1
|
Zhang X, He N, Zhang L, Dai T, Sun Z, Shi Y, Li S, Yu N. Application of high intensity focused ultrasound combined with nanomaterials in anti-tumor therapy. Drug Deliv 2024; 31:2342844. [PMID: 38659328 PMCID: PMC11047217 DOI: 10.1080/10717544.2024.2342844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
High intensity focused ultrasound (HIFU) has demonstrated its safety, efficacy and noninvasiveness in the ablation of solid tumor. However, its further application is limited by its inherent deficiencies, such as postoperative recurrence caused by incomplete ablation and excessive intensity affecting surrounding healthy tissues. Recent research has indicated that the integration of nanomaterials with HIFU exhibits a promising synergistic effect in tumor ablation. The concurrent utilization of nanomaterials with HIFU can help overcome the limitations of HIFU by improving targeting and ablation efficiency, expanding operation area, increasing operation accuracy, enhancing stability and bio-safety during the process. It also provides a platform for multi-therapy and multi-mode imaging guidance. The present review comprehensively expounds upon the synergistic mechanism between nanomaterials and HIFU, summarizes the research progress of nanomaterials as cavitation nuclei and drug carriers in combination with HIFU for tumor ablation. Furthermore, this review highlights the potential for further exploration in the development of novel nanomaterials that enhance the synergistic effect with HIFU on tumor ablation.
Collapse
Affiliation(s)
- Xuehui Zhang
- Department of Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Liang Zhang
- Department of Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tong Dai
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Zihan Sun
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Yuqing Shi
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Ning Yu
- Department of Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Jiang F, Liu S, Wang L, Chen H, Huang Y, Cao Y, Wang X, Lin M, Zhang J. ROS-Responsive Nanoprobes for Bimodal Imaging-Guided Cancer Targeted Combinatorial Therapy. Int J Nanomedicine 2024; 19:8071-8090. [PMID: 39130685 PMCID: PMC11317049 DOI: 10.2147/ijn.s467512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/17/2024] [Indexed: 08/13/2024] Open
Abstract
Purpose Chemotherapy mediated by Reactive oxygen species (ROS)-responsive drug delivery systems can potentially mitigate the toxic side effects of chemotherapeutic drugs and significantly enhance their therapeutic efficacy. However, achieving precise targeted drug delivery and real-time control of ROS-responsive drug release at tumor sites remains a formidable challenge. Therefore, this study aimed to describe a ROS-responsive drug delivery system with specific tumor targeting capabilities for mitigating chemotherapy-induced toxicity while enhancing therapeutic efficacy under guidance of Fluorescence (FL) and Magnetic resonance (MR) bimodal imaging. Methods Indocyanine green (ICG), Doxorubicin (DOX) prodrug pB-DOX and Superparamagnetic iron oxide (SPIO, Fe3O4) were encapsulated in poly(lactic-co-glycolic acid) (PLGA) by double emulsification method to prepare ICG/ pB-DOX/ Fe3O4/ PLGA nanoparticles (IBFP NPs). The surface of IBFP NPs was functionalized with mammaglobin antibodies (mAbs) by carbodiimide method to construct the breast cancer-targeting mAbs/ IBFP NPs (MIBFP NPs). Thereafter, FL and MR bimodal imaging ability of MIBFP NPs was evaluated in vitro and in vivo. Finally, the combined photodynamic therapy (PDT) and chemotherapy efficacy evaluation based on MIBFP NPs was studied. Results The multifunctional MIBFP NPs exhibited significant targeting efficacy for breast cancer. FL and MR bimodal imaging clearly displayed the distribution of the targeting MIBFP NPs in vivo. Upon near-infrared laser irradiation, the MIBFP NPs loaded with ICG effectively generated ROS for PDT, enabling precise tumor ablation. Simultaneously, it triggered activation of the pB-DOX by cleaving its sensitive moiety, thereby restoring DOX activity and achieving ROS-responsive targeted chemotherapy. Furthermore, the MIBFP NPs combined PDT and chemotherapy to enhance the efficiency of tumor ablation under guidance of bimodal imaging. Conclusion MIBFP NPs constitute a novel dual-modality imaging-guided drug delivery system for targeted breast cancer therapy and offer precise and controlled combined treatment options.
Collapse
Affiliation(s)
- Fujie Jiang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing, 400030, People’s Republic of China
| | - Shuling Liu
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing, 400030, People’s Republic of China
| | - Lu Wang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing, 400030, People’s Republic of China
| | - Huifang Chen
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing, 400030, People’s Republic of China
| | - Yao Huang
- School of Medicine, Chongqing University, Chongqing, 400030, People’s Republic of China
| | - Ying Cao
- School of Medicine, Chongqing University, Chongqing, 400030, People’s Republic of China
| | - Xiaoxia Wang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing, 400030, People’s Republic of China
| | - Meng Lin
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing, 400030, People’s Republic of China
| | - Jiuquan Zhang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing, 400030, People’s Republic of China
| |
Collapse
|
3
|
Zhang H, Zhao R, Zhang F, Xia J, Wang Z. Enhancing electrochemiluminescence for chloramphenicol detection based on the synergistic effect of doped Ti 3C 2 with ultrasound. Food Chem 2024; 448:139003. [PMID: 38547710 DOI: 10.1016/j.foodchem.2024.139003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/28/2024] [Accepted: 03/09/2024] [Indexed: 04/24/2024]
Abstract
Chloramphenicol (CAP) is known to be harmful to the environment and food, posing a threat to human health. Developing an effective and convenient method for detecting CAP is crucial. An electrochemiluminescence (ECL) biosensor has been designed for sensitive detection of CAP. The improved ECL behavior was attributed to the synergistic effect of N and P co-doped Ti3C2-Apt1 (N, P-Ti3C2-Apt1) nanoprobes and high intensity focused ultrasound (HIFU) pretreatment. The doping of N and P could improve the electrochemical performance of Ti3C2. HIFU pretreatment generated more reactive oxygen species (ROS) in the luminol-O2 system. N, P-Ti3C2 could aggregate and catalyze ROS, causing an increase in ECL intensity. Furthermore, N, P-Ti3C2 as a carrier loaded more aptamer, which could recognize CAP with high specificity. The detection limit was 0.01 ng/mL. This biosensor has been successfully applied in milk and environmental water samples, highlighting its potential in the field of food and environmental analysis.
Collapse
Affiliation(s)
- Huixin Zhang
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao Application Technology Innovation Center of Photoelectric Biosensing for Clinical Diagnosis and Treatment, Instrumental Analysis Center of Qingdao University, Qingdao University, Qingdao 266071, China; School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| | - Rui Zhao
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao Application Technology Innovation Center of Photoelectric Biosensing for Clinical Diagnosis and Treatment, Instrumental Analysis Center of Qingdao University, Qingdao University, Qingdao 266071, China
| | - Feifei Zhang
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao Application Technology Innovation Center of Photoelectric Biosensing for Clinical Diagnosis and Treatment, Instrumental Analysis Center of Qingdao University, Qingdao University, Qingdao 266071, China.
| | - Jianfei Xia
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao Application Technology Innovation Center of Photoelectric Biosensing for Clinical Diagnosis and Treatment, Instrumental Analysis Center of Qingdao University, Qingdao University, Qingdao 266071, China
| | - Zonghua Wang
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao Application Technology Innovation Center of Photoelectric Biosensing for Clinical Diagnosis and Treatment, Instrumental Analysis Center of Qingdao University, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
4
|
Xu H, Kim D, Zhao YY, Kim C, Song G, Hu Q, Kang H, Yoon J. Remote Control of Energy Transformation-Based Cancer Imaging and Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402806. [PMID: 38552256 DOI: 10.1002/adma.202402806] [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: 02/23/2024] [Revised: 03/24/2024] [Indexed: 04/06/2024]
Abstract
Cancer treatment requires precise tumor-specific targeting at specific sites that allows for high-resolution diagnostic imaging and long-term patient-tailorable cancer therapy; while, minimizing side effects largely arising from non-targetability. This can be realized by harnessing exogenous remote stimuli, such as tissue-penetrative ultrasound, magnetic field, light, and radiation, that enable local activation for cancer imaging and therapy in deep tumors. A myriad of nanomedicines can be efficiently activated when the energy of such remote stimuli can be transformed into another type of energy. This review discusses the remote control of energy transformation for targetable, efficient, and long-term cancer imaging and therapy. Such ultrasonic, magnetic, photonic, radiative, and radioactive energy can be transformed into mechanical, thermal, chemical, and radiative energy to enable a variety of cancer imaging and treatment modalities. The current review article describes multimodal energy transformation where a serial cascade or multiple types of energy transformation occur. This review includes not only mechanical, chemical, hyperthermia, and radiation therapy but also emerging thermoelectric, pyroelectric, and piezoelectric therapies for cancer treatment. It also illustrates ultrasound, magnetic resonance, fluorescence, computed tomography, photoluminescence, and photoacoustic imaging-guided cancer therapies. It highlights afterglow imaging that can eliminate autofluorescence for sustained signal emission after the excitation.
Collapse
Affiliation(s)
- Hai Xu
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Dahee Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Yuan-Yuan Zhao
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Chowon Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Qiongzheng Hu
- Qilu University of Technology (Shandong Academy of Sciences), Shandong Analysis and Test Center, Jinan, 250014, China
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| |
Collapse
|
5
|
Sun T, Li J, Zhou Y, Zeng C, Luo C, Luo X, Li H. Metal-Organic Framework-Mediated Synergistic Hypoxia-Activated Chemo-Immunotherapy Induced by High Intensity Focused Ultrasound for Enhanced Cancer Theranostics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306338. [PMID: 38072817 DOI: 10.1002/smll.202306338] [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: 07/26/2023] [Revised: 10/04/2023] [Indexed: 05/03/2024]
Abstract
High intensity focused ultrasound (HIFU) has attracted considerable attention as a noninvasive, efficient, and economic therapeutic modality for solid tumors. However, HIFU surgery has its intrinsic limitation in completely ablating tumors, leading to residual tumor tissue. Furthermore, the severely hypoxic environment ensuring after surgery can exacerbate the unrestricted proliferation and metabolism of residual tumor cells, leading to tumor recurrence and metastasis. To address these limitations, a versatile HIFU-specific metal-organic framework nanosystem (called ADMOFs) is developed by coordinating hypoxia-activated prodrug AQ4N, Mn2+, and DOX based on the postoperative response to changes in the tumor microenvironment. ADMOFs loaded with AQ4N/Mn2+ exhibited remarkable tumor-targeting behavior in vivo and enhanced photoacoustic/magnetic resonance imaging effects, enabling more accurate guidance for HIFU surgery. After surgery, the ADMOFs exploited the severely hypoxic tumor environment induced by HIFU, overcoming hypoxia-associated drug resistance, and inducing immunogenic cell death. Finally, it effectively inhibited tumor growth and eliminated lung metastasis. Transcriptome studies revealed that this strategy significantly up-regulated genes involved in apoptosis, cell cycle, and HIF-1 signaling pathway while downregulating genes related to tumor proliferation and metastasis. These findings suggest that combining hypoxia-activated chemo-immunotherapy with HIFU is a promising strategy for enhancing cancer theranostics.
Collapse
Affiliation(s)
- Tingyu Sun
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Jingnan Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Yinglin Zhou
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Chao Zeng
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Chengyan Luo
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Xirui Luo
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Huanan Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, P. R. China
| |
Collapse
|
6
|
Qin W, Qiao L, Wang Q, Gao M, Zhou M, Sun Q, Zhang H, Yang T, Shan G, Yao W, Yi X, He X. Advancing Precision: A Controllable Self-Synergistic Nanoplatform Initiating Pyroptosis-Based Immunogenic Cell Death Cascade for Targeted Tumor Therapy. ACS NANO 2024; 18:1582-1598. [PMID: 38170456 DOI: 10.1021/acsnano.3c09499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Heterogeneity of the tumor microenvironment (TME) is primarily responsible for ineffective tumor treatment and uncontrolled tumor progression. Pyroptosis-based immunogenic cell death (ICD) therapy is an ideal strategy to overcome TME heterogeneity and obtain a satisfactory antitumor effect. However, the efficiency of current pyroptosis therapeutics, which mainly depends on a single endogenous or exogenous stimulus, is limited by the intrinsic pathological features of malignant cells. Thus, it is necessary to develop a synergistic strategy with a high tumor specificity and modulability. Herein, a synergistic nanoplatform is constructed by combining a neutrophil camouflaging shell and a self-synergistic reactive oxygen species (ROS) supplier-loaded polymer. The covered neutrophil membranes endow the nanoplatform with stealthy properties and facilitate sufficient tumor accumulation. Under laser irradiation, the photosensitizer (indocyanine green) exogenously triggers ROS generation and converts the laser irradiation into heat to upregulate NAD(P)H:quinone oxidoreductase 1, which further catalyzes β-Lapachone to self-produce sufficient endogenous ROS, resulting in amplified ICD outcomes. The results confirm that the continuously amplified ROS production not only eliminates the primary tumor but also concurrently enhances gasdermin E-mediated pyroptosis, initiates an ICD cascade, re-educates the heterogeneous TME, and promotes a systemic immune response to suppress distant tumors. Overall, this self-synergistic nanoplatform provides an efficient and durable method for redesigning the immune system for targeted tumor inhibition.
Collapse
Affiliation(s)
- Weiji Qin
- School of Life Sciences, Anhui Medical University, Hefei 230011, P. R. China
| | - Lei Qiao
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Qian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Min Gao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Man Zhou
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, P. R. China
| | - Qiuting Sun
- School of Life Sciences, Anhui Medical University, Hefei 230011, P. R. China
| | - Huiru Zhang
- School of Life Sciences, Anhui Medical University, Hefei 230011, P. R. China
| | - Tianhao Yang
- School of Life Sciences, Anhui Medical University, Hefei 230011, P. R. China
| | - Guisong Shan
- School of Life Sciences, Anhui Medical University, Hefei 230011, P. R. China
| | - Wanqing Yao
- School of Life Sciences, Anhui Medical University, Hefei 230011, P. R. China
| | - Xiaoqing Yi
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, P. R. China
| | - Xiaoyan He
- School of Life Sciences, Anhui Medical University, Hefei 230011, P. R. China
| |
Collapse
|
7
|
Sun W, Xiao H, Zhu J, Hao Z, Sun J, Wang D, Wang X, Ramalingam M, Xie S, Wang R. Multifunctional Oxygen-Generating Nanoflowers for Enhanced Tumor Therapy. ACS APPLIED BIO MATERIALS 2023; 6:4998-5008. [PMID: 37880964 DOI: 10.1021/acsabm.3c00678] [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: 10/27/2023]
Abstract
Sonodynamic therapy (SDT) and chemotherapy have received great attention as effective methods for tumor treatment. However, the inherent hypoxia of the tumor greatly hinders its therapeutic efficacy. In this work, a tumor microenvironment-responsive biodegradable nanoplatform SiO2-MnO2-PEG-Ce6&DOX (designated as SMPC&D) is fabricated by encapsulating manganese oxide (MnO2) into silica nanoparticles and anchoring poly(ethylene glycol) (PEG) onto the surface for tumor hypoxia relief and delivery, then loaded with sonosensitizer Chlorin e6 (Ce6) and chemotherapeutic drug doxorubicin (DOX) for hypoxic tumor treatment. We evaluated the physicochemical properties of SMPC&D nanoparticles and the tumor therapeutic effects of chemotherapy and SDT under ultrasound stimulation in vitro and in vivo. After endocytosis by tumor cells, highly expressed glutathione (GSH) triggers biodegradation of the nanoplatform and MnO2 catalyzes hydrogen peroxide (H2O2) to generate oxygen (O2), thereby alleviating tumor hypoxia. Depleting GSH and self-supplying O2 effectively improve the SDT efficiency both in vitro and in vivo. Ultrasonic stimulation promoted the release and cellular uptake of chemotherapy drugs. In addition, the relieved hypoxia reduced the efflux of chemotherapy drugs by downregulating the expression of the P-gp protein, which jointly improved the effect of chemotherapy. This study demonstrates that the degradable SMPC&D as a therapeutic agent can achieve efficient chemotherapy and SDT synergistic therapy for hypoxic tumors.
Collapse
Affiliation(s)
- Wanru Sun
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
- Science Fund of Shandong Laboratory of Advanced Materials and Green Manufacturing at Yantai, Yantai 264000, People's Republic of China
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Huifang Xiao
- Zhongnan Hospital of Wuhan University, Wuhan 430062, People's Republic of China
| | - Jiazhi Zhu
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Zhaokun Hao
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Jian Sun
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Deqiang Wang
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai 264100, People's Republic of China
| | - Xin Wang
- Department of Rehabilitation Medicine, Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Murugan Ramalingam
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country(UPV/EHU), 01006 Vitoria-Gasteiz, Spain
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, 28029 Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
- Joint Research Laboratory (JRL), Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
- Drug Formulation Unit 10, Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
- Bioprinting and Precisión Medicine, Centro de investigación Lascaray Ikergunea, Avenida Miguel de Unamuno, 01006 Vitoria-Gasteiz, Spain
- School of Basic Medical Science, Chengdu University, Chengdu 610106, China
- Department of Metallurgical and Materials Engineering, Atilim University, Ankara 06830, Turkey
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, 78054 Villingen-Schwennigen, Germany
| | - Shuyang Xie
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Ranran Wang
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
- Science Fund of Shandong Laboratory of Advanced Materials and Green Manufacturing at Yantai, Yantai 264000, People's Republic of China
| |
Collapse
|
8
|
Siddiqui L, Hasan N, Mishra PK, Gupta N, Singh AT, Madaan A, Jaggi M, Saad S, Ekielski A, Iqbal Z, Kesharwani P, Talegaonkar S. CD44 mediated colon cancer targeting mutlifaceted lignin nanoparticles: Synthesis, in vitro characterization and in vivo efficacy studies. Int J Pharm 2023; 643:123270. [PMID: 37499773 DOI: 10.1016/j.ijpharm.2023.123270] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 07/10/2023] [Accepted: 07/23/2023] [Indexed: 07/29/2023]
Abstract
Hyaluronic acid (HA) coated irinotecan loaded lignin nanoparticles (HDLNPs) were synthesized using ionic interaction method. Optimized nanoparticles were characterized for their active chemotherapeutic targeting potential to CD44 receptors overly-expressed on cancer cells. Blood component interaction studies supported hemocompatible nature of HDLNPs and also demonstrated their sustained plasma residence property. Cell anti-proliferation and mitochondrial depolarization studies on HT-29 cells suggest significantly (p < 0.01) improved chemotherapeutic efficacy of HDLNPs. In vitro cell based studies showed that nanoparticles have retained antioxidant activity of lignin that can prevent cancer relapse. In vivo biodistribution studies in tumor-bearing Balb/c mice confirmed improved drug localization in tumor site for longer duration. Tumor regression and histopathological studies indicated the efficacy ofligand-assisted targeting chemotherapy over the conventional therapy. Hematological and biochemical estimation suggested that irinotecan-associated myelosuppression, liver steatosis and rare kidney failure can be avoided by its encapsulation in HA-coated lignin nanoparticles. HDLNPs were found to be stable over a period of 12 months.
Collapse
Affiliation(s)
- Lubna Siddiqui
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Pawan K Mishra
- Faculty of Business and Economics, Mendel University in Brno, Brno, Czech Republic.
| | - Neha Gupta
- Cell Biology Lab, Dabur Research Foundation, Ghaziabad, UP, India
| | - Anu T Singh
- Cell Biology Lab, Dabur Research Foundation, Ghaziabad, UP, India
| | - Alka Madaan
- Cell Biology Lab, Dabur Research Foundation, Ghaziabad, UP, India
| | - Manu Jaggi
- Cell Biology Lab, Dabur Research Foundation, Ghaziabad, UP, India
| | - Suma Saad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Adam Ekielski
- Department of Production Engineering, Warsaw University of Life Sciences, Poland
| | - Zeenat Iqbal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Sushama Talegaonkar
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Govt. of NCT of Delhi, New Delhi, India.
| |
Collapse
|
9
|
Li Y, Feng M, Guo T, Wang Z, Zhao Y. Tailored Beta-Lapachone Nanomedicines for Cancer-Specific Therapy. Adv Healthc Mater 2023; 12:e2300349. [PMID: 36970948 DOI: 10.1002/adhm.202300349] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/16/2023] [Indexed: 03/29/2023]
Abstract
Nanotechnology shows the power to improve efficacy and reduce the adverse effects of anticancer agents. As a quinone-containing compound, beta-lapachone (LAP) is widely employed for targeted anticancer therapy under hypoxia. The principal mechanism of LAP-mediated cytotoxicity is believed due to the continuous generation of reactive oxygen species with the aid of NAD(P)H: quinone oxidoreductase 1 (NQO1). The cancer selectivity of LAP relies on the difference between NQO1 expression in tumors and that in healthy organs. Despite this, the clinical translation of LAP faces the problem of narrow therapeutic window that is challenging for dose regimen design. Herein, the multifaceted anticancer mechanism of LAP is briefly introduced, the advance of nanocarriers for LAP delivery is reviewed, and the combinational delivery approaches to enhance LAP potency in recent years are summarized. The mechanisms by which nanosystems boost LAP efficacy, including tumor targeting, cellular uptake enhancement, controlled cargo release, enhanced Fenton or Fenton-like reaction, and multidrug synergism, are also presented. The problems of LAP anticancer nanomedicines and the prospective solutions are discussed. The current review may help to unlock the potential of cancer-specific LAP therapy and speed up its clinical translation.
Collapse
Affiliation(s)
- Yaru Li
- School of Pharmaceutical Science and Technology, Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin, 300072, China
| | - Meiyu Feng
- School of Pharmaceutical Science and Technology, Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin, 300072, China
| | - Tao Guo
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, 300120, China
| | - Zheng Wang
- School of Pharmaceutical Science and Technology, Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin, 300072, China
| | - Yanjun Zhao
- School of Pharmaceutical Science and Technology, Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin, 300072, China
| |
Collapse
|
10
|
Wang Z, Jin A, Yang Z, Huang W. Advanced Nitric Oxide Generating Nanomedicine for Therapeutic Applications. ACS NANO 2023; 17:8935-8965. [PMID: 37126728 PMCID: PMC10395262 DOI: 10.1021/acsnano.3c02303] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Nitric oxide (NO), a gaseous transmitter extensively present in the human body, regulates vascular relaxation, immune response, inflammation, neurotransmission, and other crucial functions. Nitrite donors have been used clinically to treat angina, heart failure, pulmonary hypertension, and erectile dysfunction. Based on NO's vast biological functions, it further can treat tumors, bacteria/biofilms and other infections, wound healing, eye diseases, and osteoporosis. However, delivering NO is challenging due to uncontrolled blood circulation release and a half-life of under five seconds. With advanced biotechnology and the development of nanomedicine, NO donors packaged with multifunctional nanocarriers by physically embedding or chemically conjugating have been reported to show improved therapeutic efficacy and reduced side effects. Herein, we review and discuss recent applications of NO nanomedicines, their therapeutic mechanisms, and the challenges of NO nanomedicines for future scientific studies and clinical applications. As NO enables the inhibition of the replication of DNA and RNA in infectious microbes, including COVID-19 coronaviruses and malaria parasites, we highlight the potential of NO nanomedicines for antipandemic efforts. This review aims to provide deep insights and practical hints into design strategies and applications of NO nanomedicines.
Collapse
Affiliation(s)
- Zhixiong Wang
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Albert Jin
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Zhen Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian 350117, China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, Fujian 350117, China
| | - Wei Huang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian 350117, China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, Fujian 350117, China
| |
Collapse
|
11
|
Gong J, Shi T, Liu J, Pei Z, Liu J, Ren X, Li F, Qiu F. Dual-drug codelivery nanosystems: An emerging approach for overcoming cancer multidrug resistance. Biomed Pharmacother 2023; 161:114505. [PMID: 36921532 DOI: 10.1016/j.biopha.2023.114505] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Multidrug resistance (MDR) promotes tumor recurrence and metastasis and heavily reduces anticancer efficiency, which has become a primary reason for the failure of clinical chemotherapy. The mechanisms of MDR are so complex that conventional chemotherapy usually fails to achieve an ideal therapeutic effect and even accelerates the occurrence of MDR. In contrast, the combination of chemotherapy with dual-drug has significant advantages in tumor therapy. A novel dual-drug codelivery nanosystem, which combines dual-drug administration with nanotechnology, can overcome the application limitation of free drugs. Both the characteristics of nanoparticles and the synergistic effect of dual drugs contribute to circumventing various drug-resistant mechanisms in tumor cells. Therefore, developing dual-drug codelivery nanosystems with different multidrug-resistant mechanisms has an important reference value for reversing MDR and enhancing the clinical antitumor effect. In this review, the advantages, principles, and common codelivery nanocarriers in the application of dual-drug codelivery systems are summarized. The molecular mechanisms of MDR and the dual-drug codelivery nanosystems designed based on different mechanisms are mainly introduced. Meanwhile, the development prospects and challenges of codelivery nanosystems are also discussed, which provide guidelines to exploit optimized combined chemotherapy strategies in the future.
Collapse
Affiliation(s)
- Jianing Gong
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Taoran Shi
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jinfeng Liu
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zerong Pei
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingbo Liu
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, China
| | - Xiaoliang Ren
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Fengyun Li
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
12
|
Petrikaite V, D'Avanzo N, Celia C, Fresta M. Nanocarriers overcoming biological barriers induced by multidrug resistance of chemotherapeutics in 2D and 3D cancer models. Drug Resist Updat 2023; 68:100956. [PMID: 36958083 DOI: 10.1016/j.drup.2023.100956] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023]
Abstract
Multidrug resistance (MDR) is currently a big challenge in cancer therapy and limits its success in several patients. Tumors use the MDR mechanisms to colonize the host and reduce the efficacy of chemotherapeutics that are injected as single agents or combinations. MDR mechanisms are responsible for inactivation of drugs and formbiological barriers in cancer like the drug efflux pumps, aberrant extracellular matrix, hypoxic areas, altered cell death mechanisms, etc. Nanocarriers have some potential to overcome these barriers and improve the efficacy of chemotherapeutics. In fact, they are versatile and can deliver natural and synthetic biomolecules, as well as RNAi/DNAi, thus providing a controlled release of drugs and a synergistic effect in tumor tissues. Biocompatible and safe multifunctional biopolymers, with or without specific targeting molecules, modify the surface and interface properties of nanocarriers. These modifications affect the interaction of nanocarriers with cellular models as well as the selection of suitable models for in vitro experiments. MDR cancer cells, and particularly their 2D and 3D models, in combination with anatomical and physiological structures of tumor tissues, can boost the design and preparation of nanomedicines for anticancer therapy. 2D and 3D cancer cell cultures are suitable models to study the interaction, internalization, and efficacy of nanocarriers, the mechanisms of MDR in cancer cells and tissues, and they are used to tailor a personalized medicine and improve the efficacy of anticancer treatment in patients. The description of molecular mechanisms and physio-pathological pathways of these models further allow the design of nanomedicine that can efficiently overcome biological barriers involved in MDR and test the activity of nanocarriers in 2D and 3D models of MDR cancer cells.
Collapse
Affiliation(s)
- Vilma Petrikaite
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Sukilėlių pr. 13, LT-50162 Kaunas, Lithuania; Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania.
| | - Nicola D'Avanzo
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, 66100 Chieti, Italy; Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro Campus Universitario-Germaneto, Viale Europa, 88100 Catanzaro, Italy
| | - Christian Celia
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Sukilėlių pr. 13, LT-50162 Kaunas, Lithuania; Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, 66100 Chieti, Italy
| | - Massimo Fresta
- Department of Health Sciences, University of Catanzaro "Magna Graecia", Viale "S. Venuta" s.n.c., 88100 Catanzaro, Italy
| |
Collapse
|
13
|
Zhang H, Zhou X, Zhang F, Xia J, Wang Z. Ultrasound-pretreatment combined with Ti 3C 2-TiO 2-AuNPs enhancing the electrogenerated chemiluminescence of the air-saturated luminol for exosomes detection. ULTRASONICS SONOCHEMISTRY 2023; 94:106330. [PMID: 36805412 PMCID: PMC9969320 DOI: 10.1016/j.ultsonch.2023.106330] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/30/2023] [Accepted: 02/11/2023] [Indexed: 06/18/2023]
Abstract
It is still a great challenge to develop effective strategies to improve the low electrogenerated chemiluminescence (ECL) of air-saturated luminol. Herein, the synergistic effects of Ti3C2-TiO2-AuNPs nano hybrid and high-intensity focused ultrasound pretreatment (ultrasound-pretreatment) were used to significantly improve the ECL emission of the air-saturated luminol, and the mechanism was proposed. The ultrasound-pretreatment as a green method with the cavitation effect could form O2-• and H2O2 in situ as an initiator. TiO2 and Au nanoparticles (AuNPs) were in situ decorated on the Ti3C2 surface to form Ti3C2-TiO2-AuNPs, and it was proved as a highly efficient booster which could catalyze and aggregate H2O2 to the O2-•. The utilization rate of intermediates has been greatly improved. Exosomes as model targets can be sensitively detected by the ECL sensor. The detection limit was 195 particles μL-1. The detection results of exosomes in actual samples are satisfactory. We believe that the ultrasound-pretreatment strategy could be extended to the sensitive detection in the biological sample.
Collapse
Affiliation(s)
- Huixin Zhang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Shandong Sino-Japanese Centre for Collaborative Research of Carbon Nanomaterials, Instrumental Analysis Centre of Qingdao University, Qingdao University, Qingdao, Shandong 266071, China
| | - Xin Zhou
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Shandong Sino-Japanese Centre for Collaborative Research of Carbon Nanomaterials, Instrumental Analysis Centre of Qingdao University, Qingdao University, Qingdao, Shandong 266071, China
| | - Feifei Zhang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Shandong Sino-Japanese Centre for Collaborative Research of Carbon Nanomaterials, Instrumental Analysis Centre of Qingdao University, Qingdao University, Qingdao, Shandong 266071, China
| | - Jianfei Xia
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Shandong Sino-Japanese Centre for Collaborative Research of Carbon Nanomaterials, Instrumental Analysis Centre of Qingdao University, Qingdao University, Qingdao, Shandong 266071, China
| | - Zonghua Wang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Shandong Sino-Japanese Centre for Collaborative Research of Carbon Nanomaterials, Instrumental Analysis Centre of Qingdao University, Qingdao University, Qingdao, Shandong 266071, China.
| |
Collapse
|
14
|
Tang R, He H, Lin X, Wu N, Wan L, Chen Q, Hu Y, Cheng C, Cao Y, Guo X, Zhou Y, Xiong X, Zheng M, Wang Q, Li F, Zhou Y, Li P. Novel combination strategy of high intensity focused ultrasound (HIFU) and checkpoint blockade boosted by bioinspired and oxygen-supplied nanoprobe for multimodal imaging-guided cancer therapy. J Immunother Cancer 2023; 11:jitc-2022-006226. [PMID: 36650023 PMCID: PMC9853265 DOI: 10.1136/jitc-2022-006226] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND High-intensity focused ultrasound (HIFU) has shown considerable promise in treating solid tumors, but its ultrasonic energy is easily attenuated, resulting in insufficient energy accumulation in the target area. Moreover, HIFU ablation alone may inevitably lead to the presence of residual tumors, which may cause tumor recurrence and metastasis. Here, we describe a synergistic regimen combining HIFU facilitation with immunomodulation based on a novel oxygen-carrying biomimetic perfluorocarbon nanoparticle (M@P-SOP) to stimulate immunogenic cell death in tumor cells while alleviating immune suppression tumor microenvironment. METHODS M@P-SOP was prepared by double emulsion and film extrusion method. The anticancer and antimetastatic effects of M@P-SOP were evaluated on a preclinical transplanted 4T1 tumor model by combining HIFU and immunotherapy. Flow cytometry and immunofluorescence were used to clarify the potential mechanism of HIFU+M@P-SOP and their role in anti-programmed death ligand-1 (PD-L1) therapy. RESULTS Guided by photoacoustic/MR/ultrasound (US) multimodal imaging, M@P-SOP was abundantly enriched in tumor, which greatly enhanced HIFU's killing of tumor tissue in situ, induced stronger tumor immunogenic cell death, stimulated dendritic cell maturation and activated CD8+ T cells. At the same time, M@P-SOP released oxygen to alleviate the tumor hypoxic environment, repolarizing the protumor M2-type macrophages into antitumor M1-type. With concurrent anti-PD-L1 treatment, the antitumor immune response was further amplified to the whole body, and the growth of mimic distant tumor was effectively suppressed. CONCLUSIONS Our findings offer a highly promising HIFU synergist for effectively ameliorating acoustic and hypoxia environment, eventually inhibiting tumor growth and metastasis by stimulating host's antitumor immunity under HIFU ablation, especially in synergizing with PD-L1 antibody immunotherapy.
Collapse
Affiliation(s)
- Rui Tang
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Hongye He
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Xiaohong Lin
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China,Department of Ultrasound, Chongqing General Hospital, Chongqing, China
| | - Nianhong Wu
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Li Wan
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Qiaoqi Chen
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Yaqin Hu
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Chen Cheng
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Yuting Cao
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Xun Guo
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Ying Zhou
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Xialin Xiong
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Min Zheng
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Qi Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Faqi Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Yang Zhou
- Department of ultrasound, The Third People's Hospital of Chengdu City, Chengdu, People's Republic of China
| | - Pan Li
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
Sun T, Li J, Zeng C, Luo C, Luo X, Li H. Banoxantrone Coordinated Metal-Organic Framework for Photoacoustic Imaging-Guided High Intensity Focused Ultrasound Therapy. Adv Healthc Mater 2023; 12:e2202348. [PMID: 36281898 DOI: 10.1002/adhm.202202348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/18/2022] [Indexed: 01/18/2023]
Abstract
Photoacoustic (PA) imaging with high spatial resolution has great potential as desired monitoring means in the high-intensity focused ultrasound (HIFU) surgery of tumor. However, its penetration depth in the tissue is insufficient for achieving accurate intraoperative navigation, leading to residual tumor tissue. Nanomedicine provides a new opportunity for PA imaging to guide HIFU surgery. Studies have found that the hypoxic heterogeneity of tumor is effectively reversed by HIFU. Herein, a specific metal-organic framework nanosystem, constructed by coordination of banoxantrone (AQ4N) and Mn2+ , is designed based on HIFU to reverse the hypoxic heterogeneity of tumors. It can provide exogenous light-absorbing substances, thus improving the penetrability of PA imaging signal through the deep tissue and achieving clearer PA imaging for guiding HIFU surgery. In turn, AQ4N, in the hypoxic homogenous environment of the tumor provided by HIFU, is activated sequentially to specifically treat the residual hypoxic tumor cells. This combination treatment manifests higher tumor suppressors activation and lower expression of genes related to tumor progression. This strategy addresses the dissatisfaction with PA imaging-guided HIFU therapy and is promising for translation into a clinical combination regimen.
Collapse
Affiliation(s)
- Tingyu Sun
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Jingnan Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Chao Zeng
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Chengyan Luo
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Xirui Luo
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Huanan Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, P. R. China
| |
Collapse
|
16
|
Prospects for hypoxia-based drug delivery platforms for the elimination of advanced metastatic tumors: From 3D modeling to clinical concepts. J Control Release 2023; 353:1002-1022. [PMID: 36516901 DOI: 10.1016/j.jconrel.2022.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/30/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022]
Abstract
Hypoxia is a unique characteristic of the solid tumor microenvironment. Hypoxia contributes to multi-drug resistance, metastasis and cancer relapse through numerous molecular pathways, but at the same time provides an opportunity for the development of novel drugs or modalities specifically targeting hypoxic tumor regions. Given the high significance of tumor hypoxia in therapeutic results, we here discuss a variety of hypoxia-adopted strategies, and their potential and utility in the treatment of deep-seated hypoxic tumor cells. We discuss the merits and demerits of these approaches, as well as their combination with other approaches such as photodynamic therapy. We also survey the currently available 3D hypoxia modeling systems, in particular organoid-based microfluidics. Finally, we discuss the potential and the current status of preclinical tumor hypoxia approaches in clinical trials for advanced cancer. We believe that multi-modal imaging and therapeutic hypoxia adopted drug delivery platforms could provide better efficacy and safety profiles, and more importantly personalized therapy. Determining the hypoxia status of tumors could offer a second chance for the clinical translation of hypoxia-based agents, such as hypoxia activated prodrugs (HAPs) from bench to bedside.
Collapse
|
17
|
Gao D, Asghar S, Hu R, Chen S, Niu R, Liu J, Chen Z, Xiao Y. Recent advances in diverse nanosystems for nitric oxide delivery in cancer therapy. Acta Pharm Sin B 2022; 13:1498-1521. [PMID: 37139410 PMCID: PMC10149905 DOI: 10.1016/j.apsb.2022.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/26/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Gas therapy has been proven to be a promising and advantageous treatment option for cancers. Studies have shown that nitric oxide (NO) is one of the smallest structurally significant gas molecules with great potential to suppress cancer. However, there is controversy and concern about its use as it exhibits the opposite physiological effects based on its levels in the tumor. Therefore, the anti-cancer mechanism of NO is the key to cancer treatment, and rationally designed NO delivery systems are crucial to the success of NO biomedical applications. This review summarizes the endogenous production of NO, its physiological mechanisms of action, the application of NO in cancer treatment, and nano-delivery systems for delivering NO donors. Moreover, it briefly reviews challenges in delivering NO from different nanoparticles and the issues associated with its combination treatment strategies. The advantages and challenges of various NO delivery platforms are recapitulated for possible transformation into clinical applications.
Collapse
Affiliation(s)
- Dan Gao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Sajid Asghar
- Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Rongfeng Hu
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230012, China
| | - Su Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Ruixin Niu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jia Liu
- Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin 214499, China
- Corresponding authors. Tel./fax: +86 510 86700000 (Jia Liu); +86 25 85811050 (Zhipeng Chen); +86 25 83271079 (Yanyu Xiao).
| | - Zhipeng Chen
- Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Corresponding authors. Tel./fax: +86 510 86700000 (Jia Liu); +86 25 85811050 (Zhipeng Chen); +86 25 83271079 (Yanyu Xiao).
| | - Yanyu Xiao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Corresponding authors. Tel./fax: +86 510 86700000 (Jia Liu); +86 25 85811050 (Zhipeng Chen); +86 25 83271079 (Yanyu Xiao).
| |
Collapse
|
18
|
Zhang H, Du L, Wei Z, Wang X, Sojic N, Zhou X, Wang Z. Boosting the electrochemiluminescence of luminol-O 2 system by high-intensity focused ultrasound. Anal Bioanal Chem 2022; 414:8309-8315. [PMID: 36239751 DOI: 10.1007/s00216-022-04365-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/23/2022] [Accepted: 09/27/2022] [Indexed: 12/07/2022]
Abstract
Electrochemiluminescence (ECL) of luminol is a well-established methodology in analytical chemistry and bioimaging. Developing novel strategies to enhance the ECL signal of this model emitter is a challenging but rewarding task. In this work, we introduced the high-intensity focused ultrasound (HIFU), as a pretreatment means and a non-invasive way to trigger and boost the ECL signal with a 40-fold significant enhancement in the luminol-O2 system without the addition of exogenous co-reactants. The superoxide anion (O2-•) generated in situ by HIFU was the key initiator for boosting the ECL emission as demonstrated in this study for the first time. This promising co-reactant-free strategy could find potential applications for ultrasensitive ECL detection in the analysis of complex biological entities.
Collapse
Affiliation(s)
- Huixin Zhang
- College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Instrumental Analysis Center of Qingdao University, Qingdao University, Shandong, 266071, Qingdao, China
| | - Lin Du
- College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Instrumental Analysis Center of Qingdao University, Qingdao University, Shandong, 266071, Qingdao, China
| | - Zhihao Wei
- College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Instrumental Analysis Center of Qingdao University, Qingdao University, Shandong, 266071, Qingdao, China
| | - Xuemei Wang
- College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Instrumental Analysis Center of Qingdao University, Qingdao University, Shandong, 266071, Qingdao, China
| | - Neso Sojic
- Bordeaux INP, ISM, UMR CNRS 5255, University of Bordeaux, 33607, Pessac, France
- Department of Chemistry, South Ural State University, Chelyabinsk, Russian Federation, 454080
| | - Xin Zhou
- College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Instrumental Analysis Center of Qingdao University, Qingdao University, Shandong, 266071, Qingdao, China.
| | - Zonghua Wang
- College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Instrumental Analysis Center of Qingdao University, Qingdao University, Shandong, 266071, Qingdao, China.
| |
Collapse
|
19
|
Wang Y, Tang Y, Du Y, Lin L, Zhang Z, Ou X, Chen S, Wang Q, Zou J. Genetically engineered bacteria-mediated multi-functional nanoparticles for synergistic tumor-targeting therapy. Acta Biomater 2022; 150:337-352. [PMID: 35931281 DOI: 10.1016/j.actbio.2022.07.056] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/24/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022]
Abstract
Focused ultrasonic ablation surgery (FUAS) for tumor treatment has emerged as an effective non-invasive therapeutic approach, but its widespread clinical utilization is limited by its low therapeutic efficiency caused by inadequate tumor targeting, single imaging modality, and possible tumor recurrence following surgery. Therefore, this study aimed to develop a biological targeting synergistic system consisting of genetically engineered bacteria and multi-functional nanoparticles to overcome these limitations. Escherichia coli was genetically modified to carry an acoustic reporter gene encoding the formation of gas vesicles (GVs) and then target the tumor hypoxic environment in mice. After E. coli producing GVs (GVs-E. coli) colonized the tumor target area, ultrasound imaging and collaborative FUAS were performed; multi-functional nanoparticles were then enriched in the tumor target area through electrostatic adsorption. Multi-functional cationic lipid nanoparticles containing IR780, perfluorohexane, and banoxantrone dihydrochloride (AQ4N) were coloaded in the tumor to realize targeted multimodal imaging and enhance the curative effect of FUAS. AQ4N was stimulated by the tumor hypoxic environment and synergistically cooperated with FUAS to kill tumor cells. In sum, synergistic tumor therapy involving multi-functional nanoparticles mediated by genetically engineered bacteria overcomes the limitations and improves the curative effect of existing FUAS. STATEMENT OF SIGNIFICANCE: Inadequate tumor targeting, single image monitoring mode, and prone tumor recurrence following surgery remain significant challenges yet critical for tumor therapy. This study proposes a strategy for genetically engineered bacteria-mediated multifunctional nanoparticles for synergistic tumor therapy. The multifunctional genetically engineered biological targeting synergistic agent can accomplish tumor-targeting therapy, synergistic FUAS ablation, hypoxia-activated chemotherapy combined with FUAS ablation, and multiple-imaging guidance and monitoring all at the same time, thereby compensating for the shortcomings of FUAS treatment. This strategy could pave the way for the progress of tumor therapy.
Collapse
Affiliation(s)
- Yaotai Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yu Tang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yan Du
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Li Lin
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Zhong Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Xia Ou
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Sheng Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Qi Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Jianzhong Zou
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
20
|
Ding X, Wang T, Bai S, Wan Y, Zhu S, Li T, Peng N, Qiu T, Liu Y. Multifunction in One Nanoparticle for Anticancer Therapy: Bowl-Shaped Au@PDA Yolk-Shell NPs. ACS APPLIED MATERIALS & INTERFACES 2022; 14:27733-27742. [PMID: 35675694 DOI: 10.1021/acsami.2c07671] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Multifunctional nanoparticles (NPs) with simultaneous multimodal therapeutic and imaging capabilities are very necessary for biomedical applications. We successfully prepared bowl-shaped gold@polydopamine yolk-shell NPs (bowl-shaped Au@PDA YNPs) by a novel and facile method. The unique bowl-like structure enables a drug loading rate of 92% (920 μg mg-1). The bowl-shaped Au@PDA YNPs are biocompatible, have good photothermal conversion and strong near-infrared (NIR) absorption, and can control drug release under pH/NIR dual response. Bowl-shaped Au@PDA YNPs can also be employed as contrast agents for computed tomography/photoacoustic imaging for dual-modal imaging-guided chemotherapy and photothermal therapy due to the presence of Au NPs.
Collapse
Affiliation(s)
- Xin Ding
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province & Institute of Advanced Technology and Nanomaterials, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P. R. China
| | - Tianyu Wang
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Shiwei Bai
- Department of Chemistry, Northeast Normal University, Changchun, Jilin 130024, P. R. China
| | - Yunfeng Wan
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province & Institute of Advanced Technology and Nanomaterials, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P. R. China
| | - Shuai Zhu
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province & Institute of Advanced Technology and Nanomaterials, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P. R. China
| | - Tao Li
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province & Institute of Advanced Technology and Nanomaterials, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P. R. China
| | - Na Peng
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province & Institute of Advanced Technology and Nanomaterials, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P. R. China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Yi Liu
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province & Institute of Advanced Technology and Nanomaterials, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P. R. China
- State Key Laboratory of Separation Membrane and Membrane Process & Tianjin Key Laboratory of Green Chemical Technology and Process Engineering, School of Chemistry, Tiangong University, Tianjin 300387, P. R. China
| |
Collapse
|
21
|
Jiang L, Chen D, Jin Z, Xia C, Xu Q, Fan M, Dai Y, Liu J, Li Y, He Q. Light-triggered nitric oxide release and structure transformation of peptide for enhanced intratumoral retention and sensitized photodynamic therapy. Bioact Mater 2021; 12:303-313. [PMID: 35128178 PMCID: PMC8783071 DOI: 10.1016/j.bioactmat.2021.09.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/14/2021] [Accepted: 09/23/2021] [Indexed: 01/26/2023] Open
Abstract
Tumor-targeted delivery of nanomedicine is of great importance to improve therapeutic efficacy of cancer and minimize systemic side effects. Unfortunately, nowadays the targeting efficiency of nanomedicine toward tumor is still quite limited and far from clinical requirements. In this work, we develop an innovative peptide-based nanoparticle to realize light-triggered nitric oxide (NO) release and structural transformation for enhanced intratumoral retention and simultaneously sensitizing photodynamic therapy (PDT). The designed nanoparticle is self-assembled from a chimeric peptide monomer, TPP-RRRKLVFFK-Ce6, which contains a photosensitive moiety (chlorin e6, Ce6), a β-sheet-forming peptide domain (Lys-Leu-Val-Phe-Phe, KLVFF), an oligoarginine domain (RRR) as NO donor and a triphenylphosphonium (TPP) moiety for targeting mitochondria. When irradiated by light, the constructed nanoparticles undergo rapid structural transformation from nanosphere to nanorod, enabling to achieve a significantly higher intratumoral accumulation by 3.26 times compared to that without light irradiation. More importantly, the conversion of generated NO and reactive oxygen species (ROS) in a light-responsive way to peroxynitrite anions (ONOO-) with higher cytotoxicity enables NO to sensitize PDT in cancer treatment. Both in vitro and in vivo studies demonstrate that NO sensitized PDT based on the well-designed transformable nanoparticles enables to eradicate tumors efficiently. The light-triggered transformable nanoplatform developed in this work provides a new strategy for enhanced intratumoral retention and improved therapeutic outcome.
Collapse
Affiliation(s)
- Lingdong Jiang
- Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, Guangdong, China
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA
| | - Danyang Chen
- Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China
| | - Zhaokui Jin
- Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China
| | - Chao Xia
- Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China
| | - Qingqing Xu
- Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China
| | - Mingjian Fan
- Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Jia Liu
- Central Laboratory, Longgang District People's Hospital of Shenzhen & The Third Affiliated Hospital (Provisional) of The Chinese University of Hong Kong, Shenzhen, 518172, Guangdong, China
- Corresponding author.
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA
- Corresponding author.
| | - Qianjun He
- Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China
- Central Laboratory, Longgang District People's Hospital of Shenzhen & The Third Affiliated Hospital (Provisional) of The Chinese University of Hong Kong, Shenzhen, 518172, Guangdong, China
- Corresponding author. Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China.
| |
Collapse
|
22
|
Liang W, He X, Bi J, Hu T, Sun Y. Role of reactive oxygen species in tumors based on the 'seed and soil' theory: A complex interaction (Review). Oncol Rep 2021; 46:208. [PMID: 34328200 PMCID: PMC8329912 DOI: 10.3892/or.2021.8159] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor microenvironment (TME) can serve as the 'soil' for the growth and survival of tumor cells and function synergically with tumor cells to mediate tumor progression and therapeutic resistance. Reactive oxygen species (ROS) is somewhat of a double‑edged sword for tumors. Accumulating evidence has reported that regulating ROS levels can serve an anti‑tumor role in the TME, including the promotion of cancer cell apoptosis, inhibition of angiogenesis, preventing immune escape, manipulating tumor metabolic reorganization and improving drug resistance. In the present review, the potential role of ROS in anti‑tumor therapy was summarized, including the possibility of directly or indirectly targeting the TME.
Collapse
Affiliation(s)
- Wei Liang
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Affiliated Hospital of Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Xinying He
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Affiliated Hospital of Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Jianqiang Bi
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Affiliated Hospital of Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Tingting Hu
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Affiliated Hospital of Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Yunchuan Sun
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Affiliated Hospital of Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| |
Collapse
|