Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 159: 105492, 2021 11.
Article in English | MEDLINE | ID: mdl-34478849

ABSTRACT

Repulsive guidance molecule A (RGMa) is a potent inhibitor of axonal growth and a regulator of neuronal cell death. It is up-regulated following neuronal injury and accumulates in chronic neurodegenerative diseases. Neutralizing RGMa has the potential to promote neuroregeneration and neuroprotection. Previously we reported that a rat anti-N terminal RGMa (N-RGMa) antibody r5F9 and its humanized version h5F9 (ABT-207) promote neuroprotection and neuroregeneration in preclinical neurodegenerative disease models. However, due to its cross-reactivity to RGMc/hemojuvelin, ABT-207 causes iron accumulation in vivo, which could present a safety liability. Here we report the generation and characterization of a novel RGMa-selective anti-N-RGMa antibody elezanumab, which is currently under Phase 2 clinical evaluation in multiple disease indications. Elezanumab, a human monoclonal antibody generated by in vitro PROfusion mRNA display technology, competes with ABT-207 in binding to N-RGMa but lacks RGMc cross-reactivity with no impact on iron metabolism. It neutralizes repulsive activity of soluble RGMa in vitro and blocks membrane RGMa mediated BMP signaling. In the optic nerve crush and optic neuritis models, elezanumab promotes axonal regeneration and prevents retinal nerve fiber layer degeneration. In the spinal targeted experimental autoimmune encephalomyelitis (EAE) model, elezanumab promotes axonal regeneration and remyelination, decreases inflammatory lesion area and improves functional recovery. Finally, in the mouse cuprizone model, elezanumab reduces demyelination, which is consistent with its inhibitory effect on BMP signaling. Taken together, these preclinical data demonstrate that elezanumab has neuroregenerative and neuroprotective activities without impact on iron metabolism, thus providing a compelling rationale for its clinical development in neurodegenerative diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , GPI-Linked Proteins , Nerve Regeneration , Nerve Tissue Proteins , Neuroprotection , Optic Nerve Injuries , Optic Nerve , Optic Neuritis , Recovery of Function , Retina , Animals , Mice , Cuprizone/toxicity , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/physiopathology , GPI-Linked Proteins/antagonists & inhibitors , Monoamine Oxidase Inhibitors/toxicity , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Nerve Tissue Proteins/antagonists & inhibitors , Neuroprotection/drug effects , Optic Nerve/drug effects , Optic Nerve/physiology , Optic Nerve Injuries/physiopathology , Optic Neuritis/physiopathology , Recovery of Function/drug effects , Recovery of Function/physiology , Retina/drug effects , Surface Plasmon Resonance
2.
J Immunol ; 206(5): 1102-1113, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33495237

ABSTRACT

CTLA4-Ig/abatacept dampens activation of naive T cells by blocking costimulation via CD28. It is an approved drug for rheumatoid arthritis but failed to deliver efficacy in a number of other autoimmune diseases. One explanation is that activated T cells rely less on CD28 signaling and use alternate coreceptors for effector function. ICOS is critical for activation of T-dependent humoral immune responses, which drives pathophysiology of IgG-mediated autoimmune diseases. In this study, we asked whether CD28 and ICOS play nonredundant roles for maintenance of T-dependent responses in mouse models. Using a hapten-protein immunization model, we show that during an ongoing germinal center response, combination treatment with CTLA4-Ig and ICOS ligand (ICOSL) blocking Ab completely dissolves ongoing germinal center responses, whereas single agents show only partial activity. Next, we took two approaches to engineer a therapeutic molecule that blocks both pathways. First, we engineered CTLA4-Ig to enhance binding to ICOSL while retaining affinity to CD80/CD86. Using a library approach, binding affinity of CTLA4-Ig to human ICOSL was increased significantly from undetectable to 15-42 nM; however, the affinity was still insufficient to completely block binding of ICOSL to ICOS. Second, we designed a bispecific costimulation inhibitor with high-affinity CTLA4 extracellular domains fused to anti-ICOSL Ab termed bifunctional costimulation inhibitor. With this bispecific approach, we achieved complete inhibition of CD80 and CD86 binding to CD28 as well as ICOS binding to ICOSL. Such bispecific molecules may provide greater therapeutic benefit in IgG-mediated inflammatory diseases compared with CTLA4-Ig alone.


Subject(s)
CD28 Antigens/metabolism , CTLA-4 Antigen/antagonists & inhibitors , Immune Checkpoint Inhibitors/pharmacology , Inducible T-Cell Co-Stimulator Protein/antagonists & inhibitors , Signal Transduction/drug effects , T-Lymphocytes/drug effects , Abatacept/pharmacology , Animals , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Female , Germinal Center/drug effects , Germinal Center/metabolism , Immunity, Humoral/drug effects , Immunoglobulin G/metabolism , Inducible T-Cell Co-Stimulator Ligand/metabolism , Inflammation/metabolism , Mice , Mice, Inbred C57BL , T-Lymphocytes/metabolism
3.
MAbs ; 9(4): 715-734, 2017.
Article in English | MEDLINE | ID: mdl-28375048

ABSTRACT

Protein glycosylation is arguably the paramount post-translational modification on recombinant glycoproteins, and highly cited in the literature for affecting the physiochemical properties and the efficacy of recombinant glycoprotein therapeutics. Glycosylation of human immunoglobulins follows a reasonably well-understood metabolic pathway, which gives rise to a diverse range of asparagine-linked (N-linked), or serine/threonine-linked (O-linked) glycans. In N-linked glycans, fucose levels have been shown to have an inverse relationship with the degree of antibody-dependent cell-mediated cytotoxicity, and high mannose levels have been implicated in potentially increasing immunogenicity and contributing to less favorable pharmacokinetic profiles. Here, we demonstrate a novel approach to potentially reduce the presence of high-mannose species in recombinant human immunoglobulin preparations, as well as facilitate an approximate 100% replacement of fucosylation with arabinosylation in Chinese hamster ovary cell culture through media supplementation with D-arabinose, an uncommonly used mammalian cell culture sugar substrate. The replacement of fucose with arabinose was very effective and practical to implement, since no cell line engineering or cellular adaptation strategies were required. Arabinosylated recombinant IgGs and the accompanying reduction in high mannose glycans, facilitated a reduction in dendritic cell uptake, increased FcγRIIIa signaling, and significantly increased the levels of ADCC. These aforementioned effects were without any adverse changes to various structural or functional attributes of multiple recombinant human antibodies and a bispecific DVD-Ig. Protein arabinosylation represents an expansion of the N-glycan code in mammalian expressed glycoproteins.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Arabinose/pharmacology , Immunoglobulin G/blood , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibody-Dependent Cell Cytotoxicity , CHO Cells , Cricetulus , Glycosylation/drug effects , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
4.
MAbs ; 9(2): 269-284, 2017.
Article in English | MEDLINE | ID: mdl-27929753

ABSTRACT

Exudative age-related macular degeneration (AMD) is the most common cause of moderate and severe vision loss in developed countries. Intraocular injections of vascular endothelial growth factor (VEGF or VEGF-A)-neutralizing proteins provide substantial benefit, but frequent, long-term injections are needed. In addition, many patients experience initial visual gains that are ultimately lost due to subretinal fibrosis. Preclinical studies and early phase clinical trials suggest that combined suppression of VEGF and platelet-derived growth factor-BB (PDGF-BB) provides better outcomes than suppression of VEGF alone, due to more frequent regression of neovascularization (NV) and suppression of subretinal fibrosis. We generated a dual variable domain immunoglobulin molecule, ABBV642 that specifically and potently binds and neutralizes VEGF and PDGF-BB. ABBV642 has been optimized for treatment of exudative AMD based on the following design characteristics: 1) high affinity binding to all VEGF-A isoforms and both soluble and extracellular matrix (ECM)-associated PDGF-BB; 2) potential for extended residence time in the vitreous cavity to decrease the frequency of intraocular injections; 3) rapid clearance from systemic circulation compared with molecules with wild type Fc region for normal FcRn binding, which may reduce the risk of systemic complications; and 4) low risk of potential effector function. The bispecificity of ABBV642 allows for a single injection of a single therapeutic agent, and thus a more streamlined development and regulatory path compared with combination products. In a mouse model of exudative AMD, ABBV642 was observed to be more effective than aflibercept. ABBV642 has potential to improve efficacy with reduced injection frequency in patients with exudative AMD, thereby reducing the enormous disease burden for patients and society.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Bispecific/pharmacology , Macular Degeneration/drug therapy , Proto-Oncogene Proteins c-sis/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Becaplermin , Female , Humans , Male , Mice , Mice, Transgenic , Protein Engineering , Rabbits
5.
MAbs ; 7(3): 605-19, 2015.
Article in English | MEDLINE | ID: mdl-25764208

ABSTRACT

Interleukin-1 (IL-1) cytokines such as IL-1α, IL-1ß, and IL-1Ra contribute to immune regulation and inflammatory processes by exerting a wide range of cellular responses, including expression of cytokines and chemokines, matrix metalloproteinases, and nitric oxide synthetase. IL-1α and IL-1ß bind to IL-1R1 complexed to the IL-1 receptor accessory protein and induce similar physiological effects. Preclinical and clinical studies provide significant evidence for the role of IL-1 in the pathogenesis of osteoarthritis (OA), including cartilage degradation, bone sclerosis, and synovial proliferation. Here, we describe the generation and characterization of ABT-981, a dual variable domain immunoglobulin (DVD-Ig) of the IgG1/k subtype that specifically and potently neutralizes IL-1α and IL-1ß. In ABT-981, the IL-1ß variable domain resides in the outer domain of the DVD-Ig, whereas the IL-1α variable domain is located in the inner position. ABT-981 specifically binds to IL-1α and IL-1ß, and is physically capable of binding 2 human IL-1α and 2 human IL-1ß molecules simultaneously. Single-dose intravenous and subcutaneous pharmacokinetics studies indicate that ABT-981 has a half-life of 8.0 to 10.4 d in cynomolgus monkey and 10.0 to 20.3 d in rodents. ABT-981 exhibits suitable drug-like-properties including affinity, potency, specificity, half-life, and stability for evaluation in human clinical trials. ABT-981 offers an exciting new approach for the treatment of OA, potentially addressing both disease modification and symptom relief as a disease-modifying OA drug.


Subject(s)
Antibodies, Neutralizing/chemistry , Immunoglobulin G/chemistry , Immunoglobulin Variable Region/chemistry , Interleukin-1alpha/antagonists & inhibitors , Interleukin-1beta/antagonists & inhibitors , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Antibody Affinity , Antibody Specificity , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Immunoglobulin Variable Region/immunology , Immunoglobulin Variable Region/pharmacology , Interleukin-1alpha/chemistry , Interleukin-1alpha/immunology , Interleukin-1beta/chemistry , Interleukin-1beta/immunology , Mice
6.
MAbs ; 5(4): 595-607, 2013.
Article in English | MEDLINE | ID: mdl-23774760

ABSTRACT

Efficient production of large quantities of therapeutic antibodies is becoming a major goal of the pharmaceutical industry. We developed a proprietary expression system using a polyprotein precursor-based approach to antibody expression in mammalian cells. In this approach, the coding regions for heavy and light chains are included within a single open reading frame (sORF) separated by an in-frame intein gene. A single mRNA and subsequent polypeptide are produced upon transient and stable transfection into HEK293 and CHO cells, respectively. Heavy and light chains are separated by the autocatalytic action of the intein and antibody processing proceeds to produce active, secreted antibody. Here, we report advances in sORF technology toward establishment of a viable manufacturing platform for therapeutic antibodies in CHO cells. Increasing expression levels and improving antibody processing by intein and signal peptide selection are discussed.


Subject(s)
Gene Expression , Genetic Vectors/genetics , Inteins , Open Reading Frames , Single-Chain Antibodies , Animals , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/biosynthesis , Immunoglobulin Light Chains/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/genetics
7.
Clin Immunol ; 131(2): 308-16, 2009 May.
Article in English | MEDLINE | ID: mdl-19188093

ABSTRACT

The TNF antagonists adalimumab, infliximab, and etanercept are effective treatments for rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, and psoriasis, but only adalimumab and infliximab have been found to be efficacious in Crohn's disease. The present studies evaluated the TNF-binding and complement-activating properties of adalimumab, infliximab, and etanercept to determine whether these properties may explain differences in their clinical efficacy profiles. Association and dissociation rates of binding to soluble TNF were measured by surface plasmon resonance, and were found to be similar for adalimumab, infliximab, and etanercept, as were their calculated binding affinities. Avidity of binding to soluble TNF, measured by KinExA technology, was 10- to 20-fold greater for soluble etanercept (K(D)=0.4 picomolars [pM]) than for soluble adalimumab or infliximab (K(D)=8.6 and 4.2 pM, respectively). (125)I-adalimumab, -infliximab, and -etanercept bound to membrane TNF (mTNF) on mTNF-transfected cells with similar affinities (K(D)=483, 468, and 445 pM, respectively) that were each lower than for soluble TNF. Complement-dependent cytotoxicity (CDC) was induced in mTNF-transfected cells by adalimumab and infliximab, but was not induced in activated normal human PBMC by any of the 3 agents. In conclusion, the binding properties of adalimumab, infliximab, and etanercept were similar for soluble TNF, and very similar for mTNF, yet none of the 3 was able to induce CDC in activated PBMC. These results suggest that the different clinical efficacy profiles of these agents are not explained by differences in either TNF-intrinsic binding properties or complement lysis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antibody Affinity/drug effects , Cell Membrane/immunology , Complement Activation/drug effects , Tumor Necrosis Factor-alpha/metabolism , Adalimumab , Anti-Inflammatory Agents/metabolism , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Etanercept , Humans , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , Infliximab , Kinetics , Membrane Proteins/metabolism , Protein Binding/drug effects , Receptors, Tumor Necrosis Factor/metabolism , Solubility , Tumor Necrosis Factor-alpha/antagonists & inhibitors
8.
MAbs ; 1(4): 339-47, 2009.
Article in English | MEDLINE | ID: mdl-20068402

ABSTRACT

Signal transduction through the interleukin-1 receptor (IL-1R) pathway mediates a strong pro-inflammatory response, which contributes to a number of human diseases such as rheumatoid arthritis. Within the IL-1 family, IL-1alpha and IL-1beta are both agonistic ligands for IL-1R, whereas IL-1 receptor antagonist (IL-1ra) is an endogenous antagonist that binds to IL-R, but does not signal. Therefore, the ideal therapeutic strategy would be blocking both IL-1alpha and IL-1beta, but not IL-1ra. However, due to low sequence homology between the three members of the family, it has been exceedingly difficult to identify potent therapeutic agents, e.g., monoclonal antibodies (mAbs), that selectively recognize both IL-1alpha and IL-1beta, but not IL-1ra. Currently, several anti-IL-1 therapeutic agents in clinical development either inhibit only IL-1beta (i.e., anti-IL-1beta mAb), or recognize all three ligands (i.e., anti-IL-1R mAb or IL-1R Trap). We have recently developed a novel dual variable domain immunoglobulin (or DVD-Ig) technology that enables engineering the distinct specificities of two mAbs into a single functional, dual-specific, tetravalent IgG-like molecule. Based on this approach, we have developed anti-human IL-1alpha/beta DVD-Ig molecules using several pairs of monoclonal antibodies with therapeutic potential, and present a case study for optimal design of a DVD-Ig agent for a specific target pair combination.


Subject(s)
Drug Design , Immunoglobulin Variable Region/genetics , Interleukin-1alpha/antagonists & inhibitors , Interleukin-1beta/antagonists & inhibitors , Models, Molecular , Protein Engineering , Electrophoresis, Polyacrylamide Gel , Humans , Immunoglobulin Variable Region/chemistry , Molecular Weight
9.
Nat Biotechnol ; 25(11): 1290-7, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17934452

ABSTRACT

For complex diseases in which multiple mediators contribute to overall disease pathogenesis by distinct or redundant mechanisms, simultaneous blockade of multiple targets may yield better therapeutic efficacy than inhibition of a single target. However, developing two separate monoclonal antibodies for clinical use as combination therapy is impractical, owing to regulatory hurdles and cost. Multi-specific, antibody-based molecules have been investigated; however, their therapeutic use has been hampered by poor pharmacokinetics, stability and manufacturing feasibility. Here, we describe a generally applicable model of a dual-specific, tetravalent immunoglobulin G (IgG)-like molecule--termed dual-variable-domain immunoglobulin (DVD-Ig)--that can be engineered from any two monoclonal antibodies while preserving activities of the parental antibodies. This molecule can be efficiently produced from mammalian cells and exhibits good physicochemical and pharmacokinetic properties. Preclinical studies of a DVD-Ig protein in an animal disease model demonstrate its potential for therapeutic application in human diseases.


Subject(s)
Antibodies, Bispecific/biosynthesis , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/biosynthesis , Arthritis, Experimental/drug therapy , Immunoglobulin Variable Region/biosynthesis , Protein Engineering , Animals , Antibodies, Bispecific/pharmacokinetics , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Arthritis, Experimental/pathology , CHO Cells , Cricetinae , Cricetulus , Disease Models, Animal , Humans , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Immunoglobulin Variable Region/therapeutic use , Interleukin-12/antagonists & inhibitors , Interleukin-12/immunology , Interleukin-18/antagonists & inhibitors , Interleukin-18/immunology , Mice , Protein Structure, Tertiary , Rats
10.
J Biol Chem ; 279(11): 10593-605, 2004 Mar 12.
Article in English | MEDLINE | ID: mdl-14676205

ABSTRACT

Here we identified two novel proteins denoted EH domain protein 2 (EHD2) and EHD2-binding protein 1 (EHBP1) that link clathrin-mediated endocytosis to the actin cytoskeleton. EHD2 contains an N-terminal P-loop and a C-terminal EH domain that interacts with NPF repeats in EHBP1. Disruption of EHD2 or EHBP1 function by small interfering RNA-mediated gene silencing inhibits endocytosis of transferrin into EEA1-positive endosomes as well as GLUT4 endocytosis into cultured adipocytes. EHD2 localizes with cortical actin filaments, whereas EHBP1 contains a putative actin-binding calponin homology domain. High expression of EHD2 or EHBP1 in intact cells mediates extensive actin reorganization. Thus EHD2 appears to connect endocytosis to the actin cytoskeleton through interactions of its N-terminal domain with membranes and its C-terminal EH domain with the novel EHBP1 protein.


Subject(s)
Actins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/physiology , Cytoskeleton/metabolism , Endocytosis , 3T3-L1 Cells , Actins/chemistry , Adipocytes/metabolism , Amino Acid Sequence , Animals , Biological Transport , Blotting, Northern , Blotting, Western , COS Cells , Carrier Proteins/metabolism , Cell Membrane/metabolism , DNA, Complementary/metabolism , Endosomes/metabolism , Gene Silencing , Glucose/metabolism , Green Fluorescent Proteins , Humans , Immunohistochemistry , Luminescent Proteins/metabolism , Mice , Microscopy, Electron , Microscopy, Fluorescence , Models, Biological , Models, Genetic , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Rats , Rhodamines/chemistry , Time Factors , Tissue Distribution , Transfection , Transferrin/chemistry , Transferrin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...