1
|
Maeta S, Nakakido M, Matsuura H, Sakai N, Hirata K, Kuroda D, Fukunaga A, Tsumoto K. Arginine cluster introduction on framework region in anti-lysozyme antibody improved association rate constant by changing conformational diversity of CDR loops. Protein Sci 2023; 32:e4745. [PMID: 37550885 PMCID: PMC10461459 DOI: 10.1002/pro.4745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/30/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023]
Abstract
Antibodies are used for many therapeutic and biotechnological purposes. Because the affinity of an antibody to the antigen is critical for clinical efficacy of pharmaceuticals, many affinity maturation strategies have been developed. Although we previously reported an affinity maturation strategy in which the association rate of the antibody toward its antigen is improved by introducing a cluster of arginine residues into the framework region of the antibody, the detailed molecular mechanism responsible for this improvement has been unknown. In this study, we introduced five arginine residues into an anti-hen egg white lysozyme antibody (HyHEL10) Fab fragment to create the R5-mutant and comprehensively characterized the interaction between antibody and antigen using thermodynamic analysis, X-ray crystallography, and molecular dynamics (MD) simulations. Our results indicate that introduction of charged residues strongly enhanced the association rate, as previously reported, and the antibody-antigen complex structure was almost the same for the R5-mutant and wild-type Fabs. The MD simulations indicate that the mutation increased conformational diversity in complementarity-determining region loops and thereby enhanced the association rate. These observations provide the molecular basis of affinity maturation by R5 mutation.
Collapse
Affiliation(s)
- Shingo Maeta
- Bio‐Diagnostic Reagent Technology CenterSysmex CorporationKobeJapan
- Department of Bioengineering, School of EngineeringThe University of TokyoTokyoJapan
| | - Makoto Nakakido
- Department of Bioengineering, School of EngineeringThe University of TokyoTokyoJapan
- Department of Chemistry and Biotechnology, Graduate School of EngineeringThe University of TokyoTokyoJapan
| | - Hiroaki Matsuura
- Life Science Research Infrastructure Group, RIKEN SPring‐8 CenterSaitamaJapan
| | - Naoki Sakai
- Life Science Research Infrastructure Group, RIKEN SPring‐8 CenterSaitamaJapan
| | - Kunio Hirata
- Life Science Research Infrastructure Group, RIKEN SPring‐8 CenterSaitamaJapan
| | - Daisuke Kuroda
- Department of Bioengineering, School of EngineeringThe University of TokyoTokyoJapan
- Department of Chemistry and Biotechnology, Graduate School of EngineeringThe University of TokyoTokyoJapan
- Research Center for Drug and Vaccine DevelopmentNational Institute of Infectious DiseasesTokyoJapan
| | - Atsushi Fukunaga
- Bio‐Diagnostic Reagent Technology CenterSysmex CorporationKobeJapan
| | - Kouhei Tsumoto
- Department of Bioengineering, School of EngineeringThe University of TokyoTokyoJapan
- Department of Chemistry and Biotechnology, Graduate School of EngineeringThe University of TokyoTokyoJapan
- Institute of Medical ScienceThe University of TokyoTokyoJapan
| |
Collapse
|
2
|
Khatib SE, Salla M. The mosaic puzzle of the therapeutic monoclonal antibodies and antibody fragments - A modular transition from full-length immunoglobulins to antibody mimetics. Leuk Res Rep 2022; 18:100335. [PMID: 35832747 PMCID: PMC9272380 DOI: 10.1016/j.lrr.2022.100335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 06/09/2022] [Accepted: 06/25/2022] [Indexed: 01/07/2023] Open
Abstract
The use of monoclonal antibodies represents an important and efficient diagnostic and therapeutic tool in disease management and modern science but remains limited by several factors including the uneven distribution in diseased tissues as well as undesired activation of side immune reactions. Major scientific advancements including Recombinant DNA Technology, Hybridoma Technology, and Polymerase Chain Reaction have considerably impacted the use of monoclonal antibodies providing technical and effective solutions to overcome the shortcomings encountered with conventional antibodies. Initially, the introduction of antibody fragments allowed a more uniform and deeper penetration of the targeted tissue and reduced unwanted activation of Fc-mediated immune reactions. On another level, the immunogenicity of murine-derived antibodies was overcome by humanizing their encoding genes with specific sequences of human origin andtransgenic mice able to synthesize fully human antibodies were successfully created. Moreover, the advancement of genetic engineering techniques supported by the modular structure of antibody coding genes paved the way for the development of a new generation of antibody fragments with a wide spectrum of monospecific and bispecific agents. These later could be monovalent, bivalent, or multivalent, and either expressed as a single chain, assembled in multimeric forms or stringed in tandem. This has conferred improved affinity, stability, and solubility to antibody targetting. Lately, a new array of monoclonal antibody fragments was introduced with the engineering of nanobody and antibody mimetics as non-immunoglobulin-derived fragments with promising diagnostic and therapeutic applications. In this review, we decipher the molecular basis of monoclonal antibody engineering with a detailed screening of the antibody derivatives that provides new perspectives to expand the use of monoclonal fragments into previously unexplored fields.
Collapse
Affiliation(s)
- Sami El Khatib
- Lebanese International University, Department of Biomedical Sciences, Bekaa Campus, Khiyara, West Bekaa, Lebanon
| | - Mohamed Salla
- University of Alberta. Biochemistry Department, Faculty of Medicine and Dentistry,116St & 85 Ave, Edmonton, AB, T6G 2R3, Canada
| |
Collapse
|
3
|
Farhadi T, Fakharian A, Hashemian SM. Affinity Improvement of a Humanized Antiviral Antibody by Structure-Based Computational Design. Int J Pept Res Ther 2017. [DOI: 10.1007/s10989-017-9660-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
4
|
Swindells MB, Porter CT, Couch M, Hurst J, Abhinandan KR, Nielsen JH, Macindoe G, Hetherington J, Martin ACR. abYsis: Integrated Antibody Sequence and Structure-Management, Analysis, and Prediction. J Mol Biol 2016; 429:356-364. [PMID: 27561707 DOI: 10.1016/j.jmb.2016.08.019] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/08/2016] [Accepted: 08/15/2016] [Indexed: 01/20/2023]
Abstract
abYsis is a web-based antibody research system that includes an integrated database of antibody sequence and structure data. The system can be interrogated in numerous ways-from simple text and sequence searches to sophisticated queries that apply 3D structural constraints. The publicly available version includes pre-analyzed sequence data from the European Molecular Biology Laboratory European Nucleotide Archive (EMBL-ENA) and Kabat as well as structure data from the Protein Data Bank. A researcher's own sequences can also be analyzed through the web interface. A defining characteristic of abYsis is that the sequences are automatically numbered with a series of popular schemes such as Kabat and Chothia and then annotated with key information such as complementarity-determining regions and potential post-translational modifications. A unique aspect of abYsis is a set of residue frequency tables for each position in an antibody, allowing "unusual residues" (those rarely seen at a particular position) to be highlighted and decisions to be made on which mutations may be acceptable. This is especially useful when comparing antibodies from different species. abYsis is useful for any researcher specializing in antibody engineering, especially those developing antibodies as drugs. abYsis is available at www.abysis.org.
Collapse
Affiliation(s)
| | - Craig T Porter
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK
| | - Matthew Couch
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK
| | - Jacob Hurst
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK
| | - K R Abhinandan
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK
| | - Jens H Nielsen
- Research Software Development Group, Research IT Services, University College London, Gower Street, London WC1E 6BT, UK
| | - Gary Macindoe
- Research Software Development Group, Research IT Services, University College London, Gower Street, London WC1E 6BT, UK
| | - James Hetherington
- Research Software Development Group, Research IT Services, University College London, Gower Street, London WC1E 6BT, UK
| | - Andrew C R Martin
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
5
|
Shaw S, Bourne T, Meier C, Carrington B, Gelinas R, Henry A, Popplewell A, Adams R, Baker T, Rapecki S, Marshall D, Moore A, Neale H, Lawson A. Discovery and characterization of olokizumab: a humanized antibody targeting interleukin-6 and neutralizing gp130-signaling. MAbs 2014; 6:774-82. [PMID: 24670876 PMCID: PMC4011921 DOI: 10.4161/mabs.28612] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Interleukin-6 (IL-6) is a critical regulator of the immune system and has been widely implicated in autoimmune disease. Here, we describe the discovery and characterization of olokizumab, a humanized antibody to IL-6. Data from structural biology, cell biology and primate pharmacology demonstrate the therapeutic potential of targeting IL-6 at “Site 3”, blocking the interaction with the signaling co-receptor gp130.
Collapse
|
6
|
Shaw S, Bourne T, Meier C, Carrington B, Gelinas R, Henry A, Popplewell A, Adams R, Baker T, Rapecki S, Marshall D, Moore A, Neale H, Lawson A. Discovery and characterization of olokizumab: a humanized antibody targeting interleukin-6 and neutralizing gp130-signaling. MAbs 2014. [PMID: 24670876 DOI: 10.4161/mabs.28612;] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Interleukin-6 (IL-6) is a critical regulator of the immune system and has been widely implicated in autoimmune disease. Here, we describe the discovery and characterization of olokizumab, a humanized antibody to IL-6. Data from structural biology, cell biology and primate pharmacology demonstrate the therapeutic potential of targeting IL-6 at "Site 3", blocking the interaction with the signaling co-receptor gp130.
Collapse
|
7
|
Kiyoshi M, Caaveiro JMM, Miura E, Nagatoishi S, Nakakido M, Soga S, Shirai H, Kawabata S, Tsumoto K. Affinity improvement of a therapeutic antibody by structure-based computational design: generation of electrostatic interactions in the transition state stabilizes the antibody-antigen complex. PLoS One 2014; 9:e87099. [PMID: 24475232 PMCID: PMC3903617 DOI: 10.1371/journal.pone.0087099] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 12/19/2013] [Indexed: 12/18/2022] Open
Abstract
The optimization of antibodies is a desirable goal towards the development of better therapeutic strategies. The antibody 11K2 was previously developed as a therapeutic tool for inflammatory diseases, and displays very high affinity (4.6 pM) for its antigen the chemokine MCP-1 (monocyte chemo-attractant protein-1). We have employed a virtual library of mutations of 11K2 to identify antibody variants of potentially higher affinity, and to establish benchmarks in the engineering of a mature therapeutic antibody. The most promising candidates identified in the virtual screening were examined by surface plasmon resonance to validate the computational predictions, and to characterize their binding affinity and key thermodynamic properties in detail. Only mutations in the light-chain of the antibody are effective at enhancing its affinity for the antigen in vitro, suggesting that the interaction surface of the heavy-chain (dominated by the hot-spot residue Phe101) is not amenable to optimization. The single-mutation with the highest affinity is L-N31R (4.6-fold higher affinity than wild-type antibody). Importantly, all the single-mutations showing increase affinity incorporate a charged residue (Arg, Asp, or Glu). The characterization of the relevant thermodynamic parameters clarifies the energetic mechanism. Essentially, the formation of new electrostatic interactions early in the binding reaction coordinate (transition state or earlier) benefits the durability of the antibody-antigen complex. The combination of in silico calculations and thermodynamic analysis is an effective strategy to improve the affinity of a matured therapeutic antibody.
Collapse
Affiliation(s)
- Masato Kiyoshi
- Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Jose M. M. Caaveiro
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory of Medical Proteomics, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Eri Miura
- Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Satoru Nagatoishi
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Makoto Nakakido
- Laboratory of Medical Proteomics, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Shinji Soga
- Molecular Medicine Research Laboratories, Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Hiroki Shirai
- Molecular Medicine Research Laboratories, Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Shigeki Kawabata
- Molecular Medicine Research Laboratories, Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Kouhei Tsumoto
- Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory of Medical Proteomics, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
8
|
Effect of glucose feeding on the glycosylation quality of antibody produced by a human cell line, F2N78, in fed-batch culture. Appl Microbiol Biotechnol 2014; 98:3509-15. [DOI: 10.1007/s00253-013-5462-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 12/03/2013] [Accepted: 12/09/2013] [Indexed: 11/24/2022]
|
9
|
Hristodorov D, Fischer R, Linden L. With or without sugar? (A)glycosylation of therapeutic antibodies. Mol Biotechnol 2013; 54:1056-68. [PMID: 23097175 DOI: 10.1007/s12033-012-9612-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Antibodies and antibody-based drugs are currently the fastest-growing class of therapeutics. Over the last three decades, more than 30 therapeutic monoclonal antibodies and derivatives thereof have been approved for and successfully applied in diverse indication areas including cancer, organ transplants, autoimmune/inflammatory disorders, and cardiovascular disease. The isotype of choice for antibody therapeutics is human IgG, whose Fc region contains a ubiquitous asparagine residue (N297) that acts as an acceptor site for N-linked glycans. The nature of these glycans can decisively influence the therapeutic performance of a recombinant antibody, and their absence or modification can lead to the loss of Fc effector functions, greater immunogenicity, and unfavorable pharmacokinetic profiles. However, recent studies have shown that aglycosylated antibodies can be genetically engineered to display novel or enhanced effector functions and that favorable pharmacokinetic properties can be preserved. Furthermore, the ability to produce aglycosylated antibodies in lower eukaryotes and bacteria offers the potential to broaden and simplify the production platforms and avoid the problem of antibody heterogeneity, which occurs when mammalian cells are used for production. In this review, we discuss the importance of Fc glycosylation focusing on the use of aglycosylated and glyco-engineered antibodies as therapeutic proteins.
Collapse
Affiliation(s)
- Dmitrij Hristodorov
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Forckenbeckstrasse 6, 52074 Aachen, Germany
| | | | | |
Collapse
|
10
|
Figini M, Orlandi R. New Techniques for the Production of Therapeutic Recombinant Human Monoclonal Antibodies. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/bf03259294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
11
|
Jung ST, Kang TH, Kelton W, Georgiou G. Bypassing glycosylation: engineering aglycosylated full-length IgG antibodies for human therapy. Curr Opin Biotechnol 2011; 22:858-67. [PMID: 21420850 DOI: 10.1016/j.copbio.2011.03.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 03/01/2011] [Accepted: 03/02/2011] [Indexed: 11/17/2022]
Abstract
In recent years a number of aglycosylated therapeutic antibodies have entered the clinic. The clinical evaluation of these antibodies has served to dispel concerns that the absence of the ubiquitous N297 glycan in the Fc of IgG might result in immunogenicity, poor in vivo stability or unfavorable pharmacokinetics. Importantly, recent studies have now demonstrated that aglycosylated antibodies can be engineered to display novel effector functions and mechanisms of action that do not appear to be possible with their glycosylated counterparts. Moreover, the ability to manufacture aglycosylated antibodies in lower eukaryotes or in bacteria provides significant bioprocessing advantages in terms of shorter bioprocess development and running times and by completely bypassing the problems associated with the glycan heterogeneity of conventional antibodies. These advantages are poised to catapult aglycosylated antibodies to the forefront of protein therapeutics.
Collapse
Affiliation(s)
- Sang Taek Jung
- Department of Chemical Engineering, University of Texas, Austin, TX 78712, United States
| | | | | | | |
Collapse
|
12
|
Ni M, Yu B, Huang Y, Tang Z, Lei P, Shen X, Xin W, Zhu H, Shen G. Homology modelling and bivalent single-chain Fv construction of anti-HepG2 single-chain immunoglobulin Fv fragments from a phage display library. J Biosci 2009; 33:691-7. [PMID: 19179757 DOI: 10.1007/s12038-008-0089-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
We prepared single-chain immunoglobulin Fv fragments (scFv) SLH10 specific for the HepG2 cell line after biopanning from a large human-naive phage display library (Griffin. 1 Library). The three-dimensional (3D) structure of SLH10 was modelled by the Insight II molecule simulation software.The structure was refined using the molecular dynamics method.The structures with the least steric clashes and lowest energy were determined finally. The optimized structures of heavy (VH) and light (VL) variable chains of SLH10 scFv were obtained.Then SLH10 bivalent single-chain Fv (BsFv) was constructed that would be suitable for high-affinity targeting.SLH10 BsFv was generated by linking scFvs together and identified by sequencing. Its expression products were confirmed by western blot analysis.The relative molecular masses of scFv and BsFv were approximately 30 kDa and 60 kDa,respectively. Flow cytometry revealed that SLH10 BsFv bound the selected cell lines with greater signal intensity than the parental scFv. The improved antigen binding of SLH10 BsFv may be useful for immunodiagnostics or targeted gene therapy for liver cancer.
Collapse
Affiliation(s)
- Ming Ni
- Department of Infectious Diseases,Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology,Wuhan 430030, China
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Tateishi Y, Nishimichi N, Horiuchi H, Furusawa S, Matsuda H. Construction of chicken-mouse chimeric antibody and immunogenicity in mice. J Vet Med Sci 2008; 70:397-400. [PMID: 18460836 DOI: 10.1292/jvms.70.397] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Chicken monoclonal antibodies are potentially useful for diagnostic research and have clinical applications, as chicken show higher potential for antibody production with mammalian-conserved biological molecules. However, the applications of chicken antibodies are limited because of their immunogenicity in mammals. To overcome this problem, we have constructed a chicken-mouse chimeric antibody containing the chicken variable region and the mouse constant region. This chimeric antibody retained similar binding affinities as the parental chicken antibody. The chimeric antibody was also producible as an ascitic antibody in BALB/c mice. Furthermore, when the chimeric antibody was administered to mice, it did not provoke the mouse anti-chicken antibody response. These results indicate that the chimeric antibody is suitable for application to preclinical mouse studies.
Collapse
Affiliation(s)
- Yoshiko Tateishi
- Laboratory of Immunobiology, Department of Molecular and Applied Bioscience, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Japan
| | | | | | | | | |
Collapse
|
14
|
Sandborn WJ, Hanauer SB. Antitumor necrosis factor therapy for inflammatory bowel disease: A review of agents, pharmacology, clinical results, and safety. Inflamm Bowel Dis 2007. [DOI: 10.1002/ibd.3780050209] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
|
15
|
Abstract
Maximizing the expression yields of recombinant whole antibodies and antibody fragments such as Fabs, single-chain Fvs and single-domain antibodies is highly desirable since it leads to lower production costs. Various eukaryotic and prokaryotic expression systems have been exploited to accommodate antibody expression but Escherichia coli systems have enjoyed popularity, in particular with respect to antibody fragments, because of their low cost and convenience. In many instances, product yields have been less than adequate and intrinsic and extrinsic variables have been investigated in an effort to improve yields. This review deals with various aspects of antibody expression in E. coli with a particular focus on single-domain antibodies.
Collapse
Affiliation(s)
- Mehdi Arbabi-Ghahroudi
- Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario.
| | | | | |
Collapse
|
16
|
Lohse L, Nielsen J, Kamstrup S, Oleksiewicz MB, Eriksen L. Porcine humoral immune responses to multiple injections of murine monoclonal antibodies. APMIS 2005; 113:489-96. [PMID: 16086818 DOI: 10.1111/j.1600-0463.2005.apm_62.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In humans and cattle, multiple injections of murine monoclonal antibodies (m-mAbs) induce anti-mouse antibody responses. The objectives of the present study were to investigate whether a similar response could be seen when pigs were subjected to m-mAb therapy, and to study the kinetics of such a response. In two separate animal experiments, long-term treatment was performed with m-mAbs at low-dose levels and therapeutic levels, respectively. Two specific m-mAbs that recognized cognate antigen in the pigs (CD4 and CD8 surface antigens on T-lymphocytes) and two irrelevant control m-mAbs having no cognate antigen in the pigs were used. Enzyme-linked immunosorbent assays (ELISA) were used to quantitate the circulating m-mAbs, as well as the induced pig anti-mouse antibodies (PAMA), in serum samples from m-mAb-treated pigs. As expected, we generally saw vigorous PAMA responses within 10 days after the start of m-mAb treatment with the specific m-mAbs. However, the different mAbs showed striking differences in the kinetics and levels of PAMA responses, differences that might be ascribed to the m-mAb formulation and epitope specificity. In conclusion, treatment of pigs with m-mAbs against T-cell surface antigens induced rapid PAMA responses. This may influence and possibly decrease the effect of the m-mAb treatment by narrowing the time period where m-mAbs can efficiently be used for cell depletion.
Collapse
Affiliation(s)
- Louise Lohse
- Danish Institute for Food and Veterinary Research, Department of Virology, Lindholm, Kalvehave, Denmark
| | | | | | | | | |
Collapse
|
17
|
El-Hariry I, Pignatelli M. Adhesion molecules: opportunities for modulation and a paradigm for novel therapeutic approaches in cancer. Expert Opin Investig Drugs 2005; 6:1465-78. [PMID: 15989513 DOI: 10.1517/13543784.6.10.1465] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In the past decade, there have been major advances in the elucidation of processes underlying tumour invasion and metastasis, in which adhesion molecules play a critical role. These advances have revolutionised our ability to devise novel approaches for cancer treatment. This review gives an insight into the adhesion pathways, and highlights the current status of adhesion molecules as potential therapeutic targets.
Collapse
Affiliation(s)
- I El-Hariry
- Cell Adhesion Laboratory, Department of Histopathology, Royal Postgraduate Medical School, Hammersmith Hospital, Du Cane Road, London, W12 ONN, UK
| | | |
Collapse
|
18
|
Hubbard RE. 3D structure and the drug-discovery process. MOLECULAR BIOSYSTEMS 2005. [DOI: 10.1039/b514814f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
19
|
Berger M, Shankar V, Vafai A. Therapeutic applications of monoclonal antibodies. Am J Med Sci 2002; 324:14-30. [PMID: 12120821 PMCID: PMC7093874 DOI: 10.1097/00000441-200207000-00004] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2001] [Revised: 12/21/2001] [Indexed: 12/31/2022]
Abstract
Researchers have sought therapeutic applications for monoclonal antibodies since their development in 1975. However, murine-derived monoclonal antibodies may cause an immunogenic response in human patients, reducing their therapeutic efficacy. Chimeric and humanized antibodies have been developed that are less likely to provoke an immune reaction in human patients than are murine-derived antibodies. Antibody fragments, bispecific antibodies, and antibodies produced through the use of phage display systems and genetically modified plants and animals may aid researchers in developing new uses for monoclonal antibodies in the treatment of disease. Monoclonal antibodies may have a number of promising potential therapeutic applications in the treatment of asthma, autoimmune diseases, cancer, poisoning, septicemia, substance abuse, viral infections, and other diseases.
Collapse
Affiliation(s)
- Mitchell Berger
- Emory University School of Public Health, Atlanta, Georgia, USA
| | | | | |
Collapse
|
20
|
|
21
|
Abiko Y. Passive immunization against dental caries and periodontal disease: development of recombinant and human monoclonal antibodies. CRITICAL REVIEWS IN ORAL BIOLOGY AND MEDICINE : AN OFFICIAL PUBLICATION OF THE AMERICAN ASSOCIATION OF ORAL BIOLOGISTS 2002; 11:140-58. [PMID: 12002812 DOI: 10.1177/10454411000110020101] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Indigenous micro-organisms in the oral cavity can cause two major diseases, dental caries and periodontal diseases. There is neither agreement nor consensus as to the actual mechanisms of pathogenesis of the specific virulence factors of these micro-organisms. The complexity of the bacterial community in dental plaque has made it difficult for the single bacterial agent of dental caries to be determined. However, there is considerable evidence that Streptococcus mutans is implicated as the primary causative organism of dental caries, and the cell-surface protein antigen (SA I/II) as well as glucosyltransferases (GTFs) produced by S. mutans appear to be major colonization factors. Various forms of periodontal diseases are closely associated with specific subgingival bacteria. Porphyromonas gingivalis has been implicated as an important etiological agent of adult periodontitis. Adherence of bacteria to host tissues is a prerequisite for colonization and one of the important steps in the disease process. Bacterial coaggregation factors and hemagglutinins likely play major roles in colonization in the subgingival area. Emerging evidence suggests that inhibition of these virulence factors may protect the host against caries and periodontal disease. Active and passive immunization approaches have been developed for immunotherapy of these diseases. Recent advances in mucosal immunology and the introduction of novel strategies for inducing mucosal immune responses now raise the possibility that effective and safe vaccines can be constructed. In this regard, some successful results have been reported in animal experimental models. Nevertheless, since the public at large might be skeptical about the seriousness of oral diseases, immunotherapy must be carried out with absolute safety. For this goal to be achieved, the development of safe antibodies for passive immunization is significant and important. In this review, salient advances in passive immunization against caries and periodontal diseases are summarized, and the biotechnological approaches for developing recombinant and human-type antibodies are introduced. Furthermore, our own attempts to construct single-chain variable fragments (ScFv) and human-type antibodies capable of neutralizing virulence factors are discussed.
Collapse
Affiliation(s)
- Y Abiko
- Department of Biochemistry, Nihon University School of Dentistry at Matsudo, Chiba, Japan.
| |
Collapse
|
22
|
|
23
|
Abstract
Antibodies are unique in their high affinity and specificity for a binding partner, a quality that has made them one of the most useful molecules for biotechnology and biomedical applications. The field of antibody engineering has changed rapidly in the past 10 years, fueled by novel technologies for the in vitro isolation of antibodies from combinatorial libraries and their functional expression in bacteria. This review presents an overview of the methods available for the de novo generation of human antibodies, for engineering antibodies with increased antigen affinity, and for the production of antibody fragments. Select applications of recombinant antibodies are also presented.
Collapse
Affiliation(s)
- J Maynard
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712, USA
| | | |
Collapse
|
24
|
Dillman RO. Monoclonal antibodies in the treatment of malignancy: basic concepts and recent developments. Cancer Invest 2002; 19:833-41. [PMID: 11768037 DOI: 10.1081/cnv-100107745] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Antibodies have long been considered to be potential anticancer agents because of their specificity for cell-membrane antigens. Applications of hybridoma and recombinant DNA technology have led to the production of unlimited quantities of clinical-grade murine, chimeric, and humanized monoclonal antibodies for clinical use. Whole antibodies may produce anticancer effects in conjunction with the immune system by interaction with complement proteins and/or effector cells via the Fc portion of the antibody molecule. Antibodies may also neutralize circulating ligands or block cell membrane receptors and thus interrupt ligand/receptor interactions and signal transduction that are associated with proliferative or anti-apoptotic effects. The anti-idiotype network cascade provides a rationale for antibodies as vaccine therapy. Antibodies may also serve as the guiding or targeting system for other cytotoxic pharmaceutical products such as (i) radiolabeled antibodies for radioimmunodetection and radioimmunotherapy; (ii) immunotoxins; (iii) chemotherapy/antibody conjugates; (iv) cytokine/antibody conjugates; and (v) immune cell/antibody conjugates. After years of anticipation, as of late 1999 there were four monoclonal antibodies that had been approved by the U.S. Food and Drug Administration based on activity against human malignancy, all of which are in widespread clinical use. Several other products are in various stages of clinical trial testing. Monoclonal antibodies have joined interferon-alpha, interleukin-2 (IL-2), and various hematopoietic growth factors as well-established components of biological therapy, the fourth modality of cancer treatment.
Collapse
Affiliation(s)
- R O Dillman
- Hoag Cancer Center, Newport Beach, California 92658, USA.
| |
Collapse
|
25
|
Abstract
PURPOSE The purpose of this article is to review the historical development and recent advances in the application of monoclonal antibodies for the treatment of lymphoma. OVERVIEW The history of clinical applications of monoclonal antibodies has been intertwined with that of lymphomas. The first report of a complete remission in 1981 described a patient with follicular lymphoma who was treated with a murine anti-idiotype antibody. Later that decade there appeared additional encouraging reports of radiolabeled monoclonal antibodies, immunotoxins, and other antibodies with antitumor effects against lymphoma and chronic lymphocytic leukemia. Monoclonal antibodies as a treatment of malignancy became reality in late 1997 when the US Food and Drug Administration approved the anti-CD20 chimeric monoclonal antibody rituximab for the treatment of B-cell lymphoma. Since that time an anti-CD25 monoclonal antibody (dacliximab) and an anti-CD25 immunotoxin fusion product (denileukin diftitox) have become clinically available. Several radio- labeled antibodies, including the murine anti-CD20 products (131)I-tositumomab and (90)Y-ibritumomab tiuxetan, are in advanced stages of clinical testing as are other unlabeled monoclonal antibodies with antilymphoma activity. Other antilymphoma immunotoxins that react with CD25, CD19, and CD22 also have shown promise. CLINICAL IMPLICATIONS The therapeutic arsenal against lymphoma has been significantly changed by the addition of these antibody products that are active as single agents, and are synergistic, additive, or both with other antilymphoma treatments.
Collapse
Affiliation(s)
- R O Dillman
- Laboratory and Clinical Research, Hoag Cancer Center, Newport Beach, California 92658, USA
| |
Collapse
|
26
|
Green LL. Antibody engineering via genetic engineering of the mouse: XenoMouse strains are a vehicle for the facile generation of therapeutic human monoclonal antibodies. J Immunol Methods 1999; 231:11-23. [PMID: 10648924 DOI: 10.1016/s0022-1759(99)00137-4] [Citation(s) in RCA: 114] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The major impediment to the development of murine monoclonal antibodies (mAbs) for therapy in humans has been the difficulty in reducing their potential immunogenicity. XenoMouse¿trade mark omitted¿ mice obviate this problem while retaining the relative ease of generating mAbs from a mouse. XenoMouse strains include germline-configured, megabase-sized YACs carrying portions of the human IgH and Igkappa loci, including the majority of the variable region repertoire, the genes for Cmicro, Cdelta and either Cgamma1, Cgamma2, or Cgamma4, as well as the cis elements required for their function. The IgH and Igkappa transgenes were bred onto a genetic background deficient in production of murine immunoglobulin. The large and complex human variable region repertoire encoded on the Ig transgenes in XenoMouse strains support the development of large peripheral B cell compartments and the generation of a diverse primary immune repertoire similar to that from adult humans. Immunization of XenoMouse mice with human antigens routinely results in a robust secondary immune response, which can ultimately be captured as a large panel of antigen-specific fully human IgGkappa mAbs of sub-nanomolar affinities. Monoclonal antibodies from XenoMouse animals have been shown to have therapeutic potential both in vitro and in vivo, and appear to have the pharmacokinetics of normal human antibodies based on human clinical trials. The utility of XenoMouse strains for the generation of large panels of high-affinity, fully human mAbs can be made available to researchers in the academic and private sectors, and should accelerate the development and application of mAbs as therapeutics for human disease.
Collapse
Affiliation(s)
- L L Green
- Abgenix, 7601 Dumbarton Circle, Fremont, CA 94555, USA.
| |
Collapse
|
27
|
Bruce CJ, Howard CJ, Thomas LH, Tempest PR, Taylor G. Depletion of bovine CD8+ T cells with chCC63, a chimaeric mouse-bovine antibody. Vet Immunol Immunopathol 1999; 71:215-31. [PMID: 10587302 DOI: 10.1016/s0165-2427(99)00098-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In order to investigate the role of T cells in immune responses to infectious pathogens, depletion of individual T cell subsets using monoclonal antibodies (mAbs) is commonly undertaken. Since most mAbs are of murine origin, such depletion studies in cattle are restricted by the bovine anti-mouse antibody (BAMA) response to the mouse mAbs used for the depletions. In this study, we describe the use of antibody engineering to overcome the BAMA response. The variable region cDNA from CC63, a monoclonal mouse anti-bovine CD8 antibody, has been expressed in conjunction with bovine constant region genes to produce a mouse-bovine chimaeric antibody (chCC63). Characterisation of chCC63 showed that the antibody contained a bovine constant region and specifically bound bovine CD8+ T cells. Furthermore, chCC63 blocked the binding of the original mouse antibody, CC63, and mediated complement-dependent lysis of bovine CD8+ cells in vitro. In vivo, chCC63 depleted calves of CD8+ T cells as effectively as CC63 and provoked a BAMA response that was about one-tenth of that seen with the mouse antibody.
Collapse
Affiliation(s)
- C J Bruce
- Institute for Animal Health, Compton, Newbury, Berks, UK
| | | | | | | | | |
Collapse
|
28
|
Dubowchik GM, Walker MA. Receptor-mediated and enzyme-dependent targeting of cytotoxic anticancer drugs. Pharmacol Ther 1999; 83:67-123. [PMID: 10511457 DOI: 10.1016/s0163-7258(99)00018-2] [Citation(s) in RCA: 198] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
This review is a survey of various approaches to targeting cytotoxic anticancer drugs to tumors primarily through biomolecules expressed by cancer cells or associated vasculature and stroma. These include monoclonal antibody immunoconjugates; enzyme prodrug therapies, such as antibody-directed enzyme prodrug therapy, gene-directed enzyme prodrug therapy, and bacterial-directed enzyme prodrug therapy; and metabolism-based therapies that seek to exploit increased tumor expression of, e.g., proteases, low-density lipoprotein receptors, hormones, and adhesion molecules. Following a discussion of factors that positively and negatively affect drug delivery to solid tumors, we concentrate on a mechanistic understanding of selective drug release or generation at the tumor site.
Collapse
Affiliation(s)
- G M Dubowchik
- Bristol-Myers Squibb Pharmaceutical Research Institute, Wallingford, CT 06492-7660, USA.
| | | |
Collapse
|
29
|
Sandborn WJ, Hanauer SB. Antitumor necrosis factor therapy for inflammatory bowel disease: a review of agents, pharmacology, clinical results, and safety. Inflamm Bowel Dis 1999; 5:119-33. [PMID: 10338381 DOI: 10.1097/00054725-199905000-00008] [Citation(s) in RCA: 255] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor necrosis factor-alpha (TNFalpha), a proinflammatory cytokine, plays an important role in the pathogenesis of inflammatory bowel disease (IBD). Biotechnology agents including a chimeric monoclonal anti-TNF antibody (infliximab), a humanized monoclonal anti-TNF antibody (CDP571), and a recombinant TNF receptor fusion protein (etanercept) have been used to inhibit TNFalpha activity. Controlled trials have demonstrated efficacy for infliximab in moderately to severely active Crohn's disease (CD) and fistulizing CD sufficient to justify recent U.S. Food and Drug Administration (FDA) approval. Additional trials have been completed in rheumatoid arthritis (RA). Similarly, preliminary controlled trials have suggested efficacy for CDP571 in active CD and RA. Larger controlled trials have demonstrated efficacy for etanercept in RA patients who have failed disease modifying antirheumatic drug (DMARD) therapy leading to FDA approval for RA. Toxicities observed with anti-TNF therapies have included formation of human antichimeric antibodies (HACA) with associated acute and delayed hypersensitivity infusion reactions, human antihuman antibodies (HAHAs), and formation of autoantibodies with rare instances of drug-induced lupus. Several cases of non-Hodgkin's lymphoma also has been described. Future studies should evaluate optimal timing and duration of anti-TNF therapy, the utility of adjuvant medical treatments during anti-TNF therapy, and evaluate long-term safety and efficacy of the various anti-TNF agents.
Collapse
Affiliation(s)
- W J Sandborn
- Inflammatory Bowel Disease Clinic, Mayo Clinic and Mayo Foundation, Rochester, Minnesota 55905, USA
| | | |
Collapse
|
30
|
Abstract
LEARNING OBJECTIVES Reading this article will increase the readers' knowledge of the biology of interleukin-5 (IL-5), an important cytokine. The immune and inflammatory responses of any organism are the basis of the defense mechanism ensuring its survival. The role of IL-5 in these processes, as well as in the pathogenesis of various diseases has been discussed along with the effects of various pharmacologic agents on the production and function of IL-5. DATA SOURCES A detailed literature search was performed. Studies considered relevant and important, in all languages, which involved humans and animals were used. STUDY SELECTION Information was obtained only from peer reviewed journals. RESULTS Interleukin-5 is normally produced by T-cells, mast cells, and eosinophils while Reed Sternberg and Epstein Barr virus (EBV) transformed cells also produce IL-5. Monoclonal antibodies (mAb) to IL-5 are potent inhibitors of IL-5 mediated tissue damage, secondary to eosinophil infiltration. The majority of the studies on IL-5 are preliminary, often the information is obtained from animal studies or in vitro systems and occasionally from pathologic tissue analysis. This along with the absence of confirmatory studies is a limiting factor. Nonetheless, the role of IL-5 in allergic and immunologic disease and asthma may be central to their pathogenesis. CONCLUSIONS Interleukin-5 is an important molecule that is participant to many processes that maintain health and are involved directly or indirectly in the pathogenesis of disease. Some pharmacologic agents can modify IL-5 production in vivo. Development of selective inhibitors of IL-5 may have a potential use for specific therapy of certain autoimmune, inflammatory, and neoplastic diseases.
Collapse
Affiliation(s)
- T Lalani
- Department of Oral Medicine and Diagnostic Sciences, Harvard School of Dental Medicine, Boston, Massachusetts 02112, USA
| | | | | |
Collapse
|
31
|
Papanastasiou D, Mamalaki A, Eliopoulos E, Poulas K, Liolitsas C, Tzartos SJ. Construction and characterization of a humanized single chain Fv antibody fragment against the main immunogenic region of the acetylcholine receptor. J Neuroimmunol 1999; 94:182-95. [PMID: 10376952 DOI: 10.1016/s0165-5728(98)00249-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The single chain Fv fragment of mAb198 (scFv198) directed against the main immunogenic region (MIR) of the nicotinic acetylcholine receptor (AChR), can efficiently protect the AChR in muscle cell cultures against the destructive activity of human myasthenic autoantibodies. Humanization of the scFv198 antibody fragment should prove useful for therapeutic application by reducing its immunogenicity. Framework sequences from human immunoglobulins homologous to the rat scFv198 sequences were selected and a totally synthetic humanized scFv198 antibody fragment was constructed in vitro. Humanized VH and VL domains were synthesized using two overlapping sets of 225 bases long oligonucleotides overlap extension and polymerase chain reaction (PCR), then assembled into a full-length gene by overlap extension of single-stranded DNA (ssDNA) fragments and PCR. The initial humanized antibody fragment had a very low affinity for the AChR. Molecular modeling was then performed and four residues from the framework regions (FR) of the humanized VH domain were selected to be replaced by the corresponding amino acid from the rat sequence. Three mutants were constructed by overlap extension, using PCR. The humanized variant containing replacements at VH residues 27, 29, 30 and 71 showed very good recovery of AChR binding activity; its binding affinities for Torpedo or human AChR (K(D): 8.5 or 323 nM, respectively) being only four times lower than those of the parental scFv198 (K(D): 2 or 80 nM, respectively). This variant was able to protect the human AChR against the binding of anti-MIR mAb and anti-alpha autoantibodies from a myasthenic patient. It was also able to protect AChR against antigenic modulation induced by the anti-MIR mAb198.
Collapse
Affiliation(s)
- D Papanastasiou
- Department of Biochemistry, Hellenic Pasteur Institute, Athens, Greece
| | | | | | | | | | | |
Collapse
|
32
|
Vaswani SK, Hamilton RG. Humanized antibodies as potential therapeutic drugs. Ann Allergy Asthma Immunol 1998; 81:105-15; quiz 115-6, 119. [PMID: 9723555 DOI: 10.1016/s1081-1206(10)62794-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Antibodies have been used therapeutically to treat a variety of clinical conditions. The introduction of monoclonal antibodies and more recently, engineered humanized antibodies has greatly refined and expanded the therapeutic potential of this modality of treatment. LEARNING OBJECTIVES To reinforce the reader's knowledge of the therapeutic application of antibody in the treatment for different diseases. More specifically, to enhance reader's understanding of basic methods employed in the production and clinical use of humanized antibodies. DATA SOURCE The MEDLINE database was used to review the humanized antibody related literature. CONCLUSION Humanized antibodies provide a novel approach for the treatment of a broad range of diseases. Expanded use will depend on improvement in their efficacy (avidity and specificity), demonstration of their safety, and reduction of their immunogenicity.
Collapse
Affiliation(s)
- S K Vaswani
- Johns Hopkins University School of Medicine, Division of Allergy and Clinical Immunology, Baltimore, Maryland, USA
| | | |
Collapse
|
33
|
Kolb AF, Siddell SG. Expression of a recombinant monoclonal antibody from a bicistronic mRNA. Hybridoma (Larchmt) 1997; 16:421-6. [PMID: 9388025 DOI: 10.1089/hyb.1997.16.421] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The variable regions of the murine monoclonal antibody A1, which effectively neutralizes the infection of susceptible cells by the murine hepatitis virus strain JHM, were cloned, sequenced, and expressed in mammalian cells as a functional recombinant antibody. To accomplish the concurrent synthesis of both antibody chains, the light- and heavy-chain-coding regions were inserted into a bicistronic expression cassette based on the encephalomyocarditis virus internal ribosomal entry site. The strategy of combining both coding regions in one bicistronic mRNA allows for the rapid isolation of cell clones expressing high levels of recombinant antibody.
Collapse
Affiliation(s)
- A F Kolb
- Institute of Virology and Immunology, University of Würzburg, Germany
| | | |
Collapse
|
34
|
Abstract
IL-5 is a prominent and perhaps an essential element in the induction of allergic inflammation in human asthma and other allergic diseases. Despite the strong biochemical and clinical correlates between lung eosinophilia and asthma, there is no clear understanding of how eosinophils exacerbate asthma. Antigen administration to sensitized animals produces eosinophilic infiltration that is very similar to that in man, and is prevented by administration of a neutralizing monoclonal antibody against IL-5. Mice in which the IL-5 gene is absent are unable to mount eosinophilic responses to antigen and do not sustain lung damage, but otherwise develop normally. The study of the biology of IL-5 has not only clarified the links between eosinophilia and airway hyperreactivity, but also strongly suggests that anti-IL-5 therapy may be an effective, safe, and novel way of treating human asthma and perhaps other eosinophilic diseases. There are many different potential approaches to the inhibition of IL-5, but the one most likely to provide "proof of principle" in "asthma in the wild" in man is a monoclonal antibody against IL-5.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/immunology
- Antibodies, Blocking/therapeutic use
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Asthma/immunology
- Asthma/therapy
- Dermatitis, Atopic/therapy
- Eosinophils/immunology
- Eosinophils/metabolism
- Guinea Pigs
- Haplorhini
- Humans
- Hypersensitivity, Immediate/immunology
- Hypersensitivity, Immediate/therapy
- Interleukin-5/antagonists & inhibitors
- Mice
- Mice, Knockout
- Nasal Polyps/therapy
- Rhinitis, Allergic, Perennial/immunology
- Rhinitis, Allergic, Perennial/therapy
Collapse
Affiliation(s)
- M Danzig
- Department of Clinical Research, Schering-Plough Research Institute, Kenilworth, NJ, USA
| | | |
Collapse
|
35
|
Ghetie MA, Podar EM, Ilgen A, Gordon BE, Uhr JW, Vitetta ES. Homodimerization of tumor-reactive monoclonal antibodies markedly increases their ability to induce growth arrest or apoptosis of tumor cells. Proc Natl Acad Sci U S A 1997; 94:7509-14. [PMID: 9207122 PMCID: PMC23852 DOI: 10.1073/pnas.94.14.7509] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/1997] [Indexed: 02/04/2023] Open
Abstract
Monoclonal antibodies (mAbs) that exert antitumor activity can do so by virtue of their effector function and/or their ability to signal growth arrest or cell death. In this study, we demonstrate that mAbs which have little or no signaling activity-i.e., anti-CD19, CD20, CD21, CD22 and Her-2-can become potent antitumor agents when they are converted into IgG-IgG homodimers. The homodimers exert antigrowth activity by signaling G0/G1 arrest or apoptosis, depending upon which cell surface molecule they bind. This activity is specific and, in the case of the anti-CD19 mAb, did not require an Fc portion. These results offer the possibility that homodimers of other tumor-reactive mAbs which have little antitumor activity as monomers might be potent, antitumor agents.
Collapse
Affiliation(s)
- M A Ghetie
- Cancer Immunobiology Center and Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | | | | | | | | | | |
Collapse
|
36
|
Construction of expression vectors and study on single-chain antibody and reshaping single-domain antibody against CD3. ACTA ACUST UNITED AC 1997; 40:270-6. [DOI: 10.1007/bf02879087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/1996] [Indexed: 10/22/2022]
|
37
|
Furebring C, Ohlin M, Pettersson S, Borrebaeck CA. Evaluation of novel control elements by construction of eukaryotic expression vectors. Gene X 1997; 188:191-8. [PMID: 9133591 DOI: 10.1016/s0378-1119(96)00791-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
A novel mammalian eukaryotic expression vector for the production of immunoglobulin heavy chain (IgH) genes has been designed. This expression vector contains the variable heavy chain (VH) promoter, the IgH intron enhancer (muE) and the IgH 3' enhancer (3'E). This construct, designated pTIF-1, was stably transfected into the myeloma cell line J558L. A fivefold increase in the expression level of a rearranged IgH gene was observed when using the pTIF-1 vector containing the 3'E compared to an expression vector lacking this enhancer. Interestingly, this positive effect on the expression level of the 3' enhancer appears to be position independent. The introduction of two recently identified Ig control elements, HS3 and HS4, to the vector cassette did not further elevate the expression level in the cell line tested. The pTIF-1 vector can be used for expression of any antibody specificity, using PCR amplification of the VDJ region of interest. Furthermore, the constant region can easily be exchanged, which further facilitates studies to dissect different effector functions of IgH constant genes.
Collapse
Affiliation(s)
- C Furebring
- Department of Immunotechnology, Lund University, Sweden
| | | | | | | |
Collapse
|
38
|
Koren E, Milotic F, Neethling FA, Cooper DKC. Neutralization of the Cytotoxic Effect of Anti-αGal Antibodies with Monoclonal Anti-idiotypic Antibodies. Xenotransplantation 1997. [DOI: 10.1007/978-3-642-60572-7_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
39
|
Froyen G, Billiau A. Potential therapeutic use of antibodies directed towards HuIFN-gamma. BIOTHERAPY (DORDRECHT, NETHERLANDS) 1997; 10:49-57. [PMID: 9261550 DOI: 10.1007/bf02678217] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
IFN-gamma is an important regulator of immune responses and inflammation. Studies in animal models of inflammation, autoimmunity, cancer, transplant rejection and delayed-type hypersensitivity have indicated that administration of antibodies against IFN-gamma can prevent the occurrence of diseases or alleviate disease manifestations. Therefore, it is speculated that such antibodies may have therapeutical efficacy in human diseases. Since animal-derived antibodies are immunogenic in patients several strategies are being developed in order to reduce or abolish this human anti-mouse antibody (HAMA) response. In our laboratory, we have constructed a single-chain variable fragment (scFv) derived from a mouse antibody with neutralizing potential for human IFN-gamma. A scFv consists of only variable domains tethered together by a flexible linker. The scFv was demonstrated to neutralize the antiviral activity of HuIFN-gamma in vitro and therefore might be considered as a candidate for human therapy.
Collapse
Affiliation(s)
- G Froyen
- Rega Institute for Medical Research, University of Leuven, Belgium
| | | |
Collapse
|
40
|
Keck PC, Huston JS. Symmetry of Fv architecture is conducive to grafting a second antibody binding site in the Fv region. Biophys J 1996; 71:2002-11. [PMID: 8889174 PMCID: PMC1233666 DOI: 10.1016/s0006-3495(96)79398-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Molecular modeling studies on antibody Fv regions have been pursued to design a second antigen-binding site (chi-site) in a chimeric single-chain Fv (chi sFv) species of about 30 kDa. This analysis has uncovered an architectural basis common to many Fv regions that permits grafting a chi-site onto the Fv surface that diametrically opposes the normal combining site. By using molecular graphics analysis, chimeric complementarity-determining regions (chi CDRs) were defined that comprised most of the CDRs from an antibody binding site of interest. The chain directionality of chi CDRs was consistent with that of specific bottom loops of the sFv, which allowed for grafting of chi CDRs with an overall geometry approximating CDRs in the parent combining site. Analysis of 10 different Fv crystal structures indicates that the positions for inserting chi CDRs are very highly conserved, as are the corresponding chi CDR boundaries in the parent binding site. The results of this investigation suggest that it should be possible to generally apply this approach to the development of chimeric bispecific antibody binding site (chi BABS) proteins.
Collapse
Affiliation(s)
- P C Keck
- Creative BioMolecules, Hopkinton, Massachusetts 01748, USA
| | | |
Collapse
|
41
|
Koren E, Milotic F, Neethling FA, Koscec M, Fei D, Kobayashi T, Taniguchi S, Cooper DK. Monoclonal antiidiotypic antibodies neutralize cytotoxic effects of anti-alphaGal antibodies. Transplantation 1996; 62:837-43. [PMID: 8824486 DOI: 10.1097/00007890-199609270-00023] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The aims of this study were to produce and characterize mouse monoclonal antiidiotypic antibodies (AIAs) that specifically bind human antipig (anti-alphaGal) antibodies and to select those AIAs that neutralize the cytotoxicity of human or baboon serum to pig (PK15) cells. Mice were immunized with human anti-pig antibodies, and hybridomas were produced using conventional techniques. From a total of 480 clones, 11 produced AIAs that bound with high affinity to human anti-alpha Gal IgG and F(ab')2 fragments, and individually reduced serum cytotoxicity to pig cells by 40 - 90%. Seven of the AIAs also bound to human peripheral B lymphocytes (that express the same idiotypes as the antibodies produce). Several combinations of two or three AIAs provided 100% protection of PK15 cells. Selected AIAs injected intravenously into baboons reduced the cytotoxicity of subsequently drawn sera by 50--80% for >48 hr. The cytotoxicity studies also indicated that there are at least two dominant idiotypes expressed in the human anti-alphaGal population. We conclude that AIAs against anti-alphaGal antibodies could be successfully used in 1) preoperative characterization of a recipient's anti-alphaGal profile, (2) prevention of hyperacute rejection of a pig organ, and (3) specific immunosuppression through elimination of anti-alphaGal-producing B lymphocytes.
Collapse
Affiliation(s)
- E Koren
- Oklahoma Medical Research Foundation, Oklahoma City, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Böldicke T, Haase B, Böcher M, Lindenmaier W. Human monoclonal antibodies to cytomegalovirus. Characterization and recombinant expression of a glycoprotein-B-specific antibody. EUROPEAN JOURNAL OF BIOCHEMISTRY 1995; 234:397-405. [PMID: 8536681 DOI: 10.1111/j.1432-1033.1995.397_b.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Human monoclonal antibodies (mAb) to human cytomegalovirus (HCMV) were established from spleen cells of a HCMV-positive donor. The antibodies (gamma 3, lambda) secreted from a stable heterohybridoma cell line were further characterized by immunoprecipitation and immune-fluorescence microscopy using HCMV infected cells and recombinant cell lines expressing HCMV glycoprotein B. The antibody reacted with the entire glycoprotein B or the extracellular domain expressed as glycoprotein-B--beta-galactosidase fusion protein in the native state, but the antibody was not neutralizing HCMV. Denatured and reduced forms of glycoprotein B were not recognized by this antibody, however, native glycoprotein B on the surface of infected cells was detected efficiently. The genes encoding the Fab part of the antibody were cloned and expressed in Escherichia coli. Recombinant Fab fragments specifically binding the extracellular domain of glycoprotein B could easily be isolated from the periplasmic space. Recombinant antibodies provide the opportunity to modify effector functions and to add tags to diagnostic antibodies for more efficient detection of CMV-infected cells.
Collapse
Affiliation(s)
- T Böldicke
- Gesellschaft für Biotechnologische Forschung mbH, Department of Applied Genetics, Braunschweig, Germany
| | | | | | | |
Collapse
|
43
|
Taylor G, Thomas LH, Wyld SG, Furze J, Sopp P, Howard CJ. Role of T-lymphocyte subsets in recovery from respiratory syncytial virus infection in calves. J Virol 1995; 69:6658-64. [PMID: 7474075 PMCID: PMC189575 DOI: 10.1128/jvi.69.11.6658-6664.1995] [Citation(s) in RCA: 108] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The role of T-cell subsets in respiratory syncytial virus (RSV) infection was investigated by using monoclonal antibodies (MAbs) to selectively deplete gnotobiotic calves of CD4+, CD8+, or WC1+ gamma delta T-cell receptor+ lymphocytes. Injection of these MAbs produced specific reductions of the target cell populations in the circulation and tissues. Ten days after RSV infection, immunoglobulin M (IgM), IgG1, and IgA antibodies were detected in sera and lung washings from control calves. Depletion of CD8+ T cells had no effect on either the serum or local antibody responses to RSV, whereas depletion of CD4+ T cells suppressed the antibody responses in two of three calves. The IgM and IgA responses were significantly increased in the lung washings of calves from which WC1+ T cells were depleted. Depletion of CD4+ or WC1+ T cells caused no significant delay in virus clearance, although an increase in the extent of pneumonic consolidation was observed in anti-CD4-treated calves. Nasopharyngeal excretion of RSV was prolonged in calves depleted of CD8+ T cells, and virus was isolated in high titers from lung washings of these animals 10 days after infection, whereas virus had been cleared from lung washings of all other animals. The delayed virus clearance was associated with an increase in the severity of pneumonic consolidation in three of four of the calves from which CD8+ T cells were depleted. This study shows that CD8+ T cells play a dominant role in the recovery of calves from RSV infection.
Collapse
Affiliation(s)
- G Taylor
- Division of Immunology and Pathology, Institute for Animal Health, Compton, Near Newbury, Berks, England
| | | | | | | | | | | |
Collapse
|
44
|
|
45
|
Abraham R, Buxbaum S, Link J, Smith R, Venti C, Darsley M. Screening and kinetic analysis of recombinant anti-CEA antibody fragments. J Immunol Methods 1995; 183:119-25. [PMID: 7602129 DOI: 10.1016/0022-1759(95)00039-d] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Four different carcinoembryonic antigen (CEA)-binding antibody fragments were prepared using the genes of the variable regions of the T84 epitope-specific antibody 7F7 and phage display techniques. The genes were successfully cloned and expressed in the pCANTAB5 phage display vector to investigate the kinetic binding parameters of each synthesized construct. Single chain fragments, Fab fragments, and two diabodies were purified and compared in their CEA-binding properties with the parent IgG using surface plasmon resonance detection. The on-rates for all these molecules were in the same order of magnitude (about 1 x 10(5) M-1 s-1) whereas major differences were detected in the off-rates. IgG and diabodies had slow off-rates due to bivalent binding, while single chain and Fab fragments dissociated rather fast. We also present a method for the immobilization of large amounts of CEA on CM5 sensorchips. These high density surfaces can be used for observing mass transport limited binding of CEA-specific molecules and are convenient tools for screening and quality control.
Collapse
Affiliation(s)
- R Abraham
- IGEN, Inc., Rockville, MD 20852, USA
| | | | | | | | | | | |
Collapse
|
46
|
Affiliation(s)
- B Henderson
- Maxillofacial Surgery Research Unit, Eastman Dental Institute for Oral Health Care Sciences, London, United Kingdom
| |
Collapse
|
47
|
2nd International meeting on synovium cell biology, physiology and pathology. Canterbury, United Kingdom, 21-23 September 1994. Proceedings and abstracts. Ann Rheum Dis 1995; 54:501-28. [PMID: 7632095 PMCID: PMC1009911 DOI: 10.1136/ard.54.6.501-a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
48
|
Navarro-Teulon I, Peraldi-Roux S, Bernardi T, Marin M, Piechaczyk M, Shire D, Pau B, Biard-Piechaczyk M. Expression in Escherichia coli of soluble and M13 phage-displayed forms of a single-chain antibody fragment specific for digoxin: assessment in a novel drug immunoassay. IMMUNOTECHNOLOGY : AN INTERNATIONAL JOURNAL OF IMMUNOLOGICAL ENGINEERING 1995; 1:41-52. [PMID: 9373332 DOI: 10.1016/1380-2933(95)00004-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A high affinity anti-digoxin single-chain Fv antibody fragment (scFv) was cloned from the mouse 2C2 hybridoma cell line and was functionally expressed both in the Escherichia coli periplasm as a soluble molecule and at the surface of the filamentous M13 bacteriophage as a fusion protein with the gene III minor coat protein. The 2C2 scFv sequence significantly differs from that of all the other anti-digoxin antibodies previously described. The 2C2 scFv shares with its parental monoclonal antibody a high specificity for digoxin, a cross-reactivity with active digoxin metabolites, but none with inactive metabolites. M13 phages displaying the 2C2 scFv at their surface have a high apparent affinity constant for digoxin (6.6 x 10(8) M-1) and were directly used to set up a novel type of immunoenzymatic assay for monitoring digoxin in sera of patients treated for either congestive heart failure or cardiac arrythmias. We thus report for the first time that phages displaying scFv may constitute a large source of important new reagents in the field of immunodiagnosis.
Collapse
|
49
|
Welschof M, Terness P, Kolbinger F, Zewe M, Dübel S, Dörsam H, Hain C, Finger M, Jung M, Moldenhauer G. Amino acid sequence based PCR primers for amplification of rearranged human heavy and light chain immunoglobulin variable region genes. J Immunol Methods 1995; 179:203-14. [PMID: 7876568 DOI: 10.1016/0022-1759(94)00286-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Previously described primers for PCR amplification of variable immunoglobulin (Ig) genes were based on gene sequences. To include the large number of amino acid sequences of antibodies whose DNA has not been sequenced and to ensure a maximal fit to rearranged human Ig variable region genes, we have made a comprehensive comparison of both protein and nucleotide sequences. The resulting set of 15 primers was able to amplify a wide range of rearranged antibody variable region genes. Restriction sites included in the primers facilitate cloning of the PCR products into various expression vectors. Sequence analyses of PCR-amplified cDNA derived from a polyclonal B cell population showed that maximal enrichment is obtained for highly represented variable Ig gene subgroups. Rarely occurring V kappa 4 and V lambda 5 subgroups were not detected. Rearranged Ig variable region genes from each of 19 human B cell lines were also amplified. Comparisons to germline sequences allowed the allocation of rearranged genes to the original Ig genes. This primer set should be very useful for generating large repertoires of rearranged V genes and for amplifying genes of individual B cell clones.
Collapse
Affiliation(s)
- M Welschof
- Department of Transplantation Immunology, University of Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Revillard JP, Robinet E, Goldman M, Bazin H, Latinne D, Chatenoud L. In vitro correlates of the acute toxic syndrome induced by some monoclonal antibodies: a rationale for the design of predictive tests. Toxicology 1995; 96:51-8. [PMID: 7863511 DOI: 10.1016/0300-483x(94)02975-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|