1
|
Fan Y, Zhang R, Shi J, Tian F, Zhang Y, Zhang L, Liao G, Yang M. Mild near-infrared laser-triggered photo-immunotherapy potentiates immune checkpoint blockade via an all-in-one theranostic nanoplatform. J Colloid Interface Sci 2025; 678:1088-1103. [PMID: 39276517 DOI: 10.1016/j.jcis.2024.09.020] [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: 07/04/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024]
Abstract
One of the primary challenges for immune checkpoint blockade (ICB)-based therapy is the limited infiltration of T lymphocytes (T cells) into tumors, often referred to as immunologically "cold" tumors. A promising strategy to enhance the anti-tumor efficacy of ICB is to increase antigen exposure, thereby enhancing T cell activation and converting "cold" tumors into "hot" ones. Herein, we present an innovative all-in-one therapeutic nanoplatform to realize local mild photothermal- and photodynamic-triggered antigen exposure, thereby improving the anti-tumor efficacy of ICB. This nanoplatform involves conjugating programmed death-ligand 1 antibody (aPD-L1) with gadolinium-doped near-infrared (NIR)-emitting carbon dots (aPD-L1@GdCDs), which displays negligible cytotoxicity in the absence of light. But under controlled NIR laser irradiation, the GdCDs produce combined photothermal and photodynamic effects. This not only results in tumor ablation but also induces immunogenic cell death (ICD), facilitating enhanced infiltration of CD8+ T cells in the tumor area. Importantly, the combination of aPD-L1 with photothermal and photodynamic therapies via aPD-L1@GdCDs significantly boosts CD8+ T cell infiltration, reduces tumor size, and improves anti-metastasis effects compared to either GdCDs-based phototherapy or aPD-L1 alone. In addition, the whole treatment process can be monitored by multi-modal fluorescence/photoacoustic/magnetic resonance imaging (FLI/PAI/MRI). Our study highlights a promising nanoplatform for cancer diagnosis and therapy, as well as paves the way to promote the efficacy of ICB therapy through mild photothermal- and photodynamic-triggered immunotherapy.
Collapse
Affiliation(s)
- Yadi Fan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Ruolin Zhang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Jingyu Shi
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Feng Tian
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Yu Zhang
- Department of Mechanical and Automotive Engineering, Royal Melbourne Institute of Technology, Melbourne, VIC 3000, Australia
| | - Li Zhang
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan 430200, China.
| | - Guangfu Liao
- College of Material Engineering, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| | - Mo Yang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China; The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Joint Research Center of Biosensing and Precision Theranostics, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China.
| |
Collapse
|
2
|
Xiong G, Chen Q, Wang Q, Wang X, Xiao Y, Jin L, Yan K, Zhang X, Hu F. Multifaceted role of nanocomposite hydrogels in diabetic wound healing: enhanced biomedical applications and detailed molecular mechanisms. Biomater Sci 2024; 12:6196-6223. [PMID: 39494707 DOI: 10.1039/d4bm01088d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
The complex microenvironment of diabetic wounds, which is characterized by persistent hyperglycemia, excessive inflammatory responses, and hypoxic conditions, significantly impedes the efficacy of traditional hydrogels. Nanocomposite hydrogels, which combine the high-water content and biocompatibility of hydrogels with the unique functionalities of nanomaterials, offer a promising solution. These hydrogels exhibit enhanced antibacterial, antioxidant, and drug-release properties. Incorporating nanomaterials increases the mechanical strength and bioactivity of hydrogels, allowing for dynamic regulation of the wound microenvironment and promoting cell migration, proliferation, and angiogenesis, thereby accelerating wound healing. This review provides a comprehensive overview of the latest advances in nanocomposite hydrogels for diabetic wound treatment and discusses their advantages and molecular mechanisms at various healing stages. The study aims to provide a theoretical foundation and practical guidance for future research and clinical applications. Furthermore, it highlights the challenges related to the mechanical durability, antimicrobial performance, resistance issues, and interactions with the cellular environments of these hydrogels. Future directions include optimizing smart drug delivery systems and personalized medical approaches to enhance the clinical applicability of nanocomposite hydrogels.
Collapse
Affiliation(s)
- Gege Xiong
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, PR China.
| | - Qiwei Chen
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Qiuyu Wang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, PR China.
| | - Xiaoxue Wang
- Department of Stomatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan 528000, PR China.
| | - Yaomu Xiao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, PR China.
| | - Liuli Jin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, PR China.
| | - Kaichong Yan
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, PR China.
| | - Xueyang Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, PR China.
- Department of Stomatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan 528000, PR China.
| | - Fei Hu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, PR China.
| |
Collapse
|
3
|
Jiang ZB, Xu C, Xu P, Huang DH, Kang LP. Lycorine Suppresses Non-Small-Cell Lung Cancer Progression Through Activating STING Pathway and Stimulating an Antitumor Immune Response. Chem Biol Drug Des 2024; 104:e70036. [PMID: 39707625 DOI: 10.1111/cbdd.70036] [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: 07/08/2024] [Revised: 11/29/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024]
Abstract
Non-small-cell lung cancer (NSCLC) stands as a primary contributor to cancer-related deaths worldwide. It has been demonstrated that Lycorine (LYD), a naturally occurring active sesquiterpene present in Chinese medicinal plants, exhibits anti-cancer properties across various cancer cell lines. However, the underlying mechanisms of LYD-induced anti-tumor in NSCLC are not fully known. This study demonstrated that LYD significantly reduced the proliferation of NSCLC and induced apoptosis by increasing intracellular ROS levels. The inhibition of ROS using N-acetylcysteine (NAC) eliminated the apoptosis effects of LYD, resulting in increased cell viability. Additionally, LYD treatment significantly activated the STING pathway in NSCLC and induced the expression of CXCL10, CXCL9 and CCL5 in NSCLC cells. Mechanistically, LYD was found to significantly reduce the protein levels of P70S6K and S6K, which are key proteins involved in cell growth and survival. Notably, in vivo experiments demonstrated that LYD significantly inhibited the growth of H358 xenograft and LLC1 tumor, exhibiting anti-tumor activity by elevating CD8+ T cells in the NSCLC mouse model. Our findings suggest that LYD possesses potent anti-cancer properties in NSCLC by inducing apoptosis through ROS generation and modulating the STING pathway and key chemokines. Furthermore, LYD also exerts its antitumor effects by inhibiting crucial proteins involved in cell growth. Overall, LYD shows promise as a potential therapeutic agent for NSCLC treatment.
Collapse
Affiliation(s)
- Ze-Bo Jiang
- Zhuhai Hospital of Integrated Traditional Chinese & Western Medicine, Zhuhai, Guangdong, China
| | - Cong Xu
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Pan Xu
- Zhuhai Hospital of Integrated Traditional Chinese & Western Medicine, Zhuhai, Guangdong, China
| | - Dong-Hui Huang
- Zhuhai Hospital of Integrated Traditional Chinese & Western Medicine, Zhuhai, Guangdong, China
| | - Li-Ping Kang
- Zhuhai Hospital of Integrated Traditional Chinese & Western Medicine, Zhuhai, Guangdong, China
| |
Collapse
|
4
|
Shrestha P, Duwa R, Lee S, Kwon TK, Jeong JH, Yook S. ROS-responsive thioketal nanoparticles delivering system for targeted ulcerative colitis therapy with potent HDAC6 inhibitor, tubastatin A. Eur J Pharm Sci 2024; 201:106856. [PMID: 39032536 DOI: 10.1016/j.ejps.2024.106856] [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: 03/05/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Ulcerative colitis (UC) is a common gastrointestinal problem characterized by the mucosal injury primarily affecting the large intestine. Currently available therapies are not satisfactory as evidenced by high relapse rate and adverse effects. In this study we aimed to develop an effective drug delivery system using reactive oxygen species (ROS)-responsive thioketal nanoparticles (TKNP), to deliver tubastatin A, a potent HDAC6 inhibitor, to the inflamed colon in mice with ulcerative colitis (UC). TKNPs were synthesized by step-growth polymerization from an acetal exchange reaction while TUBA-TKNP was prepared using the single emulsion solvent evaporation technique. Our developed nanoparticle showed release of tubastatin A only in presence of ROS which is found to be highly present at the site of inflamed colon. Oral administration of TUBA-TKNP resulted in the higher accumulation of tubastatin A at the inflamed colon site and decreased the inflammation as evidenced by reduced infiltration of immune cells and decreased level of pro-inflammatory cytokines in TUBA-TKNP treated mice. In summary, our results show the successful localization of tubastatin A at the site of colon inflammation through TUBA-TKNP delivery, as well as resolution of clinical features of UC in mice.
Collapse
Affiliation(s)
- Prabhat Shrestha
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ramesh Duwa
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Radiology, Molecular Imaging Program at Standford (MIPS), School of Medicine, Standford University, Standford, California 94305, USA
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu, 42601, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Simmyung Yook
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
5
|
Zhou Z, Wang J, Xu X, Wang Z, Mao L, Zhang S, Zhang H, Li Y, Yu Q, Jiang N, Zhang G, Gan Z, Ning Z. Lignin-Based Nanoparticles for Combination of Tumor Oxidative Stress Amplification and Reactive Oxygen Species Responsive Drug Release. Bioconjug Chem 2024; 35:1207-1217. [PMID: 38989881 DOI: 10.1021/acs.bioconjchem.4c00261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
In this study, maleic anhydride-modified lignin (LG-M), a ROS-cleavable thioketal (TK) bond, and polyethylene glycol (PEG) were used to synthesize a lignin-based copolymer (LG-M(TK)-PEG). Doxorubicin (DOX) was attached to the ROS-cleavable bond in the LG-M(TK)-PEG for the preparation of the ROS-activatable DOX prodrug (LG-M(TK-DOX)-PEG). Nanoparticles (NPs) with a size of 125.7 ± 3.1 nm were prepared by using LG-M(TK-DOX)-PEG, and they exhibited enhanced uptake by cancer cells compared to free DOX. Notably, the presence of lignin in the nanoparticles could boost ROS production in breast cancer 4T1 cells while showing little effect on L929 normal cells. This selective effect facilitated the specific activation of the DOX prodrug in the tumor microenvironment, resulting in the superior tumor inhibitory effects and enhanced biosafety relative to free DOX. This work demonstrates the potential of the LG-M(TK-DOX)-PEG NPs as an efficient drug delivery system for cancer treatment.
Collapse
Affiliation(s)
- Ziwei Zhou
- Beijing Laboratory of Biomedical Materials, State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, No. 15 Bei San Huan Dong Lu, Chaoyang District, Beijing 100029, China
| | - Jin Wang
- Beijing Laboratory of Biomedical Materials, State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, No. 15 Bei San Huan Dong Lu, Chaoyang District, Beijing 100029, China
| | - Xin Xu
- Department of Urology, China Japan Friendship Hospital, Beijing 100029, China
| | - Zhuang Wang
- Beijing Laboratory of Biomedical Materials, State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, No. 15 Bei San Huan Dong Lu, Chaoyang District, Beijing 100029, China
| | - Lingchen Mao
- Beijing Laboratory of Biomedical Materials, State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, No. 15 Bei San Huan Dong Lu, Chaoyang District, Beijing 100029, China
| | - Shanhu Zhang
- Beijing Laboratory of Biomedical Materials, State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, No. 15 Bei San Huan Dong Lu, Chaoyang District, Beijing 100029, China
| | - Huanhuan Zhang
- Department of General Medicine, the Fourth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Yuqiang Li
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, China
| | - Qingsong Yu
- Beijing Laboratory of Biomedical Materials, State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, No. 15 Bei San Huan Dong Lu, Chaoyang District, Beijing 100029, China
| | - Ni Jiang
- Beijing Laboratory of Biomedical Materials, State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, No. 15 Bei San Huan Dong Lu, Chaoyang District, Beijing 100029, China
| | - Guan Zhang
- Department of Urology, China Japan Friendship Hospital, Beijing 100029, China
| | - Zhihua Gan
- Beijing Laboratory of Biomedical Materials, State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, No. 15 Bei San Huan Dong Lu, Chaoyang District, Beijing 100029, China
| | - Zhenbo Ning
- Beijing Laboratory of Biomedical Materials, State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, No. 15 Bei San Huan Dong Lu, Chaoyang District, Beijing 100029, China
| |
Collapse
|
6
|
Li Y, Xin X, Zhou X, Liu J, Liu H, Yuan S, Liu H, Hao W, Sun J, Wang Y, Gong W, Yang M, Li Z, Han Y, Gao C, Yang Y. ROS-responsive biomimetic nanosystem camouflaged by hybrid membranes of platelet-exosomes engineered with neuronal targeting peptide for TBI therapy. J Control Release 2024; 372:531-550. [PMID: 38851535 DOI: 10.1016/j.jconrel.2024.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Recovery and survival following traumatic brain injury (TBI) depends on optimal amelioration of secondary injuries at lesion site. Delivering mitochondria-protecting drugs to neurons may revive damaged neurons at sites secondarily traumatized by TBI. Pioglitazone (PGZ) is a promising candidate for TBI treatment, limited by its low brain accumulation and poor targetability to neurons. Herein, we report a ROS-responsive nanosystem, camouflaged by hybrid membranes of platelets and engineered extracellular vesicles (EVs) (C3-EPm-|TKNPs|), that can be used for targeted delivery of PGZ for TBI therapy. Inspired by intrinsic ability of macrophages for inflammatory chemotaxis, engineered M2-like macrophage-derived EVs were constructed by fusing C3 peptide to EVs membrane integrator protein, Lamp2b, to confer them with ability to target neurons in inflamed lesions. Platelets provided hybridized EPm with capabilities to target hemorrhagic area caused by trauma via surface proteins. Consequently, C3-EPm-|PGZ-TKNPs| were orientedly delivered to neurons located in the traumatized hemisphere after intravenous administration, and triggered the release of PGZ from TKNPs via oxidative stress. The current work demonstrate that C3-EPm-|TKNPs| can effectively deliver PGZ to alleviate mitochondrial damage via mitoNEET for neuroprotection, further reversing behavioral deficits in TBI mice. Our findings provide proof-of-concept evidence of C3-EPm-|TKNPs|-derived nanodrugs as potential clinical approaches against neuroinflammation-related intracranial diseases.
Collapse
Affiliation(s)
- Yi Li
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Xin Xin
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Xun Zhou
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China; College of Pharmacy, Henan University, Kaifeng 475000, People's Republic of China
| | - Jingzhou Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Hangbing Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China; School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Shuo Yuan
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China; School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Hanhan Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Wenyan Hao
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Jiejie Sun
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Yuli Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Wei Gong
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Meiyan Yang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Zhiping Li
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Yang Han
- School of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| | - Chunsheng Gao
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China.
| | - Yang Yang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China.
| |
Collapse
|
7
|
Zhu S, Huo L, Zeng J, Chen R, Sun Y, Tan M, Fan M, Liu M, Zhao J, Huang G, Wang Y, Xiao Z, Zhao Z. Differentiated management of ROS level in tumor and kidney to alleviate Cis-platinum induced acute kidney injury with improved efficacy. J Nanobiotechnology 2024; 22:436. [PMID: 39044240 PMCID: PMC11267679 DOI: 10.1186/s12951-024-02710-2] [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: 01/20/2024] [Accepted: 07/05/2024] [Indexed: 07/25/2024] Open
Abstract
Cisplatin (DDP) is a prevalent chemotherapeutic agent used in tumor therapy, yet DDP-induced acute kidney injury (AKI) severely limits its clinical application. Antioxidants as reactive oxygen species (ROS) scavengers can circumvent this adverse effect while leading to the decrease of efficacy to tumor. Herein, we report ultrasmall ruthenium nanoparticles (URNPs) as switchable ROS scavengers/generators to alleviate DDP-induced AKI and improve its therapeutic efficacy. In the physiological environment of the kidney, URNPs mimic multi-enzyme activities, such as superoxide dismutase and catalase, effectively protecting the renal cell and tissue by down-regulating the increased ROS level caused by DDP and alleviating AKI. Specifically, URNPs are oxidized by high levels of H2O2 in the tumor microenvironment (TME), resulting in the generation of oxygen vacancies and Ru3+/Ru4+ ions. This unique structure transformation endows URNPs to generate singlet oxygen (1O2) under laser irradiation and hydroxyl radicals (∙OH) through a Fenton-like reaction in tumor cell and tissue. The simultaneous generation of multifarious ROS effectively improves the efficacy of DDP in vitro and in vivo. This TME-responsive ROS scavenger/generator acts as an adjuvant therapeutic agent to minimize side effects and improve the efficacy of chemotherapy drugs, providing a new avenue to chemotherapy and facilitating clinical tumor therapy.
Collapse
Affiliation(s)
- Shiqi Zhu
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Linlin Huo
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Jie Zeng
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Rong Chen
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Yutong Sun
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Mingya Tan
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Mengke Fan
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Meiling Liu
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Jiayi Zhao
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Guoming Huang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Yi Wang
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Zhibo Xiao
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China.
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Zhenghuan Zhao
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
8
|
Bertoncini B, Xiao Z, Zacchini S, Biancalana L, Gasser G, Marchetti F. Aminocarbyne-Alkyne Coupling in Diruthenium Complexes: Exploring the Anticancer Potential of the Resulting Vinyliminium Complexes and Comparison with Diiron Homologues. Inorg Chem 2024; 63:12485-12497. [PMID: 38912873 DOI: 10.1021/acs.inorgchem.4c01119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
New diruthenium complexes based on the scaffold Ru2Cp2(CO)2 (Cp = η5-C5H5) and containing a bridging vinyliminium ligand, [2a-d]CF3SO3, were synthesized through regioselective coupling of alkynes with an aminocarbyne precursor (85-90% yields). The reaction involving phenylacetylene proceeded with the formation of a diruthenacyclobutene byproduct, [4]CF3SO3 (10% yield). Complexes [2a-d]+ undergo partial alkyne extrusion in contact with alumina or CDCl3. All products were characterized by elemental analysis, infrared and multinuclear NMR spectroscopy, and single crystal X-ray diffraction in two cases. Complexes [2a-d]+ revealed an outstanding stability in DMEM cell culture medium at 37 °C (<1% degradation over 72 h). These complexes exhibited cytotoxicity in human colon colorectal adenocarcinoma HT-29 cells in the low micromolar range, with lower IC50 values than those obtained with the homologous diiron complexes previously reported. Evaluation of ROS (reactive oxygen species) production and O2 consumption rate (OCR) highlighted the higher potential of Ru2 complexes, compared to the Fe2 counterparts, to impact mitochondrial activity, with the heterometallic Ru2-ferrocenyl complex [2d]+ showing the best performance.
Collapse
Affiliation(s)
- Benedetta Bertoncini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy
| | - Zhimei Xiao
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health, 11 Rue Pierre et Marie Curie, 75005 Paris, France
| | - Stefano Zacchini
- Department of Industrial Chemistry "Toso Montanari", University of Bologna, Via P. Gobetti 85, I-40129 Bologna, Italy
| | - Lorenzo Biancalana
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health, 11 Rue Pierre et Marie Curie, 75005 Paris, France
| | - Fabio Marchetti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy
| |
Collapse
|
9
|
Tang L, Wang YJ, Wang YY, Li ST, Kong L, Li XT, Ma LL, Liu XX. Construction of ROS-Responsive Hyaluronic Acid Modified Paclitaxel and Diosgenin Liposomes and Study on Synergistic Enhancement of Anti-Ovarian Cancer Efficacy. Int J Nanomedicine 2024; 19:5193-5211. [PMID: 38859958 PMCID: PMC11162966 DOI: 10.2147/ijn.s455942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024] Open
Abstract
Purpose Ovarian cancer is a fatal gynecologic malignancy with a high rate of abdominal metastasis. Chemotherapy still has a poor clinical prognosis for ovarian cancer patients, with cell proliferation and angiogenesis leading to invasion, migration, and recurrence. To overcome these obstacles, we constructed a novel HA-modified paclitaxel and diosgenin liposome (PEG-TK-HA-PDLPs) using two novel functional materials, DSPE-PEG2000-HA and DSPE-PEG2000-TK-PEG5000, to specifically deliver the drugs to the tumor site in order to reduce OC cell proliferation and anti-angiogenic generation, thereby inhibiting invasion and migration. Methods and Results PEG-TK-HA-PDLPs were prepared by film dispersion, with ideal physicochemical properties and exhibits active targeting for enhanced cellular uptake. The ZIP synergy score for PTX and Dios was calculated using the online SynergyFinder software to be 3.15, indicating synergy. In vitro results showed that PEG-TK-HA-PDLPs were highly cytotoxic to ID8 cells, induced ID8 cell apoptosis, and inhibited ID8 cell migration and invasion. In vivo studies showed that PEG-TK-HA-PDLPs could prolong the circulation time in the blood, accumulate significantly in the tumor site, and effectively fight against angiogenesis with significant anti-tumor effects. Conclusion The production of PEG-TK-HA-PDLPs is an effective strategy for the treatment of OC.
Collapse
Affiliation(s)
- Ling Tang
- Department of Obstetrics and Gynecology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Yu-Jia Wang
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Yuan-Yuan Wang
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Shu-Tong Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, 116600, People’s Republic of China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, 116600, People’s Republic of China
| | - Xue-Tao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, 116600, People’s Republic of China
| | - Ling-Ling Ma
- Department of Obstetrics and Gynecology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Xiu-Xiu Liu
- Department of Obstetrics and Gynecology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| |
Collapse
|
10
|
Tu P, Pan Y, Wang L, Li B, Sun X, Liang Z, Liu M, Zhao Z, Wu C, Wang J, Wang Z, Song Y, Zhang Y, Ma Y, Guo Y. CD62E- and ROS-Responsive ETS Improves Cartilage Repair by Inhibiting Endothelial Cell Activation through OPA1-Mediated Mitochondrial Homeostasis. Biomater Res 2024; 28:0006. [PMID: 38439927 PMCID: PMC10911934 DOI: 10.34133/bmr.0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/03/2024] [Indexed: 03/06/2024] Open
Abstract
Background: In the environment of cartilage injury, the activation of vascular endothelial cell (VEC), marked with excessive CD62E and reactive oxygen species (ROS), can affect the formation of hyaluronic cartilage. Therefore, we developed a CD62E- and ROS-responsive drug delivery system using E-selectin binding peptide, Thioketal, and silk fibroin (ETS) to achieve targeted delivery and controlled release of Clematis triterpenoid saponins (CS) against activated VEC, and thus promote cartilage regeneration. Methods: We prepared and characterized ETS/CS and verified their CD62E- and ROS-responsive properties in vitro. We investigated the effect and underlying mechanism of ETS/CS on inhibiting VEC activation and promoting chondrogenic differentiation of bone marrow stromal cells (BMSCs). We also analyzed the effect of ETS/CS on suppressing the activated VEC-macrophage inflammatory cascade in vitro. Additionally, we constructed a rat knee cartilage defect model and administered ETS/CS combined with BMSC-containing hydrogels. We detected the cartilage differentiation, the level of VEC activation and macrophage in the new tissue, and synovial tissue. Results: ETS/CS was able to interact with VEC and inhibit VEC activation through the carried CS. Coculture experiments verified ETS/CS promoted chondrogenic differentiation of BMSCs by inhibiting the activated VEC-induced inflammatory cascade of macrophages via OPA1-mediated mitochondrial homeostasis. In the rat knee cartilage defect model, ETS/CS reduced VEC activation, migration, angiogenesis in new tissues, inhibited macrophage infiltration and inflammation, promoted chondrogenic differentiation of BMSCs in the defective areas. Conclusions: CD62E- and ROS-responsive ETS/CS promoted cartilage repair by inhibiting VEC activation and macrophage inflammation and promoting BMSC chondrogenesis. Therefore, it is a promising therapeutic strategy to promote articular cartilage repair.
Collapse
Affiliation(s)
- Pengcheng Tu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Yalan Pan
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Lining Wang
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Bin Li
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Xiaoxian Sun
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Zhongqing Liang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education,
Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Acupuncture and Tuina, School of Health and Rehabilitation,
Nanjing University of Chinese Medicine, Nanjing 210046, Jiangsu, China
| | - Mengmin Liu
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Zitong Zhao
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Chengjie Wu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Jianwei Wang
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease,
Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi 214072, P.R. China
| | - Zhifang Wang
- Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, P.R. China
| | - Yu Song
- Zhangjiagang First People’s Hospital Affiliated to Soochow University, Zhangjiagang 215638, P.R. China
| | - Yafeng Zhang
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease,
Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi 214072, P.R. China
| | - Yong Ma
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease,
Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi 214072, P.R. China
| | - Yang Guo
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease,
Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi 214072, P.R. China
| |
Collapse
|
11
|
Liu Z, Liu X, Zhang W, Gao R, Wei H, Yu CY. Current advances in modulating tumor hypoxia for enhanced therapeutic efficacy. Acta Biomater 2024; 176:1-27. [PMID: 38232912 DOI: 10.1016/j.actbio.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/08/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
Hypoxia is a common feature of most solid tumors, which promotes the proliferation, invasion, metastasis, and therapeutic resistance of tumors. Researchers have been developing advanced strategies and nanoplatforms to modulate tumor hypoxia to enhance therapeutic effects. A timely review of this rapidly developing research topic is therefore highly desirable. For this purpose, this review first introduces the impact of hypoxia on tumor development and therapeutic resistance in detail. Current developments in the construction of various nanoplatforms to enhance tumor treatment in response to hypoxia are also systematically summarized, including hypoxia-overcoming, hypoxia-exploiting, and hypoxia-disregarding strategies. We provide a detailed discussion of the rationale and research progress of these strategies. Through a review of current trends, it is hoped that this comprehensive overview can provide new prospects for clinical application in tumor treatment. STATEMENT OF SIGNIFICANCE: As a common feature of most solid tumors, hypoxia significantly promotes tumor progression. Advanced nanoplatforms have been developed to modulate tumor hypoxia to enhanced therapeutic effects. In this review, we first introduce the impact of hypoxia on tumor progression. Current developments in the construction of various nanoplatforms to enhance tumor treatment in response to hypoxia are systematically summarized, including hypoxia-overcoming, hypoxia-exploiting, and hypoxia-disregarding strategies. We discuss the rationale and research progress of the above strategies in detail, and finally introduce future challenges for treatment of hypoxic tumors. By reviewing the current trends, this comprehensive overview can provide new prospects for clinical translatable tumor therapy.
Collapse
Affiliation(s)
- Zihan Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xinping Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Wei Zhang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ruijie Gao
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
12
|
Zhou Z, Wang H, Li J, Jiang X, Li Z, Shen J. Recent progress, perspectives, and issues of engineered PD-L1 regulation nano-system to better cure tumor: A review. Int J Biol Macromol 2024; 254:127911. [PMID: 37939766 DOI: 10.1016/j.ijbiomac.2023.127911] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 10/29/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Currently, immune checkpoint blockade (ICB) therapies that target the programmed cell death ligand-1 (PD-L1) have been used as revolutionary cancer treatments in the clinic. Apart from restoring the antitumor response of cytotoxic T cells by blocking the interaction between PD-L1 on tumor cells and programmed cell death-1 (PD-1) on T cells, PD-L1 proteins were also newly revealed to possess the capacity to accelerate DNA damage repair (DDR) and enhance tumor growth through multiple mechanisms, leading to the impaired efficacy of tumor therapies. Nevertheless, current free anti-PD-1/PD-L1 therapy still suffered from poor therapeutic outcomes in most solid tumors due to the non-selective tumor accumulation, ineludible severe cytotoxic effects, as well as the common occurrence of immune resistance. Recently, nanoparticles with efficient tumor-targeting capacity, tumor-responsive prosperity, and versatility for combination therapy were identified as new avenues for PD-L1 targeting cancer immunotherapies. In this review, we first summarized the multiple functions of PD-L1 protein in promoting tumor growth, accelerating DDR, as well as depressing immunotherapy efficacy. Following this, the effects and mechanisms of current clinically widespread tumor therapies on tumor PD-L1 expression were discussed. Then, we reviewed the recent advances in nanoparticles for anti-PD-L1 therapy via using PD-L1 antibodies, small interfering RNA (siRNA), microRNA (miRNA), clustered, regularly interspaced, short palindromic repeats (CRISPR), peptide, and small molecular drugs. At last, we discussed the challenges and perspectives to promote the clinical application of nanoparticles-based PD-L1-targeting therapy.
Collapse
Affiliation(s)
- Zaigang Zhou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Haoxiang Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jie Li
- College of Pharmacy, Wenzhou Medical University, Wenzhou 325000, China
| | - Xin Jiang
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhangping Li
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang 324000, China.
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China.
| |
Collapse
|
13
|
Nguyen TTK, Pham KY, Yook S. Engineered therapeutic proteins for sustained-release drug delivery systems. Acta Biomater 2023; 171:131-154. [PMID: 37717712 DOI: 10.1016/j.actbio.2023.09.018] [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: 03/01/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 09/19/2023]
Abstract
Proteins play a vital role in diverse biological processes in the human body, and protein therapeutics have been applied to treat different diseases such as cancers, genetic disorders, autoimmunity, and inflammation. Protein therapeutics have demonstrated their advantages, such as specific pharmaceutical effects, low toxicity, and strong solubility. However, several disadvantages arise in clinical applications, including short half-life, immunogenicity, and low permeation, leading to reduced drug effectiveness. The structure of protein therapeutics can be modified to increase molecular size, leading to prolonged stability and increased plasma half-life. Notably, the controlled-release delivery systems for the sustained release of protein drugs and preserving the stability of cargo proteins are envisioned as a potential approach to overcome these challenges. In this review, we summarize recent research progress related to structural modifications (PEGylation, glycosylation, poly amino acid modification, and molecular biology-based strategies) and promising long-term delivery systems, such as polymer-based systems (injectable gel/implants, microparticles, nanoparticles, micro/nanogels, functional polymers), lipid-based systems (liposomes, solid lipid nanoparticles, nanostructured lipid carriers), and inorganic nanoparticles exploited for protein therapeutics. STATEMENT OF SIGNIFICANCE: In this review, we highlight recent advances concerning modifying proteins directly to enhance their stability and functionality and discuss state-of-the-art methods for the delivery and controlled long-term release of active protein therapeutics to their target site. In terms of drug modifications, four widely used strategies, including PEGylation, poly amino acid modification, glycosylation, and genetic, are discussed. As for drug delivery systems, we emphasize recent progress relating to polymer-based systems, lipid-based systems developed, and inorganic nanoparticles for protein sustained-release delivery. This review points out the areas requiring focused research attention before the full potential of protein therapeutics for human health and disease can be realized.
Collapse
Affiliation(s)
- Thoa Thi Kim Nguyen
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea
| | - Khang-Yen Pham
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea.
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
14
|
Banstola A, Pandit M, Duwa R, Chang J, Jeong J, Yook S. Reactive oxygen species-responsive dual-targeted nanosystem promoted immunogenic cell death against breast cancer. Bioeng Transl Med 2023; 8:e10379. [PMID: 37693071 PMCID: PMC10487313 DOI: 10.1002/btm2.10379] [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: 03/28/2022] [Revised: 07/05/2022] [Accepted: 07/16/2022] [Indexed: 09/12/2023] Open
Abstract
The development of an optimal treatment modality to improve the therapeutic outcome of breast cancer patients is still difficult. Poor antigen presentation to T cells is a major challenge in cancer immunotherapy. In this study, a synergistic immunotherapy strategy for breast cancer incorporating immune cell infiltration, immunogenic cell death (ICD), and dendritic cell (DC) maturation through a reactive oxygen species (ROS)-responsive dual-targeted smart nanosystem (anti-PD-L1-TKNP) for the simultaneous release of DOX, R848, and MIP-3α in the tumor microenvironment is reported. Following local injection, anti-PD-L1-DOX-R848-MIP-3α/thioketal nanoparticle (TKNP) converts tumor cells to a vaccine owing to the combinatorial effect of DOX-induced ICD, R848-mediated immunostimulatory properties, and MIP-3α-induced immune cell recruitment in the tumor microenvironment. Intratumoral injection of anti-PD-L1-DOX-R848-MIP-3α/TKNP caused significant regression of breast cancer. Mechanistic studies reveal that anti-PD-L1-DOX-R848-MIP-3α/TKNP specifically targets tumor tissue, resulting in maximum exposure of calreticulin (CRT) and HMGB1 in tumors, and significantly enhances intratumoral infiltration of CD4+ and CD8+ T cells in tumors. Therefore, a combined strategy using dual-targeted ROS-responsive TKNP highlights the significant application of nanoparticles in modulating the tumor microenvironment and could be a clinical treatment strategy for effective breast cancer management.
Collapse
Affiliation(s)
- Asmita Banstola
- College of PharmacyKeimyung UniversityDaeguSouth Korea
- Department of Dermatology, Harvard Medical SchoolWellman Center for Photomedicine, Massachusetts General HospitalBostonMassachusettsUSA
| | - Mahesh Pandit
- College of PharmacyYeungnam UniversityGyeongsanGyeongbukSouth Korea
| | - Ramesh Duwa
- College of PharmacyKeimyung UniversityDaeguSouth Korea
| | - Jae‐Hoon Chang
- College of PharmacyYeungnam UniversityGyeongsanGyeongbukSouth Korea
| | - Jee‐Heon Jeong
- Department of Precision Medicine, School of MedicineSungkyunkwan UniversitySuwonSouth Korea
| | - Simmyung Yook
- College of PharmacyKeimyung UniversityDaeguSouth Korea
| |
Collapse
|
15
|
Adjei-Sowah E, Benoit DSW, Loiselle AE. Drug Delivery Approaches to Improve Tendon Healing. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:369-386. [PMID: 36888543 PMCID: PMC10442691 DOI: 10.1089/ten.teb.2022.0188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/18/2023] [Indexed: 03/09/2023]
Abstract
Tendon injuries disrupt the transmission of forces from muscle to bone, leading to chronic pain, disability, and a large socioeconomic burden. Tendon injuries are prevalent; there are over 300,000 tendon repair procedures a year in the United States to address acute trauma or chronic tendinopathy. Successful restoration of function after tendon injury remains challenging clinically. Despite improvements in surgical and physical therapy techniques, the high complication rate of tendon repair procedures motivates the use of therapeutic interventions to augment healing. While many biological and tissue engineering approaches have attempted to promote scarless tendon healing, there is currently no standard clinical treatment to improve tendon healing. Moreover, the limited efficacy of systemic delivery of several promising therapeutic candidates highlights the need for tendon-specific drug delivery approaches to facilitate translation. This review article will synthesize the current state-of-the-art methods that have been used for tendon-targeted delivery through both systemic and local treatments, highlight emerging technologies used for tissue-specific drug delivery in other tissue systems, and outline future challenges and opportunities to enhance tendon healing through targeted drug delivery.
Collapse
Affiliation(s)
- Emmanuela Adjei-Sowah
- Department of Biomedical Engineering and University of Rochester, Rochester, New York, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Danielle S. W. Benoit
- Department of Biomedical Engineering and University of Rochester, Rochester, New York, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Cell Biology of Disease Program, University of Rochester, Rochester, New York, USA
- Department of Chemical Engineering, University of Rochester, Rochester, New York, USA
- Materials Science Program, University of Rochester, Rochester, New York, USA
- Knight Campus Department of Bioengineering, University of Oregon, Eugene, Oregan, USA
| | - Alayna E. Loiselle
- Department of Biomedical Engineering and University of Rochester, Rochester, New York, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
- Cell Biology of Disease Program, University of Rochester, Rochester, New York, USA
| |
Collapse
|
16
|
Tian Z, Wu X, Peng L, Yu N, Gou G, Zuo W, Yang J. pH-responsive bufadienolides nanocrystals decorated by chitosan quaternary ammonium salt for treating colon cancer. Int J Biol Macromol 2023; 242:124819. [PMID: 37178894 DOI: 10.1016/j.ijbiomac.2023.124819] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/27/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023]
Abstract
Due to its poor prognosis and propensity for metastasizing, colon cancer, a frequent cancer of the gastrointestinal system, has a high morbidity and mortality rate. However, the harsh physiological conditions of the gastrointestinal tract can cause the anti-cancer medicine bufadienolides (BU) to lose some of its structure, impairing its ability to fight cancer. In this study, pH-responsive bufadienolides nanocrystals decorated by chitosan quaternary ammonium salt (HE BU NCs) were successfully constructed by a solvent evaporation method to improve the bioavailability, release characteristics and intestinal transport ability of BU. In vitro, studies have shown that HE BU NCs could improve BU internalization, significantly induce apoptosis, decrease mitochondrial membrane potential, and increase ROS levels in tumour cells. In vivo, experiments showed that HE BU NCs effectively targeted intestinal sites, increased their retention time, and exerted antitumor activity through Caspase-3 and Bax/Bcl-2 ratio pathways. In conclusion, pH-responsive bufadienolides nanocrystals decorated by chitosan quaternary ammonium salt could protect bufadienolides from the destruction of an acidic environment, achieve synergistic release in the intestinal site, improve oral bioavailability, and ultimately exert anti-colon cancer effects, which is a promising strategy for the treatment of colon cancer.
Collapse
Affiliation(s)
- Zonghua Tian
- Departmert of Pharmaceutics, School of Phammacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
| | - Xia Wu
- Departmert of Pharmaceutics, School of Phammacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
| | - Li Peng
- Department of Hospital Pharmacy, General Hospital of Ningxia Medical University, No. 804 Shengli South Street, Yinchuan 750004, PR China
| | - Na Yu
- Department of Medical Chemistry, School of Basic Medicine, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
| | - Guojing Gou
- Departmert of Pharmaceutics, School of Phammacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China
| | - Wenbao Zuo
- Departmert of Pharmaceutics, School of Phammacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China.
| | - Jianhong Yang
- Departmert of Pharmaceutics, School of Phammacy, Ningxia Medical University, No. 1160 Shengli South Street, Yinchuan 750004, PR China.
| |
Collapse
|
17
|
Ma GL, Lin WF. Immune checkpoint inhibition mediated with liposomal nanomedicine for cancer therapy. Mil Med Res 2023; 10:20. [PMID: 37106400 PMCID: PMC10142459 DOI: 10.1186/s40779-023-00455-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
Immune checkpoint blockade (ICB) therapy for cancer has achieved great success both in clinical results and on the market. At the same time, success drives more attention from scientists to improve it. However, only a small portion of patients are responsive to this therapy, and it comes with a unique spectrum of side effects termed immune-related adverse events (irAEs). The use of nanotechnology could improve ICBs' delivery to the tumor, assist them in penetrating deeper into tumor tissues and alleviate their irAEs. Liposomal nanomedicine has been investigated and used for decades, and is well-recognized as the most successful nano-drug delivery system. The successful combination of ICB with liposomal nanomedicine could help improve the efficacy of ICB therapy. In this review, we highlighted recent studies using liposomal nanomedicine (including new emerging exosomes and their inspired nano-vesicles) in associating ICB therapy.
Collapse
Affiliation(s)
- Guang-Long Ma
- Faculty of Medicine, Centre for Cancer Immunology, University of Southampton, Southampton, SO16 6YD UK
| | - Wei-Feng Lin
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, 76100 Rehovot, Israel
- Key Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing, 100191 China
| |
Collapse
|
18
|
Liu N, Zhang R, Shi Q, Jiang H, Zhou Q. Intelligent delivery system targeting PD-1/PD-L1 pathway for cancer immunotherapy. Bioorg Chem 2023; 136:106550. [PMID: 37121105 DOI: 10.1016/j.bioorg.2023.106550] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 05/02/2023]
Abstract
The drugs targeting the PD-1/PD-L1 pathway have gained abundant clinical applications for cancer immunotherapy. However, only a part of patients benefit from such immunotherapy. Thus, brilliant novel tactic to increase the response rate of patients is on the agenda. Nanocarriers, particularly the rationally designed intelligent delivery systems with controllable therapeutic agent release ability and improved tumor targeting capacity, are firmly recommended. In light of this, state-of-the-art nanocarriers that are responsive to tumor-specific microenvironments (internal stimuli, including tumor acidic microenvironment, high level of GSH and ROS, specifically upregulated enzymes) or external stimuli (e.g., light, ultrasound, radiation) and release the target immunomodulators at tumor sites feature the advantages of increased anti-tumor potency but decreased off-target toxicity. Given the fantastic past achievements and the rapid developments in this field, the future is promising. In this review, intelligent delivery platforms targeting the PD-1/PD-L1 axis are attentively appraised. Specifically, mechanisms of the action of these stimuli-responsive drug release platforms are summarized to raise some guidelines for prior PD-1/PD-L1-based nanocarrier designs. Finally, the conclusion and outlook in intelligent delivery system targeting PD-1/PD-L1 pathway for cancer immunotherapy are outlined.
Collapse
Affiliation(s)
- Ning Liu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Renshuai Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Qiang Shi
- Moji-Nano Technology Co. Ltd., Yantai 264006, China
| | - Hongfei Jiang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China.
| | - Qihui Zhou
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266071, China; Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin 300038, China; Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
19
|
Li Y, Lin L, Xie J, Wei L, Xiong S, Yu K, Zhang B, Wang S, Li Z, Tang Y, Chen G, Li Z, Yu Z, Wang X. ROS-Triggered Self-Assembled Nanoparticles Based on a Chemo-Sonodynamic Combinational Therapy Strategy for the Noninvasive Elimination of Hypoxic Tumors. ACS APPLIED MATERIALS & INTERFACES 2023; 15:15893-15906. [PMID: 36940438 DOI: 10.1021/acsami.3c00990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The hypopermeability and hypoxia in the tumor milieu are important factors that limit multiple treatments. Herein, the reactive oxygen species (ROS)-triggered self-assembled nanoparticles (RP-NPs) was constructed. The natural small molecule Rhein (Rh) was encapsulated into RP-NPs as a sonosensitizer highly accumulated at the tumor site. Then highly tissue-permeable ultrasound (US) irradiation induced apoptosis of tumor cells through the excitation of Rh and acoustic cavitation, which prompted the rapid production of large amounts of ROS in the hypoxic tumor microenvironment. In addition, the thioketal bond structures in the innovatively designed prodrug LA-GEM were triggered and broken by ROS to achieve rapid targeted release of the gemcitabine (GEM). Sonodynamic therapy (SDT) increased the tissue permeability of solid tumors and actively disrupted redox homeostasis via mitochondrial pathways to kill hypoxic tumor cells, and the triggered response mechanism to GEM synergistically amplified the effect of chemotherapy. The chemo-sonodynamic combinational treatment approach is highly effective and noninvasive, with promising applications for hypoxic tumor elimination, such as in cervical cancer (CCa) patients who want to maintain their reproductive function.
Collapse
Affiliation(s)
- Yibing Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510630, China
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, China
| | - Ling Lin
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
| | - Jiashan Xie
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Lixue Wei
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Shuping Xiong
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Kunyi Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
| | - Bingchen Zhang
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
- Department of Oncology, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
| | - Shengtao Wang
- Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University (Foshan Maternity & Child Healthcare Hospital), Foshan, 528000, China
| | - Zibo Li
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
| | - Yan Tang
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
| | - Guimei Chen
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
| | - Zhongjun Li
- Department of Obstetrics and Gynecology, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
- Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, Dongguan, Guangdong 523058, China
| | - Zhiqiang Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
- Department of Oncology, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523058, China
| | - Xuefeng Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong 510630, China
| |
Collapse
|
20
|
|
21
|
Liang Z, Chen D. Targeting therapy effects of composite hyaluronic acid/chitosan nanosystems containing inclusion complexes. Drug Deliv 2022; 29:2734-2741. [PMID: 35983590 PMCID: PMC9397479 DOI: 10.1080/10717544.2022.2112995] [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] [Indexed: 12/03/2022] Open
Abstract
In order to solve the difficulties in the treatment of Staphylococcus aureus infections, a novel enrofloxacin-cyclodextrin (β-CD) inclusion complexes (IC) containing hyaluronic acid/chitosan (HA/CS) self-assemble composite nanosystems covered by poloxamer 188 was designed in our previous study. In this study, the sustained release peforemance, targeting delivery, and therapy effects of the enrofloxacin-composite nanosystems were evaluated in vivo. The enrofloxacin-composite nanosystems had uniform size and smooth surface with drug loading capacity (LC) of 9.92 ± 0.3%. Thermogravimetric analysis (TGA) showed that the material used for the preparation of the enrofloxacin-composite nanosystems did not affect the thermal stability of enrofloxacin. Compared with enrofloxacin injection and enrofloxacin polymeric nanoparticles, the enrofloxacin-composite nanosystems had excellent sustained-release performance in vivo. The enrofloxacin-composite nanosystems have specific targeting to the infection site of Staphylococcus aureus. The excellent sustained release and targeting delivery properties ensure that the anti-infective treatment effect of the enrofloxacin-composite nanosystems in vivo was higher than that of enrofloxacin injection and enrofloxacin polymeric nanoparticles. It can more effectively promote the wound healing. These results suggest that our previous designed enrofloxacin-composite nanosystems will be a promising formulation for effective targeting therapy of Staphylococcus aureus infections.
Collapse
Affiliation(s)
- Zhiwei Liang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei, China
| | - Dongmei Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei, China.,MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei, China
| |
Collapse
|
22
|
He S, Wu L, Sun H, Wu D, Wang C, Ren X, Shao Q, York P, Tong J, Zhu J, Li Z, Zhang J. Antioxidant Biodegradable Covalent Cyclodextrin Frameworks as Particulate Carriers for Inhalation Therapy against Acute Lung Injury. ACS APPLIED MATERIALS & INTERFACES 2022; 14:38421-38435. [PMID: 35948492 DOI: 10.1021/acsami.2c05220] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Drug therapies for acute lung injury (ALI) are far from satisfactory, primarily because drugs cannot specifically target the lungs. Direct delivery of drugs to the deep alveolar regions by inhalation administration is crucial for the treatment of ALI. However, conventional inhalable carriers such as lactose and mannitol are generally inactive. Therefore, the use of a novel pharmacologically active carrier for pulmonary delivery may produce synergetic effects in treating ALI. Considering the pathophysiological environment of ALI, which typically featured excessive reactive oxygen species (ROS) and acute inflammation, we synthesized a novel kind of biodegradable and ROS-sensitive cross-linked covalent cyclodextrin frameworks (OC-COF) with uniform inhalable particle size to treat ALI. OC-COF was devised to incorporate H2O2-scavenging peroxalate ester linkages, which could hydrolyze and eliminate ROS generated in inflammatory sites. Ligustrazine (LIG), an antioxidant and anti-inflammatory natural compound, was loaded into OC-COF and evaluated as a dry powder inhaler (LIG@OC-COF) in vitro and in vivo, showing favorable aerodynamic properties and prominent antioxidant and anti-inflammatory capacities for the synergistic effects of OC-COF and LIG. In ALI rats, inhalation of LIG@OC-COF with a one-fifth LIG dose significantly alleviated the inflammation, oxidant stress, and lung damage. Western blot analysis demonstrated that LIG@OC-COF protected the lungs by regulating the Nrf2/NF-κB signaling pathway. In summary, this study provides a novel ROS-responsive material as an inhalable particulate carrier for the improved treatment of ALI and other medical conditions.
Collapse
Affiliation(s)
- Siyu He
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Wu
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hongyu Sun
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Di Wu
- Anhui University of Chinese Medicine, Hefei 230012, China
| | - Caifen Wang
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaohong Ren
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Qun Shao
- Institute of Pharmaceutical Innovation, University of Bradford, Bradford, West Yorkshire BD7 1DP, United Kingdom
| | - Peter York
- Institute of Pharmaceutical Innovation, University of Bradford, Bradford, West Yorkshire BD7 1DP, United Kingdom
| | - Jiabing Tong
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, China
| | - Jie Zhu
- Anhui University of Chinese Medicine, Hefei 230012, China
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Zegeng Li
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, China
| | - Jiwen Zhang
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, No.2 Tiantan Xili, Beijing 100050, China
| |
Collapse
|
23
|
Wang H, Hao W, Yang L, Yan P, Wei S. Preconditioning with procyanidin B2 protects MAC-T cells against heat exposure-induced mitochondrial dysfunction and inflammation. Mol Immunol 2022; 147:126-135. [DOI: 10.1016/j.molimm.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 11/25/2022]
|
24
|
Raju GSR, Pavitra E, Varaprasad GL, Bandaru SS, Nagaraju GP, Farran B, Huh YS, Han YK. Nanoparticles mediated tumor microenvironment modulation: current advances and applications. J Nanobiotechnology 2022; 20:274. [PMID: 35701781 PMCID: PMC9195263 DOI: 10.1186/s12951-022-01476-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/23/2022] [Indexed: 12/20/2022] Open
Abstract
The tumor microenvironment (TME) plays a key role in cancer development and emergence of drug resistance. TME modulation has recently garnered attention as a potential approach for reprogramming the TME and resensitizing resistant neoplastic niches to existing cancer therapies such as immunotherapy or chemotherapy. Nano-based solutions have important advantages over traditional platform and can be specifically targeted and delivered to desired sites. This review explores novel nano-based approaches aimed at targeting and reprogramming aberrant TME components such as macrophages, fibroblasts, tumor vasculature, hypoxia and ROS pathways. We also discuss how nanoplatforms can be combined with existing anti-tumor regimens such as radiotherapy, immunotherapy, phototherapy or chemotherapy to enhance clinical outcomes in solid tumors.
Collapse
Affiliation(s)
- Ganji Seeta Rama Raju
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul, 04620, Republic of Korea
| | - Eluri Pavitra
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC), Inha University, Incheon, 22212, Republic of Korea
| | - Ganji Lakshmi Varaprasad
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC), Inha University, Incheon, 22212, Republic of Korea
| | | | | | - Batoul Farran
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA.
| | - Yun Suk Huh
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC), Inha University, Incheon, 22212, Republic of Korea.
| | - Young-Kyu Han
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul, 04620, Republic of Korea.
| |
Collapse
|
25
|
Choi HS, Mathew AP, Uthaman S, Vasukutty A, Kim IJ, Suh SH, Kim CS, Ma SK, Graham SA, Kim SW, Park IK, Bae EH. Inflammation-sensing catalase-mimicking nanozymes alleviate acute kidney injury via reversing local oxidative stress. J Nanobiotechnology 2022; 20:205. [PMID: 35477452 PMCID: PMC9044883 DOI: 10.1186/s12951-022-01410-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 04/03/2022] [Indexed: 11/10/2022] Open
Abstract
Background The reactive oxygen species (ROS) and inflammation, a critical contributor to tissue damage, is well-known to be associated with various disease. The kidney is susceptible to hypoxia and vulnerable to ROS. Thus, the vicious cycle between oxidative stress and renal hypoxia critically contributes to the progression of chronic kidney disease and finally, end-stage renal disease. Thus, delivering therapeutic agents to the ROS-rich inflammation site and releasing the therapeutic agents is a feasible solution. Results We developed a longer-circulating, inflammation-sensing, ROS-scavenging versatile nanoplatform by stably loading catalase-mimicking 1-dodecanethiol stabilized Mn3O4 (dMn3O4) nanoparticles inside ROS-sensitive nanomicelles (PTC), resulting in an ROS-sensitive nanozyme (PTC-M). Hydrophobic dMn3O4 nanoparticles were loaded inside PTC micelles to prevent premature release during circulation and act as a therapeutic agent by ROS-responsive release of loaded dMn3O4 once it reached the inflammation site. Conclusions The findings of our study demonstrated the successful attenuation of inflammation and apoptosis in the IRI mice kidneys, suggesting that PTC-M nanozyme could possess promising potential in AKI therapy. This study paves the way for high-performance ROS depletion in treating various inflammation-related diseases. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01410-z.
Collapse
Affiliation(s)
- Hong Sang Choi
- Departments of Internal Medicine, Chonnam National University Medical School, 160, Baekseo‑ro, Dong‑gu, Gwangju, 61469, Republic of Korea.,Departments of Internal Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Ansuja Pulickal Mathew
- Department of Biomedical Sciences, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, 160, Baekseo‑ro, Dong‑gu, Gwangju, 61469, Republic of Korea.,BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun-gun, Jeollanam-do, Republic of Korea
| | - Saji Uthaman
- Department of Biomedical Sciences, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, 160, Baekseo‑ro, Dong‑gu, Gwangju, 61469, Republic of Korea.,BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun-gun, Jeollanam-do, Republic of Korea
| | - Arathy Vasukutty
- Department of Biomedical Sciences, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, 160, Baekseo‑ro, Dong‑gu, Gwangju, 61469, Republic of Korea.,BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun-gun, Jeollanam-do, Republic of Korea
| | - In Jin Kim
- Departments of Internal Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Sang Heon Suh
- Departments of Internal Medicine, Chonnam National University Medical School, 160, Baekseo‑ro, Dong‑gu, Gwangju, 61469, Republic of Korea.,Departments of Internal Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Chang Seong Kim
- Departments of Internal Medicine, Chonnam National University Medical School, 160, Baekseo‑ro, Dong‑gu, Gwangju, 61469, Republic of Korea.,Departments of Internal Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Seong Kwon Ma
- Departments of Internal Medicine, Chonnam National University Medical School, 160, Baekseo‑ro, Dong‑gu, Gwangju, 61469, Republic of Korea.,Departments of Internal Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Sontyana Adonijah Graham
- Department of Biomedical Sciences, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, 160, Baekseo‑ro, Dong‑gu, Gwangju, 61469, Republic of Korea.,BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun-gun, Jeollanam-do, Republic of Korea
| | - Soo Wan Kim
- Departments of Internal Medicine, Chonnam National University Medical School, 160, Baekseo‑ro, Dong‑gu, Gwangju, 61469, Republic of Korea.,Departments of Internal Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Sciences, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, 160, Baekseo‑ro, Dong‑gu, Gwangju, 61469, Republic of Korea. .,BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun-gun, Jeollanam-do, Republic of Korea.
| | - Eun Hui Bae
- Departments of Internal Medicine, Chonnam National University Medical School, 160, Baekseo‑ro, Dong‑gu, Gwangju, 61469, Republic of Korea. .,Departments of Internal Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea.
| |
Collapse
|
26
|
Katila N, Duwa R, Bhurtel S, Khanal S, Maharjan S, Jeong JH, Lee S, Choi DY, Yook S. Enhancement of blood–brain barrier penetration and the neuroprotective effect of resveratrol. J Control Release 2022; 346:1-19. [DOI: 10.1016/j.jconrel.2022.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/01/2022] [Accepted: 04/03/2022] [Indexed: 12/11/2022]
|
27
|
Dutta SD, Hexiu J, Kim J, Sarkar S, Mondal J, An JM, Lee YK, Moniruzzaman M, Lim KT. Two-photon excitable membrane targeting polyphenolic carbon dots for long-term imaging and pH-responsive chemotherapeutic drug delivery for synergistic tumor therapy. Biomater Sci 2022; 10:1680-1696. [DOI: 10.1039/d1bm01832a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Long-term dynamic tracking of cells with theranostics properties remains challenging due to difficulty in preparing and delivering drugs by the probes. Herein, we developed a highly fluorescent one- and two-photon...
Collapse
|
28
|
Ravi Kiran AVVV, Kusuma Kumari G, Krishnamurthy PT, Khaydarov RR. Tumor microenvironment and nanotherapeutics: intruding the tumor fort. Biomater Sci 2021; 9:7667-7704. [PMID: 34673853 DOI: 10.1039/d1bm01127h] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over recent years, advancements in nanomedicine have allowed new approaches to diagnose and treat tumors. Nano drug delivery systems exploit the enhanced permeability and retention (EPR) effect and enter the tumor tissue's interstitial space. However, tumor barriers play a crucial role, and cause inefficient EPR or the homing effect. Mounting evidence supports the hypothesis that the components of the tumor microenvironment, such as the extracellular matrix, and cellular and physiological components collectively or cooperatively hinder entry and distribution of drugs, and therefore, limit the theragnostic applications of cancer nanomedicine. This abnormal tumor microenvironment plays a pivotal role in cancer nanomedicine and was recently recognized as a promising target for improving nano-drug delivery and their therapeutic outcomes. Strategies like passive or active targeting, stimuli-triggered nanocarriers, and the modulation of immune components have shown promising results in achieving anticancer efficacy. The present review focuses on the tumor microenvironment and nanoparticle-based strategies (polymeric, inorganic and organic nanoparticles) for intruding the tumor barrier and improving therapeutic effects.
Collapse
Affiliation(s)
- Ammu V V V Ravi Kiran
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Garikapati Kusuma Kumari
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Renat R Khaydarov
- Institute of Nuclear Physics, Uzbekistan Academy of Sciences, Tashkent, 100047, Uzbekistan.
| |
Collapse
|
29
|
Liang W, He X, Bi J, Hu T, Sun Y. Role of reactive oxygen species in tumors based on the 'seed and soil' theory: A complex interaction (Review). Oncol Rep 2021; 46:208. [PMID: 34328200 PMCID: PMC8329912 DOI: 10.3892/or.2021.8159] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor microenvironment (TME) can serve as the 'soil' for the growth and survival of tumor cells and function synergically with tumor cells to mediate tumor progression and therapeutic resistance. Reactive oxygen species (ROS) is somewhat of a double‑edged sword for tumors. Accumulating evidence has reported that regulating ROS levels can serve an anti‑tumor role in the TME, including the promotion of cancer cell apoptosis, inhibition of angiogenesis, preventing immune escape, manipulating tumor metabolic reorganization and improving drug resistance. In the present review, the potential role of ROS in anti‑tumor therapy was summarized, including the possibility of directly or indirectly targeting the TME.
Collapse
Affiliation(s)
- Wei Liang
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Affiliated Hospital of Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Xinying He
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Affiliated Hospital of Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Jianqiang Bi
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Affiliated Hospital of Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Tingting Hu
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Affiliated Hospital of Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Yunchuan Sun
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Affiliated Hospital of Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| |
Collapse
|
30
|
Banstola A, Poudel K, Kim JO, Jeong JH, Yook S. Recent progress in stimuli-responsive nanosystems for inducing immunogenic cell death. J Control Release 2021; 337:505-520. [PMID: 34314800 DOI: 10.1016/j.jconrel.2021.07.038] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 01/10/2023]
Abstract
Low immunogenicity and immunosuppressive tumor microenvironments are major hurdles in the application of cancer immunotherapy. To date, several immunogenic cell death (ICD) inducers have been reported to boost cancer immunotherapy by triggering ICD. ICD is characterized by the release of proinflammatory cytokines, danger-associated molecular patterns (DAMPs) and tumor associated antigens which will generate anticancer immunity by triggering adaptive immune cells. However, application of ICD inducers is limited due to severe toxicity issues and inefficient localization in the tumor microenvironment. To circumvent these challenges, stimuli-responsive nanoparticles have been exploited for improving cancer immunotherapy by limiting its toxicity. The combination of stimuli-responsive nanoparticles with an ICD inducer serves as a promising strategy for increasing the clinical applications of ICD induction in cancer immunotherapy. Here, we outline recent advances in ICD mediated by stimuli-responsive nanoparticles that may be near-infrared (NIR)-responsive, pH-responsive, redox responsive, pH and enzyme responsive, or pH and redox responsive, and evaluate their significant potential for successful clinical translation in cancer immunotherapy.
Collapse
Affiliation(s)
- Asmita Banstola
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Kishwor Poudel
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea.
| |
Collapse
|