Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
J Biol Chem ; 293(25): 9880-9891, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29720399

ABSTRACT

4-1BBL is a member of the tumor necrosis factor (TNF) superfamily and is the ligand for the TNFR superfamily receptor, 4-1BB. 4-1BB plays an immunomodulatory role in T cells and NK cells, and agonists of this receptor have garnered strong attention as potential immunotherapy agents. Broadly speaking, the structural features of TNF superfamily members, their receptors, and ligand-receptor complexes are similar. However, a published crystal structure of human 4-1BBL suggests that it may be unique in this regard, exhibiting a three-bladed propeller-like trimer assembly that is distinctly different from that observed in other family members. This unusual structure also suggests that the human 4-1BB/4-1BBL complex may be structurally unique within the TNF/TNFR superfamily, but to date no structural data have been reported. Here we report the crystal structure of the human 4-1BB/4-1BBL complex at 2.4-Å resolution. In this structure, 4-1BBL does not adopt the unusual trimer assembly previously reported, but instead forms a canonical bell-shaped trimer typical of other TNF superfamily members. The structure of 4-1BB is also largely canonical as is the 4-1BB/4-1BBL complex. Mutational data support the 4-1BBL structure reported here as being biologically relevant, suggesting that the previously reported structure is not. Together, the data presented here offer insight into structure/function relationships in the 4-1BB/4-1BBL system and improve our structural understanding of the TNF/TNFR superfamily more broadly.


Subject(s)
4-1BB Ligand/chemistry , 4-1BB Ligand/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/chemistry , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Binding Sites , Crystallography, X-Ray , HEK293 Cells , Humans , Ligands , Protein Binding , Protein Conformation , Protein Multimerization
2.
J Biol Chem ; 293(22): 8439-8448, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29669810

ABSTRACT

Pathways of human epidermal growth factor (EGF) receptors are activated upon ligand-dependent or -independent homo- or heterodimerization and their subsequent transphosphorylation. Overexpression of these receptors positively correlates with transphosphorylation rates and increased tumor growth rates. MEDI4276, an anti-human epidermal growth factor receptor 2 (HER2) biparatopic antibody-drug conjugate, has two paratopes within each antibody arm. One, 39S, is aiming at the HER2 site involved in receptor dimerization and the second, single chain fragment (scFv), mimicking trastuzumab. Here we present the cocrystal structure of the 39S Fab-HER2 complex and, along with biophysical and functional assays, determine the corresponding epitope of MEDI4276 and its underlying mechanism of action. Our results reveal that MEDI4276's uniqueness is based first on the ability of its 39S paratope to block HER2 homo- or heterodimerization and second on its ability to cluster the receptors on the surface of receptor-overexpressing cells.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Breast Neoplasms/drug therapy , Protein Multimerization , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology , Amino Acid Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Crystallography, X-Ray , Female , Humans , Models, Molecular , Phosphorylation , Protein Conformation , Sequence Homology , Tumor Cells, Cultured
3.
PLoS Genet ; 12(4): e1005895, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27093186

ABSTRACT

Small cell lung cancer (SCLC) is an aggressive disease with poor survival. A few sequencing studies performed on limited number of samples have revealed potential disease-driving genes in SCLC, however, much still remains unknown, particularly in the Asian patient population. Here we conducted whole exome sequencing (WES) and transcriptomic sequencing of primary tumors from 99 Chinese SCLC patients. Dysregulation of tumor suppressor genes TP53 and RB1 was observed in 82% and 62% of SCLC patients, respectively, and more than half of the SCLC patients (62%) harbored TP53 and RB1 mutation and/or copy number loss. Additionally, Serine/Arginine Splicing Factor 1 (SRSF1) DNA copy number gain and mRNA over-expression was strongly associated with poor survival using both discovery and validation patient cohorts. Functional studies in vitro and in vivo demonstrate that SRSF1 is important for tumorigenicity of SCLC and may play a key role in DNA repair and chemo-sensitivity. These results strongly support SRSF1 as a prognostic biomarker in SCLC and provide a rationale for personalized therapy in SCLC.


Subject(s)
Carcinoma, Small Cell/genetics , Lung Neoplasms/genetics , Oncogene Proteins/genetics , Serine-Arginine Splicing Factors/genetics , Adult , Aged , DNA Copy Number Variations , DNA Damage , Female , Gene Silencing , Humans , Male , Middle Aged , Mutation
4.
Angew Chem Int Ed Engl ; 58(25): 8489-8493, 2019 06 17.
Article in English | MEDLINE | ID: mdl-31018033

ABSTRACT

Here, we describe a diene-containing noncanonical amino acid (ncAA) capable of undergoing fast and selective normal electron-demand Diels-Alder (DA) reactions following its incorporation into antibodies. A cyclopentadiene derivative of lysine (CpHK) served as the reactive handle for DA transformations and the substrate for genetic incorporation. CpHK incorporated into antibodies with high efficiency and was available for maleimide conjugation or self-reaction depending on position in the amino acid sequence. CpHK at position K274 reacted with the maleimide drug-linker AZ1508 at a rate of ≈79 m-1 s-1 to produce functional antibody-drug conjugates (ADCs) in a one-step process. Incorporation of CpHK at position S239 resulted in dimerization, which covalently linked antibody heavy chains together. The diene ncAA described here is capable of producing therapeutic protein conjugates with clinically validated and widely available maleimide compounds, while also enabling proximity-based stapling through a DA dimerization reaction.


Subject(s)
Alkadienes/chemistry , Amino Acids/chemistry , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry , Maleimides/chemistry , Cycloaddition Reaction , Dimerization , Humans , Models, Molecular , Molecular Structure
5.
J Biol Chem ; 292(6): 2510-2518, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28011641

ABSTRACT

Selectin interactions with fucosylated glycan ligands mediate leukocyte rolling in the vasculature under shear forces. Crystal structures of P- and E-selectin suggest a two-state model in which ligand binding to the lectin domain closes loop 83-89 around the Ca2+ coordination site, enabling Glu-88 to engage Ca2+ and fucose. This triggers further allostery that opens the lectin/EGF domain hinge. The model posits that force accelerates transition from the bent (low affinity) to the extended (high affinity) state. However, transition intermediates have not been described, and the role of Glu-88 in force-assisted allostery has not been examined. Here we report the structure of the lectin and EGF domains of L-selectin bound to a fucose mimetic; that is, a terminal mannose on an N-glycan attached to a symmetry-related molecule. The structure is a transition intermediate where loop 83-89 closes to engage Ca2+ and mannose without triggering allostery that opens the lectin/EGF domain hinge. We used three complementary assays to compare ligand binding to WT selectins and to E88D selectins that replaced Glu-88 with Asp. Soluble P-selectinE88D bound with an ∼9-fold lower affinity to PSGL-1, a physiological ligand, due to faster dissociation. Adhesion frequency experiments with a biomembrane force probe could not detect interactions of P-selectinE88D with PSGL-1. Cells expressing transmembrane P-selectinE88D or L-selectinE88D detached from immobilized ligands immediately after initiating flow. Cells expressing E-selectinE88D rolled but detached faster. Our data support a two-state model for selectins in which Glu-88 must engage ligand to trigger allostery that stabilizes the high affinity state under force.


Subject(s)
Glutamic Acid/metabolism , L-Selectin/metabolism , Polysaccharides/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray , Humans , L-Selectin/chemistry , Membrane Glycoproteins/metabolism , Protein Conformation
6.
J Biol Chem ; 290(24): 14979-85, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25925951

ABSTRACT

We report the three-dimensional structure of human interferon α-2A (IFN-α2A) bound to the Fab fragment of a therapeutic monoclonal antibody (sifalimumab; IgG1/κ). The structure of the corresponding complex was solved at a resolution of 3.0 Å using molecular replacement and constitutes the first reported structure of a human type I IFN bound to a therapeutic antibody. This study revealed the major contribution made by the first complementarity-determining region in each of sifalimumab light and heavy chains. These data also provided the molecular basis for sifalimumab mechanism of action. We propose that its interferon-neutralizing properties are the result of direct competition for IFN-α2A binding to the IFN receptor subunit 1 (IFNAR1) and do not involve inhibiting IFN-α2A binding to the IFN receptor subunit 2 (IFNAR2).


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Interferon-alpha/drug effects , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Crystallography, X-Ray , Interferon alpha-2 , Interferon-alpha/immunology , Models, Molecular , Protein Conformation , Recombinant Proteins/drug effects , Recombinant Proteins/immunology
7.
J Biol Chem ; 290(7): 4282-90, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25538249

ABSTRACT

The Fc domain of IgG has been the target of multiple mutational studies aimed at altering the pH-dependent IgG/FcRn interaction to modulate IgG pharmacokinetics. These studies have yielded antibody variants with disparate pharmacokinetic characteristics, ranging from extended in vivo half-life to those exhibiting extremely rapid clearance. To better understand pH-dependent binding parameters that govern these outcomes and limit FcRn-mediated half-life extension, we generated a panel of novel Fc variants with high affinity binding at acidic pH that vary in pH 7.4 affinities and assessed pharmacokinetic outcomes. Pharmacokinetic studies in human FcRn transgenic mice and cynomolgus monkeys showed that multiple variants with increased FcRn affinities at acidic pH exhibited extended serum half-lives relative to the parental IgG. Importantly, the results reveal an underappreciated affinity threshold of neutral pH binding that determines IgG recycling efficiency. Variants with pH 7.4 FcRn affinities below this threshold recycle efficiently and can exhibit increased serum persistence. Increasing neutral pH FcRn affinity beyond this threshold reduced serum persistence by offsetting the benefits of increased pH 6.0 binding. Ultra-high affinity binding to FcRn at both acidic and neutral pH leads to rapid serum clearance.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Histocompatibility Antigens Class I/physiology , Immunoglobulin G/physiology , Protein Engineering , Receptors, Fc/physiology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibody Affinity , Bacteriophages , Female , Humans , Hydrogen-Ion Concentration , Macaca fascicularis , Male , Mice , Mice, Transgenic , Peptide Fragments/immunology , Peptide Fragments/metabolism , Peptide Library , Protein Binding , Protein Conformation , Surface Plasmon Resonance , Tissue Distribution
8.
Antimicrob Agents Chemother ; 60(9): 5312-21, 2016 09.
Article in English | MEDLINE | ID: mdl-27324766

ABSTRACT

Staphylococcus aureus infections lead to an array of illnesses ranging from mild skin infections to serious diseases, such endocarditis, osteomyelitis, and pneumonia. Alpha-toxin (Hla) is a pore-forming toxin, encoded by the hla gene, that is thought to play a key role in S. aureus pathogenesis. A monoclonal antibody targeting Hla, MEDI4893, is in clinical development for the prevention of S. aureus ventilator-associated pneumonia (VAP). The presence of the hla gene and Hla protein in 994 respiratory isolates collected from patients in 34 countries in Asia, Europe, the United States, Latin America, the Middle East, Africa, and Australia was determined. Hla levels were correlated with the geographic location, age of the subject, and length of stay in the hospital. hla gene sequence analysis was performed, and mutations were mapped to the Hla crystal structure. S. aureus supernatants containing Hla variants were tested for susceptibility or resistance to MEDI4893. The hla gene was present and Hla was expressed in 99.0% and 83.2% of the isolates, respectively, regardless of geographic region, hospital locale, or age of the subject. More methicillin-susceptible than methicillin-resistant isolates expressed Hla (86.9% versus 78.8%; P = 0.0007), and S. aureus isolates from pediatric patients expressed the largest amounts of Hla. Fifty-seven different Hla subtypes were identified, and 91% of the isolates encoded an Hla subtype that was neutralized by MED4893. This study demonstrates that Hla is conserved in diverse S. aureus isolates from around the world and is an attractive target for prophylactic monoclonal antibody (MAb) or vaccine development.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Bacterial Toxins/antagonists & inhibitors , Hemolysin Proteins/antagonists & inhibitors , Pneumonia, Ventilator-Associated/prevention & control , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Amino Acid Sequence , Anti-Inflammatory Agents/chemistry , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/chemistry , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Broadly Neutralizing Antibodies , Child , Child, Preschool , Conserved Sequence , Epidemiological Monitoring , Female , Global Health , Hemolysin Proteins/chemistry , Hemolysin Proteins/genetics , Hemolysin Proteins/immunology , Humans , Infant , Infant, Newborn , Length of Stay/statistics & numerical data , Male , Middle Aged , Models, Molecular , Mutation , Pneumonia, Ventilator-Associated/epidemiology , Pneumonia, Ventilator-Associated/microbiology , Pneumonia, Ventilator-Associated/pathology , Protein Conformation , Sequence Analysis, DNA , Staphylococcal Infections/epidemiology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Staphylococcus aureus/isolation & purification
9.
J Biol Chem ; 289(11): 7812-24, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24469444

ABSTRACT

We report the three-dimensional structure of human neonatal Fc receptor (FcRn) bound concurrently to its two known ligands. More particularly, we solved the crystal structure of the complex between human FcRn, wild-type human serum albumin (HSA), and a human Fc engineered for improved pharmacokinetics properties (Fc-YTE). The crystal structure of human FcRn bound to wild-type HSA alone is also presented. HSA domain III exhibits an extensive interface of contact with FcRn, whereas domain I plays a lesser role. A molecular explanation for the HSA recycling mechanism is provided with the identification of FcRn His(161) as the only potential direct contributor to the corresponding pH-dependent process. At last, this study also allows an accurate structural definition of residues considered for decades as important to the human IgG/FcRn interaction and reveals Fc His(310) as a significant contributor to pH-dependent binding. Finally, we explain various structural mechanisms by which several Fc mutations (including YTE) result in increased human IgG binding to FcRn. Our study provides an unprecedented relevant understanding of the molecular basis of human Fc interaction with human FcRn.


Subject(s)
Histocompatibility Antigens Class I/chemistry , Immunoglobulin G/chemistry , Receptors, Fc/chemistry , Serum Albumin/chemistry , Crystallization , Crystallography, X-Ray , HEK293 Cells , Humans , Hydrogen Bonding , Hydrogen-Ion Concentration , Immunoglobulin Fc Fragments/chemistry , Ligands , Models, Molecular , Mutation , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry
10.
J Biol Chem ; 289(43): 29874-80, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25210036

ABSTRACT

MEDI4893 is a neutralizing human monoclonal antibody that targets α-toxin (AT) and is currently undergoing evaluation in the field of Staphylococcus aureus-mediated diseases. We have solved the crystal structure of MEDI4893 Fab bound to monomeric AT at a resolution of 2.56 Å and further characterized its epitope using various engineered AT variants. We have found that MEDI4893 recognizes a novel epitope in the so-called "rim" domain of AT and exerts its neutralizing effect through a dual mechanism. In particular, MEDI4893 not only sterically blocks binding of AT to its cell receptor but also prevents it from adopting a lytic heptameric trans-membrane conformation.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Bacterial Toxins/immunology , Hemolysin Proteins/immunology , Neutralization Tests , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/chemistry , Broadly Neutralizing Antibodies , Cell Line , Crystallography, X-Ray , Epitope Mapping , Humans , Immunoglobulin Fab Fragments/chemistry , Models, Molecular , Protein Binding , Recombinant Proteins/chemistry , Surface Plasmon Resonance
11.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 11): 2354-61, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26527150

ABSTRACT

The three-dimensional structure of a human IgG1 Fc fragment bound to wild-type human FcγRI is reported. The structure of the corresponding complex was solved at a resolution of 2.4 Šusing molecular replacement; this is the highest resolution achieved for an unmutated FcγRI molecule. This study highlights the critical structural and functional role played by the second extracellular subdomain of FcγRI. It also explains the long-known major energetic contribution of the Fc `LLGG' motif at positions 234-237, and particularly of Leu235, via a `lock-and-key' mechanism. Finally, a previously held belief is corrected and a differing view is offered on the recently proposed direct role of Fc carbohydrates in the corresponding interaction. Structural evidence is provided that such glycan-related effects are strictly indirect.


Subject(s)
Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Receptors, IgG/chemistry , Receptors, IgG/metabolism , Cell Line , Crystallography, X-Ray , Humans , Models, Molecular , Polysaccharides/metabolism , Protein Binding , Protein Interaction Domains and Motifs
12.
Sci Transl Med ; 16(753): eado2817, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38924429

ABSTRACT

The evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in variants that can escape neutralization by therapeutic antibodies. Here, we describe AZD3152, a SARS-CoV-2-neutralizing monoclonal antibody designed to provide improved potency and coverage against emerging variants. AZD3152 binds to the back left shoulder of the SARS-CoV-2 spike protein receptor binding domain and prevents interaction with the human angiotensin-converting enzyme 2 receptor. AZD3152 potently neutralized a broad panel of pseudovirus variants, including the currently dominant Omicron variant JN.1 but has reduced potency against XBB subvariants containing F456L. In vitro studies confirmed F456L resistance and additionally identified T415I and K458E as escape mutations. In a Syrian hamster challenge model, prophylactic administration of AZD3152 protected hamsters from weight loss and inflammation-related lung pathologies and reduced lung viral load. In the phase 1 sentinel safety cohort of the ongoing SUPERNOVA study (ClinicalTrials.gov: NCT05648110), a single 600-mg intramuscular injection of AZD5156 (containing 300 mg each of AZD3152 and cilgavimab) was well tolerated in adults through day 91. Observed serum concentrations of AZD3152 through day 91 were similar to those observed with cilgavimab and consistent with predictions for AZD7442, a SARS-CoV-2-neutralizing antibody combination of cilgavimab and tixagevimab, in a population pharmacokinetic model. On the basis of its pharmacokinetic characteristics, AZD3152 is predicted to provide durable protection against symptomatic coronavirus disease 2019 caused by susceptible SARS-CoV-2 variants, such as JN.1, in humans.


Subject(s)
Antibodies, Neutralizing , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , SARS-CoV-2/drug effects , Humans , COVID-19/virology , Antibodies, Neutralizing/immunology , Spike Glycoprotein, Coronavirus/metabolism , Cricetinae , COVID-19 Drug Treatment , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacokinetics , Mesocricetus , Female , Male , Adult , Antibodies, Viral/immunology , Mutation/genetics , Antibodies, Monoclonal , Angiotensin-Converting Enzyme 2/metabolism , Viral Load/drug effects
13.
Article in English | MEDLINE | ID: mdl-23519809

ABSTRACT

Staphylococcus aureus alpha toxin (AT) has been crystallized in complex with the Fab fragment of a human antibody (MEDI4893). This constitutes the first reported crystals of AT bound to an antibody. The monoclinic crystals belonged to space group P21, with unit-cell parameters a=85.52, b=148.50, c=93.82 Å, ß=99.82°. The diffraction of the crystals extended to 2.56 Šresolution. The asymmetric unit contained two MEDI4893 Fab-AT complexes. This corresponds to a crystal volume per protein weight (VM) of 2.3 Å3 Da(-1) and a solvent content of 47%. The three-dimensional structure of this complex will contribute to an understanding of the molecular basis of the interaction of MEDI4893 with AT. It will also shed light on the mechanism of action of this antibody, the current evaluation of which in the field of S. aureus-mediated diseases makes it a particularly interesting case study. Finally, this study will provide the three-dimensional structure of AT in a monomeric state for the first time.


Subject(s)
Antibodies, Anti-Idiotypic/chemistry , Antibodies, Monoclonal/chemistry , Bacterial Toxins/chemistry , Immunoglobulin Fab Fragments/chemistry , Staphylococcus aureus/chemistry , Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal/immunology , Bacterial Toxins/immunology , Bacterial Toxins/isolation & purification , Crystallization , Crystallography, X-Ray , Humans , Immunoglobulin Fab Fragments/immunology , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism
14.
Article in English | MEDLINE | ID: mdl-23989160

ABSTRACT

Tn3 proteins are a novel class of binding molecules based on the third fibronectin type III domain of human tenascin C. Target-specific Tn3 proteins are selected from combinatorial libraries in which three surface-exposed loops have been diversified. Here, the cocrystallization of two different Tn3 proteins in complex with CD40L, a therapeutic target for immunological disease, is reported. These crystal structures are the first to be reported of Tn3 proteins and will help to reveal how these engineered molecules achieve specific recognition of a cognate target.


Subject(s)
CD40 Ligand/chemistry , Fibronectins/chemistry , Peptides/chemistry , Amino Acid Sequence , Binding Sites , CD40 Ligand/genetics , CD40 Ligand/isolation & purification , Crystallography, X-Ray , Escherichia coli/genetics , Fibronectins/genetics , Gene Expression , Humans , Molecular Sequence Data , Peptide Library , Peptides/genetics , Peptides/isolation & purification , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Sequence Alignment
15.
MAbs ; 15(1): 2273449, 2023.
Article in English | MEDLINE | ID: mdl-37930310

ABSTRACT

Bispecific antibodies represent an increasingly large fraction of biologics in therapeutic development due to their expanded scope in functional capabilities. Asymmetric monovalent bispecific IgGs (bsIgGs) have the additional advantage of maintaining a native antibody-like structure, which can provide favorable pharmacology and pharmacokinetic profiles. The production of correctly assembled asymmetric monovalent bsIgGs, however, is a complex engineering endeavor due to the propensity for non-cognate heavy and light chains to mis-pair. Previously, we introduced the DuetMab platform as a general solution for the production of bsIgGs, which utilizes an engineered interchain disulfide bond in one of the CH1-CL domains to promote orthogonal chain pairing between heavy and light chains. While highly effective in promoting cognate heavy and light chain pairing, residual chain mispairing could be detected for specific combinations of Fv pairs. Here, we present enhancements to the DuetMab design that improve chain pairing and production through the introduction of novel electrostatic steering mutations at the CH1-CL interface with lambda light chains (CH1-Cλ). These mutations work together with previously established charge-pair mutations at the CH1-CL interface with kappa light chains (CH1-Cκ) and Fab disulfide engineering to promote cognate heavy and light chain pairing and enable the reliable production of bsIgGs. Importantly, these enhanced DuetMabs do not require engineering of the variable domains and are robust when applied to a panel of bsIgGs with diverse Fv sequences. We present a comprehensive biochemical, biophysical, and functional characterization of the resulting DuetMabs to demonstrate compatibility with industrial production benchmarks. Overall, this enhanced DuetMab platform substantially streamlines process development of these disruptive biotherapeutics.


Subject(s)
Antibodies, Bispecific , Antibodies, Bispecific/genetics , Static Electricity , Disulfides , Mutation , Immunoglobulin G/genetics
16.
Commun Biol ; 4(1): 1048, 2021 09 08.
Article in English | MEDLINE | ID: mdl-34497355

ABSTRACT

In a biologic therapeutic landscape that requires versatility in targeting specificity, valency and half-life modulation, the monomeric Fc fusion platform holds exciting potential for the creation of a class of monovalent protein therapeutics that includes fusion proteins and bispecific targeting molecules. Here we report a structure-guided approach to engineer monomeric Fc molecules to adapt multiple versions of half-life extension modifications. Co-crystal structures of these monomeric Fc variants with Fc neonatal receptor (FcRn) shed light into the binding interactions that could serve as a guide for engineering the half-life of antibody Fc fragments. These engineered monomeric Fc molecules also enabled the generation of a novel monovalent bispecific molecular design, which translated the FcRn binding enhancement to improvement of in vivo serum half-life.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Immunoglobulin Fc Fragments/metabolism , Receptors, Fc/metabolism , Animals , Half-Life , Histocompatibility Antigens Class I/pharmacology , Humans , Immunoglobulin Fc Fragments/pharmacology , Mice , Mice, Transgenic , Protein Engineering
17.
Article in English | MEDLINE | ID: mdl-20516612

ABSTRACT

The recombinant N-terminal domain of human ephrin type-A receptor 2 (rEphA2) has been crystallized in complex with the recombinantly produced Fab fragment of a fully human antibody (1C1; IgG1/kappa). These are the first reported crystals of an ephrin receptor bound to an antibody. The orthorhombic crystals belonged to space group C222(1) (the 00l reflections obey the l = 2n rule), with unit-cell parameters a = 78.93, b = 120.79, c = 286.20 A. The diffraction of the crystals extended to 2.0 A resolution. However, only data to 2.55 A resolution were considered to be useful owing to spot overlap caused by the long unit-cell parameter. The asymmetric unit is most likely to contain two 1C1 Fab-rEphA2 complexes. This corresponds to a crystal volume per protein weight (V(M)) of 2.4 A(3) Da(-1) and a solvent content of 49.5%. The three-dimensional structure of this complex will shed light on the molecular basis of 1C1 specificity. This will also contribute to a better understanding of the mechanism of action of this antibody, the current evaluation of which as an antibody-drug conjugate in cancer therapy makes it a particularly interesting case study.


Subject(s)
Antigen-Antibody Complex/chemistry , Immunoglobulin Fab Fragments/chemistry , Peptide Fragments/chemistry , Receptor, EphA2/chemistry , Antigen-Antibody Complex/immunology , Crystallization , Crystallography, X-Ray , Humans , Immunoglobulin Fab Fragments/immunology , Peptide Fragments/immunology , Receptor, EphA2/immunology
18.
Sci Rep ; 10(1): 17257, 2020 10 14.
Article in English | MEDLINE | ID: mdl-33057063

ABSTRACT

Neuregulin protein 1 (NRG1) is a large (> 60-amino-acid) natural peptide ligand for the ErbB protein family members HER3 and HER4. We developed an agonistic antibody modality, termed antibody ligand mimetics (ALM), by incorporating complex ligand agonists such as NRG1 into an antibody scaffold. We optimized the linker and ligand length to achieve native ligand activity in HEK293 cells and cardiomyocytes derived from induced pluripotent stem cells (iPSCs) and used a monomeric Fc-ligand fusion platform to steer the ligand specificity toward HER4-dominant agonism. With the help of selectivity engineering, these enhanced ALM molecules can provide an antibody scaffold with increased receptor specificity and the potential to greatly improve the pharmacokinetics, stability, and downstream developability profiles from the natural ligand approach. This ligand mimetic design and optimization approach can be expanded to apply to other cardiovascular disease targets and emerging therapeutic areas, providing differentiated drug molecules with increased specificity and extended half-life.


Subject(s)
Antibodies, Monoclonal/chemistry , Neuregulin-1/chemistry , Receptor, ErbB-4/agonists , Antibodies, Monoclonal/metabolism , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/chemistry , Induced Pluripotent Stem Cells/metabolism , Kinetics , Ligands , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/metabolism , Neuregulin-1/metabolism , Protein Binding , Receptor, ErbB-4/metabolism , Signal Transduction
19.
Article in English | MEDLINE | ID: mdl-19153447

ABSTRACT

Recombinant human interferon alpha-2A (rhIFN-alpha-2A) has been crystallized in complex with the recombinantly produced Fab fragment of a therapeutic monoclonal antibody (MEDI545; IgG1/kappa) which targets several human interferon alpha subtypes. This constitutes the first reported crystals of a human type I interferon bound to an antibody. The orthorhombic crystals belonged to either space group I222 or I2(1)2(1)2(1), with unit-cell parameters a = 134.82, b = 153.26, c = 163.49 A. The diffraction of the crystals extended to 3.0 A resolution. The asymmetric unit contained two Fab-rhIFN-alpha-2A complexes. This corresponded to a crystal volume per protein weight (V(M)) of 3.02 A(3) Da(-1) and a solvent content of 59.3%. The corresponding three-dimensional structure is expected to shed light on the mechanism of action of MEDI545 and the molecular basis of its specificity.


Subject(s)
Antibodies, Anti-Idiotypic/chemistry , Interferon-alpha/chemistry , Interferons/chemistry , Antibodies, Monoclonal/chemistry , Crystallization , Electrophoresis, Polyacrylamide Gel , Humans , Imaging, Three-Dimensional , Immunoglobulin Fab Fragments/chemistry , Interferon alpha-2 , Molecular Conformation , Molecular Structure , Recombinant Proteins , X-Ray Diffraction
20.
Mol Immunol ; 45(7): 1872-82, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18078997

ABSTRACT

We report here the three-dimensional structure of a human Fc fragment engineered for enhanced antibody dependent cell mediated cytotoxicity (ADCC). The triple mutation S239D/A330L/I332E ('3M') was introduced into the C(H)2 portion of a human immunoglobulin G1 (IgG1) Fc. These three substitutions typically result in an about 10-100-fold increase in human IgG1 binding to human Fc gamma RIIIA (CD16). The recombinantly produced Fc/3M fragment was crystallized and its structure solved at a resolution of 2.5A using molecular replacement. No dramatic structural changes were observed in Fc/3M when compared with unmutated human Fc fragments. However, we found that the relative positions of its C(H)2 domains allowed for an unusually 'open' conformation of the entire fragment. Although this particular structural feature could be due to crystallization artifacts or intrinsic variability, we propose that molecular mechanisms at the basis of the enhanced interaction between Fc/3M and CD16 could include enhanced Fc openness as well as the introduction of additional hydrophobic contacts, hydrogen bonds and/or electrostatic interactions at the corresponding interface. The existence of a more pronounced cleft between the two Fc chains as well as of repulsive, electrostatic intra-chain interactions may also account in part for the decreased thermostability of both Fc/3M and a 3M-modified humanized anti-human EphA2 IgG1 when compared with their respective unmutated counterparts.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Immunoglobulin Fc Fragments/chemistry , Mutant Proteins/chemistry , Calorimetry, Differential Scanning , Carbohydrates/analysis , Cell Line , Crystallography, X-Ray , Humans , Immunoglobulin G/chemistry , Ligands , Metals/metabolism , Models, Molecular , Protein Binding , Protein Structure, Secondary , Receptors, IgG/chemistry , Structure-Activity Relationship , Thermodynamics , Transition Temperature
SELECTION OF CITATIONS
SEARCH DETAIL