Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nat Chem Biol ; 19(2): 159-167, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36253549

RESUMEN

The human gut bacterial genotoxin colibactin is a possible key driver of colorectal cancer (CRC) development. Understanding colibactin's biological effects remains difficult owing to the instability of the proposed active species and the complexity of the gut microbiota. Here, we report small molecule boronic acid inhibitors of colibactin biosynthesis. Designed to mimic the biosynthetic precursor precolibactin, these compounds potently inhibit the colibactin-activating peptidase ClbP. Using biochemical assays and crystallography, we show that they engage the ClbP binding pocket, forming a covalent bond with the catalytic serine. These inhibitors reproduce the phenotypes observed in a clbP deletion mutant and block the genotoxic effects of colibactin on eukaryotic cells. The availability of ClbP inhibitors will allow precise, temporal control over colibactin production, enabling further study of its contributions to CRC. Finally, application of our inhibitors to related peptidase-encoding pathways highlights the power of chemical tools to probe natural product biosynthesis.


Asunto(s)
Microbioma Gastrointestinal , Policétidos , Humanos , Mutágenos/metabolismo , Mutágenos/toxicidad , Escherichia coli/metabolismo , Policétidos/química , Péptido Hidrolasas/química
2.
Cell ; 141(1): 166-77, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20371352

RESUMEN

It has been recently reported that treatment with an anti-placenta growth factor (PlGF) antibody inhibits metastasis and primary tumor growth. Here we show that, although anti-PlGF treatment inhibited wound healing, extravasation of B16F10 cells, and growth of a tumor engineered to overexpress the PlGF receptor (VEGFR-1), neutralization of PlGF using four novel blocking antibodies had no significant effect on tumor angiogenesis in 15 models. Also, genetic ablation of the tyrosine kinase domain of VEGFR-1 in the host did not result in growth inhibition of the anti-VEGF-A sensitive or resistant tumors tested. Furthermore, combination of anti-PlGF with anti-VEGF-A antibodies did not result in greater antitumor efficacy than anti-VEGF-A monotherapy. In conclusion, our data argue against an important role of PlGF during primary tumor growth in most models and suggest that clinical evaluation of anti-PlGF antibodies may be challenging.


Asunto(s)
Neoplasias/irrigación sanguínea , Neovascularización Patológica , Proteínas Gestacionales/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos BALB C , Factor de Crecimiento Placentario , Proteínas Gestacionales/antagonistas & inhibidores , Factores de Crecimiento Endotelial Vascular
3.
EMBO J ; 31(17): 3513-23, 2012 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-22773185

RESUMEN

Angiogenesis plays a crucial role during tumorigenesis and much progress has been recently made in elucidating the role of VEGF and other growth factors in the regulation of angiogenesis. Recently, microRNAs (miRNAs) have been shown to modulate a variety of physiogical and pathological processes. We identified a set of differentially expressed miRNAs in microvascular endothelial cells co-cultured with tumour cells. Unexpectedly, most miRNAs were derived from tumour cells, packaged into microvesicles (MVs), and then directly delivered to endothelial cells. Among these miRNAs, we focused on miR-9 due to the strong morphological changes induced in cultured endothelial cells. We found that exogenous miR-9 effectively reduced SOCS5 levels, leading to activated JAK-STAT pathway. This signalling cascade promoted endothelial cell migration and tumour angiogenesis. Remarkably, administration of anti-miR-9 or JAK inhibitors suppressed MV-induced cell migration in vitro and decreased tumour burden in vivo. Collectively, these observations suggest that tumour-secreted miRNAs participate in intercellular communication and function as a novel pro-angiogenic mechanism.


Asunto(s)
Células Endoteliales/fisiología , MicroARNs/biosíntesis , Neoplasias/genética , Neovascularización Patológica/genética , Animales , Células COS , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Chlorocebus aethiops , Humanos , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/metabolismo , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/metabolismo , Ratones , Ratones Endogámicos BALB C , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Proc Natl Acad Sci U S A ; 108(28): 11590-5, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21709213

RESUMEN

PlGF, one of the ligands for VEGFR-1, has been implicated in tumor angiogenesis. However, more recent studies indicate that genetic or pharmacological inhibition of PlGF signaling does not result in reduction of microvascular density in a variety of tumor models. Here we screened 12 human tumor cell lines and identified 3 that are growth inhibited by anti-PlGF antibodies in vivo. We found that efficacy of anti-PlGF treatment strongly correlates with VEGFR-1 expression in tumor cells, but not with antiangiogenesis. In addition, PlGF induced VEGFR-1 signaling and biological responses in tumor cell lines sensitive to anti-PlGF, but not in refractory tumor cell lines or in endothelial cells. Also, genetic ablation of VEGFR-1 signaling in the host did not affect the efficacy of PlGF blockade. Collectively, these findings suggest that the role of PlGF in tumorigenesis largely consists of promoting autocrine/paracrine growth of tumor cells expressing a functional VEGFR-1 rather than stimulation of angiogenesis.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Neoplasias/inmunología , Neoplasias/terapia , Proteínas Gestacionales/antagonistas & inhibidores , Proteínas Gestacionales/inmunología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Tumoral , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , Neoplasias/irrigación sanguínea , Neoplasias/etiología , Neovascularización Patológica , Factor de Crecimiento Placentario , Proteínas Gestacionales/farmacología , ARN Interferente Pequeño/genética , Transducción de Señal , Células del Estroma/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
5.
Nature ; 450(7171): 825-31, 2007 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-18064003

RESUMEN

Bone-marrow-derived cells facilitate tumour angiogenesis, but the molecular mechanisms of this facilitation are incompletely understood. We have previously shown that the related EG-VEGF and Bv8 proteins, also known as prokineticin 1 (Prok1) and prokineticin 2 (Prok2), promote both tissue-specific angiogenesis and haematopoietic cell mobilization. Unlike EG-VEGF, Bv8 is expressed in the bone marrow. Here we show that implantation of tumour cells in mice resulted in upregulation of Bv8 in CD11b+Gr1+ myeloid cells. We identified granulocyte colony-stimulating factor as a major positive regulator of Bv8 expression. Anti-Bv8 antibodies reduced CD11b+Gr1+ cell mobilization elicited by granulocyte colony-stimulating factor. Adenoviral delivery of Bv8 into tumours was shown to promote angiogenesis. Anti-Bv8 antibodies inhibited growth of several tumours in mice and suppressed angiogenesis. Anti-Bv8 treatment also reduced CD11b+Gr1+ cells, both in peripheral blood and in tumours. The effects of anti-Bv8 antibodies were additive to those of anti-Vegf antibodies or cytotoxic chemotherapy. Thus, Bv8 modulates mobilization of CD11b+Gr1+ cells from the bone marrow during tumour development and also promotes angiogenesis locally.


Asunto(s)
Hormonas Gastrointestinales/metabolismo , Células Mieloides/metabolismo , Neoplasias/irrigación sanguínea , Neovascularización Patológica , Neuropéptidos/metabolismo , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Antineoplásicos/farmacología , División Celular/efectos de los fármacos , Línea Celular Tumoral , Hormonas Gastrointestinales/antagonistas & inhibidores , Hormonas Gastrointestinales/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Ratones , Ratones Desnudos , Células Mieloides/efectos de los fármacos , Trasplante de Neoplasias , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico , Neuropéptidos/antagonistas & inhibidores , Neuropéptidos/inmunología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
6.
Toxicol In Vitro ; 84: 105434, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35820568

RESUMEN

In vitro testing of Electronic Nicotine Delivery System (ENDS) aerosol condensates is important in evaluating their potential toxicity. Collecting sufficient condensate for these tests is a time consuming and costly procedure. The "triple puff (TP)" is a novel system which collects the aerosol from three ENDS devices sequentially into a single filter pad and impinger. The TP substantially reduces condensate collection time relative to the conventional single ENDS, single puff (SP), device system. Both the TP and SP (using two puffing profiles) were used to generate condensates from JUUL ENDS e-liquid Mint 5.0% (nicotine by weight). Aerosols were collected using the filter pad and ethanol-containing impinger method. Condensates produced with the SP and TP were compared for concentrations of primary constituents and carbonyl compounds as well as for their cytotoxicity (OECD 129), mutagenicity (OECD 471) and genotoxicity (OECD 487). Condensates generated with the SP and TP, regardless of puffing regimen, were very similar chemically and equivalent in the biological assays tested (not cytotoxic, mutagenic, or genotoxic). The TP device significantly reduces production time of ENDS condensates relative to the standard SP method and thus may facilitate further research by reducing the time and effort required to collect ENDS condensates.


Asunto(s)
Sistemas Electrónicos de Liberación de Nicotina , Aerosoles/química , Mutágenos , Nicotina/toxicidad
7.
Nat Commun ; 12(1): 1913, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33772014

RESUMEN

Diffusion is a major molecular transport mechanism in biological systems. Quantifying direction-dependent (i.e., anisotropic) diffusion is vitally important to depicting how the three-dimensional (3D) tissue structure and composition affect the biochemical environment, and thus define tissue functions. However, a tool for noninvasively measuring the 3D anisotropic extracellular diffusion of biorelevant molecules is not yet available. Here, we present light-sheet imaging-based Fourier transform fluorescence recovery after photobleaching (LiFT-FRAP), which noninvasively determines 3D diffusion tensors of various biomolecules with diffusivities up to 51 µm2 s-1, reaching the physiological diffusivity range in most biological systems. Using cornea as an example, LiFT-FRAP reveals fundamental limitations of current invasive two-dimensional diffusion measurements, which have drawn controversial conclusions on extracellular diffusion in healthy and clinically treated tissues. Moreover, LiFT-FRAP demonstrates that tissue structural or compositional changes caused by diseases or scaffold fabrication yield direction-dependent diffusion changes. These results demonstrate LiFT-FRAP as a powerful platform technology for studying disease mechanisms, advancing clinical outcomes, and improving tissue engineering.


Asunto(s)
Córnea/metabolismo , Espacio Extracelular/metabolismo , Recuperación de Fluorescencia tras Fotoblanqueo/métodos , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Tendones/metabolismo , Animales , Anisotropía , Colágeno/química , Colágeno/metabolismo , Difusión , Análisis de Fourier , Microscopía Confocal/métodos , Microscopía Electrónica de Rastreo/métodos , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Porcinos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química
8.
Nat Biomed Eng ; 4(4): 446-462, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32284552

RESUMEN

Environmental factors are the largest contributors to cardiovascular disease. Here we show that cardiac organoids that incorporate an oxygen-diffusion gradient and that are stimulated with the neurotransmitter noradrenaline model the structure of the human heart after myocardial infarction (by mimicking the infarcted, border and remote zones), and recapitulate hallmarks of myocardial infarction (in particular, pathological metabolic shifts, fibrosis and calcium handling) at the transcriptomic, structural and functional levels. We also show that the organoids can model hypoxia-enhanced doxorubicin cardiotoxicity. Human organoids that model diseases with non-genetic pathological factors could help with drug screening and development.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Corazón/efectos de los fármacos , Modelos Cardiovasculares , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Organoides/efectos de los fármacos , Cardiotoxicidad/metabolismo , Cardiotoxicidad/patología , Desarrollo de Medicamentos , Humanos , Infarto del Miocardio/inducido químicamente , Infarto del Miocardio/genética , Organoides/metabolismo , Organoides/patología , Oxígeno/metabolismo
9.
Tissue Eng Part A ; 25(7-8): 620-631, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30226442

RESUMEN

IMPACT STATEMENT: Human adipose-derived stem cells (hADSCs) spheroids have displayed remarkable potential for treating ischemic injury. However, low nutrient (i.e., glucose and oxygen) availability in ischemic environments results in limited tissue viability posttransplantation. To develop an understanding of the effects of nutrient availability on spheroid survival, we utilized both in vitro and computational models to examine the limiting factors in metabolic supply for avascular microtissues, revealing the critical role of glucose to improve hADSC spheroid survival in ischemic conditions. These results may impact future strategies for improving hADSC transplantation efficacy through codelivery of metabolic substrates.


Asunto(s)
Tejido Adiposo/citología , Esferoides Celulares/citología , Células Madre/citología , Diferenciación Celular/fisiología , Células Cultivadas , Humanos , Isquemia/metabolismo , Modelos Teóricos
10.
Acta Biomater ; 51: 495-504, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28087483

RESUMEN

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) provide an unlimited cell source to treat cardiovascular diseases, the leading cause of death worldwide. However, current hiPSC-CMs retain an immature phenotype that leads to difficulties for integration with adult myocardium after transplantation. To address this, we recently utilized electrically conductive silicon nanowires (e-SiNWs) to facilitate self-assembly of hiPSC-CMs to form nanowired hiPSC cardiac spheroids. Our previous results showed addition of e-SiNWs effectively enhanced the functions of the cardiac spheroids and improved the cellular maturation of hiPSC-CMs. Here, we examined two important factors that can affect functions of the nanowired hiPSC cardiac spheroids: (1) cell number per spheroid (i.e., size of the spheroids), and (2) the electrical conductivity of the e-SiNWs. To examine the first factor, we prepared hiPSC cardiac spheroids with four different sizes by varying cell number per spheroid (∼0.5k, ∼1k, ∼3k, ∼7k cells/spheroid). Spheroids with ∼3k cells/spheroid was found to maximize the beneficial effects of the 3D spheroid microenvironment. This result was explained with a semi-quantitative theory that considers two competing factors: 1) the improved 3D cell-cell adhesion, and 2) the reduced oxygen supply to the center of spheroids with the increase of cell number. Also, the critical role of electrical conductivity of silicon nanowires has been confirmed in improving tissue function of hiPSC cardiac spheroids. These results lay down a solid foundation to develop suitable nanowired hiPSC cardiac spheroids as an innovative cell delivery system to treat cardiovascular diseases. STATEMENT OF SIGNIFICANCE: Cardiovascular disease is the leading cause of death and disability worldwide. Due to the limited regenerative capacity of adult human hearts, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have received significant attention because they provide a patient specific cell source to regenerate damaged hearts. Despite the progress, current human hiPSC-CMs retain an immature phenotype that leads to difficulties for integration with adult myocardium after transplantation. To address this, we recently utilized electrically conductive silicon nanowires (e-SiNWs) to facilitate self-assembly of hiPSC-CMs to form nanowired hiPSC cardiac spheroids. Our previous results showed addition of e-SiNWs effectively enhanced the functions of the cardiac spheroids and improved the cellular maturation of hiPSC-CMs. In this manuscript, we examined the effects of two important factors on the functions of nanowired hiPSC cardiac spheroids: (1) cell number per spheroid (i.e., size of the spheroids), and (2) the electrical conductivity of the e-SiNWs. The results from these studies will allow for the development of suitable nanowired hiPSC cardiac spheroids to effectively deliver hiPSC-CMs for heart repair.


Asunto(s)
Conductividad Eléctrica , Miocitos Cardíacos/citología , Nanocables/química , Silicio/química , Esferoides Celulares/citología , Recuento de Células , Tamaño de la Célula , Técnica del Anticuerpo Fluorescente , Humanos , Células Madre Pluripotentes Inducidas/citología , Nanocables/ultraestructura , Esferoides Celulares/metabolismo , Esferoides Celulares/ultraestructura
11.
Sci Signal ; 6(271): ra25, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23592840

RESUMEN

The vascular endothelial growth factor (VEGF) signaling pathway plays a pivotal role in normal development and also represents a major therapeutic target for tumors and intraocular neovascular disorders. The VEGF receptor tyrosine kinases promote angiogenesis by phosphorylating downstream proteins in endothelial cells. We applied a large-scale proteomic approach to define the VEGF-regulated phosphoproteome and its temporal dynamics in human umbilical vein endothelial cells and then used siRNA (small interfering RNA) screens to investigate the function of a subset of these phosphorylated proteins in VEGF responses. The PI3K (phosphatidylinositol 3-kinase)-mTORC2 (mammalian target of rapamycin complex 2) axis emerged as central in activating VEGF-regulated phosphorylation and increasing endothelial cell viability by suppressing the activity of the transcription factor FoxO1 (forkhead box protein O1), an effect that limited cellular apoptosis and feedback activation of receptor tyrosine kinases. This FoxO1-mediated feedback loop not only reduced the effectiveness of mTOR inhibitors at decreasing protein phosphorylation and cell survival but also rendered cells more susceptible to PI3K inhibition. Collectively, our study provides a global and dynamic view of VEGF-regulated phosphorylation events and implicates the mTORC2-FoxO1 axis in VEGF receptor signaling and reprogramming of receptor tyrosine kinases in human endothelial cells.


Asunto(s)
Activación Enzimática/fisiología , Factores de Transcripción Forkhead/metabolismo , Complejos Multiproteicos/metabolismo , Fosfopéptidos/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Apoptosis/fisiología , Proteína Forkhead Box O1 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Diana Mecanicista del Complejo 2 de la Rapamicina , Fosforilación , Proteómica
12.
Clin Cancer Res ; 19(16): 4433-45, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23812669

RESUMEN

PURPOSE: Our goal was to develop a potent humanized antibody against mouse/human CXCL12. This report summarized its in vitro and in vivo activities. EXPERIMENTAL DESIGN: Cell surface binding and cell migration assays were used to select neutralizing hamster antibodies, followed by testing in several animal models. Monoclonal antibody (mAb) 30D8 was selected for humanization based on its in vitro and in vivo activities. RESULTS: 30D8, a hamster antibody against mouse and human CXCL12α, CXCL12ß, and CXCL12γ, was shown to dose-dependently block CXCL12α binding to CXCR4 and CXCR7, and CXCL12α-induced Jurkat cell migration in vitro. Inhibition of primary tumor growth and/or metastasis was observed in several models. 30D8 alone significantly ameliorated arthritis in a mouse collagen-induced arthritis model (CIA). Combination with a TNF-α antagonist was additive. In addition, 30D8 inhibited 50% of laser-induced choroidal neovascularization (CNV) in mice. Humanized 30D8 (hu30D8) showed similar in vitro and in vivo activities as the parental hamster antibody. A crystal structure of the hu30D8 Fab/CXCL12α complex in combination with mutational analysis revealed a "hot spot" around residues Asn(44)/Asn(45) of CXCL12α and part of the RFFESH region required for CXCL12α binding to CXCR4 and CXCR7. Finally, hu30D8 exhibited fast clearance in cynomolgus monkeys but not in rats. CONCLUSION: CXCL12 is an attractive target for treatment of cancer and inflammation-related diseases; hu30D8 is suitable for testing this hypothesis in humans.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Quimiocina CXCL12/antagonistas & inhibidores , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/química , Antiinflamatorios/farmacología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/química , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/metabolismo , Línea Celular Tumoral , Quimiocina CXCL12/química , Quimiocina CXCL12/metabolismo , Cricetinae , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Mapeo Epitopo , Femenino , Humanos , Ratones , Modelos Moleculares , Metástasis de la Neoplasia , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Conformación Proteica , Carga Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Clin Cancer Res ; 19(13): 3681-92, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23685835

RESUMEN

PURPOSE: The aim of this study was to identify conserved pharmacodynamic and potential predictive biomarkers of response to anti-VEGF therapy using gene expression profiling in preclinical tumor models and in patients. EXPERIMENTAL DESIGN: Surrogate markers of VEGF inhibition [VEGF-dependent genes or VEGF-dependent vasculature (VDV)] were identified by profiling gene expression changes induced in response to VEGF blockade in preclinical tumor models and in human biopsies from patients treated with anti-VEGF monoclonal antibodies. The potential value of VDV genes as candidate predictive biomarkers was tested by correlating high or low VDV gene expression levels in pretreatment clinical samples with the subsequent clinical efficacy of bevacizumab (anti-VEGF)-containing therapy. RESULTS: We show that VDV genes, including direct and more distal VEGF downstream endothelial targets, enable detection of VEGF signaling inhibition in mouse tumor models and human tumor biopsies. Retrospective analyses of clinical trial data indicate that patients with higher VDV expression in pretreatment tumor samples exhibited improved clinical outcome when treated with bevacizumab-containing therapies. CONCLUSIONS: In this work, we identified surrogate markers (VDV genes) for in vivo VEGF signaling in tumors and showed clinical data supporting a correlation between pretreatment VEGF bioactivity and the subsequent efficacy of anti-VEGF therapy. We propose that VDV genes are candidate biomarkers with the potential to aid the selection of novel indications as well as patients likely to respond to anti-VEGF therapy. The data presented here define a diagnostic biomarker hypothesis based on translational research that warrants further evaluation in additional retrospective and prospective trials.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/farmacología , Bevacizumab , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Neoplasias/genética , Neoplasias/mortalidad , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA