1
|
Zhang H, Wei Z, Wang Y, Bi Z, Han W, Shi M, Chen T, Sun Y, Wang L, Zhang S. Au 3+-Functionalized Metal-Organic Framework Coordinated Nanotherapeutics for Substrate Self-Supplied Parallel Catalytic and Calcium-Overload-Mediated Therapy of Cancer. ACS APPLIED BIO MATERIALS 2025; 8:446-456. [PMID: 39829267 DOI: 10.1021/acsabm.4c01423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The multiple enzymatic properties of the Au3+-modified metal-organic framework (Au3+-MOFs) have made it a functional catalytic system for antitumor treatment. However, in the face of insufficient catalytic substrates in tumor tissue, it is still impossible to achieve efficient treatment of tumors. Herein, Au3+-MOFs loaded with hyaluronic acid (HA)-modified calcium peroxide nanoparticles (CaO2 NPs) were used to construct a nanozyme (Au3+-MOF/CaO2/HA) for substrate self-supplied and parallel catalytic/calcium-overload-mediated therapy of cancer. Due to the specific targeted ability and retention (EPR) effect of the HA, the built nanozyme can effectively accumulate at the tumor site. Due to the oxidase-like (OXD) activity and peroxidase-like (POD) activity of Au3+-MOFs, superoxide radical anion (O2•-) and hydroxyl radicals (·OH) were cooperatively formed for parallel catalytic therapy (PCT) of cancer. Subsequently, CaO2 NPs were decomposed to Ca2+, H2O2, and O2 in the weak acidic environment of the tumor microenvironment (TME). Thus, self-supplementation of O2 as well as H2O2 was achieved, alleviating the deficiency of Au3+-MOF nanozyme catalytic substrate. In addition, Ca2+ can lead to oxidative stress for tumor calcification and calcium-overload-mediated therapy (COMT) to promote tumor necrosis in vivo. An effective paradigm of tumor PCT/COMT therapy with a self-supplying substrate has been successfully established for considerably enhanced therapeutic efficacy.
Collapse
Affiliation(s)
- Huairong Zhang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, College of Medicine, Linyi University, Linyi 276005, China
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Zizhen Wei
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Yuqi Wang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Zhiru Bi
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Wenxiu Han
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Minghui Shi
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, College of Medicine, Linyi University, Linyi 276005, China
| | - Tingting Chen
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Yongbiao Sun
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, College of Medicine, Linyi University, Linyi 276005, China
| | - Linjing Wang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, College of Medicine, Linyi University, Linyi 276005, China
| | - Shusheng Zhang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, College of Medicine, Linyi University, Linyi 276005, China
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| |
Collapse
|
2
|
Li X, Zhang X, Song L, Li Y, Liu A, Li L, Nešić MD, Li D, Peng L, Wang C, Lin Q. Nanozyme as Tumor Energy Homeostasis Disruptor to Augment Cascade Catalytic Therapy. ACS NANO 2024; 18:34656-34670. [PMID: 39661982 DOI: 10.1021/acsnano.4c09982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Breaking the balance of the tumor microenvironment and reshaping it sustainably remain major challenges in lung cancer treatment. Here, a "tumor energy homeostasis disruptor", the Cu2O@Au nanozyme was developed, which exhibits excellent glucose oxidase-like activity, enabling it to be used for starvation therapy and as a mimic peroxidase for chemodynamic therapy (CDT), producing •OH. Cu2O@Au nanozymes consume glucose at the tumor site to block the tumor's energy supply, produce H2O2 continuously, and lower the pH to enhance the efficiency of CDT, initiating a cascade reaction that leads to a storm of reactive oxygen species (ROS). Furthermore, Cu2O@Au nanozyme consumes glutathione and reduces the expression of the SLC7A11 (xCT) protein to decrease cancer cell uptake of cysteine, further enhancing the burst of ROS, resulting in lipid peroxidation in tumor cells and ultimately leading to ferroptosis. The excellent photothermal performance of Cu2O@Au can further enhance CDT. Additionally, Cu2O@Au nanozyme also has computed tomography (CT) and photothermal imaging capabilities. In conclusion, Cu2O@Au nanozymes, acting as tumor energy homeostasis disruptor, can effectively inhibit tumor growth and successfully achieve the synergistic effects of starvation therapy/CDT/photothermal therapy (PTT). This multifunctional nanozyme holds promise for providing valuable insights and therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Xingchen Li
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun 130012, China
| | - Xia Zhang
- Department of General Practice, The First Hospital of Jilin University, Changchun 130021, China
| | - Lei Song
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, China
| | - Yuan Li
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun 130012, China
| | - Annan Liu
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun 130012, China
| | - Lei Li
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun 130012, China
| | - Maja D Nešić
- Center for Light-Based Research and Technologies COHERENCE, Department of Atomic Physics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
| | - Dan Li
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, China
| | - Liping Peng
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, China
| | - Chunyan Wang
- Department of General Practice, The First Hospital of Jilin University, Changchun 130021, China
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
3
|
Sun Y, Chen Y, Wu B, Li H, Wang Y, Wang X, Deng L, Yang K, Wang X, Cheng W. Synergistic SDT/cuproptosis therapy for liver hepatocellular carcinoma: enhanced antitumor efficacy and specific mechanisms. J Nanobiotechnology 2024; 22:762. [PMID: 39696275 DOI: 10.1186/s12951-024-02995-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/06/2024] [Indexed: 12/20/2024] Open
Abstract
The efficacy of sonodynamic therapy (SDT), an emerging approach for tumor treatment, is hindered by the high levels of the antioxidant glutathione (GSH) in the tumor microenvironment (TME). In this study, we constructed nanobubbles loaded with the sonosensitizer HMME and the tumor-targeting peptide RGD (HMME-RGD@C3F8 NBs) for synergistic SDT/cuproptosis therapy of liver hepatocellular carcinoma (LIHC) in combination with Elesclomol-Cu as cuproptosis inducers. Endogenous GSH is consumed by Cu2+ to modulate the complex TME, thereby amplifying oxidative stress and further improving SDT performance. Additionally, intracellular Cu2+ overload can induce cuproptosis, which is further amplified by SDT, to initiate irreversible protein toxicity. The specific mechanism of synergistic SDT/cuproptosis therapy in LIHC was investigated by RNA sequencing analysis. The synergistic SDT/cuproptosis therapy reprogrammed the TME to improve the efficacy of immune checkpoint inhibitor-based immunotherapy. Furthermore, a risk-scoring model was created and displayed significant promise in the prognosis of LIHC.
Collapse
Affiliation(s)
- Yucao Sun
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Yichi Chen
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Bolin Wu
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Helin Li
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Yijun Wang
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Xiaodong Wang
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Liwen Deng
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Kuikun Yang
- School of Life Science and Technology, Harbin Institute of Technology, No. 92, Xidazhi Street, Nangang District, Harbin, 150081, Heilongjiang, China.
| | - Xiuhong Wang
- Department of Biochemistry and Molecular Biology, Heilongjiang Provincial Science and Technology Innovation Team in Higher Education Institutes for Infection and Immunity, Harbin Medical University, No. 157, Baojian Road, Nangang District, Harbin, 150081, Heilongjiang, China.
| | - Wen Cheng
- Department of Ultrasound, Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, No. 150, Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
4
|
Liu L, Zhang Y, Ju T, Chen X, Li X, Wu LA. Nanozymes: a promising solution for dental antibacterial applications. RSC Adv 2024; 14:36945-36959. [PMID: 39569116 PMCID: PMC11577344 DOI: 10.1039/d4ra07303g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/10/2024] [Indexed: 11/22/2024] Open
Abstract
Dental diseases pose significant public health challenges globally, affecting millions with conditions exacerbated by microbial-induced inflammation. Traditional natural enzymes, despite their antibacterial and anti-inflammatory capabilities, are limited by operational stability and environmental sensitivity. This review explores the revolutionary realm of nanozyme-artificial enzymes made from nanomaterials-which offer enhanced stability, cost-effectiveness, and ease of modification. We discuss the advent of nanozymes since their first recognition in 2007, emphasizing their enzyme-mimicking capabilities and applications in dental medicine, particularly for dental caries, pulpitis, periodontitis and peri-implantitis. This paper presents a comprehensive analysis of nanozymes' classification, mechanisms, and emerging applications, shedding light on their potential to revolutionize dental antibacterial treatments and addressing current challenges and future perspectives in their development.
Collapse
Affiliation(s)
- Lipeng Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University China
- Department of Immunology, School of Basic Medicine, The Fourth Military Medical University China
| | - Yaoyuan Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University China
| | - Tianjuan Ju
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University China
| | - Xutao Chen
- Department of Immunology, School of Basic Medicine, The Fourth Military Medical University China
| | - Xinwei Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University China
| | - Li-An Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University China
| |
Collapse
|
5
|
Zhang Q, Shi X, Gao T, Xing Y, Jin H, Hao J, Liu X, Liu X, Liu P. Precision management of Fusarium fujikuroi in rice through seed coating with an enhanced nanopesticide using a tannic acid-Zn II formulation. J Nanobiotechnology 2024; 22:717. [PMID: 39550576 PMCID: PMC11568670 DOI: 10.1186/s12951-024-02938-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/19/2024] [Indexed: 11/18/2024] Open
Abstract
Seed coating with fungicides is a common practice in controlling seed-borne diseases, but conventional methods often result in high toxicity to plants and soil. In this study, a nanoparticle formulation was successfully developed using the metal-organic framework UiO-66 as a carrier of the fungicide ipconazole (IPC), with a tannic acid (TA)-ZnII coating serving as a protective layer. The IPC@UiO-66-TA-ZnII nanoparticles provided a controlled release, triggered and regulated by environmental factors such as pH and temperature. This formulation efficiently controlled the proliferation of Fusarium fujikuroi spores, with high penetration into both rice roots and fungal mycelia. The product exhibited high antifungal activity, achieving control efficacy rates of 84.09% to 93.10%, low biotoxicity, and promoted rice growth. Compared to the IPC flowable suspension formula, IPC@UiO-66-TA-ZnII improved the physicochemical properties and enzymatic activities in soil. Importantly, it showed potential for mitigating damage to beneficial soil bacteria. This study provides a promising approach for managing plant diseases using nanoscale fungicides in seed treatment.
Collapse
Affiliation(s)
- Qizhen Zhang
- College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Xin Shi
- College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Tuqiang Gao
- College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Yaochun Xing
- College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Haisheng Jin
- College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Jianjun Hao
- School of Food and Agriculture, University of Maine, Orono, ME, 04469, USA
| | - Xiaofang Liu
- College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Xili Liu
- College of Plant Protection, China Agricultural University, Beijing, 100193, China.
| | - Pengfei Liu
- College of Plant Protection, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
6
|
Nejabat M, Samie A, Khojastehnezhad A, Hadizadeh F, Ramezani M, Alibolandi M, Abnous K, Taghdisi SM, Siaj M. Stimuli-Responsive Covalent Organic Frameworks for Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:51837-51859. [PMID: 39163539 DOI: 10.1021/acsami.4c07040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Chemotherapy as a common anticancer therapeutic modality is often challenged by various obstacles such as poor stability, low solubility, and severe side effects of chemotherapeutic agents as well as multidrug resistance of cancerous cells. Nanoparticles in the role of carriers for chemotherapeutic drugs and platforms for combining different therapeutic approaches have effectively participated in overcoming such drawbacks. In particular, nanoparticles able to induce their therapeutic effect in response to specific stimuli like tumor microenvironment characteristics (e.g., hypoxia, acidic pH, high levels of glutathione, and overexpressed hydrogen peroxide) or extrinsic stimulus of laser light bring about more precise and selective treatments. Among them, nanostructures of covalent organic frameworks (COFs) have drawn great interest in biomedical fields during recent years. Possessing large surface area, high porosity, structural stability, and customizable architecture, these biocompatible porous crystalline polymers properly translate to promising platforms for drug delivery and induction of combination therapies. With the focus on stimuli-responsive characteristics of nanoscale COFs, this study aims to propose an overview of their potentiality in cancer treatment on the basis of chemotherapy alone or in combination with sonodynamic, chemodynamic, photodynamic, and photothermal therapies.
Collapse
Affiliation(s)
- Masoud Nejabat
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
| | - Ali Samie
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
| | - Amir Khojastehnezhad
- Department of Chemistry, University of Quebec at Montreal, Montreal, Quebec H3C 3P8, Canada
| | - Farzin Hadizadeh
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
| | - Mohamed Siaj
- Department of Chemistry, University of Quebec at Montreal, Montreal, Quebec H3C 3P8, Canada
| |
Collapse
|
7
|
Chen R, Li S, Ren S, Han D, Qin K, Jia X, Zhou H, Gao Z. Micro-/nanostructures for surface-enhanced Raman spectroscopy: Recent advances and perspectives. Adv Colloid Interface Sci 2024; 331:103235. [PMID: 38908042 DOI: 10.1016/j.cis.2024.103235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/04/2024] [Accepted: 06/11/2024] [Indexed: 06/24/2024]
Abstract
Surface-enhanced Raman spectroscopy (SERS) has great potential for the analysis of molecules adsorbed on metals with rough surfaces or substrates with micro-/nanostructures. Plasmonic coupling between metal nanoparticles and the morphology of the rough metal surface can produce "hot spots" that enhance Raman scattering by adsorbed molecules, typically at micro- to nanomolar concentrations, although high enhancement factors can also facilitate single-molecule detection. This phenomenon is widely applicable for chemical analysis and sensing in various fields. In this review, the latest research progress on SERS micro-/nanosensors is evaluated, and the sensors are classified according to their individual functions. Furthermore, the design principles and working mechanisms of reported SERS-active micro-/nanostructured substrates are analyzed, and the design features adopted to overcome the difficulties associated with precision detection are explored. Finally, challenges and directions for future development in this field are discussed. This review serves as a design guide for novel SERS-active substrates.
Collapse
Affiliation(s)
- Ruipeng Chen
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Military Medical Sciences Academy, Tianjin 300050, China
| | - Shuang Li
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Military Medical Sciences Academy, Tianjin 300050, China
| | - Shuyue Ren
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Military Medical Sciences Academy, Tianjin 300050, China
| | - Dianpeng Han
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Military Medical Sciences Academy, Tianjin 300050, China
| | - Kang Qin
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Military Medical Sciences Academy, Tianjin 300050, China
| | - Xuexia Jia
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Military Medical Sciences Academy, Tianjin 300050, China
| | - Huanying Zhou
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Military Medical Sciences Academy, Tianjin 300050, China.
| | - Zhixian Gao
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Military Medical Sciences Academy, Tianjin 300050, China.
| |
Collapse
|
8
|
Lu X, Zheng Y, Liu Y, Li D, Lin J, Wei L, Gao S, Liu J, Zhang W, Chen Y. Orchestrating apoptosis and ferroptosis through enhanced sonodynamic therapy using amorphous UIO-66-CoO x. J Colloid Interface Sci 2024; 667:91-100. [PMID: 38621335 DOI: 10.1016/j.jcis.2024.04.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/17/2024]
Abstract
The development of efficient and multifunctional sonosensitizers is crucial for enhancing the efficacy of sonodynamic therapy (SDT). Herein, we have successfully constructed a CoOx-loaded amorphous metal-organic framework (MOF) UIO-66 (A-UIO-66-CoOx) sonosensitizer with excellent catalase (CAT)- and glutathione-oxidase (GSH-OXD)-like activities. The A-UIO-66-CoOx exhibits a 2.6-fold increase in singlet oxygen (1O2) generation under ultrasound (US) exposure compared to crystalline UIO-66 sonosensitizer, which is attributed to its superior charge transfer efficiency and consistent oxygen (O2) supply. Additionally, the A-UIO-66-CoOx composite reduces the expression of glutathione peroxidase (GPX4) by depleting glutathione (GSH) through Co3+ and Co2+ valence changes. The high levels of highly cytotoxic 1O2 and deactivation of GPX4 can lead to lethal lipid peroxidation, resulting in concurrent apoptosis and ferroptosis. Both in vitro and vivo tumor models comprehensively confirmed the enhanced SDT antitumor effect using A-UIO-66-CoOx sonosensitizer. Overall, this study emphasizes the possibility of utilizing amorphization engineering to improve the effectiveness of MOFs-based sonosensitizers for combined cancer therapies.
Collapse
Affiliation(s)
- Xiuxin Lu
- Department of Research, Department of Ultrasonography, Guangxi Medical University Cancer Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yang Zheng
- Department of Plastic Surgery, The Second Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yan Liu
- Department of Breast, Bone and Soft Tissue Oncology, Guangxi Medical University Cancer Hospital, Nanning, Nanning 530021, China; Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi Department of Education, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Dan Li
- Department of Research, Department of Ultrasonography, Guangxi Medical University Cancer Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jiaxin Lin
- Department of Research, Department of Ultrasonography, Guangxi Medical University Cancer Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Lineng Wei
- Department of Research, Department of Ultrasonography, Guangxi Medical University Cancer Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Song Gao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Junjie Liu
- Department of Research, Department of Ultrasonography, Guangxi Medical University Cancer Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China.
| | - Weiqing Zhang
- Department of Research, Department of Ultrasonography, Guangxi Medical University Cancer Hospital, Guangxi Medical University, Nanning, Guangxi 530021, China.
| | - Yanbo Chen
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China.
| |
Collapse
|
9
|
容 逍, 李 哲, 左 妍, 邱 逦. [Salphen-Based Fe-N 2O 2@C Nanomaterial Applied in Synergistic Sonodynamic and Chemodynamic Therapy of Tumors]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:813-825. [PMID: 39170026 PMCID: PMC11334295 DOI: 10.12182/20240760509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Indexed: 08/23/2024]
Abstract
Objective To synthesize a Salphen-based Fe-N2O2@C material with high peroxidase (POD)-mimicking activity and sonosensitivity for the synergistic sonodynamic (SDT) and chemodynamic (CDT) therapy of tumors. Methods Fe-N2O2 was synthesized via the hydrothermal method, and Fe-N2O2@C was prepared by incorporating a ketjen black substrate. The morphology, structure, composition, enzyme mimic activity for reactive oxygen species (ROS) production, and sonosensitivity of the material were characterized. The ability and mechanism of Fe-N2O2@C to perform synergistic SDT and CDT killing of 4T1 mouse breast cancer cells were explored through in vitro experiments. The in vivo tumor-killing ability of Fe-N2O2@C combined with ultrasound irradiation was investigated using a subcutaneous 4T1 tumor-bearing mouse model. Results FFe-N2O2 and Fe-N2O2@C were both irregularly shaped nanospheres with average particle sizes of 25.9 nm and 36.2 nm, respectively. XRD, FTIR, and XPS analyses confirmed that both Fe-N2O2 and Fe-N2O2@C possessed a Salphen covalent organic framework structure with M-N2O2 coordination, and the ketjen black loading had no significant impact on this structure. Compared to Fe-N2O2, Fe-N2O2@C exhibited high POD-mimicking activity (with K m reduced from 19.32 to 5.82 mmol/L and v max increased from 2.51×10-8 to 8.92×10-8 mol/[L·s]) and sonosensitivity. Fe-N2O2@C in combination with ultrasound irradiation could produce a large amount of ROS within cells and a subsequent significant decrease in mitochondrial membrane potential, thereby inducing TEM-observable mitochondrial damage and causing cell apoptosis and death. In addition, in vivo experiments showed that Fe-N2O2@C in combination with ultrasound irradiation could effectively inhibit tumor growth in a 4T1 subcutaneous tumor-bearing mouse model without significant in vivo toxicity. Conclusion In this study, we prepared a Salphen-based Fe-N2O2@C material with good biocompatibility, which can be used in combination with ultrasound irradiation to achieve SDT and CDT synergistic killing of tumor cells and inhibit tumor growth. This Salphen-based Fe-N2O2@C nanomaterial shows promising potential for multimodal tumor therapy.
Collapse
Affiliation(s)
- 逍 容
- 四川大学华西医院 超声医学科 (成都 610041)Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 哲轩 李
- 四川大学华西医院 超声医学科 (成都 610041)Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 妍 左
- 四川大学华西医院 超声医学科 (成都 610041)Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 逦 邱
- 四川大学华西医院 超声医学科 (成都 610041)Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
10
|
Xuan J, Wang Z, Huang Y, Liu Y, Han Y, Li M, Xiao M. DNA response element-based smart drug delivery systems for precise drug release. Biomater Sci 2024; 12:3550-3564. [PMID: 38832670 DOI: 10.1039/d4bm00138a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Smart drug delivery systems (DDSs) that respond to, interact with, or are actuated by biological signals or pathological abnormalities (e.g., the tumor microenvironment) for controllable drug release are appealing therapeutic platforms for cancer treatment. Owing to their inherent self-assembled nature, nucleic acids have emerged as programmable materials for the development of multifunctional structures. In response to external environmental stimuli, DNA response elements can serve as switches to trigger conformational changes in DNA structures. Their stimulus-responsive properties make them promising candidates for constructing smart DDSs, and advancements in DNA response element-based DDSs in the field of biomedicine have been made. This review summarizes different types of DNA response elements, including DNA aptamers, DNAzymes, disulfide bond-modified DNA, pH-responsive DNA motifs, and photocleavable DNA building blocks, and highlights the advancements in DNA response element-based smart DDSs for precise drug release. Finally, future challenges and perspectives in this field are discussed.
Collapse
Affiliation(s)
- Jinnan Xuan
- Hubei Key Laboratory of Photoelectric Materials and Devices, School of Materials Science and Engineering, Hubei Normal University, 11 Cihu Road, Huangshi 435002, P. R. China
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, P. R. China.
| | - Zhen Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Shaanxi Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P. R. China
| | - Yuting Huang
- Department of Radiotherapy, Chaohu Hospital of Anhui Medical University, 64 Chaohu North Road, Chaohu 238000, P. R. China
| | - Yisi Liu
- Hubei Key Laboratory of Photoelectric Materials and Devices, School of Materials Science and Engineering, Hubei Normal University, 11 Cihu Road, Huangshi 435002, P. R. China
| | - Yuqiang Han
- Hubei Key Laboratory of Photoelectric Materials and Devices, School of Materials Science and Engineering, Hubei Normal University, 11 Cihu Road, Huangshi 435002, P. R. China
| | - Man Li
- Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Shaanxi Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P. R. China
| | - Mingshu Xiao
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, P. R. China.
| |
Collapse
|
11
|
Chen Y, Li X, Luo K, Wang T, Liu T, Lu E, Wang R, Luo Y, Sha X. Hyperthermia/glutathione-triggered ferritin nanoparticles amplify the ferroptosis for synergistic tumor therapy. Mater Today Bio 2024; 26:101085. [PMID: 38765248 PMCID: PMC11098959 DOI: 10.1016/j.mtbio.2024.101085] [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] [Scholar Register] [Received: 03/06/2024] [Revised: 04/22/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024] Open
Abstract
Breast cancer is the most diagnosed malignancy in women globally, and drug resistance is among the major obstacles to effective breast cancer treatment. Emerging evidence indicates that photothermal therapy and ferroptosis are both promising therapeutic techniques for the treatment of drug-resistant breast tumors. In this study, we proposed a thermal/ferroptosis/magnetic resonance imaging (MRI) triple functional nanoparticle (I@P-ss-FRT) in which ferritin, an iron storage material with excellent cellular uptake capacity, was attached via disulfide bonds onto polydopamine coated iron oxide nanoparticle (I@P) as photothermal transduction agent and MRI probe. I@P-ss-FRT converted the near-infrared light (NIR) into localized heat which accelerated the release of ferrous ions from ferritin accomplished by glutathione reduction and subsequently induced ferroptosis. The drug-resistant cancer cell lines exhibited a more significant uptake of I@P-ss-FRT and sensitivity to PTT/ferroptosis compared with normal cancer cell lines. In vivo, I@P-ss-FRT plus NIR displayed the best tumor-killing potential with inhibitory rate of 83.46 %, along with a decline in GSH/GPX-4 content and an increase in lipid peroxides generation at tumor sites. Therefore, I@P-ss-FRT can be applied to combat drug-resistant breast cancer.
Collapse
Affiliation(s)
- Yiting Chen
- Key Laboratory of Smart Drug Delivery (Ministry of Education), School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Xinhong Li
- Key Laboratory of Smart Drug Delivery (Ministry of Education), School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Kuankuan Luo
- Key Laboratory of Smart Drug Delivery (Ministry of Education), School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Tao Wang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Tongyao Liu
- Key Laboratory of Smart Drug Delivery (Ministry of Education), School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Enhao Lu
- Key Laboratory of Smart Drug Delivery (Ministry of Education), School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Rui Wang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Yu Luo
- Key Laboratory of Smart Drug Delivery (Ministry of Education), School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Xianyi Sha
- Key Laboratory of Smart Drug Delivery (Ministry of Education), School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, China
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200030, China
| |
Collapse
|
12
|
Wang L, Zhang L, Chen F, Li Q, Zhu B, Tang Y, Yang Z, Cheng C, Qiu L, Ma L. Polymerized Network-Based Artificial Peroxisome Reprogramming Macrophages for Photoacoustic Imaging-Guided Treatment of Rheumatoid Arthritis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:25856-25868. [PMID: 38726921 DOI: 10.1021/acsami.4c04000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
Artificial peroxisomes (AP) with enzyme-mimetic catalytic activity and recruitment ability have drawn a great deal of attention in fabricating protocell systems for scavenging reactive oxygen species (ROS), modulating the inflammatory microenvironment, and reprogramming macrophages, which is of great potential in treating inflammatory diseases such as rheumatoid arthritis (RA). Herein, a macrophage membrane-cloaked Cu-coordinated polyphthalocyanine-based AP (CuAP) is prepared with a macrocyclic conjugated polymerized network and embedded Cu-single atomic active center, which mimics the catalytic activity and coordination environment of natural superoxide dismutase and catalase, possesses the inflammatory recruitment ability of macrophages, and performs photoacoustic imaging (PAI)-guided treatment. The results of both in vitro cellular and in vivo animal experiments demonstrated that the CuAP under ultrasound and microbubbles could efficiently scavenge excess ROS in cells and tissues, modulate microenvironmental inflammatory cytokines such as interleukin-1β, tumor necrosis factor-α, and arginase-1, and reprogram macrophages by polarization of M1 (proinflammatory phenotype) to M2 (anti-inflammatory phenotype). We believe this study offers a proof of concept for engineering multifaceted AP and a promising approach for a PAI-guided treatment platform for RA.
Collapse
Affiliation(s)
- Liyun Wang
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lingyan Zhang
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fan Chen
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Qian Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Bihui Zhu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuanjiao Tang
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhengbao Yang
- Department of Mechanical and Aerospace Engineering, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong 999077, China
| | - Chong Cheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Li Qiu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lang Ma
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
13
|
Zhang Y, Williams GR, Wang T, Zheng Y, Xu J, Nguyen VC, Yao L, Wang H, Zhu LM. Cisplatin-loaded mesoporous polydopamine nanoparticles capped with MnO 2 and coated with platelet membrane provide synergistic anti-tumor therapy. Int J Pharm 2024; 656:124093. [PMID: 38583822 DOI: 10.1016/j.ijpharm.2024.124093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/26/2024] [Accepted: 04/05/2024] [Indexed: 04/09/2024]
Abstract
A multifunctional nanoplatform was constructed in this work, with the goal of ameliorating the challenges faced with traditional cancer chemotherapy. Cisplatin (CP) was loaded into mesoporous polydopamine (mPDA) nanoparticles (NPs) with a drug loading of 15.8 ± 0.1 %, and MnO2 used as pore sealing agent. Finally, the NPs were wrapped with platelet membrane (PLTM). P-selectin on the PLTM can bind to CD44, which is highly expressed on the tumor cell membrane, so as to improve the targeting performance of the NPs. In addition, the CD47 on the PLTM can prevent the NPs from being phagocytosed by macrophages, which is conducive to immune escape. The final PLTM-CP@mPDA/MnO2 NPs were found to have a particle size of approximately 198 nm. MnO2 is degraded into Mn2+ in the tumor microenvironment, leading to CP release from the pores in the mPDA. CP both acts as a chemotherapy agent and can also increase the concentration of H2O2 in cells. Mn2+ can catalyze the conversion of H2O2 to OH, resulting in oxidative damage and chemodynamic therapy. In addition, Mn2+ can be used as a contrast agent in magnetic resonance imaging (MRI). In vitro and in vivo experiments were performed to explore the therapeutic effect of the NPs. When the concentration of CP is 30 μg/mL, the NPs cause approximately 50 % cell death. It was found that the PLTM-CP@mPDA/MnO2 NPs are targeted to cancerous cells, and in the tumor site cause extensive apoptosis. Tumor growth is thereby repressed. No negative off-target side effects were noted. MRI could be used to confirm the presence of the NPs in the tumor site. Overall, the nano-platform developed here provides cooperative chemotherapy and chemodynamic therapy, and can potentially be used for effective cancer treatment which could be monitored by MRI.
Collapse
Affiliation(s)
- Yanyan Zhang
- College of Biological Science and Medical Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, PR China
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Tong Wang
- College of Biological Science and Medical Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, PR China
| | - Yilu Zheng
- College of Biological Science and Medical Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, PR China
| | - Jianxiang Xu
- College of Biological Science and Medical Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, PR China
| | - Van Cuong Nguyen
- Faculty of Chemical Engineering, Industrial University of Ho Chi Minh City, 12 Nguyen Van Bao, Ho Chi Minh City 70000, Viet Nam
| | - Lili Yao
- Ri Zhao Central Hospital, Ri'zhao 276800, China.
| | - Haijun Wang
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an 271016, China.
| | - Li-Min Zhu
- College of Biological Science and Medical Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, PR China.
| |
Collapse
|
14
|
Moradi Hasan-Abad A, Shabankare A, Atapour A, Hamidi GA, Salami Zavareh M, Sobhani-Nasab A. The application of peroxidase mimetic nanozymes in cancer diagnosis and therapy. Front Pharmacol 2024; 15:1339580. [PMID: 38333005 PMCID: PMC10851941 DOI: 10.3389/fphar.2024.1339580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/16/2024] [Indexed: 02/10/2024] Open
Abstract
In recent decades, scholarly investigations have predominantly centered on nanomaterials possessing enzyme-like characteristics, commonly referred to as nanozymes. These nanozymes have emerged as viable substitutes for natural enzymes, offering simplicity, stability, and superior performance across various applications. Inorganic nanoparticles have been extensively employed in the emulation of enzymatic activity found in natural systems. Nanoparticles have shown a strong ability to mimic a number of enzyme-like functions. These systems have made a lot of progress thanks to the huge growth in nanotechnology research and the unique properties of nanomaterials. Our presentation will center on the kinetics, processes, and applications of peroxidase-like nanozymes. In this discourse, we will explore the various characteristics that exert an influence on the catalytic activity of nanozymes, with a particular emphasis on the prevailing problems and prospective consequences. This paper presents a thorough examination of the latest advancements achieved in the domain of peroxidase mimetic nanozymes in the context of cancer diagnosis and treatment. The primary focus is on their use in catalytic cancer therapy, alongside chemotherapy, phototherapy, sonodynamic therapy, radiation, and immunotherapy. The primary objective of this work is to offer theoretical and technical assistance for the prospective advancement of anticancer medications based on nanozymes. Moreover, it is anticipated that this will foster the investigation of novel therapeutic strategies aimed at achieving efficacious tumor therapy.
Collapse
Affiliation(s)
- Amin Moradi Hasan-Abad
- Autoimmune Diseases Research Center, Shahid Beheshti Hospital, Kashan University of Medical Sciences, Kashan, Iran
| | - Atefe Shabankare
- Islamic Azad University, Tehran Medical Sciences Branch, Tehran, Iran
| | - Amir Atapour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholam Ali Hamidi
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahmoud Salami Zavareh
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Sobhani-Nasab
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
15
|
Khan S, Falahati M, Cho WC, Vahdani Y, Siddique R, Sharifi M, Jaragh-Alhadad LA, Haghighat S, Zhang X, Ten Hagen TLM, Bai Q. Core-shell inorganic NP@MOF nanostructures for targeted drug delivery and multimodal imaging-guided combination tumor treatment. Adv Colloid Interface Sci 2023; 321:103007. [PMID: 37812992 DOI: 10.1016/j.cis.2023.103007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 08/16/2023] [Accepted: 09/23/2023] [Indexed: 10/11/2023]
Abstract
It is well known that metal-organic framework (MOF) nanostructures have unique characteristics such as high porosity, large surface areas and adjustable functionalities, so they are ideal candidates for developing drug delivery systems (DDSs) as well as theranostic platforms in cancer treatment. Despite the large number of MOF nanostructures that have been discovered, conventional MOF-derived nanosystems only have a single biofunctional MOF source with poor colloidal stability. Accordingly, developing core-shell MOF nanostructures with good colloidal stability is a useful method for generating efficient drug delivery, multimodal imaging and synergistic therapeutic systems. The preparation of core-shell MOF nanostructures has been done with a variety of materials, but inorganic nanoparticles (NPs) are highly effective for drug delivery and imaging-guided tumor treatment. Herein, we aimed to overview the synthesis of core-shell inorganic NP@MOF nanostructures followed by the application of core-shell MOFs derived from magnetic, quantum dots (QDs), gold (Au), and gadolinium (Gd) NPs in drug delivery and imaging-guided tumor treatment. Afterward, we surveyed different factors affecting prolonged drug delivery and cancer therapy, cellular uptake, biocompatibility, biodegradability, and enhanced permeation and retention (EPR) effect of core-shell MOFs. Last but not least, we discussed the challenges and the prospects of the field. We envision this article may hold great promise in providing valuable insights regarding the application of hybrid nanostructures as promising and potential candidates for multimodal imaging-guided combination cancer therapy.
Collapse
Affiliation(s)
- Suliman Khan
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mojtaba Falahati
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus MC, Rotterdam, the Netherlands.
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR, China
| | - Yasaman Vahdani
- Department of Biochemistry and Molecular Medicine, University of Montreal, Canada
| | - Rabeea Siddique
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Majid Sharifi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran; Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | | | - Setareh Haghighat
- Department of Microbiology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Xiaoju Zhang
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou, China
| | - Timo L M Ten Hagen
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus MC, Rotterdam, the Netherlands.
| | - Qian Bai
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
16
|
Peng X, Xu L, Zeng M, Dang H. Application and Development Prospect of Nanoscale Iron Based Metal-Organic Frameworks in Biomedicine. Int J Nanomedicine 2023; 18:4907-4931. [PMID: 37675409 PMCID: PMC10479543 DOI: 10.2147/ijn.s417543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/19/2023] [Indexed: 09/08/2023] Open
Abstract
Metal-organic frameworks (MOFs) are coordination polymers that comprise metal ions/clusters and organic ligands. MOFs have been extensively employed in different fields (eg, gas adsorption, energy storage, chemical separation, catalysis, and sensing) for their versatility, high porosity, and adjustable geometry. To be specific, Fe2+/Fe3+ exhibits unique redox chemistry, photochemical and electrical properties, as well as catalytic activity. Fe-based MOFs have been widely investigated in numerous biomedical fields over the past few years. In this study, the key index requirements of Fe-MOF materials in the biomedical field are summarized, and a conclusion is drawn in terms of the latest application progress, development prospects, and future challenges of Fe-based MOFs as drug delivery systems, antibacterial therapeutics, biocatalysts, imaging agents, and biosensors in the biomedical field.
Collapse
Affiliation(s)
- Xiujuan Peng
- Department of Clinical Laboratory, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, 621000, People’s Republic of China
| | - Li Xu
- Department of Clinical Laboratory, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, 621000, People’s Republic of China
| | - Min Zeng
- School of Materials and Chemistry, Southwest University of Science and Technology, Mianyang, Sichuan, 621010, People’s Republic of China
| | - Hao Dang
- Department of Clinical Laboratory, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, 621000, People’s Republic of China
| |
Collapse
|
17
|
Elmehrath S, Nguyen HL, Karam SM, Amin A, Greish YE. BioMOF-Based Anti-Cancer Drug Delivery Systems. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:953. [PMID: 36903831 PMCID: PMC10005089 DOI: 10.3390/nano13050953] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/19/2023] [Accepted: 03/03/2023] [Indexed: 06/18/2023]
Abstract
A variety of nanomaterials have been developed specifically for biomedical applications, such as drug delivery in cancer treatment. These materials involve both synthetic and natural nanoparticles and nanofibers of varying dimensions. The efficacy of a drug delivery system (DDS) depends on its biocompatibility, intrinsic high surface area, high interconnected porosity, and chemical functionality. Recent advances in metal-organic framework (MOF) nanostructures have led to the achievement of these desirable features. MOFs consist of metal ions and organic linkers that are assembled in different geometries and can be produced in 0, 1, 2, or 3 dimensions. The defining features of MOFs are their outstanding surface area, interconnected porosity, and variable chemical functionality, which enable an endless range of modalities for loading drugs into their hierarchical structures. MOFs, coupled with biocompatibility requisites, are now regarded as highly successful DDSs for the treatment of diverse diseases. This review aims to present the development and applications of DDSs based on chemically-functionalized MOF nanostructures in the context of cancer treatment. A concise overview of the structure, synthesis, and mode of action of MOF-DDS is provided.
Collapse
Affiliation(s)
- Sandy Elmehrath
- Department of Chemistry, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Ha L. Nguyen
- Department of Chemistry University of California—Berkeley, Kavli Energy Nanoscience Institute at UC Berkeley, and Berkeley Global Science Institute, Berkeley, CA 94720, USA
- Joint UAEU−UC Berkeley Laboratories for Materials Innovations, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Sherif M. Karam
- Department of Anatomy, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
- Zayed Centre for Health Sciences, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Amr Amin
- Zayed Centre for Health Sciences, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
- Department of Biology, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Yaser E. Greish
- Department of Chemistry, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
- Joint UAEU−UC Berkeley Laboratories for Materials Innovations, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
- Zayed Centre for Health Sciences, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| |
Collapse
|
18
|
Li Y, Wang R, Liu X, Li K, Xu Q. Recent advances in MOF-bio-interface: a review. NANOTECHNOLOGY 2023; 34:202002. [PMID: 36796094 DOI: 10.1088/1361-6528/acbc81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 02/15/2023] [Indexed: 06/18/2023]
Abstract
Metal-organic frameworks (MOFs), as a class of promising material with adjustable function and controllable structure, have been widely used in the food industry, chemical industry, biological medicine, and sensors. Biomacromolecules and living systems play a critical role in the world. However, the insufficiency in stability, recyclability, and efficiency, significantly impedes their further utilization in slightly harsh conditions. MOF-bio-interface engineering effectively address the above-mentioned shortages of biomacromolecules and living systems, and thereby attracting considerable attentions. Herein, we systematically review the achievements in the area of MOF-bio-interface. In particular, we summarize the interface between MOFs and proteins (enzymes and non-enzymatic proteins), polysaccharides, DNA, cells, microbes, and viruses. Meanwhile, we discuss the limitations of this approach and propose future research directions. We expect that this review could provide new insights and inspire new research efforts towards life science and material science.
Collapse
Affiliation(s)
- Yingfeng Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People's Republic of China
| | - Ru Wang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People's Republic of China
| | - Xue Liu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People's Republic of China
| | - Ke Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People's Republic of China
| | - Qing Xu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People's Republic of China
| |
Collapse
|
19
|
Ding Y, Pan Q, Gao W, Pu Y, Luo K, He B. Reactive oxygen species-upregulating nanomedicines towards enhanced cancer therapy. Biomater Sci 2023; 11:1182-1214. [PMID: 36606593 DOI: 10.1039/d2bm01833k] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Reactive oxygen species (ROS) play a crucial role in physiological and pathological processes, emerging as a therapeutic target in cancer. Owing to the high concentration of ROS in solid tumor tissues, ROS-based treatments, such as photodynamic therapy and chemodynamic therapy, and ROS-responsive drug delivery systems have been widely explored to powerfully and specifically suppress tumors. However, their anticancer efficacy is still hampered by the heterogeneous ROS levels, and thus comprehensively upregulating the ROS levels in tumor tissues can ensure an enhanced therapeutic effect, which can further sensitize and/or synergize with other therapies to inhibit tumor growth and metastasis. Herein, we review the recently emerging drug delivery strategies and technologies for increasing the H2O2, ˙OH, 1O2, and ˙O2- concentrations in cancer cells, including the efficient delivery of natural enzymes, nanozymes, small molecular biological molecules, and nanoscale Fenton-reagents and semiconductors and neutralization of intracellular antioxidant substances and localized input of mechanical and electromagnetic waves (such as ultrasound, near infrared light, microwaves, and X-rays). The applications of these ROS-upregulating nanosystems in enhancing and synergizing cancer therapies including chemotherapy, chemodynamic therapy, phototherapy, and immunotherapy are surveyed. In addition, we discuss the challenges of ROS-upregulating systems and the prospects for future studies.
Collapse
Affiliation(s)
- Yuanyuan Ding
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| | - Qingqing Pan
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Wenxia Gao
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325027, China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and molecular imaging Key Laboratory of Sichuan Province, Sichuan University, Chengdu 610041, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
20
|
Zheng J, Huang J, Zhang L, Wang M, Xu L, Dou X, Leng X, Fang M, Sun Y, Wang Z. Drug-loaded microbubble delivery system to enhance PD-L1 blockade immunotherapy with remodeling immune microenvironment. Biomater Res 2023; 27:9. [PMID: 36759928 PMCID: PMC9909878 DOI: 10.1186/s40824-023-00350-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/29/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Although programmed cell death protein 1 (PD-1)/ programmed cell death-ligand protein 1 (PD-L1) checkpoint blockade immunotherapy demonstrates great promise in cancer treatment, poor infiltration of T cells resulted from tumor immunosuppressive microenvironment (TIME) and insufficient accumulation of anti-PD-L1 (αPD-L1) in tumor sites diminish the immune response. Herein, we reported a drug-loaded microbubble delivery system to overcome these obstacles and enhance PD-L1 blockade immunotherapy. METHODS Docetaxel (DTX) and imiquimod (R837)-loaded microbubbles (RD@MBs) were synthesized via a typical rotary evaporation method combined with mechanical oscillation. The targeted release of drugs was achieved by using the directional "bursting" capability of ultrasound-targeted microbubble destruction (UTMD) technology. The antitumor immune response by RD@MBs combining αPD-L1 were evaluated on 4T1 and CT26 tumor models. RESULTS The dying tumor cells induced by DTX release tumor-associated antigens (TAAs), together with R837, promoted the activation, proliferation and recruitment of T cells. Besides, UTMD technology and DTX enhanced the accumulation of αPD-L1 in tumor sites. Moreover, RD@MBs remolded TIME, including the polarization of M2-phenotype tumor-associated macrophages (TAMs) to M1-phenotype, and reduction of myeloid-derived suppressor cells (MDSCs). The RD@MBs + αPD-L1 synergistic therapy not only effectively inhibited the growth of primary tumors, but also significantly inhibited the mimic distant tumors as well as lung metastases. CONCLUSION PD-L1 blockade immunotherapy was enhanced by RD@MBs delivery system.
Collapse
Affiliation(s)
- Jun Zheng
- grid.412461.40000 0004 9334 6536State Key Laboratory of Ultrasound in Medicine and Engineering, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| | - Ju Huang
- grid.412461.40000 0004 9334 6536State Key Laboratory of Ultrasound in Medicine and Engineering, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| | - Liang Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China. .,Ultrasound Department, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, People's Republic of China.
| | - Mengna Wang
- grid.203458.80000 0000 8653 0555Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016 People’s Republic of China
| | - Lihong Xu
- grid.203458.80000 0000 8653 0555Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016 People’s Republic of China
| | - Xiaoyun Dou
- grid.203458.80000 0000 8653 0555Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016 People’s Republic of China
| | - Xiaojing Leng
- grid.412461.40000 0004 9334 6536State Key Laboratory of Ultrasound in Medicine and Engineering, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| | - Mingxiao Fang
- grid.412461.40000 0004 9334 6536State Key Laboratory of Ultrasound in Medicine and Engineering, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010 People’s Republic of China
| | - Yang Sun
- State Key Laboratory of Ultrasound in Medicine and Engineering, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China.
| | - Zhigang Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China.
| |
Collapse
|
21
|
Recent Advances in Metal-Organic Framework (MOF) Asymmetric Membranes/Composites for Biomedical Applications. Symmetry (Basel) 2023. [DOI: 10.3390/sym15020403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Metal-organic frameworks (MOFs) are a new class of porous crystalline materials composed of metal and organic material. MOFs have fascinating properties, such as fine tunability, large specific surface area, and high porosity. MOFs are widely used for environmental protection, biosensors, regenerative medicine, medical engineering, cell therapy, catalysts, and drug delivery. Recent studies have reported various significant properties of MOFs for biomedical applications, such as drug detection and delivery. In contrast, MOFs have limitations such as low stability and low specificity in binding to the target. MOF-based membranes improve the stability and specificity of conventional MOFs by increasing the surface area and developing the possibility of MOF-ligand binding, while conjugated membranes dramatically increase the area of active functional groups. This special property makes them attractive for drug and biosensor fabrication, as both the spreading and solubility components of the porosity can be changed. Asymmetric membranes are a structure with high potential in the biomedical field, due to the different characteristics on its two surfaces, the possibility of adjusting various properties such as the size of porosity, transfer rate and selectivity, and surface properties such as hydrophilicity and hydrophobicity. MOF assisted asymmetric membranes can provide a platform with different properties and characteristics in the biomedical field. The latest version of MOF materials/membranes has several potential applications, especially in medical engineering, cell therapy, drug delivery, and regenerative medicine, which will be discussed in this review, along with their advantages, disadvantages, and challenges.
Collapse
|
22
|
Qin Y, Geng X, Sun Y, Zhao Y, Chai W, Wang X, Wang P. Ultrasound nanotheranostics: Toward precision medicine. J Control Release 2023; 353:105-124. [PMID: 36400289 DOI: 10.1016/j.jconrel.2022.11.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/24/2022]
Abstract
Ultrasound (US) is a mechanical wave that can penetrate biological tissues and trigger complex bioeffects. The mechanisms of US in different diagnosis and treatment are different, and the functional application of commercial US is also expanding. In particular, recent developments in nanotechnology have led to a wider use of US in precision medicine. In this review, we focus on US in combination with versatile micro and nanoparticles (NPs)/nanovesicles for tumor theranostics. We first introduce US-assisted drug delivery as a stimulus-responsive approach that spatiotemporally regulates the deposit of nanomedicines in target tissues. Multiple functionalized NPs and their US-regulated drug-release curves are analyzed in detail. Moreover, as a typical representative of US therapy, sonodynamic antitumor strategy is attracting researchers' attention. The collaborative efficiency and mechanisms of US and various nano-sensitizers such as nano-porphyrins and organic/inorganic nanosized sensitizers are outlined in this paper. A series of physicochemical processes during ultrasonic cavitation and NPs activation are also discussed. Finally, the new applications of US and diagnostic NPs in tumor-monitoring and image-guided combined therapy are summarized. Diagnostic NPs contain substances with imaging properties that enhance US contrast and photoacoustic imaging. The development of such high-resolution, low-background US-based imaging methods has contributed to modern precision medicine. It is expected that the integration of non-invasive US and nanotechnology will lead to significant breakthroughs in future clinical applications.
Collapse
Affiliation(s)
- Yang Qin
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xiaorui Geng
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yue Sun
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yitong Zhao
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Wenyu Chai
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xiaobing Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| | - Pan Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
23
|
Liang Z, Li X, Chen X, Zhou J, Li Y, Peng J, Lin Z, Liu G, Zeng X, Li C, Hang L, Li H. Fe/MOF based platform for NIR laser induced efficient PDT/PTT of cancer. Front Bioeng Biotechnol 2023; 11:1156079. [PMID: 37064235 PMCID: PMC10098195 DOI: 10.3389/fbioe.2023.1156079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/15/2023] [Indexed: 04/18/2023] Open
Abstract
Introduction: Photodynamic therapy (PDT) and photothermal therapy (PTT) are widely used in the treatment of tumors. However, their application in the treatment of clinical tumors is limited by the complexity and irreversible hypoxia environment generated by tumor tissues. To overcome this limitation, a nanoparticle composed of indocyanine green (ICG) and Fe-MOF-5 was developed. Methods: We prepared F-I@FM5 and measured its morphology, particle size, and stability. Its enzyme like ability and optical effect was verified. Then we used MTT, staining and flow cytometry to evaluated the anti-tumor effect on EMT-6 cells in vitro. Finally, the anti-tumor effect in vivo has been studied on EMT-6 tumor bearing mice. Results: For the composite nanoparticle, we confirmed that Fe-MOF-5 has the best nanozyme activity. In addition, it has excellent photothermal conversion efficiency and generates reactive oxygen species (ROS) under near-infrared light irradiation (808 nm). The composite nanoparticle showed good tumor inhibition effect in vitro and in vivo, which was superior to the free ICG or Fe-MOF-5 alone. Besides, there was no obvious cytotoxicity in major organs within the effective therapeutic concentration. Discussion: Fe-MOF-5 has the function of simulating catalase, which can promote the decomposition of excessive H2O2 in the tumor microenvironment and produce oxygen to improve the hypoxic environment. The improvement of tumor hypoxia can enhance the efficacy of PDT and PTT. This research not only provides an efficient and stable anti-tumor nano platform, but also has broad application prospects in the field of tumor therapy, and provides a new idea for the application of MOF as an important carrier material in the field of photodynamic therapy.
Collapse
Affiliation(s)
- Zixing Liang
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiaofeng Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiaofang Chen
- Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jiawei Zhou
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Yanan Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Jianhui Peng
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Zhousheng Lin
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Gai Liu
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiancheng Zeng
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Cheng Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
- Jinan University, Guangzhou, China
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Hainan, China
- *Correspondence: Hailiang Li, ; Cheng Li, ; Lifeng Hang,
| | - Lifeng Hang
- Guangdong Second Provincial General Hospital, Guangzhou, China
- *Correspondence: Hailiang Li, ; Cheng Li, ; Lifeng Hang,
| | - Hailiang Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
- *Correspondence: Hailiang Li, ; Cheng Li, ; Lifeng Hang,
| |
Collapse
|
24
|
Engineering a hierarchical carbon supported magnetite nanoparticles composite from metal organic framework and graphene oxide for lithium-ion storage. J Colloid Interface Sci 2023; 630:86-98. [DOI: 10.1016/j.jcis.2022.10.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/23/2022] [Accepted: 10/17/2022] [Indexed: 11/21/2022]
|
25
|
Ma R, Hu X, Zhang X, Wang W, Sun J, Su Z, Zhu C. Strategies to prevent, curb and eliminate biofilm formation based on the characteristics of various periods in one biofilm life cycle. Front Cell Infect Microbiol 2022; 12:1003033. [PMID: 36211965 PMCID: PMC9534288 DOI: 10.3389/fcimb.2022.1003033] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Biofilms are colonies of bacteria embedded inside a complicated self-generating intercellular. The formation and scatter of a biofilm is an extremely complex and progressive process in constant cycles. Once formed, it can protect the inside bacteria to exist and reproduce under hostile conditions by establishing tolerance and resistance to antibiotics as well as immunological responses. In this article, we reviewed a series of innovative studies focused on inhibiting the development of biofilm and summarized a range of corresponding therapeutic methods for biological evolving stages of biofilm. Traditionally, there are four stages in the biofilm formation, while we systematize the therapeutic strategies into three main periods precisely:(i) period of preventing biofilm formation: interfering the colony effect, mass transport, chemical bonds and signaling pathway of plankton in the initial adhesion stage; (ii) period of curbing biofilm formation:targeting several pivotal molecules, for instance, polysaccharides, proteins, and extracellular DNA (eDNA) via polysaccharide hydrolases, proteases, and DNases respectively in the second stage before developing into irreversible biofilm; (iii) period of eliminating biofilm formation: applying novel multifunctional composite drugs or nanoparticle materials cooperated with ultrasonic (US), photodynamic, photothermal and even immune therapy, such as adaptive immune activated by stimulated dendritic cells (DCs), neutrophils and even immunological memory aroused by plasmocytes. The multitargeted or combinational therapies aim to prevent it from developing to the stage of maturation and dispersion and eliminate biofilms and planktonic bacteria simultaneously.
Collapse
Affiliation(s)
| | | | | | | | | | - Zheng Su
- *Correspondence: Chen Zhu, ; Zheng Su,
| | - Chen Zhu
- *Correspondence: Chen Zhu, ; Zheng Su,
| |
Collapse
|
26
|
Wang W, Yu Y, Jin Y, Liu X, Shang M, Zheng X, Liu T, Xie Z. Two-dimensional metal-organic frameworks: from synthesis to bioapplications. J Nanobiotechnology 2022; 20:207. [PMID: 35501794 PMCID: PMC9059454 DOI: 10.1186/s12951-022-01395-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/23/2022] [Indexed: 12/19/2022] Open
Abstract
As a typical class of crystalline porous materials, metal-organic framework possesses unique features including versatile functionality, structural and compositional tunability. After being reduced to two-dimension, ultrathin metal-organic framework layers possess more external excellent properties favoring various technological applications. In this review article, the unique structural properties of the ultrathin metal-organic framework nanosheets benefiting from the planar topography were highlighted, involving light transmittance, and electrical conductivity. Moreover, the design strategy and versatile fabrication methodology were summarized covering discussions on their applicability and accessibility, especially for porphyritic metal-organic framework nanosheet. The current achievements in the bioapplications of two-dimensional metal-organic frameworks were presented comprising biocatalysis, biosensor, and theranostic, with an emphasis on reactive oxygen species-based nanomedicine for oncology treatment. Furthermore, current challenges confronting the utilization of two-dimensional metal-organic frameworks and future opportunities in emerging research frontiers were presented.
Collapse
Affiliation(s)
- Weiqi Wang
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Yuting Yu
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Yilan Jin
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Xiao Liu
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Min Shang
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Xiaohua Zheng
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu Province, China.
| | - Tingting Liu
- Department of Medical Imaging, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China.
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.
| |
Collapse
|
27
|
Falahati M, Sharifi M, Hagen TLMT. Explaining chemical clues of metal organic framework-nanozyme nano-/micro-motors in targeted treatment of cancers: benchmarks and challenges. J Nanobiotechnology 2022; 20:153. [PMID: 35331244 PMCID: PMC8943504 DOI: 10.1186/s12951-022-01375-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/12/2022] [Indexed: 02/07/2023] Open
Abstract
Nowadays, nano-/micro-motors are considered as powerful tools in different areas ranging from cleaning all types of contaminants, to development of Targeted drug delivery systems and diagnostic activities. Therefore, the development and application of nano-/micro-motors based on metal-organic frameworks with nanozyme activity (abbreviated as: MOF-NZs) in biomedical activities have received much interest recently. Therefore, after investigating the catalytic properties and applications of MOF-NZs in the treatment of cancer, this study intends to point out their key role in the production of biocompatible nano-/micro-motors. Since reducing the toxicity of MOF-NZ nano-/micro-motors can pave the way for medical activities, this article examines the methods of making biocompatible nanomotors to address the benefits and drawbacks of the required propellants. In the following, an analysis of the amplified directional motion of MOF-NZ nano-/micro-motors under physiological conditions is presented, which can improve the motor behaviors in the propulsion function, conductivity, targeting, drug release, and possible elimination. Meanwhile, by explaining the use of MOF-NZ nano-/micro-motors in the treatment of cancer through the possible synergy of nanomotors with different therapies, it was revealed that MOF-NZ nano-/micro-motors can be effective in the treatment of cancer. Ultimately, by analyzing the potential challenges of MOF-NZ nano-/micro-motors in the treatment of cancers, we hope to encourage researchers to develop MOF-NZs-based nanomotors, in addition to opening up new ideas to address ongoing problems.
Collapse
Affiliation(s)
- Mojtaba Falahati
- Laboratory Experimental Oncology, Department of Pathology, Erasmus MC, 3015GD, Rotterdam, The Netherlands.
| | - Majid Sharifi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.
- Depatment of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.
| | - Timo L M Ten Hagen
- Laboratory Experimental Oncology, Department of Pathology, Erasmus MC, 3015GD, Rotterdam, The Netherlands.
| |
Collapse
|
28
|
Liu C, Ren L, Li X, Fan N, Chen J, Zhang D, Yang W, Ding S, Xu W, Min X. Self-electrochemiluminescence biosensor based on CRISPR/Cas12a and PdCuBP@luminol nanoemitter for highly sensitive detection of cytochrome c oxidase subunit III gene of acute kidney injury. Biosens Bioelectron 2022; 207:114207. [PMID: 35339823 DOI: 10.1016/j.bios.2022.114207] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 03/06/2022] [Accepted: 03/19/2022] [Indexed: 12/23/2022]
Abstract
The cytochrome c oxidase subunit III (COX III) gene is a powerful biomarker for the early diagnosis of acute kidney injury. However, current methods for COX III gene detection are usually laborious and time-consuming, with limited sensitivity. Herein, we report a novel self-electrochemiluminescence (ECL) biosensor for highly sensitive detection of the COX III gene based on CRISPR/Cas12a and nanoemitters of luminol-loaded multicomponent metal-metalloid PdCuBP alloy mesoporous nanoclusters. The nanoemitter with excellent self-ECL in neutral media exhibited a high specific surface area for binding luminol and outstanding oxidase-like catalytic activity toward dissolved O2. Meanwhile, the CRISPR/Cas12a system, as a target-trigger, was employed to specifically recognize the COX III gene and efficiently cleave the interfacial quencher of dopamine-labeled hairpin DNA. As a result, the ECL biosensor showed superior analytical performance for COX III gene detection without exogenous coreactant. Benefiting from the high-efficiency ECL emission of the nanoemitter and Cas12a-mediated interfacial cleavage of the quencher, the developed ECL biosensor exhibited high sensitivity to COX III with a low detection limit of 0.18 pM. The established ECL biosensing method possessed excellent practical performance in urine samples. Meaningfully, the proposed strategy presents promising prospects for nucleic acid detection in the field of clinical diagnostics.
Collapse
Affiliation(s)
- Changjin Liu
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, China; Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Lei Ren
- Health Management Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xinmin Li
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Ningke Fan
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Junman Chen
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Decai Zhang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Wei Yang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Shijia Ding
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Wenchun Xu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China.
| | - Xun Min
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, China.
| |
Collapse
|
29
|
Lou H, Chu L, Zhou W, Dou J, Teng X, Tan W, Zhou B. Diselenium-bridged covalent organic framework with pH/GSH/photo-triple-responsiveness for highly controlled drug release toward joint chemo/photothermal/chemodynamic cancer therapy. J Mater Chem B 2022; 10:7955-7966. [PMID: 35792081 DOI: 10.1039/d2tb01015a] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Here, a novel joint chemo/photothermal/chemodynamic therapy was developed using a pH/GSH/photo triple-responsive 2D-covalent organic framework (COFs) drug carriers for passive target treatment of tumor with extraordinarily high efficiency. The well-designed...
Collapse
Affiliation(s)
- Han Lou
- School of Pharmacy, Weifang Medical University, Weifang, 261053, Shandong, P. R. China.
- Department of Urology, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Shandong, P. R. China
| | - Lichao Chu
- Department of Anesthesiology, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, 261031, Shandong, P. R. China
| | - Wenbin Zhou
- Department of Urology, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Shandong, P. R. China
| | - Jinli Dou
- School of Pharmacy, Weifang Medical University, Weifang, 261053, Shandong, P. R. China.
| | - Xiaotong Teng
- Department of Respiratory Medicine, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, 261031, Shandong, P. R. China
| | - Wei Tan
- Department of Respiratory Medicine, The First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, 261031, Shandong, P. R. China
| | - Baolong Zhou
- School of Pharmacy, Weifang Medical University, Weifang, 261053, Shandong, P. R. China.
| |
Collapse
|
30
|
Liu H, Tao Y, Wu T, Li H, Zhang X, Huang F, Bian H. A {Zn
5
} cluster‐based metal–organic framework: Multifunctional detection of Ag
+
, Cr
2
O
7
2−
, and 2,4,6‐trinitrophenol (TNP). Appl Organomet Chem 2022. [DOI: 10.1002/aoc.6456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Han‐Fu Liu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences Guangxi Normal University Guilin China
| | - Ye Tao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences Guangxi Normal University Guilin China
| | - Tai‐Xue Wu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences Guangxi Normal University Guilin China
| | - Hai‐Ye Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences Guangxi Normal University Guilin China
| | - Xiu‐Qing Zhang
- College of Chemistry and Bioengineering, Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials Guilin University of Technology Guilin China
| | - Fu‐Ping Huang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences Guangxi Normal University Guilin China
| | - He‐Dong Bian
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences Guangxi Normal University Guilin China
- School of Chemistry and Chemical Engineering, Guangxi University for Nationalities Key Laboratory of Chemistry and Engineering of Forest Products Nanning China
| |
Collapse
|
31
|
Shi Q, Yu T, Wu R, Liu J. Metal-Support Interactions of Single-Atom Catalysts for Biomedical Applications. ACS APPLIED MATERIALS & INTERFACES 2021; 13:60815-60836. [PMID: 34913673 DOI: 10.1021/acsami.1c18797] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The development of single-atom catalysts (SACs) has become a rapidly growing research field. It is a critical challenge to understand the interactions between the single-atom metal active sites and the support materials. Recently, original research reports of SACs in biomedical applications have emerged in the literature, yet this topic has seldom been reviewed. Here, this review focuses on the latest advances in single-atom catalysis for biomedical applications and highlights the keys for the design of SACs, such as understanding the interactions between metals and supports and classifying various enzyme-like activities. This review helps bridge the knowledge of multiple disciplines and provides prospects regarding the development of SACs for biomedicine.
Collapse
Affiliation(s)
- Qiaolan Shi
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215006, Jiangsu, P. R. China
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Tianrong Yu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215006, Jiangsu, P. R. China
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Renfei Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215006, Jiangsu, P. R. China
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Jian Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215006, Jiangsu, P. R. China
| |
Collapse
|
32
|
Shin Low S, Nong Lim C, Yew M, Siong Chai W, Low LE, Manickam S, Ti Tey B, Show PL. Recent ultrasound advancements for the manipulation of nanobiomaterials and nanoformulations for drug delivery. ULTRASONICS SONOCHEMISTRY 2021; 80:105805. [PMID: 34706321 PMCID: PMC8555278 DOI: 10.1016/j.ultsonch.2021.105805] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/08/2021] [Accepted: 10/20/2021] [Indexed: 05/04/2023]
Abstract
Recent advances in ultrasound (US) have shown its great potential in biomedical applications as diagnostic and therapeutic tools. The coupling of US-assisted drug delivery systems with nanobiomaterials possessing tailor-made functions has been shown to remove the limitations of conventional drug delivery systems. The low-frequency US has significantly enhanced the targeted drug delivery effect and efficacy, reducing limitations posed by conventional treatments such as a limited therapeutic window. The acoustic cavitation effect induced by the US-mediated microbubbles (MBs) has been reported to replace drugs in certain acute diseases such as ischemic stroke. This review briefly discusses the US principles, with particular attention to the recent advancements in drug delivery applications. Furthermore, US-assisted drug delivery coupled with nanobiomaterials to treat different diseases (cancer, neurodegenerative disease, diabetes, thrombosis, and COVID-19) are discussed in detail. Finally, this review covers the future perspectives and challenges on the applications of US-mediated nanobiomaterials.
Collapse
Affiliation(s)
- Sze Shin Low
- Continental-NTU Corporate Lab, Nanyang Technological University, 50 Nanyang Drive, Singapore 637553, Singapore; Department of Chemical and Environmental Engineering, Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, Semenyih 43500, Selangor Darul Ehsan, Malaysia
| | - Chang Nong Lim
- School of Engineering and Physical Sciences, Heriot-Watt University Malaysia, No. 1, Jalan Venna P5/2, Precinct 5, Putrajaya 62200, Malaysia
| | - Maxine Yew
- Department of Mechanical, Materials and Manufacturing Engineering, University of Nottingham Ningbo China, 199 Taikang East Road, Ningbo 315100, Zhejiang, China
| | - Wai Siong Chai
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, Guangdong, China
| | - Liang Ee Low
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; Advanced Engineering Platform, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China.
| | - Sivakumar Manickam
- Petroleum and Chemical Engineering, Faculty of Engineering, Universiti Teknologi Brunei, Jalan Tungku Link Gadong, Bandar Seri Begawan, BE1410, Brunei Darussalam.
| | - Beng Ti Tey
- Advanced Engineering Platform, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; Chemical Engineering Discipline, School of Engineering, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
| | - Pau Loke Show
- Department of Chemical and Environmental Engineering, Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, Semenyih 43500, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
33
|
Thangudu S, Su CH. Peroxidase Mimetic Nanozymes in Cancer Phototherapy: Progress and Perspectives. Biomolecules 2021; 11:1015. [PMID: 34356639 PMCID: PMC8301984 DOI: 10.3390/biom11071015] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/31/2022] Open
Abstract
Nanomaterial-mediated cancer therapeutics is a fast developing field and has been utilized in potential clinical applications. However, most effective therapies, such as photodynamic therapy (PDT) and radio therapy (RT), are strongly oxygen-dependent, which hinders their practical applications. Later on, several strategies were developed to overcome tumor hypoxia, such as oxygen carrier nanomaterials and oxygen generated nanomaterials. Among these, oxygen species generation on nanozymes, especially catalase (CAT) mimetic nanozymes, convert endogenous hydrogen peroxide (H2O2) to oxygen (O2) and peroxidase (POD) mimetic nanozymes converts endogenous H2O2 to water (H2O) and reactive oxygen species (ROS) in a hypoxic tumor microenvironment is a fascinating approach. The present review provides a detailed examination of past, present and future perspectives of POD mimetic nanozymes for effective oxygen-dependent cancer phototherapeutics.
Collapse
Affiliation(s)
- Suresh Thangudu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
| | - Chia-Hao Su
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| |
Collapse
|