Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Med Chem ; 65(1): 757-784, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34967602

RESUMEN

A diaryl ketone series was identified as vanin-1 inhibitors from a high-throughput screening campaign. While this novel scaffold provided valuable probe 2 that was used to build target confidence, concerns over the ketone moiety led to the replacement of this group. The successful replacement of this moiety was achieved with pyrimidine carboxamides derived from cyclic secondary amines that were extensively characterized using biophysical and crystallographic methods as competitive inhibitors of vanin-1. Through optimization of potency and physicochemical and ADME properties, and guided by co-crystal structures with vanin-1, 3 was identified with a suitable profile for advancement into preclinical development.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Piridinas/síntesis química , Piridinas/farmacología , Animales , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Cristalografía por Rayos X , Sulfato de Dextran , Perros , Descubrimiento de Drogas , Femenino , Proteínas Ligadas a GPI/antagonistas & inhibidores , Ensayos Analíticos de Alto Rendimiento , Cetonas/química , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Piridinas/farmacocinética , Ratas , Relación Estructura-Actividad
2.
J Thromb Haemost ; 18(6): 1278-1290, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32108991

RESUMEN

BACKGROUND: Previous studies have demonstrated that the A1A2A3 domains of von Willebrand factor (VWF) play a key role in regulating macrophage-mediated clearance in vivo. In particular, the A1-domain has been shown to modulate interaction with macrophage low-density lipoprotein receptor-related protein-1 (LRP1) clearance receptor. Furthermore, N-linked glycans within the A2-domain have been shown to protect VWF against premature LRP1-mediated clearance. Importantly, however, the specific regions within A1A2A3 that enable macrophage binding have not been defined. OBJECTIVE AND METHODS: To address this, we utilized site-directed PEGylation and introduced novel targeted N-linked glycosylation within A1A2A3-VWF and subsequently examined VWF clearance. RESULTS: Conjugation with a 40-kDa polyethylene glycol (PEG) moiety significantly extended the half-life of A1A2A3-VWF in VWF-/- mice in a site-specific manner. For example, PEGylation at specific sites within the A1-domain (S1286) and A3-domain (V1803, S1807) attenuated VWF clearance in vivo, compared to wild-type A1A2A3-VWF. Furthermore, PEGylation at these specific sites ablated binding to differentiated THP-1 macrophages and LRP1 cluster II and cluster IV in-vitro. Conversely, PEGylation at other positions (Q1353-A1-domain and M1545-A2-domain) had limited effects on VWF clearance or binding to LRP1.Novel N-linked glycan chains were introduced at N1803 and N1807 in the A3-domain. In contrast to PEGylation at these sites, no significant extension in half-life was observed with these N-glycan variants. CONCLUSIONS: These novel data demonstrate that site specific PEGylation but not site specific N-glycosylation modifies LRP1-dependent uptake of the A1A2A3-VWF by macrophages. This suggests that PEGylation, within the A1- and A3-domains in particular, may be used to attenuate LRP1-mediated clearance of VWF.


Asunto(s)
Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Factor de von Willebrand , Animales , Glicosilación , Cinética , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Polisacáridos , Unión Proteica , Factor de von Willebrand/metabolismo
3.
AAPS J ; 21(3): 52, 2019 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-30976993

RESUMEN

This paper presents a systemic investigation of ADA development and ADA impact of a human coagulation factor in nonclinical species during drug development and provides insights into potential implications in human if a similar ADA occurs. FXaI16L-induced ADA response was characterized in monkey, mouse, rat, and dog in different studies, and ADA effects on pharmacokinetic and/or pharmacodynamics of FXaI16L were further examined in ADA-negative and ADA-positive animals. After repeated administrations, FXaI16L elicited a dose and exposure day-dependent ADA response which ranged from no response to a transient or persistent response. Increase in exposure day and increase in dose generally enhanced ADA incidence except for a decrease in ADA incidence was observed in monkeys after repeated high-dose administrations. The observable ADA impact on pharmacokinetics was only found in some ADA+ animals and included decrease in clearance and increase in systemic exposure but no increase in half-life. In addition, no or limited effect on pharmacodynamics by ADA was observed. The earliest ADA response was observed after three exposure days, marked elevation of drug exposure was observed in some animals at log titer > 2.0, and the highest antibody titer excited was about 4 (Log10) in all species. A correlation between ADA induction and accumulative exposure after various repeat treatments in different species was found for FXaI16L. In addition, potential immunogenicity risk and mitigation of ADA in clinics are discussed.


Asunto(s)
Factor Xa/inmunología , Hemofilia A/tratamiento farmacológico , Animales , Coagulación Sanguínea/efectos de los fármacos , Coagulación Sanguínea/inmunología , Modelos Animales de Enfermedad , Perros , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Factor Xa/administración & dosificación , Factor Xa/genética , Femenino , Semivida , Hemofilia A/sangre , Hemofilia A/diagnóstico , Hemofilia A/inmunología , Humanos , Macaca fascicularis , Masculino , Ratones , Tiempo de Tromboplastina Parcial , Tiempo de Protrombina , Ratas , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Homología de Secuencia de Aminoácido , Especificidad de la Especie
4.
AAPS J ; 21(3): 46, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30927117

RESUMEN

Immunogenicity is a major challenge for protein therapeutics which can potentially reduce drug efficacy and safety and is often being monitored by anti-drug antibody (ADA) and neutralizing antibody (NAb) assays. Circulating targets and residual drugs in matrices can have significant impacts on accuracy of results from ADA and NAb assays, and sufficient drug and target tolerance for these assays are necessary. Here, we report the development of a competitive ligand binding (CLB) NAb assay for an anti-TFPI (tissue factor pathway inhibitor) monoclonal antibody (PF-06741086) with high drug and target tolerance to support ongoing clinical studies. A double acid affinity capture elution approach was used to mitigate drug interference, and a robust target removal strategy was employed to enhance target tolerance. The validated NAb assay has sensitivity of 313 ng/mL, drug tolerance of 50 µg/mL, and target tolerance of 1200 ng/mL. A step-by-step tutorial of assay development is described in this manuscript along with the rationale for using a high drug/target tolerant NAb assay. The NAb assay cut point factor obtained was 0.78. Other assay performance characteristics, e.g., precision and selectivity, are also discussed. This validated method demonstrated a superior drug and target tolerance to warrant specific and precise characterization of the NAb responses in support of ongoing clinical studies.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Neutralizantes/inmunología , Bioensayo/métodos , Desarrollo de Medicamentos/métodos , Lipoproteínas/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados/farmacología , Unión Competitiva , Tolerancia a Medicamentos/inmunología , Humanos , Tolerancia Inmunológica , Inmunoensayo/métodos , Ligandos , Unión Proteica , Proteínas Recombinantes/metabolismo
5.
AAPS J ; 21(1): 4, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30402825

RESUMEN

Insufficient drug tolerance presents a major challenge in the development of neutralizing antibody (NAb) assays for biotherapeutics. Sample pre-treatment using solid-phase extraction with acid dissociation (SPEAD) is widely reported to improve drug tolerance. In this paper, a case study is presented in which SPEAD was used in conjunction with a competitive ligand binding NAb assay format. A significant degree of biotin-drug conjugate leaching was observed resulting in the reporting of both false positive and false negative results in NAb assay. Mitigation steps have been evaluated to address drug/biotin-drug conjugate leaching. These steps included assessment of the streptavidin-coated plate in conjunction with biotin-drug conjugates at various biotin molar challenge ratios (MCR). In addition, an alternative method based on covalent capture of the drug on an aldehyde-activated plate was assessed. Both approaches were compared for the degree of drug/biotin-drug conjugate leaching during the second elution step of the SPEAD procedure. Moreover, the impact of various conditions on the assay performance was assessed, including elution pH, sample incubation time, and biotin MCR. For the covalent drug capture method, capture conditions were evaluated. Optimized conditions in both streptavidin capture and covalent capture methods enabled a significant reduction of drug/biotin-drug conjugate leaching. A streptavidin high binding capacity approach using biotin-drug conjugate with a MCR of 50:1 was chosen as the optimal method yielding a NAb assay with a fit for purpose sensitivity (153 ng/mL) and a drug tolerance of up to 50 µg/mL with 500 ng/mL PC.


Asunto(s)
Anticuerpos Neutralizantes/aislamiento & purificación , Indicadores y Reactivos/química , Pruebas de Neutralización/métodos , Extracción en Fase Sólida/métodos , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Neutralizantes/inmunología , Productos Biológicos/química , Productos Biológicos/inmunología , Productos Biológicos/farmacología , Biotina/análogos & derivados , Biotina/química , Química Farmacéutica , Cromatografía de Afinidad/métodos , Tolerancia a Medicamentos , Reacciones Falso Negativas , Reacciones Falso Positivas , Humanos , Estreptavidina/química , Succinimidas/química
6.
J Pharm Sci ; 107(7): 1995-2004, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29571739

RESUMEN

Tissue factor pathway inhibitor (TFPI) exhibits multiple isoforms, which are known to present in multiple locations such as plasma, endothelium, and platelets. TFPI is an endogenous negative modulator of the coagulation pathway, and therefore, neutralization of TFPI function can potentially increase coagulation activity. A human monoclonal antibody, PF-06741086, which interacts with all isoforms of TFPI is currently being tested in clinic for treating hemophilia patients with and without inhibitors. To support clinical development of PF-06741086, pharmacokinetics (PK) and pharmacodynamics of PF-06741086 were characterized in monkeys. In addition, a mechanistic model approach was used to estimate PK parameters in monkeys and simulate PK profiles in human. The results show that PF-06741086 exhibited target-mediated drug disposition and had specific effects on various hemostatic markers including diluted prothrombin time, thrombin generation, and thrombin-antithrombin complex in monkeys after administration. The model-predicted and observed human exposures were compared retrospectively, and the result indicates that the exposure prediction was reasonable within less than 2-fold deviation. This study demonstrated in vivo efficacy of PF-06741086 in monkeys and the utility of a rational mechanistic approach to describe PK for a monoclonal antibody with complex target binding.


Asunto(s)
Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/farmacología , Coagulación Sanguínea/efectos de los fármacos , Hemostáticos/sangre , Hemostáticos/farmacología , Lipoproteínas/antagonistas & inhibidores , Animales , Humanos , Lipoproteínas/metabolismo , Macaca fascicularis , Masculino , Modelos Biológicos
7.
J Pharmacol Toxicol Methods ; 55(1): 103-12, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-16769228

RESUMEN

INTRODUCTION: Transparency is a unique attribute of zebrafish that permits direct assessment of drug effects on the nervous system using whole mount antibody immunostaining and histochemistry. METHODS: To assess pharmacological effects of drugs on the optic nerves, motor neurons, and dopaminergic neurons, we performed whole mount immunostaining and visualized different neuronal cell types in vivo. In addition, we assessed neuronal apoptosis, proliferation, oxidation and the integrity of the myelin sheath using TUNEL staining, immunostaining and in situ hybridization. The number of dopaminergic neurons was examined and morphometric analysis was performed to quantify the staining signals for myelin basic protein and apoptosis. RESULTS: We showed that compounds that induce neurotoxicity in humans caused similar neurotoxicity in zebrafish. For example, ethanol induced defects in optic nerves and motor neurons and affected neuronal proliferation; 6-hydroxydopamine caused neuronal oxidation and dopaminergic neuron loss; acrylamide induced demyelination; taxol, neomycin, TCDD and retinoic acid induced neuronal apoptosis. DISCUSSION: Effects of drug treatment on different neurons can easily be visually assessed and quantified in intact animals. These results support the use of zebrafish as a predictive model for assessing neurotoxicity.


Asunto(s)
Neurotoxinas/toxicidad , Pruebas de Toxicidad/métodos , Acrilamida/toxicidad , Animales , Antracenos/toxicidad , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión no Mamífero/efectos de los fármacos , Etanol/toxicidad , Glutaratos/toxicidad , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Neuronas Motoras/efectos de los fármacos , Vaina de Mielina/efectos de los fármacos , Neomicina/toxicidad , Neuronas/citología , Neuronas/efectos de los fármacos , Nervio Óptico/efectos de los fármacos , Oxidopamina/toxicidad , Paclitaxel/toxicidad , Dibenzodioxinas Policloradas/toxicidad , Tretinoina/toxicidad , Pez Cebra
8.
J Pharm Sci ; 106(8): 2136-2143, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28389265

RESUMEN

FXaI16L is a recombinant human FXa variant which is currently being evaluated in the clinic for treating intracerebral hemorrhage. The aim of our studies is to investigate overall pharmacokinetics, pharmacodynamics, and distribution of FXaI16L in preclinical species, and to understand its potential implication in human. Pharmacokinetics of FXaI16L was examined using active site probes and the results showed that FXaI16L displayed fast clearance, low volume of distribution, and a very short plasma resident time in mice, rats, and monkeys. When pharmacodynamics was examined in monkeys, concentration effects of FXaI16L on shortening of active partial prothrombin time and formation of thrombin-antithrombin complex were observed. Furthermore, biodistribution study was conducted in mice using radiolabeled FXaI16L, and showed that 125I-FXaI16L has high plasma protein binding and significant liver and kidney distribution. Human pharmacokinetic prediction for first-in-human dosing was evaluated using allometric scaling, liver blood flow, and a fixed coefficient method, and single species allometric scaling using monkey data was most predictive for human pharmacokinetics of FXaI16L.


Asunto(s)
Factor Xa/farmacología , Factor Xa/farmacocinética , Animales , Coagulación Sanguínea/efectos de los fármacos , Proteínas Sanguíneas/metabolismo , Factor Xa/metabolismo , Humanos , Macaca fascicularis , Masculino , Ratones , Modelos Biológicos , Unión Proteica , Ratas , Ratas Wistar , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología , Distribución Tisular
9.
Neurotoxicol Teratol ; 28(4): 509-16, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16814516

RESUMEN

In this study, we developed an in vivo method to determine drug effects on oxidation-induced apoptosis in the zebrafish brain caused by treatment with L-hydroxyglutaric acid (LGA). We confirmed that LGA-induced apoptosis was caused by oxidation by examining the presence of an oxidative product, nitrotyrosine. Next, we examined the effects of 14 characterized neuroprotectants on LGA-treated zebrafish, including: D-methionine (D-Met), Indole-3-carbinol, deferoxamine (DFO), dihydroxybenzoate (DHB), deprenyl, L-NAME (N(G)-nitro-L-arginine methyl ester), n-acetyl L-cysteine (L-NAC), 2-oxothiazolidine-4-carboxylate (OTC), lipoic acid, minocycline, isatin, cortisone, ascorbic acid and alpha-tocopherol. Eleven of 14 neuroprotectants and 7 of 7 synthetic anti-oxidants exhibit significant protection in zebrafish. Buthionine sulfoximine (BSO), used as a negative control, exhibited no significant protective effects. In addition, three blood-brain barrier (BBB) impermeable compounds exhibited no significant effects. Our results in zebrafish were similar to results reported in mammals supporting the utility of this in vivo method for identifying potential neuroprotective anti-oxidants.


Asunto(s)
Fármacos Neuroprotectores/farmacología , Pez Cebra/fisiología , Acridinas/toxicidad , Animales , Anticuerpos , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Biomarcadores , Encéfalo/citología , Encéfalo/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Glutaratos/farmacología , Interpretación de Imagen Asistida por Computador , Microscopía Fluorescente , Óxido Nítrico/biosíntesis , Oxidación-Reducción , Tirosina/análogos & derivados , Tirosina/inmunología
10.
Curr Opin Drug Discov Devel ; 8(1): 100-6, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15679177

RESUMEN

Toxicity, due to complications of in vivo adsorption, distribution, metabolism and excretion (ADME), is a major cause of failure during drug development; many drugs shown to be safe in cell culture prove toxic in animal studies. Effective in vivo toxicity screening early in the development process can reduce the number of compounds that progress to laborious and costly late-stage animal testing. The transparent zebrafish provides accessibility to internal organs, tissues and even cells, and has emerged as an invaluable model organism for toxicity testing and drug discovery. Straightforward in vivo zebrafish assays can serve as an intermediate step between cell-based and mammalian testing, thus streamlining the drug development time-line.


Asunto(s)
Toxicología/métodos , Pez Cebra/fisiología , Animales , Carcinógenos/toxicidad , Enfermedades Cardiovasculares/inducido químicamente , Sistema Inmunológico/efectos de los fármacos , Enfermedades del Laberinto/inducido químicamente , Síndromes de Neurotoxicidad/patología , Toxicogenética , Xenobióticos/metabolismo
11.
Hear Res ; 208(1-2): 79-88, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16014323

RESUMEN

Zebrafish and other fish exhibit hair cells in the lateral-line neuromasts which are structurally and functionally similar to mammalian inner ear hair cells. To facilitate drug screening for ototoxic or otoprotective agents, we report a straightforward, quantitative in vivo assay to determine potential ototoxicity of drug candidates and to screen otoprotective agents in zebrafish larva. In this study, a fluorescent vital dye, DASPEI (2-(4-(dimethylamino)styryl)-N-ethylpyridinium iodide), was used to stain zebrafish hair cells in vivo and morphometric analysis was performed to quantify fluorescence intensity and convert images to numerical endpoints. Various therapeutics, including gentamicin, cisplatin, vinblastine sulfate, quinine, and neomycin, which cause ototoxicity in humans, also resulted in hair cell loss in zebrafish. In addition, protection against cisplatin-induced ototoxicity was observed in zebrafish larva co-treated with cisplatin and different antioxidants including, glutathione (GSH), allopurinol (ALO), N-acetyl l-cysteine (l-NAC), 2-oxothiazolidine-4-carboxylate (OTC) and d-methionine (d-MET). Our data indicate that results of ototoxicity and otoprotection in zebrafish correlated with results in humans, supporting use of zebrafish for preliminary drug screening.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Oído Interno/efectos de los fármacos , Pez Cebra/anatomía & histología , Animales , Antibacterianos/toxicidad , Antineoplásicos/toxicidad , Antioxidantes/farmacología , Cisplatino/toxicidad , Oído Interno/patología , Colorantes Fluorescentes , Gentamicinas/toxicidad , Glutatión/farmacología , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/patología , Humanos , Larva/efectos de los fármacos , Modelos Animales , Compuestos de Piridinio , Especificidad de la Especie
12.
Assay Drug Dev Technol ; 1(1 Pt 1): 41-8, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15090155

RESUMEN

The zebrafish embryo has become an important vertebrate model for assessing drug effects. It is well suited for studies in genetics, embryology, development, and cell biology. Zebrafish embryos exhibit unique characteristics, including ease of maintenance and drug administration, short reproductive cycle, and transparency that permits visual assessment of developing cells and organs. Because of these advantages, zebrafish bioassays are cheaper and faster than mouse assays, and are suitable for large-scale drug screening. Here we describe the use of zebrafish bioassays for assessing toxicity, angiogenesis, and apoptosis. Using 18 chemicals, we demonstrated that toxic response, teratogenic effects, and LC(50) in zebrafish are comparable to results in mice. The effects of compounds on various organs, including the heart, brain, intestine, pancreas, cartilage, liver, and kidney, were observed in the transparent animals without complicated processing, demonstrating the efficiency of toxicity assays using zebrafish embryos. Using endogenous alkaline phosphatase staining and a whole-animal enzyme assay, we demonstrated that SU5416 and flavopiridol, compounds shown to have antiangiogenic effects in mammals, inhibit blood vessel growth in zebrafish, and this bioassay is suitable for high-throughput screening using a 96-well microplate reader. We also demonstrated that in vivo acridine orange staining can be used to visualize apoptotic events in embryos treated with brefeldin A, neomycin, or caspase inhibitors. After in vivo staining, acridine orange can be extracted and quantitated using a fluorescence microplate reader, providing a screening system for agents that modulate apoptosis.


Asunto(s)
Evaluación Preclínica de Medicamentos , Pez Cebra/fisiología , Naranja de Acridina , Fosfatasa Alcalina/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Apoptosis/efectos de los fármacos , Bioensayo , Embrión no Mamífero , Flavonoides/farmacología , Colorantes Fluorescentes , Indoles/farmacología , Dosificación Letal Mediana , Ratones , Piperidinas/farmacología , Pirroles/farmacología , Pruebas de Toxicidad , Pez Cebra/genética
13.
AAPS J ; 15(4): 1141-54, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23990500

RESUMEN

A mathematical pharmacokinetic/anti-drug-antibody (PK/ADA) model was constructed for quantitatively assessing immunogenicity for therapeutic proteins. The model is inspired by traditional pharmacokinetic/pharmacodynamic (PK/PD) models, and is based on the observed impact of ADA on protein drug clearance. The hypothesis for this work is that altered drug PK contains information about the extent and timing of ADA generation. By fitting drug PK profiles while accounting for ADA-mediated drug clearance, the model provides an approach to characterize ADA generation during the study, including the maximum ADA response, sensitivity of ADA response to drug dose level, affinity maturation rate, time lag to observe an ADA response, and the elimination rate for ADA-drug complex. The model also provides a mean to estimate putative concentration-time profiles for ADA, ADA-drug complex, and ADA binding affinity-time profile. When simulating ADA responses to various drug dose levels, bell-shaped dose-response curves were generated. The model contains simultaneous quantitative modeling and provides estimation of the characteristics of therapeutic protein drug PK and ADA responses in vivo. With further experimental validation, the model may be applied to the simulation of ADA response to therapeutic protein drugs in silico, or be applied in subsequent PK/PD models.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Fenómenos Inmunogenéticos/efectos de los fármacos , Fenómenos Inmunogenéticos/fisiología , Modelos Teóricos , Animales , Anticuerpos Monoclonales/inmunología , Relación Dosis-Respuesta Inmunológica , Macaca fascicularis , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología
14.
Autoimmunity ; 43(8): 628-39, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20402568

RESUMEN

SHG2210, a fusion protein containing the N-terminus of human nicotinic acetylcholine receptor α (AchR-α; aa1-210) and human transferrin (TF), was characterized as a potential therapeutic for myasthenia gravis (MG) caused predominately by α subunit autoantibodies. SHG2210 was shown to be able to bind to α subunit autoantibodies and the TF receptor (TFR). SHG2210 and SHG2210-anti-AchR antibody complex are internalized through TFR-mediated endocytosis. The SHG2210 and SHG2210-anti-AchR antibody complex is present in Lamp1-positive lysosomal compartments after internalization; however, neither SHG2210 nor SHG2210-antibody complex is present in Rab11-positive recycling endosomes. SHG2210 bound to α subunit of AChR autoantibodies may be cleared by the lysosome, resulting in short cellular half-life relative to SHG2210. SHG2210 is shown to have a protective effect on antigenic modulation of the AChR induced by serum from select patients with MG, suggesting that a fusion protein approach may be an effective therapeutic for treating MG.


Asunto(s)
Miastenia Gravis/inmunología , Receptores Nicotínicos/inmunología , Receptores de Transferrina/inmunología , Proteínas Recombinantes/farmacología , Transferrina/inmunología , Unión Competitiva/inmunología , Células HeLa , Humanos , Microscopía Confocal , Miastenia Gravis/tratamiento farmacológico , Receptores Nicotínicos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapéutico , Transferrina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA