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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Drug Metab Dispos ; 50(7): 898-908, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35545256

RESUMEN

Fragment crystallizable (Fc) fusion is commonly used for extending the half-life of biotherapeutics such as cytokines. In this work, we studied the pharmacokinetics of Fc-fused interleukin-10 (IL-10) proteins that exhibited potent antitumor activity in mouse syngeneic tumor models. At pharmacologically active doses of ≥0.1 mg/kg, both mouse Fc-mouse IL-10 and human Fc-human IL-10, constructed as the C terminus of the Fc domain fused with IL-10 via a glycine-serine polypeptide linker, exhibited nonlinear pharmacokinetics after intravenous administration to mice at the doses of 0.05, 0.5, and 5 mg/kg. With a nominal dose ratio of 1:10:100; the ratio of the area under the curve for mouse Fc-mouse IL-10 and human Fc-human IL-10 was 1:181:1830 and 1:75:633, respectively. In contrast, recombinant mouse or human IL-10 proteins exhibited linear pharmacokinetics in mice. Compartmental analysis, using the Michaelis-Menten equation with the in vitro IL-10 receptor alpha binding affinity inputted as the Km, unified the pharmacokinetic data across the dose range. Additionally, nontarget-mediated clearance estimated for fusion proteins was ∼200-fold slower than that for cytokines, causing the manifestation of target-mediated drug disposition (TMDD) in the fusion protein pharmacokinetics. The experimental data generated with a mouse IL-10 receptor alpha-blocking antibody and a human Fc-human IL-10 mutant with a reduced receptor binding affinity showed significant improvements in pharmacokinetics, supporting TMDD as the cause of nonlinearity. Target expression and its effect on pharmacokinetics must be determined when considering using Fc as a half-life extension strategy, and pharmacokinetic evaluations need to be performed at a range of doses covering pharmacological activity. SIGNIFICANCE STATEMENT: Target-mediated drug disposition can manifest to affect the pharmacokinetics of a fragment crystallizable (Fc)-fused cytokine when the nontarget-mediated clearance of the cytokine is decreased due to neonatal Fc receptor-mediated recycling and molecular weight increases that reduce the renal clearance. The phenomenon was demonstrated with interleukin-10 Fc-fusion proteins in mice at pharmacologically active doses. Future drug designs using Fc as a half-life extension approach for cytokines need to consider target expression and its effect on pharmacokinetics at relevant doses.


Asunto(s)
Interleucina-10 , Animales , Semivida , Humanos , Interleucina-10/farmacocinética , Ratones , Receptores de Interleucina-10 , Proteínas Recombinantes de Fusión/farmacocinética
2.
Lancet Oncol ; 20(11): 1544-1555, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31563517

RESUMEN

BACKGROUND: IL-10 has anti-inflammatory and CD8+ T-cell stimulating activities. Pegilodecakin (pegylated IL-10) is a first-in-class, long-acting IL-10 receptor agonist that induces oligoclonal T-cell expansion and has single-agent activity in advanced solid tumours. We assessed the safety and activity of pegilodecakin with anti-PD-1 monoclonal antibody inhibitors in patients with advanced solid tumours. METHODS: We did a multicentre, multicohort, open-label, phase 1b trial (IVY) at 12 cancer research centres in the USA. Patients were assigned sequentially into cohorts. Here, we report on all enrolled patients from two cohorts treated with pegilodecakin combined with anti-PD-1 inhibitors. Eligible patients were aged at least 18 years with histologically or cytologically confirmed advanced malignant solid tumours refractory to previous therapies, and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients with uncontrolled infectious diseases were excluded. Pegilodecakin was provided in single-use 3 mL vials and was self-administered subcutaneously by injection at home at 10 µg/kg or 20 µg/kg once per day in combination with pembrolizumab (2 mg/kg every 3 weeks or 200 mg every 3 weeks) or nivolumab (3 mg/kg every 2 weeks or 240 mg every 2 weeks or 480 mg every 4 weeks at the approved dosing), both of which were given intravenously at the study site. Patients received pembrolizumab or nivolumab with pegilodecakin until disease progression, toxicity necessitating treatment discontinuation, patient withdrawal of consent, or study end. The primary endpoints were safety and tolerability, assessed in all patients enrolled in the study who received any amount of study medication including at least one dose of pegilodecakin, and pharmacokinetics (previously published). Secondary endpoints included objective response by immune-related response criteria in all patients who were treated and had evaluable measurements. The study is active but no longer recruiting, and is registered with ClinicalTrials.gov, NCT02009449. FINDINGS: Between Feb 13, 2015, and Sept 12, 2017, 111 patients were enrolled in the two cohorts. 53 received pegilodecakin plus pembrolizumab, and 58 received pegilodecakin plus nivolumab. 34 (31%) of 111 patients had non-small-cell lung cancer, 37 (33%) had melanoma, and 38 (34%) had renal cell carcinoma; one (<1%) patient had triple-negative breast cancer and one (<1%) had bladder cancer. Data cutoff was July 1, 2018. Median follow-up was 26·9 months (IQR 22·3-31·5) for patients with non-small-cell lung cancer, 33·0 months (29·2-35·1) for those with melanoma, and 22·7 months (20·9-27·0) for those with renal cell carcinoma. At least one treatment-related adverse event occurred in 103 (93%) of 111 patients. Grade 3 or 4 events occurred in 73 (66%) of 111 patients (35 [66%] of 53 in the pembrolizumab group and 38 [66%] of 58 in the nivolumab group), the most common of which were anaemia (12 [23%] in the pembrolizumab group and 16 [28%] in the nivolumab group), thrombocytopenia (14 [26%] in the pembrolizumab group and 12 [21%] in the nivolumab group), fatigue (11 [21%] in the pembrolizumab group and 6 [10%] in the nivolumab group) and hypertriglyceridaemia (three [6%] in the pembrolizumab group and eight [14%] in the nivolumab group). There were no fatal adverse events determined to be related to the study treatments. Of the patients evaluable for response, objective responses were 12 (43%) of 28 (non-small-cell lung cancer), three (10%) of 31 (melanoma), and 14 (40%) of 35 (renal cell carcinoma). INTERPRETATION: In this patient population, pegilodecakin with anti-PD-1 monoclonal antibodies had a manageable toxicity profile and preliminary antitumour activity. Pegilodecakin with pembrolizumab or nivolumab could provide a new therapeutic opportunity for previously treated patients with renal cell carcinoma and non-small-cell carcinoma. FUNDING: ARMO BioSciences, a wholly owned subsidiary of Eli Lilly and Company.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Antineoplásicos Inmunológicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Interleucina-10/administración & dosificación , Neoplasias/tratamiento farmacológico , Nivolumab/administración & dosificación , Polietilenglicoles/administración & dosificación , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Anciano , Anticuerpos Monoclonales Humanizados/efectos adversos , Antineoplásicos Inmunológicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Femenino , Humanos , Interleucina-10/efectos adversos , Interleucina-10/farmacocinética , Masculino , Persona de Mediana Edad , Neoplasias/inmunología , Neoplasias/patología , Nivolumab/efectos adversos , Polietilenglicoles/efectos adversos , Polietilenglicoles/farmacocinética , Receptor de Muerte Celular Programada 1/inmunología , Estados Unidos
3.
Nano Lett ; 14(11): 6449-55, 2014 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-25333768

RESUMEN

Protein therapeutics have gained attention recently for treatment of a myriad of human diseases due to their high potency and unique mechanisms of action. We present the development of a novel polymeric thermosponge nanoparticle for efficient delivery of labile proteins using a solvent-free polymer thermo-expansion mechanism with clinical potential, capable of effectively delivering a range of therapeutic proteins in a sustained manner with no loss of bioactivity, with improved biological half-lives and efficacy in vivo.


Asunto(s)
Antiinflamatorios/administración & dosificación , Preparaciones de Acción Retardada/química , Hipoglucemiantes/administración & dosificación , Insulina/administración & dosificación , Interleucina-10/administración & dosificación , Nanopartículas/química , Polímeros/química , Animales , Antiinflamatorios/farmacocinética , Antiinflamatorios/farmacología , Línea Celular , Sistemas de Liberación de Medicamentos , Humanos , Hipoglucemiantes/farmacocinética , Hipoglucemiantes/farmacología , Insulina/farmacocinética , Insulina/farmacología , Interleucina-10/farmacocinética , Interleucina-10/farmacología , Ratones , Nanopartículas/ultraestructura , Temperatura
4.
Drug Metab Dispos ; 40(2): 360-73, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22083830

RESUMEN

Interleukin 10 (IL-10) is a potent cytokine homodimer with multiple immunoregulatory functions. Here, we have characterized the effects of PEGylation and formation of human IL-10 (hIL-10)/humanized anti-human IL-10 (hαhIL-10) immune complexes in the pharmacokinetics, biodistribution, and biotransformation of IL-10 in mice. To assess the fate of native, PEGylated, and antibody-bound IL-10; we implemented an analytical set of fluorescence emission-linked assays. Plasma size exclusion chromatography analysis indicated that fluoro-labeled native and PEGylated murine IL-10 (PEG-mIL-10) are stable in the circulation. PEGylation of IL-10 resulted in a 21-fold increased exposure, 2.7-fold increase in half-life, and 20-fold reduction in clearance. Kidney is the major organ of disposition for both native and PEGylated mIL-10 with renal uptake directly related to systemic clearance. The fluorescence signal in the kidneys reached tissue/blood ratios up to 150 and 20 for native and PEG-mIL-10, respectively. hIL-10/hαhIL-10 immune complexes are detectable in the circulation without evidence of unbound or degraded hIL-10. The exposure of hIL-10 present in immune complexes versus that of hIL-10 alone increased from 0.53 to 11.28 µg · day/ml, with a half-life of 1.16 days and a 23-fold reduction in clearance. Unlike hIL-10 alone, antibody-bound hIL-10 was targeted mainly to the liver with minimal renal distribution. In addition, we found an 11-fold reduction (from 9.9 to 113 nM) in binding to the neonatal Fc receptor (FcRn) when the hαhIL10 antibody is conjugated to hIL-10. The potential changes in FcRn binding in vivo and increased liver uptake may explain the unique pharmacokinetic properties of hIL-10/hαhIL-10 immune complexes.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Reacciones Antígeno-Anticuerpo , Interleucina-10/química , Interleucina-10/farmacocinética , Polietilenglicoles/química , Animales , Antiinflamatorios no Esteroideos/antagonistas & inhibidores , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/metabolismo , Complejo Antígeno-Anticuerpo/análisis , Femenino , Semivida , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inyecciones Intravenosas , Inyecciones Subcutáneas , Interleucina-10/antagonistas & inhibidores , Interleucina-10/metabolismo , Riñón/citología , Riñón/inmunología , Riñón/metabolismo , Hígado/citología , Hígado/inmunología , Hígado/metabolismo , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos , Receptores Fc/metabolismo , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacocinética , Distribución Tisular
5.
Protein Expr Purif ; 83(2): 152-6, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22484198

RESUMEN

Interleukin (IL)-10 is an anti-inflammatory cytokine that could be potentially applied for clinical therapy. However, its short circulating half-life in the serum limits its clinical applications. In this study, we designed a fusion protein containing human IL-10 and an IgG Fc fragment (hIL-10/Fc), and expressed it in Pichia pastoris. This hIL-10/Fc fusion protein was purified from the culture supernatant using MabSelect affinity chromatography and size-exclusion chromatography. The hIL-10/Fc yield was about 5mg/L in shake flasks, with purity exceeding 95%. In addition, the hIL-10/Fc fusion protein suppressed the phytohemagglutinin-induced IFN-γ production in human peripheral blood mononuclear cells. Pharmacokinetic study also revealed that hIL-10/Fc has a prolonged circulating half-life of about 30h in rats. More importantly, the hIL-10/Fc fusion protein displayed highly specific biological activity, which was slightly higher than that of the commercial recombinant human IL-10 (rhIL-10). Therefore, P. pastoris is useful in the large-scale production of hIL-10/Fc fusion protein for both research and therapeutic applications.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas/aislamiento & purificación , Interleucina-10/aislamiento & purificación , Proteínas Recombinantes de Fusión/aislamiento & purificación , Animales , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/genética , Interferón gamma/análisis , Interferón gamma/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-10/farmacocinética , Leucocitos Mononucleares/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacocinética
6.
Biotechnol Bioeng ; 108(8): 1977-86, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21391205

RESUMEN

Interleukin-10 (IL-10) is an anti-inflammatory cytokine, which active form is a non-covalent homodimer. Given the potential of IL-10 for application in various medical conditions, it is essential to develop systems for its effective delivery. In previous work, it has been shown that a dextrin nanogel effectively incorporated and stabilized rIL-10, enabling its release over time. In this work, the delivery system based on dextrin nanogels was further analyzed. The biocompatibility of the nanogel was comprehensively analyzed, through cytotoxicity (lactate dehydrogenase (LDH) release, MTS, Live, and Dead) and genotoxicity (comet) assays. The release profile of rIL-10 and its biological activity were evaluated in vivo, using C57BL/6 mice. Although able to maintain a stable concentration of IL-10 for at least 4 h in mice serum, the amount of protein released was rather low. Despite this, the amount of rIL-10 released from the complex was biologically active inhibiting TNF-α production, in vivo, by LPS-challenged mice. In spite of the significant stabilization achieved using the nanogel, rIL-10 still denatures rather quickly. An additional effort is thus necessary to develop an effective delivery system for this cytokine, able to release active protein over longer periods of time. Nevertheless, the good biocompatibility, the protein stabilization effect and the ability to perform as a carrier with controlled release suggest that self-assembled dextrin nanogels may be useful protein delivery systems.


Asunto(s)
Dextrinas/administración & dosificación , Portadores de Fármacos/administración & dosificación , Factores Inmunológicos/farmacología , Factores Inmunológicos/farmacocinética , Interleucina-10/farmacología , Interleucina-10/farmacocinética , Polietilenglicoles/administración & dosificación , Polietileneimina/administración & dosificación , Animales , Dextrinas/efectos adversos , Portadores de Fármacos/efectos adversos , Ratones , Ratones Endogámicos C57BL , Nanogeles , Polietilenglicoles/efectos adversos , Polietileneimina/efectos adversos , Desnaturalización Proteica , Suero/química
7.
Macromol Biosci ; 19(1): e1800248, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30259658

RESUMEN

Shear-thinning hydrogels are useful for biomedical applications, from 3D bioprinting to injectable biomaterials. Although they have the appropriate properties for injection, it may be advantageous to decouple injectability from the controlled release of encapsulated therapeutics. Toward this, composites of hydrogels and encapsulated microgels are introduced with microgels that are fabricated via microfluidics. The microgel cross-linker controls degradation and entrapped molecule release, and the concentration of microgels alters composite hydrogel rheological properties. For the treatment of myocardial infarction (MI), interleukin-10 (IL-10) is encapsulated in microgels and released from composites. In a rat model of MI, composites with IL-10 reduce macrophage density after 1 week and improve scar thickness, ejection fraction, cardiac output, and the size of vascular structures after 4 weeks when compared to saline injection. Improvements are also observed with the composite without IL-10 over saline, emphasizing the role of injectable hydrogels alone on tissue repair.


Asunto(s)
Materiales Biocompatibles , Hidrogeles , Interleucina-10 , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacocinética , Materiales Biocompatibles/farmacología , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Modelos Animales de Enfermedad , Hidrogeles/química , Hidrogeles/farmacocinética , Hidrogeles/farmacología , Interleucina-10/química , Interleucina-10/farmacocinética , Interleucina-10/farmacología , Infarto del Miocardio/metabolismo , Ratas
8.
Acta Biomater ; 90: 179-191, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30936036

RESUMEN

Although there are numerous medical applications to recover damaged skin tissue, scarless wound healing is being extensively investigated to provide a better therapeutic outcome. The exogenous delivery of therapeutic growth factors (GFs) is one of the engineering strategies for skin regeneration. This study presents an exogenous GF delivery platform developed using coacervates (Coa), a tertiary complex of poly(ethylene argininyl aspartate diglyceride) (PEAD) polycation, heparin, and cargo GFs (i.e., transforming growth factor beta 3 (TGF-ß3) and interleukin 10 (IL-10)). Coa encompasses the advantage of high biocompatibility, facile preparation, protection of cargo GFs, and sustained GF release. We therefore speculated that coacervate-mediated dual delivery of TGF-ß3/IL-10 would exhibit synergistic effects for the reduction of scar formation during physiological wound healing. Our results indicate that the exogenous administration of dual GF via Coa enhances the proliferation and migration of skin-related cells. Gene expression profiles using RT-PCR revealed up-regulation of ECM formation at early stage of wound healing and down-regulation of scar-related genes at later stages. Furthermore, direct injection of the dual GF Coa into the edges of damaged skin in a rat skin wound defect model demonstrated accelerated wound closure and skin regeneration after 3 weeks. Histological evaluation and immunohistochemical staining also revealed enhanced formation of the epidermal layer along with facilitated angiogenesis following dual GF Coa delivery. Based on these results, we conclude that polycation-mediated Coa fabrication and exogenous dual GF delivery via the Coa platform effectively augments both the quantity and quality of regenerated skin tissues without scar formation. STATEMENT OF SIGNIFICANCE: This study was conducted to develop a simple administration platform for scarless skin regeneration using polycation-based coacervates with dual GFs. Both in vitro and in vivo studies were performed to confirm the therapeutic efficacy of this platform toward scarless wound healing. Our results demonstrate that the platform developed by us enhances the proliferation and migration of skin-related cells. Sequential modulation in various gene expression profiles suggests a balanced collagen-remodeling process by dual GFs. Furthermore, in vivo histological evaluation demonstrates that our technique enhances clear epidermis formation with less scab and thicker woven structure of collagen bundle, similar to that of a normal tissue. We propose that simple administration of dual GFs with Coa has the potential to be applied as a clinical approach for fundamental scarless skin regeneration.


Asunto(s)
Cicatriz/prevención & control , Dermis/metabolismo , Sistemas de Liberación de Medicamentos , Fibroblastos/metabolismo , Interleucina-10 , Factor de Crecimiento Transformador beta3 , Cicatrización de Heridas/efectos de los fármacos , Cicatriz/metabolismo , Cicatriz/patología , Dermis/patología , Fibroblastos/patología , Humanos , Interleucina-10/química , Interleucina-10/farmacocinética , Interleucina-10/farmacología , Factor de Crecimiento Transformador beta3/química , Factor de Crecimiento Transformador beta3/farmacocinética , Factor de Crecimiento Transformador beta3/farmacología
9.
Cancer Cell ; 35(6): 901-915.e4, 2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-31185213

RESUMEN

Increasing evidence demonstrates that interleukin-10 (IL-10), known as an immunosuppressive cytokine, induces antitumor effects depending on CD8+ T cells. However, it remains elusive how immunosuppressive effects of IL-10 contribute to CD8+ T cell-mediated antitumor immunity. We generated Cetuximab-based IL-10 fusion protein (CmAb-(IL10)2) to prolong its half-life and allow tumor-targeted delivery of IL-10. Our results demonstrated potent antitumor effects of CmAb-(IL10)2 with reduced toxicity. Moreover, we revealed a mechanism of CmAb-(IL10)2 preventing dendritic cell (DC)-mediated CD8+ tumor-infiltrating lymphocyte apoptosis through regulating IFN-γ production. When combined with immune checkpoint blockade, CmAb-(IL10)2 significantly improves antitumor effects in mice with advanced tumors. Our findings reveal a DC-regulating role of IL-10 to potentiate CD8+ T cell-mediated antitumor immunity and provide a potential strategy to improve cancer immunotherapy.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Apoptosis/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Cetuximab/farmacología , Células Dendríticas/efectos de los fármacos , Interleucina-10/farmacología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos Inmunológicos/farmacocinética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Comunicación Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cetuximab/farmacocinética , Técnicas de Cocultivo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Resistencia a Antineoplásicos , Femenino , Humanos , Interleucina-10/farmacocinética , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Terapia Molecular Dirigida , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Recombinantes de Fusión/farmacología , Transducción de Señal , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cytokine ; 40(3): 183-92, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17980615

RESUMEN

The powerful anti-inflammatory and immunosuppressive activities of IL-10 make it attractive for supplemental therapy in translational tolerance induction protocols. This is bolstered by reports of IL-10-mediated inhibition of innate immunity, association of human stem cell and nonhuman primate (NHP) islet allograft tolerance with elevated serum IL-10, and evidence that systemic IL-10 therapy enhanced pig islets survival in mice. IL-10 has not been examined as adjunctive immunosuppression in NHP. To enable such studies, we cloned and expressed rhesus macaque (RM) IL-10 fused to a mutated hinge region of human IgG1 Fc to generate IL-10/Fc(ala-ala). RM IL-10/Fc(ala-ala) was purified to approximately 98% homogeneity by affinity chromatography and shown to be endotoxin-free (<0.008 EU/microg protein). The biological activity of IL-10/Fc(ala-ala) was demonstrated by (1) costimulation of the mouse mast cell line, MC/9 proliferation in a dose-dependent fashion, (2) suppression of LPS-induced septic shock in mice and (3) abrogation of LPS-induced secretion of proinflammatory cytokines/chemokines in vitro and in vivo in NHP. Notably, RM IL-10/Fc(ala-ala) had significantly greater potency than human IL-10/Fc(ala-ala) and exhibited a circulating half-life of approximately 14 days. The availability of this reagent will facilitate definitive studies to determine whether supplemental therapy with RM IL-10/Fc(ala-ala) can influence tolerance outcomes in NHP.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Fragmentos Fc de Inmunoglobulinas/farmacología , Interleucina-10/farmacología , Mastocitos/inmunología , Proteínas Recombinantes de Fusión/farmacología , Choque Séptico/tratamiento farmacológico , Tolerancia al Trasplante/efectos de los fármacos , Animales , Línea Celular , Clonación Molecular , Relación Dosis-Respuesta a Droga , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/inmunología , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Inmunoglobulina G/farmacología , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/trasplante , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-10/farmacocinética , Lipopolisacáridos/toxicidad , Macaca mulatta , Ratones , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacocinética , Choque Séptico/inducido químicamente , Choque Séptico/inmunología , Porcinos , Trasplante Heterólogo
11.
J Clin Oncol ; 34(29): 3562-3569, 2016 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-27528724

RESUMEN

Purpose Interleukin-10 (IL-10) stimulates the expansion and cytotoxicity of tumor-infiltrating CD8+ T cells and inhibits inflammatory CD4+ T cells. Pegylation prolongs the serum concentration of IL-10 without changing the immunologic profile. This phase I study sought to determine the safety and antitumor activity of AM0010. Patients and Methods Patients with selected advanced solid tumors were treated with AM0010 in a dose-escalation study, which was followed by a renal cell cancer (RCC) dose-expansion cohort. AM0010 was self-administered subcutaneously at doses of 1 to 40 µg/kg once per day. Primary end points were safety and tolerability; clinical activity and immune activation were secondary end points. Results In the dose-escalation and -expansion cohorts, 33 and 18 patients, respectively, were treated with daily subcutaneous injection of AM0010. AM0010 was tolerated in a heavily pretreated patient population. Treatment-related adverse events (AEs) included anemia, fatigue, thrombocytopenia, fever, and injection site reactions. Grade 3 to 4 nonhematopoietic treatment-related AEs, including rash (n = 2) and transaminitis (n = 1), were observed in five of 33 patients. Grade 3 to 4 anemia or thrombocytopenia was observed in five patients. Most treatment-related AEs were transient or reversible. AM0010 led to systemic immune activation with elevated immune-stimulatory cytokines and reduced transforming growth factor beta in the serum. Partial responses were observed in one patient with uveal melanoma and four of 15 evaluable patients with RCC treated at 20 µg/kg (overall response rate, 27%). Prolonged stable disease of at least 4 months was observed in four patients, including one with colorectal cancer with disease stabilization for 20 months. Conclusion AM0010 has an acceptable toxicity profile with early evidence of antitumor activity, particularly in RCC. These data support the further evaluation of AM0010 both alone and in combination with other immune therapies and chemotherapies.


Asunto(s)
Citocinas/sangre , Interleucina-10/efectos adversos , Neoplasias/tratamiento farmacológico , Polietilenglicoles/efectos adversos , Adulto , Anciano , Anciano de 80 o más Años , Anemia/inducido químicamente , Carcinoma de Células Renales/tratamiento farmacológico , Erupciones por Medicamentos/etiología , Exantema/inducido químicamente , Fatiga/inducido químicamente , Femenino , Fiebre/inducido químicamente , Humanos , Inyecciones Subcutáneas/efectos adversos , Interferón gamma/sangre , Interleucina-10/inmunología , Interleucina-10/farmacocinética , Interleucina-10/uso terapéutico , Interleucina-4/sangre , Interleucina-8/sangre , Neoplasias Renales/tratamiento farmacológico , Masculino , Melanoma/tratamiento farmacológico , Persona de Mediana Edad , Neoplasias/patología , Polietilenglicoles/farmacocinética , Polietilenglicoles/uso terapéutico , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapéutico , Trombocitopenia/inducido químicamente , Factor de Crecimiento Transformador beta/sangre , Neoplasias de la Úvea/tratamiento farmacológico , Adulto Joven
12.
Circulation ; 100(10): 1102-8, 1999 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-10477536

RESUMEN

BACKGROUND: The T helper cell type 2-associated cytokine interleukin (IL)-10 has a variety of immunomodulatory properties. However, the effects of the cytokine on viral myocarditis remain unclear. METHODS AND RESULTS: We studied the effects of recombinant human IL-10 (rhIL-10) fully active on mouse cells in a murine experimental model of acute viral myocarditis caused by the encephalomyocarditis virus (EMCV). Four-week-old DBA/2 mice were inoculated with EMCV (day 0). rhIL-10 (10 microg/mouse) was administered once daily, starting on day 0, and control mice received vehicle only. Survival rates were determined on day 14. Myocardial histopathology, cytokine levels in the heart by ELISA assay, and myocardial virus concentration were examined on day 6, and the expression levels of myocardial inducible nitric oxide synthase (iNOS) mRNA were measured by competitive polymerase chain reaction. The 14-day survival in mice treated with rhIL-10 was significantly higher (80%) than in the control group (30%, n=10 in each, P<0.05). rhIL-10 treatment significantly attenuated myocardial lesions and suppressed tumor necrosis factor-alpha and IL-2 in the heart. rhIL-10 treatment had little effect on myocardial virus concentration. The expression levels of myocardial iNOS mRNA were significantly decreased in the group treated with rhIL-10 (8.6+/-4.7 amol/mg total RNA in treated versus 26.5+/-7.1 amol/mg total RNA in control mice, P<0.05). CONCLUSIONS: These findings provide new insights into the in vivo effects of IL-10 on viral infection and suggest a therapeutic effect of IL-10 on viral myocarditis.


Asunto(s)
Infecciones por Cardiovirus , Virus de la Encefalomiocarditis , Interleucina-10/uso terapéutico , Miocarditis/tratamiento farmacológico , Miocarditis/virología , Animales , Anticuerpos Monoclonales/uso terapéutico , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Humanos , Interleucina-10/administración & dosificación , Interleucina-10/inmunología , Interleucina-10/farmacocinética , Masculino , Ratones , Ratones Endogámicos DBA , Miocardio/metabolismo , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapéutico , Análisis de Supervivencia , Factores de Tiempo
13.
Circulation ; 101(8): 908-16, 2000 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-10694531

RESUMEN

BACKGROUND: Intimal hyperplasia after stent implantation is the main cause of in-stent restenosis. Activated monocytes play a key role in intimal growth. The anti-inflammatory cytokine interleukin-10 (IL-10) is a potent monocyte deactivator, endogenously produced in the atherosclerotic plaque. We tested the hypothesis that exogenous IL-10 may limit postangioplasty intimal hyperplasia after balloon angioplasty or stenting. METHODS AND RESULTS: Hypercholesterolemic rabbits were treated with recombinant human IL-10 (rhuIL-10) for 3 days after balloon angioplasty or 28 days after stent implantation. High IL-10 serum levels and intense deactivation of circulating monocytic cells, assessed by inhibition of IL-1beta release by lipopolysaccharide-stimulated whole blood, were detected for at least 8 hours after rhuIL-10 intravenous injection (ELISA). Morphometric analyses, performed 28 days after injury, indicated that rhuIL-10 reduced intimal growth by approximately 50% after balloon angioplasty or stenting, resulting in more preserved lumen in stented arteries. Moreover, rhuIL-10 reduced macrophage infiltration by 67% and proliferative activity by 81% in the intima and the media. No toxic effect was detected except minor changes in blood cell count. CONCLUSIONS: The anti-inflammatory cytokine rhuIL-10 reduces postinjury intimal hyperplasia. The potent attenuation of in-stent intimal growth by rhuIL-10 and its favorable toxicity profile suggest that rhuIL-10 may be useful in the prevention of in-stent restenosis.


Asunto(s)
Cateterismo/efectos adversos , Enfermedad Coronaria/prevención & control , Hipercolesterolemia/complicaciones , Interleucina-10/uso terapéutico , Stents/efectos adversos , Túnica Íntima/patología , Animales , Enfermedad Coronaria/etiología , Enfermedad Coronaria/terapia , Depresión Química , Dieta Aterogénica , Humanos , Hiperplasia , Interleucina-1/metabolismo , Interleucina-10/farmacocinética , Interleucina-10/farmacología , Interleucina-10/toxicidad , Leucocitos/efectos de los fármacos , Lipopolisacáridos/farmacología , Masculino , Conejos , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapéutico , Proteínas Recombinantes/toxicidad , Recurrencia , Especificidad de la Especie , Túnica Íntima/lesiones , Vasculitis/etiología , Vasculitis/patología , Vasculitis/prevención & control
14.
Clin Cancer Res ; 6(7): 2661-9, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10914707

RESUMEN

A pilot dose-escalation study of recombinant human interleukin 12 (rhIL-12) was conducted in Japanese patients with advanced malignancies. Cohorts of three patients received escalating doses of rhIL-12 that increased from 50 to 300 ng/kg/day s.c. three times a week for 2 weeks followed by 1-week rest. The same dosage and schedule was repeated for two additional courses. Sixteen previously treated patients were registered, and 15 were evaluated. Common toxicities were fever and leukopenia; the abnormality of laboratory tests included elevations in aspartate aminotransferase, alanine aminotransferase, alkaline phosphatase, C-reactive protein, and beta2-microglobin. Dose-limiting toxicity was the grade 3 elevation of aminotransferases, and was observed in two of six patients at the 300-ng/kg dose level after the first course in one patient and after the third course in the other. Leukopenia was observed at all of the dose levels; two of six patients at 300 ng/kg experienced grade 3 leukopenia. Thus, 300 ng/kg was determined to be the maximum acceptable dose. Peak plasma levels of rhIL-12 decreased in the second courses, but the areas under the curve were almost the same in the first and second courses. Biological effects included increases of plasma levels of IFN-gamma, tumor necrosis factor-alpha, IL-6, IL-10, and neopterin. In two patients with renal cell carcinoma, complete response and partial response of metastatic tumors were observed with 50 and 300 ng/kg; the responses lasted for 5 and 3.5 months, respectively. Although immunological response to rhIL-12 varies depending on administration route and schedule and on patients' physiological conditions, the recommended dose for Phase II studies is 300 ng/kg s.c. three times a week for 2 weeks followed by 1-week rest.


Asunto(s)
Interleucina-10/efectos adversos , Interleucina-10/farmacocinética , Neoplasias/tratamiento farmacológico , Adulto , Anciano , Carcinoma de Células Renales/tratamiento farmacológico , Estudios de Cohortes , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Infusiones Intravenosas , Interferón gamma/sangre , Interleucina-10/administración & dosificación , Interleucina-10/sangre , Interleucina-6/sangre , Japón , Neoplasias Renales/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Neoplasias/sangre , Neoplasias/inmunología , Neopterin/sangre , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/farmacocinética , Factor de Necrosis Tumoral alfa/análisis
15.
Clin Pharmacol Ther ; 62(2): 171-80, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9284853

RESUMEN

Interleukin-10 inhibits T-lymphocyte activation and proliferation and lipopolysaccharide-induced monocyte production of proinflammatory cytokines. Fifty-four healthy volunteers received single doses of recombinant human interleukin-10 (1.0, 2.5, 5.0, 10, 25, or 50 micrograms/kg) or placebo by subcutaneous injection (randomized double-blind assignment). Clinical adverse events were infrequent at doses below 50 micrograms/kg (five of six subjects had mild flu-like syndrome). Mean serum interleukin-10 concentrations were dose related. The mean terminal-phase half-life ranged from 2.7 to 4.5 hours, and the apparent volume of distribution ranged from 0.70 to 1.35 L/kg. Hematologic changes included transient mild to moderate increases of neutrophil counts, decreases of lymphocyte counts, and a delayed decrease of platelet counts. Recombinant human interleukin-10 significantly suppressed production of the proinflammatory cytokines interleukin-1 beta and tumor necrosis factor-alpha by whole blood stimulated ex vivo with Escherichia coli lipopolysaccharide.


Asunto(s)
Interleucina-10/farmacología , Activación de Linfocitos/efectos de los fármacos , Proteínas Recombinantes/farmacología , Linfocitos T/efectos de los fármacos , Adulto , Antígenos CD/metabolismo , Método Doble Ciego , Escherichia coli , Femenino , Semivida , Humanos , Inyecciones Subcutáneas , Proteína Antagonista del Receptor de Interleucina 1 , Interleucina-1/metabolismo , Interleucina-10/efectos adversos , Interleucina-10/farmacocinética , Lipopolisacáridos/farmacología , Masculino , Receptores de Interleucina-1/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/farmacocinética , Seguridad , Sialoglicoproteínas/metabolismo , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
16.
Brain Res Mol Brain Res ; 114(2): 168-71, 2003 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-12829328

RESUMEN

Interleukin (IL)-10 exerts beneficial effects on the central nervous system (CNS) after peripheral administration, but its penetration across the blood-brain barrier (BBB) has not been quantified. We show that 125I-IL-10 is stable in circulating blood but does not cross the intact BBB after intravenous delivery. Thus, peripheral IL-10 probably can serve as a CNS therapeutic only when the BBB is disrupted.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Interleucina-10/farmacocinética , Albúminas/farmacocinética , Animales , Barrera Hematoencefálica/fisiología , Encéfalo/metabolismo , Encefalopatías/tratamiento farmacológico , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Difusión/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Interleucina-10/uso terapéutico , Radioisótopos de Yodo , Masculino , Lípidos de la Membrana/metabolismo , Tasa de Depuración Metabólica/efectos de los fármacos , Ratones , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Tecnecio
17.
Rheum Dis Clin North Am ; 24(3): 629-39, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9710891

RESUMEN

IL-10 has anti-inflammatory and immunoregulatory properties that suggest a potential therapeutic role in RA. IL-10 inhibits proinflammatory cytokine and chemokine production in addition to blocking T-cell responses to specific antigens. It acts primarily through inhibition of costimulatory properties of macrophages. IL-10 stimulates proliferation and differentiation of antibody-forming B-cells. Preclinical studies in a variety of animal models, including collagen-induced arthritis, have shown that IL-10 is effective in preventing or inhibiting inflammation and autoreactivity. Although in RA, circulating and synovial levels of IL-10 are increased, accumulated evidence suggests that there may be a relative deficit of available IL-10. Moreover, exogenous addition of IL-10 in vitro has been shown to affect the immunopathological processes involved in RA. Preliminary studies of human recombinant IL-10 in patients with RA have demonstrated a trend towards efficacy with a good safety profile. Taken together, the data support a therapeutic role for IL-10 in the treatment of RA.


Asunto(s)
Artritis Reumatoide/terapia , Interleucina-10/uso terapéutico , Artritis Reumatoide/inmunología , Artritis Reumatoide/fisiopatología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Humanos , Inflamación/inmunología , Interleucina-10/farmacocinética , Interleucina-10/farmacología , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Líquido Sinovial
18.
J Clin Pharmacol ; 39(10): 1015-20, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10516935

RESUMEN

The pharmacokinetics of intravenously administered recombinant human interleukin-10 (rHuIL-10) were evaluated in 18 subjects with creatinine clearances (Clcr) between 2.7 and 116.7 mL/min/1.73 m2. Serum samples for rHuIL-10 were obtained over a 48-hour period after a single 25 micrograms/kg i.v. bolus infusion. AUC, total body clearance (Clp), and steady-state volume of distribution (Vdss) were derived by compartmental methods. Analysis of serum concentrations showed statistically significant group differences for log-transformed AUC and original scale Clp (p < 0.01). The AUC and effective half-life increased, while the mean Clp of rHuIL-10 decreased as renal function declined. A linear relationship between AUC and Clcr as well as Clp and Clcr demonstrates that the disposition of rHuIL-10 is altered in subjects with renal insufficiency. No serious adverse events were noted.


Asunto(s)
Interleucina-10/farmacocinética , Riñón/fisiología , Adulto , Anciano , Área Bajo la Curva , Creatinina/orina , Interpretación Estadística de Datos , Fiebre/inducido químicamente , Rubor/inducido químicamente , Cefalea/inducido químicamente , Humanos , Interleucina-10/efectos adversos , Interleucina-10/sangre , Pruebas de Función Renal , Tasa de Depuración Metabólica , Persona de Mediana Edad , Enfermedades Musculares/inducido químicamente , Dolor/inducido químicamente , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/sangre , Proteínas Recombinantes/farmacocinética
19.
J Clin Pharmacol ; 39(6): 624-35, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10354967

RESUMEN

The pharmacokinetic and adrenal interactions of recombinant human interleukin-10 and prednisolone were examined in this open-label, randomized, four-way crossover study in 12 healthy adult male volunteers. Single doses of IL-10 (8 micrograms/kg s.c.), IL-10 with prednisone (15 mg p.o.), placebo with prednisone, or placebo were administered on four separate occasions with at least 3-week interceding washout periods. Measurements included plasma prednisone, prednisolone and cortisol, unbound prednisolone, and serum IL-10 concentrations. Pharmacokinetic parameters were determined using noncompartmental and model-fitting analysis, while area analysis and an indirect response model were used to assess cortisol dynamics. IL-10 exhibited prolonged serum concentrations owing to dual-absorption processes that were largely unaffected by prednisone. The Cmax values were about 3 ng/mL, while the tmax occurred at 7 to 9 hours. Prednisolone exhibited rapid systemic kinetics with a Cmax of 235 ng/mL, tmax at 1.11 hours, and t1/2 of 2.54 hours with no significant alterations owing to IL-10. Both prednisolone and prednisolone/IL-10 caused marked suppression of cortisol concentrations with similar magnitude and IC50 values; however, IL-10 alone significantly increased the 24-hour AUC of cortisol by 20%. Thus, IL-10 and prednisolone do not interact in disposition or adrenal suppression to a clinically significant degree.


Asunto(s)
Glándulas Suprarrenales/efectos de los fármacos , Antiinflamatorios/farmacocinética , Interleucina-10/farmacocinética , Prednisona/farmacocinética , Glándulas Suprarrenales/metabolismo , Adulto , Área Bajo la Curva , Estudios Cruzados , Interacciones Farmacológicas , Humanos , Hidrocortisona/sangre , Masculino , Tasa de Depuración Metabólica
20.
Eur J Pharmacol ; 321(2): 231-9, 1997 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-9063693

RESUMEN

The pro-inflammatory peptide tumor necrosis factor-alpha (TNF) stimulates production of the anti-inflammatory cytokine-interleukin-10 by monocytes which in turn inhibits the synthesis of TNF. This inhibitory effect of interleukin-10 may contribute to the balance of pro- and anti-inflammatory cytokines in several diseases, e.g., chronic inflammatory bowel disease. In the present study we addressed the question whether interleukin-10 in combination with other TNF-suppressing agents leads to enhanced suppression of TNF synthesis. We investigated the inhibitory potency of interleukin-10 in combination with rolipram, a specific type IV phosphodiesterase inhibitor, or with cicaprost, a stable prostacyclin analogue in lipopolysaccharide-stimulated human peripheral blood mononuclear cells. Peripheral blood mononuclear cells were stimulated with 10 ng/ml lipopolysaccharide in the absence or presence of interleukin-10 or one of the cAMP-elevating agents. First, we confirmed the TNF-suppressing effect of interleukin-10, rolipram and cicaprost alone and determined the IC50 for these substances. Second, for the combination of interleukin-10 with one of the cAMP-elevating substances we were able to demonstrate enhanced TNF inhibition. Of these, the combination of interleukin-10 and rolipram revealed an additive effect. The maximal TNF synthesis of 5.5 +/- 1.1 ng/ml after lipopolysaccharide stimulation alone was inhibited by 0.1 ng/ml interleukin-10 to 2.7 +/- 0.6 ng/ml TNF and by 100 nM rolipram to 3.1 +/- 0.6 ng/ml TNF. Both substances combined suppressed TNF synthesis to 1.5 +/- 0.3 ng/ml. After stimulation with Staphylococcus epidermidis we could demonstrate a more pronounced inhibition of TNF synthesis by interleukin-10 compared to rolipram which was more effective after stimulation with lipopolysaccharide. Finally, the additive inhibitory effect of interleukin-10 and rolipram could be confirmed on the level of TNF mRNA. The results obtained in the present investigation could form a prerequisite to study the combination of interleukin-10 and cAMP-elevating agents in in vivo models of acute or chronic inflammatory diseases.


Asunto(s)
Antineoplásicos/farmacología , AMP Cíclico/biosíntesis , Epoprostenol/análogos & derivados , Interleucina-10/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Pirrolidinonas/farmacología , Factor de Necrosis Tumoral alfa/biosíntesis , Northern Blotting , Células Cultivadas , AMP Cíclico/sangre , Sinergismo Farmacológico , Epoprostenol/farmacología , Humanos , Interleucina-10/farmacocinética , Cinética , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/farmacología , Inhibidores de Fosfodiesterasa/farmacocinética , Pirrolidinonas/farmacocinética , ARN Mensajero/metabolismo , Rolipram , Staphylococcus epidermidis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA