1
|
Protein folding stabilities are a major determinant of oxidation rates for buried methionine residues. J Biol Chem 2022; 298:101872. [PMID: 35346688 PMCID: PMC9062257 DOI: 10.1016/j.jbc.2022.101872] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 12/20/2022] Open
Abstract
The oxidation of protein-bound methionines to form methionine sulfoxides has a broad range of biological ramifications, making it important to delineate factors that influence methionine oxidation rates within a given protein. This is especially important for biopharmaceuticals, where oxidation can lead to deactivation and degradation. Previously, neighboring residue effects and solvent accessibility have been shown to impact the susceptibility of methionine residues to oxidation. In this study, we provide proteome-wide evidence that oxidation rates of buried methionine residues are also strongly influenced by the thermodynamic folding stability of proteins. We surveyed the Escherichia coli proteome using several proteomic methodologies and globally measured oxidation rates of methionine residues in the presence and absence of tertiary structure, as well as the folding stabilities of methionine-containing domains. These data indicated that buried methionines have a wide range of protection factors against oxidation that correlate strongly with folding stabilities. Consistent with this, we show that in comparison to E. coli, the proteome of the thermophile Thermus thermophilus is significantly more stable and thus more resistant to methionine oxidation. To demonstrate the utility of this correlation, we used native methionine oxidation rates to survey the folding stabilities of E. coli and T. thermophilus proteomes at various temperatures and propose a model that relates the temperature dependence of the folding stabilities of these two species to their optimal growth temperatures. Overall, these results indicate that oxidation rates of buried methionines from the native state of proteins can be used as a metric of folding stability.
Collapse
|
2
|
Sankar K, Trainor K, Blazer L, Adams J, Sidhu S, Day T, Meiering E, Maier J. A Descriptor set for Quantitative Structure-Property Relationship Prediction in Biologics. Mol Inform 2022; 41:e2100240. [PMID: 35277930 DOI: 10.1002/minf.202100240] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 03/11/2022] [Indexed: 11/12/2022]
Abstract
In addition to attaining the desired binding to their targets, a crucial aspect in the development of biotherapeutics is 'developability', which includes several desirable properties such as high solubility, low viscosity and aggregation, physico-chemical stability and low immunogenicity. The lack of any of these properties can lead to significant obstacles in advancing them to clinic; thus in silico methods capable of raising warning flags in earlier stages of development are highly beneficial. We have developed a computational framework based on a large and diverse set of protein specific descriptors ideal for making liability predictions using a machine-learning approach. This set offers a high degree of feature diversity classifiable by sequence, structure and surface patches. We assess the sensitivity and applicability of these descriptors in four dedicated case studies that are believed to be representative of biophysical characterizations commonly employed during the development process. In addition to data sets obtained from public sources, we have validated the descriptors on novel experimental data sets in order to address antibody developability and to generate prospective predictions on Adnectins. The results demonstrate that the descriptors are well suited to assist in the improvement of properties of systems that exhibit poor solubility or aggregation.
Collapse
|
3
|
Gao X, Wang W, Tesar D, Wei B, Eschelbach J, Kelley RF, Jiang G. An Approach to Bioactivity Assessment for Critical Quality Attribute Identification Based on Antibody-Antigen Complex Structure. J Pharm Sci 2020; 110:1652-1660. [PMID: 33383056 DOI: 10.1016/j.xphs.2020.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/06/2020] [Accepted: 12/21/2020] [Indexed: 11/25/2022]
Abstract
Identification of critical quality attributes (CQAs) is an important step for development of biopharmaceuticals with intended performance. An accurate CQA assessment is needed to ensure product quality and focusing on development efforts where control is needed. The assignment of criticality is based on safety and efficacy. Efficacy is related to PK and bioactivity. Here, we developed a novel approach based on antibody-antigen complex structure and modeling as a complementary method for bioactivity assessment. To validate this approach, common product related quality attributes and mutagenesis data from several IgGs were assessed using available antibody-antigen complex structures, and results were compared with experimental data from bioactivity or binding affinity measurements. A stepwise evaluation scheme for structural based analysis is proposed; based on systematic assessment following the scheme, good correlation has been observed between structural analysis and experimental data. This demonstrates that such an approach can be applied as a complementary tool for bioactivity assessment. Main applications are 1) To decouple multiple attributes to achieve amino acid resolution for bioactivity assessment, 2) To assess bioactivity of attributes that cannot be experimentally generated, 3) To provide molecular mechanism for experimental observation and understand structure function relationship. Examples are provided to illustrate these applications.
Collapse
Affiliation(s)
- Xuan Gao
- Analytical Development and Quality Control, Genentech, South San Francisco, CA.
| | - Weiru Wang
- Structural Biology, Genentech, South San Francisco, CA
| | - Devin Tesar
- Drug Delivery, Genentech, South San Francisco, CA
| | - Bingchuan Wei
- Small Molecule Analytical Chemistry, Genentech, South San Francisco, CA
| | - John Eschelbach
- Protein Analytical Chemistry, Genentech, South San Francisco, CA
| | | | - Guoying Jiang
- Analytical Development and Quality Control, Genentech, South San Francisco, CA.
| |
Collapse
|
4
|
Nicolas A, Dejoux A, Poirier C, Aubrey N, Péan JM, Velge-Roussel F. Contribution of Intrinsic Fluorescence to the Design of a New 3D-Printed Implant for Releasing SDABS. Pharmaceutics 2020; 12:pharmaceutics12100921. [PMID: 32993086 PMCID: PMC7601711 DOI: 10.3390/pharmaceutics12100921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 11/23/2022] Open
Abstract
Single-domain antibodies (sdAbs) offer great features such as increased stability but are hampered by a limited serum half-life. Many strategies have been developed to improve the sdAb half-life, such as protein engineering and controlled release systems (CRS). In our study, we designed a new product that combined a hydrogel with a 3D-printed implant. The results demonstrate the implant’s ability to sustain sdAb release up to 13 days through a reduced initial burst release followed by a continuous release. Furthermore, formulation screening helped to identify the best sdAb formulation conditions and improved our understanding of our CRS. Through the screening step, we gained knowledge about the influence of the choice of polymer and about potential interactions between the sdAb and the polymer. To conclude, this feasibility study confirmed the ability of our CRS to extend sdAb release and established the fundamental role of formulation screening for maximizing knowledge about our CRS.
Collapse
Affiliation(s)
- Alexandre Nicolas
- GICC EA 7501, Faculty of Medicine, University of Tours, 37032 Tours, France;
- PEX DPH, Technologie Servier, 45000 Orleans, France; (A.D.); (C.P.); (J.-M.P.)
| | - Alice Dejoux
- PEX DPH, Technologie Servier, 45000 Orleans, France; (A.D.); (C.P.); (J.-M.P.)
| | - Cécile Poirier
- PEX DPH, Technologie Servier, 45000 Orleans, France; (A.D.); (C.P.); (J.-M.P.)
| | - Nicolas Aubrey
- ISP UMR 1282, INRA, Team BioMAP, University of Tours, 37200 Tours, France;
| | - Jean-Manuel Péan
- PEX DPH, Technologie Servier, 45000 Orleans, France; (A.D.); (C.P.); (J.-M.P.)
| | - Florence Velge-Roussel
- GICC EA 7501, Faculty of Medicine, University of Tours, 37032 Tours, France;
- Correspondence: ; Tel.: +33-(0)2-4736-6058
| |
Collapse
|
5
|
Sankar K, Hoi KH, Yin Y, Ramachandran P, Andersen N, Hilderbrand A, McDonald P, Spiess C, Zhang Q. Prediction of methionine oxidation risk in monoclonal antibodies using a machine learning method. MAbs 2018; 10:1281-1290. [PMID: 30252602 PMCID: PMC6284603 DOI: 10.1080/19420862.2018.1518887] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 08/15/2018] [Accepted: 08/28/2018] [Indexed: 12/22/2022] Open
Abstract
Monoclonal antibodies (mAbs) have become a major class of protein therapeutics that target a spectrum of diseases ranging from cancers to infectious diseases. Similar to any protein molecule, mAbs are susceptible to chemical modifications during the manufacturing process, long-term storage, and in vivo circulation that can impair their potency. One such modification is the oxidation of methionine residues. Chemical modifications that occur in the complementarity-determining regions (CDRs) of mAbs can lead to the abrogation of antigen binding and reduce the drug's potency and efficacy. Thus, it is highly desirable to identify and eliminate any chemically unstable residues in the CDRs during the therapeutic antibody discovery process. To provide increased throughput over experimental methods, we extracted features from the mAbs' sequences, structures, and dynamics, used random forests to identify important features and develop a quantitative and highly predictive in silico methionine oxidation model.
Collapse
Affiliation(s)
- Kannan Sankar
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Kam Hon Hoi
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
- Department of Bioinformatics and Computational Biology, Genentech, South San Francisco, CA, USA
| | - Yizhou Yin
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
- Institute for Bioscience and Biotechnology Research, Biological Sciences Graduate Program, University of Maryland, Rockville, MD, USA
| | - Prasanna Ramachandran
- Department of Analytical Development and Quality Control, Genentech, South San Francisco, CA, USA
| | - Nisana Andersen
- Department of Analytical Development and Quality Control, Genentech, South San Francisco, CA, USA
| | - Amy Hilderbrand
- Department of Analytical Development and Quality Control, Genentech, South San Francisco, CA, USA
| | - Paul McDonald
- Department of Purification Development and Bioprocess Development, Genentech, South San Francisco, CA, USA
| | - Christoph Spiess
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Qing Zhang
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
- Department of Bioinformatics and Computational Biology, Genentech, South San Francisco, CA, USA
| |
Collapse
|
6
|
Choi Y, Ndong C, Griswold KE, Bailey-Kellogg C. Computationally driven antibody engineering enables simultaneous humanization and thermostabilization. Protein Eng Des Sel 2016; 29:419-426. [PMID: 27334453 DOI: 10.1093/protein/gzw024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 05/25/2016] [Indexed: 12/22/2022] Open
Abstract
Humanization reduces the immunogenicity risk of therapeutic antibodies of non-human origin. Thermostabilization can be critical for clinical development and application of therapeutic antibodies. Here, we show that the computational antibody redesign method Computationally Driven Antibody Humanization (CoDAH) enables these two goals to be accomplished simultaneously and seamlessly. A panel of CoDAH designs for the murine parent of cetuximab, a chimeric anti-EGFR antibody, exhibited both substantially improved thermostabilities and substantially higher levels of humanness, while retaining binding activity near the parental level. The consistently high quality of the turnkey CoDAH designs, over a whole panel of variants, suggests that the computationally directed approach encapsulates key determinants of antibody structure and function.
Collapse
Affiliation(s)
- Yoonjoo Choi
- Department of Computer Science, Dartmouth College, Hanover, NH 03755, USA
| | - Christian Ndong
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Karl E Griswold
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA.,Norris Cotton Cancer Center at Dartmouth, Lebanon, NH 03766, USA.,Department of Biological Sciences, Dartmouth, Hanover, NH 03755, USA
| | | |
Collapse
|
7
|
Understanding the Supersensitive Anti-Drug Antibody Assay: Unexpected High Anti-Drug Antibody Incidence and Its Clinical Relevance. J Immunol Res 2016; 2016:3072586. [PMID: 27340678 PMCID: PMC4906211 DOI: 10.1155/2016/3072586] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/08/2016] [Indexed: 12/20/2022] Open
Abstract
Numbers of biotherapeutic products in development have increased over past decade. Despite providing significant benefits to patients with unmet needs, almost all protein-based biotherapeutics could induce unwanted immunogenicity, which result in a loss of efficacy and/or increase the risk of adverse reactions, such as infusion reactions, anaphylaxis, and even life-threatening response to endogenous proteins. Recognizing these possibilities, regulatory agencies request that immunogenicity be assessed as part of the approval process for biotherapeutics. Great efforts have been made to reduce drug immunogenicity through protein engineering. Accordingly the immunogenicity incidence has been reduced from around 80% in murine derived products to 0–10% in fully human products. However, recent improvements in immunogenicity assays have led to unexpectedly high immunogenicity rates, even in fully human products, leading to new challenges in assessing immunogenicity and its clinical relevance. These new immunogenicity assays are becoming supersensitive and able to detect more of anti-drug antibodies (ADA) than with earlier assays. This paper intends to review and discuss our understanding of the supersensitive ADA assay and the unexpected high ADA incidence and its potential clinical relevance.
Collapse
|
8
|
The Effects of Light-Accelerated Degradation on the Aggregation of Marketed Therapeutic Monoclonal Antibodies Evaluated by Size-Exclusion Chromatography With Diode Array Detection. J Pharm Sci 2016; 105:1405-18. [PMID: 26952878 DOI: 10.1016/j.xphs.2016.01.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 08/28/2015] [Accepted: 12/15/2015] [Indexed: 11/24/2022]
Abstract
Research into the effects that exposure to light can have on therapeutic proteins is essential for ensuring the quality and safety of the medicines in which they are used. It is important to understand the effects of light on aggregation to help avoid undesirable colloidal instabilities, both in the original medicines and in the formats in which they are finally administered. In this study, 5 marketed therapeutic mAbs, namely bevacizumab, cetuximab, infliximab, rituximab, and trastuzumab, were investigated for this purpose. The medicines and 2 diluted preparations in 0.9 NaCl (2 mg/mL and 5 mg/mL)-commonly used in clinical practice-were subjected to controlled light-accelerated degradation. The formation of aggregates was monitored by size-exclusion chromatography. The results indicated that light induced protein aggregation. This process of protein damage was influenced above all by mAb concentration, although the particular characteristics of each mAb were also important. Photodegradation also produced the fragmentation of the mAbs. The damage caused to the mAbs as a result of light-induced aggregation and/or fragmentation was demonstrated both in the medicines and in the diluted preparation forms. These findings should be carefully considered when handling the medicines for administration and when recommending beyond-use dates in normal hospital conditions.
Collapse
|
9
|
Yang Y, Zhao J, Geng S, Hou C, Li X, Lang X, Qiao C, Li Y, Feng J, Lv M, Shen B, Zhang B. Improving Trastuzumab’s Stability Profile by Removing the Two Degradation Hotspots. J Pharm Sci 2015; 104:1960-1970. [DOI: 10.1002/jps.24435] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 02/15/2015] [Accepted: 03/03/2015] [Indexed: 12/20/2022]
|
10
|
D’Amico E, Caserta C, Patti F. Monoclonal antibody therapy in multiple sclerosis: critical appraisal and new perspectives. Expert Rev Neurother 2015; 15:251-68. [DOI: 10.1586/14737175.2015.1008458] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
11
|
Gómez-Mantilla JD, Trocóniz IF, Parra-Guillén Z, Garrido MJ. Review on modeling anti-antibody responses to monoclonal antibodies. J Pharmacokinet Pharmacodyn 2014; 41:523-36. [PMID: 25027160 DOI: 10.1007/s10928-014-9367-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 06/25/2014] [Indexed: 10/25/2022]
Abstract
Monoclonal antibodies (mAbs) represent a therapeutic strategy that has been increasingly used in different diseases. mAbs are highly specific for their targets leading to induce specific effector functions. Despite their therapeutic benefits, the presence of immunogenic reactions is of growing concern. The immunogenicity identified as anti-drug antibodies (ADA) production due to the continuous administration of mAbs may affect the pharmacokinetics (PK) and/or the pharmacodynamics (PD) of mAbs administered to patients. Therefore, the immunogenicity and its clinical impact have been studied by several authors using PK modeling approaches. In this review, the authors try to present all those models under a unique theoretical mechanism-based framework incorporating the main considerations related to ADA formation, and how ADA may affect the efficacy or toxicity profile of some therapeutic biomolecules.
Collapse
Affiliation(s)
- José David Gómez-Mantilla
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, 31080, Spain
| | | | | | | |
Collapse
|
12
|
Structure-based prediction of asparagine and aspartate degradation sites in antibody variable regions. PLoS One 2014; 9:e100736. [PMID: 24959685 PMCID: PMC4069079 DOI: 10.1371/journal.pone.0100736] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 05/30/2014] [Indexed: 12/29/2022] Open
Abstract
Monoclonal antibodies (mAbs) and proteins containing antibody domains are the most prevalent class of biotherapeutics in diverse indication areas. Today, established techniques such as immunization or phage display allow for an efficient generation of new mAbs. Besides functional properties, the stability of future therapeutic mAbs is a key selection criterion which is essential for the development of a drug candidate into a marketed product. Therapeutic proteins may degrade via asparagine (Asn) deamidation and aspartate (Asp) isomerization, but the factors responsible for such degradation remain poorly understood. We studied the structural properties of a large, uniform dataset of Asn and Asp residues in the variable domains of antibodies. Their structural parameters were correlated with the degradation propensities measured by mass spectrometry. We show that degradation hotspots can be characterized by their conformational flexibility, the size of the C-terminally flanking amino acid residue, and secondary structural parameters. From these results we derive an accurate in silico prediction method for the degradation propensity of both Asn and Asp residues in the complementarity-determining regions (CDRs) of mAbs.
Collapse
|
13
|
Patterson KG, Dixon Pittaro JL, Bastedo PS, Hess DA, Haeryfar SMM, McCormick JK. Control of established colon cancer xenografts using a novel humanized single chain antibody-streptococcal superantigen fusion protein targeting the 5T4 oncofetal antigen. PLoS One 2014; 9:e95200. [PMID: 24736661 PMCID: PMC3988171 DOI: 10.1371/journal.pone.0095200] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 03/25/2014] [Indexed: 01/21/2023] Open
Abstract
Superantigens (SAgs) are microbial toxins that cross-link T cell receptors with major histocompatibility class II (MHC-II) molecules leading to the activation of large numbers of T cells. Herein, we describe the development and preclinical testing of a novel tumor-targeted SAg (TTS) therapeutic built using the streptococcal pyrogenic exotoxin C (SpeC) SAg and targeting cancer cells expressing the 5T4 tumor-associated antigen (TAA). To inhibit potentially harmful widespread immune cell activation, a SpeC mutation within the high-affinity MHC-II binding interface was generated (SpeCD203A) that demonstrated a pronounced reduction in mitogenic activity, yet this mutant could still induce immune cell-mediated cancer cell death in vitro. To target 5T4+ cancer cells, we engineered a humanized single chain variable fragment (scFv) antibody to recognize 5T4 (scFv5T4). Specific targeting of scFv5T4 was verified. SpeCD203A fused to scFv5T4 maintained the ability to activate and induce immune cell-mediated cytotoxicity of colorectal cancer cells. Using a xenograft model of established human colon cancer, we demonstrated that the SpeC-based TTS was able to control the growth and spread of large tumors in vivo. This required both TAA targeting by scFv5T4 and functional SAg activity. These studies lay the foundation for the development of streptococcal SAgs as ‘next-generation’ TTSs for cancer immunotherapy.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/genetics
- Antibodies, Monoclonal, Humanized/immunology
- Antigens, Neoplasm/immunology
- Cell Line, Tumor
- Cell Proliferation
- Cell Transformation, Neoplastic
- Colonic Neoplasms/immunology
- Colonic Neoplasms/pathology
- Colonic Neoplasms/therapy
- Humans
- Immunotherapy/methods
- Mice
- Models, Molecular
- Protein Conformation
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Single-Chain Antibodies/genetics
- Single-Chain Antibodies/immunology
- Streptococcus/immunology
- Superantigens/genetics
- Superantigens/immunology
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Kelcey G. Patterson
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | | | - Peter S. Bastedo
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - David A. Hess
- Department of Physiology and Pharmacology, Western University, London Ontario, Canada
- Vascular Biology Research Group, Robarts Research Institute, London, Ontario, Canada
| | - S. M. Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| | - John K. McCormick
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
14
|
Yang J, Goetze AM, Flynn GC. Assessment of naturally occurring covalent and total dimer levels in human IgG1 and IgG2. Mol Immunol 2013; 58:108-15. [PMID: 24321397 DOI: 10.1016/j.molimm.2013.11.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 11/13/2013] [Accepted: 11/14/2013] [Indexed: 10/25/2022]
Abstract
Antibody dimers, two self-associated monomers, have been detected on both recombinantly expressed and endogenous human IgG proteins. Nearly 10 years ago, Yoo et al. (2003) described low levels of IgG2 covalent dimer, in human serum, but did not quantify the levels. Here we quantify the total and covalent dimer levels of IgG2 and IgG1 in human blood, and study the origin of covalent dimer formation. Low levels (<1%) of total IgG1 and IgG2 dimers were measured in freshly prepared human plasma. Both IgG1 and IgG2 covalent dimers were also found in plasma. Whereas IgG1 covalent dimer levels were significantly reduced by steps intended to eliminate artifacts during sample preparation, IgG2 covalent dimer levels remain stable in such conditions. About 0.4% of IgG2 in plasma was in a covalent dimer form, yet very little (<0.03%) of IgG1 covalent dimer could be considered naturally occurring. IgG2 dimer also formed in vitro under conditions designed to mimic those in blood, suggesting that formation occurs in vivo during circulation. Thus, small amounts of covalent IgG2 dimer do appear to form naturally.
Collapse
Affiliation(s)
- Jane Yang
- Department of Process and Product Development, Amgen Inc., Thousand Oaks, 91320, United States
| | - Andrew M Goetze
- Department of Process and Product Development, Amgen Inc., Thousand Oaks, 91320, United States
| | - Gregory C Flynn
- Department of Process and Product Development, Amgen Inc., Thousand Oaks, 91320, United States.
| |
Collapse
|
15
|
A novel antibody humanization method based on epitopes scanning and molecular dynamics simulation. PLoS One 2013; 8:e80636. [PMID: 24278299 PMCID: PMC3836750 DOI: 10.1371/journal.pone.0080636] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 10/05/2013] [Indexed: 11/19/2022] Open
Abstract
1-17-2 is a rat anti-human DEC-205 monoclonal antibody that induces internalization and delivers antigen to dendritic cells (DCs). The potentially clinical application of this antibody is limited by its murine origin. Traditional humanization method such as complementarity determining regions (CDRs) graft often leads to a decreased or even lost affinity. Here we have developed a novel antibody humanization method based on computer modeling and bioinformatics analysis. First, we used homology modeling technology to build the precise model of Fab. A novel epitope scanning algorithm was designed to identify antigenic residues in the framework regions (FRs) that need to be mutated to human counterpart in the humanization process. Then virtual mutation and molecular dynamics (MD) simulation were used to assess the conformational impact imposed by all the mutations. By comparing the root-mean-square deviations (RMSDs) of CDRs, we found five key residues whose mutations would destroy the original conformation of CDRs. These residues need to be back-mutated to rescue the antibody binding affinity. Finally we constructed the antibodies in vitro and compared their binding affinity by flow cytometry and surface plasmon resonance (SPR) assay. The binding affinity of the refined humanized antibody was similar to that of the original rat antibody. Our results have established a novel method based on epitopes scanning and MD simulation for antibody humanization.
Collapse
|
16
|
Jiang A, Zhang W, Wu Q, Jin W, Tang Y, Zhang J, Liu JN. Construction and characterization of a novel chimeric antibody c3C7 specific for the integrin αIIbβ3 complex. Appl Microbiol Biotechnol 2013; 98:105-14. [PMID: 24113827 DOI: 10.1007/s00253-013-5284-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/09/2013] [Accepted: 09/11/2013] [Indexed: 11/26/2022]
Abstract
A murine monoclonal antibody (mAb) 3C7 against integrin αIIbβ3 was previously obtained as a potential antithrombotic agent in our laboratory. The epitope of 3C7 is a specific conformation of the αIIbβ3 complex, but not either of the two subunits, which makes it different from abciximab, a supplementary antibody drug used in percutaneous coronary intervention which has a cross-reaction with other integrins sharing the β3 subunit. To reduce the human anti-mouse antibody reactions of 3C7, the variable regions of this antibody were cloned and fused with the constant counterparts of human IgG1. Two vectors of light and heavy chains were constructed and co-transfected into CHO-dhfr(-) cells. The chimeric antibody c3C7 was purified and the properties of c3C7 were compared with 3C7. Identical to its parent antibody 3C7, c3C7 binds to the αIIbβ3 complex, but not to either of the subunits. The K(d) value of c3C7 was in the same order of magnitude as 3C7 (1.570 ± 0.326 vs 0.780 ± 0.182 nmol/L). Human platelet aggregation induced by adenosine diphosphate was effectively inhibited by c3C7 in a dose-dependent manner. In conclusion, after the modification, c3C7 retained the properties of its parent mAb with no loss of its biological activity. Therefore, c3C7 has the potential to become a novel agent for the treatment of thrombosis.
Collapse
Affiliation(s)
- Aiqin Jiang
- Institute of Molecular Medicine, Nanjing University, Nanjing, 210093, China,
| | | | | | | | | | | | | |
Collapse
|
17
|
Debaene F, Wagner-Rousset E, Colas O, Ayoub D, Corvaïa N, Van Dorsselaer A, Beck A, Cianférani S. Time Resolved Native Ion-Mobility Mass Spectrometry to Monitor Dynamics of IgG4 Fab Arm Exchange and “Bispecific” Monoclonal Antibody Formation. Anal Chem 2013; 85:9785-92. [DOI: 10.1021/ac402237v] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- François Debaene
- Laboratoire
de Spectrométrie de Masse BioOrganique (LSMBO), IPHC-DSA, Université de Strasbourg, CNRS, UMR7178, 25 rue Becquerel, 67087 Strasbourg, France
| | - Elsa Wagner-Rousset
- Centre d’Immunologie Pierre-Fabre (CIPF), 5 Av. Napoléon III, BP
60497, 74164 Saint-Julien-en-Genevois, France
| | - Olivier Colas
- Centre d’Immunologie Pierre-Fabre (CIPF), 5 Av. Napoléon III, BP
60497, 74164 Saint-Julien-en-Genevois, France
| | - Daniel Ayoub
- Centre d’Immunologie Pierre-Fabre (CIPF), 5 Av. Napoléon III, BP
60497, 74164 Saint-Julien-en-Genevois, France
| | - Nathalie Corvaïa
- Centre d’Immunologie Pierre-Fabre (CIPF), 5 Av. Napoléon III, BP
60497, 74164 Saint-Julien-en-Genevois, France
| | - Alain Van Dorsselaer
- Laboratoire
de Spectrométrie de Masse BioOrganique (LSMBO), IPHC-DSA, Université de Strasbourg, CNRS, UMR7178, 25 rue Becquerel, 67087 Strasbourg, France
| | - Alain Beck
- Centre d’Immunologie Pierre-Fabre (CIPF), 5 Av. Napoléon III, BP
60497, 74164 Saint-Julien-en-Genevois, France
| | - Sarah Cianférani
- Laboratoire
de Spectrométrie de Masse BioOrganique (LSMBO), IPHC-DSA, Université de Strasbourg, CNRS, UMR7178, 25 rue Becquerel, 67087 Strasbourg, France
| |
Collapse
|
18
|
Von Kreudenstein TS, Escobar-Carbrera E, Lario PI, D'Angelo I, Brault K, Kelly J, Durocher Y, Baardsnes J, Woods RJ, Xie MH, Girod PA, Suits MDL, Boulanger MJ, Poon DKY, Ng GYK, Dixit SB. Improving biophysical properties of a bispecific antibody scaffold to aid developability: quality by molecular design. MAbs 2013; 5:646-54. [PMID: 23924797 DOI: 10.4161/mabs.25632] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
While the concept of Quality-by-Design is addressed at the upstream and downstream process development stages, we questioned whether there are advantages to addressing the issues of biologics quality early in the design of the molecule based on fundamental biophysical characterization, and thereby reduce complexities in the product development stages. Although limited number of bispecific therapeutics are in clinic, these developments have been plagued with difficulty in producing materials of sufficient quality and quantity for both preclinical and clinical studies. The engineered heterodimeric Fc is an industry-wide favorite scaffold for the design of bispecific protein therapeutics because of its structural, and potentially pharmacokinetic, similarity to the natural antibody. Development of molecules based on this concept, however, is challenged by the presence of potential homodimer contamination and stability loss relative to the natural Fc. We engineered a heterodimeric Fc with high heterodimeric specificity that also retains natural Fc-like biophysical properties, and demonstrate here that use of engineered Fc domains that mirror the natural system translates into an efficient and robust upstream stable cell line selection process as a first step toward a more developable therapeutic.
Collapse
|
19
|
Immunogenicity testing strategy and bioanalytical assays for antibody–drug conjugates. Bioanalysis 2013; 5:1041-55. [DOI: 10.4155/bio.13.10] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background: Immunogenicity testing is an important component of clinical development for large-molecule biotherapeutics. New complex types of large molecules, such as antibody–drug conjugates (ADCs), require careful evaluation of the testing strategy and bioanalytical assays used to monitor the development of antitherapeutic antibodies. Results: An electrochemiluminescence-based immunoassay for the detection and epitope characterization of anti-ADC antibodies was validated. Using this assay format, antibodies directed against the monoclonal antibody and linker–drug components of the ADC were successfully detected in a multiple-dose rat toxicity study. Conclusion: Immunogenicity assays incorporating epitope determination may provide additional information about the characteristics of induced antitherapeutic antibodies, including the magnitude and timing of the various types of antibody responses.
Collapse
|
20
|
Overcoming soluble target interference in an anti-therapeutic antibody screening assay for an antibody-drug conjugate therapeutic. Bioanalysis 2013; 4:2013-26. [PMID: 22946917 DOI: 10.4155/bio.12.165] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The standard safety evaluation of biotherapeutics includes assessment of immunogenicity. Anti-therapeutic antibodies (ATA) can be detected in serum using immunoassays with a bridging format. However, these assays can be subject to interference. RESULTS In the bridging ATA assay for 3A5 TDC, an antibody-drug conjugate that binds to the multimeric extracellular domain of MUC16 (CA125), soluble CA125 in the serum caused false-positive results by binding to the ATA assay reagents. This interaction was blocked by wheat germ agglutinin lectin as it binds to the glycans in CA125; thus, the specificity of the assay improved. CONCLUSION The assay development and validation results showed that the addition of wheat germ agglutinin eliminates the interference from circulating CA125 without impacting the ability to detect ATA.
Collapse
|
21
|
Ball C, Fox B, Hufton S, Sharp G, Poole S, Stebbings R, Eastwood D, Findlay L, Parren PWHI, Thorpe R, Bristow A, Thorpe SJ. Antibody C Region Influences TGN1412-like Functional Activity In Vitro. THE JOURNAL OF IMMUNOLOGY 2012; 189:5831-40. [DOI: 10.4049/jimmunol.1201795] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
22
|
Chirmule N, Jawa V, Meibohm B. Immunogenicity to therapeutic proteins: impact on PK/PD and efficacy. AAPS JOURNAL 2012; 14:296-302. [PMID: 22407289 DOI: 10.1208/s12248-012-9340-y] [Citation(s) in RCA: 234] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 02/21/2012] [Indexed: 01/13/2023]
Abstract
The development of therapeutic proteins requires the understanding of the relationship between the dose, exposure, efficacy, and toxicity of these molecules. Several intrinsic and extrinsic factors contribute to the challenges for measuring therapeutic proteins in a precise and accurate manner. In addition, induction of an immune response to therapeutic protein results in additional complexities in the analysis of the pharmacokinetic profile, toxicity, safety, and efficacy of this class of molecules. Assessment of immunogenicity of therapeutic proteins is a required aspect of regulatory filings for a licensing application and for the safe and efficacious use of these compounds. A systematic strategy and well-defined criteria for measuring anti-drug antibodies (ADA) have been established, to a large extent, through coordinated efforts. These recommendations are based on risk assessment and include the determination of ADA content (concentration/titer), affinity, immunoglobulin isotype/subtype, and neutralization capacity. This manuscript reviews the requirements necessary for understanding the nature of an ADA response in order to discern the impact of immunogenicity on pharmacokinetics/pharmacodynamics and efficacy.
Collapse
|
23
|
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: 7.9] [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]
|
24
|
Miletich J, Eich G, Grampp G, Mounho B. Biosimilars 2.0: guiding principles for a global "patients first" standard. MAbs 2011; 3:318-25. [PMID: 21512318 DOI: 10.4161/mabs.3.3.15599] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
In the European Union, biosimilar products have been approved since 2006 under an abbreviated pathway that leverages their similarity to an existing "reference" biological product. The products approved to date are based on recombinant versions of endogenous proteins with well-understood structures and pharmacology, but complicated safety and immunogenicity profiles. The period during the 2000s that included the first reviews, approvals, sale and use of biosimilars, is referred to herein as "Biosimilars 1.0." Over the next several years, a new and advanced tranche of biosimilars will be developed for complex reference products, including medicines used in the treatment of cancer and autoimmune diseases. A global market for biosimilars is developing, and this may well foreshadow the beginning of the second era of product development. This Biosimilars 2.0 period will likely be characterized by the development of complex products, global harmonization of standards, and the increasing demand for long-term monitoring of pharmaceuticals. The products developed in this period should exhibit high levels of fidelity to the reference products and should be rigorously evaluated in analytical, non-clinical and clinical comparisons. Additionally, Biosimilars 2.0 manufacturers should strive for transparency in their labels and take proactive strides to be accountable to providers and patients for the quality of their products. An important opportunity now exists for the healthcare community, industry and regulators to work in partnership to outline the appropriate standards for these products to facilitate increased access while meeting patients' needs.
Collapse
|
25
|
Frandsen TP, Naested H, Rasmussen SK, Hauptig P, Wiberg FC, Rasmussen LK, Jensen AMV, Persson P, Wikén M, Engström A, Jiang Y, Thorpe SJ, Förberg C, Tolstrup AB. Consistent manufacturing and quality control of a highly complex recombinant polyclonal antibody product for human therapeutic use. Biotechnol Bioeng 2011; 108:2171-81. [PMID: 21495017 DOI: 10.1002/bit.23166] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 03/28/2011] [Accepted: 03/30/2011] [Indexed: 01/31/2023]
Abstract
The beneficial effect of antibody therapy in human disease has become well established mainly for the treatment of cancer and immunological disorders. The inherent monospecificity of mAbs present limitations to mAb therapy which have become apparent notably in addressing complex entities like infectious agents or heterogenic endogenous targets. For such indications mixtures of antibodies comprising a combination of specificities would convey more potent biological effect which could translate into therapeutic efficacy. Recombinant polyclonal antibodies (rpAb) consisting of a defined number of well-characterized mAbs constitute a new class of target specific antibody therapy. We have developed a cost-efficient cell banking and single-batch manufacturing concept for the production of such products and demonstrate that a complex pAb composition, rozrolimupab, comprising 25 individual antibodies can be manufactured in a highly consistent manner in a scaled-up manufacturing process. We present a strategy for the release and characterization of antibody mixtures which constitute a complete series of chemistry, manufacturing, and control (CMC) analytical methods to address identity, purity, quantity, potency, and general characteristics. Finally we document selected quality attributes of rozrolimupab based on a battery of assays at the genetic-, protein-, and functional level and demonstrate that the manufactured rozrolimupab batches are highly pure and very uniform in their composition.
Collapse
|
26
|
Challenges in developing bioanalytical assays for characterization of antibody–drug conjugates. Bioanalysis 2011; 3:677-700. [DOI: 10.4155/bio.11.30] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
With more than 34 targets being investigated and nearly 20 clinical trials at various phases of development, antibody–drug conjugates (ADCs) hold a lot of promise for improving oncological malignancy therapy. This therapeutic strategy designed to specifically or preferentially deliver a cytotoxic agent to tumor cells through conjugation to a monoclonal antibody is not new. Although this approach is relatively simple conceptually, the history of ADCs clearly attests to the high degree of complexity in their development. Each component of an ADC is important to achieve efficacy with minimal toxicity, and the ability to monitor this multicomponent therapeutic entity is deemed to be critical for their successful optimization. In this article we review the different bioanalytical strategies that have been implemented to characterize various ADCs and discuss the challenges and issues associated with these approaches.
Collapse
|
27
|
Capturing the natural diversity of the human antibody response against vaccinia virus. J Virol 2010; 85:1820-33. [PMID: 21147924 DOI: 10.1128/jvi.02127-10] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The eradication of smallpox (variola) and the subsequent cessation of routine vaccination have left modern society vulnerable to bioterrorism employing this devastating contagious disease. The existing, licensed vaccines based on live vaccinia virus (VACV) are contraindicated for a substantial number of people, and prophylactic vaccination of large populations is not reasonable when there is little risk of exposure. Consequently, there is an emerging need to develop efficient and safe therapeutics to be used shortly before or after exposure, either alone or in combination with vaccination. We have characterized the human antibody response to smallpox vaccine (VACV Lister) in immunized volunteers and isolated a large number of VACV-specific antibodies that recognize a variety of different VACV antigens. Using this broad antibody panel, we have generated a fully human, recombinant analogue to plasma-derived vaccinia immunoglobulin (VIG), which mirrors the diversity and specificity of the human antibody immune response and offers the advantage of unlimited supply and reproducible specificity and activity. The recombinant VIG was found to display a high specific binding activity toward VACV antigens, potent in vitro VACV neutralizing activity, and a highly protective efficacy against VACV challenge in the mouse tail lesion model when given either prophylactically or therapeutically. Altogether, the results suggest that this compound has the potential to be used as an effective postexposure prophylaxis or treatment of disease caused by orthopoxviruses.
Collapse
|
28
|
|
29
|
Singh SK. Impact of product-related factors on immunogenicity of biotherapeutics. J Pharm Sci 2010; 100:354-87. [PMID: 20740683 DOI: 10.1002/jps.22276] [Citation(s) in RCA: 260] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 05/13/2010] [Accepted: 05/24/2010] [Indexed: 12/12/2022]
Abstract
All protein therapeutics have the potential to be immunogenic. Several factors, including patient characteristics, disease state, and the therapy itself, influence the generation of an immune response. Product-related factors such as the molecule design, the expression system, post-translational modifications, impurities, contaminants, formulation and excipients, container, closure, as well as degradation products are all implicated. However, a critical examination of the available data shows that clear unequivocal evidence for the impact of these latter factors on clinical immunogenicity is lacking. No report could be found that clearly deconvolutes the clinical impact of the product attributes on patient susceptibility. Aggregation carries the greatest concern as a risk factor for immunogenicity, but the impact of aggregates is likely to depend on their structure as well as on the functionality (e.g., immunostimulatory or immunomodulatory) of the therapeutic. Preclinical studies are not yet capable of assessing the clinically relevant immunogenicity potential of these product-related factors. Simply addressing these risk factors as part of product development will not eliminate immunogenicity. Minimization of immunogenicity has to begin at the molecule design stage by reducing or eliminating antigenic epitopes and building in favorable physical and chemical properties.
Collapse
Affiliation(s)
- Satish Kumar Singh
- Pfizer, Inc., BioTherapeutics Pharmaceutical Sciences, Pharmaceutical Research and Development, Chesterfield, Missouri 63017, USA.
| |
Collapse
|
30
|
Persson P, Engström A, Rasmussen LK, Holmberg E, Frandsen TP. Development of Mass Spectrometry Based Techniques for the Identification and Determination of Compositional Variability in Recombinant Polyclonal Antibody Products. Anal Chem 2010; 82:7274-82. [DOI: 10.1021/ac101175w] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Pia Persson
- Swedish Orphan Biovitrum AB, Tomtebodavägen 23 A, SE-112 76 Stockholm, Sweden, and Symphogen A/S, Elektrovej building 375, 2800 Lyngby, Denmark
| | - Anders Engström
- Swedish Orphan Biovitrum AB, Tomtebodavägen 23 A, SE-112 76 Stockholm, Sweden, and Symphogen A/S, Elektrovej building 375, 2800 Lyngby, Denmark
| | - Lone K. Rasmussen
- Swedish Orphan Biovitrum AB, Tomtebodavägen 23 A, SE-112 76 Stockholm, Sweden, and Symphogen A/S, Elektrovej building 375, 2800 Lyngby, Denmark
| | - Erland Holmberg
- Swedish Orphan Biovitrum AB, Tomtebodavägen 23 A, SE-112 76 Stockholm, Sweden, and Symphogen A/S, Elektrovej building 375, 2800 Lyngby, Denmark
| | - Torben P. Frandsen
- Swedish Orphan Biovitrum AB, Tomtebodavägen 23 A, SE-112 76 Stockholm, Sweden, and Symphogen A/S, Elektrovej building 375, 2800 Lyngby, Denmark
| |
Collapse
|
31
|
Wuchner K, Büchler J, Spycher R, Dalmonte P, Volkin DB. Development of a Microflow Digital Imaging Assay to Characterize Protein Particulates During Storage of a High Concentration IgG1 Monoclonal Antibody Formulation. J Pharm Sci 2010; 99:3343-61. [DOI: 10.1002/jps.22123] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
32
|
Bond MD, Panek ME, Zhang Z, Wang D, Mehndiratta P, Zhao H, Gunton K, Ni A, Nedved ML, Burman S, Volkin DB. Evaluation of a dual-wavelength size exclusion HPLC method with improved sensitivity to detect protein aggregates and its use to better characterize degradation pathways of an IgG1 monoclonal antibody. J Pharm Sci 2010; 99:2582-97. [PMID: 20039394 DOI: 10.1002/jps.22034] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The evaluation of a dual wavelength size exclusion high performance liquid chromatography (DW-SE-HPLC) method with improved sensitivity to detect aggregates in a high concentration IgG1 monoclonal antibody formulation is presented. This technique utilizes ultraviolet detection at two different wavelengths to monitor the levels of monomer, aggregate, and fragments and was shown to have improved sensitivity for the detection aggregates and fragments compared to light scattering (LS) detection. After assay optimization including the use of column conditioning, the limit of quantitation for aggregates was determined to be 0.04% with essentially complete recovery of aggregates from the column (>99.5%). The DW-SE-HPLC method was used to evaluate the level of protein aggregates generated by different environmental conditions such as exposure to elevated temperatures/acidic pH or intense light. The detection and characterization of protein aggregates by DW-SE-HPLC was compared with an orthogonal biophysical technique (sedimentation velocity analytical ultracentrifugation, SV-AUC). A good overall correlation was observed for levels of monomer, aggregates (dimer and multimers), and fragments as measured by the two analytical techniques (e.g., 6.0% vs. 5.3% and 14% vs. 11% for dimeric aggregates generated by elevated temperature/acidic pH and light exposure, respectively). The stability profile of a high concentration IgG1 monoclonal antibody formulation was investigated under stressed storage conditions (40 degrees C over 3 months) using the DW-SE-HPLC method including the loss of monomeric species with the concomitant accumulation of both aggregates and fragments. The nature and composition of the aggregates (primarily noncovalent dimers) and fragments (primarily loss of Fab from an intact IgG1) formed during storage were further characterized by a combination of LS measurements and mass spectroscopy analysis of deglycosylated IgG1 samples isolated by preparative SE-HPLC. The combination of DW-SE-HPLC, SV-AUC, LS, and mass spectroscopy results provided a detailed overall understanding the monomer, aggregate, fragment degradation pathway(s) for a high concentration IgG1 monoclonal antibody formulation during storage.
Collapse
Affiliation(s)
- Michael D Bond
- BioPharmaceutical Development and Marketed Product Support, Centocor R&D, Inc., 145 King of Prussia Road, Radnor, Pennsylvania 19087, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Human monoclonal antibodies (mAbs) have become drugs of choice for the management of an increasing number of human diseases. Human antibody repertoires provide a rich source for human mAbs. Here we review the characteristics of natural and non-natural human antibody repertoires and their mining with non-combinatorial and combinatorial strategies. In particular, we discuss the selection of human mAbs from naïve, immune, transgenic, and synthetic human antibody repertoires using methods based on hybridoma technology, clonal expansion of peripheral B cells, single-cell PCR, phage display, yeast display, and mammalian cell display. Our reliance on different strategies is shifting as we gain experience and refine methods to the efficient generation of human mAbs with superior pharmacokinetic and pharmacodynamic properties.
Collapse
Affiliation(s)
| | - Christoph Rader
- 2Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute
| |
Collapse
|
34
|
Beck A, Wagner-Rousset E, Wurch T, Corvaia N. [Therapeutic antibodies and related products: choosing the right structure for success]. Med Sci (Paris) 2010; 25:1024-32. [PMID: 20035674 DOI: 10.1051/medsci/200925121024] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Monoclonal antibodies (mAb) and related-products represent the fastest growing class of therapeutics in the biotechnological and pharmaceutical industry. In just as short as 20 years, more than 30 immunoglobulins (IgG) and derivatives have been approved in a wide range of indications (oncology, inflammation and auto-immunity, transplantation, angioplasty, hematology, ophthalmology, viral infections, allergy). The mAb structure toolbox contains mouse, chimeric, humanized and human antibodies from different isotypes (IgG1, 2 and 4), as well as IgG-related products (immunoconjugates, radio-immunoconjugates, Fab fragments, Fc-fusion proteins and peptides, bispecifics). Furthermore from a structural point of view, mAb glycosylation is linked to their production systems and may impact on their effector functions and immunogenicity. Based on the current knowledge, choosing the right antibody format, isotype and glycosylation profile are some of the key issues to address early during the lead selection.
Collapse
Affiliation(s)
- Alain Beck
- Centre d'immunologie Pierre Fabre (www.cipf.com), 5, avenue Napoléon III, 74160 Saint-Julien-en-Genevois, France.
| | | | | | | |
Collapse
|
35
|
Strategies and challenges for the next generation of therapeutic antibodies. Nat Rev Immunol 2010; 10:345-52. [PMID: 20414207 DOI: 10.1038/nri2747] [Citation(s) in RCA: 623] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Antibodies and related products are the fastest growing class of therapeutic agents. By analysing the regulatory approvals of IgG-based biotherapeutic agents in the past 10 years, we can gain insights into the successful strategies used by pharmaceutical companies so far to bring innovative drugs to the market. Many challenges will have to be faced in the next decade to bring more efficient and affordable antibody-based drugs to the clinic. Here, we discuss strategies to select the best therapeutic antigen targets, to optimize the structure of IgG antibodies and to design related or new structures with additional functions.
Collapse
|
36
|
Beck A, Reichert JM, Wurch T. 5th European Antibody Congress 2009: November 30–December 2, 2009, Geneva, Switzerland. MAbs 2010; 2:108-28. [PMID: 20179425 PMCID: PMC2840230 DOI: 10.4161/mabs.2.2.11302] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 01/26/2010] [Indexed: 12/15/2022] Open
Affiliation(s)
- Alain Beck
- Physico-Chemistry Department, Centre d’Immunologie Pierre Fabre, Saint Julien en Genevois, France.
| | | | | |
Collapse
|
37
|
|
38
|
Stubenrauch K, Wessels U, Regula JT, Kettenberger H, Schleypen J, Kohnert U. Impact of molecular processing in the hinge region of therapeutic IgG4 antibodies on disposition profiles in cynomolgus monkeys. Drug Metab Dispos 2010; 38:84-91. [PMID: 19850673 DOI: 10.1124/dmd.109.029751] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
The IgG4 isotype antibody is a potential candidate for immunotherapy when reduced effector functions are desirable. However, antigen binding fragment (Fab) arm exchange leads to functional monovalency with potentially reduced therapeutic efficacy. Mutagenesis studies suggested that the CH3 domain and not the core hinge is dominantly involved in in vivo molecular processing. This work investigated whether stabilization of the core hinge of a therapeutic IgG4 antibody by mutation of Ser228 to Pro (S228P) would be sufficient to prevent in vivo Fab arm exchange. In vitro experiments evaluated the influence of different levels of oxidation-reduction conditions in buffer and serum on Fab arm exchange (swapping) of wild-type (WT) IgG4 and IgG1 and of IgG4 S228P, which included a sterically neutral second mutation (Leu235 replaced by Glu). The objective of single-dose pharmacokinetic experiments in cynomolgus monkeys was to determine whether the mutation reduced IgG4 swapping in vivo. The results indicated that S228P mutation did not completely prevent Fab arm exchange in vitro in buffer under reducing conditions relative to IgG4 WT. The immunoassay findings were confirmed by mass spectrometry measurements. Results of the in vivo studies suggested that the therapeutic IgG4 WT antibody exchanged Fab arms with endogenous cynomolgus monkey IgG4, resulting in bispecific IgG4 antibodies with monovalency for the therapeutic target. In contrast, serum from cynomolgus monkeys dosed with the IgG4 mutant was virtually free of swapped IgG4. In conclusion, the results indicated that IgG4 swapping in vivo was markedly attenuated by S228P mutation.
Collapse
MESH Headings
- Amino Acid Substitution/genetics
- Amino Acid Substitution/immunology
- Animals
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/metabolism
- Antibodies, Bispecific/pharmacokinetics
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/therapeutic use
- Buffers
- Enzyme-Linked Immunosorbent Assay
- Female
- Hinge Exons/genetics
- Humans
- Immunoglobulin Fab Fragments/immunology
- Immunoglobulin Fab Fragments/metabolism
- Immunoglobulin G/genetics
- Immunoglobulin G/metabolism
- Immunoglobulin G/therapeutic use
- Macaca fascicularis
- Male
- Mice
- OX40 Ligand/immunology
- Oxidation-Reduction
- Rats
- Receptors, Interleukin-1/immunology
- Receptors, Interleukin-13/immunology
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Recombinant Proteins/metabolism
- Recombinant Proteins/pharmacokinetics
- Reducing Agents/metabolism
- Serum/immunology
- Serum/metabolism
- Spectrometry, Mass, Electrospray Ionization
Collapse
Affiliation(s)
- Kay Stubenrauch
- Departments of Bioanalytics, Pharma Research Penzberg, Roche Diagnostics, Nonnenwald 2, 82377 Penzberg, Germany.
| | | | | | | | | | | |
Collapse
|
39
|
Rathore AS. Follow-on protein products: scientific issues, developments and challenges. Trends Biotechnol 2009; 27:698-705. [PMID: 19846228 DOI: 10.1016/j.tibtech.2009.09.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Revised: 09/02/2009] [Accepted: 09/09/2009] [Indexed: 10/20/2022]
Abstract
Scientific and regulatory issues around approval of follow-on protein products, referred to as biosimilars in Europe, have been a topic of great interest and debate recently. The central issue is our limited understanding of how the different quality attributes of a product have an impact on its safety and efficacy. Crucial gaps in our knowledge include a lack of standardization in the way in which data are collected, analyzed and reported, and limitations in the ability of non-clinical tools for predicting clinical safety and efficacy. Complexity of protein products with respect to the numerous quality attributes and complexity of the biotechnology processes and the raw materials add to the challenges. In this paper, recommendations are presented to help at least partially alleviate these challenges.
Collapse
Affiliation(s)
- Anurag S Rathore
- Department of Chemical Engineering, Indian Institute of Technology, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
40
|
Hung TT, Wu JY, Liu JF, Cheng HC. Epitope analysis of the rat dipeptidyl peptidase IV monoclonal antibody 6A3 that blocks pericellular fibronectin-mediated cancer cell adhesion. FEBS J 2009; 276:6548-59. [DOI: 10.1111/j.1742-4658.2009.07352.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
41
|
Abstract
Over the past decade, monoclonal antibodies have dramatically impacted the treatment of haematological malignancies, as evidenced by the effect of rituximab on the response rate and survival of patients with follicular and diffuse large B cell non-Hodgkin's lymphoma. Currently, only two monoclonal antibodies - the anti-CD33 immunotoxin gemtuzumab ozogamicin and the CD52-directed antibody alemtuzumab - are approved for treatment of relapsed acute myeloid leukaemia in older patients and B cell chronic lymphocytic leukaemia, respectively. Although not approved for such treatment, alemtuzumab is also active against T cell prolymphocytic leukaemia, cutaneous T cell lymphoma and Sézary syndrome, and adult T cell leukaemia and lymphoma. In addition, rituximab has demonstrated activity against B cell chronic lymphocytic and hairy cell leukaemia. Monoclonal antibodies targeting CD4, CD19, CD20, CD22, CD23, CD25, CD45, CD66 and CD122 are now being studied in the clinic for the treatment of leukaemia. Here, we discuss how these new antibodies have been engineered to reduce immunogenicity and improve antibody targeting and binding. Improved interactions with Fc receptors on immune effector cells can enhance destruction of target cells through antibody-dependent cellular cytotoxicity and complement-mediated cell lysis. The antibodies can also be armed with cellular toxins or radionuclides to enhance the destruction of leukaemia cells.
Collapse
Affiliation(s)
- John C Morris
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892-1457, USA.
| | | |
Collapse
|
42
|
Gerspach J, Pfizenmaier K, Wajant H. Improving TNF as a cancer therapeutic: tailor-made TNF fusion proteins with conserved antitumor activity and reduced systemic side effects. Biofactors 2009; 35:364-72. [PMID: 19484741 DOI: 10.1002/biof.50] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tumor necrosis factor (TNF) is highly pleiotropic cytokine regulating diverse cellular processes such as proliferation, cell migration, angiogenesis, differentiation, apoptosis, necrosis, but also survival. Because of its name-giving tumor necrosis-inducing capabilities, TNF has attracted attention very early for antitumor therapy. Although TNF is in clinical use for treatment of soft tissue sarcoma in isolated limb perfusion, its broad use in tumor therapy is prevented so far by its strong systemic proinflammatory effects. Nevertheless, over the past decade, a variety of tailor-made TNF variants have been developed with the aim to reduce TNFs systemic activity without losing its antitumoral effects. Here, we review the progress made toward improving the efficacy of TNF by genetic engineering, tumor targeting, and introduction of prodrug concepts.
Collapse
Affiliation(s)
- Jeannette Gerspach
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.
| | | | | |
Collapse
|
43
|
Johnson PR, Schnepp BC, Zhang J, Connell MJ, Greene SM, Yuste E, Desrosiers RC, Clark KR. Vector-mediated gene transfer engenders long-lived neutralizing activity and protection against SIV infection in monkeys. Nat Med 2009; 15:901-6. [PMID: 19448633 PMCID: PMC2723177 DOI: 10.1038/nm.1967] [Citation(s) in RCA: 251] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Accepted: 04/11/2009] [Indexed: 12/24/2022]
Abstract
The holy grail for HIV vaccine development is an immunogen that elicits persisting antibodies with broad neutralizing activity against field strains of the virus. Unfortunately, very little progress has been made in finding or designing such immunogens. Using the SIV model, we have taken a markedly different approach: delivery of an adeno-associated virus (AAV) gene transfer vector to muscle for the expression of antibodies or antibody-like immunoadhesins having predetermined anti-SIV specificity. With this approach, anti-SIV molecules are endogenously synthesized in myofibers and passively distributed to the circulatory system. Using such an approach in monkeys, we have now generated long-lasting neutralizing activity in serum and observed complete protection against intravenous challenge with virulent SIV. In essence, this strategy bypasses the adaptive immune system and holds significant promise as a novel approach to an effective HIV vaccine.
Collapse
Affiliation(s)
- Philip R Johnson
- The Children's Hospital of Philadelphia and the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
The twenty two monoclonal antibodies (mAbs) currently marketed in the U.S. have captured almost half of the top-20 U.S. therapeutic biotechnology sales for 2007. Eight of these products have annual sales each of more than $1 B, were developed in the relatively short average period of six years, qualified for FDA programs designed to accelerate drug approval, and their cost has been reimbursed liberally by payers. With growth of the product class driven primarily by advancements in protein engineering and the low probability of generic threats, mAbs are now the largest class of biological therapies under development. The high cost of these drugs and the lack of generic competition conflict with a financially stressed health system, setting reimbursement by payers as the major limiting factor to growth. Advances in mAb engineering are likely to result in more effective mAb drugs and an expansion of the therapeutic indications covered by the class. The parallel development of biomarkers for identifying the patient subpopulations most likely to respond to treatment may lead to a more cost-effective use of these drugs. To achieve the success of the current top-tier mAbs, companies developing new mAb products must adapt to a significantly more challenging commercial environment.
Collapse
Affiliation(s)
- Pablo A Scolnik
- The Scolnik Group Biotechnology Consultants, LLC, Hillsborough, NC 27278, USA.
| |
Collapse
|
45
|
Beck A, Hanala S, Reichert JM. 4th European Antibody Congress 2008: December 1-3, 2008, Geneva, Switzerland. MAbs 2009; 1:93-103. [PMID: 20061813 DOI: 10.4161/mabs.1.2.7890] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The Fourth European Antibody meeting, organized by Terrapin Ltd., was held in Geneva, a center of the European biopharmaceutical industry. Merck-Serono, NovImmune, Pierre Fabre and Therapeomic are located nearby, as are R&D centers of Boehringer-Ingelheim, Novartis, Roche and Sanofi-Aventis. Over 40 speakers and more than 200 delegates attended the event. Companies represented included Abbott, Ablynx, Adnexus/ BMS, Astra-Zeneca/ CAT/ Medimmune, BiogenIdec, BioRad, Centocor (Johnson & Johnson), Crucell/ DSM, Domantis, Dyax, Genmab, Genzyme, Glycart/ Roche, Haptogen, Immunogen, Kyowa-Kirin, LFB, Medarex, Merck-Serono, Micromet, Novartis, Pierre Fabre Laboratories, Roche, Sanofi-Aventis, Seattle-Genetics, Transgene, UCB Celltech and Wyeth. Other attendees included those based in academe or government (University of Amsterdam, University of Zurich, University Hospital-Lyon, Ecole Polytechnique Federale de Lausanne, INSERM, Tufts University, US National Institutes of Health), consultants, and patent attorneys (Edwards, Angell, Palmer & Dodge). The meeting was very interactive and included exchanges during the many scheduled networking times (exhibitions, speed-networking, lunches and evening receptions). The first day of the three day conference was dedicated to advances in understanding antibody structure-function relationships. Challenges and opportunities in antibody development were the focus of the second day and the third day featured discussion of innovative antibodies and antibody alternatives.
Collapse
Affiliation(s)
- Alain Beck
- Department of Physico-Chemistry; Center of Immunology Pierre Fabre, Saint-Julienen-Genevois, France
| | | | | |
Collapse
|