1
|
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
|
2
|
Gulyak EL, Komarova OA, Prokopenko YA, Faizullina EA, Malabuiok DM, Ibragimova AR, Mokrushina YA, Serova OV, Popova GP, Zhitlov MY, Nikitin TD, Brylev VA, Ustinov AV, Alferova VA, Korshun VA, Smirnov IV, Terekhov SS, Sapozhnikova KA. Branched Linkers for Homogeneous Antibody-Drug Conjugates: How Long Is Long Enough? Int J Mol Sci 2024; 25:13356. [PMID: 39769122 PMCID: PMC11678271 DOI: 10.3390/ijms252413356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Homogeneous antibody-drug conjugates (ADCs) exhibit significantly improved pharmacological properties compared to their heterogeneous counterparts. Site-specific conjugation of the payload to the IgG required for homogeneity can be achieved using enzymes. One example is microbial transglutaminase (MTGase), which can selectively perform transamidation on the Q295 residue of human Fc when N297 glycans are removed. As a result, two modifications can be introduced per IgG molecule; however, achieving higher drug-to-antibody ratios (DARs) requires the use of branched linkers. While several such linkers have been reported, little information is available on the relationship between linker structure and ADC properties. To address this gap, we synthesized two branched amino triazide linkers, differing by a PEG4 fragment inserted after the branching point, which were used to prepare two homogeneous trastuzumab-based DAR 6 ADCs (a "short" and a "long" one). This was achieved by a two-step process consisting of enzymatic linker conjugation followed by bioorthogonal coupling with a cleavable linker bearing monomethyl auristatin E (MMAE). Two other trastuzumab-MMAE conjugates were used as controls: a heterogeneous DAR 6 ADC, made using conventional thiol-maleimide chemistry, and a homogeneous DAR 2 ADC. We found that, while the four conjugates had identical affinity for HER2, their cytotoxicity differed significantly: the "long" homogeneous DAR 6 ADC was just as active as its heterogeneous counterpart, but the "short" DAR 6 ADC was an order of magnitude less potent, inferior even to the DAR 2 conjugate. Our findings indicate that the length of the branched linker critically affects the cytotoxic activity of ADCs, possibly due to steric hindrance influencing the rate of linker cleavage by lysosomal enzymes.
Collapse
Affiliation(s)
- Evgeny L. Gulyak
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Olga A. Komarova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
- Higher Chemical College of the Russian Academy of Sciences, D. Mendeleev University of Chemical Technology of Russia, Miusskaya Square 9, 125047 Moscow, Russia
| | - Yury A. Prokopenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Elina A. Faizullina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Diana M. Malabuiok
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Aigul R. Ibragimova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Yuliana A. Mokrushina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Oxana V. Serova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Galina P. Popova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Mikhail Y. Zhitlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Timofei D. Nikitin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Vladimir A. Brylev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Alexey V. Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Vera A. Alferova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Vladimir A. Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Ivan V. Smirnov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Stanislav S. Terekhov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| | - Ksenia A. Sapozhnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (O.A.K.); (Y.A.P.); (E.A.F.); (D.M.M.); (A.R.I.); (Y.A.M.); (O.V.S.); (G.P.P.); (M.Y.Z.); (T.D.N.); (V.A.B.); (A.V.U.); (V.A.A.); (V.A.K.); (I.V.S.); (S.S.T.)
| |
Collapse
|
3
|
Barker JE, Richings GW, Xie Y, Rho JY, Ferguson CTJ, O'Reilly RK, Habershon S. Integrated computational and experimental design of fluorescent heteroatom-functionalised maleimide derivatives. Chem Sci 2024:d4sc04816d. [PMID: 39512926 PMCID: PMC11536195 DOI: 10.1039/d4sc04816d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024] Open
Abstract
The bottom-up design and synthesis of organic molecular species with tailored photophysical properties stands as an important challenge to both computational and experimental chemical science. Overcoming this challenge presents the potential to usher in new tools and approaches to improve our ability to develop new technologies, such as molecular sensors and attuned molecular switches. Here, we report the bottom-up design and characterisation of new fluorescent maleimide derivatives using coupled computational and experimental insights. Using an extensive set of experimentally-measured UV/visible spectra for different functionalized maleimides in different solvents, we train an artificial neural network (ANN) to rapidly correlate maleimide structure (and solvent) with emission spectra characteristics. We subsequently use this computational predictor to identify design principles for novel functionalised maleimide structures with targeted photophysical properties; synthesis and characterisation of several new maleimides demonstrates how this combined strategy can offer new directions for tuning photochemistry, for example opening new routes to designing tailor-made fluorescent probes.
Collapse
Affiliation(s)
- Jake E Barker
- School of Chemistry, The University of Birmingham, University Rd W Birmingham B15 2TT UK
| | | | - Yujie Xie
- School of Medicine, Shanghai University Shanghai P. R. China
| | - Julia Y Rho
- School of Chemistry, The University of Birmingham, University Rd W Birmingham B15 2TT UK
| | - Calum T J Ferguson
- School of Chemistry, The University of Birmingham, University Rd W Birmingham B15 2TT UK
| | - Rachel K O'Reilly
- School of Chemistry, The University of Birmingham, University Rd W Birmingham B15 2TT UK
| | - Scott Habershon
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
| |
Collapse
|
4
|
Swift-Ramirez W, Whalen LA, Thompson LK, Shoemaker KE, Rubio AV, Weiss GA. Catalyst-Free, Three-Component Synthesis of Amidinomaleimides. J Org Chem 2024; 89:13756-13761. [PMID: 39178144 PMCID: PMC11421025 DOI: 10.1021/acs.joc.4c01485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 08/25/2024]
Abstract
Maleimide and amidine functionalities often appear in medicinal and natural product targets. We describe a catalyst-free, three-component coupling reaction for the synthesis of amidinomaleimides. This one-pot reaction fuses a broad range of secondary amines and aldehydes with azidomaleimides. The conditions are mild, simple, modular, high yielding, and amenable to aqueous solvents. Most reaction products can be sufficiently purified without column chromatography. The synthesis creates complex, multifunctional molecules with four different molecules, including a tripeptide, arrayed around an amidinomaleimide core.
Collapse
Affiliation(s)
- Wyatt
R. Swift-Ramirez
- Department
of Chemistry, University of California,
Irvine, 1102 Natural Sciences 2, Irvine, California 92697-2025, United States
| | - Lindsay A. Whalen
- Department
of Chemistry, University of California,
Irvine, 1102 Natural Sciences 2, Irvine, California 92697-2025, United States
| | - Lia K. Thompson
- Department
of Chemistry, University of California,
Irvine, 1102 Natural Sciences 2, Irvine, California 92697-2025, United States
| | - Kaylee E. Shoemaker
- Department
of Chemistry, University of California,
Irvine, 1102 Natural Sciences 2, Irvine, California 92697-2025, United States
| | - Aris V. Rubio
- Department
of Chemistry, University of California,
Irvine, 1102 Natural Sciences 2, Irvine, California 92697-2025, United States
| | - Gregory A. Weiss
- Department
of Chemistry, University of California,
Irvine, 1102 Natural Sciences 2, Irvine, California 92697-2025, United States
- Department
of Molecular Biology and Biochemistry, University
of California, Irvine, 3205 McGaugh Hall, Irvine, California 92697-3900, United States
- Department
of Pharmaceutical Sciences, University of
California, Irvine, 856
Health Sciences Road, Suite 5400, Irvine, California 92697-3958, United States
| |
Collapse
|
5
|
Yap SY, Butcher T, Spears RJ, McMahon C, Thanasi IA, Baker JR, Chudasama V. Chemo- and regio-selective differential modification of native cysteines on an antibody via the use of dehydroalanine forming reagents. Chem Sci 2024; 15:8557-8568. [PMID: 38846383 PMCID: PMC11151841 DOI: 10.1039/d4sc00392f] [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: 01/17/2024] [Accepted: 04/29/2024] [Indexed: 06/09/2024] Open
Abstract
Protein modification has garnered increasing interest over the past few decades and has become an important tool in many aspects of chemical biology. In recent years, much effort has focused on site-selective modification strategies that generate more homogenous bioconjugates, and this is particularly so in the antibody modification space. Modifying native antibodies by targeting solvent-accessible cysteines liberated by interchain disulfide reduction is, perhaps, the predominant strategy for achieving more site-selectivity on an antibody scaffold. This is evidenced by numerous approved antibody therapeutics that have utilised cysteine-directed conjugation reagents and the plethora of methods/strategies focused on antibody cysteine modification. However, all of these methods have a common feature in that after the reduction of native solvent-accessible cystines, the liberated cysteines are all reacted in the same manner. Herein, we report the discovery and application of dehydroalanine forming reagents (including novel reagents) capable of regio- and chemo-selectively modifying these cysteines (differentially) on a clinically relevant antibody fragment and a full antibody. We discovered that these reagents could enable differential reactivity between light chain C-terminal cysteines, heavy chain hinge region cysteines (cysteines with an adjacent proline residue, Cys-Pro), and other heavy chain internal cysteines. This differential reactivity was also showcased on small molecules and on the peptide somatostatin. The application of these dehydroalanine forming reagents was exemplified in the preparation of a dually modified antibody fragment and full antibody. Additionally, we discovered that readily available amide coupling agents can be repurposed as dehydroalanine forming reagents, which could be of interest to the broader field of chemical biology.
Collapse
Affiliation(s)
- Steven Y Yap
- 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
| | - Richard J Spears
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Clíona McMahon
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Ioanna A Thanasi
- 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
|
6
|
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
|
7
|
Ahad A, K. Saeed H, del Solar V, López-Hernández JE, Michel A, Mathew J, Lewis JS, Contel M. Shifting the Antibody-Drug Conjugate Paradigm: A Trastuzumab-Gold-Based Conjugate Demonstrates High Efficacy against Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer Mouse Model. ACS Pharmacol Transl Sci 2023; 6:1972-1986. [PMID: 38093840 PMCID: PMC10714425 DOI: 10.1021/acsptsci.3c00270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 01/17/2024]
Abstract
Antibody-drug conjugates (ADCs) combine the selectivity of monoclonal antibodies (mAbs) with the efficacy of chemotherapeutics to target cancers without toxicity to normal tissue. Clinically, most chemotherapeutic ADCs are based on complex organic molecules, while the conjugation of metallodrugs to mAbs has been overlooked, despite the resurgent interest in metal-based drugs as cancer chemotherapeutics. In 2019, we described the first gold ADCs containing gold-triphenylphosphane fragments as a proof of concept. The ADCs (based on the antibody trastuzumab) were selective and highly active against HER2-positive breast cancer cells. In this study, we developed site-specific ADCs (Thio-1b and Thio-2b) using the cysteine-engineered trastuzumab derivative THIOMAB antibody technology with gold(I)-containing phosphanes and a maleimide-based linker amenable to bioconjugation (1b and 2b). In addition, we developed lysine-directed ADCs with gold payloads based on phosphanes and N-heterocyclic carbenes featuring an activated ester moiety (2c and 5c) with trastuzumab (Tras-2c and Tras-5c) and another anti-HER2 antibody, pertuzumab (Per-2c and Per-5c). Both sets of ADCs demonstrated significant anticancer potency in vitro assays. Based on these results, one ADC (Tras-2c), containing the [Au(PEt3)] fragment present in FDA-approved auranofin, was selected for an in vivo antitumor efficacy study. Immunocompromised mice xenografted with the HER2-positive human cancer cell line SKBR-3 exhibited almost complete tumor reduction and low toxicity with intravenous administration of Tras-2c. With this highly selective targeting system, we demonstrated that a subnanomolar cytotoxicity profile in cells is not required for an impressive antitumor effect in a mouse xenograft model.
Collapse
Affiliation(s)
- Afruja Ahad
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biology
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
| | - Hiwa K. Saeed
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
| | - Virginia del Solar
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
| | - Javier E. López-Hernández
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biochemistry
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
| | - Alexa Michel
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
| | - Joshua Mathew
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
| | - Jason S. Lewis
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
- Molecular
Pharmacology Program, Memorial Sloan Kettering
Cancer Center, New York, New York 10065, United States
- Radiochemistry
and Molecular Imaging Probes Core, Memorial
Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Maria Contel
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biology
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Chemistry
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Biochemistry
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
| |
Collapse
|
8
|
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
|
9
|
Xu T, Zhang F, Chen D, Sun L, Tomazela D, Fayadat-Dilman L. Interrogating heterogeneity of cysteine-engineered antibody-drug conjugates and antibody-oligonucleotide conjugates by capillary zone electrophoresis-mass spectrometry. MAbs 2023; 15:2229102. [PMID: 37381585 DOI: 10.1080/19420862.2023.2229102] [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: 11/29/2022] [Revised: 06/11/2023] [Accepted: 06/20/2023] [Indexed: 06/30/2023] Open
Abstract
Production of site-specific cysteine-engineered antibody-drug conjugates (ADCs) in mammalian cells may produce developability challenges, fragments, and heterogenous molecules, leading to potential product critical quality attributes in later development stages. Liquid phase chromatography with mass spectrometry (LC-MS) is widely used to evaluate antibody impurities and drug-to-antibody ratio, but faces challenges in analysis of fragment product variants of cysteine-engineered ADCs and oligonucleotide-to-antibody ratio (OAR) species of antibody-oligonucleotide conjugates (AOCs). Here, for the first time, we report novel capillary zone electrophoresis (CZE)-MS approaches to address the challenges above. CZE analysis of six ADCs made with different parent monoclonal antibodies (mAbs) and small molecule drug-linker payloads revealed that various fragment impurities, such as half mAbs with one/two drugs, light chains with one/two drugs, light chains with C-terminal cysteine truncation, heavy chain clippings, were well resolved from the main species. However, most of these fragments were coeluted or had signal suppression during LC-MS analysis. Furthermore, the method was optimized on both ionization and separation aspects to enable the characterization of two AOCs. The method successfully achieved baseline separation and accurate quantification of their OAR species, which were also highly challenging using conventional LC-MS methods. Finally, we compared the migration time and CZE separation profiles among ADCs and their parent mAbs, and found that properties of mAbs and linker payloads significantly influenced the separation of product variants by altering their size or charge. Our study showcases the good performance and broad applicability of CZE-MS techniques for monitoring the heterogeneity of cysteine-engineered ADCs and AOCs.
Collapse
Affiliation(s)
- Tian Xu
- Department of Chemistry Michigan State University, East Lansing MI 48824 USA
| | - Fan Zhang
- Discovery Biologics, Protein Sciences, Merck & Co., Inc, South San Francisco, CA 94080 USA
| | - Daoyang Chen
- Discovery Biologics, Protein Sciences, Merck & Co., Inc, South San Francisco, CA 94080 USA
| | - Liangliang Sun
- Department of Chemistry Michigan State University, East Lansing MI 48824 USA
| | - Daniela Tomazela
- Discovery Biologics, Protein Sciences, Merck & Co., Inc, South San Francisco, CA 94080 USA
| | | |
Collapse
|
10
|
Corogeanu D, Zaki K, Beavil AJ, Arnold JN, Diebold SS. Antibody conjugates for targeted delivery of Toll-like receptor 9 agonist to the tumor tissue. PLoS One 2023; 18:e0282831. [PMID: 36913398 PMCID: PMC10010539 DOI: 10.1371/journal.pone.0282831] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 02/23/2023] [Indexed: 03/14/2023] Open
Abstract
Imiquimod, a Toll-like receptor 7 (TLR7) agonist is routinely used for topical administration in basal cell carcinoma and stage zero melanoma. Similarly, the TLR agonist Bacillus Calmette-Guérin is used for the local treatment of bladder cancer and clinical trials showed treatment efficacy of intratumoral injections with TLR9 agonists. However, when administered systemically, endosomal TLR agonists cause adverse responses due to broad immune activation. Hence, strategies for targeted delivery of TLR agonists to the tumor tissue are needed to enable the widespread use of endosomal TLR agonists in the context of tumor immunotherapy. One strategy for targeted delivery of TLR agonist is their conjugation to tumor antigen-specific therapeutic antibodies. Such antibody-TLR agonist conjugates act synergistically by inducing local TLR-mediated innate immune activation which complements the anti-tumor immune mechanisms induced by the therapeutic antibody. In this study, we explored different conjugation strategies for TLR9 agonists to immunoglobulin G (IgG). We evaluated biochemical conjugation of immunostimulatory CpG oligodesoxyribonucleotides (ODN) to the HER2-specific therapeutic antibody Trastuzumab with different cross-linkers comparing stochastic with site-specific conjugation. The physiochemical make-up and biological activities of the generated Trastuzumab-ODN conjugates were characterized in vitro and demonstrated that site-specific conjugation of CpG ODN is crucial for maintaining the antigen-binding capabilities of Trastuzumab. Furthermore, site-specific conjugate was effective in promoting anti-tumor immune responses in vivo in a pseudo-metastasis mouse model with engineered human HER2-transgenic tumor cells. In this in vivo model, co-delivery of Trastuzumab and CpG ODN in form of site-specific conjugates was superior to co-injection of unconjugated Trastuzumab, CpG ODN or stochastic conjugate in promoting T cell activation and expansion. Thereby, this study highlights that site-specific conjugation of CpG ODN to therapeutic antibodies targeting tumor markers is a feasible and more reliable approach for generation of conjugates which retain and combine the functional properties of the adjuvant and the antibody.
Collapse
Affiliation(s)
- Diana Corogeanu
- National Institute for Biological Standards and Control (NIBSC), Biotherapeutics Division, Medicines and Healthcare products Regulatory Agency, Potters Bar, United Kingdom
| | - Kam Zaki
- National Institute for Biological Standards and Control (NIBSC), Advanced Therapies Division, Medicines and Healthcare products Regulatory Agency, Potters Bar, United Kingdom
| | - Andrew J Beavil
- King's College London, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, Guy's Hospital, London, United Kingdom
| | - James N Arnold
- King's College London, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, Guy's Hospital, London, United Kingdom
| | - Sandra S Diebold
- National Institute for Biological Standards and Control (NIBSC), Biotherapeutics Division, Medicines and Healthcare products Regulatory Agency, Potters Bar, United Kingdom
| |
Collapse
|
11
|
Hansen RA, Märcher A, Gothelf KV. One-Step Conversion of NHS Esters to Reagents for Site-Directed Labeling of IgG Antibodies. Bioconjug Chem 2022; 33:1811-1817. [PMID: 36202104 DOI: 10.1021/acs.bioconjchem.2c00392] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Antibody conjugates are extensively used for diagnostics and therapeutics, and as a tool for molecular biology. To prepare such conjugates N-hydroxysuccinimide (NHS) esters are most often used due to the straightforward experimental procedure and the commercial accessibility of the reagents. Such conjugates are however highly heterogeneous, since only the reactivity of the lysines determines the distribution of labels. This has inspired the development of methods that experimentally are as facile but produce conjugates of higher quality. Herein, we report the development of a reagent that can, in one step, be activated with an NHS ester of choice and subsequently can be directly used for site-directed labeling of antibodies. The reagent can be prepared in three synthetic steps and produces conjugates with similar ease as for NHS esters, however in a site-directed manner. We show that the reagent is quantitatively activated by a variety of NHS esters, and we use these to functionalize IgG1, IgG2, and IgG4 antibodies.
Collapse
Affiliation(s)
- Rikke A Hansen
- Department of Chemistry and Interdisciplinary Nanoscience center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus, Denmark
| | - Anders Märcher
- Department of Chemistry and Interdisciplinary Nanoscience center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus, Denmark
| | - Kurt V Gothelf
- Department of Chemistry and Interdisciplinary Nanoscience center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus, Denmark
| |
Collapse
|
12
|
Pander G, Uhl P, Kühl N, Haberkorn U, Anderl J, Mier W. Antibody-drug conjugates: What drives their progress? Drug Discov Today 2022; 27:103311. [PMID: 35787480 DOI: 10.1016/j.drudis.2022.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/01/2022] [Accepted: 06/29/2022] [Indexed: 12/15/2022]
Abstract
Antibody-drug conjugates (ADCs) are on the brink of widespread use for the targeted treatment of cancer. ADCs manage the toxicity of drugs with unacceptable narrow therapeutic windows by guiding highly toxic compounds to the target cells, thereby sparing healthy cells. In this review, we describe approved ADCs and discuss their modes of action, together with medicinal chemical aspects, to evaluate the potential for improvement and to combat tumor-acquired resistance. A recent research focus has centered on the stimulation of immune responses to induce immunogenic cell death and the influence on the tumor microenvironment to enhance bystander effects.
Collapse
Affiliation(s)
- Giulia Pander
- Heidelberg University Hospital, Department of Nuclear Medicine, INF 400, 69120 Heidelberg, Germany
| | - Philipp Uhl
- Heidelberg University Hospital, Department of Nuclear Medicine, INF 400, 69120 Heidelberg, Germany
| | - Nikos Kühl
- Heidelberg University, Institute of Pharmacy and Molecular Biotechnology, INF 364, 69120 Heidelberg, Germany
| | - Uwe Haberkorn
- Heidelberg University Hospital, Department of Nuclear Medicine, INF 400, 69120 Heidelberg, Germany
| | - Jan Anderl
- Merck KGaA, Antibody Drug Conjugates & Targeted NBE Therapeutics, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Walter Mier
- Heidelberg University Hospital, Department of Nuclear Medicine, INF 400, 69120 Heidelberg, Germany.
| |
Collapse
|
13
|
Postupalenko V, Marx L, Viertl D, Gsponer N, Gasilova N, Denoel T, Schaefer N, Prior JO, Hagens G, Lévy F, Garrouste P, Segura JM, Nyanguile O. Template directed synthesis of antibody Fc conjugates with concomitant ligand release. Chem Sci 2022; 13:3965-3976. [PMID: 35440989 PMCID: PMC8985508 DOI: 10.1039/d1sc06182h] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 02/16/2022] [Indexed: 11/29/2022] Open
Abstract
Antibodies are an attractive therapeutic modality for cancer treatment as they allow the increase of the treatment response rate and avoid the severe side effects of chemotherapy. Notwithstanding the strong benefit of antibodies, the efficacy of anti-cancer antibodies can dramatically vary among patients and ultimately result in no response to the treatment. Here, we have developed a novel means to regioselectively label the Fc domain of any therapeutic antibody with a radionuclide chelator in a single step chemistry, with the aim to study by SPECT/CT imaging if the radiolabeled antibody is capable of targeting cancer cells in vivo. A Fc-III peptide was used as bait to bring a carbonate electrophilic site linked to a metal chelator and to a carboxyphenyl leaving group in close proximity with an antibody Fc nucleophile amino acid (K317), thereby triggering the covalent linkage of the chelator to the antibody lysine, with the concomitant release of the carboxyphenyl Fc-III ligand. Using CHX-A''-DTPA, we radiolabeled trastuzumab with indium-111 and showed in biodistribution and imaging experiments that the antibody accumulated successfully in the SK-OV-3 xenograft tumour implanted in mice. We found that our methodology leads to homogeneous conjugation of CHX-A''-DTPA to the antibody, and confirmed that the Fc domain can be selectively labeled at K317, with a minor level of unspecific labeling on the Fab domain. The present method can be developed as a clinical diagnostic tool to predict the success of the therapy. Furthermore, our Fc-III one step chemistry concept paves the way to a broad array of other applications in antibody bioengineering.
Collapse
Affiliation(s)
- Viktoriia Postupalenko
- Institute of Life Technologies, HES-SO Valais-Wallis Rue de l'Industrie 23 CH-1950 Sion Switzerland
| | - Léo Marx
- Debiopharm Research & Manufacturing SA, Campus "après-demain" Rue du Levant 146 1920 Martigny Switzerland
| | - David Viertl
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital CH-1011 Lausanne Switzerland
- In Vivo Imaging Facility, Department of Research and Training, University of Lausanne CH-1011 Lausanne Switzerland
| | - Nadège Gsponer
- Institute of Life Technologies, HES-SO Valais-Wallis Rue de l'Industrie 23 CH-1950 Sion Switzerland
| | - Natalia Gasilova
- EPFL Valais Wallis, MSEAP, ISIC-GE-VS rue de l'Industrie 17 1951 Sion Switzerland
| | - Thibaut Denoel
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital CH-1011 Lausanne Switzerland
| | - Niklaus Schaefer
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital CH-1011 Lausanne Switzerland
| | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital CH-1011 Lausanne Switzerland
| | - Gerrit Hagens
- Institute of Life Technologies, HES-SO Valais-Wallis Rue de l'Industrie 23 CH-1950 Sion Switzerland
| | - Frédéric Lévy
- Debiopharm International SA, Forum "après-demain" Chemin Messidor 5-7 Case postale 5911 1002 Lausanne Switzerland
| | - Patrick Garrouste
- Debiopharm Research & Manufacturing SA, Campus "après-demain" Rue du Levant 146 1920 Martigny Switzerland
| | - Jean-Manuel Segura
- Institute of Life Technologies, HES-SO Valais-Wallis Rue de l'Industrie 23 CH-1950 Sion Switzerland
| | - Origène Nyanguile
- Institute of Life Technologies, HES-SO Valais-Wallis Rue de l'Industrie 23 CH-1950 Sion Switzerland
| |
Collapse
|
14
|
Singh K, Canakci M, Kanjilal P, Williams N, Shanthalingam S, Osborne BA, Thayumanavan S. Evaluation of Cellular Targeting by Fab' vs Full-Length Antibodies in Antibody-Nanoparticle Conjugates (ANCs) Using CD4 T-cells. Bioconjug Chem 2022; 33:486-495. [PMID: 35139308 PMCID: PMC9254259 DOI: 10.1021/acs.bioconjchem.2c00024] [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] [Indexed: 11/28/2022]
Abstract
Targeted delivery of chemotherapeutic drugs can improve their therapeutic efficiency by localizing their toxic effects at the diseased site. This is often achieved either by direct conjugation of drugs to antibodies targeting overexpressed receptors on cancer cells (antibody-drug conjugates/ADCs) or by conjugating antibodies to nanoparticles bearing drugs (antibody-nanoparticle conjugates/ANCs). Here, we report a platform for utilizing hinge cysteines on antigen-binding fragment (Fab') of an anti-CD4 antibody for site-specific conjugation to nanoparticles giving rise to anti-CD4 Fab'-nanoparticle conjugates (Fab'-NCs). We demonstrate a convenient route for obtaining functional anti-CD4 Fab' from full-length antibody and examine the targeted delivery efficiencies of anti-CD4 Fab'-NCs vs ANCs for selective delivery to CD4high mT-ALL cells. Our results indicate that higher avidity of full-length anti-CD4 antibody, i.e., protein alone translated to higher binding ability to CD4high mT-ALL cells in comparison with anti-CD4 Fab' alone. However, the targeted delivery efficiency of anti-CD4 Fab'-NCs was comparable to ANCs indicating that the avidity of Fab' is restored in a nanoparticle-conjugate format. Fab'-NCs are equally capable of achieving targeted drug delivery to CD4high T-cells as ANCs and are a versatile alternative to ANCs by offering site-selective modification strategy while retaining their advantages.
Collapse
Affiliation(s)
- Khushboo Singh
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences University of Massachusetts, Amherst, Amherst, Massachusetts 01003, United States
| | - Mine Canakci
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Pintu Kanjilal
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences University of Massachusetts, Amherst, Amherst, Massachusetts 01003, United States
| | - Natalie Williams
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Sudarvili Shanthalingam
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Barbara A Osborne
- Center for Bioactive Delivery, Institute for Applied Life Sciences University of Massachusetts, Amherst, Amherst, Massachusetts 01003, United States
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences University of Massachusetts, Amherst, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| |
Collapse
|
15
|
Malde R, Parkes MA, Staniforth M, Woolley JM, Stavros VG, Chudasama V, Fielding HH, Baker JR. Intramolecular thiomaleimide [2 + 2] photocycloadditions: stereoselective control for disulfide stapling and observation of excited state intermediates by transient absorption spectroscopy. Chem Sci 2022; 13:2909-2918. [PMID: 35382459 PMCID: PMC8905992 DOI: 10.1039/d1sc06804k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/16/2022] [Indexed: 11/21/2022] Open
Abstract
Thiomaleimides undergo efficient intermolecular [2 + 2] photocycloaddition reactions and offer applications from photochemical peptide stapling to polymer crosslinking; however, the reactions are limited to the formation of the exo head-to-head isomers. Herein, we present an intramolecular variation which completely reverses the stereochemical outcome of this photoreaction, quantitatively generating endo adducts which minimise the structural disturbance of the disulfide staple and afford a 10-fold increase in quantum yield. We demonstrate the application of this reaction on a protein scaffold, using light to confer thiol stability to an antibody fragment conjugate. To understand more about this intriguing class of [2 + 2] photocycloadditions, we have used transient absorption spectroscopy (electronic and vibrational) to study the excited states involved. The initially formed S2 (π1π*) excited state is observed to decay to the S1 (n1π*) state before intersystem crossing to a triplet state. An accelerated intramolecular C-C bond formation provides evidence to explain the increased efficiency of the reaction, and the impact of the various excited states on the carbonyl vibrational modes is discussed.
Collapse
Affiliation(s)
- Roshni Malde
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Michael A Parkes
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Michael Staniforth
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Jack M Woolley
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Vasilios G Stavros
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Vijay Chudasama
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Helen H Fielding
- 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
| |
Collapse
|
16
|
Kang MS, Kong TWS, Khoo JYX, Loh TP. Recent developments in chemical conjugation strategies targeting native amino acids in proteins and their applications in antibody-drug conjugates. Chem Sci 2021; 12:13613-13647. [PMID: 34760149 PMCID: PMC8549674 DOI: 10.1039/d1sc02973h] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/27/2021] [Indexed: 12/15/2022] Open
Abstract
Many fields in chemical biology and synthetic biology require effective bioconjugation methods to achieve their desired functions and activities. Among such biomolecule conjugates, antibody-drug conjugates (ADCs) need a linker that provides a stable linkage between cytotoxic drugs and antibodies, whilst conjugating in a biologically benign, fast and selective fashion. This review focuses on how the development of novel organic synthesis can solve the problems of traditional linker technology. The review shall introduce and analyse the current developments in the modification of native amino acids on peptides or proteins and their applicability to ADC linker. Thereafter, the review shall discuss in detail each endogenous amino acid's intrinsic reactivity and selectivity aspects, and address the research effort to construct an ADC using each conjugation method.
Collapse
Affiliation(s)
- Min Sun Kang
- Division of Chemistry & Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University 21 Nanyang Link 637371 Singapore
| | - Theresa Wai See Kong
- Division of Chemistry & Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University 21 Nanyang Link 637371 Singapore
| | - Joycelyn Yi Xin Khoo
- Division of Chemistry & Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University 21 Nanyang Link 637371 Singapore
| | - Teck-Peng Loh
- Division of Chemistry & Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University 21 Nanyang Link 637371 Singapore
| |
Collapse
|
17
|
Laserna V, Abegg D, Afonso CF, Martin EM, Adibekian A, Ravn P, Corzana F, Bernardes GJL. Dichloro Butenediamides as Irreversible Site‐Selective Protein Conjugation Reagent. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202108791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Victor Laserna
- Yusuf Hamied Department of Chemistry University of Cambridge Lensfield Road CB2 1EW Cambridge UK
| | - Daniel Abegg
- Department of Chemistry The Scripps Research Institute 130 Scripps Way Jupiter Fl 33458 USA
| | - Cláudia F. Afonso
- Instituto de Medicina Molecular João Lobo Antunes Faculdade de Medicina Universidade de Lisboa Avenida Professor Egas Moniz 1649-028 Lisboa Portugal
| | - Esther M. Martin
- AstraZeneca R&D BioPharmaceuticals Unit
- Antibody Discovery & Protein Engineering (ADPE), Milstein Building Granta Park Cambridge CB21 6GH UK
| | - Alexander Adibekian
- Department of Chemistry The Scripps Research Institute 130 Scripps Way Jupiter Fl 33458 USA
| | - Peter Ravn
- AstraZeneca R&D BioPharmaceuticals Unit
- Antibody Discovery & Protein Engineering (ADPE), Milstein Building Granta Park Cambridge CB21 6GH UK
- Department of Biotherapeutic Discovery H. Lundbeck A/S Ottiliavej 9 2500 Valby Denmark
| | - Francisco Corzana
- Departamento de Química Centro de Investigación en Síntesis Química Universidad de La Rioja 26006 Logroño Spain
| | - Gonçalo J. L. Bernardes
- Yusuf Hamied Department of Chemistry University of Cambridge Lensfield Road CB2 1EW Cambridge UK
- Instituto de Medicina Molecular João Lobo Antunes Faculdade de Medicina Universidade de Lisboa Avenida Professor Egas Moniz 1649-028 Lisboa Portugal
| |
Collapse
|
18
|
Laserna V, Abegg D, Afonso CF, Martin EM, Adibekian A, Ravn P, Corzana F, Bernardes GJL. Dichloro Butenediamides as Irreversible Site-Selective Protein Conjugation Reagent. Angew Chem Int Ed Engl 2021; 60:23750-23755. [PMID: 34472678 PMCID: PMC8596790 DOI: 10.1002/anie.202108791] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/04/2021] [Indexed: 01/21/2023]
Abstract
We describe maleic-acid derivatives as robust cysteine-selective reagents for protein labelling with comparable kinetics and superior stability relative to maleimides. Diamide and amido-ester derivatives proved to be efficient protein-labelling species with a common mechanism in which a spontaneous cyclization occurs upon addition to cysteine. Introduction of chlorine atoms in their structures triggers ring hydrolysis or further conjugation with adjacent residues, which results in conjugates that are completely resistant to retro-Michael reactions in the presence of biological thiols and human plasma. By controlling the microenvironment of the reactive site, we can control selectivity towards the hydrolytic pathway, forming homogeneous conjugates. The method is applicable to several scaffolds and enables conjugation of different payloads. The synthetic accessibility of these reagents and the mild conditions required for fast and complete conjugation together with the superior stability of the conjugates make this strategy an important alternative to maleimides in bioconjugation.
Collapse
Affiliation(s)
- Victor Laserna
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW, Cambridge, UK
| | - Daniel Abegg
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Fl, 33458, USA
| | - Cláudia F Afonso
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Esther M Martin
- AstraZeneca, R&D BioPharmaceuticals Unit
- Antibody Discovery & Protein Engineering (ADPE), Milstein Building, Granta Park, Cambridge, CB21 6GH, UK
| | - Alexander Adibekian
- Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, Jupiter, Fl, 33458, USA
| | - Peter Ravn
- AstraZeneca, R&D BioPharmaceuticals Unit
- Antibody Discovery & Protein Engineering (ADPE), Milstein Building, Granta Park, Cambridge, CB21 6GH, UK.,Department of Biotherapeutic Discovery, H. Lundbeck A/S, Ottiliavej 9, 2500, Valby, Denmark
| | - Francisco Corzana
- Departamento de Química, Centro de Investigación en Síntesis Química, Universidad de La Rioja, 26006, Logroño, Spain
| | - Gonçalo J L Bernardes
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW, Cambridge, UK.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| |
Collapse
|
19
|
Matsuda Y, Mendelsohn BA. Recent Advances in Drug-Antibody Ratio Determination of Antibody-Drug Conjugates. Chem Pharm Bull (Tokyo) 2021; 69:976-983. [PMID: 34602579 DOI: 10.1248/cpb.c21-00258] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Antibody-drug conjugates (ADCs) are biopharmaceuticals produced by chemically linking small molecules (payloads) to antibodies that possess specific affinity for the target cell. The ADCs currently on the commercially market are the result of a stochastic conjugation of highly-potent payloads to multiple sites on the monoclonal antibody, resulting in a heterogeneous drug-antibody ratio (DAR) and drug distribution. The heterogeneity inherent to ADCs not produced site-specifically may not only be detrimental to the quality of the drug but also is less-desirable from the perspective of regulatory science. An ideal method or unified approach used to measure the DAR for ADCs, a critical aspect of their analysis and characterization, has not yet been established in the ADC field and remains an often-challenging issue for bioanalytical chemists. In this review we describe, compare, and evaluate the characteristics of various DAR determination methods for ADCs featuring recently reported technologies. The future landscape of bioconjugate DAR analysis is also discussed.
Collapse
|
20
|
You J, Zhang J, Wang J, Jin M. Cysteine-Based Coupling: Challenges and Solutions. Bioconjug Chem 2021; 32:1525-1534. [PMID: 34105345 DOI: 10.1021/acs.bioconjchem.1c00213] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Antibody-drug conjugates (ADCs) have attracted great attention in recent years in the wake of an accelerated FDA approval rate and several large-scale acquisitions. To date, there are ten ADC drugs on the market and more than 70 in various stages of clinical trials. Yet, due to the complicated nature of ADC molecules, considerations need to cover many aspects for the success of ADCs, including target specificity, linker-payload stability, tumor permeability, and clearance rate. This topical review summarizes and discusses current methods used to increase stability and homogeneity of ADCs of cysteine conjugation. We believe that they will lead to improvement of efficacy and pharmacokinetics (PK) of ADC drugs.
Collapse
Affiliation(s)
- Jianwei You
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.,WuXi Biologics Co, Ltd., Shanghai 200131, China
| | - Juan Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Jun Wang
- WuXi Biologics Co, Ltd., Shanghai 200131, China
| | - Mingzhi Jin
- WuXi Biologics Co, Ltd., Shanghai 200131, China
| |
Collapse
|
21
|
Chigoho DM, Lecocq Q, Awad RM, Breckpot K, Devoogdt N, Keyaerts M, Caveliers V, Xavier C, Bridoux J. Site-Specific Radiolabeling of a Human PD-L1 Nanobody via Maleimide-Cysteine Chemistry. Pharmaceuticals (Basel) 2021; 14:ph14060550. [PMID: 34201323 PMCID: PMC8228271 DOI: 10.3390/ph14060550] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 01/13/2023] Open
Abstract
Immune checkpoint inhibitors targeting the programmed cell death-1 (PD-1) and its ligand PD-L1 have proven to be efficient cancer therapies in a subset of patients. From all the patients with various cancer types, only 20% have a positive response. Being able to distinguish patients that do express PD-1/PD-L1 from patients that do not allows patients to benefit from a more personalized and efficient treatment of tumor lesion(s). Expression of PD-1 and PD-L1 is typically assessed via immunohistochemical detection in a tumor biopsy. However, this method does not take in account the expression heterogeneity within the lesion, nor the possible metastasis. To visualize whole-body PD-L1 expression by PET imaging, we developed a nanobody-based radio-immunotracer targeting PD-L1 site-specifically labeled with gallium-68. The cysteine-tagged nanobody was site-specifically conjugated with a maleimide (mal)-NOTA chelator and radiolabeling was tested at different nanobody concentrations and temperatures. Affinity and specificity of the tracer, referred to as [68Ga]Ga-NOTA-mal-hPD-L1 Nb, were assayed by surface plasmon resonance and on PD-L1POS or PD-L1NEG 624-MEL cells. Xenografted athymic nude mice bearing 624-MEL PD-L1POS or PD-L1NEG tumors were injected with the tracer and ex vivo biodistribution was performed 1 h 20 min post-injection. Ideal 68Ga-labeling conditions were found at 50 °C for 15 min. [68Ga]Ga-NOTA-mal-hPD-L1 Nb was obtained in 80 ± 5% DC-RCY with a RCP > 99%, and was stable in injection buffer and human serum up to 3 h (>99% RCP). The in vitro characterization showed that the NOTA-functionalized Nb retained its affinity and specificity. Ex vivo biodistribution revealed a tracer uptake of 1.86 ± 0.67% IA/g in the positive tumors compared with 0.42 ± 0.04% IA/g in the negative tumors. Low background uptake was measured in the other organs and tissues, except for the kidneys and bladder, due to the expected excretion route of Nbs. The data obtained show that the site-specific 68Ga-labeled NOTA-mal-hPD-L1 Nb is a promising PET radio-immunotracer due to its ease of production, stability and specificity for PD-L1.
Collapse
Affiliation(s)
- Dora Mugoli Chigoho
- In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Medical Imaging Department (MIMA), Vrije Universiteit Brussel, 1090 Brussels, Belgium; (D.M.C.); (N.D.); (M.K.); (V.C.); (C.X.)
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (Q.L.); (R.M.A.); (K.B.)
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (Q.L.); (R.M.A.); (K.B.)
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy (LMCT), Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (Q.L.); (R.M.A.); (K.B.)
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Medical Imaging Department (MIMA), Vrije Universiteit Brussel, 1090 Brussels, Belgium; (D.M.C.); (N.D.); (M.K.); (V.C.); (C.X.)
| | - Marleen Keyaerts
- In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Medical Imaging Department (MIMA), Vrije Universiteit Brussel, 1090 Brussels, Belgium; (D.M.C.); (N.D.); (M.K.); (V.C.); (C.X.)
- Department of Nuclear Medicine, UZ Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Vicky Caveliers
- In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Medical Imaging Department (MIMA), Vrije Universiteit Brussel, 1090 Brussels, Belgium; (D.M.C.); (N.D.); (M.K.); (V.C.); (C.X.)
- Department of Nuclear Medicine, UZ Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Catarina Xavier
- In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Medical Imaging Department (MIMA), Vrije Universiteit Brussel, 1090 Brussels, Belgium; (D.M.C.); (N.D.); (M.K.); (V.C.); (C.X.)
| | - Jessica Bridoux
- In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Medical Imaging Department (MIMA), Vrije Universiteit Brussel, 1090 Brussels, Belgium; (D.M.C.); (N.D.); (M.K.); (V.C.); (C.X.)
- Correspondence: ; Tel.: +32-24774991
| |
Collapse
|
22
|
Boschanski M, Krüger T, Karsten L, Falck G, Alam S, Gerlach M, Müller B, Müller KM, Sewald N, Dierks T. Site-Specific Conjugation Strategy for Dual Antibody-Drug Conjugates Using Aerobic Formylglycine-Generating Enzymes. Bioconjug Chem 2021; 32:1167-1174. [PMID: 34060308 DOI: 10.1021/acs.bioconjchem.1c00246] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Multiple, site-specific protein conjugation is increasingly attractive for the generation of antibody-drug conjugates (ADCs). As it is important to control the number and position of cargoes in an ADC, position-selective generation of reactive sites in the protein of interest is required. Formylglycine (FGly) residues are generated by enzymatic conversion of cysteine residues embedded in a certain amino acid sequence motif with a formylglycine-generating enzyme (FGE). The addition of copper ions increases FGE activity leading to the conversion of cysteines within less readily accepted sequences. With this tuned enzyme activity, it is possible to address two different recognition sequences using two aerobic formylglycine-generating enzymes. We demonstrate an improved and facile strategy for the functionalization of a DARPin (designed ankyrin repeat protein) and the single-chain antibody scFv425-Fc, both directed against the epidermal growth factor receptor (EGFR). The single-chain antibody was conjugated with monomethyl auristatin E (MMAE) and carboxyfluorescein (CF) and successfully tested for receptor binding, internalization, and cytotoxicity in cell culture, respectively.
Collapse
Affiliation(s)
- Mareile Boschanski
- Biochemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Tobias Krüger
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Lennard Karsten
- Cellular and Molecular Biotechnology, Faculty of Technology, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Georg Falck
- Cellular and Molecular Biotechnology, Faculty of Technology, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Sarfaraz Alam
- Biochemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Marcus Gerlach
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | | | - Kristian M Müller
- Cellular and Molecular Biotechnology, Faculty of Technology, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Thomas Dierks
- Biochemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| |
Collapse
|
23
|
Reiber T, Zavoiura O, Dose C, Yushchenko DA. Fluorophore Multimerization as an Efficient Approach towards Bright Protein Labels. European J Org Chem 2021. [DOI: 10.1002/ejoc.202100117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Thorge Reiber
- Department of Chemical Biology Miltenyi Biotec B.V. & Co. KG Friedrich-Ebert Straße 68 51429 Bergisch Gladbach Germany
| | - Oleksandr Zavoiura
- Department of Chemical Biology Miltenyi Biotec B.V. & Co. KG Friedrich-Ebert Straße 68 51429 Bergisch Gladbach Germany
| | - Christian Dose
- Department of Chemical Biology Miltenyi Biotec B.V. & Co. KG Friedrich-Ebert Straße 68 51429 Bergisch Gladbach Germany
| | - Dmytro A. Yushchenko
- Department of Chemical Biology Miltenyi Biotec B.V. & Co. KG Friedrich-Ebert Straße 68 51429 Bergisch Gladbach Germany
- Laboratory of Chemical Biology The Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences Flemingovo namesti 2 16610 Prague 6 Czech Republic
| |
Collapse
|
24
|
Costa AM, Bosch L, Petit E, Vilarrasa J. Computational Study of the Addition of Methanethiol to 40+ Michael Acceptors as a Model for the Bioconjugation of Cysteines. J Org Chem 2021; 86:7107-7118. [PMID: 33914532 PMCID: PMC8631706 DOI: 10.1021/acs.joc.1c00349] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Indexed: 12/17/2022]
Abstract
A long series of Michael acceptors are studied computationally as potential alternatives to the maleimides that are used in most antibody-drug conjugates to link Cys of mAbs with cytotoxic drugs. The products of the reaction of methanethiol (CH3SH/MeSH, as a simple model of Cys) with N-methylated ethynesulfonamide, 2-ethynylpyridinium ion, propynamide, and methyl ethynephosphonamidate (that is, with HC≡C-EWG) are predicted by the M06-2X/6-311+G(d,p) method to be thermodynamically more stable, in relation to their precursors, than that of MeSH with N-methylmaleimide and, in general, with H2C═CH-EWG; calculations with AcCysOMe and tBuSH are also included. However, for the addition of the anion (MeS-), which is the reactive species, the order changes and N-methylated 2-vinylpyridinium ion, 2,3-butadienamide, and maleimide may give more easily the anionic adducts than several activated triple bonds; moreover, the calculated ΔG⧧ values increase following the order HC≡C-SO2NHMe, N-methylmaleimide, HC≡C-PO(OMe)NHMe, and HC≡C-CONHMe. In other words, MeS- is predicted to react more rapidly with maleimides than with ethynephosphonamidates and with propynamides, in agreement with the experimental results. New mechanistic details are disclosed regarding the advantageous use of some amides, especially of ethynesulfonamides, which, however, are more prone to double additions and exchange reactions.
Collapse
Affiliation(s)
- Anna M. Costa
- Organic
Chemistry Section,
Facultat de Química, Universitat
de Barcelona, Diagonal 645, Barcelona 08028, Catalonia, Spain
| | - Lluís Bosch
- Organic
Chemistry Section,
Facultat de Química, Universitat
de Barcelona, Diagonal 645, Barcelona 08028, Catalonia, Spain
| | - Elena Petit
- Organic
Chemistry Section,
Facultat de Química, Universitat
de Barcelona, Diagonal 645, Barcelona 08028, Catalonia, Spain
| | - Jaume Vilarrasa
- Organic
Chemistry Section,
Facultat de Química, Universitat
de Barcelona, Diagonal 645, Barcelona 08028, Catalonia, Spain
| |
Collapse
|
25
|
The Chemistry Behind ADCs. Pharmaceuticals (Basel) 2021; 14:ph14050442. [PMID: 34067144 PMCID: PMC8152005 DOI: 10.3390/ph14050442] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/29/2021] [Accepted: 05/02/2021] [Indexed: 02/07/2023] Open
Abstract
Combining the selective targeting of tumor cells through antigen-directed recognition and potent cell-killing by cytotoxic payloads, antibody-drug conjugates (ADCs) have emerged in recent years as an efficient therapeutic approach for the treatment of various cancers. Besides a number of approved drugs already on the market, there is a formidable follow-up of ADC candidates in clinical development. While selection of the appropriate antibody (A) and drug payload (D) is dictated by the pharmacology of the targeted disease, one has a broader choice of the conjugating linker (C). In the present paper, we review the chemistry of ADCs with a particular emphasis on the medicinal chemistry perspective, focusing on the chemical methods that enable the efficient assembly of the ADC from its three components and the controlled release of the drug payload.
Collapse
|
26
|
BF 3-OEt 2 Catalyzed C3-Alkylation of Indole: Synthesis of Indolylsuccinimidesand Their Cytotoxicity Studies. Molecules 2021; 26:molecules26082202. [PMID: 33920456 PMCID: PMC8069703 DOI: 10.3390/molecules26082202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/27/2021] [Accepted: 04/02/2021] [Indexed: 11/17/2022] Open
Abstract
A simple and efficient BF3-OEt2 promoted C3-alkylation of indole has been developed to obtain3-indolylsuccinimidesfrom commercially available indoles and maleimides, with excellent yields under mild reaction conditions. Furthermore, anti-proliferative activity of these conjugates was evaluated against HT-29 (Colorectal), Hepg2 (Liver) and A549 (Lung) human cancer cell lines. One of the compounds, 3w, having N,N-Dimethylatedindolylsuccinimide is a potent congener amongst the series with IC50 value 0.02 µM and 0.8 µM against HT-29 and Hepg2 cell lines, respectively, and compound 3i was most active amongst the series with IC50 value 1.5 µM against A549 cells. Molecular docking study and mechanism of reaction have briefly beendiscussed. This method is better than previous reports in view of yield and substrate scope including electron deficient indoles.
Collapse
|
27
|
Märcher A, Palmfeldt J, Nisavic M, Gothelf KV. A Reagent for Amine‐Directed Conjugation to IgG1 Antibodies. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202013911] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Anders Märcher
- Department of Chemistry and Interdisciplinary Nanoscience Centre (iNANO) Aarhus University Gustav Wieds Vej 14 8000 Aarhus Denmark
| | - Johan Palmfeldt
- Department of Clinical Medicine Aarhus University Brendstrupgårdsvej 21A 8200 Aarhus N Denmark
| | - Marija Nisavic
- Department of Chemistry and Department of Clinical Medicine Aarhus University Brendstrupgårdsvej 21A 8200 Aarhus N Denmark
| | - Kurt V. Gothelf
- Department of Chemistry and Interdisciplinary Nanoscience Centre (iNANO) Aarhus University Gustav Wieds Vej 14 8000 Aarhus Denmark
| |
Collapse
|
28
|
Märcher A, Palmfeldt J, Nisavic M, Gothelf KV. A Reagent for Amine‐Directed Conjugation to IgG1 Antibodies. Angew Chem Int Ed Engl 2021; 60:6539-6544. [DOI: 10.1002/anie.202013911] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/28/2020] [Indexed: 01/24/2023]
Affiliation(s)
- Anders Märcher
- Department of Chemistry and Interdisciplinary Nanoscience Centre (iNANO) Aarhus University Gustav Wieds Vej 14 8000 Aarhus Denmark
| | - Johan Palmfeldt
- Department of Clinical Medicine Aarhus University Brendstrupgårdsvej 21A 8200 Aarhus N Denmark
| | - Marija Nisavic
- Department of Chemistry and Department of Clinical Medicine Aarhus University Brendstrupgårdsvej 21A 8200 Aarhus N Denmark
| | - Kurt V. Gothelf
- Department of Chemistry and Interdisciplinary Nanoscience Centre (iNANO) Aarhus University Gustav Wieds Vej 14 8000 Aarhus Denmark
| |
Collapse
|
29
|
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: 254] [Impact Index Per Article: 63.5] [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
|
30
|
Richardson MB, Gabriel KN, Garcia JA, Ashby SN, Dyer RP, Kim JK, Lau CJ, Hong J, Le Tourneau RJ, Sen S, Narel DL, Katz BB, Ziller JW, Majumdar S, Collins PG, Weiss GA. Pyrocinchonimides Conjugate to Amine Groups on Proteins via Imide Transfer. Bioconjug Chem 2020; 31:1449-1462. [PMID: 32302483 DOI: 10.1021/acs.bioconjchem.0c00143] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Advances in bioconjugation, the ability to link biomolecules to each other, small molecules, surfaces, and more, can spur the development of advanced materials and therapeutics. We have discovered that pyrocinchonimide, the dimethylated analogue of maleimide, undergoes a surprising transformation with biomolecules. The reaction targets amines and involves an imide transfer, which has not been previously reported for bioconjugation purposes. Despite their similarity to maleimides, pyrocinchonimides do not react with free thiols. Though both lysine residues and the N-termini of proteins can receive the transferred imide, the reaction also exhibits a marked preference for certain amines that cannot solely be ascribed to solvent accessibility. This property is peculiar among amine-targeting reactions and can reduce combinatorial diversity when many available reactive amines are available, such as in the formation of antibody-drug conjugates. Unlike amides, the modification undergoes very slow reversion under high pH conditions. The reaction offers a thermodynamically controlled route to single or multiple modifications of proteins for a wide range of applications.
Collapse
Affiliation(s)
- Mark B Richardson
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Kristin N Gabriel
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Joseph A Garcia
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Shareen N Ashby
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Rebekah P Dyer
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Joshua K Kim
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Calvin J Lau
- Department of Physics & Astronomy, University of California, Irvine, Irvine, California 92697, United States
| | - John Hong
- School of Medicine, University of California, Irvine, Irvine, California 92697, United States
| | - Ryan J Le Tourneau
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Sanjana Sen
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, California 92697, United States
| | - David L Narel
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Benjamin B Katz
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Joseph W Ziller
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Sudipta Majumdar
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Philip G Collins
- Department of Physics & Astronomy, University of California, Irvine, Irvine, California 92697, United States
| | - Gregory A Weiss
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States.,Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, California 92697, United States
| |
Collapse
|
31
|
Liewen H, Markuly N, Läubli H, Liu Y, Matter MS, Liewen N, Renner C, Zippelius A, Stenner F. Therapeutic Targeting of Golgi Phosphoprotein 2 (GOLPH2) with Armed Antibodies: A Preclinical Study of Anti-GOLPH2 Antibody Drug Conjugates in Lung and Colorectal Cancer Models of Patient Derived Xenografts (PDX). Target Oncol 2020; 14:577-590. [PMID: 31541350 DOI: 10.1007/s11523-019-00667-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Golgi phosphoprotein 2 (GOLPH2) has been shown to be involved in chronic inflammatory processes and carcinogenesis. GOLPH2 is prominently overexpressed in hepatocellular carcinoma, melanoma, glioblastoma, prostate, lung, and colorectal cancer. With a low and tightly regulated expression in non-malignant tissues, GOLPH2 has been proposed as an attractive target for cancer therapy. However, GOLPH2 is predominantly located intracellularly and when situated outside of the cell it is proteolytically cleaved and shed from the cell surface. Until now, GOLPH2 has been regarded as an "undruggable" target. OBJECTIVE We sought to create antibodies that specifically bind to GOLPH2 overexpressing tumor cells. PATIENTS AND METHODS Antibodies binding to membranous GOLPH2 despite shedding of the protein were generated from a scFV library screening. These antibodies target the part of GOLPH2 that remains at the cell surface after proteolytic cleavage. These antibodies were then tested in vitro and in vivo. RESULTS Two candidates (G2-1 and G2-2) showed target specific binding in vitro. Utilizing a tumor array (n = 128 tumors) with G2-2 and a reference antibody, a GOLPH2 expression scoring system was established. Rapid internalization of the antibodies was noted so this was exploited to deliver a toxic payload of pyrrolobenzodiazepine (PBD). In two patient-derived xenograft (PDX)-models, colorectal and lung cancer, the G2-2 antibody drug conjugate (ADC) displayed high efficacy with significant tumor responses (P = 0.001; P = 0.013) and improved survival (P = 0.0001; P = 0.0011) compared with controls. CONCLUSIONS Treatment with GOLPH2-directed antibodies induces durable responses in colorectal and lung cancer models. With a robust companion assay for GOLPH2 positivity at hand our findings prepare for the translation into a clinical trial.
Collapse
Affiliation(s)
- Heike Liewen
- Cureab GmbH, Benkenstrasse 254c, Technologiezentrum, 4108, Witterswil, Switzerland
| | - Norbert Markuly
- Cureab GmbH, Benkenstrasse 254c, Technologiezentrum, 4108, Witterswil, Switzerland
| | - Heinz Läubli
- Medical Oncology, University Hospital Basel & Laboratory Cancer Immunology, Department Biomedicine, University Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Yang Liu
- Medical Oncology, University Hospital Basel & Laboratory Cancer Immunology, Department Biomedicine, University Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Matthias S Matter
- Institute of Pathology, University Hospital Basel, 4031, Basel, Switzerland
| | - Nora Liewen
- University of Cologne, Albertus-Magnus-Platz, 50923, Cologne, Germany
| | - Christoph Renner
- Medical Oncology, University Hospital Basel & Laboratory Cancer Immunology, Department Biomedicine, University Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Alfred Zippelius
- Medical Oncology, University Hospital Basel & Laboratory Cancer Immunology, Department Biomedicine, University Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Frank Stenner
- Medical Oncology, University Hospital Basel & Laboratory Cancer Immunology, Department Biomedicine, University Basel, Petersgraben 4, 4031, Basel, Switzerland.
| |
Collapse
|
32
|
Antibody-Drug Conjugate Using Ionized Cys-Linker-MMAE as the Potent Payload Shows Optimal Therapeutic Safety. Cancers (Basel) 2020; 12:cancers12030744. [PMID: 32245171 PMCID: PMC7140114 DOI: 10.3390/cancers12030744] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/17/2020] [Accepted: 03/19/2020] [Indexed: 12/29/2022] Open
Abstract
Monomethyl auristatin E (MMAE) is the most popular and widely used cytotoxin in the development of antibody-drug conjugates (ADCs). However, current MMAE-based ADCs are all constructed using cleavable linkers, and this design concept still has insurmountable drawbacks. Their potential instabilities and lipophilic MMAE-induced “bystander effect” inevitably increase the toxicity to normal tissues. Herein, we overturn previous negative views of MMAE-based ADCs with non-cleavable linkers and propose using ionized L-Cysteine (Cys)-linker-MMAE as a novel payload, which can ingeniously enrich and enter tumor cells through receptor-mediated endocytosis of antibodies while its lower permeability helps to avoid further off-target toxicity. We demonstrate that Cys-linker-MMAE maintains high potency similar to free MMAE at the tubulin molecular level and can also be efficiently released in target cells. As a result, the preferred ADC (mil40-15) not only exhibits ideal plasma stability and maintains potent cytotoxicity as MMAE (IC50: 10−11 M), but also shows improved safety with lower bystander toxicity (IC50: 10−9 M), its maximum tolerated dose approaching the level of the naked antibody (160 mg/kg). This study indicated that Cys-linker-MMAE has the potential as a potent payload for ADCs, which is expected to provide novel strategies for the development of MMAE-based ADCs.
Collapse
|
33
|
Wall A, Nicholls K, Caspersen MB, Skrivergaard S, Howard KA, Karu K, Chudasama V, Baker JR. Optimised approach to albumin-drug conjugates using monobromomaleimide-C-2 linkers. Org Biomol Chem 2020; 17:7870-7873. [PMID: 31410415 DOI: 10.1039/c9ob00721k] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Conjugation of therapeutics to human serum albumin (HSA) using bromomaleimides represents a promising platform for half-life extension. We show here that the Cys-34 crevice substantially reduces the rate of serum stabilising maleimide hydrolysis in these conjugates, necessitating reagent optimisation. This improved reagent design is applied to the construction of an HSA-paclitaxel conjugate, preventing drug loss during maleimide hydrolysis.
Collapse
Affiliation(s)
- Archie Wall
- Department of Chemistry, UCL, 20 Gordon St, London, WC1H 0AJ, UK.
| | - Karl Nicholls
- Albumedix Ltd, Castle Court, 59 Castle Boulevard, Nottingham NG7 1FD, UK
| | - Mikael B Caspersen
- Albumedix Ltd, Castle Court, 59 Castle Boulevard, Nottingham NG7 1FD, UK
| | - Stig Skrivergaard
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Kenneth A Howard
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Kersti Karu
- Department of Chemistry, UCL, 20 Gordon St, London, WC1H 0AJ, UK.
| | - Vijay Chudasama
- Department of Chemistry, UCL, 20 Gordon St, London, WC1H 0AJ, UK. and Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - James R Baker
- Department of Chemistry, UCL, 20 Gordon St, London, WC1H 0AJ, UK.
| |
Collapse
|
34
|
Abstract
The prototypical ADC mechanism involving antigen-mediated uptake and lysosomal release is both elegantly simple and scientifically compelling. However, recent clinical-stage failures have prompted a reevaluation of this delivery paradigm and have resulted in an array of new technologies that have the potential to improve the safety and efficacy of up and coming programs. These innovations can generally be categorized into seven areas that will be elaborated on in this chapter: (1) Exploiting new payload mechanisms; (2) Increasing the drug-antibody ratio (DAR); (3) Increasing the antibody penetration; (4) Overcoming ADC resistance mechanisms; (5) Increasing the efficiency of ADC uptake and processing; (6) Mitigating off-target payload exposure; and (7) Employment of noncytotoxic payloads. It is our belief that these seven areas capture the current "landscape" of innovations that are taking place in the design of next-generation ADCs. Together, these advancements are reshaping the ADC field and providing a path forward in the face of the recent clinical setbacks.
Collapse
Affiliation(s)
- L Nathan Tumey
- Department of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA.
- Pfizer Inc., Groton, CT, USA.
| |
Collapse
|
35
|
Lim SI. Site-specific bioconjugation and self-assembly technologies for multi-functional biologics: on the road to the clinic. Drug Discov Today 2019; 25:168-176. [PMID: 31610287 DOI: 10.1016/j.drudis.2019.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/28/2019] [Accepted: 10/03/2019] [Indexed: 01/02/2023]
Abstract
The expanding portfolio of biotherapeutics both in the research and development (R&D) and market sectors is shaping new opportunities towards multifunctional biologics (MFBs). The combination of new or pre-existing therapeutic agents into a single multifunctional format makes it possible to develop new pharmacological actions to significantly improve their efficacy and safety. In this review, I focus on novel platform technologies that are being exploited in the biotech industry to produce MFBs with potential therapeutic benefits that include half-life extension, targeted delivery, T cell engagement, and improved vaccination. In this regard, technologies of key importance are site-specific bioconjugation and self-assembly, which allow homogeneous, defined, and scalable process developments for several MFBs that are advancing towards clinical applications.
Collapse
Affiliation(s)
- Sung In Lim
- Department of Chemical Engineering, Pukyong National University, 45 Yongso-ro, Nam-gu, Busan, Republic of Korea.
| |
Collapse
|
36
|
Peltek OO, Muslimov AR, Zyuzin MV, Timin AS. Current outlook on radionuclide delivery systems: from design consideration to translation into clinics. J Nanobiotechnology 2019; 17:90. [PMID: 31434562 PMCID: PMC6704557 DOI: 10.1186/s12951-019-0524-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/14/2019] [Indexed: 02/06/2023] Open
Abstract
Radiopharmaceuticals have proven to be effective agents, since they can be successfully applied for both diagnostics and therapy. Effective application of relevant radionuclides in pre-clinical and clinical studies depends on the choice of a sufficient delivery platform. Herein, we provide a comprehensive review on the most relevant aspects in radionuclide delivery using the most employed carrier systems, including, (i) monoclonal antibodies and their fragments, (ii) organic and (iii) inorganic nanoparticles, and (iv) microspheres. This review offers an extensive analysis of radionuclide delivery systems, the approaches of their modification and radiolabeling strategies with the further prospects of their implementation in multimodal imaging and disease curing. Finally, the comparative outlook on the carriers and radionuclide choice, as well as on the targeting efficiency of the developed systems is discussed.
Collapse
Affiliation(s)
- Oleksii O Peltek
- Russian Research Center of Radiology and Surgical Technologies (RRCRST) of Ministry of Public Health, Leningradskaya Street 70 Pesochny, Saint-Petersburg, 197758, Russian Federation
| | - Albert R Muslimov
- Russian Research Center of Radiology and Surgical Technologies (RRCRST) of Ministry of Public Health, Leningradskaya Street 70 Pesochny, Saint-Petersburg, 197758, Russian Federation
| | - Mikhail V Zyuzin
- Faculty of Physics and Engineering, ITMO University, St. Petersburg, 197101, Russia
| | - Alexander S Timin
- Russian Research Center of Radiology and Surgical Technologies (RRCRST) of Ministry of Public Health, Leningradskaya Street 70 Pesochny, Saint-Petersburg, 197758, Russian Federation.
- Research School of Chemical and Biomedical Engineering, National Research Tomsk Polytechnic University, Lenin Avenue 30, Tomsk, 634050, Russia.
| |
Collapse
|
37
|
Wang Y, Fan S, Xiao D, Xie F, Li W, Zhong W, Zhou X. Novel Silyl Ether-Based Acid-Cleavable Antibody-MMAE Conjugates with Appropriate Stability and Efficacy. Cancers (Basel) 2019; 11:cancers11070957. [PMID: 31288450 PMCID: PMC6678733 DOI: 10.3390/cancers11070957] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/29/2019] [Accepted: 07/04/2019] [Indexed: 02/07/2023] Open
Abstract
Antibody-drug conjugate (ADC) is a novel efficient drug delivery system that has been successfully used in clinical practice, and it has become a research hotspot in the anti-tumor drug field. Acid-cleavable linkers were first used in clinical ADCs, but their structural variety (e.g., hydrazone and carbonate) is still limited, and their stability is usually insufficient. Designing novel acid-cleavable linkers for the conjugation of the popular cytotoxin monomethyl auristatin E (MMAE) has always been a significant topic. In this paper, we generate a novel, silyl ether-based acid-cleavable antibody-MMAE conjugate, which skillfully achieves efficient combination of amino-conjugated MMAE with the acid-triggered silyl ether group by introducing p-hydroxybenzyl alcohol (PHB). The stability, acid-dependence cleavage, effective mechanism, efficacy and safety of the resulting ADC were systematically studied; the results show that it exhibits a significant improvement in stability, while maintaining appropriate efficacy and controlled therapeutic toxicity. This strategy is expected to expand a new type of acid-cleavable linkers for the development of ADCs with highly potent payloads.
Collapse
Affiliation(s)
- Yanming Wang
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Shiyong Fan
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Dian Xiao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Fei Xie
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Wei Li
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Xinbo Zhou
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| |
Collapse
|
38
|
Lee BS, Lee Y, Park J, Jeong BS, Jo M, Jung ST, Yoo TH. Construction of an immunotoxin via site-specific conjugation of anti-Her2 IgG and engineered Pseudomonas exotoxin A. J Biol Eng 2019; 13:56. [PMID: 31285754 PMCID: PMC6588878 DOI: 10.1186/s13036-019-0188-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/12/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Immunotoxins consisting of a toxin from bacteria or plants and a targeting module have been developed as potent anti-cancer therapeutics. The majority of them, especially those in preclinical or clinical testing stages, are fusion proteins of a toxin and antibody fragment. Immunotoxins based on full-length antibodies are less studied, even though the fragment crystallizable (Fc) domain plays an important role in regulating the concentration of immunoglobulin G (IgG) in the serum and in antibody-mediated immune responses against pathogens. RESULTS We devised a method to site-specifically conjugate IgG and another protein using a cysteine residue introduced into the IgG and a bio-orthogonally reactive unnatural amino acid incorporated into the other protein. The human epidermal growth factor receptor 2 (Her2)-targeting IgG, trastuzumab, was engineered to have an unpaired cysteine in the heavy chain, and an unnatural amino acid with the azido group was incorporated into an engineered Pseudomonas exotoxin A (PE24). The two protein molecules were conjugated site-specifically using a bifunctional linker having dibenzocyclooctyne and maleimide groups. Binding to Her2 and interaction with various Fc receptors of trastuzumab were not affected by the conjugation with PE24. The trastuzumab-PE24 conjugate was cytotoxic to Her2-overexpressing cell lines, which involved the inhibition of cellular protein synthesis due to the modification of elongation factor-2. CONCLUSIONS We constructed the site-specifically conjugated immunotoxin based on IgG and PE24, which induced target-specific cytotoxicity. To evaluate the molecule as a cancer therapeutic, animal studies are planned to assess tumor regression, half-life in blood, and in vivo immunogenicity. In addition, we expect that the site-specific conjugation method can be used to develop other antibody-protein conjugates for applications in therapeutics and diagnostics.
Collapse
Affiliation(s)
- Byeong Sung Lee
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 16499 South Korea
| | - Yumi Lee
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 16499 South Korea
| | - Jisoo Park
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 16499 South Korea
| | - Bo Seok Jeong
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 16499 South Korea
| | - Migyeong Jo
- Department of Applied Chemistry, Kookmin University, 77 Jeongneung-ro, Seongbuk-gu, Seoul, 02707 South Korea
| | - Sang Taek Jung
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, Seongbuk-gu, Seoul, 02841 South Korea
| | - Tae Hyeon Yoo
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 16499 South Korea
- Department of Applied Chemistry and Biological Engineering, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, 16499 South Korea
| |
Collapse
|
39
|
Is hydrophobic interaction chromatography the most suitable technique to characterize site-specific antibody-drug conjugates? J Chromatogr A 2019; 1586:149-153. [DOI: 10.1016/j.chroma.2018.12.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/04/2018] [Accepted: 12/10/2018] [Indexed: 12/23/2022]
|
40
|
Ravasco JMJM, Faustino H, Trindade A, Gois PMP. Bioconjugation with Maleimides: A Useful Tool for Chemical Biology. Chemistry 2018; 25:43-59. [PMID: 30095185 DOI: 10.1002/chem.201803174] [Citation(s) in RCA: 330] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Indexed: 02/06/2023]
Abstract
Maleimide chemistry stands out in the bioconjugation toolbox by virtue of its synthetic accessibility, excellent reactivity, and practicability. The second-generation of clinically approved antibody-drug conjugates (ADC) and much of the current ADC pipeline in clinical trials contain the maleimide linkage. However, thiosuccinimide linkages are now known to be less robust than once thought, and ergo, are correlated with suboptimal pharmacodynamics, pharmacokinetics, and safety profiles in some ADC constructs. Rational design of novel generations of maleimides and maleimide-type reagents have been reported to address the shortcomings of classical maleimides, allowing for the formation of robust bioconjugate linkages. This review highlights the main strategies for rational reagent design that have allowed irreversible bioconjugations in cysteines, reversible labelling strategies and disulfide re-bridging.
Collapse
Affiliation(s)
- João M J M Ravasco
- Bioorganic Chemistry Department, Research Institute for Medicines, (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Hélio Faustino
- Bioorganic Chemistry Department, Research Institute for Medicines, (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Alexandre Trindade
- Bioorganic Chemistry Department, Research Institute for Medicines, (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal.,School of Chemistry, University of Leeds, Leeds, LS2 9JT, UK
| | - Pedro M P Gois
- Bioorganic Chemistry Department, Research Institute for Medicines, (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| |
Collapse
|
41
|
Morais M, Ma MT. Site-specific chelator-antibody conjugation for PET and SPECT imaging with radiometals. DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 30:91-104. [PMID: 30553525 PMCID: PMC6291455 DOI: 10.1016/j.ddtec.2018.10.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 11/17/2022]
Abstract
Antibodies and their derivatives radiolabelled with positron- and gamma-emitting radiometals enable sensitive and quantitative molecular Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT) imaging of antibody distribution in vivo. Chelators that are covalently attached to antibodies allow radiolabelling with metallic PET and SPECT radioisotopes. Conventional strategies for chelator-protein conjugation generate heterogeneous mixtures of bioconjugates that can exhibit reduced affinity for their receptor targets, and undesirable biodistribution and pharmacokinetics. Recent advances in bioconjugation technology enable site-specific modification to generate well-defined constructs with superior properties. Herein we survey existing site-specific chelator-protein conjugation methods. These include chelator attachment to cysteines/disulfide bonds or the glycan region of the antibody, enzyme-mediated chelator conjugation, and incorporation of sequences of amino acids that chelate the radiometal. Such technology will allow better use of PET and SPECT imaging in the development of antibody-based therapies.
Collapse
Affiliation(s)
- Mauricio Morais
- School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom.
| | - Michelle T Ma
- School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| |
Collapse
|
42
|
Renault K, Fredy JW, Renard PY, Sabot C. Covalent Modification of Biomolecules through Maleimide-Based Labeling Strategies. Bioconjug Chem 2018; 29:2497-2513. [PMID: 29954169 DOI: 10.1021/acs.bioconjchem.8b00252] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Since their first use in bioconjugation more than 50 years ago, maleimides have become privileged chemical partners for the site-selective modification of proteins via thio-Michael addition of biothiols and, to a lesser extent, via Diels-Alder (DA) reactions with biocompatible dienes. Prominent examples include immunotoxins and marketed maleimide-based antibody-drug conjugates (ADCs) such as Adcetris, which are used in cancer therapies. Among the key factors in the success of these groups is the availability of several maleimides that can be N-functionalized by fluorophores, affinity tags, spin labels, and pharmacophores, as well as their unique reactivities in terms of selectivity and kinetics. However, maleimide conjugate reactions have long been considered irreversible, and only recently have systematic studies regarding their reversibility and stability toward hydrolysis been reported. This review provides an overview of the diverse applications for maleimides in bioconjugation, highlighting their strengths and weaknesses, which are being overcome by recent strategies. Finally, the fluorescence quenching ability of maleimides was leveraged for the preparation of fluorogenic probes, which are mainly used for the specific detection of thiol analytes. A summary of the reported structures, their photophysical features, and their relative efficiencies is discussed in the last part of the review.
Collapse
Affiliation(s)
- Kévin Renault
- Normandie Univ, CNRS, UNIROUEN, INSA Rouen, COBRA (UMR 6014) , 76000 Rouen , France
| | - Jean Wilfried Fredy
- Normandie Univ, CNRS, UNIROUEN, INSA Rouen, COBRA (UMR 6014) , 76000 Rouen , France
| | - Pierre-Yves Renard
- Normandie Univ, CNRS, UNIROUEN, INSA Rouen, COBRA (UMR 6014) , 76000 Rouen , France
| | - Cyrille Sabot
- Normandie Univ, CNRS, UNIROUEN, INSA Rouen, COBRA (UMR 6014) , 76000 Rouen , France
| |
Collapse
|