Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biopharm Drug Dispos ; 41(3): 111-125, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32080869

RESUMEN

The immunogenicity of biotherapeutics presents a major challenge during the clinical development of new protein drugs including monoclonal antibodies. To address this, multiple humanization and de-immunization techniques that employ in silico algorithms and in vitro test systems have been proposed and implemented. However, the success of these approaches has been variable and to date, the ability of these techniques to predict immunogenicity has not been systematically tested in humans or other primates. This study tested whether antibody humanization and de-immunization strategies reduce the risk of anti-drug antibody (ADA) development using cynomolgus macaque as a surrogate for human. First human-cyno chimeric antibodies were constructed by grafting the variable domains of the adalimumab and golimumab monoclonal antibodies onto cynomolgus macaque IgG1 and Igκ constant domains followed by framework germlining to cyno to reduce the xenogenic content. Next, B and T cell epitopes and aggregation-prone regions were identified using common in silico methods to select domains with an ADA risk for additional modification. The resultant engineered antibodies had a comparable affinity for TNFα, demonstrated similar biophysical properties, and exhibited significantly reduced ADA levels in cynomolgus macaque compared with the parental antibodies, with a corresponding improvement in the pharmacokinetic profile. Notably, plasma concentrations of the engineered antibodies were quantifiable through 504 hours (chimeric) and 840 hours (germlined/de-immunized), compared with only 336 hours (adalimumab) or 336-672 hours (golimumab). The results point to the significant value in the investment in these engineering strategies as an important guide for monoclonal antibody optimization that can contribute to improved clinical outcomes.


Asunto(s)
Adalimumab/inmunología , Anticuerpos Monoclonales/inmunología , Inmunoglobulina G/inmunología , Animales , Femenino , Humanos , Inmunización , Macaca fascicularis , Masculino , Factor de Necrosis Tumoral alfa/inmunología
2.
Immunogenetics ; 71(10): 605-615, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31776588

RESUMEN

Immunogenicity of biomolecules is one of the largest concerns in biological therapeutic drug development. Adverse immune responses as a result of immunogenicity to biotherapeutics range from mild hypersensitivity reactions to potentially life-threatening anaphylactic reactions and can negatively impact human health and drug efficacy. Numerous confounding patient-, product- or treatment-related factors can influence the development of an immune reaction against therapeutic proteins. The goal of this study was to investigate the relationship between pre-existing drug reactivity (PE-ADA), individual immunogenetics (MHC class II haplotypes), and development of treatment-induced antidrug antibodies (TE-ADA) in cynomolgus macaque. PE-ADA refers to the presence of antibodies immunoreactive against the biotherapeutic in treatment-naïve individuals. We observed that PE-ADA frequency against four different bispecific antibodies in naïve cynomolgus macaque is similar to that reported in humans. Additionally, we report a trend towards an increased incidence of TE-ADA development in macaques with high PE-ADA levels. In order to explore the relationship between MHC class II alleles and risk of ADA development, we obtained full-length MHC class II sequences from 60 cynomolgus macaques in our colony. We identified a total of 248 DR, DP, and DQ alleles and 236 unique haplotypes in our cohort indicating a genetically complex set of animals potentially reflective of the human population. Based on our observations, we propose the evaluation of the magnitude/frequency of pre-existing reactivity and consideration of MHC class II genetics as additional useful tools to understand the immunogenic potential of biotherapeutics.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Hipersensibilidad a las Drogas/inmunología , Genes MHC Clase II/genética , Genes MHC Clase II/inmunología , Inmunogenética , Animales , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/sangre , Hipersensibilidad a las Drogas/genética , Frecuencia de los Genes , Haplotipos , Macaca fascicularis , Masculino , Filogenia
3.
Cephalalgia ; 39(10): 1241-1248, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31003588

RESUMEN

OBJECTIVE: The objective of this investigation was to examine the distribution of galcanezumab and a control immunoglobulin 4 antibody containing the same constant regions as galcanezumab, into peripheral and central tissues. METHODS: Galcanezumab and a control immunoglobulin 4 antibody were radioiodinated with Iodine-125 to specific activities of 0.11 mCi/mg and 0.16 mCi/mg, respectively. At 24, 72, and 168 hours following subcutaneous injection of either antibody (4 mg/kg), cerebrospinal fluid and plasma were obtained followed by saline perfusion to remove residual blood and collection of selected tissues for determination of Iodine-125 content by gamma counting. RESULTS: The peak plasma levels of Iodine-125 galcanezumab and Iodine-125 control immunoglobulin 4 were observed at 72 hours and remained high at 168 hours post-dose. The rank order of tissue levels was dura mater = spleen > trigeminal ganglia ≫hypothalamus = spinal cord = prefrontal cortex = cerebellum. Iodine-125 galcanezumab levels in peripheral tissue (dura mater, spleen, and trigeminal ganglia) averaged 5% to 11% of plasma, whereas all of the central nervous system (CNS) tissue levels and the cerebrospinal fluid levels were < 0.4% of plasma. Distribution of the antibodies into the dura mater and the trigeminal ganglia was similar to that observed in the spleen and significantly greater than exposure in the brain or spinal cord. CONCLUSIONS: The central levels of galcanezumab were relatively low, which would favor the dura mater and trigeminal ganglia as sites of action for its observed clinical efficacy. However, a central site of action cannot be excluded.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacocinética , Encéfalo/metabolismo , Médula Espinal/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Duramadre/metabolismo , Radioisótopos de Yodo , Masculino , Ratas , Ratas Sprague-Dawley , Bazo/metabolismo , Distribución Tisular , Ganglio del Trigémino/metabolismo
4.
J Biol Chem ; 291(21): 11337-47, 2016 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-27022022

RESUMEN

Despite the increasing number of multivalent antibodies, bispecific antibodies, fusion proteins, and targeted nanoparticles that have been generated and studied, the mechanism of multivalent binding to cell surface targets is not well understood. Here, we describe a conceptual and mathematical model of multivalent antibody binding to cell surface antigens. Our model predicts that properties beyond 1:1 antibody:antigen affinity to target antigens have a strong influence on multivalent binding. Predicted crucial properties include the structure and flexibility of the antibody construct, the target antigen(s) and binding epitope(s), and the density of antigens on the cell surface. For bispecific antibodies, the ratio of the expression levels of the two target antigens is predicted to be critical to target binding, particularly for the lower expressed of the antigens. Using bispecific antibodies of different valencies to cell surface antigens including MET and EGF receptor, we have experimentally validated our modeling approach and its predictions and observed several nonintuitive effects of avidity related to antigen density, target ratio, and antibody affinity. In some biological circumstances, the effect we have predicted and measured varied from the monovalent binding interaction by several orders of magnitude. Moreover, our mathematical framework affords us a mechanistic interpretation of our observations and suggests strategies to achieve the desired antibody-antigen binding goals. These mechanistic insights have implications in antibody engineering and structure/activity relationship determination in a variety of biological contexts.


Asunto(s)
Reacciones Antígeno-Anticuerpo , Modelos Inmunológicos , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/metabolismo , Afinidad de Anticuerpos , Antígenos de Superficie/química , Antígenos de Superficie/metabolismo , Línea Celular , Receptores ErbB/química , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Humanos , Cinética , Unión Proteica , Proteínas Proto-Oncogénicas c-met/química , Proteínas Proto-Oncogénicas c-met/inmunología , Proteínas Proto-Oncogénicas c-met/metabolismo
5.
J Lipid Res ; 56(11): 2124-32, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26392590

RESUMEN

Lilly PCSK9 antibody LY3015014 (LY) is a monoclonal antibody (mAb) that neutralizes proprotein convertase subtilisin-kexin type 9 (PCSK9). LY decreases LDL cholesterol in monkeys and, unlike other PCSK9 mAbs, does not cause an accumulation of intact PCSK9 in serum. Comparing the epitope of LY with other clinically tested PCSK9 mAbs, it was noted that the LY epitope excludes the furin cleavage site in PCSK9, whereas other mAbs span this site. In vitro exposure of PCSK9 to furin resulted in degradation of PCSK9 bound to LY, whereas cleavage was blocked by other mAbs. These other mAbs caused a significant accumulation of serum PCSK9 and displayed a shorter duration of LDL-cholesterol lowering than LY when administered to mice expressing the WT human PCSK9. In mice expressing a noncleavable variant of human PCSK9, LY behaved like a cleavage-blocking mAb, in that it caused significant PCSK9 accumulation, its duration of LDL lowering was reduced, and its clearance (CL) from serum was accelerated. Thus, LY neutralizes PCSK9 and allows its proteolytic degradation to proceed, which limits PCSK9 accumulation, reduces the CL rate of LY, and extends its duration of action. PCSK9 mAbs with this property are likely to achieve longer durability and require lower doses than mAbs that cause antigen to accumulate.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticolesterolemiantes/farmacología , Proproteína Convertasas/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacocinética , Anticolesterolemiantes/química , Anticolesterolemiantes/farmacocinética , LDL-Colesterol/sangre , Evaluación Preclínica de Medicamentos , Estabilidad de Medicamentos , Furina/química , Semivida , Humanos , Hipercolesterolemia/tratamiento farmacológico , Macaca fascicularis , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proproteína Convertasa 9 , Proproteína Convertasas/inmunología , Unión Proteica , Proteolisis , Serina Endopeptidasas/inmunología , Resultado del Tratamiento
6.
J Pharmacol Exp Ther ; 354(3): 350-7, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26116630

RESUMEN

LY2951742, a monoclonal antibody targeting calcitonin gene-related peptide (CGRP), is being developed for migraine prevention and osteoarthritis pain. To support the clinical development of LY2951742, capsaicin-induced dermal blood flow (DBF) was used as a target engagement biomarker to assess CGRP activity in nonhuman primates and healthy volunteers. Inhibition of capsaicin-induced DBF in nonhuman primates, measured with laser Doppler imaging, was dose dependent and sustained for at least 29 days after a single intravenous injection of the CGRP antibody. This information was used to generate a pharmacokinetic/pharmacodynamic model, which correctly predicted inhibition of capsaicin-induced DBF in humans starting at a single subcutaneous 5-mg dose. As expected, the degree of inhibition in capsaicin-induced DBF increased with higher LY2951742 plasma concentrations. Utilization of this pharmacodynamic biomarker with pharmacokinetic data collected in phase I studies provided the dose-response relationship that assisted in dose selection for the phase II clinical development of LY2951742.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Péptido Relacionado con Gen de Calcitonina/metabolismo , Capsaicina/farmacología , Flujo Sanguíneo Regional/efectos de los fármacos , Administración Cutánea , Adolescente , Adulto , Animales , Biomarcadores/metabolismo , Método Doble Ciego , Humanos , Flujometría por Láser-Doppler/métodos , Macaca fascicularis , Masculino , Persona de Mediana Edad , Dolor/tratamiento farmacológico , Adulto Joven
7.
Drug Metab Dispos ; 43(10): 1477-83, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26175543

RESUMEN

Basal insulin peglispro (BIL) comprises insulin lispro covalently bound to a 20-kDa polyethylene glycol (PEG) at lysine B28. The biologic fate of BIL and unconjugated PEG were examined in rats given a single 0.5-mg/kg i.v. or 2-mg/kg s.c. dose of BIL with (14)C label in 20-kDa PEG or (125)I label in lispro. Unconjugated (14)C-labeled 20-kDa PEG was dosed at 10 mg/kg i.v. or s.c. Blood, urine, and feces were collected up to 336 hours after dosing. Radioactivity was measured by scintillation spectrometry, and BIL was quantitated by enzyme-linked immunosorbent assay. Area under the curve and half-life for immunoreactive BIL were lower than those for both (14)C and (125)I after subcutaneous and intravenous administration. The half-lives of (14)C after BIL and PEG dosing were similar. The clearance of immunoreactive BIL was 2.4-fold faster than that of (14)C and 1.6-fold faster than (125)I. After a subcutaneous dose of BIL, immunoreactive BIL accounted for 31% of the circulating (125)I and 16% of the circulating (14)C, indicating extensive catabolism of BIL. Subcutaneous bioavailability of BIL was 23%-29%; bioavailability for unconjugated PEG was 78%. For unconjugated PEG, most of the (14)C dose was recovered in urine. For BIL, ≥86% of (125)I was eliminated in urine and (14)C was eliminated about equally in urine and feces. The major (14)C-labeled catabolism product of BIL in urine was 20-kDa PEG with lysine attached. The attachment of 20-kDa PEG to lispro in BIL led to a different elimination pathway for PEG compared with unconjugated 20-kDa PEG.


Asunto(s)
Insulina/análogos & derivados , Polietilenglicoles/administración & dosificación , Polietilenglicoles/metabolismo , Animales , Disponibilidad Biológica , Inyecciones Intravenosas , Inyecciones Subcutáneas , Insulina/administración & dosificación , Insulina/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
8.
Drug Metab Dispos ; 42(11): 1867-72, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25024401

RESUMEN

Monoclonal antibodies (mAbs) represent an important class of therapeutic modalities. To optimize their pharmaceutical properties, studies have focused on improving mAb pharmacokinetic/pharmacodynamic profiles by modulating their interactions with the neonatal Fc receptor (FcRn). The influence of both the chemical and physical properties of IgGs has been examined in the context of FcRn interactions. In this regard, a variety of FcRn binding assays and tools have been developed and used to characterize the interaction with IgGs. However, a predictive relationship between the FcRn binding interaction of IgGs in vitro and their pharmacokinetics in vivo broadly across mAbs remains elusive. Many studies have increasingly suggested that the interplay between the characteristics of the mAb and the nature of its target can influence disposition and elimination. Thus, it is becoming increasingly evident that along with FcRn interactions, consideration of the non-FcRn-based biologic processes active in mAb disposition should be integrated into mAb development and optimization. Herein, we describe how the pharmacokinetics of mAbs can be modulated through FcRn interactions and provide perspectives on interpreting the receptor binding parameters in relation to other mechanisms involved in antibody disposition to aid in guiding mAb development.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos de Histocompatibilidad Clase I/metabolismo , Receptores Fc/metabolismo , Animales , Humanos , Farmacocinética , Unión Proteica
9.
Thorax ; 67(11): 985-92, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22735687

RESUMEN

BACKGROUND: Therapeutic strategies to modulate the host response to bacterial pneumonia are needed to improve outcomes during community-acquired pneumonia. This study used mice with impaired Fas signalling to examine susceptibility to pneumococcal pneumonia and decoy receptor 3 analogue (DcR3-a) to correct factors associated with increased susceptibility. METHODS: Wild-type mice and those with varying degrees of impairment of Fas (lpr) or Fas ligand signalling (gld) were challenged with Streptococcus pneumoniae and microbiological and immunological outcomes measured in the presence or absence of DcR3-a. RESULTS: During established pneumonia, neutrophils became the predominant cell in the airway and gld mice were less able to clear bacteria from the lungs, demonstrating localised impairment of pulmonary neutrophil function in comparison to lpr or wild-type mice. T-cells from gld mice had enhanced activation and reduced apoptosis in comparison to wild-type and lpr mice during established pneumonia. Treatment with DcR3-a reduced T-cell activation and corrected the defect in pulmonary bacterial clearance in gld mice. CONCLUSIONS: The results suggest that imbalance in tumour necrosis factor superfamily signalling and excessive T-cell activation can impair bacterial clearance in the lung but that DcR3-a treatment can reduce T-cell activation, restore optimal pulmonary neutrophil function and enhance bacterial clearance during S pneumoniae infection.


Asunto(s)
Proteína Ligando Fas/metabolismo , Neutrófilos/inmunología , Fagocitos/inmunología , Neumonía Neumocócica/inmunología , Miembro 6b de Receptores del Factor de Necrosis Tumoral/farmacología , Animales , Modelos Animales de Enfermedad , Proteína Ligando Fas/farmacología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Neutrófilos/efectos de los fármacos , Fagocitos/efectos de los fármacos , Neumonía Neumocócica/metabolismo , Neumonía Neumocócica/microbiología , Neumonía Neumocócica/terapia , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/prevención & control , Transducción de Señal/efectos de los fármacos , Streptococcus pneumoniae/inmunología
10.
Drug Metab Dispos ; 40(8): 1545-55, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22584253

RESUMEN

The pH-dependent binding of IgGs to the neonatal Fc receptor (FcRn) plays a critical role in regulating IgG homeostasis in vivo. Enhancing interactions between Fc and FcRn via protein engineering has been successfully used as an approach for improving the pharmacokinetics of monoclonal antibodies (mAbs). Although the quantitative translatability of the in vitro FcRn affinity enhancement to an in vivo pharmacokinetic benefit has been supported by several studies, there are also published reports indicating a disconnect in this relation. The body of literature suggests there are likely additional biochemical and biophysical properties of the mAbs along with their FcRn affinity that influence the in vivo pharmacokinetics. Herein, we more broadly evaluate the in vitro Fc-FcRn interactions and biochemical properties of five humanized IgG4 antibodies each with two Fc variant sequences (T250Q/M428L and V308P) and their corresponding pharmacokinetics in cynomolgus monkeys. Our findings indicate that the FcRn affinity-pharmacokinetic relationship does not show a direct correlation either across different IgGs or between the two variant sequences within a platform. Other parameters that have been suggested to contribute to mAb pharmacokinetic properties, such as the pH-dependent dissociation of the FcRn-IgG complexes, mAb biophysical properties, and nonspecific/charge binding characteristics of the mAbs, also did not independently explain the differing pharmacokinetic behaviors. Our results suggest that there is likely not a single in vitro parameter that readily predicts in vivo pharmacokinetics, but that the relative contribution and interplay of several factors along with the FcRn binding affinity are important determinants of mAb pharmacokinetic properties.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunoglobulina G/metabolismo , Receptores Fc/metabolismo , Animales , Línea Celular , Humanos , Inmunoglobulina G/química , Técnicas In Vitro , Macaca fascicularis , Unión Proteica
11.
Blood ; 113(21): 5277-86, 2009 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-19293425

RESUMEN

The anemia of chronic disease (ACD) is characterized by macrophage iron retention induced by cytokines and the master regulator hepcidin. Hepcidin controls cellular iron efflux on binding to the iron export protein ferroportin. Many patients, however, present with both ACD and iron deficiency anemia (ACD/IDA), the latter resulting from chronic blood loss. We used a rat model of ACD resulting from chronic arthritis and mimicked ACD/IDA by additional phlebotomy to define differing iron-regulatory pathways. Iron retention during inflammation occurs in macrophages and the spleen, but not in the liver. In rats and humans with ACD, serum hepcidin concentrations are elevated, which is paralleled by reduced duodenal and macrophage expression of ferroportin. Individuals with ACD/IDA have significantly lower hepcidin levels than ACD subjects, and ACD/IDA persons, in contrast to ACD subjects, were able to absorb dietary iron from the gut and to mobilize iron from macrophages. Circulating hepcidin levels affect iron traffic in ACD and ACD/IDA and are more responsive to the erythropoietic demands for iron than to inflammation. Hepcidin determination may aid to differentiate between ACD and ACD/IDA and in selecting appropriate therapy for these patients.


Asunto(s)
Anemia Ferropénica/patología , Anemia/patología , Homeostasis , Hierro/metabolismo , Anemia/sangre , Anemia/metabolismo , Anemia Ferropénica/sangre , Anemia Ferropénica/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/sangre , Estudios de Casos y Controles , Proteínas de Transporte de Catión/análisis , Enfermedad Crónica , Modelos Animales de Enfermedad , Duodeno/metabolismo , Hepcidinas , Humanos , Inflamación , Macrófagos/metabolismo , Macrófagos/patología , Ratas , Ratas Endogámicas Lew , Bazo/metabolismo , Bazo/patología
12.
Br J Haematol ; 148(3): 449-55, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19863534

RESUMEN

Recently, the iron and erythropoiesis-controlled growth differentiation factor 15 (GDF15) has been shown to inhibit the expression of hepcidin in beta-thalassaemia patients, thereby increasing iron absorption despite iron overload. To access the diagnostic and pathogenic impact of GDF15 in inflammatory anaemia the association of GDF15 expression with serum iron parameters and hepcidin was studied in patients suffering from iron deficiency anaemia (IDA), anaemia of chronic disease (ACD) and ACD subjects with true iron deficiency (ACD/IDA). GDF15 was significantly increased in both ACD and ACD/IDA, but not in IDA subjects as compared to controls. In contrast, hepcidin levels were significantly lower in IDA and ACD/IDA subjects than in ACD patients. IDA and ACD/IDA, but not ACD, showed an association between GDF15 and soluble transferrin receptor, an indicator of iron requirement for erythropoiesis. However, GDF15 did not correlate to hepcidin in either patient group. While GDF15 levels were linked to the needs for erythropoiesis and iron homeostasis in IDA, the immunity-driven increase of GDF15 may not primarily affect iron homeostasis and hepcidin expression. This indicates that other ACD-related factors may overcome the regulatory effects of GDF15 on hepcidin expression during inflammation.


Asunto(s)
Anemia/sangre , Factor 15 de Diferenciación de Crecimiento/sangre , Adulto , Anciano , Anemia/etiología , Anemia Ferropénica/sangre , Péptidos Catiónicos Antimicrobianos/sangre , Biomarcadores/sangre , Estudios de Casos y Controles , Enfermedad Crónica , Femenino , Factor 15 de Diferenciación de Crecimiento/fisiología , Hepcidinas , Humanos , Masculino , Persona de Mediana Edad
13.
Clin Chem ; 56(11): 1725-32, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20847326

RESUMEN

BACKGROUND: Hepcidin, a key regulator of iron metabolism, binds to the iron transporter ferroportin to cause its degradation. In humans, hepcidin deficiency has been linked to hemochromatosis and iron overload, whereas increased concentrations have been reported in anemia of cancer and chronic disease. There is currently an unmet clinical need for a specific immunoassay with a low limit of quantification to measure serum concentrations of hepcidin-25, the active form of the protein. METHODS: We generated 2 antihepcidin-25 monoclonal antibodies and used them to build a sandwich ELISA. We correlated ELISA results to hepcidin-25 measurements by LC-MS and used ELISA to measure serum hepcidin-25 concentrations in normal individuals, cancer patients, and patients with rheumatoid arthritis. RESULTS: The sandwich ELISA was highly specific for hepcidin-25, having a limit of quantification of 0.01 µg/L (10 pg/mL). Serum concentrations of hepcidin-25 measured by ELISA correlated with hepcidin-25 concentrations measured by using an independent LC-MS assay (r = 0.98, P < 0.001). Hepcidin-25 concentrations were increased in patients with cancer (median 54.8 µg/L, 25%-75% range 23.2-93.5 µg/L, n = 34) and rheumatoid arthritis (median 10.6 µg/L, 25%-75% range 5.9-18.4 µg/L, n = 76) compared with healthy individuals (median 1.20 µg/L, 25%-75% range 0.42-3.07 µg/L, n = 100). CONCLUSIONS: The use of 2 monoclonal antibodies in a sandwich ELISA format provides a robust and convenient method for measuring concentrations of the active form of hepcidin. This ELISA should help to improve our understanding of the role of hepcidin in regulating iron metabolism.


Asunto(s)
Anticuerpos Monoclonales , Péptidos Catiónicos Antimicrobianos/sangre , Adolescente , Adulto , Péptidos Catiónicos Antimicrobianos/inmunología , Artritis Reumatoide/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Hepcidinas , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/sangre , Grupos Raciales , Valores de Referencia , Factores Sexuales , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Adulto Joven
14.
Br J Haematol ; 144(5): 789-93, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19120353

RESUMEN

Iron absorption is inadequately increased in patients with chronic haemolytic anaemia, which is commonly complicated by iron overload. Growth differentiation factor 15 (GDF15) has been identified as a bone marrow-derived factor that abrogates hepcidin-mediated protection from iron overload under conditions of increased erythropoiesis. Increased concentrations of GDF15 have been reported in beta-thalassaemia patients and GDF15 has been found to suppress hepcidin expression in vitro. To further study the interdependencies of iron metabolism and erythropoiesis in vivo, the concentrations of hepcidin and GDF15 were determined in sera from 22 patients with pyruvate kinase deficiency (PKD) and 21 healthy control subjects. In PKD patients, serum hepcidin levels were 13-fold lower than in controls (2.0 ng/ml vs. 26.2 ng/ml) and GDF15 was significantly higher (859 pg/ml vs. 528 pg/ml). Serum hepcidin concentrations correlated positively with haemoglobin and negatively with serum GDF15. These results suggest that GDF15 contributes to low hepcidin expression and iron loading in PKD.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/sangre , Factor 15 de Diferenciación de Crecimiento/sangre , Sobrecarga de Hierro/sangre , Hierro/metabolismo , Piruvato Quinasa/deficiencia , Adolescente , Adulto , Anciano , Péptidos Catiónicos Antimicrobianos/metabolismo , Estudios de Casos y Controles , Eritropoyesis/fisiología , Femenino , Factor 15 de Diferenciación de Crecimiento/metabolismo , Hemoglobinas/análisis , Hepcidinas , Humanos , Masculino , Persona de Mediana Edad , Recuento de Reticulocitos , Estadísticas no Paramétricas , Adulto Joven
15.
MAbs ; 11(6): 1175-1190, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31181988

RESUMEN

We describe a bispecific dual-antagonist antibody against human B cell activating factor (BAFF) and interleukin 17A (IL-17). An anti-IL-17 single-chain variable fragment (scFv) derived from ixekizumab (Taltz®) was fused via a glycine-rich linker to anti-BAFF tabalumab. The IgG-scFv bound both BAFF and IL-17 simultaneously with identical stoichiometry as the parental mAbs. Stability studies of the initial IgG-scFv revealed chemical degradation and aggregation not observed in either parental antibody. The anti-IL-17 scFv showed a high melting temperature (Tm) by differential scanning calorimetry (73.1°C), but also concentration-dependent, initially reversible, protein self-association. To engineer scFv stability, three parallel approaches were taken: labile complementary-determining region (CDR) residues were replaced by stable, affinity-neutral amino acids, CDR charge distribution was balanced, and a H44-L100 interface disulfide bond was introduced. The Tm of the disulfide-stabilized scFv was largely unperturbed, yet it remained monodispersed at high protein concentration. Fluorescent dye binding titrations indicated reduced solvent exposure of hydrophobic residues and decreased proteolytic susceptibility was observed, both indicative of enhanced conformational stability. Superimposition of the H44-L100 scFv (PDB id: 6NOU) and ixekizumab antigen-binding fragment (PDB id: 6NOV) crystal structures revealed nearly identical orientation of the frameworks and CDR loops. The stabilized bispecific molecule LY3090106 (tibulizumab) potently antagonized both BAFF and IL-17 in cell-based and in vivo mouse models. In cynomolgus monkey, it suppressed B cell development and survival and remained functionally intact in circulation, with a prolonged half-life. In summary, we engineered a potent bispecific antibody targeting two key cytokines involved in human autoimmunity amenable to clinical development.


Asunto(s)
Anticuerpos Biespecíficos , Enfermedades Autoinmunes/tratamiento farmacológico , Factor Activador de Células B/antagonistas & inhibidores , Interleucina-17/antagonistas & inhibidores , Anticuerpos de Cadena Única , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Factor Activador de Células B/inmunología , Femenino , Células HEK293 , Células HT29 , Humanos , Interleucina-17/inmunología , Macaca fascicularis , Ratones , Ratones Transgénicos , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/farmacología
16.
J Clin Invest ; 115(6): 1627-35, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15902306

RESUMEN

Diabetes mellitus is a major health concern, affecting more than 5% of the population. Here we describe a potential novel therapeutic agent for this disease, FGF-21, which was discovered to be a potent regulator of glucose uptake in mouse 3T3-L1 and primary human adipocytes. FGF-21-transgenic mice were viable and resistant to diet-induced obesity. Therapeutic administration of FGF-21 reduced plasma glucose and triglycerides to near normal levels in both ob/ob and db/db mice. These effects persisted for at least 24 hours following the cessation of FGF-21 administration. Importantly, FGF-21 did not induce mitogenicity, hypoglycemia, or weight gain at any dose tested in diabetic or healthy animals or when overexpressed in transgenic mice. Thus, we conclude that FGF-21, which we have identified as a novel metabolic factor, exhibits the therapeutic characteristics necessary for an effective treatment of diabetes.


Asunto(s)
Diabetes Mellitus/tratamiento farmacológico , Factores de Crecimiento de Fibroblastos/administración & dosificación , Hipoglucemiantes/administración & dosificación , Adipocitos/citología , Adipocitos/metabolismo , Animales , Glucemia/análisis , División Celular/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus/sangre , Diabetes Mellitus/patología , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Hiperglucemia/sangre , Hiperglucemia/genética , Hiperglucemia/metabolismo , Hiperglucemia/patología , Hipoglucemiantes/metabolismo , Ratones , Ratones Obesos , Ratones Transgénicos , Triglicéridos/sangre , Aumento de Peso/efectos de los fármacos , Aumento de Peso/genética
17.
Transfusion ; 48(10): 2197-204, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18657084

RESUMEN

BACKGROUND: Individuals donating whole blood 13 times in a 2-year period without development of iron deficiency anemia (superdonors) are a self-selected population that is deferred for low hematocrit (Hct) level less frequently than other donors. STUDY DESIGN AND METHODS: Iron metabolism was assessed in 138 superdonors through a questionnaire and measurement of Hct, serum ferritin, serum hepcidin, and serum growth differentiation factor 15 (GDF15). Genetic testing for HFE and JAK-2 mutations was also performed. RESULTS AND CONCLUSIONS: Iron deficiency (ferritin level, <30 microg/L) is present in more than 60 percent of superdonors. Behaviors altering iron status included casual use of iron supplements in males, but not in females, and cigarette smoking that produced increased Hct associated with decreased ferritin. The striking biochemical characteristic of superdonors is greatly decreased serum hepcidin, consistent with their need to absorb maximal amounts of dietary iron to replace that lost from blood donation. GDF15 is normal in most superdonors, indicating that GDF15 overexpression arising from the expanded erythroid pool necessary to replace donated red cells is not the biochemical mechanism for the decreased serum hepcidin. Mutations in JAK-2 were not found, indicating that undiagnosed polycythemia vera is not a common cause for successful repeated blood donation by superdonors. Mutations in HFE associated with hemochromatosis were present in superdonors at the same frequency as the normal population. However, superdonors heterozygous for the H63D mutation in HFE had significantly decreased hepcidin : ferritin ratios demonstrating for the first time that the heterozygous state for HFE mutations is associated with alterations in hepcidin expression.


Asunto(s)
Anemia Ferropénica/sangre , Anemia Ferropénica/genética , Donantes de Sangre , Citocinas/genética , Antígenos de Histocompatibilidad Clase I/genética , Janus Quinasa 2/genética , Proteínas de la Membrana/genética , Anemia Ferropénica/tratamiento farmacológico , Péptidos Catiónicos Antimicrobianos/sangre , Femenino , Ferritinas/sangre , Factor 15 de Diferenciación de Crecimiento , Hematócrito , Proteína de la Hemocromatosis , Hepcidinas , Humanos , Hierro/farmacología , Hierro de la Dieta/farmacología , Masculino , Persona de Mediana Edad , Policitemia Vera/genética , Fumar/sangre , Encuestas y Cuestionarios
18.
Endocrinology ; 148(2): 774-81, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17068132

RESUMEN

Fibroblast growth factor (FGF)-21 has been recently characterized as a potent metabolic regulator. Systemic administration of FGF-21 reduced plasma glucose and triglycerides to near normal levels in genetically compromised diabetic rodents. Importantly, these effects were durable and did not come at the expense of weight gain, hypoglycemia, or mitogenicity. To explore the therapeutic properties of FGF-21 in a nongenetically modified primate species, and thus demonstrate the potential for efficacy in humans, we evaluated its bioactivity in diabetic nonhuman primates. When administered daily for 6 wk to diabetic rhesus monkeys, FGF-21 caused a dramatic decline in fasting plasma glucose, fructosamine, triglycerides, insulin, and glucagon. Of significant importance in regard to safety, hypoglycemia was not observed at any point during the study. FGF-21 administration also led to significant improvements in lipoprotein profiles, including lowering of low-density lipoprotein cholesterol and raising of high-density lipoprotein cholesterol, beneficial changes in the circulating levels of several cardiovascular risk markers/factors, and the induction of a small but significant weight loss. These data support the development of FGF-21 for the treatment of diabetes and other metabolic diseases.


Asunto(s)
Diabetes Mellitus/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Diabetes Mellitus/sangre , Diabetes Mellitus/etiología , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/administración & dosificación , Factores de Crecimiento de Fibroblastos/farmacocinética , Factores de Crecimiento de Fibroblastos/farmacología , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos , Ratones Mutantes , Obesidad/complicaciones , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología , Triglicéridos/sangre
19.
Oncotarget ; 8(55): 94619-94634, 2017 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-29212254

RESUMEN

Stromal cell-derived factor-1 (SDF-1) and its receptor CXCR4 play a critical role in mobilization and redistribution of immune cells and hematopoietic stem cells (HSCs). We evaluated effects of two CXCR4-targeting agents, peptide antagonist LY2510924 and monoclonal antibody LY2624587, on mobilizing HSCs and white blood cells (WBCs) in humans, monkeys, and mice. Biochemical analysis showed LY2510924 peptide blocked SDF-1/CXCR4 binding in all three species; LY2624587 antibody blocked binding in human and monkey, with minimal activity in mouse. Cellular analysis showed LY2624587 antibody, but not LY2510924 peptide, down-regulated cell surface CXCR4 and induced hematological tumor cell death; both agents have been shown to inhibit SDF-1/CXCR4 interaction and downstream signaling. In animal models, LY2510924 peptide induced robust, prolonged, dose- and time-dependent WBC and HSC increases in mice and monkeys, whereas LY2624587 antibody induced only moderate, transient increases in monkeys. In clinical trials, similar pharmacodynamic effects were observed in patients with advanced cancer: LY2510924 peptide induced sustained WBC and HSC increases, while LY2624587 antibody induced only minimal, transient WBC changes. These distinct pharmacodynamic effects in two different classes of CXCR4 inhibitors are clinically important and should be carefully considered when designing combination studies with immune checkpoint inhibitors or other agents for cancer therapy.

20.
MAbs ; 9(2): 285-296, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27981884

RESUMEN

A recent report described a novel mechanism of action for an anti-proprotein convertase subtilisin-kexin type 9 (PCSK9) monoclonal antibody (LY3015014, or LY), wherein the antibody has improved potency and duration of action due to the PCSK9 epitope for LY binding. Unlike other antibodies, proteolysis of PCSK9 can occur when LY is bound to PCSK9. We hypothesized that this allowance of PCSK9 cleavage potentially improves LY efficiency through two pathways, namely lack of accumulation of intact PCSK9 and reduced clearance of LY. A quantitative modeling approach is necessary to further understand this novel mechanism of action. We developed a mechanism-based model to characterize the relationship between antibody pharmacokinetics, PCSK9 and LDL cholesterol levels in animals, and used the model to better understand the underlying drivers for the improved efficiency of LY. Simulations suggested that the allowance of cleavage of PCSK9 resulting in a lack of accumulation of intact PCSK9 is the major driver of the improved potency and durability of LY. The modeling reveals that this novel 'proteolysis-permitting' mechanism of LY is a means by which an efficient antibody can be developed with a total antibody dosing rate that is lower than the target production rate. We expect this engineering approach may be applicable to other targets and that the mathematical models presented herein will be useful in evaluating similar approaches.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Modelos Teóricos , Inhibidores de PCSK9 , Animales , LDL-Colesterol/metabolismo , Humanos , Macaca fascicularis , Ratones , Proteolisis/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA