1
|
Li J, Kang G, Wang J, Yuan H, Wu Y, Meng S, Wang P, Zhang M, Wang Y, Feng Y, Huang H, de Marco A. Affinity maturation of antibody fragments: A review encompassing the development from random approaches to computational rational optimization. Int J Biol Macromol 2023; 247:125733. [PMID: 37423452 DOI: 10.1016/j.ijbiomac.2023.125733] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
Routinely screened antibody fragments usually require further in vitro maturation to achieve the desired biophysical properties. Blind in vitro strategies can produce improved ligands by introducing random mutations into the original sequences and selecting the resulting clones under more and more stringent conditions. Rational approaches exploit an alternative perspective that aims first at identifying the specific residues potentially involved in the control of biophysical mechanisms, such as affinity or stability, and then to evaluate what mutations could improve those characteristics. The understanding of the antigen-antibody interactions is instrumental to develop this process the reliability of which, consequently, strongly depends on the quality and completeness of the structural information. Recently, methods based on deep learning approaches critically improved the speed and accuracy of model building and are promising tools for accelerating the docking step. Here, we review the features of the available bioinformatic instruments and analyze the reports illustrating the result obtained with their application to optimize antibody fragments, and nanobodies in particular. Finally, the emerging trends and open questions are summarized.
Collapse
Affiliation(s)
- Jiaqi Li
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Guangbo Kang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Jiewen Wang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Haibin Yuan
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Yili Wu
- Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and the Affiliated Kangning Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Oujiang Laboratory, Wenzhou, Zhejiang 325035, China
| | - Shuxian Meng
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Ping Wang
- New Technology R&D Department, Tianjin Modern Innovative TCM Technology Company Limited, Tianjin 300392, China
| | - Miao Zhang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; China Resources Biopharmaceutical Company Limited, Beijing 100029, China
| | - Yuli Wang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; Tianjin Pharmaceutical Da Ren Tang Group Corporation Limited, Traditional Chinese Pharmacy Research Institute, Tianjin Key Laboratory of Quality Control in Chinese Medicine, Tianjin 300457, China; State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin 300193, China
| | - Yuanhang Feng
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - He Huang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China.
| | - Ario de Marco
- Laboratory for Environmental and Life Sciences, University of Nova Gorica, Nova Gorica, Slovenia.
| |
Collapse
|
2
|
Hemadou A, Fontayne A, Laroche-Traineau J, Ottones F, Mondon P, Claverol S, Ducasse É, Sanchez S, Mohamad S, Lorenzato C, Duonor-Cerutti M, Clofent-Sanchez G, Jacobin-Valat MJ. In Vivo Human Single-Chain Fragment Variable Phage Display-Assisted Identification of Galectin-3 as a New Biomarker of Atherosclerosis. J Am Heart Assoc 2021; 10:e016287. [PMID: 34569248 PMCID: PMC8649142 DOI: 10.1161/jaha.120.016287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Atherosclerosis is a complex pathology in which dysfunctional endothelium, activated leucocytes, macrophages, and lipid‐laden foam cells are implicated, and in which plaque disruption is driven by many putative actors. This study aimed to identify accurate targetable biomarkers using new in vivo approaches to propose tools for improved diagnosis and treatment. Methods and Results Human scFv (single‐chain fragment variable) selected by in vivo phage display in a rabbit model of atherosclerosis was reformatted as scFv fused to the scFv‐Fc (single‐chain fragment variable fused to the crystallizable fragment of immunoglobulin G format) antibodies. Their reactivity was tested using flow cytometry and immunoassays, and aorta sections from animal models and human carotid and coronary artery specimens. A pool of atherosclerotic proteins from human endarterectomies was co‐immunoprecipitated with the selected scFv‐Fc followed by mass spectrometry for target identification. Near‐infrared fluorescence imaging was performed in Apoe−/− mice after injection of an Alexa Fluor 647–labeled scFv‐Fc‐2c antibody produced in a baculovirus system with 2 additional cysteine residues (ie, 2c) for future coupling to nano‐objects for theranostic applications. One scFv‐Fc clone (P3) displayed the highest cross‐reactivity against atherosclerotic lesion sections (rabbit, mouse, and human) and was chosen for translational development. Mass spectrometry identified galectin‐3, a β‐galactoside‐binding lectin, as the leader target. ELISA and immunofluorescence assays with a commercial anti‐galectin‐3 antibody confirmed this specificity. P3 scFv‐Fc‐2c specifically targeted atherosclerotic plaques in the Apoe−/− mouse model. Conclusions These results provide evidence that the P3 antibody holds great promise for molecular imaging of atherosclerosis and other inflammatory pathologies involving macrophages. Recently, galectin‐3 was proposed as a high‐value biomarker for the assessment of coronary and carotid atherosclerosis.
Collapse
Affiliation(s)
- Audrey Hemadou
- CRMSB (Centre de Resonance Magnétique des Systèmes Biologiques)UMR5536 CNRS (Centre National de Recherche Scientifique)INSB (Institut National des Sciences Biologiques) Bordeaux France
| | - Alexandre Fontayne
- LFB (Laboratoire Français de Fractionnement et de Biotechnologies) Biotechnologies Lille France.,BE4S (Bio-Experts for Success) Croix France
| | - Jeanny Laroche-Traineau
- CRMSB (Centre de Resonance Magnétique des Systèmes Biologiques)UMR5536 CNRS (Centre National de Recherche Scientifique)INSB (Institut National des Sciences Biologiques) Bordeaux France
| | - Florence Ottones
- CRMSB (Centre de Resonance Magnétique des Systèmes Biologiques)UMR5536 CNRS (Centre National de Recherche Scientifique)INSB (Institut National des Sciences Biologiques) Bordeaux France
| | - Philippe Mondon
- LFB (Laboratoire Français de Fractionnement et de Biotechnologies) Biotechnologies Lille France
| | - Stéphane Claverol
- Protéome Pole CGFB (Centre de Génomique Fonctionnelle de Bordeaux) Bordeaux France
| | | | - Stéphane Sanchez
- CRMSB (Centre de Resonance Magnétique des Systèmes Biologiques)UMR5536 CNRS (Centre National de Recherche Scientifique)INSB (Institut National des Sciences Biologiques) Bordeaux France
| | - Sarah Mohamad
- CRMSB (Centre de Resonance Magnétique des Systèmes Biologiques)UMR5536 CNRS (Centre National de Recherche Scientifique)INSB (Institut National des Sciences Biologiques) Bordeaux France
| | - Cyril Lorenzato
- CRMSB (Centre de Resonance Magnétique des Systèmes Biologiques)UMR5536 CNRS (Centre National de Recherche Scientifique)INSB (Institut National des Sciences Biologiques) Bordeaux France
| | | | - Gisèle Clofent-Sanchez
- CRMSB (Centre de Resonance Magnétique des Systèmes Biologiques)UMR5536 CNRS (Centre National de Recherche Scientifique)INSB (Institut National des Sciences Biologiques) Bordeaux France
| | - Marie-Josée Jacobin-Valat
- CRMSB (Centre de Resonance Magnétique des Systèmes Biologiques)UMR5536 CNRS (Centre National de Recherche Scientifique)INSB (Institut National des Sciences Biologiques) Bordeaux France
| |
Collapse
|
3
|
Lim CC, Choong YS, Lim TS. Cognizance of Molecular Methods for the Generation of Mutagenic Phage Display Antibody Libraries for Affinity Maturation. Int J Mol Sci 2019; 20:E1861. [PMID: 30991723 PMCID: PMC6515083 DOI: 10.3390/ijms20081861] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/10/2019] [Accepted: 04/12/2019] [Indexed: 12/25/2022] Open
Abstract
Antibodies leverage on their unique architecture to bind with an array of antigens. The strength of interaction has a direct relation to the affinity of the antibodies towards the antigen. In vivo affinity maturation is performed through multiple rounds of somatic hypermutation and selection in the germinal centre. This unique process involves intricate sequence rearrangements at the gene level via molecular mechanisms. The emergence of in vitro display technologies, mainly phage display and recombinant DNA technology, has helped revolutionize the way antibody improvements are being carried out in the laboratory. The adaptation of molecular approaches in vitro to replicate the in vivo processes has allowed for improvements in the way recombinant antibodies are designed and tuned. Combinatorial libraries, consisting of a myriad of possible antibodies, are capable of replicating the diversity of the natural human antibody repertoire. The isolation of target-specific antibodies with specific affinity characteristics can also be accomplished through modification of stringent protocols. Despite the ability to screen and select for high-affinity binders, some 'fine tuning' may be required to enhance antibody binding in terms of its affinity. This review will provide a brief account of phage display technology used for antibody generation followed by a summary of different combinatorial library characteristics. The review will focus on available strategies, which include molecular approaches, next generation sequencing, and in silico approaches used for antibody affinity maturation in both therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Chia Chiu Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang 11800, Malaysia.
| | - Yee Siew Choong
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang 11800, Malaysia.
| | - Theam Soon Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang 11800, Malaysia.
- Analytical Biochemistry Research Centre, Universiti Sains Malaysia, Penang 11800, Malaysia.
| |
Collapse
|
4
|
An innovative flow cytometry method to screen human scFv-phages selected by in vivo phage-display in an animal model of atherosclerosis. Sci Rep 2018; 8:15016. [PMID: 30302027 PMCID: PMC6177473 DOI: 10.1038/s41598-018-33382-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 08/29/2018] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is a chronic, progressive inflammatory disease that may develop into vulnerable lesions leading to thrombosis. This pathology is characterized by the deposition of lipids within the arterial wall and infiltration of immune cells leading to amplification of inflammation. Nowadays there is a rising interest to assess directly the molecular and cellular components that underlie the clinical condition of stroke and myocardial infarction. Single chain fragment variable (scFv)-phages issuing from a human combinatorial library were selected on the lesions induced in a rabbit model of atherosclerosis after three rounds of in vivo phage display. We further implemented a high-throughput flow cytometry method on rabbit protein extracts to individually test one thousand of scFv-phages. Two hundred and nine clones were retrieved on the basis of their specificity for atherosclerotic proteins. Immunohistochemistry assays confirmed the robustness of the designed cytometry protocol. Sequencing of candidates demonstrated their high diversity in VH and VL germline usage. The large number of candidates and their diversity open the way in the discovery of new biomarkers. Here, we successfully showed the capacity of combining in vivo phage display and high-throughput cytometry strategies to give new insights in in vivo targetable up-regulated biomarkers in atherosclerosis.
Collapse
|
5
|
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: 2.0] [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
|
6
|
Forloni M, Liu AY, Wajapeyee N. Random Mutagenesis Using Error-Prone DNA Polymerases. Cold Spring Harb Protoc 2018; 2018:2018/3/pdb.prot097741. [PMID: 29496818 DOI: 10.1101/pdb.prot097741] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
"Random mutagenesis" is a technique that allows researchers to develop large libraries of variants of a particular DNA sequence. Once developed, these libraries can then be used for several purposes, including structure-function and directed evolution studies. Random mutagenesis is different from other mutagenesis techniques in that it does not require the researcher to have any prior knowledge about the structural properties of the DNA sequence being targeted, thus allowing for the unbiased discovery of novel or beneficial mutations. For this reason, random mutagenesis is especially useful for protein evolution studies. This protocol describes mutagenic replication in vitro by a low-fidelity DNA polymerase followed by selective polymerase chain reaction (PCR) amplification of the newly mutated sequences. The initial mutagenic DNA replication step is accomplished by heat-denaturing the template DNA and annealing primers possessing 5' extensions that are not complementary to the template. The purpose of the noncomplementary extensions on the primers is to allow for future selection of only the mutant strands. DNA replication is then performed by a low-fidelity DNA polymerase of choice (polymerase β, η, or ι, or any combination of the three). After mutations have been incorporated into the template, the mutagenized strands are then selectively amplified using PCR. Selective amplification of the mutant strands is accomplished by performing a PCR procedure consisting of a first cycle with a low hybridization temperature followed by subsequent selection cycles under higher hybridization temperatures that do not allow amplification of the original unmutagenized template.
Collapse
|
7
|
Monnet C, Jorieux S, Urbain R, Fournier N, Bouayadi K, De Romeuf C, Behrens CK, Fontayne A, Mondon P. Selection of IgG Variants with Increased FcRn Binding Using Random and Directed Mutagenesis: Impact on Effector Functions. Front Immunol 2015; 6:39. [PMID: 25699055 PMCID: PMC4316771 DOI: 10.3389/fimmu.2015.00039] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/21/2015] [Indexed: 12/26/2022] Open
Abstract
Despite the reasonably long half-life of immunoglogulin G (IgGs), market pressure for higher patient convenience while conserving efficacy continues to drive IgG half-life improvement. IgG half-life is dependent on the neonatal Fc receptor (FcRn), which among other functions, protects IgG from catabolism. FcRn binds the Fc domain of IgG at an acidic pH ensuring that endocytosed IgG will not be degraded in lysosomal compartments and will then be released into the bloodstream. Consistent with this mechanism of action, several Fc-engineered IgG with increased FcRn affinity and conserved pH dependency were designed and resulted in longer half-life in vivo in human FcRn-transgenic mice (hFcRn), cynomolgus monkeys, and recently in healthy humans. These IgG variants were usually obtained by in silico approaches or directed mutagenesis in the FcRn-binding site. Using random mutagenesis, combined with a pH-dependent phage display selection process, we isolated IgG variants with improved FcRn-binding, which exhibited longer in vivo half-life in hFcRn mice. Interestingly, many mutations enhancing Fc/FcRn interaction were located at a distance from the FcRn-binding site validating our random molecular approach. Directed mutagenesis was then applied to generate new variants to further characterize our IgG variants and the effect of the mutations selected. Since these mutations are distributed over the whole Fc sequence, binding to other Fc effectors, such as complement C1q and FcγRs, was dramatically modified, even by mutations distant from these effectors’ binding sites. Hence, we obtained numerous IgG variants with increased FcRn-binding and different binding patterns to other Fc effectors, including variants without any effector function, providing distinct “fit-for-purpose” Fc molecules. We therefore provide evidence that half-life and effector functions should be optimized simultaneously as mutations can have unexpected effects on all Fc receptors that are critical for IgG therapeutic efficacy.
Collapse
|
8
|
Deyev SM, Lebedenko EN, Petrovskaya LE, Dolgikh DA, Gabibov AG, Kirpichnikov MP. Man-made antibodies and immunoconjugates with desired properties: function optimization using structural engineering. RUSSIAN CHEMICAL REVIEWS 2015. [DOI: 10.1070/rcr4459] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
9
|
Zhao A, Tohidkia MR, Siegel DL, Coukos G, Omidi Y. Phage antibody display libraries: a powerful antibody discovery platform for immunotherapy. Crit Rev Biotechnol 2014; 36:276-89. [PMID: 25394539 DOI: 10.3109/07388551.2014.958978] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Phage display technology (PDT), a combinatorial screening approach, provides a molecular diversity tool for creating libraries of peptides/proteins and discovery of new recombinant therapeutics. Expression of proteins such as monoclonal antibodies (mAbs) on the surface of filamentous phage can permit the selection of high affinity and specificity therapeutic mAbs against virtually any target antigen. Using a number of diverse selection platforms (e.g. solid phase, solution phase, whole cell and in vivo biopannings), phage antibody libraries (PALs) from the start point provides great potential for the isolation of functional mAb fragments with diagnostic and/or therapeutic purposes. Given the pivotal role of PDT in the discovery of novel therapeutic/diagnostic mAbs, in the current review, we provide an overview on PALs and discuss their impact in the advancement of engineered mAbs.
Collapse
Affiliation(s)
- Aizhi Zhao
- a Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania , Philadelphia , PA , USA
| | - Mohammad R Tohidkia
- b Research Center for Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Donald L Siegel
- c Division of Transfusion Medicine, Department of Pathology & Laboratory Medicine , University of Pennsylvania School of Medicine , Philadelphia , PA , USA , and
| | - George Coukos
- a Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania , Philadelphia , PA , USA .,d Ludwig Center for Cancer Research, University of Lausanne , Lausanne , Switzerland
| | - Yadollah Omidi
- a Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania , Philadelphia , PA , USA .,b Research Center for Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
10
|
Monnet C, Jorieux S, Souyris N, Zaki O, Jacquet A, Fournier N, Crozet F, de Romeuf C, Bouayadi K, Urbain R, Behrens CK, Mondon P, Fontayne A. Combined glyco- and protein-Fc engineering simultaneously enhance cytotoxicity and half-life of a therapeutic antibody. MAbs 2014; 6:422-36. [PMID: 24492301 DOI: 10.4161/mabs.27854] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
While glyco-engineered monoclonal antibodies (mAbs) with improved antibody-dependent cell-mediated cytotoxicity (ADCC) are reaching the market, extensive efforts have also been made to improve their pharmacokinetic properties to generate biologically superior molecules. Most therapeutic mAbs are human or humanized IgG molecules whose half-life is dependent on the neonatal Fc receptor FcRn. FcRn reduces IgG catabolism by binding to the Fc domain of endocytosed IgG in acidic lysosomal compartments, allowing them to be recycled into the blood. Fc-engineered mAbs with increased FcRn affinity resulted in longer in vivo half-life in animal models, but also in healthy humans. These Fc-engineered mAbs were obtained by alanine scanning, directed mutagenesis or in silico approach of the FcRn binding site. In our approach, we applied a random mutagenesis technology (MutaGen™) to generate mutations evenly distributed over the whole Fc sequence of human IgG1. IgG variants with improved FcRn-binding were then isolated from these Fc-libraries using a pH-dependent phage display selection process. Two successive rounds of mutagenesis and selection were performed to identify several mutations that dramatically improve FcRn binding. Notably, many of these mutations were unpredictable by rational design as they were located distantly from the FcRn binding site, validating our random molecular approach. When produced on the EMABling(®) platform allowing effector function increase, our IgG variants retained both higher ADCC and higher FcRn binding. Moreover, these IgG variants exhibited longer half-life in human FcRn transgenic mice. These results clearly demonstrate that glyco-engineering to improve cytotoxicity and protein-engineering to increase half-life can be combined to further optimize therapeutic mAbs.
Collapse
|
11
|
Anti-HER3 domain 1 and 3 antibodies reduce tumor growth by hindering HER2/HER3 dimerization and AKT-induced MDM2, XIAP, and FoxO1 phosphorylation. Neoplasia 2013; 15:335-47. [PMID: 23479511 DOI: 10.1593/neo.121960] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 01/16/2013] [Accepted: 01/18/2013] [Indexed: 12/17/2022] Open
Abstract
Blockade of the human epidermal growth factor receptor 3 (HER3) and of the downstream phosphatidylinositide 3-kinase (PI3K)/AKT pathway is a prerequisite for overcoming drug resistance and to develop novel treatments for cancers that are not eligible for the currently approved targeted therapies. To this end, we generated specific antibodies (Abs) against domain 1 (D1) and domain 3 (D3) of HER3 that recognize epitopes that do not overlap with the neuregulin-binding site. The fully human H4B-121 Ab and the mouse monoclonal Abs 16D3-C1 and 9F7-F11 inhibited tumor growth in nude mice xenografted with epidermoid, pancreatic, or triple-negative breast cancer cells. The combination of one anti-HER3 Ab and trastuzumab improved tumor growth inhibition in mice xenografted with HER2(low) cancer cell lines, for which trastuzumab alone shows no or moderate efficiency. Ab-induced disruption of tumor growth was associated with G1 cell cycle arrest, proliferation inhibition, and apoptosis of cancer cells. Anti-HER3 Abs blocked HER2/HER3 heterodimerization and HER3 phosphorylation at the cell membrane, leading to inhibition of phosphorylation of the downstream AKT targets murine double minute 2, X-linked inhibitor of apoptosis, and forkhead box O1. This study demonstrates that anti-HER3 D1 and D3 Abs could represent a new option for immunotherapy of pancreatic and triple-negative breast cancers.
Collapse
|
12
|
|
13
|
Renaut L, Monnet C, Dubreuil O, Zaki O, Crozet F, Bouayadi K, Kharrat H, Mondon P. Affinity maturation of antibodies: optimized methods to generate high-quality ScFv libraries and isolate IgG candidates by high-throughput screening. Methods Mol Biol 2012; 907:451-61. [PMID: 22907368 DOI: 10.1007/978-1-61779-974-7_26] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
As a growing number of therapeutic antibodies are developed, robust methods to efficiently improve the affinity and/or specificity of antibody candidates are needed. Here we describe our powerful platform that combines scFv affinity maturation and IgG high-throughput screening. After creating diversity with our random mutagenesis technology (MutaGen™), the scFv libraries are fully cleaned using a fusion system introducing the beta-lactamase gene to select in-frame and stop codon free variants on the basis of ampicillin resistance. The high-quality scFv libraries thereby constructed are then selected on the target in vitro using phage display technology. Contrary to standard procedures, instead of producing a limited number of affinity matured scFv as IgG molecules, we developed a cloning system to directly transfer the entire pool of selected scFv into an IgG expression vector permitting rapid IgG small-scale production (96 wells) in mammalian cells. Our integrated process allows us to generate high-quality scFv libraries and test numerous IgG variants, increasing the chances to select the best therapeutic antibody candidate.
Collapse
|
14
|
Deev SM, Labedenko EN. [Antibody engineering: barnase-barstar module as a molecular constructor]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2010; 35:761-78. [PMID: 20208576 DOI: 10.1134/s1068162009060041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Today, antibody engineering for clinical applications is a rapidly progressing field of science and a big business. The basic functions of an antibody can be spatially differentiated and attributed to various structural domains of a molecule. Therefore, each of them may be an object for engineering with the aim of using a definite antibody function. In this sense, the potential of antibodies is unique. In this article, recent achievements and current problems of antibody engineering are briefly reviewed. The main attention is focused on a molecular constructor that allows for obtaining, with the help of a versatile barnase-barstar module, mono- and multiva-lent miniantibodies and their derivatives with outlined properties.
Collapse
|
15
|
Mondon P, Grand D, Souyris N, Emond S, Bouayadi K, Kharrat H. Mutagen: a random mutagenesis method providing a complementary diversity generated by human error-prone DNA polymerases. Methods Mol Biol 2010; 634:373-86. [PMID: 20676997 DOI: 10.1007/978-1-60761-652-8_26] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Random mutagenesis is one of the most effective methodologies to generate variant libraries for directed protein evolution. Indeed, this approach requires no structural or mechanistic information and can uncover unexpected beneficial mutations. Here, we describe a new random mutagenesis method based on the use of human error-prone DNA polymerases (pol beta, pol eta and pol iota). This approach allows the random introduction of mutations through a single replication step followed by a selective PCR amplification of the replicated mutated sequences. The libraries generated using this methodology display different mutation rates and complementary mutational spectra. By taking advantage of the mutation bias of naturally highly error-prone DNA polymerases, MutaGen thus appears as a very useful tool for gene and protein randomization.
Collapse
Affiliation(s)
- Philippe Mondon
- Antibody Engineering and Molecular Evolution Department, MilleGen SA, Labège, France.
| | | | | | | | | | | |
Collapse
|
16
|
Saerens D, Huang L, Bonroy K, Muyldermans S. Antibody Fragments as Probe in Biosensor Development. SENSORS 2008; 8:4669-4686. [PMID: 27873779 PMCID: PMC3705465 DOI: 10.3390/s8084669] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Revised: 08/06/2008] [Accepted: 08/07/2008] [Indexed: 11/30/2022]
Abstract
Today's proteomic analyses are generating increasing numbers of biomarkers, making it essential to possess highly specific probes able to recognize those targets. Antibodies are considered to be the first choice as molecular recognition units due to their target specificity and affinity, which make them excellent probes in biosensor development. However several problems such as difficult directional immobilization, unstable behavior, loss of specificity and steric hindrance, may arise from using these large molecules. Luckily, protein engineering techniques offer designed antibody formats suitable for biomarker analysis. Minimization strategies of antibodies into Fab fragments, scFv or even single-domain antibody fragments like VH, VL or VHHs are reviewed. Not only the size of the probe but also other issues like choice of immobilization tag, type of solid support and probe stability are of critical importance in assay development for biosensing. In this respect, multiple approaches to specifically orient and couple antibody fragments in a generic one-step procedure directly on a biosensor substrate are discussed.
Collapse
Affiliation(s)
- Dirk Saerens
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium.
- Department of Molecular and Cellular Interactions, VIB, Brussels, Belgium.
| | - Lieven Huang
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
- Department of Molecular and Cellular Interactions, VIB, Brussels, Belgium
- Department of Molecular Biology, Technologiepark 927, B-9052 Zwijnaarde, Ghent University, Ghent, Belgium
- Department for Molecular Biomedical Research, VIB, Ghent, Belgium
| | | | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
- Department of Molecular and Cellular Interactions, VIB, Brussels, Belgium
| |
Collapse
|
17
|
Emond S, André I, Jaziri K, Potocki-Véronèse G, Mondon P, Bouayadi K, Kharrat H, Monsan P, Remaud-Simeon M. Combinatorial engineering to enhance thermostability of amylosucrase. Protein Sci 2008; 17:967-76. [PMID: 18441231 DOI: 10.1110/ps.083492608] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Amylosucrase is a transglucosidase that catalyzes amylose-like polymer synthesis from sucrose substrate. About 60,000 amylosucrase variants from two libraries generated by the MutaGen random mutagenesis method were submitted to an in vivo selection procedure leading to the isolation of more than 7000 active variants. These clones were then screened for increased thermostability using an automated screening process. This experiment yielded three improved variants (two double mutants and one single mutant) showing 3.5- to 10-fold increased half-lives at 50 degrees C compared to the wild-type enzyme. Structural analysis revealed that the main differences between wild-type amylosucrase and the most improved variant (R20C/A451T) might reside in the reorganization of salt bridges involving the surface residue R20 and the introduction of a hydrogen-bonding interaction between T451 of the B' domain and D488 of flexible loop 8. This double mutant is the most thermostable amylosucrase known to date and the only one usable at 50 degrees C. At this temperature, amylose synthesis by this variant using high sucrose concentration (600 mM) led to the production of amylose chains twice as long as those obtained by the wild-type enzyme at 30 degrees C.
Collapse
Affiliation(s)
- Stéphane Emond
- Université de Toulouse, INSA, UPS, INP, LISBP, F-31077 Toulouse, France
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Moutel S, Perez F. "Antibodies--Europe. Engineering the next generation of antibodies". Biotechnol J 2008; 3:298-300. [PMID: 18348242 DOI: 10.1002/biot.200800011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
19
|
Emond S, Mondon P, Pizzut-Serin S, Douchy L, Crozet F, Bouayadi K, Kharrat H, Potocki-Véronèse G, Monsan P, Remaud-Simeon M. A novel random mutagenesis approach using human mutagenic DNA polymerases to generate enzyme variant libraries. Protein Eng Des Sel 2008; 21:267-74. [DOI: 10.1093/protein/gzn004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|