Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Biol Chem ; 405(7-8): 461-470, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-38373142

ABSTRACT

In this work we have generated cattle-derived chimeric ultralong CDR-H3 antibodies targeting tumor necrosis factor α (TNF-α) via immunization and yeast surface display. We identified one particular ultralong CDR-H3 paratope that potently neutralized TNF-α. Interestingly, grafting of the knob architecture onto a peripheral loop of the CH3 domain of the Fc part of an IgG1 resulted in the generation of a TNF-α neutralizing Fc (Fcknob) that did not show any potency loss compared with the parental chimeric IgG format. Eventually, grafting this knob onto the CH3 region of adalimumab enabled the engineering of a novel TNF-α targeting antibody architecture displaying augmented TNF-α inhibition.


Subject(s)
Adalimumab , Tumor Necrosis Factor-alpha , Adalimumab/immunology , Adalimumab/pharmacology , Adalimumab/chemistry , Animals , Cattle , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Humans , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/pharmacology , Complementarity Determining Regions/immunology , Complementarity Determining Regions/chemistry
2.
J Immunol ; 209(9): 1724-1735, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36104113

ABSTRACT

In this work, we have generated novel Fc-comprising NK cell engagers (NKCEs) that bridge human NKp30 on NK cells to human epidermal growth factor receptor (EGFR) on tumor cells. Camelid-derived VHH single-domain Abs specific for human NKp30 and a humanized Fab derived from the EGFR-specific therapeutic Ab cetuximab were used as binding arms. By combining camelid immunization with yeast surface display, we were able to isolate a diverse panel of NKp30-specific VHHs against different epitopes on NKp30. Intriguingly, NKCEs built with VHHs that compete for binding to NKp30 with B7-H6, the natural ligand of NKp30, were significantly more potent in eliciting tumor cell lysis of EGFR-positive tumor cells than NKCEs harboring VHHs that target different epitopes on NKp30 from B7-H6. We demonstrate that the NKCEs can be further improved with respect to killing capabilities by concomitant engagement of FcγRIIIa and that soluble B7-H6 does not impede cytolytic capacities of all scrutinized NKCEs at significantly higher B7-H6 concentrations than observed in cancer patients. Moreover, we show that physiological processes requiring interactions between membrane-bound B7-H6 and NKp30 on NK cells are unaffected by noncompeting NKCEs still eliciting tumor cell killing at low picomolar concentrations. Ultimately, the NKCEs generated in this study were significantly more potent in eliciting NK cell-mediated tumor cell lysis than cetuximab and elicited a robust release of proinflammatory cytokines, both features which might be beneficial for antitumor therapy.


Subject(s)
Cytokines , Natural Cytotoxicity Triggering Receptor 3 , Humans , B7 Antigens/metabolism , Cell Death , Cetuximab/pharmacology , Epitopes , ErbB Receptors , Killer Cells, Natural , Ligands , Natural Cytotoxicity Triggering Receptor 3/metabolism
3.
Int J Mol Sci ; 25(4)2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38396776

ABSTRACT

The toolbox of modern antibody engineering allows the design of versatile novel functionalities exceeding nature's repertoire. Many bispecific antibodies comprise heterodimeric Fc portions recently validated through the approval of several bispecific biotherapeutics. While heterodimerization methodologies have been established for low-throughput large-scale production, few approaches exist to overcome the bottleneck of large combinatorial screening efforts that are essential for the identification of the best possible bispecific antibody. This report presents a novel, robust and miniaturized heterodimerization process based on controlled Fab-arm exchange (cFAE), which is applicable to a variety of heterodimeric formats and compatible with automated high-throughput screens. Proof of applicability was shown for two therapeutic molecule classes and two relevant functional screening read-outs. First, the miniaturized production of biparatopic anti-c-MET antibody-drug conjugates served as a proof of concept for their applicability in cytotoxic screenings on tumor cells with different target expression levels. Second, the automated workflow enabled a large unbiased combinatorial screening of biparatopic antibodies and the identification of hits mediating potent c-MET degradation. The presented workflow utilizes standard equipment and may serve as a facile, efficient and robust method for the discovery of innovative therapeutic agents in many laboratories worldwide.


Subject(s)
Antibodies, Bispecific , Immunoconjugates , Antibodies, Bispecific/therapeutic use , Immunoconjugates/pharmacology
4.
J Immunol ; 206(1): 225-236, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33268483

ABSTRACT

Activating NK cell receptors represent promising target structures to elicit potent antitumor immune responses. In this study, novel immunoligands were generated that bridge the activating NK cell receptor NKp30 on NK cells with epidermal growth factor receptor (EGFR) on tumor cells in a bispecific IgG-like format based on affinity-optimized versions of B7-H6 and the Fab arm derived from cetuximab. To enhance NKp30 binding, the solitary N-terminal IgV domain of B7-H6 (ΔB7-H6) was affinity matured by an evolutionary library approach combined with yeast surface display. Biochemical and functional characterization of 36 of these novel ΔB7-H6-derived NK cell engagers revealed an up to 45-fold-enhanced affinity for NKp30 and significantly improved NK cell-mediated, EGFR-dependent killing of tumor cells compared with the NK cell engager based on the wild-type ΔB7-H6 domain. In this regard, potencies (EC50 killing) of the best immunoligands were substantially improved by up to 87-fold. Moreover, release of IFN-γ and TNF-α was significantly increased. Importantly, equipment of the ΔB7-H6-based NK cell engagers with a human IgG1 Fc part competent in Fc receptor binding resulted in an almost 10-fold superior killing of EGFR-overexpressing tumor cells compared with molecules either triggering FcγRIIIa or NKp30. Additionally, INF-γ and TNF-α release was increased compared with molecules solely triggering FcγRIIIa, including the clinically approved Ab cetuximab. Thus, incorporating affinity-matured ligands for NK cell-activating receptors might represent an effective strategy for the generation of potent novel therapeutic agents with unique effector functions in cancer immunotherapy.


Subject(s)
B7 Antigens/metabolism , Immunotherapy/methods , Killer Cells, Natural/immunology , Natural Cytotoxicity Triggering Receptor 3/metabolism , Neoplasms/immunology , Antibodies, Bispecific/genetics , Antibodies, Bispecific/metabolism , B7 Antigens/genetics , Cell Line, Tumor , Cetuximab/genetics , Cytokines/metabolism , Cytotoxicity, Immunologic , ErbB Receptors/immunology , ErbB Receptors/metabolism , Genetic Engineering , Humans , Immunoglobulin Fab Fragments/genetics , Inflammation Mediators/metabolism , Killer Cells, Natural/transplantation , Lymphocyte Activation , Natural Cytotoxicity Triggering Receptor 3/immunology , Neoplasms/therapy , Protein Binding , Signal Transduction
5.
Biol Chem ; 403(5-6): 455-477, 2022 04 26.
Article in English | MEDLINE | ID: mdl-33759431

ABSTRACT

Antibody display technologies enable the successful isolation of antigen-specific antibodies with therapeutic potential. The key feature that facilitates the selection of an antibody with prescribed properties is the coupling of the protein variant to its genetic information and is referred to as genotype phenotype coupling. There are several different platform technologies based on prokaryotic organisms as well as strategies employing higher eukaryotes. Among those, phage display is the most established system with more than a dozen of therapeutic antibodies approved for therapy that have been discovered or engineered using this approach. In recent years several other technologies gained a certain level of maturity, most strikingly mammalian display. In this review, we delineate the most important selection systems with respect to antibody generation with an emphasis on recent developments.


Subject(s)
Antibodies , Peptide Library , Animals , Antibodies/genetics , Antibodies/therapeutic use , Mammals/genetics
6.
Clin Exp Immunol ; 209(1): 22-32, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35325068

ABSTRACT

Natural killer (NK) cells exert an important role in cancer immune surveillance. Recognition of malignant cells and controlled activation of effector functions are facilitated by the expression of activating and inhibitory receptors, which is a complex interplay that allows NK cells to discriminate malignant cells from healthy tissues. Due to their unique profile of effector functions, the recruitment of NK cells is attractive in cancer treatment and a key function of NK cells in antibody therapy is widely appreciated. In recent years, besides the low-affinity fragment crystallizable receptor for immunoglobulin G (FcγRIIIA), the activating natural killer receptors p30 (NKp30) and p46 (NKp46), as well as natural killer group 2 member D (NKG2D), have gained increasing attention as potential targets for bispecific antibody-derivatives to redirect NK cell cytotoxicity against tumors. Beyond modulation of the receptor activity on NK cells, therapeutic targeting of the respective ligands represents an attractive approach. Here, novel therapeutic approaches to unleash NK cells by engagement of activating NK-cell receptors and alternative strategies targeting their tumor-expressed ligands in cancer therapy are summarized.


Subject(s)
Immunotherapy , Neoplasms , Receptors, Natural Killer Cell , Humans , Killer Cells, Natural , Ligands , NK Cell Lectin-Like Receptor Subfamily K/metabolism
7.
Int J Mol Sci ; 21(18)2020 Sep 08.
Article in English | MEDLINE | ID: mdl-32911608

ABSTRACT

Recent years have shown a tremendous increase and diversification in antibody-based therapeutics with advances in production techniques and formats. The plethora of currently investigated bi- to multi-specific antibody architectures can be harnessed to elicit a broad variety of specific modes of actions in oncology and immunology, spanning from enhanced selectivity to effector cell recruitment, all of which cannot be addressed by monospecific antibodies. Despite continuously growing efforts and methodologies, the identification of an optimal bispecific antibody as the best possible combination of two parental monospecific binders, however, remains challenging, due to tedious cloning and production, often resulting in undesired extended development times and increased expenses. Although automated high throughput screening approaches have matured for pharmaceutical small molecule development, it was only recently that protein bioconjugation technologies have been developed for the facile generation of bispecific antibodies in a 'plug and play' manner. In this review, we provide an overview of the most relevant methodologies for bispecific screening purposes-the DuoBody concept, paired light chain single cell production approaches, Sortase A and Transglutaminase, the SpyTag/SpyCatcher system, and inteins-and elaborate on the benefits as well as drawbacks of the different technologies.


Subject(s)
Antibodies, Bispecific/analysis , Antibodies, Bispecific/immunology , High-Throughput Screening Assays/methods , Animals , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Immunoglobulin G/metabolism , Protein Engineering/methods
8.
Biol Chem ; 400(3): 383-393, 2019 02 25.
Article in English | MEDLINE | ID: mdl-30465712

ABSTRACT

Antibodies can be successfully engineered and isolated by yeast or phage display of combinatorial libraries. Still, generation of libraries comprising heavy chain as well as light chain diversities is a cumbersome process involving multiple steps. Within this study, we set out to compare the output of yeast display screening of antibody Fab libraries from immunized rodents that were generated by Golden Gate Cloning (GGC) with the conventional three-step method of individual heavy- and light-chain sub-library construction followed by chain combination via yeast mating (YM). We demonstrate that the GGC-based one-step process delivers libraries and antibodies from heavy- and light-chain diversities with similar quality to the traditional method while being significantly less complex and faster. Additionally, we show that this method can also be used to successfully screen and isolate chimeric chicken/human antibodies following avian immunization.


Subject(s)
Immunoglobulin Heavy Chains/analysis , Immunoglobulin Light Chains/analysis , Saccharomyces cerevisiae/chemistry , Animals , Chickens , Cloning, Molecular , Humans , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/immunology , Peptide Library , Protein Engineering , Saccharomyces cerevisiae/immunology , Surface Properties
9.
Microb Cell Fact ; 17(1): 3, 2018 Jan 09.
Article in English | MEDLINE | ID: mdl-29316915

ABSTRACT

BACKGROUND: Yeast surface display (YSD) has proven to be a versatile platform technology for antibody discovery. However, the construction of antibody Fab libraries typically is a tedious three-step process that involves the generation of heavy chain as well as light chain display plasmids in different haploid yeast strains followed by yeast mating. RESULTS: Within this study, we aimed at implementing a focused Golden Gate Cloning approach for the generation of YSD libraries. For this, antibodies heavy and light chains were encoded on one single plasmid. Fab display on yeast cells was either mediated by a two-directional promoter system (2dir) or by ribosomal skipping (bicis). The general applicability of this methodology was proven by the functional display of a therapeutic antibody. Subsequently, we constructed large antibody libraries with heavy chain diversities derived from CEACAM5 immunized animals in combination with a common light chain. Target-specific antibodies from both display systems were readily obtained after three rounds of fluorescence activated cell sorting. Isolated variants exhibited high affinities in the nanomolar and subnanomolar range as well as appropriate biophysical properties. CONCLUSION: We demonstrated that Golden Gate Cloning appears to be a valid tool for the generation of large yeast surface display antibody Fab libraries. This procedure simplifies the hit discovery process of antibodies from immune repertoires.


Subject(s)
Cloning, Molecular/methods , Immunoglobulin Fab Fragments/isolation & purification , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Antibodies/metabolism , Antibody Affinity , Flow Cytometry , Immunoglobulin Fab Fragments/immunology , Peptide Library , Promoter Regions, Genetic , Surface Properties
10.
Biol Chem ; 403(5-6): 453, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35355499

Subject(s)
Protein Engineering
11.
Immunopharmacol Immunotoxicol ; 38(1): 21-8, 2016.
Article in English | MEDLINE | ID: mdl-26551147

ABSTRACT

Single-domain antibodies are the smallest antigen-binding units of antibodies, consisting either only of one variable domain or one engineered constant domain that solely facilitates target binding. This class of antibody derivatives comprises naturally occurring variable domains derived from camelids and sharks as well as engineered human variable or constant antibody domains of the heavy or light chain. Because of their high affinity and specificity as well as stability, small size and benefit of multiple re-formatting opportunities, those molecules emerged as promising candidates for biomedical applications and some of these entities have already proven to be successful in clinical development.


Subject(s)
Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Animals , Camelus/genetics , Camelus/immunology , Humans , Sharks/genetics , Sharks/immunology , Single-Chain Antibodies/genetics
12.
Angew Chem Int Ed Engl ; 55(16): 5085-9, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-26991930

ABSTRACT

Multivalent ligands of death receptors hold particular promise as tumor cell-specific therapeutic agents because they induce an apoptotic cascade in cancerous cells. Herein, we present a modular approach to generate death receptor 5 (DR5) binding constructs comprising multiple copies of DR5 targeting peptide (DR5TP) covalently bound to biomolecular scaffolds of peptidic nature. This strategy allows for efficient oligomerization of synthetic DR5TP-derived peptides in different spatial orientations using a set of enzyme-promoted conjugations or recombinant production. Heptameric constructs based on a short (60-75 residues) scaffold of a C-terminal oligomerization domain of human C4b binding protein showed remarkable proapoptotic activity (EC50=3 nm) when DR5TP was ligated to its carboxy terminus. Our data support the notion that inter-ligand distance, relative spatial orientation and copy number of receptor-binding modules are key prerequisites for receptor activation and cell killing.


Subject(s)
Apoptosis , Peptides/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Humans
13.
Angew Chem Int Ed Engl ; 53(46): 12618-23, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25070515

ABSTRACT

The immobilization of bioactive molecules onto nanocellulose leads to constructs that combine the properties of the grafted compounds with the biocompatibility and low cytotoxicity of cellulose carriers and the advantages given by their nanometer dimensions. However, the methods commonly used for protein grafting suffer from lack of selectivity, long reaction times, nonphysiological pH ranges and solvents, and the necessity to develop a tailor-made reaction strategy for each individual case. To overcome these restrictions, a generic two-step procedure was developed that takes advantage of the highly efficient oxime ligation combined with enzyme-mediated protein coupling onto the surface of peptide-modified crystalline nanocellulose. The described method is based on efficient and orthogonal transformations, requires no organic solvents, and takes place under physiological conditions. Being site-directed and regiospecific, it could be applied to a vast number of functional proteins.


Subject(s)
Cellulose/chemistry , Immobilized Proteins/chemistry , Nanoparticles/chemistry , Humans , Models, Molecular , Nanoparticles/ultrastructure , Oximes/chemistry , Peptides/chemistry , Surface Properties
14.
Cancer Treat Res Commun ; 39: 100805, 2024.
Article in English | MEDLINE | ID: mdl-38492435

ABSTRACT

BACKGROUND: Targeting the costimulatory receptor CD137 has shown promise as a therapeutic approach for cancer immunotherapy, resulting in anti-tumor efficacy demonstrated in clinical trials. However, the initial CD137 agonistic antibodies, urelumab and utomilumab, faced challenges in clinical trials due to the liver toxicity or lack of efficacy, respectively. Concurrently, c-MET has been identified as a highly expressed tumor-associated antigen (TAA) in various solid and soft tumors. METHODS: In this study, we aimed to develop a bispecific antibody (BsAb) that targets both c-MET and CD137, optimizing the BsAb format and CD137 binder for efficient delivery of the CD137 agonist to the tumor microenvironment (TME). We employed a monovalent c-MET motif and a trimeric CD137 Variable Heavy domain of Heavy chain (VHH) for the BsAb design. RESULTS: Our results demonstrate that the c-MET x CD137 BsAb provides co-stimulation to T cells through cross-linking by c-MET-expressing tumor cells. Functional immune assays confirmed the enhanced efficacy and potency of the c-MET x CD137 BsAb, as indicated by activation of CD137 signaling, target cell killing, and cytokine release in various tumor cell lines. Furthermore, the combination of c-MET x CD137 BsAb with Pembrolizumab showed a dose-dependent enhancement of target-induced T cell cytokine release. CONCLUSION: Overall, the c-MET x CD137 BsAb exhibits a promising developability profile as a tumor-targeted immune agonist by minimizing off-target effects while effectively delivering immune agonism. It has the potential to overcome resistance to anti-PD-(L)1 therapies.


Subject(s)
Antibodies, Bispecific , Immunotherapy , Proto-Oncogene Proteins c-met , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Animals , Humans , Mice , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Cell Line, Tumor , Immunotherapy/methods , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/therapy , Proto-Oncogene Proteins c-met/immunology , Tumor Microenvironment/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
15.
MAbs ; 16(1): 2302386, 2024.
Article in English | MEDLINE | ID: mdl-38214660

ABSTRACT

Optimal combinations of paratopes assembled into a biparatopic antibody have the capacity to mediate high-grade target cross-linking on cell membranes, leading to degradation of the target, as well as antibody and payload delivery in the case of an antibody-drug conjugate (ADC). In the work presented here, molecular docking suggested a suitable paratope combination targeting c-MET, but hydrophobic patches in essential binding regions of one moiety necessitated engineering. In addition to rational design of HCDR2 and HCDR3 mutations, site-specific spiking libraries were generated and screened in yeast and mammalian surface display approaches. Comparative analyses revealed similar positions amendable for hydrophobicity reduction, with a broad combinatorial diversity obtained from library outputs. Optimized variants showed high stability, strongly reduced hydrophobicity, retained affinities supporting the desired functionality and enhanced producibility. The resulting biparatopic anti-c-MET ADCs were comparably active on c-MET expressing tumor cell lines as REGN5093 exatecan DAR6 ADC. Structural molecular modeling of paratope combinations for preferential inter-target binding combined with protein engineering for manufacturability yielded deep insights into the capabilities of rational and library approaches. The methodologies of in silico hydrophobicity identification and sequence optimization could serve as a blueprint for rapid development of optimal biparatopic ADCs targeting further tumor-associated antigens in the future.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Animals , Immunoconjugates/genetics , Immunoconjugates/chemistry , Molecular Docking Simulation , Cell Line, Tumor , Hydrophobic and Hydrophilic Interactions , Mammals
16.
MAbs ; 16(1): 2315640, 2024.
Article in English | MEDLINE | ID: mdl-38372053

ABSTRACT

Natural killer (NK) cells emerged as a promising effector population that can be harnessed for anti-tumor therapy. In this work, we constructed NK cell engagers (NKCEs) based on NKp30-targeting single domain antibodies (sdAbs) that redirect the cytotoxic potential of NK cells toward epidermal growth factor receptor (EGFR)-expressing tumor cells. We investigated the impact of crucial parameters such as sdAb location, binding valencies, the targeted epitope on NKp30, and the overall antibody architecture on the redirection capacity. Our study exploited two NKp30-specific sdAbs, one of which binds a similar epitope on NKp30 as its natural ligand B7-H6, while the other sdAb addresses a non-competing epitope. For EGFR-positive tumor targeting, humanized antigen-binding domains of therapeutic antibody cetuximab were used. We demonstrate that NKCEs bivalently targeting EGFR and bivalently engaging NKp30 are superior to monovalent NKCEs in promoting NK cell-mediated tumor cell lysis and that the architecture of the NKCE can substantially influence killing capacities depending on the NKp30-targeting sdAb utilized. While having a pronounced impact on NK cell killing efficacy, the capabilities of triggering antibody-dependent cellular phagocytosis or complement-dependent cytotoxicity were not significantly affected comparing the bivalent IgG-like NKCEs with cetuximab. However, the fusion of sdAbs can have a slight impact on the NK cell release of immunomodulatory cytokines, as well as on the pharmacokinetic profile of the NKCE due to unfavorable spatial orientation within the molecule architecture. Ultimately, our findings reveal novel insights for the engineering of potent NKCEs triggering the NKp30 axis.


Subject(s)
Epidermal Growth Factor , Killer Cells, Natural , Cetuximab/metabolism , Epidermal Growth Factor/metabolism , Binding Sites, Antibody , ErbB Receptors/metabolism , Epitopes/metabolism
17.
Methods Mol Biol ; 2681: 161-173, 2023.
Article in English | MEDLINE | ID: mdl-37405648

ABSTRACT

In vitro antibody display libraries have emerged as powerful tools for a streamlined discovery of novel antibody binders. While in vivo antibody repertoires are matured and selected as a specific pair of variable heavy and light chains (VH and VL) with optimal specificity and affinity, during the recombinant generation of in vitro libraries, the native sequence pairing is not maintained. Here we describe a cloning method that combines the flexibility and versatility of in vitro antibody display with the advantages of natively paired VH-VL antibodies. In this regard, VH-VL amplicons are cloned via a two-step Golden Gate cloning procedure, allowing the display of Fab fragments on yeast cells.


Subject(s)
Antibodies , Immunoglobulin Fab Fragments , Immunoglobulin Fab Fragments/genetics , Cloning, Molecular , Peptide Library
18.
Methods Mol Biol ; 2702: 227-243, 2023.
Article in English | MEDLINE | ID: mdl-37679622

ABSTRACT

The adaptive immune system of sharks comprises a unique heavy chain-only antibody isotype, termed immunoglobulin new antigen receptor (IgNAR), in which antigen binding is mediated by a single variable domain, referred to as vNAR. In recent years, efforts were made to harness these domains for biomedical and biotechnological applications particularly due to their high affinity and specificity combined with a small size and high stability. Herein, we describe protocols for the construction of semisynthetic, CDR3-randomized vNAR libraries for the isolation of target-specific paratopes by yeast surface display. Additionally, we provide guidance for affinity maturation of a panel of antigen-enriched vNAR domains through CDR1 diversification of the FACS-selected, antigen-enriched population and sublibrary establishment.


Subject(s)
Antibodies , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Receptors, Antigen, B-Cell , Antibodies, Fungal , Immunoglobulin Isotypes , Immunoglobulin Heavy Chains
19.
Methods Mol Biol ; 2681: 47-60, 2023.
Article in English | MEDLINE | ID: mdl-37405642

ABSTRACT

Since its development in the 1980s, the Nobel Prize-awarded phage display technology has been one of the most commonly used in vitro selection technologies for the discovery of therapeutic and diagnostic antibodies. Besides the importance of selection strategy, one key component of the successful isolation of highly specific recombinant antibodies is the construction of high-quality phage display libraries. However, previous cloning protocols relied on a tedious multistep process with subsequent cloning steps for the introduction of first heavy and then light chain variable genetic antibody fragments (VH and VL). This resulted in reduced cloning efficiency, higher frequency of missing VH or VL sequences, as well as truncated antibody fragments. With the emergence of Golden Gate Cloning (GGC) for the generation of antibody libraries, the possibility of more facile library cloning has arisen. Here, we describe a streamlined one-step GGC strategy for the generation of camelid heavy chain only variable phage display libraries as well as the simultaneous introduction of heavy chain and light chain variable regions from the chicken into a scFv phage display vector.


Subject(s)
Bacteriophages , Single-Chain Antibodies , Peptide Library , Cell Surface Display Techniques/methods , Recombinant Proteins/genetics , Immunoglobulin Light Chains/genetics , Antibodies/genetics , Bacteriophages/genetics , Immunoglobulin Fragments/genetics , Single-Chain Antibodies/genetics , Cloning, Molecular
20.
MAbs ; 15(1): 2261149, 2023.
Article in English | MEDLINE | ID: mdl-37766540

ABSTRACT

In this study, we generated a novel library approach for high throughput de novo identification of humanized single-domain antibodies following camelid immunization. To achieve this, VHH-derived complementarity-determining regions-3 (CDR3s) obtained from an immunized llama (Lama glama) were grafted onto humanized VHH backbones comprising moderately sequence-diversified CDR1 and CDR2 regions similar to natural immunized and naïve antibody repertoires. Importantly, these CDRs were tailored toward favorable in silico developability properties, by considering human-likeness as well as excluding potential sequence liabilities and predicted immunogenic motifs. Target-specific humanized single-domain antibodies (sdAbs) were readily obtained by yeast surface display. We demonstrate that, by exploiting this approach, high affinity sdAbs with an optimized in silico developability profile can be generated. These sdAbs display favorable biophysical, biochemical, and functional attributes and do not require any further sequence optimization. This approach is generally applicable to any antigen upon camelid immunization and has the potential to significantly accelerate candidate selection and reduce risks and attrition rates in sdAb development.


Subject(s)
Single-Domain Antibodies , Humans , Immunization , Gene Library , Antigens , Complementarity Determining Regions/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL