Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
MAbs ; 15(1): 2195517, 2023.
Article in English | MEDLINE | ID: mdl-37074212

ABSTRACT

Single-chain fragment variable (scFv) domains play an important role in antibody-based therapeutic modalities, such as bispecifics, multispecifics and chimeric antigen receptor T cells or natural killer cells. However, scFv domains exhibit lower stability and increased risk of aggregation due to transient dissociation ("breathing") and inter-molecular reassociation of the two domains (VL and VH). We designed a novel strategy, referred to as stapling, that introduces two disulfide bonds between the scFv linker and the two variable domains to minimize scFv breathing. We named the resulting molecules stapled scFv (spFv). Stapling increased thermal stability (Tm) by an average of 10°C. In multiple scFv/spFv multispecifics, the spFv molecules display significantly improved stability, minimal aggregation and superior product quality. These spFv multispecifics retain binding affinity and functionality. Our stapling design was compatible with all antibody variable regions we evaluated and may be widely applicable to stabilize scFv molecules for designing biotherapeutics with superior biophysical properties.


Subject(s)
Antibodies , Immunoglobulin Variable Region , Immunoglobulin Variable Region/chemistry , Immunoglobulin Fragments
2.
Cell Host Microbe ; 31(5): 751-765.e11, 2023 05 10.
Article in English | MEDLINE | ID: mdl-37098341

ABSTRACT

Treating and preventing infections by antimicrobial-resistant bacterial pathogens is a worldwide problem. Pathogens such as Staphylococcus aureus produce an array of virulence determinants, making it difficult to identify single targets for the development of vaccines or monoclonal therapies. We described a human-derived anti-S. aureus monoclonal antibody (mAb)-centyrin fusion protein ("mAbtyrin") that simultaneously targets multiple bacterial adhesins, resists proteolysis by bacterial protease GluV8, avoids Fc engagement by S. aureus IgG-binding proteins SpA and Sbi, and neutralizes pore-forming leukocidins via fusion with anti-toxin centyrins, while maintaining Fc- and complement-mediated functions. Compared with the parental mAb, mAbtyrin protected human phagocytes and boosted phagocyte-mediated killing. The mAbtyrin also reduced pathology, reduced bacterial burden, and protected from different types of infections in preclinical animal models. Finally, mAbtyrin synergized with vancomycin, enhancing pathogen clearance in an animal model of bacteremia. Altogether, these data establish the potential of multivalent mAbs for treating and preventing S. aureus diseases.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Animals , Humans , Staphylococcus aureus , Staphylococcal Infections/drug therapy , Staphylococcal Infections/prevention & control , Staphylococcal Infections/microbiology , Antibodies, Monoclonal/therapeutic use , Phagocytes/metabolism , Leukocidins/metabolism , Leukocidins/therapeutic use
3.
J Pharmacol Exp Ther ; 371(2): 375-384, 2019 11.
Article in English | MEDLINE | ID: mdl-31451508

ABSTRACT

JNJ-64179375 (JNJ-9375) is a recombinant human IgG4 monoclonal antibody engineered to mimic an IgA antibody that was identified in a patient who exhibited markedly prolonged clotting times but without spontaneous bleeding episodes over several years of follow-up. The crystal structure of the JNJ-9375 antigen-binding fragment/thrombin complex showed an almost identical binding mode to that of the patient IgA. In the current study, we characterized the in vitro and in vivo properties of JNJ-9375. Surface plasmon resonance studies demonstrated that JNJ-9375 binds to α-thrombin with high affinity and specificity (K D: 0.8 nM for human thrombin). JNJ-9375 produced concentration-dependent prolongation of in vitro clotting assays in human plasma, including thrombin time (TT), ecarin clotting time, prothrombin time, and activated partial thromboplastin time, with EC2X values of 4.4, 12.4, 172.6, and 202.7 µg/ml, respectively. JNJ-9375 inhibited thrombin-induced platelet aggregation in human plasma with an IC50 value of 52.6 nM (7.8 µg/ml) and produced concentration-dependent prolongation of reaction time tested by thromboelastography. JNJ-9375 pretreatment resulted in dose-dependent reduction in thrombus formation in the rat arteriovenous (AV) shunt model of thrombosis. Robust efficacy was observed at 0.3 mg/kg accompanied by 1.5× of TT. Bleeding was increased at 3 mg/kg in a rat tail transection bleeding model demonstrating a therapeutic index of 10× compared with 1× for apixaban in the same models. Our data suggest that thrombin exosite I inhibition is efficacious against thrombosis in a pretreatment prevention animal model. SIGNIFICANCE STATEMENT: JNJ-9375 is a novel, fully human monoclonal antibody that binds to the exosite I region of thrombin with high affinity and specificity. JNJ-9375 concentration dependently prolonged clotting times and inhibited thrombin-induced platelet aggregation in in vitro assays based on its mechanism of action. In an in vivo rat AV shunt model, JNJ-9375 prevented thrombus formation in a dose-dependent fashion while demonstrating reduced bleeding risk. The present study demonstrated the antithrombotic effects of inhibiting the exosite I region of thrombin when given in a prevention mode in preclinical animal models.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antithrombins/pharmacology , Immunoglobulin G/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Recombinant Proteins/pharmacology , Animals , Antibodies, Monoclonal/metabolism , Antithrombins/metabolism , Blood Platelets/drug effects , Blood Platelets/metabolism , Dose-Response Relationship, Drug , Humans , Immunoglobulin G/metabolism , Macaca fascicularis , Male , Mice , Platelet Aggregation Inhibitors/metabolism , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism
4.
Mol Metab ; 10: 87-99, 2018 04.
Article in English | MEDLINE | ID: mdl-29453154

ABSTRACT

OBJECTIVE: Insulin resistance is a key feature of Type 2 Diabetes (T2D), and improving insulin sensitivity is important for disease management. Allosteric modulation of the insulin receptor (IR) with monoclonal antibodies (mAbs) can enhance insulin sensitivity and restore glycemic control in animal models of T2D. METHODS: A novel human mAb, IRAB-A, was identified by phage screening using competition binding and surface plasmon resonance assays with the IR extracellular domain. Cell based assays demonstrated agonist and sensitizer effects of IRAB-A on IR and Akt phosphorylation, as well as glucose uptake. Lean and diet-induced obese mice were used to characterize single-dose in vivo pharmacological effects of IRAB-A; multiple-dose IRAB-A effects were tested in obese mice. RESULTS: In vitro studies indicate that IRAB-A exhibits sensitizer and agonist properties distinct from insulin on the IR and is translated to downstream signaling and function; IRAB-A bound specifically and allosterically to the IR and stabilized insulin binding. A single dose of IRAB-A given to lean mice rapidly reduced fed blood glucose for approximately 2 weeks, with concomitant reduced insulin levels suggesting improved insulin sensitivity. Phosphorylated IR (pIR) from skeletal muscle and liver were increased by IRAB-A; however, phosphorylated Akt (pAkt) levels were only elevated in skeletal muscle and not liver vs. control; immunochemistry analysis (IHC) confirmed the long-lived persistence of IRAB-A in skeletal muscle and liver. Studies in diet-induced obese (DIO) mice with IRAB-A reduced fed blood glucose and insulinemia yet impaired glucose tolerance and led to protracted insulinemia during a meal challenge. CONCLUSION: Collectively, the data suggest IRAB-A acts allosterically on the insulin receptor acting non-competitively with insulin to both activate the receptor and enhance insulin signaling. While IRAB-A produced a decrease in blood glucose in lean mice, the data in DIO mice indicated an exacerbation of insulin resistance; these data were unexpected and suggested the interplay of complex unknown pharmacology. Taken together, this work suggests that IRAB-A may be an important tool to explore insulin receptor signaling and pharmacology.


Subject(s)
Allosteric Site , Antibodies, Monoclonal/pharmacology , Hypoglycemic Agents/pharmacology , Receptor, Insulin/agonists , 3T3 Cells , Allosteric Regulation , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Blood Glucose/metabolism , Cell Line, Tumor , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/immunology , Insulin/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Receptor, Insulin/chemistry , Receptor, Insulin/immunology , Signal Transduction
5.
Immunol Lett ; 197: 1-8, 2018 05.
Article in English | MEDLINE | ID: mdl-29476755

ABSTRACT

In therapeutic antibody discovery and early development, mice and cynomolgus monkey are used as animal models to assess toxicity, efficacy and other properties of candidate molecules. As more candidate antibodies are based on human immunoglobulin (IgG) subclasses, many strategies are pursued to simulate the human system in the test animal. However, translation rate from a successful preclinical trial to an approved drug is extremely low. This may partly be due to differences in interaction of human IgG based candidate molecules to endogenous Fcγ receptors of model animals in comparison to those of human Fcγ receptors. In this study, we compare binding characteristics of human IgG subclasses commonly used in drug development (IgG1, IgG2, IgG4) and their respective Fc silent versions (IgG1σ, IgG2σ, IgG4 PAA) to human, mouse, and cynomolgus monkey Fcγ receptors. To control interactions between Fab and Fc domains, the test IgGs all have the same variable region sequences. We found distinct variations of interaction of human IgG subclasses to model animal Fcγ receptors in comparison to their human counterparts. Particularly, cynomolgus monkey Fcγ receptors showed consistently tighter binding to human IgGs than human Fcγ receptors. Moreover, the presumably Fc silent human IgG4 PAA framework bound to cynomolgus monkey FcγRI with nanomolar affinity while only very weak binding was observed for the human FcγRI. Our results highlighted the need for a thorough in vitro affinity characterization of candidate IgGs against model animal Fcγ receptors and careful design of preclinical studies.


Subject(s)
Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/metabolism , Immunoglobulin Isotypes/metabolism , Immunotherapy/methods , Receptors, IgG/metabolism , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Immunoglobulin G/genetics , Immunoglobulin G/therapeutic use , Immunoglobulin Variable Region/genetics , Macaca fascicularis , Mice , Protein Binding , Translational Research, Biomedical
6.
Diabetes ; 66(1): 206-217, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27797911

ABSTRACT

A hallmark of type 2 diabetes is impaired insulin receptor (IR) signaling that results in dysregulation of glucose homeostasis. Understanding the molecular origins and progression of diabetes and developing therapeutics depend on experimental models of hyperglycemia, hyperinsulinemia, and insulin resistance. We present a novel monoclonal antibody, IRAB-B, that is a specific, potent IR antagonist that creates rapid and long-lasting insulin resistance. IRAB-B binds to the IR with nanomolar affinity and in the presence of insulin efficiently blocks receptor phosphorylation within minutes and is sustained for at least 3 days in vitro. We further confirm that IRAB-B antagonizes downstream signaling and metabolic function. In mice, a single dose of IRAB-B induces rapid onset of hyperglycemia within 6 h, and severe hyperglycemia persists for 2 weeks. IRAB-B hyperglycemia is normalized in mice treated with exendin-4, suggesting that this model can be effectively treated with a GLP-1 receptor agonist. Finally, a comparison of IRAB-B with the IR antagonist S961 shows distinct antagonism in vitro and in vivo. IRAB-B appears to be a powerful tool to generate both acute and chronic insulin resistance in mammalian models to elucidate diabetic pathogenesis and evaluate therapeutics.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Insulin Resistance/physiology , Receptor, Insulin/metabolism , Animals , Blotting, Western , Cell Line , Diabetes Mellitus, Type 2 , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Mice , Mice, Inbred C57BL , Peptides/pharmacology , Phosphorylation , Protein Binding , Receptor, Insulin/antagonists & inhibitors , Signal Transduction/drug effects
7.
Proteins ; 84(4): 427-34, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26800003

ABSTRACT

Microtubule-associated protein tau becomes abnormally phosphorylated in Alzheimer's disease and other tauopathies and forms aggregates of paired helical filaments (PHF-tau). AT8 is a PHF-tau-specific monoclonal antibody that is a commonly used marker of neuropathology because of its recognition of abnormally phosphorylated tau. Previous reports described the AT8 epitope to include pS202/pT205. Our studies support and extend previous findings by also identifying pS208 as part of the binding epitope. We characterized the phosphoepitope of AT8 through both peptide binding studies and costructures with phosphopeptides. From the cocrystal structure of AT8 Fab with the diphosphorylated (pS202/pT205) peptide, it appeared that an additional phosphorylation at S208 would also be accommodated by AT8. Phosphopeptide binding studies showed that AT8 bound to the triply phosphorylated tau peptide (pS202/pT205/pS208) 30-fold stronger than to the pS202/pT205 peptide, supporting the role of pS208 in AT8 recognition. We also show that the binding kinetics of the triply phosphorylated peptide pS202/pT205/pS208 was remarkably similar to that of PHF-tau. The costructure of AT8 Fab with a pS202/pT205/pS208 peptide shows that the interaction interface involves all six CDRs and tau residues 202-209. All three phosphorylation sites are recognized by AT8, with pT205 acting as the anchor. Crystallization of the Fab/peptide complex under acidic conditions shows that CDR-L2 is prone to unfolding and precludes peptide binding, and may suggest a general instability in the antibody.


Subject(s)
Antibodies, Monoclonal/chemistry , Epitopes/chemistry , Immunoglobulin Fab Fragments/chemistry , Phosphopeptides/chemistry , tau Proteins/chemistry , Amino Acid Sequence , Antibodies, Monoclonal/biosynthesis , Binding Sites, Antibody , Crystallography, X-Ray , Epitope Mapping , Epitopes/metabolism , Gene Expression , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/biosynthesis , Models, Molecular , Phosphopeptides/chemical synthesis , Phosphorylation , Protein Binding , Protein Folding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry , Serine/metabolism , Threonine/chemistry , Threonine/metabolism , tau Proteins/genetics , tau Proteins/metabolism
8.
Monoclon Antib Immunodiagn Immunother ; 34(6): 373-80, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26683175

ABSTRACT

CCL22 inactivation in vivo occurs by cleavage at the N-terminus; however, it is unclear whether this encompasses the entire site of CCR4 interaction. CCL17 also binds CCR4 and its function requires binding via two discrete binding sites. Using monoclonal antibodies (MAbs), we report that there are two separate sites on CCL22 that are required for CCR4-mediated function. The CCL22-specific antibodies bind with affinities of 632 ± 297 pM (MC2B7) and 308 ± 43 pM (MAB4391) and neither exhibited detectable binding to CCL17. Both antibodies are comparable in their ability to inhibit CCL22-mediated calcium mobilization; however, competition binding studies demonstrate that MC2B7 and MAB4391 bind to distinct epitopes on CCL22. Both antibodies inhibit function through CCR4, which is demonstrated by loss of ß-arrestin recruitment in a reporter cell line. In both assays, blocking either site independently abolished CCL22 function, suggesting that concurrent engagement of both sites with CCR4 is necessary for function. This is the first demonstration that CCL22 has two distinct binding sites that are required for CCR4 function. These antibodies are valuable tools for better understanding the interaction and function of CCL22 and CCR4 and will potentially help further understanding of the differential outcomes of CCL17 and CCL22 interaction with CCR4.


Subject(s)
Antibodies, Monoclonal/chemistry , Arrestins/immunology , Chemokine CCL22/immunology , Epitopes/immunology , Receptors, CCR4/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Arrestins/genetics , Binding Sites , Binding, Competitive , Cell Line , Chemokine CCL17/genetics , Chemokine CCL17/immunology , Chemokine CCL22/genetics , Dendritic Cells/cytology , Dendritic Cells/immunology , Epitopes/chemistry , Epitopes/genetics , Gene Expression Regulation , Humans , Immunity, Innate , Mice , Protein Binding , Protein Structure, Tertiary , Receptors, CCR4/genetics , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , beta-Arrestins
9.
J Biol Chem ; 290(41): 24689-704, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26260789

ABSTRACT

The efficacy of engaging multiple drug targets using bispecific antibodies (BsAbs) is affected by the relative cell-surface protein levels of the respective targets. In this work, the receptor density values were correlated to the in vitro activity of a BsAb (JNJ-61186372) targeting epidermal growth factor receptor (EGFR) and hepatocyte growth factor receptor (c-MET). Simultaneous binding of the BsAb to both receptors was confirmed in vitro. By using controlled Fab-arm exchange, a set of BsAbs targeting EGFR and c-MET was generated to establish an accurate receptor quantitation of a panel of lung and gastric cancer cell lines expressing heterogeneous levels of EGFR and c-MET. EGFR and c-MET receptor density levels were correlated to the respective gene expression levels as well as to the respective receptor phosphorylation inhibition values. We observed a bias in BsAb binding toward the more highly expressed of the two receptors, EGFR or c-MET, which resulted in the enhanced in vitro potency of JNJ-61186372 against the less highly expressed target. On the basis of these observations, we propose an avidity model of how JNJ-61186372 engages EGFR and c-MET with potentially broad implications for bispecific drug efficacy and design.


Subject(s)
Antibodies, Bispecific/immunology , ErbB Receptors/immunology , ErbB Receptors/metabolism , Gene Expression Regulation , Molecular Targeted Therapy , Proto-Oncogene Proteins c-met/immunology , Proto-Oncogene Proteins c-met/metabolism , Antigens, Surface/chemistry , Antigens, Surface/genetics , Antigens, Surface/immunology , Antigens, Surface/metabolism , Cell Line, Tumor , ErbB Receptors/chemistry , ErbB Receptors/genetics , Humans , Immunoglobulin Fab Fragments/immunology , Models, Molecular , Mutation , Phosphorylation , Protein Multimerization , Protein Structure, Quaternary , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Protein Eng Des Sel ; 27(6): 199-206, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24742503

ABSTRACT

CCL17 is a homeostatic chemokine associated with several human inflammatory pathologies. This makes CCL17 a potential point of intervention in inflammatory diseases. Using a Fab-pIX phage display system we were able to select antibodies that specifically bind to CCL17 and neutralize CCL17-mediated signaling. Many of the selected antibodies belong to the VH1-69 germline gene family. The VH1-69 germline gene is represented at a high frequency in the human antibody repertoire and is seen in the early immune response to a variety of pathogens. The heavy chain CDR2 of this germline gene is notably hydrophobic and can insert into hydrophobic pockets of antigens, providing much of the binding energy for these antibodies. Affinity maturation of our primary binders by light chain mutagenesis produced antibodies with sub-nanomolar affinities, with affinity improvements up to 100-fold. These were screened for non-specific protein-protein interactions as a filter for solubility. All of our high affinity antibodies were found to have high levels of non-specific protein-protein interactions. We speculated that this was due to the hydrophobicity within the germline heavy chain CDR1 and CDR2. To ameliorate this problem, we generated a phage display library for one of the clones, where the surface-exposed residues within H-CDR1 and H-CDR2 were randomized. High stringency panning of this library against human CCL17 resulted in further affinity improvement, along with reduction in protein-protein interaction in some new variants. In addition, we improved the cross-reactivity to cynomolgus CCL17. We demonstrate that affinity maturation through targeted libraries in the VH1-69 germline gene can improve both affinity and biophysical characteristics of antibodies derived from this gene scaffold.


Subject(s)
Chemokine CCL17/immunology , Immunoglobulin Heavy Chains/genetics , Animals , Antibody Affinity , Calcium Signaling , Cell Line , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Immunoglobulin G/isolation & purification , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/isolation & purification , Macaca fascicularis , Peptide Library , Protein Binding , Protein Engineering
11.
Methods ; 65(1): 114-26, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23872058

ABSTRACT

The Fc variant of IgG2, designated as IgG2σ, was engineered with V234A/G237A /P238S/H268A/V309L/A330S/P331S substitutions to eliminate affinity for Fcγ receptors and C1q complement protein and consequently, immune effector functions. IgG2σ was compared to other previously well-characterized Fc 'muted' variants, including aglycosylated IgG1, IgG2m4 (H268Q/V309L/A330S/P331S, changes to IgG4), and IgG4 ProAlaAla (S228P/L234A/L235A) in its capacity to bind FcγRs and activate various immune-stimulatory responses. In contrast to the previously characterized muted Fc variants, which retain selective FcγR binding and effector functions, IgG2σ shows no detectable binding to the Fcγ receptors in affinity and avidity measurements, nor any detectable antibody-dependent cytotoxicity, phagocytosis, complement activity, or Fc-mediated cytokine release. Moreover, IgG2σ shows minimal immunogenic potential by T-cell epitope analysis. The circulating half-life of IgG2σ in monkeys is extended relative to IgG1 and IgG2, in spite of similar in vitro binding to recombinant FcRn. The three-dimensional structure of the Fc, needed for assessing the basis for the absence of effector function, was compared with that of IgG2 revealing a number of conformational differences near the hinge region of the CH2 domain that result from the amino acid substitutions. Modeling reveals that at least one of the key interactions with FcγRs is disrupted by a conformational change that reorients P329 to a position that prevents it from interacting with conserved W90 and W113 residues of the FcγRs. Inspection of the structure also indicated significant changes to the conformations of D270 and P329 in the CH2 domain that could negatively impact C1q binding. Thus, structural perturbations of the Fc provide a rationale for the loss of function. In toto, these properties of IgG2σ suggest that it is a superior alternative to previously described IgG variants of minimal effector function, for future therapeutic applications of non-immunostimulatory mAb and Fc-fusion platforms.


Subject(s)
Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry , Immunologic Factors/chemistry , Amino Acid Substitution , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Antibody Affinity , Antibody-Dependent Cell Cytotoxicity , Binding Sites , Crystallography, X-Ray , Cytokines/metabolism , HEK293 Cells , Half-Life , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin G/genetics , Immunoglobulin G/pharmacology , Immunologic Factors/genetics , Immunologic Factors/pharmacology , Macaca fascicularis , Male , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Secondary , Receptor, ErbB-2/immunology , Receptors, IgG/chemistry
12.
Protein Eng Des Sel ; 25(10): 531-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22915597

ABSTRACT

Some antibodies have a tendency to self-associate leading to precipitation at relatively low concentrations. CNTO607, a monoclonal antibody, precipitates irreversibly in phosphate-buffered saline at concentrations above 13 mg/ml. Previous mutagenesis work based on the Fab crystal structure pinpointed a three residue fragment in the heavy chain CDR-3, (99)FHW(100a), as an aggregation epitope that is anchored by two salt bridges. Biophysical characterization of variants reveals that F99 and W100a, but not H100, contribute to the intermolecular interaction. A K210T/K215T mutant designed to disrupt the charge interactions in the aggregation model yielded an antibody that does not precipitate but forms reversible aggregates. An isotype change from IgG1 to IgG4 prevents the antibody from precipitating at low concentration yet the solution viscosity is elevated. To further understand the nature of the antibody self-association, studies on the Fab fragment found high solubility but significant self- and cross-interactions remain. Dynamic light scattering data provides evidence for higher order Fab structure at increased concentrations. Our results provide direct support for the aggregation model that CNTO607 precipitation results primarily from the specific interaction of the Fab arms of neighboring antibodies followed by the development of an extensive network of antibodies inducing large-scale aggregation and precipitation.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Interleukin-13/immunology , Animals , Antibodies, Monoclonal/genetics , Cell Line , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Immunoglobulin G/genetics , Models, Molecular , Mutagenesis , Mutation , Protein Conformation , Solubility
13.
J Mol Recognit ; 25(3): 136-46, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22407977

ABSTRACT

In the human adaptation and optimization of a mouse anti-human respiratory syncytial virus neutralizing antibody, affinity assessment was crucial to distinguish among potential candidates and to evaluate whether this correlated with function in vitro and in vivo. This affinity assessment was complicated by the trimeric nature of the antigen target, respiratory syncytial virus F (RSV-F) glycoprotein. In the initial affinity screen, surface plasmon resonance was used to determine the intrinsic binding affinities of anti-RSV-F Fab and immunoglobulin G (IgG) to the extracellular domain of RSV-F. This assessment required minimal biotinylation of the RSV-F protein and design of a capture strategy to minimize avidity effects. Approximately 30 Fabs were selected from three optimization phage display libraries on the basis of an initial ELISA screen. Surface plasmon resonance analysis demonstrated the success of optimization with some candidates from the screened libraries having low picomolar dissociation constants, more than 700-fold tighter than the parental monoclonal antibody (B21M). The affinities of these antibodies were further evaluated by a kinetic exclusion assay, a solution binding technology. One IgG (monoclonal antibody 029) displayed a low picomolar K(D) comparable with that of motavizumab, an RSV antibody in clinical study. Kinetic exclusion assay showed that two other of the matured IgGs (011 and 019) had sub-picomolar dissociation constants that could not be resolved further. We discuss the relevance of these interaction analysis results in the light of recently published data on the mechanism of F-driven viral fusion during paramyxoviral infection and 101F epitope conservation revealed from the recent crystal structure of RSV-F in the post-fusion state.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/chemistry , Antibodies, Neutralizing/chemistry , Antibody Affinity , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/immunology , Animals , Biotinylation , Humans , Hydrogen-Ion Concentration , Immobilized Proteins/chemistry , Immobilized Proteins/immunology , Immunoglobulin Fab Fragments/chemistry , Kinetics , Mice , Peptide Library , Protein Binding , Protein Structure, Quaternary , Protein Structure, Secondary , Viral Fusion Proteins/chemistry
14.
J Mol Recognit ; 25(3): 184-91, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22407982

ABSTRACT

Interleukin 13 (IL-13) is a pleiotropic cytokine secreted by activated T cells. Both IL-13 and its polymorphic variant (IL-13-R110Q) have been shown to be associated with multiple diseases such as asthma and allergy. Two IL-13 receptors have been identified, IL-13R alpha-1 receptor (IL-13Rα1) and IL-13R alpha-2 receptor (IL-13Rα2). It has been well established that IL-13 binds to IL-13Rα1 alone with low nM affinity while binding to the IL-13Rα1/IL-4R receptor complex is significantly tighter (pM). The affinity between IL-13 and IL-13Rα2, however, remains elusive. Several values have been reported in the literature varying from 20 pM to 2.5 nM. The affinities previously reported were obtained using surface plasmon resonance (SPR) or Scatchard analysis of (125) I-IL-13 binding data. This report presents the results for the kinetics and equilibrium binding analysis studies performed using label-free kinetic exclusion assay (KEA) for the interaction of human IL-13 and IL-13Rα2. KEA equilibrium analysis showed that the affinities of IL-13Rα2 are 107 and 56 pM for IL-13 and its variant (IL-13-R110Q), respectively. KEA kinetic analysis showed that a tight and very stable complex is formed between IL-13Rα2 and IL-13, as shown by calculated dissociation rate constants slower than 5 × 10(-5) per second. Kinetic analysis also showed significant differences in the kinetic behavior of wild type (wt) versus IL-13-R110Q. IL-13-R110Q not only associates to IL-13Rα2 slower than wt human IL-13 (wt-IL-13), as previously reported, but IL-13-R110Q also dissociates slower than wt-IL-13. These results show that IL-13Rα2 is a high affinity receptor and provide a new perspective on kinetic behavior that could have significant implications in the understanding of the role of IL-13-R110Q in the disease state.


Subject(s)
Interleukin-13 Receptor alpha2 Subunit/chemistry , Interleukin-13/chemistry , Amino Acid Substitution , Humans , Immobilized Proteins/chemistry , Interleukin-13/genetics , Interleukin-13 Receptor alpha2 Subunit/isolation & purification , Kinetics , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Surface Plasmon Resonance
15.
J Mol Biol ; 402(5): 797-812, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20691190

ABSTRACT

Interleukin (IL)-12 and IL-23 are heterodimeric proinflammatory cytokines that share a common p40 subunit, paired with p35 and p19 subunits, respectively. They represent an attractive class of therapeutic targets for the treatment of psoriasis and other immune-mediated diseases. Ustekinumab is a fully human monoclonal antibody (mAb) that binds specifically to IL-12/IL-23p40 and neutralizes human IL-12 and IL-23 bioactivity. The crystal structure of ustekinumab Fab (antigen binding fragment of mAb), in complex with human IL-12, has been determined by X-ray crystallography at 3.0 Å resolution. Ustekinumab Fab binds the D1 domain of the p40 subunit in a 1:1 ratio in the crystal, consistent with a 2 cytokines:1 mAb stoichiometry, as measured by isothermal titration calorimetry. The structure indicates that ustekinumab binds to the same epitope on p40 in both IL-12 and IL-23 with identical interactions. Mutational analyses confirm that several residues identified in the IL-12/IL-23p40 epitope provide important molecular binding interactions with ustekinumab. The electrostatic complementarity between the mAb antigen binding site and the p40 D1 domain epitope appears to play a key role in antibody/antigen recognition specificity. Interestingly, this structure also reveals significant structural differences in the p35 subunit and p35/p40 interface, compared with the published crystal structure of human IL-12, suggesting unusual and potentially functionally relevant structural flexibility of p35, as well as p40/p35 recognition. Collectively, these data describe unique observations about IL-12p35 and ustekinumab interactions with p40 that account for its dual binding and neutralization of IL-12 and IL-23.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Immunologic Factors/chemistry , Immunologic Factors/metabolism , Interleukin-12/metabolism , Interleukin-23/metabolism , Amino Acid Substitution/genetics , Antibodies, Monoclonal, Humanized , Calorimetry , Crystallography, X-Ray , Epitopes/genetics , Epitopes/immunology , Humans , Interleukin-12/genetics , Interleukin-23/genetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Quaternary , Ustekinumab
16.
Protein Eng Des Sel ; 23(8): 643-51, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20543007

ABSTRACT

Protein aggregation is of great concern to pharmaceutical formulations and has been implicated in several diseases. We engineered an anti-IL-13 monoclonal antibody CNTO607 for improved solubility. Three structure-based engineering approaches were employed in this study: (i) modifying the isoelectric point (pI), (ii) decreasing the overall surface hydrophobicity and (iii) re-introducing an N-linked carbohydrate moiety within a complementarity-determining region (CDR) sequence. A mutant was identified with a modified pI that had a 2-fold improvement in solubility while retaining the binding affinity to IL-13. Several mutants with decreased overall surface hydrophobicity also showed moderately improved solubility while maintaining a similar antigen affinity. Structural studies combined with mutagenesis data identified an aggregation 'hot spot' in heavy-chain CDR3 (H-CDR3) that contains three residues ((99)FHW(100a)). The same residues, however, were found to be essential for high affinity binding to IL-13. On the basis of the spatial proximity and germline sequence, we reintroduced the consensus N-glycosylation site in H-CDR2 which was found in the original antibody, anticipating that the carbohydrate moiety would shield the aggregation 'hot spot' in H-CDR3 while not interfering with antigen binding. Peptide mapping and mass spectrometric analysis revealed that the N-glycosylation site was generally occupied. This variant showed greatly improved solubility and bound to IL-13 with affinity similar to CNTO607 without the N-linked carbohydrate. All three engineering approaches led to improved solubility and adding an N-linked carbohydrate to the CDR was the most effective route for enhancing the solubility of CNTO607.


Subject(s)
Antibodies, Monoclonal/chemistry , Protein Conformation , Protein Engineering/methods , Protein Stability , Amino Acid Sequence , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Binding Sites , Calorimetry, Differential Scanning , Electrophoresis, Polyacrylamide Gel , Humans , Hydrophobic and Hydrophilic Interactions , Interleukin-13/antagonists & inhibitors , Interleukin-13/metabolism , Isoelectric Focusing , Isoelectric Point , Models, Molecular , Molecular Sequence Data , Peptide Mapping , Protein Multimerization , Solubility , Temperature
17.
MAbs ; 2(4): 428-39, 2010.
Article in English | MEDLINE | ID: mdl-20519961

ABSTRACT

We prepared and characterized golimumab (CNTO148), a human IgG1 tumor necrosis factor alpha (TNFα) antagonist monoclonal antibody chosen for clinical development based on its molecular properties. Golimumab was compared with infliximab, adalimumab and etanercept for affinity and in vitro TNFα neutralization. The affinity of golimumab for soluble human TNFα, as determined by surface plasmon resonance, was similar to that of etanercept (18 pM versus 11 pM), greater than that of infliximab (44 pM) and significantly greater than that of adalimumab (127 pM, p=0.018).  The concentration of golimumab necessary to neutralize TNFα-induced E-selectin expression on human endothelial cells by 50% was significantly less than those for infliximab (3.2 fold; p=0.017) and adalimumab (3.3-fold; p=0.008) and comparable to that for etanercept. The conformational stability of golimumab was greater than that of infliximab (primary melting temperature [Tm] 74.8 °C vs. 69.5 °C) as assessed by differential scanning calorimetry.  In addition, golimumab showed minimal aggregation over the intended shelf life when formulated as a high concentration liquid product (100 mg/mL) for subcutaneous administration.  In vivo, golimumab at doses of 1 and 10 mg/kg significantly delayed disease progression in a mouse model of human TNFα-induced arthritis when compared with untreated mice, while infliximab was effective only at 10 mg/kg. Golimumab also significantly reduced histological scores for arthritis severity and cartilage damage, as well as serum levels of pro-inflammatory cytokines and chemokines associated with arthritis. Thus, we have demonstrated that golimumab is a highly stable human monoclonal antibody with high affinity and capacity to neutralize human TNFα in vitro and in vivo.


Subject(s)
Antibodies, Monoclonal/pharmacology , Arthritis/immunology , Cartilage/drug effects , Immunoglobulin G/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Adalimumab , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal, Humanized/pharmacology , Antibody Affinity , Arthritis/chemically induced , Cartilage/pathology , Disease Models, Animal , Disease Progression , E-Selectin/genetics , E-Selectin/metabolism , Etanercept , Gene Expression Regulation/drug effects , Hybridomas , Immunoglobulin G/isolation & purification , Inflammation Mediators/metabolism , Infliximab , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Protein Conformation , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor-alpha/immunology
18.
Anal Biochem ; 382(1): 66-8, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18675772

ABSTRACT

Monoclonal antibodies are a major subclass of biopharmaceuticals. They are structurally different from other biopharmaceuticals in size and quaternary structure. Here we demonstrate a correlation between chemical stability of antibodies and thermal stability. We show that overall thermal protein stability can be predicted based on the measurement of free sulfhydryl (-SH) content on applying mildly denaturing conditions. We propose that this method can be adapted to a high-throughput screening format and used either as an absolute measure of thermal stability or for ranking a panel of possible variants.


Subject(s)
Antibodies, Monoclonal/chemistry , Sulfhydryl Compounds/analysis , Sulfhydryl Compounds/chemistry , Animals , Antibodies, Monoclonal/metabolism , Calibration , Cattle , Fluorescence , Humans , Protein Denaturation , Transition Temperature
19.
Mol Cell Biol ; 27(9): 3530-41, 2007 May.
Article in English | MEDLINE | ID: mdl-17325029

ABSTRACT

The cyclin-dependent kinase inhibitor p21(Cip1) regulates multiple cellular functions and protects cells from genotoxic and other cellular stresses. Activation of apoptosis signal-regulating kinase 1 (ASK1) induced by inhibition of mTOR signaling leads to sustained phospho-c-Jun that is suppressed in cells with functional p53 or by forced expression of p21(Cip1). Here we show that small deletions of p21(Cip1) around S98 abrogate its association with ASK1 but do not affect binding to Cdk1, hence distinguishing between the cell cycle-regulating functions of p21(Cip1) and its ability to suppress activation of the ASK1/Jun N-terminal protein kinase (JNK) pathway. p21(Cip1) is phosphorylated in vitro by both ASK1 and JNK1 at S98. In vivo phosphorylation of p21(Cip1), predominantly carried out by ASK1, is associated with binding to ASK1 and inactivation of ASK1 kinase function. Binding of p21(Cip1) to ASK1 requires ASK1 kinase function and may involve phosphorylation of S98.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Phosphoserine/metabolism , Anthracenes/pharmacology , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/genetics , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinase 5/genetics , Mutation/genetics , Nuclear Localization Signals , Protein Binding , Protein Kinase Inhibitors/pharmacology , Sirolimus/pharmacology
20.
Hum Antibodies ; 16(3-4): 117-25, 2007.
Article in English | MEDLINE | ID: mdl-18334747

ABSTRACT

The human CCL2 chemokine is implicated in many chronic inflammatory conditions. In the mouse, there are two CCL2 homologues, CCL2 (MCP-1/JE) and CCL12 (MCP-5). Both are potent monocyte chemoattractants and bind to and activate the same receptor, CCR2. The overlapping activities of these chemokines complicate the design of mouse model studies that are intended to mimic human disease. To study the roles of CCL2 and CCL12, we generated neutralizing antibodies specific to each chemokine. Consistent with binding and affinity analyses, the antibodies specifically inhibited CCL2- or CCL12- mediated Ca(2+) mobilization in THP-1 cells. When tested in nude mice bearing human PANC-1 pancreatic tumor cells in Matrigel plugs, CCL2 and CCL12 antibodies potently inhibited tumor angiogenesis, indicating that both CCL2 and CCL12 may contribute to tumor angiogenesis.


Subject(s)
Antibodies/immunology , Chemokine CCL2/immunology , Monocyte Chemoattractant Proteins/immunology , Amino Acid Sequence , Animals , Calcium/metabolism , Cell Line , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Mice, Nude , Molecular Sequence Data , Neovascularization, Pathologic/prevention & control , Neutralization Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...