1
|
Tao J, Gu Y, Zhou W, Wang Y. Dual-payload antibody-drug conjugates: Taking a dual shot. Eur J Med Chem 2024; 281:116995. [PMID: 39481229 DOI: 10.1016/j.ejmech.2024.116995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024]
Abstract
Antibody-drug conjugates (ADCs) enable the precise delivery of cytotoxic agents by conjugating small-molecule drugs with monoclonal antibodies (mAbs). Over recent decades, ADCs have demonstrated substantial clinical efficacy. However, conventional ADCs often encounter various clinical challenges, including suboptimal efficacy, significant adverse effects, and the development of drug resistance, limiting their broader clinical application. Encouragingly, a next-generation approach-dual-payload ADCs-has emerged as a pioneering strategy to address these challenges. Dual-payload ADCs are characterized by the incorporation of two distinct therapeutic payloads on the same antibody, enhancing treatment efficacy by promoting synergistic effects and reducing the risk of drug resistance. However, the synthesis of dual-payload ADCs is complex due to the presence of multiple functional groups on antibodies. In this review, we comprehensively summarize the construction strategies for dual-payload ADCs, ranging from the design of ADC components to orthogonal chemistry. The subsequent sections explore current challenges and propose prospective strategies, highlighting recent advancements in dual-payload ADC research, thereby laying the foundation for the development of next-generation ADCs.
Collapse
Affiliation(s)
- Junjie Tao
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yilin Gu
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wei Zhou
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China.
| | - Yuxi Wang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| |
Collapse
|
2
|
Journeaux T, Bernardes GJL. Homogeneous multi-payload antibody-drug conjugates. Nat Chem 2024; 16:854-870. [PMID: 38760431 DOI: 10.1038/s41557-024-01507-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 03/14/2024] [Indexed: 05/19/2024]
Abstract
Many systemic cancer chemotherapies comprise a combination of drugs, yet all clinically used antibody-drug conjugates (ADCs) contain a single-drug payload. These combination regimens improve treatment outcomes by producing synergistic anticancer effects and slowing the development of drug-resistant cell populations. In an attempt to replicate these regimens and improve the efficacy of targeted therapy, the field of ADCs has moved towards developing techniques that allow for multiple unique payloads to be attached to a single antibody molecule with high homogeneity. However, the methods for generating such constructs-homogeneous multi-payload ADCs-are both numerous and complex owing to the plethora of reactive functional groups that make up the surface of an antibody. Here, by summarizing and comparing the methods of both single- and multi-payload ADC generation and their key preclinical and clinical results, we provide a timely overview of this relatively new area of research. The methods discussed range from branched linker installation to the incorporation of unnatural amino acids, with a generalized comparison tool of the most promising modification strategies also provided. Finally, the successes and challenges of this rapidly growing field are critically evaluated, and from this, future areas of research and development are proposed.
Collapse
Affiliation(s)
- Toby Journeaux
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Gonçalo J L Bernardes
- Department of Chemistry, University of Cambridge, Cambridge, UK.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
3
|
Yamazaki S, Ito K, Aoki T, Arashida N, Watanabe T, Fujii T, Matsuda Y. Biological Evaluation of Antibody-Drug Conjugates Produced by Tag-Free Lipoate Ligase A Modification. Biochemistry 2024; 63:644-650. [PMID: 38350078 DOI: 10.1021/acs.biochem.3c00513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
The concept of tag-free protein modification has attracted considerable interest in chemical biology because of its flexible and straightforward reaction process. In 2021, a groundbreaking approach using lipoate ligase A (LplA) for tag-free enzymatic modification of antibodies was unveiled, demonstrating its potential for the generation of precise antibody conjugates. In this study, to further explore LplA-mediated antibody-drug conjugate (ADC) synthesis, we performed initial biological evaluations of ADCs synthesized using LplA. Using the anti-HER2 antibody trastuzumab, we introduced octanoic acid azide using LplA and subsequently obtained an ADC using click chemistry with the drug DBCO-VC-PAB-MMAE. The bioactivity of the synthesized anti-HER2-ADC was evaluated using HER2-positive SKBR-3 and HER2-negative MCF7 cells. Its toxicity and selectivity were found to be comparable to those of the FDA-approved Kadcyla. In addition, a stability study involving rat and human plasma demonstrated the stability of the LplA-mediated ADC. Additionally, the affinity for the neonatal Fc receptor (FcRn) was retained after conjugation. These preliminary in vitro evaluations suggested that LplA-derived ADCs can have considerable pharmaceutical potential. Our results can set the stage for further in vivo evaluations and safety assessments. We suggest that the integration of tag-free LplA methods into the production of ADCs can offer a novel and promising approach for biopharmaceutical manufacturing.
Collapse
Affiliation(s)
- Shunsuke Yamazaki
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki 210-8681, Kanagawa, Japan
| | - Kenichiro Ito
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki 210-8681, Kanagawa, Japan
| | - Tsubasa Aoki
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki 210-8681, Kanagawa, Japan
| | - Naoko Arashida
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki 210-8681, Kanagawa, Japan
| | - Tomohiro Watanabe
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki 210-8681, Kanagawa, Japan
| | - Tomohiro Fujii
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki 210-8681, Kanagawa, Japan
| | - Yutaka Matsuda
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki 210-8681, Kanagawa, Japan
| |
Collapse
|
4
|
Shikida N, Yamazaki S, Takahashi K, Matsuda Y, Shimbo K. Analytical studies on the conjugation site specificity of trastuzumab modified by Escherichia coli lipoate ligase A: multiple-enzyme digestion approach for peptide mapping. Anal Bioanal Chem 2023; 415:6461-6469. [PMID: 37702772 DOI: 10.1007/s00216-023-04922-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/14/2023]
Abstract
Tag-free protein modification has received considerable attention in the field of chemical biology owing to the versatility and simplicity of the reaction sequence. In 2021, a novel tag-free enzymatic modification of antibodies utilizing lipoate ligase A (LplA) was reported to reveal its potential in the production of site-specific antibody conjugates. Primary peptide mapping analysis revealed the biased site specificity of antibodies modified by LplA; however, quantitative analysis remains challenging because of the complicated heterogeneity derived from biased selective modification. In an effort to further understand the site occupancy of LplA-modified antibodies, this study employed numerous unconventional techniques and strategies. Optimization of HPLC conditions and utilization of enzymes such as trypsin, Glu-C, and chymotrypsin significantly increased sequence data coverage. The transition from traditional spectral counting to a more accurate peak area-based label-free quantification helped better analyze peptide modification levels. The results obtained indicate that LplA-induced modifications are specific lysines, particularly the light chain Lys188/190 site, which have an increased modification rate compared to chemically induced modifications. This study not only contributes to the understanding of peptide modification, but also presents an improved methodology that promises to stimulate further research in this field.
Collapse
Affiliation(s)
- Natsuki Shikida
- Ajinomoto Co., Inc., 1-1 Suzuki-Cho, Kawasaki, Kanagawa, 210-8681, Japan
| | - Shunsuke Yamazaki
- Ajinomoto Co., Inc., 1-1 Suzuki-Cho, Kawasaki, Kanagawa, 210-8681, Japan
| | | | - Yutaka Matsuda
- Ajinomoto Co., Inc., 1-1 Suzuki-Cho, Kawasaki, Kanagawa, 210-8681, Japan.
| | - Kazutaka Shimbo
- Ajinomoto Co., Inc., 1-1 Suzuki-Cho, Kawasaki, Kanagawa, 210-8681, Japan.
| |
Collapse
|
5
|
Qian L, Lin X, Gao X, Khan RU, Liao JY, Du S, Ge J, Zeng S, Yao SQ. The Dawn of a New Era: Targeting the "Undruggables" with Antibody-Based Therapeutics. Chem Rev 2023. [PMID: 37186942 DOI: 10.1021/acs.chemrev.2c00915] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The high selectivity and affinity of antibodies toward their antigens have made them a highly valuable tool in disease therapy, diagnosis, and basic research. A plethora of chemical and genetic approaches have been devised to make antibodies accessible to more "undruggable" targets and equipped with new functions of illustrating or regulating biological processes more precisely. In this Review, in addition to introducing how naked antibodies and various antibody conjugates (such as antibody-drug conjugates, antibody-oligonucleotide conjugates, antibody-enzyme conjugates, etc.) work in therapeutic applications, special attention has been paid to how chemistry tools have helped to optimize the therapeutic outcome (i.e., with enhanced efficacy and reduced side effects) or facilitate the multifunctionalization of antibodies, with a focus on emerging fields such as targeted protein degradation, real-time live-cell imaging, catalytic labeling or decaging with spatiotemporal control as well as the engagement of antibodies inside cells. With advances in modern chemistry and biotechnology, well-designed antibodies and their derivatives via size miniaturization or multifunctionalization together with efficient delivery systems have emerged, which have gradually improved our understanding of important biological processes and paved the way to pursue novel targets for potential treatments of various diseases.
Collapse
Affiliation(s)
- Linghui Qian
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xuefen Lin
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xue Gao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Rizwan Ullah Khan
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jia-Yu Liao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shubo Du
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Jingyan Ge
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore, 117544
| |
Collapse
|
6
|
Subhan MA, Torchilin VP. Advances in Targeted Therapy of Breast Cancer with Antibody-Drug Conjugate. Pharmaceutics 2023; 15:1242. [PMID: 37111727 PMCID: PMC10144345 DOI: 10.3390/pharmaceutics15041242] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/12/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are a potential and promising therapy for a wide variety of cancers, including breast cancer. ADC-based drugs represent a rapidly growing field of breast cancer therapy. Various ADC drug therapies have progressed over the past decade and have generated diverse opportunities for designing of state-of-the-art ADCs. Clinical progress with ADCs for the targeted therapy of breast cancer have shown promise. Off-target toxicities and drug resistance to ADC-based therapy have hampered effective therapy development due to the intracellular mechanism of action and limited antigen expression on breast tumors. However, innovative non-internalizing ADCs targeting the tumor microenvironment (TME) component and extracellular payload delivery mechanisms have led to reduced drug resistance and enhanced ADC effectiveness. Novel ADC drugs may deliver potent cytotoxic agents to breast tumor cells with reduced off-target effects, which may overcome difficulties related to delivery efficiency and enhance the therapeutic efficacy of cytotoxic cancer drugs for breast cancer therapy. This review discusses the development of ADC-based targeted breast cancer therapy and the clinical translation of ADC drugs for breast cancer treatment.
Collapse
Affiliation(s)
- Md Abdus Subhan
- Department of Chemistry, ShahJalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Vladimir P. Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, North Eastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, North Eastern University, Boston, MA 02115, USA
| |
Collapse
|
7
|
Yamazaki S, Inoue K, Mihara Y, Matsuda Y. Tag‐Free Antibody Modification Mediated by Lipoic Acid Ligase A: Application to Antibody‐Drug Conjugates Production. ChemistrySelect 2023. [DOI: 10.1002/slct.202204706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Affiliation(s)
| | - Kota Inoue
- Ajinomoto Co., Inc. 1-1 Suzuki-cho 210-8681 Kawasaki Kanagawa Japan
| | - Yasuhiro Mihara
- Ajinomoto Co., Inc. 1-1 Suzuki-cho 210-8681 Kawasaki Kanagawa Japan
| | - Yutaka Matsuda
- Ajinomoto Bio-Pharma Services 11040 Roselle Street 92121 San Diego CA United States
| |
Collapse
|
8
|
Yamazaki S, Matsuda Y. Tag‐Free Enzymatic Modification for Antibody−Drug Conjugate Production. ChemistrySelect 2022. [DOI: 10.1002/slct.202203753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
| | - Yutaka Matsuda
- Ajinomoto Bio-Pharma Services 11040 Roselle Street San Diego CA 92121 United States
| |
Collapse
|
9
|
Zeng Y, Shi W, Dong Q, Li W, Zhang J, Ren X, Tang C, Liu B, Song Y, Wu Y, Diao X, Zhou H, Huang H, Tang F, Huang W. A Traceless Site‐Specific Conjugation on Native Antibodies Enables Efficient One‐Step Payload Assembly. Angew Chem Int Ed Engl 2022; 61:e202204132. [DOI: 10.1002/anie.202204132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Indexed: 11/05/2022]
Affiliation(s)
- Yue Zeng
- School of Pharmaceutical Science and Technology Hangzhou Institute of Advanced Study Hangzhou 310024 China
- CAS Key Laboratory of Receptor Research CAS Center for Excellence in Molecular Cell Science Shanghai Institute of Materia Medica Chinese Academy of Sciences No. 555 Zuchongzhi Road Pudong Shanghai 201203 China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
| | - Wei Shi
- School of Pharmaceutical Science and Technology Hangzhou Institute of Advanced Study Hangzhou 310024 China
- CAS Key Laboratory of Receptor Research CAS Center for Excellence in Molecular Cell Science Shanghai Institute of Materia Medica Chinese Academy of Sciences No. 555 Zuchongzhi Road Pudong Shanghai 201203 China
| | - Qian Dong
- School of Pharmaceutical Science and Technology Hangzhou Institute of Advanced Study Hangzhou 310024 China
- CAS Key Laboratory of Receptor Research CAS Center for Excellence in Molecular Cell Science Shanghai Institute of Materia Medica Chinese Academy of Sciences No. 555 Zuchongzhi Road Pudong Shanghai 201203 China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
| | - Wanzhen Li
- School of Chinese Materia Medica Nanjing University of Chinese Medicine No. 138 Xianlin Road Nanjing 210023 China
| | - Jianxin Zhang
- School of Chinese Materia Medica Nanjing University of Chinese Medicine No. 138 Xianlin Road Nanjing 210023 China
| | - Xuelian Ren
- Shanghai Institute of Materia Medica Chinese Academy of Sciences No. 555 Zuchongzhi Road Pudong Shanghai 201203 China
| | - Caihong Tang
- CAS Key Laboratory of Receptor Research CAS Center for Excellence in Molecular Cell Science Shanghai Institute of Materia Medica Chinese Academy of Sciences No. 555 Zuchongzhi Road Pudong Shanghai 201203 China
| | - Bo Liu
- School of Pharmaceutical Science and Technology Hangzhou Institute of Advanced Study Hangzhou 310024 China
- CAS Key Laboratory of Receptor Research CAS Center for Excellence in Molecular Cell Science Shanghai Institute of Materia Medica Chinese Academy of Sciences No. 555 Zuchongzhi Road Pudong Shanghai 201203 China
| | - Yuanli Song
- Shanghai Institute of Materia Medica Chinese Academy of Sciences No. 555 Zuchongzhi Road Pudong Shanghai 201203 China
| | - Yali Wu
- Shanghai Institute of Materia Medica Chinese Academy of Sciences No. 555 Zuchongzhi Road Pudong Shanghai 201203 China
| | - Xingxing Diao
- Shanghai Institute of Materia Medica Chinese Academy of Sciences No. 555 Zuchongzhi Road Pudong Shanghai 201203 China
| | - Hu Zhou
- Shanghai Institute of Materia Medica Chinese Academy of Sciences No. 555 Zuchongzhi Road Pudong Shanghai 201203 China
| | - He Huang
- Shanghai Institute of Materia Medica Chinese Academy of Sciences No. 555 Zuchongzhi Road Pudong Shanghai 201203 China
| | - Feng Tang
- School of Pharmaceutical Science and Technology Hangzhou Institute of Advanced Study Hangzhou 310024 China
- CAS Key Laboratory of Receptor Research CAS Center for Excellence in Molecular Cell Science Shanghai Institute of Materia Medica Chinese Academy of Sciences No. 555 Zuchongzhi Road Pudong Shanghai 201203 China
| | - Wei Huang
- School of Pharmaceutical Science and Technology Hangzhou Institute of Advanced Study Hangzhou 310024 China
- CAS Key Laboratory of Receptor Research CAS Center for Excellence in Molecular Cell Science Shanghai Institute of Materia Medica Chinese Academy of Sciences No. 555 Zuchongzhi Road Pudong Shanghai 201203 China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
- School of Chinese Materia Medica Nanjing University of Chinese Medicine No. 138 Xianlin Road Nanjing 210023 China
| |
Collapse
|
10
|
Hanby AR, Walsh SJ, Counsell AJ, Ashman N, Mortensen KT, Carroll JS, Spring DR. Antibody dual-functionalisation enabled through a modular divinylpyrimidine disulfide rebridging strategy. Chem Commun (Camb) 2022; 58:9401-9404. [PMID: 35912884 PMCID: PMC9387566 DOI: 10.1039/d2cc02515a] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/05/2022] [Indexed: 01/17/2023]
Abstract
Herein we report the development of a methodology for the dual-functionalisation of IgG antibodies. This is accomplished through the combination of disulfide rebridging divinylpyrimidine technology, with bicyclononyne and methylcyclopropene handles to facilitate sequential SPAAC and IEDDA reactions. Advantageously, the strategy does not require metal catalysis and avoids the need for purification between functionalisation steps.
Collapse
Affiliation(s)
- Abigail R Hanby
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | - Stephen J Walsh
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Andrew J Counsell
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | - Nicola Ashman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Kim T Mortensen
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - David R Spring
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| |
Collapse
|
11
|
Zeng Y, Shi W, Dong Q, Li W, Zhang J, Ren X, Tang C, Liu B, Song Y, Wu Y, Diao X, Zhou H, Huang H, Tang F, Huang W. A Traceless Site‐Specific Conjugation on Native Antibodies Enables Efficient One‐Step Payload Assembly. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202204132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Yue Zeng
- Shanghai Institute of Materia Medica CAS: Shanghai Institute of Materia Medica Chinese Academy of Sciences Biotherapeutic center CHINA
| | - Wei Shi
- Shanghai Institute of Materia Medica CAS: Shanghai Institute of Materia Medica Chinese Academy of Sciences Biotherapeutic center CHINA
| | - Qian Dong
- Shanghai Institute of Materia Medica CAS: Shanghai Institute of Materia Medica Chinese Academy of Sciences Biotherapeutic center CHINA
| | - Wanzhen Li
- Shanghai Institute of Materia Medica CAS: Shanghai Institute of Materia Medica Chinese Academy of Sciences Biotherapeutic center CHINA
| | - Jianxin Zhang
- Shanghai Institute of Materia Medica CAS: Shanghai Institute of Materia Medica Chinese Academy of Sciences Biotherapeutic center CHINA
| | - Xuelian Ren
- Shanghai Institute of Materia Medica CAS: Shanghai Institute of Materia Medica Chinese Academy of Sciences Biotherapeutic center CHINA
| | - Caihong Tang
- Shanghai Institute of Materia Medica CAS: Shanghai Institute of Materia Medica Chinese Academy of Sciences Biotherapeutic center CHINA
| | - Bo Liu
- Shanghai Institute of Materia Medica CAS: Shanghai Institute of Materia Medica Chinese Academy of Sciences Biotherapeutic center CHINA
| | - Yuanli Song
- Shanghai Institute of Materia Medica CAS: Shanghai Institute of Materia Medica Chinese Academy of Sciences Biotherapeutic center CHINA
| | - Yali Wu
- Shanghai Institute of Materia Medica CAS: Shanghai Institute of Materia Medica Chinese Academy of Sciences Biotherapeutic center 555 Zuchongzhi Rd CHINA
| | - Xingxing Diao
- Shanghai Institute of Materia Medica CAS: Shanghai Institute of Materia Medica Chinese Academy of Sciences Biotherapeutic center 555 Zuchongzhi Rd CHINA
| | - Hu Zhou
- Shanghai Institute of Materia Medica CAS: Shanghai Institute of Materia Medica Chinese Academy of Sciences Biotherapeutic center CHINA
| | - He Huang
- Shanghai Institute of Materia Medica CAS: Shanghai Institute of Materia Medica Chinese Academy of Sciences Biotherapeutic center CHINA
| | - Feng Tang
- Shanghai Institute of Materia Medica CAS: Shanghai Institute of Materia Medica Chinese Academy of Sciences Biotherapeutic center CHINA
| | - Wei Huang
- Shanghai Institute of Materia Medica Chinese Academy of Sciences Medicinal Chemistry Zuchongzhi Road 555 201203 Shanghai CHINA
| |
Collapse
|
12
|
Yamazaki S, Shikida N, Takahashi K, Matsuda Y, Inoue K, Shimbo K, Mihara Y. Lipoate-acid ligase a modification of native antibody: Synthesis and conjugation site analysis. Bioorg Med Chem Lett 2021; 51:128360. [PMID: 34537330 DOI: 10.1016/j.bmcl.2021.128360] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/10/2021] [Indexed: 12/23/2022]
Abstract
Bioconjugation is an important chemical biology research focus, especially in the development of methods to produce pharmaceutical bioconjugates and antibody-drug conjugates (ADCs). In this report, an enzyme-catalyzed conjugation method combined with a chemical reaction was used to modify a native antibody under mild reaction conditions. Our investigation revealed that lipoic-acid ligase (LplA) modifies native IgG1 with biased site-specificity. An intact mass analysis revealed that 98.3% of IgG1 was modified by LplA and possessed at least one molecule of octanocic acid. The average number of modifications per antibody was calculated to be 4.6. Peptide mapping analysis revealed that the modified residues were K225, K249 and K363 in the Fc region, and K30, K76 and K136 in the heavy chain and K39/K42, K169, K188 and K190 in the light chain of the Fab region. Careful evaluation including solvent exposed amino acid analysis suggested that these conjugate sites were not only solvent exposed but also biased by the site-specificity of LplA. Furthermore, antibody fragment conjugation may be able to take advantage of this enzymatic approach. This feasibility study serves as a demonstration for preparing enzymatically modified antibodies with conjugation site analysis.
Collapse
Affiliation(s)
- Shunsuke Yamazaki
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan.
| | - Natsuki Shikida
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan
| | | | - Yutaka Matsuda
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan
| | - Kota Inoue
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan
| | - Kazutaka Shimbo
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan.
| | - Yasuhiro Mihara
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan
| |
Collapse
|
13
|
Le Gall CM, van der Schoot JMS, Ramos-Tomillero I, Khalily MP, van Dalen FJ, Wijfjes Z, Smeding L, van Dalen D, Cammarata A, Bonger KM, Figdor CG, Scheeren FA, Verdoes M. Dual Site-Specific Chemoenzymatic Antibody Fragment Conjugation Using CRISPR-Based Hybridoma Engineering. Bioconjug Chem 2021; 32:301-310. [PMID: 33476135 PMCID: PMC7898269 DOI: 10.1021/acs.bioconjchem.0c00673] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
![]()
Functionalized antibodies
and antibody fragments have found applications
in the fields of biomedical imaging, theranostics, and antibody–drug
conjugates (ADC). In addition, therapeutic and theranostic approaches
benefit from the possibility to deliver more than one type of cargo
to target cells, further challenging stochastic labeling strategies.
Thus, bioconjugation methods to reproducibly obtain defined homogeneous
conjugates bearing multiple different cargo molecules, without compromising
target affinity, are in demand. Here, we describe a straightforward
CRISPR/Cas9-based strategy to rapidly engineer hybridoma cells to
secrete Fab′ fragments bearing two distinct site-specific labeling
motifs, which can be separately modified by two different sortase
A mutants. We show that sequential genetic editing of the heavy chain
(HC) and light chain (LC) loci enables the generation of a stable
cell line that secretes a dual tagged Fab′ molecule (DTFab′),
which can be easily isolated. To demonstrate feasibility, we functionalized
the DTFab′ with two distinct cargos in a site-specific manner.
This technology platform will be valuable in the development of multimodal
imaging agents, theranostics, and next-generation ADCs.
Collapse
Affiliation(s)
- Camille M Le Gall
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.,Oncode Institute, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Johan M S van der Schoot
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.,Oncode Institute, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Iván Ramos-Tomillero
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.,Institute for Chemical Immunology, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, Netherlands
| | - Melek Parlak Khalily
- Department of Synthetic Organic Chemistry, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Floris J van Dalen
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Zacharias Wijfjes
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.,Institute for Chemical Immunology, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, Netherlands
| | - Liyan Smeding
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Duco van Dalen
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Anna Cammarata
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Kimberly M Bonger
- Institute for Chemical Immunology, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, Netherlands.,Department of Synthetic Organic Chemistry, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.,Oncode Institute, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.,Institute for Chemical Immunology, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, Netherlands
| | - Ferenc A Scheeren
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Martijn Verdoes
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.,Institute for Chemical Immunology, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, Netherlands
| |
Collapse
|
14
|
Walsh SJ, Bargh JD, Dannheim FM, Hanby AR, Seki H, Counsell AJ, Ou X, Fowler E, Ashman N, Takada Y, Isidro-Llobet A, Parker JS, Carroll JS, Spring DR. Site-selective modification strategies in antibody-drug conjugates. Chem Soc Rev 2021; 50:1305-1353. [PMID: 33290462 DOI: 10.1039/d0cs00310g] [Citation(s) in RCA: 220] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antibody-drug conjugates (ADCs) harness the highly specific targeting capabilities of an antibody to deliver a cytotoxic payload to specific cell types. They have garnered widespread interest in drug discovery, particularly in oncology, as discrimination between healthy and malignant tissues or cells can be achieved. Nine ADCs have received approval from the US Food and Drug Administration and more than 80 others are currently undergoing clinical investigations for a range of solid tumours and haematological malignancies. Extensive research over the past decade has highlighted the critical nature of the linkage strategy adopted to attach the payload to the antibody. Whilst early generation ADCs were primarily synthesised as heterogeneous mixtures, these were found to have sub-optimal pharmacokinetics, stability, tolerability and/or efficacy. Efforts have now shifted towards generating homogeneous constructs with precise drug loading and predetermined, controlled sites of attachment. Homogeneous ADCs have repeatedly demonstrated superior overall pharmacological profiles compared to their heterogeneous counterparts. A wide range of methods have been developed in the pursuit of homogeneity, comprising chemical or enzymatic methods or a combination thereof to afford precise modification of specific amino acid or sugar residues. In this review, we discuss advances in chemical and enzymatic methods for site-specific antibody modification that result in the generation of homogeneous ADCs.
Collapse
Affiliation(s)
- Stephen J Walsh
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Sadiki A, Vaidya SR, Abdollahi M, Bhardwaj G, Dolan ME, Turna H, Arora V, Sanjeev A, Robinson TD, Koid A, Amin A, Zhou ZS. Site-specific conjugation of native antibody. Antib Ther 2020; 3:271-284. [PMID: 33644685 PMCID: PMC7906296 DOI: 10.1093/abt/tbaa027] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Traditionally, non-specific chemical conjugations, such as acylation of amines on lysine or alkylation of thiols on cysteines, are widely used; however, they have several shortcomings. First, the lack of site-specificity results in heterogeneous products and irreproducible processes. Second, potential modifications near the complementarity-determining region may reduce binding affinity and specificity. Conversely, site-specific methods produce well-defined and more homogenous antibody conjugates, ensuring developability and clinical applications. Moreover, several recent side-by-side comparisons of site-specific and stochastic methods have demonstrated that site-specific approaches are more likely to achieve their desired properties and functions, such as increased plasma stability, less variability in dose-dependent studies (particularly at low concentrations), enhanced binding efficiency, as well as increased tumor uptake. Herein, we review several standard and practical site-specific bioconjugation methods for native antibodies, i.e., those without recombinant engineering. First, chemo-enzymatic techniques, namely transglutaminase (TGase)-mediated transamidation of a conserved glutamine residue and glycan remodeling of a conserved asparagine N-glycan (GlyCLICK), both in the Fc region. Second, chemical approaches such as selective reduction of disulfides (ThioBridge) and N-terminal amine modifications. Furthermore, we list site-specific antibody–drug conjugates in clinical trials along with the future perspectives of these site-specific methods.
Collapse
Affiliation(s)
- Amissi Sadiki
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Shefali R Vaidya
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Mina Abdollahi
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Gunjan Bhardwaj
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Michael E Dolan
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA.,Downstream Development, Biologics Process Development, Millennium Pharmaceuticals, Inc., (a wholly-owned subsidiary of Takeda Pharmaceuticals Company Limited), Cambridge, Massachusetts 02139, USA
| | - Harpreet Turna
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Varnika Arora
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Athul Sanjeev
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Timothy D Robinson
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Andrea Koid
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Aashka Amin
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| | - Zhaohui Sunny Zhou
- Department of Chemistry and Chemical Biology, Northeastern University Boston, Massachusetts 02115-5000, USA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University Boston, Massachusetts 02115-5000, USA
| |
Collapse
|
16
|
Abel M, Burkenroad A, Sun A, Lu E, Stefanoudakis D, Drakaki A. The Evolving Landscape of Antibody-Drug Conjugates for Urothelial Carcinoma. Clin Genitourin Cancer 2020; 19:183-193. [PMID: 33558159 DOI: 10.1016/j.clgc.2020.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/07/2020] [Accepted: 11/18/2020] [Indexed: 12/24/2022]
Abstract
Metastatic urothelial carcinoma (UC) carries a poor prognosis and a 5-year overall survival of less than 5%, despite standard of care therapy using cisplatin-based chemotherapy and immune checkpoint inhibitors. Thus, novel agents that improve survival and have an acceptable toxicity profile are urgently needed. Antibody-drug conjugates (ADCs) represent a promising new treatment option that utilizes the targeting ability of an antibody to deliver cytotoxic drugs directly to tumors. Many ADCs are currently being investigated for treatment of UC, with enfortumab vedotin being recently approved by the US Food and Drug Administration for treatment of metastatic UC with progressive disease after chemotherapy and/or immune checkpoint inhibitors. Overall, ADCs hold promise as a long-awaited treatment option for UC.
Collapse
Affiliation(s)
- Melissa Abel
- Ronald Reagan UCLA Medical Center, Los Angeles, CA.
| | | | | | - Eric Lu
- Ronald Reagan UCLA Medical Center, Los Angeles, CA; UCLA Health Division of Hematology and Oncology, Los Angeles, CA
| | - Dimitrios Stefanoudakis
- Ronald Reagan UCLA Medical Center, Los Angeles, CA; University of Athens Medical School, Athens, Greece
| | - Alexandra Drakaki
- Ronald Reagan UCLA Medical Center, Los Angeles, CA; UCLA Health Division of Hematology and Oncology, Los Angeles, CA
| |
Collapse
|
17
|
Zhao P, Zhang Y, Li W, Jeanty C, Xiang G, Dong Y. Recent advances of antibody drug conjugates for clinical applications. Acta Pharm Sin B 2020; 10:1589-1600. [PMID: 33088681 PMCID: PMC7564033 DOI: 10.1016/j.apsb.2020.04.012] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/25/2020] [Accepted: 03/08/2020] [Indexed: 02/06/2023] Open
Abstract
Antibody drug conjugates (ADCs) normally compose of a humanized antibody and small molecular drug via a chemical linker. After decades of preclinical and clinical studies, a series of ADCs have been widely used for treating specific tumor types in the clinic such as brentuximab vedotin (Adcetris®) for relapsed Hodgkin's lymphoma and systemic anaplastic large cell lymphoma, gemtuzumab ozogamicin (Mylotarg®) for acute myeloid leukemia, ado-trastuzumab emtansine (Kadcyla®) for HER2-positive metastatic breast cancer, inotuzumab ozogamicin (Besponsa®) and most recently polatuzumab vedotin-piiq (Polivy®) for B cell malignancies. More than eighty ADCs have been investigated in different clinical stages from approximately six hundred clinical trials to date. This review summarizes the key elements of ADCs and highlights recent advances of ADCs, as well as important lessons learned from clinical data, and future directions.
Collapse
|
18
|
Baalmann M, Neises L, Bitsch S, Schneider H, Deweid L, Werther P, Ilkenhans N, Wolfring M, Ziegler MJ, Wilhelm J, Kolmar H, Wombacher R. A Bioorthogonal Click Chemistry Toolbox for Targeted Synthesis of Branched and Well-Defined Protein-Protein Conjugates. Angew Chem Int Ed Engl 2020; 59:12885-12893. [PMID: 32342666 PMCID: PMC7496671 DOI: 10.1002/anie.201915079] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/23/2020] [Indexed: 01/19/2023]
Abstract
Bioorthogonal chemistry holds great potential to generate difficult-to-access protein-protein conjugate architectures. Current applications are hampered by challenging protein expression systems, slow conjugation chemistry, use of undesirable catalysts, or often do not result in quantitative product formation. Here we present a highly efficient technology for protein functionalization with commonly used bioorthogonal motifs for Diels-Alder cycloaddition with inverse electron demand (DAinv ). With the aim of precisely generating branched protein chimeras, we systematically assessed the reactivity, stability and side product formation of various bioorthogonal chemistries directly at the protein level. We demonstrate the efficiency and versatility of our conjugation platform using different functional proteins and the therapeutic antibody trastuzumab. This technology enables fast and routine access to tailored and hitherto inaccessible protein chimeras useful for a variety of scientific disciplines. We expect our work to substantially enhance antibody applications such as immunodetection and protein toxin-based targeted cancer therapies.
Collapse
Affiliation(s)
- Mathis Baalmann
- Institute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityIm Neuenheimer Feld 36469120HeidelbergGermany
| | - Laura Neises
- Institute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityIm Neuenheimer Feld 36469120HeidelbergGermany
| | - Sebastian Bitsch
- Institute for Organic Chemistry and BiochemistryTechnische Universität DarmstadtAlarich-Weiss-Straße 464287DarmstadtGermany
| | - Hendrik Schneider
- Institute for Organic Chemistry and BiochemistryTechnische Universität DarmstadtAlarich-Weiss-Straße 464287DarmstadtGermany
| | - Lukas Deweid
- Institute for Organic Chemistry and BiochemistryTechnische Universität DarmstadtAlarich-Weiss-Straße 464287DarmstadtGermany
| | - Philipp Werther
- Institute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityIm Neuenheimer Feld 36469120HeidelbergGermany
| | - Nadja Ilkenhans
- Institute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityIm Neuenheimer Feld 36469120HeidelbergGermany
| | - Martin Wolfring
- Institute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityIm Neuenheimer Feld 36469120HeidelbergGermany
| | - Michael J. Ziegler
- Institute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityIm Neuenheimer Feld 36469120HeidelbergGermany
| | - Jonas Wilhelm
- Institute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityIm Neuenheimer Feld 36469120HeidelbergGermany
| | - Harald Kolmar
- Institute for Organic Chemistry and BiochemistryTechnische Universität DarmstadtAlarich-Weiss-Straße 464287DarmstadtGermany
| | - Richard Wombacher
- Institute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityIm Neuenheimer Feld 36469120HeidelbergGermany
| |
Collapse
|
19
|
Walsh SJ, Iegre J, Seki H, Bargh JD, Sore HF, Parker JS, Carroll JS, Spring DR. General dual functionalisation of biomacromolecules via a cysteine bridging strategy. Org Biomol Chem 2020; 18:4224-4230. [PMID: 32432632 DOI: 10.1039/d0ob00907e] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Site-selective modification of peptides and proteins has resulted in the development of a host of novel tools for the study of cellular systems or the synthesis of enhanced biotherapeutics. There is a need for useful methodologies that enable site-selective modification of native peptides or proteins, which is even more prevalent when modification of the biomolecule with multiple payloads is desired. Herein, we report the development of a novel dual functional divinylpyrimidine (dfDVP) platform that enables robust and modular modification of peptides, antibody fragments and antibodies. These biomacromolecules could be easily functionalised with a range of functional payloads (e.g. fluorescent dyes, cytotoxic warheads or cell-penetrating tags). Importantly, the dual functionalised peptides and antibodies demonstrated exquisite bioactivity in a range of in vitro cellular assays, showcasing the enhanced utility of these bioactive conjugates.
Collapse
Affiliation(s)
- Stephen J Walsh
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK. and Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Jessica Iegre
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | - Hikaru Seki
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | - Jonathan D Bargh
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | - Hannah F Sore
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | - Jeremy S Parker
- Early Chemical Development, Pharmaceutical Development, R&D, AstraZeneca, Macclesfield, UK
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - David R Spring
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| |
Collapse
|
20
|
Baalmann M, Neises L, Bitsch S, Schneider H, Deweid L, Werther P, Ilkenhans N, Wolfring M, Ziegler MJ, Wilhelm J, Kolmar H, Wombacher R. A Bioorthogonal Click Chemistry Toolbox for Targeted Synthesis of Branched and Well‐Defined Protein–Protein Conjugates. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201915079] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Mathis Baalmann
- Institute of Pharmacy and Molecular Biotechnology Heidelberg University Im Neuenheimer Feld 364 69120 Heidelberg Germany
| | - Laura Neises
- Institute of Pharmacy and Molecular Biotechnology Heidelberg University Im Neuenheimer Feld 364 69120 Heidelberg Germany
| | - Sebastian Bitsch
- Institute for Organic Chemistry and Biochemistry Technische Universität Darmstadt Alarich-Weiss-Straße 4 64287 Darmstadt Germany
| | - Hendrik Schneider
- Institute for Organic Chemistry and Biochemistry Technische Universität Darmstadt Alarich-Weiss-Straße 4 64287 Darmstadt Germany
| | - Lukas Deweid
- Institute for Organic Chemistry and Biochemistry Technische Universität Darmstadt Alarich-Weiss-Straße 4 64287 Darmstadt Germany
| | - Philipp Werther
- Institute of Pharmacy and Molecular Biotechnology Heidelberg University Im Neuenheimer Feld 364 69120 Heidelberg Germany
| | - Nadja Ilkenhans
- Institute of Pharmacy and Molecular Biotechnology Heidelberg University Im Neuenheimer Feld 364 69120 Heidelberg Germany
| | - Martin Wolfring
- Institute of Pharmacy and Molecular Biotechnology Heidelberg University Im Neuenheimer Feld 364 69120 Heidelberg Germany
| | - Michael J. Ziegler
- Institute of Pharmacy and Molecular Biotechnology Heidelberg University Im Neuenheimer Feld 364 69120 Heidelberg Germany
| | - Jonas Wilhelm
- Institute of Pharmacy and Molecular Biotechnology Heidelberg University Im Neuenheimer Feld 364 69120 Heidelberg Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry Technische Universität Darmstadt Alarich-Weiss-Straße 4 64287 Darmstadt Germany
| | - Richard Wombacher
- Institute of Pharmacy and Molecular Biotechnology Heidelberg University Im Neuenheimer Feld 364 69120 Heidelberg Germany
| |
Collapse
|
21
|
Doti N, Caporale A, Monti A, Sandomenico A, Selis F, Ruvo M. A recent update on the use of microbial transglutaminase for the generation of biotherapeutics. World J Microbiol Biotechnol 2020; 36:53. [PMID: 32172335 DOI: 10.1007/s11274-020-02829-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/07/2020] [Indexed: 01/12/2023]
Abstract
The recent scientific progresses on the use of enzyme-mediated reactions in organic, non-aqueous and aqueous media have significantly supported the growing demand of new biotechnological and/or pharmacological products. Today, a plethora of microbial enzymes, used as biocatalysts, are available. Among these, microbial transglutaminases (MTGs) are broadly used for their ability to catalyse the formation of an isopeptide bond between the γ-amide group of glutamines and the ε-amino group of lysine. Due to their promiscuity towards primary amine-containing substrates and the more stringent specificity for glutamine-containing peptide sequences, several combined approaches can be tailored for different settings, making MTGs very attractive catalysts for generating protein-protein and protein small molecule's conjugates. The present review offers a recent update on the modifications attainable by MTG-catalysed bioreactions as reported between 2014 and 2019. In particular, we present a detailed and comparative overview on the MTG-based methods for proteins and antibodies engineering, with a particular outlook on the synthesis of homogeneous antibody-drug conjugates.
Collapse
Affiliation(s)
- N Doti
- Institute of Biostructure and Bioimaging, CNR (IBB-CNR), Via Mezzocannone, 16, 80134, Naples, Italy.
| | - A Caporale
- Institute of Crystallography, CNR (IC-CNR), c/o Area Science Park s.s. 14 Km 163.5, Basovizza, 34149, Trieste, Italy
| | - Alessandra Monti
- Institute of Biostructure and Bioimaging, CNR (IBB-CNR), Via Mezzocannone, 16, 80134, Naples, Italy.,Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABIF), University L. Vanvitelli, Via Vivaldi, 43, 80100, Caserta, Italy
| | - A Sandomenico
- Institute of Biostructure and Bioimaging, CNR (IBB-CNR), Via Mezzocannone, 16, 80134, Naples, Italy
| | - Fabio Selis
- BioVIIIx R&D, Via B. Brin, 59C, 80142, Naples, Italy
| | - M Ruvo
- Institute of Biostructure and Bioimaging, CNR (IBB-CNR), Via Mezzocannone, 16, 80134, Naples, Italy.
| |
Collapse
|
22
|
Recent progress in transglutaminase-mediated assembly of antibody-drug conjugates. Anal Biochem 2020; 595:113615. [PMID: 32035039 DOI: 10.1016/j.ab.2020.113615] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/17/2020] [Accepted: 02/04/2020] [Indexed: 02/08/2023]
Abstract
Antibody-drug conjugates (ADCs) are hybrid molecules intended to overcome the drawbacks of conventional small molecule chemotherapy and therapeutic antibodies by merging beneficial characteristics of both molecule classes to develop more efficient and patient-friendly options for cancer treatment. During the last decades a versatile bioconjugation toolbox that comprises numerous chemical and enzymatic technologies have been developed to covalently attach a cytotoxic cargo to a tumor-targeting antibody. Microbial transglutaminase (mTG) that catalyzes isopeptide bond formation between proteinaceous or peptidic glutamines and lysines, provides many favorable properties that are beneficial for the manufacturing of these conjugates. However, to efficiently utilize the enzyme for the constructions of ADCs, different drawbacks had to be overcome that originate from the enzyme's insufficiently understood substrate specificity. Within this review, pioneering methodologies, recent achievements and remaining limitations of mTG-assisted assembly of ADCs will be highlighted.
Collapse
|
23
|
Responsive Antibody Conjugates Enable Quantitative Determination of Intracellular Bond Degradation Rate. Cell Chem Biol 2019; 26:1643-1651.e4. [PMID: 31604616 DOI: 10.1016/j.chembiol.2019.09.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/06/2019] [Accepted: 09/13/2019] [Indexed: 01/24/2023]
Abstract
Degradable crosslinkers that respond to intracellular biological stimuli are a critical component of many drug delivery systems. With numerous stimuli-responsive drug delivery systems in development, it is important to quantitatively study their intracellular processing. Herein we report a framework for quantifying the rate of intracellular bond degradation in the endocytic pathway. Toward this end, we devised and synthesized a reduction-sensitive FRET-based crosslinker that can be readily conjugated to a variety of targeting ligands. This crosslinker was conjugated to trastuzumab, a humanized monoclonal antibody against the HER2 receptor. We developed a model based on mass-action kinetics to describe the intracellular processing of this conjugate. The kinetic model was developed in conjunction with live-cell experiments to extract the rate constant for intracellular disulfide bond degradation. This framework may be applied to other endocytosis pathways, bond types, and cell types to quantify this fundamental degradation rate parameter.
Collapse
|
24
|
Lee MD, Tong WY, Nebl T, Pearce LA, Pham TM, Golbaz-Hagh A, Puttick S, Rose S, Adams TE, Williams CC. Dual Site-Specific Labeling of an Antibody Fragment through Sortase A and π-Clamp Conjugation. Bioconjug Chem 2019; 30:2539-2543. [DOI: 10.1021/acs.bioconjchem.9b00639] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Michael D. Lee
- CSIRO Manufacturing, Parkville, Victoria 3052, Australia
| | - Wing Yin Tong
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Thomas Nebl
- CSIRO Manufacturing, Parkville, Victoria 3052, Australia
| | | | - Tam M. Pham
- CSIRO Manufacturing, Parkville, Victoria 3052, Australia
| | - Arghavan Golbaz-Hagh
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, 4072, Australia
| | - Simon Puttick
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, 4072, Australia
- CSIRO Health and Biosecurity, Herston, Queensland 4029, Australia
| | - Stephen Rose
- CSIRO Health and Biosecurity, Herston, Queensland 4029, Australia
| | | | | |
Collapse
|