1
|
Fawcett C, Watson J, Richards S, Doherty AE, Seki H, Love EA, Coles CH, Coe DM, Jamieson C. Comparative Study of Click Handle Stability in Common Ligation Conditions. Bioconjug Chem 2025. [PMID: 40287825 DOI: 10.1021/acs.bioconjchem.5c00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
Click chemistry efficiently ligates molecular building blocks in a robust and high-yielding manner and has found major application in the rapid modification of important molecular actors in biological systems. However, the high reactivity of click handles often correlates with decreased stability, which presents a significant challenge in the practical application of these systems. In the current study, we describe a survey of the stability of commonly deployed click manifolds across a range of widely used ligation conditions. Incompatible click handle and ligation condition combinations are identified, with kinetic half-lives and side products of each undesired reaction determined, including the assessment of stability over extended periods and in a protein environment. This data set provides researchers with a roadmap to expediently determine the most appropriate click reaction conditions for any given bioorthogonal application, thus elevating the probability of success of procedures that utilize click chemistry.
Collapse
Affiliation(s)
- Caitlin Fawcett
- Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Joe Watson
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Stephen Richards
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Alfred E Doherty
- Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Hikaru Seki
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Elizabeth A Love
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Charlotte H Coles
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Diane M Coe
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Craig Jamieson
- Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom
| |
Collapse
|
2
|
Thanasi IA, Bouloc N, McMahon C, Wang N, Szijj PA, Butcher T, Rochet LNC, Love EA, Merritt A, Baker JR, Chudasama V. Formation of mono- and dual-labelled antibody fragment conjugates via reversible site-selective disulfide modification and proximity induced lysine reactivity. Chem Sci 2025; 16:2763-2776. [PMID: 39811008 PMCID: PMC11726237 DOI: 10.1039/d4sc06500j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Many protein bioconjugation strategies focus on the modification of lysine residues owing to the nucleophilicity of their amine side-chain, the generally high abundance of lysine residues on a protein's surface and the ability to form robustly stable amide-based bioconjugates. However, the plethora of solvent accessible lysine residues, which often have similar reactivity, is a key inherent issue when searching for regioselectivity and/or controlled loading of an entity. A relevant example is the modification of antibodies and/or antibody fragments, whose conjugates offer potential for a wide variety of applications. Thus, research in this area for the controlled loading of an entity via reaction with lysine residues is of high importance. In this article, we present an approach to achieve this by exploiting the quantitative and reversible site-selective modification of disulfides using pyridazinediones, which facilitates near-quantitative proximity-induced reactions with lysines to enable controlled loading of an entity. The strategy was appraised on several clinically relevant antibody fragments and enabled the formation of mono-labelled lysine-modified antibody fragment conjugates via the formation of stable amide bonds and the use of click chemistry for modular modification. Furthermore, through the use of multiple cycles of this novel strategy, an orthogonally bis-labelled lysine-modified antibody fragment conjugate was also furnished.
Collapse
Affiliation(s)
- Ioanna A Thanasi
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Nathalie Bouloc
- LifeArc, Accelerator Building Open Innovation Campus Stevenage SG1 2FX UK
| | - Clíona McMahon
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Ning Wang
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Peter A Szijj
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Tobias Butcher
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Léa N C Rochet
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Elizabeth A Love
- LifeArc, Accelerator Building Open Innovation Campus Stevenage SG1 2FX UK
| | - Andy Merritt
- LifeArc, Accelerator Building Open Innovation Campus Stevenage SG1 2FX UK
| | - James R Baker
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Vijay Chudasama
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| |
Collapse
|
3
|
Vaur V, Koutsopetras I, Erb S, Jackowska B, Benazza R, Cahuzac H, Detappe A, Hernandez-Alba O, Cianférani S, Scott CJ, Chaubet G. Chemical Production of Cytotoxic Bispecific Antibodies Using the Ugi Multicomponent Reaction. Chembiochem 2024; 25:e202400170. [PMID: 38713134 DOI: 10.1002/cbic.202400170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 05/08/2024]
Abstract
Bispecific antibodies (bsAbs) have recently emerged as a promising platform for the treatment of several conditions, most importantly cancer. Based on the combination of two different antigen-binding motifs in a single macromolecule; bsAbs can either display the combined characteristics of their parent antibodies, or new therapeutic features, inaccessible by the sole combination of two distinct antibodies. While bsAbs are traditionally produced by molecular biology techniques, the chemical development of bsAbs holds great promises and strategies have just begun to surface. In this context, we took advantage of a chemical strategy based on the use of the Ugi reaction for the site-selective conjugation of whole antibodies and coupled the resulting conjugates in a bioorthogonal manner with Fab fragments, derived from various antibodies. We thus managed to produce five different bsAbs with 2 : 1 valency, with yields ranging from 20 % to 48 %, and showed that the affinity of the parent antibody was preserved in all bsAbs. We further demonstrated the interest of our strategy by producing two other bsAbs behaving as cytotoxic T cell engagers with IC50 values in the picomolar range in vitro.
Collapse
Affiliation(s)
- Valentine Vaur
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament Strasbourg, University of Strasbourg, 74 Route du Rhin, 67400, Illkirch-Graffenstaden, France
| | - Ilias Koutsopetras
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament Strasbourg, University of Strasbourg, 74 Route du Rhin, 67400, Illkirch-Graffenstaden, France
| | - Stéphane Erb
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, 67000, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI - FR2048, 67087, Strasbourg, France
| | - Bianka Jackowska
- Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, BT9 7BL, U.K
| | - Rania Benazza
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, 67000, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI - FR2048, 67087, Strasbourg, France
| | - Héloïse Cahuzac
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament Strasbourg, University of Strasbourg, 74 Route du Rhin, 67400, Illkirch-Graffenstaden, France
| | | | - Oscar Hernandez-Alba
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, 67000, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI - FR2048, 67087, Strasbourg, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, CNRS, IPHC UMR 7178, 67000, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI - FR2048, 67087, Strasbourg, France
| | - Christopher J Scott
- Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, BT9 7BL, U.K
| | - Guilhem Chaubet
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament Strasbourg, University of Strasbourg, 74 Route du Rhin, 67400, Illkirch-Graffenstaden, France
| |
Collapse
|
4
|
Shao C, Tang B, Chu JCH, Lau KM, Wong WT, Che CM, Tai WCS, Wong WT, Wong CTT. Macrophage-engaging peptidic bispecific antibodies (pBsAbs) for immunotherapy via a facile bioconjugation strategy. Chem Sci 2024; 15:11272-11278. [PMID: 39055004 PMCID: PMC11268508 DOI: 10.1039/d4sc00851k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/29/2024] [Indexed: 07/27/2024] Open
Abstract
Bispecific antibodies are artificial molecules that fuse two different antigen-binding sites of monoclonal antibodies into one single entity. They have emerged as a promising next-generation anticancer treatment. Despite the fascinating applications of bispecific antibodies, the design and production of bispecific antibodies remain tedious and challenging, leading to a long R&D process and high production costs. We herein report an unprecedented strategy to cyclise and conjugate tumour-targeting peptides on the surface of a monoclonal antibody to form a novel type of bispecific antibody, namely the peptidic bispecific antibody (pBsAb). Such design combines the merits of highly specific monoclonal antibodies and serum-stable cyclic peptides that endows an additional tumour-targeting ability to the monoclonal antibody for binding with two different antigens. Our results show that the novel pBsAb, which comprises EGFR-binding cyclic peptides and an anti-SIRP-α monoclonal antibody, could serve as a macrophage-engaging bispecific antibody to initiate enhanced macrophage-cancer cell interaction and block the "don't eat me" signal between CD47-SIRP-α, as well as promoting antibody-dependent cellular phagocytosis and 3D cell spheroid infiltration. These findings give rise to a new type of bispecific antibody and a new platform for the rapid generation of new bispecific antibodies for research and potential therapeutic uses.
Collapse
Affiliation(s)
- Chihao Shao
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University Kowloon Hong Kong China
| | - Bo Tang
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University Kowloon Hong Kong China
| | - Jacky C H Chu
- Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F, Building 17W, Hong Kong Science Park New Territories Hong Kong China
| | - Kwai Man Lau
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University Kowloon Hong Kong China
| | - Wai-Ting Wong
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University Kowloon Hong Kong China
| | - Chi-Ming Che
- Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F, Building 17W, Hong Kong Science Park New Territories Hong Kong China
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, The University of Hong Kong Pokfulam Road Hong Kong China
| | - William C S Tai
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University Kowloon Hong Kong China
| | - Wing-Tak Wong
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University Kowloon Hong Kong China
| | - Clarence T T Wong
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University Kowloon Hong Kong China
| |
Collapse
|
5
|
Okon A, Yang J, Giancola JB, Molina OJ, Sayers J, Cheah KM, Li Y, Strieter ER, Raines RT. Facile Access to Branched Multispecific Proteins. Bioconjug Chem 2024; 35:954-962. [PMID: 38879814 PMCID: PMC11254548 DOI: 10.1021/acs.bioconjchem.4c00162] [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] [Indexed: 07/18/2024]
Abstract
Approaches that leverage orthogonal chemical reactions to generate protein-protein conjugates have expanded access to bespoke chimeras. Although the literature is replete with examples of the semisynthesis of bispecific proteins, few methods exist for the semisynthesis of protein conjugates of higher complexity (i.e., greater than two-protein fusions). The recent emergence of trispecific cell engagers for immune cell redirection therapies necessitates the development of chemical methods for the construction of trispecific proteins that would otherwise be inaccessible via natural protein synthesis. Here, we demonstrate that 3-bromo-5-methylene pyrrolone (3Br-5MP) can be used to effect the facile chemical synthesis of trispecific peptides and proteins with exquisite control over the addition of each monomer. The multimeric complexes maintain epitope functionality both in human cells and upon immobilization. We anticipate that facile access to trispecific proteins using this 3Br-5MP will have broad utility in basic science research and will quicken the pace of research to establish novel, multimeric immune cell redirection therapies.
Collapse
Affiliation(s)
- Aniekan Okon
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jinyi Yang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - JoLynn B. Giancola
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Oscar J. Molina
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jessica Sayers
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Keith M. Cheah
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Yanfeng Li
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Eric R. Strieter
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Ronald T. Raines
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
6
|
Rocha Tapia A, Abgottspon F, Nilvebrant J, Nygren PÅ, Duclos Ivetich S, Bello Hernandez AJ, Thanasi IA, Szijj PA, Sekkat G, Cuenot FM, Chudasama V, Aceto N, deMello AJ, Richards DA. Site-directed conjugation of single-stranded DNA to affinity proteins: quantifying the importance of conjugation strategy. Chem Sci 2024; 15:8982-8992. [PMID: 38873052 PMCID: PMC11168188 DOI: 10.1039/d4sc01838a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 04/27/2024] [Indexed: 06/15/2024] Open
Abstract
Affinity protein-oligonucleotide conjugates are increasingly being explored as diagnostic and therapeutic tools. Despite growing interest, these probes are typically constructed using outdated, non-selective chemistries, and little has been done to investigate how conjugation to oligonucleotides influences the function of affinity proteins. Herein, we report a novel site-selective conjugation method for furnishing affinity protein-oligonucleotide conjugates in a 93% yield within fifteen minutes. Using SPR, we explore how the choice of affinity protein, conjugation strategy, and DNA length impact target binding and reveal the deleterious effects of non-specific conjugation methods. Furthermore, we show that these adverse effects can be minimised by employing our site-selective conjugation strategy, leading to improved performance in an immuno-PCR assay. Finally, we investigate the interactions between affinity protein-oligonucleotide conjugates and live cells, demonstrating the benefits of site-selective conjugation. This work provides critical insight into the importance of conjugation strategy when constructing affinity protein-oligonucleotide conjugates.
Collapse
Affiliation(s)
- Andres Rocha Tapia
- Institute for Chemical and Bioengineering, ETH Zurich Vladimir-Prelog-Weg 1 8093 Zürich Switzerland
| | - Fabrice Abgottspon
- Institute for Chemical and Bioengineering, ETH Zurich Vladimir-Prelog-Weg 1 8093 Zürich Switzerland
| | - Johan Nilvebrant
- Department of Protein Science, KTH Royal Institute of Technology, AlbaNova University Center 106 91 Stockholm Sweden
| | - Per-Åke Nygren
- Department of Protein Science, KTH Royal Institute of Technology, AlbaNova University Center 106 91 Stockholm Sweden
| | - Sarah Duclos Ivetich
- Institute for Chemical and Bioengineering, ETH Zurich Vladimir-Prelog-Weg 1 8093 Zürich Switzerland
| | | | - Ioanna A Thanasi
- Department of Chemistry, University College London 20 Gordon Street WC1H 0AJ London UK
| | - Peter A Szijj
- Department of Chemistry, University College London 20 Gordon Street WC1H 0AJ London UK
| | - Ghali Sekkat
- Institute for Chemical and Bioengineering, ETH Zurich Vladimir-Prelog-Weg 1 8093 Zürich Switzerland
| | - François M Cuenot
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich Otto-Stern-Weg 7 8093 Zürich Switzerland
| | - Vijay Chudasama
- Department of Chemistry, University College London 20 Gordon Street WC1H 0AJ London UK
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich Otto-Stern-Weg 7 8093 Zürich Switzerland
| | - Andrew J deMello
- Institute for Chemical and Bioengineering, ETH Zurich Vladimir-Prelog-Weg 1 8093 Zürich Switzerland
| | - Daniel A Richards
- Institute for Chemical and Bioengineering, ETH Zurich Vladimir-Prelog-Weg 1 8093 Zürich Switzerland
| |
Collapse
|
7
|
Chauhan P, V R, Kumar M, Molla R, Mishra SD, Basa S, Rai V. Chemical technology principles for selective bioconjugation of proteins and antibodies. Chem Soc Rev 2024; 53:380-449. [PMID: 38095227 DOI: 10.1039/d3cs00715d] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Proteins are multifunctional large organic compounds that constitute an essential component of a living system. Hence, control over their bioconjugation impacts science at the chemistry-biology-medicine interface. A chemical toolbox for their precision engineering can boost healthcare and open a gateway for directed or precision therapeutics. Such a chemical toolbox remained elusive for a long time due to the complexity presented by the large pool of functional groups. The precise single-site modification of a protein requires a method to address a combination of selectivity attributes. This review focuses on guiding principles that can segregate them to simplify the task for a chemical method. Such a disintegration systematically employs a multi-step chemical transformation to deconvolute the selectivity challenges. It constitutes a disintegrate (DIN) theory that offers additional control parameters for tuning precision in protein bioconjugation. This review outlines the selectivity hurdles faced by chemical methods. It elaborates on the developments in the perspective of DIN theory to demonstrate simultaneous regulation of reactivity, chemoselectivity, site-selectivity, modularity, residue specificity, and protein specificity. It discusses the progress of such methods to construct protein and antibody conjugates for biologics, including antibody-fluorophore and antibody-drug conjugates (AFCs and ADCs). It also briefs how this knowledge can assist in developing small molecule-based covalent inhibitors. In the process, it highlights an opportunity for hypothesis-driven routes to accelerate discoveries of selective methods and establish new targetome in the precision engineering of proteins and antibodies.
Collapse
Affiliation(s)
- Preeti Chauhan
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Ragendu V
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Mohan Kumar
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Rajib Molla
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Surya Dev Mishra
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Sneha Basa
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Vishal Rai
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| |
Collapse
|
8
|
Fawcett C, Tickle JR, Coles CH. Facilitating high throughput bispecific antibody production and potential applications within biopharmaceutical discovery workflows. MAbs 2024; 16:2311992. [PMID: 39674918 DOI: 10.1080/19420862.2024.2311992] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/08/2024] [Accepted: 01/25/2024] [Indexed: 12/17/2024] Open
Abstract
A major driver for the recent investment surge in bispecific antibody (bsAb) platforms and products is the multitude of distinct mechanisms of action that bsAbs offer compared to a combination of two monoclonal antibodies. Four bsAb products were granted first regulatory approvals in the US or EU during 2023 and the biopharmaceutical industry pipeline is brimming with bsAb candidates across a broad range of therapeutic applications. In previously reported bsAb discovery campaigns, following a hypothesis-based choice of two specific target proteins, selections and screening activities have often been performed in mono-specific formats. The conversion to bispecific modalities has usually been positioned toward the end of the discovery process and has involved small numbers of lead molecules, largely due to challenges in expressing, purifying, and analyzing large numbers of bsAbs. In this review, we discuss emerging strategies to facilitate the production of expanded bsAb panels, focusing particularly upon combinatorial methods to generate bsAb matrices. Such technologies will enable screening in. bispecific formats at earlier stages of discovery campaigns, not only widening the accessible protein space to maximize chances of success, but also advancing empirical bi-target validation activities to assess initial target selection hypotheses.
Collapse
Affiliation(s)
- Caitlin Fawcett
- Large Molecule Discovery, GSK, GSK Medicines Research Centre, Stevenage, UK
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Joseph R Tickle
- Large Molecule Discovery, GSK, GSK Medicines Research Centre, Stevenage, UK
| | - Charlotte H Coles
- Large Molecule Discovery, GSK, GSK Medicines Research Centre, Stevenage, UK
| |
Collapse
|
9
|
Shajan I, Rochet LNC, Tracey SR, Jackowska B, Benazza R, Hernandez-Alba O, Cianférani S, Scott CJ, van Delft FL, Chudasama V, Albada B. Rapid Access to Potent Bispecific T Cell Engagers Using Biogenic Tyrosine Click Chemistry. Bioconjug Chem 2023; 34:2215-2220. [PMID: 37962868 PMCID: PMC10739583 DOI: 10.1021/acs.bioconjchem.3c00357] [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: 08/09/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023]
Abstract
Bispecific antibodies as T cell engagers designed to display binding capabilities to both tumor-associated antigens and antigens on T cells are considered promising agents in the fight against cancer. Even though chemical strategies to develop such constructs have emerged, a method that readily converts a therapeutically applied antibody into a bispecific construct by a fully non-genetic process is not yet available. Herein, we report the application of a biogenic, tyrosine-based click reaction utilizing chemoenzymatic modifications of native IgG1 antibodies to generate a synthetic bispecific antibody construct that exhibits tumor-killing capability at picomolar concentrations. Control experiments revealed that a covalent linkage of the different components is required for the observed biological activities. In view of the highly potent nature of the constructs and the modular approach that relies on convenient synthetic methods utilizing therapeutically approved biomolecules, our method expedites the production of potent bispecific antibody constructs with tunable cell killing efficacy with significant impact on therapeutic properties.
Collapse
Affiliation(s)
- Irene Shajan
- Laboratory
of Organic Chemistry, Wageningen University
& Research, Stippeneng 4, Wageningen 6807 WE, The Netherlands
| | - Léa N. C. Rochet
- Department
of Chemistry, University College London, 20 Gordon St, London WC1H 0AJ, U.K.
| | - Shannon R. Tracey
- Patrick
G Johnston Centre for Cancer Research, School of Medicine, Dentistry
and Biomedical Sciences, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7BL, U.K.
| | - Bianka Jackowska
- Patrick
G Johnston Centre for Cancer Research, School of Medicine, Dentistry
and Biomedical Sciences, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7BL, U.K.
| | - Rania Benazza
- Laboratoire
de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg 67000, France
- Infrastructure
Nationale de Protéomique ProFI − FR2048, Strasbourg 67087, France
| | - Oscar Hernandez-Alba
- Laboratoire
de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg 67000, France
- Infrastructure
Nationale de Protéomique ProFI − FR2048, Strasbourg 67087, France
| | - Sarah Cianférani
- Laboratoire
de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg 67000, France
- Infrastructure
Nationale de Protéomique ProFI − FR2048, Strasbourg 67087, France
| | - Christopher J. Scott
- Patrick
G Johnston Centre for Cancer Research, School of Medicine, Dentistry
and Biomedical Sciences, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7BL, U.K.
| | - Floris L. van Delft
- Laboratory
of Organic Chemistry, Wageningen University
& Research, Stippeneng 4, Wageningen 6807 WE, The Netherlands
- Synaffix
BV − A Lonza Company, Kloosterstraat 9, Oss 5349 AB, The Netherlands
| | - Vijay Chudasama
- Department
of Chemistry, University College London, 20 Gordon St, London WC1H 0AJ, U.K.
| | - Bauke Albada
- Laboratory
of Organic Chemistry, Wageningen University
& Research, Stippeneng 4, Wageningen 6807 WE, The Netherlands
| |
Collapse
|
10
|
Richard M, Martin Aubert S, Denis C, Dubois S, Nozach H, Truillet C, Kuhnast B. Fluorine-18 and Radiometal Labeling of Biomolecules via Disulfide Rebridging. Bioconjug Chem 2023; 34:2123-2132. [PMID: 37881943 DOI: 10.1021/acs.bioconjchem.3c00440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
Biomolecules labeled with positron-emitting radionuclides like fluorine-18 or radiometals like copper-64 and zirconium-89 are increasingly employed in nuclear medicine for diagnosis purposes. Given the fragility and complexity of these compounds, their labeling requires mild conditions. Besides, it is essential to develop methods inducing minimal modification of the tertiary structure, as it is fundamental for the biological activity of such complex entities. Given these requirements, disulfide rebridging represents a promising possibility since it allows protein modification as well as conservation of the tertiary structure. In this context, we have developed an original radiofluorinated dibromopyridazine dione prosthetic group for labeling of disulfide-containing biomolecules via rebridging. We employed it to radiolabel octreotide, a somatostatin analogue, and to radiolabel fragment antigen binding (Fab) targeting programmed death-ligand 1 (PD-L1), whose properties were then evaluated in vitro and in vivo by positron emission tomography (PET) imaging. We next extended our strategy to the radiolabeling of cetuximab, a monoclonal antibody, with various radiometals commonly used in PET imaging (zirconium-89, copper-64) by developing various rebridging molecules bearing the appropriate chelators. The stabilities of the radiolabeled antibody conjugates were assessed in biological conditions.
Collapse
Affiliation(s)
- Mylène Richard
- CEA, CNRS, Inserm, BioMaps, SHFJ, Paris-Saclay University, Orsay 91401, France
| | | | - Caroline Denis
- CEA, CNRS, Inserm, BioMaps, SHFJ, Paris-Saclay University, Orsay 91401, France
| | - Steven Dubois
- CEA, INRAE, Medicines and Healthcare Technologies Department, SIMoS, Paris-Saclay University, Gif-sur-Yvette 91191, France
| | - Hervé Nozach
- CEA, INRAE, Medicines and Healthcare Technologies Department, SIMoS, Paris-Saclay University, Gif-sur-Yvette 91191, France
| | - Charles Truillet
- CEA, CNRS, Inserm, BioMaps, SHFJ, Paris-Saclay University, Orsay 91401, France
| | - Bertrand Kuhnast
- CEA, CNRS, Inserm, BioMaps, SHFJ, Paris-Saclay University, Orsay 91401, France
| |
Collapse
|
11
|
Szijj PA, Gray MA, Ribi MK, Bahou C, Nogueira JCF, Bertozzi CR, Chudasama V. Chemical generation of checkpoint inhibitory T cell engagers for the treatment of cancer. Nat Chem 2023; 15:1636-1647. [PMID: 37488375 PMCID: PMC10624612 DOI: 10.1038/s41557-023-01280-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 06/21/2023] [Indexed: 07/26/2023]
Abstract
Bispecific T cell engagers (BiTEs), a subset of bispecific antibodies (bsAbs), can promote a targeted cancer cell's death by bringing it close to a cytotoxic T cell. Checkpoint inhibitory T cell engagers (CiTEs) comprise a BiTE core with an added immunomodulatory protein, which serves to reverse cancer-cell immune-dampening strategies, improving efficacy. So far, protein engineering has been the main approach to generate bsAbs and CiTEs, but improved chemical methods for their generation have recently been developed. Homogeneous fragment-based bsAbs constructed from fragment antigen-binding regions (Fabs) can be generated using click chemistry. Here we describe a chemical method to generate biotin-functionalized three-protein conjugates, which include two CiTE molecules, one containing an anti-PD-1 Fab and the other containing an immunomodulatory enzyme, Salmonella typhimurium sialidase. The CiTEs' efficacy was shown to be superior to that of the simpler BiTE scaffold, with the sialidase-containing CiTE inducing substantially enhanced T cell-mediated cytotoxicity in vitro. The chemical method described here, more generally, enables the generation of multi-protein constructs with further biological applications.
Collapse
Affiliation(s)
- Peter A Szijj
- Department of Chemistry, University College London, London, UK
| | - Melissa A Gray
- Department of Chemistry, Sarafan ChEM-H, and Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Mikaela K Ribi
- Department of Chemistry, Sarafan ChEM-H, and Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Calise Bahou
- Department of Chemistry, University College London, London, UK
| | | | - Carolyn R Bertozzi
- Department of Chemistry, Sarafan ChEM-H, and Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| | - Vijay Chudasama
- Department of Chemistry, University College London, London, UK.
| |
Collapse
|
12
|
Zhao Y, Chudasama V, Baker JR. Trifunctional Dibromomaleimide Reagents Built Around A Lysine Scaffold Deliver Site-selective Dual-modality Antibody Conjugation. Chembiochem 2023; 24:e202300356. [PMID: 37548625 DOI: 10.1002/cbic.202300356] [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: 05/11/2023] [Revised: 07/11/2023] [Indexed: 08/08/2023]
Abstract
We describe the synthesis and application of a selection of trifunctional reagents for the dual-modality modification of native, solvent accessible disulfide bonds in trastuzumab. The reagents were developed from the dibromomaleimide (DBM) platform with two orthogonal clickable functional groups built around a lysine core. We also describe the development of an aryl diselenide additive which enables antibody disulfide reduction in 4 minutes and a rapid overall reduction-bridging-double click sequence.
Collapse
Affiliation(s)
- Yanbo Zhao
- Department of Chemistry, University College London, 20 Gordon St, London, WC1H 0AJ, UK
| | - Vijay Chudasama
- Department of Chemistry, University College London, 20 Gordon St, London, WC1H 0AJ, UK
| | - James R Baker
- Department of Chemistry, University College London, 20 Gordon St, London, WC1H 0AJ, UK
| |
Collapse
|
13
|
Thoreau F, Rochet LNC, Baker JR, Chudasama V. Enabling the formation of native mAb, Fab' and Fc-conjugates using a bis-disulfide bridging reagent to achieve tunable payload-to-antibody ratios (PARs). Chem Sci 2023; 14:3752-3762. [PMID: 37035695 PMCID: PMC10074397 DOI: 10.1039/d2sc06318b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Either as full IgGs or as fragments (Fabs, Fc, etc.), antibodies have received tremendous attention in the development of new therapeutics such as antibody-drug conjugates (ADCs). The production of ADCs involves the grafting of active payloads onto an antibody, which is generally enabled by the site-selective modification of native or engineered antibodies via chemical or enzymatic methods. Whatever method is employed, controlling the payload-antibody ratio (PAR) is a challenge in terms of multiple aspects including: (i) obtaining homogeneous protein conjugates; (ii) obtaining unusual PARs (PAR is rarely other than 2, 4 or 8); (iii) using a single method to access a range of different PARs; (iv) applicability to various antibody formats; and (v) flexibility for the production of heterofunctional antibody-conjugates (e.g. attachment of multiple types of payloads). In this article, we report a single pyridazinedione-based trifunctional dual bridging linker that enables, in a two-step procedure (re-bridging/click), the generation of either mAb-, Fab'-, or Fc-conjugates from native mAb, (Fab')2 or Fc formats, respectively. Fc and (Fab')2 formats were generated via enzymatic digestion of native mAbs. Whilst the same reduction and re-bridging protocols were applied to all three of the protein formats, the subsequent click reaction(s) employed to graft payload(s) drove the generation of a range of PARs, including heterofunctional PARs. As such, exploiting click reactivity and/or orthogonality afforded mAb-conjugates with PARs of 6, 4, 2 or 4 + 2, and Fab'- and Fc-conjugates with a PAR of 3, 2, 1 or 2 + 1 on-demand. We believe that the homogeneity, novelty and variety in accessible PARs, as well as the applicability to various antibody-conjugate formats enabled by our non-recombinant method could be a suitable tool for antibody-drug conjugates optimisation (optimal PAR value, optimal payloads combination) and boost the development of new antibody therapeutics (Fab'- and Fc-conjugates).
Collapse
Affiliation(s)
- Fabien Thoreau
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Léa N C Rochet
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - James R Baker
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Vijay Chudasama
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| |
Collapse
|
14
|
Thoreau F, Szijj PA, Greene MK, Rochet LNC, Thanasi IA, Blayney JK, Maruani A, Baker JR, Scott CJ, Chudasama V. Modular Chemical Construction of IgG-like Mono- and Bispecific Synthetic Antibodies (SynAbs). ACS CENTRAL SCIENCE 2023; 9:476-487. [PMID: 36968530 PMCID: PMC10037451 DOI: 10.1021/acscentsci.2c01437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Indexed: 06/18/2023]
Abstract
In recent years there has been rising interest in the field of protein-protein conjugation, especially related to bispecific antibodies (bsAbs) and their therapeutic applications. These constructs contain two paratopes capable of binding two distinct epitopes on target molecules and are thus able to perform complex biological functions (mechanisms of action) not available to monospecific mAbs. Traditionally these bsAbs have been constructed through protein engineering, but recently chemical methods for their construction have started to (re)emerge. While these have been shown to offer increased modularity, speed, and for some methods even the inherent capacity for further functionalization (e.g., with small molecule cargo), most of these approaches lacked the ability to include a fragment crystallizable (Fc) modality. The Fc component of IgG antibodies offers effector function and increased half-life. Here we report a first-in-class disulfide rebridging and click-chemistry-based method for the generation of Fc-containing, IgG-like mono- and bispecific antibodies. These are in the FcZ-(FabX)-FabY format, i.e., two distinct Fabs and an Fc, potentially all from different antibodies, attached in a homogeneous and covalent manner. We have dubbed these molecules synthetic antibodies (SynAbs). We have constructed a T cell-engager (TCE) SynAb, FcCD20-(FabHER2)-FabCD3, and have confirmed that it exhibits the expected biological functions, including the ability to kill HER2+ target cells in a coculture assay with T cells.
Collapse
Affiliation(s)
- Fabien Thoreau
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - Peter A. Szijj
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - Michelle K. Greene
- Patrick
G Johnston Centre for Cancer Research, School of Medicine, Dentistry
and Biomedical Sciences, Queen’s
University Belfast, Belfast BT9 7AEU.K.
| | - Léa N. C. Rochet
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - Ioanna A. Thanasi
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - Jaine K. Blayney
- Patrick
G Johnston Centre for Cancer Research, School of Medicine, Dentistry
and Biomedical Sciences, Queen’s
University Belfast, Belfast BT9 7AEU.K.
| | - Antoine Maruani
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - James R. Baker
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - Christopher J. Scott
- Patrick
G Johnston Centre for Cancer Research, School of Medicine, Dentistry
and Biomedical Sciences, Queen’s
University Belfast, Belfast BT9 7AEU.K.
| | - Vijay Chudasama
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| |
Collapse
|
15
|
Chrzastek A, Thanasi IA, Irving JA, Chudasama V, Baker JR. Dual reactivity disulfide bridging reagents; enabling new approaches to antibody fragment bioconjugation. Chem Sci 2022; 13:11533-11539. [PMID: 36320392 PMCID: PMC9555722 DOI: 10.1039/d2sc04531a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 09/16/2022] [Indexed: 01/27/2024] Open
Abstract
Disulfide bridging, also known as disulfide stapling, is a powerful strategy for the construction of site-selective protein bioconjugates. Here we describe the first examples of a new class of such reagents, containing a 'stable-labile' design. These dual-reactive reagents are designed to form a stable bond to one cysteine and a labile bond to the second; resulting in a robust attachment to the protein with one end of the bridge, whilst the other end serves as a reactive handle for subsequent bioconjugation. By incorporating thioesters into these bridges, we demonstrate that they are primed for native chemical ligation (NCL) with N-terminal cysteines; offering an alternative to the requirement for C-terminal thioesters for use in such ligations. Alternatively, the use of hydrazine as the ligating nucleophile enables a separate cargo to be attached to each cysteine residue, which are exploited to insert variably cleavable linkers. These methodologies are demonstrated on an antibody fragment, and serve to expand the scope of disulfide bridging strategies whilst offering a convenient route to the construction of multifunctional antibody fragment conjugates.
Collapse
Affiliation(s)
- Alina Chrzastek
- Department of Chemistry, University College London 20 Gordon Street WC1H OAJ London UK
| | - Ioanna A Thanasi
- Department of Chemistry, University College London 20 Gordon Street WC1H OAJ London UK
| | - James A Irving
- UCL Respiratory, Rayne Institute, University College London WC1E 6JF London UK
| | - Vijay Chudasama
- Department of Chemistry, University College London 20 Gordon Street WC1H OAJ London UK
| | - James R Baker
- Department of Chemistry, University College London 20 Gordon Street WC1H OAJ London UK
| |
Collapse
|
16
|
Matsushita T, Maruyama N, Koyama T, Hatano K, Matsuoka K. Modification of Fab Fragments by Dibromopyridazinediones Carrying Mono- and Double-Biotin Functionalities. ACS OMEGA 2022; 7:34554-34562. [PMID: 36188280 PMCID: PMC9520716 DOI: 10.1021/acsomega.2c04379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/02/2022] [Indexed: 06/16/2023]
Abstract
To verify the potencies of dibromopyridazinediones with mono- and double-biotin groups, the functions as cysteine-selective biotinylation reagents were evaluated through conjugation with a goat anti-mouse IgG Fab fragment as a functional protein model. The starting Fab was reduced with tris(2-carboxyethyl)phosphine to cleave the disulfide bond and then treated with the reagents. These reagents simultaneously introduced biotin groups into the reduced Fab and re-bridged the disulfide moiety. Furthermore, we demonstrated that the biotin-labeled Fabs were reactive to an antigen and streptavidin.
Collapse
Affiliation(s)
- Takahiko Matsushita
- Area
for Molecular Function, Division of Material Science, Graduate School
of Science and Engineering, Saitama University, Sakura, Saitama 338-8570, Japan
- Medical
Innovation Research Unit (MiU), Advanced Institute of Innovative Technology
(AIIT), Saitama University, Sakura, Saitama 338-8570, Japan
- Health
Sciences and Technology Research Area, Strategic Research Center, Saitama University, Sakura, Saitama 338-8570, Japan
| | - Naoto Maruyama
- Area
for Molecular Function, Division of Material Science, Graduate School
of Science and Engineering, Saitama University, Sakura, Saitama 338-8570, Japan
| | - Tetsuo Koyama
- Area
for Molecular Function, Division of Material Science, Graduate School
of Science and Engineering, Saitama University, Sakura, Saitama 338-8570, Japan
| | - Ken Hatano
- Area
for Molecular Function, Division of Material Science, Graduate School
of Science and Engineering, Saitama University, Sakura, Saitama 338-8570, Japan
- Medical
Innovation Research Unit (MiU), Advanced Institute of Innovative Technology
(AIIT), Saitama University, Sakura, Saitama 338-8570, Japan
- Health
Sciences and Technology Research Area, Strategic Research Center, Saitama University, Sakura, Saitama 338-8570, Japan
| | - Koji Matsuoka
- Area
for Molecular Function, Division of Material Science, Graduate School
of Science and Engineering, Saitama University, Sakura, Saitama 338-8570, Japan
- Medical
Innovation Research Unit (MiU), Advanced Institute of Innovative Technology
(AIIT), Saitama University, Sakura, Saitama 338-8570, Japan
- Health
Sciences and Technology Research Area, Strategic Research Center, Saitama University, Sakura, Saitama 338-8570, Japan
| |
Collapse
|
17
|
Taylor RJ, Geeson MB, Journeaux T, Bernardes GJL. Chemical and Enzymatic Methods for Post-Translational Protein-Protein Conjugation. J Am Chem Soc 2022; 144:14404-14419. [PMID: 35912579 PMCID: PMC9389620 DOI: 10.1021/jacs.2c00129] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Indexed: 11/28/2022]
Abstract
Fusion proteins play an essential role in the biosciences but suffer from several key limitations, including the requirement for N-to-C terminal ligation, incompatibility of constituent domains, incorrect folding, and loss of biological activity. This perspective focuses on chemical and enzymatic approaches for the post-translational generation of well-defined protein-protein conjugates, which overcome some of the limitations faced by traditional fusion techniques. Methods discussed range from chemical modification of nucleophilic canonical amino acid residues to incorporation of unnatural amino acid residues and a range of enzymatic methods, including sortase-mediated ligation. Through summarizing the progress in this rapidly growing field, the key successes and challenges associated with using chemical and enzymatic approaches are highlighted and areas requiring further development are discussed.
Collapse
Affiliation(s)
- Ross J. Taylor
- Department
of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, U.K.
| | - Michael B. Geeson
- Department
of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, U.K.
| | - Toby Journeaux
- Department
of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, U.K.
| | - Gonçalo J. L. Bernardes
- Department
of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, U.K.
- Instituto
de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| |
Collapse
|
18
|
Taylor RJ, Aguilar Rangel M, Geeson MB, Sormanni P, Vendruscolo M, Bernardes GJL. π-Clamp-Mediated Homo- and Heterodimerization of Single-Domain Antibodies via Site-Specific Homobifunctional Conjugation. J Am Chem Soc 2022; 144:13026-13031. [PMID: 35834748 PMCID: PMC9335888 DOI: 10.1021/jacs.2c04747] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Indexed: 01/07/2023]
Abstract
Post-translational protein-protein conjugation produces bioconjugates that are unavailable via genetic fusion approaches. A method for preparing protein-protein conjugates using π-clamp-mediated cysteine arylation with pentafluorophenyl sulfonamide functional groups is described. Two computationally designed antibodies targeting the SARS-CoV-2 receptor binding domain were produced (KD = 146, 581 nM) with a π-clamp sequence near the C-terminus and dimerized using this method to provide a 10-60-fold increase in binding (KD = 8-15 nM). When two solvent-exposed cysteine residues were present on the second protein domain, the π-clamp cysteine residue was selectively modified over an Asp-Cys-Glu cysteine residue, allowing for subsequent small-molecule conjugation. With this strategy, we build molecule-protein-protein conjugates with complete chemical control over the sites of modification.
Collapse
Affiliation(s)
- Ross J. Taylor
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Mauricio Aguilar Rangel
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Michael B. Geeson
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Pietro Sormanni
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Michele Vendruscolo
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Gonçalo J. L. Bernardes
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
- Instituto
de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
19
|
Bahou C, Chudasama V. The use of bromopyridazinedione derivatives in chemical biology. Org Biomol Chem 2022; 20:5879-5890. [PMID: 35373804 DOI: 10.1039/d2ob00310d] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Tools that facilitate the chemical modification of peptides and proteins are gaining an increasing amount of interest across many avenues of chemical biology as they enable a plethora of therapeutic, imaging and diagnostic applications. Cysteine residues and disulfide bonds have been highlighted as appealing targets for modification due to the highly homogenous nature of the products that can be formed through their site-selective modification. Amongst the reagents available for the site-selective modification of cysteine(s)/disulfide(s), pyridazinediones (PDs) have played a particularly important and enabling role. In this review, we outline the unique chemical features that make PDs especially well-suited to cysteine/disulfide modification on a wide variety of proteins and peptides, as well as provide context as to the problems solved (and applications enabled) by this technology.
Collapse
Affiliation(s)
- Calise Bahou
- UCL Department of Chemistry, 20 Gordon Street, London WC1H 0AJ, UK.
| | - Vijay Chudasama
- UCL Department of Chemistry, 20 Gordon Street, London WC1H 0AJ, UK.
| |
Collapse
|
20
|
Thoreau F, Chudasama V. Enabling the next steps in cancer immunotherapy: from antibody-based bispecifics to multispecifics, with an evolving role for bioconjugation chemistry. RSC Chem Biol 2022; 3:140-169. [PMID: 35360884 PMCID: PMC8826860 DOI: 10.1039/d1cb00082a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 10/22/2021] [Indexed: 12/02/2022] Open
Abstract
In the past two decades, immunotherapy has established itself as one of the leading strategies for cancer treatment, as illustrated by the exponentially growing number of related clinical trials. This trend was, in part, prompted by the clinical success of both immune checkpoint modulation and immune cell engagement, to restore and/or stimulate the patient's immune system's ability to fight the disease. These strategies were sustained by progress in bispecific antibody production. However, despite the decisive progress made in the treatment of cancer, toxicity and resistance are still observed in some cases. In this review, we initially provide an overview of the monoclonal and bispecific antibodies developed with the objective of restoring immune system functions to treat cancer (cancer immunotherapy), through immune checkpoint modulation, immune cell engagement or a combination of both. Their production, design strategy and impact on the clinical trial landscape are also addressed. In the second part, the concept of multispecific antibody formats, notably MuTICEMs (Multispecific Targeted Immune Cell Engagers & Modulators), as a possible answer to current immunotherapy limitations is investigated. We believe it could be the next step to take for cancer immunotherapy research and expose why bioconjugation chemistry might play a key role in these future developments.
Collapse
Affiliation(s)
- Fabien Thoreau
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Vijay Chudasama
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| |
Collapse
|
21
|
Spears RJ, McMahon C, Shamsabadi M, Bahou C, Thanasi IA, Rochet LNC, Forte N, Thoreau F, Baker JR, Chudasama V. A novel thiol-labile cysteine protecting group for peptide synthesis based on a pyridazinedione (PD) scaffold. Chem Commun (Camb) 2022; 58:645-648. [PMID: 34747956 DOI: 10.1039/d1cc03802h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Herein we report a thiol-labile cysteine protecting group based on an unsaturated pyridazinedione (PD) scaffold. We establish compatibility of the PD in conventional solid phase peptide synthesis (SPPS), showcasing this in the on-resin synthesis of biologically relevant oxytocin. Furthermore, we establish the applicability of the PD protecting group towards both microwave-assisted SPPS and native chemical ligation (NCL) in a model system.
Collapse
Affiliation(s)
- Richard J Spears
- UCL Department of Chemistry, 20 Gordon Street, London, WC1H 0AJ, UK.
| | - Clíona McMahon
- UCL Department of Chemistry, 20 Gordon Street, London, WC1H 0AJ, UK.
| | - Monika Shamsabadi
- UCL Department of Chemistry, 20 Gordon Street, London, WC1H 0AJ, UK.
| | - Calise Bahou
- UCL Department of Chemistry, 20 Gordon Street, London, WC1H 0AJ, UK.
| | - Ioanna A Thanasi
- UCL Department of Chemistry, 20 Gordon Street, London, WC1H 0AJ, UK.
| | - Léa N C Rochet
- UCL Department of Chemistry, 20 Gordon Street, London, WC1H 0AJ, UK.
| | - Nafsika Forte
- UCL Department of Chemistry, 20 Gordon Street, London, WC1H 0AJ, UK.
| | - Fabien Thoreau
- UCL Department of Chemistry, 20 Gordon Street, London, WC1H 0AJ, UK.
| | - James R Baker
- UCL Department of Chemistry, 20 Gordon Street, London, WC1H 0AJ, UK.
| | - Vijay Chudasama
- UCL Department of Chemistry, 20 Gordon Street, London, WC1H 0AJ, UK.
| |
Collapse
|
22
|
Sornay C, Vaur V, Wagner A, Chaubet G. An overview of chemo- and site-selectivity aspects in the chemical conjugation of proteins. ROYAL SOCIETY OPEN SCIENCE 2022; 9:211563. [PMID: 35116160 PMCID: PMC8790347 DOI: 10.1098/rsos.211563] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/20/2021] [Indexed: 05/03/2023]
Abstract
The bioconjugation of proteins-that is, the creation of a covalent link between a protein and any other molecule-has been studied for decades, partly because of the numerous applications of protein conjugates, but also due to the technical challenge it represents. Indeed, proteins possess inner physico-chemical properties-they are sensitive and polynucleophilic macromolecules-that make them complex substrates in conjugation reactions. This complexity arises from the mild conditions imposed by their sensitivity but also from selectivity issues, viz the precise control of the conjugation site on the protein. After decades of research, strategies and reagents have been developed to address two aspects of this selectivity: chemoselectivity-harnessing the reacting chemical functionality-and site-selectivity-controlling the reacting amino acid residue-most notably thanks to the participation of synthetic chemistry in this effort. This review offers an overview of these chemical bioconjugation strategies, insisting on those employing native proteins as substrates, and shows that the field is active and exciting, especially for synthetic chemists seeking new challenges.
Collapse
Affiliation(s)
- Charlotte Sornay
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, Illkirch-Graffenstaden 67400, France
| | - Valentine Vaur
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, Illkirch-Graffenstaden 67400, France
| | - Alain Wagner
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, Illkirch-Graffenstaden 67400, France
| | - Guilhem Chaubet
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, Illkirch-Graffenstaden 67400, France
| |
Collapse
|
23
|
Dimasi N, Kumar A, Gao C. Generation of bispecific antibodies using chemical conjugation methods. DRUG DISCOVERY TODAY. TECHNOLOGIES 2021; 40:13-24. [PMID: 34916015 DOI: 10.1016/j.ddtec.2021.08.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 08/18/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022]
Abstract
Bispecific antibodies combine the specificity of two antibodies into one molecule. During the past two decades, advancement in protein engineering enabled the development of more than 100 bispecific formats, three of which are approved by the FDA for clinical use. In parallel to protein engineering methods, advancement in conjugation chemistries have spurred the use of chemical engineering approaches to generate bispecific antibodies. Herein, we review selected chemical strategies employed to generate bispecific antibodies that cannot be made using protein engineering methods.
Collapse
Affiliation(s)
- Nazzareno Dimasi
- Antibody Discovery and Protein Engineering, AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Amit Kumar
- Antibody Discovery and Protein Engineering, AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Changshou Gao
- Antibody Discovery and Protein Engineering, AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA.
| |
Collapse
|
24
|
van Hest J, Zheng G, Rotello VM. Bioorthogonal Chemistry and Bioconjugation: Synergistic Tools for Biology and Biomedicine. Bioconjug Chem 2021; 32:1409-1410. [PMID: 34323066 DOI: 10.1021/acs.bioconjchem.1c00355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
25
|
Farleigh M, Pham TT, Yu Z, Kim J, Sunassee K, Firth G, Forte N, Chudasama V, Baker JR, Long NJ, Rivas C, Ma MT. New Bifunctional Chelators Incorporating Dibromomaleimide Groups for Radiolabeling of Antibodies with Positron Emission Tomography Imaging Radioisotopes. Bioconjug Chem 2021; 32:1214-1222. [PMID: 33724798 PMCID: PMC8299457 DOI: 10.1021/acs.bioconjchem.0c00710] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/20/2021] [Indexed: 01/23/2023]
Abstract
Positron Emission Tomography (PET) imaging with antibody-based contrast agents frequently uses the radioisotopes [64Cu]Cu2+ and [89Zr]Zr4+. The macrobicyclic chelator commonly known as sarcophagine (sar) is ideal for labeling receptor-targeted biomolecules with [64Cu]Cu2+. The siderophore chelator, desferrioxamine-B (dfo), has been widely used to incorporate [89Zr]Zr4+ into antibodies. Here, we describe new bifunctional chelators of sar and dfo: these chelators have been functionalized with dibromomaleimides (dbm), that enable site-specific and highly stable attachment of molecular cargoes to reduced, solvent-accessible, interstrand native disulfide groups. The new sar-dbm and dfo-dbm derivatives can be easily conjugated with the IgG antibody trastuzumab via reaction with reduced interstrand disulfide groups to give site-specifically modified dithiomaleamic acid (dtm) conjugates, sar-dtm-trastuzumab and dfo-dtm-trastuzumab, in which interstrand disulfides are rebridged covalently with a small molecule linker. Both sar- and dfo-dtm-trastuzumab conjugates have been radiolabeled with [64Cu]Cu2+ and [89Zr]Zr4+, respectively, in near quantitative radiochemical yield (>99%). Serum stability studies, in vivo PET imaging, and biodistribution analyses using these radiolabeled immunoconjugates demonstrate that both [64Cu]Cu-sar-dtm-trastuzumab and [89Zr]Zr-dfo-dtm-trastuzumab possess high stability in biological milieu. Dibromomaleimide technology can be easily applied to enable stable, site-specific attachment of radiolabeled chelators, such as sar and dfo, to native interstrand disulfide regions of antibodies, enabling tracking of antibodies with PET imaging.
Collapse
Affiliation(s)
- Matthew Farleigh
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Truc Thuy Pham
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Zilin Yu
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Jana Kim
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Kavitha Sunassee
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - George Firth
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Nafsika Forte
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - Vijay Chudasama
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - James R. Baker
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - Nicholas J. Long
- Department
of Chemistry, Imperial College London, Molecular
Sciences Research Hub, London W12 0BZ, U.K.
| | - Charlotte Rivas
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Michelle T. Ma
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| |
Collapse
|
26
|
Richards DA, Thomas MR, Szijj PA, Foote J, Chen Y, Nogueira JCF, Chudasama V, Stevens MM. Employing defined bioconjugates to generate chemically functionalised gold nanoparticles for in vitro diagnostic applications. NANOSCALE 2021; 13:11921-11931. [PMID: 34190286 PMCID: PMC8280965 DOI: 10.1039/d1nr02584h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/22/2021] [Indexed: 06/13/2023]
Abstract
Novel methods for introducing chemical and biological functionality to the surface of gold nanoparticles serve to increase the utility of this class of nanomaterials across a range of applications. To date, methods for functionalising gold surfaces have relied upon uncontrollable non-specific adsorption, bespoke chemical linkers, or non-generalisable protein-protein interactions. Herein we report a versatile method for introducing functionality to gold nanoparticles by exploiting the strong interaction between chemically functionalised bovine serum albumin (f-BSA) and citrate-capped gold nanoparticles (AuNPs). We establish the generalisability of the method by introducing a variety of functionalities to gold nanoparticles using cheap, commercially available chemical linkers. The utility of this approach is further demonstrated through the conjugation of the monoclonal antibody Ontruzant to f-BSA-AuNPs using inverse electron-demand Diels-Alder (iEDDA) click chemistry, a hitherto unexplored chemistry for AuNP-IgG conjugation. Finally, we show that the AuNP-Ontruzant particles generated via f-BSA-AuNPs have a greater affinity for their target in a lateral flow format when compared to conventional physisorption, highlighting the potential of this technology for producing sensitive diagnostic tests.
Collapse
Affiliation(s)
- Daniel A. Richards
- Department of Materials, Imperial College LondonLondonUK
- Department of Bioengineering, Imperial College LondonUK
- Institute of Biomedical Engineering, Imperial College LondonLondonUK
| | - Michael R. Thomas
- Department of Materials, Imperial College LondonLondonUK
- Department of Bioengineering, Imperial College LondonUK
- Institute of Biomedical Engineering, Imperial College LondonLondonUK
- London Centre for Nanotechnology, University College LondonLondonUK
- Department of Biochemical Engineering, University College LondonWC1E 6BT LondonUK
| | - Peter A. Szijj
- Department of Chemistry, University College LondonLondonUK
| | - James Foote
- Department of Materials, Imperial College LondonLondonUK
- Department of Bioengineering, Imperial College LondonUK
- Institute of Biomedical Engineering, Imperial College LondonLondonUK
| | - Yiyun Chen
- Department of Materials, Imperial College LondonLondonUK
- Department of Bioengineering, Imperial College LondonUK
- Institute of Biomedical Engineering, Imperial College LondonLondonUK
| | | | | | - Molly M. Stevens
- Department of Materials, Imperial College LondonLondonUK
- Department of Bioengineering, Imperial College LondonUK
- Institute of Biomedical Engineering, Imperial College LondonLondonUK
- Department of Medical Biochemistry and BiophysicsStockholmSweden
| |
Collapse
|
27
|
Hu Y, Schomaker JM. Recent Developments and Strategies for Mutually Orthogonal Bioorthogonal Reactions. Chembiochem 2021; 22:3254-3262. [PMID: 34261195 DOI: 10.1002/cbic.202100164] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/12/2021] [Indexed: 12/23/2022]
Abstract
Over the past decade, several different metal-free bioorthogonal reactions have been developed to enable simultaneous double-click labeling with minimal-to-no competing cross-reactivities; such transformations are termed 'mutually orthogonal'. More recently, several examples of successful triple ligation strategies have also been described. In this minireview, we discuss selected aspects of the development of orthogonal bioorthogonal reactions over the past decade, including general strategies to drive future innovations to achieve simultaneous, mutually orthogonal click reactions in one pot.
Collapse
Affiliation(s)
- Yun Hu
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI, 53706, USA
| | - Jennifer M Schomaker
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI, 53706, USA
| |
Collapse
|
28
|
Laserna V, Istrate A, Kafuta K, Hakala TA, Knowles TPJ, Alcarazo M, Bernardes GJL. Protein Conjugation by Electrophilic Alkynylation Using 5-(Alkynyl)dibenzothiophenium Triflates. Bioconjug Chem 2021; 32:1570-1575. [PMID: 34232618 DOI: 10.1021/acs.bioconjchem.1c00317] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
5-(Alkynyl)dibenzothiophenium triflates are introduced as new reagents to prepare different protein conjugates through site-selective cysteine alkynylation. The protocol developed allows a highly efficient label of free cysteine-containing proteins with relevant biological roles, such as ubiquitin, the C2A domain of Synaptotagmin-I, or HER2 targeting nanobodies. An electrophilic bis-alkynylating reagent was also designed. The second alkynylating handle thus introduced in the desired protein enables access to protein-thiol, protein-peptide, and protein-protein conjugates, and even diubiquitin dimers can be prepared through this approach. The low excess of reagent needed, mild reaction conditions used, short reaction times, and stability of the S-C(alkyne) bonds at physiological conditions make this approach an interesting addition to the toolbox of classical, site-selective cysteine-conjugation methods.
Collapse
Affiliation(s)
- Victor Laserna
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Alena Istrate
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Kevin Kafuta
- Institut für Organische und Biomolekulare Chemie, Georg-August-Universität Göttingen, Tammannstr. 2, 37077-Göttingen, Germany
| | - Tuuli A Hakala
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Tuomas P J Knowles
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom.,Cavendish Laboratory, Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, United Kingdom
| | - Manuel Alcarazo
- Institut für Organische und Biomolekulare Chemie, Georg-August-Universität Göttingen, Tammannstr. 2, 37077-Göttingen, Germany
| | - Gonçalo J L Bernardes
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
29
|
Xu L, Kuan SL, Weil T. Contemporary Approaches for Site-Selective Dual Functionalization of Proteins. Angew Chem Int Ed Engl 2021; 60:13757-13777. [PMID: 33258535 PMCID: PMC8248073 DOI: 10.1002/anie.202012034] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Indexed: 12/16/2022]
Abstract
Site-selective protein functionalization serves as an invaluable tool for investigating protein structures and functions in complicated cellular environments and accomplishing semi-synthetic protein conjugates such as traceable therapeutics with improved features. Dual functionalization of proteins allows the incorporation of two different types of functionalities at distinct location(s), which greatly expands the features of native proteins. The attachment and crosstalk of a fluorescence donor and an acceptor dye provides fundamental insights into the folding and structural changes of proteins upon ligand binding in their native cellular environments. Moreover, the combination of drug molecules with different modes of action, imaging agents or stabilizing polymers provides new avenues to design precision protein therapeutics in a reproducible and well-characterizable fashion. This review aims to give a timely overview of the recent advancements and a future perspective of this relatively new research area. First, the chemical toolbox for dual functionalization of proteins is discussed and compared. The strengths and limitations of each strategy are summarized in order to enable readers to select the most appropriate method for their envisaged applications. Thereafter, representative applications of these dual-modified protein bioconjugates benefiting from the synergistic/additive properties of the two synthetic moieties are highlighted.
Collapse
Affiliation(s)
- Lujuan Xu
- Max Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Institute of Inorganic Chemistry IUlm UniversityAlbert-Einstein-Allee 1189081UlmGermany
| | - Seah Ling Kuan
- Max Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Institute of Inorganic Chemistry IUlm UniversityAlbert-Einstein-Allee 1189081UlmGermany
| | - Tanja Weil
- Max Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Institute of Inorganic Chemistry IUlm UniversityAlbert-Einstein-Allee 1189081UlmGermany
| |
Collapse
|
30
|
Bahou C, Szijj PA, Spears RJ, Wall A, Javaid F, Sattikar A, Love EA, Baker JR, Chudasama V. A Plug-and-Play Platform for the Formation of Trifunctional Cysteine Bioconjugates that also Offers Control over Thiol Cleavability. Bioconjug Chem 2021; 32:672-679. [PMID: 33710874 PMCID: PMC8154211 DOI: 10.1021/acs.bioconjchem.1c00057] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/04/2021] [Indexed: 02/08/2023]
Abstract
Linkers that enable the site-selective synthesis of chemically modified proteins are of great interest to the field of chemical biology. Homogenous bioconjugates often show advantageous pharmacokinetic profiles and consequently increased efficacy in vivo. Cysteine residues have been exploited as a route to site-selectively modify proteins, and many successfully approved therapeutics make use of cysteine directed conjugation reagents. However, commonly used linkers, including maleimide-thiol conjugates, are not stable to the low concentrations of thiol present in blood. Furthermore, only a few cysteine-targeting reagents enable the site-selective attachment of multiple functionalities: a useful tool in the fields of theranostics and therapeutic blood half-life extension. Herein, we demonstrate the application of the pyridazinedione motif to enable site-selective attachment of three functionalities to a protein bearing a single cysteine residue. Extending upon previously documented dual modification work, here we demonstrate that by exploiting a bromide leaving group as an additional reactive point on the pyridazinedione scaffold, a thiol or aniline derivative can be added to a protein, post-conjugation. Thiol cleavability appraisal of the resultant C-S and C-N linked thio-bioconjugates demonstrated C-S functionalized linkers to be cleavable and C-N functionalized linkers to be noncleavable when incubated in an excess of glutathione. The plug-and-play trifunctional platform was exemplified by attaching clinically relevant motifs: biotin, fluorescein, a polyethylene glycol chain, and a model peptide. This platform provides a rare opportunity to combine up to three functionalities on a protein in a site-selective fashion. Furthermore, by selecting the use of a thiol or an amine for functionalization, we provide unique control over linker cleavability toward thiols, allowing this novel linker to be applied in a range of physiological environments.
Collapse
Affiliation(s)
- Calise Bahou
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Peter A. Szijj
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Richard J. Spears
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Archie Wall
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Faiza Javaid
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Afrah Sattikar
- LifeArc,
Accelerator Building, SBC Open Innovation Campus, SG1 2FX, Stevenage, United Kingdom
| | - Elizabeth A. Love
- LifeArc,
Accelerator Building, SBC Open Innovation Campus, SG1 2FX, Stevenage, United Kingdom
| | - James R. Baker
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Vijay Chudasama
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
- Research
Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| |
Collapse
|
31
|
Xu L, Kuan SL, Weil T. Contemporary Approaches for Site‐Selective Dual Functionalization of Proteins. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202012034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Lujuan Xu
- Max Planck Institute for Polymer Research Ackermannweg 10 55128 Mainz Germany
- Institute of Inorganic Chemistry I Ulm University Albert-Einstein-Allee 11 89081 Ulm Germany
| | - Seah Ling Kuan
- Max Planck Institute for Polymer Research Ackermannweg 10 55128 Mainz Germany
- Institute of Inorganic Chemistry I Ulm University Albert-Einstein-Allee 11 89081 Ulm Germany
| | - Tanja Weil
- Max Planck Institute for Polymer Research Ackermannweg 10 55128 Mainz Germany
- Institute of Inorganic Chemistry I Ulm University Albert-Einstein-Allee 11 89081 Ulm Germany
| |
Collapse
|
32
|
Abstract
Bispecific antibodies (bsAbs) target two different epitopes. These are an up-and-coming class of biologics, with two such therapeutics (emicizumab and blinatumomab) FDA approved and on the market, and many more in clinical trials. While the first reported bsAbs were constructed by chemical methods, this approach has fallen out of favour with the advent of modern genetic engineering techniques and, nowadays, the vast majority of bsAbs are produced by protein engineering. However, in recent years, relying on innovations in the fields of bioconjugation and bioorthogonal click chemistry, new chemical methods have appeared that have the potential to be competitive with protein engineering techniques and, indeed, hold some advantages. These approaches offer modularity, reproducibility and batch-to-batch consistency, as well as the integration of handles, whereby additional cargo molecules can be attached easily, e.g. to generate bispecific antibody-drug conjugates. The linker between the antibodies/antibody fragments can also be easily varied, and new formats (types, defined by structural properties or by construction methodology) can be generated rapidly. These attributes offer the potential to revolutionize the field. Here, we review chemical methods for the generation of bsAbs, showing that the newest examples of these techniques are worthy competitors to the industry-standard expression-based strategies.
Collapse
|
33
|
Park J, Lee S, Kim Y, Yoo TH. Methods to generate site-specific conjugates of antibody and protein. Bioorg Med Chem 2021; 30:115946. [DOI: 10.1016/j.bmc.2020.115946] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
|
34
|
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: 3.4] [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
|