Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Endocr Rev ; 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-38091968

RESUMEN

Our overview covers several key areas related to recent results obtained for collagen type VI and endotrophin (ETP): i) An introduction to the history of ETP, including how it was identified, how it is released and its function and potential receptors. ii) An introduction to the collagen family, with a focus on what differentiates collagen type VI from an evolutionary standpoint. iii) An overview of collagen type VI, the six individual chains (COL6A1, A2, A3, A4, A5 and A6), their differences and similarities, as well as their expression profiles and function. iv) A detailed analysis of COL6A3, including the cleaved product endotrophin, and what separates it from the other five collagen 6 molecules, including its suggested function based on insights gained from knockout and gain of function mouse models. v) An introduction to the history of ETP, including how it was identified, how it is released and its function and potential receptors. vi) The pathology of ETP. What leads to its presence and release and what are the consequences thereof? vii) Functional implications of circulating ETP. Here we review the data with the functional roles of ETP in mind. viii) We propose that ETP is a mediator for fibrotic (or fibro-inflammatory? ) disorders. Based on what we know about ETP, we have to consider it as a target for the treatment of fibrotic (or fibro-inflammatory) disorders. What segment(s) of the patient population would most dramatically respond to an ETP-targeted intervention? How can we find the population that would profit most from an intervention? We aim to present a broad overview over the ETP field at large, providing an assessment of where the future research efforts need to be placed to tap into the vast potential of ETP, both as a marker and as a target in different diseases.

2.
J Immunother Cancer ; 11(6)2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37328286

RESUMEN

BACKGROUND: Immune exclusion (IE) where tumors deter the infiltration of immune cells into the tumor microenvironment has emerged as a key mechanism underlying immunotherapy resistance. We recently reported a novel role of discoidin domain-containing receptor 1 (DDR1) in promoting IE in breast cancer and validated its critical role in IE using neutralizing rabbit monoclonal antibodies (mAbs) in multiple mouse tumor models. METHODS: To develop a DDR1-targeting mAb as a potential cancer therapeutic, we humanized mAb9 with a complementarity-determining region grafting strategy. The humanized antibody named PRTH-101 is currently being tested in a Phase 1 clinical trial. We determined the binding epitope of PRTH-101 from the crystal structure of the complex between DDR1 extracellular domain (ECD) and the PRTH-101 Fab fragment with 3.15 Å resolution. We revealed the underlying mechanisms of action of PRTH-101 using both cell culture assays and in vivo study in a mouse tumor model. RESULTS: PRTH-101 has subnanomolar affinity to DDR1 and potent antitumor efficacy similar to the parental rabbit mAb after humanization. Structural information illustrated that PRTH-101 interacts with the discoidin (DS)-like domain, but not the collagen-binding DS domain of DDR1. Mechanistically, we showed that PRTH-101 inhibited DDR1 phosphorylation, decreased collagen-mediated cell attachment, and significantly blocked DDR1 shedding from the cell surface. Treatment of tumor-bearing mice with PRTH-101 in vivo disrupted collagen fiber alignment (a physical barrier) in the tumor extracellular matrix (ECM) and enhanced CD8+ T cell infiltration in tumors. CONCLUSIONS: This study not only paves a pathway for the development of PRTH-101 as a cancer therapeutic, but also sheds light on a new therapeutic strategy to modulate collagen alignment in the tumor ECM for enhancing antitumor immunity.


Asunto(s)
Anticuerpos Monoclonales , Receptor con Dominio Discoidina 1 , Neoplasias , Animales , Ratones , Colágeno/metabolismo , Receptor con Dominio Discoidina 1/metabolismo , Matriz Extracelular/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Microambiente Tumoral , Anticuerpos Monoclonales/farmacología
3.
Front Immunol ; 14: 1084887, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37033994

RESUMEN

Background: The immune cell topography of solid tumors has been increasingly recognized as an important predictive factor for progression of disease and response to immunotherapy. The distribution pattern of immune cells in solid tumors is commonly classified into three categories - namely, "Immune inflamed", "Immune desert" and "Immune excluded" - which, to some degree, connect immune cell presence and positioning within the tumor microenvironment to anti-tumor activity. Materials and methods: In this review, we look at the ways immune exclusion has been defined in published literature and identify opportunities to develop consistent, quantifiable definitions, which in turn, will allow better determination of the underlying mechanisms that span cancer types and, ultimately, aid in the development of treatments to target these mechanisms. Results: The definitions of tumor immune phenotypes, especially immune exclusion, have largely been conceptual. The existing literature lacks in consistency when it comes to practically defining immune exclusion, and there is no consensus on a definition. Majority of the definitions use somewhat arbitrary cut-offs in an attempt to place each tumor into a distinct phenotypic category. Tumor heterogeneity is often not accounted for, which limits the practical application of a definition. Conclusions: We have identified two key issues in existing definitions of immune exclusion, establishing clinically relevant cut-offs within the spectrum of immune cell infiltration as well as tumor heterogeneity. We propose an approach to overcome these limitations, by reporting the degree of immune cell infiltration, tying cut-offs to clinically meaningful outcome measures, maximizing the number of regions of a tumor that are analyzed and reporting the degree of heterogeneity. This will allow for a consensus practical definition for operationalizing this categorization into clinical trial and signal-seeking endpoints.


Asunto(s)
Neoplasias , Escape del Tumor , Humanos , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Microambiente Tumoral/inmunología , Consenso , Escape del Tumor/inmunología
4.
Br J Pharmacol ; 175(7): 1054-1065, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29329501

RESUMEN

BACKGROUND AND PURPOSE: Anaemia of chronic disease (ACD) has been linked to iron-restricted erythropoiesis imposed by high circulating levels of hepcidin, a 25 amino acid hepatocyte-derived peptide that controls systemic iron homeostasis. Here, we report the engineering of the human lipocalin-derived, small protein-based anticalin PRS-080 hepcidin antagonist with high affinity and selectivity. EXPERIMENTAL APPROACH: Anticalin- and hepcidin-specific pharmacokinetic (PK)/pharmacodynamic modelling (PD) was used to design and select the suitable drug candidate based on t1/2 extension and duration of hepcidin suppression. The development of a novel free hepcidin assay enabled accurate analysis of bioactive hepcidin suppression and elucidation of the observed plasma iron levels after PRS-080-PEG30 administration in vivo. KEY RESULTS: PRS-080 had a hepcidin-binding affinity of 0.07 nM and, after coupling to 30 kD PEG (PRS-080-PEG30), a t1/2 of 43 h in cynomolgus monkeys. Dose-dependent iron mobilization and hepcidin suppression were observed after a single i.v. dose of PRS-080-PEG30 in cynomolgus monkeys. Importantly, in these animals, suppression of free hepcidin and subsequent plasma iron elevation were sustained during repeated s.c. dosing. After repeated dosing and followed by a treatment-free interval, all iron parameters returned to pre-dose values. CONCLUSIONS AND IMPLICATIONS: In conclusion, we developed a dose-dependent and safe approach for the direct suppression of hepcidin, resulting in prolonged iron mobilization to alleviate iron-restricted erythropoiesis that can address the root cause of ACD. PRS-080-PEG30 is currently in early clinical development.


Asunto(s)
Hepcidinas/antagonistas & inhibidores , Hepcidinas/sangre , Hierro/sangre , Animales , Femenino , Macaca fascicularis , Masculino , Modelos Biológicos
5.
Biosens Bioelectron ; 100: 139-147, 2018 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-28886458

RESUMEN

Schizophrenia treatment may see a paradigm shift due to development of new atypical antipsychotic drugs (APDs), with better tolerability due to more selective dopamine (DA) receptor blockade. Monitoring of these APD candidates in biological fluids is of great importance to reduce the development cost, to clarify the mechanism of action and ultimately to support the demonstration of efficacy of these molecules. Electrochemical approaches have attracted great attention for monitoring DA and APD levels but none of the methods developed so far aimed to screen APD candidates. Herein, by this work, we propose for the first time an electrochemical ligand-binding approach for antipsychotic drug screening where competitive binding of a novel APD and DA to a dopamine D3 receptor (D3R) was investigated by looking at electrochemical signals of DA and drug before and after D3R interaction. D3R peptide was incubated with DA and/or drug first and then changes in electrochemical oxidation signals of free DA and the drug was measured by cyclic voltammetry (CV) and differential pulse voltammetry (DPV). Circular Dichroism spectroscopy was used to investigate the secondary structure of the peptide upon binding with either drug and/or DA.


Asunto(s)
Antipsicóticos/farmacología , Técnicas Biosensibles/métodos , Dopamina/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Técnicas Electroquímicas/métodos , Receptores de Dopamina D3/metabolismo , Humanos , Modelos Moleculares , Estructura Secundaria de Proteína/efectos de los fármacos , Receptores de Dopamina D3/química , Esquizofrenia/tratamiento farmacológico
6.
PLoS One ; 12(6): e0179991, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28662116

RESUMEN

BACKGROUND: Actinic keratoses (AK) are pre-malignant cutaneous lesions caused by prolonged exposure to ultraviolet radiation. As AKs lesions are generally accepted to be the initial lesions in a disease continuum that progresses to squamous cell carcinoma (SCC), AK lesions have to be treated. They are also the second most common reason for visits to the dermatologist. Several treatments are available but their efficacy still needs to be improved. The UV-B-induced KA lesion mouse model is used in preclinical studies to assess the efficacy of novel molecules, even though it is often more representative of advanced AK or SCC. OBJECTIVES: Here we report on a translational study, comparing the various stages of AK development in humans and in the UV-B irradiated mouse model, as well as the optimization of photograph acquisition of AK lesions on mouse skin. METHODS: Human and mouse skin lesions were analysed by histology and immunohistochemistry. Mouse lesions were also assessed using a digital dermatoscope. RESULTS: An histological and phenotypic analysis, including p53, Ki67 and CD3 expression detection, performed on human and mouse AK lesions, shows that overall AK modelling in mice is relevant in the clinical situation. Some differences are observed, such as disorganization of keratinocytes of the basal layer and a number of atypical nuclei which are more numerous in human AK, whereas much more pronounced acanthosis is observed in skin lesion in mice. Thanks to this translational study, we are able to select appropriate experimental conditions for establishing either early or advanced stage AK or an SCC model. Furthermore, we optimized photograph acquisition of AK lesions on mouse skin by using a digital dermatoscope which is also used in clinics and allows reproducible photograph acquisition for further reliable assessment of mouse lesions. Use of this camera is illustrated through a pharmacological study assessing the activity of CARAC®. CONCLUSION: These data demonstrate that this mouse model of UV-B-induced skin lesions is predictive for the identification of novel therapeutic treatments for both early and advanced stages of the disease.


Asunto(s)
Modelos Animales de Enfermedad , Queratosis Actínica/patología , Investigación Biomédica Traslacional , Animales , Dermoscopía , Fluorouracilo/uso terapéutico , Humanos , Inmunosupresores/uso terapéutico , Queratosis Actínica/tratamiento farmacológico , Ratones , Ratones Pelados , Rayos Ultravioleta
7.
Angiogenesis ; 19(1): 79-94, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26650228

RESUMEN

Human tear lipocalin (Tlc) was utilized as a protein scaffold to engineer an Anticalin that specifically binds and functionally blocks vascular endothelial growth factor A (VEGF-A), a pivotal inducer of physiological angiogenesis that also plays a crucial role in several neovascular diseases. Starting from a naive combinatorial library where residues that form the natural ligand-binding site of Tlc were randomized, followed by affinity maturation, the final Anticalin PRS-050 was selected to bind all major splice forms of VEGF-A with picomolar affinity. Moreover, this Anticalin cross-reacts with the murine ortholog. PRS-050 efficiently antagonizes the interaction between VEGF-A and its cellular receptors, and it inhibits VEGF-induced mitogenic signaling as well as proliferation of primary human endothelial cells with subnanomolar IC50 values. Intravitreal administration of the Anticalin suppressed VEGF-induced blood-retinal barrier breakdown in a rabbit model. To allow lasting systemic neutralization of VEGF-A in vivo, the plasma half-life of the Anticalin was extended by site-directed PEGylation. The modified Anticalin efficiently blocked VEGF-mediated vascular permeability as well as growth of tumor xenografts in nude mice, concomitantly with reduction in microvessel density. In contrast to bevacizumab, the Anticalin did not trigger platelet aggregation and thrombosis in human FcγRIIa transgenic mice, thus suggesting an improved safety profile. Since neutralization of VEGF-A activity is well known to exert beneficial effects in cancer and other neovascular diseases, including wet age-related macular degeneration, this Anticalin offers a novel potent small protein antagonist for differentiated therapeutic intervention in oncology and ophthalmology.


Asunto(s)
Lipocalinas/farmacología , Terapia Molecular Dirigida , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Bevacizumab/farmacología , Bevacizumab/uso terapéutico , Barrera Hematorretinal/patología , Permeabilidad Capilar , Proliferación Celular/efectos de los fármacos , Femenino , Semivida , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Lipocalinas/farmacocinética , Lipocalinas/uso terapéutico , Ratones Desnudos , Ratones Transgénicos , Mitógenos/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Polietilenglicoles/química , Ingeniería de Proteínas , Conejos , Receptores de IgG/metabolismo , Transducción de Señal , Resonancia por Plasmón de Superficie , Trombosis/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
J Nucl Med ; 55(4): 665-71, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24614223

RESUMEN

UNLABELLED: Anticalins are a novel class of biopharmaceuticals, displaying highly desirable attributes as imaging agents. The anticalin PRS-110 was rationally engineered to target the oncogene MET with high affinity and specificity. The aim of this study was to visualize MET expression and analyze biodistribution of (89)Zr-labeled PRS-110 in human tumor-bearing mice. METHODS: (89)Zr-PRS-110 was generated. For biodistribution studies (96 h after injection of tracer) 10 µg of (89)Zr-PRS-110 (with 0-490 µg of unlabeled PRS-110) were injected into BALB/c mice bearing high MET-expressing H441 non-small cell lung cancer xenografts. Further characterization with PET imaging was performed at 6, 24, 48, and 96 h after injection of 50 µg of (89)Zr-PRS-110 into mice bearing H441, primary glioblastoma U87-MG (intermediate MET), or ovarian cancer A2780 (low MET) xenografts. Drug distribution was also analyzed ex vivo using fluorescently labeled PRS-110. RESULTS: Biodistribution analyses showed a dose-dependent tumor uptake of (89)Zr-PRS-110, with the highest fractional tumor uptake at 10 µg of (89)Zr-PRS-110, with no unlabeled PRS-110. Small-animal PET imaging supported by biodistribution data revealed specific tumor uptake of (89)Zr-PRS-110 in the MET-expressing H441 and U87-MG tumors whereas the MET-negative A2780 tumor model showed a lower uptake similar to a non-MET binder anticalin control. Tumor uptake increased up to 24 h after tracer injection and remained high, whereas uptake in other organs decreased over time. Ex vivo fluorescence revealed intracellular presence of PRS-110. CONCLUSION: (89)Zr-PRS-110 specifically accumulates in MET-expressing tumors in a receptor density-dependent manner. PET imaging provides real-time noninvasive information about PRS-110 distribution and tumor accumulation in preclinical models.


Asunto(s)
Proteínas , Proteínas Proto-Oncogénicas c-met/biosíntesis , Radiofármacos , Animales , Anticuerpos Monoclonales Humanizados , Bevacizumab , Unión Competitiva , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Marcaje Isotópico , Lipocalinas , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Control de Calidad , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
9.
PLoS One ; 8(12): e83232, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24349470

RESUMEN

BACKGROUND: To report the nonrandomized first-in-human phase I trial of PRS-050, a novel, rationally engineered Anticalin based on human tear lipocalin that targets and antagonizes vascular endothelial growth factor A (VEGF-A). METHODS: Patients with advanced solid tumors received PRS-050 at 0.1 mg/kg to 10 mg/kg by IV in successive dosing cohorts according to the 3+3 escalation scheme. The primary end point was safety. RESULTS: Twenty-six patients were enrolled; 25 were evaluable. Two patients experienced dose-limiting toxicity, comprising grade (G) 3 hypertension and G3 pyrexia, respectively. The maximum tolerated dose was not reached. Most commonly reported treatment-emergent adverse events (AEs) included chills (52%; G3, 4%), fatigue (52%; G3, 4%), hypertension (44%; G3, 16%), and nausea (40%, all G1/2). No anti-PRS-050 antibodies following multiple administration of the drug were detected. PRS-050 showed dose-proportional pharmacokinetics (PK), with a terminal half-life of approximately 6 days. Free VEGF-A was detectable at baseline in 9/25 patients, becoming rapidly undetectable after PRS-050 infusion for up to 3 weeks. VEGF-A/PRS-050 complex was detectable for up to 3 weeks at all dose levels, including in patients without detectable baseline-free VEGF-A. We also detected a significant reduction in circulating matrix metalloproteinase 2, suggesting this end point could be a pharmacodynamic (PD) marker of the drug's activity. CONCLUSIONS: PRS-050, a novel Anticalin with high affinity for VEGF-A, was well-tolerated when administered at the highest dose tested, 10 mg/kg. Based on target engagement and PK/PD data, the recommended phase II dose is 5 mg/kg every 2 weeks administered as a 120-minute infusion. TRIAL REGISTRATION: ClinicalTrials.gov NCT01141257 http://clinicaltrials.gov/ct2/show/NCT01141257.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/farmacocinética , Lipocalina 1 , Lipocalinas/administración & dosificación , Lipocalinas/farmacocinética , Neoplasias/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Adulto , Anciano , Inhibidores de la Angiogénesis/efectos adversos , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Lipocalinas/efectos adversos , Metaloproteinasa 2 de la Matriz/sangre , Persona de Mediana Edad , Neoplasias/sangre , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/sangre
10.
Mol Cancer Ther ; 12(11): 2459-71, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24002935

RESUMEN

Activation of the MET oncogenic pathway has been implicated in the development of aggressive cancers that are difficult to treat with current chemotherapies. This has led to an increased interest in developing novel therapies that target the MET pathway. However, most existing drug modalities are confounded by their inability to specifically target and/or antagonize this pathway. Anticalins, a novel class of monovalent small biologics, are hypothesized to be "fit for purpose" for developing highly specific and potent antagonists of cancer pathways. Here, we describe a monovalent full MET antagonist, PRS-110, displaying efficacy in both ligand-dependent and ligand-independent cancer models. PRS-110 specifically binds to MET with high affinity and blocks hepatocyte growth factor (HGF) interaction. Phosphorylation assays show that PRS-110 efficiently inhibits HGF-mediated signaling of MET receptor and has no agonistic activity. Confocal microscopy shows that PRS-110 results in the trafficking of MET to late endosomal/lysosomal compartments in the absence of HGF. In vivo administration of PRS-110 resulted in significant, dose-dependent tumor growth inhibition in ligand-dependent (U87-MG) and ligand-independent (Caki-1) xenograft models. Analysis of MET protein levels on xenograft biopsy samples show a significant reduction in total MET following therapy with PRS-110 supporting its ligand-independent mechanism of action. Taken together, these data indicate that the MET inhibitor PRS-110 has potentially broad anticancer activity that warrants evaluation in patients.


Asunto(s)
Lipocalinas/farmacología , Neoplasias Experimentales/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas/farmacología , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Sitios de Unión/efectos de los fármacos , Células CHO , Línea Celular Tumoral , Cricetulus , Relación Dosis-Respuesta a Droga , Mapeo Epitopo , Femenino , Células HT29 , Factor de Crecimiento de Hepatocito/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ligandos , Lipocalinas/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Experimentales/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas/uso terapéutico , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Glia ; 59(2): 208-18, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21125641

RESUMEN

Neuropathic pain produced by damage to or dysfunction of the nervous system is a common and severely disabling state that affects millions of people worldwide. Recent evidence indicates that activated microglia are key cellular intermediaries in the pathogenesis of neuropathic pain and that ATP serves as the mediator. However, the in vivo mechanism underlying the retention of activated microglia in the injured region has not yet been completely elucidated. Prostaglandin E(2) (PGE(2)) is the principal proinflammatory prostanoid and plays versatile roles by acting via four PGE receptor subtypes, EP1-EP4. In the present study, we investigated the role of PGE(2) in spinal microglial activation in relation to neuropathic pain by using genetic and pharmacological methods. Mice deficient in microsomal prostaglandin E synthase-1 impaired the activation of microglia and the NMDA-nitric oxide (NO) cascade in spinal neurons in the dorsal horn and did not exhibit mechanical allodynia after peripheral nerve injury. The intrathecal injection of indomethacin, a nonsteroidal anti-inflammatory drug, ONO-8713, a selective EP1 antagonist, or 7-nitroindole, a neuronal NO synthase inhibitor, attenuated mechanical allodynia and the increase in activated microglia observed in the established neuropathic-pain state. We further demonstrated that ATP-induced microglial migration was blocked in vitro by PGE(2) via EP2 and by S-nitrosoglutathione, an NO donor. Taken together, the present study suggests that PGE(2) participated in the maintenance of neuropathic pain in vivo not only by activating spinal neurons, but also by retaining microglia in the central terminals of primary afferent fibers via EP2 subtype and via EP1-mediated NO production.


Asunto(s)
Movimiento Celular/fisiología , Dinoprostona/metabolismo , Microglía/fisiología , Neuralgia/metabolismo , Neuralgia/patología , Médula Espinal/patología , Adenosina Trifosfato/farmacología , Animales , Movimiento Celular/genética , Corteza Cerebral/citología , Cinamatos/farmacología , Cinamatos/uso terapéutico , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Indazoles/farmacología , Indazoles/uso terapéutico , Oxidorreductasas Intramoleculares/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Microglía/efectos de los fármacos , Neuralgia/complicaciones , Neuralgia/tratamiento farmacológico , Neuronas/metabolismo , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Prostaglandina-E Sintasas , S-Nitrosoglutatión/farmacología , Médula Espinal/efectos de los fármacos , Nervios Espinales/lesiones
12.
Biologics ; 4: 163-71, 2010 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-20631820

RESUMEN

Biological therapies, such as monoclonal antibodies (mAbs) that target tumor-associated antigens have been considered an effective therapeutic approach in oncology. In considering Notch-1 receptor as a potential target, we performed immunohistochemistry on tissue microarrays to determine 1) whether the receptor is overexpressed in tumor cells as compared to their corresponding normal tissues and 2) the clinical significance of its expression levels in human breast, colorectal, lung and prostate cancers. We found that the expression of Notch-1 protein was overexpressed in primary colorectal adenocarcinoma and nonsmall cell lung carcinoma (NSCLC), but not in primary ductal breast carcinoma or prostate adenocarcinoma. Further analysis revealed that higher levels of Notch-1 protein expression were significantly associated with poorer differentiation of breast and prostate tumors. Strikingly, for NSCLC, the expression levels of Notch-1 protein were found to be inversely correlated with tumor differentiation and progression. For colorectal tumors, however, no correlation of Notch-1 protein expression was found with any tumor clinicopathological parameters, in spite of its overexpression in tumor cells. Our data demonstrated the complexity of Notch-1 protein expression in human solid tumors and further supported the notion that the roles of Notch-1 expression in tumorigenesis are highly context-dependent. The findings could provide the basis for development of distinct therapeutic strategies of Notch-1 mAbs for its applications in the treatment of suitable types of human cancers.

13.
PLoS One ; 5(2): e9094, 2010 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-20161710

RESUMEN

BACKGROUND: Notch receptors normally play a key role in guiding a variety of cell fate decisions during development and differentiation of metazoan organisms. On the other hand, dysregulation of Notch1 signaling is associated with many different types of cancer as well as tumor angiogenesis, making Notch1 a potential therapeutic target. PRINCIPAL FINDINGS: Here we report the in vitro activities of inhibitory Notch1 monoclonal antibodies derived from cell-based and solid-phase screening of a phage display library. Two classes of antibodies were found, one directed against the EGF-repeat region that encompasses the ligand-binding domain (LBD), and the second directed against the activation switch of the receptor, the Notch negative regulatory region (NRR). The antibodies are selective for Notch1, inhibiting Jag2-dependent signaling by Notch1 but not by Notch 2 and 3 in reporter gene assays, with EC(50) values as low as 5+/-3 nM and 0.13+/-0.09 nM for the LBD and NRR antibodies, respectively, and fail to recognize Notch4. While more potent, NRR antibodies are incomplete antagonists of Notch1 signaling. The antagonistic activity of LBD, but not NRR, antibodies is strongly dependent on the activating ligand. Both LBD and NRR antibodies bind to Notch1 on human tumor cell lines and inhibit the expression of sentinel Notch target genes, including HES1, HES5, and DTX1. NRR antibodies also strongly inhibit ligand-independent signaling in heterologous cells transiently expressing Notch1 receptors with diverse NRR "class I" point mutations, the most common type of mutation found in human T-cell acute lymphoblastic leukemia (T-ALL). In contrast, NRR antibodies failed to antagonize Notch1 receptors bearing rare "class II" or "class III" mutations, in which amino acid insertions generate a duplicated or constitutively sensitive metalloprotease cleavage site. Signaling in T-ALL cell lines bearing class I mutations is partially refractory to inhibitory antibodies as compared to cell-penetrating gamma-secretase inhibitors. CONCLUSIONS/SIGNIFICANCE: Antibodies that compete with Notch1 ligand binding or that bind to the negative regulatory region can act as potent inhibitors of Notch1 signaling. These antibodies may have clinical utility for conditions in which inhibition of signaling by wild-type Notch1 is desired, but are likely to be of limited value for treatment of T-ALLs associated with aberrant Notch1 activation.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Mutación , Receptor Notch1/inmunología , Transducción de Señal/efectos de los fármacos , Células 3T3 , Animales , Especificidad de Anticuerpos/inmunología , Sitios de Unión/genética , Sitios de Unión/inmunología , Unión Competitiva , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-2 , Ligandos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Receptor Notch1/genética , Receptor Notch1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Hypertension ; 55(2): 531-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20065147

RESUMEN

Prostaglandin (PG)E(2) has multiple actions that may affect blood pressure. It is synthesized from arachidonic acid by the sequential actions of phospholipases, cyclooxygenases, and PGE synthases. Although microsomal PGE synthase (mPGES)1 is the only genetically verified PGE synthase, results of previous studies examining the consequences of mPGES1 deficiency on blood pressure (BP) are conflicting. To determine whether genetic background modifies the impact of mPGES1 on BP, we generated mPGES1(-/-) mice on 2 distinct inbred backgrounds, DBA/1lacJ and 129/SvEv. On the DBA/1 background, baseline BP was similar between wild-type (WT) and mPGES1(-/-) mice. By contrast, on the 129 background, baseline BPs were significantly higher in mPGES1(-/-) animals than WT controls. During angiotensin II infusion, the DBA/1 mPGES1(-/-) and WT mice developed mild hypertension of similar magnitude, whereas 129-mPGES1(-/-) mice developed more severe hypertension than WT controls. DBA/1 animals developed only minimal albuminuria in response to angiotensin II infusion. By contrast, WT 129 mice had significantly higher levels of albumin excretion than WT DBA/1 and the extent of albuminuria was further augmented in 129 mPGES1(-/-) animals. In WT mice of both strains, the increase in urinary excretion of PGE(2) with angiotensin II was attenuated in mPGES1(-/-) animals. Urinary excretion of thromboxane was unaffected by angiotensin II in the DBA/1 lines but increased more than 4-fold in 129 mPGES1(-/-) mice. These data indicate that genetic background significantly modifies the BP response to mPGES1 deficiency. Exaggerated production of thromboxane may contribute to the robust hypertension and albuminuria in 129 mPGES1-deficient mice.


Asunto(s)
Albuminuria/metabolismo , Hipertensión/enzimología , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Análisis de Varianza , Angiotensina II/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica , Hipertensión/inducido químicamente , Oxidorreductasas Intramoleculares/deficiencia , Riñón/efectos de los fármacos , Riñón/metabolismo , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Microsomas/metabolismo , Probabilidad , Prostaglandina-E Sintasas , Prostaglandina-Endoperóxido Sintasas/metabolismo , Prostaglandinas/metabolismo , ARN Mensajero/genética , Distribución Aleatoria , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
MAbs ; 2(1): 14-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20081379

RESUMEN

Approval of an anti-CD20 chimeric monoclonal antibody, rituximab, has revolutionized cancer treatment and also validated CD20 targeting for providing benefit and improvement of overall response rate in B cell malignancies. Although many patients have benefited from the treatment of rituximab, there are still significant numbers of patients who are refractory or develop resistance to the treatment. Here we discuss pre-clinically well-defined potential mechanisms of action for rituximab and review the ways next generation anti-CD20 monoclonal antibodies can potentially exploit them to further enhance the treatment of B cell malignancies. Although the relative importance of each of these mechanism remains to be established in the clinic, well-designed clinical trials will help to define the efficacy and understanding of which effector activity of modified next generation anti-CD20 mAb will be important in the treatment of B-cell malignancies.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Linfocitos B/efectos de los fármacos , Inmunoterapia , Linfoma de Células B/tratamiento farmacológico , Oncología Médica , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales de Origen Murino , Apoptosis/inmunología , Linfocitos B/inmunología , Ensayos Clínicos como Asunto , Resistencia a Antineoplásicos , Humanos , Linfoma de Células B/inmunología , Rituximab , Transducción de Señal/inmunología
16.
Prostaglandins Other Lipid Mediat ; 88(3-4): 73-81, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19010439

RESUMEN

Prostaglandin E(2) (PGE(2)) plays an important role in the normal physiology of many organ systems. Increased levels of this lipid mediator are associated with many disease states, and it potently regulates inflammatory responses. Three enzymes capable of in vitro synthesis of PGE(2) from the cyclooxygenase metabolite PGH(2) have been described. Here, we examine the contribution of one of these enzymes to PGE(2) production, mPges-2, which encodes microsomal prostaglandin synthase-2 (mPGES-2), by generating mice homozygous for the null allele of this gene. Loss of mPges-2 expression did not result in a measurable decrease in PGE(2) levels in any tissue or cell type examined from healthy mice. Taken together, analysis of the mPGES-2 deficient mouse lines does not substantiate the contention that mPGES-2 is a PGE(2) synthase.


Asunto(s)
Dinoprostona/biosíntesis , Oxidorreductasas Intramoleculares/fisiología , Animales , Northern Blotting , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Interferón gamma/farmacología , Oxidorreductasas Intramoleculares/genética , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Mutantes , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Prostaglandina-E Sintasas
17.
J Pharmacol Exp Ther ; 326(3): 754-63, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18524979

RESUMEN

Microsomal prostaglandin E synthase-1 (mPGES-1) is a terminal prostaglandin E(2) (PGE(2)) synthase in the cyclooxygenase pathway. Inhibitors of mPGES-1 may block PGE(2) production and relieve inflammatory symptoms. To test the hypothesis, we evaluated the antipyretic and analgesic properties of a novel and selective mPGES-1 inhibitor, MF63 [2-(6-chloro-1H-phenanthro-[9,10-d]imidazol-2-yl)isophthalonitrile], in animal models of inflammation. MF63 potently inhibited the human mPGES-1 enzyme (IC(50) = 1.3 nM), with a high degree (>1000-fold) of selectivity over other prostanoid synthases. In rodent species, MF63 strongly inhibited guinea pig mPGES-1 (IC(50) = 0.9 nM) but not the mouse or rat enzyme. When tested in the guinea pig and a knock-in (KI) mouse expressing human mPGES-1, the compound selectively suppressed the synthesis of PGE(2), but not other prostaglandins inhibitable by nonsteroidal anti-inflammatory drugs (NSAIDs), yet retained NSAID-like efficacy at inhibiting lipopolysaccharide-induced pyresis, hyperalgesia, and iodoacetate-induced osteoarthritic pain. In addition, MF63 did not cause NSAID-like gastrointestinal toxic effects, such as mucosal erosions or leakage in the KI mice or nonhuman primates, although it markedly inhibited PGE(2) synthesis in the KI mouse stomach. Our data demonstrate that mPGES-1 inhibition leads to effective relief of both pyresis and inflammatory pain in preclinical models of inflammation and may be a useful approach for treating inflammatory diseases.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Fiebre/enzimología , Imidazoles/farmacología , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Microsomas/enzimología , Dolor/enzimología , Fenantrenos/farmacología , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/uso terapéutico , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Femenino , Fiebre/tratamiento farmacológico , Fiebre/genética , Cobayas , Humanos , Imidazoles/química , Imidazoles/uso terapéutico , Oxidorreductasas Intramoleculares/biosíntesis , Oxidorreductasas Intramoleculares/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microsomas/efectos de los fármacos , Dolor/tratamiento farmacológico , Dolor/genética , Fenantrenos/química , Fenantrenos/uso terapéutico , Antagonistas de Prostaglandina/química , Antagonistas de Prostaglandina/farmacología , Antagonistas de Prostaglandina/uso terapéutico , Prostaglandina-E Sintasas , Ratas , Saimiri
18.
Mol Med ; 14(7-8): 374-82, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18475307

RESUMEN

Multiple lines of evidence suggest that inhibition of Type I Interferons, including IFN-alpha, may provide a therapeutic benefit for autoimmune diseases. Using a chemical genomics approach integrated with cellular and in vivo assays, we screened a small compound library to identify modulators of IFN-alpha biological effects. A genomic fingerprint was developed from both ex vivo patient genomic information and in vitro gene modulation from IFN-alpha cell-based stimulation. A high throughput genomic-based screen then was applied to prioritize 268 small molecule inhibitors targeting 41 different intracellular signaling pathways. Active compounds were profiled further for their ability to inhibit the activation and differentiation of human monocytes using disease-related stimuli. Inhibitors targeting NF-kappaB or Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) signaling emerged as "dissociated inhibitors" because they did not modulate IFN-alpha anti-viral effects against HSV-1 but potently inhibited other immune-related functions. This work describes a novel strategy to identify small molecule inhibitors for the treatment of autoimmune disorders.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Interferón-alfa/antagonistas & inhibidores , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Bibliotecas de Moléculas Pequeñas/análisis , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antivirales/antagonistas & inhibidores , Células Cultivadas , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Femenino , Genoma Humano , Genómica/instrumentación , Genómica/métodos , Humanos , Factores Inmunológicos/antagonistas & inhibidores , Ratones , Ratones Endogámicos , Modelos Biológicos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología
19.
Arthritis Rheum ; 58(5): 1354-65, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18438856

RESUMEN

OBJECTIVE: Prostaglandins (PGs) are found in high levels in the synovial fluid of patients with rheumatoid arthritis, and nonsteroidal blockade of these bioactive lipids plays a role in patient care. The aim of this study was to explore the relative contribution of cyclooxygenase (COX) isoforms and PG species in the autoantibody-driven K/BxN serum-transfer arthritis. METHODS: The prostanoid content of arthritic ankles was assessed in ankle homogenates, and the importance of this pathway was confirmed with pharmacologic blockade. The presence of COX isoforms was assessed by Western blotting and their functional contribution was compared using COX-1-/- and COX-2-/- mice as well as isoform-specific inhibitors. The relative importance of PGE2 and PGI2 (prostacyclin) was determined using mice deficient in microsomal PGE synthase 1 (mPGES-1) and in the receptors for PGI2. RESULTS: High levels of PGE2 and 6-keto-PGF1alpha (a stable metabolite of PGI2) were detected in arthritic joint tissues, correlating strongly with the intensity of synovitis. Pharmacologic inhibition of PG synthesis prevented arthritis and ameliorated active disease. While both COX isoforms were found in inflamed joint tissues, only COX-1 contributed substantially to clinical disease; COX-1-/- mice were fully resistant to disease, whereas COX-2-/- mice remained susceptible. These findings were confirmed by isoform-specific pharmacologic inhibition. Mice lacking mPGES-1 (and therefore PGE2) developed arthritis normally, whereas mice incapable of responding to PGI2 exhibited a significantly attenuated arthritis course, confirming a role of PGI2 in this arthritis model. CONCLUSION: These findings challenge previous paradigms of distinct "housekeeping" versus inflammatory functions of the COX isoforms and highlight the potential pathogenic contribution of prostanoids synthesized via COX-1, in particular PGI2, to inflammatory arthritis.


Asunto(s)
Artritis/enzimología , Artritis/inmunología , Autoanticuerpos/inmunología , Ciclooxigenasa 1/fisiología , Ciclooxigenasa 2/fisiología , Prostaglandinas/fisiología , Animales , Ratones , Ratones Endogámicos C57BL
20.
Circulation ; 117(13): 1701-10, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18347209

RESUMEN

BACKGROUND: Pharmacological inhibition of cyclooxygenase-2 increases the risk of myocardial infarction (MI) and stroke. Microsomal prostaglandin (PG) E(2) synthase-1 (mPGES-1), encoded by the Ptges gene, functions downstream from cyclooxygenase-2 in the inducible PGE(2) biosynthetic pathway. We caused acute MI in Ptges(+/+) and Ptges(-/-) mice to define the role of mPGES-1 in cardiac ischemic injury. METHODS AND RESULTS: Twenty-eight days after MI, Ptges(-/-) mice develop more left ventricular (LV) dilation, have worse LV systolic and diastolic function, and have higher LV end-diastolic pressure than Ptges(+/+) mice but have similar pulmonary wet-to-dry weight ratios, cardiac mass, infarct size, and mortality. The length-to-width ratio of individual cardiomyocytes is significantly greater in Ptges(-/-) than Ptges(+/+) mice after MI, a finding consistent with eccentric cardiomyocyte hypertrophy in Ptges(-/-) mice. Expression of atrial natriuretic peptide, brain natriuretic peptide, and alpha- and beta-myosin heavy chain, markers of ventricular hypertrophy, is higher in the LV of Ptges(-/-) than Ptges(+/+) mice after MI. Ptges(+/+) mice express cyclooxygenase-2 and mPGES-1 protein in inflammatory cells adjacent to the infarct after MI but do not express these proteins in cardiomyocytes. Ptges(-/-) mice express cyclooxygenase-2 in inflammatory cells adjacent to the infarct and do not express mPGES-1 in any cells in the heart. Levels of PGE(2) but not PGD(2), thromboxane A(2), PGI(2), or PGF(2alpha) are higher in the infarct and LV remote from the infarct after MI in Ptges(+/+) than Ptges(-/-) mice. CONCLUSIONS: In Ptges(+/+) mice, mPGES-1 in inflammatory cells catalyzes PGE(2) biosynthesis in the LV after MI. Deletion of mPGES-1 leads to eccentric cardiac myocyte hypertrophy, LV dilation, and impaired LV contractile function after acute MI.


Asunto(s)
Eliminación de Gen , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Microsomas/enzimología , Infarto del Miocardio/enzimología , Infarto del Miocardio/fisiopatología , Remodelación Ventricular/genética , Animales , Oxidorreductasas Intramoleculares/fisiología , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Infarto del Miocardio/genética , Prostaglandina-E Sintasas , Remodelación Ventricular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA