1
|
Hou DY, You Q, Zhang P, Li XP, Wu JC, Wang Y, You HH, Lv MY, Wu G, Liu X, Guo P, Cheng DB, Chen X, Xu W. Cascade-Activatable Nanoprodrug System Augments Sonochemotherapy of Bladder Cancer. ACS NANO 2024; 18:35507-35519. [PMID: 39686741 DOI: 10.1021/acsnano.4c12967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/18/2024]
Abstract
Sonochemotherapy (SCT) has emerged as a powerful modality for cancer treatment by triggering excessive production of reactive oxygen species (ROS) and controlled release of chemotherapeutic agents under ultrasound. However, achieving spatiotemporally controlled release of chemotherapeutic agents during ROS generation is still an enormous challenge. In this work, we developed a cascade-activated nanoprodrug (CAN) system that utilizes a reversible covalent Schiff base mixed with a hypoxia-activatable camptothecin (CPT) prodrug. Briefly, the designed fluorinated CAN system is self-assembled into nanoparticles under aqueous conditions, which could penetrate deep tumors to offer sufficient oxygen for ultrasound-triggered ROS production. Consequently, the nanoparticles substantially exacerbated the hypoxia of the tumor microenvironment (TME) by elevating oxygen consumption. The aggravated hypoxia in turn served as a positive amplifier to boost the tumor-specific CPT release of Azo-CPT prodrug, which made up for the insufficient treatment efficacy of sonodynamic therapy (SDT). On this basis, we observed a substantial reduction, approximately 3.5-fold, in the half-maximal inhibitory concentration (IC50) of the CAN system compared to that of free CPT in bladder cancer cell lines (T24). Furthermore, the CAN system demonstrated potent antitumor efficacy with reduced side effects, resulting in regression and eradication of T24 tumors in various mouse models. In summary, the CAN system can be easily extended by incorporating different chemotherapeutic agents, showing great potential to revolutionize the clinical management paradigm of bladder cancer.
Collapse
Affiliation(s)
- Da-Yong Hou
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of PET-CT/MRI, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Qing You
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Peng Zhang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Xiang-Peng Li
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Jiong-Cheng Wu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Yueze Wang
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Hui-Hui You
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Mei-Yu Lv
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Gege Wu
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Xiao Liu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Pengyu Guo
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Wanhai Xu
- NHC Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| |
Collapse
|
2
|
Fu X, Hu X. Ultrasound-Controlled Prodrug Activation: Emerging Strategies in Polymer Mechanochemistry and Sonodynamic Therapy. ACS APPLIED BIO MATERIALS 2024; 7:8040-8058. [PMID: 38698527 PMCID: PMC11653258 DOI: 10.1021/acsabm.4c00150] [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] [Academic Contribution Register] [Received: 01/31/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
Ultrasound has gained prominence in biomedical applications due to its noninvasive nature and ability to penetrate deep tissue with spatial and temporal resolution. The burgeoning field of ultrasound-responsive prodrug systems exploits the mechanical and chemical effects of ultrasonication for the controlled activation of prodrugs. In polymer mechanochemistry, materials scientists exploit the sonomechanical effect of acoustic cavitation to mechanochemically activate force-sensitive prodrugs. On the other hand, researchers in the field of sonodynamic therapy adopt fundamentally distinct methodologies, utilizing the sonochemical effect (e.g., generation of reactive oxygen species) of ultrasound in the presence of sonosensitizers to induce chemical transformations that activate prodrugs. This cross-disciplinary review comprehensively examines these two divergent yet interrelated approaches, both of which originated from acoustic cavitation. It highlights molecular and materials design strategies and potential applications in diverse therapeutic contexts, from chemotherapy to immunotherapy and gene therapy methods, and discusses future directions in this rapidly advancing domain.
Collapse
Affiliation(s)
- Xuancheng Fu
- Department
of Chemistry, BioInspired Institute, Syracuse
University, Syracuse, New York 13244, United States
| | - Xiaoran Hu
- Department
of Chemistry, BioInspired Institute, Syracuse
University, Syracuse, New York 13244, United States
| |
Collapse
|
3
|
Zhang M, Sun D, Huang H, Yang D, Song X, Feng W, Jing X, Chen Y. Nanosonosensitizer Optimization for Enhanced Sonodynamic Disease Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409663. [PMID: 39308222 DOI: 10.1002/adma.202409663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 07/05/2024] [Revised: 08/16/2024] [Indexed: 11/16/2024]
Abstract
Low-intensity ultrasound-mediated sonodynamic therapy (SDT), which, by design, integrates sonosensitizers and molecular oxygen to generate therapeutic substances (e.g., toxic hydroxyl radicals, superoxide anions, or singlet oxygen) at disease sites, has shown enormous potential for the effective treatment of a variety of diseases. Nanoscale sonosensitizers play a crucial role in the SDT process because their structural, compositional, physicochemical, and biological characteristics are key determinants of therapeutic efficacy. In particular, advances in materials science and nanotechnology have invigorated a series of optimization strategies for augmenting the therapeutic efficacy of nanosonosensitizers. This comprehensive review systematically summarizes, discusses, and highlights state-of-the-art studies on the current achievements of nanosonosensitizer optimization in enhanced sonodynamic disease treatment, with an emphasis on the general design principles of nanosonosensitizers and their optimization strategies, mainly including organic and inorganic nanosonosensitizers. Additionally, recent advancements in optimized nanosonosensitizers for therapeutic applications aimed at treating various diseases, such as cancer, bacterial infections, atherosclerosis, and autoimmune diseases, are clarified in detail. Furthermore, the biological effects of the improved nanosonosensitizers for versatile SDT applications are thoroughly discussed. The review concludes by highlighting the current challenges and future opportunities in this rapidly evolving research field to expedite its practical clinical translation and application.
Collapse
Affiliation(s)
- Min Zhang
- Department of Ultrasound, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, P. R. China
| | - Dandan Sun
- Department of Ultrasound, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, P. R. China
| | - Hui Huang
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Dayan Yang
- Department of Ultrasound, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, P. R. China
| | - Xinran Song
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Wei Feng
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Xiangxiang Jing
- Department of Ultrasound, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, P. R. China
| | - Yu Chen
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang, 325088, P. R. China
- Shanghai Institute of Materdicine, Shanghai, 200051, P. R. China
| |
Collapse
|
4
|
Li Y, Chang P, Xu L, Zhu Z, Hu M, Cen J, Li S, Zhao YE. TiO2-Nanoparticle-Enhanced Sonodynamic Therapy for Prevention of Posterior Capsular Opacification and Ferroptosis Exploration of Its Mechanism. Invest Ophthalmol Vis Sci 2024; 65:24. [PMID: 39417751 PMCID: PMC11500051 DOI: 10.1167/iovs.65.12.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/05/2024] [Accepted: 07/30/2024] [Indexed: 10/19/2024] Open
Abstract
Purpose To explore the application and potential ferroptosis mechanisms of sonodynamic therapy (SDT) using titanium dioxide nanoparticles (TiO2-NPs) as sonosensitizers for the prevention of posterior capsule opacification (PCO). Methods We fabricated TiO2-NP-coated intraocular lenses (TiO2-IOLs) using the spin-coating method, followed by ultrasound activation of the photosensitizer TiO2. In vitro experiments were performed with human lens epithelial cells (HLECs) to explore the appropriate concentration of TiO2 and ultrasonic parameters. Investigations included reactive oxygen species (ROS) generation, glutathione (GSH) depletion, glutathione peroxidase 4 (GPX4) western blot analysis, lipid peroxidation assays, and transcriptomics analysis. Finally, TiO2-IOLs were implanted in rabbit eyes to explore the in vivo performance of SDT. Results Through both in vitro and in vivo experiments, the study determined that the ultrasound parameters of 5-minute duration, 1-MHz frequency, 50% duty cycle, and 1.2-W/cm2 intensity were reliable and valid for killing HLECs without damaging other ocular structures. In vitro experiments demonstrated that SDT generated excess ROS, which disrupted the mitochondrial membrane potential and significantly reduced the GSH content. Additionally, the downregulation of GPX4, accumulation of lipid peroxides, and alteration of mitochondrial morphology were observed, suggesting that ferroptosis may be the underlying mechanism. The RNA-sequencing analysis results also showed an increase in the expression of multiple pro-ferroptosis genes and the ferroptosis marker gene PTGS2. Animal experiments preliminarily demonstrated the safety and effectiveness of SDT in treating PCO in vivo. Conclusions TiO2-IOLs combined with SDT effectively prevented PCO by generating ROS and intracellular ferroptosis.
Collapse
Affiliation(s)
- Yuanyuan Li
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| | - Pingjun Chang
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| | - Liming Xu
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| | - Zehui Zhu
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| | - Man Hu
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| | - Jiaying Cen
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| | - Siyan Li
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| | - Yun-e Zhao
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Eye Hospital of Wenzhou Medical University Hangzhou Branch, Hangzhou, China
| |
Collapse
|
5
|
Chen X, Wu D, Chen Z. Biomedical applications of stimuli-responsive nanomaterials. MedComm (Beijing) 2024; 5:e643. [PMID: 39036340 PMCID: PMC11260173 DOI: 10.1002/mco2.643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/25/2023] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/23/2024] Open
Abstract
Nanomaterials have aroused great interests in drug delivery due to their nanoscale structure, facile modifiability, and multifunctional physicochemical properties. Currently, stimuli-responsive nanomaterials that can respond to endogenous or exogenous stimulus display strong potentials in biomedical applications. In comparison with conventional nanomaterials, stimuli-responsive nanomaterials can improve therapeutic efficiency and reduce the toxicity of drugs toward normal tissues through specific targeting and on-demand drug release at pathological sites. In this review, we summarize the responsive mechanism of a variety of stimulus, including pH, redox, and enzymes within pathological microenvironment, as well as exogenous stimulus such as thermal effect, magnetic field, light, and ultrasound. After that, biomedical applications (e.g., drug delivery, imaging, and theranostics) of stimuli-responsive nanomaterials in a diverse array of common diseases, including cardiovascular diseases, cancer, neurological disorders, inflammation, and bacterial infection, are presented and discussed. Finally, the remaining challenges and outlooks of future research directions for the biomedical applications of stimuli-responsive nanomaterials are also discussed. We hope that this review can provide valuable guidance for developing stimuli-responsive nanomaterials and accelerate their biomedical applications in diseases diagnosis and treatment.
Collapse
Affiliation(s)
- Xiaojie Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang ProvinceSchool of Pharmaceutical SciencesDepartment of NeurologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| | - Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang ProvinceSchool of Pharmaceutical SciencesDepartment of NeurologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang ProvinceSchool of Pharmaceutical SciencesDepartment of NeurologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| |
Collapse
|
6
|
Yu N, Zhou J, Ding M, Li M, Peng S, Li J. Sono-Triggered Cascade Lactate Depletion by Semiconducting Polymer Nanoreactors for Cuproptosis-Immunotherapy of Pancreatic Cancer. Angew Chem Int Ed Engl 2024; 63:e202405639. [PMID: 38708791 DOI: 10.1002/anie.202405639] [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] [Academic Contribution Register] [Received: 03/22/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/07/2024]
Abstract
The high level of lactate in tumor microenvironment not only promotes tumor development and metastasis, but also induces immune escape, which often leads to failures of various tumor therapy strategies. We here report a sono-triggered cascade lactate depletion strategy by using semiconducting polymer nanoreactors (SPNLCu) for cancer cuproptosis-immunotherapy. The SPNLCu mainly contain a semiconducting polymer as sonosensitizer, lactate oxidase (LOx) conjugated via a reactive oxygen species (ROS)-cleavable linker and chelated Cu2+. Upon ultrasound (US) irradiation, the semiconducting polymer generates singlet oxygen (1O2) to cut ROS-cleavable linker to allow the release of LOx that catalyzes lactate depletion to produce hydrogen peroxide (H2O2). The Cu2+ will be reduced to Cu+ in tumor microenvironment, which reacts with the produced H2O2 to obtain hydroxyl radical (⋅OH) that further improves LOx release via destroying ROS-cleavable linkers. As such, sono-triggered cascade release of LOx achieves effective lactate depletion, thus relieving immunosuppressive roles of lactate. Moreover, the toxic Cu+ induces cuproptosis to cause immunogenic cell death (ICD) for activating antitumor immunological effect. SPNLCu are used to treat both subcutaneous and deep-tissue orthotopic pancreatic cancer with observably enhanced efficacy in restricting the tumor growths. This study thus provides a precise and effective lactate depletion tactic for cancer therapy.
Collapse
Affiliation(s)
- Ningyue Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Jianhui Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Mengbin Ding
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Meng Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Shaojun Peng
- Center for Biological Science and Technology & College of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| |
Collapse
|
7
|
Zhao P, Wang J, Huang H, Chen Z, Wang H, Lin Q. Exosomes-based dual drug-loaded nanocarrier for targeted and multiple proliferative vitreoretinopathy therapy. Regen Biomater 2024; 11:rbae081. [PMID: 39040514 PMCID: PMC11262591 DOI: 10.1093/rb/rbae081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/12/2024] [Revised: 05/31/2024] [Accepted: 06/16/2024] [Indexed: 07/24/2024] Open
Abstract
Proliferative vitreoretinopathy (PVR) is a common cause of vision loss after retinal reattachment surgery and ocular trauma. The key pathogenic mechanisms of PVR development include the proliferation, migration and epithelial-mesenchymal transition (EMT) of retinal pigment epithelial cells (RPEs) activated by the growth factors and cytokines after surgery. Although some drugs have been tried in PVR treatments as basic investigations, the limited efficacy remains an obstacle, which may be due to the single pharmacological action and lack of targeting. Herein, the anti-proliferative Daunorubicin and anti-inflammatory Dexamethasone were co-loaded in the RPEs-derived exosomes (Exos), obtaining an Exos-based dual drug-loaded nanocarrier (Exos@D-D), and used for multiple PVR therapy. Owing to the advantages of homologous Exos and the dual drug loading, Exos@D-D showed good RPEs targeting as well as improved uptake efficiency, and could inhibit the proliferation, migration, as well as EMT of RPEs effectively. The animal studies have also demonstrated that Exos@D-D effectively inhibits the production of proliferative membranes and prevents the further development of inflammation, shows significant therapeutic effects on PVR and good biocompatibility. Such Exos-based dual drug-loaded nanocarrier investigation not only provides a promising approach for multifunctional exosome drug delivery systems construction, but also has great potential in PVR clinical therapy application.
Collapse
Affiliation(s)
- Peiyi Zhao
- National Engineering Research Center of Ophthalmology and Optometry, Department of Biomaterials, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jiahao Wang
- National Engineering Research Center of Ophthalmology and Optometry, Department of Biomaterials, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Huiying Huang
- National Engineering Research Center of Ophthalmology and Optometry, Department of Biomaterials, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Zhirong Chen
- National Engineering Research Center of Ophthalmology and Optometry, Department of Biomaterials, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Hui Wang
- National Engineering Research Center of Ophthalmology and Optometry, Department of Biomaterials, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Quankui Lin
- National Engineering Research Center of Ophthalmology and Optometry, Department of Biomaterials, School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
8
|
Liu Y, Jiang Z, Yang X, Wang Y, Yang B, Fu Q. Engineering Nanoplatforms for Theranostics of Atherosclerotic Plaques. Adv Healthc Mater 2024; 13:e2303612. [PMID: 38564883 DOI: 10.1002/adhm.202303612] [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] [Academic Contribution Register] [Received: 10/20/2023] [Revised: 03/28/2024] [Indexed: 04/04/2024]
Abstract
Atherosclerotic plaque formation is considered the primary pathological mechanism underlying atherosclerotic cardiovascular diseases, leading to severe cardiovascular events such as stroke, acute coronary syndromes, and even sudden cardiac death. Early detection and timely intervention of plaques are challenging due to the lack of typical symptoms in the initial stages. Therefore, precise early detection and intervention play a crucial role in risk stratification of atherosclerotic plaques and achieving favorable post-interventional outcomes. The continuously advancing nanoplatforms have demonstrated numerous advantages including high signal-to-noise ratio, enhanced bioavailability, and specific targeting capabilities for imaging agents and therapeutic drugs, enabling effective visualization and management of atherosclerotic plaques. Motivated by these superior properties, various noninvasive imaging modalities for early recognition of plaques in the preliminary stage of atherosclerosis are comprehensively summarized. Additionally, several therapeutic strategies are proposed to enhance the efficacy of treating atherosclerotic plaques. Finally, existing challenges and promising prospects for accelerating clinical translation of nanoplatform-based molecular imaging and therapy for atherosclerotic plaques are discussed. In conclusion, this review provides an insightful perspective on the diagnosis and therapy of atherosclerotic plaques.
Collapse
Affiliation(s)
- Yuying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Zeyu Jiang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Bin Yang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
9
|
Dang J, Li Y, Yan J, Wu J, Cai K, Yin L, Zhou Z. Reversal of Chemoresistance via Staged Liberation of Chemodrug and siRNA in Hierarchical Response to ROS Gradient. Adv Healthc Mater 2024; 13:e2304130. [PMID: 38427696 DOI: 10.1002/adhm.202304130] [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] [Academic Contribution Register] [Received: 11/23/2023] [Revised: 01/29/2024] [Indexed: 03/03/2024]
Abstract
P-glycoprotein (P-gp)-mediated multidrug resistance (MDR) often leads to the failure of antitumor chemotherapy, and codelivery of chemodrug with P-gp siRNA (siP-gp) represents a promising approach for treating chemoresistant tumors. To maximize the antitumor efficacy, it is desired that the chemodrug be latently released upon completion of siP-gp-mediated gene silencing, which however, largely remains an unmet demand. Herein, core-shell nanocomplexes (NCs) are developed to overcome MDR via staged liberation of siP-gp and chemodrug (doxorubicin, Dox) in hierarchical response to reactive oxygen species (ROS) concentration gradients. The NCs are constructed from mesoporous silica nanoparticles (MSNs) surface-decorated with cRGD-modified, PEGylated, ditellurium-crosslinked polyethylenimine (RPPT), wherein thioketal-linked dimeric doxorubicin (TK-Dox2) and photosensitizer are coencapsulated inside MSNs while siP-gp is embedded in the RPPT polymeric layer. RPPT with ultrahigh ROS-sensitivity can be efficiently degraded by the low-concentration ROS inside cancer cells to trigger siP-gp release. Upon siP-gp-mediated gene silencing and MDR reversal, light irradiation is performed to generate high-concentration, lethal amount of ROS, which cleaves thioketal with low ROS-sensitivity to liberate the monomeric Dox. Such a latent release profile greatly enhances Dox accumulation in Dox-resistant cancer cells (MCF-7/ADR) in vitro and in vivo, which cooperates with the generated ROS to efficiently eradicate MCF-7/ADR xenograft tumors.
Collapse
MESH Headings
- Humans
- Reactive Oxygen Species/metabolism
- Doxorubicin/pharmacology
- Doxorubicin/chemistry
- RNA, Small Interfering/chemistry
- Drug Resistance, Neoplasm/drug effects
- Animals
- Nanoparticles/chemistry
- Mice
- Mice, Nude
- Female
- Silicon Dioxide/chemistry
- Cell Line, Tumor
- MCF-7 Cells
- Mice, Inbred BALB C
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- Drug Resistance, Multiple/drug effects
- Photosensitizing Agents/chemistry
- Photosensitizing Agents/pharmacology
Collapse
Affiliation(s)
- Juanjuan Dang
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Yongjuan Li
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Jing Yan
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Jianhua Wu
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Kaimin Cai
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign Urbana, IL, 61801, USA
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Zhuchao Zhou
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| |
Collapse
|
10
|
Dai X, Du Y, Li Y, Yan F. Nanomaterials-based precision sonodynamic therapy enhancing immune checkpoint blockade: A promising strategy targeting solid tumor. Mater Today Bio 2023; 23:100796. [PMID: 37766898 PMCID: PMC10520454 DOI: 10.1016/j.mtbio.2023.100796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/03/2023] [Revised: 08/24/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Burgeoning is an evolution from conventional photodynamic therapy (PDT). Thus, sonodynamic therapy (SDT) regulated by nanoparticles (NPs) possesses multiple advantages, including stronger penetration ability, better biological safety, and not reactive oxygen species (ROS)-dependent tumor-killing effect. However, the limitation to tumor inhibition instead of shrinkage and the incapability of eliminating metastatic tumors hinder the clinical potential for SDT. Fortunately, immune checkpoint blockade (ICB) can revive immunological function and induce a long-term immune memory against tumor rechallenges. Hence, synergizing NPs-based SDT with ICB can provide a promising therapeutic outcome for solid tumors. Herein, we briefly reviewed the progress in NPs-based SDT and ICB therapy. We highlighted the synergistic anti-tumor mechanisms and summarized the representative preclinical trials on SDT-assisted immunotherapy. Compared to other reviews, we provided comprehensive and unique perspectives on the innovative sonosensitizers in each trial. Moreover, we also discussed the current challenges and future corresponding solutions.
Collapse
Affiliation(s)
- Xinlun Dai
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Yangyang Du
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Yumei Li
- Department of Pediatric Intensive Care Unit, First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Fei Yan
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
11
|
Ding M, Zhang Y, Yu N, Zhou J, Zhu L, Wang X, Li J. Augmenting Immunogenic Cell Death and Alleviating Myeloid-Derived Suppressor Cells by Sono-Activatable Semiconducting Polymer Nanopartners for Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2302508. [PMID: 37165741 DOI: 10.1002/adma.202302508] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 03/17/2023] [Revised: 05/09/2023] [Indexed: 05/12/2023]
Abstract
Inducing immunogenic cell death (ICD) by sonodynamic therapy (SDT) is promising for cancer immunotherapy, which however is inefficient due to oxygen depletion that compromises SDT effect and mediates recruitment of immunosuppressive myeloid-derived suppressor cells (MDSCs). The fabrication of sono-activatable semiconducting polymer nanopartners (SPNTi ) to simultaneously augment ICD and alleviate MDSCs for immunotherapy is reported. A sonodynamic semiconducting polymer, hydrophobic hypoxia-responsive tirapazamine (TPZ)-conjugate, and MDSC-targeting drug (ibrutinib) are encapsulated inside such SPNTi with surface shell of a singlet oxygen (1 O2 )-cleavable amphiphilic polymer. TPZ and ibrutinib serve as drug partners to enlarge immunotherapeutic effect. Upon sono-activation, SPNTi generate 1 O2 to break 1 O2 -cleavable polymers for in situ liberations of TPZ-conjugate and ibrutinib in tumor sites, and oxygen is consumed to create severe hypoxic tumor microenvironment, in which, TPZ-conjugate is activated for augmenting ICD action, while ibrutinib alleviates MDSCs for promoting antitumor immunological effect. In a bilateral tumor mouse model, SPNTi -mediated sono-activatable immunotherapy results in growth restraints of primary and distant tumors and noteworthy precaution of tumor metastases. This study thus provides a sono-activatable immunotherapeutic strategy with high precision and safety for cancer via overcoming post-treatment hypoxia and targeting MDSCs.
Collapse
Affiliation(s)
- Mengbin Ding
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Yijing Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Ningyue Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Jianhui Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Liyun Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Xing Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| |
Collapse
|
12
|
Wang F, Pu K, Li J. Activating Nanomedicines with Electromagnetic Energy for Deep-Tissue Induction of Immunogenic Cell Death in Cancer Immunotherapy. SMALL METHODS 2023; 7:e2201083. [PMID: 36316270 DOI: 10.1002/smtd.202201083] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 08/19/2022] [Revised: 10/12/2022] [Indexed: 05/17/2023]
Abstract
Immunotherapy is an attractive approach for cancer therapy, while its antitumor efficacy is still limited, especially for non-immunogenic tumors. Nanomedicines can be utilized to convert the non-immunogenic "cold" tumors to immunogenic "hot" tumors via inducing immunogenic cell death (ICD), thereby promoting the antitumor immune response. Some nanomedicines that can produce local heat and reactive oxygen species upon the stimulation of electromagnetic energy are the main candidates for inducing the ICD effect. However, their applications are often restricted due to the poor tissue penetration depths of electromagnetic energy, such as light. By contrast, ultrasound, X-ray, alternating magnetic field, and microwave show excellent tissue penetration depths and thereby can be used for sonodynamic therapy, radiotherapy, magnetic hyperthermia therapy, and microwave ablation therapy, all of which can effectively induce ICD. Herein, the combination of deep-tissue electromagnetic energy with nanomedicines for inducing ICD and cancer immunotherapy are summarized. In particular, the designs of nanomedicines to amplify ICD effect in the presence of deep-tissue electromagnetic energy and sensitize tumors to various immunotherapies will be discussed. At the end of this review, a brief conclusion and discussion of current challenges and further perspectives in this subfield are provided.
Collapse
Affiliation(s)
- Fengshuo Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 637457, Singapore
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| |
Collapse
|
13
|
Li Y, Wu P, Zhu M, Liang M, Zhang L, Zong Y, Wan M. High-Performance Delivery of a CRISPR Interference System via Lipid-Polymer Hybrid Nanoparticles Combined with Ultrasound-Mediated Microbubble Destruction for Tumor-Specific Gene Repression. Adv Healthc Mater 2023; 12:e2203082. [PMID: 36591868 DOI: 10.1002/adhm.202203082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/28/2022] [Indexed: 01/03/2023]
Abstract
The dCas9-based CRISPR interference (CRISPRi) system efficiently silences genes without causing detectable off-target activity, thus showing great potential for the treatment of cancer at the transcriptional level. However, due to the large size of the commonly used CRISPRi system, effective delivery of the system has been a challenge that hinders its application in the clinic. Herein, a combination of pH-responsive lipid-polymer hybrid nanoparticles (PLPNs) and ultrasound-mediated microbubble destruction (UMMD) is used for the delivery of the CRISPRi system. The core-shell structure of PLPNs can effectively be loaded with the CRISPRi plasmid, and increases the time spent in the circulating in vivo, and "actively target" cancer cells. Moreover, the combination of PLPNs with UMMD achieves a higher cellular uptake of the CRISPRi plasmid in vitro and retention in vivo. Furthermore, when PLPNs loaded with a CRISPRi plasmid that targets microRNA-10b (miR-10b) are used in combination with UMMD, it results in the effective repression of miR-10b in breast cancer, simultaneous disturbance of multiple cell migration and invasion-related signaling pathways, and a significant inhibition of lung metastasis. Thus, the established system presents a versatile, highly efficient, and safe strategy for delivery of the CRISPRi system both in vitro and in vivo.
Collapse
Affiliation(s)
- Yan Li
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Pengying Wu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Mingting Zhu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Meiling Liang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Lei Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Yujin Zong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Mingxi Wan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
14
|
Liu D, Dai X, Ye L, Wang H, Qian H, Cheng H, Wang X. Nanotechnology meets glioblastoma multiforme: Emerging therapeutic strategies. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1838. [PMID: 35959642 DOI: 10.1002/wnan.1838] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 03/19/2022] [Revised: 06/24/2022] [Accepted: 07/11/2022] [Indexed: 01/31/2023]
Abstract
Glioblastoma multiforme (GBM) represents the most common and fatal form of primary invasive brain tumors as it affects a great number of patients each year and has a median overall survival of approximately 14.6 months after diagnosis. Despite intensive treatment, almost all patients with GBM experience recurrence, and their 5-year survival rate is approximately 5%. At present, the main clinical treatment strategy includes surgical resection, radiotherapy, and chemotherapy. However, tumor heterogeneity, blood-brain barrier, glioma stem cells, and DNA damage repair mechanisms hinder efficient GBM treatment. The emergence of nanometer-scale diagnostic and therapeutic approaches in cancer medicine due to the establishment of nanotechnology provides novel and promising tools that will allow us to overcome these difficulties. This review summarizes the application and recent progress in nanotechnology-based monotherapies (e.g., chemotherapy) and combination cancer treatment strategies (chemotherapy-based combined cancer therapy) for GBM and describes the synergistic enhancement between these combination therapies as well as the current standard therapy for brain cancer and its deficiencies. These combination therapies that can reduce individual drug-related toxicities and significantly enhance therapeutic efficiency have recently undergone rapid development. The mechanisms underlying these different nanotechnology-based therapies as well as the application of nanotechnology in GBM (e.g., in photodynamic therapy and chemodynamic therapy) have been systematically summarized here in an attempt to review recent developments and to identify promising directions for future research. This review provides novel and clinically significant insights and directions for the treatment of GBM, which is of great clinical importance. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > In Vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Dongdong Liu
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, China.,Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xingliang Dai
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lei Ye
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Haisheng Qian
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, China
| | - Hongwei Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, China
| |
Collapse
|
15
|
Meng X, Sun S, Gong C, Yang J, Yang Z, Zhang X, Dong H. Ag-Doped Metal-Organic Frameworks' Heterostructure for Sonodynamic Therapy of Deep-Seated Cancer and Bacterial Infection. ACS NANO 2022; 17:1174-1186. [PMID: 36583572 DOI: 10.1021/acsnano.2c08687] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 06/17/2023]
Abstract
Metal-organic frameworks (MOF) have attracted great potential in sonodynamic therapy (SDT) owing to large sonosensitizers' loading and fast reactive oxygen species' (ROS) diffusion; however, the low ligand-to-metal charge transfer efficiency sharply impairs the SDT effect. Herein, we report the design of MIL@Ag heterostructures with high electron-hole pairs separation efficiency and enhanced diverse ROS generation ability for deep-seated cancer treatment and bacterial infection. The MIL@Ag heterostructure is composed of Ti-based MOFs (named MIL), on which are in situ assembled silver nanoparticles (Ag NPs). The electrochemical experiments and density functional theory calculations verify that the introduction of Ag NPs can significantly improve the electron transfer efficiency and O2 adsorption capacity of MIL. Under ultrasound irradiation, the doped Ag NPs can trap the activated electrons from MIL to reduce surrounding O2 and produce superoxide radicals (•O2-), while the activated holes enable oxidizing H2O to produce hydroxyl radicals (•OH). Thus, they efficiently improve the therapeutic efficiency of SDT. MIL@Ag-PEG-mediated SDT implements A549 cancer cells' killing under a tissue barrier of 2 cm and eradicates the bacterial infection of Staphylococcus aureus, thus promoting wound healing. Therefore, MIL@Ag-PEG provides a promising strategy for augmenting SDT performance by rational heterostructure design of sonosensitizers.
Collapse
Affiliation(s)
- Xiangdan Meng
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Research Center for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, People's Republic of China
| | - Sirong Sun
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, People's Republic of China
| | - Chenchen Gong
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, People's Republic of China
| | - Junyan Yang
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, People's Republic of China
| | - Zhou Yang
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, People's Republic of China
| | - Xueji Zhang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Research Center for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, People's Republic of China
- Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Health Science Centre, Shenzhen University, Shenzhen 518071, People's Republic of China
| | - Haifeng Dong
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Research Center for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, People's Republic of China
- Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Health Science Centre, Shenzhen University, Shenzhen 518071, People's Republic of China
| |
Collapse
|
16
|
Lim YY, Zaidi AMA, Miskon A. Composing On-Program Triggers and On-Demand Stimuli into Biosensor Drug Carriers in Drug Delivery Systems for Programmable Arthritis Therapy. Pharmaceuticals (Basel) 2022; 15:1330. [PMID: 36355502 PMCID: PMC9698912 DOI: 10.3390/ph15111330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/21/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 08/31/2023] Open
Abstract
Medication in arthritis therapies is complex because the inflammatory progression of rheumatoid arthritis (RA) and osteoarthritis (OA) is intertwined and influenced by one another. To address this problem, drug delivery systems (DDS) are composed of four independent exogenous triggers and four dependent endogenous stimuli that are controlled on program and induced on demand, respectively. However, the relationships between the mechanisms of endogenous stimuli and exogenous triggers with pathological alterations remain unclear, which results in a major obstacle in terms of clinical translation. Thus, the rationale for designing a guidance system for these mechanisms via their key irritant biosensors is in high demand. Many approaches have been applied, although successful clinical translations are still rare. Through this review, the status quo in historical development is highlighted in order to discuss the unsolved clinical difficulties such as infiltration, efficacy, drug clearance, and target localisation. Herein, we summarise and discuss the rational compositions of exogenous triggers and endogenous stimuli for programmable therapy. This advanced active pharmaceutical ingredient (API) implanted dose allows for several releases by remote controls for endogenous stimuli during lesion infections. This solves the multiple implantation and local toxic accumulation problems by using these flexible desired releases at the specified sites for arthritis therapies.
Collapse
Affiliation(s)
- Yan Yik Lim
- Faculty of Defence Science and Technology, National Defence University of Malaysia, Sungai Besi Prime Camp, Kuala Lumpur 57000, Malaysia
| | - Ahmad Mujahid Ahmad Zaidi
- Faculty of Defence Science and Technology, National Defence University of Malaysia, Sungai Besi Prime Camp, Kuala Lumpur 57000, Malaysia
| | - Azizi Miskon
- Faculty of Engineering, National Defence University of Malaysia, Sungai Besi Prime Camp, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
17
|
Sofuni A, Itoi T. Current status and future perspective of sonodynamic therapy for cancer. J Med Ultrason (2001) 2022:10.1007/s10396-022-01263-x. [PMID: 36224458 DOI: 10.1007/s10396-022-01263-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/15/2022] [Accepted: 09/08/2022] [Indexed: 12/07/2022]
Abstract
There is a tremendous need for prevention and effective treatment of cancer due to the associated morbidity and mortality. In this study, we introduce sonodynamic therapy (SDT), which is expected to be a new cancer treatment modality. SDT is a promising option for minimally invasive treatment of solid tumors and comprises three different components: sonosensitizers, ultrasound, and molecular oxygen. These components are harmless individually, but in combination they generate cytotoxic reactive oxygen species (ROS). We will explore the molecular mechanism by which SDT kills cancer cells, the class of sonosensitizers, drug delivery methods, and in vitro and in vivo studies. At the same time, we will highlight clinical applications for cancer treatment. The progress of SDT research suggests that it has the potential to become an advanced field of cancer treatment in clinical application. In this article, we will focus on the mechanism of action of SDT and its application to cancer treatment, and explain key factors to aid in developing strategies for future SDT development.
Collapse
Affiliation(s)
- Atsushi Sofuni
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku Shinjuku-ku, Tokyo, 160-0023, Japan.
| | - Takao Itoi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku Shinjuku-ku, Tokyo, 160-0023, Japan
| |
Collapse
|
18
|
Hersh AM, Bhimreddy M, Weber-Levine C, Jiang K, Alomari S, Theodore N, Manbachi A, Tyler BM. Applications of Focused Ultrasound for the Treatment of Glioblastoma: A New Frontier. Cancers (Basel) 2022; 14:4920. [PMID: 36230843 PMCID: PMC9563027 DOI: 10.3390/cancers14194920] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/10/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/21/2022] Open
Abstract
Glioblastoma (GBM) is an aggressive primary astrocytoma associated with short overall survival. Treatment for GBM primarily consists of maximal safe surgical resection, radiation therapy, and chemotherapy using temozolomide. Nonetheless, recurrence and tumor progression is the norm, driven by tumor stem cell activity and a high mutational burden. Focused ultrasound (FUS) has shown promising results in preclinical and clinical trials for treatment of GBM and has received regulatory approval for the treatment of other neoplasms. Here, we review the range of applications for FUS in the treatment of GBM, which depend on parameters, including frequency, power, pulse duration, and duty cycle. Low-intensity FUS can be used to transiently open the blood-brain barrier (BBB), which restricts diffusion of most macromolecules and therapeutic agents into the brain. Under guidance from magnetic resonance imaging, the BBB can be targeted in a precise location to permit diffusion of molecules only at the vicinity of the tumor, preventing side effects to healthy tissue. BBB opening can also be used to improve detection of cell-free tumor DNA with liquid biopsies, allowing non-invasive diagnosis and identification of molecular mutations. High-intensity FUS can cause tumor ablation via a hyperthermic effect. Additionally, FUS can stimulate immunological attack of tumor cells, can activate sonosensitizers to exert cytotoxic effects on tumor tissue, and can sensitize tumors to radiation therapy. Finally, another mechanism under investigation, known as histotripsy, produces tumor ablation via acoustic cavitation rather than thermal effects.
Collapse
Affiliation(s)
- Andrew M. Hersh
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Meghana Bhimreddy
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Carly Weber-Levine
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kelly Jiang
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Safwan Alomari
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Nicholas Theodore
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Amir Manbachi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Mechanical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Electrical and Computer Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Betty M. Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
19
|
Fan CH, Ho YJ, Lin CW, Wu N, Chiang PH, Yeh CK. State-of-the-art of ultrasound-triggered drug delivery from ultrasound-responsive drug carriers. Expert Opin Drug Deliv 2022; 19:997-1009. [PMID: 35930441 DOI: 10.1080/17425247.2022.2110585] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The development of new tools to locally and non-invasively transferring therapeutic substances at the desired site in deep living tissue has been a long sought-after goal within the drug delivery field. Among the established methods, ultrasound (US) with US-responsive carriers holds great promise and demonstrates on-demand delivery of a variety of functional substances with spatial precision of several millimeters in deep-seated tissues in animal models and humans. These properties have motivated several explorations of US with US responsive carriers as a modality for neuromodulation and the treatment of various diseases, such as stroke and cancer. AREAS COVERED This article briefly discussed three specific mechanisms that enhance in vivo drug delivery via US with US-responsive carriers: 1) permeabilizing cellular membrane, 2) increasing the permeability of vessels, and 3) promoting cellular endocytotic uptake. Besides, a series of US-responsive drug carriers are discussed, with an emphasis on the relation between structural feature and therapeutic outcome. EXPERT OPINION This article summarized current development for each of US-responsive drug carrier, focusing on the routes of enhancing delivery and applications. The mechanisms of interaction between US-responsive carriers and US energy, such as cavitation, hyperthermia, and reactive oxygen species, as well as how these interactions can improve drug delivery into target cell/tissue. It can be expected that there are serval efforts to further identification of US-responsive particles, design of novel US waveform sequence, and survey of optimal combination between US parameters and US-responsive carriers for better controlling the spatiotemporal drug release profile, stability, and safety in vivo. The authors believe these will provide novel tools for precisely designing treatment strategies and significantly benefit the clinical management of several diseases.
Collapse
Affiliation(s)
- Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan.,Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ju Ho
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Chia-Wei Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Nan Wu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Pei-Hua Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
20
|
Wang Z, Yu N, Zhang J, Ren Q, Li M, Chen Z. Nanoscale Hf-hematoporphyrin frameworks for synergetic sonodynamic/radiation therapy of deep-seated tumors. J Colloid Interface Sci 2022; 626:803-814. [DOI: 10.1016/j.jcis.2022.06.174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/18/2022] [Revised: 06/14/2022] [Accepted: 06/29/2022] [Indexed: 10/31/2022]
|
21
|
Guo QL, Dai XL, Yin MY, Cheng HW, Qian HS, Wang H, Zhu DM, Wang XW. Nanosensitizers for sonodynamic therapy for glioblastoma multiforme: current progress and future perspectives. Mil Med Res 2022; 9:26. [PMID: 35676737 PMCID: PMC9178901 DOI: 10.1186/s40779-022-00386-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/23/2021] [Accepted: 05/22/2022] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor, and it is associated with poor prognosis. Its characteristics of being highly invasive and undergoing heterogeneous genetic mutation, as well as the presence of the blood-brain barrier (BBB), have reduced the efficacy of GBM treatment. The emergence of a novel therapeutic method, namely, sonodynamic therapy (SDT), provides a promising strategy for eradicating tumors via activated sonosensitizers coupled with low-intensity ultrasound. SDT can provide tumor killing effects for deep-seated tumors, such as brain tumors. However, conventional sonosensitizers cannot effectively reach the tumor region and kill additional tumor cells, especially brain tumor cells. Efforts should be made to develop a method to help therapeutic agents pass through the BBB and accumulate in brain tumors. With the development of novel multifunctional nanosensitizers and newly emerging combination strategies, the killing ability and selectivity of SDT have greatly improved and are accompanied with fewer side effects. In this review, we systematically summarize the findings of previous studies on SDT for GBM, with a focus on recent developments and promising directions for future research.
Collapse
Affiliation(s)
- Qing-Long Guo
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, 230032, China.,Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Xing-Liang Dai
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Meng-Yuan Yin
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, 230032, China.,Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Hong-Wei Cheng
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| | - Hai-Sheng Qian
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, 230032, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Dao-Ming Zhu
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, the First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Xian-Wen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
22
|
Wen M, Yu N, Wu S, Huang M, Qiu P, Ren Q, Zhu M, Chen Z. On-demand assembly of polymeric nanoparticles for longer-blood-circulation and disassembly in tumor for boosting sonodynamic therapy. Bioact Mater 2022; 18:242-253. [PMID: 35387175 PMCID: PMC8961299 DOI: 10.1016/j.bioactmat.2022.03.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/15/2021] [Revised: 02/18/2022] [Accepted: 03/06/2022] [Indexed: 12/14/2022] Open
Abstract
Sonodynamic therapy (SDT) is one of the promising strategies for tumor therapy, but its application is usually hindered by fast clearance in blood-circulation, abnormal tumor microenvironment, and inefficient generation of reactive oxygen species. To solve these problems, we proposed an on-demand assembly-disassembly strategy, where the assembly is favorable for longer-blood-circulation and then the disassembly in tumor is favorable for boosting SDT. Hematoporphyrin monomethyl ether (HMME) as the model of organic sonosensitizers were conjugated with hyaluronic acid (HA). Then HA-HMME was mixed with catalase (CAT) and assembled into polymeric nanoparticles (CAT@HA-HMME NPs) with size of ∼80 nm. CAT@HA-HMME NPs exhibit good biocompatibility and a longer blood half-time (t1/2 = 4.17 h) which is obviously longer than that (∼0.82 h) of HMME molecules. After HA receptor-mediated endocytosis of cancer cells, CAT@HA-HMME NPs can be cleaved by endogenous hyaluronidase, resulting in the on-demand disassembly in tumor to release HA-HMME molecules and CAT. The CAT catalyzes the endogenous H2O2 into O2 to relieve the hypoxic microenvironment, and the released HA-HMME exhibits a higher ROS generation ability, greatly boosting SDT for the inhibition of tumor growth. Therefore, the on-demand assembly-disassembly strategy may provide some insight in the design and development of nanoagents for tumor therapy. On-demand assembly from molecules to nanoparticles for longer-blood-circulation. On-demand disassembly in presence of hyaluronidase (in tumor) for boosting sonodynamic effects. Efficient damage on cancer cells in-vitro and Significant inhibition of the tumor growth due to the enhanced SDT.
Collapse
|
23
|
Applications of the ROS-Responsive Thioketal Linker for the Production of Smart Nanomedicines. Polymers (Basel) 2022; 14:polym14040687. [PMID: 35215600 PMCID: PMC8874672 DOI: 10.3390/polym14040687] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/20/2022] [Revised: 02/06/2022] [Accepted: 02/08/2022] [Indexed: 12/16/2022] Open
Abstract
Reactive oxygen species (ROS)-sensitive drug delivery systems (DDS) specifically responding to altered levels of ROS in the pathological microenvironment have emerged as an effective means to enhance the pharmaceutical efficacy of conventional nanomedicines, while simultaneously reducing side effects. In particular, the use of the biocompatible, biodegradable, and non-toxic ROS-responsive thioketal (TK) functional group in the design of smart DDS has grown exponentially in recent years. In the design of TK-based DDS, different technological uses of TK have been proposed to overcome the major limitations of conventional DDS counterparts including uncontrolled drug release and off-target effects. This review will focus on the different technological uses of TK-based biomaterials in smart nanomedicines by using it as a linker to connect a drug on the surface of nanoparticles, form prodrugs, as a core component of the DDS to directly control its structure, to control the opening of drug-releasing gates or to change the conformation of the nano-systems. A comprehensive view of the various uses of TK may allow researchers to exploit this reactive linker more consciously while designing nanomedicines to be more effective with improved disease-targeting ability, providing novel therapeutic opportunities in the treatment of many diseases.
Collapse
|
24
|
Deep and precise lighting-up/combat diseases through sonodynamic agents integrating molecular imaging and therapy modalities. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214333] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022]
|
25
|
Huang B, Wang L, Tang K, Chen S, Xu Y, Liao H, Niu C. IR780 Based Sonotherapeutic Nanoparticles to Combat Multidrug-Resistant Bacterial Infections. Front Chem 2022; 10:840598. [PMID: 35141201 PMCID: PMC8818736 DOI: 10.3389/fchem.2022.840598] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/21/2021] [Accepted: 01/04/2022] [Indexed: 11/23/2022] Open
Abstract
Multidrug-resistant (MDR) bacterial strains have emerged and weakened the therapeutic effects of antibacterial drugs. Sonodynamic therapy (SDT) takes advantage of noninvasiveness and deep tissue-penetrating features and has been rejuvenated to combat MDR bacteria and their biofilm-associated infections. To improve the efficacy of antibacterial SDT, we first developed IR780-based PLGA nanoparticles as sonosensitizers for high-frequency ultrasound (US)-monitored antibacterial SDT of MRSA myositis by therapeutic low-frequency US. In this study, the developed shell-core-structured IR780@PLGA nanoparticles were designed with a polymer shell PLGA with the sonosensitizer IR780 loaded on. High-frequency diagnostic US was introduced to monitor the sonotherapeutic progression of bacterial myositis by therapeutic low-frequency US. Importantly, the in vitro and in vivo results confirmed that IR780@PLGA nanoparticles combined with US irradiation possess high efficiency for antibacterial therapy. This approach provides a simple and efficient strategy to monitor and combat MDR bacterial infection.
Collapse
Affiliation(s)
- Biying Huang
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Long Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Kui Tang
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Sijie Chen
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yan Xu
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Haiqin Liao
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chengcheng Niu
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
26
|
Song S, Ma D, Xu L, Wang Q, Liu L, Tong X, Yan H. Low-intensity pulsed ultrasound-generated singlet oxygen induces telomere damage leading to glioma stem cell awakening from quiescence. iScience 2022; 25:103558. [PMID: 34988401 PMCID: PMC8693467 DOI: 10.1016/j.isci.2021.103558] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/18/2021] [Revised: 09/09/2021] [Accepted: 12/01/2021] [Indexed: 12/27/2022] Open
Abstract
Cancer stem cells, quiescent and drug resistant, have become a therapeutic target. Unlike high-intensity focused ultrasound directly killing tumor, low-intensity pulsed ultrasound (LIPUS), a new noninvasive physical device, promotes pluripotent stem cell differentiation and is primarily applied in tissue engineering but rarely in oncotherapy. We explored the effect and mechanism of LIPUS on glioma stem cell (GSC) expulsion from quiescence. Here, we observed that LIPUS led to attenuated expression of GSC biomarkers, promoted GSC escape from G0 quiescence, and significantly weakened the Wnt and Hh pathways. Of note, LIPUS transferred sonomechanical energy into cytochrome c and B5 proteins, which converted oxygen molecules into singlet oxygen, triggering telomere crisis. The in vivo and in vitro results confirmed that LIPUS enhanced the GSC sensitivity to temozolomide. These results demonstrated that LIPUS "waked up" GSCs to improve their sensitivity to chemotherapy, and importantly, we confirmed the direct targeted proteins of LIPUS in GSCs.
Collapse
Affiliation(s)
- Sirong Song
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300070, China
| | - Dongbin Ma
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300070, China
| | - Lixia Xu
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin 300350, P.R.China
| | - Qiong Wang
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin 300350, P.R.China
| | - Lanxiang Liu
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, No. 258 Wenhua Road, Qinhuangdao 066000, Hebei Province, P.R. China
| | - Xiaoguang Tong
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin 300350, P.R.China
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350, P.R. China
- Corresponding author
| | - Hua Yan
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin 300350, P.R.China
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300350, P.R. China
- Corresponding author
| |
Collapse
|
27
|
Zhao LY, Chao X, Yang BS, Wang GG, Zou JZ, Wu F. Phase-shift Perfluoropentane Nanoemulsions Enhance Pulsed High-intensity Focused Ultrasound Ablation in an Isolated Perfused Liver System and Their Potential Value for Cancer Therapy. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2022; 41:107-121. [PMID: 33724514 DOI: 10.1002/jum.15686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 08/12/2019] [Revised: 01/26/2021] [Accepted: 02/13/2021] [Indexed: 06/12/2023]
Abstract
PURPOSE To investigate whether phase-shift perfluoropetane (PFP) nanoemulsions can enhance pulsed high-intensity focused ultrasound (HIFU) ablation. METHODS PFP was encapsulated by poly(lactic-co-glycolic acid) (PLGA) to form a nanometer-sized droplet (PLGA-PFP), which was added to an isolated perfused liver system. Meanwhile, phosphate-buffered saline (PBS) was used as a control. The perfused liver was exposed to HIFU (150 W, t = 3/5/10 s) at various duty cycles (DCs). The ultrasound images, cavitation emissions, and temperature were recorded. Rabbits with subcutaneous VX2 tumors were exposed to HIFU (150 W) at various DCs with or without PLGA-PFP. After ablation, necrosis volume and energy efficiency factor were calculated. Pathologic characteristics were observed. RESULTS Compared to the PBS control, PLGA-PFP nanoemulsions markedly enhanced HIFU-induced necrosis volume in both perfused livers and subcutaneous VX2 tumor-bearing rabbits (P <.05). Inertial cavitation was much stronger in the pulsed-HIFU exposure at 10% than that in the continuous-wave HIFU exposure (P <.01). Peak temperature at 100% DC was significantly higher than that at 10% (P <.05). Compared to 100% DC HIFU exposure, the mean necrosis volume induced by 10 s exposure at 50% DC was significantly larger (P <.005) but lower at 10% DC in the isolated perfused livers (P <.05). In addition, the mean necrosis volume in subcutaneous VX2 tumor-bearing rabbits was significantly increased after HIFU exposure at 10% DC when compared to those at 100% DC (P <.05). Histopathologic analysis showed liquefaction necrosis in pulsed HIFU. CONCLUSION PLGA-PFP nanoemulsions can enhance HIFU ablation in the isolated perfused livers and promote tumor ablation in the subcutaneous xenograft rabbit model. Appropriate pulsed HIFU exposure may increase the necrosis volume and reduce total ultrasound energy required for HIFU ablation.
Collapse
Affiliation(s)
- Lu-Yan Zhao
- Transformational Medical Center, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
- The College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xu Chao
- The College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Bing-She Yang
- Transformational Medical Center, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Guo-Guan Wang
- The College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jian-Zhong Zou
- College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Feng Wu
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
28
|
Highlights in ultrasound-targeted microbubble destruction-mediated gene/drug delivery strategy for treatment of malignancies. Int J Pharm 2021; 613:121412. [PMID: 34942327 DOI: 10.1016/j.ijpharm.2021.121412] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/02/2021] [Revised: 12/06/2021] [Accepted: 12/17/2021] [Indexed: 01/05/2023]
Abstract
Ultrasound is one of the safest and most advanced medical imaging technologies that is widely used in clinical practice. Ultrasound microbubbles, traditionally used for contrast-enhanced imaging, are increasingly applied in Ultrasound-targeted Microbubble Destruction (UTMD) technology which enhances tissue and cell membrane permeability through cavitation and sonoporation, to result in a promising therapeutic gene/drug delivery strategy. Here, we review recent developments in the application of UTMD-mediated gene and drug delivery in the diagnosis and treatment of tumors, including the concept, mechanism of action, clinical application status, and advantages of UTMD. Furthermore, the future perspectives that should be paid more attention to in this field are prospected.
Collapse
|
29
|
Zhou J, Hou J, Liu S, Xu J, Luo Y, Zheng J, Li X, Wang Z, Ran H, Guo D. Theranostic Nanoplatform with Sequential SDT and ADV Effects in Response to Well-Programmed LIFU Irradiation for Cervical Cancer. Int J Nanomedicine 2021; 16:7995-8012. [PMID: 34916791 PMCID: PMC8669754 DOI: 10.2147/ijn.s339257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/14/2021] [Accepted: 11/15/2021] [Indexed: 11/23/2022] Open
Abstract
Background Some patients with cervical cancer have the need to preserve fertility; therefore, a minimally invasive treatment option that can effectively inactivate tumors in these patients is necessary. Methods In this paper, we designed and prepared nanoparticles (NPs) carrying IR780 and perfluorohexane (PFH) and characterized their properties. We focused on the promotion of programmed low-intensity focused ultrasound (LIFU) irradiation on the penetration and treatment of cervical cancer. First we used penetration-enhancing LIFU irradiation to promote the penetration of the NPs into 3D multicellular tumor spheroids (MCTSs) and tumors in tumor-bearing nude mice. Then we used re-therapeutic LIFU irradiation to achieve antitumor effects in vitro and in vivo. Photoacoustic (PA) and magnetic resonance (MR) imaging were used to monitor and evaluate the targeting and therapeutic effects of these NPs on tumor tissues. Results The NPs prepared in this paper exhibited high affinity for HeLa cells, and can selectively achieve mitochondrial localization in the cell due to IR780 assistance. The penetration-enhancing LIFU irradiation have the ability to promote the penetration of the NPs into cervical cancer models in vivo and in vitro. Under LIFU irradiation, the cytotoxic reactive oxygen species (ROS) produced by IR780 during the first half of the re-therapeutic LIFU irradiation and the physical acoustic droplet vaporization (ADV) effect after PFH phase transition during the second half of the re-therapeutic LIFU irradiation can achieve synergistic minimally invasive treatment of tumors, which can be visualized and evaluated by PA and MR imaging in vivo. Conclusion Well-programmed LIFU irradiation can promote NP penetration into deep tumor tissue and achieve antitumor effects simultaneously. Linking ROS + ADV effects can induce cell coagulation necrosis and lead to a comprehensive, long-term impact on tumor tissue, providing a conceptual theranostic nanoplatform for cervical cancer.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Jingxin Hou
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China.,Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Shuling Liu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Jie Xu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Ying Luo
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Jun Zheng
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Xin Li
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Dajing Guo
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| |
Collapse
|
30
|
Hu D, Pan M, Yang Y, Sun A, Chen Y, Yuan L, Huang K, Qu Y, He C, Wei Q, Qian Z. Trimodal Sono/Photoinduced Focal Therapy for Localized Prostate Cancer: Single‐Drug‐Based Nanosensitizer under Dual‐Activation. ADVANCED FUNCTIONAL MATERIALS 2021. [DOI: 10.1002/adfm.202104473] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 02/05/2023]
Affiliation(s)
- DanRong Hu
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine Key Laboratory of Rehabilitation Medicine in Sichuan Province State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center Chengdu Sichuan 610041 P. R. China
| | - Meng Pan
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine Key Laboratory of Rehabilitation Medicine in Sichuan Province State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center Chengdu Sichuan 610041 P. R. China
| | - Yun Yang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine Key Laboratory of Rehabilitation Medicine in Sichuan Province State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center Chengdu Sichuan 610041 P. R. China
| | - Ao Sun
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine Key Laboratory of Rehabilitation Medicine in Sichuan Province State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center Chengdu Sichuan 610041 P. R. China
| | - Yu Chen
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine Key Laboratory of Rehabilitation Medicine in Sichuan Province State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center Chengdu Sichuan 610041 P. R. China
| | - LiPing Yuan
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine Key Laboratory of Rehabilitation Medicine in Sichuan Province State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center Chengdu Sichuan 610041 P. R. China
| | - KangKang Huang
- Department of Orthopedics West China Hospital Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Ying Qu
- Department of Hematology and Research Laboratory of Hematology State Key Laboratory of Biotherapy West China Hospital Sichuan University Collaborative Innovation Center Chengdu Sichuan 610041 P. R. China
| | - ChengQi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine Key Laboratory of Rehabilitation Medicine in Sichuan Province State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center Chengdu Sichuan 610041 P. R. China
| | - Quan Wei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine Key Laboratory of Rehabilitation Medicine in Sichuan Province State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center Chengdu Sichuan 610041 P. R. China
| | - ZhiYong Qian
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine Key Laboratory of Rehabilitation Medicine in Sichuan Province State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center Chengdu Sichuan 610041 P. R. China
| |
Collapse
|
31
|
Meng Z, Zhang Y, Shen E, Li W, Wang Y, Sathiyamoorthy K, Gao W, C. Kolios M, Bai W, Hu B, Wang W, Zheng Y. Marriage of Virus-Mimic Surface Topology and Microbubble-Assisted Ultrasound for Enhanced Intratumor Accumulation and Improved Cancer Theranostics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2004670. [PMID: 34258156 PMCID: PMC8261514 DOI: 10.1002/advs.202004670] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 12/04/2020] [Revised: 03/11/2021] [Indexed: 05/13/2023]
Abstract
The low delivery efficiency of nanoparticles to solid tumors greatly reduces the therapeutic efficacy and safety which is closely related to low permeability and poor distribution at tumor sites. In this work, an "intrinsic plus extrinsic superiority" administration strategy is proposed to dramatically enhance the mean delivery efficiency of nanoparticles in prostate cancer to 6.84% of injected dose, compared to 1.42% as the maximum in prostate cancer in the previously reported study. Specifically, the intrinsic superiority refers to the virus-mimic surface topology of the nanoparticles for enhanced nano-bio interactions. Meanwhile, the extrinsic stimuli of microbubble-assisted low-frequency ultrasound is to enhance permeability of biological barriers and improve intratumor distribution. The enhanced intratumor enrichment can be verified by photoacoustic resonance imaging, fluorescence imaging, and magnetic resonance imaging in this multifunctional nanoplatform, which also facilitates excellent anticancer effect of photothermal treatment, photodynamic treatment, and sonodynamic treatment via combined laser and ultrasound irradiation. This study confirms the significant advance in nanoparticle accumulation in multiple tumor models, which provides an innovative delivery paradigm to improve intratumor accumulation of nanotherapeutics.
Collapse
Affiliation(s)
- Zheying Meng
- Department of Ultrasound in MedicineShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233P. R. China
- Shanghai Institute of Ultrasound in MedicineShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233P. R. China
| | - Yang Zhang
- Department of Ultrasound in MedicineShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233P. R. China
- Shanghai Institute of Ultrasound in MedicineShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233P. R. China
| | - E Shen
- Department of Ultrasound in MedicineShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233P. R. China
- Shanghai Institute of Ultrasound in MedicineShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233P. R. China
| | - Wei Li
- Department of ChemistryShanghai Key Laboratory of Molecular Catalysis and Innovative MaterialsFudan UniversityShanghai200433P. R. China
| | - Yanjie Wang
- Department of PhysicsRyerson UniversityTorontoOntarioM5B 2K3Canada
| | | | - Wei Gao
- Department of Ultrasound in MedicineShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233P. R. China
- Shanghai Institute of Ultrasound in MedicineShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233P. R. China
| | | | - Wenkun Bai
- Department of Ultrasound in MedicineShanghai Jiao Tong University Affiliated Sixth People's Hospital, Institute of Medical ImagingShanghai Jiao Tong UniversityShanghai200233P. R. China
| | - Bing Hu
- Department of Ultrasound in MedicineShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233P. R. China
- Shanghai Institute of Ultrasound in MedicineShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233P. R. China
| | - Wenxing Wang
- Department of ChemistryShanghai Key Laboratory of Molecular Catalysis and Innovative MaterialsFudan UniversityShanghai200433P. R. China
| | - Yuanyi Zheng
- Department of Ultrasound in MedicineShanghai Jiao Tong University Affiliated Sixth People's HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200032P. R. China
| |
Collapse
|
32
|
Wang S, Tian R, Zhang X, Cheng G, Yu P, Chang J, Chen X. Beyond Photo: Xdynamic Therapies in Fighting Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007488. [PMID: 33987898 DOI: 10.1002/adma.202007488] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 11/02/2020] [Revised: 12/02/2020] [Indexed: 05/14/2023]
Abstract
Reactive oxygen species (ROS)-related therapeutic approaches are developed as a promising modality for cancer treatment because the aberrant increase of intracellular ROS level can cause cell death due to nonspecific oxidation damage to key cellular biomolecules. However, the most widely considered strategy, photodynamic therapy (PDT), suffers from critical limitations such as limited tissue-penetration depth, high oxygen dependence, and phototoxicity. Non-photo-induced ROS generation strategies, which are defined as Xdynamic therapies (X = sono, radio, microwave, chemo, thermo, and electro), show good potential to overcome the drawbacks of PDT. Herein, recent advances in the development of Xdynamic therapies, including the design of systems, the working mechanisms, and examples of cancer therapy application, are introduced. Furthermore, the approaches to enhance treatment efficiency of Xdynamic therapy are highlighted. Finally, the perspectives and challenges of these strategies are also discussed.
Collapse
Affiliation(s)
- Sheng Wang
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Rui Tian
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Xu Zhang
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Guohui Cheng
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Peng Yu
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Jin Chang
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology and Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Departments of Chemical and Biomolecular Engineering, and, Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| |
Collapse
|
33
|
Abstract
IR780, a small molecule with a strong optical property and excellent photoconversion efficiency following near infrared (NIR) irradiation, has attracted increasing attention in the field of cancer treatment and imaging. This review is focused on different IR780-based nanoplatforms and the application of IR780-based nanomaterials for cancer bioimaging and therapy. Thus, this review summarizes the overall aspects of IR780-based nanomaterials that positively impact cancer biomedical applications.
Collapse
Affiliation(s)
- Long Wang
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China. and Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Chengcheng Niu
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China. and Department of Ultrasound Diagnosis and Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
34
|
Bai L, Wang M, Zhang L, Zhao M, Ren M, Zheng L, Lei M, Shen H. Poly(Amino Acid) Coordination Nanoparticle as a Potent Sonosensitizer for Cancer Therapy. ACS APPLIED BIO MATERIALS 2021. [DOI: 10.1021/acsabm.0c01383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/28/2022]
Affiliation(s)
- Lintao Bai
- State Key Laboratory of Organic−Inorganic Composites, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Mingkun Wang
- State Key Laboratory of Organic−Inorganic Composites, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Lin Zhang
- State Key Laboratory of Chemical Resource Engineering, Institute of Computational Chemistry, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Meijun Zhao
- State Key Laboratory of Organic−Inorganic Composites, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Mei Ren
- State Key Laboratory of Organic−Inorganic Composites, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Lirong Zheng
- Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Ming Lei
- State Key Laboratory of Chemical Resource Engineering, Institute of Computational Chemistry, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Heyun Shen
- State Key Laboratory of Organic−Inorganic Composites, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| |
Collapse
|
35
|
Um W, E. K. PK, Lee J, Kim CH, You DG, Park JH. Recent advances in nanomaterial-based augmented sonodynamic therapy of cancer. Chem Commun (Camb) 2021; 57:2854-2866. [DOI: 10.1039/d0cc07750j] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/15/2022]
Abstract
This review focuses on recent advances in augmented sonodynamic therapy (SDT) using engineered nanomaterials, and the mechanism of SDT for discussing future perspectives.
Collapse
Affiliation(s)
- Wooram Um
- School of Chemical Engineering
- College of Engineering
- Sungkyunkwan University
- Suwon
- Republic of Korea
| | - Pramod Kumar E. K.
- School of Chemical Engineering
- College of Engineering
- Sungkyunkwan University
- Suwon
- Republic of Korea
| | - Jeongjin Lee
- Department of Health Sciences and Technology
- SAIHST
- Sungkyunkwan University
- Seoul
- Republic of Korea
| | - Chan Ho Kim
- School of Chemical Engineering
- College of Engineering
- Sungkyunkwan University
- Suwon
- Republic of Korea
| | - Dong Gil You
- School of Chemical Engineering
- College of Engineering
- Sungkyunkwan University
- Suwon
- Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering
- College of Engineering
- Sungkyunkwan University
- Suwon
- Republic of Korea
| |
Collapse
|
36
|
Abstract
Various therapeutic techniques have been studied for treating cancer precisely and effectively, such as targeted drug delivery, phototherapy, tumor-specific catalytic therapy, and synergistic therapy, which, however, evoke numerous challenges due to the inherent limitations of these therapeutic modalities and intricate biological circumstances as well. With the remarkable advances of nanotechnology, nanoplatform-based cascade engineering, as an efficient and booming strategy, has been tactfully introduced to optimize these cancer therapies. Based on the designed nanoplatforms, pre-supposed cascade processes could be triggered under specific conditions to generate/deliver more therapeutic species or produce stronger tumoricidal effects inside tumors, aiming to achieve cancer therapy with increased anti-tumor efficacy and diminished side effects. In this review, the recent advances in nanoplatform-based cascade engineering for cancer therapy are summarized and discussed, with an emphasis on the design of smart nanoplatforms with unique structures, compositions and properties, and the implementation of specific cascade processes by means of endogenous tumor microenvironment (TME) resources and/or exogenous energy inputs. This fascinating strategy presents unprecedented potential in the enhancement of cancer therapies, and offers better controllability, specificity and effectiveness of therapeutic functions compared to the corresponding single components/functions. In the end, challenges and prospects of such a burgeoning strategy in the field of cancer therapy will be discussed, hopefully to facilitate its further development to meet the personalized treatment demands.
Collapse
Affiliation(s)
- Jiajie Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China.
| | | | | | | |
Collapse
|
37
|
Zhang R, Zhang L, Ran H, Li P, Huang J, Tan M, Yang Y, Wang Z. A mitochondria-targeted anticancer nanoplatform with deep penetration for enhanced synergistic sonodynamic and starvation therapy. Biomater Sci 2020; 8:4581-4594. [PMID: 32691765 DOI: 10.1039/d0bm00408a] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/16/2022]
Abstract
Sonodynamic therapy (SDT), as an emerging technique, gives rise to reactive oxygen species (ROS)-induced apoptosis of tumor cells. However, nonselective enrichment and unsatisfactory penetration depth of sonosensitizers in tumor tissues limit its application. In this study, we synthesized core/shell (glucose oxidase (GOx) in the core/hematoporphyrin monomethyl ether (HMME) and IR780 in the shell) structured polylactic-co-glycolic acid (PLGA) nanoparticles (NPs) with deep tumor penetration and mitochondrial targeting capability for synergistic sonodynamic and starvation therapy. After passing through the endothelial space of tumor vasculatures, by virtue of IR780, these NPs can selectively accumulate towards cancer cells/sites, especially in mitochondria and diffuse into deep tumour centres. Upon ultrasound (US) exposure, the overproduced ROS cause tumor cell apoptosis. Sonodynamic effects can be amplified by mitochondrial targeting because mitochondria are susceptible to ROS. GOx blocks glucose (energy) supply, further suppressing the growth of malignant tumors. This synergistic therapy exhibited a superb response to treatment (4.7-fold lower tumor growth in volume than the control group). In addition, these NPs also serve as excellent photoacoustic (PA)/fluorescent (FL) imaging contrast agents to simultaneously monitor and guide cancer therapy. This study paves a promising way to achieve an ideal strategy for cancer therapy.
Collapse
Affiliation(s)
- Ruo Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Ji P, Wang L, Wang S, Zhang Y, Qi X, Tao J, Wu Z. Hyaluronic acid-coated metal-organic frameworks benefit the ROS-mediated apoptosis and amplified anticancer activity of artesunate. J Drug Target 2020; 28:1096-1109. [PMID: 32552125 DOI: 10.1080/1061186x.2020.1781136] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/24/2022]
Abstract
Artesunate (AS), as an effective new tumour treatment drug, induces cancer cell death based on high intracellular reactive oxygen species (ROS) produced by interacting with ferrous ions. However, the relatively low intracellular ferrous iron ion concentrations and the low efficiency of ROS generation limit its clinical application. Herein, we developed a metal-organic framework-Fe2+ (MOF), and AS was loaded in the MOF and then coated with hyaluronic acid (HA) on the surface of the MOF (HA@MOF-AS) for targeted and enhanced cancer treatment. HA@MOF-AS has high loading efficiency, good monodispersity, biocompatibility, strong cell uptake capacity and high intracellular ROS production, and it can target tumour tissues. In addition, in vivo anticancer studies have shown that HA@MOF-AS not only has high accumulation in tumours but also significantly inhibits tumour growth without significant damage to major organs. Therefore, HA@MOF-AS has excellent potential and may open a new approach for targeted cancer treatment.
Collapse
Affiliation(s)
- Peng Ji
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, PR China
| | - Le Wang
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, PR China
| | - Siqi Wang
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, PR China
| | - Yongxin Zhang
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, PR China
| | - Xiaole Qi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, PR China
| | - Juan Tao
- Department of Pharmacy and Traditional Chinese Pharmacy, Jiangsu College of Nursing, Huaian, PR China
| | - Zhenghong Wu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing, PR China
| |
Collapse
|
39
|
Huang B, Chen S, Pei W, Xu Y, Jiang Z, Niu C, Wang L. Oxygen-Sufficient Nanoplatform for Chemo-Sonodynamic Therapy of Hypoxic Tumors. Front Chem 2020; 8:358. [PMID: 32411675 PMCID: PMC7199163 DOI: 10.3389/fchem.2020.00358] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/08/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022] Open
Abstract
Modulation of hypoxia is an essential factor for enhancing the effects of antitumor therapies, especially sonodynamic therapy and chemotherapy. To improve the efficacy of combination therapy by reversing the hypoxic tumor microenvironment, we developed shell-core structured PPID-NPs, which were designed with a polymer shell onto the sonosensitizer and a chemotherapeutic drug were loaded and a perfluorocarbon core loaded with oxygen. The perfluorocarbon core provides sufficient oxygen not only for causing the sonosensitizer to produce more singlet oxygen to induce cell apoptosis but also for reducing drug resistance to enhance therapeutic efficacy. Furthermore, the release of chemotherapeutic drugs at the tumor site can be controlled. Thus, PPID-NPs can efficiently inhibit the growth of breast cancer by synergistic therapy under ultrasound exposure. We believe that our oxygen-sufficient nanoplatform could be an ideal therapeutic system for hypoxic tumors.
Collapse
Affiliation(s)
- Biying Huang
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Sijie Chen
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wenjing Pei
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yan Xu
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zichao Jiang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Chengcheng Niu
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Long Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
40
|
Choi V, Rajora MA, Zheng G. Activating Drugs with Sound: Mechanisms Behind Sonodynamic Therapy and the Role of Nanomedicine. Bioconjug Chem 2020; 31:967-989. [DOI: 10.1021/acs.bioconjchem.0c00029] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/12/2022]
Affiliation(s)
- Victor Choi
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, Ontario, Canada M5G 1L7
- School of Pharmacy, University College London, 29-39 Brunswick Square, London, United Kingdom WC1N 1AX
| | - Maneesha A. Rajora
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, Ontario, Canada M5G 1L7
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario, Canada M5S 3G9
| | - Gang Zheng
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, Ontario, Canada M5G 1L7
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario, Canada M5S 3G9
- Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, Ontario, Canada M5G 1L7
| |
Collapse
|