1
|
Aboul-Ella H, Gohar A, Ali AA, Ismail LM, Mahmoud AEER, Elkhatib WF, Aboul-Ella H. Monoclonal antibodies: From magic bullet to precision weapon. MOLECULAR BIOMEDICINE 2024; 5:47. [PMID: 39390211 PMCID: PMC11467159 DOI: 10.1186/s43556-024-00210-1] [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: 05/05/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
Monoclonal antibodies (mAbs) are used to prevent, detect, and treat a broad spectrum of non-communicable and communicable diseases. Over the past few years, the market for mAbs has grown exponentially with an expected compound annual growth rate (CAGR) of 11.07% from 2024 (237.64 billion USD estimated at the end of 2023) to 2033 (679.03 billion USD expected by the end of 2033). Ever since the advent of hybridoma technology introduced in 1975, antibody-based therapeutics were realized using murine antibodies which further progressed into humanized and fully human antibodies, reducing the risk of immunogenicity. Some benefits of using mAbs over conventional drugs include a drastic reduction in the chances of adverse reactions, interactions between drugs, and targeting specific proteins. While antibodies are very efficient, their higher production costs impede the process of commercialization. However, their cost factor has been improved by developing biosimilar antibodies as affordable versions of therapeutic antibodies. Along with the recent advancements and innovations in antibody engineering have helped and will furtherly help to design bio-better antibodies with improved efficacy than the conventional ones. These novel mAb-based therapeutics are set to revolutionize existing drug therapies targeting a wide spectrum of diseases, thereby meeting several unmet medical needs. This review provides comprehensive insights into the current fundamental landscape of mAbs development and applications and the key factors influencing the future projections, advancement, and incorporation of such promising immunotherapeutic candidates as a confrontation approach against a wide list of diseases, with a rationalistic mentioning of any limitations facing this field.
Collapse
Affiliation(s)
- Hassan Aboul-Ella
- Department of Microbiology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.
| | - Asmaa Gohar
- Department of Microbiology and Immunology, Faculty of Pharmacy, Galala University, Suez, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ahram Canadian University (ACU), Giza, Egypt
- Egyptian Drug Authority (EDA), Giza, Egypt
| | - Aya Ahmed Ali
- Department of Microbiology and Immunology, Faculty of Pharmacy, Sinai University, Sinai, Egypt
| | - Lina M Ismail
- Department of Biotechnology and Molecular Chemistry, Faculty of Science, Cairo University, Giza, Egypt
- Creative Egyptian Biotechnologists (CEB), Giza, Egypt
| | | | - Walid F Elkhatib
- Department of Microbiology and Immunology, Faculty of Pharmacy, Galala University, Suez, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Heba Aboul-Ella
- Department of Pharmacognosy, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University (ECU), Cairo, Egypt
- Scientific Research Group in Egypt (SRGE), Cairo, Egypt
| |
Collapse
|
2
|
Yamaguchi A, Aino H, Tsumuraya T, Hirama M, Fujii I. Humanization and characterization of an anti-ciguatoxin CTX3C monoclonal antibody. Toxicon 2024; 247:107848. [PMID: 38964619 DOI: 10.1016/j.toxicon.2024.107848] [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: 02/29/2024] [Revised: 05/02/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Ciguatera poisoning (CP), caused by ciguatoxins (CTXs), is one of the most common food-borne diseases, affecting more than 50,000 people each year. In most cases, CP are managed with symptomatic and supportive remedies, and no specific treatment has been devised. In this study, toward the development of therapeutic antibodies for CP, we examined to humanize mouse anti-CTX3C antibody 10C9 (m10C9), which exhibited neutralizing activity against ciguatoxin in vitro and in vivo. The complementarity determining regions were grafted onto a human germline sequence with high sequence identity to m10C9, and the backmutations were examined to maintain the binding affinity. The optimized humanized antibody, Opt.h10C9Fab, showed a strong binding affinity to CTX3C with a high affinity (KD = 19.0 nM), and only two backmutations of ArgL46 and CysH94 in the framework regions were involved in determining the antigen binding affinity.
Collapse
Affiliation(s)
- Asako Yamaguchi
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Sakai, Osaka, 599-8531, Japan; Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, Sakai, Osaka, 599-8531, Japan
| | - Hiroaki Aino
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Sakai, Osaka, 599-8531, Japan
| | - Takeshi Tsumuraya
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Sakai, Osaka, 599-8531, Japan; Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, Sakai, Osaka, 599-8531, Japan
| | - Masahiro Hirama
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, Sakai, Osaka, 599-8531, Japan
| | - Ikuo Fujii
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Sakai, Osaka, 599-8531, Japan; Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, Sakai, Osaka, 599-8531, Japan.
| |
Collapse
|
3
|
Busato D, Capolla S, Durigutto P, Mossenta M, Bozzer S, Sblattero D, Macor P, Dal Bo M, Toffoli G. A novel complement-fixing IgM antibody targeting GPC1 as a useful immunotherapeutic strategy for the treatment of pancreatic ductal adenocarcinoma. J Transl Med 2023; 21:864. [PMID: 38017492 PMCID: PMC10685509 DOI: 10.1186/s12967-023-04745-9] [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: 10/06/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancers with a very low survival rate at 5 years. The use of chemotherapeutic agents results in only modest prolongation of survival and is generally associated with the occurrence of toxicity effects. Antibody-based immunotherapy has been proposed for the treatment of PDAC, but its efficacy has so far proved limited. The proteoglycan glypican-1 (GPC1) may be a useful immunotherapeutic target because it is highly expressed on the surface of PDAC cells, whereas it is not expressed or is expressed at very low levels in benign neoplastic lesions, chronic pancreatitis, and normal adult tissues. Here, we developed and characterized a specific mouse IgM antibody (AT101) targeting GPC1. METHODS We developed a mouse monoclonal antibody of the IgM class directed against an epitope of GPC1 in close proximity to the cell membrane. For this purpose, a 46 amino acid long peptide of the C-terminal region was used to immunize mice by an in-vivo electroporation protocol followed by serum titer and hybridoma formation. RESULTS The ability of AT101 to bind the GPC1 protein was demonstrated by ELISA, and by flow cytometry and immunofluorescence analysis in the GPC1-expressing "PDAC-like" BXPC3 cell line. In-vivo experiments in the BXPC3 xenograft model showed that AT101 was able to bind GPC1 on the cell surface and accumulate in the BXPC3 tumor masses. Ex-vivo analyses of BXPC3 tumor masses showed that AT101 was able to recruit immunological effectors (complement system components, NK cells, macrophages) to the tumor site and damage PDAC tumor tissue. In-vivo treatment with AT101 reduced tumor growth and prolonged survival of mice with BXPC3 tumor (p < 0.0001). CONCLUSIONS These results indicate that AT101, an IgM specific for an epitope of GPC1 close to PDAC cell surface, is a promising immunotherapeutic agent for GPC1-expressing PDAC, being able to selectively activate the complement system and recruit effector cells in the tumor microenvironment, thus allowing to reduce tumor mass growth and improve survival in treated mice.
Collapse
Affiliation(s)
- Davide Busato
- Experimental and Clinical Pharmacology, Centro Di Riferimento Oncologico (CRO) Di Aviano IRCCS, 33081, Aviano, Italy
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Sara Capolla
- Experimental and Clinical Pharmacology, Centro Di Riferimento Oncologico (CRO) Di Aviano IRCCS, 33081, Aviano, Italy
| | - Paolo Durigutto
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Monica Mossenta
- Experimental and Clinical Pharmacology, Centro Di Riferimento Oncologico (CRO) Di Aviano IRCCS, 33081, Aviano, Italy
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Sara Bozzer
- Experimental and Clinical Pharmacology, Centro Di Riferimento Oncologico (CRO) Di Aviano IRCCS, 33081, Aviano, Italy
| | - Daniele Sblattero
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology, Centro Di Riferimento Oncologico (CRO) Di Aviano IRCCS, 33081, Aviano, Italy.
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology, Centro Di Riferimento Oncologico (CRO) Di Aviano IRCCS, 33081, Aviano, Italy
| |
Collapse
|
4
|
Fernández-Quintero ML, Pomarici ND, Fischer ALM, Hoerschinger VJ, Kroell KB, Riccabona JR, Kamenik AS, Loeffler JR, Ferguson JA, Perrett HR, Liedl KR, Han J, Ward AB. Structure and Dynamics Guiding Design of Antibody Therapeutics and Vaccines. Antibodies (Basel) 2023; 12:67. [PMID: 37873864 PMCID: PMC10594513 DOI: 10.3390/antib12040067] [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: 09/05/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/25/2023] Open
Abstract
Antibodies and other new antibody-like formats have emerged as one of the most rapidly growing classes of biotherapeutic proteins. Understanding the structural features that drive antibody function and, consequently, their molecular recognition is critical for engineering antibodies. Here, we present the structural architecture of conventional IgG antibodies alongside other formats. We emphasize the importance of considering antibodies as conformational ensembles in solution instead of focusing on single-static structures because their functions and properties are strongly governed by their dynamic nature. Thus, in this review, we provide an overview of the unique structural and dynamic characteristics of antibodies with respect to their antigen recognition, biophysical properties, and effector functions. We highlight the numerous technical advances in antibody structure prediction and design, enabled by the vast number of experimentally determined high-quality structures recorded with cryo-EM, NMR, and X-ray crystallography. Lastly, we assess antibody and vaccine design strategies in the context of structure and dynamics.
Collapse
Affiliation(s)
- Monica L. Fernández-Quintero
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80/82, A-6020 Innsbruck, Austria
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nancy D. Pomarici
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80/82, A-6020 Innsbruck, Austria
| | - Anna-Lena M. Fischer
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80/82, A-6020 Innsbruck, Austria
| | - Valentin J. Hoerschinger
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80/82, A-6020 Innsbruck, Austria
| | - Katharina B. Kroell
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80/82, A-6020 Innsbruck, Austria
| | - Jakob R. Riccabona
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80/82, A-6020 Innsbruck, Austria
| | - Anna S. Kamenik
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80/82, A-6020 Innsbruck, Austria
| | - Johannes R. Loeffler
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80/82, A-6020 Innsbruck, Austria
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - James A. Ferguson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Hailee R. Perrett
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Klaus R. Liedl
- Institute of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80/82, A-6020 Innsbruck, Austria
| | - Julianna Han
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
5
|
Zhang Y, Li Q, Luo L, Duan C, Shen J, Wang Z. Application of germline antibody features to vaccine development, antibody discovery, antibody optimization and disease diagnosis. Biotechnol Adv 2023; 65:108143. [PMID: 37023966 DOI: 10.1016/j.biotechadv.2023.108143] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
Although the efficacy and commercial success of vaccines and therapeutic antibodies have been tremendous, designing and discovering new drug candidates remains a labor-, time- and cost-intensive endeavor with high risks. The main challenges of vaccine development are inducing a strong immune response in broad populations and providing effective prevention against a group of highly variable pathogens. Meanwhile, antibody discovery faces several great obstacles, especially the blindness in antibody screening and the unpredictability of the developability and druggability of antibody drugs. These challenges are largely due to poorly understanding of germline antibodies and the antibody responses to pathogen invasions. Thanks to the recent developments in high-throughput sequencing and structural biology, we have gained insight into the germline immunoglobulin (Ig) genes and germline antibodies and then the germline antibody features associated with antigens and disease manifestation. In this review, we firstly outline the broad associations between germline antibodies and antigens. Moreover, we comprehensively review the recent applications of antigen-specific germline antibody features, physicochemical properties-associated germline antibody features, and disease manifestation-associated germline antibody features on vaccine development, antibody discovery, antibody optimization, and disease diagnosis. Lastly, we discuss the bottlenecks and perspectives of current and potential applications of germline antibody features in the biotechnology field.
Collapse
Affiliation(s)
- Yingjie Zhang
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Qing Li
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Liang Luo
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Changfei Duan
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Zhanhui Wang
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China.
| |
Collapse
|
6
|
Das NC, Chakraborty P, Bayry J, Mukherjee S. Comparative Binding Ability of Human Monoclonal Antibodies against Omicron Variants of SARS-CoV-2: An In Silico Investigation. Antibodies (Basel) 2023; 12:17. [PMID: 36975364 PMCID: PMC10045060 DOI: 10.3390/antib12010017] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/26/2023] Open
Abstract
Mutation(s) in the spike protein is the major characteristic trait of newly emerged SARS-CoV-2 variants such as Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), and Delta-plus. Omicron (B.1.1.529) is the latest addition and it has been characterized by high transmissibility and the ability to escape host immunity. Recently developed vaccines and repurposed drugs exert limited action on Omicron strains and hence new therapeutics are immediately needed. Herein, we have explored the efficiency of twelve therapeutic monoclonal antibodies (mAbs) targeting the RBD region of the spike glycoprotein against all the Omicron variants bearing a mutation in spike protein through molecular docking and molecular dynamics simulation. Our in silico evidence reveals that adintivimab, beludivimab, and regadanivimab are the most potent mAbs to form strong biophysical interactions and neutralize most of the Omicron variants. Considering the efficacy of mAbs, we incorporated CDRH3 of beludavimab within the framework of adintrevimab, which displayed a more intense binding affinity towards all of the Omicron variants viz. BA.1, BA.2, BA.2.12.1, BA.4, and BA.5. Furthermore, the cDNA of chimeric mAb was cloned in silico within pET30ax for recombinant production. In conclusion, the present study represents the candidature of human mAbs (beludavimab and adintrevimab) and the therapeutic potential of designed chimeric mAb for treating Omicron-infected patients.
Collapse
Affiliation(s)
- Nabarun Chandra Das
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713 340, India
| | - Pritha Chakraborty
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713 340, India
| | - Jagadeesh Bayry
- Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad 678 623, India
| | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713 340, India
| |
Collapse
|
7
|
Lefranc MP, Lefranc G. IMGT ®Homo sapiens IG and TR Loci, Gene Order, CNV and Haplotypes: New Concepts as a Paradigm for Jawed Vertebrates Genome Assemblies. Biomolecules 2022; 12:381. [PMID: 35327572 PMCID: PMC8945572 DOI: 10.3390/biom12030381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 02/04/2023] Open
Abstract
IMGT®, the international ImMunoGeneTics information system®, created in 1989, by Marie-Paule Lefranc (Université de Montpellier and CNRS), marked the advent of immunoinformatics, a new science which emerged at the interface between immunogenetics and bioinformatics for the study of the adaptive immune responses. IMGT® is based on a standardized nomenclature of the immunoglobulin (IG) and T cell receptor (TR) genes and alleles from fish to humans and on the IMGT unique numbering for the variable (V) and constant (C) domains of the immunoglobulin superfamily (IgSF) of vertebrates and invertebrates, and for the groove (G) domain of the major histocompatibility (MH) and MH superfamily (MhSF) proteins. IMGT® comprises 7 databases, 17 tools and more than 25,000 pages of web resources for sequences, genes and structures, based on the IMGT Scientific chart rules generated from the IMGT-ONTOLOGY axioms and concepts. IMGT® reference directories are used for the analysis of the NGS high-throughput expressed IG and TR repertoires (natural, synthetic and/or bioengineered) and for bridging sequences, two-dimensional (2D) and three-dimensional (3D) structures. This manuscript focuses on the IMGT®Homo sapiens IG and TR loci, gene order, copy number variation (CNV) and haplotypes new concepts, as a paradigm for jawed vertebrates genome assemblies.
Collapse
Affiliation(s)
- Marie-Paule Lefranc
- IMGT®, The International ImMunoGeneTics Information System®, Laboratoire d’Immuno Génétique Moléculaire (LIGM), Institut de Génétique Humaine (IGH), Université de Montpellier (UM), Centre National de la Recherche Scientifique (CNRS), UMR 9002 CNRS-UM, 141 rue de la Cardonille, CEDEX 5, 34396 Montpellier, France
| | - Gérard Lefranc
- IMGT®, The International ImMunoGeneTics Information System®, Laboratoire d’Immuno Génétique Moléculaire (LIGM), Institut de Génétique Humaine (IGH), Université de Montpellier (UM), Centre National de la Recherche Scientifique (CNRS), UMR 9002 CNRS-UM, 141 rue de la Cardonille, CEDEX 5, 34396 Montpellier, France
| |
Collapse
|
8
|
Avril A, Tournier JN, Paucod JC, Fournes B, Thullier P, Pelat T. Antibodies against Anthrax Toxins: A Long Way from Benchlab to the Bedside. Toxins (Basel) 2022; 14:172. [PMID: 35324669 PMCID: PMC8955606 DOI: 10.3390/toxins14030172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 02/01/2023] Open
Abstract
Anthrax is an acute disease caused by the bacterium Bacillus anthracis, and is a potential biowarfare/bioterrorist agent. Its pulmonary form, caused by inhalation of the spores, is highly lethal and is mainly related to injury caused by the toxins secretion. Antibodies neutralizing the toxins of B. anthracis are regarded as promising therapeutic drugs, and two are already approved by the Federal Drug Administration. We developed a recombinant human-like humanized antibody, 35PA83 6.20, that binds the protective antigen and that neutralized anthrax toxins in-vivo in White New Zealand rabbits infected with the lethal 9602 strain by intranasal route. Considering these promising results, the preclinical and clinical phase one development was funded and a program was started. Unfortunately, after 5 years, the preclinical development was cancelled due to industrial and scientific issues. This shutdown underlined the difficulty particularly, but not only, for an academic laboratory to proceed to clinical development, despite the drug candidate being promising. Here, we review our strategy and some preliminary results, and we discuss the issues that led to the no-go decision of the pre-clinical development of 35PA83 6.20 mAb. Our review provides general information to the laboratories planning a (pre-)clinical development.
Collapse
Affiliation(s)
- Arnaud Avril
- Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France; (J.-N.T.); (J.-C.P.); (P.T.); (T.P.)
| | - Jean-Nicolas Tournier
- Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France; (J.-N.T.); (J.-C.P.); (P.T.); (T.P.)
- Ecole du Val-de-Grâce, 75005 Paris, France
| | - Jean-Charles Paucod
- Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France; (J.-N.T.); (J.-C.P.); (P.T.); (T.P.)
| | - Bénédicte Fournes
- Laboratoire Français du Fractionnement et des Biotechnologies, 91940 Les Ulis, France;
| | - Philippe Thullier
- Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France; (J.-N.T.); (J.-C.P.); (P.T.); (T.P.)
| | - Thibaut Pelat
- Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France; (J.-N.T.); (J.-C.P.); (P.T.); (T.P.)
| |
Collapse
|
9
|
Hsieh YC, Liao JM, Chuang KH, Ho KW, Hong ST, Liu HJ, Huang BC, Chen IJ, Liu YL, Wang JY, Tsai HL, Su YC, Wang YT, Cheng TL. A universal in silico V(D)J recombination strategy for developing humanized monoclonal antibodies. J Nanobiotechnology 2022; 20:58. [PMID: 35101043 PMCID: PMC8805405 DOI: 10.1186/s12951-022-01259-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/12/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Humanization of mouse monoclonal antibodies (mAbs) is crucial for reducing their immunogenicity in humans. However, humanized mAbs often lose their binding affinities. Therefore, an in silico humanization method that can prevent the loss of the binding affinity of mAbs is needed. METHODS We developed an in silico V(D)J recombination platform in which we used V(D)J human germline gene sequences to design five humanized candidates of anti-tumor necrosis factor (TNF)-α mAbs (C1-C5) by using different human germline templates. The candidates were subjected to molecular dynamics simulation. In addition, the structural similarities of their complementarity-determining regions (CDRs) to those of original mouse mAbs were estimated to derive the weighted interatomic root mean squared deviation (wRMSDi) value. Subsequently, the correlation of the derived wRMSDi value with the half maximal effective concentration (EC50) and the binding affinity (KD) of the humanized anti-TNF-α candidates was examined. To confirm whether our in silico estimation method can be used for other humanized mAbs, we tested our method using the anti-epidermal growth factor receptor (EGFR) a4.6.1, anti-glypican-3 (GPC3) YP9.1 and anti-α4β1 integrin HP1/2L mAbs. RESULTS The R2 value for the correlation between the wRMSDi and log(EC50) of the recombinant Remicade and those of the humanized anti-TNF-α candidates was 0.901, and the R2 value for the correlation between wRMSDi and log(KD) was 0.9921. The results indicated that our in silico V(D)J recombination platform could predict the binding affinity of humanized candidates and successfully identify the high-affinity humanized anti-TNF-α antibody (Ab) C1 with a binding affinity similar to that of the parental chimeric mAb (5.13 × 10-10). For the anti-EGFR a4.6.1, anti-GPC3 YP9.1, and anti-α4β1 integrin HP1/2L mAbs, the wRMSDi and log(EC50) exhibited strong correlations (R2 = 0.9908, 0.9999, and 0.8907, respectively). CONCLUSIONS Our in silico V(D)J recombination platform can facilitate the development of humanized mAbs with low immunogenicity and high binding affinities. This platform can directly transform numerous mAbs with therapeutic potential to humanized or even human therapeutic Abs for clinical use.
Collapse
Affiliation(s)
- Yuan-Chin Hsieh
- Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan
- School of Medicine for International Students, I-Shou University, No.8, Yida Rd., Jiaosu Village Yanchao District, Kaohsiung, 82445, Taiwan
| | - Jun-Min Liao
- Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan
| | - Kuo-Hsiang Chuang
- Graduate Institute of Pharmacognosy, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
- Ph.D. Program for Clinical Drug Discovery From Botanical Herbs, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Kai-Wen Ho
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan
| | - Shih-Ting Hong
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan
| | - Hui-Ju Liu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan
| | - Bo-Cheng Huang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, 70 Lien-hai Road, Kaohsiung, 804, Taiwan
| | - I-Ju Chen
- School of Medicine, I-Shou University, No.8, Yida Rd., Jiaosu Village Yanchao District, Kaohsiung, 82445, Taiwan
| | - Yen-Ling Liu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan
| | - Jaw-Yuan Wang
- Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan
- Department of Surgery, Faculty of Medicine, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, No.100, Tzyou 1st Rd., Sanmin Dist., Kaohsiung, 80756, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan
| | - Hsiang-Lin Tsai
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan
- Department of Surgery, Faculty of Medicine, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan
| | - Yu-Cheng Su
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, No. 1001, Daxue Rd. East Dist., Hsin-Chu, 300, Taiwan
| | - Yen-Tseng Wang
- Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan.
- School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan.
| | - Tian-Lu Cheng
- Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan.
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
10
|
Das NC, Chakraborty P, Bayry J, Mukherjee S. In Silico Analyses on the Comparative Potential of Therapeutic Human Monoclonal Antibodies Against Newly Emerged SARS-CoV-2 Variants Bearing Mutant Spike Protein. Front Immunol 2022; 12:782506. [PMID: 35082779 PMCID: PMC8784557 DOI: 10.3389/fimmu.2021.782506] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/07/2021] [Indexed: 12/19/2022] Open
Abstract
Since the start of the pandemic, SARS-CoV-2 has already infected more than 250 million people globally, with more than five million fatal cases and huge socio-economic losses. In addition to corticosteroids, and antiviral drugs like remdesivir, various immunotherapies including monoclonal antibodies (mAbs) to S protein of SARS-CoV-2 have been investigated to treat COVID-19 patients. These mAbs were initially developed against the wild-type SARS-CoV-2; however, emergence of variant forms of SARS-CoV-2 having mutations in the spike protein in several countries including India raised serious questions on the potential use of these mAbs against SARS-CoV-2 variants. In this study, using an in silico approach, we have examined the binding abilities of eight mAbs against several SARS-CoV-2 variants of Alpha (B.1.1.7) and Delta (B.1.617.2) lineages. The structure of the Fab region of each mAb was designed in silico and subjected to molecular docking against each mutant protein. mAbs were subjected to two levels of selection based on their binding energy, stability, and conformational flexibility. Our data reveal that tixagevimab, regdanvimab, and cilgavimab can efficiently neutralize most of the SARS-CoV-2 Alpha strains while tixagevimab, bamlanivimab, and sotrovimab can form a stable complex with the Delta variants. Based on these data, we have designed, by in silico, a chimeric antibody by conjugating the CDRH3 of regdanivimab with a sotrovimab framework to combat the variants that could potentially escape from the mAb-mediated neutralization. Our finding suggests that though currently available mAbs could be used to treat COVID-19 caused by the variants of SARS-CoV-2, better results could be expected with the chimeric antibodies.
Collapse
Affiliation(s)
- Nabarun Chandra Das
- Integrative Biochemistry and Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, India
| | - Pritha Chakraborty
- Integrative Biochemistry and Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, India
| | - Jagadeesh Bayry
- Department of Biological Sciences and Engineering, Indian Institute of Technology Palakkad, Palakkad, India
| | - Suprabhat Mukherjee
- Integrative Biochemistry and Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, India
| |
Collapse
|
11
|
Lefranc MP, Lefranc G. Immunoglobulins or Antibodies: IMGT ® Bridging Genes, Structures and Functions. Biomedicines 2020; 8:E319. [PMID: 32878258 PMCID: PMC7555362 DOI: 10.3390/biomedicines8090319] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/18/2022] Open
Abstract
IMGT®, the international ImMunoGeneTics® information system founded in 1989 by Marie-Paule Lefranc (Université de Montpellier and CNRS), marked the advent of immunoinformatics, a new science at the interface between immunogenetics and bioinformatics. For the first time, the immunoglobulin (IG) or antibody and T cell receptor (TR) genes were officially recognized as 'genes' as well as were conventional genes. This major breakthrough has allowed the entry, in genomic databases, of the IG and TR variable (V), diversity (D) and joining (J) genes and alleles of Homo sapiens and of other jawed vertebrate species, based on the CLASSIFICATION axiom. The second major breakthrough has been the IMGT unique numbering and the IMGT Collier de Perles for the V and constant (C) domains of the IG and TR and other proteins of the IG superfamily (IgSF), based on the NUMEROTATION axiom. IMGT-ONTOLOGY axioms and concepts bridge genes, sequences, structures and functions, between biological and computational spheres in the IMGT® system (Web resources, databases and tools). They provide the IMGT Scientific chart rules to identify, to describe and to analyse the IG complex molecular data, the huge diversity of repertoires, the genetic (alleles, allotypes, CNV) polymorphisms, the IG dual function (paratope/epitope, effector properties), the antibody humanization and engineering.
Collapse
Affiliation(s)
- Marie-Paule Lefranc
- IMGT, The International ImMunoGeneTics Information System, Laboratoire d’ImmunoGénétique Moléculaire LIGM, Institut de Génétique Humaine IGH, Université de Montpellier UM, Centre National de la Recherche Scientifique CNRS, UMR 9002 CNRS-UM, 141 Rue de la Cardonille, CEDEX 5, 34396 Montpellier, France
| | - Gérard Lefranc
- IMGT, The International ImMunoGeneTics Information System, Laboratoire d’ImmunoGénétique Moléculaire LIGM, Institut de Génétique Humaine IGH, Université de Montpellier UM, Centre National de la Recherche Scientifique CNRS, UMR 9002 CNRS-UM, 141 Rue de la Cardonille, CEDEX 5, 34396 Montpellier, France
| |
Collapse
|
12
|
Kuroda D, Tsumoto K. Engineering Stability, Viscosity, and Immunogenicity of Antibodies by Computational Design. J Pharm Sci 2020; 109:1631-1651. [DOI: 10.1016/j.xphs.2020.01.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/25/2019] [Accepted: 01/10/2020] [Indexed: 12/18/2022]
|
13
|
Lu RM, Hwang YC, Liu IJ, Lee CC, Tsai HZ, Li HJ, Wu HC. Development of therapeutic antibodies for the treatment of diseases. J Biomed Sci 2020; 27:1. [PMID: 31894001 PMCID: PMC6939334 DOI: 10.1186/s12929-019-0592-z] [Citation(s) in RCA: 1099] [Impact Index Per Article: 274.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022] Open
Abstract
It has been more than three decades since the first monoclonal antibody was approved by the United States Food and Drug Administration (US FDA) in 1986, and during this time, antibody engineering has dramatically evolved. Current antibody drugs have increasingly fewer adverse effects due to their high specificity. As a result, therapeutic antibodies have become the predominant class of new drugs developed in recent years. Over the past five years, antibodies have become the best-selling drugs in the pharmaceutical market, and in 2018, eight of the top ten bestselling drugs worldwide were biologics. The global therapeutic monoclonal antibody market was valued at approximately US$115.2 billion in 2018 and is expected to generate revenue of $150 billion by the end of 2019 and $300 billion by 2025. Thus, the market for therapeutic antibody drugs has experienced explosive growth as new drugs have been approved for treating various human diseases, including many cancers, autoimmune, metabolic and infectious diseases. As of December 2019, 79 therapeutic mAbs have been approved by the US FDA, but there is still significant growth potential. This review summarizes the latest market trends and outlines the preeminent antibody engineering technologies used in the development of therapeutic antibody drugs, such as humanization of monoclonal antibodies, phage display, the human antibody mouse, single B cell antibody technology, and affinity maturation. Finally, future applications and perspectives are also discussed.
Collapse
Affiliation(s)
- Ruei-Min Lu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Yu-Chyi Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan
| | - I-Ju Liu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Chi-Chiu Lee
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Han-Zen Tsai
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Hsin-Jung Li
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan. .,, 128 Academia Rd., Section 2, Nankang, Taipei, 11529, Taiwan.
| |
Collapse
|
14
|
Chiu ML, Goulet DR, Teplyakov A, Gilliland GL. Antibody Structure and Function: The Basis for Engineering Therapeutics. Antibodies (Basel) 2019; 8:antib8040055. [PMID: 31816964 PMCID: PMC6963682 DOI: 10.3390/antib8040055] [Citation(s) in RCA: 243] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 12/11/2022] Open
Abstract
Antibodies and antibody-derived macromolecules have established themselves as the mainstay in protein-based therapeutic molecules (biologics). Our knowledge of the structure–function relationships of antibodies provides a platform for protein engineering that has been exploited to generate a wide range of biologics for a host of therapeutic indications. In this review, our basic understanding of the antibody structure is described along with how that knowledge has leveraged the engineering of antibody and antibody-related therapeutics having the appropriate antigen affinity, effector function, and biophysical properties. The platforms examined include the development of antibodies, antibody fragments, bispecific antibody, and antibody fusion products, whose efficacy and manufacturability can be improved via humanization, affinity modulation, and stability enhancement. We also review the design and selection of binding arms, and avidity modulation. Different strategies of preparing bispecific and multispecific molecules for an array of therapeutic applications are included.
Collapse
Affiliation(s)
- Mark L. Chiu
- Drug Product Development Science, Janssen Research & Development, LLC, Malvern, PA 19355, USA
- Correspondence:
| | - Dennis R. Goulet
- Department of Medicinal Chemistry, University of Washington, P.O. Box 357610, Seattle, WA 98195-7610, USA;
| | - Alexey Teplyakov
- Biologics Research, Janssen Research & Development, LLC, Spring House, PA 19477, USA; (A.T.); (G.L.G.)
| | - Gary L. Gilliland
- Biologics Research, Janssen Research & Development, LLC, Spring House, PA 19477, USA; (A.T.); (G.L.G.)
| |
Collapse
|
15
|
Kuramochi T, Igawa T, Tsunoda H, Hattori K. Humanization and Simultaneous Optimization of Monoclonal Antibody. Methods Mol Biol 2019; 1904:213-230. [PMID: 30539472 DOI: 10.1007/978-1-4939-8958-4_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Antibody humanization is an essential technology for reducing the potential risk of immunogenicity associated with animal-derived antibodies and has been applied to a majority of the therapeutic antibodies on the market. For developing an antibody molecule as a pharmaceutical at the current biotechnology level, however, other properties also have to be considered in parallel with humanization in antibody generation and optimization. This section describes the critical properties of therapeutic antibodies that should be sufficiently qualified, including immunogenicity, binding affinity, physicochemical stability, expression in host cells and pharmacokinetics, and the basic methodologies of antibody engineering involved. By simultaneously optimizing the antibody molecule in light of these properties, it should prove possible to shorten the research and development period necessary to identify a highly qualified clinical candidate and consequently accelerate the start of the clinical trial.
Collapse
Affiliation(s)
| | - Tomoyuki Igawa
- Chugai Pharmabody Research Pte. Ltd., Singapore, Singapore
| | - Hiroyuki Tsunoda
- Research Division, Chugai Pharmaceutical, Kamakura, Kanagawa, Japan
| | - Kunihiro Hattori
- Research Division, Chugai Pharmaceutical, Kamakura, Kanagawa, Japan
| |
Collapse
|
16
|
Mayrhofer P, Kunert R. Nomenclature of humanized mAbs: Early concepts, current challenges and future perspectives. Hum Antibodies 2019; 27:37-51. [PMID: 30103312 PMCID: PMC6294595 DOI: 10.3233/hab-180347] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Nomenclature of monoclonal antibodies traditionally followed a strict scheme indicating target and species information. Because of the rapid advances in this field, emphasized by approval of four humanized and six human antibodies in 2017, the International Nonproprietary Name of new antibodies was updated profoundly by removing the species substem completely. In this review we give an overview about what developments led to the preference of the scientific community towards human-like antibodies. We summarize the major updates in naming schemes that tried to classify antibodies according to their humanization technique or to the final primary sequence and how this led to the erroneous perception to indicate expected immunogenicity. Following the new 2017 nomenclature update, there will not be any information available about the species origin in the names of new antibodies, which emphasizes the need for providing additional supplemental information to the scientific community and develop tools to accurately estimate and control the safety of new monoclonal antibody molecules.
Collapse
Affiliation(s)
- Patrick Mayrhofer
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), 1190 Vienna, Austria
| | - Renate Kunert
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), 1190 Vienna, Austria
| |
Collapse
|
17
|
Avril A. Therapeutic Antibodies for Biodefense. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1053:173-205. [PMID: 29549640 DOI: 10.1007/978-3-319-72077-7_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Diseases can be caused naturally by biological agents such as bacteria, viruses and toxins (natural risk). However, such biological agents can be intentionally disseminated in the environment by a State (military context) or terrorists to cause diseases in a population or livestock, to destabilize a nation by creating a climate of terror, destabilizing the economy and undermining institutions. Biological agents can be classified according to the severity of illness they cause, its mortality and how easily the agent can be spread. The Centers for Diseases Control and Prevention (CDC) classify biological agents in three categories (A, B and C); Category A consists of the six pathogens most suitable for use as bioweapons (Bacillus anthracis, Yersinia pestis, Francisella tularensis, botulinum neurotoxins, smallpox and viral hemorrhagic fevers). Antibodies represent a perfect biomedical countermeasure as they present both prophylactic and therapeutic properties, act fast and are highly specific to the target. This review focuses on the main biological agents that could be used as bioweapons, the history of biowarfare and antibodies that have been developed to neutralize these agents.
Collapse
Affiliation(s)
- Arnaud Avril
- Département des maladies infectieuses, Unité biothérapies anti-infectieuses et immunité, Institut de Recherche Biomédical des Armées, Brétigny-sur-Orge, France.
| |
Collapse
|
18
|
Estupina P, Fontayne A, Barret JM, Kersual N, Dubreuil O, Le Blay M, Pichard A, Jarlier M, Pugnière M, Chauvin M, Chardès T, Pouget JP, Deshayes E, Rossignol A, Abache T, de Romeuf C, Terrier A, Verhaeghe L, Gaucher C, Prost JF, Pèlegrin A, Navarro-Teulon I. The anti-tumor efficacy of 3C23K, a glyco-engineered humanized anti-MISRII antibody, in an ovarian cancer model is mainly mediated by engagement of immune effector cells. Oncotarget 2018; 8:37061-37079. [PMID: 28427157 PMCID: PMC5513714 DOI: 10.18632/oncotarget.15715] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/11/2017] [Indexed: 01/06/2023] Open
Abstract
Ovarian cancer is the leading cause of death in women with gynecological cancers and despite recent advances, new and more efficient therapies are crucially needed. Müllerian Inhibiting Substance type II Receptor (MISRII, also named AMHRII) is expressed in most ovarian cancer subtypes and is a novel potential target for ovarian cancer immunotherapy. We previously developed and tested 12G4, the first murine monoclonal antibody (MAb) against human MISRII. Here, we report the humanization, affinity maturation and glyco-engineering steps of 12G4 to generate the Fc-optimized 3C23K MAb, and the evaluation of its in vivo anti-tumor activity. The epitopes of 3C23K and 12G4 were strictly identical and 3C23K affinity for MISRII was enhanced by a factor of about 14 (KD = 5.5 × 10−11 M vs 7.9 × 10−10 M), while the use of the EMABling® platform allowed the production of a low-fucosylated 3C23K antibody with a 30-fold KD improvement of its affinity to FcγRIIIa. In COV434-MISRII tumor-bearing mice, 3C23K reduced tumor growth more efficiently than 12G4 and its combination with carboplatin was more efficient than each monotherapy with a mean tumor size of 500, 1100 and 100 mm3 at the end of treatment with 3C23K (10 mg/kg, Q3-4D12), carboplatin (60 mg/kg, Q7D4) and 3C23K+carboplatin, respectively. Conversely, 3C23K-FcKO, a 3C23K form without affinity for the FcγRIIIa receptor, did not display any anti-tumor effect in vivo. These results strongly suggested that 3C23K mechanisms of action are mainly Fc-related. In vitro, antibody-dependent cytotoxicity (ADCC) and antibody-dependent cell phagocytosis (ADCP) were induced by 3C23K, as demonstrated with human effector cells. Using human NK cells, 50% of the maximal lysis was obtained with a 46-fold lower concentration of low-fucosylated 3C23K (2.9 ng/ml) than of 3C23K expressed in CHO cells (133.35 ng/ml). As 3C23K induced strong ADCC with human PBMC but almost none with murine PBMC, antibody-dependent cell phagocytosis (ADCP) was then investigated. 3C23K-dependent (100 ng/ml) ADCP was more active with murine than human macrophages (only 10% of living target cells vs. about 25%). These in vitro results suggest that the reduced ADCC with murine effectors could be partially balanced by ADCP activity in in vivo experiments. Taken together, these preclinical data indicate that 3C23K is a new promising therapeutic candidate for ovarian cancer immunotherapy and justify its recent introduction in a phase I clinical trial.
Collapse
Affiliation(s)
- Pauline Estupina
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France.,INSERM, U896, Montpellier, F-34298, France.,Université Montpellier, Montpellier, F-34298, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, F-34298, France
| | | | | | - Nathalie Kersual
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France.,INSERM, U896, Montpellier, F-34298, France.,Université Montpellier, Montpellier, F-34298, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, F-34298, France
| | | | - Marion Le Blay
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France.,INSERM, U896, Montpellier, F-34298, France.,Université Montpellier, Montpellier, F-34298, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, F-34298, France
| | - Alexandre Pichard
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France.,INSERM, U896, Montpellier, F-34298, France.,Université Montpellier, Montpellier, F-34298, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, F-34298, France
| | - Marta Jarlier
- Institut Régional du Cancer de Montpellier, ICM, Montpellier, F-34298, France
| | - Martine Pugnière
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France.,INSERM, U896, Montpellier, F-34298, France.,Université Montpellier, Montpellier, F-34298, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, F-34298, France
| | - Maëva Chauvin
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France.,INSERM, U896, Montpellier, F-34298, France.,Université Montpellier, Montpellier, F-34298, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, F-34298, France
| | - Thierry Chardès
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France.,INSERM, U896, Montpellier, F-34298, France.,Université Montpellier, Montpellier, F-34298, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, F-34298, France
| | - Jean-Pierre Pouget
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France.,INSERM, U896, Montpellier, F-34298, France.,Université Montpellier, Montpellier, F-34298, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, F-34298, France
| | - Emmanuel Deshayes
- Institut Régional du Cancer de Montpellier, ICM, Montpellier, F-34298, France
| | | | | | | | | | | | | | | | - André Pèlegrin
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France.,INSERM, U896, Montpellier, F-34298, France.,Université Montpellier, Montpellier, F-34298, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, F-34298, France
| | - Isabelle Navarro-Teulon
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France.,INSERM, U896, Montpellier, F-34298, France.,Université Montpellier, Montpellier, F-34298, France.,Institut Régional du Cancer de Montpellier, ICM, Montpellier, F-34298, France
| |
Collapse
|
19
|
Abstract
IMGT®, the international ImMunoGeneTics information system® ( http://www.imgt.org ), was created in 1989 by Marie-Paule Lefranc (Université de Montpellier and CNRS) to manage the huge diversity of the antigen receptors, immunoglobulins (IG) or antibodies, and T cell receptors (TR). The founding of IMGT® marked the advent of immunoinformatics, which emerged at the interface between immunogenetics and bioinformatics. Standardized sequence and structure analysis of antibody using IMGT® databases and tools allow one to bridge, for the first time, the gap between antibody sequences and three-dimensional (3D) structures. This is achieved through the IMGT Scientific chart rules, based on the IMGT-ONTOLOGY concepts of classification (IMGT gene and allele nomenclature), description (IMGT standardized labels), and numerotation (IMGT unique numbering and IMGT Collier de Perles). IMGT® is acknowledged as the global reference for immunogenetics and immunoinformatics, and its standards are particularly useful for antibody engineering and humanization. IMGT® databases for antibody nucleotide sequences and genes include IMGT/LIGM-DB and IMGT/GENE-DB, respectively, and nucleotide sequence analysis is performed by the IMGT/V-QUEST and IMGT/JunctionAnalysis tools and for NGS by IMGT/HighV-QUEST. In this chapter, we focus on IMGT® databases and tools for amino acid sequences, two-dimensional (2D) and three-dimensional (3D) structures: the IMGT/DomainGapAlign and IMGT Collier de Perles tools and the IMGT/2Dstructure-DB and IMGT/3Dstructure-DB database. IMGT/mAb-DB provides the query interface for monoclonal antibodies (mAb), fusion proteins for immune applications (FPIA), and composite proteins for clinical applications (CPCA) and related proteins of interest (RPI) and links to the proposed and recommended lists of the World Health Organization International Nonproprietary Name (WHO INN) programme, to IMGT/2Dstructure-DB for amino acid sequences, and to IMGT/3Dstructure-DB and its associated tools (IMGT/StructuralQuery, IMGT/DomainSuperimpose) for crystallized antibodies.
Collapse
|
20
|
Banisadr A, Safdari Y, Kianmehr A, Pourafshar M. Production of a germline-humanized cetuximab scFv and evaluation of its activity in recognizing EGFR- overexpressing cancer cells. Hum Vaccin Immunother 2017; 14:856-863. [PMID: 29185855 DOI: 10.1080/21645515.2017.1407482] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
The aim of this study was to produce a humanized single chain antibody (scFv) as a potential improved product design to target EGFR (Epidermal Growth Factor Receptor) overexpressing cancer cells. To this end, CDR loops of cetuximab (an FDA-approved anti-EGFR antibody) were grafted on framework regions derived from type 3 (VH3 and VL3 kappa) human germline sequences to obtain recombinant VH and VL domainslinked together with a flexible linker [(Gly4Ser)3] to form a scFv. Codon optimized synthetic gene encoding the scFv (with NH2-VH-linker-VL-COOH orientation) was expressed in E. coli Origami™ 2(DE3) cells and the resultant scFv purified by using Ni-NTA affinity chromatography. The scFv, called cet.Hum scFv, was evaluated in ELISA and immunoblot to determine whether it can recognize EGFR. The scFv was able to recognize EGFR over-expressing cancer cells (A-431) but failed to detect cancer cells with low levels of EGFR (MCF-7 cells). Although the affinity of the scFv forA-431 cells was 9 fold lower than that of cetuximab, it was strong enough to recognize these cells. Considering its ability to bind EGFR molecules, the scFv may exhibit a potential application for the detection of EGFR-overexpressing cancer cells.
Collapse
Affiliation(s)
- Arsham Banisadr
- a Department of Medical Biotechnology , School of Advanced Technologies in Medicine, Golestan University of Medical Sciences , Gorgan , Iran
| | - Yaghoub Safdari
- b Golestan Research Center of Gastroenterology & Hepatology (GRCGH), Golestan University of Medical Sciences , Gorgan , Iran
| | - Anvarsadat Kianmehr
- c Cancer Research Center, Golestan University of Medical Sciences , Gorgan , Iran
| | - Mahdieh Pourafshar
- a Department of Medical Biotechnology , School of Advanced Technologies in Medicine, Golestan University of Medical Sciences , Gorgan , Iran
| |
Collapse
|
21
|
Steven J, Müller MR, Carvalho MF, Ubah OC, Kovaleva M, Donohoe G, Baddeley T, Cornock D, Saunders K, Porter AJ, Barelle CJ. In Vitro Maturation of a Humanized Shark VNAR Domain to Improve Its Biophysical Properties to Facilitate Clinical Development. Front Immunol 2017; 8:1361. [PMID: 29109729 PMCID: PMC5660122 DOI: 10.3389/fimmu.2017.01361] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/04/2017] [Indexed: 11/13/2022] Open
Abstract
Molecular engineering to increase the percentage identity to common human immunoglobulin sequences of non-human therapeutic antibodies and scaffolds has become standard practice. This strategy is often used to reduce undesirable immunogenic responses, accelerating the clinical development of candidate domains. The first humanized shark variable domain (VNAR) was reported by Kovalenko and colleagues and used the anti-human serum albumin (HSA) domain, clone E06, as a model to construct a number of humanized versions including huE06v1.10. This study extends this work by using huE06v1.10 as a template to isolate domains with improved biophysical properties and reduced antigenicity. Random mutagenesis was conducted on huE06v1.10 followed by refinement of clones through an off-rate ranking-based selection on target antigen. Many of these next-generation binders retained high affinity for target, together with good species cross-reactivity. Lead domains were assessed for any tendency to dimerize, tolerance to N- and C-terminal fusions, affinity, stability, and relative antigenicity in human dendritic cell assays. Functionality of candidate clones was verified in vivo through the extension of serum half-life in a typical drug format. From these analyses the domain, BA11, exhibited negligible antigenicity, high stability and high affinity for mouse, rat, and HSA. When these attributes were combined with demonstrable functionality in a rat model of PK, the BA11 clone was established as our clinical candidate.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Thomas Baddeley
- Department of Chemistry, University of Aberdeen, Aberdeen, United Kingdom
| | - Dawn Cornock
- Department of Chemistry, University of Aberdeen, Aberdeen, United Kingdom
| | | | - Andrew J Porter
- Elasmogen Ltd., Aberdeen, United Kingdom.,Department of Molecular and Cell Biology, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | | |
Collapse
|
22
|
Neutralization of Botulinum Neurotoxin Type E by a Humanized Antibody. Toxins (Basel) 2016; 8:toxins8090257. [PMID: 27626446 PMCID: PMC5037483 DOI: 10.3390/toxins8090257] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/15/2016] [Accepted: 08/22/2016] [Indexed: 12/21/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) cause botulism and are the deadliest naturally-occurring substances known to humans. BoNTs have been classified as one of the category A agents by the Centers for Disease Control and Prevention, indicating their potential use as bioweapons. To counter bio-threat and naturally-occurring botulism cases, well-tolerated antibodies by humans that neutralize BoNTs are relevant. In our previous work, we showed the neutralizing potential of macaque (Macaca fascicularis)-derived scFv-Fc (scFv-Fc ELC18) by in vitro endopeptidase immunoassay and ex vivo mouse phrenic nerve-hemidiaphragm assay by targeting the light chain of the botulinum neurotoxin type E (BoNT/E). In the present study, we germline-humanized scFv-Fc ELC18 into a full IgG hu8ELC18 to increase its immunotolerance by humans. We demonstrated the protection and prophylaxis capacity of hu8ELC18 against BoNT/E in a mouse model. A concentration of 2.5 ng/mouse of hu8ELC18 protected against 5 mouse lethal dose (MLD) in a mouse protection assay and complete neutralization of 1 LD50 of pure BoNT/E toxin was achieved with 8 ng of hu8ELC18 in mouse paralysis assay. Furthermore, hu8ELC18 protected mice from 5 MLD if injected up to 14 days prior to intraperitoneal BoNT/E administration. This newly-developed humanized IgG is expected to have high tolerance in humans.
Collapse
|
23
|
Miethe S, Mazuet C, Liu Y, Tierney R, Rasetti-Escargueil C, Avril A, Frenzel A, Thullier P, Pelat T, Urbain R, Fontayne A, Sesardic D, Hust M, Popoff MR. Development of Germline-Humanized Antibodies Neutralizing Botulinum Neurotoxin A and B. PLoS One 2016; 11:e0161446. [PMID: 27560688 PMCID: PMC4999263 DOI: 10.1371/journal.pone.0161446] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/07/2016] [Indexed: 12/20/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are counted among the most toxic substances known and are responsible for human botulism, a life-threatening disease characterized by flaccid muscle paralysis that occurs naturally by food poisoning or colonization of the gastrointestinal tract by BoNT-producing clostridia. To date, 7 serologically distinct serotypes of BoNT (serotype A-G) are known. Due to the high toxicity of BoNTs the Centers for Disease Control and Prevention (CDC) have classified BoNTs as category A agent, including the six biological agents with the highest potential risk of use as bioweapons. Well tolerated antibodies neutralizing BoNTs are required to deal with the potential risk. In a previous work, we described the development of scFv and scFv-Fc (Yumab) from macaque origin (Macaca fascicularis) neutralizing BoNT/A and B by targeting the heavy and light chain of each serotype. In the present study, we humanized the macaque antibodies SEM120-IIIC1 (anti-BoNT/A light chain), A1HC38 (anti-BoNT/A heavy chain), BLC3 (anti-BoNT/B light chain) and B2-7 (anti-BoNT/B heavy chain) by germline-humanization to obtain a better potential immunotolerance in humans. We increased the Germinality Index (GI) of SEM120-IIIC1 to 94.5%, for A1HC38, to 95% for BLC3 and to 94.4% for B2-7. Furthermore, the neutralization efficacies of the germline-humanized antibodies were analyzed in lethal and non-lethal in vivo mouse assays as full IgG. The germline-humanized IgGs hu8SEM120-IIIC1, hu8A1HC38, hu8BLC3 and hu8B2-7 were protective in vivo, when anti-heavy and anti-light chain antibodies were combined. The synergistic effect and high humanness of the selected IgGs makes them promising lead candidates for further clinical development.
Collapse
Affiliation(s)
- Sebastian Miethe
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Christelle Mazuet
- Institut Pasteur, Unité des Bactéries anaérobies et Toxines, Paris, France
| | - Yvonne Liu
- National Institute for Biological Standards and Control (NIBSC), Division of Bacteriology, Potters Bar, United Kingdom
| | - Robert Tierney
- National Institute for Biological Standards and Control (NIBSC), Division of Bacteriology, Potters Bar, United Kingdom
| | - Christine Rasetti-Escargueil
- National Institute for Biological Standards and Control (NIBSC), Division of Bacteriology, Potters Bar, United Kingdom
| | - Arnaud Avril
- Institut de Recherche Biomédicale des Armées (IRBA) Département des Maladies Infectieuses, Unité Interaction Hôte-Pathogène, Brétigny-sur-Orge, France
| | - André Frenzel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Philippe Thullier
- Institut de Recherche Biomédicale des Armées (IRBA) Département des Maladies Infectieuses, Unité Interaction Hôte-Pathogène, Brétigny-sur-Orge, France
| | - Thibaut Pelat
- Institut de Recherche Biomédicale des Armées (IRBA) Département des Maladies Infectieuses, Unité Interaction Hôte-Pathogène, Brétigny-sur-Orge, France
| | - Remi Urbain
- LFB Biotechnologies, Therapeutic Innovation Department, Lille, France
| | | | - Dorothea Sesardic
- National Institute for Biological Standards and Control (NIBSC), Division of Bacteriology, Potters Bar, United Kingdom
| | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
- * E-mail: (MRP); (MH)
| | - Michel Robert Popoff
- Institut Pasteur, Unité des Bactéries anaérobies et Toxines, Paris, France
- * E-mail: (MRP); (MH)
| |
Collapse
|
24
|
Martin AC, Rees AR. Extracting human antibody sequences from public databases for antibody humanization: high frequency of species assignment errors. Protein Eng Des Sel 2016; 29:403-408. [DOI: 10.1093/protein/gzw018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 05/09/2016] [Indexed: 11/13/2022] Open
|
25
|
Respaud R, Marchand D, Pelat T, Tchou-Wong KM, Roy CJ, Parent C, Cabrera M, Guillemain J, Mac Loughlin R, Levacher E, Fontayne A, Douziech-Eyrolles L, Junqua-Moullet A, Guilleminault L, Thullier P, Guillot-Combe E, Vecellio L, Heuzé-Vourc'h N. Development of a drug delivery system for efficient alveolar delivery of a neutralizing monoclonal antibody to treat pulmonary intoxication to ricin. J Control Release 2016; 234:21-32. [PMID: 27173943 DOI: 10.1016/j.jconrel.2016.05.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/06/2016] [Accepted: 05/07/2016] [Indexed: 12/13/2022]
Abstract
The high toxicity of ricin and its ease of production have made it a major bioterrorism threat worldwide. There is however no efficient and approved treatment for poisoning by ricin inhalation, although there have been major improvements in diagnosis and therapeutic strategies. We describe the development of an anti-ricin neutralizing monoclonal antibody (IgG 43RCA-G1) and a device for its rapid and effective delivery into the lungs for an application in humans. The antibody is a full-length IgG and binds to the ricin A-chain subunit with a high affinity (KD=53pM). Local administration of the antibody into the respiratory tract of mice 6h after pulmonary ricin intoxication allowed the rescue of 100% of intoxicated animals. Specific operational constraints and aerosolization stresses, resulting in protein aggregation and loss of activity, were overcome by formulating the drug as a dry-powder that is solubilized extemporaneously in a stabilizing solution to be nebulized. Inhalation studies in mice showed that this formulation of IgG 43RCA-G1 did not induce pulmonary inflammation. A mesh nebulizer was customized to improve IgG 43RCA-G1 deposition into the alveolar region of human lungs, where ricin aerosol particles mostly accumulate. The drug delivery system also comprises a semi-automatic reconstitution system to facilitate its use and a specific holding chamber to maximize aerosol delivery deep into the lung. In vivo studies in monkeys showed that drug delivery with the device resulted in a high concentration of IgG 43RCA-G1 in the airways for at least 6h after local deposition, which is consistent with the therapeutic window and limited passage into the bloodstream.
Collapse
Affiliation(s)
- Renaud Respaud
- Université François-Rabelais de Tours, UMR 1100, CHRU de Tours, Service de Pharmacie, F-37032 Tours, France; INSERM, Centre d'Etude des Pathologies Respiratoires, UMR 1100, F-37032 Tours, France
| | - Denis Marchand
- Université François Rabelais, UMR 1100, F-37032 Tours, France; INSERM, Centre d'Etude des Pathologies Respiratoires, UMR 1100, F-37032 Tours, France; Aerodrug, F-37032 Tours, France
| | - Thibaut Pelat
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines; Brétigny sur Orge, France; BIOTEM, Parc d'activité Bièvre Dauphine, Apprieu, France
| | - Kam-Meng Tchou-Wong
- NYU School of Medicine, Department of Environmental Medicine, 57 Old Forge Road, Tuxedo, New York 10987, USA
| | - Chad J Roy
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Division of Microbiology, Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Christelle Parent
- Université François Rabelais, UMR 1100, F-37032 Tours, France; INSERM, Centre d'Etude des Pathologies Respiratoires, UMR 1100, F-37032 Tours, France
| | - Maria Cabrera
- Université François Rabelais, UMR 1100, F-37032 Tours, France; INSERM, Centre d'Etude des Pathologies Respiratoires, UMR 1100, F-37032 Tours, France
| | - Joël Guillemain
- SESAME, Expertise en toxicologie, Chambray-les-tours, France
| | | | | | | | | | | | - Laurent Guilleminault
- Université François Rabelais, UMR 1100, F-37032 Tours, France; INSERM, Centre d'Etude des Pathologies Respiratoires, UMR 1100, F-37032 Tours, France
| | - Philippe Thullier
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines; Brétigny sur Orge, France
| | - Emmanuelle Guillot-Combe
- DGA, Direction de la Stratégie (DS), Mission pour la recherche et l'Innovation scientifique (MRIS), France
| | - Laurent Vecellio
- Université François Rabelais, UMR 1100, F-37032 Tours, France; INSERM, Centre d'Etude des Pathologies Respiratoires, UMR 1100, F-37032 Tours, France; Aerodrug, F-37032 Tours, France
| | - Nathalie Heuzé-Vourc'h
- Université François Rabelais, UMR 1100, F-37032 Tours, France; INSERM, Centre d'Etude des Pathologies Respiratoires, UMR 1100, F-37032 Tours, France.
| |
Collapse
|
26
|
Miethe S, Rasetti-Escargueil C, Avril A, Liu Y, Chahboun S, Korkeala H, Mazuet C, Popoff MR, Pelat T, Thullier P, Sesardic D, Hust M. Development of Human-Like scFv-Fc Neutralizing Botulinum Neurotoxin E. PLoS One 2015; 10:e0139905. [PMID: 26440796 PMCID: PMC4595074 DOI: 10.1371/journal.pone.0139905] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/17/2015] [Indexed: 11/18/2022] Open
Abstract
Background Botulinum neurotoxins (BoNTs) are considered to be the most toxic substances known on earth and are responsible for human botulism, a life-threatening disease characterized by flaccid muscle paralysis that occurs naturally by food-poisoning or colonization of the gastrointestinal tract by BoNT-producing clostridia. BoNTs have been classified as category A agent by the Centers of Disease Control and Prevention (CDC) and are listed among the six agents with the highest risk to be used as bioweapons. Neutralizing antibodies are required for the development of effective anti-botulism therapies to deal with the potential risk of exposure. Results In this study, a macaque (Macaca fascicularis) was immunized with recombinant light chain of BoNT/E3 and an immune phage display library was constructed. After a multi-step panning, several antibody fragments (scFv, single chain fragment variable) with nanomolar affinities were isolated, that inhibited the endopeptidase activity of pure BoNT/E3 in vitro by targeting its light chain. Furthermore, three scFv were confirmed to neutralize BoNT/E3 induced paralysis in an ex vivo mouse phrenic nerve-hemidiaphragm assay. The most effective neutralization (20LD50/mL, BoNT/E3) was observed with scFv ELC18, with a minimum neutralizing concentration at 0.3 nM. Furthermore, ELC18 was highly effective in vivo when administered as an scFv-Fc construct. Complete protection of 1LD50 BoNT/E3 was observed with 1.6 ng/dose in the mouse flaccid paralysis assay. Conclusion These scFv-Fcs antibodies are the first recombinant antibodies neutralizing BoNT/E by targeting its light chain. The human-like nature of the isolated antibodies is predicting a good tolerance for further clinical development.
Collapse
Affiliation(s)
- Sebastian Miethe
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Spielmannstr. 7, 38106 Braunschweig, Germany
| | - Christine Rasetti-Escargueil
- Division of Bacteriology, National Institute for Biological Standards and Control (NIBSC), a centre of Medicines and Healthcare products Regulatory Agency, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, United Kingdom
| | - Arnaud Avril
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA), Département de Microbiologie, Unité de biotechnologie des anticorps et des toxines, 24 avenue des Maquis du Grésivaudan, B.P. 87, 38702 La Tronche Cedex, France
| | - Yvonne Liu
- Division of Bacteriology, National Institute for Biological Standards and Control (NIBSC), a centre of Medicines and Healthcare products Regulatory Agency, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, United Kingdom
| | - Siham Chahboun
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA), Département de Microbiologie, Unité de biotechnologie des anticorps et des toxines, 24 avenue des Maquis du Grésivaudan, B.P. 87, 38702 La Tronche Cedex, France
| | - Hannu Korkeala
- University of Helsinki, Faculty of Veterinary Medicine, Centre of Excellence in Microbial Food Safety Research, Department of Food Hygiene and Environmental Health, P.O. Box 66 (Agnes Sjöbergin katu 2), 00014 Helsinki University, Helsinki, Finland
| | - Christelle Mazuet
- Unité des Bactéries anaérobies et Toxines, Institut Pasteur, 25 avenue du Docteur Roux, 75015, Paris, France
| | - Michel-Robert Popoff
- Unité des Bactéries anaérobies et Toxines, Institut Pasteur, 25 avenue du Docteur Roux, 75015, Paris, France
| | - Thibaut Pelat
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA), Département de Microbiologie, Unité de biotechnologie des anticorps et des toxines, 24 avenue des Maquis du Grésivaudan, B.P. 87, 38702 La Tronche Cedex, France
| | - Philippe Thullier
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA), Département de Microbiologie, Unité de biotechnologie des anticorps et des toxines, 24 avenue des Maquis du Grésivaudan, B.P. 87, 38702 La Tronche Cedex, France
| | - Dorothea Sesardic
- Division of Bacteriology, National Institute for Biological Standards and Control (NIBSC), a centre of Medicines and Healthcare products Regulatory Agency, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, United Kingdom
| | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Spielmannstr. 7, 38106 Braunschweig, Germany
- * E-mail:
| |
Collapse
|
27
|
Rasetti-Escargueil C, Avril A, Chahboun S, Tierney R, Bak N, Miethe S, Mazuet C, Popoff MR, Thullier P, Hust M, Pelat T, Sesardic D. Development of human-like scFv-Fc antibodies neutralizing Botulinum toxin serotype B. MAbs 2015; 7:1161-77. [PMID: 26381852 PMCID: PMC4966489 DOI: 10.1080/19420862.2015.1082016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are responsible for human botulism, a life-threatening disease characterized by flaccid muscle paralysis that occurs naturally by food poisoning or colonization of the gastrointestinal tract by BoNT-producing clostridia. BoNTs have been classified as category A agents by the Centers for Disease Control and Prevention. To date, 7 subtypes of BoNT/B were identified showing that subtypes B1 (16 strains) and B2 (32 strains) constitute the vast majority of BoNT/B strains. Neutralizing antibodies are required for the development of anti-botulism drugs to deal with the potential risk. In this study, macaques (Macaca fascicularis) were immunized with recombinant light chain (LC) or heavy chain (HC) of BoNT/B2, followed by the construction of 2 hyper-immune phage display libraries. The best single-chain variable fragments (scFvs) isolated from each library were selected according to their affinities and cross reactivity with BoNT/B1 toxin subtype. These scFvs against LC and HC were further analyzed by assessing the inhibition of in vitro endopeptidase activity of BoNT/B1 and B2 and neutralization of BoNT/B1 and B2 toxin-induced paralysis in the mouse ex vivo phrenic nerve assay. The antibodies B2–7 (against HC) and BLC3 (against LC) were produced as scFv-Fc, and, when tested individually, neutralized BoNT/B1 and BoNT/B2 in a mouse ex vivo phrenic nerve assay. Whereas only scFv-Fc BLC3 alone protected mice against BoNT/B2-induced paralysis in vivo, when B2–7 and BLC3 were combined they exhibited potent synergistic protection. The present study provided an opportunity to assess the extent of antibody-mediated neutralization of BoNT/B1 and BoNT/B2 subtypes in ex vivo and in vitro assays, and to confirm the benefit of the synergistic effect of antibodies targeting the 2 distinct functional domains of the toxin in vivo. Notably, the framework regions of the most promising antibodies (B2–7 and BLC3) are close to the human germline sequences, which suggest that they may be well tolerated in potential clinical development.
Collapse
Affiliation(s)
- Christine Rasetti-Escargueil
- a Division of Bacteriology; National Institute for Biological Standards and Control (NIBSC), a centre of Medicines and Healthcare products Regulatory Agency ; Hertfordshire UK
| | - Arnaud Avril
- b Département des Maladies Infectieuses ; Unité Interaction Hôte-Pathogène; Institut de Recherche Biomédicale des Armées (IRBA) ; Brétigny-sur-Orge , France
| | - Siham Chahboun
- b Département des Maladies Infectieuses ; Unité Interaction Hôte-Pathogène; Institut de Recherche Biomédicale des Armées (IRBA) ; Brétigny-sur-Orge , France
| | - Rob Tierney
- a Division of Bacteriology; National Institute for Biological Standards and Control (NIBSC), a centre of Medicines and Healthcare products Regulatory Agency ; Hertfordshire UK
| | - Nicola Bak
- a Division of Bacteriology; National Institute for Biological Standards and Control (NIBSC), a centre of Medicines and Healthcare products Regulatory Agency ; Hertfordshire UK
| | - Sebastian Miethe
- c Institut für Biochemie, Biotechnologie und Bioinformatik; Technische Universität Braunschweig ; Braunschweig , Germany
| | - Christelle Mazuet
- d Unité des Bactéries anaérobies et Toxines; Institut Pasteur ; Paris , France
| | - Michel R Popoff
- d Unité des Bactéries anaérobies et Toxines; Institut Pasteur ; Paris , France
| | - Philippe Thullier
- b Département des Maladies Infectieuses ; Unité Interaction Hôte-Pathogène; Institut de Recherche Biomédicale des Armées (IRBA) ; Brétigny-sur-Orge , France
| | - Michael Hust
- c Institut für Biochemie, Biotechnologie und Bioinformatik; Technische Universität Braunschweig ; Braunschweig , Germany
| | - Thibaut Pelat
- b Département des Maladies Infectieuses ; Unité Interaction Hôte-Pathogène; Institut de Recherche Biomédicale des Armées (IRBA) ; Brétigny-sur-Orge , France
| | - Dorothea Sesardic
- a Division of Bacteriology; National Institute for Biological Standards and Control (NIBSC), a centre of Medicines and Healthcare products Regulatory Agency ; Hertfordshire UK
| |
Collapse
|
28
|
Isolation of nanomolar scFvs of non-human primate origin, cross-neutralizing botulinum neurotoxins A1 and A2 by targeting their heavy chain. BMC Biotechnol 2015; 15:86. [PMID: 26382731 PMCID: PMC4574468 DOI: 10.1186/s12896-015-0206-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 09/11/2015] [Indexed: 12/14/2022] Open
Abstract
Background Botulism is a naturally occurring disease, mainly caused by the ingestion of food contaminated by the botulinum neurotoxins (BoNTs). Botulinum neurotoxins are the most lethal. They are classified among the six major biological warfare agents by the Centers for Disease Control. BoNTs act on the cholinergic motoneurons, where they cleave proteins implicated in acetylcholine vesicle exocytosis. This exocytosis inhibition induces a flaccid paralysis progressively affecting all the muscles and generally engendering a respiratory distress. BoNTs are also utilized in medicine, mainly for the treatment of neuromuscular disorders, preventing large scale vaccination. Botulism specific treatment requires injections of antitoxins, usually of equine origin and thus poorly tolerated. Therefore, development of human or human-like neutralizing antibodies is of a major interest, and it is the subject of the European framework project called “AntiBotABE”. Results In this study, starting from a macaque immunized with the recombinant heavy chain of BoNT/A1 (BoNT/A1-HC), an immune antibody phage-display library was generated and antibody fragments (single chain Fragment variable) with nanomolar affinity were isolated and further characterized. The neutralization capacities of these scFvs were analyzed in the mouse phrenic nerve-hemidiaphragm assay. Conclusions After a three-round panning, 24 antibody fragments with affinity better than 10 nM were isolated. Three of them neutralized BoNT/A1 efficiently and two cross-neutralized BoNT/A1 and BoNT/A2 subtypes in the mouse phrenic nerve-hemidiaphragm assay. These are the first monoclonal human-like antibodies cross-neutralizing both BoNT/A1 and BoNT/A2. The antibody A1HC38 was selected for further development, and could be clinically developed for the prophylaxis and treatment of botulism. Electronic supplementary material The online version of this article (doi:10.1186/s12896-015-0206-0) contains supplementary material, which is available to authorized users.
Collapse
|
29
|
VNARs: An Ancient and Unique Repertoire of Molecules That Deliver Small, Soluble, Stable and High Affinity Binders of Proteins. Antibodies (Basel) 2015. [DOI: 10.3390/antib4030240] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
30
|
Ko BK, Choi S, Cui LG, Lee YH, Hwang IS, Kim KT, Shim H, Lee JS. Affinity Maturation of Monoclonal Antibody 1E11 by Targeted Randomization in CDR3 Regions Optimizes Therapeutic Antibody Targeting of HER2-Positive Gastric Cancer. PLoS One 2015. [PMID: 26225765 PMCID: PMC4520604 DOI: 10.1371/journal.pone.0134600] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Anti-HER2 murine monoclonal antibody 1E11 has strong and synergistic anti-tumor activity in HER2-overexpressing gastric cancer cells when used in combination with trastuzumab. We presently optimized this antibody for human therapeutics. First, the complementarity determining regions (CDRs) of the murine antibody were grafted onto human germline immunoglobulin variable genes. No difference in affinity and biological activity was observed between chimeric 1E11 (ch1E11) and humanized 1E11 (hz1E11). Next, affinity maturation of hz1E11 was performed by the randomization of CDR-L3 and H3 residues followed by stringent biopanning selection. Milder selection pressure favored the selection of more diverse clones, whereas higher selection stringency resulted in the convergence of the panning output to a smaller number of clones with improved affinity. Clone 1A12 had four amino acid substitutions in CDR-L3, and showed a 10-fold increase in affinity compared to the parental clone and increased potency in an in vitro anti-proliferative activity assay with HER2-overepxressing gastric cancer cells. Clone 1A12 inhibited tumor growth of NCI-N87 xenograft model with similar efficacy to trastuzumab alone, and the combination treatment of 1A12 and trastuzumab completely removed the established tumors. These results suggest that humanized and affinity matured monoclonal antibody 1A12 is a highly optimized molecule for future therapeutic development against HER2-positive tumors.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal, Humanized/chemistry
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibody Affinity
- Cell Line, Tumor
- Complementarity Determining Regions/immunology
- Female
- Genes, erbB-2
- Humans
- Mice
- Mice, Nude
- Molecular Sequence Data
- Sequence Homology, Amino Acid
- Stomach Neoplasms/genetics
- Stomach Neoplasms/therapy
Collapse
Affiliation(s)
- Bong-Kook Ko
- Therapeutic antibody research center, AbClon Inc., Seoul, Korea
| | - Soyoung Choi
- Therapeutic antibody research center, AbClon Inc., Seoul, Korea
| | - Lei Guang Cui
- Therapeutic antibody research center, AbClon Inc., Seoul, Korea
| | - Young-Ha Lee
- Therapeutic antibody research center, AbClon Inc., Seoul, Korea
| | - In-Sik Hwang
- Therapeutic antibody research center, AbClon Inc., Seoul, Korea
| | - Kyu-Tae Kim
- Therapeutic antibody research center, AbClon Inc., Seoul, Korea
- * E-mail: (HS); (KTK)
| | - Hyunbo Shim
- Departments of Bioinspired Science and Life Science, Ewha Womans University, Seoul, Korea
- * E-mail: (HS); (KTK)
| | - Jong-Seo Lee
- Therapeutic antibody research center, AbClon Inc., Seoul, Korea
| |
Collapse
|
31
|
Roh J, Byun SJ, Seo Y, KIm M, Lee JH, Kim S, Lee Y, Lee KW, Kim JK, Kwon MH. Generation of a chickenized catalytic anti-nucleic acid antibody by complementarity-determining region grafting. Mol Immunol 2015; 63:513-20. [PMID: 25458312 DOI: 10.1016/j.molimm.2014.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/07/2014] [Accepted: 10/11/2014] [Indexed: 12/24/2022]
Abstract
In contrast to a number of studies on the humanization of non-human antibodies, the reshaping of a non-human antibody into a chicken antibody has never been attempted. Therefore, nothing is known about the animal species-dependent compatibility of the framework regions (FRs) that sustain the appropriate conformation of the complementarity-determining regions (CDRs). In this study, we attempted the reshaping of the variable domains of the mouse catalytic anti-nucleic acid antibody 3D8 (m3D8) into the FRs of a chicken antibody (“chickenization”) by CDR grafting, which is a common method for the humanization of antibodies. CDRs of the acceptor chicken antibody that showed a high homology to the FRs of m3D8 were replaced with those of m3D8, resulting in the chickenized antibody (ck3D8). ck3D8 retained the biochemical properties (DNA binding, DNA hydrolysis, and cellular internalizing activities) and three-dimensional structure of m3D8 and showed reduced immunogenicity in chickens. Our study demonstrates that CDR grafting can be applied to the chickenization of a mouse antibody, probably due to the interspecies compatibility of the FRs.
Collapse
|
32
|
Chromikova V, Mader A, Hofbauer S, Göbl C, Madl T, Gach JS, Bauernfried S, Furtmüller PG, Forthal DN, Mach L, Obinger C, Kunert R. Introduction of germline residues improves the stability of anti-HIV mAb 2G12-IgM. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:1536-44. [PMID: 25748881 PMCID: PMC4582045 DOI: 10.1016/j.bbapap.2015.02.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/16/2015] [Accepted: 02/24/2015] [Indexed: 11/02/2022]
Abstract
Immunoglobulins M (IgMs) are gaining increasing attention as biopharmaceuticals since their multivalent mode of binding can give rise to high avidity. Furthermore, IgMs are potent activators of the complement system. However, they are frequently difficult to express recombinantly and can suffer from low conformational stability. Here, the broadly neutralizing anti-HIV-1 antibody 2G12 was class-switched to IgM and then further engineered by introduction of 17 germline residues. The impact of these changes on the structure and conformational stability of the antibody was then assessed using a range of biophysical techniques. We also investigated the effects of the class switch and germline substitutions on the ligand-binding properties of 2G12 and its capacity for HIV-1 neutralization. Our results demonstrate that the introduced germline residues improve the conformational and thermal stability of 2G12-IgM without altering its overall shape and ligand-binding properties. Interestingly, the engineered protein was found to exhibit much lower neutralization potency than its wild-type counterpart, indicating that potent antigen recognition is not solely responsible for IgM-mediated HIV-1 inactivation.
Collapse
Affiliation(s)
- Veronika Chromikova
- Department of Biotechnology, Vienna Institute of BioTechnology at BOKU, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Alexander Mader
- Department of Biotechnology, Vienna Institute of BioTechnology at BOKU, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Stefan Hofbauer
- Department of Chemistry, Division of Biochemistry, Vienna Institute of BioTechnology at BOKU, University of Natural Resources and Life Sciences, Vienna, Austria; Department for Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, Austria
| | - Christoph Göbl
- Center for Integrated Protein Science Munich at Chair of Biomolecular NMR Spectroscopy, Department of Chemistry, Technical University Munich, Garching, Germany; Institute of Structural Biology, Helmholtz Center Munich, Neuherberg, Germany
| | - Tobias Madl
- Center for Integrated Protein Science Munich at Chair of Biomolecular NMR Spectroscopy, Department of Chemistry, Technical University Munich, Garching, Germany; Institute of Structural Biology, Helmholtz Center Munich, Neuherberg, Germany; Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Austria
| | - Johannes S Gach
- Department of Medicine, Division of Infectious Diseases, University of CA, Irvine, USA
| | - Stefan Bauernfried
- Department of Biotechnology, Vienna Institute of BioTechnology at BOKU, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Paul G Furtmüller
- Department of Chemistry, Division of Biochemistry, Vienna Institute of BioTechnology at BOKU, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Donald N Forthal
- Department of Medicine, Division of Infectious Diseases, University of CA, Irvine, USA
| | - Lukas Mach
- Department of Applied Genetics and Cell Biology, Vienna Institute of BioTechnology at BOKU, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Christian Obinger
- Department of Chemistry, Division of Biochemistry, Vienna Institute of BioTechnology at BOKU, University of Natural Resources and Life Sciences, Vienna, Austria.
| | - Renate Kunert
- Department of Biotechnology, Vienna Institute of BioTechnology at BOKU, University of Natural Resources and Life Sciences, Vienna, Austria.
| |
Collapse
|
33
|
Lefranc MP. Immunoglobulins: 25 years of immunoinformatics and IMGT-ONTOLOGY. Biomolecules 2014; 4:1102-39. [PMID: 25521638 PMCID: PMC4279172 DOI: 10.3390/biom4041102] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 12/02/2014] [Accepted: 12/03/2014] [Indexed: 11/17/2022] Open
Abstract
IMGT®, the international ImMunoGeneTics information system® (CNRS and Montpellier University) is the global reference in immunogenetics and immunoinformatics. By its creation in 1989, IMGT® marked the advent of immunoinformatics, which emerged at the interface between immunogenetics and bioinformatics. IMGT® is specialized in the immunoglobulins (IG) or antibodies, T cell receptors (TR), major histocompatibility (MH), and IgSF and MhSF superfamilies. IMGT® has been built on the IMGT-ONTOLOGY axioms and concepts, which bridged the gap between genes, sequences and three-dimensional (3D) structures. The concepts include the IMGT® standardized keywords (identification), IMGT® standardized labels (description), IMGT® standardized nomenclature (classification), IMGT unique numbering and IMGT Colliers de Perles (numerotation). IMGT® comprises seven databases, 15,000 pages of web resources and 17 tools. IMGT® tools and databases provide a high-quality analysis of the IG from fish to humans, for basic, veterinary and medical research, and for antibody engineering and humanization. They include, as examples: IMGT/V-QUEST and IMGT/JunctionAnalysis for nucleotide sequence analysis and their high-throughput version IMGT/HighV-QUEST for next generation sequencing, IMGT/DomainGapAlign for amino acid sequence analysis of IG domains, IMGT/3Dstructure-DB for 3D structures, contact analysis and paratope/epitope interactions of IG/antigen complexes, and the IMGT/mAb-DB interface for therapeutic antibodies and fusion proteins for immunological applications (FPIA).
Collapse
Affiliation(s)
- Marie-Paule Lefranc
- IMGT®, the international ImMunoGenetics information system®, Laboratoire d'ImmunoGénétique Moléculaire LIGM, Institut de Génétique Humaine IGH, UPR CNRS 1142, Montpellier University, 141 rue de la Cardonille, 34396 Montpellier cedex 5, France.
| |
Collapse
|
34
|
Ahmadzadeh V, Farajnia S, Feizi MAH, Nejad RAK. Antibody humanization methods for development of therapeutic applications. Monoclon Antib Immunodiagn Immunother 2014; 33:67-73. [PMID: 24746146 DOI: 10.1089/mab.2013.0080] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Recombinant antibody technologies are rapidly becoming available and showing considerable clinical success. However, the immunogenicity of murine-derived monoclonal antibodies is restrictive in cancer immunotherapy. Humanized antibodies can overcome these problems and are considered to be a promising alternative therapeutic agent. There are several approaches for antibody humanization. In this article we review various methods used in the antibody humanization process.
Collapse
Affiliation(s)
- Vahideh Ahmadzadeh
- 1 Biotechnology Research Center, Tabriz University of Medical Sciences , Tabriz, Iran
| | | | | | | |
Collapse
|
35
|
Design, expression and characterization of a single chain anti-CD20 antibody; a germline humanized antibody derived from Rituximab. Protein Expr Purif 2014; 102:45-51. [PMID: 25088934 DOI: 10.1016/j.pep.2014.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 07/20/2014] [Accepted: 07/22/2014] [Indexed: 12/31/2022]
Abstract
CD20 is a B cell lineage specific surface antigen involved in various B cell malignancies. So far, several murine and chimeric antibodies have been produced against this antigen among which Rituximab is a commercially approved antibody widely used in treatment of cancers associated with CD20 overexpression. The current study reports the production and characterization of a humanized single chain version of Rituximab through CDR grafting method. For either heavy or light chain variable domains, a human antibody with the highest sequence homology to Rituximab was selected from human germline sequences and used as framework donors. Vernier zone residues in framework regions were replaced with those of Rituximab to retain the antigen binding affinity of parental antibody. The reactivity of humanized single chain antibody with CD20 was examined by ELISA and dot blot assays. The ability of antibody to suppress the growth of CD20 overexpressing Raji cells was tested by MTT assay. Analysis of reactivity with CD20 antigen revealed that the humanized single chain antibody reacted to the target antigen with high affinity. Proliferation inhibition assay showed that humanized scFv could suppress the proliferation of Raji cells efficiently in a dose-dependent manner. This successful production of a humanized scFv with the ability to inhibit growth of CD20-expressing cancer cell may provide a promising alternative strategy for CD20 targeted therapy.
Collapse
|
36
|
Safdari Y, Farajnia S, Asgharzadeh M, Khosroshahi SA, Veisi K, Ahmadzadeh V, Khalili M. Protein L: a robust enzyme-conjugated molecule for detection of humanized single chain antibodies. Monoclon Antib Immunodiagn Immunother 2014; 32:409-12. [PMID: 24328745 DOI: 10.1089/mab.2013.0049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The current study was aimed at introducing an efficient enzyme-conjugated molecule for use in the detection of humanized single chain antibodies. Because they are composite in their variable domain sequences and also devoid of constant domains, humanized single chain antibodies require a suitable secondary enzyme-conjugated antibody (more precisely enzyme-conjugated protein molecule) to be efficiently recognized and detected in ELISA, dot blot, and other detection tests, guaranteeing a more precise evaluation of their quantity, affinity, and other features. In the current study we examined the ability of three HRP-conjugated protein molecules including protein L, anti-human IgG antibody, and anti-mouse IgG antibody in the detection of three humanized single chain antibodies (anti-CD20, anti-EGFRvIII, and anti-EGFR) in ELISA and dot blot tests. The results signify the HRP-conjugated protein L as the most efficient molecule for detecting humanized single chain antibody.
Collapse
Affiliation(s)
- Yaghoub Safdari
- 1 Biotechnology Research Center, Tabriz University of Medical Sciences , Tabriz, Iran
| | | | | | | | | | | | | |
Collapse
|
37
|
Hülseweh B, Rülker T, Pelat T, Langermann C, Frenzel A, Schirrmann T, Dübel S, Thullier P, Hust M. Human-like antibodies neutralizing Western equine encephalitis virus. MAbs 2014; 6:718-27. [PMID: 24518197 PMCID: PMC4011916 DOI: 10.4161/mabs.28170] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
This study describes the development of the first neutralizing antibodies against Western equine encephalitis virus (WEEV), a member of the genus Alphavirus. WEEV is transmitted by mosquitoes and can spread to the human central nervous system, causing symptoms ranging from mild febrile reactions to life-threatening encephalitis. WEEV has been classified as a biological warfare agent by the US Centers for Disease Control and Prevention. No anti-WEEV drugs are currently commercially available. Neutralizing antibodies are useful for the pre- and post-exposure treatment of WEEV infections. In this study, two immune antibody gene libraries were constructed from two macaques immunized with inactivated WEEV. Four antibodies were selected from these libraries and recloned as scFv-Fc, with a human Fc part. These antibodies bound WEEV specifically in ELISA with little or no cross-reaction with other alphaviruses. They were further analyzed by immunohistochemistry. All binders were suitable for the intracellular detection of WEEV particles. Neutralizing activity was determined in vitro. Three of the four antibodies were found to be neutralizing; about 1 ng/mL of the best antibody (ToR69–3A2) neutralized 50% of 5x104 TCID50/mL. Due to its human-like nature with a germinality index of 89% (VH) and 91% (VL), the ToR69–3A2 antibody is a promising candidate for future passive vaccine development.
Collapse
Affiliation(s)
- Birgit Hülseweh
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS); ABC-Schutz; Munster, Germany
| | - Torsten Rülker
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie und Bioinformatik; Braunschweig, Germany
| | - Thibaut Pelat
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines; La Tronche, France
| | - Claudia Langermann
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS); ABC-Schutz; Munster, Germany
| | - Andrè Frenzel
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie und Bioinformatik; Braunschweig, Germany
| | - Thomas Schirrmann
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie und Bioinformatik; Braunschweig, Germany
| | - Stefan Dübel
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie und Bioinformatik; Braunschweig, Germany
| | - Philippe Thullier
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines; La Tronche, France
| | - Michael Hust
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie und Bioinformatik; Braunschweig, Germany
| |
Collapse
|
38
|
Lefranc MP. Immunoglobulin and T Cell Receptor Genes: IMGT(®) and the Birth and Rise of Immunoinformatics. Front Immunol 2014; 5:22. [PMID: 24600447 PMCID: PMC3913909 DOI: 10.3389/fimmu.2014.00022] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 01/15/2014] [Indexed: 11/13/2022] Open
Abstract
IMGT(®), the international ImMunoGeneTics information system(®) (1), (CNRS and Université Montpellier 2) is the global reference in immunogenetics and immunoinformatics. By its creation in 1989, IMGT(®) marked the advent of immunoinformatics, which emerged at the interface between immunogenetics and bioinformatics. IMGT(®) is specialized in the immunoglobulins (IG) or antibodies, T cell receptors (TR), major histocompatibility (MH), and proteins of the IgSF and MhSF superfamilies. IMGT(®) has been built on the IMGT-ONTOLOGY axioms and concepts, which bridged the gap between genes, sequences, and three-dimensional (3D) structures. The concepts include the IMGT(®) standardized keywords (concepts of identification), IMGT(®) standardized labels (concepts of description), IMGT(®) standardized nomenclature (concepts of classification), IMGT unique numbering, and IMGT Colliers de Perles (concepts of numerotation). IMGT(®) comprises seven databases, 15,000 pages of web resources, and 17 tools, and provides a high-quality and integrated system for the analysis of the genomic and expressed IG and TR repertoire of the adaptive immune responses. Tools and databases are used in basic, veterinary, and medical research, in clinical applications (mutation analysis in leukemia and lymphoma) and in antibody engineering and humanization. They include, for example IMGT/V-QUEST and IMGT/JunctionAnalysis for nucleotide sequence analysis and their high-throughput version IMGT/HighV-QUEST for next-generation sequencing (500,000 sequences per batch), IMGT/DomainGapAlign for amino acid sequence analysis of IG and TR variable and constant domains and of MH groove domains, IMGT/3Dstructure-DB for 3D structures, contact analysis and paratope/epitope interactions of IG/antigen and TR/peptide-MH complexes and IMGT/mAb-DB interface for therapeutic antibodies and fusion proteins for immune applications (FPIA).
Collapse
Affiliation(s)
- Marie-Paule Lefranc
- The International ImMunoGenetics Information System (IMGT), Laboratoire d’ImmunoGénétique Moléculaire (LIGM), Institut de Génétique Humaine, UPR CNRS, Université Montpellier 2, Montpellier, France
| |
Collapse
|
39
|
Miethe S, Rasetti-Escargueil C, Liu Y, Chahboun S, Pelat T, Avril A, Frenzel A, Schirrmann T, Thullier P, Sesardic D, Hust M. Development of neutralizing scFv-Fc against botulinum neurotoxin A light chain from a macaque immune library. MAbs 2014; 6:446-59. [PMID: 24492304 PMCID: PMC3984333 DOI: 10.4161/mabs.27773] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Botulinum toxins (BoNTs) are among the most toxic substances on earth, with serotype A toxin being the most toxic substance known. They are responsible for human botulism, a disease characterized by flaccid muscle paralysis that occurs naturally through food poisoning or the colonization of the gastrointestinal tract by BoNT-producing clostridia. BoNT has been classified as a category A agent by the Centers for Disease Control, and it is one of six agents with the highest potential risk of use as bioweapons. Human or human-like neutralizing antibodies are thus required for the development of anti-botulinum toxin drugs to deal with this possibility. In this study, Macaca fascicularis was hyperimmunized with a recombinant light chain of BoNT/A. An immune phage display library was constructed and, after multistep panning, several scFv with nanomolar affinities that inhibited the endopeptidase activity of BoNT/A1 in vitro as scFv-Fc, with a molar ratio (ab binding site:toxin) of up to 1:1, were isolated. The neutralization of BoNT/A-induced paralysis by the SEM120-IID5, SEM120-IIIC1 and SEM120-IIIC4 antibodies was demonstrated in mouse phrenic nerve-hemidiaphragm preparations with the holotoxin. The neutralization observed is the strongest ever measured in the phrenic nerve-hemidiaphragm assay for BoNT/A1 for a monoclonal antibody. Several scFv-Fc inhibiting the endopeptidase activity of botulinum neurotoxin A were isolated. For SEM120-IID5, SEM120-IIIC1, and SEM120-IIIC4, inhibitory effects in vitro and protection against the toxin ex vivo were observed. The human-like nature of these antibodies makes them promising lead candidates for further development of immunotherapeutics for this disease.
Collapse
Affiliation(s)
- Sebastian Miethe
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie, und Bioinformatik; Abteilung Biotechnologie; Braunschweig, Germany
| | - Christine Rasetti-Escargueil
- National Institute for Biological Standards and Control (NIBSC); Medicines and Healthcare Products Regulatory Agency; Division of Bacteriology; Potters Bar, UK
| | - Yvonne Liu
- National Institute for Biological Standards and Control (NIBSC); Medicines and Healthcare Products Regulatory Agency; Division of Bacteriology; Potters Bar, UK
| | - Siham Chahboun
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines ; La Tronche Cedex, France
| | - Thibaut Pelat
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines ; La Tronche Cedex, France
| | - Arnaud Avril
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines ; La Tronche Cedex, France
| | - André Frenzel
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie, und Bioinformatik; Abteilung Biotechnologie; Braunschweig, Germany
| | - Thomas Schirrmann
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie, und Bioinformatik; Abteilung Biotechnologie; Braunschweig, Germany
| | - Philippe Thullier
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines ; La Tronche Cedex, France
| | - Dorothea Sesardic
- National Institute for Biological Standards and Control (NIBSC); Medicines and Healthcare Products Regulatory Agency; Division of Bacteriology; Potters Bar, UK
| | - Michael Hust
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie, und Bioinformatik; Abteilung Biotechnologie; Braunschweig, Germany
| |
Collapse
|
40
|
Kuramochi T, Igawa T, Tsunoda H, Hattori K. Humanization and simultaneous optimization of monoclonal antibody. Methods Mol Biol 2014; 1060:123-137. [PMID: 24037839 DOI: 10.1007/978-1-62703-586-6_7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Antibody humanization is an essential technology for reducing the potential risk of immunogenicity associated with animal-derived antibodies and has been applied to a majority of the therapeutic antibodies on the market. For developing an antibody molecule as a pharmaceutical at the current biotechnology level, however, other properties also have to be considered in parallel with humanization in antibody generation and optimization. This section describes the critical properties of therapeutic antibodies that should be sufficiently qualified, including immunogenicity, binding affinity, physiochemical stability, expression in host cells and pharmacokinetics, and the basic methodologies of antibody engineering involved. By simultaneously optimizing the antibody molecule in the light of these properties, it should prove possible to shorten the research and development period necessary to identify a highly qualified clinical candidate and consequently accelerate the start of the clinical trial.
Collapse
Affiliation(s)
- T Kuramochi
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | | | | | | |
Collapse
|
41
|
humMR1, a highly specific humanized single chain antibody for targeting EGFRvIII. Int Immunopharmacol 2013; 18:304-10. [PMID: 24370392 DOI: 10.1016/j.intimp.2013.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 11/30/2013] [Accepted: 12/11/2013] [Indexed: 11/23/2022]
Abstract
Production of an efficient humanized single chain antibody is reported here to specifically target EGFRvIII, a truncated receptor expressed in a wide variety of human cancers. CDR loops of MR1, a phage display-derived murine single chain antibody developed against this mutant receptor, were grafted on human frameworks that had been selected based on similarity to MR1 in terms of two distinct parameters, variable domain protein sequence and CDR canonical structures. Moreover, two point mutations were introduced in CDR-H2 and CDR-H3 loops of the humanized antibody to destroy its cross-reactivity to wild-type EGFR. The resultant antibody, referred to as humMR1, was found by MTT assay, ELISA and western blot techniques to be highly specific for EGFRvIII. The affinity of this antibody for EGFRvIII-specific 14-amino acid synthetic peptide and HC2 cells were measured to be 1.87 × 10(10) and 2.17 × 10(10)/M respectively. This humanized antibody leads to 78.5% inhibition in proliferation of EGFRvIII-overexpressing cells.
Collapse
|
42
|
Safdari Y, Farajnia S, Asgharzadeh M, Khalili M. Antibody humanization methods – a review and update. Biotechnol Genet Eng Rev 2013; 29:175-86. [DOI: 10.1080/02648725.2013.801235] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
43
|
Gao SH, Huang K, Tu H, Adler AS. Monoclonal antibody humanness score and its applications. BMC Biotechnol 2013; 13:55. [PMID: 23826749 PMCID: PMC3729710 DOI: 10.1186/1472-6750-13-55] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 06/20/2013] [Indexed: 01/27/2023] Open
Abstract
Background Monoclonal antibody therapeutics are rapidly gaining in popularity for the treatment of a myriad of diseases, ranging from cancer to autoimmune diseases and neurological diseases. Multiple forms of antibody therapeutics are in use today that differ in the amount of human sequence present in both the constant and variable regions, where antibodies that are more human-like usually have reduced immunogenicity in clinical trials. Results Here we present a method to quantify the humanness of the variable region of monoclonal antibodies and show that this method is able to clearly distinguish human and non-human antibodies with excellent specificity. After creating and analyzing a database of human antibody sequences, we conducted an in-depth analysis of the humanness of therapeutic antibodies, and found that increased humanness score is correlated with decreased immunogenicity of antibodies. We further discovered a surprisingly similarity in the immunogenicity of fully human antibodies and humanized antibodies that are more human-like based on their humanness score. Conclusions Our results reveal that in most cases humanizing an antibody and confirming the humanness of the final form may be sufficient to eliminate immunogenicity issues to the same extent as using fully human antibodies. We created a public website to calculate the humanness score of any input antibody sequence based on our human antibody database. This tool will be of great value during the preclinical drug development process for new monoclonal antibody therapeutics.
Collapse
Affiliation(s)
- Sean H Gao
- LakePharma, Inc,, 530 Harbor Boulevard, Belmont, CA 94002, USA
| | | | | | | |
Collapse
|
44
|
Meng W, Pan W, Zhang AJX, Li Z, Wei G, Feng L, Dong Z, Li C, Hu X, Sun C, Luo Q, Yuen KY, Zhong N, Chen L. Rapid Generation of Human-Like Neutralizing Monoclonal Antibodies in Urgent Preparedness for Influenza Pandemics and Virulent Infectious Diseases. PLoS One 2013; 8:e66276. [PMID: 23824680 PMCID: PMC3688872 DOI: 10.1371/journal.pone.0066276] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Accepted: 05/03/2013] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The outbreaks of emerging infectious diseases caused by pathogens such as SARS coronavirus, H5N1, H1N1, and recently H7N9 influenza viruses, have been associated with significant mortality and morbidity in humans. Neutralizing antibodies from individuals who have recovered from an infection confer therapeutic protection to others infected with the same pathogen. However, survivors may not always be available for providing plasma or for the cloning of monoclonal antibodies (mAbs). METHODOLOGY/PRINCIPAL FINDINGS The genome and the immunoglobulin genes in rhesus macaques and humans are highly homologous; therefore, we investigated whether neutralizing mAbs that are highly homologous to those of humans (human-like) could be generated. Using the H5N1 influenza virus as a model, we first immunized rhesus macaques with recombinant adenoviruses carrying a synthetic gene encoding hemagglutinin (HA). Following screening an antibody phage display library derived from the B cells of immunized monkeys, we cloned selected macaque immunoglobulin heavy chain and light chain variable regions into the human IgG constant region, which generated human-macaque chimeric mAbs exhibiting over 97% homology to human antibodies. Selected mAbs demonstrated potent neutralizing activities against three clades (0, 1, 2) of the H5N1 influenza viruses. The in vivo protection experiments demonstrated that the mAbs effectively protected the mice even when administered up to 3 days after infection with H5N1 influenza virus. In particular, mAb 4E6 demonstrated sub-picomolar binding affinity to HA and superior in vivo protection efficacy without the loss of body weight and obvious lung damage. The analysis of the 4E6 escape mutants demonstrated that the 4E6 antibody bound to a conserved epitope region containing two amino acids on the globular head of HA. CONCLUSIONS/SIGNIFICANCE Our study demonstrated the generation of neutralizing mAbs for potential application in humans in urgent preparedness against outbreaks of new influenza infections or other virulent infectious diseases.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/immunology
- Antibodies, Neutralizing/biosynthesis
- Antibodies, Neutralizing/immunology
- Communicable Diseases, Emerging/epidemiology
- Communicable Diseases, Emerging/immunology
- Communicable Diseases, Emerging/therapy
- Disease Outbreaks
- Humans
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza, Human/epidemiology
- Influenza, Human/immunology
- Influenza, Human/therapy
- Macaca mulatta
- Mice
Collapse
Affiliation(s)
- Weixu Meng
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Weiqi Pan
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Anna J. X. Zhang
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | - Zhengfeng Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guowei Wei
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Liqiang Feng
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhenyuan Dong
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chufang Li
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiangjing Hu
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Caijun Sun
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qinfang Luo
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kwok-Yung Yuen
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | - Nanshan Zhong
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
45
|
Mapping the epitopes of a neutralizing antibody fragment directed against the lethal factor of Bacillus anthracis and cross-reacting with the homologous edema factor. PLoS One 2013; 8:e65855. [PMID: 23741517 PMCID: PMC3669279 DOI: 10.1371/journal.pone.0065855] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 05/03/2013] [Indexed: 11/19/2022] Open
Abstract
The lethal toxin (LT) of Bacillus anthracis, composed of the protective antigen (PA) and the lethal factor (LF), plays an essential role in anthrax pathogenesis. PA also interacts with the edema factor (EF, 20% identity with LF) to form the edema toxin (ET), which has a lesser role in anthrax pathogenesis. The first recombinant antibody fragment directed against LF was scFv 2LF; it neutralizes LT by blocking the interaction between PA and LF. Here, we report that scFv 2LF cross-reacts with EF and cross-neutralizes ET, and we present an in silico method taking advantage of this cross-reactivity to map the epitope of scFv 2LF on both LF and EF. This method identified five epitope candidates on LF, constituted of a total of 32 residues, which were tested experimentally by mutating the residues to alanine. This combined approach precisely identified the epitope of scFv 2LF on LF as five residues (H229, R230, Q234, L235 and Y236), of which three were missed by the consensus epitope candidate identified by pre-existing in silico methods. The homolog of this epitope on EF (H253, R254, E258, L259 and Y260) was experimentally confirmed to constitute the epitope of scFv 2LF on EF. Other inhibitors, including synthetic molecules, could be used to target these epitopes for therapeutic purposes. The in silico method presented here may be of more general interest.
Collapse
|
46
|
Use of IMGT(®) databases and tools for antibody engineering and humanization. Methods Mol Biol 2012; 907:3-37. [PMID: 22907343 DOI: 10.1007/978-1-61779-974-7_1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
IMGT(®), the international ImMunoGeneTics information system(®) (http://www.imgt.org), was created in 1989 to manage the huge diversity of the antigen receptors, immunoglobulins (IG) or antibodies, and T cell receptors (TR). Standardized sequence and structure analysis of antibody using IMGT(®) databases and tools allows one to bridge, for the first time, the gap between antibody sequences and three-dimensional (3D) structures. This is achieved through the IMGT Scientific chart rules, based on the IMGT-ONTOLOGY concepts of classification (IMGT gene and allele nomenclature), description (IMGT standardized labels), and numerotation (IMGT unique numbering and IMGT Colliers de Perles). IMGT(®) is the international reference for immunogenetics and immunoinformatics and its standards are particularly useful for antibody humanization and evaluation of immunogenicity. IMGT(®) databases for antibody nucleotide sequences and genes include IMGT/LIGM-DB and IMGT/GENE-DB, respectively, whereas nucleotide sequence analysis is performed by the IMGT/V-QUEST, IMGT/HighV-QUEST, and IMGT/JunctionAnalysis tools. In this chapter, we focus on IMGT(®) databases and tools for amino acid sequences, two-dimensional (2D) and three-dimensional (3D) structures: the IMGT/DomainGapAlign and IMGT/Collier-de-Perles tools, the IMGT/2Dstructure-DB database for amino acid sequences of monoclonal antibodies (mAb, suffix -mab) and fusion proteins for immune applications (FPIA, suffix -cept) of the World Health Organization/International Nonproprietary Name (WHO/INN) programme and other proteins of interest, and the IMGT/3Dstructure-DB database for crystallized antibodies and its associated tools (IMGT/StructuralQuery, IMGT/DomainSuperimpose).
Collapse
|
47
|
Rülker T, Voß L, Thullier P, O' Brien LM, Pelat T, Perkins SD, Langermann C, Schirrmann T, Dübel S, Marschall HJ, Hust M, Hülseweh B. Isolation and characterisation of a human-like antibody fragment (scFv) that inactivates VEEV in vitro and in vivo. PLoS One 2012; 7:e37242. [PMID: 22666347 PMCID: PMC3364240 DOI: 10.1371/journal.pone.0037242] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 04/18/2012] [Indexed: 11/18/2022] Open
Abstract
Venezuelan equine encephalitis virus (VEEV) belongs to the Alphavirus genus and several species of this family are pathogenic to humans. The viruses are classified as potential agents of biological warfare and terrorism and sensitive detection as well as effective prophylaxis and antiviral therapies are required.In this work, we describe the isolation of the anti-VEEV single chain Fragment variable (scFv), ToR67-3B4, from a non-human primate (NHP) antibody gene library. We report its recloning into the bivalent scFv-Fc format and further immunological and biochemical characterisation.The scFv-Fc ToR67-3B4 recognised viable as well as formalin and ß-propionolactone (ß-Pl) inactivated virus particles and could be applied for immunoblot analysis of VEEV proteins and immuno-histochemistry of VEEV infected cells. It detected specifically the viral E1 envelope protein of VEEV but did not react with reduced viral glycoprotein preparations suggesting that recognition depends upon conformational epitopes. The recombinant antibody was able to detect multiple VEEV subtypes and displayed only marginal cross-reactivity to other Alphavirus species except for EEEV. In addition, the scFv-Fc fusion described here might be of therapeutic use since it successfully inactivated VEEV in a murine disease model. When the recombinant antibody was administered 6 hours post challenge, 80% to 100% of mice survived lethal VEEV IA/B or IE infection. Forty to sixty percent of mice survived when scFv-Fc ToR67-3B4 was applied 6 hours post challenge with VEEV subtypes II and former IIIA. In combination with E2-neutralising antibodies the NHP antibody isolated here could significantly improve passive protection as well as generic therapy of VEE.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/genetics
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/isolation & purification
- Antibodies, Viral/genetics
- Antibodies, Viral/immunology
- Antibodies, Viral/isolation & purification
- Cloning, Molecular
- Encephalitis Virus, Venezuelan Equine/immunology
- Gene Library
- Genetic Vectors/genetics
- Humans
- Immunization, Passive
- Macaca fascicularis
- Male
- Mice
- Mice, Inbred BALB C
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/isolation & purification
- Sequence Analysis
- Single-Chain Antibodies/genetics
- Single-Chain Antibodies/immunology
- Single-Chain Antibodies/isolation & purification
Collapse
Affiliation(s)
- Torsten Rülker
- Technische Universität Braunschweig, Institut für Biochemie und Biotechnologie, Braunschweig, Germany
| | - Luzie Voß
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS) – ABC-Schutz, Munster, Germany
| | - Philippe Thullier
- Centre de Recherche du Service de Santé des Armées (CRSSA-IRBA), La Tronche, France
| | - Lyn M. O' Brien
- Defence Science and Technology Laboratory, Biomedical Sciences Department, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - Thibaut Pelat
- Centre de Recherche du Service de Santé des Armées (CRSSA-IRBA), La Tronche, France
| | - Stuart D. Perkins
- Defence Science and Technology Laboratory, Biomedical Sciences Department, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - Claudia Langermann
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS) – ABC-Schutz, Munster, Germany
| | - Thomas Schirrmann
- Technische Universität Braunschweig, Institut für Biochemie und Biotechnologie, Braunschweig, Germany
| | - Stefan Dübel
- Technische Universität Braunschweig, Institut für Biochemie und Biotechnologie, Braunschweig, Germany
| | - Hans-Jürgen Marschall
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS) – ABC-Schutz, Munster, Germany
| | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie und Biotechnologie, Braunschweig, Germany
| | - Birgit Hülseweh
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS) – ABC-Schutz, Munster, Germany
| |
Collapse
|
48
|
Abstract
Antibodies make up the largest, growing segment of protein therapeutics in the pharmaceutical and biotechnology industries. The development or engineering of therapeutic antibodies is based to a large extent on our knowledge of antibody structure and requires sophisticated methods that continue to evolve. In this chapter, after a review of what is known about the structure and functional properties of antibodies, the current, state-of-the-art antibody engineering methods are described. These methods include antibody humanization, antigen-affinity optimization, Fc engineering for modulated effector function and extended half-life, and engineering for improved stability and biophysical properties. X-ray crystallographic structures of antibody fragments and their complexes can play a critical role in guiding and, in some cases, accelerating these processes. These approaches represent guidelines for developing antibody therapeutics with the desired affinity, effector function, and biophysical properties.
Collapse
|
49
|
Hust M, Frenzel A, Meyer T, Schirrmann T, Dübel S. Construction of human naive antibody gene libraries. Methods Mol Biol 2012; 907:85-107. [PMID: 22907347 DOI: 10.1007/978-1-61779-974-7_5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human antibodies are valuable tools for proteome research and diagnostics. Furthermore, antibodies are a rapidly growing class of therapeutic agents, mainly for inflammation and cancer therapy. The first therapeutic antibodies are of murine origin and were chimerized or humanized. The later-developed antibodies are fully human antibodies. Here, two technologies are competing the hybridoma technology using transgenic mice with human antibody gene loci and antibody phage display. The starting point for the selection of human antibodies against any target is the construction of an antibody phage display gene library.In this review we describe the construction of human naive and immune antibody gene libraries for antibody phage display.
Collapse
Affiliation(s)
- Michael Hust
- Institut für Biochemie und Biotechnologie, Techische Universität Braunschweig, Braunschweig, Germany.
| | | | | | | | | |
Collapse
|
50
|
Chahboun S, Hust M, Liu Y, Pelat T, Miethe S, Helmsing S, Jones RG, Sesardic D, Thullier P. Isolation of a nanomolar scFv inhibiting the endopeptidase activity of botulinum toxin A, by single-round panning of an immune phage-displayed library of macaque origin. BMC Biotechnol 2011; 11:113. [PMID: 22111995 PMCID: PMC3252318 DOI: 10.1186/1472-6750-11-113] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Accepted: 11/23/2011] [Indexed: 01/06/2023] Open
Abstract
Background Botulinum neurotoxin A (BoNT/A), mainly represented by subtype A1, is the most toxic substance known. It causes naturally-occurring food poisoning, and is among the biological agents at the highest risk of being weaponized. Several antibodies neutralizing BoNT/A by targeting its heavy chain (BoNT/A-H) have been isolated in the past. For the first time however, an IgG (4LCA) recently isolated by hybridoma technology and targeting the BoNT/A light chain (BoNT/A-L), was shown to inhibit BoNT/A endopeptidase activity and protect in vivo against BoNT/A. In the present study, a phage-displayed library was constructed from a macaque (Macaca fascicularis) hyper-immunized with BoNTA/L in order to isolate scFvs inhibiting BoNT/A endopeptidase activity for clinical use. Results Diversity of the scFvs constituting the library was limited due to the frequent presence, within the genes intended to be part of the library, of restriction sites utilized for its construction. After screening with several rounds of increasing stringency, as is usual with phage technology, the library got overwhelmed by phagemids encoding incomplete scFvs. The screening was successfully re-performed with a single round of high stringency. In particular, one of the isolated scFvs, 2H8, bound BoNT/A1 with a 3.3 nM affinity and effectively inhibited BoNT/A1 endopeptidase activity. The sequence encoding 2H8 was 88% identical to human germline genes and its average G-score was -0.72, quantifying the high human-like quality of 2H8. Conclusions The presence of restrictions sites within many of the sequences that were to be part of the library did not prevent the isolation of an scFv, 2H8, by an adapted panning strategy. ScFv 2H8 inhibited toxin endopeptidase activity in vitro and possessed human-like quality required for clinical development. More generally, the construction and screening of phage-displayed libraries built from hyper-immunized non-human primates is an efficient solution to isolate antibody fragments with therapeutic potential.
Collapse
Affiliation(s)
- Siham Chahboun
- Unité de Biotechnologie des Anticorps, et des Toxines, Département de Microbiologie, Institut de Recherche Biomédicale des Armées (IRBA-CRSSA), 24 Avenue des Maquis du Grésivaudan, BP 87, 38702 La Tronche Cedex, France.
| | | | | | | | | | | | | | | | | |
Collapse
|