Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Cereb Blood Flow Metab ; 38(4): 727-740, 2018 04.
Article in English | MEDLINE | ID: mdl-29140158

ABSTRACT

Current methods for examining antibody trafficking are either non-quantitative such as immunocytochemistry or require antibody labeling with tracers. We have developed a multiplexed quantitative method for antibody 'tracking' in endosomal compartments of brain endothelial cells. Rat brain endothelial cells were co-incubated with blood-brain barrier (BBB)-crossing FC5, monovalent FC5Fc or bivalent FC5Fc fusion antibodies and control antibodies. Endosomes were separated using sucrose-density gradient ultracentrifugation and analyzed using multiplexed mass spectrometry to simultaneously quantify endosomal markers, receptor-mediated transcytosis (RMT) receptors and the co-incubated antibodies in each fraction. The quantitation showed that markers of early endosomes were enriched in high-density fractions (HDF), whereas markers of late endosomes and lysosomes were enriched in low-density fractions (LDF). RMT receptors, including transferrin receptor, showed a profile similar to that of early endosome markers. The in vitro BBB transcytosis rates of antibodies were directly proportional to their partition into early endosome fractions of brain endothelial cells. Addition of the Fc domain resulted in facilitated antibody 'redistribution' from LDF into HDF and additionally into multivesicular bodies (MVB). Sorting of various FC5 antibody formats away from late endosomes and lysosomes and into early endosomes and a subset of MVB results in increased antibody transcytosis at the abluminal side of the BBB.


Subject(s)
Antibodies/metabolism , Blood-Brain Barrier/physiology , Endosomes/physiology , Transcytosis/physiology , Animals , Antibodies/cerebrospinal fluid , Antigens, CD , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Lysosomes/metabolism , Mass Spectrometry , Rats , Rats, Sprague-Dawley , Receptors, Transferrin
2.
Adv Pharmacol ; 71: 301-35, 2014.
Article in English | MEDLINE | ID: mdl-25307221

ABSTRACT

The development and approval of antibody-based therapeutics have progressed rapidly over the past decade. However, poor blood-brain barrier (BBB) permeability hinders the progress of antibody therapies for conditions in which the target is located in the central nervous system (CNS). Increased brain penetration of therapeutic antibodies can be achieved by engineering bispecific antibodies in which one antibody binding specificity recognizes a BBB receptor that undergoes receptor-mediated transcytosis (RMT) from the circulatory compartment into brain parenchyma, and the second binding specificity recognizes a therapeutic target within the CNS. These bispecific antibodies can be built using various antibody fragments as "building blocks," including monomeric single-domain antibodies, the smallest antigen-binding fragments of immunoglobulins. The development of BBB-crossing bispecific antibodies requires targeted antibody engineering to optimize multiple characteristics of "BBB carrier" and therapeutic arms, as well as other antibody properties impacting pharmacokinetics and effector function. Whereas several BBB-crossing bispecific antibodies have been developed using transferrin receptor antibodies as BBB carriers, the principal obstacle for capitalizing on the future promise of CNS-active antibodies remains the scarcity of known, characterized RMT receptors which could be exploited for the development of BBB carriers. This chapter reviews the recent advances and guiding principles for designing, engineering, and evaluating BBB-crossing bispecific antibodies and discusses approaches to identify and characterize novel BBB-crossing antibodies and RMT receptors.


Subject(s)
Antibodies, Bispecific , Blood-Brain Barrier/metabolism , Animals , Antibodies, Bispecific/pharmacokinetics , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Drug Design , Humans , Models, Biological , Permeability
3.
FASEB J ; 28(11): 4764-78, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25070367

ABSTRACT

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.


Subject(s)
Antibodies, Bispecific/metabolism , Biological Products/metabolism , Blood-Brain Barrier/metabolism , Animals , Antibodies, Bispecific/immunology , Biological Transport/physiology , Brain/metabolism , Humans , Immunoconjugates/metabolism , Male , Protein Engineering/methods , Rats, Wistar , Recombinant Fusion Proteins/metabolism
4.
Methods Mol Biol ; 763: 383-401, 2011.
Article in English | MEDLINE | ID: mdl-21874466

ABSTRACT

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.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Histocompatibility Antigens Class I/metabolism , Imaging, Three-Dimensional/methods , Immunoglobulin G/metabolism , Molecular Imaging/methods , Receptors, Fc/metabolism , Transcytosis/physiology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Brain/cytology , Cell Line , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Enzyme-Linked Immunosorbent Assay , Fluorescent Dyes/analysis , Fluorescent Dyes/metabolism , Half-Life , Kinetics , Male , Rats , Rats, Wistar , Software
5.
Protein Sci ; 19(5): 954-66, 2010 May.
Article in English | MEDLINE | ID: mdl-20198683

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunoglobulin G/chemistry , Membrane Proteins/immunology , Mutagenesis, Site-Directed/methods , Nerve Tissue Proteins/immunology , Protein Engineering/methods , Amino Acid Sequence , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Area Under Curve , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Immunoglobulin Class Switching , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Stability , Solubility , Temperature
6.
MAbs ; 1(2): 128-41, 2009.
Article in English | MEDLINE | ID: mdl-20061822

ABSTRACT

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.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Lymphotoxin beta Receptor/immunology , Neoplasms/therapy , Protein Engineering/methods , Protein Stability , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Amino Acid Sequence , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibody Specificity , Cell Line, Tumor , Cell Proliferation , Humans , Lymphotoxin beta Receptor/genetics , Mice , Mice, SCID , Models, Molecular , Molecular Sequence Data , Neoplasms/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Single-Chain Antibodies/chemistry , Xenograft Model Antitumor Assays
7.
Biochemistry ; 45(33): 10117-28, 2006 Aug 22.
Article in English | MEDLINE | ID: mdl-16906770

ABSTRACT

LTbetaR is a member of the TNF receptor family of proteins. It binds to two different cell surface ligands, LIGHT, a homotypic trimer, and LTalpha1beta2, a heterotypic trimer. We have measured the affinities of the dimeric IgG fusion protein, LTbetaRIgG, and monomeric LTbetaR protein binding to both LIGHT and LTalpha1beta2 using surface plasmon resonance and found values of <0.1 and 38 nM for LIGHT and <0.1 and 48 nM for LTalpha1beta2, respectively. We also determined the stoichiometries of binding for both forms of the receptor LTbetaRIgG and LTbetaR binding to LIGHT. The data obtained from several biophysical methods are consistent with receptor polypeptide to trimeric ligand ratios of 2:1. The determined masses of the complexes using SEC-LS corresponded to a single LTbetaRIgG bound to a LIGHT trimer, or two LTbetaR bound per LIGHT. Sedimentation velocity of varied ratios of LTbetaR to a fixed concentration of LIGHT were analyzed by SEDANAL and were successfully fit with a model with two tight binding sites on LIGHT and one poor affinity site. Isothermal calorimetric titration of LIGHT with either LTbetaR or LTbetaRIgG also demonstrated stoichiometries of 1:2 and 1:1, respectively. The binding of LTbetaR to LIGHT was endothermic and, hence, entropy-driven. TNFR p55 (extracellular domain) complexed with the trimeric ligand, TNFbeta, exhibits a 3:1 receptor/ligand stoichiometry. This complex has been used as the prototypical model setting the receptor-ligand complexation paradigm for the entire TNF family. The LTbetaR/LIGHT binding stoichiometry of 2:1 demonstrated here does not fit the paradigm. This has numerous implications for cell biology including signaling requiring only dimerization of LTbetaR rather than trimerization as expected from the structural paradigm.


Subject(s)
Lymphotoxin-alpha/chemistry , Lymphotoxin-alpha/metabolism , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/metabolism , Binding Sites , Calorimetry/methods , Cell Physiological Phenomena , Chromatography, Gel , Dimerization , Electrophoresis, Polyacrylamide Gel , Immunoglobulin G/metabolism , Ligands , Models, Molecular , Molecular Weight , Recombinant Fusion Proteins/metabolism , Surface Plasmon Resonance , Thermodynamics
8.
Biochemistry ; 45(7): 2006-13, 2006 Feb 21.
Article in English | MEDLINE | ID: mdl-16475789

ABSTRACT

The oligomeric state of BAFF (B cell activing factor), a tumor necrosis factor (TNF) family cytokine that plays a critical role in B cell development and survival, has been the subject of recent debate. Myc-tagged BAFF starting at residue Gln136 was previously reported to crystallize as trimers at pH 4.5, whereas a histidine-tagged construct of BAFF, starting at residue Ala134, formed a virus-like cluster containing 60 monomers when crystallized at pH 9.0. The formation of the BAFF 60-mer was pH dependent, requiring pH >or= 7.0. More recently, 60-mer formation was suggested to be artificially induced by the histidine tag, and it was proposed that BAFF, like all other TNF family members, is trimeric. We report here that a construct of BAFF with no amino-terminal tag (Ala134-BAFF) can form a 60-mer in solution. Using size exclusion chromatography and static light scattering to monitor trimer to 60-mer ratios in BAFF preparations, we find that 60-mer formation is pH-dependent and requires histidine 218 within the DE loop of BAFF. Biacore measurements established that the affinity of Ala134-BAFF for the BAFF receptor BAFFR/BR3 is similar to that of myc-Gln136-BAFF, which is exclusively trimeric in solution. However, Ala134-BAFF is more efficacious than myc-Gln136-BAFF in inducing B cell proliferation in vitro. We additionally show that BAFF that is processed and secreted by 293T cells transfected with full-length BAFF, or by a histiocytic lymphoma cell line (U937) that expresses BAFF endogenously, forms a pH-dependent 60-mer in solution. Our results indicate that the formation of the 60-mer in solution by the BAFF extracellular domain is an intrinsic property of the protein, and therefore that this more active form of BAFF may be physiologically relevant.


Subject(s)
Membrane Proteins/physiology , Protein Structure, Quaternary , Tumor Necrosis Factor-alpha/physiology , Animals , B-Cell Activating Factor , Chromatography, Gel , Humans , Hydrogen-Ion Concentration , Light , Mice , Molecular Weight , Pichia/metabolism , Scattering, Radiation
9.
Science ; 310(5750): 1022-5, 2005 Nov 11.
Article in English | MEDLINE | ID: mdl-16284179

ABSTRACT

We have identified a small-molecule inhibitor of tumor necrosis factor alpha (TNF-alpha) that promotes subunit disassembly of this trimeric cytokine family member. The compound inhibits TNF-alpha activity in biochemical and cell-based assays with median inhibitory concentrations of 22 and 4.6 micromolar, respectively. Formation of an intermediate complex between the compound and the intact trimer results in a 600-fold accelerated subunit dissociation rate that leads to trimer dissociation. A structure solved by x-ray crystallography reveals that a single compound molecule displaces a subunit of the trimer to form a complex with a dimer of TNF-alpha subunits.


Subject(s)
Indoles/chemistry , Indoles/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/chemistry , Biotinylation , Chemical Phenomena , Chemistry, Physical , Crystallography, X-Ray , Dimerization , Fluorescence , Hydrogen/chemistry , Hydrophobic and Hydrophilic Interactions , Indoles/chemical synthesis , Kinetics , Mass Spectrometry , Models, Chemical , Models, Molecular , Molecular Conformation , Molecular Structure , Protein Conformation , Protein Subunits/chemistry , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...