1
|
Collins M, Ibeanu N, Grabowska WR, Awwad S, Khaw PT, Brocchini S, Khalili H. Bispecific FpFs: a versatile tool for preclinical antibody development. RSC Chem Biol 2024:d4cb00130c. [PMID: 39347456 PMCID: PMC11427889 DOI: 10.1039/d4cb00130c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/08/2024] [Indexed: 10/01/2024] Open
Abstract
We previously described FpFs 1̲ (Fab-PEG-Fab) as binding mimetics of IgGs. FpFs are prepared with di(bis-sulfone) conjugation reagents 3̲ that undergo disulfide rebridging conjugation with the accessible disulfide of each Fab (Scheme 1). We have now prepared bispecific FpFs 2̲ (bsFpF and Fab1-PEG-Fab2) as potential bispecific antibody mimetics with the intent that bsFpFs could be used in preclinical antibody development since sourcing bispecific antibodies may be challenging during preclinical research. The di(bis-sulfone) reagent 3̲ was first used to prepare a bsFpF 2̲ by the sequential conjugation of a first Fab and then a second Fab to another target (Scheme 2). Seeking to improve bsFpF synthesis, the asymmetric conjugation reagent, bis-sulfone bis-sulfide 1̲6̲, with different thiol conjugation reactivities at each terminus (Scheme 4) was examined and the bsFpFs appeared to be formed at similar conversion to the di(bis-sulfone) reagent 3̲. To explore the advantages of using common intermediates in the preparation of bsFpF families, we investigated bsFpF synthesis with a protein conjugation-ligation approach (Scheme 5). Reagents with a bis-sulfone moiety for conjugation on one PEG terminus and a ligation moiety on the other terminus were examined. Bis-sulfone PEG trans-cyclooctene (TCO) 2̲8̲ and bis-sulfone PEG tetrazine (Tz) 3̲0̲ were used to prepare several bsFpFs targeting various therapeutic targets (TNF-α, IL6R, IL17, and VEGF) and tissue affinity targets (hyaluronic acid and collagen II). Surface plasmon resonance (SPR) binding studies indicated that there was little difference between the dissociation rate constant (k d) for the unmodified Fab, mono-conjugated PEG-Fab and the corresponding Fab in a bsFpF. The Fab association rate (k a) in the bsFpF was slower than for PEG-Fab, which may be because of mass differences that influence SPR results. These observations suggest that each Fab will bind to its target independently of the other Fab and that bsFpF binding profiles can be estimated using the corresponding PEG-Fab conjugates.
Collapse
Affiliation(s)
- Matthew Collins
- School of Health, Sport and Bioscience, University of East London London UK
| | - Nkiru Ibeanu
- School of Pharmacy, University College London London UK
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology London EC1V 9EL UK
| | | | - Sahar Awwad
- School of Pharmacy, University College London London UK
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology London EC1V 9EL UK
| | - Peng T Khaw
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology London EC1V 9EL UK
| | | | - Hanieh Khalili
- School of Pharmacy, University College London London UK
- School of Biomedical Science, University of West London London W5 5RF UK
| |
Collapse
|
2
|
Giacon N, Lo Cascio E, Pennacchietti V, De Maio F, Santarelli G, Sibilia D, Tiberio F, Sanguinetti M, Lattanzi W, Toto A, Arcovito A. PDZ2-conjugated-PLGA nanoparticles are tiny heroes in the battle against SARS-CoV-2. Sci Rep 2024; 14:13059. [PMID: 38844490 PMCID: PMC11156922 DOI: 10.1038/s41598-024-63239-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/27/2024] [Indexed: 06/09/2024] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has highlighted the urgent need for innovative antiviral strategies to fight viral infections. Although a substantial part of the overall effort has been directed at the Spike protein to create an effective global vaccination strategy, other proteins have also been examined and identified as possible therapeutic targets. Among them, although initially underestimated, there is the SARS-CoV-2 E-protein, which turned out to be a key factor in viral pathogenesis due to its role in virus budding, assembly and spreading. The C-terminus of E-protein contains a PDZ-binding motif (PBM) that plays a key role in SARS-CoV-2 virulence as it is recognized and bound by the PDZ2 domain of the human tight junction protein ZO-1. The binding between the PDZ2 domain of ZO-1 and the C-terminal portion of SARS-CoV-2 E-protein has been extensively characterized. Our results prompted us to develop a possible adjuvant therapeutic strategy aimed at slowing down or inhibiting virus-mediated pathogenesis. Such innovation consists in the design and synthesis of externally PDZ2-ZO1 functionalized PLGA-based nanoparticles to be used as intracellular decoy. Contrary to conventional strategies, this innovative approach aims to capitalize on the E protein-PDZ2 interaction to prevent virus assembly and replication. In fact, the conjugation of the PDZ2 domain to polymeric nanoparticles increases the affinity toward the E protein effectively creating a "molecular sponge" able to sequester E proteins within the intracellular environment of infected cells. Our in vitro studies on selected cellular models, show that these nanodevices significantly reduce SARS-CoV-2-mediated virulence, emphasizing the importance of exploiting viral-host interactions for therapeutic benefit.
Collapse
Affiliation(s)
- Noah Giacon
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy
| | - Ettore Lo Cascio
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy
| | - Valeria Pennacchietti
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Sapienza Università di Roma, P.le A. Moro 5, 00185, Rome, Italy
| | - Flavio De Maio
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
| | - Giulia Santarelli
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
| | - Diego Sibilia
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy
| | - Federica Tiberio
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy
| | - Maurizio Sanguinetti
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
| | - Wanda Lattanzi
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
| | - Angelo Toto
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Sapienza Università di Roma, P.le A. Moro 5, 00185, Rome, Italy.
| | - Alessandro Arcovito
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy.
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy.
| |
Collapse
|
3
|
Bento C, Katz M, Santos MMM, Afonso CAM. Striving for Uniformity: A Review on Advances and Challenges To Achieve Uniform Polyethylene Glycol. Org Process Res Dev 2024; 28:860-890. [PMID: 38660381 PMCID: PMC11036406 DOI: 10.1021/acs.oprd.3c00428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 04/26/2024]
Abstract
Poly(ethylene glycol) (PEG) is the polymer of choice in drug delivery systems due to its biocompatibility and hydrophilicity. For over 20 years, this polymer has been widely used in the drug delivery of small drugs, proteins, oligonucleotides, and liposomes, improving the stability and pharmacokinetics of many drugs. However, despite the extensive clinical experience with PEG, concerns have emerged related to its use. These include hypersensitivity, purity, and nonbiodegradability. Moreover, conventional PEG is a mixture of polymers that can complicate drug synthesis and purification leading to unwanted immunogenic reactions. Studies have shown that uniform PEGylated drugs may be more effective than conventional PEGylated drugs as they can overcome issues related to molecular heterogeneity and immunogenicity. This has led to significant research efforts to develop synthetic procedures to produce uniform PEGs (monodisperse PEGs). As a result, iterative step-by-step controlled synthesis methods have been created over time and have shown promising results. Nonetheless, these procedures have presented numerous challenges due to their iterative nature and the requirement for multiple purification steps, resulting in increased costs and time consumption. Despite these challenges, the synthetic procedures went through several improvements. This review summarizes and discusses recent advances in the synthesis of uniform PEGs and its derivatives with a focus on overall yields, scalability, and purity of the polymers. Additionally, the available characterization methods for assessing polymer monodispersity are discussed as well as uniform PEG applications, side effects, and possible alternative polymers that can overcome the drawbacks.
Collapse
Affiliation(s)
- Cláudia Bento
- Hovione
Farmaciência S.A., Estrada do Paço do Lumiar, Campus do Lumiar, Edifício
R, 1649-038 Lisboa, Portugal
- Research
Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Marianna Katz
- Hovione
Farmaciência S.A., Estrada do Paço do Lumiar, Campus do Lumiar, Edifício
R, 1649-038 Lisboa, Portugal
| | - Maria M. M. Santos
- Research
Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Carlos A. M. Afonso
- Research
Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| |
Collapse
|
4
|
Zhang X, Geng C, Fu Y, Lv Z, Wei Z, Wang H, Wang S. A Facile and Green Approach for the Preparation of Amine-Functionalized Poly(ethylene glycol) by Reducing Poly(ethylene glycol) Azide with Dithiothreitol. Biomacromolecules 2024; 25:1972-1977. [PMID: 38359265 DOI: 10.1021/acs.biomac.3c01370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
A facile and green approach for the preparation of PEGn-NH2s from PEGn-N3s in water with DTT as the reduction reagent has been developed, avoiding the introduction of metal ions and difficulties in purification compared to the traditional synthesis process of PEGn-NH2s. A series of high-purity linear and multiarm PEGn-NH2s with different molecular weights were synthesized, demonstrating the versatility of this method. Additionally, HS-PEG45-NH2 with high fidelity of thiol and amine was easily prepared through the one-step two functional group conversion of N3-PEG45-S-S-PEG45-N3, and the PEG-based NH2-PEG@AuNPs were also prepared. This technology will promote the application of PEGn-NH2s in the fields of medicine and biomaterials.
Collapse
Affiliation(s)
- Xuemei Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. of China
| | - Chao Geng
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. of China
| | - Yang Fu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. of China
| | - Zhen Lv
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. of China
| | - Zhonglin Wei
- Department of Organic Chemistry, College of Chemistry, Jilin University, Changchun 130021, Jilin, P.R. of China
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. of China
| | - Shixue Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. of China
| |
Collapse
|
5
|
Li C, Li T, Tian X, An W, Wang Z, Han B, Tao H, Wang J, Wang X. Research progress on the PEGylation of therapeutic proteins and peptides (TPPs). Front Pharmacol 2024; 15:1353626. [PMID: 38523641 PMCID: PMC10960368 DOI: 10.3389/fphar.2024.1353626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/22/2024] [Indexed: 03/26/2024] Open
Abstract
With the rapid advancement of genetic and protein engineering, proteins and peptides have emerged as promising drug molecules for therapeutic applications. Consequently, there has been a growing interest in the field of chemical modification technology to address challenges associated with their clinical use, including rapid clearance from circulation, immunogenicity, physical and chemical instabilities (such as aggregation, adsorption, deamination, clipping, oxidation, etc.), and enzymatic degradation. Polyethylene glycol (PEG) modification offers an effective solution to these issues due to its favorable properties. This review presents recent progress in the development and application of PEGylated therapeutic proteins and peptides (TPPs). For this purpose, firstly, the physical and chemical properties as well as classification of PEG and its derivatives are described. Subsequently, a detailed summary is provided on the main sites of PEGylated TPPs and the factors that influence their PEGylation. Furthermore, notable instances of PEG-modified TPPs (including antimicrobial peptides (AMPs), interferon, asparaginase and antibodies) are highlighted. Finally, we propose the chemical modification of TPPs with PEG, followed by an analysis of the current development status and future prospects of PEGylated TPPs. This work provides a comprehensive literature review in this promising field while facilitating researchers in utilizing PEG polymers to modify TPPs for disease treatment.
Collapse
Affiliation(s)
- Chunxiao Li
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Ting Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, China
| | - Xinya Tian
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Wei An
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Zhenlong Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Bing Han
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Hui Tao
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Jinquan Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Xiumin Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, China
| |
Collapse
|
6
|
Rochet LNC, Bahou C, Wojciechowski JP, Koutsopetras I, Britton P, Spears RJ, Thanasi IA, Shao B, Zhong L, Bučar DK, Aliev AE, Porter MJ, Stevens MM, Baker JR, Chudasama V. Use of pyridazinediones for tuneable and reversible covalent cysteine modification applied to peptides, proteins and hydrogels. Chem Sci 2023; 14:13743-13754. [PMID: 38075666 PMCID: PMC10699563 DOI: 10.1039/d3sc04976k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/27/2023] [Indexed: 05/02/2024] Open
Abstract
Reversible cysteine modification has been found to be a useful tool for a plethora of applications such as selective enzymatic inhibition, activity-based protein profiling and/or cargo release from a protein or a material. However, only a limited number of reagents display reliable dynamic/reversible thiol modification and, in most cases, many of these reagents suffer from issues of stability, a lack of modularity and/or poor rate tunability. In this work, we demonstrate the potential of pyridazinediones as novel reversible and tuneable covalent cysteine modifiers. We show that the electrophilicity of pyridazinediones correlates to the rates of the Michael addition and retro-Michael deconjugation reactions, demonstrating that pyridazinediones provide an enticing platform for readily tuneable and reversible thiol addition/release. We explore the regioselectivity of the novel reaction and unveil the reason for the fundamental increased reactivity of aryl bearing pyridazinediones by using DFT calculations and corroborating findings with SCXRD. We also applied this fundamental discovery to making more rapid disulfide rebridging agents in related work. We finally provide the groundwork for potential applications in various areas with exemplification using readily functionalised "clickable" pyridazinediones on clinically relevant cysteine and disulfide conjugated proteins, as well as on a hydrogel material.
Collapse
Affiliation(s)
- Léa N C Rochet
- 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
| | - Jonathan P Wojciechowski
- Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London London SW7 2AZ UK
| | - Ilias Koutsopetras
- Bio-Functional Chemistry (UMR 7199), Institut du Médicament de Strasbourg, University of Strasbourg 74 Route du Rhin 67400 Illkirch-Graffenstaden France
| | - Phyllida Britton
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Richard J Spears
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Ioanna A Thanasi
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Baihao Shao
- Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London London SW7 2AZ UK
| | - Lisha Zhong
- Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London London SW7 2AZ UK
| | - Dejan-Krešimir Bučar
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Abil E Aliev
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Michael J Porter
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London London SW7 2AZ UK
| | - James R Baker
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Vijay Chudasama
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| |
Collapse
|
7
|
Wang Z, Jing R, Li Y, Song D, Wan Y, Fukui N, Shinokubo H, Kuang Z, Xia A. Intrinsic Photostability in Dithiolonaphthalimide Achieved by Disulfide Bond-Induced Excited-State Quenching. J Phys Chem Lett 2023; 14:8485-8492. [PMID: 37721763 DOI: 10.1021/acs.jpclett.3c02260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Disulfide bridges common in proteins show excellent photostability achieved by ultrafast internal conversion and maintain the stability of the tertiary structure. When disulfide bonds exist in aromatic compounds, the rigid chemical structure may affect the cleavage and reforming dynamics of disulfide bonds. In this work, a model compound with a disulfide five-membered-ring structure, 4,5-dithiolo-N-(2,6-dimethylphenyl)-1,8-naphthalimide (DTDPNI), is selected to elaborate the effect of disulfide modification on the excited-state deactivation mechanism. Quantum chemical calculations show that the S-S stretching leads to a dramatic decrease in the energy gap between the S1 and S0 states, similar to the situation in 1,2-dithiane. Due to the efficient nonradiative process, the excited-state lifetime of DTDPNI resolved by ultrafast spectroscopy is determined to be ∼20 ps. It is found that the excellent photostability is achieved by ultrafast excited-state quenching induced by the S-S stretching, rather than the cleavage of the disulfide bond; even the disulfide bridge is in a very rigid aromatic molecular system.
Collapse
Affiliation(s)
- Zeming Wang
- State Key Laboratory of Information Photonic and Optical Communications, and School of Science, Beijing University of Posts and Telecommunications (BUPT), Beijing 100876, P. R. China
| | - Rui Jing
- State Key Laboratory of Information Photonic and Optical Communications, and School of Science, Beijing University of Posts and Telecommunications (BUPT), Beijing 100876, P. R. China
| | - Yang Li
- State Key Laboratory of Information Photonic and Optical Communications, and School of Science, Beijing University of Posts and Telecommunications (BUPT), Beijing 100876, P. R. China
| | - Di Song
- State Key Laboratory of Information Photonic and Optical Communications, and School of Science, Beijing University of Posts and Telecommunications (BUPT), Beijing 100876, P. R. China
| | - Yan Wan
- College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Norihito Fukui
- Department of Molecular and Macromolecular Chemistry, Graduate School of Engineering, Nagoya University, Furo-chi, Chikusa-ku, Nagoya 464-8603, Japan
| | - Hiroshi Shinokubo
- Department of Molecular and Macromolecular Chemistry, Graduate School of Engineering, Nagoya University, Furo-chi, Chikusa-ku, Nagoya 464-8603, Japan
| | - Zhuoran Kuang
- State Key Laboratory of Information Photonic and Optical Communications, and School of Science, Beijing University of Posts and Telecommunications (BUPT), Beijing 100876, P. R. China
| | - Andong Xia
- State Key Laboratory of Information Photonic and Optical Communications, and School of Science, Beijing University of Posts and Telecommunications (BUPT), Beijing 100876, P. R. China
| |
Collapse
|
8
|
Zhao Y, Chudasama V, Baker JR. Trifunctional Dibromomaleimide Reagents Built Around A Lysine Scaffold Deliver Site-selective Dual-modality Antibody Conjugation. Chembiochem 2023; 24:e202300356. [PMID: 37548625 DOI: 10.1002/cbic.202300356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/11/2023] [Indexed: 08/08/2023]
Abstract
We describe the synthesis and application of a selection of trifunctional reagents for the dual-modality modification of native, solvent accessible disulfide bonds in trastuzumab. The reagents were developed from the dibromomaleimide (DBM) platform with two orthogonal clickable functional groups built around a lysine core. We also describe the development of an aryl diselenide additive which enables antibody disulfide reduction in 4 minutes and a rapid overall reduction-bridging-double click sequence.
Collapse
Affiliation(s)
- Yanbo Zhao
- Department of Chemistry, University College London, 20 Gordon St, London, WC1H 0AJ, UK
| | - Vijay Chudasama
- Department of Chemistry, University College London, 20 Gordon St, London, WC1H 0AJ, UK
| | - James R Baker
- Department of Chemistry, University College London, 20 Gordon St, London, WC1H 0AJ, UK
| |
Collapse
|
9
|
Pinto M, Silva V, Barreiro S, Silva R, Remião F, Borges F, Fernandes C. Brain drug delivery and neurodegenerative diseases: Polymeric PLGA-based nanoparticles as a forefront platform. Ageing Res Rev 2022; 79:101658. [PMID: 35660114 DOI: 10.1016/j.arr.2022.101658] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/20/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023]
Abstract
The discovery of effective drugs for the treatment of neurodegenerative disorders (NDs) is a deadlock. Due to their complex etiology and high heterogeneity, progresses in the development of novel NDs therapies have been slow, raising social/economic and medical concerns. Nanotechnology and nanomedicine evolved exponentially in recent years and presented a panoply of tools projected to improve diagnosis and treatment. Drug-loaded nanosystems, particularly nanoparticles (NPs), were successfully used to address numerous drug glitches, such as efficacy, bioavailability and safety. Polymeric nanoparticles (PNPs), mainly based on polylactic-co-glycolic acid (PLGA), have been already validated and approved for the treatment of cancer, neurologic dysfunctions and hormonal-related diseases. Despite promising no PNPs-based therapy for neurodegenerative disorders is available up to date. To stimulate the research in the area the studies performed so far with polylactic-co-glycolic acid (PLGA) nanoparticles as well as the techniques aimed to improve PNPs BBB permeability and drug targeting were revised. Bearing in mind NDs pharmacological therapy landscape huge efforts must be done in finding new therapeutic solutions along with the translation of the most promising results to the clinic, which hopefully will converge in the development of effective drugs in a foreseeable future.
Collapse
|
10
|
Rose DA, Treacy JW, Yang ZJ, Ko JH, Houk KN, Maynard HD. Self-Immolative Hydroxybenzylamine Linkers for Traceless Protein Modification. J Am Chem Soc 2022; 144:6050-6058. [PMID: 35321547 DOI: 10.1021/jacs.2c01136] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Traceless self-immolative linkers are widely used for the reversible modification of proteins and peptides. This article describes a new class of traceless linkers based on ortho- or para-hydroxybenzylamines. The introduction of electron-donating substituents on the aromatic core stabilizes the quinone methide intermediate, thus providing a platform for payload release that can be modulated. To determine the extent to which the electronics affect the rate of release, we prepared a small library of hydroxybenzylamine linkers with varied electronics in the aromatic core, resulting in half-lives ranging from 20 to 144 h. Optimization of the linker design was carried out with mechanistic insights from density functional theory (DFT) and the in silico design of an intramolecular trapping agent through the use of DFT and intramolecular distortion energy calculations. This resulted in the development of a faster self-immolative linker with a half-life of 4.6 h. To demonstrate their effectiveness as traceless linkers for bioconjugation, reversible protein-polyethylene glycol conjugates with a model protein lysozyme were prepared, which had reduced protein activity but recovered ≥94% activity upon traceless release of the polymer. This new class of linkers with tunable release rates expands the traceless linkers toolbox for a variety of bioconjugation applications.
Collapse
Affiliation(s)
- Douglas A Rose
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Joseph W Treacy
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Zhongyue J Yang
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Jeong Hoon Ko
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - K N Houk
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| | - Heather D Maynard
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
| |
Collapse
|
11
|
Harms M, Hansson RF, Carmali S, Almeida-Hernández Y, Sanchez-Garcia E, Münch J, Zelikin AN. Dimerization of the Peptide CXCR4-Antagonist on Macromolecular and Supramolecular Protraction Arms Affords Increased Potency and Enhanced Plasma Stability. Bioconjug Chem 2022; 33:594-607. [PMID: 35293739 DOI: 10.1021/acs.bioconjchem.2c00034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Peptides are prime drug candidates due to their high specificity of action but are disadvantaged by low proteolytic stability. Here, we focus on the development of stabilized analogues of EPI-X4, an endogenous peptide antagonist of CXCR4. We synthesized macromolecular peptide conjugates and performed side-by-side comparison with their albumin-binding counterparts and considered monovalent conjugates, divalent telechelic conjugates, and Y-shaped peptide dimers. All constructs were tested for competition with the CXCR4 antibody-receptor engagement, inhibition of receptor activation, and inhibition of the CXCR4-tropic human immunodeficiency virus infection. We found that the Y-shaped conjugates were more potent than the parent peptide and at the same time more stable in human plasma, with a favorable outlook for translational studies.
Collapse
Affiliation(s)
- Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Rikke Fabech Hansson
- Department of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| | - Sheiliza Carmali
- Department of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| | - Yasser Almeida-Hernández
- Computational Biochemistry, Center of Medical Biotechnology, University Duisburg-Essen, D-45141 Essen, Germany
| | - Elsa Sanchez-Garcia
- Computational Biochemistry, Center of Medical Biotechnology, University Duisburg-Essen, D-45141 Essen, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Alexander N Zelikin
- Department of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| |
Collapse
|
12
|
Tagmose TM, Pedersen KM, Pridal L, Stidsen CE, Pedersen MØ, Lin Z, Zhang Y, Wan Z, Ferreras M, Naver H, Nielsen PK, Cao Z, Wang Y, Lykke L, Christensen JL, Jensen VS, Manfè V, Pedersen TÅ, Johansson E, Madsen P, Kodra JT, Münzel M, De Maria L, Nishimura E, Kjeldsen TB. Molecular Engineering of Efficacious Mono-Valent Ultra-Long Acting Two-Chain Insulin-Fc Conjugates. J Med Chem 2022; 65:2633-2645. [PMID: 35104142 DOI: 10.1021/acs.jmedchem.1c02039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Here, we describe molecular engineering of monovalent ultra-long acting two-chain insulin-Fc conjugates. Insulin-Fc conjugates were synthesized using trifunctional linkers with one amino reactive group for reaction with a lysine residue of insulin and two thiol reactive groups used for re-bridging of a disulfide bond within the Fc molecule. The ultra-long pharmacokinetic profile of the insulin-Fc conjugates was the result of concertedly slowing insulin receptor-mediated clearance by (1) introduction of amino acid substitutions that lowered the insulin receptor affinity and (2) conjugating insulin to the Fc element. Fc conjugation leads to recycling by the neonatal Fc receptor and increase in the molecular size, both contributing to the ultra-long pharmacokinetic and pharmacodynamic profiles.
Collapse
Affiliation(s)
- Tina M Tagmose
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | | | - Lone Pridal
- Novo Nordisk A/S, Global Drug Discovery, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Carsten E Stidsen
- Novo Nordisk A/S, Global Drug Discovery, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Marie Ø Pedersen
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Zhaosheng Lin
- Novo Nordisk A/S, Global Research Technologies, 20 Life Science Park Road, Changping District, 102206 Beijing, China
| | - Yuanyuan Zhang
- Novo Nordisk A/S, Global Research Technologies, 20 Life Science Park Road, Changping District, 102206 Beijing, China
| | - Zhe Wan
- Novo Nordisk A/S, Global Research Technologies, 20 Life Science Park Road, Changping District, 102206 Beijing, China
| | - Mercedes Ferreras
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Helle Naver
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Peter K Nielsen
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Zheng Cao
- Novo Nordisk A/S, Global Research Technologies, 20 Life Science Park Road, Changping District, 102206 Beijing, China
| | - Yi Wang
- Novo Nordisk A/S, Global Research Technologies, 20 Life Science Park Road, Changping District, 102206 Beijing, China
| | - Lennart Lykke
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | | | - Victoria S Jensen
- Novo Nordisk A/S, Global Drug Discovery, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Valentina Manfè
- Novo Nordisk A/S, Global Drug Discovery, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Thomas Å Pedersen
- Novo Nordisk A/S, Global Drug Discovery, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Eva Johansson
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Peter Madsen
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - János T Kodra
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Martin Münzel
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Leonardo De Maria
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Erica Nishimura
- Novo Nordisk A/S, Global Drug Discovery, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| | - Thomas B Kjeldsen
- Novo Nordisk A/S, Global Research Technologies, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| |
Collapse
|
13
|
Cheng L, Niu MM, Yan T, Ma Z, Huang K, Yang L, Zhong X, Li C. Bioresponsive micro-to-nano albumin-based systems for targeted drug delivery against complex fungal infections. Acta Pharm Sin B 2021; 11:3220-3230. [PMID: 34729311 PMCID: PMC8546853 DOI: 10.1016/j.apsb.2021.04.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/09/2021] [Accepted: 03/25/2021] [Indexed: 12/14/2022] Open
Abstract
As a typical human pathogenic fungus, Cryptococcus neoformans is a life-threatening invasive fungal pathogen with a worldwide distribution causing ∼700,000 deaths annually. Cryptococcosis is not just an infection with multi-organ involvement, intracellular survival and extracellular multiplication of the fungus also play important roles in the pathogenesis of C. neoformans infections. Because adequate accumulation of drugs at target organs and cells is still difficult to achieve, an effective delivery strategy is desperately required to treat these infections. Here, we report a bioresponsive micro-to-nano (MTN) system that effectively clears the C. neoformans in vivo. This strategy is based on our in-depth study of the overexpression of matrix metalloproteinase 3 (MMP-3) in infectious microenvironments (IMEs) and secreted protein acidic and rich in cysteine (SPARC) in several associated target cells. In this MTN system, bovine serum albumin (BSA, a natural ligand of SPARC) was used for the preparation of nanoparticles (NPs), and then microspheres were constructed by conjugation with a special linker, which mainly consisted of a BSA-binding peptide and an MMP-3-responsive peptide. This MTN system was mechanically captured by the smallest capillaries of the lungs after intravenous injection, and then hydrolyzed into BSA NPs by MMP-3 in the IMEs. The NPs further targeted the lung tissue, brain and infected macrophages based on the overexpression of SPARC, reaching multiple targets and achieving efficient treatment. We have developed a size-tunable strategy where microspheres "shrink" to NPs in IMEs, which effectively combines active and passive targeting and may be especially powerful in the fight against complex fungal infections.
Collapse
Key Words
- Albumin
- AmB, amphotericin B
- BBB, blood‒brain barrier
- BSA, bovine serum albumin
- Complex fungal infection
- DDS, drug delivery system
- IME, infectious microenvironment
- MMP-3
- MMP-3, matrix metalloproteinase 3
- MTN, micro-to-nano
- Microenvironment responsive
- NP, nanoparticle
- PEG, polyethylene glycol
- PMVECs, pulmonary microvascular endothelial cells
- RFP, red fluorescent protein
- SPARC
- SPARC, secreted protein acidic and rich in cysteine
- Size-tunable strategy
Collapse
Affiliation(s)
- Liting Cheng
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Miao-Miao Niu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing 210009, China
| | - Tong Yan
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhongyi Ma
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Kexin Huang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Ling Yang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xin Zhong
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Chong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
14
|
Herr K, Fleckenstein M, Brodrecht M, Höfler MV, Heise H, Aussenac F, Gutmann T, Reggelin M, Buntkowsky G. A novel strategy for site selective spin-labeling to investigate bioactive entities by DNP and EPR spectroscopy. Sci Rep 2021; 11:13714. [PMID: 34211027 PMCID: PMC8249612 DOI: 10.1038/s41598-021-92975-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/18/2021] [Indexed: 11/09/2022] Open
Abstract
A novel specific spin-labeling strategy for bioactive molecules is presented for eptifibatide (integrilin) an antiplatelet aggregation inhibitor, which derives from the venom of certain rattlesnakes. By specifically labeling the disulfide bridge this molecule becomes accessible for analytical techniques such as Electron Paramagnetic Resonance (EPR) and solid state Dynamic Nuclear Polarization (DNP). The necessary spin-label was synthesized and inserted into the disulfide bridge of eptifibatide via reductive followed by insertion by a double Michael addition under physiological conditions. This procedure is universally applicable for disulfide containing biomolecules and is expected to preserve their tertiary structure with minimal change due to the small size of the label and restoring of the previous disulfide connection. HPLC and MS analysis show the successful introduction of the spin label and EPR spectroscopy confirms its activity. DNP-enhanced solid state NMR experiments show signal enhancement factors of up to 19 in 13C CP MAS experiments which corresponds to time saving factors of up to 361. This clearly shows the high potential of our new spin labeling strategy for the introduction of site selective radical spin labels into biomolecules and biosolids without compromising its conformational integrity for structural investigations employing solid-state DNP or advanced EPR techniques.
Collapse
Affiliation(s)
- Kevin Herr
- Institute of Physical Chemistry, Technical University Darmstadt, Alarich-Weiss-Straße 8, 64287, Darmstadt, Germany
| | - Max Fleckenstein
- Institute of Organic Chemistry, Technical University Darmstadt, Alarich-Weiss-Straße 4, 64287, Darmstadt, Germany
| | - Martin Brodrecht
- Institute of Physical Chemistry, Technical University Darmstadt, Alarich-Weiss-Straße 8, 64287, Darmstadt, Germany
| | - Mark V Höfler
- Institute of Physical Chemistry, Technical University Darmstadt, Alarich-Weiss-Straße 8, 64287, Darmstadt, Germany
| | - Henrike Heise
- Structural Biochemistry (ICS-6), Institute of Complex Systems, Forschungszentrum Jülich, 52425, Jülich, Germany.,Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany
| | - Fabien Aussenac
- Bruker France SAS, 34 rue de l'industrie, 67160, Wissembourg, France
| | - Torsten Gutmann
- Institute of Physical Chemistry, Technical University Darmstadt, Alarich-Weiss-Straße 8, 64287, Darmstadt, Germany
| | - Michael Reggelin
- Institute of Organic Chemistry, Technical University Darmstadt, Alarich-Weiss-Straße 4, 64287, Darmstadt, Germany.
| | - Gerd Buntkowsky
- Institute of Physical Chemistry, Technical University Darmstadt, Alarich-Weiss-Straße 8, 64287, Darmstadt, Germany.
| |
Collapse
|
15
|
Bahou C, Szijj PA, Spears RJ, Wall A, Javaid F, Sattikar A, Love EA, Baker JR, Chudasama V. A Plug-and-Play Platform for the Formation of Trifunctional Cysteine Bioconjugates that also Offers Control over Thiol Cleavability. Bioconjug Chem 2021; 32:672-679. [PMID: 33710874 PMCID: PMC8154211 DOI: 10.1021/acs.bioconjchem.1c00057] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/04/2021] [Indexed: 02/08/2023]
Abstract
Linkers that enable the site-selective synthesis of chemically modified proteins are of great interest to the field of chemical biology. Homogenous bioconjugates often show advantageous pharmacokinetic profiles and consequently increased efficacy in vivo. Cysteine residues have been exploited as a route to site-selectively modify proteins, and many successfully approved therapeutics make use of cysteine directed conjugation reagents. However, commonly used linkers, including maleimide-thiol conjugates, are not stable to the low concentrations of thiol present in blood. Furthermore, only a few cysteine-targeting reagents enable the site-selective attachment of multiple functionalities: a useful tool in the fields of theranostics and therapeutic blood half-life extension. Herein, we demonstrate the application of the pyridazinedione motif to enable site-selective attachment of three functionalities to a protein bearing a single cysteine residue. Extending upon previously documented dual modification work, here we demonstrate that by exploiting a bromide leaving group as an additional reactive point on the pyridazinedione scaffold, a thiol or aniline derivative can be added to a protein, post-conjugation. Thiol cleavability appraisal of the resultant C-S and C-N linked thio-bioconjugates demonstrated C-S functionalized linkers to be cleavable and C-N functionalized linkers to be noncleavable when incubated in an excess of glutathione. The plug-and-play trifunctional platform was exemplified by attaching clinically relevant motifs: biotin, fluorescein, a polyethylene glycol chain, and a model peptide. This platform provides a rare opportunity to combine up to three functionalities on a protein in a site-selective fashion. Furthermore, by selecting the use of a thiol or an amine for functionalization, we provide unique control over linker cleavability toward thiols, allowing this novel linker to be applied in a range of physiological environments.
Collapse
Affiliation(s)
- Calise Bahou
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Peter A. Szijj
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Richard J. Spears
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Archie Wall
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Faiza Javaid
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Afrah Sattikar
- LifeArc,
Accelerator Building, SBC Open Innovation Campus, SG1 2FX, Stevenage, United Kingdom
| | - Elizabeth A. Love
- LifeArc,
Accelerator Building, SBC Open Innovation Campus, SG1 2FX, Stevenage, United Kingdom
| | - James R. Baker
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
| | - Vijay Chudasama
- Department
of Chemistry, University College London, 20 Gordon Street, WC1H OAJ, London, United Kingdom
- Research
Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| |
Collapse
|
16
|
Kuan SL, Raabe M. Solid-Phase Protein Modifications: Towards Precision Protein Hybrids for Biological Applications. ChemMedChem 2021; 16:94-104. [PMID: 32667697 PMCID: PMC7818443 DOI: 10.1002/cmdc.202000412] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Indexed: 12/13/2022]
Abstract
Proteins have attracted increasing attention as biopharmaceutics and diagnostics due to their high specificity, biocompatibility, and biodegradability. The biopharmaceutical sector in particular is experiencing rapid growth, which has led to an increase in the production and sale of protein drugs and diagnostics over the last two decades. Since the first-generation biopharmaceutics dominated by native proteins, both recombinant and chemical technologies have evolved and transformed the outlook of this rapidly developing field. This review article presents updates on the fabrication of covalent and supramolecular fusion hybrids, as well as protein-polymer hybrids using solid-phase approaches that hold great promise for preparing protein hybrids with precise control at the macromolecular level to incorporate additional features. In addition, the applications of the resultant protein hybrids in medicine and diagnostics are highlighted where possible.
Collapse
Affiliation(s)
- Seah Ling Kuan
- Max Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Institute of Inorganic Chemistry IUlm UniversityAlbert-Einstein-Allee 1189081UlmGermany
| | - Marco Raabe
- Max Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Institute of Inorganic Chemistry IUlm UniversityAlbert-Einstein-Allee 1189081UlmGermany
| |
Collapse
|
17
|
Yang Z, Li P, Chen Y, Gan Q, Feng Z, Jin Y, Zhou C, He Z, Wang C, Liu Y, Feng C. Construction of pH/glutathione responsive chitosan nanoparticles by a self-assembly/self-crosslinking method for photodynamic therapy. Int J Biol Macromol 2020; 167:46-58. [PMID: 33271181 DOI: 10.1016/j.ijbiomac.2020.11.141] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/12/2020] [Accepted: 11/20/2020] [Indexed: 01/04/2023]
Abstract
A novel pH/glutathione (GSH) multi-responsive chitosan nanoparticles (NPs) material has been successfully designed and prepared by a self-assembly/self-crosslinking method for photodynamic therapy (PDT), which overcomes the shortcomings of traditional photosensitizer carriers, such as poor chemical stability, low loading efficiency and single-responsive photosensitizer release. Amphiphilic sulfhydryl chitosan (SA-CS-NAC) is first prepared by modifying chitosan (CS) with stearic acid (SA) and N-acetyl-L-cysteine (NAC), and then subject to self-assembly and self-crosslinking in the presence of photosensitizer, indocyanine green (ICG), to form the ICG-loaded amphiphilic sulfhydryl chitosan nanoparticles (SA-CS-NAC@ICG NPs). The ICG entrapment efficiency and loading efficiency of the NPs are found to be 95.2% and 27.6%, respectively. The multi-responsive ICG release of the NPs to the low pH and high GSH content of the microenvironment in tumor cells is successfully achieved. Under the laser irradiation, the SA-CS-NAC@ICG NPs produce the amount of reactive oxygen species (ROS) twice of that generated by free ICG under the same conditions. The in vitro cell experiment confirmed the strong cellular uptake ability, low biotoxicity and good tumor inhibition of the NPs. Our work has provided a new strategy for the targeted photosensitizer delivery for PDT.
Collapse
Affiliation(s)
- Ziming Yang
- Beijing Institute of Technology, Beijing 100081, PR China; South Subtropical Crop Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong 524091, PR China
| | - Puwang Li
- South Subtropical Crop Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong 524091, PR China
| | - Yu Chen
- Beijing Institute of Technology, Beijing 100081, PR China.
| | - Qiang Gan
- Beijing Institute of Technology, Beijing 100081, PR China
| | - Zhipan Feng
- Beijing Institute of Technology, Beijing 100081, PR China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | - Chuang Zhou
- South Subtropical Crop Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong 524091, PR China
| | - Zuyu He
- South Subtropical Crop Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong 524091, PR China
| | - Chao Wang
- South Subtropical Crop Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong 524091, PR China
| | - Yunhao Liu
- South Subtropical Crop Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang, Guangdong 524091, PR China
| | - Changgen Feng
- Beijing Institute of Technology, Beijing 100081, PR China.
| |
Collapse
|
18
|
Wu LH, Zhou S, Luo QF, Tian JS, Loh TP. Dichloroacetophenone Derivatives: A Class of Bioconjugation Reagents for Disulfide Bridging. Org Lett 2020; 22:8193-8197. [PMID: 33052688 DOI: 10.1021/acs.orglett.0c02477] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A mild and biocompatible method for the construction of disulfide bridging in peptides using dichloroacetophenone derivatives is developed. This method is highly selective (chemo, diastereo, regio, etc.) and atom economic and works under biocompatible reaction conditions (metal-free, water, pH 7, rt, etc.).
Collapse
Affiliation(s)
- Liu-Hai Wu
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| | - Shuguang Zhou
- Institute of Advanced Synthesis (IAS), Northwestern Polytechnical University (NPU), Xi'an 710072, China.,Yangtze River Delta Research Institute of NPU, Taicang, Jiangsu 215400, China
| | - Qun-Feng Luo
- Institute of Advanced Synthesis (IAS), Northwestern Polytechnical University (NPU), Xi'an 710072, China.,Yangtze River Delta Research Institute of NPU, Taicang, Jiangsu 215400, China
| | - Jie-Sheng Tian
- Institute of Advanced Synthesis (IAS), Northwestern Polytechnical University (NPU), Xi'an 710072, China.,Yangtze River Delta Research Institute of NPU, Taicang, Jiangsu 215400, China.,Institute of Advanced Synthesis (IAS), Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Teck-Peng Loh
- Institute of Advanced Synthesis (IAS), Northwestern Polytechnical University (NPU), Xi'an 710072, China.,Yangtze River Delta Research Institute of NPU, Taicang, Jiangsu 215400, China.,Institute of Advanced Synthesis (IAS), Nanjing Tech University (NanjingTech), Nanjing 211816, China.,Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| |
Collapse
|
19
|
|
20
|
Jiang F, Liu K, Zhao M, Tao X, Hu X, Lu S. Tunable High-Molecular-Weight Silk Fibroin Polypeptide Materials: Fabrication and Self-Assembly Mechanism. ACS APPLIED BIO MATERIALS 2020; 3:3248-3259. [PMID: 35025367 DOI: 10.1021/acsabm.0c00231] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Silk fibroin is a multisegment natural protein composed of a heavy (H) chain, a light (L) chain and a P25 glycoprotein chain. Herein, we developed a dialysis separation technique under reducing conditions to break the disulfide bond between the H-chain and L-chain and remove the low-molecular-weight portions of the protein. Thus, a high-molecular-weight silk fibroin polypeptide (HSF) material was obtained. SDS-PAGE electrophoresis showed that the molecular weight of HSF was over 80 kDa, similar to the size of the silk fibroin H-chain. Amino acid analysis result demonstrated that the amino acid composition of HSF was almost identical to that of H-chain composition. Importantly, the HSF material obtained has a high surface activity, which can reduce the surface tension of water to below 20 mN/m; at high temperature and high concentration, it can also form a unique nanofibrous network with a lamellar crystalline structure. HSF can further form a rod-shaped structure in a strong polar environment and become a star-shaped fibrous network in a weak polar environment. When the pH value of HSF solution was adjusted from 6 to 8, a structural transition from a folded crank sheet-like structure with micellar beads to a ring-like fibrous structure was observed. During the conversion of HSF from colloidal particles to nanofibers, its molecular conformation also transformed from random coils to β-sheets. These tunable properties indicate that HSF materials have a wide range of applications in biomedical and green chemistry fields.
Collapse
Affiliation(s)
- Fujian Jiang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Kai Liu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Meihui Zhao
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Xiaocheng Tao
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Xiao Hu
- Department of Physics and Astronomy, Department of Biomedical Engineering, Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, New Jersey 08028, United States
| | - Shenzhou Lu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| |
Collapse
|
21
|
Draper SRE, Ashton DS, Conover BM, Carter AJ, Stern KL, Xiao Q, Price JL. PEGylation near a Patch of Nonpolar Surface Residues Increases the Conformational Stability of the WW Domain. J Org Chem 2020; 85:1725-1730. [PMID: 31749365 DOI: 10.1021/acs.joc.9b02615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Many proteins have one or more surface-exposed patches of nonpolar residues; our observations here suggest that PEGylation near such locations might be a useful strategy for increasing protein conformational stability. Specifically, we show that conjugating a PEG-azide to a propargyloxyphenylalanine via the copper(I)-catalyzed azide-alkyne cycloaddition can increase the conformational stability of the WW domain due to a favorable synergistic effect that depends on the hydrophobicity of a nearby patch of nonpolar surface residues.
Collapse
Affiliation(s)
- Steven R E Draper
- Department of Chemistry and Biochemistry , Brigham Young University , Provo , Utah 84602 , United States
| | - Dallin S Ashton
- Department of Chemistry and Biochemistry , Brigham Young University , Provo , Utah 84602 , United States
| | - Benjamin M Conover
- Department of Chemistry and Biochemistry , Brigham Young University , Provo , Utah 84602 , United States
| | - Anthony J Carter
- Department of Chemistry and Biochemistry , Brigham Young University , Provo , Utah 84602 , United States
| | - Kimberlee L Stern
- Department of Chemistry and Biochemistry , Brigham Young University , Provo , Utah 84602 , United States
| | - Qiang Xiao
- Department of Chemistry and Biochemistry , Brigham Young University , Provo , Utah 84602 , United States
| | - Joshua L Price
- Department of Chemistry and Biochemistry , Brigham Young University , Provo , Utah 84602 , United States
| |
Collapse
|
22
|
Arisawa M, Fukumoto K, Yamaguchi M. Rhodium-catalyzed phosphorylation reaction of water-soluble disulfides using hypodiphosphoric acid tetraalkyl esters in water. RSC Adv 2020; 10:13820-13823. [PMID: 35492965 PMCID: PMC9051538 DOI: 10.1039/d0ra02377a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022] Open
Abstract
RhCl3catalyzed the phosphorylation reaction of water-soluble disulfides, including unprotected glutathione disulfide, with hypodiphosphoric acid tetraalkyl esters in homogeneous water.
Collapse
Affiliation(s)
- Mieko Arisawa
- Department of Organic Chemistry
- Graduate School of Pharmaceutical Sciences
- Tohoku University
- Sendai
- Japan
| | - Kohei Fukumoto
- Department of Organic Chemistry
- Graduate School of Pharmaceutical Sciences
- Tohoku University
- Sendai
- Japan
| | - Masahiko Yamaguchi
- Department of Organic Chemistry
- Graduate School of Pharmaceutical Sciences
- Tohoku University
- Sendai
- Japan
| |
Collapse
|
23
|
Nielsen T, Märcher A, Drobňáková Z, Hučko M, Štengl M, Balšánek V, Wiberg C, Nielsen PF, Nielsen TE, Gothelf KV, Cló E. Disulphide-mediated site-directed modification of proteins. Org Biomol Chem 2020; 18:4717-4722. [DOI: 10.1039/d0ob00861c] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Site-directed addition of a single thiols handle to proteins by means of temporary disulphide rebridging of solvent exposed disulphides is obtained with a new labelling reagent.
Collapse
Affiliation(s)
- Thorbjørn Nielsen
- Interdisciplinary Nanoscience Center
- and the Dept. of Chemistry
- Aarhus University
- 8000 Aarhus C
- Denmark
| | - Anders Märcher
- Interdisciplinary Nanoscience Center
- and the Dept. of Chemistry
- Aarhus University
- 8000 Aarhus C
- Denmark
| | | | | | | | | | | | | | | | - Kurt V. Gothelf
- Interdisciplinary Nanoscience Center
- and the Dept. of Chemistry
- Aarhus University
- 8000 Aarhus C
- Denmark
| | | |
Collapse
|
24
|
|
25
|
Duarte Y, Márquez-Miranda V, Miossec MJ, González-Nilo F. Integration of target discovery, drug discovery and drug delivery: A review on computational strategies. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1554. [PMID: 30932351 DOI: 10.1002/wnan.1554] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/14/2018] [Accepted: 01/23/2019] [Indexed: 12/22/2022]
Abstract
Most of the computational tools involved in drug discovery developed during the 1980s were largely based on computational chemistry, quantitative structure-activity relationship (QSAR) and cheminformatics. Subsequently, the advent of genomics in the 2000s gave rise to a huge number of databases and computational tools developed to analyze large quantities of data, through bioinformatics, to obtain valuable information about the genomic regulation of different organisms. Target identification and validation is a long process during which evidence for and against a target is accumulated in the pursuit of developing new drugs. Finally, the drug delivery system appears as a novel approach to improve drug targeting and releasing into the cells, leading to new opportunities to improve drug efficiency and avoid potential secondary effects. In each area: target discovery, drug discovery and drug delivery, different computational strategies are being developed to accelerate the process of selection and discovery of new tools to be applied to different scientific fields. Research on these three topics is growing rapidly, but still requires a global view of this landscape to detect the most challenging bottleneck and how computational tools could be integrated in each topic. This review describes the current state of the art in computational strategies for target discovery, drug discovery and drug delivery and how these fields could be integrated. Finally, we will discuss about the current needs in these fields and how the continuous development of databases and computational tools will impact on the improvement of those areas. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Yorley Duarte
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Valeria Márquez-Miranda
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Matthieu J Miossec
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Fernando González-Nilo
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile.,Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
26
|
Richter F, Zettlitz KA, Seifert O, Herrmann A, Scheurich P, Pfizenmaier K, Kontermann RE. Monovalent TNF receptor 1-selective antibody with improved affinity and neutralizing activity. MAbs 2019; 11:166-177. [PMID: 30252601 PMCID: PMC6343807 DOI: 10.1080/19420862.2018.1524664] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/29/2018] [Accepted: 09/10/2018] [Indexed: 01/08/2023] Open
Abstract
Selective inhibition of tumor necrosis factor (TNF) signaling through the proinflammatory axis of TNF-receptor 1 (TNFR1) while leaving pro-survival and regeneration-promoting signals via TNFR2 unaffected is a promising strategy to circumvent limitations of complete inhibition of TNF action by the approved anti-TNF drugs. A previously developed humanized antagonistic TNFR1-specific antibody, ATROSAB, showed potent inhibition of TNFR1-mediated cellular responses. Because the parental mouse antibody H398 possesses even stronger inhibitory potential, we scrutinized the specific binding parameters of the two molecules and revealed a faster dissociation of ATROSAB compared to H398. Applying affinity maturation and re-engineering of humanized variable domains, we generated a monovalent Fab derivative (13.7) of ATROSAB that exhibited increased binding to TNFR1 and superior inhibition of TNF-mediated TNFR1 activation, while lacking any agonistic activity even in the presence of cross-linking antibodies. In order to improve its pharmacokinetic properties, several Fab13.7-derived molecules were generated, including a PEGylated Fab, a mouse serum albumin fusion protein, a half-IgG with a dimerization-deficient Fc, and a newly designed Fv-Fc format, employing the knobs-into-holes technology. Among these derivatives, the Fv13.7-Fc displayed the best combination of improved pharmacokinetic properties and antagonistic activity, thus representing a promising candidate for further clinical development.
Collapse
Affiliation(s)
- Fabian Richter
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Kirstin A. Zettlitz
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Oliver Seifert
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | | | - Peter Scheurich
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Klaus Pfizenmaier
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
27
|
Forte N, Chudasama V, Baker JR. Homogeneous antibody-drug conjugates via site-selective disulfide bridging. DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 30:11-20. [PMID: 30553515 DOI: 10.1016/j.ddtec.2018.09.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/19/2018] [Indexed: 06/09/2023]
Abstract
Antibody-drug conjugates (ADCs) constructed using site-selective labelling methodologies are likely to dominate the next generation of these targeted therapeutics. To this end, disulfide bridging has emerged as a leading strategy as it allows the production of highly homogeneous ADCs without the need for antibody engineering. It consists of targeting reduced interchain disulfide bonds with reagents which reconnect the resultant pairs of cysteine residues, whilst simultaneously attaching drugs. The 3 main reagent classes which have been exemplified for the construction of ADCs by disulfide bridging will be discussed in this review; bissulfones, next generation maleimides and pyridazinediones, along with others in development.
Collapse
Affiliation(s)
- Nafsika Forte
- Department of Chemistry, University College London, London, UK
| | - Vijay Chudasama
- Department of Chemistry, University College London, London, UK.
| | - James R Baker
- Department of Chemistry, University College London, London, UK.
| |
Collapse
|
28
|
Englert C, Brendel JC, Majdanski TC, Yildirim T, Schubert S, Gottschaldt M, Windhab N, Schubert US. Pharmapolymers in the 21st century: Synthetic polymers in drug delivery applications. Prog Polym Sci 2018. [DOI: 10.1016/j.progpolymsci.2018.07.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
29
|
Morais M, Ma MT. Site-specific chelator-antibody conjugation for PET and SPECT imaging with radiometals. DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 30:91-104. [PMID: 30553525 PMCID: PMC6291455 DOI: 10.1016/j.ddtec.2018.10.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 11/17/2022]
Abstract
Antibodies and their derivatives radiolabelled with positron- and gamma-emitting radiometals enable sensitive and quantitative molecular Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT) imaging of antibody distribution in vivo. Chelators that are covalently attached to antibodies allow radiolabelling with metallic PET and SPECT radioisotopes. Conventional strategies for chelator-protein conjugation generate heterogeneous mixtures of bioconjugates that can exhibit reduced affinity for their receptor targets, and undesirable biodistribution and pharmacokinetics. Recent advances in bioconjugation technology enable site-specific modification to generate well-defined constructs with superior properties. Herein we survey existing site-specific chelator-protein conjugation methods. These include chelator attachment to cysteines/disulfide bonds or the glycan region of the antibody, enzyme-mediated chelator conjugation, and incorporation of sequences of amino acids that chelate the radiometal. Such technology will allow better use of PET and SPECT imaging in the development of antibody-based therapies.
Collapse
Affiliation(s)
- Mauricio Morais
- School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom.
| | - Michelle T Ma
- School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| |
Collapse
|
30
|
Ramirez-Paz J, Saxena M, Delinois LJ, Joaquín-Ovalle FM, Lin S, Chen Z, Rojas-Nieves VA, Griebenow K. Thiol-maleimide poly(ethylene glycol) crosslinking of L-asparaginase subunits at recombinant cysteine residues introduced by mutagenesis. PLoS One 2018; 13:e0197643. [PMID: 30052638 PMCID: PMC6063399 DOI: 10.1371/journal.pone.0197643] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022] Open
Abstract
L-Asparaginase is an enzyme successfully being used in the treatment of acute lymphoblastic leukemia, acute myeloid leukemia, and non-Hodgkin’s lymphoma. However, some disadvantages still limit its full application potential, e.g., allergic reactions, pancreatitis, and blood clotting impairment. Therefore, much effort has been directed at improving its performance. A popular strategy is to randomly conjugate L-asparaginase with mono-methoxy polyethylene glycol, which became a commercial FDA approved formulation widely used in recent years. To improve this formulation by PEGylation, herein we performed cysteine-directed conjugation of the L-asparaginase subunits to prevent dissociation-induced loss of activity. The recombinant cysteine conjugation sites were introduced by mutagenesis at surface-exposed positions on the protein to avoid affecting the catalytic activity. Three conjugates were obtained using different linear PEGs of 1000, 2000, and 5000 g/mol, with physical properties ranging from a semi-solid gel to a fully soluble state. The soluble-conjugate exhibited higher catalytic activity than the non-conjugated mutant, and the same activity than the native enzyme. The cysteine-directed crosslinking of the L-asparaginase subunits produced a higher molecular weight conjugate compared to the native tetrameric enzyme. This strategy might improve L-asparaginase efficiency for leukemia treatment by reducing glomerular filtration due to the increase in hydrodynamic size thus extending half-live, while at the same time retaining full catalytic activity.
Collapse
Affiliation(s)
- Josell Ramirez-Paz
- Department of Chemistry, Faculty of Natural Sciences, University of Puerto Rico at Rio Piedras, San Juan, Puerto Rico, United States of America
| | - Manoj Saxena
- Department of Chemistry, Faculty of Natural Sciences, University of Puerto Rico at Rio Piedras, San Juan, Puerto Rico, United States of America
| | - Louis J. Delinois
- Department of Chemistry, Faculty of Natural Sciences, University of Puerto Rico at Rio Piedras, San Juan, Puerto Rico, United States of America
| | - Freisa M. Joaquín-Ovalle
- Department of Chemistry, Faculty of Natural Sciences, University of Puerto Rico at Rio Piedras, San Juan, Puerto Rico, United States of America
| | - Shiru Lin
- Department of Chemistry, Faculty of Natural Sciences, University of Puerto Rico at Rio Piedras, San Juan, Puerto Rico, United States of America
| | - Zhongfang Chen
- Department of Chemistry, Faculty of Natural Sciences, University of Puerto Rico at Rio Piedras, San Juan, Puerto Rico, United States of America
| | - Virginia A. Rojas-Nieves
- Department of Chemistry, Faculty of Natural Sciences, University of Puerto Rico at Rio Piedras, San Juan, Puerto Rico, United States of America
| | - Kai Griebenow
- Department of Chemistry, Faculty of Natural Sciences, University of Puerto Rico at Rio Piedras, San Juan, Puerto Rico, United States of America
- * E-mail:
| |
Collapse
|
31
|
Shao S, Tsai MH, Lu J, Yu T, Jin J, Xiao D, Jiang H, Han M, Wang M, Wang J. Site-specific and hydrophilic ADCs through disulfide-bridged linker and branched PEG. Bioorg Med Chem Lett 2018; 28:1363-1370. [DOI: 10.1016/j.bmcl.2018.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 02/28/2018] [Accepted: 03/02/2018] [Indexed: 01/19/2023]
|
32
|
Gu Z, Gao D, Al-Zubaydi F, Li S, Singh Y, Rivera K, Holloway J, Szekely Z, Love S, Sinko PJ. The effect of size and polymer architecture of doxorubicin-poly(ethylene) glycol conjugate nanocarriers on breast duct retention, potency and toxicity. Eur J Pharm Sci 2018; 121:118-125. [PMID: 29698706 DOI: 10.1016/j.ejps.2018.04.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 04/22/2018] [Indexed: 11/17/2022]
Abstract
Although systemic administration of chemotherapeutic agents is routinely used for treating invasive breast cancer, the only therapeutic options for ductal carcinoma in situ (DCIS) are surgery and radiation. Treating DCIS by delivering drugs locally to the affected milk duct offers significant advantages over systemic administration, including reduced systemic and breast toxicities, as well as a greatly reduced need for surgery and radiation. In this study, mammary gland retention and toxicity of intraductally administered poly(ethylene) glycol-doxorubicin (PEG-DOX) polymeric conjugate nanocarriers of varying molecular sizes and architectures were investigated. Nanocarriers were formed by conjugating one or more copies of doxorubicin to PEG polymers, of varying molecular weights (5, 10, 20, and 40 kDa) and architectures (linear, four-arm and eight-arm). Cytotoxicity against MCF7 cells, a human breast cancer cell line, was assessed, and IC50 values were calculated. The nanocarriers were intraductally administered into the mammary glands of female retired breeder Sprague-Dawley rats. Whole body images were captured using in vivo optical imaging, and changes in ductal structure as well local inflammation were monitored. Fluorescence intensities were monitored, over time, to evaluate nanocarrier mammary gland retention half-lives (t1/2). The IC50 values of PEG-DOX nanocarriers against MCF7 cells were 40 kDa PEG-(DOX)4 (1.23 μM) < 5 kDa PEG-DOX (1.76 μM) < 40 kDa PEG-(DOX)8 (3.49 μM) < 10 kDa PEG-DOX (3.86 μM) < 20 kDa PEG-DOX (8.96 μM) < 40 kDa PEG-DOX (18.11 μM), whereas the IC50 of free DOX was only 0.14 μM. The t1/2 of linear 5, 20, and 40 kDa nanocarriers were 2.2 ± 0.3, 3.6 ± 0.6, and 13.1 ± 3.4 h, whereas the retention t1/2 of 4- and 8-arm 40 kDa nanocarriers were 14.9 ± 5.6 h and 11.9 ± 2.9 h, respectively. The retention t1/2 of free doxorubicin was 2.0 ± 0.4 h, which was significantly shorter than that of the linear and branched 40 kDa PEG-DOX nanocarriers. Increased molecular weight and decreased branching both demonstrated a strong correlation to enhanced mammary gland retention. Intraductally administered free doxorubicin resulted in ductal damage, severe inflammation and generation of atypical cell neoplasms, whereas PEG-DOX nanocarriers induced only minor and transient inflammation (i.e., damaged epithelial cells and detached cellular debris). The 40 kDa 4-arm PEG-DOX nanocarrier demonstrated the longest ductal retention half-life, the lowest IC50 (i.e., most potent), and minimal ductal damage and inflammation. The current results suggest that PEG-DOX nanocarriers with prolonged ductal retention may present the best option for intraductal treatment of DCIS, due to their low local toxicity and potential for sustained therapeutic effect.
Collapse
Affiliation(s)
- Zichao Gu
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| | - Dayuan Gao
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| | - Firas Al-Zubaydi
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| | - Shike Li
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| | - Yashveer Singh
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA; Department of Chemistry, Center for Biomedical Engineering (CBME), Indian Institute of Technology Ropar, Nangal Road, Rupnagar, Punjab 140001, India.
| | - Kristia Rivera
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| | - Jennifer Holloway
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| | - Zoltan Szekely
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA; Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08901, USA.
| | - Susan Love
- Dr. Susan Love Research Foundation, 16133 Ventura Suite 1000, Encino, CA 91436, USA.
| | - Patrick J Sinko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA; Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08901, USA.
| |
Collapse
|
33
|
Wilding KM, Smith AK, Wilkerson JW, Bush DB, Knotts TA, Bundy BC. The Locational Impact of Site-Specific PEGylation: Streamlined Screening with Cell-Free Protein Expression and Coarse-Grain Simulation. ACS Synth Biol 2018; 7:510-521. [PMID: 29295615 DOI: 10.1021/acssynbio.7b00316] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Although polyethylene glycol (PEG) is commonly used to improve protein stability and therapeutic efficacy, the optimal location for attaching PEG onto proteins is not well understood. Here, we present a cell-free protein synthesis-based screening platform that facilitates site-specific PEGylation and efficient evaluation of PEG attachment efficiency, thermal stability, and activity for different variants of PEGylated T4 lysozyme, including a di-PEGylated variant. We also report developing a computationally efficient coarse-grain simulation model as a potential tool to narrow experimental screening candidates. We use this simulation method as a novel tool to evaluate the locational impact of PEGylation. Using this screen, we also evaluated the predictive impact of PEGylation site solvent accessibility, conjugation site structure, PEG size, and double PEGylation. Our findings indicate that PEGylation efficiency, protein stability, and protein activity varied considerably with PEGylation site, variations that were not well predicted by common PEGylation guidelines. Overall our results suggest current guidelines are insufficiently predictive, highlighting the need for experimental and simulation screening systems such as the one presented here.
Collapse
Affiliation(s)
- Kristen M. Wilding
- Department of Chemical Engineering, Brigham Young University, Provo, Utah 84602, United States
| | - Addison K. Smith
- Department of Chemical Engineering, Brigham Young University, Provo, Utah 84602, United States
| | - Joshua W. Wilkerson
- Department of Chemical Engineering, Brigham Young University, Provo, Utah 84602, United States
| | - Derek B. Bush
- Department of Chemical Engineering, Brigham Young University, Provo, Utah 84602, United States
| | - Thomas A. Knotts
- Department of Chemical Engineering, Brigham Young University, Provo, Utah 84602, United States
| | - Bradley C. Bundy
- Department of Chemical Engineering, Brigham Young University, Provo, Utah 84602, United States
| |
Collapse
|
34
|
Maso K, Grigoletto A, Pasut G. Transglutaminase and Sialyltransferase Enzymatic Approaches for Polymer Conjugation to Proteins. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 112:123-142. [PMID: 29680235 DOI: 10.1016/bs.apcsb.2018.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Proteins hold a central role in medicine and biology, also confirmed by the several therapeutic applications based on biologic drugs. Such therapies are of great relevance thanks to high potency and safety of proteins. Nevertheless, many proteins as therapeutics might present issues like fast kidney clearance, rapid enzymatic degradation, or immunogenicity. Such defects implicate frequent administrations or administrations at high doses of the therapeutics, thus yielding or exacerbating potential side effects. A successful technology for improving the clinical profiles of proteins is the conjugation of polymers to the protein surface. The design of a protein-polymer conjugate presents critical aspects that determine the efficacy and safety of the final product. The control over stoichiometry and conjugation site is a strict criterion on which researchers have been intensively focused during the years, in order to obtain homogeneous and batch-to-batch reproducible products. An innovative site-specific conjugation strategy relies on the use of enzymes as tools to mediate polymer conjugation. Enzymatic approaches are attractive because they allow site-selective polymer conjugation at specific protein amino acids. In these reactions, the polymer is a substrate analog that replaces the native substrate. Furthermore, enzymes can count other advantages such as high yields of conversion and physiological conditions of reaction. This chapter provides a meaningful description of protein-polymer conjugation through transglutaminase-mediated and sialyltransferase-mediated enzymatic strategies, reporting the mechanism of action and some relevant examples.
Collapse
Affiliation(s)
| | | | - Gianfranco Pasut
- University of Padua, Padua, Italy; Veneto Institute of Oncology IOV-IRCCS, Padua, Italy.
| |
Collapse
|
35
|
|
36
|
Agrawalla BK, Wang T, Riegger A, Domogalla MP, Steinbrink K, Dörfler T, Chen X, Boldt F, Lamla M, Michaelis J, Kuan SL, Weil T. Chemoselective Dual Labeling of Native and Recombinant Proteins. Bioconjug Chem 2017; 29:29-34. [PMID: 29231709 PMCID: PMC6242188 DOI: 10.1021/acs.bioconjchem.7b00675] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The attachment of two different functionalities in a site-selective fashion represents a great challenge in protein chemistry. We report site specific dual functionalizations of peptides and proteins capitalizing on reactivity differences of cysteines in their free (thiol) and protected, oxidized (disulfide) forms. The dual functionalization of interleukin 2 and EYFP proceeded with no loss of bioactivity in a stepwise fashion applying maleimide and disulfide rebridging allyl-sulfone groups. In order to ensure broader applicability of the functionalization strategy, a novel, short peptide sequence that introduces a disulfide bridge was designed and site-selective dual labeling in the presence of biogenic groups was successfully demonstrated.
Collapse
Affiliation(s)
| | - Tao Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University , Chengdu 610031, P.R. China
| | - Andreas Riegger
- Max-Planck-Institute for Polymer Research , Ackermannweg 10, 55128 Mainz, Germany
| | - Matthias P Domogalla
- Max-Planck-Institute for Polymer Research , Ackermannweg 10, 55128 Mainz, Germany.,Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg-University Mainz , Mainz D-55099, Germany
| | - Kerstin Steinbrink
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg-University Mainz , Mainz D-55099, Germany
| | | | | | | | | | | | - Seah Ling Kuan
- Max-Planck-Institute for Polymer Research , Ackermannweg 10, 55128 Mainz, Germany
| | - Tanja Weil
- Max-Planck-Institute for Polymer Research , Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
37
|
Awwad S, Al-Shohani A, Khaw PT, Brocchini S. Comparative Study of In Situ Loaded Antibody and PEG-Fab NIPAAM Gels. Macromol Biosci 2017; 18. [PMID: 29205853 DOI: 10.1002/mabi.201700255] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/18/2017] [Indexed: 01/01/2023]
Abstract
Hydrogels can potentially prolong the release of a therapeutic protein, especially to treat blinding conditions. One challenge is to ensure that the protein and hydrogel are intimately mixed by better protein entanglement within the hydrogel. N-isopropylacrylamide (NIPAAM) gels are optimized with poly(ethylene glycol) diacrylate (PEDGA) crosslinker in the presence of either bevacizumab or PEG conjugated ranibizumab (PEG10 -Fabrani ). The release profiles of the hydrogels are evaluated using an outflow model of the eye, which is previously validated for human clearance of proteins. Release kinetics of in situ loaded bevacizumab-NIPAAM gels displays a prolonged bimodal release profile in phosphate buffered saline compared to bevacizumab loaded into a preformed NIPAAM gel. Bevacizumab release in simulated vitreous from in situ loaded gels is similar to bevacizumab control indicating that diffusion through the vitreous rather than from the gel is rate limiting. Ranibizumab is site-specifically PEGylated by disulfide rebridging conjugation. Prolonged and continuous release is observed with the in situ loaded PEG10 -Fabrani -NIPAAM gels compared to PEG10 -Fabrani injection (control). Compared to an unmodified protein, there is better mixing due to PEG entanglement and compatibility of PEG10 -Fabrani within the NIPAAM-PEDGA hydrogel. These encouraging results suggest that the extended release of PEGylated proteins in the vitreous can be achieved using injectable hydrogels.
Collapse
Affiliation(s)
- Sahar Awwad
- UCL School of Pharmacy, London, WC1N 1AX, UK.,National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, EC1V 9EL, UK
| | - Athmar Al-Shohani
- UCL School of Pharmacy, London, WC1N 1AX, UK.,National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, EC1V 9EL, UK
| | - Peng T Khaw
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, EC1V 9EL, UK
| | - Steve Brocchini
- UCL School of Pharmacy, London, WC1N 1AX, UK.,National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, EC1V 9EL, UK
| |
Collapse
|
38
|
Raghupathi K, Eron SJ, Anson F, Hardy JA, Thayumanavan S. Utilizing Inverse Emulsion Polymerization To Generate Responsive Nanogels for Cytosolic Protein Delivery. Mol Pharm 2017; 14:4515-4524. [PMID: 29053277 PMCID: PMC5714657 DOI: 10.1021/acs.molpharmaceut.7b00643] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Therapeutic biologics have various advantages over synthetic drugs in terms of selectivity, their catalytic nature, and, thus, therapeutic efficacy. These properties offer the potential for more effective treatments that may also overcome the undesirable side effects observed due to off-target toxicities of small molecule drugs. Unfortunately, systemic administration of biologics is challenging due to cellular penetration, renal clearance, and enzymatic degradation difficulties. A delivery vehicle that can overcome these challenges and deliver biologics to specific cellular populations has the potential for significant therapeutic impact. In this work, we describe a redox-responsive nanoparticle platform, which can encapsulate hydrophilic proteins and release them only in the presence of a reducing stimulus. We have formulated these nanoparticles using an inverse emulsion polymerization (IEP) methodology, yielding inverse nanoemulsions, or nanogels. We have demonstrated our ability to overcome the liabilities that contribute to activity loss by delivering a highly challenging cargo, functionally active caspase-3, a cysteine protease susceptible to oxidative and self-proteolytic insults, to the cytosol of HeLa cells by encapsulation inside a redox-responsive nanogel.
Collapse
Affiliation(s)
| | - Scott J. Eron
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003
| | - Francesca Anson
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003
| | - Jeanne A. Hardy
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003
- Center for Bioactive Delivery at the Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003
| | - S. Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003
- Center for Bioactive Delivery at the Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003
| |
Collapse
|
39
|
Li Z, Huang R, Xu H, Chen J, Zhan Y, Zhou X, Chen H, Jiang B. Divinylsulfonamides as Specific Linkers for Stapling Disulfide Bonds in Peptides. Org Lett 2017; 19:4972-4975. [PMID: 28880566 DOI: 10.1021/acs.orglett.7b02464] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A new class of N-phenyl-divinylsulfonamides which can be easily prepared have been successfully developed and utilized as efficient linkers in the field of disulfide bond modification. Functional divinylsulfonamides provide opportunities for the specific introduction of various functionalities, including affinity probes, fluorescent tags, and drugs, into peptides.
Collapse
Affiliation(s)
- Zhihong Li
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University , Shanghai 201210, China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203, China.,University of Chinese Academy of Sciences , Beijing 100049, China
| | - Rong Huang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University , Shanghai 201210, China
| | - Hongtao Xu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University , Shanghai 201210, China.,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203, China
| | - Jiakang Chen
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University , Shanghai 201210, China
| | - Yuexiong Zhan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University , Shanghai 201210, China
| | - Xianhao Zhou
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University , Shanghai 201210, China
| | - Hongli Chen
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University , Shanghai 201210, China
| | - Biao Jiang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University , Shanghai 201210, China
| |
Collapse
|
40
|
Herrington-Symes A, Choi JW, Brocchini S. Interferon dimers: IFN-PEG-IFN. J Drug Target 2017; 25:881-890. [PMID: 28817988 DOI: 10.1080/1061186x.2017.1363214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Increasingly complex proteins can be made by a recombinant chemical approach where proteins that can be made easily can be combined by site-specific chemical conjugation to form multifunctional or more active protein therapeutics. Protein dimers may display increased avidity for cell surface receptors. The increased size of protein dimers may also increase circulation times. Cytokines bind to cell surface receptors that dimerise, so much of the solvent accessible surface of a cytokine is involved in binding to its target. Interferon (IFN) homo-dimers (IFN-PEG-IFN) were prepared by two methods: site-specific bis-alkylation conjugation of PEG to the two thiols of a native disulphide or to two imidazoles on a histidine tag of two His8-tagged IFN (His8IFN). Several control conjugates were also prepared to assess the relative activity of these IFN homo-dimers. The His8IFN-PEG20-His8IFN obtained by histidine-specific conjugation displayed marginally greater in vitro antiviral activity compared to the IFN-PEG20-IFN homo-dimer obtained by disulphide re-bridging conjugation. This result is consistent with previous observations in which enhanced retention of activity was made possible by conjugation to an N-terminal His-tag on the IFN. Comparison of the antiviral and antiproliferative activities of the two IFN homo-dimers prepared by disulphide re-bridging conjugation indicated that IFN-PEG10-IFN was more biologically active than IFN-PEG20-IFN. This result suggests that the size of PEG may influence the antiviral activity of IFN-PEG-IFN homo-dimers.
Collapse
Affiliation(s)
| | - Ji-Won Choi
- a Abzena , Babraham Research Campus , Babraham, Cambridge , UK
| | | |
Collapse
|
41
|
Zhao J, Lin JD, Chen JC, Chen G, Li XL, Wang XQ, Chen MX. α-chymotrypsin activated and stabilized by self-assembled polypseudorotaxane fabricated with bis-thiolated poly(ethylene glycol) and α-cyclodextrin: Spectroscopic and mechanistic analysis. Int J Biol Macromol 2017; 102:1266-1273. [PMID: 28495630 DOI: 10.1016/j.ijbiomac.2017.05.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 05/04/2017] [Accepted: 05/05/2017] [Indexed: 10/19/2022]
Abstract
The self-assembled polypseudorotaxane (PPRX) fabricated with bis-thiolated poly(ethylene glycol) (PEG) and α-cyclodextrin (α-CyD) acted as an activator for α-chymotrypsin (CT) and retained the activity of CT for a long time up to 7days. The stabilization mechanism was studied, and the interaction between CT and PPRX was analyzed by using circular dichroism, fluorescence spectra and X-ray powder diffraction (XRD). The bis-thiolated PEG and its assembled PPRX with α-CyD exhibited the interaction with the C-terminal region of the CT's B-chain probably through PEGylation of the surface disulfide bridge of CT. It caused the aromatic chromophores more exposed to the hydrophilic microenvironment, leading to conformational variation of CT that was revealed by spectroscopic analysis. It rendered the peptide chains in a more flexible and active state. As a comparison, the non-thiolated components could not decorate the surface of CT and performed almost no effect on its stability, which demonstrated that the decoration of the surface disulfide bridge was a key factor in retaining the activity of CT. Due to the activation and stabilization effect, bis-thiolated PEG/α-CyD PPRX was an excellent soft-immobilized carrier for CT, and provided an intriguing method for enzyme's stabilization.
Collapse
Affiliation(s)
- Jun Zhao
- Department of Bioengineering and Biotechnology, College of Chemical Engineering, Huaqiao University, 668 Jimei Avenue, Amoy, 361021, China; Key Laboratory of Chemical Biology and Molecular Engineering (Huaqiao University), Fujian Province University, 668 Jimei Avenue, Amoy, 361021, China; Institute of Oil and Natural Products, Huaqiao University, 668 Jimei Avenue, Amoy, 361021, China.
| | - Ji-Duan Lin
- Department of Bioengineering and Biotechnology, College of Chemical Engineering, Huaqiao University, 668 Jimei Avenue, Amoy, 361021, China; Key Laboratory of Chemical Biology and Molecular Engineering (Huaqiao University), Fujian Province University, 668 Jimei Avenue, Amoy, 361021, China
| | - Jia-Chen Chen
- Department of Bioengineering and Biotechnology, College of Chemical Engineering, Huaqiao University, 668 Jimei Avenue, Amoy, 361021, China
| | - Guo Chen
- Department of Bioengineering and Biotechnology, College of Chemical Engineering, Huaqiao University, 668 Jimei Avenue, Amoy, 361021, China; Key Laboratory of Chemical Biology and Molecular Engineering (Huaqiao University), Fujian Province University, 668 Jimei Avenue, Amoy, 361021, China
| | - Xia-Lan Li
- Department of Bioengineering and Biotechnology, College of Chemical Engineering, Huaqiao University, 668 Jimei Avenue, Amoy, 361021, China; Key Laboratory of Chemical Biology and Molecular Engineering (Huaqiao University), Fujian Province University, 668 Jimei Avenue, Amoy, 361021, China
| | - Xiao-Qin Wang
- Department of Bioengineering and Biotechnology, College of Chemical Engineering, Huaqiao University, 668 Jimei Avenue, Amoy, 361021, China; Key Laboratory of Chemical Biology and Molecular Engineering (Huaqiao University), Fujian Province University, 668 Jimei Avenue, Amoy, 361021, China; Institute of Oil and Natural Products, Huaqiao University, 668 Jimei Avenue, Amoy, 361021, China
| | - Ming-Xia Chen
- Department of Bioengineering and Biotechnology, College of Chemical Engineering, Huaqiao University, 668 Jimei Avenue, Amoy, 361021, China; Key Laboratory of Chemical Biology and Molecular Engineering (Huaqiao University), Fujian Province University, 668 Jimei Avenue, Amoy, 361021, China
| |
Collapse
|
42
|
Makwana H, Mastrotto F, Magnusson JP, Sleep D, Hay J, Nicholls KJ, Allen S, Alexander C. Engineered Polymer–Transferrin Conjugates as Self-Assembling Targeted Drug Delivery Systems. Biomacromolecules 2017; 18:1532-1543. [DOI: 10.1021/acs.biomac.7b00101] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Hiteshri Makwana
- School
of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | - Francesca Mastrotto
- School
of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | - Johannes P. Magnusson
- School
of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | - Darrell Sleep
- Albumedix, Ltd., 59 Castle Boulevard, Nottingham NG7 1FD, United Kingdom
| | - Joanna Hay
- Albumedix, Ltd., 59 Castle Boulevard, Nottingham NG7 1FD, United Kingdom
| | - Karl J Nicholls
- Albumedix, Ltd., 59 Castle Boulevard, Nottingham NG7 1FD, United Kingdom
| | - Stephanie Allen
- School
of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | - Cameron Alexander
- School
of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| |
Collapse
|
43
|
Abstract
The formulation in which therapeutic proteins are administered plays a key role in retaining their biological activity. Enzyme wrapping, using synthetic polymers, is a strategy employed to provide enzymes with lower immunogenicity, longer circulation times, and better targeting capabilities. Protein-polymer complexation methods, involving covalent, noncovalent, and electrostatic interactions, that can provide means to develop formulations for retaining enzyme stability are discussed in this chapter. Amphiphilic self-cross-linkable polymer was used to encapsulate capsase-3 enzyme in the nanogel, while inverse emulsion polymerization method was used to entrap α-glucosidase enzyme in the nanogel. These nanogels were characterized by dynamic light scattering, transmission electron microscopy, and gel electrophoresis. Upon release of caspase-3 enzyme from polymeric nanogel, it retained nearly 86% of its original activity. Similarly, α-glucosidase that was encased in the acid cleavable polymeric nanogel exhibited substantial activity after release under acidic conditions (pH 5, 48h). Nano-armoring of the enzymes were nearly complete and provided high yields of the encased enzyme.
Collapse
|
44
|
Abstract
SIGNIFICANCE Secreted proteins are important both as signaling molecules and potential biomarkers. Recent Advances: Protein can undergo different types of oxidation, both in physiological conditions or under oxidative stress. Several redox proteomics techniques have been successfully applied to the identification of glutathionylated proteins, an oxidative post-translational modification consisting in the formation of a mixed disulfide between a protein cysteine and glutathione. Redox proteomics has also been used to study other forms of protein oxidation. CRITICAL ISSUES Because of the highest proportion of free cysteines in the cytosol, redox proteomics of protein thiols has focused, so far, on intracellular proteins. However, plasma proteins, such as transthyretin and albumin, have been described as glutathionylated or cysteinylated. The present review discusses the redox state of protein cysteines in relation to their cellular distribution. We describe the various approaches used to detect secreted glutathionylated proteins, the only thiol modification studied so far in secreted proteins, and the specific problems presented in the study of the secretome. FUTURE DIRECTIONS This review focusses on glutathionylated proteins secreted under inflammatory conditions and that may act as soluble mediators (cytokines). Future studies on the redox secretome (including other forms of oxidation) might identify new soluble mediators and biomarkers of oxidative stress. Antioxid. Redox Signal. 26, 299-312.
Collapse
Affiliation(s)
- Pietro Ghezzi
- 1 Brighton & Sussex Medical School , Brighton, United Kingdom
| | - Philippe Chan
- 2 PISSARO Proteomic Platform, Institute for Research and Innovation in Biomedicine, University of Rouen , Rouen, France
| |
Collapse
|
45
|
Carmali S, Murata H, Cummings C, Matyjaszewski K, Russell AJ. Polymer-Based Protein Engineering: Synthesis and Characterization of Armored, High Graft Density Polymer-Protein Conjugates. Methods Enzymol 2017; 590:347-380. [PMID: 28411645 DOI: 10.1016/bs.mie.2016.12.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Atom transfer radical polymerization (ATRP) from the surface of a protein can generate remarkably dense polymer shells that serve as armor and rationally tune protein function. Using straightforward chemistry, it is possible to covalently couple or display multiple small molecule initiators onto a protein surface. The chemistry is fine-tuned to be sequence specific (if one desires a single targeted site) at controlled density. Once the initiator is anchored on the protein surface, ATRP is used to grow polymers on protein surface, in situ. The technique is so powerful that a single-protein polymer conjugate molecule can contain more than 90% polymer coating by weight. If desired, stimuli-responsive polymers can be "grown" from the initiated sites to prepare enzyme conjugates that respond to external triggers such as temperature or pH, while still maintaining enzyme activity and stability. Herein, we focus mainly on the synthesis of chymotrypsin-polymer conjugates. Control of the number of covalently coupled initiator sites by changing the stoichiometric ratio between enzyme and the initiator during the synthesis of protein-initiator complexes allowed fine-tuning of the grafting density. For example, very high grafting density chymotrypsin conjugates were prepared from protein-initiator complexes to grow the temperature-responsive polymers, poly(N-isopropylacrylamide), and poly[N,N'-dimethyl(methacryloyloxyethyl) ammonium propane sulfonate]. Controlled growth of polymers from protein surfaces enables one to predictably manipulate enzyme kinetics and stability without the need for molecular biology-dependent mutagenesis.
Collapse
Affiliation(s)
- Sheiliza Carmali
- Center for Polymer-Based Protein Engineering, ICES, Carnegie Mellon University, Pittsburgh, PA, United States; Carnegie Mellon University, Pittsburgh, PA, United States
| | - Hironobu Murata
- Center for Polymer-Based Protein Engineering, ICES, Carnegie Mellon University, Pittsburgh, PA, United States; Carnegie Mellon University, Pittsburgh, PA, United States
| | - Chad Cummings
- Center for Polymer-Based Protein Engineering, ICES, Carnegie Mellon University, Pittsburgh, PA, United States; Carnegie Mellon University, Pittsburgh, PA, United States
| | - Krzysztof Matyjaszewski
- Center for Polymer-Based Protein Engineering, ICES, Carnegie Mellon University, Pittsburgh, PA, United States; Carnegie Mellon University, Pittsburgh, PA, United States
| | - Alan J Russell
- Center for Polymer-Based Protein Engineering, ICES, Carnegie Mellon University, Pittsburgh, PA, United States; Carnegie Mellon University, Pittsburgh, PA, United States.
| |
Collapse
|
46
|
Kuan SL, Wang T, Weil T. Site-Selective Disulfide Modification of Proteins: Expanding Diversity beyond the Proteome. Chemistry 2016; 22:17112-17129. [PMID: 27778400 PMCID: PMC5600100 DOI: 10.1002/chem.201602298] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Indexed: 01/06/2023]
Abstract
The synthetic transformation of polypeptides with molecular accuracy holds great promise for providing functional and structural diversity beyond the proteome. Consequently, the last decade has seen an exponential growth of site-directed chemistry to install additional features into peptides and proteins even inside living cells. The disulfide rebridging strategy has emerged as a powerful tool for site-selective modifications since most proteins contain disulfide bonds. In this Review, we present the chemical design, advantages and limitations of the disulfide rebridging reagents, while summarizing their relevance for synthetic customization of functional protein bioconjugates, as well as the resultant impact and advancement for biomedical applications.
Collapse
Affiliation(s)
- Seah Ling Kuan
- Institute of Organic Chemistry IIIUlm UniversityAlbert-Einstein-Allee 1189081UlmGermany
- Max Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Tao Wang
- Institute of Organic Chemistry IIIUlm UniversityAlbert-Einstein-Allee 1189081UlmGermany
- School of Materials Science and EngineeringSouthwest Jiaotong UniversityChengdu610031P.R. China
| | - Tanja Weil
- Institute of Organic Chemistry IIIUlm UniversityAlbert-Einstein-Allee 1189081UlmGermany
- Max Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| |
Collapse
|
47
|
Kantner T, Watts AG. Characterization of Reactions between Water-Soluble Trialkylphosphines and Thiol Alkylating Reagents: Implications for Protein-Conjugation Reactions. Bioconjug Chem 2016; 27:2400-2406. [PMID: 27602944 DOI: 10.1021/acs.bioconjchem.6b00375] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Water-soluble trialkylphosphines such as tris(carboxyethyl)phosphine (TCEP) and trishydroxypropyl phosphine (THPP) are effective agents for reducing disulfide bonds in proteins and are increasingly becoming the reagents of choice for bioconjugation strategies that modify cysteine (thiol containing) amino acids. These reducing agents are often considered as being chemically compatible with Michael acceptors such as maleimides and, as such, are often not removed prior to performing protein conjugation reactions. Here, we demonstrate the rapid and irreversible reaction of both TCEP and THPP with derivatives of the commonly employed thiol alkylating groups, maleimide and vinyl sulfone. Mechanistic investigations revealed distinct differences between the reactions of TCEP and THPP with maleimide, leading to the production of either nonproductive ylenes or succidimidyl derivatives, respectively. Importantly, we also demonstrate the incorporation of nonproductive ylenes formed between maleimide and TCEP into the Pneumococcal capsular polysaccharide Pn6b following strategies employed toward the production of conjugate vaccines.
Collapse
Affiliation(s)
- Terrence Kantner
- Department of Pharmacy and Pharmacology, University of Bath , Claverton Down, Bath BA2 7AY, United Kingdom
| | - Andrew G Watts
- Department of Pharmacy and Pharmacology, University of Bath , Claverton Down, Bath BA2 7AY, United Kingdom
| |
Collapse
|
48
|
Nanda P, JagadeeshBabu PE, Gupta P, Prasad AG. Development of a Spectrophotometric Biphasic Assay for the Estimation of mPEG-maleimide in Thiol PEGylation Reaction Mixtures. CHEM ENG COMMUN 2016. [DOI: 10.1080/00986445.2016.1204604] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Pooja Nanda
- Department of Chemical Engineering, National Institute of Technology Karnataka, Surathkal, Karnataka, India
| | - P. E. JagadeeshBabu
- Department of Chemical Engineering, National Institute of Technology Karnataka, Surathkal, Karnataka, India
| | - Purva Gupta
- Department of Chemical Engineering, National Institute of Technology Karnataka, Surathkal, Karnataka, India
| | - Aprameya Ganesh Prasad
- Department of Chemical Engineering, National Institute of Technology Karnataka, Surathkal, Karnataka, India
| |
Collapse
|
49
|
Aviv O, Amir N, Laout N, Ratner S, Basu A, Domb AJ. Poly(hexamethylene guanidine)-poly(ethylene glycol) solid blend for water microbial deactivation. Polym Degrad Stab 2016. [DOI: 10.1016/j.polymdegradstab.2016.04.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
50
|
Ginn C, Choi JW, Brocchini S. Disulfide-bridging PEGylation during refolding for the more efficient production of modified proteins. Biotechnol J 2016; 11:1088-99. [DOI: 10.1002/biot.201600035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 03/07/2016] [Accepted: 05/17/2016] [Indexed: 01/18/2023]
Affiliation(s)
| | - Ji-won Choi
- PolyTherics Ltd; Babraham Research Campus, Babraham; Cambridge UK
| | | |
Collapse
|