1
|
Ma S, Chen H, Liu S, Huang X, Mo T, Liu WQ, Zhang W, Ding W, Zhang Q. A gene-encoded aldehyde tag repurposed from RiPP cyclophane-forming pathway. Bioorg Med Chem Lett 2024; 101:129653. [PMID: 38360420 DOI: 10.1016/j.bmcl.2024.129653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/19/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
Gene-encoded aldehyde tag technology has been widely utilized in protein bioorthogonal chemistry and biotechnological application. Herein, we report utilization of the promiscuous rSAM cyclophane synthase SjiB involved in triceptide biosynthesis as a dedicated and highly efficient formylglycine synthase. The new aldehyde tag sequence in this system, YQSSI, is biosynthetically orthogonal to the known aldehyde tag (C/S)x(P/A)xR. The potential use of SjiB/YQSSI aldehyde tag system was further validated in fluorescent labelling of model proteins.
Collapse
Affiliation(s)
- Suze Ma
- Department of Chemistry, Fudan University, Shanghai 200433, China
| | - Heng Chen
- Department of Chemistry, Fudan University, Shanghai 200433, China; State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shuxun Liu
- Department of Chemistry, Fudan University, Shanghai 200433, China
| | - Xuedong Huang
- Department of Chemistry, Fudan University, Shanghai 200433, China
| | - Tianlu Mo
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Wan-Qiu Liu
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wei Zhang
- Key Laboratory of Extreme Environmental Microbial Resources and Engineering of Gansu Province, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Wei Ding
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qi Zhang
- Department of Chemistry, Fudan University, Shanghai 200433, China.
| |
Collapse
|
2
|
Dudchak R, Podolak M, Holota S, Szewczyk-Roszczenko O, Roszczenko P, Bielawska A, Lesyk R, Bielawski K. Click chemistry in the synthesis of antibody-drug conjugates. Bioorg Chem 2024; 143:106982. [PMID: 37995642 DOI: 10.1016/j.bioorg.2023.106982] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/31/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
Antibody-Drug Conjugates (ADC) are a new class of anticancer therapeutics with immense potential. They have been rapidly advancing in the last two decades. This fast speed of development has become possible due to several new technologies and methods. One of them is Click Chemistry, an approach that was created only two decades ago, but already is actively utilized for bioconjugation, material science and drug discovery. In this review, we researched the impact of Click Chemistry reactions on the synthesis and development of ADCs. The information about the most frequently utilized reactions, such as Michael's addition, Copper-catalyzed azide-alkyne [3+2] cycloaddition (CuAAC), Strain-promoted azide-alkyne [3+2] cycloaddition (SPAAC), oxime bond formation, hydrazine-iso-Pictet-Spengler Ligation (HIPS), Diels-Alder reactions have been summarized. The implementation of thiol-maleimide Click Chemistry reaction in the synthesis of numerous FDA-approved Antibody-Drug Conjugates has been reported. The data amassed in the present review provides better understanding of the importance of Click Chemistry in the synthesis, development and improvement of the Antibody-Drug Conjugates and it will be helpful for further researches related to ADCs.
Collapse
Affiliation(s)
- Rostyslav Dudchak
- Department of Synthesis and Technology of Drugs, Faculty of Pharmacy, Medical University of Bialystok, Jana Kilińskiego 1, Bialystok 15-089, Poland
| | - Magdalena Podolak
- Department of Biotechnology, Faculty of Pharmacy, Medical University of Bialystok, Jana Kilińskiego 1, Bialystok 15-089, Poland
| | - Serhii Holota
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, Lviv 79010, Ukraine
| | - Olga Szewczyk-Roszczenko
- Department of Synthesis and Technology of Drugs, Faculty of Pharmacy, Medical University of Bialystok, Jana Kilińskiego 1, Bialystok 15-089, Poland
| | - Piotr Roszczenko
- Department of Biotechnology, Faculty of Pharmacy, Medical University of Bialystok, Jana Kilińskiego 1, Bialystok 15-089, Poland
| | - Anna Bielawska
- Department of Biotechnology, Faculty of Pharmacy, Medical University of Bialystok, Jana Kilińskiego 1, Bialystok 15-089, Poland
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, Lviv 79010, Ukraine.
| | - Krzysztof Bielawski
- Department of Synthesis and Technology of Drugs, Faculty of Pharmacy, Medical University of Bialystok, Jana Kilińskiego 1, Bialystok 15-089, Poland
| |
Collapse
|
3
|
Tufail A, Akkad S, Hatton NE, Yates NDJ, Spears RJ, Keenan T, Parkin A, Signoret N, Fascione MA. Cross aldol OPAL bioconjugation outcompetes intramolecular hemiaminal cyclisation of proline adjacent N-terminal α-oxo aldehydes at acidic pH. RSC Adv 2024; 14:3723-3729. [PMID: 38268544 PMCID: PMC10806391 DOI: 10.1039/d3ra08776j] [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: 12/22/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024] Open
Abstract
Novel methods to construct small molecule-protein bioconjugates are integral to the development of new biomedicines for a variety of diseases. C-C linked bioconjugates are increasingly desirable in this application due to their in vivo stability and can be accessed through cross aldol bioconjugation of reactive α-oxo aldehyde handles easily introduced at the N-terminus of proteins by periodate oxidation. We previously developed an organocatalyst-mediated protein aldol ligation (OPAL) for chemical modification of these reactive aldehydes, but the efficiency of this method was limited when a proline residue was directly adjacent to the N-terminus due to intramolecular hemiaminal formation. Herein we explore the competition between this cyclisation and the OPAL modification and demonstrate bioconjugation can be favoured through use of acidic pH for both oxidation and OPAL, and optimisation of reaction conditions and organocatalyst. We then showcase the utility of this acidic-OPAL in modification of the cholera toxin B-subunit (CTB), a homo-pentameric protein of biomedical promise.
Collapse
Affiliation(s)
- Afzaal Tufail
- Department of Chemistry, University of York Heslington York YO10 5DD UK
- Hull York Medical School, University of York YO10 5DD UK
| | - Saeed Akkad
- Department of Chemistry, University of York Heslington York YO10 5DD UK
| | - Natasha E Hatton
- Department of Chemistry, University of York Heslington York YO10 5DD UK
| | | | - Richard J Spears
- Department of Chemistry, University of York Heslington York YO10 5DD UK
| | - Tessa Keenan
- Department of Chemistry, University of York Heslington York YO10 5DD UK
| | - Alison Parkin
- Department of Chemistry, University of York Heslington York YO10 5DD UK
| | | | - Martin A Fascione
- Department of Chemistry, University of York Heslington York YO10 5DD UK
| |
Collapse
|
4
|
Lee D, Latour S, Emblem M, Clark HJ, Santos JT, Jang J, McGuigan AP, Nitz M. Characterization of an N-Allylglyoxylamide-Based Bioorthogonal Nitrone Trap. Bioconjug Chem 2023; 34:2358-2365. [PMID: 38051144 DOI: 10.1021/acs.bioconjchem.3c00463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Aldehydes are attractive bioorthogonal coupling partners. The ease of manipulation of aldehydes and their orthogonality to other classes of bioorthogonal reactions have inspired the exploration of chemistries, which generate irreversible conjugates. Similarly, nitrones have been shown to be potent 1,3-dipoles in bioorthogonal reactions when paired with strained alkynes. Here, we combine the reactivity of nitrones with the simplicity of aldehydes using an N-allylglyoxylamide, in a cascade reaction with an N-alkylhydroxylamine to produce a bicyclic isoxazolidine. The reaction is found to be catalyzed by 5-methoxyanthranilic acid and proceeds at pH 7 with favorable kinetics. Using the HaloTag7 protein bearing an N-alkylhydroxylamine, we show the reaction to be bioorthogonal in a complex cell lysate and to proceed well at the surface of a HEK293 cell. Furthermore, the reaction is compatible with a typical strain-promoted alkyne-azide click reaction. The characteristics of this reaction suggest it will be a useful addition to the pallet of bioorthogonal reactions that have revolutionized chemical biology.
Collapse
Affiliation(s)
- Daniel Lee
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Simon Latour
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E6, Canada
| | - Michael Emblem
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Hunter J Clark
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Jobette T Santos
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Jaewan Jang
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Alison P McGuigan
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E6, Canada
| | - Mark Nitz
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| |
Collapse
|
5
|
Balamkundu S, Liu CF. Lysosomal-Cleavable Peptide Linkers in Antibody-Drug Conjugates. Biomedicines 2023; 11:3080. [PMID: 38002080 PMCID: PMC10669454 DOI: 10.3390/biomedicines11113080] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Antibody-drug Conjugates (ADCs) are a powerful therapeutic modality for cancer treatment. ADCs are multi-functional biologics in which a disease-targeting antibody is conjugated to an effector payload molecule via a linker. The success of currently used ADCs has been largely attributed to the development of linker systems, which allow for the targeted release of cytocidal payload drugs inside cancer cells. Many lysosomal proteases are over expressed in human cancers. They can effectively cleave a variety of peptide sequences, which can be exploited for the design of ADC linker systems. As a well-established linker, valine-citrulline-p-aminobenzyl carbamate (ValCitPABC) is used in many ADCs that are already approved or under preclinical and clinical development. Although ValCitPABC and related linkers are readily cleaved by cathepsins in the lysosome while remaining reasonably stable in human plasma, many studies have shown that they are susceptible to carboxylesterase 1C (Ces1C) in mouse and rat plasma, which hinders the preclinical evaluation of ADCs. Furthermore, neutropenia and thrombocytopenia, two of the most commonly observed dose-limiting adverse effects of ADCs, are believed to result from the premature hydrolysis of ValCitPABC by human neutrophil elastase. In addition to ValCitPABC, the GGFG tetrapeptidyl-aminomethoxy linker is also cathepsin-cleavable and is used in the highly successful ADC drug, DS8201a. In addition to cathepsin-cleavable linkers, there is also growing interest in legumain-sensitive linkers for ADC development. Increasing plasma stability while maintaining lysosomal cleavability of ADC linkers is an objective of intensive current research. This review reports recent advances in the design and structure-activity relationship studies of various peptide/peptidomimetic linkers in this field.
Collapse
Affiliation(s)
| | - Chuan-Fa Liu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore;
| |
Collapse
|
6
|
Sasso J, Tenchov R, Bird R, Iyer KA, Ralhan K, Rodriguez Y, Zhou QA. The Evolving Landscape of Antibody-Drug Conjugates: In Depth Analysis of Recent Research Progress. Bioconjug Chem 2023; 34:1951-2000. [PMID: 37821099 PMCID: PMC10655051 DOI: 10.1021/acs.bioconjchem.3c00374] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Antibody-drug conjugates (ADCs) are targeted immunoconjugate constructs that integrate the potency of cytotoxic drugs with the selectivity of monoclonal antibodies, minimizing damage to healthy cells and reducing systemic toxicity. Their design allows for higher doses of the cytotoxic drug to be administered, potentially increasing efficacy. They are currently among the most promising drug classes in oncology, with efforts to expand their application for nononcological indications and in combination therapies. Here we provide a detailed overview of the recent advances in ADC research and consider future directions and challenges in promoting this promising platform to widespread therapeutic use. We examine data from the CAS Content Collection, the largest human-curated collection of published scientific information, and analyze the publication landscape of recent research to reveal the exploration trends in published documents and to provide insights into the scientific advances in the area. We also discuss the evolution of the key concepts in the field, the major technologies, and their development pipelines with company research focuses, disease targets, development stages, and publication and investment trends. A comprehensive concept map has been created based on the documents in the CAS Content Collection. We hope that this report can serve as a useful resource for understanding the current state of knowledge in the field of ADCs and the remaining challenges to fulfill their potential.
Collapse
Affiliation(s)
- Janet
M. Sasso
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Rumiana Tenchov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Robert Bird
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | | | - Yacidzohara Rodriguez
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
7
|
Richter D, Lakis E, Piel J. Site-specific bioorthogonal protein labelling by tetrazine ligation using endogenous β-amino acid dienophiles. Nat Chem 2023; 15:1422-1430. [PMID: 37400596 PMCID: PMC10533398 DOI: 10.1038/s41557-023-01252-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 05/24/2023] [Indexed: 07/05/2023]
Abstract
The tetrazine ligation is an inverse electron-demand Diels-Alder reaction widely used for bioorthogonal modifications due to its versatility, site specificity and fast reaction kinetics. A major limitation has been the incorporation of dienophiles in biomolecules and organisms, which relies on externally added reagents. Available methods require the incorporation of tetrazine-reactive groups by enzyme-mediated ligations or unnatural amino acid incorporation. Here we report a tetrazine ligation strategy, termed TyrEx (tyramine excision) cycloaddition, permitting autonomous dienophile generation in bacteria. It utilizes a unique aminopyruvate unit introduced by post-translational protein splicing at a short tag. Tetrazine conjugation occurs rapidly with a rate constant of 0.625 (15) M-1 s-1 and was applied to produce a radiolabel chelator-modified Her2-binding Affibody and intracellular, fluorescently labelled cell division protein FtsZ. We anticipate the labelling strategy to be useful for intracellular studies of proteins, as a stable conjugation method for protein therapeutics, as well as other applications.
Collapse
Affiliation(s)
- Daniel Richter
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Zürich, Switzerland
| | - Edgars Lakis
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Zürich, Switzerland
| | - Jörn Piel
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Zürich, Switzerland.
| |
Collapse
|
8
|
Debon A, Siirola E, Snajdrova R. Enzymatic Bioconjugation: A Perspective from the Pharmaceutical Industry. JACS AU 2023; 3:1267-1283. [PMID: 37234110 PMCID: PMC10207132 DOI: 10.1021/jacsau.2c00617] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/10/2023] [Accepted: 01/10/2023] [Indexed: 05/27/2023]
Abstract
Enzymes have firmly established themselves as bespoke catalysts for small molecule transformations in the pharmaceutical industry, from early research and development stages to large-scale production. In principle, their exquisite selectivity and rate acceleration can also be leveraged for modifying macromolecules to form bioconjugates. However, available catalysts face stiff competition from other bioorthogonal chemistries. In this Perspective, we seek to illuminate applications of enzymatic bioconjugation in the face of an expanding palette of new drug modalities. With these applications, we wish to highlight some examples of current successes and pitfalls of using enzymes for bioconjugation along the pipeline and try to illustrate opportunities for further development.
Collapse
Affiliation(s)
- Aaron Debon
- Global
Discovery Chemistry, Novartis Institute
for Biomedical Research, Basel 4108, Switzerland
| | - Elina Siirola
- Global
Discovery Chemistry, Novartis Institute
for Biomedical Research, Basel 4108, Switzerland
| | - Radka Snajdrova
- Global
Discovery Chemistry, Novartis Institute
for Biomedical Research, Basel 4108, Switzerland
| |
Collapse
|
9
|
Ni Y, Wang Y, Tabor AB, Ward JM, Hailes HC. The use of tyrosinases in a chemoenzymatic cascade as a peptide ligation strategy. RSC Chem Biol 2023; 4:132-137. [PMID: 36794017 PMCID: PMC9906322 DOI: 10.1039/d2cb00237j] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Peptides play many key roles in biological systems and numerous methods have been developed to generate both natural and unnatural peptides. However, straightforward, reliable coupling methods that can be achieved under mild reactions conditions are still sought after. In this work, a new N-terminal tyrosine-containing peptide ligation method with aldehydes, utilising a Pictet-Spengler reaction is described. In a key step, tyrosinase enzymes have been used to convert l-tyrosine to l-3,4-dihydroxyphenyl alanine (l-DOPA) residues, generating suitable functionality for the Pictet-Spengler coupling. This new chemoenzymatic coupling strategy can be used for fluorescent-tagging and peptide ligation purposes.
Collapse
Affiliation(s)
- Yeke Ni
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Yu Wang
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Alethea B. Tabor
- Department of Chemistry, University College London20 Gordon StreetLondonWC1H 0AJUK
| | - John M. Ward
- Department of Biochemical Engineering, University College LondonBernard Katz Building, Gower StreetLondon WC1E 6BTUK
| | - Helen C. Hailes
- Department of Chemistry, University College London20 Gordon StreetLondonWC1H 0AJUK
| |
Collapse
|
10
|
Site-Specific Antibody Conjugation with Payloads beyond Cytotoxins. Molecules 2023; 28:molecules28030917. [PMID: 36770585 PMCID: PMC9921355 DOI: 10.3390/molecules28030917] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 01/18/2023] Open
Abstract
As antibody-drug conjugates have become a very important modality for cancer therapy, many site-specific conjugation approaches have been developed for generating homogenous molecules. The selective antibody coupling is achieved through antibody engineering by introducing specific amino acid or unnatural amino acid residues, peptides, and glycans. In addition to the use of synthetic cytotoxins, these novel methods have been applied for the conjugation of other payloads, including non-cytotoxic compounds, proteins/peptides, glycans, lipids, and nucleic acids. The non-cytotoxic compounds include polyethylene glycol, antibiotics, protein degraders (PROTAC and LYTAC), immunomodulating agents, enzyme inhibitors and protein ligands. Different small proteins or peptides have been selectively conjugated through unnatural amino acid using click chemistry, engineered C-terminal formylglycine for oxime or click chemistry, or specific ligation or transpeptidation with or without enzymes. Although the antibody protamine peptide fusions have been extensively used for siRNA coupling during early studies, direct conjugations through engineered cysteine or lysine residues have been demonstrated later. These site-specific antibody conjugates containing these payloads other than cytotoxic compounds can be used in proof-of-concept studies and in developing new therapeutics for unmet medical needs.
Collapse
|
11
|
Cheng-Sánchez I, Moya-Utrera F, Porras-Alcalá C, López-Romero JM, Sarabia F. Antibody-Drug Conjugates Containing Payloads from Marine Origin. Mar Drugs 2022; 20:md20080494. [PMID: 36005497 PMCID: PMC9410405 DOI: 10.3390/md20080494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 12/10/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are an important class of therapeutics for the treatment of cancer. Structurally, an ADC comprises an antibody, which serves as the delivery system, a payload drug that is a potent cytotoxin that kills cancer cells, and a chemical linker that connects the payload with the antibody. Unlike conventional chemotherapy methods, an ADC couples the selective targeting and pharmacokinetic characteristics related to the antibody with the potent cytotoxicity of the payload. This results in high specificity and potency by reducing off-target toxicities in patients by limiting the exposure of healthy tissues to the cytotoxic drug. As a consequence of these outstanding features, significant research efforts have been devoted to the design, synthesis, and development of ADCs, and several ADCs have been approved for clinical use. The ADC field not only relies upon biology and biochemistry (antibody) but also upon organic chemistry (linker and payload). In the latter, total synthesis of natural and designed cytotoxic compounds, together with the development of novel synthetic strategies, have been key aspects of the consecution of clinical ADCs. In the case of payloads from marine origin, impressive structural architectures and biological properties are observed, thus making them prime targets for chemical synthesis and the development of ADCs. In this review, we explore the molecular and biological diversity of ADCs, with particular emphasis on those containing marine cytotoxic drugs as the payload.
Collapse
Affiliation(s)
- Iván Cheng-Sánchez
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Correspondence:
| | - Federico Moya-Utrera
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, 29071 Málaga, Spain; (F.M.-U.); (C.P.-A.); (J.M.L.-R.); (F.S.)
| | - Cristina Porras-Alcalá
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, 29071 Málaga, Spain; (F.M.-U.); (C.P.-A.); (J.M.L.-R.); (F.S.)
| | - Juan M. López-Romero
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, 29071 Málaga, Spain; (F.M.-U.); (C.P.-A.); (J.M.L.-R.); (F.S.)
| | - Francisco Sarabia
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, 29071 Málaga, Spain; (F.M.-U.); (C.P.-A.); (J.M.L.-R.); (F.S.)
| |
Collapse
|
12
|
Gaviña D, Escolano M, Rabasa Alcañiz F, Díaz Oltra S, Sanchez-Rosello M, del Pozo C. Tandem Asymmetric Cycloaromatization/intramolecular Pictet‐Spengler‐type Reaction. An Entry to Polycyclic Pyrroles. Adv Synth Catal 2022. [DOI: 10.1002/adsc.202200399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
13
|
Stump B. Click Bioconjugation - Modifying Proteins using Click-Like Chemistry. Chembiochem 2022; 23:e202200016. [PMID: 35491526 DOI: 10.1002/cbic.202200016] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/27/2022] [Indexed: 11/11/2022]
Abstract
Bioconjugation is dealing with the chemical modification of proteins. The reactions used exploit either the intrinsic chemical reactivity of the biomolecule or introduce functionalities that can then be subsequently reacted without interfering with other functional groups of the biological entity. Perfectly selective, high yielding chemical transformations are needed that can be run in aqueous environment at mild pH conditions. Requirements that have an obvious overlap with the definition of click chemistry. This review shows a selection of successfully applied click-type reactions in bioconjugation as well as some recent developments to broaden the chemical toolbox to meet the challenge of a selective, bioorthogonal modification of biomolecules.
Collapse
Affiliation(s)
- Bernhard Stump
- Lonza AG: Lonza Ltd, Bioconjugates, Rottenstr, 3930, Visp, SWITZERLAND
| |
Collapse
|
14
|
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: 7] [Impact Index Per Article: 3.5] [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
|
15
|
Bivalent EGFR-Targeting DARPin-MMAE Conjugates. Int J Mol Sci 2022; 23:ijms23052468. [PMID: 35269611 PMCID: PMC8909960 DOI: 10.3390/ijms23052468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 02/14/2022] [Accepted: 02/18/2022] [Indexed: 02/04/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) is a validated tumor marker overexpressed in various cancers such as squamous cell carcinoma (SSC) of the head and neck and gliomas. We constructed protein-drug conjugates based on the anti-EGFR Designed Ankyrin Repeat Protein (DARPin) E01, and compared the bivalent DARPin dimer (DD1) and a DARPin-Fc (DFc) to the monomeric DARPin (DM) and the antibody derived scFv425-Fc (scFvFc) in cell culture and a mouse model. The modular conjugation system, which was successfully applied for the preparation of protein-drug and -dye conjugates, uses bio-orthogonal protein-aldehyde generation by the formylglycine-generating enzyme (FGE). The generated carbonyl moiety is addressed by a bifunctional linker with a pyrazolone for a tandem Knoevenagel reaction and an azide for strain-promoted azide-alkyne cycloaddition (SPAAC). The latter reaction with a PEGylated linker containing a dibenzocyclooctyne (DBCO) for SPAAC and monomethyl auristatin E (MMAE) as the toxin provided the stable conjugates DD1-MMAE (drug-antibody ratio, DAR = 2.0) and DFc-MMAE (DAR = 4.0) with sub-nanomolar cytotoxicity against the human squamous carcinoma derived A431 cells. In vivo imaging of Alexa Fluor 647-dye conjugates in A431-xenografted mice bearing subcutaneous tumors as the SCC model revealed unspecific binding of bivalent DARPins to the ubiquitously expressed EGFR. Tumor-targeting was verified 6 h post-injection solely for DD1 and scFvFc. The total of four administrations of 6.5 mg/kg DD1-MMAE or DFc-MMAE twice weekly did not cause any sequela in mice. MMAE conjugates showed no significant anti-tumor efficacy in vivo, but a trend towards increased necrotic areas (p = 0.2213) was observed for the DD1-MMAE (n = 5).
Collapse
|
16
|
Wu D, Yang K, Zhang Z, Feng Y, Rao L, Chen X, Yu G. Metal-free bioorthogonal click chemistry in cancer theranostics. Chem Soc Rev 2022; 51:1336-1376. [PMID: 35050284 DOI: 10.1039/d1cs00451d] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bioorthogonal chemistry is a powerful tool to site-specifically activate drugs in living systems. Bioorthogonal reactions between a pair of biologically reactive groups can rapidly and specifically take place in a mild physiological milieu without perturbing inherent biochemical processes. Attributed to their high selectivity and efficiency, bioorthogonal reactions can significantly decrease background signals in bioimaging. Compared with metal-catalyzed bioorthogonal click reactions, metal-free click reactions are more biocompatible without the metal catalyst-induced cytotoxicity. Although a great number of bioorthogonal chemistry-based strategies have been reported for cancer theranostics, a comprehensive review is scarce to highlight the advantages of these strategies. In this review, recent progress in cancer theranostics guided by metal-free bioorthogonal click chemistry will be depicted in detail. The elaborate design as well as the advantages of bioorthogonal chemistry in tumor theranostics are summarized and future prospects in this emerging field are emphasized.
Collapse
Affiliation(s)
- Dan Wu
- College of Materials Science and Engineering, Zhejiang University of Technology Hangzhou, 310014, P. R. China.
| | - Kuikun Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, P. R. China
| | - Zhankui Zhang
- College of Materials Science and Engineering, Zhejiang University of Technology Hangzhou, 310014, P. R. China.
| | - Yunxuan Feng
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China.
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, P. R. China.
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore.
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China.
| |
Collapse
|
17
|
Janson N, Heinks T, Beuel T, Alam S, Höhne M, Bornscheuer UT, Fischer von Mollard G, Sewald N. Efficient Site‐Selective Immobilization of Aldehyde‐Tagged Peptides and Proteins by Knoevenagel Ligation. ChemCatChem 2021. [DOI: 10.1002/cctc.202101485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Nils Janson
- Faculty of Chemistry Organic and Bioorganic Chemistry Bielefeld University Universitätsstraße 25 33615 Bielefeld Germany
| | - Tobias Heinks
- TFaculty of Chemistry, Biochemistry Bielefeld University Universitätsstraße 25 33615 Bielefeld Germany
| | - Tobias Beuel
- TFaculty of Chemistry, Biochemistry Bielefeld University Universitätsstraße 25 33615 Bielefeld Germany
| | - Sarfaraz Alam
- TFaculty of Chemistry, Biochemistry Bielefeld University Universitätsstraße 25 33615 Bielefeld Germany
| | - Matthias Höhne
- Institute of Biochemistry Greifswald University Felix-Hausdorff-Straße 4 17487 Greifswald Germay
| | - Uwe T. Bornscheuer
- Institute of Biochemistry Greifswald University Felix-Hausdorff-Straße 4 17487 Greifswald Germay
| | | | - Norbert Sewald
- Faculty of Chemistry Organic and Bioorganic Chemistry Bielefeld University Universitätsstraße 25 33615 Bielefeld Germany
| |
Collapse
|
18
|
Manicardi A, Cadoni E, Madder A. Hydrolysis of 5-methylfuran-2-yl to 2,5-dioxopentanyl allows for stable bio-orthogonal proximity-induced ligation. Commun Chem 2021; 4:146. [PMID: 36697666 PMCID: PMC9814669 DOI: 10.1038/s42004-021-00584-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/14/2021] [Indexed: 01/28/2023] Open
Abstract
Ligation methodologies featuring bio-orthogonal units and leading to the formation of a stable adduct are the ideal candidates for being applied in a biological context. However, most of the available strategies rely on highly reactive species that require careful handling, or on the activation of pro-reactive functional groups. We here report on a proximity-induced ligation reaction that relies on a stable 2,5-dione, that can be conveniently generated under acidic conditions from a 2,5-dialkylfuran building block, and hydrazine nucleophiles. This bio-orthogonal ligation, which proceeds under physiological conditions, does not require any stimulus or trigger and leads to the formation of a pyridazinium adduct that demonstrates excellent stability under harsh conditions (24 h at 90 °C). The reaction was applied to the formation of PNA-PNA adducts, DNA- and RNA-templated ligations, and for the formation of peptide-peptide adducts in solution. This convenient methodology was further implemented on plastic and glass surfaces to realize self-addressable covalent constructs.
Collapse
Affiliation(s)
- Alex Manicardi
- grid.5342.00000 0001 2069 7798Organic and Biomimetic Chemistry Research Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, 9000 Ghent, Belgium
| | - Enrico Cadoni
- grid.5342.00000 0001 2069 7798Organic and Biomimetic Chemistry Research Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, 9000 Ghent, Belgium
| | - Annemieke Madder
- grid.5342.00000 0001 2069 7798Organic and Biomimetic Chemistry Research Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, 9000 Ghent, Belgium
| |
Collapse
|
19
|
Abstract
Antibodies, particularly of the immunoglobulin G (IgG) isotype, are a group of biomolecules that are extensively used as affinity reagents for many applications in research, disease diagnostics, and therapy. Most of these applications require antibodies to be modified with specific functional moieties, including fluorophores, drugs, and proteins. Thus, a variety of methodologies have been developed for the covalent labeling of antibodies. The most common methods stably attach functional molecules to lysine or cysteine residues, which unavoidably results in heterogeneous products that cannot be further purified. In an effort to prepare homogeneous antibody conjugates, bioorthogonal handles have been site-specifically introduced via enzymatic treatment, genetic code expansion, or genetically encoded tagging, followed by functionalization using bioorthogonal conjugation reactions. The resulting homogeneous products have proven superior to their heterogeneous counterparts for both in vitro and in vivo usage. Nevertheless, additional chemical treatment or protein engineering of antibodies is required for incorporation of the bioorthogonal handles, processes that often affect antibody folding, stability, and/or production yield and cost. Accordingly, concurrent with advances in the fields of bioorthogonal chemistry and protein engineering, there is growing interest in site-specifically labeling native (nonengineered) antibodies without chemical or enzymatic treatments. In this review, we highlight recent strategies for producing site-specific native antibody conjugates and provide a comprehensive summary of the merits and disadvantages of these strategies.
Collapse
Affiliation(s)
- Kuan-Lin Wu
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Chenfei Yu
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Catherine Lee
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Chao Zuo
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Zachary T Ball
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Han Xiao
- Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
- Department of Biosciences, Rice University, 6100 Main Street, Houston, Texas 77005, United States
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| |
Collapse
|
20
|
Ahn G, Banik SM, Bertozzi CR. Degradation from the outside in: Targeting extracellular and membrane proteins for degradation through the endolysosomal pathway. Cell Chem Biol 2021; 28:1072-1080. [PMID: 33770486 PMCID: PMC8286304 DOI: 10.1016/j.chembiol.2021.02.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/17/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022]
Abstract
Targeted protein degradation (TPD) is a promising strategy to remove deleterious proteins for therapeutic benefit and to probe biological pathways. The past two decades have witnessed a surge in the development of technologies that rely on intracellular machinery to degrade challenging cytosolic targets. However, these TPD platforms leave the majority of extracellular and membrane proteins untouched. To enable degradation of these classes of proteins, internalizing receptors can be co-opted to traffic extracellular proteins to the lysosome. Sweeping antibodies and Seldegs use Fc receptors in conjunction with engineered antibodies to degrade soluble proteins. Recently, lysosome-targeting chimeras (LYTACs) have emerged as a strategy to degrade both secreted and membrane-anchored targets. Together with other newcomer technologies, including antibody-based proteolysis-targeting chimeras, modalities that degrade extracellular proteins have promising translational potential. This perspective will give an overview of TPD platforms that degrade proteins via outside-in approaches and focus on the recent development of LYTACs.
Collapse
Affiliation(s)
- Green Ahn
- Department of Chemistry and Stanford ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Steven M Banik
- Department of Chemistry and Stanford ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Carolyn R Bertozzi
- Department of Chemistry and Stanford ChEM-H, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford, CA 94305, USA.
| |
Collapse
|
21
|
Heiss TK, Dorn RS, Prescher JA. Bioorthogonal Reactions of Triarylphosphines and Related Analogues. Chem Rev 2021; 121:6802-6849. [PMID: 34101453 PMCID: PMC10064493 DOI: 10.1021/acs.chemrev.1c00014] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bioorthogonal phosphines were introduced in the context of the Staudinger ligation over 20 years ago. Since that time, phosphine probes have been used in myriad applications to tag azide-functionalized biomolecules. The Staudinger ligation also paved the way for the development of other phosphorus-based chemistries, many of which are widely employed in biological experiments. Several reviews have highlighted early achievements in the design and application of bioorthogonal phosphines. This review summarizes more recent advances in the field. We discuss innovations in classic Staudinger-like transformations that have enabled new biological pursuits. We also highlight relative newcomers to the bioorthogonal stage, including the cyclopropenone-phosphine ligation and the phospha-Michael reaction. The review concludes with chemoselective reactions involving phosphite and phosphonite ligations. For each transformation, we describe the overall mechanism and scope. We also showcase efforts to fine-tune the reagents for specific functions. We further describe recent applications of the chemistries in biological settings. Collectively, these examples underscore the versatility and breadth of bioorthogonal phosphine reagents.
Collapse
|
22
|
Abstract
Click chemistry has been established rapidly as one of the most valuable methods for the chemical transformation of complex molecules. Due to the rapid rates, clean conversions to the products, and compatibility of the reagents and reaction conditions even in complex settings, it has found applications in many molecule-oriented disciplines. From the vast landscape of click reactions, approaches have emerged in the past decade centered around oxidative processes to generate in situ highly reactive synthons from dormant functionalities. These approaches have led to some of the fastest click reactions know to date. Here, we review the various methods that can be used for such oxidation-induced "one-pot" click chemistry for the transformation of small molecules, materials, and biomolecules. A comprehensive overview is provided of oxidation conditions that induce a click reaction, and oxidation conditions are orthogonal to other click reactions so that sequential "click-oxidation-click" derivatization of molecules can be performed in one pot. Our review of the relevant literature shows that this strategy is emerging as a powerful approach for the preparation of high-performance materials and the generation of complex biomolecules. As such, we expect that oxidation-induced "one-pot" click chemistry will widen in scope substantially in the forthcoming years.
Collapse
Affiliation(s)
- Bauke Albada
- Laboratory of Organic Chemistry, Wageningen University & Research, Stippeneng 4, 6807 WE Wageningen, The Netherlands
| | - Jordi F Keijzer
- Laboratory of Organic Chemistry, Wageningen University & Research, Stippeneng 4, 6807 WE Wageningen, The Netherlands
| | - Han Zuilhof
- Laboratory of Organic Chemistry, Wageningen University & Research, Stippeneng 4, 6807 WE Wageningen, The Netherlands.,School of Pharmaceutical Sciences and Technology, Tianjin University, Tianjin 300072, China.,Department of Chemical and Materials Engineering, Faculty of Engineering, King Abdulaziz University, 21589 Jeddah, Saudi Arabia
| | - Floris van Delft
- Laboratory of Organic Chemistry, Wageningen University & Research, Stippeneng 4, 6807 WE Wageningen, The Netherlands.,Synaffix BV, Industrielaan 63, 5349 AE, Oss, The Netherlands
| |
Collapse
|
23
|
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: 4.7] [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
|
24
|
Scinto SL, Bilodeau DA, Hincapie R, Lee W, Nguyen SS, Xu M, am Ende CW, Finn MG, Lang K, Lin Q, Pezacki JP, Prescher JA, Robillard MS, Fox JM. Bioorthogonal chemistry. NATURE REVIEWS. METHODS PRIMERS 2021; 1:30. [PMID: 34585143 PMCID: PMC8469592 DOI: 10.1038/s43586-021-00028-z] [Citation(s) in RCA: 195] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/05/2021] [Indexed: 12/11/2022]
Abstract
Bioorthogonal chemistry represents a class of high-yielding chemical reactions that proceed rapidly and selectively in biological environments without side reactions towards endogenous functional groups. Rooted in the principles of physical organic chemistry, bioorthogonal reactions are intrinsically selective transformations not commonly found in biology. Key reactions include native chemical ligation and the Staudinger ligation, copper-catalysed azide-alkyne cycloaddition, strain-promoted [3 + 2] reactions, tetrazine ligation, metal-catalysed coupling reactions, oxime and hydrazone ligations as well as photoinducible bioorthogonal reactions. Bioorthogonal chemistry has significant overlap with the broader field of 'click chemistry' - high-yielding reactions that are wide in scope and simple to perform, as recently exemplified by sulfuryl fluoride exchange chemistry. The underlying mechanisms of these transformations and their optimal conditions are described in this Primer, followed by discussion of how bioorthogonal chemistry has become essential to the fields of biomedical imaging, medicinal chemistry, protein synthesis, polymer science, materials science and surface science. The applications of bioorthogonal chemistry are diverse and include genetic code expansion and metabolic engineering, drug target identification, antibody-drug conjugation and drug delivery. This Primer describes standards for reproducibility and data deposition, outlines how current limitations are driving new research directions and discusses new opportunities for applying bioorthogonal chemistry to emerging problems in biology and biomedicine.
Collapse
Affiliation(s)
- Samuel L. Scinto
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| | - Didier A. Bilodeau
- Department of Chemistry and Biomolecular Science, University of Ottawa, Ottawa, Ontario, Canada
- These authors contributed equally: Didier A. Bilodeau, Robert Hincapie, Wankyu Lee, Sean S. Nguyen, Minghao Xu
| | - Robert Hincapie
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
- These authors contributed equally: Didier A. Bilodeau, Robert Hincapie, Wankyu Lee, Sean S. Nguyen, Minghao Xu
| | - Wankyu Lee
- Pfizer Worldwide Research and Development, Cambridge, MA, USA
- These authors contributed equally: Didier A. Bilodeau, Robert Hincapie, Wankyu Lee, Sean S. Nguyen, Minghao Xu
| | - Sean S. Nguyen
- Department of Chemistry, University of California, Irvine, CA, USA
- These authors contributed equally: Didier A. Bilodeau, Robert Hincapie, Wankyu Lee, Sean S. Nguyen, Minghao Xu
| | - Minghao Xu
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
- These authors contributed equally: Didier A. Bilodeau, Robert Hincapie, Wankyu Lee, Sean S. Nguyen, Minghao Xu
| | | | - M. G. Finn
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Kathrin Lang
- Department of Chemistry, Technical University of Munich, Garching, Germany
- Laboratory of Organic Chemistry, ETH Zurich, Zurich, Switzerland
| | - Qing Lin
- Department of Chemistry, State University of New York at Buffalo, Buffalo, NY, USA
| | - John Paul Pezacki
- Department of Chemistry and Biomolecular Science, University of Ottawa, Ottawa, Ontario, Canada
| | - Jennifer A. Prescher
- Department of Chemistry, University of California, Irvine, CA, USA
- Molecular Biology & Biochemistry, University of California, Irvine, CA, USA
| | | | - Joseph M. Fox
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| |
Collapse
|
25
|
Detection of Genomic Uracil Patterns. Int J Mol Sci 2021; 22:ijms22083902. [PMID: 33918885 PMCID: PMC8070346 DOI: 10.3390/ijms22083902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/28/2021] [Accepted: 04/05/2021] [Indexed: 01/06/2023] Open
Abstract
The appearance of uracil in the deoxyuridine moiety of DNA is among the most frequently occurring genomic modifications. Three different routes can result in genomic uracil, two of which do not require specific enzymes: spontaneous cytosine deamination due to the inherent chemical reactivity of living cells, and thymine-replacing incorporation upon nucleotide pool imbalances. There is also an enzymatic pathway of cytosine deamination with multiple DNA (cytosine) deaminases involved in this process. In order to describe potential roles of genomic uracil, it is of key importance to utilize efficient uracil-DNA detection methods. In this review, we provide a comprehensive and critical assessment of currently available uracil detection methods with special focus on genome-wide mapping solutions. Recent developments in PCR-based and in situ detection as well as the quantitation of genomic uracil are also discussed.
Collapse
|
26
|
LYTACs that engage the asialoglycoprotein receptor for targeted protein degradation. Nat Chem Biol 2021; 17:937-946. [PMID: 33767387 PMCID: PMC8387313 DOI: 10.1038/s41589-021-00770-1] [Citation(s) in RCA: 222] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 02/10/2021] [Indexed: 02/06/2023]
Abstract
Selective protein degradation platforms have afforded new development opportunities for therapeutics and tools for biological inquiry. The first lysosome targeting chimeras (LYTACs) targeted extracellular and membrane proteins for degradation by bridging a target protein to the cation-independent mannose-6-phosphate receptor (CI-M6PR). Here, we developed LYTACs that engage the asialoglycoprotein receptor (ASGPR), a liver-specific lysosomal targeting receptor, to degrade extracellular proteins in a cell type-specific manner. We conjugated binders to a tri-GalNAc motif that engages ASGPR to drive downregulation of proteins. Degradation of EGFR by GalNAc-LYTAC attenuated EGFR signaling compared to inhibition with an antibody. Furthermore, we demonstrated that a LYTAC comprising a 3.4 kDa peptide binder linked to a tri-GalNAc ligand degrades integrins and reduces cancer cell proliferation. Degradation with a single tri-GalNAc ligand prompted site-specific conjugation on antibody scaffolds, which improved the pharmacokinetic profile of GalNAc-LYTACs in vivo. GalNAc-LYTACs thus represent an avenue for cell-type restricted protein degradation.
Collapse
|
27
|
Chuprakov S, Ogunkoya AO, Barfield RM, Bauzon M, Hickle C, Kim YC, Yeo D, Zhang F, Rabuka D, Drake PM. Tandem-Cleavage Linkers Improve the In Vivo Stability and Tolerability of Antibody-Drug Conjugates. Bioconjug Chem 2021; 32:746-754. [PMID: 33689309 DOI: 10.1021/acs.bioconjchem.1c00029] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Although peptide motifs represent the majority of cleavable linkers used in clinical-stage antibody-drug conjugates (ADCs), the sequences are often sensitive to cleavage by extracellular enzymes, such as elastase, which leads to systemic release of the cytotoxic payload. This action reduces the therapeutic index by causing off-target toxicities that can be dose-limiting. For example, a common side-effect of ADCs made using peptide-cleavable linkers is myelosuppression, including neutropenia. Only a few reports describe methods for optimizing peptide linkers to maintain efficient and potent tumor payload delivery while enhancing circulating stability. Herein, we address these critical limitations through the development of a tandem-cleavage linker strategy, where two sequential enzymatic cleavage events mediate payload release. We prepared dipeptides that are protected from degradation in the circulation by a sterically encumbering glucuronide moiety. Upon ADC internalization and lysosomal degradation, the monosaccharide is removed and the exposed dipeptide is degraded, which liberates the attached payload inside the target cell. We used CD79b-targeted monomethyl auristatin E (MMAE) conjugates as our model system and compared the stability, efficacy, and tolerability of ADCs made with tandem-cleavage linkers to ADCs made using standard technology with the vedotin linker. The results, where rat studies showed dramatically improved tolerability in the hematopoietic compartment, highlight the role that linker stability plays in efficacy and tolerability and also offer a means of improving an ADC's therapeutic index for improved patient outcomes.
Collapse
Affiliation(s)
- Stepan Chuprakov
- Catalent Pharma Solutions, 5959 Horton Street, Suite 400, Emeryville, California 94608, United States
| | - Ayodele O Ogunkoya
- Catalent Pharma Solutions, 5959 Horton Street, Suite 400, Emeryville, California 94608, United States
| | - Robyn M Barfield
- Catalent Pharma Solutions, 5959 Horton Street, Suite 400, Emeryville, California 94608, United States
| | - Maxine Bauzon
- Catalent Pharma Solutions, 5959 Horton Street, Suite 400, Emeryville, California 94608, United States
| | - Colin Hickle
- Catalent Pharma Solutions, 5959 Horton Street, Suite 400, Emeryville, California 94608, United States
| | - Yun Cheol Kim
- Catalent Pharma Solutions, 5959 Horton Street, Suite 400, Emeryville, California 94608, United States
| | - Dominick Yeo
- Catalent Pharma Solutions, 5959 Horton Street, Suite 400, Emeryville, California 94608, United States
| | - Fangjiu Zhang
- Catalent Pharma Solutions, 5959 Horton Street, Suite 400, Emeryville, California 94608, United States
| | - David Rabuka
- Catalent Pharma Solutions, 5959 Horton Street, Suite 400, Emeryville, California 94608, United States
| | - Penelope M Drake
- Catalent Pharma Solutions, 5959 Horton Street, Suite 400, Emeryville, California 94608, United States
| |
Collapse
|
28
|
Walsh SJ, Bargh JD, Dannheim FM, Hanby AR, Seki H, Counsell AJ, Ou X, Fowler E, Ashman N, Takada Y, Isidro-Llobet A, Parker JS, Carroll JS, Spring DR. Site-selective modification strategies in antibody-drug conjugates. Chem Soc Rev 2021; 50:1305-1353. [PMID: 33290462 DOI: 10.1039/d0cs00310g] [Citation(s) in RCA: 220] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antibody-drug conjugates (ADCs) harness the highly specific targeting capabilities of an antibody to deliver a cytotoxic payload to specific cell types. They have garnered widespread interest in drug discovery, particularly in oncology, as discrimination between healthy and malignant tissues or cells can be achieved. Nine ADCs have received approval from the US Food and Drug Administration and more than 80 others are currently undergoing clinical investigations for a range of solid tumours and haematological malignancies. Extensive research over the past decade has highlighted the critical nature of the linkage strategy adopted to attach the payload to the antibody. Whilst early generation ADCs were primarily synthesised as heterogeneous mixtures, these were found to have sub-optimal pharmacokinetics, stability, tolerability and/or efficacy. Efforts have now shifted towards generating homogeneous constructs with precise drug loading and predetermined, controlled sites of attachment. Homogeneous ADCs have repeatedly demonstrated superior overall pharmacological profiles compared to their heterogeneous counterparts. A wide range of methods have been developed in the pursuit of homogeneity, comprising chemical or enzymatic methods or a combination thereof to afford precise modification of specific amino acid or sugar residues. In this review, we discuss advances in chemical and enzymatic methods for site-specific antibody modification that result in the generation of homogeneous ADCs.
Collapse
Affiliation(s)
- Stephen J Walsh
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
|
30
|
Janson N, Krüger T, Karsten L, Boschanski M, Dierks T, Müller KM, Sewald N. Bifunctional Reagents for Formylglycine Conjugation: Pitfalls and Breakthroughs. Chembiochem 2020; 21:3580-3593. [PMID: 32767537 PMCID: PMC7756428 DOI: 10.1002/cbic.202000416] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/05/2020] [Indexed: 12/28/2022]
Abstract
Formylglycine-generating enzymes specifically oxidize cysteine within the consensus sequence CxPxR to Cα -formylglycine (FGly). This noncanonical electrophilic amino acid can subsequently be addressed selectively by bioorthogonal hydrazino-iso-Pictet-Spengler (HIPS) or Knoevenagel ligation to attach payloads like fluorophores or drugs to proteins to obtain a defined payload-to-protein ratio. However, the disadvantages of these conjugation techniques include the need for a large excess of conjugation building block, comparably low reaction rates and limited stability of FGly-containing proteins. Therefore, functionalized clickable HIPS and tandem Knoevenagel building blocks were synthesized, conjugated to small proteins (DARPins) and subsequently linked to strained alkyne-containing payloads for protein labeling. This procedure allowed the selective bioconjugation of one or two DBCO-carrying payloads with nearly stoichiometric amounts at low concentrations. Furthermore, an azide-modified tandem Knoevenagel building block enabled the synthesis of branched PEG linkers and the conjugation of two fluorophores, resulting in an improved signal-to-noise ratio in live-cell fluorescence-imaging experiments targeting the EGF receptor.
Collapse
Affiliation(s)
- Nils Janson
- Faculty of ChemistryOrganic and Bioorganic ChemistryBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| | - Tobias Krüger
- Faculty of ChemistryOrganic and Bioorganic ChemistryBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| | - Lennard Karsten
- Cellular and Molecular BiotechnologyBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| | - Mareile Boschanski
- Faculty of ChemistryBiochemistryBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| | - Thomas Dierks
- Faculty of ChemistryBiochemistryBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| | - Kristian M. Müller
- Cellular and Molecular BiotechnologyBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| | - Norbert Sewald
- Faculty of ChemistryOrganic and Bioorganic ChemistryBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| |
Collapse
|
31
|
Gray MA, Stanczak MA, Mantuano NR, Xiao H, Pijnenborg JFA, Malaker SA, Miller CL, Weidenbacher PA, Tanzo JT, Ahn G, Woods EC, Läubli H, Bertozzi CR. Targeted glycan degradation potentiates the anticancer immune response in vivo. Nat Chem Biol 2020; 16:1376-1384. [PMID: 32807964 PMCID: PMC7727925 DOI: 10.1038/s41589-020-0622-x] [Citation(s) in RCA: 196] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/08/2020] [Indexed: 12/24/2022]
Abstract
Currently approved immune checkpoint inhibitor therapies targeting the PD-1 and CTLA-4 receptor pathways are powerful treatment options for certain cancers; however, most patients across cancer types still fail to respond. Consequently, there is interest in discovering and blocking alternative pathways that mediate immune suppression. One such mechanism is an upregulation of sialoglycans in malignancy, which has been recently shown to inhibit immune cell activation through multiple mechanisms and therefore represents a targetable glycoimmune checkpoint. Since these glycans are not canonically druggable, we designed an αHER2 antibody-sialidase conjugate that potently and selectively strips diverse sialoglycans from breast cancer cells. In syngeneic breast cancer models, desialylation enhanced immune cell infiltration and activation and prolonged the survival of mice, an effect that was dependent on expression of the Siglec-E checkpoint receptor found on tumor-infiltrating myeloid cells. Thus, antibody-sialidase conjugates represent a promising modality for glycoimmune checkpoint therapy.
Collapse
MESH Headings
- Allografts
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/metabolism
- B7-H1 Antigen/genetics
- B7-H1 Antigen/immunology
- Cell Line, Tumor
- Humans
- Hydrolysis
- Immunoconjugates/chemistry
- Immunoconjugates/metabolism
- Immunoconjugates/pharmacology
- Immunotherapy/methods
- Killer Cells, Natural/cytology
- Killer Cells, Natural/immunology
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/mortality
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Molecular
- Molecular Targeted Therapy
- Neuraminidase/chemistry
- Neuraminidase/genetics
- Neuraminidase/immunology
- Polysaccharides/chemistry
- Polysaccharides/immunology
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/immunology
- Protein Binding
- Protein Interaction Domains and Motifs
- Protein Structure, Secondary
- Receptor, ErbB-2/chemistry
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Sialic Acid Binding Immunoglobulin-like Lectins/chemistry
- Sialic Acid Binding Immunoglobulin-like Lectins/genetics
- Sialic Acid Binding Immunoglobulin-like Lectins/immunology
- Survival Analysis
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Melissa A Gray
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Michal A Stanczak
- Cancer Immunology Laboratory, Department of Biomedicine, University Hospital, Basel, Switzerland
- Division of Oncology, Department of Internal Medicine, University Hospital, Basel, Switzerland
| | - Natália R Mantuano
- Cancer Immunology Laboratory, Department of Biomedicine, University Hospital, Basel, Switzerland
- Division of Oncology, Department of Internal Medicine, University Hospital, Basel, Switzerland
| | - Han Xiao
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | | | - Stacy A Malaker
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Caitlyn L Miller
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | - Julia T Tanzo
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Green Ahn
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Elliot C Woods
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Heinz Läubli
- Cancer Immunology Laboratory, Department of Biomedicine, University Hospital, Basel, Switzerland
- Division of Oncology, Department of Internal Medicine, University Hospital, Basel, Switzerland
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
32
|
Wang W, Han N, Xu Y, Zhao Y, Shi L, Filmus J, Li F. Assembling custom side chains on proteoglycans to interrogate their function in living cells. Nat Commun 2020; 11:5915. [PMID: 33219207 PMCID: PMC7679400 DOI: 10.1038/s41467-020-19765-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Proteoglycans (PGs) are composed of a core protein and one or more chains of glycosaminoglycans (GAGs). The highly heterogeneous GAG chains play an irreplaceable role in the functions of PGs. However, the lack of an approach to control the exact structure of GAG chains conjugated to PGs tremendously hinders functional studies of PGs. Herein, by using glypican-3 as a model, we establish an aldehyde tag-based approach to assemble PGs with specific GAG chains on the surface of living cells. We show that the engineered glypican-3 can regulate Wnt and Hedgehog signaling like the wild type. Furthermore, we also present a method for studying the interaction of PGs with their target glycoproteins by combining the assembly of PGs carrying specific GAG chains with metabolic glycan labeling, and most importantly, we obtain evidence of GPC3 directly interacting with Frizzled. In conclusion, this study provides a very useful platform for structural and functional studies of PGs with specific GAG chains. Currently, it is not possible to generate proteoglycans displaying glycosaminoglycan chains with specific structures. Here the authors show that by using an aldehyde tag-based methodology it is possible to insert these specific chains onto proteoglycans expressed on the cell surface.
Collapse
Affiliation(s)
- Wenshuang Wang
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, China
| | - Naihan Han
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, China.,Shandong Police College, Jinan, China
| | - Yingying Xu
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, China
| | - Yunxue Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Liran Shi
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, China
| | - Jorge Filmus
- Sunnybrook Health Science Centre, University of Toronto, Toronto, Ontario, Canada
| | - Fuchuan Li
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, China.
| |
Collapse
|
33
|
Mailig M, Hymel D, Liu F. Further Exploration of Hydrazine-Mediated Bioconjugation Chemistries. Org Lett 2020; 22:6677-6681. [PMID: 32786214 DOI: 10.1021/acs.orglett.0c02545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The hydrazine group serves as a great anchor for bioconjugation; however, the application of hydrazone ligation has been limited by poor product stability. We aim to resolve such issues by optimizing the recently established pyrazolone ligation and investigating a new pyrazole ligation. We have identified a new, electron-deficient pyrazolone ligation and a regiospecific pyrazole ligation, both offering aqueous buffer stable and chemically inert products possessing triazole-like structures while not involving any heavy metal catalyst.
Collapse
Affiliation(s)
- Melrose Mailig
- Novo Nordisk Research Center, 530 Fairview Avenue North, Seattle, Washington 98109, United States
| | - David Hymel
- Novo Nordisk Research Center, 530 Fairview Avenue North, Seattle, Washington 98109, United States
| | - Fa Liu
- Novo Nordisk Research Center, 530 Fairview Avenue North, Seattle, Washington 98109, United States
| |
Collapse
|
34
|
Mao X, Li W, Zhu S, Zou J, Tian H, Duan Y, Wang Y, Fei J, Wang X. Bifunctional pyridoxal derivatives as efficient bioorthogonal reagents for biomacromolecule modifications. Chem Commun (Camb) 2020; 56:7601-7604. [PMID: 32514510 DOI: 10.1039/d0cc02722g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Two types of pyridoxal analogs, azido pyridoxal (PL-N3) and carboxyl pyridoxal (PL-COOH), were developed as novel bifunctional bioorthogonal molecules. These molecules showed fast imine formation with hydrazinyl groups and stable covalent linkages via azido/carboxyl groups, and thus were of great use for site-specific peptide and protein modifications.
Collapse
Affiliation(s)
- Xianxian Mao
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Bioconjugation chemistries are critical tools in biotherapeutics discovery. The past efforts have been exclusively focused on two-segment conjugations. However, emerging research directions, such as polypharmacy biotherapeutics, desire multiple-component bioconjugations where more than two pharmacologically related biomolecules can be assembled into a single construct in high efficiency. We present here a set of sequential bioconjugation chemistries centered on a pyrazolone structural motif. It starts with a clickable “pyrazolone ligation” between a hydrazine group and a β-ketoester moiety followed by the conjugation between the newly formed pyrazolone core and an aldehyde-bearing biomolecule through a Knoevenagel reaction forming a Michael addition acceptor that can effectively capture a thiol-bearing biomolecule. When utilized intermolecularly, it quickly assembles four segments together forming a quadruple functional construct. When applied intramolecularly, it offers a set of highly diverse biomolecule scaffolds including stapled peptides and poly-macrocyclic peptides. We envision broad utilities of such sequential ligation chemistries. A multiple component sequential bioconjugation chemistry establishes upon the joined force of hydrazine, β-keto ester, thiol and aldehyde.![]()
Collapse
Affiliation(s)
- Melrose Mailig
- Novo Nordisk Research Center 530 Fairview Avenue North Seattle WA 98109 USA
| | - Fa Liu
- Novo Nordisk Research Center 530 Fairview Avenue North Seattle WA 98109 USA
| |
Collapse
|
36
|
Liu J, Barfield RM, Rabuka D. Site-Specific Bioconjugation Using SMARTag ® Technology: A Practical and Effective Chemoenzymatic Approach to Generate Antibody-Drug Conjugates. Methods Mol Biol 2020; 2033:131-147. [PMID: 31332752 DOI: 10.1007/978-1-4939-9654-4_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
As a critical feature of the next generation of antibody-drug conjugates (ADCs), site-specific bioconjugation approaches can help to optimize stability, pharmacokinetics, efficacy, and safety as well as improve manufacturing consistency. The SMARTag® technology platform offers a practical and efficient chemoenzymatic solution for site-specific protein modifications. A bioorthogonal aldehyde handle is introduced through the oxidation of a cysteine residue, embedded in a specific peptide sequence (CxPxR), to the aldehyde-bearing formylglycine (fGly). This enzymatic modification is carried out by the formylglycine-generating enzyme (FGE). The broad recognition of this short sequence by FGE within the context of heterologous proteins allows for the introduction of fGly residues at chosen sites in proteins expressed in prokaryotic and eukaryotic systems. The protocol presented here describes the methods for expressing fGly-containing antibodies in eukaryotic cells and subsequent site-specific conjugation with a payload-linker using aldehyde-specific Hydrazino-Iso-Pictet-Spengler (HIPS) chemistry.
Collapse
Affiliation(s)
- Junjie Liu
- Catalent Pharma Solutions, Richmond, CA, USA
| | | | | |
Collapse
|
37
|
Gauzy-Lazo L, Sassoon I, Brun MP. Advances in Antibody–Drug Conjugate Design: Current Clinical Landscape and Future Innovations. SLAS DISCOVERY 2020; 25:843-868. [DOI: 10.1177/2472555220912955] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The targeted delivery of potent cytotoxic molecules into cancer cells is considered a promising anticancer strategy. The design of clinically effective antibody–drug conjugates (ADCs), in which biologically active drugs are coupled through chemical linkers to monoclonal antibodies, has presented challenges for pharmaceutical researchers. After 30 years of intensive research and development activities, only seven ADCs have been approved for clinical use; two have received fast-track designation and two breakthrough therapy designation from the Food and Drug Administration. There is continued interest in the field, as documented by the growing number of candidates in clinical development. This review aims to summarize the most recent innovations that have been applied to the design of ADCs undergoing early- and late-stage clinical trials. Discovery and rational optimization of new payloads, chemical linkers, and antibody formats have improved the therapeutic index of next-generation ADCs, ultimately resulting in improved clinical benefit for the patients.
Collapse
Affiliation(s)
| | - Ingrid Sassoon
- Immuno-Oncology Therapeutic Area, Sanofi, Vitry-sur-Seine, France
| | | |
Collapse
|
38
|
Bispecific Antibodies and Antibody-Drug Conjugates for Cancer Therapy: Technological Considerations. Biomolecules 2020; 10:biom10030360. [PMID: 32111076 PMCID: PMC7175114 DOI: 10.3390/biom10030360] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/20/2020] [Accepted: 02/23/2020] [Indexed: 01/07/2023] Open
Abstract
The ability of monoclonal antibodies to specifically bind a target antigen and neutralize or stimulate its activity is the basis for the rapid growth and development of the therapeutic antibody field. In recent years, traditional immunoglobulin antibodies have been further engineered for better efficacy and safety, and technological developments in the field enabled the design and production of engineered antibodies capable of mediating therapeutic functions hitherto unattainable by conventional antibody formats. Representative of this newer generation of therapeutic antibody formats are bispecific antibodies and antibody–drug conjugates, each with several approved drugs and dozens more in the clinical development phase. In this review, the technological principles and challenges of bispecific antibodies and antibody–drug conjugates are discussed, with emphasis on clinically validated formats but also including recent developments in the fields, many of which are expected to significantly augment the current therapeutic arsenal against cancer and other diseases with unmet medical needs.
Collapse
|
39
|
Calcaterra A, Mangiardi L, Delle Monache G, Quaglio D, Balducci S, Berardozzi S, Iazzetti A, Franzini R, Botta B, Ghirga F. The Pictet-Spengler Reaction Updates Its Habits. Molecules 2020; 25:E414. [PMID: 31963860 PMCID: PMC7024544 DOI: 10.3390/molecules25020414] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/05/2020] [Accepted: 01/09/2020] [Indexed: 12/31/2022] Open
Abstract
The Pictet-Spengler reaction (P-S) is one of the most direct, efficient, and variable synthetic method for the construction of privileged pharmacophores such as tetrahydro-isoquinolines (THIQs), tetrahydro-β-carbolines (THBCs), and polyheterocyclic frameworks. In the lustro (five-year period) following its centenary birthday, the P-S reaction did not exit the stage but it came up again on limelight with new features. This review focuses on the interesting results achieved in this period (2011-2015), analyzing the versatility of this reaction. Classic P-S was reported in the total synthesis of complex alkaloids, in combination with chiral catalysts as well as for the generation of libraries of compounds in medicinal chemistry. The P-S has been used also in tandem reactions, with the sequences including ring closing metathesis, isomerization, Michael addition, and Gold- or Brønsted acid-catalyzed N-acyliminium cyclization. Moreover, the combination of P-S reaction with Ugi multicomponent reaction has been exploited for the construction of highly complex polycyclic architectures in few steps and high yields. The P-S reaction has also been successfully employed in solid-phase synthesis, affording products with different structures, including peptidomimetics, synthetic heterocycles, and natural compounds. Finally, the enzymatic version of P-S has been reported for biosynthesis, biotransformations, and bioconjugations.
Collapse
Affiliation(s)
- Andrea Calcaterra
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (L.M.); (G.D.M.); (D.Q.); (S.B.); (A.I.); (R.F.); (B.B.)
| | - Laura Mangiardi
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (L.M.); (G.D.M.); (D.Q.); (S.B.); (A.I.); (R.F.); (B.B.)
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy;
| | - Giuliano Delle Monache
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (L.M.); (G.D.M.); (D.Q.); (S.B.); (A.I.); (R.F.); (B.B.)
| | - Deborah Quaglio
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (L.M.); (G.D.M.); (D.Q.); (S.B.); (A.I.); (R.F.); (B.B.)
| | - Silvia Balducci
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (L.M.); (G.D.M.); (D.Q.); (S.B.); (A.I.); (R.F.); (B.B.)
| | - Simone Berardozzi
- Department of Chemistry and Applied Biosciences, ETH-Zürich, Vladimir-Prelog Weg 4, 8093 Zürich, Switzerland
| | - Antonia Iazzetti
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (L.M.); (G.D.M.); (D.Q.); (S.B.); (A.I.); (R.F.); (B.B.)
| | - Roberta Franzini
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (L.M.); (G.D.M.); (D.Q.); (S.B.); (A.I.); (R.F.); (B.B.)
| | - Bruno Botta
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (L.M.); (G.D.M.); (D.Q.); (S.B.); (A.I.); (R.F.); (B.B.)
| | - Francesca Ghirga
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy;
| |
Collapse
|
40
|
Peng Q, Zang B, Zhao W, Li D, Ren J, Ji F, Jia L. Efficient continuous-flow aldehyde tag conversion using immobilized formylglycine generating enzyme. Catal Sci Technol 2020. [DOI: 10.1039/c9cy01856e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Immobilized formylglycine generating enzyme for efficient aldehyde tag conversion under continuous flow conditions.
Collapse
Affiliation(s)
- Qiang Peng
- Liaoning Key Laboratory of Molecular Recognition and Imaging
- School of Life Science and Biotechnology
- Dalian University of Technology
- Dalian
- P. R. China
| | - Berlin Zang
- Liaoning Key Laboratory of Molecular Recognition and Imaging
- School of Life Science and Biotechnology
- Dalian University of Technology
- Dalian
- P. R. China
| | - Wei Zhao
- Liaoning Key Laboratory of Molecular Recognition and Imaging
- School of Life Science and Biotechnology
- Dalian University of Technology
- Dalian
- P. R. China
| | - Da Li
- Liaoning Key Laboratory of Molecular Recognition and Imaging
- School of Life Science and Biotechnology
- Dalian University of Technology
- Dalian
- P. R. China
| | - Jun Ren
- Liaoning Key Laboratory of Molecular Recognition and Imaging
- School of Life Science and Biotechnology
- Dalian University of Technology
- Dalian
- P. R. China
| | - Fangling Ji
- Liaoning Key Laboratory of Molecular Recognition and Imaging
- School of Life Science and Biotechnology
- Dalian University of Technology
- Dalian
- P. R. China
| | - Lingyun Jia
- Liaoning Key Laboratory of Molecular Recognition and Imaging
- School of Life Science and Biotechnology
- Dalian University of Technology
- Dalian
- P. R. China
| |
Collapse
|
41
|
Abstract
Click chemistry has found wide application in bioconjugation, enabling control over the site of modification in biomolecules. Demonstrations of this chemistry to construct chemically defined antibody-drug conjugates (ADCs) have increased in recent years, following studies that support benefits of homogeneity and site-specificity of drug placement on the antibody. In this chapter, a brief history of early applications of this chemistry in ADCs is presented. Examples of click chemistries that are utilized for ADC synthesis, including those currently undergoing clinical investigations, are enumerated. Protocols for two common conjugation methods based on carbonyl-aminooxy coupling and strain-promoted azide-alkyne cycloaddition are described.
Collapse
|
42
|
Harel ET, Drake PM, Barfield RM, Lui I, Farr-Jones S, Van’t Veer L, Gartner ZJ, Green EM, Lourenço AL, Cheng Y, Hann BC, Rabuka D, Craik CS. Antibody-Drug Conjugates Targeting the Urokinase Receptor (uPAR) as a Possible Treatment of Aggressive Breast Cancer. Antibodies (Basel) 2019; 8:antib8040054. [PMID: 31694242 PMCID: PMC6963874 DOI: 10.3390/antib8040054] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/20/2019] [Accepted: 10/23/2019] [Indexed: 12/22/2022] Open
Abstract
A promising molecular target for aggressive cancers is the urokinase receptor (uPAR). A fully human, recombinant antibody that binds uPAR to form a stable complex that blocks uPA-uPAR interactions (2G10) and is internalized primarily through endocytosis showed efficacy in a mouse xenograft model of highly aggressive, triple negative breast cancer (TNBC). Antibody-drug conjugates (ADCs) of 2G10 were designed and produced bearing tubulin inhibitor payloads ligated through seven different linkers. Aldehyde tag technology was employed for linking, and either one or two tags were inserted into the antibody heavy chain, to produce site-specifically conjugated ADCs with drug-to-antibody ratios of either two or four. Both cleavable and non-cleavable linkers were combined with two different antimitotic toxins—MMAE (monomethylauristatin E) and maytansine. Nine different 2G10 ADCs were produced and tested for their ability to target uPAR in cell-based assays and a mouse model. The anti-uPAR ADC that resulted in tumor regression comprised an MMAE payload with a cathepsin B cleavable linker, 2G10-RED-244-MMAE. This work demonstrates in vitro activity of the 2G10-RED-244-MMAE in TNBC cell lines and validates uPAR as a therapeutic target for TNBC.
Collapse
Affiliation(s)
- Efrat T. Harel
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA; (E.T.H.); (I.L.); (Z.J.G.); (A.L.L.)
| | - Penelope M. Drake
- Catalent Biologics, West, Emeryville, CA 94608, USA; (P.M.D.); (R.M.B.); (D.R.)
| | - Robyn M. Barfield
- Catalent Biologics, West, Emeryville, CA 94608, USA; (P.M.D.); (R.M.B.); (D.R.)
| | - Irene Lui
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA; (E.T.H.); (I.L.); (Z.J.G.); (A.L.L.)
| | - Shauna Farr-Jones
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94110, USA;
| | - Laura Van’t Veer
- Laboratory Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA;
| | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA; (E.T.H.); (I.L.); (Z.J.G.); (A.L.L.)
| | - Evan M. Green
- Biophysics Graduate Program and Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA;
| | - André Luiz Lourenço
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA; (E.T.H.); (I.L.); (Z.J.G.); (A.L.L.)
| | - Yifan Cheng
- Howard Hughes Medical Institute, University of California San Francisco, and Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA;
| | - Byron C. Hann
- Preclinical Therapeutics Core, UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA;
| | - David Rabuka
- Catalent Biologics, West, Emeryville, CA 94608, USA; (P.M.D.); (R.M.B.); (D.R.)
| | - Charles S. Craik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA; (E.T.H.); (I.L.); (Z.J.G.); (A.L.L.)
- Correspondence: ; Tel.: +1-415-476-8146
| |
Collapse
|
43
|
Hernandez-Alba O, Houel S, Hessmann S, Erb S, Rabuka D, Huguet R, Josephs J, Beck A, Drake PM, Cianférani S. A Case Study to Identify the Drug Conjugation Site of a Site-Specific Antibody-Drug-Conjugate Using Middle-Down Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2019; 30:2419-2429. [PMID: 31429052 DOI: 10.1007/s13361-019-02296-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 06/10/2023]
Abstract
Middle-down mass spectrometry (MD MS) has emerged as a promising alternative to classical bottom-up approaches for protein characterization. Middle-level experiments after enzymatic digestion are routinely used for subunit analysis of monoclonal antibody (mAb)-related compounds, providing information on drug load distribution and average drug-to-antibody ratio (DAR). However, peptide mapping is still the gold standard for primary amino acid sequence assessment, post-translational modifications (PTM), and drug conjugation identification and localization. However, peptide mapping strategies can be challenging when dealing with more complex and heterogeneous mAb formats, like antibody-drug conjugates (ADCs). We report here, for the first time, MD MS analysis of a third-generation site-specific DAR4 ADC using different fragmentation techniques, including higher-energy collisional- (HCD), electron-transfer (ETD) dissociation and 213 nm ultraviolet photodissociation (UVPD). UVPD used as a standalone technique for ADC subunit analysis afforded, within the same liquid chromatography-MS/MS run, enhanced performance in terms of primary sequence coverage compared to HCD- or ETD-based MD approaches, and generated substantially more MS/MS fragments containing either drug conjugation or glycosylation site information, leading to confident drug/glycosylation site identification. In addition, our results highlight the complementarity of ETD and UVPD for both primary sequence validation and drug conjugation/glycosylation site assessment. Altogether, our results highlight the potential of UVPD for ADC MD MS analysis for drug conjugation/glycosylation site assessment, and indicate that MD MS strategies can improve structural characterization of empowered next-generation mAb-based formats, especially for PTMs and drug conjugation sites validation.
Collapse
Affiliation(s)
- Oscar Hernandez-Alba
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS IPHC UMR 7178, Université de Strasbourg, ECPM R5-0 - 25 Rue Becquerel, Cedex 2, 67087, Strasbourg, France
| | - Stéphane Houel
- Thermo Fisher Scientific, 355 River Oaks Pkwy, San Jose, CA, 95134, USA
| | - Steve Hessmann
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS IPHC UMR 7178, Université de Strasbourg, ECPM R5-0 - 25 Rue Becquerel, Cedex 2, 67087, Strasbourg, France
| | - Stéphane Erb
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS IPHC UMR 7178, Université de Strasbourg, ECPM R5-0 - 25 Rue Becquerel, Cedex 2, 67087, Strasbourg, France
| | - David Rabuka
- Catalent Biologics West, 5703 Hollis Street, Emeryville, CA, 94530, USA
| | - Romain Huguet
- Thermo Fisher Scientific, 355 River Oaks Pkwy, San Jose, CA, 95134, USA
| | - Jonathan Josephs
- Thermo Fisher Scientific, 355 River Oaks Pkwy, San Jose, CA, 95134, USA
| | - Alain Beck
- IRPF, Centre d'Immunologie Pierre-Fabre (CIPF), Saint-Julien-en-Genevois, France
| | - Penelope M Drake
- Catalent Biologics West, 5703 Hollis Street, Emeryville, CA, 94530, USA
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS IPHC UMR 7178, Université de Strasbourg, ECPM R5-0 - 25 Rue Becquerel, Cedex 2, 67087, Strasbourg, France.
| |
Collapse
|
44
|
Walker JA, Sorkin MR, Ledesma F, Kabaria SR, Barfield RM, Rabuka D, Alabi CA. Hydrophilic Sequence-Defined Cross-Linkers for Antibody–Drug Conjugates. Bioconjug Chem 2019; 30:2982-2988. [DOI: 10.1021/acs.bioconjchem.9b00713] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Joshua A. Walker
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 113 Ho Plaza, Ithaca, New York 14850, United States
| | - Michelle R. Sorkin
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 113 Ho Plaza, Ithaca, New York 14850, United States
| | - Francis Ledesma
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 113 Ho Plaza, Ithaca, New York 14850, United States
| | - Sneha R. Kabaria
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 113 Ho Plaza, Ithaca, New York 14850, United States
| | - Robyn M. Barfield
- Catalent Biologics, 5703 Hollis Street, Emeryville, California 94608, United States
| | - David Rabuka
- Catalent Biologics, 5703 Hollis Street, Emeryville, California 94608, United States
| | - Christopher A. Alabi
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 113 Ho Plaza, Ithaca, New York 14850, United States
| |
Collapse
|
45
|
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: 2.4] [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
|
46
|
CRISPR-Cas9 screens identify regulators of antibody-drug conjugate toxicity. Nat Chem Biol 2019; 15:949-958. [PMID: 31451760 DOI: 10.1038/s41589-019-0342-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 07/09/2019] [Indexed: 01/01/2023]
Abstract
Antibody-drug conjugates (ADCs) selectively deliver chemotherapeutic agents to target cells and are important cancer therapeutics. However, the mechanisms by which ADCs are internalized and activated remain unclear. Using CRISPR-Cas9 screens, we uncover many known and novel endolysosomal regulators as modulators of ADC toxicity. We identify and characterize C18ORF8/RMC1 as a regulator of ADC toxicity through its role in endosomal maturation. Through comparative analysis of screens with ADCs bearing different linkers, we show that a subset of late endolysosomal regulators selectively influence toxicity of noncleavable linker ADCs. Surprisingly, we find cleavable valine-citrulline linkers can be processed rapidly after internalization without lysosomal delivery. Lastly, we show that sialic acid depletion enhances ADC lysosomal delivery and killing in diverse cancer cell types, including with FDA (US Food and Drug Administration)-approved trastuzumab emtansine (T-DM1) in Her2-positive breast cancer cells. Together, these results reveal new regulators of endolysosomal trafficking, provide important insights for ADC design and identify candidate combination therapy targets.
Collapse
|
47
|
Krüger T, Weiland S, Boschanski M, Sinha PK, Falck G, Müller KM, Dierks T, Sewald N. Conversion of Serine‐Type Aldehyde Tags by the Radical SAM Protein AtsB from
Methanosarcina mazei. Chembiochem 2019; 20:2074-2078. [DOI: 10.1002/cbic.201900322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Indexed: 01/09/2023]
Affiliation(s)
- Tobias Krüger
- Organische und Bioorganische ChemieFakultät für ChemieUniversität Bielefeld Universitätsstrasse 25 33615 Bielefeld Germany
| | - Stefanie Weiland
- Biochemie IFakultät für ChemieUniversität Bielefeld Universitätsstrasse 25 33615 Bielefeld Germany
| | - Mareile Boschanski
- Biochemie IFakultät für ChemieUniversität Bielefeld Universitätsstrasse 25 33615 Bielefeld Germany
| | - Prem Kumar Sinha
- Biochemie IFakultät für ChemieUniversität Bielefeld Universitätsstrasse 25 33615 Bielefeld Germany
| | - Georg Falck
- Zelluläre und Molekulare BiotechnologieTechnische FakultätUniversität Bielefeld Universitätsstrasse 25 33615 Bielefeld Germany
| | - Kristian M. Müller
- Zelluläre und Molekulare BiotechnologieTechnische FakultätUniversität Bielefeld Universitätsstrasse 25 33615 Bielefeld Germany
| | - Thomas Dierks
- Biochemie IFakultät für ChemieUniversität Bielefeld Universitätsstrasse 25 33615 Bielefeld Germany
| | - Norbert Sewald
- Organische und Bioorganische ChemieFakultät für ChemieUniversität Bielefeld Universitätsstrasse 25 33615 Bielefeld Germany
| |
Collapse
|
48
|
Yamada K, Ito Y. Recent Chemical Approaches for Site‐Specific Conjugation of Native Antibodies: Technologies toward Next‐Generation Antibody–Drug Conjugates. Chembiochem 2019; 20:2729-2737. [DOI: 10.1002/cbic.201900178] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Indexed: 11/09/2022]
Affiliation(s)
- Kei Yamada
- Ajinomoto Co., Inc. 1-1 Suzuki-Cho Kawasaki-Ku Kawasaki-Shi Kanagawa 210-8681 Japan
- Department of Chemistry and BioscienceGraduate School of Science and EngineeringKagoshima University 1-21-35 Korimoto Kagoshima 890-0065 Japan
| | - Yuji Ito
- Department of Chemistry and BioscienceGraduate School of Science and EngineeringKagoshima University 1-21-35 Korimoto Kagoshima 890-0065 Japan
| |
Collapse
|
49
|
Liu ZJ, Martínez Cuesta S, van Delft P, Balasubramanian S. Sequencing abasic sites in DNA at single-nucleotide resolution. Nat Chem 2019; 11:629-637. [PMID: 31209299 PMCID: PMC6589398 DOI: 10.1038/s41557-019-0279-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 04/30/2019] [Indexed: 12/27/2022]
Abstract
In DNA, the loss of a nucleobase by hydrolysis generates an abasic site. Formed as a result of DNA damage, as well as a key intermediate during the base excision repair pathway, abasic sites are frequent DNA lesions that can lead to mutations and strand breaks. Here we present snAP-seq, a chemical approach that selectively exploits the reactive aldehyde moiety at abasic sites to reveal their location within DNA at single-nucleotide resolution. Importantly, the approach resolves abasic sites from other aldehyde functionalities known to exist in genomic DNA. snAP-seq was validated on synthetic DNA and then applied to two separate genomes. We studied the distribution of thymine modifications in the Leishmania major genome by enzymatically converting these modifications into abasic sites followed by abasic site mapping. We also applied snAP-seq directly to HeLa DNA to provide a map of endogenous abasic sites in the human genome.
Collapse
Affiliation(s)
- Zheng J Liu
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Sergio Martínez Cuesta
- Department of Chemistry, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Shankar Balasubramanian
- Department of Chemistry, University of Cambridge, Cambridge, UK.
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
- School of Clinical Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
50
|
Mathaes R, Mahler HC. Next Generation Biopharmaceuticals: Product Development. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2019; 165:253-276. [PMID: 29637223 DOI: 10.1007/10_2016_57] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Therapeutic proteins show a rapid market growth. The relatively young biotech industry already represents 20 % of the total global pharma market. The biotech industry environment has traditionally been fast-pasted and intellectually stimulated. Nowadays the top ten best selling drugs are dominated by monoclonal antibodies (mABs).Despite mABs being the biggest medical breakthrough in the last 25 years, technical innovation does not stand still.The goal remains to preserve the benefits of a conventional mAB (serum half-life and specificity) whilst further improving efficacy and safety and to open new and better avenues for treating patients, e.g., improving the potency of molecules, target binding, tissue penetration, tailored pharmacokinetics, and reduced adverse effects or immunogenicity.The next generation of biopharmaceuticals can pose specific chemistry, manufacturing, and control (CMC) challenges. In contrast to conventional proteins, next-generation biopharmaceuticals often require lyophilization of the final drug product to ensure storage stability over shelf-life time. In addition, next-generation biopharmaceuticals require analytical methods that cover different ways of possible degradation patterns and pathways, and product development is a long way from being straight forward. The element of "prior knowledge" does not exist equally for most novel formats compared to antibodies, and thus the assessment of critical quality attributes (CQAs) and the definition of CQA assessment criteria and specifications is difficult, especially in early-stage development.
Collapse
Affiliation(s)
- Roman Mathaes
- Drug Product Services, Lonza AG, Münchensteiner Strasse 38, 4002, Basel, Switzerland.
| | | |
Collapse
|