Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Arthritis Res Ther ; 17: 234, 2015 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-26335795

RESUMEN

INTRODUCTION: CD40 ligand (CD40L) blockade has demonstrated efficacy in experimental autoimmune models. However, clinical trials of hu5c8, an anti-human CD40L IgG1 antibody, in systemic lupus erythematosus (SLE) were halted due to an increased incidence of thrombotic events. This study evaluated CDP7657, a high affinity PEGylated monovalent Fab' anti-CD40L antibody fragment, to assess whether an Fc-deficient molecule retains efficacy while avoiding the increased risk of thrombotic events observed with hu5c8. METHODS: The potency and cross-reactivity of CDP7657 was assessed in in vitro assays employing human and non-human primate leukocytes, and the capacity of different antibody formats to activate platelets in vitro was assessed using aggregometry and dense granule release assays. Given the important role CD40L plays in regulating humoral immunity, in vivo efficacy was assessed by investigating the capacity of Cynomolgus monkeys to generate immune responses to the tetanus toxoid antigen while the potential to induce thrombotic events in vivo was evaluated after repeat dosing of antibodies to Rhesus monkeys. A PEGylated anti-mouse CD40L was generated to assess efficacy in the New Zealand Black/White (NZB/W) mouse model of SLE. RESULTS: CDP7657 dose-dependently inhibited antigen-specific immune responses to tetanus toxoid in Cynomolgus monkeys, and in contrast to hu5c8, there was no evidence of pulmonary thrombovasculopathy in Rhesus monkeys. Aglycosyl hu5c8, which lacks Fc receptor binding function, also failed to induce thrombotic events in Rhesus monkeys. In vitro experiments confirmed that antibody constructs lacking an Fc, including CDP7657, did not induce human or monkey platelet activation. A PEGylated monovalent Fab' anti-mouse CD40L antibody also inhibited disease activity in the NZB/W mouse model of SLE after administration using a therapeutic dosing regimen where mice received antibodies only after they had displayed severe proteinuria. CONCLUSIONS: These findings demonstrate for the first time that anti-CD40L antibodies lacking a functional Fc region do not induce thrombotic events in Rhesus monkeys and fail to activate platelets in vitro but, nevertheless retain pharmacological activity and support the investigation of CDP7657 as a potential therapy for systemic lupus erythematosus and other autoimmune diseases.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Ligando de CD40/inmunología , Inmunidad Humoral/inmunología , Trombosis/inmunología , Animales , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacología , Formación de Anticuerpos/inmunología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/prevención & control , Modelos Animales de Enfermedad , Humanos , Inmunidad Humoral/efectos de los fármacos , Fragmentos Fab de Inmunoglobulinas/inmunología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/prevención & control , Macaca fascicularis , Macaca mulatta , Ratones Endogámicos NZB , Polietilenglicoles/química , Toxoide Tetánico/inmunología , Trombosis/inducido químicamente
2.
FASEB J ; 28(11): 4764-78, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25070367

RESUMEN

The blood-brain barrier (BBB) prevents the access of therapeutic antibodies to central nervous system (CNS) targets. The engineering of bispecific antibodies in which a therapeutic "arm" is combined with a BBB-transcytosing arm can significantly enhance their brain delivery. The BBB-permeable single-domain antibody FC5 was previously isolated by phenotypic panning of a naive llama single-domain antibody phage display library. In this study, FC5 was engineered as a mono- and bivalent fusion with the human Fc domain to optimize it as a modular brain delivery platform. In vitro studies demonstrated that the bivalent fusion of FC5 with Fc increased the rate of transcytosis (Papp) across brain endothelial monolayer by 25% compared with monovalent fusion. Up to a 30-fold enhanced apparent brain exposure (derived from serum and cerebrospinal fluid pharmacokinetic profiles) of FC5- compared with control domain antibody-Fc fusions after systemic dosing in rats was observed. Systemic pharmacological potency was evaluated in the Hargreaves model of inflammatory pain using the BBB-impermeable neuropeptides dalargin and neuropeptide Y chemically conjugated with FC5-Fc fusion proteins. Improved serum pharmacokinetics of Fc-fused FC5 contributed to a 60-fold increase in pharmacological potency compared with the single-domain version of FC5; bivalent and monovalent FC5 fusions with Fc exhibited similar systemic pharmacological potency. The study demonstrates that modular incorporation of FC5 as the BBB-carrier arm in bispecific antibodies or antibody-drug conjugates offers an avenue to develop pharmacologically active biotherapeutics for CNS indications.


Asunto(s)
Anticuerpos Biespecíficos/metabolismo , Productos Biológicos/metabolismo , Barrera Hematoencefálica/metabolismo , Animales , Anticuerpos Biespecíficos/inmunología , Transporte Biológico/fisiología , Encéfalo/metabolismo , Humanos , Inmunoconjugados/metabolismo , Masculino , Ingeniería de Proteínas/métodos , Ratas Wistar , Proteínas Recombinantes de Fusión/metabolismo
4.
Methods Mol Biol ; 763: 383-401, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21874466

RESUMEN

The neonatal Fc receptor, FcRn, mediates endocytic recycling pathway that prevents degradation of IgG and is expressed in most endothelial cells. The blood-brain barrier (BBB), formed by brain endothelial cells sealed with tight junctions, restricts transport of IgG from the blood to the brain. In contrast, it has been suggested that IgG undergoes efflux from the brain parenchyma via reverse transcytosis across the BBB mediated by FcRn. The fast elimination of therapeutic antibodies from the brain via this route may limit their therapeutic potency. In vitro and in vivo methods described in this chapter were developed to facilitate research into mechanisms and dynamics of brain efflux of compounds, including FcRn-mediated reverse transcytosis across the BBB. The in vitro model uses immortalized adult rat brain endothelial cells which express high levels of FcRn. In vivo models use Prospective optical imaging to measure the clearance rate of intracerebrally injected FcRn-transported molecules tagged with near-infrared fluorescent probes.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Imagenología Tridimensional/métodos , Inmunoglobulina G/metabolismo , Imagen Molecular/métodos , Receptores Fc/metabolismo , Transcitosis/fisiología , Animales , Astrocitos/citología , Astrocitos/metabolismo , Encéfalo/citología , Línea Celular , Células Endoteliales/citología , Endotelio Vascular/citología , Ensayo de Inmunoadsorción Enzimática , Colorantes Fluorescentes/análisis , Colorantes Fluorescentes/metabolismo , Semivida , Cinética , Masculino , Ratas , Ratas Wistar , Programas Informáticos
5.
J Pharmacol Exp Ther ; 339(2): 519-29, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21807883

RESUMEN

LINGO-1 (leucine-rich repeat and Ig domain containing NOGO receptor interacting protein-1) is a negative regulator of myelination and repair of damaged axons in the central nervous system (CNS). Blocking LINGO-1 function leads to robust remyelination. The anti-LINGO-1 Li81 antibody is currently being evaluated in clinical trials for multiple sclerosis (MS) and is the first MS therapy that directly targets myelin repair. LINGO-1 is selectively expressed in brain and spinal cord but not in peripheral tissues. Perhaps the greatest concern for Li81 therapy is the limited access of the drug to the CNS. Here, we measured Li81 concentrations in brain, spinal cord, and cerebral spinal fluid in rats after systemic administration and correlated them with dose-efficacy responses in rat lysolecithin and experimental autoimmune encephalomyelitis spinal cord models of remyelination. Remyelination was dose-dependent, and levels of Li81 in spinal cord that promoted myelination correlated well with affinity measurements for the binding of Li81 to LINGO-1. Observed Li81 concentrations in the CNS of 0.1 to 0.4% of blood levels are consistent with values reported for other antibodies. To understand the features of the antibody that affect CNS penetration, we also evaluated the pharmacokinetics of Li81 Fab2, Fab, and poly(ethylene glycol)-modified Fab. The reagents all showed similar CNS exposure despite large differences in their sizes, serum half-lives, and volumes of distribution, and area under the curve (AUC) measurements in the CNS directly correlated with AUC measurements in serum. These studies demonstrate that exposure levels achieved by passive diffusion of the Li81 monoclonal antibody into the CNS are sufficient and lead to robust remyelination.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/inmunología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/inmunología , Médula Espinal/efectos de los fármacos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Líquido Cefalorraquídeo/química , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Lisofosfatidilcolinas , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Regeneración , Médula Espinal/metabolismo , Médula Espinal/patología
6.
MAbs ; 3(4): 362-75, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21697654

RESUMEN

TWEAK, a TNF family ligand with pleiotropic cellular functions, was originally described as capable of inducing tumor cell death in vitro. TWEAK functions by binding its receptor, Fn14, which is up-regulated on many human solid tumors. Herein, we show that intratumoral administration of TWEAK, delivered either by an adenoviral vector or in an immunoglobulin Fc-fusion form, results in significant inhibition of tumor growth in a breast xenograft model. To exploit the TWEAK-Fn14 pathway as a therapeutic target in oncology, we developed an anti-Fn14 agonistic antibody, BIIB036. Studies described herein show that BIIB036 binds specifically to Fn14 but not other members of the TNF receptor family, induces Fn14 signaling, and promotes tumor cell apoptosis in vitro. In vivo, BIIB036 effectively inhibits growth of tumors in multiple xenograft models, including colon (WiDr), breast (MDA-MB-231), and gastric (NCI-N87) tumors, regardless of tumor cell growth inhibition response observed to BIIB036 in vitro. The anti-tumor activity in these cell lines is not TNF-dependent. Increasing the antigen-binding valency of BIB036 significantly enhances its anti-tumor effect, suggesting the contribution of higher order cross-linking of the Fn14 receptor. Full Fc effector function is required for maximal activity of BIIB036 in vivo, likely due to the cross-linking effect and/or ADCC mediated tumor killing activity. Taken together, the anti-tumor properties of BIIB036 validate Fn14 as a promising target in oncology and demonstrate its potential therapeutic utility in multiple solid tumor indications.


Asunto(s)
Antineoplásicos , Apoptosis/efectos de los fármacos , Neoplasias/terapia , Receptores del Factor de Necrosis Tumoral/agonistas , Factores de Necrosis Tumoral/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Antineoplásicos/administración & dosificación , Antineoplásicos/inmunología , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Citocina TWEAK , Células HT29 , Humanos , Ligandos , Ratones , Neoplasias/inmunología , Unión Proteica , Receptores del Factor de Necrosis Tumoral/inmunología , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptor de TWEAK , Factores de Necrosis Tumoral/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Pharm Biomed Anal ; 55(1): 168-75, 2011 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-21300512

RESUMEN

Natalizumab is a humanized IgG4 monoclonal antibody which binds human α4 integrin and is approved for treatment of multiple sclerosis and Crohn's disease. Assessment of the in vivo disposition of natalizumab presents a unique assay development challenge due to the ability of human IgG4 antibodies to undergo half-antibody exchange in vivo. Such exchange generates IgG4 molecules of mixed specificity comprising a natalizumab heavy-light chain pair coupled to an IgG4 heavy-light chain pair of unknown specificity. Since exchanged and non-exchanged species cannot be quantified independently using a single enzyme linked immunosorbent assay (ELISA), a novel quantitation strategy was developed employing two ELISAs: one measuring total natalizumab including both intact and exchanged molecules, and the second measuring only intact natalizumab. The presence and amount of exchanged natalizumab in serum is calculated by the difference in values obtained in the two assays. To evaluate assay performance, a control reagent was created from natalizumab and an irrelevant humanized monoclonal IgG4 antibody. Subsequent validation demonstrated that both assays are specific, accurate, and precise within the working ranges of the assays (1.5-10µg/mL for total and 0.5-12µg/mL for intact natalizumab assays). The mean accuracy, intra- and inter-assay precision for both assays were 82-113%, ≤9% and ≤20%, respectively. Additionally, the limits of detection of intact and exchanged natalizumab were established using statistical methods. The utility of the two-assay strategy was confirmed by analyzing samples from a pharmacokinetic study in rats using different variants of natalizumab administered along with another human IgG4 antibody as an exchange partner.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Inmunoensayo/métodos , Inmunoglobulina G/metabolismo , Región Variable de Inmunoglobulina/metabolismo , Integrina alfa4/metabolismo , Algoritmos , Animales , Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados , Enfermedad de Crohn/tratamiento farmacológico , Humanos , Inmunoglobulina G/sangre , Región Variable de Inmunoglobulina/inmunología , Límite de Detección , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Proteínas Mutantes/metabolismo , Proteínas Mutantes/farmacocinética , Natalizumab , Replegamiento Proteico , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Tecnología Farmacéutica
8.
Bioconjug Chem ; 22(2): 200-10, 2011 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-21254764

RESUMEN

The use of LINGO-1 antagonists to promote repair of damaged myelin is an emerging therapeutic opportunity for treatment of CNS diseases caused by demyelination such as multiple sclerosis. The Li33 anti-LINGO-1 antibody is a potent inducer of myelination in vitro and in vivo, but aggregation issues prevented the engineering of an optimal development candidate. PEGylated Li33 Fab' is one of several versions of the Li33 antibody that is being investigated in an attempt to identify the most favorable anti-LINGO-1 antibody design. For targeted PEGylation, a Li33 Fab' construct was engineered with a single unpaired cysteine in the heavy-chain hinge sequence. The Fab' was expressed in CHO cells, purified, and PEGylated with 20 kDa methoxy-poly(ethylene glycol) maleimide using a reaction strategy optimized to improve the yield of the PEG-Fab'. Biochemical analysis of the Li33 PEG-Fab' verified the selectivity of the PEGylation reaction. The in vitro and in vivo attributes of the PEG-Fab' were benchmarked against a Li33 full antibody. Both the Li33 PEG-Fab' and intact antibody bound LINGO-1 with nanomolar affinity, promoted myelination in an in vitro signaling assay, and promoted the repair of damaged myelin in the rat lysolecithin model. These studies extend our understanding of the biological activity of the Li33 mAb and validate the use of an anti-LINGO-1 PEG-Fab' for treatment of CNS diseases caused by demyelination.


Asunto(s)
Anticuerpos Monoclonales/química , Fragmentos Fab de Inmunoglobulinas/química , Polietilenglicoles/química , Animales , Anticuerpos Monoclonales/inmunología , Células CHO , Cricetinae , Cricetulus , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/aislamiento & purificación , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Modelos Animales , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Ratas
9.
Biotechnol Appl Biochem ; 57(1): 31-45, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20815818

RESUMEN

NgRI (Nogo-66 receptor) is part of a signalling complex that inhibits axon regeneration in the central nervous system. Truncated soluble versions of NgRI have been used successfully to promote axon regeneration in animal models of spinal-cord injury, raising interest in this protein as a potential therapeutic target. The LRR (leucine-rich repeat) regions in NgRI are flanked by N- and C-terminal disulfide-containing 'cap' domains (LRRNT and LRRCT respectively). In the present work we show that, although functionally active, the NgRI(310)-Fc fusion protein contains mislinked and heterogeneous disulfide patterns in the LRRCT domain, and we report the generation of a series of variant molecules specifically designed to prevent this heterogeneity. Using these variants we explored the effects of modifying the NgRI truncation site or the spacing between the NgRI and Fc domains, or replacing cysteines within the NgRI or IgG hinge regions. One variant, which incorporates replacements of Cys²66 and Cys³°9 with alanine residues, completely eliminated disulfide scrambling while maintaining functional in vitro and in vivo efficacy. This modified NgRI-Fc molecule represents a significantly improved candidate for further pharmaceutical development, and may serve as a useful model for the optimization of other IgG fusion proteins made from LRR proteins.


Asunto(s)
Disulfuros/metabolismo , Proteínas de la Mielina/química , Ingeniería de Proteínas/métodos , Receptores de Superficie Celular/química , Proteínas Recombinantes de Fusión/química , Secuencia de Aminoácidos , Animales , Conducta Animal/efectos de los fármacos , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Electroforesis en Gel de Poliacrilamida , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/genética , Humanos , Masculino , Anotación de Secuencia Molecular , Datos de Secuencia Molecular , Proteínas de la Mielina/genética , Receptor Nogo 1 , Estabilidad Proteica , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Raíces Nerviosas Espinales/lesiones
10.
J Clin Invest ; 120(8): 2767-81, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20628202

RESUMEN

Studies in mice and humans have revealed that the T cell, immunoglobulin, mucin (TIM) genes are associated with several atopic diseases. TIM-1 is a type I membrane protein that is expressed on T cells upon stimulation and has been shown to modulate their activation. In addition to a recently described interaction with dendritic cells, TIM-1 has also been identified as a phosphatidylserine recognition molecule, and several protein ligands have been proposed. Our understanding of its activity is complicated by the possibility that TIM-1 possesses multiple and diverse binding partners. In order to delineate the function of TIM-1, we generated monoclonal antibodies directed to a cleft formed within the IgV domain of TIM-1. We have shown here that antibodies that bind to this defined cleft antagonize TIM-1 binding to specific ligands and cells. Notably, these antibodies exhibited therapeutic activity in a humanized SCID model of experimental asthma, ameliorating inflammation, and airway hyperresponsiveness. Further experiments demonstrated that the effects of the TIM-1-specific antibodies were mediated via suppression of Th2 cell proliferation and cytokine production. These results demonstrate that modulation of the TIM-1 pathway can critically influence activated T cells in a humanized disease model, suggesting that TIM-1 antagonists may provide potent therapeutic benefit in asthma and other immune-mediated disorders.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Asma/prevención & control , Glicoproteínas de Membrana/antagonistas & inhibidores , Proteínas de la Membrana/antagonistas & inhibidores , Receptores Virales/antagonistas & inhibidores , Animales , Asma/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Receptor Celular 1 del Virus de la Hepatitis A , Humanos , Activación de Linfocitos , Glicoproteínas de Membrana/fisiología , Proteínas de la Membrana/fisiología , Ratones , Ratones SCID , Fosfatidilserinas/metabolismo , Receptores Virales/fisiología
11.
J Immunol ; 185(3): 1577-83, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20585032

RESUMEN

Anti-CD40L immunotherapy in systemic lupus erythematosus patients was associated with thromboembolism of unknown cause. We previously showed that monoclonal anti-CD40L immune complexes (ICs) activated platelets in vitro via the IgG receptor (FcgammaRIIa). In this study, we examined the prothrombotic effects of anti-CD40L ICs in vivo. Because mouse platelets lack FcgammaRIIa, we used FCGR2A transgenic mice. FCGR2A mice were injected i.v. with preformed ICs consisting of either anti-human CD40L mAb (M90) plus human CD40L, or a chimerized anti-mouse CD40L mAb (hMR1) plus mouse CD40L. ICs containing an aglycosylated form of hMR1, which does not bind FcgammaRIIa, were also injected. M90 IC caused shock and thrombocytopenia in FCGR2A but not in wild-type mice. Animals injected with hMR1 IC also experienced these effects, whereas those injected with aglycosylated-hMR1 IC did not, demonstrating that anti-CD40L IC-induced platelet activation in vivo is FcgammaRIIa-dependent. Sequential injections of individual IC components caused similar effects, suggesting that ICs were able to assemble in circulation. Analysis of IC-injected mice revealed pulmonary thrombi consisting of platelet aggregates and fibrin. Mice pretreated with a thrombin inhibitor became moderately thrombocytopenic in response to anti-CD40L ICs and had pulmonary platelet-thrombi devoid of fibrin. In conclusion, we have shown for the first time that anti-CD40L IC-induced thrombosis can be replicated in mice transgenic for FcgammaRIIa. This molecular mechanism may be important for understanding thrombosis associated with CD40L immunotherapy. The FCGR2A mouse model may also be useful for assessing the hemostatic safety of other therapeutic Abs.


Asunto(s)
Complejo Antígeno-Anticuerpo/fisiología , Autoanticuerpos/toxicidad , Ligando de CD40/inmunología , Activación Plaquetaria/inmunología , Receptores de IgG/genética , Trombosis/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/toxicidad , Complejo Antígeno-Anticuerpo/administración & dosificación , Complejo Antígeno-Anticuerpo/toxicidad , Autoanticuerpos/administración & dosificación , Autoanticuerpos/uso terapéutico , Humanos , Hibridomas , Ratones , Ratones Noqueados , Ratones Transgénicos , Activación Plaquetaria/genética , Receptores de IgG/deficiencia , Receptores de IgG/fisiología , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/fisiología , Proteínas Recombinantes de Fusión/toxicidad , Trombosis/sangre
12.
Protein Sci ; 19(5): 954-66, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20198683

RESUMEN

Monoclonal antibodies (Mabs) are a favorite drug platform of the biopharmaceutical industry. Currently, over 20 Mabs have been approved and several hundred others are in clinical trials. The anti-LINGO-1 Mab Li33 was selected from a large panel of antibodies by Fab phage display technology based on its extraordinary biological activity in promoting oligodendrocyte differentiation and myelination in vitro and in animal models of remyelination. However, the Li33 Fab had poor solubility when converted into a full antibody in an immunoglobulin G1 framework. A detailed analysis of the biochemical and structural features of the antibody revealed several possible reasons for its propensity to aggregate. Here, we successfully applied three molecular approaches (isotype switching, targeted mutagenesis of complementarity determining region residues, and glycosylation site insertion mutagenesis) to address the solubility problem. Through these efforts we were able to improve the solubility of the Li33 Mab from 0.3 mg/mL to >50 mg/mL and reduce aggregation to an acceptable level. These strategies can be readily applied to other proteins with solubility issues.


Asunto(s)
Anticuerpos Monoclonales/química , Inmunoglobulina G/química , Proteínas de la Membrana/inmunología , Mutagénesis Sitio-Dirigida/métodos , Proteínas del Tejido Nervioso/inmunología , Ingeniería de Proteínas/métodos , Secuencia de Aminoácidos , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Área Bajo la Curva , Cristalografía por Rayos X , Electroforesis en Gel de Poliacrilamida , Cambio de Clase de Inmunoglobulina , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/metabolismo , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Estabilidad Proteica , Solubilidad , Temperatura
13.
J Biol Chem ; 284(47): 32686-94, 2009 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-19783658

RESUMEN

Misincorporation of amino acids in proteins expressed in Escherichia coli has been well documented but not in proteins expressed in mammalian cells under normal recombinant protein production conditions. Here we report for the first time that Ser can be incorporated at Asn positions in proteins expressed in Chinese hamster ovary cells. This misincorporation was discovered as a result of intact mass measurement, peptide mapping analysis, and tandem mass spectroscopy sequencing. Our analyses showed that the substitution was not related to specific protein molecules or DNA codons and was not site-specific. We believe that the incorporation of Ser at sites coded for Asn was due to mischarging of tRNA(Asn) rather than to codon misreading. The rationale for substitution of Asn by Ser and not by other amino acids is also discussed. Further investigation indicated that the substitution was due to the starvation for Asn in the cell culture medium and that the substitution could be limited by using the Asn-rich feed. These observations demonstrate that the quality of expressed proteins should be closely monitored when altering cell culture conditions.


Asunto(s)
Asparagina/química , Proteínas Recombinantes/química , Serina/química , Animales , Células CHO , Clonación Molecular , Cricetinae , Cricetulus , Escherichia coli/metabolismo , Glicosilación , Espectrometría de Masas/métodos , Modelos Biológicos , Mapeo Peptídico , Péptidos/química , ARN de Transferencia/metabolismo
14.
J Biol Chem ; 284(15): 10254-67, 2009 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-19211557

RESUMEN

Therapeutic antibodies directed against the type 1 insulin-like growth factor receptor (IGF-1R) have recently gained significant momentum in the clinic because of preliminary data generated in human patients with cancer. These antibodies inhibit ligand-mediated activation of IGF-1R and the resulting down-stream signaling cascade. Here we generated a panel of antibodies against IGF-1R and screened them for their ability to block the binding of both IGF-1 and IGF-2 at escalating ligand concentrations (>1 microm) to investigate allosteric versus competitive blocking mechanisms. Four distinct inhibitory classes were found as follows: 1) allosteric IGF-1 blockers, 2) allosteric IGF-2 blockers, 3) allosteric IGF-1 and IGF-2 blockers, and 4) competitive IGF-1 and IGF-2 blockers. The epitopes of representative antibodies from each of these classes were mapped using a purified IGF-1R library containing 64 mutations. Most of these antibodies bound overlapping surfaces on the cysteine-rich repeat and L2 domains. One class of allosteric IGF-1 and IGF-2 blocker was identified that bound a separate epitope on the outer surface of the FnIII-1 domain. Using various biophysical techniques, we show that the dual IGF blockers inhibit ligand binding using a spectrum of mechanisms ranging from highly allosteric to purely competitive. Binding of IGF-1 or the inhibitory antibodies was associated with conformational changes in IGF-1R, linked to the ordering of dynamic or unstructured regions of the receptor. These results suggest IGF-1R uses disorder/order within its polypeptide sequence to regulate its activity. Interestingly, the activity of representative allosteric and competitive inhibitors on H322M tumor cell growth in vitro was reflective of their individual ligand-blocking properties. Many of the antibodies in the clinic likely adopt one of the inhibitory mechanisms described here, and the outcome of future clinical studies may reveal whether a particular inhibitory mechanism leads to optimal clinical efficacy.


Asunto(s)
Epítopos/química , Receptores de Somatomedina/química , Sitio Alostérico , Animales , Células CHO , Rastreo Diferencial de Calorimetría , Cricetinae , Cricetulus , Mapeo Epitopo , Humanos , Factor II del Crecimiento Similar a la Insulina/química , Cinética , Ligandos , Conformación Molecular , Receptor IGF Tipo 1/metabolismo
15.
MAbs ; 1(2): 128-41, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20061822

RESUMEN

Bispecific antibodies (BsAbs) represent an emerging class of biologics that achieve dual targeting with a single agent. Recombinant DNA technologies have facilitated a variety of creative bispecific designs with many promising therapeutic applications; however, practical methods for producing high quality BsAbs that have good product stability, long serum half-life, straightforward purification, and scalable production have largely been limiting. Here we describe a protein-engineering approach for producing stable, scalable tetravalent IgG-like BsAbs. The stability-engineered IgG-like BsAb was envisioned to target and crosslink two TNF family member receptors, TRAIL-R2 (TNF-Related Apoptosis Inducing Ligand Receptor-2) and LTbetaR (Lymphotoxin-beta Receptor), expressed on the surface of epithelial tumor cells with the goal of triggering an enhanced anti-tumor effect. Our IgG-like BsAbs consists of a stability-engineered anti-LTbetaR single chain Fv (scFv) genetically fused to either the N- or C-terminus of the heavy chain of a fulllength anti-TRAIL-R2 IgG1 monoclonal antibody. Both N- or C-terminal BsAbs were active in inhibiting tumor cell growth in vitro, and with some cell lines demonstrated enhanced activity relative to the combination of parental Abs. Pharmacokinetic studies in mice revealed long serum half-lives for the BsAbs. In murine tumor xenograft models, therapeutic treatment with the BsAbs resulted in reduction in tumor volume either comparable to or greater than the combination of parental antibodies, indicating that simultaneously targeting and cross-linking receptor pairs is an effective strategy for treating tumor cells. These studies support that stability-engineering is an enabling step for producing scalable IgG-like BsAbs with properties desirable for biopharmaceutical development.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Receptor beta de Linfotoxina/inmunología , Neoplasias/terapia , Ingeniería de Proteínas/métodos , Estabilidad Proteica , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/genética , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Especificidad de Anticuerpos , Línea Celular Tumoral , Proliferación Celular , Humanos , Receptor beta de Linfotoxina/genética , Ratones , Ratones SCID , Modelos Moleculares , Datos de Secuencia Molecular , Neoplasias/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Anticuerpos de Cadena Única/química , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Biochem Biophys Res Commun ; 377(1): 215-20, 2008 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-18835250

RESUMEN

Cripto is a cell surface protein highly expressed in certain solid tumors, and overexpression of Cripto protein is oncogenic. Cripto-1 protein is encoded by CRIPTO1 gene. CRIPTO3, a presumed pseudogene, has an open reading frame with six amino acid differences from Cripto-1. We show that CRIPTO3 mRNA is the CRIPTO message expressed in many cancer samples. A CRIPTO3 SAGE tag was found in several cancer SAGE libraries, while the CRIPTO1 tag was found in ES cell libraries. In vitro experiments indicate both Cripto-1 and Cripto-3 proteins are functional in the Nodal-dependent signal pathway. Our data indicate that CRIPTO3 is an expressed gene, particularly in certain cancers, and suggest a potentially novel mechanism of oncogenesis through activation of a retrogene.


Asunto(s)
Transformación Celular Neoplásica/genética , Factor de Crecimiento Epidérmico/genética , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias , Glicoproteínas de Membrana/genética , Proteínas de Neoplasias/genética , Neoplasias/genética , Seudogenes , Secuencia de Aminoácidos , Línea Celular Tumoral , Proteínas Ligadas a GPI , Humanos , Péptidos y Proteínas de Señalización Intercelular , Datos de Secuencia Molecular , Transcripción Genética
17.
Nat Med ; 13(10): 1228-33, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17906634

RESUMEN

Demyelinating diseases, such as multiple sclerosis, are characterized by the loss of the myelin sheath around neurons, owing to inflammation and gliosis in the central nervous system (CNS). Current treatments therefore target anti-inflammatory mechanisms to impede or slow disease progression. The identification of a means to enhance axon myelination would present new therapeutic approaches to inhibit and possibly reverse disease progression. Previously, LRR and Ig domain-containing, Nogo receptor-interacting protein (LINGO-1) has been identified as an in vitro and in vivo negative regulator of oligodendrocyte differentiation and myelination. Here we show that loss of LINGO-1 function by Lingo1 gene knockout or by treatment with an antibody antagonist of LINGO-1 function leads to functional recovery from experimental autoimmune encephalomyelitis. This is reflected biologically by improved axonal integrity, as confirmed by magnetic resonance diffusion tensor imaging, and by newly formed myelin sheaths, as determined by electron microscopy. Antagonism of LINGO-1 or its pathway is therefore a promising approach for the treatment of demyelinating diseases of the CNS.


Asunto(s)
Axones/fisiología , Encefalomielitis Autoinmune Experimental/inducido químicamente , Proteínas de la Membrana/antagonistas & inhibidores , Vaina de Mielina/fisiología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Traumatismos de la Médula Espinal/terapia , Animales , Axones/diagnóstico por imagen , Axones/ultraestructura , Encefalomielitis Autoinmune Experimental/patología , Inyecciones Espinales , Proteínas de la Membrana/administración & dosificación , Proteínas de la Membrana/fisiología , Ratones , Ratones Noqueados , Proteínas de la Mielina , Vaina de Mielina/ultraestructura , Glicoproteína Asociada a Mielina/inmunología , Glicoproteína Asociada a Mielina/farmacología , Glicoproteína Mielina-Oligodendrócito , Proteínas del Tejido Nervioso/administración & dosificación , Proteínas del Tejido Nervioso/fisiología , Ratas , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Tomografía Computarizada por Rayos X
18.
Biochem Biophys Res Commun ; 355(3): 751-7, 2007 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-17321501

RESUMEN

Although the functional properties of IgGs are well known, little has been published concerning the stability of whole IgG molecules. Stability is, however, a requirement for the development of antibodies for therapeutic or diagnostic applications. The hypervariable antigen-binding region (Fv) is responsible for stability variations between IgGs of identical subclass. To determine the range of stabilities that may be expected for human(ized) antibodies, differential scanning calorimetry was performed on 17 human(ized) antibodies from various in-house programs. The antigen-binding fragments (Fabs) of these antibodies exhibited thermal unfolding transitions with midpoints (T(M)s) varying from 57 to 82 degrees C. Antibodies with very low Fab stabilities were found to aggregate and express poorly. Fab instability was often associated with high levels of uncommonly observed amino acids or CDR loop lengths particularly within the variable heavy chain domain. Overall, the study provides a thermostability range for IgGs and suggests possible stability guidelines for developing antibody diagnostics or therapeutics.


Asunto(s)
Fragmentos Fab de Inmunoglobulinas/química , Inmunoglobulina G/química , Región Variable de Inmunoglobulina/química , Rastreo Diferencial de Calorimetría , Calor , Humanos , Inmunoglobulina G/uso terapéutico , Estructura Terciaria de Proteína
19.
Cancer Res ; 66(19): 9617-24, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17018619

RESUMEN

The lymphotoxin-beta receptor (LT beta R) is a tumor necrosis factor receptor family member critical for the development and maintenance of various lymphoid microenvironments. Herein, we show that agonistic anti-LT beta R monoclonal antibody (mAb) CBE11 inhibited tumor growth in xenograft models and potentiated tumor responses to chemotherapeutic agents. In a syngeneic colon carcinoma tumor model, treatment of the tumor-bearing mice with an agonistic antibody against murine LT beta R caused increased lymphocyte infiltration and necrosis of the tumor. A pattern of differential gene expression predictive of cellular and xenograft response to LT beta R activation was identified in a panel of colon carcinoma cell lines and when applied to a panel of clinical colorectal tumor samples indicated 35% likelihood a tumor response to CBE11. Consistent with this estimate, CBE11 decreased tumor size and/or improved long-term animal survival with two of six independent orthotopic xenografts prepared from surgical colorectal carcinoma samples. Targeting of LT beta R with agonistic mAbs offers a novel approach to the treatment of colorectal and potentially other types of cancers.


Asunto(s)
Adenocarcinoma/terapia , Anticuerpos Monoclonales/uso terapéutico , Neoplasias del Colon/terapia , Receptor beta de Linfotoxina/agonistas , Neoplasias del Cuello Uterino/terapia , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Camptotecina/análogos & derivados , Camptotecina/uso terapéutico , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Terapia Combinada , Sinergismo Farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/uso terapéutico , Inmunoglobulina M/inmunología , Inmunoglobulina M/uso terapéutico , Irinotecán , Linfocitos Infiltrantes de Tumor/inmunología , Receptor beta de Linfotoxina/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Distribución Aleatoria , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Método Simple Ciego , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/patología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Protein Eng Des Sel ; 19(7): 317-24, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16682434

RESUMEN

Monocyte chemoattractant proteins (MCPs) are cytokines that direct immune cells bearing appropriate receptors to sites of inflammation or injury and are therefore attractive therapeutic targets for inhibitory molecules. 11K2 is a blocking mouse monoclonal antibody active against several human and murine MCPs. A 2.5 A structure of the Fab fragment of this antibody in complex with human MCP-1 has been solved. The Fab blocks CCR2 receptor binding to MCP-1 through an adjacent but distinct binding site. The orientation of the Fab indicates that a single MCP-1 dimer will bind two 11K2 antibodies. Several key residues on the antibody and on human MCPs were predicted to be involved in antibody selectivity. Mutational analysis of these residues confirms their involvement in the antibody-chemokine interaction. In addition to mutations that decreased or disrupted binding, one antibody mutation resulted in a 70-fold increase in affinity for human MCP-2. A key residue missing in human MCP-3, a chemokine not recognized by the antibody, was identified and engineering the preferred residue into the chemokine conferred binding to the antibody.


Asunto(s)
Anticuerpos Bloqueadores/farmacología , Citocinas/farmacología , Fragmentos Fab de Inmunoglobulinas/farmacología , Proteínas Quimioatrayentes de Monocitos/farmacología , Receptores de Quimiocina/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Anticuerpos Bloqueadores/inmunología , Sitios de Unión , Quimiocina CCL2/química , Quimiocina CCL2/farmacología , Quimiocina CCL7 , Quimiocina CCL8 , Citocinas/metabolismo , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Inflamación/tratamiento farmacológico , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Quimioatrayentes de Monocitos/química , Mutación , Receptores CCR2 , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/química , Relación Estructura-Actividad , Heridas y Lesiones/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...