1
|
Liu Y, Zong Q, Tu Y, Zhang X, Tan Q, Ullah I, Yuan Y. A tumor heterogeneity-independent antigen-responsive nanocarrier enabled by bioorthogonal pre-targeting and click-activated self-immolative polymer. Biomaterials 2025; 319:123200. [PMID: 39987854 DOI: 10.1016/j.biomaterials.2025.123200] [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: 12/05/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
Bioorthogonal pre-targeting alleviate the limitations of traditional nanomedicines in passive and active targeting delivery. However, the high selectivity of bioorthogonal pre-targeting depends on the high expression level of antigens in lesion sites, and there are very limited targets with sufficient overexpression. Herein, we propose a tumor heterogeneity-independent antigen-responsive nanocarrier utilizing bioorthogonal pre-targeting and click-activated self-immolative polymers for stimulus signal conversion and amplification. This approach comprises a tetrazine (Tz) conjugated with trastuzumab (T-Tz), and a bioorthogonally activatable nanocarrier CONP which self-assembled by isocyanide and polyethylene glycol-modified poly (thiocarbamate) (NC-PTC-PEG) and hydrogen sulfide (H2S)-responsive self-immolative polymers. In practice, T-Tz is first injected to actively pretarget HER2-positive tumor cells and followed by the second injection of nanocarrier CONP. The NC-PTC-PEG in CONP undergoes a click reaction with Tz to generate H2S, thereby achieving the transformation from antigen signal to H2S signal. Finally, NO2-PTC-PEG responds to H2S stimulation and undergoes a head-to-tail depolymerization process similar to dominoes to produce a large amount of H2S, further amplifying the stimulus signal. This bioorthogonal pre-targeting combine with click-activated self-immolative polymers is anticipated to enhance the effectiveness of existing pre-targeting strategies for tumor imaging and therapy, with the potential to overcome challenges posed by tumor heterogeneity.
Collapse
Affiliation(s)
- Ye Liu
- Department of Radiology, The Second Affiliated Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China
| | - Qingyu Zong
- Department of Radiology, The Second Affiliated Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China
| | - Yalan Tu
- Scientific Research and Teaching Department, Public Health Clinical Center of Chengdu, Sichuan, 610061, PR China.
| | - Xingzu Zhang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 511442, PR China
| | - Qiaoling Tan
- Department of Radiology, The Second Affiliated Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China
| | - Ihsan Ullah
- Department of Radiology, The Second Affiliated Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China
| | - Youyong Yuan
- Department of Radiology, The Second Affiliated Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, PR China.
| |
Collapse
|
2
|
Yin W, Chen X, Bai L, Li Y, Chen W, Jiang Y, He Y, Yang Y, Lin Y, Tian T, Cai X. BBPs-functionalized tetrahedral framework nucleic acid hydrogel scaffold captures endogenous BMP-2 to promote bone regeneration. Biomaterials 2025; 319:123194. [PMID: 39970843 DOI: 10.1016/j.biomaterials.2025.123194] [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: 10/22/2024] [Revised: 02/03/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Bone Morphogenetic Protein-2 (BMP-2) is a key growth factor for inducing osteogenic differentiation and promoting bone remodeling. However, the exogenous application of delivery systems for BMP-2 has been hampered by various postoperative complications, poor stability and high price. Hence, in situ enrichment of endogenous BMP-2 is promising. The discovery of a small molecule BMP-2 binding peptide (BBP) that binds specifically to BMP-2 with high affinity lays the foundation for the construction of bioactive materials that capture endogenous BMP-2. In contrast, conventional enrichment strategies have low binding efficiency due to steric hindrance caused by the disordered arrangement of BBPs. Tetrahedral framework nucleic acid (tFNA) exhibits good editability and unique three-dimensional spatial structure that enables topological control of multivalent ligands in spatial distribution. The BBPs are further designed to be stably modified on tFNA (BBPs-tFNA) via click chemistry of the azide-alkyne addition to achieve the orderly arrangement of BBPs in spatial organization, to improve the binding efficiency of BMP-2. Therefore, in this study, BBPs-tFNA is modified on biocompatible hyaluronic acid methacryloyl (HAMA) to construct the functionalized bioactive composite hydrogel scaffolds, with the aim of achieving precise and efficient capture of endogenous BMP-2, stimulating osteogenic differentiation and promoting in situ osteogenesis for bone defect repair.
Collapse
Affiliation(s)
- Wumeng Yin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xingyu Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Long Bai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yong Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wen Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yueying Jiang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yutian He
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yichen Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Taoran Tian
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China.
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
3
|
Zhang W, Wang S, Xing Y, Luo X, Wang R, Yu F. Bioorthogonal SERS-bioluminescence dual-modal imaging for real-time tracking of triple-negative breast cancer metastasis. Acta Biomater 2025; 197:431-443. [PMID: 40101869 DOI: 10.1016/j.actbio.2025.03.019] [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: 11/19/2024] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 03/20/2025]
Abstract
Triple-negative breast cancer (TNBC) represents an aggressive subtype of breast cancer, characterized by early metastasis and a poor prognosis. Traditional imaging modalities often lack the sensitivity and molecular specificity required for the early detection of metastatic lesions. In this study, we developed a dual-modal imaging strategy that integrates surface-enhanced Raman scattering (SERS) and bioluminescence imaging probes, utilizing bioorthogonal labeling to track TNBC organ metastasis. The SERS probes were encapsulated with azide-labeled macrophage membranes to extend circulation time and enhance targeting efficiency. Additionally, bioorthogonal metabolic glycolengineering was employed to modify luciferase-labeled tumor cells (4T1-Luc) with bicyclo[6.1.0]nonyne (BCN) groups, facilitating precise binding between the probes and 4T1-Luc cells through click chemistry reactions. This dual-modal imaging approach enabled real-time monitoring of small metastatic lesions with high sensitivity, providing a non-invasive and accurate method for assessing tumor metastasis and therapeutic response in vivo. Our findings indicate that the dual-modal imaging technique, combining SERS and bioluminescence with bioorthogonal labeling, holds significant potential for advanced applications in oncology. STATEMENT OF SIGNIFICANCE: This study devised a surface-enhanced Raman scattering (SERS) and bioluminescence dual-modal imaging strategy integrated with a bioorthogonal label to address the challenge of tracking the metastasis of aggressive triple-negative breast cancer (TNBC). In contrast to conventional methods, this approach facilitated real-time, whole-body monitoring of tumor dissemination through bioluminescence. Simultaneously, it achieved the detection of micro-metastases in organs using SERS, thereby exceeding the sensitivity limitations of existing imaging techniques. Clinical validation with human samples further demonstrated its potential for non-invasive therapeutic assessment and early intervention. By bridging preclinical innovation and clinical requirements, this research offered a transformative tool for precision oncology. It is expected to attract the interest of researchers in the fields of biomedicine, nanotechnology, and cancer therapeutics.
Collapse
Affiliation(s)
- Wei Zhang
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China
| | - Sisi Wang
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China; Department of Breast and Thyroid Surgery, The Second Affiliated Hospital, Hainan Medical University, Haikou 571199, PR China
| | - Yanlong Xing
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China
| | - Xianzhu Luo
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China
| | - Rui Wang
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China.
| | - Fabiao Yu
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China.
| |
Collapse
|
4
|
Ediriweera GR, Li M, Fletcher NL, Houston ZH, Ahamed M, Blakey I, Thurecht KJ. Harnessing nanoparticles and bioorthogonal chemistries for improving precision of nuclear medicine. Biomater Sci 2025; 13:2297-2319. [PMID: 40135276 DOI: 10.1039/d4bm01387e] [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: 03/27/2025]
Abstract
The convergence of nanotechnology, radiopharmaceutical development and molecular imaging has unveiled exciting opportunities for the progress of innovative diagnostic and therapeutic strategies, paving the way for significant advancements in biomedical research, especially in relation to cancer. For example, the use of highly sensitive and quantitative nuclear imaging techniques including PET and SPECT, together with nanoparticles for tumour imaging and therapy has recently expanded rapidly. While the long circulating properties of many nanomaterials are beneficial for prodrug chemotherapy formulations, due to the constant decay processes involved in nuclear medicines, directly labelled materials result in prolonged systemic radiation exposure and reduced therapeutic indices due to the unfavourable target-to-background ratios. This is due to the tendency for long circulating nanomaterials to distribute within the blood to other organs, such as the liver and spleen. The recent integration of bioorthogonal chemistry with nanotechnology and molecular imaging/radiotherapy has revolutionized the field by allowing the decoupling of the targeting molecule (i.e. nanomaterial with a bioorthogonal tag) and the imaging/therapeutic radioisotope. In this way, the detection/therapeutic element can be administered as a secondary "chase" molecule that contains the bioorthogonal partner, thereby creating an avenue to improve therapeutic index and provide imaging and treatments with reduced risk. This review will provide an overview of the progress made thus far in the field of nuclear imaging and radiotherapy for cancer using the combination of nanomaterials and bioorthogonal chemistry. We also provide a critical evaluation of the challenges and opportunities for using these approaches to better understand disease and treatment mechanisms, with the potential for downstream clinical translation.
Collapse
Affiliation(s)
- Gayathri R Ediriweera
- Centre for Advanced Imaging and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
- ARC Research Hub for Advanced Manufacture of Targeted Radiopharmaceuticals, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Mengdie Li
- Centre for Advanced Imaging and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Nicholas L Fletcher
- Centre for Advanced Imaging and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
- ARC Research Hub for Advanced Manufacture of Targeted Radiopharmaceuticals, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Zachary H Houston
- Centre for Advanced Imaging and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Muneer Ahamed
- Centre for Advanced Imaging and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Idriss Blakey
- Centre for Advanced Imaging and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
- ARC Research Hub for Advanced Manufacture of Targeted Radiopharmaceuticals, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Kristofer J Thurecht
- Centre for Advanced Imaging and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
- ARC Research Hub for Advanced Manufacture of Targeted Radiopharmaceuticals, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
5
|
Lim H, Lee JH, Park SH, Lee JH, Jang H, Yang SB, Seo M, Lee S, Park J. Tumor-specific biochemical nanoconversion of self-assembled peptide-conjugated paclitaxel-docetaxel-based nanoparticles. NANO CONVERGENCE 2025; 12:20. [PMID: 40285925 PMCID: PMC12033163 DOI: 10.1186/s40580-025-00487-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
Docetaxel (DTX, 1) and paclitaxel (PTX, 2) are famous cytotoxic agents widely used in cancer therapy, however, their low specificity for tumor cells often results in severe systemic toxicity. Beyond conventional prodrug strategies, this study introduces a novel nanoconversion technology that chemically modifies DTX to form self-assembled nanoparticles (NPs), which subsequently convert into a paclitaxel-mimicking molecule (PTXm, 3). Hydrophilic acetylated Phe-Arg-Arg-Phe peptide ((Ac)FRRF, 4) and hydrophobic docetaxel were conjugated to prepare self-assembled (Ac)FRRF-DTX NPs. The selective cleavage of the Arg-Phe bond by cathepsin B, which is abundant in cancer cells, facilitated the nanoconversion of PTXm (3) from (Ac)FRRF-DTX NPs, demonstrating effective cytotoxic effects. Utilizing the cleavage site of peptide and specific sequences (ex. Arg-Arg-Phe), this approach does not simply act as a prodrug but allows the nanomaterial to transform into another cytotoxic biomolecule within tumors. (Ac)FRRF-DTX NPs exhibited remarkable physicochemical properties, superior anti-cancer efficacy, and low toxicity, showcasing an innovative conversion in peptide-conjugated nanomedicine. Unlike traditional prodrug chemistry, this tumor-specific nanoconversion process involves the biochemical transformation of DTX (1) into PTXm (3) via enzymatic action. Overall, this study provides an outstanding example of chemical drug molecular modification through the concept of nanoconversion.
Collapse
Affiliation(s)
- Hansol Lim
- Department of Applied Life Science, BK21 Program, Konkuk University, Chungju, 27478, Republic of Korea
| | - Jae-Hyeon Lee
- Department of Applied Life Science, BK21 Program, Konkuk University, Chungju, 27478, Republic of Korea
| | - So-Hyeon Park
- Department of Applied Life Science, BK21 Program, Konkuk University, Chungju, 27478, Republic of Korea
| | - Jun-Hyuck Lee
- Department of Applied Life Science, BK21 Program, Konkuk University, Chungju, 27478, Republic of Korea
| | - Hyesu Jang
- College of Pharmacy, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Seong-Bin Yang
- Department of Applied Life Science, BK21 Program, Konkuk University, Chungju, 27478, Republic of Korea
| | - Minho Seo
- Department of Applied Life Science, BK21 Program, Konkuk University, Chungju, 27478, Republic of Korea
| | - Seokwoo Lee
- College of Pharmacy, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Jooho Park
- Department of Applied Life Science, BK21 Program, Konkuk University, Chungju, 27478, Republic of Korea.
- Department of Biomedical Chemistry, College of Biomedical and Health Science, Konkuk University, Chungju, 27478, Republic of Korea.
| |
Collapse
|
6
|
Ma J, Wang X, Hu Y, Ma J, Ma Y, Chen H, Han Z. Recent Advances in Augmenting the Therapeutic Efficacy of Peptide-Drug Conjugates. J Med Chem 2025. [PMID: 40267310 DOI: 10.1021/acs.jmedchem.5c00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
There is an urgent need for the development of safe and effective modalities for the treatment of diseases owing to drug resistance, undesired side effects, and poor clinical outcomes. Combining cell-targeting and efficient cell-killing properties, peptide-drug conjugates (PDCs) have demonstrated superior efficacy compared with peptides and payloads alone. However, innovative molecular designs of PDCs are essential for further improving targeting precision, protease resistance and stability, cell permeability, and overall treatment efficacy. Several strategies have been developed to address these challenges, such as multivalency approaches, bispecific targeting, and long-acting PDCs. Other novel strategies, including overcoming biological barriers, conjugating novel functional payloads, and targeting macropinocytosis, have also shown promise. This perspective compiles the most recent strategies for enhancing PDC treatment efficacy, highlights key advancements in PDC, and provides insights on future directions for the development of novel PDCs.
Collapse
Affiliation(s)
- Jiahui Ma
- Gansu Provincial Key Laboratory of Environmental Oncology, Department of Tumor Center, Lanzhou University Second Hospital, Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Xuedan Wang
- School of Life Sciences and Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Yonghua Hu
- Gansu Provincial Key Laboratory of Environmental Oncology, Department of Tumor Center, Lanzhou University Second Hospital, Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
- Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Jianping Ma
- School of Life Sciences and Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Yaping Ma
- Shenzhen DIVBIO Pharmaceutical, Shenzhen 518057, China
| | - Hao Chen
- Gansu Provincial Key Laboratory of Environmental Oncology, Department of Tumor Center, Lanzhou University Second Hospital, Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Zhijian Han
- Gansu Provincial Key Laboratory of Environmental Oncology, Department of Tumor Center, Lanzhou University Second Hospital, Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
7
|
Cedrún-Morales M, Migliavacca M, Ceballos M, Perez-Maseda M, Zampini G, Alameda Felgueiras MT, Ostolaza-Paraiso J, Juanes M, Rincón I, Fairen-Jimenez D, Montenegro J, Horcajada P, Polo E, Pelaz B, Del Pino P. Clickable Polymer-Based Coatings for Modulating the Interaction of Metal-Organic Framework Nanocrystals with Living Cells. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40257304 DOI: 10.1021/acsami.5c01695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Nanosized microporous metal-organic-frameworks (NMOFs) serve as versatile drug delivery systems capable of navigating complex microenvironments and interacting with cells in specific tissues. The physicochemical properties of NMOFs, such as size, composition, porosity, colloidal stability, and external surface functionalization are essential for their success as efficient carriers. This study introduces a flexible, clickable coating using an amphiphilic polymer derivatized with dibenzo cyclooctyne groups as a universal, postsynthetic functionalization tool. To prove its universality, nanosized MOFs with different structure and composition (UiO-67, NU-1000, PCN-222, and ZIF-8) were produced with high monodispersity and were coated with a clickable, amphiphilic polymer. The resulting polymer-coated NMOFs display exceptional colloidal and structural stability in different biologically relevant media. For comparative purposes, we selected two size-equivalent NMOFs, ZIF-8 and UiO-67, which were functionalized with a library of biologically relevant azide-derivatized (macro)molecules, including poly(ethylene glycol), mannose, and a dynein-binding cell-penetrating peptide, using a bioorthogonal reaction. The choice of ZIF-8 and UiO-67, both 150 nm in size but with distinct coordination and surface chemistries, is pivotal due to their differing acid and base stability characteristics, which may potentially influence their performance in cellular environments. To track their performance in vitro, the NMOFs were loaded with cresyl violet, a common histological stain and lysosomal marker. Cellular internalization of the surface-functionalized NMOFs was markedly governed by their distinct (macro)molecule characteristics. This demonstrates that surface properties critically influence uptake efficiency, while also highlighting the versatility and effectiveness of the proposed coating strategy. In particular, the one functionalized with the dynein-binding peptide demonstrated a markedly higher rate of cellular internalization compared to other NMOFs. In contrast, derivatizations with mannose and poly(ethylene glycol) are associated with a substantial reduction in cellular uptake, suggesting stealth behavior. These results provide a bioorthogonal and versatile alternative for the external surface engineering of NMOFs, aiming to improve targeted drug delivery effectiveness.
Collapse
Affiliation(s)
- Manuela Cedrún-Morales
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Física de Partículas, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Martina Migliavacca
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Manuel Ceballos
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Física de Partículas, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Marta Perez-Maseda
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Física de Partículas, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Giulia Zampini
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - María Teresa Alameda Felgueiras
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Jon Ostolaza-Paraiso
- The Adsorption and Advanced Materials Laboratory (A2ML), Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Marisa Juanes
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Irene Rincón
- Advanced Porous Materials Unit (APMU), IMDEA Energy Institute, Av. Ramón de la Sagra 3, 28935 Móstoles-Madrid, Spain
| | - David Fairen-Jimenez
- The Adsorption and Advanced Materials Laboratory (A2ML), Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Javier Montenegro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Patricia Horcajada
- Advanced Porous Materials Unit (APMU), IMDEA Energy Institute, Av. Ramón de la Sagra 3, 28935 Móstoles-Madrid, Spain
| | - Ester Polo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Bioquímica y Biología Molecular, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Beatriz Pelaz
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Pablo Del Pino
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Física de Partículas, Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain
| |
Collapse
|
8
|
Tan Z, Zheng L, Bo Y, Kambar N, Wang H, Leal C. Click Lipid Nanoparticles for the Delivery of mRNA to Metabolically Labeled Cancer Cells. Biochemistry 2025; 64:1807-1816. [PMID: 40181500 DOI: 10.1021/acs.biochem.4c00699] [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: 04/05/2025]
Abstract
Lipid nanoparticle (LNP)-based mRNA delivery has a lot of potential in combating a wide range of diseases, but delivering mRNA to specific cell types continues to be challenging. Despite recent advances in organ and cell specificity, the majority of clinical LNP systems cannot fully release their payload to a targeted site. Incorporating active targeting moieties into LNPs is highly desired to expand nanomedicine applications. In this Letter, we developed LNPs that harness the power of bioorthogonal "click" azide-alkyne chemical reactions. We show that the plasma membranes of cancer cells can be labeled with azide groups by metabolic sugar labeling, and these azide groups can react with dibenzocyclooctyne (DBCO) on LNPs to achieve specific binding. To achieve this, we synthesized new and versatile lipids by functionalizing DBCO groups to phospholipids with or without a poly(ethylene glycol) (PEG) linker. The DBCO lipids were successfully formulated into DBCO-LNPs comprising other standard lipid compounds. When using these DBCO-LNPs to deliver mRNA to metabolically labeled cells, DBCO-LNPs showed a remarkable ability to preferentially deliver mRNA to azide-labeled cells. Removing PEG linkers from DBCO lipids enables better integration and retention in the LNP, and the higher the amount of DBCO lipid, the stronger the targeting effect. This work demonstrates that cell-specific targeting can be achieved utilizing azide-alkyne ″click″ chemistry and could inspire the development of the next generation of LNPs for active cyto-tropic nanomedicines.
Collapse
Affiliation(s)
- Zhengzhong Tan
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Lining Zheng
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yang Bo
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Nurila Kambar
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Hua Wang
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Cecilia Leal
- Department of Materials Science and Engineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
9
|
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
|
10
|
Schauenburg D, Weil T. Not So Bioorthogonal Chemistry. J Am Chem Soc 2025; 147:8049-8062. [PMID: 40017419 PMCID: PMC11912343 DOI: 10.1021/jacs.4c15986] [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: 11/12/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/01/2025]
Abstract
The advent of bioorthogonal chemistry has transformed scientific research, offering a powerful tool for selective and noninvasive labeling of (bio)molecules within complex biological environments. This innovative approach has facilitated the study of intricate cellular processes, protein dynamics, and interactions. Nevertheless, a number of challenges remain to be addressed, including the need for improved reaction kinetics, enhanced biocompatibility, and the development of a more diverse and orthogonal set of reactions. While scientists continue to search for veritable solutions, bioorthogonal chemistry remains a transformative tool with a vast potential for advancing our understanding of biology and medicine. This Perspective offers insights into reactions commonly classified as "bioorthogonal", which, however, may not always demonstrate the desired selectivity regarding the interactions between their components and the additives or catalysts used under the reaction conditions.
Collapse
Affiliation(s)
- Dominik Schauenburg
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Tanja Weil
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
11
|
Cao S, Zhou P, Shen G, Ivanov T, Yan X, Landfester K, Caire da Silva L. Binary peptide coacervates as an active model for biomolecular condensates. Nat Commun 2025; 16:2407. [PMID: 40069227 PMCID: PMC11897134 DOI: 10.1038/s41467-025-57772-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Biomolecular condensates formed by proteins and nucleic acids are critical for cellular processes. Macromolecule-based coacervate droplets formed by liquid-liquid phase separation serve as synthetic analogues, but are limited by complex compositions and high molecular weights. Recently, short peptides have emerged as an alternative component of coacervates, but tend to form metastable microdroplets that evolve into rigid nanostructures. Here we present programmable coacervates using binary mixtures of diphenylalanine-based short peptides. We show that the presence of different short peptides stabilizes the coacervate phase and prevents the formation of rigid structures, allowing peptide coacervates to be used as stable adaptive compartments. This approach allows fine control of droplet formation and dynamic morphological changes in response to physiological triggers. As compartments, short peptide coacervates sequester hydrophobic molecules and enhance bio-orthogonal catalysis. In addition, the incorporation of coacervates into model synthetic cells enables the design of Boolean logic gates. Our findings highlight the potential of short peptide coacervates for creating adaptive biomimetic systems and provide insight into the principles of phase separation in biomolecular condensates.
Collapse
Affiliation(s)
- Shoupeng Cao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, PR China
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Peng Zhou
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Science, Beijing, 100190, PR China
| | - Guizhi Shen
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Science, Beijing, 100190, PR China
| | - Tsvetomir Ivanov
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Xuehai Yan
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Science, Beijing, 100190, PR China.
| | | | - Lucas Caire da Silva
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany.
- Department of Chemistry, McGill University, Montreal, H3A 0B8, Canada.
| |
Collapse
|
12
|
Bresinsky M, Goepferich A. Control of biomedical nanoparticle distribution and drug release in vivo by complex particle design strategies. Eur J Pharm Biopharm 2025; 208:114634. [PMID: 39826847 DOI: 10.1016/j.ejpb.2025.114634] [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: 10/16/2024] [Revised: 01/06/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
The utilization of targeted nanoparticles as a selective drug delivery system is a powerful tool to increase the amount of active substance reaching the target site. This can increase therapeutic efficacy while reducing adverse drug effects. However, nanoparticles face several challenges: upon injection, the immediate adhesion of plasma proteins may mask targeting ligands, thereby diminishing the target cell selectivity. In addition, opsonization can lead to premature clearance and the widespread presence of receptors or enzymes limits the accuracy of target cell recognition. Nanoparticles may also suffer from endosomal entrapment, and controlled drug release can be hindered by premature burst release or insufficient particle retention at the target site. Various strategies have been developed to address these adverse events, such as the implementation of switchable particle properties, regulating the composition of the formed protein corona, or using click-chemistry based targeting approaches. This has resulted in increasingly complex particle designs, raising the question of whether this development actually improves the therapeutic efficacy in vivo. This review provides an overview of the challenges in targeted drug delivery and explores potential solutions described in the literature. Subsequently, appropriate strategies for the development of nanoparticular drug delivery concepts are discussed.
Collapse
Affiliation(s)
- Melanie Bresinsky
- Department of Pharmaceutical Technology, University of Regensburg 93053 Regensburg, Bavaria, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg 93053 Regensburg, Bavaria, Germany.
| |
Collapse
|
13
|
Zhang Y, Huang Q, Lei F, Qian W, Zhang C, Wang Q, Liu C, Ji H, Wang F. Exploring New Bioorthogonal Catalysts: Scaffold Diversity in Catalysis for Chemical Biology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404431. [PMID: 39921286 PMCID: PMC11884534 DOI: 10.1002/advs.202404431] [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: 04/25/2024] [Revised: 01/11/2025] [Indexed: 02/10/2025]
Abstract
Bioorthogonal catalysis has revolutionized the field of chemical biology by enabling selective and controlled chemical transformations within living systems. Research has converged on the development of innovative catalyst scaffolds, seeking to broaden the scope of bioorthogonal reactions, boost their efficiency, and surpass the limitations of conventional catalysts. This review provides a comprehensive overview of the latest advancements in bioorthogonal catalyst research based on different scaffold materials. Through an in-depth analysis of fabrication strategies and applications of bioorthogonal catalysts, this review discusses the design principles, mechanisms of action, and applications of these novel catalysts in chemical biology. Current challenges and future directions in exploring the scaffold diversity are also highlighted. The integration of diverse catalyst scaffolds offers exciting prospects for precise manipulation of biomolecules and the development of innovative therapeutic strategies in chemical biology. In addition, the review fills in the gaps in previous reviews, such as in fully summarizing the presented scaffold materials applied in bioorthogonal catalysts, emphasizing the potential impact on advancing bioorthogonal chemistry, and offering prospects for future development in this field.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Special Environmental MedicineNantong UniversityNantong226019China
| | - Qizhen Huang
- School of Public HealthNantong UniversityNantong226019China
| | - Fang Lei
- School of Public HealthNantong UniversityNantong226019China
| | - Wanlong Qian
- Institute of Special Environmental MedicineNantong UniversityNantong226019China
| | - Chengfeng Zhang
- Institute of Special Environmental MedicineNantong UniversityNantong226019China
| | - Qi Wang
- School of Public HealthNantong UniversityNantong226019China
| | - Chaoqun Liu
- School of PharmacyHenan UniversityKaifeng475004China
| | - Haiwei Ji
- School of Public HealthNantong UniversityNantong226019China
| | - Faming Wang
- School of Public HealthNantong UniversityNantong226019China
| |
Collapse
|
14
|
Yang S, Zhu H, Jin H, Wang K, Song J, Sun N, Liu Y, Yin X, Wang R, Wu X, Liu H, Zhang C, Zhao W, Yu F. Bio-orthogonal-labeled exosomes reveals specific distribution in vivo and provides potential application in ARDS therapy. Biomaterials 2025; 319:123208. [PMID: 40023928 DOI: 10.1016/j.biomaterials.2025.123208] [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: 11/07/2024] [Revised: 02/15/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
Exosomes derived from specific cells may be useful for targeted drug delivery, but tracking them in vivo is essential for their clinical application. However, their small size and complex structure challenge the development of exosome-tracking techniques, and traditional labeling methods are limited by weak affinity and potential toxicity. To address these issues, here we developed a novel bio-orthogonal labeling strategy based on phosphatidylinositol derivatives to fluorescently label exosomes from various human and mouse cell types. The different cell-derived exosomes revealed organ-specific distribution patterns and a favorable safety profile. Notably, 4T1 cell-derived exosomes specifically targeted the lungs. When used as drug carriers loaded with anti-inflammatory resveratrol, these exosomes showed significant therapeutic efficacy in mice with acute respiratory distress syndrome (ARDS), effectively reducing inflammatory responses, mitigating pulmonary fibrosis, and restoring lung tissue morphology and function. Our findings provide a novel exosome labeling strategy and an invaluable tool for their in vivo tracking and targeting screening, while exosomes that specifically target the lungs offer a potential therapeutic strategy for organ-specific diseases such as ARDS.
Collapse
Affiliation(s)
- Song Yang
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China; State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Haomiao Zhu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China; Department of Pharmacy, Qilu Hospital, Shandong University, No.107 Cultural West Road, Jinan, 250012, China
| | - Hongzhen Jin
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China; State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Kun Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Junna Song
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Na Sun
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China
| | - Yonghui Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China; School of Chemistry, Tiangong University, No.399 BinShuiXi Road, Tianjin, 300387, China
| | - Xiaona Yin
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Rui Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Xiao Wu
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China
| | - Huadong Liu
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China
| | - Chunling Zhang
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China.
| | - Wei Zhao
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China; State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China.
| | - Fan Yu
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China; State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China.
| |
Collapse
|
15
|
Hu W, Li M, Feng Y, Wang X, Yang S, Gao Y, Jiang D, Lan X. Molecular Imaging for Biomimetic Nanomedicine in Cancer Therapy: Current Insights and Challenges. ACS APPLIED MATERIALS & INTERFACES 2025; 17:10231-10245. [PMID: 39878693 DOI: 10.1021/acsami.4c19720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Coating biological membranes onto biomimetic nanocarriers improves biocompatibility, prolongs circulation, and enhances targeted delivery for cancer precision medicine. To better understand the biodistribution profiles of these biomimetic nanosystems, molecular imaging techniques, including optical imaging, radionuclide imaging, magnetic resonance imaging, and ultrasound imaging, have been widely employed for in vivo tracking and dynamic imaging. Here in this review, we delve into the profound role of these imaging modalities in visualizing changes in the tumor microenvironment, particularly in monitoring oxygen consumption and immune response dynamics, highlighting their potential to improve cancer therapies. We also briefly discuss current applications of molecular imaging in synergistic cancer therapies and future perspectives. Finally, we offer insights into the potential of integrating biomimetic nanomedicine with molecular imaging for clinical translation.
Collapse
Affiliation(s)
- Wenzhu Hu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Mengting Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan 430022, China
| | - Yuan Feng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xingyi Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Shaowen Yang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yu Gao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan 430022, China
| |
Collapse
|
16
|
Ferreira TM, Navo CD, Agnes JP, Cardozo F, de Oliveira DC, Zanotto-Filho A, Jiménez-Osés G, Domingos JB. Mechanistic Insights into Chloride-Dependent Uncaging Reaction of Propargyl and Allene-Protected Substrates by Pd-Complexes. Chemistry 2025; 31:e202403987. [PMID: 39620662 DOI: 10.1002/chem.202403987] [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: 10/28/2024] [Indexed: 12/12/2024]
Abstract
This study investigates the effect of chloride levels on the mode of action of palladium complexes for the activation of propargyl- and allene-protected fluorophores and chemotherapeutic drugs through uncaging reactions. Four Pd(II) complexes were synthesized and characterized using various spectroscopic techniques to confirm their structure and electronic properties. Kinetic studies and density functional theory calculations revealed that chloride ions in phosphate buffered saline (PBS) significantly enhance catalytic efficiency, particularly for allenyl-protected substrates compared to propargylic counterparts. This enhancement is attributed to the neutral charge nature of the Pd complex. The data suggest that neutral complexes are less prone to chloride exchange by water molecules. Additionally, chloride ligands counterbalance the unusually high stability of the key σ-bound η1-Pd intermediates in the aquo complexes in PB, leading to an overall higher reactivity. These results highlight the impact of fine-tuning the electronic properties of the metal center through both designed ligands and environmental factors. Bench evaluations and tests with living breast cancer cells demonstrated that a Pd catalyst complex with a bidentate ligand effectively activates the prodrugs propargyl-5-fluorouracil (Prop-5FU) and allene-5-Fluorouracil (Alle-5FU), the latter being a novel prodrug. The Pd catalyst successfully released the active drug, inducing significant cytotoxicity, especially with Alle-5FU, which operates at lower catalyst concentrations than Pro-5FU.
Collapse
Affiliation(s)
- Thuany M Ferreira
- Laboratory of Biomimetic Catalysis (LaCBio), Department of Chemistry, Federal University of Santa Catarina (UFSC), Campus Trindade, 88040-900, Florianópolis - SC, Brazil
| | - Claudio D Navo
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160, Derio, Spain
| | - Jonathan P Agnes
- Laboratory of Biomimetic Catalysis (LaCBio), Department of Chemistry, Federal University of Santa Catarina (UFSC), Campus Trindade, 88040-900, Florianópolis - SC, Brazil
| | - Filipe Cardozo
- Laboratory of Biomimetic Catalysis (LaCBio), Department of Chemistry, Federal University of Santa Catarina (UFSC), Campus Trindade, 88040-900, Florianópolis - SC, Brazil
| | - Daniela C de Oliveira
- Brazilian Synchrotron Light Laboratory (LNLS), C. P. 6192, 13083-970, Campinas - SP, Brazil
| | - Alfeu Zanotto-Filho
- Laboratory of Cancer Pharmacology (LabCancer) Department of Pharmacology, Federal University of Santa Catarina (UFSC), Campus Trindade, 88040-900, Florianópolis - SC, Brazil
| | - Gonzalo Jiménez-Osés
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160, Derio, Spain
- Ikerbasque Basque Foundation for Science, 48013, Bilbao, Spain
| | - Josiel B Domingos
- Laboratory of Biomimetic Catalysis (LaCBio), Department of Chemistry, Federal University of Santa Catarina (UFSC), Campus Trindade, 88040-900, Florianópolis - SC, Brazil
| |
Collapse
|
17
|
Chen Y, Liu L, Li M, Chen X, Li Y, Tao J, Deng Y. Nanoparticle-enabled In Situ drug potency activation for enhanced tumor-specific therapy. Eur J Pharm Sci 2025; 205:106989. [PMID: 39675436 DOI: 10.1016/j.ejps.2024.106989] [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: 10/16/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
Cancer treatment faces significant challenges including inadequate tumor specificity, drug resistance, and severe side effects, often resulting in unsatisfactory patient outcomes. Nanomedicines offer a transformative platform for tumor-targeted drug delivery and antitumor potency activation, providing an indispensable strategy for overcoming the severe damage to normal tissues caused by the inherent "always-on" cytotoxicity of conventional therapeutic agents. This review focuses on the emerging concept of "nanoparticle-enabled in situ drug potency activation", where inactive or minimally toxic agents are selectively activated within tumors to enhance the therapeutic efficacy and minimize the adverse effects. We systematically analyzed literature from PubMed and Web of Science databases spanning the last two decades, emphasizing experimental evidence supporting this in situ drug potency activation concept. Key strategies including stimuli-responsive prodrug nanoparticles, metal-induced activation, and bioorthogonal reactions are critically evaluated for their potential to overcome limitations in current cancer therapies. The findings highlight the potential of in situ potency activation as a promising alternative to conventional therapeutics, with far-reaching implications for advancing effective and safe cancer treatments.
Collapse
Affiliation(s)
- Yitian Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Lishan Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Ming Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Xiaolian Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yaoqi Li
- Department of Pharmacy, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jing Tao
- Department of Pharmacy, the First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Yibin Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China; State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China.
| |
Collapse
|
18
|
Di Ianni E, Jeon J, Hu H, Quintana JM, Lee C, Haidar EA, Mohammadi S, Zargani-Piccardi A, Mahamdeh M, Hernández IC, Ng TSC, Breyne K, Lee H, Breakefield XO, Miller MA. Enhanced mRNA delivery using ultrasound-delivered click reactive anchors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635330. [PMID: 39975219 PMCID: PMC11838329 DOI: 10.1101/2025.01.28.635330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Therapeutic nucleic acid delivery has many potential applications, but it remains challenging to target extrahepatic tissues in a flexible and image-guided manner. To address this issue, we report a bioorthogonal pre-targeting strategy that uses focused ultrasound to promote the delivery of mRNA-loaded lipid nanoparticles (mRNA-LNP). We synthesized amphiphilic click reactive anchors (ACRAs) consisting of a phospholipid PEG-conjugate functionalized with transcyclooctene (TCO) or its companion reactive partner methyltetrazine (mTz), yielding ACRA-TCO and ACRA-mTz. ACRA derivatives were screened for cellular activity, yielding functionalized DOPE-PEG (1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N- (polyethylene glycol)) derivatives outperforming those containing saturated lipid or branched PEG. Nanobubbles encapsulating ultrasound-responsive gas precursor delivered ACRA-TCO to targeted cells and tissues using focused ultrasound, and this pre-targeting promoted the subsequent delivery of mRNA- LNP functionalized with companion ACRA-mTz. In cell cultures and in mice, ultrasound pre-targeting enhanced the accumulation of mTz-functionalized small molecule and nanoparticle compounds by 75% and 3.6-fold, respectively, and increased gene expression using mRNA-LNP in vivo . Taken together, this report presents a modular, ultrasound-enabled strategy for enhancing nucleic acid delivery in targeted tissues.
Collapse
|
19
|
Miao Y, Wang Y, Chen Y, Huang Z, Lu C, Liu Y, Chen F, Wen X, Zhang J, Zhu S, Zhao P, Chen Y, Tian T, Zhang Y, Xie H, Lin J, Ye D. Pretargeted Multimodal Tumor Imaging by Enzymatic Self-Immobilization Labeling and Bioorthogonal Reaction. J Am Chem Soc 2025; 147:2809-2821. [PMID: 39801138 DOI: 10.1021/jacs.4c15896] [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: 01/23/2025]
Abstract
Covalent modification of cell membranes has shown promise for tumor imaging and therapy. However, existing membrane labeling techniques face challenges such as slow kinetics and poor selectivity for cancer cells, leading to off-target effects and suboptimal in vivo efficacy. Here, we present an enzyme-triggered self-immobilization labeling strategy, termed E-SIM, which enables rapid and selective labeling of tumor cell membranes with bioorthogonal trans-cycloctene (TCO) handles in vivo. E-SIM utilizes P-TCO, an alkaline phosphatase (ALP) responsive quinone methide (QM) precursor with a TCO group, facilitating the rapid conjugation of high-density TCO handles onto tumor cell membranes via proximity labeling. These TCO groups then react efficiently with tetrazine (Tz)-bearing reporters via a fast bioorthogonal reaction, resulting in significant enrichment of reporters of various sizes and imaging modalities on tumor cell membranes. We demonstrate the efficacy of E-SIM labeling and bioorthogonal reaction for pretargeted multimodality imaging of tumors in vivo. Notably, we achieve selective and efficient installation of Tz-modified Renilla luciferase on tumor cells in vivo, thereby offering highly sensitive bioluminescence signals for detecting and guiding the surgical removal of small human HepG2 liver tumor peritoneal metastases. E-SIM represents a robust tool for precise tumor cell labeling in complex in vivo environments, feasible for pretargeted enrichment of various reporters in tumors for multimodal imaging applications.
Collapse
Affiliation(s)
- Yinxing Miao
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Yuqi Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Yefeng Chen
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zheng Huang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Chunmei Lu
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Yili Liu
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Fangfang Chen
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xidan Wen
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Junya Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Shiliang Zhu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Pengke Zhao
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Yunhan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Tian Tian
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Yan Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| | - Hexin Xie
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jianguo Lin
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Deju Ye
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Road, Nanjing 210023, China
| |
Collapse
|
20
|
Huang G, He Y, Chen X, Yin T, Ma A, Zhu L, Chen L, Liang R, Zhang P, Pan H, Cai L. Bioorthogonal oncolytic-virus nanovesicles combined bio-immunotherapy with CAR-T cells for solid tumors. Biomater Sci 2025; 13:457-465. [PMID: 39607022 DOI: 10.1039/d4bm01305k] [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: 11/29/2024]
Abstract
Various oncolytic viruses (OVs) have been adopted as therapeutic tools to increase the efficacy of chimeric antigen receptor (CAR)-T cells against solid tumors. However, the therapeutic effect of OVs has been limited by pre-existing neutralizing antibodies and poor targeting delivery for systemic administration. Herein, we propose using bioorthogonal OV nanovesicles to boost the antitumor effects of CAR-T cells in solid tumors by reshaping the tumor microenvironment. Using a cell-membrane nanomimetic technique, we embedded artificial chemical ligands on cancer cell surfaces and then encapsulated lysoviral particles to obtain dual-targeted OV nanovesicles with bioorthogonal targeting and homologous recognition. OVs can be directly encapsulated into cancer cell nanovesicles and exhibit a liposome-like nanostructure, efficient loading, and excellent tumor-targeting capability. Encouragingly, OV nanovesicles efficiently induced tumor-cell apoptosis while sparing normal tissues and cells, thereby inhibiting tumor growth. Administration of viral nanovesicles effectively increased the secretion of anti-tumor cytokines such as IL-2, TNF-α and IFN-γ, and significantly promoted the infiltration and activation of CD8+CAR-T cells in tumors. Our data suggest that bioorthogonal OV nanovesicles hold great potential to overcome the limitations of CAR-T cells as monotherapies against solid tumors and, thus, drive the clinical application of combination therapy.
Collapse
Affiliation(s)
- Guojun Huang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
| | - Yiran He
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaocong Chen
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
| | - Ting Yin
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
| | - Aiqing Ma
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
| | - Lizhen Zhu
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
| | - Liqi Chen
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ruijing Liang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Pengfei Zhang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hong Pan
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Sino-European Center of Biomedicine and Health, Luohu, Shenzhen 518024, China
| |
Collapse
|
21
|
Hu H, Zhang C. Conjugation of Multiple Proteins Onto the Surface of PLGA/Lipid Hybrid Nanoparticles. J Biomed Mater Res A 2025; 113:e37807. [PMID: 39420678 DOI: 10.1002/jbm.a.37807] [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: 06/30/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024]
Abstract
Nanoparticles are increasingly being used in the development of vaccines for disease prevention or treatment. Recent research has demonstrated that conjugating a protein onto the surface of nanoparticles can significantly increase its immunogenicity. Considering various pathogens that threaten human health, multivalent vaccines are often desirable. Up to now, nanoparticle-based vaccines are mostly limited to one protein per nanoparticle. No research has been conducted to explore the possibility of conjugating more than one protein onto the surface of a nanoparticle. Here we developed a specific conjugation strategy to conjugate multiple proteins to the PLGA/lipid hybrid nanoparticle surface. The maleimide-thiol Michael addition, Aizde-DBCO (Dibenzocyclooctyne), and TCO (trans-cycloctene)-Tetrazine click chemistry were employed to conjugate three different proteins, subunit keyhole limpet hemocyanin (sKLH), Ovalbumin (OVA), and cross-reactive material 197 (CRM197), to the surface of PLGA/lipid hybrid nanoparticles (hNPs). The successful results of this study pave the way for developing multivalent vaccines against different pathogens.
Collapse
Affiliation(s)
- He Hu
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, Virginia, USA
| | - Chenming Zhang
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
22
|
Liu Y, Wang MY, Li FF, Jia WL, Ma RN, Zhang W, Shang L, Li XJ, Wang HS, Jia LP. Enhanced-electrochemiluminescence biosensor for detecting miRNA-21 based on a CuO-mediated click reaction and catalytic hairpin self-assembly. Talanta 2025; 282:127033. [PMID: 39406082 DOI: 10.1016/j.talanta.2024.127033] [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: 08/06/2024] [Revised: 09/10/2024] [Accepted: 10/10/2024] [Indexed: 11/20/2024]
Abstract
MicroRNAs (miRNAs) are closely associated with cancer and have been considered cancer biomarkers. Herein, we propose an electrochemiluminescence (ECL) biosensor for detecting miRNA-21 based on target-induced catalytic hairpin self-assembly (CHA) and CuO-mediated azide-alkyne cycloaddition. Two hairpin DNAs were employed: one was immobilized on magnetic beads (HP2) and another was labeled with CuO (HP1-CuO). HP1 and HP2 formed a duplex through CHA induced by miRNA-21, resulting in the immobilization of CuO on magnetic beads and in the recycling of miRNA-21. After magnetic separation, CuO was treated with hydrochloric acid to release Cu2+, which concentration is quantitatively proportional to the target concentration. Subsequently, Cu2+ was reduced to Cu+, which catalyzed the click reaction between Fc-C CH and SH-DNA-N3+ immobilized on a Au/g-C3N4 modified electrode. Thus, the ECL of Au/g-C3N4 was quenched by Fc, and miRNA-21 was indirectly detected through a change in ECL intensity. Benefiting from the amplification effect of CuO nanoparticle loading, CHA-based target recycling, and the catalytic effect of click reaction, the proposed ECL biosensor showed high sensitivity. Experimental results indicate that the ECL biosensor proposed for detecting miRNA-21 exhibits a wide linear range from 1 fM to 1 nM and a low detection limit of 0.26 fM (3σ/S). Furthermore, the ECL sensor was capable of measuring miRNA-21 in real serum with high selectivity, indicating its notable applicable potential in biomedicine and clinical diagnosis.
Collapse
Affiliation(s)
- Yu Liu
- College of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong, 252000, China
| | - Ming-Yue Wang
- College of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong, 252000, China
| | - Fei-Fei Li
- College of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong, 252000, China
| | - Wen-Li Jia
- College of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong, 252000, China
| | - Rong-Na Ma
- College of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong, 252000, China
| | - Wei Zhang
- College of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong, 252000, China
| | - Lei Shang
- College of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong, 252000, China
| | - Xiao-Jian Li
- College of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong, 252000, China
| | - Huai-Sheng Wang
- College of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong, 252000, China.
| | - Li-Ping Jia
- College of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong, 252000, China.
| |
Collapse
|
23
|
Robles-Remacho A, Martos-Jamai I, Tabraue-Chávez M, Aguilar-González A, Laz-Ruiz JA, Cano-Cortés MV, López-Delgado FJ, Guardia-Monteagudo JJ, Pernagallo S, Diaz-Mochon JJ, Sanchez-Martin RM. Click chemistry-based dual nanosystem for microRNA-122 detection with single-base specificity from tumour cells. J Nanobiotechnology 2024; 22:791. [PMID: 39710710 DOI: 10.1186/s12951-024-03071-6] [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: 10/15/2024] [Accepted: 12/11/2024] [Indexed: 12/24/2024] Open
Abstract
MicroRNAs (miRNAs) have been recognised as potential biomarkers due to their specific expression patterns in different biological tissues and their changes in expression under pathological conditions. MicroRNA-122 (miR-122) is a vertebrate-specific miRNA that is predominantly expressed in the liver and plays an important role in liver metabolism and development. Dysregulation of miR-122 expression is associated with several liver-related diseases, including hepatocellular carcinoma and drug-induced liver injury (DILI). Given the potential of miR-122 as a biomarker, its effective detection is important for accurate diagnosis. However, miRNA detection methods still face challenges, particularly in terms of accurately identifying miRNA isoforms that may differ by only a single base. Here, with the aim of advancing accessible methods for the detection of miRNAs with single-base specificity, we have developed a robust dual nanosystem that leverages the simplicity of click chemistry reactions. Using the dual nanosystem, we successfully detected miR-122 at single-base resolution using flow cytometry and analysed its expression in various tumour cell lines with high specificity and strong correlation with TaqMan assay results. We also detected miR-122 in serum and identified four single nucleotide variations in its sequence. The chemistry employed in this dual nanosystem is highly versatile and offers a promising opportunity to develop nanoparticle-based strategies that incorporate click chemistry and bioorthogonal chemistry for the detection of miRNAs and their isoforms.
Collapse
Affiliation(s)
- Agustín Robles-Remacho
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016, Granada, Spain.
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, School of Pharmacy, University of Granada, Campus Cartuja s/n, 18071, Granada, Spain.
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain.
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden.
| | - Ismael Martos-Jamai
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016, Granada, Spain
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, School of Pharmacy, University of Granada, Campus Cartuja s/n, 18071, Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
| | - Mavys Tabraue-Chávez
- DESTINA Genomica S.L, PTS Granada, Avenida de la Innovación 1, Edificio BIC, 18016, Armilla, Spain
| | - Araceli Aguilar-González
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016, Granada, Spain
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, School of Pharmacy, University of Granada, Campus Cartuja s/n, 18071, Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
| | - Jose A Laz-Ruiz
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016, Granada, Spain
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, School of Pharmacy, University of Granada, Campus Cartuja s/n, 18071, Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
| | - M Victoria Cano-Cortés
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016, Granada, Spain
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, School of Pharmacy, University of Granada, Campus Cartuja s/n, 18071, Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
| | - F Javier López-Delgado
- DESTINA Genomica S.L, PTS Granada, Avenida de la Innovación 1, Edificio BIC, 18016, Armilla, Spain
| | | | - Salvatore Pernagallo
- DESTINA Genomica S.L, PTS Granada, Avenida de la Innovación 1, Edificio BIC, 18016, Armilla, Spain
| | - Juan J Diaz-Mochon
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016, Granada, Spain.
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, School of Pharmacy, University of Granada, Campus Cartuja s/n, 18071, Granada, Spain.
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain.
| | - Rosario M Sanchez-Martin
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Avenida de la Ilustración, 18016, Granada, Spain
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, School of Pharmacy, University of Granada, Campus Cartuja s/n, 18071, Granada, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, Granada, Spain
| |
Collapse
|
24
|
Wang F, Bao C, Cui S, Han G, Yang W, Yu Y. Enzyme-free fluorescent DNA detection based on nucleic acid-templated click reaction via controllable synthesis of Cu 2O as heterogeneous nanocatalyst. Talanta 2024; 280:126692. [PMID: 39128313 DOI: 10.1016/j.talanta.2024.126692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 08/13/2024]
Abstract
In the field of nucleic acid amplification assays, developing enzyme-free, easy-to-use, and highly sensitive amplification approaches remains a challenge. In this work, we synthesized a heterogeneous Cu2O nanocatalyst (hnCu2O) with different particle sizes and shapes, which was used for developing enzyme- and label-free nucleic acid amplification methods based on the nucleic acid-templated azide-alkyne cycloaddition (AAC) reaction catalyzed by hnCu2O. The hnCu2O exhibited size- and shape-dependent catalytic activity, with smaller sizes and spherical-like shapes exhibiting superior activity. Spherical-like hnCu2O (61 ± 8 nm) not only achieved a ligation yield of up to 84.2 ± 3.9 % in 3 min but also exhibited faster kinetics in the nucleic acid-templated hnCu2O-catalyzed AAC reaction, with a high reaction rate of 0.65 min-1 and a half-life of 1.07 ± 0.09 min. Based on this result, we developed nucleic acid-templated click ligation linear amplification reaction (NA-CLLAR) and nucleic acid-templated click ligation exponential amplification reaction (NA-CLEAR) approach. By combining the recognition (complementary to the target sequence) and signal output (split G-quadruplex sequence) elements into a DNA probe, the NA-CLLAR and NA-CLEAR fluorescence assays achieved highly specific detection of target nucleic acids, with a detection limit of 2.8 aM based on G-quadruplex-enhanced fluorescence. This work is a valuable reference and will inspire researchers to design enzyme-free nucleic acid signal amplification strategies by developing different types of Cu(I) catalysts with improved catalytic activity.
Collapse
Affiliation(s)
- Fan Wang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, PR China
| | - Chenglong Bao
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, PR China
| | - Susu Cui
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, PR China
| | - Guanghui Han
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, PR China
| | - Weiwei Yang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, PR China.
| | - Yongsheng Yu
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, PR China.
| |
Collapse
|
25
|
İlem-Özdemir D, Santos-Oliveira R. Can radiopharmaceuticals be delivered by quantum dots? Expert Opin Drug Deliv 2024; 21:1689-1691. [PMID: 39420518 DOI: 10.1080/17425247.2024.2419446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/18/2024] [Accepted: 10/17/2024] [Indexed: 10/19/2024]
Affiliation(s)
- Derya İlem-Özdemir
- Faculty of Pharmacy, Department of Radiopharmacy, Ege University, Bornova, Izmir, Turkey
| | - Ralph Santos-Oliveira
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Laboratory of Synthesis of Novel Radiopharmaceuticals and Nanoradiopharmacy, Rio de Janeiro, Brazil
- Laboratory of Nanoradiopharmaceuticals and Radiopharmacy, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
26
|
Liu Z, Zhang W, Zhao H, Sun M, Zhao C, Ren J, Qu X. Light-Controlled Bioorthogonal Chemistry Altered Natural Killer Cell Activity for Boosted Adoptive Immunotherapy. Angew Chem Int Ed Engl 2024; 63:e202411905. [PMID: 39112373 DOI: 10.1002/anie.202411905] [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: 06/25/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 10/15/2024]
Abstract
Natural killer (NK) cell-based immunotherapy has received much attention in recent years. However, its practical application is still suffering from the decreased function and inadequate infiltration of NK cells in the immunosuppressive microenvironment of solid tumors. Herein, we construct light-responsive porphyrin Fe array-armed NK cells (denoted as NK@p-Fe) for cell behavior modulation via bioorthogonal catalysis. By installing cholesterol-modified porphyrin Fe molecules on the NK cell surface, a catalytic array with light-harvesting capabilities is formed. This functionality transforms NK cells into cellular factories capable of catalyzing the production of active agents in a light-controlled manner. NK@p-Fe can generate the active antineoplastic drug doxorubicin through bioorthogonal reactions to enhance the cytotoxic function of NK cells. Beyond drug synthesis, NK@p-Fe can also bioorthogonally catalyze the production of the FDA-approved immune agonist imiquimod (IMQ). The activated immune agonist plays a dual role, inducing dendritic cell maturation for NK cell activation and reshaping the tumor immunosuppressive microenvironment for NK cell infiltration. This work represents a paradigm for the modulation of adoptive cell behaviors to boost cancer immunotherapy by bioorthogonal catalysis.
Collapse
Affiliation(s)
- Zhengwei Liu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Wenting Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Huisi Zhao
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Mengyu Sun
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Chuanqi Zhao
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
27
|
Zheng J, Wang X, Qin H, Hou Y, Yang Q, Zhang X, Hun X. Target-Navigated CBT-Cys "Stapling" Coupled with CRISPR/Cas12a Amplification for the Photoelectrochemical Nucleic Acid Assay. Anal Chem 2024; 96:18011-18019. [PMID: 39331475 DOI: 10.1021/acs.analchem.4c03254] [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: 09/29/2024]
Abstract
Generally, rolling circle amplification (RCA) is based on an enzyme-linked padlock extension reaction. Herein, rapid linking that utilizes click chemistry for joining sticky ends of DNA molecules was developed. The ends of nucleic acid were modified with 2-cyano-6-aminobenzothiazole (CBT) and cystine (Cys-Cys), while glutathione was introduced to break the disulfide bond under target navigation and promote the linkage between CBT and Cys at the terminus of the nucleic acid at pH 7.4. Subsequently, RCA was performed using phi29 polymerase. CRISPR/Cas12a cleavage was triggered by the product of RCA amplification. Assisted by alkaline phosphatase, the electron exchange process between the photoelectroactive Sb@Co(OH)F nanorod and p-aminophenol (p-AP) was collected in the form of photoelectrochemical (PEC) signals. Mass spectrometry, gel electrophoresis, and PEC signals were employed to verify the linking process and the RCA coupled with CRISPR/Cas12a cleavage amplification. CBT-Cys connection exhibited a high reaction rate (23.79 M-1·s-1). This enzyme-free linking process was superior to traditional enzyme catalysis in terms of the reaction environment and linking rate. This efficient nonenzymatic joining system holds great potential for constructing nonhomologous end joining, modifying DNA with molecules, and facilitating nucleic acid-protein modification processes.
Collapse
Affiliation(s)
- Jie Zheng
- College of Biological Engineering, College of Chemistry and Molecular Engineering, Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Xiaoyu Wang
- College of Chemistry and Molecular Engineering, Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Hongqing Qin
- College of Biological Engineering, College of Chemistry and Molecular Engineering, Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Yaxiao Hou
- College of Chemistry and Molecular Engineering, Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Qianqian Yang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, P. R. China
| | - Xuzhi Zhang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, P. R. China
| | - Xu Hun
- College of Chemistry and Molecular Engineering, Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| |
Collapse
|
28
|
Liao M, Zhang Q, Huang J, Huang X, Cheng C, Tu J, Zhang D, Lu Q, Ma L. Near-infrared and ultrasound triggered Pt/Pd-engineered cluster bombs for the treatment of solid tumors. J Control Release 2024; 375:331-345. [PMID: 39278358 DOI: 10.1016/j.jconrel.2024.09.024] [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: 07/18/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024]
Abstract
Owing to the dense extracellular matrix and high interstitial fluid pressure in the tumor microenvironment, methods which enhance the permeation and retention of nano drugs into liver tumors remain unsatisfactory for successful tumor treatment. We designed a near-infrared (NIR)- and ultrasound (US)-triggered Pt/Pd-engineered "cluster bomb" (Pt/Pd-CB) which actively penetrates liver cancer cell membranes and achieves photothermal and sonodynamic therapy (SDT). The physical forces generated by the fast expansion and collapse of perfluoropentane nanodroplets eject "sub bombs" (Pt/Pd nanoalloys) into liver cancer cells upon activation by NIR and US. Pt/Pd nanoalloys can then convert H2O2 into O2 to alleviate hypoxia and boost SDT efficiency while exhibiting a highly efficient photothermal response under NIR irradiation. Our findings might especially be promising for the treatment of solid tumors.
Collapse
Affiliation(s)
- Min Liao
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qi Zhang
- School of Physics, Nanjing University, Nanjing 210093, China
| | - Jianbo Huang
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaotong Huang
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chong Cheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Juan Tu
- School of Physics, Nanjing University, Nanjing 210093, China
| | - Dong Zhang
- School of Physics, Nanjing University, Nanjing 210093, China.
| | - Qiang Lu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Lang Ma
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
29
|
Lee LC, Lo KK. Leveraging the Photofunctions of Transition Metal Complexes for the Design of Innovative Phototherapeutics. SMALL METHODS 2024; 8:e2400563. [PMID: 39319499 PMCID: PMC11579581 DOI: 10.1002/smtd.202400563] [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: 04/19/2024] [Revised: 08/03/2024] [Indexed: 09/26/2024]
Abstract
Despite the advent of various medical interventions for cancer treatment, the disease continues to pose a formidable global health challenge, necessitating the development of new therapeutic approaches for more effective treatment outcomes. Photodynamic therapy (PDT), which utilizes light to activate a photosensitizer to produce cytotoxic reactive oxygen species (ROS) for eradicating cancer cells, has emerged as a promising approach for cancer treatment due to its high spatiotemporal precision and minimal invasiveness. However, the widespread clinical use of PDT faces several challenges, including the inefficient production of ROS in the hypoxic tumor microenvironment, the limited penetration depth of light in biological tissues, and the inadequate accumulation of photosensitizers at the tumor site. Over the past decade, there has been increasing interest in the utilization of photofunctional transition metal complexes as photosensitizers for PDT applications due to their intriguing photophysical and photochemical properties. This review provides an overview of the current design strategies used in the development of transition metal complexes as innovative phototherapeutics, aiming to address the limitations associated with PDT and achieve more effective treatment outcomes. The current challenges and future perspectives on the clinical translation of transition metal complexes are also discussed.
Collapse
Affiliation(s)
- Lawrence Cho‐Cheung Lee
- Department of ChemistryCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
| | - Kenneth Kam‐Wing Lo
- Department of ChemistryCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
- State Key Laboratory of Terahertz and Millimeter WavesCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
| |
Collapse
|
30
|
Liu YQ, Chao YC, Xu SQ, Peng YR, Syu JJ, Yang XH, Pan YK, Lin PC, Weng LL, Chen IC, Tan KT. Surface Functionalization of Gold Nanoparticles Using Alkyne Derivatives: Applications in Chemical Sensing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58262-58273. [PMID: 39425641 PMCID: PMC11533169 DOI: 10.1021/acsami.4c12063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024]
Abstract
Colloidal gold nanoparticles (AuNPs) are important nanomaterials for chemical sensing and therapeutics. For their application, it is vital to develop a reliable and robust surface functionalization method that can be applied to diverse functional molecules and offer better stability under harsh biological conditions. Currently, thiol (SH) is the most commonly used functional group for forming stable covalent bonds with AuNPs. However, thiolated molecules typically require complicated preparation procedures, are susceptible to oxidation, and are not compatible with many electrophiles and reducing groups. In this study, we report that surface functionalization of AuNPs can be achieved using alkyne derivatives, which exhibit several advantages over classical thiolation and peptide-bond methods, including straightforward preparation of alkyne derivatives, rapid and simple conjugation in buffers and complex media, higher conjugation efficiency, long-term stability, and resistance to decomposition under harsh conditions. Several alkynylated biotin and fluorescein derivatives are prepared, and the alkynylated-AuNPs are characterized using a lateral flow assay, gel electrophoresis, and spectroscopy techniques to investigate the conjugation efficiencies, size distributions, protein interaction properties, and binding mode of the Au-alkyne bond. We also demonstrate that alkynylated-AuNPs can be used for the sensitive detection of hydrogen peroxide and streptavidin proteins.
Collapse
Affiliation(s)
- Yun-Qiao Liu
- Department
of Chemistry, National Tsing Hua University, 101 Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Yi-Cheng Chao
- Department
of Chemistry, National Tsing Hua University, 101 Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Shun-Qiang Xu
- Department
of Chemistry, National Tsing Hua University, 101 Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Yun-Rong Peng
- Department
of Chemistry, National Tsing Hua University, 101 Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Jhih-Jie Syu
- Department
of Chemistry, National Tsing Hua University, 101 Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Xiang-He Yang
- Department
of Chemistry, National Tsing Hua University, 101 Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Yung-Kun Pan
- Department
of Chemistry, National Tsing Hua University, 101 Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Po-Cheng Lin
- Department
of Chemistry, National Tsing Hua University, 101 Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Ling-Ling Weng
- Department
of Chemistry, National Tsing Hua University, 101 Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - I-Chia Chen
- Department
of Chemistry, National Tsing Hua University, 101 Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Kui-Thong Tan
- Department
of Chemistry, National Tsing Hua University, 101 Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
- Department
of Medicinal and Applied Chemistry, Kaohsiung
Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
31
|
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
|
32
|
Dong F, Hao L, Wang L, Huang Y. Clickable nanozyme enhances precise colonization of probiotics for ameliorating inflammatory bowel disease. J Control Release 2024; 373:749-765. [PMID: 39084465 DOI: 10.1016/j.jconrel.2024.07.064] [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: 04/11/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Convincing evidence suggests that aberrant gut microbiota changes play a critical role in the progression and pathogenesis of inflammatory bowel disease (IBD). Probiotic therapeutic interventions targeting the microbiota may provide alternative avenues to treat IBD, but currently available probiotics often suffer from low intestinal colonization and limited targeting capability. Here, we developed azido (N3)-modified Prussian blue nanozyme (PB@N3) spatio-temporal guidance enhances the targeted colonization of probiotics to alleviate intestinal inflammation. First, clickable PB@N3 targets intestinal inflammation, simultaneously, it scavenges reactive oxygen species (ROS). Subsequently, utilizing "click" chemistry to spatio-temporally guide targeted colonization of dibenzocyclooctyne (DBCO)-modified Lactobacillus reuteri DSM 17938 (LR@DBCO). The "click" reaction between PB@N3 and LR@DBCO has excellent specificity and efficacy both in vivo and in vitro. Despite the complex physiological environment of IBD, "click" reaction can prolong the retention time of probiotics in the intestine. Dextran sulfate sodium (DSS)-induced colitis mice model, demonstrates that the combination of PB@N3 and LR@DBCO effectively mitigates levels of ROS, enhances the colonization of probiotics, modulates intestinal flora composition and function, regulates immune profiles, restores intestinal barrier function, and alleviates intestinal inflammation. Hence, PB@N3 spatio-temporal guidance enhances targeted colonization of LR@DBCO provides a promising medical treatment strategy for IBD.
Collapse
Affiliation(s)
- Fang Dong
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Liangwen Hao
- The Institute for Biomedical Engineering and Nano Science School of Medicine, Tongji University, Shanghai 200072, China
| | - Lin Wang
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ying Huang
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai 201102, China.
| |
Collapse
|
33
|
Zhang Y, Lei F, Qian W, Zhang C, Wang Q, Liu C, Ji H, Liu Z, Wang F. Designing intelligent bioorthogonal nanozymes: Recent advances of stimuli-responsive catalytic systems for biomedical applications. J Control Release 2024; 373:929-951. [PMID: 39097195 DOI: 10.1016/j.jconrel.2024.07.073] [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: 04/29/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Bioorthogonal nanozymes have emerged as a potent tool in biomedicine due to their unique ability to perform enzymatic reactions that do not interfere with native biochemical processes. The integration of stimuli-responsive mechanisms into these nanozymes has further expanded their potential, allowing for controlled activation and targeted delivery. As such, intelligent bioorthogonal nanozymes have received more and more attention in developing therapeutic approaches. This review provides a comprehensive overview of the recent advances in the development and application of stimuli-responsive bioorthogonal nanozymes. By summarizing the design outlines for anchoring bioorthogonal nanozymes with stimuli-responsive capability, this review seeks to offer valuable insights and guidance for the rational design of these remarkable materials. This review highlights the significant progress made in this exciting field with different types of stimuli and the various applications. Additionally, it also examines the current challenges and limitations in the design, synthesis, and application of these systems, and proposes potential solutions and research directions. This review aims to stimulate further research toward the development of more efficient and versatile stimuli-responsive bioorthogonal nanozymes for biomedical applications.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Fang Lei
- School of Public Health, Nantong University, Nantong 226019, China
| | - Wanlong Qian
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Chengfeng Zhang
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China
| | - Qi Wang
- School of Public Health, Nantong University, Nantong 226019, China
| | - Chaoqun Liu
- School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Haiwei Ji
- School of Public Health, Nantong University, Nantong 226019, China
| | - Zhengwei Liu
- Precision Immunology Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York 10029, USA.
| | - Faming Wang
- School of Public Health, Nantong University, Nantong 226019, China.
| |
Collapse
|
34
|
Zahednezhad F, Allahyari S, Sarfraz M, Zakeri-Milani P, Feyzizadeh M, Valizadeh H. Liposomal drug delivery systems for organ-specific cancer targeting: early promises, subsequent problems, and recent breakthroughs. Expert Opin Drug Deliv 2024; 21:1363-1384. [PMID: 39282895 DOI: 10.1080/17425247.2024.2394611] [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: 03/22/2024] [Accepted: 08/16/2024] [Indexed: 10/02/2024]
Abstract
INTRODUCTION Targeted liposomal systems for cancer intention have been recognized as a specific and robust approach compared to conventional liposomal delivery systems. Cancer cells have a unique microenvironment with special over-expressed receptors on their surface, providing opportunities for discovering novel and effective drug delivery systems using active targeting. AREAS COVERED Smartly targeted liposomes, responsive to internal or external stimulations, enhance the delivery efficiency by increasing accumulation of the encapsulated anti-cancer agent in the tumor site. The application of antibodies and aptamers against the prevalent cell surface receptors is a potent and ever-growing field. Moreover, immuno-liposomes and cancer vaccines as adjuvant chemotherapy are also amenable to favorable immune modulation. Combinational and multi-functional systems are also attractive in this regard. However, potentially active targeted liposomal drug delivery systems have a long path to clinical acceptance, chiefly due to cross-interference and biocompatibility affairs of the functionalized moieties. EXPERT OPINION Engineered liposomal formulations have to be designed based on tissue properties, including surface chemistry, charge, and microvasculature. In this paper, we aimed to investigate the updated targeted liposomal systems for common cancer therapy worldwide.
Collapse
Affiliation(s)
- Fahimeh Zahednezhad
- Student Research Committee and Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
| | - Saeideh Allahyari
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Parvin Zakeri-Milani
- Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Feyzizadeh
- Student Research Committee and Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
| | - Hadi Valizadeh
- Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
| |
Collapse
|
35
|
Lee LCC, Lo KKW. Shining New Light on Biological Systems: Luminescent Transition Metal Complexes for Bioimaging and Biosensing Applications. Chem Rev 2024; 124:8825-9014. [PMID: 39052606 PMCID: PMC11328004 DOI: 10.1021/acs.chemrev.3c00629] [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: 07/27/2024]
Abstract
Luminescence imaging is a powerful and versatile technique for investigating cell physiology and pathology in living systems, making significant contributions to life science research and clinical diagnosis. In recent years, luminescent transition metal complexes have gained significant attention for diagnostic and therapeutic applications due to their unique photophysical and photochemical properties. In this Review, we provide a comprehensive overview of the recent development of luminescent transition metal complexes for bioimaging and biosensing applications, with a focus on transition metal centers with a d6, d8, and d10 electronic configuration. We elucidate the structure-property relationships of luminescent transition metal complexes, exploring how their structural characteristics can be manipulated to control their biological behavior such as cellular uptake, localization, biocompatibility, pharmacokinetics, and biodistribution. Furthermore, we introduce the various design strategies that leverage the interesting photophysical properties of luminescent transition metal complexes for a wide variety of biological applications, including autofluorescence-free imaging, multimodal imaging, organelle imaging, biological sensing, microenvironment monitoring, bioorthogonal labeling, bacterial imaging, and cell viability assessment. Finally, we provide insights into the challenges and perspectives of luminescent transition metal complexes for bioimaging and biosensing applications, as well as their use in disease diagnosis and treatment evaluation.
Collapse
Affiliation(s)
- Lawrence Cho-Cheung Lee
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Units 1503-1511, 15/F, Building 17W, Hong Kong Science Park, New Territories, Hong Kong, P. R. China
| | - Kenneth Kam-Wing Lo
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
- State Key Laboratory of Terahertz and Millimeter Waves, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
| |
Collapse
|
36
|
Liu Y, Wang S, Zhang J, Sun Q, Xiao Y, Chen J, Yao M, Zhang G, Huang Q, Zhao T, Huang Q, Shi X, Feng C, Ai K, Bai Y. Reprogramming the myocardial infarction microenvironment with melanin-based composite nanomedicines in mice. Nat Commun 2024; 15:6651. [PMID: 39103330 PMCID: PMC11300711 DOI: 10.1038/s41467-024-50854-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/24/2024] [Indexed: 08/07/2024] Open
Abstract
Myocardial infarction (MI) has a 5-year mortality rate of more than 50% due to the lack of effective treatments. Interactions between cardiomyocytes and the MI microenvironment (MIM) can determine the progression and fate of infarcted myocardial tissue. Here, a specially designed Melanin-based composite nanomedicines (MCN) is developed to effectively treat MI by reprogramming the MIM. MCN is a nanocomposite composed of polydopamine (P), Prussian blue (PB) and cerium oxide (CexOy) with a Mayuan-like structure, which reprogramming the MIM by the efficient conversion of detrimental substances (H+, reactive oxygen species, and hypoxia) into beneficial status (O2 and H2O). In coronary artery ligation and ischemia reperfusion models of male mice, intravenously injecting MCN specifically targets the damaged area, resulting in restoration of cardiac function. With its promising therapeutic effects, MCN constitutes a new agent for MI treatment and demonstrates potential for clinical application.
Collapse
Affiliation(s)
- Yamei Liu
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Shuya Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China
| | - Jiaxiong Zhang
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Quan Sun
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Yi Xiao
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Jing Chen
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Meilian Yao
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Guogang Zhang
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Qun Huang
- Department of Child Health Care, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, Hunan, P.R. China
| | - Tianjiao Zhao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China
| | - Qiong Huang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Xiaojing Shi
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Can Feng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China.
| | - Yongping Bai
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China.
| |
Collapse
|
37
|
Gao P, Duan Z, Xu G, Gong Q, Wang J, Luo K, Chen J. Harnessing and Mimicking Bacterial Features to Combat Cancer: From Living Entities to Artificial Mimicking Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405075. [PMID: 39136067 DOI: 10.1002/adma.202405075] [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: 04/08/2024] [Revised: 07/23/2024] [Indexed: 08/29/2024]
Abstract
Bacterial-derived micro-/nanomedicine has garnered considerable attention in anticancer therapy, owing to the unique natural features of bacteria, including specific targeting ability, immunogenic benefits, physicochemical modifiability, and biotechnological editability. Besides, bacterial components have also been explored as promising drug delivery vehicles. Harnessing these bacterial features, cutting-edge physicochemical and biotechnologies have been applied to attenuated tumor-targeting bacteria with unique properties or functions for potent and effective cancer treatment, including strategies of gene-editing and genetic circuits. Further, the advent of bacteria-inspired micro-/nanorobots and mimicking artificial systems has furnished fresh perspectives for formulating strategies for developing highly efficient drug delivery systems. Focusing on the unique natural features and advantages of bacteria, this review delves into advances in bacteria-derived drug delivery systems for anticancer treatment in recent years, which has experienced a process from living entities to artificial mimicking systems. Meanwhile, a summary of relative clinical trials is provided and primary challenges impeding their clinical application are discussed. Furthermore, future directions are suggested for bacteria-derived systems to combat cancer.
Collapse
Affiliation(s)
- Peng Gao
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenyu Duan
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Gang Xu
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiyong Gong
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, 361000, China
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Kui Luo
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Jie Chen
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
38
|
Li M, Jia L, Zhu A, Li J, Li J, Liu X, Xie X. Engineered Leukocyte Biomimetic Colorimetric Sensor Enables High-Efficient Detection of Tumor Cells Based on Bioorthogonal Chemistry. ACS APPLIED MATERIALS & INTERFACES 2024; 16:36106-36116. [PMID: 38955781 DOI: 10.1021/acsami.4c06272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Accurate detection of heterogeneous circulating tumor cells (CTCs) is critical as they can make tumor cells more aggressive, drug-resistant, and metastasizing. Although the leukocyte membrane coating strategy is promising in meeting the challenge of detecting heterogeneous CTCs due to its inherent antiadhesive properties, it is still limited by the reduction or loss of expression of known markers. Bioorthogonal glycol-metabolic engineering is expected to break down this barrier by feeding the cells with sugar derivatives with a unique functional group to establish artificial targets on the surface of tumor cells. Herein, an engineered leukocyte biomimetic colorimetric sensor was accordingly fabricated for high-efficient detection of heterogeneous CTCs. Compared with conventional leukocyte membrane coating, the sensor could covalently bound to the heterogeneous CTCs models fed with Ac4ManNAz in vitro through the synergy of bioorthogonal chemistry and metabolic glycoengineering, ignoring the phenotypic changes of heterogeneous CTCs. Meanwhile, a sandwich structure composed of leukocyte biomimetic layer/CTCs/MoS2 nanosheet was formed for visual detection of HeLa cells as low as 10 cells mL-1. Overall, this approach can overcome the dependence of conventional cell membrane biomimetic technology on specific cell phenotypes and provide a new viewpoint to highly efficiently detect heterogeneous CTCs.
Collapse
Affiliation(s)
- Min Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an 710061, China
| | - Lanlan Jia
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an 710061, China
| | - Aihong Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jiaqi Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an 710061, China
| | - Jing Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xia Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoyu Xie
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China
| |
Collapse
|
39
|
Hua Y, Zou Z, Prescimone A, Ward TR, Mayor M, Köhler V. NSPs: chromogenic linkers for fast, selective, and irreversible cysteine modification. Chem Sci 2024; 15:10997-11004. [PMID: 39027294 PMCID: PMC11253191 DOI: 10.1039/d4sc01710b] [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: 03/13/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
The addition of a sulfhydryl group to water-soluble N-alkyl(o-nitrostyryl)pyridinium ions (NSPs) followed by fast and irreversible cyclization and aromatization results in a stable S-C sp2-bond. The reaction sequence, termed Click & Lock, engages accessible cysteine residues under the formation of N-hydroxy indole pyridinium ions. The accompanying red shift of >70 nm to around 385 nm enables convenient monitoring of the labeling yield by UV-vis spectroscopy at extinction coefficients of ≥2 × 104 M-1 cm-1. The versatility of the linker is demonstrated in the stapling of peptides and the derivatization of proteins, including the modification of reduced trastuzumab with Val-Cit-PAB-MMAE. The high stability of the linker in human plasma, fast reaction rates (k app up to 4.4 M-1 s-1 at 20 °C), high selectivity for cysteine, favorable solubility of the electrophilic moiety and the bathochromic properties of the Click & Lock reaction provide an appealing alternative to existing methods for cysteine conjugation.
Collapse
Affiliation(s)
- Yong Hua
- Department of Chemistry, University of Basel St. Johannsring 19 CH-4056 Basel Switzerland
- Department of Chemistry, University of Basel Mattenstrasse 22 CH-4058 Basel Switzerland
| | - Zhi Zou
- Department of Chemistry, University of Basel St. Johannsring 19 CH-4056 Basel Switzerland
- Department of Chemistry, University of Basel Mattenstrasse 22 CH-4058 Basel Switzerland
| | - Alessandro Prescimone
- Department of Chemistry, University of Basel St. Johannsring 19 CH-4056 Basel Switzerland
- Department of Chemistry, University of Basel Mattenstrasse 22 CH-4058 Basel Switzerland
| | - Thomas R Ward
- Department of Chemistry, University of Basel St. Johannsring 19 CH-4056 Basel Switzerland
- Department of Chemistry, University of Basel Mattenstrasse 22 CH-4058 Basel Switzerland
- National Center of Competence in Research (NCCR) "Molecular Systems Engineering" 4058 Basel Switzerland
| | - Marcel Mayor
- Department of Chemistry, University of Basel St. Johannsring 19 CH-4056 Basel Switzerland
- Department of Chemistry, University of Basel Mattenstrasse 22 CH-4058 Basel Switzerland
- Institute for Nanotechnology (INT) and Karlsruhe Nano Micro Facility (KNMFi) Karlsruhe Institute of Technology (KIT) P.O. Box 3640 DE-76021 Karlsruhe Eggenstein-Leopoldshafen Germany
- Lehn Institute of Functional Materials (LIFM), School of Chemistry, Sun Yat-Sen University (SYSU) XinGangXi Road 135 510275 Guangzhou P. R. China
| | - Valentin Köhler
- Department of Chemistry, University of Basel St. Johannsring 19 CH-4056 Basel Switzerland
- Department of Chemistry, University of Basel Mattenstrasse 22 CH-4058 Basel Switzerland
| |
Collapse
|
40
|
Li Y, Shan S, Zhang R, Sun C, Hu X, Fan J, Wang Y, Duan R, Gao M. Imaging and Downstaging Bladder Cancer with the 177Lu-Labeled Bioorthogonal Nanoprobe. ACS NANO 2024; 18:17209-17217. [PMID: 38904444 DOI: 10.1021/acsnano.4c04303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Efforts on bladder cancer treatment have been shifting from extensive surgery to organ preservation in the past decade. To this end, we herein develop a multifunctional nanoagent for bladder cancer downstaging and bladder-preserving therapy by integrating mucosa penetration, reduced off-target effects, and internal irradiation therapy into a nanodrug. Specifically, an iron oxide nanoparticle was used as a carrier that was coated with hyaluronic acid (HA) for facilitating mucosa penetration. Dibenzocyclooctyne (DBCO) was introduced into the HA coating layer to react through bioorthogonal reaction with azide as an artificial receptor of bladder cancer cells, to improve the cellular internalization of the nanoprobe labeled with 177Lu. Through magnetic resonance imaging, the targeted imaging of both nonmuscle-invasive bladder cancer (NMIBC) and muscle-invasive bladder cancer (MIBC) was realized after intravesical instillation of the multifunctional probe, both NMIBC and MIBC were found downstaged, and the metastasis was inhibited, which demonstrates the potential of the multifunctional nanoprobe for bladder preservation in bladder cancer treatment.
Collapse
Affiliation(s)
- Yueping Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Shanshan Shan
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Ruru Zhang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Chaoping Sun
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Xuelan Hu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Jiada Fan
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Yi Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Ruixue Duan
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
| | - Mingyuan Gao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, P. R. China
- Clinical Translation Center of State Key Lab, the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
41
|
Ling B, Gungoren B, Yao Y, Dutka P, Vassallo R, Nayak R, Smith CAB, Lee J, Swift MB, Shapiro MG. Truly Tiny Acoustic Biomolecules for Ultrasound Imaging and Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307106. [PMID: 38409678 PMCID: PMC11602542 DOI: 10.1002/adma.202307106] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 02/01/2024] [Indexed: 02/28/2024]
Abstract
Nanotechnology offers significant advantages for medical imaging and therapy, including enhanced contrast and precision targeting. However, integrating these benefits into ultrasonography is challenging due to the size and stability constraints of conventional bubble-based agents. Here bicones, truly tiny acoustic contrast agents based on gas vesicles (GVs), a unique class of air-filled protein nanostructures naturally produced in buoyant microbes, are described. It is shown that these sub-80 nm particles can be effectively detected both in vitro and in vivo, infiltrate tumors via leaky vasculature, deliver potent mechanical effects through ultrasound-induced inertial cavitation, and are easily engineered for molecular targeting, prolonged circulation time, and payload conjugation.
Collapse
Affiliation(s)
- Bill Ling
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Bilge Gungoren
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Yuxing Yao
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Przemysław Dutka
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Reid Vassallo
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Rohit Nayak
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Cameron A. B. Smith
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Justin Lee
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Margaret B. Swift
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Mikhail G. Shapiro
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
- Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA, USA
- Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA, USA
| |
Collapse
|
42
|
Blanco-Gómez A, Díaz-Abellás M, Montes de Oca I, Peinador C, Pazos E, García MD. Host-Guest Stimuli-Responsive Click Chemistry. Chemistry 2024; 30:e202400743. [PMID: 38597381 DOI: 10.1002/chem.202400743] [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: 02/23/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/11/2024]
Abstract
Click chemistry has reached its maturity as the weapon of choice for the irreversible ligation of molecular fragments, with over 20 years of research resulting in the development or improvement of highly efficient kinetically controlled conjugation reactions. Nevertheless, traditional click reactions can be disadvantageous not only in terms of efficiency (side products, slow kinetics, air/water tolerance, etc.), but also because they completely avoid the possibility to reversibly produce and control bound/unbound states. Recently, non-covalent click chemistry has appeared as a more efficient alternative, in particular by using host-guest self-assembled systems of high thermodynamic stability and kinetic lability. This review discusses the implementation of molecular switches in the development of such non-covalent ligation processes, resulting in what we have termed stimuli-responsive click chemistry, in which the bound/unbound constitutional states of the system can be favored by external stimulation, in particular using host-guest complexes. As we exemplify with handpicked selected examples, these supramolecular systems are well suited for the development of human-controlled molecular conjugation, by coupling thermodynamically regulated processes with appropriate temporally resolved extrinsic control mechanisms, thus mimicking nature and advancing our efforts to develop a more function-oriented chemical synthesis.
Collapse
Affiliation(s)
- Arturo Blanco-Gómez
- CICA - Centro Interdisciplinar de Química e Bioloxía and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, A Coruña, 15071, A Coruña, Spain
| | - Mauro Díaz-Abellás
- CICA - Centro Interdisciplinar de Química e Bioloxía and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, A Coruña, 15071, A Coruña, Spain
| | - Iván Montes de Oca
- CICA - Centro Interdisciplinar de Química e Bioloxía and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, A Coruña, 15071, A Coruña, Spain
| | - Carlos Peinador
- CICA - Centro Interdisciplinar de Química e Bioloxía and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, A Coruña, 15071, A Coruña, Spain
| | - Elena Pazos
- CICA - Centro Interdisciplinar de Química e Bioloxía and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, A Coruña, 15071, A Coruña, Spain
| | - Marcos D García
- CICA - Centro Interdisciplinar de Química e Bioloxía and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, A Coruña, 15071, A Coruña, Spain
| |
Collapse
|
43
|
Guerricchio L, Barile L, Bollini S. Evolving Strategies for Extracellular Vesicles as Future Cardiac Therapeutics: From Macro- to Nano-Applications. Int J Mol Sci 2024; 25:6187. [PMID: 38892376 PMCID: PMC11173118 DOI: 10.3390/ijms25116187] [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: 04/12/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Cardiovascular disease represents the foremost cause of mortality and morbidity worldwide, with a steadily increasing incidence due to the growth of the ageing population. Cardiac dysfunction leading to heart failure may arise from acute myocardial infarction (MI) as well as inflammatory- and cancer-related chronic cardiomyopathy. Despite pharmacological progress, effective cardiac repair represents an unmet clinical need, with heart transplantation being the only option for end-stage heart failure. The functional profiling of the biological activity of extracellular vesicles (EVs) has recently attracted increasing interest in the field of translational research for cardiac regenerative medicine. The cardioprotective and cardioactive potential of human progenitor stem/cell-derived EVs has been reported in several preclinical studies, and EVs have been suggested as promising paracrine therapy candidates for future clinical translation. Nevertheless, some compelling aspects must be properly addressed, including optimizing delivery strategies to meet patient needs and enhancing targeting specificity to the cardiac tissue. Therefore, in this review, we will discuss the most relevant aspects of the therapeutic potential of EVs released by human progenitors for cardiovascular disease, with a specific focus on the strategies that have been recently implemented to improve myocardial targeting and administration routes.
Collapse
Affiliation(s)
- Laura Guerricchio
- Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy;
| | - Lucio Barile
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, CH-6500 Bellinzona, Switzerland;
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera Italiana, CH-6900 Lugano, Switzerland
| | - Sveva Bollini
- Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy;
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| |
Collapse
|
44
|
Scott C, Miller S, Moenne-Loccoz P, Barnes C, Ralle M. Biocompatible Cobalt Oxide Nanoparticles for X-ray Fluorescence Microscopy. RESEARCH SQUARE 2024:rs.3.rs-4312367. [PMID: 38883752 PMCID: PMC11177975 DOI: 10.21203/rs.3.rs-4312367/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The synthesis of water-soluble nanoparticles is a well-developed field for ferrite-based nanoparticles with the majority consisting of iron oxide or mixed metal iron oxide nanoparticles. However, the synthesis of non-agglomerated non-ferrite metal/metal oxide NPs is not as well established. The synthesis and characterization of uniform 20 nm, biologically compatible cobalt oxide (CoO) nanoparticles (NPs) is described. These nanoparticles have two principle components: 1) a CoO core of suitable size to contain enough cobalt atoms to be visualized by X-ray fluorescence microscopy (XFM) and 2) a robust coating that inhibits NP aggregation as well as renders them water-soluble and biocompatible (i.e. stealth coatings). Stable cobalt oxide NPs are obtained with octadecyl amine coatings as reported by Bhattacharjee. Two strategies for solubilizing these NPs in water were investigated with varying degrees of success. Exchanging the octadecyl amine coating for a nitrodopamine anchored PEG coating yielded the desired water-soluble NPs but in very low yield. Alternately, leaving the octadecyl amine coating on the NP and interdigitating this with a maleic anhydride-vinyl copolymer with different hydrophobic sidechains followed by opening the maleic anhydride ring with amine substituted PEG polymers (the water solubilizing component), yielded the desired water soluble NPS were obtained in good yield. Characterization data for the nanoparticles and the components of the coatings required for bioorthogonal reactions to ligate them with biotargeting agents are also described.
Collapse
|
45
|
Darguzyte M, Rama E, Rix A, Baier J, Hermann J, Rezvantalab S, Khedri M, Jankowski J, Kiessling F. Riboflavin-targeted polymers improve tolerance of paclitaxel while maintaining therapeutic efficacy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 58:102751. [PMID: 38705222 DOI: 10.1016/j.nano.2024.102751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 03/15/2024] [Accepted: 04/26/2024] [Indexed: 05/07/2024]
Abstract
Active targeting can enhance precision and efficacy of drug delivery systems (DDS) against cancers. Riboflavin (RF) is a promising ligand for active targeting due to its biocompatibility and high riboflavin-receptor expression in cancers. In this study, RF-targeted 4-arm polyethylene glycol (PEG) stars conjugated with Paclitaxel (PTX), named PEG PTX RF, were evaluated as a targeted DDS. In vitro, PEG PTX RF exhibited higher toxicity against tumor cells compared to the non-targeted counterpart (PEG PTX), while free PTX displayed the highest acute toxicity. In vivo, all treatments were similarly effective, but PEG PTX RF-treated tumors showed fewer proliferating cells, pointing to sustained therapy effects. Moreover, PTX-treated animals' body and liver weights were significantly reduced, whereas both remained stable in PEG PTX and PEG PTX RF-treated animals. Overall, our targeted and non-targeted DDS reduced PTX's adverse effects, with RF targeting promoted drug uptake in cancer cells for sustained therapeutic effect.
Collapse
Affiliation(s)
- Milita Darguzyte
- Institute for Experimental Molecular Imaging, University Hospital Aachen, Forckenbeckstrasse 55, 52074 Aachen, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf, Center for Molecular Medicine Cologne, 50931 Cologne, Germany; Institute for Translational Immune-Oncology, Cancer Research Center Cologne-Essen (CCCE), University of Cologne, 50931 Cologne, Germany
| | - Elena Rama
- Institute for Experimental Molecular Imaging, University Hospital Aachen, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Anne Rix
- Institute for Experimental Molecular Imaging, University Hospital Aachen, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Jasmin Baier
- Institute for Experimental Molecular Imaging, University Hospital Aachen, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Juliane Hermann
- Institute of Molecular Cardiovascular Research, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Sima Rezvantalab
- Department of Chemical Engineering, Urmia University of Technology, 57166-419, Urmia, Iran
| | - Mohammad Khedri
- Computational Biology and Chemistry Group (CBCG) Universal Scientific Education and Research Network (USERN), 19839-63113 Tehran, Iran
| | - Joachim Jankowski
- Institute of Molecular Cardiovascular Research, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, University Hospital Aachen, Forckenbeckstrasse 55, 52074 Aachen, Germany; Fraunhofer MEVIS, Institute for Medical Image Computing, Forckenbeckstrasse 55, 52074 Aachen, Germany.
| |
Collapse
|
46
|
Yu W, Lu X, Xiong L, Teng J, Chen C, Li B, Liao BQ, Lin H, Shen L. Thiol-Ene Click Reaction in Constructing Liquid Separation Membranes for Water Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310799. [PMID: 38213014 DOI: 10.1002/smll.202310799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/25/2023] [Indexed: 01/13/2024]
Abstract
In the evolving landscape of water treatment, membrane technology has ascended to an instrumental role, underscored by its unmatched efficacy and ubiquity. Diverse synthesis and modification techniques are employed to fabricate state-of-the-art liquid separation membranes. Click reactions, distinguished by their rapid kinetics, minimal byproduct generation, and simple reaction condition, emerge as a potent paradigm for devising eco-functional materials. While the metal-free thiol-ene click reaction is acknowledged as a viable approach for membrane material innovation, a systematic elucidation of its applicability in liquid separation membrane development remains conspicuously absent. This review elucidates the pre-functionalization strategies of substrate materials tailored for thiol-ene reactions, notably highlighting thiolation and introducing unsaturated moieties. The consequential implications of thiol-ene reactions on membrane properties-including trade-off effect, surface wettability, and antifouling property-are discussed. The application of thiol-ene reaction in fabricating various liquid separation membranes for different water treatment processes, including wastewater treatment, oil/water separation, and ion separation, are reviewed. Finally, the prospects of thiol-ene reaction in designing novel liquid separation membrane, including pre-functionalization, products prediction, and solute-solute separation membrane, are proposed. This review endeavors to furnish invaluable insights, paving the way for expanding the horizons of thiol-ene reaction application in liquid separation membrane fabrication.
Collapse
Affiliation(s)
- Wei Yu
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua, 321004, China
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, Zhejiang Normal University, Jinhua, 321004, China
| | - Xinyi Lu
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua, 321004, China
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, Zhejiang Normal University, Jinhua, 321004, China
| | - Liping Xiong
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua, 321004, China
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, Zhejiang Normal University, Jinhua, 321004, China
| | - Jiaheng Teng
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua, 321004, China
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, Zhejiang Normal University, Jinhua, 321004, China
| | - Cheng Chen
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua, 321004, China
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, Zhejiang Normal University, Jinhua, 321004, China
| | - Bisheng Li
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua, 321004, China
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, Zhejiang Normal University, Jinhua, 321004, China
| | - Bao-Qiang Liao
- Department of Chemical Engineering, Lakehead University, 955 Oliver Road, Thunder Bay, Ontario, P7B 5E1, Canada
| | - Hongjun Lin
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua, 321004, China
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, Zhejiang Normal University, Jinhua, 321004, China
| | - Liguo Shen
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua, 321004, China
- Key Laboratory of Watershed Earth Surface Processes and Ecological Security, Zhejiang Normal University, Jinhua, 321004, China
| |
Collapse
|
47
|
Xu X, Li J, Lu Y, Shan Y, Shen Z, Sun F, Zhu J, Chen W, Shi H. Extracellular Vesicles in the Repair of Bone and Cartilage Injury: From Macro‐Delivery to Micro‐Modification. ADVANCED THERAPEUTICS 2024; 7. [DOI: 10.1002/adtp.202300428] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Indexed: 01/06/2025]
Abstract
AbstractExtracellular vesicles (EVs) are intermediaries in intercellular signal transmission and material exchange and have attracted significant attention from researchers in bone and cartilage repair. These nanoscale vesicles hold immense potential in facilitating bone and cartilage repair and regeneration by regulating the microenvironment at an injury site. However, their in vivo utilization is limited by their self‐clearance and random distribution. Therefore, various delivery platforms have been developed to improve EV targeting and retention rates in target organs while achieving a controlled release of EVs. Additionally, engineering modification of EVs has been proposed to effectively enhance EVs' intrinsic targeting and drug‐loading abilities and further improve their therapeutic effects on bone and cartilage injuries. This review aims to introduce the biogenesis of EVs and their regulatory mechanisms in the microenvironment of bone and cartilage injuries and comprehensively discuss the application of EV‐delivery platforms of different materials and various EV engineering modification methods in treating bone and cartilage injuries. The review's findings can help advance EV research and develop new strategies for improving the therapy of bone and cartilage injuries.
Collapse
Affiliation(s)
- Xiangyu Xu
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| | - Jialu Li
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| | - Yi Lu
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| | - Yibo Shan
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| | - Zhiming Shen
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| | - Fei Sun
- Department of Thoracic Surgery Taizhou People's Hospital Affiliated to Nanjing Medical University Taizhou 225300 China
| | - Jianwei Zhu
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| | - Wenxuan Chen
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| | - Hongcan Shi
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| |
Collapse
|
48
|
Fan J, Gong H, Wang F, Wang L, Yu Y, Liu D, Yang W. Multiplexed electrochemical nucleic acid sensor based on visible light-mediated metal-free thiol-yne click reaction for simultaneous detection of different nucleic acid targets. Talanta 2024; 273:125856. [PMID: 38442565 DOI: 10.1016/j.talanta.2024.125856] [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: 10/13/2023] [Revised: 12/26/2023] [Accepted: 02/28/2024] [Indexed: 03/07/2024]
Abstract
Simultaneous detection of multiple tumor biomarkers with a simple and low-cost assay is crucial for early cancer detection and diagnosis. Herein, we presented a low-cost and simple assay for multiplexed detection of tumor biomarkers using a spatially separated electrodes strategy. The sensor is fabricated based on a metal-free thiol-yne click reaction, which is mediated by visible light, on commercially available indium tin oxide (ITO) electrodes. Four biomarkers, including p53 DNA, Brca2 DNA, K-ras DNA, and MicroRNA-204 RNA, were used as model analytes, and the corresponding oligonucleotide probes were modified on the desired electrode units sequentially with 530 nm irradiation light in the presence of photosensitizer Eosin Y. By this visible light-mediated coupling reaction, oligonucleotide probe densities of up to 9.2 ± 0.7 × 1010 molecules/cm2 were readily obtained on the ITO electrode surface. The proposed multiplexed E-NA sensor could detect four different nucleic acid targets concurrently without crosstalk among adjacent electrodes and was also successfully applied for detecting targets in a 20% fetal calf serum sample. The detection limits for p53 DNA, Brca2 DNA, K-ras DNA, and MicroRNA-204 RNA were 0.72 nM, 0.97 nM, 2.15 nM, and 1.73 nM, respectively. The developed approach not only has a great potential for developing cost-effective biosensors on affordable substrates for nucleic acid target detection, but also be easily extended to detect other targets by modifying the specific oligonucleotide probes anchored on the electrode.
Collapse
Affiliation(s)
- Jinlong Fan
- School of Material Science and Chemical Engineering, Harbin University of Science and Technology, Harbin 150040, China; MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Hanlin Gong
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Fan Wang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Li Wang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150001, China
| | - Yongsheng Yu
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Danqing Liu
- School of Material Science and Chemical Engineering, Harbin University of Science and Technology, Harbin 150040, China.
| | - Weiwei Yang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
49
|
da S. Santos B, Finelli FG, Spring DR. Photoredox C(2)-Arylation of Indole- and Tryptophan-Containing Biomolecules. Org Lett 2024; 26:4065-4070. [PMID: 38696591 PMCID: PMC11194849 DOI: 10.1021/acs.orglett.4c01019] [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: 03/25/2024] [Revised: 04/17/2024] [Accepted: 04/30/2024] [Indexed: 05/04/2024]
Abstract
We introduce a novel and straightforward methodology for photoredox arylation of an indole scaffold using aryldiazonium salts under mild and metal-free conditions. Our approach enables the regioselective and chemoselective introduction of several aryl groups to the C(2) position of indoles and tryptophan, even in competition with other amino acids. This approach extends to the late-stage functionalization of peptides and lysozyme, heralding the unprecedented arylation of tryptophan residues in wild-type proteins and offering broad utility in chemical biology.
Collapse
Affiliation(s)
- Bruno
M. da S. Santos
- Instituto
de Pesquisas de Produtos Naturais, Universidade
Federal do Rio de Janeiro, Rio de Janeiro 21941-599, Brazil
| | - Fernanda G. Finelli
- Instituto
de Pesquisas de Produtos Naturais, Universidade
Federal do Rio de Janeiro, Rio de Janeiro 21941-599, Brazil
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Cambridge CB2 1EW, U.K.
| | - David R. Spring
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Cambridge CB2 1EW, U.K.
| |
Collapse
|
50
|
Abstract
The recent emergence of nanomedicine has revolutionized the therapeutic landscape and necessitated the creation of more sophisticated drug delivery systems. Polymeric nanoparticles sit at the forefront of numerous promising drug delivery designs, due to their unmatched control over physiochemical properties such as size, shape, architecture, charge, and surface functionality. Furthermore, polymeric nanoparticles have the ability to navigate various biological barriers to precisely target specific sites within the body, encapsulate a diverse range of therapeutic cargo and efficiently release this cargo in response to internal and external stimuli. However, despite these remarkable advantages, the presence of polymeric nanoparticles in wider clinical application is minimal. This review will provide a comprehensive understanding of polymeric nanoparticles as drug delivery vehicles. The biological barriers affecting drug delivery will be outlined first, followed by a comprehensive description of the various nanoparticle designs and preparation methods, beginning with the polymers on which they are based. The review will meticulously explore the current performance of polymeric nanoparticles against a myriad of diseases including cancer, viral and bacterial infections, before finally evaluating the advantages and crucial challenges that will determine their wider clinical potential in the decades to come.
Collapse
Affiliation(s)
- Maximilian
A. Beach
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Umeka Nayanathara
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yanting Gao
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Changhe Zhang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yijun Xiong
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yufu Wang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Georgina K. Such
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|