1
|
Tao J, Gu Y, Zhou W, Wang Y. Dual-payload antibody-drug conjugates: Taking a dual shot. Eur J Med Chem 2024; 281:116995. [PMID: 39481229 DOI: 10.1016/j.ejmech.2024.116995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024]
Abstract
Antibody-drug conjugates (ADCs) enable the precise delivery of cytotoxic agents by conjugating small-molecule drugs with monoclonal antibodies (mAbs). Over recent decades, ADCs have demonstrated substantial clinical efficacy. However, conventional ADCs often encounter various clinical challenges, including suboptimal efficacy, significant adverse effects, and the development of drug resistance, limiting their broader clinical application. Encouragingly, a next-generation approach-dual-payload ADCs-has emerged as a pioneering strategy to address these challenges. Dual-payload ADCs are characterized by the incorporation of two distinct therapeutic payloads on the same antibody, enhancing treatment efficacy by promoting synergistic effects and reducing the risk of drug resistance. However, the synthesis of dual-payload ADCs is complex due to the presence of multiple functional groups on antibodies. In this review, we comprehensively summarize the construction strategies for dual-payload ADCs, ranging from the design of ADC components to orthogonal chemistry. The subsequent sections explore current challenges and propose prospective strategies, highlighting recent advancements in dual-payload ADC research, thereby laying the foundation for the development of next-generation ADCs.
Collapse
Affiliation(s)
- Junjie Tao
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yilin Gu
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wei Zhou
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China.
| | - Yuxi Wang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| |
Collapse
|
2
|
Chen Y, Xiong T, Peng Q, Du J, Sun W, Fan J, Peng X. Self-reporting photodynamic nanobody conjugate for precise and sustainable large-volume tumor treatment. Nat Commun 2024; 15:6935. [PMID: 39138197 PMCID: PMC11322375 DOI: 10.1038/s41467-024-51253-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 08/02/2024] [Indexed: 08/15/2024] Open
Abstract
Nanobodies (Nbs), the smallest antigen-binding fragments with high stability and affinity derived from the variable domain of naturally occurring heavy-chain-only antibodies in camelids, have been shown as an efficient way to improve the specificity to tumors for photodynamic therapy (PDT). Nonetheless, the rapid clearance of Nbs in vivo restricts the accumulation and retention of the photosensitizer at the tumor site causing insufficient therapeutic outcome, especially in large-volume tumors. Herein, we develop photodynamic conjugates, MNB-Pyra Nbs, through site-specific conjugation between 7D12 Nbs and type I photosensitizer MNB-Pyra (morpholine-modified nile blue structure connected to pyrazolinone) in a 1:2 ratio. The photosensitizers with long-term retention can be released at the tumor site by reactive oxygen species cleavage after illumination, accompanied with fluorescence recovery for self-reporting the occurrence of PDT. Ultimately, a single dose of MNB-Pyra Nbs demonstrate highly effective tumor suppression with high biosafety in the large-volume tumor models after three rounds of PDT. This nanobody conjugate provides a paradigm for the design of precise long-time retention photosensitizers and is expected to promote the development of PDT.
Collapse
Affiliation(s)
- Yingchao Chen
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
| | - Tao Xiong
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
| | - Qiang Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
| | - Jianjun Du
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, Dalian, 116024, China.
- Liaoning Binhai Laboratory, Dalian, 116023, China.
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, Dalian, 116024, China
- Liaoning Binhai Laboratory, Dalian, 116023, China
| |
Collapse
|
3
|
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
|
4
|
Gulyak EL, Alferova VA, Korshun VA, Sapozhnikova KA. Introduction of Carbonyl Groups into Antibodies. Molecules 2023; 28:7890. [PMID: 38067618 PMCID: PMC10707781 DOI: 10.3390/molecules28237890] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/26/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Antibodies and their derivatives (scFv, Fabs, etc.) represent a unique class of biomolecules that combine selectivity with the ability to target drug delivery. Currently, one of the most promising endeavors in this field is the development of molecular diagnostic tools and antibody-based therapeutic agents, including antibody-drug conjugates (ADCs). To meet this challenge, it is imperative to advance methods for modifying antibodies. A particularly promising strategy involves the introduction of carbonyl groups into the antibody that are amenable to further modification by biorthogonal reactions, namely aliphatic, aromatic, and α-oxo aldehydes, as well as aliphatic and aryl-alkyl ketones. In this review, we summarize the preparation methods and applications of site-specific antibody conjugates that are synthesized using this approach.
Collapse
Affiliation(s)
| | | | | | - Ksenia A. Sapozhnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (V.A.A.); (V.A.K.)
| |
Collapse
|
5
|
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
|
6
|
Peng Q, Zang B, Xiong T, Huang C, Xu T, Zhang C, Ren J, Ji F, Jia L. Single and dual functionalization of proteins using site-specific nucleophilic carbon ligations. Chem Commun (Camb) 2022; 58:6316-6319. [PMID: 35522979 DOI: 10.1039/d2cc01630c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We here found that while Meldrum's acid as the reactive warhead allows for the attachment of a single chemical modification on aldehyde-containing proteins, pyrazolone derivatives in combination with a phosphine nucleophile enable protein dual site-specific conjugation with the same or distinct moieties. These reactions are efficient and convergent under biocompatible conditions and allow access to protein bioconjugates with superior stability, homogeneity and flexibility. Our work expands the repertoire of bioconjugation chemistries and offers opportunities to construct bioconjugates with defined structure that have potential for medical and biomaterial applications.
Collapse
Affiliation(s)
- Qiang Peng
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, 116024, P. R. China.
| | - Berlin Zang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, 116024, P. R. China.
| | - Tao Xiong
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, P. R. China
| | - Chundong Huang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, 116024, P. R. China.
| | - Ting Xu
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, 116024, P. R. China.
| | - Chong Zhang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, 116024, P. R. China.
| | - Jun Ren
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, 116024, P. R. China.
| | - Fangling Ji
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, 116024, P. R. China.
| | - Lingyun Jia
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, 116024, P. R. China.
| |
Collapse
|
7
|
Angelastro A, Barkhanskiy A, Mattey AP, Pallister EG, Spiess R, Goundry W, Barran P, Flitsch SL. Galactose Oxidase Enables Modular Assembly of Conjugates from Native Antibodies with High Drug-to-Antibody Ratios. CHEMSUSCHEM 2022; 15:e202102592. [PMID: 34931761 PMCID: PMC9303943 DOI: 10.1002/cssc.202102592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/20/2021] [Indexed: 05/31/2023]
Abstract
The potential of antibody conjugates with high drug loading in anticancer therapy has recently been highlighted by the approval of Trastuzumab deruxtecan and Sacituzumab govitecan. These biopharmaceutical approaches have spurred interest in bioconjugation strategies with high and defined degrees of drug-to-antibody ratio (DAR), in particular on native antibodies. Here, a glycoengineering methodology was developed to generate antibody drug conjugates with DAR of up to eight, by combining highly selective enzymatic galactosylation and oxidation with biorthogonal tandem Knoevenagel-Michael addition chemistry. This four-step approach offers a selective route to conjugates from native antibodies with high drug loading, and thus illustrates how biocatalysis can be used for the generation of biopharmaceuticals using mild reaction conditions.
Collapse
Affiliation(s)
- Antonio Angelastro
- School of Chemistry and Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DN
| | - Alexey Barkhanskiy
- School of Chemistry and Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DN
| | - Ashley P. Mattey
- School of Chemistry and Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DN
| | - Edward G. Pallister
- School of Chemistry and Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DN
| | - Reynard Spiess
- School of Chemistry and Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DN
| | - William Goundry
- The Department of Pharmaceutical SciencesAstraZenecaSilk Road Business ParkMacclesfieldSK10 2NAUK
| | - Perdita Barran
- School of Chemistry and Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DN
| | - Sabine L. Flitsch
- School of Chemistry and Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DN
| |
Collapse
|
8
|
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
|
9
|
Seckler D, Dea CM, Rios EAM, de Godoi M, Rampon DDS, D’Oca MGM, D'Oca CDRM. Rice straw ash extract/glycerol: an efficient sustainable approach for Knoevenagel condensation. NEW J CHEM 2022. [DOI: 10.1039/d1nj05755c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
WERSA/glycerol is described as a powerful combination for the green approach of the Knoevenagel reaction, without any additional catalyst, base or promoter.
Collapse
Affiliation(s)
- Diego Seckler
- Laboratory of Polymers and Catalysis (LAPOCA), Department of Chemistry, Federal University of Paraná – UFPR, P. O. Box 19032, Curitiba, PR, 81531-990, Brazil
- Kolbe's Laboratory of Organic Synthesis, Department of Chemistry, Federal University of Paraná – UFPR, P. O. Box 19032, Curitiba, PR, 81531-990, Brazil
| | - Camila M. Dea
- Laboratory of Polymers and Catalysis (LAPOCA), Department of Chemistry, Federal University of Paraná – UFPR, P. O. Box 19032, Curitiba, PR, 81531-990, Brazil
| | - Elise Ane Maluf Rios
- Laboratory of Polymers and Catalysis (LAPOCA), Department of Chemistry, Federal University of Paraná – UFPR, P. O. Box 19032, Curitiba, PR, 81531-990, Brazil
| | - Marcelo de Godoi
- Food and Chemistry School, Federal University of Rio Grande – FURG, RS 95500-000, Brazil
| | - Daniel da Silveira Rampon
- Laboratory of Polymers and Catalysis (LAPOCA), Department of Chemistry, Federal University of Paraná – UFPR, P. O. Box 19032, Curitiba, PR, 81531-990, Brazil
| | - Marcelo Gonçalves Montes D’Oca
- Kolbe's Laboratory of Organic Synthesis, Department of Chemistry, Federal University of Paraná – UFPR, P. O. Box 19032, Curitiba, PR, 81531-990, Brazil
| | - Caroline Da Ros Montes D'Oca
- Laboratory of Polymers and Catalysis (LAPOCA), Department of Chemistry, Federal University of Paraná – UFPR, P. O. Box 19032, Curitiba, PR, 81531-990, Brazil
| |
Collapse
|
10
|
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
|
11
|
Khairy M, Mohamed M, Ismael M. Condensation of Active Methylene and Substituted Aldehydes over Mesoporous Nickel Oxides Nanostructures: A Combined Experimental and DFT Study. ChemistrySelect 2021. [DOI: 10.1002/slct.202100675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Mohamed Khairy
- Chemistry Department, Faculty of Science Sohag University Egypt 82524 Sohag
| | - Mounir Mohamed
- Chemistry Department, Faculty of Science Sohag University Egypt 82524 Sohag
| | - Mohamed Ismael
- Chemistry Department, Faculty of Science Sohag University Egypt 82524 Sohag
| |
Collapse
|
12
|
Suazo KF, Park KY, Distefano MD. A Not-So-Ancient Grease History: Click Chemistry and Protein Lipid Modifications. Chem Rev 2021; 121:7178-7248. [PMID: 33821625 PMCID: PMC8820976 DOI: 10.1021/acs.chemrev.0c01108] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Protein lipid modification involves the attachment of hydrophobic groups to proteins via ester, thioester, amide, or thioether linkages. In this review, the specific click chemical reactions that have been employed to study protein lipid modification and their use for specific labeling applications are first described. This is followed by an introduction to the different types of protein lipid modifications that occur in biology. Next, the roles of click chemistry in elucidating specific biological features including the identification of lipid-modified proteins, studies of their regulation, and their role in diseases are presented. A description of the use of protein-lipid modifying enzymes for specific labeling applications including protein immobilization, fluorescent labeling, nanostructure assembly, and the construction of protein-drug conjugates is presented next. Concluding remarks and future directions are presented in the final section.
Collapse
Affiliation(s)
- Kiall F. Suazo
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455 USA
| | - Keun-Young Park
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455 USA
| | - Mark D. Distefano
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455 USA
| |
Collapse
|
13
|
Gong K, Zhang D, Wang Y, Li C, Zhang H, Li H, Feng H. Biguanide-functionalized hierarchical porous covalent organic frameworks for efficient catalysis of condensation reactions. MOLECULAR CATALYSIS 2021. [DOI: 10.1016/j.mcat.2021.111663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
14
|
Toward Homogenous Antibody Drug Conjugates Using Enzyme-Based Conjugation Approaches. Pharmaceuticals (Basel) 2021; 14:ph14040343. [PMID: 33917962 PMCID: PMC8068374 DOI: 10.3390/ph14040343] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/02/2021] [Accepted: 04/04/2021] [Indexed: 01/17/2023] Open
Abstract
In the last few decades, antibody-based diagnostic and therapeutic applications have been well established in medicine and have revolutionized cancer managements by improving tumor detection and treatment. Antibodies are unique medical elements due to their powerful properties of being able to recognize specific antigens and their therapeutic mechanisms such as blocking specific pathways, antibody-dependent cellular cytotoxicity, and complement-dependent cytotoxicity. Furthermore, modification techniques have paved the way for improving antibody properties and to develop new classes of antibody-conjugate-based diagnostic and therapeutic agents. These techniques allow arming antibodies with various effector molecules. However, these techniques are utilizing the most frequently used amino acid residues for bioconjugation, such as cysteine and lysine. These bioconjugation approaches generate heterogeneous products with different functional and safety profiles. This is mainly due to the abundance of lysine and cysteine side chains. To overcome these limitations, different site-direct conjugation methods have been applied to arm the antibodies with therapeutic or diagnostics molecules to generate unified antibody conjugates with tailored properties. This review summarizes some of the enzyme-based site-specific conjugation approaches.
Collapse
|
15
|
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
|
16
|
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
|
17
|
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
|
18
|
Monehzadeh F, Rafiee Z. Application of GO/COF as a novel, efficient and recoverable catalyst in the Knoevenagel reaction. Appl Organomet Chem 2020. [DOI: 10.1002/aoc.5631] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Fatemeh Monehzadeh
- Department of ChemistryYasouj University Yasouj 75918‐74831 Islamic Republic of Iran
| | - Zahra Rafiee
- Department of ChemistryYasouj University Yasouj 75918‐74831 Islamic Republic of Iran
| |
Collapse
|
19
|
Zare E, Rafiee Z. Cellulose stabilized Fe
3
O
4
and carboxylate‐imidazole and Co‐based MOF growth as an exceptional catalyst for the Knoevenagel reaction. Appl Organomet Chem 2020. [DOI: 10.1002/aoc.5516] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Elham Zare
- Department of ChemistryYasouj University Yasouj 75918‐74831 Iran
| | - Zahra Rafiee
- Department of ChemistryYasouj University Yasouj 75918‐74831 Iran
| |
Collapse
|
20
|
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
|
21
|
Wagh YB, Padvi SA, Mahulikar PP, Dalal DS. CsF promoted rapid synthesis of spirooxindole‐pyran annulated heterocycles at room temperature in ethanol. J Heterocycl Chem 2019. [DOI: 10.1002/jhet.3846] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Yogesh B. Wagh
- School of Chemical SciencesKavayitri Bahinabai Chaudhari North Maharashtra University Jalgaon India
| | | | - Pramod P. Mahulikar
- School of Chemical SciencesKavayitri Bahinabai Chaudhari North Maharashtra University Jalgaon India
| | - Dipak S. Dalal
- School of Chemical SciencesKavayitri Bahinabai Chaudhari North Maharashtra University Jalgaon India
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
Chio TI, Gu H, Mukherjee K, Tumey LN, Bane SL. Site-Specific Bioconjugation and Multi-Bioorthogonal Labeling via Rapid Formation of a Boron-Nitrogen Heterocycle. Bioconjug Chem 2019; 30:1554-1564. [PMID: 31026151 PMCID: PMC6585445 DOI: 10.1021/acs.bioconjchem.9b00246] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Precise control of covalent bond formation in the presence of multiple functional groups is pertinent in the development of many next-generation bioconjugates and materials. Strategies derived from bioorthogonal chemistries are contributing greatly in that regard; however, the gain of chemoselectivity is often compromised by the slow rates of many of these existing chemistries. Recent work on a variation of the classical aldehyde/ketone condensation based on ortho-carbonylphenylboronic acids has uncovered markedly accelerated rates compared to those of the simple carbonyl counterparts. The products of these reactions are distinct, often in the form of boron-nitrogen heterocycles. In particular, we have shown that 2-formylphenylboronic acid (2fPBA), when coupled with an α-amino-hydrazide, produces a unique zwitterionic and stable 2,3,1-benzodiazaborine derivative. In this work, we apply this chemistry to generate chemically defined and functional bioconjugates, herein illustrated with immunoconjugates. We show that an antibody and a fluorophore (as payload) equipped with the relevant reactive handles undergo rapid conjugation at near-stoichiometric ratios, displaying a reaction half-life of only ∼5 min with 2 equiv of the linker payload. Importantly, the reaction can be extended to multicomponent labeling by partnering with the popular strain-promoted azide-alkyne cycloaddition and tetrazine- trans-cyclooctene (Tz-TCO) ligation. The mutual orthogonality to both of these chemistries allows simultaneous triple bioorthogonal conjugations, a rare feat thus far that will widen the scope of various multilabeling applications. Further collaboration with the Tz-TCO reaction enables rapid one-pot synthesis of a site-specific dual-payload antibody conjugate. Altogether, we envision that the 2fPBA-α-amino-hydrazide ligation will facilitate efficient assembly of diverse bioconjugates and materials, enabling access to more complex modalities via partnership with other orthogonal chemistries.
Collapse
Affiliation(s)
- Tak Ian Chio
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - Han Gu
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - Kamalika Mukherjee
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - L. Nathan Tumey
- Department of Pharmaceutical Sciences, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| | - Susan L. Bane
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902, United States
| |
Collapse
|
25
|
|
26
|
Renault K, Guillou C, Renard PY, Sabot C. Investigation of tetrazine reactivity towards C-nucleophiles: pyrazolone-based modification of biomolecules. Org Biomol Chem 2019; 17:388-396. [PMID: 30601507 DOI: 10.1039/c8ob02108b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Chemoselective, biocompatible ligation reactions are the key components for efficient and modular access to biomolecular scaffolds. Tetrazine ligation leads to the formation of a mixture of isomers, which makes reaction monitoring, purification and characterization of conjugates difficult. We report herein a modified tetrazine ligation strategy based on the use of a pyrazolone coupling partner, which provides a single molecule conjugate.
Collapse
Affiliation(s)
- Kévin Renault
- Normandie Univ, CNRS, UNIROUEN, INSA Rouen, COBRA (UMR 6014), 76000 Rouen, France.
| | | | | | | |
Collapse
|
27
|
Faridoon F, Shi W, Qin K, Tang Y, Li M, Guan D, Tian X, Jiang B, Dong J, Tang F, Huang W. New linker structures applied in glycosite-specific antibody drug conjugates. Org Chem Front 2019. [DOI: 10.1039/c9qo00646j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Two new linkers employed in glycosite-specific antibody–drug conjugates demonstrate efficient conjugation, enhanced stability, and fluorescence properties.
Collapse
|
28
|
Zhang Y, Park KY, Suazo KF, Distefano MD. Recent progress in enzymatic protein labelling techniques and their applications. Chem Soc Rev 2018; 47:9106-9136. [PMID: 30259933 PMCID: PMC6289631 DOI: 10.1039/c8cs00537k] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Protein-based conjugates are valuable constructs for a variety of applications. Conjugation of proteins to fluorophores is commonly used to study their cellular localization and the protein-protein interactions. Modification of therapeutic proteins with either polymers or cytotoxic moieties greatly enhances their pharmacokinetics or potency. To label a protein of interest, conventional direct chemical reaction with the side-chains of native amino acids often yields heterogeneously modified products. This renders their characterization complicated, requires difficult separation steps and may impact protein function. Although modification can also be achieved via the insertion of unnatural amino acids bearing bioorthogonal functional groups, these methods can have lower protein expression yields, limiting large scale production. As a site-specific modification method, enzymatic protein labelling is highly efficient and robust under mild reaction conditions. Significant progress has been made over the last five years in modifying proteins using enzymatic methods for numerous applications, including the creation of clinically relevant conjugates with polymers, cytotoxins or imaging agents, fluorescent or affinity probes to study complex protein interaction networks, and protein-linked materials for biosensing. This review summarizes developments in enzymatic protein labelling over the last five years for a panel of ten enzymes, including sortase A, subtiligase, microbial transglutaminase, farnesyltransferase, N-myristoyltransferase, phosphopantetheinyl transferases, tubulin tyrosin ligase, lipoic acid ligase, biotin ligase and formylglycine generating enzyme.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | | | | | |
Collapse
|
29
|
Krüger T, Dierks T, Sewald N. Formylglycine-generating enzymes for site-specific bioconjugation. Biol Chem 2018; 400:289-297. [DOI: 10.1515/hsz-2018-0358] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 09/28/2018] [Indexed: 01/01/2023]
Abstract
Abstract
Site-specific bioconjugation strategies offer many possibilities for directed protein modifications. Among the various enzyme-based conjugation protocols, formylglycine-generating enzymes allow to posttranslationally introduce the amino acid Cα-formylglycine (FGly) into recombinant proteins, starting from cysteine or serine residues within distinct consensus motifs. The aldehyde-bearing FGly-residue displays orthogonal reactivity to all other natural amino acids and can, therefore, be used for site-specific labeling reactions on protein scaffolds. In this review, the state of research on catalytic mechanisms and consensus motifs of different formylglycine-generating enzymes, as well as labeling strategies and applications of FGly-based bioconjugations are presented.
Collapse
Affiliation(s)
- Tobias Krüger
- Organic and Bioorganic Chemistry, Faculty of Chemistry , Bielefeld University , Universitätsstraße 25 , D-33615 Bielefeld , Germany
| | - Thomas Dierks
- Biochemistry I, Faculty of Chemistry , Bielefeld University , Universitätsstraße 25 , D-33615 Bielefeld , Germany
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Faculty of Chemistry , Bielefeld University , Universitätsstraße 25 , D-33615 Bielefeld , Germany
| |
Collapse
|
30
|
Huang BCB, Kim YC, Bañas S, Barfield RM, Drake PM, Rupniewski I, Haskins WE, Rabuka D. Antibody-drug conjugate library prepared by scanning insertion of the aldehyde tag into IgG1 constant regions. MAbs 2018; 10:1182-1189. [PMID: 30252630 PMCID: PMC6284588 DOI: 10.1080/19420862.2018.1512327] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The advantages of site-specific over stochastic bioconjugation technologies include homogeneity of product, minimal perturbation of protein structure/function, and – increasingly – the ability to perform structure activity relationship studies at the conjugate level. When selecting the optimal location for site-specific payload placement, many researchers turn to in silico modeling of protein structure to identify regions predicted to offer solvent-exposed conjugatable sites while conserving protein function. Here, using the aldehyde tag as our site-specific technology platform and human IgG1 antibody as our target protein, we demonstrate the power of taking an unbiased scanning approach instead. Scanning insertion of the human formylglycine generating enzyme (FGE) recognition sequence, LCTPSR, at each of the 436 positions in the light and heavy chain antibody constant regions followed by co-expression with FGE yielded a library of antibodies bearing an aldehyde functional group ready for conjugation. Each of the variants was expressed, purified, and conjugated to a cytotoxic payload using the Hydrazinyl Iso-Pictet-Spengler ligation to generate an antibody-drug conjugate (ADC), which was analyzed in terms of conjugatability (assessed by drug-to-antibody ratio, DAR) and percent aggregate. We searched for insertion sites that could generate manufacturable ADCs, defined as those variants yielding reasonable antibody titers, DARs of ≥ 1.3, and ≥ 95% monomeric species. Through this process, we discovered 58 tag insertion sites that met these metrics, including 14 sites in the light chain, a location that had proved refractory to the placement of manufacturable tag sites using in silico modeling/rational approaches.
Collapse
|
31
|
Krüger T, Weiland S, Falck G, Gerlach M, Boschanski M, Alam S, Müller KM, Dierks T, Sewald N. Zweifach-bioorthogonale Derivatisierung durch verschiedene Formylglycin-generierende Enzyme. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201803183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Tobias Krüger
- Organische und Bioorganische Chemie; Fakultät für Chemie; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Deutschland
| | - Stefanie Weiland
- Biochemie I; Fakultät für Chemie; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Deutschland
| | - Georg Falck
- Zelluläre und Molekulare Biotechnologie, Technische Fakultät; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Deutschland
| | - Marcus Gerlach
- Organische und Bioorganische Chemie; Fakultät für Chemie; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Deutschland
| | - Mareile Boschanski
- Biochemie I; Fakultät für Chemie; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Deutschland
| | - Sarfaraz Alam
- Biochemie I; Fakultät für Chemie; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Deutschland
| | - Kristian M. Müller
- Zelluläre und Molekulare Biotechnologie, Technische Fakultät; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Deutschland
| | - Thomas Dierks
- Biochemie I; Fakultät für Chemie; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Deutschland
| | - Norbert Sewald
- Organische und Bioorganische Chemie; Fakultät für Chemie; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Deutschland
| |
Collapse
|
32
|
Krüger T, Weiland S, Falck G, Gerlach M, Boschanski M, Alam S, Müller KM, Dierks T, Sewald N. Two-fold Bioorthogonal Derivatization by Different Formylglycine-Generating Enzymes. Angew Chem Int Ed Engl 2018; 57:7245-7249. [DOI: 10.1002/anie.201803183] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Tobias Krüger
- Organische und Bioorganische Chemie; Fakultät für Chemie; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Germany
| | - Stefanie Weiland
- Biochemie I; Fakultät für Chemie; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Germany
| | - Georg Falck
- Zelluläre und Molekulare Biotechnologie, Technische Fakultät; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Germany
| | - Marcus Gerlach
- Organische und Bioorganische Chemie; Fakultät für Chemie; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Germany
| | - Mareile Boschanski
- Biochemie I; Fakultät für Chemie; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Germany
| | - Sarfaraz Alam
- Biochemie I; Fakultät für Chemie; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Germany
| | - Kristian M. Müller
- Zelluläre und Molekulare Biotechnologie, Technische Fakultät; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Germany
| | - Thomas Dierks
- Biochemie I; Fakultät für Chemie; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Germany
| | - Norbert Sewald
- Organische und Bioorganische Chemie; Fakultät für Chemie; Universität Bielefeld; Universitätsstraße 25 33615 Bielefeld Germany
| |
Collapse
|
33
|
Nittoli T, Kelly MP, Delfino F, Rudge J, Kunz A, Markotan T, Spink J, Chen Z, Shan J, Navarro E, Tait M, Provoncha K, Giurleo J, Zhao F, Jiang X, Hylton D, Makonnen S, Hickey C, Kirshner JR, Thurston G, Papadopoulos N. Antibody drug conjugates of cleavable amino-alkyl and aryl maytansinoids. Bioorg Med Chem 2018; 26:2271-2279. [PMID: 29605304 DOI: 10.1016/j.bmc.2018.02.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 02/09/2018] [Accepted: 02/15/2018] [Indexed: 10/18/2022]
Abstract
Natural products have been used for many medicinal purposes for centuries. Antibody drug conjugates (ADCs) have utilized this rich source of small molecule therapeutics to produce several clinically useful treatments. ADCs based on the natural product maytansine have been successful clinically. The authors further the utility of the anti-cancer natural product maytansine by developing efficacious payloads and linker-payloads for conjugating to antibodies. The success of our approach was realized in the EGFRvIII targeting ADC EGFRvIII-16. The ADC was able to regress tumors in 2 tumor models (U251/EGFRvIII and MMT/EGFRvIII). When compared to a positive control ADC, the efficacy observed was similar or improved while the isotype control ADCs had no effect.
Collapse
Affiliation(s)
- Thomas Nittoli
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States.
| | - Marcus P Kelly
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Frank Delfino
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - John Rudge
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Arthur Kunz
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Thomas Markotan
- Abzena, 360 George Patterson Blvd, Bristol, PA 19007, United States
| | - Jan Spink
- Abzena, 360 George Patterson Blvd, Bristol, PA 19007, United States
| | - Zhaoyuan Chen
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Jing Shan
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Elizabeth Navarro
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Michele Tait
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Kathleen Provoncha
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Jason Giurleo
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Feng Zhao
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Xiaobo Jiang
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Donna Hylton
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Sosina Makonnen
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Carlos Hickey
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Jessica R Kirshner
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Gavin Thurston
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Nicholas Papadopoulos
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| |
Collapse
|
34
|
Yu J, Shen D, Zhang H, Yin Z. Rapid, Stoichiometric, Site-Specific Modification of Aldehyde-Containing Proteins Using a Tandem Knoevenagel-Intra Michael Addition Reaction. Bioconjug Chem 2018; 29:1016-1020. [DOI: 10.1021/acs.bioconjchem.8b00086] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Jian Yu
- Center of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100084, People’s Republic of China
| | - Da Shen
- Center of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100084, People’s Republic of China
| | - Hanjie Zhang
- Center of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100084, People’s Republic of China
| | - Zheng Yin
- Center of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100084, People’s Republic of China
| |
Collapse
|
35
|
Brabham RL, Spears RJ, Walton J, Tyagi S, Lemke EA, Fascione MA. Palladium-unleashed proteins: gentle aldehyde decaging for site-selective protein modification. Chem Commun (Camb) 2018; 54:1501-1504. [DOI: 10.1039/c7cc07740h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
A bioorthogonal decaging strategy has been developed to expose protein aldehydes using one equivalent of palladium, allowing site-selective protein labelling.
Collapse
Affiliation(s)
| | | | - Julia Walton
- Department of Chemistry
- University of York
- Heslington Road
- UK
| | - Swati Tyagi
- EMBL
- Meyerhofstrasse 1
- 69117 Heidelberg
- Germany
| | | | | |
Collapse
|
36
|
Yu J, Chen X, Jiang M, Wang A, Yang L, Pei X, Zhang P, Wu SG. Efficient promiscuous Knoevenagel condensation catalyzed by papain confined in Cu3(PO4)2 nanoflowers. RSC Adv 2018; 8:2357-2364. [PMID: 35541490 PMCID: PMC9077389 DOI: 10.1039/c7ra12940h] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 12/29/2017] [Indexed: 11/21/2022] Open
Abstract
To develop an efficient and green immobilized biocatalyst for promiscuous catalysis which has a broad scope of applications, hybrid nanoflower (hNF) confined papain as a biocatalyst has been proposed and characterized in this study. hNFs were firstly prepared through mixing CuSO4 aqueous solution with papain in phosphate saline (PBS) at room temperature. The resulting hNFs were characterized by SEM and verified through a hydrolysis reaction with N-benzoyl-dl-arginine amide as substrate. Under optimal conditions, this nano-biocatalyst demonstrated a 15-fold hydrolytic activity compared with papain of free form, along with better thermal stability. A series of reaction factors (reaction temperature, time, and solvent) have been investigated for Knoevenagel condensation reactions with hNFs as catalyst. At optimal conditions, product yield of the hNFs catalyzed reaction was 1.3 fold higher than that of the free enzyme with benzaldehyde and acetylacetone as substrates. A few aldehydes and methylene compounds have also been used to test the generality and scope of this new enzymatic promiscuity. To sum up, the obtained hNFs demonstrate better catalytic properties than free papain and the inorganic metal-salt crystal can function as both support and promotor in biocatalysis. Knoevenagel condensation was catalyzed and enhanced by Cu2+ and papain on hybrid nanoflowers (hNFs) in the promiscuous catalysis.![]()
Collapse
Affiliation(s)
- Jianyun Yu
- College of Materials, Chemistry and Chemical Engineering
- Hangzhou Normal University
- Hangzhou 310014
- P. R. China
| | - Xinxin Chen
- College of Materials, Chemistry and Chemical Engineering
- Hangzhou Normal University
- Hangzhou 310014
- P. R. China
| | - Min Jiang
- College of Materials, Chemistry and Chemical Engineering
- Hangzhou Normal University
- Hangzhou 310014
- P. R. China
| | - Anming Wang
- College of Materials, Chemistry and Chemical Engineering
- Hangzhou Normal University
- Hangzhou 310014
- P. R. China
| | - Linlin Yang
- College of Materials, Chemistry and Chemical Engineering
- Hangzhou Normal University
- Hangzhou 310014
- P. R. China
| | - Xiaolin Pei
- College of Materials, Chemistry and Chemical Engineering
- Hangzhou Normal University
- Hangzhou 310014
- P. R. China
| | - Pengfei Zhang
- College of Materials, Chemistry and Chemical Engineering
- Hangzhou Normal University
- Hangzhou 310014
- P. R. China
| | - Stephen Gang Wu
- Department of Energy, Environmental and Chemical Engineering
- Washington University
- St. Louis
- USA
| |
Collapse
|
37
|
Hamblett KJ. HER2-Targeted ADCs: At the Forefront of ADC Technology Development. CANCER DRUG DISCOVERY AND DEVELOPMENT 2018. [DOI: 10.1007/978-3-319-78154-9_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
38
|
Revisiting the Knoevenagel condensations: A universal and flexible bis-ammoniated fiber catalyst for the mild synthesis of α,β-unsaturated compounds. J IND ENG CHEM 2017. [DOI: 10.1016/j.jiec.2017.05.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
39
|
Botzanowski T, Erb S, Hernandez-Alba O, Ehkirch A, Colas O, Wagner-Rousset E, Rabuka D, Beck A, Drake PM, Cianférani S. Insights from native mass spectrometry approaches for top- and middle- level characterization of site-specific antibody-drug conjugates. MAbs 2017; 9:801-811. [PMID: 28406343 DOI: 10.1080/19420862.2017.1316914] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Antibody-drug conjugates (ADCs) have emerged as a family of compounds with promise as efficient immunotherapies. First-generation ADCs were generated mostly via reactions on either lysine side-chain amines or cysteine thiol groups after reduction of the interchain disulfide bonds, resulting in heterogeneous populations with a variable number of drug loads per antibody. To control the position and the number of drug loads, new conjugation strategies aiming at the generation of more homogeneous site-specific conjugates have been developed. We report here the first multi-level characterization of a site-specific ADC by state-of-the-art mass spectrometry (MS) methods, including native MS and its hyphenation to ion mobility (IM-MS). We demonstrate the versatility of native MS methodologies for site-specific ADC analysis, with the unique ability to provide several critical quality attributes within one single run, along with a direct snapshot of ADC homogeneity/heterogeneity without extensive data interpretation. The capabilities of native IM-MS to directly access site-specific ADC conformational information are also highlighted. Finally, the potential of these techniques for assessing an ADC's heterogeneity/homogeneity is illustrated by comparing the analytical characterization of a site-specific DAR4 ADC to that of first-generation ADCs. Altogether, our results highlight the compatibility, versatility, and benefits of native MS approaches for the analytical characterization of all types of ADCs, including site-specific conjugates. Thus, we envision integrating native MS and IM-MS approaches, even in their latest state-of-the-art forms, into workflows that benchmark bioconjugation strategies.
Collapse
Affiliation(s)
- Thomas Botzanowski
- a Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS , Strasbourg , France
| | - Stéphane Erb
- a Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS , Strasbourg , France
| | - Oscar Hernandez-Alba
- a Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS , Strasbourg , France
| | - Anthony Ehkirch
- a Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS , Strasbourg , France
| | - Olivier Colas
- b Centre d'Immunologie Pierre-Fabre (CIPF) , Saint-Julien-en-Genevois , France
| | - Elsa Wagner-Rousset
- b Centre d'Immunologie Pierre-Fabre (CIPF) , Saint-Julien-en-Genevois , France
| | - David Rabuka
- c Catalent Biologics West , Emeryville , CA , USA
| | - Alain Beck
- b Centre d'Immunologie Pierre-Fabre (CIPF) , Saint-Julien-en-Genevois , France
| | | | - Sarah Cianférani
- a Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS , Strasbourg , France
| |
Collapse
|