1
|
Fawcett C, Watson J, Richards S, Doherty AE, Seki H, Love EA, Coles CH, Coe DM, Jamieson C. Comparative Study of Click Handle Stability in Common Ligation Conditions. Bioconjug Chem 2025. [PMID: 40287825 DOI: 10.1021/acs.bioconjchem.5c00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
Click chemistry efficiently ligates molecular building blocks in a robust and high-yielding manner and has found major application in the rapid modification of important molecular actors in biological systems. However, the high reactivity of click handles often correlates with decreased stability, which presents a significant challenge in the practical application of these systems. In the current study, we describe a survey of the stability of commonly deployed click manifolds across a range of widely used ligation conditions. Incompatible click handle and ligation condition combinations are identified, with kinetic half-lives and side products of each undesired reaction determined, including the assessment of stability over extended periods and in a protein environment. This data set provides researchers with a roadmap to expediently determine the most appropriate click reaction conditions for any given bioorthogonal application, thus elevating the probability of success of procedures that utilize click chemistry.
Collapse
Affiliation(s)
- Caitlin Fawcett
- Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Joe Watson
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Stephen Richards
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Alfred E Doherty
- Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Hikaru Seki
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Elizabeth A Love
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Charlotte H Coles
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Diane M Coe
- Research Technologies, GSK, Gunnels Wood Road, Stevenage SG1 2NY, United Kingdom
| | - Craig Jamieson
- Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom
| |
Collapse
|
2
|
Ragab SS. Signature of click chemistry in advanced techniques for cancer therapeutics. RSC Adv 2025; 15:10583-10601. [PMID: 40190630 PMCID: PMC11970365 DOI: 10.1039/d5ra01196e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Click chemistry has made a revolution in the field of chemical biology owing to its high efficiency, specificity, and mild reaction conditions. The copper(i)-catalyzed azide-alkyne cycloaddition (CuAAC) and strain-promoted [3 + 2] azide-alkyne cycloaddition (SPAAC) stand out as the most popular click reactions that construct a stable triazole ring by reacting an azide with an alkyne. These two reactions represent an ideal choice for biological applications due to its specificity, reliability, and biocompatibility. As a powerful modular synthetic approach for creating new molecular entities, it has seen increasing use in anticancer drug discovery. The present "state of the art" focuses mainly on the signature of click chemistry (CuAAC and SPAAC) in advanced techniques for cancer therapeutics, which includes cancer immunotherapy, antibody-drug conjugates, development of proteolysis-targeting chimeras, targeted dual-agent combination therapy for cancer, exosome modification for cancer therapy, and photodynamic therapy (PDT).
Collapse
Affiliation(s)
- Sherif Shaban Ragab
- Photochemistry Department, Chemical Industries Research Institute, National Research Centre El-Buhouth St, P.O. 12622, Dokki Giza Egypt
| |
Collapse
|
3
|
Le Stum M, Romero E, Molander GA. Photocatalyzed elaboration of antibody-based bioconjugates. Beilstein J Org Chem 2025; 21:616-629. [PMID: 40130177 PMCID: PMC11931643 DOI: 10.3762/bjoc.21.49] [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: 12/21/2024] [Accepted: 03/05/2025] [Indexed: 03/26/2025] Open
Abstract
Antibody-drug conjugates (ADCs) represent a promising class of targeted therapeutics, combining the specificity of antibodies with the potency of cytotoxic drugs to enhance therapeutic efficacy while minimizing off-target effects. The development of new chemical methods for bioconjugation is essential to generate ADCs and to optimize their stability, efficacy, and safety. Traditional conjugation methods often face challenges related to site-selectivity and heterogeneous product mixtures, highlighting the need to develop new, innovative chemical strategies. Photoredox chemistry emerges as a powerful tool in this context, enabling precise, mild, and selective modifications of peptides and proteins. By harnessing light to drive chemical transformations, photoredox techniques can facilitate the synthesis of antibody bioconjugates. This perspective will discuss the drive to develop and empower photoredox methods applied to antibody functionalization.
Collapse
Affiliation(s)
- Marine Le Stum
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-Sur-Yvette, France
| | - Eugénie Romero
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-Sur-Yvette, France
| | - Gary A Molander
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-Sur-Yvette, France
| |
Collapse
|
4
|
Sayed MT, Mady MF. A review of click chemistry in the synthesis of organophosphorus triazoles and their biological activities. Eur J Med Chem 2025; 286:117270. [PMID: 39827489 DOI: 10.1016/j.ejmech.2025.117270] [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: 11/23/2024] [Revised: 12/20/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
Organophosphorus compounds, characterized by the incorporation of phosphorus into organic molecules, play a critical role in various fields such as medicine, agriculture, and industry. Their unique electronic properties and versatility make them essential in developing therapeutic agents, pesticides, and materials. One prominent class of organophosphorus compounds is organophosphorus heterocycles, which combine the benefits of both phosphorus and cyclic structures. Triazoles, a class of nitrogen-containing heterocyclic compounds, are particularly notable for their broad biological activities, including anticancer, antiviral, antibacterial, and antioxidant effects. Traditional methods for synthesizing triazoles often encounter challenges such as low yields and non-selective products, whereas click chemistry provides a more efficient and reliable alternative. The copper-catalyzed azide-alkyne [3 + 2] cycloaddition, a cornerstone of click chemistry, allows for the rapid and selective formation of triazoles under mild conditions. When functionalized with organophosphorus groups, triazoles not only retain but often enhance their biological activities, improving their potency, selectivity, and stability. This review covers the synthesis of organophosphorus-functionalized triazoles via click chemistry and explores their molecular structure, including the coordination chemistry of these compounds. The behavior and interactions of these organophosphorus derivatives with various metal ions are also addressed, as these interactions significantly influence their chemical reactivity, stability, and bioactivity.
Collapse
Affiliation(s)
- Mariam T Sayed
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha, 2713, Qatar
| | - Mohamed F Mady
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha, 2713, Qatar.
| |
Collapse
|
5
|
Zhang J, Yang Z, Liu Y, Liu Y, Qu J, Pan X. Recent Advances in Smart Linkage Strategies for Developing Drug Conjugates for Targeted Delivery. Top Curr Chem (Cham) 2025; 383:13. [PMID: 40080285 DOI: 10.1007/s41061-025-00497-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: 06/11/2024] [Accepted: 02/16/2025] [Indexed: 03/15/2025]
Abstract
Targeted drug delivery systems effectively solve the problem of off-target toxicity of chemotherapeutic drugs by combining chemotherapeutic drugs with antibodies or peptides, thereby promoting drug targeting to the tumor site and bringing further hope for cancer treatment. The development of stimulus-responsive smart linkage technologies has led to the emergence of drug conjugates. Linkage technologies play a crucial role in the design, synthesis, and in vivo circulation of drug conjugates, as they determine the release of cytotoxic drugs from the conjugates and their subsequent therapeutic efficacy. This article reviews some of the smart linkage strategies used in designing drug conjugates, with a focus on the tumor microenvironment and exogenous stimuli as conditions influencing controlled drug release. This review introduces linker classifications and cleavage mechanisms, discusses modular linkers that promote the efficient synthesis of conjugates, and discusses the differences between linkage strategies. Furthermore, this article focuses on the implementation of self-assembly in drug conjugates, which is currently of great interest. Related concepts are introduced and relevant examples of their applications are provided. Furthermore, a comprehensive discourse is presented on the challenges that may arise in the research and clinical implementation of diverse linkage strategies, along with the associated enhancement measures. Finally, the factors that should be considered when designing linkage strategies for drug conjugates are summarized, offering strategies and ideas for scientists involved in drug conjugate research. It is particularly noteworthy that appropriate linkage strategies allow for the intracellular release of drugs after internalization of the conjugates, thereby maximizing their tumor cell-killing effect.
Collapse
Affiliation(s)
- Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zeyu Yang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yu Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yuying Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jingkun Qu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyan Pan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
6
|
Segujja F, Duruksu G, Eren EB, İsayeva A, Yazır Y, Erdem A. Diels-Alder-based IPN hydrogels with tunable mechanical and protein release properties for tissue engineering. Int J Biol Macromol 2025; 306:141779. [PMID: 40049464 DOI: 10.1016/j.ijbiomac.2025.141779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/19/2025] [Accepted: 03/04/2025] [Indexed: 04/12/2025]
Abstract
Advancing hydrogel technology with tunable mechanical strength and sustained release is critical for therapeutic applications in drug delivery and tissue engineering. Conventional single polymer networks, including semi-interpenetrating polymer network (SIPN) hydrogels, often lack mechanical robustness and controlled release needed for therapeutic use. In this study, we fabricated a biocompatible interpenetrating polymer network (IPN) hydrogel with improved properties for controlled protein release. We employed a facile one-pot synthesis approach that integrated aqueous Diels-Alder (DA) 'click' chemistry with photopolymerization methods to crosslink gelatin methacryloyl (GelMA) within a polymeric framework of poly(ethylene) glycol bismaleimide (PEGMI) and multi-furan-modified polyethylene glycol (PEGFU). Spectroscopy (FTIR and 1H NMR) confirmed the chemical composition of the hydrogels. The effect of varying polymer ratios on hydrogel properties was assessed to optimize protein release and mechanical behavior. Fully crosslinked IPN hydrogels exhibited enhanced energy dissipation and compressive moduli 2.5- to 3.5-fold relative to SIPN hydrogels across various polymer ratios. Release kinetics followed the Korsmeyer-Peppas mathematical model, indicating sustained release. IPN hydrogels demonstrated good water absorption, moderate degradation, and favorable biocompatibility with 3 T3 fibroblast cells. Overall, these findings highlight the potential of IPN hydrogels as a promising drug delivery platform for advancing regenerative therapies and targeted treatment strategies.
Collapse
Affiliation(s)
- Farouk Segujja
- Department of Biomedical Engineering, Faculty of Technology, Kocaeli University, 41001, Kocaeli, Turkey.
| | - Gökhan Duruksu
- Department of Stem Cell, Institute of Health Sciences, Kocaeli University, 41001, Kocaeli, Turkey; Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, 41001, Kocaeli, Turkey.
| | - Elif Beyza Eren
- Department of Biomedical Engineering, Faculty of Technology, Kocaeli University, 41001, Kocaeli, Turkey
| | - Aygun İsayeva
- Department of Biomedical Engineering, Faculty of Technology, Kocaeli University, 41001, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, 41001, Kocaeli, Turkey
| | - Yusufhan Yazır
- Department of Stem Cell, Institute of Health Sciences, Kocaeli University, 41001, Kocaeli, Turkey; Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, 41001, Kocaeli, Turkey
| | - Ahmet Erdem
- Department of Biomedical Engineering, Faculty of Technology, Kocaeli University, 41001, Kocaeli, Turkey.
| |
Collapse
|
7
|
Luo Q, Liu S, Hua Y, Long C, Lv S, Li J, Zhang Y. Heterobifunctional cross-linker with dinitroimidazole and azide modules for protein and oligonucleotide functionalization. RSC Adv 2025; 15:4526-4531. [PMID: 39931413 PMCID: PMC11808663 DOI: 10.1039/d4ra07987f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
Dinitroimidazole (DNIm) was recently identified as a powerful bioconjugation agent that could selectively modify thiol over amine on biomolecules at an ultrahigh speed in an aqueous buffer. However, its derivative containing a DNIm module and a terminal alkyne module failed to construct functional agents bearing a DNIm warhead via the CuAAC reaction. To solve this problem, a heterobifunctional cross-linker was designed and synthesized by linking a DNIm module with an azide module via an oxoaliphatic amido bond spacer arm. Its two modules, DNIm and azide, reacted with a thiol and cyclooctyne, respectively, in an orthogonal way. The cross-linker facilitated the preparation of various functional agents bearing a DNIm warhead via SPAAC reaction and was further applied to protein functionalization (including biotinylation and fluorescence labeling) and oligonucleotide functionalization (including PEGylation, oligonucleotide-peptide and oligonucleotide-protein conjugate). Thus, the cross-linker not only provided convenient access to those functional agents bearing a DNIm warhead but also combined DNIm chemistry with click chemistry of SPAAC to enlarge their respective application range in the bioconjugation field.
Collapse
Affiliation(s)
- Qunfeng Luo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University Nanchang Jiangxi 330006 People's Republic of China
| | - Shuli Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University Nanchang Jiangxi 330006 People's Republic of China
| | - Yaoguang Hua
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University Nanchang Jiangxi 330006 People's Republic of China
| | - Chunqiu Long
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University Nanchang Jiangxi 330006 People's Republic of China
| | - Sijia Lv
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University Nanchang Jiangxi 330006 People's Republic of China
| | - Juncheng Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University Nanchang Jiangxi 330006 People's Republic of China
| | - Yuzhi Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University Nanchang Jiangxi 330006 People's Republic of China
| |
Collapse
|
8
|
Zaleski MH, Chase LS, Hood ED, Wang Z, Nong J, Espy CL, Zamora ME, Wu J, Morrell LJ, Muzykantov VR, Myerson JW, Brenner JS. Conjugation Chemistry Markedly Impacts Toxicity and Biodistribution of Targeted Nanoparticles, Mediated by Complement Activation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2409945. [PMID: 39663706 PMCID: PMC11795710 DOI: 10.1002/adma.202409945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/21/2024] [Indexed: 12/13/2024]
Abstract
Conjugation chemistries are a major enabling technology for the development of drug delivery systems, from antibody-drug conjugates to antibody-targeted lipid nanoparticles inspired by the success of the COVID-19 vaccine. However, here it is shown that for antibody-targeted nanoparticles, the most popular conjugation chemistries directly participate in the activation of the complement cascade of plasma proteins. Their activation of complement leads to large changes in the biodistribution of nanoparticles (up to 140-fold increased uptake into phagocytes of the lungs) and multiple toxicities, including a 50% drop in platelet count. It is founded that the mechanism of complement activation varies dramatically between different conjugation chemistries. Dibenzocyclooctyne, a commonly used click-chemistry, caused aggregation of conjugated antibodies, but only on the surface of nanoparticles (not in bulk solution). By contrast, thiol-maleimide chemistry do not activate complement via its effects on antibodies, but rather because free maleimide bonded to albumin in plasma, and clustered albumin is then attacked by complement. Using these mechanistic insights, solutions are engineered that reduced the activation of complement for each class of conjugation chemistry. These results highlight that while conjugation chemistry is essential for the future of nanomedicine, it is not innocuous and must be designed with opsonins like complement in mind.
Collapse
Affiliation(s)
- Michael H. Zaleski
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Liam S. Chase
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Elizabeth D. Hood
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Zhicheng Wang
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Jia Nong
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Carolann L. Espy
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Marco E. Zamora
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Jichuan Wu
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Lianne J. Morrell
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Vladimir R. Muzykantov
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Jacob W. Myerson
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
| | - Jacob S. Brenner
- Department of Systems Pharmacology and Translational TherapeuticsThe Perelman School of MedicineUniversity of Pennsylvania421 Curie Blvd., 354 BRB II/IIIPhiladelphiaPA19104USA
- Department of MedicineUniversity of Pennsylvania3400 Civic Center BoulevardPhiladelphiaPA19104USA
| |
Collapse
|
9
|
Yamaguchi H, Sugawa H, Takahashi H, Nagai R. Rapid and Efficient Synthesis of Succinated Thiol Compounds via Maleic Anhydride Derivatization. Molecules 2025; 30:571. [PMID: 39942675 PMCID: PMC11820211 DOI: 10.3390/molecules30030571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/24/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Succination is a non-enzymatic post-translational modification of cysteine (Cys) residues, resulting in the formation of S-(2-succino)cysteine (2SC). While hundreds of 2SC-modified proteins have been identified and are associated with the dysfunction of proteins, the underlying molecular mechanisms remain poorly understood. Conventional methods for synthesizing succinated compounds, such as 2SC, often require prolonged reaction times and/or HCl hydrolysis. In this study, we present a rapid and efficient synthesis method for succinated compounds using maleic anhydride, enabling more effective in vivo studies of succination mechanisms. This method was tested on thiol compounds with varying molecular weights, including Cys derivatives, Cys-containing peptides, and reduced bovine serum albumin. By incubating these compounds in an aqueous buffer with maleic anhydride dissolved in an organic solvent like diethyl ether, we achieved significantly improved succination efficiency compared to conventional methods. The succination efficiency using maleic anhydride surpassed that of fumaric acid or maleic acid. Notably, this approach facilitated the succination of amino acids, peptides, and proteins within minutes at 25 °C, without requiring acid hydrolysis. Our findings provide a straightforward, time-efficient strategy for synthesizing succinated thiol compounds, offering a valuable tool to enhance the understanding of succination's molecular mechanisms and its role in protein function and dysfunction.
Collapse
Affiliation(s)
- Hiroshi Yamaguchi
- Department of Food and Life Science, School of Agriculture, Tokai University, 871-12 Sugido, Mashiki, Kamimashiki, Kumamoto 861-2205, Japan; (H.S.); (R.N.)
- Graduate School of Bioscience, Tokai University, 871-12 Sugido, Mashiki, Kamimashiki, Kumamoto 861-2205, Japan;
| | - Hikari Sugawa
- Department of Food and Life Science, School of Agriculture, Tokai University, 871-12 Sugido, Mashiki, Kamimashiki, Kumamoto 861-2205, Japan; (H.S.); (R.N.)
| | - Himeno Takahashi
- Graduate School of Bioscience, Tokai University, 871-12 Sugido, Mashiki, Kamimashiki, Kumamoto 861-2205, Japan;
| | - Ryoji Nagai
- Department of Food and Life Science, School of Agriculture, Tokai University, 871-12 Sugido, Mashiki, Kamimashiki, Kumamoto 861-2205, Japan; (H.S.); (R.N.)
- Graduate School of Bioscience, Tokai University, 871-12 Sugido, Mashiki, Kamimashiki, Kumamoto 861-2205, Japan;
| |
Collapse
|
10
|
Aboshouk DR, Youssef MA, Panda SS, Kariuki BM, Bekheit MS, Hamed AR, Fayad W, Soliman AAF, Girgis AS. Design and synthesis of antiproliferative 2-oxoindolin-3-ylidenes incorporating urea function with potential VEGFR-2 inhibitory properties. Sci Rep 2025; 15:618. [PMID: 39753596 PMCID: PMC11699130 DOI: 10.1038/s41598-024-82005-6] [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: 07/31/2024] [Accepted: 12/02/2024] [Indexed: 01/06/2025] Open
Abstract
Targeted therapy is preferable over other therapeutics due to its limitation of drawbacks and better pharmaceutical outcomes. VEGF and its receptors have been observed to be hyper-activated in many cancer types and are considered promising targets for assigning anticancer agents. The current study is directed towards synthesis of novel antiproliferative 2-oxoindolin-3-ylidenes incorporating urea function with VEGFR-2 properties. The targeted agents were obtained through a two-step reaction. Addition of the appropriate 1-(acetylphenyl)-3-phenylurea 9a,b to the corresponding isatin 10a-f in ethanol containing a quantitative amount of Et2NH followed by acidic dehydration (AcOH/HCl) afforded the targeted agents 12a-j. Promising antiproliferation properties (MTT assay) were observed for most of the synthesized agents against HCT116 (colon), MCF7 (breast) and PaCa2 (pancreatic) cancer cell lines relative to sunitinib. VEGFR-2 inhibitory properties are consistent with the antiproliferation properties exhibited against the tested cell lines. Compound 12b (R = 4-NHCONHPh, R' = H; % inhibition = 87.2) is the most promising/potent anti-VEGFR-2 agent synthesized with activity close to that of sunitinib (% inhibition = 89.4) at 10 μM. Molecular docking studies (PDB: 3WZE and 3AGD) support the antiproliferation effects against cancer cell lines tested with VEGFR-2 inhibitory properties. The results are consistent with collaboration of the pharmacophores considered (2-oxoindolyl heterocycle and urea) in improving the bio-properties.
Collapse
Affiliation(s)
- Dalia R Aboshouk
- Department of Pesticide Chemistry, National Research Centre, Dokki, 12622, Giza, Egypt
| | - M Adel Youssef
- Department of Chemistry, Faculty of Science, Helwan University, Helwan, Egypt
| | - Siva S Panda
- Department of Chemistry and Biochemistry & Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, 30912, USA
| | - Benson M Kariuki
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff, CF10 3AT, UK
| | - Mohamed S Bekheit
- Department of Pesticide Chemistry, National Research Centre, Dokki, 12622, Giza, Egypt
| | - Ahmed R Hamed
- Chemistry of Medicinal Plants Department, National Research Centre, Dokki, 12622, Giza, Egypt
| | - Walid Fayad
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, 12622, Giza, Egypt
| | - Ahmed A F Soliman
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, 12622, Giza, Egypt
| | - Adel S Girgis
- Department of Pesticide Chemistry, National Research Centre, Dokki, 12622, Giza, Egypt.
| |
Collapse
|
11
|
Zhang H, Guo L, Li X, Liu H, Zhao Z, Ji G, Huang Y, Wang X. A Modular Approach to Obtain HER2-Targeting DM1-Loaded Nanoparticles for Gastric Cancer Therapy. ACS OMEGA 2024; 9:48598-48606. [PMID: 39676924 PMCID: PMC11635515 DOI: 10.1021/acsomega.4c07442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 11/10/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024]
Abstract
Antibody-based tumor-targeting nanomedicines, despite their high efficacy, present significant challenges in preparation and long-term storage. We introduce a novel approach for the synthesis of durable, ready-to-use, antibody-coupled nanomedical drugs. Our research centers on the development of HER2-targeting DM1-loaded nanoparticles for gastric cancer treatment using a modular methodology. We synthesized Fc-PLG-Mal, conjugated DM1 through a "click" reaction, and subsequently bound the resultant compound with the HER2 antibody trastuzumab. The nanoparticles demonstrated a high drug loading content, stable particle size, and effective HER2 targeting. HER2-PLG-DM1 exhibited significant cytotoxicity against NCI-N87 gastric cancer cells, with an IC50 of 0.35 nM. Biodistribution revealed rapid and substantial tumor accumulation, 6-fold higher than that of nontargeting IgG-PLG-DM1. HER2-PLG-DM1 significantly inhibited tumor growth in NCI-N87 tumor-bearing mice, achieving a 90.8% tumor inhibition rate, and displayed dose-dependent effects without significant liver and kidney toxicity. These studies offer an efficient and stable method for the preparation of antibody-coupled drugs.
Collapse
Affiliation(s)
- Hui Zhang
- Department
of Clinical Laboratory Medicine, The Affiliated
Hospital to Changchun University of Chinese Medicine, Changchun, Jilin Province 130021, China
| | - Lijiao Guo
- Department
of Clinical Laboratory Medicine, The Affiliated
Hospital to Changchun University of Chinese Medicine, Changchun, Jilin Province 130021, China
| | - Xue Li
- Department
of Clinical Laboratory Medicine, The Affiliated
Hospital to Changchun University of Chinese Medicine, Changchun, Jilin Province 130021, China
| | - Hongtao Liu
- Jilin
Academy
of Chinese Medicine Sciences, Changchun, Jilin Province 130117, China
| | - Zibin Zhao
- Jilin
Academy
of Chinese Medicine Sciences, Changchun, Jilin Province 130117, China
| | - Guangling Ji
- Gastroenteric
Medicine and Digestive Endoscopy Center, The Second Hospital of Jilin University, Changchun, Jilin Province 130041, China
| | - Yue Huang
- Key Laboratory
of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun 130022, China
| | - Xiaodong Wang
- Gastroenteric
Medicine and Digestive Endoscopy Center, The Second Hospital of Jilin University, Changchun, Jilin Province 130041, China
| |
Collapse
|
12
|
Jing L, Wu G, Zhao F, Jiang X, Liu N, Feng D, Sun Y, Zhang T, De Clercq E, Pannecouque C, Kang D, Liu X, Zhan P. Discovery of potent HIV-1 NNRTIs by CuAAC click-chemistry-based miniaturized synthesis, rapid screening and structure optimization. Eur J Med Chem 2024; 277:116772. [PMID: 39167895 DOI: 10.1016/j.ejmech.2024.116772] [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: 06/20/2024] [Revised: 07/31/2024] [Accepted: 08/11/2024] [Indexed: 08/23/2024]
Abstract
In addressing the urgent need for novel HIV-1 non-nucleoside reverse transcriptase inhibitors (NNRTIs) to combat drug resistance, we employed CuAAC click chemistry to construct a diverse 312-member diarylpyrimidine (DAPY) derivative library. This rapid synthesis approach facilitated the identification of A6N36, demonstrating exceptional HIV-1 RT inhibitory activity. Moreover, it was demonstrated with EC50 values of 1.8-8.7 nM for mutant strains L100I, K103 N, Y181C, and E138K, being equipotent or superior to that of ETR. However, A6N36's efficacy was compromised against specific resistant strains (Y188L, F227L + V106A and RES056), highlighting a need for further optimization. Through scaffold hopping, we optimized this lead to develop 10c, which exhibited broad-spectrum activity with EC50 values ranging from 3.2 to 57.5 nM and superior water solubility. Molecular docking underscored the key interactions of 10c within the NNIBP. Our findings present 10c as a promising NNRTI lead, illustrating the power of click chemistry and rational design in combatting HIV-1 resistance.
Collapse
Affiliation(s)
- Lanlan Jing
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China
| | - Gaochan Wu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China
| | - Fabao Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China
| | - Xiangyi Jiang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China
| | - Na Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China
| | - Da Feng
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China
| | - Yanying Sun
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China
| | - Tao Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China
| | - Erik De Clercq
- Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, K.U. Leuven, Leuven, B-3000, Belgium
| | - Christophe Pannecouque
- Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, K.U. Leuven, Leuven, B-3000, Belgium
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China.
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China.
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China.
| |
Collapse
|
13
|
Tarab-Ravski D, Stotsky-Oterin L, Elisha A, Kundoor GR, Ramishetti S, Hazan-Halevy I, Haas H, Peer D. The future of genetic medicines delivered via targeted lipid nanoparticles to leukocytes. J Control Release 2024; 376:286-302. [PMID: 39401676 DOI: 10.1016/j.jconrel.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Genetic medicines hold vast therapeutic potential, offering the ability to silence or induce gene expression, knock out genes, and even edit DNA fragments. Applying these therapeutic modalities to leukocytes offers a promising path for treating various conditions yet overcoming the obstacles of specific and efficient delivery to leukocytes remains a major bottleneck in their clinical translation. Lipid nanoparticles (LNPs) have emerged as the leading delivery system for nucleic acids due to their remarkable versatility and ability to improve their in vivo stability, pharmacokinetics, and therapeutic benefits. Equipping LNPs with targeting moieties can promote their specific cellular uptake and internalization to leukocytes, making targeted LNPs (tLNPs) an inseparable part of developing leukocyte-targeted gene therapy. However, despite the significant advancements in research, genetic medicines for leukocytes using targeted delivery approaches have not been translated into the clinic yet. Herein, we discuss the important aspects of designing tLNPs and highlight the considerations for choosing an appropriate bioconjugation strategy and targeting moiety. Furthermore, we provide our insights on limiting challenges and identify key areas for further research to advance these exciting therapies for patient care.
Collapse
Affiliation(s)
- Dana Tarab-Ravski
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Lior Stotsky-Oterin
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Aviad Elisha
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Govinda Reddy Kundoor
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | | | - Inbal Hazan-Halevy
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Heinrich Haas
- NeoVac Ltd. 127 Olympic Ave., OX14 4SA, Milton Park, Oxfordshire, UK; Department of Biopharmaceutics and Pharmaceutical Technology, Johannes Gutenberg-University, Mainz, Germany
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
14
|
Raja A, Kasana A, Verma V. Next-Generation Therapeutic Antibodies for Cancer Treatment: Advancements, Applications, and Challenges. Mol Biotechnol 2024:10.1007/s12033-024-01270-y. [PMID: 39222285 DOI: 10.1007/s12033-024-01270-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
The field of cancer treatment has evolved significantly over the last decade with the emergence of next-generation therapeutic antibodies. Conventional treatments like chemotherapy pose significant challenges, including adverse side effects. Monoclonal antibodies have paved the way for more targeted and effective interventions. The evolution from chimeric to humanized and fully human antibodies has led to a reduction in immunogenicity and enhanced tolerance in vivo. The advent of next-generation antibodies, including bispecific antibodies, nanobodies, antibody-drug conjugates, glyco-engineered antibodies, and antibody fragments, represents a leap forward in cancer therapy. These innovations offer increased potency, adaptability, and reduced drug resistance. Challenges such as target validation, immunogenicity, and high production costs exist. However, technological advancements in antibody engineering techniques provide optimism for addressing these issues. The future promises a paradigm shift, where ongoing research will propel these powerful antibodies to the forefront, revolutionizing the fight against cancer and creating new preventive and curative treatments. This review provides an overview of three next-generation antibody-based molecules, namely bispecific antibodies, antibody-drug conjugates, and nanobodies that have shown promising results in cancer treatment. It discusses the evolution of antibodies from conventional forms to next-generation molecules, along with their applications in cancer treatment, production methods, and associated challenges. The review aims to offer researchers insights into the evolving landscape of next-generation antibody-based cancer therapeutics and their potential to revolutionize treatment strategies.
Collapse
Affiliation(s)
- Abhavya Raja
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, 201310, Uttar Pradesh, India
| | - Abhishek Kasana
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, 201310, Uttar Pradesh, India
| | - Vaishali Verma
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, 201310, Uttar Pradesh, India.
| |
Collapse
|
15
|
Wang J, Zheng Y, Huang H, Ma Y, Zhao X. An overview of signal amplification strategies and construction methods on phage-based biosensors. Food Res Int 2024; 191:114727. [PMID: 39059923 DOI: 10.1016/j.foodres.2024.114727] [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: 04/22/2024] [Revised: 06/12/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024]
Abstract
Phages are a class of viruses that specifically infect host bacteria. Compared to other recognition elements, phages offer several advantages such as high specificity, easy to obtain and good environmental tolerance, etc. These advantages underscore the potential of phages as recognition elements in the construction of biosensors. Therefore, the phage-based biosensors are currently garnering widespread attention for detecting pathogens in recent years. However, the test performance such as detection limit, sensitivity and stability of exicting phage-based biosensors require enhancement. In the design of sensors, the selection of various materials and construction methods significantly influences the test performance of the sensor, and employing appropriate signal amplification strategies and construction methods to devise biosensors based on different principles is an effective strategy to enhance sensor performance. The manuscript primarily focuses on the signal amplification strategies and construction methods employed in phage-based biosensors recent ten years, and summarizes the advantages and disadvantages of different signal amplification strategies and construction methods. Meanwhile, the manuscript discusses the relationship between sensor performance and various materials and construction methods, and reviews the application progress of phage-based electrochemical biosensors in the detection of foodborne bacteria. Furthermore, the manuscript points out the present limitations and the future research direction for the field of phage-based biosensors, so as to provide the reference for developing high-performance phage-based biosensors.
Collapse
Affiliation(s)
- Jiahao Wang
- College of Light Industry and Food Science, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China.
| | - Yuqing Zheng
- College of Light Industry and Food Science, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China.
| | - Hongkai Huang
- College of Light Industry and Food Science, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China.
| | - Ya Ma
- College of Light Industry and Food Science, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China.
| | - Xiaojuan Zhao
- College of Light Industry and Food Science, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China.
| |
Collapse
|
16
|
Morishita M, Makabe M, Shinohara C, Fukumori A, Morita S, Terada Y, Miyai S, Katsumi H, Yamamoto A. Versatile functionalization of Bifidobacteria-derived extracellular vesicles using amino acid metabolic labeling and click chemistry for immunotherapy. Int J Pharm 2024; 661:124410. [PMID: 38954931 DOI: 10.1016/j.ijpharm.2024.124410] [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: 03/03/2024] [Revised: 06/02/2024] [Accepted: 06/29/2024] [Indexed: 07/04/2024]
Abstract
Extracellular vesicles (EVs) are nanoparticles secreted by various organisms. Methods for modifying EVs functionally have garnered attention for developing EV-based therapeutic systems. However, most technologies used to integrate these functions are limited to mammalian-derived EVs and a promising modification method for bacteria-derived EVs has not yet been developed. In this study, we propose a novel method for the versatile functionalization of immunostimulatory probiotic Bifidobacteria-derived EVs (B-EVs) using amino acid metabolic labeling and azide-alkyne click reaction. Azide D-alanine (ADA), a similar molecule to D-alanine in bacteria cell-wall peptidoglycan, was selected as an azide group-functionalized amino acid. Azide-modified B-EVs were isolated from Bifidobacteria incubated with ADA. The physicochemical and compositional characteristics, as well as adjuvanticity of B-EVs against immune cells were not affected by azide loading, demonstrating that this functionalization approach can retain the endogenous usefulness of B-EVs. By using the fluorescent B-EVs obtained by this method, the intracellular trafficking of B-EVs after uptake by immune cells was successfully observed. Furthermore, this method enabled the formulation of B-EVs for hydrogelation and enhanced adjuvanticity in the host. Our findings will be helpful for further development of EV-based immunotherapy.
Collapse
Affiliation(s)
- Masaki Morishita
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan.
| | - Mizuho Makabe
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Chisa Shinohara
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Ami Fukumori
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Shiori Morita
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Yuki Terada
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Syunsuke Miyai
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Hidemasa Katsumi
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| |
Collapse
|
17
|
Geisler HC, Ghalsasi AA, Safford HC, Swingle KL, Thatte AS, Mukalel AJ, Gong N, Hamilton AG, Han EL, Nachod BE, Padilla MS, Mitchell MJ. EGFR-targeted ionizable lipid nanoparticles enhance in vivo mRNA delivery to the placenta. J Control Release 2024; 371:455-469. [PMID: 38789090 PMCID: PMC11259947 DOI: 10.1016/j.jconrel.2024.05.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 05/15/2024] [Accepted: 05/19/2024] [Indexed: 05/26/2024]
Abstract
The full potential of ionizable lipid nanoparticles (LNPs) as an in vivo nucleic acid delivery platform has not yet been realized given that LNPs primarily accumulate in the liver following systemic administration, limiting their success to liver-centric conditions. The engineering of LNPs with antibody targeting moieties can enable extrahepatic tropism by facilitating site-specific LNP tethering and driving preferential LNP uptake into receptor-expressing cell types via receptor-mediated endocytosis. Obstetric conditions stemming from placental dysfunction, such as preeclampsia, are characterized by overexpression of cellular receptors, including the epidermal growth factor receptor (EGFR), making targeted LNP platforms an exciting potential treatment strategy for placental dysfunction during pregnancy. Herein, an EGFR antibody-conjugated LNP (aEGFR-LNP) platform was developed by engineering LNPs with increasing densities of antibody functionalization. aEGFR-LNPs were screened in vitro in immortalized placental trophoblasts and in vivo in non-pregnant and pregnant mice and compared to non-targeted formulations for extrahepatic, antibody-targeted mRNA LNP delivery to the placenta. Our top performing LNP with an intermediate density of antibody functionalization (1:5 aEGFR-LNP) mediated a ∼twofold increase in mRNA delivery in murine placentas and a ∼twofold increase in LNP uptake in EGFR-expressing trophoblasts compared to non-targeted counterparts. These results demonstrate the potential of antibody-conjugated LNPs for achieving extrahepatic tropism, and the ability of aEGFR-LNPs in promoting mRNA delivery to EGFR-expressing cell types in the placenta.
Collapse
Affiliation(s)
- Hannah C Geisler
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Aditi A Ghalsasi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Hannah C Safford
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Kelsey L Swingle
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Ajay S Thatte
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Alvin J Mukalel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Alex G Hamilton
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Emily L Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Benjamin E Nachod
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Marshall S Padilla
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States; Penn Institute for RNA Innovation, Perelman School of Medicine, Philadelphia, PA, United States; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
18
|
Nemecz D, Nowak WA, Nemecz Á. VHH Nanobody Versatility against Pentameric Ligand-Gated Ion Channels. J Med Chem 2024; 67:8502-8518. [PMID: 38829690 PMCID: PMC11181324 DOI: 10.1021/acs.jmedchem.4c00231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/17/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024]
Abstract
Pentameric ligand-gated ion channels provide rapid chemical-electrical signal transmission between cells in the central and peripheral nervous system. Their dysfunction is associated with many nervous system disorders. They are composed of five identical (homomeric receptors) or homologous (heteromeric receptors) subunits. VHH nanobodies, or single-chain antibodies, are the variable domain, VHH, of antibodies that are composed of the heavy chain only from camelids. Their unique structure results in many specific biochemical and biophysical properties that make them an excellent alternative to conventional antibodies. This Perspective explores the published VHH nanobodies which have been isolated against pentameric ligand-gated ion channel subfamilies. It outlines the genetic and chemical modifications available to alter nanobody function. An assessment of the available functional and structural data indicate that it is feasible to create therapeutic agents and impart, through their modification, a given desired modulatory effect of its target receptor for current stoichiometric-specific VHH nanobodies.
Collapse
Affiliation(s)
- Dorota Nemecz
- Biochemistry
Department, Nicolaus Copernicus University
in Torun, 87-100 Torun, Poland
| | - Weronika A. Nowak
- Biochemistry
Department, Nicolaus Copernicus University
in Torun, 87-100 Torun, Poland
| | - Ákos Nemecz
- Biochemistry
Department, Nicolaus Copernicus University
in Torun, 87-100 Torun, Poland
| |
Collapse
|
19
|
Aboshouk DR, Youssef MA, Bekheit MS, Hamed AR, Girgis AS. Antineoplastic indole-containing compounds with potential VEGFR inhibitory properties. RSC Adv 2024; 14:5690-5728. [PMID: 38362086 PMCID: PMC10866129 DOI: 10.1039/d3ra08962b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 01/29/2024] [Indexed: 02/17/2024] Open
Abstract
Cancer is one of the most significant health challenges worldwide. Various techniques, tools and therapeutics/materials have been developed in the last few decades for the treatment of cancer, together with great interest, funding and efforts from the scientific society. However, all the reported studies and efforts seem insufficient to combat the various types of cancer, especially the advanced ones. The overexpression of tyrosine kinases is associated with cancer proliferation and/or metastasis. VEGF, an important category of tyrosine kinases, and its receptors (VEGFR) are hyper-activated in different cancers. Accordingly, they are known as important factors in the angiogenesis of different tumors and are considered in the development of effective therapeutic approaches for controlling many types of cancer. In this case, targeted therapeutic approaches are preferable to the traditional non-selective approaches to minimize the side effects and drawbacks associated with treatment. Several indole-containing compounds have been identified as effective agents against VEGFR. Herein, we present a summary of the recent indolyl analogs reported within the last decade (2012-2023) with potential antineoplastic and VEGFR inhibitory properties. The most important drugs, natural products, synthesized potent compounds and promising hits/leads are highlighted. Indoles functionalized and conjugated with various heterocycles beside spiroindoles are also considered.
Collapse
Affiliation(s)
- Dalia R Aboshouk
- Department of Pesticide Chemistry, National Research Centre Dokki Giza 12622 Egypt
| | - M Adel Youssef
- Department of Chemistry, Faculty of Science, Helwan University Helwan Egypt
| | - Mohamed S Bekheit
- Department of Pesticide Chemistry, National Research Centre Dokki Giza 12622 Egypt
| | - Ahmed R Hamed
- Chemistry of Medicinal Plants Department, National Research Centre Dokki Giza 12622 Egypt
| | - Adel S Girgis
- Department of Pesticide Chemistry, National Research Centre Dokki Giza 12622 Egypt
| |
Collapse
|
20
|
Podolak M, Holota S, Deyak Y, Dziduch K, Dudchak R, Wujec M, Bielawski K, Lesyk R, Bielawska A. Tubulin inhibitors. Selected scaffolds and main trends in the design of novel anticancer and antiparasitic agents. Bioorg Chem 2024; 143:107076. [PMID: 38163424 DOI: 10.1016/j.bioorg.2023.107076] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/02/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
Design of tubulin inhibitors as anticancer drugs dynamically developed over the past 20 years. The modern arsenal of potential tubulin-targeting anticancer agents is represented by small molecules, monoclonal antibodies, and antibody-drug conjugates. Moreover, targeting tubulin has been a successful strategy in the development of antiparasitic drugs. In the present review, an overall picture of the research and development of potential tubulin-targeting agents using small molecules between 2018 and 2023 is provided. The data about some most often used and prospective chemotypes of small molecules (privileged heterocycles, moieties of natural molecules) and synthetic methodologies (analogue-based, fragment-based drug design, molecular hybridization) applied for the design of novel agents with an impact on the tubulin system are summarized. The design and prospects of multi-target agents with an impact on the tubulin system were also highlighted. Reported in the review data contribute to the "structure-activity" profile of tubulin-targeting small molecules as anticancer and antiparasitic agents and will be useful for the application by medicinal chemists in further exploration, design, improvement, and optimization of this class of molecules.
Collapse
Affiliation(s)
- Magdalena Podolak
- Department of Biotechnology, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland
| | - Serhii Holota
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine
| | - Yaroslava Deyak
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine; Department of Pharmaceutical Disciplines, Uzhhorod National University, Narodna Square 3, 88000 Uzhhorod, Ukraine
| | - Katarzyna Dziduch
- Doctoral School, Medical University of Lublin, Chodzki 7, 20-093 Lublin, Poland
| | - Rostyslav Dudchak
- Department of Biotechnology, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland
| | - Monika Wujec
- Department of Organic Chemistry, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland
| | - Krzysztof Bielawski
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine.
| | - Anna Bielawska
- Department of Biotechnology, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland
| |
Collapse
|