251
|
Guo Y, Yao L, Luo L, Wang HX, Yang Z, Wang Z, Ai SL, Zhang Y, Zou QC, Zhang HL. Alkylaminomaleimide fluorophores: synthesis via air oxidation and emission modulation by twisted intramolecular charge transfer. Org Chem Front 2021. [DOI: 10.1039/d0qo01285h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
A novel strategy to synthesize 3-alkylaminomaleimide fluorophores via air oxidation is developed, and the structural features for the designed TICT fluorophores with bright emission are established.
Collapse
|
252
|
Hill SA, Steinfort R, Hartmann L. Progress, challenges and future directions of heterocycles as building blocks in iterative methodologies towards sequence-defined oligomers and polymers. Polym Chem 2021. [DOI: 10.1039/d1py00425e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Heterocyclic building blocks for iterative methodologies leading to sequence-defined oligomers and polymers are reviewed. Solid- as well as solution-phase methods, challenges surrounding these systems and potential future directions are presented.
Collapse
Affiliation(s)
- Stephen A. Hill
- Institute of Organic and Macromolecular Chemistry
- Heinrich Heine University Düsseldorf
- 40225 Düsseldorf
- Germany
| | - Robert Steinfort
- Institute of Organic and Macromolecular Chemistry
- Heinrich Heine University Düsseldorf
- 40225 Düsseldorf
- Germany
| | - Laura Hartmann
- Institute of Organic and Macromolecular Chemistry
- Heinrich Heine University Düsseldorf
- 40225 Düsseldorf
- Germany
| |
Collapse
|
253
|
|
254
|
António JPM, Faustino H, Gois PMP. A 2-formylphenylboronic acid (2FPBA)-maleimide crosslinker: a versatile platform for Cys-peptide-hydrazine conjugation and interplay. Org Biomol Chem 2021; 19:6221-6226. [PMID: 34198316 DOI: 10.1039/d1ob00917f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In this work, we describe the preparation of a heterobifunctional 2-formylphenylboronic acid (2-FPBA)-maleimide crosslinker and explore its versatility in the preparation of various bioconjugates. We demonstrate the straightforward attachment of hydrazine payloads to cysteine residues in peptides, as well as the crosslinking of different thiol-bearing peptides or payloads with N-terminal cysteine peptides. Importantly, the dynamic nature of the 2-FPBA handle enables an interplay between the thiazolidine and diazaborine forms, which allows obtaining various products controlled by (and in some cases independent of) the order of addition of the components.
Collapse
Affiliation(s)
- João P M António
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal. and Chimie ParisTech, PSL University, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, F-75005 Paris, France
| | - Hélio Faustino
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal. and Association BLC3-Innovation and Technology Campus, Oliveira do Hospital, Portugal
| | - Pedro M P Gois
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
255
|
Park J, Lee S, Kim Y, Yoo TH. Methods to generate site-specific conjugates of antibody and protein. Bioorg Med Chem 2021; 30:115946. [DOI: 10.1016/j.bmc.2020.115946] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
|
256
|
Verdoliva V, Digilio G, Saviano M, De Luca S. Thio-conjugation of substituted benzofurazans to peptides: molecular sieves catalyze nucleophilic attack on unsaturated fused rings. Catal Sci Technol 2021. [DOI: 10.1039/d0cy02004d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
An efficient procedure to selectively introduce benzofurazan moieties into peptides was developed. It employs zeolite to catalyze the S-conjugation reaction.
Collapse
Affiliation(s)
- Valentina Verdoliva
- Institute of Biostructures and Bioimaging
- National Research Council
- 80134 Naples
- Italy
| | - Giuseppe Digilio
- Department of Science and Technologic Innovation
- Università del Piemonte Orientale “A. Avogadro”
- 15121 Alessandria
- Italy
| | - Michele Saviano
- Institute of Crystallography
- National Research Council
- 70126 Bari
- Italy
| | - Stefania De Luca
- Institute of Biostructures and Bioimaging
- National Research Council
- 80134 Naples
- Italy
| |
Collapse
|
257
|
Bartlett ME, Shuler SA, Rose DJ, Gilbert LM, Hegab RA, Lawton TJ, Messersmith RE. Paintable proteins: biofunctional coatings via covalent incorporation of proteins into a polymer network. NEW J CHEM 2021. [DOI: 10.1039/d1nj04687j] [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
Attaching proteins to surfaces while maintaining bioactivity is a promising avenue for developing new functional materials.
Collapse
Affiliation(s)
- Mairead E. Bartlett
- Research and Exploratory Development Department, The Johns Hopkins University Applied Physics Laboratory, 11100 Johns Hopkins Road, Laurel, Maryland 20723, USA
| | - Scott A. Shuler
- Research and Exploratory Development Department, The Johns Hopkins University Applied Physics Laboratory, 11100 Johns Hopkins Road, Laurel, Maryland 20723, USA
| | - Daniel J. Rose
- Research and Exploratory Development Department, The Johns Hopkins University Applied Physics Laboratory, 11100 Johns Hopkins Road, Laurel, Maryland 20723, USA
| | - Lindsey M. Gilbert
- Research and Exploratory Development Department, The Johns Hopkins University Applied Physics Laboratory, 11100 Johns Hopkins Road, Laurel, Maryland 20723, USA
| | - Rachel A. Hegab
- Research and Exploratory Development Department, The Johns Hopkins University Applied Physics Laboratory, 11100 Johns Hopkins Road, Laurel, Maryland 20723, USA
| | - Thomas J. Lawton
- Research and Exploratory Development Department, The Johns Hopkins University Applied Physics Laboratory, 11100 Johns Hopkins Road, Laurel, Maryland 20723, USA
| | - Reid E. Messersmith
- Research and Exploratory Development Department, The Johns Hopkins University Applied Physics Laboratory, 11100 Johns Hopkins Road, Laurel, Maryland 20723, USA
| |
Collapse
|
258
|
Thanzeel FY, Wolf C. Chemoselective bioconjugation based on modular click chemistry with 4-halocoumarins and aryl sulfonates. RSC Adv 2021; 11:18960-18965. [PMID: 35478620 PMCID: PMC9033492 DOI: 10.1039/d1ra03271b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 05/21/2021] [Indexed: 12/25/2022] Open
Abstract
We report chemoselective and modular peptide bioconjugation using stoichiometric amounts of 4-halocoumarin and arylsulfonate agents that undergo metal-free C(sp2)-heteroatom bond formation at micromolar concentrations. The underlying ipso-substitution click chemistry is irreversible and generates stable and inherently fluorescent bioconjugates, and the broad selection of coumarin tags offers high labeling flexibility and versatility. Different coumarins and arylsulfonates can be selectively attached to amino and thiol groups in the small peptides glutathione and ornipressin, and both free as well as latent thiols captured in disulfide bridges can be targeted if desired. The broad utility, ease of use, storage, and preparation of 4-halocoumarins and arylsulfonates are very attractive features that extend currently available dual bioconjugation capabilities. We report chemoselective and modular peptide bioconjugation using stoichiometric amounts of 4-halocoumarin and arylsulfonate agents that undergo metal-free C(sp2)-heteroatom bond formation at micromolar concentrations.![]()
Collapse
Affiliation(s)
| | - Christian Wolf
- Department of Chemistry
- Georgetown University
- Washington
- USA
| |
Collapse
|
259
|
|
260
|
Feng Y, Zhou Z, McDougald D, Meshaw RL, Vaidyanathan G, Zalutsky MR. Site-specific radioiodination of an anti-HER2 single domain antibody fragment with a residualizing prosthetic agent. Nucl Med Biol 2021; 92:171-183. [PMID: 32448731 PMCID: PMC7657985 DOI: 10.1016/j.nucmedbio.2020.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 11/28/2022]
Abstract
INTRODUCTION As a consequence of their small size, high stability and high affinity, single domain antibody fragments (sdAbs) are appealing targeting vectors for radiopharmaceutical development. With sdAbs binding to internalizing receptors like HER2, residualizing prosthetic agents can enhance tumor retention of radioiodine, which until now has been done with random labeling approaches. Herein we evaluate a site-specific strategy utilizing a radioiodinated, residualizing maleimido moiety and the anti-HER2 sdAb 5F7 bearing a GGC tail for conjugation. METHODS Maleimidoethyl 3-(guanidinomethyl)-5-iodobenzoate ([131I]MEGMB) and its N-succinimidyl ester analogue, iso-[125I]SGMIB, were labeled by halodestannylation and conjugated with 5F7GGC and 5F7, respectively. Radiochemical purity, immunoreactivity and binding affinity were determined. Paired-label experiments directly compared iso-[125I]SGMIB-5F7 and [131I]MEGMIB-5F7GGC with regard to internalization/residualization and affinity on HER2-expressing SKOV-3 ovarian carcinoma cells as well as biodistribution and metabolite distribution in athymic mice with subcutaneous SKOV-3 xenografts. RESULTS [131I]MEGMIB-5F7GGC had an immunoreactivity of 81.3% and Kd = 0.94 ± 0.27 nM. Internalization assays demonstrated high intracellular trapping for both conjugates, For example, at 1 h, intracellular retention was 50.30 ± 3.36% for [131I]MEGMIB-5F7GGC and 55.95 ± 3.27% for iso-[125I]SGMIB-5F7, while higher retention was seen for iso-[125I]SGMIB-5F7 at later time points. Peak tumor uptake was similar for both conjugates (8.35 ± 2.66%ID/g and 8.43 ± 2.84%ID/g for iso-[125I]SGMIB-5F7 and [131I]MEGMIB-5F7GGC at 1 h, respectively); however, more rapid normal tissue clearance was seen for [131I]MEGMIB-5F7GGC, with a 2-fold higher tumor-to-kidney ratio and a 3-fold higher tumor-to-liver ratio compared with co-injected iso-[125I]SGMIB-5F7. Consisted with this, generation of labeled catabolites in the kidneys was higher for [131I]MEGMIB-5F7GGC. CONCLUSION [131I]MEGMIB-5F7GGC offers similar tumor targeting as iso-[125I]SGMIB-5F7 but with generally lower normal tissue uptake. ADVANCES IN KNOWLEDGE AND IMPLICATION FOR PATIENT CARE The site specific nature of the [131I]MEGMIB reagent may facilitate clinical translation, particularly for sdAb with compromised affinity after random labeling.
Collapse
Affiliation(s)
- Yutian Feng
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Zhengyuan Zhou
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Darryl McDougald
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Rebecca L Meshaw
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | - Michael R Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
261
|
Verhelst SHL, Bonger KM, Willems LI. Bioorthogonal Reactions in Activity-Based Protein Profiling. Molecules 2020; 25:E5994. [PMID: 33352858 PMCID: PMC7765892 DOI: 10.3390/molecules25245994] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 12/27/2022] Open
Abstract
Activity-based protein profiling (ABPP) is a powerful technique to label and detect active enzyme species within cell lysates, cells, or whole animals. In the last two decades, a wide variety of applications and experimental read-out techniques have been pursued in order to increase our understanding of physiological and pathological processes, to identify novel drug targets, to evaluate selectivity of drugs, and to image probe targets in cells. Bioorthogonal chemistry has substantially contributed to the field of ABPP, as it allows the introduction of tags, which may be bulky or have unfavorable physicochemical properties, at a late stage in the experiment. In this review, we give an overview of the bioorthogonal reactions that have been implemented in ABPP, provide examples of applications of bioorthogonal chemistry in ABPP, and share some thoughts on future directions.
Collapse
Affiliation(s)
- Steven H. L. Verhelst
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven, Herestr. 49, Box 802, 3000 Leuven, Belgium
- AG Chemical Proteomics, Leibniz Institute for Analytical Sciences ISAS, e.V., Otto-Hahn-Str. 6b, 44227 Dortmund, Germany
| | - Kimberly M. Bonger
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Lianne I. Willems
- York Structural Biology Laboratory, Department of Chemistry, University of York, York YO10 5DD, UK
| |
Collapse
|
262
|
Dhanjee HH, Buslov I, Windsor IW, Raines RT, Pentelute BL, Buchwald SL. Palladium-Protein Oxidative Addition Complexes by Amine-Selective Acylation. J Am Chem Soc 2020; 142:21237-21242. [PMID: 33319995 DOI: 10.1021/jacs.0c09180] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Palladium oxidative addition complexes (OACs) are traditionally accessed by treating an aryl halide-containing substrate with a palladium(0) source. Here, a new strategy to selectively prepare stable OACs from amino groups on native proteins is presented. The approach relies on an amine-selective acylation reaction that occurs without modification of a preformed palladium(II)-aryl group. Once transferred onto a protein substrate, the palladium(II)-aryl group facilitates conjugation by undergoing reaction with a second, cysteine-containing protein. This operationally simple method is applicable to native, nonengineered enzymes as well as antibodies and is carried out in an aqueous setting and open to air. The resulting Pd-protein OACs are stable, storable reagents that retain biological activity and can be used to achieve protein-protein cross-coupling at nanomolar concentrations within hours.
Collapse
Affiliation(s)
- Heemal H Dhanjee
- Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Ivan Buslov
- Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Ian W Windsor
- Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Ronald T Raines
- Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Bradley L Pentelute
- Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Stephen L Buchwald
- Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
263
|
Ferreira VFC, Oliveira BL, D'Onofrio A, Farinha CM, Gano L, Paulo A, Bernardes GJL, Mendes F. In Vivo Pretargeting Based on Cysteine-Selective Antibody Modification with IEDDA Bioorthogonal Handles for Click Chemistry. Bioconjug Chem 2020; 32:121-132. [PMID: 33295756 DOI: 10.1021/acs.bioconjchem.0c00551] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pretargeted imaging has emerged as an effective multistep strategy aiming to improve imaging contrast and reduce patient radiation exposure through decoupling of the radioactivity from the targeting vector. The inverse electron-demand Diels-Alder (IEDDA) reaction between a trans-cyclooctene (TCO)-conjugated antibody and a labeled tetrazine holds great promise for pretargeted imaging applications due to its bioorthogonality, rapid kinetics under mild conditions, and formation of stable products. Herein, we describe the use of functionalized carbonylacrylic reagents for site-specific incorporation of TCO onto a human epidermal growth factor receptor 2 (HER2) antibody (THIOMAB) containing an engineered unpaired cysteine residue, generating homogeneous conjugates. Precise labeling of THIOMAB-TCO with a fluorescent or radiolabeled tetrazine revealed the potential of the TCO-functionalized antibody for imaging the HER2 after pretargeting in a cellular context in a HER2 positive breast cancer cell line. Control studies with MDA-MD-231 cells, which do not express HER2, further confirmed the target specificity of the modified antibody. THIOMAB-TCO was also evaluated in vivo after pretargeting and subsequent administration of an 111In-labeled tetrazine. Biodistribution studies in breast cancer tumor-bearing mice showed a significant activity accumulation on HER2+ tumors, which was 2.6-fold higher than in HER2- tumors. Additionally, biodistribution studies with THIOMAB without the TCO handle also resulted in a decreased uptake of 111In-DOTA-Tz on HER2+ tumors. Altogether, these results clearly indicate the occurrence of the click reaction at the tumor site, i.e., pretargeting of SK-BR-3 HER2-expressing cells with THIOMAB-TCO and reaction through the TCO moiety present in the antibody. The combined advantages of site-selectivity and stability of TCO tagged-antibodies could allow application of biorthogonal chemistry strategies for pretargeting imaging with minimal side-reactions and background.
Collapse
Affiliation(s)
- Vera F C Ferreira
- Center for Nuclear Sciences and Technologies (C2TN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - Bruno L Oliveira
- Instituto de Medicina Molecular João Lobo Antunes (iMM-JLA), Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Alice D'Onofrio
- Center for Nuclear Sciences and Technologies (C2TN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - Carlos M Farinha
- Biosystems and Integrative Sciences Institute (BioISI), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Lurdes Gano
- Center for Nuclear Sciences and Technologies (C2TN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal.,Departamento de Engenharia e Ciências Nucleares (DECN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - António Paulo
- Center for Nuclear Sciences and Technologies (C2TN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal.,Departamento de Engenharia e Ciências Nucleares (DECN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - Gonçalo J L Bernardes
- Instituto de Medicina Molecular João Lobo Antunes (iMM-JLA), Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal.,Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Filipa Mendes
- Center for Nuclear Sciences and Technologies (C2TN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal.,Departamento de Engenharia e Ciências Nucleares (DECN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| |
Collapse
|
264
|
Hübner R, Cheng X, Wängler B, Wängler C. Functional Hybrid Molecules for the Visualization of Cancer: PESIN-Homodimers Combined with Multimodal Molecular Imaging Probes for Positron Emission Tomography and Optical Imaging: Suited for Tracking of GRPR-Positive Malignant Tissue*. Chemistry 2020; 26:16349-16356. [PMID: 32618007 PMCID: PMC7756681 DOI: 10.1002/chem.202002386] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Indexed: 12/16/2022]
Abstract
We describe multimodal imaging probes for gastrin-releasing peptide receptor (GRPR)-specific targeting suited for positron emission tomography and optical imaging (PET/OI), consisting of PESIN (PEG3 -BBN7-14 ) dimers connected to multimodal imaging subunits. These multimodal agents comprise a fluorescent dye for OI and the chelator ((1,4,7-triazacyclononane-4,7-diyl)diacetic acid-1-glutaric acid) (NODA-GA) for PET radiometal isotope labelling. Special focus was put on the influence of the used dyes on the properties of the whole bioconjugates. For this, several compounds with different fluorescent dyes and non-dye carrying subunits were synthesized and investigated. As fluorescent dyes, dansyl, NBD, derivatives of fluorescein, coumarin and rhodamine as well as three pyrilium-based dyes were employed. Considerable influence of the charge of the colored unit on hydrophilicity as well as in vitro target receptor binding was observed and classified. High radiochemical yields and purities were found during radiolabeling of the multimodal imaging subunits as well as their GRPR-specific bioconjugates with 68 Ga. Examinations of the photophysical properties of both molecule species displayed no loss or alteration of fluorescence characteristics.
Collapse
Affiliation(s)
- Ralph Hübner
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear MedicineMedical Faculty Mannheim of Heidelberg UniversityTheodor-Kutzer-Ufer 1–368167MannheimGermany
| | - Xia Cheng
- Molecular Imaging and RadiochemistryDepartment of Clinical Radiology and Nuclear MedicineMedical Faculty Mannheim of Heidelberg UniversityTheodor-Kutzer-Ufer 1–368167MannheimGermany
| | - Björn Wängler
- Molecular Imaging and RadiochemistryDepartment of Clinical Radiology and Nuclear MedicineMedical Faculty Mannheim of Heidelberg UniversityTheodor-Kutzer-Ufer 1–368167MannheimGermany
| | - Carmen Wängler
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear MedicineMedical Faculty Mannheim of Heidelberg UniversityTheodor-Kutzer-Ufer 1–368167MannheimGermany
| |
Collapse
|
265
|
Matsuda Y, Mendelsohn BA. An overview of process development for antibody-drug conjugates produced by chemical conjugation technology. Expert Opin Biol Ther 2020; 21:963-975. [PMID: 33141625 DOI: 10.1080/14712598.2021.1846714] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: We discuss chemical conjugation strategies for antibody-drug conjugates (ADCs) from an industrial perspective and compare three promising chemical conjugation technologies to produce site-specific ADCs.Areas covered: Currently, nine ADCs are commercially approved and all are produced by chemical conjugation technology. However, seven of these ADCs contain a relatively broad drug distribution, potentially limiting their therapeutic indices. In 2019, the first site-specific ADC was launched on the market by Daiichi-Sankyo. This achievement, and an analysis of clinical trials over the last decade, indicates that current industrial interest in the ADC field is shifting toward site-specific conjugation technologies. From an industrial point of view, we aim to provide guidance regarding established conjugation methodologies that have already been applied to scale-up stages. With an emphasis on highly productive, scalable, and synthetic process robustness, conjugation methodologies for ADC production is discussed herein.Expert opinion: All three chemical conjugation technologies described in this review have various advantages and disadvantages, therefore drug developers can utilize these depending on their biological and/or protein targets. The future landscape of the ADC field is also discussed.
Collapse
Affiliation(s)
- Yutaka Matsuda
- Research Institute for Bioscience Products & Fine Chemicals, Ajinomoto Co Inc., 1-1 Suzuki-cho, Kawasaki-ku, Kawasaki 210-8681, Japan
| | - Brian A Mendelsohn
- Process Development & Tech Transfer, Ajinomoto Bio-Pharma Services, 11040 Roselle Street, San Diego, CA 92121, United States
| |
Collapse
|
266
|
Lossouarn A, Renard PY, Sabot C. Tailored Bioorthogonal and Bioconjugate Chemistry: A Source of Inspiration for Developing Kinetic Target-Guided Synthesis Strategies. Bioconjug Chem 2020; 32:63-72. [PMID: 33232599 DOI: 10.1021/acs.bioconjchem.0c00568] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Kinetic target-guided synthesis (KTGS) is a promising tool for the discovery of biologically active compounds. It relies on the identification of potent ligands that are covalently assembled by the biological targets themselves from a pool of reagents. Significant effort is devoted to developing new KTGS strategies; however, only a handful of biocompatible reactions are available, which may be insufficient to meet the specificities (stability, dynamics, active site topology, etc.) of a wide range of biological targets with therapeutic potential. This Topical Review proposes a retrospective analysis of existing KTGS ligation tools, in terms of their kinetics and analogy with other biocompatible reactions, and provides new clues to expand the KTGS toolkit. By way of examples, a nonexhaustive selection of such chemical ligation tools belonging to different classes of reactions as promising candidate reactions for KTGS are suggested.
Collapse
Affiliation(s)
- Alexis Lossouarn
- Normandie Université, Centre National de la Recherche Scientifique, UNIROUEN, INSA Rouen, COBRA, UMR 6014 & FR 3038, 76000, Rouen, France
| | - Pierre-Yves Renard
- Normandie Université, Centre National de la Recherche Scientifique, UNIROUEN, INSA Rouen, COBRA, UMR 6014 & FR 3038, 76000, Rouen, France
| | - Cyrille Sabot
- Normandie Université, Centre National de la Recherche Scientifique, UNIROUEN, INSA Rouen, COBRA, UMR 6014 & FR 3038, 76000, Rouen, France
| |
Collapse
|
267
|
Hu H, Steinmetz NF. Cisplatin Prodrug-Loaded Nanoparticles Based on Physalis Mottle Virus for Cancer Therapy. Mol Pharm 2020; 17:4629-4636. [PMID: 33186039 DOI: 10.1021/acs.molpharmaceut.0c00834] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Nanoparticle-based prodrugs offer an effective strategy to improve the safety and delivery of small-molecule therapeutics while reducing the risk of drug resistance. Here, we conjugated a maleimide-functionalized cisplatin prodrug containing Pt(IV) to the internal and/or external surface of virus-like particles (VLPs) derived from Physalis mottle virus (PhMV) to develop a pH-sensitive drug delivery system. The internally loaded and PEGylated VLPs (Pt-PhMVCy5.5-PEG) were taken up efficiently by cancer cells where they released platinum, presumably as a reduced, DNA-reactive Pt(II) complex, rapidly under acidic conditions in vitro (>80% in 30 h). The efficacy of the VLP-based drug delivery system was demonstrated against a panel of cancer cell lines, including cell lines resistant to platinum therapy. Furthermore, Pt-PhMVCy5.5-PEG successfully inhibited the growth of xenograft MDA-MB-231 breast tumors in vivo and significantly prolonged the survival of mice compared to free cisplatin and cisplatin-maleimide. Pt-PhMVCy5.5-PEG therefore appears promising as a prodrug to overcome the limitations of conventional platinum-based drugs for cancer therapy.
Collapse
Affiliation(s)
- He Hu
- Department of NanoEngineering, University of California-San Diego, La Jolla, California 92093, United States
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California-San Diego, La Jolla, California 92093, United States.,Department of Bioengineering, University of California-San Diego, La Jolla, California 92093, United States.,Department of Radiology, University of California-San Diego, La Jolla, California 92093, United States.,Moores Cancer Center, University of California-San Diego, La Jolla, California 92093, United States.,Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla, California 92093, United States
| |
Collapse
|
268
|
Chan HJ, Lin XH, Fan SY, Ru Hwu J, Tan KT. Rapid and Selective Labeling of Endogenous Transmembrane Proteins in Living Cells with a Difluorophenyl Ester Affinity-Based Probe. Chem Asian J 2020; 15:3416-3420. [PMID: 32931625 DOI: 10.1002/asia.202001049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Indexed: 12/28/2022]
Abstract
The long-term stability of affinity-based protein labeling probes is crucial to obtain reproducible protein labeling results. However, highly stable probes generally suffer from low protein labeling efficiency and pose significant challenges when labeling low abundance native proteins in living cells. In this paper, we report that protein labeling probes based on an ortho-difluorophenyl ester reactive module exhibit long-term stability in DMSO stock solution and aqueous buffer, yet they can undergo rapid and selective labeling of native proteins. This novel electrophile can be customized with a wide range of different protein ligands and is particularly well-suited for the labeling and imaging of transmembrane proteins. With this probe design, the identity and relative levels of basal and hypoxia-induced transmembrane carbonic anhydrases were revealed by live cell imaging and in-gel fluorescence analysis. We believe that the extension of this difluorophenyl ester reactive module would allow for the specific labeling of various endogenous membrane proteins, facilitating in-depth studies of their distribution and functions in biological processes.
Collapse
Affiliation(s)
- Hsin-Ju Chan
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan (Republic of China
| | - Xin-Hui Lin
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan (Republic of China
| | - Syuan-Yun Fan
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan (Republic of China
| | - Jih Ru Hwu
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan (Republic of China.,Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan (Republic of China
| | - Kui-Thong Tan
- Department of Chemistry, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan (Republic of China.,Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 101 Section 2, Kuang Fu Road, Hsinchu, 30013, Taiwan (Republic of China.,Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan (Republic of China
| |
Collapse
|
269
|
Vamisetti GB, Satish G, Sulkshane P, Mann G, Glickman MH, Brik A. On-Demand Detachment of Succinimides on Cysteine to Facilitate (Semi)Synthesis of Challenging Proteins. J Am Chem Soc 2020; 142:19558-19569. [PMID: 33136379 PMCID: PMC7705887 DOI: 10.1021/jacs.0c07663] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
![]()
The
maleimide group is a widely used reagent for bioconjugation
of peptides, proteins, and oligonucleotides employing Michael addition
and Diels–Alder cycloaddition reactions. However, the utility
of this functionality in chemical synthesis of peptides and proteins
remains unexplored. We report, for the first time that PdII complexes can mediate the efficient removal of various succinimide
derivatives in aqueous conditions. Succinimide removal by PdII was applied for the synthesis of two ubiquitin activity-based probes
(Ub-ABPs) employing solid phase chemical ligation (SPCL). SPCL was
achieved through a sequential three segment ligation on a polymer
support via a maleimide anchor. The obtained probes successfully formed
the expected covalent complexes with deubiquitinating enzymes (DUBs)
USP2 and USP7, highlighting the use of our new method for efficient
preparation of unique synthetic proteins. Importantly, we demonstrate
the advantages of our newly developed method for the protection and
deprotection of native cysteine with a succinimide group in a peptide
fragment derived from thioredoxin-1 (Trx-1) obtained via intein based
expression to enable ligation/desulfurization and subsequent disulfide
bond formation in a one-pot process.
Collapse
Affiliation(s)
- Ganga B Vamisetti
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Haifa 3200008, Israel
| | - Gandhesiri Satish
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Haifa 3200008, Israel
| | - Prasad Sulkshane
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200008, Israel
| | - Guy Mann
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Haifa 3200008, Israel
| | - Michael H Glickman
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200008, Israel
| | - Ashraf Brik
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Haifa 3200008, Israel
| |
Collapse
|
270
|
Bam R, Daryaei I, Abou-Elkacem L, Vilches-Moure JG, Meuillet EJ, Lutz A, Marinelli ER, Unger EC, Gambhir SS, Paulmurugan R. Toward the Clinical Development and Validation of a Thy1-Targeted Ultrasound Contrast Agent for the Early Detection of Pancreatic Ductal Adenocarcinoma. Invest Radiol 2020; 55:711-721. [PMID: 32569010 PMCID: PMC7541735 DOI: 10.1097/rli.0000000000000697] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Early detection of pancreatic ductal adenocarcinoma (PDAC) represents the most significant step toward the treatment of this aggressive lethal disease. Previously, we engineered a preclinical Thy1-targeted microbubble (MBThy1) contrast agent that specifically recognizes Thy1 antigen overexpressed in the vasculature of murine PDAC tissues by ultrasound (US) imaging. In this study, we adopted a single-chain variable fragment (scFv) site-specific bioconjugation approach to construct clinically translatable MBThy1-scFv and test for its efficacy in vivo in murine PDAC imaging, and functionally evaluated the binding specificity of scFv ligand to human Thy1 in patient PDAC tissues ex vivo. MATERIALS AND METHODS We recombinantly expressed the Thy1-scFv with a carboxy-terminus cysteine residue to facilitate its thioether conjugation to the PEGylated MBs presenting with maleimide functional groups. After the scFv-MB conjugations, we tested binding activity of the MBThy1-scFv to MS1 cells overexpressing human Thy1 (MS1Thy1) under liquid shear stress conditions in vitro using a flow chamber setup at 0.6 mL/min flow rate, corresponding to a wall shear stress rate of 100 seconds, similar to that in tumor capillaries. For in vivo Thy1 US molecular imaging, MBThy1-scFv was tested in the transgenic mouse model (C57BL/6J - Pdx1-Cre; KRas; Ink4a/Arf) of PDAC and in control mice (C57BL/6J) with L-arginine-induced pancreatitis or normal pancreas. To facilitate its clinical feasibility, we further produced Thy1-scFv without the bacterial fusion tags and confirmed its recognition of human Thy1 in cell lines by flow cytometry and in patient PDAC frozen tissue sections of different clinical grades by immunofluorescence staining. RESULTS Under shear stress flow conditions in vitro, MBThy1-scFv bound to MS1Thy1 cells at significantly higher numbers (3.0 ± 0.8 MB/cell; P < 0.01) compared with MBNontargeted (0.5 ± 0.5 MB/cell). In vivo, MBThy1-scFv (5.3 ± 1.9 arbitrary units [a.u.]) but not the MBNontargeted (1.2 ± 1.0 a.u.) produced high US molecular imaging signal (4.4-fold vs MBNontargeted; n = 8; P < 0.01) in the transgenic mice with spontaneous PDAC tumors (2-6 mm). Imaging signal from mice with L-arginine-induced pancreatitis (n = 8) or normal pancreas (n = 3) were not significantly different between the two MB constructs and were significantly lower than PDAC Thy1 molecular signal. Clinical-grade scFv conjugated to Alexa Fluor 647 dye recognized MS1Thy1 cells but not the parental wild-type cells as evaluated by flow cytometry. More importantly, scFv showed highly specific binding to VEGFR2-positive vasculature and fibroblast-like stromal components surrounding the ducts of human PDAC tissues as evaluated by confocal microscopy. CONCLUSIONS Our findings summarize the development and validation of a clinically relevant Thy1-targeted US contrast agent for the early detection of human PDAC by US molecular imaging.
Collapse
Affiliation(s)
- Rakesh Bam
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Palo Alto, CA
| | | | - Lotfi Abou-Elkacem
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Palo Alto, CA
| | | | | | - Amelie Lutz
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Palo Alto, CA
| | | | | | - Sanjiv S. Gambhir
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Palo Alto, CA
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Palo Alto, CA
| |
Collapse
|
271
|
David V, Moldoveanu SC, Galaon T. Derivatization procedures and their analytical performances for HPLC determination in bioanalysis. Biomed Chromatogr 2020; 35:e5008. [PMID: 33084080 DOI: 10.1002/bmc.5008] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023]
Abstract
Derivatization, or chemical structure modification, is often used in bioanalysis performed by liquid chromatography technique in order to enhance detectability or to improve the chromatographic performance for the target analytes. The derivatization process is discussed according to the analytical procedure used to achieve the reaction between the reagent and the target compounds (containing hydroxyl, thiol, amino, carbonyl and carboxyl as the main functional groups involved in derivatization). Important procedures for derivatization used in bioanalysis are in situ or based on extraction processes (liquid-liquid, solid-phase and related techniques) applied to the biomatrix. In the review, chiral, isotope-labeling, hydrophobicity-tailored and post-column derivatizations are also included, based on representative publications in the literature during the last two decades. Examples of derivatization reagents and brief reaction conditions are included, together with some bioanalytical applications and performances (chromatographic conditions, detection limit, stability and sample biomatrix).
Collapse
Affiliation(s)
- Victor David
- Faculty of Chemistry, Department of Analytical Chemistry, University of Bucharest, Bucharest, Romania
| | | | - Toma Galaon
- National Research and Development Institute for Industrial Ecology - ECOIND, Bucharest-6, Romania
| |
Collapse
|
272
|
Wall A, Wills AG, Forte N, Bahou C, Bonin L, Nicholls K, Ma MT, Chudasama V, Baker JR. One-pot thiol-amine bioconjugation to maleimides: simultaneous stabilisation and dual functionalisation. Chem Sci 2020; 11:11455-11460. [PMID: 34094388 PMCID: PMC8162801 DOI: 10.1039/d0sc05128d] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 09/23/2020] [Indexed: 11/21/2022] Open
Abstract
Maleimide chemistry is widely used in the site-selective modification of proteins. However, hydrolysis of the resultant thiosuccinimides is required to provide robust stability to the bioconjugates. Herein, we present an alternative approach that affords simultaneous stabilisation and dual functionalisation in a one pot fashion. By consecutive conjugation of a thiol and an amine to dibromomaleimides, we show that aminothiomaleimides can be generated extremely efficiently. Furthermore, the amine serves to deactivate the electrophilicity of the maleimide, precluding further reactivity and hence generating stable conjugates. We have applied this conjugation strategy to peptides and proteins to generate stabilised trifunctional conjugates. We propose that this stabilisation-dual modification strategy could have widespread use in the generation of diverse conjugates.
Collapse
Affiliation(s)
- Archie Wall
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Alfie G Wills
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Nafsika Forte
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Calise Bahou
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Lisa Bonin
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | | | - Michelle T Ma
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London SE1 7EH UK
| | - Vijay Chudasama
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa Lisbon Portugal
| | - James R Baker
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| |
Collapse
|
273
|
Recent advances of thiol-selective bioconjugation reactions. Curr Opin Chem Biol 2020; 58:28-36. [DOI: 10.1016/j.cbpa.2020.04.017] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 01/09/2023]
|
274
|
Miller LM, Herman R, Gyulev I, Krauss TF, Thomas GH, Duhme-Klair AK. Synthesis and biochemical evaluation of cephalosporin analogues equipped with chemical tethers. RSC Adv 2020; 10:36485-36494. [PMID: 35517937 PMCID: PMC9056950 DOI: 10.1039/d0ra04893c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/25/2020] [Indexed: 02/04/2023] Open
Abstract
Molecular probes typically require structural modifications to allow for the immobilisation or bioconjugation with a desired substrate but the effects of these changes are often not evaluated. Here, we set out to determine the effects of attaching functional handles to a first-generation cephalosporin. A series of cephalexin derivatives was prepared, equipped with chemical tethers suitable for the site-selective conjugation of antibiotics to functionalised surfaces. The tethers were positioned remotely from the β-lactam ring to ensure minimal effect to the antibiotic's pharmacophore. Herein, the activity of the modified antibiotics was evaluated for binding to the therapeutic target, the penicillin binding proteins, and shown to maintain binding interactions. In addition, the deactivation of the modified drugs by four β-lactamases (TEM-1, CTX-M-15, AmpC, NDM-1) was investigated and the effect of the tethers on the catalytic efficiencies determined. CTX-M-15 was found to favour hydrolysis of the parent antibiotic without a tether, whereas AmpC and NDM-1 were found to favour the modified analogues. Furthermore, the antimicrobial activity of the derivatives was evaluated to investigate the effect of the structural modifications on the antimicrobial activity of the parent drug, cephalexin. Tethered β-lactam antibiotics provide insights into designing chemical tools to target specific β-lactamases.![]()
Collapse
Affiliation(s)
- Lisa M Miller
- Department of Chemistry, University of York Heslington York YO10 5DD UK
| | - Reyme Herman
- Department of Biology, University of York Heslington York YO10 5DD UK
| | - Ivan Gyulev
- Department of Biology, University of York Heslington York YO10 5DD UK
| | - Thomas F Krauss
- Department of Physics, University of York Heslington York YO10 5DD UK
| | - Gavin H Thomas
- Department of Biology, University of York Heslington York YO10 5DD UK
| | | |
Collapse
|
275
|
Mann FA, Herrmann N, Opazo F, Kruss S. Quantum Defects as a Toolbox for the Covalent Functionalization of Carbon Nanotubes with Peptides and Proteins. Angew Chem Int Ed Engl 2020; 59:17732-17738. [PMID: 32511874 PMCID: PMC7540668 DOI: 10.1002/anie.202003825] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/24/2020] [Indexed: 12/16/2022]
Abstract
Single-walled carbon nanotubes (SWCNTs) are a 1D nanomaterial that shows fluorescence in the near-infrared (NIR, >800 nm). In the past, covalent chemistry was less explored to functionalize SWCNTs as it impairs NIR emission. However, certain sp3 defects (quantum defects) in the carbon lattice have emerged that preserve NIR fluorescence and even introduce a new, red-shifted emission peak. Here, we report on quantum defects, introduced using light-driven diazonium chemistry, that serve as anchor points for peptides and proteins. We show that maleimide anchors allow conjugation of cysteine-containing proteins such as a GFP-binding nanobody. In addition, an Fmoc-protected phenylalanine defect serves as a starting point for conjugation of visible fluorophores to create multicolor SWCNTs and in situ peptide synthesis directly on the nanotube. Therefore, these quantum defects are a versatile platform to tailor both the nanotube's photophysical properties as well as their surface chemistry.
Collapse
Affiliation(s)
- Florian A. Mann
- Institute of Physical ChemistryGeorg-August UniversitätTammannstraße 637077GöttingenGermany
| | - Niklas Herrmann
- Institute of Physical ChemistryGeorg-August UniversitätTammannstraße 637077GöttingenGermany
| | - Felipe Opazo
- Center for Biostructural Imaging of NeurodegenerationVon-Siebold-Straße 3a37075GöttingenGermany
| | - Sebastian Kruss
- Institute of Physical ChemistryGeorg-August UniversitätTammannstraße 637077GöttingenGermany
| |
Collapse
|
276
|
Tran TT, Rabah J, Ha-Thi MH, Allard E, Nizinski S, Burdzinski G, Aloïse S, Fensterbank H, Baczko K, Nasrallah H, Vallée A, Clavier G, Miomandre F, Pino T, Méallet-Renault R. Photoinduced Electron Transfer and Energy Transfer Processes in a Flexible BODIPY-C 60 Dyad. J Phys Chem B 2020; 124:9396-9410. [PMID: 32897728 DOI: 10.1021/acs.jpcb.0c05187] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A new donor-acceptor dyad composed of a BODIPY (4,4'-difluoro-4-bora-3a,4a-diaza-s-indacene) donor and a fullerene C60 acceptor has been synthesized and characterized. This derivative has been prepared using a clickable fullerene building block that bears an alkyne moiety and a maleimide unit. The post-functionalization of the maleimide group by a BODIPY thiol leads to a BODIPY-C60 dyad, leaving the alkyne moiety for further functional arrangement. On the basis of the combination of semi-empirical and density functional theory (DFT) calculations, spectroelectrochemical experiments, and steady-state and time-resolved spectroscopies, the photophysical properties of this new BODIPY-C60 dyad were thoroughly studied. By using semi-empirical calculations, the equilibrium of three conformations of the BODIPY-C60 dyad has been deduced, and their molecular orbital structures have been analyzed using DFT calculations. Two short fluorescence lifetimes were attributed to two extended conformers displaying variable donor-acceptor distances (17.5 and 20.0 Å). Additionally, the driving force for photoinduced electron transfer from the singlet excited state of BODIPY to the C60 moiety was calculated using redox potentials determined with electrochemical studies. Spectroelectrochemical measurements were also carried out to investigate the absorption profiles of radicals in the BODIPY-C60 dyad in order to assign the transient species in pump-probe experiments. Under selective photoexcitation of the BODIPY moiety, occurrences of both energy and electron transfers were demonstrated for the dyad by femtosecond and nanosecond transient absorption spectroscopies. Photoinduced electron transfer occurs in the folded conformer, while energy transfer is observed in extended conformers.
Collapse
Affiliation(s)
- Thu-Trang Tran
- Université Paris-Saclay, CNRS, Institut des Sciences Moléculaires d'Orsay, 91405 Orsay, France.,Faculty of Physics and Technology, Thai Nguyen University of Science, Thai Nguyen 24000, Vietnam
| | - Jad Rabah
- Université Paris-Saclay, UVSQ, CNRS, Institut Lavoisier de Versailles, 78000 Versailles, France
| | - Minh-Huong Ha-Thi
- Université Paris-Saclay, CNRS, Institut des Sciences Moléculaires d'Orsay, 91405 Orsay, France
| | - Emmanuel Allard
- Université Paris-Saclay, UVSQ, CNRS, Institut Lavoisier de Versailles, 78000 Versailles, France
| | - Stanislaw Nizinski
- Adam Mickiewicz University in Poznan, Fac Phys, Quantum Elect Lab, PL-61614 Poznan, Poland
| | - Gotard Burdzinski
- Adam Mickiewicz University in Poznan, Fac Phys, Quantum Elect Lab, PL-61614 Poznan, Poland
| | - Stéphane Aloïse
- Laboratoire de Spectrochimie Infrarouge et Raman, UMR-CNRS 8516, Université de Lille, F-59000 Lille, France
| | - Hélène Fensterbank
- Université Paris-Saclay, UVSQ, CNRS, Institut Lavoisier de Versailles, 78000 Versailles, France
| | - Krystyna Baczko
- Université Paris-Saclay, UVSQ, CNRS, Institut Lavoisier de Versailles, 78000 Versailles, France
| | - Houssein Nasrallah
- Université Paris-Saclay, UVSQ, CNRS, Institut Lavoisier de Versailles, 78000 Versailles, France
| | - Anne Vallée
- Université Paris-Saclay, UVSQ, CNRS, Institut Lavoisier de Versailles, 78000 Versailles, France
| | - Gilles Clavier
- PPSM, UMR-CNRS 8531, ENS Paris Saclay, 61 Avenue du Président Wilson, 94235 Cachan, France
| | - Fabien Miomandre
- PPSM, UMR-CNRS 8531, ENS Paris Saclay, 61 Avenue du Président Wilson, 94235 Cachan, France
| | - Thomas Pino
- Université Paris-Saclay, CNRS, Institut des Sciences Moléculaires d'Orsay, 91405 Orsay, France
| | - Rachel Méallet-Renault
- Université Paris-Saclay, CNRS, Institut des Sciences Moléculaires d'Orsay, 91405 Orsay, France
| |
Collapse
|
277
|
Tamshen K, Wang Y, Jamieson SM, Perry JK, Maynard HD. Genetic Code Expansion Enables Site-Specific PEGylation of a Human Growth Hormone Receptor Antagonist through Click Chemistry. Bioconjug Chem 2020; 31:2179-2190. [PMID: 32786367 PMCID: PMC8291075 DOI: 10.1021/acs.bioconjchem.0c00365] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Regulation of human growth hormone (GH) signaling has important applications in the remediation of several diseases including acromegaly and cancer. Growth hormone receptor (GHR) antagonists currently provide the most effective means for suppression of GH signaling. However, these small 22 kDa recombinantly engineered GH analogues exhibit short plasma circulation times. To improve clinical viability, between four and six molecules of 5 kDa poly(ethylene glycol) (PEG) are nonspecifically conjugated to the nine amines of the GHR antagonist designated as B2036 in the FDA-approved therapeutic pegvisomant. PEGylation increases the molecular weight of B2036 and considerably extends its circulation time, but also dramatically reduces its bioactivity, contributing to high dosing requirements and increased cost. As an alternative to nonspecific PEGylation, we report the use of genetic code expansion technology to site-specifically incorporate the unnatural amino acid propargyl tyrosine (pglY) into B2036 with the goal of producing site-specific protein-polymer conjugates. Substitution of tyrosine 35 with pglY yielded a B2036 variant containing an alkyne functional group without compromising bioactivity, as verified by a cellular assay. Subsequent conjugation of 5, 10, and 20 kDa azide-containing PEGs via the copper-catalyzed click reaction yielded high purity, site-specific conjugates with >89% conjugation efficiencies. Site-specific attachment of PEG to B2036 is associated with substantially improved in vitro bioactivity values compared to pegvisomant, with an inverse relationship between polymer size and activity observed. Notably, the B2036-20 kDa PEG conjugate has a molecular weight comparable to pegvisomant, while exhibiting a 12.5 fold improvement in half-maximal inhibitory concentration in GHR-expressing Ba/F3 cells (103.3 nM vs 1289 nM). We expect that this straightforward route to achieve site-specific GHR antagonists will be useful for GH signal regulation.
Collapse
Affiliation(s)
- Kyle Tamshen
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095-1569, United States
| | - Yue Wang
- Liggins Institute, University of Auckland, Auckland 1203, New Zealand
| | - Stephen M.F. Jamieson
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland 1023, New Zealand
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Jo K. Perry
- Liggins Institute, University of Auckland, Auckland 1203, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland 1023, New Zealand
| | - Heather D. Maynard
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095-1569, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095-1569, United States
- Department of Bioengineering, University of California, Los Angeles, California 90095-1569, United States
| |
Collapse
|
278
|
Lidický O, Klener P, Machová D, Vočková P, Pokorná E, Helman K, Mavis C, Janoušková O, Etrych T. Overcoming resistance to rituximab in relapsed non-Hodgkin lymphomas by antibody-polymer drug conjugates actively targeted by anti-CD38 daratumumab. J Control Release 2020; 328:160-170. [PMID: 32860930 DOI: 10.1016/j.jconrel.2020.08.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/11/2020] [Accepted: 08/21/2020] [Indexed: 11/30/2022]
Abstract
B-cell non-Hodgkin lymphomas (B-NHL) represent the most common type of hematologic malignancies in the Western hemisphere. The therapy of all B-NHL is based on the combination of different genotoxic cytostatics and anti-CD20 monoclonal antibody (mAb) rituximab. Unfortunately, many patients relapse after the mentioned front-line treatment approaches. The therapy of patients with relapsed/refractory (R/R) B-NHL represents an unmet medical need. We designed, developed and tested novel actively targeted hybrid mAb-polymer-drug conjugate (APDC) containing anti-CD20, anti-CD38 or anti-CD19 mAbs. Biocompatible copolymers based on N-(2-hydroxypropyl)methacrylamide (HPMA) with cytostatic agent doxorubicin attached via stimuli-sensitive hydrazone bond were employed for the mAb grafting. Anti-lymphoma efficacy of the APDC nanotherapeutics was evaluated in vivo on a panel of three patient-derived lymphoma xenografts derived from two patients with R/R B-NHL and one patient with so far untreated B-NHL. In both PDX models derived from patients with R/R B-NHL, the targeting with anti-CD38 antibody daratumumab demonstrated highly improved anti-lymphoma efficacy compared to the targeting with anti-CD20 rituximab, two experimental anti-CD19 antibodies and non-targeted controls. The results represent a proof-of-concept of a new algorithm of personalized anti-tumor therapy based on highly innovative APDC biomaterials.
Collapse
Affiliation(s)
- Ondřej Lidický
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Pavel Klener
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic; Institute of Pathological Physiology, First Faculty of Medicine, Charles University, U Nemocnice 5, 128 53 Prague 2, Czech Republic; First Department of Internal Medicine- Hematology, University General Hospital and First Faculty of Medicine, Charles University, U Nemocnice 2, 128 08 Prague 2, Czech Republic
| | - Daniela Machová
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Petra Vočková
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, U Nemocnice 5, 128 53 Prague 2, Czech Republic; First Department of Internal Medicine- Hematology, University General Hospital and First Faculty of Medicine, Charles University, U Nemocnice 2, 128 08 Prague 2, Czech Republic
| | - Eva Pokorná
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, U Nemocnice 5, 128 53 Prague 2, Czech Republic
| | - Karel Helman
- Faculty of Informatics and Statistics, University of Economics, Prague, Czech Republic
| | - Cory Mavis
- Department of Medicine Lymphoma/Myeloma, Roswell Park, Comprehensive Cancer Center, Elm & Carlton Sts, Buffalo, NY 14263, United States of America
| | - Olga Janoušková
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic.
| |
Collapse
|
279
|
Tobiesen HN, Leth LA, Iversen MV, Næsborg L, Bertelsen S, Jørgensen KA. Stereoselective Oxidative Bioconjugation of Amino Acids and Oligopeptides to Aldehydes. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202008513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Henriette N. Tobiesen
- Department of Chemistry Aarhus University 8000 Aarhus C Denmark
- Research Chemistry, Global Research Technologies Novo Nordisk A/S 2760 Maaloev Denmark
| | - Lars A. Leth
- Department of Chemistry Aarhus University 8000 Aarhus C Denmark
| | - Marc V. Iversen
- Department of Chemistry Aarhus University 8000 Aarhus C Denmark
| | - Line Næsborg
- Department of Chemistry Aarhus University 8000 Aarhus C Denmark
| | - Søren Bertelsen
- Research Chemistry, Global Research Technologies Novo Nordisk A/S 2760 Maaloev Denmark
| | | |
Collapse
|
280
|
Tobiesen HN, Leth LA, Iversen MV, Næsborg L, Bertelsen S, Jørgensen KA. Stereoselective Oxidative Bioconjugation of Amino Acids and Oligopeptides to Aldehydes. Angew Chem Int Ed Engl 2020; 59:18490-18494. [DOI: 10.1002/anie.202008513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Indexed: 01/09/2023]
Affiliation(s)
- Henriette N. Tobiesen
- Department of Chemistry Aarhus University 8000 Aarhus C Denmark
- Research Chemistry, Global Research Technologies Novo Nordisk A/S 2760 Maaloev Denmark
| | - Lars A. Leth
- Department of Chemistry Aarhus University 8000 Aarhus C Denmark
| | - Marc V. Iversen
- Department of Chemistry Aarhus University 8000 Aarhus C Denmark
| | - Line Næsborg
- Department of Chemistry Aarhus University 8000 Aarhus C Denmark
| | - Søren Bertelsen
- Research Chemistry, Global Research Technologies Novo Nordisk A/S 2760 Maaloev Denmark
| | | |
Collapse
|
281
|
Izraylit V, Hommes-Schattmann PJ, Neffe AT, Gould OE, Lendlein A. Polyester urethane functionalizable through maleimide side-chains and cross-linkable by polylactide stereocomplexes. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2020.109916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
282
|
Mann FA, Herrmann N, Opazo F, Kruss S. Quantendefekte als Werkzeugkasten für die kovalente Funktionalisierung von Kohlenstoffnanoröhren mit Peptiden und Proteinen. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202003825] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Florian A. Mann
- Institut für Physikalische Chemie Georg-August Universität Tammannstraße 6 37077 Göttingen Deutschland
| | - Niklas Herrmann
- Institut für Physikalische Chemie Georg-August Universität Tammannstraße 6 37077 Göttingen Deutschland
| | - Felipe Opazo
- Center for Biostructural Imaging of Neurodegeneration Von-Siebold-Straße 3a 37075 Göttingen Deutschland
| | - Sebastian Kruss
- Institut für Physikalische Chemie Georg-August Universität Tammannstraße 6 37077 Göttingen Deutschland
| |
Collapse
|
283
|
Lynch DM, Scanlan EM. Thiyl Radicals: Versatile Reactive Intermediates for Cyclization of Unsaturated Substrates. Molecules 2020; 25:E3094. [PMID: 32646036 PMCID: PMC7412111 DOI: 10.3390/molecules25133094] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 01/11/2023] Open
Abstract
Sulfur centered radicals are widely employed in chemical synthesis, in particular for alkene and alkyne hydrothiolation towards thioether bioconjugates. The steadfast radical chain process that enables efficient hydrothiolation has been explored in the context of cascade reactions to furnish complex molecular architectures. The use of thiyl radicals offers a much cheaper and less toxic alternative to the archetypal organotin-based radical methods. This review outlines the development of thiyl radicals as reactive intermediates for initiating carbocyclization cascades. Key developments in cascade cyclization methodology are presented and applications for natural product synthesis are discussed. The review provides a chronological account of the field, beginning in the early seventies up to very recent examples; a span of almost 50 years.
Collapse
Affiliation(s)
| | - Eoin M. Scanlan
- School of Chemistry and Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, The University of Dublin, Dublin 2, Ireland;
| |
Collapse
|
284
|
Piestansky J, Barath P, Majerova P, Galba J, Mikus P, Kovacech B, Kovac A. A simple and rapid LC-MS/MS and CE-MS/MS analytical strategy for the determination of therapeutic peptides in modern immunotherapeutics and biopharmaceutics. J Pharm Biomed Anal 2020; 189:113449. [PMID: 32622303 DOI: 10.1016/j.jpba.2020.113449] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023]
Abstract
Modern therapy of metabolic, neurodegenerative, inflammation, or cancer diseases is recently based on an immunotherapeutic approach. The peptide conjugates represent innovative and effective therapeutics that are better tolerated and are much more specific than small molecule-based medicines. The nature and manufacturing process of these therapeutics make their analysis very challenging. Here, two robust analytical methods based on an on-line combination of ultra-high-performance liquid chromatography with tandem mass spectrometry (UHPLC-MS/MS) and capillary electrophoresis with tandem mass spectrometry (CE-MS/MS) were developed for fast determination of immunogenic synthetic peptide (peptide sequence CADNLHKVVGQST) in a conjugate with bovine serum albumin (BSA) as a carrier protein and is a peptide, conjugate formulated with a vaccine adjuvant - Alhydrogel® 2 %. An effective non-enzymatic release step of the peptide from the final peptide conjugate based on acid hydrolysis with the use of 2% formic acid was successfully tested and implemented. The proposed methods were validated according to the ICH guideline and parameters such as linearity, precision, and accuracy, the limit of detection (LOD) or limit of quantification (LOQ) were assessed. Calibration curves were linear within the range of 1-30 μg.mL-1 and the correlation coefficients were higher than 0.99. The intraday and interday precisions were 3.2-8.1 % (UHPLC-MS/MS), 1.6-9.3 % (CE-MS/MS) and 3.6-10.3 % (UHPLC-MS/MS), 4.1-10.2 % (CE-MS/MS), respectively. The recovery ranged in the interval of 98.4-107.4 % for UHPLC-MS/MS method and 100.3-103.2 % for CE-MS/MS method. The presented approaches represent an effective tool for simple, rapid and robust quantification of immunogens in modern immunotherapeutics and other biopharmaceuticals with appropriate peptide sequences.
Collapse
Affiliation(s)
- Juraj Piestansky
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovak Republic; Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, SK-832 32 Bratislava, Slovak Republic.
| | - Peter Barath
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, SK-845 38 Bratislava, Slovak Republic.
| | - Petra Majerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, SK-845 10 Bratislava, Slovak Republic.
| | - Jaroslav Galba
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovak Republic.
| | - Peter Mikus
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, SK-832 32 Bratislava, Slovak Republic; Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, SK-832 32 Bratislava, Slovak Republic.
| | - Branislav Kovacech
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, SK-845 10 Bratislava, Slovak Republic.
| | - Andrej Kovac
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, SK-845 10 Bratislava, Slovak Republic.
| |
Collapse
|
285
|
Schaffer A, Kränzlein M, Rieger B. Synthesis and Application of Functional Group-Bearing Pyridyl-Based Initiators in Rare Earth Metal-Mediated Group Transfer Polymerization. Macromolecules 2020. [DOI: 10.1021/acs.macromol.0c00642] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Andreas Schaffer
- WACKER-Chair of Macromolecular Chemistry, Technical University of Munich, Lichtenbergstraße 4, 85748 Garching near Munich, Germany
| | - Moritz Kränzlein
- WACKER-Chair of Macromolecular Chemistry, Technical University of Munich, Lichtenbergstraße 4, 85748 Garching near Munich, Germany
| | - Bernhard Rieger
- WACKER-Chair of Macromolecular Chemistry, Technical University of Munich, Lichtenbergstraße 4, 85748 Garching near Munich, Germany
| |
Collapse
|
286
|
Stereospecific Si-C coupling and remote control of axial chirality by enantioselective palladium-catalyzed hydrosilylation of maleimides. Nat Commun 2020; 11:2904. [PMID: 32518227 PMCID: PMC7283218 DOI: 10.1038/s41467-020-16716-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/20/2020] [Indexed: 01/23/2023] Open
Abstract
Hydrosilylation of unsaturated carbon-carbon bonds with hydrosilanes is a very important process to access organosilicon compounds and ranks as one of the most fundamental reactions in organic chemistry. However, catalytic asymmetric hydrosilylation of activated alkenes and internal alkenes has proven elusive, due to competing reduction of carbon-carbon double bond or isomerization processes. Herein, we report a highly enantioselective Si-C coupling by hydrosilylation of carbonyl-activated alkenes using a palladium catalyst with a chiral TADDOL-derived phosphoramidite ligand, which inhibits O-hydrosilylation/olefin reduction. The stereospecific Si-C coupling/hydrosilylation of maleimides affords a series of silyl succinimides with up to 99% yield, >99:1 diastereoselectivity and >99:1 enantioselectivity. The high degree of stereoselectivity exerts remote control of axial chirality, leading to functionalized, axially chiral succinimides which are versatile building blocks. The product utility is highlighted by the enantioselective construction of N-heterocycles bearing up to three stereocenters. Catalytic asymmetric hydrosilylation of internal alkenes has proven elusive due to more favourable double bond reduction or isomerization. Here, the authors show an enantioselective Si-C coupling by hydrosilylation of activated alkenes using a palladium/phosphoramidite catalyst affording axially chiral succinimides.
Collapse
|
287
|
McAulay K, Hoyt EA, Thomas M, Schimpl M, Bodnarchuk MS, Lewis HJ, Barratt D, Bhavsar D, Robinson DM, Deery MJ, Ogg DJ, Bernardes GJL, Ward RA, Waring MJ, Kettle JG. Alkynyl Benzoxazines and Dihydroquinazolines as Cysteine Targeting Covalent Warheads and Their Application in Identification of Selective Irreversible Kinase Inhibitors. J Am Chem Soc 2020; 142:10358-10372. [PMID: 32412754 DOI: 10.1021/jacs.9b13391] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
With a resurgence in interest in covalent drugs, there is a need to identify new moieties capable of cysteine bond formation that are differentiated from commonly employed systems such as acrylamide. Herein, we report on the discovery of new alkynyl benzoxazine and dihydroquinazoline moieties capable of covalent reaction with cysteine. Their utility as alternative electrophilic warheads for chemical biological probes and drug molecules is demonstrated through site-selective protein modification and incorporation into kinase drug scaffolds. A potent covalent inhibitor of JAK3 kinase was identified with superior selectivity across the kinome and improvements in in vitro pharmacokinetic profile relative to the related acrylamide-based inhibitor. In addition, the use of a novel heterocycle as a cysteine reactive warhead is employed to target Cys788 in c-KIT, where acrylamide has previously failed to form covalent interactions. These new reactive and selective heterocyclic warheads supplement the current repertoire for cysteine covalent modification while avoiding some of the limitations generally associated with established moieties.
Collapse
Affiliation(s)
| | - Emily A Hoyt
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | | | - Marianne Schimpl
- Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge CB4 0WG, U.K
| | | | | | - Derek Barratt
- Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Deepa Bhavsar
- Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | | | - Michael J Deery
- Cambridge Centre for Proteomics, Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, U.K
| | - Derek J Ogg
- Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Gonçalo J L Bernardes
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.,Instituto de Medicina Molecular, Faculdade de Medicina de Universidad de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | | | - Michael J Waring
- Northern Institute for Cancer Research, Chemistry, School of Natural and Environmental Sciences, Newcastle University, Bedson Building, Newcastle upon Tyne NE1 7RU, U.K
| | | |
Collapse
|
288
|
Gahoual R, Bolbach G, Ould-Melha I, Clodic G, François YN, Scherman D, Mignet N, Houzé P. Kinetic and structural characterization of therapeutic albumin chemical functionalization using complementary mass spectrometry techniques. J Pharm Biomed Anal 2020; 185:113242. [DOI: 10.1016/j.jpba.2020.113242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/03/2020] [Accepted: 03/05/2020] [Indexed: 01/06/2023]
|
289
|
Synthesis of Pharmacologically Relevant New Derivatives of Maleimides via Ligand-Free Pd-Catalyzed Suzuki–Miyaura Cross-Coupling Reactions. ARABIAN JOURNAL FOR SCIENCE AND ENGINEERING 2020. [DOI: 10.1007/s13369-020-04450-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
290
|
Sepay N, Sepay N, Al Hoque A, Mondal R, Halder UC, Muddassir M. In silico fight against novel coronavirus by finding chromone derivatives as inhibitor of coronavirus main proteases enzyme. Struct Chem 2020; 31:1831-1840. [PMID: 32412544 PMCID: PMC7220622 DOI: 10.1007/s11224-020-01537-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/13/2020] [Indexed: 01/10/2023]
Abstract
Novel coronavirus, 2019-nCoV is a danger to the world and is spreading rapidly. Very little structural information about 2019-nCoV make this situation more difficult for drug designing. Benzylidenechromanones, naturally occurring oxygen heterocyclic compounds, having capability to inhibit various protein and receptors, have been designed here to block mutant variety of coronavirus main protease enzyme (SARC-CoV-2 Mpro) isolated from 2019-nCoV with the assistance of molecular docking, bioinformatics and molecular electrostatic potential. (Z)-3-(4'-chlorobenzylidene)-thiochroman-4-one showed highest binding affinity to the protein. Binding of a compound to this protein actually inhibits the replication and transcription of the virus and, ultimately, stop the virus multiplication. Incorporation of any functional groups to the basic benzylidenechromanones enhances their binding ability. Chloro and bromo substitutions amplify the binding affinity. ADME studies of all these compounds indicate they are lipophilic, high gastro intestine absorbable and blood-brain barrier permeable. The outcome reveals that the investigated benzylidenechromanones can be examined in the case of 2019-nCoV as potent inhibitory drug of SARC-CoV-2 Mpro, for their strong inhibition ability, high reactivity and effective pharmacological properties.
Collapse
Affiliation(s)
- Nayim Sepay
- Department of Chemistry, Jadavpur University, Kolkata, 700032 India
| | - Nadir Sepay
- Plant Biotechnology Laboratory, Post Graduate Department of Botany, Ramakrishna Mission Vivekananda Centenary College, Rahara, Kolkata, 700118 India
| | - Ashique Al Hoque
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032 India
| | - Rina Mondal
- Department of Chemistry, Uluberia College, Howrah, West Bengal 711 315 India
| | | | - Mohd. Muddassir
- Department of Chemistry, College of Science, King Saud University, Riyadh, 11451 Saudi Arabia
| |
Collapse
|
291
|
Tessier R, Nandi RK, Dwyer BG, Abegg D, Sornay C, Ceballos J, Erb S, Cianférani S, Wagner A, Chaubet G, Adibekian A, Waser J. Ethynylation of Cysteine Residues: From Peptides to Proteins in Vitro and in Living Cells. Angew Chem Int Ed Engl 2020; 59:10961-10970. [DOI: 10.1002/anie.202002626] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Romain Tessier
- Laboratory of Catalysis and Organic SynthesisEcole Polytechnique Fédérale de Lausanne EPFL SB ISIC LCSO, BCH 4306 1015 Lausanne Switzerland
- Present address: Department of Chemical BiologyMax Planck Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Raj Kumar Nandi
- Laboratory of Catalysis and Organic SynthesisEcole Polytechnique Fédérale de Lausanne EPFL SB ISIC LCSO, BCH 4306 1015 Lausanne Switzerland
- Present address: Department of ChemistryDiamond Harbour Women's University Sarisha South 24 Parganas West Bengal 743368 India
| | - Brendan G. Dwyer
- Department of ChemistryThe Scripps Research Institute 130 Scripps Way Jupiter FL 33458 USA
| | - Daniel Abegg
- Department of ChemistryThe Scripps Research Institute 130 Scripps Way Jupiter FL 33458 USA
| | - Charlotte Sornay
- Bio-Functional Chemistry (UMR 7199)LabEx Medalis, University of Strasbourg 74 Route du Rhin 67400 Illkirch-Graffenstaden France
| | - Javier Ceballos
- Laboratory of Catalysis and Organic SynthesisEcole Polytechnique Fédérale de Lausanne EPFL SB ISIC LCSO, BCH 4306 1015 Lausanne Switzerland
| | - Stéphane Erb
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO)Université de StrasbourgCNRS, IPHC UMR 7178 67000 Strasbourg France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO)Université de StrasbourgCNRS, IPHC UMR 7178 67000 Strasbourg France
| | - Alain Wagner
- Bio-Functional Chemistry (UMR 7199)LabEx Medalis, University of Strasbourg 74 Route du Rhin 67400 Illkirch-Graffenstaden France
| | - Guilhem Chaubet
- Bio-Functional Chemistry (UMR 7199)LabEx Medalis, University of Strasbourg 74 Route du Rhin 67400 Illkirch-Graffenstaden France
| | - Alexander Adibekian
- Department of ChemistryThe Scripps Research Institute 130 Scripps Way Jupiter FL 33458 USA
| | - Jerome Waser
- Laboratory of Catalysis and Organic SynthesisEcole Polytechnique Fédérale de Lausanne EPFL SB ISIC LCSO, BCH 4306 1015 Lausanne Switzerland
| |
Collapse
|
292
|
Tessier R, Nandi RK, Dwyer BG, Abegg D, Sornay C, Ceballos J, Erb S, Cianférani S, Wagner A, Chaubet G, Adibekian A, Waser J. Ethynylation of Cysteine Residues: From Peptides to Proteins in Vitro and in Living Cells. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202002626] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Romain Tessier
- Laboratory of Catalysis and Organic SynthesisEcole Polytechnique Fédérale de Lausanne EPFL SB ISIC LCSO, BCH 4306 1015 Lausanne Switzerland
- Present address: Department of Chemical BiologyMax Planck Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Raj Kumar Nandi
- Laboratory of Catalysis and Organic SynthesisEcole Polytechnique Fédérale de Lausanne EPFL SB ISIC LCSO, BCH 4306 1015 Lausanne Switzerland
- Present address: Department of ChemistryDiamond Harbour Women's University Sarisha South 24 Parganas West Bengal 743368 India
| | - Brendan G. Dwyer
- Department of ChemistryThe Scripps Research Institute 130 Scripps Way Jupiter FL 33458 USA
| | - Daniel Abegg
- Department of ChemistryThe Scripps Research Institute 130 Scripps Way Jupiter FL 33458 USA
| | - Charlotte Sornay
- Bio-Functional Chemistry (UMR 7199)LabEx Medalis, University of Strasbourg 74 Route du Rhin 67400 Illkirch-Graffenstaden France
| | - Javier Ceballos
- Laboratory of Catalysis and Organic SynthesisEcole Polytechnique Fédérale de Lausanne EPFL SB ISIC LCSO, BCH 4306 1015 Lausanne Switzerland
| | - Stéphane Erb
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO)Université de StrasbourgCNRS, IPHC UMR 7178 67000 Strasbourg France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO)Université de StrasbourgCNRS, IPHC UMR 7178 67000 Strasbourg France
| | - Alain Wagner
- Bio-Functional Chemistry (UMR 7199)LabEx Medalis, University of Strasbourg 74 Route du Rhin 67400 Illkirch-Graffenstaden France
| | - Guilhem Chaubet
- Bio-Functional Chemistry (UMR 7199)LabEx Medalis, University of Strasbourg 74 Route du Rhin 67400 Illkirch-Graffenstaden France
| | - Alexander Adibekian
- Department of ChemistryThe Scripps Research Institute 130 Scripps Way Jupiter FL 33458 USA
| | - Jerome Waser
- Laboratory of Catalysis and Organic SynthesisEcole Polytechnique Fédérale de Lausanne EPFL SB ISIC LCSO, BCH 4306 1015 Lausanne Switzerland
| |
Collapse
|
293
|
Richardson MB, Gabriel KN, Garcia JA, Ashby SN, Dyer RP, Kim JK, Lau CJ, Hong J, Le Tourneau RJ, Sen S, Narel DL, Katz BB, Ziller JW, Majumdar S, Collins PG, Weiss GA. Pyrocinchonimides Conjugate to Amine Groups on Proteins via Imide Transfer. Bioconjug Chem 2020; 31:1449-1462. [PMID: 32302483 DOI: 10.1021/acs.bioconjchem.0c00143] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Advances in bioconjugation, the ability to link biomolecules to each other, small molecules, surfaces, and more, can spur the development of advanced materials and therapeutics. We have discovered that pyrocinchonimide, the dimethylated analogue of maleimide, undergoes a surprising transformation with biomolecules. The reaction targets amines and involves an imide transfer, which has not been previously reported for bioconjugation purposes. Despite their similarity to maleimides, pyrocinchonimides do not react with free thiols. Though both lysine residues and the N-termini of proteins can receive the transferred imide, the reaction also exhibits a marked preference for certain amines that cannot solely be ascribed to solvent accessibility. This property is peculiar among amine-targeting reactions and can reduce combinatorial diversity when many available reactive amines are available, such as in the formation of antibody-drug conjugates. Unlike amides, the modification undergoes very slow reversion under high pH conditions. The reaction offers a thermodynamically controlled route to single or multiple modifications of proteins for a wide range of applications.
Collapse
Affiliation(s)
- Mark B Richardson
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Kristin N Gabriel
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Joseph A Garcia
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Shareen N Ashby
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Rebekah P Dyer
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Joshua K Kim
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Calvin J Lau
- Department of Physics & Astronomy, University of California, Irvine, Irvine, California 92697, United States
| | - John Hong
- School of Medicine, University of California, Irvine, Irvine, California 92697, United States
| | - Ryan J Le Tourneau
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Sanjana Sen
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, California 92697, United States
| | - David L Narel
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Benjamin B Katz
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Joseph W Ziller
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Sudipta Majumdar
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Philip G Collins
- Department of Physics & Astronomy, University of California, Irvine, Irvine, California 92697, United States
| | - Gregory A Weiss
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States.,Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, California 92697, United States
| |
Collapse
|
294
|
Abstract
Immuno-positron emission tomography (immunoPET) is a paradigm-shifting molecular imaging modality combining the superior targeting specificity of monoclonal antibody (mAb) and the inherent sensitivity of PET technique. A variety of radionuclides and mAbs have been exploited to develop immunoPET probes, which has been driven by the development and optimization of radiochemistry and conjugation strategies. In addition, tumor-targeting vectors with a short circulation time (e.g., Nanobody) or with an enhanced binding affinity (e.g., bispecific antibody) are being used to design novel immunoPET probes. Accordingly, several immunoPET probes, such as 89Zr-Df-pertuzumab and 89Zr-atezolizumab, have been successfully translated for clinical use. By noninvasively and dynamically revealing the expression of heterogeneous tumor antigens, immunoPET imaging is gradually changing the theranostic landscape of several types of malignancies. ImmunoPET is the method of choice for imaging specific tumor markers, immune cells, immune checkpoints, and inflammatory processes. Furthermore, the integration of immunoPET imaging in antibody drug development is of substantial significance because it provides pivotal information regarding antibody targeting abilities and distribution profiles. Herein, we present the latest immunoPET imaging strategies and their preclinical and clinical applications. We also emphasize current conjugation strategies that can be leveraged to develop next-generation immunoPET probes. Lastly, we discuss practical considerations to tune the development and translation of immunoPET imaging strategies.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705, United States
| |
Collapse
|
295
|
Liu S, Jin R, Wang M, Li G. Nanoparticle Delivery of CD147 Antagonistic Peptide-9 Protects against Acute Ischemic Brain Injury and tPA-Induced Intracerebral Hemorrhage in Mice. ACS APPLIED BIO MATERIALS 2020; 3:1976-1985. [PMID: 34124605 PMCID: PMC8195622 DOI: 10.1021/acsabm.9b01141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
CD147 has emerged as a potential therapeutic target in many human diseases. We have demonstrated that inhibition of CD147 using its function-blocking antibody ameliorates acute ischemic brain injury and promotes long-term functional recovery in mice. Recently, peptide-nanoparticle conjugates have emerged as powerful tools for biomedical applications. The present study aimed to investigate the therapeutic potential of CD147 antagonist peptide-9 (AP9) in acute ischemic stroke in mice using nanomaterial as the drug delivery vehicles. AP9-conjugated nanoparticles (APN), with an average size of about 40 nm, were fabricated by maleimide linkage and characterized using dynamic light scattering and transmission electron microscopy. We found that APN specifically bound to CD147 in cultured mouse brain endothelial cells (bEnd.3) and to ischemia-induced CD147 in mouse cerebral microvessels. Using a mouse model of transient middle cerebral artery occlusion (tMCAO), we demonstrated, for the first time, that systemic delivery of APN (2.5 mg/kg, I.V.) initiated at 1 h after tMCAO significantly reduced brain infarct size, improved functional outcome, and attenuated delayed (5 h after tMCAO) tPA-induced intracerebral hemorrhage in acute ischemic stroke. These protective effects were associated with profound inhibition of MMP-9 and MMP-3 in both ischemic brain and plasma. In conclusion, the CD147 antagonist peptide-9 represents a potentially promising therapeutic candidate for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Shan Liu
- Department of Neurosurgery, The Pennsylvania State, University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Rong Jin
- Department of Neurosurgery, The Pennsylvania State, University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Min Wang
- Department of Neurosurgery, The Pennsylvania State, University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Guohong Li
- Department of Neurosurgery, The Pennsylvania State, University College of Medicine, Hershey, Pennsylvania 17033, United States
| |
Collapse
|
296
|
Chen HA, Ma YH, Hsu TY, Chen JP. Preparation of Peptide and Recombinant Tissue Plasminogen Activator Conjugated Poly(Lactic-Co-Glycolic Acid) (PLGA) Magnetic Nanoparticles for Dual Targeted Thrombolytic Therapy. Int J Mol Sci 2020; 21:2690. [PMID: 32294917 PMCID: PMC7215398 DOI: 10.3390/ijms21082690] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/11/2022] Open
Abstract
Recombinant tissue plasminogen activator (rtPA) is the only thrombolytic agent that has been approved by the FDA for treatment of ischemic stroke. However, a high dose intravenous infusion is required to maintain effective drug concentration, owing to the short half-life of the thrombolytic drug, whereas a momentous limitation is the risk of bleeding. We envision a dual targeted strategy for rtPA delivery will be feasible to minimize the required dose of rtPA for treatment. For this purpose, rtPA and fibrin-avid peptide were co-immobilized to poly(lactic-co-glycolic acid) (PLGA) magnetic nanoparticles (PMNP) to prepare peptide/rtPA conjugated PMNPs (pPMNP-rtPA). During preparation, PMNP was first surface modified with avidin, which could interact with biotin. This is followed by binding PMNP-avidin with biotin-PEG-rtPA (or biotin-PEG-peptide), which was prepared beforehand by binding rtPA (or peptide) to biotin-PEG-maleimide while using click chemistry between maleimide and the single -SH group in rtPA (or peptide). The physicochemical property characterization indicated the successful preparation of the magnetic nanoparticles with full retention of rtPA fibrinolysis activity, while biological response studies underlined the high biocompatibility of all magnetic nanoparticles from cytotoxicity and hemolysis assays in vitro. The magnetic guidance and fibrin binding effects were also confirmed, which led to a higher thrombolysis rate in vitro using PMNP-rtPA or pPMNP-rtPA when compared to free rtPA after static or dynamic incubation with blood clots. Using pressure-dependent clot lysis model in a flow system, dual targeted pPMNP-rtPA could reduce the clot lysis time for reperfusion by 40% when compared to free rtPA at the same drug dosage. From in vivo targeted thrombolysis in a rat embolic model, pPMNP-rtPA was used at 20% of free rtPA dosage to restore the iliac blood flow in vascular thrombus that was created by injecting a blood clot to the hind limb area.
Collapse
Affiliation(s)
- Huai-An Chen
- Department of Chemical and Materials and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan;
| | - Yunn-Hwa Ma
- Department of Physiology and Pharmacology and Healthy Aging Research Center, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (Y.-H.M.); (T.-Y.H.)
| | - Tzu-Yuan Hsu
- Department of Physiology and Pharmacology and Healthy Aging Research Center, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (Y.-H.M.); (T.-Y.H.)
| | - Jyh-Ping Chen
- Department of Chemical and Materials and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan;
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan
- Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33302, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan
| |
Collapse
|
297
|
Gessner I, Neundorf I. Nanoparticles Modified with Cell-Penetrating Peptides: Conjugation Mechanisms, Physicochemical Properties, and Application in Cancer Diagnosis and Therapy. Int J Mol Sci 2020; 21:E2536. [PMID: 32268473 PMCID: PMC7177461 DOI: 10.3390/ijms21072536] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/28/2020] [Accepted: 04/03/2020] [Indexed: 12/27/2022] Open
Abstract
Based on their tunable physicochemical properties and the possibility of producing cell-specific platforms through surface modification with functional biomolecules, nanoparticles (NPs) represent highly promising tools for biomedical applications. To improve their potential under physiological conditions and to enhance their cellular uptake, combinations with cell-penetrating peptides (CPPs) represent a valuable strategy. CPPs are often cationic peptide sequences that are able to translocate across biological membranes and to carry attached cargos inside cells and have thus been recognized as versatile tools for drug delivery. Nevertheless, the conjugation of CPP to NP surfaces is dependent on many properties from both individual components, and further insight into this complex interplay is needed to allow for the fabrication of highly stable but functional vectors. Since CPPs per se are nonselective and enter nearly all cells likewise, additional decoration of NPs with homing devices, such as tumor-homing peptides, enables the design of multifunctional platforms for the targeted delivery of chemotherapeutic drugs. In this review, we have updated the recent advances in the field of CPP-NPs, focusing on synthesis strategies, elucidating the influence of different physicochemical properties, as well as their application in cancer research.
Collapse
Affiliation(s)
- Isabel Gessner
- Department of Chemistry, Inorganic Chemistry, University of Cologne, Greinstr 6, 50939 Cologne, Germany;
| | - Ines Neundorf
- Department of Chemistry, Biochemistry, University of Cologne, Zuelpicher Str. 47a, 50674 Cologne, Germany
| |
Collapse
|
298
|
Meier-Menches SM, Casini A. Design Strategies and Medicinal Applications of Metal-Peptidic Bioconjugates. Bioconjug Chem 2020; 31:1279-1288. [DOI: 10.1021/acs.bioconjchem.0c00152] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Samuel M. Meier-Menches
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 38, 1090 Vienna, Austria
| | - Angela Casini
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, Technical University of Munich, Lichtenbergstrasse 4, 85748 Garching, Germany
| |
Collapse
|
299
|
Quirós-Ovies R, Vázquez Sulleiro M, Vera-Hidalgo M, Prieto J, Gómez IJ, Sebastián V, Santamaría J, Pérez EM. Controlled Covalent Functionalization of 2 H-MoS 2 with Molecular or Polymeric Adlayers. Chemistry 2020; 26:6629-6634. [PMID: 32101348 DOI: 10.1002/chem.202000068] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/25/2020] [Indexed: 01/01/2023]
Abstract
Most air-stable 2D materials are relatively inert, which makes their chemical modification difficult. In particular, in the case of MoS2 , the semiconducting 2 H-MoS2 is much less reactive than its metallic counterpart, 1T-MoS2 . As a consequence, there are hardly any reliable methods for the covalent modification of 2 H-MoS2 . An ideal method for the chemical functionalization of such materials should be both mild, not requiring the introduction of a large number of defects, and versatile, allowing for the decoration with as many different functional groups as possible. Herein, a comprehensive study on the covalent functionalization of 2 H-MoS2 with maleimides is presented. The use of a base (Et3 N) leads to the in situ formation of a succinimide polymer layer, covalently connected to MoS2 . In contrast, in the absence of base, functionalization stops at the molecular level. Moreover, the functionalization protocol is mild (occurs at room temperature), fast (nearly complete in 1 h), and very flexible (11 different solvents and 10 different maleimides tested). In practical terms, the procedures described here allow for the chemist to manipulate 2 H-MoS2 in a very flexible way, decorating it with polymers or molecules, and with a wide range of functional groups for subsequent modification. Conceptually, the spurious formation of an organic polymer might be general to other methods of functionalization of 2D materials, where a large excess of molecular reagents is typically used.
Collapse
Affiliation(s)
- Ramiro Quirós-Ovies
- IMDEA Nanociencia, Ciudad Universitaria de Cantoblanco, C/Faraday 9, 28049, Madrid, Spain
| | | | - Mariano Vera-Hidalgo
- IMDEA Nanociencia, Ciudad Universitaria de Cantoblanco, C/Faraday 9, 28049, Madrid, Spain
| | - Javier Prieto
- IMDEA Nanociencia, Ciudad Universitaria de Cantoblanco, C/Faraday 9, 28049, Madrid, Spain
| | - I Jénnifer Gómez
- CEITEC Masaryk University, Kamenice 5, 62500, Brno, Czech Republic
| | - Víctor Sebastián
- Department of Chemical and Environmental Engineering, Institute of Nanoscience of Aragon (INA), Campus Rio Ebro, 50018, Zaragoza, Spain.,Instituto de Ciencia de Materiales de Aragon (ICMA), Consejo Superior de Investigaciones Científicas, (CSIC-Universidad de Zaragoza), 50018, Zaragoza, Spain.,Networking Research Center on Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| | - Jesús Santamaría
- Department of Chemical and Environmental Engineering, Institute of Nanoscience of Aragon (INA), Campus Rio Ebro, 50018, Zaragoza, Spain.,Instituto de Ciencia de Materiales de Aragon (ICMA), Consejo Superior de Investigaciones Científicas, (CSIC-Universidad de Zaragoza), 50018, Zaragoza, Spain.,Networking Research Center on Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| | - Emilio M Pérez
- IMDEA Nanociencia, Ciudad Universitaria de Cantoblanco, C/Faraday 9, 28049, Madrid, Spain
| |
Collapse
|
300
|
Thiol-Reactive PODS-Bearing Bifunctional Chelators for the Development of EGFR-Targeting [ 18F]AlF-Affibody Conjugates. Molecules 2020; 25:molecules25071562. [PMID: 32235296 PMCID: PMC7180749 DOI: 10.3390/molecules25071562] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 01/18/2023] Open
Abstract
Site-selective bioconjugation of cysteine-containing peptides and proteins is currently achieved via a maleimide–thiol reaction (Michael addition). When maleimide-functionalized chelators are used and the resulting bioconjugates are subsequently radiolabeled, instability has been observed both during radiosynthesis and post-injection in vivo, reducing radiochemical yield and negatively impacting performance. Recently, a phenyloxadiazolyl methylsulfone derivative (PODS) was proposed as an alternative to maleimide for the site-selective conjugation and radiolabeling of proteins, demonstrating improved in vitro stability and in vivo performance. Therefore, we have synthesized two novel PODS-bearing bifunctional chelators (NOTA-PODS and NODAGA-PODS) and attached them to the EGFR-targeting affibody molecule ZEGFR:03115. After radiolabeling with the aluminum fluoride complex ([18F]AlF), both conjugates showed good stability in murine serum. When injected in high EGFR-expressing tumor-bearing mice, [18F]AlF-NOTA-PODS-ZEGFR:03115 and [18F]AlF-NODAGA-PODS-ZEGFR:03115 showed similar pharmacokinetics and a specific tumor uptake of 14.1 ± 5.3% and 16.7 ± 4.5% ID/g at 1 h post-injection, respectively. The current results are encouraging for using PODS as an alternative to maleimide-based thiol-selective bioconjugation reactions.
Collapse
|