Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
CPT Pharmacometrics Syst Pharmacol ; 9(6): 342-352, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32419339

RESUMEN

Quantitative understanding about the dynamics of drug-target interactions in biological systems is essential, especially in rare disease programs with small patient populations. Follistatin, by antagonism of myostatin and activin, which are negative regulators of skeletal muscle and inflammatory response, is a promising therapeutic target for Duchenne Muscular Dystrophy. In this study, we constructed a quantitative systems pharmacology model for FS-EEE-Fc, a follistatin recombinant protein to investigate its efficacy from dual target binding, and, subsequently, to project its human efficacious dose. Based on model simulations, with an assumed efficacy threshold of 7-10% muscle volume increase, 3-5 mg/kg weekly dosing of FS-EEE-Fc is predicted to achieve meaningful clinical outcome. In conclusion, the study demonstrated an application of mechanism driven approach at early stage of a rare disease drug development to support lead compound optimization, enable human dose, pharmacokinetics, and efficacy predictions.


Asunto(s)
Folistatina/farmacocinética , Modelos Biológicos , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne/tratamiento farmacológico , Fármacos Neuromusculares/farmacocinética , Biología de Sistemas , Receptores de Activinas Tipo II/antagonistas & inhibidores , Receptores de Activinas Tipo II/metabolismo , Animales , Cálculo de Dosificación de Drogas , Folistatina/administración & dosificación , Humanos , Ligandos , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatología , Miostatina/antagonistas & inhibidores , Miostatina/metabolismo , Fármacos Neuromusculares/administración & dosificación , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/farmacocinética , Transducción de Señal
2.
J Pharmacol Exp Ther ; 366(2): 291-302, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29752426

RESUMEN

Follistatin (FS) is an important regulatory protein, a natural antagonist for transforming growth factor-ß family members activin and myostatin. The diverse biologic roles of the activin and myostatin signaling pathways make FS a promising therapeutic target for treating human diseases exhibiting inflammation, fibrosis, and muscle disorders, such as Duchenne muscular dystrophy. However, rapid heparin-mediated hepatic clearance of FS limits its therapeutic potential. We targeted the heparin-binding loop of FS for site-directed mutagenesis to improve clearance parameters. By generating a series of FS variants with one, two, or three negative amino acid substitutions, we demonstrated a direct and proportional relationship between the degree of heparin-binding affinity in vitro and the exposure in vivo. The triple mutation K(76,81,82)E abolished heparin-binding affinity, resulting in ∼20-fold improved in vivo exposure. This triple mutant retains full functional activity and an antibody-like pharmacokinetic profile, and shows a superior developability profile in physical stability and cell productivity compared with FS variants, which substitute the entire heparin-binding loop with alternative sequences. Our surgical approach to mutagenesis should also reduce the immunogenicity risk. To further lower this risk, we introduced a novel glycosylation site into the heparin-binding loop. This hyperglycosylated variant showed a 10-fold improved exposure and decreased clearance in mice compared with an IgG1 Fc fusion protein containing the native FS sequence. Collectively, our data highlight the importance of improving pharmacokinetic properties by manipulating heparin-binding affinity and glycosylation content and provide a valuable guideline to design desirable therapeutic FS molecules.


Asunto(s)
Folistatina/genética , Folistatina/farmacocinética , Ingeniería de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacocinética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Femenino , Folistatina/metabolismo , Folistatina/uso terapéutico , Glicosilación , Heparina/metabolismo , Humanos , Ratones , Mutación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Distribución Tisular
3.
Reprod Domest Anim ; 53(1): 3-10, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29134682

RESUMEN

Several oocyte-derived genes/proteins are essential to early embryonic development. The expression and stability of these proteins are influenced by the autocrine/paracrine activity of factors released by oocytes and cumulus cells. This study investigated the paracrine and autocrine activity of follistatin (FS), which is secreted by oocytes and cumulus cells as part of porcine embryogenesis. Immunohistochemical (IHC) localization of follistatin was conducted on 100 randomly selected early- and late-cleaving two-cell embryos. Dissociated cumulus cells were treated with various doses of follistatin for determination of the follistatin gene (FST) mRNA expression levels by quantitative real-time PCR analysis. Microinjection of siRNA induced a downregulation of FST mRNA during embryonic development, thereby decreasing the proportion embryos developing to the blastocyst stage (19.33%). Immunolocalization analysis showed enhanced staining for follistatin in early-cleavage stage embryos. Quantitative real-time PCR indicated a significantly lower FST transcript level in cumulus cells after application of the highest dose of follistatin (100 ng/ml). Exogenous follistatin treatment of in vitro maturation embryos resulted in statistically significant dose-dependent changes during development. Application of the highest concentration (100 ng/ml) of follistatin decreased the maturation rate of the oocytes. On the other hand, the application of 10 ng/ml follistatin resulted in an increase in the number of embryos. The observed differential effect of exogenous follistatin might be due to maternal FST and autocrine/paracrine factors secreted by cumulus cells.


Asunto(s)
Células del Cúmulo/efectos de los fármacos , Folistatina/farmacocinética , Oocitos/efectos de los fármacos , Porcinos/embriología , Animales , Comunicación Autocrina , Blastocisto , Células del Cúmulo/metabolismo , Desarrollo Embrionario/efectos de los fármacos , Femenino , Folistatina/genética , Expresión Génica , Silenciador del Gen , Técnicas de Maduración In Vitro de los Oocitos/veterinaria , Oocitos/metabolismo , Comunicación Paracrina , ARN Mensajero , Reacción en Cadena en Tiempo Real de la Polimerasa , Porcinos/genética , Porcinos/metabolismo
4.
Drug Metab Dispos ; 43(12): 1882-90, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26354950

RESUMEN

Follistatin 315 heparan sulfate-binding deficient mutant human IgG4 Fc fusion (FST-ΔHBS-Fc) is a follistatin (FST) based Fc fusion protein currently being developed as a novel therapy for several potential indications, including muscle wasting. Previous assessments of the pharmacokinetics and therapeutic activity of FST-ΔHBS-Fc have shown a close association of the exposure-response relationship. The current work builds upon these initial studies by investigating the glycosylation characteristics of FST-ΔHBS-Fc after recombinant expression and its impact on the pharmacokinetics in mice and Cynomolgus monkeys. The data presented indicate that FST-ΔHBS-Fc is heterogeneously glycosylated at the three putative sites in FST when recombinantly expressed in stably transfected Chinese hamster ovary cells. Such carbohydrate heterogeneity, especially with regards to sialic acid incorporation, directly results in sugar-dependent clearance in both mice and Cynomolgus monkeys. Examination of the pharmacokinetics of FST-ΔHBS-Fc molecules containing variable sialic acid content in asialoglycoprotein receptor 1 (ASPGR-1) knockout mice supports the receptor's role as part of the clearance mechanism of the molecules. Based on the evaluation of several variably sialylated lots of material in pharmacokinetic assessments, we define specifications for average sialic acid incorporation into FST-ΔHBS-Fc that result in limited sugar-mediated clearance. Taken together, these studies highlight the importance of establishing an early understanding of the glycosylation/pharmacokinetic relationships of FST-ΔHBS-Fc, which will provide a basis for future application toward optimal systemic drug delivery and dosing strategies.


Asunto(s)
Terapia Biológica/tendencias , Folistatina/química , Folistatina/farmacocinética , Animales , Células CHO , Cricetinae , Cricetulus , Glicosilación , Células HEK293 , Humanos , Macaca fascicularis , Masculino , Ratones , Ratones Noqueados , Ratones SCID
5.
J Pharmacol Exp Ther ; 344(3): 616-23, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23249626

RESUMEN

Human follistatin is a regulatory glycoprotein with widespread biologic functions, including antiinflammatory activities, wound-healing properties, and muscle-stimulating effects. The role of follistatin in a wide range of biologic activities shows promise for potential clinical application, which has prompted considerable interest in the investigation of the protein as a potential disease-modifying agent. In spite of this potential, the development of follistatin as a broad use biotherapeutic has been severely hindered by a poor understanding and characterization of its pharmacokinetic/pharmacodynamic (PK/PD) relationships. Therefore, to better define these relationships, we performed in-depth analyses of the PK/PD relationships of native follistatin-315 (FST315). Our data indicate that the intrinsic PK/PD properties of native FST315 are poorly suited for acting as a parentally administered biotherapeutic with broad systemic effects. Here, we leveraged protein engineering to modify the PK characteristics of the native molecule by fusing FST315 to a murine IgG(1) Fc and removing the intrinsic heparan sulfate-binding activity of follistatin. The engineered variant molecule had ~100- and ~1600-fold improvements in terminal half-life and exposure, respectively. In contrast to the native FST315, the variant showed a robust, dose-dependent pharmacological effect when administered subcutaneously on a weekly basis in mouse models of muscle atrophy and degeneration. These studies highlight the underappreciated and critical relationship between optimizing multiple physical and chemical properties of follistatin on its overall PK/PD profile. Moreover, our findings provide the first documented strategy toward the development of a follistatin therapeutic with potential use in patients affected with skeletal muscle diseases.


Asunto(s)
Folistatina/farmacología , Folistatina/farmacocinética , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/farmacocinética , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Folistatina/genética , Células HEK293 , Semivida , Heparina/metabolismo , Humanos , Inmunoglobulina G/metabolismo , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Atrofia Muscular/tratamiento farmacológico , Atrofia Muscular/metabolismo , Unión Proteica , Ingeniería de Proteínas/métodos , Proteínas Recombinantes/genética , Sefarosa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...