1
|
Vesamäki S, Meteling H, Nasare R, Siiskonen A, Patrakka J, Roas-Escalona N, Linder M, Virkki M, Priimagi A. Strategies to control humidity sensitivity of azobenzene isomerisation kinetics in polymer thin films. COMMUNICATIONS MATERIALS 2024; 5:209. [PMID: 39371916 PMCID: PMC11446815 DOI: 10.1038/s43246-024-00642-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/13/2024] [Indexed: 10/08/2024]
Abstract
Azobenzenes are versatile photoswitches that garner interest in applications ranging from photobiology to energy storage. Despite their great potential, transforming azobenzene-based discoveries and proof-of-concept demonstrations from the lab to the market is highly challenging. Herein we give an overview of a journey that started from a discovery of hydroxyazobenzene's humidity sensitive isomerisation kinetics, developed into commercialization efforts of azobenzene-containing thin film sensors for optical monitoring of the relative humidity of air, and arrives to the present work aiming for better design of such sensors by understanding the different factors affecting the humidity sensitivity. Our concept is based on thermal isomerisation kinetics of tautomerizable azobenzenes in polymer matrices which, using pre-defined calibration curves, can be converted to relative humidity at known temperature. We present a small library of tautomerizable azobenzenes exhibiting humidity sensitive isomerisation kinetics in hygroscopic polymer films. We also investigate how water absorption properties of the polymer used, and the isomerisation kinetics are linked and how the azobenzene content in the thin film affects both properties. Based on our findings we propose simple strategies for further development of azobenzene-based optical humidity sensors.
Collapse
Affiliation(s)
- Sami Vesamäki
- Faculty of Engineering and Natural Sciences, Tampere University, Tampere, Finland
| | - Henning Meteling
- Faculty of Engineering and Natural Sciences, Tampere University, Tampere, Finland
| | - Roshan Nasare
- Faculty of Engineering and Natural Sciences, Tampere University, Tampere, Finland
| | - Antti Siiskonen
- Faculty of Engineering and Natural Sciences, Tampere University, Tampere, Finland
| | - Jani Patrakka
- Faculty of Engineering and Natural Sciences, Tampere University, Tampere, Finland
| | | | - Markus Linder
- Department of Bioproducts and Biosystems, Aalto University, Espoo, Finland
| | - Matti Virkki
- VTT Technical Research Centre of Finland Ltd, Oulu, Finland
| | - Arri Priimagi
- Faculty of Engineering and Natural Sciences, Tampere University, Tampere, Finland
| |
Collapse
|
2
|
Zhao Y, Huang Q, Liu Y. Recent Advances of Light/Hypoxia-Responsive Azobenzene in Nanomedicine Design. Chembiochem 2024:e202400635. [PMID: 39252178 DOI: 10.1002/cbic.202400635] [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: 07/30/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 09/11/2024]
Abstract
Azobenzene (Azo) and its derivatives are versatile stimuli-responsive molecules. Their reversible photoisomerization and susceptibility to reduction-mediated cleavage make them valuable for various biomedical applications. Upon exposure to the UV light, Azo units undergo a thermodynamically stable trans-to-cis transition, which can be reversed by heating in the dark or irradiation with visible light. Additionally, the N=N bonds in azobenzenes can be cleaved under hypoxic conditions by azoreductase, making azobenzenes useful as hypoxia-responsive linkers. The integration of azobenzenes into nanomedicines holds promise for enhancing therapeutic efficacy, particularly in tumor targeting and controllable drug release. In this Concept paper, recent advances in the design and applications of azobenzene-based nanomedicines are updated, and future development opportunities are also summarized.
Collapse
Affiliation(s)
- Yu Zhao
- College of Chemistry, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, China
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Qingqing Huang
- College of Chemistry, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Yang Liu
- College of Chemistry, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, 300071, China
| |
Collapse
|
3
|
Alhamdi HW, Alfaifi MY, Shati AA, Elbehairi SEI, Er-Rajy M, Elshaarawy RFM, Hassan YA, Zakrya R. New multifunctional hybrids as modulators of apoptosis markers and topoisomerase II in breast cancer therapy: synthesis, characterization, and in vitro and in silico studies. RSC Adv 2024; 14:28555-28568. [PMID: 39247509 PMCID: PMC11378026 DOI: 10.1039/d4ra04219k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024] Open
Abstract
Recently, molecular hybrids of two or more active pharmacophores have shown promise for designing and synthesizing anticancer drugs. Herein, a new multifunctional hybrid (PAHMQ), combining azobenzene and quinoline pharmacophores, and its M(ii) complexes (MPAHMQ) have been successfully developed and structurally characterized. The MTT assay revealed CuBHTP as the most efficient and safe breast cancer treatment, with an IC50 of 11.18 ± 0.39 μg mL-1 and a high selectivity index (SI) of 5.63 for cancer MCF-7 cells over healthy MCF10A cells. Moreover, the CuPAHMQ-treated MCF-7 cells experience a dramatic impact with regard to key apoptotic markers, including an increase in P53 and Bax expression, with a decrease in Bcl-2 expression levels compared to the untreated MCF-7 cells. Additionally, CuPAHMQ effectively halted the growth and division of MCF-7 cells by inducing cell cycle arrest in the crucial G1 and S phases, ultimately inhibiting both Topo II activity and cell proliferation. Molecular docking investigations validated the CuPAHMQ complex's groove binding and topoisomerase II binding, establishing it as a potent anticancer drug.
Collapse
Affiliation(s)
- Heba W Alhamdi
- College of Sciences, Biology Department, King Khalid University Abha 61413 Saudi Arabia
| | - Mohammad Y Alfaifi
- King Khalid University, Faculty of Science, Biology Department Abha 9004 Saudi Arabia
- Tissue Culture and Cancer Biology Research Laboratory, King Khalid University Abha 9004 Saudi Arabia
| | - Ali A Shati
- King Khalid University, Faculty of Science, Biology Department Abha 9004 Saudi Arabia
- Tissue Culture and Cancer Biology Research Laboratory, King Khalid University Abha 9004 Saudi Arabia
| | - Serag Eldin I Elbehairi
- King Khalid University, Faculty of Science, Biology Department Abha 9004 Saudi Arabia
- Tissue Culture and Cancer Biology Research Laboratory, King Khalid University Abha 9004 Saudi Arabia
- Cell Culture Lab, Egyptian Organization for Biological Products and Vaccines (VACSERA Holding Company) 51 Wezaret El-Zeraa St., Agouza Giza Egypt
| | - Mohammed Er-Rajy
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University Fez Morocco
| | - Reda F M Elshaarawy
- Department of Chemistry, Faculty of Science, Suez University 43533 Suez Egypt
| | - Yasser A Hassan
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology Gamasa Egypt
| | - Rozan Zakrya
- Chemistry Department, Faculty of Science, Port-Said University Port-Said Egypt
| |
Collapse
|
4
|
Qausain S, Basheeruddin M. Therapeutic Applications of Azo Dye Reduction: Insights From Methyl Orange Degradation for Biomedical Innovations. Cureus 2024; 16:e69952. [PMID: 39445263 PMCID: PMC11496386 DOI: 10.7759/cureus.69952] [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: 08/20/2024] [Accepted: 09/22/2024] [Indexed: 10/25/2024] Open
Abstract
This paper emphasizes the possible application of methyl orange reduction as a therapeutic technique, highlighting the potential of azo dye reduction in biomedical fields. The generally used azo dyes are toxic and carcinogenic; hence, they implicitly threaten the environment and health. The degradation of methyl orange, a famous example of azo dyes, is used to describe the degradation process for other azo dyes. This work discusses the ability of different methyl orange degradation methods, focusing on biocatalysts and nanomaterials, among the methods that identified enzymatic degradation with azoreductase enzymes as the method that quickly breaks down azo dyes under mild conditions as the most appropriate method, as well as its specificity as environmentally friendly. Moreover, metal nanoparticles such as silver and gold impellers increase the reducing efficiency because they offer a pivotal surface for the reduction reactions that undergo electron transfer. The complete breakdown of methyl orange is essential in biomedical usage. The strategies for treating azo dye reduction can be extended to next-generation drug delivery systems (DDS), biosensors, and therapeutic agents. Organisms involved in degradation can be functionalized to selectively degrade specific cells or tissues, thus presenting a new targeted therapy. Knowledge of degradation pathways and non-toxic products is essential in creating programs that build better and more efficient therapeutic agents. This work endeavors to illustrate the development of enzymatic and nanomaterials-based approaches to achieve sustainable azo dye decolorisation to open the gateway to developing other biomedical applications that tend to promote environmental and health-friendly solutions.
Collapse
Affiliation(s)
- Sana Qausain
- Biochemistry, Jawaharlal Nehru Medical College, Wardha, IND
| | | |
Collapse
|
5
|
Li M, Dong Y, Wang Z, Zhao Y, Dai Y, Zhang B. Engineering hypoxia-responsive 6-aminonicotinamide prodrugs for on-demand NADPH depletion and redox manipulation. J Mater Chem B 2024; 12:8067-8075. [PMID: 39129477 DOI: 10.1039/d4tb01338g] [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: 08/13/2024]
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) is a promising target in cancer therapy. However, poor cellular uptake and off-target toxicity have impeded the clinical translation of a canonical G6PD inhibitor (6-aminonicotinamide/6AN). Here, we report a prodrug strategy to address this issue. The tailored 6AN prodrug contains an azo-bearing protection moiety. The hydrophobic prodrug showed increased cellular uptake than 6AN and was vulnerable to hypoxia, resulting in NAD(P)H quinone dehydrogenase 1 (NQO1)-triggered cleavage of azo bonds. Intriguingly, the prodrug showed configuration-dependent anti-cancer potency. Despite the lower thermodynamic stability, the cis isomer showed enhanced cellular uptake compared to the trans counterpart due to the increased aqueous solubility. Moreover, the boosted potency of the cis isomer compared to the trans isomer arose from the enhancement of NOQ1-catalyzed 6AN release under hypoxia, a hallmark of solid tumors. The discovery of hypoxia-responsive 6AN prodrugs in the current work opens up new avenues for G6PD-targeting cancer medicines.
Collapse
Affiliation(s)
- Mingye Li
- Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, School of Pharmaceutical Science & Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China.
| | - Yuyu Dong
- Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, School of Pharmaceutical Science & Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China.
| | - Zheng Wang
- Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, School of Pharmaceutical Science & Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China.
| | - Yanjun Zhao
- Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, School of Pharmaceutical Science & Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China.
| | - Yujie Dai
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, No. 29 of 13th Street, TEDA, Tianjin 300457, China.
| | - Baoxin Zhang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Huimin District, Hohhot, 010000, China.
| |
Collapse
|
6
|
Khan A. Cleavable azobenzene linkers for the design of stimuli-responsive materials. Chem Commun (Camb) 2024; 60:6591-6602. [PMID: 38872512 DOI: 10.1039/d4cc02311k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
The azo linkage (NN) is one of the very few functional groups in organic chemistry that exhibits sensitivity towards thermal, chemical, photochemical, and biological stimuli. Consequently, this property has given rise to a distinct class of responsive materials. For example, thermal sensitivity has led to generation of free radical initiators useful in curing and polymerization applications. Chemically-induced cleavage has aided the development of self-immolative polymers and reactive scaffolds for proteomics applications. Photo-isomerization capability has given rise to photo-responsive systems. Azobenzene cleavage in biologically reducing environments, such as that of the colon, and under tumor hypoxia conditions has led to diagnostic, therapeutic, and delivery materials. Such conditions have also allowed for control over formation (assembly) and disruption (disassembly) of micellar nanoparticles. The aim of this review article is to look beyond the prevalent photosensitivity aspect of the aromatic azo compounds and draw attention to the azo scission reaction as a trigger of the change in the structure and properties of organic materials. Thus, the main discussion begins with the mechanism of the reductive cleavage. Then, its application in the design of molecules that can be activated as drugs and fluorescent sensors, (nano)materials with potential to release active substances, and polymers with side-chain and main-chain self-immolative capacity is discussed. Finally, the status and future challenges in this field are discussed.
Collapse
Affiliation(s)
- Anzar Khan
- National Institute for Research and Development of Isotopic and Molecular Technologies - INCDTIM, 67-103 Donat Street, 400293 Cluj-Napoca, Romania.
| |
Collapse
|
7
|
Chen H, Tang Z, Yang Y, Hao Y, Chen W. Recent Advances in Photoswitchable Fluorescent and Colorimetric Probes. Molecules 2024; 29:2521. [PMID: 38893396 PMCID: PMC11173890 DOI: 10.3390/molecules29112521] [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: 04/28/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
In recent years, significant advancements have been made in the research of photoswitchable probes. These probes undergo reversible structural and electronic changes upon light exposure, thus exhibiting vast potential in molecular detection, biological imaging, material science, and information storage. Through precisely engineered molecular structures, the photoswitchable probes can toggle between "on" and "off" states at specific wavelengths, enabling highly sensitive and selective detection of targeted analytes. This review systematically presents photoswitchable fluorescent and colorimetric probes built on various molecular photoswitches, primarily focusing on the types involving photoswitching in their detection and/or signal response processes. It begins with an analysis of various molecular photoswitches, including their photophysical properties, photoisomerization and photochromic mechanisms, and fundamental design concepts for constructing photoswitchable probes. The article then elaborates on the applications of these probes in detecting diverse targets, including cations, anions, small molecules, and biomacromolecules. Finally, it offers perspectives on the current state and future development of photoswitchable probes. This review aims to provide a clear introduction for researchers in the field and guidance for the design and application of new, efficient fluorescent and colorimetric probes.
Collapse
Affiliation(s)
- Hongjuan Chen
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China; (H.C.); (Y.Y.)
| | - Zilong Tang
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China; (H.C.); (Y.Y.)
| | - Yewen Yang
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China; (H.C.); (Y.Y.)
| | - Yuanqiang Hao
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China; (H.C.); (Y.Y.)
| | - Wansong Chen
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410017, China
| |
Collapse
|
8
|
Sanna AL, Pachova T, Catellani A, Calzolari A, Sforazzini G. Meta-Substituted Asymmetric Azobenzenes: Insights into Structure-Property Relationship. Molecules 2024; 29:1929. [PMID: 38731420 PMCID: PMC11085191 DOI: 10.3390/molecules29091929] [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: 04/03/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
This article presents a comprehensive investigation into the functionalization of methoxyphenylazobenzene using electron-directing groups located at the meta position relative to the azo group. Spectroscopic analysis of meta-functionalized azobenzenes reveals that the incorporation of electron-withdrawing units significantly influences the absorption spectra of both E and Z isomers, while electron-donating functionalities lead to more subtle changes. The thermal relaxation process from Z to E result in almost twice as prolonged for electron-withdrawing functionalized azobenzenes compared to their electron-rich counterparts. Computational analysis contributes a theoretical understanding of the electronic structure and properties of meta-substituted azobenzenes. This combined approach, integrating experimental and computational techniques, yields significant insights into the structure-property relationship of meta-substituted asymmetrical phenolazobenzenes.
Collapse
Affiliation(s)
- Anna Laura Sanna
- Department of Chemical and Geological Sciences, Università degli Studi di Cagliari, SS 554, Bivio per Sestu, 09042 Cagliari, Italy
| | - Tatiana Pachova
- Laboratory of Macromolecular and Organic Materials, Institute of Material Science and Engineering, Ecole Polytechnique Federale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | | | - Arrigo Calzolari
- CNR-NANO, Istituto Nanoscienze, Via Giuseppe Campi, 213, 41125 Modena, Italy
| | - Giuseppe Sforazzini
- Department of Chemical and Geological Sciences, Università degli Studi di Cagliari, SS 554, Bivio per Sestu, 09042 Cagliari, Italy
- Laboratory of Macromolecular and Organic Materials, Institute of Material Science and Engineering, Ecole Polytechnique Federale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
9
|
Patra R, Halder S, Saha R, Jana K, Sarkar K. Highly Efficient Photoswitchable Smart Polymeric Nanovehicle for Gene and Anticancer Drug Delivery in Triple-Negative Breast Cancer. ACS Biomater Sci Eng 2024; 10:2299-2323. [PMID: 38551335 DOI: 10.1021/acsbiomaterials.4c00115] [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] [Indexed: 04/09/2024]
Abstract
Over the past few decades, there has been significant interest in smart drug delivery systems capable of carrying multiple drugs efficiently, particularly for treating genetic diseases such as cancer. Despite the development of various drug delivery systems, a safe and effective method for delivering both anticancer drugs and therapeutic genes for cancer therapy remains elusive. In this study, we describe the synthesis of a photoswitchable smart polymeric vehicle comprising a photoswitchable spiropyran moiety and an amino-acid-based cationic monomer-based block copolymer using reversible addition-fragmentation chain transfer (RAFT) polymerization. This system aims at diagnosing triple-negative breast cancer and subsequently delivering genes and anticancer agents. Triple-negative breast cancer patients have elevated concentrations of Cu2+ ions, making them excellent targets for diagnosis. The polymer can detect Cu2+ ions with a low limit of detection value of 9.06 nM. In vitro studies on doxorubicin drug release demonstrated sustained delivery at acidic pH level similar to the tumor environment. Furthermore, the polymer exhibited excellent blood compatibility even at the concentration as high as 500 μg/mL. Additionally, it displayed a high transfection efficiency of approximately 82 ± 5% in MDA-MB-231 triple-negative breast cancer cells at an N/P ratio of 50:1. It is observed that mitochondrial membrane depolarization and intracellular reactive oxygen species generation are responsible for apoptosis and the higher number of apoptotic cells, which occurred through the arrest of the G2/M phase of the cell cycle were observed. Therefore, the synthesized light-responsive cationic polymer may be an effective system for diagnosis, with an efficient anticancer drug and gene carrier for the treatment of triple-negative breast cancer in the future.
Collapse
Affiliation(s)
- Rishik Patra
- Gene Therapy and Tissue Engineering Lab, Department of Polymer Science and Technology, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| | - Satyajit Halder
- Division of Molecular Medicine, Centenary Campus, Bose Institute, P-1/12 C.I.T. Scheme VII-M, Kolkata 700054, India
| | - Rima Saha
- Gene Therapy and Tissue Engineering Lab, Department of Polymer Science and Technology, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| | - Kuladip Jana
- Division of Molecular Medicine, Centenary Campus, Bose Institute, P-1/12 C.I.T. Scheme VII-M, Kolkata 700054, India
| | - Kishor Sarkar
- Gene Therapy and Tissue Engineering Lab, Department of Polymer Science and Technology, University of Calcutta, 92, A.P.C. Road, Kolkata 700009, India
| |
Collapse
|
10
|
Romanovska A, Schmidt M, Brandt V, Tophoven J, Tiller JC. Controlling the function of bioactive worm micelles by enzyme-cleavable non-covalent inter-assembly cross-linking. J Control Release 2024; 368:15-23. [PMID: 38346504 DOI: 10.1016/j.jconrel.2024.02.013] [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: 11/26/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/23/2024]
Abstract
Drugs that form self-assembled supramolecular structures to be most-active is a promising way of creating new highly specific and active pharmaceuticals. Controlling the activity of bioactive supramolecular structures such as drug-loaded micelles is possible by both core/shell and inter-assembly cross-linking. However, if the flexibility of the assembly is mandatory for the activity cross-linking is not feasible. Thus, such structures cannot be manipulated in their activity. The present study demonstrates a novel concept to control the activity of not drug-releasing, non-cross-linked bioactive superstructures. This is achieved by formation of nanostructured nanoparticles derived by non-covalent inter-assembly cross-linking of the superstructures. This is shown on the example of amphiphilic diblock-copolymers conjugated with the antibiotic ciprofloxacin (CIP). These polymer-antibiotic conjugates form worm micelles, which greatly activate the conjugated antibiotic without releasing it. Non-covalent inter-assembly cross-linking of these CIP-worm-micelles with amphiphilic triblock copolymers terminated with lipase-cleavable esters leads to nanostructured nanoparticles that resemble cross-linked worm micelles and show an up to 135-fold lower activity than the free worm micelles. The activity of the worm-micelles can be fully recovered by cleaving the end groups of the polymeric cross-linker with lipase.
Collapse
Affiliation(s)
- Alina Romanovska
- Biomaterials and Polymer Science, Department of Bio- and Chemical Engineering, TU Dortmund, Emil-Figge-Straße 66, 44227 Dortmund, Germany
| | - Martin Schmidt
- Biomaterials and Polymer Science, Department of Bio- and Chemical Engineering, TU Dortmund, Emil-Figge-Straße 66, 44227 Dortmund, Germany
| | - Volker Brandt
- Biomaterials and Polymer Science, Department of Bio- and Chemical Engineering, TU Dortmund, Emil-Figge-Straße 66, 44227 Dortmund, Germany
| | - Jonas Tophoven
- Biomaterials and Polymer Science, Department of Bio- and Chemical Engineering, TU Dortmund, Emil-Figge-Straße 66, 44227 Dortmund, Germany
| | - Joerg C Tiller
- Biomaterials and Polymer Science, Department of Bio- and Chemical Engineering, TU Dortmund, Emil-Figge-Straße 66, 44227 Dortmund, Germany.
| |
Collapse
|
11
|
Luo W, Zeng Y, Song Q, Wang Y, Yuan F, Li Q, Liu Y, Li S, Jannatun N, Zhang G, Li Y. Strengthening the Combinational Immunotherapy from Modulating the Tumor Inflammatory Environment via Hypoxia-Responsive Nanogels. Adv Healthc Mater 2024; 13:e2302865. [PMID: 38062634 DOI: 10.1002/adhm.202302865] [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: 08/28/2023] [Revised: 11/17/2023] [Indexed: 12/19/2023]
Abstract
Despite the success of immuno-oncology in clinical settings, the therapeutic efficacy is lower than the expectation due to the immunosuppressive inflammatory tumor microenvironment (TME) and the lack of functional lymphocytes caused by exhaustion. To enhance the efficacy of immuno-oncotherapy, a synergistic strategy should be used that can effectively improve the inflammatory TME and increase the tumor infiltration of cytotoxic T lymphocytes (CTLs). Herein, a TME hypoxia-responsive nanogel (NG) is developed to enhance the delivery and penetration of diacerein and (-)-epigallocatechin gallate (EGCG) in tumors. After systemic administration, diacerein effectively improves the tumor immunosuppressive condition through a reduction of MDSCs and Tregs in TME, and induces tumor cell apoptosis via the inhibition of IL-6/STAT3 signal pathway, realizing a strong antitumor effect. Additionally, EGCG can effectively inhibit the expression of PD-L1, restoring the tumor-killing function of CTLs. The infiltration of CTLs increases at the tumor site with activation of systemic immunity after the combination of TIM3 blockade therapy, ultimately resulting in a strong antitumor immune response. This study provides valuable insights for future research on eliciting effective antitumor immunity by suppressing adverse tumor inflammation. The feasible strategy proposed in this work may solve the urgent clinical concerns of the dissatisfactory checkpoint-based immuno-oncotherapy.
Collapse
Affiliation(s)
- Wenhe Luo
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Yanqiao Zeng
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Qingle Song
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Yu Wang
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Feng Yuan
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Qi Li
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Yingnan Liu
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Su Li
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Nahar Jannatun
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Guofang Zhang
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Yang Li
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
- Laboratory of Immunology and Nanomedicine & China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| |
Collapse
|
12
|
El-Gaby MSA, Abdel Reheim MAM, Akrim ZSM, Naguib BH, Saleh NM, El-Adasy ABAAM, El-Adl K, Mohamady S. 2-Thioxo-3,4-dihydropyrimidine and thiourea endowed with sulfonamide moieties as dual EGFR T790M and VEGFR-2 inhibitors: Design, synthesis, docking, and anticancer evaluations. Drug Dev Res 2024; 85:e22143. [PMID: 38349267 DOI: 10.1002/ddr.22143] [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: 10/03/2023] [Revised: 11/18/2023] [Accepted: 12/16/2023] [Indexed: 02/15/2024]
Abstract
The effectiveness of a new series of thiopyrimidine and thiourea containing sulfonamides moieties was tested on HCT-116, MCF-7, HepG2, and A549. HepG2 cell line was the one that all the new derivatives affected the most. The greatest potent compounds against the four HepG2, HCT116, MCF-7, and A549 cell lines were 8f and 8g with IC50 = 4.13, 6.64, 5.74, 6.85 µM and 4.09, 4.36, 4.22, 7.25 µM correspondingly. Compound 8g exhibited higher activity than sorafenib against HCT116 and MCF-7 but exhibited lower activity against HepG2 and A549. Moreover, compounds 8f and 8g exhibited higher activities than erlotinib on HepG2, HCT116, and MCF-7 but demonstrated lower activity on A549. The most potent cytotoxic derivatives 6f, 6g, 8c, 8d, 8e, 8f, and 8g were examined on normal VERO cell lines. Our derivatives have low toxicity on VERO cells with IC50 values ranging from 32.05 to 53.15 μM. Additionally, all compounds were assessed for dual VEGFR-2 and EGFRT790M inhibition effects. Compounds 8f and 8g were the most potent derivatives inhibited VEGFR-2 at IC50 value of 0.88 and 0.90 µM, correspondingly. As well, derivatives 8f and 8g could inhibit EGFRT790M demonstrating strongest effects with IC50 = 0.32 and 0.33 µM sequentially. Additionally, the greatest active derivatives ADMET profile was evaluated in relationship with sorafenib and erlotinib as reference agents. The data attained from docking were greatly related to that achieved from the biological testing.
Collapse
Affiliation(s)
- Mohamed S A El-Gaby
- Department of Chemistry, Faculty of Science, Al-Azhar University, Nasr City, Cairo, Egypt
| | | | - Zuhir S M Akrim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Omar Almukhtar University Al-Bayda, Libya
| | - Bassem H Naguib
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Nashwa M Saleh
- Department of Chemistry, Faculty of Science, Al-Azhar University (Girls Branch), Cairo, Egypt
| | | | - Khaled El-Adl
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt
| | - Samy Mohamady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| |
Collapse
|
13
|
Zhang J, Tang K, Liu Z, Zhang Z, Duan S, Wang H, Yang H, Yang D, Fan W. Tumor microenvironment-responsive degradable silica nanoparticles: design principles and precision theranostic applications. NANOSCALE HORIZONS 2024; 9:186-214. [PMID: 38164973 DOI: 10.1039/d3nh00388d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Silica nanoparticles have emerged as promising candidates in the field of nanomedicine due to their remarkable versatility and customizable properties. However, concerns about their potential toxicity in healthy tissues and organs have hindered their widespread clinical translation. To address this challenge, significant attention has been directed toward a specific subset of silica nanoparticles, namely degradable silica nanoparticles, primarily because of their excellent biocompatibility and responsive biodegradability. In this review, we provide a comprehensive understanding of degradable silica nanoparticles, categorizing them into two distinct groups: inorganic species-doped and organic moiety-doped silica nanoparticles based on their framework components. Next, the recent progress of tumor microenvironment (TME)-responsive degradable silica nanoparticles for precision theranostic applications is summarized in detail. Finally, current bottlenecks and future opportunities of theranostic nanomedicines based on degradable silica nanoparticles in clinical applications are also outlined and discussed. The aim of this comprehensive review is to shed light on the potential of degradable silica nanoparticles in addressing current challenges in nanomedicine, offering insights into their design, applications in tumor diagnosis and treatment, and paving the way for future advancements in clinical theranostic nanomedicines.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu 233030, P. R. China.
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, P. R. China
| | - Kaiyuan Tang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu 233030, P. R. China.
| | - Zilu Liu
- School of Fundamental Sciences, Bengbu Medical College, Bengbu 233030, P. R. China.
| | - Zhijing Zhang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu 233030, P. R. China.
| | - Shufan Duan
- School of Fundamental Sciences, Bengbu Medical College, Bengbu 233030, P. R. China.
| | - Hui Wang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu 233030, P. R. China.
| | - Hui Yang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu 233030, P. R. China.
| | - Dongliang Yang
- Nanjing Polytechnic Institute, Nanjing 210048, P. R. China.
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, P. R. China.
| |
Collapse
|
14
|
Liu D, Liang M, Fan A, Bing W, Qi J. Hypoxia-responsive AIEgens for precise disease theranostics. LUMINESCENCE 2024; 39:e4659. [PMID: 38286609 DOI: 10.1002/bio.4659] [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: 09/15/2023] [Revised: 11/01/2023] [Accepted: 12/04/2023] [Indexed: 01/31/2024]
Abstract
Specific biomarker-activatable probes have revolutionized theranostics, being beneficial for precision medicine. Hypoxia is a critical pathological characteristic prevalent in numerous major diseases such as cancers, cardiovascular disorders, inflammatory diseases, and acute ischemia. Aggregation-induced emission luminogens (AIEgens) have emerged as a promising tool to tackle the biomedical issues. Of particular significance are the hypoxia-responsive AIEgens, representing a kind of crucial probe capable of delicately sensing and responding to the hypoxic microenvironment, thereby enhancing the precision of disease diagnosis and treatment. In this review, we summarize the recent advances of hypoxia-responsive AIEgens for varied biomedical applications. The hypoxia-responsive structures based on AIEgens, such as azobenzene, nitrobenzene, and N-oxide are presented, which are in response to the reduction property to bring about significant alternations in response spectra and/or fluorescence intensity. The bioapplications including imaging and therapy of tumor and ischemia diseases are discussed. Moreover, the review sheds light on the future challenges and prospects in this field. This review aims to provide comprehensive guidance and understanding into the development of activatable bioprobes, especially the hypoxia-responsive AIEgens for improving the diagnosis and therapy outcome of related diseases.
Collapse
Affiliation(s)
- Dongfang Liu
- Key Laboratory of Bionic Engineering, Ministry of Education, Jilin University, Changchun, China
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, China
| | - Mengyun Liang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| | - Aohua Fan
- Key Laboratory of Bionic Engineering, Ministry of Education, Jilin University, Changchun, China
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, China
| | - Wei Bing
- Key Laboratory of Bionic Engineering, Ministry of Education, Jilin University, Changchun, China
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, China
| | - Ji Qi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
15
|
Anwer KE, El-Hddad SSA, Abd El-Sattar NEA, El-Morsy A, Khedr F, Mohamady S, Keshek DE, Salama SA, El-Adl K, Hanafy NS. Five and six membered heterocyclic rings endowed with azobenzene as dual EGFR T790M and VEGFR-2 inhibitors: design, synthesis, in silico ADMET profile, molecular docking, dynamic simulation and anticancer evaluations. RSC Adv 2023; 13:35321-35338. [PMID: 38053688 PMCID: PMC10695193 DOI: 10.1039/d3ra06614b] [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: 09/28/2023] [Accepted: 11/28/2023] [Indexed: 12/07/2023] Open
Abstract
Novel azobenzene scaffold-joined heterocyclic isoxazole, pyrazole, triazole, and/or triazine moieties have been developed and synthesized utilizing microwave and traditional methods. Our compounds were tested for growth inhibition of A549, MCF-7, HCT-116, and HepG2 tumors by dual targeting the VEGFR-2 and EGFRT790M enzymes. The suggested compound's manner of binding with EGFRT790M and VEGFR-2 active sites was explored through molecular design and MD modeling. The information from the results of the biological screening and the docking studies was highly correlated. The A549 cell line was the one that responded to the novel compound's effects most effectively. Having IC50 values of 5.15, 6.37, 8.44 and 6.23 μM, respectively, 14 was the most effective derivative on the four A549, MCF-7, HCT116 and HepG2 cancer cells. It had greater activity than erlotinib and slightly inferior activities on the tested cell lines than sorafenib, respectively. The cytotoxicity of the most effective derivatives, 5, 6, 10 and 14, was evaluated against typical VERO cell lines. Having IC50 values ranging from 42.32 to 55.20 μM, the results showed that the investigated drugs have modest toxicity against VERO normal cells. Additionally all derivatives were assessed for their dual VEGFR-2 and EGFRT790M inhibitory effects. Among them, derivatives 14, 5 and 10 were established as the greatest inhibitors of VEGFR-2 at IC50 values of 0.95, 1.25 and 1.50 μM correspondingly. As well, derivatives 14, 6, 5 and 10 could inhibit EGFRT790M activity demonstrating strongest effects with IC50 = 0.25, 0.35, 0.40 and 0.50 μM respectively. Furthermore, the ADMET profile was evaluated for compounds 5, 6, 10 and 14 in contrast to reference drugs sorafenib and erlotinib.
Collapse
Affiliation(s)
- Kurls E Anwer
- Department of Chemistry, Faculty of Science, Ain Shams University Abbassia Cairo Egypt
| | | | - Nour E A Abd El-Sattar
- Department of Chemistry, Faculty of Science, Ain Shams University Abbassia Cairo Egypt
- Basic & Medical Sciences Department, Faculty of Dentistry, Alryada University for Science & Technology Egypt
| | - Ahmed El-Morsy
- Pharmaceutical Chemistry Department, College of Pharmacy, The Islamic University Najaf Iraq
| | - Fathalla Khedr
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University Nasr City 11884 Cairo Egypt
| | - Samy Mohamady
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, The British University in Egypt Cairo Egypt
| | - Doaa E Keshek
- Department of Biology, Jumum College University, Umm Al-Qura University P.O. Box 7388 Makkah 21955 Sudia Arabia
- Agriculture Genetic Engineering Research Institute (AGERI), Agriculture Research Centre Giza Egypt
| | - Samir A Salama
- Division of Biochemistry, Department of Pharmacology, College of Pharmacy, Taif University P.O. Box 11099 Taif 21944 Kingdom of Saudi Arabia
| | - Khaled El-Adl
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University Nasr City 11884 Cairo Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Heliopolis University for Sustainable Development Cairo Egypt
| | - Noura S Hanafy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Heliopolis University for Sustainable Development Cairo Egypt
| |
Collapse
|
16
|
Zhang R, Yu J, Guo Z, Jiang H, Wang C. Camptothecin-based prodrug nanomedicines for cancer therapy. NANOSCALE 2023; 15:17658-17697. [PMID: 37909755 DOI: 10.1039/d3nr04147f] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Camptothecin (CPT) is a cytotoxic alkaloid that attenuates the replication of cancer cells via blocking DNA topoisomerase 1. Despite its encouraging and wide-spectrum antitumour activity, its application is significantly restricted owing to its instability, low solubility, significant toxicity, and acquired tumour cell resistance. This has resulted in the development of many CPT-based therapeutic agents, especially CPT-based nanomedicines, with improved pharmacokinetic and pharmacodynamic profiles. Specifically, smart CPT-based prodrug nanomedicines with stimuli-responsive release capacity have been extensively explored owing to the advantages such as high drug loading, improved stability, and decreased potential toxicity caused by the carrier materials in comparison with normal nanodrugs and traditional delivery systems. In this review, the potential strategies and applications of CPT-based nanoprodrugs for enhanced CPT delivery toward cancer cells are summarized. We appraise in detail the chemical structures and release mechanisms of these nanoprodrugs and guide materials chemists to develop more powerful nanomedicines that have real clinical therapeutic capacities.
Collapse
Affiliation(s)
- Renshuai Zhang
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
| | - Jing Yu
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao Municipal Hospital, Qingdao, 266071, China
| | - Zhu Guo
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
- The Affiliated Hospital of Qingdao University, Qingdao 266061, China
| | - Hongfei Jiang
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
| | - Chao Wang
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
| |
Collapse
|
17
|
Chen S, Zhu J. Probing Near-infrared Absorbance of E and Z Diazene Isomers via Antiaromaticity. J Org Chem 2023; 88:12183-12193. [PMID: 37579502 DOI: 10.1021/acs.joc.3c00010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
The photoswitching behaviors of heteroaryl azos and azobenzenes have attracted considerable interest due to their applications from material science to pharmacology. However, the use of UV light limits their application, especially in biomedicine and photopharmacology. In this work, using several aromaticity descriptors, including anisotropy of the induced current density analysis and nucleus-independent chemical shifts, we systematically investigate the relationship between anti-aromaticity and the absorption of a series of heterocyclic azos. We have demonstrated that the antiaromatic heterocycles substituted with diazenes enable the significant red shifts of the n → π* and π → π* transition bands of E and Z isomers via density functional theory calculations. Moreover, introducing substituents into heterocycles could further tune the absorption. Finally, the λmax of the first transition bands of the E (ca. 1026 nm) and Z isomers (ca. 1167 nm) of azos is achieved in the near-infrared region.
Collapse
Affiliation(s)
- Shuwen Chen
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry and Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jun Zhu
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry and Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
18
|
Li D, Wang X, Han K, Sun Y, Ren T, Sun G, Zhang N, Zhao L, Zhong R. Hypoxia and CD44 receptors dual-targeted nano-micelles with AGT-inhibitory activity for the targeting delivery of carmustine. Int J Biol Macromol 2023; 246:125657. [PMID: 37399878 DOI: 10.1016/j.ijbiomac.2023.125657] [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: 11/26/2022] [Revised: 02/17/2023] [Accepted: 06/30/2023] [Indexed: 07/05/2023]
Abstract
Carmustine (BCNU) is a typical chemotherapy used for treatment of cerebroma and other solid tumors, which exerts antitumor effect by inducing DNA damage at O6 position of guanine. However, the clinical application of BCNU was extremely limited due to the drug resistance mainly mediated by O6-alkylguanine-DNA alkyltransferase (AGT) and absence of tumor-targeting ability. To overcome these limitations, we developed a hypoxia-responsive nanomicelle with AGT inhibitory activity, which was successfully loaded with BCNU. In this nano-system, hyaluronic acid (HA) acts as an active tumor-targeting ligand to bind the overexpressing CD44 receptors on the surface of tumor cells. An azo bond selectively breaks in hypoxic tumor microenvironment to release O6-benzylguanine (BG) as AGT inhibitor and BCNU as DNA alkylating agent. The obtained HA-AZO-BG NPs with shell core structure had an average particle size of 176.98 ± 11.19 nm and exhibited good stability. Meanwhile, HA-AZO-BG NPs possessed a hypoxia-responsive drug release profile. After immobilizing BCNU into HA-AZO-BG NPs, the obtained HA-AZO-BG/BCNU NPs exhibited obvious hypoxia-selectivity and superior cytotoxicity in T98G, A549, MCF-7 and SMMC-7721 cells with IC50 at 189.0, 183.2, 90.1 and 100.1 μm, respectively, under hypoxic condition. Near-infrared imaging in HeLa tumor xenograft models showed that HA-AZO-BG/DiR NPs could effectively accumulate in tumor site at 4 h of post-injection, suggesting its good tumor-targetability. In addition, in vivo anti-tumor efficacy and toxicity evaluation indicated that HA-AZO-BG/BCNU NPs was more effective and less harmful compared to the other groups. After treatment, the tumor weight of HA-AZO-BG/BCNU NPs group was 58.46 % and 63.33 % of the control group and BCNU group, respectively. Overall, HA-AZO-BG/BCNU NPs was expected to be a promising candidate for targeted delivery of BCNU and elimination of chemoresistance.
Collapse
Affiliation(s)
- Duo Li
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China
| | - Xiaoli Wang
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China
| | - Kaishuo Han
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China
| | - Yaqian Sun
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China
| | - Ting Ren
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China
| | - Guohui Sun
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China
| | - Na Zhang
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China
| | - Lijiao Zhao
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China.
| | - Rugang Zhong
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
19
|
Li Y, Feng M, Guo T, Wang Z, Zhao Y. Tailored Beta-Lapachone Nanomedicines for Cancer-Specific Therapy. Adv Healthc Mater 2023; 12:e2300349. [PMID: 36970948 DOI: 10.1002/adhm.202300349] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/16/2023] [Indexed: 03/29/2023]
Abstract
Nanotechnology shows the power to improve efficacy and reduce the adverse effects of anticancer agents. As a quinone-containing compound, beta-lapachone (LAP) is widely employed for targeted anticancer therapy under hypoxia. The principal mechanism of LAP-mediated cytotoxicity is believed due to the continuous generation of reactive oxygen species with the aid of NAD(P)H: quinone oxidoreductase 1 (NQO1). The cancer selectivity of LAP relies on the difference between NQO1 expression in tumors and that in healthy organs. Despite this, the clinical translation of LAP faces the problem of narrow therapeutic window that is challenging for dose regimen design. Herein, the multifaceted anticancer mechanism of LAP is briefly introduced, the advance of nanocarriers for LAP delivery is reviewed, and the combinational delivery approaches to enhance LAP potency in recent years are summarized. The mechanisms by which nanosystems boost LAP efficacy, including tumor targeting, cellular uptake enhancement, controlled cargo release, enhanced Fenton or Fenton-like reaction, and multidrug synergism, are also presented. The problems of LAP anticancer nanomedicines and the prospective solutions are discussed. The current review may help to unlock the potential of cancer-specific LAP therapy and speed up its clinical translation.
Collapse
Affiliation(s)
- Yaru Li
- School of Pharmaceutical Science and Technology, Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin, 300072, China
| | - Meiyu Feng
- School of Pharmaceutical Science and Technology, Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin, 300072, China
| | - Tao Guo
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, 300120, China
| | - Zheng Wang
- School of Pharmaceutical Science and Technology, Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin, 300072, China
| | - Yanjun Zhao
- School of Pharmaceutical Science and Technology, Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin, 300072, China
| |
Collapse
|
20
|
Zhao H, Yu J, Zhang R, Chen P, Jiang H, Yu W. Doxorubicin prodrug-based nanomedicines for the treatment of cancer. Eur J Med Chem 2023; 258:115612. [PMID: 37441851 DOI: 10.1016/j.ejmech.2023.115612] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023]
Abstract
The chemotherapeutic drug of doxorubicin (DOX) has witnessed widespread applications for treating various cancers. DOX-treated dying cells bear cellular modifications which allow enhanced presentation of tumor antigen and neighboring dendritic cell activation. Furthermore, DOX also facilitate the immune-mediated clearance of tumor cells. However, disadvantages such as severe off-target toxicity, and prominent hydrophobicity have resulted in unsatisfactory clinical therapeutic outcomes. The effective delivery of DOX drug molecules is still challenging despite the rapid advances in nanotechnology and biomaterials. Huge progress has been witnessed in DOX nanoprodrugs owing to their brilliant benefits such as tumor stimuli-responsive drug release capacity, high drug loading efficiency and so on. This review summarized recent progresses of DOX prodrug-based nanomedicines to provide deep insights into future development and inspire researchers to explore DOX nanoprodrugs with real clinical applications.
Collapse
Affiliation(s)
- Haibo Zhao
- Cancer Institute of the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Jing Yu
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao Municipal Hospital, Qingdao, 266071, China
| | - Renshuai Zhang
- Cancer Institute of the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Pengwei Chen
- Hainan Key Laboratory for Research and Development of Natural Product from Li Folk Medicine, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
| | - Hongfei Jiang
- Cancer Institute of the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Wanpeng Yu
- Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
21
|
Shamsipur M, Ghavidast A, Pashabadi A. Phototriggered structures: Latest advances in biomedical applications. Acta Pharm Sin B 2023; 13:2844-2876. [PMID: 37521863 PMCID: PMC10372844 DOI: 10.1016/j.apsb.2023.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 03/12/2023] [Accepted: 04/11/2023] [Indexed: 08/01/2023] Open
Abstract
Non-invasive control of the drug molecules accessibility is a key issue in improving diagnostic and therapeutic procedures. Some studies have explored the spatiotemporal control by light as a peripheral stimulus. Phototriggered drug delivery systems (PTDDSs) have received interest in the past decade among biological researchers due to their capability the control drug release. To this end, a wide range of phototrigger molecular structures participated in the DDSs to serve additional efficiency and a high-conversion release of active fragments under light irradiation. Up to now, several categories of PTDDSs have been extended to upgrade the performance of controlled delivery of therapeutic agents based on well-known phototrigger molecular structures like o-nitrobenzyl, coumarinyl, anthracenyl, quinolinyl, o-hydroxycinnamate and hydroxyphenacyl, where either of one endows an exclusive feature and distinct mechanistic approach. This review conveys the design, photochemical properties and essential mechanism of the most important phototriggered structures for the release of single and dual (similar or different) active molecules that have the ability to quickly reason of the large variety of dynamic biological phenomena for biomedical applications like photo-regulated drug release, synergistic outcomes, real-time monitoring, and biocompatibility potential.
Collapse
|
22
|
Huang R, Fan D, Cheng H, Huo J, Wang S, He H, Zhang G. Multi-Site Attack, Neutrophil Membrane-Camouflaged Nanomedicine with High Drug Loading for Enhanced Cancer Therapy and Metastasis Inhibition. Int J Nanomedicine 2023; 18:3359-3375. [PMID: 37361388 PMCID: PMC10290460 DOI: 10.2147/ijn.s415139] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023] Open
Abstract
Background Advanced breast cancer is a highly metastatic tumor with high mortality. Simultaneous elimination of primary tumor and inhibition of neutrophil-circulation tumor cells (CTCs) cluster formation are urgent issues for cancer therapy. Unfortunately, the drug delivery efficiency to tumors and anti-metastasis efficacy of nanomedicine are far from satisfactory. Methods To address these problems, we designed a multi-site attack, neutrophil membrane-camouflaged nanoplatform encapsulating hypoxia-responsive dimeric prodrug hQ-MMAE2 (hQNM-PLGA) for enhanced cancer and anti-metastasis therapy. Results Encouraged by the natural tendency of neutrophils to inflammatory tumor sites, hQNM-PLGA nanoparticles (NPs) could target delivery of drug to tumor, and the acute hypoxic environment of advanced 4T1 breast tumor promoted hQ-MMAE2 degradation to release MMAE, thus eliminating the primary tumor cells to achieve remarkable anticancer efficacy. Alternatively, NM-PLGA NPs inherited the similar adhesion proteins of neutrophils so that NPs could compete with neutrophils to interrupt the formation of neutrophil-CTC clusters, leading to a reduction in extravasation of CTCs and inhibition of tumor metastasis. The in vivo results further revealed that hQNM-PLGA NPs possessed a perfect safety and ability to inhibit tumor growth and spontaneous lung metastasis. Conclusion This study demonstrates the multi-site attack strategy provides a prospective avenue with the potential to improve anticancer and anti-metastasis therapeutic efficacy.
Collapse
Affiliation(s)
- Ran Huang
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, 450046, People’s Republic of China
| | - Daopeng Fan
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, 450046, People’s Republic of China
| | - Hanghang Cheng
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, 450046, People’s Republic of China
| | - Jian Huo
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, 450046, People’s Republic of China
| | - Shuqi Wang
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, 450046, People’s Republic of China
| | - Hua He
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, 450046, People’s Republic of China
| | - Gaiping Zhang
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, 450046, People’s Republic of China
- Longhu Laboratory, Zhengzhou, 450046, People’s Republic of China
| |
Collapse
|
23
|
Li JJ, Rong RX, Yang Y, Hu ZY, Hu B, Zhao YY, Li HB, Hu XY, Wang KR, Guo DS. Triple targeting host-guest drug delivery system based on lactose-modified azocalix[4]arene for tumor ablation. MATERIALS HORIZONS 2023; 10:1689-1696. [PMID: 36825769 DOI: 10.1039/d3mh00018d] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Host-guest drug delivery systems (HGDDSs) have been studied in an effort to modify the characteristics of therapeutic agents through noncovalent interactions, reduce toxic side effects and improve therapeutic effects. However, it is still an important task to continuously improve the targeting ability of HGDDSs, which is conducive to the development of precision medicine. Herein, we utilize the lactose-modified azocalix[4]arene (LacAC4A) as a triple targeting drug carrier customized for antitumor purposes. LacAC4A integrates three targeting features, passive targeting through the enhancing permeability and retention effect, active targeting by the interactions of lactose and the asialoglycoprotein receptors on the surface of tumor cells, and stimuli-responsive targeting via the reduction of the azo group under a hypoxia microenvironment. After loading doxorubicin (DOX) in LacAC4A, the supramolecular nanoformulation DOX@LacAC4A clearly showed the effective suppression of tumor growth through in vivo experiments. LacAC4A can achieve effective targeting, rapid release, and improve drug bioavailability. This design principle will provide a new material for drug delivery systems.
Collapse
Affiliation(s)
- Juan-Juan Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China.
| | - Rui-Xue Rong
- Department of Medical Microbiology and Immunology, School of Basic Medical Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Medical Comprehensive Experimental Center, Hebei University, Baoding 071002, China
| | - Yan Yang
- Department of Medical Microbiology and Immunology, School of Basic Medical Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Medical Comprehensive Experimental Center, Hebei University, Baoding 071002, China
| | - Zong-Ying Hu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China.
| | - Bing Hu
- College of Chemistry and Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding 071002, China.
| | - Ying-Ying Zhao
- College of Chemistry and Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding 071002, China.
| | - Hua-Bin Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China.
| | - Xin-Yue Hu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China.
| | - Ke-Rang Wang
- College of Chemistry and Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding 071002, China.
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China.
| |
Collapse
|
24
|
Albanese P, Cataldini S, Ren CZJ, Valletti N, Brunetti J, Chen JLY, Rossi F. Light-Switchable Membrane Permeability in Giant Unilamellar Vesicles. Pharmaceutics 2022; 14:2777. [PMID: 36559270 PMCID: PMC9780837 DOI: 10.3390/pharmaceutics14122777] [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: 10/31/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
In this work, giant unilamellar vesicles (GUVs) were synthesized by blending the natural phospholipid 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) with a photoswitchable amphiphile (1) that undergoes photoisomerization upon irradiation with UV-A (E to Z) and blue (Z to E) light. The mixed vesicles showed marked changes in behavior in response to UV light, including changes in morphology and the opening of pores. The fine control of membrane permeability with consequent cargo release could be attained by modulating either the UV irradiation intensity or the membrane composition. As a proof of concept, the photocontrolled release of sucrose from mixed GUVs is demonstrated using microscopy (phase contrast) and confocal studies. The permeability of the GUVs to sucrose could be increased to ~4 × 10-2 μm/s when the system was illuminated by UV light. With respect to previously reported systems (entirely composed of synthetic amphiphiles), our findings demonstrate the potential of photosensitive GUVs that are mainly composed of natural lipids to be used in medical and biomedical applications, such as targeted drug delivery and localized topical treatments.
Collapse
Affiliation(s)
- Paola Albanese
- Department of Earth, Environmental & Physical Sciences, University of Siena, Pian Mantellini 44, 53100 Siena, Italy
| | - Simone Cataldini
- Department of Biotechnology, Chemistry and Pharmaceutical Sciences, University of Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Chloe Z-J Ren
- Centre for Biomedical and Chemical Sciences, School of Science, Auckland University of Technology, Auckland 1142, New Zealand
| | - Nadia Valletti
- Department of Earth, Environmental & Physical Sciences, University of Siena, Pian Mantellini 44, 53100 Siena, Italy
| | - Jlenia Brunetti
- MedBiotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Jack L-Y Chen
- Department of Biotechnology, Chemistry and Pharmaceutical Sciences, University of Siena, Via Aldo Moro, 53100 Siena, Italy
- Centre for Biomedical and Chemical Sciences, School of Science, Auckland University of Technology, Auckland 1142, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Victoria University of Wellington, Wellington 6140, New Zealand
| | - Federico Rossi
- Department of Earth, Environmental & Physical Sciences, University of Siena, Pian Mantellini 44, 53100 Siena, Italy
| |
Collapse
|
25
|
Shen X, Zhang C, Lan F, Su Z, Zheng Y, Zheng T, Xiong Q, Xie X, Du G, Zhao X, Hu C, Deng P, Yu Z. Dibenzo[
b
,
f
][1,4,5]chalcogenadiazepine Photoswitches: Conversion of Excitation Energy into Ring Strain. Angew Chem Int Ed Engl 2022; 61:e202209441. [DOI: 10.1002/anie.202209441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Xin Shen
- Key Laboratory of Green Chemistry & Technology of Ministry of Education College of Chemistry Sichuan University 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Cefei Zhang
- Key Laboratory of Green Chemistry & Technology of Ministry of Education College of Chemistry Sichuan University 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Fengying Lan
- Key Laboratory of Green Chemistry & Technology of Ministry of Education College of Chemistry Sichuan University 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Zhishan Su
- Key Laboratory of Green Chemistry & Technology of Ministry of Education College of Chemistry Sichuan University 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Yuanqin Zheng
- Key Laboratory of Green Chemistry & Technology of Ministry of Education College of Chemistry Sichuan University 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Tingting Zheng
- Key Laboratory of Green Chemistry & Technology of Ministry of Education College of Chemistry Sichuan University 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Qin Xiong
- Key Laboratory of Green Chemistry & Technology of Ministry of Education College of Chemistry Sichuan University 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Xinyu Xie
- Key Laboratory of Green Chemistry & Technology of Ministry of Education College of Chemistry Sichuan University 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Guangxi Du
- Key Laboratory of Green Chemistry & Technology of Ministry of Education College of Chemistry Sichuan University 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Xiaohu Zhao
- Key Laboratory of Green Chemistry & Technology of Ministry of Education College of Chemistry Sichuan University 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Changwei Hu
- Key Laboratory of Green Chemistry & Technology of Ministry of Education College of Chemistry Sichuan University 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Pengchi Deng
- Analytical & Testing Center Sichuan University 29 Wangjiang Road Chengdu 610064 P. R. China
| | - Zhipeng Yu
- Key Laboratory of Green Chemistry & Technology of Ministry of Education College of Chemistry Sichuan University 29 Wangjiang Road Chengdu 610064 P. R. China
| |
Collapse
|
26
|
Tian H, Zhang T, Qin S, Huang Z, Zhou L, Shi J, Nice EC, Xie N, Huang C, Shen Z. Enhancing the therapeutic efficacy of nanoparticles for cancer treatment using versatile targeted strategies. J Hematol Oncol 2022; 15:132. [PMID: 36096856 PMCID: PMC9469622 DOI: 10.1186/s13045-022-01320-5] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Poor targeting of therapeutics leading to severe adverse effects on normal tissues is considered one of the obstacles in cancer therapy. To help overcome this, nanoscale drug delivery systems have provided an alternative avenue for improving the therapeutic potential of various agents and bioactive molecules through the enhanced permeability and retention (EPR) effect. Nanosystems with cancer-targeted ligands can achieve effective delivery to the tumor cells utilizing cell surface-specific receptors, the tumor vasculature and antigens with high accuracy and affinity. Additionally, stimuli-responsive nanoplatforms have also been considered as a promising and effective targeting strategy against tumors, as these nanoplatforms maintain their stealth feature under normal conditions, but upon homing in on cancerous lesions or their microenvironment, are responsive and release their cargoes. In this review, we comprehensively summarize the field of active targeting drug delivery systems and a number of stimuli-responsive release studies in the context of emerging nanoplatform development, and also discuss how this knowledge can contribute to further improvements in clinical practice.
Collapse
Affiliation(s)
- Hailong Tian
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Tingting Zhang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jiayan Shi
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, 3800, VIC, Australia
| | - Edouard C Nice
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China
| | - Na Xie
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China. .,West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China.
| | - Canhua Huang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.
| |
Collapse
|
27
|
Wang X, Sun B, Ye Z, Zhang W, Xu W, Gao S, Zhou N, Wu F, Shen J. Enzyme-Responsive COF-Based Thiol-Targeting Nanoinhibitor for Curing Bacterial Infections. ACS APPLIED MATERIALS & INTERFACES 2022; 14:38483-38496. [PMID: 35989491 DOI: 10.1021/acsami.2c08845] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Pathogen infections impose severe challenges in clinical practice, especially for patients infected with antibiotic-resistant microbes. The thioredoxin (Trx) system in Gram-positive bacteria serves as an ideal antimicrobial target for novel medicine design due to the structural differences from corresponding system in mammals. However, a backup thiol-dependent antioxidant glutathione (GSH) system limits the effectiveness of drugs in many Gram-negative bacteria. Herein, we synthesize a thiol-targeting nanoinhibitor based on an enzyme-responsive covalent organic framework (COF) coloaded with silver nanoparticles (AgNPs) and ebselen (EBS) (Ag-TA-CON@EBS@PEG) to exert synergistic antibacterial effects. Since azoreductase can dissociate the enzyme-responsive COF, we adopt this strategy to achieve the accurate release of EBS and Ag+ at infection sites. Our research identifies that the functionalized nanoinhibitor shows excellent bactericidal performance for Gram-positive and Gram-negative bacteria in vitro and exhibits low toxicity to normal cells. Besides, the nanoinhibitor presents favorable biocompatibility, anti-inflammatory property, and effective wound healing ability in mice. This paper provides a promising clinical strategy for synergistic antibacterial therapy and enhanced wound healing properties via an optimized combination of the targeted nanomedicines with an intelligent drug conveying platform.
Collapse
Affiliation(s)
- Xinye Wang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Baohong Sun
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Ziqiu Ye
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Wenjia Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Wang Xu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Shurui Gao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Ninglin Zhou
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Fan Wu
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|
28
|
Shen X, Zhang C, Lan F, Su Z, Zheng Y, Zheng T, Xiong Q, Xie X, Du G, Zhao X, Hu C, Deng P, Yu Z. Dibenzo[b,f][1,4,5]chalcogenadiazepine Photoswitches: Conversion of Excitation Energy into Ring Strain. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202209441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Xin Shen
- Sichuan University Department of Chemistry 610000 Chengdu CHINA
| | - Cefei Zhang
- Sichuan University College of Chemistry CHINA
| | - Fengying Lan
- Sichuan University Department of Chemistry CHINA
| | - Zhishan Su
- Sichuan University College of Chemistry CHINA
| | | | | | - Qin Xiong
- Sichuan University Department of Chemistry CHINA
| | - Xinyu Xie
- Sichuan University Department of Chemistry CHINA
| | - Guangxi Du
- Sichuan University Department of Chemistry CHINA
| | - Xiaohu Zhao
- Sichuan University Department of Chemistry CHINA
| | - Changwei Hu
- Sichuan University College of Chemistry CHINA
| | - Pengchi Deng
- Sichuan University Analytical & Testing Center CHINA
| | - Zhipeng Yu
- Sichuan University - Wangjiang Campus: Sichuan University College of Chemistry College of Chemistry29 Wangjianglu, Jiuyanqiao 610064 Chengdu CHINA
| |
Collapse
|