1
|
Li S, Wu Q, Chen Z, Guo W, Tan L, Ren X, Fu C, Jiang G, Huang Z, Meng X. Synchronous Interference of Dual Metabolic Pathways Mediated by H 2S Gas/GOx for Augmenting Tumor Microwave Thermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 39798012 DOI: 10.1021/acsami.4c18641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2025]
Abstract
Sublethal tumor cells have an urgent need for energy, making it common for them to switch metabolic phenotypes between glycolysis and oxidative phosphorylation (OXPHOS) for compensatory energy supply; thus, the synchronous interference of dual metabolic pathways for limiting energy level is essential in inhibiting sublethal tumor growth. Herein, a multifunctional nanoplatform of Co-MOF-loaded anethole trithione (ADT) and myristyl alcohol (MA), modified with GOx and hyaluronic acid (HA) was developed, namely, CAMGH. It could synchronously interfere with dual metabolic pathways including glycolysis and OXPHOS to restrict the adenosine triphosphate (ATP) supply, achieving the inhibition to sublethal tumors after microwave (MW) thermal therapy. Under low-power MW irradiation, CAMGH induced certain tumor thermal damage while ensuring the safety of the surrounding normal tissues. The loaded GOx consumed glucose in tumors, undoubtedly blocking the main energy supply pathway, the glycolytic pathway. Then, H2O2 generated from GOx reacted with Co2+ to produce cytotoxic ·OH, combining with the released H2S from ADT to co-obstruct OXPHOS and then synergy with the above glycolysis blocking for a more effective ATP inhibition. The powerful depletion of ATP caused significant suppression of damage resistance protein upregulated after thermal stimulation, i.e., HSP90, and then the activation of caspase-3, achieving the simultaneous reversal of heat resistance and apoptosis resistance. Altogether, the CAMGH-based synchronous interference of dual metabolic pathways shows the potential to break down tumor self-repair, presenting an alternative strategy to enhance the therapeutic effect of tumor MW thermal therapy.
Collapse
Affiliation(s)
- Shimei Li
- Key Laboratory of Cryogenics Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Qiong Wu
- Key Laboratory of Cryogenics Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Zengzhen Chen
- Key Laboratory of Cryogenics Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Wenna Guo
- Key Laboratory of Cryogenics Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- Guangdong Second Provincial General Hospital, Jinan University, Guangzhou 510310, China
| | - Longfei Tan
- Key Laboratory of Cryogenics Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiangling Ren
- Key Laboratory of Cryogenics Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Changhui Fu
- Key Laboratory of Cryogenics Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Guihua Jiang
- Guangdong Second Provincial General Hospital, Jinan University, Guangzhou 510310, China
| | - Zhongbing Huang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Xianwei Meng
- Key Laboratory of Cryogenics Science and Technology, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
2
|
Liang S, Liu Y, Zhu H, Liao G, Zhu W, Zhang L. Emerging nitric oxide gas-assisted cancer photothermal treatment. EXPLORATION (BEIJING, CHINA) 2024; 4:20230163. [PMID: 39713202 PMCID: PMC11655315 DOI: 10.1002/exp.20230163] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/08/2024] [Indexed: 12/24/2024]
Abstract
Photothermal therapy (PTT) has garnered significant attention in recent years, but the standalone application of PTT still faces limitations that hinder its ability to achieve optimal therapeutic outcomes. Nitric oxide (NO), being one of the most extensively studied gaseous molecules, presents itself as a promising complementary candidate for PTT. In response, various nanosystems have been developed to enable the simultaneous utilization of PTT and NO-mediated gas therapy (GT), with the integration of photothermal agents (PTAs) and thermally-sensitive NO donors being the prevailing approach. This combination seeks to leverage the synergistic effects of PTT and GT while mitigating the potential risks associated with gas toxicity through the use of a single laser irradiation. Furthermore, additional internal or external stimuli have been employed to trigger NO release when combined with different types of PTAs, thereby further enhancing therapeutic efficacy. This comprehensive review aims to summarize recent advancements in NO gas-assisted cancer photothermal treatment. It commences by providing an overview of various types of NO donors and precursors, including those sensitive to photothermal, light, ultrasound, reactive oxygen species, and glutathione. These NO donors and precursors are discussed in the context of dual-modal PTT/GT. Subsequently, the incorporation of other treatment modalities such as chemotherapy (CHT), photodynamic therapy (PDT), alkyl radical therapy, radiation therapy, and immunotherapy (IT) in the creation of triple-modal therapeutic nanoplatforms is presented. The review further explores tetra-modal therapies, such as PTT/GT/CHT/PDT, PTT/GT/CHT/chemodynamic therapy (CDT), PTT/GT/PDT/IT, PTT/GT/starvation therapy (ST)/IT, PTT/GT/Ca2+ overload/IT, PTT/GT/ferroptosis (FT)/IT, and PTT/GT/CDT/IT. Finally, potential challenges and future perspectives concerning these novel paradigms are discussed. This comprehensive review is anticipated to serve as a valuable resource for future studies focused on the development of innovative photothermal/NO-based cancer nanotheranostics.
Collapse
Affiliation(s)
- Shuang Liang
- Department of Radiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yufei Liu
- Department of Radiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hongquan Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Guangfu Liao
- College of Material EngineeringFujian Agriculture and Forestry UniversityFuzhouChina
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Li Zhang
- Department of Critical Care MedicineShenzhen HospitalSouthern Medical UniversityShenzhenGuangdongChina
- Department of ChemistryCity University of Hong KongKowloonHong Kong SARChina
| |
Collapse
|
3
|
Opoku-Damoah Y, Xu ZP, Ta HT, Zhang R. Ultrasound-Responsive Lipid Nanoplatform with Nitric Oxide and Carbon Monoxide Release for Cancer Sono-Gaso-Therapy. ACS APPLIED BIO MATERIALS 2024; 7:7585-7594. [PMID: 39509170 DOI: 10.1021/acsabm.4c01165] [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: 11/15/2024]
Abstract
Local gas therapy is emerging as a potential cancer treatment approach due to its specificity as gas-containing molecules can be packed into a nanodelivery system to release the corresponding gaseous molecules around the tumor site upon a suitable stimulus. Single-gas therapy has been reported, while synergistic dual-gas therapy has rarely been reported. Herein, we report a dual-gas-containing nanoplatform for synergistic cancer gasotherapy upon ultrasound irradiation. First, a robust ultrasound-responsive lipid-coated nanosystem was prepared with suitable particle size and characteristics. A low-intensity ultrasound (1.25 W/cm2) was found to simultaneously modulate carbon monoxide (CO) and nitric oxide (NO) release from the nanosystem in media and CT26 colon cancer cells for efficient therapeutic effect. The intracellular release promoted the overgeneration of reactive oxygen species (ROS) and triggered cancer cell apoptosis synergistically. The in vivo test demonstrated that the optimal dual-gas-containing formulation efficiently inhibited tumor growth (by ∼87%) at relatively low doses upon ultrasound irradiation (1.25 W/cm2, 5 min). This therapeutic efficacy shows that the current responsive lipid-coated delivery system has potential for ultrasound-triggered dual-gas therapy of both superficially and deeply seated cancers.
Collapse
Affiliation(s)
- Yaw Opoku-Damoah
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- Institute of Biomedical Health Technology and Engineering and Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, P. R. China
| | - Hang T Ta
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
- Queensland Micro and Nanotechnology Centre, Griffith University, Brisbane, QLD 4111, Australia
| | - Run Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
4
|
Ouyang Q, Zhao Y, Xu K, He Y, Qin M. Hyaluronic Acid Receptor-Mediated Nanomedicines and Targeted Therapy. SMALL METHODS 2024; 8:e2400513. [PMID: 39039982 DOI: 10.1002/smtd.202400513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/25/2024] [Indexed: 07/24/2024]
Abstract
Hyaluronic acid (HA) is a naturally occurring polysaccharide found in the extracellular matrix with broad applications in disease treatment. HA possesses good biocompatibility, biodegradability, and the ability to interact with various cell surface receptors. Its wide range of molecular weights and modifiable chemical groups make it an effective drug carrier for drug delivery. Additionally, the overexpression of specific receptors for HA on cell surfaces in many disease states enhances the accumulation of drugs at pathological sites through receptor binding. In this review, the modification of HA with drugs, major receptor proteins, and the latest advances in receptor-targeted nano drug delivery systems (DDS) for the treatment of tumors and inflammatory diseases are summarized. Furthermore, the functions of HA with varying molecular weights of HA in vivo and the selection of drug delivery methods for different diseases are discussed.
Collapse
Affiliation(s)
- Qiuhong Ouyang
- Department of Lung Cancer Center and Center for Preclinical Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Zhao
- Department of Lung Cancer Center and Center for Preclinical Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kunyao Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yuechen He
- Department of Lung Cancer Center and Center for Preclinical Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Qin
- Department of Lung Cancer Center and Center for Preclinical Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
5
|
Chu B, Deng H, Niu T, Qu Y, Qian Z. Stimulus-Responsive Nano-Prodrug Strategies for Cancer Therapy: A Focus on Camptothecin Delivery. SMALL METHODS 2024; 8:e2301271. [PMID: 38085682 DOI: 10.1002/smtd.202301271] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/15/2023] [Indexed: 08/18/2024]
Abstract
Camptothecin (CPT) is a highly cytotoxic molecule with excellent antitumor activity against various cancers. However, its clinical application is severely limited by poor water solubility, easy inactivation, and severe toxicity. Structural modifications and nanoformulations represent two crucial avenues for camptothecin's development. However, the potential for further structural modifications is limited, and camptothecin nanoparticles fabricated via physical loading have the drawbacks of low drug loading and leakage. Prodrug-based CPT nanoformulations have shown unique advantages, including increased drug loading, reduced burst release, improved bioavailability, and minimal toxic side effects. Stimulus-responsive CPT nano-prodrugs that respond to various endogenous or exogenous stimuli by introducing various activatable linkers to achieve spatiotemporally responsive drug release at the tumor site. This review comprehensively summarizes the latest research advances in stimulus-responsive CPT nano-prodrugs, including preparation strategies, responsive release mechanisms, and their applications in cancer therapy. Special focus is placed on the release mechanisms and characteristics of various stimulus-responsive CPT nano-prodrugs and their application in cancer treatment. Furthermore, clinical applications of CPT prodrugs are discussed. Finally, challenges and future research directions for CPT nano-prodrugs are discussed. This review to be valuable to readers engaged in prodrug research is expected.
Collapse
Affiliation(s)
- Bingyang Chu
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hanzhi Deng
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Niu
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Qu
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhiyong Qian
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
6
|
Luo R, Le H, Wu Q, Gong C. Nanoplatform-Based In Vivo Gene Delivery Systems for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312153. [PMID: 38441386 DOI: 10.1002/smll.202312153] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/05/2024] [Indexed: 07/26/2024]
Abstract
Gene therapy uses modern molecular biology methods to repair disease-causing genes. As a burgeoning therapeutic, it has been widely applied for cancer therapy. Since 1989, there have been numerous clinical gene therapy cases worldwide. However, a few are successful. The main challenge of clinical gene therapy is the lack of efficient and safe vectors. Although viral vectors show high transfection efficiency, their application is still limited by immune rejection and packaging capacity. Therefore, the development of non-viral vectors is overwhelming. Nanoplatform-based non-viral vectors become a hotspot in gene therapy. The reasons are mainly as follows. 1) Non-viral vectors can be engineered to be uptaken by specific types of cells or tissues, providing effective targeting capability. 2) Non-viral vectors can protect goods that need to be delivered from degradation. 3) Nanoparticles can transport large-sized cargo such as CRISPR/Cas9 plasmids and nucleoprotein complexes. 4) Nanoparticles are highly biosafe, and they are not mutagenic in themselves compared to viral vectors. 5) Nanoparticles are easy to scale preparation, which is conducive to clinical conversion and application. Here, an overview of the categories of nanoplatform-based non-viral gene vectors, the limitations on their development, and their applications in cancer therapy.
Collapse
Affiliation(s)
- Rui Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hao Le
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qinjie Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Changyang Gong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
7
|
Chu B, Chen Z, Wu X, Shi H, Jin X, Song B, Cui M, Zhao Y, Zhao Y, He Y, Wang H, Dong F. Photoactivated Gas-Generating Nanocontrast Agents for Long-Term Ultrasonic Imaging-Guided Combined Therapy of Tumors. ACS NANO 2024; 18:15590-15606. [PMID: 38847586 DOI: 10.1021/acsnano.4c01041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2024]
Abstract
To date, long-term and continuous ultrasonic imaging for guiding the puncture biopsy remains a challenge. In order to address this issue, a multimodality imaging and therapeutic method was developed in the present study to facilitate long-term ultrasonic and fluorescence imaging-guided precision diagnosis and combined therapy of tumors. In this regard, certain types of photoactivated gas-generating nanocontrast agents (PGNAs), capable of exhibiting both ultrasonic and fluorescence imaging ability along with photothermal and sonodynamic function, were designed and fabricated. The advantages of these fabricated PGNAs were then utilized against tumors in vivo, and high therapeutic efficacy was achieved through long-term ultrasonic imaging-guided treatment. In particular, the as-prepared multifunctional PGNAs were applied successfully for the fluorescence-based determination of patient tumor samples collected through puncture biopsy in clinics, and superior performance was observed compared to the clinically used SonoVue contrast agents that are incapable of specifically distinguishing the tumor in ex vivo tissues.
Collapse
Affiliation(s)
- Binbin Chu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
| | - Zhiming Chen
- Department of Ultrasound, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiaofeng Wu
- Department of Ultrasound, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Haoliang Shi
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
| | - Xiangbowen Jin
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
| | - Bin Song
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
| | - Mingyue Cui
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
| | - Yadan Zhao
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
| | - Yingying Zhao
- Department of Ultrasound, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Yao He
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
- Macao Translational Medicine Center, Macau University of Science and Technology, Taipa, 999078 Macau SAR, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, 999078 Macau SAR, China
| | - Houyu Wang
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou 215123 China
| | - Fenglin Dong
- Department of Ultrasound, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| |
Collapse
|
8
|
Qin W, Yang Q, Zhu C, Jiao R, Lin X, Fang C, Guo J, Zhang K. A Distinctive Insight into Inorganic Sonosensitizers: Design Principles and Application Domains. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2311228. [PMID: 38225708 DOI: 10.1002/smll.202311228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 12/29/2023] [Indexed: 01/17/2024]
Abstract
Sonodynamic therapy (SDT) as a promising non-invasive anti-tumor means features the preferable penetration depth, which nevertheless, usually can't work without sonosensitizers. Sonosensitizers produce reactive oxygen species (ROS) in the presence of ultrasound to directly kill tumor cells, and concurrently activate anti-tumor immunity especially after integration with tumor microenvironment (TME)-engineered nanobiotechnologies and combined therapy. Current sonosensitizers are classified into organic and inorganic ones, and current most reviews only cover organic sonosensitizers and highlighted their anti-tumor applications. However, there have few specific reviews that focus on inorganic sonosensitizers including their design principles, microenvironment regulation, etc. In this review, inorganic sonosensitizers are first classified according to their design rationales rather than composition, and the action rationales and underlying chemistry features are highlighted. Afterward, what and how TME is regulated based on the inorganic sonosensitizers-based SDT nanoplatform with an emphasis on the TME targets-engineered nanobiotechnologies are elucidated. Additionally, the combined therapy and their applications in non-cancer diseases are also outlined. Finally, the setbacks and challenges, and proposed the potential solutions and future directions is pointed out. This review provides a comprehensive and detailed horizon on inorganic sonosensitizers, and will arouse more attentions on SDT.
Collapse
Affiliation(s)
- Wen Qin
- State Key Laboratory of Targeting Oncology, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
- Department of Pharmacy and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, Sichuan, 610072, P. R. China
| | - Qiaoling Yang
- State Key Laboratory of Targeting Oncology, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
- Department of Pharmacy and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, Sichuan, 610072, P. R. China
| | - Chunyan Zhu
- Department of Pharmacy and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, Sichuan, 610072, P. R. China
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Yanchangzhong Road, Shanghai, 200072, P. R. China
| | - Rong Jiao
- State Key Laboratory of Targeting Oncology, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
- Department of Pharmacy and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, Sichuan, 610072, P. R. China
| | - Xia Lin
- State Key Laboratory of Targeting Oncology, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
- Department of Pharmacy and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, Sichuan, 610072, P. R. China
| | - Chao Fang
- Department of Pharmacy and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, Sichuan, 610072, P. R. China
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Yanchangzhong Road, Shanghai, 200072, P. R. China
| | - Jiaming Guo
- Department of Radiation Medicine, College of Naval Medicine, Naval Medical University, No. 800 Xiangyin Road, Shanghai, 200433, P. R. China
| | - Kun Zhang
- Department of Pharmacy and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, Sichuan, 610072, P. R. China
| |
Collapse
|
9
|
Wu D, Wang J, Du X, Cao Y, Ping K, Liu D. Cucurbit[8]uril-based supramolecular theranostics. J Nanobiotechnology 2024; 22:235. [PMID: 38725031 PMCID: PMC11084038 DOI: 10.1186/s12951-024-02349-z] [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: 12/23/2023] [Accepted: 02/20/2024] [Indexed: 05/12/2024] Open
Abstract
Different from most of the conventional platforms with dissatisfactory theranostic capabilities, supramolecular nanotheranostic systems have unparalleled advantages via the artful combination of supramolecular chemistry and nanotechnology. Benefiting from the tunable stimuli-responsiveness and compatible hierarchical organization, host-guest interactions have developed into the most popular mainstay for constructing supramolecular nanoplatforms. Characterized by the strong and diverse complexation property, cucurbit[8]uril (CB[8]) shows great potential as important building blocks for supramolecular theranostic systems. In this review, we summarize the recent progress of CB[8]-based supramolecular theranostics regarding the design, manufacture and theranostic mechanism. Meanwhile, the current limitations and corresponding reasonable solutions as well as the potential future development are also discussed.
Collapse
Affiliation(s)
- Dan Wu
- Department of Vascular Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, People's Republic of China
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Jianfeng Wang
- Department of Radiotherapy, China-Japan Union Hospital, Jilin University, Changchun, 130033, People's Republic of China
| | - Xianlong Du
- Bethune First Clinical Medical College, Jilin University, Changchun, 130012, People's Republic of China
| | - Yibin Cao
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Kunmin Ping
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Dahai Liu
- Department of Vascular Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, People's Republic of China.
| |
Collapse
|
10
|
Dash P, Panda PK, Su C, Lin YC, Sakthivel R, Chen SL, Chung RJ. Near-infrared-driven upconversion nanoparticles with photocatalysts through water-splitting towards cancer treatment. J Mater Chem B 2024; 12:3881-3907. [PMID: 38572601 DOI: 10.1039/d3tb01066j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Water splitting is promising, especially for energy and environmental applications; however, there are limited studies on the link between water splitting and cancer treatment. Upconversion nanoparticles (UCNPs) can be used to convert near-infrared (NIR) light to ultraviolet (UV) or visible (Vis) light and have great potential for biomedical applications because of their profound penetration ability, theranostic approaches, low self-fluorescence background, reduced damage to biological tissue, and low toxicity. UCNPs with photocatalytic materials can enhance the photocatalytic activities that generate a shorter wavelength to increase the tissue penetration depth in the biological microenvironment under NIR light irradiation. Moreover, UCNPs with a photosensitizer can absorb NIR light and convert it into UV/vis light and emit upconverted photons, which excite the photoinitiator to create H2, O2, and/or OH˙ via water splitting processes when exposed to NIR irradiation. Therefore, combining UCNPs with intensified photocatalytic and photoinitiator materials may be a promising therapeutic approach for cancer treatment. This review provides a novel strategy for explaining the principles and mechanisms of UCNPs and NIR-driven UCNPs with photocatalytic materials through water splitting to achieve therapeutic outcomes for clinical applications. Moreover, the challenges and future perspectives of UCNP-based photocatalytic materials for water splitting for cancer treatment are discussed in this review.
Collapse
Affiliation(s)
- Pranjyan Dash
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan.
| | - Pradeep Kumar Panda
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Taoyuan City 32003, Taiwan
| | - Chaochin Su
- Institute of Organic and Polymeric Materials, Research and Development Center for Smart Textile Technology, National Taipei University of Technology (Taipei Tech), Taipei 10608, Taiwan
| | - Yu-Chien Lin
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan.
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
- ZhongSun Co., LTD, New Taipei City 220031, Taiwan
| | - Rajalakshmi Sakthivel
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan.
| | - Sung-Lung Chen
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan.
| | - Ren-Jei Chung
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan.
- High-value Biomaterials Research and Commercialization Center, National Taipei University of Technology (Taipei Tech), Taipei 10608, Taiwan
| |
Collapse
|
11
|
Chang K, Sun X, Fu M, Han B, Jiang X, Qi Q, Zhang Y, Ni T, Ge C, Yang Z. H 2O 2-triggered controllable carbon monoxide delivery for photothermally augmented gas therapy. J Mater Chem B 2024; 12:2737-2745. [PMID: 38379390 DOI: 10.1039/d3tb02399k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Carbon monoxide (CO) gas therapy has shown great potential as a very promising approach in the ongoing fight against tumors. However, delivering unstable CO to the tumor site and safely releasing it for maximum efficacy still have unsatisfactory outcomes. In this study, we've developed nanotheranostics (IN-DPPCO NPs) based on conjugated polymer IN-DPP and carbon monoxide (CO) carrier polymer mPEG(CO) for photothermal augmented gas therapy. The IN-DPPCO NPs can release CO with the hydrogen peroxide (H2O2) overexpressed in the tumor microenvironment. Meanwhile, IN-DPPCO NPs exhibit strong absorption in the near-infrared window, showing a high photothermal conversion efficiency of up to 41.5% under 808 nm laser irradiation. In vitro and in vivo experiments demonstrate that these nanotheranostics exhibit good biocompatibility. Furthermore, the synergistic CO/photothermal therapy shows enhanced therapeutic efficacy compared to gas therapy alone. This work highlights the great promise of conjugated polymer nanoparticles as versatile nanocarriers for spatiotemporally controlled and on-demand delivery of gaseous messengers to achieve precision cancer theranostics.
Collapse
Affiliation(s)
- Kaiwen Chang
- Key Laboratory of Medical Molecular Probes, Department of Medical Chemistry, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, P. R. China.
| | - Xiaolin Sun
- Key Laboratory of Medical Molecular Probes, Department of Medical Chemistry, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, P. R. China.
- Department of Scientific Research, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang 453003, P. R. China
| | - Mingying Fu
- Key Laboratory of Medical Molecular Probes, Department of Medical Chemistry, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, P. R. China.
| | - Bing Han
- Key Laboratory of Medical Molecular Probes, Department of Medical Chemistry, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, P. R. China.
| | - Xiaopeng Jiang
- Key Laboratory of Medical Molecular Probes, Department of Medical Chemistry, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, P. R. China.
| | - Qiaofang Qi
- Key Laboratory of Medical Molecular Probes, Department of Medical Chemistry, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, P. R. China.
| | - Yang Zhang
- Key Laboratory of Medical Molecular Probes, Department of Medical Chemistry, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, P. R. China.
| | - Tianjun Ni
- Key Laboratory of Medical Molecular Probes, Department of Medical Chemistry, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, P. R. China.
| | - Chunpo Ge
- Key Laboratory of Medical Molecular Probes, Department of Medical Chemistry, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, P. R. China.
| | - Zhijun Yang
- Key Laboratory of Medical Molecular Probes, Department of Medical Chemistry, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, P. R. China.
| |
Collapse
|
12
|
Li XT, Peng SY, Feng SM, Bao TY, Li SZ, Li SY. Recent Progress in Phage-Based Nanoplatforms for Tumor Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307111. [PMID: 37806755 DOI: 10.1002/smll.202307111] [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: 08/18/2023] [Revised: 09/18/2023] [Indexed: 10/10/2023]
Abstract
Nanodrug delivery systems have demonstrated a great potential for tumor therapy with the development of nanotechnology. Nonetheless, traditional drug delivery systems are faced with issues such as complex synthetic procedures, low reproducibility, nonspecific distribution, impenetrability of biological barrier, systemic toxicity, etc. In recent years, phage-based nanoplatforms have attracted increasing attention in tumor treatment for their regular structure, fantastic carrying property, high transduction efficiency and biosafety. Notably, therapeutic or targeting peptides can be expressed on the surface of the phages through phage display technology, enabling the phage vectors to possess multifunctions. As a result, the drug delivery efficiency on tumor will be vastly improved, thereby enhancing the therapeutic efficacy while reducing the side effects on normal tissues. Moreover, phages can overcome the hindrance of biofilm barrier to elicit antitumor effects, which exhibit great advantages compared with traditional synthetic drug delivery systems. Herein, this review not only summarizes the structure and biology of the phages, but also presents their potential as prominent nanoplatforms against tumor in different pathways to inspire the development of effective nanomedicine.
Collapse
Affiliation(s)
- Xiao-Tong Li
- Department of Anesthesiology, the Second Clinical School of Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Shu-Yi Peng
- Department of Anesthesiology, the Second Clinical School of Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Shao-Mei Feng
- Department of Anesthesiology, the Second Clinical School of Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Ting-Yu Bao
- Department of Clinical Medicine, the Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Sheng-Zhang Li
- Department of Clinical Medicine, the Second Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Shi-Ying Li
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| |
Collapse
|
13
|
Qian G, Mao Y, Zhao H, Zhang L, Xiong L, Long Z. pH-Responsive nanoplatform synergistic gas/photothermal therapy to eliminate biofilms in poly(L-lactic acid) scaffolds. J Mater Chem B 2024; 12:1379-1392. [PMID: 38247429 DOI: 10.1039/d3tb02600k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
To date, implant-associated infection is still a significant clinical challenge, which cannot be effectively eliminated by single therapies due to the formation of microbial biofilms. Herein, a pH-responsive nanoplatform was constructed via the in situ growth of zinc sulfide (ZnS) nanoparticles on the surface of Ti3C2 MXene nanosheets, which was subsequently introduced in poly(L-lactic acid) (PLLA) to prepare a composite bone scaffold via selective laser sintering technology. In the acidic biofilm microenvironment, the degradation of ZnS released hydrogen sulfide (H2S) gas to eliminate the biofilm extracellular DNA (eDNA), thus destroying the compactness of the biofilm. Then, the bacterial biofilm became sensitive to hyperthermia, which could be further destroyed under near-infrared light irradiation due to the excellent photothermal property of MXene, finally achieving gas/photothermal synergistic antibiofilm and efficient sterilization. The results showed that the synergistic gas/photothermal therapy for the composite scaffold not only evidently inhibited the formation of biofilms, but also effectively eradicated the eDNA of the already-formed biofilms and killed 90.4% of E. coli and 84.2% of S. aureus under near infrared light irradiation compared with single gas or photothermal therapy. In addition, the composite scaffold promoted the proliferation and osteogenic differentiation of mouse bone marrow mesenchymal stem cells. Thus, the designed scaffold with excellent biofilm elimination and osteogenesis ability has great potential as an alternative treatment for implant-associated bone infections.
Collapse
Affiliation(s)
- Guowen Qian
- School of Energy and Mechanical Engineering, Jiangxi University of Science and Technology, Nanchang 330013, P. R. China.
- Department of Orthopedics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, P. R. China.
| | - Yuqian Mao
- School of Energy and Mechanical Engineering, Jiangxi University of Science and Technology, Nanchang 330013, P. R. China.
| | - Huihui Zhao
- School of Energy and Mechanical Engineering, Jiangxi University of Science and Technology, Nanchang 330013, P. R. China.
| | - Lemin Zhang
- School of Energy and Mechanical Engineering, Jiangxi University of Science and Technology, Nanchang 330013, P. R. China.
| | - Long Xiong
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, P. R. China
| | - Zhisheng Long
- Department of Orthopedics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, P. R. China.
| |
Collapse
|
14
|
Cheng Q, Shi X, Li Q, Wang L, Wang Z. Current Advances on Nanomaterials Interfering with Lactate Metabolism for Tumor Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305662. [PMID: 37941489 PMCID: PMC10797484 DOI: 10.1002/advs.202305662] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/15/2023] [Indexed: 11/10/2023]
Abstract
Increasing numbers of studies have shown that tumor cells prefer fermentative glycolysis over oxidative phosphorylation to provide a vast amount of energy for fast proliferation even under oxygen-sufficient conditions. This metabolic alteration not only favors tumor cell progression and metastasis but also increases lactate accumulation in solid tumors. In addition to serving as a byproduct of glycolytic tumor cells, lactate also plays a central role in the construction of acidic and immunosuppressive tumor microenvironment, resulting in therapeutic tolerance. Recently, targeted drug delivery and inherent therapeutic properties of nanomaterials have attracted great attention, and research on modulating lactate metabolism based on nanomaterials to enhance antitumor therapy has exploded. In this review, the advanced tumor therapy strategies based on nanomaterials that interfere with lactate metabolism are discussed, including inhibiting lactate anabolism, promoting lactate catabolism, and disrupting the "lactate shuttle". Furthermore, recent advances in combining lactate metabolism modulation with other therapies, including chemotherapy, immunotherapy, photothermal therapy, and reactive oxygen species-related therapies, etc., which have achieved cooperatively enhanced therapeutic outcomes, are summarized. Finally, foreseeable challenges and prospective developments are also reviewed for the future development of this field.
Collapse
Affiliation(s)
- Qian Cheng
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalHuazhong University of Science and TechnologyWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhongUniversity of Science and TechnologyWuhan430022China
| | - Xiao‐Lei Shi
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalHuazhong University of Science and TechnologyWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhongUniversity of Science and TechnologyWuhan430022China
| | - Qi‐Lin Li
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalHuazhong University of Science and TechnologyWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhongUniversity of Science and TechnologyWuhan430022China
| | - Lin Wang
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalHuazhong University of Science and TechnologyWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhongUniversity of Science and TechnologyWuhan430022China
| | - Zheng Wang
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhongUniversity of Science and TechnologyWuhan430022China
| |
Collapse
|
15
|
Xiao W, Zhao L, Sun Y, Yang X, Fu Q. Stimuli-Responsive Nanoradiosensitizers for Enhanced Cancer Radiotherapy. SMALL METHODS 2024; 8:e2301131. [PMID: 37906050 DOI: 10.1002/smtd.202301131] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/10/2023] [Indexed: 11/02/2023]
Abstract
Radiotherapy (RT) has been a classical therapeutic method of cancer for several decades. It attracts tremendous attention for the precise and efficient treatment of local tumors with stimuli-responsive nanomaterials, which enhance RT. However, there are few systematic reviews summarizing the newly emerging stimuli-responsive mechanisms and strategies used for tumor radio-sensitization. Hence, this review provides a comprehensive overview of recently reported studies on stimuli-responsive nanomaterials for radio-sensitization. It includes four different approaches for sensitized RT, namely endogenous response, exogenous response, dual stimuli-response, and multi stimuli-response. Endogenous response involves various stimuli such as pH, hypoxia, GSH, and reactive oxygen species (ROS), and enzymes. On the other hand, exogenous response encompasses X-ray, light, and ultrasound. Dual stimuli-response combines pH/enzyme, pH/ultrasound, and ROS/light. Lastly, multi stimuli-response involves the combination of pH/ROS/GSH and X-ray/ROS/GSH. By elaborating on these responsive mechanisms and applying them to clinical RT diagnosis and treatment, these methods can enhance radiosensitive efficiency and minimize damage to surrounding normal tissues. Finally, this review discusses the additional challenges and perspectives related to stimuli-responsive nanomaterials for tumor radio-sensitization.
Collapse
Affiliation(s)
- Wenjing Xiao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Lin Zhao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yang Sun
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
16
|
Das S, Ghosh B, Sahoo RN, Nayak AK. Recent Advancements in Bioelectronic Medicine: A Review. Curr Drug Deliv 2024; 21:1445-1459. [PMID: 38173212 DOI: 10.2174/0115672018286832231218112557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/17/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024]
Abstract
Bioelectronic medicine is a multidisciplinary field that combines molecular medicine, neurology, engineering, and computer science to design devices for diagnosing and treating diseases. The advancements in bioelectronic medicine can improve the precision and personalization of illness treatment. Bioelectronic medicine can produce, suppress, and measure electrical activity in excitable tissue. Bioelectronic devices modify specific neural circuits using electrons rather than pharmaceuticals and uses of bioelectronic processes to regulate the biological processes underlining various diseases. This promotes the potential to address the underlying causes of illnesses, reduce adverse effects, and lower costs compared to conventional medication. The current review presents different important aspects of bioelectronic medicines with recent advancements. The area of bioelectronic medicine has a lot of potential for treating diseases, enabling non-invasive therapeutic intervention by regulating brain impulses. Bioelectronic medicine uses electricity to control biological processes, treat illnesses, or regain lost capability. These new classes of medicines are designed by the technological developments in the detection and regulation of electrical signaling methods in the nervous system. Peripheral nervous system regulates a wide range of processes in chronic diseases; it involves implanting small devices onto specific peripheral nerves, which read and regulate the brain signaling patterns to achieve therapeutic effects specific to the signal capacity of a particular organ. The potential for bioelectronic medicine field is vast, as it investigates for treatment of various diseases, including rheumatoid arthritis, diabetes, hypertension, paralysis, chronic illnesses, blindness, etc.
Collapse
Affiliation(s)
- Sudipta Das
- Department of Pharmaceutics, Netaji Subhas Chandra Bose Institute of Pharmacy, Chakdaha, Nadia - 741222, West Bengal, India
| | - Baishali Ghosh
- Department of Pharmaceutics, Netaji Subhas Chandra Bose Institute of Pharmacy, Chakdaha, Nadia - 741222, West Bengal, India
| | - Rudra Narayan Sahoo
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, 751003, Odisha, India
| | - Amit Kumar Nayak
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, 751003, Odisha, India
| |
Collapse
|
17
|
Opoku-Damoah Y, Zhang R, Ta HT, Xu ZP. Simultaneous Light-Triggered Release of Nitric Oxide and Carbon Monoxide from a Lipid-Coated Upconversion Nanosystem Inhibits Colon Tumor Growth. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 38038959 DOI: 10.1021/acsami.3c13165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Gas therapy has gained noteworthy attention in biomedical research, with the rise of gas-releasing molecules enhancing their therapeutic potential, especially when integrated into nano-based drug delivery systems. Herein, we present a lipid-coated gas delivery system to simultaneously shuttle two gas-releasing molecules carrying nitric oxide (NO) and carbon monoxide (CO), respectively. Upconversion nanoparticles (UCNPs) are designed to generate photons at 360 nm upon 808 nm of near-infrared (NIR) irradiation. These in situ-generated UV photons trigger simultaneous NO and CO release from S-nitrosoglutathione (GSNO) and the CO-releasing molecule (CORM), respectively, which are coloaded into lipid-coated UCNP/GSNO/CORM/FA nanoparticles (LUGCF). LUGCF with a GSNO/CORM mass ratio of 2:1 is determined to be optimal in terms of synergistically instigating apoptosis in HCT116 and CT26 colon cancer cells, where both NO/CO are released and subsequent production of ROS are detected. This CO/NO combination nanoplatform exhibits a very effective inhibition of colon tumor growth in vivo at relatively low doses upon a mild 808 nm irradiation. Overall, we effectively integrated two therapeutic gas-releasing molecules in one NIR-responsive nanosystem, presenting a promising therapeutic strategy for future biomedical applications in dual-gas cancer therapy.
Collapse
Affiliation(s)
- Yaw Opoku-Damoah
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Run Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Hang T Ta
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
- School of Environment and Science, Griffith University, Brisbane, Queensland 4111, Australia
- Queensland Micro and Nanotechnology Centre, Griffith University, Brisbane, Queensland 4111, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
18
|
Wu S, Yan M, Liang M, Yang W, Chen J, Zhou J. Supramolecular host-guest nanosystems for overcoming cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:805-827. [PMID: 38263983 PMCID: PMC10804391 DOI: 10.20517/cdr.2023.77] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/31/2023] [Accepted: 11/15/2023] [Indexed: 01/25/2024]
Abstract
Cancer drug resistance has become one of the main challenges for the failure of chemotherapy, greatly limiting the selection and use of anticancer drugs and dashing the hopes of cancer patients. The emergence of supramolecular host-guest nanosystems has brought the field of supramolecular chemistry into the nanoworld, providing a potential solution to this challenge. Compared with conventional chemotherapeutic platforms, supramolecular host-guest nanosystems can reverse cancer drug resistance by increasing drug uptake, reducing drug efflux, activating drugs, and inhibiting DNA repair. Herein, we summarize the research progress of supramolecular host-guest nanosystems for overcoming cancer drug resistance and discuss the future research direction in this field. It is hoped that this review will provide more positive references for overcoming cancer drug resistance and promoting the development of supramolecular host-guest nanosystems.
Collapse
Affiliation(s)
- Sha Wu
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, Liaoning, China
| | - Miaomiao Yan
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, Liaoning, China
| | - Minghao Liang
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, Liaoning, China
| | - Wenzhi Yang
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, Liaoning, China
| | - Jingyu Chen
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, Liaoning, China
| | - Jiong Zhou
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, Liaoning, China
- Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou 510632, Guangdong, China
| |
Collapse
|
19
|
He M, Dan Y, Chen M, Dong CM. Biocompatible Polymer-Modified Nanoplatform for Ferroptosis-Enhanced Combination Cancer Therapy. Macromol Biosci 2023; 23:e2300215. [PMID: 37363952 DOI: 10.1002/mabi.202300215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/08/2023] [Indexed: 06/28/2023]
Abstract
Ferroptosis is a novel type of iron-dependent non-apoptotic pathway that regulates cell death and shows unique mechanisms including causing lipid peroxide accumulation, sensitizing drug-resistant cancers, priming immunity by immunogenic cell death, and cooperatively acting with other anticancer modalities for eradicating aggressive malignancies and tumor relapse. Recently, there has been a great deal of effort to design and develop anticancer biocompatible polymeric nanoplatforms including polypeptide and PEGylated ones to achieve effective ferroptosis therapy (FT) and synergistic combination therapies including chemotherapy (CT), photodynamic therapy (PDT), sonodynamic therapy (SDT), photothermal therapy (PTT), gas therapy (GT) including nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2 S), and immunotherapy (IT). To be noted, the combo therapies such as FT-CT, FT-PTT, FT-GT, and FT-IT are attracting much efforts to fight against intractable and metastatic tumors as they can generate synergistic antitumor effects and immunogenic cell death (ICD) effects or modulate immunosuppressive tumor microenvironments to initiate strong antitumor immunity and memory effects. The polymeric Fenton nano-agents with good biosafety and high anticancer efficacy will provide a guarantee for their applications. In this review, various biocompatible polymer-modified nanoplatforms designed for FT and combo treatments are summarized for anticancer therapies and discussed for potential clinical transitions.
Collapse
Affiliation(s)
- Meng He
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Yuxin Dan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Mingsheng Chen
- Shanghai Public Health Clinic Center, Fudan University, Shanghai, 201508, P. R. China
| | - Chang-Ming Dong
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
20
|
Zhang T, Pan Y, Suo M, Lyu M, Lam JWY, Jin Z, Ning S, Tang BZ. Photothermal-Triggered Sulfur Oxide Gas Therapy Augments Type I Photodynamic Therapy for Potentiating Cancer Stem Cell Ablation and Inhibiting Radioresistant Tumor Recurrence. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304042. [PMID: 37559173 PMCID: PMC10582409 DOI: 10.1002/advs.202304042] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Indexed: 08/11/2023]
Abstract
Despite advances in cancer therapy, the existence of self-renewing cancer stem cells (CSC) can lead to tumor recurrence and radiation resistance, resulting in treatment failure and high mortality in patients. To address this issue, a near-infrared (NIR) laser-induced synergistic therapeutic platform has been developed by incorporating aggregation-induced emission (AIE)-active phototheranostic agents and sulfur dioxide (SO2 ) prodrug into a biocompatible hydrogel, namely TBH, to suppress malignant CSC growth. Outstanding hydroxyl radical (·OH) generation and photothermal effect of the AIE phototheranostic agent actualizes Type I photodynamic therapy (PDT) and photothermal therapy through 660 nm NIR laser irradiation. Meanwhile, a large amount of SO2 is released from the SO2 prodrug in thermo-sensitive TBH gel, which depletes upregulated glutathione in CSC and consequentially promotes ·OH generation for PDT enhancement. Thus, the resulting TBH hydrogel can diminish CSC under 660 nm laser irradiation and finally restrain tumor recurrence after radiotherapy (RT). In comparison, the tumor in the mice that were only treated with RT relapsed rapidly. These findings reveal a double-boosting ·OH generation protocol, and the synergistic combination of AIE-mediated PDT and gas therapy provides a novel strategy for inhibiting CSC growth and cancer recurrence after RT, which presents great potential for clinical treatment.
Collapse
Affiliation(s)
- Tianfu Zhang
- School of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510182China
- Department of Chemistrythe Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstructionand Guangdong‐Hong Kong‐Macro Joint Laboratory of Optoelectronic and Magnetic Functional MaterialsThe Hong Kong University of Science and TechnologyClear Water BayKowloonHong Kong999077China
| | - You Pan
- Guangxi Medical University Cancer HospitalNanning530000China
| | - Meng Suo
- School of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510182China
| | - Meng Lyu
- Department of Gastrointestinal Surgery & Department of GeriatricsShenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdong518020China
| | - Jacky Wing Yip Lam
- Department of Chemistrythe Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstructionand Guangdong‐Hong Kong‐Macro Joint Laboratory of Optoelectronic and Magnetic Functional MaterialsThe Hong Kong University of Science and TechnologyClear Water BayKowloonHong Kong999077China
| | - Zhaokui Jin
- School of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510182China
| | - Shipeng Ning
- Guangxi Medical University Cancer HospitalNanning530000China
| | - Ben Zhong Tang
- Department of Chemistrythe Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstructionand Guangdong‐Hong Kong‐Macro Joint Laboratory of Optoelectronic and Magnetic Functional MaterialsThe Hong Kong University of Science and TechnologyClear Water BayKowloonHong Kong999077China
- School of Science and EngineeringShenzhen Institute of Aggregate Science and TechnologyThe Chinese University of Hong KongShenzhenGuangdong518172China
| |
Collapse
|
21
|
Zhang Z, Yan H, Cao W, Xie S, Ran P, Wei K, Li X. Ultrasound-Chargeable Persistent Luminescence Nanoparticles to Generate Self-Propelled Motion and Photothermal/NO Therapy for Synergistic Tumor Treatment. ACS NANO 2023; 17:16089-16106. [PMID: 37515593 DOI: 10.1021/acsnano.3c04906] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/31/2023]
Abstract
Cancer phototherapy indicates advantages in ease of manipulation, negligible drug resistance, and spatiotemporal control but is confronted with challenges in tumor cell accessibility and intermittent light excitation. Herein, we propose a strategy with persistent luminescence (PL)-excited photothermal therapy (PTT), concurrent thermophoresis-propelled motion, and PL-triggered NO release, where PL emission is chargeable by ultrasonication for readily applicable to deep tumors. Mechanoluminescent (ML) nanodots of SrAl2O4:Eu2+ (SAOE) and PL nanodots of ZnGa2O4:Cr3+ (ZGC) were deposited on mesoporous silicates to obtain mSZ nanoparticles (NPs), followed by partially coating with polydopamine (PDA) caps and loading NO donors to prepare Janus mSZ@PDA-NO NPs. The ML emission bands of SAOE nanodots overlap with the excitation band of ZGC, and the persistent near-infrared (NIR) emission could be repeatedly activated by ultrasonication. The PL emission acts as an internal NIR source to produce a thermophoretic force and NO gas propellers to drive the motion of Janus NPs. Compared with the commonly used intermittent NIR illumination at both 660 and 808 nm, the persistent motion of ultrasound-activated NPs enhances cellular uptake and long-lasting PTT and intracellular NO levels to combat tumor cells without the use of any chemotherapeutic drugs. The ultrasound-activated persistent motion promotes intratumoral accumulation and tumor distribution of PTT/NO therapeutics and exhibits significantly higher tumor growth inhibition, longer animal survival, and larger intratumoral NO levels than those who experience external NIR illumination. Thus, this study demonstrates a strategy to activate PL emissions and construct PL-excited nanomotors for phototherapy in deep tissues.
Collapse
Affiliation(s)
- Zhanlin Zhang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Hui Yan
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Wenxiong Cao
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Shuang Xie
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Pan Ran
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Kun Wei
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Xiaohong Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| |
Collapse
|
22
|
Yu YL, Wu JJ, Lin CC, Qin X, Tay FR, Miao L, Tao BL, Jiao Y. Elimination of methicillin-resistant Staphylococcus aureus biofilms on titanium implants via photothermally-triggered nitric oxide and immunotherapy for enhanced osseointegration. Mil Med Res 2023; 10:21. [PMID: 37143145 PMCID: PMC10158155 DOI: 10.1186/s40779-023-00454-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 04/07/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Treatment of methicillin-resistant Staphylococcus aureus (MRSA) biofilm infections in implant placement surgery is limited by the lack of antimicrobial activity of titanium (Ti) implants. There is a need to explore more effective approaches for the treatment of MRSA biofilm infections. METHODS Herein, an interfacial functionalization strategy is proposed by the integration of mesoporous polydopamine nanoparticles (PDA), nitric oxide (NO) release donor sodium nitroprusside (SNP) and osteogenic growth peptide (OGP) onto Ti implants, denoted as Ti-PDA@SNP-OGP. The physical and chemical properties of Ti-PDA@SNP-OGP were assessed by scanning electron microscopy, X-ray photoelectron spectroscope, water contact angle, photothermal property and NO release behavior. The synergistic antibacterial effect and elimination of the MRSA biofilms were evaluated by 2',7'-dichlorofluorescein diacetate probe, 1-N-phenylnaphthylamine assay, adenosine triphosphate intensity, o-nitrophenyl-β-D-galactopyranoside hydrolysis activity, bicinchoninic acid leakage. Fluorescence staining, assays for alkaline phosphatase activity, collagen secretion and extracellular matrix mineralization, quantitative real‑time reverse transcription‑polymerase chain reaction, and enzyme-linked immunosorbent assay (ELISA) were used to evaluate the inflammatory response and osteogenic ability in bone marrow stromal cells (MSCs), RAW264.7 cells and their co-culture system. Giemsa staining, ELISA, micro-CT, hematoxylin and eosin, Masson's trichrome and immunohistochemistry staining were used to evaluate the eradication of MRSA biofilms, inhibition of inflammatory response, and promotion of osseointegration of Ti-PDA@SNP-OGP in vivo. RESULTS Ti-PDA@SNP-OGP displayed a synergistic photothermal and NO-dependent antibacterial effect against MRSA following near-infrared light irradiation, and effectively eliminated the formed MRSA biofilms by inducing reactive oxygen species (ROS)-mediated oxidative stress, destroying bacterial membrane integrity and causing leakage of intracellular components (P < 0.01). In vitro experiments revealed that Ti-PDA@SNP-OGP not only facilitated osteogenic differentiation of MSCs, but also promoted the polarization of pro-inflammatory M1 macrophages to the anti-inflammatory M2-phenotype (P < 0.05 or P < 0.01). The favorable osteo-immune microenvironment further facilitated osteogenesis of MSCs and the anti-inflammation of RAW264.7 cells via multiple paracrine signaling pathways (P < 0.01). In vivo evaluation confirmed the aforementioned results and revealed that Ti-PDA@SNP-OGP induced ameliorative osseointegration in an MRSA-infected femoral defect implantation model (P < 0.01). CONCLUSIONS These findings suggest that Ti-PDA@SNP-OGP is a promising multi-functional material for the high-efficient treatment of MRSA infections in implant replacement surgeries.
Collapse
Affiliation(s)
- Yong-Lin Yu
- Department of Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 Guizhou China
| | - Jun-Jie Wu
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Chuan-Chuan Lin
- Department of Blood Transfusion, Laboratory of Radiation Biology, the Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037 China
| | - Xian Qin
- Department of Reproductive Endocrinology, Chongqing Health Center for Women and Children, Chongqing, 401147 China
| | - Franklin R. Tay
- The Graduate School, Augusta University, Augusta, GA 30912 USA
| | - Li Miao
- Department of Stomatology, the Seventh Medical Center of PLA General Hospital, Beijing, 100700 China
| | - Bai-Long Tao
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 China
| | - Yang Jiao
- Department of Stomatology, the Seventh Medical Center of PLA General Hospital, Beijing, 100700 China
| |
Collapse
|
23
|
Nie Y, Chen W, Kang Y, Yuan X, Li Y, Zhou J, Tao W, Ji X. Two-dimensional porous vermiculite-based nanocatalysts for synergetic catalytic therapy. Biomaterials 2023; 295:122031. [PMID: 36731367 DOI: 10.1016/j.biomaterials.2023.122031] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/09/2023] [Accepted: 01/27/2023] [Indexed: 01/30/2023]
Abstract
This study reports an ultrasound-mediated and two-dimensional (2D) porous vermiculite nanosheets (VMT NSs)-based nanocatalyst platform (Arg@VMT@PDA-PEG) that synergistically harnessed the Fenton reaction-based chemodynamic therapy (CDT), 2D semiconductor-based sonodynamic therapy (SDT) and nitric oxide (NO)-based gas therapy for combination cancer therapy. The tumor microenvironment responsive degradation of polydopamine (PDA) shell could not only prevent L-Arg, a NO donor, leakage during blood circulation, but also selectively release the active sites of VMT NSs for catalytic reactions in tumor cells. Additionally, the Fenton reactions mediated by the abundant Fe2+/Fe3+ in VMT NSs could efficiently produce ·OH and consume glutathione (GSH) for CDT. Moreover, the reactive oxygen species (ROS, ·OH and ·O2-) produced by ultrasound-triggered Arg@VMT@PDA-PEG could not only execute SDT but also oxidize L-Arg to NO for synergetic gas therapy. The results show that the transformation of ROS to NO can enhance curative efficacy owing to the ability of NO with much longer life-time in freely diffusing into cells from intercellular space. This biodegradable Arg@VMT@PDA-PEG nanocatalytic platform integrating three different catalytic reactions provides a new therapeutic paradigm for combination cancer therapy.
Collapse
Affiliation(s)
- Yichu Nie
- Clinical Research Institute, First People's Hospital of Foshan, Foshan, Guangdong, 528000, China
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yong Kang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, 300072, China
| | - Xue Yuan
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, 300072, China
| | - Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jun Zhou
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, 300072, China; Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
24
|
Yang S, Dai W, Zheng W, Wang J. Non-UV-activated persistent luminescence phosphors for sustained bioimaging and phototherapy. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
25
|
Pan M, Hu D, Yuan L, Yu Y, Li Y, Qian Z. Newly developed gas-assisted sonodynamic therapy in cancer treatment. Acta Pharm Sin B 2022. [PMID: 37521874 PMCID: PMC10372842 DOI: 10.1016/j.apsb.2022.12.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Sonodynamic therapy (SDT) is an emerging noninvasive treatment modality that utilizes low-frequency and low-intensity ultrasound (US) to trigger sensitizers to kill tumor cells with reactive oxygen species (ROS). Although SDT has attracted much attention for its properties including high tumor specificity and deep tissue penetration, its anticancer efficacy is still far from satisfactory. As a result, new strategies such as gas-assisted therapy have been proposed to further promote the effectiveness of SDT. In this review, the mechanisms of SDT and gas-assisted SDT are first summarized. Then, the applications of gas-assisted SDT for cancer therapy are introduced and categorized by gas types. Next, therapeutic systems for SDT that can realize real-time imaging are further presented. Finally, the challenges and perspectives of gas-assisted SDT for future clinical applications are discussed.
Collapse
|
26
|
Metal-phenolic networks with ferroptosis to deliver NIR-responsive CO for synergistic therapy. J Control Release 2022; 352:313-327. [PMID: 36272661 DOI: 10.1016/j.jconrel.2022.10.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
As an endogenous gasotransmitter, CO has achieved tremendous advances in cancer treatment through selectively killing cancer cells. However, the application of CO in tumor immunotherapy has not been reported and the tumor targeting delivery is still a tremendous challenge. Herein, thermosensitive boronic acid group-containing CO prodrug was synthesized and fabricated with tannic acid (TA) and iron (Fe) to form metal-phenolic networks, and then loaded with near-infrared (NIR) photothermal agent IR820 to form FeCO-IR820@FeIIITA for combinational therapy of CO and photothermal therapy. Ferroptosis can also be enhanced due to the Fe3+ incorporation. After TA reduced Fe3+ into Fe2+, Fe2+ might lead to intracellular Fenton reaction. Furthermore, in combination with CTLA-4 blockade immunotherapy, FeCO-IR820@FeIIITA remarkably inhibited breast tumor growth, suppressed the lung metastasis and improved the antitumor immune response. To summarize, FeCO-IR820@FeIIITA provides a potential novel option for CO/photothermal/immune synergistic therapy with enhanced ferroptosis through simple compositions and facile synthesis process.
Collapse
|
27
|
Zhang L, Liu Y, Huang H, Xie H, Zhang B, Xia W, Guo B. Multifunctional nanotheranostics for near infrared optical imaging-guided treatment of brain tumors. Adv Drug Deliv Rev 2022; 190:114536. [PMID: 36108792 DOI: 10.1016/j.addr.2022.114536] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/03/2022] [Accepted: 09/07/2022] [Indexed: 02/08/2023]
Abstract
Malignant brain tumors, a heterogeneous group of primary and metastatic neoplasms in the central nervous system (CNS), are notorious for their highly invasive and devastating characteristics, dismal prognosis and low survival rate. Recently, near-infrared (NIR) optical imaging modalities including fluorescence imaging (FLI) and photoacoustic imaging (PAI) have displayed bright prospect in innovation of brain tumor diagnoses, due to their merits, like noninvasiveness, high spatiotemporal resolution, good sensitivity and large penetration depth. Importantly, these imaging techniques have been widely used to vividly guide diverse brain tumor therapies in a real-time manner with high accuracy and efficiency. Herein, we provide a systematic summary of the state-of-the-art NIR contrast agents (CAs) for brain tumors single-modal imaging (e.g., FLI and PAI), dual-modal imaging (e.g., FLI/PAI, FLI/magnetic resonance imaging (MRI) and PAI/MRI) and triple-modal imaging (e.g., MRI/FLI/PAI and MRI/PAI/computed tomography (CT) imaging). In addition, we update the most recent progress on the NIR optical imaging-guided therapies, like single-modal (e.g., photothermal therapy (PTT), chemotherapy, surgery, photodynamic therapy (PDT), gene therapy and gas therapy), dual-modal (e.g., PTT/chemotherapy, PTT/surgery, PTT/PDT, PDT/chemotherapy, PTT/chemodynamic therapy (CDT) and PTT/gene therapy) and triple-modal (e.g., PTT/PDT/chemotherapy, PTT/PDT/surgery, PTT/PDT/gene therapy and PTT/gene/chemotherapy). Finally, we discuss the opportunities and challenges of the CAs and nanotheranostics for future clinic translation.
Collapse
Affiliation(s)
- Li Zhang
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China
| | - Yue Liu
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China
| | - Haiyan Huang
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China
| | - Hui Xie
- Chengdu Institute of Organic Chemistry, Chinese Academy of Sciences, Chengdu, 610041 China
| | - Baozhu Zhang
- Department of Oncology, People's Hospital of Shenzhen Baoan District, The Second Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518101, China
| | - Wujiong Xia
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China.
| |
Collapse
|
28
|
Ji X, Zhong Z. External stimuli-responsive gasotransmitter prodrugs: Chemistry and spatiotemporal release. J Control Release 2022; 351:81-101. [PMID: 36116579 DOI: 10.1016/j.jconrel.2022.09.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/28/2022]
Abstract
Gasotransmitters like nitric oxide, carbon monoxide, and hydrogen sulfide with unique pleiotropic pharmacological effects in mammals are an emerging therapeutic modality for different human diseases including cancer, infection, ischemia-reperfusion injuries, and inflammation; however, their clinical translation is hampered by the lack of a reliable delivery form, which delivers such gasotransmitters to the action site with precisely controlled dosage. The external stimuli-responsive prodrug strategy has shown tremendous potential in developing gasotransmitter prodrugs, which affords precise temporospatial control and better dose control compared with endogenous stimuli-sensitive prodrugs. The promising external stimuli employed for gasotransmitter activation range from photo, ultrasound, and bioorthogonal click chemistry to exogenous enzymes. Herein, we highlight the recent development of external stimuli-mediated decaging chemistry for the temporospatial delivery of gasotransmitters including nitric oxide, carbon monoxide, hydrogen sulfide and sulfur dioxide, and discuss the pros and cons of different designs.
Collapse
Affiliation(s)
- Xingyue Ji
- College of Pharmaceutical Sciences, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| | - Zhiyuan Zhong
- College of Pharmaceutical Sciences, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
29
|
Shukla A, Maiti P. Nanomedicine and versatile therapies for cancer treatment. MedComm (Beijing) 2022; 3:e163. [PMID: 35992969 PMCID: PMC9386439 DOI: 10.1002/mco2.163] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/11/2022] [Accepted: 06/14/2022] [Indexed: 12/19/2022] Open
Abstract
The higher prevalence of cancer is related to high rates of mortality and morbidity worldwide. By virtue of the properties of matter at the nanoscale, nanomedicine is proven to be a powerful tool to develop innovative drug carriers with greater efficacies and fewer side effects than conventional therapies. In this review, different nanocarriers for controlled drug release and their routes of administration have been discussed in detail, especially for cancer treatment. Special emphasis has been given on the design of drug delivery vehicles for sustained release and specific application methods for targeted delivery to the affected areas. Different polymeric vehicles designed for the delivery of chemotherapeutics have been discussed, including graft copolymers, liposomes, hydrogels, dendrimers, micelles, and nanoparticles. Furthermore, the effect of dimensional properties on chemotherapy is vividly described. Another integral section of the review focuses on the modes of administration of nanomedicines and emerging therapies, such as photothermal, photodynamic, immunotherapy, chemodynamic, and gas therapy, for cancer treatment. The properties, therapeutic value, advantages, and limitations of these nanomedicines are highlighted, with a focus on their increased performance versus conventional molecular anticancer therapies.
Collapse
Affiliation(s)
- Aparna Shukla
- School of Materials Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| | - Pralay Maiti
- School of Materials Science and TechnologyIndian Institute of Technology (Banaras Hindu University)VaranasiIndia
| |
Collapse
|
30
|
Tian L, Pei R, Zhang X, Li K, Zhong Y, Luo Y, Zhou SF, Chen L. Tumor Cell-Specific and Lipase-Responsive Delivery of Hydrogen Sulfide for Sensitizing Chemotherapy of Pancreatic Cancer. Front Bioeng Biotechnol 2022; 10:934151. [PMID: 35898641 PMCID: PMC9309817 DOI: 10.3389/fbioe.2022.934151] [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: 05/02/2022] [Accepted: 06/08/2022] [Indexed: 11/23/2022] Open
Abstract
The inability of small molecule drugs to diffuse into tumor interstitium is responsible for the relatively low effectiveness of chemotherapy. Herein, a hydrogen sulfide (H2S) gas–involved chemosensitization strategy is proposed for pancreatic cancer treatment by developing a tumor-specific lipase-responsive nanomedicine based on aptamer-conjugated DATS/Dox co-loaded PCL-b-PEO micelle (DA/D@Ms-A). After receptor-mediated endocytosis and subsequent digestion of PCL blocks by intracellular lipase, the nanomedicine releases Dox and DATS, which then react with intracellular glutathione to produce H2S. The cytotoxicity result indicates that H2S can enhance Dox chemotherapy efficiency owing to the synergetic therapeutic effect of Dox and H2S. Moreover, the nanomedicine is featured with well tumor penetration capability benefitting from the targeting ability of aptamers and high in vivo biocompatibility due to the high density of PEO and biodegradable PCL. The nanomedicine capable of synergetic gas-chemotherapy holds great potential for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Libing Tian
- College of Chemical Engineering, Huaqiao University, Xiamen, China
| | - Rui Pei
- College of Chemical Engineering, Huaqiao University, Xiamen, China
| | - Xiaojun Zhang
- College of Chemical Engineering, Huaqiao University, Xiamen, China
| | - Kun Li
- College of Chemical Engineering, Huaqiao University, Xiamen, China
| | - Yuting Zhong
- College of Chemical Engineering, Huaqiao University, Xiamen, China
| | - Yougen Luo
- Department of Basic Medical Science, Jiangsu Vocational College of Medicine, Yancheng, China
- *Correspondence: Lichan Chen, ; Shu-Feng Zhou, ; Yougen Luo,
| | - Shu-Feng Zhou
- College of Chemical Engineering, Huaqiao University, Xiamen, China
- *Correspondence: Lichan Chen, ; Shu-Feng Zhou, ; Yougen Luo,
| | - Lichan Chen
- College of Chemical Engineering, Huaqiao University, Xiamen, China
- *Correspondence: Lichan Chen, ; Shu-Feng Zhou, ; Yougen Luo,
| |
Collapse
|