1
|
Nguyen JB, Liu S, Howie DA, Oberholtzer ZR, Ong ET, Rao R, Prinston JE, Dikiy I, Wu J, Wu Z, Zhao Y, Li M, Molden R, Molina G, Provoncha K, Sandu C, Qiu H, Li N, Matousek W, Rosconi MP, Pyles EA. Comparison of enriched charge variants from different anti-CD3 bispecific antibodies reveals differential susceptibility of each bispecific arm to post-translational modification. Protein Sci 2025; 34:e70079. [PMID: 40116211 PMCID: PMC11926628 DOI: 10.1002/pro.70079] [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: 11/18/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 03/23/2025]
Abstract
Charge heterogeneity is an important quality attribute of therapeutic antibodies, and a detailed understanding of charge heterogeneity arising from post-translational modifications (PTMs) is required by regulatory agencies during drug development. Among antibody therapeutics, the bispecific antibody with two distinct Fab domains targeting distinct antigens provides additional complexity to the charge profile. In this study, charge variant species were enriched from three bispecific antibodies (bsAbs) each containing one anti-CD3 binding arm designed with differential affinity to CD3. The charge heterogeneity corresponding to each anti-CD3 arm within each enriched fraction was evaluated using a domain-specific, digestion-assisted imaged capillary isoelectric focusing (icIEF) method known as DiCE. Through fractionation, we observed that the anti-CD3 arm of each bispecific antibody exhibited different distributions of acidic variants, even when the anti-CD3 arms were identical based on primary sequence. Reduced peptide mapping was performed on specific fractions to identify unique site-specific PTMs that were uncovered or enriched through fractionation. In each case, the bispecific arm that was most susceptible to PTMs exhibited a more basic isoelectric point. Conformational stability analysis of each bispecific antibody using differential scanning calorimetry suggested that the more basic Fab arm tended to be correlated with a lower melting temperature, although it is unclear the extent to which PTMs on the basic arm may contribute to reduced conformational stability. Overall, these results provide additional evidence that each of the two arms of a bispecific antibody may exhibit differential susceptibility to post-translational modification and that this susceptibility is likely correlated with subtle differences in overall bispecific antibody structure, which is influenced by electrostatic properties inherent to the primary sequence. Future studies to obtain high-resolution structures of full-length bispecific antibodies by crystallography or cryo-electron microscopy may help to elucidate the driving force for susceptibility to PTMs in bispecific antibodies.
Collapse
Affiliation(s)
| | - Sophia Liu
- Protein BiochemistryRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | - Dylan A. Howie
- Protein BiochemistryRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | | | - Eric T. Ong
- Protein BiochemistryRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | - Ramya Rao
- Protein BiochemistryRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | | | - Igor Dikiy
- Protein BiochemistryRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | - Jikang Wu
- Analytical ChemistryRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | - Zhijie Wu
- Analytical ChemistryRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | - Yimeng Zhao
- Analytical ChemistryRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | - Meinuo Li
- Analytical ChemistryRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | - Rosalynn Molden
- Analytical ChemistryRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | - Guido Molina
- Therapeutic ProteinsRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | | | - Cristinel Sandu
- Therapeutic ProteinsRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | - Haibo Qiu
- Analytical ChemistryRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | - Ning Li
- Analytical ChemistryRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | - William Matousek
- Protein BiochemistryRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | | | - Erica A. Pyles
- Protein BiochemistryRegeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| |
Collapse
|
2
|
Gonzàlez Gutierrez C, Aimard A, Biarnes-Pélicot M, Kerfelec B, Puech PH, Robert P, Piazza F, Chames P, Limozin L. Decoupling Individual Host Response and Immune Cell Engager Cytotoxic Potency. ACS NANO 2025; 19:2089-2098. [PMID: 39791371 DOI: 10.1021/acsnano.4c08541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Immune cell engagers are molecular agents, usually antibody-based constructs, engineered to recruit immune cells against cancer cells and kill them. They are versatile and powerful tools for cancer immunotherapy. Despite the multiplication of engagers tested and accepted in the clinic, how molecular and cellular parameters influence their actions is poorly understood. In particular, disentangling the respective roles of host immune cells and engager biophysical characteristics is needed to improve their design and efficiency. Focusing here on harnessing antibody-dependent Natural Killer cell cytotoxicity, we measure the efficiency of 6 original bispecific antibodies (bsAb), associating an anti-HER2 nanobody and an anti-CD16 nanobody. In vitro cytotoxicity data using primary human NK cells on different target cell lines exposing different antigen densities were collected, exhibiting a wide range of bsAb dose response. In order to rationalize our observations, we introduce a simple multiscale model, postulating that the density of bsAb bridging the two cells is the main parameter triggering the cytotoxic response. We introduce two microscopic parameters: the surface cooperativity describing bsAb affinity at the bridging step and the threshold of bridge density determining the donor-dependent response. Both parameters permit ranking Abs and donors and predicting bsAb potency as a function of antibodies bulk affinities and receptor surface densities on cells. Our approach thus provides a general way to decouple donor response from immune engager characteristics, rationalizing the landscape of molecule design.
Collapse
Affiliation(s)
| | - Adrien Aimard
- Aix-Marseille Univ., CNRS, INSERM, Institut Paoli Calmettes, CRCM, 13009 Marseille, France
| | | | - Brigitte Kerfelec
- Aix-Marseille Univ., CNRS, INSERM, Institut Paoli Calmettes, CRCM, 13009 Marseille, France
| | - Pierre-Henri Puech
- Aix-Marseille Univ., CNRS, INSERM, LAI, Centuri Living Systems, 13009 Marseille, France
| | - Philippe Robert
- Aix-Marseille Univ., CNRS, INSERM, LAI, Centuri Living Systems, 13009 Marseille, France
- Assistance Publique Hôpitaux de Marseille, 13005 Marseille, France
| | - Francesco Piazza
- CNRS, Univ. Orleans, CBM, 45000 Orleans, France
- Dipartimento di Fisica e Astronomia, Università di Firenze and INFN sezione di Firenze, 50019 Sesto Fiorentino, Italy
| | - Patrick Chames
- Aix-Marseille Univ., CNRS, INSERM, Institut Paoli Calmettes, CRCM, 13009 Marseille, France
| | - Laurent Limozin
- Aix-Marseille Univ., CNRS, INSERM, LAI, Centuri Living Systems, 13009 Marseille, France
| |
Collapse
|
3
|
Saito S, Nakayama M, Yamazaki K, Miyamoto Y, Hiraishi K, Tomioka D, Takagi‐Maeda S, Usami K, Takahashi N, Nara S, Imai E. Engineering and physicochemical characterization of a novel, stable, symmetric bispecific antibody with dual target-binding using a common light chain. Protein Sci 2024; 33:e5121. [PMID: 39276019 PMCID: PMC11401053 DOI: 10.1002/pro.5121] [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: 11/28/2023] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 09/16/2024]
Abstract
Bispecific antibodies (BsAbs) have emerged as a major class of antibody therapeutics owing to their substantial potential in disease treatment. While several BsAbs have been successfully approved in recent years, ongoing development efforts continue to focus on optimizing various BsAbs tailored to particular antigens and action mechanisms, aiming to achieve favorable physicochemical properties. BsAbs generally encounter challenges due to their unfavorable physicochemical characteristics and poor manufacturing efficiencies, highlighting the need for optimization to achieve reliable productivity and developability. Herein, we describe the development of a novel symmetric BsAb, REGULGENT™ (N-term/C-term), comprising two Fab domains, using a common light chain. The heavy chain fragment encoded two antigen-binding determinants in one chain. The design and production of REGULGENT™ (N-term/C-term) are simple owing to the use of the same light chain, which does not induce heavy and light chain mispairing, frequently observed with the asymmetric BsAb format. REGULGENT™ (N-term/C-term) exhibited high expression and low aggregation characteristics during cell culture and stress treatment under low pH conditions. Differential scanning calorimetric data indicated that REGULGENT™ molecules had high conformational stability, similar to that of stabilized monoclonal antibodies. Surface plasmon resonance data showed that REGULGENT™ (N-term/C-term) could bind to two antigens simultaneously and exhibited a high affinity for two antigens. In summary, the symmetric BsAb format of REGULGENT™ confers its desirable IgG-like physicochemical properties, thus making it an excellent candidate for commercial development. The findings demonstrate a novel BsAb with substantial development potential for clinical applications.
Collapse
Affiliation(s)
- Seiji Saito
- Molecular Analysis Center, R&D DivisionKyowa Kirin Co., Ltd.TokyoJapan
| | - Makoto Nakayama
- Research Core Function Laboratories, R&D DivisionKyowa Kirin Co., Ltd.TokyoJapan
| | - Kaori Yamazaki
- Molecular Analysis Center, R&D DivisionKyowa Kirin Co., Ltd.TokyoJapan
| | - Yuya Miyamoto
- Molecular Analysis Center, R&D DivisionKyowa Kirin Co., Ltd.TokyoJapan
| | - Keiko Hiraishi
- Molecular Analysis Center, R&D DivisionKyowa Kirin Co., Ltd.TokyoJapan
| | - Daisuke Tomioka
- Molecular Analysis Center, R&D DivisionKyowa Kirin Co., Ltd.TokyoJapan
| | | | - Katsuaki Usami
- Modality Research Laboratories, R&D DivisionKyowa Kirin Co., Ltd.TokyoJapan
| | | | - Shinji Nara
- Molecular Analysis Center, R&D DivisionKyowa Kirin Co., Ltd.TokyoJapan
| | - Eiichiro Imai
- Molecular Analysis Center, R&D DivisionKyowa Kirin Co., Ltd.TokyoJapan
| |
Collapse
|
4
|
Li Y, Zhao W, Shen Y, Xu Y, Chen S, Pan L. T Cell Receptor-Directed Bispecific T Cell Engager Targeting MHC-Linked NY-ESO-1 for Tumor Immunotherapy. Biomedicines 2024; 12:776. [PMID: 38672132 PMCID: PMC11048172 DOI: 10.3390/biomedicines12040776] [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: 01/27/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Antibody-based bispecific T cell engagers (TCEs) that redirect T cells to kill tumor cells have shown a promising therapeutic effect on hematologic malignancies. However, tumor-specific targeting is still a challenge for TCEs, impeding the development of TCEs for solid tumor therapy. The major histocompatibility complex (MHC) presents almost all intracellular peptides (including tumor-specific peptides) on the cell surface to be scanned by the TCR on T cells. With the premise of choosing optimal peptides, the final complex peptide-MHC could be the tumor-specific target for TCEs. Here, a novel TCR-directed format of a TCE targeting peptide-MHC was designed named IgG-T-TCE, which was modified from the IgG backbone and prepared in a mammalian cell expression system. The recombinant IgG-T-TCE-NY targeting NY-ESO-1157-165/HLA-A*02:01 could be generated in HEK293 cells with a glycosylated TCR and showed potency in T cell activation and redirecting T cells to specifically kill target tumor cells. We also found that the in vitro activity of IgG-T-TCE-NY could be leveraged by various anti-CD3 antibodies and Fc silencing. The IgG-T-TCE-NY efficiently inhibited tumor growth in a tumor-PBMC co-engrafted mouse model without any obvious toxicities.
Collapse
Affiliation(s)
- Yiming Li
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (Y.L.); (W.Z.); (Y.S.); (Y.X.)
| | - Wenbin Zhao
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (Y.L.); (W.Z.); (Y.S.); (Y.X.)
- Zhejiang University Innovation Institute for Artificial Intelligence in Medicine, Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou 310018, China
| | - Ying Shen
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (Y.L.); (W.Z.); (Y.S.); (Y.X.)
- Zhejiang University Innovation Institute for Artificial Intelligence in Medicine, Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou 310018, China
| | - Yingchun Xu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (Y.L.); (W.Z.); (Y.S.); (Y.X.)
| | - Shuqing Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (Y.L.); (W.Z.); (Y.S.); (Y.X.)
| | - Liqiang Pan
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (Y.L.); (W.Z.); (Y.S.); (Y.X.)
| |
Collapse
|
5
|
Cattaneo I, Choblet S, Valgardsdottir R, Roth M, Massafra A, Beeg M, Gobbi M, Duonor-Cerutti M, Golay J. Development of a Bispecific IgG1 Antibody Targeting BCMA and PDL1. Antibodies (Basel) 2024; 13:15. [PMID: 38390876 PMCID: PMC10885062 DOI: 10.3390/antib13010015] [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: 12/07/2023] [Revised: 02/12/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024] Open
Abstract
We designed, produced, and purified a novel IgG1-like, bispecific antibody (bsAb) directed against B-cell maturation antigen (BCMA), expressed by multiple myeloma (MM) cells, and an immune checkpoint inhibitor (ICI), PDL1, expressed in the MM microenvironment. The BCMA×PDL1 bsAb was fully characterized in vitro. BCMA×PDL1 bound specifically and simultaneously, with nM affinity, to both native membrane-bound antigens and to the recombinant soluble antigen fragments, as shown by immunophenotyping analyses and surface plasmon resonance (SPR), respectively. The binding affinity of bsAb for PDL1 and BCMA was similar to each other, but PDL1 affinity was about 10-fold lower in the bsAb compared to parent mAb, probably due to the steric hindrance associated with the more internal anti-PDL1 Fab. The bsAb was also able to functionally block both antigen targets with IC50 in the nM range. The bsAb Fc was functional, inducing human-complement-dependent cytotoxicity as well as ADCC by NK cells in 24 h killing assays. Finally, BCMA×PDL1 was effective in 7-day killing assays with peripheral blood mononuclear cells as effectors, inducing up to 75% of target MM cell line killing at a physiologically attainable, 6 nM, concentration. These data provide the necessary basis for future optimization and in vivo testing of this novel bsAb.
Collapse
Affiliation(s)
- Irene Cattaneo
- Division of Hematology, Center of Cellular Therapy "G. Lanzani", Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, 24122 Bergamo, Italy
| | - Sylvie Choblet
- Centre National de la Recherche Scientifique UAR3426 "Baculovirus et Therapie", 30380 Saint-Christol-Lez-Alès, France
| | - Rut Valgardsdottir
- Division of Hematology, Center of Cellular Therapy "G. Lanzani", Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, 24122 Bergamo, Italy
| | - Muriel Roth
- Centre National de la Recherche Scientifique UAR3426 "Baculovirus et Therapie", 30380 Saint-Christol-Lez-Alès, France
| | - Annamaria Massafra
- Laboratory of Pharmacodynamics and Pharmacokinetics, Department of Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, 20157 Milan, Italy
| | - Marten Beeg
- Laboratory of Pharmacodynamics and Pharmacokinetics, Department of Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, 20157 Milan, Italy
| | - Marco Gobbi
- Laboratory of Pharmacodynamics and Pharmacokinetics, Department of Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, 20157 Milan, Italy
| | - Martine Duonor-Cerutti
- Centre National de la Recherche Scientifique UAR3426 "Baculovirus et Therapie", 30380 Saint-Christol-Lez-Alès, France
| | - Josée Golay
- Division of Hematology, Center of Cellular Therapy "G. Lanzani", Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, 24122 Bergamo, Italy
| |
Collapse
|
6
|
Ullah A, Shin G, Lim SI. Human serum albumin binders: A piggyback ride for long-acting therapeutics. Drug Discov Today 2023; 28:103738. [PMID: 37591409 DOI: 10.1016/j.drudis.2023.103738] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 07/29/2023] [Accepted: 08/10/2023] [Indexed: 08/19/2023]
Abstract
Human serum albumin (HSA) is the most abundant protein in the blood and has desirable properties as a drug carrier. One of the most promising ways to exploit HSA as a carrier is to append an albumin-binding moiety (ABM) to a drug for in situ HSA binding upon administration. Nature- and library-derived ABMs vary in size, affinity, and epitope, differentially improving the pharmacokinetics of an appended drug. In this review, we evaluate the current state of knowledge regarding various aspects of ABMs and the unique advantages of ABM-mediated drug delivery. Furthermore, we discuss how ABMs can be specifically modulated to maximize potential benefits in clinical development.
Collapse
Affiliation(s)
- Aziz Ullah
- Department of Chemical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Gomal Centre of Pharmaceutical Sciences, Faculty of Pharmacy, Gomal University, Dera Ismail Khan 29050, Khyber Pakhtunkhwa, Pakistan
| | - Goeun Shin
- Department of Chemical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Nbios Inc, 7, Jukheon-gil, Gangneung-si, Gangwon-do, Republic of Korea
| | - Sung In Lim
- Department of Chemical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Marine BioResource Co., Ltd., 365, Sinseon-ro, Nam-gu, Busan 48548, Republic of Korea.
| |
Collapse
|
7
|
Bai Z, Wang J, Li J, Yuan H, Wang P, Zhang M, Feng Y, Cao X, Cao X, Kang G, de Marco A, Huang H. Design of nanobody-based bispecific constructs by in silico affinity maturation and umbrella sampling simulations. Comput Struct Biotechnol J 2022; 21:601-613. [PMID: 36659922 PMCID: PMC9822835 DOI: 10.1016/j.csbj.2022.12.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Random mutagenesis is the natural opportunity for proteins to evolve and biotechnologically it has been exploited to create diversity and identify variants with improved characteristics in the mutant pools. Rational mutagenesis based on biophysical assumptions and supported by computational power has been proposed as a faster and more predictable strategy to reach the same aim. In this work we confirm that substantial improvements in terms of both affinity and stability of nanobodies can be obtained by using combinations of algorithms, even for binders with already high affinity and elevated thermal stability. Furthermore, in silico approaches allowed the development of an optimized bispecific construct able to bind simultaneously the two clinically relevant antigens TNF-α and IL-23 and, by means of its enhanced avidity, to inhibit effectively the apoptosis of TNF-α-sensitive L929 cells. The results revealed that salt bridges, hydrogen bonds, aromatic-aromatic and cation-pi interactions had a critical role in increasing affinity. We provided a platform for the construction of high-affinity bispecific constructs based on nanobodies that can have relevant applications for the control of all those biological mechanisms in which more than a single antigen must be targeted to increase the treatment effectiveness and avoid resistance mechanisms.
Collapse
Affiliation(s)
- Zixuan Bai
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Jiewen Wang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
- Institute of Shaoxing, Tianjin University, Zhejiang 312300, China
| | - Jiaqi Li
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
- Institute of Shaoxing, Tianjin University, Zhejiang 312300, China
| | - Haibin Yuan
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Ping Wang
- Tianjin Modern Innovative TCM Technology Co. Ltd., Tianjin, China
| | - Miao Zhang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
- China Resources Biopharmaceutical Company Limited, Beijing, China
| | - Yuanhang Feng
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Xiangtong Cao
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Xiangan Cao
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Guangbo Kang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
- Institute of Shaoxing, Tianjin University, Zhejiang 312300, China
| | - Ario de Marco
- Laboratory for Environmental and Life Sciences, University of Nova Gorica, Nova Gorica, Slovenia
| | - He Huang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
- Institute of Shaoxing, Tianjin University, Zhejiang 312300, China
| |
Collapse
|
8
|
You G, Won J, Lee Y, Moon D, Park Y, Lee SH, Lee SW. Bispecific Antibodies: A Smart Arsenal for Cancer Immunotherapies. Vaccines (Basel) 2021; 9:724. [PMID: 34358141 PMCID: PMC8310217 DOI: 10.3390/vaccines9070724] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/05/2021] [Accepted: 06/28/2021] [Indexed: 12/13/2022] Open
Abstract
Following the clinical success of cancer immunotherapies such as immune checkpoint inhibitors blocking B7/CTLA-4 or PD-1/PD-L1 signaling and ongoing numerous combination therapies in the clinic,3 bispecific antibodies (BsAbs) are now emerging as a growing class of immunotherapies with the potential to improve clinical efficacy and safety further. Here, we describe four classes of BsAbs: (a) immune effector cell redirectors; (b) tumor-targeted immunomodulators; (c) dual immunomodulators; and (d) dual tumor-targeting BsAbs. This review describes each of these classes of BsAbs and presents examples of BsAbs in development. We reviewed the biological rationales and characteristics of BsAbs and summarized the current status and limitations of clinical development of BsAbs and strategies to overcome limitations. The field of BsAb-based cancer immunotherapy is growing, and more data from clinical trials are accumulating. Thus, BsAbs could be the next generation of new treatment options for cancer patients.
Collapse
Affiliation(s)
- Gihoon You
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (G.Y.); (D.M.)
| | - Jonghwa Won
- ABL Bio Inc., Seongnam 13488, Korea; (J.W.); (Y.L.); (S.H.L.)
| | - Yangsoon Lee
- ABL Bio Inc., Seongnam 13488, Korea; (J.W.); (Y.L.); (S.H.L.)
| | - Dain Moon
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (G.Y.); (D.M.)
| | - Yunji Park
- Biotechcenter, POSTECH, Pohang 37673, Korea;
| | - Sang Hoon Lee
- ABL Bio Inc., Seongnam 13488, Korea; (J.W.); (Y.L.); (S.H.L.)
| | - Seung-Woo Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (G.Y.); (D.M.)
| |
Collapse
|
9
|
Omar MH, Scott JD. AKAP Signaling Islands: Venues for Precision Pharmacology. Trends Pharmacol Sci 2020; 41:933-946. [PMID: 33082006 DOI: 10.1016/j.tips.2020.09.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 12/19/2022]
Abstract
Regulatory enzymes often have different roles in distinct subcellular compartments. Yet, most drugs indiscriminately saturate the cell. Thus, subcellular drug-delivery holds promise as a means to reduce off-target pharmacological effects. A-kinase anchoring proteins (AKAPs) sequester combinations of signaling enzymes within subcellular microdomains. Targeting drugs to these 'signaling islands' offers an opportunity for more precise delivery of therapeutics. Here, we review mechanisms that bestow protein kinase A (PKA) versatility inside the cell, appraise recent advances in exploiting AKAPs as platforms for precision pharmacology, and explore the impact of methodological innovations on AKAP research.
Collapse
Affiliation(s)
- Mitchell H Omar
- Department of Pharmacology, University of Washington, Seattle, WA, 98195, USA
| | - John D Scott
- Department of Pharmacology, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
10
|
Guo ZS, Lotze MT, Zhu Z, Storkus WJ, Song XT. Bi- and Tri-Specific T Cell Engager-Armed Oncolytic Viruses: Next-Generation Cancer Immunotherapy. Biomedicines 2020; 8:E204. [PMID: 32664210 PMCID: PMC7400484 DOI: 10.3390/biomedicines8070204] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 02/07/2023] Open
Abstract
Oncolytic viruses (OVs) are potent anti-cancer biologics with a bright future, having substantial evidence of efficacy in patients with cancer. Bi- and tri-specific antibodies targeting tumor antigens and capable of activating T cell receptor signaling have also shown great promise in cancer immunotherapy. In a cutting-edge strategy, investigators have incorporated the two independent anti-cancer modalities, transforming them into bi- or tri-specific T cell engager (BiTE or TriTE)-armed OVs for targeted immunotherapy. Since 2014, multiple research teams have studied this combinatorial strategy, and it showed substantial efficacy in various tumor models. Here, we first provide a brief overview of the current status of oncolytic virotherapy and the use of multi-specific antibodies for cancer immunotherapy. We then summarize progress on BiTE and TriTE antibodies as a novel class of cancer therapeutics in preclinical and clinical studies, followed by a discussion of BiTE- or TriTE-armed OVs for cancer therapy in translational models. In addition, T cell receptor mimics (TCRm) have been developed into BiTEs and are expected to greatly expand the application of BiTEs and BiTE-armed OVs for the effective targeting of intracellular tumor antigens. Future applications of such innovative combination strategies are emerging as precision cancer immunotherapies.
Collapse
Affiliation(s)
- Zong Sheng Guo
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Michael T Lotze
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Zhi Zhu
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Walter J Storkus
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | |
Collapse
|