1
|
Sun Y, Chen M, Ye P, Yang J, Fang K, Wang C, Li R, Fan T, Shi S, Dong C. Dual inhibition of DNA damage repair sensitizes photodynamic therapy for triple negative breast cancer. Mater Today Bio 2025; 32:101706. [PMID: 40230647 PMCID: PMC11995139 DOI: 10.1016/j.mtbio.2025.101706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/21/2025] [Accepted: 03/23/2025] [Indexed: 04/16/2025] Open
Abstract
Photodynamic therapy (PDT) has emerged as an ideal candidate among reactive oxygen species (ROS)-mediated tumor therapies. However, the self-repair of DNA, a hallmark of mechanism induced by excessive intracellular ROS is one of the leading causes of PDT resistance. To overcome this challenge, we construct a therapeutic system, Ce6@MSN-ZOP-HA, wrapped with hyaluronic acid (HA) for binding to the highly expressed CD44 on triple negative breast cancer (TNBC) and loaded with DNA damage repair inhibitor (olaparib) and p38 inhibitor (SB203580). After arriving at tumor site, HA mediates drug internalization, intracellular acid microenvironment mediated ZIF8 cleavage, thus releases olaparib and SB203580. Ce6 produces ROS under 650 nm laser, SB203580 as well as olaparib increase the expression of DNA damage related molecule γH2AX, and consequent cell apoptosis, dually enhancing the PDT sensitivity of TNBC. Eventually, Ce6@MSN-ZOP-HA provides a new thought for the development of PDT synergistic nanodrugs with low biotoxicity and high anti-tumor efficiency.
Collapse
Affiliation(s)
- Yanting Sun
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, PR China
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, PR China
| | - Mengyao Chen
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, PR China
- WuXi City College of Vocational Technology, Wuxi, Jiangsu, 214153, PR China
| | - Pingting Ye
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, PR China
| | - Jingxian Yang
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, PR China
| | - Kang Fang
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, PR China
| | - Chunhui Wang
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, PR China
| | - Ruihao Li
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, PR China
| | - Ting Fan
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, PR China
| | - Shuo Shi
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, PR China
| | - Chunyan Dong
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, PR China
| |
Collapse
|
2
|
Qu H, Hang L, Diao Y, Wang H, Fang L, Liu W, Liu J, Sun H, Wang J, Meng X, Li H, Jiang G. Mn-doped MOF nanoparticles mitigating hypoxia via in-situ substitution strategy for dual-imaging guided combination treatment of microwave dynamic therapy and chemotherapy. J Colloid Interface Sci 2025; 685:912-926. [PMID: 39874828 DOI: 10.1016/j.jcis.2025.01.202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/31/2024] [Accepted: 01/23/2025] [Indexed: 01/30/2025]
Abstract
Microwave dynamic therapy (MWDT) destroy tumor cells using reactive oxygen species (ROS), but its effectiveness is limited by low ROS production and intracellular oxygen (O2) availability. This study presents a novel strategy using manganese (II) ion (Mn2+) doped iron (Fe)-based metal-organic framework (Fe MOF) nanoparticles (NPs) to enhance both O2 generation and ROS production for improved MWDT. Incorporating Mn2+ into Fe MOF narrows the bandgap from 0.673 eV to 0.429 eV, improving the separation of electronic-hole pair and increasing ROS yield. Meanwhile, Mn-porphyrin nanocomplexes facilitate the decomposition of hydrogen peroxide to O2in situ. Additionally, encapsulating the chemotherapeutic drug gemcitabine (GEM) within NPs and surface-modifying with Pluronic F127 creates Mn-Fe MOF@GEM@F127 (MMGF) NPs, which are suitable for photoacoustic/magnetic resonance imaging guidance (relaxivity, r1: 2.007 mM-1s-1). The microwave (MW)/pH dual responsive GEM release works synergistically with MWDT, thereby more effectively disrupting tumor cells. This strategy differs from monotherapy by using MW sensitizers to enhance O2 production, which not only increases the efficiency of ROS generation in MWDT but also makes subsequent chemotherapy more effective while reducing the side effects of conventional chemotherapy. This combined treatment reduced HONE-1 cell proliferation and tumor growth by 89.95 % and 96.12 %, respectively. The study proposes a versatile strategy to significantly improve both MWDT and chemotherapy efficacy with potential applications to various cancers.
Collapse
Affiliation(s)
- Hong Qu
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Jinan University, Guangzhou 518037 China; College of Chemistry and Materials Science, Jinan University, Guangzhou 510632 China
| | - Lifeng Hang
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Jinan University, Guangzhou 518037 China
| | - Yanzhao Diao
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Jinan University, Guangzhou 518037 China
| | - Haiying Wang
- Guangdong Medical University, Zhanjiang 524023 China
| | - Laiping Fang
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Jinan University, Guangzhou 518037 China
| | - Wangzi Liu
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632 China
| | - Jinwu Liu
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Jinan University, Guangzhou 518037 China
| | - Hui Sun
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Jinan University, Guangzhou 518037 China
| | - Jizhuang Wang
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632 China
| | - Xianwei Meng
- Laboratory of Controllable Preparation and Application of Nanomaterials Laboratory of Cryogenics, Technical Institute of Physics and Chemistry Chinese Academy of Sciences, Beijing 100190 China.
| | - Hong Li
- College of Chemistry and Materials Science, Jinan University, Guangzhou 510632 China.
| | - Guihua Jiang
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Jinan University, Guangzhou 518037 China.
| |
Collapse
|
3
|
He J, Wang G, Zhou Y, Li B, Shang P. Recent advances in polydopamine-coated metal-organic frameworks for cancer therapy. Front Bioeng Biotechnol 2025; 13:1553653. [PMID: 40291560 PMCID: PMC12023280 DOI: 10.3389/fbioe.2025.1553653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/04/2025] [Indexed: 04/30/2025] Open
Abstract
The creation and development of classical multifunctional nanomaterials are crucial for the advancement of nanotherapeutic treatments for tumors. Currently, metal-organic frameworks (MOFs) modified with polydopamine (PDA) are at the forefront of nanomedicine research, particularly in tumor diagnostics and therapy, owing to their exceptional biocompatibility, expansive specific surface area, multifaceted functionalities, and superior photothermal properties, which led to significant advancements in anti-tumor research. Consequently, a range of anti-cancer strategies has been devised by leveraging the exceptional capabilities of MOFs, including intelligent drug delivery systems, photodynamic therapy, and photothermal therapy, which are particularly tailored for the tumor microenvironment. In order to gain deeper insight into the role of MOFs@PDA in cancer diagnosis and treatment, it is essential to conduct a comprehensive review of existing research outcomes and promptly analyze the challenges associated with their biological applications. This will provide valuable perspectives on the potential of MOFs@PDA in clinical settings.
Collapse
Affiliation(s)
- Jingchao He
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Key Laboratory of the Jiangsu Higher Education Institutions for Nucleic Acid and Cell Fate Regulation, Yangzhou University, Yangzhou, China
| | - Guangtian Wang
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Yongfang Zhou
- Department of Oncology, Jining Cancer Hospital, Jining, China
| | - Bin Li
- Department of Biochemistry and Molecular Biology, Medical College, Guangxi University of Science and Technology, Liuzhou, China
| | - Pan Shang
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, China
| |
Collapse
|
4
|
Akbari Oryani M, Tarin M, Rahnama Araghi L, Rastin F, Javid H, Hashemzadeh A, Karimi-Shahri M. Synergistic cancer treatment using porphyrin-based metal-organic Frameworks for photodynamic and photothermal therapy. J Drug Target 2025; 33:473-491. [PMID: 39618308 DOI: 10.1080/1061186x.2024.2433551] [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: 09/23/2024] [Revised: 11/03/2024] [Accepted: 11/18/2024] [Indexed: 02/25/2025]
Abstract
Recent advancements in multifunctional nanomaterials for cancer therapy have highlighted porphyrin-based metal-organic frameworks (MOFs) as promising candidates due to their unique properties and versatile applications. This overview focuses on the use of porphyrin-based MOFs for combined photodynamic therapy (PDT) and photothermal therapy (PTT) in cancer treatment. Porphyrin-based MOFs offer high porosity, tuneable structures, and excellent stability, making them ideal for drug delivery and therapeutic applications. The incorporation of porphyrin molecules into the MOF framework enhances light absorption and energy transfer, leading to improved photodynamic and photothermal effects. Additionally, the porosity of MOFs allows for the encapsulation of therapeutic agents, further enhancing efficacy. In PDT, porphyrin-based MOFs generate reactive oxygen species (ROS) upon light activation, destroying cancer cells. The photothermal properties enable the conversion of light energy into heat, resulting in localised hyperthermia and tumour ablation. The combination of PDT and PTT in a single platform offers synergistic effects, leading to better therapeutic outcomes, reduced side effects, and improved selectivity. This dual-modal treatment strategy provides precise spatiotemporal control over the treatment process, paving the way for next-generation therapeutics with enhanced efficacy and reduced side effects. Further research and optimisation are needed for clinical applications.
Collapse
Affiliation(s)
- Mahsa Akbari Oryani
- Department of Pathology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Tarin
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Leila Rahnama Araghi
- Department of Biotechnology, Faculty of Science, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Farangis Rastin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Hashemzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
5
|
Xiang G, Wang H, Lu C, Yu S, Wu A, Wang X. Luteolin-Manganese Nanozyme Induces Apoptosis and Ferroptosis for Enhanced Cancer Therapy. Inorg Chem 2025; 64:3885-3897. [PMID: 39969912 DOI: 10.1021/acs.inorgchem.4c05083] [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: 02/20/2025]
Abstract
Cancer presents a significant global public health challenge that impacts millions of individuals worldwide. The incorporation of natural products into cancer treatment has the potential to mitigate many of the side effects commonly associated with chemotherapy. This study builds on the advantages of enhancing the anticancer activity of natural flavonoids through metal chelation by synthesizing a natural antioxidant flavonoid complex, termed Lu-Mn nanozyme, which involves the chelation of luteolin with manganese ions. In vitro experiments demonstrated that Lu-Mn exhibits a strong affinity for hydrogen peroxide (H2O2) and effectively catalyzes the generation of hydroxyl radicals (•OH) from H2O2 within the tumor microenvironment. The administration of the Lu-Mn nanozyme not only induced apoptosis in tumor cells by upregulating the expression of cleaved caspase3 and caspase9 but also activated ferroptosis through downregulation of the NRF2-GPX4 signaling pathway. Furthermore, animal studies have shown that Lu-Mn possesses significant antitumor efficacy and a favorable safety profile. Collectively, these findings suggest that luteolin, through its chelation with metal ions, has considerable potential for application in cancer treatment.
Collapse
Affiliation(s)
- Gang Xiang
- College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China
| | - Hui Wang
- College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China
| | - Changfang Lu
- College of Science, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Siyuan Yu
- College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China
| | - Aimin Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Xianxiang Wang
- College of Science, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| |
Collapse
|
6
|
Wang R, Liu Y, Liu M, Zhang M, Li C, Xu S, Tang S, Ma Y, Wu X, Fei W. Combating tumor PARP inhibitor resistance: Combination treatments, nanotechnology, and other potential strategies. Int J Pharm 2025; 669:125028. [PMID: 39638266 DOI: 10.1016/j.ijpharm.2024.125028] [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: 09/04/2024] [Revised: 11/14/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
PARP (poly (ADP-ribose) polymerase) inhibitors (PARPi) have demonstrated significant potential in cancer treatment, particularly in tumors with breast cancer susceptibility gene (BRCA) mutations and other DNA repair deficiencies. However, the development of resistance to PARPi has become a major challenge in their clinical application. The emergence of drug resistance leads to reduced efficacy of the PARPi over time, impacting long-term treatment outcomes and survival rates. PARPi resistance in tumors often arises as cells activate alternative DNA repair pathways or evade the effect of PARPi, diminishing therapeutic effectiveness. Consequently, overcoming resistance is crucial for maintaining treatment efficacy and improving patient prognosis. This paper reviews the strategies to overcome PARPi resistance through combination treatment and nanotechnology therapy. We first review the current combination therapies with PARPi, including anti-angiogenic therapies, radiotherapies, immunotherapies, and chemotherapies, and elucidate their mechanisms for overcoming PARPi resistance. Additionally, this paper focuses on the application of nanotechnology in improving the effectiveness of PARPi and overcoming drug resistance. Subsequently, this paper presents several promising strategies to tackle PARPi resistance, including but not limited to: structural modifications of PARPi, deployment of gene editing systems, implementation of "membrane lipid therapy," and modulation of cellular metabolism in tumors. By integrating these strategies, this research will provide comprehensive approaches to overcome the resistance of PARPi in cancer treatment and offer guidance for future research and clinical practice.
Collapse
Affiliation(s)
- Rong Wang
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yunxi Liu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Mingqi Liu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Meng Zhang
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Chaoqun Li
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Shanshan Xu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Sangsang Tang
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yidan Ma
- YiPeng Subdistrict Community Healthcare Center, Hangzhou 311225, China
| | - Xiaodong Wu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Weidong Fei
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
7
|
Umar AK, Limpikirati PK, Rivai B, Ardiansah I, Sriwidodo S, Luckanagul JA. Complexed hyaluronic acid-based nanoparticles in cancer therapy and diagnosis: Research trends by natural language processing. Heliyon 2025; 11:e41246. [PMID: 39811313 PMCID: PMC11729671 DOI: 10.1016/j.heliyon.2024.e41246] [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: 08/11/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Hyaluronic acid (HA) is a popular surface modifier in targeted cancer delivery due to its receptor-binding abilities. However, HA alone faces limitations in lipid solubility, biocompatibility, and cell internalization, making it less effective as a standalone delivery system. This comprehensive study aimed to explore a dynamic landscape of complexation in HA-based nanoparticles in cancer therapy, examining diverse aspects from influential modifiers to emerging trends in cancer diagnostics. We discovered that certain active substances, such as 5-aminolevulinic acid, adamantane, and protamine, have been on trend in terms of their usage over the past decade. Dextran, streptavidin, and catechol emerge as intriguing conjugates for HA, coupled with nanostar, quantum dots, and nanoprobe structures for optimal drug delivery and diagnostics. Strategies like hypoxic conditioning, dual responsiveness, and pulse laser activation enhance controlled release, targeted delivery, and real-time diagnostic techniques like ultrasound imaging and X-ray computed tomography (X-ray CT). Based on our findings, conventional bibliometric tools fail to highlight relevant topics in this area, instead producing merely abstract and broad-meaning keywords. Extraction using Named Entity Recognition and topic search with Latent Dirichlet Allocation successfully revealed five representative topics with the ability to exclude irrelevant keywords. A shift in research focuses from optimizing chemical toxicity to particular targeting tactics and precise release mechanisms is evident. These findings reflect the dynamic landscape of HA-based nanoparticle research in cancer therapy, emphasizing advancements in targeted drug delivery, therapeutic efficacy, and multimodal diagnostic approaches to improve overall patient outcomes.
Collapse
Affiliation(s)
- Abd Kakhar Umar
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Medical Informatics Laboratory, ETFLIN, Palu City, 94225, Indonesia
| | - Patanachai K. Limpikirati
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Metabolomics for Life Sciences Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Bachtiar Rivai
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Medical Informatics Laboratory, ETFLIN, Palu City, 94225, Indonesia
| | - Ilham Ardiansah
- Department of Animal Husbandry, Faculty Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Medical Informatics Laboratory, ETFLIN, Palu City, 94225, Indonesia
| | - Sriwidodo Sriwidodo
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| | - Jittima Amie Luckanagul
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
8
|
Hassanzadeh Goji N, Alibolandi M, Ramezani M, Saljooghi AS, Dayyani M, Nekooei S. A four in one nanoplatform: Theranostic bismuth-containing nanoMOFs for chemo-photodynamic- radiation therapy and CT scan imaging. Int J Pharm 2025; 668:124971. [PMID: 39566700 DOI: 10.1016/j.ijpharm.2024.124971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/16/2024] [Accepted: 11/16/2024] [Indexed: 11/22/2024]
Abstract
Integration of different therapeutic performances into one platform is an innovative development for using multiple applications in real-time. In this paper, for the first time we exploited the concurrent capacity of radio and photosensitizing in a theranostic nanoMOFs based on bismuth, zirconium, and porphyrin. The porosity of nanoMOFs provided the capability of doxorubicin loading and chemotherapy besides enhanced photodynamic and radiation therapy (PDT & RT). Its PEGylation and aptamer (MUC1) immobilization endowed the platform with high biocompatibility and targeted tumor killing, respectively. In vitro assay exhibited that this aptamer immobilized DOX-loaded PEGylated MOF (Apt@DOX) produced more toxicity against 4 T1 cells compared to non-targeted nanoparticles (NP@DOX), especially when the treatment combined with PDT or/and RT. In vivo experiment also provided great results for tumor growth, survival rate, and body weight for 4 T1 bearing mice injected by Apt@DOX in combination with irradiation by 660 nm laser and/or exposure to 3 Gy dosage of X-ray radiation. The CT imaging of injected mice with targeted and non-targeted bismuth-based MOF introduced this nanoplatform as a promising CT contrast agent. Resultantly, we can present our as-synthesized nanoplatform as an efficient multifunctional theranostics with the ability of multimodal therapy and diagnostic performance.
Collapse
Affiliation(s)
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Amir Sh Saljooghi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Mahdieh Dayyani
- Radiation Oncology Department, Reza Radiotherapy and Oncology Center, Mashhad, Iran
| | - Sirous Nekooei
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Mondal S, Park S, Nguyen VT, Doan VHM, Choi J, Ly CD, Phan DT, Truong TT, Vo TH, Nguyen DT, Pal U, Lee B, Oh J. Precision Cancer Therapy Enabled Anti-Epidermal Growth Factor Receptor-Conjugated Manganese Core Phthalocyanine Bismuth Nanocomposite for Dual Imaging-Guided Breast Cancer Treatment. Biomater Res 2024; 2024:0092. [PMID: 39525484 PMCID: PMC11542904 DOI: 10.34133/bmr.0092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/04/2024] [Accepted: 09/24/2024] [Indexed: 11/16/2024] Open
Abstract
Cancer remains a formidable global health challenge, demanding the exploration of innovative treatment modalities with minimized side effects. One promising avenue involves the synergistic integration of targeted photothermal/photodynamic therapy (PTT/PDT), utilizing specially designed functional nanomaterials for precise cancer diagnosis and treatment. This study introduces a composite biomaterial, anti-epidermal growth factor receptor-conjugated manganese core phthalocyanine bismuth (anti-EGFR-MPB), synthesized for precise cancer imaging and treatment. The biomaterial, synthesized via a solvothermal process, effectively treats and images breast cancer in mouse models. Its biomimetic design targets cancer cells precisely, with dual imaging for real-time monitoring. The biomimetic design of the composite enables precise targeting of cancer cells, whereas the dual imaging allows for real-time visualization and monitoring of the treatment. In vivo examinations confirm substantial damage to tumor tissues with no recurrence following 808-nm laser irradiation. The composite shows strong fluorescence/photoacoustic imaging (PAI) contrast, aiding malignancy detection. Biological assays and histological analyses confirmed the efficacy of the nanocomposite in inducing apoptosis in cancer cells. The integrated targeted dual image-guided phototherapy offered by this composite substantially enhances the precision and efficacy of cancer therapy, achieving an impressive photothermal efficiency of ~33.8%. Our findings demonstrate the utility of the anti-EGFR-MPB nanocomposite for both in vitro and in vivo photoacoustic image-guided PTT and PDT. The optimal treatment strategy for triple-negative breast cancer is found to be the use of 250 μg/ml of nanocomposite irradiated with 1.0 W/cm2 808-nm laser for 7 min.
Collapse
Affiliation(s)
- Sudip Mondal
- Digital Healthcare Research Center, Pukyong National University
| | - Sumin Park
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Van Tu Nguyen
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Vu Hoang Minh Doan
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Jaeyeop Choi
- Smart Gym-Based Translational Research Center for Active Senior’s Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Cao Duong Ly
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Duc Tri Phan
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Thi Thuy Truong
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Tan Hung Vo
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Dinh Tuan Nguyen
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Umapada Pal
- Institute of Physics, Autonomous University of Puebla, Puebla, Pue. 72570, Mexico
| | - Byeongil Lee
- Digital Healthcare Research Center, Pukyong National University
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea
- Smart Gym-Based Translational Research Center for Active Senior’s Healthcare, Pukyong National University, Busan 48513, Republic of Korea
- Department of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Junghwan Oh
- Digital Healthcare Research Center, Pukyong National University
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea
- Smart Gym-Based Translational Research Center for Active Senior’s Healthcare, Pukyong National University, Busan 48513, Republic of Korea
- Department of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
- Ohlabs Corp., Busan 48513, Republic of Korea
| |
Collapse
|
10
|
Li J, Lei D, Cao Y, Xin F, Zhang Z, Liu X, Wu M, Yao C. Nanozyme Decorated Metal-Organic Framework Nanosheet for Enhanced Photodynamic Therapy Against Hypoxic Tumor. Int J Nanomedicine 2024; 19:9727-9739. [PMID: 39315364 PMCID: PMC11418915 DOI: 10.2147/ijn.s466011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024] Open
Abstract
INTRODUCTION Photodynamic therapy (PDT) has attracted increasing attention in the clinical treatment of epidermal and luminal tumors. However, the PDT efficacy in practice is severely impeded by tumor hypoxia and the adverse factors associated with hydrophobic photosensitizers (PSs), including low delivery capacity, poor photoactivity and limited ROS diffusion. In this study, Pt nanozymes decorated two-dimensional (2D) porphyrin metal-organic framework (MOF) nanosheets (PMOF@HA) were fabricated and investigated to conquer the obstacles of PDT against hypoxic tumors. MATERIALS AND METHODS PMOF@HA was synthesized by the coordination of transition metal iron (Zr4+) and PS (TCPP), in situ generation of Pt nanozyme and surface modification with hyaluronic acid (HA). The abilities of hypoxic relief and ROS generation were evaluated by detecting the changes of O2 and 1O2 concentration. The cellular uptake was investigated using flow cytometry and confocal laser scanning microscopy. The SMMC-7721 cells and the subcutaneous tumor-bearing mice were used to demonstrate the PDT efficacy of PMOF@HA in vitro and in vivo, respectively. RESULTS Benefiting from the 2D structure and inherent properties of MOF materials, the prepared PMOF@HA could not only serve as nano-PS with high PS loading but also ensure the rational distance between PS molecules to avoid aggregation-induced quenching, enhance the photosensitive activity and promote the rapid diffusion of generated radical oxide species (ROS). Meanwhile, Pt nanozymes with catalase-like activity effectively catalyzed intratumoral overproduced H2O2 into O2 to alleviate tumor hypoxia. Additionally, PMOF@HA, with the help of externally coated HA, significantly improved the stability and increased the cell uptake by CD44 overexpressed tumor cells to strengthen O2 self-supply and PDT efficacy. CONCLUSION This study provided a new strategy of integrating 2D porphyrin MOF nanosheets with nanozymes to conquer the obstacles of PDT against hypoxic tumors.
Collapse
Affiliation(s)
- Jiong Li
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Photonics and Sensing, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Dongqin Lei
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Photonics and Sensing, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Yanbing Cao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People’s Republic of China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, People’s Republic of China
| | - Fuli Xin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People’s Republic of China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, People’s Republic of China
| | - Zhenxi Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Photonics and Sensing, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People’s Republic of China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, People’s Republic of China
| | - Ming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People’s Republic of China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, People’s Republic of China
| | - Cuiping Yao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Photonics and Sensing, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| |
Collapse
|
11
|
Wang Y, Gao N, Li X, Ling G, Zhang P. Metal organic framework-based variable-size nanoparticles for tumor microenvironment-responsive drug delivery. Drug Deliv Transl Res 2024; 14:1737-1755. [PMID: 38329709 DOI: 10.1007/s13346-023-01500-x] [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] [Accepted: 12/13/2023] [Indexed: 02/09/2024]
Abstract
Nanoparticles (NPs) have been designed for the treatment of tumors increasingly. However, the drawbacks of single-size NPs are still worth noting, as their circulation and metabolism in the blood are negatively correlated with their accumulation at the tumor site. If the size of single-size NPs is too small, it will be quickly cleared in the blood circulation, while, the size is too large, the distribution of NPs in the tumor site will be reduced, and the widespread distribution of NPs throughout the body will cause systemic toxicity. Therefore, a class of variable-size NPs with metal organic frameworks (MOFs) as the main carrier, and size conversion in compliance with the characteristics of the tumor microenvironment (TME), was designed. MOF-based variable-size NPs can simultaneously extend the time of blood circulation and metabolism, then enhance the targeting ability of the tumor site. In this review, MOF NPs are categorized and exemplified from a new perspective of NP size variation; the advantages, mechanisms, and significance of MOF-based variable-size NPs were summarized, and the potential and challenges in delivering anti-tumor drugs and multimodal combination therapy were discussed.
Collapse
Affiliation(s)
- Yu Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Nan Gao
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Xiaodan Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Guixia Ling
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China.
| | - Peng Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China.
| |
Collapse
|
12
|
Bai Z, Huang J, Lu H, Wang N, Li H, Zhu Y. Based on polydopamine-coated metal organic framework multifunctional nanoplatform for enhanced photothermal/sonodynamicand treatment combined with checkpoint blockade therapy. Int J Biol Macromol 2024; 269:132207. [PMID: 38723823 DOI: 10.1016/j.ijbiomac.2024.132207] [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: 01/19/2024] [Revised: 04/08/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024]
Abstract
To overcome the low efficacy of sonodynamic therapy (SDT) caused by hypoxia in the tumor microenvironment, we developed a multiple anti-tumor nanoplatform with synergistic SDT, photothermal therapy (PTT), and ferroptosis effects. PCN-224@FcCaO2/Mn/dihydroartemisinin/imiquimod/PDA (PFC) was prepared by modified with dihydroartemisinin (DHA), imiquimod (R837), CaO2, ferrocene (Fc) and Mn2+ on the PCN-224 (Cu) to achieve self-replenishment of H2O2/O2 and GSH consumption. FcCaO2 decomposed into H2O2 in the tumor microenvironment, triggering the Fenton effect to produce OH, and Cu2+ reduced the potential loss of OH by the depletion of GSH. Under ultrasonic (US) and laser irradiation, PFC exhibits exciting PTT and SDT effects from polydopamine (PDA) and PCN-224. Mn2+ not only promoted the reaction of H2O2 to produce O2 to effectively enhance SDT but also induced tumor cell apoptosis by Mn2+ combined with DHA. PFC induced ferroptosis via Fe interaction with DHA to produce ROS and reduce the expression of GPX4. The released R837 and tumor-associated antigens from SDT/PTT can produce damage associated molecular patterns (DAMPs), which can initiate adaptive immune responses to kill cancer cells, and released again to promote the tumor immune cycle. What's more, SDT/PTT and ferroptosis combined with aPD-L1 can effectively suppress both primary and distant tumor growth.
Collapse
Affiliation(s)
- Zhihao Bai
- College of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, China; State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China
| | - JianFeng Huang
- Department of nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China
| | - HaiZhen Lu
- Department of nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China
| | - Nannan Wang
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China.
| | - HaoYu Li
- Department of nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China.
| | - Yanqiu Zhu
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China; College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter EX4 4QF, UK.
| |
Collapse
|
13
|
Hang L, Li M, Zhang Y, Li W, Fang L, Chen Y, Zhou C, Qu H, Shao L, Jiang G. Mn(II) Optimized Sono/Chemodynamic Effect of Porphyrin-Metal-Organic Framework Nanosheets for MRI-Guided Colon Cancer Therapy and Metastasis Suppression. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306364. [PMID: 37997202 DOI: 10.1002/smll.202306364] [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: 07/26/2023] [Revised: 10/10/2023] [Indexed: 11/25/2023]
Abstract
Sonodynamic therapy (SDT) offers a remarkable non-invasive ultrasound (US) treatment by activating sonosensitizer and generating reactive oxygen species (ROS) to inhibit tumor growth. The development of multifunctional, biocompatible, and highly effective sonosensitizers remains a current priority for SDT. Herein, the first report that Mn(II) ions chelated Gd-TCPP (GMT) nanosheets (NSs) are synthesized via a simple reflux method and encapsulated with pluronic F-127 to form novel sonosensitizers (GMTF). The GMTF NSs produce a high yield of ROS under US irradiation due to the decreased highest occupied molecular orbital-lowest unoccupied molecular orbital gap energy (2.7-1.28 eV). Moreover, Mn(II) ions endow GMTF with a fascinating Fenton-like activity to produce hydroxyl radicals in support of chemodynamic therapy (CDT). It is also effectively used in magnetic resonance imaging (MRI) with high relaxation rate (r 1: 4.401 mM-1 s-1) to track the accumulation of NSs in tumors. In vivo results indicate that the SDT and CDT in combination with programmed cell death protein 1 antibody (anti-PD-1) show effective metastasis prevention effects, and 70% of the mice in the GMTF + US + anti-PD-1 group survived for 60 days. In conclusion, this study develops a sonosensitizer with promising potential for utilizing both MRI-guided SDT and CDT strategies.
Collapse
Affiliation(s)
- Lifeng Hang
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, Guangdong Second Provincial General Hospital, Guangzhou, 518037, P. R. China
| | - Meng Li
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, Guangdong Second Provincial General Hospital, Guangzhou, 518037, P. R. China
| | - Yuxuan Zhang
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, P. R. China
| | - Wuming Li
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, Guangdong Second Provincial General Hospital, Guangzhou, 518037, P. R. China
| | - Laiping Fang
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, Guangdong Second Provincial General Hospital, Guangzhou, 518037, P. R. China
| | - Yiyu Chen
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, Guangdong Second Provincial General Hospital, Guangzhou, 518037, P. R. China
| | - Chunze Zhou
- Interventional Radiology Department, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, P. R. China
| | - Hong Qu
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, Guangdong Second Provincial General Hospital, Guangzhou, 518037, P. R. China
| | - Lianyi Shao
- School of Materials and Energy, Guangdong University of Technology, Guangzhou, 510006, P. R. China
| | - Guihua Jiang
- The Department of Medical Imaging, Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, Guangdong Second Provincial General Hospital, Guangzhou, 518037, P. R. China
| |
Collapse
|
14
|
Akbarzadeh F, Khoshgard K. Enhancement of the effect of novel targeted 5-aminolevulinic acid conjugated bismuth oxide nanoparticles-based photodynamic therapy by simultaneous radiotherapy on KB cells. Photodiagnosis Photodyn Ther 2024; 46:104025. [PMID: 38403143 DOI: 10.1016/j.pdpdt.2024.104025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/30/2024] [Accepted: 02/21/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND Selective accumulation of photosensitizers into cancerous cells is one of the most important factors affecting photodynamic therapy (PDT) efficacy. 5-aminolevulinic acid (5-ALA) is the precursor of a strong photosensitizer, protoporphyrin-IX; but it has poor permeability into the cells. Folate receptors are overexpressed on the surface of many tumor cells. In the present study, folic acid (FA) and 5-ALA conjugated bismuth oxide nanoparticles were synthesized; and used in PDT, radiotherapy (RT), and concurrent PDT & RT against nasopharyngeal carcinoma (KB cell line). METHODS The KB cells were incubated with the synthesized nanoparticles (NPs) for 2 h; then illuminated using a custom-made LED lamp at the light dose of 26 J/cm2. Irradiation of the cells was carried out using X-ray 6 MV (2 Gy); and synergistic effect of the simultaneous RT and PDT treatments was evaluated using fractional product values. Efficacy of the treatments was determined using MTT and Caspase-3 enzyme activity assays. RESULTS Targeting of folic acid receptors enables the selective endocytosis of the conjugated NPs. RT results in the presence of Bi2O3 NPs showed a significant radiosensitizer potential of these NPs. Fractional product values of 1.49±0.05, 1.36±0.06, and 1.05±0.06 obtained in the presence of FA-5-ALA conjugated NPs, 5-ALA conjugated NPs, and in the absence of the NPs, respectively. Therefore, simultaneous RT and PDT in the presence of these conjugated NPs is superior to RT in the presence of the NPs. CONCLUSION Simultaneous PDT and RT in the presence of FA-5-ALA conjugated bismuth oxide NPs can be introduced as a promising therapeutic approach in controlling KB cancer cells.
Collapse
Affiliation(s)
- Fatemeh Akbarzadeh
- Students Research Committee, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Karim Khoshgard
- Department of Medical Physics, School of Medicine, Kermanshah University of Medical Sciences, Sorkheh-Lizhe Blvd, Kermanshah, P.O.Box:1568, Iran.
| |
Collapse
|
15
|
Zhang H, Yuan W. Self-healable oxide sodium alginate/carboxymethyl chitosan nanocomposite hydrogel loading Cu 2+-doped MOF for enhanced synergistic and precise cancer therapy. Int J Biol Macromol 2024; 262:129996. [PMID: 38342271 DOI: 10.1016/j.ijbiomac.2024.129996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/25/2024] [Accepted: 02/04/2024] [Indexed: 02/13/2024]
Abstract
The limitations of traditional therapeutic methods such as chemotherapy serious restricted the application in tumor treatment, including poor targeting, toxic side effects and poor precision. It is important to develop non-chemotherapeutic systems to achieve precise and efficient tumor treatment. Therefore, a functional metal-organic framework material (MOF) with porphyrin core and doped with Cu2+ and surface-modified with polydopamine (PDA), namely PCN-224(Cu)@PDA (PCP) was designed and prepared. After loaded into the injectable and self-healable hydrogels by dynamic Schiff base bonding of oxidized sodium alginate (OSA) and carboxymethyl chitosan (CMC), the multifunctional nanocomposite hydrogels were obtained, in which Cu2+ in MOF converts to Cu+ by reacting with glutathione (GSH) which reduces the tumor antioxidant activity to improve the CDT effect. The Cu2+/Cu+ induces Fenton-like reaction in tumor cells to produce a toxic hydroxyl radical (OH). PDA achieves photothermal conversion under NIR light for photothermal therapy (PTT), and porphyrin core as a ligand generates reactive oxygen species (ROS), presenting highly efficient photodynamic therapy (PDT). Injectable self-healing hydrogel as a loading platform can be in situ injected to tumor site to release PCP and endocytosed by tumor cells to achieve precise and synergistic CDT-PDT-PTT therapy.
Collapse
Affiliation(s)
- Hanyan Zhang
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China
| | - Weizhong Yuan
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China.
| |
Collapse
|
16
|
Dai R, Liu Q, Zhang B, Zhang X, Gao M, Li D, Kang W, Chen L, Zhao M, Zheng Z, Zhang R. A Single NIR-II Laser-Triggered Self-Enhancing Photo/Enzyme-coupled Three-in-One Nanosystems for Breast Cancer Phototheranostics. Adv Healthc Mater 2024; 13:e2302783. [PMID: 38016674 DOI: 10.1002/adhm.202302783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 11/22/2023] [Indexed: 11/30/2023]
Abstract
Multifunctional phototheranostics, employing precise and non-invasive techniques, is widely developed to enhance theranostic efficiency of breast cancer (BC), reduce side-effects, and improve quality of life. Integrating all phototheranostic modalities into a single photosensitizer for highly effective BC treatment is particularly challenging due to the potential inefficiency and time consumption associated with repeated switching of multiple-wavelength lasers. Herein, a novel single NIR-II laser-triggered three-in-one nanosystem(PdCu NY) is rationally designed, which enables dual-modal (NIR-II FL/NIR-II PA) imaging-guided self-enhancing photothermal-photodynamic therapy (PTT-PDT) in NIR-II window. The PdCu NY based on optimal Pd/Cu molar-ratio(1:11) can be easily fabricated and large-scale production for simultaneous PTT-PDT against BC under a single 1064nm laser irradiation. Significantly, the PdCu NY acted as a promising photocatalyst for decomposition of H2O into O2 upon the same laser irradiation. In addition, the inherent catalase (CAT)-like activity of PdCu NYs enables photo-enzyme dual-catalytic O2 supply to effectively alleviate hypoxia, achieving self-enhanced PDT efficiency. These PTT-PDT self-enhanced nanosystems demonstrate precise lesion localization and complete tumor ablation using a single 1064nm laser source by "one-laser, multi-functions" strategy. More importantly, this study not only reports a three-in-one PdCu-based phototheranostic agent, but also sheds light on the exploration of versatile biosafety nanosystems for clinical applications.
Collapse
Affiliation(s)
- Rong Dai
- Department of Radiology, First hospital of Shanxi Medical University, Taiyuan, 030001, China
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
| | - Qi Liu
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
- Department of Radiology, Shanxi cardiovascular hospital, Taiyuan, 030000, China
| | - Binyue Zhang
- Department of Radiology, First hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Xin Zhang
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
| | - Mengting Gao
- Department of Radiology, First hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Dongsheng Li
- Department of Radiology, First hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Weiwei Kang
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
| | - Lin Chen
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
| | - Mingxin Zhao
- Department of Radiology, First hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Ziliang Zheng
- Department of Radiology, First hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Ruiping Zhang
- Department of Radiology, First hospital of Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
17
|
Chen Y, Lu Z, Wang D. Multifunctional Nanoplatform for Single NIR Laser-Regulated Efficient PDT/PTT/Chemotherapy. Biomacromolecules 2024; 25:1038-1046. [PMID: 38242167 DOI: 10.1021/acs.biomac.3c01100] [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: 01/21/2024]
Abstract
The combination of phototherapy and chemotherapy with superior advantages is a promising strategy for cancer therapy. However, combination therapy is generally regulated by two different wavelengths of light or other stimuli, which results in complex operations and inevitable systemic side effects, even affecting therapeutic efficacy. Herein, we design a signal NIR light-regulated nanoplatform via the self-assembly process of reactive oxygen species (ROS)-sensitive prodrug (DTD), human serum albumin (HSA), and IR780 for combined photothermal/photodynamic therapy and chemotherapy. Upon 808 nm laser irradiation, IR780 in nanoparticles generates abundant ROS and a significant photothermal effect to achieve photothermal/photodynamic therapy. Meanwhile, the generating ROS further cleans up the thioketal link to release DOX for chemotherapy. Hence, signal NIR light can effectively control the process of combination therapy. In vivo and in vitro experiment results demonstrate that the multifunctional nanoparticles exhibit excellent antitumor efficacy via the combination of phototherapy and chemotherapy controlled by a signal NIR laser. Overall, the signal NIR light-regulated nanoparticles with combination therapy performance provide a versatile platform for enhancing antitumor efficacy.
Collapse
Affiliation(s)
- Yu Chen
- Key Laboratory of Textile Fiber and Products, Ministry of Education, Wuhan Textile University, Wuhan 430200, China
| | - Zhentan Lu
- Key Laboratory of Textile Fiber and Products, Ministry of Education, Wuhan Textile University, Wuhan 430200, China
| | - Dong Wang
- Key Laboratory of Textile Fiber and Products, Ministry of Education, Wuhan Textile University, Wuhan 430200, China
| |
Collapse
|
18
|
Zhang S, Zhang M, Zhang J, Li G, Lu X, Sun F, Liu W. Photoresponsive metal-organic framework with combined photodynamic therapy and hypoxia-activated chemotherapy for the targeted treatment of rheumatoid arthritis. Colloids Surf B Biointerfaces 2024; 234:113707. [PMID: 38181689 DOI: 10.1016/j.colsurfb.2023.113707] [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: 09/09/2023] [Revised: 11/20/2023] [Accepted: 12/12/2023] [Indexed: 01/07/2024]
Abstract
Activated M1-type macrophages, which produce inflammatory factors that exacerbate rheumatoid arthritis (RA), represent crucial target cells for inhibiting the disease process. In this study, we developed a novel photoresponsive targeted drug delivery system (TPNPs-HA) that can effectively deliver the hypoxia-activated prodrug tirapazamine (TPZ) specifically to activated macrophages. After administration, this metal-organic framework, PCN-224, constructed uing the photosensitizer porphyrin, exhibits the ability to generate excessive toxic reactive oxygen species (ROS) when exposed to near-infrared light. Additionally, the oxygen-consumed hypoxic environment further activates the chemotherapeutic effect of TPZ, thus creating a synergistic combination of photodynamic therapy (PDT) and hypoxia-activated chemotherapy (HaCT) to promote the elimination of activated M1-type macrophages. The results highlight the significantly potential of this photoresponsive nano-delivery system in providing substantial relief for RA. Furthermore, these findings support its effectiveness in inhibiting the disease process of RA, thereby offering new possibilities for the development of precise and accurate strategies for RA.
Collapse
Affiliation(s)
- Shixin Zhang
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun 130022, China; School of Life Sciences, Jilin University, Changchun 130012, China
| | - Miaomiao Zhang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jingbo Zhang
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Ge Li
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xinyue Lu
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Fengying Sun
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wenhua Liu
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun 130022, China.
| |
Collapse
|
19
|
Tehrani Nejad S, Rahimi R, Najafi M, Rostamnia S. Sustainable Gold Nanoparticle (Au-NP) Growth within Interspaces of Porphyrinic Zirconium-Based Metal-Organic Frameworks: Green Synthesis of PCN-224/Au-NPs and Its Anticancer Effect on Colorectal Cancer Cells Assay. ACS APPLIED MATERIALS & INTERFACES 2024; 16:3162-3170. [PMID: 38194287 DOI: 10.1021/acsami.3c15398] [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: 01/10/2024]
Abstract
In this work, a simple green synthesis method of the novel metal-organic framework (MOF) nanocomposite PCN-224/Au-NPs (Au-NPs = gold nanoparticles) is described. In this regard, initially, PCN-224 was synthesized. Afterward, in a single-step, one-pot procedure, under visible-light irradiation, Au-NPs were fabricated on PCN-224. The cytotoxicity effect of the synthesized PCN-224/Au-NPs nanocomposite was investigated in human colon cancer cells. Determination of the apoptosis induction was done by the Annexin- V/propidium iodide flow cytometry method. Besides, to ascertain the biocompatibility of the synthesized sample, the cytotoxicity of PCN-224/Au-NPs was evaluated on the human embryonic kidney (HEK)-293 cell line. The substantial anticancer activity with the biocompatibility of the structure, the green facile synthesis, and the MOF surface of the synthesized nanocomposite make it special for utilization in therapeutic applications.
Collapse
Affiliation(s)
- Sajedeh Tehrani Nejad
- Department of Chemistry, Iran University of Science and Technology, Tehran 16846-13114, Iran
| | - Rahmatollah Rahimi
- Department of Chemistry, Iran University of Science and Technology, Tehran 16846-13114, Iran
| | - Mina Najafi
- Department of Chemistry, Iran University of Science and Technology, Tehran 16846-13114, Iran
| | - Sadegh Rostamnia
- Department of Chemistry, Iran University of Science and Technology, Tehran 16846-13114, Iran
| |
Collapse
|
20
|
Yang M, Zhang Y, Hu Z, Xie H, Tian W, Liu Z. Application of hyaluronic acid-based nanoparticles for cancer combination therapy. Int J Pharm 2023; 646:123459. [PMID: 37778513 DOI: 10.1016/j.ijpharm.2023.123459] [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: 05/07/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/03/2023]
Abstract
Cancer is a significant public health problem in the world. The treatment methods include surgery, chemotherapy, phototherapy, and immunotherapy. Due to their respective limitations, the treatment effect is often unsatisfactory, laying hidden dangers for metastasis and recurrence. Since their exceptional biocompatibility and excellent targeting capabilities, hyaluronic acid-based biomaterials have generated great interest as drug delivery methods for tumor therapy. Moreover, modified HA can self-assemble into hydrogels or nanoparticles (NPs) for precise drug administration. This article summarizes the application of HA-based NPs in combination therapy. Ultimately, it is anticipated that this research will offer guidance for creating various HA-based NPs utilized in numerous cancer therapies.
Collapse
Affiliation(s)
- Mengru Yang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Ying Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Zheming Hu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Haonan Xie
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Wenli Tian
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Zhidong Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
21
|
Feng C, Chen B, Fan R, Zou B, Han B, Guo G. Polyphenol-Based Nanosystems for Next-Generation Cancer Therapy: Multifunctionality, Design, and Challenges. Macromol Biosci 2023; 23:e2300167. [PMID: 37266916 DOI: 10.1002/mabi.202300167] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/15/2023] [Indexed: 06/03/2023]
Abstract
With the continuous updating of cancer treatment methods and the rapid development of precision medicine in recent years, there are higher demands for advanced and versatile drug delivery systems. Scientists are committed to create greener and more effective nanomedicines where the carrier is no longer limited to a single function of drug delivery. Polyphenols, which can act as both active ingredients and fundamental building blocks, are being explored as potential multifunctional carriers that are efficient and safe for design purposes. Due to their intrinsic anticancer activity, phenolic compounds have shown surprising expressiveness in ablation of tumor cells, overcoming cancer multidrug resistance (MDR), and enhancing immunotherapeutic efficacy. This review provides an overview of recent advances in the design, synthesis, and application of versatile polyphenol-based nanosystems for cancer therapy in various modes. Moreover, the merits of polyphenols and the challenges for their clinical translation are also discussed, and it is pointed out that the novel polyphenol delivery system requires further optimization and validation.
Collapse
Affiliation(s)
- Chenqian Feng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bo Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rangrang Fan
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bingwen Zou
- Department of Thoracic Oncology and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bo Han
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, 832002, China
| | - Gang Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
22
|
Liu K, Yao Y, Xue S, Zhang M, Li D, Xu T, Zhi F, Liu Y, Ding D. Recent Advances of Tumor Microenvironment-Responsive Nanomedicines-Energized Combined Phototherapy of Cancers. Pharmaceutics 2023; 15:2480. [PMID: 37896240 PMCID: PMC10610502 DOI: 10.3390/pharmaceutics15102480] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/02/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023] Open
Abstract
Photodynamic therapy (PDT) has emerged as a powerful tumor treatment tool due to its advantages including minimal invasiveness, high selectivity and thus dampened side effects. On the other side, the efficacy of PDT is severely frustrated by the limited oxygen level in tumors, thus promoting its combination with other therapies, particularly photothermal therapy (PTT) for bolstered tumor treatment outcomes. Meanwhile, nanomedicines that could respond to various stimuli in the tumor microenvironment (TME) provide tremendous benefits for combined phototherapy with efficient hypoxia relief, tailorable drug release and activation, improved cellular uptake and intratumoral penetration of nanocarriers, etc. In this review, we will introduce the merits of combining PTT with PDT, summarize the recent important progress of combined phototherapies and their combinations with the dominant tumor treatment regimen, chemotherapy based on smart nanomedicines sensitive to various TME stimuli with a focus on their sophisticated designs, and discuss the challenges and future developments of nanomedicine-mediated combined phototherapies.
Collapse
Affiliation(s)
- Kehan Liu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (K.L.); (S.X.); (M.Z.); (T.X.)
| | - Yao Yao
- Department of Gerontology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian 223800, China;
| | - Shujuan Xue
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (K.L.); (S.X.); (M.Z.); (T.X.)
| | - Mengyao Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (K.L.); (S.X.); (M.Z.); (T.X.)
| | - Dazhao Li
- Department of Neurosurgery, The First People’s Hospital of Changzhou, Changzhou 213003, China; (D.L.); (F.Z.)
- Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Tao Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (K.L.); (S.X.); (M.Z.); (T.X.)
- School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), D02 NY74 Dublin, Ireland
| | - Feng Zhi
- Department of Neurosurgery, The First People’s Hospital of Changzhou, Changzhou 213003, China; (D.L.); (F.Z.)
- Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Yang Liu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (K.L.); (S.X.); (M.Z.); (T.X.)
| | - Dawei Ding
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (K.L.); (S.X.); (M.Z.); (T.X.)
| |
Collapse
|
23
|
Huang Z, Xian T, Meng X, Hu H, Gao L, Huang J, Yang D, Ou K, Wang B, Zhang Y. Multifunctional Novel Nanoplatform for Effective Synergistic Chemo-Photodynamic Therapy of Breast Cancer by Enhancing DNA Damage and Disruptions of Its Reparation. Molecules 2023; 28:6972. [PMID: 37836815 PMCID: PMC10574765 DOI: 10.3390/molecules28196972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023] Open
Abstract
Photodynamic therapy (PDT) is an effective noninvasive therapeutic strategy that has been widely used for anti-tumor therapy by the generation of excessive highly cytotoxic ROS. However, the poor water solubility of the photosensitizer, reactive oxygen species (ROS) depleting by high concentrations of glutathione (GSH) in the tumor microenvironment and the activation of DNA repair pathways to combat the oxidative damage, will significantly limit the therapeutic effect of PDT. Herein, we developed a photosensitizer prodrug (CSP) by conjugating the photosensitizer pyropheophorbide a (PPa) and the DNA-damaging agent Chlorambucil (Cb) with a GSH-responsive disulfide linkage and demonstrated a multifunctional co-delivery nanoplatform (CSP/Ola nanoparticles (NPs)) together with DSPE-PEG2000 and PARP inhibitor Olaparib (Ola). The CSP/Ola NPs features excellent physiological stability, efficient loading capacity, much better cellular uptake behavior and photodynamic performance. Specifically, the nanoplatform could induce elevated intracellular ROS levels upon the in situ generation of ROS during PDT, and decrease ROS consumption by reducing intracellular GSH level. Moreover, the CSP/Ola NPs could amplify DNA damage by released Cb and inhibit the activation of Poly(ADP-ribose) polymerase (PARP), promote the upregulation of γ-H2AX, thereby blocking the DNA repair pathway to sensitize tumor cells for PDT. In vitro investigations revealed that CSP/Ola NPs showed excellent phototoxicity and the IC50 values of CSP/Ola NPs against MDA-MB-231 breast cancer cells were as low as 0.05-01 μM after PDT. As a consequence, the co-delivery nanoplatform greatly promotes the tumor cell apoptosis and shows a high antitumor performance with combinational chemotherapy and PDT. Overall, this work provides a potential alternative to improve the therapeutic efficiency of triple negative breast cancer cell (TNBC) treatment by synergistically enhancing DNA damage and disrupting DNA damage repair.
Collapse
Affiliation(s)
- Zheng Huang
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing Collaborative Innovation Center of Targeted and Innovative Therapeutics, College of Pharmacy, Chongqing University of Arts and Sciences, Chongqing 402160, China; (Z.H.); (T.X.); (X.M.); (H.H.); (L.G.); (J.H.); (D.Y.); (K.O.)
- Key Laboratory of Bio-Theological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400045, China;
| | - Tong Xian
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing Collaborative Innovation Center of Targeted and Innovative Therapeutics, College of Pharmacy, Chongqing University of Arts and Sciences, Chongqing 402160, China; (Z.H.); (T.X.); (X.M.); (H.H.); (L.G.); (J.H.); (D.Y.); (K.O.)
| | - Xiangyi Meng
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing Collaborative Innovation Center of Targeted and Innovative Therapeutics, College of Pharmacy, Chongqing University of Arts and Sciences, Chongqing 402160, China; (Z.H.); (T.X.); (X.M.); (H.H.); (L.G.); (J.H.); (D.Y.); (K.O.)
| | - Huaisong Hu
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing Collaborative Innovation Center of Targeted and Innovative Therapeutics, College of Pharmacy, Chongqing University of Arts and Sciences, Chongqing 402160, China; (Z.H.); (T.X.); (X.M.); (H.H.); (L.G.); (J.H.); (D.Y.); (K.O.)
| | - Lixia Gao
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing Collaborative Innovation Center of Targeted and Innovative Therapeutics, College of Pharmacy, Chongqing University of Arts and Sciences, Chongqing 402160, China; (Z.H.); (T.X.); (X.M.); (H.H.); (L.G.); (J.H.); (D.Y.); (K.O.)
| | - Jiuhong Huang
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing Collaborative Innovation Center of Targeted and Innovative Therapeutics, College of Pharmacy, Chongqing University of Arts and Sciences, Chongqing 402160, China; (Z.H.); (T.X.); (X.M.); (H.H.); (L.G.); (J.H.); (D.Y.); (K.O.)
| | - Donglin Yang
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing Collaborative Innovation Center of Targeted and Innovative Therapeutics, College of Pharmacy, Chongqing University of Arts and Sciences, Chongqing 402160, China; (Z.H.); (T.X.); (X.M.); (H.H.); (L.G.); (J.H.); (D.Y.); (K.O.)
| | - Kepeng Ou
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing Collaborative Innovation Center of Targeted and Innovative Therapeutics, College of Pharmacy, Chongqing University of Arts and Sciences, Chongqing 402160, China; (Z.H.); (T.X.); (X.M.); (H.H.); (L.G.); (J.H.); (D.Y.); (K.O.)
| | - Bochu Wang
- Key Laboratory of Bio-Theological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400045, China;
| | - Yimei Zhang
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing Collaborative Innovation Center of Targeted and Innovative Therapeutics, College of Pharmacy, Chongqing University of Arts and Sciences, Chongqing 402160, China; (Z.H.); (T.X.); (X.M.); (H.H.); (L.G.); (J.H.); (D.Y.); (K.O.)
| |
Collapse
|
24
|
Guo S, Gu D, Yang Y, Tian J, Chen X. Near-infrared photodynamic and photothermal co-therapy based on organic small molecular dyes. J Nanobiotechnology 2023; 21:348. [PMID: 37759287 PMCID: PMC10523653 DOI: 10.1186/s12951-023-02111-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Near-infrared (NIR) organic small molecule dyes (OSMDs) are effective photothermal agents for photothermal therapy (PTT) due to their advantages of low cost and toxicity, good biodegradation, and strong NIR absorption over a wide wavelength range. Nevertheless, OSMDs have limited applicability in PTT due to their low photothermal conversion efficiency and inadequate destruction of tumor regions that are nonirradiated by NIR light. However, they can also act as photosensitizers (PSs) to produce reactive oxygen species (ROS), which can be further eradicated by using ROS-related therapies to address the above limitations of PTT. In this review, the synergistic mechanism, composition, and properties of photodynamic therapy (PDT)-PTT nanoplatforms were comprehensively discussed. In addition, some specific strategies for further improving the combined PTT and PDT based on OSMDs for cancer to completely eradicate cancer cells were outlined. These strategies include performing image-guided co-therapy, enhancing tumor infiltration, increasing H2O2 or O2 in the tumor microenvironment, and loading anticancer drugs onto nanoplatforms to enable combined therapy with phototherapy and chemotherapy. Meanwhile, the intriguing prospects and challenges of this treatment modality were also summarized with a focus on the future trends of its clinical application.
Collapse
Affiliation(s)
- Shuang Guo
- School of Light Industry and Chemical Engineering, Dalian Polytechnic University, Dalian, 116034, China
| | - Dongyu Gu
- College of Marine Science and Environment, Dalian Ocean University, Dalian, 116023, China
| | - Yi Yang
- School of Light Industry and Chemical Engineering, Dalian Polytechnic University, Dalian, 116034, China.
| | - Jing Tian
- School of Biological Engineering, Dalian Polytechnic University, Dalian, 116034, China.
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine, Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore.
| |
Collapse
|
25
|
Mahmut Z, Zhang C, Ruan F, Shi N, Zhang X, Wang Y, Zheng X, Tang Z, Dong B, Gao D, Sun J. Medical Applications and Advancement of Near Infrared Photosensitive Indocyanine Green Molecules. Molecules 2023; 28:6085. [PMID: 37630337 PMCID: PMC10459369 DOI: 10.3390/molecules28166085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/30/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Indocyanine green (ICG) is an important kind of near infrared (NIR) photosensitive molecules for PTT/PDT therapy as well as imaging. When exposed to NIR light, ICG can produce reactive oxygen species (ROS), which can kill cancer cells and pathogenic bacteria. Moreover, the absorbed light can also be converted into heat by ICG molecules to eliminate cancer cells. In addition, it performs exceptionally well in optical imaging-guided tumor therapy and antimicrobial therapy due to its deeper tissue penetration and low photobleaching properties in the near-infrared region compared to other dyes. In order to solve the problems of water and optical stability and multi-function problem of ICG molecules, composite nanomaterials based on ICG have been designed and widely used, especially in the fields of tumors and sterilization. So far, ICG molecules and their composite materials have become one of the most famous infrared sensitive materials. However, there have been no corresponding review articles focused on ICG molecules. In this review, the molecular structure and properties of ICG, composite material design, and near-infrared light- triggered anti-tumor, and antibacterial, and clinical applications are reviewed in detail, which of great significance for related research.
Collapse
Affiliation(s)
- Zulpya Mahmut
- Department of Cell Biology and Medical Genetics, College of Basic Medical Science, Jilin University, Changchun 130021, China; (Z.M.); (C.Z.); (X.Z.); (Y.W.); (X.Z.)
| | - Chunmei Zhang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Science, Jilin University, Changchun 130021, China; (Z.M.); (C.Z.); (X.Z.); (Y.W.); (X.Z.)
| | - Fei Ruan
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130012, China; (F.R.); (Z.T.)
| | - Nan Shi
- Department of Respiratory Medicine, No. 964 Hospital of People’s Liberation Army, 4799 Xi’an Road, Changchun 130062, China;
| | - Xinyao Zhang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Science, Jilin University, Changchun 130021, China; (Z.M.); (C.Z.); (X.Z.); (Y.W.); (X.Z.)
| | - Yuda Wang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Science, Jilin University, Changchun 130021, China; (Z.M.); (C.Z.); (X.Z.); (Y.W.); (X.Z.)
| | - Xianhong Zheng
- Department of Cell Biology and Medical Genetics, College of Basic Medical Science, Jilin University, Changchun 130021, China; (Z.M.); (C.Z.); (X.Z.); (Y.W.); (X.Z.)
| | - Zixin Tang
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130012, China; (F.R.); (Z.T.)
| | - Biao Dong
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130012, China; (F.R.); (Z.T.)
| | - Donghui Gao
- Department of Anesthesiology and Operating Room, School and Hospital of Stomatology, Jilin University, Changchun 130012, China
| | - Jiao Sun
- Department of Cell Biology and Medical Genetics, College of Basic Medical Science, Jilin University, Changchun 130021, China; (Z.M.); (C.Z.); (X.Z.); (Y.W.); (X.Z.)
| |
Collapse
|
26
|
Huang J, Liao D, Han Y, Chen Y, Raza S, Lu C, Liu J, Lan Q. Current status of porous coordination networks (PCNs) derived porphyrin spacers for cancer therapy. Expert Opin Drug Deliv 2023; 20:1209-1229. [PMID: 37776531 DOI: 10.1080/17425247.2023.2260309] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/14/2023] [Indexed: 10/02/2023]
Abstract
INTRODUCTION Porous coordination networks (PCNs) have been widely used in large number of applications such as light harvesting, catalysis, and biomedical applications. Inserting porphyrins into PCNs scaffolds can alleviate the solubility and chemical stability problems associated with porphyrin ligands and add functionality to PCNs. The discovery that some PCNs materials have photosensitizer and acoustic sensitizer properties has attracted significant attention in the field of biomedicine, particularly in cancer therapy. This article describes the latest applications of the porphyrin ligand-based family of PCNs in cancer chemodynamic therapy (CDT), photodynamic therapy (PDT), sonodynamic therapy (SDT), photothermal therapy (PTT), and combination therapies and offers some observations and reflections on them. AREAS COVERED This article discusses the use of the PCN family of MOFs in cancer treatment, specifically focusing on chemodynamic therapy, sonodynamic therapy, photodynamic therapy, photothermal therapy, and combination therapy. EXPERT OPINION Although a large number of PCNs have been developed for use in novel cancer therapeutic approaches, further improvements are needed to advance the use of PCNs in the clinic. For example, the main mechanism of action of PCNs against cancer and the metabolic processes in organisms, and how to construct PCNs that maintain good stability in the complex environment of organisms.
Collapse
Affiliation(s)
- Jeifeng Huang
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Donghui Liao
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Yuting Han
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Ying Chen
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Saleem Raza
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, P.R. China
| | - Chengyu Lu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Jianqiang Liu
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, China
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Qian Lan
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| |
Collapse
|
27
|
Jiang X, Zhao Y, Sun S, Xiang Y, Yan J, Wang J, Pei R. Research development of porphyrin-based metal-organic frameworks: targeting modalities and cancer therapeutic applications. J Mater Chem B 2023. [PMID: 37305964 DOI: 10.1039/d3tb00632h] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Porphyrins are naturally occurring organic molecules that have attracted widespread attention for their potential in the field of biomedical research. Porphyrin-based metal-organic frameworks (MOFs) that utilize porphyrin molecules as organic ligands have gained attention from researchers due to their excellent results as photosensitizers in tumor photodynamic therapy (PDT). Additionally, MOFs hold significant promise and potential for other tumor therapeutic approaches due to their tunable size and pore size, excellent porosity, and ultra-high specific surface area. Active delivery of nanomaterials via targeted molecules for tumor therapy has demonstrated greater accumulation, lower drug doses, higher therapeutic efficacy, and reduced side effects relative to passive targeting through the enhanced permeation and retention effect (EPR). This paper presents a comprehensive review of the targeting methods employed by porphyrin-based MOFs in tumor targeting therapy over the past few years. It further discusses the applications of porphyrin-based MOFs for targeted cancer therapy through various therapeutic methods. The objective of this paper is to provide a valuable reference and source of ideas for targeted therapy using porphyrin-based MOF materials and to inspire further exploration of their potential in the field of cancer therapy.
Collapse
Affiliation(s)
- Xiang Jiang
- College of Mechanics and Materials, Hohai University, Nanjing, 210098, China.
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Yuewu Zhao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Shengkai Sun
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Ying Xiang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Jincong Yan
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Jine Wang
- College of Mechanics and Materials, Hohai University, Nanjing, 210098, China.
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
- Jiangxi Institute of Nanotechnology, Nanchang, 330200, China
| | - Renjun Pei
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| |
Collapse
|
28
|
Yu S, Xu K, Wang Z, Zhang Z, Zhang Z. Bibliometric and visualized analysis of metal-organic frameworks in biomedical application. Front Bioeng Biotechnol 2023; 11:1190654. [PMID: 37234479 PMCID: PMC10206306 DOI: 10.3389/fbioe.2023.1190654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Background: Metal-organic frameworks (MOFs) are hybrid materials composed of metal ions or clusters and organic ligands that spontaneously assemble via coordination bonds to create intramolecular pores, which have recently been widely used in biomedicine due to their porosity, structural, and functional diversity. They are used in biomedical applications, including biosensing, drug delivery, bioimaging, and antimicrobial activities. Our study aims to provide scholars with a comprehensive overview of the research situations, trends, and hotspots in biomedical applications of MOFs through a bibliometric analysis of publications from 2002 to 2022. Methods: On 19 January 2023, the Web of Science Core Collection was searched to review and analyze MOFs applications in the biomedical field. A total of 3,408 studies published between 2002 and 2022 were retrieved and examined, with information such as publication year, country/region, institution, author, journal, references, and keywords. Research hotspots were extracted and analyzed using the Bibliometrix R-package, VOSviewer, and CiteSpace. Results: We showed that researchers from 72 countries published articles on MOFs in biomedical applications, with China producing the most publications. The Chinese Academy of Science was the most prolific contributor to these publications among 2,209 institutions that made contributions. Reference co-citation analysis classifies references into 8 clusters: synergistic cancer therapy, efficient photodynamic therapy, metal-organic framework encapsulation, selective fluorescence, luminescent probes, drug delivery, enhanced photodynamic therapy, and metal-organic framework-based nanozymes. Keyword co-occurrence analysis divided keywords into 6 clusters: biosensors, photodynamic therapy, drug delivery, cancer therapy and bioimaging, nanoparticles, and antibacterial applications. Research frontier keywords were represented by chemodynamic therapy (2020-2022) and hydrogen peroxide (2020-2022). Conclusion: Using bibliometric methods and manual review, this review provides a systematic overview of research on MOFs in biomedical applications, filling an existing gap. The burst keyword analysis revealed that chemodynamic therapy and hydrogen peroxide are the prominent research frontiers and hot spots. MOFs can catalyze Fenton or Fenton-like reactions to generate hydroxyl radicals, making them promising materials for chemodynamic therapy. MOF-based biosensors can detect hydrogen peroxide in various biological samples for diagnosing diseases. MOFs have a wide range of research prospects for biomedical applications.
Collapse
Affiliation(s)
- Sanyang Yu
- The VIP Department, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Kaihao Xu
- The VIP Department, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Zhenhua Wang
- Department of Physiology, School of Life Sciences, China Medical University, Shenyang, China
| | - Zhichang Zhang
- Department of Computer, School of Intelligent Medicine, China Medical University, Shenyang, China
| | - Zhongti Zhang
- The VIP Department, School and Hospital of Stomatology, China Medical University, Shenyang, China
| |
Collapse
|
29
|
Li J, Wang S, Fontana F, Tapeinos C, Shahbazi MA, Han H, Santos HA. Nanoparticles-based phototherapy systems for cancer treatment: Current status and clinical potential. Bioact Mater 2023; 23:471-507. [PMID: 36514388 PMCID: PMC9727595 DOI: 10.1016/j.bioactmat.2022.11.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 11/16/2022] [Accepted: 11/20/2022] [Indexed: 12/11/2022] Open
Abstract
Remarkable progress in phototherapy has been made in recent decades, due to its non-invasiveness and instant therapeutic efficacy. In addition, with the rapid development of nanoscience and nanotechnology, phototherapy systems based on nanoparticles or nanocomposites also evolved as an emerging hotspot in nanomedicine research, especially in cancer. In this review, first we briefly introduce the history of phototherapy, and the mechanisms of phototherapy in cancer treatment. Then, we summarize the representative development over the past three to five years in nanoparticle-based phototherapy and highlight the design of the innovative nanoparticles thereof. Finally, we discuss the feasibility and the potential of the nanoparticle-based phototherapy systems in clinical anticancer therapeutic applications, aiming to predict future research directions in this field. Our review is a tutorial work, aiming at providing useful insights to researchers in the field of nanotechnology, nanoscience and cancer.
Collapse
Affiliation(s)
- Jiachen Li
- Department of Biomedical Engineering, W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, the Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Shiqi Wang
- Drug Research Program Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Flavia Fontana
- Drug Research Program Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Christos Tapeinos
- Drug Research Program Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, the Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Huijie Han
- Department of Biomedical Engineering, W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, the Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Hélder A Santos
- Department of Biomedical Engineering, W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, the Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
- Drug Research Program Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
30
|
Tian J, Wang J, Xu H, Zou B, Chen W, Liu Y, Chen J, Zhang R. Nanoscale metal-organic framework delivers rapamycin to induce tissue immunogenic cell death and potentiates cancer immunotherapy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 50:102678. [PMID: 37044194 DOI: 10.1016/j.nano.2023.102678] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/21/2023] [Accepted: 04/02/2023] [Indexed: 04/14/2023]
Abstract
Rapamycin has great potential in the antitumor application, but its therapeutic effect is seriously affected by poor water solubility, targeting ability, and low bioavailability. Here, we constructed a novel composite nanomaterial with PCN-224 as a drug carrier and loaded rapamycin, named R@BP@HA. The nanoplate not only improves targeting, but also synergizes rapamycin with PCN-224 to effectively promote tumor cell apoptosis, which subsequently causes immunogenic cell death (ICD), and shows strong therapeutic effect in 4T1 breast cancer model. The treatment effect depends on three main points:(i)Proapoptotic effect of rapamycin on tumor cells;(ii)ROS production by PCN-224-mediated photodynamic therapy;(iii)ICD induced DC maturation, increased immune response and promoted T cell proliferation and differentiation. This nanoplate offers potential antitumor efficacy in combination with chemotherapy, photodynamic therapy, and immunotherapy.
Collapse
Affiliation(s)
- Jihua Tian
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan 030001, China.
| | - Jing Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan 030001, China
| | - Huanyu Xu
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan 030001, China
| | - Bocheng Zou
- Department of The Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Weihao Chen
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan 030001, China
| | - Yulong Liu
- Department of The Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Jingshu Chen
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan 030001, China
| | - Ruiping Zhang
- Department of The Radiology Department of First Hospital of Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
31
|
Yang C, Wang K, Tian S, Mo L, Lin W. Functionalized photosensitive metal-organic framework as a theranostic nanoplatform for turn-on detection of MicroRNA and photodynamic therapy. Anal Chim Acta 2023; 1239:340689. [PMID: 36628708 DOI: 10.1016/j.aca.2022.340689] [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: 09/20/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022]
Abstract
Developing a theranostic platform integrating precise diagnostic and efficient treatment is significant but challenging. Here, we reported a new theranostic platform - hairpin probe - photosensitizing MOFs (HPMOF) composed of photosensitizing MOFs (PMOFs) and hairpin probes labeled with fluorophore and quencher, in which PMOF played the role of photosensitizer and nanocarrier of the hairpin probe. The HPMOF was covered with a layer of ZIF-8 to achieve the dual-layered nanotheranostics (HPMOF@ZIF-8). The HPMOF@ZIF-8 achieved high DNA loading capacity and intracellular delivery for tumor-related miRNA imaging. Moreover, HPMOF@ZIF-8 could generate reactive oxygen species with high efficiency, which induced cell apoptosis, leading to efficient photodynamic therapy. Due to the different expression of miRNA between normal cells and cancer cells, the HPMOF@ZIF-8 could recognize cancer cells through imaging of miRNA, leading to more accurate treatment of cancer, providing a promising theranostic nanoplatform.
Collapse
Affiliation(s)
- Chan Yang
- Guangxi Key Laboratory of Electrochemical Energy Materials, Institute of Optical Materials and Chemical Biology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Kun Wang
- Guangxi Key Laboratory of Electrochemical Energy Materials, Institute of Optical Materials and Chemical Biology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Shuo Tian
- Guangxi Key Laboratory of Electrochemical Energy Materials, Institute of Optical Materials and Chemical Biology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Liuting Mo
- Guangxi Key Laboratory of Electrochemical Energy Materials, Institute of Optical Materials and Chemical Biology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Weiying Lin
- Guangxi Key Laboratory of Electrochemical Energy Materials, Institute of Optical Materials and Chemical Biology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China.
| |
Collapse
|
32
|
Alavi N, Maghami P, Pakdel AF, Rezaei M, Avan A. Antibody-modified Gold Nanobiostructures: Advancing Targeted Photodynamic Therapy for Improved Cancer Treatment. Curr Pharm Des 2023; 29:3103-3122. [PMID: 37990429 DOI: 10.2174/0113816128265544231102065515] [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: 06/10/2023] [Accepted: 10/03/2023] [Indexed: 11/23/2023]
Abstract
Photodynamic therapy (PDT) is an innovative, non-invasive method of treating cancer that uses light-activated photosensitizers to create reactive oxygen species (ROS). However, challenges associated with the limited penetration depth of light and the need for precise control over photosensitizer activation have hindered its clinical translation. Nanomedicine, particularly gold nanobiostructures, offers promising solutions to overcome these limitations. This paper reviews the advancements in PDT and nanomedicine, focusing on applying antibody-modified gold nanobiostructures as multifunctional platforms for enhanced PDT efficacy and improved cancer treatment outcomes. The size, shape, and composition of gold nanobiostructures can significantly influence their PDT efficacy, making synthetic procedures crucial. Functionalizing the surface of gold nanobiostructures with various molecules, such as antibodies or targeting agents, bonding agents, PDT agents, photothermal therapy (PTT) agents, chemo-agents, immunotherapy agents, and imaging agents, allows composition modification. Integrating gold nanobiostructures with PDT holds immense potential for targeted cancer therapy. Antibody-modified gold nanobiostructures, in particular, have gained significant attention due to their tunable plasmonic characteristics, biocompatibility, and surface functionalization capabilities. These multifunctional nanosystems possess unique properties that enhance the efficacy of PDT, including improved light absorption, targeted delivery, and enhanced ROS generation. Passive and active targeting of gold nanobiostructures can enhance their localization near cancer cells, leading to efficient eradication of tumor tissues upon light irradiation. Future research and clinical studies will continue to explore the potential of gold nanobiostructures in PDT for personalized and effective cancer therapy. The synthesis, functionalization, and characterization of gold nanobiostructures, their interaction with light, and their impact on photosensitizers' photophysical and photochemical properties, are important areas of investigation. Strategies to enhance targeting efficiency and the evaluation of gold nanobiostructures in vitro and in vivo studies will further advance their application in PDT. The integrating antibody-modified gold nanobiostructures in PDT represents a promising strategy for targeted cancer therapy. These multifunctional nanosystems possess unique properties that enhance PDT efficacy, including improved light absorption, targeted delivery, and enhanced ROS generation. Continued research and development in this field will contribute to the advancement of personalized and effective cancer treatment approaches.
Collapse
Affiliation(s)
- Negin Alavi
- Department of Biology, Islamic Azad University Science and Research Branch, Tehran, Iran
| | - Parvaneh Maghami
- Department of Biology, Islamic Azad University Science and Research Branch, Tehran, Iran
| | - Azar Fani Pakdel
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Rezaei
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane 4059, Australia
| |
Collapse
|
33
|
Xie Y, Wang M, Sun Q, Wang D, Li C. Recent Advances in Tetrakis (4‐Carboxyphenyl) Porphyrin‐Based Nanocomposites for Tumor Therapy. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Yulin Xie
- Institute of Molecular Sciences and Engineering Institute of Frontier and Interdisciplinary Science Shandong University Qingdao 266237 P.R. China
| | - Man Wang
- Institute of Molecular Sciences and Engineering Institute of Frontier and Interdisciplinary Science Shandong University Qingdao 266237 P.R. China
| | - Qianqian Sun
- Institute of Molecular Sciences and Engineering Institute of Frontier and Interdisciplinary Science Shandong University Qingdao 266237 P.R. China
| | - Dongmei Wang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials College of Chemistry and Life Sciences Zhejiang Normal University Jinhua 321004 P.R. China
| | - Chunxia Li
- Institute of Molecular Sciences and Engineering Institute of Frontier and Interdisciplinary Science Shandong University Qingdao 266237 P.R. China
| |
Collapse
|
34
|
Copper-olsalazine metal-organic frameworks as a nanocatalyst and epigenetic modulator for efficient inhibition of colorectal cancer growth and metastasis. Acta Biomater 2022; 152:495-506. [PMID: 36087871 DOI: 10.1016/j.actbio.2022.08.076] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/16/2022] [Accepted: 08/31/2022] [Indexed: 11/23/2022]
Abstract
Despite the extensive explorations of nanoscale metal-organic frameworks (nanoMOFs) in drug delivery, the intrinsic bioactivity of nanoMOFs, such as anticancer activity, is severely underestimated owing to the overlooked integration of the hierarchical components including nanosized MOFs and molecular-level organic ligands and metal-organic complexes. Herein, we propose a de novo design of multifunctional bioactive nanoMOFs ranging from molecular to nanoscale level, and demonstrate this proof-of-concept by a copper-olsalazine (Olsa, a clinically approved drug for inflammatory bowel disease, here as a bioactive linker and DNA hypomethylating agent) nanoMOF displaying a multifaceted anticancer mechanism: (1) Cu-Olsa nanoMOF-mediated redox dyshomeostasis for enhanced catalytic tumor therapy, (2) targeting downregulation of cyclooxygenase-2 by the organic complex of Cu2+ and Olsa, and (3) Olsa-mediated epigenetic regulation. Cu-Olsa nanoMOF displayed an enzyme-like catalytic activity to generate cancericidal species ·OH and 1O2 from rich H2O2 in tumors, improved the expression of tumor suppressors TIMP3 and AXIN2 by epigenetic modulation, and fulfilled selective inhibition of colorectal cancer cells over normal cells. The hyaluronic acid-modified nanoMOF further verified the efficient suppression of CT26 colorectal tumor growth and metastasis in murine models. Overall, these results suggest that Olsa-based MOF presents a platform of epigenetic therapy-synergized nanomedicine for efficient cancer treatment and provides a powerful strategy for the design of intrinsically bioactive nanoMOFs. STATEMENT OF SIGNIFICANCE: Metal-organic frameworks (MOFs) with intrinsic bioactivities such as anticancer and antibacterial activity are of great interest. Herein, we reported a bioactive copper-olsalazine (Cu-Olsa) nanoMOF as a nanodrug for colorectal cancer treatment. This nanoMOF per se displayed enzyme-like catalytic activity to generate cancericidal species ·OH and 1O2 from rich H2O2 in tumors for nanocatalytic tumor therapy. Upon dissociation into small molecular copper-organic complex and olsalazine in cancer cells, COX-2 inhibition and epigenetic modulation were fulfilled for selective inhibition of colorectal cancer growth and metastasis.
Collapse
|
35
|
Wang Y, Zhang Y, Zhang X, Zhang Z, She J, Wu D, Gao W. High Drug-Loading Nanomedicines for Tumor Chemo-Photo Combination Therapy: Advances and Perspectives. Pharmaceutics 2022; 14:pharmaceutics14081735. [PMID: 36015361 PMCID: PMC9415722 DOI: 10.3390/pharmaceutics14081735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 11/28/2022] Open
Abstract
The combination of phototherapy and chemotherapy (chemo−photo combination therapy) is an excellent attempt for tumor treatment. The key requirement of this technology is the high drug-loading nanomedicines, which can load either chemotherapy drugs or phototherapy agents at the same nanomedicines and simultaneously deliver them to tumors, and play a multimode therapeutic role for tumor treatment. These nanomedicines have high drug-loading efficiency (>30%) and good tumor combination therapeutic effect with important clinical application potential. Although there are many reports of high drug-loading nanomedicines for tumor therapy at present, systematic analyses on those nanomedicines remain lacking and a comprehensive review is urgently needed. In this review, we systematically analyze the current status of developed high drug-loading nanomedicines for tumor chemo−photo combination therapy and summarize their types, methods, drug-loading properties, in vitro and in vivo applications. The shortcomings of the existing high drug-loading nanomedicines for tumor chemo−photo combination therapy and the possible prospective development direction are also discussed. We hope to attract more attention for researchers in different academic fields, provide new insights into the research of tumor therapy and drug delivery system and develop these nanomedicines as the useful tool for tumor chemo−photo combination therapy in the future.
Collapse
Affiliation(s)
- Ya Wang
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an 710061, China
| | - Yujie Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an 710061, China
| | - Xiaojiang Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an 710061, China
| | - Zhe Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an 710061, China
| | - Junjun She
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an 710061, China
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an 710061, China
- Correspondence: (J.S.); (D.W.); (W.G.)
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Correspondence: (J.S.); (D.W.); (W.G.)
| | - Wei Gao
- Department of Anesthesiology & Center for Brain Science & Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- Correspondence: (J.S.); (D.W.); (W.G.)
| |
Collapse
|
36
|
Sun Y, Fang K, Hu X, Yang J, Jiang Z, Feng L, Li R, Rao Y, Shi S, Dong C. NIR-light-controlled G-quadruplex hydrogel for synergistically enhancing photodynamic therapy via sustained delivery of metformin and catalase-like activity in breast cancer. Mater Today Bio 2022; 16:100375. [PMID: 35983175 PMCID: PMC9379686 DOI: 10.1016/j.mtbio.2022.100375] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 07/16/2022] [Accepted: 07/19/2022] [Indexed: 12/27/2022] Open
Abstract
Severely hypoxic condition of tumour represents a notable obstacle against the efficiency of photodynamic therapy (PDT). While mitochondria targeted therapy by metformin has been considered as a promising strategy for reducing oxygen consumption in tumours, its low treatment sensitivity, short half-life and narrow absorption window in vivo remain the intractable challenges. In this report, 5′-guanosine monophosphate (5′GMP), indocyanine green (ICG), hemin and metformin, were combined to construct a smart G-quadruplex (G4) hydrogel named HMI@GEL for breast cancer (BC) treatment. Benefiting from the photothermal (PTT) effect of ICG, HMI@GEL exhibited excellent characteristics of NIR-light-triggered and persistent drug delivery to maintain high intratumoral concentration of metformin. Furthermore, drug loading concentration of metformin reached an amazing 300 mg mL−1 in HMI@GEL. To our knowledge, it might be the highest loading efficiency in the reported literatures. With the combination of catalase-mimicking Hemin@mil88, metformin could inhibit tumour mitochondrial respiratory significantly, which sequentially permitted in situ efficient oxygen generation. Remarkable apoptosis and necrosis were achieved by the combination of PTT and synergistically enhanced PDT as well as the activated tumour immunotherapy. Collectively, the HMI@GEL in situ injectable platform showed a promising strategy for enhanced PDT by metformin, and opened new perspectives for treating BC versatilely. A NIR-light-controlled G-quadruplex hydrogel HMI@GEL loading metformin was prepared for precision breast cancer therapy. The extremely high drug loading capacity (300 mg mL−1) and persistent delivery of metformin was realized for the first time. The combination of catalase-mimicking Hemin@mil88 and metformin dual enhanced intracellular ROS generation. The tumour immune microenvironment was dramatically reshaped by synthetic photodynamic/photothermal therapy.
Collapse
Affiliation(s)
- Yanting Sun
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Kang Fang
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Xiaochun Hu
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Jingxian Yang
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Zhengyang Jiang
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Lei Feng
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Ruihao Li
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Yiming Rao
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
| | - Shuo Shi
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
- Corresponding author.
| | - Chunyan Dong
- Department of Oncology, East Hospital Affiliated to Tongji University, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, PR China
- Corresponding author.
| |
Collapse
|
37
|
Cheng K, Guo Q, Shen Z, Yang W, Wang Y, Sun Z, Wu H. Bibliometric Analysis of Global Research on Cancer Photodynamic Therapy: Focus on Nano-Related Research. Front Pharmacol 2022; 13:927219. [PMID: 35784740 PMCID: PMC9243586 DOI: 10.3389/fphar.2022.927219] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 05/30/2022] [Indexed: 01/10/2023] Open
Abstract
A growing body of research has illuminated that photodynamic therapy (PDT) serves as an important therapeutic strategy in oncology and has become a hot topic in recent years. Although numerous papers related to cancer PDT (CPDT) have been published, no bibliometric studies have been conducted to summarize the research landscape, and highlight the research trends and hotspots in this field. This study collected 5,804 records on CPDT published between 2000 and 2021 from Web of Science Core Collection. Bibliometric analysis and visualization were conducted using VOSviewer, CiteSpace, and one online platform. The annual publication and citation results revealed significant increasing trends over the past 22 years. China and the United States, contributing 56.24% of the total publications, were the main driving force in this field. Chinese Academy of Sciences was the most prolific institution. Photodiagnosis and Photodynamic Therapy and Photochemistry and Photobiology were the most productive and most co-cited journals, respectively. All keywords were categorized into four clusters including studies on nanomaterial technology, clinical applications, mechanism, and photosensitizers. “nanotech-based PDT” and “enhanced PDT” were current research hotspots. In addition to several nano-related topics such as “nanosphere,” “nanoparticle,” “nanomaterial,” “nanoplatform,” “nanomedicine” and “gold nanoparticle,” the following topics including “photothermal therapy,” “metal organic framework,” “checkpoint blockade,” “tumor microenvironment,” “prodrug” also deserve further attention in the near future.
Collapse
Affiliation(s)
- Kunming Cheng
- Department of Intensive Care Unit, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiang Guo
- Department of Orthopaedics, Baodi Clinical College of Tianjin Medical University, Tianjin, China
| | - Zefeng Shen
- Department of Graduate School, Sun Yat-sen University, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Weiguang Yang
- Department of Graduate School of Tianjin Medical University, Tianjin, China
- Department of Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Yulin Wang
- Department of Graduate School of Tianjin Medical University, Tianjin, China
- Department of Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Zaijie Sun
- Department of Orthopaedic Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
- *Correspondence: Kunming Cheng, ; Zaijie Sun, ; Haiyang Wu,
| | - Haiyang Wu
- Department of Graduate School of Tianjin Medical University, Tianjin, China
- Department of Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- *Correspondence: Kunming Cheng, ; Zaijie Sun, ; Haiyang Wu,
| |
Collapse
|
38
|
Hu J, Hu J, Wu W, Qin Y, Fu J, Liu C, Seeberger PH, Yin J. Bimodal Treatment of Hepatocellular Carcinoma by Targeted Minimally Interventional Photodynamic/Chemotherapy Using Glyco-Covalent-Organic Frameworks-Guided Porphyrin/Sorafenib. Acta Biomater 2022; 148:206-217. [PMID: 35697198 DOI: 10.1016/j.actbio.2022.06.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/27/2022] [Accepted: 06/06/2022] [Indexed: 11/01/2022]
Abstract
Very limited treatment options are available to fight hepatocellular carcinoma (HCC), a serious global health concern with high morbidity and mortality. The integration of multiple therapies into one nanoplatform to exert synergistic therapeutic effects offers advantages over monotherapies. Here, we describe the construction of the nanoplatform Sor@GR-COF-366 for synergistic chemotherapy and photodynamic therapy (PDT) for HCC using a porphyrin-based covalent organic framework (COF-366) coated with N-acetyl-galactosamine (GalNAc) and rhodamine B (RhB), and loaded with the first-line agent, Sorafenib (Sor). The nanoplatform is targeted towards ASGPR-overexpressed HCC cells and liver tissues by GalNAc and observed by real-time imaging of RhB in vitro and in vivo. The nanoplatform Sor@GR-COF-366 exerts an enhanced synergistic tumor suppression effect in a subcutaneous HCC mouse model with a tumor inhibition rate (TGI) of 97% while significantly prolonging survival at very low toxicity. The potent synergistic therapeutic outcome is confirmed in an orthotopic mouse model of HCC with the TGI of 98% with a minimally invasive interventional PDT (IPDT). Sor@GR-COF-366 is a promising candidate to be combined with chemo-IPDT for the treatment of HCC. STATEMENT OF SIGNIFICANCE: This work describes the construction of covalent-organic frameworks (COFs) modified with glyco-moieties to serve as hepato-targeted multitherapy delivery systems. They combine minimally invasive interventional photodynamic therapy (IPDT) triggered synergism with chemotherapy treatment for hepatocellular carcinoma (HCC). With the aid of minimally invasive intervention, PDT can elicit potent anti-cancer activity for deep solid tumors. This platform shows strong therapeutic outcomes in both subcutaneous and orthotopic mouse models, which can significantly prolong survival. This work showed an effective combination of a biomedical nano-formulation with the clinical operational means in cancer treatment, which is greatly promising in clinical translation.
Collapse
Affiliation(s)
- Jun Hu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Jing Hu
- Wuxi School of Medicine, Jiangnan University, Lihu Avenue1800, Wuxi 214122, China
| | - Wenrui Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Department of Biliary-Pancreat Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yufei Qin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Department of Biliary-Pancreat Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Junjie Fu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Chao Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Department of Biliary-Pancreat Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Peter H Seeberger
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Potsdam 14476, Germany
| | - Jian Yin
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
39
|
Li R, Hu X, Shang F, Wu W, Zhang H, Wang Y, Pan J, Shi S, Dong C. Treatment of triple negative breast cancer by near infrared light triggered mild temperature photothermal therapy combined with oxygen-independent cytotoxic free radicals. Acta Biomater 2022; 148:218-229. [DOI: 10.1016/j.actbio.2022.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/28/2022] [Accepted: 06/06/2022] [Indexed: 11/01/2022]
|
40
|
Fan X, Luo Z, Chen Y, Yeo JCC, Li Z, Wu YL, He C. Oxygen self-supplied enzyme nanogels for tumor targeting with amplified synergistic starvation and photodynamic therapy. Acta Biomater 2022; 142:274-283. [PMID: 35114372 DOI: 10.1016/j.actbio.2022.01.056] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 02/06/2023]
Abstract
Tumor tissues need vast supply of nutrients and energy to sustain the rapid proliferation of cancer cells. Cutting off the glucose supply represents a promising cancer therapy approach. Herein, a tumor tissue-targeted enzyme nanogel (rGCP nanogel) with self-supply oxygen capability was developed. The enzyme nanogel synergistically enhanced starvation therapy and photodynamic therapy (PDT) to mitigate the rapid proliferation of cancer cells. The rGCP nanogel was fabricated by copolymerizing two monomers, porphyrin and cancer cells-targeted, Arg-Gly-Asp (RGD), onto the glucose oxidase (GOX) and catalase (CAT) surfaces. The cascade reaction within the rGCP nanogel could efficiently consume intracellular glucose catalyzed by GOX. Concurrently, CAT safely decomposed the produced H2O2 with systemic toxicity to promote oxygen generation and achieved low toxicity starvation therapy. The produced oxygen subsequently facilitated the glucose oxidation reaction and significantly enhanced the generation of cytotoxic singlet oxygen (1O2) in the presence of 660 nm light irradiation. Combining starvation therapy and PDT, the designed enzyme nanogel system presented an amplified synergic cancer therapy effect. This approach potentially paved a new way to fabricate a combinatorial therapy approach by employing cascaded catalytic nanomedicines with good tumor selectivity and efficient anti-cancer effect. STATEMENT OF SIGNIFICANCE: The performance of starvation and photodynamic therapy (PDT) is usually suppressed by intrinsic tumorous hypoxia. Herein, an oxygen self-supplied and tumor tissue-targeted enzyme nanogel was created by copolymerization of two monomers, porphyrin and cancer cell-targeted Arg-Gly-Asp (RGD), onto the surface of glucose oxidase (GOX) and catalase (CAT), which synergistically enhanced starvation therapy and PDT. Moreover, the enzyme nanogels possessed high stability and could be synthesized straightforwardly. This anti-cancer system provides an approach for constructing a combinatorial therapy approach by employing cascaded catalytic nanomedicine with good tumor selectivity and therapeutic efficacy.
Collapse
Affiliation(s)
- Xiaotong Fan
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Zheng Luo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Ying Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Jayven Chee Chuan Yeo
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore; Institute of Materials Research and Engineering, Agency for Science, Technology, and Research (A*STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Singapore
| | - Zibiao Li
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore; Institute of Materials Research and Engineering, Agency for Science, Technology, and Research (A*STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Singapore.
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.
| | - Chaobin He
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore; Institute of Materials Research and Engineering, Agency for Science, Technology, and Research (A*STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Singapore.
| |
Collapse
|