA