1
|
Fang B, Pan F, Shan T, Chen H, Peng W, Tian W, Huang F, Mao Z, Ding Y. An Integrated Virtual Screening Platform to Identify Potent Co-Assembled Nanodrugs for Cancer Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414154. [PMID: 39988868 DOI: 10.1002/adma.202414154] [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: 09/19/2024] [Revised: 01/18/2025] [Indexed: 02/25/2025]
Abstract
Co-assembled nanodrugs provide significant advantages in cancer treatment and drug delivery, yet effective screening methods to identify molecular combinations for co-assembly are lacking. This study presents a screening strategy integrating ligand-based virtual screening (LBVS) and density functional theory (DFT) calculations to explore new molecular combinations with co-assembly capabilities. The accuracy of this screening was validated by synthesizing various co-assembled nanodrugs under mild conditions. Vinpocetine (Vin) and lenvatinib (Len) are representative co-assembly combinations that can directly co-assemble into nanoparticles (NPs) through hydrogen bonding, van der Waals forces, and π-π interactions. These NPs were further functionalized with polyethylene glycol (PEG), resulting in PEG-L/V NPs that exhibited enhanced stability and biocompatibility. In addition, PEG-L/V NPs can respond to acidic conditions and release Vin and Len, working synergistically to induce cell cycle arrest and apoptosis in tumor cells in vitro while also inhibiting xenograft tumor growth in vivo. RNA sequencing (RNA-seq) analysis revealed that the co-assembled nanodrugs exhibited mechanisms that are distinct from those of single drugs. This study demonstrates the feasibility of utilizing a computational approach combining LBVS and DFT to identify small molecules with co-assembly capabilities, leading to innovative anticancer strategies.
Collapse
Affiliation(s)
- Bo Fang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
- Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Fei Pan
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100093, China
| | - Tianyu Shan
- Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
- Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Hualei Chen
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100093, China
| | - Wenjun Peng
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100093, China
| | - Wenli Tian
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, 100093, China
| | - Feihe Huang
- Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
- Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Zhengwei Mao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, 310009, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, 310009, China
- Center for Medical Research and Innovation in Digestive System Tumors Ministry of Education, China, Hangzhou, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
- ZJU-Pujian Research and Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, 310058, China
| |
Collapse
|
2
|
Arandhara A, Bhuyan P, Das BK. Exploring lung cancer microenvironment: pathways and nanoparticle-based therapies. Discov Oncol 2025; 16:159. [PMID: 39934547 DOI: 10.1007/s12672-025-01902-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Lung cancer stands out as a significant global health burden, with staggering incidence and mortality rates primarily linked to smoking and environmental carcinogens. The tumor microenvironment (TME) emerges as a critical determinant of cancer progression and treatment outcomes, comprising a complex interplay of cells, signaling molecules, and extracellular matrix. Through a comprehensive literature review, we elucidate current research trends and therapeutic prospects, aiming to advance our understanding of TME modulation strategies and their clinical implications for lung cancer treatment. Dysregulated immune responses within the TME can facilitate tumor evasion, limiting the efficacy of immune checkpoint inhibitors (ICI). Consequently, TME modulation strategies have become potential avenues to enhance therapeutic responses. However, conventional TME-targeted therapies often face challenges. In contrast, nanoparticle (NP)-based therapies offer promising prospects for improved drug delivery and reduced toxicity, leveraging the enhanced permeability and retention (EPR) effect. Despite NP design and delivery advancements, obstacles like poor tumor cell uptake and off-target effects persist, necessitating further optimization. This review underscores the pivotal role of TME in lung cancer management, emphasizing the synergistic potential of immunotherapy and nano-therapy.
Collapse
Affiliation(s)
- Arunabh Arandhara
- Assam Pharmacy Institute, Titabar, Amgurikhat, Jorhat, Assam, 785632, India
| | - Pallabi Bhuyan
- School of Pharmacy, The Assam Kaziranga University, Koraikhowa, Jorhat, Assam, 785006, India
| | - Bhrigu Kumar Das
- Department of Pharmacology, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Azara, Guwahati, Assam, 781017, India.
| |
Collapse
|
3
|
Ghaffari-Bohlouli P, Jafari H, Nie L, Kakkar A, Shavandi A. Enzymes in Addressing Hypoxia for Biomaterials Engineering. Adv Healthc Mater 2024; 13:e2401713. [PMID: 39183514 DOI: 10.1002/adhm.202401713] [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: 05/09/2024] [Revised: 08/05/2024] [Indexed: 08/27/2024]
Abstract
Oxygen is essential for normal cellular functions. Hypoxia impacts various cellular processes, such as metabolism, growth, proliferation, angiogenesis, metastasis, tumorigenesis, microbial infection, and immune response, mediated by hypoxia-inducible factors (HIFs). Hypoxia contributes to the progression and development of cancer, cardiovascular diseases, metabolic disorders, kidney diseases, and infections. The potential alleviation of hypoxia has been explored through the enzymatic in situ decomposition of hydrogen peroxide, leading to the generation of oxygen. However, challenges such as limited stability restrict the effectiveness of enzymes such as catalase in biomedical and in vivo applications. To overcome these limitations, targeted delivery of the enzymes has been proposed. This review offers a critical comparison of i) current approaches to enhance the in vivo stability of catalase; and ii) the structure, mechanism of action, and kinetics of catalase and catalase-like nanozymes.
Collapse
Affiliation(s)
- Pejman Ghaffari-Bohlouli
- 3BIO-BioMatter, École Polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec, H3A 0B8, Canada
| | - Hafez Jafari
- 3BIO-BioMatter, École Polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| | - Lei Nie
- College of Life Sciences, Xinyang Normal University, Xinyang, 464000, China
| | - Ashok Kakkar
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec, H3A 0B8, Canada
| | - Amin Shavandi
- 3BIO-BioMatter, École Polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| |
Collapse
|
4
|
Zu HL, Zhuang PP, Peng Y, Peng C, Peng C, Zhu ZJ, Yao Y, Yue J, Wang QS, Zhou WH, Wang HY. Dual-Drug Nanomedicine Assembly with Synergistic Anti-Aneurysmal Effects via Inflammation Suppression and Extracellular Matrix Stabilization. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402141. [PMID: 38953313 DOI: 10.1002/smll.202402141] [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: 03/18/2024] [Revised: 06/03/2024] [Indexed: 07/04/2024]
Abstract
Abdominal aortic aneurysm (AAA) represents a critical cardiovascular condition characterized by localized dilation of the abdominal aorta, carrying a significant risk of rupture and mortality. Current treatment options are limited, necessitating novel therapeutic approaches. This study investigates the potential of a pioneering nanodrug delivery system, RAP@PFB, in mitigating AAA progression. RAP@PFB integrates pentagalloyl glucose (PGG) and rapamycin (RAP) within a metal-organic-framework (MOF) structure through a facile assembly process, ensuring remarkable drug loading capacity and colloidal stability. The synergistic effects of PGG, a polyphenolic antioxidant, and RAP, an mTOR inhibitor, collectively regulate key players in AAA pathogenesis, such as macrophages and smooth muscle cells (SMCs). In macrophages, RAP@PFB efficiently scavenges various free radicals, suppresses inflammation, and promotes M1-to-M2 phenotype repolarization. In SMCs, it inhibits apoptosis and calcification, thereby stabilizing the extracellular matrix and reducing the risk of AAA rupture. Administered intravenously, RAP@PFB exhibits effective accumulation at the AAA site, demonstrating robust efficacy in reducing AAA progression through multiple mechanisms. Moreover, RAP@PFB demonstrates favorable biosafety profiles, supporting its potential translation into clinical applications for AAA therapy.
Collapse
Affiliation(s)
- Hong Lin Zu
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Pei Pei Zhuang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Ying Peng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Chao Peng
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Cheng Peng
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Zi Jia Zhu
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Ye Yao
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Jie Yue
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Qing Shan Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Wen Hu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Hai Yang Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| |
Collapse
|
5
|
Wang T, Chen S, Sun J, Li K. Functional co-delivery nanoliposomes based on improving hypoxia for increasing photoimmunotherapy efficacy of cold tumors. Int J Pharm 2024; 663:124581. [PMID: 39137819 DOI: 10.1016/j.ijpharm.2024.124581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/04/2024] [Accepted: 08/10/2024] [Indexed: 08/15/2024]
Abstract
Cold tumors lack T cells infiltration and have low immunogenicity, resulting insufficient immunotherapy response. Therefore, how to realize the transformation from cold tumor to hot tumor is an urgent problem to be solved. Photodynamic therapy can induce immunogenic death of tumor cells (ICD) and activate T lymphocytes to produce tumor immune response. However, hypoxia in the cold tumor microenvironment limits the effectiveness of photodynamic therapy. So in this article, MET-HMME/CAT-HMME@Nlip as a functional co-delivery nanoliposomes was constructed based on overcoming the above problems. Firstly, the oxygen-deficient state could be improved by the following two ways, one is catalase loaded in CAT-HMME@Nlip can decompose high concentration hydrogen peroxide to produce oxygen, and the other is metformin loaded in MET-HMME@Nlip can decrease oxygen consumption by inhibiting of mitochondrial respiration. And then with the increase of substrate oxygen concentration, the sensitivity of photodynamic therapy can be greatly improved and the anti-tumor immune response by PDT-induced ICD can also be enhanced obviously. In addition, metformin could act as a small molecule immune checkpoint inhibitor to reduce the expression of PD-L1 on the surface of tumor cells, thereby effectively improving the specific killing ability of cytotoxic T cells to tumor cells which could not only erasing the primary tumor, but also inhibiting the growth of simulated distant tumors through the immune memory function. This study provides a new idea for improving the clinical treatment effect of hypoxic cold tumors, especially for tumors that could not benefit from immunotherapy due to low or no expression of PD-L1 protein on the surface of tumor cells.
Collapse
Affiliation(s)
- Tian Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Siqi Chen
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Jianing Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Kexin Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| |
Collapse
|
6
|
Wan Y, Li C, Fu LH, Feng T, Zhang Y, Li Y, Lin J, Huang P, Cui DX. Erythrocyte Membrane Camouflaged Nanotheranostics for Optical Molecular Imaging-Escorted Self-Oxygenation Photodynamic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309026. [PMID: 38477698 DOI: 10.1002/smll.202309026] [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: 10/08/2023] [Revised: 02/27/2024] [Indexed: 03/14/2024]
Abstract
Hypoxic tumor microenvironment (TME) hampers the application of oxygen (O2)-dependent photodynamic therapy (PDT) in solid tumors. To address this problem, a biomimetic nanotheranostics (named MMCC@EM) is developed for optical molecular imaging-escorted self-oxygenation PDT. MMCC@EM is synthesized by encapsulating chlorin e6 (Ce6) and catalase (CAT) in metal-organic framework (MOF) nanoparticles with erythrocyte membrane (EM) camouflage. Based on the biomimetic properties of EM, MMCC@EM efficiently accumulates in tumor tissues. The enriched MMCC@EM achieves TME-activatable drug release, thereby releasing CAT and Ce6, and this process can be monitored through fluorescence (FL) imaging. In addition, endogenous hydrogen peroxide (H2O2) will be decomposed by CAT to produce O2, which can be reflected by the measurement of intratumoral oxygen concentration using photoacoustic (PA) imaging. Such self-oxygenation nanotheranostics effectively mitigate tumor hypoxia and improve the generation of singlet oxygen (1O2). The 1O2 disrupts mitochondrial function and triggers caspase-3-mediated cellular apoptosis. Furthermore, MMCC@EM triggers immunogenic cell death (ICD) effect, leading to an increased infiltration of cytotoxic T lymphocytes (CTLs) into tumor tissues. As a result, MMCC@EM exhibits good therapeutic effects in 4T1-tumor bearing mice under the navigation of FL/PA duplex imaging.
Collapse
Affiliation(s)
- Yilin Wan
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Chunying Li
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Lian-Hua Fu
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Ting Feng
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Yifan Zhang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Youyan Li
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, China
| | - Da-Xiang Cui
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| |
Collapse
|
7
|
Wang Y, Zhou X, Yao L, Hu Q, Liu H, Zhao G, Wang K, Zeng J, Sun M, Lv C. Capsaicin Enhanced the Efficacy of Photodynamic Therapy Against Osteosarcoma via a Pro-Death Strategy by Inducing Ferroptosis and Alleviating Hypoxia. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306916. [PMID: 38221813 DOI: 10.1002/smll.202306916] [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: 08/12/2023] [Revised: 01/04/2024] [Indexed: 01/16/2024]
Abstract
Ferroptosis, a novel form of nonapoptotic cell death, can effectively enhance photodynamic therapy (PDT) performance by disrupting intracellular redox homeostasis and promoting apoptosis. However, the extremely hypoxic tumor microenvironment (TME) together with highly expressed hypoxia-inducible factor-1α (HIF-1α) presents a considerable challenge for clinical PDT against osteosarcoma (OS). Hence, an innovative nanoplatform that enhances antitumor PDT by inducing ferroptosis and alleviating hypoxia is fabricated. Capsaicin (CAP) is widely reported to specifically activate transient receptor potential vanilloid 1 (TRPV1) channel, trigger an increase in intracellular Ca2+ concentration, which is closely linked with ferroptosis, and participate in decreased oxygen consumption by inhibiting HIF-1α in tumor cells, potentiating PDT antitumor efficiency. Thus, CAP and the photosensitizer IR780 are coencapsulated into highly biocompatible human serum albumin (HSA) to construct a nanoplatform (CI@HSA NPs) for synergistic tumor treatment under near-infrared (NIR) irradiation. Furthermore, the potential underlying signaling pathways of the combination therapy are investigated. CI@HSA NPs achieve real-time dynamic distribution monitoring and exhibit excellent antitumor efficacy with superior biosafety in vivo. Overall, this work highlights a promising NIR imaging-guided "pro-death" strategy to overcome the limitations of PDT for OS by promoting ferroptosis and alleviating hypoxia, providing inspiration and support for future innovative tumor therapy approaches.
Collapse
Affiliation(s)
- Yang Wang
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610064, P. R. China
| | - Xueru Zhou
- West China School of Pharmacy, Sichuan University, Chengdu, 610064, P. R. China
| | - Li Yao
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610064, P. R. China
| | - Qin Hu
- Emergency and Trauma College, Hainan Medical University, Haikou, 571199, P. R. China
| | - Haoran Liu
- Emergency and Trauma College, Hainan Medical University, Haikou, 571199, P. R. China
| | - Guosheng Zhao
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Kai Wang
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610064, P. R. China
| | - Jun Zeng
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610064, P. R. China
| | - Mingwei Sun
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610064, P. R. China
| | - Chuanzhu Lv
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610064, P. R. China
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, 571199, P. R. China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, P. R. China
| |
Collapse
|
8
|
Wang Z, Yang L. Natural-product-based, carrier-free, noncovalent nanoparticles for tumor chemo-photodynamic combination therapy. Pharmacol Res 2024; 203:107150. [PMID: 38521285 DOI: 10.1016/j.phrs.2024.107150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/22/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
Cancer, with its diversity, heterogeneity, and complexity, is a significant contributor to global morbidity, disability, and mortality, highlighting the necessity for transformative treatment approaches. Photodynamic therapy (PDT) has aroused continuous interest as a viable alternative to conventional cancer treatments that encounter drug resistance. Nanotechnology has brought new advances in medicine and has shown great potential in drug delivery and cancer treatment. For precise and efficient therapeutic utilization of such a tumor therapeutic approach with high spatiotemporal selectivity and minimal invasiveness, the carrier-free noncovalent nanoparticles (NPs) based on chemo-photodynamic combination therapy is essential. Utilizing natural products as the foundation for nanodrug development offers unparalleled advantages, including exceptional pharmacological activity, easy functionalization/modification, and well biocompatibility. The natural-product-based, carrier-free, noncovalent NPs revealed excellent synergistic anticancer activity in comparison with free photosensitizers and free bioactive natural products, representing an alternative and favorable combination therapeutic avenue to improve therapeutic efficacy. Herein, a comprehensive summary of current strategies and representative application examples of carrier-free noncovalent NPs in the past decade based on natural products (such as paclitaxel, 10-hydroxycamptothecin, doxorubicin, etoposide, combretastatin A4, epigallocatechin gallate, and curcumin) for tumor chemo-photodynamic combination therapy. We highlight the insightful design and synthesis of the smart carrier-free NPs that aim to enhance PDT efficacy. Meanwhile, we discuss the future challenges and potential opportunities associated with these NPs to provide new enlightenment, spur innovative ideas, and facilitate PDT-mediated clinical transformation.
Collapse
Affiliation(s)
- Zhonglei Wang
- Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China; School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus, Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Liyan Yang
- School of Physics and Physical Engineering, Qufu Normal University, Qufu 273165, PR China; Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China.
| |
Collapse
|
9
|
Negut I, Bita B. Polymersomes as Innovative, Stimuli-Responsive Platforms for Cancer Therapy. Pharmaceutics 2024; 16:463. [PMID: 38675124 PMCID: PMC11053450 DOI: 10.3390/pharmaceutics16040463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
This review addresses the urgent need for more targeted and less toxic cancer treatments by exploring the potential of multi-responsive polymersomes. These advanced nanocarriers are engineered to deliver drugs precisely to tumor sites by responding to specific stimuli such as pH, temperature, light, hypoxia, and redox conditions, thereby minimizing the side effects associated with traditional chemotherapy. We discuss the design, synthesis, and recent applications of polymersomes, emphasizing their ability to improve therapeutic outcomes through controlled drug release and targeted delivery. Moreover, we highlight the critical areas for future research, including the optimization of polymersome-biological interactions and biocompatibility, to facilitate their clinical adoption. Multi-responsive polymersomes emerge as a promising development in nanomedicine, offering a pathway to safer and more effective cancer treatments.
Collapse
Affiliation(s)
- Irina Negut
- Faculty of Physics, University of Bucharest, 077125 Magurele, Romania;
| | - Bogdan Bita
- Faculty of Physics, University of Bucharest, 077125 Magurele, Romania;
- National Institute for Lasers, Plasma and Radiation Physics, 077125 Magurele, Romania
| |
Collapse
|
10
|
Chen X, Mendes B, Zhuang Y, Conniot J, Mercado Argandona S, Melle F, Sousa DP, Perl D, Chivu A, Patra HK, Shepard W, Conde J, Fairen-Jimenez D. A Fluorinated BODIPY-Based Zirconium Metal-Organic Framework for In Vivo Enhanced Photodynamic Therapy. J Am Chem Soc 2024; 146:1644-1656. [PMID: 38174960 PMCID: PMC10797627 DOI: 10.1021/jacs.3c12416] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024]
Abstract
Photodynamic therapy (PDT), an emergent noninvasive cancer treatment, is largely dependent on the presence of efficient photosensitizers (PSs) and a sufficient oxygen supply. However, the therapeutic efficacy of PSs is greatly compromised by poor solubility, aggregation tendency, and oxygen depletion within solid tumors during PDT in hypoxic microenvironments. Despite the potential of PS-based metal-organic frameworks (MOFs), addressing hypoxia remains challenging. Boron dipyrromethene (BODIPY) chromophores, with excellent photostability, have exhibited great potential in PDT and bioimaging. However, their practical application suffers from limited chemical stability under harsh MOF synthesis conditions. Herein, we report the synthesis of the first example of a Zr-based MOF, namely, 69-L2, exclusively constructed from the BODIPY-derived ligands via a single-crystal to single-crystal post-synthetic exchange, where a direct solvothermal method is not applicable. To increase the PDT performance in hypoxia, we modify 69-L2 with fluorinated phosphate-functionalized methoxy poly(ethylene glycol). The resulting 69-L2@F is an oxygen carrier, enabling tumor oxygenation and simultaneously acting as a PS for reactive oxygen species (ROS) generation under LED irradiation. We demonstrate that 69-L2@F has an enhanced PDT effect in triple-negative breast cancer MDA-MB-231 cells under both normoxia and hypoxia. Following positive results, we evaluated the in vivo activity of 69-L2@F with a hydrogel, enabling local therapy in a triple-negative breast cancer mice model and achieving exceptional antitumor efficacy in only 2 days. We envision BODIPY-based Zr-MOFs to provide a solution for hypoxia relief and maximize efficacy during in vivo PDT, offering new insights into the design of promising MOF-based PSs for hypoxic tumors.
Collapse
Affiliation(s)
- Xu Chen
- The
Adsorption & Advanced Materials Laboratory (AML),
Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Bárbara
B. Mendes
- ToxOmics,
NOVA Medical School, Faculdade de Ciências Médicas,
NMS|FCM, Universidade Nova de Lisboa, Lisboa 2775-405, Portugal
| | - Yunhui Zhuang
- The
Adsorption & Advanced Materials Laboratory (AML),
Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - João Conniot
- ToxOmics,
NOVA Medical School, Faculdade de Ciências Médicas,
NMS|FCM, Universidade Nova de Lisboa, Lisboa 2775-405, Portugal
| | - Sergio Mercado Argandona
- The
Adsorption & Advanced Materials Laboratory (AML),
Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Francesca Melle
- The
Adsorption & Advanced Materials Laboratory (AML),
Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Diana P. Sousa
- ToxOmics,
NOVA Medical School, Faculdade de Ciências Médicas,
NMS|FCM, Universidade Nova de Lisboa, Lisboa 2775-405, Portugal
| | - David Perl
- Synchrotron
SOLEIL-UR1, L’Orme des Merisiers, Départementale 128, 91190 Saint-Aubin, France
| | - Alexandru Chivu
- Department
of Surgical Biotechnology, University College
London, London NW3 2PF, U.K.
| | - Hirak K. Patra
- Department
of Surgical Biotechnology, University College
London, London NW3 2PF, U.K.
| | - William Shepard
- Synchrotron
SOLEIL-UR1, L’Orme des Merisiers, Départementale 128, 91190 Saint-Aubin, France
| | - João Conde
- ToxOmics,
NOVA Medical School, Faculdade de Ciências Médicas,
NMS|FCM, Universidade Nova de Lisboa, Lisboa 2775-405, Portugal
| | - David Fairen-Jimenez
- The
Adsorption & Advanced Materials Laboratory (AML),
Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| |
Collapse
|
11
|
Liu L, Zhang Y, Li X, Deng J. Microenvironment of pancreatic inflammation: calling for nanotechnology for diagnosis and treatment. J Nanobiotechnology 2023; 21:443. [PMID: 37996911 PMCID: PMC10666376 DOI: 10.1186/s12951-023-02200-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Acute pancreatitis (AP) is a common and life-threatening digestive disorder. However, its diagnosis and treatment are still impeded by our limited understanding of its etiology, pathogenesis, and clinical manifestations, as well as by the available detection methods. Fortunately, the progress of microenvironment-targeted nanoplatforms has shown their remarkable potential to change the status quo. The pancreatic inflammatory microenvironment is typically characterized by low pH, abundant reactive oxygen species (ROS) and enzymes, overproduction of inflammatory cells, and hypoxia, which exacerbate the pathological development of AP but also provide potential targeting sites for nanoagents to achieve early diagnosis and treatment. This review elaborates the various potential targets of the inflammatory microenvironment of AP and summarizes in detail the prospects for the development and application of functional nanomaterials for specific targets. Additionally, it presents the challenges and future trends to develop multifunctional targeted nanomaterials for the early diagnosis and effective treatment of AP, providing a valuable reference for future research.
Collapse
Affiliation(s)
- Lu Liu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Street, Nanchong, 637001, China
| | - Yiqing Zhang
- Institute of Burn Research Southwest Hospital State Key Lab of Trauma Burn and Combined Injury Chongqing Key Laboratory for Disease Proteomics Army Medical University, Chongqing, 400038, China
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese PLA General Hospita, PLA Medical College, 28 Fu Xing Road, Beijing, 100853, China
| | - Xinghui Li
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Street, Nanchong, 637001, China.
| | - Jun Deng
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Street, Nanchong, 637001, China.
- Institute of Burn Research Southwest Hospital State Key Lab of Trauma Burn and Combined Injury Chongqing Key Laboratory for Disease Proteomics Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
12
|
Zhang C, Hu X, Jin L, Lin L, Lin H, Yang Z, Huang W. Strategic Design of Conquering Hypoxia in Tumor for Advanced Photodynamic Therapy. Adv Healthc Mater 2023; 12:e2300530. [PMID: 37186515 DOI: 10.1002/adhm.202300530] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/08/2023] [Indexed: 05/17/2023]
Abstract
Photodynamic therapy (PDT), with its advantages of high targeting, minimally invasive, and low toxicity side effects, has been widely used in the clinical therapy of various tumors, especially superficial tumors. However, the tumor microenvironment (TME) presents hypoxia due to the low oxygen (O2 ) supply caused by abnormal vascularization in neoplastic tissues and high O2 consumption induced by the rapid proliferation of tumor cells. The efficacy of oxygen-consumping PDT can be hampered by a hypoxic TME. To address this problem, researchers have been developing advanced nanoplatforms and strategies to enhance the therapeutic effect of PDT in tumor treatment. This review summarizes recent advanced PDT therapeutic strategies to against the hypoxic TME, thus enhancing PDT efficacy, including increasing O2 content in TME through delivering O2 to the tumors and in situ generations of O2 ; decreasing the O2 consumption during PDT by design of type I photosensitizers. Moreover, recent synergistically combined therapy of PDT and other therapeutic methods such as chemotherapy, photothermal therapy, immunotherapy, and gas therapy is accounted for by addressing the challenging problems of mono PDT in hypoxic environments, including tumor resistance, proliferation, and metastasis. Finally, perspectives of the opportunities and challenges of PDT in future clinical research and translations are provided.
Collapse
Affiliation(s)
- Cheng Zhang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
| | - Xiaoming Hu
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, P. R. China
- Jiangxi Key Laboratory of Nanobiomaterials, School of Materials Science and Engineering, East China Jiaotong University, Nanchang, 330013, P. R. China
| | - Long Jin
- Department of Pathology, Shengli Clinical Medical College, Fujian Medical University, Fuzhou, 350001, P. R. China
| | - Lisheng Lin
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
| | - Hongxin Lin
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
| | - Zhen Yang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, P. R. China
| | - Wei Huang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Strait Institute of Flexible Electronics (SIFE Future Technologies), Fujian Normal University, Fuzhou, 350007, P. R. China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, P. R. China
- Frontiers Science Center for Flexible Electronics (FSCFE), MIIT Key Laboratory of Flexible Electronics (KLoFE) Northwestern Polytechnical University Xi'an, Xi'an, 710072, P. R. China
| |
Collapse
|
13
|
Zhang Z, Ding C, Sun T, Wang L, Chen C. Tumor Therapy Strategies Based on Microenvironment-Specific Responsive Nanomaterials. Adv Healthc Mater 2023; 12:e2300153. [PMID: 36933000 DOI: 10.1002/adhm.202300153] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/10/2023] [Indexed: 03/19/2023]
Abstract
The tumor microenvironment (TME) is a complex and variable region characterized by hypoxia, low pH, high redox status, overexpression of enzymes, and high-adenosine triphosphate concentrations. In recent years, with the continuous in-depth study of nanomaterials, more and more TME-specific response nanomaterials are used for tumor treatment. However, the complexity of the TME causes different types of responses with various strategies and mechanisms of action. Aiming to systematically demonstrate the recent advances in research on TME-responsive nanomaterials, this work summarizes the characteristics of TME and outlines the strategies of different TME responses. Representative reaction types are illustrated and their merits and demerits are analyzed. Finally, forward-looking views on TME-response strategies for nanomaterials are presented. It is envisaged that such emerging strategies for the treatment of cancer are expected to exhibit dramatic trans-clinical capabilities, demonstrating the extensive potential for the diagnosis and therapy of cancer.
Collapse
Affiliation(s)
- Zhaocong Zhang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Chengwen Ding
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Tiedong Sun
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Lei Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Chunxia Chen
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| |
Collapse
|
14
|
Zhang J, Tang K, Fang R, Liu J, Liu M, Ma J, Wang H, Ding M, Wang X, Song Y, Yang D. Nanotechnological strategies to increase the oxygen content of the tumor. Front Pharmacol 2023; 14:1140362. [PMID: 36969866 PMCID: PMC10034070 DOI: 10.3389/fphar.2023.1140362] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/01/2023] [Indexed: 03/12/2023] Open
Abstract
Hypoxia is a negative prognostic indicator of solid tumors, which not only changes the survival state of tumors and increases their invasiveness but also remarkably reduces the sensitivity of tumors to treatments such as radiotherapy, chemotherapy and photodynamic therapy. Thus, developing therapeutic strategies to alleviate tumor hypoxia has recently been considered an extremely valuable target in oncology. In this review, nanotechnological strategies to elevate oxygen levels in tumor therapy in recent years are summarized, including (I) improving the hypoxic tumor microenvironment, (II) oxygen delivery to hypoxic tumors, and (III) oxygen generation in hypoxic tumors. Finally, the challenges and prospects of these nanotechnological strategies for alleviating tumor hypoxia are presented.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Kaiyuan Tang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Runqi Fang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Jiaming Liu
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Ming Liu
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Jiayi Ma
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Hui Wang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Meng Ding
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- *Correspondence: Meng Ding, ; Xiaoxiao Wang, ; Dongliang Yang,
| | - Xiaoxiao Wang
- Biochemical Engineering Research Center, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma’anshan, China
- *Correspondence: Meng Ding, ; Xiaoxiao Wang, ; Dongliang Yang,
| | - Yanni Song
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, China
| | - Dongliang Yang
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, China
- *Correspondence: Meng Ding, ; Xiaoxiao Wang, ; Dongliang Yang,
| |
Collapse
|
15
|
Feng Q, Xu J, Zhuang C, Xiong J, Wang H, Xiao K. Mitochondria-Targeting and Multiresponsive Nanoplatform Based on AIEgens for Synergistic Chemo-Photodynamic Therapy and Enhanced Immunotherapy. Biomacromolecules 2023; 24:977-990. [PMID: 36703538 DOI: 10.1021/acs.biomac.2c01416] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Although photodynamic therapy (PDT) has become an attractive strategy for cancer treatment, its clinical application still suffers from some limitations, including insufficient delivery of photosensitizers, hypoxic tumor environment, and the development of PDT resistance. To address these limitations, a new class of mitochondria-targeting and fluorinated polymer with aggregation-induced emission characteristics was fabricated to sensitize PDT and co-deliver chemotherapeutic drugs. The amphiphilic fluoropolymer was able to efficiently carry oxygen and SN-38 (the active metabolite of irinotecan) and self-assemble into multifunctional micellar nanoparticles (SN-38-TTCF@O2 NPs). Upon internalization into tumor cells, these NPs could successfully escape lysosomes, selectively target mitochondria, efficiently produce reactive oxygen species (ROS) under light irradiation, and release drugs in response to ROS. In the HCT116 tumor xenograft model, they preferentially accumulated in tumor tissue and significantly alleviated tumor hypoxia, resulting in synergistic chemo-PDT efficacy without distinct toxicity. Furthermore, the nanoscale chemo-PDT induced immunogenic cell death, promoted the recruitment and activation of cytotoxic T lymphocytes, and ultimately augmented the anti-tumor efficacy of anti-PD-1 antibody in the murine CT26 tumor model. These results may provide novel insights into the development of efficient chemo-PDT nanomedicine to improve the outcome of immunotherapy.
Collapse
Affiliation(s)
- Qiyi Feng
- Precision Medicine Research Center, Sichuan Provincial Key Laboratory of Precision Medicine and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Junhuai Xu
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Cheng Zhuang
- Precision Medicine Research Center, Sichuan Provincial Key Laboratory of Precision Medicine and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Junjie Xiong
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Haibo Wang
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Kai Xiao
- Precision Medicine Research Center, Sichuan Provincial Key Laboratory of Precision Medicine and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, P. R. China.,Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| |
Collapse
|
16
|
Yi M, Xiong B, Li Y, Guo W, Huang Y, Lu B. Manipulate tumor hypoxia for improved photodynamic therapy using nanomaterials. Eur J Med Chem 2023; 247:115084. [PMID: 36599230 DOI: 10.1016/j.ejmech.2022.115084] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/20/2022] [Accepted: 12/30/2022] [Indexed: 01/01/2023]
Abstract
Due to its low adverse effects, minimal invasiveness, and outstanding patient compliance, photodynamic therapy (PDT) has drawn a great deal of interest, which is achieved through incomplete reduction of O2 by a photosensitizer under light illumination that produces amounts of reactive oxygen species (ROS). However, tumor hypoxia significantly hinders the therapeutic effect of PDT so that tumor cells cannot be eliminated, which results in tumor cells proliferating, invading, and metastasizing. Additionally, O2 consumption during PDT exacerbates hypoxia in tumors, leading to several adverse events after PDT treatment. In recent years, various investigations have focused on conquering or using tumor hypoxia by nanomaterials to amplify PDT efficacy, which is summarized in this review. This comprehensive review's objective is to present novel viewpoints on the advancement of oxygenation nanomaterials in this promising field, which is motivated by hypoxia-associated anti-tumor therapy.
Collapse
Affiliation(s)
- Mengqi Yi
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Bei Xiong
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Yuyang Li
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Wei Guo
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Yunhan Huang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Bo Lu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China.
| |
Collapse
|
17
|
Paul M, Itoo AM, Ghosh B, Biswas S. Hypoxia alleviating platinum(IV)/chlorin e6-based combination chemotherapeutic-photodynamic nanomedicine for oropharyngeal carcinoma. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 238:112627. [PMID: 36525775 DOI: 10.1016/j.jphotobiol.2022.112627] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/09/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022]
Abstract
Hypoxia is an important pathological hallmark of the tumor microenvironment, associated with metabolic alterations, cell proliferation, aggressiveness, metastasis, and therapy resistance in cancers. Hypoxia impedes the outcome of photodynamic therapy (PDT), which is largely dependent on molecular oxygen to generate cytotoxic 1O2. Here, a near-infrared light activatable, oxygen-generating nanomicellar PDT-chemotherapy system (mPPCPN Ms) constituted of amphiphilic mPEG-PLA, photosensitizer Ce6, and tetravalent platinum prodrug Pt(IV)-diazide was developed for oral squamous cell carcinoma. The polymer conjugate self-assemble to nanosize (115 ± 2.35 nm) micelles, which, upon irradiation (660 nm laser), activated Ce6, and photodecomposed to produce cytotoxic Pt(II), azidyl radical, and molecular oxygen. The strategically fabricated PDT-chemotherapy produced a strong antitumor response in vitro using oral squamous cell carcinoma and in vivo in oral cancer-xenografted mouse models, revealing its significant potential in chemo-photodynamic combination therapy with the benefit of reversing hypoxia.
Collapse
Affiliation(s)
- Milan Paul
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| | - Asif Mohd Itoo
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| | - Balaram Ghosh
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| | - Swati Biswas
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India.
| |
Collapse
|
18
|
Li X, Chen L, Huang M, Zeng S, Zheng J, Peng S, Wang Y, Cheng H, Li S. Innovative strategies for photodynamic therapy against hypoxic tumor. Asian J Pharm Sci 2023; 18:100775. [PMID: 36896447 PMCID: PMC9989661 DOI: 10.1016/j.ajps.2023.100775] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/15/2022] [Accepted: 01/05/2023] [Indexed: 01/19/2023] Open
Abstract
Photodynamic therapy (PDT) is applied as a robust therapeutic option for tumor, which exhibits some advantages of unique selectivity and irreversible damage to tumor cells. Among which, photosensitizer (PS), appropriate laser irradiation and oxygen (O2) are three essential components for PDT, but the hypoxic tumor microenvironment (TME) restricts the O2 supply in tumor tissues. Even worse, tumor metastasis and drug resistance frequently happen under hypoxic condition, which further deteriorate the antitumor effect of PDT. To enhance the PDT efficiency, critical attention has been received by relieving tumor hypoxia, and innovative strategies on this topic continue to emerge. Traditionally, the O2 supplement strategy is considered as a direct and effective strategy to relieve TME, whereas it is confronted with great challenges for continuous O2 supply. Recently, O2-independent PDT provides a brand new strategy to enhance the antitumor efficiency, which can avoid the influence of TME. In addition, PDT can synergize with other antitumor strategies, such as chemotherapy, immunotherapy, photothermal therapy (PTT) and starvation therapy, to remedy the inadequate PDT effect under hypoxia conditions. In this paper, we summarized the latest progresses in the development of innovative strategies to improve PDT efficacy against hypoxic tumor, which were classified into O2-dependent PDT, O2-independent PDT and synergistic therapy. Furthermore, the advantages and deficiencies of various strategies were also discussed to envisage the prospects and challenges in future study.
Collapse
Affiliation(s)
- Xiaotong Li
- Department of Anesthesiology, the Second Clinical School of Guangzhou Medical University, Guangzhou 510182, China
| | - Lei Chen
- Department of Anesthesiology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Miaoting Huang
- Department of Anesthesiology, the Second Clinical School of Guangzhou Medical University, Guangzhou 510182, China
| | - Shaoting Zeng
- Department of Anesthesiology, the Second Clinical School of Guangzhou Medical University, Guangzhou 510182, China
| | - Jiayi Zheng
- Department of Anesthesiology, the Second Clinical School of Guangzhou Medical University, Guangzhou 510182, China
| | - Shuyi Peng
- Department of Anesthesiology, the Second Clinical School of Guangzhou Medical University, Guangzhou 510182, China
| | - Yuqing Wang
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Hong Cheng
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Shiying Li
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
19
|
Chang Y, Cui P, Zhou S, Qiu L, Jiang P, Chen S, Wang C, Wang J. Metal-phenolic network for cancer therapy. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
20
|
Wang C, Li F, Zhang T, Yu M, Sun Y. Recent advances in anti-multidrug resistance for nano-drug delivery system. Drug Deliv 2022; 29:1684-1697. [PMID: 35616278 PMCID: PMC9154776 DOI: 10.1080/10717544.2022.2079771] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy for tumors occasionally results in drug resistance, which is the major reason for the treatment failure. Higher drug doses could improve the therapeutic effect, but higher toxicity limits the further treatment. For overcoming drug resistance, functional nano-drug delivery system (NDDS) has been explored to sensitize the anticancer drugs and decrease its side effects, which are applied in combating multidrug resistance (MDR) via a variety of mechanisms including bypassing drug efflux, controlling drug release, and disturbing metabolism. This review starts with a brief report on the major MDR causes. Furthermore, we searched the papers from NDDS and introduced the recent advances in sensitizing the chemotherapeutic drugs against MDR tumors. Finally, we concluded that the NDDS was based on several mechanisms, and we looked forward to the future in this field.
Collapse
Affiliation(s)
- Changduo Wang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Fashun Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Tianao Zhang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Min Yu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| |
Collapse
|
21
|
Mfouo-Tynga IS, Mouinga-Ondeme AG. Photodynamic Therapy: A Prospective Therapeutic Approach for Viral Infections and Induced Neoplasia. Pharmaceuticals (Basel) 2022; 15:ph15101273. [PMID: 36297385 PMCID: PMC9608479 DOI: 10.3390/ph15101273] [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: 09/18/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022] Open
Abstract
The recent COVID-19 pandemic outbreak and arising complications during treatments have highlighted and demonstrated again the evolving ability of microorganisms, especially viral resistance to treatment as they develop into new and strong strains. The search for novel and effective treatments to counter the effects of ever-changing viruses is undergoing. Although it is an approved procedure for treating cancer, photodynamic therapy (PDT) was first used against bacteria and has now shown potential against viruses and certain induced diseases. PDT is a multi-stage process and uses photosensitizing molecules (PSs) that accumulate in diseased tissues and eradicates them after being light-activated in the presence of oxygen. In this review, studies describing viruses and their roles in disrupting cell regulation mechanisms and signaling pathways and facilitating tumorigenesis were described. With the development of innovative “or smart” PSs through the use of nanoparticles and two-photon excitation, among other strategies, PDT can boost immune responses, inactivate viral infections, and eradicate neoplastic cells. Visualization and monitoring of biological processes can be achieved in real-time with nanomedicines and better tissue penetration strategies. After photodynamic inactivation of viruses, signaling pathways seem to be restored but the underlying mechanisms are still to be elucidated. Light-mediated treatments are suitable to manage both oncogenic viral infections and induced neoplasia.
Collapse
|
22
|
Zeng Y, Xu G, Kong X, Ye G, Guo J, Lu C, Nezamzadeh-Ejhieh A, Shahnawaz Khan M, Liu J, Peng Y. Recent advances of the core-shell MOFs in tumour therapy. Int J Pharm 2022; 627:122228. [PMID: 36162610 DOI: 10.1016/j.ijpharm.2022.122228] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 12/15/2022]
Abstract
Coordination chemistry has always been vital to explore the material prominence of metal-organic systems. The metal-organic chemistry plays a fundamental role in decisive structural features, which are accountable for tuning the properties of materials. Tumour therapy has become an important research field of medical treatment in the world. Metal-organic frameworks (MOFs) have attracted extensive interest in medical science research due to their large effective surface area, clear pore network, and critical catalytic performance. Compared with traditional MOF materials, MOF materials with core-shell structures have a higher loading rate and better stability, which can overcome a single function. They have been successfully used in tumour medical research and have excellent prospects for diagnosing and treating various tumours. The current review article thoroughly describes the various synthetic approaches for engineering core-shell MOF materials, the structural types, and the potential functional applications. We also discussed core-shell MOF materials for the various treatment of tumours, such as tumour chemotherapy, tumour phototherapy and tumour microenvironment anti-hypoxia therapy. In this paper, the synthesized procedures of core-shell MOFs and their applications for tumour treatment have been discussed, and their future research has prospected. The current improved strategies, challenges, and prospects are also presented because of the metal-organic chemistry governing the structural modification of core-shell MOFs for tumour therapy applications. Therefore, the present review article opens a new door for medicinal chemists to tune the structural features of the core-shell MOF materials to modulate tumour therapy with simple, low-cost materials for better human lives.
Collapse
Affiliation(s)
- Yana Zeng
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China; Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
| | - Guihua Xu
- Department of Science and Education, The Dongguan Affiliated Hospital of Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan 523900, China
| | - Xiangyang Kong
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China; Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
| | - Gaomin Ye
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
| | - Jian Guo
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, PR China.
| | - Chengyu Lu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
| | | | - M Shahnawaz Khan
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Jianqiang Liu
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China; Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China.
| | - Yanqiong Peng
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
23
|
Xiao X, Chen M, Zhang Y, Li L, Peng Y, Li J, Zhou W. Hemin-incorporating DNA nanozyme enabling catalytic oxygenation and GSH depletion for enhanced photodynamic therapy and synergistic tumor ferroptosis. J Nanobiotechnology 2022; 20:410. [PMID: 36109814 PMCID: PMC9479271 DOI: 10.1186/s12951-022-01617-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/31/2022] [Indexed: 11/18/2022] Open
Abstract
Photodynamic therapy (PDT) has emerged as a promising tumor treatment method via light-triggered generation of reactive oxygen species (ROS) to kill tumor cells. However, the efficacy of PDT is usually restricted by several biological limitations, including hypoxia, excess glutathione (GSH) neutralization, as well as tumor resistance. To tackle these issues, herein we developed a new kind of DNA nanozyme to realize enhanced PDT and synergistic tumor ferroptosis. The DNA nanozyme was constructed via rolling circle amplification, which contained repeat AS1411 G quadruplex (G4) units to form multiple G4/hemin DNAzymes with catalase-mimic activity. Both hemin, an iron-containing porphyrin cofactor, and chlorine e6 (Ce6), a photosensitizer, were facilely inserted into G4 structure with high efficiency, achieving in-situ catalytic oxygenation and photodynamic ROS production. Compared to other self-oxygen-supplying tools, such DNA nanozyme is advantageous for high biological stability and compatibility. Moreover, the nanostructure could achieve tumor cells targeting internalization and intranuclear transport of Ce6 by virtue of specific nucleolin binding of AS1411. The nanozyme could catalyze the decomposition of intracellular H2O2 into oxygen for hypoxia relief as evidenced by the suppression of hypoxia-inducible factor-1α (HIF-1α), and moreover, GSH depletion and cell ferroptosis were also achieved for synergistic tumor therapy. Upon intravenous injection, the nanostructure could effectively accumulate into tumor, and impose multi-modal tumor therapy with excellent biocompatibility. Therefore, by integrating the capabilities of O2 generation and GSH depletion, such DNA nanozyme is a promising nanoplatform for tumor PDT/ferroptosis combination therapy.
Collapse
Affiliation(s)
- Xiaoxiong Xiao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Min Chen
- Department of Thoracic Surgery, The Second People's Hospital of Huaihua City, Huaihua, China
| | - Yuchen Zhang
- Department of Pharmacy, Yichun People's Hospital, Yichun, Jiangxi, China
| | - Liang Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
| | - Ying Peng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
| | - Junyu Li
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China.
| | - Wenhu Zhou
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China.
| |
Collapse
|
24
|
Tian H, Zhang T, Qin S, Huang Z, Zhou L, Shi J, Nice EC, Xie N, Huang C, Shen Z. Enhancing the therapeutic efficacy of nanoparticles for cancer treatment using versatile targeted strategies. J Hematol Oncol 2022; 15:132. [PMID: 36096856 PMCID: PMC9469622 DOI: 10.1186/s13045-022-01320-5] [Citation(s) in RCA: 166] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Poor targeting of therapeutics leading to severe adverse effects on normal tissues is considered one of the obstacles in cancer therapy. To help overcome this, nanoscale drug delivery systems have provided an alternative avenue for improving the therapeutic potential of various agents and bioactive molecules through the enhanced permeability and retention (EPR) effect. Nanosystems with cancer-targeted ligands can achieve effective delivery to the tumor cells utilizing cell surface-specific receptors, the tumor vasculature and antigens with high accuracy and affinity. Additionally, stimuli-responsive nanoplatforms have also been considered as a promising and effective targeting strategy against tumors, as these nanoplatforms maintain their stealth feature under normal conditions, but upon homing in on cancerous lesions or their microenvironment, are responsive and release their cargoes. In this review, we comprehensively summarize the field of active targeting drug delivery systems and a number of stimuli-responsive release studies in the context of emerging nanoplatform development, and also discuss how this knowledge can contribute to further improvements in clinical practice.
Collapse
Affiliation(s)
- Hailong Tian
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Tingting Zhang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jiayan Shi
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, 3800, VIC, Australia
| | - Edouard C Nice
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China
| | - Na Xie
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China. .,West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China.
| | - Canhua Huang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.
| |
Collapse
|
25
|
Du Y, Han J, Jin F, Du Y. Recent Strategies to Address Hypoxic Tumor Environments in Photodynamic Therapy. Pharmaceutics 2022; 14:pharmaceutics14091763. [PMID: 36145513 PMCID: PMC9505114 DOI: 10.3390/pharmaceutics14091763] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 12/02/2022] Open
Abstract
Photodynamic therapy (PDT) has become a promising method of cancer treatment due to its unique properties, such as noninvasiveness and low toxicity. The efficacy of PDT is, however, significantly reduced by the hypoxia tumor environments, because PDT involves the generation of reactive oxygen species (ROS), which requires the great consumption of oxygen. Moreover, the consumption of oxygen caused by PDT would further exacerbate the hypoxia condition, which leads to angiogenesis, invasion of tumors to other parts, and metastasis. Therefore, many research studies have been conducted to design nanoplatforms that can alleviate tumor hypoxia and enhance PDT. Herein, the recent progress on strategies for overcoming tumor hypoxia is reviewed, including the direct transport of oxygen to the tumor site by O2 carriers, the in situ generation of oxygen by decomposition of oxygen-containing compounds, reduced O2 consumption, as well as the regulation of tumor microenvironments. Limitations and future perspectives of these technologies to improve PDT are also discussed.
Collapse
|
26
|
Yuan Z, Liu C, Sun Y, Li Y, Wu H, Ma S, Shang J, Zhan Y, Yin P, Gao F. Bufalin exacerbates Photodynamic therapy of colorectal cancer by targeting SRC-3/HIF-1α pathway. Int J Pharm 2022; 624:122018. [PMID: 35839982 DOI: 10.1016/j.ijpharm.2022.122018] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/28/2022] [Accepted: 07/10/2022] [Indexed: 10/17/2022]
Abstract
Photodynamic therapy (PDT) induces tumour cell death by producing reactive oxygen species (ROS), and hypoxia is one of the main factors that limits its efficiency. In our previous study, bufalin (BU) enhanced photosensitizer mTHPC-mediated PDT therapy in colorectal cancer (CRC) cells, but its mechanism was not elucidated. To explore a strategy for improving the efficacy of PDT, we designed iRGD-modified nanoparticles to co-capsuled mTHPC and BU for simultaneous delivery to the tumour site and explored the underlying mechanism of the synergistic anti-CRC effect. In our study, mTHPC&BU@VES-CSO/TPGS-RGD nanoparticles (T-B@NP) had a particle size of 148.3 ± 2.5 nm and a zeta potential of 22.8 ± 2.0 mV. Specifically, these nanoparticles passively accumulated in tumour cells, and under laser irradiation, mTHPC induced cell apoptosis and death. In addition, the sustained release of BU inhibited HIF-1α and reduced VEGF-mediated angiogenesis by targeting the SRC-3/HIF-1α pathway, which induced a strong PDT effect against CRC. In vivo studies demonstrated that codelivery of the nanoparticles under laser irradiation exhibited a superior antitumour effect (84.2%) and significantly prolonged survival time of mice, with the mechanisms of alleviating hypoxia and inhibiting angiogenesis. In summary, mTHPC and BU codelivery via nanoparticles efficiently enhances the therapeutic effects of PDT by inhibiting the SRC-3/HIF-1α pathway in CRC. This work provides an effective strategy to combat hypoxia-induced tumour resistance and overcome the barriers of PDT treatment.
Collapse
Affiliation(s)
- Zeting Yuan
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, Shanghai, 200237, China; Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai 200062, China; Central Lab, Shanghai 200062, China
| | - Chaolian Liu
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, Shanghai, 200237, China; Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai 200062, China; Central Lab, Shanghai 200062, China
| | - Yuji Sun
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, Shanghai, 200237, China; Shanghai Key Laboratory of Functional Materials Chemistry, Shanghai, 200237, China; Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yue Li
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai 200062, China; Central Lab, Shanghai 200062, China
| | - Honglei Wu
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai 200062, China; Central Lab, Shanghai 200062, China
| | - Shuli Ma
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, Shanghai, 200237, China; Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai 200062, China; Central Lab, Shanghai 200062, China
| | - Jing Shang
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai 200062, China; Central Lab, Shanghai 200062, China
| | - Yueping Zhan
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai 200062, China; Central Lab, Shanghai 200062, China
| | - Peihao Yin
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai 200062, China; Central Lab, Shanghai 200062, China; Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Feng Gao
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, Shanghai, 200237, China; Shanghai Key Laboratory of Functional Materials Chemistry, Shanghai, 200237, China; Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
27
|
Zi Y, Yang K, He J, Wu Z, Liu J, Zhang W. Strategies to enhance drug delivery to solid tumors by harnessing the EPR effects and alternative targeting mechanisms. Adv Drug Deliv Rev 2022; 188:114449. [PMID: 35835353 DOI: 10.1016/j.addr.2022.114449] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/03/2022] [Accepted: 07/06/2022] [Indexed: 12/20/2022]
Abstract
The Enhanced Permeability and Retention (EPR) effect has been recognized as the central paradigm in tumor-targeted delivery in the last decades. In the wake of this concept, nanotechnologies have reached phenomenal levels in research. However, clinical tumors display a poor manifestation of EPR effect. Factors including tumor heterogeneity, complicating tumor microenvironment, and discrepancies between laboratory models and human tumors largely contribute to poor efficiency in tumor-targeted delivery and therapeutic failure in clinical translation. In this article, approaches for evaluation of EPR effect in human tumor were overviewed as guidance to employ EPR effect for cancer treatment. Strategies to augment EPR-mediated tumoral delivery are discussed in different dimensions including enhancement of vascular permeability, depletion of tumor extracellular matrix and optimization of nanoparticle design. Besides, the recent development in alternative tumor-targeted delivery mechanisms are highlighted including transendothelial pathway, endogenous cell carriers and non-immunogenic bacteria-mediated delivery. In addition, the emerging preclinical models better reflect human tumors are introduced. Finally, more rational applications of EPR effect in other disease and field are proposed. This article elaborates on fundamental reasons for the gaps between theoretical expectation and clinical outcomes, attempting to provide some perspective directions for future development of cancer nanomedicines in this still evolving landscape.
Collapse
Affiliation(s)
- Yixuan Zi
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Kaiyun Yang
- School of Pharmacy, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Jianhua He
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Zimei Wu
- School of Pharmacy, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
28
|
Guo L, Zhong S, Liu P, Guo M, Ding J, Zhou W. Radicals Scavenging MOFs Enabling Targeting Delivery of siRNA for Rheumatoid Arthritis Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2202604. [PMID: 35661593 DOI: 10.1002/smll.202202604] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Indexed: 06/15/2023]
Abstract
Macrophages play essential roles in the progression of rheumatoid arthritis (RA), which are polarized into the pro-inflammatory M1 phenotype with significant oxidative stress and cytokines excretion. Herein, an active targeting nanomedicine based on metal-organic frameworks (MOFs) to re-educate the diseased macrophages for RA therapy is reported. The MOFs are prepared via coordination between tannic acid (TA) and Fe3+ , and anti-TNF-α siRNA is loaded via a simple sonication process, achieving high loading capacity comparable to cationic vectors. The MOFs show excellent biocompatibility, and enable rapid endo/lysosome escape of siRNA via the proton-sponge effect for effective cytokines down-regulation. Importantly, such nanomedicine displays intrinsic radicals scavenging capability to eliminate a broad spectrum of reactive oxygen and nitrogen species (RONS), which in turn repolarizes the M1 macrophages into anti-inflammatory M2 phenotypes for enhanced RA therapy in combination with siRNA. The MOFs are further modified with bovine serum albumin (BSA) to allow cascade RA joint and diseased macrophages targeted delivery. As a result, an excellent anti-RA efficacy is achieved in a collagen-induced arthritis mice model. This work provides a robust gene vector with great translational potential, and offers a vivid example of rationally designing MOF structure with multifunctionalities to synergize with its payload for enhanced disease treatment.
Collapse
Affiliation(s)
- Lina Guo
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Shenghui Zhong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
- School of Medicine, Yichun University, Yichun, Jiangxi, 336000, China
| | - Peng Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Man Guo
- Academician Workstation, Changsha Medical University, Changsha, 410219, China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
- Academician Workstation, Changsha Medical University, Changsha, 410219, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Changsha, Hunan, 410008, China
| |
Collapse
|
29
|
Duan W, Li B, Zhang W, Li J, Yao X, Tian Y, Zheng J, Li D. Two-photon responsive porphyrinic metal-organic framework involving Fenton-like reaction for enhanced photodynamic and sonodynamic therapy. J Nanobiotechnology 2022; 20:217. [PMID: 35524276 PMCID: PMC9074235 DOI: 10.1186/s12951-022-01436-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/24/2022] [Indexed: 11/17/2022] Open
Abstract
Designing new oxygenation nanomaterials by oxygen-generating or oxygen-carrying strategies in hypoxia-associated anti-tumor therapy is a high priority target yet challenge. In this work, we fabricated a nanoplatform involving Fenton-like reaction, Pd@MOF-525@HA, to relieve tumor hypoxia via oxygen-generating strategy for enhanced oxygen-dependent anti-tumor therapy. Thereinto, the porphyrinic MOF-525 can produce singlet oxygen (1O2) via light or ultrasonic irradiation for photodynamic and sonodynamic therapy. Notably, the well-dispersed Pd nanocubes within MOF-525 can convert H2O2 into O2 to mitigate the hypoxic environment for enhanced therapy outcome. Moreover, the two-photon activity and cancer cell specific targeting capability of Pd@MOF-525@HA gave rise to deeper tissue penetration and near-infrared light-induced fluorescence imaging to achieve precise guidance for cancer therapy. This work provides a feasible way in designing new oxygenation nanomaterials to relieve tumor hypoxia for enhanced cancer treatment.
Collapse
Affiliation(s)
- Wenyao Duan
- Institutes of Physics Science and Information Technology, Key Laboratory of Structure and Functional Regulation of Hybrid Materials, Ministry of Education, Anhui University, Hefei, 230601, People's Republic of China
| | - Bo Li
- Institutes of Physics Science and Information Technology, Key Laboratory of Structure and Functional Regulation of Hybrid Materials, Ministry of Education, Anhui University, Hefei, 230601, People's Republic of China
| | - Wen Zhang
- Department of Chemistry, Key Laboratory of Functional Inorganic Material Chemistry of Anhui Province, Anhui University, Hefei, 230601, People's Republic of China
| | - Jiaqi Li
- Institutes of Physics Science and Information Technology, Key Laboratory of Structure and Functional Regulation of Hybrid Materials, Ministry of Education, Anhui University, Hefei, 230601, People's Republic of China
| | - Xin Yao
- Department of Chemistry, Key Laboratory of Functional Inorganic Material Chemistry of Anhui Province, Anhui University, Hefei, 230601, People's Republic of China
| | - Yupeng Tian
- Department of Chemistry, Key Laboratory of Functional Inorganic Material Chemistry of Anhui Province, Anhui University, Hefei, 230601, People's Republic of China
| | - Jun Zheng
- Institutes of Physics Science and Information Technology, Key Laboratory of Structure and Functional Regulation of Hybrid Materials, Ministry of Education, Anhui University, Hefei, 230601, People's Republic of China.
| | - Dandan Li
- Institutes of Physics Science and Information Technology, Key Laboratory of Structure and Functional Regulation of Hybrid Materials, Ministry of Education, Anhui University, Hefei, 230601, People's Republic of China. .,Department of Chemistry, Key Laboratory of Functional Inorganic Material Chemistry of Anhui Province, Anhui University, Hefei, 230601, People's Republic of China.
| |
Collapse
|
30
|
Lv K, Wang L, Ma Y, Zhang F, Guo W, Yu K, Qu F, Lin H. Biodegradation Mn-CoS@carbon di-shell nanoheterostructure with enhanced nanozyme-mediated phototherapy. BIOMATERIALS ADVANCES 2022; 136:212778. [PMID: 35929316 DOI: 10.1016/j.bioadv.2022.212778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/09/2022] [Accepted: 03/24/2022] [Indexed: 06/15/2023]
Abstract
The efficacy of phototherapy is dependent on intracellular O2 concentration and NIR harvest. Here, a simple nanoplatform with nanoenzyme mediated phototherapy enhances anticancer capacity. Mn-CoS@carbon (CMS/C) di-shell hollow nanospheres (50 nm) are synthesized successfully through two-step consecutive Kirkendall process. The nanoheterostructure reveals the higher near-infrared (NIR) light absorption and photothermal conversion rate of 66.3% than pure CoS (45.5%), owing to the decreased band gap and multi-reflection of incident light in the hollow structure. And CMS/C reveals the reactive oxygen species (ROS) production and nanoenzyme activities (mimic peroxidase and catalase) that are 6 and 2 times than those of pure CoS. Furthermore, the nanoenzyme exhibits NIR-enhanced abilities to produce more OH and O2 facilitating anticancer. In addition, it also depletes glutathione (mimicking glutathione oxidase), to disturb intracellular redox-homeostasis, boosting the increase of oxidative stress. With grafting bovine serum albumin (BSA) and drug loading, CMS/C@BSA-Dox integrated multi-therapy make the great anticancer effect in vitro and vivo. After that, the nanocomposite could be biodegraded and eliminated via urinary and feces within 14 days. Based on this work, the efficient charge-separation can be designed to reveal high performance nanoenzymes as well as photosensitizers for anticancer.
Collapse
Affiliation(s)
- Kexin Lv
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Limin Wang
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Yajie Ma
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Feng Zhang
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Wei Guo
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Kai Yu
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Fengyu Qu
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China.
| | - Huiming Lin
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China; Laboratory for Photon and Electronic Bandgap Materials, Ministry of Education, Harbin Normal University, Harbin 150025, China.
| |
Collapse
|
31
|
Core-shell structured nanoparticles for photodynamic therapy-based cancer treatment and related imaging. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214427] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
32
|
An J, Tang S, Hong G, Chen W, Chen M, Song J, Li Z, Peng X, Song F, Zheng WH. An unexpected strategy to alleviate hypoxia limitation of photodynamic therapy by biotinylation of photosensitizers. Nat Commun 2022; 13:2225. [PMID: 35469028 PMCID: PMC9038921 DOI: 10.1038/s41467-022-29862-9] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 04/04/2022] [Indexed: 01/25/2023] Open
Abstract
The most common working mechanism of photodynamic therapy is based on high-toxicity singlet oxygen, which is called Type II photodynamic therapy. But it is highly dependent on oxygen consumption. Recently, Type I photodynamic therapy has been found to have better hypoxia tolerance to ease this restriction. However, few strategies are available on the design of Type I photosensitizers. We herein report an unexpected strategy to alleviate the limitation of traditional photodynamic therapy by biotinylation of three photosensitizers (two fluorescein-based photosensitizers and the commercially available Protoporphyrin). The three biotiylated photosensitizers named as compound 1, 2 and 3, exhibit impressive ability in generating both superoxide anion radicals and singlet oxygen. Moreover, compound 1 can be activated upon low-power white light irradiation with stronger ability of anion radicals generation than the other two. The excellent combinational Type I / Type II photodynamic therapy performance has been demonstrated with the photosensitizers 1. This work presents a universal protocol to provide tumor-targeting ability and enhance or trigger the generation of anion radicals by biotinylation of Type II photosensitizers against tumor hypoxia. Type I photodynamic therapy (PDT) sensitizers show good hypoxia tolerance but only few strategies are available for the design of purely organic Type I photosensitizers (PS). Here, the authors use biotinylation as design strategy to obtain PS-Biotin sensitizers with high efficiency for the generation of superoxide anion radicals and singlet oxygen.
Collapse
Affiliation(s)
- Jing An
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Shanliang Tang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Gaobo Hong
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Wenlong Chen
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Miaomiao Chen
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Jitao Song
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, 266237, Qingdao, China
| | - Zhiliang Li
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, 266237, Qingdao, China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China
| | - Fengling Song
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 116024, Dalian, China. .,Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, 266237, Qingdao, China.
| | - Wen-Heng Zheng
- Department of Interventional Therapy, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, 110042, Shenyang, China.
| |
Collapse
|
33
|
Liu P, Shi X, Peng Y, Hu J, Ding J, Zhou W. Anti-PD-L1 DNAzyme Loaded Photothermal Mn 2+ /Fe 3+ Hybrid Metal-Phenolic Networks for Cyclically Amplified Tumor Ferroptosis-Immunotherapy. Adv Healthc Mater 2022; 11:e2102315. [PMID: 34841741 DOI: 10.1002/adhm.202102315] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Indexed: 12/11/2022]
Abstract
Ferroptosis can activate immune response via inducing tumor cells immunogenic cell death (ICD), and antitumor immunity in turn boosts the efficacy of ferroptosis by excreting interferon gamma (IFN-γ), which shows a promising combo for synergistically amplified tumor treatment. However, their combination is strictly limited by the complexity of tumor microenvironment, including poor ferroptosis response and immunosuppressive factors in tumor. Herein, a metal-phenolic networks (MPNs) nanoplatform with all-active components is constructed to favor the ferroptosis-immunotherapy cyclical synergism. The photothermal MPNs are assembled via coordination between tannic acid (TA) and metal-ion complex of Fe3+ /Mn2+ , through which a PD-L1 inhibiting DNAzyme (DZ) is loaded to regulate the immunosuppressive PD-1/PD-L1 pathway. After intracellular delivery, each component of MPNs exerts their respective functions: Fe2+ is in situ generated from Fe3+ by TA reduction to trigger ferroptosis, while DZ is activated by Mn2+ to effectively silence PD-L1. With external laser irradiation, photothermal therapy is initiated to synergize with ferroptosis for enhanced ICD, which induces strong antitumor immunes. Combined with DZ-mediated PD-L1 suppression, a cyclically amplified tumor ferroptosis-immunotherapy is achieved, resulting in obliteration of both primary and distant tumor. This work provides a smart, simple, yet robust nanomedicine-based combination for self-amplified tumor treatment.
Collapse
Affiliation(s)
- Peng Liu
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410013 China
| | - Xinyi Shi
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410013 China
| | - Ying Peng
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410013 China
| | - Jianming Hu
- Department of Pathology the First Affiliated Hospital Shihezi University School of Medicine Shihezi Xinjiang 832003 China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410013 China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410013 China
- Henan Key Laboratory of Biomolecular Recognition and Sensing Shangqiu Normal University Shangqiu Henan 476000 China
| |
Collapse
|
34
|
Zhao Z, Liu Z, Hua Y, Pan Y, Yi G, Wu S, He C, Zhang Y, Yang Y. Biomimetic ZIF8 Nanosystem With Tumor Hypoxia Relief Ability to Enhance Chemo-Photothermal Synergistic Therapy. Front Pharmacol 2022; 13:850534. [PMID: 35401170 PMCID: PMC8988193 DOI: 10.3389/fphar.2022.850534] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/31/2022] [Indexed: 11/23/2022] Open
Abstract
Tumor hypoxic microenvironment can reduce the therapeutic effects of chemotherapy, radiotherapy, photodynamic therapy, immunotherapy, etc. It is also a potential source of tumor recurrence and metastasis. A biomimetic nanosystem based on zeolitic imidazolate framework 8 (ZIF8), which had multifunctions of hypoxia relief, chemotherapy, and photothermal therapy, was established to improve tumor hypoxic microenvironment and overcome the corresponding therapeutic resistance. ZIF8 enveloped with DOX and CuS nanoparticles (DC@ZIF8) was synthesized by a sedimentation method. Red blood cell membrane and catalase (CAT) were coated onto DC@ZIF8 and biomimetic nanosystem (DC@ZIF8-MEMC) was formed. The designed DC@ZIF8-MEMC had a shape of polyhedron with an average particle size around 254 nm. The loading content of DOX, CAT, and CuS was 4.9%, 6.2%, and 2.5%, separately. The release of DOX from DC@ZIF8-MEMC was pH dependent and significantly faster at pH 5 due to the degradation of ZIF8. DC@ZIF8-MEMC exhibited outstanding photothermal conversion properties and excellent antitumor effect in vitro and in vivo. Moreover, the hypoxia relief by CAT was proved to have good sensitization effect on chemo-photothermal combined therapy. DC@ZIF8-MEMC is a prospective nanosystem, which can realize great chemo-photothermal synergistic antitumor effect under the sensitization of CAT. The biomimetic multifunctional nanoplatform provides a potential strategy of chemo-photothermal synergistic antitumor effect under the sensitization of CAT.
Collapse
Affiliation(s)
- Ziming Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Department of Pharmaceutics, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Zhaorong Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Yabing Hua
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Department of Pharmaceutics, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Yuanjie Pan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Ge Yi
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Shengyue Wu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Cong He
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Yanzhuo Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Department of Pharmaceutics, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Yanzhuo Zhang, ; Yihua Yang,
| | - Yihua Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Department of Pharmaceutics, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Yanzhuo Zhang, ; Yihua Yang,
| |
Collapse
|
35
|
Sun W, Xu Y, Yao Y, Yue J, Wu Z, Li H, Shen G, Liao Y, Wang H, Zhou W. Self-oxygenation mesoporous MnO2 nanoparticles with ultra-high drug loading capacity for targeted arteriosclerosis therapy. J Nanobiotechnology 2022; 20:88. [PMID: 35183183 PMCID: PMC8858544 DOI: 10.1186/s12951-022-01296-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/05/2022] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis (AS) is a leading cause of vascular diseases that severely threats the human health due to the lack of efficient therapeutic methods. During the development and progress of AS, macrophages play critical roles, which are polarized into pro-inflammatory M1 phenotype to excrete abundant cytokines and overproduce reactive oxygen species (ROS), and take up excess amount of lipid to form foam cells. In this work, we developed a MnO2-based nanomedicine to re-educate macrophages for targeting AS therapy. The MnO2 was one-pot synthesized under mild condition, showing intrinsic catalase-mimic activity for self-oxygenation by using endogenous H2O2 as substrate. Moreover, the mesoporous structure as well as the abundant metal coordination sites in MnO2 structure facilitated the loading of an anti-AS drug of curcumin (Cur), achieving extraordinarily high drug loading capacity of 54%. Cur displayed a broad spectrum of anti-oxidant and anti-inflammatory capabilities to repolarize M1 macrophages into M2 phenotype, and the catalytic MnO2 recovered the function of lipid efflux transporter to remove lipid from cells by suppressing HIF-1α. Collectively, the nanocarrier and the payload drug functioned as an all-active nanoplatform to synergistically alleviate the syndromes of AS. In ApoE−/− mice model, the nanosystem could significantly prolong the circulation half-life of Cur by sixfold, and enhance drug accumulation in atherosclerotic lesion by 3.5-fold after intravenous injection by virtue of surface hyaluronic acid (HA) modification. As a result, a robust anti-AS efficacy was achieved as evidenced by the decrease of atherosclerotic lesion, plaque area, lipid level.
Collapse
|
36
|
Mei H, Cai S, Huang D, Gao H, Cao J, He B. Carrier-free nanodrugs with efficient drug delivery and release for cancer therapy: From intrinsic physicochemical properties to external modification. Bioact Mater 2022; 8:220-240. [PMID: 34541398 PMCID: PMC8424425 DOI: 10.1016/j.bioactmat.2021.06.035] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/23/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022] Open
Abstract
The considerable development of carrier-free nanodrugs has been achieved due to their high drug-loading capability, simple preparation method, and offering "all-in-one" functional platform features. However, the native defects of carrier-free nanodrugs limit their delivery and release behavior throughout the in vivo journey, which significantly compromise the therapeutic efficacy and hinder their further development in cancer treatment. In this review, we summarized and discussed the recent strategies to enhance drug delivery and release of carrier-free nanodrugs for improved cancer therapy, including optimizing the intrinsic physicochemical properties and external modification. Finally, the corresponding challenges that carrier-free nanodrugs faced are discussed and the future perspectives for its application are presented. We hope this review will provide constructive information for the rational design of more effective carrier-free nanodrugs to advance therapeutic treatment.
Collapse
Affiliation(s)
- Heng Mei
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Shengsheng Cai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Dennis Huang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78731, USA
| | - Huile Gao
- West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Jun Cao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Bin He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| |
Collapse
|
37
|
Zhou R, Zeng X, Zhao H, Chen Q, Wu P. Combating the hypoxia limit of photodynamic therapy through reversing the survival-related pathways of cancer cells. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
38
|
Zhang X, He C, Xiang G. Engineering nanomedicines to inhibit hypoxia-inducible Factor-1 for cancer therapy. Cancer Lett 2022; 530:110-127. [PMID: 35041892 DOI: 10.1016/j.canlet.2022.01.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/18/2021] [Accepted: 01/10/2022] [Indexed: 11/02/2022]
Abstract
Hypoxia-inducible factor-1 (HIF-1), an essential promoter of tumor progression, has attracted increasing attention as a therapeutic target. In addition to hypoxic cellular conditions, HIF-1 activation can be triggered by cancer treatment, which causes drug tolerance and therapeutic failure. To date, a series of effective strategies have been explored to suppress HIF-1 function, including silencing the HIF-1α gene, inhibiting HIF-1α protein translation, degrading HIF-1α protein, and inhibiting HIF-1 transcription. Furthermore, nanoparticle-based drug delivery systems have been widely developed to improve the stability and pharmacokinetics of HIF-1 inhibitors or achieve HIF-1-targeted combination therapies as a nanoplatform. In this review, we summarize the current literature on nanomedicines targeting HIF-1 to combat cancer and discuss their potential for future development.
Collapse
Affiliation(s)
- Xiaojuan Zhang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chuanchuan He
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guangya Xiang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
39
|
Ma Y, Qu X, Liu C, Xu Q, Tu K. Metal-Organic Frameworks and Their Composites Towards Biomedical Applications. Front Mol Biosci 2022; 8:805228. [PMID: 34993235 PMCID: PMC8724581 DOI: 10.3389/fmolb.2021.805228] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 11/22/2021] [Indexed: 01/10/2023] Open
Abstract
Owing to their unique features, including high cargo loading, biodegradability, and tailorability, metal–organic frameworks (MOFs) and their composites have attracted increasing attention in various fields. In this review, application strategies of MOFs and their composites in nanomedicine with emphasis on their functions are presented, from drug delivery, therapeutic agents for different diseases, and imaging contrast agents to sensor nanoreactors. Applications of MOF derivatives in nanomedicine are also introduced. Besides, we summarize different functionalities related to MOFs, which include targeting strategy, biomimetic modification, responsive moieties, and other functional decorations. Finally, challenges and prospects are highlighted about MOFs in future applications.
Collapse
Affiliation(s)
- Yana Ma
- School of Basic Medical Sciences, Xi'an Key Laboratory of Immune Related Diseases, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| | - Xianglong Qu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Cui Liu
- School of Basic Medical Sciences, Xi'an Key Laboratory of Immune Related Diseases, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| | - Qiuran Xu
- Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China.,Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
40
|
Qin S, Xu Y, Li H, Chen H, Yuan Z. Recent advances in in situ oxygen-generating and oxygen-replenishing strategies for hypoxic-enhanced photodynamic therapy. Biomater Sci 2021; 10:51-84. [PMID: 34882762 DOI: 10.1039/d1bm00317h] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cancer is a leading cause of death worldwide, accounting for an estimated 10 million deaths by 2020. Over the decades, various strategies for tumor therapy have been developed and evaluated. Photodynamic therapy (PDT) has attracted increasing attention due to its unique characteristics, including low systemic toxicity and minimally invasive nature. Despite the excellent clinical promise of PDT, hypoxia is still the Achilles' heel associated with its oxygen-dependent nature related to increased tumor proliferation, angiogenesis, and distant metastases. Moreover, PDT-mediated oxygen consumption further exacerbates the hypoxia condition, which will eventually lead to the poor effect of drug treatment and resistance and irreversible tumor metastasis, even limiting its effective application in the treatment of hypoxic tumors. Hypoxia, with increased oxygen consumption, may occur in acute and chronic hypoxia conditions in developing tumors. Tumor cells farther away from the capillaries have much lower oxygen levels than cells in adjacent areas. However, it is difficult to change the tumor's deep hypoxia state through different ways to reduce the tumor tissue's oxygen consumption. Therefore, it will become more difficult to cure malignant tumors completely. In recent years, numerous investigations have focused on improving PDT therapy's efficacy by providing molecular oxygen directly or indirectly to tumor tissues. In this review, different molecular oxygen supplementation methods are summarized to alleviate tumor hypoxia from the innovative perspective of using supplemental oxygen. Besides, the existing problems, future prospects and potential challenges of this strategy are also discussed.
Collapse
Affiliation(s)
- Shuheng Qin
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China.
| | - Yue Xu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China.
| | - Hua Li
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China.
| | - Haiyan Chen
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China.
| | - Zhenwei Yuan
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing 210009, China.
| |
Collapse
|
41
|
Sun Y, Wang Y, Liu Y, Weng B, Yang H, Xiang Z, Ran J, Wang H, Yang C. Intelligent Tumor Microenvironment-Activated Multifunctional Nanoplatform Coupled with Turn-on and Always-on Fluorescence Probes for Imaging-Guided Cancer Treatment. ACS APPLIED MATERIALS & INTERFACES 2021; 13:53646-53658. [PMID: 34748304 DOI: 10.1021/acsami.1c17642] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Intrinsic tumor microenvironment (TME)-related therapeutic resistance and nontumor-specific imaging have limited the application of imaging-guided cancer therapy. Herein, a TME-responsive MnO2-based nanoplatform coupled with turn-on and always-on fluorescence probes was designed through a facile biomineralization method for imaging-guided photodynamic/chemodynamic/photothermal therapy (PDT/CDT/PTT). After the tumor-targeting delivery of the AuNCs@MnO2-ICG@AS1411 (AMIT) nanoplatform via aptamer AS1411, the TME-responsive dissociation of MnO2 generated sufficient O2 and Mn2+ with the consumption of GSH for improving PDT efficacy and Fenton-like reaction-mediated CDT. Simultaneously, the released small-sized ICG and AuNCs facilitated PDT and PTT efficacy via the deep tumor penetration. Moreover, the turn-on fluorescence of AuNCs revealed the real-time TME-responsive MnO2 degradation process, and the always-on ICG fluorescence enabled the in situ monitoring of the payload distribution in vitro and in vivo. The AMIT NPs also provided magnetic resonance and thermal imaging guidance for the enhanced PDT, CDT, and PTT. Therefore, this all-in-one nanosystem provides a simple and versatile strategy for multiple imaging-guided theranostic applications.
Collapse
Affiliation(s)
- Yudong Sun
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
- College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Yifei Wang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
- College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Yaqi Liu
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
- College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Benrui Weng
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
- College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Huiran Yang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
- College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Zhouxuan Xiang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
- College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Jiabing Ran
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
- College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Huimin Wang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
- College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Changying Yang
- Hubei Key Laboratory of Natural Products Research and Development, China Three Gorges University, Yichang 443002, China
- College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| |
Collapse
|
42
|
Zhou X, Huang JQ, Liu LS, Deng FA, Liu YB, Li YM, Chen AL, Yu XY, Li SY, Cheng H. Self-Remedied Nanomedicine for Surmounting the Achilles' Heel of Photodynamic Tumor Therapy. ACS APPLIED BIO MATERIALS 2021; 4:8023-8032. [PMID: 35006783 DOI: 10.1021/acsabm.1c00938] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Oxygen-dependent photodynamic therapy (PDT) could exacerbate tumor hypoxia to induce the upregulation of hypoxia-inducible factor-1α (HIF-1α), which would promote tumor growth and metastasis. In this paper, a self-remedied nanomedicine is developed based on a photosensitizer and a HIF-1α inhibitor to surmount the Achilles' heel of PDT for enhanced antitumor efficacy. Specifically, the nanomedicine (designated as CYC-1) is prepared by the self-assembly of chlorine e6 (Ce6) and 3-(5'-hydroxy-methyl-2'-furyl)-1-benzylindazole (YC-1) through π-π stacking and hydrophobic interactions. Of special note, carrier-free CYC-1 holds an extremely high drug loading rate and avoids excipient-triggered adverse reactions. Intravenously administered CYC-1 prefers to accumulate in the tumor tissue for effective cellular uptake. More importantly, it is verified that CYC-1 is capable of inhibiting the HIF-1α activity, thereby improving its PDT efficacy on tumor suppression. Besides, CYC-1 has the overwhelming superiority in restraining tumor proliferation over the combined administration of Ce6 and YC-1, which highlights the advantage of this self-remedied strategy in drug delivery and tumor therapy. This study sheds light on the development of self-delivery nanomedicine for efficient PDT against malignancies.
Collapse
Affiliation(s)
- Xiang Zhou
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Jia-Qi Huang
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Ling-Shan Liu
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Fu-An Deng
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Yi-Bin Liu
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Yan-Mei Li
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - A-Li Chen
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Xi-Yong Yu
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Shi-Ying Li
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Hong Cheng
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| |
Collapse
|
43
|
Lu J, Ni C, Huang J, Liu Y, Tao Y, Hu P, Wang Y, Zheng S, Shi M. Biocompatible Mesoporous Silica-Polydopamine Nanocomplexes as MR/Fluorescence Imaging Agent for Light-Activated Photothermal-Photodynamic Cancer Therapy In Vivo. Front Bioeng Biotechnol 2021; 9:752982. [PMID: 34858959 PMCID: PMC8630682 DOI: 10.3389/fbioe.2021.752982] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/13/2021] [Indexed: 11/13/2022] Open
Abstract
Conventional cancer phototherapy with single modality suffers from low therapeutic efficacy and undesired posttreatment damage for adjacent normal tissues. Therefore, the lower NIR laser irradiation power is vital to the reduction or preclusion of risk of scalds and burns in normal tissues. Herein, we rationally proposed a novel multifunctional nanocomplex, which enabled good magnetic resonance (MR) imaging contrast effect and promising photothermal conversion efficacy. The prepared core/shell nanocomplexes [MSN-Ce6@PDA (Mn)] were composed of chlorin e6-embedded mesoporous silica/nanoparticle composites as the cores, and then polydopamine and manganese ions were conjugated on the cores to form protective shells. The MSN-Ce6@PDA (Mn) nanocomplexes revealed superior properties in colloidal stability, photothermal conversion, reaction oxygen species generation, magnetic resonance imaging, etc. Under the guidance of MR and fluorescence imaging, these MSN-Ce6@PDA (Mn) nanocomplexes were found to be primarily accumulated in the MDA-MB-231 tumor area. Furthermore, the combined photodynamic and photothermal therapy exhibited strong inhibition to the growth of MDA-MB-231 tumor in vitro and in vivo. Besides, the MSN-Ce6@PDA (Mn) nanocomplexes also exhibited excellent biocompatibility and low damage to the healthy animals. Hence, the results demonstrated that the prepared MSN-Ce6@PDA (Mn) nanocomplex would be a promising potential for multimodal imaging-guided phototherapy.
Collapse
Affiliation(s)
- Jiahui Lu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, China
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, China
| | - Chen Ni
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, China
| | - Jie Huang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, China
| | - Yawen Liu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, China
| | - Yingkai Tao
- Department of Dermatology and Venereal Diseases, The First People’s Hospital of Changzhou, Changzhou, China
| | - Pengcheng Hu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, China
| | - Yong Wang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, China
| | - Shaohui Zheng
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, China
- Institute of Medical Imaging and Digital Medicine, Xuzhou Medical University, Xuzhou, China
| | - Meilin Shi
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, China
- Institute of Medical Imaging and Digital Medicine, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
44
|
Le JQ, Yang F, Yin MD, Zhao RR, Zhang BC, Li C, Lin JF, Fang YF, Lin YT, Shao JW. Biomimetic polyphenol-coated nanoparticles by Co-assembly of mTOR inhibitor and photosensitizer for synergistic chemo-photothermal therapy. Colloids Surf B Biointerfaces 2021; 209:112177. [PMID: 34749194 DOI: 10.1016/j.colsurfb.2021.112177] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/12/2021] [Accepted: 10/20/2021] [Indexed: 12/28/2022]
Abstract
Rapamycin (RAPA) functions as effectively clinical immunosuppressive agent, its significant tumor growth suppression effect via various pathways in diverse cancers, especially combined with photothermal therapy, is gaining a burgeoning attention. However, its critical defects, low solubility and poor stability, have severely hampered its further application. Herein, RAPA, indocyanine green (ICG) and epigallocatechin gallate (EGCG) serving as chemotherapeutic drug, photosensitizer and biomimetic coatings, respectively, were co-assembled into carrier-free, high biocompatible ICG-RAPA-EGCG nanoparticles (IRE NPs) for synergistic cancer therapy. Particularly, the bioinspired EGCG coatings not only improved the stability of IRE NPs under physiological conditions to avert NPs disassembly and drug release, but also maintained the photostability of ICG to achieve excellent photothermal response. The results indicated that the as-prepared IRE NPs displayed good monodispersity and enhanced stability at various stored media after introducing of EGCG. Compared with monotherapy of RAPA or ICG, IRE NPs showed higher dose-dependent toxicity in MCF-7 cells, HepG2 cells and HeLa cells, especially plus near-infrared laser irradiation. Furthermore, IRE NPs exhibited quicker uptake in cells, higher accumulation in tumor region (even in 48 h) than free ICG and effectively inhibited tumor growth without side effect in H22 tumor-bearing mice. Collectively, the carrier-free IRE NPs provided a simply alternative approach to fabricate RAPA/photosensitizer co-loaded nanoparticles for combinatorial tumor therapy.
Collapse
Affiliation(s)
- Jing-Qing Le
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Fang Yang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Meng-Die Yin
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Rui-Rui Zhao
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Bing-Chen Zhang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Chao Li
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Juan-Fang Lin
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Yi-Fan Fang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Yu-Ting Lin
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Jing-Wei Shao
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China.
| |
Collapse
|
45
|
Wang C, Li Y, Yang W, Zhou L, Wei S. Nanozyme with Robust Catalase Activity by Multiple Mechanisms and Its Application for Hypoxic Tumor Treatment. Adv Healthc Mater 2021; 10:e2100601. [PMID: 34390206 DOI: 10.1002/adhm.202100601] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/21/2021] [Indexed: 01/02/2023]
Abstract
Utilizing catalase-mimicking nanozymes to produce O2 is an effective method to overcome tumor hypoxia. However, it is challenging to fabricate nanozymes with ultrahigh catalytic activity. Palladium nanosheet (Pd NS), a photothermal agent for photothermal therapy (PTT), has superior catalase-mimicking activity. Here, titanium dioxide (TiO2 ) is used to modify Pd NS (denoted Pd@TiO2 ) by a simple one-step method to improve its catalytic activity about 8 times. The enhancement mechanism's fundamental insights are discussed through experiments and density functional theory calculations. Next, zinc phthalocyanine is loaded on Pd@TiO2 to form a nanomotor (denoted PTZCs) with the synergistic activities of photodynamic therapy and PTT. PTZCs inherit the catalase activity of Pd@TiO2 to facilitate the decomposition of endogenous H2 O2 to O2 , which can relieve tumor hypoxia and propel PTZC migration to expand the reach of PTZCs, further enhancing its synergistic treatment outcome both in vitro and in vivo. It is proposed that this work can provide a simple and effective strategy for catalytic activity enhancement and bring a critical new perspective to studying and guiding the nanozyme design.
Collapse
Affiliation(s)
- Chongchong Wang
- College of Chemistry and Materials Science Jiangsu Key Laboratory of Biofunctional Materials Jiangsu Collaborative Innovation Center of Biomedical Functional Materials Key Laboratory of Applied Photochemistry Nanjing Normal University Nanjing 210023 China
| | - Yanqing Li
- College of Chemistry and Materials Science Jiangsu Key Laboratory of Biofunctional Materials Jiangsu Collaborative Innovation Center of Biomedical Functional Materials Key Laboratory of Applied Photochemistry Nanjing Normal University Nanjing 210023 China
| | - Weijie Yang
- Department of Power Engineering School of Energy Power and Mechanical Engineering North China Electric Power University Baoding 071003 China
| | - Lin Zhou
- College of Chemistry and Materials Science Jiangsu Key Laboratory of Biofunctional Materials Jiangsu Collaborative Innovation Center of Biomedical Functional Materials Key Laboratory of Applied Photochemistry Nanjing Normal University Nanjing 210023 China
| | - Shaohua Wei
- College of Chemistry and Materials Science Jiangsu Key Laboratory of Biofunctional Materials Jiangsu Collaborative Innovation Center of Biomedical Functional Materials Key Laboratory of Applied Photochemistry Nanjing Normal University Nanjing 210023 China
- School of Chemistry and Chemical Engineering Yancheng Institute of Technology Yancheng 224051 China
| |
Collapse
|
46
|
Wan X, Zhang H, Pan W, Li N, Tang B. An enzyme nanopocket based on covalent organic frameworks for long-term starvation therapy and enhanced photodynamic therapy of cancer. Chem Commun (Camb) 2021; 57:5402-5405. [PMID: 33942842 DOI: 10.1039/d0cc07544b] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
An enzyme nanopocket constructed from a porphyrin-based covalent organic framework (COF) was developed to co-load glucose oxidase (GOx) and catalase (CAT) for long-term starvation therapy and enhanced photodynamic therapy (PDT).
Collapse
Affiliation(s)
- Xiuyan Wan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Huiwen Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| |
Collapse
|
47
|
Zheng Q, Liu X, Zheng Y, Yeung KWK, Cui Z, Liang Y, Li Z, Zhu S, Wang X, Wu S. The recent progress on metal-organic frameworks for phototherapy. Chem Soc Rev 2021; 50:5086-5125. [PMID: 33634817 DOI: 10.1039/d1cs00056j] [Citation(s) in RCA: 238] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Some infectious or malignant diseases such as cancers are seriously threatening the health of human beings all over the world. The commonly used antibiotic therapy cannot effectively treat these diseases within a short time, and also bring about adverse effects such as drug resistance and immune system damage during long-term systemic treatment. Phototherapy is an emerging antibiotic-free strategy to treat these diseases. Upon light irradiation, phototherapeutic agents can generate cytotoxic reactive oxygen species (ROS) or induce a temperature increase, which leads to the death of targeted cells. These two kinds of killing strategies are referred to as photodynamic therapy (PDT) and photothermal therapy (PTT), respectively. So far, many photo-responsive agents have been developed. Among them, the metal-organic framework (MOF) is becoming one of the most promising photo-responsive materials because its structure and chemical compositions can be easily modulated to achieve specific functions. MOFs can have intrinsic photodynamic or photothermal ability under the rational design of MOF construction, or serve as the carrier of therapeutic agents, owing to its tunable porosity. MOFs also provide feasibility for various combined therapies and targeting methods, which improves the efficiency of phototherapy. In this review, we firstly investigated the principles of phototherapy, and comprehensively summarized recent advances of MOF in PDT, PTT and synergistic therapy, from construction to modification. We expect that our demonstration will shed light on the future development of this field, and bring it one step closer to clinical trials.
Collapse
Affiliation(s)
- Qiyao Zheng
- School of Materials Science & Engineering, The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China.
| | - Xiangmei Liu
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science & Engineering, Hubei University, Wuhan 430062, China.
| | - Yufeng Zheng
- State Key Laboratory for Turbulence and Complex System and Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Kelvin W K Yeung
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Zhenduo Cui
- School of Materials Science & Engineering, The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China.
| | - Yanqin Liang
- School of Materials Science & Engineering, The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China.
| | - Zhaoyang Li
- School of Materials Science & Engineering, The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China.
| | - Shengli Zhu
- School of Materials Science & Engineering, The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China.
| | - Xianbao Wang
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, School of Materials Science & Engineering, Hubei University, Wuhan 430062, China.
| | - Shuilin Wu
- School of Materials Science & Engineering, The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
48
|
Zeng Y, Zhou H, Ding J, Zhou W. Cell membrane inspired nano-shell enabling long-acting Glucose Oxidase for Melanoma starvation therapy via microneedles-based percutaneous delivery. Theranostics 2021; 11:8270-8282. [PMID: 34373741 PMCID: PMC8344000 DOI: 10.7150/thno.60758] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/05/2021] [Indexed: 12/27/2022] Open
Abstract
Rationale: Glucose oxidase (GOx) has gained tremendous research interest recently as a glucose-consuming enzyme for tumor starvation therapy, while its in vivo applications are strictly limited by rapid deactivation, as well as side effects of non-specific catalysis. Methods: To address these issues, here we report a protective nano-shell to encapsule GOx for localized melanoma therapy delivered by dissolving microneedles (MNs). Inspired by cell membrane that separates and protects cell organelles and components from outside environment while selectively ingesting nutrition sources, we designed polydopamine (PDA)-structured nano-shell to allow free transportation of glucose for catalytic reaction, while impede the penetration of GOx, proteinase, and other GOx-deactivating macromolecules across the shell membrane. Results: GOx was well protected in core layer with persistent catalytic activity for at least 6 d under various biological matrixes (e.g., PBS, serum, and cell lysate) and surviving different harsh conditions (e.g., acid/base treatments, and proteinase-induced degradation). Such long-acting nano-catalyst can be easily integrated into MNs as topical delivery carrier for effective glucose consumption in melanoma tissue, achieving significant tumor growth inhibition via starvation therapy with minimized side effects as compared to systemic administration. Conclusion: This work provides an elegant platform for in vivo delivery of GOx, and our cell-mimicking nano-system can also be applied for other enzyme-based therapeutics.
Collapse
|
49
|
Zhang LJ, Huang R, Shen YW, Liu J, Wu Y, Jin JM, Zhang H, Sun Y, Chen HZ, Luan X. Enhanced anti-tumor efficacy by inhibiting HIF-1α to reprogram TAMs via core-satellite upconverting nanoparticles with curcumin mediated photodynamic therapy. Biomater Sci 2021; 9:6403-6415. [PMID: 34259235 DOI: 10.1039/d1bm00675d] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tumor hypoxic stress after photodynamic therapy (PDT) will be inevitably exacerbated by the vascular blocking effects and oxygen consumption in the tumor microenvironment (TME) which usually leads to compromised efficacy and clinical performance. Increasing evidence links the hypoxia induced up-regulation of hypoxia inducible factor 1α (HIF-1α) with immunosuppressive TME, including the polarization of M2 phenotype tumor associated macrophages (TAMs), which promote the recurrence and metastasis. Here, we reported NIR-triggered core-satellite upconverting nanoparticles (CSNPs) with curcumin (Cur) embedded as a difunctional photosensitizer, which could realize PDT in deep tumors with long excitation wavelength (980 nm) and reverse the immunosuppressive TME induced by up-regulated HIF-1α at the same time. This Cur-loaded CSNPs (Cur-CSNPs)-mediated PDT could successfully induce the immunogenic cell death (ICD) of triple negative breast cancer (TNBC) cell lines (4T1 and MDA-MB-231) in vitro and repolarize the 4T1 cells co-cultured TAMs from pro-tumor M2 to the anti-tumor M1 phenotype. Furthermore, Cur-CSNPs-mediated PDT could suppress the 4T1 tumor growth in primary and distant sites through the synergistic immunotherapeutic effects in vivo by priming M1 type TAMs and CD4+/CD8+ T cells' infiltration. Our data highlight the novel application of CSNPs-embedded Cur as a difunctional photosensitizer to enhance the anti-tumor efficacy of PDT.
Collapse
Affiliation(s)
- Li-Jun Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Rui Huang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yi-Wen Shen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jie Liu
- Department of Research and Development & Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai 201321, China.
| | - Ye Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jin-Mei Jin
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Hong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yun Sun
- Department of Research and Development & Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai 201321, China.
| | - Hong-Zhuan Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China. and Department of Pharmacology, Shanghai Jiao Tong University School of Medicine, W. Building 3, Room 407, 280 Chongqing Road, Shanghai, 200025, China
| | - Xin Luan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
50
|
Wu H, Jiang Q, Luo K, Zhu C, Xie M, Wang S, Fei Z, Zhao J. Synthesis of iridium-based nanocomposite with catalase activity for cancer phototherapy. J Nanobiotechnology 2021; 19:203. [PMID: 34233696 PMCID: PMC8265148 DOI: 10.1186/s12951-021-00948-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/29/2021] [Indexed: 11/10/2022] Open
Abstract
The combination of photothermal therapy (PTT) and photodynamic therapy (PDT) has attracted attention due to its enhanced tumor therapy effect. This study proposes a novel nanoenzyme-based theranostic nanoplatform, IrO2@MSN@PDA-BSA(Ce6), for the combined PTT and PDT of tumors. IrO2 was prepared by a simple hydrolysis method and coated with a thin layer of mesoporous silica (MSN) to facilitate the physical adsorption of Chlorin e6 (Ce6). The PDA coating and IrO2 NPs of the nanoplatform demonstrated an improved photothermal conversion efficiency of 29.8% under NIR irradiation. Further, the Ce6 loading imparts materials with the ability to produce reactive oxygen species (ROS) under 660 nm NIR laser irradiation. It was also proved that the IrO2 NPs could catalyze the hydrogen peroxide (H2O2) in the tumor microenvironment (TME) to generate endogenous oxygen (O2), thereby enhancing the efficiency of PDT. The in vitro and in vivo experiments indicated that the nanocomposite was highly biocompatible and could produce a satisfactory tumor therapeutic effect. Thus, the findings of the present study demonstrate the viability of using theranostic nanoenzymes for translational medicine.
Collapse
Affiliation(s)
- Hang Wu
- Department of Breast Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200433, People's Republic of China
| | - Qi Jiang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, No. 168 Changhai Road, Shanghai, 200433, People's Republic of China
| | - Keyi Luo
- College of Science, University of Shanghai for Science and Technology, No. 334 Jungong Road, Shanghai, 200093, People's Republic of China
| | - Chunping Zhu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, No. 168 Changhai Road, Shanghai, 200433, People's Republic of China
| | - Mengmeng Xie
- College of Science, University of Shanghai for Science and Technology, No. 334 Jungong Road, Shanghai, 200093, People's Republic of China
| | - Shige Wang
- College of Science, University of Shanghai for Science and Technology, No. 334 Jungong Road, Shanghai, 200093, People's Republic of China
| | - Zhewei Fei
- Department of Breast Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200433, People's Republic of China.
| | - Jiulong Zhao
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, No. 168 Changhai Road, Shanghai, 200433, People's Republic of China.
| |
Collapse
|