Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Chem Inf Model ; 63(22): 6964-6971, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-37934909

ABSTRACT

The electrostatic properties of proteins arise from the number and distribution of polar and charged residues. Electrostatic interactions in proteins play a critical role in numerous processes such as molecular recognition, protein solubility, viscosity, and antibody developability. Thus, characterizing and quantifying electrostatic properties of a protein are prerequisites for understanding these processes. Here, we present PEP-Patch, a tool to visualize and quantify the electrostatic potential on the protein surface in terms of surface patches, denoting separated areas of the surface with a common physical property. We highlight its applicability to elucidate protease substrate specificity and antibody-antigen recognition and predict heparin column retention times of antibodies as an indicator of pharmacokinetics.


Subject(s)
Antibodies , Proteins , Static Electricity , Proteins/chemistry , Solubility , Viscosity
2.
MAbs ; 15(1): 2171248, 2023.
Article in English | MEDLINE | ID: mdl-36823021

ABSTRACT

Beyond potency, a good developability profile is a key attribute of a biological drug. Selecting and screening for such attributes early in the drug development process can save resources and avoid costly late-stage failures. Here, we review some of the most important developability properties that can be assessed early on for biologics. These include the influence of the source of the biologic, its biophysical and pharmacokinetic properties, and how well it can be expressed recombinantly. We furthermore present in silico, in vitro, and in vivo methods and techniques that can be exploited at different stages of the discovery process to identify molecules with liabilities and thereby facilitate the selection of the most optimal drug leads. Finally, we reflect on the most relevant developability parameters for injectable versus orally delivered biologics and provide an outlook toward what general trends are expected to rise in the development of biologics.


Subject(s)
Biological Products , Drug Discovery , Drug Discovery/methods , Antibodies, Monoclonal
3.
Front Mol Biosci ; 9: 960194, 2022.
Article in English | MEDLINE | ID: mdl-36120542

ABSTRACT

While antibody-based therapeutics have grown to be one of the major classes of novel medicines, some antibody development candidates face significant challenges regarding expression levels, solubility, as well as stability and aggregation, under physiological and storage conditions. A major determinant of those properties is surface hydrophobicity, which promotes unspecific interactions and has repeatedly proven problematic in the development of novel antibody-based drugs. Multiple computational methods have been devised for in-silico prediction of antibody hydrophobicity, often using hydrophobicity scales to assign values to each amino acid. Those approaches are usually validated by their ability to rank potential therapeutic antibodies in terms of their experimental hydrophobicity. However, there is significant diversity both in the hydrophobicity scales and in the experimental methods, and consequently in the performance of in-silico methods to predict experimental results. In this work, we investigate hydrophobicity of monoclonal antibodies using hydrophobicity scales. We implement several scoring schemes based on the solvent-accessibility and the assigned hydrophobicity values, and compare the different scores and scales based on their ability to predict retention times from hydrophobic interaction chromatography. We provide an overview of the strengths and weaknesses of several commonly employed hydrophobicity scales, thereby improving the understanding of hydrophobicity in antibody development. Furthermore, we test several datasets, both publicly available and proprietary, and find that the diversity of the dataset affects the performance of hydrophobicity scores. We expect that this work will provide valuable guidelines for the optimization of biophysical properties in future drug discovery campaigns.

5.
Front Mol Biosci ; 9: 812750, 2022.
Article in English | MEDLINE | ID: mdl-35155578

ABSTRACT

As the current biotherapeutic market is dominated by antibodies, the design of different antibody formats, like bispecific antibodies and other new formats, represent a key component in advancing antibody therapy. When designing new formats, a targeted modulation of pairing preferences is key. Several existing approaches are successful, but expanding the repertoire of design possibilities would be desirable. Cognate immunoglobulin G antibodies depend on homodimerization of the fragment crystallizable regions of two identical heavy chains. By modifying the dimeric interface of the third constant domain (CH3-CH3), with different mutations on each domain, the engineered Fc fragments form rather heterodimers than homodimers. The first constant domain (CH1-CL) shares a very similar fold and interdomain orientation with the CH3-CH3 dimer. Thus, numerous well-established design efforts for CH3-CH3 interfaces, have also been applied to CH1-CL dimers to reduce the number of mispairings in the Fabs. Given the high structural similarity of the CH3-CH3 and CH1-CL domains we want to identify additional opportunities in comparing the differences and overlapping interaction profiles. Our vision is to facilitate a toolkit that allows for the interchangeable usage of different design tools from crosslinking the knowledge between these two interface types. As a starting point, here, we use classical molecular dynamics simulations to identify differences of the CH3-CH3 and CH1-CL interfaces and already find unexpected features of these interfaces shedding new light on possible design variations. Apart from identifying clear differences between the similar CH3-CH3 and CH1-CL dimers, we structurally characterize the effects of point-mutations in the CH3-CH3 interface on the respective dynamics and interface interaction patterns. Thus, this study has broad implications in the field of antibody engineering as it provides a structural and mechanistical understanding of antibody interfaces and thereby presents a crucial aspect for the design of bispecific antibodies.

6.
MAbs ; 13(1): 1993769, 2021.
Article in English | MEDLINE | ID: mdl-34711143

ABSTRACT

A growing body of evidence supports the important role of molecular charge on antibody pharmacokinetics (PK), yet a quantitative description of the effect of charge on systemic and tissue disposition of antibodies is still lacking. Consequently, we have systematically engineered complementarity-determining regions (CDRs) of trastuzumab to create a series of variants with an isoelectric point (pI) range of 6.3-8.9 and a variable region (Fv) charge range of -8.9 to +10.9 (at pH 5.5), and have investigated in vitro and in vivo disposition of these molecules. These monoclonal antibodies (mAbs) exhibited incrementally enhanced binding to cell surfaces and cellular uptake with increased positive charge in antigen-negative cells. After single intravenous dosing in mice, a bell-shaped relationship between systemic exposure and Fv charge was observed, with both extended negative and positive charge patches leading to more rapid nonspecific clearance. Whole-body PK experiments revealed that, although overall exposures of most variants in the tissues were very similar, positive charge of mAbs led to significantly enhanced tissue:plasma concentration ratios for most tissues. In well-perfused organs such as liver, spleen, and kidney, the positive charge variants show superior accumulation. In tissues with continuous capillaries such as fat, muscle, skin, and bone, plasma concentrations governed tissue exposures. The in vitro and in vivo disposition data presented here facilitate better understanding of the impact of charge modifications on antibody PK, and suggest that alteration in the charge may help to improve tissue:plasma concentration ratios for mAbs in certain tissues. The data presented here also paves the way for the development of physiologically based pharmacokinetic models of mAbs that incorporate charge variations.


Subject(s)
Antibodies, Monoclonal , Antineoplastic Agents, Immunological , Animals , Antigens , Complementarity Determining Regions , Isoelectric Point , Mice
7.
Front Immunol ; 12: 675655, 2021.
Article in English | MEDLINE | ID: mdl-34447370

ABSTRACT

Antibodies have emerged as one of the fastest growing classes of biotherapeutic proteins. To improve the rational design of antibodies, we investigate the conformational diversity of 16 different germline combinations, which are composed of 4 different kappa light chains paired with 4 different heavy chains. In this study, we systematically show that different heavy and light chain pairings strongly influence the paratope, interdomain interaction patterns and the relative VH-VL interface orientations. We observe changes in conformational diversity and substantial population shifts of the complementarity determining region (CDR) loops, resulting in distinct dominant solution structures and differently favored canonical structures. Additionally, we identify conformational changes in the structural diversity of the CDR-H3 loop upon different heavy and light chain pairings, as well as upon changes in sequence and structure of the neighboring CDR loops, despite having an identical CDR-H3 loop amino acid sequence. These results can also be transferred to all CDR loops and to the relative VH-VL orientation, as certain paratope states favor distinct interface angle distributions. Furthermore, we directly compare the timescales of sidechain rearrangements with the well-described transition kinetics of conformational changes in the backbone of the CDR loops. We show that sidechain flexibilities are strongly affected by distinct heavy and light chain pairings and decipher germline-specific structural features co-determining stability. These findings reveal that all CDR loops are strongly correlated and that distinct heavy and light chain pairings can result in different paratope states in solution, defined by a characteristic combination of CDR loop conformations and VH-VL interface orientations. Thus, these results have broad implications in the field of antibody engineering, as they clearly show the importance of considering paired heavy and light chains to understand the antibody binding site, which is one of the key aspects in the design of therapeutics.


Subject(s)
Binding Sites, Antibody , Germ Cells/immunology , Molecular Dynamics Simulation , Complementarity Determining Regions/chemistry , Humans , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Light Chains/chemistry , Immunoglobulin Variable Region/chemistry , Protein Conformation
8.
Mol Pharm ; 18(6): 2208-2217, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34014104

ABSTRACT

The current standard of care for antivascular endothelial growth factor (VEGF) treatment requires frequent intravitreal (IVT) injections of protein therapeutics, as a result of limited retention within the eye. A thorough understanding of the determinants of ocular pharmacokinetics (PK) and its translation across species is an essential prerequisite for developing more durable treatments. In this work, we studied the ocular PK in macaques of the protein formats that comprise today's anti-VEGF standard of care. Cynomolgus monkeys received a single IVT injection of a single-chain variable fragment (scFv, brolucizumab), antigen-binding fragment (Fab, ranibizumab), fragment crystallizable-fusion protein (Fc-fusion, aflibercept), or immunoglobulin G monoclonal antibody (IgG, VA2 CrossMAb). Drug concentrations were determined in aqueous humor samples collected up to 42 days postinjection using immunoassay methods. The ocular half-life (t1/2) was 2.28, 2.62, 3.13, and 3.26 days for scFv, Fab, Fc-fusion, and IgG, respectively. A correlation with human t1/2 values from the literature confirmed the translational significance of the cynomolgus monkey as an animal model for ocular research. The relation between ocular t1/2 and molecular size was also investigated. Size was inferred from the molecular weight (MW) or determined experimentally by dynamic light scattering. The MW and hydrodynamic radius were found to be good predictors for the ocular t1/2 of globular proteins. The analysis showed that molecular size is a determinant of ocular disposition and may be used in lieu of dedicated PK studies in animals.


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , Aqueous Humor/metabolism , Vitreous Body/metabolism , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/pharmacokinetics , Half-Life , Intravitreal Injections , Macaca fascicularis , Models, Animal , Molecular Weight , Ranibizumab/administration & dosage , Ranibizumab/chemistry , Ranibizumab/pharmacokinetics , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Receptors, Vascular Endothelial Growth Factor/chemistry , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics
9.
Nat Commun ; 12(1): 708, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33514724

ABSTRACT

We report the development of a platform of dual targeting Fab (DutaFab) molecules, which comprise two spatially separated and independent binding sites within the human antibody CDR loops: the so-called H-side paratope encompassing HCDR1, HCDR3 and LCDR2, and the L-side paratope encompassing LCDR1, LCDR3 and HCDR2. Both paratopes can be independently selected and combined into the desired bispecific DutaFabs in a modular manner. X-ray crystal structures illustrate that DutaFabs are able to bind two target molecules simultaneously at the same Fv region comprising a VH-VL heterodimer. In the present study, this platform is applied to generate DutaFabs specific for VEGFA and PDGF-BB, which show high affinities, physico-chemical stability and solubility, as well as superior efficacy over anti-VEGF monotherapy in vivo. These molecules exemplify the usefulness of DutaFabs as a distinct class of antibody therapeutics, which is currently being evaluated in patients.


Subject(s)
Antibodies, Bispecific/pharmacology , Choroidal Neovascularization/drug therapy , Drug Development/methods , Immunoglobulin Fab Fragments/pharmacology , Protein Engineering , Amino Acid Sequence/genetics , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/therapeutic use , Antibodies, Bispecific/ultrastructure , Becaplermin/antagonists & inhibitors , Binding Sites, Antibody/genetics , Crystallography, X-Ray , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/therapeutic use , Immunoglobulin Fab Fragments/ultrastructure , Inhibitory Concentration 50 , Intravitreal Injections , Male , Models, Molecular , Proof of Concept Study , Protein Conformation , Rats , Vascular Endothelial Growth Factor A/antagonists & inhibitors
10.
Biophys J ; 120(1): 143-157, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33220303

ABSTRACT

A major challenge in the development of antibody biotherapeutics is their tendency to aggregate. One root cause for aggregation is exposure of hydrophobic surface regions to the solvent. Many current techniques predict the relative aggregation propensity of antibodies via precalculated scales for the hydrophobicity or aggregation propensity of single amino acids. However, those scales cannot describe the nonadditive effects of a residue's surrounding on its hydrophobicity. Therefore, they are inherently limited in their ability to describe the impact of subtle differences in molecular structure on the overall hydrophobicity. Here, we introduce a physics-based approach to describe hydrophobicity in terms of the hydration free energy using grid inhomogeneous solvation theory (GIST). We apply this method to assess the effects of starting structures, conformational sampling, and protonation states on the hydrophobicity of antibodies. Our results reveal that high-quality starting structures, i.e., crystal structures, are crucial for the prediction of hydrophobicity and that conformational sampling can compensate errors introduced by the starting structure. On the other hand, sampling of protonation states only leads to good results when combined with high-quality structures, whereas it can even be detrimental otherwise. We conclude by pointing out that a single static homology model may not be adequate for predicting hydrophobicity.


Subject(s)
Amino Acids , Hydrophobic and Hydrophilic Interactions , Molecular Conformation , Molecular Structure , Solvents
11.
Mol Pharm ; 17(2): 695-709, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31876425

ABSTRACT

Therapeutic antibodies administered intravitreally are the current standard of care to treat retinal diseases. The ocular half-life (t1/2) is a key determinant of the duration of target suppression. To support the development of novel, longer-acting drugs, a reliable determination of t1/2 is needed together with an improved understanding of the factors that influence it. A model-based meta-analysis was conducted in humans and nonclinical species (rat, rabbit, monkey, and pig) to determine consensus values for the ocular t1/2 of IgG antibodies and Fab fragments. Results from multiple literature and in-house pharmacokinetic studies are presented within a mechanistic framework that assumes diffusion-controlled drug elimination from the vitreous. Our analysis shows, both theoretically and experimentally, that the ocular t1/2 increases in direct proportion to the product of the hydrodynamic radius of the macromolecule (3.0 nm for Fab and 5.0 nm for IgG) and the square of the radius of the vitreous globe, which varies approximately 24-fold from the rat to the human. Interspecies differences in the proportionality factors are observed and discussed in mechanistic terms. In addition, mathematical formulae are presented that allow prediction of the ocular t1/2 for molecules of interest. The utility of these formulae is successfully demonstrated in case studies of aflibercept, brolucizumab, and PEGylated Fabs, where the predicted ocular t1/2 values are found to be in reasonable agreement with the experimental data available for these molecules.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Biological Products/administration & dosage , Immunoglobulin Fab Fragments/administration & dosage , Immunoglobulin G/administration & dosage , Intravitreal Injections/methods , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Animals , Antibodies, Monoclonal, Humanized/pharmacokinetics , Biological Products/pharmacokinetics , Diffusion , Half-Life , Haplorhini , Humans , Hydrodynamics , Rabbits , Rats , Recombinant Fusion Proteins/pharmacokinetics , Retinal Diseases/drug therapy , Swine , Tissue Distribution , Vitreous Body/drug effects , Vitreous Body/metabolism
12.
MAbs ; 12(1): 1683432, 2020.
Article in English | MEDLINE | ID: mdl-31769731

ABSTRACT

The pharmacokinetic (PK) properties of therapeutic antibodies directly affect efficacy, dose and dose intervals, application route and tissue penetration. In indications where health-care providers and patients can choose between several efficacious and safe therapeutic options, convenience (determined by dosing interval or route of application), which is mainly driven by PK properties, can affect drug selection. Therapeutic antibodies can have greatly different PK even if they have identical Fc domains and show no target-mediated drug disposition. Biophysical properties like surface charge or hydrophobicity, and binding to surrogates for high abundant off-targets (e.g., baculovirus particles, Chinese hamster ovary cell membrane proteins) were proposed to be responsible for these differences. Here, we used heparin chromatography to separate a polyclonal mix of endogenous human IgGs (IVIG) into fractions that differ in their PK properties. Heparin was chosen as a surrogate for highly negatively charged glycocalyx components on endothelial cells, which are among the main contributors to nonspecific clearance. By directly correlating heparin retention time with clearance, we identified heparin chromatography as a tool to assess differences in unspecific cell-surface interaction and the likelihood for increased pinocytotic uptake and degradation. Building on these results, we combined predictors for FcRn-mediated recycling and cell-surface interaction. The combination of heparin and FcRn chromatography allow identification of antibodies with abnormal PK by mimicking the major root causes for fast, non-target-mediated, clearance of therapeutic, Fc-containing proteins.


Subject(s)
Chromatography/methods , Endothelial Cells/metabolism , Immunoglobulins, Intravenous/chemistry , Animals , CHO Cells , Cricetulus , Heparin/chemistry , Histocompatibility Antigens Class I/metabolism , Humans , Immunoglobulins, Intravenous/metabolism , Metabolic Clearance Rate , Pinocytosis , Protein Binding , Proteolysis , Receptors, Fc/metabolism
13.
Drug Metab Dispos ; 47(12): 1443-1456, 2019 12.
Article in English | MEDLINE | ID: mdl-31748266

ABSTRACT

For therapeutic proteins, the currently established standard development path generally does not foresee biotransformation studies by default because it is well known that the clearance of therapeutic proteins proceeds via degradation to small peptides and individual amino acids. In contrast to small molecules, there is no general need to identify enzymes involved in biotransformation because this information is not relevant for drug-drug interaction assessment and for understanding the clearance of a therapeutic protein. Nevertheless, there are good reasons to embark on biotransformation studies, especially for complex therapeutic proteins. Typical triggers are unexpected rapid clearance, species differences in clearance not following the typical allometric relationship, a mismatch in the pharmacokinetics/pharmacodynamics (PK/PD) relationship, and the need to understand observed differences between the results of multiple bioanalytical methods (e.g., total vs. target-binding competent antibody concentrations). Early on during compound optimization, knowledge on protein biotransformation may help to design more stable drug candidates with favorable in vivo PK properties. Understanding the biotransformation of a therapeutic protein may also support designing and understanding the bioanalytical assay and ultimately the PK/PD assessment. Especially in cases where biotransformation products are pharmacologically active, quantification and assessment of their contribution to the overall pharmacological effect can be important for establishing a PK/PD relationship and extrapolation to humans. With the increasing number of complex therapeutic protein formats, the need for understanding the biotransformation of therapeutic proteins becomes more urgent. This article provides an overview on biotransformation processes, proteases involved, strategic considerations, regulatory guidelines, literature examples for in vitro and in vivo biotransformation, and technical approaches to study protein biotransformation. SIGNIFICANCE STATEMENT: Understanding the biotransformation of complex therapeutic proteins can be crucial for establishing a pharmacokinetic/pharmacodynamic relationship. This article will highlight scientific, strategic, regulatory, and technological features of protein biotransformation.


Subject(s)
Pharmaceutical Preparations/metabolism , Proteins/pharmacokinetics , Small Molecule Libraries/pharmacokinetics , Animals , Biotransformation , Drug Interactions , Humans , Pharmaceutical Preparations/administration & dosage , Proteins/administration & dosage , Proteins/pharmacology , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/pharmacology
14.
MAbs ; 11(8): 1402-1414, 2019.
Article in English | MEDLINE | ID: mdl-31526159

ABSTRACT

High specificity accompanied with the ability to recruit immune cells has made recombinant therapeutic antibodies an integral part of drug development. Here we present a generic approach to generate two novel IgG-derived antibody formats that are based on a modification of the CrossMab technology. MoAbs harbor two heavy chains (HCs) resulting in one binding entity and one fragment crystallizable region (Fc), whereas DuoMabs are composed of four HCs harboring two binding entities and two Fc regions linked at a disulfide-bridged hinge. The latter bivalent format is characterized by avidity-enhanced target cell binding while simultaneously increasing the 'Fc-load' on the surface. DuoMabs were shown to be producible in high yield and purity and bind to surface cells with affinities comparable to IgGs. The increased Fc load directed at the surface of target cells by DuoMabs modulates their antibody-dependent cell-mediated cytotoxicity competency toward target cells, making them attractive for applications that require or are modulated by FcR interactions.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Antibodies, Bispecific/chemistry , Antibodies, Monoclonal/chemistry , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry
15.
J Labelled Comp Radiopharm ; 62(11): 751-757, 2019 09.
Article in English | MEDLINE | ID: mdl-31369163

ABSTRACT

The number of therapeutic antibodies in research and development as well as their complexity increases from year to year. Novel therapeutic protein formats, such as Fc-fusions, bispecific, or multivalent antibodies, are currently in preclinical and clinical development. Therefore, the need for biodistribution and imaging studies, eg, with radiolabeled proteins are very high. However, the labeling process or the label itself can have an impact on binding to cellular receptors, eg, to neonatal Fc receptor (FcRn), which can lead to altered PK properties compared with the unlabeled antibody. FcRn affinity chromatography allows the assessment of immunoglobulin G (IgG) samples with respect to their pH-dependent FcRn interaction. We analyzed IgGs with different types of labels, namely, direct iodination with 125 I; chelating agents, such as DOTA and DOTAM; and [3 H]propionate. Direct radio-iodination leads to shifts in FcRn column retention time, which might indicate a potentially faster clearance. Furthermore, high conjugation ratios of chelator lower the affinity to FcRn successively and thus may influence the lysosomal degradation of the antibody in endothelial cells. In contrast, IgGs labeled with [3 H]propionate did not show any timeshifts in FcRn affinity chromatography. This article is based on the oral presentation at the IIS 2018 Prague and highlights the importance of an affinity chromatography for characterization of potential changes in affinity to FcRn itself or charge and hydrophobicity.


Subject(s)
Histocompatibility Antigens Class I/immunology , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Receptors, Fc/immunology , Chelating Agents/chemistry , Halogenation , Isotope Labeling , Metals/chemistry , Propionates/chemistry , Radioisotopes/chemistry , Succinimides/chemistry
16.
Redox Biol ; 20: 146-156, 2019 01.
Article in English | MEDLINE | ID: mdl-30312900

ABSTRACT

The diphthamide modification of translation elongation factor 2 is highly conserved in eukaryotes and archaebacteria. Nevertheless, cells lacking diphthamide can carry out protein synthesis and are viable. We have analyzed the phenotypes of diphthamide deficient cells and found that diphthamide deficiency reduces selenocysteine incorporation into selenoproteins. Additional phenotypes resulting from diphthamide deficiency include altered tRNA-synthetase and selenoprotein transcript levels, hypersensitivity to oxidative stress and increased selenite tolerance. Diphthamide-eEF2 occupies the aminoacyl-tRNA translocation site at which UGA either stalls translation or decodes selenocysteine. Its position is in close proximity and mutually exclusive to the ribosomal binding site of release/recycling factor ABCE1, which harbors a redox-sensitive Fe-S cluster and, like diphthamide, is present in eukaryotes and archaea but not in eubacteria. Involvement of diphthamide in UGA-SECIS decoding may explain deregulated selenoprotein expression and as a consequence oxidative stress, NFkB activation and selenite tolerance in diphthamide deficient cells.


Subject(s)
Gene Expression Regulation/drug effects , Histidine/analogs & derivatives , Selenoproteins/genetics , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Cell Line, Tumor , Histidine/pharmacology , Humans , NF-kappa B/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Selenious Acid/pharmacology , Selenocysteine/metabolism , Selenoproteins/metabolism
17.
MAbs ; 8(4): 811-27, 2016.
Article in English | MEDLINE | ID: mdl-26984378

ABSTRACT

The epidermal growth factor receptor (EGFR) and the insulin-like growth factor-1 receptor (IGF-1R) play critical roles in tumor growth, providing a strong rationale for the combined inhibition of IGF-1R and EGFR signaling in cancer therapy. We describe the design, affinity maturation, in vitro and in vivo characterization of the bispecific anti-IGF-1R/EGFR antibody XGFR*. XGFR* is based on the bispecific IgG antibody XGFR, which enabled heterodimerization of an IGF-1R binding scFab heavy chain with an EGFR-binding light and heavy chain by the "knobs-into-holes" technology. XGFR* is optimized for monovalent binding of human EGFR and IGF-1R with increased binding affinity for IGF-1R due to affinity maturation and highly improved protein stability to oxidative and thermal stress. It bears an afucosylated Fc-portion for optimal induction of antibody-dependent cell-mediated cytotoxicity (ADCC). Stable Chinese hamster ovary cell clones with production yields of 2-3 g/L were generated, allowing for large scale production of the bispecific antibody. XGFR* potently inhibits EGFR- and IGF-1R-dependent receptor phosphorylation, reduces tumor cell proliferation in cells with heterogeneous levels of IGF-1R and EGFR receptor expression and induces strong ADCC in vitro. A comparison of pancreatic and colorectal cancer lines demonstrated superior responsiveness to XGFR*-mediated signaling and tumor growth inhibition in pancreatic cancers that frequently show a high degree of IGF-1R/EGFR co-expression. XGFR* showed potent anti-tumoral efficacy in the orthotopic MiaPaCa-2 pancreatic xenograft model, resulting in nearly complete tumor growth inhibition with significant number of tumor remissions. In summary, the bispecific anti-IGF-1R/EGFR antibody XGFR* combines potent signaling and tumor growth inhibition with enhanced ADCC induction and represents a clinical development candidate for the treatment of pancreatic cancer.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Pancreatic Neoplasms/immunology , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Antibodies, Bispecific/biosynthesis , Antibody Affinity , Antibody-Dependent Cell Cytotoxicity , CHO Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cricetinae , Cricetulus , Humans , Mice , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
18.
MAbs ; 8(3): 562-73, 2016.
Article in English | MEDLINE | ID: mdl-26864324

ABSTRACT

Vascular endothelial growth factor (VEGF)-A blockade has been validated clinically as a treatment for human cancers. Angiopoietin-2 (Ang-2) is a key regulator of blood vessel remodeling and maturation. In tumors, Ang-2 is up-regulated and an unfavorable prognostic factor. Recent data demonstrated that Ang-2 inhibition mediates anti-tumoral effects. We generated a tetravalent bispecific antibody (Ang-2-VEGF-TAvi6) targeting VEGF-A with 2 arms based on bevacizumab (Avastin®), and targeting Ang-2 with 2 arms based on a novel anti-Ang-2 antibody (LC06). The two Ang-2-targeting single-chain variable fragments are disulfide-stabilized and fused to the C-terminus of the heavy chain of bevacizumab. Treatment with Ang-2-VEGF-A-TAvi6 led to a complete abrogation of angiogenesis in the cornea micropocket assay. Metastatic spread and tumor growth of subcutaneous, orthotopic and anti-VEGF-A resistant tumors were also efficiently inhibited. These data further establish Ang-2-VEGF bispecific antibodies as a promising anti-angiogenic, anti-metastatic and anti-tumor agent for the treatment of cancer.


Subject(s)
Angiopoietin-2/antagonists & inhibitors , Antibodies, Bispecific , Antibodies, Neoplasm , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Experimental , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/pharmacology , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Mice , Neoplasm Metastasis , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Xenograft Model Antitumor Assays
19.
Int J Mol Sci ; 16(11): 27497-507, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26593903

ABSTRACT

TriFabs are IgG-shaped bispecific antibodies (bsAbs) composed of two regular Fab arms fused via flexible linker peptides to one asymmetric third Fab-sized binding module. This third module replaces the IgG Fc region and is composed of the variable region of the heavy chain (VH) fused to CH3 with "knob"-mutations, and the variable region of the light chain (VL) fused to CH3 with matching "holes". The hinge region does not contain disulfides to facilitate antigen access to the third binding site. To compensate for the loss of hinge-disulfides between heavy chains, CH3 knob-hole heterodimers are linked by S354C-Y349C disulphides, and VH and VL of the stem region may be linked via VH44C-VL100C disulphides. TriFabs which bind one antigen bivalent in the same manner as IgGs and the second antigen monovalent "in between" these Fabs can be applied to simultaneously engage two antigens, or for targeted delivery of small and large (fluorescent or cytotoxic) payloads.


Subject(s)
Antibodies, Bispecific , Immunoglobulin Fab Fragments , Immunoglobulin G , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Antibody Affinity/immunology , Binding Sites , Disulfides/chemistry , Drug Carriers , Drug Delivery Systems , Epitopes/immunology , Genetic Engineering , Humans , Immunoconjugates/immunology , Immunoconjugates/metabolism , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Protein Binding , Protein Multimerization , Protein Stability , Temperature
20.
Proc Natl Acad Sci U S A ; 112(19): 5997-6002, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25918417

ABSTRACT

Here, we investigated the influence of the variable fragment (Fv) of IgG antibodies on the binding to the neonatal Fc receptor (FcRn) as well as on FcRn-dependent pharmacokinetics (PK). FcRn plays a key role in IgG homeostasis, and specific manipulation in the crystallizable fragment (Fc) is known to affect FcRn-dependent PK. Although the influence of the antigen-binding fragment (Fab) on FcRn interactions has been reported, the underlying mechanism is hitherto only poorly understood. Therefore, we analyzed the two IgG1 antibodies, briakinumab and ustekinumab, that have similar Fc parts but different terminal half-lives in human and systematically engineered variants of them with cross-over exchanges and varied charge distribution. Using FcRn affinity chromatography, molecular dynamics simulation, and in vivo PK studies in human FcRn transgenic mice, we provide evidence that the charge distribution on the Fv domain is involved in excessive FcRn binding. This excessive binding prevents efficient FcRn-IgG dissociation at physiological pH, thereby reducing FcRn-dependent terminal half-lives. Furthermore, we observed a linear correlation between FcRn column retention times of the antibody variants and the terminal half-lives in vivo. Taken together, our study contributes to a better understanding of the FcRn-IgG interaction, and it could also provide profound potential in FcRn-dependent antibody engineering of the variable Fab region.


Subject(s)
Histocompatibility Antigens Class I/chemistry , Receptors, Fc/chemistry , Animals , Antibodies/chemistry , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized/chemistry , Antigen-Antibody Reactions , Chromatography, Affinity , Female , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen-Ion Concentration , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry , Mice , Mice, Transgenic , Microscopy, Confocal , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Protein Engineering , Protein Multimerization , Static Electricity , Surface Plasmon Resonance , Ustekinumab , beta 2-Microglobulin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...