1
|
Seo H, Hirota K, Ohta K. Molecular mechanisms of avian immunoglobulin gene diversification and prospect for industrial applications. Front Immunol 2024; 15:1453833. [PMID: 39346918 PMCID: PMC11427246 DOI: 10.3389/fimmu.2024.1453833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
Poultry immunoglobulin genes undergo diversification through homologous recombination (HR) and somatic hypermutation (SHM). Most animals share a similar system in immunoglobulin diversification, with the rare exception that human and murine immunoglobulin genes diversify through V(D)J recombination. Poultry possesses only one functional variable gene for each immunoglobulin heavy (HC) and light chains (LC), with clusters of non-productive pseudogenes upstream. During the B cell development, the functional variable gene is overwritten by sequences from the pseudo-variable genes via a process known as gene conversion (GC), a kind of HR. Point mutations caused in the functional variable gene also contribute to immunoglobulin diversification. This review discusses the latest findings on the molecular mechanisms of antibody gene diversification in poultry, using chickens as a model. Additionally, it will outline how these basic research findings have recently been applied especially in the medical field.
Collapse
Affiliation(s)
- Hidetaka Seo
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Kouji Hirota
- Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Kunihiro Ohta
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
2
|
Leighton PA, Ching K, Reynolds K, Vuong CN, Zeng B, Zhang Y, Gupta A, Morales J, Rivera GS, Srivastava DB, Cotter R, Pedersen D, Collarini E, Izquierdo S, van de Lavoir MC, Harriman W. Chickens with a Truncated Light Chain Transgene Express Single-Domain H Chain-Only Antibodies. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1744-1753. [PMID: 38629917 PMCID: PMC11102025 DOI: 10.4049/jimmunol.2300617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 03/22/2024] [Indexed: 05/20/2024]
Abstract
H chain-only Igs are naturally produced in camelids and sharks. Because these Abs lack the L chain, the Ag-binding domain is half the size of a traditional Ab, allowing this type of Ig to bind to targets in novel ways. Consequently, the H chain-only single-domain Ab (sdAb) structure has the potential to increase the repertoire and functional range of an active humoral immune system. The majority of vertebrates use the standard heterodimeric (both H and L chains) structure and do not produce sdAb format Igs. To investigate if other animals are able to support sdAb development and function, transgenic chickens (Gallus gallus) were designed to produce H chain-only Abs by omitting the L chain V region and maintaining only the LC region to serve as a chaperone for Ab secretion from the cell. These birds produced 30-50% normal B cell populations within PBMCs and readily expressed chicken sequence sdAbs. Interestingly, the H chains contained a spontaneous CH1 deletion. Although no isotype switching to IgY or IgA occurred, the IgM repertoire was diverse, and immunization with a variety of protein immunogens rapidly produced high and specific serum titers. mAbs of high affinity were efficiently recovered by single B cell screening. In in vitro functional assays, the sdAbs produced by birds immunized against SARS-CoV-2 were also able to strongly neutralize and prevent viral replication. These data suggest that the truncated L chain design successfully supported sdAb development and expression in chickens.
Collapse
|
3
|
Thomas B, Chockalingam K, Chen Z. Methods for Engineering Binders to Multi-Pass Membrane Proteins. Bioengineering (Basel) 2023; 10:1351. [PMID: 38135942 PMCID: PMC10741020 DOI: 10.3390/bioengineering10121351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/11/2023] [Accepted: 11/19/2023] [Indexed: 12/24/2023] Open
Abstract
Numerous potential drug targets, including G-protein-coupled receptors and ion channel proteins, reside on the cell surface as multi-pass membrane proteins. Unfortunately, despite advances in engineering technologies, engineering biologics against multi-pass membrane proteins remains a formidable task. In this review, we focus on the different methods used to prepare/present multi-pass transmembrane proteins for engineering target-specific biologics such as antibodies, nanobodies and synthetic scaffold proteins. The engineered biologics exhibit high specificity and affinity, and have broad applications as therapeutics, probes for cell staining and chaperones for promoting protein crystallization. We primarily cover publications on this topic from the past 10 years, with a focus on the different formats of multi-pass transmembrane proteins. Finally, the remaining challenges facing this field and new technologies developed to overcome a number of obstacles are discussed.
Collapse
Affiliation(s)
- Benjamin Thomas
- Interdisciplinary Graduate Program in Genetics and Genomics, Texas A&M University, College Station, TX 77845, USA;
| | - Karuppiah Chockalingam
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA;
| | - Zhilei Chen
- Interdisciplinary Graduate Program in Genetics and Genomics, Texas A&M University, College Station, TX 77845, USA;
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, TX 77807, USA;
| |
Collapse
|
4
|
Chen YC, Saito D, Suzuki T, Takemoto T. An inducible germ cell ablation chicken model for high-grade germline chimeras. Development 2023; 150:dev202079. [PMID: 37665168 PMCID: PMC10560566 DOI: 10.1242/dev.202079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/24/2023] [Indexed: 09/05/2023]
Abstract
Chicken embryos are a powerful and widely used animal model in developmental biology studies. Since the development of CRISPR technology, gene-edited chickens have been generated by transferring primordial germ cells (PGCs) into recipients after genetic modifications. However, low inheritance caused by competition between host germ cells and the transferred cells is a common complication and greatly reduces production efficiency. Here, we generated a gene-edited chicken, in which germ cells can be ablated in a drug-dependent manner, as recipients for gene-edited PGC transfer. We used the nitroreductase/metronidazole (NTR/Mtz) system for cell ablation, in which nitroreductase produces cytotoxic alkylating agents from administered metronidazole, causing cell apoptosis. The chicken Vasa homolog (CVH) gene locus was used to drive the expression of the nitroreductase gene in a germ cell-specific manner. In addition, a fluorescent protein gene, mCherry, was also placed in the CVH locus to visualize the PGCs. We named this system 'germ cell-specific autonomous removal induction' (gSAMURAI). gSAMURAI chickens will be an ideal recipient to produce offspring derived from transplanted exogenous germ cells.
Collapse
Affiliation(s)
- Yi-Chen Chen
- Division of Research and Development, Setsuro Tech Inc., Tokushima 770-8503, Japan
- Laboratory for Embryology, Institute for Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Daisuke Saito
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Takayuki Suzuki
- Department of Biology, Graduate School of Science, Osaka Metropolitan University, Osaka 558-8585, Japan
| | - Tatsuya Takemoto
- Division of Research and Development, Setsuro Tech Inc., Tokushima 770-8503, Japan
- Laboratory for Embryology, Institute for Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| |
Collapse
|
5
|
Harwardt J, Carrara SC, Bogen JP, Schoenfeld K, Grzeschik J, Hock B, Kolmar H. Generation of a symmetrical trispecific NK cell engager based on a two-in-one antibody. Front Immunol 2023; 14:1170042. [PMID: 37081888 PMCID: PMC10110854 DOI: 10.3389/fimmu.2023.1170042] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023] Open
Abstract
To construct a trispecific IgG-like antibody at least three different binding moieties need to be combined, which results in a complex architecture and challenging production of these molecules. Here we report for the first time the construction of trispecific natural killer cell engagers based on a previously reported two-in-one antibody combined with a novel anti-CD16a common light chain module identified by yeast surface display (YSD) screening of chicken-derived immune libraries. The resulting antibodies simultaneously target epidermal growth factor receptor (EGFR), programmed death-ligand 1 (PD-L1) and CD16a with two Fab fragments, resulting in specific cellular binding properties on EGFR/PD-L1 double positive tumor cells and a potent ADCC effect. This study paves the way for further development of multispecific therapeutic antibodies derived from avian immunization with desired target combinations, valencies, molecular symmetries and architectures.
Collapse
Affiliation(s)
- Julia Harwardt
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Stefania C. Carrara
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Biologics Technology and Development, Ferring Darmstadt Laboratory, Darmstadt, Germany
| | - Jan P. Bogen
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Biologics Technology and Development, Ferring Darmstadt Laboratory, Darmstadt, Germany
| | - Katrin Schoenfeld
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Julius Grzeschik
- Biologics Technology and Development, Ferring Biologics Innovation Centre, Epalinges, Switzerland
| | - Björn Hock
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany
- *Correspondence: Harald Kolmar,
| |
Collapse
|
6
|
El-Kafrawy SA, Abbas AT, Oelkrug C, Tahoon M, Ezzat S, Zumla A, Azhar EI. IgY antibodies: The promising potential to overcome antibiotic resistance. Front Immunol 2023; 14:1065353. [PMID: 36742328 PMCID: PMC9896010 DOI: 10.3389/fimmu.2023.1065353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 01/05/2023] [Indexed: 01/22/2023] Open
Abstract
Antibiotic resistant bacteria are a growing threat to global health security. Whilst the emergence of antimicrobial resistance (AMR) is a natural phenomenon, it is also driven by antibiotic exposure in health care, agriculture, and the environment. Antibiotic pressure and inappropriate use of antibiotics are important factors which drive resistance. Apart from their use to treat bacterial infections in humans, antibiotics also play an important role in animal husbandry. With limited antibiotic options, alternate strategies are required to overcome AMR. Passive immunization through oral, nasal and topical administration of egg yolk-derived IgY antibodies from immunized chickens were recently shown to be effective for treating bacterial infections in animals and humans. Immunization of chickens with specific antigens offers the possibility of creating specific antibodies targeting a wide range of antibiotic-resistant bacteria. In this review, we describe the growing global problem of antimicrobial resistance and highlight the promising potential of the use of egg yolk IgY antibodies for the treatment of bacterial infections, particularly those listed in the World Health Organization priority list.
Collapse
Affiliation(s)
- Sherif A El-Kafrawy
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Clinical Pathology, National Liver Institute, Menoufia University, Shebin El-Kom, Egypt
| | - Aymn T Abbas
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Biotechnology Research Laboratories, Gastroenterology, Surgery Centre, Mansoura University, Mansoura, Egypt
| | | | - Marwa Tahoon
- Epidemiology and Preventive Medicine Department, National Liver Institute, Menoufia University, Shebin El-Kom, Egypt
| | - Sameera Ezzat
- Epidemiology and Preventive Medicine Department, National Liver Institute, Menoufia University, Shebin El-Kom, Egypt.,MARC for Medical Services and Scientific Research, 6th of October City, Giza, Egypt
| | - Alimuddin Zumla
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Infection, Division of Infection and Immunity, Centre for Clinical Microbiology, University College London, London, United Kingdom.,National Institute for Health and Care Research (NIHR) Biomedical Research Centre, University College London Hospitals, London, United Kingdom
| | - Esam I Azhar
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
7
|
Banik SSR, Kushnir N, Doranz BJ, Chambers R. Breaking barriers in antibody discovery: harnessing divergent species for accessing difficult and conserved drug targets. MAbs 2023; 15:2273018. [PMID: 38050985 DOI: 10.1080/19420862.2023.2273018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/16/2023] [Indexed: 12/07/2023] Open
Abstract
To exploit highly conserved and difficult drug targets, including multipass membrane proteins, monoclonal antibody discovery efforts increasingly rely on the advantages offered by divergent species such as rabbits, camelids, and chickens. Here, we provide an overview of antibody discovery technologies, analyze gaps in therapeutic antibodies that stem from the historic use of mice, and examine opportunities to exploit previously inaccessible targets through discovery now possible in alternate species. We summarize the clinical development of antibodies raised from divergent species, discussing how these animals enable robust immune responses against highly conserved binding sites and yield antibodies capable of penetrating functional pockets via long HCDR3 regions. We also discuss the value of pan-reactive molecules often produced by these hosts, and how these antibodies can be tested in accessible animal models, offering a faster path to clinical development.
Collapse
|
8
|
Gul H, Habib G, Khan IM, Rahman SU, Khan NM, Wang H, Khan NU, Liu Y. Genetic resilience in chickens against bacterial, viral and protozoal pathogens. Front Vet Sci 2022; 9:1032983. [PMID: 36439341 PMCID: PMC9691405 DOI: 10.3389/fvets.2022.1032983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/28/2022] [Indexed: 06/13/2024] Open
Abstract
The genome contributes to the uniqueness of an individual breed, and enables distinctive characteristics to be passed from one generation to the next. The allelic heterogeneity of a certain breed results in a different response to a pathogen with different genomic expression. Disease resistance in chicken is a polygenic trait that involves different genes that confer resistance against pathogens. Such resistance also involves major histocompatibility (MHC) molecules, immunoglobulins, cytokines, interleukins, T and B cells, and CD4+ and CD8+ T lymphocytes, which are involved in host protection. The MHC is associated with antigen presentation, antibody production, and cytokine stimulation, which highlight its role in disease resistance. The natural resistance-associated macrophage protein 1 (Nramp-1), interferon (IFN), myxovirus-resistance gene, myeloid differentiation primary response 88 (MyD88), receptor-interacting serine/threonine kinase 2 (RIP2), and heterophile cells are involved in disease resistance and susceptibility of chicken. Studies related to disease resistance genetics, epigenetics, and quantitative trait loci would enable the identification of resistance markers and the development of disease resistance breeds. Microbial infections are responsible for significant outbreaks and have blighted the poultry industry. Breeding disease-resistant chicken strains may be helpful in tackling pathogens and increasing the current understanding on host genetics in the fight against communicable diseases. Advanced technologies, such as the CRISPR/Cas9 system, whole genome sequencing, RNA sequencing, and high-density single nucleotide polymorphism (SNP) genotyping, aid the development of resistant breeds, which would significantly decrease the use of antibiotics and vaccination in poultry. In this review, we aimed to reveal the recent genetic basis of infection and genomic modification that increase resistance against different pathogens in chickens.
Collapse
Affiliation(s)
- Haji Gul
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Gul Habib
- Department of Microbiology, Abbottabad University of Science and Technology, Abbottabad, Pakistan
| | - Ibrar Muhammad Khan
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Sajid Ur Rahman
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Nazir Muhammad Khan
- Department of Zoology, University of Science and Technology, Bannu, Pakistan
| | - Hongcheng Wang
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Najeeb Ullah Khan
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, Pakistan
| | - Yong Liu
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| |
Collapse
|
9
|
Chockalingam K, Kumar A, Song J, Chen Z. Chicken-derived CD20 antibodies with potent B-cell depletion activity. Br J Haematol 2022; 199:560-571. [PMID: 36039695 PMCID: PMC9649889 DOI: 10.1111/bjh.18438] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/26/2022] [Accepted: 08/17/2022] [Indexed: 12/01/2022]
Abstract
We report four novel anti-human CD20 (hCD20) monoclonal antibodies (mAbs) discovered from a phylogenetically distant species-chickens. The chicken-human chimaeric antibodies exhibit at least 10-fold enhanced antibody-dependent cellular cytotoxicity (ADCC) and 4-8-fold stronger complement-dependent cytotoxicity (CDC) relative to the clinically used mouse-human chimaeric anti-hCD20 antibody rituximab (RTX). Thus, to our knowledge these mAbs are the first to significantly outperform RTX in both Fc-mediated mechanisms of action. The antibodies show 20-100-fold superior depletion of B cells in whole blood from healthy humans relative to RTX and retain efficacy in vivo. One of the mAbs, AC1, can bind mouse CD20, indicating specificity for a novel hCD20 epitope inaccessible to current (mouse-derived) anti-hCD20 mAbs. A humanized version of one antibody, hAC11-10, was created by complementarity-determining region (CDR) grafting into a human variable region framework and this molecule retained the ADCC, in vitro human whole-blood B-cell depletion, and in vivo lymphoma cell depletion activities of the parent. These mAbs represent promising monotherapy candidates for improving upon current less-than-ideal clinical outcomes in lymphoid malignancies and provide an arsenal of biologically relevant molecules for the development of next-generation CD20-mediated immunotherapies including bispecific T-cell engagers (BiTE), antibody-drug conjugates (ADC) and chimaeric antigen receptor-engineered T (CAR-T) cells.
Collapse
Affiliation(s)
- Karuppiah Chockalingam
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center
| | - Anil Kumar
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center
| | - Zhilei Chen
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center
| |
Collapse
|
10
|
IgY Antibodies as Biotherapeutics in Biomedicine. Antibodies (Basel) 2022; 11:antib11040062. [PMID: 36278615 PMCID: PMC9590010 DOI: 10.3390/antib11040062] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/05/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Since the discovery of antibodies by Emil Von Behring and Shibasaburo Kitasato during the 19th century, their potential for use as biotechnological reagents has been exploited in different fields, such as basic and applied research, diagnosis, and the treatment of multiple diseases. Antibodies are relatively easy to obtain from any species with an adaptive immune system, but birds are animals characterized by relatively easy care and maintenance. In addition, the antibodies they produce can be purified from the egg yolk, allowing a system for obtaining them without performing invasive practices, which favors the three “rs” of animal care in experimentation, i.e., replacing, reducing, and refining. In this work, we carry out a brief descriptive review of the most outstanding characteristics of so-called “IgY technology” and the use of IgY antibodies from birds for basic experimentation, diagnosis, and treatment of human beings and animals.
Collapse
|
11
|
PD-1/PD-L1 Checkpoint Inhibitors Are Active in the Chicken Embryo Model and Show Antitumor Efficacy In Ovo. Cancers (Basel) 2022; 14:cancers14133095. [PMID: 35804865 PMCID: PMC9264844 DOI: 10.3390/cancers14133095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 12/28/2022] Open
Abstract
Simple Summary Cancer immunotherapy, also known as immuno-oncology (IO), has made impressive progress in recent decades and is becoming an essential approach for cancer treatments. For IO drug development, a pertinent preclinical model is indispensable for the rapid and efficient transition from preclinical evaluation through to clinical progress. To date, rodents represent the most-often used models for preclinical evaluation. However, their use presents several drawbacks, including ethical constraints, and time-consuming and costly experiments, which could slow down IO drug development. The aim of our study was to assess the use of the chicken embryo (in ovo) model as an alternative in vivo model for evaluating IO drugs. We confirmed in ovo the anti-tumor efficacy of programmed cell death protein-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) checkpoint inhibitors based on the Chicken Chorioallantoic Membrane (CAM) assay, revealing the pertinence of the chicken embryo model in its use for IO research. Abstract (1) Purpose: To assess the use of the chicken embryo (in ovo) model as an alternative in vivo model for immuno-oncology (IO) drug development, focusing on programmed cell death protein-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) immune checkpoint inhibitors. (2) Methods: First, the presence of immune cells in the model was detected through the immunophenotyping of chicken peripheral blood mononuclear cells (PBMCs) based on fluorescence activated cell sorting (FACS) analysis and the immunohistochemistry (IHC) analysis of in ovo tumor-infiltrating lymphocytes. Second, the cross-reactivity between one anti-human PD-1 Ab, pembrolizumab (KEYTRUDA®), and chicken PD-1 was verified through the labelling of chicken splenocytes with pembrolizumab by FACS analysis. Third, the blockade effect of pembrolizumab on chicken PBMCs was assessed in vitro through cytotoxicity assay based on MTT. Fourth, the CAM assay was used to estimate the anti-tumor performance of pembrolizumab through the analyses of tumor growth and chicken immune cell infiltration in tumors. Finally, the efficacy of several PD-1 or PD-L1 inhibitors (nivolumab, atezolizumab and avelumab) on tumor growth was further assessed using the CAM assay. (3) Results: The presence of CD3+, CD4+, CD8+ T lymphocytes and monocytes was confirmed by FACS and IHC analyses. During in vitro assays, pembrolizumab cross-reacted with chicken lymphocytes and induced PD-1/PD-L1 blockade, which permitted the restoration of chicken T-cell’s cytotoxicity against human lung cancer H460 tumor cells. All these in vitro results were correlated with in ovo findings based on the CAM assay: pembrolizumab inhibited H460 tumor growth and induced evident chicken immune cell infiltration (with significant chicken CD45, CD3, CD4, CD8 and CD56 markers) in tumors. Furthermore, the potency of the CAM assay was not limited to the application of pembrolizumab. Nivolumab, atezolizumab and avelumab also led to tumor growth inhibition in ovo, on different tumor models. (4) Conclusions: The chicken embryo affords a physiological, immune reactive, in vivo environment for IO research, which allows observation of how the immune system defense against tumor cells, as well as the different immune tolerance mechanisms leading to tumor immune escape. The encouraging results obtained with PD-1/PD-L1 inhibitors in this study reveal the potential use of the chicken embryo model as an alternative, fast, and reliable in vivo model in the different fields of IO drug discovery.
Collapse
|
12
|
Harwardt J, Bogen JP, Carrara SC, Ulitzka M, Grzeschik J, Hock B, Kolmar H. A Generic Strategy to Generate Bifunctional Two-in-One Antibodies by Chicken Immunization. Front Immunol 2022; 13:888838. [PMID: 35479092 PMCID: PMC9036444 DOI: 10.3389/fimmu.2022.888838] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/18/2022] [Indexed: 12/21/2022] Open
Abstract
Various formats of bispecific antibodies exist, among them Two-in-One antibodies in which each Fab arm can bind to two different antigens. Their IgG-like architecture accounts for low immunogenicity and also circumvents laborious engineering and purification steps to facilitate correct chain pairing. Here we report for the first time the identification of a Two‐in‐One antibody by yeast surface display (YSD) screening of chicken-derived immune libraries. The resulting antibody simultaneously targets the epidermal growth factor receptor (EGFR) and programmed death‐ligand 1 (PD-L1) at the same Fv fragment with two non-overlapping paratopes. The dual action Fab is capable of inhibiting EGFR signaling by binding to dimerization domain II as well as blocking the PD-1/PD-L1 interaction. Furthermore, the Two-in-One antibody demonstrates specific cellular binding properties on EGFR/PD-L1 double positive tumor cells. The presented strategy relies solely on screening of combinational immune-libraries and obviates the need for any additional CDR engineering as described in previous reports. Therefore, this study paves the way for further development of therapeutic antibodies derived from avian immunization with novel and tailor-made binding properties.
Collapse
Affiliation(s)
- Julia Harwardt
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Jan P. Bogen
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Stefania C. Carrara
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Michael Ulitzka
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Julius Grzeschik
- Ferring Biologics Innovation Centre, Biologics Technology and Development, Epalinges, Switzerland
| | - Björn Hock
- Ferring Biologics Innovation Centre, Biologics Technology and Development, Epalinges, Switzerland
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Centre for Synthtic Biology, Technical University of Darmstadt, Darmstadt, Germany
- *Correspondence: Harald Kolmar,
| |
Collapse
|
13
|
Use of Genome Editing Techniques to Produce Transgenic Farm Animals. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1354:279-297. [PMID: 34807447 PMCID: PMC9810480 DOI: 10.1007/978-3-030-85686-1_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recombinant proteins are essential for the treatment and diagnosis of clinical human ailments. The availability and biological activity of recombinant proteins is heavily influenced by production platforms. Conventional production platforms such as yeast, bacteria, and mammalian cells have biological and economical challenges. Transgenic livestock species have been explored as an alternative production platform for recombinant proteins, predominantly through milk secretion; the strategy has been demonstrated to produce large quantities of biologically active proteins. The major limitation of utilizing livestock species as bioreactors has been efforts required to alter the genome of livestock. Advancements in the genome editing field have drastically improved the ability to genetically engineer livestock species. Specifically, genome editing tools such as the CRISPR/Cas9 system have lowered efforts required to generate genetically engineered livestock, thus minimizing restrictions on the type of genetic modification in livestock. In this review, we discuss characteristics of transgenic animal bioreactors and how the use of genome editing systems enhances design and availability of the animal models.
Collapse
|
14
|
Zhang X, Chelliappan B, S R, Antonysamy M. Recent Advances in Applications of Bioactive Egg Compounds in Nonfood Sectors. Front Bioeng Biotechnol 2021; 9:738993. [PMID: 34976961 PMCID: PMC8716877 DOI: 10.3389/fbioe.2021.738993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
Egg, a highly nutritious food, contains high-quality proteins, vitamins, and minerals. This food has been reported for its potential pharmacological properties, including antibacterial, anti-cancer, anti-inflammatory, angiotensin-converting enzyme (ACE) inhibition, immunomodulatory effects, and use in tissue engineering applications. The significance of eggs and their components in disease prevention and treatment is worth more attention. Eggs not only have been known as a "functional food" to combat diseases and facilitate the promotion of optimal health, but also have numerous industrial applications. The current review focuses on different perceptions and non-food applications of eggs, including cosmetics. The versatility of eggs from an industrial perspective makes them a potential candidate for further exploration of several novel components.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Chinese-German Joint Laboratory for Natural Product Research, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
- Centre of Molecular and Environmental Biology, University of Minho, Department of Biology, Braga, Portugal
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Brindha Chelliappan
- Chinese-German Joint Laboratory for Natural Product Research, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, China
- Department of Microbiology, PSG College of Arts & Science, Bharathiar University, Coimbatore, India
| | - Rajeswari S
- Department of Microbiology, PSG College of Arts & Science, Bharathiar University, Coimbatore, India
| | - Michael Antonysamy
- Department of Microbiology, PSG College of Arts & Science, Bharathiar University, Coimbatore, India
| |
Collapse
|
15
|
Zlatina K, Galuska SP. Immunoglobulin Glycosylation - An Unexploited Potential for Immunomodulatory Strategies in Farm Animals. Front Immunol 2021; 12:753294. [PMID: 34733284 PMCID: PMC8558360 DOI: 10.3389/fimmu.2021.753294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/23/2021] [Indexed: 01/01/2023] Open
Abstract
The function of antibodies, namely the identification and neutralization of pathogens, is mediated by their antigen binding site (Fab). In contrast, the subsequent signal transduction for activation of the immune system is mediated by the fragment crystallizable (Fc) region, which interacts with receptors or other components of the immune system, such as the complement system. This aspect of binding and interaction is more precise, readjusted by covalently attached glycan structures close to the hinge region of immunoglobulins (Ig). This fine-tuning of Ig and its actual state of knowledge is the topic of this review. It describes the function of glycosylation at Ig in general and the associated changes due to corresponding glycan structures. We discuss the functionality of IgG glycosylation during different physiological statuses, like aging, lactation and pathophysiological processes. Further, we point out what is known to date about Ig glycosylation in farm animals and how new achievements in vaccination may contribute to improved animal welfare.
Collapse
Affiliation(s)
- Kristina Zlatina
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Sebastian P Galuska
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| |
Collapse
|
16
|
Karachaliou CE, Vassilakopoulou V, Livaniou E. IgY technology: Methods for developing and evaluating avian immunoglobulins for the in vitro detection of biomolecules. World J Methodol 2021; 11:243-262. [PMID: 34631482 PMCID: PMC8472547 DOI: 10.5662/wjm.v11.i5.243] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/10/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
The term “IgY technology” was introduced in the literature in the mid 1990s to describe a procedure involving immunization of avian species, mainly laying hens and consequent isolation of the polyclonal IgYs from the “immune” egg yolk (thus avoiding bleeding and animal stress). IgYs have been applied to various fields of medicine and biotechnology. The present article will deal with specific aspects of IgY technology, focusing on the currently reported methods for developing, isolating, evaluating and storing polyclonal IgYs. Other topics such as current information on isolation protocols or evaluation of IgYs from different avian species are also discussed. Specific advantages of IgY technology (e.g., novel antibody specificities that may emerge via the avian immune system) will also be discussed. Recent in vitro applications of polyclonal egg yolk-derived IgYs to the field of disease diagnosis in human and veterinary medicine through in vitro immunodetection of target biomolecules will be presented. Moreover, ethical aspects associated with animal well-being as well as new promising approaches that are relevant to the original IgY technology (e.g., development of monoclonal IgYs and IgY-like antibodies through the phage display technique or in transgenic chickens) and future prospects in the area will also be mentioned.
Collapse
Affiliation(s)
- Chrysoula-Evangelia Karachaliou
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research “Demokritos”, Athens 15310, Greece
| | - Vyronia Vassilakopoulou
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research “Demokritos”, Athens 15310, Greece
| | - Evangelia Livaniou
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research “Demokritos”, Athens 15310, Greece
| |
Collapse
|
17
|
Elter A, Bogen JP, Habermann J, Kolmar H. Vom Huhn abgeleitete Antikörper für Diagnostik und Immuntherapie. BIOSPEKTRUM 2021; 27:500-504. [PMID: 34511735 PMCID: PMC8417631 DOI: 10.1007/s12268-021-1623-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
AbstractDue to the large evolutionary distance between birds (Aves) und humans, immunization of chickens with human proteins results in a strong response of the bird’s adaptive immune system to proteins of mammalian origin. Additionally, chicken-derived antibodies display less undesired cross-reactivity in analytical setups than conventional rodent-derived antibodies. Due to these features as well as the facile amplification of antibody-coding genes, chicken-derived antibodies emerged as promising molecules for the immunotherapy and various biotechnological applications.
Collapse
|
18
|
Valldorf B, Hinz SC, Russo G, Pekar L, Mohr L, Klemm J, Doerner A, Krah S, Hust M, Zielonka S. Antibody display technologies: selecting the cream of the crop. Biol Chem 2021; 403:455-477. [PMID: 33759431 DOI: 10.1515/hsz-2020-0377] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
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.
Collapse
Affiliation(s)
- Bernhard Valldorf
- Chemical and Pharmaceutical Development, Merck KGaA, Frankfurter Strasse 250, D-64293Darmstadt, Germany
| | - Steffen C Hinz
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287Darmstadt, Germany
| | - Giulio Russo
- Abcalis GmbH, Inhoffenstrasse 7, D-38124Braunschweig, Germany.,Institut für Biochemie, Biotechnologie und Bioinformatik, Technische Universität Braunschweig, Spielmannstrasse 7, D-38106Braunschweig, Germany
| | - Lukas Pekar
- Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, D-64293Darmstadt, Germany
| | - Laura Mohr
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, University of Frankfurt, Max-von-Laue-Strasse 13, D-60438Frankfurt am Main, Germany
| | - Janina Klemm
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287Darmstadt, Germany
| | - Achim Doerner
- Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, D-64293Darmstadt, Germany
| | - Simon Krah
- Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, D-64293Darmstadt, Germany
| | - Michael Hust
- Institut für Biochemie, Biotechnologie und Bioinformatik, Technische Universität Braunschweig, Spielmannstrasse 7, D-38106Braunschweig, Germany
| | - Stefan Zielonka
- Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, D-64293Darmstadt, Germany
| |
Collapse
|
19
|
Ching KH, Berg K, Reynolds K, Pedersen D, Macias A, Abdiche YN, Harriman WD, Leighton PA. Common light chain chickens produce human antibodies of high affinity and broad epitope coverage for the engineering of bispecifics. MAbs 2021; 13:1862451. [PMID: 33491549 PMCID: PMC7849766 DOI: 10.1080/19420862.2020.1862451] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Bispecific antibodies are an important and growing segment in antibody therapeutics, particularly in the immuno-oncology space. Manufacturing of a bispecific antibody with two different heavy chains is greatly simplified if the light chains can be the same for both arms of the antibody. Here, we introduce a strain of common light chain chickens, called OmniClic®, that produces antibody repertoires largely devoid of light chain diversity. The antibody repertoire in these chickens is composed of diverse human heavy chain variable regions capable of high-affinity antigen-specific binding and broad epitope diversity when paired with the germline human kappa light chain. OmniClic birds can be used in immunization campaigns for discovery of human heavy chains to different targets. Subsequent pairing of the heavy chain with a germline human kappa light chain serves to facilitate bispecific antibody production by increasing the efficiency of correct pairing. Abbreviations: AID: activation-induced cytidine deaminase; bsAb: bispecific antibody; CDR: complementarity-determining region; CL: light chain constant region; CmLC: common light chain; D: diversity region; ELISA: enzyme-linked immunosorbent assay; FACS: fluorescence-activated cell sorting; Fc: fragment crystallizable; FcRn: neonatal Fc receptor; FR: framework region; GEM: gel-encapsulated microenvironment; Ig: immunoglobulin; IMGT: the international ImMunoGeneTics information system®; J: joining region; KO: knockout; mAb: monoclonal antibody; NGS: next-generation sequencing; PBS: phosphate-buffered saline; PCR: polymerase chain reaction; PGC: primordial germ cell; PGRN: progranulin; TCR: T cell receptor; V: variable region; VK: kappa light chain variable region; VL: light chain variable region; VH: heavy chain variable region
Collapse
Affiliation(s)
- Kathryn H Ching
- Department of Research and Development, Ligand Pharmaceuticals, Inc ., Emeryville, CA, USA
| | - Kimberley Berg
- Department of Research and Development, Ligand Pharmaceuticals, Inc ., Emeryville, CA, USA.,Department of Molecular and Cellular Biology, Harvard University , Cambridge, MA, USA
| | - Kevin Reynolds
- Department of Research and Development, Ligand Pharmaceuticals, Inc ., Emeryville, CA, USA
| | - Darlene Pedersen
- Department of Research and Development, Ligand Pharmaceuticals, Inc ., Emeryville, CA, USA
| | - Alba Macias
- Department of Structural Biology, Vernalis , Cambridge, UK
| | - Yasmina N Abdiche
- Department of Research and Development, Carterra, Inc. Salt LakeCity, USA(Currently at ImmunoPrecise Antibodies , Fargo, UT, USA
| | - William D Harriman
- Department of Research and Development, Ligand Pharmaceuticals, Inc ., Emeryville, CA, USA
| | - Philip A Leighton
- Department of Research and Development, Ligand Pharmaceuticals, Inc ., Emeryville, CA, USA
| |
Collapse
|
20
|
Transgenic Animals for the Generation of Human Antibodies. LEARNING MATERIALS IN BIOSCIENCES 2021. [DOI: 10.1007/978-3-030-54630-4_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
21
|
Bogen JP, Carrara SC, Fiebig D, Grzeschik J, Hock B, Kolmar H. Expeditious Generation of Biparatopic Common Light Chain Antibodies via Chicken Immunization and Yeast Display Screening. Front Immunol 2020; 11:606878. [PMID: 33424853 PMCID: PMC7786285 DOI: 10.3389/fimmu.2020.606878] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022] Open
Abstract
Bispecific (BsAb) and biparatopic (BpAb) antibodies emerged as promising formats for therapeutic biologics exhibiting tailor-made functional properties. Over recent years, chicken-derived antibodies have gained traction for diagnostic and therapeutic applications due to their broad epitope coverage and convenience of library generation. Here we report the first generation of a biparatopic common light chain (cLC) chicken-derived antibody by an epitope binning-based screening approach using yeast surface display. The resulting monospecific antibodies target conformational epitopes on domain II or III of the epidermal growth factor receptor (EGFR) with lower double- or single-digit nanomolar affinities, respectively. Furthermore, the domain III targeting variant was shown to interfere with epidermal growth factor (EGF) binding. Utilizing the Knob-into-Hole technology (KiH), a biparatopic antibody with subnanomolar affinity was generated that facilitates clustering of soluble and cell-bound EGFR and displayed enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) compared to the parental antibodies. This strategy for generating cLC-based biparatopic antibodies from immunized chickens may pave the way for their further development in therapeutic settings.
Collapse
Affiliation(s)
- Jan P Bogen
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany.,Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Stefania C Carrara
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany.,Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - David Fiebig
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany.,Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Julius Grzeschik
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Björn Hock
- Ferring International Center S.A., Saint-Prex, Switzerland
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| |
Collapse
|
22
|
Ros F, Offner S, Klostermann S, Thorey I, Niersbach H, Breuer S, Zarnt G, Lorenz S, Puels J, Siewe B, Schueler N, Dragicevic T, Ostler D, Hansen-Wester I, Lifke V, Kaluza B, Kaluza K, van Schooten W, Buelow R, Tissot AC, Platzer J. Rabbits transgenic for human IgG genes recapitulating rabbit B-cell biology to generate human antibodies of high specificity and affinity. MAbs 2020; 12:1846900. [PMID: 33228444 PMCID: PMC7780963 DOI: 10.1080/19420862.2020.1846900] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Transgenic animals incorporating human antibody genes are extremely attractive for drug development because they obviate subsequent antibody humanization procedures required for therapeutic translation. Transgenic platforms have previously been established using mice, but also more recently rats, chickens, and cows and are now in abundant use for drug development. However, rabbit-based antibody generation, with a strong track record for specificity and affinity, is able to include gene conversion mediated sequence diversification, thereby enhancing binder maturation and improving the variance/selection of output antibodies in a different way than in rodents. Since it additionally frequently permits good binder generation against antigens that are only weakly immunogenic in other organisms, it is a highly interesting species for therapeutic antibody generation. We report here on the generation, utilization, and analysis of the first transgenic rabbit strain for human antibody production. Through the knockout of endogenous IgM genes and the introduction of human immunoglobulin sequences, this rabbit strain has been engineered to generate a highly diverse human IgG antibody repertoire. We further incorporated human CD79a/b and Bcl2 (B-cell lymphoma 2) genes, which enhance B-cell receptor expression and B-cell survival. Following immunization against the angiogenic factor BMP9 (Bone Morphogenetic Proteins 9), we were able to isolate a set of exquisitely affine and specific neutralizing antibodies from these rabbits. Sequence analysis of these binders revealed that both somatic hypermutation and gene conversion are fully operational in this strain, without compromising the very high degree of humanness. This powerful new transgenic strategy will allow further expansion of the use of endogenous immune mechanisms in drug development.
Collapse
Affiliation(s)
- Francesca Ros
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich , Penzberg, Germany
| | - Sonja Offner
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich , Penzberg, Germany
| | - Stefan Klostermann
- Roche Pharmaceutical Research and Early Development, Informatics, Roche Innovation Center Munich , Penzberg, Germany
| | - Irmgard Thorey
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich , Penzberg, Germany
| | - Helmut Niersbach
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich , Penzberg, Germany
| | - Sebastian Breuer
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich , Penzberg, Germany
| | - Grit Zarnt
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich , Penzberg, Germany
| | - Stefan Lorenz
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich , Penzberg, Germany
| | | | - Basile Siewe
- THE JACKSON LABORATORY JMCRS, Sacramento, CA, USA
| | - Nicole Schueler
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich , Penzberg, Germany
| | - Tajana Dragicevic
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich , Penzberg, Germany
| | - Dominique Ostler
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich , Penzberg, Germany
| | - Imke Hansen-Wester
- Supplier Quality Management, Global External Quality Roche Diagnostics GmbH , Penzberg, Germany
| | - Valeria Lifke
- Personalized Healthcare Solution, Immunoassay Development and System Integration, Roche Diagnostics GmbH , Penzberg, Germany
| | - Brigitte Kaluza
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich , Penzberg, Germany
| | - Klaus Kaluza
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich , Penzberg, Germany
| | | | | | - Alain C Tissot
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich , Penzberg, Germany
| | - Josef Platzer
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich , Penzberg, Germany
| |
Collapse
|
23
|
Elter A, Bogen JP, Hinz SC, Fiebig D, Macarrón Palacios A, Grzeschik J, Hock B, Kolmar H. Humanization of Chicken-Derived scFv Using Yeast Surface Display and NGS Data Mining. Biotechnol J 2020; 16:e2000231. [PMID: 33078896 DOI: 10.1002/biot.202000231] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/06/2020] [Indexed: 01/17/2023]
Abstract
Generation of high-affinity monoclonal antibodies by immunization of chickens is a valuable strategy, particularly for obtaining antibodies directed against epitopes that are conserved in mammals. A generic procedure is established for the humanization of chicken-derived antibodies. To this end, high-affinity binders of the epidermal growth factor receptor extracellular domain are isolated from immunized chickens using yeast surface display. Complementarity determining regions (CDRs) of two high-affinity binders are grafted onto a human acceptor framework. Simultaneously, Vernier zone residues, responsible for spatial CDR arrangement, are partially randomized. A yeast surface display library comprising ≈300 000 variants is screened for high-affinity binders in the scFv and Fab formats. Next-generation sequencing discloses humanized antibody variants with restored affinity and improved protein characteristics compared to the parental chicken antibodies. Furthermore, the sequencing data give new insights into the importance of antibody format, used during the humanization process. Starting from the antibody repertoire of immunized chickens, this work features an effective and fast high-throughput approach for the generation of multiple humanized antibodies with potential therapeutic relevance.
Collapse
Affiliation(s)
- Adrian Elter
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Strasse 4, Darmstadt, D-64287, Germany.,Merck Lab @ Technical University of Darmstadt, Alarich-Weiss-Strasse 4, Darmstadt, D-64287, Germany
| | - Jan P Bogen
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Strasse 4, Darmstadt, D-64287, Germany.,Ferring Darmstadt Laboratory, Biologics Technology and Development, Alarich-Weiss-Strasse 4, Darmstadt, D-64287, Germany
| | - Steffen C Hinz
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Strasse 4, Darmstadt, D-64287, Germany.,Merck Lab @ Technical University of Darmstadt, Alarich-Weiss-Strasse 4, Darmstadt, D-64287, Germany
| | - David Fiebig
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Strasse 4, Darmstadt, D-64287, Germany.,Ferring Darmstadt Laboratory, Biologics Technology and Development, Alarich-Weiss-Strasse 4, Darmstadt, D-64287, Germany
| | - Arturo Macarrón Palacios
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Strasse 4, Darmstadt, D-64287, Germany
| | - Julius Grzeschik
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Alarich-Weiss-Strasse 4, Darmstadt, D-64287, Germany
| | - Björn Hock
- Ferring International Center S.A., Chemin de la Vergognausaz 50, Saint-Prex, 1162, Switzerland
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Strasse 4, Darmstadt, D-64287, Germany.,Merck Lab @ Technical University of Darmstadt, Alarich-Weiss-Strasse 4, Darmstadt, D-64287, Germany
| |
Collapse
|
24
|
Cameron B, Dabdoubi T, Berthou-Soulié L, Gagnaire M, Arnould I, Severac A, Soubrier F, Morales J, Leighton PA, Harriman W, Ching K, Abdiche Y, Radošević K, Bouquin T. Complementary epitopes and favorable developability of monoclonal anti-LAMP1 antibodies generated using two transgenic animal platforms. PLoS One 2020; 15:e0235815. [PMID: 32673351 PMCID: PMC7365404 DOI: 10.1371/journal.pone.0235815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 06/23/2020] [Indexed: 01/08/2023] Open
Abstract
Monoclonal antibodies (mAbs) for therapeutic applications should be as similar to native human antibodies as possible to minimize their immunogenicity in patients. Several transgenic animal platforms are available for the generation of fully human mAbs. Attributes such as specificity, efficacy and Chemistry, Manufacturing and Controls (CMC) developability of antibodies against a specific target are typically established for antibodies obtained from one platform only. In this study, monoclonal antibodies (mAbs) cross-reactive against human and cynomolgus LAMP1 were derived from the human immunoglobulin transgenic TRIANNI mouse and OmniChicken® platforms and assessed for their specificity, sequence diversity, ability to bind to and internalize into tumor cells, expected immunogenicity and CMC developability. Our results show that the two platforms were complementary at providing a large diversity of mAbs with respect to epitope coverage and antibody sequence diversity. Furthermore, most antibodies originating from either platform exhibited good manufacturability characteristics.
Collapse
Affiliation(s)
| | | | | | | | | | - Anne Severac
- Biologics Research, Sanofi R&D, Boufféré, France
| | | | - Jacqueline Morales
- Ligand Pharmaceuticals Inc., San Diego, California, United States of America
| | - Philip A. Leighton
- Ligand Pharmaceuticals Inc., San Diego, California, United States of America
| | - William Harriman
- Ligand Pharmaceuticals Inc., San Diego, California, United States of America
| | - Kathryn Ching
- Ligand Pharmaceuticals Inc., San Diego, California, United States of America
| | - Yasmina Abdiche
- Carterra Inc., Salt Lake City, Utah, United States of America
| | | | | |
Collapse
|
25
|
Parray HA, Shukla S, Samal S, Shrivastava T, Ahmed S, Sharma C, Kumar R. Hybridoma technology a versatile method for isolation of monoclonal antibodies, its applicability across species, limitations, advancement and future perspectives. Int Immunopharmacol 2020; 85:106639. [PMID: 32473573 PMCID: PMC7255167 DOI: 10.1016/j.intimp.2020.106639] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/06/2020] [Accepted: 05/22/2020] [Indexed: 02/06/2023]
Abstract
The advancements in technology and manufacturing processes have allowed the development of new derivatives, biosimilar or advanced improved versions for approved antibodies each year for treatment regimen. There are more than 700 antibody-based molecules that are in different stages of phase I/II/ III clinical trials targeting new unique targets. To date, approximately more than 80 monoclonal antibodies (mAbs) have been approved. A total of 7 novel antibody therapeutics had been granted the first approval either in the United States or European Union in the year 2019, representing approximately 20% of the total number of approved drugs. Most of these licenced mAbs or their derivatives are either of hybridoma origin or their improvised engineered versions. Even with the recent development of high throughput mAb generation technologies, hybridoma is the most favoured method due to its indigenous nature to preserve natural cognate antibody pairing information and preserves innate functions of immune cells. The recent advent of antibody engineering technology has superseded the species level barriers and has shown success in isolation of hybridoma across phylogenetically distinct species. This has led to the isolation of monoclonal antibodies against human targets that are conserved and non-immunogenic in the rodent. In this review, we have discussed in detail about hybridoma technology, its expansion towards different animal species, the importance of antibodies isolated from different animal sources that are useful in biological applications, advantages, and limitations. This review also summarizes the challenges and recent progress associated with hybridoma development, and how it has been overcome in these years to provide new insights for the isolation of mAbs.
Collapse
Affiliation(s)
- Hilal Ahmed Parray
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Shivangi Shukla
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Sweety Samal
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Tripti Shrivastava
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Shubbir Ahmed
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Chandresh Sharma
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana 121001, India.
| | - Rajesh Kumar
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana 121001, India.
| |
Collapse
|
26
|
Streamlined human antibody generation and optimization by exploiting designed immunoglobulin loci in a B cell line. Cell Mol Immunol 2020; 18:1545-1561. [PMID: 32457406 PMCID: PMC8166883 DOI: 10.1038/s41423-020-0440-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 01/30/2023] Open
Abstract
Monoclonal antibodies (mAbs) are widely utilized as therapeutic drugs for various diseases, such as cancer, autoimmune diseases, and infectious diseases. Using the avian-derived B cell line DT40, we previously developed an antibody display technology, namely, the ADLib system, which rapidly generates antigen-specific mAbs. Here, we report the development of a human version of the ADLib system and showcase the streamlined generation and optimization of functional human mAbs. Tailored libraries were first constructed by replacing endogenous immunoglobulin genes with designed human counterparts. From these libraries, clones producing full-length human IgGs against distinct antigens can be isolated, as exemplified by the selection of antagonistic mAbs. Taking advantage of avian biology, effective affinity maturation was achieved in a straightforward manner by seamless diversification of the parental clones into secondary libraries followed by single-cell sorting, quickly affording mAbs with improved affinities and functionalities. Collectively, we demonstrate that the human ADLib system could serve as an integrative platform with unique diversity for rapid de novo generation and optimization of therapeutic or diagnostic antibody leads. Furthermore, our results suggest that libraries can be constructed by introducing exogenous genes into DT40 cells, indicating that the ADLib system has the potential to be applied for the rapid and effective directed evolution and optimization of proteins in various fields beyond biomedicine.
Collapse
|
27
|
Chockalingam K, Peng Z, Vuong CN, Berghman LR, Chen Z. Golden Gate assembly with a bi-directional promoter (GBid): A simple, scalable method for phage display Fab library creation. Sci Rep 2020; 10:2888. [PMID: 32076016 PMCID: PMC7031318 DOI: 10.1038/s41598-020-59745-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 02/03/2020] [Indexed: 11/09/2022] Open
Abstract
Fabs offer an attractive platform for monoclonal antibody discovery/engineering, but library construction can be cumbersome. We report a simple method – Golden Gate assembly with a bi-directional promoter (GBid) – for constructing phage display Fab libraries. In GBid, the constant domains of the Fabs are located in the backbone of the phagemid vector and the library insert comprises only the variable regions of the antibodies and a central bi-directional promoter. This vector design reduces the process of Fab library construction to “scFv-like” simplicity and the double promoter ensures robust expression of both constituent chains. To maximize the library size, the 3 fragments comprising the insert – two variable chains and one bi-directional promoter – are assembled via a 3-fragment overlap extension PCR and the insert is incorporated into the vector via a high-efficiency one-fragment, one-pot Golden Gate assembly. The reaction setup requires minimal preparatory work and enzyme quantities, making GBid highly scalable. Using GBid, we constructed a chimeric chicken-human Fab phage display library comprising 1010 variants targeting the multi-transmembrane protein human CD20 (hCD20). Selection/counter-selection on transfected whole cells yielded hCD20-specific antibodies in four rounds of panning. The simplicity and scalability of GBid makes it a powerful tool for the discovery/engineering of Fabs and IgGs.
Collapse
Affiliation(s)
- Karuppiah Chockalingam
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, Texas, 77843, USA
| | - Zeyu Peng
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, Texas, 77843, USA.,Biosion, Inc., Nanjing, 210061, China
| | - Christine N Vuong
- Department of Poultry Science, Texas A&M University, College Station, Texas, 77843, USA.,Department of Poultry Science, University of Arkansas, Fayetteville, Arkansas, 72703, USA
| | - Luc R Berghman
- Department of Poultry Science, Texas A&M University, College Station, Texas, 77843, USA
| | - Zhilei Chen
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, Texas, 77843, USA.
| |
Collapse
|
28
|
Production of TRPV2-targeting functional antibody ameliorating dilated cardiomyopathy and muscular dystrophy in animal models. J Transl Med 2020; 100:324-337. [PMID: 31896817 DOI: 10.1038/s41374-019-0363-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 12/28/2022] Open
Abstract
Abnormal Ca2+ handling is essential in the pathophysiology of degenerative muscle disorders, such as dilated cardiomyopathy (DCM) and muscular dystrophy (MD). Transient receptor potential cation channel, subfamily V, member 2 (TRPV2) is a candidate for Ca2+ entry and a potential therapeutic target for degenerative muscle disorders, there are few specific inhibitors for TRPV2. In this study, we produced a monoclonal antibody (designated mAb88-2) and two polyclonal antibodies (pAb591 and pAb592) that selectively recognize TRPV2 from the outside of cells and interact with the turret region of the pore-forming outer gate. These antibodies inhibited Ca2+ influx via TRPV2 in cultured cells and substantially reduced TRPV2 in the plasma membrane via cellular internalization. We evaluated the therapeutic efficacy of the functional antibody in δ-sarcoglycan-deficient hamster (J2N-k) models of DCM and MD and in the 4C30 DCM model of murine heart failure. The intraperitoneal administration of the functional antibody (0.5 mg/kg) for 2 weeks (once a week) prevented the progression of cardiac dysfunction, as evaluated by echocardiography and histological staining, and improved the abnormal Ca2+ handling (high diastolic Ca2+ level and small Ca2+ transient peak) in cardiomyocytes isolated from J2N-k hamsters and prevented skeletal muscle damage. Further, the antibody effectively prevented heart failure in the 4C30 mouse model with end-stage DCM. Interestingly, endogenous TRPV2 that accumulated in the cardiac and skeletal muscle sarcolemma disappeared upon antibody administration. Thus, the newly produced antibodies are capable of ameliorating DCM and MD by promoting the cellular internalization of TRPV2; antibodies specific to human TRPV2 may substantially improve the treatment of patients with degenerative muscle diseases.
Collapse
|
29
|
Ching KH, Berg K, Morales J, Pedersen D, Harriman WD, Abdiche YN, Leighton PA. Expression of human lambda expands the repertoire of OmniChickens. PLoS One 2020; 15:e0228164. [PMID: 31995598 PMCID: PMC6988971 DOI: 10.1371/journal.pone.0228164] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/08/2020] [Indexed: 12/20/2022] Open
Abstract
Most of the approved monoclonal antibodies used in the clinic were initially discovered in mice. However, many targets of therapeutic interest are highly conserved proteins that do not elicit a robust immune response in mice. There is a need for non-mammalian antibody discovery platforms which would allow researchers to access epitopes that are not recognized in mammalian hosts. Recently, we introduced the OmniChicken®, a transgenic animal carrying human VH3-23 and VK3-15 at its immunoglobulin loci. Here, we describe a new version of the OmniChicken which carries VH3-23 and either VL1-44 or VL3-19 at its heavy and light chain loci, respectively. The Vλ-expressing birds showed normal B and T populations in the periphery. A panel of monoclonal antibodies demonstrated comparable epitope coverage of a model antigen compared to both wild-type and Vκ-expressing OmniChickens. Kinetic analysis identified binders in the picomolar range. The Vλ-expressing bird increases the antibody diversity available in the OmniChicken platform, further enabling discovery of therapeutic leads.
Collapse
Affiliation(s)
- Kathryn H. Ching
- Ligand Pharmaceuticals Incorporated, Emeryville, California, United States of America
| | - Kimberley Berg
- Ligand Pharmaceuticals Incorporated, Emeryville, California, United States of America
| | - Jacqueline Morales
- Ligand Pharmaceuticals Incorporated, Emeryville, California, United States of America
| | - Darlene Pedersen
- Ligand Pharmaceuticals Incorporated, Emeryville, California, United States of America
| | - William D. Harriman
- Ligand Pharmaceuticals Incorporated, Emeryville, California, United States of America
| | | | - Philip A. Leighton
- Ligand Pharmaceuticals Incorporated, Emeryville, California, United States of America
| |
Collapse
|
30
|
Sellmann C, Pekar L, Bauer C, Ciesielski E, Krah S, Becker S, Toleikis L, Kügler J, Frenzel A, Valldorf B, Hust M, Zielonka S. A One-Step Process for the Construction of Phage Display scFv and VHH Libraries. Mol Biotechnol 2020; 62:228-239. [DOI: 10.1007/s12033-020-00236-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
31
|
Sim J, Sockolosky JT, Sangalang E, Izquierdo S, Pedersen D, Harriman W, Wibowo AS, Carter J, Madan A, Doyle L, Harrabi O, Kauder SE, Chen A, Kuo TC, Wan H, Pons J. Discovery of high affinity, pan-allelic, and pan-mammalian reactive antibodies against the myeloid checkpoint receptor SIRPα. MAbs 2019; 11:1036-1052. [PMID: 31257988 PMCID: PMC6748616 DOI: 10.1080/19420862.2019.1624123] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Targeting the CD47-signal-regulatory protein α (SIRPα) pathway represents a novel therapeutic approach to enhance anti-cancer immunity by promoting both innate and adaptive immune responses. Unlike CD47, which is expressed ubiquitously, SIRPα expression is mainly restricted to myeloid cells and neurons. Therefore, compared to CD47-targeted therapies, targeting SIRPα may result in differential safety and efficacy profiles, potentially enabling lower effective doses and improved pharmacokinetics and pharmacodynamics. The development of effective SIRPα antagonists is restricted by polymorphisms within the CD47-binding domain of SIRPα, necessitating pan-allele reactive anti-SIRPα antibodies for therapeutic intervention in diverse patient populations. We immunized wild-type and human antibody transgenic chickens with a multi-allele and multi-species SIRPα regimen in order to discover pan-allelic and pan-mammalian reactive anti-SIRPα antibodies suitable for clinical translation. A total of 200 antibodies were isolated and screened for SIRPα reactivity from which approximately 70 antibodies with diverse SIRPα binding profiles, sequence families, and epitopes were selected for further characterization. A subset of anti-SIRPα antibodies bound to both human SIRPα v1 and v2 alleles with high affinity ranging from low nanomolar to picomolar, potently antagonized the CD47/SIRPα interaction, and potentiated macrophage-mediated antibody-dependent cellular phagocytosis in vitro. X-ray crystal structures of five anti-SIRPα antigen-binding fragments, each with unique epitopes, in complex with SIRPα (PDB codes 6NMV, 6NMU, 6NMT, 6NMS, and 6NMR) are reported. Furthermore, some of the anti-SIRPα antibodies cross-react with cynomolgus SIRPα and various mouse SIRPα alleles (BALB/c, NOD, BL/6), which can facilitate preclinical to clinical development. These properties provide an attractive rationale to advance the development of these anti-SIRPα antibodies as a novel therapy for advanced malignancies. Abbreviations: ADCC: antibody-dependent cellular cytotoxicity; ADCP: antibody-dependent cellular phagocytosis; CFSE: carboxyfluorescein succinimidyl ester; Fab: fragment antigen binding; Fc: fragment crystallizable; FcγR: Fcγ receptor; Ig: immunoglobulin; IND: investigational new drug; MDM⊘: monocyte-derived macrophage; NOD: non-obese diabetic; scFv: single chain fragment variable; SCID: severe combined immunodeficiency; SIRP: signal-regulatory protein.
Collapse
Affiliation(s)
- Janet Sim
- a ALX Oncology, Departments of Protein Sciences and Translational Biology , Burlingame , CA , USA
| | - Jonathan T Sockolosky
- a ALX Oncology, Departments of Protein Sciences and Translational Biology , Burlingame , CA , USA
| | - Emma Sangalang
- a ALX Oncology, Departments of Protein Sciences and Translational Biology , Burlingame , CA , USA
| | | | | | | | | | | | - Anup Madan
- d Covance Genomics Laboratory , Redmond , WA , USA
| | - Laura Doyle
- a ALX Oncology, Departments of Protein Sciences and Translational Biology , Burlingame , CA , USA
| | - Ons Harrabi
- a ALX Oncology, Departments of Protein Sciences and Translational Biology , Burlingame , CA , USA
| | - Steven E Kauder
- a ALX Oncology, Departments of Protein Sciences and Translational Biology , Burlingame , CA , USA
| | - Amy Chen
- a ALX Oncology, Departments of Protein Sciences and Translational Biology , Burlingame , CA , USA
| | - Tracy C Kuo
- a ALX Oncology, Departments of Protein Sciences and Translational Biology , Burlingame , CA , USA
| | - Hong Wan
- a ALX Oncology, Departments of Protein Sciences and Translational Biology , Burlingame , CA , USA
| | - Jaume Pons
- a ALX Oncology, Departments of Protein Sciences and Translational Biology , Burlingame , CA , USA
| |
Collapse
|
32
|
Gjetting T, Gad M, Fröhlich C, Lindsted T, Melander MC, Bhatia VK, Grandal MM, Dietrich N, Uhlenbrock F, Galler GR, Strandh M, Lantto J, Bouquin T, Horak ID, Kragh M, Pedersen MW, Koefoed K. Sym021, a promising anti-PD1 clinical candidate antibody derived from a new chicken antibody discovery platform. MAbs 2019; 11:666-680. [PMID: 31046547 PMCID: PMC6601539 DOI: 10.1080/19420862.2019.1596514] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Discovery of therapeutic antibodies is a field of intense development, where immunization of rodents remains a major source of antibody candidates. However, high orthologue protein sequence homology between human and rodent species disfavors generation of antibodies against functionally conserved binding epitopes. Chickens are phylogenetically distant from mammals. Since chickens generate antibodies from a restricted set of germline genes, the possibility of adapting the Symplex antibody discovery platform to chicken immunoglobulin genes and combining it with high-throughput humanization of antibody frameworks by “mass complementarity-determining region grafting” was explored. Hence, wild type chickens were immunized with an immune checkpoint inhibitor programmed cell death 1 (PD1) antigen, and a repertoire of 144 antibodies was generated. The PD1 antibody repertoire was successfully humanized, and we found that most humanized antibodies retained affinity largely similar to that of the parental chicken antibodies. The lead antibody Sym021 blocked PD-L1 and PD-L2 ligand binding, resulting in elevated T-cell cytokine production in vitro. Detailed epitope mapping showed that the epitope recognized by Sym021 was unique compared to the clinically approved PD1 antibodies pembrolizumab and nivolumab. Moreover, Sym021 bound human PD1 with a stronger affinity (30 pM) compared to nivolumab and pembrolizumab, while also cross-reacting with cynomolgus and mouse PD1. This enabled direct testing of Sym021 in the syngeneic mouse in vivo cancer models and evaluation of preclinical toxicology in cynomolgus monkeys. Preclinical in vivo evaluation in various murine and human tumor models demonstrated a pronounced anti-tumor effect of Sym021, supporting its current evaluation in a Phase 1 clinical trial. Abbreviations: ADCC, antibody-dependent cellular cytotoxicity; CD, cluster of differentiation; CDC, complement-dependent cytotoxicity; CDR, complementarity determining region; DC, dendritic cell; ELISA, enzyme-linked immunosorbent assay; FACS, fluorescence activated cell sorting; FR, framework region; GM-CSF, granulocyte-macrophage colony-stimulating factor; HRP, horseradish peroxidase; IgG, immunoglobulin G; IL, interleukin; IFN, interferon; mAb, monoclonal antibody; MLR, mixed lymphocyte reaction; NK, natural killer; PBMC, peripheral blood mono-nuclear cell; PD1, programmed cell death 1; PDL1, programmed cell death ligand 1; RT-PCR, reverse transcription polymerase chain reaction; SEB, Staphylococcus Enterotoxin B; SPR, surface Plasmon Resonance; VL, variable part of light chain; VH, variable part of heavy chain
Collapse
Affiliation(s)
- Torben Gjetting
- a Antibody Discovery, Antibody Discovery , Ballerup , Denmark
| | - Monika Gad
- b Cancer Biology and Immunology, Symphogen A/S , Ballerup , Denmark
| | | | - Trine Lindsted
- b Cancer Biology and Immunology, Symphogen A/S , Ballerup , Denmark
| | | | - Vikram K Bhatia
- a Antibody Discovery, Antibody Discovery , Ballerup , Denmark
| | | | | | | | | | - Magnus Strandh
- a Antibody Discovery, Antibody Discovery , Ballerup , Denmark
| | - Johan Lantto
- d Global Research and Development, Symphogen A/S , Ballerup , Denmark
| | - Thomas Bouquin
- a Antibody Discovery, Antibody Discovery , Ballerup , Denmark
| | - Ivan D Horak
- d Global Research and Development, Symphogen A/S , Ballerup , Denmark
| | - Michael Kragh
- c Antibody Pharmacology, Symphogen A/S , Ballerup , Denmark
| | | | - Klaus Koefoed
- a Antibody Discovery, Antibody Discovery , Ballerup , Denmark
| |
Collapse
|
33
|
Roth L, Grzeschik J, Hinz SC, Becker S, Toleikis L, Busch M, Kolmar H, Krah S, Zielonka S. Facile generation of antibody heavy and light chain diversities for yeast surface display by Golden Gate Cloning. Biol Chem 2018; 400:383-393. [DOI: 10.1515/hsz-2018-0347] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/11/2018] [Indexed: 01/03/2023]
Abstract
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.
Collapse
Affiliation(s)
- Lukas Roth
- Institute for Organic Chemistry and Biochemistry , Technische Universität Darmstadt , Alarich-Weiss-Strasse 4 , D-64287 Darmstadt , Germany
| | - Julius Grzeschik
- Institute for Organic Chemistry and Biochemistry , Technische Universität Darmstadt , Alarich-Weiss-Strasse 4 , D-64287 Darmstadt , Germany
| | - Steffen C. Hinz
- Institute for Organic Chemistry and Biochemistry , Technische Universität Darmstadt , Alarich-Weiss-Strasse 4 , D-64287 Darmstadt , Germany
| | - Stefan Becker
- Protein Engineering and Antibody Technologies, Merck KGaA , Frankfurter Strasse 250 , D-64293 Darmstadt , Germany
| | - Lars Toleikis
- Protein Engineering and Antibody Technologies, Merck KGaA , Frankfurter Strasse 250 , D-64293 Darmstadt , Germany
| | - Michael Busch
- Discovery Pharmacology, Merck KGaA , Frankfurter Strasse 250 , D-64293 Darmstadt , Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry , Technische Universität Darmstadt , Alarich-Weiss-Strasse 4 , D-64287 Darmstadt , Germany
| | - Simon Krah
- Protein Engineering and Antibody Technologies, Merck KGaA , Frankfurter Strasse 250 , D-64293 Darmstadt , Germany
| | - Stefan Zielonka
- Protein Engineering and Antibody Technologies, Merck KGaA , Frankfurter Strasse 250 , D-64293 Darmstadt , Germany
| |
Collapse
|
34
|
Sid H, Schusser B. Applications of Gene Editing in Chickens: A New Era Is on the Horizon. Front Genet 2018; 9:456. [PMID: 30356667 PMCID: PMC6189320 DOI: 10.3389/fgene.2018.00456] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/18/2018] [Indexed: 01/15/2023] Open
Abstract
The chicken represents a valuable model for research in the area of immunology, infectious diseases as well as developmental biology. Although it was the first livestock species to have its genome sequenced, there was no reverse genetic technology available to help understanding specific gene functions. Recently, homologous recombination was used to knockout the chicken immunoglobulin genes. Subsequent studies using immunoglobulin knockout birds helped to understand different aspects related to B cell development and antibody production. Furthermore, the latest advances in the field of genome editing including the CRISPR/Cas9 system allowed the introduction of site specific gene modifications in various animal species. Thus, it may provide a powerful tool for the generation of genetically modified chickens carrying resistance for certain pathogens. This was previously demonstrated by targeting the Trp38 region which was shown to be effective in the control of avian leukosis virus in chicken DF-1 cells. Herein we review the current and future prospects of gene editing and how it possibly contributes to the development of resistant chickens against infectious diseases.
Collapse
Affiliation(s)
| | - Benjamin Schusser
- Department of Animal Sciences, Reproductive Biotechnology, School of Life Sciences Weihenstephan, Technical University Munich, Freising, Germany
| |
Collapse
|
35
|
Persson H, Kirik U, Thörnqvist L, Greiff L, Levander F, Ohlin M. In Vitro Evolution of Antibodies Inspired by In Vivo Evolution. Front Immunol 2018; 9:1391. [PMID: 29977238 PMCID: PMC6021498 DOI: 10.3389/fimmu.2018.01391] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 06/05/2018] [Indexed: 01/16/2023] Open
Abstract
In vitro generation of antibodies often requires variable domain sequence evolution to adapt the protein in terms of affinity, specificity, or developability. Such antibodies, including those that are of interest for clinical development, may have their origins in a diversity of immunoglobulin germline genes. Others and we have previously shown that antibodies of different origins tend to evolve along different, preferred trajectories. Apart from substitutions within the complementary determining regions, evolution may also, in a germline gene-origin-defined manner, be focused to residues in the framework regions, and even to residues within the protein core, in many instances at a substantial distance from the antibody’s antigen-binding site. Examples of such germline origin-defined patterns of evolution are described. We propose that germline gene-preferred substitution patterns offer attractive alternatives that should be considered in efforts to evolve antibodies intended for therapeutic use with respect to appropriate affinity, specificity, and product developability. We also hypothesize that such germline gene-origin-defined in vitro evolution hold potential to result in products with limited immunogenicity, as similarly evolved antibodies will be parts of conventional, in vivo-generated antibody responses and thus are likely to have been seen by the immune system in the past.
Collapse
Affiliation(s)
- Helena Persson
- Drug Discovery and Development Platform, Science for Life Laboratory, Stockholm, Sweden.,School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology, Stockholm, Sweden
| | - Ufuk Kirik
- Department of Immunotechnology, Lund University, Lund, Sweden
| | | | - Lennart Greiff
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Otorhinolaryngology, Head & Neck Surgery, Skåne University Hospital, Lund, Sweden
| | | | - Mats Ohlin
- Department of Immunotechnology, Lund University, Lund, Sweden.,Human Antibody Therapeutics, Drug Discovery and Development Platform, Science for Life Laboratory, Lund University, Lund, Sweden
| |
Collapse
|
36
|
Leighton PA, Morales J, Harriman WD, Ching KH. V(D)J Rearrangement Is Dispensable for Producing CDR-H3 Sequence Diversity in a Gene Converting Species. Front Immunol 2018; 9:1317. [PMID: 29951062 PMCID: PMC6008532 DOI: 10.3389/fimmu.2018.01317] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/28/2018] [Indexed: 12/20/2022] Open
Abstract
An important characteristic of chickens is that the antibody repertoire is based on a single framework, with diversity found mainly in the CDRs of the light and heavy chain variable regions. Despite this apparent limitation in the antibody repertoire, high-affinity antibodies can be raised to a wide variety of targets, including those that are highly conserved. Transgenic chickens have previously been generated that express a humanized antibody repertoire, with a single framework that incorporates diversity by the process of gene conversion, as in wild-type chickens. Here, we compare the sequences and antibodies that are generated purely by gene conversion/somatic hypermutation of a pre-rearranged heavy chain, with the diversity obtained by V(D)J rearrangement followed by gene conversion and somatic hypermutation. In a gene converting species, CDR-H3 lengths are more variable with V(D)J rearrangement, but similar levels of amino acid diversity are obtainable with gene conversion/somatic hypermutation alone.
Collapse
|
37
|
Chen WC, Murawsky CM. Strategies for Generating Diverse Antibody Repertoires Using Transgenic Animals Expressing Human Antibodies. Front Immunol 2018; 9:460. [PMID: 29563917 PMCID: PMC5845867 DOI: 10.3389/fimmu.2018.00460] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/21/2018] [Indexed: 01/14/2023] Open
Abstract
Therapeutic molecules derived from antibodies have become a dominant class of drugs used to treat human disease. Increasingly, therapeutic antibodies are discovered using transgenic animal systems that have been engineered to express human antibodies. While the engineering details differ, these platforms share the ability to raise an immune response that is comprised of antibodies with fully human idiotypes. Although the predominant transgenic host species has been mouse, the genomes of rats, rabbits, chickens, and cows have also been modified to express human antibodies. The creation of transgenic animal platforms expressing human antibody repertoires has revolutionized therapeutic antibody drug discovery. The observation that the immune systems of these animals are able to recognize and respond to a wide range of therapeutically relevant human targets has led to a surge in antibody-derived drugs in current development. While the clinical success of fully human monoclonal antibodies derived from transgenic animals is well established, recent trends have seen increasingly stringent functional design goals and a shift in difficulty as the industry attempts to tackle the next generation of disease-associated targets. These challenges have been met with a number of novel approaches focused on the generation of large, high-quality, and diverse antibody repertoires. In this perspective, we describe some of the strategies and considerations we use for manipulating the immune systems of transgenic animal platforms (such as XenoMouse®) with a focus on maximizing the diversity of the primary response and steering the ensuing antibody repertoire toward a desired outcome.
Collapse
Affiliation(s)
- Weihsu C Chen
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, BC, Canada
| | - Christopher M Murawsky
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, BC, Canada
| |
Collapse
|
38
|
Almagro JC, Daniels-Wells TR, Perez-Tapia SM, Penichet ML. Progress and Challenges in the Design and Clinical Development of Antibodies for Cancer Therapy. Front Immunol 2018; 8:1751. [PMID: 29379493 PMCID: PMC5770808 DOI: 10.3389/fimmu.2017.01751] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/24/2017] [Indexed: 11/14/2022] Open
Abstract
The remarkable progress in engineering and clinical development of therapeutic antibodies in the last 40 years, after the seminal work by Köhler and Milstein, has led to the approval by the United States Food and Drug Administration (FDA) of 21 antibodies for cancer immunotherapy. We review here these approved antibodies, with emphasis on the methods used for their discovery, engineering, and optimization for therapeutic settings. These methods include antibody engineering via chimerization and humanization of non-human antibodies, as well as selection and further optimization of fully human antibodies isolated from human antibody phage-displayed libraries and immunization of transgenic mice capable of generating human antibodies. These technology platforms have progressively led to the development of therapeutic antibodies with higher human content and, thus, less immunogenicity. We also discuss the genetic engineering approaches that have allowed isotype switching and Fc modifications to modulate effector functions and bioavailability (half-life), which together with the technologies for engineering the Fv fragment, have been pivotal in generating more efficacious and better tolerated therapeutic antibodies to treat cancer.
Collapse
Affiliation(s)
| | - Tracy R Daniels-Wells
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | | | - Manuel L Penichet
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, United States.,The Molecular Biology Institute, University of California, Los Angeles, CA, United States.,UCLA AIDS Institute, Los Angeles, CA, United States
| |
Collapse
|