1
|
Liu S, Nguyen JB, Zhao Y, Schussler S, Kim S, Qiu H, Li N, Rosconi MP, Pyles EA. Development of a platform method for rapid detection and characterization of domain-specific post-translational modifications in bispecific antibodies. J Pharm Biomed Anal 2024; 244:116120. [PMID: 38547650 DOI: 10.1016/j.jpba.2024.116120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/14/2024] [Accepted: 03/17/2024] [Indexed: 04/29/2024]
Abstract
Charge heterogeneity is inherent to all therapeutic antibodies and arises from post-translational modifications (PTMs) and/or protein degradation events that may occur during manufacturing. Among therapeutic antibodies, the bispecific antibody (bsAb) containing two unique Fab arms directed against two different targets presents an additional layer of complexity to the charge profile. In the context of a bsAb, a single domain-specific PTM within one of the Fab domains may be sufficient to compromise target binding and could potentially impact the stability, safety, potency, and efficacy of the drug product. Therefore, characterization and routine monitoring of domain-specific modifications is critical to ensure the quality of therapeutic bispecific antibody products. We developed a Digestion-assisted imaged Capillary isoElectric focusing (DiCE) method to detect and quantitate domain-specific charge variants of therapeutic bispecific antibodies (bsAbs). The method involves enzymatic digestion using immunoglobulin G (IgG)-degrading enzyme of S. pyogenes (IdeS) to generate F(ab)2 and Fc fragments, followed by imaged capillary isoelectric focusing (icIEF) under reduced, denaturing conditions to separate the light chains (LCs) from the Fd domains. Our results suggest that DiCE is a highly sensitive method that is capable of quantitating domain-specific PTMs of a bsAb. In one case study, DiCE was used to quantitate unprocessed C-terminal lysine and site-specific glycation of Lys98 in the complementarity-determining region (CDR) of a bsAb that could not be accurately quantitated using conventional, platform-based charge variant analysis, such as intact icIEF. Quantitation of these PTMs by DiCE was comparable to results from peptide mapping, demonstrating that DiCE is a valuable orthogonal method for ensuring product quality. This method may also have potential applications for characterizing fusion proteins, antibody-drug conjugates, and co-formulated antibody cocktails.
Collapse
Affiliation(s)
- Sophia Liu
- Protein Biochemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Jennifer B Nguyen
- Protein Biochemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States.
| | - Yimeng Zhao
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Svetlana Schussler
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Sunnie Kim
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Haibo Qiu
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Ning Li
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Michael P Rosconi
- Protein Biochemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Erica A Pyles
- Protein Biochemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| |
Collapse
|
2
|
Du Y, Xu J. Engineered Bifunctional Proteins for Targeted Cancer Therapy: Prospects and Challenges. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2103114. [PMID: 34585802 DOI: 10.1002/adma.202103114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 08/08/2021] [Indexed: 06/13/2023]
Abstract
Bifunctional proteins (BFPs) are a class of therapeutic agents produced through genetic engineering and protein engineering, and are increasingly used to treat various human diseases, including cancer. These proteins usually have two or more biological functions-specifically recognizing different molecular targets to regulate the related signaling pathways, or mediating effector molecules/cells to kill tumor cells. Unlike conventional small-molecule or single-target drugs, BFPs possess stronger biological activity but lower systemic toxicity. Hence, BFPs are considered to offer many benefits for the treatment of heterogeneous tumors. In this review, the authors briefly describe the unique structural feature of BFP molecules and innovatively divide them into bispecific antibodies, cytokine-based BFPs (immunocytokines), and protein toxin-based BFPs (immunotoxins) according to their mode of action. In addition, the latest advances in the development of BFPs are discussed and the potential limitations or problems in clinical applications are outlined. Taken together, future studies need to be centered on understanding the characteristics of BFPs for optimizing and designing more effective such drugs.
Collapse
Affiliation(s)
- Yue Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Jian Xu
- Laboratory of Molecular Biology, Center for Cancer Research, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
3
|
Khanna V, Panyam J, Griffith TS. Exploiting antibody biology for the treatment of cancer. Immunotherapy 2020; 12:255-267. [DOI: 10.2217/imt-2019-0118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Over the last decade, antibodies have become an important component in the arsenal of cancer therapeutics. High-specificity, low off-target effects, desirable pharmacokinetics and high success rate are a few of the many attributes that make antibodies amenable for development as drugs. To design antibodies for successful clinical applications, however, it is critical to have an understanding of their structure, functions, mechanisms of action and pharmacokinetic/pharmacodynamic properties. This review highlights some of these key aspects, as well as certain limitations encountered, with monoclonal antibody therapy. Further, we discuss rational combination therapies for clinical applications, some of which could help overcome the limitations.
Collapse
Affiliation(s)
- Vidhi Khanna
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jayanth Panyam
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- School of Pharmacy, Temple University, Philadelphia, PA 19140, USA
| | - Thomas S Griffith
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Urology, Universityof Minnesota, Minneapolis, MN 55455, USA
- Center for Immunology, Universityof Minnesota, Minneapolis, MN 55455, USA
- Microbiology, Immunology, & Cancer Biology Graduate Program, University of Minnesota,Minneapolis, MN 55455, USA
| |
Collapse
|
4
|
Schrade A, Bujotzek A, Spick C, Wagner M, Goerl J, Wezler X, Georges G, Kontermann RE, Brinkmann U. Back-to-Germline (B2G) Procedure for Antibody Devolution. Antibodies (Basel) 2019; 8:antib8030045. [PMID: 31544851 PMCID: PMC6784197 DOI: 10.3390/antib8030045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/13/2019] [Accepted: 08/16/2019] [Indexed: 11/16/2022] Open
Abstract
Bispecific antibodies (bsAbs) with avidity-enhanced specificity can be used to address target cells with increased specificity, ideally binding efficiently to cells that express two cognate antigens, yet not to cells that express only one of those. Building blocks required to generate such bsAbs are binders that recognize the two antigens with high specificity yet with various (including very low monovalent) affinities. The herein described ‘back-to-germline’ (B2G) procedure defines such derivatives. It converts parent antibodies with high specificity to derivatives that retain specificity but modulate affinity. The approach defines mutations to be introduced into antibody complementarity-determining regions (CDRs) regions without requiring structures of antibody-antigen complexes. Instead, it reverses the B-cell maturation process that increases affinities, with preference on CDR residues with high antigen contact probability. Placing germline residues at those positions generates VH and VL domains and Fv-combinations thereof that retain specificities but are ‘de-matured’ to different degrees. De-maturation influences on-rates and off-rates, and can produce entities with extremely low affinity for which binding can only be detected in bivalent formats. A comparison with alanine replacement in CDRs (so far, the most frequently applied technology) indicates that B2G may be more reliable/predictable without introduction of stickiness or poly-reactivity. The applicability for generating sets of affinity-modulated monospecific variants is exemplarily shown for antibodies that bind CD138, Her2/neu, and EGFR.
Collapse
Affiliation(s)
- Anja Schrade
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Alexander Bujotzek
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Christian Spick
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Martina Wagner
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Johannes Goerl
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Xenia Wezler
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Guy Georges
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, 82377 Penzberg, Germany
| | - Roland E Kontermann
- Institute of Cell Biology & Immunology, Stuttgart University, 70569 Stuttgart, Germany
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, 82377 Penzberg, Germany.
| |
Collapse
|
5
|
Jalilzadeh-Razin S, Mantegi M, Tohidkia MR, Pazhang Y, Pourseif MM, Barar J, Omidi Y. Phage antibody library screening for the selection of novel high-affinity human single-chain variable fragment against gastrin receptor: an in silico and in vitro study. ACTA ACUST UNITED AC 2019; 27:21-34. [PMID: 30607886 DOI: 10.1007/s40199-018-0233-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/03/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND As a membrane G protein coupled receptors (GPCRs) family, gastrin/cholecystokinin-2 receptor (CCK2R) plays a key role in the initiation and development of gastric cancer. OBJECTIVES Targeting CCK2R by immunotherapeutics such as single-chain variable fragments (scFvs) may provide an effective treatment modality against gastric cancer. Thus, the main objective of this study was to isolate scFvs specific to CCK2R. METHODS To isolate scFvs specific to the CCK2R, we capitalized on a semi-synthetic diverse phage antibody library (PAL) and a solution-phase biopanning process. The library was panned against a biotinylated peptide of the second extracellular loop (ECL2) of CCK2R. After four rounds of biopanning, the selected soluble scFv clones were screened by enzyme-linked immunosorbent assay (ELISA) and examined for specific binding to the peptide. The selected scFvs were purified using immobilized metal affinity chromatography (IMAC). The binding affinity and specificity of the scFvs were examined by the surface plasmon resonance (SPR), immunoblotting and flow cytometry assays and molecular docking using ZDOCK v3.0.2. RESULTS Ten different scFvs were isolated, which displayed binding affinity ranging from 0.68 to 8.0 (nM). Immunoblotting and molecular docking analysis revealed that eight scFvs were able to detect the denatured form of CCK2R protein. Of the isolated scFvs, two scFvs showed high-binding affinity to the human gastric adenocarcinoma AGS cells. CONCLUSIONS Based on our findings, a couple of the selected scFvs showed markedly high-binding affinity to immobilized CCK2R peptide and CCK2R-overexpressing AGS cells. Therefore, these scFvs are proposed to serve as targeting and/or treatment agents in the diagnosis and immunotherapy of CCK2R-positive tumors. Graphical abstract ᅟ.
Collapse
Affiliation(s)
- Sepideh Jalilzadeh-Razin
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Malihe Mantegi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Islamic Azad University of Urmia, Urmia, Iran
| | - Mohammad R Tohidkia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Yaghub Pazhang
- Department of Biochemistry, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Mohammad M Pourseif
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Tustian AD, Laurin L, Ihre H, Tran T, Stairs R, Bak H. Development of a novel affinity chromatography resin for platform purification of bispecific antibodies with modified protein a binding avidity. Biotechnol Prog 2018; 34:650-658. [PMID: 29464924 PMCID: PMC6099419 DOI: 10.1002/btpr.2622] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 02/16/2018] [Indexed: 12/30/2022]
Abstract
There is strong interest in the production of bispecific monoclonal antibodies that can simultaneously bind two distinct targets or epitopes to achieve novel mechanisms of action and efficacy. Regeneron's bispecific technology, based upon a standard IgG, consists of a heterodimer of two different heavy chains, and a common light chain. Coexpression of two heavy chains leads to the formation of two parental IgG impurities, the removal of which is facilitated by a dipeptide substitution in the Fc portion of one of the heavy chains that ablates Fc Protein A binding. Therefore, the affinity capture (Protein A) step of the purification process must perform both bulk capture and high resolution of these mAb impurities, a task current commercially available resins are not designed for. Resolution can be further impaired by the ability of Protein A to bind some antibodies in the variable region of the heavy chain (VH ). This article details development of a novel Protein A resin. This resin combines an alkali stable ligand with a base matrix exhibiting excellent mass transfer properties to allow high capacity single step capture and resolution of bispecific antibodies (bsAbs) with high yields. The developed resin, named MabSelect SuRe™ pcc, is implemented in GMP production processes for several bsAbs. © 2018 The Authors Biotechnology Progress published by Wiley Periodicals, Inc. on behalf of American Institute of Chemical Engineers Biotechnol. Prog., 34:650-658, 2018.
Collapse
Affiliation(s)
| | | | | | - Travis Tran
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, 10591
| | - Robert Stairs
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, 10591
| | - Hanne Bak
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, 10591
| |
Collapse
|
7
|
Petrilli R, Eloy JO, Lee RJ, Lopez RFV. Preparation of Immunoliposomes by Direct Coupling of Antibodies Based on a Thioether Bond. Methods Mol Biol 2018; 1674:229-237. [PMID: 28921442 DOI: 10.1007/978-1-4939-7312-5_19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Drug delivery is of paramount importance, since the drug needs to be delivered to a specific site, in adequate concentration, avoiding degradation in order to provide therapeutic efficacy. Different nanocarriers have been used over the years for this purpose and liposomes are well-established systems due to the high biocompatibility and the possibility to vehiculate both hydrophilic and lipophilic drugs. In order to circumvent the rapid clearance by the reticuloendothelial system and to avoid the healthy cells exposure to the drug, long circulating liposomes containing polyethyleneglycol (PEG) and functionalized liposomes for targeted delivery have been developed. Immunoliposomes consist of liposomes containing antibodies or antibody fragments attached at the membrane surface. This attachment can be performed using PEG lipids, containing a reactive terminal group such as maleimide and thiolated antibodies. Additionaly, the use of PEG chains as spacers increases antibody-antigen affinity, since the antibody is not shielded by the steric hindrance of PEG and also due to the correct orientation of antibodies for interaction with receptors on cell surface. In this chapter, we describe and discuss in details the protocol to prepare anti-epidermal growth factor receptor (anti-EGFR) and anti-human epidermal growth factor receptor 2 (anti-HER2) liposomes using cetuximab and trastuzumab as antibodies. We present the direct coupling method based on the maleimide thioether reaction for these immunoliposomes preparation and present some characterization steps and in vitro studies in cell culture which can be used for better understanding these nanocarriers.
Collapse
Affiliation(s)
- Raquel Petrilli
- School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Av. do Cafe s/n, 14040-903, Ribeirão Preto, SP, Brazil
| | - Josimar O Eloy
- School of Pharmaceutical Sciences, UNESP, Araraquara, Rodovia Araraquara-Jau, km. 1, 14801-902, Araraquara, SP, Brazil
- UNL, CONICET, FBCB, Cell Culture Laboratory, de Santa Fe, Argentina
| | - Robert J Lee
- College of Pharmacy, The Ohio State University, Columbus, 500 W. 12th Ave., Columbus, OH, 43210, USA
| | - Renata F V Lopez
- School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Av. do Cafe s/n, 14040-903, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
8
|
Yu J, Wang X, Xu T, Jin Q, Duan J, Wu J, Wu H, Xu T, Ye S. A rational approach to enhancing antibody Fc homodimer formation for robust production of antibody mixture in a single cell line. J Biol Chem 2017; 292:17885-17896. [PMID: 28878018 DOI: 10.1074/jbc.m116.771188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 08/19/2017] [Indexed: 11/06/2022] Open
Abstract
Combinations of different antibodies have been shown to be more effective for managing certain diseases than monotherapy. Co-expression of the antibody mixture in a single cell line is key to reducing complexity during antibody development and manufacturing. However, co-transfection of multiple light and heavy chains into cells often leads to production of mismatched, heterodimeric by-products that are inactive, making the development of co-expression systems that robustly and efficiently produce highly active antibody mixtures a high priority. In this study, we modified the CH3 domain interface of the antibody fragment crystallizable (Fc) region by changing several charge pairs to create electrostatic interactions favoring Fc homodimer formation and disfavoring Fc heterodimer formation. When co-expressed, these modified antibodies with altered charge polarity across the Fc dimer interface preferentially formed homodimers that fully preserved the functions of each component, rather than inactive heterodimers whose formation was reduced because of rationally designed repulsive interactions. We designed eight different combinations and experimentally screened the best one, which enabled us to produce a binary antibody mixture against the EGF receptor with a minimal heterodimer contaminant. We further determined the crystal structure of a triple-mutated Fc variant in the best combination, and we elucidated the molecular interactions favoring Fc homodimer over heterodimer formation, which provided a structural basis for further optimization. The approach presented here demonstrates the feasibility of rational antibody modification for efficient and consistent production of monoclonal antibody mixtures in a single cell line and thus broadens our options for manufacturing more effective antibody-based therapeutic agents.
Collapse
Affiliation(s)
- Jie Yu
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058 and
| | | | - Tao Xu
- Alphamab Co. Ltd., Suzhou 215125, China
| | - Qiuheng Jin
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058 and
| | - Jinyuan Duan
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058 and
| | - Jie Wu
- Alphamab Co. Ltd., Suzhou 215125, China
| | - Haiyan Wu
- Alphamab Co. Ltd., Suzhou 215125, China
| | - Ting Xu
- Alphamab Co. Ltd., Suzhou 215125, China
| | - Sheng Ye
- From the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058 and
| |
Collapse
|
9
|
Krah S, Sellmann C, Rhiel L, Schröter C, Dickgiesser S, Beck J, Zielonka S, Toleikis L, Hock B, Kolmar H, Becker S. Engineering bispecific antibodies with defined chain pairing. N Biotechnol 2017; 39:167-173. [PMID: 28137467 DOI: 10.1016/j.nbt.2016.12.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/21/2016] [Accepted: 12/23/2016] [Indexed: 01/07/2023]
Abstract
Bispecific IgG-like antibodies can simultaneously interact with two epitopes on the same or on different antigens. Therefore, these molecules facilitate novel modes of action, which cannot be addressed by conventional monospecific IgGs. However, the generation of such antibodies still appears to be demanding due to their specific architecture comprising four different polypeptide chains that need to assemble correctly. This review focusses on different strategies to circumvent this issue or to enforce a correct chain association with a focus on common-chain bispecific antibodies.
Collapse
Affiliation(s)
- Simon Krah
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany; Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany
| | - Carolin Sellmann
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany; Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany
| | - Laura Rhiel
- Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany
| | - Christian Schröter
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany; Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany
| | - Stephan Dickgiesser
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany; Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany
| | - Jan Beck
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany; Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany
| | - Stefan Zielonka
- Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany
| | - Lars Toleikis
- Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany
| | - Björn Hock
- Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany.
| | - Stefan Becker
- Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany.
| |
Collapse
|
10
|
Scott MJ, Lee JA, Wake MS, Batt KV, Wattam TA, Hiles ID, Batuwangala TD, Ashman CI, Steward M. 'In-Format' screening of a novel bispecific antibody format reveals significant potency improvements relative to unformatted molecules. MAbs 2016; 9:85-93. [PMID: 27786601 DOI: 10.1080/19420862.2016.1249078] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Bispecific antibodies (BsAbs) are emerging as an important class of biopharmaceutical. The majority of BsAbs are created from conventional antibodies or fragments engineered into more complex configurations. A recurring challenge in their development, however, is the identification of components that are optimised for inclusion in the final format in order to deliver both efficacy and robust biophysical properties. Using a modular BsAb format, the mAb-dAb, we assessed whether an 'in-format' screening approach, designed to select format-compatible domain antibodies, could expedite lead discovery. Human nerve growth factor (NGF) was selected as an antigen to validate the approach; domain antibody (dAb) libraries were screened, panels of binders identified, and binding affinities and potencies compared for selected dAbs and corresponding mAb-dAbs. A number of dAbs that exhibited high potency (IC50) when assessed in-format were identified. In contrast, the corresponding dAb monomers had ∼1000-fold lower potency than the formatted dAbs; such dAb monomers would therefore have been omitted from further characterization. Subsequent stoichiometric analyses of mAb-dAbs bound to NGF, or an additional target antigen (vascular endothelial growth factor), suggested different target binding modes; this indicates that the observed potency improvements cannot be attributed simply to an avidity effect offered by the mAb-dAb format. We conclude that, for certain antigens, screening naïve selection outputs directly in-format enables the identification of a subset of format-compatible dAbs, and that this offers substantial benefits in terms of molecular properties and development time.
Collapse
Affiliation(s)
| | | | | | - Kelly V Batt
- a Biopharm Discovery, GlaxoSmithKline , Stevenage , UK
| | | | - Ian D Hiles
- a Biopharm Discovery, GlaxoSmithKline , Stevenage , UK
| | | | | | | |
Collapse
|
11
|
Iida M, Bahrar H, Brand TM, Pearson HE, Coan JP, Orbuch RA, Flanigan BG, Swick AD, Prabakaran PJ, Lantto J, Horak ID, Kragh M, Salgia R, Kimple RJ, Wheeler DL. Targeting the HER Family with Pan-HER Effectively Overcomes Resistance to Cetuximab. Mol Cancer Ther 2016; 15:2175-86. [PMID: 27422810 PMCID: PMC5010956 DOI: 10.1158/1535-7163.mct-16-0012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 07/01/2016] [Indexed: 12/31/2022]
Abstract
Cetuximab, an antibody against the EGFR, has shown efficacy in treating head and neck squamous cell carcinoma (HNSCC), metastatic colorectal cancer, and non-small cell lung cancer (NSCLC). Despite the clinical success of cetuximab, many patients do not respond to cetuximab. Furthermore, virtually all patients who do initially respond become refractory, highlighting both intrinsic and acquired resistance to cetuximab as significant clinical problems. To understand mechanistically how cancerous cells acquire resistance, we previously developed models of acquired resistance using the H226 NSCLC and UM-SCC1 HNSCC cell lines. Cetuximab-resistant clones showed a robust upregulation and dependency on the HER family receptors EGFR, HER2, and HER3. Here, we examined pan-HER, a mixture of six antibodies targeting these receptors on cetuximab-resistant clones. In cells exhibiting acquired or intrinsic resistance to cetuximab, pan-HER treatment decreased all three receptors' protein levels and downstream activation of AKT and MAPK. This correlated with decreased cell proliferation in cetuximab-resistant clones. To determine whether pan-HER had a therapeutic benefit in vivo, we established de novo cetuximab-resistant mouse xenografts and treated resistant tumors with pan-HER. This regimen resulted in a superior growth delay of cetuximab-resistant xenografts compared with mice continued on cetuximab. Furthermore, intrinsically cetuximab-resistant HNSCC patient-derived xenograft tumors treated with pan-HER exhibited significant growth delay compared with vehicle/cetuximab controls. These results suggest that targeting multiple HER family receptors simultaneously with pan-HER is a promising treatment strategy for tumors displaying intrinsic or acquired resistance to cetuximab. Mol Cancer Ther; 15(9); 2175-86. ©2016 AACR.
Collapse
Affiliation(s)
- Mari Iida
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Harsh Bahrar
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin. Radboud Department of Radiation Oncology, University Medical Centre Nijmegen, Nijmegen, the Netherlands
| | - Toni M Brand
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Hannah E Pearson
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - John P Coan
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Rachel A Orbuch
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Bailey G Flanigan
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Adam D Swick
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Prashanth J Prabakaran
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | | | | | | | | | - Randy J Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Deric L Wheeler
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| |
Collapse
|
12
|
Fattore L, Malpicci D, Marra E, Belleudi F, Noto A, De Vitis C, Pisanu ME, Coluccia P, Camerlingo R, Roscilli G, Ribas A, Di Napoli A, Torrisi MR, Aurisicchio L, Ascierto PA, Mancini R, Ciliberto G. Combination of antibodies directed against different ErbB3 surface epitopes prevents the establishment of resistance to BRAF/MEK inhibitors in melanoma. Oncotarget 2016. [PMID: 26208478 PMCID: PMC4694796 DOI: 10.18632/oncotarget.4485] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Patients with metastatic melanoma bearing V600 mutations in BRAF oncogene clinically benefit from the treatment with BRAF inhibitors alone or in combination with MEK inhibitors. However, a limitation to such treatment is the occurrence of resistance. Tackling the adaptive changes helping cells survive from drug treatment may offer new therapeutic opportunities. Very recently the ErbB3 receptor has been shown to act as a central node promoting survival of BRAF mutated melanoma. In this paper we first demonstrate that ErbB3/AKT hyperphosphorylation occurs in BRAF mutated melanoma cell lines following exposure to BRAF and/or MEK inhibitors. This strongly correlates with increased transcriptional activation of its ligand neuregulin. Anti-ErbB3 antibodies impair the establishment of de novo cell resistance to BRAF inhibition in vitro. In order to more potently ablate ErbB3 activity we used a combination of two anti-ErbB3 antibodies directed against distinct epitopes of its extracellular domain. These two antibodies in combo with BRAF/MEK inhibitors potently inhibit in vitro cell growth and tumor regrowth after drug withdrawal in an in vivo xenograft model. Importantly, residual tumor masses from mice treated by the antibodies and BRAF/ERK inhibitors combo are characterized almost exclusively by large necrotic areas with limited residual areas of tumor growth. Taken together, our findings support the concept that triple therapy directed against BRAF/MEK/ErbB3 may be able to provide durable control of BRAF mutated metastatic melanoma.
Collapse
Affiliation(s)
- Luigi Fattore
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples, Italy.,Dipartimento di Chirurgia "P. Valdoni", Sapienza Università di Roma, Rome, Italy
| | - Debora Malpicci
- Dipartimento di Chirurgia "P. Valdoni", Sapienza Università di Roma, Rome, Italy.,Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Catanzaro "Magna Graecia", Catanzaro, Italy
| | | | - Francesca Belleudi
- Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy.,Istituto Pasteur Fondazione Cenci Bolognetti, Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy
| | - Alessia Noto
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples, Italy.,Dipartimento di Chirurgia "P. Valdoni", Sapienza Università di Roma, Rome, Italy
| | - Claudia De Vitis
- Dipartimento di Chirurgia "P. Valdoni", Sapienza Università di Roma, Rome, Italy.,Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy
| | - Maria Elena Pisanu
- Dipartimento di Chirurgia "P. Valdoni", Sapienza Università di Roma, Rome, Italy.,Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Pierpaolo Coluccia
- Dipartimento di Chirurgia "P. Valdoni", Sapienza Università di Roma, Rome, Italy
| | - Rosa Camerlingo
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples, Italy
| | - Giuseppe Roscilli
- Takis S.r.l., Rome, Italy.,Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy
| | - Antoni Ribas
- Department of Medicine, Division of Hematology/Oncology, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Arianna Di Napoli
- Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy.,Azienda Ospedaliera S. Andrea, Rome, Italy
| | - Maria Rosaria Torrisi
- Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy.,Istituto Pasteur Fondazione Cenci Bolognetti, Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy.,Azienda Ospedaliera S. Andrea, Rome, Italy
| | | | - Paolo Antonio Ascierto
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples, Italy
| | - Rita Mancini
- Dipartimento di Chirurgia "P. Valdoni", Sapienza Università di Roma, Rome, Italy.,Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy
| | - Gennaro Ciliberto
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples, Italy
| |
Collapse
|
13
|
Corti D, Kearns JD. Promises and pitfalls for recombinant oligoclonal antibodies-based therapeutics in cancer and infectious disease. Curr Opin Immunol 2016; 40:51-61. [PMID: 26995095 PMCID: PMC7127534 DOI: 10.1016/j.coi.2016.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 02/29/2016] [Accepted: 03/01/2016] [Indexed: 02/08/2023]
Abstract
Monoclonal antibodies (mAbs) have revolutionized the diagnosis and treatment of many human diseases and the application of combinations of mAbs has demonstrated improved therapeutic activity in both preclinical and clinical testing. Combinations of antibodies have several advantages such as the capacities to target multiple and mutating antigens in complex pathogens and to engage varied epitopes on multiple disease-related antigens (e.g. receptors) to overcome heterogeneity and plasticity. Oligoclonal antibodies are an emerging therapeutic format in which a novel antibody combination is developed as a single drug product. Here, we will provide historical context on the use of oligoclonal antibodies in oncology and infectious diseases and will highlight practical considerations related to their preclinical and clinical development programs.
Collapse
Affiliation(s)
| | - Jeffrey D Kearns
- Merrimack Pharmaceuticals, Inc., One Kendall Square, Suite B7201, Cambridge, MA 02139, USA.
| |
Collapse
|
14
|
Wu X, Sereno AJ, Huang F, Lewis SM, Lieu RL, Weldon C, Torres C, Fine C, Batt MA, Fitchett JR, Glasebrook AL, Kuhlman B, Demarest SJ. Fab-based bispecific antibody formats with robust biophysical properties and biological activity. MAbs 2016; 7:470-82. [PMID: 25774965 DOI: 10.1080/19420862.2015.1022694] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
A myriad of innovative bispecific antibody (BsAb) platforms have been reported. Most require significant protein engineering to be viable from a development and manufacturing perspective. Single-chain variable fragments (scFvs) and diabodies that consist only of antibody variable domains have been used as building blocks for making BsAbs for decades. The drawback with Fv-only moieties is that they lack the native-like interactions with CH1/CL domains that make antibody Fab regions stable and soluble. Here, we utilize a redesigned Fab interface to explore 2 novel Fab-based BsAbs platforms. The redesigned Fab interface designs limit heavy and light chain mixing when 2 Fabs are co-expressed simultaneously, thus allowing the use of 2 different Fabs within a BsAb construct without the requirement of one or more scFvs. We describe the stability and activity of a HER2×HER2 IgG-Fab BsAb, and compare its biophysical and activity properties with those of an IgG-scFv that utilizes the variable domains of the same parental antibodies. We also generated an EGFR × CD3 tandem Fab protein with a similar format to a tandem scFv (otherwise known as a bispecific T cell engager or BiTE). We show that the Fab-based BsAbs have superior biophysical properties compared to the scFv-based BsAbs. Additionally, the Fab-based BsAbs do not simply recapitulate the activity of their scFv counterparts, but are shown to possess unique biological activity.
Collapse
Key Words
- BiTE, bispecific T cell engager
- BsAb, bispecific antibody
- CD, circular dichroism
- DSC, differential scanning calorimetry
- Fab interface design
- Fab, antigen binding antibody fragment
- Fv, variable domains antibody fragment
- HC, antibody heavy chain
- IgG-Fab
- LC, antibody light chain
- LCMS, liquid chromatography with in-line mass spectrometry
- SEC-LC, size exclusion chromatography with in-line static light scattering
- T cell
- Tm, temperature at the midpoint of thermal unfolding
- bispecific antibody
- mAb, monoclonal antibody
- scFv, single chain Fv
- tandem Fab
Collapse
Affiliation(s)
- Xiufeng Wu
- a Eli Lilly Biotechnology Center ; San Diego , CA , USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Tustian AD, Endicott C, Adams B, Mattila J, Bak H. Development of purification processes for fully human bispecific antibodies based upon modification of protein A binding avidity. MAbs 2016; 8:828-38. [PMID: 26963837 PMCID: PMC4966828 DOI: 10.1080/19420862.2016.1160192] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
There is strong interest in the design of bispecific monoclonal antibodies (bsAbs) that can simultaneously bind 2 distinct targets or epitopes to achieve novel mechanisms of action and efficacy. Multiple bispecific formats have been proposed and are currently under development. Regeneron's bispecific technology is based upon a standard fully human IgG antibody in order to minimize immunogenicity and improve the pharmacokinetic profile. A single common light chain and 2 distinct heavy chains combine to form the bispecific molecule. One of the heavy chains contains a chimeric Fc sequence form (called Fc*) that ablates binding to Protein A via the constant region. As a result of co-expression of the 2 heavy chains and the common light chain, 3 products are created, 2 of which are homodimeric for the heavy chains and one that is the desired heterodimeric bispecific product. The Fc* sequence allows selective purification of the FcFc* bispecific product on commercially available affinity columns, due to intermediate binding affinity for Protein A compared to the high avidity FcFc heavy chain homodimer, or the weakly binding Fc*Fc* homodimer. This platform requires the use of Protein A chromatography in both a capture and polishing modality. Several challenges, including variable region Protein A binding, resin selection, selective elution optimization, and impacts upon subsequent non-affinity downstream unit operations, were addressed to create a robust and selective manufacturing process.
Collapse
Affiliation(s)
| | | | | | - John Mattila
- a Regeneron Pharmaceuticals Inc. , Tarrytown , NY , USA
| | - Hanne Bak
- a Regeneron Pharmaceuticals Inc. , Tarrytown , NY , USA
| |
Collapse
|
16
|
Mazor Y, Oganesyan V, Yang C, Hansen A, Wang J, Liu H, Sachsenmeier K, Carlson M, Gadre DV, Borrok MJ, Yu XQ, Dall'Acqua W, Wu H, Chowdhury PS. Improving target cell specificity using a novel monovalent bispecific IgG design. MAbs 2015; 7:377-89. [PMID: 25621507 PMCID: PMC4622537 DOI: 10.1080/19420862.2015.1007816] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Monovalent bispecific IgGs cater to a distinct set of mechanisms of action but are difficult to engineer and manufacture because of complexities associated with correct heavy and light chain pairing. We have created a novel design, “DuetMab,” for efficient production of these molecules. The platform uses knobs-into-holes (KIH) technology for heterodimerization of 2 distinct heavy chains and increases the efficiency of cognate heavy and light chain pairing by replacing the native disulfide bond in one of the CH1-CL interfaces with an engineered disulfide bond. Using two pairs of antibodies, cetuximab (anti-EGFR) and trastuzumab (anti-HER2), and anti-CD40 and anti-CD70 antibodies, we demonstrate that DuetMab antibodies can be produced in a highly purified and active form, and show for the first time that monovalent bispecific IgGs can concurrently bind both antigens on the same cell. This last property compensates for the loss of avidity brought about by monovalency and improves selectivity toward the target cell.
Collapse
Key Words
- ADCC, antibody-dependent cell-mediated cytotoxicity
- Biotechnology
- CDR, complementarity determining region
- CH1, 2 and 3-heavy chain constant domain 1, 2 and 3
- CL-, light chain constant domain
- DSC-differential scanning calorimetry
- E:T, ratio of effector to target cells
- EGFR
- EGFR, epidermal growth factor receptor
- FcRn, neonatal Fc receptor
- FcγR, receptor for IgG Fc
- HER2
- IGFR, insulin like growth factor receptor
- IL-6, interleukin 6
- IgG, Immunoglobulin G
- PNGase, protein N-glycanase
- Q1q, first component of complement 1
- RAGE, receptor for advanced glycosylation
- antibody engineering
- bispecific antibody
- cancer
- disulfide
- mAbs, monoclonal antibodies
- multi-targeting
Collapse
Affiliation(s)
- Yariv Mazor
- a Department of Antibody Discovery and Protein Engineering ; MedImmune ; Gaithersburg , MD USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Jacobsen HJ, Poulsen TT, Dahlman A, Kjær I, Koefoed K, Sen JW, Weilguny D, Bjerregaard B, Andersen CR, Horak ID, Pedersen MW, Kragh M, Lantto J. Pan-HER, an Antibody Mixture Simultaneously Targeting EGFR, HER2, and HER3, Effectively Overcomes Tumor Heterogeneity and Plasticity. Clin Cancer Res 2015; 21:4110-22. [PMID: 25908781 DOI: 10.1158/1078-0432.ccr-14-3312] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 04/01/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Accumulating evidence indicates a high degree of plasticity and compensatory signaling within the human epidermal growth factor receptor (HER) family, leading to resistance upon therapeutic intervention with HER family members. EXPERIMENTAL DESIGN/RESULTS We have generated Pan-HER, a mixture of six antibodies targeting each of the HER family members EGFR, HER2, and HER3 with synergistic pairs of antibodies, which simultaneously remove all three targets, thereby preventing compensatory tumor promoting mechanisms within the HER family. Pan-HER induces potent growth inhibition in a range of cancer cell lines and xenograft models, including cell lines with acquired resistance to therapeutic antibodies. Pan-HER is also highly efficacious in the presence of HER family ligands, indicating that it is capable of overcoming acquired resistance due to increased ligand production. All three target specificities contribute to the enhanced efficacy, demonstrating a distinct benefit of combined HER family targeting when compared with single-receptor targeting. CONCLUSIONS Our data show that simultaneous targeting of three receptors provides broader efficacy than targeting a single receptor or any combination of two receptors in the HER family, especially in the presence of HER family ligands. Pan-HER represents a novel strategy to deal with primary and acquired resistance due to tumor heterogeneity and plasticity in terms of HER family dependency and as such may be a viable alternative in the clinic.
Collapse
Affiliation(s)
| | | | | | - Ida Kjær
- Symphogen A/S, Ballerup, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Wilmes GM, Carey KL, Hicks SW, Russell HH, Stevenson JA, Kocjan P, Lutz SR, Quesenberry RS, Shulga-Morskoy SV, Lewis ME, Clark E, Medik V, Cooper AB, Reczek EE. Non-viral adeno-associated virus-based platform for stable expression of antibody combination therapeutics. MAbs 2014; 6:957-67. [PMID: 24758837 DOI: 10.4161/mabs.28917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Antibody combination therapeutics (ACTs) are polyvalent biopharmaceuticals that are uniquely suited for the control of complex diseases, including antibiotic resistant infectious diseases, autoimmune disorders and cancers. However, ACTs also represent a distinct manufacturing challenge because the independent manufacture and subsequent mixing of monoclonal antibodies quickly becomes cost prohibitive as more complex mixtures are envisioned. We have developed a virus-free recombinant protein expression platform based on adeno-associated viral (AAV) elements that is capable of rapid and consistent production of complex antibody mixtures in a single batch format. Using both multiplexed immunoassays and cation exchange (CIEX) chromatography, cell culture supernatants generated using our system were assessed for stability of expression and ratios of the component antibodies over time. Cultures expressing combinations of three to ten antibodies maintained consistent expression levels and stable ratios of component antibodies for at least 60 days. Cultures showed remarkable reproducibility following cell banking, and AAV-based cultures showed higher stability and productivity than non-AAV based cultures. Therefore, this non-viral AAV-based expression platform represents a predictable, reproducible, quick and cost effective method to manufacture or quickly produce for preclinical testing recombinant antibody combination therapies and other recombinant protein mixtures.
Collapse
|
19
|
Abstract
Therapeutic antibodies have been used since the end of nineteenth century, but their use is progressively increased and recently, with the availability of monoclonal antibodies, they are successfully employed in a large disease spectrum, which transversally covers different fields of medicine. Hyperimmune polyclonal immune globulin has been used against infectious diseases, in a period in which anti-microbial drugs were not yet available, and it still maintains a relevant place in prophylaxis/therapy. Although immune globulin should be considered life-saving as replacement therapy in humoral immunodeficiencies, its place in the immune-modulating treatment is not usually first-choice, but it should be considered as support to standard approved treatments. Despite therapeutic monoclonal antibodies have been lastly introduced in therapy, their extreme potentiality is reflected by the large number of approved molecules, addressed toward different immunological targets and able to heavily influence the prognosis and quality of life of a wide range of different diseases.
Collapse
Affiliation(s)
- Simonetta Salemi
- Sapienza Università di Roma -Facoltà di Medicina e Psicologia , Azienda Ospedaliera S. Andrea, Roma , Italy
| | | | | | | |
Collapse
|
20
|
Abstract
Antibody therapeutics are one of the fastest growing classes of pharmaceuticals, with an annual US market over $20 billion, developed to treat a variety of diseases including cancer, auto-immune and infectious diseases. Most are currently administered as a single molecule to treat a single disease, however there is mounting evidence that cocktails of multiple antibodies, each with a unique binding specificity and protective mechanism, may improve clinical efficacy. Here, we review progress in the development of oligoclonal combinations of antibodies to treat disease, focusing on identification of synergistic antibodies. We then discuss the application of modern antibody engineering technologies to produce highly potent antibody preparations, including oligoclonal antibody cocktails and truly recombinant polyclonal antibodies. Specific examples illustrating the synergy conferred by multiple antibodies will be provided for diseases caused by botulinum toxin, cancer and immune thrombocytopenia. The bioprocessing and regulatory options for these preparations will be discussed.
Collapse
Affiliation(s)
- Xian-zhe Wang
- Department of Chemistry, University of Texas at Austin, Austin, TX 78712, United States
| | - Vincent W Coljee
- Department of Physics, Harvard University, Cambridge, MA 02138, United States
| | - Jennifer A Maynard
- Chemical Engineering, University of Texas at Austin, Austin, TX 78712, United States
| |
Collapse
|
21
|
Woods RJ, Xie MH, Von Kreudenstein TS, Ng GYK, Dixit SB. LC-MS characterization and purity assessment of a prototype bispecific antibody. MAbs 2013; 5:711-22. [PMID: 23884083 PMCID: PMC3851224 DOI: 10.4161/mabs.25488] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Bispecific IgG asymmetric (heterodimeric) antibodies offer enhanced therapeutic efficacy, but present unique challenges for drug development. These challenges are related to the proper assembly of heavy and light chains. Impurities such as symmetric (homodimeric) antibodies can arise with improper assembly. A new method to assess heterodimer purity of such bispecific antibody products is needed because traditional separation-based purity assays are unable to separate or quantify homodimer impurities. This paper presents a liquid chromatography-mass spectrometry (LC-MS)-based method for evaluating heterodimeric purity of a prototype asymmetric antibody containing two different heavy chains and two identical light chains. The heterodimer and independently expressed homodimeric standards were characterized by two complementary LC-MS techniques: Intact protein mass measurement of deglycosylated antibody and peptide map analyses. Intact protein mass analysis was used to check molecular integrity and composition. LC-MSE peptide mapping of Lys-C digests was used to verify protein sequences and characterize post-translational modifications, including C-terminal truncation species. Guided by the characterization results, a heterodimer purity assay was demonstrated by intact protein mass analysis of pure deglycosylated heterodimer spiked with each deglycosylated homodimeric standard. The assay was capable of detecting low levels (2%) of spiked homodimers in conjunction with co-eluting half antibodies and multiple mass species present in the homodimer standards and providing relative purity differences between samples. Detection of minor homodimer and half-antibody C-terminal truncation species at levels as low as 0.6% demonstrates the sensitivity of the method. This method is suitable for purity assessment of heterodimer samples during process and purification development of bispecific antibodies, e.g., clone selection.
Collapse
|
22
|
Rasmussen SK, Næsted H, Müller C, Tolstrup AB, Frandsen TP. Recombinant antibody mixtures: Production strategies and cost considerations. Arch Biochem Biophys 2012; 526:139-45. [DOI: 10.1016/j.abb.2012.07.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 06/19/2012] [Accepted: 07/06/2012] [Indexed: 10/28/2022]
|
23
|
Kanakaraj P, Puffer BA, Yao XT, Kankanala S, Boyd E, Shah RR, Wang G, Patel D, Krishnamurthy R, Kaithamana S, Smith RG, LaFleur DW, Barbas CF, Hilbert DM, Kiener PA, Roschke VV. Simultaneous targeting of TNF and Ang2 with a novel bispecific antibody enhances efficacy in an in vivo model of arthritis. MAbs 2012; 4:600-13. [PMID: 22864384 PMCID: PMC3499301 DOI: 10.4161/mabs.21227] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Despite the clinical success of anti-tumor necrosis factor (TNF) therapies in the treatment of inflammatory conditions such as rheumatoid arthritis, Crohn disease and psoriasis, full control of the diseases only occurs in a subset of patients and there is a need for new therapeutics with improved efficacy against broader patient populations. One possible approach is to combine biological therapeutics, but both the cost of the therapeutics and the potential for additional toxicities needs to be considered. In addition to the various mediators of immune and inflammatory pathways, angiogenesis is reported to contribute substantially to the overall pathogenesis of inflammatory diseases. The combination of an anti-angiogenic agent with anti-TNF into one molecule could be more efficacious without the risk of severe immunosuppression. To evaluate this approach with our Zybody technology, we generated bispecific antibodies that contain an Ang2 targeting peptide genetically fused to the anti-TNF antibody adalimumab (Humira®). The bispecific molecules retain the binding and functional characteristics of the anti-TNF antibody, but with additional activity that neutralizes Ang2. In a TNF transgenic mouse model of arthritis, the bispecific anti-TNF-Ang2 molecules showed a dose-dependent reduction in both clinical symptoms and histological scores that were significantly better than that achieved by adalimumab alone.
Collapse
|
24
|
Kelton C, Wesolowski JS, Soloviev M, Schweickhardt R, Fischer D, Kurosawa E, McKenna SD, Gross AW. Anti-EGFR biparatopic-SEED antibody has enhanced combination-activity in a single molecule. Arch Biochem Biophys 2012; 526:219-25. [PMID: 22426455 DOI: 10.1016/j.abb.2012.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 03/02/2012] [Accepted: 03/04/2012] [Indexed: 01/11/2023]
Abstract
Certain combinations of non-competitive anti-EGFR antibodies have been reported to produce new effects on cells compared to either antibody used separately. New and enhanced combination-activity includes increased inhibition of signaling, increased receptor internalization and degradation, reduced proliferation of tumor cell lines and induction of complement-dependent cytotoxicity (CDC) effector function. To test requirements and mechanisms to elicit enhanced combination-activity with different EGFR binding domains, we created an anti-EGFR biparatopic antibody. A biparatopic antibody interacts through two different antigen-binding sites to a single antigen. A heterodimeric antibody with one binding domain derived from the C225 antibody and one binding domain derived from the humanized 425 (hu425) antibody was built on the strand-exchange engineered domain (SEED) scaffold. This anti-EGFR biparatopic-SEED antibody was compared to parental antibodies used alone and in combination, and to the corresponding monovalent anti-EGFR-SEED antibodies used alone or in combination. We found that the anti-EGFR biparatopic-SEED had enhanced activity, similar to the combination of the two parental antibodies. Combinations of monovalent anti-EGFR-SEED antibodies did not produce enhanced effectiveness in cellular assays. Our results show that the anti-EGFR biparatopic antibody created using the SEED scaffold has enhanced combination-activity in a single molecule. Furthermore, these data suggest that the potential to cross-link the two different epitopes is an important requirement in the mechanism of enhanced combination-activity.
Collapse
Affiliation(s)
- Christie Kelton
- EMD Serono Research Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Monoclonal antibodies are widely used for the treatment of cancer, inflammatory and infectious diseases and other disorders. Most of the marketed antibodies are monospecific and therefore capable of interacting and interfering with a single target. However, complex diseases are often multifactorial in nature, and involve redundant or synergistic action of disease mediators or upregulation of different receptors, including crosstalk between their signaling networks. Consequently, blockade of multiple, different pathological factors and pathways may result in improved therapeutic efficacy. This result can be achieved by combining different drugs, or use of the dual targeting strategies applying bispecific antibodies that have emerged as an alternative to combination therapy. This review discusses the various dual targeting strategies for which bispecific antibodies have been developed and provides an overview of the established bispecific antibody formats.
Collapse
Affiliation(s)
- Roland E Kontermann
- Institut für Zellbiologie und Immunologie; Universität Stuttgart; Stuttgart, Germany
| |
Collapse
|