Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
PLoS One ; 16(10): e0252558, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34634047

RESUMEN

The identification of antibody variable regions in the heavy (VH) and light (VL) chains from hybridomas is necessary for the production of recombinant, sequence-defined monoclonal antibodies (mAbs) and antibody derivatives. This process has received renewed attention in light of recent reports of hybridomas having unintended specificities due to the production of non-antigen specific heavy and/or light chains for the intended antigen. Here we report a surprising finding and potential pitfall in variable domain sequencing of an anti-human CD63 hybridoma. We amplified multiple VL genes from the hybridoma cDNA, including the well-known aberrant Sp2/0 myeloma VK and a unique, full-length VL. After finding that the unique VL failed to yield a functional antibody, we discovered an additional full-length sequence with surprising similarity (~95% sequence identify) to the non-translated myeloma kappa chain but with a correction of its key frameshift mutation. Expression of the recombinant mAb confirmed that this highly homologous sequence is the antigen-specific light chain. Our results highlight the complexity of PCR-based cloning of antibody genes and strategies useful for identification of correct sequences.


Asunto(s)
Anticuerpos Monoclonales/genética , Hibridomas/fisiología , Cadenas Ligeras de Inmunoglobulina/genética , Mieloma Múltiple/genética , Secuencia de Aminoácidos , Animales , Células CHO , Línea Celular , Clonación Molecular/métodos , Cricetulus , ADN Complementario/genética , Mutación del Sistema de Lectura/genética , Genes de Inmunoglobulinas/genética , Células HEK293 , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Ratones , Proteínas Recombinantes/genética , Tetraspanina 30/genética
2.
Methods Mol Biol ; 2355: 163-173, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34386959

RESUMEN

Single-chain protein affinity ligands are recombinant polypeptides that recreate the antigen-binding site of parental, monoclonal antibodies (mAbs) or present unique binding surfaces derived from display technologies, computational design, or other approaches. These diverse ligands have several advantages over full-length mAbs as agents for delivery of small molecule, protein, and nanoparticle cargoes to desired sites in the body. However, they present unique challenges for modification and bioconjugation. Fusion of a LPXTGG motif, or "sortag," and a 5-amino acid, flexible linker to the C-terminus of these affinity ligands enables high-efficiency transpeptidation by the bacterial enzyme, Sortase A, and site-specific addition of fluorophores, radiolabels, or functional groups for oriented and stoichiometrically controlled bioconjugation. We describe in detail this method and address several challenges and pitfalls in the purification and characterization of modified single-chain affinity ligands.


Asunto(s)
Fluorescencia , Anticuerpos Monoclonales , Antígenos , Péptidos
3.
Proc Natl Acad Sci U S A ; 117(7): 3405-3414, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32005712

RESUMEN

Drug targeting to inflammatory brain pathologies such as stroke and traumatic brain injury remains an elusive goal. Using a mouse model of acute brain inflammation induced by local tumor necrosis factor alpha (TNFα), we found that uptake of intravenously injected antibody to vascular cell adhesion molecule 1 (anti-VCAM) in the inflamed brain is >10-fold greater than antibodies to transferrin receptor-1 and intercellular adhesion molecule 1 (TfR-1 and ICAM-1). Furthermore, uptake of anti-VCAM/liposomes exceeded that of anti-TfR and anti-ICAM counterparts by ∼27- and ∼8-fold, respectively, achieving brain/blood ratio >300-fold higher than that of immunoglobulin G/liposomes. Single-photon emission computed tomography imaging affirmed specific anti-VCAM/liposome targeting to inflamed brain in mice. Intravital microscopy via cranial window and flow cytometry showed that in the inflamed brain anti-VCAM/liposomes bind to endothelium, not to leukocytes. Anti-VCAM/LNP selectively accumulated in the inflamed brain, providing de novo expression of proteins encoded by cargo messenger RNA (mRNA). Anti-VCAM/LNP-mRNA mediated expression of thrombomodulin (a natural endothelial inhibitor of thrombosis, inflammation, and vascular leakage) and alleviated TNFα-induced brain edema. Thus VCAM-directed nanocarriers provide a platform for cerebrovascular targeting to inflamed brain, with the goal of normalizing the integrity of the blood-brain barrier, thus benefiting numerous brain pathologies.


Asunto(s)
Anticuerpos/administración & dosificación , Barrera Hematoencefálica/efectos de los fármacos , Encefalitis/tratamiento farmacológico , Endotelio Vascular/efectos de los fármacos , Nanomedicina/métodos , Animales , Barrera Hematoencefálica/inmunología , Encefalitis/genética , Encefalitis/inmunología , Endotelio Vascular/inmunología , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Ratones , Receptores de Transferrina/genética , Receptores de Transferrina/inmunología , Trombomodulina/genética , Trombomodulina/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/inmunología
4.
J Control Release ; 301: 54-61, 2019 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-30871995

RESUMEN

New advances in intra-arterial (IA) catheters offer clinically proven local interventions in the brain. Here we tested the effect of combining local IA delivery and vascular immunotargeting. Microinjection of tumor necrosis factor alpha (TNFα) in the brain parenchyma causes cerebral overexpression of Inter-Cellular Adhesion Molecule-1 (ICAM-1) in mice. Systemic intravenous injection of ICAM-1 antibody (anti-ICAM-1) and anti-ICAM-1/liposomes provided nearly an order of magnitude higher uptake in the inflamed vs normal brain (from ~0.1 to 0.8%ID/g for liposomes). Local injection of anti-ICAM-1 and anti-ICAM-1/liposomes via carotid artery catheter provided an additional respective 2-fold and 5-fold elevation of uptake in the inflamed brain vs levels attained by IV injection. The uptake in the inflamed brain of respective untargeted IgG counterparts was markedly lower (e.g., uptake of anti-ICAM-1/liposomes was 100-fold higher vs IgG/liposomes). These data affirm the specificity of the combined effect of the first pass and immunotargeting. Intravital real-time microscopy via cranial window revealed that anti-ICAM-1/liposomes, but not IgG/liposomes bind to the lumen of blood vessels in the inflamed brain within minutes after injection. This straightforward framework provides the basis for translational efforts towards local vascular drug targeting to the brain.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Encéfalo/metabolismo , Encefalitis/metabolismo , Molécula 1 de Adhesión Intercelular/inmunología , Animales , Anticuerpos Monoclonales/farmacocinética , Transporte Biológico , Encéfalo/irrigación sanguínea , Encefalitis/inducido químicamente , Liposomas , Pulmón/metabolismo , Masculino , Ratones Endogámicos C57BL , Nanoestructuras/administración & dosificación , Poliestirenos/administración & dosificación , Poliestirenos/farmacocinética , Factor de Necrosis Tumoral alfa
5.
Sci Rep ; 8(1): 1760, 2018 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-29379029

RESUMEN

Site-specific modification of antibodies has become a critical aspect in the development of next-generation immunoconjugates meeting criteria of clinically acceptable homogeneity, reproducibility, efficacy, ease of manufacturability, and cost-effectiveness. Using CRISPR/Cas9 genomic editing, we developed a simple and novel approach to produce site-specifically modified antibodies. A sortase tag was genetically incorporated into the C-terminal end of the third immunoglobulin heavy chain constant region (CH3) within a hybridoma cell line to manufacture antibodies capable of site-specific conjugation. This enabled an effective enzymatic site-controlled conjugation of fluorescent and radioactive cargoes to a genetically tagged mAb without impairment of antigen binding activity. After injection in mice, these immunoconjugates showed almost doubled specific targeting in the lung vs. chemically conjugated maternal mAb, and concomitant reduction in uptake in the liver and spleen. The approach outlined in this work provides a facile method for the development of more homogeneous, reproducible, effective, and scalable antibody conjugates for use as therapeutic and diagnostic tools.


Asunto(s)
Anticuerpos Monoclonales/genética , Sistemas CRISPR-Cas/genética , Inmunoconjugados/genética , Animales , Antígenos/genética , Línea Celular Tumoral , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Cisteína/genética , Humanos , Ratones , Ratas , Reproducibilidad de los Resultados
6.
Blood Adv ; 2(3): 165-176, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29365311

RESUMEN

Carriage of drugs by red blood cells (RBCs) modulates pharmacokinetics, pharmacodynamics, and immunogenicity. However, optimal targets for attaching therapeutics to human RBCs and adverse effects have not been studied. We engineered nonhuman-primate single-chain antibody fragments (scFvs) directed to human RBCs and fused scFvs with human thrombomodulin (hTM) as a representative biotherapeutic cargo (hTM-scFv). Binding fusions to RBCs on band 3/glycophorin A (GPA; Wright b [Wrb] epitope) and RhCE (Rh17/Hr0 epitope) similarly endowed RBCs with hTM activity, but differed in their effects on RBC physiology. scFv and hTM-scFv targeted to band 3/GPA increased membrane rigidity and sensitized RBCs to hemolysis induced by mechanical stress, while reducing sensitivity to hypo-osmotic hemolysis. Similar properties were seen for other ligands bound to GPA and band 3 on human and murine RBCs. In contrast, binding of scFv or hTM-scFv to RhCE did not alter deformability or sensitivity to mechanical and osmotic stress at similar copy numbers bound per RBCs. Contrasting responses were also seen for immunoglobulin G antibodies against band 3, GPA, and RhCE. RBC-bound hTM-scFv generated activated protein C (APC) in the presence of thrombin, but RhCE-targeted hTM-scFv demonstrated greater APC generation per bound copy. Both Wrb- and RhCE-targeted fusion proteins inhibited fibrin deposition induced by tumor necrosis factor-α in an endothelialized microfluidic model using human whole blood. RhCE-bound hTM-scFv more effectively reduced platelet and leukocyte adhesion, whereas anti-Wrb scFv appeared to promote platelet adhesion. These data provide a translational framework for the development of engineered affinity ligands to safely couple therapeutics to human RBCs.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Eritrocitos/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Trombosis/tratamiento farmacológico , Animales , Proteína 1 de Intercambio de Anión de Eritrocito/inmunología , Humanos , Inflamación/tratamiento farmacológico , Macaca , Ratones , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Sistema del Grupo Sanguíneo Rh-Hr/inmunología , Anticuerpos de Cadena Única/administración & dosificación , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Trombomodulina/administración & dosificación , Trombomodulina/genética , Trombosis/patología
7.
Bioconjug Chem ; 29(1): 56-66, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29200285

RESUMEN

The conjugation of antibodies to drugs and drug carriers improves delivery to target tissues. Widespread implementation and effective translation of this pharmacologic strategy awaits the development of affinity ligands capable of a defined degree of modification and highly efficient bioconjugation without loss of affinity. To date, such ligands are lacking for the targeting of therapeutics to vascular endothelial cells. To enable site-specific, click-chemistry conjugation to therapeutic cargo, we used the bacterial transpeptidase, sortase A, to attach short azidolysine containing peptides to three endothelial-specific single chain antibody fragments (scFv). While direct fusion of a recognition motif (sortag) to the scFv C-terminus generally resulted in low levels of sortase-mediated modification, improved reaction efficiency was observed for one protein, in which two amino acids had been introduced during cloning. This prompted insertion of a short, semi-rigid linker between scFv and sortag. The linker significantly enhanced modification of all three proteins, to the extent that unmodified scFv could no longer be detected. As proof of principle, purified, azide-modified scFv was conjugated to the antioxidant enzyme, catalase, resulting in robust endothelial targeting of functional cargo in vitro and in vivo.


Asunto(s)
Química Clic/métodos , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/farmacocinética , Secuencia de Aminoácidos , Aminoaciltransferasas/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Catalasa/metabolismo , Línea Celular , Cisteína Endopeptidasas/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/administración & dosificación , Molécula 1 de Adhesión Intercelular/química , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/administración & dosificación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/química , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/farmacocinética , Anticuerpos de Cadena Única/administración & dosificación , Anticuerpos de Cadena Única/metabolismo , Distribución Tisular
8.
PLoS One ; 12(1): e0169537, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28085903

RESUMEN

Monoclonal antibodies (mAbs) directed to extracellular epitopes of human and mouse Platelet Endothelial Cell Adhesion Molecule-1 (CD31 or PECAM-1) stimulate binding of other mAbs to distinct adjacent PECAM-1 epitopes. This effect, dubbed Collaborative Enhancement of Paired Affinity Ligands, or CEPAL, has been shown to enhance delivery of mAb-targeted drugs and nanoparticles to the vascular endothelium. Here we report new insights into the mechanism underlying this effect, which demonstrates equivalent amplitude in the following models: i) cells expressing a full length PECAM-1 and mutant form of PECAM-1 unable to form homodimers; ii) isolated fractions of cellular membranes; and, iii) immobilized recombinant PECAM-1. These results indicate that CEPAL is mediated not by interference in cellular functions or homophilic PECAM-1 interactions, but rather by conformational changes within the cell adhesion molecule induced by ligand binding. This mechanism, mediated by exposure of partially occult epitopes, is likely to occur in molecules other than PECAM-1 and may represent a generalizable phenomenon with valuable practical applications.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Adhesión Celular/inmunología , Membrana Celular/inmunología , Endotelio Vascular/metabolismo , Epítopos/inmunología , Neoplasias Pulmonares/metabolismo , Mesotelioma/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Animales , Anticuerpos Monoclonales/metabolismo , Células Cultivadas , Endotelio Vascular/inmunología , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Mesotelioma/inmunología , Mesotelioma/patología , Mesotelioma Maligno , Ratones , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo
9.
Blood Adv ; 1(18): 1452-1465, 2017 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-29296786

RESUMEN

Diverse human illnesses are characterized by loss or inactivation of endothelial thrombomodulin (TM), predisposing to microvascular inflammation, activation of coagulation, and tissue ischemia. Single-chain antibody fragment (scFv)/TM) fusion proteins, previously protective against end-organ injury in murine models of inflammation, are attractive candidates to treat inflammatory thrombosis. However, animal models have inherent differences in TM and coagulation biology, are limited in their ability to resolve and control endothelial biology, and do not allow in-depth testing of "humanized" scFv/TM fusion proteins, which are necessary for translation to the clinical domain. To address these challenges, we developed a human whole-blood, microfluidic model of inflammatory, tissue factor (TF)-driven coagulation that features a multichannel format for head-to-head comparison of therapeutic approaches. In this model, fibrin deposition, leukocyte adhesion, and platelet adhesion and aggregation showed a dose-dependent response to tumor necrosis factor-α activation and could be quantified via real-time microscopy. We used this model to compare hTM/R6.5, a humanized, intracellular adhesion molecule 1 (ICAM-1)-targeted scFv/TM biotherapeutic, to untargeted antithrombotic agents, including soluble human TM (shTM), anti-TF antibodies, and hirudin. The targeted hTM/R6.5 more effectively inhibited TF-driven coagulation in a protein C (PC)-dependent manner and demonstrated synergy with supplemental PC. These results support the translational prospects of ICAM-targeted scFv/TM and illustrate the utility of the microfluidic system as a platform to study humanized therapeutics at the interface of endothelium and whole blood under flow.

10.
Artículo en Inglés | MEDLINE | ID: mdl-25946066

RESUMEN

Targeted drug delivery aims to improve therapeutic effects and enable mechanisms that are not feasible for untargeted agents (e.g., due to impermeable biological barriers). To achieve targeting, a drug or its carrier should possess properties providing specific accumulation from circulation at the desired site. There are several examples of systems-inspired approaches that have been applied to achieve this goal. First, proteomics analysis of plasma membrane fraction of the vascular endothelium has identified a series of target molecules and their ligands (e.g., antibodies) that deliver conjugated cargoes to well-defined vascular cells and subcellular compartments. Second, selection of ligands binding to cells of interest using phage display libraries in vitro and in vivo has provided peptides and polypeptides that bind to normal and pathologically altered cells. Finally, large-scale high-throughput combinatorial synthesis and selection of lipid- and polymer-based nanocarriers varying their chemical components has yielded a series of carriers accumulating in diverse organs and delivering RNA interference agents to diverse cells. Together, these approaches offer a basis for systems-based design and selection of targets, targeting molecules, and targeting vehicles. Current studies focus on expanding the arsenal of these and alternative targeting strategies, devising drug delivery systems capitalizing on these strategies and evaluation of their benefit/risk ratio in adequate animal models of human diseases. These efforts, combined with better understanding of mechanisms and unintended consequences of these targeted interventions, need to be ultimately translated into industrial development and the clinical domain.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Diseño de Fármacos , Nanopartículas , Biología de Sistemas/métodos , Animales , Sistemas de Liberación de Medicamentos/instrumentación , Humanos , Proteómica/métodos , Interferencia de ARN
11.
FASEB J ; 29(8): 3483-92, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25953848

RESUMEN

Anchoring pharmacologic agents to the vascular lumen has the potential to modulate critical processes at the blood-tissue interface, avoiding many of the off-target effects of systemically circulating agents. We report a novel strategy for endothelial dual targeting of therapeutics, which both enhances drug delivery and enables targeted agents to partner enzymatically to generate enhanced biologic effect. Based on the recent discovery that paired antibodies directed to adjacent epitopes of platelet endothelial cell adhesion molecule (PECAM)-1 stimulate each other's binding, we fused single-chain fragments (scFv) of paired anti-mouse PECAM-1 antibodies to recombinant murine thrombomodulin (TM) and endothelial protein C receptor (EPCR), endothelial membrane proteins that partner in activation of protein C (PC). scFv/TM and scFv/EPCR bound to mouse endothelial PECAM-1 with high affinity (EC50 1.5 and 3.8 nM, respectively), and codelivery induced a 5-fold increase in PC activation not seen when TM and EPCR are anchored to distinct cell adhesion molecules. In a mouse model of acute lung injury, dual targeting reduces both the expression of lung inflammatory markers and trans-endothelial protein leak by as much as 40%, as compared to either agent alone. These findings provide proof of principle for endothelial dual targeting, an approach with numerous potential biomedical applications.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Preparaciones Farmacéuticas/administración & dosificación , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos/métodos , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Epítopos/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteína C/metabolismo , Receptores de Superficie Celular/metabolismo , Trombomodulina/metabolismo
12.
J Control Release ; 163(2): 161-9, 2012 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-22974832

RESUMEN

Oxidant stress caused by pathological elevation of reactive oxygen species (ROS) production in the endothelial cells lining the vascular lumen is an important component of many vascular and pulmonary disease conditions. NADPH oxidase (NOX) activated by pathological mediators including angiotensin and cytokines is a major source of endothelial ROS. In order to intercept this pathological pathway, we have encapsulated an indirect NOX inhibitor, MJ33, into immunoliposomes (Ab-MJ33/IL) targeted to endothelial marker platelet endothelial cell adhesion molecule (PECAM-1). Ab-MJ33/IL, but not control IgG-MJ33/IL are specifically bound to endothelium and attenuated angiotensin-induced ROS production in vitro and in vivo. Additionally, Ab-MJ33/IL inhibited endothelial expression of the inflammatory marker vascular cell adhesion molecule (VCAM) in cells and animals challenged with the cytokine TNF. Furthermore, Ab-MJ33/IL alleviated pathological disruption of endothelial permeability barrier function in cells exposed to vascular endothelial growth factor (VEGF) and in the lungs of mice challenged with lipopolysaccharide (LPS). Of note, the latter beneficial effect has been achieved both by prophylactic and therapeutic injection of Ab-MJ33/IL in animals. Therefore, specific suppression of ROS production by NOX in endothelium, attainable by Ab-MJ33/IL targeting, may help deciphering mechanisms of vascular oxidative stress and inflammation, and potentially improve treatment of these conditions.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antioxidantes/administración & dosificación , Glicerofosfatos/administración & dosificación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Angiotensina II/farmacología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Inhibidores Enzimáticos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/metabolismo , Lipopolisacáridos , Liposomas , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas/antagonistas & inhibidores , Fosfolipasas A/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Estreptavidina/química , Distribución Tisular , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología
13.
Curr Hypertens Rep ; 13(6): 436-41, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21826469

RESUMEN

Sunitinib is a highly potent, multitargeted anticancer agent. However, there is growing clinical evidence that sunitinib induces cardiac dysfunction. Disruption of multiple signaling pathways, which are important in the maintenance of adult cardiac function, is likely to result in cardiovascular toxicity. Basic and translational evidence implicates a potential role for specific growth factor signaling pathways. This review discusses the relevant translational data from animal models of heart failure, focusing on three key pathways that are inhibited by sunitinib: AMP-activated protein kinase (AMPK), platelet-derived growth factor receptors (PDGFRs), and the vascular endothelial growth factor receptors (VEGFRs) 1, 2, and 3. We hypothesize that disruption of these pathways by sunitinib results in cardiotoxicity, and present direct and indirect evidence to support the notion that sunitinib-induced cardiac dysfunction likely involves a variety of molecular mechanisms that are critical for cardiac homeostasis.


Asunto(s)
Inhibidores de la Angiogénesis/efectos adversos , Antineoplásicos/efectos adversos , Cardiotoxinas , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/inducido químicamente , Indoles/efectos adversos , Pirroles/efectos adversos , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Animales , Carcinoma de Células Renales/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Humanos , Neoplasias Renales/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Receptores de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Factores de Riesgo , Transducción de Señal/efectos de los fármacos , Sunitinib
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA