1
|
Liu J, Zhu J. Progresses of T-cell-engaging bispecific antibodies in treatment of solid tumors. Int Immunopharmacol 2024; 138:112609. [PMID: 38971103 DOI: 10.1016/j.intimp.2024.112609] [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: 05/27/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/08/2024]
Abstract
T-cell-engaging bispecific antibody (TCB) therapies have emerged as a promising immunotherapeutic approach, effectively redirecting effector T cells to selectively eliminate tumor cells. The therapeutic potential of TCBs has been well recognized, particularly with the approval of multiple TCBs in recent years for the treatment of hematologic malignancies as well as some solid tumors. However, TCBs encounter multiple challenges in treating solid tumors, such as on-target off-tumor toxicity, cytokine release syndrome (CRS), and T cell dysfunction within the immunosuppressive tumor microenvironment, all of which may impact their therapeutic efficacy. In this review, we summarize clinical data on TCBs for solid tumor treatment, highlight the challenges faced, and discuss potential solutions based on emerging strategies from current clinical and preclinical research. These solutions include TCB structural optimization, target selection, and combination strategies. This comprehensive analysis aims to guide the development of TCBs from design to clinical application, addressing the evolving landscape of cancer immunotherapy.
Collapse
Affiliation(s)
- Junjun Liu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianwei Zhu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; Jecho Laboratories, Inc., Frederick, MD 21704, USA.
| |
Collapse
|
2
|
Sebastião MJ, Hoffman M, Escandell J, Tousi F, Zhang J, Figueroa B, DeMaria C, Gomes-Alves P. Identification of Mispairing Omic Signatures in Chinese Hamster Ovary (CHO) Cells Producing a Tri-Specific Antibody. Biomedicines 2023; 11:2890. [PMID: 38001891 PMCID: PMC10669571 DOI: 10.3390/biomedicines11112890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023] Open
Abstract
Monoclonal antibody-based therapy has shown efficacy against cancer, autoimmune, infectious, and inflammatory diseases. Multispecific antibodies (MsAbs), including trispecifics (tsAbs), offer enhanced therapeutic potential by targeting different epitopes. However, when co-expressed from three or more different polypeptide chains, MsAb production can lead to incorrect chain assembly and co-production of mispaired species with impaired biological activity. Moreover, mispairing carries significant challenges for downstream purification, decreasing yields and increasing the cost of bioprocess development. In this study, quantitative transcriptomics and proteomics analyses were employed to investigate which signaling pathways correlated with low and high mispairing clone signatures. Gene and protein expression profiles of Chinese hamster ovary (CHO) clones producing an tsAb were analyzed in the exponential growth and stationary (tsAb production) phase of fed-batch culture. Functional analysis revealed activated endoplasmic reticulum stress in high mispairing clones in both culture phases, while low mispairing clones exhibited expression profiles indicative of activated protein translation, as well as higher endocytosis and target protein degradation, suggesting the clearance of unfolded proteins through ubiquitin-mediated mechanisms. In addition, through transcriptomic profiling, we identified a group of genes that have the potential to be used as a biomarker panel tool for identifying high mispairing levels in the early stages of bioprocess development.
Collapse
Affiliation(s)
- Maria João Sebastião
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; (M.J.S.)
- ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Michael Hoffman
- Sanofi Cell Line and Cell Bank Development, Mammalian Platform, Global CMC Development, Framingham, MA 01701, USA (B.F.)
| | - José Escandell
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; (M.J.S.)
- ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Fatemeh Tousi
- Sanofi Bioanalytics Development, Global CMC Development, Framingham, MA 01701, USA
| | - Jin Zhang
- Sanofi Cell Line and Cell Bank Development, Mammalian Platform, Global CMC Development, Framingham, MA 01701, USA (B.F.)
| | - Bruno Figueroa
- Sanofi Cell Line and Cell Bank Development, Mammalian Platform, Global CMC Development, Framingham, MA 01701, USA (B.F.)
| | - Christine DeMaria
- Sanofi Cell Line and Cell Bank Development, Mammalian Platform, Global CMC Development, Framingham, MA 01701, USA (B.F.)
| | - Patrícia Gomes-Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; (M.J.S.)
- ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
3
|
Koga H, Yamano T, Betancur J, Nagatomo S, Ikeda Y, Yamaguchi K, Nabuchi Y, Sato K, Teranishi-Ikawa Y, Sato M, Hirayama H, Hayasaka A, Torizawa T, Haraya K, Sampei Z, Shiraiwa H, Kitazawa T, Igawa T, Kuramochi T. Efficient production of bispecific antibody by FAST-Ig TM and its application to NXT007 for the treatment of hemophilia A. MAbs 2023; 15:2222441. [PMID: 37339067 PMCID: PMC10283433 DOI: 10.1080/19420862.2023.2222441] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/22/2023] Open
Abstract
Efficient production of bispecific antibodies (BsAbs) in single mammalian cells is essential for basic research and industrial manufacturing. However, preventing unwanted pairing of heavy chains (HCs) and light chains (LCs) is a challenging task. To address this, we created an engineering technology for preferential cognate HC/LC and HC/HC paring called FAST-Ig (Four-chain Assembly by electrostatic Steering Technology - Immunoglobulin), and applied it to NXT007, a BsAb for the treatment of hemophilia A. We introduced charged amino-acid substitutions at the HC/LC interface to facilitate the proper assembly for manufacturing a standard IgG-type BsAb. We generated CH1/CL interface-engineered antibody variants that achieved > 95% correct HC/LC pairing efficiency with favorable pharmacological properties and developability. Among these, we selected a design (C3) that allowed us to separate the mis-paired species with an unintended pharmacological profile using ion-exchange chromatography. Crystal structure analysis demonstrated that the C3 design did not affect the overall structure of both Fabs. To determine the final design for HCs-heterodimerization, we compared the stability of charge-based and knobs into hole-based Fc formats in acidic conditions and selected the more stable charge-based format. FAST-Ig was also applicable to stable CHO cell lines for industrial production and demonstrated robust chain pairing with different subclasses of parent BsAbs. Thus, it can be applied to a wide variety of BsAbs both preclinically and clinically.
Collapse
Affiliation(s)
- Hikaru Koga
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Takashi Yamano
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Juan Betancur
- API Process Development Department, Chugai Pharmaceutical Co., Ltd, Ukima, Tokyo, Japan
| | - Satoko Nagatomo
- Analytical Development Department, Chugai Pharmaceutical Co, Ltd, Ukima, Tokyo, Japan
| | - Yousuke Ikeda
- Analytical Development Department, Chugai Pharmaceutical Co, Ltd, Ukima, Tokyo, Japan
| | - Kazuki Yamaguchi
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Yoshiaki Nabuchi
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Kazuki Sato
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | | | - Motohiko Sato
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Hiroyuki Hirayama
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Akira Hayasaka
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Takuya Torizawa
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Kenta Haraya
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Zenjiro Sampei
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Hirotake Shiraiwa
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Takehisa Kitazawa
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Tomoyuki Igawa
- Translational Research Division, Chugai Pharmaceutical Co., Ltd, Chuo-Ku, Tokyo, Japan
| | - Taichi Kuramochi
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| |
Collapse
|
4
|
Bispecific Antibodies: A Novel Approach for the Treatment of Solid Tumors. Pharmaceutics 2022; 14:pharmaceutics14112442. [PMID: 36432631 PMCID: PMC9694302 DOI: 10.3390/pharmaceutics14112442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Advancement in sequencing technologies allows for the identification of molecular pathways involved in tumor progression and treatment resistance. Implementation of novel agents targeting these pathways, defined as targeted therapy, significantly improves the prognosis of cancer patients. Targeted therapy also includes the use of monoclonal antibodies (mAbs). These drugs recognize specific oncogenic proteins expressed in cancer cells. However, as with many other types of targeting agents, mAb-based therapy usually fails in the long-term control of cancer progression due to the development of resistance. In many cases, resistance is caused by the activation of alternative pathways involved in cancer progression and the development of immune evasion mechanisms. To overcome this off-target resistance, bispecific antibodies (bsAbs) were developed to simultaneously target differential oncogenic pathway components, tumor-associated antigens (TAA) and immune regulatory molecules. As a result, in the last few years, several bsAbs have been tested or are being tested in cancer patients. A few of them are currently approved for the treatment of some hematologic malignancies but no bsAbs are approved in solid tumors. In this review, we will provide an overview of the state-of-the-art of bsAbs for the treatment of solid malignancies outlining their classification, design, main technologies utilized for production, mechanisms of action, updated clinical evidence and potential limitations.
Collapse
|
5
|
Segués A, Huang S, Sijts A, Berraondo P, Zaiss DM. Opportunities and challenges of bi-specific antibodies. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 369:45-70. [PMID: 35777864 DOI: 10.1016/bs.ircmb.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The recent clinical approval of different Bi-specific antibodies (BsAbs) has revealed the great therapeutic potential of this novel class of biologicals. For example, the bispecific T-cell engager (BiTE), Blinatumomab, demonstrated the unique capacity of BsAbs to link T-cells with tumor cells, inducing targeted tumor cell removal. Additionally, Amivantamab, recognizing the EGFR and cMet in cis, revealed a substantial improvement of therapeutic efficacy by concomitantly targeting two tumor antigens. Cis-targeting BsAbs furthermore allow discerning cell populations which concurrently express two antigens, for which each antigen expression pattern in itself might not be selective. In this way, BsAbs harbor the great prospect of being more specific and showing fewer side effects than monoclonal antibodies. Nevertheless, BsAbs have also faced major obstacles, for instance, in ensuring reliable assembly and clinical-grade purification. In this review, we summarize the different available antibody platforms currently used for the generation of IgG-like and non-IgG-like BsAbs and explain which approaches have been used to assemble those BsAbs which are currently approved for clinical application. By focusing on the example of regulatory T-cells (Tregs) and the different, ongoing approaches to develop BsAbs specifically targeting Tregs within the tumor microenvironment, our review highlights the huge potential as well as the pitfalls BsAb face in order to emerge as one of the most effective therapeutic biologicals targeting desired cell populations in a highly selective way. Such BsAb may improve treatment efficacy and reduce side effects, thereby opening novel treatment opportunities for a range of different diseases, such as cancer or autoimmune diseases.
Collapse
Affiliation(s)
- Aina Segués
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom; Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Shuyu Huang
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom; Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Alice Sijts
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Dietmar M Zaiss
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom; Department of Immune Medicine, University Regensburg, Regensburg, Germany; Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany; Institute of Pathology, University Regensburg, Regensburg, Germany.
| |
Collapse
|
6
|
Dettling DE, Kwok E, Quach L, Datt A, Degenhardt JD, Panchal A, Seto P, Krakow JL, Wall R, Hillier BJ, Zhu Y, Vinogradova M, DuBridge RB, May C. Regression of EGFR positive established solid tumors in mice with the conditionally active T cell engager TAK-186. J Immunother Cancer 2022; 10:jitc-2021-004336. [PMID: 35728872 PMCID: PMC9214390 DOI: 10.1136/jitc-2021-004336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2022] [Indexed: 11/16/2022] Open
Abstract
Background Despite clinical success with T cell engagers (TCEs) targeting hematological malignancies, achieving a safe and efficacious dose in patients with solid tumors remains challenging. Due to potency, low levels of target antigen expression on normal tissues may not be tolerated. To overcome this, we engineered a novel conditionally active TCE design called COBRA (Conditional Bispecific Redirected Activation). Administered as prodrugs, COBRAs bind to cell surface antigens on both normal and tumor tissues but are preferentially activated within the tumor microenvironment. Methods A COBRA was engineered to target EGFR, TAK-186. The potency of precleaved TAK-186 relative to a non-cleavable control was assessed in vitro. Mice bearing established solid tumors expressing a range of EGFR levels were administered a single bolus of human T cells, and concurrently treated with TAK-186 and associated controls intravenously. We assessed the plasma and tumor exposure of intact and cleaved TAK-186. Results TAK-186 shows potent redirected T cell killing of antigen expressing tumor cells. In vivo efficacy studies demonstrate regressions of established solid tumors, dependent on intratumoral COBRA cleavage. Pharmacokinetic studies reveal TAK-186 is stable in circulation, but once activated is rapidly cleared due to loss of its albumin-binding half-life extension domain. Conclusions The studies shown support the advancement of TAK-186, and the pursuit of additional COBRA TCEs for the treatment of solid tumors.
Collapse
Affiliation(s)
- Danielle E Dettling
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Eilene Kwok
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Lucy Quach
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Aakash Datt
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Jeremiah D Degenhardt
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Anand Panchal
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Pui Seto
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Jessica L Krakow
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Russell Wall
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Brian J Hillier
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Ying Zhu
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Maia Vinogradova
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Robert B DuBridge
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| | - Chad May
- Oncology Drug Development Unit, Takeda Development Centers America, Inc (TDCA), Lexington, Massachusetts, USA
| |
Collapse
|
7
|
Panchal A, Seto P, Wall R, Hillier BJ, Zhu Y, Krakow J, Datt A, Pongo E, Bagheri A, Chen THT, Degenhardt JD, Culp PA, Dettling DE, Vinogradova MV, May C, DuBridge RB. COBRA™: a highly potent conditionally active T cell engager engineered for the treatment of solid tumors. MAbs 2021; 12:1792130. [PMID: 32684124 PMCID: PMC7531513 DOI: 10.1080/19420862.2020.1792130] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Conditionally active COBRA™ (COnditional Bispecific Redirected Activation) T cell engagers are engineered to overcome the limitations of inherently active first-generation T cell engagers, which are unable to discern between tumor and healthy tissues. Designed to be administered as prodrugs, COBRAs target cell surface antigens upon administration, but engage T cells only after they are activated within the tumor microenvironment (TME). This allows COBRAs to be preferentially turned on in tumors while safely remaining inactive in healthy tissue. Here, we describe the development of the COBRA design and the characterization of these conditionally active T cell engagers. Upon administration COBRAs are engineered to bind to tumor-associated antigens (TAAs) and serum albumin (to extend their half-life in circulation), but are inhibited from interacting with the T cell receptor complex signaling molecule CD3. In the TME, a matrix metalloproteinase (MMP)-mediated linker cleavage event occurs within the COBRA construct, which rearranges the molecule, allowing it to co-engage TAAs and CD3, thereby activating T cells against the tumor. COBRAs are conditionally activated through cleavage with MMP9, and once active are highly potent, displaying sub-pM EC50s in T cell killing assays. Studies in tumor-bearing mice demonstrate COBRA administration completely regresses established solid tumor xenografts. These results strongly support the further characterization of the novel COBRA design in preclinical development studies.
Collapse
Affiliation(s)
- Anand Panchal
- Research and Discovery, Maverick Therapeutics , Brisbane, CA, USA
| | - Pui Seto
- Research and Discovery, Maverick Therapeutics , Brisbane, CA, USA
| | - Russell Wall
- Research and Discovery, Maverick Therapeutics , Brisbane, CA, USA
| | - Brian J Hillier
- Research and Discovery, Maverick Therapeutics , Brisbane, CA, USA
| | - Ying Zhu
- Research and Discovery, Maverick Therapeutics , Brisbane, CA, USA
| | - Jessica Krakow
- Research and Discovery, Maverick Therapeutics , Brisbane, CA, USA
| | - Aakash Datt
- Research and Development, Maverick Therapeutics , Brisbane, CA, USA
| | - Elizabeth Pongo
- Research and Discovery, Maverick Therapeutics , Brisbane, CA, USA
| | - Andisheh Bagheri
- Research and Discovery, Maverick Therapeutics , Brisbane, CA, USA
| | - Tseng-Hui T Chen
- Research and Discovery, Maverick Therapeutics , Brisbane, CA, USA
| | | | - Patricia A Culp
- Research and Discovery, Maverick Therapeutics , Brisbane, CA, USA
| | | | | | - Chad May
- Research and Development, Maverick Therapeutics , Brisbane, CA, USA
| | | |
Collapse
|
8
|
Hosseini SS, Khalili S, Baradaran B, Bidar N, Shahbazi MA, Mosafer J, Hashemzaei M, Mokhtarzadeh A, Hamblin MR. Bispecific monoclonal antibodies for targeted immunotherapy of solid tumors: Recent advances and clinical trials. Int J Biol Macromol 2020; 167:1030-1047. [PMID: 33197478 DOI: 10.1016/j.ijbiomac.2020.11.058] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023]
Abstract
Bispecific antibodie (BsAbs) combine two or more epitope-recognizing sequences into a single protein molecule. The first therapeutic applications of BsAbs were focused on cancer therapy. However, these antibodies have grown to cover a wider disease spectrum, including imaging, diagnosis, prophylaxis, and therapy of inflammatory and autoimmune diseases. BsAbs can be categorized into IgG-like formats and non-IgG-like formats. Different technologies have been used for the construction of BsAbs including "CrossMAb", "Quadroma", "knobs-into-holes" and molecular cloning. The mechanism of action for BsAbs includes the induction of CDC, ADCC, ADCP, apoptosis, and recruitment of cell surface receptors, as well as activation or inhibition of signaling pathways. The first clinical trials included mainly leukemia and lymphoma, but solid tumors are now being investigated. The BsAbs bind to a tumor-specific antigen using one epitope, while the second epitope binds to immune cell receptors such as CD3, CD16, CD64, and CD89, with the goal of stimulating the immune response against cancer cells. Currently, over 20 different commercial methods have been developed for the construction of BsAbs. Three BsAbs are currently clinically approved and marketed, and more than 85 clinical trials are in progress. In the present review, we discuss recent trends in the design, engineering, clinical applications, and clinical trials of BsAbs in solid tumors.
Collapse
Affiliation(s)
- Seyed Samad Hosseini
- Department of Biotechnology, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Faculty of Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negar Bidar
- Department of Biotechnology, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Jafar Mosafer
- Nanotechnology Research center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology, School of Paramedical Science, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mahmoud Hashemzaei
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02114, USA; Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA; Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| |
Collapse
|
9
|
A novel efficient bispecific antibody format, combining a conventional antigen-binding fragment with a single domain antibody, avoids potential heavy-light chain mis-pairing. J Immunol Methods 2020; 483:112811. [PMID: 32569598 DOI: 10.1016/j.jim.2020.112811] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/21/2020] [Accepted: 06/15/2020] [Indexed: 01/04/2023]
Abstract
Due to the technical innovations in generating bispecific antibodies (BsAbs) in recent years, BsAbs have become important reagents for diagnostic and therapeutic applications. However, the difficulty of producing a heterodimer consisting of two different arms with high yield and purity constituted a major limitation for their application in academic and clinical settings. Here, we describe a novel Fc-containing BsAb format (Fab × sdAb-Fc) composed of a conventional antigen-binding fragment (Fab), and a single domain antibody (sdAb), which avoids heavy-light chain mis-pairing during antibody assembly. In this study, the Fab x sdAb-Fc BsAbs were efficiently produced by three widely used heavy-heavy chain heterodimerization methods: Knobs-into-holes (KIH), Charge-pairs (CP) and controlled Fab-arm exchange (cFAE), respectively. The novel Fab x sdAb-Fc format provided a rapid and efficient strategy to generate BsAb with high purity and a unique possibility to further purify desired BsAbs from undesired antibodies based on molecular weight (MW). Compared to conventional BsAb formats, the advantages of Fab x sdAb-Fc format may thus provide a straightforward opportunity to apply bispecific antibody principles to research and development of novel targets and pathways in diseases such as cancer and autoimmunity.
Collapse
|
10
|
Dietrich S, Gross AW, Becker S, Hock B, Stadlmayr G, Rüker F, Wozniak-Knopp G. Constant domain-exchanged Fab enables specific light chain pairing in heterodimeric bispecific SEED-antibodies. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2019; 1868:140250. [PMID: 31295556 DOI: 10.1016/j.bbapap.2019.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/13/2019] [Accepted: 07/03/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Bispecific antibodies promise to broadly expand the clinical utility of monoclonal antibody technology. Several approaches for heterodimerization of heavy chains have been established to produce antibodies with two different Fab arms, but promiscuous pairing of heavy and light chains remains a challenge for their manufacturing. METHODS We have designed a solution in which the CH1 and CL domain pair in one of the Fab fragments is replaced with a CH3-domain pair and heterodimerized to facilitate correct modified Fab-chain pairing in bispecific heterodimeric antibodies based on a strand-exchange engineered domain (SEED) scaffold with specificity for epithelial growth factor receptor and either CD3 or CD16 (FcγRIII). RESULTS Bispecific antibodies retained binding to their target antigens and redirected primary T cells or NK cells to induce potent killing of target cells. All antibodies were expressed at a high yield in Expi293F cells, were detected as single sharp symmetrical peaks in size exclusion chromatography and retained high thermostability. Mass spectrometric analysis revealed specific heavy-to-light chain pairing for the bispecific SEED antibodies as well as for one-armed SEED antibodies co-expressed with two different competing light chains. CONCLUSION Incorporation of a constant domain-exchanged Fab fragment into a SEED antibody yields functional molecules with favorable biophysical properties. GENERAL SIGNIFICANCE Our results show that the novel engineered bispecific SEED antibody scaffold with an incorporated Fab fragment with CH3-exchanged constant domains is a promising tool for the generation of complete heterodimeric bispecific antibodies with correct light chain pairing.
Collapse
Affiliation(s)
- Sylvia Dietrich
- Christian Doppler Laboratory for Antibody Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Alec W Gross
- Protein Engineering and Antibody Technologies, EMD Serono Research and Development Institute, Inc., 45A Middlesex Turnpike, Billerica, MA 01821, USA
| | - Stefan Becker
- Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Björn Hock
- Protein Engineering and Antibody Technologies, Merck KGaA, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Gerhard Stadlmayr
- Christian Doppler Laboratory for Innovative Immunotherapeutics, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Florian Rüker
- Christian Doppler Laboratory for Innovative Immunotherapeutics, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Gordana Wozniak-Knopp
- Christian Doppler Laboratory for Innovative Immunotherapeutics, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|
11
|
Benschop RJ, Chow CK, Tian Y, Nelson J, Barmettler B, Atwell S, Clawson D, Chai Q, Jones B, Fitchett J, Torgerson S, Ji Y, Bina H, Hu N, Ghanem M, Manetta J, Wroblewski VJ, Lu J, Allan BW. Development of tibulizumab, a tetravalent bispecific antibody targeting BAFF and IL-17A for the treatment of autoimmune disease. MAbs 2019; 11:1175-1190. [PMID: 31181988 PMCID: PMC6748573 DOI: 10.1080/19420862.2019.1624463] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/26/2019] [Accepted: 05/22/2019] [Indexed: 12/23/2022] Open
Abstract
We describe a bispecific dual-antagonist antibody against human B cell activating factor (BAFF) and interleukin 17A (IL-17). An anti-IL-17 single-chain variable fragment (scFv) derived from ixekizumab (Taltz®) was fused via a glycine-rich linker to anti-BAFF tabalumab. The IgG-scFv bound both BAFF and IL-17 simultaneously with identical stoichiometry as the parental mAbs. Stability studies of the initial IgG-scFv revealed chemical degradation and aggregation not observed in either parental antibody. The anti-IL-17 scFv showed a high melting temperature (Tm) by differential scanning calorimetry (73.1°C), but also concentration-dependent, initially reversible, protein self-association. To engineer scFv stability, three parallel approaches were taken: labile complementary-determining region (CDR) residues were replaced by stable, affinity-neutral amino acids, CDR charge distribution was balanced, and a H44-L100 interface disulfide bond was introduced. The Tm of the disulfide-stabilized scFv was largely unperturbed, yet it remained monodispersed at high protein concentration. Fluorescent dye binding titrations indicated reduced solvent exposure of hydrophobic residues and decreased proteolytic susceptibility was observed, both indicative of enhanced conformational stability. Superimposition of the H44-L100 scFv (PDB id: 6NOU) and ixekizumab antigen-binding fragment (PDB id: 6NOV) crystal structures revealed nearly identical orientation of the frameworks and CDR loops. The stabilized bispecific molecule LY3090106 (tibulizumab) potently antagonized both BAFF and IL-17 in cell-based and in vivo mouse models. In cynomolgus monkey, it suppressed B cell development and survival and remained functionally intact in circulation, with a prolonged half-life. In summary, we engineered a potent bispecific antibody targeting two key cytokines involved in human autoimmunity amenable to clinical development.
Collapse
Affiliation(s)
- Robert J. Benschop
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company Corporate Center, Indianapolis, IN, USA
| | - Chi-Kin Chow
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company Corporate Center, Indianapolis, IN, USA
| | - Yu Tian
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company Corporate Center, Indianapolis, IN, USA
| | - James Nelson
- Biotechnology Discovery Research, Applied Molecular Evolution, Eli Lilly and Company, San Diego, CA, USA
| | - Barbra Barmettler
- Biotechnology Discovery Research, Applied Molecular Evolution, Eli Lilly and Company, San Diego, CA, USA
| | - Shane Atwell
- Biotechnology Discovery Research, Applied Molecular Evolution, Eli Lilly and Company, San Diego, CA, USA
| | - David Clawson
- Discovery Chemistry Research and Technologies, Eli Lilly and Company Corporate Center, Indianapolis, IN, USA
| | - Qing Chai
- Biotechnology Discovery Research, Applied Molecular Evolution, Eli Lilly and Company, San Diego, CA, USA
| | - Bryan Jones
- Biotechnology Discovery Research, Applied Molecular Evolution, Eli Lilly and Company, San Diego, CA, USA
| | - Jon Fitchett
- Biotechnology Discovery Research, Applied Molecular Evolution, Eli Lilly and Company, San Diego, CA, USA
| | - Stacy Torgerson
- Department of Drug Disposition Development/Commercialization; Lilly Research Laboratories, Eli Lilly and Company Corporate Center, Indianapolis, IN, USA
| | | | - Holly Bina
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company Corporate Center, Indianapolis, IN, USA
| | - Ningjie Hu
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company Corporate Center, Indianapolis, IN, USA
| | | | - Joseph Manetta
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company Corporate Center, Indianapolis, IN, USA
| | - Victor J. Wroblewski
- Department of Drug Disposition Development/Commercialization; Lilly Research Laboratories, Eli Lilly and Company Corporate Center, Indianapolis, IN, USA
| | - Jirong Lu
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company Corporate Center, Indianapolis, IN, USA
| | - Barrett W. Allan
- Biotechnology Discovery Research, Applied Molecular Evolution, Eli Lilly and Company, San Diego, CA, USA
| |
Collapse
|
12
|
Wu X, Demarest SJ. Building blocks for bispecific and trispecific antibodies. Methods 2018; 154:3-9. [PMID: 30172007 DOI: 10.1016/j.ymeth.2018.08.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/23/2018] [Accepted: 08/25/2018] [Indexed: 01/07/2023] Open
Abstract
Bispecific antibodies (BsAbs), which target two antigens or epitopes, incorporate the specificities and properties of two distinct monoclonal antibodies (mAbs) into a single molecule. As such, BsAbs can elicit synergistic activities and provide the capacity for enhanced therapeutic efficacy and/or safety compared to what can be achieved with conventional monospecific IgGs. There are many building block formats to generate BsAbs and Trispecific antibodies (TsAbs) based on combining the antigen recognition domains of monoclonal antibodies (mAbs). This review describes the many and varied antibody-based building blocks used to achieve multivalency and multispecificity. These diverse building blocks provide opportunities to tailor the design of BsAbs and TsAbs to match the desired applications.
Collapse
Affiliation(s)
- Xiufeng Wu
- Lilly Biotechnology Center, 10290 Campus Point Dr., San Diego, CA 92121, United States.
| | - Stephen J Demarest
- Lilly Biotechnology Center, 10290 Campus Point Dr., San Diego, CA 92121, United States
| |
Collapse
|
13
|
Regula JT, Imhof-Jung S, Mølhøj M, Benz J, Ehler A, Bujotzek A, Schaefer W, Klein C. Variable heavy-variable light domain and Fab-arm CrossMabs with charged residue exchanges to enforce correct light chain assembly. Protein Eng Des Sel 2018; 31:289-299. [PMID: 30169707 PMCID: PMC6277175 DOI: 10.1093/protein/gzy021] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/26/2018] [Accepted: 08/03/2018] [Indexed: 01/07/2023] Open
Abstract
Technologies for the production of bispecific antibodies need to overcome two major challenges. The first one is correct heavy chain assembly, which was solved by knobs-into-holes technology or charge interactions in the CH3 domains. The second challenge is correct light chain assembly. This can be solved by engineering the Fab-arm interfaces or applying the immunoglobulin domain crossover approach. There are three different crossovers possible, namely Fab-arm, constant domain and variable domain crossovers. The CrossMabCH1-CL exchange does not lead to the formation of unexpected side products, whereas the CrossMabFab and the CrossMabVH-VL formats result in the formation of typical side products. Thus, CrossMabCH1-CL was initially favored for therapeutic antibody development. Here, we report a novel improved CrossMab design principle making use of site-specific positional exchanges of charged amino acid pairs in the constant domain of these CrossMabs to enable the correct light chain assembly in the CrossMabVH-VL and improvements for the CrossMabFab design.
Collapse
Affiliation(s)
- Joerg Thomas Regula
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Sabine Imhof-Jung
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Michael Mølhøj
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Joerg Benz
- Roche Pharmaceutical Research and Early Development, Chemical Biology, Roche Innovation Center Basel, Basel, Switzerland
| | - Andreas Ehler
- Roche Pharmaceutical Research and Early Development, Chemical Biology, Roche Innovation Center Basel, Basel, Switzerland
| | - Alexander Bujotzek
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Wolfgang Schaefer
- Roche Pharmaceutical Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Christian Klein
- Roche Pharmaceutical Research and Early Development, Discovery Oncology, Roche Innovation Center Zurich, Schlieren, Switzerland
| |
Collapse
|
14
|
Leem J, Georges G, Shi J, Deane CM. Antibody side chain conformations are position-dependent. Proteins 2018; 86:383-392. [PMID: 29318667 DOI: 10.1002/prot.25453] [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: 09/26/2017] [Revised: 12/15/2017] [Accepted: 01/05/2018] [Indexed: 11/11/2022]
Abstract
Side chain prediction is an integral component of computational antibody design and structure prediction. Current antibody modelling tools use backbone-dependent rotamer libraries with conformations taken from general proteins. Here we present our antibody-specific rotamer library, where rotamers are binned according to their immunogenetics (IMGT) position, rather than their local backbone geometry. We find that for some amino acid types at certain positions, only a restricted number of side chain conformations are ever observed. Using this information, we are able to reduce the breadth of the rotamer sampling space. Based on our rotamer library, we built a side chain predictor, position-dependent antibody rotamer swapper (PEARS). On a blind test set of 95 antibody model structures, PEARS had the highest average χ1 and χ1+2 accuracy (78.7% and 64.8%) compared to three leading backbone-dependent side chain predictors. Our use of IMGT position, rather than backbone ϕ/ψ, meant that PEARS was more robust to errors in the backbone of the model structure. PEARS also achieved the lowest number of side chain-side chain clashes. PEARS is freely available as a web application at http://opig.stats.ox.ac.uk/webapps/pears.
Collapse
Affiliation(s)
- Jinwoo Leem
- Department of Statistics, University of Oxford, 24-29 St Giles, Oxford, OX1 3LB, United Kingdom
| | - Guy Georges
- Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Nonnenwald 2, Penzberg, 82377, Germany
| | - Jiye Shi
- Chemistry Department, UCB, 208 Bath Road, Slough, SL1 3WE, United Kingdom
| | - Charlotte M Deane
- Department of Statistics, University of Oxford, 24-29 St Giles, Oxford, OX1 3LB, United Kingdom
| |
Collapse
|
15
|
Brinkmann U, Kontermann RE. The making of bispecific antibodies. MAbs 2017; 9:182-212. [PMID: 28071970 PMCID: PMC5297537 DOI: 10.1080/19420862.2016.1268307] [Citation(s) in RCA: 605] [Impact Index Per Article: 86.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/18/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022] Open
Abstract
During the past two decades we have seen a phenomenal evolution of bispecific antibodies for therapeutic applications. The 'zoo' of bispecific antibodies is populated by many different species, comprising around 100 different formats, including small molecules composed solely of the antigen-binding sites of two antibodies, molecules with an IgG structure, and large complex molecules composed of different antigen-binding moieties often combined with dimerization modules. The application of sophisticated molecular design and genetic engineering has solved many of the technical problems associated with the formation of bispecific antibodies such as stability, solubility and other parameters that confer drug properties. These parameters may be summarized under the term 'developability'. In addition, different 'target product profiles', i.e., desired features of the bispecific antibody to be generated, mandates the need for access to a diverse panel of formats. These may vary in size, arrangement, valencies, flexibility and geometry of their binding modules, as well as in their distribution and pharmacokinetic properties. There is not 'one best format' for generating bispecific antibodies, and no single format is suitable for all, or even most of, the desired applications. Instead, the bispecific formats collectively serve as a valuable source of diversity that can be applied to the development of therapeutics for various indications. Here, a comprehensive overview of the different bispecific antibody formats is provided.
Collapse
Affiliation(s)
- Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Im Nonnenwald, Penzberg, Germany
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Nobelstraße, Stuttgart, Germany
| |
Collapse
|
16
|
Bönisch M, Sellmann C, Maresch D, Halbig C, Becker S, Toleikis L, Hock B, Rüker F. Novel CH1:CL interfaces that enhance correct light chain pairing in heterodimeric bispecific antibodies. Protein Eng Des Sel 2017; 30:685-696. [PMID: 28981885 PMCID: PMC5914326 DOI: 10.1093/protein/gzx044] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/14/2017] [Accepted: 08/02/2017] [Indexed: 11/14/2022] Open
Abstract
Targeting two unique antigens with a single bispecific antibody is an attractive approach with potential broad therapeutic applicability. However, the production of heterodimeric bispecific antibodies (bsAbs) presents a challenge, requiring the co-expression and accurate pairing of two distinct heavy and light chain units. Several undesirable by-products can be formed in the production process, including heavy chain homodimers and non-cognate light chain pairings. Although additional downstream purification methods exist, they are often time consuming and restrict practical large-scale production. In this study, we identify and validate novel Fab interface mutations that increase cognate light chain pairing efficiencies within heterodimeric bsAbs. Importantly, the variable domains remain unaltered as interface mutations were restricted to the CH1 and CL domains. We performed several biochemical assays to demonstrate that the novel engineered interfaces do not adversely impact bispecific antibody expression, stability, affinity and biological function. The designs reported here can easily be applied in a generic manner to use existing antibodies as building blocks for bsAbs which will help to accelerate the identification and production of next generation bispecific antibody therapeutics.
Collapse
Affiliation(s)
- Maximilian Bönisch
- Christian Doppler Laboratory for Antibody Engineering at Department of Chemistry and Department of Biotechnology, BOKU—University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, A-1190 Vienna, Austria
| | - 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
| | - Daniel Maresch
- Department of Chemistry, BOKU—University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, A-1190 Vienna, Austria
| | - Claudia Halbig
- Christian Doppler Laboratory for Antibody Engineering at Department of Chemistry and Department of Biotechnology, BOKU—University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, A-1190 Vienna, Austria
| | - Stefan Becker
- 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
| | - Florian Rüker
- Christian Doppler Laboratory for Antibody Engineering at Department of Chemistry and Department of Biotechnology, BOKU—University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, A-1190 Vienna, Austria
| |
Collapse
|
17
|
Dillon M, Yin Y, Zhou J, McCarty L, Ellerman D, Slaga D, Junttila TT, Han G, Sandoval W, Ovacik MA, Lin K, Hu Z, Shen A, Corn JE, Spiess C, Carter PJ. Efficient production of bispecific IgG of different isotypes and species of origin in single mammalian cells. MAbs 2016; 9:213-230. [PMID: 27929752 PMCID: PMC5297516 DOI: 10.1080/19420862.2016.1267089] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Bispecific IgG production in single host cells has been a much sought-after goal to support the clinical development of these complex molecules. Current routes to single cell production of bispecific IgG include engineering heavy chains for heterodimerization and redesign of Fab arms for selective pairing of cognate heavy and light chains. Here, we describe novel designs to facilitate selective Fab arm assembly in conjunction with previously described knobs-into-holes mutations for preferential heavy chain heterodimerization. The top Fab designs for selective pairing, namely variants v10 and v11, support near quantitative assembly of bispecific IgG in single cells for multiple different antibody pairs as judged by high-resolution mass spectrometry. Single-cell and in vitro-assembled bispecific IgG have comparable physical, in vitro biological and in vivo pharmacokinetics properties. Efficient single-cell production of bispecific IgG was demonstrated for human IgG1, IgG2 and IgG4 thereby allowing the heavy chain isotype to be tailored for specific therapeutic applications. Additionally, a reverse chimeric bispecific IgG2a with humanized variable domains and mouse constant domains was generated for preclinical proof-of-concept studies in mice. Efficient production of a bispecific IgG in stably transfected mammalian (CHO) cells was shown. Individual clones with stable titer and bispecific IgG composition for >120 days were readily identified. Such long-term cell line stability is needed for commercial manufacture of bispecific IgG. The single-cell bispecific IgG designs developed here may be broadly applicable to biotechnology research, including screening bispecific IgG panels, and to support clinical development.
Collapse
Affiliation(s)
- Michael Dillon
- a Department of Antibody Engineering , Genentech, Inc. , South San Francisco , CA , USA
| | - Yiyuan Yin
- a Department of Antibody Engineering , Genentech, Inc. , South San Francisco , CA , USA
| | - Jianhui Zhou
- a Department of Antibody Engineering , Genentech, Inc. , South San Francisco , CA , USA
| | - Luke McCarty
- b Department of Protein Chemistry , Genentech, Inc. , South San Francisco , CA , USA
| | - Diego Ellerman
- b Department of Protein Chemistry , Genentech, Inc. , South San Francisco , CA , USA
| | - Dionysos Slaga
- c Department of Translational Oncology , Genentech, Inc. , South San Francisco , CA , USA
| | - Teemu T Junttila
- c Department of Translational Oncology , Genentech, Inc. , South San Francisco , CA , USA
| | - Guanghui Han
- d Department of Microchemistry, Proteomics and Lipidomics , Genentech, Inc. , South San Francisco , CA , USA
| | - Wendy Sandoval
- d Department of Microchemistry, Proteomics and Lipidomics , Genentech, Inc. , South San Francisco , CA , USA
| | - Meric A Ovacik
- e Department of Preclinical and Translational Pharmacokinetics , Genentech, Inc. , South San Francisco , CA , USA
| | - Kedan Lin
- e Department of Preclinical and Translational Pharmacokinetics , Genentech, Inc. , South San Francisco , CA , USA
| | - Zhilan Hu
- f Department of Early Stage Cell Culture , Genentech, Inc. , South San Francisco , CA , USA
| | - Amy Shen
- f Department of Early Stage Cell Culture , Genentech, Inc. , South San Francisco , CA , USA
| | - Jacob E Corn
- g Department of Early Discovery Biochemistry, Genentech, Inc. , South San Francisco , CA , USA
| | - Christoph Spiess
- a Department of Antibody Engineering , Genentech, Inc. , South San Francisco , CA , USA
| | - Paul J Carter
- a Department of Antibody Engineering , Genentech, Inc. , South San Francisco , CA , USA
| |
Collapse
|
18
|
Crystal structures of mono- and bi-specific diabodies and reduction of their structural flexibility by introduction of disulfide bridges at the Fv interface. Sci Rep 2016; 6:34515. [PMID: 27682821 PMCID: PMC5041106 DOI: 10.1038/srep34515] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/14/2016] [Indexed: 12/20/2022] Open
Abstract
Building a sophisticated protein nano-assembly requires a method for linking protein components in a predictable and stable structure. Diabodies are engineered antibody fragments that are composed of two Fv domains connected by short peptide linkers. They are attractive candidates for mediators in assembling protein nano-structures because they can simultaneously bind to two different proteins and are rigid enough to be crystallized. However, comparison of previous crystal structures demonstrates that there is substantial structural diversity in the Fv interface region of diabodies and, therefore, reliable prediction of its structure is not trivial. Here, we present the crystal structures of ten mono- and bi-specific diabodies. We found that changing an arginine residue in the Fv interface to threonine greatly reduced the structural diversity of diabodies. We also found that one of the bispecific diabodies underwent an unexpected process of chain swapping yielding a non-functional monospecific diabody. In order to further reduce structural flexibility and prevent chain shuffling, we introduced disulfide bridges in the Fv interface regions. The disulfide-bridged diabodies have rigid and predictable structures and may have applications in crystallizing proteins, analyzing cryo-electron microscopic images and building protein nano-assemblies.
Collapse
|
19
|
Abstract
Cancer immunotherapy has recently generated much excitement after the continuing success of the immunomodulating anti-CTLA-4 and anti-PD-1 antibodies against various types of cancers. Aside from these immunomodulating antibodies, bispecific antibodies, chimeric antigen receptor T cells, and other technologies are being actively studied. Among the various approaches to cancer immunotherapy, 2 bispecific antibodies are currently approved for patient care. Many more bispecific antibodies are now in various phases of clinical development and will become the next generation of antibody-based therapies. Further understanding of immunology and advances in protein engineering will help to generate a greater variety of bispecific antibodies to fight cancer. Here, we focus on bispecific antibodies that recruit immune cells to engage and kill tumor cells.
Collapse
Affiliation(s)
- Siqi Chen
- a School of Pharmaceutical Sciences, Sun Yat-Sen University , Guangzhou , China.,b Center for Cellular & Structural Biology, Sun Yat-Sen University , Guangzhou , China
| | - Jing Li
- a School of Pharmaceutical Sciences, Sun Yat-Sen University , Guangzhou , China.,b Center for Cellular & Structural Biology, Sun Yat-Sen University , Guangzhou , China
| | - Qing Li
- a School of Pharmaceutical Sciences, Sun Yat-Sen University , Guangzhou , China.,b Center for Cellular & Structural Biology, Sun Yat-Sen University , Guangzhou , China
| | - Zhong Wang
- a School of Pharmaceutical Sciences, Sun Yat-Sen University , Guangzhou , China.,b Center for Cellular & Structural Biology, Sun Yat-Sen University , Guangzhou , China
| |
Collapse
|
20
|
Electrostatic engineering of the interface between heavy and light chains promotes antibody Fab fragment production. Cytotechnology 2016; 69:469-475. [PMID: 26856589 DOI: 10.1007/s10616-016-9955-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 02/01/2016] [Indexed: 12/11/2022] Open
Abstract
Monoclonal antibodies and antibody fragments are used for diverse diagnostic and therapeutic applications. We have investigated the secretory production of Fab fragments from insect cells cotransfected with plasmid vectors carrying heavy- and light-chain genes. In the present study, to promote the formation of the disulfide bond between the heavy and light chains, some positively charged amino acid residues were introduced near the cysteine residue for the disulfide bond at the C-terminus of CL, while some negatively charged amino acid residues were added near the cysteine residue for the disulfide bond at the C-terminus of CH1. This electrostatic steering led to an increase in Fab secretions from insect cells.
Collapse
|
21
|
Schaefer W, Völger HR, Lorenz S, Imhof-Jung S, Regula JT, Klein C, Mølhøj M. Heavy and light chain pairing of bivalent quadroma and knobs-into-holes antibodies analyzed by UHR-ESI-QTOF mass spectrometry. MAbs 2015; 8:49-55. [PMID: 26496506 DOI: 10.1080/19420862.2015.1111498] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The quadroma antibody represents the first attempt to produce a bispecific heterodimeric IgG antibody by somatic fusion of 2 hybridoma cells each expressing monoclonal antibodies with distinctive specificities. However, because of random heavy and light chain pairing, the desired functional bispecific antibody represents only a small fraction of the protein produced. Subsequently, the knobs-into-holes (KiH) approach was developed to enforce correct heavy chain heterodimerization. Assuming equimolar expression of 4 unmodified chains comprising 2 heavy and 2 light chains, the statistical distribution of all paired combinations can be calculated. With equimolar expression as the goal, we transfected HEK cells with 1:1:1:1 plasmid ratios and analyzed the protein A affinity-purified antibodies from the quadroma and KiH approaches qualitatively and quantitatively with regard to the estimated relative amounts of the products using electrospray quadrupole time-of-flight mass spectrometry. Our results show that all expected species are formed, and that, within the methodological limits, the species distribution in the mixtures corresponds approximately to the statistical distribution.
Collapse
Affiliation(s)
- Wolfgang Schaefer
- a Roche Pharma Research and Early Development; Large Molecule Research, Roche Innovation Center Penzberg ; Penzberg , Germany
| | - Hans R Völger
- a Roche Pharma Research and Early Development; Large Molecule Research, Roche Innovation Center Penzberg ; Penzberg , Germany
| | - Stefan Lorenz
- a Roche Pharma Research and Early Development; Large Molecule Research, Roche Innovation Center Penzberg ; Penzberg , Germany
| | - Sabine Imhof-Jung
- a Roche Pharma Research and Early Development; Large Molecule Research, Roche Innovation Center Penzberg ; Penzberg , Germany
| | - Jörg T Regula
- a Roche Pharma Research and Early Development; Large Molecule Research, Roche Innovation Center Penzberg ; Penzberg , Germany
| | - Christian Klein
- b Oncology Discovery & Translational Area, Roche Innovation Center Zurich ; Schlieren , Switzerland
| | - Michael Mølhøj
- a Roche Pharma Research and Early Development; Large Molecule Research, Roche Innovation Center Penzberg ; Penzberg , Germany
| |
Collapse
|
22
|
Spiess C, Zhai Q, Carter PJ. Alternative molecular formats and therapeutic applications for bispecific antibodies. Mol Immunol 2015; 67:95-106. [DOI: 10.1016/j.molimm.2015.01.003] [Citation(s) in RCA: 417] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 12/30/2014] [Accepted: 01/02/2015] [Indexed: 12/21/2022]
|
23
|
Liu Z, Leng EC, Gunasekaran K, Pentony M, Shen M, Howard M, Stoops J, Manchulenko K, Razinkov V, Liu H, Fanslow W, Hu Z, Sun N, Hasegawa H, Clark R, Foltz IN, Yan W. A novel antibody engineering strategy for making monovalent bispecific heterodimeric IgG antibodies by electrostatic steering mechanism. J Biol Chem 2015; 290:7535-62. [PMID: 25583986 DOI: 10.1074/jbc.m114.620260] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Producing pure and well behaved bispecific antibodies (bsAbs) on a large scale for preclinical and clinical testing is a challenging task. Here, we describe a new strategy for making monovalent bispecific heterodimeric IgG antibodies in mammalian cells. We applied an electrostatic steering mechanism to engineer antibody light chain-heavy chain (LC-HC) interface residues in such a way that each LC strongly favors its cognate HC when two different HCs and two different LCs are co-expressed in the same cell to assemble a functional bispecific antibody. We produced heterodimeric IgGs from transiently and stably transfected mammalian cells. The engineered heterodimeric IgG molecules maintain the overall IgG structure with correct LC-HC pairings, bind to two different antigens with comparable affinity when compared with their parental antibodies, and retain the functionality of parental antibodies in biological assays. In addition, the bispecific heterodimeric IgG derived from anti-HER2 and anti-EGF receptor (EGFR) antibody was shown to induce a higher level of receptor internalization than the combination of two parental antibodies. Mouse xenograft BxPC-3, Panc-1, and Calu-3 human tumor models showed that the heterodimeric IgGs strongly inhibited tumor growth. The described approach can be used to generate tools from two pre-existent antibodies and explore the potential of bispecific antibodies. The asymmetrically engineered Fc variants for antibody-dependent cellular cytotoxicity enhancement could be embedded in monovalent bispecific heterodimeric IgG to make best-in-class therapeutic antibodies.
Collapse
Affiliation(s)
- Zhi Liu
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119,
| | - Esther C Leng
- From the Departments of Therapeutic Discovery and Amgen Inc., Burnaby, British Columbia V5A 1V7, Canada
| | - Kannan Gunasekaran
- From the Departments of Therapeutic Discovery and Amgen Inc., Thousand Oaks, California 91320, and
| | - Martin Pentony
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Min Shen
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Monique Howard
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Janelle Stoops
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Kathy Manchulenko
- From the Departments of Therapeutic Discovery and Amgen Inc., Burnaby, British Columbia V5A 1V7, Canada
| | - Vladimir Razinkov
- Amgen Inc., Seattle, Washington 98119, Process and Product Development
| | - Hua Liu
- Amgen Inc., Seattle, Washington 98119, Therapeutic Innovation Unit
| | - William Fanslow
- Amgen Inc., Seattle, Washington 98119, Therapeutic Innovation Unit
| | - Zhonghua Hu
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Nancy Sun
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Haruki Hasegawa
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Rutilio Clark
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Ian N Foltz
- From the Departments of Therapeutic Discovery and Amgen Inc., Burnaby, British Columbia V5A 1V7, Canada
| | - Wei Yan
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119,
| |
Collapse
|
24
|
Generation of bispecific IgG antibodies by structure-based design of an orthogonal Fab interface. Nat Biotechnol 2014; 32:191-8. [PMID: 24463572 DOI: 10.1038/nbt.2797] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 12/10/2013] [Indexed: 01/22/2023]
Abstract
Robust generation of IgG bispecific antibodies has been a long-standing challenge. Existing methods require extensive engineering of each individual antibody, discovery of common light chains, or complex and laborious biochemical processing. Here we combine computational and rational design approaches with experimental structural validation to generate antibody heavy and light chains with orthogonal Fab interfaces. Parental monoclonal antibodies incorporating these interfaces, when simultaneously co-expressed, assemble into bispecific IgG with improved heavy chain-light chain pairing. Bispecific IgGs generated with this approach exhibit pharmacokinetic and other desirable properties of native IgG, but bind target antigens monovalently. As such, these bispecific reagents may be useful in many biotechnological applications.
Collapse
|
25
|
|
26
|
Klein C, Sustmann C, Thomas M, Stubenrauch K, Croasdale R, Schanzer J, Brinkmann U, Kettenberger H, Regula JT, Schaefer W. Progress in overcoming the chain association issue in bispecific heterodimeric IgG antibodies. MAbs 2012; 4:653-63. [PMID: 22925968 DOI: 10.4161/mabs.21379] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The development of bispecific antibodies has attracted substantial interest, and many different formats have been described. Those specifically containing an Fc part are mostly tetravalent, such as stabilized IgG-scFv fusions or dual-variable domain (DVD) IgGs. However, although they exhibit IgG-like properties and technical developability, these formats differ in size and geometry from classical IgG antibodies. Thus, considerable efforts focus on bispecific heterodimeric IgG antibodies that more closely mimic natural IgG molecules. The inherent chain association problem encountered when producing bispecific heterodimeric IgG antibodies can be overcome by several methods. While technologies like knobs-into-holes (KiH) combined with a common light chain or the CrossMab technology enforce the correct chain association, other approaches, e.g., the dual-acting Fab (DAF) IgGs, do not rely on a heterodimeric Fc part. This review discusses the state of the art in bispecific heterodimeric IgG antibodies, with an emphasis on recent progress.
Collapse
Affiliation(s)
- Christian Klein
- Discovery Oncology, Roche Pharma Research and Early Development pRED, Roche Glycart AG, Schlieren, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Bax HJ, Keeble AH, Gould HJ. Cytokinergic IgE Action in Mast Cell Activation. Front Immunol 2012; 3:229. [PMID: 22888332 PMCID: PMC3412263 DOI: 10.3389/fimmu.2012.00229] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 07/14/2012] [Indexed: 12/20/2022] Open
Abstract
Some 10 years ago it emerged that at sufficiently high concentrations certain monoclonal mouse IgEs exert previously unsuspected effects on mast cells. Thus they can both promote survival and induce activation of mast cells without the requirement for antigens. This was a wake up call that appears to have been missed (or dismissed) by the majority of immunologists. The structural attributes responsible for the potency of the so-called “highly cytokinergic” or HC IgEs have not yet been determined, but the events that ensue when such IgEs bind to the high-affinity receptor, FcεRI, on mast cells have been thoroughly studied, and are strikingly similar to those engendered by antigens when they form cross-linked complexes with the receptors. We review the evidence for the cytokinergic activity of IgE, and the structural features and known properties of immunoglobulins, and of IgE in particular, most likely to be implicated in the phenomenon. We suggest that IgEs with cytokinergic activity may be generated by local germinal center reactions in the target organs of allergy. We consider also the important implications that the existence of cytokinergic IgE may have for a fuller understanding of adaptive immunity and of the action of IgE in asthma and other diseases.
Collapse
Affiliation(s)
- Heather J Bax
- Randall Division of Cell and Molecular Biophysics, King's College London London, UK
| | | | | |
Collapse
|
28
|
Vendel MC, Favis M, Snyder WB, Huang F, Capili AD, Dong J, Glaser SM, Miller BR, Demarest SJ. Secretion from bacterial versus mammalian cells yields a recombinant scFv with variable folding properties. Arch Biochem Biophys 2012; 526:188-93. [PMID: 22230329 DOI: 10.1016/j.abb.2011.12.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 12/19/2011] [Accepted: 12/21/2011] [Indexed: 11/24/2022]
Abstract
Escherichia coli (E. coli) is the most commonly used organism for expressing antibody fragments such as single chain antibody Fvs (scFvs). Previously, we have utilized E. coli to express well-folded scFvs for characterization and engineering purposes with the goal of using these engineered proteins as building blocks for generating IgG-like bispecific antibodies (BsAbs). In the study, described here, we observed a significant difference in the secondary structure of an scFv produced in E. coli and the same scFv expressed and secreted from chinese hamster ovary (CHO) cells as part of a BsAb. We devised a proteolytic procedure to separate the CHO-derived scFv from its antibody-fusion partner and compared its properties with those of the E. coli-derived scFv. In comparison to the CHO-derived scFv, the E. coli-derived scFv was found trapped in a misfolded, but monomeric state that was stable for months at 4 °C. The misfolded state bound antigen in a heterogeneous fashion that included non-specific binding, which made functional characterization challenging. This odd incidence of obtaining a misfolded scFv from bacteria suggests careful characterization of the folded properties of bacterially expressed scFvs is warranted if anomalous issues with antigen-binding or non-specificity occur during an engineering campaign. Additionally, our proteolytic methodology for obtaining significant levels of intact scFvs from highly expressed IgG-like antibody proteins serves as a robust method for producing scFvs in CHO without the use of designed cleavage motifs.
Collapse
|