Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(19): e2322934121, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38701119

RESUMO

EPH receptors (EPHs), the largest family of tyrosine kinases, phosphorylate downstream substrates upon binding of ephrin cell surface-associated ligands. In a large cohort of endometriotic lesions from individuals with endometriosis, we found that EPHA2 and EPHA4 expressions are increased in endometriotic lesions relative to normal eutopic endometrium. Because signaling through EPHs is associated with increased cell migration and invasion, we hypothesized that chemical inhibition of EPHA2/4 could have therapeutic value. We screened DNA-encoded chemical libraries (DECL) to rapidly identify EPHA2/4 kinase inhibitors. Hit compound, CDD-2693, exhibited picomolar/nanomolar kinase activity against EPHA2 (Ki: 4.0 nM) and EPHA4 (Ki: 0.81 nM). Kinome profiling revealed that CDD-2693 bound to most EPH family and SRC family kinases. Using NanoBRET target engagement assays, CDD-2693 had nanomolar activity versus EPHA2 (IC50: 461 nM) and EPHA4 (IC50: 40 nM) but was a micromolar inhibitor of SRC, YES, and FGR. Chemical optimization produced CDD-3167, having picomolar biochemical activity toward EPHA2 (Ki: 0.13 nM) and EPHA4 (Ki: 0.38 nM) with excellent cell-based potency EPHA2 (IC50: 8.0 nM) and EPHA4 (IC50: 2.3 nM). Moreover, CDD-3167 maintained superior off-target cellular selectivity. In 12Z endometriotic epithelial cells, CDD-2693 and CDD-3167 significantly decreased EFNA5 (ligand) induced phosphorylation of EPHA2/4, decreased 12Z cell viability, and decreased IL-1ß-mediated expression of prostaglandin synthase 2 (PTGS2). CDD-2693 and CDD-3167 decreased expansion of primary endometrial epithelial organoids from patients with endometriosis and decreased Ewing's sarcoma viability. Thus, using DECL, we identified potent pan-EPH inhibitors that show specificity and activity in cellular models of endometriosis and cancer.


Assuntos
Inibidores de Proteínas Quinases , Humanos , Feminino , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/química , Endometriose/tratamento farmacológico , Endometriose/metabolismo , Endometriose/patologia , DNA/metabolismo , Receptores da Família Eph/metabolismo , Receptores da Família Eph/antagonistas & inibidores , Receptor EphA2/metabolismo , Receptor EphA2/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Bibliotecas de Moléculas Pequenas/química , Movimento Celular/efeitos dos fármacos
2.
Mol Cell ; 79(3): 390-405.e7, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32619402

RESUMO

Despite their apparent lack of catalytic activity, pseudokinases are essential signaling molecules. Here, we describe the structural and dynamic properties of pseudokinase domains from the Wnt-binding receptor tyrosine kinases (PTK7, ROR1, ROR2, and RYK), which play important roles in development. We determined structures of all pseudokinase domains in this family and found that they share a conserved inactive conformation in their activation loop that resembles the autoinhibited insulin receptor kinase (IRK). They also have inaccessible ATP-binding pockets, occluded by aromatic residues that mimic a cofactor-bound state. Structural comparisons revealed significant domain plasticity and alternative interactions that substitute for absent conserved motifs. The pseudokinases also showed dynamic properties that were strikingly similar to those of IRK. Despite the inaccessible ATP site, screening identified ATP-competitive type-II inhibitors for ROR1. Our results set the stage for an emerging therapeutic modality of "conformational disruptors" to inhibit or modulate non-catalytic functions of pseudokinases deregulated in disease.


Assuntos
Moléculas de Adesão Celular/química , Inibidores de Proteínas Quinases/farmacologia , Receptores Proteína Tirosina Quinases/química , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/química , Sequência de Aminoácidos , Animais , Baculoviridae/genética , Baculoviridae/metabolismo , Sítios de Ligação , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Clonagem Molecular , Cristalografia por Raios X , Expressão Gênica , Humanos , Camundongos , Modelos Moleculares , Células Precursoras de Linfócitos B/citologia , Células Precursoras de Linfócitos B/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Inibidores de Proteínas Quinases/química , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/antagonistas & inibidores , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Receptores da Família Eph/antagonistas & inibidores , Receptores da Família Eph/química , Receptores da Família Eph/genética , Receptores da Família Eph/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Sf9 , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Spodoptera , Homologia Estrutural de Proteína , Especificidade por Substrato
3.
J Mol Neurosci ; 70(12): 2001-2006, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32488844

RESUMO

The search for novel neuroprotection strategies in ischemic stroke continues, as revascularization using tissue-plasminogen activator is the only pharmacological method currently available to patients. The purpose of this review article is to summarize research findings regarding the erythropoietin-producing hepatocellular receptor pathway as an emerging novel molecular target for neuroprotection in ischemic stroke. Ephrin-Eph interactions represent a new strategy in neuroprotection. Potential therapeutic targets include the different cellular locations within the neurovascular unit (e.g. astrocytes and neurons) and the different ephrin receptor subtypes. In particular, ephrin-B2/EphB4 receptor stimulation seems to exert neuroprotective effects, while stimulation of other ligands/receptors results in deleterious effects, during the post-ischemic stroke recovery phase. Neuroprotection, assessed by either a decrease in neurovascular unit injury markers or improvement in motor function tests, can be achieved by modulating the activity of different ephrin-Eph receptor subtypes. These novel molecular targets provide multiple potential neuroprotective therapeutic benefits, with meaningful clinical outcomes.


Assuntos
Efrinas/metabolismo , AVC Isquêmico/metabolismo , Receptores da Família Eph/metabolismo , Animais , Humanos , AVC Isquêmico/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Receptores da Família Eph/antagonistas & inibidores , Transdução de Sinais
4.
Pharmacol Res ; 159: 105038, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32565311

RESUMO

Ephrin-Eph signaling is a receptor tyrosine kinase signaling pathway involved in a variety of cellular mechanisms, of which many are related to the adhesion or migration of cells. Both the Eph receptor and ephrin ligand are abundantly present on a wide variety of cell types, and strongly evolutionary conserved. This review provides an overview of how 18 genetically diverse viruses utilize the Eph receptor (Eph), ephrin ligand (ephrin) or ephrin-Eph signaling to their advantage in their viral life cycle. Both Ephs and ephrins have been shown to serve as entry receptors for a variety of viruses, via both membrane fusion and endocytosis. Ephs and ephrins are also involved in viral transmission by vectors, associated with viral replication or persistence and lastly to neurological damage caused by viral infection. Although therapeutic opportunities targeting Ephs or ephrins do not seem feasible yet, the current research does propose two models for the viral usage of ephrin-Eph signaling. Firstly, the viral entry model, in which membrane molecules are used for viral entry, leading to cells being used for replication or as a transporter. Secondly, the advantageous expression ephrin-Eph signaling model, where viruses adapt the expression of Ephs or ephrins to change cell-cell interaction to their advantage. These models can guide future research questions on the usage of Ephs or ephrins by viruses and therapeutic opportunities.


Assuntos
Efrinas/metabolismo , Receptores da Família Eph/metabolismo , Receptores Virais/metabolismo , Viroses/virologia , Internalização do Vírus , Vírus/patogenicidade , Animais , Antivirais/uso terapêutico , Endocitose , Interações Hospedeiro-Patógeno , Humanos , Ligantes , Receptores da Família Eph/antagonistas & inibidores , Receptores Virais/antagonistas & inibidores , Viroses/tratamento farmacológico , Viroses/metabolismo , Viroses/transmissão , Internalização do Vírus/efeitos dos fármacos , Replicação Viral , Vírus/crescimento & desenvolvimento , Vírus/metabolismo
5.
Adv Cancer Res ; 147: 303-317, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32593404

RESUMO

Receptor tyrosine kinases (RTKs) are integral membrane sensors that govern cell differentiation, proliferation and mobility, and enable rapid communication between cells and their environment. Of the 20 RTK subfamilies currently known, Eph receptors are the largest group. Together with their corresponding ephrin ligands, Eph receptors regulate a diverse array of physiologic processes including axonal guidance, bone remodeling, and immune cell development and trafficking. Deregulation of Eph signaling pathways is linked to cancer and other proliferative diseases and, because RTKs play critical roles in cancer development, the specific targeting of these molecules in malignancies provides a promising treatment approach. Monoclonal antibodies targeting RTKs represent a potentially attractive modality for pharmaceutical development due to their relatively high target specificity and low off-target binding rates. Therefore, new technologies to generate antibodies able to target RTKs in their native in vivo context are likely to facilitate pre-clinical and clinical development of antibody-based therapies. Our group has recently reported a platform discovery methodology termed Selection of Phage-displayed Accessible Recombinant Targeted Antibodies (SPARTA). SPARTA is a novel and robust stepwise method, which combines the attributes of in vitro screenings of a naïve human recombinant antibody library against known tumor targets with those features of in vivo selections based on tumor-homing capabilities of a pre-enriched antibody pool. This unique approach overcomes several rate-limiting challenges to generate human monoclonal antibodies amenable to rapid translation into medical applications.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias/tratamento farmacológico , Receptores da Família Eph/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacologia , Antineoplásicos Imunológicos/farmacologia , Antineoplásicos Imunológicos/uso terapêutico , Humanos , Terapia de Alvo Molecular , Neoplasias/enzimologia , Neoplasias/patologia , Receptores da Família Eph/metabolismo , Transdução de Sinais
6.
Expert Opin Investig Drugs ; 29(6): 567-582, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32348169

RESUMO

INTRODUCTION: Erythropoietin-producing human hepatocellular (Eph) receptors are among the largest family of tyrosine kinases that are divided into two classes: EphA and EphB receptors. Over the past two decades, their role in cancer has become more evident. AREAS COVERED: There is a need for new anticancer treatments and more insight in the emerging role of Eph receptors in cancer. Molecular mechanisms underlying the pro-tumorigenic effects of Eph receptors could be exploited for future therapeutic strategies. This review describes the variability in expression levels and different effects on oncogenic and tumor suppressive downstream signaling of Eph receptors in various cancer types, and the small molecules, antibodies and peptides that target these receptors. EXPERT OPINION: The complexity of Eph signaling is a challenge for the definition of clear targets for cancer treatment. Nevertheless, numerous drugs that target EphA2 and EphB4 are currently in clinical trials. However, some Eph targeted drugs also inhibit other tyrosine kinases, so it is unclear to what extent the targeting of Eph receptors contributes to their efficacy. Future research is warranted for an improved understanding of the full network in which Eph receptors function. This will be critical for the improvement of the anticancer effects of drugs that target the Eph receptors.


Assuntos
Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Receptores da Família Eph/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Desenvolvimento de Medicamentos , Regulação Neoplásica da Expressão Gênica , Humanos , Terapia de Alvo Molecular , Neoplasias/genética , Neoplasias/patologia , Receptores da Família Eph/genética , Receptores da Família Eph/metabolismo
7.
Org Biomol Chem ; 18(16): 3104-3116, 2020 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-32253415

RESUMO

Eph receptor tyrosine kinases, particularly EphA2 and EphB4, represent promising candidates for molecular imaging due to their essential role in cancer progression and therapy resistance. Xanthine derivatives were identified to be potent Eph receptor inhibitors with IC50 values in the low nanomolar range (1-40 nm). These compounds occupy the hydrophobic pocket of the ATP-binding site in the kinase domain. Based on lead compound 1, we designed two fluorine-18-labelled receptor tyrosine kinase inhibitors ([18F]2/3) as potential tracers for positron emission tomography (PET). Docking into the ATP-binding site allowed us to find the best position for radiolabelling. The replacement of the methyl group at the uracil residue ([18F]3) rather than the methyl group of the phenoxy moiety ([18F]2) by a fluoropropyl group was predicted to preserve the affinity of the lead compound 1. Herein, we point out a synthesis route to [18F]2 and [18F]3 and the respective tosylate precursors as well as a labelling procedure to insert fluorine-18. After radiolabelling, both radiotracers were obtained in approximately 5% radiochemical yield with high radiochemical purity (>98%) and a molar activity of >10 GBq µmol-1. In line with the docking studies, first cell experiments revealed specific, time-dependent binding and uptake of [18F]3 to EphA2 and EphB4-overexpressing A375 human melanoma cells, whereas [18F]2 did not accumulate at these cells. Since both tracers [18F]3 and [18F]2 are stable in rat blood, the novel radiotracers might be suitable for in vivo molecular imaging of Eph receptors with PET.


Assuntos
Radioisótopos de Flúor/química , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/síntese química , Receptores da Família Eph/análise , Xantinas/química , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Efrina-A2/análise , Humanos , Melanoma/diagnóstico por imagem , Melanoma/patologia , Imagem Molecular/métodos , Ratos , Receptor EphA2 , Receptor EphB4/análise , Receptores da Família Eph/antagonistas & inibidores
8.
J Pharm Biomed Anal ; 180: 113067, 2020 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-31891876

RESUMO

The interest on the role of gut microbiota in the biotransformation of drugs and xenobiotics has grown over the last decades and a deeper understanding of the mutual interactions is expected to help future improvements in the fields of drug development, toxicological risk assessment and precision medicine. In this paper, a microbiome drug metabolism case is presented, involving a lipophilic small molecule, N-(3ß-hydroxy-Δ5-cholen-24-oyl)-l-tryptophan, UniPR1331, active as antagonist of the Eph-ephrin system and effective in vivo in a murine orthotopic model of glioblastoma multiforme (GBM). Following the administration of a single 30 mg/kg dose (p.o.) to mice, maximal plasma levels were reached 30 min after dosing and rapidly declined thereafter. To explain the observed in vivo behaviour, in vitro phase I and II metabolism assays were conducted employing mouse and human liver subcellular fractions and profiling main metabolites by means of tandem (HPLC-ESI-MS/MS) and high resolution mass spectrometry (HPLC-ESI-HR-MS). In the presence of in vitro mouse liver fractions, UniPR1331 showed a low phase I metabolic clearance, despite the identification of a 3-oxo and several hydroxylated metabolites. Conversely, after oral administration of UniPR1331 to mice, a novel isobaric metabolite was detected that (i) was subjected, as parent UniPR1331, to enterohepatic circulation (ii) had not been previously identified in vitro in mouse liver microsomes and (iii) was not observed forming after intraperitoneal (i.p.) administration of UniPR1331. An in vitro faecal fermentation assay produced the same chemical entity supporting a major role of gut microbiota in the in vivo clearance of UniPR1331.


Assuntos
Efrinas/antagonistas & inibidores , Microbioma Gastrointestinal/fisiologia , Microssomos Hepáticos/metabolismo , Receptores da Família Eph/antagonistas & inibidores , Animais , Bile/metabolismo , Cromatografia Líquida de Alta Pressão , Fezes/química , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Masculino , Taxa de Depuração Metabólica , Desintoxicação Metabólica Fase I , Desintoxicação Metabólica Fase II , Camundongos , Camundongos Endogâmicos C57BL , Microssomos Hepáticos/efeitos dos fármacos , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem
9.
Neuro Oncol ; 20(9): 1185-1196, 2018 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-29982664

RESUMO

Background: Meningiomas are the most common primary brain tumor in adults, and somatic loss of the neurofibromatosis 2 (NF2) tumor suppressor gene is a frequent genetic event. There is no effective treatment for tumors that recur or continue to grow despite surgery and/or radiation. Therefore, targeted therapies that either delay tumor progression or cause tumor shrinkage are much needed. Our earlier work established mammalian target of rapamycin complex mTORC1/mTORC2 activation in NF2-deficient meningiomas. Methods: High-throughput kinome analyses were performed in NF2-null human arachnoidal and meningioma cell lines to identify functional kinome changes upon NF2 loss. Immunoblotting confirmed the activation of kinases and demonstrated effectiveness of drugs to block the activation. Drugs, singly and in combination, were screened in cells for their growth inhibitory activity. Antitumor drug efficacy was tested in an orthotopic meningioma model. Results: Erythropoietin-producing hepatocellular receptor tyrosine kinases (EPH RTKs), c-KIT, and Src family kinase (SFK) members, which are biological targets of dasatinib, were among the top candidates activated in NF2-null cells. Dasatinib significantly inhibited phospho-EPH receptor A2 (pEPHA2), pEPHB1, c-KIT, and Src/SFK in NF2-null cells, showing no cross-talk with mTORC1/2 signaling. Posttreatment kinome analyses showed minimal adaptive changes. While dasatinib treatment showed some activity, dual mTORC1/2 inhibitor and its combination with dasatinib elicited stronger growth inhibition in meningiomas. Conclusion: Co-targeting mTORC1/2 and EPH RTK/SFK pathways could be a novel effective treatment strategy for NF2-deficient meningiomas.


Assuntos
Antineoplásicos/farmacologia , Biomarcadores Tumorais/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Meníngeas/patologia , Meningioma/patologia , Neurofibromina 2/deficiência , Receptores da Família Eph/antagonistas & inibidores , Animais , Apoptose , Proliferação de Células , Humanos , Neoplasias Meníngeas/tratamento farmacológico , Neoplasias Meníngeas/metabolismo , Meningioma/tratamento farmacológico , Meningioma/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Receptores da Família Eph/genética , Receptores da Família Eph/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Res ; 78(17): 5023-5037, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29945963

RESUMO

Glioblastoma (GBM) carries a dismal prognosis and inevitably relapses despite aggressive therapy. Many members of the Eph receptor tyrosine kinase (EphR) family are expressed by GBM stem cells (GSC), which have been implicated in resistance to GBM therapy. In this study, we identify several EphRs that mark a therapeutically targetable GSC population in treatment-refractory, recurrent GBM (rGBM). Using a highly specific EphR antibody panel and CyTOF (cytometry by time-of-flight), we characterized the expression of all 14 EphR in primary and recurrent patient-derived GSCs to identify putative rGBM-specific EphR. EPHA2 and EPHA3 coexpression marked a highly tumorigenic cell population in rGBM that was enriched in GSC marker expression. Knockdown of EPHA2 and EPHA3 together led to increased expression of differentiation marker GFAP and blocked clonogenic and tumorigenic potential, promoting significantly higher survival in vivo Treatment of rGBM with a bispecific antibody against EPHA2/A3 reduced clonogenicity in vitro and tumorigenic potential of xenografted recurrent GBM in vivo via downregulation of AKT and ERK and increased cellular differentiation. In conclusion, we show that EPHA2 and EPHA3 together mark a GSC population in rGBM and that strategic cotargeting of EPHA2 and EPHA3 presents a novel and rational therapeutic approach for rGBM.Significance: Treatment of rGBM with a novel bispecific antibody against EPHA2 and EPHA3 reduces tumor burden, paving the way for the development of therapeutic approaches against biologically relevant targets in rGBM. Cancer Res; 78(17); 5023-37. ©2018 AACR.


Assuntos
Efrina-A2/genética , Glioblastoma/genética , Recidiva Local de Neoplasia/genética , Receptores Proteína Tirosina Quinases/genética , Animais , Biomarcadores Tumorais/genética , Carcinogênese/genética , Diferenciação Celular/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Efrina-A2/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/radioterapia , Humanos , Camundongos , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/radioterapia , Células-Tronco Neoplásicas/patologia , Prognóstico , Radiação , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptor EphA3 , Receptores da Família Eph/antagonistas & inibidores , Receptores da Família Eph/genética , Temozolomida/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Hematol Oncol ; 10(1): 161, 2017 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-28985758

RESUMO

BACKGROUND: EPH (erythropoietin-producing hepatocellular) receptors are clinically relevant targets in several malignancies. This report describes the effects of GLPG1790, a new potent pan-EPH inhibitor, in human embryonal rhabdomyosarcoma (ERMS) cell lines. METHODS: EPH-A2 and Ephrin-A1 mRNA expression was quantified by real-time PCR in 14 ERMS tumour samples and in normal skeletal muscle (NSM). GLPG1790 effects were tested in RD and TE671 cell lines, two in vitro models of ERMS, by performing flow cytometry analysis, Western blotting and immunofluorescence experiments. RNA interfering experiments were performed to assess the role of specific EPH receptors. Radiations were delivered using an x-6 MV photon linear accelerator. GLPG1790 (30 mg/kg) in vivo activity alone or in combination with irradiation (2 Gy) was determined in murine xenografts. RESULTS: Our study showed, for the first time, a significant upregulation of EPH-A2 receptor and Ephrin-A1 ligand in ERMS primary biopsies in comparison to NSM. GLPG1790 in vitro induced G1-growth arrest as demonstrated by Rb, Cyclin A and Cyclin B1 decrease, as well as by p21 and p27 increment. GLPG1790 reduced migratory capacity and clonogenic potential of ERMS cells, prevented rhabdosphere formation and downregulated CD133, CXCR4 and Nanog stem cell markers. Drug treatment committed ERMS cells towards skeletal muscle differentiation by inducing a myogenic-like phenotype and increasing MYOD1, Myogenin and MyHC levels. Furthermore, GLPG1790 significantly radiosensitized ERMS cells by impairing the DNA double-strand break repair pathway. Silencing of both EPH-A2 and EPH-B2, two receptors preferentially targeted by GLPG1790, closely matched the effects of the EPH pharmacological inhibition. GLPG1790 and radiation combined treatments reduced tumour mass by 83% in mouse TE671 xenografts. CONCLUSIONS: Taken together, our data suggest that altered EPH signalling plays a key role in ERMS development and that its pharmacological inhibition might represent a potential therapeutic strategy to impair stemness and to rescue myogenic program in ERMS cells.


Assuntos
Receptores da Família Eph/antagonistas & inibidores , Rabdomiossarcoma Embrionário/tratamento farmacológico , Rabdomiossarcoma Embrionário/radioterapia , Adolescente , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Camundongos , Camundongos Nus , Tolerância a Radiação/efeitos dos fármacos , Receptores da Família Eph/metabolismo , Rabdomiossarcoma Embrionário/enzimologia , Rabdomiossarcoma Embrionário/patologia , Transdução de Sinais , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Exp Hematol ; 54: 31-39, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28751189

RESUMO

The use of monoclonal antibodies (mAbs) and molecules derived from them has achieved considerable attention and success in recent years, establishing this mode of therapy as an important therapeutic strategy in many cancers, in particular hematological tumors. mAbs recognize cell surface antigens expressed on target cells and mediate their function through various mechanisms such as antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, or immune system modulation. The efficacy of mAb therapy can be improved when they are conjugated to a highly potent payloads, including cytotoxic drugs and radiolabeled isotopes. The Eph family of proteins has received considerable attention in recent years as therapeutic targets for treatment of both solid and hematological cancers. High expression of Eph receptors on cancer cells compared with low expression levels in normal adult tissues makes them an attractive candidate for cancer immunotherapy. In this review, we detail the modes of action of antibody-based therapies with a focus on the Eph family of proteins as potential targets for therapy in hematological malignancies.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Regulação Neoplásica da Expressão Gênica , Neoplasias Hematológicas/tratamento farmacológico , Terapia de Alvo Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Receptores da Família Eph/antagonistas & inibidores , Anticorpos Monoclonais/biossíntese , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Antineoplásicos/uso terapêutico , Ativação do Complemento/efeitos dos fármacos , Citotoxinas/química , Citotoxinas/uso terapêutico , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/patologia , Humanos , Imunoconjugados/química , Imunoconjugados/uso terapêutico , Imunomodulação/efeitos dos fármacos , Imunoterapia/métodos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Receptores da Família Eph/genética , Receptores da Família Eph/imunologia
13.
Eur J Med Chem ; 103: 312-24, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26363867

RESUMO

The Eph receptor-ephrin system is an emerging target for the development of novel anti-angiogenic therapies. Research programs aimed at developing small-molecule antagonists of the Eph receptors are still in their initial stage as available compounds suffer from pharmacological drawbacks, limiting their application in vitro and in vivo. In the present work, we report the design, synthesis and evaluation of structure-activity relationships of a class of Δ(5)-cholenoyl-amino acid conjugates as Eph-ephrin antagonists. As a major achievement of our exploration, we identified N-(3ß-hydroxy-Δ(5)-cholen-24-oyl)-L-tryptophan (UniPR1331) as the first small molecule antagonist of the Eph-ephrin system effective as an anti-angiogenic agent in endothelial cells, bioavailable in mice by the oral route and devoid of biological activity on G protein-coupled and nuclear receptors targeted by bile acid derivatives.


Assuntos
Aminoácidos/farmacologia , Inibidores da Angiogênese/química , Inibidores da Angiogênese/farmacologia , Efrinas/antagonistas & inibidores , Receptores da Família Eph/antagonistas & inibidores , Aminoácidos/síntese química , Aminoácidos/química , Inibidores da Angiogênese/síntese química , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Efrinas/química , Humanos , Masculino , Camundongos , Modelos Moleculares , Estrutura Molecular , Receptores da Família Eph/química , Relação Estrutura-Atividade
14.
Curr Drug Targets ; 16(10): 1048-56, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26302794

RESUMO

Eph-ephrin system is emerging as a new potential target in several diseases including cancer, diabetes, neurodegenerative diseases and inflammation. In the last decade, several efforts have been made to develop small molecule antagonists of Eph receptors. Both natural and synthetic compounds were discovered with (poly) phenol and steroidal derivatives on one side and the α1 agonist doxazosin, 2,5-dimethylpyrrol- 1-yl-benzoic acids and amino acid conjugates of lithocholic acid on the other. In the present paper we critically present available data for these compounds and discuss their potential usefulness as pharmacological tools or as candidates for a lead-optimization program.


Assuntos
Descoberta de Drogas/métodos , Efrinas/metabolismo , Preparações de Plantas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Receptores da Família Eph/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Benzoatos/química , Benzoatos/farmacologia , Ácidos Cólicos/química , Ácidos Cólicos/farmacologia , Doxazossina/química , Doxazossina/farmacologia , Humanos , Modelos Moleculares , Estrutura Molecular , Preparações de Plantas/química , Ligação Proteica , Mapeamento de Interação de Proteínas , Mapas de Interação de Proteínas , Inibidores de Proteínas Quinases/química , Receptores da Família Eph/agonistas , Bibliotecas de Moléculas Pequenas/química
15.
Curr Drug Targets ; 16(10): 1021-30, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26028047

RESUMO

The Eph receptors are the largest sub-family of Receptor Tyrosine Kinases (RTK). They, together with their ephrin ligands, play central roles in cell-cell communication during development, and also in the maintenance of a normal adult physiology. Their malfunction, therefore, can contribute to various human diseases. Since the structures of the Eph receptors and ephrins are by now well characterized, there has been extensive recent work to develop ways to manipulate their action in order to achieve therapeutic benefits. Although few reagents have progressed to clinical trials thus far, it is evident that the Eph receptors are valid targets for therapeutic drugs. In this review we first summarize studies on the three-dimensional structures of Eph receptors. We then give an overview on small molecule inhibitors and activators using Ephs as targets. We put a special focus on the latest developments in the field of monoclonal antibodies and antibody fragments for inhibiting or activating the Eph/ephrin signaling.


Assuntos
Descoberta de Drogas , Efrinas/metabolismo , Receptores da Família Eph/metabolismo , Anticorpos de Cadeia Única/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Sítios de Ligação , Humanos , Modelos Moleculares , Conformação Proteica , Receptores da Família Eph/agonistas , Receptores da Família Eph/antagonistas & inibidores , Receptores da Família Eph/química , Transdução de Sinais/efeitos dos fármacos , Anticorpos de Cadeia Única/química , Bibliotecas de Moléculas Pequenas/química
16.
Artigo em Inglês | MEDLINE | ID: mdl-25772170

RESUMO

Evidence to show that the Eph/ephrin system is involved in inflammation induced by infection, injury, inflammatory diseases, and atherosclerosis has been increased. Although the roles of the Eph/ephrin system in both neural and vascular development as well as cell motility are well documented, its involvement in inflammatory processes has not yet been elucidated in detail. Moreover, the soluble form of artificially oligomerized or dimerized Fc-fused ephrin-A1 has been widely used in in vitro and/or in vivo studies to activate the EphA receptors, whereas its physiological functions as a membrane-anchored protein remain largely unknown. Recent studies using clinical samples reported that the overexpression of Ephs and ephrins in some tumors such as hepatocellular carcinoma positively correlated with both malignancy of tumors and the poor prognosis of cancer patients. However, the molecular mechanisms underlying malignancy of tumors are not fully understood. The author herein summarizes the molecular mechanisms of the Eph/ephrin system involved in the immune system and inflammatory processes. Especially, the author focuses on inflammation-induced physiological changes in vascular endothelial cells leading to vascular hyper-permeability and described them in this review. The author also introduces those that contribute to ephrin-A1-mediated lung metastasis.


Assuntos
Anti-Inflamatórios/uso terapêutico , Efrinas/antagonistas & inibidores , Mediadores da Inflamação/antagonistas & inibidores , Inflamação/tratamento farmacológico , Neoplasias/tratamento farmacológico , Receptores da Família Eph/antagonistas & inibidores , Animais , Antineoplásicos/uso terapêutico , Permeabilidade Capilar , Desenho de Fármacos , Células Endoteliais/metabolismo , Efrinas/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Terapia de Alvo Molecular , Metástase Neoplásica , Neoplasias/metabolismo , Neoplasias/patologia , Receptores da Família Eph/metabolismo , Transdução de Sinais/efeitos dos fármacos
17.
Cytokine Growth Factor Rev ; 26(1): 1-6, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24933439

RESUMO

Eph receptor tyrosine kinases (RTKs) and their ligands, ephrins, play critical roles in development, tissue homeostasis, and cancer. Because Eph receptors are expressed in most adult stem cell niches and in many types of cancers, it has been long suspected that this family of RTKs may also regulate the function of cancer stem-like cells (CSCs). This review will focus on recent studies to elucidate the contribution of Eph/ephrin molecules in CSC self-renewal and tumorigenicity, as well as describe efforts to target these molecules in cancer. Because CSCs are often resistant to therapeutic intervention and have been shown to depend on Eph RTKs for self-renewal, targeting Eph receptors may hold promise for the treatment of drug-resistant cancers.


Assuntos
Efrinas/metabolismo , Células-Tronco Neoplásicas/metabolismo , Receptores da Família Eph/metabolismo , Animais , Divisão Celular , Homeostase , Humanos , Camundongos , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Receptores da Família Eph/agonistas , Receptores da Família Eph/antagonistas & inibidores , Transdução de Sinais
18.
Neuroscience ; 284: 546-554, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25453775

RESUMO

EphB receptors and their ephrinB ligands are implicated in modulating of spinal nociceptive information processing. Here, we investigated whether protein kinase A (PKA), acts as a downstream effector, participates in the modulation spinal nociceptive information related to ephrinB-EphB signaling. Intrathecal injection of ephrinB2-Fc caused thermal hyperalgesia and mechanical allodynia, which were accompanied by increased expression of spinal PKA catalytic subunit (PKAca) and phosphorylated cAMP-response element-binding protein (p-CREB). Pre-treatment with H89, a PKA inhibitor, prevented the activation of CREB by ephrinB2-Fc. Inhibition of spinal PKA signaling prevented and reversed pain behaviors induced by the intrathecal injection of ephrinB2-Fc. Furthermore, blockade of the EphB receptors by intrathecal injection of EphB2-Fc reduced formalin-induced inflammatory, chronic constrictive injury (CCI)-induced neuropathic, and tibia bone cavity tumor cell implantation (TCI)-induced bone cancer pain behaviors, which were accompanied by decreased expression of spinal PKAca and p-CREB. Overall, these results confirmed the important involvement of PKA in the modulation of spinal nociceptive information related to ephrinBs-EphBs signaling. This finding may have important implications for exploring the roles and mechanisms of ephrinB-EphB signaling in physiologic and pathologic pain.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Efrina-B2/metabolismo , Nociceptividade/fisiologia , Receptores da Família Eph/metabolismo , Medula Espinal/metabolismo , Animais , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/fisiopatologia , Linhagem Celular Tumoral , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Modelos Animais de Doenças , Efrina-B2/administração & dosagem , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Inflamação/tratamento farmacológico , Inflamação/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neuralgia/tratamento farmacológico , Neuralgia/fisiopatologia , Nociceptividade/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Receptores da Família Eph/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos
19.
Growth Factors ; 32(6): 176-89, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25391995

RESUMO

Eph receptor tyrosine kinases control cell-cell interactions during normal and oncogenic development, and are implicated in a range of processes including angiogenesis, stem cell maintenance and metastasis. They are thus of great interest as targets for cancer therapy. EphA3, originally isolated from leukemic and melanoma cells, is presently one of the most promising therapeutic targets, with multiple tumor-promoting roles in a variety of cancer types. This review focuses on EphA3, its functions in controlling cellular behavior, both in normal and pathological development, and most particularly in cancer.


Assuntos
Neoplasias/metabolismo , Receptores da Família Eph/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Humanos , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Receptores da Família Eph/antagonistas & inibidores , Receptores da Família Eph/química , Receptores da Família Eph/genética
20.
Growth Factors ; 32(6): 207-13, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25413947

RESUMO

Several Eph receptor tyrosine kinases (RTKs) are commonly over-expressed in epithelial and mesenchymal cancers and are recognized as promising therapeutic targets. Although normal interaction between Eph receptors and their ephrin ligands stimulates kinase activity and is generally tumor suppressive, significant Eph over-expression allows activation of ligand- and/or kinase-independent signaling pathways that promote oncogenesis. Single-agent kinase inhibitors are widely used to target RTK-driven tumors but acquired and de novo resistance to such agents is a major limitation to effective clinical use. Accumulating evidence suggests that Ephs can be inhibited by "leaky" or low-specificity kinase inhibitors targeted at other RTKs. Such off-target effects may therefore inadvertently promote ligand- and/or kinase-independent oncogenic Eph signaling, thereby providing a new mechanism by which resistance to the RTK inhibitors can emerge. We propose that combining specific, non-leaky kinase inhibitors with tumor-suppressive stimulators of Eph signaling may provide more effective treatment options for overcoming treatment-induced resistance and clinical failure.


Assuntos
Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Neoplasias/metabolismo , Inibidores de Proteínas Quinases/uso terapêutico , Receptores da Família Eph/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Humanos , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Receptores da Família Eph/genética , Receptores da Família Eph/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...