101
|
Lehmann A, Wixted JHF, Shapovalov MV, Roder H, Dunbrack RL, Robinson MK. Stability engineering of anti-EGFR scFv antibodies by rational design of a lambda-to-kappa swap of the VL framework using a structure-guided approach. MAbs 2015; 7:1058-71. [PMID: 26337947 DOI: 10.1080/19420862.2015.1088618] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Phage-display technology facilitates rapid selection of antigen-specific single-chain variable fragment (scFv) antibodies from large recombinant libraries. ScFv antibodies, composed of a VH and VL domain, are readily engineered into multimeric formats for the development of diagnostics and targeted therapies. However, the recombinant nature of the selection strategy can result in VH and VL domains with sub-optimal biophysical properties, such as reduced thermodynamic stability and enhanced aggregation propensity, which lead to poor production and limited application. We found that the C10 anti-epidermal growth factor receptor (EGFR) scFv, and its affinity mutant, P2224, exhibit weak production from E. coli. Interestingly, these scFv contain a fusion of lambda3 and lambda1 V-region (LV3 and LV1) genes, most likely the result of a PCR aberration during library construction. To enhance the biophysical properties of these scFvs, we utilized a structure-based approach to replace and redesign the pre-existing framework of the VL domain to one that best pairs with the existing VH. We describe a method to exchange lambda sequences with a more stable kappa3 framework (KV3) within the VL domain that incorporates the original lambda DE-loop. The resulting scFvs, C10KV3_LV1DE and P2224KV3_LV1DE, are more thermodynamically stable and easier to produce from bacterial culture. Additionally, C10KV3_LV1DE and P2224KV3_LV1DE retain binding affinity to EGFR, suggesting that such a dramatic framework swap does not significantly affect scFv binding. We provide here a novel strategy for redesigning the light chain of problematic scFvs to enhance their stability and therapeutic applicability.
Collapse
Affiliation(s)
- Andreas Lehmann
- a Molecular Therapeutics Program, Fox Chase Cancer Center , Philadelphia , PA.,b Current address: Biogen , Cambridge MA
| | | | - Maxim V Shapovalov
- a Molecular Therapeutics Program, Fox Chase Cancer Center , Philadelphia , PA
| | - Heinrich Roder
- a Molecular Therapeutics Program, Fox Chase Cancer Center , Philadelphia , PA
| | - Roland L Dunbrack
- a Molecular Therapeutics Program, Fox Chase Cancer Center , Philadelphia , PA
| | - Matthew K Robinson
- a Molecular Therapeutics Program, Fox Chase Cancer Center , Philadelphia , PA
| |
Collapse
|
102
|
Chen L, Kutskova YA, Hong F, Memmott JE, Zhong S, Jenkinson MD, Hsieh CM. Preferential germline usage and VH/VL pairing observed in human antibodies selected by mRNA display. Protein Eng Des Sel 2015; 28:427-35. [PMID: 26337062 DOI: 10.1093/protein/gzv042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/31/2015] [Indexed: 11/13/2022] Open
Abstract
Since the invention of phage display, in vitro antibody display technologies have revolutionized the field of antibody discovery. In combination with antibody libraries constructed with sequences of human origin, such technologies enable accelerated therapeutic antibody discovery while bypassing the laborious animal immunization and hybridoma generation processes. Many in vitro display technologies developed since aim to differentiate from phage display by displaying full-length IgG proteins, utilizing eukaryotic translation system and codons, increasing library size or real-time kinetic selection by fluorescent activated cell sorting. We report here the development of an mRNA display technology and an accompanying HCDR3 size spectratyping monitor for human antibody discovery. Importantly, the mRNA display technology maintains a monovalent linkage between the mRNA (genotype) and display binding protein (phenotype), which minimizes avidity effect common in other display systems and allows for a stringent affinity and off-rate selection. The mRNA display technology successfully identified 100 human antibodies in 15 different selections against various targets from naïve human antibody libraries. These antibodies in general have high affinity and diversity. By analyzing the germline usage and combination of antibodies selected by the mRNA display technology, we identified trends and determined the productivity of each germline subgroup in the libraries that could serve as the knowledge base for constructing fully synthetic, next generation antibody libraries.
Collapse
Affiliation(s)
- Lei Chen
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Yuliya A Kutskova
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Feng Hong
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - John E Memmott
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Suju Zhong
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Megan D Jenkinson
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Chung-Ming Hsieh
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| |
Collapse
|
103
|
Choi DK, Bae J, Shin SM, Shin JY, Kim S, Kim YS. A general strategy for generating intact, full-length IgG antibodies that penetrate into the cytosol of living cells. MAbs 2015; 6:1402-14. [PMID: 25484049 DOI: 10.4161/mabs.36389] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Full-length IgG antibodies cannot cross cell membranes of living cells; this limits their use for direct targeting of cytosolic proteins. Here, we describe a general strategy for the generation of intact, full-length IgG antibodies, herein called cytotransmabs, which internalize into living cells and localize in the cytosol. We first generated a humanized light chain variable domain (VL) that could penetrate into the cytosol of living cells and was engineered for association with various subtypes of human heavy chain variable domains (VHs). When light chains with humanized VL were co-expressed with 3 heavy chains (HCs), including 2 HCs of the clinically approved adalimumab (Humira®) and bevacizumab (Avastin®), all 3 purified IgG antibodies were internalized into the cytoplasm of living cells. Cytotransmabs primarily internalized into living cells by the clathrin-mediated endocytic pathway through interactions with heparin sulfate proteoglycan that was expressed on the cell surface. The cytotransmabs escaped into the cytosol from early endosomes without being further transported into other cellular compartments, like the lysosomes, endoplasmic reticulum, Golgi apparatus, and nucleus. Furthermore, we generated a cytotransmab that co-localized with the targeted cytosolic protein when it was incubated with living cells, demonstrating that the cytotransmab can directly target cytosolic proteins. Internalized cytotransmabs did not show any noticeable cytotoxicity and remained in the cytosol for more than 6 h before being degraded by proteosomes. These results suggest that cytotransmabs, which efficiently enter living cells and reach the cytosolic space, will find widespread uses as research, diagnostic, and therapeutic agents.
Collapse
Affiliation(s)
- Dong-Ki Choi
- a Department of Molecular Science and Technology ; Ajou University ; Suwon , Korea
| | | | | | | | | | | |
Collapse
|
104
|
McConnell AD, Zhang X, Macomber JL, Chau B, Sheffer JC, Rahmanian S, Hare E, Spasojevic V, Horlick RA, King DJ, Bowers PM. A general approach to antibody thermostabilization. MAbs 2015; 6:1274-82. [PMID: 25517312 DOI: 10.4161/mabs.29680] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Antibody engineering to enhance thermostability may enable further application and ease of use of antibodies across a number of different areas. A modified human IgG framework has been developed through a combination of engineering approaches, which can be used to stabilize antibodies of diverse specificity. This is achieved through a combination of complementarity-determining region (CDR)-grafting onto the stable framework, mammalian cell display and in vitro somatic hypermutation (SHM). This approach allows both stabilization and maturation to affinities beyond those of the original antibody, as shown by the stabilization of an anti-HA33 antibody by approximately 10°C and affinity maturation of approximately 300-fold over the original antibody. Specificities of 10 antibodies of diverse origin were successfully transferred to the stable framework through CDR-grafting, with 8 of these successfully stabilized, including the therapeutic antibodies adalimumab, stabilized by 9.9°C, denosumab, stabilized by 7°C, cetuximab stabilized by 6.9°C and to a lesser extent trastuzumab stabilized by 0.8°C. This data suggests that this approach may be broadly useful for improving the biophysical characteristics of antibodies across a number of applications.
Collapse
Key Words
- CDR, complementarity-determining region
- CH2, heavy chain constant domain 2
- CH3, heavy chain constant domain 3
- DSC, differential scanning calorimetry
- HC, heavy chain
- LC, light chain
- NGF, β-nerve growth factor
- SHM, somatic hypermutation
- SPR, surface plasmon resonance
- TNF, tumor necrosis factor
- Tm, melting temperature
- VH, heavy chain variable region
- VL, light chain variable region
- affinity maturation
- monoclonal antibodies
- protein engineering
- solubility
- somatic hypermutation
- thermostability
Collapse
|
105
|
Sedlák E, Schaefer JV, Marek J, Gimeson P, Plückthun A. Advanced analyses of kinetic stabilities of iggs modified by mutations and glycosylation. Protein Sci 2015; 24:1100-13. [PMID: 25966898 DOI: 10.1002/pro.2691] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 04/11/2015] [Accepted: 04/29/2015] [Indexed: 01/07/2023]
Abstract
The stability of Immunoglobulin G (IgG) affects production, storage and usability, especially in the clinic. The complex thermal and isothermal transitions of IgGs, especially their irreversibilities, pose a challenge to the proper determination of parameters describing their thermodynamic and kinetic stability. Here, we present a reliable mathematical model to study the irreversible thermal denaturations of antibody variants. The model was applied to two unrelated IgGs and their variants with stabilizing mutations as well as corresponding non-glycosylated forms of IgGs and Fab fragments. Thermal denaturations of IgGs were analyzed with three transitions, one reversible transition corresponding to C(H)2 domain unfolding followed by two consecutive irreversible transitions corresponding to Fab and C(H)3 domains, respectively. The parameters obtained allowed us to examine the effects of these mutations on the stabilities of individual domains within the full-length IgG. We found that the kinetic stability of the individual Fab fragment is significantly lowered within the IgG context, possibly because of intramolecular aggregation upon heating, while the stabilizing mutations have an especially beneficial effect. Thermal denaturations of non-glycosylated variants of IgG consist of more than three transitions and could not be analyzed by our model. However, isothermal denaturations demonstrated that the lack of glycosylation affects the stability of all and not just of the C(H)2 domain, suggesting that the partially unfolded domains may interact with each other during unfolding. Investigating thermal denaturation of IgGs according to our model provides a valuable tool for detecting subtle changes in thermodynamic and/or kinetic stabilities of individual domains.
Collapse
Affiliation(s)
- Erik Sedlák
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.,Centre for Interdisciplinary Biosciences, P.J. Šafárik University, Moyzesova 11, Košice, 040 01, Slovakia.,Department of Biochemistry, P.J. Šafárik University, Moyzesova 11, Košice, 040 01, Slovakia
| | - Jonas V Schaefer
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Jozef Marek
- Department of Biophysics, Institute of Experimental Physics, Watsonova 47, Košice, 040 01, Slovakia
| | - Peter Gimeson
- Malvern Instruments Inc., Northampton, Massachusetts, 01060-2327
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| |
Collapse
|
106
|
Jarasch A, Koll H, Regula JT, Bader M, Papadimitriou A, Kettenberger H. Developability Assessment During the Selection of Novel Therapeutic Antibodies. J Pharm Sci 2015; 104:1885-1898. [DOI: 10.1002/jps.24430] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 02/28/2015] [Accepted: 03/03/2015] [Indexed: 01/02/2023]
|
107
|
Zhang YF, Phung Y, Gao W, Kawa S, Hassan R, Pastan I, Ho M. New high affinity monoclonal antibodies recognize non-overlapping epitopes on mesothelin for monitoring and treating mesothelioma. Sci Rep 2015; 5:9928. [PMID: 25996440 PMCID: PMC4440525 DOI: 10.1038/srep09928] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 03/23/2015] [Indexed: 12/17/2022] Open
Abstract
Mesothelin is an emerging cell surface target in mesothelioma and other solid tumors. Most antibody drug candidates recognize highly immunogenic Region I (296–390) on mesothelin. Here, we report a group of high-affinity non-Region I rabbit monoclonal antibodies. These antibodies do not compete for mesothelin binding with the immunotoxin SS1P that binds Region I of mesothelin. One pair of antibodies (YP218 and YP223) is suitable to detect soluble mesothelin in a sandwich ELISA with high sensitivity. The new assay can also be used to measure serum mesothelin concentration in mesothelioma patients, indicating its potential use for monitoring patients treated with current antibody therapies targeting Region I. The antibodies are highly specific and sensitive in immunostaining of mesothelioma. To explore their use in tumor therapy, we have generated the immunotoxins based on the Fv of these antibodies. One immunotoxin (YP218 Fv-PE38) exhibits potent anti-tumor cytotoxicity towards primary mesothelioma cell lines in vitro and an NCI-H226 xenograft tumor in mice. Furthermore, we have engineered a humanized YP218 Fv that retains full binding affinity for mesothelin-expressing cancer cells. In conclusion, with their unique binding properties, these antibodies may be promising candidates for monitoring and treating mesothelioma and other mesothelin-expressing cancers.
Collapse
Affiliation(s)
- Yi-Fan Zhang
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Bethesda, MD 20892, United States
| | - Yen Phung
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Bethesda, MD 20892, United States
| | - Wei Gao
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Bethesda, MD 20892, United States
| | - Seiji Kawa
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Bethesda, MD 20892, United States
| | - Raffit Hassan
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, United States
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Bethesda, MD 20892, United States
| | - Mitchell Ho
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Bethesda, MD 20892, United States
| |
Collapse
|
108
|
Singh S, Kroe-Barrett RR, Canada KA, Zhu X, Sepulveda E, Wu H, He Y, Raymond EL, Ahlberg J, Frego LE, Amodeo LM, Catron KM, Presky DH, Hanke JH. Selective targeting of the IL23 pathway: Generation and characterization of a novel high-affinity humanized anti-IL23A antibody. MAbs 2015; 7:778-91. [PMID: 25905918 PMCID: PMC4622456 DOI: 10.1080/19420862.2015.1032491] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Herein, we describe the generation and characterization of BI 655066, a novel, highly potent neutralizing anti-interleukin-23 (IL23) monoclonal antibody in clinical development for autoimmune conditions, including psoriasis and Crohn's disease. IL23 is a key driver of the differentiation, maintenance, and activity of a number of immune cell subsets, including T helper 17 (Th17) cells, which are believed to mediate the pathogenesis of several immune-mediated disorders. Thus, IL23 neutralization is an attractive therapeutic approach. Designing an antibody for clinical activity and convenience for the patient requires certain properties, such as high affinity, specificity, and solubility. These properties were achieved by directed design of the immunization, lead identification, and humanization procedures. Favorable substance and pharmacokinetic properties were established by biophysical assessments and studies in cynomolgus monkeys.
Collapse
Key Words
- ADCC, antibody-dependent cell-mediated cytotoxicity
- AUC, analytical ultracentrifugation
- BI 655066
- CCG, Chemical Computing Group
- CDRs, complementarity-determining regions
- CH, constant region
- Cκ, constant kappa
- DMF, dimethylformamide
- EOF, electro-osmotic flow
- ESI, electrospray ionization
- F, phenylalanine
- G, glycine
- GAHA, goat anti-human IgG gamma antibody
- HCLF, high concentration liquid formulation
- IL12, Interleukin 12
- IL12RB1, IL12 receptor subunit beta 1
- IL23, Interleukin-23
- IL23R, IL23 receptor
- JAK2, Janus kinase 2
- PBS, phosphate-buffered saline
- PK, pharmacokinetic
- RU, resonance units
- SEC, size-exclusion chromatography
- SPR, surface plasmon resonance
- Th17, T helper 17 cells
- UV, ultraviolet
- V, variable
- VH, variable heavy
- Vκ, variable kappa
- Y, tyrosine
- biophysical assessment
- humanization
- immunogen design
- pharmacokinetic profile
- tyk2, tyrosine kinase 2
Collapse
Affiliation(s)
- Sanjaya Singh
- a Department of Biotherapeutics Research; Boehringer Ingelheim Pharmaceuticals Inc. ; Ridgefield , CT , USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Rouet R, Dudgeon K, Christie M, Langley D, Christ D. Fully Human VH Single Domains That Rival the Stability and Cleft Recognition of Camelid Antibodies. J Biol Chem 2015; 290:11905-17. [PMID: 25737448 DOI: 10.1074/jbc.m114.614842] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Indexed: 01/01/2023] Open
Abstract
Human VH single domains represent a promising class of antibody fragments with applications as therapeutic modalities. Unfortunately, isolated human VH domains also generally display poor biophysical properties and a propensity to aggregate. This has encouraged the development of non-human antibody domains as alternative means of antigen recognition and, in particular, camelid (VHH) domains. Naturally devoid of light chain partners, these domains are characterized by favorable biophysical properties and propensity for cleft binding, a highly desirable characteristic, allowing the targeting of cryptic epitopes. In contrast, previously reported structures of human VH single domains had failed to recapitulate this property. Here we report the engineering and characterization of phage display libraries of stable human VH domains and the selection of binders against a diverse set of antigens. Unlike "camelized" human domains, the domains do not rely on potentially immunogenic framework mutations and maintain the structure of the VH/VL interface. Structure determination in complex with hen egg white lysozyme revealed an extended VH binding interface, with complementarity-determining region 3 deeply penetrating into the active site cleft, highly reminiscent of what has been observed for camelid domains. Taken together, our results demonstrate that fully human VH domains can be constructed that are not only stable and well expressed but also rival the cleft binding properties of camelid antibodies.
Collapse
Affiliation(s)
- Romain Rouet
- From the Department of Immunology, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia and
| | - Kip Dudgeon
- From the Department of Immunology, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia and
| | - Mary Christie
- From the Department of Immunology, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia and the Faculty of Medicine, St. Vincent's Clinical School, University of New South Wales, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - David Langley
- From the Department of Immunology, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia and
| | - Daniel Christ
- From the Department of Immunology, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia and the Faculty of Medicine, St. Vincent's Clinical School, University of New South Wales, Darlinghurst, Sydney, New South Wales 2010, Australia
| |
Collapse
|
110
|
Friedel T, Hanisch LJ, Muth A, Honegger A, Abken H, Plückthun A, Buchholz CJ, Schneider IC. Receptor-targeted lentiviral vectors are exceptionally sensitive toward the biophysical properties of the displayed single-chain Fv. Protein Eng Des Sel 2015; 28:93-106. [PMID: 25715658 DOI: 10.1093/protein/gzv005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
An increasing number of applications require the expression of single-chain variable fragments (scFv) fusion proteins in mammalian cells at the cell surface membrane. Here we assessed the CD30-specific scFv HRS3, which is used in immunotherapy, for its ability to retarget lentiviral vectors (LVs) to CD30 and to mediate selective gene transfer into CD30-positive cells. Fused to the C-terminus of the type-II transmembrane protein hemagglutinin (H) of measles virus and expressed in LV packaging cells, gene transfer mediated by the released LV particles was inefficient. A series of point mutations in the scFv framework regions addressing its biophysical properties, which substantially improved production and increased the melting temperature without impairing its kinetic binding behavior to CD30, also improved the performance of LV particles. Gene transfer into CD30-positive cells increased ∼100-fold due to improved transport of the H-scFv protein to the plasma membrane. Concomitantly, LV particle aggregation and syncytia formation in packaging cells were substantially reduced. The data suggest that syncytia formation can be triggered by trans-cellular dimerization of H-scFv proteins displayed on adjacent cells. Taken together, we show that the biophysical properties of the targeting ligand have a decisive role for the gene transfer efficiency of receptor-targeted LVs.
Collapse
Affiliation(s)
- Thorsten Friedel
- Section of Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, Langen 63225, Germany
| | - Lydia J Hanisch
- Roche Pharmaceutical Research and Early Development, Protein Engineering Group, Roche Innovation Center Zürich, Schlieren 8952, Switzerland
| | - Anke Muth
- Section of Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, Langen 63225, Germany
| | - Annemarie Honegger
- Department of Biochemistry, University of Zürich, Zürich 8057, Switzerland
| | - Hinrich Abken
- Center for Molecular Medicine Cologne, University of Cologne, Cologne 50931, Germany Department I of Internal Medicine, University Hospital Cologne, Cologne 50931, Germany
| | - Andreas Plückthun
- Department of Biochemistry, University of Zürich, Zürich 8057, Switzerland
| | - Christian J Buchholz
- Section of Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, Langen 63225, Germany
| | - Irene C Schneider
- Section of Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, Langen 63225, Germany
| |
Collapse
|
111
|
Exploiting light chains for the scalable generation and platform purification of native human bispecific IgG. Nat Commun 2015; 6:6113. [PMID: 25672245 PMCID: PMC4339886 DOI: 10.1038/ncomms7113] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 12/15/2014] [Indexed: 12/23/2022] Open
Abstract
Bispecific antibodies enable unique therapeutic approaches but it remains a challenge to produce them at the industrial scale, and the modifications introduced to achieve bispecificity often have an impact on stability and risk of immunogenicity. Here we describe a fully human bispecific IgG devoid of any modification, which can be produced at the industrial scale, using a platform process. This format, referred to as a κλ-body, is assembled by co-expressing one heavy chain and two different light chains, one κ and one λ. Using ten different targets, we demonstrate that light chains can play a dominant role in mediating specificity and high affinity. The κλ-bodies support multiple modes of action, and their stability and pharmacokinetic properties are indistinguishable from therapeutic antibodies. Thus, the κλ-body represents a unique, fully human format that exploits light-chain variable domains for antigen binding and light-chain constant domains for robust downstream processing, to realize the potential of bispecific antibodies. Bispecific antibodies allow for novel therapeutic approaches but industrial-scale production and immunogenicity represent significant challenges. Here Fischer et al. describe a unique human bispecific antibody format that exploits differing light chains to overcome these obstacles.
Collapse
|
112
|
Enever C, Pupecka-Swider M, Sepp A. Stress selections on domain antibodies: 'what doesn't kill you makes you stronger'. Protein Eng Des Sel 2015; 28:59-66. [PMID: 25655396 DOI: 10.1093/protein/gzu057] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In addition to the desired specificity and affinity for their respective therapeutic targets, antibody-based drugs must also demonstrate an ability to be manufactured and formulated at the concentrations needed for therapeutic application and to remain resistant to aggregation during storage to reduce the risk of induced immunogenicity. Improvements to the thermodynamic stability of the folded state of the protein are considered to be critical for decreasing the aggregation propensity of the protein. In this work, we have improved the biophysical properties of a number of human domain antibodies (dAbs) by identifying mutations which decrease the propensity for dAb self-aggregation without compromising the affinity for their respective target antigen. The mutations were identified by subjecting phage-displayed error-prone PCR-generated libraries to a variety of generic environmental conditions (temperature, pH and protease) followed by antigen capture, facilitating selection for improved thermodynamic stability of the protein. The results indicate that sufficient sequence diversity usually exists within the complementarity determining regions of dAbs to allow for mutations that lead to improvements to biophysical properties with full retention of parent lead biochemical and biological properties. Improved biophysical properties were often accompanied by higher apparent melting temperature values, while alternative selection pressures often identified similar features, suggesting generic nature of these mutations.
Collapse
Affiliation(s)
- C Enever
- Biopharm Innovation, RD Biopharm R&D, GlaxoSmithKline Plc., 315 Cambridge Science Park, Cambridge CB4 0WG, UK
| | - M Pupecka-Swider
- Biopharm Innovation, RD Biopharm R&D, GlaxoSmithKline Plc., 315 Cambridge Science Park, Cambridge CB4 0WG, UK
| | - A Sepp
- Biopharm Innovation, RD Biopharm R&D, GlaxoSmithKline Plc., 315 Cambridge Science Park, Cambridge CB4 0WG, UK
| |
Collapse
|
113
|
Razinkov VI, Treuheit MJ, Becker GW. Accelerated formulation development of monoclonal antibodies (mAbs) and mAb-based modalities: review of methods and tools. ACTA ACUST UNITED AC 2015; 20:468-83. [PMID: 25576149 DOI: 10.1177/1087057114565593] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
More therapeutic monoclonal antibodies and antibody-based modalities are in development today than ever before, and a faster and more accurate drug discovery process will ensure that the number of candidates coming to the biopharmaceutical pipeline will increase in the future. The process of drug product development and, specifically, formulation development is a critical bottleneck on the way from candidate selection to fully commercialized medicines. This article reviews the latest advances in methods of formulation screening, which allow not only the high-throughput selection of the most suitable formulation but also the prediction of stability properties under manufacturing and long-term storage conditions. We describe how the combination of automation technologies and high-throughput assays creates the opportunity to streamline the formulation development process starting from early preformulation screening through to commercial formulation development. The application of quality by design (QbD) concepts and modern statistical tools are also shown here to be very effective in accelerated formulation development of both typical antibodies and complex modalities derived from them.
Collapse
|
114
|
Dow BA, Tatulian SA, Davidson VL. Use of the amicyanin signal sequence for efficient periplasmic expression in E. coli of a human antibody light chain variable domain. Protein Expr Purif 2015; 108:9-12. [PMID: 25573388 DOI: 10.1016/j.pep.2014.12.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 12/24/2014] [Accepted: 12/27/2014] [Indexed: 01/03/2023]
Abstract
Periplasmic localization of recombinant proteins offers advantages over cytoplasmic protein expression. In this study signal sequence of amicyanin, which is encoded by the mauC gene of Paracoccus denitrificans, was used to express the light chain variable domain of the human κIO8/O18 germline antibody in the periplasm of Escherichiacoli. The expressed protein was purified in good yield (70mg/L of culture) in one step from the periplasmic fraction by affinity chromatography using an engineered hexahistidine tag. Circular dichroism spectroscopy was used to determine if the secondary and tertiary structures of the protein and its thermal stability corresponded to those of the native folded protein. The expressed and purified protein was indeed properly folded and exhibited a reasonable thermal transition temperature of 53°C. These results indicate that the amicyanin signal sequence may be particularly useful for prokaryotic expression of proteins which are prone to mis-folding, aggregation or formation of inclusion bodies, all of which were circumvented in this study.
Collapse
Affiliation(s)
- Brian A Dow
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, United States
| | - Suren A Tatulian
- Department of Physics, University of Central Florida, Orlando, FL 32816, United States
| | - Victor L Davidson
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, United States.
| |
Collapse
|
115
|
Abstract
The unbiased, paired analysis of T-cell receptor (TCR) α- and β-chain usage at the single-cell level provides a valuable window of understanding into the TCR repertoire and the nature of the immune response. Earlier technologies for TCR repertoire analysis were often limited to examining TCR complementarity-determining region 3 (CDR3) β expression or required in vitro cloning procedures that can artificially skew the TCR repertoire from its in vivo state. We describe here a direct ex vivo, single-cell-based strategy for the clonotypic analysis of TCRαβ repertoires that utilizes multiplexed panels of TCRα and TCRβ-specific primers in a nested PCR to amplify expressed transcripts from individual, epitope-specific T cells. This strategy yields the paired TCRαβ sequences of any given population of αβ T cells of interest.
Collapse
Affiliation(s)
- Pradyot Dash
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place/MS 351, Memphis, TN, 38105, USA.
| | - George C Wang
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place/MS 351, Memphis, TN, 38105, USA.,Center of Excellence in Geriatric Medicine, Newton Medical Center, Sparta, NJ, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place/MS 351, Memphis, TN, 38105, USA.
| |
Collapse
|
116
|
Remmele RL, Bee JS, Phillips JJ, Mo WD, Higazi DR, Zhang J, Lindo V, Kippen AD. Characterization of Monoclonal Antibody Aggregates and Emerging Technologies. ACS SYMPOSIUM SERIES 2015. [DOI: 10.1021/bk-2015-1202.ch005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Richard L. Remmele
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune One MedImmune Way, Gaithersburg, Maryland 20878, United States
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune Granta Park, Cambridge CB21 6GH, United Kingdom
| | - Jared S. Bee
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune One MedImmune Way, Gaithersburg, Maryland 20878, United States
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune Granta Park, Cambridge CB21 6GH, United Kingdom
| | - Jonathan J. Phillips
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune One MedImmune Way, Gaithersburg, Maryland 20878, United States
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune Granta Park, Cambridge CB21 6GH, United Kingdom
| | - Wenjun David Mo
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune One MedImmune Way, Gaithersburg, Maryland 20878, United States
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune Granta Park, Cambridge CB21 6GH, United Kingdom
| | - Daniel R. Higazi
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune One MedImmune Way, Gaithersburg, Maryland 20878, United States
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune Granta Park, Cambridge CB21 6GH, United Kingdom
| | - Jifeng Zhang
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune One MedImmune Way, Gaithersburg, Maryland 20878, United States
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune Granta Park, Cambridge CB21 6GH, United Kingdom
| | - Vivian Lindo
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune One MedImmune Way, Gaithersburg, Maryland 20878, United States
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune Granta Park, Cambridge CB21 6GH, United Kingdom
| | - Alistair D. Kippen
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune One MedImmune Way, Gaithersburg, Maryland 20878, United States
- Analytical Biotechnology, Biopharmaceutical Development, MedImmune Granta Park, Cambridge CB21 6GH, United Kingdom
| |
Collapse
|
117
|
Zielonka S, Weber N, Becker S, Doerner A, Christmann A, Christmann C, Uth C, Fritz J, Schäfer E, Steinmann B, Empting M, Ockelmann P, Lierz M, Kolmar H. Shark Attack: high affinity binding proteins derived from shark vNAR domains by stepwise in vitro affinity maturation. J Biotechnol 2014; 191:236-45. [PMID: 24862193 DOI: 10.1016/j.jbiotec.2014.04.023] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 04/08/2014] [Accepted: 04/28/2014] [Indexed: 11/17/2022]
Abstract
A novel method for stepwise in vitro affinity maturation of antigen-specific shark vNAR domains is described that exclusively relies on semi-synthetic repertoires derived from non-immunized sharks. Target-specific molecules were selected from a CDR3-randomized bamboo shark (Chiloscyllium plagiosum) vNAR library using yeast surface display as platform technology. Various antigen-binding vNAR domains were easily isolated by screening against several therapeutically relevant antigens, including the epithelial cell adhesion molecule (EpCAM), the Ephrin type-A receptor 2 (EphA2), and the human serine protease HTRA1. Affinity maturation was demonstrated for EpCAM and HTRA1 by diversifying CDR1 of target-enriched populations which allowed for the rapid selection of nanomolar binders. EpCAM-specific vNAR molecules were produced as soluble proteins and more extensively characterized via thermal shift assays and biolayer interferometry. Essentially, we demonstrate that high-affinity binders can be generated in vitro without largely compromising the desirable high thermostability of the vNAR scaffold.
Collapse
Affiliation(s)
- Stefan Zielonka
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Niklas Weber
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Stefan Becker
- Protein Engineering and Antibody Technologies, Merck Serono, Merck KGaA, Frankfurter Straße 250, D-64293 Darmstadt, Germany
| | - Achim Doerner
- Protein Engineering and Antibody Technologies, Merck Serono, Merck KGaA, Frankfurter Straße 250, D-64293 Darmstadt, Germany
| | - Andreas Christmann
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Christine Christmann
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Christina Uth
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Janine Fritz
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Elena Schäfer
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Björn Steinmann
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Martin Empting
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Department Drug Design and Optimization, Saarland University, Campus C2.3, D-66123 Saarbrücken, Germany
| | - Pia Ockelmann
- Goethe-University Frankfurt, Faculty of Biosciences, Max-von-Laue-Str. 13, D-60438 Frankfurt am Main, Germany; University Hospital Frankfurt, Department of Anesthesiology, Intensive-Care Medicine and Pain Therapy, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Michael Lierz
- Clinic for Birds, Reptiles, Amphibians and Fish, Justus-Liebig University, Gießen, Frankfurter Str. 91-93, D-35392 Giessen, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany.
| |
Collapse
|
118
|
Koerber JT, Hornsby MJ, Wells JA. An improved single-chain Fab platform for efficient display and recombinant expression. J Mol Biol 2014; 427:576-86. [PMID: 25481745 DOI: 10.1016/j.jmb.2014.11.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 11/17/2014] [Accepted: 11/25/2014] [Indexed: 12/13/2022]
Abstract
Antibody phage display libraries combined with high-throughput selections have recently demonstrated tremendous promise to create the next generation of renewable, recombinant antibodies to study proteins and their many post-translational modification states; however, many challenges still remain, such as optimized antibody scaffolds. Recently, a single-chain fragment antigen binding (Fab) (scFab) format, in which the carboxy-terminus of the light chain is linked to the amino-terminus of the heavy chain, was described to potentially combine the high display levels of a single-chain fragment variable with the high stability of purified Fabs. However, this format required removal of the interchain disulfide bond to achieve modest display levels and subsequent bacterial expression resulted in high levels of aggregated scFab, hindering further use of scFabs. Here, we developed an improved scFab format that retains the interchain disulfide bond by increasing the linker length between the light and heavy chains to improve display and bacterial expression levels to 1-3 mg/L. Furthermore, rerouting of the scFab to the co-translational signal recognition particle pathway combined with reengineering of the signal peptide sequence results in display levels 24-fold above the original scFab format and 3-fold above parent Fab levels. This optimized scFab scaffold can be easily reformatted in a single step for expression in a bacterial or mammalian host to produce stable (Tm of 81 °C), predominantly monomeric (>90%) antibodies at a high yield. Ultimately, this new scFab format will advance high-throughput antibody generation platforms to discover the next generation of research and therapeutic antibodies.
Collapse
Affiliation(s)
- James T Koerber
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, 94158, USA; Recombinant Antibody Network, University of California, San Francisco, CA, 94158, USA
| | - Michael J Hornsby
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, 94158, USA; Recombinant Antibody Network, University of California, San Francisco, CA, 94158, USA
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, 94158, USA; Recombinant Antibody Network, University of California, San Francisco, CA, 94158, USA.
| |
Collapse
|
119
|
Kim DY, Hussack G, Kandalaft H, Tanha J. Mutational approaches to improve the biophysical properties of human single-domain antibodies. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1844:1983-2001. [DOI: 10.1016/j.bbapap.2014.07.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 07/05/2014] [Accepted: 07/11/2014] [Indexed: 01/06/2023]
|
120
|
Kim DY, To R, Kandalaft H, Ding W, van Faassen H, Luo Y, Schrag JD, St-Amant N, Hefford M, Hirama T, Kelly JF, MacKenzie R, Tanha J. Antibody light chain variable domains and their biophysically improved versions for human immunotherapy. MAbs 2014; 6:219-35. [PMID: 24423624 PMCID: PMC3929445 DOI: 10.4161/mabs.26844] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
We set out to gain deeper insight into the potential of antibody light chain variable domains (VLs) as immunotherapeutics. To this end, we generated a naïve human VL phage display library and, by using a method previously shown to select for non-aggregating antibody heavy chain variable domains (VHs), we isolated a diversity of VL domains by panning the library against B cell super-antigen protein L. Eight domains representing different germline origins were shown to be non-aggregating at concentrations as high as 450 µM, indicating VL repertoires are a rich source of non-aggregating domains. In addition, the VLs demonstrated high expression yields in E. coli, protein L binding and high reversibility of thermal unfolding. A side-by-side comparison with a set of non-aggregating human VHs revealed that the VLs had similar overall profiles with respect to melting temperature (Tm), reversibility of thermal unfolding and resistance to gastrointestinal proteases. Successful engineering of a non-canonical disulfide linkage in the core of VLs did not compromise the non-aggregation state or protein L binding properties. Furthermore, the introduced disulfide bond significantly increased their Tms, by 5.5–17.5 °C, and pepsin resistance, although it somewhat reduced expression yields and subtly changed the structure of VLs. Human VLs and engineered versions may make suitable therapeutics due to their desirable biophysical features. The disulfide linkage-engineered VLs may be the preferred therapeutic format because of their higher stability, especially for oral therapy applications that necessitate high resistance to the stomach’s acidic pH and pepsin.
Collapse
Affiliation(s)
- Dae Young Kim
- Human Health Therapeutics; National Research Council Canada; Ottawa, ON Canada
| | - Rebecca To
- Human Health Therapeutics; National Research Council Canada; Ottawa, ON Canada
| | - Hiba Kandalaft
- Human Health Therapeutics; National Research Council Canada; Ottawa, ON Canada
| | - Wen Ding
- Human Health Therapeutics; National Research Council Canada; Ottawa, ON Canada
| | - Henk van Faassen
- Human Health Therapeutics; National Research Council Canada; Ottawa, ON Canada
| | - Yan Luo
- Human Health Therapeutics; National Research Council Canada; Ottawa, ON Canada
| | - Joseph D Schrag
- Human Health Therapeutics; National Research Council Canada; Montréal, QC Canada
| | - Nadereh St-Amant
- Centre for Vaccine Evaluation; Biologics and Genetic Therapies Directorate;, Health Canada; Ottawa, ON Canada
| | - Mary Hefford
- Centre for Vaccine Evaluation; Biologics and Genetic Therapies Directorate;, Health Canada; Ottawa, ON Canada
| | - Tomoko Hirama
- Human Health Therapeutics; National Research Council Canada; Ottawa, ON Canada
| | - John F Kelly
- Human Health Therapeutics; National Research Council Canada; Ottawa, ON Canada
| | - Roger MacKenzie
- Human Health Therapeutics; National Research Council Canada; Ottawa, ON Canada; School of Environmental Sciences; Ontario Agricultural College; University of Guelph; Guelph, ON Canada
| | - Jamshid Tanha
- Human Health Therapeutics; National Research Council Canada; Ottawa, ON Canada; School of Environmental Sciences; Ontario Agricultural College; University of Guelph; Guelph, ON Canada; Department of Biochemistry, Microbiology, and Immunology; University of Ottawa; Ottawa, ON Canada
| |
Collapse
|
121
|
Yim SS, Bang HB, Kim YH, Lee YJ, Jeong GM, Jeong KJ. Rapid isolation of antibody from a synthetic human antibody library by repeated fluorescence-activated cell sorting (FACS). PLoS One 2014; 9:e108225. [PMID: 25303314 PMCID: PMC4193741 DOI: 10.1371/journal.pone.0108225] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 08/27/2014] [Indexed: 11/19/2022] Open
Abstract
Antibodies and their derivatives are the most important agents in therapeutics and diagnostics. Even after the significant progress in the technology for antibody screening from huge libraries, it takes a long time to isolate an antibody, which prevents a prompt action against the spread of a disease. Here, we report a new strategy for isolating desired antibodies from a combinatorial library in one day by repeated fluorescence-activated cell sorting (FACS). First, we constructed a library of synthetic human antibody in which single-chain variable fragment (scFv) was expressed in the periplasm of Escherichia coli. After labeling the cells with fluorescent antigen probes, the highly fluorescent cells were sorted by using a high-speed cell sorter, and these cells were reused without regeneration in the next round of sorting. After repeating this sorting, the positive clones were completely enriched in several hours. Thus, we screened the library against three viral antigens, including the H1N1 influenza virus, Hepatitis B virus, and Foot-and-mouth disease virus. Finally, the potential antibody candidates, which show KD values between 10 and 100 nM against the target antigens, could be successfully isolated even though the library was relatively small (∼106). These results show that repeated FACS screening without regeneration of the sorted cells can be a powerful method when a rapid response to a spreading disease is required.
Collapse
Affiliation(s)
- Sung Sun Yim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST) (BK21 plus program), Daejeon, Republic of Korea
| | - Hyun Bae Bang
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST) (BK21 plus program), Daejeon, Republic of Korea
| | - Young Hwan Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST) (BK21 plus program), Daejeon, Republic of Korea
| | - Yong Jae Lee
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST) (BK21 plus program), Daejeon, Republic of Korea
| | - Gu Min Jeong
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST) (BK21 plus program), Daejeon, Republic of Korea
| | - Ki Jun Jeong
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST) (BK21 plus program), Daejeon, Republic of Korea
- Institute for the BioCentury, KAIST, Daejeon, Republic of Korea
- * E-mail:
| |
Collapse
|
122
|
Chen W, Gong R, Ying T, Prabakaran P, Zhu Z, Feng Y, Dimitrov DS. Discovery of novel candidate therapeutics and diagnostics based on engineered human antibody domains. Curr Drug Discov Technol 2014; 11:28-40. [PMID: 23863097 DOI: 10.2174/15701638113109990032] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 07/02/2013] [Accepted: 07/03/2013] [Indexed: 12/11/2022]
Abstract
The smallest independently folded antibody fragments, the domains, are emerging as promising scaffolds for candidate therapeutics and diagnostics that bind specifically targets of interest. The discovery of such binders is based on several technologies including structure-based design and generation of libraries of mutants displayed on phage or yeast, next-generation sequencing for diversity analysis, panning and screening of the libraries, affinity maturation of selected binders, and their expression, purification, and characterization for specific binding, function, and aggregation propensity. In this review, we describe these technologies as applied for the generation of engineered antibody domains (eAds), especially those derived from the human immunoglobulin heavy chain variable region (VH) and the second domain of IgG1 heavy chain constant region (CH2) as potential candidate therapeutics and diagnostics, and discuss examples of eAds against HIV-1 and cancer-related proteins.
Collapse
|
123
|
Molecular design of recombinant scFv antibodies for site-specific photocoupling to β-cyclodextrin in solution and onto solid support. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1844:2164-73. [PMID: 25172394 DOI: 10.1016/j.bbapap.2014.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 08/18/2014] [Accepted: 08/19/2014] [Indexed: 12/14/2022]
Abstract
The ability to design and tailor-make antibodies to meet the biophysical demands required by the vast range of current and future antibody-based applications within biotechnology and biomedicine will be essential. In this proof-of-concept study, we have for the first time tailored human recombinant scFv antibodies for site-specific photocoupling through the use of an unnatural amino acid (UAA) and the dock'n'flash technology. In more detail, we have successfully explored the possibility to expand the genetic code of E. coli and introduced the photoreactive UAA p-benzoyl-L-phenylalanine (pBpa), and showed that the mutated scFv antibody could be expressed in E. coli with retained structural and functional properties, as well as binding affinity. The pBpa group was then used for affinity capture of the mutated antibody by β-cyclodextrin (β-CD), which provided the hydrogen atoms to be abstracted in the subsequent photocoupling process upon irradiation at 365nm. The results showed that the pBpa mutated antibody could be site-specifically photocoupled to free and surface (array) immobilized β-CD. Taken together, this paves the way for novel means of tailoring recombinant scFv antibodies for site-specific photochemical-based tagging, functionalization and immobilization in numerous applications.
Collapse
|
124
|
Nokwe CN, Zacharias M, Yagi H, Hora M, Reif B, Goto Y, Buchner J. A residue-specific shift in stability and amyloidogenicity of antibody variable domains. J Biol Chem 2014; 289:26829-26846. [PMID: 25096580 DOI: 10.1074/jbc.m114.582247] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Variable (V) domains of antibodies are essential for antigen recognition by our adaptive immune system. However, some variants of the light chain V domains (VL) form pathogenic amyloid fibrils in patients. It is so far unclear which residues play a key role in governing these processes. Here, we show that the conserved residue 2 of VL domains is crucial for controlling its thermodynamic stability and fibril formation. Hydrophobic side chains at position 2 stabilize the domain, whereas charged residues destabilize and lead to amyloid fibril formation. NMR experiments identified several segments within the core of the VL domain to be affected by changes in residue 2. Furthermore, molecular dynamic simulations showed that hydrophobic side chains at position 2 remain buried in a hydrophobic pocket, and charged side chains show a high flexibility. This results in a predicted difference in the dissociation free energy of ∼10 kJ mol(-1), which is in excellent agreement with our experimental values. Interestingly, this switch point is found only in VL domains of the κ family and not in VLλ or in VH domains, despite a highly similar domain architecture. Our results reveal novel insight into the architecture of variable domains and the prerequisites for formation of amyloid fibrils. This might also contribute to the rational design of stable variable antibody domains.
Collapse
Affiliation(s)
- Cardine N Nokwe
- Center for Integrated Protein Science, Department of Chemie, Technische Universität München, Lichtenbergstrasse 4, D-85747 Garching, Germany
| | - Martin Zacharias
- Center for Integrated Protein Science, Department of Physik, Technische Universität München, James-Franck-Strasse 1, D-85748 Garching, Germany
| | - Hisashi Yagi
- Department of Chemistry and Biotechnology, Graduate School of Engineering and Center for Research on Green Sustainable Chemistry, Tottori University, 4-101 Koyamatyo-minami, Tottori 680-8550, Japan, and; Division of Protein Structural Biology, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Manuel Hora
- Center for Integrated Protein Science, Department of Chemie, Technische Universität München, Lichtenbergstrasse 4, D-85747 Garching, Germany
| | - Bernd Reif
- Center for Integrated Protein Science, Department of Chemie, Technische Universität München, Lichtenbergstrasse 4, D-85747 Garching, Germany
| | - Yuji Goto
- Division of Protein Structural Biology, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Johannes Buchner
- Center for Integrated Protein Science, Department of Chemie, Technische Universität München, Lichtenbergstrasse 4, D-85747 Garching, Germany,.
| |
Collapse
|
125
|
Swift J, Saing S, Rouet R, Dudgeon K, Schofield P, Sewell W, Christ D. Identification of aggregation inhibitors of the human antibody light chain repertoire by phage display. Protein Eng Des Sel 2014; 27:405-9. [DOI: 10.1093/protein/gzu026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
126
|
Messih MA, Lepore R, Marcatili P, Tramontano A. Improving the accuracy of the structure prediction of the third hypervariable loop of the heavy chains of antibodies. ACTA ACUST UNITED AC 2014; 30:2733-40. [PMID: 24930144 PMCID: PMC4173008 DOI: 10.1093/bioinformatics/btu194] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
MOTIVATION Antibodies are able to recognize a wide range of antigens through their complementary determining regions formed by six hypervariable loops. Predicting the 3D structure of these loops is essential for the analysis and reengineering of novel antibodies with enhanced affinity and specificity. The canonical structure model allows high accuracy prediction for five of the loops. The third loop of the heavy chain, H3, is the hardest to predict because of its diversity in structure, length and sequence composition. RESULTS We describe a method, based on the Random Forest automatic learning technique, to select structural templates for H3 loops among a dataset of candidates. These can be used to predict the structure of the loop with a higher accuracy than that achieved by any of the presently available methods. The method also has the advantage of being extremely fast and returning a reliable estimate of the model quality. AVAILABILITY AND IMPLEMENTATION The source code is freely available at http://www.biocomputing.it/H3Loopred/ .
Collapse
Affiliation(s)
- Mario Abdel Messih
- Department of Physics, Sapienza University, 00185 Rome, Italy, Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, Lyngby, Denmark and Istituto Pasteur-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| | - Rosalba Lepore
- Department of Physics, Sapienza University, 00185 Rome, Italy, Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, Lyngby, Denmark and Istituto Pasteur-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| | - Paolo Marcatili
- Department of Physics, Sapienza University, 00185 Rome, Italy, Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, Lyngby, Denmark and Istituto Pasteur-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| | - Anna Tramontano
- Department of Physics, Sapienza University, 00185 Rome, Italy, Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, Lyngby, Denmark and Istituto Pasteur-Fondazione Cenci Bolognetti, 00185 Rome, Italy Department of Physics, Sapienza University, 00185 Rome, Italy, Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, Lyngby, Denmark and Istituto Pasteur-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| |
Collapse
|
127
|
Banach M, Konieczny L, Roterman I. The fuzzy oil drop model, based on hydrophobicity density distribution, generalizes the influence of water environment on protein structure and function. J Theor Biol 2014; 359:6-17. [PMID: 24859428 DOI: 10.1016/j.jtbi.2014.05.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/25/2014] [Accepted: 05/05/2014] [Indexed: 12/24/2022]
Abstract
In this paper we show that the fuzzy oil drop model represents a general framework for describing the generation of hydrophobic cores in proteins and thus provides insight into the influence of the water environment upon protein structure and stability. The model has been successfully applied in the study of a wide range of proteins, however this paper focuses specifically on domains representing immunoglobulin-like folds. Here we provide evidence that immunoglobulin-like domains, despite being structurally similar, differ with respect to their participation in the generation of hydrophobic core. It is shown that β-structural fragments in β-barrels participate in hydrophobic core formation in a highly differentiated manner. Quantitatively measured participation in core formation helps explain the variable stability of proteins and is shown to be related to their biological properties. This also includes the known tendency of immunoglobulin domains to form amyloids, as shown using transthyretin to reveal the clear relation between amyloidogenic properties and structural characteristics based on the fuzzy oil drop model.
Collapse
Affiliation(s)
- Mateusz Banach
- Department of Bioinformatics and Telemedicine - Jagiellonian University - Medical College, Krakow, Poland; Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Leszek Konieczny
- Chair of Medical Chemistry - Jagiellonian University - Medical College, Krakow, Poland
| | - Irena Roterman
- Department of Bioinformatics and Telemedicine - Jagiellonian University - Medical College, Krakow, Poland.
| |
Collapse
|
128
|
Ohlin M. A new look at a poorly immunogenic neutralization epitope on cytomegalovirus glycoprotein B. Is there cause for antigen redesign? Mol Immunol 2014; 60:95-102. [PMID: 24802891 DOI: 10.1016/j.molimm.2014.03.015] [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: 02/26/2014] [Revised: 03/28/2014] [Accepted: 03/31/2014] [Indexed: 11/25/2022]
Abstract
The immune response is able to control cytomegalovirus infection in most subjects. However, in some patient groups the virus is not well contained resulting in disease and severe morbidity. The development of efficacious vaccines is therefore a high priority. Antibodies may contribute to protection against disease caused by CMV but the most efficient targets for protective humoral immunity are not precisely known. Glycoprotein B (gB) is a protein that is targeted by virus-neutralizing antibodies. One epitope on gB, AD-2, is poorly immunogenic following natural infection and vaccination. It is consequently not effectively exploited as a target for antibodies by the immune system. However, antibodies specific for this epitope, when they develop, display important functional activities that may play a role in protection against infection. In this study critical features of human antibody recognition of this epitope are re-assessed based on structural and immunochemical data. The analysis suggests that the immune system may only be able to develop an AD-2 specific antibody response through rare, very specific rearrangement events that by chance create a naïve B cell that can be recruited into an AD-2 specific immune response. These results reinvigorate the notion that if we are to be able to effectively exploit AD-2 specific humoral immunity we need to readdress the nature of the antigen incorporated into vaccines so as to more effectively recruit B cells into the response against this epitope.
Collapse
Affiliation(s)
- Mats Ohlin
- Dept. of Immunotechnology, Lund University, Medicon Village, Building 406, S-22381 Lund, Sweden.
| |
Collapse
|
129
|
Neuber T, Frese K, Jaehrling J, Jäger S, Daubert D, Felderer K, Linnemann M, Höhne A, Kaden S, Kölln J, Tiller T, Brocks B, Ostendorp R, Pabst S. Characterization and screening of IgG binding to the neonatal Fc receptor. MAbs 2014; 6:928-42. [PMID: 24802048 DOI: 10.4161/mabs.28744] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The neonatal Fc receptor (FcRn) protects immunoglobulin G (IgG) from degradation and increases the serum half-life of IgG, thereby contributing to a higher concentration of IgG in the serum. Because altered FcRn binding may result in a reduced or prolonged half-life of IgG molecules, it is advisable to characterize Fc receptor binding of therapeutic antibody lead candidates prior to the start of pre-clinical and clinical studies. In this study, we characterized the interactions between FcRn of different species (human, cynomolgus monkey, mouse and rat) and nine IgG molecules from different species and isotypes with common variable heavy (VH) and variable light chain (VL) domains. Binding was analyzed at acidic and neutral pH using surface plasmon resonance (SPR) and biolayer interferometry (BLI). Furthermore, we transferred the well-accepted, but low throughput SPR-based method for FcRn binding characterization to the BLI-based Octet platform to enable a higher sample throughput allowing the characterization of FcRn binding already during early drug discovery phase. We showed that the BLI-based approach is fit-for-purpose and capable of discriminating between IgG molecules with significant differences in FcRn binding affinities. Using this high-throughput approach we investigated FcRn binding of 36 IgG molecules that represented all VH/VL region combinations available in the fully human, recombinant antibody library Ylanthia®. Our results clearly showed normal FcRn binding profiles for all samples. Hence, the variations among the framework parts, complementarity-determining region (CDR) 1 and CDR2 of the fragment antigen binding (Fab) domain did not significantly change FcRn binding.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Anne Höhne
- MorphoSys AG; Martinsried/Planegg, Germany
| | | | | | | | | | | | | |
Collapse
|
130
|
Pauly F, Smedby KE, Jerkeman M, Hjalgrim H, Ohlsson M, Rosenquist R, Borrebaeck CAK, Wingren C. Identification of B-cell lymphoma subsets by plasma protein profiling using recombinant antibody microarrays. Leuk Res 2014; 38:682-90. [PMID: 24754901 DOI: 10.1016/j.leukres.2014.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/13/2014] [Accepted: 03/15/2014] [Indexed: 12/14/2022]
Abstract
B-cell lymphoma (BCL) heterogeneity represents a key issue, often making the classification and clinical management of these patients challenging. In this pilot study, we outlined the first resolved view of BCL disease heterogeneity on the protein level by deciphering disease-associated plasma biomarkers, specific for chronic lymphocytic leukemia, diffuse large B-cell lymphoma, follicular lymphoma, and mantle cell lymphoma, using recombinant antibody microarrays targeting mainly immunoregulatory proteins. The results showed the BCLs to be heterogeneous, and revealed potential novel subgroups of each BCL. In the case of diffuse large B-cell lymphoma, we also indicated a link between the novel subgroups and survival.
Collapse
Affiliation(s)
- Frida Pauly
- Department of Immunotechnology, Lund University, Lund, Sweden; CREATE Health, Lund University, Lund, Sweden
| | - Karin E Smedby
- Department of Medicine Solna, Clinical Epidemiology Unit, Karolinska Institutet, Stockholm, Sweden
| | - Mats Jerkeman
- Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Henrik Hjalgrim
- Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark
| | - Mattias Ohlsson
- Computational Biology & Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, Lund, Sweden; CREATE Health, Lund University, Lund, Sweden
| | - Richard Rosenquist
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Carl A K Borrebaeck
- Department of Immunotechnology, Lund University, Lund, Sweden; CREATE Health, Lund University, Lund, Sweden
| | - Christer Wingren
- Department of Immunotechnology, Lund University, Lund, Sweden; CREATE Health, Lund University, Lund, Sweden.
| |
Collapse
|
131
|
Chromikova V, Mader A, Steinfellner W, Kunert R. Evaluating the bottlenecks of recombinant IgM production in mammalian cells. Cytotechnology 2014; 67:343-56. [PMID: 24615530 DOI: 10.1007/s10616-014-9693-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 01/10/2014] [Indexed: 11/26/2022] Open
Abstract
Despite the fact, that monoclonal antibodies are the fastest growing group of biopharmaceuticals in development, this is not true for the IgM class, which remains as enigmatic as ever. While more examples of usefulness of IgMs for medical applications are emerging, their recombinant production is still not common. In our study, stable monoclonal IgM producing CHO DG44 and HEK 293 cell lines, expressing two model IgM molecules (IgM-617 and IgM-012) were established. Recombinant cell lines were compared in regard of specific productivity, specific growth rate, maximal achieved antibody titer, gene copy numbers and transcription levels of transgene. IgM-617 cell lines were identified as high while IgM-012 clones were low producers. Although differences in gene copy numbers as well as in transcription levels were observed, they did not seem to be a limitation. Levels of relevant endoplasmic reticulum-stress related proteins were analyzed and no indications of unfolded protein response were detected. This could indicate that the difference in the intrinsic protein stability of our model proteins (as was previously observed on purified samples) might cause lower yields of IgM-012. Transcriptomics and/or proteomics follow up studies might be necessary for identification of potential bottlenecks in IgM producing cell lines.
Collapse
Affiliation(s)
- Veronika Chromikova
- Department of Biotechnology, Vienna Institute of BioTechnology (BOKU - VIBT), University of Natural Resources and Life Sciences, Muthgasse 11, 1190, Vienna, Austria
| | | | | | | |
Collapse
|
132
|
Malpiedi LP, Nerli BB, Abdalla DSP, Pessoa A. Assessment of the effect of triton X-114 on the physicochemical properties of an antibody fragment. Biotechnol Prog 2014; 30:554-61. [DOI: 10.1002/btpr.1882] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 01/29/2014] [Indexed: 01/05/2023]
Affiliation(s)
- Luciana P. Malpiedi
- Biochemical and Pharmaceutical Technology Dept./FCF; University of São Paulo. Av. Prof. Lineu Prestes; 580-Bloco 16 CEP 05508-000 São Paulo Brazil
- Physical Chemistry Dept., Faculty of Biochemistry and Pharmaceutical Sciences; National University of Rosario; Suipacha 570 S2002LRK Rosario Argentina
| | - Bibiana B. Nerli
- Physical Chemistry Dept., Faculty of Biochemistry and Pharmaceutical Sciences; National University of Rosario; Suipacha 570 S2002LRK Rosario Argentina
| | | | - Adalberto Pessoa
- Biochemical and Pharmaceutical Technology Dept./FCF; University of São Paulo. Av. Prof. Lineu Prestes; 580-Bloco 16 CEP 05508-000 São Paulo Brazil
| |
Collapse
|
133
|
|
134
|
Abstract
Antibody-based microarrays are a novel technology that hold great promise in proteomics. Microarrays can be printed with thousands of recombinant antibodies carrying the desired specificities, the biologic sample (e.g., an entire proteome) and any specifically bound analytes detected. The microarray patterns that are generated can then be converted into proteomic maps, or molecular fingerprints, revealing the composition of the proteome. Using this tool, global proteome analysis and protein expression profiling will thus provide new opportunities for biomarker discovery, drug target identification and disease diagnostics, as well as providing insights into disease biology. Intense work is currently underway to develop this novel technology platform into the high-throughput proteomic tool required by the research community.
Collapse
Affiliation(s)
- Christer Wingren
- Department of Immunotechnology, Lund University, PO Box 7031, Lund, Sweden.
| | | |
Collapse
|
135
|
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: 15] [Impact Index Per Article: 1.4] [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
|
136
|
Chatel A, Kumpalume P, Hoare M. Ultra scale-down characterization of the impact of conditioning methods for harvested cell broths on clarification by continuous centrifugation-Recovery of domain antibodies from rec E. coli. Biotechnol Bioeng 2013; 111:913-24. [PMID: 24284936 PMCID: PMC4153950 DOI: 10.1002/bit.25164] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 11/19/2013] [Accepted: 11/19/2013] [Indexed: 11/30/2022]
Abstract
The processing of harvested E. coli cell broths is examined where the expressed protein product has been released into the extracellular space. Pre-treatment methods such as freeze–thaw, flocculation, and homogenization are studied. The resultant suspensions are characterized in terms of the particle size distribution, sensitivity to shear stress, rheology and solids volume fraction, and, using ultra scale-down methods, the predicted ability to clarify the material using industrial scale continuous flow centrifugation. A key finding was the potential of flocculation methods both to aid the recovery of the particles and to cause the selective precipitation of soluble contaminants. While the flocculated material is severely affected by process shear stress, the impact on the very fine end of the size distribution is relatively minor and hence the predicted performance was only diminished to a small extent, for example, from 99.9% to 99.7% clarification compared with 95% for autolysate and 65% for homogenate at equivalent centrifugation conditions. The lumped properties as represented by ultra scale-down centrifugation results were correlated with the basic properties affecting sedimentation including particle size distribution, suspension viscosity, and solids volume fraction. Grade efficiency relationships were used to allow for the particle and flow dynamics affecting capture in the centrifuge. The size distribution below a critical diameter dependant on the broth pre-treatment type was shown to be the main determining factor affecting the clarification achieved. Biotechnol. Bioeng. 2014;111: 913–924. © 2013 The Authors. Biotechnology and Bioengineering Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alex Chatel
- Department of Biochemical Engineering, UCL, Torrington Place, London, WC1E 7JE, UK
| | | | | |
Collapse
|
137
|
Pybus LP, James DC, Dean G, Slidel T, Hardman C, Smith A, Daramola O, Field R. Predicting the expression of recombinant monoclonal antibodies in Chinese hamster ovary cells based on sequence features of the CDR3 domain. Biotechnol Prog 2013; 30:188-97. [DOI: 10.1002/btpr.1839] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 10/27/2013] [Indexed: 11/11/2022]
Affiliation(s)
- Leon P. Pybus
- ChELSI Institute; Dept. of Chemical and Biological Engineering; University of Sheffield; Mappin Street, Sheffield S1 3JD U.K
| | - David C. James
- ChELSI Institute; Dept. of Chemical and Biological Engineering; University of Sheffield; Mappin Street, Sheffield S1 3JD U.K
| | - Greg Dean
- MedImmune Ltd.; Granta Park Cambridge CB21 6GH U.K
| | - Tim Slidel
- MedImmune Ltd.; Granta Park Cambridge CB21 6GH U.K
| | | | - Andrew Smith
- MedImmune Ltd.; Granta Park Cambridge CB21 6GH U.K
| | | | - Ray Field
- MedImmune Ltd.; Granta Park Cambridge CB21 6GH U.K
| |
Collapse
|
138
|
Riaño-Umbarila L, Olamendi-Portugal T, Morelos-Juárez C, Gurrola GB, Possani LD, Becerril B. A novel human recombinant antibody fragment capable of neutralizing Mexican scorpion toxins. Toxicon 2013; 76:370-6. [DOI: 10.1016/j.toxicon.2013.09.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 08/08/2013] [Accepted: 09/12/2013] [Indexed: 11/24/2022]
|
139
|
Rouet R, Lowe D, Christ D. Stability engineering of the human antibody repertoire. FEBS Lett 2013; 588:269-77. [PMID: 24291820 DOI: 10.1016/j.febslet.2013.11.029] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 11/20/2013] [Accepted: 11/20/2013] [Indexed: 10/26/2022]
Abstract
Human monoclonal antibodies often display limited thermodynamic and colloidal stabilities. This behavior hinders their production, and places limitations on the development of novel formulation conditions and therapeutic applications. Antibodies are highly diverse molecules, with much of the sequence variation observed within variable domain families and, in particular, their complementarity determining regions. This has complicated the development of comprehensive strategies for the stability engineering of the human antibody repertoire. Here we provide an overview of the field, and discuss recent advances in the development of robust and aggregation resistant antibody therapeutics.
Collapse
Affiliation(s)
- Romain Rouet
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia.
| | - David Lowe
- MedImmune, Milstein Building, Granta Park, Cambridge CB21 6GH, United Kingdom
| | - Daniel Christ
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; The University of New South Wales, Faculty of Medicine, St Vincent's Clinical School, Darlinghurst, Sydney, NSW 2010, Australia
| |
Collapse
|
140
|
Mandrup OA, Friis NA, Lykkemark S, Just J, Kristensen P. A novel heavy domain antibody library with functionally optimized complementarity determining regions. PLoS One 2013; 8:e76834. [PMID: 24116173 PMCID: PMC3792991 DOI: 10.1371/journal.pone.0076834] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 08/30/2013] [Indexed: 11/29/2022] Open
Abstract
Today a number of synthetic antibody libraries of different formats have been created and used for the selection of a large number of recombinant antibodies. One of the determining factors for successful isolation of recombinant antibodies from libraries lies in the quality of the libraries i.e. the number of correctly folded, functional antibodies contained in the library. Here, we describe the construction of a novel, high quality, synthetic single domain antibody library dubbed Predator. The library is based on the HEL4 domain antibody with the addition of recently reported mutations concerning the amino acid composition at positions critical for the folding characteristics and aggregation propensities of domain antibodies. As a unique feature, the CDR3 of the library was designed to mimic the natural human immune response by designating amino acids known to be prevalent in functional antibodies to the diversity in CDR3. CDR randomizations were performed using trinucleotide synthesis to avoid the presence of stop codons. Furthermore a novel cycle free elongation method was used for the conversion of the synthesized single stranded DNA containing the randomized CDRs into double stranded DNA of the library. In addition a modular approach has been adopted for the scaffold in which each CDR region is flanked by unique restrictions sites, allowing easy affinity maturation of selected clones by CDR shuffling. To validate the quality of the library, one round phage display selections were performed on purified antigens and highly complex antigen mixtures such as cultured eukaryotic cells resulting in several specific binders. The further characterization of some of the selected clones, however, indicates a reduction in thermodynamic stability caused by the inclusion the additional mutations to the HEL4 scaffold.
Collapse
Affiliation(s)
| | - Niels Anton Friis
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Simon Lykkemark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Sino-Danish Centre for Education and Research, Aarhus, Denmark
| | - Jesper Just
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Peter Kristensen
- Department of Engineering, Aarhus University, Aarhus, Denmark
- * E-mail:
| |
Collapse
|
141
|
Gong R, Wang Y, Ying T, Feng Y, Streaker E, Prabakaran P, Dimitrov DS. N-terminal truncation of an isolated human IgG1 CH2 domain significantly increases its stability and aggregation resistance. Mol Pharm 2013; 10:2642-52. [PMID: 23641816 PMCID: PMC3795862 DOI: 10.1021/mp400075f] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Isolated human immunoglobulin G (IgG) CH2 domains are promising scaffolds for novel candidate therapeutics. Unlike other human IgG domains, CH2 is not involved in strong interchain interactions, and isolated CH2 is relatively stable. However, isolated single CH2 is prone to aggregation. In native IgG and Fc molecules, the N-terminal residues of CH2 from the two heavy chains interact with each other and form hinge regions. By contrast, the N-terminal residues are highly disordered in isolated CH2. We have hypothesized that the removal of the CH2 N-terminal residues may not only increase its stability but also its aggregation resistance. To test this hypothesis we constructed a shortened variant of IgG1 CH2 (CH2s) where the first seven residues of the N-terminus were deleted. We found that the thermal stability of CH2s was increased by 5 °C compared to CH2. Importantly, we demonstrated that CH2s is significantly less prone to aggregation than CH2 as measured by Thioflavin T (ThT) fluorescence, turbidity, and light scattering. We also found that the CH2s exhibited pH-dependent binding to a soluble single-chain human neonatal Fc receptor (shFcRn) which was significantly stronger than the very weak binding of CH2 to shFcRn as measured by flow cytometry. Computer modeling suggested a possible mode of CH2 aggregation involving its N-terminal residues. Therefore, deletion of the N-terminal residues could increase drugability of CH2-based therapeutic candidates. This strategy to increase stability and aggregation resistance could also be applicable to other Ig-related proteins.
Collapse
Affiliation(s)
- Rui Gong
- Antibody Engineering Group, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- Protein Interactions Group, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Yanping Wang
- Protein Interactions Group, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
- Basic Research Program, Science Applications International Corporation-Frederick, Inc., Frederick, MD 21702, USA
| | - Tianlei Ying
- Protein Interactions Group, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Yang Feng
- Protein Interactions Group, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Emily Streaker
- Protein Interactions Group, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
- Basic Research Program, Science Applications International Corporation-Frederick, Inc., Frederick, MD 21702, USA
| | - Ponraj Prabakaran
- Protein Interactions Group, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
- Basic Research Program, Science Applications International Corporation-Frederick, Inc., Frederick, MD 21702, USA
| | - Dimiter S. Dimitrov
- Protein Interactions Group, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| |
Collapse
|
142
|
Olimpieri PP, Chailyan A, Tramontano A, Marcatili P. Prediction of site-specific interactions in antibody-antigen complexes: the proABC method and server. ACTA ACUST UNITED AC 2013; 29:2285-91. [PMID: 23803466 PMCID: PMC3753563 DOI: 10.1093/bioinformatics/btt369] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Motivation: Antibodies or immunoglobulins are proteins of paramount importance in the immune system. They are extremely relevant as diagnostic, biotechnological and therapeutic tools. Their modular structure makes it easy to re-engineer them for specific purposes. Short of undergoing a trial and error process, these experiments, as well as others, need to rely on an understanding of the specific determinants of the antibody binding mode. Results: In this article, we present a method to identify, on the basis of the antibody sequence alone, which residues of an antibody directly interact with its cognate antigen. The method, based on the random forest automatic learning techniques, reaches a recall and specificity as high as 80% and is implemented as a free and easy-to-use server, named prediction of Antibody Contacts. We believe that it can be of great help in re-design experiments as well as a guide for molecular docking experiments. The results that we obtained also allowed us to dissect which features of the antibody sequence contribute most to the involvement of specific residues in binding to the antigen. Availability:http://www.biocomputing.it/proABC. Contact:anna.tramontano@uniroma1.it or paolo.marcatili@gmail.com Supplementary information:Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Pier Paolo Olimpieri
- Department of Physics, Sapienza University and Istituto Pasteur - Fondazione Cenci Bolognetti, 00185 Rome, Italy
| | | | | | | |
Collapse
|
143
|
Dudgeon K, Rouet R, Christ D. Rapid prediction of expression and refolding yields using phage display. Protein Eng Des Sel 2013; 26:671-4. [DOI: 10.1093/protein/gzt019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
144
|
Kovalenko OV, Olland A, Piché-Nicholas N, Godbole A, King D, Svenson K, Calabro V, Müller MR, Barelle CJ, Somers W, Gill DS, Mosyak L, Tchistiakova L. Atypical antigen recognition mode of a shark immunoglobulin new antigen receptor (IgNAR) variable domain characterized by humanization and structural analysis. J Biol Chem 2013; 288:17408-19. [PMID: 23632026 DOI: 10.1074/jbc.m112.435289] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The immunoglobulin new antigen receptors (IgNARs) are a class of Ig-like molecules of the shark immune system that exist as heavy chain-only homodimers and bind antigens by their single domain variable regions (V-NARs). Following shark immunization and/or in vitro selection, V-NARs can be generated as soluble, stable, and specific high affinity monomeric binding proteins of ∼12 kDa. We have previously isolated a V-NAR from an immunized spiny dogfish shark, named E06, that binds specifically and with high affinity to human, mouse, and rat serum albumins. Humanization of E06 was carried out by converting over 60% of non-complementarity-determining region residues to those of a human germ line Vκ1 sequence, DPK9. The resulting huE06 molecules have largely retained the specificity and affinity of antigen binding of the parental V-NAR. Crystal structures of the shark E06 and its humanized variant (huE06 v1.1) in complex with human serum albumin (HSA) were determined at 3- and 2.3-Å resolution, respectively. The huE06 v1.1 molecule retained all but one amino acid residues involved in the binding site for HSA. Structural analysis of these V-NARs has revealed an unusual variable domain-antigen interaction. E06 interacts with HSA in an atypical mode that utilizes extensive framework contacts in addition to complementarity-determining regions that has not been seen previously in V-NARs. On the basis of the structure, the roles of various elements of the molecule are described with respect to antigen binding and V-NAR stability. This information broadens the general understanding of antigen recognition and provides a framework for further design and humanization of shark IgNARs.
Collapse
Affiliation(s)
- Oleg V Kovalenko
- Global Biotherapeutics Technologies, Pfizer Research and Development, Cambridge, Massachusetts 02140, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Fenn S, Schiller CB, Griese JJ, Duerr H, Imhof-Jung S, Gassner C, Moelleken J, Regula JT, Schaefer W, Thomas M, Klein C, Hopfner KP, Kettenberger H. Crystal structure of an anti-Ang2 CrossFab demonstrates complete structural and functional integrity of the variable domain. PLoS One 2013; 8:e61953. [PMID: 23613981 PMCID: PMC3629102 DOI: 10.1371/journal.pone.0061953] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 03/15/2013] [Indexed: 11/18/2022] Open
Abstract
Bispecific antibodies are considered as a promising class of future biotherapeutic molecules. They comprise binding specificities for two different antigens, which may provide additive or synergistic modes of action. There is a wide variety of design alternatives for such bispecific antibodies, including the “CrossMab” format. CrossMabs contain a domain crossover in one of the antigen-binding (Fab) parts, together with the “knobs-and-holes” approach, to enforce the correct assembly of four different polypeptide chains into an IgG-like bispecific antibody. We determined the crystal structure of a hAng-2-binding Fab in its crossed and uncrossed form and show that CH1-CL-domain crossover does not induce significant perturbations of the structure and has no detectable influence on target binding.
Collapse
Affiliation(s)
- Sebastian Fenn
- Large Molecule Research, Pharma Research and Early Development (pRED), Roche Dignostics GmbH, Penzberg, Germany
| | - Christian B. Schiller
- Department of Biochemistry, Gene Center, Ludwig Maximilians University Munich, Munich, Germany
| | - Julia J. Griese
- Department of Biochemistry, Gene Center, Ludwig Maximilians University Munich, Munich, Germany
| | - Harald Duerr
- Large Molecule Research, Pharma Research and Early Development (pRED), Roche Dignostics GmbH, Penzberg, Germany
| | - Sabine Imhof-Jung
- Large Molecule Research, Pharma Research and Early Development (pRED), Roche Dignostics GmbH, Penzberg, Germany
| | - Christian Gassner
- Large Molecule Research, Pharma Research and Early Development (pRED), Roche Dignostics GmbH, Penzberg, Germany
| | - Joerg Moelleken
- Large Molecule Research, Pharma Research and Early Development (pRED), Roche Dignostics GmbH, Penzberg, Germany
| | - Joerg Thomas Regula
- Large Molecule Research, Pharma Research and Early Development (pRED), Roche Dignostics GmbH, Penzberg, Germany
| | - Wolfgang Schaefer
- Large Molecule Research, Pharma Research and Early Development (pRED), Roche Dignostics GmbH, Penzberg, Germany
| | - Markus Thomas
- Discovery Oncology, Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany
| | - Christian Klein
- Discovery Oncology; Pharma Research and Early Development (pRED); Roche Glycart AG, Schlieren, Switzerland
| | - Karl-Peter Hopfner
- Department of Biochemistry, Gene Center, Ludwig Maximilians University Munich, Munich, Germany
- * E-mail: (KH); (HK)
| | - Hubert Kettenberger
- Large Molecule Research, Pharma Research and Early Development (pRED), Roche Dignostics GmbH, Penzberg, Germany
- * E-mail: (KH); (HK)
| |
Collapse
|
146
|
Tiller T, Schuster I, Deppe D, Siegers K, Strohner R, Herrmann T, Berenguer M, Poujol D, Stehle J, Stark Y, Heßling M, Daubert D, Felderer K, Kaden S, Kölln J, Enzelberger M, Urlinger S. A fully synthetic human Fab antibody library based on fixed VH/VL framework pairings with favorable biophysical properties. MAbs 2013; 5:445-70. [PMID: 23571156 DOI: 10.4161/mabs.24218] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
This report describes the design, generation and testing of Ylanthia, a fully synthetic human Fab antibody library with 1.3E+11 clones. Ylanthia comprises 36 fixed immunoglobulin (Ig) variable heavy (VH)/variable light (VL) chain pairs, which cover a broad range of canonical complementarity-determining region (CDR) structures. The variable Ig heavy and Ig light (VH/VL) chain pairs were selected for biophysical characteristics favorable to manufacturing and development. The selection process included multiple parameters, e.g., assessment of protein expression yield, thermal stability and aggregation propensity in fragment antigen binding (Fab) and IgG1 formats, and relative Fab display rate on phage. The framework regions are fixed and the diversified CDRs were designed based on a systematic analysis of a large set of rearranged human antibody sequences. Care was taken to minimize the occurrence of potential posttranslational modification sites within the CDRs. Phage selection was performed against various antigens and unique antibodies with excellent biophysical properties were isolated. Our results confirm that quality can be built into an antibody library by prudent selection of unmodified, fully human VH/VL pairs as scaffolds.
Collapse
|
147
|
Aggregates, crystals, gels, and amyloids: intracellular and extracellular phenotypes at the crossroads of immunoglobulin physicochemical property and cell physiology. Int J Cell Biol 2013; 2013:604867. [PMID: 23533417 PMCID: PMC3603282 DOI: 10.1155/2013/604867] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 01/27/2013] [Indexed: 12/20/2022] Open
Abstract
Recombinant immunoglobulins comprise an important class of human therapeutics. Although specific immunoglobulins can be purposefully raised against desired antigen targets by various methods, identifying an immunoglobulin clone that simultaneously possesses potent therapeutic activities and desirable manufacturing-related attributes often turns out to be challenging. The variable domains of individual immunoglobulins primarily define the unique antigen specificities and binding affinities inherent to each clone. The primary sequence of the variable domains also specifies the unique physicochemical properties that modulate various aspects of individual immunoglobulin life cycle, starting from the biosynthetic steps in the endoplasmic reticulum, secretory pathway trafficking, secretion, and the fate in the extracellular space and in the endosome-lysosome system. Because of the diverse repertoire of immunoglobulin physicochemical properties, some immunoglobulin clones' intrinsic properties may manifest as intriguing cellular phenotypes, unusual solution behaviors, and serious pathologic outcomes that are of scientific and clinical importance. To gain renewed insights into identifying manufacturable therapeutic antibodies, this paper catalogs important intracellular and extracellular phenotypes induced by various subsets of immunoglobulin clones occupying different niches of diverse physicochemical repertoire space. Both intrinsic and extrinsic factors that make certain immunoglobulin clones desirable or undesirable for large-scale manufacturing and therapeutic use are summarized.
Collapse
|
148
|
Somatic hypermutation maintains antibody thermodynamic stability during affinity maturation. Proc Natl Acad Sci U S A 2013; 110:4261-6. [PMID: 23440204 DOI: 10.1073/pnas.1301810110] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Somatic hypermutation and clonal selection lead to B cells expressing high-affinity antibodies. Here we show that somatic mutations not only play a critical role in antigen binding, they also affect the thermodynamic stability of the antibody molecule. Somatic mutations directly involved in antigen recognition by antibody 93F3, which binds a relatively small hapten, reduce the melting temperature compared with its germ-line precursor by up to 9 °C. The destabilizing effects of these mutations are compensated by additional somatic mutations located on surface loops distal to the antigen binding site. Similarly, somatic mutations enhance both the affinity and thermodynamic stability of antibody OKT3, which binds the large protein antigen CD3. Analysis of the crystal structures of 93F3 and OKT3 indicates that these somatic mutations modulate antibody stability primarily through the interface of the heavy and light chain variable domains. The historical view of antibody maturation has been that somatic hypermutation and subsequent clonal selection increase antigen-antibody specificity and binding energy. Our results suggest that this process also optimizes protein stability, and that many peripheral mutations that were considered to be neutral are required to offset deleterious effects of mutations that increase affinity. Thus, the immunological evolution of antibodies recapitulates on a much shorter timescale the natural evolution of enzymes in which function and thermodynamic stability are simultaneously enhanced through mutation and selection.
Collapse
|
149
|
Mahon CM, Lambert MA, Glanville J, Wade JM, Fennell BJ, Krebs MR, Armellino D, Yang S, Liu X, O'Sullivan CM, Autin B, Oficjalska K, Bloom L, Paulsen J, Gill D, Damelin M, Cunningham O, Finlay WJJ. Comprehensive interrogation of a minimalist synthetic CDR-H3 library and its ability to generate antibodies with therapeutic potential. J Mol Biol 2013; 425:1712-30. [PMID: 23429058 DOI: 10.1016/j.jmb.2013.02.015] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 01/31/2013] [Accepted: 02/11/2013] [Indexed: 11/17/2022]
Abstract
We have generated large libraries of single-chain Fv antibody fragments (>10(10) transformants) containing unbiased amino acid diversity that is restricted to the central combining site of the stable, well-expressed DP47 and DPK22 germline V-genes. Library WySH2A was constructed to examine the potential for synthetic complementarity-determining region (CDR)-H3 diversity to act as the lone source of binding specificity. Library WySH2B was constructed to assess the necessity for diversification in both the H3 and L3. Both libraries provided diverse, specific antibodies, yielding a total of 243 unique hits against 7 different targets, but WySH2B produced fewer hits than WySH2A when selected in parallel. WySH2A also consistently produced hits of similar quality to WySH2B, demonstrating that the diversification of the CDR-L3 reduces library fitness. Despite the absence of deliberate bias in the library design, CDR length was strongly associated with the number of hits produced, leading to a functional loop length distribution profile that mimics the biases observed in the natural repertoire. A similar trend was also observed for the CDR-L3. After target selections, several key amino acids were enriched in the CDR-H3 (e.g., small and aromatic residues) while others were reduced (e.g., strongly charged residues) in a manner that was specific to position, preferentially occurred in CDR-H3 stem positions, and tended towards residues associated with loop stabilization. As proof of principle for the WySH2 libraries to produce viable lead candidate antibodies, 114 unique hits were produced against Delta-like ligand 4 (DLL4). Leads exhibited nanomolar binding affinities, highly specific staining of DLL4+ cells, and biochemical neutralization of DLL4-NOTCH1 interaction.
Collapse
Affiliation(s)
- Ciara M Mahon
- Pfizer, Global Biotherapeutics Technologies, Grange Castle Business Park, Clondalkin, Dublin 22, Ireland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Xu L, Kohli N, Rennard R, Jiao Y, Razlog M, Zhang K, Baum J, Johnson B, Tang J, Schoeberl B, Fitzgerald J, Nielsen U, Lugovskoy AA. Rapid optimization and prototyping for therapeutic antibody-like molecules. MAbs 2013; 5:237-54. [PMID: 23392215 PMCID: PMC3893234 DOI: 10.4161/mabs.23363] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Multispecific antibody-like molecules have the potential to advance the standard-of-care in many human diseases. The design of therapeutic molecules in this class, however, has proven to be difficult and, despite significant successes in preclinical research, only one trivalent antibody, catumaxomab, has demonstrated clinical utility. The challenge originates from the complexity of the design space where multiple parameters such as affinity, avidity, effector functions, and pharmaceutical properties need to be engineered in concurrent fashion to achieve the desired therapeutic efficacy. Here, we present a rapid prototyping approach that allows us to successfully optimize these parameters within one campaign cycle that includes modular design, yeast display of structure focused antibody libraries and high throughput biophysical profiling. We delineate this approach by presenting a design case study of MM-141, a tetravalent bispecific antibody targeting two compensatory signaling growth factor receptors: insulin-like growth factor 1 receptor (IGF-1R) and v-erb-b2 erythroblastic leukemia viral oncogene homolog 3 (ErbB3). A MM-141 proof-of-concept (POC) parent molecule did not meet initial design criteria due to modest bioactivity and poor stability properties. Using a combination of yeast display, structured-guided antibody design and library-scale thermal challenge assay, we discovered a diverse set of stable and active anti-IGF-1R and anti-ErbB3 single-chain variable fragments (scFvs). These optimized modules were reformatted to create a diverse set of full-length tetravalent bispecific antibodies. These re-engineered molecules achieved complete blockade of growth factor induced pro-survival signaling, were stable in serum, and had adequate activity and pharmaceutical properties for clinical development. We believe this approach can be readily applied to the optimization of other classes of bispecific or even multispecific antibody-like molecules.
Collapse
Affiliation(s)
- Lihui Xu
- Merrimack Pharmaceuticals, Inc. Cambridge, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|