551
|
Beck A, Lambert J, Sun M, Lin K. Fourth World Antibody-Drug Conjugate Summit: February 29-March 1, 2012, Frankfurt, Germany. MAbs 2012; 4:637-47. [PMID: 22909934 PMCID: PMC3502230 DOI: 10.4161/mabs.21697] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The 4th World Antibody Drug Conjugate (WADC) Summit, organized by Hanson Wade was held on February 29‑March 1, 2012 in Frankfurt, Germany, which was also the location for the Antibody Drug Conjugate Summit Europe held in February 2011. During the one year between these meetings, antibody drug conjugates (ADCs) have confirmed their technological maturity and their clinical efficacy in oncology. Brentuximab vedotin (ADCETRISTM) gained approval by the US Food and Drug Administration in August 2011 and trastuzumab emtansine (T-DM1) confirmed impressive clinical efficacy responses in a large cohort of breast cancer patients. During the 4th WADC meeting, antibody-maytansinoid conjugates were showcased by representatives of ImmunoGen (T-DM1, SAR3419, lorvotuzumab mertansine/IMGN801, IMGN529 and IMG853) and Biotest (BT-062). Data on antibody-auristatin conjugates were presented by scientists and clinicians from Seattle Genetics and Takeda (brentuximab vedotin), Pfizer (5T4-MMAF), Agensys/Astella (AGS-16M8F), Progenics (PSMA-ADC) and Genmab (anti-TF ADCs). Alternative payloads such as calicheamicins and duocarmycin used for preparation of ADCs were discussed by Pfizer and Synthon representatives, respectively. In addition, emerging technologies, including site-directed conjugation (Ambrx), a protein toxin as payload (Viventia), hapten-binding bispecific antibodies (Roche), and use of light activated drugs (Photobiotics), were also presented. Last but not least, progresses in solving Chemistry Manufacturing and Control, and pharmacokinetic issues were addressed by scientists from Genentech, Pfizer, Novartis and Pierre Fabre.
Collapse
Affiliation(s)
- Alain Beck
- Centre d'Immunologie Pierre Fabre, Saint-Julien-en-Genevois, France.
| | | | | | | |
Collapse
|
552
|
Rosati S, Thompson NJ, Barendregt A, Hendriks LJA, Bakker ABH, de Kruif J, Throsby M, van Duijn E, Heck AJR. Qualitative and Semiquantitative Analysis of Composite Mixtures of Antibodies by Native Mass Spectrometry. Anal Chem 2012; 84:7227-32. [DOI: 10.1021/ac301611d] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Sara Rosati
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for
Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The
Netherlands
- Netherlands Proteomics Centre, Padualaan 8, 3584 CH
Utrecht, The Netherlands
| | - Natalie J. Thompson
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for
Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The
Netherlands
- Netherlands Proteomics Centre, Padualaan 8, 3584 CH
Utrecht, The Netherlands
| | - Arjan Barendregt
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for
Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The
Netherlands
- Netherlands Proteomics Centre, Padualaan 8, 3584 CH
Utrecht, The Netherlands
| | - Linda J. A. Hendriks
- Merus Biopharmaceuticals, Postvak 133,
Padualaan 8, 3584 CH Utrecht, The Netherlands
| | | | - John de Kruif
- Merus Biopharmaceuticals, Postvak 133,
Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Mark Throsby
- Merus Biopharmaceuticals, Postvak 133,
Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Esther van Duijn
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for
Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The
Netherlands
- Netherlands Proteomics Centre, Padualaan 8, 3584 CH
Utrecht, The Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for
Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The
Netherlands
- Netherlands Proteomics Centre, Padualaan 8, 3584 CH
Utrecht, The Netherlands
| |
Collapse
|
553
|
McAtee CP, Hornbuckle J. Isolation of monoclonal antibody charge variants by displacement chromatography. CURRENT PROTOCOLS IN PROTEIN SCIENCE 2012; Chapter 8:8.10.1-8.10.13. [PMID: 22851499 DOI: 10.1002/0471140864.ps0810s69] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
This unit discusses the important parameters in designing and optimizing a separation of monoclonal antibody (mAb) charge variants from process streams by ion-exchange displacement chromatography, including sample preparation and selection of matrix, column, and appropriate buffer. A protocol is provided for determination of optimal column binding and displacement conditions, including cleaning and regeneration of the displacement columns.
Collapse
|
554
|
Lingg N, Zhang P, Song Z, Bardor M. The sweet tooth of biopharmaceuticals: importance of recombinant protein glycosylation analysis. Biotechnol J 2012; 7:1462-72. [PMID: 22829536 DOI: 10.1002/biot.201200078] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 06/06/2012] [Accepted: 06/18/2012] [Indexed: 11/10/2022]
Abstract
Biopharmaceuticals currently represent the fastest growing sector of the pharmaceutical industry, mainly driven by a rapid expansion in the manufacture of recombinant protein-based drugs. Glycosylation is the most prominent post-translational modification occurring on these protein drugs. It constitutes one of the critical quality attributes that requires thorough analysis for optimal efficacy and safety. This review examines the functional importance of glycosylation of recombinant protein drugs, illustrated using three examples of protein biopharmaceuticals: IgG antibodies, erythropoietin and glucocerebrosidase. Current analytical methods are reviewed as solutions for qualitative and quantitative measurements of glycosylation to monitor quality target product profiles of recombinant glycoprotein drugs. Finally, we propose a framework for designing the quality target product profile of recombinant glycoproteins and planning workflow for glycosylation analysis with the selection of available analytical methods and tools.
Collapse
Affiliation(s)
- Nico Lingg
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | | | | | | |
Collapse
|
555
|
Schaefer JV, Plückthun A. Transfer of engineered biophysical properties between different antibody formats and expression systems. Protein Eng Des Sel 2012; 25:485-506. [PMID: 22763265 DOI: 10.1093/protein/gzs039] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recombinant antibodies and their derivatives are receiving ever increasing attention for many applications. Nevertheless, they differ widely in biophysical properties, from stable monomers to metastable aggregation-prone mixtures of oligomers. Previous work from our laboratory presented the combination of structure-based analysis with family consensus alignments as being able to improve the properties of immunoglobulin variable domains. We had identified a series of mutations in the variable domains that greatly influenced both the stability and the expression level of single-chain Fv (scFv) fragments produced in the periplasm of Escherichia coli. We now investigated whether these effects are transferable to Fab fragments and immunoglobulin G (IgG) produced in bacteria, Pichia pastoris, and mammalian cells. Taken together, our data indicate that engineered mutations can increase functional expression levels only for periplasmic expression in prokaryotes. In contrast, stability against thermal and denaturant-induced unfolding is improved by the same mutations in all formats tested, including scFv, Fab and IgG, independent of the expression system. The mutations in V(H) also influenced the structural homogeneity of full-length IgG, and the reducibility of the distant C(H)1-C(L) inter-chain disulfide bond. These results confirm the potential of structure-based protein engineering in the context of full-length IgGs and the transferability of stability improvements discovered with smaller antibody fragments.
Collapse
Affiliation(s)
- Jonas V Schaefer
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | | |
Collapse
|
556
|
Beck A, Reichert JM. Marketing approval of mogamulizumab: a triumph for glyco-engineering. MAbs 2012; 4:419-25. [PMID: 22699226 PMCID: PMC3499336 DOI: 10.4161/mabs.20996] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Therapeutic properties of antibodies strongly depend on the composition of their glycans. Most of the currently approved antibodies are produced in mammalian cell lines, which yield mixtures of different glycoforms that are close to those of humans, but not fully identical. Glyco-engineering is being developed as a method to control the composition of carbohydrates and to enhance the pharmacological properties of mAbs. The recent approval in Japan of mogamulizumab (POTELIGEO®), the first glyco-engineered antibody to reach the market, is a landmark in the field of therapeutic antibodies. Mogamulizumab is a humanized mAb derived from Kyowa Hakko Kirin’s POTELLIGENT® technology, which produces antibodies with enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) activity. The approval was granted April 30, 2012 by the Japanese Ministry of Health, Labour and Welfare for patients with relapsed or refractory CCR4-positive adult T-cell leukemia-lymphoma.
Collapse
|
557
|
Pauwels PJ, Dumontet C, Reichert JM, Beck A, Goetsch L, Corvaia N, Klein C, Coiffier B, Teicher B. 7th cancer scientific forum of theCancéropôle Lyon Auvergne Rhône-Alpes: March 20-21, 2012, Lyon, France. MAbs 2012; 4:434-44. [PMID: 22684281 PMCID: PMC3499338 DOI: 10.4161/mabs.20869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The Innovative Approaches in Anti-Cancer Monoclonal Antibodies meeting, held on March 20, 2012 in Lyon, was organized by Cancéropôle Lyon Auvergne-Rhône-Alps in partnership with the French competitiveness cluster Lyonbiopôle. CLARA is one of the seven cancer research clusters within France in charge of facilitating Translational Oncology Research by taking into account the objectives of the French National Cancer Plans I and II and, in coordination with the French National Cancer Institute and local authorities (mainly Grand Lyon, Rhône County and Rhône-Alpes Region), to perform economic development of research findings. The contribution of lectures by outstanding speakers as described in this report, the organization of two-round tables: "Antibody treatment in cancer: Unmet needs in solid tumors and hematological malignancies," and "From chimeric to more than human antibodies," together with face-to-face meetings, was shared by over 230 participants. The lectures provided an overview of the commercial pipeline of monoclonal antibody (mAb) therapeutics for cancer; discussion of the distinction between biosimilar, biobetter and next generation therapeutic antibodies for cancer; updates on obinutuzumab and the use of mAbs in lymphoma; and discussion of antibody-drug conjugates.
Collapse
|
558
|
Eissler N, Ruf P, Mysliwietz J, Lindhofer H, Mocikat R. Trifunctional bispecific antibodies induce tumor-specific T cells and elicit a vaccination effect. Cancer Res 2012; 72:3958-66. [PMID: 22745368 DOI: 10.1158/0008-5472.can-12-0146] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A major goal of tumor immunotherapy is the induction of long-lasting systemic T-cell immunity. Bispecific antibodies (bsAbs) that lack the immunoglobulin Fc region confer T-cell-mediated killing of tumor cells but do not induce long-term memory. In contrast, trifunctional bsAbs comprise an appropriate Fc region and, therefore, not only recruit T cells but also accessory cells that bear activating Fcγ receptors (FcγR), providing additional T-cell-activating signals and securing presentation of tumor-derived antigens to T cells. In this study, we show that trifunctional bsAbs induce a polyvalent T-cell response and, therefore, a vaccination effect. Mice were treated with melanoma cells and with a trifunctional bsAb directed against the melanoma target antigen ganglioside GD2 in addition to murine CD3. The trifunctional bsAb activated dendritic cells and induced a systemic immune response that was not replicated by treatment with the F(ab')2-counterpart lacking the Fc region. Restimulation of spleen and lymph node cells in vitro yielded T-cell lines that specifically produced interferon-γ in response to tumor. In addition, trifunctional bsAb-induced T cells recognized various specific peptides derived from melanoma-associated antigens. Moreover, these polyvalent responses proved to be tumor-suppressive and could not be induced by the corresponding bsF(ab')2-fragment. Taken together, our findings provide preclinical proof of concept that trifunctional bsAbs can induce tumor-specific T cells with defined antigen specificity.
Collapse
Affiliation(s)
- Nina Eissler
- Helmholtz-Zentrum München, Institut für Molekulare Immunologie; Trion Research GmbH, München, Germany
| | | | | | | | | |
Collapse
|
559
|
Both L, Banyard AC, van Dolleweerd C, Horton DL, Ma JKC, Fooks AR. Passive immunity in the prevention of rabies. THE LANCET. INFECTIOUS DISEASES 2012; 12:397-407. [PMID: 22541629 DOI: 10.1016/s1473-3099(11)70340-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Prevention of clinical disease in those exposed to viral infection is an important goal of human medicine. Using rabies virus infection as an example, we discuss the advances in passive immunoprophylaxis, most notably the shift from the recommended polyclonal human or equine immunoglobulins to monoclonal antibody therapies. The first rabies-specific monoclonal antibodies are undergoing clinical trials, so passive immunisation might finally become an accessible, affordable, and routinely used part of global health practices for rabies. Coupled with an adequate supply of modern tissue-culture vaccines, replacing the less efficient and unsafe nerve-tissue-derived rabies vaccines, the burden of this disease could be substantially reduced.
Collapse
Affiliation(s)
- Leonard Both
- Hotung Molecular Immunology Unit, Division of Clinical Sciences, St George's University of London, London, UK
| | | | | | | | | | | |
Collapse
|
560
|
Ballow M, Akdis CA, Casale TB, Wardlaw AJ, Wenzel SE, Ballas Z, Lötvall J. Immune response modifiers in the treatment of asthma: A PRACTALL document of the American Academy of Allergy, Asthma & Immunology and the European Academy of Allergy and Clinical Immunology. J Allergy Clin Immunol 2012; 130:311-24. [PMID: 22713596 DOI: 10.1016/j.jaci.2012.04.046] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 04/09/2012] [Indexed: 11/27/2022]
Affiliation(s)
- Mark Ballow
- Division of Allergy, Immunology & Pediatric Rheumatology, SUNY Buffalo School of Medicine, Buffalo, NY 14222, USA.
| | | | | | | | | | | | | |
Collapse
|
561
|
Reynaud CA, Moutel S, Dieu-Nosjean MC, Laskov R, Teillaud JL. 1(st) French-Israeli International Conference on B Cells and therapeutic antibodies: October 23-25, 2011 Jerusalem, Israel. MAbs 2012; 4:426-33. [PMID: 22664457 PMCID: PMC3499337 DOI: 10.4161/mabs.20289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Claude-Agnès Reynaud
- INSERM U. 783, Paris Descartes University, Faculty of Medicine, Necker-Enfants Malades site, Paris, France
| | | | | | | | | |
Collapse
|
562
|
Madhumathi J, Verma RS. Therapeutic targets and recent advances in protein immunotoxins. Curr Opin Microbiol 2012; 15:300-9. [PMID: 22647353 DOI: 10.1016/j.mib.2012.05.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2012] [Accepted: 05/14/2012] [Indexed: 12/25/2022]
Abstract
Targeted therapy has replaced the conventional methods of disease management with the advances in recombinant technology and increased understanding of molecular mechanisms of diseases. The immunotoxin strategy for diseases like cancer and a variety of autoimmune disorders has been used successfully in the past since its discovery. Since bacterial, fungal and plant toxins have various limitations like toxicity and immunogenicity, studies on fully humanized immunotoxins have gained attraction recently, which reduced toxicity significantly. Improved methods of antibody engineering have led to the emergence of various new formats of immunotoxins. This review summarizes the target moieties used in immunotoxin constructs in different diseases and describes the recent advances in immunotoxin targeting.
Collapse
Affiliation(s)
- Madhumathi J
- Stem Cell and Molecular Biology Laboratory, Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, Tamilnadu, India
| | | |
Collapse
|
563
|
Furlong MT, Ouyang Z, Wu S, Tamura J, Olah T, Tymiak A, Jemal M. A universal surrogate peptide to enable LC-MS/MS bioanalysis of a diversity of human monoclonal antibody and human Fc-fusion protein drug candidates in pre-clinical animal studies. Biomed Chromatogr 2012; 26:1024-32. [PMID: 22623136 DOI: 10.1002/bmc.2759] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 04/01/2012] [Accepted: 04/23/2012] [Indexed: 11/06/2022]
Abstract
For the development of human antibody Fc (fraction crystallizable) region-containing therapeutic protein candidates, which can be either monoclonal antibodies (mAbs) or pharmacologically active proteins/peptides fused to the Fc region of human Immunoglobulin G (IgG), reliable quantification of these proteins in animal pharmacokinetic study plasma samples is critical. LC-MS/MS has emerged as a promising assay platform for this purpose. LC-MS/MS assays used for bioanalysis of human antibody Fc region-containing therapeutic protein candidates frequently rely upon quantification of a 'signature' surrogate peptide whose sequence is unique to the protein analyte of interest. One drawback of the signature peptide approach is that a new LC-MS/MS assay must be developed for each new human Fc region-containing therapeutic protein. To address this issue, we propose an alternative 'universal surrogate peptide' approach for the quantification of human antibody Fc region-containing therapeutic protein candidates in plasma samples from all nonclinical species. A single surrogate tryptic peptide was identified in the Fc region of most human antibody Fc-containing therapeutic protein candidates. An LC-MS-MS method based upon this peptide was shown to be capable of supporting bioanalysis of a diversity of human Fc region-containing therapeutic protein candidates in plasma samples of all commonly used animal species.
Collapse
Affiliation(s)
- Michael T Furlong
- Analytical Research and Development, Research and Development, Bristol-Myers Squibb, Princeton, NJ 08543-4000, USA
| | | | | | | | | | | | | |
Collapse
|
564
|
Beck A, Sanglier-Cianférani S, Van Dorsselaer A. Biosimilar, biobetter, and next generation antibody characterization by mass spectrometry. Anal Chem 2012; 84:4637-46. [PMID: 22510259 DOI: 10.1021/ac3002885] [Citation(s) in RCA: 196] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This Feature will introduce the strategies of therapeutic antibodies (mAbs) in-depth characterization by mass spectrometry (MS) and discuss analytical comparison of biosimilar to originator mAbs, with the cases of trastuzumab and cetuximab. In addition, the structural and functional insights gained both by state-of-the art and emerging MS methods used for biobetters and next generation antibodies design and optimization will also be highlighted.
Collapse
Affiliation(s)
- Alain Beck
- Centre d'Immunologie Pierre Fabre (CIPF), 5 Av. Napoléon III, BP 60497, 74164 Saint-Julien-en-Genevois, France.
| | | | | |
Collapse
|
565
|
Unique approach for B lymphoma therapy. Blood 2012; 119:3647-8. [PMID: 22517869 DOI: 10.1182/blood-2012-02-409052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
566
|
Abstract
The ability of antibodies to bind to target molecules with high affinity and specificity has led to their widespread use in diagnostic and therapeutic applications. Nevertheless, a limitation of antibodies is their propensity to self-associate and aggregate at high concentrations and elevated temperatures. The large size and multidomain architecture of full-length monoclonal antibodies have frustrated systematic analysis of how antibody sequence and structure regulate antibody solubility. In contrast, analysis of single and multidomain antibody fragments that retain the binding activity of mono-clonal antibodies has provided valuable insights into the determinants of antibody aggregation. Here we review advances in engineering antibody frameworks, domain interfaces, and antigen-binding loops to prevent aggregation of natively and nonnatively folded antibody fragments. We also highlight advances and unmet challenges in developing robust strategies for engineering large, multidomain antibodies to resist aggregation.
Collapse
Affiliation(s)
- Joseph M Perchiacca
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | | |
Collapse
|
567
|
Abstract
The use of monoclonal antibodies (mAbs) for cancer therapy has achieved considerable success in recent years. Antibody-drug conjugates are powerful new treatment options for lymphomas and solid tumours, and immunomodulatory antibodies have also recently achieved remarkable clinical success. The development of therapeutic antibodies requires a deep understanding of cancer serology, protein-engineering techniques, mechanisms of action and resistance, and the interplay between the immune system and cancer cells. This Review outlines the fundamental strategies that are required to develop antibody therapies for cancer patients through iterative approaches to target and antibody selection, extending from preclinical studies to human trials.
Collapse
Affiliation(s)
- Andrew M Scott
- Ludwig Institute for Cancer Research; University of Melbourne; and Centre for PET, Austin Hospital, Melbourne, Victoria 3084, Australia.
| | | | | |
Collapse
|
568
|
Yu CM, Peng HP, Chen IC, Lee YC, Chen JB, Tsai KC, Chen CT, Chang JY, Yang EW, Hsu PC, Jian JW, Hsu HJ, Chang HJ, Hsu WL, Huang KF, Ma AC, Yang AS. Rationalization and design of the complementarity determining region sequences in an antibody-antigen recognition interface. PLoS One 2012; 7:e33340. [PMID: 22457753 PMCID: PMC3310866 DOI: 10.1371/journal.pone.0033340] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 02/14/2012] [Indexed: 12/01/2022] Open
Abstract
Protein-protein interactions are critical determinants in biological systems. Engineered proteins binding to specific areas on protein surfaces could lead to therapeutics or diagnostics for treating diseases in humans. But designing epitope-specific protein-protein interactions with computational atomistic interaction free energy remains a difficult challenge. Here we show that, with the antibody-VEGF (vascular endothelial growth factor) interaction as a model system, the experimentally observed amino acid preferences in the antibody-antigen interface can be rationalized with 3-dimensional distributions of interacting atoms derived from the database of protein structures. Machine learning models established on the rationalization can be generalized to design amino acid preferences in antibody-antigen interfaces, for which the experimental validations are tractable with current high throughput synthetic antibody display technologies. Leave-one-out cross validation on the benchmark system yielded the accuracy, precision, recall (sensitivity) and specificity of the overall binary predictions to be 0.69, 0.45, 0.63, and 0.71 respectively, and the overall Matthews correlation coefficient of the 20 amino acid types in the 24 interface CDR positions was 0.312. The structure-based computational antibody design methodology was further tested with other antibodies binding to VEGF. The results indicate that the methodology could provide alternatives to the current antibody technologies based on animal immune systems in engineering therapeutic and diagnostic antibodies against predetermined antigen epitopes.
Collapse
Affiliation(s)
- Chung-Ming Yu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hung-Pin Peng
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Institute of Biomedical Informatics, National Yang-Ming University, Taipei, Taiwan
- Bioinformatics Program, Taiwan International Graduate Program, Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | - Ing-Chien Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Ching Lee
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Jun-Bo Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Department of Computer Science, National Tsing-Hua University, Hsinchu, Taiwan
| | | | - Ching-Tai Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Institute of Bioinformatics and Systems Biology, National Chiao-Tung University, Hsinchu, Taiwan
- Institute of Information Sciences, Academia Sinica, Taipei, Taiwan
- Bioinformatics Program, Taiwan International Graduate Program, Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | - Jeng-Yih Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Ei-Wen Yang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Institute of Information Sciences, Academia Sinica, Taipei, Taiwan
| | - Po-Chiang Hsu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Jhih-Wei Jian
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Institute of Biomedical Informatics, National Yang-Ming University, Taipei, Taiwan
- Bioinformatics Program, Taiwan International Graduate Program, Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | - Hung-Ju Hsu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense University, Taipei, Taiwan
| | - Hung-Ju Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Institute of Biochemical Science, National Taiwan University, Taipei, Taiwan
- Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Wen-Lian Hsu
- Institute of Information Sciences, Academia Sinica, Taipei, Taiwan
| | - Kai-Fa Huang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Alex Che Ma
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - An-Suei Yang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
569
|
Hudak JE, Barfield RM, de Hart GW, Grob P, Nogales E, Bertozzi CR, Rabuka D. Synthesis of Heterobifunctional Protein Fusions Using Copper-Free Click Chemistry and the Aldehyde Tag. Angew Chem Int Ed Engl 2012. [DOI: 10.1002/ange.201108130] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
570
|
Hudak JE, Barfield RM, de Hart GW, Grob P, Nogales E, Bertozzi CR, Rabuka D. Synthesis of heterobifunctional protein fusions using copper-free click chemistry and the aldehyde tag. Angew Chem Int Ed Engl 2012; 51:4161-5. [PMID: 22407566 PMCID: PMC3379715 DOI: 10.1002/anie.201108130] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Indexed: 01/16/2023]
Affiliation(s)
- Jason E Hudak
- Department of Chemistry, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | | | | | | | | | | | | |
Collapse
|
571
|
Lugovskoy AA, Reichert JM, Beck A. 7th annual European Antibody Congress 2011: November 29-December 1, 2011, Geneva, Switzerland. MAbs 2012; 4:134-52. [PMID: 22453093 DOI: 10.4161/mabs.4.2.19426] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The 7th European Antibody Congress (EAC), organized by Terrapin Ltd., was again held in Geneva, Switzerland, following on the tradition established with the 4th EAC. The 2011 version of the EAC was attended by nearly 250 delegates who learned of the latest advances and trends in the global development of antibody-based therapeutics. The first day focused on advances in understanding structure-function relationships, choosing the best format, glycoengineering biobetter antibodies, improving the efficacy and drugability of mAbs and epitope mapping. On the second day, the discovery of novel targets for mAb therapy, clinical pipeline updates, use of antibody combinations to address resistance, generation and identification of mAbs against new targets and biosimilar mAb development were discussed. Antibody-drug conjugates, domain antibodies and new scaffolds and bispecific antibodies were the topics of the third day. In total, nearly 50 speakers provided updates of programs related to antibody research and development on-going in the academic, government and commercial sectors.
Collapse
Affiliation(s)
| | | | - Alain Beck
- Centre d'Immunologie Pierre Fabre; Saint-Julien en Genevois, France
| |
Collapse
|
572
|
DeSesso JM, Williams AL, Ahuja A, Bowman CJ, Hurtt ME. The placenta, transfer of immunoglobulins, and safety assessment of biopharmaceuticals in pregnancy. Crit Rev Toxicol 2012; 42:185-210. [PMID: 22348352 DOI: 10.3109/10408444.2011.653487] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
573
|
Blanc V, Bousseau A, Caron A, Carrez C, Lutz RJ, Lambert JM. SAR3419: an anti-CD19-Maytansinoid Immunoconjugate for the treatment of B-cell malignancies. Clin Cancer Res 2012; 17:6448-58. [PMID: 22003072 DOI: 10.1158/1078-0432.ccr-11-0485] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
SAR3419 is a novel anti-CD19 humanized monoclonal antibody conjugated to a maytansine derivate through a cleavable linker for the treatment of B-cell malignancies. SAR3419 combines the strengths of a high-potency tubulin inhibitor and the exquisite B-cell selectivity of an anti-CD19 antibody. The internalization and processing of SAR3419, following its binding at the surface of CD19-positive human lymphoma cell lines and xenograft models, release active metabolites that trigger cell-cycle arrest and apoptosis, leading to cell death and tumor regression. SAR3419 has also been shown to be active in different lymphoma xenograft models, including aggressive diffuse large B-cell lymphoma, resulting in complete regressions and tumor-free survival. In these models, the activity of SAR3419 compared favorably with rituximab and lymphoma standard of care chemotherapy. Two phase I trials with 2 different schedules of SAR3419 as a single agent were conducted in refractory/relapsed B-cell non-Hodgkin lymphoma. Activity was reported in both schedules, in heavily pretreated patients of both follicular and diffuse large B-cell lymphoma subtypes, with a notable lack of significant hematological toxicity, validating SAR3419 as an effective antibody-drug conjugate and opening opportunities in the future. Numerous B-cell-specific anti-CD19 biologics are available to treat B-cell non-Hodgkin lymphoma, and early phase I results obtained with SAR3419 suggest that it is a promising candidate for further development in this disease. In addition, thanks to the broad expression of CD19, SAR3419 may provide treatment options for B-cell leukemias that are often CD20-negative.
Collapse
Affiliation(s)
- Veronique Blanc
- Oncology Business Division, Sanofi, Vitry sur Seine, France.
| | | | | | | | | | | |
Collapse
|
574
|
Abstract
IgG antibodies trigger leukocyte activation and inflammation by forming immune complexes that crosslink activating Fcγ receptors (FcγRs). This is essential to combat infection, but detrimental if antibodies target or cross-react with autoantigens. The high specificity and long serum half-life of IgG antibodies confers tremendous therapeutic potential. Indeed, antibodies have been successfully employed to target cancers, autoreactive B cells, and pro-inflammatory cytokines. Conversely, IgG antibodies can also initiate anti-inflammatory responses. In the form of intravenous immunoglobulin (IVIG), IgGs are routinely administered to treat inflammatory autoimmune diseases. Importantly, the N-linked glycans on the IgG Fc are absolutely required for initiating these IgG effector functions. In fact, the Fc glycan composition dictates IgG affinity to individual FcγRs, and in a broader sense, binding to different FcγRs classes: activating, inhibitory, and anti-inflammatory (dendritic cell-specific ICAM-3 grabbing nonintegrin, DC-SIGN). The Fc glycan requirements to initiate and suppress inflammation will be discussed herein.
Collapse
Affiliation(s)
- Robert M Anthony
- Leonard Wagner Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York 10065, USA.
| | | | | |
Collapse
|
575
|
Killer peptide: a novel paradigm of antimicrobial, antiviral and immunomodulatory auto-delivering drugs. Future Med Chem 2012; 3:1209-31. [PMID: 21806382 DOI: 10.4155/fmc.11.71] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The incidence of life-threatening viral and microbial infections has dramatically increased over recent decades. Despite significant developments in anti-infective chemotherapy, many issues have increasingly narrowed the therapeutic options, making it imperative to discover new effective molecules. Among them, small peptides are arousing great interest. This review will focus in particular on a killer peptide, engineered from an anti-idiotypic recombinant antibody that mimics the activity of a wide-spectrum antimicrobial yeast killer toxin targeting β-glucan cell-wall receptors. The in vitro and in vivo antimicrobial, antiviral and immunomodulatory activities of killer peptide and its ability to spontaneously and reversibly self-assemble and slowly release its active dimeric form over time will be discussed as a novel paradigm of targeted auto-delivering drugs.
Collapse
|
576
|
Monoclonal antibodies for the treatment of cancer. Semin Cancer Biol 2012; 22:3-13. [PMID: 22245472 DOI: 10.1016/j.semcancer.2011.12.009] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 12/15/2011] [Indexed: 02/07/2023]
Abstract
Over the past decade, the clinical utility of monoclonal antibodies has been realized and antibodies are now a mainstay for the treatment of cancer. Antibodies have the unique capacity to target and kill tumor cells while simultaneously activating immune effectors to kill tumor cells through the complement cascade or antibody-dependent cellular cytotoxicity (ADCC). This multifaceted mechanism of action combined with target specificity underlies the capacity of antibodies to elicit anti-tumor responses while minimizing the frequency and magnitude of adverse events. This review will focus on mechanisms of action, clinical applications and putative mechanisms of resistance to monoclonal antibody therapy in the context of cancer.
Collapse
|
577
|
Hofmann M, Große-Hovest L, Nübling T, Pyż E, Bamberg ML, Aulwurm S, Bühring HJ, Schwartz K, Haen SP, Schilbach K, Rammensee HG, Salih HR, Jung G. Generation, selection and preclinical characterization of an Fc-optimized FLT3 antibody for the treatment of myeloid leukemia. Leukemia 2012; 26:1228-37. [PMID: 22289926 DOI: 10.1038/leu.2011.372] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The therapeutic efficacy of humanized or chimeric second-generation antitumor antibodies is clearly established, but often limited. In recent years, defined modifications of the glycosylation pattern or the amino-acid sequence of the human immunoglobulin G1 Fc part have resulted in the development of third-generation antibodies with improved capability to recruit Fc receptor-bearing effector cells. The first antibodies of this kind, currently evaluated in early clinical trials, are directed against lymphoma-associated antigens. Fc-engineered antibodies targeting myeloid leukemia are not yet available. We here report on the generation and preclinical characterization of an Fc-optimized antibody directed to the FMS-related tyrosine kinase 3 (FLT3), an antigen expressed on the leukemic blasts of all investigated patients with acute myeloid leukemia (AML). This antibody, termed 4G8SDIEM, mediated markedly enhanced cellular cytotoxicity against FLT3-expressing cell lines as well as blasts of AML patients. FLT3 expression levels on AML cells varied between 300 and 4600 molecules/cell and, in most cases, were substantially higher than those detected on normal hematopoietic precursor cells and dendritic cells (approximately 300 molecules/cell). Antibody-mediated cytotoxicity against these normal cells was not detectable. 4G8SDIEM has been produced in pharmaceutical quality in a university-owned production unit and is currently used for the treatment of leukemia patients.
Collapse
Affiliation(s)
- M Hofmann
- Department of Immunology, Eberhard-Karls University, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
578
|
Abstract
Producing recombinant mammalian proteins in native or near-native conformation is fundamental to many aspects of biology. Unfortunately, it is also a task whose outcome is extremely unpredictable. A protein that has been shaped over millions of generations of evolution for expression at a level appropriate to a specific cell type or in a particular developmental stage, may be toxic to a new host cell, or become insoluble (among many possible obstacles) when overexpressed in vitro. The object of this volume, "Protein Expression in Mammalian Cells," is to offer guidance for those who wish (or who have been forced by circumstance) to overexpress a mammalian protein in mammalian cells.
Collapse
Affiliation(s)
- James L Hartley
- Protein Expression Laboratory, SAIC-Frederick, Inc., National Cancer Institute, Frederick, MD, USA.
| |
Collapse
|
579
|
|
580
|
Abstract
Antibodies make up the largest, growing segment of protein therapeutics in the pharmaceutical and biotechnology industries. The development or engineering of therapeutic antibodies is based to a large extent on our knowledge of antibody structure and requires sophisticated methods that continue to evolve. In this chapter, after a review of what is known about the structure and functional properties of antibodies, the current, state-of-the-art antibody engineering methods are described. These methods include antibody humanization, antigen-affinity optimization, Fc engineering for modulated effector function and extended half-life, and engineering for improved stability and biophysical properties. X-ray crystallographic structures of antibody fragments and their complexes can play a critical role in guiding and, in some cases, accelerating these processes. These approaches represent guidelines for developing antibody therapeutics with the desired affinity, effector function, and biophysical properties.
Collapse
|
581
|
Collins JC, James K. Emac – a comparative index for the assessment of macrocyclization efficiency. MEDCHEMCOMM 2012. [DOI: 10.1039/c2md20176c] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Based upon a critical analysis of macrocyclization reactions published over the last three years, a macrocyclization efficiency index, Emac, is proposed, which takes into account both yield and concentration, as a means of determining the true efficiency of a macrocyclization reaction.
Collapse
Affiliation(s)
| | - Keith James
- Department of Chemistry
- The Scripps Research Institute
- USA
| |
Collapse
|
582
|
Yan B, Boyd D, Kaschak T, Tsukuda J, Shen A, Lin Y, Chung S, Gupta P, Kamath A, Wong A, Vernes JM, Meng GY, Totpal K, Schaefer G, Jiang G, Nogal B, Emery C, Vanderlaan M, Carter P, Harris R, Amanullah A. Engineering upper hinge improves stability and effector function of a human IgG1. J Biol Chem 2011; 287:5891-7. [PMID: 22203673 DOI: 10.1074/jbc.m111.311811] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Upper hinge is vulnerable to radical attacks that result in breakage of the heavy-light chain linkage and cleavage of the hinge of an IgG1. To further explore mechanisms responsible for the radical induced hinge degradation, nine mutants were designed to determine the roles that the upper hinge Asp and His play in the radical reactions. The observation that none of these substitutions could inhibit the breakage of the heavy-light chain linkage suggests that the breakage may result from electron transfer from Cys(231) directly to the heavy-light chain linkage upon radical attacks, and implies a pathway separate from His(229)-mediated hinge cleavage. On the other hand, the substitution of His(229) with Tyr showed promising advantages over the native antibody and other substitutions in improving the stability and function of the IgG1. This substitution inhibited the hinge cleavage by 98% and suggests that the redox active nature of Tyr did not enable it to replicate the ability of His to facilitate radical induced degradation. We propose that the lower redox potential of Tyr, a residue that may be the ultimate sink for oxidizing equivalents in proteins, is responsible for the inhibition. More importantly, the substitution increased the antibody's binding to FcγRIII receptors by 2-3-fold, and improved ADCC activity by 2-fold, while maintaining a similar pharmacokinetic profile with respect to the wild type. Implications of these observations for antibody engineering and development are discussed.
Collapse
Affiliation(s)
- Boxu Yan
- Department of Pharma Technical Development, Genentech, Oceanside, California 92056, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
583
|
Buechele C, Baessler T, Schmiedel BJ, Schumacher CE, Grosse-Hovest L, Rittig K, Salih HR. 4-1BB ligand modulates direct and Rituximab-induced NK-cell reactivity in chronic lymphocytic leukemia. Eur J Immunol 2011; 42:737-48. [PMID: 22144129 DOI: 10.1002/eji.201141920] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 11/04/2011] [Accepted: 11/21/2011] [Indexed: 12/17/2022]
Abstract
NK cells play an important role in tumor immunosurveillance and largely contribute to the therapeutic success of anti-tumor antibodies like Rituximab. Here, we studied the role of the TNF family member 4-1BB ligand (4-1BBL) during the interaction of NK cells with chronic lymphocytic leukemia (CLL) cells. 4-1BBL was highly expressed on patient B-CLL cells in all 56 investigated cases. Signaling via 4-1BBL following interaction with 4-1BB, which was detected on NK cells of CLL patients but not healthy individuals, led to the release of immunoregulatory cytokines including TNF by CLL cells. CLL patient sera contained elevated levels of TNF and induced 4-1BB upregulation on NK cells, which in turn impaired direct and Rituximab-induced NK-cell reactivity against 4-1BBL-expressing targets. NK-cell reactivity was not only enhanced by blocking the interaction of NK cell-expressed 4-1BB with 4-1BBL expressed by CLL cells, but also by preventing 4-1BB upregulation on NK cells via neutralization of TNF in patient serum with Infliximab. Our data indicate that 4-1BBL mediates NK-cell immunosubversion in CLL, and thus might contribute to the reportedly compromised efficacy of Rituximab to induce NK-cell reactivity in the disease, and that TNF neutralization may serve to enhance the efficacy of Rituximab treatment in CLL.
Collapse
Affiliation(s)
- Corina Buechele
- Department of Hematology, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | | | | | | | | | | | | |
Collapse
|
584
|
Abstract
Antibodies are the most successful affinity tools used today, in both fundamental and applied research (diagnostics, purification and therapeutics). Nonetheless, antibodies do have their limitations, including high production costs and low stability. Alternative affinity tools based on nucleic acids (aptamers), polypeptides (engineered binding proteins) and inorganic matrices (molecular imprinted polymers) have received considerable attention. A major advantage of these alternatives concerns the efficient (microbial) production and in vitro selection procedures. The latter approach allows for the high-throughput optimization of aptamers and engineered binding proteins, e.g. aiming at enhanced chemical and physical stability. This has resulted in a rapid development of the fields of nucleic acid- and protein-based affinity tools and, although they are certainly not as widely used as antibodies, the number of their applications has steadily increased in recent years. In the present review, we compare the properties of the more conventional antibodies with these innovative affinity tools. Recent advances of affinity tool developments are described, both in a medical setting (e.g. diagnostics, therapeutics and drug delivery) and in several niche areas for which antibodies appear to be less attractive. Furthermore, an outlook is provided on anticipated future developments.
Collapse
|
585
|
Latypov RF, Hogan S, Lau H, Gadgil H, Liu D. Elucidation of acid-induced unfolding and aggregation of human immunoglobulin IgG1 and IgG2 Fc. J Biol Chem 2011; 287:1381-96. [PMID: 22084250 DOI: 10.1074/jbc.m111.297697] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Understanding the underlying mechanisms of Fc aggregation is an important prerequisite for developing stable and efficacious antibody-based therapeutics. In our study, high resolution two-dimensional nuclear magnetic resonance (NMR) was employed to probe structural changes in the IgG1 Fc. A series of (1)H-(15)N heteronuclear single-quantum correlation NMR spectra were collected between pH 2.5 and 4.7 to assess whether unfolding of C(H)2 domains precedes that of C(H)3 domains. The same pH range was subsequently screened in Fc aggregation experiments that utilized molecules of IgG1 and IgG2 subclasses with varying levels of C(H)2 glycosylation. In addition, differential scanning calorimetry data were collected over a pH range of 3-7 to assess changes in C(H)2 and C(H)3 thermostability. As a result, compelling evidence was gathered that emphasizes the importance of C(H)2 stability in determining the rate and extent of Fc aggregation. In particular, we found that Fc domains of the IgG1 subclass have a lower propensity to aggregate compared with those of the IgG2 subclass. Our data for glycosylated, partially deglycosylated, and fully deglycosylated molecules further revealed the criticality of C(H)2 glycans in modulating Fc aggregation. These findings provide important insights into the stability of Fc-based therapeutics and promote better understanding of their acid-induced aggregation process.
Collapse
Affiliation(s)
- Ramil F Latypov
- Drug Product Development, Amgen Inc., Seattle, Washington 98119, USA.
| | | | | | | | | |
Collapse
|
586
|
Fernández-Marrero Y, López-Requena A. Lonely killers: effector cell- and complement-independent non-proapoptotic cytotoxic antibodies inducing membrane lesions. MAbs 2011; 3:528-34. [PMID: 22123064 DOI: 10.4161/mabs.3.6.17770] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The majority of the most effective monoclonal antibodies (mAbs) currently in the clinics bind to cancer or immune cells. Classic mechanisms of cell killing by therapeutic mAbs include antibody-dependent cell-mediated cytotoxicity, complement-dependent cytotoxicity and induction of apoptosis by engagement of specific cell ligands. A few reports have described mAbs whose cytotoxic activity is Fc-independent and that do not induce the morphological and biochemical changes associated with the apoptosis-type of cell death. Even fewer works describe mAbs able to directly induce membrane lesions. Here, we discuss the available data on those molecules and their cell killing activity, with particular attention to the case of a mAb specific for the tumor-associated N-glycolyl (Neu5Gc)-GM3 ganglioside (GM3(Neu5Gc)). Some similarities are found in the cell death pathways triggered by these mAbs, but data are not abundant. We conclude that the usefulness of mAbs with a direct cytotoxic activity for immunotherapeutic strategies deserves deeper research.
Collapse
|
587
|
|
588
|
Östner G, Lindström V, Postnikov AB, Solovyeva TI, Emilsson ÖI, Grubb A. High throughput testing of drug library substances and monoclonal antibodies for capacity to reduce formation of cystatin C dimers to identify candidates for treatment of hereditary cystatin C amyloid angiopathy. Scandinavian Journal of Clinical and Laboratory Investigation 2011; 71:676-82. [DOI: 10.3109/00365513.2011.621026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
589
|
Popov J, Kapanen AI, Turner C, Ng R, Tucker C, Chiu G, Klasa R, Bally MB, Chikh G. Multivalent rituximab lipid nanoparticles as improved lymphoma therapies: indirect mechanisms of action and in vivo activity. Nanomedicine (Lond) 2011; 6:1575-91. [PMID: 22011314 DOI: 10.2217/nnm.11.50] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
AIMS The activity of therapeutic antibodies can be enhanced by creating multivalent constructs, such as antibody lipid nanoparticles (LNPs). Here, we examine differences between rituximab (Ritux) and Ritux-LNPs in terms of their indirect mechanisms of action: complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC). MATERIALS & METHODS We employed two mantle-cell lymphoma cell lines, Z138 and JVM2, which exhibit different in vivo sensitivities to Ritux along with variable expression levels of cell-surface proteins that regulate ADCC and CDC. RESULTS In both cell lines, CDC and ADCC were found to be significantly enhanced after treatment with Ritux-LNPs compared with Ritux. In vivo efficacy studies, however, suggested that the therapeutic activities of Ritux and Ritux-LNPs were equivalent, which was subsequently explained in part by pharmacokinetic studies indicating rapid elimination of Ritux-LNP. CONCLUSION Although indirect and direct mechanisms of multivalent Ritux are enhanced, its further development requires methods to improve its circulation lifetime.
Collapse
Affiliation(s)
- Jesse Popov
- Department of Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Ave., Vancouver BC, V5Z 1L3, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
590
|
Muda M, Gross AW, Dawson JP, He C, Kurosawa E, Schweickhardt R, Dugas M, Soloviev M, Bernhardt A, Fischer D, Wesolowski JS, Kelton C, Neuteboom B, Hock B. Therapeutic assessment of SEED: a new engineered antibody platform designed to generate mono- and bispecific antibodies. Protein Eng Des Sel 2011; 24:447-54. [PMID: 21498564 DOI: 10.1093/protein/gzq123] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The strand-exchange engineered domain (SEED) platform was designed to generate asymmetric and bispecific antibody-like molecules, a capability that expands therapeutic applications of natural antibodies. This new protein engineered platform is based on exchanging structurally related sequences of immunoglobulin within the conserved CH3 domains. Alternating sequences from human IgA and IgG in the SEED CH3 domains generate two asymmetric but complementary domains, designated AG and GA. The SEED design allows efficient generation of AG/GA heterodimers, while disfavoring homodimerization of AG and GA SEED CH3 domains. Using a clinically validated antibody (C225), we tested whether Fab derivatives constructed on the SEED platform retain desirable therapeutic antibody features such as in vitro and in vivo stability, favorable pharmacokinetics, ligand binding and effector functions including antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity. In addition, we tested SEED with combinations of binder domains (scFv, VHH, Fab). Mono- and bivalent Fab-SEED fusions retain full binding affinity, have excellent biochemical and biophysical stability, and retain desirable antibody-like characteristics conferred by Fc domains. Furthermore, SEED is compatible with different combinations of Fab, scFv and VHH domains. Our assessment shows that the new SEED platform expands therapeutic applications of natural antibodies by generating heterodimeric Fc-analog proteins.
Collapse
Affiliation(s)
- Marco Muda
- EMD Serono Research Institute, Inc, One Technology Place, Rockland, MA 02370, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
591
|
Koti M, Nagy E, Kaushik AK. A single point mutation in framework region 3 of heavy chain affects viral neutralization dynamics of single-chain Fv against bovine herpes virus type 1. Vaccine 2011; 29:7905-12. [DOI: 10.1016/j.vaccine.2011.08.077] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 07/20/2011] [Accepted: 08/15/2011] [Indexed: 10/17/2022]
|
592
|
Hart MH, de Vrieze H, Wouters D, Wolbink GJ, Killestein J, de Groot ER, Aarden LA, Rispens T. Differential effect of drug interference in immunogenicity assays. J Immunol Methods 2011; 372:196-203. [DOI: 10.1016/j.jim.2011.07.019] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 06/22/2011] [Accepted: 07/20/2011] [Indexed: 01/13/2023]
|
593
|
Abstract
Hematologic malignancies were the first diseases in clinical oncology for which the potential of harnessing the immune system as targeted therapy was unequivocally demonstrated. Unfortunately, the use of this highly efficacious modality has been limited to only a subset of patients and diseases because of immune-mediated toxicities resulting from incomplete specificity, and disease-specific determinants of sensitivity versus resistance to immune effector mechanisms. Recent studies, however, have begun to elucidate the molecular basis of the observed clinical effects allowing the rational development of next generation of immunotherapeutic combinations. We discuss here cancer antigen targets in hematologic malignancies and the specific approaches to induce immunity being pursued, the importance of modulating the host immunoregulatory environment, and the special features of immunological monitoring in clinical investigation. The hematologic malignancies represent an ideal setting for the development of immunotherapy due to logistical, clinical monitoring, and disease biology factors and may represent an exemplar for immune-based treatment in other cancer types.
Collapse
Affiliation(s)
- Christopher S Hourigan
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231-1000, USA
| | | |
Collapse
|
594
|
Model simulation and experimental verification of a cation-exchange IgG capture step in batch and continuous chromatography. J Chromatogr A 2011; 1218:5195-204. [DOI: 10.1016/j.chroma.2011.05.103] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 05/24/2011] [Accepted: 05/30/2011] [Indexed: 11/21/2022]
|
595
|
Regulatory considerations for development of bioanalytical assays for biotechnology products. Bioanalysis 2011; 3:597-603. [PMID: 21417729 DOI: 10.4155/bio.11.27] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Ligand-binding assays are the predominant method used for determination of concentrations of biotechnology products in serum or other matrices, as well as for the determination of antidrug antibodies in nonclinical and clinical studies. The challenges regarding the design and validation of these assays are well understood. The US FDA published a Guidance for Industry on Bioanalytical Method Validation and a Draft Guidance for Industry on Assay Development for Immunogenicity Testing of Therapeutic Proteins. The purpose of this article is to highlight specific elements in these guidance documents that should also apply to new methods, discuss the application of new generation ligand-binding methods and LC-MS for these purposes and provide a scientific and regulatory perspective on the specific challenges assessing the pharmacokinetics and immunogenicity of monoclonal antibodies.
Collapse
|
596
|
High-level production of a kringle domain variant by high-cell-density cultivation of Escherichia coli. Appl Microbiol Biotechnol 2011; 92:327-36. [DOI: 10.1007/s00253-011-3423-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 05/24/2011] [Accepted: 05/29/2011] [Indexed: 11/25/2022]
|
597
|
Immunoglobulin domain crossover as a generic approach for the production of bispecific IgG antibodies. Proc Natl Acad Sci U S A 2011; 108:11187-92. [PMID: 21690412 DOI: 10.1073/pnas.1019002108] [Citation(s) in RCA: 327] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We describe a generic approach to assemble correctly two heavy and two light chains, derived from two existing antibodies, to form human bivalent bispecific IgG antibodies without use of artificial linkers. Based on the knobs-into-holes technology that enables heterodimerization of the heavy chains, correct association of the light chains and their cognate heavy chains is achieved by exchange of heavy-chain and light-chain domains within the antigen binding fragment (Fab) of one half of the bispecific antibody. This "crossover" retains the antigen-binding affinity but makes the two arms so different that light-chain mispairing can no longer occur. Applying the three possible "CrossMab" formats, we generated bispecific antibodies against angiopoietin-2 (Ang-2) and vascular endothelial growth factor A (VEGF-A) and show that they can be produced by standard techniques, exhibit stabilities comparable to natural antibodies, and bind both targets simultaneously with unaltered affinity. Because of its superior side-product profile, the CrossMab(CH1-CL) was selected for in vivo profiling and showed potent antiangiogenic and antitumoral activity.
Collapse
|
598
|
Christen S, Holdener M, Beerli C, Thoma G, Bayer M, Pfeilschifter JM, Hintermann E, Zerwes HG, Christen U. Small molecule CXCR3 antagonist NIBR2130 has only a limited impact on type 1 diabetes in a virus-induced mouse model. Clin Exp Immunol 2011; 165:318-28. [PMID: 21649647 DOI: 10.1111/j.1365-2249.2011.04426.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
CXCL10 is one of the key chemokines involved in trafficking of autoaggressive T cells to the islets of Langerhans during the autoimmune destruction of beta cells in type 1 diabetes (T1D). Blockade of CXCL10 or genetic deletion of its receptor CXCR3 results in a reduction of T1D in animal models. As an alternative to the use of neutralizing monoclonal antibodies to CXCL10 or CXCR3 we evaluated the small molecule CXCR3 antagonist NIBR2130 in a virus-induced mouse model for T1D. We found that the overall frequency of T1D was not reduced in mice administered with NIBR2130. An initial slight delay of diabetes onset was not stable over time, because the mice turned diabetic upon removal of the antagonist. Accordingly, no significant differences were found in the islet infiltration rate and the frequency and activity of islet antigen-specific T cells between protected mice administered with NIBR2130 and control mice. Our data indicate that in contrast to direct inhibition of CXCL10, blockade of CXCR3 with the small molecule antagonist NIBR2130 has no impact on trafficking and/or activation of autoaggressive T cells and is not sufficient to prevent T1D.
Collapse
Affiliation(s)
- S Christen
- Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe Universität, Frankfurt am Main, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
599
|
Immediate adverse reactions to biologicals: from pathogenic mechanisms to prophylactic management. Curr Opin Allergy Clin Immunol 2011; 11:262-8. [DOI: 10.1097/aci.0b013e3283464bcd] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
600
|
Targeting siglecs--a novel pharmacological strategy for immuno- and glycotherapy. Biochem Pharmacol 2011; 82:323-32. [PMID: 21658374 DOI: 10.1016/j.bcp.2011.05.018] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 05/17/2011] [Indexed: 01/13/2023]
Abstract
The immune system must be tightly held in check to avoid bystander tissue damage as well as autoreactivity caused by overwhelming immune reactions. A novel family of immunoregulatory, carbohydrate-binding receptors, the Siglecs (sialic acid binding immunoglobulin-like lectins), has received particular attention in light of their capacity to mediate cell death, anti-proliferative effects and to regulate a variety of cellular activities. Siglec receptors are mainly expressed on leukocytes in a cell type-specific and differentiation-dependent manner. Siglecs might potentially be exploited as targets of novel immune- and glycotherapeutics for cell-directed therapies in autoimmune and allergic diseases, as well as in haematologic malignancies. Here we present novel insights on structural and functional characteristics, expression patterns and evolutionary aspects of Siglecs and their ligands. Pharmacological strategies using Siglec agonistic cross-linking therapeutics, such as monoclonal or engineered antibodies, intravenous immunoglobulin (IVIG), or glycomimetics are discussed. Modulation of immune responses by targeting Siglecs using agonistic or antagonistic therapeutics may have important clinical implications and may pave the way for novel pharmacological avenues for the treatment of autoimmune and allergic diseases or for tumor immunotherapy.
Collapse
|