Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Proteins ; 90(1): 270-281, 2022 01.
Article in English | MEDLINE | ID: mdl-34405904

ABSTRACT

This study uses differential scanning calorimetry, X-ray crystallography, and molecular dynamics simulations to investigate the structural basis for the high thermal stability (melting temperature 97.5°C) of a FN3-like protein domain from thermophilic bacteria Thermoanaerobacter tengcongensis (FN3tt). FN3tt adopts a typical FN3 fold with a three-stranded beta sheet packing against a four-stranded beta sheet. We identified three solvent exposed arginine residues (R23, R25, and R72), which stabilize the protein through salt bridge interactions with glutamic acid residues on adjacent strands. Alanine mutation of the three arginine residues reduced melting temperature by up to 22°C. Crystal structures of the wild type (WT) and a thermally destabilized (∆Tm -19.7°C) triple mutant (R23L/R25T/R72I) were found to be nearly identical, suggesting that the destabilization is due to interactions of the arginine residues. Molecular dynamics simulations showed that the salt bridge interactions in the WT were stable and provided a dynamical explanation for the cooperativity observed between R23 and R25 based on calorimetry measurements. In addition, folding free energy changes computed using free energy perturbation molecular dynamics simulations showed high correlation with melting temperature changes. This work is another example of surface salt bridges contributing to the enhanced thermal stability of thermophilic proteins. The molecular dynamics simulation methods employed in this study may be broadly useful for in silico surface charge engineering of proteins.


Subject(s)
Bacterial Proteins/chemistry , Fibronectin Type III Domain , Sodium Chloride/chemistry , Thermoanaerobacter/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Hot Temperature , Molecular Dynamics Simulation , Protein Domains , Protein Stability , Thermoanaerobacter/genetics
2.
Antibodies (Basel) ; 8(4)2019 Dec 03.
Article in English | MEDLINE | ID: mdl-31816964

ABSTRACT

Antibodies and antibody-derived macromolecules have established themselves as the mainstay in protein-based therapeutic molecules (biologics). Our knowledge of the structure-function relationships of antibodies provides a platform for protein engineering that has been exploited to generate a wide range of biologics for a host of therapeutic indications. In this review, our basic understanding of the antibody structure is described along with how that knowledge has leveraged the engineering of antibody and antibody-related therapeutics having the appropriate antigen affinity, effector function, and biophysical properties. The platforms examined include the development of antibodies, antibody fragments, bispecific antibody, and antibody fusion products, whose efficacy and manufacturability can be improved via humanization, affinity modulation, and stability enhancement. We also review the design and selection of binding arms, and avidity modulation. Different strategies of preparing bispecific and multispecific molecules for an array of therapeutic applications are included.

3.
Proteins ; 86(5): 495-500, 2018 05.
Article in English | MEDLINE | ID: mdl-29490423

ABSTRACT

CD19 is a transmembrane protein expressed on malignant B cells, but not in other lineages or other tissues, which makes it an attractive target for monoclonal antibody-mediated immunotherapy. Anti-CD19 antibody B43 was utilized in a bispecific T-cell engager (BiTE) blinatumomab that demonstrated potency for the treatment of relapsed acute lymphoblastic leukemia. To gain insight into the mechanism of action of the antibody, the crystal structure of B43 Fab was determined in complex with CD19 and in the unbound form. The structure revealed the binding epitope, explained the lack of cross-reactivity toward non-human species, and suggested the key-and-lock mechanism of antigen recognition. Most unexpectedly, the structure revealed a unique molecular topology of CD19. Rather than a tandem of c-type immunoglobulin folds predicted from the amino acid sequence, the extracellular domain of CD19 exhibits an elongated ß-sandwich formed by two immunoglobulin folds by swapping their C-terminal halves. This is the first structure of CD19, which has no sequence homologs.


Subject(s)
Antibodies, Monoclonal/chemistry , Antigens, CD19/chemistry , Amino Acid Sequence , Animals , B-Lymphocytes/cytology , Binding Sites , Crystallography, X-Ray , HEK293 Cells , Humans , Mice , Models, Molecular , Protein Binding , Protein Folding , Protein Structure, Secondary
4.
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
5.
MAbs ; 10(2): 269-277, 2018.
Article in English | MEDLINE | ID: mdl-29283291

ABSTRACT

Murine antibody 10H10 raised against human tissue factor is unique in that it blocks the signaling pathway, and thus inhibits angiogenesis and tumor growth without interfering with coagulation. As a potential therapeutic, the antibody was humanized in a two-step procedure. Antigen-binding loops were grafted onto selected human frameworks and the resulting chimeric antibody was subjected to affinity maturation by using phage display libraries. The results of humanization were analyzed from the structural perspective through comparison of the structure of a humanized variant with the parental mouse antibody. This analysis revealed several hot spots in the framework region that appear to affect antigen binding, and therefore should be considered in human germline selection. In addition, some positions in the Vernier zone, e.g., residue 71 in the heavy chain, that are traditionally thought to be crucial appear to tolerate amino acid substitutions without any effect on binding. Several humanized variants were produced using both short and long forms of complementarity-determining region (CDR) H2 following the difference in the Kabat and Martin definitions. Comparison of such pairs indicated consistently higher thermostability of the variants with short CDR H2. Analysis of the binding data in relation to the structures singled out the ImMunoGeneTics information system® germline IGHV1-2*01 as dubious owing to two potentially destabilizing mutations as compared to the other alleles of the same germline and to other human germlines.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Antibody Affinity/physiology , Thromboplastin/immunology , Animals , Antibodies, Monoclonal, Humanized/immunology , Complementarity Determining Regions/chemistry , Humans , Mice , Models, Molecular , Protein Engineering/methods
6.
Biochem Biophys Rep ; 13: 27-31, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29264403

ABSTRACT

The homeostatic chemokine CCL17, also known as thymus and activation regulated chemokine (TARC), has been associated with various diseases such as asthma, idiopathic pulmonary fibrosis, atopic dermatitis and ulcerative colitis. Neutralization of CCL17 by antibody treatment ameliorates the impact of disease by blocking influx of T cells. Monoclonal antibody M116 derived from a combinatorial library shows potency in neutralizing CCL17-induced signaling. To gain insight into the structural determinants of antigen recognition, the crystal structure of M116 Fab was determined in complex with CCL17 and in the unbound form. Comparison of the structures revealed an unusual induced-fit mechanism of antigen recognition that involves cis-trans isomerization in two CDRs. The structure of the CCL17-M116 complex revealed the antibody binding epitope, which does not overlap with the putative receptor epitope, suggesting that the current model of chemokine-receptor interactions, as observed in the CXCR4-vMIP-II system, may not be universal.

7.
Protein Sci ; 27(3): 798-808, 2018 03.
Article in English | MEDLINE | ID: mdl-29168245

ABSTRACT

The Protein Data Bank (PDB) is the global archive for structural information on macromolecules, and a popular resource for researchers, teachers, and students, amassing more than one million unique users each year. Crystallographic structure models in the PDB (more than 100,000 entries) are optimized against the crystal diffraction data and geometrical restraints. This process of crystallographic refinement typically ignored hydrogen bond (H-bond) distances as a source of information. However, H-bond restraints can improve structures at low resolution where diffraction data are limited. To improve low-resolution structure refinement, we present methods for deriving H-bond information either globally from well-refined high-resolution structures from the PDB-REDO databank, or specifically from on-the-fly constructed sets of homologous high-resolution structures. Refinement incorporating HOmology DErived Restraints (HODER), improves geometrical quality and the fit to the diffraction data for many low-resolution structures. To make these improvements readily available to the general public, we applied our new algorithms to all crystallographic structures in the PDB: using massively parallel computing, we constructed a new instance of the PDB-REDO databank (https://pdb-redo.eu). This resource is useful for researchers to gain insight on individual structures, on specific protein families (as we demonstrate with examples), and on general features of protein structure using data mining approaches on a uniformly treated dataset.


Subject(s)
Computational Biology/methods , Proteins/chemistry , Algorithms , Crystallography, X-Ray , Data Mining , Databases, Protein , Hydrogen Bonding , Models, Molecular , Protein Conformation
8.
MAbs ; 9(8): 1306-1316, 2017.
Article in English | MEDLINE | ID: mdl-28898162

ABSTRACT

The increased number of bispecific antibodies (BsAb) under therapeutic development has resulted in a need for mouse surrogate BsAbs. Here, we describe a one-step method for generating highly pure mouse BsAbs suitable for in vitro and in vivo studies. We identify two mutations in the mouse IgG2a and IgG2b Fc region: one that eliminates protein A binding and one that enhances protein A binding by 8-fold. We show that BsAbs harboring these mutations can be purified from the residual parental monoclonal antibodies in one step using protein A affinity chromatography. The structural basis for the effects of these mutations was analyzed by X-ray crystallography. While the mutation that disrupted protein A binding also inhibited FcRn interaction, a bispecific mutant in which one subunit retained the ability to bind protein A could still interact with FcRn. Pharmacokinetic analysis of the serum half-lives of the mutants showed that the mutant BsAb had a serum half-life comparable to a wild-type Ab. The results describe a rapid method for generating panels of mouse BsAbs that could be used in mouse studies.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Histocompatibility Antigens Class I/immunology , Receptors, Fc/immunology , Staphylococcal Protein A/immunology , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/metabolism , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Crystallography, X-Ray , Histocompatibility Antigens Class I/metabolism , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Mice , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/immunology , Mutant Proteins/metabolism , Mutation , Protein Binding/immunology , Protein Domains , Receptors, Fc/metabolism , Staphylococcal Protein A/metabolism
9.
MAbs ; 9(7): 1129-1142, 2017 10.
Article in English | MEDLINE | ID: mdl-28758875

ABSTRACT

Immunostimulatory receptors belonging to the tumor necrosis factor receptor (TNFR) superfamily are emerging as promising targets for cancer immunotherapies. To optimize the agonism of therapeutic antibodies to these receptors, Fc engineering of antibodies was applied to facilitate the clustering of cell surface TNFRs to activate downstream signaling pathways. One engineering strategy is to identify Fc mutations that facilitate antibody multimerization on the cell surface directly. From the analyses of the crystal packing of IgG1 structures, we identified a novel set of Fc mutations, T437R and K248E, that facilitated antibody multimerization upon binding to antigens on cell surface. In a NF-κB reporter assay, the engineered T437R/K248E mutations could facilitate enhanced agonism of an anti-OX40 antibody without the dependence on FcγRIIB crosslinking. Nonetheless, the presence of cells expressing FcγRIIB could facilitate a boost of the agonism of the engineered antibody with mutations on IgG1 Fc, but not on the silent IgG2σ Fc. The Fc engineered antibody also showed enhanced effector functions, including antibody-dependent cell-meditated cytotoxicity, antibody-dependent cellular phagocytosis, and complement-dependent cytotoxicity, depending on the IgG subtypes. Also, the engineered antibodies showed normal FcRn binding and pharmacokinetic profiles in mice. In summary, this study elucidated a novel Fc engineering approach to promote antibody multimerization on a cell surface, which could enhance agonism and improve effector function for anti-TNFR antibodies as well as other therapeutic antibodies.


Subject(s)
Immunoglobulin Fc Fragments/immunology , Immunotherapy/methods , Protein Engineering/methods , Receptors, OX40/agonists , Animals , Antibody-Dependent Cell Cytotoxicity , Humans , Mice , Mutation
10.
Alzheimers Res Ther ; 9(1): 66, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28830506

ABSTRACT

BACKGROUND: ß-Amyloid (Aß) peptide is believed to play a pivotal role in the development of Alzheimer's disease. Passive immunization with anti-Aß monoclonal antibodies may facilitate the clearance of Aß in the brain and may thus prevent the downstream pathology. Antibodies targeting the immunodominant N-terminal epitope of Aß and capable of binding both the plaques and soluble species have been most efficacious in animal models. Structural studies of such antibodies with bound Aß peptides provided the basis for understanding the mechanisms of action and the differences in potency. To gain further insight into the structural determinants of antigen recognition and the preferential Aß conformations, we determined the crystal structure of murine antibody C706 in complex with the N-terminal Aß 1-16 peptide sequence. METHODS: The antigen-binding fragment of C706 was expressed in HEK293 cells and was crystallized in complex with the Aß peptide. The X-ray structure was determined at 1.9-Å resolution. RESULTS: The binding epitope of C706 is centered on residues Arg5 and His6, which provide the majority of interactions. Unlike most antibodies, C706 recognizes a coiled rather than extended conformation of Aß. CONCLUSIONS: Comparison with other antibodies targeting the N-terminal section of Aß suggests that the conformation of the bound peptide may be linked to the immunization protocol and may reflect the preference for the extended conformation in the context of a longer Aß peptide as opposed to the coiled conformation in the isolated short peptide.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Peptide Fragments/chemistry , Peptide Fragments/immunology , Animals , Antibody Specificity , Crystallography, X-Ray , HEK293 Cells , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Mice , Models, Molecular , Protein Conformation , Recombinant Fusion Proteins/chemistry
11.
Acta Crystallogr F Struct Biol Commun ; 73(Pt 5): 294-299, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28471362

ABSTRACT

CD27 is a T-cell and B-cell co-stimulatory glycoprotein of the tumor necrosis factor (TNF) receptor superfamily that is dependent on the availability of the TNF-like ligand CD70. Therapeutic approaches to treating autoimmune diseases and cancers with antagonistic and agonistic anti-CD27 monoclonal antibodies (mAbs), respectively, have recently been developed. Mouse anti-human CD27 mAb 2177 shows potency in neutralizing CD70-induced signaling; however, it does not block the binding of soluble CD70. To provide insight into the mechanism of action of the mAb, the crystal structure of the CD27 extracellular domain in complex with the Fab fragment of mAb 2177 was determined at 1.8 Šresolution. CD27 exhibits the assembly of cysteine-rich domains characteristic of the TNF receptor superfamily. The structure reveals a unique binding site of mAb 2177 at the edge of the receptor molecule, which allows the mAb to sterically block the cell-bound form of CD70 from reaching CD27 while leaving the ligand epitope clear. This mode of action suggests a potential dual use of mAb 2177 either as an antagonist or as an agonist.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Antigen-Antibody Complex/chemistry , CD27 Ligand/chemistry , Immunoglobulin Fab Fragments/chemistry , Tumor Necrosis Factor Receptor Superfamily, Member 7/chemistry , Amino Acid Motifs , Animals , Antibodies, Monoclonal/genetics , Antibodies, Neutralizing/genetics , Antigen-Antibody Complex/genetics , Baculoviridae/genetics , Baculoviridae/metabolism , Binding Sites , CD27 Ligand/genetics , CD27 Ligand/immunology , Cloning, Molecular , Crystallography, X-Ray , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/genetics , Ligands , Mice , Models, Molecular , Protein Binding , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sequence Alignment , Sf9 Cells , Spodoptera , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
12.
Cell Signal ; 36: 139-144, 2017 08.
Article in English | MEDLINE | ID: mdl-28483635

ABSTRACT

Tissue factor (TF) initiates the extrinsic pathway of blood coagulation through sequential binding and activation of coagulation factors VII (FVII) and X (FX). In addition, through activation of G-protein-coupled protease activated receptors (PARs) TF induces cell signaling that is related to cancer, angiogenesis and inflammation. Monoclonal antibodies (mAbs) proved to be a useful tool for studying the interplay between TF signaling and coagulation. MAb 10H10 is unique in that it blocks the signaling pathway and thus inhibits angiogenesis and tumor growth without interfering with coagulation. It was also presumed that mAb 10H10 recognizes the cryptic pool of TF devoid of procoagulant activity. The crystal structure of the 10H10 Fab was determined in the absence and in the presence of the TF extracellular domain (ECD). The structures show that the antibody operates by the key-and-lock mechanism causing no conformational changes in either Fab or TF. The TF:10H10 interface is extensive and includes five segments of TF in both the N-terminal and C-terminal domains of the ECD. Neither the known epitope of FVII, nor the putative epitope of FX overlaps with the 10H10 binding site. The 10H10 epitope points to the likely location of the PAR2 exosite. It is also the hypothetical site of TF interaction with integrins that may play a major role in the encryption-decryption process.


Subject(s)
Antibodies, Monoclonal/metabolism , Epitopes/metabolism , Signal Transduction , Thromboplastin/chemistry , Thromboplastin/metabolism , Animals , Crystallography, X-Ray , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Mice , Models, Molecular , Protein Structure, Secondary
13.
Mol Immunol ; 83: 92-99, 2017 03.
Article in English | MEDLINE | ID: mdl-28119207

ABSTRACT

CD27 is a T and B cell co-stimulatory protein of the TNF receptor superfamily dependent on the availability of the TNF-like ligand CD70. Two anti-CD27 neutralizing monoclonal antibodies were obtained from mouse hybridoma and subsequently humanized and optimized for binding the target. The two antibodies are similar in terms of their CD27-binding affinity and ability to block NF-κB signaling, however their clearance rates in monkeys are very different. The pharmacokinetics profiles could be epitope dependent. To identify the epitopes, we determined the crystal structure of the ternary complex between CD27 and the Fab fragments of these non-competing antibodies. The structure reveals the binding modes of the antibodies suggesting that their mechanisms of action are distinctly different and provides a possible explanation of the in vivo data.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibody Affinity , CD27 Ligand/chemistry , CD27 Ligand/immunology , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Half-Life , Humans , Macaca fascicularis , Mice
14.
Antibodies (Basel) ; 6(3)2017 Sep 01.
Article in English | MEDLINE | ID: mdl-31548527

ABSTRACT

Engineering of fragment crystallizable (Fc) domains of therapeutic immunoglobulin (IgG) antibodies to eliminate their immune effector functions while retaining other Fc characteristics has numerous applications, including blocking antigens on Fc gamma (Fcγ) receptor-expressing immune cells. We previously reported on a human IgG2 variant termed IgG2σ with barely detectable activity in antibody-dependent cellular cytotoxicity, phagocytosis, complement activity, and Fcγ receptor binding assays. Here, we extend that work to IgG1 and IgG4 antibodies, alternative subtypes which may offer advantages over IgG2 antibodies. In several in vitro and in vivo assays, the IgG1σ and IgG4σ variants showed equal or even lower Fc-related activities than the corresponding IgG2σ variant. In particular, IgG1σ and IgG4σ variants demonstrate complete lack of effector function as measured by antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, antibody-dependent cellular phagocytosis, and in vivo T-cell activation. The IgG1σ and IgG4σ variants showed acceptable solubility and stability, and typical human IgG1 pharmacokinetic profiles in human FcRn-transgenic mice and cynomolgus monkeys. In silico T-cell epitope analyses predict a lack of immunogenicity in humans. Finally, crystal structures and simulations of the IgG1σ and IgG4σ Fc domains can explain the lack of Fc-mediated immune functions. These variants show promise for use in those therapeutic antibodies and Fc fusions for which the Fc domain should be immunologically "silent".

15.
Protein Eng Des Sel ; 30(1): 31-37, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27881684

ABSTRACT

Designed ankyrin repeat proteins (DARPin®) are artificial non-immunoglobulin binding proteins with potential applications as therapeutic molecules. DARPin 6G9 binds interleukin-13 with high affinity and blocks the signaling pathway and as such is promising for the treatment of asthma and other atopic diseases. The crystal structures of DARPin 6G9 in the unbound form and in complex with IL-13 were determined at high resolution. The DARPin competes for the same epitope as the IL-13 receptor chain 13Rα1 but does not interfere with the binding of the other receptor chain, IL-4Rα. Analysis of multiple copies of the DARPin molecule in the crystal indicates the conformational instability in the N-terminal cap that was predicted from molecular dynamics simulations. Comparison of the DARPin structures in the free state and in complex with IL-13 reveals a concerted movement of the ankyrin repeats upon binding resulted in the opening of the binding site. The induced-fit mode of binding employed by DARPin 6G9 is very unusual for DARPins since they were designed as particularly stable and rigid molecules. This finding shows that DARPins can operate by various binding mechanisms and suggests that some flexibility in the scaffold may be an advantage.


Subject(s)
Ankyrin Repeat , Antibodies/chemistry , Antibodies/immunology , Interleukin-13/immunology , Amino Acid Sequence , Animals , Antibodies/genetics , Crystallography, X-Ray , Humans , Macaca fascicularis , Models, Molecular , Protein Engineering , Protein Structure, Secondary
16.
Curr Opin Struct Biol ; 38: 163-73, 2016 06.
Article in English | MEDLINE | ID: mdl-27525816

ABSTRACT

The successful introduction of antibody-based protein therapeutics into the arsenal of treatments for patients has within a few decades fostered intense innovation in the production and engineering of antibodies. Reviewed here are the methods currently used to produce antibodies along with how our knowledge of the structural and functional characterization of immunoglobulins has resulted in the engineering of antibodies to produce protein therapeutics with unique properties, both biological and biophysical, that are leading to novel therapeutic approaches. Antibody engineering includes the introduction of the antibody combining site (variable regions) into a host of architectures including bi and multi-specific formats that further impact the therapeutic properties leading to further advantages and successes in patient treatment.


Subject(s)
Antibodies/genetics , Antibodies/therapeutic use , Protein Engineering/methods , Animals , Antibodies/chemistry , Antibodies/metabolism , Humans , Tissue Distribution
17.
MAbs ; 8(6): 1045-63, 2016.
Article in English | MEDLINE | ID: mdl-27210805

ABSTRACT

To support antibody therapeutic development, the crystal structures of a set of 16 germline variants composed of 4 different kappa light chains paired with 4 different heavy chains have been determined. All four heavy chains of the antigen-binding fragments (Fabs) have the same complementarity-determining region (CDR) H3 that was reported in an earlier Fab structure. The structure analyses include comparisons of the overall structures, canonical structures of the CDRs and the VH:VL packing interactions. The CDR conformations for the most part are tightly clustered, especially for the ones with shorter lengths. The longer CDRs with tandem glycines or serines have more conformational diversity than the others. CDR H3, despite having the same amino acid sequence, exhibits the largest conformational diversity. About half of the structures have CDR H3 conformations similar to that of the parent; the others diverge significantly. One conclusion is that the CDR H3 conformations are influenced by both their amino acid sequence and their structural environment determined by the heavy and light chain pairing. The stem regions of 14 of the variant pairs are in the 'kinked' conformation, and only 2 are in the extended conformation. The packing of the VH and VL domains is consistent with our knowledge of antibody structure, and the tilt angles between these domains cover a range of 11 degrees. Two of 16 structures showed particularly large variations in the tilt angles when compared with the other pairings. The structures and their analyses provide a rich foundation for future antibody modeling and engineering efforts.


Subject(s)
Antibody Diversity , Complementarity Determining Regions/chemistry , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Light Chains/chemistry , Single-Domain Antibodies/chemistry , HEK293 Cells , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/genetics , Models, Molecular , Protein Conformation , Protein Engineering , Single-Domain Antibodies/genetics , Synchrotrons
18.
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
19.
Proteins ; 83(6): 1191-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25900776

ABSTRACT

The crystal structure of DARPin 44C12V5 that neutralizes IL-4 signaling has been determined alone and bound to human IL-4. A significant conformational change occurs in the IL-4 upon DARPin binding. The DARPin binds to the face of IL-4 formed by the A and C α-helices. The structure of the DARPin remains virtually unchanged. The conformational changes in IL-4 include a reorientation of the C-helix Trp91 side chain and repositioning of CD-loop residue Leu96. Both side chains move by >9 Å, becoming buried in the central hydrophobic region of the IL-4:DARPin interface. This hydrophobic region is surrounded by a ring of hydrophilic interactions comprised of hydrogen bonds and salt bridges and represents a classical "hotspot." The structures also reveal how the DARPin neutralizes IL-4 signaling. Comparing the IL-4:DARPin complex structure with the structures of IL-4 bound to its receptors (Hage et al., Cell 1999; 97, 271-281; La Porte et al., Cell 2008, 132, 259-272), it is found that the DARPin binds to the same IL-4 face that interacts with the junction of the D1 and D2 domains of the IL-4Rα receptors. Signaling is blocked since IL-4 cannot bind to this receptor, which it must do first before initiating a productive receptor complex with either the IL-13α1 or the γc receptor.


Subject(s)
Interleukin-4/chemistry , Interleukin-4/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Ankyrin Repeat , Humans , Models, Molecular , Protein Conformation , Recombinant Proteins/pharmacology
20.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 8): 1107-15, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25084393

ABSTRACT

The crystallization of 16 human antibody Fab fragments constructed from all pairs of four different heavy chains and four different light chains was enabled by employing microseed matrix screening (MMS). In initial screening, diffraction-quality crystals were obtained for only three Fabs, while many Fabs produced hits that required optimization. Application of MMS, using the initial screens and/or refinement screens, resulted in diffraction-quality crystals of these Fabs. Five Fabs that failed to give hits in the initial screen were crystallized by cross-seeding MMS followed by MMS optimization. The crystallization protocols and strategies that resulted in structure determination of all 16 Fabs are presented. These results illustrate the power of MMS and provide a basis for developing future strategies for macromolecular crystallization.


Subject(s)
Immunoglobulin Fab Fragments/immunology , Proteins/chemistry , Crystallization , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...